Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 20
Filter
1.
Int J Mol Sci ; 23(6)2022 Mar 17.
Article in English | MEDLINE | ID: mdl-35328679

ABSTRACT

Osteosarcoma (OS) is a highly malignant bone tumour that has seen little improvement in treatment modalities in the past 30 years. Understanding what molecules contribute to OS biology could aid in the discovery of novel therapies. Extracellular vesicles (EVs) serve as a mode of cell-to-cell communication and have the potential to uncover novel protein signatures. In our research, we developed a novel pipeline to isolate, characterize, and profile EVs from normal bone and osteosarcoma tissue explants from canine OS patients. Proteomic analysis of vesicle preparations revealed a protein signature related to protein metabolism. One molecule of interest, PSMD14/Rpn11, was explored further given its prognostic potential in human and canine OS, and its targetability with the drug capzimin. In vitro experiments demonstrated that capzimin induces apoptosis and reduces clonogenic survival, proliferation, and migration in two metastatic canine OS cell lines. Capzimin also reduces the viability of metastatic human OS cells cultured under 3D conditions that mimic the growth of OS cells at secondary sites. This unique pipeline can improve our understanding of OS biology and identify new prognostic markers and molecular targets for both canine and human OS patients.


Subject(s)
Bone Neoplasms , Extracellular Vesicles , Osteosarcoma , Animals , Bone Neoplasms/metabolism , Cell Line, Tumor , Cell Proliferation , Dogs , Extracellular Vesicles/metabolism , Humans , Osteosarcoma/metabolism , Proteasome Endopeptidase Complex/metabolism , Proteomics , Trans-Activators/metabolism
2.
Pharmaceuticals (Basel) ; 14(10)2021 Sep 25.
Article in English | MEDLINE | ID: mdl-34681195

ABSTRACT

Osteosarcoma (OS) is an aggressive mesenchymal cell tumor that carries a poor long-term prognosis. Despite definitive surgery for the primary tumor and adjuvant chemotherapy, pulmonary metastasis is common and is the primary cause of morbidity. To improve outcomes for patients, we have developed and optimized a phenotypic screen for drugs that may target OS disseminated tumor cells (DTCs) and inhibit their metastatic outbreak rather than merely screening for cytotoxic activity against proliferating cells, as is commonly conducted in conventional drug discovery approaches. We report on the validation of a previously described 3D reconstituted basement membrane extract (3D BME) model system for tumor dormancy and metastatic outgrowth adapted to clonal pairs of high and low metastatic OS cells. A post-hoc validation of the assay was possible by comparing the activity of a drug in our assay with early evidence of activity in human OS clinical trials (regorafenib and saracatinib). In this validation, we found concordance between our assay and human clinical trial experience We then explored an approved veterinary small molecule inhibitor of Janus kinase-1 (oclacitinib) as a potential drug candidate to take advantage of the high prevalence of OS in pet dogs and its translational value to humans. Despite the biological rationale, we found no evidence to support the use of oclacitinib as an antimetastatic agent in OS. The findings support our 3D BME assay as a highly efficient method to examine drugs for activity in targeting OS DTCs.

3.
Front Oncol ; 11: 659963, 2021.
Article in English | MEDLINE | ID: mdl-33987095

ABSTRACT

Cancer recurrence remains a great fear for many cancer survivors following their initial, apparently successful, therapy. Despite significant improvement in the overall survival of many types of cancer, metastasis accounts for ~90% of all cancer mortality. There is a growing understanding that future therapeutic practices must accommodate this unmet medical need in preventing metastatic recurrence. Accumulating evidence supports dormant disseminated tumor cells (DTCs) as a source of cancer recurrence and recognizes the need for novel strategies to target these tumor cells. This review presents strategies to target dormant quiescent DTCs that reside at secondary sites. These strategies aim to prevent recurrence by maintaining dormant DTCs at bay, or eradicating them. Various approaches are presented, including: reinforcing the niche where dormant DTCs reside in order to keep dormant DTCs at bay; promoting cell intrinsic mechanisms to induce dormancy; preventing the engagement of dormant DTCs with their supportive niche in order to prevent their reactivation; targeting cell-intrinsic mechanisms mediating long-term survival of dormant DTCs; sensitizing dormant DTCs to chemotherapy treatments; and, inhibiting the immune evasion of dormant DTCs, leading to their demise. Various therapeutic approaches, some of which utilize drugs that are already approved, or have been tested in clinical trials and may be considered for repurposing, will be discussed. In addition, clinical evidence for the presence of dormant DTCs will be reviewed, along with potential prognostic biomarkers to enable the identification and stratification of patients who are at high risk of recurrence, and who could benefit from novel dormant DTCs targeting therapies. Finally, we will address the shortcomings of current trial designs for determining activity against dormant DTCs and provide novel approaches.

4.
BMC Genomics ; 17 Suppl 2: 444, 2016 06 23.
Article in English | MEDLINE | ID: mdl-27357693

ABSTRACT

BACKGROUND: Next generation sequencing (NGS) provides a key technology for deciphering the genetic underpinnings of human diseases. Typical NGS analyses of a patient depict tens of thousands non-reference coding variants, but only one or very few are expected to be significant for the relevant disorder. In a filtering stage, one employs family segregation, rarity in the population, predicted protein impact and evolutionary conservation as a means for shortening the variation list. However, narrowing down further towards culprit disease genes usually entails laborious seeking of gene-phenotype relationships, consulting numerous separate databases. Thus, a major challenge is to transition from the few hundred shortlisted genes to the most viable disease-causing candidates. RESULTS: We describe a novel tool, VarElect ( http://ve.genecards.org ), a comprehensive phenotype-dependent variant/gene prioritizer, based on the widely-used GeneCards, which helps rapidly identify causal mutations with extensive evidence. The GeneCards suite offers an effective and speedy alternative, whereby >120 gene-centric automatically-mined data sources are jointly available for the task. VarElect cashes on this wealth of information, as well as on GeneCards' powerful free-text Boolean search and scoring capabilities, proficiently matching variant-containing genes to submitted disease/symptom keywords. The tool also leverages the rich disease and pathway information of MalaCards, the human disease database, and PathCards, the unified pathway (SuperPaths) database, both within the GeneCards Suite. The VarElect algorithm infers direct as well as indirect links between genes and phenotypes, the latter benefitting from GeneCards' diverse gene-to-gene data links in GenesLikeMe. Finally, our tool offers an extensive gene-phenotype evidence portrayal ("MiniCards") and hyperlinks to the parent databases. CONCLUSIONS: We demonstrate that VarElect compares favorably with several often-used NGS phenotyping tools, thus providing a robust facility for ranking genes, pointing out their likelihood to be related to a patient's disease. VarElect's capacity to automatically process numerous NGS cases, either in stand-alone format or in VCF-analyzer mode (TGex and VarAnnot), is indispensable for emerging clinical projects that involve thousands of whole exome/genome NGS analyses.


Subject(s)
Computational Biology/methods , High-Throughput Nucleotide Sequencing/methods , Algorithms , Data Mining , Databases, Genetic , Genome, Human , Humans , Phenotype
5.
Curr Protoc Bioinformatics ; 54: 1.30.1-1.30.33, 2016 06 20.
Article in English | MEDLINE | ID: mdl-27322403

ABSTRACT

GeneCards, the human gene compendium, enables researchers to effectively navigate and inter-relate the wide universe of human genes, diseases, variants, proteins, cells, and biological pathways. Our recently launched Version 4 has a revamped infrastructure facilitating faster data updates, better-targeted data queries, and friendlier user experience. It also provides a stronger foundation for the GeneCards suite of companion databases and analysis tools. Improved data unification includes gene-disease links via MalaCards and merged biological pathways via PathCards, as well as drug information and proteome expression. VarElect, another suite member, is a phenotype prioritizer for next-generation sequencing, leveraging the GeneCards and MalaCards knowledgebase. It automatically infers direct and indirect scored associations between hundreds or even thousands of variant-containing genes and disease phenotype terms. VarElect's capabilities, either independently or within TGex, our comprehensive variant analysis pipeline, help prepare for the challenge of clinical projects that involve thousands of exome/genome NGS analyses. © 2016 by John Wiley & Sons, Inc.


Subject(s)
Data Mining/methods , Databases, Genetic , Genomics/methods , Sequence Analysis/methods , High-Throughput Nucleotide Sequencing , Humans , Phenotype , Proteome , Software/standards
6.
OMICS ; 20(3): 139-51, 2016 Mar.
Article in English | MEDLINE | ID: mdl-26983021

ABSTRACT

Postgenomics data are produced in large volumes by life sciences and clinical applications of novel omics diagnostics and therapeutics for precision medicine. To move from "data-to-knowledge-to-innovation," a crucial missing step in the current era is, however, our limited understanding of biological and clinical contexts associated with data. Prominent among the emerging remedies to this challenge are the gene set enrichment tools. This study reports on GeneAnalytics™ ( geneanalytics.genecards.org ), a comprehensive and easy-to-apply gene set analysis tool for rapid contextualization of expression patterns and functional signatures embedded in the postgenomics Big Data domains, such as Next Generation Sequencing (NGS), RNAseq, and microarray experiments. GeneAnalytics' differentiating features include in-depth evidence-based scoring algorithms, an intuitive user interface and proprietary unified data. GeneAnalytics employs the LifeMap Science's GeneCards suite, including the GeneCards®--the human gene database; the MalaCards-the human diseases database; and the PathCards--the biological pathways database. Expression-based analysis in GeneAnalytics relies on the LifeMap Discovery®--the embryonic development and stem cells database, which includes manually curated expression data for normal and diseased tissues, enabling advanced matching algorithm for gene-tissue association. This assists in evaluating differentiation protocols and discovering biomarkers for tissues and cells. Results are directly linked to gene, disease, or cell "cards" in the GeneCards suite. Future developments aim to enhance the GeneAnalytics algorithm as well as visualizations, employing varied graphical display items. Such attributes make GeneAnalytics a broadly applicable postgenomics data analyses and interpretation tool for translation of data to knowledge-based innovation in various Big Data fields such as precision medicine, ecogenomics, nutrigenomics, pharmacogenomics, vaccinomics, and others yet to emerge on the postgenomics horizon.


Subject(s)
Computational Biology/methods , Gene Regulatory Networks , Genome, Human , High-Throughput Nucleotide Sequencing/statistics & numerical data , Microarray Analysis/statistics & numerical data , Software , Algorithms , Data Mining , Databases, Factual , Databases, Genetic , Humans , Metabolic Networks and Pathways/genetics
7.
Regen Med ; 9(5): 649-72, 2014.
Article in English | MEDLINE | ID: mdl-25372080

ABSTRACT

Cell therapies aim to repair the mechanisms underlying disease initiation and progression, achieved through trophic effect or by cell replacement. Multiple cell types can be utilized in such therapies, including stem, progenitor or primary cells. This review covers the current state of cell therapies designed for the prominent disorders, including cardiovascular, neurological (Parkinson's disease, amyotrophic lateral sclerosis, stroke, spinal cord injury), autoimmune (Type 1 diabetes, multiple sclerosis, Crohn's disease), ophthalmologic, renal, liver and skeletal (osteoarthritis) diseases. Various cell therapies have reached advanced clinical trial phases with potential marketing approvals in the near future, many of which are based on mesenchymal stem cells. Advances in pluripotent stem cell research hold great promise for regenerative medicine. The information presented in this review is based on the analysis of the cell therapy collection detailed in LifeMap Discovery(®) (LifeMap Sciences Inc., USA) the database of embryonic development, stem cell research and regenerative medicine.


Subject(s)
Cell- and Tissue-Based Therapy/trends , Regenerative Medicine/trends , Stem Cell Transplantation , Autoimmune Diseases/therapy , Humans , Kidney Diseases/therapy , Liver Diseases/therapy , Nervous System Diseases/therapy , Osteoarthritis/therapy , Spinal Cord Injuries/therapy , Vascular Diseases/therapy
8.
PLoS One ; 8(7): e66629, 2013.
Article in English | MEDLINE | ID: mdl-23874394

ABSTRACT

LifeMap Discovery™ provides investigators with an integrated database of embryonic development, stem cell biology and regenerative medicine. The hand-curated reconstruction of cell ontology with stem cell biology; including molecular, cellular, anatomical and disease-related information, provides efficient and easy-to-use, searchable research tools. The database collates in vivo and in vitro gene expression and guides translation from in vitro data to the clinical utility, and thus can be utilized as a powerful tool for research and discovery in stem cell biology, developmental biology, disease mechanisms and therapeutic discovery. LifeMap Discovery is freely available to academic nonprofit institutions at http://discovery.lifemapsc.com.


Subject(s)
Embryonic Development , Regenerative Medicine , Animals , Cell Differentiation , Data Mining , Gene Expression , Humans , Protein Biosynthesis , Stem Cells/cytology
9.
Chemosphere ; 69(3): 500-6, 2007 Sep.
Article in English | MEDLINE | ID: mdl-17555789

ABSTRACT

Previous work in this laboratory has confirmed that the bacteria Mycobacterium sp. strain RJGII.135 and Sphingomonas yanoikuyae strain B1 and the green alga Selanastrum capricornutum strain UTEX 1648 degrade benzo[a]pyrene (BaP) to various BaP intermediates. S. capricornutum was first grown with BaP for 4 days. The organic extract of this media was then introduced into separate cultures of strain RJGII.135 and strain B1; separate cultures were grown with BaP for comparison. Cultures grown with BaP and those grown with the algal/BaP extract showed similar mineralization patterns. The quantity of total metabolites formed was greater in bacterial cultures grown with the algal/BaP extract than those grown with BaP alone. For strain RJGII.135, only 27% of the original BaP remained in cultures grown with the algal/BaP extract; 59% remained in cultures grown with BaP. For strain B1, only 6% of the original BaP remained in cultures grown with the algal/BaP extract; 38% remained in cultures grown with BaP. These results indicate that strategies utilizing organisms together may be necessary in being able to degrade large, recalcitrant polycyclic aromatic hydrocarbons (PAHs) such as BaP.


Subject(s)
Benzo(a)pyrene/metabolism , Chlorophyta/metabolism , Mycobacterium/metabolism , Chromatography, High Pressure Liquid , Oxidation-Reduction
10.
Dev Cell ; 12(1): 57-71, 2007 Jan.
Article in English | MEDLINE | ID: mdl-17199041

ABSTRACT

X chromosome inactivation (XCI) is initiated by expression of the noncoding Xist RNA in the female embryo. Tsix, the antisense noncoding partner of Xist, serves as its regulator during both imprinted and random XCI. Here, we show that Tsix in part acts through a 34mer repeat, DXPas34. DXPas34 contains bidirectional promoter activity, producing overlapping forward and reverse transcripts. We generate three new Tsix alleles in mouse embryonic stem cells and show that, while the Tsix promoter is unexpectedly dispensable, DXPas34 plays dual positive-negative functions. At the onset of XCI, DXPas34 stimulates Tsix expression through its enhancer activity. Once XCI is established, DXPas34 becomes repressive and stably silences Tsix. Germline transmission of the DXPas34 mutation demonstrates its necessity for both random and imprinted XCI in mice. Intriguingly, sequence analysis suggests that DXPas34 could potentially have descended from an ancient retrotransposon. We hypothesize that DXPas34 was acquired by Tsix to regulate antisense function.


Subject(s)
Genomic Imprinting , RNA, Untranslated/genetics , Repetitive Sequences, Nucleic Acid/genetics , X Chromosome Inactivation/genetics , Animals , Base Sequence , Consensus Sequence , Down-Regulation , Embryo, Mammalian/cytology , Embryo, Mammalian/embryology , Embryo, Mammalian/metabolism , Embryonic Stem Cells/metabolism , Female , Gene Expression Regulation, Developmental , Gene Targeting , In Situ Hybridization, Fluorescence , Mice , Models, Genetic , Molecular Sequence Data , Phylogeny , Promoter Regions, Genetic/genetics , RNA, Long Noncoding , Sequence Deletion , Up-Regulation , X Chromosome/genetics
11.
Toxicol Lett ; 162(2-3): 246-9, 2006 Apr 10.
Article in English | MEDLINE | ID: mdl-16256281

ABSTRACT

UNLABELLED: Studies of the impact of phase 1 enzyme polymorphisms on genetic damage have yielded mixed results. We studied how genetic damage would be altered when specific genes were ablated under low dose conditions. METHODS: Knockouts (KO) were generated from c57bl6/J mice with mutations in Cyp1a2 or Ahr receptor that eliminated gene product function. Animals were treated topically with either 4-aminobiphenyl (4ABP) 10mg/kg, benzo(a)pyrene (BaP) 33.3mg/kg or dibenzo(c,g)carbazole (DBC) 8 mg/kg, and sacrificed after 24h. DNA from livers, skin and/or urinary bladders were isolated and (32)P-post labelled. RESULTS: Cyp1a2-/- mice did not differ in 4ABP DNA adduct levels in either urinary bladder or liver compared to wildtype. There was a sex difference in the organ affected. Cyp1a2 knockout reduced skin BAP adduct levels 50% and AHR knockout reduced skin BAP adduct levels by 90%. There was no impact of either knockout on the levels of DBC-DNA adducts in any tissue. CONCLUSIONS: Ablation of specific metabolizing enzymes had compound- and tissue-specific effects in mice. Phenotypic variability in single CYP enzymes may have minor impact in humans at low doses, but variation in the ability to induce the family of CYPs may have a greater impact.


Subject(s)
Carcinogens/toxicity , Cytochrome P-450 CYP1A2/genetics , DNA Adducts , Receptors, Aryl Hydrocarbon/genetics , Aminobiphenyl Compounds/toxicity , Animals , Basic Helix-Loop-Helix Transcription Factors , Benzo(a)pyrene/toxicity , Carbazoles/toxicity , Cytochrome P-450 CYP1A2/deficiency , Environmental Monitoring , Genetic Predisposition to Disease , Genetic Variation , Liver/drug effects , Liver/metabolism , Lung/drug effects , Lung/metabolism , Mice , Mice, Inbred C57BL , Mice, Knockout , Polychlorinated Dibenzodioxins/toxicity , Receptors, Aryl Hydrocarbon/deficiency , Skin/drug effects , Skin/metabolism
12.
Toxicol Appl Pharmacol ; 206(1): 73-93, 2005 Aug 01.
Article in English | MEDLINE | ID: mdl-15963346

ABSTRACT

Polycyclic aromatic hydrocarbons (PAHs) and heterocyclic aromatic compounds (HACs) constitute a major class of chemical carcinogens present in the environment. These compounds require activation to electrophilic metabolites to exert their mutagenic or carcinogenic effects. There are three principal pathways currently proposed for metabolic activation of PAH and HAC: the pathway via bay region dihydrodiol epoxide by cytochrome P450 enzymes (CYPs), the pathway via radical cation by one-electron oxidation, and the ortho-quinone pathway by dihydrodiol dehydrogenase (DD). In addition to these major pathways, a brief description of a minor metabolic activation pathway, sulfonation, for PAHs that contain a primary benzylic alcoholic group or secondary hydroxyl group(s) is included in this review. The DNA damages caused through the reactive metabolites of PAH/HAC are described involving the DNA covalent binding to form stable or depurinating adducts, the formation of apurinic sites, and the oxidative damage. The review emphasizes the chemical/biochemical reactions involved in the metabolic processes and the chemical structures of metabolites and DNA adducts.


Subject(s)
Carcinogens/metabolism , DNA Adducts/biosynthesis , Epoxy Compounds/metabolism , Heterocyclic Compounds/metabolism , Polycyclic Aromatic Hydrocarbons/metabolism , Animals , Biotransformation , Carcinogens/pharmacokinetics , Epoxy Compounds/chemical synthesis , Epoxy Compounds/pharmacokinetics , Heterocyclic Compounds/pharmacokinetics , Polycyclic Aromatic Hydrocarbons/pharmacokinetics , Stereoisomerism , Structure-Activity Relationship
13.
Pathobiology ; 71(5): 253-60, 2004.
Article in English | MEDLINE | ID: mdl-15459484

ABSTRACT

OBJECTIVE: In the 7,12-dimethylbenz[a]anthracene (DMBA) model of rat mammary carcinogenesis, microvascular density and angiogenic potential increase with progression from normal to invasive disease, but the mechanisms involved are unknown. Using RT-PCR, we determined the expression of angiogenic regulators in DMBA-induced intraductal hyperplasia (IDP), carcinoma in situ (CIS), invasive tumors (INV), as well as normal tissue. METHODS: RT-PCR was performed on frozen tissue sections of each type of pathology for factors known to regulate angiogenesis in other systems. RESULTS: MMP-2, MMP-9, uPA, PAI-1, IGF-2, BFGF, VEGF, ANG-1, IRS-1, and TSP-1 were significantly (p < or =0.05) upregulated in CIS and INV, whereas TIMP-1, ANG-2, MASPIN, IGF1-R and HBEGF were unchanged. IGF-1 was uniquely elevated in IDP. SPARC was downregulated in CIS. Inhibition of IGF-1R by the tyrphostin, AG1024, blocked endothelial tubulogenesis in vitro, confirming that IGF-1 functions as a regulator of angiogenesis. CONCLUSIONS: These data support the involvement of specific angiogenic mediators in mammary tumor formation. Angiogenesis at different stages of tumorigenesis may be regulated by unique factors.


Subject(s)
9,10-Dimethyl-1,2-benzanthracene/toxicity , Angiogenic Proteins/biosynthesis , Carcinogens/toxicity , Carcinoma in Situ/chemically induced , Carcinoma, Ductal/chemically induced , Gene Expression Regulation/drug effects , Mammary Glands, Animal/drug effects , Mammary Neoplasms, Experimental/chemically induced , Angiogenic Proteins/genetics , Angiopoietin-1/biosynthesis , Angiopoietin-1/genetics , Animals , Carcinoma in Situ/genetics , Carcinoma, Ductal/genetics , Female , Fibroblast Growth Factor 2/biosynthesis , Fibroblast Growth Factor 2/genetics , Gene Expression Profiling , Hyperplasia , Insulin Receptor Substrate Proteins , Insulin-Like Growth Factor I/biosynthesis , Insulin-Like Growth Factor I/genetics , Insulin-Like Growth Factor I/physiology , Insulin-Like Growth Factor II/biosynthesis , Insulin-Like Growth Factor II/genetics , Mammary Glands, Animal/metabolism , Mammary Neoplasms, Experimental/genetics , Matrix Metalloproteinase 2/biosynthesis , Matrix Metalloproteinase 2/genetics , Matrix Metalloproteinase 9/biosynthesis , Matrix Metalloproteinase 9/genetics , Osteonectin/genetics , Osteonectin/physiology , Phosphoproteins/biosynthesis , Phosphoproteins/genetics , Plasminogen Activator Inhibitor 1/biosynthesis , Plasminogen Activator Inhibitor 1/genetics , Rats , Rats, Sprague-Dawley , Reverse Transcriptase Polymerase Chain Reaction , Thrombospondin 1/biosynthesis , Thrombospondin 1/genetics , Urokinase-Type Plasminogen Activator/biosynthesis , Urokinase-Type Plasminogen Activator/genetics , Vascular Endothelial Growth Factor A/biosynthesis , Vascular Endothelial Growth Factor A/genetics
14.
Lab Invest ; 84(8): 989-98, 2004 Aug.
Article in English | MEDLINE | ID: mdl-15170218

ABSTRACT

Preinvasive mammary pathologies in humans and rat chemical carcinogenesis model systems have an increased microvascular density relative to normal tissue. This suggests the possibility of preventing invasive breast cancer by inhibiting angiogenesis. Vascular endothelial cell growth factor (VEGF) is a potent angiogenic growth factor, commonly involved in tumor-induced angiogenesis. Here, we show that both VEGF and VEGFR2 expression increase with histological progression to invasive disease in the rat 7,12-dimethylbenz[a]anthracene (DMBA) model. Other VEGF receptors, VEGFR1, neuropilin 1 and neuropilin 2, are constitutively expressed throughout progression. To examine whether VEGF signaling is functionally relevant to tumor-induced endothelial tubule formation in vitro and for tumor formation in vivo, we utilized the VEGFR2 inhibitor, ZD6474. In vitro endothelial cell tubulogenesis induced by isolated mammary organoids or carcinoma in situ from DMBA-treated rats is inhibited by ZD6474, in a dose-dependent fashion. The administration of ZD6474 to DMBA-treated rats inhibits the formation of atypical ductal hyperplasia and carcinoma in situ by greater than 95% (P < 0.05), when administered 1 week or 6 weeks post-DMBA initiation. Invasive disease was absent in all ZD6474 cohorts. These data support the hypothesis that progression of DMBA-induced preinvasive mammary pathologies to palpable disease requires angiogenesis via a VEGF-dependent mechanism.


Subject(s)
Mammary Neoplasms, Experimental/prevention & control , Vascular Endothelial Growth Factor Receptor-2/antagonists & inhibitors , 9,10-Dimethyl-1,2-benzanthracene/toxicity , Animals , Base Sequence , Carcinoma in Situ/blood supply , Carcinoma in Situ/chemically induced , Carcinoma in Situ/pathology , Carcinoma in Situ/prevention & control , DNA Primers/genetics , Female , Humans , Hyperplasia , Mammary Neoplasms, Experimental/blood supply , Mammary Neoplasms, Experimental/chemically induced , Mammary Neoplasms, Experimental/pathology , Neovascularization, Pathologic/prevention & control , Piperidines/pharmacology , Quinazolines/pharmacology , Rats , Rats, Sprague-Dawley , Vascular Endothelial Growth Factor Receptor-2/genetics , Vascular Endothelial Growth Factor Receptor-2/physiology
15.
Lab Invest ; 83(7): 1001-11, 2003 Jul.
Article in English | MEDLINE | ID: mdl-12861040

ABSTRACT

In many women pathologic lesions, such as hyperplasia and carcinoma in situ, precede invasive breast cancer. We have shown that tissue vascularity increases with histologic progression to invasive disease. Similarly, in the well-characterized 7,12-dimethylbenz[a]anthracene (DMBA) model of mammary tumorigenesis, preinvasive lesions exhibit increased vascularity with progression. Using this model we asked whether inhibition of angiogenesis would block progression and if so, at which stage. We treated rats with DMBA followed by the potent angiogenic inhibitor, TNP-470, and/or tamoxifen starting 1 day or 6 weeks later. Histopathology and in vitro angiogenic potential of mammary organoids were evaluated 3 months after DMBA. All statistical tests were two-sided. Early TNP-470 and tamoxifen treatment inhibited the formation of carcinoma in situ (p < 0.001) and invasive disease (p < 0.001). However, their effects were not additive, despite their unique mechanisms of action. TNP-470 administration begun at the time of microscopic carcinoma in situ formation was unable to prevent the further development of carcinoma in situ or invasive breast cancer, whereas tamoxifen was highly effective (p = 0.001). There was no added benefit of combining TNP-470 and tamoxifen. TNP-470 therapy, unlike tamoxifen, did not inhibit the angiogenic potential of DMBA-treated normal mammary organoids, supporting its lack of a direct effect on the epithelium. These data provide proof-in-principle that inhibition of angiogenesis early in mammary tumorigenesis prevents mammary tumor formation in a hormone-sensitive model, indicating that angiogenesis is a potential target for cancer chemoprevention. Interactions with other chemopreventive strategies and the timing of administration must be thoroughly examined in vivo.


Subject(s)
Angiogenesis Inhibitors/therapeutic use , Antibiotics, Antineoplastic/therapeutic use , Carcinoma in Situ/prevention & control , Carcinoma, Intraductal, Noninfiltrating/prevention & control , Mammary Neoplasms, Experimental/prevention & control , Precancerous Conditions/prevention & control , Sesquiterpenes/therapeutic use , Tamoxifen/therapeutic use , 9,10-Dimethyl-1,2-benzanthracene/toxicity , Animals , Carcinogens/toxicity , Carcinoma in Situ/blood supply , Carcinoma in Situ/pathology , Carcinoma, Intraductal, Noninfiltrating/blood supply , Carcinoma, Intraductal, Noninfiltrating/pathology , Cyclohexanes , Disease Models, Animal , Drug Interactions , Female , Mammary Neoplasms, Experimental/blood supply , Mammary Neoplasms, Experimental/chemically induced , Mammary Neoplasms, Experimental/pathology , Neovascularization, Pathologic , O-(Chloroacetylcarbamoyl)fumagillol , Precancerous Conditions/blood supply , Precancerous Conditions/pathology , Rats , Rats, Sprague-Dawley
16.
Toxicol Lett ; 139(1): 11-23, 2003 Mar 20.
Article in English | MEDLINE | ID: mdl-12595154

ABSTRACT

Arylamine N-acetyltransferase (NAT) enzymes catalyze the addition of an acetyl group from acetyl-CoA to a terminal nitrogen on a suitable substrate such as environmentally relevant compounds and pharmaceuticals. In human, there are two highly polymorphic active allozymes, NAT1 and -2, and one inactive pseudogene, NATP. The expression of these enzymes is tissue-specific such that NAT1 is ubiquitously expressed and NAT2 is confined mainly to liver and colorectal tissues. We hypothesized that these genes would be tissue-specifically transcriptionally regulated, and so we isolated putative proximal control regions for both the NAT genes, which were inserted into luciferase vectors and transiently transfected into human liver and bladder cells. The transfected cells were dosed with 4-aminosalicylic acid, sulfamethazine or solvent and the resulting luciferase activity was measured. We found that both NAT1 and -2 regions were inducible in liver cells by both xenobiotics but only one of the NAT1 regions was inducible again by both xenobiotics in bladder cells. These results suggest that the NAT genes may be tissue-specifically transcriptionally regulated.


Subject(s)
Aminosalicylic Acid/pharmacology , Arylamine N-Acetyltransferase/genetics , Sulfamethazine/pharmacology , Xenobiotics/pharmacology , Base Sequence , Cell Line , Enzyme Induction/drug effects , Genes, Reporter/genetics , Humans , Molecular Sequence Data , Promoter Regions, Genetic/genetics
17.
J Toxicol Environ Health A ; 65(23): 2029-52, 2002 Dec 13.
Article in English | MEDLINE | ID: mdl-12490046

ABSTRACT

Manufactured gas plant (MGP) sites are contaminated with coal tar and may contain metals such as arsenic (As), cadmium (Cd), chromium (Cr), nickel (Ni), and vanadium (V). These metals are known to cause cancer or other adverse health conditions in humans, and the extent and cost of remediating MGP sites may be influenced by the presence of these metals. Studies assessed the distribution of these metals in female B6C3F1 mice ingesting (1) a metal mixture in water or (2) an MGP mixture in NIH-31 feed. The highest metal levels were measured in the small intestine and kidneys of mice receiving the metal mixture in water. For mice receiving the metal mixture in water, levels of As, Cd, and Cr, in the small intestine, levels of As, Cd, Cr, and V in the kidneys, levels of As and Cd in the pancreas, and levels of Cr and V in the femur were significantly greater than controls at 4, 8, 12, 16, and 24 wk. Except for Ni levels in the small intestine and femur and Cr levels in the kidneys, levels of metals were much lower in mice administered the MGP mixture in feed. The highest concentrations of metals in mice ingesting the MGP mixture in feed were found in the small intestine and kidneys, but few were significantly greater than controls. Levels of As in the small intestine at 6 and 18 wk and levels of Cr in the kidneys at 12, 18, and 24 wk were significantly greater than in controls. The data suggest that tissue burdens in small intestine, kidneys, pancreas, and femur of arsenic, cadmium, chromium, and vanadium are less when metals are present as an MGP mixture in feed than as a mixture in water. The reduced distribution and accumulation of metals in the organs of mice ingesting the MGP mixture in feed compared to the levels in organs of mice ingesting the metal mixture in water suggests that metals may be less likely to accumulate in humans ingesting MGP mixtures, thereby presenting a lower overall human health risk. The data presented indicate that the matrix in which metals are present will affect the uptake of individual metals and the organ specificity.


Subject(s)
Arsenic/pharmacokinetics , Metals, Heavy/pharmacokinetics , Administration, Oral , Animal Feed , Animals , Coal Tar , Female , Humans , Industrial Waste , Intestine, Small/chemistry , Kidney/chemistry , Rats , Tissue Distribution , Water
18.
Chem Res Toxicol ; 15(7): 915-21, 2002 Jul.
Article in English | MEDLINE | ID: mdl-12119001

ABSTRACT

7H-dibenzo[c,g]carbazole (DBC) is a potent multispecies, multisite carcinogen present in the environment. The metabolic activation pathways of DBC are not completely known. It is hypothesized that DBC may be metabolically activated by oxidation to the reactive Michael acceptor o-quinones, which can form stable and depurinating DNA adducts. The synthesis of DBC-3,4-dione has been previously reported by this research group. In the present article, we describe the synthesis and chemical structural elucidation of nine DBC-nucleic acid adducts produced from reactions of DBC-3,4-dione with Ade, Cyt, 2'-deoxyguanosine (dGuo), 2'-deoxycytidine (dCyd), and Guo. Adducts were isolated from reaction mixtures by HPLC and analyzed using MS including elemental compositions and collision-activated dissociation (CAD), (1)H NMR, and two-dimensional chemical shift correlation spectroscopy (COSY) NMR. The adducts, 7-[3,4-dione-DBC-1-yl]-Ade, N4-[3,4-dione-DBC-1-yl]-Cyt, 5-[3,4-dione-DBC-1-yl]-Cyt, two conformational isomers of N2-[3,4-dihydroxy-DBC-1-yl]-dGuo, and two conformational isomers of N2-[3,4-dihydroxy-DBC-1-yl]-Guo, were characterized. Two adducts from reactions of DBC-3,4-dione with dCyd were identified by MS but not fully characterized by NMR due to instability of the adducts. Under similar conditions, the reactions of DBC-3,4-dione with Gua and 2'-deoxyadenosine (dAdo) did not result in an identifiable adduct. Liver DNA adducts from mice treated topically with DBC-3,4-dione (100 microg) in dimethyl sulfoxide/acetone (15/85, 100 microL) were identified with 32P-postlabeling. The major adduct chromatographically matched one of the adducts formed from livers of DBC-treated mouse (adduct 3) using identical conditions.


Subject(s)
Carbazoles/pharmacokinetics , Carcinogens, Environmental/pharmacokinetics , DNA Adducts/metabolism , DNA/metabolism , Nucleosides/metabolism , Administration, Topical , Animals , Biotransformation , Carbazoles/chemistry , Carcinogens, Environmental/chemistry , Chromatography, High Pressure Liquid , DNA/chemistry , DNA Adducts/chemistry , Female , Liver/metabolism , Magnetic Resonance Spectroscopy , Mice , Mice, Inbred Strains , Nucleosides/chemistry , Spectrometry, Mass, Electrospray Ionization
19.
Toxicol Lett ; 132(1): 71-9, 2002 Jun 07.
Article in English | MEDLINE | ID: mdl-12084622

ABSTRACT

Tamoxifen (TAM), an antiestrogenic compound, has been approved for the treatment of breast cancer in high risk women. TAM has been shown to be an effective agent for prevention of breast cancer in women of varying degrees of risk and has been proposed to be used prophylactically in women whose genetic background suggests a high risk for breast cancer. However, it is not known whether TAM given prophylactically will alter the response of women to carcinogens from common environmental exposures such as tobacco smoke. Therefore, we studied the effects of TAM pretreatment on mammary DNA adducts of the model carcinogen, 7,12-dimethylbenz[a]anthracene (DMBA), in the female rats to assess whether TAM would alter the adduct pattern of DMBA. TAM (0.3 mg/day) was given for 7 days prior to a single 20 mg DMBA gavage treatment that is considered a carcinogenic dose in the rat. At 7 days post-DMBA, there was a reduction in the major DMBA-DNA adduct and a significant reduction in the minor DMBA-DNA adduct in mammary glands (P=0.002) of TAM pretreated rats compared to control rats. These data indicate that TAM may alter either metabolic steps in the formation of DNA binding species and/or enhance adduct removal. These data suggest that TAM given to women prior to the development of breast cancer may modulate the impact of environmental exposures, for example, tobacco smoke. Furthermore, research is needed to determine if modulation will be positive, or negative as in the current study.


Subject(s)
9,10-Dimethyl-1,2-benzanthracene/metabolism , Anticarcinogenic Agents/pharmacology , Carcinogens/metabolism , DNA Adducts/metabolism , Mammary Glands, Animal/metabolism , Tamoxifen/pharmacology , Animals , Autoradiography , Biotransformation , DNA/genetics , DNA/isolation & purification , DNA Adducts/drug effects , Female , Hydrolysis , Mammary Glands, Animal/drug effects , Rats , Rats, Sprague-Dawley
20.
Environ Toxicol Chem ; 21(2): 253-9, 2002 Feb.
Article in English | MEDLINE | ID: mdl-11833792

ABSTRACT

Mycobacterium sp. strain RJGII-135 is capable of degrading a wide range of polycyclic aromatic hydrocarbons (PAHs), including benzo[a]pyrene (BaP). In this study, critical aspects of degradation were investigated, including compound uptake, relative rates of PAH degradation, and the effects of co-occurring PAH substrates on BaP degradation and mineralization to CO2. Mycobacterium sp. strain RJGII-135 was capable of degrading phenanthrene, anthracene, and pyrene at a 10- to 20-fold greater rate than benz[a]anthracene (BaA) and BaP. A significant amount of phenanthrene and pyrene, 30% and 10%, respectively, was completely mineralized, whereas less than 4% of anthracene, BaA, and BaP was mineralized. The PAH uptake assays demonstrated that high amounts of BaP and BaA, 81% and 75% of added compound, respectively, could be recovered from bacterial cell fractions after a 4-h incubation compared with pyrene (61%), anthracene (53%), and phenanthrene (47%). The half-saturation constant (Km) for pyrene was threefold lower for pyrene over BaP, suggesting that the degradation system in Mycobacterium sp. strain RJGII-135 has a higher affinity for pyrene, reaching maximal degradative activity at lower concentrations. No hybridization to dioxygenase gene probes nahAc, bphA1, or tolC1C2 was detected. Studies to investigate competition between different PAH substrates demonstrated that the rate of BaP metabolism was influenced by the presence of a second PAH substrate. The BaP metabolism was inhibited when coincubated with BaA, pyrene, and anthracene. Phenanthrene did not inhibit but enhanced BaP metabolism sixfold. These data suggest that induction effects of components of complex mixtures may be as important as competitive metabolism when assessing the ability of bacteria to effectively degrade high-molecular-weight PAHs in the environment.


Subject(s)
Benzo(a)pyrene/metabolism , Carcinogens/metabolism , Mycobacterium/physiology , Polycyclic Aromatic Hydrocarbons/chemistry , Biodegradation, Environmental , Drug Interactions , Kinetics , Minerals , Pyrenes/chemistry , Pyrenes/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL