Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 15 de 15
Filter
1.
Arch Toxicol ; 92(3): 1311-1322, 2018 Mar.
Article in English | MEDLINE | ID: mdl-29170806

ABSTRACT

Low molecular weight (LMW) polycyclic aromatic hydrocarbons (PAH) are the most abundant PAHs environmentally, occupationally, and are in cigarette smoke; however, little is known about their carcinogenic potential. We hypothesized that LMW PAHs act as co-carcinogens in the presence of a known carcinogen (benzo[a]pyrene (B[a]P)) in a mouse non-tumorigenic type II cell line (C10 cells). Gap junctions are commonly suppressed and inflammation induced during tumor promotion, while DNA-adduct formation is observed during the initiation stage of cancer. We used these endpoints together as markers of carcinogenicity in these lung adenocarcinoma progenitor cells. LMW PAHs (1-methylanthracene and fluoranthene, 1-10 µM total in a 1:1 ratio) were used based on previous studies as well as B[a]P (0-3 µM) as the classic carcinogen; non-cytotoxic doses were used. B[a]P-induced inhibition of gap junctional intercellular communication (GJIC) was observed at low doses and further reduced in the presence of the LMW PAH mixture (P < 0.05), supporting a role for GJIC suppression in cancer development. Benzo[a]pyrene diol-epoxide (BPDE)-DNA adduct levels were significantly induced in B[a]P-treated C10 cells and additionally increased with the LMW PAH mixture (P < 0.05). Significant increases in cyclooxygenase (Cox-2) were observed in response to the B[a]P/LMW PAH mixture combinations. DNA adduct formation coincided with the inhibition of GJIC and increase in Cox-2 mRNA expression. Significant cytochrome p4501b1 increases and connexin 43 decreases in gene expression were also observed. These studies suggest that LMW PAHs in combination with B[a]P can elicit increased carcinogenic potential. Future studies will further address the mechanisms of co-carcinogenesis driving these responses.


Subject(s)
Carcinogens/toxicity , Polycyclic Aromatic Hydrocarbons/toxicity , Pulmonary Alveoli/drug effects , Animals , Anthracenes/toxicity , Benzo(a)pyrene/toxicity , Cell Line , Connexin 43/genetics , Connexin 43/metabolism , Cyclooxygenase 2/genetics , Cytochrome P-450 CYP1B1/genetics , DNA Adducts , Epithelial Cells/drug effects , Fluorenes/toxicity , Gap Junctions/drug effects , Gap Junctions/pathology , Gene Expression Regulation/drug effects , Mice, Inbred BALB C , Polycyclic Aromatic Hydrocarbons/chemistry , Pulmonary Alveoli/cytology , Pulmonary Alveoli/pathology
2.
Mol Carcinog ; 56(1): 94-105, 2017 01.
Article in English | MEDLINE | ID: mdl-26894620

ABSTRACT

Adenocarcinoma accounts for ∼40% of lung cancer, equating to ∼88 500 new patients in 2015, most of who will succumb to this disease, thus, the public health burden is evident. Unfortunately, few early biomarkers as well as effective therapies exist, hence the need for novel targets in lung cancer treatment. We previously identified epiregulin (Ereg), an EGF-like ligand, as a biomarker in several mouse lung cancer models. In the present investigation we used a primary two-stage initiation/promotion model to test our hypothesis that Ereg deficiency would reduce lung tumor promotion in mice. We used 3-methylcholanthrene (initiator) or oil vehicle followed by multiple weekly exposures to butylated hydroxytoluene (BHT; promoter) in mice lacking Ereg (Ereg-/- ) and wildtype controls (BALB/ByJ; Ereg+/+ ) and examined multiple time points and endpoints (bronchoalveolar lavage analysis, tumor analysis, mRNA expression, ELISA, wound assay) during tumor promotion. At the early time points (4 and 12 wk), we observed significantly reduced amounts of inflammation (macrophages, PMNs) in the Ereg-/- mice compared to controls (Ereg+/+ ). At 20 wk, tumor multiplicity was also significantly decreased in the Ereg-/- mice versus controls (Ereg+/+ ). IL10 expression, an anti-inflammatory mediator, and downstream signaling events (Stat3) were significantly increased in the Ereg-/- mice in response to BHT, supporting both reduced inflammation and tumorigenesis. Lastly, wound healing was significantly increased with recombinant Ereg in both human and mouse lung epithelial cell lines. These results indicate that Ereg has proliferative potential and may be utilized as an early cancer biomarker as well as a novel potential therapeutic target. © 2016 Wiley Periodicals, Inc.


Subject(s)
Adenocarcinoma/genetics , Carcinogenesis/genetics , Epiregulin/genetics , Gene Expression Regulation, Neoplastic , Lung Neoplasms/genetics , Lung/pathology , Adenocarcinoma/chemically induced , Adenocarcinoma/pathology , Adenocarcinoma of Lung , Animals , Butylated Hydroxytoluene , Carcinogenesis/chemically induced , Carcinogenesis/pathology , Gene Deletion , Humans , Inflammation/chemically induced , Inflammation/genetics , Inflammation/pathology , Lung Neoplasms/chemically induced , Lung Neoplasms/pathology , Male , Mice , Mice, Inbred BALB C
3.
Exp Lung Res ; 42(3): 154-73, 2016 Apr.
Article in English | MEDLINE | ID: mdl-27093379

ABSTRACT

Tumor promotion is an early and critical stage during lung adenocarcinoma (ADC). We previously demonstrated that Tlr4 mutant mice were more susceptible to butylated hydroxytoluene (BHT)-induced pulmonary inflammation and tumor promotion in comparison to Tlr4-sufficient mice. Our study objective was to elucidate the underlying differences in Tlr4 mutant mice in innate immune cell populations, their functional responses, and the influence of these cellular differences on ADC progenitor (type II) cells following BHT-treatment. BALB (Tlr4-sufficient) and C.C3-Tlr4(Lps-d)/J (BALB(Lpsd); Tlr4 mutant) mice were treated with BHT (promoter) followed by bronchoalveolar lavage (BAL) and flow cytometry processing on the lungs. ELISAs, Club cell enrichment, macrophage function, and RNA isolation were also performed. Bone marrow-derived macrophages (BMDM) co-cultured with a type II cell line were used for wound healing assays. Innate immune cells significantly increased in whole lung in BHT-treated BALB(Lpsd) mice compared to BALB mice. BHT-treated BALB(Lpsd) mice demonstrated enhanced macrophage functionality, increased epithelial wound closure via BMDMs, and increased Club cell number in BALB(Lpsd) mice, all compared to BALB BHT-treated mice. Cytokine/chemokine (Kc, Mcp1) and growth factor (Igf1) levels also significantly differed among the strains and within macrophages, gene expression, and cell surface markers collectively demonstrated a more plastic phenotype in BALB(Lpsd) mice. Therefore, these correlative studies suggest that distinct innate immune cell populations are associated with the differences observed in the Tlr4-mutant model. Future studies will investigate the macrophage origins and the utility of the pathways identified herein as indicators of immune system deficiencies and lung tumorigenesis.


Subject(s)
Carcinogenesis/genetics , Carcinogenesis/immunology , Immunity, Innate/immunology , Lung Neoplasms/immunology , Lung Neoplasms/pathology , Toll-Like Receptor 4/immunology , Adenocarcinoma/genetics , Adenocarcinoma/immunology , Adenocarcinoma/pathology , Adenocarcinoma of Lung , Animals , Bronchoalveolar Lavage/methods , Butylated Hydroxytoluene/pharmacology , Carcinogenesis/pathology , Chemokines/immunology , Gene Expression/genetics , Gene Expression/immunology , Immunity, Innate/genetics , Lung/immunology , Lung/pathology , Lung Neoplasms/genetics , Macrophages/immunology , Macrophages/pathology , Male , Mice , Mice, Inbred BALB C , Mutation/genetics , Mutation/immunology , Pneumonia/chemically induced , Pneumonia/genetics , Pneumonia/immunology , Pneumonia/pathology , Toll-Like Receptor 4/genetics
4.
J Neurochem ; 123(6): 919-31, 2012 Dec.
Article in English | MEDLINE | ID: mdl-23020846

ABSTRACT

Glucagon-like peptide-1 (GLP-1)-based therapies are currently available for the treatment of type 2 diabetes, based on their actions on pancreatic ß cells. GLP-1 is also known to exert neuroprotective actions. To determine its mechanism of action, we developed a neuron-rich cell culture system by differentiating human neuroprogenitor cells in the presence of a combination of neurotrophins and retinoic acid. The neuronal nature of these cells was characterized by neurogenesis pathway-specific array. GLP-1 receptor expression was seen mainly in the neuronal population. Culture of neurons in the presence of Aß oligomers resulted in the induction of apoptosis as shown by the activation of caspase-3 and caspase-6. Exendin-4, a long-acting analog of GLP-1, protected the neurons from apoptosis induced by Aß oligomers. Exendin-4 stimulated cyclic AMP response element binding protein phosphorylation, a regulatory step in its activation. A transient transfection assay showed induction of a reporter linked to CRE site-containing human brain-derived neurotrophic factor promoter IV, by the growth factor through multiple signaling pathways. The anti-apoptotic action of exendin-4 was lost following down-regulation of cAMP response element binding protein. Withdrawal of neurotrophins resulted in the loss of neuronal phenotype of differentiated neuroprogenitor cells, which was prevented by incubation in the presence of exendin-4. Diabetes is a risk factor in the pathogenesis of Alzheimer's disease. Our findings suggest that GLP-1-based therapies can decrease the incidence of Alzheimer's disease among aging diabetic population.


Subject(s)
Cell Differentiation/physiology , Glucagon-Like Peptide 1/physiology , Neural Stem Cells/cytology , Neuroprotective Agents/pharmacology , Stem Cells/cytology , Brain-Derived Neurotrophic Factor/biosynthesis , Brain-Derived Neurotrophic Factor/genetics , Cells, Cultured , Cyclic AMP Response Element-Binding Protein/physiology , Gene Expression Regulation/physiology , Glucagon-Like Peptide 1/metabolism , Humans , Neural Pathways/physiology , Neural Stem Cells/metabolism , Promoter Regions, Genetic , Signal Transduction/genetics , Stem Cells/metabolism
5.
J Virol ; 85(13): 6678-86, 2011 Jul.
Article in English | MEDLINE | ID: mdl-21525352

ABSTRACT

Primary varicella-zoster virus (VZV) infection in humans produces varicella (chickenpox), after which the virus becomes latent in ganglionic neurons. Analysis of the physical state of viral nucleic acid and virus gene expression during latency requires postmortem acquisition of fresh human ganglia. To provide an additional way to study the VZV-host relationship in neurons, we developed an in vitro model of infected differentiated human neural stem cells (NSCs). NSCs were induced to differentiate in culture dishes coated with poly-l-lysine and mouse laminin in the presence of fibroblast growth factor 2 (FGF-2), nerve growth factor (NGF), brain-derived neurotropic factor (BDNF), dibutyryl cyclic AMP, and retinoic acid. Immunostaining with neuronal (MAP2a and ß-tubulin), astrocyte (GFAP), and oligodendrocyte (CNPase) markers revealed that differentiated neurons constituted approximately 90% of the cell population. These neurons were maintained in culture for up to 8 weeks. No cytopathic effect (CPE) developed in neurons infected with cell-free VZV (Zostavax vaccine) compared to human fetal lung fibroblasts infected with VZV. Weeks later, VZV DNA virus-specific transcripts (open reading frames [ORFs] 21, 29, 62, and 63) were detected in infected neurons, and dual immunofluorescence staining revealed the presence of VZV IE63 and gE exclusively in healthy-appearing neurons, but not in astrocytes. Neither the tissue culture medium nor a homogenate prepared from VZV-infected neurons produced a CPE in fibroblasts. VZV induced apoptosis in fibroblasts, as shown by activation of caspase 3 and by terminal deoxynucleotidyltransferase-mediated dUTP-biotin nick end labeling (TUNEL) staining, but not in neurons. This model provides a unique in vitro system to study the VZV-neuronal relationship.


Subject(s)
Cell Differentiation , Herpesvirus 3, Human/pathogenicity , Neural Stem Cells/cytology , Neural Stem Cells/virology , Neurons/virology , Animals , Apoptosis , Astrocytes/cytology , Astrocytes/virology , Cells, Cultured , Cytopathogenic Effect, Viral , Fibroblasts/virology , Fluorescent Antibody Technique , Herpesvirus 3, Human/genetics , Herpesvirus 3, Human/physiology , Humans , Lung/cytology , Lung/virology , Mice , Neurons/cytology , Viral Proteins/genetics , Viral Proteins/metabolism , Virology/methods
6.
Cancers (Basel) ; 11(4)2019 Apr 23.
Article in English | MEDLINE | ID: mdl-31018556

ABSTRACT

Polycyclic aromatic hydrocarbons (PAHs), prevalent contaminants in our environment, in many occupations, and in first and second-hand smoke, pose significant adverse health effects. Most research focused on the genotoxic high molecular weight PAHs (e.g., benzo[a]pyrene), however, the nongenotoxic low molecular weight (LMW) PAHs are emerging as potential co-carcinogens and tumor promoters known to dysregulate gap junctional intercellular communication (GJIC), activate mitogen activated protein kinase pathways, and induce the release of inflammatory mediators. We hypothesize that inflammatory mediators resulting from LMW PAH exposure in mouse lung epithelial cell lines are involved in the dysregulation of GJIC. We used mouse lung epithelial cell lines and an alveolar macrophage cell line in the presence of a binary PAH mixture (1:1 ratio of fluoranthene and 1-methylanthracene; PAH mixture). Parthenolide, a pan-inflammation inhibitor, reversed the PAH-induced inhibition of GJIC, the decreased CX43 expression, and the induction of KC and TNF. To further determine the direct role of a cytokine in regulating GJIC, recombinant TNF (rTNF) was used to inhibit GJIC and this response was further enhanced in the presence of the PAH mixture. Collectively, these findings support a role for inflammation in regulating GJIC and the potential to target these early stage cancer pathways for therapeutics.

7.
Environ Sci Process Impacts ; 21(8): 1342-1352, 2019 Aug 14.
Article in English | MEDLINE | ID: mdl-31049512

ABSTRACT

In this study, we investigated the airborne particles released during paper printing and paper shredding processes in an attempt to characterize and differentiate these particles. Particle characteristics were studied with real time instruments (RTIs) to measure concentrations and with samplers to collect particles for subsequent microscopy and cytotoxicity analysis. The particles released by paper shredding were evaluated for cytotoxicity by using in vitro human lung epithelial cell models. A substantial amount of particles were released during both the shredding and printing processes. We found that the printing process caused substantial release of particles with sizes of less than 300 nm in the form of metal granules and graphite. These released particles contained various elements including Al, Ca, Cu, Fe, Mg, N, K, P, S and Si. The particles released by the paper shredding processes were primarily nanoparticles and had a peak size between 27.4 nm and 36.5 nm. These paper particles contained elements including Al, Br Ca, Cl, Cr, Cu, Fe, Mg, N, Na, Ni P, S and Si, as determined by scanning electron microscope-energy dispersive X-ray spectroscopy (SEM-EDS) and single-particle inductively coupled plasma-mass spectroscopy (SP-ICP-MS) analysis. Although various metals were identified in the paper particles, these particles did not elicit cytotoxicity to simian virus-transformed bronchial epithelial cells (BEAS2B) and immortalized normal human bronchial epithelial cells (HBE1). However, future studies should investigate other cytotoxicity effects of these paper particles in various types of lung cells to identify potential health effects of the particles.


Subject(s)
Air Pollutants/analysis , Air Pollution, Indoor/analysis , Environmental Monitoring/methods , Paper , Particulate Matter/analysis , Printing , Graphite/analysis , Humans , Metals/analysis , Particle Size , Printing/instrumentation
8.
Toxicol Sci ; 169(1): 180-193, 2019 05 01.
Article in English | MEDLINE | ID: mdl-30690640

ABSTRACT

Low molecular weight polycyclic aromatic hydrocarbons (LMW PAHs; < 206.3 g/mol) are under regulated environmental contaminants (eg, secondhand smoke) that lead to gap junction dysregulation, p38 MAPK activation, and increased mRNA production of inflammatory mediators, such as cytokines and cyclooxygenase (COX2), in lung epithelial cells. However, the early mechanisms involving lipid signaling through the arachidonic acid pathway and subsequent eicosanoid production leading to these downstream events are not known. Common human exposures are to mixtures of LMW PAHs, thus C10 cells (a mouse lung epithelial cell line) were exposed to a representative binary PAH mixture, 1-methylanthracene (1-MeA) and fluoranthene (Flthn), for 30 min-24 h with and without p38 and cytosolic phospholipase A2 (cPLA2) inhibitors. Cytosolic phospholipase A2 inhibition reversed PAH-induced phospho-p38 MAPK activation and gap junction dysregulation at 30 min. A significant biphasic increase in cPLA2 protein was observed at 30 min, 2, and 4 h, as well as COX2 protein at 2 and 8 h. Untargeted metabolomics demonstrated a similar trend with significantly changing metabolites at 30 min and 4 h of exposure relative to 1 h; a "cPLA2-like" subset of metabolites within the biphasic response were predominately phospholipids. Targeted metabolomics showed several eicosanoids (eg, prostaglandin D2 (PGD2), PGE2α) were significantly increased at 4, 8, and 12 h following exposure to the binary PAH mixture and this effect was p38-dependent. Finally, PAH metabolism was not observed until after 8 h. These results indicate an early lipid signaling mechanism of LMW PAH toxicity in lung epithelial cells due to parent PAH compounds.


Subject(s)
Alveolar Epithelial Cells/drug effects , Anthracenes/toxicity , Eicosanoids/metabolism , Fluorenes/toxicity , Signal Transduction/drug effects , Alveolar Epithelial Cells/metabolism , Alveolar Epithelial Cells/pathology , Animals , Anthracenes/chemistry , Cell Line , Cyclooxygenase 2/metabolism , Enzyme Activation , Fluorenes/chemistry , Group IV Phospholipases A2/metabolism , Metabolomics , Mice, Inbred BALB C , Molecular Weight , Phosphorylation , Time Factors , Up-Regulation , p38 Mitogen-Activated Protein Kinases/metabolism
9.
JCI Insight ; 3(15)2018 08 09.
Article in English | MEDLINE | ID: mdl-30089720

ABSTRACT

With more than 150,000 deaths per year in the US alone, lung cancer has the highest number of deaths for any cancer. These poor outcomes reflect a lack of treatment for the most common form of lung cancer, non-small cell lung carcinoma (NSCLC). Lung adenocarcinoma (ADC) is the most prevalent subtype of NSCLC, with the main oncogenic drivers being KRAS and epidermal growth factor receptor (EGFR). Whereas EGFR blockade has led to some success in lung ADC, effective KRAS inhibition is lacking. KRAS-mutant ADCs are characterized by high levels of gel-forming mucin expression, with the highest mucin levels corresponding to worse prognoses. Despite these well-recognized associations, little is known about roles for individual gel-forming mucins in ADC development causatively. We hypothesized that MUC5AC/Muc5ac, a mucin gene known to be commonly expressed in NSCLC, is crucial in KRAS/Kras-driven lung ADC. We found that MUC5AC was a significant determinant of poor prognosis, especially in patients with KRAS-mutant tumors. In addition, by using mice with lung ADC induced chemically with urethane or transgenically by mutant-Kras expression, we observed significantly reduced tumor development in animals lacking Muc5ac compared with controls. Collectively, these results provide strong support for MUC5AC as a potential therapeutic target for lung ADC, a disease with few effective treatments.


Subject(s)
Adenocarcinoma of Lung/pathology , Carcinogenesis/pathology , Carcinoma, Non-Small-Cell Lung/pathology , Lung Neoplasms/pathology , Mucin 5AC/metabolism , Adenocarcinoma of Lung/genetics , Adenocarcinoma of Lung/mortality , Animals , Biomarkers, Tumor , Carcinogenesis/genetics , Carcinoma, Non-Small-Cell Lung/genetics , Carcinoma, Non-Small-Cell Lung/mortality , ErbB Receptors/genetics , Female , Humans , Lung/pathology , Lung Neoplasms/genetics , Lung Neoplasms/mortality , Male , Mice , Mice, Transgenic , Mucin 5AC/genetics , Mutation , Neoplasms, Experimental/genetics , Neoplasms, Experimental/pathology , Prognosis , Proto-Oncogene Proteins p21(ras)/genetics , Survival Analysis
10.
Toxicol Sci ; 157(1): 156-171, 2017 05 01.
Article in English | MEDLINE | ID: mdl-28329830

ABSTRACT

Low molecular weight polycyclic aromatic hydrocarbons (LMW PAHs; < 206.3 g/mol) are prevalent and ubiquitous environmental contaminants, presenting a human health concern, and have not been as thoroughly studied as the high MW PAHs. LMW PAHs exert their pulmonary effects, in part, through P38-dependent and -independent mechanisms involving cell-cell communication and the production of pro-inflammatory mediators known to contribute to lung disease. Specifically, we determined the effects of two representative LMW PAHs, 1-methylanthracene (1-MeA) and fluoranthene (Flthn), individually and as a binary PAH mixture on the dysregulation of gap junctional intercellular communication (GJIC) and connexin 43 (Cx43), activation of mitogen activated protein kinases (MAPK), and induction of inflammatory mediators in a mouse non-tumorigenic alveolar type II cell line (C10). Both 1-MeA, Flthn, and the binary PAH mixture of 1-MeA and Flthn dysregulated GJIC in a dose and time-dependent manner, reduced Cx43 protein, and activated the following MAPKs: P38, ERK1/2, and JNK. Inhibition of P38 MAPK prevented PAH-induced dysregulation of GJIC, whereas inhibiting ERK and JNK did not prevent these PAHs from dysregulating GJIC indicating a P38-dependent mechanism. A toxicogenomic approach revealed significant P38-dependent and -independent pathways involved in inflammation, steroid synthesis, metabolism, and oxidative responses. Genes in these pathways were significantly altered by the binary PAH mixture when compared with 1-MeA and Flthn alone suggesting interactive effects. Exposure to the binary PAH mixture induced the production and release of cytokines and metalloproteinases from the C10 cells. Our findings with a binary mixture of PAHs suggest that combinations of LMW PAHs may elicit synergistic or additive inflammatory responses which warrant further investigation and confirmation.


Subject(s)
Inflammation/metabolism , Lung/drug effects , Mitogens/metabolism , Polycyclic Aromatic Hydrocarbons/toxicity , Signal Transduction/drug effects , Tobacco Smoke Pollution , Animals , Cell Communication/drug effects , Cell Line , Connexin 43/metabolism , Dose-Response Relationship, Drug , Enzyme Activation , Epithelial Cells/drug effects , Epithelial Cells/metabolism , Gap Junctions/drug effects , Lung/metabolism , Lung/pathology , Mice , Mitogen-Activated Protein Kinases/metabolism , Transcriptome
11.
PLoS One ; 8(6): e65150, 2014.
Article in English | MEDLINE | ID: mdl-23755184

ABSTRACT

Polycyclic aromatic hydrocarbons (PAHs) are ubiquitous environmental and occupational toxicants, which are a major human health concern in the U.S. and abroad. Previous research has focused on the genotoxic events caused by high molecular weight PAHs, but not on non-genotoxic events elicited by low molecular weight PAHs. We used an isomeric pair of low molecular weight PAHs, namely 1-Methylanthracene (1-MeA) and 2-Methylanthracene (2-MeA), in which only 1-MeA possessed a bay-like region, and hypothesized that 1-MeA, but not 2-MeA, would affect non-genotoxic endpoints relevant to tumor promotion in murine C10 lung cells, a non-tumorigenic type II alveolar pneumocyte and progenitor cell type of lung adenocarcinoma. The non-genotoxic endpoints assessed were dysregulation of gap junction intercellular communication function and changes in the major pulmonary connexin protein, connexin 43, using fluorescent redistribution and immunoblots, activation of mitogen activated protein kinases (MAPK) using phosphospecific MAPK antibodies for immunoblots, and induction of inflammatory genes using quantitative RT-PCR. 2-MeA had no effect on any of the endpoints, but 1-MeA dysregulated gap junctional communication in a dose and time dependent manner, reduced connexin 43 protein expression, and altered membrane localization. 1-MeA also activated ERK1/2 and p38 MAP kinases. Inflammatory genes, such as cyclooxygenase 2, and chemokine ligand 2 (macrophage chemoattractant 2), were also upregulated in response to 1-MeA only. These results indicate a possible structure-activity relationship of these low molecular weight PAHs relevant to non-genotoxic endpoints of the promoting aspects of cancer. Therefore, our novel findings may improve the ability to predict outcomes for future studies with additional toxicants and mixtures, identify novel targets for biomarkers and chemotherapeutics, and have possible implications for future risk assessment for these PAHs.


Subject(s)
Carcinogenesis/pathology , Inflammation/pathology , Lung/pathology , Polycyclic Aromatic Hydrocarbons/chemistry , Polycyclic Aromatic Hydrocarbons/toxicity , Signal Transduction/drug effects , Animals , Anthracenes/chemistry , Anthracenes/toxicity , Carcinogenesis/metabolism , Cell Communication/drug effects , Cell Death/drug effects , Cell Line, Tumor , Connexin 43/metabolism , Enzyme Activation/drug effects , Gap Junctions/metabolism , Humans , Mice , Mitogen-Activated Protein Kinases/metabolism , Molecular Weight , Phosphorylation/drug effects , Protein Kinase Inhibitors/pharmacology , Time Factors
12.
J Mol Biomark Diagn ; 5(1)2013 Dec 27.
Article in English | MEDLINE | ID: mdl-25035812

ABSTRACT

TLR4 protects against lung tumor promotion and pulmonary inflammation in mice. Connexin 43 (Cx43), a gap junction gene, was increased in Tlr4 wildtype compared to Tlr4-mutant mice in response to promotion, which suggests gap junctional intercellular communication (GJIC) may be compromised. We hypothesized that the early tumor microenvironment, represented by Bronchoalveolar Lavage Fluid (BALF) from Butylated hydroxytoluene (BHT; promoter)-treated mice, would produce TLR4-dependent changes in pulmonary epithelium, including dysregulation of GJIC in the Tlr4-mutant (BALB Lps-d ) compared to the Tlr4-sufficient (BALB; wildtype) mice. BHT (4 weekly doses) was injected ip followed by BALF collection at 24 h. BALF total protein and total macrophages were significantly elevated in BHT-treated BALB Lps-d over BALB mice, similar to previous findings. BALF was then utilized in an ex vivo manner to treat C10 cells, a murine alveolar type II cell line, followed by the scrape-load dye transfer assay (GJIC), Cx43 immunostaining, and quantitative RT-PCR (Mcp-1, monocyte chemotactic protein 1). GJIC was markedly reduced in C10 cells treated with BHT-treated BALB Lps-d BALF for 4 and 24 h compared to BALB and control BALF from the respective mice (p < 0.05). Mcp-1, a chemokine, was also significantly increased in the BHT-treated BALB Lps-d BALF compared to the BALB mice, and Cx43 protein expression in the cell membrane altered. These novel findings suggest signaling from the BALF milieu is involved in GJIC dysregulation associated with promotion and links gap junctions to pulmonary TLR4 protection in a novel ex vivo model that could assist in future potential tumor promoter screening.

13.
Endocrinology ; 153(3): 1116-28, 2012 Mar.
Article in English | MEDLINE | ID: mdl-22253425

ABSTRACT

Islets isolated from cadaveric donor pancreas are functionally viable and can be transplanted in diabetic patients to reduce insulin requirements. This therapeutic approach is less efficient because a significant portion of functional islets is lost due to oxidative stress, inflammation, and hypoxia. Exendin-4, a glucagon-like peptide-1 receptor agonist, is known to improve islet survival through activation of the transcription factor, cAMP response element binding protein (CREB). However, isolated human islets are exposed to several stresses known to down-regulate CREB. The objective of the present study was to determine whether the cytoprotective actions of exendin-4 in human islets can be augmented by increasing the levels of CREB. Simulation of ischemia/reperfusion injury and exposure to hypoxic conditions in cultured human islets resulted in decreased CREB activation and induction of apoptosis. Islets were transduced with adenoviral CREB followed by exposure to exendin-4 as a strategy for improving their survival. This combination increased the levels of several proteins needed for ß-cell survival and function, including insulin receptor substrate-2, Bcl-2, and baculoviral IAP repeat-containing 3, and suppressed the expression of proapoptotic and inflammatory genes. A combination of CREB and exendin-4 exerted enhanced antiapoptotic action in cultured islets against hypoxia and cytokines. More significantly, transplantation of human islets transduced with adenoviral CREB and treated with exendin-4 showed improved glycemic control over a 30-d period in diabetic athymic nude mice. These observations have significant implications in the therapeutic potential of exendin-4 and CREB in the islet transplantation setting as well as in preserving ß-cell mass of diabetic patients.


Subject(s)
Apoptosis , Cyclic AMP Response Element-Binding Protein/metabolism , Cytokines/metabolism , Hypoxia , Peptides/pharmacology , Venoms/pharmacology , Animals , Cell Line , Cells, Cultured , Computer Simulation , Exenatide , Humans , Insulin Receptor Substrate Proteins/metabolism , Insulin-Secreting Cells/cytology , Islets of Langerhans/cytology , Male , Mice , Mice, Nude , Pancreas/metabolism , Reperfusion Injury/pathology , Signal Transduction
14.
Free Radic Biol Med ; 51(6): 1146-54, 2011 Sep 15.
Article in English | MEDLINE | ID: mdl-21712085

ABSTRACT

Hydroxychalcones are naturally occurring compounds that continue to attract considerable interest because of their anti-inflammatory and antiangiogenic properties. They have been reported to inhibit the synthesis of the inducible nitric oxide synthase and to induce the expression of heme oxygenase-1. This study examines the mechanisms by which 2',5'-dihydroxychalcone (2',5'-DHC) induces an increase in cellular glutathione (GSH) levels using a cell line stably expressing a luciferase reporter gene driven by antioxidant-response elements (MCF-7/AREc32). The 2',5'-DHC-induced increase in cellular GSH levels was partially inhibited by the catalytic antioxidant MnTDE-1,3-IP(5+), suggesting that reactive oxygen species (ROS) mediate the antioxidant adaptive response. 2',5'-DHC treatment induced phosphorylation of the c-Jun N-terminal kinase (JNK) pathway, which was also inhibited by MnTDE-1,3-IP(5+). These findings suggest a ROS-dependent activation of the AP-1 transcriptional response. However, whereas 2',5'-DHC triggered the NF-E2-related factor 2 (Nrf2) transcriptional response, cotreatment with MnTDE-1,3-IP(5+) did not decrease 2',5'-DHC-induced Nrf2/ARE activity, showing that this pathway is not dependent on ROS. Moreover, pharmacological inhibitors of mitogen-activated protein kinase (MAPK) pathways showed a role for JNK and p38MAPK in mediating the 2',5'-DHC-induced Nrf2 response. These findings suggest that the 2',5'-DHC-induced increase in GSH levels results from a combination of ROS-dependent and ROS-independent pathways.


Subject(s)
Antioxidants/pharmacology , Chalcone/pharmacology , Glutathione/metabolism , JNK Mitogen-Activated Protein Kinases/metabolism , NF-E2-Related Factor 2/metabolism , Cell Line, Tumor , Chalcone/analogs & derivatives , Chalcone/chemistry , Humans , Metalloporphyrins/pharmacology , Oxidative Stress/drug effects , Reactive Oxygen Species/metabolism , Response Elements/genetics , Signal Transduction/drug effects , Transcription Factor AP-1/metabolism , Transcriptional Activation/drug effects
15.
Free Radic Biol Med ; 46(3): 430-40, 2009 Feb 01.
Article in English | MEDLINE | ID: mdl-19056485

ABSTRACT

Protandim is an antioxidant supplement that consists of five ingredients, namely, ashwagandha, bacopa extract, green tea extract, silymarin, and curcumin, each with known therapeutic properties. Protandim was formulated with the objective of combining multiple phytochemicals at low nontoxic doses to gain synergy among them. A recent clinical study demonstrated the in vivo antioxidant effects of Protandim (S.K. Nelson et al., 2006, Free Radic. Biol. Med. 40, 341-347). The objective of the present study was to determine if the components of Protandim induce heme oxygenase-1 (HO-1) in a synergistic manner in cultured MIN6 cells, a mouse beta-cell line, and in SK-N-MC cells, a human neuroblastoma cell line. When the components of Protandim were tested alone at low doses, curcumin showed minimal induction, whereas the others were unable to induce the HO-1 promoter, assayed by transient transfection. All components together, however, produced a strongly synergistic induction of around three- to ninefold in a dose-dependent manner, greatly exceeding the sum of the parts. Similar findings were obtained for the expression of HO-1 at the mRNA and protein levels. Protandim-mediated HO-1 induction involved the presence of ARE sites in the HO-1 promoter and nuclear translocalization of the transcription factor Nrf2, which binds to ARE sites. The involvement of multiple signaling pathways, including PI3-kinase/Akt, p38MAPK, and PKCdelta, in HO-1 induction seems to be the probable mechanism of synergy between the components of Protandim. There were significant increases in the levels of total glutathione in Protandim-treated cells. These findings suggest that the use of a combination of phytochemicals may be an efficient method for the induction of antioxidant enzymes.


Subject(s)
Antioxidants/pharmacology , Enzyme Induction/drug effects , Heme Oxygenase-1/metabolism , NF-E2-Related Factor 2/metabolism , Active Transport, Cell Nucleus/drug effects , Animals , Antioxidants/chemistry , Cell Line , Cell Nucleus/metabolism , Curcumin/pharmacology , Dose-Response Relationship, Drug , Drug Synergism , Drugs, Chinese Herbal/chemistry , Drugs, Chinese Herbal/pharmacology , Glutathione/genetics , Glutathione/metabolism , Heme Oxygenase-1/genetics , Humans , Insulin-Secreting Cells/drug effects , Insulin-Secreting Cells/enzymology , Mice , NF-E2-Related Factor 2/genetics , Pharmaceutical Preparations/chemistry , Phosphatidylinositol 3-Kinases/metabolism , Promoter Regions, Genetic , Protein Kinase C-delta/metabolism , Response Elements/drug effects , Signal Transduction , p38 Mitogen-Activated Protein Kinases/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL