Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 23
Filter
1.
Sci Rep ; 10(1): 8886, 2020 06 01.
Article in English | MEDLINE | ID: mdl-32483272

ABSTRACT

This paper examines the effect of TET1 expression on survival in glioma patients using open-access data from the Genomic Data Commons. A neural network-based survival model was built on expression data from a selection of genes most affected by TET1 knockdown with a median cross-validated survival concordance of 82.5%. A synthetic experiment was then conducted that linked two separately trained neural networks: a multitask model estimating cancer hallmark gene expression from TET1 expression, and a survival neural network. This experiment quantified the mediation of the TET1 survival effect through eight cancer hallmarks: apoptosis, cell cycle, cell death, cell motility, DNA repair, immune response, two phosphorylation pathways, and a randomized gene sets. Immune response, DNA repair, and apoptosis displayed greater mediation than the randomized gene set. Cell motility was inversely associated with only 12.5% mediated concordance. We propose the neural network linkage mediation experiment as an approach to collecting evidence of hazard mediation relationships with prognostic capacity useful for designing interventions.


Subject(s)
Brain Neoplasms/mortality , Gene Regulatory Networks , Glioma/mortality , Mixed Function Oxygenases/genetics , Proto-Oncogene Proteins/genetics , Brain Neoplasms/genetics , Databases, Genetic , Gene Expression Profiling , Gene Expression Regulation, Neoplastic , Gene Knockdown Techniques , Glioma/genetics , Humans , Mutation , Neural Networks, Computer , Sequence Analysis, RNA , Survival Analysis
2.
Toxicol Appl Pharmacol ; 380: 114646, 2019 10 01.
Article in English | MEDLINE | ID: mdl-31278917

ABSTRACT

Glioblastomas are the most aggressive of malignant brain cancers with a median patient survival of approximately 18 months. We recently demonstrated that Tet methylcytosine dioxygenase 1(TET1) is involved in cellular responses to ionizing radiation (IR) in glial-, glioblastoma-, and non-tumor-derived cells. This study used a lentiviral-mediated knockdown of TET1 to further dissect the contribution of TET1 to the DNA damage response in glial cell lines by evaluating its role in DNA repair. TET1-deficient glial cell lines displayed attenuated cytotoxicity compared to non-targeted knockdown after treatment with IR but these differences were not observed between control and TET1 deficient in response to inhibitors of Na+/K+-ATPase. Additionally, the percentage of glial cells displaying γH2A.x foci was greatly reduced in TET1-deficient glial cells compared to non-targeted knockdown conditions in response to IR and topoisomerase inhibitors. We also observed a lower percentage and a delay in 53BP1 foci formation, a marker of non-homologous end-joining, in response to IR and topoisomerase inhibitors in TET1-deficient glial cells. DNA-PK, another marker of non-homologous end-joining, was also lower in TET1-deficient glial cell lines. Interestingly, TET1-deficient glial cells displayed higher numbers of DNA strand breaks compared to control cells and repaired DNA breaks less efficiently in Comet assays. We suggest that attenuated DNA repair in TET1 deficient gliomas leads to genomic instability, which underlies poor patient survival.


Subject(s)
DNA Repair , Mixed Function Oxygenases/genetics , Neuroglia , Proto-Oncogene Proteins/genetics , Radiation, Ionizing , Cell Line , DNA Damage , Glioma/genetics , Humans
3.
Epigenetics ; 12(10): 854-864, 2017.
Article in English | MEDLINE | ID: mdl-28758831

ABSTRACT

Recent studies have shown that loss of TET1 may play a significant role in the formation of tumors. Because genomic instability is a hallmark of cancer, we examined the potential involvement of 10-11 translocation 1 (TET1) in the DNA damage response (DDR). Here we demonstrate that, in response to clinically relevant doses of ionizing radiation (IR), human glial cells made TET1-deficient with lentiviral vectors displayed greater numbers of colony forming units and lower levels of apoptotic markers compared with glial cells transduced with control vectors; yet, they harbored greater DNA strand breaks. The G2/M check point and expression of cyclin B1 were greatly diminished in TET1-deficient cells, and TET1-deficient cells displayed lower levels of γH2A.x following exposure to IR. Levels of DNA-PKcs, which are DNA-PK complex members, were lower in TET1-deficient cells compared with control cell lines. However, levels of ATM were similar in both cell lines. Cyclin B1, DNA-PKcs, and γH2A.x levels were each rescued by reintroduction of the TET1 catalytic domain. Finally, cytosine methylation within intron 1 of PRKDC, the gene encoding DNA-PKcs, was significantly higher upon depletion of TET1. Taken together, this study illustrates the involvement of TET1 in the different arms of the DDR and suggests its loss results in the continued survival of cells with genomic instability.


Subject(s)
DNA Damage/radiation effects , DNA Methylation/genetics , Genomic Instability/genetics , Mixed Function Oxygenases/genetics , Proto-Oncogene Proteins/genetics , Cyclin B1/genetics , Cytosine/metabolism , DNA Damage/genetics , DNA Methylation/radiation effects , DNA Repair/genetics , DNA-Activated Protein Kinase/genetics , Gene Expression Regulation/radiation effects , Genomic Instability/radiation effects , Histones/genetics , Humans , Lentivirus/genetics , Neoplasms/genetics , Neoplasms/pathology , Neuroglia/radiation effects , Nuclear Proteins/genetics , Radiation, Ionizing
4.
Clin Epigenetics ; 9: 29, 2017.
Article in English | MEDLINE | ID: mdl-28360946

ABSTRACT

BACKGROUND: Genetics explains a small proportion of variance in body mass index at the population level. Epigenetics, commonly measured by gene methylation, holds promise for understanding obesity risk factors and mechanisms. METHODS: Participants were 431 adolescents aged 10-15 years. BMI z-score, waist circumference z-score, and percent body fat were measured. Saliva samples were collected and methylation of promoter regions of four candidate genes or sequences (LEP, ICAM-1, CRH, and LINE-1) were measured in 3-4 CpG sites each. Linear regression was used to identify associations of methylation with obesity-related outcomes. RESULTS: After adjusting for age, in sex-stratified analysis, the three obesity-related outcomes were negatively associated with LEP methylation in obese boys only. There were no associations of methylation of the other genes or sequences and the obesity-related outcomes. CONCLUSIONS: Our results are consistent with prior studies that reported sex differences in associations of obesity-related outcomes with LEP methylation, and also as would be expected in adipose tissue, the source of circulating leptin. The findings suggest that saliva might be an acceptable tissue for epigenetics studies in adolescents.


Subject(s)
Corticotropin-Releasing Hormone/genetics , DNA Methylation , Intercellular Adhesion Molecule-1/genetics , Leptin/genetics , Long Interspersed Nucleotide Elements , Obesity/genetics , Adolescent , Body Mass Index , Child , Epigenesis, Genetic , Female , Humans , Male , Sex Characteristics , Waist Circumference
5.
J Neuroimmunol ; 283: 23-9, 2015 Jun 15.
Article in English | MEDLINE | ID: mdl-26004152

ABSTRACT

Knocking down methyl CpG binding protein 2 (MeCP2) enhances NF-κB activation in human peripheral blood mononuclear cells (PBMC). In this study, we examined whether this caused the expression of cytokines to be elevated. Increased levels of TNFα, IL-6, and IL-3 mRNAs were observed in human PBMC made MeCP2 deficient with a lentiviral shRNA MeCP2 vector and in splenocytes from MeCP2-null mice. TNFα neutralizing antibody attenuated expression of IL-6 and TNFα but did not affect expression of IL-3. Lipopolysaccharide-mediated increases in TNFα, IL-6, and IL-3 mRNAs were also enhanced in MeCP2-deficient PBMC. Two inhibitors of NF-κB blocked the increased levels of IL-6, TNFα, and IL-3 in MeCP2-deficient PBMC treated with lipopolysaccharide. MeCP2 deficiency also enhanced expression of IL-6 and TNFα mRNAs in the THP1 human monocyte cell line, which were also attenuated by the NF-κB inhibitors. In chromatin immunoprecipitation assays, the binding of the NF-κB family member p65 and acetylated H3 to the TNFα promoter was greater after treatment with LPS in MeCP2-deficient THP1 cells. MeCP2 did not bind to the TNFα promoter. In summary, the data indicates that MeCP2 deficiency increases expression of TNFα and other inflammatory cytokines by enhancing NF-κB signaling.


Subject(s)
Methyl-CpG-Binding Protein 2/physiology , NF-kappa B/metabolism , Animals , Cell Line , Humans , Inflammation , Interleukin-3/biosynthesis , Interleukin-3/genetics , Interleukin-6/biosynthesis , Interleukin-6/genetics , Leukocytes, Mononuclear/metabolism , Lipopolysaccharides/pharmacology , Methyl-CpG-Binding Protein 2/deficiency , Methyl-CpG-Binding Protein 2/genetics , Mice , Mice, Inbred C57BL , Mice, Knockout , Monocytes/metabolism , Promoter Regions, Genetic , RNA Interference , RNA, Small Interfering/pharmacology , Signal Transduction/physiology , Spleen/cytology , Transcription Factor RelA/metabolism , Transcription, Genetic , Tumor Necrosis Factor-alpha/biosynthesis , Tumor Necrosis Factor-alpha/genetics
6.
J Neuroimmunol ; 265(1-2): 61-7, 2013 Dec 15.
Article in English | MEDLINE | ID: mdl-24268627

ABSTRACT

The signaling pathway involved in regulating glutamate release was investigated. Glutaminase mRNA levels were higher in microglia isolated from mice treated with lipopolysaccharide. Pam3CSK and lipopolysaccharide stimulated glutamate release in neonatal mouse microglia cultures, which was attenuated by NF-κB inhibitors. Higher levels of glutaminase mRNA and glutamate release were observed in human peripheral blood mononuclear cells made MeCP2 deficient with MeCP2 shRNA, which was blocked by NF-κB inhibitors. NF-κB inhibitors also decreased the higher levels of glutaminase mRNA in MeCP2 deficient THP1 monocyte cell lines. Finally, an inverse relation was observed between MeCP2 levels and NF-κB reporter activity in THP1 cells. We suggest that NF-κB pathway is involved in the release of glutamate in MeCP2 deficient cells from the myeloid lineage.


Subject(s)
Glutamic Acid/metabolism , Methyl-CpG-Binding Protein 2/deficiency , Myeloid Cells/metabolism , NF-kappa B/metabolism , Signal Transduction/physiology , Animals , Animals, Newborn , Brain/cytology , Brain/drug effects , Brain/metabolism , CD11b Antigen/metabolism , Cells, Cultured , Enzyme Inhibitors/pharmacology , Glutamic Acid/genetics , Glutaminase/genetics , Glutaminase/metabolism , Leukocytes, Mononuclear/metabolism , Lipopolysaccharides/pharmacology , Male , Mice , Microglia/drug effects , Microglia/metabolism , Myeloid Cells/drug effects , Myeloid Cells/immunology , Nitric Oxide/metabolism , RNA, Messenger/genetics , RNA, Small Interfering/genetics , RNA, Small Interfering/metabolism , Signal Transduction/drug effects , beta-Galactosidase/metabolism
7.
J Biol Chem ; 288(40): 28792-800, 2013 Oct 04.
Article in English | MEDLINE | ID: mdl-23940045

ABSTRACT

DNA methylation regulates gene expression throughout development and in a wide range of pathologies such as cancer and neurological disorders. Pathways controlling the dynamic levels and targets of methylation are known to be disrupted by chemicals and are therefore of great interest in both prevention and clinical contexts. Benzene and its metabolite hydroquinone have been shown to lead to decreased levels of DNA methylation, although the mechanism is not known. This study employs a cell culture model to investigate the mechanism of hydroquinone-mediated changes in DNA methylation. Exposures that do not affect HEK293 cell viability led to genomic and methylated reporter DNA demethylation. Hydroquinone caused reactivation of a methylated reporter plasmid that was prevented by the addition of N-acetylcysteine. Hydroquinone also caused an increase in Ten Eleven Translocation 1 activity and global levels of 5-hydroxymethylcytosine. 5-Hydroxymethylcytosine was found enriched at LINE-1 prior to a decrease in both 5-hydroxymethylcytosine and 5-methylcytosine. Ten Eleven Translocation-1 knockdown decreased 5-hydroxymethylcytosine formation following hydroquinone exposure as well as the induction of glutamate-cysteine ligase catalytic subunit and 14-3-3σ. Finally, Ten Eleven Translocation 1 knockdown decreased the percentage of cells accumulating in G2+M following hydroquinone exposure, indicating that it may have a role in cell cycle changes in response to toxicants. This work demonstrates that hydroquinone exposure leads to active and functional DNA demethylation in HEK293 cells in a mechanism involving reactive oxygen species and Ten Eleven Translocation 1 5-methylcytosine dioxygenase.


Subject(s)
Cytosine/analogs & derivatives , DNA-Binding Proteins/metabolism , Dioxygenases/metabolism , Hydroquinones/pharmacology , Proto-Oncogene Proteins/metabolism , 5-Methylcytosine/analogs & derivatives , Cell Cycle/drug effects , Cell Cycle/genetics , Cell Nucleus/drug effects , Cell Nucleus/metabolism , Cytosine/metabolism , DNA/metabolism , DNA Methylation/drug effects , Gene Expression Regulation/drug effects , Genome, Human/genetics , HEK293 Cells , Humans , Mixed Function Oxygenases , Reactive Oxygen Species/metabolism
8.
Biochim Biophys Acta ; 1833(3): 460-7, 2013 Mar.
Article in English | MEDLINE | ID: mdl-23195226

ABSTRACT

Chemicals that affect cellular differentiation through epigenetic mechanisms have potential utility in treating a wide range of diseases. Hydralazine decreases DNA methylation in some cell types but its effect on differentiation has not been well explored. After five days of exposure to hydralazine, P19 embryocarcinoma cells displayed a giant cell morphology and were binucleate, indicative of a trophoblast-like morphology. Other trophoblast-like properties included the intermediary filament Troma-1/cytokeratin 8 and the transcription factor Tead4. A decrease in CpG methylation at three sites in the TEAD4 promoter and the B1 repeated sequence was observed. Knocking down expression of Tead4 with siRNA blocked the increase in Troma-1/cytokeratin 8 and over expression of Tead4 induced the expression of Troma-1/cytokeratin 8. Cells treated for 5days with hydralazine were no longer capable of undergoing retinoic acid-mediated neuronal differentiation. An irreversible loss of the pluripotent transcription factor Oct-4 was observed following hydralazine exposure. In summary, hydralazine induces P19 cells to assume a trophoblast-like phenotype by upregulating Tead4 expression through a mechanism involving DNA demethylation.


Subject(s)
Cell Differentiation/drug effects , DNA Methylation/drug effects , Embryonal Carcinoma Stem Cells/pathology , Gene Expression Regulation, Developmental/drug effects , Hydralazine/pharmacology , Trophoblasts/pathology , Animals , Apoptosis/drug effects , Blotting, Western , Cell Proliferation/drug effects , DNA-Binding Proteins/genetics , DNA-Binding Proteins/metabolism , Embryonal Carcinoma Stem Cells/drug effects , Embryonal Carcinoma Stem Cells/metabolism , Immunoenzyme Techniques , Keratin-8/genetics , Keratin-8/metabolism , Mice , Muscle Proteins/genetics , Muscle Proteins/metabolism , Neurons/metabolism , Neurons/pathology , Phenotype , RNA, Messenger/genetics , Real-Time Polymerase Chain Reaction , Reverse Transcriptase Polymerase Chain Reaction , TEA Domain Transcription Factors , Transcription Factors/genetics , Transcription Factors/metabolism , Tretinoin/pharmacology , Trophoblasts/drug effects , Trophoblasts/metabolism , Tumor Cells, Cultured
9.
Pediatr Neurol ; 47(5): 337-40, 2012 Nov.
Article in English | MEDLINE | ID: mdl-23044014

ABSTRACT

The study examined whether maternal serum antibodies from mothers of autistic children preferentially bind to lymphocytes of their autistic children compared with unaffected siblings. In a previous study, maternal serum antibodies from mothers mediated cytotoxicity with complement to lymphocytes of their autistic children. Here, maternal serum antibody binding was examined by flow cytometry. We compared levels of mothers' serum binding against peripheral blood monocytes of their autistic children vs unaffected siblings. Because the level of binding to peripheral blood monocytes could be low, binding was examined in specific lymphocyte subpopulations. In 19 samples, the mean level of maternal serum immunoglobulin G binding to CD4 and CD8 T cells, B cells, natural killer cells, and macrophages was not significantly different from the mean level of binding to unaffected siblings. The percentages of different subpopulations were not significantly different between autistic children and unaffected siblings, although a trend (P < 0.1) emerged, i.e., autistic children displayed a higher percentage of natural killer cells and a lower percentage of B cells. These findings cast doubt on a direct effect of maternal antibodies, but do not preclude potential intrauterine pathogenic immune mechanisms in autism.


Subject(s)
Autistic Disorder/immunology , CD4-Positive T-Lymphocytes/immunology , CD8-Positive T-Lymphocytes/immunology , Histocompatibility, Maternal-Fetal/immunology , Adult , B-Lymphocytes/immunology , B-Lymphocytes/metabolism , Binding Sites, Antibody/immunology , CD4-Positive T-Lymphocytes/metabolism , CD8-Positive T-Lymphocytes/metabolism , Child , Child, Preschool , Female , Humans , Killer Cells, Natural/immunology , Killer Cells, Natural/metabolism , Leukocytes, Mononuclear/immunology , Leukocytes, Mononuclear/metabolism , Male , Middle Aged , Siblings
10.
J Toxicol Environ Health A ; 75(1): 25-35, 2012.
Article in English | MEDLINE | ID: mdl-22047161

ABSTRACT

The aim of this study was to determine whether multiwalled carbon nanotubes (MWNCT) are taken up by and are toxic to human intestinal enterocytes using the Caco-2 cell model. Caco-2 cells were exposed to 50 µg/ml MWCNT (oxidized or pristine) for 24 h, and experiments were repeated in the presence of 2.5 mg/L natural organic matter. Cells displayed many of the properties that characterize enterocytes, such as apical microvilli, basolateral basement membrane, and glycogen. The cell monolayers also displayed tight junctions and electrical resistance. Exposure to pristine and oxidized MWCNT, with or without natural organic matter, did not markedly affect viability, which was assessed by measuring activity of released lactate dehydrogenase (LDH) and staining with propidium iodide. Ultrastructural analysis revealed some damage to microvilli colocalized with the MWCNT; however, neither type of MWCNT was taken up by Caco-2 cells. In contrast, pristine and oxidized MWCNT were taken up by the macrophage RAW 264.7 line. Our study suggests that intestinal enterocytes cells do not take up MWCNT.


Subject(s)
Enterocytes/drug effects , Nanotubes, Carbon/toxicity , Caco-2 Cells , Cell Line , Cell Membrane/drug effects , Cell Membrane/metabolism , Cell Membrane/ultrastructure , Enterocytes/metabolism , Enterocytes/ultrastructure , Humans , L-Lactate Dehydrogenase/metabolism , Microvilli/drug effects , Microvilli/metabolism , Microvilli/ultrastructure , Nanotubes, Carbon/chemistry , Nanotubes, Carbon/ultrastructure , Propidium/chemistry , Tight Junctions/drug effects , Time Factors
11.
Methods Mol Biol ; 758: 153-68, 2011.
Article in English | MEDLINE | ID: mdl-21815064

ABSTRACT

The blood brain barrier regulates the transport of chemicals from entering and leaving the brain. Brain capillaries establish the barrier and restrict transport into the brain by providing a physical and chemical barrier. The physical barrier is due to tight membrane junctions separating the capillary endothelial cells resulting in limited paracellular transport. The chemical barrier is due to the expression of multidrug transporters that mediate the efflux of a broad range of hydrophobic chemicals. Because of the unusual nutrient demands of the brain, this limited permeability is compensated by the expression of a large number of transporters that are responsive to the metabolic demands of the brain. Consequently, the blood brain barrier indirectly regulates brain function by directly controlling the uptake of nutrients. Two widely used methods for studying the blood brain are a cell culture model using rat, pig, or cow brain endothelial cells and isolated microvessels. The cell culture model is more popular likely because it is easier to use and less costly compared to isolated microvessels. In some laboratories, brain endothelial cells are cocultured with astrocyte- or astroglial-conditioned media. The endothelial cells express many of the transporters displayed in vivo but not all. Although cell culture models vary, none express the tight barrier observed in vivo. Because microvessels are isolated directly from the brain, they express all of the transporters displayed in vivo. Their disadvantage is that the preparation is laborious, requires animals, and has a shorter lifespan in vitro. We present an approach in which transport is first verified in isolated microvessels, and then the mechanism is studied in cell culture.


Subject(s)
Blood-Brain Barrier/cytology , Animals , Biological Transport , Blood-Brain Barrier/metabolism , Brain/blood supply , Capillary Permeability , Cell Line , Electrophysiological Phenomena , Endothelial Cells/metabolism , In Vitro Techniques , Mice , Microvessels/cytology , Microvessels/metabolism , Rats
12.
J Pediatr Neurol ; 9(1): 1-8, 2011 Mar.
Article in English | MEDLINE | ID: mdl-29056860

ABSTRACT

Fluoxetine, a selective serotonin reuptake inhibitor (SSRI), has shown favorable effects in some children with autism. There are no previous studies evaluating the connection between clinical outcome and markers of clinical response to fluoxetine treatment. We examined serum brain derived neurotrophic factor (BDNF) concentrations and serotonin transporter (SERT) binding in the medial frontal cortex and midbrain, measured by single photon emission computed tomography (SPECT) scanning, in a group of 13 autistic children and adolescents (12 males, one female; age 5-16 years), who were treated for six months with fluoxetine at a dose range of 10-40 mg/day. Clinical response was evaluated by the Autism Treatment Evaluation Checklist (ATEC). Serum concentrations of BDNF and SERT binding were measured at baseline and two months after termination of fluoxetine treatment. At baseline, before starting fluoxetine treatment, the serum concentration of BDNF had a bimodal distribution in the autism group with either a low concentration (n = 8, mean 1497 pg/mL) or a high concentration (n = 5, mean 14062 pg/mL) with respect to controls (n = 15, mean 9652 pg/mL), and SERT binding was uniformly low in the autistic subjects in medial frontal cortex and midbrain. Fluoxetine treatment led to positive effects in several aspects of communication, socialization and cognitive awareness, with 6 out 13 subjects being particularly good responders. These six also had a significant decrease in BDNF (p = 0.03) and minimal change in SERT binding after therapy. The other 7 subjects showed a trend towards an increase in BDNF and SERT binding. Our results indicate that fluoxetine may improve core autistic symptoms, and that this clinical response is linked to a decrease in serum BDNF.

13.
PLoS One ; 5(9): e12911, 2010 Sep 23.
Article in English | MEDLINE | ID: mdl-20886113

ABSTRACT

BACKGROUND: Neurons are more likely to die through apoptosis in the immature brain after injury whereas adult neurons in the mature brain die by necrosis. Several studies have suggested that this maturational change in the mechanism of cell death is regulated, in part, by thyroid hormone. We examined the involvement of the hairless (Hr) gene which has been suspected of having a role in cell cycle regulation and apoptosis in the hair follicle and is strongly regulated by the thyroid hormone in the brain. METHODOLOGY: Forced expression of Hr by transfection decreased the number of apoptotic nuclei, levels of caspase-3 activity, and cytosolic cytochrome C in COS cells exposed to staurosporine and tunicamycin. Similarly, caspase-3 activity was lower and the decrease in mitochondrial membrane potential was smaller in cultures of adult cerebellar granule neurons from wild type mice compared to Hr knockout mice induced to undergo apoptosis. In vivo, apoptosis as detected by positive TUNEL labeling and caspase 3 activity was lower in wild-type mice compared to Hr knockouts after exposure to trimethyltin. Hr expression lowered levels of p53, p53 mediated reporter gene activity, and lower levels of the pro-apoptotic Bcl2 family member Bax in COS cells. Finally, Hr expression did not attenuate apoptosis in mouse embryonic fibroblasts from p53 knockout mice but was effective in mouse embryonic fibroblasts from wild type mice. CONCLUSIONS/SIGNIFICANCE: Overall, our studies demonstrate that Hr evokes an anti-apoptotic response by repressing expression of p53 and pro-apoptotic events regulated by p53.


Subject(s)
Apoptosis , Transcription Factors/genetics , Tumor Suppressor Protein p53/metabolism , Animals , Brain/cytology , Brain/metabolism , COS Cells , Cells, Cultured , Chlorocebus aethiops , Gene Expression , Humans , Mice , Mice, Hairless , Mice, Knockout , Proto-Oncogene Proteins c-bcl-2/genetics , Proto-Oncogene Proteins c-bcl-2/metabolism , Transcription Factors/metabolism , Tumor Suppressor Protein p53/genetics
14.
Cancer Res ; 67(19): 9389-97, 2007 Oct 01.
Article in English | MEDLINE | ID: mdl-17909048

ABSTRACT

Bioluminescence imaging (BLI) is becoming indispensable to the study of transgene expression during development and, in many in vivo models of disease such as cancer, for high throughput drug screening in vitro. Because reaction of d-luciferin with firefly luciferase (fLuc) produces photons of sufficiently long wavelength to permit imaging in intact animals, use of this substrate and enzyme pair has become the method of choice for performing BLI in vivo. We now show that expression of the ATP-binding cassette (ABC) family transporter ABCG2/BCRP affects BLI signal output from the substrate d-luciferin. In vitro studies show that d-luciferin is a substrate for ABCG2/BCRP but not for the MDR1 P-glycoprotein (ABCB1/Pgp), multidrug resistance protein 1 (MRP1/ABCC1), or multidrug resistance protein 2 (MRP2/ABCC2). d-Luciferin uptake within cells is shown to be modulated by ABC transporter inhibitors, including the potent and selective ABCG2/BCRP inhibitor fumitremorgin C. Images of xenografts engineered to express transgenic ABCG2/BCRP, as well as xenografts derived from the human prostate cancer cell line 22Rv1 that naturally express ABCG2/BCRP, show that ABCG2/BCRP expression and function within regions of interest substantially influence d-luciferin-dependent bioluminescent output in vivo. These findings highlight the need to consider ABCG2/BCRP effects during d-luciferin-based BLI and suggest novel high throughput methods for identifying new ABCG2/BCRP inhibitors.


Subject(s)
ATP-Binding Cassette Transporters/biosynthesis , Firefly Luciferin/metabolism , Neoplasm Proteins/biosynthesis , ATP Binding Cassette Transporter, Subfamily G, Member 2 , Animals , Cell Line, Tumor , Dogs , Female , Firefly Luciferin/analysis , Humans , Luciferases, Firefly/metabolism , Luminescent Agents/analysis , Luminescent Agents/metabolism , Luminescent Measurements , Male , Mice , Mice, Nude , Multidrug Resistance-Associated Protein 2 , Prostatic Neoplasms/metabolism , Substrate Specificity
15.
Hum Exp Toxicol ; 26(3): 221-9, 2007 Mar.
Article in English | MEDLINE | ID: mdl-17439925

ABSTRACT

The transport of essential metals and other nutrients across tight membrane barriers such as the gastrointestinal tract and blood-brain barrier is mediated by specific transport mechanisms. Specific transporters take up metals at the apical surface and export them at the basolateral surface, and are involved in their intracellular distribution. Transporters for each of the major essential metals, calcium, iron and zinc, have been identified. These transporters also mediate the transport of non-essential metals across tight membrane barriers. For example, the intestinal iron transporter divalent metal transporter 1 mediates the uptake of lead and cadmium. The levels of essential metals are strictly regulated by transporters. When dietary levels of essential metals are low, levels of the corresponding transporters increase in the intestine, after which there is a greater potential for increased transport of toxic metals. In the brain, the strict regulation of metals prevents injury that potentially would result from oxidative damage induced by the essential metals iron, copper and zinc. Indeed, the oxidative damage found in neurodegenerative diseases is likely to be due to higher levels of these metals. Involvement of intracellular transporters for copper and zinc has been shown in animal models of Alzheimer's disease, raising the possibility that higher levels of iron, zinc and copper might be due to a disruption in the activity of transporters. Accordingly, exposure to toxicants that affect the activity of transporters potentially could contribute to the aetiology/progression of neurodegenerative diseases.


Subject(s)
Blood-Brain Barrier/metabolism , Intestinal Mucosa/metabolism , Membrane Transport Proteins/metabolism , Metals/toxicity , Animals , Environmental Exposure/adverse effects , Environmental Pollutants/toxicity , Homeostasis/drug effects , Humans , Neurodegenerative Diseases/chemically induced , Neurodegenerative Diseases/metabolism , Neurotoxicity Syndromes/metabolism
16.
Toxicol Appl Pharmacol ; 220(3): 349-56, 2007 May 01.
Article in English | MEDLINE | ID: mdl-17376497

ABSTRACT

Aluminum and other trivalent metals were shown to stimulate uptake of transferrin bound iron and nontransferrin bound iron in erytholeukemia and hepatoma cells. Because of the association between aluminum and Alzheimer's Disease, and findings of higher levels of iron in Alzheimer's disease brains, the effects of aluminum on iron homeostasis were examined in a human glial cell line. Aluminum stimulated dose- and time-dependent uptake of nontransferrin bound iron and iron bound to transferrin. A transporter was likely involved in the uptake of nontransferrin iron because uptake reached saturation, was temperature-dependent, and attenuated by inhibitors of protein synthesis. Interestingly, the effects of aluminum were not blocked by inhibitors of RNA synthesis. Aluminum also decreased the amount of iron bound to ferritin though it did not affect levels of divalent metal transporter 1. These results suggest that aluminum disrupts iron homeostasis in the brain by several mechanisms including the transferrin receptor, a nontransferrin iron transporter, and ferritin.


Subject(s)
Aluminum Compounds/pharmacology , Chlorides/pharmacology , Iron/metabolism , Neuroglia/drug effects , Transferrin/metabolism , 4,4'-Diisothiocyanostilbene-2,2'-Disulfonic Acid/pharmacology , Adult , Aluminum Chloride , Apoproteins/metabolism , Cell Line, Tumor , Cinnamates/pharmacology , Citrates/pharmacology , Dose-Response Relationship, Drug , Ferrous Compounds/pharmacology , Humans , Iron/pharmacokinetics , Methionine/metabolism , Neuroglia/metabolism , Neuroglia/pathology , Nucleic Acid Synthesis Inhibitors/pharmacology , Protein Binding , Protein Synthesis Inhibitors/pharmacology , Receptors, Transferrin/metabolism , Time Factors , Tin/pharmacology , Transferrin/pharmacokinetics , Tunicamycin/pharmacology
17.
Methods Mol Biol ; 341: 1-15, 2006.
Article in English | MEDLINE | ID: mdl-16799185

ABSTRACT

This chapter describes in vitro methods for studying the blood-brain barrier. These methods include a cell line and isolated brain microvessels. The rat brain endothelial cell line 4 (RBE4) express many properties that are expressed by brain endothelial cells in vivo. Tissue culture methods allow the investigator to design experiments for studying transporters and permeability that would be much more difficult in vivo. A method for making preparations of isolated brain microvessels also is described. These preparations are highly enriched and also can be used for studying transport in vitro, but their short life span is a limitation. Two methods are discussed for measuring transport in cell culture. In one method, permeability is measured across a cell mono-layer. This method is useful for measuring luminal and abluminal transport. The second method is especially designed for measuring the families of efflux transporters. These in vitro methods will complement many of the in vivo techniques, and they may be used as screening for more timely and expensive experiments, and also reducing the need for experimental animals.


Subject(s)
Blood-Brain Barrier/physiology , Cell Communication/physiology , Endothelial Cells/physiology , Membrane Transport Proteins/metabolism , Animals , Biological Transport, Active/physiology , Blood-Brain Barrier/cytology , Cell Culture Techniques/methods , Cell Line , Cell Membrane Permeability/physiology , Endothelial Cells/cytology , Rats
18.
Front Biosci ; 10: 160-5, 2005 Jan 01.
Article in English | MEDLINE | ID: mdl-15574358

ABSTRACT

We have characterized membrane-associated substrates of Ca2+-dependent kinases in primary rat astrocytes by in vitro phosphorylation, 2-dimensional gel electrophoresis and autoradiography. The most prominent among these were three acidic, protein kinase C (PKC) substrates. These are important because they likely transduce cytokine and other neuro-immune modulatory signals mediated by PKC. We now show that one of these phosphoproteins is myristoylated alanine-rich PKC kinase substrate (MARCKS) or phosphomyristin C. The identity was corroborated by one- and 2- dimensional immunoblotting with an MARCKS-specific polyclonal antibody. Exposing primary astrocytes to phorbol 12-myristate 13-acetate stimulated phosphorylation of this protein. The level of MARCKS appeared inversely proportional to the proliferative potential of astrocytes because it was lower in spontaneously transformed as compared to passaged or confluent cells. These data are consistent with previous reports and indicate that one of three major acidic membrane-associated PKC substrates in astrocytes is MARCKS. Thus, MARCKS is likely near-proximal transducer of PKC-mediated signals in astrocytes.


Subject(s)
Astrocytes/cytology , Intracellular Signaling Peptides and Proteins/physiology , Membrane Proteins/physiology , Animals , Astrocytes/metabolism , Autoradiography , Cell Membrane/metabolism , Cell Proliferation , Cells, Cultured , Cytokines/metabolism , Electrophoresis, Gel, Two-Dimensional , GAP-43 Protein/chemistry , Intracellular Signaling Peptides and Proteins/metabolism , Membrane Proteins/metabolism , Myristoylated Alanine-Rich C Kinase Substrate , Neuroglia/metabolism , Protein Kinase C/metabolism , Rats , Tetradecanoylphorbol Acetate/pharmacology
19.
Ann N Y Acad Sci ; 1012: 142-52, 2004 Mar.
Article in English | MEDLINE | ID: mdl-15105261

ABSTRACT

The effect of exposure to cadmium (Cd) and lead (Pb) on human health has been recognized for many years and recent information suggests that minimal exposure levels are themselves too high. Common scenarios for Pb exposure include occupational, residential, and/or behavioral (hand-to-mouth activity) settings. The main source of Cd exposure for nonsmokers is dietary, through plants or animals that accumulate the metal. Specific cellular importers for Pb and Cd are unlikely as these metals are nonessential and toxic. Accordingly, in the intestine, the operational mechanism is assumed to be inadvertent uptake through pathways intended for essential nutrients such as iron. Results from experimental and epidemiological studies indicated that diets low in iron (Fe) result in increased absorption of Pb and Cd, suggesting common molecular mechanisms of Cd and Pb transport. Indeed, recent mechanistic studies found that the intestinal transporter for nonheme iron, divalent metal transporter 1 (DMT1), mediates the transport of Pb and Cd. DMT1 is regulated, in part, by dietary iron, and chemical species of Cd and Pb that are transported by DMT1 would be made available through digestion and are also found in plasma. Accordingly, the involvement of DMT1 in metal uptake offers a mechanistic explanation for why an iron-deficient diet is a risk factor for Pb and Cd poisoning. It also suggests that diets rich in iron-containing food could be protective against heavy metal poisoning.


Subject(s)
Cadmium/metabolism , Cation Transport Proteins/metabolism , Ion Transport/physiology , Iron-Binding Proteins/metabolism , Lead/metabolism , Animals , Environmental Exposure , Humans , Intestinal Mucosa/metabolism , Intestines/cytology , Iron/metabolism
20.
Toxicol Sci ; 78(2): 241-7, 2004 Apr.
Article in English | MEDLINE | ID: mdl-14976354

ABSTRACT

The effect of the organophosphorous insecticide paraoxon on the integrity of the blood-brain barrier (BBB) and permeability of pyridostigmine (PYR), a peripheral inhibitor of cholinesterase activity, was examined in Long Evans rats. The integrity of the BBB was examined by measuring the number of capillaries leaking horseradish peroxidase, which was injected into the heart. Treatment with paraoxon at 100 microg/kg, intramuscularly, resulted in a 3- to 4-fold increase in the number of leaky capillaries in young rats (25 to 30 days old) but not in older rats (90 days old). Interestingly, young rats treated with PYR (30 mg/kg, po) 50 min before treatment with paraoxon showed an inhibited effect of paraoxon on the BBB. Furthermore, no increase in the degree of inhibition of acetylcholinesterase activity was observed in young rats treated with PYR before paraoxon compared with young rats treated with paraoxon alone. Cholinergic toxicity, as assessed by changes in behavior, was not observed in young rats treated with paraoxon alone; but, slight signs of cholinergic toxicity were observed in rats treated with PYR. Young rats treated with both PYR and paraoxon did not exhibit more extensive signs of toxicity than rats treated with paraoxon alone or PYR alone. The results indicate that treatment with paraoxon can compromise BBB permeability at dosages that do not induce cholinergic toxicity, but only in young rats. Also, PYR pre-exposure appears to protect the BBB from the paraoxon-induced alterations.


Subject(s)
Behavior, Animal/drug effects , Blood-Brain Barrier/drug effects , Cholinesterase Inhibitors/pharmacology , Insecticides/toxicity , Paraoxon/toxicity , Pyridostigmine Bromide/pharmacology , Age Factors , Animals , Blood-Brain Barrier/metabolism , Brain/enzymology , Brain/metabolism , Capillaries/drug effects , Capillaries/enzymology , Capillaries/physiopathology , Cholinesterase Inhibitors/metabolism , Cholinesterases/analysis , Cholinesterases/metabolism , Drug Interactions , Horseradish Peroxidase/administration & dosage , Horseradish Peroxidase/analysis , Humans , Male , Paraoxon/antagonists & inhibitors , Persian Gulf Syndrome/etiology , Persian Gulf Syndrome/metabolism , Pyridostigmine Bromide/metabolism , Rats , Rats, Long-Evans
SELECTION OF CITATIONS
SEARCH DETAIL