75 results on '"Bjornsti MA"'
Search Results
2. Domain Interactions Affecting Human DNA Topoisomerase I Catalysis and Camptothecin Sensitivity. Molecular Phamacology 56:1105-1115 (1999)
- Author
-
Fiorani, P., Amatruda, Jf, Silvestri, Bjornsti, Ma, Butler, Rh, and Benedetti, Pietro
- Published
- 1999
3. A Small Molecule, UAB126, Reverses Diet-Induced Obesity and its Associated Metabolic Disorders.
- Author
-
Ren G, Kim T, Kim HS, Young ME, Muccio DD, Atigadda VR, Blum SI, Tse HM, Habegger KM, Bhatnagar S, Coric T, Bjornsti MA, Shalev A, Frank SJ, and Kim JA
- Subjects
- Animals, Fatty Liver blood, Hyperlipidemias blood, Lipids blood, Male, Mice, Obesity blood, Diet, High-Fat, Fatty Liver drug therapy, Hyperlipidemias drug therapy, Insulin Resistance physiology, Liver drug effects, Obesity drug therapy, Retinoid X Receptors agonists
- Abstract
Targeting retinoid X receptor (RXR) has been proposed as one of the therapeutic strategies to treat individuals with metabolic syndrome, as RXR heterodimerizes with multiple nuclear receptors that regulate genes involved in metabolism. Despite numerous efforts, RXR ligands (rexinoids) have not been approved for clinical trials to treat metabolic syndrome due to the serious side effects such as hypertriglyceridemia and altered thyroid hormone axis. In this study, we demonstrate a novel rexinoid-like small molecule, UAB126, which has positive effects on metabolic syndrome without the known side effects of potent rexinoids. Oral administration of UAB126 ameliorated obesity, insulin resistance, hepatic steatosis, and hyperlipidemia without changes in food intake, physical activity, and thyroid hormone levels. RNA-sequencing analysis revealed that UAB126 regulates the expression of genes in the liver that are modulated by several nuclear receptors, including peroxisome proliferator-activated receptor α and/or liver X receptor in conjunction with RXR. Furthermore, UAB126 not only prevented but also reversed obesity-associated metabolic disorders. The results suggest that optimized modulation of RXR may be a promising strategy to treat metabolic disorders without side effects. Thus, the current study reveals that UAB126 could be an attractive therapy to treat individuals with obesity and its comorbidities., (© 2020 by the American Diabetes Association.)
- Published
- 2020
- Full Text
- View/download PDF
4. Topoisomerases and cancer chemotherapy: recent advances and unanswered questions.
- Author
-
Bjornsti MA and Kaufmann SH
- Subjects
- DNA, DNA Damage, DNA Replication, Humans, DNA Topoisomerases physiology, Neoplasms
- Abstract
DNA topoisomerases are enzymes that catalyze changes in the torsional and flexural strain of DNA molecules. Earlier studies implicated these enzymes in a variety of processes in both prokaryotes and eukaryotes, including DNA replication, transcription, recombination, and chromosome segregation. Studies performed over the past 3 years have provided new insight into the roles of various topoisomerases in maintaining eukaryotic chromosome structure and facilitating the decatenation of daughter chromosomes at cell division. In addition, recent studies have demonstrated that the incorporation of ribonucleotides into DNA results in trapping of topoisomerase I (TOP1)-DNA covalent complexes during aborted ribonucleotide removal. Importantly, such trapped TOP1-DNA covalent complexes, formed either during ribonucleotide removal or as a consequence of drug action, activate several repair processes, including processes involving the recently described nuclear proteases SPARTAN and GCNA-1. A variety of new TOP1 inhibitors and formulations, including antibody-drug conjugates and PEGylated complexes, exert their anticancer effects by also trapping these TOP1-DNA covalent complexes. Here we review recent developments and identify further questions raised by these new findings., Competing Interests: Competing interests: Dr. Kaufmann indicates that he is the named co-inventor on a patent held by Mayo Clinic regarding the use of antibodies to TOPccs as theranostic reagents. Dr Bjornsti declared that she has no competing interests. No competing interests were disclosed.No competing interests were disclosed.No competing interests were disclosed., (Copyright: © 2019 Bjornsti MA and Kaufmann SH.)
- Published
- 2019
- Full Text
- View/download PDF
5. UBC9 Mutant Reveals the Impact of Protein Dynamics on Substrate Selectivity and SUMO Chain Linkages.
- Author
-
Wright CM, Whitaker RH, Onuiri JE, Blackburn T, McGarity S, Bjornsti MA, and Placzek WJ
- Subjects
- Amino Acid Sequence, Amino Acid Substitution, Catalytic Domain, Cysteine chemistry, Humans, Leucine chemistry, Leucine genetics, Mutation, Proline chemistry, Proline genetics, Saccharomyces cerevisiae chemistry, Sequence Alignment, Substrate Specificity, Sumoylation, Ubiquitin-Conjugating Enzymes chemistry, Ubiquitin-Conjugating Enzymes genetics, Ubiquitin-Conjugating Enzyme UBC9, Saccharomyces cerevisiae Proteins metabolism, Small Ubiquitin-Related Modifier Proteins metabolism, Ubiquitin-Conjugating Enzymes metabolism
- Abstract
SUMO, a conserved ubiquitin-like protein, is conjugated to a multitude of cellular proteins to maintain genomic integrity and resist genotoxic stress. Studies of the SUMO E2 conjugating enzyme mutant, UBC9
P123L , suggested that altered substrate specificity enhances cell sensitivity to DNA damaging agents. Using nuclear magnetic resonance chemical shift studies, we confirm that the mutation does not alter the core globular fold of UBC9, while15 N relaxation measurements demonstrate mutant-induced stabilization of distinct chemical states in residues near the active site cysteine and substrate recognition motifs. We further demonstrate that the P123L substitution induces a switch from the preferential addition of SUMO to lysine residues in unstructured sites to acceptor lysines embedded in secondary structures, thereby also inducing alterations in SUMO chain linkages. Our results provide new insights regarding the impact that structural dynamics of UBC9 have on substrate selection and specifically SUMO chain formation. These findings highlight the potential contribution of nonconsensus SUMO targets and/or alternative SUMO chain linkages on DNA damage response and chemotherapeutic sensitivity.- Published
- 2019
- Full Text
- View/download PDF
6. DNA topoisomerase-targeting chemotherapeutics: what's new?
- Author
-
Cuya SM, Bjornsti MA, and van Waardenburg RCAM
- Subjects
- Animals, DNA Breaks, Drug Design, Humans, Mitochondria enzymology, Mitochondria genetics, Molecular Targeted Therapy, Neoplasms enzymology, Antineoplastic Agents pharmacology, DNA Topoisomerases metabolism, Neoplasms drug therapy
- Abstract
To resolve the topological problems that threaten the function and structural integrity of nuclear and mitochondrial genomes and RNA molecules, human cells encode six different DNA topoisomerases including type IB enzymes (TOP1 and TOP1mt), type IIA enzymes (TOP2α and TOP2β) and type IA enzymes (TOP3α and TOP3β). DNA entanglements and the supercoiling of DNA molecules are regulated by topoisomerases through the introduction of transient enzyme-linked DNA breaks. The covalent topoisomerase-DNA complexes are the cellular targets of a diverse group of cancer chemotherapeutics, which reversibly stabilize these reaction intermediates. Here we review the structure-function and catalytic mechanisms of each family of eukaryotic DNA topoisomerases and the topoisomerase-targeting agents currently approved for patient therapy or in clinical trials, and highlight novel developments and challenges in the clinical development of these agents.
- Published
- 2017
- Full Text
- View/download PDF
7. Molecular Mechanism of DNA Topoisomerase I-Dependent rDNA Silencing: Sir2p Recruitment at Ribosomal Genes.
- Author
-
D'Alfonso A, Di Felice F, Carlini V, Wright CM, Hertz MI, Bjornsti MA, and Camilloni G
- Subjects
- Chromatin Immunoprecipitation, DNA Topoisomerases, Type I genetics, Gene Deletion, Genetic Complementation Test, Protein Binding, Saccharomyces cerevisiae genetics, Transcription, Genetic, DNA Topoisomerases, Type I metabolism, DNA, Ribosomal metabolism, Gene Expression Regulation, Fungal, RNA, Ribosomal biosynthesis, Saccharomyces cerevisiae metabolism, Silent Information Regulator Proteins, Saccharomyces cerevisiae metabolism, Sirtuin 2 metabolism
- Abstract
Saccharomyces cerevisiae sir2Δ or top1Δ mutants exhibit similar phenotypes involving ribosomal DNA, including (i) loss of transcriptional silencing, resulting in non-coding RNA hyperproduction from cryptic RNA polymerase II promoters; (ii) alterations in recombination; and (iii) a general increase in histone acetylation. Given the distinct enzymatic activities of Sir2 and Top1 proteins, a histone deacetylase and a DNA topoisomerase, respectively, we investigated whether genetic and/or physical interactions between the two proteins could explain the shared ribosomal RNA genes (rDNA) phenotypes. We employed an approach of complementing top1Δ cells with yeast, human, truncated, and chimeric yeast/human TOP1 constructs and of assessing the extent of non-coding RNA silencing and histone H4K16 deacetylation. Our findings demonstrate that residues 115-125 within the yeast Top1p N-terminal domain are required for the complementation of the top1∆ rDNA phenotypes. In chromatin immunoprecipitation and co-immunoprecipitation experiments, we further demonstrate the physical interaction between Top1p and Sir2p. Our genetic and biochemical studies support a model whereby Top1p recruits Sir2p to the rDNA and clarifies a structural role of DNA topoisomerase I in the epigenetic regulation of rDNA, independent of its known catalytic activity., (Copyright © 2016 Elsevier Ltd. All rights reserved.)
- Published
- 2016
- Full Text
- View/download PDF
8. Acoustic Droplet Ejection Technology and Its Application in High-Throughput RNA Interference Screening.
- Author
-
Nebane NM, Coric T, McKellip S, Woods L, Sosa M, Rasmussen L, Bjornsti MA, and White EL
- Subjects
- Acoustics, Solutions, Biomedical Technology methods, Genetic Association Studies methods, High-Throughput Screening Assays methods, RNA Interference
- Abstract
The development of acoustic droplet ejection (ADE) technology has resulted in many positive changes associated with the operations in a high-throughput screening (HTS) laboratory. Originally, this liquid transfer technology was used to simply transfer DMSO solutions of primarily compounds. With the introduction of Labcyte's Echo 555, which has aqueous dispense capability, the application of this technology has been expanded beyond its original use. This includes the transfer of many biological reagents solubilized in aqueous buffers, including siRNAs. The Echo 555 is ideal for siRNA dispensing because it is accurate at low volumes and a step-down dilution is not necessary. The potential for liquid carryover and cross-contamination is eliminated, as no tips are needed. Herein, we describe the siRNA screening platform at Southern Research's HTS Center using the ADE technology. With this technology, an siRNA library can be dispensed weeks or even months in advance of the assay itself. The protocol has been optimized to achieve assay parameters comparable to small-molecule screening parameters, and exceeding the norm reported for genomewide siRNA screens., Competing Interests: Declaration of Conflicting Interests The authors declared no potential conflicts of interest with respect to the research, authorship, and/or publication of this article., (© 2015 Society for Laboratory Automation and Screening.)
- Published
- 2016
- Full Text
- View/download PDF
9. Point mutations of the mTOR-RHEB pathway in renal cell carcinoma.
- Author
-
Ghosh AP, Marshall CB, Coric T, Shim EH, Kirkman R, Ballestas ME, Ikura M, Bjornsti MA, and Sudarshan S
- Subjects
- Adaptor Proteins, Signal Transducing genetics, Adaptor Proteins, Signal Transducing metabolism, Biomarkers, Tumor metabolism, Carcinoma, Renal Cell drug therapy, Carcinoma, Renal Cell metabolism, Carcinoma, Renal Cell pathology, Cell Proliferation drug effects, DNA Mutational Analysis, Databases, Genetic, Drug Resistance, Neoplasm genetics, GTPase-Activating Proteins genetics, GTPase-Activating Proteins metabolism, Genetic Predisposition to Disease, HEK293 Cells, Humans, Intracellular Signaling Peptides and Proteins genetics, Intracellular Signaling Peptides and Proteins metabolism, Kidney Neoplasms drug therapy, Kidney Neoplasms metabolism, Kidney Neoplasms pathology, Mechanistic Target of Rapamycin Complex 1, Mechanistic Target of Rapamycin Complex 2, Multiprotein Complexes genetics, Multiprotein Complexes metabolism, Phenotype, Protein Kinase Inhibitors pharmacology, Protein Structure, Tertiary, Ras Homolog Enriched in Brain Protein, Signal Transduction drug effects, Sirolimus pharmacology, TOR Serine-Threonine Kinases antagonists & inhibitors, TOR Serine-Threonine Kinases metabolism, Transfection, Tuberous Sclerosis Complex 2 Protein, Tumor Suppressor Proteins genetics, Tumor Suppressor Proteins metabolism, Biomarkers, Tumor genetics, Carcinoma, Renal Cell genetics, Kidney Neoplasms genetics, Monomeric GTP-Binding Proteins genetics, Neuropeptides genetics, Point Mutation, TOR Serine-Threonine Kinases genetics
- Abstract
Aberrations in the mTOR (mechanistic target of rapamycin) axis are frequently reported in cancer. Using publicly available tumor genome sequencing data, we identified several point mutations in MTOR and its upstream regulator RHEB (Ras homolog enriched in brain) in patients with clear cell renal cell carcinoma (ccRCC), the most common histology of kidney cancer. Interestingly, we found a prominent cluster of hyperactivating mutations in the FAT (FRAP-ATM-TTRAP) domain of mTOR in renal cell carcinoma that led to an increase in both mTORC1 and mTORC2 activities and led to an increased proliferation of cells. Several of the FAT domain mutants demonstrated a decreased binding of DEPTOR (DEP domain containing mTOR-interacting protein), while a subset of these mutations showed altered binding of the negative regulator PRAS40 (proline rich AKT substrate 40). We also identified a recurrent mutation in RHEB in ccRCC patients that leads to an increase in mTORC1 activity. In vitro characterization of this RHEB mutation revealed that this mutant showed considerable resistance to TSC2 (Tuberous Sclerosis 2) GAP (GTPase activating protein) activity, though its interaction with TSC2 remained unaltered. Mutations in the FAT domain of MTOR and in RHEB remained sensitive to rapamycin, though several of these mutations demonstrated residual mTOR kinase activity after treatment with rapamycin at clinically relevant doses. Overall, our data suggests that point mutations in the mTOR pathway may lead to downstream mTOR hyperactivation through multiple different mechanisms to confer a proliferative advantage to a tumor cell.
- Published
- 2015
- Full Text
- View/download PDF
10. DNA topoisomerase I domain interactions impact enzyme activity and sensitivity to camptothecin.
- Author
-
Wright CM, van der Merwe M, DeBrot AH, and Bjornsti MA
- Subjects
- Amino Acid Sequence, Catalysis, Humans, Models, Molecular, Molecular Sequence Data, Protein Structure, Tertiary, Recombinant Fusion Proteins chemistry, Sequence Homology, Amino Acid, Species Specificity, Camptothecin chemistry, DNA Topoisomerases, Type I chemistry, Saccharomyces cerevisiae enzymology, Topoisomerase I Inhibitors chemistry
- Abstract
During processes such as DNA replication and transcription, DNA topoisomerase I (Top1) catalyzes the relaxation of DNA supercoils. The nuclear enzyme is also the cellular target of camptothecin (CPT) chemotherapeutics. Top1 contains four domains: the highly conserved core and C-terminal domains involved in catalysis, a coiled-coil linker domain of variable length, and a poorly conserved N-terminal domain. Yeast and human Top1 share a common reaction mechanism and domain structure. However, the human Top1 is ∼100-fold more sensitive to CPT. Moreover, substitutions of a conserved Gly(717) residue, which alter intrinsic enzyme sensitivity to CPT, induce distinct phenotypes in yeast. To address the structural basis for these differences, reciprocal swaps of yeast and human Top1 domains were engineered in chimeric enzymes. Here we report that intrinsic Top1 sensitivity to CPT is dictated by the composition of the conserved core and C-terminal domains. However, independent of CPT, biochemically similar chimeric enzymes produced strikingly distinct phenotypes in yeast. Expression of a human Top1 chimera containing the yeast linker domain proved toxic, even in the context of a catalytically inactive Y723F enzyme. Lethality was suppressed either by splicing the yeast N-terminal domain into the chimera, deleting the human N-terminal residues, or in enzymes reconstituted by polypeptide complementation. These data demonstrate a functional interaction between the N-terminal and linker domains, which, when mispaired between yeast and human enzymes, induces cell lethality. Because toxicity was independent of enzyme catalysis, the inappropriate coordination of N-terminal and linker domains may induce aberrant Top1-protein interactions to impair cell growth., (© 2015 by The American Society for Biochemistry and Molecular Biology, Inc.)
- Published
- 2015
- Full Text
- View/download PDF
11. Tyrosyl-DNA phosphodiesterase I catalytic mutants reveal an alternative nucleophile that can catalyze substrate cleavage.
- Author
-
Comeaux EQ, Cuya SM, Kojima K, Jafari N, Wanzeck KC, Mobley JA, Bjornsti MA, and van Waardenburg RC
- Subjects
- Catalysis, Catalytic Domain genetics, Crystallography, X-Ray, DNA chemistry, DNA Adducts genetics, DNA Damage genetics, DNA Repair genetics, DNA Topoisomerases, Type I chemistry, DNA Topoisomerases, Type I genetics, Humans, Multiprotein Complexes chemistry, Multiprotein Complexes genetics, Mutant Proteins genetics, Phosphoric Diester Hydrolases chemistry, Spinocerebellar Ataxias genetics, Spinocerebellar Ataxias pathology, DNA genetics, DNA Adducts chemistry, Mutant Proteins chemistry, Phosphoric Diester Hydrolases genetics
- Abstract
Tyrosyl-DNA phosphodiesterase I (Tdp1) catalyzes the repair of 3'-DNA adducts, such as the 3'-phosphotyrosyl linkage of DNA topoisomerase I to DNA. Tdp1 contains two conserved catalytic histidines: a nucleophilic His (His(nuc)) that attacks DNA adducts to form a covalent 3'-phosphohistidyl intermediate and a general acid/base His (His(gab)), which resolves the Tdp1-DNA linkage. A His(nuc) to Ala mutant protein is reportedly inactive, whereas the autosomal recessive neurodegenerative disease SCAN1 has been attributed to the enhanced stability of the Tdp1-DNA intermediate induced by mutation of His(gab) to Arg. However, here we report that expression of the yeast His(nuc)Ala (H182A) mutant actually induced topoisomerase I-dependent cytotoxicity and further enhanced the cytotoxicity of Tdp1 His(gab) mutants, including H432N and the SCAN1-related H432R. Moreover, the His(nuc)Ala mutant was catalytically active in vitro, albeit at levels 85-fold less than that observed with wild type Tdp1. In contrast, the His(nuc)Phe mutant was catalytically inactive and suppressed His(gab) mutant-induced toxicity. These data suggest that the activity of another nucleophile when His(nuc) is replaced with residues containing a small side chain (Ala, Asn, and Gln), but not with a bulky side chain. Indeed, genetic, biochemical, and mass spectrometry analyses show that a highly conserved His, immediately N-terminal to His(nuc), can act as a nucleophile to catalyze the formation of a covalent Tdp1-DNA intermediate. These findings suggest that the flexibility of Tdp1 active site residues may impair the resolution of mutant Tdp1 covalent phosphohistidyl intermediates and provide the rationale for developing chemotherapeutics that stabilize the covalent Tdp1-DNA intermediate., (© 2015 by The American Society for Biochemistry and Molecular Biology, Inc.)
- Published
- 2015
- Full Text
- View/download PDF
12. Development and histopathological characterization of tumorgraft models of pancreatic ductal adenocarcinoma.
- Author
-
Garcia PL, Council LN, Christein JD, Arnoletti JP, Heslin MJ, Gamblin TL, Richardson JH, Bjornsti MA, and Yoon KJ
- Subjects
- Animals, DNA Mutational Analysis, Humans, Mice, Proto-Oncogene Proteins p21(ras) metabolism, Carcinoma, Pancreatic Ductal physiopathology, Disease Models, Animal, Heterografts physiopathology, Pancreatic Neoplasms physiopathology
- Abstract
Pancreatic cancer is the one of the deadliest of all malignancies. The five year survival rate for patients with this disease is 3-5%. Thus, there is a compelling need for novel therapeutic strategies to improve the clinical outcome for patients with pancreatic cancer. Several groups have demonstrated for other types of solid tumors that early passage human tumor xenograft models can be used to define some genetic and molecular characteristics of specific human tumors. Published studies also suggest that murine tumorgraft models (early passage xenografts derived from direct implantation of primary tumor specimens) may be useful in identifying compounds with efficacy against specific tumor types. Because pancreatic cancer is a fatal disease and few well-characterized model systems are available for translational research, we developed and characterized a panel of pancreatic tumorgraft models for biological evaluation and therapeutic drug testing. Of the 41 primary tumor specimens implanted subcutaneously into mice, 35 produced viable tumorgraft models. We document the fidelity of histological and morphological characteristics and of KRAS mutation status among primary (F0), F1, and F2 tumors for the twenty models that have progressed to the F3 generation. Importantly, our procedures produced a take rate of 85%, higher than any reported in the literature. Primary tumor specimens that failed to produce tumorgrafts were those that either contained <10% tumor cells or that were obtained from significantly smaller primary tumors. In view of the fidelity of characteristics of primary tumor specimens through at least the F2 generation in mice, we propose that these tumorgraft models represent a useful tool for identifying critical characteristics of pancreatic tumors and for evaluating potential therapies.
- Published
- 2013
- Full Text
- View/download PDF
13. High-throughput RNA interference screening: tricks of the trade.
- Author
-
Nebane NM, Coric T, Whig K, McKellip S, Woods L, Sosa M, Sheppard R, Rasmussen L, Bjornsti MA, and White EL
- Subjects
- Animals, Genetic Testing instrumentation, HEK293 Cells, Humans, Microfluidic Analytical Techniques standards, Reproducibility of Results, Genetic Testing methods, High-Throughput Screening Assays, RNA Interference, RNA, Small Interfering genetics
- Abstract
The process of validating an assay for high-throughput screening (HTS) involves identifying sources of variability and developing procedures that minimize the variability at each step in the protocol. The goal is to produce a robust and reproducible assay with good metrics. In all good cell-based assays, this means coefficient of variation (CV) values of less than 10% and a signal window of fivefold or greater. HTS assays are usually evaluated using Z' factor, which incorporates both standard deviation and signal window. A Z' factor value of 0.5 or higher is acceptable for HTS. We used a standard HTS validation procedure in developing small interfering RNA (siRNA) screening technology at the HTS center at Southern Research. Initially, our assay performance was similar to published screens, with CV values greater than 10% and Z' factor values of 0.51 ± 0.16 (average ± standard deviation). After optimizing the siRNA assay, we got CV values averaging 7.2% and a robust Z' factor value of 0.78 ± 0.06 (average ± standard deviation). We present an overview of the problems encountered in developing this whole-genome siRNA screening program at Southern Research and how equipment optimization led to improved data quality., Competing Interests: The authors declared no potential conflicts of interest with respect to the research, authorship, and/or publication of this article.
- Published
- 2013
- Full Text
- View/download PDF
14. 4-Hydroxytamoxifen induces autophagic death through K-Ras degradation.
- Author
-
Kohli L, Kaza N, Coric T, Byer SJ, Brossier NM, Klocke BJ, Bjornsti MA, Carroll SL, and Roth KA
- Subjects
- Autophagy genetics, Caspases genetics, Caspases metabolism, Cell Death genetics, Cell Line, Tumor, Down-Regulation drug effects, ErbB Receptors genetics, ErbB Receptors metabolism, HCT116 Cells, Humans, MCF-7 Cells, Mitogen-Activated Protein Kinase Kinases genetics, Mitogen-Activated Protein Kinase Kinases metabolism, Nerve Sheath Neoplasms enzymology, Nerve Sheath Neoplasms metabolism, Nerve Sheath Neoplasms pathology, Protein Kinase C genetics, Protein Kinase C metabolism, Proteolysis, Proto-Oncogene Proteins genetics, Proto-Oncogene Proteins p21(ras), Receptors, Estrogen genetics, Receptors, Estrogen metabolism, Tamoxifen pharmacology, ras Proteins genetics, Autophagy drug effects, Cell Death drug effects, Nerve Sheath Neoplasms drug therapy, Proto-Oncogene Proteins metabolism, Tamoxifen analogs & derivatives, ras Proteins metabolism
- Abstract
Tamoxifen is widely used to treat estrogen receptor-positive breast cancer. Recent findings that tamoxifen and its derivative 4-hydroxytamoxifen (OHT) can exert estrogen receptor-independent cytotoxic effects have prompted the initiation of clinical trials to evaluate its use in estrogen receptor-negative malignancies. For example, tamoxifen and OHT exert cytotoxic effects in malignant peripheral nerve sheath tumors (MPNST) where estrogen is not involved. In this study, we gained insights into the estrogen receptor-independent cytotoxic effects of OHT by studying how it kills MPNST cells. Although caspases were activated following OHT treatment, caspase inhibition provided no protection from OHT-induced death. Rather, OHT-induced death in MPNST cells was associated with autophagic induction and attenuated by genetic inhibition of autophagic vacuole formation. Mechanistic investigations revealed that OHT stimulated autophagic degradation of K-Ras, which is critical for survival of MPNST cells. Similarly, we found that OHT induced K-Ras degradation in breast, colon, glioma, and pancreatic cancer cells. Our findings describe a novel mechanism of autophagic death triggered by OHT in tumor cells that may be more broadly useful clinically in cancer treatment., (©2013 AACR.)
- Published
- 2013
- Full Text
- View/download PDF
15. Cellular strategies for regulating DNA supercoiling: a single-molecule perspective.
- Author
-
Koster DA, Crut A, Shuman S, Bjornsti MA, and Dekker NH
- Subjects
- Animals, Cell Physiological Phenomena, DNA chemistry, DNA metabolism, DNA, Superhelical metabolism, Humans, DNA Topoisomerases, Type I metabolism, DNA, Superhelical chemistry
- Abstract
Entangling and twisting of cellular DNA (i.e., supercoiling) are problems inherent to the helical structure of double-stranded DNA. Supercoiling affects transcription, DNA replication, and chromosomal segregation. Consequently the cell must fine-tune supercoiling to optimize these key processes. Here, we summarize how supercoiling is generated and review experimental and theoretical insights into supercoil relaxation. We distinguish between the passive dissipation of supercoils by diffusion and the active removal of supercoils by topoisomerase enzymes. We also review single-molecule studies that elucidate the timescales and mechanisms of supercoil removal., (Copyright 2010 Elsevier Inc. All rights reserved.)
- Published
- 2010
- Full Text
- View/download PDF
16. Disulfide cross-links reveal conserved features of DNA topoisomerase I architecture and a role for the N terminus in clamp closure.
- Author
-
Palle K, Pattarello L, van der Merwe M, Losasso C, Benedetti P, and Bjornsti MA
- Subjects
- DNA Topoisomerases, Type I genetics, DNA Topoisomerases, Type I metabolism, Disulfides metabolism, Humans, Protein Structure, Tertiary physiology, Saccharomyces cerevisiae genetics, Saccharomyces cerevisiae Proteins genetics, Saccharomyces cerevisiae Proteins metabolism, DNA Breaks, DNA Topoisomerases, Type I chemistry, Disulfides chemistry, Saccharomyces cerevisiae enzymology, Saccharomyces cerevisiae Proteins chemistry
- Abstract
In eukaryotes, DNA topoisomerase I (Top1) catalyzes the relaxation of supercoiled DNA by a conserved mechanism of transient DNA strand breakage, rotation, and religation. The unusual architecture of the monomeric human enzyme comprises a conserved protein clamp, which is tightly wrapped about duplex DNA, and an extended coiled-coil linker domain that appropriately positions the C-terminal active site tyrosine domain against the Top1 core to form the catalytic pocket. A structurally undefined N-terminal domain, dispensable for enzyme activity, mediates protein-protein interactions. Previously, reversible disulfide bonds were designed to assess whether locking the Top1 clamp around duplex DNA would restrict DNA strand rotation within the covalent Top1-DNA intermediate. The active site proximal disulfide bond in full-length Top1-clamp(534) restricted DNA rotation (Woo, M. H., Losasso, C., Guo, H., Pattarello, L., Benedetti, P., and Bjornsti, M. A. (2003) Proc. Natl. Acad. Sci. U. S. A. 100, 13767-13772), whereas the more distal disulfide bond of the N-terminally truncated Topo70-clamp(499) did not (Carey, J. F., Schultz, S. J., Sisson, L., Fazzio, T. G., and Champoux, J. J. (2003) Proc. Natl. Acad. Sci. U. S. A. 100, 5640-5645). To assess the contribution of the N-terminal domain to the dynamics of Top1 clamping of DNA, the same disulfide bonds were engineered into full-length Top1 and truncated Topo70, and the activities of these proteins were assessed in vitro and in yeast. Here we report that the N terminus impacts the opening and closing of the Top1 protein clamp. We also show that the architecture of yeast and human Top1 is conserved in so far as cysteine substitutions of the corresponding residues suffice to lock the Top1-clamp. However, the composition of the divergent N-terminal/linker domains impacts Top1-clamp activity and stability in vivo.
- Published
- 2008
- Full Text
- View/download PDF
17. Mutation of Gly721 alters DNA topoisomerase I active site architecture and sensitivity to camptothecin.
- Author
-
van der Merwe M and Bjornsti MA
- Subjects
- Base Sequence, Binding Sites, Catalysis, Catalytic Domain, Crystallography, X-Ray methods, Enzyme Inhibitors pharmacology, Models, Molecular, Molecular Conformation, Molecular Sequence Data, Protein Structure, Tertiary, Camptothecin pharmacology, DNA Topoisomerases, Type I chemistry, Glycine chemistry, Mutation, Saccharomyces cerevisiae metabolism
- Abstract
DNA topoisomerase I (Top1p) catalyzes the relaxation of supercoiled DNA via a concerted mechanism of DNA strand cleavage and religation. Top1p is the cellular target of the anti-cancer drug camptothecin (CPT), which reversibly stabilizes a covalent enzyme-DNA intermediate. Top1p clamps around duplex DNA, wherein the core and C-terminal domains are connected by extended alpha-helices (linker domain), which position the active site Tyr of the C-terminal domain within the catalytic pocket. The physical connection of the linker with the Top1p clamp as well as linker flexibility affect enzyme sensitivity to CPT. Crystallographic data reveal that a conserved Gly residue (located at the juncture between the linker and C-terminal domains) is at one end of a short alpha-helix, which extends to the active site Tyr covalently linked to the DNA. In the presence of drug, the linker is rigid and this alpha-helix extends to include Gly and the preceding Leu. We report that mutation of this conserved Gly in yeast Top1p alters enzyme sensitivity to CPT. Mutating Gly to Asp, Glu, Asn, Gln, Leu, or Ala enhanced enzyme CPT sensitivity, with the acidic residues inducing the greatest increase in drug sensitivity in vivo and in vitro. By contrast, Val or Phe substituents rendered the enzyme CPT-resistant. Mutation-induced alterations in enzyme architecture preceding the active site Tyr suggest these structural transitions modulate enzyme sensitivity to CPT, while enhancing the rate of DNA cleavage. We postulate that this conserved Gly residue provides a flexible hinge within the Top1p catalytic pocket to facilitate linker dynamics and the structural alterations that accompany drug binding of the covalent enzyme-DNA intermediate.
- Published
- 2008
- Full Text
- View/download PDF
18. TOR signaling is a determinant of cell survival in response to DNA damage.
- Author
-
Shen C, Lancaster CS, Shi B, Guo H, Thimmaiah P, and Bjornsti MA
- Subjects
- Adult, Cell Cycle Proteins genetics, Cell Cycle Proteins metabolism, Checkpoint Kinase 2, Cycloheximide metabolism, Humans, Methyl Methanesulfonate metabolism, Multiprotein Complexes, Mutagens metabolism, Protein Serine-Threonine Kinases genetics, Protein Serine-Threonine Kinases metabolism, Protein Subunits genetics, Protein Subunits metabolism, Ribonucleoside Diphosphate Reductase genetics, Ribonucleoside Diphosphate Reductase metabolism, S Phase physiology, Saccharomyces cerevisiae cytology, Saccharomyces cerevisiae genetics, Saccharomyces cerevisiae metabolism, Saccharomyces cerevisiae Proteins genetics, Sirolimus metabolism, Transcription Factors genetics, Transcription Factors metabolism, Cell Survival, DNA Damage, Saccharomyces cerevisiae Proteins metabolism, Signal Transduction physiology
- Abstract
The conserved TOR (target of rapamycin) kinase is part of a TORC1 complex that regulates cellular responses to environmental stress, such as amino acid starvation and hypoxia. Dysregulation of Akt-TOR signaling has also been linked to the genesis of cancer, and thus, this pathway presents potential targets for cancer chemotherapeutics. Here we report that rapamycin-sensitive TORC1 signaling is required for the S-phase progression and viability of yeast cells in response to genotoxic stress. In the presence of the DNA-damaging agent methyl methanesulfonate (MMS), TOR-dependent cell survival required a functional S-phase checkpoint. Rapamycin inhibition of TORC1 signaling suppressed the Rad53 checkpoint-mediated induction of ribonucleotide reductase subunits Rnr1 and Rnr3, thereby abrogating MMS-induced mutagenesis and enhancing cell lethality. Moreover, cells deleted for RNR3 were hypersensitive to rapamycin plus MMS, providing the first demonstration that Rnr3 contributes to the survival of cells exposed to DNA damage. Our findings support a model whereby TORC1 acts as a survival pathway in response to genotoxic stress by maintaining the deoxynucleoside triphosphate pools necessary for error-prone translesion DNA polymerases. Thus, TOR-dependent cell survival in response to DNA-damaging agents coincides with increased mutation rates, which may contribute to the acquisition of chemotherapeutic drug resistance.
- Published
- 2007
- Full Text
- View/download PDF
19. Mutation of a conserved active site residue converts tyrosyl-DNA phosphodiesterase I into a DNA topoisomerase I-dependent poison.
- Author
-
He X, van Waardenburg RCAM, Babaoglu K, Price AC, Nitiss KC, Nitiss JL, Bjornsti MA, and White SW
- Subjects
- Amino Acid Sequence, Animals, Crystallography, X-Ray, DNA Adducts, Humans, Models, Molecular, Molecular Sequence Data, Molecular Structure, Sequence Alignment, Substrate Specificity, Binding Sites, DNA Topoisomerases, Type I metabolism, Mutation, Phosphoric Diester Hydrolases chemistry, Phosphoric Diester Hydrolases genetics, Phosphoric Diester Hydrolases metabolism, Phosphoric Diester Hydrolases toxicity, Protein Structure, Tertiary, Saccharomyces cerevisiae Proteins chemistry, Saccharomyces cerevisiae Proteins genetics, Saccharomyces cerevisiae Proteins metabolism, Saccharomyces cerevisiae Proteins toxicity
- Abstract
Tyrosyl-DNA phosphodiesterase 1 (Tdp1) catalyzes the resolution of 3' and 5' phospho-DNA adducts. A defective mutant, associated with the recessive neurodegenerative disease SCAN1, accumulates Tdp1-DNA complexes in vitro. To assess the conservation of enzyme architecture, a 2.0 A crystal structure of yeast Tdp1 was determined that is very similar to human Tdp1. Poorly conserved regions of primary structure are peripheral to an essentially identical catalytic core. Enzyme mechanism was also conserved, because the yeast SCAN1 mutant (H(432)R) enhanced cell sensitivity to the DNA topoisomerase I (Top1) poison camptothecin. A more severe Top1-dependent lethality of Tdp1H(432)N was drug-independent, coinciding with increased covalent Top1-DNA and Tdp1-DNA complex formation in vivo. However, both H(432) mutants were recessive to wild-type Tdp1. Thus, yeast H(432) acts in the general acid/base catalytic mechanism of Tdp1 to resolve 3' phosphotyrosyl and 3' phosphoamide linkages. However, the distinct pattern of mutant Tdp1 activity evident in yeast cells, suggests a more severe defect in Tdp1H(432)N-catalyzed resolution of 3' phospho-adducts.
- Published
- 2007
- Full Text
- View/download PDF
20. Antitumour drugs impede DNA uncoiling by topoisomerase I.
- Author
-
Koster DA, Palle K, Bot ES, Bjornsti MA, and Dekker NH
- Subjects
- Humans, Magnetics, Nanotechnology, Saccharomyces cerevisiae genetics, DNA Topoisomerases, Type I metabolism, DNA, Superhelical metabolism, Enzyme Inhibitors pharmacology, Topoisomerase I Inhibitors, Topotecan pharmacology
- Abstract
Increasing the ability of chemotherapeutic drugs to kill cancer cells is often hampered by a limited understanding of their mechanism of action. Camptothecins, such as topotecan, induce cell death by poisoning DNA topoisomerase I, an enzyme capable of removing DNA supercoils. Topotecan is thought to stabilize a covalent topoisomerase-DNA complex, rendering it an obstacle to DNA replication forks. Here we use single-molecule nanomanipulation to monitor the dynamics of human topoisomerase I in the presence of topotecan. This allowed us to detect the binding and unbinding of an individual topotecan molecule in real time and to quantify the drug-induced trapping of topoisomerase on DNA. Unexpectedly, our findings also show that topotecan significantly hinders topoisomerase-mediated DNA uncoiling, with a more pronounced effect on the removal of positive (overwound) versus negative supercoils. In vivo experiments in the budding yeast verified the resulting prediction that positive supercoils would accumulate during transcription and replication as a consequence of camptothecin poisoning of topoisomerase I. Positive supercoils, however, were not induced by drug treatment of cells expressing a catalytically active, camptothecin-resistant topoisomerase I mutant. This combination of single-molecule and in vivo data suggests a cytotoxic mechanism for camptothecins, in which the accumulation of positive supercoils ahead of the replication machinery induces potentially lethal DNA lesions.
- Published
- 2007
- Full Text
- View/download PDF
21. Structure of a SUMO-binding-motif mimic bound to Smt3p-Ubc9p: conservation of a non-covalent ubiquitin-like protein-E2 complex as a platform for selective interactions within a SUMO pathway.
- Author
-
Duda DM, van Waardenburg RC, Borg LA, McGarity S, Nourse A, Waddell MB, Bjornsti MA, and Schulman BA
- Subjects
- Amino Acid Sequence, Catalytic Domain, Cloning, Molecular, Crystallography, X-Ray, Cysteine chemistry, Humans, Molecular Conformation, Molecular Sequence Data, Protein Binding, Saccharomyces cerevisiae metabolism, Sequence Homology, Amino Acid, Small Ubiquitin-Related Modifier Proteins, Ubiquitin-Conjugating Enzyme UBC9, Proteins chemistry, Repressor Proteins chemistry, Saccharomyces cerevisiae Proteins chemistry, Ubiquitin chemistry, Ubiquitin-Conjugating Enzymes chemistry
- Abstract
The SUMO ubiquitin-like proteins play regulatory roles in cell division, transcription, DNA repair, and protein subcellular localization. Paralleling other ubiquitin-like proteins, SUMO proteins are proteolytically processed to maturity, conjugated to targets by E1-E2-E3 cascades, and subsequently recognized by specific downstream effectors containing a SUMO-binding motif (SBM). SUMO and its E2 from the budding yeast Saccharomyces cerevisiae, Smt3p and Ubc9p, are encoded by essential genes. Here we describe the 1.9 A resolution crystal structure of a non-covalent Smt3p-Ubc9p complex. Unexpectedly, a heterologous portion of the crystallized complex derived from the expression construct mimics an SBM, and binds Smt3p in a manner resembling SBM binding to human SUMO family members. In the complex, Smt3p binds a surface distal from Ubc9's catalytic cysteine. The structure implies that a single molecule of Smt3p cannot bind concurrently to both the non-covalent binding site and the catalytic cysteine of a single Ubc9p molecule. However, formation of higher-order complexes can occur, where a single Smt3p covalently linked to one Ubc9p's catalytic cysteine also binds non-covalently to another molecule of Ubc9p. Comparison with other structures from the SUMO pathway suggests that formation of the non-covalent Smt3p-Ubc9p complex occurs mutually exclusively with many other Smt3p and Ubc9p interactions in the conjugation cascade. By contrast, high-resolution insights into how Smt3p-Ubc9p can also interact with downstream recognition machineries come from contacts with the SBM mimic. Interestingly, the overall architecture of the Smt3p-Ubc9p complex is strikingly similar to recent structures from the ubiquitin pathway. The results imply that non-covalent ubiquitin-like protein-E2 complexes are conserved platforms, which function as parts of larger assemblies involved in many protein post-translational regulatory pathways.
- Published
- 2007
- Full Text
- View/download PDF
22. Inhibition of topoisomerase I cleavage activity by thiol-reactive compounds: importance of vicinal cysteines 504 and 505.
- Author
-
Montaudon D, Palle K, Rivory LP, Robert J, Douat-Casassus C, Quideau S, Bjornsti MA, and Pourquier P
- Subjects
- Arsenicals pharmacology, Camptothecin pharmacology, DNA Topoisomerases, Type I genetics, DNA Topoisomerases, Type I metabolism, Ethylmaleimide pharmacology, Humans, Mutagenesis, Site-Directed, Protein Conformation, Cysteine genetics, DNA Cleavage drug effects, Enzyme Inhibitors pharmacology, Sulfhydryl Compounds pharmacology, Topoisomerase I Inhibitors
- Abstract
DNA topoisomerase I (Top1) is a nuclear enzyme that plays a crucial role in the removal of DNA supercoiling associated with replication and transcription. It is also the target of the anticancer agent, camptothecin (CPT). Top1 contains eight cysteines, including two vicinal residues (504 and 505), which are highly conserved across species. In this study, we show that thiol-reactive compounds such as N-ethylmaleimide and phenylarsine oxide can impair Top1 catalytic activity. We demonstrate that in contrast to CPT, which inhibits Top1-catalyzed religation, thiolation of Top1 inhibited the DNA cleavage step of the reaction. This inhibition was more pronounced when Top1 was preincubated with the thiol-reactive compound and could be reversed in the presence of dithiothreitol. We also established that phenylarsine oxide-mediated inhibition of Top1 cleavage involved the two vicinal cysteines 504 and 505, as this effect was suppressed when cysteines were mutated to alanines. Interestingly, mutation of Cys-505 also altered Top1 sensitivity to CPT, even in the context of the double Cys-504 to Cys-505 mutant, which relaxed supercoiled DNA with a comparable efficiency to that of wild-type Top1. This indicates that cysteine 505, which is located in the lower Lip domain of human Top1, is critical for optimal poisoning of the enzyme by CPT and its analogs. Altogether, our results suggest that conserved vicinal cysteines 504 and 505 of human Top1 play a critical role in enzyme catalytic activity and are the target of thiol-reactive compounds, which may be developed as efficient Top1 catalytic inhibitors.
- Published
- 2007
- Full Text
- View/download PDF
23. Alterations in linker flexibility suppress DNA topoisomerase I mutant-induced cell lethality.
- Author
-
Losasso C, Cretaio E, Palle K, Pattarello L, Bjornsti MA, and Benedetti P
- Subjects
- Amino Acid Substitution drug effects, Apoptosis drug effects, Apoptosis genetics, Base Sequence, Camptothecin pharmacology, Cell Death, DNA Topoisomerases, Type I genetics, Drug Resistance, Fungal genetics, Molecular Sequence Data, Saccharomyces cerevisiae drug effects, Saccharomyces cerevisiae enzymology, Amino Acid Substitution genetics, Apoptosis physiology, DNA Topoisomerases, Type I physiology, Mutation, Topoisomerase I Inhibitors
- Abstract
Eukaryotic DNA topoisomerase I (Top1p) catalyzes changes in DNA topology via the formation of a covalent enzyme-DNA intermediate, which is reversibly stabilized by the anticancer agent camptothecin (CPT). Crystallographic studies of the 70-kDa C terminus of human Top1p bound to duplex DNA describe a monomeric protein clamp circumscribing the DNA helix. The structures, which lack the N-terminal domain, comprise the conserved clamp, an extended linker domain, and the conserved C-terminal active site Tyr domain. CPT bound to the covalent Top1p-DNA complex limits linker flexibility, allowing structural determination of this domain. We previously reported that mutation of Ala(653) to Pro in the linker increases the rate of enzyme-catalyzed DNA religation, thereby rendering Top1A653Pp resistant to CPT (Fiorani, P., Bruselles, A., Falconi, M., Chillemi, G., Desideri, A., and Benedetti P. (2003) J. Biol. Chem. 278, 43268-43275). Molecular dynamics studies suggested mutation-induced increases in linker flexibility alter Top1p catalyzed DNA religation. To address the functional consequences of linker flexibility on enzyme catalysis and drug sensitivity, we investigated the interactions of the A653P linker mutation with a self-poisoning T718A mutation within the active site of Top1p. The A653P mutation suppressed the lethal phenotype of Top1T718Ap in yeast, yet did not restore enzyme sensitivity to CPT. However, the specific activity of the double mutant was decreased in vivo and in vitro, consistent with a decrease in DNA binding. These findings support a model where changes in the flexibility or orientation of the linker alter the geometry of the active site and thereby the kinetics of DNA cleavage/religation catalyzed by Top1p.
- Published
- 2007
- Full Text
- View/download PDF
24. Trapping of DNA topoisomerase I on nick-containing DNA in cell free extracts of Saccharomyces cerevisiae.
- Author
-
Lebedeva N, Auffret Vander Kemp P, Bjornsti MA, Lavrik O, and Boiteux S
- Subjects
- Affinity Labels, Base Sequence, Cell-Free System, Cross-Linking Reagents, DNA Damage, DNA Polymerase beta metabolism, DNA Repair, DNA, Fungal chemistry, DNA, Fungal genetics, Poly(ADP-ribose) Polymerases metabolism, Saccharomyces cerevisiae genetics, Saccharomyces cerevisiae Proteins metabolism, Substrate Specificity, DNA Topoisomerases, Type I metabolism, DNA, Fungal metabolism, Saccharomyces cerevisiae metabolism
- Abstract
The aim of the present study was to identify proteins that bind nicked DNA intermediates formed in the course of base excision repair (BER) in cell free extracts of Saccharomyces cerevisiae. In mammalian cells, nicks in DNA are targets of proteins such as PARP-1 or XRCC1 that have no homologues in yeast. One of the most promising methodologies to trap proteins that interact with damaged DNA lies in using a photocrosslinking technique with photoactivable dNTP analogues such as exo-N-{2-[N-(4-azido-2,5-difluoro-3-chloropyridine-6-yl)-3-aminopropionyl]-aminoethyl}-2'-deoxycytidine-5'-triphosphate (FAP-dCTP) for enzymatic synthesis of DNA probes with a photoreactive dNMP residue at the 3'-margin of a nick. Using this approach, we identified a major covalent DNA-protein adduct between a nick-containing 34-mer DNA duplex and a protein of a molecular mass of around 100-kDa. Unexpectedly, the formation of the 100-kDa adduct did not require the incorporation of the photoreactive dNMP residue at the 3'-margin of the nick nor exposure to near UV-light. However, the formation of the 100-kDa adduct strictly required a nick or a short gap in the DNA probe. Furthermore, the 100-kDa adduct was not detected in yeast extracts lacking DNA topoisomerase I (Top1). To further establish the nature of crosslinked protein, yeast Top1 was tagged with a Myc-epitope. In this case, the mobility of the Top1-DNA adduct increased by 7- kDa. Therefore, our data speak in favor of Top1 trapping by nicked DNA. In support of this hypothesis, purified yeast Top1 was also crosslinked to nicked DNA structures. Undamaged, uracil- and abasic (AP) site-containing DNAs were unable to trap Top1 under the same assay conditions. Since nicked DNA structures are frequently formed in the course of BER, their covalent linkage to Top1 has the potential to interfere with BER in vivo.
- Published
- 2006
- Full Text
- View/download PDF
25. Distinct functional domains of Ubc9 dictate cell survival and resistance to genotoxic stress.
- Author
-
van Waardenburg RC, Duda DM, Lancaster CS, Schulman BA, and Bjornsti MA
- Subjects
- Amino Acid Sequence, Humans, Molecular Sequence Data, Protein Structure, Tertiary genetics, Recombinant Fusion Proteins chemistry, Recombinant Fusion Proteins genetics, Saccharomyces cerevisiae cytology, Saccharomyces cerevisiae genetics, Saccharomyces cerevisiae Proteins genetics, Saccharomyces cerevisiae Proteins metabolism, Substrate Specificity, Ubiquitin metabolism, Ubiquitin-Conjugating Enzymes genetics, Ubiquitin-Conjugating Enzymes metabolism, Ubiquitin-Protein Ligases metabolism, X-Ray Diffraction, Ubiquitin-Conjugating Enzyme UBC9, DNA Damage genetics, Saccharomyces cerevisiae enzymology, Saccharomyces cerevisiae Proteins chemistry, Ubiquitin-Conjugating Enzymes chemistry
- Abstract
Covalent modification with SUMO alters protein function, intracellular localization, or protein-protein interactions. Target recognition is determined, in part, by the SUMO E2 enzyme, Ubc9, while Siz/Pias E3 ligases may facilitate select interactions by acting as substrate adaptors. A yeast conditional Ubc9P(123)L mutant was viable at 36 degrees C yet exhibited enhanced sensitivity to DNA damage. To define functional domains in Ubc9 that dictate cellular responses to genotoxic stress versus those necessary for cell viability, a 1.75-A structure of yeast Ubc9 that demonstrated considerable conservation of backbone architecture with human Ubc9 was solved. Nevertheless, differences in side chain geometry/charge guided the design of human/yeast chimeras, where swapping domains implicated in (i) binding residues within substrates that flank canonical SUMOylation sites, (ii) interactions with the RanBP2 E3 ligase, and (iii) binding of the heterodimeric E1 and SUMO had distinct effects on cell growth and resistance to DNA-damaging agents. Our findings establish a functional interaction between N-terminal and substrate-binding domains of Ubc9 and distinguish the activities of E3 ligases Siz1 and Siz2 in regulating cellular responses to genotoxic stress.
- Published
- 2006
- Full Text
- View/download PDF
26. Defects in SUMO (small ubiquitin-related modifier) conjugation and deconjugation alter cell sensitivity to DNA topoisomerase I-induced DNA damage.
- Author
-
Jacquiau HR, van Waardenburg RC, Reid RJ, Woo MH, Guo H, Johnson ES, and Bjornsti MA
- Subjects
- Saccharomyces cerevisiae genetics, Saccharomyces cerevisiae metabolism, Saccharomyces cerevisiae Proteins genetics, Saccharomyces cerevisiae Proteins metabolism, DNA Damage, DNA Topoisomerases, Type I metabolism, SUMO-1 Protein metabolism
- Abstract
Eukaryotic DNA topoisomerase I (Top1p) has important functions in DNA replication, transcription, and recombination. This enzyme also constitutes the cellular target of camptothecin (CPT), which induces S-phase-dependent cytotoxicity. To define cellular pathways that regulate cell sensitivity to Top1p-induced DNA lesions, we described a yeast genetic screen for conditional tah (top1T722A-hypersensitive) mutants with enhanced sensitivity to low levels of the CPT mimetic mutant top1T722A (Reid, R. J., Fiorani, P., Sugawara, M., and Bjornsti, M. A. (1999) Proc. Natl. Acad. Sci. U. S. A. 96, 11440-11445; Fiorani, P., Reid, R. J., Schepis, A., Jacquiau, H. R., Guo, H., Thimmaiah, P., Benedetti, P., and Bjornsti, M. A. (2004) J. Biol. Chem. 279, 21271-21281). Here we report that tah mutant ubc9-10 harbors a hypomorphic allele of UBC9, which encodes the essential SUMO (small ubiquitin-related modifier) E2-conjugating enzyme. The same conditional ubc9P123L mutant was also isolated in an independent screen for enhanced sensitivity to a distinct Top1p poison, Top1N726Hp. The ubc9-10 mutant exhibited a decrease in global protein sumoylation and increased sensitivity to a wide range of DNA-damaging agents independent of Top1p. Deletion of the Ulp2 SUMO protease failed to restore ubc9-10 cell resistance to Top1p poisons or hydroxyurea yet adversely affected wild-type TOP1 cell genetic stability and sensitivity to hydroxyurea. Moreover, although mutation of different consensus SUMO sites in the N terminus and linker region of yeast Top1p failed to recapitulate ubc9-10 mutant phenotypes, they revealed distinct and subtle effects on cell sensitivity to CPT. These results provide insights into the complexities of SUMO conjugation and the confounding effects of SUMO modification on Top1p function and cell sensitivity to genotoxic agents.
- Published
- 2005
- Full Text
- View/download PDF
27. Enhanced antitumor activity of irofulven in combination with irinotecan in pediatric solid tumor xenograft models.
- Author
-
Woo MH, Peterson JK, Billups C, Liang H, Bjornsti MA, and Houghton PJ
- Subjects
- Animals, Antineoplastic Agents, Phytogenic administration & dosage, Camptothecin administration & dosage, Drug Administration Schedule, Female, Irinotecan, Mice, Mice, Inbred ICR, Sesquiterpenes administration & dosage, Treatment Failure, Antineoplastic Combined Chemotherapy Protocols therapeutic use, Brain Neoplasms drug therapy, Camptothecin analogs & derivatives, Xenograft Model Antitumor Assays methods
- Abstract
Purpose: Irofulven, a novel chemotherapeutic agent with a broad spectrum of activity, is effective against preclinical models of pediatric tumors. The cytotoxic activity of irofulven is augmented when combined with agents that interact with DNA topoisomerase I; however, none of the reported studies have used the protracted dosing schedule found to be active clinically in treatment of childhood cancers. The objective of this study was to evaluate the antitumor activity of irofulven in combination with irinotecan administered on a protracted schedule in a panel of pediatric solid tumor xenografts., Methods: Irofulven and irinotecan were evaluated alone or in combination against eight independent xenografts, which included childhood brain tumors (n=5), neuroblastoma (n=1), and rhabdomyosarcoma (n=2). Irofulven was administered i.v. daily for 5 days with courses repeated every 21 days for a total of three cycles. Doses of irofulven ranged from 1.33 to 4.6 mg/kg. Irinotecan was given i.v. daily for 5 days each week for 2 weeks repeated every 21 days for three cycles at doses between 0.28 and 1.25 mg/kg., Results: Irofulven and irinotecan, given as single agents, induced few responses in pediatric solid tumor xenografts at the selected doses. At the same doses, irofulven in combination with irinotecan demonstrated superior antitumor activity, inducing complete responses in seven of the eight xenograft lines., Conclusions: These studies show that the cytotoxic activity of irofulven is greater when combined with protracted administration of irinotecan. Although the systemic exposure of irofulven required to induce objective responses in this panel of pediatric solid tumors was in excess of that achievable in patients receiving maximally tolerated doses using this schedule of drug administration, the enhanced activity of irofulven in combination with irinotecan supports the pursuit of alternative administration strategies and combinations.
- Published
- 2005
- Full Text
- View/download PDF
28. Platinated DNA adducts enhance poisoning of DNA topoisomerase I by camptothecin.
- Author
-
van Waardenburg RC, de Jong LA, van Eijndhoven MA, Verseyden C, Pluim D, Jansen LE, Bjornsti MA, and Schellens JH
- Subjects
- Breast Neoplasms, Cisplatin pharmacology, Colonic Neoplasms, Cytarabine pharmacology, DNA chemistry, DNA Adducts chemistry, DNA Topoisomerases, Type I chemistry, Drug Synergism, Female, Humans, Models, Molecular, Ovarian Neoplasms, Topotecan chemistry, Topotecan pharmacology, Tumor Cells, Cultured, Antineoplastic Agents, Phytogenic pharmacology, Camptothecin pharmacology, DNA Adducts pharmacology, Enzyme Inhibitors pharmacology, Platinum Compounds pharmacology, Topoisomerase I Inhibitors
- Abstract
Camptothecins constitute a novel class of chemotherapeutics that selectively target DNA topoisomerase I (Top1) by reversibly stabilizing a covalent enzyme-DNA intermediate. This cytotoxic mechanism contrasts with that of platinum drugs, such as cisplatin, which induce inter- and intrastrand DNA adducts. In vitro combination studies using platinum drugs combined with Top1 poisons, such as topotecan, showed a schedule-dependent synergistic activity, with promising results in the clinic. However, whereas the molecular mechanism of these single agents may be relatively well understood, the mode of action of these chemotherapeutic agents in combination necessitates a more complete understanding. Indeed, we recently reported that a functional homologous recombination pathway is required for cisplatin and topotecan synergy yet represses the synergistic toxicity of 1-beta-D-arabinofuranosyl cytidine in combination with topotecan (van Waardenburg, R. C., de Jong, L. A., van Delft, F., van Eijndhoven, M. A., Bohlander, M., Bjornsti, M. A., Brouwer, J., and Schellens, J. H. (2004) Mol. Cancer Ther. 3, 393-402). Here we provide direct evidence for Pt-1,3-d(GTG) poisoning of Top1 in vitro and demonstrate that persistent Pt-DNA adducts correlate with increased covalent Top1-DNA complexes in vivo. This contrasts with a lack of persistent lesions induced by the alkylating agent bis[chloroethyl]nitrosourea, which exhibits only additive activity with topotecan in a range of cell lines. In human IGROV-1 ovarian cancer cells, the synergistic activity of cisplatin with topotecan requires processive DNA polymerization, whereas overexpression of Top1 enhances yeast cell sensitivity to cisplatin. These results indicate that the cytotoxic activity of cisplatin is due, in part, to poisoning of Top1, which is exacerbated in the presence of topotecan.
- Published
- 2004
- Full Text
- View/download PDF
29. Substitution of conserved residues within the active site alters the cleavage religation equilibrium of DNA topoisomerase I.
- Author
-
Colley WC, van der Merwe M, Vance JR, Burgin AB Jr, and Bjornsti MA
- Subjects
- Asparagine, Binding Sites, Camptothecin metabolism, Camptothecin pharmacology, Conserved Sequence, DNA Topoisomerases, Type I genetics, DNA, Superhelical metabolism, Enzyme Inhibitors pharmacology, Histidine, Mutagenesis, Phenylalanine, Saccharomyces cerevisiae enzymology, Structure-Activity Relationship, Tyrosine, DNA metabolism, DNA Topoisomerases, Type I chemistry, DNA Topoisomerases, Type I metabolism
- Abstract
Eukaryotic DNA topoisomerase I (Top1p) catalyzes the relaxation of supercoiled DNA and constitutes the cellular target of camptothecin (CPT). Mutation of conserved residues in close proximity to the active site tyrosine (Tyr(727) of yeast Top1p) alters the DNA cleavage religation equilibrium, inducing drug-independent cell lethality. Previous studies indicates that yeast Top1T722Ap and Top1N726Hp cytotoxicity results from elevated levels of covalent enzyme-DNA intermediates. Here we show that Top1T722Ap acts as a CPT mimetic by exhibiting reduced rates of DNA religation, whereas increased Top1N726Hp.DNA complexes result from elevated DNA binding and cleavage. We also report that the combination of the T722A and N726H mutations in a single protein potentiates the cytotoxic action of the enzyme beyond that induced by co-expression of the single mutants. Moreover, the addition of CPT to cells expressing the double top1T722A/N726H mutant did not enhance cell lethality. Thus, independent alterations in DNA cleavage and religation contribute to the lethal phenotype. The formation of distinct cytotoxic lesions was also evidenced by the different responses induced by low levels of these self-poisoning enzymes in isogenic strains defective for the Rad9 DNA damage checkpoint, processive DNA replication, or ubiquitin-mediated proteolysis. Substitution of Asn(726) with Phe or Tyr also produces self-poisoning enzymes, implicating stacking interactions in the increased kinetics of DNA cleavage by Top1N726Hp and Top1N726Fp. In contrast, replacing the amide side chain of Asn(726) with Gln renders Top1N726Qp resistant to CPT, suggesting that the orientation of the amide within the active site is critical for effective CPT binding.
- Published
- 2004
- Full Text
- View/download PDF
30. Lost in translation: dysregulation of cap-dependent translation and cancer.
- Author
-
Bjornsti MA and Houghton PJ
- Subjects
- Animals, Cell Transformation, Neoplastic, Gene Expression Regulation, Neoplastic, Genes, Tumor Suppressor, Humans, Models, Biological, Mutation, Nuclear Proteins metabolism, Phosphatidylinositol 3-Kinases metabolism, Phosphorylation, Phosphotransferases (Alcohol Group Acceptor) metabolism, Saccharomyces cerevisiae metabolism, Saccharomyces cerevisiae Proteins metabolism, Signal Transduction, Apoptosis, Neoplasms pathology, Protein Biosynthesis
- Abstract
Activation of the phosphatidylinositol 3' kinase-Akt pathway has long been associated with malignant transformation and antiapoptotic signaling. Mutations downstream of Akt that activate the TOR kinase are found in tumor-prone syndromes, while overexpression of translation initiation complex components, such as eIF4E, occurs frequently in human cancer. However, direct roles for TOR signaling or eIF4E overexpression, in the genesis of cancer, have been lacking. Recent papers, including one by in this issue of Cancer Cell, clearly establish that dysregulation of cap-dependent translation confers malignant characteristics and induces cancer by suppressing apoptosis, underscoring the potential of therapeutics that selectively target the Akt-TOR-eIF4E pathway.
- Published
- 2004
- Full Text
- View/download PDF
31. The deubiquitinating enzyme Doa4p protects cells from DNA topoisomerase I poisons.
- Author
-
Fiorani P, Reid RJ, Schepis A, Jacquiau HR, Guo H, Thimmaiah P, Benedetti P, and Bjornsti MA
- Subjects
- Carrier Proteins genetics, Cell Cycle physiology, Cytoskeletal Proteins, Endopeptidases genetics, Endosomal Sorting Complexes Required for Transport, Genotype, Hydroxyurea pharmacology, Mutagenesis, Saccharomyces cerevisiae drug effects, Saccharomyces cerevisiae enzymology, Saccharomyces cerevisiae genetics, Saccharomyces cerevisiae Proteins genetics, Ubiquitin Thiolesterase, Endopeptidases metabolism, Enzyme Inhibitors pharmacology, Saccharomyces cerevisiae Proteins metabolism, Topoisomerase I Inhibitors
- Abstract
DNA topoisomerase I (Top1p) catalyzes changes in DNA topology via the formation of an enzyme-DNA covalent complex that is reversibly stabilized by the antitumor drug, camptothecin (CPT). During S-phase, collisions with replication forks convert these complexes into cytotoxic DNA lesions that trigger cell cycle arrest and cell death. To investigate cellular responses to CPT-induced DNA damage, a yeast genetic screen identified conditional tah mutants with enhanced sensitivity to self-poisoning DNA topoisomerase I mutant (Top1T722Ap), which mimics the action of CPT. Mutant alleles of three genes, DOA4, SLA1 and SLA2, were recovered. A nonsense mutation in DOA4 eliminated the catalytic residues of the Doa4p deubiquitinating enzyme, yet retained the rhodanase domain. At 36 degrees C, this doa4-10 mutant exhibited increased sensitivity to CPT, osmotic stress, and hydroxyurea, and a reversible petite phenotype. However, the accumulation of pre-vacuolar class E vesicles that was observed in doa4Delta cells was not detected in the doa4-10 mutant. Mutations in SLA1 or SLA2, which alter actin cytoskeleton architecture, induced a conditional synthetic lethal phenotype in combination with doa4-10 in the absence of DNA damage. Here actin cytoskeleton defects coincided with the enhanced fragility of large-budded cells. In contrast, the enhanced sensitivity of doa4-10 mutant cells to Top1T722Ap was unrelated to alterations in endocytosis and was selectively suppressed by increased dosage of the ribonucleotide reductase inhibitor Sml1p. Additional studies suggest a role for Doa4p in the Rad9p checkpoint response to Top1p poisons. These findings indicate a functional link between ubiquitin-mediated proteolysis and cellular resistance to CPT-induced DNA damage.
- Published
- 2004
- Full Text
- View/download PDF
32. The TOR pathway: a target for cancer therapy.
- Author
-
Bjornsti MA and Houghton PJ
- Subjects
- Antibiotics, Antineoplastic pharmacology, Humans, Phosphatidylinositol 3-Kinases physiology, Protein Serine-Threonine Kinases physiology, Sirolimus pharmacology, TOR Serine-Threonine Kinases, Cell Transformation, Neoplastic drug effects, Protein Kinases physiology, Signal Transduction physiology, Tacrolimus Binding Proteins physiology
- Published
- 2004
- Full Text
- View/download PDF
33. Homologous recombination is a highly conserved determinant of the synergistic cytotoxicity between cisplatin and DNA topoisomerase I poisons.
- Author
-
van Waardenburg RC, de Jong LA, van Delft F, van Eijndhoven MA, Bohlander M, Bjornsti MA, Brouwer J, and Schellens JH
- Subjects
- Animals, CHO Cells, Cell Survival drug effects, Cell Survival radiation effects, Cricetinae, Cricetulus, Cytarabine pharmacology, DNA Damage drug effects, DNA Repair, DNA Topoisomerases, Type I metabolism, DNA-Binding Proteins genetics, DNA-Binding Proteins metabolism, Drug Synergism, Rad52 DNA Repair and Recombination Protein, Saccharomyces cerevisiae drug effects, Saccharomyces cerevisiae genetics, Saccharomyces cerevisiae metabolism, Saccharomyces cerevisiae radiation effects, Saccharomyces cerevisiae Proteins, Schizosaccharomyces drug effects, Schizosaccharomyces genetics, Schizosaccharomyces metabolism, Schizosaccharomyces radiation effects, X-Rays, Camptothecin toxicity, Cisplatin toxicity, Enzyme Inhibitors toxicity, Recombination, Genetic drug effects, Topoisomerase I Inhibitors
- Abstract
Phase I and II clinical trails are currently investigating the antitumor activity of cisplatin and camptothecins (CPTs; DNA topoisomerase I poisons), based on the dramatic synergistic cytotoxicity of these agents in some preclinical models. However, the mechanistic basis for this synergism is poorly understood. By exploiting the evolutionary conservation of DNA repair pathways from genetically tractable organisms such as budding and fission yeasts to mammalian cells, we demonstrate that the synergism of CPT and cisplatin requires homologous recombination. In yeast and mammalian cell lines defective for RAD52 and XRCC2/3, respectively, the combination of these agents proved antagonistic, while greater than additive activity was evident in isogenic wild-type cells. Homologous recombination appears to mediate a similar interaction of X-rays and CPT, but antagonizes the synergism of cytarabine (Ara-C) with CPT. These findings suggest that homologous recombination comprises an evolutionarily conserved determinant of cellular sensitivity when CPTs are used in combination with other therapeutics.
- Published
- 2004
34. Locking the DNA topoisomerase I protein clamp inhibits DNA rotation and induces cell lethality.
- Author
-
Woo MH, Losasso C, Guo H, Pattarello L, Benedetti P, and Bjornsti MA
- Subjects
- Camptothecin pharmacology, Catalytic Domain, DNA Topoisomerases, Type I genetics, Disulfides chemistry, Macromolecular Substances, Models, Molecular, Mutation, Nucleic Acid Conformation, Protein Conformation, Rotation, Saccharomyces cerevisiae cytology, Saccharomyces cerevisiae drug effects, Saccharomyces cerevisiae genetics, Saccharomyces cerevisiae metabolism, DNA Topoisomerases, Type I chemistry, DNA Topoisomerases, Type I metabolism, DNA, Fungal chemistry, DNA, Fungal metabolism
- Abstract
Eukaryotic DNA topoisomerase I (Top1) is a monomeric protein clamp that functions in DNA replication, transcription, and recombination. Opposable "lip" domains form a salt bridge to complete Top1 protein clamping of duplex DNA. Changes in DNA topology are catalyzed by the formation of a transient phosphotyrosyl linkage between the active-site Tyr-723 and a single DNA strand. Substantial protein domain movements are required for DNA binding, whereas the tight packing of DNA within the covalent Top1-DNA complex necessitates some DNA distortion to allow rotation. To investigate the effects of Top1-clamp closure on enzyme catalysis, molecular modeling was used to design a disulfide bond between residues Gly-365 and Ser-534, to crosslink protein loops more proximal to the active-site tyrosine than the protein loops held by the Lys-369-Glu-497 salt bridge. In reducing environments, Top1-Clamp was catalytically active. However, contrary to crosslinking the salt-bridge loops [Carey, J. F., Schultz, S. J., Sission, L., Fazzio, T. G. & Champoux, J. J. (2003) Proc. Natl. Acad. Sci. USA 100, 5640-5645], crosslinking the active-site proximal loops inhibited DNA rotation. Apparently, subtle alterations in Top1 clamp flexibility impact enzyme catalysis in vitro. Yet, the catalytically active Top1-Clamp was cytotoxic, even in the reducing environment of yeast cells. Remarkably, a shift in redox potential in glr1Delta cells converted the catalytically inactive Top1Y723F mutant clamp into a cellular toxin, which failed to induce an S-phase terminal phenotype. This cytotoxic mechanism is distinct from that of camptothecin chemotherapeutics, which stabilize covalent Top1-DNA complexes, and it suggests that the development of novel therapeutics that promote Top1-clamp closure is possible.
- Published
- 2003
- Full Text
- View/download PDF
35. Rapamycins: mechanism of action and cellular resistance.
- Author
-
Huang S, Bjornsti MA, and Houghton PJ
- Subjects
- Animals, Drug Resistance, Neoplasm, Humans, Neoplasms drug therapy, Neoplasms metabolism, Protein Kinases metabolism, TOR Serine-Threonine Kinases, Anti-Bacterial Agents pharmacology, Antibiotics, Antineoplastic pharmacology, Immunosuppressive Agents pharmacology, Sirolimus pharmacology
- Abstract
Rapamycins are macrocyclic lactones that possess immunosuppressive, antifungal and antitumor properties. The parent compound, rapamycin, is approved as an immunosup-pressive agent for preventing rejection in patients receiving organ transplantation. Two analogues, CCI-779 and RAD001 are currently being investigated as anticancer agents. Rapamycins first bind a cyclophilin FKBP12, and this complex binds and inhibits the function of mTOR (mammalian target of rapamycin) a serine/threonine (Ser/Thr) kinase with homology to phosphatidylinositol 3' kinase. Currently, as mTOR is the only identified target, this places rapamycins in a unique position of being the most selective kinase inhibitor known. Consequently these agents have been powerful tools in elucidating the role of mTOR in cellular growth, proliferation, survival and tumorigenesis. Increasing evidence suggests that mTOR acts as a central controller sensing cellular environment (nutritional status or mitogenic stimulation) and regulating translation initiation through the eukaryotic initiation factor 4E, and ribosomal p70 S6 kinase pathways. Here we review the conserved TOR signaling pathways, conceptual basis for tumor selectivity, and the mechanisms of resistance to this class of antitumor agent.
- Published
- 2003
- Full Text
- View/download PDF
36. A novel active DNA topoisomerase I in Leishmania donovani.
- Author
-
Villa H, Otero Marcos AR, Reguera RM, Balaña-Fouce R, García-Estrada C, Pérez-Pertejo Y, Tekwani BL, Myler PJ, Stuart KD, Bjornsti MA, and Ordóñez D
- Subjects
- Amino Acid Motifs, Amino Acid Sequence, Animals, Base Sequence, Cloning, Molecular, DNA Primers, DNA Topoisomerases, Type I chemistry, DNA Topoisomerases, Type I genetics, Genes, Protozoan, Humans, Molecular Sequence Data, Open Reading Frames, Sequence Homology, Amino Acid, DNA Topoisomerases, Type I metabolism, Leishmania donovani enzymology
- Abstract
A common feature shared by type I DNA topoisomerases is the presence of a "serine, lysine, X, X, tyrosine" motif as conventional enzyme active site. Preliminary data have shown that Leishmania donovani DNA topoisomerase I gene (LdTOP1A) lacked this conserved motif, giving rise to different theories about the reconstitution of an active DNA topoisomerase I in this parasite. We, herein, describe the molecular cloning of a new DNA topoisomerase I gene from L. donovani (LdTOP1B) containing the highly conserved serine, lysine, X, X, tyrosine motif. DNA topoisomerase I activity was detected only when both genes (LdTOP1A and LdTOP1B) were co-expressed in a yeast expression system, suggesting the existence of a dimeric DNA topoisomerase I in Leishmania parasites.
- Published
- 2003
- Full Text
- View/download PDF
37. Cancer therapeutics in yeast.
- Author
-
Bjornsti MA
- Subjects
- Humans, Models, Biological, Saccharomyces cerevisiae enzymology, Saccharomyces cerevisiae genetics, Antineoplastic Agents pharmacology, Neoplasms drug therapy, Saccharomyces cerevisiae drug effects
- Abstract
The budding yeast Saccharomyces cerevisiae is a genetically tractable model system with which to establish the cellular target of a given agent and investigate mechanisms of drug action.
- Published
- 2002
- Full Text
- View/download PDF
38. Active site mutations in DNA topoisomerase I distinguish the cytotoxic activities of camptothecin and the indolocarbazole, rebeccamycin.
- Author
-
Woo MH, Vance JR, Marcos AR, Bailly C, and Bjornsti MA
- Subjects
- Base Sequence, Binding Sites, DNA Primers, DNA Topoisomerases, Type I genetics, Humans, Mutagenesis, Recombinant Proteins antagonists & inhibitors, Recombinant Proteins genetics, Saccharomyces cerevisiae genetics, Aminoglycosides, Anti-Bacterial Agents pharmacology, Camptothecin pharmacology, Carbazoles, Enzyme Inhibitors pharmacology, Indoles, Mutation, Topoisomerase I Inhibitors
- Abstract
DNA topoisomerase I (Top1p) catalyzes topological changes in DNA and is the cellular target of the antitumor agent camptothecin (CPT). Non-CPT drugs that target Top1p, such as indolocarbazoles, are under clinical development. However, whether the cytotoxicity of indolocarbazoles derives from Top1p poisoning remains unclear. To further investigate indolocarbazole mechanism, rebeccamycin R-3 activity was examined in vitro and in yeast. Using a series of Top1p mutants, where substitution of residues around the active site tyrosine has well-defined effects on enzyme catalysis, we show that catalytically active, CPT-resistant enzymes remain sensitive to R-3. This indolocarbazole did not inhibit yeast Top1p activity, yet was effective in stabilizing Top1p-DNA complexes. Similar results were obtained with human Top1p, when Ser or His were substituted for Asn-722. The mutations altered enzyme function and sensitivity to CPT, yet R-3 poisoning of Top1p was unaffected. Moreover, top1delta, rad52delta yeast cells expressing human Top1p, but not catalytically inactive Top1Y723Fp, were sensitive to R-3. These data support hTop1p as the cellular target of R-3 and indicate that distinct drug-enzyme interactions at the active site are required for efficient poisoning by R-3 or CPT. Furthermore, resistance to one poison may potentiate cell sensitivity to structurally distinct compounds that also target Top1p.
- Published
- 2002
- Full Text
- View/download PDF
39. Drug-induced stabilization of covalent DNA topoisomerase I-DNA intermediates. DNA cleavage assays.
- Author
-
Fiorani P, Hann CL, Benedetti P, and Bjornsti MA
- Subjects
- Camptothecin analogs & derivatives, Cells, Cultured, DNA, Bacterial genetics, DNA, Bacterial metabolism, Electrophoresis, Polyacrylamide Gel, Enzyme Stability drug effects, Humans, Plasmids genetics, Sequence Analysis, DNA, Topoisomerase I Inhibitors, Antineoplastic Agents pharmacology, Camptothecin pharmacology, DNA Topoisomerases, Type I metabolism, DNA-Binding Proteins metabolism, Plasmids metabolism
- Published
- 2001
- Full Text
- View/download PDF
40. Studying DNA topoisomerase I-targeted drugs in the yeast. Saccharomyces cerevisiae.
- Author
-
Woo MH, Vance JR, and Bjornsti MA
- Subjects
- Camptothecin pharmacology, Cell Division drug effects, Cell Membrane Permeability, Cell Survival drug effects, DNA Topoisomerases, Type I genetics, DNA Topoisomerases, Type I metabolism, Gene Deletion, Genes, Fungal, Genotype, Humans, Plasmids genetics, Recombinant Proteins antagonists & inhibitors, Recombinant Proteins metabolism, Saccharomyces cerevisiae cytology, Saccharomyces cerevisiae enzymology, Saccharomyces cerevisiae genetics, Transformation, Genetic, Antineoplastic Agents pharmacology, Saccharomyces cerevisiae drug effects, Topoisomerase I Inhibitors
- Published
- 2001
- Full Text
- View/download PDF
41. Substitutions of Asn-726 in the active site of yeast DNA topoisomerase I define novel mechanisms of stabilizing the covalent enzyme-DNA intermediate.
- Author
-
Fertala J, Vance JR, Pourquier P, Pommier Y, and Bjornsti MA
- Subjects
- Amino Acid Substitution, Base Sequence, Binding Sites, Camptothecin pharmacology, Enzyme Stability, Mutagenesis, Site-Directed, Nucleic Acid Conformation, Plasmids chemistry, Saccharomyces cerevisiae drug effects, Saccharomyces cerevisiae growth & development, Substrate Specificity, Asparagine, DNA Topoisomerases, Type I chemistry, DNA Topoisomerases, Type I metabolism, Plasmids metabolism, Saccharomyces cerevisiae enzymology
- Abstract
Eukaryotic DNA topoisomerase I (Top1p) catalyzes changes in DNA topology and is the cellular target of camptothecin. Recent reports of enzyme structure highlight the importance of conserved amino acids N-terminal to the active site tyrosine and the involvement of Asn-726 in mediating Top1p sensitivity to camptothecin. To investigate the contribution of this residue to enzyme catalysis, we evaluated the effect of substituting His, Asp, or Ser for Asn-726 on yeast Top1p. Top1N726S and Top1N726D mutant proteins were resistant to camptothecin, although the Ser mutant was distinguished by a lack of detectable changes in activity. Thus, a basic residue immediately N-terminal to the active site tyrosine is required for camptothecin cytotoxicity. However, replacing Asn-726 with Asp or His interfered with distinct aspects of the catalytic cycle, resulting in cell lethality. In contrast to camptothecin, which inhibits enzyme-catalyzed religation of DNA, the His substituent enhanced the rate of DNA scission, whereas the Asp mutation diminished the enzyme binding of DNA. Yet, these effects on enzyme catalysis were not mutually exclusive as the His mutant was hypersensitive to camptothecin. These results suggest distinct mechanisms of poisoning DNA topoisomerase I may be explored in the development of antitumor agents capable of targeting different aspects of the Top1p catalytic cycle.
- Published
- 2000
- Full Text
- View/download PDF
42. Mechanisms of DNA topoisomerase I-induced cell killing in the yeast Saccharomyces cerevisiae.
- Author
-
Fiorani P and Bjornsti MA
- Subjects
- Animals, Antineoplastic Agents, Phytogenic toxicity, Camptothecin toxicity, DNA Topoisomerases, Type I genetics, DNA Topoisomerases, Type I metabolism, Enzyme Inhibitors toxicity, Saccharomyces cerevisiae cytology, Saccharomyces cerevisiae drug effects, Saccharomyces cerevisiae genetics, Topoisomerase I Inhibitors, DNA Topoisomerases, Type I physiology, Saccharomyces cerevisiae enzymology
- Abstract
DNA topoisomerase I (Top1) catalyzes the relaxation of supercoiled DNA by a mechanism of transient DNA strand cleavage characterized by the formation of a phosphotyrosyl bond between the DNA end and active site tyrosine. Camptothecin reversibly stabilizes the covalent enzyme-DNA intermediate by inhibiting DNA religation. During S-phase, collisions with advancing replication forks convert these complexes into potentially lethal lesions. To define the DNA damage induced by alterations in Top1p catalysis and the cellular processes that mediate the repair of such lesions, the yeast Saccharomyces cerevisiae was used. Substitution of conserved residues N-terminal to the active site tyrosine (Tyr-727) produced alterations in the camptothecin sensitivity or catalytic cycle of DNA Top1. For example, substituting Ala for Thr-722 in Top1T722A increased the stability of the covalent enzyme DNA intermediate. As with camptothecin, Top1T722A-induced cytotoxicity was ascribed to a reduction in DNA religation. By contrast, enhanced covalent complex formation by Top1N726H resulted from a relative increase in the rate of DNA cleavage. Conditional yeast mutants were also selected that exhibit temperature-sensitive growth only in the presence of the self-poisoning Top1T722A enzyme. Subsequent analyses of these tah mutants identified 9 genes whose function suppresses the cytotoxic action of camptothecin and Top1T722A. These include genes encoding essential DNA replication proteins (CDC45 and DPB11) and proteins involved in SUMO- or ubiquitination (UBC9 and DOA4).
- Published
- 2000
- Full Text
- View/download PDF
43. Domain interactions affecting human DNA topoisomerase I catalysis and camptothecin sensitivity.
- Author
-
Fiorani P, Amatruda JF, Silvestri A, Butler RH, Bjornsti MA, and Benedetti P
- Subjects
- Alanine genetics, Amino Acid Substitution, Catalysis, DNA drug effects, DNA metabolism, DNA Topoisomerases, Type I chemistry, DNA Topoisomerases, Type I genetics, Drug Resistance, Enzyme Stability, Glycine genetics, Humans, Models, Molecular, Mutation, Protein Conformation, Protein Structure, Tertiary, Saccharomyces cerevisiae genetics, Threonine genetics, Transfection, Camptothecin pharmacology, DNA Topoisomerases, Type I metabolism, Enzyme Inhibitors pharmacology, Topoisomerase I Inhibitors
- Abstract
DNA topoisomerase I (Top1p) relaxes supercoiled DNA by the formation of a covalent intermediate in which the active site tyrosine is transiently bound to the severed DNA strand. The antineoplastic agent camptothecin (Cpt) specifically targets Top1p and several mutations have been isolated that render the enzyme Cpt resistant. The mutated residues, although located in different regions of the enzyme, may constitute part of the Cpt binding site. To begin identifying the structural features of DNA Top1p important for Cpt-induced cytotoxicity, we developed a novel yeast genetic screen to isolate catalytically active, yet Cpt-resistant enzymes from a pool of human top1 mutants. Among the mutations isolated were substitutions of Ser or Val for Gly363, which like the Gly363 to Cys mutation previously reported by us, suppressed the Cpt sensitivity of Top1p. In contrast, each amino-acid substitution differed in its ability to suppress the lethal phenotype and catalytic activity of a human top1 mutant top1T718A that resembles Cpt by stabilizing the covalent intermediate. Biochemical analyses and molecular modeling support a model where interactions between two conserved domains, a central "lip" region containing residue Gly363 and the residues around the active site tyrosine (Tyr723), directly affect the formation of the Cpt-binding site and enzyme catalysis.
- Published
- 1999
- Full Text
- View/download PDF
44. CDC45 and DPB11 are required for processive DNA replication and resistance to DNA topoisomerase I-mediated DNA damage.
- Author
-
Reid RJ, Fiorani P, Sugawara M, and Bjornsti MA
- Subjects
- Camptothecin pharmacology, Mutation, Carrier Proteins physiology, Cell Cycle Proteins physiology, DNA Damage, DNA Replication, DNA Topoisomerases, Type I physiology, DNA-Binding Proteins, Nuclear Proteins physiology, Saccharomyces cerevisiae Proteins
- Abstract
The antitumor agent camptothecin targets DNA topoisomerase I by reversibly stabilizing a covalent enzyme-DNA intermediate. The subsequent collision of DNA replication forks with these drug-enzyme-DNA complexes produces the cytotoxic DNA lesions that signal cell cycle arrest and ultimately lead to cell death. Despite intense investigation, the character of the lesions produced and the repair processes that resolve the damage remain poorly defined. A yeast genetic screen was implemented to isolate conditional mutants with enhanced sensitivity to DNA topoisomerase I-mediated DNA damage. Cells exhibiting temperature-sensitive growth in the presence of the DNA topoisomerase I mutant, Top1T722Ap, were selected. Substitution of Ala for Thr722 increases the stability of the covalent Top1T722Ap-DNA intermediate, mimicking the cytotoxic action of camptothecin. Two mutants isolated, cdc45-10 and dpb11-10, exhibited specific defects in DNA replication and a synthetic lethal phenotype in the absence of DNA damaging agents. The accumulation of Okazaki fragments under nonpermissive conditions suggests a common function in promoting processive DNA replication through polymerase switching. These results provide a mechanistic basis for understanding the cellular processes involved in the resolution of DNA damage induced by camptothecin and DNA topoisomerase I.
- Published
- 1999
- Full Text
- View/download PDF
45. Induction of reversible complexes between eukaryotic DNA topoisomerase I and DNA-containing oxidative base damages. 7, 8-dihydro-8-oxoguanine and 5-hydroxycytosine.
- Author
-
Pourquier P, Ueng LM, Fertala J, Wang D, Park HJ, Essigmann JM, Bjornsti MA, and Pommier Y
- Subjects
- Camptothecin pharmacology, Cytosine pharmacology, DNA metabolism, DNA Topoisomerases, Type I genetics, DNA-Binding Proteins genetics, DNA-Binding Proteins metabolism, Guanosine pharmacology, Humans, Kinetics, Molecular Structure, Mutation genetics, Oligonucleotides metabolism, Oxidative Stress, Cytosine analogs & derivatives, DNA Damage genetics, DNA Topoisomerases, Type I metabolism, Guanosine analogs & derivatives
- Abstract
We recently showed that abasic sites, uracil mismatches, nicks, and gaps can trap DNA topoisomerase I (top1) when these lesions are introduced in the vicinity of a top1 cleavage site (Pourquier, P., Ueng, L.-M., Kohlhagen, G., Mazumder, A., Gupta, M., Kohn, K. W., and Pommier, Y. (1997) J. Biol. Chem. 272, 7792-7796; Pourquier, P., Pilon, A. A., Kohlhagen, G., Mazumder, A., Sharma, A., and Pommier, Y. (1997) J. Biol. Chem. 26441-26447). In this study, we investigated the effects on top1 of an abundant base damage generated by various oxidative stresses: 7,8-dihydro-8-oxoguanine (8-oxoG). Using purified eukaryotic top1 and oligonucleotides containing the 8-oxoG modification, we found a 3-7-fold increase in top1-mediated DNA cleavage when 8-oxoG was present at the +1 or +2 position relative to the cleavage site. Another oxidative lesion, 5-hydroxycytosine, also enhanced top1 cleavage by 2-fold when incorporated at the +1 position of the scissile strand. 8-oxoG at the +1 position enhanced noncovalent top1 DNA binding and had no detectable effect on DNA religation or on the incision step. top1 trapping by 8-oxoG was markedly enhanced when asparagine adjacent to the catalytic tyrosine was mutated to histidine, suggesting a direct interaction between this residue and the DNA major groove immediately downstream from the top1 cleavage site. Altogether, these results demonstrate that oxidative base lesions can increase top1 binding to DNA and induce top1 cleavage complexes.
- Published
- 1999
- Full Text
- View/download PDF
46. Cascades of mammalian caspase activation in the yeast Saccharomyces cerevisiae.
- Author
-
Kang JJ, Schaber MD, Srinivasula SM, Alnemri ES, Litwack G, Hall DJ, and Bjornsti MA
- Subjects
- Caspase 10, Caspase 3, Caspase 6, Caspase 8, Caspase 9, Caspases genetics, Catalysis, Coumarins metabolism, Enzyme Activation, Fluorescent Dyes metabolism, Galactose pharmacology, Humans, Microscopy, Fluorescence, Oligopeptides metabolism, Saccharomyces cerevisiae genetics, Schizosaccharomyces, Caspases metabolism, Enzyme Precursors metabolism, Saccharomyces cerevisiae enzymology
- Abstract
Caspases (aspartate-specific cysteine proteases) play a critical role in the execution of the mammalian apoptotic program. To address the regulation of human caspase activation, we used the yeast Saccharomyces cerevisiae, which is devoid of endogenous caspases. The apical procaspases, -8beta and -10, were efficiently processed and activated in yeast. Although protease activity, per se, was insufficient to drive cell death, caspase-10 activity had little effect on cell viability, whereas expression of caspase-8beta was cytotoxic. This lethal phenotype was abrogated by co-expression of the pan-caspase inhibitor, baculovirus p35, and by mutation of the active site cysteine of procaspase-8beta. In contrast, autoactivation of the executioner caspase-3 and -6 zymogens was not detected. Procaspase-3 activation required co-expression of procaspase-8 or -10. Surprisingly, activation of procaspase-6 required proteolytic activities other than caspase-8, -10, or -3. Caspase-8beta or -10 activity was insufficient to catalyze the maturation of procaspase-6. Moreover, a constitutively active caspase-3, although cytotoxic in its own right, was unable to induce the processing of wild-type procaspase-6 and vice versa. These results distinguish sequential modes of activation for different caspases in vivo and establish a yeast model system to examine the regulation of caspase cascades. Moreover, the distinct terminal phenotypes induced by various caspases attest to differences in the cellular targets of these apoptotic proteases, which may be defined using this system.
- Published
- 1999
- Full Text
- View/download PDF
47. Overexpression and purification of DNA topoisomerase I from yeast.
- Author
-
Bjornsti MA and Fertala J
- Subjects
- DNA Topoisomerases, Type I genetics, Plasmids, Saccharomyces cerevisiae genetics, Transformation, Genetic, DNA Topoisomerases, Type I isolation & purification, DNA Topoisomerases, Type I metabolism, Saccharomyces cerevisiae enzymology
- Published
- 1999
- Full Text
- View/download PDF
48. Resolution of DNA molecules by one-dimensional agarose-gel electrophoresis.
- Author
-
Bjornsti MA and Megonigal MD
- Subjects
- Molecular Weight, Plasmids, DNA analysis, Electrophoresis, Agar Gel methods
- Published
- 1999
- Full Text
- View/download PDF
49. Introduction to DNA topoisomerases.
- Author
-
Bjornsti MA and Osheroff N
- Subjects
- Animals, Humans, DNA Topoisomerases
- Published
- 1999
- Full Text
- View/download PDF
50. Intragenic suppressors of mutant DNA topoisomerase I-induced lethality diminish enzyme binding of DNA.
- Author
-
Hann CL, Carlberg AL, and Bjornsti MA
- Subjects
- Amino Acid Sequence, Camptothecin pharmacology, Catalytic Domain genetics, Conserved Sequence, DNA Topoisomerases, Type I toxicity, Genes, Lethal, Humans, Mutation, Protein Binding genetics, Topoisomerase I Inhibitors, DNA Topoisomerases, Type I genetics, DNA Topoisomerases, Type I metabolism, Suppression, Genetic
- Abstract
Eukaryotic DNA topoisomerase I (Top1p) catalyzes changes in DNA topology and is the cellular target of the antitumor drug camptothecin (Cpt). Mutation of several conserved residues in yeast top1 mutants is sufficient to induce cell lethality in the absence of camptothecin. Despite tremendous differences in catalytic activity, the mutant proteins Top1T722Ap and Top1R517Gp cause cell death via a mechanism similar to that of Cpt, i.e. stabilization of the covalent enzyme-DNA intermediate. To establish the interdomainal interactions required for the catalytic activity of Top1p and how alterations in enzyme structure contribute to the cytotoxic activity of Cpt or specific DNA topoisomerase I mutants, we initiated a genetic screen for intragenic suppressors of the top1T722A-lethal phenotype. Nine single amino acid substitutions were defined that map to the conserved central and C-terminal domains of Top1p as well as the nonconserved linker domain of the protein. All reduced the catalytic activity of the enzyme over 100-fold. However, detailed biochemical analyses of three suppressors, top1C273Y,T722A, top1G295V,T722A, and top1G369D,T722A, revealed this was accomplished via a mechanism of reduced affinity for the DNA substrate. The mechanistic implications of these results are discussed in the context of the known structures of yeast and human DNA topoisomerase I.
- Published
- 1998
- Full Text
- View/download PDF
Catalog
Discovery Service for Jio Institute Digital Library
For full access to our library's resources, please sign in.