Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 3.007
Filter
1.
Bioorg Chem ; 123: 105744, 2022 06.
Article in English | MEDLINE | ID: mdl-35349830

ABSTRACT

While interstrand crosslinks (ICLs) have been considered as one type of DNA damage in the past, there is mounting evidence suggesting that these highly cytotoxic lesions are processed differently by the cellular machinery depending upon the ICL structure. In this study, we examined the crosslinking ability of three mitomycins, the structure of the ICLs they produce and the cytotoxicity of the drugs toward three different cell lines. The drugs are: mitomycin C (1), decarbamoylmitomycin C (2), and a mitomycin-conjugate (3) whose mitosane moiety is linked to a N-methylpyrrole carboxamide. We found that, overall, both MC and compound 3 show strong similarities regarding their alkylation of DNA, while DMC alkylating behavior is markedly different. To gain further insight into the mode of action of these drugs, we performed high throughput gene expression and gene ontology analysis to identify gene expression and cellular pathways most impacted by each drug treatment in MCF-7 cell lines. We observed that the novel mitomycin derivative (3) specifically causes changes in the expression of genes encoding proteins involved in cell integrity and tissue structure. Further analysis using bioinformatics (IPA) indicated that the new derivative (3) displays a stronger downregulation of major signaling networks that regulate the cell cycle, DNA damage response and cell proliferation when compared to MC and DMC. Collectively, these findings demonstrate that cytotoxic mechanisms of all three drugs are complex and are not solely related to their crosslinking abilities or the structure of the ICLs they produce.


Subject(s)
DNA Adducts , Mitomycin , Alkylation , DNA/chemistry , DNA Damage , Humans , Mitomycin/chemistry , Mitomycin/pharmacology , Mitomycins/chemistry , Mitomycins/pharmacology
2.
Lasers Med Sci ; 36(2): 349-356, 2021 Mar.
Article in English | MEDLINE | ID: mdl-32435908

ABSTRACT

The purpose of this study is to compare two dacryocystorhinostomy (DCR) techniques in epiphora treatment. This study is a prospective randomized trial. Twenty-nine patients presenting persistent epiphora due to primary acquired nasolacrimal duct obstruction (PANDO) were included in the study. Two groups each consisting of 15 eyes were formed. Mechanical transnasal endoscopic DCR (MTE-DCR) was applied to the first group, while transcanalicular dacryocystorhinostomy with multidiode laser (TCML-DCR) techniques is employed in the second group. Follow-up is conducted in the first day, first week, and first month of the dacryocystorhinostomy which is followed by 4-month follow-up period, and results were compared using statistical methods. The main outcome measures were the elimination of epiphora and unrestricted flow of irrigated saline to the nose. Seven patients were male, 22 were female, and the mean age was 39.3 ± 12.5 years. Mean follow-up times were 111.3 ± 10.5 months and 93 ± 2.9 months in group 1 and group 2, respectively. Complete resolution is achieved in group 1, whereas failures stemming from canalicular stenosis and fibrosis at osteotomy site are recorded in two cases in group 2. Occlusion occurred in the fifth month in both cases. Thus, long-term success rates were 100% in the first and 86.6% in the second group (P = 0.483). MTE-DCR is a strong substitute for external DCR. Although TCML-DCR shows promising results, it is far away from becoming the gold standard technique in epiphora treatment.


Subject(s)
Dacryocystorhinostomy , Endoscopy , Lasers , Nose/surgery , Adult , Female , Humans , Lacrimal Duct Obstruction/pathology , Male , Mitomycins/pharmacology , Nasolacrimal Duct/surgery , Outcome Assessment, Health Care , Prospective Studies , Time Factors , Treatment Outcome
3.
Yonsei Med J ; 59(6): 727-735, 2018 Aug.
Article in English | MEDLINE | ID: mdl-29978609

ABSTRACT

PURPOSE: To investigate the effect of combined inhibition of protein kinase B (AKT) and SRC on the growth and metastatic potential of human pancreatic cancer cells. MATERIALS AND METHODS: AKT and SRC were inhibited using 10-DEBC and PP2, respectively. The expression of their messenger RNAs were down-regulated by specific small interfering RNA (siRNA). Changes in pancreatic cancer cell growth and metastatic potential were determined using a cell viability assay and a xenotransplant model of pancreatic cancer, as well as cell migration and invasion assays. Signal proteins were analyzed by Western blot. RESULTS: The inhibitors 10-DEBC and PP2 suppressed cell proliferation in a dose-dependent fashion in pancreatic cancer cell lines MIA PaCa-2 and PANC-1. The simultaneous inhibition of AKT and SRC at low concentrations resulted in a significant suppression of cell proliferation. Knockdown of AKT2 and SRC using siRNAs also significantly decreased cell proliferation. In a pancreatic cancer model, combined treatment with 10-DEBC and PP2 also significantly suppressed the growth of pancreatic cancer. Application of 10-DEBC with PP2 significantly reduced the metastatic potential of pancreatic cancer cells by inhibiting migration and invasion. The combined inhibition suppressed the phosphorylation of mTOR and ERK in pancreatic cancer cells. CONCLUSION: Combined targeting of AKT and SRC resulted in a synergistic efficacy against human pancreatic cancer growth and metastasis.


Subject(s)
Cell Movement/drug effects , Mitomycins/pharmacology , Pancreatic Neoplasms/drug therapy , Pancreatic Neoplasms/pathology , Protein Kinase Inhibitors/pharmacology , Proto-Oncogene Proteins c-akt/antagonists & inhibitors , src-Family Kinases/antagonists & inhibitors , Blotting, Western , Cell Line, Tumor , Cell Movement/genetics , Cell Proliferation , Cell Survival/drug effects , Drug Synergism , Humans , Molecular Targeted Therapy , Pancreatic Neoplasms/genetics , Pancreatic Neoplasms/metabolism , Phosphorylation/drug effects , Protein Kinase Inhibitors/administration & dosage , Protein-Tyrosine Kinases/antagonists & inhibitors , Protein-Tyrosine Kinases/genetics , Protein-Tyrosine Kinases/metabolism , Proto-Oncogene Proteins c-akt/drug effects , Proto-Oncogene Proteins c-akt/metabolism , RNA, Small Interfering/genetics , src-Family Kinases/genetics , src-Family Kinases/metabolism
4.
Curr Opin Pharmacol ; 41: 20-26, 2018 08.
Article in English | MEDLINE | ID: mdl-29679802

ABSTRACT

DNA crosslinking agents make up a broad class of chemotherapy agents that target rapidly dividing cancer cells by disrupting DNA synthesis. These drugs differ widely in both chemical structure and biological effect. In cells, crosslinking agents can form multiple types of DNA lesions with varying efficiencies. Inter-strand crosslinks (ICLs) are considered to be the most cytotoxic lesion, creating a covalent roadblock to replication and transcription. Despite over 50 years in the clinic, the use of crosslinking agents that specialize in the formation of ICLs remains limited, largely due to high toxicity in patients. Current ICL-based therapeutics have focused on late-stage and drug-resistant tumors, or localized treatments that limit exposure. In this article, we review the development of clinical crosslinking agents, our understanding of how cells respond to different lesions, and the potential to improve ICL-based chemotherapeutics in the future.


Subject(s)
Antineoplastic Agents/therapeutic use , Cross-Linking Reagents/therapeutic use , Neoplasms/drug therapy , Antineoplastic Agents/pharmacology , Cross-Linking Reagents/pharmacology , DNA/drug effects , Furocoumarins/pharmacology , Furocoumarins/therapeutic use , Humans , Mechlorethamine/analogs & derivatives , Mechlorethamine/therapeutic use , Mitomycins/pharmacology , Mitomycins/therapeutic use
5.
Bioorg Med Chem ; 23(23): 7378-85, 2015 Dec 01.
Article in English | MEDLINE | ID: mdl-26541587

ABSTRACT

Two synthetic aziridinomitosenes (AZMs), Me-AZM and H-AZM, structurally related to mitomycin C (MC) were evaluated for their anticancer activity against six cancer cell lines (HeLa, Jurkat, T47D, HepG2, HL-60, and HuT-78) and tested for their DNA-modifying abilities in Jurkat cells. Cytotoxicity assays showed that Me-AZM is up to 72-fold and 520-fold more potent than MC and H-AZM, respectively. Me-AZM also demonstrated increased DNA modification over MC and H-AZM in alkaline COMET and Hoechst fluorescence assays that measured crosslinks in cellular DNA. Me-AZM and H-AZM treatment of Jurkat cells was found to sponsor significant DNA-protein crosslinks using a K-SDS assay. The results clearly indicate that the AZM C6/C7 substitution pattern plays an important role in drug activity and supports both DNA-DNA and DNA-protein adduct formation as mechanisms for inducing cytotoxic effects.


Subject(s)
Antineoplastic Agents/pharmacology , Cross-Linking Reagents/pharmacology , DNA/metabolism , Mitomycins/pharmacology , Antineoplastic Agents/chemistry , Cell Line, Tumor , Cell Proliferation/drug effects , Cell Survival/drug effects , Comet Assay , Cross-Linking Reagents/chemistry , DNA Adducts/metabolism , Humans , Mitomycins/chemistry , Structure-Activity Relationship
6.
Cell Cycle ; 14(5): 744-54, 2015.
Article in English | MEDLINE | ID: mdl-25565400

ABSTRACT

Interstrand crosslinks induce DNA replication fork stalling that in turn activates the ATR-dependent checkpoint and DNA repair on nuclear chromatin. Mitomycin C (MC) and Decarbamoyl Mitomycin C (DMC) induce different types of DNA crosslinks with DMC being a more cytotoxic agent. We previously reported that the novel DMC induced ß-interstrand DNA crosslinks induce a p53-independent form of cell death. The p53-independent DMC cytotoxicity associates with the activation, and subsequent depletion, of Chk1. In this study we further dissect the novel DMC signal transduction pathway and asked how it influences chromatin-associated proteins. We found that treatment with DMC, but not MC, stimulated the disassociation of ATR from chromatin and re-localization of ATR to the cytoplasm. The chromatin eviction of ATR was coupled with the formation of nuclear Rad51 foci and the phosphorylation of Chk1. Furthermore, DMC but not MC, activated expression of gadd45α mRNA. Importantly, knocking down p53 via shRNA did not inhibit the DMC-induced disassociation of ATR from chromatin or reduce the activation of transcription of gadd45α. Our results suggest that DMC induces a p53-independent disassociation of ATR from chromatin that facilitates Chk1 checkpoint activation and Rad51 chromatin recruitment. Our findings provide evidence that ATR chromatin eviction in breast cancer cells is an area of study that should be focused on for inducing p53-independent cell death.


Subject(s)
Chromatin/metabolism , Mitomycins/pharmacology , Tumor Suppressor Protein p53/metabolism , Ataxia Telangiectasia Mutated Proteins/metabolism , Cell Cycle Proteins/metabolism , Cell Death/drug effects , Checkpoint Kinase 1 , DNA/metabolism , DNA Damage , Homologous Recombination/drug effects , Humans , MCF-7 Cells , Models, Biological , Nuclear Proteins/metabolism , Phosphorylation/drug effects , Phosphoserine/metabolism , Protein Binding/drug effects , Protein Kinases/metabolism , Rad51 Recombinase/metabolism , p38 Mitogen-Activated Protein Kinases/metabolism
7.
Angew Chem Int Ed Engl ; 53(35): 9302-5, 2014 Aug 25.
Article in English | MEDLINE | ID: mdl-25044229

ABSTRACT

A DNA crosslinking approach, which is distinct but related to the double alkylation by mitomycin C, involving a novel electrophilic spiro-cyclopropane intermediate is hypothesized. Rational design and substantial structural simplification permitted the expedient chemical synthesis and rapid discovery of MTSB-6, a mitomycin C analogue which is twice as potent as mitomycin C against the prostate cancer cells. MTSB-6 shows improvements in its selective action against noncancer prostate cells over mitomycin C. This hypothesis-driven discovery opens novel yet synthetically accessible mitosene structural space for discovering more potent and less toxic therapeutic candidates.


Subject(s)
Mitomycin/pharmacology , Mitomycins/chemistry , Mitomycins/pharmacology , Cell Line , Cell Survival/drug effects , Dose-Response Relationship, Drug , Humans , Inhibitory Concentration 50 , Mitomycin/chemistry , Mitomycins/chemical synthesis , Molecular Structure , Structure-Activity Relationship
8.
PLoS One ; 9(2): e88828, 2014.
Article in English | MEDLINE | ID: mdl-24586407

ABSTRACT

p53 is a transcription factor that regulates the response to cellular stress. Mammalian p53 functions as a tumor suppressor. The C. elegans p53, cep-1, regulates DNA-damage induced germline cell death by activating the transcription of egl-1 and ced-13. We used the C. elegans model to investigate how, in the whole animal, different forms of DNA damage can induce p53-dependent versus p53-independent cell death and DNA repair. DNA damage was induced by ultraviolet type C (UVC) radiation, or 10-decarbamoyl mitomycin C (DMC, an agent known to induce mammalian p53-independent cell death). Wild-type or cep-1 loss-of-function mutant animals were assayed for germline cell death and DNA lesions. Wild-type animals displayed greater removal of UVC-lesions over time, whereas cep-1 mutant animals displayed increased UVC-lesion retention. The cep-1 mutation increased UVC-lesion retention directly correlated with a reduction of progeny viability. Consistent with DMC inducing p53-independent cell death in mammalian cells DMC induced a C. elegans p53-independent germline cell death pathway. To examine the influence of wild-type CEP-1 and DNA damage on C. elegans tumors we used glp-1(ar202gf)/Notch germline tumor mutants. UVC treatment of glp-1 mutant animals activated the CEP-1 target gene egl-1 and reduced tumor size. In cep-1(gk138);glp-1(ar202gf) animals, UVC treatment resulted in increased susceptibility to lesions and larger tumorous germlines. Interestingly, the partial knockdown of bec-1 in adults resulted in a CEP-1-dependent increase in germline cell death and an increase in DNA damage. These results strongly support cross-talk between BEC-1 and CEP-1 to protect the C. elegans genome.


Subject(s)
Caenorhabditis elegans Proteins/metabolism , Caenorhabditis elegans/physiology , DNA Repair/physiology , Gene Expression Regulation/genetics , Tumor Suppressor Protein p53/metabolism , Vesicular Transport Proteins/metabolism , Animals , Apoptosis/drug effects , Apoptosis/physiology , Caenorhabditis elegans/genetics , DNA Damage/radiation effects , DNA Primers/genetics , DNA Repair/genetics , Germ Cells/drug effects , Germ Cells/physiology , Mitomycins/pharmacology , RNA Interference , Repressor Proteins/metabolism , Reverse Transcriptase Polymerase Chain Reaction , Ultraviolet Rays
10.
Bioorg Med Chem Lett ; 20(1): 31-4, 2010 Jan 01.
Article in English | MEDLINE | ID: mdl-19954979

ABSTRACT

The reductive activation of mitomycin C in aqueous bicarbonate buffer resulted in the formation of a previously unknown compound, characterized as an oxazolidinone derivative of cis-1-hydroxy-2,7-diaminomitosene. This compound is the result of a cyclization reaction of bicarbonate with the aziridine ring of aziridinomitosene, and was observed at bicarbonate concentrations close to those present in physiological plasma.


Subject(s)
Antibiotics, Antineoplastic/chemistry , Bicarbonates/chemistry , Mitomycin/chemistry , Mitomycins/chemistry , Oxazolidinones/chemistry , Antibiotics, Antineoplastic/metabolism , Aziridines/chemistry , Cyclization , Isomerism , Mitomycin/metabolism , Mitomycins/isolation & purification , Mitomycins/pharmacology , Oxidation-Reduction
11.
Anticancer Agents Med Chem ; 9(4): 369-80, 2009 May.
Article in English | MEDLINE | ID: mdl-19442038

ABSTRACT

The 8H-thieno[2,3-b]pyrrolizinones, some of which exert very potent cytotoxic activity against tumor cell lines in vitro, are a promising novel series of anti-cancer agents. These compounds belong to the tripentone family and are based on 9H-pyrrolo[1,2-a]indol-9-one derivatives and their heterocyclic isosteres. This paper inventories the different synthetic strategies for tripentones and reviews their biological effects and therapeutic potential.


Subject(s)
Antineoplastic Agents/chemical synthesis , Mitomycins/pharmacology , Animals , Antineoplastic Agents/pharmacology , Antineoplastic Agents/therapeutic use , Drug Screening Assays, Antitumor , Inhibitory Concentration 50 , Leukemia L1210/drug therapy , Mitomycins/chemical synthesis , Structure-Activity Relationship
12.
Chem Res Toxicol ; 21(12): 2370-8, 2008 Dec.
Article in English | MEDLINE | ID: mdl-19053323

ABSTRACT

The antitumor antibiotic and cancer chemotherapeutic agent mitomycin C (MC) alkylates and crosslinks DNA, forming six major MC-deoxyguanosine adducts of known structures in vitro and in vivo. Two of these adducts are derived from 2,7-diaminomitosene (2,7-DAM), a nontoxic reductive metabolite of MC formed in cells in situ. Several methods have been used for the analysis of MC-DNA adducts in the past; however, a need exists for a safer, more comprehensive and direct assay of the six-adduct complex. Development of an assay, based on mass spectrometry, is described. DNA from EMT6 mouse mammary tumor cells, Fanconi Anemia-A fibroblasts, normal human fibroblasts, and MCF-7 human breast cancer cells was isolated after MC or 10-decarbamoyl mitomycin C (DMC) treatment of the cells, digested to nucleosides, and submitted to liquid chromatography electrospray-tandem mass spectrometry. Two fragments of each parent ion were monitored ("multiple reaction monitoring"). Identification and quantitative analysis were based on a standard mixture of six adducts, the preparation of which is described here in detail. The lower limit of detection of adducts is estimated as 0.25 pmol. Three initial applications of this method are reported as follows: (i) differential kinetics of adduct repair in EMT6 cells, (ii) analysis of adducts in MC- or DMC-treated Fanconi Anemia cells, and (iii) comparison of the adducts generated by treatment of MCF-7 breast cancer cells with MC and DMC. Notable results are the following: Repair removal of the DNA interstrand cross-link and of the two adducts of 2,7-DAM is relatively slow; both MC and DMC generate DNA interstrand cross-links in human fibroblasts, Fanconi Anemia-A fibroblasts, and MCF-7 cells as well as EMT6 cells; and DMC shows a stereochemical preference of linkage to the guanine-2-amino group opposite from that of MC.


Subject(s)
Cross-Linking Reagents/pharmacology , DNA Adducts/biosynthesis , Mitomycin/pharmacology , Mitomycins/pharmacology , Spectrometry, Mass, Electrospray Ionization/methods , Tandem Mass Spectrometry/methods , Animals , Breast Neoplasms/drug therapy , Breast Neoplasms/genetics , Breast Neoplasms/metabolism , Cell Line, Tumor , Chromatography, High Pressure Liquid , Cross-Linking Reagents/chemistry , Cross-Linking Reagents/metabolism , DNA Adducts/chemistry , DNA Damage , DNA Repair , DNA, Neoplasm/chemistry , DNA, Neoplasm/drug effects , Fibroblasts/drug effects , Fibroblasts/metabolism , Humans , Mammary Neoplasms, Animal/drug therapy , Mammary Neoplasms, Animal/genetics , Mammary Neoplasms, Animal/metabolism , Mice , Mitomycin/chemistry , Mitomycin/metabolism , Mitomycins/chemistry , Mitomycins/metabolism
13.
Zhonghua Wei Chang Wai Ke Za Zhi ; 11(3): 276-9, 2008 May.
Article in Chinese | MEDLINE | ID: mdl-18478476

ABSTRACT

OBJECTIVE: To evaluate in vitro anti-tumor effect of chemotherapeutic drugs on human gastric cancer cells, and investigate the relationship with Bcl-2 expression. METHODS: Single cell suspension was prepared from fresh gastric cancer tissue and exposed to taxol (Tax), 5-fluorouracil (5-FU), cisplatin (CDDP), adriamycin (ADM), mitomycin (MMC) respectively for 48 hours. Metabolic activity and inhibitory rate of cells were detected by MTT assay. Expression of Bcl-2 was examined with immunohistochemistry. RESULTS: The inhibitory rates of cancer cells exposed to chemotherapeutic drugs were different and Tax, 5-FU, CDDP had remarkably higher rates than ADM and MMC. The lower differentiated gastric cancer cells were more sensitive than the higher ones. Positive expression rate of Bcl-2 was 80% and the positive cells showed resistance to 5-FU, ADM and MMC. CONCLUSIONS: Chemosensitive testing by MTT assay can constitute the prediction for the application of chemotherapeutic drugs individually. Overexpression of Bcl-2 may contribute to multiple drug-resistance of tumors.


Subject(s)
Antineoplastic Agents/pharmacology , Proto-Oncogene Proteins c-bcl-2/metabolism , Stomach Neoplasms/metabolism , Adult , Aged , Antineoplastic Agents/therapeutic use , Cell Survival , Cisplatin/pharmacology , Cisplatin/therapeutic use , Doxorubicin/pharmacology , Doxorubicin/therapeutic use , Drug Screening Assays, Antitumor , Female , Fluorouracil/pharmacology , Fluorouracil/therapeutic use , Humans , Male , Middle Aged , Mitomycin/pharmacology , Mitomycin/therapeutic use , Mitomycins/pharmacology , Mitomycins/therapeutic use , Paclitaxel/pharmacology , Paclitaxel/therapeutic use , Stomach Neoplasms/drug therapy , Stomach Neoplasms/pathology , Tumor Cells, Cultured
14.
Biomed Environ Sci ; 21(6): 492-8, 2008 Dec.
Article in English | MEDLINE | ID: mdl-19263805

ABSTRACT

OBJECTIVE: To determine the in vitro possible clastogenic and cytotoxic activities of Ulva rigida crude extracts (URE), and identify their antigenotoxic and protective effects on chemotherapeutic agent mitomycine-C (MMC). METHODS: Anti-clastogenic and anti-genotoxic activities of Ulva rigida crude extracts (URE) were studied using chromosome aberration (CA), sister chromatid exchange (SCE), and micronuclei (MN) tests in human lymphocytes cultured in vitro. RESULTS: The chromosome aberration, sister chromatid exchange or micronuclei tests showed that URE at concentrations of 10, 20, and 40 microg/mL had no clastogenic activity in human lymphocyte cell culture. Three doses of URE significantly decreased the number of chromosomal aberrations and the frequencies of SCE and MN when compared with the culture treated with MMC (P < 0.0001). CONCLUSION: Although URE itself is not a clastogenic or cytotoxic substance, it possesses strong antigenotoxic, anti-clastogenic, and protective effects on MMC in vitro.


Subject(s)
Antimutagenic Agents/pharmacology , Chromosome Aberrations/drug effects , Lymphocytes/drug effects , Mitomycins/pharmacology , Mutagens/toxicity , Plant Extracts/pharmacology , Sister Chromatid Exchange/drug effects , Antibiotics, Antineoplastic/pharmacology , Cells, Cultured , Chlorophyta , Dose-Response Relationship, Drug , Humans , Lymphocytes/metabolism , Micronucleus Tests , Plant Extracts/chemistry
15.
Drug Metab Dispos ; 35(12): 2262-9, 2007 Dec.
Article in English | MEDLINE | ID: mdl-17890447

ABSTRACT

The aryl hydrocarbon receptor repressor (AhRR) is a member of the aryl hydrocarbon receptor (AhR) signaling cascade, which mediates dioxin toxicity and is involved in regulation of cell growth and differentiation. The AhRR was described as a feedback modulator, which counteracts AhR-dependent gene expression. We investigated the molecular mechanisms of transcriptional regulation of the human AhRR by cloning its regulatory DNA region located in intron I of the AhRR. By means of reporter gene analyses and generation of deletion variants, we identified a functional, 3-methylcholanthrene-sensitive xenobiotic response element (XRE) site. Chromatin immunoprecipitation analyses revealed that the AhRR binds to this XRE, displaying an autoregulatory loop of AhRR expression. In addition we show that an adjacent GC-box is of functional relevance for AhRR transcription, since blocking of this GC-box resulted in a decrease of constitutive and inducible AhRR gene activity. The differences in constitutive AhRR mRNA level observed in HepG2, primary fibroblast, and HeLa cells are directly correlated with CYP1A1 inducibility. We show that the nonresponsiveness of high AhRR-expressing cells toward AhR-agonists is associated with a constitutive binding of the AhRR to XRE sites of CYP1A1. Treatment with the histone deacetylase inhibitor sodium butyrate restored the responsiveness of CYP1A1 in these cell lines, due to the dissociation of AhRR from the XREs. Furthermore, transient AhRR mRNA silencing in untreated HeLa cells was accompanied by an increase of basal CYP1A1 expression, pointing to a constitutive role of the AhRR in regulation of CYP1A1. The functional relevance of the AhRR in high AhRR-expressing primary fibroblasts is discussed.


Subject(s)
Cytochrome P-450 CYP1A1/genetics , Gene Expression Regulation, Enzymologic , Introns , Promoter Regions, Genetic , Repressor Proteins/genetics , Transcription, Genetic , Aryl Hydrocarbon Receptor Nuclear Translocator/metabolism , Basic Helix-Loop-Helix Transcription Factors , Butyrates/pharmacology , Cloning, Molecular , Cytochrome P-450 CYP1A1/metabolism , Enzyme Inhibitors/pharmacology , Fibroblasts/enzymology , Fibroblasts/metabolism , GC Rich Sequence , Genes, Reporter , HeLa Cells , Histone Deacetylase Inhibitors , Histone Deacetylases/metabolism , Humans , Luciferases , Methylcholanthrene/pharmacology , Mitomycins/pharmacology , Mutation , Promoter Regions, Genetic/drug effects , RNA Interference , RNA, Messenger/metabolism , RNA, Small Interfering/metabolism , Receptors, Aryl Hydrocarbon/agonists , Receptors, Aryl Hydrocarbon/metabolism , Repressor Proteins/metabolism , Transcription, Genetic/drug effects , Transfection
16.
ACS Chem Biol ; 2(6): 399-407, 2007 Jun 15.
Article in English | MEDLINE | ID: mdl-17530733

ABSTRACT

10-Decarbamoyl-mitomycin C (DMC), a mitomycin C (MC) derivative, generates an array of DNA monoadducts and interstrand cross-links stereoisomeric to those that are generated by MC. DMC was previously shown in our laboratory to exceed the cytotoxicity of MC in a human leukemia cell line that lacks a functional p53 pathway (K562). However, the molecular signal transduction pathway activated by DMCDNA adducts has not been investigated. In this study, we have compared molecular targets associated with signaling pathways activated by DMC and MC in several human cancer cell lines. In cell lines lacking wild-type p53, DMC was reproducibly more cytotoxic than MC, but it generated barely detectable signal transduction markers associated with apoptotic death. Strikingly, DMCs increased cytotoxicity was not associated with an increase in DNA double-strand breaks but was associated with early poly(ADP-ribose) polymerase (PARP) activation and Chk1 kinase depletion. Alkylating agents can induce increased PARP activity associated with programmed necrosis, and the biological activity of DMC in p53-null cell lines fits this paradigm. In cell lines with a functional p53 pathway, both MC and DMC induced apoptosis. In the presence of p53, both MC and DMC activate procaspases; however, the spectrum of procaspases involved differs for the two drugs, as does induction of p73. These studies suggest that in the absence of p53, signaling to molecular targets in cell death can shift in response to different DNA adduct structures to induce non-apoptotic cell death.


Subject(s)
DNA Adducts/physiology , Mitomycins/pharmacology , Signal Transduction/physiology , Tumor Suppressor Protein p53/physiology , Animals , Cell Death/drug effects , Cell Death/physiology , Cell Line, Tumor , DNA Adducts/genetics , Humans , Mice , Signal Transduction/drug effects
17.
Biochem Pharmacol ; 72(3): 366-76, 2006 Jul 28.
Article in English | MEDLINE | ID: mdl-16765324

ABSTRACT

NRH:quinone oxidoreductase 2 (NQO2) is a cytosolic flavoprotein that utilizes NRH as electron donor. The present studies investigate the role of NQO2 in metabolic detoxification/activation of quinones and quinone based anti-tumor drugs. Chinese hamster ovary (CHO) cells stably overexpressing cDNA derived mouse NQO2 and mouse keratinocytes from DMBA-induced skin tumors in wild-type and NQO2-null mice were generated. The CHO cells overexpressing NQO2 and mouse keratinocytes expressing or deficient in NQO2 were treated with varying concentrations of mitomycin C (MMC), CB1954, MMC analog BMY25067, EO9, menadione and BP-3,6-quinone, in the absence and presence of NRH. The cytotoxicity of the drugs was evaluated by colony formation. The CHO cells overexpressing higher levels of mouse NQO2 showed significantly increased cytotoxicity to menadione, BP-3,6-quinone and to the anti-tumor drugs MMC and CB1954 when compared to CHO cells expressing endogenous NQO2. The cytotoxicity increased in presence of NRH. Similar results were also observed with BMY25067 and EO9 treatments, but to a lesser extent. The results on keratinocytes deficient in NQO2 supported the data from CHO cells. The inclusion of NRH had no effect on cytotoxicity of quinones and drugs in keratinocytes deficient in NQO2. Mouse NQO2 protein was expressed in bacteria, purified and used to study the role of NQO2 in MMC-induced DNA cross-linking. Bacterially expressed and purified NQO2 efficiently catalyzed MMC activation that led to DNA cross-linking. These results concluded that NQO2 plays a significant role in the metabolic activation of both quinones and anti-tumor drugs leading to cytotoxicity and cell death.


Subject(s)
Antineoplastic Agents/pharmacology , Biotransformation/drug effects , Quinone Reductases/metabolism , Quinones/pharmacology , Animals , Antineoplastic Agents/metabolism , Aziridines/metabolism , Aziridines/pharmacology , Benzopyrenes/metabolism , Benzopyrenes/pharmacology , CHO Cells , Cell Survival/drug effects , Cells, Cultured , Cricetinae , Cricetulus , Cross-Linking Reagents/metabolism , Cross-Linking Reagents/pharmacology , DNA, Complementary/genetics , Dose-Response Relationship, Drug , Hydroquinones/metabolism , Hydroquinones/pharmacology , Indolequinones/metabolism , Indolequinones/pharmacology , Keratinocytes/cytology , Keratinocytes/drug effects , Keratinocytes/metabolism , Mice , Mice, Knockout , Mitomycin/metabolism , Mitomycin/pharmacology , Mitomycins/metabolism , Mitomycins/pharmacology , Quinone Reductases/genetics , Quinones/metabolism , Transfection , Vitamin K 3/metabolism , Vitamin K 3/pharmacology
18.
Clin Cancer Res ; 12(6): 1913-20, 2006 Mar 15.
Article in English | MEDLINE | ID: mdl-16551877

ABSTRACT

PURPOSE: A lipid-based prodrug of mitomycin C [MMC; 2,3-(distearoyloxy)propane-1-dithio-4'-benzyloxycarbonyl-MMC] was designed for liposome formulation. The purpose of this study was to examine the in vitro cytotoxicity, pharmacokinetics, in vivo toxicity, and in vivo antitumor activity of this new lipid-based prodrug formulated in polyethylene glycol-coated (pegylated) liposomes. EXPERIMENTAL DESIGN: MMC was released from the MMC lipid-based prodrug (MLP) by thiolytic-induced cleavage with a variety of thiol-containing reducing agents. MLP was incorporated with nearly 100% efficiency in cholesterol-free pegylated liposomes with hydrogenated phosphatidylcholine as the main component and a mean vesicle size of approximately 90 nm. This formulation was used for in vitro and in vivo tests in rodents. RESULTS: In vitro, the cytotoxic activity of pegylated liposomal MLP (PL-MLP) was drastically reduced compared with free MMC. However, in the presence of reducing agents, such as cysteine or N-acetyl-cysteine, its activity increased to nearly comparable levels to those of free MMC. Intravenous administration of PL-MLP in rats resulted in a slow clearance indicating stable prodrug retention in liposomes and long circulation time kinetics, with a pharmacokinetic profile substantially different from that of free MMC. In vivo, PL-MLP was approximately 3-fold less toxic than free MMC. The therapeutic index and absolute antitumor efficacy of PL-MLP were superior to that of free MMC in the three tumor models tested. In addition, PL-MLP was significantly more active than a formulation of doxorubicin in pegylated liposomes (DOXIL) in the M109R tumor model, a mouse tumor cell line with a multidrug-resistant phenotype. CONCLUSIONS: Delivery of MLP in pegylated liposomes is a potential approach for effective treatment of multidrug-resistant tumors while significantly buffering the toxicity of MMC.


Subject(s)
Liposomes/chemistry , Mitomycins/pharmacology , Neoplasms, Experimental/drug therapy , Prodrugs/pharmacology , Animals , Antineoplastic Agents/chemistry , Antineoplastic Agents/pharmacology , Antineoplastic Agents/therapeutic use , Cell Line, Tumor , Cell Survival/drug effects , Female , Inhibitory Concentration 50 , Mice , Mice, Inbred BALB C , Mitomycins/pharmacokinetics , Mitomycins/therapeutic use , Molecular Structure , Neoplasms, Experimental/pathology , Polyethylene Glycols/chemistry , Prodrugs/pharmacokinetics , Prodrugs/therapeutic use , Rats , Rats, Sprague-Dawley , Time Factors , Treatment Outcome
20.
Chem Res Toxicol ; 18(2): 213-23, 2005 Feb.
Article in English | MEDLINE | ID: mdl-15720125

ABSTRACT

Mitomycin C (MC) is a cytotoxic and mutagenic antitumor agent that alkylates and cross-links DNA. These effects are dependent on reductive bioactivation of MC. 2,7-Diaminomitosene (2,7-DAM) is the major metabolite of MC in tumor cells, generated by the reduction of MC. 2,7-DAM alkylates DNA in the cell in situ, forming an adduct at the N7 position of 2'-deoxyguanosine (2,7-DAM-dG-N7). To determine the biological effects of this adduct, we have synthesized an oligonucleotide containing a single 2,7-DAM-dG-N7 adduct and inserted it into an M13 bacteriophage genome. Replication of this construct in repair-competent Escherichia coli showed that the adduct was only weakly toxic and generated approximately 50% progeny as compared to control. No mutant was isolated after analysis of more than 4000 progeny phages from SOS-induced or uninduced host cells; therefore, we estimate that the mutation frequency of 2,7-DAM-dG-N7 was less than 2 x 10(-4) in E. coli. Subsequently, to determine if this adduct might be mutagenic in mammalian cells, it was incorporated into a single-stranded shuttle phagemid vector, pMS2, and replicated in simian kidney (COS-7) cells. Analysis of the progeny showed that mutational frequency of a site specific 2,7-DAM-dG-N7 was not higher than the spontaneous mutation frequency in simian kidney cells. In parallel experiments in cell free systems, template oligonucleotides containing a single 2,7-DAM-dG-N7 adduct directed selective incorporation of cytosine in the 5'-32P-labeled primer strands opposite the adducted guanine, catalyzed by Klenow (exo-) DNA polymerase. The adducted templates also supported full extension of primer strands by Klenow (exo-) and T7 (exo-) DNA polymerases and partial extension by DNA polymerase eta. The innocuous behavior of the 2,7-DAM-dG-N7 monoadduct in vivo and in vitro is in sharp contrast to that of the toxic MC-dG-N2 monoadduct reported earlier.


Subject(s)
DNA Adducts/chemistry , DNA Damage , Mitomycin/chemistry , Mitomycins/chemistry , Animals , COS Cells , DNA/drug effects , Escherichia coli/chemistry , Escherichia coli/drug effects , Mitomycins/chemical synthesis , Mitomycins/pharmacology , Molecular Structure , Mutagenicity Tests , Oligonucleotides/chemical synthesis , Oligonucleotides/chemistry , Oligonucleotides/pharmacology
SELECTION OF CITATIONS
SEARCH DETAIL
...