Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 17 de 17
Filter
Add more filters










Publication year range
1.
bioRxiv ; 2024 Jul 08.
Article in English | MEDLINE | ID: mdl-39026716

ABSTRACT

Cetylpyridinium chloride (CPC) is a quaternary ammonium antimicrobial used in numerous personal care products, human food, cosmetic products, and cleaning solutions. Yet, there is minimal published data on CPC effects on eukaryotes, immune signaling, and human health. Previously, we showed that low-micromolar CPC inhibits rat mast cell function by inhibiting antigen (Ag)-stimulated Ca 2+ mobilization, microtubule polymerization, and degranulation. In this study, we extend the findings to human mast cells (LAD2) and present data indicating that CPC's mechanism of action centers on its positively-charged quaternary nitrogen in its pyridinium headgroup. CPC's inhibitory effect is independent of signaling platform receptor architecture. Tyrosine phosphorylation events are a trigger of Ca 2+ mobilization necessary for degranulation. CPC inhibits global tyrosine phosphorylation in Ag-stimulated mast cells. Specifically, CPC inhibits tyrosine phosphorylation of specific key players Syk kinase and LAT, a substrate of Syk. In contrast, CPC does not affect Lyn kinase phosphorylation. Thus, CPC's root mechanism is electrostatic disruption of particular tyrosine phosphorylation events essential for signaling. This work outlines the biochemical mechanisms underlying the effects of CPC on immune signaling and allows the prediction of CPC effects on cell types, like T cells, that share similar signaling elements.

2.
Food Chem Toxicol ; 186: 114547, 2024 Apr.
Article in English | MEDLINE | ID: mdl-38408634

ABSTRACT

People are exposed to high concentrations of antibacterial agent cetylpyridinium chloride (CPC) via food and personal care products, despite little published information regarding CPC effects on eukaryotes. Here, we show that low-micromolar CPC exposure, which does not cause cell death, inhibits mitochondrial ATP production in primary human keratinocytes, mouse NIH-3T3 fibroblasts, and rat RBL-2H3 immune mast cells. ATP inhibition via CPC (EC50 1.7 µM) is nearly as potent as that caused by canonical mitotoxicant CCCP (EC50 1.2 µM). CPC inhibition of oxygen consumption rate (OCR) tracks with that of ATP: OCR is halved due to 1.75 µM CPC in RBL-2H3 cells and 1.25 µM in primary human keratinocytes. Mitochondrial [Ca2+] changes can cause mitochondrial dysfunction. Here we show that CPC causes mitochondrial Ca2+ efflux from mast cells via an ATP-inhibition mechanism. Using super-resolution microscopy (fluorescence photoactivation localization) in live cells, we have discovered that CPC causes mitochondrial nanostructural defects in live cells within 60 min, including the formation of spherical structures with donut-like cross section. This work reveals CPC as a mitotoxicant despite widespread use, highlighting the importance of further research into its toxicological safety.


Subject(s)
Anti-Infective Agents, Local , Anti-Infective Agents , Mice , Humans , Rats , Animals , Cetylpyridinium/chemistry , Cetylpyridinium/pharmacology , Rodentia , Anti-Infective Agents/pharmacology , Mitochondria , Adenosine Triphosphate
3.
Food Chem Toxicol ; 179: 113980, 2023 Sep.
Article in English | MEDLINE | ID: mdl-37549805

ABSTRACT

Cetylpyridinium chloride (CPC) is an antimicrobial used in numerous personal care and janitorial products and food for human consumption at millimolar concentrations. Minimal information exists on the eukaryotic toxicology of CPC. We have investigated the effects of CPC on signal transduction of the immune cell type mast cells. Here, we show that CPC inhibits the mast cell function degranulation with antigen dose-dependence and at non-cytotoxic doses ∼1000-fold lower than concentrations in consumer products. Previously we showed that CPC disrupts phosphatidylinositol 4,5-bisphosphate, a signaling lipid critical for store-operated Ca2+ entry (SOCE), which mediates degranulation. Our results indicate that CPC inhibits antigen-stimulated SOCE: CPC restricts Ca2+ efflux from endoplasmic reticulum, reduces Ca2+ uptake into mitochondria, and dampens Ca2+ flow through plasma membrane channels. While inhibition of Ca2+ channel function can be caused by alteration of plasma membrane potential (PMP) and cytosolic pH, CPC does not affect PMP or pH. Inhibition of SOCE is known to depress microtubule polymerization, and here we show that CPC indeed dose-dependently shuts down formation of microtubule tracks. In vitro data reveal that CPC inhibition of microtubules is not due to direct CPC interference with tubulin. In summary, CPC is a signaling toxicant that targets Ca2+ mobilization.


Subject(s)
Cetylpyridinium , Mast Cells , Humans , Cetylpyridinium/metabolism , Cetylpyridinium/pharmacology , Calcium/metabolism , Signal Transduction , Pharmaceutical Preparations/metabolism , Calcium Signaling
4.
bioRxiv ; 2023 May 24.
Article in English | MEDLINE | ID: mdl-37292883

ABSTRACT

Cetylpyridinium chloride (CPC) is an antimicrobial used in numerous personal care and janitorial products and food for human consumption at millimolar concentrations. Minimal information exists on the eukaryotic toxicology of CPC. We have investigated the effects of CPC on signal transduction of the immune cell type mast cells. Here, we show that CPC inhibits the mast cell function degranulation with antigen dose-dependence and at non-cytotoxic doses ∼1000-fold lower than concentrations in consumer products. Previously we showed that CPC disrupts phosphatidylinositol 4,5-bisphosphate, a signaling lipid critical for store-operated Ca 2+ entry (SOCE), which mediates degranulation. Our results indicate that CPC inhibits antigen-stimulated SOCE: CPC restricts Ca 2+ efflux from endoplasmic reticulum, reduces Ca 2+ uptake into mitochondria, and dampens Ca 2+ flow through plasma membrane channels. While inhibition of Ca 2+ channel function can be caused by alteration of plasma membrane potential (PMP) and cytosolic pH, CPC does not affect PMP or pH. Inhibition of SOCE is known to depress microtubule polymerization, and here we show that CPC indeed dose-dependently shuts down formation of microtubule tracks. In vitro data reveal that CPC inhibition of microtubules is not due to direct CPC interference with tubulin. In summary, CPC is a signaling toxicant that targets Ca 2+ mobilization.

5.
Toxicol Appl Pharmacol ; 405: 115205, 2020 10 15.
Article in English | MEDLINE | ID: mdl-32835763

ABSTRACT

Triclosan (TCS) is an antimicrobial agent that was effectively banned by the FDA from hand soaps in 2016, hospital soaps in 2017, and hand sanitizers in 2019; however, TCS can still be found in a few products. At consumer-relevant, non-cytotoxic doses, TCS inhibits the functions of both mitochondria and mast cells, a ubiquitous cell type. Via the store-operated Ca2+ entry mechanism utilized by many immune cells, mast cells undergo antigen-stimulated Ca2+ influx into the cytosol, for proper function. Previous work showed that TCS inhibits Ca2+ dynamics in mast cells, and here we show that TCS also inhibits Ca2+ mobilization in human Jurkat T cells. However, the biochemical mechanism behind the Ca2+ dampening has yet to be elucidated. Three-dimensional super-resolution microscopy reveals that TCS induces mitochondrial swelling, in line with and extending the previous finding of TCS inhibition of mitochondrial membrane potential via its proton ionophoric activity. Inhibition of plasma membrane potential (PMP) by the canonical depolarizer gramicidin can inhibit mast cell function. However, use of the genetically encoded voltage indicators (GEVIs) ArcLight (pH-sensitive) and ASAP2 (pH-insensitive), indicates that TCS does not disrupt PMP. In conjunction with data from a plasma membrane-localized, pH-sensitive reporter, these results indicate that TCS, instead, induces cytosolic acidification in mast cells and T cells. Acidification of the cytosol likely inhibits Ca2+ influx by uncoupling the STIM1/ORAI1 interaction that is required for opening of plasma membrane Ca2+ channels. These results provide a mechanistic explanation of TCS disruption of Ca2+ influx and, thus, of immune cell function.


Subject(s)
Anti-Infective Agents/toxicity , Calcium/metabolism , Cytoplasm/drug effects , Mast Cells/drug effects , T-Lymphocytes/drug effects , Triclosan/toxicity , Calcium Channels/metabolism , Cell Degranulation/drug effects , Cell Line , Cell Membrane/drug effects , Cytoplasm/metabolism , Humans , Hydrogen-Ion Concentration , Mast Cells/metabolism , Membrane Potentials/drug effects , Mitochondrial Swelling/drug effects , T-Lymphocytes/metabolism
6.
Dev Psychobiol ; 62(1): 21-35, 2020 01.
Article in English | MEDLINE | ID: mdl-31401811

ABSTRACT

Although prenatal opioid exposure and postnatal withdrawal (neonatal abstinence syndrome) are associated with infant neurobehavioral deficits, little is known about the impact of continued maternal opioid treatment in the postnatal period on maternal responsivity and relationship to mother's oxytocin release during dyadic interactions in the Still Face paradigm. Mother and infant dyads (N = 14) were recruited and comprised of mothers on opioid replacement throughout pregnancy and postpartum (opioid-exposed group, n = 7) and a demographically controlled, non-exposed group (n = 7). Salivary oxytocin was collected following 10 min of infant separation before and immediately after a 6-min Still Face paradigm. Oxytocin measures correlated strongly with sensitive and prosocial maternal behaviors in response to infant initiation. Opioid-exposed compared to non-exposed mothers had significantly lower pre-test to post-test rise in salivary oxytocin concentration level as well as fewer sensitive behaviors during the reunion condition of the Still Face paradigm. Maternal opioid dependence during early infancy may impair maternal responsivity and sensitivity through suppression of the oxytocin reflex to infant stimulation.


Subject(s)
Maternal Behavior/physiology , Mother-Child Relations , Mothers , Object Attachment , Opioid-Related Disorders/metabolism , Oxytocin/metabolism , Pregnancy Complications/metabolism , Adult , Female , Humans , Infant , Longitudinal Studies , Opiate Substitution Treatment , Opioid-Related Disorders/drug therapy , Pregnancy , Pregnancy Complications/drug therapy
7.
J Appl Toxicol ; 39(12): 1672-1690, 2019 12.
Article in English | MEDLINE | ID: mdl-31429102

ABSTRACT

Humans are exposed to the antimicrobial agent triclosan (TCS) through use of TCS-containing products. Exposed tissues contain mast cells, which are involved in numerous biological functions and diseases by secreting various chemical mediators through a process termed degranulation. We previously demonstrated that TCS inhibits both Ca2+ influx into antigen-stimulated mast cells and subsequent degranulation. To determine the mechanism linking the TCS cytosolic Ca2+ depression to inhibited degranulation, we investigated the effects of TCS on crucial signaling enzymes activated downstream of the Ca2+ rise: protein kinase C (PKC; activated by Ca2+ and reactive oxygen species [ROS]) and phospholipase D (PLD). We found that TCS strongly inhibits PLD activity within 15 minutes post-antigen, a key mechanism of TCS mast cell inhibition. In addition, experiments using fluorescent constructs and confocal microscopy indicate that TCS delays antigen-induced translocations of PKCßII, PKCδ and PKC substrate myristoylated alanine-rich C-kinase. Surprisingly, TCS does not inhibit PKC activity or overall ability to translocate, and TCS actually increases PKC activity by 45 minutes post-antigen; these results are explained by the timing of both TCS inhibition of cytosolic Ca2+ (~15+ minutes post-antigen) and TCS stimulation of ROS (~45 minutes post-antigen). These findings demonstrate that it is incorrect to assume that all Ca2+ -dependent processes will be synchronously inhibited when cytosolic Ca2+ is inhibited by a toxicant or drug. The results offer molecular predictions of the effects of TCS on other mammalian cell types, which share these crucial signal transduction elements and provide biochemical information that may underlie recent epidemiological findings implicating TCS in human health problems.


Subject(s)
Anti-Infective Agents/toxicity , Calcium/metabolism , Cell Degranulation/drug effects , Mast Cells/drug effects , Phospholipase D/antagonists & inhibitors , Triclosan/toxicity , Cell Line , Humans , Mast Cells/metabolism , Mast Cells/pathology , Mast Cells/physiology , Myristoylated Alanine-Rich C Kinase Substrate/metabolism , Protein Kinase C/metabolism , Reactive Oxygen Species/metabolism , Signal Transduction
8.
J Appl Toxicol ; 39(3): 473-484, 2019 03.
Article in English | MEDLINE | ID: mdl-30374992

ABSTRACT

Mast cells comprise a physiologically and toxicologically important cell type that is ubiquitous among species and tissues. Mast cells undergo degranulation, in which characteristic intracellular granules fuse with the plasma membrane and release many bioactive substances, such as enzymes ß-hexosaminidase and tryptase. Activity of mast cells in the toxicology model organism, zebrafish, has been monitored via tryptase release and cleavage of substrate N-α-benzoyl-dl-Arg-p-nitroanilide (BAPNA). An extensively used in vitro mast cell model for studying toxicant mechanisms is the RBL-2H3 cell line. However, instead of tryptase, granule contents such as ß-hexosaminidase have usually been employed as RBL-2H3 degranulation markers. To align RBL-2H3 cell toxicological studies to in vivo mast cell studies using zebrafish, we aimed to develop an RBL-2H3 tryptase assay. Unexpectedly, we discovered that tryptase release from RBL-2H3 cells is not detectable, using BAPNA substrate, despite optimized assay that can detect as little as 1 ng tryptase. Additional studies performed with another substrate, tosyl-Gly-Pro-Lys-pNA, and with an enzyme-linked immunosorbent assay, revealed a lack of tryptase protein released from stimulated RBL-2H3 cells. Furthermore, none of the eight rat tryptase genes (Tpsb2, Tpsab1, Tpsg1, Prss34, Gzmk, Gzma, Prss29, Prss41) is expressed in RBL-2H3 cells, even though all are found in RBL-2H3 genomic DNA and even though ß-hexosaminidase mRNA is constitutively expressed. Therefore, mast cell researchers should utilize ß-hexosaminidase or another reliable marker for RBL-2H3 degranulation studies, not tryptase. Comparative toxicity testing in RBL-2H3 cells in vitro and in zebrafish mast cells in vivo will require use of a degranulation reporter different from tryptase.


Subject(s)
Mast Cells/enzymology , Tryptases/analysis , Animals , Cell Degranulation , Cells, Cultured , Enzyme-Linked Immunosorbent Assay , Humans , Mast Cells/drug effects , Mice , Rats , Tryptases/genetics , Tryptases/metabolism , Zebrafish
9.
Toxicol Appl Pharmacol ; 349: 39-54, 2018 06 15.
Article in English | MEDLINE | ID: mdl-29630968

ABSTRACT

The antimicrobial agent triclosan (TCS) is used in products such as toothpaste and surgical soaps and is readily absorbed into oral mucosa and human skin. These and many other tissues contain mast cells, which are involved in numerous physiologies and diseases. Mast cells release chemical mediators through a process termed degranulation, which is inhibited by TCS. Investigation into the underlying mechanisms led to the finding that TCS is a mitochondrial uncoupler at non-cytotoxic, low-micromolar doses in several cell types and live zebrafish. Our aim was to determine the mechanisms underlying TCS disruption of mitochondrial function and of mast cell signaling. We combined super-resolution (fluorescence photoactivation localization) microscopy and multiple fluorescence-based assays to detail triclosan's effects in living mast cells, fibroblasts, and primary human keratinocytes. TCS disrupts mitochondrial nanostructure, causing mitochondria to undergo fission and to form a toroidal, "donut" shape. TCS increases reactive oxygen species production, decreases mitochondrial membrane potential, and disrupts ER and mitochondrial Ca2+ levels, processes that cause mitochondrial fission. TCS is 60 × more potent than the banned uncoupler 2,4-dinitrophenol. TCS inhibits mast cell degranulation by decreasing mitochondrial membrane potential, disrupting microtubule polymerization, and inhibiting mitochondrial translocation, which reduces Ca2+ influx into the cell. Our findings provide mechanisms for both triclosan's inhibition of mast cell signaling and its universal disruption of mitochondria. These mechanisms provide partial explanations for triclosan's adverse effects on human reproduction, immunology, and development. This study is the first to utilize super-resolution microscopy in the field of toxicology.


Subject(s)
Anti-Infective Agents, Local/toxicity , Calcium Signaling/drug effects , Mast Cells/drug effects , Mitochondria/drug effects , Mitochondria/ultrastructure , Triclosan/toxicity , 3T3 Cells , Animals , Cell Degranulation/drug effects , Endoplasmic Reticulum/drug effects , Fibroblasts/drug effects , Humans , Keratinocytes/drug effects , Membrane Potential, Mitochondrial/drug effects , Mice , Microtubules/drug effects , Microtubules/ultrastructure , Primary Cell Culture , Reactive Oxygen Species/metabolism
10.
Environ Pollut ; 235: 180-185, 2018 Apr.
Article in English | MEDLINE | ID: mdl-29289828

ABSTRACT

Nitrate accumulation in aquatic reservoirs from agricultural pollution has often been overlooked as a water quality hazard, yet a growing body of literature suggests negative effects on human and wildlife health following nitrate exposure. This research seeks to understand differences in oxygen consumption rates between different routes of laboratory nitrate exposure, whether via immersion or injection, in zebrafish (Danio rerio) embryos. Embryos were exposed within 1 h post fertilization (hpf) to 0, 10, and 100 mg/L NO3-N with sodium nitrate, or to counter ion control (CIC) treatments using sodium chloride. Embryos in the immersion treatments received an injection of 4 nL of appropriate treatment solution into the perivitelline space. At 24 hpf, Oxygen Consumption Rates (OCR) were measured and recorded in vivo using the Agilent Technologies XFe96 Extracellular Flux Analyzer and Spheroid Microplate. Immersion exposures did not induce significant changes in OCR, yet nitrate induced significant changes when injected through the embryo chorion. Injection of 10 and 100 mg/L NO3-N down-regulated OCR compared to the control treatment group. Injection of the 100 mg/L CIC also significantly down-regulated OCR compared to the control treatment group. Interestingly, the 100 mg/L NO3-N treatment further down-regulated OCR compared to the 100 mg/L CIC treatment, suggesting the potential for additive effects between the counter ion and the ion of interest. These data support that elevated nitrate exposure can alter normal metabolic activity by changing OCR in 24 hpf embryos. These results highlight the need for regularly examining the counter ion of laboratory nitrate compounds while conducting research with developing zebrafish, and justify examining different routes of laboratory nitrate exposure, as the chorion may act as an effective barrier to nitrate penetration in zebrafish, which may lead to conservative estimates of significant effects in other species for which nitrate more readily penetrates the chorion.


Subject(s)
Embryo, Nonmammalian/metabolism , Nitrates/toxicity , Water Pollutants, Chemical/toxicity , Animals , Chorion , Embryo, Nonmammalian/drug effects , Nitrates/metabolism , Nitrogen Oxides/metabolism , Water Pollutants, Chemical/metabolism , Zebrafish/embryology , Zebrafish/metabolism
11.
J Appl Toxicol ; 36(12): 1662-1667, 2016 12.
Article in English | MEDLINE | ID: mdl-27111768

ABSTRACT

Triclosan (TCS) is a synthetic antimicrobial agent used in many consumer goods at millimolar concentrations. As a result of exposure, TCS has been detected widely in humans. We have recently discovered that TCS is a proton ionophore mitochondrial uncoupler in multiple types of living cells. Here, we present novel data indicating that TCS is also a mitochondrial uncoupler in a living organism: 24-hour post-fertilization (hpf) zebrafish embryos. These experiments were conducted using a Seahorse Bioscience XFe 96 Extracellular Flux Analyzer modified for bidirectional temperature control, using the XF96 spheroid plate to position and measure one zebrafish embryo per well. Using this method, after acute exposure to TCS, the basal oxygen consumption rate (OCR) increases, without a decrease in survival or heartbeat rate. TCS also decreases ATP-linked respiration and spare respiratory capacity and increases proton leak: all indicators of mitochondrial uncoupling. Our data indicate, that TCS is a mitochondrial uncoupler in vivo, which should be taken into consideration when assessing the toxicity and/or pharmaceutical uses of TCS. This is the first example of usage of a Seahorse Extracellular Flux Analyzer to measure bioenergetic flux of a single zebrafish embryo per well in a 96-well assay format. The method developed in this study provides a high-throughput tool to identify previously unknown mitochondrial uncouplers in a living organism. Copyright © 2016 John Wiley & Sons, Ltd.


Subject(s)
Embryo, Nonmammalian/drug effects , Environmental Pollutants/toxicity , Mitochondria/drug effects , Triclosan/toxicity , Uncoupling Agents/toxicity , Zebrafish , Animals , Dose-Response Relationship, Drug , Mitochondria/metabolism , Oxygen Consumption/drug effects , Protons , Zebrafish/embryology , Zebrafish/metabolism
12.
J Appl Toxicol ; 36(11): 1446-59, 2016 11.
Article in English | MEDLINE | ID: mdl-27018130

ABSTRACT

Exposure to arsenic is a global health concern. We previously documented an inhibitory effect of inorganic Arsenite on IgE-mediated degranulation of RBL-2H3 mast cells (Hutchinson et al., 2011; J. Appl. Toxicol. 31: 231-241). Mast cells are tissue-resident cells that are positioned at the host-environment interface, thereby serving vital roles in many physiological processes and disease states, in addition to their well-known roles in allergy and asthma. Upon activation, mast cells secrete several mediators from cytoplasmic granules, in degranulation. The present study is an investigation of Arsenite's molecular target(s) in the degranulation pathway. Here, we report that arsenic does not affect degranulation stimulated by either the Ca(2) (+) ionophore A23187 or thapsigargin, which both bypass early signaling events. Arsenic also does not alter degranulation initiated by another non-IgE-mediated mast cell stimulant, the G-protein activator compound 48/80. However, arsenic inhibits Ca(2) (+) influx into antigen-activated mast cells. These results indicate that the target of arsenic in the degranulation pathway is upstream of the Ca(2) (+) influx. Phospho-Syk and phospho-p85 phosphoinositide 3-kinase enzyme-linked immunosorbent assays data show that arsenic inhibits early phosphorylation events. Taken together, this evidence indicates that the mechanism underlying arsenic inhibition of mast cell degranulation occurs at the early tyrosine phosphorylation steps in the degranulation pathway. Copyright © 2016 John Wiley & Sons, Ltd.


Subject(s)
Arsenites/toxicity , Calcium/metabolism , Cell Degranulation/drug effects , Environmental Pollutants/toxicity , Mast Cells/drug effects , Protein-Tyrosine Kinases/antagonists & inhibitors , Animals , Cell Line, Tumor , Cell Survival/drug effects , Mast Cells/metabolism , Mast Cells/physiology , Phosphoinositide-3 Kinase Inhibitors , Phosphorylation , Rats , Syk Kinase/antagonists & inhibitors
13.
J Appl Toxicol ; 36(6): 777-89, 2016 Jun.
Article in English | MEDLINE | ID: mdl-26204821

ABSTRACT

Triclosan (TCS) is an antimicrobial used widely in hospitals and personal care products, at ~10 mm. Human skin efficiently absorbs TCS. Mast cells are ubiquitous key players both in physiological processes and in disease, including asthma, cancer and autism. We previously showed that non-cytotoxic levels of TCS inhibit degranulation, the release of histamine and other mediators, from rat basophilic leukemia mast cells (RBL-2H3), and in this study, we replicate this finding in human mast cells (HMC-1.2). Our investigation into the molecular mechanisms underlying this effect led to the discovery that TCS disrupts adenosine triphosphate (ATP) production in RBL-2H3 cells in glucose-free, galactose-containing media (95% confidence interval EC50 = 7.5-9.7 µm), without causing cytotoxicity. Using these same glucose-free conditions, 15 µm TCS dampens RBL-2H3 degranulation by 40%. The same ATP disruption was found with human HMC-1.2 cells (EC50 4.2-13.7 µm), NIH-3 T3 mouse fibroblasts (EC50 4.8-7.4 µm) and primary human keratinocytes (EC50 3.0-4.1 µm) all with no cytotoxicity. TCS increases oxygen consumption rate in RBL-2H3 cells. Known mitochondrial uncouplers (e.g., carbonyl cyanide 3-chlorophenylhydrazone) previously were found to inhibit mast cell function. TCS-methyl, which has a methyl group in place of the TCS ionizable proton, affects neither degranulation nor ATP production at non-cytotoxic doses. Thus, the effects of TCS on mast cell function are due to its proton ionophore structure. In addition, 5 µm TCS inhibits thapsigargin-stimulated degranulation of RBL-2H3 cells: further evidence that TCS disrupts mast cell signaling. Our data indicate that TCS is a mitochondrial uncoupler, and TCS may affect numerous cell types and functions via this mechanism. Copyright © 2015 John Wiley & Sons, Ltd.


Subject(s)
Anti-Infective Agents, Local/pharmacology , Keratinocytes/drug effects , Mast Cells/drug effects , Mitochondria/drug effects , Oxidative Phosphorylation/drug effects , Triclosan/pharmacology , Uncoupling Agents/pharmacology , Animals , Anti-Infective Agents, Local/adverse effects , Anticarcinogenic Agents/adverse effects , Anticarcinogenic Agents/pharmacology , Carcinogens/antagonists & inhibitors , Carcinogens/toxicity , Cell Degranulation/drug effects , Cell Line , Cell Line, Tumor , Cell Survival/drug effects , Cells, Cultured , Humans , Keratinocytes/metabolism , Kinetics , Mast Cells/immunology , Mast Cells/metabolism , Mice , Mitochondria/metabolism , NIH 3T3 Cells , Rats , Thapsigargin/antagonists & inhibitors , Thapsigargin/toxicity , Triclosan/adverse effects , Triclosan/analogs & derivatives , Uncoupling Agents/adverse effects
14.
Immunogenetics ; 66(4): 267-79, 2014 Apr.
Article in English | MEDLINE | ID: mdl-24469064

ABSTRACT

The polymeric immunoglobulin (Ig) receptor (pIgR) is an integral transmembrane glycoprotein that plays an important role in the mammalian immune response by transporting soluble polymeric Igs across mucosal epithelial cells. Single pIgR genes, which are expressed in lymphoid organs including mucosal tissues, have been identified in several teleost species. A single pigr gene has been identified on zebrafish chromosome 2 along with a large multigene family consisting of 29 pigr-like (PIGRL) genes. Full-length transcripts from ten different PIGRL genes that encode secreted and putative inhibitory membrane-bound receptors have been characterized. Although PIGRL and pigr transcripts are detected in immune tissues, only PIGRL transcripts can be detected in lymphoid and myeloid cells. In contrast to pIgR which binds Igs, certain PIGRL proteins bind phospholipids. PIGRL transcript levels are increased after infection with Streptococcus iniae, suggesting a role for PIGRL genes during bacterial challenge. Transcript levels of PIGRL genes are decreased after infection with Snakehead rhabdovirus, suggesting that viral infection may suppress PIGRL function.


Subject(s)
Receptors, Polymeric Immunoglobulin/genetics , Receptors, Polymeric Immunoglobulin/metabolism , Zebrafish Proteins/genetics , Zebrafish Proteins/immunology , Zebrafish/genetics , Zebrafish/immunology , Amino Acid Sequence , Animals , Chromosome Mapping , Conserved Sequence , Evolution, Molecular , Fishes/genetics , Fishes/immunology , Gene Expression , Humans , Immunity, Innate/genetics , Ligands , Mammals/genetics , Mammals/immunology , Molecular Sequence Data , Multigene Family , Phospholipids/metabolism , Phylogeny , Protein Binding , Protein Structure, Tertiary , Receptors, Polymeric Immunoglobulin/chemistry , Rhabdoviridae Infections/genetics , Rhabdoviridae Infections/immunology , Rhabdoviridae Infections/metabolism , Sequence Homology, Amino Acid , Streptococcal Infections/genetics , Streptococcal Infections/immunology , Streptococcal Infections/metabolism , Zebrafish/metabolism , Zebrafish Proteins/metabolism
15.
J Vis Exp ; (81): e50671, 2013 Nov 01.
Article in English | MEDLINE | ID: mdl-24300285

ABSTRACT

Mast cells play important roles in allergic disease and immune defense against parasites. Once activated (e.g. by an allergen), they degranulate, a process that results in the exocytosis of allergic mediators. Modulation of mast cell degranulation by drugs and toxicants may have positive or adverse effects on human health. Mast cell function has been dissected in detail with the use of rat basophilic leukemia mast cells (RBL-2H3), a widely accepted model of human mucosal mast cells(3-5). Mast cell granule component and the allergic mediator ß-hexosaminidase, which is released linearly in tandem with histamine from mast cells(6), can easily and reliably be measured through reaction with a fluorogenic substrate, yielding measurable fluorescence intensity in a microplate assay that is amenable to high-throughput studies(1). Originally published by Naal et al.(1), we have adapted this degranulation assay for the screening of drugs and toxicants and demonstrate its use here. Triclosan is a broad-spectrum antibacterial agent that is present in many consumer products and has been found to be a therapeutic aid in human allergic skin disease(7-11), although the mechanism for this effect is unknown. Here we demonstrate an assay for the effect of triclosan on mast cell degranulation. We recently showed that triclosan strongly affects mast cell function(2). In an effort to avoid use of an organic solvent, triclosan is dissolved directly into aqueous buffer with heat and stirring, and resultant concentration is confirmed using UV-Vis spectrophotometry (using ε280 = 4,200 L/M/cm)(12). This protocol has the potential to be used with a variety of chemicals to determine their effects on mast cell degranulation, and more broadly, their allergic potential.


Subject(s)
Cell Degranulation/drug effects , Mast Cells/drug effects , Mast Cells/physiology , Triclosan/pharmacology , Animals , Anti-Infective Agents, Local/pharmacology , Buffers , Cell Degranulation/immunology , Cell Line, Tumor , Cytological Techniques/instrumentation , Cytological Techniques/methods , Leukemia, Basophilic, Acute/pathology , Mast Cells/cytology , Mast Cells/immunology , Rats , Solubility , Spectrophotometry, Ultraviolet , Triclosan/chemistry , Water/chemistry
16.
PLoS One ; 8(7): e68759, 2013.
Article in English | MEDLINE | ID: mdl-23874753

ABSTRACT

Understanding spatial distribution and dynamics of receptors within unperturbed membranes is essential for elucidating their role in antiviral signaling, but conventional studies of detergent-resistant membrane fractions cannot provide this information. Caveolae are integral to numerous signaling pathways and these membrane domains have been previously implicated in viral entry but not antiviral defense. This study shows, for the first time, the importance of spatio-temporal regulation of signaling receptors and the importance of the regulation of clustering for downstream signaling. A novel mechanism for virus evasion of host cell defenses is demonstrated through disruption of clusters of signaling molecules organized within caveolin-rich domains. Viral infection leads to a downregulation in Caveolin-1b (Cav-1b), disrupting clusters of CRFB1, a zebrafish type I interferon receptor (-R) subunit. Super-resolution microscopy has enabled the first single-molecule imaging of CRFB1 association with cav-1b-containing membrane domains. Strikingly, downregulation of Cav-1b, the major protein component of caveolae, caused CRFB1 clusters to disperse. Dispersal of CRFB1 clusters led to a suppressed antiviral immune response both in vitro and in vivo, through abrogation of downstream signaling. This response strongly suggests that CRFB1 organization within cav-1b-containing membrane domains is critical for IFN-mediated antiviral defense and presents a previously undescribed antiviral evasion strategy to alter IFN signaling and the antiviral immune response.


Subject(s)
Caveolin 1/metabolism , Disease Resistance , Receptors, Interferon/metabolism , Signal Transduction , Zebrafish Proteins/metabolism , Zebrafish/metabolism , Zebrafish/virology , Animals , Cell Membrane/metabolism , Disease Resistance/immunology , Fish Diseases/immunology , Fish Diseases/metabolism , Fish Diseases/virology , Immunity, Innate/genetics , Interferons/metabolism , Protein Binding , Zebrafish/immunology
17.
Sensors (Basel) ; 8(4): 2865-2885, 2008 Apr 28.
Article in English | MEDLINE | ID: mdl-27879854

ABSTRACT

This paper proposes a new methodology to improve location-sensing accuracy in wireless network environments eliminating the effects of non-line-of-sight errors. After collecting bulks of anonymous location measurements from a wireless network, the preparation stage of the proposed methodology begins. Investigating the collected location measurements in terms of signal features and geometric features, feature locations are identified. After the identification of feature locations, the non-line-of-sight error correction maps are generated. During the real-time location sensing stage, each user can request localization with a set of location measurements. With respected to the reported measurements, the pre-computed correction maps are applied. As a result, localization accuracy improves by eliminating the non-line-of-sight errors. A simulation result, assuming a typical dense urban environment, demonstrates the benefits of the proposed location sensing methodology.

SELECTION OF CITATIONS
SEARCH DETAIL
...