Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 91
Filter
Add more filters











Publication year range
1.
Vet Pathol ; 53(3): 602-13, 2016 May.
Article in English | MEDLINE | ID: mdl-26797094

ABSTRACT

Ductal plate malformations (DPMs) represent developmental biliary disorders with a wide phenotypic spectrum. This study characterizes DPM in 30 Boxer dogs. Median age was 1.5 (range, 0.3-10.0) years, with 12 dogs <1 year. Clinical features included increased serum levels of liver enzymes (28), gastrointestinal signs (16), poor body condition (14), abdominal effusion (9), and hepatic encephalopathy (2). Additional malformations included gallbladder atresia (8), atrophied left liver (2), absent quadrate lobe with left-displaced gallbladder (1), portal vasculature atresia (left liver, 1), intrahepatic portosystemic shunt (1), and complex intrahepatic arteriovenous malformation (1). All dogs had portal tracts dimensionally expanded by a moderate-to-severe multiple small bile duct phenotype embedded in abundant extracellular matrix; 80% displayed variable portal-to-portal bridging. Quantitative analysis confirmed significantly increased fibrillar collagen and a 3-fold increased portal tract area relative to 6 Boxer and 10 non-Boxer controls. Biliary phenotype was dominated by tightly formed CK19-positive ductules, typically 10 to 15 µm in diameter, with 3 to >30 profiles per portal tract, reduced luminal apertures, and negative Ki-67 immunoreactivity. CK19-positive biliary epithelium intersected directly with zone 1 hepatocytes as a signature feature when considered with other DPM characteristics. Phenotypic variation included a multiple small bile duct phenotype (all dogs), predominantly thin-walled sacculated ducts (4), well-formed saccular ducts (4), and sacculated segmental, interlobular, and intralobular ducts (Caroli malformation, 2 dogs, one with bridging portal fibrosis). Histologic evidence of portal venous hypoperfusion accompanied increased biliary profiles in every case. We propose that this spectrum of disorders be referred to as DPM with appropriate modifiers to characterize the unique phenotypes.


Subject(s)
Bile Ducts/pathology , Caroli Disease/veterinary , Dog Diseases/pathology , Liver Cirrhosis/veterinary , Liver/pathology , Animals , Bile Ducts/embryology , Bile Ducts/metabolism , Caroli Disease/embryology , Caroli Disease/metabolism , Caroli Disease/pathology , Dog Diseases/embryology , Dog Diseases/metabolism , Dogs , Female , Gallbladder/pathology , Hepatocytes/metabolism , Hepatocytes/pathology , Keratin-19/metabolism , Liver/embryology , Liver/metabolism , Liver Cirrhosis/embryology , Liver Cirrhosis/metabolism , Liver Cirrhosis/pathology , Male , Phenotype
2.
J Vet Intern Med ; 29(4): 1069-73, 2015.
Article in English | MEDLINE | ID: mdl-26118570

ABSTRACT

BACKGROUND: The prevalence of concurrent disease in hyperthyroid cats is unknown. OBJECTIVES: To identify the prevalence of concurrent intra-abdominal disease using abdominal ultrasound examination (AUS) in hyperthyroid cats referred for radioactive iodine treatment (RIT) and to determine whether the requirement for pretreatment AUS is justified. ANIMALS: Five hundred and thirty-four client-owned cats diagnosed with hyperthyroidism and referred for RIT. METHODS: Retrospective study. Age, breed, sex, body weight, clinical signs, total serum T4 concentration, blood urea nitrogen (BUN) concentration, serum creatinine concentration, urine specific gravity (USG), AUS results, and biopsy or cytology results, or both (if obtained) were collected from the medical records. RESULTS: The prevalence of concurrent disease identified using AUS in hyperthyroid cats referred for RIT was 36.1%; 22.8% of the cats in the study had renal disease and 2.4% had confirmed neoplasia. Significant differences in median USG (P value 0.032) and median BUN (P value 0.028) were found between cats that had abnormal kidneys on AUS compared to those with normal-appearing kidneys. Only 2.2% of the cats were not treated with RIT as a result of changes identified on AUS and subsequently obtained cytology or biopsy results. CONCLUSIONS AND CLINICAL IMPORTANCE: The results indicate that pretreatment AUS in hyperthyroid cats referred for RIT is unnecessary in most patients.


Subject(s)
Cat Diseases/diagnostic imaging , Hyperthyroidism/veterinary , Iodine Radioisotopes/therapeutic use , Abdomen/diagnostic imaging , Animals , Cat Diseases/radiotherapy , Cats , Female , Hyperthyroidism/complications , Hyperthyroidism/diagnostic imaging , Hyperthyroidism/radiotherapy , Kidney Diseases/complications , Kidney Diseases/diagnostic imaging , Kidney Diseases/veterinary , Male , Retrospective Studies , Ultrasonography
3.
Neuroscience ; 278: 11-9, 2014 Oct 10.
Article in English | MEDLINE | ID: mdl-25086317

ABSTRACT

Alterations in hippocampal neurogenesis affect spatial learning, though, the relative contributions of cell proliferation and cell survival on this process are poorly understood. The current study utilized mu opioid receptor (MOR-1) knockout (KO) mice on two background strains, C57BL/6 and 129S6, to assess cell survival as well as determine the impact on spatial learning using the Morris water maze. These experiments were designed to extend prior work showing that both C57BL/6 and 129S6 MOR-1 KO mice have an increased number of proliferating cells in the dentate gyrus (DG) when compared to wild-type (WT) mice. The current study indicates that newly born neurons in the DG of C57BL/6 MOR-1 KO mice exhibit enhanced survival when compared to WT mice, while new neurons in the DG of 129S6 MOR-1 KO mice do not. In addition, C57BL/6 MOR-1 KO mice have a lower number of apoptotic cells in the DG compared to WT mice while, in contrast, 129S6 MOR-1 KO mice have a higher number of apoptotic cells in this region. These alterations collectively contribute to an increase in the granule cell number in the DG of C57BL/6 MOR-1 KO mice, while the total number of granule cells in 129S6 MOR-1 KO mice is unchanged. Thus, although C57BL/6 and 129S6 MOR-1 KO mice both exhibit increased cell proliferation in the DG, the impact of the MOR-1 mutation on cell survival differs between strains. Furthermore, the decrease in DG cell survival displayed by 129S6 MOR-1 KO mice is correlated with functional deficits in spatial learning, suggesting that MOR-1-dependent alterations in the survival of new neurons in the DG, and not MOR-1-dependent changes in proliferation of progenitor cells in the DG, is important for spatial learning.


Subject(s)
Dentate Gyrus/physiology , Neurogenesis , Receptors, Opioid, mu/physiology , Spatial Learning/physiology , Animals , Cell Death , Cell Survival , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Receptors, Opioid, mu/genetics , Species Specificity
4.
Neuroscience ; 206: 49-59, 2012 Mar 29.
Article in English | MEDLINE | ID: mdl-22280973

ABSTRACT

The endogenous opioid system is involved in various physiological processes, including neurogenesis in the dentate gyrus (DG) of the hippocampus. In the current study, we investigated the role of the mu opioid receptor (MOR-1) on DG neurogenesis and measured glucocorticoid levels following several injection paradigms to supplement the neurogenesis experiments. MOR-1 knockout (KO) mice on C57BL/6 and 129S6 backgrounds were injected with bromodeoxyuridine (BrdU) using either a single injection or two different repeated injection protocols and then sacrificed at different time points. The total number of BrdU and proliferating cell nuclear antigen (PCNA) positive cells in the DG is significantly increased in MOR-1 KO mice compared with wild type (WT) on both strains after repeated injection, but not after a single injection. Plasma corticosterone (CORT) levels increased similarly in MOR-1 KO and WT mice following both single and repeated injection, indicating that the stress response is activated following any injection protocol, but that the mechanism responsible for the increase in BrdU labeling in MOR-1 KO mice is CORT-level independent. Finally, WT 129S6 mice, independent of genotype, showed higher levels of plasma CORT compared with WT C57BL/6 mice in both noninjected controls and following injection at two separate time points; these levels were inversely correlated with low numbers of BrdU cells in the DG in 129S6 mice compared with C57BL/6 mice. In summary, these data demonstrate that loss of MOR-1 increases BrdU labeling in the DG independent of CORT levels, but only following a repeated injection, illustrating the capability of injection paradigms to influence cell-proliferative responses in a genotype-dependent manner.


Subject(s)
Bromodeoxyuridine/administration & dosage , Dentate Gyrus/metabolism , Neurogenesis/physiology , Receptors, Opioid, mu/metabolism , Staining and Labeling/methods , Animals , Cortisone/blood , Fluorescent Antibody Technique , Immunohistochemistry , Male , Mice , Mice, 129 Strain , Mice, Inbred C57BL , Mice, Knockout , Radioimmunoassay , Receptors, Opioid, mu/genetics
5.
Endocrinology ; 150(4): 1739-47, 2009 Apr.
Article in English | MEDLINE | ID: mdl-19022883

ABSTRACT

Amidated peptides are critically involved in many physiological functions. Genetic deletion of peptidylglycine alpha-amidating monooxygenase (PAM), the only enzyme that can synthesize these peptides, is embryonically lethal. The goal of the present study was the identification of physiological functions impaired by haploinsufficiency of PAM. Regulation of the hypothalamic-pituitary-thyroid axis and body temperature, functions requiring contributions from multiple amidated peptides, were selected for evaluation. Based on serum T(4) and pituitary TSH-beta mRNA levels, mice heterozygous for PAM (PAM(+/-)) were euthyroid at baseline. Feedback within the hypothalamic-pituitary-thyroid axis was impaired in PAM(+/-) mice made hypothyroid using a low iodine/propylthiouracil diet. Despite their normal endocrine response to cold, PAM(+/-) mice were unable to maintain body temperature as well as wild-type littermates when kept in a 4 C environment. When provided with additional dietary copper, PAM(+/-) mice maintained body temperature as well as wild-type mice. Pharmacological activation of vasoconstriction or shivering also allowed PAM(+/-) mice to maintain body temperature. Cold-induced vasoconstriction was deficient in PAM(+/-) mice. This deficit was eliminated in PAM(+/-) mice receiving a diet with supplemental copper. These results suggest that dietary deficiency of copper, coupled with genetic deficits in PAM, could result in physiological deficits in humans.


Subject(s)
Copper/pharmacology , Mixed Function Oxygenases/deficiency , Mixed Function Oxygenases/genetics , Multienzyme Complexes/deficiency , Multienzyme Complexes/genetics , Adipose Tissue, Brown/drug effects , Adipose Tissue, Brown/metabolism , Animals , Body Temperature/drug effects , Body Temperature/genetics , Cold Temperature , Copper/administration & dosage , Dietary Supplements , Female , Genotype , Hypothalamus/drug effects , Hypothalamus/metabolism , Ion Channels/genetics , Ion Channels/metabolism , Male , Mice , Mice, Mutant Strains , Mitochondrial Proteins/genetics , Mitochondrial Proteins/metabolism , Mixed Function Oxygenases/physiology , Multienzyme Complexes/physiology , Phenylephrine/pharmacology , Piperazines/pharmacology , Pyridines/pharmacology , Radioimmunoassay , Reverse Transcriptase Polymerase Chain Reaction , Rheology , Uncoupling Protein 1 , Vasoconstriction/drug effects , Vasoconstriction/physiology
6.
Neuroscience ; 153(3): 773-88, 2008 May 15.
Article in English | MEDLINE | ID: mdl-18424009

ABSTRACT

On the basis of numerous studies that have described interactions between the dopaminergic and opioidergic systems, we have investigated whether genetic deletion of dopamine D2 receptors (D2R) might influence the expression of central opioid receptors. The levels of mu, delta, kappa and nociceptin opioid peptide receptors were determined in the brains and spinal cords of D2R knockout mice using quantitative autoradiography. The significant changes in opioid receptor binding found in the brains of heterozygous and homozygous mice were mainly restricted to the basal ganglia. In homozygous mice, a down-regulation of mu and delta receptors was observed in the striatal and pallidal areas. This alteration may be an adaptive response to the increase in enkephalin levels previously described in the striatum of these mutant mice. On the contrary, an up-regulation of kappa receptors was found in the striatal and nigral regions and might be related to a change in dynorphin levels. Significant increases in nociceptin receptor binding were also observed in homozygous mice in brain areas involved in motor behavior. At the spinal level, only kappa and nociceptin receptor binding showed significant overall differences between genotypes. The functional consequences of these adaptive changes are discussed in relation to the findings of behavioral and neurochemical studies reported to date in D2R knockout mice.


Subject(s)
Brain/metabolism , Receptors, Dopamine D2/deficiency , Receptors, Opioid/biosynthesis , Spinal Cord/metabolism , Adaptation, Physiological , Animals , Autoradiography , Male , Mice , Mice, Knockout , Receptors, Dopamine D2/genetics , Receptors, Opioid/metabolism , Nociceptin Receptor
7.
Neuroscience ; 150(4): 807-17, 2007 Dec 19.
Article in English | MEDLINE | ID: mdl-17997230

ABSTRACT

Analgesic effects of delta opioid receptor (DOR) -selective agonists are enhanced during persistent inflammation and arthritis. Although the underlying mechanisms are still unknown, membrane density of DOR was shown to be increased 72 h after induction of inflammation, an effect abolished in mu opioid receptor (MOR) -knockout (KO) mice [Morinville A, Cahill CM, Kieffer B, Collier B, Beaudet A (2004b) Mu-opioid receptor knockout prevents changes in delta-opioid receptor trafficking induced by chronic inflammatory pain. Pain 109:266-273]. In this study, we demonstrated a crucial role of MOR in DOR-mediated antihyperalgesia. Intrathecal administration of the DOR selective agonist deltorphin II failed to induce antihyperalgesic effects in MOR-KO mice, whereas it dose-dependently reversed thermal hyperalgesia in wild-type mice. The antihyperalgesic effects of deltorphin II were blocked by naltrindole but not d-Phe-Cys-Tyr-D-Trp-Orn-Thr-Pen-Thr-NH(2) (CTOP) suggesting that this agonist was mainly acting through DOR. SNC80-induced antihyperalgesic effects in MOR-KO mice were also attenuated as compared with littermate controls. In contrast, kappa opioid receptor knockout did not affect deltorphin II-induced antihyperalgesia. As evaluated using mice lacking endogenous opioid peptides, the regulation of DOR's effects was also independent of beta-endorphin, enkephalins, or dynorphin opioids known to be released during persistent inflammation. We therefore conclude that DOR-mediated antihyperalgesia is dependent on MOR expression but that activation of MOR by endogenous opioids is probably not required.


Subject(s)
Hyperalgesia/drug therapy , Receptors, Opioid, delta/metabolism , Receptors, Opioid, mu/physiology , Animals , Dose-Response Relationship, Drug , Dynorphins/deficiency , Enkephalins/deficiency , Freund's Adjuvant , Hyperalgesia/etiology , Inflammation/chemically induced , Inflammation/complications , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Motor Activity/drug effects , Naltrexone/administration & dosage , Narcotic Antagonists/administration & dosage , Oligopeptides/administration & dosage , Pain Measurement , Protein Precursors/deficiency , Reaction Time/drug effects , Receptors, Opioid, kappa/deficiency , Receptors, Opioid, mu/deficiency , Somatostatin/administration & dosage , Somatostatin/analogs & derivatives , beta-Endorphin/deficiency
8.
Neuroscience ; 147(2): 439-44, 2007 Jun 29.
Article in English | MEDLINE | ID: mdl-17544222

ABSTRACT

Opioids are extensively used analgesics yet can paradoxically increase pain sensitivity in humans and rodents. This hyperalgesia is extensively conceptualized to be a consequence of opioid receptor activity, perhaps providing an adaptive response to analgesia, and to utilize N-methyl-D-aspartate (NMDA) receptors. These assumptions were tested here in opioid receptor triple knock-out (KO) mice lacking all three genes encoding opioid receptors (mu, delta, and kappa) by comparing their thermal nociceptive responses to the opioids morphine and oxymorphone with those of B6129F(1) controls. Injecting acute opioid bolus doses in controls caused maximal analgesia that was completely abolished in KO mice, confirming the functional consequence of the KO mouse opioid receptor deficiency. Continuous opioid infusion by osmotic pump in control mice also initially caused several consecutive days of analgesia that was shortly thereafter followed by several consecutive days of hyperalgesia. In contrast, continuously infusing KO mice with opioids caused no detectable analgesic response, but only immediate and steady declines in nociceptive thresholds culminating in several days of unremitting hyperalgesia. Finally, injecting the non-competitive NMDA receptor antagonist MK-801 during opioid infusion markedly reversed hyperalgesia in control but not KO mice. These data demonstrate that sustained morphine and oxymorphone delivery causes hyperalgesia independently of prior or concurrent opioid or NMDA receptor activity or opioid analgesia, indicating the contribution of mechanisms outside of current conceptions, and are inconsistent with proposals of hyperalgesia as a causative factor of opioid analgesic tolerance.


Subject(s)
Analgesics, Opioid/pharmacology , Pain/genetics , Pain/physiopathology , Receptors, Opioid/genetics , Receptors, Opioid/physiology , Analgesics, Opioid/administration & dosage , Animals , Data Interpretation, Statistical , Dizocilpine Maleate/pharmacology , Excitatory Amino Acid Antagonists/pharmacology , Hyperalgesia/chemically induced , Hyperalgesia/genetics , Hyperalgesia/physiopathology , Mice , Mice, Knockout , Morphine/pharmacology , Oxymorphone/pharmacology , Pain Measurement/drug effects , Reaction Time/physiology , Receptors, N-Methyl-D-Aspartate/drug effects
9.
Neuroscience ; 146(4): 1795-807, 2007 Jun 08.
Article in English | MEDLINE | ID: mdl-17467916

ABSTRACT

Phosphorylation of specific sites in the second intracellular loop and in the C-terminal domain have previously been suggested to cause desensitization and internalization of the mu-opioid receptor (MOP-R). To assess sites of MOP-R phosphorylation in vivo, affinity-purified, phosphoselective antibodies were raised against either phosphothreonine-180 in the second intracellular loop (MOR-P1) or the C-terminal domain of MOP-R containing phosphothreonine-370 and phosphoserine-375 (MOR-P2). We found that MOR-P2-immunoreactivity (IR) was significantly increased within the striatum of wild-type C57BL/6 mice after injection of the agonist fentanyl. Pretreatment with the antagonist naloxone blocked the fentanyl-induced increase. Furthermore, mutant mice lacking MOP-R showed only non-specific nuclear MOR-P2-IR before or after fentanyl treatment, confirming the specificity of the MOR-P2 antibodies. To assess whether MOP-R phosphorylation occurs following endogenous opioid release, we induced chronic neuropathic pain by partial sciatic nerve ligation (pSNL), which caused a significant increase in MOR-P2-IR in the striatum. pSNL also induced signs of mu opioid receptor tolerance demonstrated by a rightward shift in the morphine dose response in the tail withdrawal assay and by a reduction in morphine conditioned place preference (CPP). Mutant mice selectively lacking all forms of the beta-endorphin peptides derived from the proopiomelanocortin (Pomc) gene did not show increased MOR-P2-IR, decreased morphine antinociception, or reduced morphine CPP following pSNL. In contrast gene deletion of either proenkephalin or prodynorphin opioids did not block the effects of pSNL. These results suggest that neuropathic pain caused by pSNL in wild-type mice activates the release of the endogenous opioid beta-endorphin, which subsequently induces MOP-R phosphorylation and opiate tolerance.


Subject(s)
Receptors, Opioid, mu/genetics , Receptors, Opioid, mu/metabolism , Sciatica/metabolism , Analgesics, Opioid/pharmacology , Analysis of Variance , Animals , Behavior, Animal , Cell Line, Transformed , Conditioning, Operant/drug effects , Conditioning, Operant/physiology , Corpus Striatum/drug effects , Corpus Striatum/metabolism , Drug Interactions , Enkephalin, Ala(2)-MePhe(4)-Gly(5)-/pharmacology , Green Fluorescent Proteins/biosynthesis , Humans , Hyperalgesia/etiology , Mice , Mice, Knockout , Mutagenesis/physiology , Naloxone/pharmacology , Narcotic Antagonists/pharmacology , Phosphorylation/drug effects , Phosphothreonine/immunology , Phosphothreonine/metabolism , Receptors, Opioid, mu/chemistry , Sciatica/complications , Sciatica/pathology , Transfection , beta-Endorphin/deficiency , beta-Endorphin/metabolism
10.
J Small Anim Pract ; 47(10): 588-95, 2006 Oct.
Article in English | MEDLINE | ID: mdl-17004951

ABSTRACT

OBJECTIVES: The differential diagnosis for young to middle-aged dogs with progressive neurological signs, focal or multifocal computed tomography/magnetic resonance imaging lesions, mononuclear cerebrospinal fluid pleocytosis and negative infectious titres includes granulomatous meningoencephalomyelitis, breed-specific meningoencephalitis, infectious meningoencephalitis of unknown origin and central nervous system neoplasia. The terminology meningoencephalitis of unknown aetiology may be preferable for cases that lack histopathological diagnoses. The safety and efficacy of a combination of cytosine arabinoside and prednisone protocol is evaluated, in this study, for the treatment of meningoencephalitis of unknown aetiology in 10 dogs. METHODS: Cases were selected based on neuroanatomical localisation, negative regional infectious disease titres, cerebrospinal fluid pleocytosis and brain imaging. Clinical response was gauged through follow-up examinations, owner and referring veterinarian surveys and review of medical records. RESULTS: Partial or complete remission was achieved in all dogs; the median survival time for the 10 dogs was 531 days (range 46 to 1025 days), with five of the 10 dogs alive at the time of writing. CLINICAL SIGNIFICANCE: Prednisone/cytosine arabinoside is a safe empirical therapy for dogs with meningoencephalitis of unknown aetiology; this drug combination may prolong survival time.


Subject(s)
Anti-Inflammatory Agents/therapeutic use , Cytarabine/therapeutic use , Dog Diseases/drug therapy , Meningoencephalitis/veterinary , Prednisone/therapeutic use , Animals , Anti-Inflammatory Agents/adverse effects , Cytarabine/adverse effects , Dog Diseases/etiology , Dog Diseases/mortality , Dogs , Drug Therapy, Combination , Female , Male , Meningoencephalitis/drug therapy , Meningoencephalitis/etiology , Meningoencephalitis/mortality , Prednisone/adverse effects , Prognosis , Retrospective Studies , Safety , Survival Analysis , Time Factors , Treatment Outcome
11.
Neuropeptides ; 39(6): 559-67, 2005 Dec.
Article in English | MEDLINE | ID: mdl-16289278

ABSTRACT

Agonist stimulation of opioid receptors increases feeding in rodents, while opioid antagonists inhibit food intake. The pan-opioid antagonist, LY255582, produces a sustained reduction in food intake and body weight in rodent models of obesity. However, the specific receptor subtype(s) responsible for this activity is unknown. To better characterize the pharmacology of LY255582, we examined the binding of a radiolabeled version of the molecule, [(3)H]-LY255582, in mouse brain using autoradiography. In mouse brain homogenates, the K(d) and B(max) for [(3)H]-LY255582 were 0.156 +/- 0.07 nM and 249 +/- 14 fmol/mg protein, respectively. [(3)H]-LY255582 bound to slide mounted sections of mouse brain with high affinity and low non-specific binding. High levels of binding were seen in areas consistent with the known localization of opioid receptors. These areas included the caudate putamen, nucleus accumbens, claustrum, medial habenula, dorsal endopiriform nucleus, basolateral nucleus of the amygdala, hypothalamus, thalamus and ventral tegmental area. We compared the binding distribution of [(3)H]-LY255582 to the opioid receptor antagonist radioligands [(3)H]-naloxone (mu), [(3)H]-naltrindole (delta) and [(3)H]-norBNI (kappa). The overall distribution of [(3)H]-LY255582 binding sites was similar to that of the other ligands. No specific [(3)H]-LY255582 binding was noted in sections of mu-, delta- and kappa-receptor combinatorial knockout mice. Therefore, it is likely that LY255582 produces its effects on feeding and body weight gain through a combination of mu-, delta- and kappa-receptor activity.


Subject(s)
Brain/metabolism , Cyclohexanes/metabolism , Piperidines/metabolism , Receptors, Opioid, delta/metabolism , Receptors, Opioid, kappa/metabolism , Receptors, Opioid, mu/metabolism , Animals , Autoradiography , Binding Sites , Brain/anatomy & histology , Cyclohexanes/chemistry , Mice , Mice, Knockout , Molecular Structure , Naloxone/metabolism , Naltrexone/analogs & derivatives , Naltrexone/metabolism , Narcotic Antagonists/metabolism , Piperidines/chemistry , Receptors, Opioid, delta/genetics , Receptors, Opioid, kappa/genetics , Receptors, Opioid, mu/genetics , Tritium/chemistry , Tritium/metabolism
12.
Neuroscience ; 130(2): 359-67, 2005.
Article in English | MEDLINE | ID: mdl-15664692

ABSTRACT

There is general agreement that dopaminergic neurons projecting from the ventral tegmental area (VTA) to the nucleus accumbens and prefrontal cortex play a key role in drug reinforcement. The activity of these neurons is strongly modulated by the inhibitory and excitatory input they receive. Activation of mu-opioid receptors, located on GABAergic neurons in the VTA, causes hyperpolarization of these GABAergic neurons, thereby causing a disinhibition of VTA dopaminergic neurons. This effect of mu-opioid receptors upon GABA neurotransmission is a likely mechanism for mu-opioid receptor modulation of drug reinforcement. We studied mu-opioid receptor signaling in relation to cocaine reinforcement in wild-type and mu-opioid receptor knockout mice using a cocaine self-administration paradigm and in vitro electrophysiology. Cocaine self-administration was reduced in mu-opioid receptor knockout mice, suggesting a critical role of mu-opioid receptors in cocaine reinforcement. The frequency of spontaneous inhibitory post-synaptic currents onto dopaminergic neurons in the ventral tegmental area was increased in mu-opioid receptor knockout mice compared with wild-type controls, while the frequency of spontaneous excitatory post-synaptic currents was unaltered. The reduced cocaine self-administration and increased GABAergic input to VTA dopaminergic neurons in mu-opioid receptor knockout mice supports the notion that suppression of GABAergic input onto dopaminergic neurons in the VTA contributes to mu-opioid receptor modulation of cocaine reinforcement.


Subject(s)
Cocaine/pharmacology , Neurons/metabolism , Receptors, Opioid, mu/genetics , Reinforcement, Psychology , Ventral Tegmental Area/drug effects , gamma-Aminobutyric Acid/metabolism , Afferent Pathways/drug effects , Afferent Pathways/metabolism , Afferent Pathways/physiopathology , Animals , Cocaine-Related Disorders/metabolism , Cocaine-Related Disorders/physiopathology , Disease Models, Animal , Dopamine/metabolism , Male , Membrane Potentials/drug effects , Membrane Potentials/physiology , Mice , Mice, Knockout , Neural Inhibition/drug effects , Neural Inhibition/physiology , Neurons/drug effects , Self Administration , Synapses/drug effects , Synapses/metabolism , Synaptic Transmission/drug effects , Synaptic Transmission/physiology , Up-Regulation/drug effects , Up-Regulation/physiology , Ventral Tegmental Area/metabolism , Ventral Tegmental Area/physiopathology
13.
Neuroscience ; 125(1): 211-20, 2004.
Article in English | MEDLINE | ID: mdl-15051160

ABSTRACT

Cocaine-induced behavioral sensitization is a complex phenomenon involving a number of neuromodulator and neurotransmitter systems. To specifically investigate the role of the micro opioid receptor (MOR) in cocaine-induced behavioral sensitization in mice, both genetic and pharmacological approaches were undertaken. MOR-1 deficient mice of varying backgrounds (C57BL/6J, 129S6, F1 hybrid 129S6xC57BL/6J and 129S6xC57BL/6J) and wild-type C57BL/6J mice exposed continuously to naltrexone, an opioid receptor antagonist, received single daily injections of saline or cocaine for 10 days. All mice received a single cocaine challenge 7 days following the last saline or cocaine injection to test for the expression of sensitization. The locomotor-stimulating and sensitizing effects of cocaine observed in MOR-1 wild-type mice were absent in MOR-1 knockout mice maintained on the mixed 129S6xC57BL/6J background. In contrast, MOR-1 deficient mice developed on a C57BL/6J background showed an accentuated sensitivity to cocaine-induced locomotion. Cocaine's psychomotor activating effects were more pronounced in the MOR-1 C57BL/6J knockouts injected daily with cocaine than in the MOR-1 wild-type mice. Similar locomotor-stimulating and sensitizing effects were found in both F1 hybrid 129S6xC57BL/6J MOR-1 wild-type and MOR-1 knockout mice, while the 129S6 strain showed an overall indifference to cocaine. That is, both the locomotor-stimulating and sensitizing effects of cocaine were absent in both MOR-1 wild-type and MOR-1 knockout mice maintained on the 129S6 background. Lastly, the locomotor-stimulating and sensitizing effects of cocaine were attenuated in C57BL/6J wild-type mice exposed continuously to naltrexone. Collectively, these data support a role for opioidergic involvement in cocaine-influenced behavior in mice. Moreover, MORs appear to differentially modulate a sensitized response to cocaine in different strains of mice as delineated by MOR-1 gene deletion and pharmacological antagonism.


Subject(s)
Adaptation, Physiological/physiology , Cocaine/pharmacology , Dopamine Uptake Inhibitors/pharmacology , Receptors, Opioid, mu/genetics , Animals , Behavior, Animal , Mice , Mice, Knockout , Motor Activity/drug effects , Naltrexone/pharmacology , Narcotic Antagonists/pharmacology , Receptors, Opioid, mu/deficiency
14.
J Endocrinol ; 179(2): 227-35, 2003 Nov.
Article in English | MEDLINE | ID: mdl-14596674

ABSTRACT

Analysis of knockout mice suggests that the neurotropin-inducible secreted polypeptide VGF (non-acronymic) plays an important role in the regulation of energy balance. VGF is synthesized by neurons in the central and peripheral nervous systems (CNS, PNS), as well as in the adult pituitary, adrenal medulla, endocrine cells of the stomach and pancreatic beta cells. Thus VGF, like cholecystokinin, leptin, ghrelin and other peptide hormones that have been shown to regulate feeding and energy expenditure, is synthesized in both the gut and the brain. Although detailed developmental studies of VGF localization in the CNS and PNS have been completed, little is known about the ontogeny of VGF expression in endocrine and neuroendocrine tIssues. Here, we report that VGF mRNA is detectable as early as embryonic day 15.5 in the developing rat gastrointestinal and esophageal lumen, pancreas, adrenal, and pituitary, and we further demonstrate that VGF mRNA is synthesized in the gravid rat uterus, together supporting possible functional roles for this polypeptide outside the nervous system and in the enteric plexus.


Subject(s)
Neurosecretory Systems/chemistry , Neurosecretory Systems/embryology , Proteins/genetics , RNA, Messenger/analysis , Adrenal Glands/chemistry , Adrenal Glands/embryology , Animals , DNA-Binding Proteins/genetics , Endocrine System/chemistry , Endocrine System/embryology , Female , Fushi Tarazu Transcription Factors , GATA2 Transcription Factor , Homeodomain Proteins/genetics , Humans , In Situ Hybridization/methods , Islets of Langerhans/chemistry , Islets of Langerhans/embryology , Neuropeptides , Paired Box Transcription Factors , Pituitary Gland/chemistry , Pituitary Gland/embryology , Pregnancy , Rats , Rats, Sprague-Dawley , Receptors, Cytoplasmic and Nuclear , Sequence Homology , Steroidogenic Factor 1 , Transcription Factors/genetics , Uterus/chemistry
15.
Neuroscience ; 117(1): 157-68, 2003.
Article in English | MEDLINE | ID: mdl-12605902

ABSTRACT

The opioid receptor-like 1 receptor is a novel member of the opioid receptor family and its endogenous peptide ligand has been termed nociceptin and orphanin FQ. Activation of the opioid receptor-like 1 receptor by nociceptin/orphanin FQ in vivo produces hyperalgesia when this peptide is given supraspinally but analgesia at the spinal level. Nociceptin/orphanin FQ also reverses stress-induced analgesia, suggesting that the peptide has anti-opioid properties. Nociceptin/orphanin FQ knockout mice show alterations in pain sensitivity and stress responses and display increased morphine dependence, suggesting an interaction of the nociceptin/orphanin FQ system with classical opioid receptor function. To determine if the behavioural phenotype of nociceptin/orphanin FQ knockout mice reflects changes in either opioid receptor-like 1 or classical opioid receptor expression, we have carried out quantitative autoradiography of the opioid receptor-like 1, mu-, delta- and kappa-opioid receptors in the brains of these animals. Receptor density was measured on coronal sections from wild-type, heterozygous and homozygous mice using [(3)H]nociceptin, [(3)H][D-Ala(2)-N-methyl-Phe(4)-Gly(5) ol] enkephalin, [(3)H]deltorphin-I, or [(3)H](-)-N-methyl-N-[7-(1-pyrrodinyl)-1-oxospiro[4,5]dec-8-yl]-4-benzofuranacetamide to label opioid receptor-like 1, mu-, delta- and kappa-receptors, respectively. A region-specific up-regulation of the opioid receptor-like 1 receptor (up to 135%) was seen in brains from homozygous mice. Mu-Receptors also showed significant differences between genotypes whilst changes in delta- and kappa- receptors were minor. In conclusion the region-specific up-regulation of the opioid receptor-like 1 receptor indicates a tonic role for nociceptin/orphanin FQ in some brain structures and may suggest the peptide regulates the receptor expression in these regions. The changes in the opioid receptor-like 1 receptor may relate to the anxiogenic phenotype of these animals but the observed change in mu-receptors does not correlate with altered morphine responses.


Subject(s)
Brain/metabolism , Receptors, Opioid/biosynthesis , Receptors, Opioid/deficiency , Up-Regulation/physiology , Animals , Brain Chemistry/physiology , Gene Expression Regulation/physiology , Mice , Mice, Knockout , Receptors, Opioid/analysis , Receptors, Opioid/genetics , Nociceptin Receptor
16.
Proc Natl Acad Sci U S A ; 99(5): 3087-92, 2002 Mar 05.
Article in English | MEDLINE | ID: mdl-11854475

ABSTRACT

The serine protease prohormone convertase 2 (PC2), principally involved in the processing of polypeptide hormone precursors in neuroendocrine tissues, requires interaction with the neuroendocrine protein 7B2 to generate an enzymatically active form. 7B2 null mice express no PC2 activity and release large quantities of uncleaved ACTH, resulting in a lethal endocrine condition that resembles pituitary Cushing's (Westphal, C. H., Muller, L., Zhou, A., Bonner-Weir, S., Schambelan, M., Steiner, D. F., Lindberg, I. & Leder, P. (1999) Cell 96, 689). Here, we have compared the 7B2 and PC2 null mouse models to determine why the 7B2 null, but not the PC2 null, exhibits a lethal disease state. Both 7B2 and PC2 nulls contained highly elevated pituitary adrenocorticotropic hormone (ACTH); the neurointermediate lobe content of ACTH in 7B2 nulls was 13-fold higher than in WT mice; that of the PC2 null was 65-fold higher. However, circulating ACTH levels were much higher in the 7B2 null than in the PC2 null. Because hypothalamic inhibitory dopaminergic control represents the major influence on intermediate lobe proopiomelanocortin-derived peptide secretion, dopamine levels were measured, and they revealed that 7B2 null pituitaries contained only one-fourth of WT pituitary dopamine. Adrenalectomized 7B2 null animals survived past the usual time of death at 5 weeks; a month after adrenalectomy, they exhibited normal levels of pituitary dopamine, circulating ACTH, and corticosterone. Elevated corticosterone, therefore, seems to play a central role in the lethal phenotype of the 7B2 null, whereas a 7B2-mediated dopaminergic deficiency state may be involved in the actual ACTH hypersecretion phenomenon. Interestingly, adrenalectomized 7B2 nulls also developed unexpectedly severe obesity.


Subject(s)
Adrenal Glands/metabolism , Adrenocorticotropic Hormone/metabolism , Cushing Syndrome/metabolism , Dopamine/metabolism , Nerve Tissue Proteins/physiology , Pituitary Hormones/physiology , Adrenalectomy , Adrenocorticotropic Hormone/genetics , Animals , Cushing Syndrome/etiology , Cushing Syndrome/mortality , Humans , In Situ Hybridization , Mice , Mice, Inbred C57BL , Mice, Knockout , Nerve Tissue Proteins/genetics , Neuroendocrine Secretory Protein 7B2 , Phenotype , Pituitary Gland/metabolism , Pituitary Gland, Anterior/metabolism , Pituitary Hormones/genetics , Pro-Opiomelanocortin/genetics , Proprotein Convertase 2 , RNA, Messenger , Subtilisins/genetics , Subtilisins/physiology
17.
Med Sci Sports Exerc ; 33(11): 1946-52, 2001 Nov.
Article in English | MEDLINE | ID: mdl-11689748

ABSTRACT

PURPOSE: The Children's OMNI Scale of Perceived Exertion was used to identify a response normalized rating of perceived exertion (RPE)-Overall, RPE-Legs, and RPE-Chest that corresponds to the ventilatory breakpoint (Vpt) in 8- to 12-yr-old female and male children. METHODS: Subjects were a priori stratified into two fitness groups on the basis of peak oxygen uptake (VO2 peak): average (A) (41.0-49.0 mL x kg(-1) x min(-1); N = 24) and above average (AA) (50.0-58.0 mL x kg(-1) x min(-1); N = 24). Vpt was determined by a progressive cycle ergometer protocol to VO2 peak. RESULTS: A gender effect was not observed for any descriptive or dependent variable. Mean VO2peak for the A group was 1.72 L x min(-1) and for the AA group 2.04 L x min(-1). Vpt corresponded to 64.0% VO2 peak for A and 74.0% VO2peak for AA. RPE-Overall (mean A and AA, 6.1), RPE-Legs (mean A and AA, 7.2), and RPE-Chest (mean A and AA, 4.5) did not differ between the fitness groups. CONCLUSION: Findings indicated that undifferentiated and differentiated RPE-Vpt were similar between female and male children who varied in VO2peak and Vpt. A comparatively stable RPE-Vpt for 8- to 12-yr-old children that vary in VO2peak and Vpt indicates a group normalized perceptual response.


Subject(s)
Anaerobic Threshold/physiology , Exercise Test/standards , Oxygen Consumption/physiology , Physical Exertion/physiology , Analysis of Variance , Child , Female , Heart Rate , Humans , Leg/physiology , Male , Physical Fitness/physiology , Reference Values , Reproducibility of Results , Sex Factors , Thorax/physiology
18.
J Neurosci ; 21(19): 7788-92, 2001 Oct 01.
Article in English | MEDLINE | ID: mdl-11567069

ABSTRACT

Dopamine systems are intimately involved with opioid actions. Pharmacological studies suggest an important modulatory effect of dopamine and its receptors on opioid analgesia. We have now examined these interactions in a knock-out model in which the dopamine(2) (D(2)) receptor has been disrupted. Loss of D(2) receptors enhances, in a dose-dependent manner, the analgesic actions of the mu analgesic morphine, the kappa(1) agonist U50,488H and the kappa(3) analgesic naloxone benzoylhydrazone. The responses to the delta opioid analgesic [d-Pen(2),d-Pen(5)]enkephalin were unaffected in the knock-out animals. Loss of D(2) receptors also potentiated spinal orphanin FQ/nociceptin analgesia. Antisense studies using a probe targeting the D(2) receptor revealed results similar to those observed in the knock-out model. The modulatory actions of D(2) receptors were independent of final sigma receptor systems because the final sigma agonist (+)-pentazocine lowered opioid analgesia in all mice, including the D(2) knock-out group. Thus, dopamine D(2) receptors represent an additional, significant modulatory system that inhibits analgesic responses to mu and kappa opioids.


Subject(s)
Analgesia , Analgesics, Opioid/pharmacology , Receptors, Dopamine D2/deficiency , 3,4-Dichloro-N-methyl-N-(2-(1-pyrrolidinyl)-cyclohexyl)-benzeneacetamide, (trans)-Isomer/pharmacology , Animals , Dopamine Antagonists/pharmacology , Dopamine D2 Receptor Antagonists , Dose-Response Relationship, Drug , Drug Synergism , Enkephalin, D-Penicillamine (2,5)-/pharmacology , Heterozygote , Mice , Mice, Knockout , Morphine/pharmacology , Naloxone/analogs & derivatives , Naloxone/pharmacology , Oligonucleotides, Antisense/pharmacology , Opioid Peptides/pharmacology , Pain Measurement/drug effects , Pentazocine/pharmacology , Receptors, Dopamine D2/genetics , Receptors, Opioid, delta/agonists , Receptors, Opioid, kappa/agonists , Receptors, Opioid, mu/agonists , Receptors, sigma/agonists , Sulpiride/pharmacology , Nociceptin
19.
J Neurosci ; 21(16): 5864-70, 2001 Aug 15.
Article in English | MEDLINE | ID: mdl-11487609

ABSTRACT

The bioactivity of neuropeptides can be regulated by a variety of post-translational modifications, including proteolytic processing. Here, gene-targeted mice producing defective prohormone convertase 2 (PC2) were used to examine the post-translational processing of two neuroendocrine prohormones, pro-opiomelanocortin (POMC) and pro-orphanin FQ (pOFQ)/nociceptin (N), in the brain. Reversed-phase HPLC and gel-exclusion chromatography were combined with specific radioimmunoassays to analyze the processing patterns of these two prohormones in the hypothalamus and the amygdala. In the case of POMC, the lack of PC2 activity completely prevented carboxy-shortening of beta-endorphins and greatly diminished conversion of beta-lipotropin to gamma-lipotropin and beta-endorphin. Although conversion of beta-lipotropin to beta-endorphin decreased, the lack of PC2 activity caused an increase in beta-lipotropin and beta-endorphin levels in the mutant animals, but no increases in POMC or biosynthetic intermediates were seen. The extent of OFQ/N production was significantly lower in PC2-deficient mice and there was an accumulation of relatively large amounts of pOFQ/N and biosynthetic intermediates. These results demonstrate that PC2 is directly involved in the biogenesis of two brain neuropeptides in vivo and suggest that the specific prohormone and cellular context influences neuropeptide processing by PCs.


Subject(s)
Brain/metabolism , Pro-Opiomelanocortin/metabolism , Protein Precursors/metabolism , Protein Processing, Post-Translational , Receptors, Opioid/metabolism , Subtilisins/biosynthesis , Amygdala/chemistry , Amygdala/metabolism , Animals , Brain Chemistry , Chromatography, Gel , Chromatography, High Pressure Liquid , Gene Targeting , Heterozygote , Homozygote , Hypothalamus/chemistry , Hypothalamus/metabolism , Mice , Mice, Knockout , Proprotein Convertase 2 , Radioimmunoassay , Subtilisins/genetics , beta-Endorphin/biosynthesis , beta-Lipotropin/biosynthesis , beta-Lipotropin/metabolism
20.
Brain Res ; 906(1-2): 13-24, 2001 Jul 06.
Article in English | MEDLINE | ID: mdl-11430857

ABSTRACT

Until recently the opioid receptor family was thought to consist of only the mu-, delta- and kappa-receptors. The cloning of opioid receptor like receptor (ORL1) and its endogenous ligand nociceptin/orphanin FQ, which displayed anti-opioid properties, has raised the issue of functional co-operativity of this system with the classical opioid system. ORL1 receptor knockout mice have been successfully developed by homologous recombination to allow the issue of potential heterogeneity of this receptor and also of compensatory changes in mu-, delta- or kappa-receptors in the absence of ORL1 to be addressed. We have carried out quantitative autoradiographic mapping of these receptors in the brains of mice that are wild-type, heterozygous and homozygous for the deletion of the ORL1 receptor. ORL1, mu-, delta- and kappa-receptors were labelled with [(3)H] leucyl-nociceptin (0.4 nM), [(3)H] DAMGO (4 nM), [(3)H] deltorphin-I (7 nM), and [(3)H] CI-977 (2.5 nM) respectively. An approximately 50% decrease in [(3)H] leucyl-nociceptin binding was seen in heterozygous ORL1 mutant mice and there was a complete absence of binding in homozygous brains indicating the single gene encodes for the ORL1 receptor and any putative subtypes. No significant gross changes in the binding to other opioid receptors were seen across genotypes in the ORL1 mutant mice demonstrating a lack of major compensation of classical opioid receptors in the absence of ORL1. There were a small number of region specific changes in the expression of classical opioid receptors that may relate to interdependent function with ORL1.


Subject(s)
Brain/metabolism , Neurons/metabolism , Receptors, Opioid, delta/metabolism , Receptors, Opioid, kappa/metabolism , Receptors, Opioid, mu/metabolism , Receptors, Opioid/deficiency , Analgesics, Opioid/pharmacology , Animals , Autoradiography , Benzofurans/pharmacology , Binding Sites/drug effects , Binding Sites/physiology , Brain/cytology , Brain/drug effects , Enkephalin, Ala(2)-MePhe(4)-Gly(5)-/pharmacokinetics , Gene Deletion , Mice , Mice, Knockout/genetics , Mice, Knockout/metabolism , Neurons/cytology , Neurons/drug effects , Neuroprotective Agents/pharmacology , Oligopeptides/pharmacology , Opioid Peptides/pharmacokinetics , Pyrrolidines/pharmacology , Radioligand Assay , Receptors, Opioid/genetics , Receptors, Opioid, delta/drug effects , Receptors, Opioid, kappa/drug effects , Receptors, Opioid, mu/drug effects , Tritium/pharmacokinetics , Nociceptin Receptor , Nociceptin
SELECTION OF CITATIONS
SEARCH DETAIL