Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 24
Filter
Add more filters










Publication year range
1.
Biochem Pharmacol ; 83(9): 1241-50, 2012 May 01.
Article in English | MEDLINE | ID: mdl-22326903

ABSTRACT

Anthracycline chemotherapeutics are well characterised as poisons of topoisomerase II, however many anthracyclines, including doxorubicin, are also capable of forming drug-DNA adducts. Anthracycline-DNA adducts present an unusual obstacle for cells as they are covalently attached to one DNA strand and stabilised by hydrogen bonding to the other strand. We now show that in cycling cells processing of anthracycline adducts through DNA replication appears dominant compared to processing via transcription-coupled pathways, and that the processing of these adducts into DNA breaks is independent of topoisomerase II. It has previously been shown that cells deficient in homologous recombination (HR) are hypersensitive to adduct forming treatments. Given that anthracycline-DNA adducts, whilst not true crosslinks, are associated with both DNA strands, the role of ICL repair pathways was investigated. Mus81 is a structure specific nuclease implicated in Holliday junction resolution and the resolution of branched DNA formed by stalled replication forks. We now show that ICL repair deficient cells (Mus81(-/-)) are hypersensitive to anthracycline-DNA adducts and ET-743, a compound which causes a chemically similar type of DNA damage. Further analysis of this mechanism showed that Mus81 does not appear to cause DNA breaks resulting from either anthracycline- or ET743-DNA adducts. This suggests Mus81 processes these novel forms of DNA damage in a fundamentally different way compared to the processing of classical covalent crosslinks. Improved understanding of the role of DNA repair in response to such adducts may lead to more effective chemotherapy for patients with BRCA1/2 mutations and other HR deficiencies.


Subject(s)
Anthracyclines/metabolism , Anthracyclines/pharmacology , DNA Adducts/metabolism , DNA Repair , DNA Replication , DNA-Binding Proteins/metabolism , Endonucleases/metabolism , Animals , Anthracyclines/chemistry , Cricetinae , Cricetulus , DNA Adducts/chemistry , DNA Breaks, Double-Stranded/drug effects , DNA Topoisomerases, Type II/metabolism , DNA-Binding Proteins/genetics , Dioxoles/pharmacology , Endonucleases/genetics , Fanconi Anemia Complementation Group D2 Protein/genetics , Fanconi Anemia Complementation Group D2 Protein/metabolism , G1 Phase/genetics , HL-60 Cells , Homologous Recombination , Humans , Tetrahydroisoquinolines/pharmacology , Trabectedin
2.
Anticancer Agents Med Chem ; 10(1): 70-7, 2010 Jan.
Article in English | MEDLINE | ID: mdl-19807684

ABSTRACT

Barminomycin is a member of the anthracycline class of anticancer agents and was originally discovered as a pink/red complex with DNA and RNA and named SN-07. The chromophore was subsequently separated from the nucleic acids by nuclease digestion and contained the four-membered anthraquinone ring system characteristic of anthracyclines, but with an unusual eight membered ring that contained a carbinolamine which readily interconverted to an imine. The imine form is analogous to the formaldehyde-activated form of other anthracyclines such as doxorubicin. The imine form confers exceptional activity to barminomycin which is 1,000-fold more cytotoxic than doxorubicin. Barminomycin rapidly forms adducts with DNA, reacting with the exocyclic amino group of guanine residues and with high selectivity for 5'-GC-3' sequences. The coupling to DNA appears to be identical to the N-C-N aminal linkage formed between doxorubicin and DNA where the carbon derives from formaldehyde for doxorubicin-DNA adducts, whereas this "activated carbon" is an inherent component of the imine group in the eight membered ring of barminomycin. Although the linkage of both drugs to DNA appears to be identical, barminomycin-DNA complexes are essentially irreversible compared to the labile doxorubicin-DNA adducts which have an in vitro (purified DNA) half-life of 25 h at 37 degrees C. A 3D model of the barminomycin-DNA complex has been defined from 307 NOE distance constraints. The enhanced stability of barminomycin-DNA adducts appears to be due primarily to protection of the aminal linkage from hydrolysis and this has provided insight into the design of new anthracycline derivatives with enhanced stability and activity. Strategies for harnessing the extreme reactivity and activity of barminomycin are also presented.


Subject(s)
Anthracyclines/chemistry , Anthracyclines/pharmacology , DNA Adducts/chemistry , Drug Discovery , Magnetic Resonance Spectroscopy , Models, Molecular
3.
Br J Cancer ; 96(11): 1667-74, 2007 Jun 04.
Article in English | MEDLINE | ID: mdl-17473824

ABSTRACT

Formaldehyde has been previously shown to play a dominant role in promoting synergy between doxorubicin (Dox) and formaldehyde-releasing butyric acid (BA) prodrugs in killing cancer cells. In this work, we report that these prodrugs also protect neonatal rat cardiomyocytes and adult mice against toxicity elicited by Dox. In cardiomyocytes treated with Dox, the formaldehyde releasing prodrugs butyroyloxymethyl diethylphosphate (AN-7) and butyroyloxymethyl butyrate (AN-1), but not the corresponding acetaldehyde-releasing butyroyloxydiethyl phosphate (AN-88) or butyroyloxyethyl butyrate (AN-11), reduced lactate dehydrogenase leakage, prevented loss of mitochondrial membrane potential (DeltaPsim) and attenuated upregulation of the proapoptotic gene Bax. In Dox-treated mice, AN-7 but not AN-88 attenuated weight-loss and mortality, and increase in serum lactate dehydrogenase. These findings show that BA prodrugs that release formaldehyde and augment Dox anticancer activity also protect against Dox cardiotoxicity. Based on these observations, clinical applications of these prodrugs for patients treated with Dox warrant further investigation.


Subject(s)
Antineoplastic Agents/toxicity , Butyric Acid/pharmacology , Cytoprotection/drug effects , Doxorubicin/toxicity , Formaldehyde/pharmacology , Myocytes, Cardiac/drug effects , Organophosphates/pharmacology , Prodrugs/pharmacology , Animals , Animals, Newborn , Antineoplastic Agents/metabolism , Antineoplastic Agents/pharmacology , Antineoplastic Combined Chemotherapy Protocols/pharmacology , Butyrates , Butyric Acid/metabolism , Cells, Cultured , Drug Evaluation, Preclinical , Female , Formaldehyde/metabolism , Gene Expression Regulation/drug effects , Histone Deacetylases/metabolism , Membrane Potential, Mitochondrial/drug effects , Mice , Mice, Inbred C57BL , Models, Biological , Myocytes, Cardiac/metabolism , Organophosphorus Compounds , Rats
4.
Curr Med Chem Anticancer Agents ; 5(5): 431-47, 2005 Sep.
Article in English | MEDLINE | ID: mdl-16178771

ABSTRACT

The anthracycline group of compounds are amongst the most effective chemotherapy agents currently in use for cancer treatment. They are generally classified as topoisomerase II inhibitors but also have a variety of other targets in cells. It has been known for some years that the anthracyclines are capable of forming DNA adducts, but the relevance and extent of these DNA adducts in cells and their role in causing cell death has remained obscure. When the adduct structure was solved, it became clear that formaldehyde was an absolute requirement for adduct formation. This led to a renewed interest in the capacity of anthracyclines to form DNA adducts, and there are now several ways in which adduct formation can be facilitated in cells. These involve strategies to provide the requisite formaldehyde in the form of anthracycline-formaldehyde conjugates, and the use of formaldehyde-releasing drugs in combination with anthracyclines. Of particular interest is the new therapeutic compound AN-9 that releases both butyric acid and formaldehyde, leading to efficient anthracycline-DNA adduct formation, and synergy between the two compounds. Targeted formation of adducts using anthracycline-formaldehyde conjugates tethered to cell surface targeted molecules is now also possible. Some of the cellular consequences of these adducts have now been studied, and it appears that their formation can overcome anthracycline-resistance mechanisms, and that they are more efficient at inducing apoptosis than when functioning primarily through impairment of topoisomerase II. The clinical application of the use of anthracyclines as DNA adduct forming agents is now being explored.


Subject(s)
Anthracyclines/pharmacology , Formaldehyde/chemistry , Animals , Anthracyclines/chemistry , Anthracyclines/metabolism , Apoptosis/drug effects , DNA Adducts/drug effects , DNA Adducts/metabolism , Formaldehyde/metabolism , Formaldehyde/pharmacology , Humans , Molecular Structure , Structure-Activity Relationship
5.
Cancer Res ; 61(22): 8194-202, 2001 Nov 15.
Article in English | MEDLINE | ID: mdl-11719450

ABSTRACT

The interaction of Adriamycin and pivaloyloxymethyl butyrate (AN-9) was investigated in IMR-32 neuroblastoma and MCF-7 breast adenocarcinoma cells. Adriamycin is a widely used anticancer drug, whereas AN-9 is an anticancer agent presently undergoing Phase II clinical trials. The anticancer activity of AN-9 has been attributed to its ability to act as a butyric acid prodrug, although it also releases formaldehyde and pivalic acid. Adriamycin and AN-9 in combination display synergy when exposed simultaneously to cells or when AN-9 treatment is up to 18 h after Adriamycin administration. However, the reverse order of addition results in antagonism. These interactions have been established using cell viability assays and classical isobologram analysis. To understand the molecular basis of this synergy, the relative levels of Adriamycin-DNA adducts were determined using various treatment combinations. Levels of Adriamycin-DNA adducts were enhanced when treatment combinations known to be synergistic were used and were diminished using those treatments known to be antagonistic. The relative timing of the addition of Adriamycin and AN-9 was critical, with a 20-fold enhancement of Adriamycin-DNA adducts occurring when AN-9 was administered 2 h after the exposure of cells to Adriamycin. The enhanced levels of these adducts and the accompanying decreased cell viability were directly related to the esterase-dependent release of formaldehyde from AN-9, providing evidence for the formaldehyde-mediated activation of Adriamycin.


Subject(s)
Antineoplastic Combined Chemotherapy Protocols/pharmacology , Butyrates/pharmacokinetics , Doxorubicin/pharmacology , Formaldehyde/pharmacology , Prodrugs/pharmacokinetics , Adenocarcinoma/drug therapy , Adenocarcinoma/metabolism , Biotransformation , Breast Neoplasms/drug therapy , Breast Neoplasms/metabolism , Butyrates/administration & dosage , Butyrates/pharmacology , DNA Adducts/biosynthesis , Doxorubicin/administration & dosage , Doxorubicin/pharmacokinetics , Drug Administration Schedule , Drug Synergism , Formaldehyde/pharmacokinetics , Humans , Neuroblastoma/drug therapy , Neuroblastoma/metabolism , Prodrugs/administration & dosage , Prodrugs/pharmacology , Tumor Cells, Cultured
6.
Chem Biol Interact ; 138(2): 137-53, 2001 Nov 28.
Article in English | MEDLINE | ID: mdl-11672697

ABSTRACT

The anthracycline Adriamycin is known to form adducts with DNA, but requires prior activation by formaldehyde. In contrast, the anthracycline barminomycin is also able to form adducts with DNA, but does not require activation by formaldehyde. Barminomycin, therefore, appears to function as a pre-activated form of Adriamycin. The DNA adducts formed by both anthracyclines are bound covalently to only one strand of DNA, but both also stabilise duplex DNA sufficiently that they can be detected as virtual interstrand crosslinks in heat denaturation electrophoretic crosslinking assays. The barminomycin-DNA adducts form extremely rapidly with DNA, and at exceedingly low concentrations (approximately 50-fold lower than with Adriamycin in the presence of excess formaldehyde), both characteristics consistent with barminomycin being in a pre-activated state, hence, undergoing a bimolecular reaction with DNA compared with the trimolecular reaction (drug, formaldehyde and DNA) required with Adriamycin. Surprisingly, barminomycin-DNA adducts are substantially more stable (essentially irreversible) than Adriamycin-DNA adducts (half life of approximately 25 h at 37 degrees C). Due to this understanding of the reactivity of barminomycin and its exceptional cytotoxicity (1000-fold more cytotoxic than Adriamycin), detailed structural studies of barminomycin-DNA adducts are now warranted, both in vitro and in tumour cells.


Subject(s)
Anthracyclines/chemistry , Antibiotics, Antineoplastic/chemistry , Antineoplastic Agents , Doxorubicin , Animals , Anthracyclines/pharmacology , Antibiotics, Antineoplastic/pharmacology , Cattle , Cross-Linking Reagents/chemistry , Cross-Linking Reagents/metabolism , DNA/chemistry , DNA/drug effects , DNA/metabolism , DNA Adducts/chemistry , DNA Adducts/metabolism , Dose-Response Relationship, Drug , Formaldehyde/chemistry , Formaldehyde/metabolism , Hot Temperature , In Vitro Techniques , Kinetics , Spectrometry, Mass, Electrospray Ionization
8.
J Inorg Biochem ; 84(1-2): 119-27, 2001 Mar.
Article in English | MEDLINE | ID: mdl-11330470

ABSTRACT

The non-covalent binding of [(en)Pt(mu-dpzm)2Pt(en)]4+ to segments of DNA containing only G and C bases has been studied to gain an understanding of the pre-covalent binding association of cationic polynuclear platinum(II) anti-cancer drugs at G/C sites. 1H-NMR and CD spectroscopy were used to study the binding of the metal complex to the oligonucleotide d(GC)5 and the polynucleotide poly(dG-dC).poly(dG-dC), respectively. NOE contacts between the metal complex protons and the oligonucleotide sugar H1' protons observed in NOESY spectra indicated that the metal complex bound in the minor groove at the central C4 to G7 region of the oligonucleotide. This result indicates that even though cationic polynuclear platinum(II) complexes bind covalently in the major groove at G/C sites, the pre-covalent binding association is favoured in the minor groove. CD spectra indicated that the addition of the metal complex to poly(dG-dC)-poly(dG-dC) induced some conformational changes, but it was not possible to conclude that [(en)Pt(mu-dpzm)2Pt(en)]4+ induced a B- to Z-type DNA transition. In addition, in vitro transcription assays using the lac UV5 promoter showed that the non-covalent binding of [(en)Pt(mu-dpzm)2Pt(en)]4+ was sufficiently stable to inhibit transcription, and at particular sites.


Subject(s)
DNA/metabolism , Organoplatinum Compounds/metabolism , Pyrazoles/metabolism , Antineoplastic Agents/chemistry , Antineoplastic Agents/metabolism , Base Composition , Base Sequence , Binding Sites , Circular Dichroism , DNA/chemistry , DNA/drug effects , In Vitro Techniques , Magnetic Resonance Spectroscopy , Molecular Sequence Data , Nucleic Acid Conformation/drug effects , Oligodeoxyribonucleotides/chemistry , Oligodeoxyribonucleotides/metabolism , Organoplatinum Compounds/chemistry , Organoplatinum Compounds/pharmacology , Polydeoxyribonucleotides/chemistry , Polydeoxyribonucleotides/metabolism , Pyrazoles/chemistry , Pyrazoles/pharmacology
9.
Proc Natl Acad Sci U S A ; 98(10): 5705-10, 2001 May 08.
Article in English | MEDLINE | ID: mdl-11331754

ABSTRACT

Neocentromeres (NCs) are fully functional centromeres that arise ectopically in noncentromeric regions lacking alpha-satellite DNA. Using telomere-associated chromosome truncation, we have produced a series of minichromosomes (MiCs) from a mardel(10) marker chromosome containing a previously characterized human NC. These MiCs range in size from approximately 0.7 to 1.8 Mb and contain single-copy intact genomic DNA from the 10q25 region. Two of these NC-based Mi-Cs (NC-MiCs) appear circular whereas one is linear. All demonstrate stability in both structure and mitotic transmission in the absence of drug selection. Presence of a functional NC is shown by binding a host of key centromere-associated proteins. These NC-MiCs provide direct evidence for mitotic segregation function of the NC DNA and represent examples of stable mammalian MiCs lacking centromeric repeats.


Subject(s)
Centromere , Chromosomes, Human , Telomere , Cell Line , Gene Transfer Techniques , Humans , In Situ Hybridization, Fluorescence
10.
J Biol Chem ; 276(19): 15953-60, 2001 May 11.
Article in English | MEDLINE | ID: mdl-11278477

ABSTRACT

Recently, we have shown that mitoxantrone can be activated by formaldehyde in vitro to form DNA adducts that are specific for CpG and CpA sites in DNA. The CpG specificity of adduct formation prompted investigations into the effect of cytosine methylation (CpG) on adduct formation, since the majority of CpG dinucleotides in the mammalian genome are methylated and hypermethylation in subsets of genes is associated with various neoplasms. Upon methylation of a 512-base pair DNA fragment (containing the lac UV5 promoter) using HpaII methylase, three CCGG sites downstream of the promoter were methylated at C5 of the internal cytosine residue. In vitro transcription studies of mitoxantrone-reacted DNA revealed a 3-fold enhancement in transcriptional blockage (and hence adduct formation) exclusively at these methylated sites. In vitro cross-linking assays also revealed that methylation enhanced mitoxantrone adduct formation by 2-3-fold, and methylation of cytosine at a single potential drug binding site on a duplex oligonucleotide also enhanced adduct levels by 3-fold. Collectively, these results indicate preferential adduct formation at methylated CpG sites. However, adducts at these methylated sites exhibited the same stability as nonmethylated sites, suggesting that cytosine methylation increases drug accessibility to DNA rather than being involved in kinetic stabilization of the adduct.


Subject(s)
Cytosine , DNA Adducts/metabolism , DNA Methylation , Dinucleoside Phosphates/metabolism , Mitoxantrone/metabolism , Base Pairing , Base Sequence , Binding Sites , DNA-Cytosine Methylases/metabolism , Formaldehyde/pharmacology , Kinetics , Mitoxantrone/pharmacology , Oligodeoxyribonucleotides/chemistry , Oligodeoxyribonucleotides/metabolism , Plasmids/chemistry , Plasmids/metabolism , Promoter Regions, Genetic , Transcription, Genetic/drug effects , beta-Galactosidase/genetics
11.
Nucleic Acids Res ; 28(4): 982-90, 2000 Feb 15.
Article in English | MEDLINE | ID: mdl-10648792

ABSTRACT

Recently we have found that mitoxantrone, like Adria-mycin, can be activated by formaldehyde and subsequently form adducts which stabilise double-stranded DNA in vitro. This activation by formaldehyde may be biologically relevant since formaldehyde levels are elevated in those tumours in which mitoxan-trone is most cytotoxic. In vitro transcription analysis revealed that these adducts block the progression of RNA polymerase during transcription and cause truncated RNA transcripts. There was an absolute requirement for both mitoxantrone and formaldehyde in transcriptional blockage formation and the activated complex was found to exhibit site specificity, with blockage occurring prior to CpG and CpA sites in the DNA (non-template strand). The stability of the adduct at 37 degrees C was site dependent. The half-lives ranged from 45 min to approximately 5 h and this was dependent on both the central 2 bp blockage site as well as flanking sequences. The CpG specificity of mitoxantrone adduct sites was also confirmed independently by a lambda exonuclease digestion assay.


Subject(s)
CpG Islands/genetics , DNA Adducts , Dinucleoside Phosphates/genetics , Formaldehyde/pharmacology , Mitoxantrone/pharmacology , Base Sequence , DNA Primers , Drug Interactions , Exodeoxyribonucleases/metabolism , Hot Temperature , Transcription, Genetic , Viral Proteins
12.
Nucleic Acids Res ; 28(4): 1019-25, 2000 Feb 15.
Article in English | MEDLINE | ID: mdl-10648796

ABSTRACT

Activation of Adriamycin by formaldehyde leads to the formation of drug-DNA adducts in vitro and these adducts stabilise the DNA to such a degree that they function as virtual interstrand cross-links. The formation of these virtual interstrand cross-links by Adriamycin was investigated in MCF-7 cells using a gene-specific interstrand cross-linking assay. Cross-linking was measured in both the nuclear-encoded DHFR gene and in mitochondrial DNA (mtDNA). Cross-link formation increased linearly with Adriamycin concentration following a 4 h exposure to the drug. The rate of formation of Adriamycin cross-links in each of the genomes was similar, reaching maximal levels of 0.55 and 0.4 cross-links/10 kb in the DHFR gene and mtDNA respectively, following exposure to 20 micro M Adriamycin for 8 h. The interstrand cross-link was short lived in both DNA compartments, with a half-life of 4.5 and 3.3 h in the DHFR gene and mtDNA respectively. The kinetics of total Adriamycin adduct formation, detected using [(14)C]Adriamycin, was similar to that of cross-link formation. Maximal adduct levels (30 lesions/10 kb) were observed following incubation at 20 micro M drug for 8 h. The formation of such high levels of adducts and cross-links could therefore be expected to contribute to the mechanism of action of Adriamycin.


Subject(s)
Cell Nucleus/chemistry , DNA, Mitochondrial/chemistry , DNA, Neoplasm/chemistry , Doxorubicin/chemistry , Carbon Radioisotopes , DNA Adducts , Humans , Kinetics , Tetrahydrofolate Dehydrogenase/genetics , Tumor Cells, Cultured
13.
Anticancer Drug Des ; 15(5): 373-86, 2000 Oct.
Article in English | MEDLINE | ID: mdl-11354313

ABSTRACT

A series of anthracyclines (comprising carminomycins I, II and III, and barminomycin) were tested for their ability to react with DNA to form site-specific adducts using an in vitro transcription assay. The requirement for drug activation by formaldehyde was also assessed using a transcription assay and HPLC analysis of GC-containing oligonucleotide duplexes. In the absence of formaldehyde, barminomycin was the most reactive compound and carminomycin I the least reactive. The DNA sequence specificity of all anthracyclines was similar (the most intense binding sites being 5'-GC sequences), although barminomycin was the most selective for 5'-GC. Barminomycin adducts were the most stable at 37 degrees C (no loss in the 48 h time frame studied) while carminomycin II and III lesions were least stable (each with a half-life of approximately 4-5 h). These results are discussed collectively in terms of the requirement and contribution of structural elements of the anthracyclines for the formation of DNA adducts.


Subject(s)
Antibiotics, Antineoplastic/chemistry , Antibiotics, Antineoplastic/pharmacology , DNA Adducts/chemistry , Anthracyclines/chemistry , Anthracyclines/pharmacology , Base Sequence , Carubicin/chemistry , Carubicin/pharmacology , Chromatography, High Pressure Liquid , Cross-Linking Reagents , DNA Adducts/drug effects , DNA, Neoplasm/chemistry , DNA, Neoplasm/drug effects , Doxorubicin/chemistry , Doxorubicin/pharmacology , Formaldehyde/chemistry , Humans , Molecular Sequence Data , Structure-Activity Relationship , Transcription, Genetic/drug effects
14.
Hum Mol Genet ; 9(2): 187-94, 2000 Jan 22.
Article in English | MEDLINE | ID: mdl-10607829

ABSTRACT

A double-stranded 9 bp GTGAAAAAG pJ alpha sequence found in human centromeric alpha-satellite DNA and a 28 bp ATGTATATATGTGTATATAGACATAAAT tandemly repeated AT28 sequence found within a cloned neo- centromere DNA have each allowed the affinity purification of a nuclear protein that we have identified as poly(ADP-ribose) polymerase (PARP). Use of other related or unrelated oligonucleotide sequences as affinity substrates has indicated either significantly reduced or no detectable PARP purification, suggesting preferential but not absolute sequence-specific binding. Immunofluorescence analysis of human and sheep metaphase cells using a polyclonal anti-PARP antibody revealed centromeric localization of PARP, with diffuse signals also seen on the chromosome arms. Similar results were observed for mouse chromosomes except for a significantly enlarged PARP-binding region around the core centromere-active domain, suggesting possible 'spreading' of PARP into surrounding non-core centromeric domains. Enhanced PARP signals were also observed on alpha-satellite-negative human neo- centromeres and on the active but not the inactive alpha-satellite-containing centromere of a human dicentric chromosome. PARP signals were absent from the q12 heterochromatin of the Y chromosome, suggesting a correlation of PARP binding with centromere function that is independent of heterochromatic properties. Preliminary cell cycle analysis indicates detectable centromeric association of PARP during S/G(2)phase and that the total proportion of PARP that is centromeric is relatively low. Strong binding of PARP to different centromere sequence motifs may offer a versatile mechanism of mammalian centromere recognition that is independent of primary DNA sequences.


Subject(s)
Centromere/enzymology , Metaphase/genetics , Poly(ADP-ribose) Polymerases/metabolism , Animals , Binding Sites/genetics , Cell Line , Centromere/genetics , Centromere/metabolism , Chromatography, Affinity , HeLa Cells , Heterochromatin/enzymology , Humans , Mice , Molecular Weight , Poly(ADP-ribose) Polymerases/genetics , Poly(ADP-ribose) Polymerases/isolation & purification , Sheep
15.
Hum Mol Genet ; 8(7): 1145-55, 1999 Jul.
Article in English | MEDLINE | ID: mdl-10369859

ABSTRACT

INCENP is a chromosomal passenger protein which relocates from the centromere to thel spindle midzone during the metaphase-anaphase transition, ultimately being discarded in the cell midbody at the completion of cytokinesis. Using homologous recombination, we have generated Incenp gene-targeted heterozygous mice that are phenotypically indistinguishable from their wild-type littermates. Intercrossing the hetero-zygotes results in no live-born homozygous Incenp -disrupted progeny, indicating an early lethality. Day 3.5 affected pre-implantation embryos contain large, morphologically abnormal cells that fail to fully develop a blastocoel cavity or thrive in utero and in culture. Chromatin and tubulin immunocytochemical stainings of these and day 2.5 affected embryos reveal a high mitotic index, no discernible metaphase or anaphase stages, complete absence of midbodies, micronuclei formation, morphologically irregular macronuclei with large chromosome complements, multipolar mitotic configurations, binucleated cells, internuclear bridges and abnormal spindle bundling. The phenotype is consistent with a defect in the modulation of microtubule dynamics, severely affecting chromosome segregation and resulting in poorly resolved chromatin masses, aberrant karyokinesis and internuclear bridge formation. These latter occurrences could pose a physical barrier blocking cytokinesis.


Subject(s)
Chromosomal Proteins, Non-Histone/genetics , Chromosome Segregation , Microtubules/physiology , Animals , Cell Nucleus , Chickens , Chromosomal Proteins, Non-Histone/physiology , Chromosomes , Embryonic and Fetal Development/genetics , Embryonic and Fetal Development/physiology , Gene Targeting , Immunohistochemistry , Mice , Mice, Knockout , Mutation , Phenotype , Tubulin/analysis
17.
J Cell Biol ; 141(2): 309-19, 1998 Apr 20.
Article in English | MEDLINE | ID: mdl-9548711

ABSTRACT

CENP-B is a constitutive centromere DNA-binding protein that is conserved in a number of mammalian species and in yeast. Despite this conservation, earlier cytological and indirect experimental studies have provided conflicting evidence concerning the role of this protein in mitosis. The requirement of this protein in meiosis has also not previously been described. To resolve these uncertainties, we used targeted disruption of the Cenpb gene in mouse to study the functional significance of this protein in mitosis and meiosis. Male and female Cenpb null mice have normal body weights at birth and at weaning, but these subsequently lag behind those of the heterozygous and wild-type animals. The weight and sperm content of the testes of Cenpb null mice are also significantly decreased. Otherwise, the animals appear developmentally and reproductively normal. Cytogenetic fluorescence-activated cell sorting and histological analyses of somatic and germline tissues revealed no abnormality. These results indicate that Cenpb is not essential for mitosis or meiosis, although the observed weight reduction raises the possibility that Cenpb deficiency may subtly affect some aspects of centromere assembly and function, and result in reduced rate of cell cycle progression, efficiency of microtubule capture, and/or chromosome movement. A model for a functional redundancy of this protein is presented.


Subject(s)
Autoantigens , Body Weight/genetics , Chromosomal Proteins, Non-Histone/physiology , DNA-Binding Proteins , Meiosis/physiology , Mitosis/physiology , Testis/growth & development , Animals , Centromere/chemistry , Centromere Protein B , Chromosomal Proteins, Non-Histone/analysis , Chromosomal Proteins, Non-Histone/genetics , Female , Karyotyping , Male , Mice , Mice, Knockout , Organ Size , Sperm Count
20.
J Biol Chem ; 271(10): 5422-9, 1996 Mar 08.
Article in English | MEDLINE | ID: mdl-8621397

ABSTRACT

Adriamycin is known to specifically induce DNA interstrand cross-links at 5'-GC sequences. Because 5'-GC sequences are a predominant feature of 5'-untranslated regions (transcription factor-binding sites, promoter, and enhancer regions), it is likely that adriamycin adducts at GC sites would affect the binding of DNA-interacting proteins. Two model systems were chosen for the analysis: the octamer-binding proteins Oct-1, N-Oct-3 and N-Oct-5, which bind to ATGCAAAT and TAATGARAT recognition sites, and Escherichia coli RNA polymerase binding to the lac UV5 promoter. Electrophoretic mobility shift studies showed that adriamycin adducts at GC sites inhibited the binding of octamer proteins to their consensus motifs at drug levels as low as 1 micoM, but no effect was observed with a control sequence lacking a GC site. Adriamycin adducts at GC sites also inhibited the binding of RNA polymerase to the lac UV5 promoter. Adriamycin may therefore function by down-regulating the expression of specific genes by means of inactivation of short but critical motifs containing one or more GC sites.


Subject(s)
Antibiotics, Antineoplastic/pharmacology , DNA Adducts/metabolism , DNA-Binding Proteins/metabolism , DNA-Directed RNA Polymerases/metabolism , DNA/metabolism , Doxorubicin/metabolism , Doxorubicin/pharmacology , Escherichia coli/genetics , Transcription Factors/metabolism , Antibiotics, Antineoplastic/metabolism , Base Sequence , Binding Sites , Consensus Sequence , DNA/drug effects , DNA-Binding Proteins/antagonists & inhibitors , DNA-Directed RNA Polymerases/drug effects , Escherichia coli/enzymology , Homeodomain Proteins/metabolism , Host Cell Factor C1 , Kinetics , Molecular Sequence Data , Octamer Transcription Factor-1 , Octamer Transcription Factor-3 , Plasmids , Promoter Regions, Genetic , Substrate Specificity , Transcription Factors/drug effects
SELECTION OF CITATIONS
SEARCH DETAIL
...