Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 93
Filter
1.
Eur J Neurosci ; 46(4): 2026-2034, 2017 Aug.
Article in English | MEDLINE | ID: mdl-28700108

ABSTRACT

The orphan receptor, GPR88, is emerging as a key player in the pathophysiology of several neuropsychiatric diseases, including psychotic disorders. Knockout (KO) mice lacking GPR88 throughout the brain exhibit many abnormalities relevant to schizophrenia including locomotor hyperactivity, behavioural hypersensitivity to dopaminergic psychostimulants and deficient sensorimotor gating. Here, we used conditional knockout (cKO) mice lacking GPR88 selectively in striatal medium spiny neurons expressing A2A receptor to determine neuronal circuits underlying these phenotypes. We first studied locomotor responses of A2A R-Gpr88 KO mice and their control littermates to psychotomimetic, amphetamine, and to selective D1 and D2 receptor agonists, SKF-81297 and quinpirole, respectively. To assess sensorimotor gating performance, mice were submitted to acoustic and visual prepulse inhibition (PPI) paradigms. Total knockout GPR88 mice were also studied for comparison. Like total GPR88 KO mice, A2A R-Gpr88 KO mice displayed a heightened sensitivity to locomotor stimulant effects of amphetamine and SKF-81297. They also exhibited enhanced locomotor activity to quinpirole, which tended to suppress locomotion in control mice. By contrast, they had normal acoustic and visual PPI, unlike total GPR88 KO mice that show impairments across different sensory modalities. Finally, none of the genetic manipulations altered central auditory temporal processing assessed by gap-PPI. Together, these findings support the role of GPR88 in the pathophysiology of schizophrenia and show that GPR88 in A2A receptor-expressing neurons modulates psychomotor behaviour but not sensorimotor gating.


Subject(s)
Dopamine Agonists/pharmacology , Motor Activity/physiology , Neurons/metabolism , Receptors, Adrenergic, alpha-2/biosynthesis , Receptors, G-Protein-Coupled/biosynthesis , Sensory Gating/physiology , Animals , Female , Gene Expression , Male , Mice , Mice, 129 Strain , Mice, Inbred C57BL , Mice, Knockout , Mice, Transgenic , Motor Activity/drug effects , Neurons/drug effects , Receptors, Adrenergic, alpha-2/genetics , Receptors, G-Protein-Coupled/genetics , Reflex, Startle/drug effects , Reflex, Startle/physiology , Sensory Gating/drug effects
2.
Biomed Res Int ; 2017: 6352159, 2017.
Article in English | MEDLINE | ID: mdl-28286770

ABSTRACT

Background. Neuroinflammation which presents as a possible mechanism of delirium is associated with MCP-1, an important proinflammatory factor which is expressed on astrocytes. It is known that dexmedetomidine (DEX) possesses potent anti-inflammatory properties. This study aimed to investigate the potential effects of DEX on the production of MCP-1 in lipopolysaccharide-stimulated astrocytes. Materials and Methods. Astrocytes were treated with LPS (10 ng/ml, 50 ng/ml, 100 ng/ml, and 1000 ng/ml), DEX (500 ng/mL), LPS (100 ng/ml), and DEX (10, 100, and 500 ng/mL) for a duration of three hours; expression levels of MCP-1 were measured by real-time PCR. The double immunofluorescence staining protocol was utilized to determine the expression of α2-adrenoceptors (α2AR) and glial fibrillary acidic protein (GFAP) on astrocytes. Results. Expressions of MCP-1 mRNA in astrocytes were induced dose-dependently by LPS. Administration of DEX significantly inhibited the expression of MCP-1 mRNA (P < 0.001). Double immunofluorescence assay showed that α2AR colocalize with GFAP, which indicates the expression of α2-adrenoceptors in astrocytes. Conclusions. DEX is a potent suppressor of MCP-1 in astrocytes induced with lipopolysaccharide through α2A-adrenergic receptors, which potentially explains its beneficial effects in the treatment of delirium by attenuating neuroinflammation.


Subject(s)
Astrocytes/metabolism , Chemokine CCL2/biosynthesis , Dexmedetomidine/pharmacology , Gene Expression Regulation/drug effects , Lipopolysaccharides/toxicity , Animals , Astrocytes/pathology , Cells, Cultured , Glial Fibrillary Acidic Protein/biosynthesis , Rats , Rats, Sprague-Dawley , Receptors, Adrenergic, alpha-2/biosynthesis
3.
Sci Rep ; 6: 37921, 2016 11 30.
Article in English | MEDLINE | ID: mdl-27901063

ABSTRACT

The molecular mechanisms that control the targeting of newly synthesized G protein-coupled receptors (GPCRs) to the functional destinations remain poorly elucidated. Here, we have determined the role of Golgi-localized, γ-adaptin ear domain homology, ADP ribosylation factor-binding proteins 1 and 2 (GGA1 and GGA2) in the cell surface transport of α2B-adrenergic receptor (α2B-AR), a prototypic GPCR, and studied the underlying mechanisms. We demonstrated that knockdown of GGA1 and GGA2 by shRNA and siRNA significantly reduced the cell surface expression of inducibly expressed α2B-AR and arrested the receptor in the perinuclear region. Knockdown of each GGA markedly inhibited the dendritic expression of α2B-AR in primary cortical neurons. Consistently, depleting GGA1 and GGA2 attenuated receptor-mediated signal transduction measured as ERK1/2 activation and cAMP inhibition. Although full length α2B-AR associated with GGA2 but not GGA1, its third intracellular loop was found to directly interact with both GGA1 and GGA2. More interestingly, further mapping of interaction domains showed that the GGA1 hinge region and the GGA2 GAE domain bound to multiple subdomains of the loop. These studies have identified an important function and revealed novel mechanisms of the GGA family proteins in the forward trafficking of a cell surface GPCR.


Subject(s)
Adaptor Proteins, Vesicular Transport/metabolism , Gene Expression Regulation/physiology , Receptors, Adrenergic, alpha-2/biosynthesis , Adaptor Proteins, Vesicular Transport/genetics , Cyclic AMP/genetics , Cyclic AMP/metabolism , HEK293 Cells , HeLa Cells , Humans , MAP Kinase Signaling System/physiology , Mitogen-Activated Protein Kinase 1/genetics , Mitogen-Activated Protein Kinase 1/metabolism , Mitogen-Activated Protein Kinase 3/genetics , Mitogen-Activated Protein Kinase 3/metabolism , Protein Domains , Protein Structure, Secondary , Protein Transport/physiology , Receptors, Adrenergic, alpha-2/genetics
4.
J Immunol ; 193(6): 3023-35, 2014 Sep 15.
Article in English | MEDLINE | ID: mdl-25114107

ABSTRACT

Adrenergic receptors are expressed on the surface of inflammation-mediating cells, but their potential role in the regulation of the inflammatory response is still poorly understood. The objectives of this work were to study the effects of α2-adrenergic agonists on the inflammatory response in vivo and to determine their mechanism of action. In two mouse models of inflammation, zymosan air pouch and thioglycolate-induced peritonitis models, the i.m. treatment with xylazine or UK14304, two α2-adrenergic agonists, reduced neutrophil migration by 60%. The α2-adrenergic antagonist RX821002 abrogated this effect. In flow cytometry experiments, the basal surface expression of L-selectin and CD11b was modified neither in murine nor in human neutrophils upon α2-agonist treatment. Similar experiments in HUVEC showed that UK14304 prevented the activation-dependent upregulation of ICAM-1. In contrast, UK14304 augmented electrical resistance and reduced macromolecular transport through a confluent HUVEC monolayer. In flow chamber experiments, under postcapillary venule-like flow conditions, the pretreatment of HUVECs, but not neutrophils, with α2-agonists decreased transendothelial migration, without affecting neutrophil rolling. Interestingly, α2-agonists prevented the TNF-α-mediated decrease in expression of the adherens junctional molecules, VE-cadherin, ß-catenin, and plakoglobin, and reduced the ICAM-1-mediated phosphorylation of VE-cadherin by immunofluorescence and confocal analysis and Western blot analysis, respectively. These findings indicate that α2-adrenoceptors trigger signals that protect the integrity of endothelial adherens junctions during the inflammatory response, thus pointing at the vascular endothelium as a therapeutic target for the management of inflammatory processes in humans.


Subject(s)
Adherens Junctions/immunology , Endothelium, Vascular/immunology , Neutrophils/immunology , Receptors, Adrenergic, alpha-2/immunology , Adrenergic alpha-2 Receptor Agonists/pharmacology , Adrenergic alpha-Antagonists/pharmacology , Animals , Antigens, CD/biosynthesis , Brimonidine Tartrate , CD11b Antigen/biosynthesis , Cadherins/biosynthesis , Humans , Idazoxan/analogs & derivatives , Idazoxan/pharmacology , Inflammation/immunology , Intercellular Adhesion Molecule-1/biosynthesis , L-Selectin/biosynthesis , Male , Mice , Peritonitis/chemically induced , Quinoxalines/pharmacology , Receptors, Adrenergic, alpha-2/biosynthesis , Thioglycolates/pharmacology , Transendothelial and Transepithelial Migration/drug effects , Transendothelial and Transepithelial Migration/immunology , Tumor Necrosis Factor-alpha/immunology , Up-Regulation/drug effects , Xylazine/pharmacology , Zymosan/pharmacology , beta Catenin/biosynthesis , gamma Catenin/biosynthesis
5.
Invest Ophthalmol Vis Sci ; 55(6): 3385-94, 2014 Apr 29.
Article in English | MEDLINE | ID: mdl-24781942

ABSTRACT

PURPOSE: Alpha2-adrenergic receptor agonists are used in glaucoma treatment and have been shown to have some neuroprotective effects. We performed this study to test the hypothesis that epidermal growth factor receptors on chicken Müller cells are transactivated by α2-adrenergic receptors and we focused on the extracellular signal-activated kinases 1/2 (ERK) pathway. METHODS: Embryonic chicken retina and cultures of primary Müller cells were stimulated by α2-adrenergic receptor agonist brimonidine. Immunostaining, quantitative RT-PCR, and Western blot techniques, in combination with Src, epidermal growth factor receptor kinase, and matrix metalloproteinase inhibitors, were used for analysis of the cellular responses. RESULTS: Our results showed that Müller cells express α2A-adrenergic receptors in vivo and in vitro and that brimonidine triggered a robust and transient phosphorylation of ERK1/2. This ERK-response was Src-kinase dependent, associated with tyrosine phosphorylation of epidermal growth factor receptors (phospho-Y1068, Y1173) and was mediated by matrix metalloproteinase activity on the Müller cells. CONCLUSIONS: Müller cells express the α2A-adrenergic receptor, and brimonidine triggers both Src-kinase- and matrix metalloproteinase-mediated autocrine ligand-dependent activation of epidermal growth factor receptors on Müller cells. This response is consistent with transactivation of epidermal growth factor receptors by stimulation of α2-adrenergic receptors.


Subject(s)
Ependymoglial Cells/metabolism , ErbB Receptors/genetics , Glaucoma/genetics , Quinoxalines/pharmacology , RNA/genetics , Retina/metabolism , Transcriptional Activation , Adrenergic alpha-2 Receptor Agonists/pharmacology , Animals , Blotting, Western , Brimonidine Tartrate , Cells, Cultured , Chick Embryo , Ependymoglial Cells/drug effects , Ependymoglial Cells/pathology , ErbB Receptors/biosynthesis , ErbB Receptors/drug effects , Glaucoma/metabolism , Glaucoma/pathology , Immunohistochemistry , MAP Kinase Signaling System/genetics , Real-Time Polymerase Chain Reaction , Receptors, Adrenergic, alpha-2/biosynthesis , Receptors, Adrenergic, alpha-2/drug effects , Receptors, Adrenergic, alpha-2/genetics , Retina/drug effects , Retina/pathology
6.
J Clin Invest ; 123(12): 5119-34, 2013 Dec.
Article in English | MEDLINE | ID: mdl-24231350

ABSTRACT

A systems pharmacological approach that capitalizes on the characterization of intracellular signaling networks can transform our understanding of human diseases and lead to therapy development. Here, we applied this strategy to identify pharmacological targets for the treatment of Stargardt disease, a severe juvenile form of macular degeneration. Diverse GPCRs have previously been implicated in neuronal cell survival, and crosstalk between GPCR signaling pathways represents an unexplored avenue for pharmacological intervention. We focused on this receptor family for potential therapeutic interventions in macular disease. Complete transcriptomes of mouse and human samples were analyzed to assess the expression of GPCRs in the retina. Focusing on adrenergic (AR) and serotonin (5-HT) receptors, we found that adrenoceptor α 2C (Adra2c) and serotonin receptor 2a (Htr2a) were the most highly expressed. Using a mouse model of Stargardt disease, we found that pharmacological interventions that targeted both GPCR signaling pathways and adenylate cyclases (ACs) improved photoreceptor cell survival, preserved photoreceptor function, and attenuated the accumulation of pathological fluorescent deposits in the retina. These findings demonstrate a strategy for the identification of new drug candidates and FDA-approved drugs for the treatment of monogenic and complex diseases.


Subject(s)
Adenylyl Cyclase Inhibitors , Adrenergic alpha-Agonists/therapeutic use , Adrenergic alpha-Antagonists/therapeutic use , Macular Degeneration/congenital , Molecular Targeted Therapy , Nerve Tissue Proteins/biosynthesis , Photoreceptor Cells, Vertebrate/drug effects , Receptor, Serotonin, 5-HT2A/biosynthesis , Receptors, Adrenergic, alpha-2/biosynthesis , Serotonin Antagonists/therapeutic use , ATP-Binding Cassette Transporters/deficiency , ATP-Binding Cassette Transporters/genetics , Adenine/analogs & derivatives , Adenine/pharmacology , Adenine/therapeutic use , Adrenergic alpha-Agonists/pharmacology , Adrenergic alpha-Antagonists/pharmacology , Alcohol Oxidoreductases/deficiency , Alcohol Oxidoreductases/genetics , Animals , Cell Survival , Disease Models, Animal , Doxazosin/pharmacology , Doxazosin/therapeutic use , Drug Evaluation, Preclinical , Guanabenz/pharmacology , Guanabenz/therapeutic use , Humans , Light/adverse effects , Macaca fascicularis , Macular Degeneration/drug therapy , Macular Degeneration/genetics , Macular Degeneration/prevention & control , Mice , Mice, Inbred BALB C , Mice, Knockout , Nerve Tissue Proteins/genetics , Photoreceptor Cells, Vertebrate/pathology , Photoreceptor Cells, Vertebrate/physiology , Photoreceptor Cells, Vertebrate/radiation effects , Reactive Oxygen Species , Receptor, Serotonin, 5-HT2A/genetics , Receptors, Adrenergic, alpha-2/genetics , Receptors, G-Protein-Coupled/biosynthesis , Receptors, G-Protein-Coupled/genetics , Serotonin Antagonists/pharmacology , Signal Transduction , Stargardt Disease
7.
Brain Res ; 1531: 9-16, 2013 Sep 19.
Article in English | MEDLINE | ID: mdl-23916734

ABSTRACT

Sleep responses to chronic sleep restriction may be very different from those observed after acute total sleep deprivation. Specifically, when sleep restriction is repeated for several consecutive days, animals express attenuated compensatory increases in sleep time and intensity during daily sleep opportunities. The neurobiological mechanisms underlying these adaptive, or more specifically, allostatic, changes in sleep homeostasis are unknown. Several lines of evidence indicate that norepinephrine may play a key role in modulating arousal states and NREM EEG delta power, which is widely recognized as a marker for sleep intensity. Therefore, we investigated time course changes in brain adrenergic receptor mRNA levels in response to chronic sleep restriction using a rat model. Here, we observed that significantly altered mRNA levels of the α1- adrenergic receptor in the basal forebrain as well as α2- and ß1-adrenergic receptor in the anterior cingulate cortex only on the first sleep restriction day. On the other hand, the frontal cortex α1-, α2-, and ß1-adrenergic receptor mRNA levels were reduced throughout the period of sleep restriction. Combined with our earlier findings on EEG that sleep time and intensity significantly increased only on the first sleep restriction days, these results suggest that alterations in the brain norepinephrine system in the basal forebrain and cingulate cortex may mediate allostatic changes in sleep time and intensity observed during chronic sleep restriction.


Subject(s)
Allostasis/physiology , Norepinephrine/physiology , Receptors, Adrenergic, alpha-1/physiology , Receptors, Adrenergic, alpha-2/physiology , Receptors, Adrenergic, beta-1/physiology , Sleep Deprivation/physiopathology , Animals , Brain/metabolism , Brain/physiology , Electroencephalography/methods , Male , RNA, Messenger/biosynthesis , Rats , Rats, Sprague-Dawley , Receptors, Adrenergic, alpha-1/biosynthesis , Receptors, Adrenergic, alpha-2/biosynthesis , Receptors, Adrenergic, beta-1/biosynthesis , Sleep Deprivation/metabolism , Time Factors
8.
J Pharmacol Exp Ther ; 344(2): 522-30, 2013 Feb.
Article in English | MEDLINE | ID: mdl-23230215

ABSTRACT

The role of dynorphin A (1-17; Dyn) and its associated kappa opioid receptor (KOR) in nociception represents a longstanding scientific conundrum: Dyn and KOR (Dyn/KOR) have variously been reported to inhibit, facilitate, or have no effect on pain. We investigated whether interactions between sex and pain type (which are usually ignored) influenced Dyn/KOR-mediated antinociception. Blockade of the spinal α(2)-noradrenergic receptor (α(2)-NAR) using yohimbine elicited comparable spinal Dyn release in females and males. Nevertheless, the yohimbine-induced antinociception exhibited sexual dimorphism that depended on the pain test used: in the intraperitoneal acetic acid-induced writhing test, yohimbine produced antinociception only in females, whereas in the intraplantar formalin-induced paw flinch test, antinociception was observed only in males. In females and males, both intrathecal Dyn antibodies and spinal KOR blockade eliminated the yohimbine-induced antinociception, indicating that Dyn/KOR mediated it. However, despite the conditional nature of spinal Dyn/KOR-mediated yohimbine antinociception, both intraplantar formalin and intraperitoneal acetic acid activated spinal Dyn neurons that expressed α(2)-NARs. Moreover, Dyn terminals apposed KOR-expressing spinal nociceptive neurons in both sexes. This similar organization suggests that the sexually dimorphic interdependent effects of sex and pain type may result from the presence of nonfunctional (silent) KORs on nociceptive spinal neurons that are responsive to intraplantar formalin (in females) versus intraperitoneal acetic acid (in males). Our findings that spinal Dyn/KOR-mediated antinociception depends on interactions between sex and pain type underscore the importance of using both sexes and multiple pain models when investigating Dyn/KOR antinociception.


Subject(s)
Dynorphins/metabolism , Nociceptive Pain/metabolism , Pain Measurement , Receptors, Adrenergic, alpha-2/metabolism , Receptors, Opioid, kappa/metabolism , Sex Characteristics , Adrenergic alpha-2 Receptor Antagonists/pharmacology , Animals , Dynorphins/biosynthesis , Female , Injections, Spinal , Male , Motor Neurons/drug effects , Motor Neurons/metabolism , Nociceptive Pain/drug therapy , Nociceptive Pain/etiology , Pain Measurement/drug effects , Rats , Rats, Sprague-Dawley , Receptors, Adrenergic, alpha-2/biosynthesis , Receptors, Opioid, kappa/biosynthesis , Spinal Cord/drug effects , Spinal Cord/metabolism , Yohimbine/pharmacology
9.
J Appl Physiol (1985) ; 112(5): 841-8, 2012 Mar.
Article in English | MEDLINE | ID: mdl-22194325

ABSTRACT

We tested the hypothesis that adrenergic and nonadrenergic receptor responsiveness and protein expression would be altered with advancing age. Young (n = 6; 22 ± 1 mo; mean ± SE) and old (n = 6; 118 ± 9 mo) beagles were instrumented with flow probes and an indwelling catheter for continuous measurement of external iliac blood flow and arterial blood pressure. Vascular conductance (VC) was calculated as hindlimb blood flow/mean arterial pressure. Selective agonists for α-1, α-2, neuropeptide-Y (NPY), and purinergic (P2X) receptors were infused at rest and during treadmill running at moderate (2.5 mph) and heavy (4 mph with 2.5% grade) exercise intensities. Feed arteries were dissected from gracilis muscles, and α-1D, α-1B, α-2A, P2X-4, P2X-1, and NPY-Y1 receptor protein expression was determined. Phenylephrine produced similar decreases (P > 0.05) in VC in young and old beagles at rest (young: -62 ± 5%; old: -59 ± 5%) and during moderate (young: -67 ± 5%; old: -62 ± 4%) and heavy (young: -54 ± 4%; old: -49 ± 3%) exercise. Clonidine caused similar (P > 0.05) decreases in VC in old compared with young dogs at rest (young: -59 ± 8%; old: -70 ± 6%) and during moderate (young: -52 ± 6%; old: -47 ± 5%)- and heavy (young: -42 ± 5%; old: -43 ± 5%)-intensity exercise. NPY infusion resulted in a similar decline in VC in young and old beagles at rest (young: -40 ± 7%; old: -39 ± 9%) and during moderate (young: -47 ± 6%; old: -40 ± 6%)- and heavy (young: -40 ± 3%; old: -38 ± 4%)-intensity exercise. α-ß-Methylene-ATP also produced similar decreases in VC in young and old beagles at rest (young: -36 ± 6%; old: -40 ± 8%) and during exercise at moderate (young: -42 ± 5%; old: -40 ± 9%) and heavy (young: -47 ± 5%; old: -42 ± 8%) intensities. α-1B receptor protein expression was elevated (P < 0.05) in old compared with young dogs, whereas there were no age-related differences in α-1D or α-2A receptor expression and nonadrenergic P2X-4, P2X-1, and NPY-Y1 receptor expression. The present findings indicate that postsynaptic adrenergic and nonadrenergic receptor responsiveness was not altered by advancing age. Moreover, the expression of adrenergic and nonadrenergic receptors in skeletal-muscle feed arteries was largely unaffected by aging.


Subject(s)
Aging/physiology , Muscle, Skeletal/physiology , Receptors, Adrenergic, alpha-1/biosynthesis , Receptors, Adrenergic, alpha-2/biosynthesis , Receptors, Neuropeptide Y/metabolism , Receptors, Purinergic P2X/biosynthesis , Adrenergic alpha-1 Receptor Agonists/pharmacology , Adrenergic alpha-2 Receptor Agonists/pharmacology , Aging/metabolism , Animals , Arteries/drug effects , Arteries/metabolism , Arteries/physiology , Blood Pressure/drug effects , Blood Pressure/physiology , Clonidine/pharmacology , Dogs , Hemodynamics/drug effects , Hemodynamics/physiology , Hindlimb/blood supply , Hindlimb/drug effects , Hindlimb/metabolism , Muscle, Skeletal/drug effects , Muscle, Skeletal/metabolism , Norepinephrine/pharmacology , Phenylephrine/pharmacology , Physical Conditioning, Animal/physiology , Purinergic P2X Receptor Agonists/pharmacology , Receptors, Adrenergic, alpha-1/genetics , Receptors, Adrenergic, alpha-1/metabolism , Receptors, Adrenergic, alpha-2/genetics , Receptors, Adrenergic, alpha-2/metabolism , Receptors, Neuropeptide Y/agonists , Receptors, Neuropeptide Y/genetics , Receptors, Purinergic P2X/genetics , Receptors, Purinergic P2X/metabolism , Regional Blood Flow/drug effects , Regional Blood Flow/physiology , Rest/physiology
10.
Psychopharmacology (Berl) ; 221(1): 53-65, 2012 May.
Article in English | MEDLINE | ID: mdl-22038538

ABSTRACT

RATIONALE: Peripheral neuropathic pain is a chronic condition that may produce plastic changes in several brain regions. The noradrenergic locus coeruleus (LC) is a crucial component of ascending and descending pain pathways, both of which are frequently compromised after nerve injury. OBJECTIVES: The objective of the study was to examine whether chronic constriction injury (CCI), a model of neuropathic pain, alters noradrenergic activity in the rat LC. METHODS: Activity in the LC was assessed by electrophysiology and microdialysis, while protein expression was monitored in western blots and by immunohistochemistry. RESULTS: The pain threshold had dropped in injured rats 7 days after inducing neuropathy. While alpha-2-adrenoceptors mediate activity in the LC and in its terminal areas, no alterations in either spontaneous neuronal activity or extracellular noradrenaline levels were observed following CCI. Moreover, alpha-2-adrenoceptor activity in the LC of CCI rats remained unchanged after systemic administration of UK14,304, RX821002 or desipramine. Accordingly, extracellular noradrenaline levels in the LC were similar in CCI and control animals following local administration of clonidine or RX821002. In addition, there were no changes in the expression of the alpha-2-adrenoceptors, Gαi/z subunits or the regulators of G-protein signaling. However, pERK1/2 (phosphorylated extracellular signal-regulated kinases 1/2) expression augmented in the spinal cord, paragigantocellularis nucleus (PGi) and dorsal raphe nucleus (DRN) following CCI. CONCLUSIONS: Neuropathic pain is not accompanied by modifications in tonic LC activity after the onset of pain. This may indicate that the signals from the PGi and DRN, the excitatory and inhibitory afferents of the LC, cancel one another out.


Subject(s)
Locus Coeruleus/physiology , Neuralgia/physiopathology , Receptors, Adrenergic, alpha-2/physiology , Action Potentials/physiology , Adrenergic Uptake Inhibitors/pharmacology , Adrenergic alpha-2 Receptor Agonists/pharmacology , Adrenergic alpha-Antagonists/pharmacology , Animals , Brimonidine Tartrate , Clonidine/pharmacology , Desipramine/pharmacology , Disease Models, Animal , Idazoxan/analogs & derivatives , Idazoxan/pharmacology , Locus Coeruleus/drug effects , Locus Coeruleus/metabolism , MAP Kinase Signaling System/physiology , Male , Medulla Oblongata/drug effects , Medulla Oblongata/metabolism , Norepinephrine/metabolism , Prefrontal Cortex/metabolism , Quinoxalines/pharmacology , Raphe Nuclei/drug effects , Raphe Nuclei/metabolism , Rats , Rats, Sprague-Dawley , Receptors, Adrenergic, alpha-2/biosynthesis , Signal Transduction/drug effects , Signal Transduction/physiology , Spinal Cord/drug effects , Spinal Cord/metabolism
11.
Mol Cell Biochem ; 362(1-2): 103-14, 2012 Mar.
Article in English | MEDLINE | ID: mdl-22038629

ABSTRACT

Hypertension is sexually dimorphic and modified by removal of endogenous sex steroids. This study tested the hypothesis that endogenous gonadal hormones exert differential effects on protein expression in the kidney and mesentery of SHR. At ~5 weeks of age male and female SHR underwent sham operation, orchidectomy, or ovariectomy (OVX). At 20-23 weeks of age, mean arterial pressure (MAP) was measured in conscious rats. The mesenteric arterial tree and kidneys were collected, processed for Western blots, and probed for Cu Zn superoxide dismutase (SOD1), soluble epoxide hydrolase (sEH), and Alpha 2A adrenergic receptor (A2AR) expression. MAP was unaffected by ovariectomy (Sham 164 ± 4: Ovariecttomy 159 ± 3 mm Hg). MAP was reduced by orchidectomy (Sham 189 ± 5:Orchidectomy 167 ± 2 mm Hg). In mesenteric artery, SOD1 expression was greater in male versus female SHR. Orchidectomy increased while ovariectomy decreased SOD1 expression. The kidney exhibited a different pattern of response. SOD1 expression was reduced in male compared to female SHR but gonadectomy had no effect. sEH expression was not significantly different among the groups in mesenteric artery. In kidney, sEH expression was greater in males compared to females. Ovariectomy but not orchidectomy increased sEH expression. A2AR expression was greater in female than male SHR in mesentery artery and kidney. Gonadectomy had no effect in either tissue. We conclude that sexually dimorphic hypertension is associated with regionally specific changes in expression of three key proteins involved in blood pressure control. These data suggest that broad spectrum inhibition or stimulation of these systems may not be the best approach for hypertension treatment. Instead regionally targeted manipulation of these systems should be investigated.


Subject(s)
Blood Pressure/physiology , Gonadal Hormones/metabolism , Hypertension/physiopathology , Animals , Epoxide Hydrolases/biosynthesis , Female , Humans , Hypertension/genetics , Hypertension/metabolism , Kidney/metabolism , Male , Mesenteric Arteries/metabolism , Orchiectomy , Ovariectomy , RNA, Messenger/genetics , RNA, Messenger/metabolism , Rats , Rats, Inbred SHR , Rats, Sprague-Dawley , Receptors, Adrenergic, alpha-2/biosynthesis , Sex Characteristics , Superoxide Dismutase/biosynthesis , Superoxide Dismutase-1
12.
Genetika ; 48(11): 1328-35, 2012 Nov.
Article in Russian | MEDLINE | ID: mdl-23297488

ABSTRACT

Using a large amount of breeding material, the idea of D. K. Belyaev on the role of selection in the appearance of new behavioral and neuronal forms was confirmed. Experiments were performed using rats of the GC (genetics + catatonia) strain, which are prone to passive defensive reactions of cataleptic freezing. At the current breeding stage, elevation of the proportion of so-called nervous animals was demonstrated, both with respect to the expression of such reactions and their frequency. At this breeding stage, in the brains of GC rats, the mRNA levels of alpha1A- and alpha2A-adrenoreceptor genes were determined. A decrease of alpha1A-adrenoreceptor gene expression in the midbrain and medulla oblongata, along with elevation of alpha2A-adrenoreceptor gene expression in the frontal cortex was observed. It was suggested that changes in the expression of alpha-adrenoreceptor genes could be caused by an increase in the proportion of nervous animals and could contribute to the akinetic behavioral component in GC rats.


Subject(s)
Behavior, Animal , Brain/metabolism , Catatonia/metabolism , Gene Expression Regulation , Receptors, Adrenergic, alpha-1/biosynthesis , Receptors, Adrenergic, alpha-2/biosynthesis , Animals , Brain/physiopathology , Breeding , Catatonia/immunology , Catatonia/physiopathology , Disease Models, Animal , RNA, Messenger/biosynthesis , RNA, Messenger/genetics , Rats , Rats, Inbred Strains , Receptors, Adrenergic, alpha-1/genetics , Receptors, Adrenergic, alpha-2/genetics
13.
J Biol Chem ; 286(41): 36063-36075, 2011 Oct 14.
Article in English | MEDLINE | ID: mdl-21859713

ABSTRACT

The neurobiological mechanisms of action underlying antidepressant drugs remain poorly understood. Desipramine (DMI) is an antidepressant classically characterized as an inhibitor of norepinephrine reuptake. Available evidence, however, suggests a mechanism more complex than simple reuptake inhibition. In the present study, we have characterized the direct interaction between DMI and the α(2A)-adrenergic receptor (α(2A)AR), a key regulator of noradrenergic neurotransmission with altered expression and function in depression. DMI alone was found to be sufficient to drive receptor internalization acutely and a robust down-regulation of α(2A)AR expression and signaling following prolonged stimulation in vitro. These effects are achieved through arrestin-biased regulation of the receptor, as DMI selectively induces recruitment of arrestin but not activation of heterotrimeric G proteins. Meanwhile, a physiologically relevant concentration of endogenous agonist (norepinephrine) was unable to sustain a down-regulation response. Prolonged in vivo administration of DMI resulted in significant down-regulation of synaptic α(2A)AR expression, a response that was lost in arrestin3-null animals. We contend that direct DMI-driven arrestin-mediated α(2A)AR down-regulation accounts for the therapeutically desirable but mechanistically unexplained adaptive alterations in receptor expression associated with this antidepressant. Our results provide novel insight into both the pharmacology of this antidepressant drug and the targeting of the α(2A)AR in depression.


Subject(s)
Antidepressive Agents, Tricyclic/pharmacology , Arrestins/metabolism , Desipramine/pharmacology , Down-Regulation/drug effects , Receptors, Adrenergic, alpha-2/biosynthesis , Synaptic Transmission/drug effects , Animals , Arrestins/genetics , Depression/drug therapy , Depression/genetics , Depression/metabolism , Down-Regulation/genetics , Drug Delivery Systems , HEK293 Cells , Heterotrimeric GTP-Binding Proteins/genetics , Heterotrimeric GTP-Binding Proteins/metabolism , Humans , Mice , Mice, Knockout , Synaptic Transmission/genetics
14.
J Pharmacol Exp Ther ; 337(1): 285-92, 2011 Apr.
Article in English | MEDLINE | ID: mdl-21248068

ABSTRACT

Diabetic neuropathy is a common cause of chronic pain that is not adequately relieved by conventional analgesics. The α(2)-adrenoceptors are involved in the regulation of glutamatergic input and nociceptive transmission in the spinal dorsal horn, but their functional changes in diabetic neuropathy are not clear. The purpose of the present study was to determine the plasticity of presynaptic and postsynaptic α(2)-adrenoceptors in the control of spinal glutamatergic synaptic transmission in painful diabetic neuropathy. Whole-cell voltage-clamp recordings of lamina II neurons were performed in spinal cord slices from streptozotocin-induced diabetic rats. The amplitude of glutamatergic excitatory postsynaptic currents (EPSCs) evoked from the dorsal root and the frequency of spontaneous EPSCs (sEPSCs) were significantly higher in diabetic than vehicle-control rats. The specific α(2)-adrenoceptor agonist 5-bromo-6-(2-imidazolin-2-ylamino)quinoxaline (UK-14304) (0.1-2 µM) inhibited the frequency of sEPSCs more in diabetic than vehicle-treated rats. UK-14304 also inhibited the amplitude of evoked monosynaptic and polysynaptic EPSCs more in diabetic than control rats. Furthermore, the amplitude of postsynaptic G protein-coupled inwardly rectifying K(+) channel (GIRK) currents elicited by UK-14304 was significantly larger in the diabetic group than in the control group. In addition, intrathecal administration of UK-14304 increased the nociceptive threshold more in diabetic than vehicle-control rats. Our findings suggest that diabetic neuropathy increases the activity of presynaptic and postsynaptic α(2)-adrenoceptors to attenuate glutamatergic transmission in the spinal dorsal horn, which accounts for the potentiated antinociceptive effect of α(2)-adrenoceptor activation in diabetic neuropathic pain.


Subject(s)
Diabetic Neuropathies/metabolism , Excitatory Postsynaptic Potentials/physiology , Pain/metabolism , Posterior Horn Cells/metabolism , Presynaptic Terminals/metabolism , Receptors, Adrenergic, alpha-2/biosynthesis , Up-Regulation/physiology , Animals , Diabetic Neuropathies/complications , Male , Pain/etiology , Pain Measurement/methods , Rats , Rats, Sprague-Dawley
15.
Br J Anaesth ; 106(3): 371-9, 2011 Mar.
Article in English | MEDLINE | ID: mdl-21169610

ABSTRACT

BACKGROUND: We defined the nature of the pharmacological interaction after intrathecal co-administration of ginsenosides with clonidine, and clarified the contribution of the α-2 adrenoceptors on the effect of ginsenosides. METHODS: Pain was evoked by injection of a formalin solution (5%, 50 µl) into the hindpaw of male Sprague-Dawley rats. Isobolographic analysis was performed to characterize the drug interaction between ginsenosides and clonidine. The antagonism of ginsenosides-mediated antinociception was determined with α-2A (BRL 44408), α-2B (ARC 239), and α-2C (JP 1302) adrenoceptor antagonists. The expression of α-2 adrenoceptor subtypes was examined by reverse transcriptase-polymerase chain reaction. RESULTS: Intrathecal ginsenosides (n=29) and clonidine (n=31) displayed an antinociceptive effect. The ED(50) values (95% confidence intervals) of ginsenosides and clonidine for phases 1 and 2 were 109.5 (63-190.3) and 110.9 (57.1-215.5), and 11.8 (3.7-37.1) and 4.9 (3.1-6.7) µg, respectively. With an isobolographic study (n=48), the ED(50) values (95% confidence intervals) of ginsenosides in the combination of ginsenosides and clonidine for phases 1 and 2 were 58.2 (38.9-87.3) and 57.2 (46.5-70.3) µg, respectively. Intrathecal BRL 44408 (n=6), ARC 239 (n=5), and JP 1302 (n=5) reversed the antinociception of ginsenosides in both phases (P<0.01, <0.001). The injection of formalin increased the expression of α-2C adrenoceptor in the spinal cord (P<0.05). CONCLUSIONS: Intrathecal ginsenosides additively interacted with clonidine in the formalin test. Furthermore, α-2A, -B, and -C adrenoceptors contributed to the antinociception of intrathecal ginsenosides.


Subject(s)
Analgesics, Non-Narcotic/pharmacology , Clonidine/pharmacology , Ginsenosides/pharmacology , Pain/prevention & control , Receptors, Adrenergic, alpha-2/drug effects , Analgesics, Non-Narcotic/administration & dosage , Analgesics, Non-Narcotic/therapeutic use , Animals , Behavior, Animal/drug effects , Clonidine/administration & dosage , Clonidine/therapeutic use , Dose-Response Relationship, Drug , Drug Evaluation, Preclinical/methods , Drug Interactions , Gene Expression Regulation/drug effects , Ginsenosides/administration & dosage , Ginsenosides/therapeutic use , Injections, Spinal , Male , Pain/chemically induced , Pain Measurement/methods , RNA, Messenger/genetics , Rats , Rats, Sprague-Dawley , Receptors, Adrenergic, alpha-2/biosynthesis , Receptors, Adrenergic, alpha-2/genetics
16.
Br J Pharmacol ; 160(7): 1727-40, 2010 Aug.
Article in English | MEDLINE | ID: mdl-20649575

ABSTRACT

BACKGROUND AND PURPOSE: We have investigated the distribution of alpha-adrenoceptors in sheep internal anal sphincter (IAS), as a model for the human tissue, and evaluated various imidazoline derivatives for potential treatment of faecal incontinence. EXPERIMENTAL APPROACH: Saturation and competition binding with (3)H-prazosin and (3)H-RX821002 were used to confirm the presence and density of alpha-adrenoceptors in sheep IAS, and the affinity of imidazoline compounds at these receptors. A combination of in vitro receptor autoradiography and immunohistochemistry was used to investigate the regional distribution of binding sites. Contractile activity of imidazoline-based compounds on sheep IAS was assessed by isometric tension recording. KEY RESULTS: Saturation binding confirmed the presence of both alpha(1)- and alpha(2)-adrenoceptors, and subsequent characterization with sub-type-selective agents, identified them as alpha(1A)- and alpha(2D)-adrenoceptor sub-types. Autoradiographic studies with (3)H-prazosin showed a positive association of alpha(1)-adrenoceptors with immunohistochemically identified smooth muscle fibres. Anti-alpha(1)-adrenoceptor immunohistochemistry revealed similar distributions of the receptor in sheep and human IAS. The imidazoline compounds caused concentration-dependent contractions of the anal sphincter, but the maximum responses were less than those elicited by l-erythro-methoxamine, a standard non-imidazoline alpha(1)-adrenoceptor agonist. Prazosin (selective alpha(1)-adrenoceptor antagonist) significantly reduced the magnitude of contraction to l-erythro-methoxamine at the highest concentration used. Both prazosin and RX811059 (a selective alpha(2)-adrenoceptor antagonist) reduced the potency (pEC(50)) of clonidine. CONCLUSIONS AND IMPLICATIONS: This study shows that both alpha(1)- and alpha(2)-adrenoceptors are expressed in the sheep IAS, and contribute (perhaps synergistically) to contractions elicited by various imidazoline derivatives. These agents may prove useful in the treatment of faecal incontinence.


Subject(s)
Anal Canal/metabolism , Receptors, Adrenergic, alpha-1/biosynthesis , Receptors, Adrenergic, alpha-2/biosynthesis , Sheep , Adrenergic alpha-1 Receptor Antagonists , Adrenergic alpha-2 Receptor Antagonists , Adrenergic alpha-Agonists/pharmacology , Adrenergic alpha-Antagonists/pharmacology , Anal Canal/drug effects , Animals , Antibodies, Monoclonal/pharmacology , Autoradiography , Binding, Competitive , Dose-Response Relationship, Drug , Fecal Incontinence/metabolism , Humans , Idazoxan/analogs & derivatives , Idazoxan/pharmacology , Immunohistochemistry , In Vitro Techniques , Muscle Contraction/drug effects , Prazosin/pharmacology , Protein Binding , Radioligand Assay , Receptors, Adrenergic, alpha-1/metabolism , Receptors, Adrenergic, alpha-2/metabolism , Sheep/metabolism
17.
Neurochem Int ; 57(4): 411-20, 2010 Nov.
Article in English | MEDLINE | ID: mdl-20380860

ABSTRACT

Recent in vivo studies have established astrocytes as a major target for locus coeruleus activation (Bekar et al., 2008), renewing interest in cell culture studies on noradrenergic effects on astrocytes in primary cultures and calling for additional information about the expression of adrenoceptor subtypes on different types of brain cells. In the present communication, mRNA expression of alpha(1)-, alpha(2)- and beta-adrenergic receptors and their subtypes was determined in freshly isolated, cell marker-defined populations of astrocytes, NG2-positive cells, microglia, endothelial cells, and Thy1-positive neurons (mainly glutamatergic projection neurons) in murine cerebral cortex. Immediately after dissection of frontal, parietal and occipital cortex of 10-12-week-old transgenic mice, which combined each cell-type marker with a specific fluorescent signal, the tissue was digested, triturated and centrifuged, yielding a solution of dissociated cells of all types, which were separated by fluorescence-activated cell sorting (FACS). mRNA expression in each cell fraction was determined by microarray analysis. alpha(1A)-Receptors were unequivocally expressed in astrocytes and NG2-positive cells, but absent in other cell types, and alpha(1B)-receptors were not expressed in any cell population. Among alpha(2)-receptors only alpha(2A)-receptors were expressed, unequivocally in astrocytes and NG-positive cells, tentatively in microglia and questionably in Thy1-positive neurons and endothelial cells. beta(1)-Receptors were unequivocally expressed in astrocytes, tentatively in microglia, and questionably in neurons and endothelial cells, whereas beta(2)-adrenergic receptors showed tentative expression in neurons and astrocytes and unequivocal expression in other cell types. This distribution was supported by immunochemical data and its relevance established by previous studies in well-differentiated primary cultures of mouse astrocytes, showing that stimulation of alpha(2)-adrenoceptors increases glycogen formation and oxidative metabolism, the latter by a mechanism depending on intramitochondrial Ca(2+), whereas alpha(1)-adrenoceptor stimulation enhances glutamate uptake, and beta-adrenoceptor activation causes glycogenolysis and increased Na(+), K(+)-ATPase activity. The Ca(2+)- and cAMP-mediated association between energy-consuming and energy-yielding processes is emphasized.


Subject(s)
Astrocytes/metabolism , Brain Chemistry/genetics , Brain Chemistry/physiology , Calcium Signaling/physiology , Receptors, Adrenergic/biosynthesis , Receptors, Adrenergic/genetics , Animals , Cell Separation , Cells, Cultured , Flow Cytometry , Gene Expression/physiology , Glucose/metabolism , Glycogen/metabolism , Mice , Mice, Transgenic/physiology , Microarray Analysis , Mitochondria/metabolism , Oxidation-Reduction , Pyruvic Acid/metabolism , RNA/biosynthesis , RNA/genetics , Receptors, Adrenergic, alpha-1/biosynthesis , Receptors, Adrenergic, alpha-1/genetics , Receptors, Adrenergic, alpha-2/biosynthesis , Receptors, Adrenergic, alpha-2/genetics , Receptors, Adrenergic, beta/biosynthesis , Receptors, Adrenergic, beta/genetics
18.
Neurosci Lett ; 471(2): 83-8, 2010 Mar 03.
Article in English | MEDLINE | ID: mdl-20080149

ABSTRACT

Leptin binding to its functional receptor stimulates the Janus kinase-signal transducer and activator of transcription (JAK-STAT) signalling pathway, finally resulting in nuclear translocation of the phosphorylated STAT3 (P-STAT3). Systemic treatment with leptin (3mg/kg; intraperitoneal injection) induced the appearance of P-STAT3-immunoreactive cells in adult mouse preoptic area (POA). Here we show that the vast majority of leptin-responsive cells were located in medial POA (mPOA), followed by the median preoptic nucleus. Rare, scattered and weakly stained cells were found in ventromedial preoptic nucleus and lateral preoptic area. Co-localization studies disclosed that mPOA leptin-responsive cells were neurons, and that a large proportion expressed the alpha(1A)- and/or alpha(2A)-adrenergic receptor (AR) isoforms. Although understanding the functional relevance of leptin-responsive POA neurons requires further investigation, the finding that they bear alpha-ARs suggests that they may be targeted by the ascending noradrenergic system, which densely innervates the mPOA, and thus be involved in thermoregulation, arousal and/or the sleep-wake cycle.


Subject(s)
Leptin/physiology , Neurons/metabolism , Preoptic Area/metabolism , Receptors, Adrenergic, alpha-1/biosynthesis , Receptors, Adrenergic, alpha-2/biosynthesis , Animals , Leptin/pharmacology , Male , Mice , Neurons/drug effects , Preoptic Area/drug effects , Protein Isoforms/biosynthesis , Recombinant Proteins/pharmacology , STAT3 Transcription Factor/metabolism
19.
J Pharmacol Exp Ther ; 333(1): 174-83, 2010 Apr.
Article in English | MEDLINE | ID: mdl-20093398

ABSTRACT

ADP-ribosylation factors (ARFs) regulate vesicular traffic through recruiting coat proteins. However, their functions in the anterograde transport of nascent G protein-coupled receptors (GPCRs) from the endoplasmic reticulum to the plasma membrane remain poorly explored. Here we show that treatment with brefeldin A, an inhibitor of guanine nucleotide exchange on ARFs, markedly attenuated the cell surface numbers of alpha(2B)-adrenergic receptor (AR), beta(2)-AR, angiotensin II type 1 receptor, and chemokine (CXC motif) receptor 4. Functional inhibition of individual ARF GTPases by transient expression of the GDP-bound, GTP-bound, and guanine nucleotide-deficient mutants showed that the five human ARFs differentially modulated receptor cell surface expression and that the ARF1 mutants produced the most profound inhibitory effect. Furthermore, expression of the ARF1 GTPase-activating protein (GAP) ARFGAP1 significantly blocked receptor transport. Interestingly, the GDP- and GTP-bound ARF1 mutants arrested the receptors in distinct intracellular compartments. Consistent with the reduced receptor cell surface expression, extracellular signal-regulated kinase 1 and 2 activation by receptor agonists was significantly attenuated by the GDP-bound mutant ARF1T31N. Moreover, coimmunoprecipitation showed that alpha(2B)-AR associated with ARF1 and glutathione transferase pull-down assay indicated that the alpha(2B)-AR C terminus directly interacted with ARF1. These data show that ARF1 GTPase is involved in the regulation of cell surface expression of GPCRs at multiple transport steps.


Subject(s)
ADP-Ribosylation Factors/physiology , Receptors, G-Protein-Coupled/metabolism , ADP-Ribosylation Factor 1/physiology , Cell Line , Enzyme Activation , Humans , Mitogen-Activated Protein Kinase 1/metabolism , Mitogen-Activated Protein Kinase 3/metabolism , Protein Transport , Receptor, Angiotensin, Type 1/biosynthesis , Receptor, Angiotensin, Type 1/metabolism , Receptors, Adrenergic, alpha-2/biosynthesis , Receptors, Adrenergic, alpha-2/metabolism , Receptors, Adrenergic, beta-2/biosynthesis , Receptors, Adrenergic, beta-2/metabolism , Receptors, CXCR4/biosynthesis , Receptors, CXCR4/metabolism , Receptors, G-Protein-Coupled/biosynthesis
20.
Bone ; 45(3): 568-78, 2009 Sep.
Article in English | MEDLINE | ID: mdl-19446054

ABSTRACT

In contrast to osteoblasts, little attention has been paid to the functional expression of adrenergic signaling machineries in chondrocytes. Expression of mRNA was for the first time demonstrated for different adrenergic receptor (AdR) subtypes in chondrogenic ATDC5 cells and mouse metatarsals isolated before vascularization in culture, but not for other molecules related to adrenergic signaling. In neonatal mouse tibial sections, beta(2)AdR and alpha(2a)AdR mRNA expression was found in chondrocytes at different developmental stages by in situ hybridization. Exposure to adrenaline significantly suppressed expression of several maturation markers through the cAMP/protein kinase A pathway activated by beta(2)AdR without affecting cellular proliferation in both cultured ATDC5 cells and metatarsals. Adrenaline also significantly inhibited gene transactivation by sry-type HMG box 9 (Sox9) family members essential for chondrogenic differentiation in a manner prevented by the general betaAdR antagonist propranolol, with a concomitant significant decrease in the levels of Sox6 mRNA and corresponding protein, in ATDC5 cells and primary cultured mouse costal chondrocytes. Systemic administration of propranolol significantly promoted the increased expression of mRNA for collagen I and collagen X, but not for collagen II, in callus of fractured femur in mice. These results suggest that adrenaline may interfere with chondrogenic differentiation through downregulation of Sox6 expression for subsequent suppression of gene transactivation mediated by Sox9 family members after activation of beta(2)AdR expressed by chondrocytes.


Subject(s)
Chondrocytes/cytology , Epinephrine/pharmacology , Receptors, Adrenergic, alpha-2/biosynthesis , Receptors, Adrenergic, beta-2/biosynthesis , SOX9 Transcription Factor/physiology , Transcriptional Activation , Adrenergic beta-2 Receptor Antagonists , Animals , Animals, Newborn , Cell Differentiation , Cells, Cultured , Chondrocytes/metabolism , Cyclic AMP/metabolism , Embryo, Mammalian , Fractures, Bone/metabolism , Fractures, Bone/pathology , Metatarsal Bones/cytology , Metatarsal Bones/physiology , Mice , RNA, Messenger/biosynthesis , Receptors, Adrenergic, alpha-2/genetics , Receptors, Adrenergic, beta-2/genetics , SOXD Transcription Factors/metabolism , Signal Transduction , Tibia/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...