Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 14 de 14
Filter
Add more filters










Publication year range
1.
Regul Toxicol Pharmacol ; 127: 105053, 2021 Dec.
Article in English | MEDLINE | ID: mdl-34619288

ABSTRACT

Lemborexant is a dual orexin receptor antagonist (DORA) approved in multiple countries including the United States, Japan, Canada and Australia for the treatment of adults with insomnia. As required for marketing approval of new compounds with central nervous system activity with sedating effects, the abuse potential of lemborexant was assessed in accordance with regulatory guidelines, which included three nonclinical studies. These assessments comprised physical dependence and drug discrimination studies in rats and a self-administration study in rhesus monkeys. There was no evidence of withdrawal signs following abrupt drug discontinuation, indicating that lemborexant does not induce physical dependence. In the drug discrimination study, lemborexant at doses up to 1000 mg/kg administered orally did not cross-generalize to the zolpidem training stimulus, although another DORA included in the same experiment, suvorexant, showed partial generalization with zolpidem. In rhesus monkeys, lemborexant treatment did not induce any gross behavioral changes, and there was no increase in self-administration rates compared with control, indicative of a lack of reinforcing effects of lemborexant. Collectively, these nonclinical studies support the position that lemborexant, which has been placed in Schedule IV by the United States Drug Enforcement Administration, has a low risk of abuse in humans.


Subject(s)
Hypnotics and Sedatives/pharmacology , Orexin Receptor Antagonists/pharmacology , Pyridines/pharmacology , Pyrimidines/pharmacology , Substance-Related Disorders/physiopathology , Animals , Dose-Response Relationship, Drug , Female , Hypnotics and Sedatives/pharmacokinetics , Male , Orexin Receptor Antagonists/pharmacokinetics , Pyridines/pharmacokinetics , Pyrimidines/pharmacokinetics , Rats , Substance Withdrawal Syndrome/physiopathology
2.
Neurosci Res ; 173: 99-105, 2021 Dec.
Article in English | MEDLINE | ID: mdl-34280429

ABSTRACT

Sleep stage scoring is important to determine sleep structure in preclinical and clinical research. The aim of this study was to develop an automatic sleep stage classification system for mice with a new deep neural network algorithm. For the purpose of base feature extraction, wake-sleep and rapid eye movement (REM) and non- rapid eye movement (NREM) models were developed by extracting defining features from mouse-derived electromyogram (EMG) and electroencephalogram (EEG) signals, respectively. The wake-sleep model and REM-NREM sleep model were integrated into three different algorithms including a rule-based integration approach, an ensemble stacking approach, and a multimodal with fine-tuning approach. The deep learning algorithm assessing sleep stages in animal experiments by the multimodal with fine-tuning approach showed high potential for increasing accuracy in sleep stage scoring in mice and promoting sleep research.


Subject(s)
Deep Learning , Algorithms , Animals , Electroencephalography , Mice , Sleep , Sleep Stages
3.
J Alzheimers Dis ; 81(3): 1151-1167, 2021.
Article in English | MEDLINE | ID: mdl-33843668

ABSTRACT

BACKGROUND: Many patients with Alzheimer's disease (AD) display circadian rhythm and sleep-wake disturbances. However, few mouse AD models exhibit these disturbances. Lemborexant, a dual orexin receptor antagonist, is under development for treating circadian rhythm disorders in dementia. OBJECTIVE: Evaluation of senescence-accelerated mouse prone-8 (SAMP8) mice as a model for sleep-wake and rhythm disturbances in AD and the effect of lemborexant by assessing sleep-wake/diurnal rhythm behavior. METHODS: SAMP8 and control senescence-accelerated mouse resistant-1 (SAMR1) mice received vehicle or lemborexant at light onset; plasma lemborexant and diurnal cerebrospinal fluid (CSF) orexin concentrations were assessed. Sleep-wake behavior and running wheel activity were evaluated. RESULTS: Plasma lemborexant concentrations were similar between strains. The peak/nadir timing of CSF orexin concentrations were approximately opposite between strains. During lights-on, SAMP8 mice showed less non-rapid eye movement (non-REM) and REM sleep than SAMR1 mice. Lemborexant treatment normalized wakefulness/non-REM sleep in SAMP8 mice. During lights-off, lemborexant-treated SAMR1 mice showed increased non-REM sleep; lemborexant-treated SAMP8 mice displayed increased wakefulness. SAMP8 mice showed differences in electroencephalogram architecture versus SAMR1 mice. SAMP8 mice exhibited more running wheel activity during lights-on. Lemborexant treatment reduced activity during lights-on and increased activity in the latter half of lights-off, demonstrating a corrective effect on overall diurnal rhythm. Lemborexant delayed the acrophase of activity in both strains by approximately 1 hour. CONCLUSION: SAMP8 mice display several aspects of sleep-wake and rhythm disturbances in AD, notably mistimed activity. These findings provide some preclinical rationale for evaluating lemborexant in patients with AD who experience sleep-wake and rhythm disturbances.


Subject(s)
Alzheimer Disease/complications , Orexin Receptor Antagonists/therapeutic use , Pyridines/therapeutic use , Pyrimidines/therapeutic use , Sleep Wake Disorders/drug therapy , Sleep/drug effects , Animals , Circadian Rhythm/drug effects , Disease Models, Animal , Male , Mice , Motor Activity/drug effects , Orexin Receptor Antagonists/pharmacology , Pyridines/pharmacology , Pyrimidines/pharmacology , Sleep Wake Disorders/complications
4.
Drug Metab Dispos ; 49(1): 31-38, 2021 01.
Article in English | MEDLINE | ID: mdl-33144331

ABSTRACT

Lemborexant is a novel dual orexin receptor antagonist recently approved for the treatment of insomnia in the United States and Japan. Here, disposition and metabolic profiles were investigated in healthy human subjects. After single oral administration of 10 mg [14C]lemborexant (100 µCi), plasma concentrations of lemborexant and radioactivity peaked at 1 hour postdose and decreased biphasically. Cumulative recovery of the administered radioactivity within 480 hours was 86.5% of the dose, with 29.1% in urine and 57.4% in feces. Unchanged lemborexant was not detected in urine but accounted for 13.0% of the dose in feces, suggesting that the main elimination pathway of lemborexant was metabolism. Metabolite analyses revealed that the major metabolic pathways of lemborexant are oxidation of the dimethylpyrimidine moiety and subsequent further oxidation and/or glucuronidation. In plasma, lemborexant was the dominant component, accounting for 26.5% of total drug-related exposure. M4, M9, M10, and M18 were detected as the major radioactive components; M10 was the only metabolite exceeding 10% of total drug-related exposure. Although M4, M9, and M10 showed binding affinity for orexin receptors comparable to that of lemborexant, their contributions to the sleep-promoting effects of lemborexant are likely low because of the limited brain penetration by P-glycoprotein. Exposure comparison between humans and nonclinical toxicology species confirmed that plasma exposure of M10 was higher in at least one animal species compared with that in humans, indicating that there is no disproportionate metabolite in humans, as defined by International Council for Harmonisation of Technical Requirements for Pharmaceuticals for Human Use M3(R2) and U.S. Food and Drug Administration Metabolite in Safety Testing guidance; therefore, no additional toxicology studies are needed. SIGNIFICANCE STATEMENT: This study provides detailed data of the disposition and metabolism of lemborexant, a novel therapeutic drug for insomnia, in humans, as well as a characterization of the circulating metabolites and assessment of their contributions to efficacy and safety. The information presented herein furthers our understanding of the pharmacokinetic profiles of lemborexant and its metabolites and will promote the safe and effective use of lemborexant in the clinic.


Subject(s)
Drug Monitoring/methods , Pyridines , Pyrimidines , Sleep Initiation and Maintenance Disorders/drug therapy , Administration, Oral , Healthy Volunteers , Humans , Metabolic Networks and Pathways , Orexin Receptor Antagonists/blood , Orexin Receptor Antagonists/pharmacokinetics , Pharmacokinetics , Pyridines/blood , Pyridines/pharmacokinetics , Pyrimidines/blood , Pyrimidines/pharmacokinetics , Radioactivity
5.
J Med Chem ; 58(11): 4648-64, 2015 Jun 11.
Article in English | MEDLINE | ID: mdl-25953512

ABSTRACT

The orexin/hypocretin receptors are a family of G protein-coupled receptors and consist of orexin-1 (OX1) and orexin-2 (OX2) receptor subtypes. Orexin receptors are expressed throughout the central nervous system and are involved in the regulation of the sleep/wake cycle. Because modulation of these receptors constitutes a promising target for novel treatments of disorders associated with the control of sleep and wakefulness, such as insomnia, the development of orexin receptor antagonists has emerged as an important focus in drug discovery research. Here, we report the design, synthesis, characterization, and structure-activity relationships (SARs) of novel orexin receptor antagonists. Various modifications made to the core structure of a previously developed compound (-)-5, the lead molecule, resulted in compounds with improved chemical and pharmacological profiles. The investigation afforded a potential therapeutic agent, (1R,2S)-2-{[(2,4-dimethylpyrimidin-5-yl)oxy]methyl}-2-(3-fluorophenyl)-N-(5-fluoropyridin-2-yl)cyclopropanecarboxamide (E2006), an orally active, potent orexin antagonist. The efficacy was demonstrated in mice in an in vivo study by using sleep parameter measurements.


Subject(s)
Amides/chemistry , Aminopyridines/pharmacology , Cyclopropanes/chemistry , Drug Design , Drug Discovery , Orexin Receptor Antagonists , Pyrimidines/pharmacology , Administration, Oral , Aminopyridines/administration & dosage , Animals , Calcium/metabolism , Cells, Cultured , Humans , Male , Mice , Mice, Inbred C57BL , Microsomes, Liver/drug effects , Microsomes, Liver/metabolism , Models, Molecular , Molecular Structure , Orexin Receptors/metabolism , Pyrimidines/administration & dosage , Structure-Activity Relationship
6.
Bioorg Med Chem ; 22(21): 6071-88, 2014 Nov 01.
Article in English | MEDLINE | ID: mdl-25267004

ABSTRACT

Herein we describe the design, synthesis, and structure-activity relationships (SARs) of a novel phenylcyclopropane series represented by 7 and 33 b as antagonists of orexin 1 and orexin 2 receptors. With 4 serving as the initial lead for the development of orexin antagonists, exploration of SAR resulted in improved binding affinity for orexin 1 and orexin 2 receptors. Among the synthesized compounds, 33 b ((-)-N-(5-cyanopyridin-2-yl)-2-[(3,4-dimethoxyphenyl)oxymethyl]-2-phenylcyclopropanecarboxamide) exhibited potent in vitro activity and oral efficacy in animal sleep measurement experiments. The results of our study suggest that compound 33 b may serve as a valuable template for the development of new orexin receptor antagonists.


Subject(s)
Cyclopropanes/chemistry , Cyclopropanes/pharmacology , Orexin Receptor Antagonists , Animals , Cyclopropanes/chemical synthesis , Cyclopropanes/pharmacokinetics , Drug Design , Humans , Male , Mice , Mice, Inbred C57BL , Models, Molecular , Orexin Receptors/metabolism , Sleep/drug effects , Sleep Initiation and Maintenance Disorders/drug therapy , Structure-Activity Relationship
7.
J Biochem ; 141(2): 173-80, 2007 Feb.
Article in English | MEDLINE | ID: mdl-17167040

ABSTRACT

Here we report the enzymatic and ligand-binding properties of a major secretory protein in the choroid plexus of cane toad, Bufo marinus, whose protein is homologous with lipocalin-type prostaglandin (PG) D synthase (L-PGDS) and is recombinantly expressed in Xenopus A6 cells and Escherichia coli. The toad protein bound all-trans retinal, bile pigment, and thyroid hormones with high affinities (K(d)=0.17 to 2.00 microM). The toad protein also catalysed the L-PGDS activity, which was accelerated in the presence of GSH or DTT, similar to the mammalian enzyme. The K(m) value for PGH(2) (17 microM) of the toad protein was almost the same as that of rat L-PGDS (14 microM), whereas the turnover number (6 min(-1)) was approximately 28 fold lower than that of rat L-PGDS. Site-directed mutagenesis based on a modeled structure of the toad protein revealed that Cys(59) and Thr(61) residues were crucial for the PGDS activity. The quadruple Gly(39)Ser/Ala(75)Ser/Ser(140)Thr/Phe(142)Tyr mutant of the toad protein, resembling mouse L-PGDS, showed a 1.6 fold increase in the turnover number and a shift in the optimum pH for the PGDS activity from 9.0 to 8.5. Our results suggest that the toad protein is a prototype of L-PGDS with a highly functional ligand-binding pocket and yet with a primitive catalytic pocket.


Subject(s)
Bufo marinus/metabolism , Choroid Plexus/enzymology , Intramolecular Oxidoreductases/chemistry , Amino Acid Sequence , Animals , Bile Pigments/metabolism , DNA, Complementary , Intramolecular Oxidoreductases/genetics , Intramolecular Oxidoreductases/metabolism , Lipocalins , Models, Molecular , Molecular Sequence Data , Mutagenesis, Site-Directed , Recombinant Proteins/chemistry , Recombinant Proteins/genetics , Recombinant Proteins/metabolism , Retinoids/metabolism , Thyroid Hormones/metabolism , Xenopus
8.
Proc Natl Acad Sci U S A ; 102(28): 9942-7, 2005 Jul 12.
Article in English | MEDLINE | ID: mdl-15983370

ABSTRACT

Neuropeptide B (NPB) and neuropeptide W (NPW) have been recently identified as ligands for the G protein-coupled receptor (GPR) 7 and GPR8. The precise in vivo role of this neuropeptide-receptor pathway has not been fully demonstrated. In this paper, we report that NPB-deficient mice manifest a mild adult-onset obesity, similar to that reported in GPR7-null mice. NPB-deficient mice also exhibit hyperalgesia in response to inflammatory pain. Hyperalgesia was not observed in response to chemical pain, thermal pain, or electrical stimulation. NPB-deficient mice demonstrated intact behavioral responses to pain, and learning from the negative reinforcement of electrical stimulation was unaltered. Baseline anxiety was also unchanged as measured in both the elevated plus maze and time spent immobile in a novel environment. These data support the idea that NPB is a factor in the modulation of responses to inflammatory pain and body weight homeostasis.


Subject(s)
Hyperalgesia/genetics , Inflammation/complications , Neuropeptides/genetics , Pain/physiopathology , Analysis of Variance , Animals , Body Weight/genetics , Formaldehyde , Hyperalgesia/etiology , In Situ Hybridization , Mice , Mice, Knockout , Pain/etiology
9.
Brain Res ; 1044(1): 116-21, 2005 May 17.
Article in English | MEDLINE | ID: mdl-15862796

ABSTRACT

Orexins (hypocretins) are neuropeptides expressed specifically in neurons in the lateral hypothalamic area and are known to be involved in the regulation of vigilance and feeding behavior. However, the relationship between orexin and emotional behaviors like anxiety is still poorly understood. Therefore, in this report we evaluated the effect of intracerebroventricular injection of orexin-A in two major anxiety tests, the light-dark exploration test (mouse) and the elevated plus-maze test (mouse, rat). Orexin increased time spent in the dark compartment in the light-dark test and time spent on the closed arms in the elevated plus-maze test. These results were not caused by a hypothetical sedative or activity-inducing effect of orexin-A because spontaneous locomotor activity did not alter upon orexin-A application under novel conditions. We therefore suggest an anxiogenic effect of orexin-A. To our knowledge, this is the first report about a relationship between orexin-A and anxiety.


Subject(s)
Anxiety/etiology , Behavior, Animal/drug effects , Intracellular Signaling Peptides and Proteins/toxicity , Neuropeptides/toxicity , Analysis of Variance , Animals , Anxiety/physiopathology , Disease Models, Animal , Dose-Response Relationship, Drug , Exploratory Behavior/drug effects , Injections, Intraventricular/methods , Male , Maze Learning/drug effects , Mice , Mice, Inbred C57BL , Motor Activity/drug effects , Orexins , Rats , Rats, Wistar , Time Factors
10.
J Neurosci ; 24(18): 4469-77, 2004 May 05.
Article in English | MEDLINE | ID: mdl-15128861

ABSTRACT

The sleep disorder narcolepsy has been linked to loss of hypothalamic neurons producing the orexin (hypocretin) neuropeptides. Here, we report the generation of transgenic rats expressing a human ataxin-3 fragment with an elongated polyglutamyl stretch under control of the human prepro-orexin promoter (orexin/ataxin-3 rats). At 17 weeks of age, the transgenic rats exhibited postnatal loss of orexin-positive neurons in the lateral hypothalamus, and orexin-containing projections were essentially undetectable. The loss of orexin production resulted in the expression of a phenotype with fragmented vigilance states, a decreased latency to rapid eye movement (REM) sleep and increased REM sleep time during the dark active phase. Wakefulness time was also reduced during the dark phase, and this effect was concentrated at the photoperiod boundaries. Direct transitions from wakefulness to REM sleep, a defining characteristic of narcolepsy, occurred frequently. Brief episodes of muscle atonia and postural collapse resembling cataplexy were also noted while rats maintained the electroencephalographic characteristics of wakefulness. These findings indicate that the orexin/ataxin-3 transgenic rat could provide a useful model of human narcolepsy.


Subject(s)
Carrier Proteins/metabolism , Intracellular Signaling Peptides and Proteins , Narcolepsy/genetics , Nerve Tissue Proteins/biosynthesis , Nerve Tissue Proteins/genetics , Neurons/metabolism , Neuropeptides/metabolism , Transgenes , Animals , Animals, Genetically Modified , Arousal/genetics , Ataxin-3 , Behavior, Animal/physiology , Disease Models, Animal , Electroencephalography , Electromyography , Humans , Male , Narcolepsy/metabolism , Narcolepsy/physiopathology , Neuropeptides/deficiency , Nuclear Proteins , Orexins , Phenotype , Photoperiod , Promoter Regions, Genetic/genetics , Rats , Rats, Wistar , Repressor Proteins , Sleep, REM/genetics , Wakefulness/genetics
11.
J Neurosci ; 23(17): 6793-7, 2003 Jul 30.
Article in English | MEDLINE | ID: mdl-12890773

ABSTRACT

Differential properties of voltage-dependent Ca2+ channels have been primarily ascribed to the alpha1 subunit, of which 10 different subtypes are currently known. For example, channels that conduct the N-type Ca2+ current possess the alpha1B subunit (Cav2.2), which has been localized, inter alia, to the piriform cortex, hippocampus, hypothalamus, locus coeruleus, dorsal raphe, thalamic nuclei, and granular layer of the cortex. Some of these regions have been previously implicated in metabolic and vigilance state control, and selective block of the N-type Ca2+ channel causes circadian rhythm disruption. In this study of Cav2.2-/- knock-out mice, we examined potential differences in feeding behavior, spontaneous locomotion, and the sleep-wake cycle. Cav2.2-/- mice did not display an overt metabolic phenotype but were hyperactive, demonstrating a 20% increase in activity under novel conditions and a 95% increase in activity under habituated conditions during the dark phase, compared with wild-type littermates. Cav2.2-/- mice also displayed vigilance state differences during the light phase, including increased consolidation of rapid-eye movement (REM) sleep and increased intervals between non-REM (NREM) and wakefulness episodes. EEG spectral power was increased during wakefulness and REM sleep and was decreased during NREM sleep in Cav2.2-/- mice. These results indicate a role of the N-type Ca2+ channel in activity and vigilance state control, which we interpret in terms of effects on neurotransmitter release.


Subject(s)
Arousal/genetics , Calcium Channels, N-Type/deficiency , Calcium Channels, N-Type/genetics , Hyperkinesis/genetics , Animals , Electroencephalography , Electromyography , Feeding Behavior/physiology , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Motor Activity/genetics , Motor Activity/physiology , Protein Subunits/deficiency , Protein Subunits/genetics , Sleep Stages/genetics , Sleep Stages/physiology , Sleep, REM/genetics , Sleep, REM/physiology , Wakefulness/genetics , Wakefulness/physiology
12.
Neuron ; 38(5): 701-13, 2003 Jun 05.
Article in English | MEDLINE | ID: mdl-12797956

ABSTRACT

Mammals respond to reduced food availability by becoming more wakeful and active, yet the central pathways regulating arousal and instinctual motor programs (such as food seeking) according to homeostatic need are not well understood. We demonstrate that hypothalamic orexin neurons monitor indicators of energy balance and mediate adaptive augmentation of arousal in response to fasting. Activity of isolated orexin neurons is inhibited by glucose and leptin and stimulated by ghrelin. Orexin expression of normal and ob/ob mice correlates negatively with changes in blood glucose, leptin, and food intake. Transgenic mice, in which orexin neurons are ablated, fail to respond to fasting with increased wakefulness and activity. These findings indicate that orexin neurons provide a crucial link between energy balance and arousal.


Subject(s)
Arousal/genetics , Energy Metabolism/genetics , Food Deprivation/physiology , Hunger/physiology , Hypothalamus/metabolism , Intracellular Signaling Peptides and Proteins , Neurons/metabolism , Neuropeptides/deficiency , Animals , Arousal/drug effects , Blood Glucose/drug effects , Blood Glucose/physiology , Carrier Proteins/genetics , Energy Metabolism/drug effects , Exploratory Behavior/drug effects , Exploratory Behavior/physiology , Extracellular Space/metabolism , Ghrelin , Glucose/metabolism , Glucose/pharmacology , Green Fluorescent Proteins , Homeostasis/drug effects , Homeostasis/genetics , Hunger/drug effects , Hypothalamus/cytology , Hypothalamus/drug effects , Leptin/metabolism , Leptin/pharmacology , Luminescent Proteins , Male , Membrane Potentials/genetics , Mice , Mice, Transgenic , Neurons/cytology , Neurons/drug effects , Neuropeptides/genetics , Orexins , Organ Culture Techniques , Peptide Hormones/metabolism , Peptide Hormones/pharmacology , Recombinant Fusion Proteins , Synaptic Transmission/genetics , Transgenes/genetics
13.
J Mol Med (Berl) ; 80(6): 329-42, 2002 Jun.
Article in English | MEDLINE | ID: mdl-12072908

ABSTRACT

The neuropeptides orexin A and orexin B (also called hypocretin 1 and 2) were recently discovered by a "reverse pharmacology" approach as ligands for two previously orphan G protein coupled receptors: orexin receptors 1 and 2. Neurons producing orexins are located exclusively in the lateral hypothalamic area but project broadly to various parts of the brain, and they have been implicated in the control of energy homeostasis and arousal maintenance. The orexin receptors are also broadly expressed in the central nervous system. Murine and canine models suggest that defective signaling in the orexin system is responsible for the sleep/wake disorder narcolepsy. Although narcoleptic patients rarely have genetic defects in the orexin system, they lack these neuropeptides in the brain and cerebrospinal fluid, indicating that human narcolepsy is an orexin deficiency syndrome in the majority of cases. A connection between sleep/wake regulation and energy homeostasis is hypothesized with orexin neuropeptides as a molecular link.


Subject(s)
Carrier Proteins/physiology , Homeostasis/physiology , Intracellular Signaling Peptides and Proteins , Narcolepsy/physiopathology , Neuropeptides/physiology , Sleep/physiology , Wakefulness/physiology , Animals , Disease Models, Animal , Dogs , Energy Metabolism , Humans , Mice , Narcolepsy/therapy , Orexin Receptors , Orexins , Receptors, G-Protein-Coupled , Receptors, Neuropeptide/metabolism
14.
J Neurosci ; 22(12): 4885-96, 2002 Jun 15.
Article in English | MEDLINE | ID: mdl-12077186

ABSTRACT

The genetic demyelinating mouse "twitcher" is a model of the human globoid cell leukodystrophy, caused by galactosylceramidase (GALC) deficiency. Demyelination in the twitcher brain is secondary to apoptotic death of oligodendrocytes (OLs). Lipocalin-type prostaglandin (PG) D synthase (L-PGDS), a protein expressed in mature OLs, was progressively upregulated in twitcher OLs; whereas expression of OL-associated proteins such as carbonic anhydrase II, myelin basic protein, and myelin-associated glycoprotein was downregulated during demyelination in twitcher brains. The upregulation of L-PGDS was more remarkable in perineuronal OLs than in interfascicular OLs. A larger number of L-PGDS-positive OLs was found in selected fiber tracts of twitcher brains where fewer apoptotic cells were detected. The distribution of L-PGDS-positive OLs was inversely related to the severity of demyelination, as assessed by accumulation of scavenger macrophages. Mice doubly deficient for L-PGDS and GALC disclosed a large number of apoptotic neurons, which were never seen in twitcher brains, in addition to an increased number of apoptotic OLs. A linear positive correlation was observed between the population of L-PGDS-positive OLs in the twitcher brain and the ratio of apoptotic nuclei in the double mutant versus those in the twitcher, suggesting a dose-dependent effect of L-PGDS against apoptosis. These lines of evidence suggest that L-PGDS is an anti-apoptotic molecule protecting neurons and OLs from apoptosis in the twitcher mouse. This is a novel example of OL-neuronal interaction.


Subject(s)
Apoptosis , Intramolecular Oxidoreductases/biosynthesis , Leukodystrophy, Globoid Cell/enzymology , Neurons/pathology , Oligodendroglia/enzymology , Animals , Brain/enzymology , Brain/pathology , Galactosylceramidase/genetics , Intramolecular Oxidoreductases/analysis , Intramolecular Oxidoreductases/genetics , Leukodystrophy, Globoid Cell/genetics , Leukodystrophy, Globoid Cell/pathology , Lipocalins , Mice , Mice, Inbred C57BL , Mice, Knockout , Mice, Neurologic Mutants , Neuroprotective Agents/metabolism , Oligodendroglia/pathology , Point Mutation , RNA, Messenger/biosynthesis , Transcriptional Activation , Up-Regulation
SELECTION OF CITATIONS
SEARCH DETAIL
...