Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 15 de 15
Filter
Add more filters










Publication year range
1.
Neurotherapeutics ; 20(6): 1808-1819, 2023 Oct.
Article in English | MEDLINE | ID: mdl-37700152

ABSTRACT

Patients with Lafora disease have a mutation in EPM2A or EPM2B, resulting in dysregulation of glycogen metabolism throughout the body and aberrant glycogen molecules that aggregate into Lafora bodies. Lafora bodies are particularly damaging in the brain, where the aggregation drives seizures with increasing severity and frequency, coupled with neurodegeneration. Previous work employed mouse genetic models to reduce glycogen synthesis by approximately 50%, and this strategy significantly reduced Lafora body formation and disease phenotypes. Therefore, an antisense oligonucleotide (ASO) was developed to reduce glycogen synthesis in the brain by targeting glycogen synthase 1 (Gys1). To test the distribution and efficacy of this drug, the Gys1-ASO was administered to Epm2b-/- mice via intracerebroventricular administration at 4, 7, and 10 months. The mice were then sacrificed at 13 months and their brains analyzed for Gys1 expression, glycogen aggregation, and neuronal excitability. The mice treated with Gys1-ASO exhibited decreased Gys1 protein levels, decreased glycogen aggregation, and reduced epileptiform discharges compared to untreated Epm2b-/- mice. This work provides proof of concept that a Gys1-ASO halts disease progression of EPM2B mutations of Lafora disease.


Subject(s)
Lafora Disease , Humans , Mice , Animals , Lafora Disease/genetics , Lafora Disease/metabolism , Glycogen Synthase/genetics , Disease Models, Animal , Mutation , Oligonucleotides, Antisense/therapeutic use , Glycogen/metabolism , Ubiquitin-Protein Ligases/genetics
2.
Nat Commun ; 14(1): 2759, 2023 05 13.
Article in English | MEDLINE | ID: mdl-37179348

ABSTRACT

Matrix assisted laser desorption/ionization imaging has greatly improved our understanding of spatial biology, however a robust bioinformatic pipeline for data analysis is lacking. Here, we demonstrate the application of high-dimensionality reduction/spatial clustering and histopathological annotation of matrix assisted laser desorption/ionization imaging datasets to assess tissue metabolic heterogeneity in human lung diseases. Using metabolic features identified from this pipeline, we hypothesize that metabolic channeling between glycogen and N-linked glycans is a critical metabolic process favoring pulmonary fibrosis progression. To test our hypothesis, we induced pulmonary fibrosis in two different mouse models with lysosomal glycogen utilization deficiency. Both mouse models displayed blunted N-linked glycan levels and nearly 90% reduction in endpoint fibrosis when compared to WT animals. Collectively, we provide conclusive evidence that lysosomal utilization of glycogen is required for pulmonary fibrosis progression. In summary, our study provides a roadmap to leverage spatial metabolomics to understand foundational biology in pulmonary diseases.


Subject(s)
Pulmonary Fibrosis , Mice , Animals , Humans , Glycogen , Metabolomics/methods , Polysaccharides , Spectrometry, Mass, Matrix-Assisted Laser Desorption-Ionization/methods
3.
Cell Rep Methods ; 3(4): 100455, 2023 04 24.
Article in English | MEDLINE | ID: mdl-37159672

ABSTRACT

Brain glucose metabolism is highly heterogeneous among brain regions and continues postmortem. In particular, we demonstrate exhaustion of glycogen and glucose and an increase in lactate production during conventional rapid brain resection and preservation by liquid nitrogen. In contrast, we show that these postmortem changes are not observed with simultaneous animal sacrifice and in situ fixation with focused, high-power microwave. We further employ microwave fixation to define brain glucose metabolism in the mouse model of streptozotocin-induced type 1 diabetes. Using both total pool and isotope tracing analyses, we identified global glucose hypometabolism in multiple brain regions, evidenced by reduced 13C enrichment into glycogen, glycolysis, and the tricarboxylic acid (TCA) cycle. Reduced glucose metabolism correlated with a marked decrease in GLUT2 expression and several metabolic enzymes in unique brain regions. In conclusion, our study supports the incorporation of microwave fixation for more accurate studies of brain metabolism in rodent models.


Subject(s)
Brain , Microwaves , Animals , Mice , Brain/diagnostic imaging , Metabolome , Glucose , Glycogen
4.
Sci Adv ; 7(46): eabi8602, 2021 Nov 12.
Article in English | MEDLINE | ID: mdl-34767443

ABSTRACT

Lactate accumulation is a hallmark of solid cancers and is linked to the immune suppressive phenotypes of tumor-infiltrating immune cells. We report herein that interleukin-4 (IL-4)­induced M0 → M2 macrophage polarization is accompanied by interchangeable glucose- or lactate-dependent tricarboxylic acid (TCA) cycle metabolism that directly drives histone acetylation, M2 gene transcription, and functional immune suppression. Lactate-dependent M0 → M2 polarization requires both mitochondrial pyruvate uptake and adenosine triphosphate­citrate lyase (ACLY) enzymatic activity. Notably, exogenous acetate rescues defective M2 polarization and histone acetylation following mitochondrial pyruvate carrier 1 (MPC1) inhibition or ACLY deficiency. Lastly, M2 macrophage­dependent tumor progression is impaired by conditional macrophage ACLY deficiency, further supporting a dominant role for glucose/lactate mitochondrial metabolism and histone acetylation in driving immune evasion. This work adds to our understanding of how mitochondrial metabolism affects macrophage functional phenotypes and identifies a unique tumor microenvironment (TME)­driven metabolic-epigenetic link in M2 macrophages.

5.
Cell Metab ; 33(7): 1404-1417.e9, 2021 07 06.
Article in English | MEDLINE | ID: mdl-34043942

ABSTRACT

Glycosylation defects are a hallmark of many nervous system diseases. However, the molecular and metabolic basis for this pathology is not fully understood. In this study, we found that N-linked protein glycosylation in the brain is metabolically channeled to glucosamine metabolism through glycogenolysis. We discovered that glucosamine is an abundant constituent of brain glycogen, which functions as a glucosamine reservoir for multiple glycoconjugates. We demonstrated the enzymatic incorporation of glucosamine into glycogen by glycogen synthase, and the release by glycogen phosphorylase by biochemical and structural methodologies, in primary astrocytes, and in vivo by isotopic tracing and mass spectrometry. Using two mouse models of glycogen storage diseases, we showed that disruption of brain glycogen metabolism causes global decreases in free pools of UDP-N-acetylglucosamine and N-linked protein glycosylation. These findings revealed fundamental biological roles of brain glycogen in protein glycosylation with direct relevance to multiple human diseases of the central nervous system.


Subject(s)
Brain/metabolism , Glucosamine/metabolism , Glycogen/physiology , Protein Processing, Post-Translational , Animals , Cells, Cultured , Disease Models, Animal , Female , Glycogen/metabolism , Glycogen Synthase/genetics , Glycogen Synthase/metabolism , Glycogenolysis/genetics , Glycosylation , Lafora Disease/genetics , Lafora Disease/metabolism , Lafora Disease/pathology , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Protein Processing, Post-Translational/genetics
6.
Neurobiol Dis ; 136: 104742, 2020 03.
Article in English | MEDLINE | ID: mdl-31931141

ABSTRACT

The Apolipoprotein E (APOE) gene is a major genetic risk factor associated with Alzheimer's disease (AD). APOE encodes for three main isoforms in humans (E2, E3, and E4). Homozygous E4 individuals have more than a 10-fold higher risk for developing late-onset AD, while E2 carriers are protected. A hallmark of AD is a reduction in cerebral glucose metabolism, alluding to a strong metabolic component in disease onset and progression. Interestingly, E4 individuals display a similar regional pattern of cerebral glucose hypometabolism decades prior to disease onset. Mapping this metabolic landscape may help elucidate the underlying biological mechanism of APOE-associated risk for AD. Efficient metabolic coupling of neurons and glia is necessary for proper neuronal function, and disruption in glial energy distribution has been proposed to contribute to neuronal cell death and AD pathology. One important function of astrocytes - canonically the primary source of apolipoprotein E in the brain - is to provide metabolic substrates (lactate, lipids, amino acids and neurotransmitters) to neurons. Here we investigate the effects of APOE on astrocyte glucose metabolism in vitro utilizing scintillation proximity assays, stable isotope tracer metabolomics, and gene expression analyses. Glucose uptake is impaired in E4 astrocytes relative to E2 or E3 with specific alterations in central carbon metabolism. Using stable isotope labeled glucose [U-13C] allowed analyses of astrocyte-specific deep metabolic networks affected by APOE, and provided insight to the effects downstream of glucose uptake. Enrichment of 13C in early steps of glycolysis was lowest in E4 astrocytes (highest in E2), while synthesis of lactate from glucose was highest in E4 astrocytes (lowest in E2). We observed an increase in glucose flux through the pentose phosphate pathway (PPP), with downstream increases in gluconeogenesis, lipid, and de novo nucleotide biosynthesis in E4 astrocytes. There was also a marked increase in 13C enrichment in the TCA cycle of E4 astrocytes - whose substrates were also incorporated into biosynthetic pathways at a higher rate. Pyruvate carboxylase (PC) and pyruvate dehydrogenase (PDH) are the two main enzymes controlling pyruvate entry to the TCA cycle. PC gene expression is increased in E4 astrocytes and the activity relative to PDH was also increased, compared to E2 or E3. Decreased enrichment in the TCA cycle of E2 and E3 astrocytes is suggestive of increased oxidation and non-glucose derived anaplerosis, which could be fueling mitochondrial ATP production. Conversely, E4 astrocytes appear to increase carbon flux into the TCA cycle to fuel cataplerosis. Together, these data demonstrate clear APOE isoform-specific effects on glucose utilization in astrocytes, including E4-associated increases in lactate synthesis, PPP flux, and de novo biosynthesis pathways.


Subject(s)
Apolipoprotein E4/metabolism , Astrocytes/metabolism , Carbon Isotopes/metabolism , Glucose/metabolism , Animals , Apolipoprotein E4/genetics , Astrocytes/chemistry , Carbon Isotopes/analysis , Cell Line, Transformed , Chromatography, Ion Exchange/methods , Glucose/analysis , Humans , Mice
7.
J Biol Chem ; 294(36): 13464-13477, 2019 09 06.
Article in English | MEDLINE | ID: mdl-31337706

ABSTRACT

Nucleotide synthesis is essential to proliferating cells, but the preferred precursors for de novo biosynthesis are not defined in human cancer tissues. We have employed multiplexed stable isotope-resolved metabolomics to track the metabolism of [13C6]glucose, D2-glycine, [13C2]glycine, and D3-serine into purine nucleotides in freshly resected cancerous and matched noncancerous lung tissues from nonsmall cell lung cancer (NSCLC) patients, and we compared the metabolism with established NSCLC PC9 and A549 cell lines in vitro Surprisingly, [13C6]glucose was the best carbon source for purine synthesis in human NSCLC tissues, in contrast to the noncancerous lung tissues from the same patient, which showed lower mitotic indices and MYC expression. We also observed that D3-Ser was preferentially incorporated into purine rings over D2-glycine in both tissues and cell lines. MYC suppression attenuated [13C6]glucose, D3-serine, and [13C2]glycine incorporation into purines and reduced proliferation in PC9 but not in A549 cells. Using detailed kinetic modeling, we showed that the preferred use of glucose as a carbon source for purine ring synthesis in NSCLC tissues involves cytoplasmic activation/compartmentation of the glucose-to-serine pathway and enhanced reversed one-carbon fluxes that attenuate exogenous serine incorporation into purines. Our findings also indicate that the substrate for de novo nucleotide synthesis differs profoundly between cancer cell lines and fresh human lung cancer tissues; the latter preferred glucose to exogenous serine or glycine but not the former. This distinction in substrate utilization in purine synthesis in human cancer tissues should be considered when targeting one-carbon metabolism for cancer therapy.


Subject(s)
Carcinoma, Non-Small-Cell Lung/metabolism , Glycine/biosynthesis , Lung Neoplasms/metabolism , Purine Nucleotides/biosynthesis , Serine/biosynthesis , A549 Cells , Carcinoma, Non-Small-Cell Lung/pathology , Cell Line, Tumor , Cell Proliferation , Humans , Lung Neoplasms/pathology , Metabolomics
8.
Proteomics ; 19(21-22): e1800486, 2019 11.
Article in English | MEDLINE | ID: mdl-31298457

ABSTRACT

Large clinical trials and model systems studies suggest that the chemical form of selenium dictates chemopreventive and chemotherapeutic efficacy. Selenite induces excess ROS production, which mediates autophagy and eventual cell death in non-small cell lung cancer adenocarcinoma A549 cells. As the mechanisms underlying these phenotypic effects are unclear, the clinical relevance of selenite for cancer therapy remains to be determined. The authors' previous stable isotope-resolved metabolomics and gene expression analysis showed that selenite disrupts glycolysis, the Krebs cycle, and polyamine metabolism in A549 cells, potentially through perturbed glutaminolysis, a vital anaplerotic process for proliferation of many cancer cells. Herein, the role of the glutaminolytic enzyme glutaminase 1 (GLS1) in selenite's toxicity in A549 cells and in patient-derived lung cancer tissues is investigated. Using [13 C6 ]-glucose and [13 C5 ,15 N2 ]-glutamine tracers, selenite's action on metabolic networks is determined. Selenite inhibits glutaminolysis and glutathione synthesis by suppressing GLS1 expression, and blocks the Krebs cycle, but transiently activates pyruvate carboxylase activity. Glutamate supplementation partially rescues these anti-proliferative and oxidative stress activities. Similar metabolic perturbations and necrosis are observed in selenite-treated human patients' cancerous lung tissues ex vivo. The results support the hypothesis that GLS1 suppression mediates part of the anti-cancer activity of selenite both in vitro and ex vivo.


Subject(s)
Glutaminase/genetics , Lung Neoplasms/drug therapy , Metabolomics , Selenious Acid/pharmacology , A549 Cells , Antineoplastic Agents/pharmacology , Autophagy/drug effects , Cell Proliferation/drug effects , Citric Acid Cycle/drug effects , Female , Gene Expression Regulation, Neoplastic/drug effects , Glucose/metabolism , Glutamic Acid/genetics , Glutamic Acid/metabolism , Humans , Lung Neoplasms/genetics , Lung Neoplasms/pathology , Male , Metabolic Networks and Pathways/genetics , Oxidative Stress/drug effects , Reactive Oxygen Species/metabolism
9.
Analyst ; 143(4): 999, 2018 02 12.
Article in English | MEDLINE | ID: mdl-29359211

ABSTRACT

Correction for 'Quantitative profiling of carbonyl metabolites directly in crude biological extracts using chemoselective tagging and nanoESI-FTMS' by Pan Deng, et al., Analyst, 2018, 143, 311-322.

10.
Analyst ; 143(1): 311-322, 2017 12 18.
Article in English | MEDLINE | ID: mdl-29192912

ABSTRACT

The extensive range of chemical structures, wide range of abundances, and chemical instability of metabolites present in the metabolome pose major analytical challenges that are difficult to address with existing technologies. To address these issues, one approach is to target a subset of metabolites that share a functional group, such as ketones and aldehydes, using chemoselective tagging. Here we report a greatly improved chemoselective method for the quantitative analysis of hydrophilic and hydrophobic carbonyl-containing metabolites directly in biological samples. This method is based on direct tissue or cells extraction with simultaneous derivatization of stable and labile carbonylated metabolites using N-[2-(aminooxy)ethyl]-N,N-dimethyl-1-dodecylammonium (QDA) and 13CD3 labeled QDA. We combined innovations of direct quenching of biological sample with frozen derivatization conditions under the catalyst N,N-dimethyl-p-phenylenediamine, which facilitated the formation of oxime stable-isotope ion pairs differing by m/z 4.02188 while minimizing metabolite degradation. The resulting oximes were extracted by HyperSep C8 tips to remove interfering compounds, and the products were detected using nano-electrospray ionization interfaced with a Thermo Fusion mass spectrometer. The quaternary ammonium tagging greatly increased electrospray MS detection sensitivity and the signature ions pairs enabled simple identification of carbonyl compounds. The improved method showed the lower limits of quantification for carbonyl standards to be in the range of 0.20-2 nM, with linearity of R2 > 0.99 over 4 orders of magnitude. We have applied the method to assign 66 carbonyls in mouse tumor tissues, many of which could not be assigned solely by accurate mass and tandem MS. Fourteen of the metabolites were quantified using authentic standards. We also demonstrated the suitability of this method for determining 13C labeled isotopologues of carbonyl metabolites in 13C6-glucose-based stable isotope-resolved metabolomic (SIRM) studies.

11.
J Biol Chem ; 292(28): 11601-11609, 2017 07 14.
Article in English | MEDLINE | ID: mdl-28592486

ABSTRACT

Metabolic reprogramming is a hallmark of cancer. The changes in metabolism are adaptive to permit proliferation, survival, and eventually metastasis in a harsh environment. Stable isotope-resolved metabolomics (SIRM) is an approach that uses advanced approaches of NMR and mass spectrometry to analyze the fate of individual atoms from stable isotope-enriched precursors to products to deduce metabolic pathways and networks. The approach can be applied to a wide range of biological systems, including human subjects. This review focuses on the applications of SIRM to cancer metabolism and its use in understanding drug actions.


Subject(s)
Energy Metabolism , Metabolomics/methods , Neoplasms/metabolism , Animals , Antineoplastic Agents/pharmacology , Antineoplastic Agents/therapeutic use , Carbon Isotopes , Cellular Reprogramming/drug effects , Drug Evaluation, Preclinical/methods , Drug Evaluation, Preclinical/trends , Energy Metabolism/drug effects , Fourier Analysis , Humans , Magnetic Resonance Spectroscopy , Mass Spectrometry , Metabolomics/trends , Neoplasms/drug therapy , Neoplasms/enzymology , Nitrogen Isotopes
12.
Pharmacol Rev ; 66(4): 1033-79, 2014 Oct.
Article in English | MEDLINE | ID: mdl-25244928

ABSTRACT

Phospholipase D is a ubiquitous class of enzymes that generates phosphatidic acid as an intracellular signaling species. The phospholipase D superfamily plays a central role in a variety of functions in prokaryotes, viruses, yeast, fungi, plants, and eukaryotic species. In mammalian cells, the pathways modulating catalytic activity involve a variety of cellular signaling components, including G protein-coupled receptors, receptor tyrosine kinases, polyphosphatidylinositol lipids, Ras/Rho/ADP-ribosylation factor GTPases, and conventional isoforms of protein kinase C, among others. Recent findings have shown that phosphatidic acid generated by phospholipase D plays roles in numerous essential cellular functions, such as vesicular trafficking, exocytosis, autophagy, regulation of cellular metabolism, and tumorigenesis. Many of these cellular events are modulated by the actions of phosphatidic acid, and identification of two targets (mammalian target of rapamycin and Akt kinase) has especially highlighted a role for phospholipase D in the regulation of cellular metabolism. Phospholipase D is a regulator of intercellular signaling and metabolic pathways, particularly in cells that are under stress conditions. This review provides a comprehensive overview of the regulation of phospholipase D activity and its modulation of cellular signaling pathways and functions.


Subject(s)
Neoplasms/pathology , Phosphatidic Acids/metabolism , Phospholipase D/metabolism , Animals , Humans , Molecular Targeted Therapy , Neoplasms/therapy , Signal Transduction/physiology
13.
J Biol Chem ; 289(2): 600-16, 2014 Jan 10.
Article in English | MEDLINE | ID: mdl-24257753

ABSTRACT

The lack of innovative drug targets for glioblastoma multiforme (GBM) limits patient survival to approximately 1 year following diagnosis. The pro-survival kinase Akt provides an ideal target for the treatment of GBM as Akt signaling is frequently activated in this cancer type. However, the central role of Akt in physiological processes limits its potential as a therapeutic target. In this report, we show that the lipid-metabolizing enzyme phospholipaseD(PLD) is a novel regulator of Akt inGBM.Studies using a combination of small molecule PLD inhibitors and siRNA knockdowns establish phosphatidic acid, the product of the PLD reaction, as an essential component for the membrane recruitment and activation of Akt. Inhibition of PLD enzymatic activity and subsequent Akt activation decreases GBM cell viability by specifically inhibiting autophagic flux. We propose a mechanism whereby phosphorylation of beclin1 by Akt prevents binding of Rubicon (RUN domain cysteine-rich domain containing beclin1-interacting protein), an interaction known to inhibit autophagic flux. These findings provide a novel framework through which Akt inhibition can be achieved without directly targeting the kinase.


Subject(s)
Autophagy/physiology , Phospholipase D/metabolism , Proto-Oncogene Proteins c-akt/metabolism , Signal Transduction/physiology , Animals , Apoptosis Regulatory Proteins/metabolism , Autophagy/drug effects , Autophagy/genetics , Autophagy-Related Proteins , Beclin-1 , Cell Line , Cell Line, Tumor , Cell Survival/genetics , Cell Survival/physiology , Culture Media, Serum-Free/pharmacology , Enzyme Inhibitors/pharmacology , Glioblastoma/genetics , Glioblastoma/metabolism , Glioblastoma/pathology , HEK293 Cells , Heterocyclic Compounds, 3-Ring/pharmacology , Humans , Immunoblotting , Intracellular Signaling Peptides and Proteins/metabolism , Membrane Proteins/metabolism , Microscopy, Confocal , Phosphatidic Acids/metabolism , Phospholipase D/antagonists & inhibitors , Phospholipase D/genetics , Phosphorylation/drug effects , Proto-Oncogene Proteins c-akt/antagonists & inhibitors , Proto-Oncogene Proteins c-akt/genetics , RNA Interference , Signal Transduction/drug effects , Signal Transduction/genetics
14.
J Clin Invest ; 122(9): 3197-210, 2012 Sep.
Article in English | MEDLINE | ID: mdl-22886302

ABSTRACT

Aberrant regulation of growth signaling is a hallmark of cancer development that often occurs through the constitutive activation of growth factor receptors or their downstream effectors. Using validation-based insertional mutagenesis (VBIM), we identified family with sequence similarity 83, member B (FAM83B), based on its ability to substitute for RAS in the transformation of immortalized human mammary epithelial cells (HMECs). We found that FAM83B coprecipitated with a downstream effector of RAS, CRAF. Binding of FAM83B with CRAF disrupted CRAF/14-3-3 interactions and increased CRAF membrane localization, resulting in elevated MAPK and mammalian target of rapamycin (mTOR) signaling. Ablation of FAM83B inhibited the proliferation and malignant phenotype of tumor-derived cells or RAS-transformed HMECs, implicating FAM83B as a key intermediary in EGFR/RAS/MAPK signaling. Analysis of human tumor specimens revealed that FAM83B expression was significantly elevated in cancer and was associated with specific cancer subtypes, increased tumor grade, and decreased overall survival. Cumulatively, these results suggest that FAM83B is an oncogene and potentially represents a new target for therapeutic intervention.


Subject(s)
Cell Transformation, Neoplastic/metabolism , ErbB Receptors/metabolism , Neoplasm Proteins/metabolism , Neoplasms/metabolism , ras Proteins/metabolism , Amino Acid Sequence , Animals , Antineoplastic Agents/pharmacology , Base Sequence , Cell Line, Tumor , Cell Proliferation , Drug Resistance, Neoplasm , Epithelial Cells/metabolism , Epithelial Cells/pathology , ErbB Receptors/antagonists & inhibitors , Erlotinib Hydrochloride , Gene Expression , Gene Knockdown Techniques , Humans , Mammary Glands, Human/metabolism , Mammary Glands, Human/pathology , Mice , Mice, Inbred NOD , Mice, SCID , Molecular Sequence Data , Neoplasm Transplantation , Oligonucleotide Array Sequence Analysis , Protein Structure, Tertiary , Proto-Oncogene Proteins c-raf/metabolism , Quinazolines/pharmacology , RNA Interference , Signal Transduction , TOR Serine-Threonine Kinases/metabolism , Transcriptome
15.
Eur J Neurosci ; 26(3): 717-28, 2007 Aug.
Article in English | MEDLINE | ID: mdl-17651428

ABSTRACT

Previous research has shown that individual differences in response to novelty predict self-administration and the locomotor response to psychostimulant drugs of abuse. The aim of the present study was to determine if individual differences in response to novelty based on inescapable or free-choice novelty tests predict dopamine transporter (DAT) function and trafficking as well as nicotine-induced modulation of DAT. Results show that the maximal velocity (Vmax) of [3H]dopamine uptake into prefrontal cortex (PFC) synaptosomes correlated negatively with the locomotor response to inescapable novelty. In contrast, Vmax correlated positively with novelty place preference in the free-choice novelty test. The divergent correlations between DAT and the two behavioral phenotypes suggest a differential contribution of DAT in these phenotypes, which are known not to be isomorphic. Furthermore, rats categorized as high responders to inescapable novelty had lower Vmax values, which were accompanied by less DAT expression at the cell surface in PFC compared with low responders, suggesting that inherent individual differences in DAT cellular localization may underlie the differential response to novelty. Compared with the saline control, nicotine increased Vmax and cell surface DAT immunoreactivity in PFC from high responders but not from low responders. Similarly, nicotine increased Vmax and cell surface DAT in PFC in rats classified as low in novelty place preference but not in rats classified as high in novelty place preference. Thus, despite the different behavioral phenotypes, the pharmacological effect of nicotine to increase DAT function and cell surface expression was apparent, such that rats with inherently lower DAT function show a greater sensitivity to the neurochemical effect of nicotine.


Subject(s)
Dopamine Plasma Membrane Transport Proteins/metabolism , Dopamine/metabolism , Exploratory Behavior/physiology , Nicotine/pharmacology , Prefrontal Cortex/metabolism , Tobacco Use Disorder/metabolism , Aconitine/analogs & derivatives , Aconitine/metabolism , Animals , Behavior, Animal/drug effects , Behavior, Animal/physiology , Binding, Competitive/drug effects , Binding, Competitive/physiology , Cocaine/analogs & derivatives , Cocaine/metabolism , Dopamine Plasma Membrane Transport Proteins/drug effects , Dopamine Uptake Inhibitors/metabolism , Exploratory Behavior/drug effects , Male , Motor Activity/drug effects , Motor Activity/physiology , Nicotinic Agonists/pharmacology , Nicotinic Antagonists/metabolism , Phenotype , Prefrontal Cortex/drug effects , Presynaptic Terminals/drug effects , Presynaptic Terminals/metabolism , Radioligand Assay , Rats , Rats, Sprague-Dawley , Receptors, Cell Surface/drug effects , Receptors, Cell Surface/metabolism , Synaptosomes , Tobacco Use Disorder/physiopathology
SELECTION OF CITATIONS
SEARCH DETAIL
...