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.
Cell ; 166(3): 729-739, 2016 Jul 28.
Article in English | MEDLINE | ID: mdl-27471967

ABSTRACT

Isogenic E. coli cells growing in a constant environment display significant variability in growth rates, division sizes, and generation times. The guiding principle appears to be that each cell, during one generation, adds a size increment that is uncorrelated to its birth size. Here, we investigate the mechanisms underlying this "adder" behavior by mapping the chromosome replication cycle to the division cycle of individual cells using fluorescence microscopy. We have found that initiation of chromosome replication is triggered at a fixed volume per chromosome independent of a cell's birth volume and growth rate. Each initiation event is coupled to a division event after a growth-rate-dependent time. We formalize our findings in a model showing that cell-to-cell variation in division timing and cell size is mainly driven by variations in growth rate. The model also explains why fast-growing cells display adder behavior and correctly predict deviations from the adder behavior at slow growth.


Subject(s)
Cell Division/physiology , Chromosomes, Bacterial , DNA Replication , DNA, Bacterial/biosynthesis , Escherichia coli/physiology , Models, Biological
2.
Neurosci Lett ; 611: 51-8, 2016 Jan 12.
Article in English | MEDLINE | ID: mdl-26610904

ABSTRACT

Progressive accumulation of α-synuclein (α-syn)-containing protein aggregates throughout the nervous system is a pathological hallmark of Parkinson's disease (PD). The mechanisms whereby α-syn exerts neurodegeneration remain to be fully understood. Here we show that overexpression of α-syn in transgenic mice leads to increased phosphorylation of glutamate NMDA receptor (NMDAR) subunits NR1 and NR2B in substantia nigra and striatum as well as reduced glucocerebrosidase (GCase) levels. Similarly, molecular studies performed in mouse N2A cells stably overexpressing human α-syn ((α-syn)N2A) showed that phosphorylation states of the same NMDAR subunits were increased, whereas GCase levels and lysosomal GCase activity were reduced. (α-syn)N2A cells showed an increased sensitivity to neurotoxicity towards 6-hydroxydopamine and NMDA. However, wildtype N2A, but not (α-syn)N2A cells, showed a further reduction in viability when co-incubated with 6-hydroxydopamine and the lysosomal inhibitors NH4Cl and leupeptin, suggesting that α-syn per se perturbs lysosomal functions. NMDA treatment reduced lysosomal GCase activity to the same extent in (α-syn)N2A cells as in wildtype N2A cells, indicating that the α-syn-dependent difference in NMDA neurotoxicity is unrelated to an altered GCase activity. Nevertheless, these data provide molecular evidence that overexpression of α-syn simultaneously induces two potential neurotoxic hits by increasing glutamate NMDA receptor phosphorylation, consistent with increased NMDA receptors functionality, and reducing GCase activity.


Subject(s)
Glucosylceramidase/metabolism , Receptors, N-Methyl-D-Aspartate/metabolism , alpha-Synuclein/metabolism , Animals , Cell Line, Tumor , Cell Survival , Corpus Striatum/metabolism , Gene Dosage , Lysosomes/metabolism , Mice , Mice, Inbred C57BL , Oxidopamine/toxicity , Phosphorylation , Protein Subunits/metabolism , Substantia Nigra/metabolism , alpha-Synuclein/genetics
3.
J Biol Chem ; 289(8): 4660-73, 2014 Feb 21.
Article in English | MEDLINE | ID: mdl-24371137

ABSTRACT

The subcellular distribution of the G protein-coupled receptor GPR37 affects cell viability and is implicated in the pathogenesis of parkinsonism. Intracellular accumulation and aggregation of GPR37 cause cell death, whereas GPR37 located in the plasma membrane provides cell protection. We define here a pathway through which the recently identified natural ligand, prosaposin, promotes plasma membrane association of GPR37. Immunoabsorption of extracellular prosaposin reduced GPR37(tGFP) surface density and decreased cell viability in catecholaminergic N2a cells. We found that GPR37(tGFP) partitioned in GM1 ganglioside-containing lipid rafts in the plasma membrane of live cells. This partitioning required extracellular prosaposin and was disrupted by lipid raft perturbation using methyl-ß-cyclodextrin or cholesterol oxidase. Moreover, complex formation between GPR37(tGFP) and the GM1 marker cholera toxin was observed in the plasma membrane. These data show functional association between GPR37, prosaposin, and GM1 in the plasma membrane. These results thus tie together the three previously defined components of the cellular response to insult. Our findings identify a mechanism through which the receptor's natural ligand and GM1 may protect against toxic intracellular GPR37 aggregates observed in parkinsonism.


Subject(s)
Cell Membrane/metabolism , G(M1) Ganglioside/metabolism , Receptors, G-Protein-Coupled/metabolism , Saposins/metabolism , Animals , Cell Membrane/drug effects , Cell Polarity/drug effects , Cell Survival/drug effects , Endocytosis/drug effects , Extracellular Space/metabolism , Flow Cytometry , Fluorescence Recovery After Photobleaching , Green Fluorescent Proteins/metabolism , Lysosomes/drug effects , Lysosomes/metabolism , Membrane Microdomains/metabolism , Mice , Mitochondria/drug effects , Mitochondria/metabolism , Nerve Growth Factors/metabolism , Peptides/metabolism , Protein Binding/drug effects , Protein Transport/drug effects , Time Factors , beta-Cyclodextrins/pharmacology
4.
J Neurochem ; 124(3): 410-7, 2013 Feb.
Article in English | MEDLINE | ID: mdl-23121049

ABSTRACT

G protein-coupled receptor 37 (GPR37) is suggested to be implicated in the pathogenesis of Parkinson's disease and is accumulating in Lewy bodies within afflicted brain regions. Over-expressed GPR37 is prone to misfolding and aggregation, causing cell death via endoplasmic reticulum stress. Although the cytotoxicity of misfolded GPR37 is well established, effects of the functional receptor on cell viability are still unknown. An N2a cell line stably expressing green fluorescent protein (GFP)-tagged human GPR37 was created to study its trafficking and effects on cell viability upon challenge with the toxins 1-methyl-4-phenylpyridinium (MPP+), rotenone and 6-hydroxydopamine (6-OHDA). Neuronal-like differentiation into a tyrosine hydroxylase expressing phenotype, using dibutyryl-cAMP, induced trafficking of GPR37 to the plasma membrane. 3-(4,5-Dimethylthiazol-2-yl)-2,5-diphenyltetrazolium bromide (MTT) cell viability and lactate dehydrogenase (LDH) cell death assays revealed that GPR37 was protective against all three toxins in differentiated cells. In undifferentiated cells, the majority of GPR37 was cytoplasmic and the protective effects were more variable: GPR37 expression protected against rotenone and MPP+ but not against 6-OHDA in MTT assays, while it protected against 6-OHDA but not against MPP+ or rotenone in lactate dehydrogenase (LDH) assays. These results suggest that GPR37 functionally trafficked to the plasma membrane protects against toxicity.


Subject(s)
1-Methyl-4-phenylpyridinium/toxicity , Oxidopamine/toxicity , Receptors, G-Protein-Coupled/metabolism , Rotenone/toxicity , Animals , Catecholamines/physiology , Cell Line, Tumor , Green Fluorescent Proteins/genetics , Herbicides/toxicity , Humans , Mice , Neuroblastoma , Protein Transport/drug effects , Protein Transport/physiology , Receptors, G-Protein-Coupled/genetics , Sympatholytics/toxicity , Uncoupling Agents/toxicity
5.
J Neurosci ; 31(47): 16928-40, 2011 Nov 23.
Article in English | MEDLINE | ID: mdl-22114263

ABSTRACT

"Ecstasy" [3,4-methylenedioxymetamphetamine (MDMA)] is of considerable interest in light of its prosocial properties and risks associated with widespread recreational use. Recently, it was found to bind trace amine-1 receptors (TA(1)Rs), which modulate dopaminergic transmission. Accordingly, using mice genetically deprived of TA(1)R (TA(1)-KO), we explored their significance to the actions of MDMA, which robustly activated human adenylyl cyclase-coupled TA(1)R transfected into HeLa cells. In wild-type (WT) mice, MDMA elicited a time-, dose-, and ambient temperature-dependent hypothermia and hyperthermia, whereas TA(1)-KO mice displayed hyperthermia only. MDMA-induced increases in dialysate levels of dopamine (DA) in dorsal striatum were amplified in TA(1)-KO mice, despite identical levels of MDMA itself. A similar facilitation of the influence of MDMA upon dopaminergic transmission was acquired in frontal cortex and nucleus accumbens, and induction of locomotion by MDMA was haloperidol-reversibly potentiated in TA(1)-KO versus WT mice. Conversely, genetic deletion of TA(1)R did not affect increases in DA levels evoked by para-chloroamphetamine (PCA), which was inactive at hTA(1) sites. The TA(1)R agonist o-phenyl-3-iodotyramine (o-PIT) blunted the DA-releasing actions of PCA both in vivo (dialysis) and in vitro (synaptosomes) in WT but not TA(1)-KO animals. MDMA-elicited increases in dialysis levels of serotonin (5-HT) were likewise greater in TA(1)-KO versus WT mice, and 5-HT-releasing actions of PCA were blunted in vivo and in vitro by o-PIT in WT mice only. In conclusion, TA(1)Rs exert an inhibitory influence on both dopaminergic and serotonergic transmission, and MDMA auto-inhibits its neurochemical and functional actions by recruitment of TA(1)R. These observations have important implications for the effects of MDMA in humans.


Subject(s)
Gene Deletion , N-Methyl-3,4-methylenedioxyamphetamine/antagonists & inhibitors , N-Methyl-3,4-methylenedioxyamphetamine/pharmacology , Receptors, G-Protein-Coupled/deficiency , Receptors, G-Protein-Coupled/physiology , Animals , Dopamine/physiology , Dose-Response Relationship, Drug , HeLa Cells , Humans , Male , Mice , Mice, 129 Strain , Mice, Inbred C57BL , Mice, Knockout , Random Allocation , Receptors, G-Protein-Coupled/genetics , Serotonin/physiology
6.
J Neurosci ; 30(12): 4369-81, 2010 Mar 24.
Article in English | MEDLINE | ID: mdl-20335473

ABSTRACT

The preoptic area/anterior hypothalamus, a region that contains neurons that control thermoregulation, is the main locus at which histamine affects body temperature. Here we report that histamine reduced the spontaneous firing rate of GABAergic preoptic neurons by activating H3 subtype histamine receptors. This effect involved a decrease in the level of phosphorylation of the extracellular signal-regulated kinase and was not dependent on synaptic activity. Furthermore, a population of non-GABAergic neurons was depolarized, and their firing rate was enhanced by histamine acting at H1 subtype receptors. In our experiments, activation of the H1R receptors was linked to the PLC pathway and Ca(2+) release from intracellular stores. This depolarization persisted in TTX or when fast synaptic potentials were blocked, indicating that it represents a postsynaptic effect. Single-cell reverse transcription-PCR analysis revealed expression of H3 receptors in a population of GABAergic neurons, while H1 receptors were expressed in non-GABAergic cells. Histamine applied in the median preoptic nucleus induced a robust, long-lasting hyperthermia effect that was mimicked by either H1 or H3 histamine receptor subtype-specific agonists. Our data indicate that histamine modulates the core body temperature by acting at two distinct populations of preoptic neurons that express H1 and H3 receptor subtypes, respectively.


Subject(s)
Body Temperature/drug effects , Histamine/pharmacology , Neurons/drug effects , Preoptic Area/cytology , Receptors, Histamine H1/metabolism , Receptors, Histamine H3/metabolism , 6-Cyano-7-nitroquinoxaline-2,3-dione/pharmacology , Action Potentials/drug effects , Animals , Animals, Newborn , Calcium/metabolism , Cells, Cultured , Enzyme Inhibitors/pharmacology , Excitatory Amino Acid Antagonists/pharmacology , Excitatory Postsynaptic Potentials/drug effects , Gene Expression Regulation/drug effects , Glutamate Decarboxylase/genetics , Glutamate Decarboxylase/metabolism , Glutamic Acid/metabolism , Green Fluorescent Proteins/genetics , Histamine Agonists/pharmacology , Histamine Antagonists/pharmacology , In Vitro Techniques , Mice , Mice, Transgenic , Motor Activity/drug effects , Motor Activity/physiology , Patch-Clamp Techniques , Receptors, Histamine H1/drug effects , Receptors, Histamine H3/drug effects , Signal Transduction/drug effects , Signal Transduction/physiology , Sodium Channel Blockers/pharmacology , Telemetry/methods , Tetrodotoxin/pharmacology , Type C Phospholipases/metabolism , gamma-Aminobutyric Acid/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...