Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 35
Filter
1.
Toxicol Pathol ; 49(4): 912-927, 2021 06.
Article in English | MEDLINE | ID: mdl-32840183

ABSTRACT

Quantitative assessment of proliferation can be an important endpoint in toxicologic pathology. Traditionally, cell proliferation is quantified by labor-intensive manual counting of positive and negative cells after immunohistochemical staining for proliferation markers (eg, Ki67, bromo-2'-deoxyuridine, or proliferating cell nuclear antigen). Currently, there is a lot of interest in replacing manual evaluation of histology end points with image analysis tools based on artificial intelligence. The aim of the present study was to explore if a commercially available image analysis software can be used to quantify epithelial proliferative activity in rat mammary gland and minipig oviduct. First, algorithms based on artificial intelligence were trained to detect epithelium in each tissue. Areas of BrdU- or Ki67-positive nuclei and negative nuclei were subsequently quantified with threshold analysis. Artificial intelligence-based and manually counted labelling indices were strongly correlated and equally well detected the estrous cycle influence on proliferation in mammary gland and oviduct epithelium, as well as the dramatically increased proliferation in rat mammary glands after treatment with estradiol and progesterone. In conclusion, quantification of epithelial proliferation in two reproductive tissues can be achieved in a reliable fashion using image analysis software based on artificial intelligence, thus avoiding time- and labor-intensive manual counting, requiring trained operators.


Subject(s)
Artificial Intelligence , Epithelial Cells , Mammary Glands, Animal , Oviducts , Animals , Cell Proliferation , Female , Mammary Glands, Animal/cytology , Mammary Glands, Animal/growth & development , Oviducts/drug effects , Oviducts/growth & development , Rats , Swine , Swine, Miniature
2.
Am J Disaster Med ; 10(2): 109-20, 2015.
Article in English | MEDLINE | ID: mdl-26312493

ABSTRACT

OBJECTIVE: New smallpox medical countermeasures are entering the marketplace, offering the opportunity to modernize existing stockpiles. However, new smallpox countermeasures are developed under the animal rule, meaning that human efficacy data are lacking, and human safety data may be limited. Also, stockpile modernization would require prioritization of increasingly limited public funds. Approaches to address these issues are needed. METHODS: Smallpox vaccine data were gathered by literature search. The financial value of vaccination in the face of an outbreak was evaluated using a threatbased cost/benefit analysis model, involving i) estimation of the efficacy of new smallpox vaccines based on available clinical data on virus-neutralizing seroconversion in vaccinees, ii) estimation of the likelihood for a smallpox outbreak in Denmark, and iii) estimation of the expected life-saving effects of postevent vaccination. RESULTS: The authors estimated that i) the likelihood of a smallpox outbreak in Denmark is very low (one event in 200,000 years), ii) the expected efficacy of currently available and new vaccines is 95 and 75 percent, respectively, iii) the expected frequency of serious side effects from vaccination is between 100 and 10,000 fold lower for new than for existing vaccines, depending on modes of action. CONCLUSIONS: Despite the very low likelihood for a smallpox outbreak, the potentially large consequences combined with the protective effect of vaccination make maintenance of the smallpox vaccine stockpile justified and valuable. For vaccination in the face of a smallpox outbreak, a high efficacy rather than a lowered rate of adverse effects would maximize the number of lives saved.


Subject(s)
Disease Outbreaks/prevention & control , Smallpox Vaccine/supply & distribution , Smallpox/prevention & control , Strategic Stockpile , Bioterrorism , Disaster Medicine , Humans , Risk Assessment
4.
J Appl Toxicol ; 35(7): 842-50, 2015 Jul.
Article in English | MEDLINE | ID: mdl-25413577

ABSTRACT

There is a medical need for new insulin analogues. Yet, molecular alterations to the insulin molecule can theoretically result in analogues with carcinogenic effects. Preclinical carcinogenicity risk assessment for insulin analogues rests to a large extent on mitogenicity assays in cell lines. We therefore optimized mitogenicity assay conditions for a panel of five cell lines. All cell lines expressed insulin receptors (IR), IGF-I receptors (IGF-IR) and hybrid receptors, and in all cell lines, insulin as well as the comparator compounds X10 and IGF-I caused phosphorylation of the IR as well as IGF-IR. Insulin exhibited mitogenicity EC(50) values in the single-digit nanomolar to picomolar range. We observed correlations across cell types between (i) mitogenic potency of insulin and IGF-IR/IR ratio, (ii) Akt phosphorylation and mitogenic potency and (iii) Akt phosphorylation and IR phosphorylation. Using siRNA-mediated knockdown of IR and IGF-IR, we observed that in HCT 116 cells the IR appeared dominant in driving the mitogenic response to insulin, whereas in MCF7 cells the IGF-IR appeared dominant in driving the mitogenic response to insulin. Together, our results show that the IR as well as IGF-IR may contribute to the mitogenic potency of insulin. While insulin was a more potent mitogen than IGF-I in cells expressing more IR than IGF-IR, the hyper-mitogenic insulin analogue X10 was a more potent mitogen than insulin across all cell types, supporting that the hyper-mitogenic effect of X10 involves the IR as well as the IGF-IR. These results are relevant for preclinical safety assessment of developmental insulin analogues.


Subject(s)
Insulin/pharmacology , Mitosis/drug effects , Receptor, IGF Type 1/metabolism , Receptor, Insulin/metabolism , Animals , Blotting, Western , Cell Line , Cell Line, Tumor , Cell Proliferation/drug effects , Enzyme-Linked Immunosorbent Assay , HCT116 Cells/drug effects , Hep G2 Cells/drug effects , Humans , Insulin/analogs & derivatives , MCF-7 Cells/drug effects , Rats
5.
Vet Microbiol ; 165(1-2): 21-8, 2013 Jul 26.
Article in English | MEDLINE | ID: mdl-23528651

ABSTRACT

Porcine reproductive and respiratory syndrome virus (PRRSV) is a major threat to European swine production. The existence of extensive genetic variation in endemic strains and the presence of highly virulent strains in other geographic regions pose the threat of devastating epidemic outbreaks. Here we describe the current knowledge of genetic variation in European PRRSV isolates, the implications for PRRSV evolution, and the presence of multiple genetic lineages of Type 2 (North American genotype) isolates in Europe. In Type 1 (European genotype) PRRSV, three genetic subtypes are recognized and a fourth subtype appears to be present. Type 2 PRRSV was considered to be genetically homogenous in Europe due to a unique presence of an introduced vaccine strain, but independent introductions of virulent Type 2 field viruses are now evident. In Type 1 PRRSV, only subtype 1 (Lelystad virus-like) circulates in Central and Western Europe and globally. In Eastern Europe, all subtypes are present. The subtypes of Type 1 PRRSV also exhibit length differences in the nucleocapsid protein, ranging in size from 124 to 132 amino acids depending on subtype. This size heterogeneity is unparalleled in the nucleocapsid proteins of Type 2 PRRSV or other viruses. Surprisingly, it affects the C-terminus, otherwise thought to be under strong structural constraints. Finally, divergent subtypes of Type 1 PRRSV have produced high rates of false-negative RT-PCR results in diagnostic tests, and may also degrade the reliability of serodiagnostic assays using the nucleocapsid protein antigen. In summary, the extensive genetic diversity of Type 1 PRRSV is of relevance for understanding nucleocapsid protein structure/function relationships. Further, the extensive genetic diversity of Type 1 PRRSV in Europe, and the presence of diverse Type 2 PRRSV strains, together emphasize the importance of relevant validation of PRRSV diagnostics. More extensive and systematic molecular phylogeny studies are needed to fully understand PRRSV diversity in Europe, to provide swine producers with reliable diagnostics, and to better assess the potential consequences of endemic spread and exotic introductions.


Subject(s)
Porcine Reproductive and Respiratory Syndrome/virology , Porcine respiratory and reproductive syndrome virus/isolation & purification , Animals , Europe/epidemiology , Evolution, Molecular , Genetic Variation , Nucleocapsid Proteins/genetics , Phylogeny , Porcine Reproductive and Respiratory Syndrome/epidemiology , Porcine respiratory and reproductive syndrome virus/classification , Porcine respiratory and reproductive syndrome virus/genetics , Swine
6.
Arch Biochem Biophys ; 526(2): 124-31, 2012 Oct 15.
Article in English | MEDLINE | ID: mdl-22705202

ABSTRACT

Today's medicine has to deal with the emergence of multi-drug resistant bacteria, and is beginning to be confronted with pan-resistant microbes. This worsening inadequacy of the antibiotics concept, which has ruled infectious medicine in the last six decades creates an increasing unmet medical need that can be addressed by passive immunization. While past experience from the pre-antibiotic era with serum therapy was in many cases encouraging, antibacterial monoclonal antibodies have so far suffered high attrition rates in the clinic, generally from lack of efficacy. Yet, we believe that recent developments in a number of areas such as infectious disease pathogenesis research, translational medicine, mAb engineering, mAb manufacturing and rapid bedside diagnostics are converging to make the medium-term future permissive for antibacterial mAb development. Here, we review antibacterial mAb-based approaches that are or were in clinical development, and may potentially act as paradigms with regards to molecular targets, antibody formats and mode-of-action, pre-clinical validation and selection of most relevant patient populations, in order to increase the likelihood of successful product development in this field.


Subject(s)
Anti-Bacterial Agents/immunology , Anti-Bacterial Agents/therapeutic use , Antibodies, Monoclonal/immunology , Antibodies, Monoclonal/therapeutic use , Bacteria/immunology , Bacterial Infections/drug therapy , Animals , Bacteria/drug effects , Bacterial Infections/immunology , Drug Discovery/methods , Drug Resistance, Bacterial , Humans
7.
Exp Toxicol Pathol ; 64(4): 321-32, 2012 May.
Article in English | MEDLINE | ID: mdl-20951558

ABSTRACT

Assessment of mammary gland proliferation in rats is an important endpoint in preclinical safety studies of pharmaceutical compounds. However, existing data on mammary gland proliferation in rats during the estrous cycle is conflicting, and it is unknown whether mammary gland proliferation differs between young and mature female virgin rats. Additionally, it is unclear which of the commonly applied markers of proliferating cells that is optimal for assessment of rat mammary gland proliferation. In this study the caudal thoracic, the abdominal and the cranial inguinal (i.e., the 3rd the 4th and the 5th) mammary gland were collected from 29 young and 26 mature non-treated, virgin female Sprague Dawley rats. Estrous cycle stage was determined from repeated vaginal smears and histological examination of the reproductive organs. Proliferation of mammary epithelium was assessed by immunohistochemistry using three markers: PCNA, Ki67, and BrdU. Proliferation of the mammary epithelium occurred mainly in the terminal end buds in the young animals. Epithelial proliferation was significantly increased during metestrus compared to the other phases. Mammary gland proliferation in pseudo-pregnant females was increased compared to proestrus, estrus and diestrus, but not metestrus. Except during estrus no difference in mammary gland proliferation was observed between young and mature female rats, and no significant differences was observed between different mammary glands. The percentages of PCNA-, Ki67- and BrdU-positive epithelial cells were significantly correlated. In conclusion, the variation in normal proliferation between estrous cycle stages and animals with an irregular estrous cycle should be considered in toxico-pathological studies of mammary gland proliferation.


Subject(s)
Aging/physiology , Estrous Cycle/physiology , Mammary Glands, Animal/cytology , Pseudopregnancy/physiopathology , Age Factors , Animals , Biomarkers/metabolism , Bromodeoxyuridine/metabolism , Cell Proliferation , Female , Ki-67 Antigen/metabolism , Mammary Glands, Animal/growth & development , Mammary Glands, Animal/metabolism , Proliferating Cell Nuclear Antigen/metabolism , Rats , Rats, Sprague-Dawley
8.
PLoS One ; 7(12): e51972, 2012.
Article in English | MEDLINE | ID: mdl-23300584

ABSTRACT

Insulin can trigger metabolic as well as mitogenic effects, the latter being pharmaceutically undesirable. An understanding of the structure/function relationships between insulin receptor (IR) binding and mitogenic/metabolic signalling would greatly facilitate the preclinical development of new insulin analogues. The occurrence of ligand agonism and antagonism is well described for G protein-coupled receptors (GPCRs) and other receptors but in general, with the exception of antibodies, not for receptor tyrosine kinases (RTKs). In the case of the IR, no natural ligand or insulin analogue has been shown to exhibit antagonistic properties, with the exception of a crosslinked insulin dimer (B29-B'29). However, synthetic monomeric or dimeric peptides targeting sites 1 or 2 of the IR were shown to be either agonists or antagonists. We found here that the S961 peptide, previously described to be an IR antagonist, exhibited partial agonistic effects in the 1-10 nM range, showing altogether a bell-shaped dose-response curve. Intriguingly, the agonistic effects of S961 were seen only on mitogenic endpoints ((3)H-thymidine incorporation), and not on metabolic endpoints ((14)C-glucose incorporation in adipocytes and muscle cells). The agonistic effects of S961 were observed in 3 independent cell lines, with complete concordance between mitogenicity ((3)H-thymidine incorporation) and phosphorylation of the IR and Akt. Together with the B29-B'29 crosslinked dimer, S961 is a rare example of a mixed agonist/antagonist for the human IR. A plausible mechanistic explanation based on the bivalent crosslinking model of IR activation is proposed.


Subject(s)
Insulin/metabolism , Peptides/pharmacology , Receptor, Insulin/agonists , Receptor, Insulin/antagonists & inhibitors , 3T3-L1 Cells , Adipocytes/cytology , Adipocytes/drug effects , Adipocytes/metabolism , Animals , Breast Neoplasms/drug therapy , Breast Neoplasms/metabolism , Breast Neoplasms/pathology , CHO Cells , Cell Differentiation/drug effects , Cells, Cultured , Cricetinae , Female , Glucose/metabolism , Humans , Insulin/chemistry , Insulin-Like Growth Factor I/metabolism , Mice , Myoblasts/cytology , Myoblasts/drug effects , Myoblasts/metabolism , Phosphorylation/drug effects , Proto-Oncogene Proteins c-akt/metabolism , Rats , Receptor, IGF Type 1/metabolism , Receptor, Insulin/metabolism , Signal Transduction/drug effects
9.
Toxicol Pathol ; 39(4): 623-40, 2011 Jun.
Article in English | MEDLINE | ID: mdl-21558470

ABSTRACT

High doses of insulin and the insulin analog AspB10 have been reported to increase mammary tumor incidence in female rats likely via receptor-mediated mechanisms, possibly involving enhanced IGF-1 receptor activation. However, insulin and IGF-1 receptor functionality and intracellular signaling in the rat mammary gland in vivo is essentially unexplored. The authors investigated the effect of a single subcutaneous dose of 600 nmol/kg human insulin or IGF-1 on Akt and ERK1/2 phosphorylation in rat liver, colon, and mammary gland. Rat tissues were examined by Western blotting and immunohistochemistry by phosphorylation-specific antibodies. Insulin as well as IGF-1 caused Akt phosphorylation in mammary epithelial cells, with myoepithelial and basal epithelial cells being most sensitive. IGF-1 caused stronger Akt phosphorylation than insulin in mammary gland epithelial cells. Phosphorylation of ERK1/2 was not influenced by insulin or IGF-1. Rather, in liver and mammary gland P-ERK1/2 appeared to correlate with estrous cycling, supporting that ERK1/2 has important physiological roles in these two organs. In short, these findings supported that the rat mammary gland epithelium expresses functional insulin and IGF-1 receptors and that phosphorylation of Akt as well as ERK1/2 may be of value in understanding the effects of exogenous insulin in the rat mammary gland and colon.


Subject(s)
Insulin-Like Growth Factor I/pharmacology , Insulin/pharmacology , Mitogen-Activated Protein Kinase 1/genetics , Mitogen-Activated Protein Kinase 3/genetics , Proto-Oncogene Proteins c-akt/genetics , Animals , Blotting, Western , Breast/cytology , Breast/drug effects , Colon/drug effects , Epithelial Cells/drug effects , Female , Humans , Immunohistochemistry , Liver/drug effects , Mammary Glands, Animal/drug effects , Mitogen-Activated Protein Kinase 1/metabolism , Mitogen-Activated Protein Kinase 3/metabolism , Phosphorylation , Proto-Oncogene Proteins c-akt/metabolism , Rats , Rats, Sprague-Dawley , Receptor, IGF Type 1/metabolism , Signal Transduction/drug effects
10.
J Appl Toxicol ; 31(4): 312-28, 2011 May.
Article in English | MEDLINE | ID: mdl-21259294

ABSTRACT

Supra-pharmacological doses of the insulin analog X10 (AspB10) increased the incidence of mammary tumors in female Sprague-Dawley rats in chronic toxicity studies, most likely via receptor-mediated mechanisms. However, little is known about the expression of the insulin receptor family in the rat mammary gland. Using laser micro-dissection, quantitative RT-PCR and immunohistochemistry, we examined the expression of IR (insulin receptor), IGF-1R (IGF-1 receptor), IRR (insulin receptor-related receptor), ERα (estrogen receptor alpha), ERß (estrogen receptor beta) and PR (progesteron receptor) in young, virgin, female Sprague-Dawley rats and compared to expression in reference organs. The mammary gland displayed the highest expression of IRR and IGF-1R. In contrast, low expression of IR transcripts was observed in the mammary gland tissue with expression of the IR-A isoform being 5-fold higher than the expression of the IR-B. By immunohistochemistry, expression of IR and IGF-1R was detected in all mammary gland epithelial cells. Expression of ERα and PR was comparable between mammary gland and ovary, whereas expression of ERß was lower in mammary gland than in the ovary. Finally, expression of IGF-1R and PR in the mammary gland varied during the estrous cycle. These findings are important for the understanding of carcinogenic effects of insulin analogs in the rat mammary gland, and relevant for development of refined short-term models for preclinical safety assessment of insulin analogs.


Subject(s)
Mammary Glands, Animal/metabolism , Receptor, IGF Type 1/biosynthesis , Receptor, Insulin/biosynthesis , Animals , Estrogen Receptor alpha/biosynthesis , Estrogen Receptor alpha/genetics , Estrogen Receptor beta/biosynthesis , Estrogen Receptor beta/genetics , Estrous Cycle/metabolism , Female , Immunohistochemistry , Mammary Neoplasms, Experimental/metabolism , Rats , Rats, Sprague-Dawley , Receptor, IGF Type 1/genetics , Receptor, Insulin/genetics , Reverse Transcriptase Polymerase Chain Reaction
11.
Vet J ; 190(1): 103-8, 2011 Oct.
Article in English | MEDLINE | ID: mdl-20952231

ABSTRACT

The function and development of the rat mammary gland is dependent on the oestrus cycle. Normalization of gene expression in mammary gland samples assessed by quantitative RT-PCR therefore requires housekeeping genes (HKGs) which are stably expressed during the oestrus cycle. mRNA expression of 10 HKGs was measured in the rat mammary gland at different phases of the oestrus cycle. In addition, mRNA expression of the HKGs was measured in a panel of other rat tissues comprising laser microdissected mammary gland alveolar lobules and interlobular connective tissue and macrodissected mammary gland, liver, skeletal muscle, colon and ovary samples. Expression and ranking of HKGs varied between tissues and oestrus cycle phases and several HKGs were necessary for normalization between samples. In the mammary gland samples, three HKGs (Sdha, Tbp, and Atp5b) were identified as the optimal combination of stably expressed genes across oestrus cycle phases. For normalization between samples from the entire panel of rat tissues, eight HKGs (Rps18, Eef1a1, B2m, Actb, Tbp, Hprt, Pgk1, and Sdha) were identified as the optimal combination. These HKGs are of general relevance for studies comparing gene expression between different rat tissues.


Subject(s)
Gene Expression Profiling/veterinary , Genes, Essential , Mammary Glands, Animal/metabolism , Rats/genetics , Animals , Biomarkers/analysis , Female , Gene Expression Profiling/methods , Microdissection/veterinary , Polymerase Chain Reaction/veterinary , RNA, Messenger/metabolism , Rats, Sprague-Dawley , Reference Standards , Tissue Distribution
12.
J Appl Toxicol ; 31(4): 329-41, 2011 May.
Article in English | MEDLINE | ID: mdl-20936651

ABSTRACT

We compared mitogenicity and intracellular signalling by human insulin and the AspB10 (X-10) human insulin analogue in MCF-7 human mammary adenocarcinoma cells. By flow analysis of phosphorylated histone H3 or cell cycle distributions, insulin and X-10 were mitogenic at physiologically relevant concentrations (2 nm to 74 pm range), with X-10 being approximately 3-fold more mitogenic than insulin. By western blotting with phospho-specific antibodies, insulin induced phosphorylation of IRS-1, Akt, p70S6K, S6 ribosomal protein, 4E-BP1, FoxO3a, FoxO1, p44/42 MAPK and the EGFR. Blocking with wortmannin, rapamycin and U0126 showed that these signalling events conformed to the canonical PI3K pathway. IRS-1 (Ser302) phosphorylation was abolished by wortmannin and rapamycin, suggesting a feedback from the PI3K pathway on insulin signalling. Compared with equimolar insulin, X-10 caused up to 2-fold higher phosphorylation of all proteins examined in this study. The phosphorylation sites that responded most strongly to insulin were not generally the same as those responding most strongly to X-10. In the PI3K pathway, the most X-10-sensitive protein localized to the translation-regulating arm (p70S6K), with FoxO3a and FoxO1 transcription factors showing a more comparable response to insulin and X-10. Using flow analysis, we confirmed the correlation between insulin-triggered translational activation in G0/G1 (S6 phosphorylation) and S-phase entry by MCF-7 cells. In summary, our findings implicate asymmetrical PI3K pathway activation and specifically stimulation of protein translation in the hypermitogenic effect of insulin analogues such as X-10. It remains to be shown whether these findings are relevant to other human mammary cancer cell types.


Subject(s)
Cell Cycle/drug effects , Insulin/analogs & derivatives , Mitogens/pharmacology , Signal Transduction/drug effects , Blotting, Western , Cell Culture Techniques , Cell Line, Tumor , Dose-Response Relationship, Drug , Elafin/metabolism , Flow Cytometry , Humans , Insulin/pharmacology , Phosphorylation , Protein Transport , Receptor, IGF Type 1/genetics , Receptor, IGF Type 1/metabolism , Receptor, Insulin/genetics , Receptor, Insulin/metabolism
13.
Exp Toxicol Pathol ; 63(4): 317-24, 2011 May.
Article in English | MEDLINE | ID: mdl-20199854

ABSTRACT

In toxicopathological studies of the rat mammary glands, the guidelines of the Registry of Industrial Toxicology and Animal Data (RITA) recommend transverse sections of the inguinal mammary gland. However, occasionally limited amounts of mammary gland tissue are found in transverse sections. We compared transverse sectioning with an alternative method comprising horizontal sections of the rat mammary glands. Normal cycling female Sprague Dawley rats were sacrificed in proestrous, estrous, metestrous and diestrous, and samples from all mammary glands were collected. Transverse sections were prepared from the left-sided glands, and horizontal sections were cut from the right-sided glands. Sections were stained with HE, and epithelial and myoepithelial cells were visualized by immunohistochemical staining of cytokeratin 18 and alpha smooth muscle actin, respectively. Area of the mammary fat pad, mammary epithelium and connective tissue were measured in randomly sampled vision fields from each section. Horizontal sections contained a significantly larger area of mammary fat pad as well as glandular and connective tissue. No differences in tissue densities of epithelial or myoepithelial cells or connective tissue were observed between transverse sections and horizontal sections. Interestingly, densities of epithelium and connective tissue varied between cranial and caudal glands as well as the phases of the estrous cycle. In conclusion, horizontal histological sections of the rat mammary gland provided significantly larger samples of mammary gland tissue with no difference in tissue composition compared to transverse sections, which are recommended in the RITA guidelines.


Subject(s)
Mammary Glands, Animal/pathology , Microtomy/methods , Animals , Female , Guidelines as Topic , Immunohistochemistry , Rats , Rats, Sprague-Dawley , Toxicology/methods
14.
J Appl Toxicol ; 30(2): 151-62, 2010 Mar.
Article in English | MEDLINE | ID: mdl-19757489

ABSTRACT

Some dual-acting PPARalpha + gamma agonists cause cancer in the rat urinary bladder, in some cases overrepresented in males, by a mechanism suggested to involve chronic stimulation of PPARalpha and PPARgamma, i.e. exaggerated pharmacology. By western blotting, we found that the rat urinary bladder urothelium expressed PPARalpha at higher levels than the liver and heart, and comparable to kidney. Urothelial expression of PPARgamma was above that of fat, heart, skeletal muscle and kidney. Male rats exhibited a higher PPARalpha/PPARgamma expression balance in the bladder urothelium than did female rats. Rats were treated by gastric gavage with rosiglitazone (PPARgamma agonist), fenofibrate (PPARalpha agonist) or a combination of rosiglitazone and fenofibrate for 7 days. In the urothelium, the transcription factor Egr-1 was induced to significantly higher levels in rats co-administered rosiglitazone and fenofibrate than in rats administered either rosiglitazone or fenofibrate alone. Egr-1 was also induced in the heart and liver of rats treated with fenofibrate, but a positive interaction between rosiglitazone and fenofibrate with regards to Egr-1 induction was only seen in the urothelium. Thus, in the rat urinary bladder urothelium, PPARalpha and PPARgamma were expressed at high levels, were functional and exhibited positive interactions. Interestingly, fenofibrate induced the peroxisome membrane protein PMP70 not only in liver, but also in the bladder urothelium, opening the possibility that oxidative stress may contribute to rat urothelial carcinogenesis by dual-acting PPARalpha + gamma agonists.


Subject(s)
PPAR alpha/metabolism , PPAR gamma/metabolism , Urinary Bladder/metabolism , Urothelium/metabolism , Animals , Blotting, Western , Female , Fenofibrate/metabolism , Liver/metabolism , Male , Proteins/metabolism , Rats , Rats, Sprague-Dawley , Rosiglitazone , Thiazolidinediones/metabolism
15.
Cell Biol Toxicol ; 26(4): 293-307, 2010 Aug.
Article in English | MEDLINE | ID: mdl-19898946

ABSTRACT

Evaluating mitogenic signaling specifically through the human insulin receptor (IR) is relevant for the preclinical safety assessment of developmental insulin analogs. It is known that overexpression of IR sensitizes cells to the mitogenic effects of insulin, but it is essentially unknown how mitogenic responses can be optimized to allow practical use of such recombinant cell lines for preclinical safety testing. We constitutively overexpressed the short isoform of the human insulin receptor (hIR-A, exon 11-negative) in L6 rat skeletal myoblasts. Because the mitogenic effect of growth factors such as insulin is expected to act in G0/G1, promoting S-phase entry, we developed a combined topoinhibition + serum deprivation strategy to explore the effect of G0/G1 synchronization as an independent parameter in the context of serum deprivation, the latter being routinely used to reduce background in mitogenicity assays. G0/G1 synchronization significantly improved the mitogenic responses of L6-hIR cells to insulin, measured by (3)H-thymidine incorporation. Comparison with the parental L6 cells using phospho-mitogen-activated protein kinase, phospho-AKT, as well as (3)H-thymidine incorporation end points supported that the majority of the mitogenic effect of insulin in L6-hIR cells was mediated by the overexpressed hIR-A. Using the optimized L6-hIR assay, we found that the X-10 insulin analog was more mitogenic than native human insulin, supporting that X-10 exhibits increased mitogenic signaling through the hIR-A. In summary, this study provides the first demonstration that serum deprivation may not be sufficient, and G0/G1 synchronization may be required to obtain optimal responsiveness of hIR-overexpressing cell lines for preclinical safety testing.


Subject(s)
Antigens, CD/metabolism , G1 Phase/drug effects , Insulin/pharmacology , Mitogens/pharmacology , Muscle Cells/cytology , Muscle Cells/metabolism , Receptor, Insulin/metabolism , Resting Phase, Cell Cycle/drug effects , Animals , Cell Membrane/drug effects , Cell Membrane/metabolism , Cell Proliferation/drug effects , Culture Media, Serum-Free , Humans , Muscle Cells/drug effects , Muscle, Skeletal/cytology , Protein Isoforms/metabolism , Rats , Signal Transduction
16.
Toxicol Pathol ; 37(7): 947-58, 2009 Dec.
Article in English | MEDLINE | ID: mdl-20008548

ABSTRACT

To facilitate studies of the rat bladder carcinogenicity of dual-acting PPAR alpha+gamma agonists, we previously identified the Egr-1 transcription factor as a candidate carcinogenicity biomarker and developed rat models based on coadministration of commercially available specific PPAR alpha and PPAR gamma agonists. Immunohistochemistry for Egr-1 with a rabbit monoclonal antibody demonstrated that male vehicle-treated rats exhibited minimal urothelial expression and specifically, no nuclear signal. In contrast, Egr-1 was induced in the nuclei of bladder, as well as kidney pelvis, urothelia within one day (2 doses) of oral dosing of rats with a combination of 8 mg/kg rosiglitazone and 200 mg/kg fenofibrate (specific PPAR gamma and PPAR alpha agonists, respectively). These findings were confirmed by Western blotting using a different Egr-1 antibody. Egr-1 was induced to similar levels in the dorsal and ventral bladder urothelium, arguing against involvement of urinary solids. Egr-1 induction sometimes occurred in a localized fashion, indicating physiological microheterogeneity in the urothelium. The rapid kinetics supported that Egr-1 induction occurred as a result of pharmacological activation of PPAR alpha and PPAR gamma, which are coexpressed at high levels in the rat urothelium. Finally, our demonstration of a nuclear localization supports that the Egr-1 induced by PPAR alpha and PPAR gamma coactivation in the rat urothelium may be biologically active.


Subject(s)
Cell Nucleus/metabolism , Early Growth Response Protein 1/metabolism , Kidney Pelvis/metabolism , PPAR alpha/agonists , PPAR gamma/agonists , Urinary Bladder/metabolism , Urothelium/metabolism , Administration, Oral , Animals , Carcinogenicity Tests , Carcinogens/administration & dosage , Carcinogens/toxicity , Cell Nucleus/genetics , Fenofibrate/administration & dosage , Fenofibrate/toxicity , Male , PPAR alpha/metabolism , PPAR gamma/metabolism , Rats , Rosiglitazone , Thiazolidinediones/administration & dosage , Thiazolidinediones/toxicity
17.
BMC Cancer ; 9: 385, 2009 Oct 30.
Article in English | MEDLINE | ID: mdl-19878561

ABSTRACT

BACKGROUND: Egr-1 (early growth response-1 transcription factor) has been proposed to be involved in invasion and metastasis processes of human bladder cancer, but Egr-1 protein expression levels in human bladder cancer have not been investigated. In the present study we investigated the expression levels of Egr-1 protein in early stages of human bladder cancer and correlated it to later progression. METHODS: Expression of Egr-1 protein in human bladder cancer was examined by immunohistochemistry, on a tissue microarray constructed from tumors from 289 patients with non-muscle invasive urothelial bladder cancer. RESULTS: The frequency of tumor cells with nuclear Egr-1 immunolabelling correlated to bladder cancer stage, grade and to later progression to muscle-invasive bladder cancer (T2-4). Stage T1 tumors exhibited significantly higher frequencies of tumor cells with nuclear Egr-1 immunolabelling than Ta tumors (P = 0.001). Furthermore, Kaplan-Meier survival analysis showed that a high frequency of tumor cells with nuclear Egr-1 immunolabelling was significantly associated with a higher risk of progression to stage T2-4 (log-rank test, P = 0.035). Tumor cells with nuclear Egr-1 immunolabelling were found to localize at the tumor front in some of the tumor biopsies. CONCLUSION: The results from this study support a potential involvement of Egr-1 in the progression from non-muscle invasive bladder cancers to muscle invasive bladder cancer.


Subject(s)
Early Growth Response Protein 1/genetics , Gene Expression Regulation, Neoplastic , Urinary Bladder Neoplasms/genetics , Urinary Bladder Neoplasms/pathology , Disease Progression , Early Growth Response Protein 1/metabolism , Female , Follow-Up Studies , Humans , Male , Neoplasm Staging , Urinary Bladder Neoplasms/metabolism
18.
Arch Toxicol ; 83(12): 1061-74, 2009 Dec.
Article in English | MEDLINE | ID: mdl-19730820

ABSTRACT

Insulin-exposed rat mammary cancer cells were flow sorted based on a c-myc reporter plasmid encoding a destabilized green fluorescent protein. Sorted cells exhibited gradual increases in c-myc levels. Cells overexpressing c-myc by only 10% exhibited phenotypic changes attributable to c-myc overexpression, such as cell cycle disturbances, increased cell size, and overexpression of the S6 ribosomal protein. Cells overexpressing c-myc by 70% exhibited additional phenotypic changes typical of c-myc overexpression, such as increased histone H3 phosphorylation, and reduced adherence. Sorted cells also exhibited overexpression of the IGF-1R, and slightly elevated expression of the IR. Increased susceptibility to the mitogenic effect of insulin was seen in a small proportion of the sorted cells, and insulin was more effective in activating the p44/42 MAPK pathway, but not the PI3K pathway, in the sorted cells than in the nonsorted cell population. To our knowledge, this is the first in vitro system allowing functional coupling between mitogenic signaling by a well-defined growth factor and gradual overexpression of the normal, endogenous c-myc gene. Thus, our flow-sorting approach provides an alternative modeling of the receptor-mediated carcinogenic process, compared to the currently used approaches of recombinant constitutive or conditional overexpression of oncogenic transmembrane receptor tyrosine kinases or oncogenic transcription factors.


Subject(s)
Adenocarcinoma/metabolism , Flow Cytometry/methods , Mammary Neoplasms, Animal/metabolism , Proto-Oncogene Proteins c-myc/metabolism , Adenocarcinoma/drug therapy , Adenocarcinoma/pathology , Animals , Cell Adhesion , Cell Line, Tumor , Cell Separation , Female , Histones/metabolism , Insulin/pharmacology , Mammary Neoplasms, Animal/drug therapy , Mammary Neoplasms, Animal/pathology , Mitogen-Activated Protein Kinase 1/metabolism , Mitogen-Activated Protein Kinase 3/metabolism , Phenotype , Phosphatidylinositol 3-Kinases/metabolism , Phosphorylation , Rats , Receptor, IGF Type 1/metabolism , Signal Transduction
19.
J Appl Toxicol ; 29(6): 470-7, 2009 Aug.
Article in English | MEDLINE | ID: mdl-19338014

ABSTRACT

Human mammary cell lines are extensively used for preclinical safety assessment of insulin analogs. However, it is essentially unknown how mitogenic responses can be optimized in mammary cell-based systems. We developed an insulin mitogenicity assay in MCF-7 human mammary adenocarcinoma cells, under low serum (0.1% FCS) and phenol red-free conditions, with 3H thymidine incorporation as endpoint. Based on EC50 values determined from 10-fold dilution series, beta-estradiol was the most potent mitogen, followed by human IGF-1, human AspB10 insulin and native human insulin. AspB10 insulin was significantly more mitogenic than native insulin, validating the ability of the assay to identify hypermitogenic human insulin analogs. With MCF-7 cells on a collagen IV surface, the ranking of mitogens was maintained, but fold mitogenic responses and dynamic range and steepness of dose-response curves were increased. Also, PI3K pathway activation by insulin was enhanced on a collagen IV surface. This study provided the first determination and ranking of the mitogenic potencies of standard reference compounds in an optimized MCF-7 assay. The optimized MCF-7 assay described here is of relevance for in vitro toxicological testing and carcinogenicity safety assessment of new insulin compounds.


Subject(s)
Adenocarcinoma/metabolism , Breast Neoplasms/metabolism , Collagen Type IV/metabolism , Insulin/metabolism , Adenocarcinoma/pathology , Animals , Breast Neoplasms/pathology , Cell Line, Tumor , Collagen Type IV/pharmacology , Dose-Response Relationship, Drug , Estradiol/metabolism , Female , Humans , Hypoglycemic Agents/metabolism , Hypoglycemic Agents/pharmacology , Insulin/analogs & derivatives , Insulin/pharmacology , Lethal Dose 50 , Mice , Mitogens/metabolism , Mitosis , Reference Standards , Reproducibility of Results , Thymidine/metabolism
20.
Arch Virol ; 153(8): 1479-88, 2008.
Article in English | MEDLINE | ID: mdl-18592131

ABSTRACT

The nucleocapsid protein of the European genotype of porcine reproductive and respiratory syndrome virus (type 1, PRRSV-1) exhibited extensive size polymorphism (124-130 amino acids), correlating with phylogenetic grouping of ORF7 as well as ORF5 nucleotide sequences, thereby validating ORF7 size as an independent PRRSV-1 subtype marker. Based on new sequence information from the Russian Federation, we propose division of European genotype PRRSV-1 into 3 subtypes: a pan-European subtype 1 and East European subtypes 2 and 3, with nucleocapsid protein sizes of 128, 125 and 124 amino acids, respectively. The genetic differences between European genotype PRRSV subtypes affected diagnostic RT-PCR primer binding sites. Using Escherichia coli-expressed ORF7 protein, we confirmed that even the relatively closely related PRRSV subtypes 2 and 3 were antigenically different. Finally, the isoelectric point (pI) correlated with the nucleocapsid protein size for European genotype PRRSV subtypes, suggesting subtype-specific compensatory structural changes associated with subtype-specific ORF7 sizes. Thus, the new ORF7-based subtype division of PRRSV-1 proposed here is biologically meaningful and practically relevant.


Subject(s)
Nucleocapsid Proteins/chemistry , Porcine Reproductive and Respiratory Syndrome/epidemiology , Porcine respiratory and reproductive syndrome virus/classification , Animals , Europe/epidemiology , Genotype , Geography , Nucleocapsid Proteins/genetics , Porcine Reproductive and Respiratory Syndrome/virology , Porcine respiratory and reproductive syndrome virus/genetics , Porcine respiratory and reproductive syndrome virus/immunology , Porcine respiratory and reproductive syndrome virus/isolation & purification , Swine Diseases/virology
SELECTION OF CITATIONS
SEARCH DETAIL
...