Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 15 de 15
Filter
1.
Mod Pathol ; 26(10): 1320-8, 2013 Oct.
Article in English | MEDLINE | ID: mdl-23599145

ABSTRACT

HRAS is mutated in ∼15% of Spitz nevi, and GNAQ or GNA11 is mutated in blue nevi (46-83% and ∼7% respectively). Epithelioid blue nevi and deep penetrating nevi show features of both blue nevi (intradermal location, pigmentation) and Spitz nevi (epithelioid morphology). Epithelioid blue nevi and deep penetrating nevi can also show overlapping features with melanoma, posing a diagnostic challenge. Although epithelioid blue nevi are considered blue nevic variants, no GNAQ or GNA11 mutations have been reported. Classification of deep penetrating nevi as blue nevic variants has also been proposed, however, no GNAQ or GNA11 mutations have been reported and none have been tested for HRAS mutations. To better characterize these tumors, we performed mutational analysis for GNAQ, GNA11, and HRAS, with blue nevi and Spitz nevi as controls. Within deep penetrating nevi, none demonstrated GNAQ or GNA11 mutations (0/38). However, 6% revealed HRAS mutation (2/32). Twenty percent of epithelioid blue nevi contained a GNAQ mutation (2/10), while none displayed GNA11 or HRAS mutation. Eighty-seven percent of blue nevi contained a GNAQ mutation (26/30), 4% a GNA11 mutation (1/28), and none an HRAS mutation. Within Spitz nevi, none demonstrated GNAQ or GNA11 mutations (0/30). Seventeen percent contained an HRAS mutation (5/30). All GNAQ and GNA11 mutations were p.Q209L (c.626A>T) point mutations, except 2 GNAQ mutations, which contained novel c.625_626CA>TT double mutations. Four HRAS mutations were in exon 2, and three in exon 3. This is the first study to identify HRAS mutations in deep penetrating nevi. The presence of HRAS mutations and absence of GNAQ or GNA11 mutations in deep penetrating nevi suggests classification of these unusual nevi within the Spitz nevus category of melanocytic tumors, rather than the blue nevus category.


Subject(s)
Nevus/genetics , Point Mutation , Proto-Oncogene Proteins p21(ras)/genetics , Skin Neoplasms/genetics , Adolescent , Adult , Aged , Aged, 80 and over , Child , Child, Preschool , DNA Mutational Analysis , Female , GTP-Binding Protein alpha Subunits/genetics , GTP-Binding Protein alpha Subunits/metabolism , GTP-Binding Protein alpha Subunits, Gq-G11 , Humans , Male , Middle Aged , Nevus/metabolism , Nevus/pathology , Proto-Oncogene Proteins p21(ras)/metabolism , Skin Neoplasms/metabolism , Skin Neoplasms/pathology
2.
Oncotarget ; 7(2): 1707-16, 2016 Jan 12.
Article in English | MEDLINE | ID: mdl-26625196

ABSTRACT

Malignant phyllodes tumor is a rare breast malignancy with sarcomatous overgrowth and with limited effective treatment options for recurrent and metastatic cases. Recent clinical trials indicated a potential for anti-angiogenic, anti-EGFR and immunotherapeutic approaches for patients with sarcomas, which led us to investigate these and other targetable pathways in malignant phyllodes tumor of the breast. Thirty-six malignant phyllodes tumors (including 8 metastatic tumors with two cases having matched primary and metastatic tumors) were profiled using gene sequencing, gene copy number analysis, whole genome expression, and protein expression. Whole genome expression analysis demonstrated consistent over-expression of genes involved in angiogenesis including VEGFA, Angiopoietin-2, VCAM1, PDGFRA, and PTTG1. EGFR protein overexpression was observed in 26/27 (96%) of cases with amplification of the EGFR gene in 8/24 (33%) cases. Two EGFR mutations were identified including EGFRvIII and a presumed pathogenic V774M mutation, respectively. The most common pathogenic mutations included TP53 (50%) and PIK3CA (15%). Cases with matched primary and metastatic tumors harbored identical mutations in both sites (PIK3CA/KRAS and RB1 gene mutations, respectively). Tumor expression of PD-L1 immunoregulatory protein was observed in 3/22 (14%) of cases. Overexpression of molecular biomarkers of increased angiogenesis, EGFR and immune checkpoints provides novel targeted therapy options in malignant phyllodes tumors of the breast.


Subject(s)
Biomarkers, Tumor/genetics , Breast Neoplasms/genetics , Gene Expression Profiling/methods , Gene Expression Regulation, Neoplastic , Phyllodes Tumor/genetics , Adolescent , Adult , Aged , Biomarkers, Tumor/metabolism , Breast Neoplasms/blood supply , Breast Neoplasms/metabolism , ErbB Receptors/genetics , Female , Humans , Immunohistochemistry , Middle Aged , Mutation , Neovascularization, Pathologic/genetics , Neovascularization, Pathologic/metabolism , Oligonucleotide Array Sequence Analysis , Phyllodes Tumor/blood supply , Phyllodes Tumor/metabolism , Sequence Analysis, DNA/methods , Young Adult
3.
Chem Biol Interact ; 153-154: 197-205, 2005 May 30.
Article in English | MEDLINE | ID: mdl-15935817

ABSTRACT

Benzene is a human carcinogen that induces hematopoietic malignancies. It is believed that benzene does not initiate leukemias directly, but rather generates DNA damage through a series of phenolic and quinone-based metabolites, especially 1,4-benzoquinone. Since the DNA damage induced by 1,4-benzoquinone is consistent with that of topoisomerase II-targeted drugs, it has been proposed that the compound initiates specific types of leukemia by acting as a topoisomerase II poison. This hypothesis, however, was not supported by initial in vitro studies. While 1,4-benzoquinone inhibited topoisomerase II catalysis, increases in enzyme-mediated DNA cleavage were not observed. Because of the potential involvement of topoisomerase II in benzene-induced leukemias, we re-examined the effects of benzene metabolites (including 1,4-benzoquinone, 1,4-hydroquinone, catechol, 1,2,4-benzenetriol, 2,2'-biphenol, and 4,4'-biphenol) on DNA cleavage mediated by human topoisomerase IIalpha. In contrast to previous reports, we found that 1,4-benzoquinone was a strong topoisomerase II poison and was more potent in vitro than the anticancer drug etoposide. Other metabolites displayed considerably less activity. DNA cleavage enhancement by 1,4-benzoquinone was unseen in previous studies due to the presence of reducing agents and the incubation of 1,4-benzoquinone with the enzyme prior to the addition of DNA. Unlike anticancer drugs such as etoposide that interact with topoisomerase IIalpha in a noncovalent manner, the actions of 1,4-benzoquinone appear to involve a covalent attachment to the enzyme. Finally, 1,4-benzoquinone stimulated DNA cleavage by topoisomerase IIalpha in cultured human cells. These findings are consistent with the hypothesis that topoisomerase IIalpha plays a role in the initiation of some benzene-induced leukemias.


Subject(s)
Benzoquinones/toxicity , DNA Damage , DNA-Binding Proteins/antagonists & inhibitors , Topoisomerase II Inhibitors , Antigens, Neoplasm/metabolism , Benzene/metabolism , Benzene/toxicity , Chromosome Breakage , DNA Topoisomerases, Type II/metabolism , DNA-Binding Proteins/metabolism , Humans , Tumor Cells, Cultured
4.
Oncotarget ; 6(23): 19819-25, 2015 Aug 14.
Article in English | MEDLINE | ID: mdl-26110571

ABSTRACT

The histiocytoses are rare tumors characterized by the primary accumulation and tissue infiltration of histiocytes and dendritic cells. Identification of the activating BRAFV600E mutation in Erdheim-Chester disease (ECD) and Langerhans cell histiocytosis (LCH) cases provided the basis for the treatment with BRAF and/or MEK inhibitors, but additional treatment options are needed. Twenty-four cases of neoplastic histiocytic diseases [11 extrapulmonary LCH, 4 ECD, 4 extranodal Rosai-Dorfman disease (RDD), 3 follicular dendritic cell sarcoma (FDCS), 1 histiocytic sarcoma (HS) and 1 blastic plasmacytoid dendritic cell neoplasm (BPDCN)] were analyzed using immunohistochemical and mutational analysis in search of biomarkers for targeted therapy. BRAF V600E mutations were detected in 4/11 LCH and 4/4 ECD cases. A pathogenic PTEN gene mutation and loss of PTEN protein expression were identified in the case of HS. Increased expression of PD-L1 (≥2+/≥5%) was seen in 3/4 ECD, 7/8 LCH, 3/3 FDCS and 1/1 HS, with overall 81% concordance between 2 antibodies used in the study (SP142 vs. MAB1561 clone). These results show for the first time significant expression of the PD-L1 immune checkpoint protein in these disorders, which may provide rationale for addition of immune check-point inhibitors in treatment of disseminated and/or refractory histiocytoses.


Subject(s)
B7-H1 Antigen/analysis , Dendritic Cell Sarcoma, Follicular/metabolism , Dendritic Cells/chemistry , Erdheim-Chester Disease/metabolism , Histiocytes/chemistry , Histiocytic Sarcoma/metabolism , Histiocytosis, Langerhans-Cell/metabolism , Histiocytosis, Sinus/metabolism , Mutation , Proto-Oncogene Proteins B-raf/genetics , Adolescent , Adult , Aged , Child , Child, Preschool , DNA Mutational Analysis , Dendritic Cell Sarcoma, Follicular/enzymology , Dendritic Cell Sarcoma, Follicular/genetics , Dendritic Cell Sarcoma, Follicular/pathology , Dendritic Cells/enzymology , Erdheim-Chester Disease/enzymology , Erdheim-Chester Disease/genetics , Erdheim-Chester Disease/pathology , Female , Genetic Markers , Histiocytes/enzymology , Histiocytes/pathology , Histiocytic Sarcoma/enzymology , Histiocytic Sarcoma/genetics , Histiocytic Sarcoma/pathology , Histiocytosis, Langerhans-Cell/enzymology , Histiocytosis, Langerhans-Cell/genetics , Histiocytosis, Langerhans-Cell/pathology , Histiocytosis, Sinus/enzymology , Histiocytosis, Sinus/genetics , Histiocytosis, Sinus/pathology , Humans , Immunohistochemistry , Male , Middle Aged , PTEN Phosphohydrolase/analysis , PTEN Phosphohydrolase/genetics , Young Adult
5.
Hum Pathol ; 46(9): 1350-9, 2015 Sep.
Article in English | MEDLINE | ID: mdl-26208846

ABSTRACT

Despite the marked improvement in the understanding of molecular mechanisms and classification of apocrine carcinoma, little is known about its specific molecular genetic alterations and potentially targetable biomarkers. In this study, we explored immunohistochemical and molecular genetic characteristics of 37 invasive apocrine carcinomas using immunohistochemistry (IHC), fluorescent in situ hybridization (FISH), multiplex ligation-dependent probe amplification (MLPA), and next-generation sequencing (NGS) assays. IHC revealed frequent E-cadherin expression (89%), moderate (16%) proliferation activity [Ki-67, phosphohistone H3], infrequent (~10%) expression of basal cell markers [CK5/6, CK14, p63, caveolin-1], loss of PTEN (83%), and overexpression of HER2 (32%), EGFR (41%), cyclin D1 (50%), and MUC-1 (88%). MLPA assay revealed gene copy gains of MYC, CCND1, ZNF703, CDH1, and TRAF4 in 50% or greater of the apocrine carcinomas, whereas gene copy losses frequently affected BRCA2 (75%), ADAM9 (54%), and BRCA1 (46%). HER2 gain, detected by MLPA in 38% of the cases, was in excellent concordance with HER2 results obtained by IHC/FISH (κ = 0.915, P < .001). TOP2A gain was observed in one case, while five cases (21%) exhibited TOP2A loss. Unsupervised hierarchical cluster analysis revealed two distinct clusters: HER2-positive and HER2-negative (P = .03 and .04, respectively). NGS assay revealed mutations of the TP53 (2 of 7, 29%), BRAF/KRAS (2 of 7, 29%), and PI3KCA/PTEN genes (7 of 7, 100%). We conclude that morphologically defined apocrine carcinomas exhibit complex molecular genetic alterations that are consistent with the "luminal-complex" phenotype. Some of the identified molecular targets are promising biomarkers; however, functional studies are needed to prove these observations.


Subject(s)
Apocrine Glands/chemistry , Biomarkers, Tumor/analysis , Biomarkers, Tumor/genetics , Breast Neoplasms/diagnosis , Carcinoma/diagnosis , Immunohistochemistry , Molecular Diagnostic Techniques , Sweat Gland Neoplasms/diagnosis , Apocrine Glands/pathology , Breast Neoplasms/chemistry , Breast Neoplasms/genetics , Breast Neoplasms/pathology , Carcinoma/chemistry , Carcinoma/genetics , Carcinoma/pathology , Cluster Analysis , Female , Gene Expression Regulation, Neoplastic , Genetic Predisposition to Disease , High-Throughput Nucleotide Sequencing , Humans , In Situ Hybridization, Fluorescence , Multiplex Polymerase Chain Reaction , Phenotype , Predictive Value of Tests , Sweat Gland Neoplasms/chemistry , Sweat Gland Neoplasms/genetics , Sweat Gland Neoplasms/pathology
6.
Medicine (Baltimore) ; 93(15): e81, 2014 Oct.
Article in English | MEDLINE | ID: mdl-25275525

ABSTRACT

Renal oncocytoma is a benign tumor with characteristic histologic findings. We describe an oncocytoma-like renal tumor with progression to high-grade oncocytic carcinoma and metastasis. A 74-year-old man with no family history of cancer presented with hematuria. Computed tomography showed an 11 cm heterogeneous multilobulated mass in the right kidney lower pole, enlarged aortocaval lymph nodes, and multiple lung nodules. In the nephrectomy specimen, approximately one third of the renal tumor histologically showed regions classic for benign oncocytoma transitioning to regions of high-grade carcinoma without sharp demarcation. With extensive genomic investigation using single nucleotide polymorphism-based array virtual karyotyping, multiregion sequencing, and expression array analysis, we were able to show a common lineage between the benign oncocytoma and high-grade oncocytic carcinoma regions in the tumor. We were also able to show karyotypic differences underlying this progression. The benign oncocytoma showed no chromosomal aberrations, whereas the high-grade oncocytic carcinoma showed loss of the 17p region housing FLCN (folliculin [Birt-Hogg-Dubé protein]), loss of 8p, and gain of 8q. Gene expression patterns supported dysregulation and activation of phosphoinositide 3-kinase (PI3K)/v-akt murine thymoma viral oncogene homolog (Akt), mitogen-activated protein kinase (MAPK)/extracellular-signal-regulated kinase (ERK), and mechanistic target of rapamycin (serine/threonine kinase) (mTOR) pathways in the high-grade oncocytic carcinoma regions. This was partly attributable to FLCN underexpression but further accentuated by overexpression of numerous genes on 8q. In the high-grade oncocytic carcinoma region, vascular endothelial growth factor A along with metalloproteinases matrix metallopeptidase 9 and matrix metallopeptidase 12 were overexpressed, facilitating angiogenesis and invasiveness. Genetic molecular testing provided evidence for the development of an aggressive oncocytic carcinoma from an oncocytoma, leading to aggressive targeted treatment but eventual death 39 months after the diagnosis.


Subject(s)
Adenoma, Oxyphilic/pathology , Kidney Neoplasms/pathology , Adenoma, Oxyphilic/genetics , Adenoma, Oxyphilic/immunology , Aged , Disease Progression , Gene Silencing , Genome Components , Humans , Immunohistochemistry , Karyotyping , Kidney Neoplasms/genetics , Kidney Neoplasms/immunology , Male , Microarray Analysis , RNA, Messenger/analysis
7.
Hum Pathol ; 44(1): 47-54, 2013 Jan.
Article in English | MEDLINE | ID: mdl-22939575

ABSTRACT

Juxtaglomerular cell tumors (JGCTs) are rare tumors characterized by renin synthesis, hyperaldosteronism and hypertension. A curious immunohistochemical overlap between JGCT and gastrointestinal stromal tumor (GIST) including the expression of vimentin, CD34, CD117, α-smooth muscle actin was previously reported, prompting us to further investigate JGCT and its phenotypic and molecular genetic characteristics. Virtual karyotyping showed gain of chromosomes 3, 4, 10, 13, 17 and 18 in one JGCT, and fluorescence in situ hybridization (FISH) study confirmed this multiple gain pattern. Additionally, loss of chromosome 9 was observed in four of six cases analyzed with FISH. A whole genome expression analysis revealed 415 up-regulated (including renin, and CD117) and 325 down-regulated genes between the 2 cases. The study confirmed earlier reports on the gain of chromosomes 4 and 10, and provided further evidence of up-regulation of the genes located on these 2 chromosomes. For the first time our study indicated the importance of the loss of chromosome 9 and loss of expression of several tumor suppressor genes located on this chromosome as possible pathogenetic events important in development of JGCT.


Subject(s)
Juxtaglomerular Apparatus/pathology , Adult , Cytoplasmic Granules/genetics , Cytoplasmic Granules/pathology , Cytoplasmic Granules/ultrastructure , Female , Humans , Immunohistochemistry , Juxtaglomerular Apparatus/chemistry , Juxtaglomerular Apparatus/metabolism , Karyotyping , Male , Proto-Oncogene Proteins c-kit/chemistry , Proto-Oncogene Proteins c-kit/genetics , Proto-Oncogene Proteins c-kit/ultrastructure , Renin/chemistry , Renin/genetics , Renin/ultrastructure , Young Adult , beta Catenin/chemistry , beta Catenin/genetics , beta Catenin/ultrastructure
8.
Nat Med ; 18(10): 1503-10, 2012 Oct.
Article in English | MEDLINE | ID: mdl-22983396

ABSTRACT

The discovery of potent inhibitors of the BRAF proto-oncogene has revolutionized therapy for melanoma harboring mutations in BRAF, yet NRAS-mutant melanoma remains without an effective therapy. Because direct pharmacological inhibition of the RAS proto-oncogene has thus far been unsuccessful, we explored systems biology approaches to identify synergistic drug combination(s) that can mimic RAS inhibition. Here, leveraging an inducible mouse model of NRAS-mutant melanoma, we show that pharmacological inhibition of mitogen-activated protein kinase kinase (MEK) activates apoptosis but not cell-cycle arrest, which is in contrast to complete genetic neuroblastoma RAS homolog (NRAS) extinction, which triggers both of these effects. Network modeling pinpointed cyclin-dependent kinase 4 (CDK4) as a key driver of this differential phenotype. Accordingly, combined pharmacological inhibition of MEK and CDK4 in vivo led to substantial synergy in therapeutic efficacy. We suggest a gradient model of oncogenic NRAS signaling in which the output is gated, resulting in the decoupling of discrete downstream biological phenotypes as a result of incomplete inhibition. Such a gated signaling model offers a new framework to identify nonobvious coextinction target(s) for combined pharmacological inhibition in NRAS-mutant melanomas.


Subject(s)
Apoptosis/drug effects , Cyclin-Dependent Kinase 4/antagonists & inhibitors , Melanoma/metabolism , Mitogen-Activated Protein Kinase Kinases/antagonists & inhibitors , Proto-Oncogene Proteins p21(ras)/antagonists & inhibitors , Proto-Oncogene Proteins p21(ras)/metabolism , Animals , Benzimidazoles/pharmacology , Cell Cycle Checkpoints/drug effects , Cell Line, Tumor , Cell Proliferation/drug effects , Cell Survival , Cyclin-Dependent Kinase 4/metabolism , Doxycycline/pharmacology , Female , Genes, ras , Humans , Melanoma/drug therapy , Melanoma/genetics , Melanoma/pathology , Mice , Mice, Knockout , Mice, Nude , Middle Aged , Mitogen-Activated Protein Kinase Kinases/metabolism , Proto-Oncogene Mas , Proto-Oncogene Proteins p21(ras)/genetics , Signal Transduction/drug effects
9.
Biochemistry ; 47(15): 4501-9, 2008 Apr 15.
Article in English | MEDLINE | ID: mdl-18355043

ABSTRACT

Etoposide is a widely prescribed anticancer agent that stabilizes topoisomerase II-mediated DNA strand breaks. The drug contains a polycyclic ring system (rings A-D), a glycosidic moiety at C4, and a pendant ring (E-ring) at C1. A recent study that focused on yeast topoisomerase II demonstrated that the H15 geminal protons of the etoposide A-ring, the H5 and H8 protons of the B-ring, and the H2', H6', 3'-methoxyl, and 5'-methoxyl protons of the E-ring contact topoisomerase II in the binary enzyme-drug complex [ Wilstermann et al. (2007) Biochemistry 46, 8217-8225 ]. No interactions with the C4 sugar were observed. The present study used DNA cleavage assays, saturation transfer difference [ (1)H] NMR spectroscopy, and enzyme-drug binding studies to further define interactions between etoposide and human topoisomerase IIalpha. Etoposide and three derivatives that lacked the C4 sugar were analyzed. Except for the sugar, 4'-demethyl epipodophyllotoxin is identical to etoposide, epipodophyllotoxin contains a 4'-methoxyl group on the E-ring, and 6,7- O, O-demethylenepipodophyllotoxin replaces the A-ring with a diol. Results suggest that etoposide-topoisomerase IIalpha binding is driven by interactions with the A- and B-rings and potentially by stacking interactions with the E-ring. We propose that the E-ring pocket on the enzyme is confined, because the addition of bulk to this ring adversely affects drug function. The A- and E-rings do not appear to contact DNA in the enzyme-drug-DNA complex. Conversely, the sugar moiety subtly alters DNA interactions. The identification of etoposide substituents that contact topoisomerase IIalpha in the binary complex has predictive value for drug behavior in the enzyme-etoposide-DNA complex.


Subject(s)
Antigens, Neoplasm/drug effects , Antineoplastic Agents, Phytogenic/chemistry , DNA Topoisomerases, Type II/drug effects , DNA-Binding Proteins/drug effects , Etoposide/chemistry , Antigens, Neoplasm/chemistry , Antigens, Neoplasm/metabolism , Antineoplastic Agents, Phytogenic/pharmacology , DNA/metabolism , DNA Topoisomerases, Type II/chemistry , DNA Topoisomerases, Type II/metabolism , DNA-Binding Proteins/chemistry , DNA-Binding Proteins/metabolism , Drug Design , Etoposide/analogs & derivatives , Etoposide/pharmacology , Humans , Podophyllotoxin/analogs & derivatives , Podophyllotoxin/chemistry , Protein Binding
10.
Chem Res Toxicol ; 20(6): 975-81, 2007 Jun.
Article in English | MEDLINE | ID: mdl-17516663

ABSTRACT

Several quinone-based metabolites of industrial and environmental toxins are potent topoisomerase II poisons. These compounds act by adducting the protein, and previous studies suggest that they increase levels of enzyme-associated DNA strand breaks by at least two potential mechanisms. Quinones act directly on the DNA cleavage-ligation equilibrium of topoisomerase II by inhibiting the rate of ligation. They also block the N-terminal gate of the protein, thereby stabilizing topoisomerase II in its "closed clamp" form and trapping DNA in the central annulus of the enzyme. It has been proposed that this latter activity enhances DNA cleavage by increasing the population of enzyme molecules with DNA in their active sites, but a causal relationship has not been established. In order to more fully characterize the mechanistic basis for quinone action against topoisomerase II, the present study characterized the sensitivity of human topoisomerase IIalpha carrying a Cys455-->Ala mutation (top2alphaC455A) toward quinones. Cys455 was identified as a site of quinone adduction by mass spectrometry. The mutant enzyme was approximately 1.5-2-fold hypersensitive to 1,4-benzoquinone and the polychlorinated biphenyl quinone 4'Cl-2,5pQ, but it displayed wild-type sensitivity to traditional topoisomerase II poisons. The ability of 1,4-benzoquinone to inhibit DNA ligation mediated by top2alphaC455A was similar to that of wild-type topoisomerase IIalpha. However, the quinone induced approximately 3 times the level of clamp closure with the mutant enzyme. These findings strongly support the hypothesis that the ability of quinones to block the N-terminal gate of the type II enzyme contributes to their actions as topoisomerase II poisons.


Subject(s)
Antigens, Neoplasm/metabolism , DNA Cleavage/drug effects , DNA Topoisomerases, Type II/metabolism , DNA-Binding Proteins/metabolism , Mutation , Quinones/pharmacology , Alanine/chemistry , Alanine/genetics , Alanine/metabolism , Amino Acid Substitution , Antigens, Neoplasm/chemistry , Antigens, Neoplasm/genetics , Benzoquinones/chemistry , Benzoquinones/pharmacology , Cysteine/chemistry , Cysteine/genetics , Cysteine/metabolism , DNA Topoisomerases, Type II/chemistry , DNA Topoisomerases, Type II/genetics , DNA, Superhelical/genetics , DNA, Superhelical/metabolism , DNA-Binding Proteins/chemistry , DNA-Binding Proteins/genetics , Etoposide/chemistry , Etoposide/pharmacology , Humans , Kinetics , Mass Spectrometry , Models, Molecular , Mutant Proteins/chemistry , Mutant Proteins/metabolism , Protein Structure, Tertiary , Quinones/chemistry , Recombinant Proteins/chemistry , Recombinant Proteins/metabolism , Temperature
11.
Biochemistry ; 46(10): 2856-64, 2007 Mar 13.
Article in English | MEDLINE | ID: mdl-17298034

ABSTRACT

Several quinone-based metabolites of drugs and environmental toxins are potent topoisomerase II poisons. These compounds act by adducting the protein and appear to increase levels of enzyme-DNA cleavage complexes by at least two potentially independent mechanisms. Treatment of topoisomerase IIalpha with quinones inhibits DNA religation and blocks the N-terminal gate of the protein by cross-linking its two protomer subunits. It is not known whether these two effects result from adduction of quinone to the same amino acid residue(s) in topoisomerase IIalpha or whether they are mediated by modification of separate residues. Therefore, this study identified amino acid residues in human topoisomerase IIalpha that are modified by quinones and determined their role in the actions of these compounds as topoisomerase II poisons. Four cysteine residues were identified by mass spectrometry as sites of quinone adduction: Cys170, Cys392, Cys405, and Cys455. Mutations (Cys --> Ala) were individually generated at each position. Only mutations at Cys392 or Cys405 reduced sensitivity ( approximately 50% resistance) to benzoquinone. Top2alphaC392A and top2alphaC405A displayed faster rates ( approximately 2-fold) of DNA religation than wild-type topoisomerase IIalpha in the presence of the quinone. In contrast, as determined by DNA binding, protein clamp closing, and protomer cross-linking experiments, mutations at Cys392 and Cys405 did not affect the ability of benzoquinone to block the N-terminal gate of topoisomerase IIalpha. These findings indicate that adduction of Cys392 and Cys405 is important for the actions of quinones against the enzyme and increases levels of cleavage complexes primarily by inhibiting DNA religation.


Subject(s)
Antigens, Neoplasm/metabolism , Benzoquinones/pharmacology , DNA Cleavage/drug effects , DNA Topoisomerases, Type II/metabolism , DNA-Binding Proteins/metabolism , Antigens, Neoplasm/genetics , Cysteine/metabolism , DNA Topoisomerases, Type II/genetics , DNA-Binding Proteins/genetics , Humans , Mutation , Quinones/pharmacology
12.
Biochemistry ; 46(28): 8217-25, 2007 Jul 17.
Article in English | MEDLINE | ID: mdl-17580961

ABSTRACT

Etoposide is one of the most successful chemotherapeutic agents used for the treatment of human cancers. The drug kills cells by inhibiting the ability of topoisomerase II to ligate nucleic acids that it cleaves during the double-stranded DNA passage reaction. Etoposide is composed of a polycyclic ring system (rings A-D), a glycosidic moiety at the C4 position, and a pendent ring (E-ring) at the C1 position. Although drug-enzyme contacts, as opposed to drug-DNA interactions, mediate the entry of etoposide into the topoisomerase II-drug-DNA complex, the substituents on etoposide that interact with the enzyme have not been identified. Therefore, saturation transfer difference [1H]-nuclear magnetic resonance spectroscopy and protein-drug competition binding assays were employed to define the groups on etoposide that associate with yeast topoisomerase II and human topoisomerase IIalpha. Results indicate that the geminal protons of the A-ring, the H5 and H8 protons of the B-ring, and the H2' and H6' protons and the 3'- and 5'-methoxyl protons of the pendent E-ring interact with both enzymes in the binary protein-ligand complexes. In contrast, no significant nuclear Overhauser enhancement signals arising from the C-ring, the D-ring, or the C4 glycosidic moiety were observed with either enzyme, suggesting that there is limited or no contact between these portions of etoposide and topoisomerase II in the binary complex. The functional importance of E-ring substituents was confirmed by topoisomerase II-mediated DNA cleavage assays.


Subject(s)
Antineoplastic Agents, Phytogenic/chemistry , DNA Topoisomerases, Type II/chemistry , Etoposide/chemistry , Antigens, Neoplasm/chemistry , Binding, Competitive , DNA-Binding Proteins/chemistry , Humans , Nuclear Magnetic Resonance, Biomolecular , Protein Structure, Tertiary
13.
Biochemistry ; 45(33): 10140-52, 2006 Aug 22.
Article in English | MEDLINE | ID: mdl-16906772

ABSTRACT

Polychlorinated biphenyls (PCBs) are associated with a broad spectrum of human health problems and cause cancer in rodents. In addition, these compounds cause chromosomal aberrations in humans and treated human cells. Although the underlying basis for the chromosomal damage induced by PCBs is not understood, it is believed that these compounds act through a series of phenolic and quinone-based metabolites. Recent studies indicate that several quinones that promote chromosomal damage also act as topoisomerase II poisons. Therefore, the effects of PCB quinone metabolites (including mono and dichlorinated compounds and p- and o-quinones) on the activity of human topoisomerase IIalpha were examined. Results indicate that these compounds are potent topoisomerase IIalpha poisons in vitro and act by adducting the enzyme. They also increase DNA cleavage by topoisomerase IIalpha in cultured human cells. In contrast, incubation of topoisomerase IIalpha with PCB metabolites in the absence of DNA leads to a rapid loss of enzyme activity. On the basis of (1) the differential ability of quinone-treated enzyme to bind circular and linear DNA molecules and (2) the generation of salt-stable noncovalent complexes between topoisomerase IIalpha and circular plasmids in the presence of PCB quinones, it appears that these compounds alter enzyme function (at least in part) by blocking the N-terminal gate of the protein. Finally, exposure to quinones generates a protein species with a molecular mass approximately twice that of a monomeric topoisomerase IIalpha protomer. This finding suggests that PCB quinones block the N-terminal gate by cross-linking the protomer subunits of topoisomerase IIalpha.


Subject(s)
Chromosome Aberrations/drug effects , DNA Damage/drug effects , DNA-Binding Proteins/antagonists & inhibitors , Polychlorinated Biphenyls/toxicity , Quinones/toxicity , Topoisomerase II Inhibitors , Antigens, Neoplasm/chemistry , Antigens, Neoplasm/metabolism , Cells, Cultured , DNA Topoisomerases, Type II/chemistry , DNA Topoisomerases, Type II/metabolism , DNA-Binding Proteins/chemistry , DNA-Binding Proteins/metabolism , Electrophoresis, Polyacrylamide Gel , Humans , Polychlorinated Biphenyls/metabolism , Quinones/metabolism
14.
Chem Res Toxicol ; 18(4): 761-70, 2005 Apr.
Article in English | MEDLINE | ID: mdl-15833037

ABSTRACT

Although benzene induces leukemias in humans, the compound is not believed to generate chromosomal damage directly. Rather, benzene is thought to act through a series of phenolic- and quinone-based metabolites, especially 1,4-benzoquinone. A recent study found that 1,4-benzoquinone is a potent topoisomerase II poison in vitro and in cultured human cells [Lindsey et al. (2004) Biochemistry 43, 7363-7374]. Because benzene is metabolized to multiple compounds in addition to 1,4-benzoquinone, we determined the effects of several phenolic metabolites, including catechol, 1,2,4-benzenetriol, 1,4-hydroquinone, 2,2'-biphenol, and 4,4'-biphenol, on the DNA cleavage activity of human topoisomerase II alpha. Only 1,4-hydroquinone generated substantial levels of topoisomerase II-mediated DNA scission. DNA cleavage with this compound approached levels observed with 1,4-benzoquinone (approximately 5- vs 8-fold) but required a considerably higher concentration (approximately 250 vs 25 microM). 1,4-Hydroquinone is a precursor to 1,4-benzoquinone in the body and can be activated to the quinone by redox cycling. It is not known whether the effects of 1,4-hydroquinone on human topoisomerase II alpha reflect a lower reactivity of the hydroquinone or a low level of activation to the quinone. The high concentration of 1,4-hydroquinone required to increase enzyme-mediated DNA cleavage is consistent with either explanation. 1,4-Hydroquinone displayed attributes against topoisomerase II alpha, including DNA cleavage specificity, that were similar to those of 1,4-benzoquinone. However, 1,4-hydroquinone consistently inhibited DNA ligation to a greater extent than 1,4-benzoquinone. This latter result implies that the hydroquinone may display (at least in part) independent activity against topoisomerase II alpha. The present findings are consistent with the hypothesis that topoisomerase II alpha plays a role in the initiation of specific types of leukemia that are induced by benzene and its metabolites.


Subject(s)
Antigens, Neoplasm/physiology , Benzene/metabolism , DNA Topoisomerases, Type II/physiology , DNA-Binding Proteins/physiology , DNA/metabolism , Hydroquinones/toxicity , Antigens, Neoplasm/drug effects , Benzene/toxicity , Cells, Cultured , DNA Topoisomerases, Type II/drug effects , DNA-Binding Proteins/drug effects , Humans , Leukemia/chemically induced
15.
Biochemistry ; 43(12): 3731-9, 2004 Mar 30.
Article in English | MEDLINE | ID: mdl-15035644

ABSTRACT

Although acetaminophen is the most widely used analgesic in the world, it is also a leading cause of toxic drug overdoses. Beyond normal therapeutic doses, the drug is hepatotoxic and genotoxic. All of the harmful effects of acetaminophen have been attributed to the production of its toxic metabolite, N-acetyl-p-benzoquinone imine (NAPQI). Since many of the cytotoxic/genotoxic events triggered by NAPQI are consistent with the actions of topoisomerase II-targeted drugs, the effects of this metabolite on human topoisomerase IIalpha were examined. NAPQI was a strong topoisomerase II poison and increased levels of enzyme-mediated DNA cleavage >5-fold at 100 microM. The compound induced scission at a number of DNA sites that were similar to those observed in the presence of the topoisomerase II-targeted anticancer drug etoposide; however, the relative site utilization differed. NAPQI strongly impaired the ability of topoisomerase IIalpha to reseal cleaved DNA molecules, suggesting that inhibition of DNA religation is the primary mechanism underlying cleavage enhancement. In addition to its effects in purified systems, NAPQI appeared to increase levels of DNA scission mediated by human topoisomerase IIalpha in cultured CEM leukemia cells. In contrast, acetaminophen did not significantly affect the DNA cleavage activity of the human enzyme in vitro or in cultured CEM cells. Furthermore, the analgesic did not interfere with the actions of etoposide against the type II enzyme. These results suggest that at least some of the cytotoxic/genotoxic effects caused by acetaminophen overdose may be mediated by the actions of NAPQI as a topoisomerase II poison.


Subject(s)
Acetaminophen/metabolism , Benzoquinones/chemistry , Benzoquinones/poisoning , DNA Topoisomerases, Type II/chemistry , Imines/chemistry , Imines/poisoning , Topoisomerase II Inhibitors , Antigens, Neoplasm , Antineoplastic Agents/chemistry , Benzoquinones/metabolism , Cell Line, Tumor , Chromosome Breakage , DNA Damage/drug effects , DNA Topoisomerases, Type I/chemistry , DNA Topoisomerases, Type II/toxicity , DNA-Binding Proteins , Etoposide/chemistry , Humans , Imines/metabolism , Mutagens/chemistry , Mutagens/metabolism , Mutagens/poisoning
SELECTION OF CITATIONS
SEARCH DETAIL