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










Publication year range
1.
Cancer Res Commun ; 4(1): 164-169, 2024 01 19.
Article in English | MEDLINE | ID: mdl-38259096

ABSTRACT

The extent to which non-genetic environmental factors, such as diet, contribute to carcinogenesis has been long debated. One potential mechanism for the effects of environmental factors is through epigenetic modifications that affect gene expression without changing the underlying DNA sequence. However, the functional cooperation between dietary factors and cancer-causing epigenetic regulation is largely unknown. Here, we use a mouse model of age-dependent p16 epimutation, in which the p16 gene activity is directly controlled by promoter DNA methylation. We show p16 epimutation is modulated by folate and cofactors in dietary supplementation, which leads to increased colon cancer risk. Importantly, our findings provide functional evidence concerning the safety of folate fortification in the general population. SIGNIFICANCE: Our study demonstrates that dietary folate and cofactors modulate tumor-suppressor gene methylation to increase intestinal tumorigenesis. Our findings highlight the need for monitoring the long-term safety of folate fortification in high-risk individuals.


Subject(s)
Carcinogenesis , Cyclin-Dependent Kinase Inhibitor p16 , Epigenesis, Genetic , Intestinal Neoplasms , Animals , Humans , Mice , Carcinogenesis/genetics , Cell Transformation, Neoplastic , Diet , Folic Acid , Intestinal Neoplasms/genetics , Cyclin-Dependent Kinase Inhibitor p16/genetics
3.
Bioorg Med Chem ; 65: 116761, 2022 07 01.
Article in English | MEDLINE | ID: mdl-35504208

ABSTRACT

Survivin, a member of the inhibitor of apoptosis protein family, exists as a homodimer and is aberrantly upregulated in a wide spectrum of cancers. It was thought to be an ideal target due to its lack of expression in most adult normal tissues and importance in cancer cell survival. However, it has been challenging to target survivin due to its "undruggable" nature. We previously attempted to target its dimerization domain with a hypothesis that inhibiting survivin dimerization would promote its degradation in proteasome, which led to identification of a lead small-molecule inhibitor, LQZ-7F. LQZ-7F consists of a flat tetracyclic aromatic core with labile hydrazone linking a 1,2,5-oxadiazole moiety. In this study, we tested the hypothesis that LQZ-7F could be developed as a prodrug because the labile hydrazone linker could be hydrolyzed, releasing the tetracyclic aromatic core. To this end, we synthesized the tetracyclic aromatic core (LQZ-7F1) using reported procedure and tested LQZ-7F1 for its biological activities. Here we show that LQZ-7F1 has a significantly improved potency with submicromolar IC50's and induces spontaneous apoptosis in prostate cancer cells. It also more effectively inhibits survivin dimerization and induces survivin degradation in a proteasome-dependent manner than LQZ-7F. We also show that the combination of LQZ-7F1 and docetaxel have strong synergism in inhibiting prostate cancer cell survival. Together, we conclude that the hydrazone linker with the oxadiazole tail is dispensable for survivin inhibition and the survivin dimerization inhibitor, LQZ-7F, may be developed as a prodrug for prostate cancer treatment and to overcome docetaxel resistance.


Subject(s)
Prodrugs , Prostatic Neoplasms , Apoptosis , Cell Line, Tumor , Dimerization , Docetaxel/pharmacology , Docetaxel/therapeutic use , Humans , Hydrazones/pharmacology , Hydrazones/therapeutic use , Inhibitor of Apoptosis Proteins/metabolism , Male , Microtubule-Associated Proteins/metabolism , Oxadiazoles/pharmacology , Oxadiazoles/therapeutic use , Prodrugs/pharmacology , Prostatic Neoplasms/drug therapy , Prostatic Neoplasms/metabolism , Proteasome Endopeptidase Complex/metabolism , Survivin/metabolism
4.
Semin Perinatol ; 45(6): 151455, 2021 10.
Article in English | MEDLINE | ID: mdl-34246487

ABSTRACT

The interaction between the gut and its eventual trillions of microbe inhabitants during microbial colonization, represents a critical time period for establishing the overall health and wellbeing of an individual. The gut microbiome represents a diverse community of microbes that are critical for many physiological roles of the host including host metabolism. These processes are controlled by a fine-tuned chemical cross talk between the host and microbiota. Although the exact mechanisms behind this cross talk remains elusive, microbiota induced epigenetic mechanisms like DNA methylation and histone modifications may be key. This review presents our perspective on the epigenome as a mediator for host-microbiota cross talk, as well as methodology to study epigenetics, the role of dysbiosis in disease, and how the gut microbiome-host axis may be used in personal medicine.


Subject(s)
Gastrointestinal Microbiome , Microbiota , Dysbiosis , Epigenome , Epigenomics , Humans
5.
J Med Chem ; 63(13): 7243-7251, 2020 07 09.
Article in English | MEDLINE | ID: mdl-32421328

ABSTRACT

Survivin, a homodimeric member of the Inhibitor of Apoptosis Protein (IAP) family, is required for cancer cell survival and overexpressed in almost all solid tumors. However, targeting survivin has been challenging due to its "undruggable" nature. Recently, we used a novel approach to target the dimerization interface and identified inhibitors of two scaffolds that can directly bind to and inhibit survivin dimerization. One of the scaffolds, represented by the compound LQZ-7, contains an undesirable labile hydrazone linker and a potentially nonfunctional furazanopyrazine ring that we attempted to eliminate in this study. We found one compound, 7I, that is more active than the parent compound, LQZ-7, and when given orally effectively inhibits xenograft tumor growth and induces survivin loss in tumors. These findings indicate that 7I with a stable linker and a quinoxaline ring can be used as a lead for further optimization of this novel class of survivin inhibitors.


Subject(s)
Antineoplastic Agents/chemical synthesis , Antineoplastic Agents/pharmacology , Survivin/metabolism , Administration, Oral , Animals , Antineoplastic Agents/administration & dosage , Cell Survival/drug effects , Dose-Response Relationship, Drug , Drug Design , Humans , Hydrazones/chemistry , Male , Mice , PC-3 Cells , Proteasome Endopeptidase Complex/metabolism , Protein Multimerization , Survivin/antagonists & inhibitors , Xenograft Model Antitumor Assays
7.
Biotechnol J ; 13(10): e1700748, 2018 Oct.
Article in English | MEDLINE | ID: mdl-29797786

ABSTRACT

Clonally derived cell lines (CDCL) from Chinese Hamster Ovary (CHO) host cell lines, remain the most popular method to manufacture therapeutic proteins. However, CHO cell pools are increasingly being used as an alternate method to produce therapeutic proteins for preclinical drug development in an effort to shorten the time required for new drug development. It is essential that these CHO pools exhibit the desired attributes of CHO CDCLs such as high protein titers and consistent product quality attributes (PQAs). In this study the authors evaluated the Leap-In Transposase®, for the expression of four different proteins (three mAbs and one Bispecific mAb). The resultant pool titers ranges from 2.0 to 5.0 g L-1 for the four proteins compared to 1.5-3.3 g L-1 from the respective control pools (generated by random gene integration). The resultant cell pools are a homogeneously expressing cell population. The average gene copy numbers are similar or lower in the evaluation pools relative to the control pools. The higher titers in the evaluation pools are attributed to higher levels of both IgG-LC and IgG-HC mRNA. In conclusion, the Leap-In transposase generates high titer, homogeneous CHO pools in a short time-period without introducing any undesired PQAs.


Subject(s)
Antibodies, Bispecific , Antibodies, Monoclonal , Cell Culture Techniques , Transposases , Animals , Antibodies, Bispecific/biosynthesis , Antibodies, Monoclonal/biosynthesis , CHO Cells , Cricetulus , Plasmids
8.
Drug Discov Today ; 22(10): 1466-1477, 2017 10.
Article in English | MEDLINE | ID: mdl-28577912

ABSTRACT

Survivin, the smallest member of the inhibitor of apoptosis protein (IAP) family, is overexpressed in cells of almost all cancers but not in most normal tissues in adults. Survivin expression is required for cancer cell survival and knocking down its expression or inhibiting its function using molecular approaches results in spontaneous apoptosis. Thus, survivin is an attractive and perhaps ideal target for cancer drug discovery. However, a US Food and Drug Administration (FDA)-approved drug targeting survivin has yet to emerge. In this Foundation Review, we examine and evaluate various strategies that have been used to target survivin and the stages of each survivin inhibitor to help understand this lack of success. We also provide future perspectives moving forward in targeting survivin for drug discovery.


Subject(s)
Antineoplastic Agents/pharmacology , Antineoplastic Agents/therapeutic use , Inhibitor of Apoptosis Proteins/metabolism , Animals , Apoptosis/drug effects , Drug Discovery/methods , Humans , Neoplasms/drug therapy , Neoplasms/metabolism , United States , United States Food and Drug Administration
9.
Biotechnol Prog ; 33(2): 534-540, 2017 03.
Article in English | MEDLINE | ID: mdl-28188692

ABSTRACT

Chinese hamster ovary (CHO) cells remain the most popular host for the production of biopharmaceutical drugs, particularly monoclonal antibodies (mAbs), bispecific antibodies, and Fc-fusion proteins. Creating and characterizing the stable CHO clonally-derived cell lines (CDCLs) needed to manufacture these therapeutic proteins is a lengthy and laborious process. Therefore, CHO pools have increasingly been used to rapidly produce protein to support and enable preclinical drug development. We recently described the generation of CHO pools yielding mAb titers as high as 7.6 g/L in a 16 day bioprocess using piggyBac transposon-mediated gene integration. In this study, we wanted to understand why the piggyBac pool titers were significantly higher (2-10 fold) than the control CHO pools. Higher titers were the result of a combination of increased average gene copy number, significantly higher messenger RNA levels and the homogeneity (i.e. less diverse population distribution) of the piggyBac pools, relative to the control pools. In order to validate the use of piggyBac pools to support preclinical drug development, we then performed an in-depth product quality analysis of purified protein. The product quality of protein obtained from the piggyBac pools was very similar to the product quality profile of protein obtained from the control pools. Finally, we demonstrated the scalability of these pools from shake flasks to 36L bioreactors. Overall, these results suggest that gram quantities of therapeutic protein can be rapidly obtained from piggyBac CHO pools without significantly changing product quality attributes. © 2017 American Institute of Chemical Engineers Biotechnol. Prog., 33:534-540, 2017.


Subject(s)
Antibodies, Monoclonal/biosynthesis , Bioreactors , Cell Proliferation/physiology , DNA Transposable Elements/genetics , Protein Engineering/methods , Animals , Antibodies, Monoclonal/genetics , Batch Cell Culture Techniques/methods , CHO Cells , Cricetulus , Pilot Projects , Recombinant Proteins/genetics , Recombinant Proteins/metabolism , Up-Regulation
10.
Biotechnol Prog ; 33(2): 469-477, 2017 03.
Article in English | MEDLINE | ID: mdl-27977915

ABSTRACT

IgG bispecific antibodies (BsAbs) represent one of the preferred formats for bispecific antibody therapeutics due to their native-like IgG properties and their monovalent binding to each target. Most reported studies utilized transient expression in HEK293 cells to produce BsAbs. However, the expression of biotherapeutic molecules using stable CHO cell lines is commonly used for biopharmaceutical manufacturing. Unfortunately, limited information is available in the scientific literature on the expression of BsAbs in CHO cell lines. In this study we describe an alternative approach to express the multiple components of IgG BsAbs using a single plasmid vector (quad vector). This single plasmid vector contains both heavy chain genes and both light chain genes required for the expression and assembly of the IgG BsAb, along with a selectable marker. We expressed, purified, and characterized four different IgG BsAbs or "hetero-mAbs" using transient CHO expression and stable CHO minipools. Transient CHO titers ranged from 90 to 160 mg/L. Stable CHO titers ranged from 0.4 to 2.3 g/L. Following a simple Protein A purification step, the percentage of correctly paired BsAbs ranged from 74% to 98% as determined by mass spectrometry. We also found that information generated from transient CHO expression was similar to information generated using stable CHO minipools. In conclusion, the quad vector approach represents a simple, but effective, alternative approach for the generation of IgG BsAbs in both transient CHO and stable CHO expression systems. © 2017 American Institute of Chemical Engineers Biotechnol. Prog., 33:469-477, 2017.


Subject(s)
Antibodies, Monoclonal/biosynthesis , Antibodies, Monoclonal/immunology , Cell Proliferation/physiology , Cloning, Molecular/methods , Immunoglobulin G/immunology , Protein Engineering/methods , Transfection/methods , Animals , Antibodies, Monoclonal/isolation & purification , CHO Cells , Cricetulus , Recombinant Proteins/immunology , Recombinant Proteins/isolation & purification , Recombinant Proteins/metabolism
11.
Biotechnol Prog ; 32(5): 1301-1307, 2016 09.
Article in English | MEDLINE | ID: mdl-27254818

ABSTRACT

Chinese hamster ovary (CHO) cells remain the default production host for many biopharmaceutical drugs, particularly monoclonal antibodies (mAb). Production of gram and kilogram quantities of protein typically requires the generation of stable CHO clones. Unfortunately, this process takes several months, significantly slowing down the drug discovery and development process. Therefore, improved technologies are needed to accelerate biopharmaceutical drug discovery and final drug substance manufacturing. In this study, we describe the generation of stable CHO pools using the piggyBac transposon system. We evaluated the system using four model antibody molecules (3 mAbs and 1 bispecific Ab). Stable CHO pools were isolated in 7-12 days. Using a simple 16-day fed-batch process, we measured titers ranging from 2.3 to 7.6 g/L for the four model antibodies. This represented a 4- to 12-fold increase relative to the controls. Additionally, we isolated stable CHO clones. We found that the stable CHO clones isolated from the piggyBac transposon pools yielded titers two to threefold higher relative to the control clones. Taken together, these results suggest that stable CHO pool and clone generation can be significantly improved by using the piggyBac transposon system. © 2016 American Institute of Chemical Engineers Biotechnol. Prog., 32:1301-1307, 2016.


Subject(s)
Antibodies/analysis , DNA Transposable Elements , Animals , Antibodies/metabolism , CHO Cells , Cells, Cultured , Clone Cells , Cricetulus
12.
Cancer Res ; 76(2): 453-62, 2016 Jan 15.
Article in English | MEDLINE | ID: mdl-26744521

ABSTRACT

Many oncoproteins are considered undruggable because they lack enzymatic activities. In this study, we present a small-molecule-based anticancer agent that acts by inhibiting dimerization of the oncoprotein survivin, thereby promoting its degradation along with spontaneous apoptosis in cancer cells. Through a combination of computational analysis of the dimerization interface and in silico screening, we identified one compound that induced proteasome-dependent survivin degradation. Analysis of a set of structural analogues led us to identify a lead compound (LQZ-7F), which was effective in blocking the survival of multiple cancer cell lines in a low micromolar concentration range. LQZ-7F induced proteasome-dependent survivin degradation, mitotic arrest, and apoptosis, and it blocked the growth of human tumors in mouse xenograft assays. In addition to providing preclinical proof of concept for a survivin-targeting anticancer agent, our work offers novel in silico screening strategies to therapeutically target homodimeric oncogenic proteins considered undruggable.


Subject(s)
Dimerization , Inhibitor of Apoptosis Proteins/genetics , Inhibitor of Apoptosis Proteins/metabolism , Animals , Apoptosis , Cell Line, Tumor , Disease Models, Animal , Humans , Mice , Models, Molecular , Survivin , Xenograft Model Antitumor Assays
13.
PLoS One ; 10(7): e0130796, 2015.
Article in English | MEDLINE | ID: mdl-26177200

ABSTRACT

Phenotypic assays have a proven track record for generating leads that become first-in-class therapies. Whole cell assays that inform on a phenotype or mechanism also possess great potential in drug repositioning studies by illuminating new activities for the existing pharmacopeia. The National Center for Advancing Translational Sciences (NCATS) pharmaceutical collection (NPC) is the largest reported collection of approved small molecule therapeutics that is available for screening in a high-throughput setting. Via a wide-ranging collaborative effort, this library was analyzed in the Open Innovation Drug Discovery (OIDD) phenotypic assay modules publicly offered by Lilly. The results of these tests are publically available online at www.ncats.nih.gov/expertise/preclinical/pd2 and via the PubChem Database (https://pubchem.ncbi.nlm.nih.gov/) (AID 1117321). Phenotypic outcomes for numerous drugs were confirmed, including sulfonylureas as insulin secretagogues and the anti-angiogenesis actions of multikinase inhibitors sorafenib, axitinib and pazopanib. Several novel outcomes were also noted including the Wnt potentiating activities of rotenone and the antifolate class of drugs, and the anti-angiogenic activity of cetaben.


Subject(s)
Drug Repositioning , Cell Line, Tumor , Drug Approval , Drug Evaluation, Preclinical , High-Throughput Screening Assays , Humans , Inhibitory Concentration 50 , Phenotype , Small Molecule Libraries/pharmacology
14.
J Biomol Screen ; 16(6): 588-602, 2011 Jul.
Article in English | MEDLINE | ID: mdl-21521801

ABSTRACT

Phenotypic lead generation strategies seek to identify compounds that modulate complex, physiologically relevant systems, an approach that is complementary to traditional, target-directed strategies. Unlike gene-specific assays, phenotypic assays interrogate multiple molecular targets and signaling pathways in a target "agnostic" fashion, which may reveal novel functions for well-studied proteins and discover new pathways of therapeutic value. Significantly, existing compound libraries may not have sufficient chemical diversity to fully leverage a phenotypic strategy. To address this issue, Eli Lilly and Company launched the Phenotypic Drug Discovery Initiative (PD(2)), a model of open innovation whereby external research groups can submit compounds for testing in a panel of Lilly phenotypic assays. This communication describes the statistical validation, operations, and initial screening results from the first PD(2) assay panel. Analysis of PD(2) submissions indicates that chemical diversity from open source collaborations complements internal sources. Screening results for the first 4691 compounds submitted to PD(2) have confirmed hit rates from 1.6% to 10%, with the majority of active compounds exhibiting acceptable potency and selectivity. Phenotypic lead generation strategies, in conjunction with novel chemical diversity obtained via open-source initiatives such as PD(2), may provide a means to identify compounds that modulate biology by novel mechanisms and expand the innovation potential of drug discovery.


Subject(s)
Drug Discovery , Phenotype , Animals , Apolipoproteins E/metabolism , Cell Cycle/drug effects , Cell Differentiation/drug effects , Cell Line , Drug Evaluation, Preclinical , HeLa Cells , Humans , Insulin/metabolism , Insulin Secretion , Mice , Neovascularization, Physiologic/drug effects , Nocodazole/pharmacology , Osteoblasts/cytology , Osteoblasts/metabolism , Protein Kinase Inhibitors/pharmacology , Rats , Reproducibility of Results , Signal Transduction/drug effects , Tubulin Modulators/pharmacology , Wnt Proteins/metabolism
15.
Cancer Res ; 71(5): 1849-57, 2011 Mar 01.
Article in English | MEDLINE | ID: mdl-21233335

ABSTRACT

Activation of the translation initiation factor 4E (eIF4E) promotes malignant transformation and metastasis. Signaling through the AKT-mTOR pathway activates eIF4E by phosphorylating the inhibitory 4E binding proteins (4E-BP). This liberates eIF4E and allows binding to eIF4G. eIF4E can then be phosphorylated at serine 209 by the MAPK-interacting kinases (Mnk), which also interact with eIF4G. Although dispensable for normal development, Mnk function and eIF4E phosphorylation promote cellular proliferation and survival and are critical for malignant transformation. Accordingly, Mnk inhibition may serve as an attractive cancer therapy. We now report the identification of a potent, selective and orally bioavailable Mnk inhibitor that effectively blocks 4E phosphorylation both in vitro and in vivo. In cultured cancer cell lines, Mnk inhibitor treatment induces apoptosis and suppresses proliferation and soft agar colonization. Importantly, a single, orally administered dose of this Mnk inhibitor substantially suppresses eIF4E phosphorylation for at least 4 hours in human xenograft tumor tissue and mouse liver tissue. Moreover, oral dosing with the Mnk inhibitor significantly suppresses outgrowth of experimental B16 melanoma pulmonary metastases as well as growth of subcutaneous HCT116 colon carcinoma xenograft tumors, without affecting body weight. These findings offer the first description of a novel, orally bioavailable MNK inhibitor and the first preclinical proof-of-concept that MNK inhibition may provide a tractable cancer therapeutic approach.


Subject(s)
Antineoplastic Agents/pharmacology , Benzofurans/pharmacology , Eukaryotic Initiation Factor-4E/metabolism , Intracellular Signaling Peptides and Proteins/antagonists & inhibitors , Lung Neoplasms/metabolism , Protein Serine-Threonine Kinases/antagonists & inhibitors , Animals , Apoptosis/drug effects , Base Sequence , Blotting, Western , Cell Proliferation/drug effects , Female , Humans , Inhibitory Concentration 50 , Intracellular Signaling Peptides and Proteins/genetics , Lung Neoplasms/drug therapy , Lung Neoplasms/pathology , Mice , Mice, Nude , Molecular Sequence Data , Neoplasm Metastasis/drug therapy , Phosphorylation , Polymerase Chain Reaction , Protein Serine-Threonine Kinases/genetics , Xenograft Model Antitumor Assays
16.
FEBS J ; 275(21): 5258-71, 2008 Nov.
Article in English | MEDLINE | ID: mdl-18795946

ABSTRACT

DNA ligases are the enzymes essential for DNA replication, repair and recombination in all organisms. The bacterial DNA ligases involved in DNA replication require NAD(+) for activity, but eukaryotic and viral DNA ligases require ATP. Because of their essential nature, unique structures and widespread existence in nature, bacterial DNA ligases represent a class of valuable targets for identifying novel and selective antibacterial agents. In this study, we cloned and expressed the ligA gene from Streptococcus pneumoniae, and characterized this ligA-encoded NAD(+)-dependent DNA ligase. We then screened small molecule chemical libraries using a biochemical assay and identified a new small molecule with a structure of 2,4-diamino-7-dimethylamino-pyrimido[4,5-d]pyrimidine. We show that this small molecule is a specific inhibitor of bacterial NAD(+)-dependent DNA ligases. Biochemical studies show that this molecule inhibits NAD(+)-dependent DNA ligases, but not ATP-dependent enzymes. The molecule inhibits NAD(+)-dependent DNA ligases competitively with respect to NAD(+) and specifically inhibits enzyme adenylation, but not DNA adenylation or ligation. Labeling studies establish that this molecule inhibits the incorporation of thymidine into DNA and that overexpression of DNA ligase in the cell abolishes this inhibition. Finally, microbiological studies show that this molecule exhibits a broad spectrum of antibacterial activity. Together, this study shows that this small molecule inhibitor identified is specific to bacterial NAD(+)-dependent DNA ligases, exhibits a broad spectrum of antibacterial activities, and has the potential to be developed into an antibacterial agent.


Subject(s)
Anti-Bacterial Agents/chemistry , DNA Ligases/antagonists & inhibitors , Enzyme Inhibitors/pharmacology , Pyrimidines/pharmacology , Streptococcus pneumoniae/enzymology , Bacterial Proteins/antagonists & inhibitors , Base Sequence , DNA Ligases/genetics , DNA Ligases/isolation & purification , Enzyme Inhibitors/chemistry , Humans , Kinetics , Pyrimidines/chemistry , Small Molecule Libraries , Streptococcus pneumoniae/genetics
17.
Anal Biochem ; 360(2): 196-206, 2007 Jan 15.
Article in English | MEDLINE | ID: mdl-17141171

ABSTRACT

Vascular endothelial growth factor (VEGF), a critical regulator in angiogenesis, exerts its angiogenic effect via binding to its receptor, VEGF receptor-2 tyrosine kinase (VEGFR2) or kinase insert domain-containing receptor (Kdr), on the surface of endothelial cells. Kdr-mediated signaling plays an important role in the proliferation, migration, differentiation, and survival of endothelial cells. Therefore, the inhibition of this signaling pathway represents a promising therapeutic approach for the discovery of novel anticancer agents by destabilizing the progression of solid tumors via abrogating tumor-induced angiogenesis. To explore Kdr as an anticancer target and further characterize the enzyme, we purified a cytoplasmic domain of human Kdr (Kdr-CD) and characterized its autophosphorylation activity. We also designed and synthesized peptides containing amino acid sequences corresponding to the autophosphorylation sites of Kdr and developed a simple, robust, high-throughput assay for measuring the phosphate transfer activity of the enzyme. This assay was validated by the experiments showing that the phosphate transfer activity of the purified Kdr-CD required Mg2+ or Mn2+ and preactivation by adenosine 5'-triphosphate (ATP) and was inhibited by known Kdr inhibitors. Using this assay, we examined effects of Mg2+ and Mn2+ on the enzyme activity; optimized the concentrations of Kdr-CD, peptide and ATP substrates, and metal ions in the assay; and determined the kinetic properties of the enzyme for the peptide and ATP as well as IC50 values of two known Kdr inhibitors. Thus, the results of these studies have validated the utilities of this assay for biochemical characterizations of the enzyme and its inhibitors. This approach of designing peptides corresponding to the autophosphorylation sites of Kdr as substrates for the enzyme has general practical implications to other kinases.


Subject(s)
Peptides/metabolism , Phosphates/metabolism , Vascular Endothelial Growth Factor Receptor-2/metabolism , Adenosine Diphosphate/metabolism , Blotting, Western , Chemistry Techniques, Analytical/methods , Electrophoresis, Polyacrylamide Gel , Enzyme Activation/drug effects , Humans , Kinetics , Magnesium/pharmacology , Manganese/pharmacology , Peptides/chemical synthesis , Phosphorylation/drug effects , Protein Binding , Reproducibility of Results , Substrate Specificity , Vascular Endothelial Growth Factor A/metabolism , Vascular Endothelial Growth Factor Receptor-2/antagonists & inhibitors
18.
J Bacteriol ; 188(13): 4737-48, 2006 Jul.
Article in English | MEDLINE | ID: mdl-16788183

ABSTRACT

Acyl carrier protein (ACP) synthase (AcpS) catalyzes the transfer of the 4'-phosphopantetheine moiety from coenzyme A (CoA) onto a serine residue of apo-ACP, resulting in the conversion of apo-ACP to the functional holo-ACP. The holo form of bacterial ACP plays an essential role in mediating the transfer of acyl fatty acid intermediates during the biosynthesis of fatty acids and phospholipids. AcpS is therefore an attractive target for therapeutic intervention. In this study, we have purified and characterized the AcpS enzymes from Escherichia coli, Streptococcus pneumoniae, and Mycoplasma pneumoniae, which exemplify gram-negative, gram-positive, and atypical bacteria, respectively. Our gel filtration column chromatography and cross-linking studies demonstrate that the AcpS enzyme from M. pneumoniae, like E. coli enzyme, exhibits a homodimeric structure, but the enzyme from S. pneumoniae exhibits a trimeric structure. Our biochemical studies show that the AcpS enzymes from M. pneumoniae and S. pneumoniae can utilize both short- and long-chain acyl CoA derivatives but prefer long-chain CoA derivatives as substrates. On the other hand, the AcpS enzyme from E. coli can utilize short-chain CoA derivatives but not the long-chain CoA derivatives tested. Finally, our biochemical studies show that M. pneumoniae AcpS is kinetically a very sluggish enzyme compared with those from E. coli and S. pneumoniae. Together, the results of these studies show that the AcpS enzymes from different bacterial species exhibit different native structures and substrate specificities with regard to the utilization of CoA and its derivatives. These findings suggest that AcpS from different microorganisms plays a different role in cellular physiology.


Subject(s)
Acyl Coenzyme A/metabolism , Bacterial Proteins/metabolism , Escherichia coli/enzymology , Transferases (Other Substituted Phosphate Groups)/physiology , Acyl Coenzyme A/chemistry , Bacterial Proteins/chemistry , Mycoplasma pneumoniae , Species Specificity , Streptococcus pneumoniae , Substrate Specificity , Transferases (Other Substituted Phosphate Groups)/chemistry , Transferases (Other Substituted Phosphate Groups)/metabolism
19.
Microbiology (Reading) ; 146 ( Pt 5): 1071-1083, 2000 May.
Article in English | MEDLINE | ID: mdl-10832634

ABSTRACT

Era, an essential GTPase, appears to play an important role in the regulation of the cell cycle and protein synthesis of bacteria and mycoplasmas. In this study, native Era, His-tagged Era (His-Era) and glutathione S-transferase (GST)-fusion Era (GST-Era) proteins from Escherichia coli were expressed and purified. It was shown that the GST-Era and His-Era proteins purified by 1-step affinity column chromatographic methods were associated with RNA and exhibited a higher GTPase activity. However, the native Era protein purified by a 3-step column chromatographic method had a much lower GTPase activity and was not associated with RNA which had been removed during purification. Purified GST-Era protein was shown to be present as a high- and a low-molecular-mass forms. The high-molecular-mass form of GST-Era was associated with RNA and exhibited a much higher GTPase activity. Removal of the RNA associated with GST-Era resulted in a significant reduction in the GTPase activity. The RNA associated with GST-Era was shown to be primarily 16S rRNA. A purified native Era protein preparation, when mixed with total cellular RNA, was found to bind to some of the RNA. The native Era protein isolated directly from the cells of a wild-type E. coli strain was also present as a high-molecular-mass form complexed with RNA and RNase treatment converted the high-molecular-mass form into a 32 kDa low-molecular-mass form, a monomer of Era. Furthermore, a C-terminally truncated Era protein, when expressed in E. coli, did not bind RNA. Finally, the GTPase activity of the Era protein free of RNA, but not the Era protein associated with the RNA, was stimulated by acetate and 3-phosphoglycerate. These carbohydrates, however, failed to activate the GTPase activity of the C-terminally truncated Era protein. Thus, the results of this study establish that the C-terminus of Era is essential for the RNA-binding activity and that the RNA and carbohydrates modulate the GTPase activity of Era possibly through a similar mechanism.


Subject(s)
Bacterial Proteins/metabolism , Escherichia coli Proteins , Escherichia coli/metabolism , GTP-Binding Proteins/metabolism , Glutathione Transferase/metabolism , RNA, Ribosomal, 16S/metabolism , RNA-Binding Proteins , Acetates/pharmacology , Bacterial Proteins/chemistry , Bacterial Proteins/genetics , Blotting, Western , Carbohydrates/pharmacology , Chromatography, Affinity , Chromatography, Gel , Chromatography, Ion Exchange , Electrophoresis, Polyacrylamide Gel , Escherichia coli/genetics , GTP-Binding Proteins/chemistry , GTP-Binding Proteins/genetics , Glutathione Transferase/chemistry , Glutathione Transferase/isolation & purification , Glyceric Acids/pharmacology , Molecular Weight , Polymerase Chain Reaction , Protein Binding , RNA, Bacterial/analysis , RNA, Bacterial/metabolism , RNA, Bacterial/pharmacology , RNA, Ribosomal, 16S/analysis , RNA, Ribosomal, 16S/pharmacology , Recombinant Proteins/metabolism
20.
Microbiology (Reading) ; 145 ( Pt 4): 791-800, 1999 Apr.
Article in English | MEDLINE | ID: mdl-10220158

ABSTRACT

Era, an essential GTPase, is present in many bacteria and Mycoplasma spp. and appears to play a major role in the cell cycle and other cellular processes. To further understand its function, an era gene from Streptococcus pneumoniae was identified and cloned, and a mutant era gene with a deletion of 68 codons from its 3'-terminus was constructed. The truncated Era protein was then purified and characterized, and the ability of the truncated era gene to complement an Escherichia coli mutant strain defective in Era production was examined. Like the full-length Era protein, the truncated Era protein was able to bind and hydrolyse GTP, but its binding activity was increased twofold and its hydrolytic activity was reduced sevenfold when compared with those of the full-length Era protein. Unlike the full-length Era protein, the truncated Era protein lost its ability to bind to the E. coli cytoplasmic membrane. The full-length era gene was able to complement the E. coli mutant deficient in Era production when carried on pACYC184, while the truncated era gene failed to do so when carried on pACYC184, pBR322 or pUC18. The cellular amounts of the truncated Era and the full-length Era proteins in E. coli and S. pneumoniae, respectively, were then determined by Western blot analysis. In addition, the minimal amount of the S. pneumoniae Era protein needed for complementation of the E. coli mutant was also measured. Taken together, these results suggest that the C-terminus of the Era protein might be responsible for the binding of the protein to the cytoplasmic membrane and be essential for function.


Subject(s)
Escherichia coli Proteins , GTP Phosphohydrolases/genetics , GTP Phosphohydrolases/metabolism , GTP-Binding Proteins/genetics , GTP-Binding Proteins/metabolism , RNA-Binding Proteins , Streptococcus pneumoniae/enzymology , Streptococcus pneumoniae/genetics , Blotting, Western , Cloning, Molecular , Escherichia coli/genetics , Escherichia coli/metabolism , GTP Phosphohydrolases/chemistry , GTP Phosphohydrolases/isolation & purification , GTP-Binding Proteins/chemistry , GTP-Binding Proteins/isolation & purification , Genes, Bacterial , Genetic Complementation Test , Guanosine Triphosphate/metabolism , Hydrolysis , Molecular Sequence Data , Recombinant Proteins/chemistry , Recombinant Proteins/isolation & purification , Recombinant Proteins/metabolism , Streptococcus pneumoniae/growth & development
SELECTION OF CITATIONS
SEARCH DETAIL
...