Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 7 de 7
Filter
Add more filters










Database
Language
Publication year range
1.
Oncotarget ; 9(54): 30225-30239, 2018 Jul 13.
Article in English | MEDLINE | ID: mdl-30100985

ABSTRACT

With the advent of high-throughput sequencing (HTS), profiling immunoglobulin (IG) repertoires has become an essential part of immunological research. Advances in sequencing technology enable the IonTorrent Personal Genome Machine (PGM) to cover the full-length of IG mRNA transcripts. Nucleotide insertions and deletions (indels) are the dominant errors of the PGM sequencing platform and can critically influence IG repertoire assessments. Here, we present a PGM-tailored IG repertoire sequencing approach combining error correction through unique molecular identifier (UID) barcoding and indel detection through ImMunoGeneTics (IMGT), the most commonly used sequence alignment database for IG sequences. Using artificially falsified sequences for benchmarking, we found that IMGT's underlying algorithms efficiently detect 98% of the introduced indels. Undetected indels are either located at the end of the sequences or produce masked frameshifts with an insertion and deletion in close proximity. The complementary determining regions 3 (CDR3s) are returned correct for up to 3 insertions or 3 deletions through conservative culling. We further show, that our PGM-tailored unique molecular identifiers result in highly accurate HTS data if combined with the presented processing strategy. In this regard, considering sequences with at least two copies from datasets with UID families of minimum 3 reads result in correct sequences with over 99% confidence. Finally, we show that the protocol can readily be used to generate homogenous datasets for bulk sequencing of murine bone marrow samples. Taken together, this approach will help to establish benchtop-scale sequencing of IG heavy chain transcripts in the field of IG repertoire research.

2.
J Trace Elem Med Biol ; 44: 76-82, 2017 Dec.
Article in English | MEDLINE | ID: mdl-28965604

ABSTRACT

One consequence of lipopolysaccharide (LPS)-induced stimulation of macrophages is the release of Interferon (IFN)-ß, and subsequently the activation of the JAK-STAT1 pathway, resulting in the expression of inducible nitric oxide synthase (iNOS). Free intracellular zinc ions (Zn2+) have a profound impact as a second messenger in LPS-dependent gene expression. Previous work had indicated a Zn2+-dependent upregulation of STAT1 mRNA in response to LPS and IFN-ß, potentially affecting STAT1-dependent downstream signaling upon pre-incubation with these agents. The aim of the present study was to investigate the long-term influence of Zn2+ chelation on cellular STAT1 levels and their effect on protein levels and activity of iNOS. The LPS- and IFN-ß-mediated increase of STAT1 mRNA and protein levels was abrogated by chelation of Zn2+ with the membrane permeable chelator N,N,N',N'-Tetrakis(2-pyridylmethyl)ethylenediamine (TPEN) in RAW 264.7 macrophages. After 48h pre-incubation together with IFN-ß, TPEN also led to reduced nitric monoxide formation in response to a second stimulation with LPS. Nonetheless, the latter was observed regardless of any pre-incubation with IFN-ß, suggesting that the effect of treatment with TPEN negatively affects iNOS induction independently from cellular STAT1 levels. In conclusion, long term Zn2+ chelation does affect STAT1 protein expression, but interferes with NO production by a different, yet unknown pathway not involving STAT1. However, as there are many additional STAT1-dependent genes, there might still be effects on targets other than iNOS.


Subject(s)
Chelating Agents/pharmacology , Interferon-beta/metabolism , Nitric Oxide Synthase Type II/metabolism , STAT1 Transcription Factor/metabolism , Up-Regulation/drug effects , Zinc/pharmacology , 3T3 Cells , Animals , Lipopolysaccharides/pharmacology , Mice , Models, Biological , Nitric Oxide/metabolism , RAW 264.7 Cells , RNA, Messenger/genetics , RNA, Messenger/metabolism , STAT1 Transcription Factor/genetics
3.
J Trace Elem Med Biol ; 37: 117-124, 2016 Sep.
Article in English | MEDLINE | ID: mdl-27134082

ABSTRACT

Lead is a widespread environmental pollutant and the highly poisonous metal compromises multiple organs in the body. Among other tissues and cells, lead ions (Pb(2+)) can affect macrophages and microglia cells. The present study observed a concentration-dependent protection of BV-2 microglia and RAW 264.7 macrophages by Pb(2+) against lipopolysaccharide (LPS)-induced toxicity. Both cell lines are potent producers of two substances that have previously been shown to mediate cytotoxic effects of LPS. These are the pro-inflammatory cytokine tumor necrosis factor (TNF)-α and nitric monoxide (NO), which creates nitrosative stress, hampering the distribution of invading pathogens and tumor cells. While the expression of TNF-α was unaffected by Pb(2+), the production of NO was significantly inhibited. Moreover, blocking NO synthesis by low molecular weight inhibitors prevented LPS-mediated toxicity, confirming the role of NO in these events. Pb(2+) exposure led to a downregulation of LPS-induced expression of the transcription factor STAT1, which is involved in iNOS transcription. Moreover, iNOS mRNA and protein levels were reduced in the presence of Pb(2+), explaining the reduced formation of NO and a subsequent increase of cellular viability in vitro. In vivo, the effect might limit collateral damage caused by excessive NO production, but also impair the efficiency of NO as a central mediator of the defense against various pathogens.


Subject(s)
Lead/pharmacology , Lipopolysaccharides/antagonists & inhibitors , Nitric Oxide Synthase Type II/biosynthesis , Nitric Oxide/toxicity , STAT1 Transcription Factor/biosynthesis , Animals , Cell Survival/drug effects , Cells, Cultured , Dose-Response Relationship, Drug , Ions/pharmacology , Lipopolysaccharides/pharmacology , Macrophages/drug effects , Macrophages/metabolism , Mice , Nitric Oxide/biosynthesis , Nitric Oxide Synthase Type II/genetics , Nitric Oxide Synthase Type II/metabolism , STAT1 Transcription Factor/genetics , STAT1 Transcription Factor/metabolism , Structure-Activity Relationship , Tumor Necrosis Factor-alpha/antagonists & inhibitors , Tumor Necrosis Factor-alpha/genetics , Tumor Necrosis Factor-alpha/metabolism
4.
J Nutr Biochem ; 29: 116-23, 2016 Mar.
Article in English | MEDLINE | ID: mdl-26895672

ABSTRACT

The essential trace element zinc is indispensable for proper immune function as zinc deficiency accompanies immune defects and dysregulations like allergies, autoimmunity and an increased presence of transplant rejection. This point to the importance of the physiological and dietary control of zinc levels for a functioning immune system. This study investigates the capacity of zinc to induce immune tolerance. The beneficial impact of physiological zinc supplementation of 6 µg/day (0.3mg/kg body weight) or 30 µg/day (1.5mg/kg body weight) on murine experimental autoimmune encephalomyelitis (EAE), an animal model for multiple sclerosis with a Th1/Th17 (Th, T helper) cell-dominated immunopathogenesis, was analyzed. Zinc administration diminished EAE scores in C57BL/6 mice in vivo (P<.05), reduced Th17 RORγT(+) cells (P<.05) and significantly increased inducible iTreg cells (P<.05). While Th17 cells decreased systemically, iTreg cells accumulated in the central nervous system. Cumulatively, zinc supplementation seems to be capable to induce tolerance in unwanted immune reactions by increasing iTreg cells. This makes zinc a promising future tool for treating autoimmune diseases without suppressing the immune system.


Subject(s)
Encephalomyelitis, Autoimmune, Experimental/immunology , T-Lymphocytes, Regulatory/immunology , Th1 Cells/immunology , Th17 Cells/immunology , Zinc/administration & dosage , Animals , Female , Mice , Mice, Inbred C57BL
5.
Metallomics ; 6(7): 1277-87, 2014 Jul.
Article in English | MEDLINE | ID: mdl-24759986

ABSTRACT

Free zinc ions (Zn(2+)) participate in several signaling pathways. The aim of the present study was to investigate a potential involvement of Zn(2+) in the PI3K/Akt pathway of interleukin (IL)-2 signaling in T-cells. The IL-2 receptor triggers three major pathways, ERK1/2, JAK/STAT5, and PI3K/Akt. We have previously shown that an IL-2-mediated release of lysosomal Zn(2+) into the cytoplasm activates ERK1/2, but not STAT5. In the present study, Akt phosphorylation in response to IL-2 was abrogated by the Zn(2+) chelator N,N,N',N'-tetrakis-2(pyridyl-methyl)ethylenediamine, and was induced by treatment with Zn(2+) and the ionophore pyrithione. The latter were ineffective in cells that were treated with siRNA against the phosphatase and tensin homolog deleted on chromosome 10 (PTEN), a phosphatase that degrades the lipid second messenger PI(3,4,5)P3, which is produced by PI3K and leads to activation of Akt. Inhibition of recombinant PTEN by Zn(2+)in vitro yielded an IC50 of 0.59 nM. Considering a resting free cytoplasmic Zn(2+) level of 0.2 nM in the T-cell line CTLL-2, this seems ideally suited for dynamic regulation by cellular Zn(2+). Oxidation with H2O2 and supplementation with Zn(2+) led to similar changes in the CD spectrum of PTEN. Moreover, Zn(2+) partially prevented the oxidation of cysteines 71 and 124. Hence, we hypothesize that zinc signals affect the IL-2-dependent PI3K/Akt pathway by inhibiting the negative regulator PTEN through binding with a sub-nanomolar affinity to cysteine residues that are essential for its catalytic activity.


Subject(s)
Extracellular Signal-Regulated MAP Kinases/metabolism , Interleukin-2/metabolism , PTEN Phosphohydrolase/antagonists & inhibitors , Phosphatidylinositol 3-Kinases/metabolism , Proto-Oncogene Proteins c-akt/metabolism , Zinc/pharmacology , Animals , Humans , Interleukin-2/pharmacology , Jurkat Cells , Male , Phosphorylation , Signal Transduction/drug effects , T-Lymphocytes/drug effects , T-Lymphocytes/metabolism
6.
J Immunol ; 191(4): 1808-17, 2013 Aug 15.
Article in English | MEDLINE | ID: mdl-23863901

ABSTRACT

Zinc signals are utilized by several immune cell receptors. One is TLR4, which causes an increase of free zinc ions (Zn(2+)) that is required for the MyD88-dependent expression of inflammatory cytokines. This study investigates the role of Zn(2+) on Toll/IL-1R domain-containing adapter inducing IFN-ß (TRIF)-dependent signals, the other major intracellular pathway activated by TLR4. Chelation of Zn(2+) with the membrane-permeable chelator N,N,N',N'-Tetrakis(2-pyridylmethyl)ethylenediamine augmented TLR4-mediated production of IFN-ß and subsequent synthesis of inducible NO synthase and production of NO. The effect is based on Zn(2+) acting as a negative regulator of the TRIF pathway via reducing IFN regulatory factor 3 activation. This was also observed with TLR3, the only TLR that signals exclusively via TRIF, but not MyD88, and does not trigger a zinc signal. In contrast, IFN-γ-induced NO production was unaffected by N,N,N',N'-Tetrakis(2-pyridylmethyl)ethylenediamine. Taken together, Zn(2+) is specifically involved in TLR signaling, where it differentially regulates MyD88 and TRIF signaling via a zinc signal or via basal Zn(2+) levels, respectively.


Subject(s)
Adaptor Proteins, Vesicular Transport/physiology , Myeloid Differentiation Factor 88/physiology , Signal Transduction/drug effects , Toll-Like Receptor 4/physiology , Zinc/pharmacology , Animals , Antibodies, Monoclonal/immunology , Antibodies, Monoclonal/pharmacology , Cell Line , Chelating Agents/pharmacology , Cytokines/biosynthesis , Cytokines/genetics , Enzyme Induction/drug effects , Ethylenediamines/pharmacology , Gene Expression Regulation/drug effects , Interferon Regulatory Factor-3/metabolism , Interferon-beta/biosynthesis , Interferon-beta/genetics , Interferon-beta/immunology , Lipopolysaccharides/pharmacology , Macrophages/immunology , Mice , Mice, Inbred C57BL , NF-kappa B/metabolism , Nitric Oxide Synthase Type II/biosynthesis , Nitric Oxide Synthase Type II/genetics , Phosphorylation/drug effects , Polystyrenes/pharmacology , Polyvinyls/pharmacology , Protein Processing, Post-Translational/drug effects , Rats , Toll-Like Receptors/physiology
7.
Toxicol In Vitro ; 25(4): 960-8, 2011 Jun.
Article in English | MEDLINE | ID: mdl-21397682

ABSTRACT

Perfluorinated compounds (PFCs), such as perfluorooctanesulfonate (PFOS) and perfluorooctanoic acid (PFOA), are xenobiotics that can be detected worldwide in the environment, wildlife, and humans. So far, the immunotoxicity of PFCs has only been investigated in rodents, but not in humans. In this study, we explore the impact of PFOS and PFOA on selected functions of human leukocytes in vitro. PFOS induced a significant decrease of natural killer-cell activity and reduced the release of the pro-inflammatory cytokine TNF-α following lipopolysaccharide (LPS)-stimulation. Furthermore, the plasma PFOS concentrations (2.09-8.98 ng/ml) found in our study subjects correlated positively with the LPS-stimulated IL-6 release. PFOA augmented significantly calcitriol-induced monocytic differentiation of the HL-60 cell line. Additionally, there was a significant linear relationship between LPS-stimulated TNF-α and IL-6 release, and the plasma PFOA (1.20-6.92ng/ml) concentrations of the study subjects. In conclusion, the investigated PFCs affect human immune cells mainly with regard to natural killer-cell cytotoxicity and the pro-inflammatory cytokine release by stimulated macrophages.


Subject(s)
Alkanesulfonic Acids/toxicity , Caprylates/toxicity , Fluorocarbons/toxicity , Leukocytes, Mononuclear/drug effects , Adult , Alkanesulfonic Acids/pharmacokinetics , Caprylates/pharmacokinetics , Female , Fluorocarbons/pharmacokinetics , HL-60 Cells , Humans , In Vitro Techniques , Killer Cells, Natural/drug effects , Killer Cells, Natural/metabolism , Leukocytes, Mononuclear/metabolism , Lipopolysaccharides/pharmacology , Macrophages/drug effects , Macrophages/metabolism , Male , Tumor Necrosis Factor-alpha/drug effects , Tumor Necrosis Factor-alpha/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...