Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 6 de 6
Filter
1.
Environ Sci Nano ; 5(2): 572-588, 2018 Feb 01.
Article in English | MEDLINE | ID: mdl-29479436

ABSTRACT

ZnO nanoparticles (nZnO) are commonly used in nanotechnology applications despite their demonstrated cytotoxicity against multiple cell types. This underscores the significant need to determine the physicochemical properties that influence nZnO cytotoxicity. In this study, we analyzed six similarly sized nZnO formulations, along with SiO2-coated nZnO, bulk ZnO and ZnSO4 as controls. Four of the nZnO samples were synthesized using various wet chemical methods, while three employed high-temperature flame spray pyrolysis (FSP) techniques. X-ray diffraction and optical analysis demonstrated the lattice parameters and electron band gap of the seven nZnO formulations were similar. However, electrophoretic mobility measures, hydrodynamic size, photocatalytic rate constants, dissolution potential, reactive oxygen species (ROS) production and, more importantly, the cytotoxicity of the variously synthesized nZnO towards Jurkat leukemic and primary CD4+ T cells displayed major differences. Surface structure analysis using FTIR, X-ray photoelectron spectroscopies (XPS) and dynamic light scattering (DLS) revealed significant differences in the surface-bound chemical groups and the agglomeration tendencies of the samples. The wet chemical nZnO, with higher cationic surface charge, faster photocatalytic rates, increased extracellular dissolution and ROS generation demonstrated greater cytotoxicity towards both cell types than those made with FSP techniques. Furthermore, principal component analysis (PCA) suggests that the synthesis procedure employed influences which physicochemical properties contribute more to the cytotoxic response. These results suggest that the synthesis approach results in unique surface chemistries and can be a determinant of cellular cytotoxicity and oxidative stress responses.

2.
Chem Res Toxicol ; 30(8): 1641-1651, 2017 08 21.
Article in English | MEDLINE | ID: mdl-28693316

ABSTRACT

Zinc oxide nanoparticles (nZnO) are one of the most highly produced nanomaterials and are used in numerous applications including cosmetics and sunscreens despite reports demonstrating their cytotoxicity. Dissolution is viewed as one of the main sources of nanoparticle (NP) toxicity; however, dissolution studies can be time-intensive to perform and complicated by issues such as particle separation from solution. Our work attempts to overcome some of these challenges by utilizing new methods using UV/vis and fluorescence spectroscopy to quantitatively assess nZnO dissolution in various biologically relevant solutions. All biological buffers tested induce rapid dissolution of nZnO. These buffers, including HEPES, MOPS, and PIPES, are commonly used in cell culture media, cellular imaging solutions, and to maintain physiological pH. Additional studies using X-ray diffraction, FT-IR, X-ray photoelectron spectroscopy, ICP-MS, and TEM were performed to understand how the inclusion of these nonessential media components impacts the behavior of nZnO in RPMI media. From these assessments, we demonstrate that HEPES causes increased dissolution kinetics, boosts the conversion of nZnO into zinc phosphate/carbonate, and, interestingly, alters the structural morphology of the complex precipitates formed with nZnO in cell culture conditions. Cell viability experiments demonstrated that the inclusion of these buffers significantly decrease the viability of Jurkat leukemic cells when challenged with nZnO. This work demonstrates that biologically relevant buffering systems dramatically impact the dynamics of nZnO including dissolution kinetics, morphology, complex precipitate formation, and toxicity profiles.


Subject(s)
Culture Media/chemistry , Metal Nanoparticles/chemistry , Zinc Oxide/chemistry , Buffers , Cell Survival/drug effects , Humans , Hydrogen-Ion Concentration , Jurkat Cells , Kinetics , Mass Spectrometry , Metal Nanoparticles/toxicity , Microscopy, Confocal , Microscopy, Electron, Transmission , Particle Size , Photoelectron Spectroscopy , Spectrometry, Fluorescence , Spectroscopy, Fourier Transform Infrared , X-Ray Diffraction
3.
J Immunol Methods ; 376(1-2): 20-31, 2012 Feb 28.
Article in English | MEDLINE | ID: mdl-22079255

ABSTRACT

Vasoactive intestinal peptide receptor-1 signaling in lymphocytes has been shown to regulate chemotaxis, proliferation, apoptosis and differentiation. During T cell activation, VPAC1 mRNA is downregulated, but the effect on its protein levels is less clear. A small number of studies have reported measurement of human VPAC1 by flow cytometry, but murine VPAC1 reagents are unavailable. Therefore, we set out to generate a reliable and highly specific α-mouse VPAC1 polyclonal antibody for use with flow cytometry. After successfully generating a rabbit α-VPAC1 polyclonal antibody (α-mVPAC1 pAb), we characterized its cross-reactivity and showed that it does not recognize other family receptors (mouse VPAC2 and PAC1, and human VPAC1, VPAC2 and PAC1) by flow cytometry. Partial purification of the rabbit α-VPAC1 sera increased the specific-activity of the α-mVPAC1 pAb by 20-fold, and immunofluorescence microscopy (IF) confirmed a plasma membrane subcellular localization for mouse VPAC1 protein. To test the usefulness of this specific α-mVPAC1 pAb, we showed that primary, resting mouse T cells express detectable levels of VPAC1 protein, with little detectable signal from activated T cells, or CD19 B cells. These data support our previously published data showing a downregulation of VPAC1 mRNA during T cell activation. Collectively, we have established a well-characterized, and highly species specific α-mVPAC1 pAb for VPAC1 surface measurement by IF and flow cytometry.


Subject(s)
Antibodies/immunology , Receptors, Vasoactive Intestinal Polypeptide, Type I/immunology , Animals , Antibodies/genetics , CHO Cells , Cricetinae , Flow Cytometry/methods , Mice , Microscopy, Fluorescence , RNA/chemistry , RNA/genetics , Rabbits , Receptors, Vasoactive Intestinal Polypeptide, Type I/genetics , Reverse Transcriptase Polymerase Chain Reaction , Transfection/methods
4.
Peptides ; 32(10): 2058-66, 2011 Oct.
Article in English | MEDLINE | ID: mdl-21878358

ABSTRACT

Successful thymocyte maturation is essential for normal, peripheral T cell function. Vasoactive intestinal peptide (VIP) is a neuropeptide which is highly expressed in the thymus that has been shown to modulate thymocyte development. VIP predominantly binds two G protein coupled receptors, termed vasoactive intestinal peptide receptor 1 (VPAC1) and VPAC2, but their expression profiles in CD4(-)/CD8(-) (double negative, DN) thymocyte subsets, termed DN1-4, have yet to be identified. We hypothesized that a high VPAC1:VPAC2 ratio in the earliest thymocyte progenitors (ETP cells) would be reversed during early lymphopoiesis as observed in activated, peripheral Th(2) cells, as the thymus is rich in Th(2) cytokines. In support of this hypothesis, high VPAC1 mRNA levels decreased 1000-fold, accompanied with a simultaneous increase in VPAC2 mRNA expression during early thymocyte progenitor (ETP/DN1)→DN3 differentiation. Moreover, arrested DN3 cells derived from an Ikaros null mouse (JE-131 cells) failed to completely reverse the VIP receptor ratio compared to wild type DN3 thymocytes. Surprisingly, VPAC2(-/-) mice did not show significant changes in relative thymocyte subset numbers. These data support the notion that both VPAC1 and VPAC2 receptors are dynamically regulated by Ikaros, a master transcriptional regulator for thymocyte differentiation, during early thymic development. Moreover, high VPAC1 mRNA is a novel marker for the ETP population making it enticing to speculate that the chemotactic VIP/VPAC1 signaling axis may play a role in thymocyte movement. Also, despite the results that VPAC2 deficiency did not affect thymic subset numbers, future studies are necessary to determine whether downstream T cell phenotypic changes manifest themselves, such as a propensity for a Th(1) versus Th(2) polarization.


Subject(s)
Lymphopoiesis/physiology , Receptors, Vasoactive Intestinal Peptide, Type II/metabolism , Receptors, Vasoactive Intestinal Polypeptide, Type I/metabolism , Animals , Ikaros Transcription Factor/genetics , Ikaros Transcription Factor/metabolism , Lymphocyte Subsets/metabolism , Mice , Mice, Inbred C57BL , Mice, Knockout , Receptors, Vasoactive Intestinal Peptide, Type II/genetics , Receptors, Vasoactive Intestinal Polypeptide, Type I/genetics , Thymocytes/cytology , Thymocytes/metabolism
5.
Mol Immunol ; 47(6): 1181-94, 2010 Mar.
Article in English | MEDLINE | ID: mdl-20117839

ABSTRACT

More than 40 years after the discovery of vasoactive intestinal peptide (VIP), its transcriptome in the immune system has still not been completely elucidated. In an attempt to understand the biological role of this neuropeptide in immunity, we chose CD4 T cells as a cellular system. Agilent Mouse Whole Genome microarrays were hybridized with fluorescently labeled total RNA isolated from resting CD4 T cells cultured +/-10(-7)M VIP for 5h or PMA/ionomycin activated CD4 T cells cultured +/-10(-7)M VIP for 5h. These VIP-regulated transcriptomes were analyzed by Significance Analysis of Microarrays (SAM) and Ingenuity Pathway Analysis (IPA) software to identify relevant signaling pathways modulated by VIP in the absence and presence of T cell activation. In resting CD4 T cells, VIP-modulated 368 genes, ranging from 3.49 to -4.78-fold. In the PMA/ionomycin activated CD4 T cells, 326 gene expression levels were changed by VIP, ranging from 2.94 to -1.66-fold. IPA analysis revealed that VIP exposure alters cellular function through EGFR signaling in resting CD4 T cells, and modulates immediate early genes, Fos and CREM/ICER, in activated CD4 T cells. These gene expression changes are suggested to explain at a molecular level how VIP can regulate T cell homing to the gut and induce regulatory T cell generation.


Subject(s)
CD4-Positive T-Lymphocytes/metabolism , Gene Expression Profiling , Gene Expression Regulation/drug effects , Lymphocyte Activation/drug effects , Lymphocyte Activation/genetics , Signal Transduction/drug effects , Vasoactive Intestinal Peptide/pharmacology , Animals , CD4-Positive T-Lymphocytes/drug effects , Female , Gene Regulatory Networks , Male , Mice , Mice, Inbred C57BL , Oligonucleotide Array Sequence Analysis , Phenotype , Receptors, Vasoactive Intestinal Peptide/genetics , Receptors, Vasoactive Intestinal Peptide/metabolism , Reproducibility of Results , Reverse Transcriptase Polymerase Chain Reaction , Tetradecanoylphorbol Acetate/pharmacology
6.
Brain Behav Immun ; 22(7): 1032-1040, 2008 Oct.
Article in English | MEDLINE | ID: mdl-18534815

ABSTRACT

Strict regulation of T cell function is imperative to control adaptive immunity, and dysregulation of T cell activation can contribute to infectious and autoimmune diseases. Vasoactive intestinal peptide receptor-1 (VPAC-1), an anti-inflammatory G-protein coupled receptor, has been reported to be downregulated during T cell activation. However, the regulatory mechanisms controlling the expression of VPAC-1 in T cells are not well understood. Therefore, mouse splenic CD4 T cells were treated in complete media+/-anti-CD3 for 24h, total RNA isolated and VPAC-1 levels measured by qPCR. Surprisingly, we discovered that T cells incubated in complete media steadily upregulated VPAC-1 mRNA levels over time (24h). Importantly, CD4 T cells isolated from blood also showed elevated VPAC-1 expression compared to splenic T cells. Collectively, these data support that the vascular environment positively influences VPAC-1 mRNA expression that is negatively regulated by TCR signaling. This research was supported by a national service award (1KO1 DK064828) to G.D., the Center for Protease Research (2P20RR015566), and INBRE (P20 RR016741).


Subject(s)
CD4-Positive T-Lymphocytes/metabolism , Receptor-CD3 Complex, Antigen, T-Cell/physiology , Receptors, Vasoactive Intestinal Polypeptide, Type I/genetics , Signal Transduction/physiology , Animals , Antibodies/pharmacology , CD4-Positive T-Lymphocytes/cytology , CD4-Positive T-Lymphocytes/drug effects , Cell Culture Techniques , Culture Media/pharmacology , Electrophoresis, Polyacrylamide Gel , Enzyme-Linked Immunosorbent Assay , Female , Gene Expression Regulation/drug effects , Lymphocyte Activation/drug effects , Male , Mice , Mice, Inbred C57BL , RNA, Messenger/genetics , RNA, Messenger/metabolism , Receptor-CD3 Complex, Antigen, T-Cell/immunology , Receptors, Vasoactive Intestinal Polypeptide, Type I/analysis , Receptors, Vasoactive Intestinal Polypeptide, Type I/metabolism , Reverse Transcriptase Polymerase Chain Reaction , Signal Transduction/drug effects
SELECTION OF CITATIONS
SEARCH DETAIL