Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 18 de 18
Filter
1.
J Appl Toxicol ; 42(8): 1371-1384, 2022 08.
Article in English | MEDLINE | ID: mdl-35152467

ABSTRACT

Toxicologic evaluation of new drug candidates routinely utilizes healthy animals. In oncology, there remains a limited understanding of the effects of novel test candidates in a diseased host. For vascular modulating agents (VMAs), an increased understanding of preclinical tumour-host interaction, and its potential to exacerbate or alleviate 'off-target' effects of anti-angiogenic administration, could aid in the prediction of adverse clinical outcomes in a defined cancer patient. We have previously reported that the implantation and growth of a range of human- and mouse-derived tumours leads to structural vascular and, potentially, functional signalling changes within host mouse endocrine tissues, indicating possible roles for tumour- and host-derived cytokines/growth factors and the liberation of myeloid-derived suppressor cells in this phenomenon. Here, we further demonstrate that the growth of the Calu-6 xenograft is associated with a resistance to VMA-induced mouse peripheral endocrine vascular rarefaction (toxicity), with potential functional impact, notably with respect to mixed tyrosine kinase inhibition. The pathogenesis of these findings indicates a potential role for both tumour- and host-derived basic fibroblast growth factor (bFGF), with associated upregulation in the intra-tumoural autotaxin-lysophosphatic acid signalling axis.


Subject(s)
Neoplasms , Neovascularization, Pathologic , Animals , Humans , Mice , Neoplasms/drug therapy , Neovascularization, Pathologic/drug therapy
2.
J Appl Toxicol ; 41(12): 1952-1965, 2021 12.
Article in English | MEDLINE | ID: mdl-33977518

ABSTRACT

Allograft and xenograft transplantation into a mouse host is frequently utilized to study cancer biology, tumor behavior, and response to treatment. Preclinical studies employing these models often focus solely upon the intra-tumoral effects of a given treatment, without consideration of systemic toxicity or tumor-host interaction, nor whether this latter relationship could modulate the toxicologic response to therapy. Here it is demonstrated that the implantation and growth of a range of human- and mouse-derived cell lines leads to structural vascular and, potentially, functional changes within peripheral endocrine tissues, a process that could conceivably ameliorate the severity of anti-angiogenic-induced fenestrated vessel attenuation. Observations suggest a multifactorial process, which may involve host- and tumor-derived cytokines/growth factors, and the liberation of myeloid-derived suppressor cells. Further investigation revealed a structurally comparable response to the administration of exogenous estrogen. These findings, in addition to providing insight into the development of clinical anti-angiogenic "adaptation," may be of significance within the "cancer-cachexia" and cancer-related anemia syndromes in man.


Subject(s)
Anemia/physiopathology , Cachexia/physiopathology , Cytokines/metabolism , Endocrine System/physiopathology , Animals , Cell Line, Tumor , Mice , Neoplasms/physiopathology
3.
J Appl Toxicol ; 37(8): 902-912, 2017 08.
Article in English | MEDLINE | ID: mdl-28186326

ABSTRACT

The study of vascular modulation has received a great deal of attention in recent years as knowledge has increased around the role of angiogenesis within disease contexts such as cancer. Despite rapidly expanding insights into the molecular processes involved and the concomitant generation of a number of anticancer vascular modulating chemotherapeutics, techniques used in the measurement of structural vascular change have advanced more modestly, particularly with regard to the preclinical quantification of off-target vascular regression within systemic, notably endocrine, blood vessels. Such changes translate into a number of major clinical side effects and there remains a need for improved preclinical screening and analysis. Here we present the generation of a novel structural biomarker, which can be incorporated into a number of contemporary image analysis platforms and used to compare tumour versus systemic host tissue vascularity. By contrasting the measurements obtained, the preclinical efficacy of vascular modulating chemotherapies can be evaluated in light of the predicted therapeutic window. Copyright © 2017 John Wiley & Sons, Ltd.


Subject(s)
Angiogenesis Inhibitors/pharmacology , Endocrine System/blood supply , Image Processing, Computer-Assisted , Lung Neoplasms/blood supply , Microvessels/drug effects , Neovascularization, Pathologic/pathology , Animals , Female , Lung Neoplasms/pathology , Mice, Inbred C57BL , Mice, Nude , Microvessels/pathology , Vascular Endothelial Growth Factor Receptor-1/antagonists & inhibitors , Vascular Endothelial Growth Factor Receptor-2/antagonists & inhibitors , Xenograft Model Antitumor Assays
4.
Am J Physiol Renal Physiol ; 312(4): F748-F759, 2017 04 01.
Article in English | MEDLINE | ID: mdl-27440778

ABSTRACT

Diabetic nephropathy (DN) remains an unmet medical challenge as its prevalence is projected to continue to increase and specific medicines for treatment remain undeveloped. Activation of the immune system, in particular T-cells, is emerging as a possible mechanism underlying DN disease progression in humans and animal models. We hypothesized that inhibition of T-cell activation will ameliorate DN. Interaction of B7-1 (CD80) on the surface of antigen presenting cells with its binding partners, CTLA4 (CD152) and CD28 on T-cells, is essential for T-cell activation. In this study we used the soluble CTLA4-Fc fusion protein Abatacept to block cell surface B7-1, preventing the cellular interaction and inhibiting T-cell activation. When Abatacept was dosed in an animal model of diabetes-induced albuminuria, it reduced albuminuria in both prevention and intervention modes. The number of T-cells infiltrating the kidneys of DN animals correlated with the degree of albuminuria, and treatment with Abatacept reduced the number of renal T-cells. As B7-1 induction has been recently proposed to underlie podocyte damage in DN, Abatacept could be efficacious in DN by protecting podocytes. However, this does not appear to be the case as B7-1 was not expressed in 1) kidneys of DN animals; 2) stimulated human podocytes in culture; or 3) glomeruli of DN patients. We conclude that Abatacept ameliorates DN by blocking systemic T-cell activation and not by interacting with podocytes.


Subject(s)
Abatacept/pharmacology , Albuminuria/drug therapy , Diabetic Nephropathies/drug therapy , Immunosuppressive Agents/pharmacology , Kidney/drug effects , Lymphocyte Activation/drug effects , T-Lymphocytes/drug effects , Albuminuria/immunology , Albuminuria/metabolism , Albuminuria/pathology , Animals , B7-1 Antigen/immunology , B7-1 Antigen/metabolism , Cell Line , Collagen Type IV/metabolism , Diabetes Mellitus, Experimental/chemically induced , Diabetes Mellitus, Experimental/immunology , Diabetes Mellitus, Type 1/chemically induced , Diabetes Mellitus, Type 1/immunology , Diabetic Nephropathies/immunology , Diabetic Nephropathies/metabolism , Diabetic Nephropathies/pathology , Diet, High-Fat , Humans , Kidney/immunology , Kidney/metabolism , Kidney/pathology , Mice, Inbred C57BL , Podocytes/drug effects , Podocytes/immunology , Podocytes/metabolism , Streptozocin , T-Lymphocytes/immunology , Time Factors
5.
Toxicol Pathol ; 44(5): 663-72, 2016 07.
Article in English | MEDLINE | ID: mdl-26936079

ABSTRACT

Modulation of the cell cycle may underlie the toxicologic or pharmacologic responses of a potential therapeutic agent and contributes to decisions on its preclinical and clinical safety and efficacy. The descriptive and quantitative assessment of normal, aberrant, and degenerate mitotic figures in tissue sections is an important end point characterizing the effect of xenobiotics on the cell cycle. Historically, pathologists used manual counting and special staining visualization techniques such as immunohistochemistry for quantification of normal, aberrant, and degenerate mitotic figures. We designed an automated image analysis algorithm for measuring these mitotic figures in hematoxylin and eosin (H&E)-stained sections. Algorithm validation methods used data generated from a subcutaneous human transitional cell carcinoma xenograft model in nude rats treated with the cell cycle inhibitor Eg5. In these studies, we scanned and digitized H&E-stained xenografts and applied a complex ruleset of sequential mathematical filters and shape discriminators for classification of cell populations demonstrating normal, aberrant, or degenerate mitotic figures. The resultant classification system enabled the representations of three identifiable degrees of morphological change associated with tumor differentiation and compound effects. The numbers of mitotic figure variants and mitotic indices data generated corresponded to a manual assessment by a pathologist and supported automated algorithm verification and application for both efficacy and toxicity studies.


Subject(s)
Algorithms , Antineoplastic Agents/pharmacology , Image Processing, Computer-Assisted/methods , Kinesins/antagonists & inhibitors , Mitosis/drug effects , Animals , Carcinoma, Transitional Cell/pathology , Cell Line, Tumor , Humans , Rats , Urinary Bladder Neoplasms/pathology , Xenograft Model Antitumor Assays
6.
Lab Invest ; 95(6): 610-24, 2015 Jun.
Article in English | MEDLINE | ID: mdl-25531566

ABSTRACT

Murine γ-herpesvirus 68 (MHV-68) infection of Mus musculus-derived strains of mice is an established model of γ-herpesvirus infection. We have previously developed an alternative system using a natural host, the wood mouse (Apodemus sylvaticus), and shown that the MHV-68 M3 chemokine-binding protein contributes significantly to MHV-68 pathogenesis. Here we demonstrate in A. sylvaticus using high-density micro-arrays that M3 influences the expression of genes involved in the host response including Scgb1a1 and Bpifa1 that encode potential innate defense proteins secreted into the respiratory tract. Further analysis of MHV-68-infected animals showed that the levels of both protein and RNA for SCGB1A1 and BPIFA1 were decreased at day 7 post infection (p.i.) but increased at day 14 p.i. as compared with M3-deficient and mock-infected animals. The modulation of expression was most pronounced in bronchioles but was also present in the bronchi and trachea. Double staining using RNA in situ hybridization and immunohistology demonstrated that much of the BPIFA1 expression occurs in club cells along with SCGB1A1 and that BPIFA1 is stored within granules in these cells. The increase in SCGB1A1 and BPIFA1 expression at day 14 p.i. was associated with the differentiation of club cells into mucus-secreting cells. Our data highlight the role of club cells and the potential of SCGB1A1 and BPIFA1 as innate defense mediators during respiratory virus infection.


Subject(s)
Gammaherpesvirinae/genetics , Glycoproteins/metabolism , Herpesviridae Infections/metabolism , Herpesviridae Infections/virology , Phosphoproteins/metabolism , Uteroglobin/metabolism , Animals , Bronchioles/chemistry , Bronchioles/cytology , Bronchioles/metabolism , Bronchioles/virology , Glycoproteins/genetics , Herpesviridae Infections/genetics , Host-Pathogen Interactions/genetics , Murinae , Phosphoproteins/genetics , Respiratory Mucosa/chemistry , Respiratory Mucosa/cytology , Respiratory Mucosa/metabolism , Respiratory Mucosa/virology , Uteroglobin/genetics
7.
Int J Exp Pathol ; 95(1): 29-48, 2014 Feb.
Article in English | MEDLINE | ID: mdl-24456331

ABSTRACT

Antagonism of the effects of glucagon as an adjunct therapy with other glucose-lowering drugs in the chronic treatment of diabetes has been suggested to aggressively control blood glucose levels. Antagonism of glucagon effects, by targeting glucagon secretion or disabling the glucagon receptor, is associated with α-cell hyperplasia. We evaluated the influence of total glucagon withdrawal on islets of Langerhans using prohormone convertase-2 knockout mice (PC2-ko), in which α-cell hyperplasia is present from a young age and persists throughout life, in order to understand whether or not sustained glucagon deficit would lead to islet tumorigenesis. PC2-ko and wild-type (WT) mice were maintained drug-free, and cohorts of these groups sampled at 3, 12 and 18 months for plasma biochemical and morphological (histological, immunohistochemical, electron microscopical and image analytical) assessments. WT mice showed no islet tumours up to termination of the study, but PC2-ko animals displayed marked changes in islet morphology from α-cell hypertrophy/hyperplasia/atypical hyperplasia, to adenomas and carcinomas, these latter being first encountered at 6-8 months. Islet hyperplasias and tumours primarily consisted of α-cells associated to varying degrees with other islet endocrine cell types. In addition to substantial increases in islet neoplasia, increased α-cell neogenesis associated primarily with pancreatic duct(ule)s was present. We conclude that absolute blockade of the glucagon signal results in tumorigenesis and that the PC2-ko mouse represents a valuable model for investigation of islet tumours and pancreatic ductal neogenesis.


Subject(s)
Carcinogenesis/pathology , Cell Transformation, Neoplastic/pathology , Glucagon-Secreting Cells/pathology , Islets of Langerhans/pathology , Proprotein Convertase 2/deficiency , Proprotein Convertase 2/genetics , Adenoma/metabolism , Adenoma/pathology , Animals , Carcinogenesis/metabolism , Cell Proliferation , Cell Transformation, Neoplastic/metabolism , Disease Models, Animal , Epithelial-Mesenchymal Transition , Female , Glucagon/antagonists & inhibitors , Glucagon/metabolism , Glucagon-Secreting Cells/metabolism , Hyperplasia , Islets of Langerhans/metabolism , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Pancreatic Neoplasms/metabolism , Pancreatic Neoplasms/pathology , Proprotein Convertase 2/metabolism
8.
Toxicol Pathol ; 41(7): 992-1000, 2013.
Article in English | MEDLINE | ID: mdl-23416962

ABSTRACT

Image analysis is now routinely employed as a tool in toxicologic pathology to help quantitate end points of efficacy and safety. It is regarded as a proficient and a sensitive technique to generate numerical data that can be easily interrogated for statistical evaluation. Traditional semiquantitative pathology scoring on the other hand is sometimes regarded as less accurate due to the limitations of the scoring systems employed and the day-to-day variations often noted between pathologists. We therefore decided to generate an optimized histochemical staining and image analysis protocol to compare the accuracy of semiquantitative scoring with computerized image analysis. In order to achieve this, we describe a standardized protocol for staining and image analysis that eliminates or minimizes as many sources of error as possible. The results of this experiment demonstrate that despite consistent variations in scoring between two independent pathologists, correlation with image analysis data of 0.91 to 0.95 (Spearman's Rho test) was achieved. These data indicate that either image analysis or traditional semiquantitative scoring can generate accurate data. As a result of this, it appears that it is equally safe to employ either method dependent upon the complexity and the practicality of the task at hand provided that the experimental conditions are rigorously optimized and rigidly adhered to.


Subject(s)
Ferrocyanides/analysis , Histocytochemistry/methods , Image Processing, Computer-Assisted/methods , Iron/analysis , Liver/chemistry , Animals , Female , Ferrocyanides/metabolism , Liver/metabolism , Male , Organ Size , Rats , Rats, Wistar , Statistics, Nonparametric
9.
Toxicol Pathol ; 41(3): 425-44, 2013.
Article in English | MEDLINE | ID: mdl-23047688

ABSTRACT

Islets of Langerhans represent a heterogeneous population in insulin resistant and diabetic animals and humans as histological appearances and function vary substantially. Mathematical representation that reflects this morphological diversity will assist in assessment of degeneration and regeneration, enabling comparisons between species, strains, and experimental investigations. Our investigative approach used a model of islet degeneration in diabetic male obese Zucker Diabetic Fatty (ZDF) rats and evaluated its prevention using rosiglitazone treatment. Immunohistochemical staining (insulin and collagens I/III) with automated image analysis reliably measured numbers, area, clustering, and staining intensity of ß-cells and degree of islet fibrosis. Finite mixture mathematical modeling for the joint probability distribution of seven islet parameters to represent islet numerical data variation provided an automatic procedure for islet category allocations as normal or abnormal. Allocations for obese ZDF controls and rosiglitazone-treated animals were significantly different, with no significant difference between the latter and lean ZDF controls, indicative of differences within islet populations of individual animals, between lean and obese rat strains and following drug treatment. Islet morphology showed clear association with mathematical characterization. Information on islet morphology obtained by histopathological assessment of single pancreatic tissue sections was confirmed by this method showing drug-induced retardation of islet of Langerhans degeneration.


Subject(s)
Diabetes Mellitus, Experimental/drug therapy , Diabetes Mellitus, Experimental/pathology , Image Processing, Computer-Assisted/methods , Immunohistochemistry/methods , Islets of Langerhans/pathology , Obesity/drug therapy , Thiazolidinediones/pharmacology , Algorithms , Animals , Collagen Type I/analysis , Collagen Type I/chemistry , Collagen Type III/analysis , Collagen Type III/chemistry , Hypoglycemic Agents/pharmacology , Insulin/analysis , Insulin/chemistry , Islets of Langerhans/drug effects , Male , Models, Statistical , Obesity/metabolism , Obesity/pathology , Rats , Rats, Zucker , Rosiglitazone
10.
J Pharmacol Toxicol Methods ; 63(1): 102-14, 2011.
Article in English | MEDLINE | ID: mdl-20619348

ABSTRACT

INTRODUCTION: We have evaluated the ability of a semi-automated, optomotor reflex method to assess drug-induced visual dysfunction, in albino and pigmented rats and mice. METHODS: Male Han Wistar (HW) and Long Evans (LE) rats and mice (CD-1 and C57BL/6) were tested in a chamber formed by 4 computer monitors displaying a rotating vertical grating, to elicit head-tracking movements. The highest visible grating frequency was taken as the threshold of visual acuity, in cycles per degree (c/d). Animals received an intravenous infusion of either sodium iodate (50mg/kg) or 0.9% w/v NaCl (aq). They were tested 2h later, then re-tested daily for a further 3 days. The time course of the effect was assessed in HW rats over a 6-week period, including electron microscopy, and immunohistochemical analysis of markers of injury and repair in the retina. RESULTS: Baseline visual acuities for HW and LE rats were 0.355 ± 0.007 and 0.530 ± 0.004 c/d, respectively, and 0.296 ± 0.003 c/d and 0.370 ± 0.001 c/d for CD-1 and C57BL/6 mice, respectively (n=10 for each). In HW rats there was a dramatic loss of visual acuity 2h after administration of sodium iodate (0.021 ± 0.021 c/d; P<0.001). Less dramatic decreases in visual acuity were seen in LE rats and in the two mouse strains. In HW rats, visual acuity was restored after 4 weeks. This paralleled the histopathological recovery of the peripheral retina, whereas the central retina did not recover. DISCUSSION: The method proved to be very convenient, and the stability of visual acuity in vehicle control rats over a 6-week period also demonstrated its suitability for inclusion in long-term toxicity studies. Both albino and pigmented mice and rats are suitable for assessment of retinotoxicity using this method, but albino rats are the most sensitive to sodium iodate.


Subject(s)
Iodates/toxicity , Retina/drug effects , Toxicity Tests/methods , Visual Acuity/drug effects , Albinism , Animals , Male , Mice , Mice, Inbred C57BL , Rats , Rats, Long-Evans , Rats, Wistar , Retina/cytology , Time Factors
11.
Drug Metab Dispos ; 38(2): 341-6, 2010 Feb.
Article in English | MEDLINE | ID: mdl-19920056

ABSTRACT

The aldo-keto reductase (AKR) phase I drug metabolism enzyme superfamily is implicated in detoxification or bioactivation of a wide variety of carbonyl-bearing compounds. In this study, we have used antibodies raised against purified recombinant rat AKR isoforms 1A3, 1B4, 1C9, 1D2, and 7A1 to characterize the expression profile of these superfamily members in the rat and define their localization by immunohistochemistry. Western blotting showed that AKR1A3, AKR1B4, and AKR1C9 are ubiquitously expressed, whereas AKR1D2 and AKR7A1 are present in liver, adrenal gland, and kidney, with the latter also present in testis, spleen, and stomach. Immunohistochemical analysis of the kidney demonstrated the localization of AKR1A3 in proximal convoluted tubules, AKR1B4 in the loop of Henle, and AKR1C9 in the pars recta S3 segment of proximal tubules. We also report localization of AKR1B4 in the adrenal gland (parenchymal cells of the zona reticularis) and testis (Sertoli cells and late spermatids), of AKR1D2 in the liver (hepatocyte nuclei), and of AKR7A1 in the pancreatic duct and bronchiolar epithelium. Previous studies have shown that expression of AKR7A1 is induced in response to dietary administration of the phenolic antioxidants butylated hydroxyanisole and ethoxyquin. Here we identify AKR1B13 and AKR1D2 as further inducible members of the rat AKR superfamily.


Subject(s)
Antioxidants/pharmacology , Gene Expression Regulation, Enzymologic/drug effects , Oxidoreductases/genetics , Oxidoreductases/metabolism , Aldehyde Reductase/genetics , Aldehyde Reductase/metabolism , Animals , Butylated Hydroxyanisole/pharmacology , Ethoxyquin/pharmacology , Female , Immunohistochemistry , Isoenzymes/genetics , Isoenzymes/metabolism , Liver/drug effects , Liver/metabolism , Male , Organ Specificity , RNA, Messenger/metabolism , Rats , Rats, Inbred F344 , Rats, Wistar , Regulatory Sequences, Nucleic Acid , Reverse Transcriptase Polymerase Chain Reaction
12.
Toxicol Pathol ; 36(2): 345-52, 2008 Feb.
Article in English | MEDLINE | ID: mdl-18362199

ABSTRACT

Rosuvastatin is a relatively new member of the statin family (HMG-CoA reductase inhibitors), with superior lipid-lowering effects and a pattern of clinical side effects, including a low incidence of myopathy, similar to other widely prescribed statins. This article describes investigations of myopathy in the rat following administration of very high doses of rosuvastatin. The nature of the changes were found to be entirely consistent with those seen with other statins, including a differential sensitivity of muscle fibers (with glycolytic fibers [type IIB] the most sensitive and oxidative fibers [type I] the least), a delay of approximately 10 days after the start of oral dosing before necrosis was apparent, and ultrastructural alterations appearing first in mitochondria. In addition, the development of myopathy was prevented by coadministration of mevalonate, the product of HMG-CoA reductase. The findings illustrate a pattern of induced myopathy in the rat directly attributable to inhibition of HMG-CoA reductase that is entirely consistent between the various statins, with the oral dose required to produce the changes being a differentiating feature (based on these new data and a previously reported study from the same laboratory): cerivastatin dose less than simvastatin, and simvastatin dose less than rosuvastatin.


Subject(s)
Fluorobenzenes/toxicity , Hydroxymethylglutaryl-CoA Reductase Inhibitors/toxicity , Muscle, Skeletal/drug effects , Muscular Diseases/chemically induced , Pyrimidines/toxicity , Sulfonamides/toxicity , Administration, Oral , Animals , Biomarkers/analysis , Body Weight/drug effects , Creatine Kinase/blood , Dose-Response Relationship, Drug , Female , Muscle Fibers, Fast-Twitch/drug effects , Muscle Fibers, Fast-Twitch/ultrastructure , Muscle Fibers, Slow-Twitch/drug effects , Muscle Fibers, Slow-Twitch/ultrastructure , Muscle, Skeletal/chemistry , Muscle, Skeletal/pathology , Muscular Diseases/blood , Muscular Diseases/pathology , Necrosis , Rats , Rats, Wistar , Rosuvastatin Calcium , Time Factors
13.
Clin Cancer Res ; 13(12): 3682-8, 2007 Jun 15.
Article in English | MEDLINE | ID: mdl-17575233

ABSTRACT

PURPOSE: In the current study, we examined the in vivo effects of AZD1152, a novel and specific inhibitor of Aurora kinase activity (with selectivity for Aurora B). EXPERIMENTAL DESIGN: The pharmacodynamic effects and efficacy of AZD1152 were determined in a panel of human tumor xenograft models. AZD1152 was dosed via several parenteral (s.c. osmotic mini-pump, i.p., and i.v.) routes. RESULTS: AZD1152 potently inhibited the growth of human colon, lung, and hematologic tumor xenografts (mean tumor growth inhibition range, 55% to > or =100%; P < 0.05) in immunodeficient mice. Detailed pharmacodynamic analysis in colorectal SW620 tumor-bearing athymic rats treated i.v. with AZD1152 revealed a temporal sequence of phenotypic events in tumors: transient suppression of histone H3 phosphorylation followed by accumulation of 4N DNA in cells (2.4-fold higher compared with controls) and then an increased proportion of polyploid cells (>4N DNA, 2.3-fold higher compared with controls). Histologic analysis showed aberrant cell division that was concurrent with an increase in apoptosis in AZD1152-treated tumors. Bone marrow analyses revealed transient myelosuppression with the drug that was fully reversible following cessation of AZD1152 treatment. CONCLUSIONS: These data suggest that selective targeting of Aurora B kinase may be a promising therapeutic approach for the treatment of a range of malignancies. In addition to the suppression of histone H3 phosphorylation, determination of tumor cell polyploidy and apoptosis may be useful biomarkers for this class of therapeutic agent. AZD1152 is currently in phase I trials.


Subject(s)
Antineoplastic Agents/pharmacology , Apoptosis/drug effects , Neoplasms, Experimental/drug therapy , Organophosphates/pharmacology , Quinazolines/pharmacology , Animals , Aurora Kinase B , Aurora Kinases , Cell Line, Tumor , Cell Proliferation/drug effects , Flow Cytometry , Humans , Mice , Mice, Nude , Protein Serine-Threonine Kinases/drug effects , Rats , Transplantation, Heterologous
14.
Exp Toxicol Pathol ; 57(3): 181-93, 2006 Jan.
Article in English | MEDLINE | ID: mdl-16410186

ABSTRACT

The use of tissue microarrays (TMAs) in the determination of novel target molecule distribution in organs is an expanding area of discovery pathology. This pilot study was carried out to assess the Chromavision automated cellular imaging system (ACIS) for quantitation of both mRNA and protein distribution in rat and dog TMAs. The targets chosen were a protein kinase, P-CIP2, for mRNA assessment and its downsteam target, peptidylglycine amidating monoxygenase (PAM), for immunohistochemistry (IHC). Oligonucleotide probes produced against P-CIP2, together with an antibody against PAM, were evaluated on rat and dog TMAs. A method for evaluation of target distribution using the ACIS was developed and involved a two-tier approach. Firstly, an initial scanning of the labelled slides identified which tissues expressed the target. Secondly, a more comprehensive analysis was made. This required operator interaction to select specific regions of interest within selected tissue cores and exclude any background labelling from the final assessment. This exacted the level of expression of P-CIP2 or PAM in different cellular populations in tissue cores. A comparative semi-quantitative analysis of the same arrays was concomitantly made by the pathologist in order to assess the relative benefits of a potentially time-consuming detailed morphological evaluation. This involved the histological identification by the pathologist of specific cell populations expressing P-CIP2 or PAM. In this study, we demonstrate the power of an image analysing system to provide quantitative data on target distribution by in situ hybridisation and IHC on normal TMAs. This methodology, together with detailed histological analysis by a pathologist, forms a guideline for future target distribution evaluation within discovery pathology.


Subject(s)
Carrier Proteins/biosynthesis , Cyclic AMP-Dependent Protein Kinases/biosynthesis , Mixed Function Oxygenases/biosynthesis , Multienzyme Complexes/biosynthesis , Pharmacokinetics , Protein Array Analysis , Animals , Automation , Carrier Proteins/analysis , Cyclic AMP-Dependent Protein Kinases/analysis , Dogs , Immunohistochemistry , In Situ Hybridization , Intracellular Signaling Peptides and Proteins , Mixed Function Oxygenases/analysis , Multienzyme Complexes/analysis , Protein Serine-Threonine Kinases , RNA, Messenger/analysis , Rats , Specimen Handling
15.
Toxicol Pathol ; 33(2): 246-57, 2005.
Article in English | MEDLINE | ID: mdl-15902968

ABSTRACT

Simvastatin and cerivastatin have been used to investigate the development of statin-induced muscle necrosis in the rat. This was similar for both statins and was treatment-duration dependent, only occurring after 10 days had elapsed even if the dose was increased, and still occurring after this time when dosing was terminated earlier as a result of morbidity. It was then widespread and affected all areas of the muscular system. However, even when myotoxicity was severe, particular individual muscles and some types of fibres within affected muscles were spared consistently. Fibre typing of spared muscles and of acutely necrotic fibres within affected muscles indicated a differential fibre sensitivity to statin-induced muscle necrosis. The fibres showed a necrotic response to statin administration that matched their oxidative/glycolytic metabolic nature: Least sensitive --> I < - > IIA < - > IID < - > IIB <-- most sensitive. Type I and IIB fibres represent metabolic extremes of a continuum of metabolic properties through the fibre types with type I fibres most oxidative in metabolism and type IIB fibres most glycolytic. In addition, in some (nonnecrotic) glycolytic fibres from muscles showing early multifocal single fibre necrosis the only subcellular alterations present in isolation of any other changes were mitochondrial. These changes were characterised by an increased incidence of vacuolation and the formation of myelinoid vesicular bodies that accumulated in the subsarcolemmal areas. These findings suggest an important early involvement of mitochondria in selective glycolytic muscle fibre necrosis following inhibition of the enzyme HMG-CoA reductase.


Subject(s)
Hydroxymethylglutaryl-CoA Reductase Inhibitors/toxicity , Muscle Fibers, Fast-Twitch/drug effects , Muscle Fibers, Slow-Twitch/drug effects , Muscle, Skeletal/drug effects , Pyridines/toxicity , Simvastatin/toxicity , Administration, Oral , Animals , Biomarkers/metabolism , Dose-Response Relationship, Drug , Female , Muscle Fibers, Fast-Twitch/metabolism , Muscle Fibers, Fast-Twitch/pathology , Muscle Fibers, Slow-Twitch/metabolism , Muscle Fibers, Slow-Twitch/pathology , Muscle, Skeletal/metabolism , Muscle, Skeletal/pathology , Myosins/metabolism , Rats , Rats, Wistar
16.
Cancer Res ; 65(10): 4389-400, 2005 May 15.
Article in English | MEDLINE | ID: mdl-15899831

ABSTRACT

Inhibition of vascular endothelial growth factor-A (VEGF) signaling is a promising therapeutic approach that aims to stabilize the progression of solid malignancies by abrogating tumor-induced angiogenesis. This may be accomplished by inhibiting the kinase activity of VEGF receptor-2 (KDR), which has a key role in mediating VEGF-induced responses. The novel indole-ether quinazoline AZD2171 is a highly potent (IC50 < 1 nmol/L) ATP-competitive inhibitor of recombinant KDR tyrosine kinase in vitro. Concordant with this activity, in human umbilical vein endothelial cells, AZD2171 inhibited VEGF-stimulated proliferation and KDR phosphorylation with IC50 values of 0.4 and 0.5 nmol/L, respectively. In a fibroblast/endothelial cell coculture model of vessel sprouting, AZD2171 also reduced vessel area, length, and branching at subnanomolar concentrations. Once-daily oral administration of AZD2171 ablated experimental (VEGF-induced) angiogenesis in vivo and inhibited endochondral ossification in bone or corpora luteal development in ovary; physiologic processes that are highly dependent upon neovascularization. The growth of established human tumor xenografts (colon, lung, prostate, breast, and ovary) in athymic mice was inhibited dose-dependently by AZD2171, with chronic administration of 1.5 mg per kg per day producing statistically significant inhibition in all models. A histologic analysis of Calu-6 lung tumors treated with AZD2171 revealed a reduction in microvessel density within 52 hours that became progressively greater with the duration of treatment. These changes are indicative of vascular regression within tumors. Collectively, the data obtained with AZD2171 are consistent with potent inhibition of VEGF signaling, angiogenesis, neovascular survival, and tumor growth. AZD2171 is being developed clinically as a once-daily oral therapy for the treatment of cancer.


Subject(s)
Neoplasms/drug therapy , Neoplasms/enzymology , Protein Kinase Inhibitors/pharmacology , Quinazolines/pharmacology , Vascular Endothelial Growth Factor Receptor-2/antagonists & inhibitors , Administration, Oral , Animals , Biological Availability , Bone Development/drug effects , Cell Proliferation/drug effects , Corpus Luteum/drug effects , Corpus Luteum/growth & development , Endothelial Cells/drug effects , Endothelial Cells/enzymology , Endothelial Cells/metabolism , Extracellular Matrix Proteins , Female , Humans , Mice , Myosin Heavy Chains , Neoplasms/blood supply , Neoplasms/pathology , Nonmuscle Myosin Type IIB , Phosphorylation/drug effects , Protein Kinase Inhibitors/pharmacokinetics , Proteins/antagonists & inhibitors , Quinazolines/pharmacokinetics , Vascular Endothelial Growth Factor A/antagonists & inhibitors , Vascular Endothelial Growth Factor Receptor-2/metabolism , Vascular Endothelial Growth Factor Receptor-3/antagonists & inhibitors , Xenograft Model Antitumor Assays
17.
J Histochem Cytochem ; 52(5): 653-62, 2004 May.
Article in English | MEDLINE | ID: mdl-15100242

ABSTRACT

Class kappa glutathione S-transferases are a poorly characterized family of detoxication enzymes whose localization has not been defined. In this study we investigated the tissue, cellular, and subcellular distribution of mouse glutathione S-transferase class kappa 1 (mGSTK1) protein using a variety of immunolocalization techniques. Western blotting analysis of mouse tissue homogenates demonstrated that mGSTK1 is expressed at relatively high levels in liver and stomach. Moderate expression was observed in kidney, heart, large intestine, testis, and lung, whereas sparse or essentially no mGSTK1 protein was detected in small intestine, brain, spleen, and skeletal muscle. Immunohistochemical (IHC) analysis for mGSTK1 revealed granular staining of hepatocytes throughout the liver, consistent with organelle staining. IHC analysis of murine kidney localized GSTK1 to the straight portion of the proximal convoluted tubule (pars recta). Staining was consistent with regions rich in mitochondria. Electron microscopy, using indirect immunocolloidal gold staining, clearly showed that mGSTK1 was localized in mitochondria in both mouse liver and kidney. These results are consistent with a role for mGST K1-1 in detoxification, and the confirmation of the intramitochondrial localization of this enzyme implies a unique role for GST class kappa as an antioxidant enzyme.


Subject(s)
Glutathione Transferase/metabolism , Subcellular Fractions/enzymology , Animals , Electron Transport Complex IV/biosynthesis , Electron Transport Complex IV/metabolism , Glutathione Transferase/biosynthesis , Kidney/enzymology , Kidney/metabolism , Kidney/ultrastructure , Liver/enzymology , Liver/metabolism , Liver/ultrastructure , Male , Mice , Mice, Inbred C57BL , Microscopy, Electron , Mitochondria/enzymology , Organ Specificity , Protein Subunits/metabolism
18.
Cancer Res ; 62(16): 4645-55, 2002 Aug 15.
Article in English | MEDLINE | ID: mdl-12183421

ABSTRACT

ZD6474 [N-(4-bromo-2-fluorophenyl)-6-methoxy-7-[(1-methylpiperidin-4-yl)methoxy]quinazolin-4-amine]is a potent, p.o. active, low molecular weight inhibitor of kinase insert domain-containing receptor [KDR/vascular endothelial growth factor receptor (VEGFR) 2] tyrosine kinase activity (IC(50) = 40 nM). This compound has some additional activity versus the tyrosine kinase activity of fms-like tyrosine kinase 4 (VEGFR3;IC(50) = 110 nM) and epidermal growth factor receptor (EGFR/HER1; IC(50) = 500 nM) and yet demonstrates selectivity against a range of other tyrosine and serine-threonine kinases. The activity of ZD6474 versus KDR tyrosine kinase translates into potent inhibition of vascular endothelial growth factor-A (VEGF)-stimulated endothelial cell (human umbilical vein endothelial cell) proliferation in vitro (IC(50) = 60 nM). Selective inhibition of VEGF signaling has been demonstrated in vivo in a growth factor-induced hypotension model in anesthetized rat: administration of ZD6474 (2.5 mg/kg, i.v.) reversed a hypotensive change induced by VEGF (by 63%) but did not significantly affect that induced by basic fibroblast growth factor. Once-daily oral administration of ZD6474 to growing rats for 14 days produced a dose-dependent increase in the femoro-tibial epiphyseal growth plate zone of hypertrophy, which is consistent with inhibition of VEGF signaling and angiogenesis in vivo. Administration of 50 mg/kg/day ZD6474 (once-daily, p.o.) to athymic mice with intradermally implanted A549 tumor cells also inhibited tumor-induced neovascularization significantly (63% inhibition after 5 days; P < 0.001). Oral administration of ZD6474 to athymic mice bearing established (0.15-0.47 cm(3)), histologically distinct (lung, prostate, breast, ovarian, colon, or vulval) human tumor xenografts or after implantation of aggressive syngeneic rodent tumors (lung, melanoma) in immunocompetent mice, produced a dose-dependent inhibition of tumor growth in all cases. Statistically significant antitumor activity was evident in each model with at least 25 mg/kg ZD6474 once daily (P < 0.05, one-tailed t test). Histological analysis of Calu-6 tumors treated with 50 mg/kg/day ZD6474 for 24 days showed a significant reduction (>70%) in CD31 (endothelial cell) staining in nonnecrotic regions. ZD6474 also restrained growth of much larger (0.9 cm(3) volume) Calu-6 lung tumor xenografts and induced profound regression in established PC-3 prostate tumors of 1.4 cm(3) volume. ZD6474 is currently in Phase I clinical development as a once-daily oral therapy in patients with advanced cancer.


Subject(s)
Angiogenesis Inhibitors/pharmacology , Antineoplastic Agents/pharmacology , Endothelial Growth Factors/antagonists & inhibitors , Lymphokines/antagonists & inhibitors , Piperidines/pharmacology , Quinazolines/pharmacology , Administration, Oral , Animals , Cell Division/drug effects , Endothelial Growth Factors/physiology , Enzyme Inhibitors/pharmacology , Female , Humans , Lymphokines/physiology , Male , Mice , Neoplasms, Experimental/blood supply , Neoplasms, Experimental/drug therapy , Neoplasms, Experimental/pathology , Neovascularization, Pathologic/drug therapy , Rats , Rats, Wistar , Receptor Protein-Tyrosine Kinases/antagonists & inhibitors , Receptors, Growth Factor/antagonists & inhibitors , Receptors, Vascular Endothelial Growth Factor , Signal Transduction/drug effects , Tumor Cells, Cultured , Vascular Endothelial Growth Factor A , Vascular Endothelial Growth Factors , Xenograft Model Antitumor Assays
SELECTION OF CITATIONS
SEARCH DETAIL
...