Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 6 de 6
Filter
Add more filters










Database
Language
Publication year range
1.
Int J Cancer ; 151(12): 2195-2205, 2022 Dec 15.
Article in English | MEDLINE | ID: mdl-35830207

ABSTRACT

The N-nitroso-trischloroethylurea (NTCU)-induced mouse model of squamous lung carcinoma recapitulates human disease from premalignant dysplasia through invasive tumors, making it suitable for preclinical chemoprevention drug testing. Pioglitazone is a peroxisome proliferator-activated receptor γ (PPARγ) agonist shown to prevent lung tumors in preclinical models. We investigated pioglitazone's effect on lesion development and markers of potential preventive mechanisms in the NTCU model. Female FVB/N mice were exposed to vehicle, NTCU or NTCU + oral pioglitazone for 32 weeks. NTCU induces the appearance of basal cells in murine airways while decreasing/changing their epithelial cell makeup, resulting in development of bronchial dysplasia. H&E and keratin 5 (KRT5) staining were used to detect and grade squamous lesions in formalin fixed lungs. mRNA expression of epithelial to mesenchymal transition (EMT) markers and basal cell markers were measured by qPCR. Dysplasia persistence markers desmoglein 3 and polo like kinase 1 were measured by immunohistochemistry. Basal cell markers KRT14 and p63, club cell specific protein and ciliated cell marker acetylated tubulin were measured by immunofluorescence. Pioglitazone treatment significantly reduced squamous lesions and the presence of airway basal cells, along with increasing normal epithelial cells in the airways of NTCU-exposed mice. Pioglitazone also significantly influenced EMT gene expression to promote a more epithelial, and less mesenchymal, phenotype. Pioglitazone reduced the presence of squamous dysplasia and maintained normal airway cell composition. This work increases the knowledge of mechanistic pathways in PPARγ agonism for lung cancer interception and provides a basis for further investigation to advance this chemoprevention strategy.


Subject(s)
Carcinoma, Non-Small-Cell Lung , Carcinoma, Squamous Cell , Lung Neoplasms , Mice , Female , Humans , Animals , PPAR gamma , Keratin-5 , Epithelial-Mesenchymal Transition , Pioglitazone/adverse effects , Tubulin , Desmoglein 3 , Carcinoma, Squamous Cell/pathology , Lung Neoplasms/drug therapy , Lung Neoplasms/prevention & control , Lung Neoplasms/chemically induced , Lung/pathology , Formaldehyde/adverse effects , RNA, Messenger
2.
Cancer Prev Res (Phila) ; 5(2): 283-9, 2012 Feb.
Article in English | MEDLINE | ID: mdl-22086679

ABSTRACT

Squamous cell carcinoma (SCC) and premalignant endobronchial lesions have been difficult to study in murine models. In this study, we evaluate the topical N-nitroso-tris-chloroethylurea (NTCU) murine SCC model, determine the extent to which resulting premalignant airway dysplasia develops, discuss clinicopathologic grading criteria in lesion progression, and confirm that immunohistochemical (IHC) staining patterns are consistent with those observed in human endobronchial dysplasia and SCC. Male and female FVB mice were treated biweekly with topical NTCU (4, 8, or 40 mmol/L) or vehicle for 32 weeks. Following sacrifice, squamous cell lesions were enumerated and categorized into the following groups: flat atypia, low-grade dysplasia, high-grade dysplasia, and invasive SCC. The 40 mmol/L NTCU concentration produced the entire spectrum of premalignant dysplasias and squamous cell carcinomas, but was associated with poor survival. Concentrations of 4 and 8 mmol/L NTCU were better tolerated and produced only significant levels of flat atypia. Squamous origin of the range of observed lesions was confirmed with IHC staining for cytokeratin 5/6, p63, thyroid transcription factor-1 (TTF-1), and Napsin-A. This study shows that topical application of high-dose NTCU produces endobronchial premalignant lesions with classic squamous characteristics and should allow for improved preclinical evaluation of potential chemopreventive agents.


Subject(s)
Bronchial Neoplasms/chemically induced , Carcinogens/toxicity , Carcinoma, Squamous Cell/chemically induced , Carmustine/analogs & derivatives , Disease Models, Animal , Precancerous Conditions/chemically induced , Administration, Topical , Animals , Bronchial Neoplasms/mortality , Bronchial Neoplasms/pathology , Carcinoma, Squamous Cell/mortality , Carcinoma, Squamous Cell/pathology , Carmustine/toxicity , Female , Immunoenzyme Techniques , Male , Mice , Mice, Inbred BALB C , Neoplasm Invasiveness , Precancerous Conditions/mortality , Precancerous Conditions/pathology , Survival Rate
3.
Lung Cancer ; 70(1): 37-42, 2010 Oct.
Article in English | MEDLINE | ID: mdl-20116128

ABSTRACT

INTRODUCTION: We hypothesized that the combination of the EGFR tyrosine kinase inhibitor (TKI) gefitinib with the powerful chemopreventive manipulation of lung-specific transgenic prostacyclin synthase (PGIS) overexpression on tumorigenesis in FVB/N mice would result in augmented chemoprevention. MATERIALS AND METHODS: Wildtype and littermate PGIS overexpressors (OE) were given urethane, 1 mg/kg i.p. followed by thrice weekly i.p. injections of gefitinib, 50 mg/kg or 100 mg/kg, or vehicle. Pulmonary adenomas were enumerated and measured. RESULTS: Gefitinib at either 50 mg/kg or 100 mg/kg administered i.p. three times weekly was effective in inhibiting EGF induced EGFR tyrosine phosphorylation and downstream signaling. The PGIS overexpressors showed significant decreases in tumor multiplicity consistent with prior studies. Gefitinib had no effect on tumor multiplicity or volume in wildtype mice. Among the PGIS overexpressors, a significant reduction in tumor multiplicity was shown in the 50 mg/kg, but not the 100 mg/kg, gefitinib treatment group vs. vehicle control animals (1.13+/-0.29 vs. 2.29+/-0.32 tumors/mouse, p=0.015). We examined the phosphorylation status in selected downstream effectors of EGFR (Erk, Akt, Src, PTEN). The major difference in the 50 mg/kg vs. 100 mg/kg group was an increase in p-Src in the PGIS OE mice receiving the higher dose. CONCLUSION: We conclude that gefitinib alone has no chemopreventive efficacy in this model; it augmented the effect of PGIS overexpression at 50 mg/kg but not 100 mg/kg. Increased p-Src is correlated with loss of efficacy at the higher dose, suggesting the potential for combined EGFR and Src inhibition strategies in chemoprevention.


Subject(s)
Cytochrome P-450 Enzyme System/biosynthesis , Intramolecular Oxidoreductases/biosynthesis , Lung Neoplasms/prevention & control , Protein Kinase Inhibitors/pharmacology , Quinazolines/pharmacology , Adenoma/enzymology , Adenoma/genetics , Adenoma/metabolism , Adenoma/prevention & control , Animals , Antineoplastic Agents/pharmacology , Cadherins/metabolism , Cytochrome P-450 Enzyme System/genetics , Dinoprostone/metabolism , Epoprostenol/metabolism , ErbB Receptors/antagonists & inhibitors , ErbB Receptors/metabolism , Female , Gefitinib , Intramolecular Oxidoreductases/genetics , Lung Neoplasms/enzymology , Lung Neoplasms/genetics , Lung Neoplasms/metabolism , Male , Mice , Mice, Transgenic , Rats , Signal Transduction/drug effects
4.
Cancer Prev Res (Phila) ; 1(5): 349-56, 2008 Oct.
Article in English | MEDLINE | ID: mdl-19138979

ABSTRACT

Overexpression of prostacyclin synthase (PGIS) decreases lung tumor multiplicity in chemical- and cigarette-smoke-induced murine lung cancer models. Prostacyclin signals through a single G-protein-coupled receptor (IP), which signals through cyclic AMP. To determine the role of this receptor in lung cancer chemoprevention by prostacyclin, PGIS-overexpressing mice were crossed to mice that lack the IP receptor [IP(-/-)]. Carcinogen-induced lung tumor incidence was similar in IP(+/+), IP(+/-), and IP(-/-) mice, and overexpression of PGIS gave equal protection in all three groups, indicating that the protective effects of prostacyclin are not mediated through activation of IP. Because prostacyclin can activate members of the peroxisomal proliferator-activated receptor (PPAR) family of nuclear receptors, we examined the role of PPARgamma in the protection of prostacyclin against lung tumorigenesis. Iloprost, a stable prostacyclin analogue, activated PPARgamma in nontransformed bronchial epithelial cells and in a subset of human non-small-cell lung cancer cell lines. Iloprost-impregnated chow fed to wild-type mice resulted in elevated lung macrophages and decreased lung tumor formation. Transgenic animals with lung-specific PPARgamma overexpression also developed fewer lung tumors. This reduction was not enhanced by administration of supplemental iloprost. These studies indicate that PPARgamma is a critical target for prostacyclin-mediated lung cancer chemoprevention and may also have therapeutic activity.


Subject(s)
Carcinoma, Non-Small-Cell Lung/prevention & control , Epoprostenol/therapeutic use , Lung Neoplasms/prevention & control , PPAR gamma/agonists , Receptors, Epoprostenol/physiology , Animals , Carcinoma, Non-Small-Cell Lung/genetics , Cytochrome P-450 Enzyme System/genetics , Drug Evaluation, Preclinical , Epoprostenol/analogs & derivatives , Epoprostenol/pharmacology , Genotype , Humans , Iloprost/pharmacology , Iloprost/therapeutic use , Intramolecular Oxidoreductases/genetics , Lung Neoplasms/genetics , Mice , Mice, Inbred C57BL , Mice, Transgenic , PPAR gamma/genetics , PPAR gamma/metabolism , PPAR gamma/physiology , Rats , Receptors, Epoprostenol/genetics , Receptors, Epoprostenol/metabolism , Respiratory Mucosa/drug effects , Respiratory Mucosa/metabolism , Tumor Cells, Cultured
5.
Anticancer Res ; 26(4B): 2857-61, 2006.
Article in English | MEDLINE | ID: mdl-16886605

ABSTRACT

BACKGROUND: Manipulating prostaglandin (PG) production modulates tumor development. Elevated PGI2 production prevents murine lung cancer, while decreasing PGE2 content protects against colon cancer. PGE2 receptor subtype 2 (EP2) -deficient mice were hypothesized to be resistant to lung tumorigenesis. MATERIALS AND METHODS: EP2 null BALB/c mice and their wild-type littermates were exposed to an initiation-promotion carcinogenesis protocol and lung tumorigenesis was examined. Chronic lung inflammation was induced to determine whether EP2 ablation influenced inflammatory cell infiltration. RESULTS: Tumor multiplicity in EP2 null mice was 34% lower than in their wild-type littermates (21.9+/-3.0 vs. 14.5+/-2.9 tumors/mouse, p<0.001). The lung tumor burden, an indicator of growth rate, also declined (57%, p<0.05). All the mice exhibited similar inflammatory cell infiltration. CONCLUSION: PGE2, acting through EP2, enhanced lung tumorigenesis through a mechanism that may be distinct from its proinflammatory activity. Thus, EP2 is a potential target for novel chemoprevention strategies.


Subject(s)
Lung Neoplasms/prevention & control , Receptors, Prostaglandin E/deficiency , Animals , Butylated Hydroxytoluene , Cell Growth Processes/physiology , Female , Leukocytes/immunology , Lung Neoplasms/chemically induced , Lung Neoplasms/metabolism , Lung Neoplasms/pathology , Male , Methylcholanthrene , Mice , Mice, Inbred BALB C , Mice, Knockout , Receptors, Prostaglandin E/genetics , Receptors, Prostaglandin E, EP2 Subtype
6.
Cancer Res ; 64(16): 5897-904, 2004 Aug 15.
Article in English | MEDLINE | ID: mdl-15313935

ABSTRACT

Increased pulmonary production of prostaglandin I2 (prostacyclin) by lung-specific overexpression of prostacyclin synthase decreases lung tumor incidence and multiplicity in chemically induced murine lung cancer models. We hypothesized that pulmonary prostacyclin synthase overexpression would prevent lung carcinogenesis in tobacco-smoke exposed mice. Murine exposure to tobacco smoke is an established model of inducing pulmonary adenocarcinomas and allows for the testing of potential chemopreventive strategies. Transgenic FVB/N mice with lung-specific prostacyclin synthase overexpression were exposed to mainstream cigarette smoke for 22 weeks and then held unexposed for an additional 20 weeks. All of the exposed animals developed bronchiolitis analogous to the respiratory bronchiolitis seen in human smokers. The transgenic mice, when compared with smoke-exposed transgene negative littermates, had significant decreases in tumor incidence and multiplicity. Significantly fewer transgenics (6 of 15; 40%) developed tumors compared with the tumor incidence in wild-type littermates (16 of 19; 84%; Fisher's exact test, P = 0.012). Tumor multiplicity was also significantly decreased in the transgenic animals (tg+ = 0.4 +/- 0.5 versus wild-type = 1.2 +/- 0.86 tumors/mouse; P < 0.001). Targeted prostaglandin levels at the time of sacrifice revealed significantly elevated prostaglandin I2 levels in the transgenic animals, coupled with significantly decreased prostaglandin E2 levels. Gene expression analysis of isolated type II pneumocytes suggests potential explanations for the observed chemoprevention, with Western blot analysis confirming decreased expression of cytochrome p450 2e1. These studies extend our previous studies and demonstrate that manipulation of prostaglandin production distal to cyclooxygenase significantly reduces lung carcinogenesis in a tobacco smoke exposure model, and gene expression studies show critical alterations in antioxidation, immune response, and cytokine pathways.


Subject(s)
Cytochrome P-450 Enzyme System/biosynthesis , Intramolecular Oxidoreductases/biosynthesis , Lung Neoplasms/enzymology , Lung Neoplasms/prevention & control , Lung/enzymology , Smoke/adverse effects , Animals , Bronchiolitis/etiology , Dinoprostone/biosynthesis , Epoprostenol/biosynthesis , Humans , Lung Neoplasms/etiology , Mice , Mice, Transgenic , Oligonucleotide Array Sequence Analysis , Pulmonary Alveoli/metabolism , Rats , Weight Gain
SELECTION OF CITATIONS
SEARCH DETAIL