Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 22
Filter
Add more filters











Publication year range
1.
Cell Rep ; 30(7): 2055-2064.e5, 2020 02 18.
Article in English | MEDLINE | ID: mdl-32075752

ABSTRACT

Mechanisms underpinning airway epithelial homeostatic maintenance and ways to prevent its dysregulation remain elusive. Herein, we identify that ß-catenin phosphorylated at Y489 (p-ß-cateninY489) emerges during human squamous lung cancer progression. This led us to develop a model of airway basal stem cell (ABSC) hyperproliferation by driving Wnt/ß-catenin signaling, resulting in a morphology that resembles premalignant lesions and loss of ciliated cell differentiation. To identify small molecules that could reverse this process, we performed a high-throughput drug screen for inhibitors of Wnt/ß-catenin signaling. Our studies unveil Wnt inhibitor compound 1 (WIC1), which suppresses T-cell factor/lymphoid enhancer-binding factor (TCF/LEF) activity, reduces ABSC proliferation, induces ciliated cell differentiation, and decreases nuclear p-ß-cateninY489. Collectively, our work elucidates a dysregulated Wnt/p-ß-cateninY489 axis in lung premalignancy that can be modeled in vitro and identifies a Wnt/ß-catenin inhibitor that promotes airway homeostasis. WIC1 may therefore serve as a tool compound in regenerative medicine studies with implications for restoring normal airway homeostasis after injury.


Subject(s)
Lung/drug effects , Lung/metabolism , Stem Cells/drug effects , Stem Cells/metabolism , Wnt Proteins/antagonists & inhibitors , Wnt Signaling Pathway/drug effects , Animals , Bronchi/cytology , Bronchi/drug effects , Bronchi/metabolism , Bronchi/pathology , Cell Differentiation/drug effects , Drug Evaluation, Preclinical/methods , Female , High-Throughput Screening Assays/methods , Homeostasis/drug effects , Humans , Lung/cytology , Lung/pathology , Male , Mice , Mice, Inbred C57BL , Precancerous Conditions/metabolism , Precancerous Conditions/pathology , Small Molecule Libraries/pharmacology , Stem Cells/cytology , Stem Cells/pathology , Transfection , Wnt Proteins/metabolism , beta Catenin/antagonists & inhibitors , beta Catenin/metabolism
2.
Proc Natl Acad Sci U S A ; 116(37): 18597-18606, 2019 09 10.
Article in English | MEDLINE | ID: mdl-31439817

ABSTRACT

Burkholderia pseudomallei (Bp) and Burkholderia mallei (Bm) are Tier-1 Select Agents that cause melioidosis and glanders, respectively. These are highly lethal human infections with limited therapeutic options. Intercellular spread is a hallmark of Burkholderia pathogenesis, and its prominent ties to virulence make it an attractive therapeutic target. We developed a high-throughput cell-based phenotypic assay and screened ∼220,000 small molecules for their ability to disrupt intercellular spread by Burkholderia thailandensis, a closely related BSL-2 surrogate. We identified 268 hits, and cross-species validation found 32 hits that also disrupt intercellular spread by Bp and/or Bm Among these were a fluoroquinolone analog, which we named burkfloxacin (BFX), which potently inhibits growth of intracellular Burkholderia, and flucytosine (5-FC), an FDA-approved antifungal drug. We found that 5-FC blocks the intracellular life cycle at the point of type VI secretion system 5 (T6SS-5)-mediated cell-cell spread. Bacterial conversion of 5-FC to 5-fluorouracil and subsequently to fluorouridine monophosphate is required for potent and selective activity against intracellular Burkholderia In a murine model of fulminant respiratory melioidosis, treatment with BFX or 5-FC was significantly more effective than ceftazidime, the current antibiotic of choice, for improving survival and decreasing bacterial counts in major organs. Our results demonstrate the utility of cell-based phenotypic screening for Select Agent drug discovery and warrant the advancement of BFX and 5-FC as candidate therapeutics for melioidosis in humans.


Subject(s)
Burkholderia pseudomallei/drug effects , Ciprofloxacin/pharmacology , Drug Repositioning , Flucytosine/pharmacology , Melioidosis/drug therapy , Animals , Burkholderia pseudomallei/pathogenicity , Ciprofloxacin/analogs & derivatives , Ciprofloxacin/therapeutic use , Cytoplasm/drug effects , Cytoplasm/microbiology , Disease Models, Animal , Drug Evaluation, Preclinical , Female , Flucytosine/therapeutic use , HEK293 Cells , High-Throughput Screening Assays , Humans , Melioidosis/microbiology , Mice , Microbial Sensitivity Tests , Treatment Outcome , Virulence
3.
Neuro Oncol ; 20(6): 764-775, 2018 05 18.
Article in English | MEDLINE | ID: mdl-29136244

ABSTRACT

Background: Clinical trials of therapies directed against nodes of the signaling axis of phosphatidylinositol-3 kinase/Akt/mammalian target of rapamycin (mTOR) in glioblastoma (GBM) have had disappointing results. Resistance to mTOR inhibitors limits their efficacy. Methods: To determine mechanisms of resistance to chronic mTOR inhibition, we performed tandem screens on patient-derived GBM cultures. Results: An unbiased phosphoproteomic screen quantified phosphorylation changes associated with chronic exposure to the mTOR inhibitor rapamycin, and our analysis implicated a role for glycogen synthase kinase (GSK)3B attenuation in mediating resistance that was confirmed by functional studies. A targeted short hairpin RNA screen and further functional studies both in vitro and in vivo demonstrated that microtubule-associated protein (MAP)1B, previously associated predominantly with neurons, is a downstream effector of GSK3B-mediated resistance. Furthermore, we provide evidence that chronic rapamycin induces microtubule stability in a MAP1B-dependent manner in GBM cells. Additional experiments explicate a signaling pathway wherein combinatorial extracellular signal-regulated kinase (ERK)/mTOR targeting abrogates inhibitory phosphorylation of GSK3B, leads to phosphorylation of MAP1B, and confers sensitization. Conclusions: These data portray a compensatory molecular signaling network that imparts resistance to chronic mTOR inhibition in primary, human GBM cell cultures and points toward new therapeutic strategies.


Subject(s)
Gene Expression Regulation, Neoplastic/drug effects , Glioblastoma/pathology , Microtubule-Associated Proteins/metabolism , RNA, Small Interfering/genetics , Sirolimus/pharmacology , TOR Serine-Threonine Kinases/metabolism , Animals , Antibiotics, Antineoplastic/pharmacology , Apoptosis , Biomarkers, Tumor/genetics , Biomarkers, Tumor/metabolism , Cell Proliferation , Extracellular Signal-Regulated MAP Kinases/antagonists & inhibitors , Extracellular Signal-Regulated MAP Kinases/genetics , Extracellular Signal-Regulated MAP Kinases/metabolism , Glioblastoma/drug therapy , Glioblastoma/genetics , Glycogen Synthase Kinase 3 beta/antagonists & inhibitors , Glycogen Synthase Kinase 3 beta/genetics , Glycogen Synthase Kinase 3 beta/metabolism , Humans , Male , Mice , Mice, Inbred NOD , Mice, SCID , Microtubule-Associated Proteins/antagonists & inhibitors , Microtubule-Associated Proteins/genetics , Phosphatidylinositol 3-Kinases/genetics , Phosphatidylinositol 3-Kinases/metabolism , Phosphoinositide-3 Kinase Inhibitors , Phosphorylation , Proto-Oncogene Proteins c-akt/antagonists & inhibitors , Proto-Oncogene Proteins c-akt/genetics , Proto-Oncogene Proteins c-akt/metabolism , Signal Transduction , TOR Serine-Threonine Kinases/antagonists & inhibitors , TOR Serine-Threonine Kinases/genetics , Tumor Cells, Cultured , Xenograft Model Antitumor Assays
4.
Nat Biomed Eng ; 2(2): 124-137, 2018 02.
Article in English | MEDLINE | ID: mdl-31015629

ABSTRACT

As cells with aberrant force-generating phenotypes can directly lead to disease, cellular force-generation mechanisms are high-value targets for new therapies. Here, we show that single-cell force sensors embedded in elastomers enable single-cell force measurements with ~100-fold improvement in throughput than was previously possible. The microtechnology is scalable and seamlessly integrates with the multi-well plate format, enabling highly parallelized time-course studies. In this regard, we show that airway smooth muscle cells isolated from fatally asthmatic patients have innately greater and faster force-generation capacity in response to stimulation than healthy control cells. By simultaneously tracing agonist-induced calcium flux and contractility in the same cell, we show that the calcium level is ultimately a poor quantitative predictor of cellular force generation. Finally, by quantifying phagocytic forces in thousands of individual human macrophages, we show that force initiation is a digital response (rather than a proportional one) to the proper immunogen. By combining mechanobiology at the single-cell level with high-throughput capabilities, this microtechnology can support drug-discovery efforts for clinical conditions associated with aberrant cellular force generation.


Subject(s)
Elastomers/chemistry , Single-Cell Analysis/methods , Asthma/pathology , Cell Differentiation , Cells, Cultured , Fluorescent Dyes/chemistry , Formoterol Fumarate/pharmacology , Heterocyclic Compounds, 4 or More Rings/pharmacology , Humans , Macrophages/cytology , Macrophages/metabolism , Mesenchymal Stem Cells/cytology , Mesenchymal Stem Cells/metabolism , Microscopy, Fluorescence , Myocardial Contraction/drug effects , Myocytes, Smooth Muscle/cytology , Myocytes, Smooth Muscle/metabolism , Phagocytosis/drug effects
5.
Nat Biomed Eng ; 2(4): 265, 2018 Apr.
Article in English | MEDLINE | ID: mdl-31015734

ABSTRACT

In the version of this Article originally published, in Fig. 1a, all cells in the top schematic were missing, and in the bottom-left schematic showing multiple pattern shapes, two cells were missing in the bottom-right corner. This figure has now been updated in all versions of the Article.

6.
Sci Rep ; 7(1): 11261, 2017 09 12.
Article in English | MEDLINE | ID: mdl-28900159

ABSTRACT

Discovery of first-in-class medicines for treating cancer is limited by concerns with their toxicity and safety profiles, while repurposing known drugs for new anticancer indications has become a viable alternative. Here, we have developed a new approach that utilizes cell cycle arresting patterns as unique molecular signatures for prioritizing FDA-approved drugs with repurposing potential. As proof-of-principle, we conducted large-scale cell cycle profiling of 884 FDA-approved drugs. Using cell cycle indexes that measure changes in cell cycle profile patterns upon chemical perturbation, we identified 36 compounds that inhibited cancer cell viability including 6 compounds that were previously undescribed. Further cell cycle fingerprint analysis and 3D chemical structural similarity clustering identified unexpected FDA-approved drugs that induced DNA damage, including clinically relevant microtubule destabilizers, which was confirmed experimentally via cell-based assays. Our study shows that computational cell cycle profiling can be used as an approach for prioritizing FDA-approved drugs with repurposing potential, which could aid the development of cancer therapeutics.


Subject(s)
Antineoplastic Agents/isolation & purification , Antineoplastic Agents/pharmacology , Cell Cycle/drug effects , Cell Proliferation/drug effects , Drug Evaluation, Preclinical/methods , Drug Repositioning/methods , Cell Line, Tumor , Computational Biology/methods , Humans
7.
BMC Complement Altern Med ; 17(1): 381, 2017 Aug 02.
Article in English | MEDLINE | ID: mdl-28768515

ABSTRACT

BACKGROUND: Epidemiological and experimental evidences have shown cancer as a leading cause of death worldwide. Although the folklore use of plants as a reliable source of health-restoring principles is well-documented, the search for more of such plants that are active against diseases, such as cancer, continues. We report here a laboratory-based evidence of the relevance of an ethanol leaf extract of Anogeissus leiocarpus (A2L) in comparison with resveratrol, a natural polyphenol, in cancer therapy. METHODS: The quantitative assessment of flavonoid and phenolic contents involved quercetin and gallic acid as standards, respectively were determined using spectrophotometry. Cytotoxicity was determined fluorometrically using propidium-iodide-staining method. Antioxidant status, adenosine triphosphate (ATP) levels, caspase activities and mitochondrial integrity were assessed using fluorometry/luminometry. RESULTS: The antioxidant assay demonstrated that A2L possesses a strong antioxidant capacity as compared with the reference compounds, ascorbic acid and butylated hydroxytoluene. This is further buttressed by the significantly high level of phenolics obtained in the quantitative assessment of the extract. A 72-h post-treatment examination indicated that both A2L and resveratrol modulate the proliferation of HepG2 liver carcinoma cells in a time- and concentration-dependent manner. Determination of the total nuclei area, propidium-iodide negative and positive nuclei areas all further buttress the modulation of cell proliferation by A2L and resveratrol with the indication that the observed cell death is due to apoptosis and necrosis at lower and higher concentrations of treatments respectively. At lower concentrations (0.39-3.13 µg/mL), resveratrol possesses higher tendencies to activate caspases 3 and 7. Bioenergetically, both resveratrol and A2L do not adversely affect the cells at lower concentrations (0.39-6.25 µg/mL for resveratrol and 12.5-100.0 µg/mL for A2L) except at higher concentrations (12.5-25.0 µg/mL for resveratrol and 200-800 µg/mL for A2L) which are more pronounced in A2L-treated cells. Furthermore, the antioxidant status of HepG2 cells is not perturbed by resveratrol as compared with A2L. Assessment of 24-h post-treatment mitochondrial function shows that resveratrol is not mitotoxic as compared with A2L which exhibits mitotoxicity at its highest concentration. CONCLUSIONS: Taken together, findings from this study showed that A2L possesses strong antiproliferative activity and its prospect in the management of hepatocellular carcinoma deserves further investigation.


Subject(s)
Antineoplastic Agents, Phytogenic/therapeutic use , Antioxidants/therapeutic use , Carcinoma, Hepatocellular/drug therapy , Combretaceae/chemistry , Liver Neoplasms/drug therapy , Phytotherapy , Stilbenes/therapeutic use , Adenosine Triphosphate/metabolism , Antineoplastic Agents, Phytogenic/pharmacology , Antioxidants/pharmacology , Apoptosis , Carcinoma, Hepatocellular/metabolism , Caspases/metabolism , Cell Proliferation , Combretaceae/classification , Flavonoids/pharmacology , Flavonoids/therapeutic use , Hep G2 Cells , Humans , Liver Neoplasms/metabolism , Mitochondria/drug effects , Mitochondria/metabolism , Necrosis , Phenols/pharmacology , Phenols/therapeutic use , Plant Extracts/pharmacology , Plant Extracts/therapeutic use , Plant Leaves , Resveratrol , Stilbenes/pharmacology
8.
Environ Mol Mutagen ; 58(7): 522-528, 2017 08.
Article in English | MEDLINE | ID: mdl-28640454

ABSTRACT

Genotoxicity is thought to be the cause of many cancers. Genotoxicity due to environmental toxins may be partly responsible for the dramatic increase in the incidence of papillary thyroid cancer over the past two decades. Here, we present a fully automatable assay platform that directly quantifies the phosphorylation of nuclear histone gamma H2AX (γH2AX), a specific cellular marker for DNA double strand breaks (DSBs) via immunohistochemistry and laser scanning cytometry. It multiplexes γH2AX with total cell number measured as propidium iodide and calculates the percentage of cells with DSBs. Validation of this assay using NTHY-ori-3-1 human thyroid cells and etoposide showed that it was an excellent choice for high throughput applications. We used the assay to test the genotoxic effects of the EPA Toxcast Phase 1 pesticide library of 309 compounds. Compounds were evaluated in dose response and the DSB was quantified. We found that 19 pesticides induce DSB in vitro, highlighting a need to further assess these pesticides for their long-term oncogenic effects on the thyroid gland. Environ. Mol. Mutagen. 58:522-528, 2017. © 2017 Wiley Periodicals, Inc.


Subject(s)
DNA Damage , High-Throughput Screening Assays/methods , Histones/genetics , Mutagenicity Tests/methods , Mutagens/toxicity , Pesticides/toxicity , Thyroid Gland/drug effects , Cell Line , Dose-Response Relationship, Drug , Humans , Thyroid Gland/pathology
9.
J Integr Med ; 15(3): 214-230, 2017 05.
Article in English | MEDLINE | ID: mdl-28494852

ABSTRACT

OBJECTIVE: The fatality of cancer is mostly dependent on the possibility of occurrence of metastasis. Thus, if the development of metastasis can be prevented through novel therapeutic strategies targeted against this process, then the success of cancer treatment will drastically increase. In this study, therefore, we evaluated the antimetastatic potentials of an extract of Khaya senegalensis and curcumin on the metastatic liver cell line HepG2, and also assessed the anticancer property of the extract. METHODS: Cells were cultured and treated with graded concentrations of test substances for 24, 48, or 72 h with provisions made for negative controls. Treated cells were assessed as follows: nanotechnologically - atomic force microscopy (AFM) was used to determine cell stiffness; biochemically - cell cytotoxicity, glutathione level and adenosine triphosphate status, caspase activation and mitochondrial toxicity were considered; and microbiologically - a carrot disk assay was used to assess the anticancer property of the extract of K. senegalensis. RESULTS: Curcumin and K. senegalensis increased the cell stiffness by 2.6- and 4.0-fold respectively, indicating their antimetastatic effects. Corresponding changes in redox (glutathione level) and energy (adenosine triphosphate) status of the cells were also demonstrated. Further mechanistic studies indicated that curcumin was not mitotoxic in HepG2 cells unlike the K. senegalensis extract. In addition, the extract potently inhibited the Agrobacterium tumefaciens-induced genetic transformation based on carrot disk assay. CONCLUSION: Cell elasticity measurement data, using AFM, strongly suggested, for the first time, that both curcumin and the extract of K. senegalensis exhibited antimetastatic properties on HepG2 cells.


Subject(s)
Antineoplastic Agents, Phytogenic/pharmacology , Curcuma , Curcumin/pharmacology , Meliaceae , Neoplasm Metastasis/prevention & control , Phytotherapy , Plant Extracts/pharmacology , Adenosine Triphosphate/metabolism , Antineoplastic Agents, Phytogenic/therapeutic use , Apoptosis , Cell Proliferation , Curcumin/therapeutic use , Elasticity , Glutathione/metabolism , Hep G2 Cells , Humans , Microscopy, Atomic Force , Neoplasm Invasiveness/prevention & control , Oxidation-Reduction , Plant Extracts/therapeutic use
10.
Analyst ; 139(5): 943-53, 2014 Mar 07.
Article in English | MEDLINE | ID: mdl-24260774

ABSTRACT

Relationships among fourteen different biological responses (including ten signaling pathway activities and four cytotoxicity effects) of murine macrophage (RAW264.7) and bronchial epithelial (BEAS-2B) cells exposed to six metal and metal oxide nanoparticles (NPs) were analyzed using both statistical and data mining approaches. Both the pathway activities and cytotoxicity effects were assessed using high-throughput screening (HTS) over an exposure period of up to 24 h and concentration range of 0.39-200 mg L(-1). HTS data were processed by outlier removal, normalization, and hit-identification (for significantly regulated cellular responses) to arrive at reliable multiparametric bioactivity profiles for the NPs. Association rule mining was then applied to the bioactivity profiles followed by a pruning process to remove redundant rules. The non-redundant association rules indicated that "significant regulation" of one or more cellular responses implies regulation of other (associated) cellular response types. Pairwise correlation analysis (via Pearson's χ(2) test) and self-organizing map clustering of the different cellular response types indicated consistency with the identified non-redundant association rules. Furthermore, in order to explore the potential use of association rules as a tool for data-driven hypothesis generation, specific pathway activity experiments were carried out for ZnO NPs. The experimental results confirmed the association rule identified for the p53 pathway and mitochondrial superoxide levels (via MitoSox reagent) and further revealed that blocking of the transcriptional activity of p53 lowered the MitoSox signal. The present approach of using association rule mining for data-driven hypothesis generation has important implications for streamlining multi-parameter HTS assays, improving the understanding of NP toxicity mechanisms, and selection of endpoints for the development of nanomaterial structure-activity relationships.


Subject(s)
High-Throughput Screening Assays/methods , Metal Nanoparticles/toxicity , Oxides/toxicity , Animals , Cell Line , Cell Survival/drug effects , Cell Survival/physiology , Mice
11.
Proc Natl Acad Sci U S A ; 110(50): E4904-12, 2013 Dec 10.
Article in English | MEDLINE | ID: mdl-24191014

ABSTRACT

Pathogenic microorganisms and toxins have evolved a variety of mechanisms to gain access to the host-cell cytosol and thereby exert virulent effects upon the host. One common mechanism of cellular entry requires trafficking to an acidified endosome, which promotes translocation across the host membrane. To identify small-molecule inhibitors that block this process, a library of 30,000 small molecules was screened for inhibitors of anthrax lethal toxin. Here we report that 4-bromobenzaldehyde N-(2,6-dimethylphenyl)semicarbazone, the most active compound identified in the screen, inhibits intoxication by lethal toxin and blocks the entry of multiple other acid-dependent bacterial toxins and viruses into mammalian cells. This compound, which we named EGA, also delays lysosomal targeting and degradation of the EGF receptor, indicating that it targets host-membrane trafficking. In contrast, EGA does not block endosomal recycling of transferrin, retrograde trafficking of ricin, phagolysosomal trafficking, or phagosome permeabilization by Franciscella tularensis. Furthermore, EGA does not neutralize acidic organelles, demonstrating that its mechanism of action is distinct from pH-raising agents such as ammonium chloride and bafilomycin A1. EGA is a powerful tool for the study of membrane trafficking and represents a class of host-targeted compounds for therapeutic development to treat infectious disease.


Subject(s)
Bacterial Toxins/antagonists & inhibitors , Endosomes/drug effects , High-Throughput Screening Assays/methods , Semicarbazones/pharmacology , Virus Internalization/drug effects , Amines , Animals , Biological Transport/physiology , Caspase 1/metabolism , Chromatography, Liquid , Endosomes/physiology , Flow Cytometry , HeLa Cells , Humans , Macrophages , Magnetic Resonance Spectroscopy , Mass Spectrometry , Mice , Mice, Transgenic , Microscopy, Fluorescence , Molecular Structure , Phagocytosis/drug effects , Phagocytosis/physiology , Semicarbazones/chemistry , Small Molecule Libraries , Structure-Activity Relationship
12.
Arch Toxicol ; 87(1): 99-109, 2013 Jan.
Article in English | MEDLINE | ID: mdl-22885792

ABSTRACT

To uncover the size influence of TiO(2) nanoparticles on their potential toxicity, the cytotoxicity of different-sized TiO(2) nanoparticles with and without photoactivation was tested. It was demonstrated that without photoactivation, TiO(2) nanoparticles were inert up to 100 µg/ml. On the contrary, with photoactivation, the toxicity of TiO(2) nanoparticles significantly increased, which correlated well with the specific surface area of the particles. Our results also suggest that the generation of hydroxyl radicals and reactive oxygen species (ROS)-mediated damage to the surface-adsorbed biomolecules could be the two major reasons for the cytotoxicity of TiO(2) nanoparticles after photoactivation. Higher ROS generation from smaller particles was detected under both biotic and abiotic conditions. Smaller particles could adsorb more proteins, which was confirmed by thermogravimetric analysis. To further investigate the influence of the generation of hydroxyl radicals and adsorption of protein, poly (ethylene-alt-maleic anhydride) (PEMA) and chitosan were used to coat TiO(2) nanoparticles. The results confirmed that surface coating of TiO(2) nanoparticles could reduce such toxicity after photoactivation, by hindering adsorption of biomolecules and generation of hydroxyl radical (·OH) during photoactivation.


Subject(s)
Dermatitis, Phototoxic , Metal Nanoparticles/toxicity , Particle Size , Adsorption , Animals , Cell Line/drug effects , Chitosan/chemistry , Chitosan/pharmacology , Coated Materials, Biocompatible/toxicity , Hydroxyl Radical/metabolism , Maleates/chemistry , Maleates/pharmacology , Metal Nanoparticles/chemistry , Metal Nanoparticles/ultrastructure , Mice , Microscopy, Electron, Scanning , Polyethylenes/chemistry , Polyethylenes/pharmacology , Proteins/metabolism , Reactive Oxygen Species/metabolism , Toxicity Tests/methods , Ultraviolet Rays
13.
Arch Toxicol ; 87(6): 1075-86, 2013 Jun.
Article in English | MEDLINE | ID: mdl-22983807

ABSTRACT

The aim of this study is to uncover the size influence of poly (lactic-co-glycolic acid) (PLGA) and titanium dioxide (TiO(2)) nanoparticles on their potential cytotoxicity. PLGA and TiO(2) nanoparticles of three different sizes were thoroughly characterized before in vitro cytotoxic tests which included viability, generation of reactive oxygen species (ROS), mitochondrial depolarization, integrity of plasma membrane, intracellular calcium influx and cytokine release. Size-dependent cytotoxic effect was observed in both RAW264.7 cells and BEAS-2B cells after cells were incubated with PLGA or TiO(2) nanoparticles for 24 h. Although PLGA nanoparticles did not trigger significantly lethal toxicity up to a concentration of 300 µg/ml, the TNF-α release after the stimulation of PLGA nanoparticles should not be ignored especially in clinical applications. Relatively more toxic TiO(2) nanoparticles triggered cell death, ROS generation, mitochondrial depolarization, plasma membrane damage, intracellular calcium concentration increase and size-dependent TNF-α release, especially at a concentration higher than 100 µg/ml. These cytotoxic effects could be due to the size-dependent interaction between nanoparticles and biomolecules, as smaller particles tend to adsorb more biomolecules. In summary, we demonstrated that the ability of protein adsorption could be an important paradigm to predict the in vitro cytotoxicity of nanoparticles, especially for low toxic nanomaterials such as PLGA and TiO(2) nanoparticles.


Subject(s)
Epithelial Cells/drug effects , Lactic Acid/toxicity , Lung/drug effects , Macrophages/drug effects , Metal Nanoparticles/toxicity , Polyglycolic Acid/toxicity , Titanium/toxicity , Adsorption , Animals , Calcium Signaling/drug effects , Cell Membrane/drug effects , Cell Membrane/metabolism , Cell Membrane/pathology , Cell Survival/drug effects , Dose-Response Relationship, Drug , Epithelial Cells/immunology , Epithelial Cells/metabolism , Epithelial Cells/pathology , Humans , Inflammation Mediators/metabolism , Lactic Acid/metabolism , Lung/immunology , Lung/metabolism , Lung/pathology , Macrophages/immunology , Macrophages/metabolism , Macrophages/pathology , Membrane Potential, Mitochondrial/drug effects , Mice , Oxidative Stress/drug effects , Particle Size , Polyglycolic Acid/metabolism , Polylactic Acid-Polyglycolic Acid Copolymer , Protein Binding , Reactive Oxygen Species/metabolism , Serum Albumin, Bovine/metabolism , Time Factors , Titanium/metabolism , Tumor Necrosis Factor-alpha/metabolism
14.
Infect Immun ; 79(8): 3302-8, 2011 Aug.
Article in English | MEDLINE | ID: mdl-21576335

ABSTRACT

Anthrax edema toxin (ET) is one of two binary toxins produced by Bacillus anthracis that contributes to the virulence of this pathogen. ET is an adenylate cyclase that generates high levels of cyclic AMP (cAMP), causing alterations in multiple host cell signaling pathways. We previously demonstrated that ET increases cell surface expression of the anthrax toxin receptors (ANTXR) in monocyte-derived cells and promotes dendritic cell (DC) migration toward the lymph node-homing chemokine MIP-3ß. In this work, we sought to determine if glycogen synthase kinase 3 (GSK-3) is important for ET-induced modulation of macrophage and DC function. We demonstrate that inhibition of GSK-3 dampens ET-induced maturation and migration processes of monocyte-derived dendritic cells (MDDCs). Additional studies reveal that the ET-induced expression of ANTXR in macrophages was decreased when GSK-3 activity was disrupted with chemical inhibitors or with small interfering RNA (siRNA) targeting GSK-3. Further examination of the ET induction of ANTXR revealed that a dominant negative form of CREB could block the ET induction of ANTXR, suggesting that CREB or a related family member was involved in the upregulation of ANTXR. Because CREB and GSK-3 activity appeared to be important for ET-induced ANTXR expression, the impact of GSK-3 on ET-induced CREB activity was examined in RAW 264.7 cells possessing a CRE-luciferase reporter. As with ANTXR expression, the ET induction of the CRE reporter was decreased by reducing GSK-3 activity. These studies not only provide insight into host pathways targeted by ET but also shed light on interactions between GSK-3 and CREB pathways in host immune cells.


Subject(s)
Anthrax/immunology , Anthrax/pathology , Antigens, Bacterial/toxicity , Bacterial Toxins/toxicity , Dendritic Cells/immunology , Glycogen Synthase Kinase 3/metabolism , Macrophages/immunology , Receptors, Peptide/metabolism , Animals , Cell Line , Host-Pathogen Interactions , Humans , Mice
15.
ACS Nano ; 5(4): 2756-2769, 2011 Apr 26.
Article in English | MEDLINE | ID: mdl-21366263

ABSTRACT

Most in vitro toxicity studies on engineered nanomaterials (ENMs) use transformed rather than primary cells for logistical reasons. However, primary cells may provide a more appropriate connection to in vivo toxicity because these cells maintain their phenotypic fidelity and are also capable of differentiating into lineages that may be differently affected by potentially hazardous ENMs. Few studies to date have focused on the role of cellular differentiation in determining ENM toxicity. We compared the response of undifferentiated and differentiated primary human bronchial epithelial (NHBE) cells to cationic mesoporous silica nanoparticles (MSNPs) that are coated with polyethyleneimine (PEI) since this polymer is known to exert differential cytotoxicity depending on its molecular weight and cationic density. The attachment of cationic PEI polymers to the MSNP surface was used to assess these materials' toxicological potential in undifferentiated and differentiated human bronchial epithelial cells, using a multiparametric assay that screens for an integrated set of sublethal and lethal response outcomes. MSNPs coated with high molecular weight (10 and 25 kD) polymers were more toxic in differentiated cells than particles coated with shorter length polymers. The increased susceptibility of the differentiated cells is in agreement with more abundant expression of a proteoglycan, syndecan-1, which contains copious heparin sulfate side chains. Pretreatment with heparinase to remove the negatively charged sulfates decreased MSNP-PEI binding to the cell surface and lowered the cytotoxic potential of the cationic particles. These data demonstrate the importance of studying cellular differentiation as an important variable in the response of primary cells to toxic ENM properties.


Subject(s)
Bronchi/drug effects , Cations , Cell Differentiation , Nanoparticles , Syndecan-1/metabolism , Blotting, Western , Bronchi/cytology , Bronchi/metabolism , Cells, Cultured , Epithelial Cells/drug effects , Epithelial Cells/metabolism , Erythrocytes/drug effects , Hemolysis/drug effects , Humans , Microscopy, Confocal , Microscopy, Electron, Transmission
16.
ACS Nano ; 5(3): 1805-17, 2011 Mar 22.
Article in English | MEDLINE | ID: mdl-21323332

ABSTRACT

Because of concerns about the safety of a growing number of engineered nanomaterials (ENM), it is necessary to develop high-throughput screening and in silico data transformation tools that can speed up in vitro hazard ranking. Here, we report the use of a multiparametric, automated screening assay that incorporates sublethal and lethal cellular injury responses to perform high-throughput analysis of a batch of commercial metal/metal oxide nanoparticles (NP) with the inclusion of a quantum dot (QD1). The responses chosen for tracking cellular injury through automated epifluorescence microscopy included ROS production, intracellular calcium flux, mitochondrial depolarization, and plasma membrane permeability. The z-score transformed high volume data set was used to construct heat maps for in vitro hazard ranking as well as showing the similarity patterns of NPs and response parameters through the use of self-organizing maps (SOM). Among the materials analyzed, QD1 and nano-ZnO showed the most prominent lethality, while Pt, Ag, SiO2, Al2O3, and Au triggered sublethal effects but without cytotoxicity. In order to compare the in vitro with the in vivo response outcomes in zebrafish embryos, NPs were used to assess their impact on mortality rate, hatching rate, cardiac rate, and morphological defects. While QDs, ZnO, and Ag induced morphological abnormalities or interfered in embryo hatching, Pt and Ag exerted inhibitory effects on cardiac rate. Ag toxicity in zebrafish differed from the in vitro results, which is congruent with this material's designation as extremely dangerous in the environment. Interestingly, while toxicity in the initially selected QD formulation was due to a solvent (toluene), supplementary testing of additional QDs selections yielded in vitro hazard profiling that reflect the release of chalcogenides. In conclusion, the use of a high-throughput screening, in silico data handling and zebrafish testing may constitute a paradigm for rapid and integrated ENM toxicological screening.


Subject(s)
Biological Assay/methods , Drug Evaluation, Preclinical/methods , Embryo, Nonmammalian/drug effects , Microscopy, Fluorescence/methods , Nanostructures/toxicity , Quantum Dots , Toxicity Tests/methods , Animals , Embryo, Nonmammalian/pathology , Zebrafish
17.
Environ Sci Technol ; 45(4): 1695-702, 2011 Feb 15.
Article in English | MEDLINE | ID: mdl-21250674

ABSTRACT

The response of a murine macrophage cell line exposed to a library of seven metal and metal oxide nanoparticles was evaluated via High Throughput Screening (HTS) assay employing luciferase-reporters for ten independent toxicity-related signaling pathways. Similarities of toxicity response among the nanoparticles were identified via Self-Organizing Map (SOM) analysis. This analysis, applied to the HTS data, quantified the significance of the signaling pathway responses (SPRs) of the cell population exposed to nanomaterials relative to a population of untreated cells, using the Strictly Standardized Mean Difference (SSMD). Given the high dimensionality of the data and relatively small data set, the validity of the SOM clusters was established via a consensus clustering technique. Analysis of the SPR signatures revealed two cluster groups corresponding to (i) sublethal pro-inflammatory responses to Al2O3, Au, Ag, SiO2 nanoparticles possibly related to ROS generation, and (ii) lethal genotoxic responses due to exposure to ZnO and Pt nanoparticles at a concentration range of 25-100 µg/mL at 12 h exposure. In addition to identifying and visualizing clusters and quantifying similarity measures, the SOM approach can aid in developing predictive quantitative-structure relations; however, this would require significantly larger data sets generated from combinatorial libraries of engineered nanoparticles.


Subject(s)
Metal Nanoparticles/toxicity , Signal Transduction/drug effects , Animals , Cell Line , Luciferases/drug effects , Luciferases/metabolism , Macrophages , Metal Nanoparticles/chemistry , Mice , Nanostructures , Oxides/chemistry , Oxides/toxicity
18.
PLoS Pathog ; 7(12): e1002469, 2011 Dec.
Article in English | MEDLINE | ID: mdl-22241984

ABSTRACT

Anthrax is a potentially fatal disease resulting from infection with Bacillus anthracis. The outcome of infection is influenced by pathogen-encoded virulence factors such as lethal toxin (LT), as well as by genetic variation within the host. To identify host genes controlling susceptibility to anthrax, a library of congenic mice consisting of strains with homozygous chromosomal segments from the LT-responsive CAST/Ei strain introgressed on a LT-resistant C57BL/6 (B6) background was screened for response to LT. Three congenic strains containing CAST/Ei regions of chromosome 11 were identified that displayed a rapid inflammatory response to LT similar to, but more severe than that driven by a LT-responsive allele of the inflammasome constituent NRLP1B. Importantly, increased response to LT in congenic mice correlated with greater resistance to infection by the Sterne strain of B. anthracis. The genomic region controlling the inflammatory response to LT was mapped to 66.36-74.67 Mb on chromosome 11, a region that encodes the LT-responsive CAST/Ei allele of Nlrp1b. However, known downstream effects of NLRP1B activation, including macrophage pyroptosis, cytokine release, and leukocyte infiltration could not fully explain the response to LT or the resistance to B. anthracis Sterne in congenic mice. Further, the exacerbated response in congenic mice is inherited in a recessive manner while the Nlrp1b-mediated response to LT is dominant. Finally, congenic mice displayed increased responsiveness in a model of sepsis compared with B6 mice. In total, these data suggest that allelic variation of one or more chromosome 11 genes in addition to Nlrp1b controls the severity of host response to multiple inflammatory stimuli and contributes to resistance to B. anthracis Sterne. Expression quantitative trait locus analysis revealed 25 genes within this region as high priority candidates for contributing to the host response to LT.


Subject(s)
Alleles , Anthrax/genetics , Bacillus anthracis , Chromosomes, Mammalian/genetics , Genetic Variation , Immunity, Innate/genetics , Animals , Anthrax/immunology , Antigens, Bacterial/immunology , Bacterial Toxins/immunology , Chromosomes, Mammalian/immunology , Inflammation/genetics , Inflammation/immunology , Mice , Quantitative Trait Loci/immunology
19.
J Immunol ; 184(1): 17-20, 2010 Jan 01.
Article in English | MEDLINE | ID: mdl-19949100

ABSTRACT

Pathogenesis of Bacillus anthracis is associated with the production of lethal toxin (LT), which activates the murine Nalp1b/Nlrp1b inflammasome and induces caspase-1-dependent pyroptotic death in macrophages and dendritic cells. In this study, we investigated the effect of allelic variation of Nlrp1b on the outcome of LT challenge and infection by B. anthracis spores. Nlrp1b allelic variation did not alter the kinetics or pathology of end-stage disease induced by purified LT, suggesting that, in contrast to previous reports, macrophage lysis does not contribute directly to LT-mediated pathology. However, animals expressing a LT-sensitive allele of Nlrp1b showed an early inflammatory response to LT and increased resistance to infection by B. anthracis. Data presented here support a model whereby LT-mediated activation of Nlrp1b and subsequent lysis of macrophages is not a mechanism used by B. anthracis to promote virulence, but rather a protective host-mediated innate immune response.


Subject(s)
Anthrax/genetics , Anthrax/immunology , Apoptosis Regulatory Proteins/genetics , Genetic Predisposition to Disease , Animals , Antigens, Bacterial/toxicity , Bacillus anthracis/immunology , Bacillus anthracis/pathogenicity , Bacterial Toxins/toxicity , Macrophages/immunology , Mice , Mice, Inbred C57BL , Mice, Transgenic
20.
Microbes Infect ; 10(6): 613-9, 2008 May.
Article in English | MEDLINE | ID: mdl-18467145

ABSTRACT

The role of macrophages in the pathogenesis of anthrax is unresolved. Macrophages are believed to support the initiation of infection by Bacillus anthracis spores, yet are also sporicidal. Furthermore, it is believed that the anthrax toxins suppress normal macrophage function. However, the significance of toxin effects on macrophages has not been addressed in an in vivo infection model. We used mutant derivatives of murine macrophage RAW264.7 cells that are toxin receptor-negative (R3D) to test the role of toxin-targeting of macrophages during a challenge with spores of the Ames strain of B. anthracis in both in vivo and in vitro models. We found that the R3D cells were able to control challenge with Ames when mice were inoculated with the cells prior to spore challenge. These findings were confirmed in vitro by high dose spore infection of macrophages. Interestingly, whereas the R3D cells provided a significantly greater survival advantage against spores than did the wild type RAW264.7 cells or R3D-complemented cells, the protection afforded the mutant and wild type cells was equivalent against a bacillus challenge. The findings appear to be the first specific test of the role of toxin targeting of macrophages during infection with B. anthracis spores.


Subject(s)
Anthrax/pathology , Antigens, Bacterial/toxicity , Bacillus anthracis/pathogenicity , Bacterial Toxins/toxicity , Macrophages/drug effects , Macrophages/microbiology , Receptors, Peptide/metabolism , Spores, Bacterial/drug effects , Animals , Anthrax/immunology , Anthrax/mortality , Antigens, Bacterial/immunology , Bacillus anthracis/physiology , Bacterial Toxins/immunology , Host-Parasite Interactions , Macrophages/physiology , Mice , Mice, Inbred BALB C , Receptors, Cell Surface/genetics , Receptors, Cell Surface/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL