Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 6 de 6
Filter
Add more filters










Database
Language
Publication year range
1.
Brain ; 139(Pt 6): 1762-82, 2016 06.
Article in English | MEDLINE | ID: mdl-27084575

ABSTRACT

The p75 neurotrophin receptor is important in multiple physiological actions including neuronal survival and neurite outgrowth during development, and after central nervous system injury. We have discovered a novel piperazine-derived compound, EVT901, which interferes with p75 neurotrophin receptor oligomerization through direct interaction with the first cysteine-rich domain of the extracellular region. Using ligand binding assays with cysteine-rich domains-fused p75 neurotrophin receptor, we confirmed that EVT901 interferes with oligomerization of full-length p75 neurotrophin receptor in a dose-dependent manner. Here we report that EVT901 reduces binding of pro-nerve growth factor to p75 neurotrophin receptor, blocks pro-nerve growth factor induced apoptosis in cells expressing p75 neurotrophin receptor, and enhances neurite outgrowth in vitro Furthermore, we demonstrate that EVT901 abrogates p75 neurotrophin receptor signalling by other ligands, such as prion peptide and amyloid-ß. To test the efficacy of EVT901 in vivo, we evaluated the outcome in two models of traumatic brain injury. We generated controlled cortical impacts in adult rats. Using unbiased stereological analysis, we found that EVT901 delivered intravenously daily for 1 week after injury, reduced lesion size, protected cortical neurons and oligodendrocytes, and had a positive effect on neurological function. After lateral fluid percussion injury in adult rats, oral treatment with EVT901 reduced neuronal death in the hippocampus and thalamus, reduced long-term cognitive deficits, and reduced the occurrence of post-traumatic seizure activity. Together, these studies provide a new reagent for altering p75 neurotrophin receptor actions after injury and suggest that EVT901 may be useful in treatment of central nervous system trauma and other neurological disorders where p75 neurotrophin receptor signalling is affected.


Subject(s)
Oligodendroglia/drug effects , Piperazines/pharmacology , Receptor, Nerve Growth Factor/antagonists & inhibitors , Animals , Apoptosis/drug effects , Brain Injuries, Traumatic/metabolism , Brain Injuries, Traumatic/pathology , Cell Count , Cell Proliferation/drug effects , Cells, Cultured , Demyelinating Diseases/pathology , Dose-Response Relationship, Drug , Humans , Male , Neurons/drug effects , Neuroprotective Agents/pharmacology , Oligodendroglia/metabolism , Phosphorylation/drug effects , Primary Cell Culture , Radioligand Assay , Rats , Receptor, Nerve Growth Factor/biosynthesis , Receptor, trkA/metabolism , Recovery of Function
2.
Hepatology ; 46(1): 122-9, 2007 Jul.
Article in English | MEDLINE | ID: mdl-17526015

ABSTRACT

This study investigated the effects of rimonabant (SR141716), an antagonist of the cannabinoid receptor type 1 (CB1), on obesity-associated hepatic steatosis and related features of metabolic syndrome: inflammation (elevated plasma levels of tumor necrosis factor alpha [TNFalpha]), dyslipidemia, and reduced plasma levels of adiponectin. We report that oral treatment of obese (fa/fa) rats with rimonabant (30 mg/kg) daily for 8 weeks abolished hepatic steatosis. This treatment reduced hepatomegaly, reduced elevation of plasma levels of enzyme markers of hepatic damage (alanine aminotransferase, gamma glutamyltransferase, and alkaline phosphatase) and decreased the high level of local hepatic TNFalpha currently associated with steatohepatitis. In parallel, treatment of obese (fa/fa) rats with rimonabant reduced the high plasma level of the proinflammatory cytokine TNFalpha and increased the reduced plasma level of the anti-inflammatory hormone adiponectin. Finally, rimonabant treatment also improved dyslipidemia by both decreasing plasma levels of triglycerides, free fatty acids, and total cholesterol and increasing the HDLc/LDLc ratio. All the effects of rimonabant found in this study were not or only slightly observed in pair-fed obese animals, highlighting the additional beneficial effects of treatment with rimonabant compared to diet. These results demonstrate that rimonabant plays a hepatoprotective role and suggest that this CB1 receptor antagonist potentially has clinical applications in the treatment of obesity-associated liver diseases and related features of metabolic syndrome.


Subject(s)
Fatty Liver/prevention & control , Metabolic Syndrome/prevention & control , Obesity/complications , Piperidines/therapeutic use , Pyrazoles/therapeutic use , Animals , Cannabinoid Receptor Antagonists , Fatty Liver/etiology , Inflammation/prevention & control , Liver/drug effects , Liver/pathology , Male , Metabolic Syndrome/etiology , RNA/genetics , RNA/isolation & purification , Rats , Rats, Zucker , Reverse Transcriptase Polymerase Chain Reaction , Rimonabant , Tumor Necrosis Factor-alpha/blood
3.
Eur J Pharmacol ; 529(1-3): 172-8, 2006 Jan 04.
Article in English | MEDLINE | ID: mdl-16325798

ABSTRACT

In diabetic patients, impairment of wound healing is a serious problem which represents a significant health burden. The effect of a highly selective beta-3 adrenoceptor agonist, SR58611A, on wound healing was assessed in animal models of type II diabetes. In db/db diabetic mice, a daily oral treatment with SR58611A (1, 3 and 10 mg/kg/day for two weeks) significantly reduced hyperglycaemia from 3 mg/kg/day onwards. The compound also normalized wound healing, starting from the lowest dose tested (1 mg/kg/day). SR58611A did not affect wound healing of control (lean) mice. An oral anti-diabetic agent, devoid of affinity for beta-3 adrenoceptors, troglitazone (130 mg/kg/day p.o.), normalized glycaemia but did not improve wound healing in db/db mice. Local application of SR58611A (200 microg/day in db/db mice) did not affect wound healing. SR58611A also normalized glucose levels in ob/ob mice, but only slightly improved wound healing in this strain. Moreover, in 17-week old db/db mice (i.e. severely insulin resistant) and in streptozotocin-induced diabetic mice, SR58611A slightly decreased hyperglycaemia and did not affect wound healing. In conclusion, SR58611A improves wound healing in animal models of non-insulin-dependent diabetes. This effect is not related to its effect on glucose levels, but probably implicates systemic effects of the compound.


Subject(s)
Adrenergic beta-3 Receptor Agonists , Adrenergic beta-Agonists/therapeutic use , Diabetes Mellitus, Experimental/physiopathology , Obesity/physiopathology , Tetrahydronaphthalenes/therapeutic use , Wound Healing/drug effects , Administration, Oral , Adrenergic beta-Agonists/administration & dosage , Adrenergic beta-Agonists/pharmacology , Animals , Blood Glucose/analysis , Diabetes Mellitus, Experimental/metabolism , Male , Mice , Mice, Inbred C57BL , Obesity/metabolism , Skin/drug effects , Skin/injuries , Skin/pathology , Tetrahydronaphthalenes/administration & dosage , Tetrahydronaphthalenes/pharmacology , Time Factors
4.
FASEB J ; 19(11): 1567-9, 2005 Sep.
Article in English | MEDLINE | ID: mdl-16009704

ABSTRACT

We investigated the molecular events involved in the long-lasting reduction of adipose mass by the selective CB1 antagonist, SR141716. Its effects were assessed at the transcriptional level both in white (WAT) and brown (BAT) adipose tissues in a diet-induced obesity model in mice. Our data clearly indicated that SR141716 reversed the phenotype of obese adipocytes at both macroscopic and genomic levels. First, oral treatment with SR141716 at 10 mg/kg/d for 40 days induced a robust reduction of obesity, as shown by the 50% decrease in adipose mass together with a major restoration of white adipocyte morphology similar to lean animals. Second, we found that the major alterations in gene expression levels induced by obesity in WAT and BAT were mostly reversed in SR141716-treated obese mice. Importantly, the transcriptional patterns of treated obese mice were similar to those obtained in the CB1 receptor knockout mice fed a high-fat regimen and which are resistant to obesity, supporting a CB1 receptor-mediated process. Functional analysis of these modulations indicated that the reduction of adipose mass by the molecule resulted from an enhanced lipolysis through the induction of enzymes of the beta-oxidation and TCA cycle, increased energy expenditure, mainly through futile cycling (calcium and substrate), and a tight regulation of glucose homeostasis. These changes accompanied a significant cellular remodeling and contributed to a reduction of the obesity-related inflammatory status. In addition to a transient reduction of food consumption, increases of both fatty acid oxidation and energy expenditure induced by the molecule summate leading to a sustained weight loss. Altogether, these data strongly indicate that the endocannabinoid system has a major role in the regulation of energy metabolism.


Subject(s)
Energy Metabolism/drug effects , Lipolysis/drug effects , Obesity/drug therapy , Piperidines/pharmacology , Pyrazoles/pharmacology , Receptor, Cannabinoid, CB1/antagonists & inhibitors , Adipocytes/drug effects , Adiponectin/genetics , Animals , Cytoskeleton/drug effects , Dietary Fats/administration & dosage , Energy Intake , Gene Expression Profiling , Glucose/metabolism , Male , Mice , Mice, Inbred C57BL , Obesity/metabolism , Phenotype , Piperidines/therapeutic use , Pyrazoles/therapeutic use , Rimonabant , Thermogenesis/drug effects
5.
J Pharmacol Exp Ther ; 310(3): 905-14, 2004 Sep.
Article in English | MEDLINE | ID: mdl-15131245

ABSTRACT

Based on binding, functional, and pharmacological data, this study introduces SR147778 [5-(4-bromophenyl)-1-(2,4-dichloro-phenyl)-4-ethyl-N-(1-piperidinyl)-1H-pyrazole-3-carboxamide] as a highly potent, selective, and orally active antagonist for the CB1 receptor. This compound displays nanomolar affinity (Ki = 0.56 and 3.5 nM) for both the rat brain and human CB1 recombinant receptors, respectively. It has low affinity (Ki = 400 nM) for both the rat spleen and human CB2 receptors. Furthermore, it shows no affinity for any of the over 100 targets investigated (IC50 > 1 microM). In vitro, SR147778 antagonizes the inhibitory effects of CP 55,940 [(1R,3R,4R)-3-[2-hydroxy-4-(1,1-dimethylheptyl)phenyl]-4-(3-hydroxypropyl)cyclohexan-1-ol] on both the mouse vas deferens contractions (pA2 value = 8.1) and on forskolin-stimulated adenylyl cyclase activity in the U373 MG cell lines (pA2 value = 8.2) but not in Chinese hamster ovary (CHO) cells permanently expressing the human peripheral cannabinoid receptor (hCB2). SR147778 is able to block the mitogen-activated protein kinase activity induced by CP 55,940 in the CHO cell line expressing human brain cannabinoid receptor (IC50 = 9.6 nM) but was inactive in cells expressing hCB2. After oral administration, SR147778 displaced the ex vivo [3H]-CP 55,940 binding to mouse brain membranes (ED50 = 3.8 mg/kg) with a long duration of action, whereas it did not interact with the CB2 receptor expressed in the mouse spleen. Using different routes of administration, SR147778 (0.3-3 mg/kg) is shown to antagonize pharmacological effects (hypothermia, analgesia, and gastrointestinal transit) induced by R-(+)-(2,3-dihydro-5-methyl-3-[[4-morpholinyl]methyl] pyrol [1,2,3-de]-1,4-benzoxazin-6-yl)(1-naphthalenyl) methanone in mice. Finally, per se, SR147778 (0.3-10 mg/kg) is able to reduce ethanol or sucrose consumption in mice and rats and food intake in fasted and nondeprived rats.


Subject(s)
Piperidines/pharmacology , Pyrazoles/pharmacology , Receptor, Cannabinoid, CB1/antagonists & inhibitors , Animals , Binding, Competitive , CHO Cells , Cricetinae , Male , Mice , Rats , Rats, Sprague-Dawley , Rats, Wistar , Species Specificity
6.
Am J Physiol Regul Integr Comp Physiol ; 284(2): R345-53, 2003 Feb.
Article in English | MEDLINE | ID: mdl-12399252

ABSTRACT

Because the CB1 receptor antagonist SR141716 was previously reported to modulate food intake in rodents, we studied its efficacy in reducing obesity in a diet-induced obesity (DIO) model widely used for research on the human obesity syndrome. During a 5-wk treatment, SR141716 (10 mg. kg(-1). day(-1) orally) induced a transient reduction of food intake (-48% on week 1) and a marked but sustained reduction of body weight (-20%) and adiposity (-50%) of DIO mice. Furthermore, SR141716 corrected the insulin resistance and lowered plasma leptin, insulin, and free fatty acid levels. Most of these effects were present, but less pronounced at 3 mg. kg(-1). day(-1). In addition to its hypophagic action, SR141716 may influence metabolic processes as the body weight loss of SR141716-treated mice was significantly higher during 24-h fasting compared with vehicle-treated animals, and when a 3-day treatment was compared with a pair feeding. SR141716 had no effect in CB1 receptor knockout mice, which confirmed the implication of CB1 receptors in the activity of the compound. These findings suggest that SR141716 has a potential as a novel anti-obesity treatment.


Subject(s)
Diet , Obesity/drug therapy , Piperidines/pharmacology , Piperidines/therapeutic use , Pyrazoles/pharmacology , Pyrazoles/therapeutic use , Receptors, Drug/antagonists & inhibitors , Animals , Binding Sites , Body Weight/drug effects , Dietary Fats/administration & dosage , Dietary Fats/pharmacology , Dose-Response Relationship, Drug , Fatty Acids, Nonesterified/blood , Insulin/blood , Leptin/blood , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Obesity/blood , Obesity/chemically induced , Obesity/physiopathology , Receptors, Cannabinoid , Receptors, Drug/genetics , Receptors, Drug/metabolism , Rimonabant , Time Factors
SELECTION OF CITATIONS
SEARCH DETAIL
...