Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 24
Filter
1.
Methods Mol Biol ; 1343: 249-64, 2015.
Article in English | MEDLINE | ID: mdl-26420722

ABSTRACT

Pyrosequencing is a technique that uses a sequencing-by-synthesis system which is designed to quantify single-nucleotide polymorphisms (SNPs). Artificial C/T SNP creation via bisulfite modification permits measurement of DNA methylation locally and globally in real time. Alteration in DNA methylation has been implicated in aging, as well as aging-related conditions such as cancer, as well as cardiovascular, neurodegenerative, and autoimmune diseases. Considering its ubiquitous presence in divergent clinical pathologies, quantitative analysis of DNA CpG methylation both globally and at individual genes helps to elucidate the regulation of genes involved in pathophysiological conditions. The ability to detect and quantify the methylation pattern of DNA has the potential to serve as an early detection marker and potential drug target for several diseases. Here, we provide a detailed technical protocol for pyrosequencing supplemented by critical information about assay design and nuances of the system that provides a strong foundation for beginners in the field.


Subject(s)
DNA Methylation , Epigenomics , Sequence Analysis, DNA , Epigenomics/methods , Sequence Analysis, DNA/methods
2.
Crit Rev Immunol ; 34(1): 1-14, 2014.
Article in English | MEDLINE | ID: mdl-24579699

ABSTRACT

Adipose tissue historically was believed to be an inert tissue, functioning primarily in the storage of energy and thermal homeostasis. However, recent discoveries point toward a critical role for adipocytes in endocrine function as well as immune regulation. Excess body fat, accumulated through aging and/or a calorie-rich diet, is associated with many chronic metabolic and inflammatory diseases. Within the stromal vascular fraction of adipose tissue, macrophages and T cells accumulate with increasing tissue mass, secreting pro- or anti-inflammatory cytokines. In this review we discuss the current understanding of immune cell function in both diet-induced and age-related obesity. In both models of obesity, the classically activated, pro-inflammatory (M1) subtype takes precedence over the alternatively activated, anti-inflammatory (M2) macrophages, causing tissue necrosis and releasing pro-inflammatory cytokines like interleukin-6. Other distinct adipose tissue macrophage subtypes have been identified by surface marker expression and their functions characterized. Adipose tissue T cell recruitment to adipose tissue is also different between aging- and diet-induced obesity. Under both conditions, T cells exhibit restricted T-cell receptor diversity and produce higher levels of pro-inflammatory signals like interferon-γ and granzyme B relative to young or healthy mice. However, numbers of regulatory T cells are dramatically different between the 2 models of obesity. Taken together, these findings suggest models of age- and diet-induced obesity may be more distinct than previously thought, with many questions yet to be resolved in this multidimensional disease.


Subject(s)
Adipose Tissue/immunology , Aging/immunology , Macrophages/immunology , Obesity/immunology , T-Lymphocytes/immunology , Animals , Cytokines/metabolism , Humans , Inflammation Mediators/metabolism , Mice
3.
Aging Cell ; 13(3): 441-8, 2014 Jun.
Article in English | MEDLINE | ID: mdl-24325345

ABSTRACT

Regulatory T-cell (Treg, CD4(+) CD25(+)) dysfunction is suspected to play a key role in immune senescence and contributes to increased susceptibility to diseases with age by suppressing T-cell responses. FoxP3 is a master regulator of Treg function, and its expression is under control of several epigenetically labile promoters and enhancers. Demethylation of CpG sites within these regions is associated with increased FoxP3 expression and development of a suppressive phenotype. We examined differences in FoxP3 expression between young (3-4 months) and aged (18-20 months) C57BL/6 mice. DNA from CD4(+) T cells is hypomethylated in aged mice, which also exhibit increased Treg numbers and FoxP3 expression. Additionally, Treg from aged mice also have greater ability to suppress effector T-cell (Teff) proliferation in vitro than Tregs from young mice. Tregs from aged mice exhibit greater redox remodeling-mediated suppression of Teff proliferation during coculture with DCs by decreasing extracellular cysteine availability to a greater extent than Tregs from young mice, creating an adverse environment for Teff proliferation. Tregs from aged mice produce higher IL-10 levels and suppress CD86 expression on DCs more strongly than Tregs from young mice, suggesting decreased T-cell activity. Taken together, these results reveal a potential mechanism of higher Treg-mediated activity that may contribute to increased immune suppression with age.


Subject(s)
Aging/physiology , CD4-Positive T-Lymphocytes/immunology , CD8-Positive T-Lymphocytes/immunology , T-Lymphocytes, Regulatory/immunology , Animals , Cell Proliferation/physiology , Epigenomics , Forkhead Transcription Factors/metabolism , Male , Methylation , Mice , Mice, Inbred C57BL , Oxidation-Reduction
4.
PLoS One ; 8(2): e56253, 2013.
Article in English | MEDLINE | ID: mdl-23437105

ABSTRACT

Atherosclerosis is an inflammatory condition of the arterial wall mediated by cells of both innate and adaptive immunity. T lymphocytes play an important role in orchestrating the pathogenic immune response involved in the acceleration of atherosclerosis. Previously, we have shown that a prenatal methyl-donor supplementation diet (MS), when fed to dams during pregnancy and lactation, decreased the T cell-mediated pro-inflammatory cytokine and chemokine response in F1 mice. In the current study, we report feeding Apolipoprotein E (ApoE(-/-)) deficient dams with the MS diet during pregnancy reduces atherosclerotic plaques in F1 mice that were fed high fat diet (HFD) after weaning. F1 mice from dams on the MS diet exhibited increased global T cell DNA methylation. T-cell chemokines and their receptors (in particular CCR2, CCR5, and CXCR3) play important roles in the inflammatory cell recruitment to vascular lesions. MS diet significantly reduced Ccr2 mRNA and protein expression in CD3+ T cells but not in CD11b+ monocytes in MS F1 mice relative to controls. F1 litter size, HFD consumption, body weight, and body fat were similar between control and MS diet groups. Moreover, serum thiol metabolite levels were similar between the two groups. However, MS diet is associated with significantly higher serum HDL and lower LDL+VLDL levels in comparison to F1 mice from dams on the control diet. Inflammatory cytokines (IL-17, TNF-α, IL-6) were also lower in MS F1 mice serum and conditioned media from T-cell culture. Altogether, these data suggest that the MS diet ameliorates development of atherosclerosis by inhibiting the T-cell Ccr2 expression, reducing inflammatory cytokines production and increasing serum HDL:LDL ratio.


Subject(s)
Apolipoproteins E/deficiency , Atherosclerosis/prevention & control , Crosses, Genetic , Diet , Dietary Supplements , Animals , Aorta/pathology , Apolipoproteins E/metabolism , Atherosclerosis/blood , Atherosclerosis/pathology , Body Composition , Cholesterol/biosynthesis , Diet, High-Fat , Feeding Behavior , Female , Gene Expression Regulation , Inflammation Mediators/metabolism , Lipoproteins/blood , Litter Size , Liver/metabolism , Liver/pathology , Lymphocyte Activation/immunology , Male , Methylation , Mice , Monocytes/metabolism , Receptors, CCR2/metabolism , Sulfhydryl Compounds/metabolism , T-Lymphocytes/metabolism , Weight Gain
5.
Sci Rep ; 3: 1045, 2013.
Article in English | MEDLINE | ID: mdl-23304436

ABSTRACT

Vimentin, an abundant intermediate filament protein, presumably has an important role in stabilizing intracellular architecture, but its function is otherwise poorly understood. In a vimentin knockout (Vim KO) mouse model, we note that Vim KO mice challenged with intraperitoneal Escherichia coli control bacterial infection better than do wild-type (WT) mice. In vitro, Vim KO phagocytes show significantly increased capacity to mediate bacterial killing by abundant production of reactive oxygen species (ROS) and nitric oxides, likely due to interactions with the p47phox active subunit of NADPH oxidase. In acute colitis induced by dextran sodium sulfate (DSS), Vim KO mice develop significantly less gut inflammation than do WT mice. Further, Vim KO mice have markedly decreased bacterial extravasation in the setting of DSS-induced acute colitis, consistent with decreased intestinal disease. Our results suggest that vimentin impedes bacterial killing and production of ROS, thereby contributing to the pathogenesis of acute colitis.


Subject(s)
Colitis/metabolism , Vimentin/metabolism , Animals , Colitis/chemically induced , Colitis/pathology , Dextran Sulfate/toxicity , Escherichia coli/pathogenicity , Macrophages/immunology , Macrophages/metabolism , Mice , Mice, Knockout , NADPH Oxidases/metabolism , Nitric Oxide/metabolism , Phagocytosis , RNA Interference , RNA, Small Interfering/metabolism , Reactive Oxygen Species/metabolism , Vimentin/antagonists & inhibitors , Vimentin/genetics
6.
Biochim Biophys Acta ; 1822(11): 1671-81, 2012 Nov.
Article in English | MEDLINE | ID: mdl-22820549

ABSTRACT

Alzheimer's disease (AD) is associated with impaired glutamate clearance and depressed Na(+)/K(+) ATPase levels in AD brain that might lead to a cellular ion imbalance. To test this hypothesis, [Na(+)] and [K(+)] were analyzed in postmortem brain samples of 12 normal and 16 AD individuals, and in cerebrospinal fluid (CSF) from AD patients and matched controls. Statistically significant increases in [Na(+)] in frontal (25%) and parietal cortex (20%) and in cerebellar [K(+)] (15%) were observed in AD samples compared to controls. CSF from AD patients and matched controls exhibited no differences, suggesting that tissue ion imbalances reflected changes in the intracellular compartment. Differences in cation concentrations between normal and AD brain samples were modeled by a 2-fold increase in intracellular [Na(+)] and an 8-15% increase in intracellular [K(+)]. Since amyloid beta peptide (Aß) is an important contributor to AD brain pathology, we assessed how Aß affects ion homeostasis in primary murine astrocytes, the most abundant cells in brain tissue. We demonstrate that treatment of astrocytes with the Aß 25-35 peptide increases intracellular levels of Na(+) (~2-3-fold) and K(+) (~1.5-fold), which were associated with reduced levels of Na(+)/K(+) ATPase and the Na(+)-dependent glutamate transporters, GLAST and GLT-1. Similar increases in astrocytic Na(+) and K(+) levels were also caused by Aß 1-40, but not by Aß 1-42 treatment. Our study suggests a previously unrecognized impairment in AD brain cell ion homeostasis that might be triggered by Aß and could significantly affect electrophysiological activity of brain cells, contributing to the pathophysiology of AD.


Subject(s)
Alzheimer Disease/metabolism , Brain , Potassium , Sodium-Potassium-Exchanging ATPase/metabolism , Sodium , Alzheimer Disease/cerebrospinal fluid , Amino Acid Transport System X-AG/metabolism , Amyloid beta-Peptides/pharmacology , Animals , Astrocytes/metabolism , Brain/metabolism , Brain/physiopathology , Excitatory Amino Acid Transporter 2/metabolism , Glutamic Acid/cerebrospinal fluid , Glutamic Acid/metabolism , HEK293 Cells , Humans , Jurkat Cells , Mice , Potassium/cerebrospinal fluid , Potassium/metabolism , Sodium/cerebrospinal fluid , Sodium/metabolism
7.
J Nutr ; 142(7): 1329-35, 2012 Jul.
Article in English | MEDLINE | ID: mdl-22649261

ABSTRACT

Prenatal environmental exposures play a critical role in determining late-life chronic disease susceptibility. However, the mechanisms linking the in utero environment and disease development in the offspring are poorly understood. Recent investigations have confirmed a central pathogenic role of T cell chemokine receptors, particularly C-C chemokine receptor (CCR) 2 and CCR5, in chronic inflammatory conditions. This study was designed to determine the effect of a synthetic prenatal micronutrient supplementation (MS) diet rich in methionine pathway metabolites on the T cell chemokine system in F1 C57Bl/6 mice. Female mice were fed either an MS or control diet 3 wk prior to mating, during pregnancy, and lactation. At 4 wk of age, F1 mice were killed for experiments or were fed the standard NIH-31 diet and allowed to age. Food consumption, maternal weight gain, and litter size were similar in dams fed the control and MS diets. However, the F1 offspring of dams fed the MS diet were smaller in size (P < 0.001). T cells from the MS F1 offspring had global hypermethylation compared with control F1 offspring (P < 0.005), corresponding to lower T cell chemokine receptor expression [CCR2 (P < 0.001), CCR5 (P < 0.001), and C-x-C chemokine receptor 3 (P < 0.01)] and cytokine expression [TNFα (P < 0.05), IL-2 (P < 0.001), and IL-4 (P < 0.01)]. Reduced T cell chemokine receptor gene expression in MS F1 mice was associated with decreased chemotaxis in vitro to C-C chemokine ligand (CCL) 2 and C-X-C chemokine ligand 10 (P < 0.01) and in vivo to CCL2 (P < 0.01). Taken together, the results suggest that epigenetic alteration through prenatal diet manipulation reduces the response to proinflammatory signals in mice.


Subject(s)
Gene Expression/drug effects , Growth/drug effects , Inflammation/prevention & control , Micronutrients/pharmacology , Prenatal Nutritional Physiological Phenomena/drug effects , Receptors, Antigen, T-Cell/metabolism , Receptors, Chemokine/metabolism , Animals , Chemotaxis/drug effects , Cytokines/genetics , Cytokines/metabolism , DNA Methylation/drug effects , Diet , Dietary Supplements , Epigenesis, Genetic , Female , Growth/genetics , Inflammation/genetics , Inflammation/metabolism , Methionine/metabolism , Mice , Mice, Inbred C57BL , Pregnancy , Prenatal Care , Prenatal Nutritional Physiological Phenomena/immunology , Receptors, Antigen, T-Cell/antagonists & inhibitors , Receptors, Antigen, T-Cell/genetics , Receptors, Chemokine/antagonists & inhibitors , Receptors, Chemokine/genetics , Signal Transduction/drug effects
8.
J Biol Chem ; 287(10): 7603-14, 2012 Mar 02.
Article in English | MEDLINE | ID: mdl-22215680

ABSTRACT

The integrated stress response mediated by eukaryotic translation initiation factor 2α (eIF2α) phosphorylation maintains cellular homeostasis under endoplasmic reticulum (ER) stress. eIF2α phosphorylation induces activating transcription factor 4 (ATF4), a basic leucine zipper transcription factor that regulates the expression of genes responsible for amino acid metabolism, cellular redox state, and anti-stress responses. Cystathionine γ-lyase (CSE) and cystathionine ß-synthase are critical enzymes in the transsulfuration pathway, which also regulate cellular redox status by modulating glutathione (GSH) levels. To determine the link between the integrated stress response and the transsulfuration pathway, we used homocysteine (Hcy) as an inducer of eIF2α phosphorylation and ATF4 gene induction. Mouse embryonic fibroblasts (MEFs) lacking ATF4 (ATF4(-/-)) had reduced GSH levels and increased reactive oxygen species and were susceptible to apoptotic cell death under normal culture conditions. Further, ATF4(-/-) MEFs were more sensitive to Hcy-induced cytotoxicity and showed significantly reduced intracellular GSH levels associated with apoptosis. ATF4(-/-) MEFs could be rescued from l-Hcy-induced apoptosis by ß-mercaptoethanol medium supplementation that increases cysteine levels and restores GSH synthesis. ATF4(-/-) MEFs showed little or no CSE protein but did express cystathionine ß-synthase. Further, ER stress-inducing agents, including tunicamycin and thapsigargin, induced the expression of CSE in ATF4(+/+) MEFs. Consistent with ATF4(-/-) MEFs, CSE(-/-) MEFs showed significantly greater apoptosis when treated with tunicamycin, thapsigargin, and l-Hcy, compared with CSE(+/+) MEFs. Liver and kidney GSH levels were also reduced in CSE(-/-) mice, suggesting that CSE is a critical factor in GSH synthesis and may act to protect the liver and kidney from a variety of conditions that cause ER stress.


Subject(s)
Endoplasmic Reticulum Stress/physiology , Fibroblasts/metabolism , Glutathione/metabolism , Homocysteine/metabolism , Lyases/metabolism , Sulfhydryl Compounds/metabolism , Activating Transcription Factor 4/genetics , Activating Transcription Factor 4/metabolism , Animals , Apoptosis/physiology , Cells, Cultured , Cystathionine gamma-Lyase , Embryo, Mammalian/cytology , Embryo, Mammalian/metabolism , Fibroblasts/cytology , Glutathione/genetics , Homocysteine/genetics , Kidney/metabolism , Liver/metabolism , Lyases/genetics , Mice , Mice, Knockout , Oxidation-Reduction
9.
Exp Neurol ; 233(1): 333-41, 2012 Jan.
Article in English | MEDLINE | ID: mdl-22079587

ABSTRACT

It is widely believed that microglia and monocyte-derived macrophages (collectively referred to as central nervous system (CNS) macrophages) cause excitotoxicity in the diseased or injured CNS. This view has evolved mostly from in vitro studies showing that neurotoxic concentrations of glutamate are released from CNS macrophages stimulated with lipopolysaccharide (LPS), a potent inflammogen. We hypothesized that excitotoxic killing by CNS macrophages is more rigorously controlled in vivo, requiring both the activation of the glutamate/cystine antiporter (system x(c)(-)) and an increase in extracellular cystine, the substrate that drives glutamate release. Here, we show that non-traumatic microinjection of low-dose LPS into spinal cord gray matter activates CNS macrophages but without causing overt neuropathology. In contrast, neurotoxic inflammation occurs when LPS and cystine are co-injected. Simultaneous injection of NBQX, an antagonist of AMPA glutamate receptors, reduces the neurotoxic effects of LPS+cystine, implicating glutamate as a mediator of neuronal cell death in this model. Surprisingly, neither LPS nor LPS+cystine adversely affects survival of oligodendrocytes or oligodendrocyte progenitor cells. Ex vivo analyses show that redox balance in microglia and macrophages is controlled by induction of system x(c)(-) and that high GSH:GSSG ratios predict the neurotoxic potential of these cells. Together, these data indicate that modulation of redox balance in CNS macrophages, perhaps through regulating system x(c)(-), could be a novel approach for attenuating injurious neuroinflammatory cascades.


Subject(s)
Excitatory Amino Acids/toxicity , Glutamic Acid/metabolism , Macrophages/metabolism , Microglia/metabolism , Spinal Cord Diseases/chemically induced , Spinal Cord Diseases/pathology , Animals , Cystine/metabolism , Disease Models, Animal , Excitatory Amino Acid Antagonists/pharmacology , Gene Expression Regulation/drug effects , Glutamic Acid/toxicity , Glutathione/metabolism , Laser Capture Microdissection/methods , Lipopolysaccharides/pharmacology , Macrophages/drug effects , Mice , Mice, Inbred C57BL , Microglia/drug effects , Nerve Tissue Proteins/metabolism , Neurons/drug effects , Oxidation-Reduction/drug effects , Quinoxalines/pharmacology
10.
J Biol Chem ; 286(37): 32002-10, 2011 Sep 16.
Article in English | MEDLINE | ID: mdl-21778230

ABSTRACT

The physiological roles of taurine, a product of cysteine degradation and one of the most abundant amino acids in the body, remain elusive. Taurine deficiency leads to heart dysfunction, brain development abnormalities, retinal degradation, and other pathologies. The taurine synthetic pathway is proposed to be incomplete in astrocytes and neurons, and metabolic cooperation between these cell types is reportedly needed to complete the pathway. In this study, we analyzed taurine synthesis capability as reported by incorporation of radioactivity from [(35)S]cysteine into taurine, in primary murine astrocytes and neurons, and in several transformed cell lines (human (SH-SY5Y) and murine (N1E-115) neuroblastoma, human astrocytoma (U-87 MG and 1321 N1), and rat glioma (C6)). Extensive incorporation of radioactivity from [(35)S]cysteine into taurine was observed in rat glioma cells as well as in primary mouse astrocytes and neurons, establishing the presence of an intact taurine synthesis pathway in these cells. Interestingly, exposure of cells to cysteine or cysteamine resulted in elevated intracellular hypotaurine without a corresponding increase in taurine levels, suggesting that oxidation of hypotaurine limits taurine synthesis in cells. Consistent with its role as an organic osmolyte, taurine synthesis was stimulated under hypertonic conditions in neurons.


Subject(s)
Astrocytes/metabolism , Neurons/metabolism , Taurine/biosynthesis , Animals , Astrocytes/cytology , Cell Line, Tumor , Cysteine/metabolism , Humans , Isotope Labeling , Mice , Mice, Inbred BALB C , Neurons/cytology , Osmotic Pressure , Oxidation-Reduction , Rats
11.
Antioxid Redox Signal ; 14(12): 2385-97, 2011 Jun 15.
Article in English | MEDLINE | ID: mdl-21235355

ABSTRACT

Astrocytes are critical for neuronal redox homeostasis providing them with cysteine needed for glutathione synthesis. In this study, we demonstrate that the astrocytic redox response signature provoked by amyloid beta (Aß) is distinct from that of a general oxidant (tertiary-butylhydroperoxide [t-BuOOH]). Acute Aß treatment increased cystathionine ß-synthase (CBS) levels and enhanced transsulfuration flux in contrast to repeated Aß exposure, which decreased CBS and catalase protein levels. Although t-BuOOH also increased transsulfuration flux, CBS levels were unaffected. The net effect of Aß treatment was an oxidative shift in the intracellular glutathione/glutathione disulfide redox potential in contrast to a reductive shift in response to peroxide. In the extracellular compartment, Aß, but not t-BuOOH, enhanced cystine uptake and cysteine accumulation, and resulted in remodeling of the extracellular cysteine/cystine redox potential in the reductive direction. The redox changes elicited by Aß but not peroxide were associated with enhanced DNA synthesis. CBS activity and protein levels tended to be lower in cerebellum from patients with Alzheimer's disease than in age-matched controls. Our study suggests that the alterations in astrocytic redox status could compromise the neuroprotective potential of astrocytes and may be a potential new target for therapeutic intervention in Alzheimer's disease.


Subject(s)
Amyloid beta-Peptides/pharmacology , Astrocytes/drug effects , Astrocytes/metabolism , Oxidation-Reduction , Alzheimer Disease/metabolism , Animals , Antioxidants/pharmacology , Astrocytes/cytology , Cells, Cultured , Cystathionine beta-Synthase/metabolism , Cysteine/metabolism , Cystine/metabolism , DNA/biosynthesis , Dose-Response Relationship, Drug , Homeostasis , Humans , Mice , Mice, Inbred BALB C , Neurons/cytology , Neurons/metabolism , Reactive Oxygen Species/metabolism , tert-Butylhydroperoxide/pharmacology
12.
Antioxid Redox Signal ; 15(1): 39-47, 2011 Jul 01.
Article in English | MEDLINE | ID: mdl-20673163

ABSTRACT

The synthesis of glutathione, a major cellular antioxidant with a critical role in T cell proliferation, is limited by cysteine. In this study, we evaluated the contributions of the x(C)(-) cystine transporter and the transsulfuration pathway to cysteine provision for glutathione synthesis and antioxidant defense in naïve versus activated T cells and in the immortalized T lymphocyte cell line, Jurkat. We show that the x(C)(-) transporter, although absent in naïve T cells, is induced after activation, releasing T cells from their cysteine dependence on antigen-presenting cells. We also demonstrate the existence of an intact transsulfuration pathway in naïve and activated T cells and in Jurkat cells. The flux through the transsulfuration pathway increases in primary but not in transformed T cells in response to oxidative challenge by peroxide. Inhibition of the transsulfuration pathway in both primary and transformed T cells decreases cell viability under oxidative-stress conditions.


Subject(s)
Lymphocyte Activation/drug effects , T-Lymphocytes/drug effects , Animals , Antigen-Presenting Cells/cytology , Antigen-Presenting Cells/drug effects , Blotting, Western , Cell Survival/drug effects , Cells, Cultured , Cysteine/metabolism , Glutathione/metabolism , Humans , Jurkat Cells/drug effects , Male , Mice , Mice, Inbred BALB C , Peroxides/pharmacology , T-Lymphocytes/cytology
13.
J Biol Chem ; 285(53): 41525-32, 2010 Dec 31.
Article in English | MEDLINE | ID: mdl-21037289

ABSTRACT

Naturally occurring CD4(+)CD25(+)Foxp3(+) regulatory T cells (Tregs) suppress proliferation of CD4(+)CD25(-) effector T cells (Teffs) by mechanisms that are not well understood. We have previously demonstrated a novel mechanism of Treg suppression, i.e. interference with extracellular redox remodeling that occurs during activation of T cells by dendritic cells. In this study, we demonstrate that Treg-mediated redox perturbation is antigen-dependent but not antigen-specific, is CTLA-4-dependent, and requires cell-cell contact. Furthermore, we show that Tregs use multiple strategies for extracellular redox remodeling, including diminished GSH synthesis in dendritic cells via decreased expression of γ-glutamylcysteine synthetase, the limiting enzyme for GSH synthesis. Tregs also consume extracellular cysteine and partition it more proficiently to the oxidation product (sulfate), whereas Teffs divert more of the cysteine pool toward protein and GSH synthesis. Tregs appear to block GSH redistribution from the nucleus to the cytoplasm in Teffs, which is abrogated by the addition of exogenous cysteine. Together, these data provide novel insights into modulation of sulfur-based redox metabolism by Tregs, leading to suppression of T cell activation and proliferation.


Subject(s)
Dendritic Cells/cytology , Glutathione/metabolism , T-Lymphocytes, Regulatory/cytology , T-Lymphocytes/cytology , Animals , Antioxidants/chemistry , Cell Proliferation , Cytokines/metabolism , Cytoplasm/metabolism , Immunochemistry/methods , Lymphocyte Activation , Mice , Mice, Inbred BALB C , Mice, Transgenic , Oxidation-Reduction , Sulfhydryl Compounds/chemistry
14.
Nat Chem Biol ; 5(10): 721-3, 2009 Oct.
Article in English | MEDLINE | ID: mdl-19718041

ABSTRACT

We demonstrate that the mechanism of redox remodeling during mouse T-cell activation involves secretion of glutathione by dendritic cells and its subsequent cleavage to cysteine. Extracellular cysteine accumulation results in a lower redox potential, which is conducive to proliferation, and changes the net redox status of exofacial protein domains. Regulatory T cells inhibit this redox metabolite signaling pathway, which represents a previously unrecognized mechanism for immunosuppression of effector T cells.


Subject(s)
Cysteine/metabolism , Dendritic Cells/metabolism , Extracellular Space/metabolism , Glutathione/metabolism , Lymphocyte Activation/immunology , T-Lymphocytes, Regulatory/immunology , Animals , Blotting, Western , Cell Proliferation/drug effects , Cells, Cultured , Coculture Techniques , Cysteine/pharmacology , Dendritic Cells/immunology , Flow Cytometry , Male , Mice , Mice, Inbred BALB C , Oxidation-Reduction , Signal Transduction/drug effects , T-Lymphocytes, Regulatory/metabolism
15.
J Neurochem ; 108(5): 1155-66, 2009 Mar.
Article in English | MEDLINE | ID: mdl-19141080

ABSTRACT

Astrocytes can either exacerbate or ameliorate secondary degeneration at sites of injury in the CNS but the contextual basis for eliciting these opposing phenotypes is poorly understood. In this study, we demonstrate that the two major cytokines produced by Th1 and Th2 cells, interferon-gamma (IFN-gamma), and interleukin-4 (IL-4), respectively, contribute differentially to shaping a neuroprotective response in astrocytes. While IFN-gamma protects the ability of oxidatively stressed murine astrocytes to clear extracellular glutamate in culture, IL-4 has no effect at any concentration that was tested (10-100 ng/mL). The enhanced release of neuroprotective thiols and lactate by astrocytes in response to T cell stimulation is mimicked by both IL-4 and IFN-gamma. When co-administered, IL-4 abrogated the protective effect of low IFN-gamma on the glutamate clearance function of oxidatively stressed astrocytes in a dose-dependent manner. Astrocyte-conditioned media obtained from cells cultured in the presence of IL-4 (10 or 100 ng/mL) or IFN-gamma (10 ng/mL) decreased by approximately 2-fold, neuronal apoptosis induced by oxidative stress in vitro. However, unlike IL-4, IFN-gamma at high concentrations (100 ng/mL) was not neuroprotective. Our studies with IFN-gamma and IL-4 suggest that a balanced Th1 and Th2 cytokine response might be needed for protecting two key astrocytic functions, glutamate clearance and thiol secretion and might be pertinent to neuroprotective approaches that are aimed at inhibition of an initial pro-inflammatory response to injury or its sustained boosting.


Subject(s)
Astrocytes/drug effects , Astrocytes/metabolism , Interferon-gamma/pharmacology , Interleukin-4/pharmacology , Neuroprotective Agents/pharmacology , Animals , Astrocytes/immunology , Brain/cytology , Brain/metabolism , Brain Injuries/metabolism , Brain Injuries/pathology , Cell Hypoxia/drug effects , Cells, Cultured , Dose-Response Relationship, Drug , Drug Interactions , Embryo, Mammalian , Glutamic Acid/metabolism , Glutathione/metabolism , In Situ Nick-End Labeling/methods , Interleukin-4/immunology , Lactic Acid/metabolism , Mice , Neurons/drug effects , Reactive Oxygen Species/metabolism , Th1 Cells/immunology , Th2 Cells/immunology , tert-Butylhydroperoxide/pharmacology
16.
Trends Biochem Sci ; 33(9): 413-9, 2008 Sep.
Article in English | MEDLINE | ID: mdl-18703339

ABSTRACT

Metabolic interdependence between specialized cells in an organ represents a strategy for energy economy by requiring expression of only a subset of pathway genes in a given cell type. In brain, sulfur metabolism exemplifies this principle of metabolic cooperation between glial and neuronal cells and furnishes three key reagents: S-adenosylmethionine, glutathione and taurine. The pathways for glutathione and taurine syntheses depend on metabolic integration between astrocytes and neurons and intersect with the glutamine-glutamate cycle, which underlies glutamatergic synaptic transmission and requires cooperation between these cell types. We propose that underlying waves of glutamate clearance by astrocytes are activation of cystine import and taurine efflux that result, respectively, from a shared transporter and an increase in solute concentration that triggers osmoregulatory responses.


Subject(s)
Sulfur/metabolism , Synaptic Transmission/physiology , Animals , Astrocytes/physiology , Brain/physiology , Cystine/metabolism , Glutamic Acid/physiology , Glutathione/biosynthesis , Humans , Models, Neurological , Neurons/physiology , Taurine/metabolism
17.
J Immunol ; 180(6): 3866-73, 2008 Mar 15.
Article in English | MEDLINE | ID: mdl-18322194

ABSTRACT

A well-controlled T cell response to CNS injury may result in increased neuronal survival. However, the precise mechanism of T cell-induced neuroprotection is unknown. In this study, we report the unexpected finding that during culture of T cells, high levels of glutamate accumulate, which are efficiently cleared if T cells are cocultured with astrocytes. The T cell-derived glutamate elicits in turn, the release of neuroprotective thiols (cysteine, glutathione, and cysteinyl-glycine) and lactate from astrocytes. Media obtained from astrocytes conditioned in the presence of T cells reduce neuronal apoptosis induced by oxidative stress in primary neuronal cultures from 48 +/- 14 to 9 +/- 4% (p < 0.001). Inhibition of glutamate-dependent signaling during astrocyte-T cell cocultivation by a glutamate uptake inhibitor, l-aspartic acid beta-hydroxamate, abolishes this neuroprotective effect. The ability of astrocytes to clear extracellular glutamate is impaired under conditions of oxidative stress. We demonstrate that T cells, via secreted cytokines, restore glutamate clearance capacity of astrocytes under oxidative conditions. Furthermore, under normoxic conditions, glutamate-buffering capacity of astrocytes is increased upon cocultivation with T cells. It is known that, following CNS injury, astrocytes can respond with beneficial or destructive effects on neurons. However, the context and signaling mechanisms for this dual astrocytic response are unknown. Our results implicate T cells as potential determinants of the context that elicits a protective role for astrocytes in the damaged CNS.


Subject(s)
Astrocytes/immunology , Astrocytes/metabolism , Glutamic Acid/physiology , Immunophenotyping , Neuroprotective Agents/metabolism , T-Lymphocyte Subsets/immunology , T-Lymphocyte Subsets/metabolism , Anaerobiosis/immunology , Animals , Cell Communication/immunology , Cells, Cultured , Coculture Techniques , Extracellular Space/immunology , Extracellular Space/metabolism , Glutamic Acid/metabolism , Glutamic Acid/toxicity , Immunity, Cellular , Lactic Acid/metabolism , Mice , Neurons/immunology , Neurons/metabolism , Oxidative Stress/immunology , Sulfhydryl Compounds/metabolism
18.
J Neuroimmune Pharmacol ; 3(2): 59-74, 2008 Jun.
Article in English | MEDLINE | ID: mdl-18202920

ABSTRACT

Microglial neuroinflammatory responses affect the onset and progression of Parkinson's disease (PD). We posit that such neuroinflammatory responses are, in part, mediated by microglial interactions with nitrated and aggregated alpha-synuclein (alpha-syn) released from Lewy bodies as a consequence of dopaminergic neuronal degeneration. As disease progresses, secretions from alpha-syn-activated microglia can engage neighboring glial cells in a cycle of autocrine and paracrine amplification of neurotoxic immune products. Such pathogenic processes affect the balance between a microglial neurotrophic and neurotoxic signature. We now report that microglia secrete both neurotoxic and neuroprotective factors after exposure to nitrated alpha-syn (N-alpha-syn). Proteomic (surface enhanced laser desorption-time of flight, 1D sodium dodecyl sulfate electrophoresis, and liquid chromatography-tandem mass spectrometry) and limited metabolomic profiling demonstrated that N-alpha-syn-activated microglia secrete inflammatory, regulatory, redox-active, enzymatic, and cytoskeletal proteins. Increased extracellular glutamate and cysteine and diminished intracellular glutathione and secreted exosomal proteins were also demonstrated. Increased redox-active proteins suggest regulatory microglial responses to N-alpha-syn. These were linked to discontinuous cystatin expression, cathepsin activity, and nuclear factor-kappa B activation. Inhibition of cathepsin B attenuated, in part, N-alpha-syn microglial neurotoxicity. These data support multifaceted microglia functions in PD-associated neurodegeneration.


Subject(s)
Microglia/drug effects , Nerve Tissue Proteins/metabolism , Nitrates/pharmacology , alpha-Synuclein/pharmacology , Animals , Cathepsin B/antagonists & inhibitors , Cathepsin B/physiology , Cells, Cultured/drug effects , Cystatins/biosynthesis , Cystatins/genetics , Cysteine/metabolism , Cytoskeletal Proteins/biosynthesis , Cytoskeletal Proteins/genetics , Dopamine/physiology , Gene Expression Regulation/drug effects , Glutamic Acid/metabolism , Glutathione/analysis , Mice , Mice, Inbred C57BL , Microglia/metabolism , NF-kappa B/metabolism , Nerve Tissue Proteins/biosynthesis , Nerve Tissue Proteins/genetics , Nitrates/toxicity , Oxidation-Reduction , Oxidative Stress , Parkinson Disease/physiopathology , Protein Array Analysis , Spectrometry, Mass, Matrix-Assisted Laser Desorption-Ionization , Tandem Mass Spectrometry , alpha-Synuclein/toxicity
19.
Clin Immunol ; 121(3): 260-4, 2006 Dec.
Article in English | MEDLINE | ID: mdl-17049310

ABSTRACT

Sphingosine 1-phosphate (S1P) has recently been reported to induce antimycobacterial activity in vitro and in a mouse model of in vivo Mycobacterium tuberculosis infection. However, its role in the course of pulmonary tuberculosis in humans is still not known. This study shows that S1P levels in airway surface fluid of tuberculosis (TB) patients are significantly less than those observed in non-TB control patients. Moreover, the in vitro stimulation of bronchoalveolar lavage cells coming from TB patients with S1P significantly reduces intracellular growth of endogenous mycobacterial isolates. These results show that, in the course of pulmonary TB, airway epithelial fluid-associated S1P may play a protective role in the containment of intracellular mycobacterial growth and that its decrease may represent a novel pathogenic mechanism through which M. tuberculosis favors its replication.


Subject(s)
Lysophospholipids/metabolism , Sphingosine/analogs & derivatives , Tuberculosis, Pulmonary/metabolism , Tuberculosis, Pulmonary/microbiology , Adult , Animals , Cells, Cultured , Cricetinae , Female , Humans , Lysophospholipids/pharmacology , Male , Middle Aged , Mycobacterium tuberculosis/drug effects , Sphingosine/metabolism , Sphingosine/pharmacology , Tuberculosis, Pulmonary/pathology
20.
Clin Diagn Lab Immunol ; 12(3): 465-73, 2005 Mar.
Article in English | MEDLINE | ID: mdl-15753260

ABSTRACT

A simple and cost-effective diagnostic tool (TB Screen Test) for the screening of patients with pulmonary and extrapulmonary tuberculosis and for differentiation of those individuals from individuals without tuberculosis, other common infections, and healthy controls has been developed. The serological responses of purified mycobacterial glycolipid antigens were examined by a liposome agglutination assay. The assay was able to detect very low antiglycolipid antibody concentrations in the infected individuals. The sera from the tuberculosis patient group had significantly higher concentrations of antiglycolipid antibody than the sera from uninfected control subjects, with 94% sensitivity and 98.3% specificity. Glycolipids of Mycobacterium tuberculosis H37Rv antigens were isolated, purified, and characterized. After interchelation with liposome particles, these purified antigens specifically bound to the antiglycolipid antibodies present in the sera of patients with tuberculosis, resulting in the formation of a blue agglutination. This protocol clearly differentiates healthy controls and M. bovis BCG-vaccinated subjects from those with active tuberculosis. The resultant diagnostic tool, the TB Screen Test, is more economical and rapid (4 min) than other currently available products and can be used for the mass screening of a heavily afflicted population.


Subject(s)
Glycolipids/analysis , Mycobacterium tuberculosis/isolation & purification , Tuberculosis/diagnosis , Agglutination Tests/methods , Agglutination Tests/standards , Antibodies, Bacterial/blood , Biomarkers/blood , Case-Control Studies , Glycolipids/immunology , Humans , Mass Screening/methods , Mycobacterium tuberculosis/chemistry , Sensitivity and Specificity
SELECTION OF CITATIONS
SEARCH DETAIL
...