Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 12 de 12
Filter
Add more filters










Publication year range
1.
Proc Natl Acad Sci U S A ; 115(40): E9499-E9506, 2018 10 02.
Article in English | MEDLINE | ID: mdl-30228117

ABSTRACT

Acetyl-CoA synthetase 2 (ACSS2) is a conserved nucleocytosolic enzyme that converts acetate to acetyl-CoA. Adult mice lacking ACSS2 appear phenotypically normal but exhibit reduced tumor burdens in mouse models of liver cancer. The normal physiological functions of this alternate pathway of acetyl-CoA synthesis remain unclear, however. Here, we reveal that mice lacking ACSS2 exhibit a significant reduction in body weight and hepatic steatosis in a diet-induced obesity model. ACSS2 deficiency reduces dietary lipid absorption by the intestine and also perturbs repartitioning and utilization of triglycerides from adipose tissue to the liver due to lowered expression of lipid transporters and fatty acid oxidation genes. In this manner, ACSS2 promotes the systemic storage or metabolism of fat according to the fed or fasted state through the selective regulation of genes involved in lipid metabolism. Thus, targeting ACSS2 may offer a therapeutic benefit for the treatment of fatty liver disease.


Subject(s)
Acetate-CoA Ligase/metabolism , Adipose Tissue/metabolism , Fatty Liver/metabolism , Gene Expression Regulation , Lipid Metabolism , Liver/metabolism , Acetate-CoA Ligase/genetics , Acetyl Coenzyme A/genetics , Acetyl Coenzyme A/metabolism , Adipose Tissue/pathology , Animals , Fatty Liver/genetics , Fatty Liver/pathology , Liver/pathology , Mice , Mice, Knockout
2.
Cell Rep ; 12(3): 371-9, 2015 Jul 21.
Article in English | MEDLINE | ID: mdl-26166573

ABSTRACT

Nitrogen permease regulator-like 2 (NPRL2) is a component of a conserved complex that inhibits mTORC1 (mammalian Target Of Rapamycin Complex 1) in response to amino acid insufficiency. Here, we show that NPRL2 is required for mouse viability and that its absence significantly compromises fetal liver hematopoiesis in developing embryos. Moreover, NPRL2 KO embryos have significantly reduced methionine levels and exhibit phenotypes reminiscent of cobalamin (vitamin B12) deficiency. Consistent with this idea, NPRL2 KO liver and mouse embryonic fibroblasts (MEFs) show defective processing of the cobalamin-transport protein transcobalamin 2, along with impaired lysosomal acidification and lysosomal gene expression. NPRL2 KO MEFs exhibit a significant defect in the cobalamin-dependent synthesis of methionine from homocysteine, which can be rescued by supplementation with cyanocobalamin. Taken together, these findings demonstrate a role for NPRL2 and mTORC1 in the regulation of lysosomal-dependent cobalamin processing, methionine synthesis, and maintenance of cellular re-methylation potential, which are important during hematopoiesis.


Subject(s)
Hematopoiesis/physiology , Methionine/metabolism , Multiprotein Complexes/antagonists & inhibitors , TOR Serine-Threonine Kinases/antagonists & inhibitors , Tumor Suppressor Proteins/metabolism , Animals , Female , Mechanistic Target of Rapamycin Complex 1 , Mice , Mice, Inbred C57BL , Mice, Knockout , Multiprotein Complexes/metabolism , TOR Serine-Threonine Kinases/metabolism
3.
Proc Natl Acad Sci U S A ; 109(42): 17016-21, 2012 Oct 16.
Article in English | MEDLINE | ID: mdl-23027932

ABSTRACT

We previously reported the discovery of P7C3, an aminopropyl carbazole having proneurogenic and neuroprotective properties in newborn neural precursor cells of the hippocampal dentate gyrus. We have further found that chemicals having efficacy in this in vivo screening assay also protect dopaminergic neurons of the substantia nigra following exposure to the neurotoxin 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine, a mouse model of Parkinson disease. Here, we provide evidence that an active analog of P7C3, known as P7C3A20, protects ventral horn spinal cord motor neurons from cell death in the G93A-SOD1 mutant mouse model of amyotrophic lateral sclerosis (ALS). P7C3A20 is efficacious in this model when administered at disease onset, and protection from cell death correlates with preservation of motor function in assays of walking gait and in the accelerating rotarod test. The prototypical member of this series, P7C3, delays disease progression in G93A-SOD1 mice when administration is initiated substantially earlier than the expected time of symptom onset. Dimebon, an antihistaminergic drug with significantly weaker proneurogenic and neuroprotective efficacy than P7C3, confers no protection in this ALS model. We propose that the chemical scaffold represented by P7C3 and P7C3A20 may provide a basis for the discovery and optimization of pharmacologic agents for the treatment of ALS.


Subject(s)
Amyotrophic Lateral Sclerosis/prevention & control , Carbazoles/pharmacology , Motor Neurons/cytology , Neuroprotective Agents/pharmacology , Spinal Cord/cytology , Animals , Carbazoles/chemical synthesis , Carbazoles/chemistry , Carbazoles/pharmacokinetics , Indoles/pharmacokinetics , Indoles/pharmacology , Mice , Motor Activity/drug effects , Motor Activity/physiology , Motor Neurons/drug effects , Polymerase Chain Reaction , Rotarod Performance Test , Spinal Cord/drug effects
4.
Proc Natl Acad Sci U S A ; 109(42): 17010-5, 2012 Oct 16.
Article in English | MEDLINE | ID: mdl-23027934

ABSTRACT

We previously reported the discovery of P7C3, an aminopropyl carbazole having proneurogenic and neuroprotective properties in newborn neural precursor cells of the dentate gyrus. Here, we provide evidence that P7C3 also protects mature neurons in brain regions outside of the hippocampus. P7C3 blocks 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine (MPTP)-mediated cell death of dopaminergic neurons in the substantia nigra of adult mice, a model of Parkinson disease (PD). Dose-response studies show that the P7C3 analog P7C3A20 blocks cell death with even greater potency and efficacy, which parallels the relative potency and efficacy of these agents in blocking apoptosis of newborn neural precursor cells of the dentate gyrus. P7C3 and P7C3A20 display similar relative effects in blocking 1-methyl-4-phenylpyridinium (MPP(+))-mediated death of dopaminergic neurons in Caenorhabditis elegans, as well as in preserving C. elegans mobility following MPP(+) exposure. Dimebon, an antihistaminergic drug that is weakly proneurogenic and neuroprotective in the dentate gyrus, confers no protection in either the mouse or the worm models of PD. We further demonstrate that the hippocampal proneurogenic efficacy of eight additional analogs of P7C3 correlates with their protective efficacy in MPTP-mediated neurotoxicity. In vivo screening of P7C3 analogs for proneurogenic efficacy in the hippocampus may thus provide a reliable means of predicting neuroprotective efficacy. We propose that the chemical scaffold represented by P7C3 and P7C3A20 provides a basis for optimizing and advancing pharmacologic agents for the treatment of patients with PD.


Subject(s)
1-Methyl-4-phenyl-1,2,3,6-tetrahydropyridine/antagonists & inhibitors , 1-Methyl-4-phenylpyridinium/antagonists & inhibitors , Carbazoles/pharmacology , Dopaminergic Neurons/drug effects , Neuroprotective Agents/pharmacology , Parkinson Disease/prevention & control , Substantia Nigra/cytology , Animals , Apoptosis/drug effects , Caenorhabditis elegans , Carbazoles/chemical synthesis , Carbazoles/chemistry , Carbazoles/pharmacokinetics , Dose-Response Relationship, Drug , Hippocampus/cytology , Hippocampus/drug effects , Indoles/pharmacokinetics , Indoles/pharmacology , Mice , Mice, Inbred C57BL , Molecular Structure , Substantia Nigra/drug effects
5.
J Neurosci ; 31(25): 9404-13, 2011 Jun 22.
Article in English | MEDLINE | ID: mdl-21697390

ABSTRACT

While unesterified cholesterol (C) is essential for remodeling neuronal plasma membranes, its role in certain neurodegenerative disorders remains poorly defined. Uptake of sterol from pericellular fluid requires processing that involves two lysosomal proteins, lysosomal acid lipase, which hydrolyzes C esters, and NPC1 (Niemann-Pick type C1). In systemic tissues, inactivation of either protein led to sterol accumulation and cell death, but in the brain, inactivation of only NPC1 caused C sequestration and neurodegeneration. When injected into the CNS of the npc1(-/-) mouse, 2-hydroxypropyl-ß-cyclodextrin (HP-ß-CD), a compound known to prevent this C accumulation, diffused throughout the brain and was excreted with a t(½) of 6.5 h. This agent caused suppression of C synthesis, elevation of C esters, suppression of sterol regulatory-binding protein 2 (SREBP2) target genes, and activation of liver X receptor-controlled genes. These findings indicated that HP-ß-CD promoted movement of the sequestered C from lysosomes to the metabolically active pool of C in the cytosolic compartment of cells in the CNS. The ED(50) for this agent in the brain was ∼0.5 mg/kg, and the therapeutic effect lasted >7 d. Continuous infusion of HP-ß-CD into the ventricular system of npc1(-/-) animals between 3 and 7 weeks of age normalized the biochemical abnormalities and completely prevented the expected neurodegeneration. These studies support the concept that neurons continuously acquire C from interstitial fluid to permit plasma membrane turnover and remodeling. Inactivation of NPC1 leads to lysosomal C sequestration and neurodegeneration, but this is prevented by the continuous, direct administration of HP-ß-CD into the CNS.


Subject(s)
Cholesterol/metabolism , Endosomes/metabolism , Lysosomes/metabolism , Neurodegenerative Diseases/metabolism , Animals , Biological Transport, Active , Esterification , Mice , Mice, Knockout , Neurodegenerative Diseases/etiology , Neurodegenerative Diseases/prevention & control
6.
J Am Chem Soc ; 133(5): 1428-37, 2011 Feb 09.
Article in English | MEDLINE | ID: mdl-21210688

ABSTRACT

Degeneration of the hippocampus is associated with Alzheimer's disease and occurs very early in the progression of the disease. Current options for treating the cognitive symptoms associated with Alzheimer's are inadequate, giving urgency to the search for novel therapeutic strategies. Pharmacologic agents that safely enhance hippocampal neurogenesis may provide new therapeutic approaches. We discovered the first synthetic molecule, named P7C3, which protects newborn neurons from apoptotic cell death, and thus promotes neurogenesis in mice and rats in the subgranular zone of the hippocampal dentate gyrus, the site of normal neurogenesis in adult mammals. We describe the results of a medicinal chemistry campaign to optimize the potency, toxicity profile, and stability of P7C3. Systematic variation of nearly every position of the lead compound revealed elements conducive toward increases in activity and regions subject to modification. We have discovered compounds that are orally available, nontoxic, stable in mice, rats, and cell culture, and capable of penetrating the blood-brain barrier. The most potent compounds are active at nanomolar concentrations. Finally, we have identified derivatives that may facilitate mode-of-action studies through affinity chromatography or photo-cross-linking.


Subject(s)
Carbazoles/chemistry , Carbazoles/pharmacology , Drug Discovery/methods , Neurogenesis/drug effects , Neuroprotective Agents/chemistry , Neuroprotective Agents/pharmacology , Animals , Carbazoles/therapeutic use , Carbazoles/toxicity , Dose-Response Relationship, Drug , Drug Stability , HeLa Cells , Humans , Male , Mice , Neurodegenerative Diseases/drug therapy , Neuroprotective Agents/therapeutic use , Neuroprotective Agents/toxicity , Structure-Activity Relationship
7.
Cell ; 142(1): 39-51, 2010 Jul 09.
Article in English | MEDLINE | ID: mdl-20603013

ABSTRACT

An in vivo screen was performed in search of chemicals capable of enhancing neuron formation in the hippocampus of adult mice. Eight of 1000 small molecules tested enhanced neuron formation in the subgranular zone of the dentate gyrus. Among these was an aminopropyl carbazole, designated P7C3, endowed with favorable pharmacological properties. In vivo studies gave evidence that P7C3 exerts its proneurogenic activity by protecting newborn neurons from apoptosis. Mice missing the gene encoding neuronal PAS domain protein 3 (NPAS3) are devoid of hippocampal neurogenesis and display malformation and electrophysiological dysfunction of the dentate gyrus. Prolonged administration of P7C3 to npas3(-/-) mice corrected these deficits by normalizing levels of apoptosis of newborn hippocampal neurons. Prolonged administration of P7C3 to aged rats also enhanced neurogenesis in the dentate gyrus, impeded neuron death, and preserved cognitive capacity as a function of terminal aging. PAPERCLIP:


Subject(s)
Carbazoles/pharmacology , Drug Evaluation, Preclinical , Neurogenesis/drug effects , Neurons/cytology , Neuroprotective Agents/pharmacology , Aging/drug effects , Animals , Basic Helix-Loop-Helix Transcription Factors/genetics , Carbazoles/chemistry , Cognition/drug effects , Dentate Gyrus/cytology , Dentate Gyrus/physiology , Female , Hippocampus/cytology , Hippocampus/drug effects , Hippocampus/physiology , Male , Mice , Mice, Inbred C57BL , Mitochondrial Membranes/drug effects , Mitochondrial Membranes/metabolism , Neuroprotective Agents/chemistry , Rats
8.
J Am Chem Soc ; 130(42): 13864-6, 2008 Oct 22.
Article in English | MEDLINE | ID: mdl-18811162

ABSTRACT

We are developing methods that restrict the conformational mobility of peptides and related heteropolymers while simultaneously altering their properties. Our experiments occur as processes wherein a conserved, lipophilic reagent is activated in stages to form composite products with unprotected polyamides in parallel. For each starting oligomer, the goal is to create not one, but rather a collection of products. The intent is for those materials to retain molecular recognition elements of the biopolymer, yet display that functionality as part of stable, cyclic structures having defined shapes and enhanced membrane solubility/permeability. Here we describe reagent 2 and its two-step integration into peptides to afford macrocyclic ethers (e.g., 4 when starting with W-W-Y). When those materials are treated with protic acid in anhydrous solvent, the cinnamyl unit migrates from the oxygen of tyrosine to distribute throughout the structure, forming new products via carbon/carbon bonding. These changes occur concomitantly with acid-promoted rearrangements/cyclizations of the dienyne appendage to generate mixtures containing unique macrocycles such as 15. Similar amalgamations of 2 with more diverse peptides is a means to begin accessing complex peptidomimetics systematically. From a library of screening fractions generated in this way, we have identified a small molecule that selectively promotes hippocampal neurogenesis in the adult mouse brain.


Subject(s)
Acids/chemistry , Cinnamates/chemistry , Macrocyclic Compounds/chemical synthesis , Peptides/chemistry , Animals , Cell Proliferation/drug effects , Cyclization , Dentate Gyrus/cytology , Dentate Gyrus/drug effects , Ions/chemistry , Macrocyclic Compounds/chemistry , Macrocyclic Compounds/pharmacology , Mice , Molecular Conformation , Polymers/chemistry , Stereoisomerism
9.
Proc Natl Acad Sci U S A ; 103(18): 7118-23, 2006 May 02.
Article in English | MEDLINE | ID: mdl-16636276

ABSTRACT

Because the transcription factor neuronal Per-Arnt-Sim-type signal-sensor protein-domain protein 2 (NPAS2) acts both as a sensor and an effector of intracellular energy balance, and because sleep is thought to correct an energy imbalance incurred during waking, we examined NPAS2's role in sleep homeostasis using npas2 knockout (npas2-/-) mice. We found that, under conditions of increased sleep need, i.e., at the end of the active period or after sleep deprivation (SD), NPAS2 allows for sleep to occur at times when mice are normally awake. Lack of npas2 affected electroencephalogram activity of thalamocortical origin; during non-rapid eye movement sleep (NREMS), activity in the spindle range (10-15 Hz) was reduced, and within the delta range (1-4 Hz), activity shifted toward faster frequencies. In addition, the increase in the cortical expression of the NPAS2 target gene period2 (per2) after SD was attenuated in npas2-/- mice. This implies that NPAS2 importantly contributes to the previously documented wake-dependent increase in cortical per2 expression. The data also revealed numerous sex differences in sleep; in females, sleep need accumulated at a slower rate, and REMS loss was not recovered after SD. In contrast, the rebound in NREMS time after SD was compromised only in npas2-/- males. We conclude that NPAS2 plays a role in sleep homeostasis, most likely at the level of the thalamus and cortex, where NPAS2 is abundantly expressed.


Subject(s)
Basic Helix-Loop-Helix Transcription Factors/metabolism , Genotype , Homeostasis , Nerve Tissue Proteins/metabolism , Sleep Stages/physiology , Transcription, Genetic , Animals , Basic Helix-Loop-Helix Transcription Factors/genetics , Brain/cytology , Brain/metabolism , Cell Cycle Proteins , Electroencephalography , Female , Humans , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Nerve Tissue Proteins/genetics , Nuclear Proteins/genetics , Nuclear Proteins/metabolism , Period Circadian Proteins , Sex Factors , Sleep Deprivation , Transcription Factors/genetics , Transcription Factors/metabolism
10.
Proc Natl Acad Sci U S A ; 102(39): 14052-7, 2005 Sep 27.
Article in English | MEDLINE | ID: mdl-16172381

ABSTRACT

The neuronal PAS domain protein 3 (NPAS3) gene encoding a brain-enriched transcription factor was recently found to be disrupted in a family suffering from schizophrenia. Mice harboring compound disruptions in the NPAS3 and related NPAS1 genes manifest behavioral and neuroanatomical abnormalities reminiscent of schizophrenia. Herein we demonstrate that Npas3-/- mice are deficient in expression of hippocampal FGF receptor subtype 1 mRNA, most notably in the dentate gyrus. In vivo BrdUrd-labeling shows that basal neural precursor cell proliferation in the dentate gyrus of Npas3-/- mice is reduced by 84% relative to wild-type littermates. We propose that a deficiency in adult neurogenesis may cause the behavioral and neuroanatomical abnormalities seen in Npas3-/- mice, and we speculate that impaired neurogenesis may be involved in the pathophysiology of schizophrenia.


Subject(s)
Basic Helix-Loop-Helix Transcription Factors/metabolism , Hippocampus/cytology , Neurons/cytology , Schizophrenia/genetics , Animals , Basic Helix-Loop-Helix Transcription Factors/genetics , Behavior, Animal , Cell Proliferation , Dentate Gyrus/anatomy & histology , Dentate Gyrus/chemistry , Dentate Gyrus/cytology , Dentate Gyrus/growth & development , Female , Fibroblast Growth Factor 2/pharmacology , Hippocampus/chemistry , Male , Mice , Mice, Neurologic Mutants , Neurons/metabolism , Neurons/physiology , Oligonucleotide Array Sequence Analysis , RNA, Messenger/metabolism , Schizophrenia/metabolism , Stem Cells/metabolism
11.
Proc Natl Acad Sci U S A ; 101(37): 13648-53, 2004 Sep 14.
Article in English | MEDLINE | ID: mdl-15347806

ABSTRACT

Laboratory mice bearing inactivating mutations in the genes encoding the NPAS1 and NPAS3 transcription factors have been shown to exhibit a spectrum of behavioral and neurochemical abnormalities. Behavioral abnormalities included diminished startle response, as measured by prepulse inhibition, and impaired social recognition. NPAS1/NPAS3-deficient mice also exhibited stereotypic darting behavior at weaning and increased locomotor activity. Immunohistochemical staining assays showed that the NPAS1 and NPAS3 proteins are expressed in inhibitory interneurons and that the viability and anatomical distribution of these neurons are unaffected by the absence of either transcription factor. Adult brain tissues from NPAS3- and NPAS1/NPAS3-deficient mice exhibited a distinct reduction in reelin, a large, secreted protein whose expression has been reported to be attenuated in the postmortem brain tissue of patients with schizophrenia. These observations raise the possibility that a regulatory program controlled in inhibitory interneurons by the NPAS1 and NPAS3 transcription factors may be either substantively or tangentially relevant to psychosis.


Subject(s)
Behavior, Animal/physiology , Brain/metabolism , Nerve Tissue Proteins/deficiency , Transcription Factors/deficiency , Aging/physiology , Animals , Basic Helix-Loop-Helix Transcription Factors , Brain/pathology , Cell Adhesion Molecules, Neuronal/metabolism , Extracellular Matrix Proteins/metabolism , Gene Deletion , Gene Expression Profiling , Homozygote , Mice , Mice, Knockout , Nerve Tissue Proteins/genetics , Nerve Tissue Proteins/metabolism , Neurons/metabolism , Neurons/pathology , Reelin Protein , Serine Endopeptidases , Social Behavior , Transcription Factors/genetics , Transcription Factors/metabolism , gamma-Aminobutyric Acid/metabolism
12.
Science ; 301(5631): 379-83, 2003 Jul 18.
Article in English | MEDLINE | ID: mdl-12843397

ABSTRACT

Animal behavior is synchronized to the 24-hour light:dark (LD) cycle by regulatory programs that produce circadian fluctuations in gene expression throughout the body. In mammals, the transcription factor CLOCK controls circadian oscillation in the suprachiasmatic nucleus of the brain; its paralog, neuronal PAS domain protein 2 (NPAS2), performs a similar function in other forebrain sites. To investigate the role of NPAS2 in behavioral manifestations of circadian rhythm, we studied locomotor activity, sleep patterns, and adaptability to both light- and restricted food-driven entrainment in NPAS2-deficient mice. Our results indicate that NPAS2 plays a substantive role in maintaining circadian behaviors in normal LD and feeding conditions and that NPAS2 is critical for adaptability to food restriction.


Subject(s)
Adaptation, Physiological , Biological Clocks/physiology , Circadian Rhythm/physiology , Motor Activity , Nerve Tissue Proteins/physiology , Sleep , Transcription Factors/physiology , Animals , Basic Helix-Loop-Helix Transcription Factors , Behavior, Animal , Body Weight , CLOCK Proteins , Crosses, Genetic , Darkness , Eating , Electroencephalography , Electromyography , Female , Food , Gene Targeting , Light , Male , Mice , Mice, Inbred C57BL , Nerve Tissue Proteins/genetics , Prosencephalon/physiology , Suprachiasmatic Nucleus/physiology , Trans-Activators/genetics , Trans-Activators/physiology , Transcription Factors/genetics
SELECTION OF CITATIONS
SEARCH DETAIL
...