Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 8 de 8
Filter
Add more filters










Database
Language
Publication year range
1.
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
2.
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
3.
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
4.
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
5.
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
6.
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
7.
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
8.
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
...