Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 15 de 15
Filter
Add more filters










Publication year range
1.
Front Pharmacol ; 12: 792600, 2021.
Article in English | MEDLINE | ID: mdl-35095503

ABSTRACT

Tranexamic Acid (TA) is a clinically used antifibrinolytic agent that acts as a Lys mimetic to block binding of Plasminogen with Plasminogen activators, preventing conversion of Plasminogen to its proteolytically activated form, Plasmin. Previous studies suggested that TA may exhibit anticancer activity by blockade of extracellular Plasmin formation. Plasmin-mediated cleavage of the CDCP1 protein may increase its oncogenic functions through several downstream pathways. Results presented herein demonstrate that TA blocks Plasmin-mediated excision of the extracellular domain of the oncoprotein CDCP1. In vitro studies indicate that TA reduces the viability of a broad array of human and murine cancer cell lines, and breast tumor growth studies demonstrate that TA reduces cancer growth in vivo. Based on the ability of TA to mimic Lys and Arg, we hypothesized that TA may perturb multiple processes that involve Lys/Arg-rich protein sequences, and that TA may alter intracellular signaling pathways in addition to blocking extracellular Plasmin production. Indeed, TA-mediated suppression of tumor cell viability is associated with multiple biochemical actions, including inhibition of protein synthesis, reduced activating phosphorylation of STAT3 and S6K1, decreased expression of the MYC oncoprotein, and suppression of Lys acetylation. Further, TA inhibited uptake of Lys and Arg by cancer cells. These findings suggest that TA or TA analogs may serve as lead compounds and inspire the production of new classes of anticancer agents that function by mimicking Lys and Arg.

2.
Mol Pharmacol ; 98(2): 168-180, 2020 08.
Article in English | MEDLINE | ID: mdl-32474444

ABSTRACT

The two major nicotinic acetylcholine receptors (nAChRs) in the brain are the α4ß2 and α7 subtypes. A "methyl scan" of the pyrrolidinium ring was used to detect differences in nicotine's interactions with these two receptors. Each methylnicotine was investigated using voltage-clamp and radioligand binding techniques. Methylation at each ring carbon elicited unique changes in nicotine's receptor interactions. Replacing the 1'-N-methyl with an ethyl group or adding a second 1'-N-methyl group significantly reduced interaction with α4ß2 but not α7 receptors. The 2'-methylation uniquely enhanced binding and agonist potency at α7 receptors. Although 3'- and 5'-trans-methylations were much better tolerated by α7 receptors than α4ß2 receptors, 4'-methylation decreased potency and efficacy at α7 receptors much more than at α4ß2 receptors. Whereas cis-5'-methylnicotine lacked agonist activity and displayed a low affinity at both receptors, trans-5'-methylnicotine retained considerable α7 receptor activity. Differences between the two 5'-methylated analogs of the potent pyridyl oxymethylene-bridged nicotine analog A84543 were consistent with what was found for the 5'-methylnicotines. Computer docking of the methylnicotines to the Lymnaea acetylcholine binding protein crystal structure containing two persistent waters predicted most of the changes in receptor affinity that were observed with methylation, particularly the lower affinities of the cis-methylnicotines. The much smaller effects of 1'-, 3'-, and 5'-methylations and the greater effects of 2'- and 4'-methylations on nicotine α7 nAChR interaction might be exploited for the design of new drugs based on the nicotine scaffold. SIGNIFICANCE STATEMENT: Using a comprehensive "methyl scan" approach, we show that the orthosteric binding sites for acetylcholine and nicotine in the two major brain nicotinic acetylcholine receptors interact differently with the pyrrolidinium ring of nicotine, and we suggest reasons for the higher affinity of nicotine for the heteromeric receptor. Potential sites for nicotine structure modification were identified that may be useful in the design of new drugs targeting these receptors.


Subject(s)
Nicotine/analogs & derivatives , Pyridines/chemical synthesis , Receptors, Nicotinic/metabolism , alpha7 Nicotinic Acetylcholine Receptor/metabolism , Animals , Binding Sites , Male , Methylation , Molecular Docking Simulation , Molecular Structure , Nicotine/chemistry , Pyridines/chemistry , Pyridines/pharmacology , Rats , Structure-Activity Relationship , Xenopus laevis
3.
Drug Metab Dispos ; 48(7): 563-569, 2020 07.
Article in English | MEDLINE | ID: mdl-32357971

ABSTRACT

Previous work has shown that hepatic levels of human glutathione transferase zeta 1 (GSTZ1) protein, involved in tyrosine catabolism and responsible for metabolism of the investigational drug dichloroacetate, increase in cytosol after birth before reaching a plateau around age 7. However, the mechanism regulating this change of expression is still unknown, and previous studies showed that GSTZ1 mRNA levels did not correlate with GSTZ1 protein expression. In this study, we addressed the hypothesis that microRNAs (miRNAs) could regulate expression of GSTZ1. We obtained liver samples from donors aged less than 1 year or older than 13 years and isolated total RNA for use in a microarray to identify miRNAs that were downregulated in the livers of adults compared with children. From a total of 2578 human miRNAs tested, 63 miRNAs were more than 2-fold down-regulated in adults, of which miR-376c-3p was predicted to bind to the 3' untranslated region of GSTZ1 mRNA. There was an inverse correlation of miR-376c-3p and GSTZ1 protein expression in the liver samples. Using cell culture, we confirmed that miR-376c-3p could downregulate GSTZ1 protein expression. Our findings suggest that miR-376c-3p prevents production of GSTZ1 through inhibition of translation. These experiments further our understanding of GSTZ1 regulation. Furthermore, our array results provide a database resource for future studies on mechanisms regulating human hepatic developmental expression. SIGNIFICANCE STATEMENT: Hepatic glutathione transferase zeta 1 (GSTZ1) is responsible for metabolism of the tyrosine catabolite maleylacetoacetate as well as the investigational drug dichloroacetate. Through examination of microRNA (miRNA) expression in liver from infants and adults and studies in cells, we showed that expression of GSTZ1 is controlled by miRNA. This finding has application to the dosing regimen of the drug dichloroacetate. The miRNA expression profiles are provided and will prove useful for future studies of drug-metabolizing enzymes in infants and adults.


Subject(s)
Aging/genetics , Down-Regulation , Gene Expression Regulation, Developmental , Glutathione Transferase/genetics , MicroRNAs/metabolism , 3' Untranslated Regions/genetics , Adolescent , Adult , Aged , Aged, 80 and over , Aging/metabolism , Female , Gene Expression Profiling , Glutathione Transferase/metabolism , HEK293 Cells , Hep G2 Cells , Hepatobiliary Elimination/genetics , Humans , Infant , Infant, Newborn , Liver/enzymology , Liver/growth & development , Male , Middle Aged , RNA, Messenger/analysis , RNA, Messenger/metabolism , Young Adult
4.
Chem Res Toxicol ; 32(10): 2042-2052, 2019 10 21.
Article in English | MEDLINE | ID: mdl-31524376

ABSTRACT

Dichloroacetate (DCA) has potential for treating mitochondrial disorders and cancer by activating the mitochondrial pyruvate dehydrogenase complex. Repeated dosing of DCA results in reduced drug clearance due to inactivation of glutathione transferase ζ1 (GSTZ1), its metabolizing enzyme. We investigated the time-course of inactivation of GSTZ1 in hepatic cytosol and mitochondria after one oral dose of 100 mg/kg DCA to female Sprague-Dawley rats aged 4 weeks (young) and 52 weeks (adult) as models for children and adults, respectively. GSTZ1 activity with both DCA and an endogenous substrate, maleylacetone (MA), as well as GSTZ1 protein expression were rapidly reduced in cytosol from both ages following DCA treatment. In mitochondria, loss of GSTZ1 protein and activity with DCA were even more rapid. The cytosolic in vivo half-lives of the loss of GSTZ1 activity with DCA were 1.05 ± 0.03 and 0.82 ± 0.02 h (mean ± S.D., n = 6) for young and adult rats, respectively, with inactivation significantly more rapid in adult rats, p < 0.001. The mitochondrial inactivation half-lives were similar in young (0.57 ± 0.02 h) and adult rats (0.54 ± 0.02 h) and were significantly (p < 0.0001) shorter than cytosolic inactivation half-lives. By 24 h after DCA administration, activity and expression remained at 10% or less than control values. The in vitro GSTZ1 inactivation half-lives following incubation with 2 mM DCA in the presence of physiological chloride (Cl-) concentrations (cytosol = 44 mM, mitochondria = 1-2 mM) exhibited marked differences between subcellular fractions, being 3 times longer in the cytosol than in the mitochondria, regardless of age, suggesting that the lower Cl- concentration in mitochondria explained the faster degradation of GSTZ1. These results demonstrate for the first time that rat mitochondrial GSTZ1 is more readily inactivated by DCA than cytosolic GSTZ1, and cytosolic GSTZ1 is inactivated more rapidly in adult than young rats.


Subject(s)
Cytosol/enzymology , Dichloroacetic Acid/pharmacology , Dichloroacetic Acid/toxicity , Glutathione Transferase/antagonists & inhibitors , Liver/drug effects , Mitochondria/drug effects , Animals , Dichloroacetic Acid/administration & dosage , Female , Glutathione Transferase/metabolism , Liver/metabolism , Mitochondria/metabolism , Rats , Rats, Sprague-Dawley
5.
Drug Metab Dispos ; 46(8): 1118-1128, 2018 08.
Article in English | MEDLINE | ID: mdl-29853471

ABSTRACT

Glutathione transferase zeta1 (GSTZ1) catalyzes glutathione (GSH)-dependent dechlorination of dichloroacetate (DCA), an investigational drug with therapeutic potential in metabolic disorders and cancer. GSTZ1 is expressed in both hepatic cytosol and mitochondria. Here, we examined the ontogeny and characterized the properties of human mitochondrial GSTZ1. GSTZ1 expression and activity with DCA were determined in 103 human hepatic mitochondrial samples prepared from livers of donors aged 1 day to 84 years. DNA from each sample was genotyped for three common GSTZ1 functional single nucleotide polymorphisms. Expression of mitochondrial GSTZ1 protein increased in an age-dependent manner to a plateau after age 21 years. Activity with DCA correlated with expression, after taking into account the somewhat higher activity of samples that were homo- or heterozygous for GSTZ1A. In samples from livers with the GSTZ1C variant, apparent enzyme kinetic constants for DCA and GSH were similar for mitochondria and cytosol after correcting for the loss of GSH observed in mitochondrial incubations. In the presence of 38 mM chloride, mitochondrial GSTZ1 exhibited shorter half-lives of inactivation compared with the cytosolic enzyme (P = 0.017). GSTZ1 protein isolated from mitochondria was shown by mass spectrometry to be identical to cytosolic GSTZ1 protein in the covered primary protein sequence. In summary, we report age-related development in the expression and activity of human hepatic mitochondrial GSTZ1 does not have the same pattern as that reported for cytosolic GSTZ1. Some properties of cytosolic and mitochondrial GSTZ1 differed, but these were not related to differences in amino acid sequence or post-translationally modified residues.


Subject(s)
Glutathione Transferase/genetics , Liver/metabolism , Mitochondria/genetics , Mitochondria/metabolism , Adolescent , Adult , Aged , Aged, 80 and over , Amino Acid Sequence , Child , Child, Preschool , Cytosol/metabolism , Dichloroacetic Acid/metabolism , Drugs, Investigational/metabolism , Female , Glutathione Transferase/metabolism , Humans , Infant , Kinetics , Male , Middle Aged , Polymorphism, Single Nucleotide/genetics , Young Adult
6.
Biochem Pharmacol ; 152: 236-243, 2018 06.
Article in English | MEDLINE | ID: mdl-29626439

ABSTRACT

Biotransformation of dichloroacetate (DCA) to glyoxylate by hepatic glutathione transferase zeta 1 (GSTZ1) is considered the principal determinant of the rate of plasma clearance of the drug. However, several other organismal and subcellular factors are also known to influence DCA metabolism. We utilized a female rat model to study these poorly understood processes. Rats aged 4 weeks (young) and 42-52 weeks (adult) were used to model children and adults, respectively. Hepatic chloride concentrations, which influence the rate of GSTZ1 inactivation by DCA, were lower in rat than in human tissues and rats did not show the age dependence previously seen in humans. We found GSTZ1 expression and activity in rat brain, heart, and kidney cell-free homogenates that were age-dependent. GSTZ1 expression in brain was higher in young rats than adult rats, whereas cardiac and renal GSTZ1 expression levels were higher in adult than young rats. GSTZ1 activity with DCA could not be measured accurately in kidney cell-free homogenates due to rapid depletion of glutathione by γ-glutamyl transpeptidase. Following oral administration of DCA, 100 mg/kg, to rats, GSTZ1 expression and activity were reduced in all rat tissues, but chloride concentrations were not affected. Together, these data extend our understanding of factors that determine the in vivo kinetics of DCA.


Subject(s)
Chlorides/metabolism , Dichloroacetic Acid/metabolism , Glutathione Transferase/metabolism , Liver/metabolism , Animals , Brain/metabolism , Female , Gene Expression Regulation, Enzymologic , Glutathione , Glutathione Transferase/genetics , Kidney/metabolism , Mitochondria/metabolism , Myocardium/metabolism , Rats , Rats, Sprague-Dawley
7.
Pharmacol Ther ; 170: 166-180, 2017 02.
Article in English | MEDLINE | ID: mdl-27771434

ABSTRACT

Dichloroacetate (DCA) has several therapeutic applications based on its pharmacological property of inhibiting pyruvate dehydrogenase kinase. DCA has been used to treat inherited mitochondrial disorders that result in lactic acidosis, as well as pulmonary hypertension and several different solid tumors, the latter through its ability to reverse the Warburg effect in cancer cells and restore aerobic glycolysis. The main clinically limiting toxicity is reversible peripheral neuropathy. Although administration of high doses to rodents can result in liver cancer, there is no evidence that DCA is a human carcinogen. In all studied species, including humans, DCA has the interesting property of inhibiting its own metabolism upon repeat dosing, resulting in alteration of its pharmacokinetics. The first step in DCA metabolism is conversion to glyoxylate catalyzed by glutathione transferase zeta 1 (GSTZ1), for which DCA is a mechanism-based inactivator. The rate of GSTZ1 inactivation by DCA is influenced by age, GSTZ1 haplotype and cellular concentrations of chloride. The effect of DCA on its own metabolism complicates the selection of an effective dose with minimal side effects.


Subject(s)
Dichloroacetic Acid/administration & dosage , Glutathione Transferase/metabolism , Protein Serine-Threonine Kinases/antagonists & inhibitors , Animals , Dichloroacetic Acid/adverse effects , Dichloroacetic Acid/pharmacology , Dose-Response Relationship, Drug , Humans , Hypertension, Pulmonary/drug therapy , Mitochondrial Diseases/drug therapy , Neoplasms/drug therapy , Neoplasms/pathology , Pyruvate Dehydrogenase Acetyl-Transferring Kinase
8.
Biochim Biophys Acta ; 1860(6): 1202-10, 2016 Jun.
Article in English | MEDLINE | ID: mdl-26850694

ABSTRACT

Dichloroacetate (DCA), commonly used to treat metabolic disorders, is under investigation as an anti-cancer therapy due to its ability to reverse the Warburg effect and induce apoptosis in tumor cells. While DCA's mechanism of action is well-studied, other factors that influence its potential as a cancer treatment have not been thoroughly investigated. Here we show that expression of glutathione transferase zeta 1 (GSTZ1), the enzyme responsible for conversion of DCA to its inactive metabolite, glyoxylate, is downregulated in liver cancer and upregulated in some breast cancers, leading to abnormal expression of the protein. The cellular concentration of chloride, an ion that influences the stability of GSTZ1 in the presence of DCA, was also found to be abnormal in tumors, with consistently higher concentrations in hepatocellular carcinoma than in surrounding non-tumor tissue. Finally, results from experiments employing two- and three-dimensional cultures of HepG2 cells, parental and transduced to express GSTZ1, demonstrate that high levels of GSTZ1 expression confers resistance to the effect of high concentrations of DCA on cell viability. These results may have important clinical implications in determining intratumoral metabolism of DCA and, consequently, appropriate oral dosing.


Subject(s)
Chlorides/metabolism , Dichloroacetic Acid/pharmacology , Glutathione Transferase/physiology , Neoplasms/drug therapy , Cell Survival/drug effects , Drug Resistance, Neoplasm , Hep G2 Cells , Humans , MicroRNAs/analysis , Neoplasms/metabolism
9.
Biochem Biophys Res Commun ; 459(3): 463-8, 2015 Apr 10.
Article in English | MEDLINE | ID: mdl-25748576

ABSTRACT

We recently reported that, in a concentration-dependent manner, chloride protects hepatic glutathione transferase zeta 1 from inactivation by dichloroacetate, an investigational drug used in treating various acquired and congenital metabolic diseases. Despite the importance of chloride ions in normal physiology, and decades of study of chloride transport across membranes, the literature lacks information on chloride concentrations in animal tissues other than blood. In this study we measured chloride concentrations in human liver samples from male and female donors aged 1 day to 84 years (n = 97). Because glutathione transferase zeta 1 is present in cytosol and, to a lesser extent, in mitochondria, we measured chloride in these fractions by high-performance liquid chromatography analysis following conversion of the free chloride to pentafluorobenzylchloride. We found that chloride concentration decreased with age in hepatic cytosol but increased in liver mitochondria. In addition, chloride concentrations in cytosol, (105.2 ± 62.4 mM; range: 24.7-365.7 mM) were strikingly higher than those in mitochondria (4.2 ± 3.8 mM; range 0.9-22.2 mM). These results suggest a possible explanation for clinical observations seen in patients treated with dichloroacetate, whereby children metabolize the drug more rapidly than adults following repeated doses, and also provide information that may influence our understanding of normal liver physiology.


Subject(s)
Aging/metabolism , Chlorides/metabolism , Liver/metabolism , Adolescent , Adult , Aged , Aged, 80 and over , Child , Child, Preschool , Chromatography, High Pressure Liquid , Cytosol/metabolism , Dichloroacetic Acid/adverse effects , Dichloroacetic Acid/pharmacokinetics , Dichloroacetic Acid/pharmacology , Enzyme Inhibitors/adverse effects , Enzyme Inhibitors/pharmacokinetics , Enzyme Inhibitors/pharmacology , Female , Glutathione Transferase/antagonists & inhibitors , Glutathione Transferase/metabolism , Humans , Infant , Infant, Newborn , Ion Transport , Liver/drug effects , Male , Metabolic Diseases/drug therapy , Metabolic Diseases/metabolism , Middle Aged , Mitochondria, Liver/metabolism , Young Adult
10.
PLoS One ; 9(6): e99927, 2014.
Article in English | MEDLINE | ID: mdl-24927123

ABSTRACT

While rapamycin and the "rapalogs" Everolimus and Temsirolimus have been approved for clinical use in the treatment of a number of forms of cancer, they have not met overarching success. Some tumors are largely refractory to rapamycin treatment, with some even undergoing an increase in growth rates. However the mechanisms by which this occurs are largely unknown. The results presented here reveal novel cell-signaling mechanisms that may lead to this resistance. The absence of TGFß signaling results in resistance to rapamycin. Additionally, we observed that treatment of some cancer cell lines with rapamycin and its analogs not only potentiates mitogenic signaling and proliferation induced by HGF, but also stimulates the pro-survival kinase Akt. Together, the data show that the effectiveness of rapamycin treatment can be influenced by a number of factors and bring to light potential biomarkers for the prediction of responsiveness to treatment, and suggest combination therapies to optimize rapalog anticancer efficacy.


Subject(s)
Cell Proliferation/drug effects , Cytostatic Agents/pharmacology , Sirolimus/pharmacology , Antibiotics, Antineoplastic/pharmacology , Cell Line, Tumor , Everolimus , Humans , Signal Transduction/drug effects , Sirolimus/analogs & derivatives , Transforming Growth Factor beta/metabolism
11.
Biochemistry ; 52(20): 3489-501, 2013 May 21.
Article in English | MEDLINE | ID: mdl-23627734

ABSTRACT

Previous studies have shown conflicting data regarding cyclin D1/cyclin-dependent kinase 2 (Cdk2) complexes, and considering the widespread overexpression of cyclin D1 in cancer, it is important to fully understand their relevance. While many have shown that cyclin D1 and Cdk2 form active complexes, others have failed to show activity or association. Here, using a novel p21-PCNA fusion protein as well as p21 mutant proteins, we show that p21 is a required scaffolding protein, with cyclin D1 and Cdk2 failing to complex in its absence. These p21/cyclin D1/Cdk2 complexes are active and also bind the trimeric PCNA complex, with each trimer capable of independently binding distinct cyclin/Cdk complexes. We also show that increased p21 levels due to treatment with chemotherapeutic agents result in increased formation and kinase activity of cyclin D1/Cdk2 complexes, and that cyclin D1/Cdk2 complexes are able to phosphorylate a number of substrates in addition to Rb. Nucleophosmin and Cdh1, two proteins important for centrosome replication and implicated in the chromosomal instability of cancer, are shown to be phosphorylated by cyclin D1/Cdk2 complexes. Additionally, polypyrimidine tract binding protein-associated splicing factor (PSF) is identified as a novel Cdk2 substrate, being phosphorylated by Cdk2 complexed with either cyclin E or cyclin D1, and given the many functions of PSF, it could have important implications on cellular activity.


Subject(s)
Cyclin D1/chemistry , Cyclin-Dependent Kinase 2/chemistry , Binding Sites , Cells, Cultured , Cyclin D1/metabolism , Cyclin-Dependent Kinase 2/metabolism , HCT116 Cells , Humans , Multiprotein Complexes/chemistry , Multiprotein Complexes/metabolism , Substrate Specificity
12.
Mol Cancer Ther ; 12(5): 610-20, 2013 May.
Article in English | MEDLINE | ID: mdl-23625935

ABSTRACT

Acetyltransferase p300 (KAT3B) plays key roles in signaling cascades that support cancer cell survival and sustained proliferation. Thus, p300 represents a potential anticancer therapeutic target. To discover novel anticancer agents that target p300, we conducted a high-throughput screening campaign. A library of 622,079 compounds was assayed for cytotoxicity to the triple-negative breast cancer (TNBC) cell line MDA-MB-231 but not to the human mammary epithelial cells. The resulting compounds were tested in a biochemical assay for inhibiting the enzymatic activity of p300. One compound (L002, NSC764414) displayed an IC50 of 1.98 µmol/L against p300 in vitro, inhibited acetylation of histones and p53, and suppressed STAT3 activation in cell-based assays. L002 could be docked to the active site of the p300 catalytic domain. Biochemical tests of a series of related compounds revealed functional groups that may impact inhibitory potency of L002 against p300. Interestingly, these analogs showed inhibitory activities against the cellular paralog of p300 (CBP), p300/CBP-associated factor, and GCN5, but not to other acetyltransferases (KAT5, KAT6B, and KAT7), histone deacetylases, and histone methyltransferases. Among the NCI-60 panel of cancer cell lines, leukemia and lymphoma cell lines were extremely sensitive to L002, whereas it is toxic to only a limited number of cell lines derived from solid tumors. Notably, breast cancer cell lines, especially those derived from TNBC, were highly susceptible to L002. In vivo, it potently suppressed tumor growth and histone acetylation of MDA-MB-468 xenografts. Thus, these new acetyltransferase inhibitors are potential anticancer therapeutics.


Subject(s)
Antineoplastic Agents/pharmacology , Drug Screening Assays, Antitumor , High-Throughput Screening Assays , Small Molecule Libraries , p300-CBP Transcription Factors/antagonists & inhibitors , Acetylation , Animals , Antineoplastic Agents/chemistry , Antineoplastic Agents/metabolism , Antineoplastic Agents/toxicity , Catalytic Domain , Cell Line , Humans , Inhibitory Concentration 50 , Mice , Molecular Docking Simulation , Protein Binding , Protein Interaction Domains and Motifs , Xenograft Model Antitumor Assays , p300-CBP Transcription Factors/chemistry , p300-CBP Transcription Factors/metabolism
13.
J Cell Sci ; 126(Pt 5): 1207-17, 2013 Mar 01.
Article in English | MEDLINE | ID: mdl-23321641

ABSTRACT

The cell has many mechanisms for protecting the integrity of its genome. These mechanisms are often weakened or absent in many cancers, leading to high rates of chromosomal instability in tumors. Control of the cell cycle is crucial for the function of these checkpoints, and is frequently lost in cancers as well. Overexpression of Cyclin D1 in a large number of breast cancers causes overactivation of the cyclin-dependent kinases, including Cdk2. Constitutive Cdk2 activation through Cyclin D1 generates tumors in mice that are aneuploid and have many characteristics indicative of chromosomal instability. Expression of these complexes in the MCF10A cell line leads to retinoblastoma protein (Rb) hyperphosphorylation, a subsequent increase in proliferation rate, and increased expression of the spindle assembly checkpoint protein Mad2. This results in a strengthening of the spindle assembly checkpoint and renders cells more sensitive to the spindle poison paclitaxel. Constitutive Rb phosphorylation also causes a weakening of the p53-dependent tetraploidy checkpoint. Cells with overactive Cdk2 fail to arrest after mitotic slippage in the presence of paclitaxel or cytokinesis failure during treatment with cytochalasin-B, generating 8N populations. This additional increase in DNA content appears to further intensify the tetraploidy checkpoint in a step-wise manner. These polyploid cells are not viable long-term, either failing to undergo division or creating daughter cells that are unable to undergo subsequent division. This study raises intriguing questions about the treatment of tumors with overactive Cdk2.


Subject(s)
Aneuploidy , Cell Cycle Checkpoints/physiology , Cyclin-Dependent Kinase 2/metabolism , Spindle Apparatus/metabolism , Tetraploidy , Cell Cycle Checkpoints/genetics , Cell Line, Tumor , Centrosome/metabolism , Chromosomal Instability/genetics , Cyclin D1/genetics , Cyclin D1/metabolism , Cyclin-Dependent Kinase 2/genetics , Flow Cytometry , Humans , Immunoblotting , Microscopy, Fluorescence , Spindle Apparatus/genetics
14.
Cancer Lett ; 326(2): 183-90, 2012 Dec 30.
Article in English | MEDLINE | ID: mdl-22906417

ABSTRACT

The epithelial to mesenchymal transition (EMT) is a process by which differentiated epithelial cells transition to a mesenchymal phenotype. EMT enables the escape of epithelial cells from the rigid structural constraints of the tissue architecture to a phenotype more amenable to cell migration and, therefore, invasion and metastasis. We characterized an in vivo model of EMT and discovered that marked changes in mitogenic signaling occurred during this process. DNA microarray analysis revealed that the expression of a number of genes varied significantly between post-EMT and pre-EMT breast cancer cells. Post-EMT cancer cells upregulated mRNA encoding c-Met and the PDGF and LPA receptors, and acquired increased responsiveness to HGF, PDGF, and LPA. This rendered the post-EMT cells responsive to the growth inhibitory effects of HGF, PDGF, and LPA receptor inhibitors/antagonists. Furthermore, post-EMT cells exhibited decreased basal Raf and Erk phosphorylation, and in comparison to pre-EMT cells, their proliferation was poorly inhibited by a MEK inhibitor. These studies suggest that therapies need to be designed to target both pre-EMT and post-EMT cancer cells and that signaling changes in post-EMT cells may allow them to take advantage of paracrine signaling from the stroma in vivo.


Subject(s)
Epithelial-Mesenchymal Transition , Mitogens/pharmacology , Animals , Cell Line, Tumor , Humans , Mice , Models, Biological , Oligonucleotide Array Sequence Analysis
15.
Proc Natl Acad Sci U S A ; 108(9): 3743-8, 2011 Mar 01.
Article in English | MEDLINE | ID: mdl-21321191

ABSTRACT

Because our in silico analysis with a human transcription factor database demonstrated the presence of several binding sites for NF-κB, a central regulator of cellular immune and inflammatory responses, in the adeno-associated virus (AAV) genome, we investigated whether AAV uses NF-κB during its life cycle. We used small molecule modulators of NF-κB in HeLa cells transduced with recombinant AAV vectors. VP16, an NF-κB activator, augmented AAV vector-mediated transgene expression up to 25-fold. Of the two NF-κB inhibitors, Bay11, which blocks both the canonical and the alternative NF-κB pathways, totally ablated transgene expression, whereas pyrrolidone dithiocarbamate, which interferes with the classical NF-κB pathway, had no effect. Western blot analyses confirmed the abundance of the nuclear p52 protein component of the alternative NF-κB pathway in the presence of VP16, which was ablated by Bay11, suggesting that AAV transduction activates the alternative NF-κB pathway. In vivo, hepatic AAV gene transfer activated the canonical NF-κB pathway within 2 h, resulting in expression of proinflammatory cytokines and chemokines (likely reflecting the sensing of viral particles by antigen-presenting cells), whereas the alternative pathway was activated by 9 h. Bay11 effectively blocked activation of both pathways without interfering with long-term transgene expression while eliminating proinflammatory cytokine expression. These studies suggest that transient immunosuppression with NF-κB inhibitors before transduction with AAV vectors should lead to a dampened immune response, which has significant implications in the optimal use of AAV vectors in human gene therapy.


Subject(s)
Dependovirus/genetics , Genetic Therapy , Genetic Vectors/genetics , Immunity/genetics , NF-kappa B/metabolism , Signal Transduction , Animals , Antigen-Presenting Cells/drug effects , Antigen-Presenting Cells/metabolism , Antigen-Presenting Cells/virology , Binding Sites , Cytokines/metabolism , Dependovirus/drug effects , Gene Expression/drug effects , HeLa Cells , Humans , Immunity/drug effects , Inflammation Mediators/metabolism , Liver/drug effects , Liver/metabolism , Liver/pathology , Liver/virology , Mice , NF-kappa B/antagonists & inhibitors , Nitriles/pharmacology , Parvoviridae Infections/genetics , Parvoviridae Infections/metabolism , Parvoviridae Infections/virology , Protein Binding/drug effects , Signal Transduction/drug effects , Sulfones/pharmacology , Terminal Repeat Sequences/genetics , Time Factors , Transcription Factors/metabolism , Transgenes
SELECTION OF CITATIONS
SEARCH DETAIL
...