Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 17 de 17
Filter
1.
Materials (Basel) ; 13(11)2020 May 29.
Article in English | MEDLINE | ID: mdl-32485911

ABSTRACT

The effect of electric potential on the sulfide capacity of the CaO-SiO2-Al2O3 system was evaluated by applying voltages in the range of -1.5 to 1.5 V at 1823 K in a C/CO gas equilibrium. When the cathodic potential (-1.5 to 0 V) was applied, it was confirmed that the sulfur partition ratio increased based on the electrochemical reaction of sulfur (S + 2e- = S2-). However, the reversibility of the electrochemical resulfurization reaction (S2- = S + 2e-) in slag was not established in the reverse (anodic) potential region (0-1.5 V), yet the sulfur partition ratio increased. In particular, sulfur evaporation was observed in the anodic potential region. Therefore, in the present study, potential anodic electro-desulfurization mechanisms based on sulfur evaporation are proposed. To verify these mechanisms, sulfur evaporation is discussed in detail as a function of the thermodynamic stability of sulfur in the slag.

2.
Cancer Genet ; 237: 19-38, 2019 09.
Article in English | MEDLINE | ID: mdl-31447063

ABSTRACT

Folate-mediated one-carbon metabolism is essential for growth and survival of cancer cells. We investigated whether the response of cancer cells to antitumor treatment may be partially influenced by variation in expression of one-carbon metabolism genes. We used cancer cell line information from the Cancer Cell Line Encyclopedia and the Genomics of Drug Sensitivity in Cancer resources to examine whether variation in pretreatment expression of one-carbon metabolism-related genes was associated with response to treatment. GART, TYMS, SHMT2, MTR, ALDH2, BHMT, MAT2B, MTHFD2, NNMT, and SLC46A1 showed modest statistically significant correlations with response to a variety of antitumor agents. Higher expression levels of SLC46A1 were associated with resistance to multiple agents, whereas elevated expression of GART, TYMS, SHMT2, MTR, BHMT, and MAT2B was associated with chemosensitivity to multiple drugs. NNMT expression was bimodally distributed and showed different directions of association with various agents. Correlation of increased NNMT expression with sensitivity to dasatinib was validated in the NCI-60 cancer cell line panel. Pretreatment expression levels were correlated among many one-carbon metabolism genes. Expression of several folate genes was strongly associated with expression of multiple components of drug target pathways. Molecular mechanisms underlying associations of one-carbon metabolism gene with drug response require further investigation.


Subject(s)
Antineoplastic Agents/therapeutic use , Carbon/metabolism , Folic Acid/metabolism , Neoplasms/drug therapy , Neoplasms/genetics , Transcription, Genetic , Cell Line, Tumor , Gene Expression Profiling , Humans
3.
Cancer Chemother Pharmacol ; 84(4): 771-780, 2019 10.
Article in English | MEDLINE | ID: mdl-31367787

ABSTRACT

PURPOSE: Genotoxic agents (GAs) including cisplatin, doxorubicin, gemcitabine, and topotecan are often used in cancer treatment. However, the response to GAs is variable among patients and predictive biomarkers are inadequate to select patients for treatment. Accurate and rapid pharmacodynamics measures of response can, thus, be useful for monitoring therapy and improve clinical outcomes. METHODS: This study focuses on integrating a database of genome-wide response to treatment (The NCI Transcriptional Pharmacodynamics Workbench) with a database of baseline gene expression (GSE32474) for the NCI-60 cell lines to identify mechanisms of response and pharmacodynamic (PD) biomarkers. RESULTS AND CONCLUSIONS: Our analysis suggests that GA-induced endoplasmic reticulum (ER) stress may signal for GA-induced cell death. Reducing the uptake of GA, activating DNA repair, and blocking ER-stress induction cooperate to prevent GA-induced cell death in the GA-resistant cells. ATF3, DDIT3, CARS, and PPP1R15A appear as possible candidate PD biomarkers for monitoring the progress of GA treatment. Further validation studies on the proposed intrinsic drug-resistant mechanism and candidate genes are needed using in vivo data from either patient-derived xenograft models or clinical chemotherapy trials.


Subject(s)
Antineoplastic Agents/pharmacokinetics , Cell Death , DNA Damage , Drug Resistance, Neoplasm/genetics , Endoplasmic Reticulum Stress , Mutagens/pharmacokinetics , Activating Transcription Factor 3/genetics , Biomarkers, Pharmacological/analysis , Cell Death/drug effects , Cell Death/physiology , Cell Line, Tumor , DNA Damage/drug effects , DNA Damage/genetics , Endoplasmic Reticulum Stress/drug effects , Endoplasmic Reticulum Stress/physiology , Gene Expression Profiling/methods , Genome-Wide Association Study/statistics & numerical data , Humans , Protein Phosphatase 1/genetics , ROC Curve , Transcription Factor CHOP/genetics
4.
Environ Technol ; 40(26): 3512-3522, 2019 Nov.
Article in English | MEDLINE | ID: mdl-29799331

ABSTRACT

A study on selective separation of Zn from a leaching solution by disposal batteries including various type batteries was carried out to understand the recovery behaviour of Zn in leaching solution. Selective recovery of Zn in leaching solution including Mn, Cd, Cu ion was difficult due to its similar physicochemical behaviour. Experiment results by present leaching solution with 279 µm undersize indicated that the best condition for leaching is 1 M H2SO4, 250 rpm, 5 vol.% H2O2 and 353 K and the leaching efficient of Zn, Co and Mn is approximately 97%, respectively. The exclusive extraction behaviour of Zn by using D2EHPA is indicated that the best conditions for solvent extraction are to be 0.6 M D2EHPA diluted with kerosene, 30% saponification, 298 K, 5-min contact time and three-stage countercurrent extraction, and the O/A ratio 1, respectively. Recovery of Zn was with approximately 99.7% selectively from Mn, Co, Ni, Cd and Li. After scrubbing 5 times by pH 2 modified solution and single stripping experiment by 1.5 M H2SO4, the solution including Zn of 9.0 g/L can be produced.


Subject(s)
Recycling , Zinc , Electric Power Supplies , Hydrogen Peroxide , Metals
5.
Anticancer Res ; 36(6): 2649-57, 2016 Jun.
Article in English | MEDLINE | ID: mdl-27272773

ABSTRACT

BACKGROUND: Triple-negative breast cancer (TNBC) is an aggressive form of breast cancer currently lacking targeted therapies. Our previous work demonstrated a therapeutic synergism with gemcitabine (GEM) and the CHK1 inhibitor (AZD7762) combination treatment in a TNBC cell line. We hypothesized that the response to this combination therapy would differ among heterogeneous TNBC patients and that addition of a SMAC mimetic (TL32711) could improve efficacy. MATERIALS AND METHODS: Therapeutic responses to GEM, GEM/AZD7762, and GEM/AZD7762/TL32711 combinations were investigated by XTT assays and western blotting of cell cycle and apoptosis-related proteins in ten TNBC cell lines. RESULTS: TNBC cell lines harboring low levels of endogenous CHK1, cIAP1 and cIAP2 were responsive to GEM alone, whereas cell lines demonstrating a minimal increase in phospho-S345 CHK1 after treatment were responsive to GEM/AZD7762 or GEM/AZD7762/TL32711 combination. CONCLUSION: The response of TNBC cells to particular therapies varies and will require development of predictive biomarkers.


Subject(s)
Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Deoxycytidine/analogs & derivatives , Dipeptides/administration & dosage , Indoles/administration & dosage , Thiophenes/administration & dosage , Triple Negative Breast Neoplasms/drug therapy , Urea/analogs & derivatives , Apoptosis/drug effects , Cell Line, Tumor , Checkpoint Kinase 1/metabolism , DNA Repair , Deoxycytidine/administration & dosage , Female , Humans , Phosphorylation , Triple Negative Breast Neoplasms/pathology , Urea/administration & dosage , Gemcitabine
6.
Blood ; 117(1): 211-20, 2011 Jan 06.
Article in English | MEDLINE | ID: mdl-20974671

ABSTRACT

The multiple myeloma SET domain (MMSET) protein is overexpressed in multiple myeloma (MM) patients with the translocation t(4;14). Although studies have shown the involvement of MMSET/Wolf-Hirschhorn syndrome candidate 1 in development, its mode of action in the pathogenesis of MM is largely unknown. We found that MMSET is a major regulator of chromatin structure and transcription in t(4;14) MM cells. High levels of MMSET correlate with an increase in lysine 36 methylation of histone H3 and a decrease in lysine 27 methylation across the genome, leading to a more open structural state of the chromatin. Loss of MMSET expression alters adhesion properties, suppresses growth, and induces apoptosis in MM cells. Consequently, genes affected by high levels of MMSET are implicated in the p53 pathway, cell cycle regulation, and integrin signaling. Regulation of many of these genes required functional histone methyl-transferase activity of MMSET. These results implicate MMSET as a major epigenetic regulator in t(4;14)+ MM.


Subject(s)
Chromosomes, Human, Pair 14/genetics , Chromosomes, Human, Pair 4/genetics , DNA Methylation , Histone-Lysine N-Methyltransferase/genetics , Histones/genetics , Multiple Myeloma/genetics , Repressor Proteins/genetics , Translocation, Genetic/genetics , Apoptosis , Biomarkers, Tumor/genetics , Biomarkers, Tumor/metabolism , Blotting, Western , Cell Adhesion , Cell Cycle , Cell Movement , Cell Proliferation , Chromatin/genetics , Chromatin Immunoprecipitation , Epigenomics , Gene Expression Profiling , Gene Expression Regulation, Neoplastic , Histone-Lysine N-Methyltransferase/antagonists & inhibitors , Histone-Lysine N-Methyltransferase/metabolism , Humans , Oligonucleotide Array Sequence Analysis , Protein Isoforms , RNA, Messenger/genetics , RNA, Small Interfering/genetics , Repressor Proteins/antagonists & inhibitors , Repressor Proteins/metabolism , Reverse Transcriptase Polymerase Chain Reaction , Tumor Cells, Cultured
7.
Cancer Cell ; 18(6): 539-41, 2010 Dec 14.
Article in English | MEDLINE | ID: mdl-21156276

ABSTRACT

In this issue of Cancer Cell, Rui et al. identify JAK2 and JMJDC2 as two contiguous, coamplified oncogenes in primary mediastinal B cell and Hodgkin lymphoma. Together, JAK2 and JMJD2C induce major changes in chromatin structure and gene expression. Targeting these proteins with small molecules represents a new avenue for therapy.

8.
Clin Cancer Res ; 16(17): 4339-52, 2010 Sep 01.
Article in English | MEDLINE | ID: mdl-20601445

ABSTRACT

PURPOSE: To understand the changes in gene expression in polycythemia vera (PV) progenitor cells and their relationship to JAK2V617F. EXPERIMENTAL DESIGN: Messenger RNA isolated from CD34(+) cells from nine PV patients and normal controls was profiled using Affymetrix arrays. Gene expression change mediated by JAK2V617F was determined by profiling CD34(+) cells transduced with the kinase and by analysis of leukemia cell lines harboring JAK2V617F, treated with an inhibitor. RESULTS: A PV expression signature was enriched for genes involved in hematopoietic development, inflammatory responses, and cell proliferation. By quantitative reverse transcription-PCR, 23 genes were consistently deregulated in all patient samples. Several of these genes such as WT1 and KLF4 were regulated by JAK2, whereas others such as NFIB and EVI1 seemed to be deregulated in PV by a JAK2-independent mechanism. Using cell line models and comparing gene expression profiles of cell lines and PV CD34(+) PV specimens, we have identified panels of 14 JAK2-dependent genes and 12 JAK2-independent genes. These two 14- and 12-gene sets could separate not only PV from normal CD34(+) specimens, but also other MPN such as essential thrombocytosis and primary myelofibrosis from their normal counterparts. CONCLUSIONS: A subset of the aberrant gene expression in PV progenitor cells can be attributed to the action of the mutant kinase, but there remain a significant number of genes characteristic of the disease but deregulated by as yet unknown mechanisms. Genes deregulated in PV as a result of the action of JAK2V617F or independent of the kinase may represent other targets for therapy.


Subject(s)
Gene Expression Profiling , Janus Kinase 2/genetics , Mutation , Polycythemia Vera/genetics , Amino Acid Substitution , Antigens, CD34/genetics , Antigens, CD34/metabolism , Bone Marrow Cells/metabolism , Cell Differentiation/drug effects , Cell Differentiation/genetics , Cell Line, Tumor , Cell Proliferation/drug effects , Cells, Cultured , Cluster Analysis , Erythroid Cells/cytology , Erythroid Cells/metabolism , Erythropoietin/pharmacology , Humans , Janus Kinase 2/metabolism , Kruppel-Like Factor 4 , Oligonucleotide Array Sequence Analysis , Polycythemia Vera/blood , Polycythemia Vera/pathology , Reverse Transcriptase Polymerase Chain Reaction , Transfection
9.
J Clin Oncol ; 26(27): 4376-84, 2008 Sep 20.
Article in English | MEDLINE | ID: mdl-18802149

ABSTRACT

PURPOSE: To identify children with acute lymphoblastic leukemia (ALL) at initial diagnosis who are at risk for inferior response to therapy by using molecular signatures. PATIENTS AND METHODS: Gene expression profiles were generated from bone marrow blasts at initial diagnosis from a cohort of 99 children with National Cancer Institute-defined high-risk ALL who were treated uniformly on the Children's Oncology Group (COG) 1961 study. For prediction of early response, genes that correlated to marrow status on day 7 were identified on a training set and were validated on a test set. An additional signature was correlated with long-term outcome, and the predictive models were validated on three large, independent patient cohorts. Results We identified a 24-probe set signature that was highly predictive of day 7 marrow status on the test set (P = .0061). Pathways were identified that may play a role in early blast regression. We have also identified a 47-probe set signature (which represents 41 unique genes) that was predictive of long-term outcome in our data set as well as three large independent data sets of patients with childhood ALL who were treated on different protocols. However, we did not find sufficient evidence for the added significance of these genes and the derived predictive models when other known prognostic features, such as age, WBC, and karyotype, were included in a multivariate analysis. CONCLUSION: Genes and pathways that play a role in early blast regression may identify patients who may be at risk for inferior responses to treatment. A fully validated predictive gene expression signature was defined for high-risk ALL that provided insight into the biologic mechanisms of treatment failure.


Subject(s)
Gene Expression Profiling , Precursor Cell Lymphoblastic Leukemia-Lymphoma/diagnosis , Precursor Cell Lymphoblastic Leukemia-Lymphoma/genetics , Adolescent , Child , Child, Preschool , Female , Gene Expression Regulation, Neoplastic/genetics , Genetic Markers , Humans , Infant , Male , Models, Statistical , Precursor Cell Lymphoblastic Leukemia-Lymphoma/therapy , Predictive Value of Tests , ROC Curve , Risk Assessment , Treatment Outcome
10.
Blood ; 111(6): 3145-54, 2008 Mar 15.
Article in English | MEDLINE | ID: mdl-18156491

ABSTRACT

MMSET, identified by its fusion to the IgH locus in t(4;14)-associated multiple myeloma, possesses domains found within chromatin regulators, including the SET domain. MMSET protein is overexpressed and highly associated with chromatin in myeloma cell lines carrying t(4;14). MMSET possesses methyltransferase activity for core histone H3 lysine 4 and histone 4 lysine 20, whereas MMSET made in cells only modified H4. Segments of MMSET fused to the Gal4 DNA binding domain repressed transcription of a chromatin-embedded Gal4 reporter gene. MMSET-mediated repression was associated with increased H4K20 methylation gene and loss of histone acetylation. Consistent with this repressive activity, MMSET could form a complex with HDAC1 and HDAC2, mSin3a, and the histone demethylase LSD1, suggesting that it is a component of corepressor complexes. Furthermore, MMSET coexpression enhances HDAC1- and HDAC2-mediated repression in transcriptional reporter assays. Finally, shRNA-mediated knockdown of MMSET compromised viability of a myeloma cell line, suggesting a biologic role for the protein in malignant cell growth. Collectively, these data suggest that, by acting directly as a modifier of chromatin as well as through binding of other chromatin-modifying enzymes, MMSET influences gene expression and potentially acts as a pathogenic agent in multiple myeloma.


Subject(s)
Gene Expression Regulation , Histone-Lysine N-Methyltransferase/metabolism , Repressor Proteins/metabolism , Transcription, Genetic/genetics , Animals , Catalysis , Cell Line , Cell Nucleus/enzymology , Cell Survival , Chromosomes, Human, Pair 4/genetics , Histone-Lysine N-Methyltransferase/chemistry , Histone-Lysine N-Methyltransferase/genetics , Histones/metabolism , Humans , Isoenzymes/genetics , Isoenzymes/metabolism , Melanoma/enzymology , Melanoma/genetics , Mice , Models, Molecular , Protein Binding , Protein Structure, Tertiary , Rabbits , Repressor Proteins/chemistry , Repressor Proteins/genetics
11.
Apoptosis ; 11(11): 1977-86, 2006 Nov.
Article in English | MEDLINE | ID: mdl-17013760

ABSTRACT

Cancer cell resistance to chemotherapy may be mediated by defects in apoptotic pathways. A prior study showed that in vivo apoptosis of Acute Lymphoblastic Leukemia (ALL) blasts in response to chemotherapy could occur through diverse pathways including both p53-dependent and -independent mechanisms. In this study we investigated the apoptotic response in more detail by using a panel of ALL cell lines that differed in respect to p53 status. Upon exposure to a uniform stimulus, expression of apoptotic proteins, including the effector caspase-3, varied among ALL cell lines partly depending on p53 transcriptional activity and caspase-8 activation. Although the expression and contribution to apoptosis differed among known members of the apoptotic pathway, apoptosis was universally mediated by mitochondrial depolarization. The NFkappaB pathway was activated in response to chemotherapy but NFkappaB inhibition appeared to not influence chemosensitivity. This study further documents the highly variable nature of cell death programs in ALL and provides the foundation for cell death pathway modulation to improve ALL cure rates without increasing chemotherapy-related toxicity.


Subject(s)
Antineoplastic Agents, Phytogenic/therapeutic use , Apoptosis , Etoposide/therapeutic use , Precursor Cell Lymphoblastic Leukemia-Lymphoma/drug therapy , Caspase 8/metabolism , Cell Line, Tumor , Humans , Mitochondria/metabolism , NF-kappa B/metabolism , Precursor Cell Lymphoblastic Leukemia-Lymphoma/pathology , Tumor Suppressor Protein p53/metabolism
12.
Blood ; 108(2): 711-7, 2006 Jul 15.
Article in English | MEDLINE | ID: mdl-16822902

ABSTRACT

Outcome for children with childhood acute lymphoblastic leukemia (ALL) who relapse is poor. To gain insight into the mechanisms of relapse, we analyzed gene-expression profiles in 35 matched diagnosis/relapse pairs as well as 60 uniformly treated children at relapse using the Affymetrix platform. Matched-pair analyses revealed significant differences in the expression of genes involved in cell-cycle regulation, DNA repair, and apoptosis between diagnostic and early-relapse samples. Many of these pathways have been implicated in tumorigenesis previously and are attractive targets for intervention strategies. In contrast, no common pattern of changes was observed among late-relapse pairs. Early-relapse samples were more likely to be similar to their respective diagnostic sample while we noted greater divergence in gene-expression patterns among late-relapse pairs. Comparison of expression profiles of early- versus late-relapse samples indicated that early-relapse clones were characterized by overexpression of biologic pathways associated with cell-cycle regulation. These results suggest that early-relapse results from the emergence of a related clone, characterized by the up-regulation of genes mediating cell proliferation. In contrast, late relapse appears to be mediated by diverse pathways.


Subject(s)
Gene Expression Profiling , Precursor Cell Lymphoblastic Leukemia-Lymphoma/pathology , Apoptosis/genetics , Cell Cycle/genetics , Cell Proliferation , Child , Clone Cells/pathology , DNA Repair/genetics , Humans , Matched-Pair Analysis , Precursor Cell Lymphoblastic Leukemia-Lymphoma/genetics , Recurrence , Time Factors
13.
Pediatr Blood Cancer ; 47(2): 130-40, 2006 Aug.
Article in English | MEDLINE | ID: mdl-16358311

ABSTRACT

BACKGROUND: T-cell acute lymphoblastic leukemia (T-ALL) and T-cell lymphoblastic lymphoma (T-LL) and are often thought to represent a spectrum of a single disease. The malignant cells in T-ALL and T-LL are morphologically indistinguishable, and they share the expression of common cell surface antigens and cytogenetic characteristics. However, despite these similarities, differences in the clinical behavior of T-ALL and T-LL are observed. PROCEDURE: We analyzed the gene expression profiles of T-ALL and T-LL samples obtained from Children's Oncology Group (COG) tumor banks using DNA arrays. Immunohistochemistry was also performed to validate the expression of selected targets. RESULTS: Unsupervised hierarchical clustering of all samples showed complete segregation of T-ALL and T-LL into distinct clusters. Next, we identified the top 201 genes that best differentiated T-ALL from T-LL using significance analysis of microarrays (SAM), a supervised statistical approach. Genes representing several functional groups were differentially expressed in T-LL and T-ALL. Prediction analysis of microarrays (PAM) identified a subset of genes, which accurately classified all 19 T-ALL and T-LL samples with an overall misclassification error rate of 0. Immunohistochemical validation of protein expression of selected genes identified by microarray analysis confirmed overexpression of MLL-1 in T-LL tumor cells compared to T-ALL and CD47 in T-ALL tumors cells when compared to T-LL. CONCLUSIONS: Despite significant similarities between the malignant T-cell precursors, clear differences in the gene expression profiles were observed between T-ALL and T-LL implying underlying differences in the biology of the two entities.


Subject(s)
Gene Expression Profiling , Leukemia-Lymphoma, Adult T-Cell/genetics , Leukemia-Lymphoma, Adult T-Cell/pathology , Precursor Cell Lymphoblastic Leukemia-Lymphoma/genetics , Precursor Cell Lymphoblastic Leukemia-Lymphoma/pathology , Bone Marrow/pathology , Child , Cluster Analysis , Gene Expression Regulation, Neoplastic/genetics , Humans , Immunohistochemistry , Oligonucleotide Array Sequence Analysis , Tumor Cells, Cultured
14.
Pediatr Blood Cancer ; 46(5): 570-8, 2006 May 01.
Article in English | MEDLINE | ID: mdl-16365862

ABSTRACT

The recent sequencing of the human genome and technical breakthroughs now make it possible to simultaneously determine mRNA expression levels of almost all of the identified genes in the human genome. DNA "chip" or microarray technology holds great promise for the development of more refined, biologically-based classification systems for childhood ALL, as well as the identification of new targets for novel therapy. To date gene expression profiles have been described that correlate with subtypes of ALL defined by morphology, immunophenotype, cytogenetic alterations, and response to therapy. Mechanistic insights into treatment failure have come from the definition of mRNA signatures that predict in vitro chemoresistance, as well as differences between blasts at relapse and new diagnosis. New bioinformatics tools optimize data mining, but validation of findings is essential since "over-fitting" the data is a common danger. In the future, genomic analysis will be complemented by evaluation of the cancer proteome.


Subject(s)
Precursor Cell Lymphoblastic Leukemia-Lymphoma/genetics , Child , Computational Biology , Gene Expression Profiling , Gene Expression Regulation, Leukemic , Genomics , Humans , Microarray Analysis , Neoplasm Proteins/genetics , Neoplasm Proteins/metabolism , Oligonucleotide Array Sequence Analysis , Precursor Cell Lymphoblastic Leukemia-Lymphoma/metabolism , Precursor Cell Lymphoblastic Leukemia-Lymphoma/pathology , Signal Transduction
15.
Bioconjug Chem ; 16(6): 1617-23, 2005.
Article in English | MEDLINE | ID: mdl-16287262

ABSTRACT

The genetic addition of hexahistidine (H(6)) tags is widely used to isolate recombinant proteins by immobilized metal-affinity chromatography (IMAC). Addition of a tyrosine residue to H(6) tags enabled proteins to be covalently cross-linked under mild conditions in a manner similar to the natural, site-specific cross-linking of tyrosines into dityrosine. A series of seven hexahistidine tags with tyrosines placed in various positions (H(6)Y tags) were added to the amino terminus of the I28 immunoglobulin domain of the human cardiac titin. The H(6)Y-tagged I28 dimerized in the presence of excess Ni(2+) with a K(D) of 200 microM. Treatment of Ni(2+)-dimerized H(6)Y-I28 with an oxidant, monoperoxyphthalic acid (MMPP) or sodium sulfite, resulted in covalent protein multimerization through chelated Ni(2+)-catalyzed cross-linking of the Y residues engineered into the H(6) tag. The protein oligomerization was observed by sodium dodecyl sulfate polyacrylamide gel electrophoresis (SDS PAGE). The presence of dityrosine in the cross-linked proteins was confirmed by fluorescence emission at 410 nm. Proteins lacking the Y residue in the H(6) tag treated with the same oxidative conditions did not cross-link or exhibit dityrosine fluorescence, despite the presence of an endogenous Y residue. The method may have potential uses in other protein conjugation applications such as protein labeling and interfacial immobilization of proteins on artificial surfaces.


Subject(s)
Cross-Linking Reagents/chemistry , Molecular Probes/chemical synthesis , Proteins/chemistry , Tyrosine/chemistry , Binding Sites , Connectin , Histidine , Humans , Muscle Proteins/chemistry , Protein Engineering , Protein Kinases/chemistry
16.
Per Med ; 2(4): 349-361, 2005 Nov.
Article in English | MEDLINE | ID: mdl-29788576

ABSTRACT

In the field of oncology, a growing emphasis is now being placed on individualizing treatment in a way that maximizes chance for cure while minimizing unwanted side effects. In childhood acute lymphoblastic leukemia (ALL), several well-established clinical and biologic prognostic variables have traditionally been used to risk stratify therapy for individual patients. While this approach has been very successful, many relapses still occur unpredictably in patients characterized as having favorable features of their disease at diagnosis. Furthermore, it is likely that other children are overtreated. Therefore, current initiatives in childhood leukemia have focused on identifying new prognostic markers to refine treatment decision-making. Recent advances, which include the sequencing of the human genome, and technical developments in high-throughput genomics and proteomics, have facilitated these efforts. This review will chart the evolution of individualized therapy for ALL, the most common malignancy of children.

17.
Article in English | MEDLINE | ID: mdl-14633779

ABSTRACT

The outcome for children with acute lymphoblastic leukemia (ALL) has improved dramatically with current therapy resulting in an event free survival exceeding 75% for most patients. However significant challenges remain including developing better methods to predict which patients can be cured with less toxic treatment and which ones will benefit from augmented therapy. In addition, 25% of patients fail therapy and novel treatments that are focused on undermining specifically the leukemic process are needed urgently. In Section I, Dr. Carroll reviews current approaches to risk classification and proposes a system that incorporates well-established clinical parameters, genetic lesions of the blast as well as early response parameters. He then provides an overview of emerging technologies in genomics and proteomics and how they might lead to more rational, biologically based classification systems. In Section II, Drs. Mary Relling and Stella Davies describe emerging findings that relate to host features that influence outcome, the role of inherited germline variation. They highlight technical breakthroughs in assessing germline differences among patients. Polymorphisms of drug metabolizing genes have been shown to influence toxicity and the best example is the gene thiopurine methyltransferase (TPMT) a key enzyme in the metabolism of 6-mercaptopurine. Polymorphisms are associated with decreased activity that is also associated with increased toxicity. The role of polymorphisms in other genes whose products play an important role in drug metabolism as well as cytokine genes are discussed. In Sections III and IV, Drs. James Downing and Cheryl Willman review their findings using gene expression profiling to classify ALL. Both authors outline challenges in applying this methodology to analysis of clinical samples. Dr. Willman describes her laboratory's examination of infant leukemia and precursor B-ALL where unsupervised approaches have led to the identification of inherent biologic groups not predicted by conventional morphologic, immunophenotypic and cytogenetic variables. Dr. Downing describes his results from a pediatric ALL expression database using over 327 diagnostic samples, with 80% of the dataset consisting of samples from patients treated on a single institutional protocol. Seven distinct leukemia subtypes were identified representing known leukemia subtypes including: BCR-ABL, E2A-PBX1, TEL-AML1, rearrangements in the MLL gene, hyperdiploid karyotype (i.e., > 50 chromosomes), and T-ALL as well as a new leukemia subtype. A subset of genes have been identified whose expression appears to be predictive of outcome but independent verification is needed before this type of analysis can be integrated into treatment assignment. Chemotherapeutic agents kill cancer cells by activating apoptosis, or programmed cell death. In Section V, Dr. John Reed describes major apoptotic pathways and the specific role of key proteins in this response. The expression level of some of these proteins, such as BCL2, BAX, and caspase 3, has been shown to be predictive of ultimate outcome in hematopoietic tumors. New therapeutic approaches that modulate the apoptotic pathway are now available and Dr. Reed highlights those that may be applicable to the treatment of childhood ALL.


Subject(s)
Precursor Cell Lymphoblastic Leukemia-Lymphoma/genetics , Precursor Cell Lymphoblastic Leukemia-Lymphoma/therapy , Child , Gene Expression Profiling/methods , Gene Expression Regulation, Neoplastic , Humans , Pharmacogenetics , Precursor Cell Lymphoblastic Leukemia-Lymphoma/classification , Prognosis , Risk Assessment
SELECTION OF CITATIONS
SEARCH DETAIL
...