Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 28
Filter
Add more filters










Publication year range
1.
Molecules ; 28(13)2023 Jul 04.
Article in English | MEDLINE | ID: mdl-37446864

ABSTRACT

This review uses the National Cancer Institute (NCI) COMPARE program to establish an extensive list of heterocyclic iminoquinones and quinones with similarities in differential growth inhibition patterns across the 60-cell line panel of the NCI Developmental Therapeutics Program (DTP). Many natural products and synthetic analogues are revealed as potential NAD(P)H:quinone oxidoreductase 1 (NQO1) substrates, through correlations to dipyridoimidazo[5,4-f]benzimidazoleiminoquinone (DPIQ), and as potential thioredoxin reductase (TrxR) inhibitors, through correlations to benzo[1,2,4]triazin-7-ones and pleurotin. The strong correlation to NQO1 infers the enzyme has a major influence on the amount of the active compound with benzo[e]perimidines, phenoxazinones, benz[f]pyrido[1,2-a]indole-6,11-quinones, seriniquinones, kalasinamide, indolequinones, and furano[2,3-b]naphthoquinones, hypothesised as prodrugs. Compounds with very strong correlations to known TrxR inhibitors had inverse correlations to the expression of both reductase enzymes, NQO1 and TrxR, including naphtho[2,3-b][1,4]oxazepane-6,11-diones, benzo[a]carbazole-1,4-diones, pyranonaphthoquinones (including kalafungin, nanaomycin A, and analogues of griseusin A), and discorhabdin C. Quinoline-5,8-dione scaffolds based on streptonigrin and lavendamycin can correlate to either reductase. Inhibitors of TrxR are not necessarily (imino)quinones, e.g., parthenolides, while oxidising moieties are essential for correlations to NQO1, as with the mitosenes. Herein, an overview of synthetic methods and biological activity of each family of heterocyclic imino(quinone) is provided.


Subject(s)
Antineoplastic Agents , Indolequinones , Neoplasms , United States , National Cancer Institute (U.S.) , Quinones/chemistry , Oxidoreductases , NAD(P)H Dehydrogenase (Quinone)/metabolism , Antineoplastic Agents/pharmacology , Antineoplastic Agents/chemistry
2.
Front Oncol ; 11: 822500, 2021.
Article in English | MEDLINE | ID: mdl-35198436

ABSTRACT

DNA lesions arising from both exogenous and endogenous sources occur frequently in DNA. During DNA replication, the presence of unrepaired DNA damage in the template can arrest replication fork progression, leading to fork collapse, double-strand break formation, and to genome instability. To facilitate completion of replication and prevent the generation of strand breaks, DNA damage tolerance (DDT) pathways play a key role in allowing replication to proceed in the presence of lesions in the template. The two main DDT pathways are translesion synthesis (TLS), which involves the recruitment of specialized TLS polymerases to the site of replication arrest to bypass lesions, and homology-directed damage tolerance, which includes the template switching and fork reversal pathways. With some exceptions, lesion bypass by TLS polymerases is a source of mutagenesis, potentially contributing to the development of cancer. The capacity of TLS polymerases to bypass replication-blocking lesions induced by anti-cancer drugs such as cisplatin can also contribute to tumor chemoresistance. On the other hand, during homology-directed DDT the nascent sister strand is transiently utilised as a template for replication, allowing for error-free lesion bypass. Given the role of DNA damage tolerance pathways in replication, mutagenesis and chemoresistance, a more complete understanding of these pathways can provide avenues for therapeutic exploitation. A number of small molecule inhibitors of TLS polymerase activity have been identified that show synergy with conventional chemotherapeutic agents in killing cancer cells. In this review, we will summarize the major DDT pathways, explore the relationship between damage tolerance and carcinogenesis, and discuss the potential of targeting TLS polymerases as a therapeutic approach.

3.
Bioorg Chem ; 84: 418-433, 2019 03.
Article in English | MEDLINE | ID: mdl-30554081

ABSTRACT

1,5-Dideoxy-1,5-imino-l-fucitol (1-deoxyfuconojirimycin, DFJ) is an iminosugar that inhibits fucosidases. Herein, N-alkyl DFJs have been synthesised and tested against the α-fucosidases of T. maritima (bacterial origin) and B. taurus (bovine origin). The N-alkyl derivatives were inactive against the bacterial fucosidase, while inhibiting the bovine enzyme. Docking of inhibitors to homology models, generated for the bovine and human fucosidases, was carried out. N-Decyl-DFJ was toxic to cancer cell lines and was more potent than the other N-alkyl DFJs studied.


Subject(s)
Enzyme Inhibitors/chemical synthesis , Sugar Alcohols/chemistry , alpha-L-Fucosidase/antagonists & inhibitors , 1-Deoxynojirimycin/analogs & derivatives , 1-Deoxynojirimycin/chemistry , 1-Deoxynojirimycin/metabolism , Bacteria/enzymology , Bacterial Proteins/antagonists & inhibitors , Bacterial Proteins/metabolism , Binding Sites , Cell Line, Tumor , Cell Survival/drug effects , Enzyme Inhibitors/metabolism , Enzyme Inhibitors/pharmacology , Humans , Immunoglobulin G/metabolism , Immunoglobulin G/pharmacology , Inhibitory Concentration 50 , Melphalan/chemical synthesis , Melphalan/metabolism , Melphalan/pharmacology , Molecular Docking Simulation , Structure-Activity Relationship , Sugar Alcohols/metabolism , alpha-L-Fucosidase/metabolism
4.
Org Lett ; 20(10): 3025-3029, 2018 05 18.
Article in English | MEDLINE | ID: mdl-29697986

ABSTRACT

Isosorbide was functionalized with furoxan for the first time to give adducts that release nitric oxide up to 7.5 times faster than the commercial vasodilator, isosorbide-5-mononitrate (Is5N). The synthesis was facilitated by MeMgCl-mediated selective acetylation of isosorbide or selective deacetylation of isosorbide-2,5-diacetate, which was rationalized in terms of a more stable 5-alkoxide magnesium salt using DFT. Isosorbide-furoxans are safer to handle than Is5N due to greater thermal stability.

5.
Molecules ; 23(3)2018 Mar 03.
Article in English | MEDLINE | ID: mdl-29510488

ABSTRACT

Cell viability studies for benzo[1,2,4]triazin-7-ones and 1,2,4-benzotriazinyl (Blatter-type) radical precursors are described with comparisons made with 2,2,6,6-tetramethyl-1-piperidinyloxy (TEMPO). All of the stable free radicals were several orders of magnitude less cytotoxic than the benzo[1,2,4]triazin-7-ones. The synthesis and evaluation of two new pyrid-2-yl benzo[1,2,4]triazin-7-ones are described, where altering the 1,3-substitution from phenyl to pyrid-2-yl increased cytotoxicity against most cancer cell lines, as indicated using National Cancer Institute (NCI) one-dose testing. COMPARE analysis of five-dose testing data from the NCI showed very strong correlations to the naturally occurring anti-cancer compound pleurotin. COMPARE is program, which analyzes similarities in cytotoxicity data of compounds, and enables quantitative expression as Pearson correlation coefficients. Compounds were also evaluated using the independent MTT assay, which was compared with SRB assay data generated at the NCI.


Subject(s)
Antineoplastic Agents/pharmacology , Benzene Derivatives/pharmacology , Heterocyclic Compounds, 4 or More Rings/pharmacology , Triazines/pharmacology , Antineoplastic Agents/chemical synthesis , Benzene Derivatives/chemical synthesis , Cell Line, Tumor , Cell Survival/drug effects , Dose-Response Relationship, Drug , Free Radicals , HT29 Cells , Heterocyclic Compounds, 4 or More Rings/chemical synthesis , Humans , MCF-7 Cells , Structure-Activity Relationship , Triazines/chemical synthesis
6.
Bioorg Med Chem ; 24(16): 3565-70, 2016 08 15.
Article in English | MEDLINE | ID: mdl-27290691

ABSTRACT

The thioredoxin (Trx)-thioredoxin reductase (TrxR) system plays a key role in maintaining the cellular redox balance with Trx being over-expressed in a number of cancers. Inhibition of TrxR is an important strategy for anti-cancer drug discovery. The natural product pleurotin is a well-known irreversible inhibitor of TrxR. The cytotoxicity data for benzo[1,2,4]triazin-7-ones showed very strong correlation (Pearson correlation coefficients ∼0.8) to pleurotin using National Cancer Institute COMPARE analysis. A new 3-CF3 substituted benzo[1,2,4]triazin-7-one gave submicromolar inhibition of TrxR, although the parent compound 1,3-diphenylbenzo[1,2,4]triazin-7-one was more cytotoxic against cancer cell lines. Benzo[1,2,4]triazin-7-ones exhibited different types of reversible inhibition of TrxR, and cyclic voltammetry showed characteristic quasi-reversible redox processes. Cell viability studies indicated strong dependence of cytotoxicity on substitution at the 6-position of the 1,3-diphenylbenzo[1,2,4]triazin-7-one ring.


Subject(s)
Antineoplastic Agents/pharmacology , Heterocyclic Compounds, 4 or More Rings/antagonists & inhibitors , Thioredoxin-Disulfide Reductase/antagonists & inhibitors , Triazines/pharmacology , Antineoplastic Agents/chemistry , Cell Line, Transformed , Drug Discovery , Drug Screening Assays, Antitumor , Humans , Spectrometry, Mass, Matrix-Assisted Laser Desorption-Ionization , Triazines/chemistry
7.
Sci Rep ; 3: 3277, 2013 Nov 20.
Article in English | MEDLINE | ID: mdl-24253929

ABSTRACT

Human cells lacking DNA polymerase η (polη) are sensitive to platinum-based cancer chemotherapeutic agents. Using DNA combing to directly investigate the role of polη in bypass of platinum-induced DNA lesions in vivo, we demonstrate that nascent DNA strands are up to 39% shorter in human cells lacking polη than in cells expressing polη. This provides the first direct evidence that polη modulates replication fork progression in vivo following cisplatin and carboplatin treatment. Severe replication inhibition in individual platinum-treated polη-deficient cells correlates with enhanced phosphorylation of the RPA2 subunit of replication protein A on serines 4 and 8, as determined using EdU labelling and immunofluorescence, consistent with formation of DNA strand breaks at arrested forks in the absence of polη. Polη-mediated bypass of platinum-induced DNA lesions may therefore represent one mechanism by which cancer cells can tolerate platinum-based chemotherapy.


Subject(s)
DNA Damage/drug effects , DNA Damage/physiology , DNA Replication/drug effects , DNA Replication/physiology , DNA-Directed DNA Polymerase/metabolism , Platinum/pharmacology , Carboplatin/pharmacology , Carboplatin/toxicity , Cell Cycle/drug effects , Cell Cycle/physiology , Cell Line , Cisplatin/pharmacology , Cisplatin/toxicity , DNA-Directed DNA Polymerase/deficiency , DNA-Directed DNA Polymerase/genetics , Drug Synergism , Gene Expression , Humans , Phosphorylation , Platinum/toxicity , Replication Protein A/metabolism
8.
Int J Hematol ; 96(5): 649-56, 2012 Nov.
Article in English | MEDLINE | ID: mdl-23076878

ABSTRACT

Anthracyclines, including doxorubicin, are widely used in the treatment of leukemia. While the effects of doxorubicin on hematopoietic cells have been characterized, less is known about the response of human mesenchymal stem cells (hMSCs) in the bone marrow stroma to anthracyclines. We characterized the effect of doxorubicin on key DNA damage responses in hMSCs, and compared doxorubicin sensitivity and DNA damage response activation between isolated hMSCs and the chronic myelogenous leukemia cell line, K562. Phosphorylation of H2AX, Chk1, and RPA2 was more strongly activated in K562 cells than in hMSCs, at equivalent doses of doxorubicin. hMSCs were relatively resistant to doxorubicin such that, following exposure to 15 µM doxorubicin, the level of cleaved caspase-3 detected by western blotting was lower in hMSCs compared to K562 cells. Flow cytometric analysis of cell cycle progression demonstrated that exposure to doxorubicin induced G2/M phase arrest in hMSCs, while 48 h after exposure, 15.6 % of cells were apoptotic, as determined from the percentage of cells having sub-G1 DNA content. We also show that the doxorubicin sensitivity of hMSCs isolated from a healthy donor was comparable to that of hMSCs isolated from a chronic lymphocytic leukemia patient. Overall, our results demonstrate that high doses of doxorubicin induce the DNA damage response in hMSCs, and that cultured hMSCs are relatively resistant to doxorubicin.


Subject(s)
Antibiotics, Antineoplastic/adverse effects , DNA Damage , Doxorubicin/adverse effects , Mesenchymal Stem Cells/metabolism , Antibiotics, Antineoplastic/pharmacology , Apoptosis/drug effects , Checkpoint Kinase 1 , Doxorubicin/pharmacology , Female , G2 Phase Cell Cycle Checkpoints/drug effects , Histones/metabolism , Humans , Inorganic Pyrophosphatase/metabolism , K562 Cells , M Phase Cell Cycle Checkpoints/drug effects , Male , Mesenchymal Stem Cells/pathology , Mitochondrial Proteins/metabolism , Phosphorylation/drug effects , Protein Kinases/metabolism
9.
Bioorg Med Chem ; 20(10): 3223-32, 2012 May 15.
Article in English | MEDLINE | ID: mdl-22522008

ABSTRACT

Synthesis and cytotoxicity of imidazo[5,4-f]benzimidazolequinones and iminoquinone derivatives is described, enabling structure-activity relationships to be obtained. The most promising compound (an iminoquinone derivative) has undergone National Cancer Institute (NCI) 60 cell line (single and five dose) screening, and using the NCI COMPARE program, has shown correlation to NQO1 activity and to other NQO1 substrates. Common structural features suggest that the iminoquinone moiety is significant with regard to NQO1 specificity. Computational docking into the active site of NQO1 was performed, and the first comprehensive mitomycin C (MMC)-NQO1 docking study is presented. Small distances for hydride reduction and high binding affinities are characteristic of MMC and of iminoquinones showing correlations with NQO1 via COMPARE analysis. Docking also indicated that the presence of a substituent capable of hydrogen bonding to the His194 residue is important in influencing the orientation of the substrate in the NQO1 active site, leading to more efficient reduction.


Subject(s)
Benzimidazoles/toxicity , NAD(P)H Dehydrogenase (Quinone)/metabolism , Quinones/chemistry , Software , Antineoplastic Agents/chemistry , Antineoplastic Agents/toxicity , Binding Sites , Cell Line, Tumor , Drug Screening Assays, Antitumor , Humans , Hydrophobic and Hydrophilic Interactions , Inhibitory Concentration 50 , Models, Molecular , NAD(P)H Dehydrogenase (Quinone)/chemistry , Quinones/toxicity , Structure-Activity Relationship , Substrate Specificity
10.
Cell Cycle ; 10(21): 3768-77, 2011 Nov 01.
Article in English | MEDLINE | ID: mdl-22037398

ABSTRACT

DNA damaging agents are widely used in treatment of hematogical malignancies and solid tumors. While effects on hematopoietic stem cells have been characterized, less is known about the DNA damage response in human mesenchymal stem cells (hMSCs) in the bone marrow stroma, progenitors of osteoblasts, chondrocytes and adipocytes. To elucidate the response of undifferentiated hMSCs to γ-irradiation and cisplatin, key DNA damage responses have been characterised in hMSCs from normal adult donors. Cisplatin and γ-irradiation activated the DNA damage response in hMSCs, including induction of p53 and p21, and activation of PI3 kinase-related protein kinase (PIKK)-dependent phosphorylation of histone H2AX on serine 139, and replication protein A2 on serine4/serine8. Chemical inhibition of ATM or DNA-PK reduced DNA damage-induced phosphorylation of H2AX, indicating a role for both PIKKs in the response of hMSCs to DNA damage. Consistent with repair of DNA strand breaks, γ-H2AX staining decreased by 24 hours following gamma-irradiation. γ-Irradiation arrested hMSCs in the G 1 phase of the cell cycle, while cisplatin induced S-phase arrest, mediated in part by the ATR/Chk1 checkpoint pathway. In hMSCs isolated from a chronic lymphocytic leukemia (CLL) patient, p53 and p21 were induced by cisplatin and γ-irradiation, while RPA2 was phosphorylated on serine4/8 in particular following cisplatin. Compared to peripheral blood lymphocytes or the leukemia cell line K562, both normal hMSCs and CLL-derived hMSCs were more resistant to cisplatin and γ-irradiation. These results provide insights into key pathways mediating the response of bone marrow-derived hMSCs to DNA damaging agents used in cancer treatment.


Subject(s)
Cisplatin/pharmacology , DNA Damage , Gamma Rays , Mesenchymal Stem Cells/drug effects , Mutagens/pharmacology , Cell Cycle Checkpoints/drug effects , Cell Cycle Checkpoints/radiation effects , Cyclin-Dependent Kinase Inhibitor p21/drug effects , Cyclin-Dependent Kinase Inhibitor p21/metabolism , DNA Adducts , DNA Breaks/radiation effects , DNA Repair/drug effects , DNA Repair/radiation effects , Histones/metabolism , Humans , Mesenchymal Stem Cells/radiation effects , Phosphorylation , Tumor Suppressor Protein p53/drug effects , Tumor Suppressor Protein p53/metabolism
11.
Org Biomol Chem ; 9(19): 6700-6, 2011 Oct 07.
Article in English | MEDLINE | ID: mdl-21808774

ABSTRACT

Anionic aromatic ipso-substitution has allowed an aziridine ring to be fused onto pyrrolo[1,2-a]benzimidazole. This diazole analogue of aziridinomitosene, and N-[(aziridinyl)methyl]-1H-benzimidazole are shown to be significantly more cytotoxic towards the human breast cancer cell lines MCF-7 and HCC1937 than towards a human normal fibroblast cell line (GM00637). The aziridinyl fused pyrrolo[1,2-a]benzimidazole is less cytotoxic than the non-ring fused aziridinyl analogue towards all three cell lines. The BRCA1-deficient HCC1937 cells are more sensitive to mitomycin C (MMC) compared to GM00637 and MCF-7 cells. The evidence provided indicates that different pathways may mediate cellular response to benzimidazole-containing aziridine compounds compared to MMC.


Subject(s)
Antineoplastic Agents/pharmacology , Aziridines/chemistry , Benzimidazoles/pharmacology , Breast Neoplasms/pathology , Breast/drug effects , Antineoplastic Agents/chemical synthesis , Antineoplastic Agents/chemistry , Benzimidazoles/chemical synthesis , Benzimidazoles/chemistry , Breast/cytology , Cell Proliferation/drug effects , Chemistry Techniques, Synthetic , Dose-Response Relationship, Drug , Drug Screening Assays, Antitumor , Female , Humans , Molecular Structure , Stereoisomerism , Structure-Activity Relationship , Tumor Cells, Cultured
12.
Prostate ; 71(2): 111-24, 2011 Feb 01.
Article in English | MEDLINE | ID: mdl-20665530

ABSTRACT

BACKGROUND: While locally advanced prostate cancer is initially treatable with androgen ablation, eventually cells develop a castrate-resistant phenotype. Currently, there are no effective treatments for this form of the disease with Docetaxel only providing a small survival advantage. In this study, the effects of novel derivatives of titanocene dichloride on prostate cancer cell lines has been investigated. METHODS: Cellular effects were assessed using the crystal violet assay and the clonogenic survival assay. Cell cycle and apoptosis were assessed by propidium iodide staining. DNA damage was analyzed by comet assay and Western analysis. DNA damage response inhibition was achieved by pre-incubation with an ATM/ATR inhibitor; CGK733 and DNA-PK inhibitor; DMNB. RESULTS: These analogs caused a reduction in cell number. In particular titanocene Y and C had significant effects in all cell lines. A reduction in clonogenic survival was found in response to titanocene Y in three cell lines while the PC-3 cells exhibited increased resistance.Further analysis showed no effect on cell cycle however, the analogs were found to induce apoptosis in a dose-dependent manner in all cell lines. These analogs associate with DNA, induce DNA damage and a differential damage response. Inhibition of key regulators of this DNA damage response sensitized the PC-3 cell line to titanocene-induced apoptosis and significantly reduced the clonogenic capacity of the cells. CONCLUSION: These results demonstrate the mechanism of action of these novel titanocene dichloride analogs and their potential use in castrate-independent advanced prostate cancer.


Subject(s)
Antineoplastic Agents/pharmacology , Apoptosis/drug effects , DNA Damage , Organometallic Compounds/pharmacology , Prostatic Neoplasms/drug therapy , Apoptosis/genetics , Blotting, Western , Cell Line, Tumor , Cell Survival/drug effects , Comet Assay , Gentian Violet/chemistry , Humans , Male , Neoplasms, Hormone-Dependent/drug therapy , Neoplasms, Hormone-Dependent/genetics , Neoplasms, Hormone-Dependent/pathology , Prostatic Neoplasms/genetics , Prostatic Neoplasms/pathology , Titanium/chemistry
13.
Cancer Genomics Proteomics ; 7(5): 279-86, 2010.
Article in English | MEDLINE | ID: mdl-20952762

ABSTRACT

Cancer chemotherapy relies heavily on DNA damaging agents such as cisplatin to induce tumour cell death. The response of cells to genotoxic insult, including cell cycle arrest, DNA repair and cell death, is mediated by the DNA damage response (DDR). To address the relationship between the DDR and the outcome of exposure, this study utilised a magnetic-activated cell sorting (MACS®)-based approach to isolate apoptotic and non-apoptotic cells from a DNA polymerase eta-deficient human cell line. The pattern of phosphorylation of the key DNA damage response protein RPA2 on serine 4/8 was altered in apoptotic cells isolated following cisplatin treatment. By combining MACS® with multi-antibody screening for phosphorylated proteins, apoptosis-associated changes were characterized in a number of key signalling pathways. Phosphorylation of Erk1 on Thr202/Tyr204, and Erk2 on Thr185/Tyr187 was increased in apoptotic cells. This approach provides novel insights into the relationship between cisplatin-induced protein phosphorylation and the cellular consequences of exposure to this chemotherapeutic agent.


Subject(s)
Annexin A5/metabolism , Antineoplastic Agents/pharmacology , Cisplatin/pharmacology , Intracellular Signaling Peptides and Proteins/metabolism , Replication Protein A/metabolism , Signal Transduction/drug effects , Antibodies , Apoptosis/drug effects , Blotting, Western , Cell Cycle/drug effects , Cell Line, Tumor , Cell Separation , DNA Damage , DNA Repair , Flow Cytometry , Humans , Phosphorylation
14.
Eur J Med Chem ; 45(9): 3762-9, 2010 Sep.
Article in English | MEDLINE | ID: mdl-20605274

ABSTRACT

A facile 6-exo-trig cyclization of sigma-aromatic radicals has allowed the synthesis of various aromatic ring fused benzimidazoles and benzimidazolequinones. The most highly conjugated naphthyl fused benzimidazolequinone, (5-methyl-5,6-dihydrobenzimidazo[2,1-a]benzo[f]isoquinoline-8,11-dione) showed the highest specificity towards human cervical (HeLa) and prostate (DU145) cancer cell lines with little toxicity towards a human normal (GM00637) cell line at doses of <1 microM. In contrast, 2-aromatic ring substituted (benzimidazole-4,7-diones) analogues, benzimidazolequinone with a pyridine ring and mitomycin C were more toxic than the highly conjugated naphthyl fused benzimidazolequinone towards the normal cell line.


Subject(s)
Antineoplastic Agents/chemical synthesis , Antineoplastic Agents/pharmacology , Benzimidazoles/chemical synthesis , Benzimidazoles/pharmacology , Hydrocarbons, Aromatic/chemistry , Antineoplastic Agents/chemistry , Antineoplastic Agents/toxicity , Benzimidazoles/chemistry , Benzimidazoles/toxicity , Cell Line, Tumor , Cell Survival/drug effects , Humans , Inhibitory Concentration 50
15.
Org Biomol Chem ; 8(14): 3149-56, 2010 Jul 21.
Article in English | MEDLINE | ID: mdl-20485753

ABSTRACT

Bu(3)SnH/1,1'-azobis(cyclohexanecarbonitrile) (ACN)-mediated five, six, and seven-membered double alkyl radical cyclizations onto imidazo[5,4-f]benzimidazole and imidazo[4,5-f]benzimidazole are described. The quinone derivatives evaluated show selective toxicity towards human cervical (HeLa) and prostate (DU145) cancer cell lines (with negligible toxicity towards a normal human cell line, GM00637). Only the Fremy oxidation of the 6-aminoimidazo[5,4-f]benzimidazole gave iminoquinone, which showed high specificity towards the prostate cancer cell line (DU145).


Subject(s)
Antineoplastic Agents/chemistry , Antineoplastic Agents/pharmacology , Benzimidazoles/chemistry , Benzimidazoles/pharmacology , Antineoplastic Agents/chemical synthesis , Benzimidazoles/chemical synthesis , Cell Line, Tumor , Humans , Inhibitory Concentration 50
16.
Curr Cancer Drug Targets ; 10(4): 411-21, 2010 Jun.
Article in English | MEDLINE | ID: mdl-20370690

ABSTRACT

Human mesenchymal stem cells (hMSCs) consist of cells that can differentiate into mesenchymal tissues, including osteoblasts, adipocytes and chondrocytes. hMSCs constitute a particular stem cell niche in the stromal compartment of the bone marrow, and also play a role in maintaining the normal function of haematopoietic stem cells. Furthermore, hMSCs localise to solid tumours, and can modulate cancer cell function through secretion of paracrine signals. While hMSCs, either in the bone marrow, or in the microenvironment of a tumour, will be targeted by DNA damaging agents used in cancer therapy, the response of the hMSC population to DNA damage is not well understood. In their role as progenitor cells, genomic DNA damage to hMSCs during cancer therapy could generate a population of surviving cells that can go on to give rise to secondary tumours. A better understanding of the response of hMSCs to DNA damage could provide new insights into the effects of cancer treatments, as well as into the development of treatment-associated secondary cancers. The article will review the relationship of hMSCs to cancer, with a focus on the response of hMSCs to DNA damaging agents.


Subject(s)
Antineoplastic Agents/pharmacology , DNA Damage/drug effects , Mesenchymal Stem Cells/drug effects , Neoplasms/drug therapy , Humans , Neoplasms/pathology
17.
Bioorg Med Chem ; 18(8): 2930-6, 2010 Apr 15.
Article in English | MEDLINE | ID: mdl-20347318

ABSTRACT

A series of new aromatic monoesters of alpha-aminoaralkylphosphonic acids were synthesized by selective hydrolysis of corresponding aromatic diesters of alpha-aminoaralkylphosphonic acids. New potential inhibitors of aminopeptidase N/CD13, an enzyme important in tumour angiogenesis, were developed. Some derivatives of the homophenylalanine and norleucine related monoaryl phosphonates displayed higher inhibition potency than corresponding alpha-aminoaralkylphosphonic acids toward aminopeptidase N/CD13. The effect of one of the new inhibitors on the growth of human PANC-1 and HT-1080 cell lines was examined, either alone or in combination with TNF-alpha.


Subject(s)
Angiogenesis Inhibitors/chemistry , CD13 Antigens/antagonists & inhibitors , Phosphinic Acids/chemistry , Protease Inhibitors/chemistry , Angiogenesis Inhibitors/chemical synthesis , Angiogenesis Inhibitors/pharmacology , CD13 Antigens/metabolism , Cell Line, Tumor , Humans , Phosphinic Acids/chemical synthesis , Phosphinic Acids/pharmacology , Protease Inhibitors/chemical synthesis , Protease Inhibitors/pharmacology , Structure-Activity Relationship , Tumor Necrosis Factor-alpha/metabolism
18.
Eur J Med Chem ; 45(5): 1873-9, 2010 May.
Article in English | MEDLINE | ID: mdl-20122765

ABSTRACT

Aziridinyl substituted benzimidazolequinones are more toxic than methoxy analogues towards normal human fibroblast cells (GM00637). The aziridinyl substituent is required for hypersensitive killing of Fanconi anaemia (FA) cells (PD20i) deficient in FANCD2. Despite lacking quinone functionality, 4,7-dimethoxy-N-[(aziridin-2-yl)methyl]benzimidazole also induces hypersensitivity from FA cells, similar to their response towards mitomycin C. Expression of FANCD2 (in PD20:RV) corrects FA cell hypersensitivity supporting cellular response via the FANC pathway.


Subject(s)
Aziridines/chemistry , Benzimidazoles/toxicity , Fanconi Anemia/pathology , Fibroblasts/drug effects , Quinones/toxicity , Benzimidazoles/chemistry , Cell Survival/drug effects , Cells, Cultured , Dose-Response Relationship, Drug , Drug Evaluation, Preclinical , Humans , Molecular Structure , Quinones/chemistry , Stereoisomerism , Structure-Activity Relationship
19.
Subcell Biochem ; 50: 189-209, 2010.
Article in English | MEDLINE | ID: mdl-20012583

ABSTRACT

Genomic DNA is constantly damaged by exposure to exogenous and endogenous agents. Bulky adducts such as UV-induced cyclobutane pyrimidine dimers (CPDs) in the template DNA present a barrier to DNA synthesis by the major eukaryotic replicative polymerases including DNA polymerase delta. Translesion synthesis (TLS) carried out by specialized DNA polymerases is an evolutionarily conserved mechanism of DNA damage tolerance. The Y family of DNA polymerases, including DNA polymerase eta (Pol eta), the subject of this chapter, play a key role in TLS. Mutations in the human POLH gene encoding Pol eta underlie the genetic disease xeroderma pigmentosum variant (XPV), characterized by sun sensitivity, elevated incidence of skin cancer, and at the cellular level, by delayed replication and hypermutability after UV-irradiation. Pol eta is a low fidelity enzyme when copying undamaged DNA, but can carry out error-free TLS at sites of UV-induced dithymine CPDs. The active site of Pol eta has an open conformation that can accommodate CPDs, as well as cisplatin-induced intrastrand DNA crosslinks. Pol eta is recruited to sites of replication arrest in a tightly regulated process through interaction with PCNA. Pol eta-deficient cells show strong activation of downstream DNA damage responses including ATR signaling, and accumulate strand breaks as a result of replication fork collapse. Thus, Pol eta plays an important role in preventing genome instability after UV- and cisplatin-induced DNA damage. Inhibition of DNA damage tolerance pathways in tumors might also represent an approach to potentiate the effects of DNA damaging agents such as cisplatin.


Subject(s)
DNA Damage , DNA Repair , DNA-Directed DNA Polymerase/metabolism , Humans
20.
Nucleic Acids Res ; 37(18): 6028-41, 2009 Oct.
Article in English | MEDLINE | ID: mdl-19671522

ABSTRACT

The human single-stranded DNA-binding protein, replication protein A (RPA), is regulated by the N-terminal phosphorylation of its 32-kDa subunit, RPA2. RPA2 is hyperphosphorylated in response to various DNA-damaging agents and also phosphorylated in a cell-cycle-dependent manner during S- and M-phase, primarily at two CDK consensus sites, S23 and S29. Here we generated two monoclonal phospho-specific antibodies directed against these CDK sites. These phospho-specific RPA2-(P)-S23 and RPA2-(P)-S29 antibodies recognized mitotically phosphorylated RPA2 with high specificity. In addition, the RPA2-(P)-S23 antibody recognized the S-phase-specific phosphorylation of RPA2, suggesting that during S-phase only S23 is phosphorylated, whereas during M-phase both CDK sites, S23 and S29, are phosphorylated. Immunofluorescence microscopy revealed that the mitotic phosphorylation of RPA2 starts at the onset of mitosis, and dephosphorylation occurs during late cytokinesis. In mitotic cells treated with ionizing radiation (IR), we observed a rapid hyperphosphorylation of RPA2 in addition to its mitotic phosphorylation at S23 and S29, associated with a significant change in the subcellular localization of RPA. Our data also indicate that the RPA2 hyperphosphorylation in response to IR is facilitated by the activity of both ATM and DNA-PK, and is associated with activation of the Chk2 pathway.


Subject(s)
DNA Damage , Mitosis , Replication Protein A/metabolism , Ataxia Telangiectasia Mutated Proteins , Cell Cycle Proteins/metabolism , DNA-Activated Protein Kinase/metabolism , DNA-Binding Proteins/metabolism , Humans , Mitosis/radiation effects , Phosphorylation , Protein Serine-Threonine Kinases/metabolism , Protein Subunits/chemistry , Protein Subunits/metabolism , Radiation, Ionizing , Replication Protein A/chemistry , Serine/metabolism , Tumor Suppressor Proteins/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...