Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 17 de 17
Filter
Add more filters










Publication year range
1.
Sci Rep ; 9(1): 11227, 2019 08 02.
Article in English | MEDLINE | ID: mdl-31375703

ABSTRACT

DNA damaging chemotherapies are successful in cancer therapy, however, the damage can be reversed by DNA repair mechanisms that may be up-regulated in cancer cells. We hypothesized that inhibiting RAD51, a protein involved in homologous recombination DNA repair, would block DNA repair and restore the effectiveness of DNA damaging chemotherapy. We used phage-display to generate a novel synthetic antibody fragment that bound human RAD51 with high affinity (KD = 8.1 nM) and inhibited RAD51 ssDNA binding in vitro. As RAD51 is an intracellular target, we created a corresponding intrabody fragment that caused a strong growth inhibitory phenotype on human cells in culture. We then used a novel cell-penetrating peptide "iPTD" fusion to generate a therapeutically relevant antibody fragment that effectively entered living cells and enhanced the cell-killing effect of a DNA alkylating agent. The iPTD may be similarly useful as a cell-penetrating peptide for other antibody fragments and open the door to numerous intracellular targets previously off-limits in living cells.


Subject(s)
Antineoplastic Agents, Alkylating/pharmacology , DNA Repair/drug effects , Immunoglobulin Fragments/therapeutic use , Rad51 Recombinase/antagonists & inhibitors , Cell-Penetrating Peptides/pharmacology , Cell-Penetrating Peptides/therapeutic use , Cells, Cultured , DNA Damage/drug effects , Humans , Immunoglobulin Fragments/pharmacology , Peptide Library
2.
Nucleic Acids Res ; 47(9): e50, 2019 05 21.
Article in English | MEDLINE | ID: mdl-30854567

ABSTRACT

Next-generation sequencing (NGS) technologies have been employed in several phage display platforms for analyzing natural and synthetic antibody sequences and for identifying and reconstructing single-chain variable fragments (scFv) and antigen-binding fragments (Fab) not found by conventional ELISA screens. In this work, we developed an NGS-assisted antibody discovery platform by integrating phage-displayed, single-framework, synthetic Fab libraries. Due to limitations in attainable read and amplicon lengths, NGS analysis of Fab libraries and selection outputs is usually restricted to either VH or VL. Since this information alone is not sufficient for high-throughput reconstruction of Fabs, we developed a rapid and simple method for linking and sequencing all diversified CDRs in phage Fab pools. Our method resulted in a reliable and straightforward platform for converting NGS information into Fab clones. We used our NGS-assisted Fab reconstruction method to recover low-frequency rare clones from phage selection outputs. While previous studies chose rare clones for rescue based on their relative frequencies in sequencing outputs, we chose rare clones for reconstruction from less-frequent CDRH3 lengths. In some cases, reconstructed rare clones (frequency ∼0.1%) showed higher affinity and better specificity than high-frequency top clones identified by Sanger sequencing, highlighting the significance of NGS-based approaches in synthetic antibody discovery.


Subject(s)
Cell Surface Display Techniques , Complementarity Determining Regions/genetics , High-Throughput Nucleotide Sequencing , Single-Chain Antibodies/genetics , Antibody Affinity/genetics , Bacteriophages/genetics , Enzyme-Linked Immunosorbent Assay/methods , Humans , Peptide Library
3.
Cell Chem Biol ; 23(3): 381-91, 2016 Mar 17.
Article in English | MEDLINE | ID: mdl-26991103

ABSTRACT

Antibiotic resistance arises from the maintenance of resistance mutations or genes acquired from the acquisition of adaptive de novo mutations or the transfer of resistance genes. Antibiotic resistance is acquired in response to antibiotic therapy by activating SOS-mediated DNA repair and mutagenesis and horizontal gene transfer pathways. Initiation of the SOS pathway promotes activation of RecA, inactivation of LexA repressor, and induction of SOS genes. Here, we have identified and characterized phthalocyanine tetrasulfonic acid RecA inhibitors that block antibiotic-induced activation of the SOS response. These inhibitors potentiate the activity of bactericidal antibiotics, including members of the quinolone, ß-lactam, and aminoglycoside families in both Gram-negative and Gram-positive bacteria. They reduce the ability of bacteria to acquire antibiotic resistance mutations and to transfer mobile genetic elements conferring resistance. This study highlights the advantage of including RecA inhibitors in bactericidal antibiotic therapies and provides a new strategy for prolonging antibiotic shelf life.


Subject(s)
Anti-Bacterial Agents/pharmacology , Drug Resistance, Microbial/drug effects , Enzyme Inhibitors/pharmacology , Gram-Negative Bacteria/drug effects , Gram-Positive Bacteria/drug effects , Indoles/pharmacology , Rec A Recombinases/antagonists & inhibitors , Animals , Anti-Bacterial Agents/chemistry , Enzyme Inhibitors/chemistry , Female , Gram-Negative Bacteria/enzymology , Gram-Positive Bacteria/enzymology , Indoles/chemistry , Mice , Microbial Sensitivity Tests , Molecular Structure , Rec A Recombinases/genetics , Rec A Recombinases/metabolism , Structure-Activity Relationship
4.
BMC Genomics ; 16: 350, 2015 May 06.
Article in English | MEDLINE | ID: mdl-25943404

ABSTRACT

BACKGROUND: DNA methylation has essential roles in transcriptional regulation, imprinting, X chromosome inactivation and other cellular processes, and aberrant CpG methylation is directly involved in the pathogenesis of human imprinting disorders and many cancers. To address the need for a quantitative and highly multiplexed bisulfite sequencing method with long read lengths for targeted CpG methylation analysis, we developed single-molecule real-time bisulfite sequencing (SMRT-BS). RESULTS: Optimized bisulfite conversion and PCR conditions enabled the amplification of DNA fragments up to ~1.5 kb, and subjecting overlapping 625-1491 bp amplicons to SMRT-BS indicated high reproducibility across all amplicon lengths (r=0.972) and low standard deviations (≤0.10) between individual CpG sites sequenced in triplicate. Higher variability in CpG methylation quantitation was correlated with reduced sequencing depth, particularly for intermediately methylated regions. SMRT-BS was validated by orthogonal bisulfite-based microarray (r=0.906; 42 CpG sites) and second generation sequencing (r=0.933; 174 CpG sites); however, longer SMRT-BS amplicons (>1.0 kb) had reduced, but very acceptable, correlation with both orthogonal methods (r=0.836-0.897 and r=0.892-0.927, respectively) compared to amplicons less than ~1.0 kb (r=0.940-0.951 and r=0.948-0.963, respectively). Multiplexing utility was assessed by simultaneously subjecting four distinct CpG island amplicons (702-866 bp; 325 CpGs) and 30 hematological malignancy cell lines to SMRT-BS (average depth of 110X), which identified a spectrum of highly quantitative methylation levels across all interrogated CpG sites and cell lines. CONCLUSIONS: SMRT-BS is a novel, accurate and cost-effective targeted CpG methylation method that is amenable to a high degree of multiplexing with minimal clonal PCR artifacts. Increased sequencing depth is necessary when interrogating longer amplicons (>1.0 kb) and the previously reported bisulfite sequencing PCR bias towards unmethylated DNA should be considered when measuring intermediately methylated regions. Coupled with an optimized bisulfite PCR protocol, SMRT-BS is capable of interrogating ~1.5 kb amplicons, which theoretically can cover ~91% of CpG islands in the human genome.


Subject(s)
DNA Methylation/drug effects , Sequence Analysis, DNA/methods , Sulfites/pharmacology , Cell Line, Tumor , Genome, Human/genetics , Humans , Polymerase Chain Reaction , Time Factors
5.
Chembiochem ; 14(16): 2119-25, 2013 Nov 04.
Article in English | MEDLINE | ID: mdl-24030821

ABSTRACT

Going against tradition: although most kinase inhibitors are ATP competitive, lariat peptides inhibit Abl kinase activity in an ATP-uncompetitive manner. Further, lariat peptides discriminated Src family kinases, and recognize the allosteric region that lies adjacent to the ATP binding pocket in the Abl kinase catalytic cleft.


Subject(s)
Peptides/metabolism , Peptides/pharmacology , Proto-Oncogene Proteins c-abl/antagonists & inhibitors , Adenosine Triphosphate/chemistry , Adenosine Triphosphate/metabolism , Allosteric Regulation , Amino Acid Sequence , Binding Sites , Catalytic Domain , Cell Line, Tumor , Enzyme Activation/drug effects , Humans , Kinetics , Peptides/chemistry , Protein Binding , Protein Kinase Inhibitors/chemistry , Protein Kinase Inhibitors/metabolism , Protein Kinase Inhibitors/pharmacology , Proto-Oncogene Proteins c-abl/metabolism
6.
Chem Biol ; 16(11): 1148-57, 2009 Nov 25.
Article in English | MEDLINE | ID: mdl-19942138

ABSTRACT

Functional genomic analyses provide information that allows hypotheses to be formulated on protein function. These hypotheses, however, need to be validated using reverse genetic approaches, which are difficult to perform on a large scale and in diploid organisms. We developed a genetic screen for isolating "lariat" peptides that function as trans dominant inhibitors of protein function. A lariat consists of a lactone-cyclized peptide with a covalently attached transcription activation domain, which allows combinatorial lariat libraries to be screened for protein interactions using the yeast two-hybrid assay. We isolated lariats against the bacterial repressor protein LexA. LexA regulates bacterial SOS response and LexA mutants that cannot undergo autoproteolysis make bacteria more sensitive to, and inhibit resistance against, cytotoxic reagents. We showed that an anti-LexA lariat blocked LexA autoproteolysis and potentiated the antimicrobial activity of mitomycin C.


Subject(s)
Bacterial Proteins/antagonists & inhibitors , Lactones/chemistry , Peptides, Cyclic/chemistry , Amino Acid Sequence , Bacterial Proteins/genetics , Bacterial Proteins/metabolism , Chromatin Immunoprecipitation , Combinatorial Chemistry Techniques , Genes, Reporter , Lactones/pharmacology , Peptide Library , Peptides, Cyclic/pharmacology , Protein Binding , SOS Response, Genetics , Serine Endopeptidases/genetics , Serine Endopeptidases/metabolism , Surface Plasmon Resonance , Transcriptional Activation , Two-Hybrid System Techniques
7.
Carcinogenesis ; 30(12): 2117-22, 2009 Dec.
Article in English | MEDLINE | ID: mdl-19843644

ABSTRACT

To evaluate loss of imprinting (LOI) and expression of the IGF2 gene in matched esophageal normal and adenocarcinoma tissues, we studied a prospective cohort of 77 patients who underwent esophageal resection between 1998 and 2003. IGF2 imprinting status was determined by reverse transcription-polymerase chain reaction (PCR) following ApaI digestion, and quantitative PCR was used to evaluate IGF2 expression, which was correlated with clinicopathologic findings, disease-free and overall survival. In total, 32% (14/44) of informative tissues showed loss of IGF2 imprinting, with a strong correlation between the tumor and normal esophageal epithelia (Kappa = 0.89, P < 0.01). Normal epithelia with LOI had increased expression of IGF2 [median: 2.91, 95% confidence interval (CI): 0.93-5.06] compared with imprinted normal epithelia (median: 1.13, 95% CI: 0.85-1.39) (P = 0.03). In contrast, tumors with LOI had significantly reduced IGF2 expression (median: 1.87, 95% CI: 0.53-5.21) compared with normally imprinted tumors (median: 6.79, 95% CI: 3.39-15.89) (P = 0.016). Patients below the age of 65 years with normally imprinted tumors had significantly reduced 5 year disease-free survival (DFS) (24%) compared with patients whose tumors had LOI for IGF2 (55%) (P = 0.03). Cox regression analysis showed that IGF2 overexpression was associated with significantly reduced disease-free survival (P = 0.04). We conclude that in a subgroup of younger patients, loss of IGF2 imprinting was associated with improved outcome following esophageal resection. Expression of IGF2 in esophageal adenocarcinoma and normal esophageal epithelia depended on imprinting status and tissue type, suggesting novel molecular regulatory mechanisms in esophageal tumorigenesis.


Subject(s)
Adenocarcinoma/metabolism , Esophageal Neoplasms/metabolism , Esophagus/metabolism , Gene Expression Regulation, Neoplastic , Gene Expression Regulation , Genomic Imprinting , Insulin-Like Growth Factor II/metabolism , Adult , Aged , Aged, 80 and over , Alleles , Cohort Studies , Disease-Free Survival , Female , Genotype , Humans , Male , Middle Aged , Prospective Studies , Treatment Outcome
8.
J Hematol Oncol ; 2: 28, 2009 Jul 14.
Article in English | MEDLINE | ID: mdl-19602237

ABSTRACT

BACKGROUND: RIZ1 expression and activity are reduced in many cancers. In AML cell lines and patient material, RIZ1 expression is reduced relative to normal bone marrow. In chronic myelogenous leukemia (CML), blastic transformation is associated with loss of heterozygosity in the region where RIZ1 is located. RIZ1 is a PR domain methyltransferase that methylates histone H3 lysine 9, a modification important for transcriptional repression. In CML blast crisis cell lines RIZ1 represses insulin-like growth factor-1 expression and autocrine signaling. Together these observations suggest that RIZ1 may have a role in the chronic phase to blast crisis transition in CML. RESULTS: In CML patient material, we observed that RIZ1 expression was decreased during progression from chronic phase to blast crisis. RIZ1 was expressed in mature myeloid and CD34+ cells demonstrating that decreased RIZ1 expression in blast crisis is not due to an increased immature cell population. Expression of RIZ1 CML blast crisis cell lines decreased proliferation, increased apoptosis, and enhanced differentiation. CONCLUSION: RIZ1 is a candidate tumor suppressor gene whose expression is decreased in blast crisis. Loss of RIZ1 activity results in decreased apoptosis and differentiation and enhanced proliferation. Together these results suggest that loss of RIZ1 expression will lead to an increase in myeloid blast cell population resulting in CML progression.


Subject(s)
DNA-Binding Proteins/physiology , Gene Expression Regulation, Leukemic , Genes, Tumor Suppressor , Leukemia, Myelogenous, Chronic, BCR-ABL Positive/genetics , Leukemia, Myelogenous, Chronic, BCR-ABL Positive/pathology , Nuclear Proteins/physiology , Transcription Factors/physiology , Blast Crisis/genetics , Blast Crisis/pathology , Cell Differentiation/genetics , Cells, Cultured , DNA-Binding Proteins/genetics , Disease Progression , Down-Regulation , Gene Expression Regulation, Leukemic/physiology , Genes, Tumor Suppressor/physiology , Histone-Lysine N-Methyltransferase , Humans , K562 Cells , Nuclear Proteins/genetics , Transcription Factors/genetics , Transfection
9.
Histol Histopathol ; 23(11): 1309-14, 2008 11.
Article in English | MEDLINE | ID: mdl-18785112

ABSTRACT

Friend leukemia integration site 1 (Fli-1) has been reported as the first nuclear marker of endothelial differentiation; it is expressed in leukocytes and recently demonstrated in melanomas. Formalin-fixed, paraffin-embedded tissue sections from 97 melanomas including 69 cases of primary and 28 metastatic melanomas were evaluated by immunohistochemistry. Five melanoma cell lines were evaluated by Western blot and immunocytochemistry. Fli-1 expression was observed in all cell lines. Fli-1 expression was higher in metastatic than in primary tumors (r=0.208, p=0.041, Spearman correlation), it positively correlated with Ki-67 expression (r=0.233, p=0.022, Spearman correlation), and the presence of an ulcer in the primary tumor (r=0.267, p=0.030, Spearman correlation). Therefore, the expression of Fli-1 in malignant melanoma appears to be associated with biologically more aggressive tumors.


Subject(s)
Biomarkers, Tumor/metabolism , Melanoma/metabolism , Proto-Oncogene Protein c-fli-1/metabolism , Adult , Aged , Aged, 80 and over , Blotting, Western , Cell Proliferation , Humans , Immunohistochemistry , Jurkat Cells , Melanoma/pathology , Middle Aged , Neoplasm Invasiveness , Neoplasm Metastasis
10.
Cancer Genet Cytogenet ; 186(1): 54-7, 2008 Oct.
Article in English | MEDLINE | ID: mdl-18786443

ABSTRACT

The case of a patient with follicular dendritic cell (FDC) sarcoma with chromosomal aberration add(21)(q11.2) is described. Cytogenetic studies showed the karyotype 46,XX,add(21)(q11.2)[3]/46,XX[17], although the encoded protein involved was not clarified. The abnormal pattern was quite simple, and different from a previous report. The clinical course of the FDC sarcoma in this case has been indolent, as for most FDC sarcoma patients. Although this patient suffered from breast carcinoma 6 years after the onset of FDC sarcoma, the carcinoma showed different histological and phenotypic profiles.


Subject(s)
Chromosome Aberrations , Chromosomes, Human, Pair 21 , Dendritic Cell Sarcoma, Follicular/genetics , Neoplasms, Multiple Primary/genetics , Adenocarcinoma, Scirrhous/genetics , Axilla , Biomarkers, Tumor/analysis , Breast Neoplasms/genetics , Carcinoma, Ductal, Breast/genetics , Dendritic Cell Sarcoma, Follicular/pathology , Fatal Outcome , Female , Humans , Immunophenotyping , Lymph Nodes/pathology , Middle Aged , Neoplasm Proteins/analysis , Receptors, Complement 3d/analysis
11.
Exp Hematol ; 35(2): 263-73, 2007 Feb.
Article in English | MEDLINE | ID: mdl-17258075

ABSTRACT

OBJECTIVE: The p15INK4B tumor suppressor is frequently silenced by promoter hypermethylation in myelodysplastic syndrome and acute myeloid leukemia (AML). Clinically approved DNA methylation inhibitors, such as 5-aza-2'-deoxycytidine, can reverse p15INK4B promoter methylation, but widespread clinical use of these inhibitors is limited by their toxicity and instability in aqueous solution. The cytidine analog zebularine is a stable DNA methylation inhibitor that has minimal toxicity in vitro and in vivo. We evaluated zebularine effects on p15INK4B reactivation and cell growth in vitro to investigate a potential role for zebularine in treating myeloid malignancies. METHODS: We examined the specific effects of zebularine on reexpression of transcriptionally silenced p15INK4B and its global effects on cell cycle and apoptosis in AML cell lines and primary patient samples. RESULTS: Zebularine treatment of AML193, which has a densely methylated p15INK4B promoter, results in a dose-dependent increase in p15INK4B expression that correlates with CpG island promoter demethylation and enrichment of local histone acetylation. We observed enhanced p15INK4B induction following co-treatment with zebularine and the histone deacetylase inhibitor Trichostatin A. Zebularine inhibits cell proliferation, arrests cells at G(2)/M, and induces apoptosis at dosages that effectively demethylate the p15INK4B promoter. Zebularine treatment of KG-1 cells and AML patient blasts with hypermethylated p15INK4B promoters also reactivates p15INK4B reexpression and induces apoptosis. CONCLUSION: Zebularine is an effective inhibitor of p15INK4B methylation and cell growth in human AML in vitro. Our results extend the spectrum of zebularine effects to nonepithelial malignancies and provide a strong rationale for evaluating its clinical utility in the treatment of myeloid malignancies.


Subject(s)
Cell Proliferation/drug effects , Cyclin-Dependent Kinase Inhibitor p15/genetics , Cytidine/analogs & derivatives , DNA Methylation/drug effects , Leukemia, Myeloid/metabolism , Promoter Regions, Genetic/drug effects , Acetylation , Acute Disease , Apoptosis/drug effects , Cell Division/drug effects , Cell Line, Tumor , Cyclin-Dependent Kinase Inhibitor p15/drug effects , Cyclin-Dependent Kinase Inhibitor p15/metabolism , Cytidine/pharmacology , Dose-Response Relationship, Drug , Drug Screening Assays, Antitumor , G2 Phase/drug effects , Gene Expression Profiling , HL-60 Cells , Histones/drug effects , Histones/metabolism , Humans , Hydroxamic Acids/pharmacology , Leukemia, Myeloid/drug therapy , Phosphorylation , Promoter Regions, Genetic/genetics , Reverse Transcriptase Polymerase Chain Reaction/methods
12.
Am J Clin Pathol ; 126(5): 798-804, 2006 Nov.
Article in English | MEDLINE | ID: mdl-17050077

ABSTRACT

We evaluated 123 formalin-fixed, paraffin-embedded samples, including neuroendocrine tumors, adult brain, mesonephric tissues, and from various other sites. A pre-B lymphoma cell line, Daudi, and a small cell carcinoma cell line, NCL-H128, were evaluated by Western blot. All tissues were immunostained by mouse monoclonal anti-Pax-5 antibody by using standard, synthetic polymer-based detection methods. Our study describes for the first time distribution of Pax-5 in adult brain tissue, including periaqueductal gray matter of the midbrain, area postrema of the medulla oblongata, and occasional cells of the spinal trigeminal nucleus (caudal nucleus). We confirm that Pax-5 is expressed regularly in poorly differentiated neuroendocrine tumors but never in well-differentiated classic carcinoid tumors. Pax-5 expression also was found readily in benign and malignant mesonephric tissues and focally in müllerian duct-derived tissues and tumors. Expression of Pax-5 in the Daudi and NCL-H128 cell lines was confirmed by Western blot. Together, these results are important for correct interpretation of results in immunophenotyping of undifferentiated tumors, for diagnosis of mesonephric carcinoma, and, potentially, for correct classification of neuroendocrine tumors in small biopsy samples.


Subject(s)
Carcinoma, Small Cell/metabolism , Neuroendocrine Tumors/metabolism , PAX5 Transcription Factor/biosynthesis , Adult , Blotting, Western , Carcinoma, Small Cell/pathology , Cell Line, Tumor , Female , Gastrointestinal Neoplasms/metabolism , Gastrointestinal Neoplasms/pathology , Humans , Immunohistochemistry , Lung Neoplasms/metabolism , Lung Neoplasms/pathology , Neuroendocrine Tumors/pathology , Pancreatic Neoplasms/metabolism , Pancreatic Neoplasms/pathology , Uterine Cervical Neoplasms/metabolism , Uterine Cervical Neoplasms/pathology
13.
Leuk Res ; 30(1): 69-76, 2006 Jan.
Article in English | MEDLINE | ID: mdl-16043219

ABSTRACT

Decitabine is a potent demethylating agent that exhibits clinical activity against myeloid malignancies. Numerous genes silenced by hypermethylation are reactivated by decitabine through a mechanism involving promoter demethylation with subsequent release of histone deacetylases (HDACs) and accumulation of acetylated histones. Recent studies indicating that decitabine also induces regional chromatin remodeling of some unmethylated genes suggest additional mechanisms of action. Decitabine reactivates unmethylated p21WAF1 in some AML cell lines but the possible occurrence of p21WAF1 methylation in AML in vivo has not been studied in detail and decitabine effects on p21WAF1 chromatin remodeling have not been reported. We found that p21WAF1 mRNA was undetectable in 6 of 24 AML patient samples and 4 of 5 AML cell lines but there was no evidence of p21WAF1 promoter methylation. However, decitabine induced p21WAF1 in AML cell lines KG-1 and KG-1a in association with release of HDAC1 and increased acetylated histone H3 at the unmethylated p21WAF1 promoter. Decitabine effects on p21WAF1 histone acetylation and induction were enhanced by the HDAC inhibitor trichostatin A and were independent of wild type p53. Our findings indicate that decitabine can relieve p21WAF1 repression in AML by a mechanism that involves release of HDAC1 without requiring promoter demethylation. Furthermore, our study provides evidence that combined decitabine and HDAC inhibitor treatment can enhance chromatin remodeling and reactivation of an unmethylated tumor suppressor gene. This latter finding is of relevance to the clinical use of these agents in AML as we found the p21WAF1 promoter to be unmethylated in vivo.


Subject(s)
Azacitidine/analogs & derivatives , Cyclin-Dependent Kinase Inhibitor p21/metabolism , DNA Methylation , Enzyme Inhibitors/pharmacology , Histone Deacetylases/metabolism , Leukemia, Myeloid, Acute/mortality , Neoplasm Proteins/metabolism , Promoter Regions, Genetic , Acylation/drug effects , Azacitidine/pharmacology , Azacitidine/therapeutic use , Chromatin Assembly and Disassembly/drug effects , DNA Methylation/drug effects , Decitabine , Enzyme Inhibitors/therapeutic use , Histone Deacetylase 1 , Histone Deacetylase Inhibitors , Histones/metabolism , Humans , Hydroxamic Acids/pharmacology , K562 Cells , Leukemia, Myeloid, Acute/drug therapy , Neoplasm Proteins/antagonists & inhibitors , Promoter Regions, Genetic/drug effects , Protein Processing, Post-Translational/drug effects , Tumor Suppressor Protein p53/metabolism
14.
Leuk Res ; 28(12): 1293-301, 2004 Dec.
Article in English | MEDLINE | ID: mdl-15475071

ABSTRACT

Epigenetic silencing of downstream components of the transforming growth factor beta pathway including the cyclin-dependent kinase inhibitors (CDKIs) p15INK4B, p27KIP1 and p21CIP1 is implicated in the pathogenesis of some hematological malignancies. Loss of p15INK4B expression due to promoter methylation occurs frequently in human T-cell acute lymphoblastic leukemia (T-ALL) but the expression and methylation status of p27KIP1 remains to be characterized in T-ALL or T-cell lymphoblastic lymphoma (T-LBL). As well, while some have reported a high frequency of p21CIP1 methylation in ALL patient samples others have found the gene to be unmethylated in this disease and the relationship between p21CIP1 expression and promoter methylation has not been examined in T-LBL. Using RNase protection assays (RPA) and methylation specific PCR (MSP), we found p27KIP1 to be expressed and its promoter unmethylated in 20 of 20 (100%) and 28 of 28 (100%) T-LBL/ALL samples, respectively. In contrast, p21CIP1 mRNA was absent in 7 of 14 (50%) T-LBL biopsies and 5 of 6 (83%) T-ALL cell lines. However, like p27KIP1 there was no evidence of p21CIP1 promoter methylation by MSP or temporal temperature gradient electrophoresis (TTGE) analysis of 35 CpG sites in any of the 28 T-LBL/ALLs analyzed. Similar to T-ALL, we found p15INK4B mRNA was absent in 13 of 14 (93%) T-LBL biopsies and its promoter methylated in 6 of 10 (64%) cases. Our results indicate that p21CIP1 mRNA is absent in human T-LBL biopsies and T-ALL cell lines at a high frequency. However, unlike p15INK4B, reduced p21CIP1 expression in T-LBL/ALL is independent of dense promoter-associated CpG methylation. In contrast to some hematological malignancies p27KIP1 methylation does not appear to contribute significantly to T-LBL/ALL pathogenesis.


Subject(s)
Cyclin-Dependent Kinases/antagonists & inhibitors , DNA Methylation , Epigenesis, Genetic , Leukemia-Lymphoma, Adult T-Cell/genetics , Transforming Growth Factor beta/metabolism , Carrier Proteins/genetics , Cell Cycle Proteins/genetics , Cyclin-Dependent Kinase Inhibitor p15 , Cyclin-Dependent Kinase Inhibitor p21 , Cyclin-Dependent Kinase Inhibitor p27 , Cyclins/genetics , DNA Primers , Gene Expression Regulation, Neoplastic , Humans , Intracellular Signaling Peptides and Proteins/genetics , Leukemia-Lymphoma, Adult T-Cell/etiology , Polymerase Chain Reaction/methods , Promoter Regions, Genetic/genetics , Tumor Suppressor Proteins/genetics
15.
Cancer ; 100(5): 913-9, 2004 Mar 01.
Article in English | MEDLINE | ID: mdl-14983485

ABSTRACT

BACKGROUND: The positive replication error (RER+) phenotype defines a distinct subgroup of tumors with specific clinical, pathologic, and molecular features that have been documented well in hereditary nonpolyposis colon carcinoma (HNPCC). More recently, this phenotype also has been described in breast carcinoma. METHODS: To determine the effect of RER phenotype on prognosis in patients with breast carcinoma, the authors examined matched archival tumor and normal tissue from 100 women with Stage I and Stage II breast carcinoma, all of whom were treated with hormonal therapy. Patients had been followed for a minimum of 5 years or until death. Seven microsatellite loci were examined, including hMLH1 (3p22, D3S1611), hMSH2 (2p16, D2S123), NM23-H1 (17q21), TP53-Dint (17p13), TP53-Penta (17p13), APC (5q21, D5S346), and HPC1 (1q24, D1S2883). The RER+ phenotype was defined as the presence of allelic shifts at three of the seven loci examined. RESULTS: Twenty-five percent of patients were classified with the RER+ phenotype based on these criteria. The two groups, women with positive RER status and women with negative RER status, were comparable in terms of other factors that may influence prognosis: age, tumor size, lymph node status, disease stage, and estrogen receptor status. The development of distant metastases to the lung, liver, or brain was correlated significantly with the positive RER phenotype, with a relative risk of 2.625 (95% confidence interval, 1.059-6.057). CONCLUSIONS: The presence of high-frequency RER+ may predict for the development of distant metastatic disease in patients with early-stage breast carcinoma who are treated with hormonal therapy.


Subject(s)
Breast Neoplasms/genetics , Carcinoma/drug therapy , Carcinoma/genetics , DNA Replication , DNA, Neoplasm/genetics , Neoplasm Invasiveness/genetics , Aged , Analysis of Variance , Antineoplastic Agents, Hormonal/therapeutic use , Biopsy, Needle , Breast Neoplasms/drug therapy , Breast Neoplasms/mortality , Breast Neoplasms/pathology , Carcinoma/secondary , Case-Control Studies , Culture Techniques , Female , Humans , Microsatellite Repeats , Middle Aged , Neoplasm Invasiveness/pathology , Neoplasm Metastasis , Neoplasm Staging , Phenotype , Polymerase Chain Reaction , Predictive Value of Tests , Probability , Prognosis , Proportional Hazards Models , Retrospective Studies , Risk Assessment , Statistics, Nonparametric , Survival Analysis
16.
Leuk Res ; 27(1): 23-34, 2003 Jan.
Article in English | MEDLINE | ID: mdl-12479849

ABSTRACT

In solid cancers, defective DNA mismatch repair (MMR) is most commonly caused by hMSH2 or hMLH1 mutations, or epigenetic silencing of hMLH1 by promoter hypermethylation, and results in the acquisition of characteristic frameshift microsatellite mutations of mononucleotide repeats located within the coding regions of defined target genes. We previously identified hMSH2 mutations in T-cell lymphoblastic lymphoma (T-LBL) patient tumor samples and others have reported coding region microsatellite mutations in T-cell acute lymphoblastic leukemia (T-ALL) cell lines. Thus, while MMR gene mutations are known to occur in some human T-lymphoblastic tumors in vivo, it is still unknown if the coding region microsatellite mutations detected in human cell lines also occur in vivo or if hMLH1 or hMSH2 promoter hypermethylation contributes to defective MMR in these tumors. We analyzed the TGFbetaRII (A)10 and caspase-5 (A)10 coding region repeats in 16 human T-LBL/ALL patient tumor samples and identified six with microsatellite mutations in one or both repeats. There was no evidence of hMSH2 or hMLH1 promoter methylation as assessed by standard methylation specific PCR or by a novel temporal temperature gradient electrophoresis (TTGE) method that analyzed 25 and 30 CpG sites in the hMLH1 and hMSH2 promoters, respectively. Our results indicate that coding region microsatellite mutations characteristic of defective MMR occur in some human T-LBL/ALL in vivo but not as a consequence of hMLH1 or hMSH2 promoter hypermethylation. Furthermore, the identification of TGFbetaRII and caspase-5 coding region mutations in vivo implicates these genes in the pathogenesis of human T-LBL/ALL.


Subject(s)
Base Pair Mismatch , Caspases/genetics , DNA Methylation , DNA Repair/genetics , DNA-Binding Proteins , Leukemia, T-Cell/genetics , Lymphoma, T-Cell/genetics , Microsatellite Repeats , Mutation , Neoplasm Proteins/genetics , Promoter Regions, Genetic/genetics , Proto-Oncogene Proteins/genetics , Receptors, Transforming Growth Factor beta/genetics , Adaptor Proteins, Signal Transducing , Adolescent , Adult , Base Sequence , Carrier Proteins , Caspases/physiology , CpG Islands , DNA Mutational Analysis , DNA, Neoplasm/genetics , Female , Humans , Male , Middle Aged , Molecular Sequence Data , MutL Protein Homolog 1 , MutS Homolog 2 Protein , Nuclear Proteins , Protein Serine-Threonine Kinases , Receptor, Transforming Growth Factor-beta Type II , Receptors, Transforming Growth Factor beta/physiology
17.
Lab Invest ; 82(9): 1103-9, 2002 Sep.
Article in English | MEDLINE | ID: mdl-12218070

ABSTRACT

The atypical cells of CD30(+) cutaneous lymphoproliferative disorders (CD30CLD) are commonly of T-cell origin and frequently have a similar morphology as Hodgkin or Reed-Sternberg cells of Hodgkin's lymphoma (HL). HL is one of the tumors associated with CD30CLD. Although most studies support a B-cell derivation of the tumor cells in HL, recently a few cases of classical HL with T-cell genotype have been reported. We report a patient who presented with CD30CLD whose lymph nodes showed classical HL of mixed cellularity subtype at presentation. By single-cell PCR, the same clonal gene rearrangements of the T cell receptor-beta gene locus could be assigned to the CD30(+) and CD15(+) cells of both skin and lymph node. In a lymph node biopsy specimen taken in relapse after several courses of chemotherapy, the CD30(+) tumor cells were abundant. The T cell-derived tumor cells displayed aberrant expression of the Pax-5 gene in all specimens. A common clonal origin of both CD30CLD and HL of the lymph node in the patient presented here suggests that HL with T-cell genotype exists in association with CD30CLD as well as in sporadic cases and may share clonal origin with the skin tumor.


Subject(s)
Hodgkin Disease/pathology , Ki-1 Antigen/analysis , Lymph Nodes/pathology , Lymphoma, T-Cell/pathology , Lymphoproliferative Disorders/pathology , Polymerase Chain Reaction/methods , Skin Diseases/pathology , DNA-Binding Proteins/analysis , Female , Gene Rearrangement, beta-Chain T-Cell Antigen Receptor , Hodgkin Disease/classification , Humans , Immunohistochemistry , Lewis X Antigen/analysis , Middle Aged , PAX5 Transcription Factor , Transcription Factors/analysis
SELECTION OF CITATIONS
SEARCH DETAIL
...