Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 12 de 12
Filter
Add more filters










Publication year range
1.
Nucleic Acid Ther ; 22(3): 205-10, 2012 Jun.
Article in English | MEDLINE | ID: mdl-22519815

ABSTRACT

The immune stimulation induced by short interfering RNAs (siRNAs) has been reported to be quieted or abrogated by methoxy or fluoro modifications of the 2' position of the ribose sugar. However, variables such as the type of modification, nucleotide preference, and strand bias have not been systematically evaluated. Here, we report the results of a screen of several modified siRNAs via a human peripheral blood monocyte cytokine induction assay. Unlike corresponding modifications of guanosine, cytidine, or uridine, 2'-fluoro modification of adenosine significantly reduced cytokine induction while retaining siRNA knockdown activity. The results of this study suggest adenosine as an optimal target for modification.


Subject(s)
Adenosine/chemistry , RNA, Small Interfering/immunology , Cytokines/biosynthesis , Cytokines/blood , Enzyme-Linked Immunosorbent Assay , Humans , RNA, Small Interfering/chemistry , beta-Galactosidase/metabolism
2.
J Am Chem Soc ; 133(24): 9200-3, 2011 Jun 22.
Article in English | MEDLINE | ID: mdl-21612237

ABSTRACT

Immune stimulation is a significant hurdle in the development of effective and safe RNA interference therapeutics. Here, we address this problem in the context of a mimic of microRNA-122 by employing novel nucleobase and known 2'-ribose modifications. The nucleobase modifications are analogues of adenosine and guanosine that contain cyclopentyl and propyl minor-groove projections. Via a site-by-site chemical modification analysis, we identify several immunostimulatory 'hot spots' within the miRNA guide strand at which single base modifications significantly reduce immune stimulation. A duplex containing one base modification on each strand proved to be most effective in preventing immune stimulation.


Subject(s)
Biomimetic Materials/adverse effects , Biomimetic Materials/chemistry , Immune System/drug effects , MicroRNAs/genetics , RNA, Double-Stranded/adverse effects , RNA, Double-Stranded/chemistry , Ribose/chemistry , Animals , Base Sequence , Biomimetic Materials/chemical synthesis , Cell Line, Tumor , Cytokines/metabolism , Gene Knockdown Techniques , Immune System/metabolism , Mice , RNA, Double-Stranded/chemical synthesis , RNA, Double-Stranded/genetics
3.
Cancer Chemother Pharmacol ; 65(4): 707-17, 2010 Mar.
Article in English | MEDLINE | ID: mdl-19649632

ABSTRACT

PURPOSE: The Aurora family of serine/threonine kinases (Aurora-A, Aurora-B, and Aurora-C) plays a key role in cells orderly progression through mitosis. Elevated expression levels of Aurora kinases have been detected in a high percentage of melanoma, colon, breast, ovarian, gastric, and pancreatic tumors. We characterized the biological and pharmacological properties of SNS-314, an ATP-competitive, selective, and potent inhibitor of Aurora kinases. METHODS: We studied the biochemical potency and selectivity of SNS-314 to inhibit Aurora kinases A, B, and C. The inhibition of cellular proliferation induced by SNS-314 was evaluated in a broad range of tumor cell lines and correlated to inhibition of histone H3 phosphorylation, inhibition of cell-cycle progression, increase in nuclear content and cell size, loss of viability, and induction of apoptosis. The dose and administration schedule of SNS-314 was optimized for in vivo efficacy in mouse xenograft models of human cancer. RESULTS: In the HCT116 human colon cancer xenograft model, administration of 50 and 100 mg/kg SNS-314 led to dose-dependent inhibition of histone H3 phosphorylation for at least 10 h, indicating effective Aurora-B inhibition in vivo. HCT116 tumors from animals treated with SNS-314 showed potent and sustained responses including reduction of phosphorylated histone H3 levels, increased caspase-3 and appearance of increased nuclear size. The compound showed significant tumor growth inhibition in a dose-dependent manner under a variety of dosing schedules including weekly, bi-weekly, and 5 days on/9 days off. CONCLUSIONS: SNS-314 is a potent small-molecule inhibitor of Aurora kinases developed as a novel anti-cancer therapeutic agent for the treatment of diverse human malignancies.


Subject(s)
Antineoplastic Agents/pharmacology , Neoplasms/prevention & control , Phenylurea Compounds/pharmacology , Protein Serine-Threonine Kinases/antagonists & inhibitors , Thiazoles/pharmacology , Animals , Antineoplastic Agents/chemistry , Apoptosis/drug effects , Aurora Kinase A , Aurora Kinase B , Aurora Kinase C , Aurora Kinases , Caspase 3/metabolism , Cell Cycle/drug effects , Cell Proliferation/drug effects , Cell Survival/drug effects , Dose-Response Relationship, Drug , Enzyme Inhibitors/chemistry , Enzyme Inhibitors/pharmacology , Female , HCT116 Cells , HT29 Cells , HeLa Cells , Histones/metabolism , Humans , Inhibitory Concentration 50 , Mice , Mice, Nude , Molecular Structure , Neoplasms/metabolism , Neoplasms/pathology , Phenylurea Compounds/chemistry , Phosphorylation/drug effects , Protein Serine-Threonine Kinases/metabolism , Thiazoles/chemistry , Tumor Burden/drug effects , Xenograft Model Antitumor Assays
4.
Mol Cancer Ther ; 8(4): 930-9, 2009 Apr.
Article in English | MEDLINE | ID: mdl-19372566

ABSTRACT

Aurora kinases play key roles in regulating centrosome maturation, mitotic spindle formation, and cytokinesis during cell division, and are considered promising drug targets due to their frequent overexpression in a variety of human cancers. SNS-314 is a selective and potent pan Aurora inhibitor currently in a dose escalation phase 1 clinical trial for the treatment of patients with advanced solid tumors. Here, we report the antiproliferative effects of SNS-314 in combination with common chemotherapeutics in cell culture and xenograft models. The HCT116 colorectal carcinoma cell line, with intact or depleted p53 protein levels, was treated with SNS-314 and a cytotoxic chemotherapeutic from a panel comprised of gemcitabine, 5-fluorouracil (5-FU), carboplatin, daunomycin, SN-38 (the active metabolite of irinotecan), docetaxel, and vincristine. Combinations were administered under either concurrent or sequential schedules. SNS-314 has predominantly additive effects when administered concurrently with commonly used anticancer agents. Sequential administration of SNS-314 with chemotherapeutic compounds showed additive antiproliferative effects with carboplatin, gemcitabine, 5-FU, daunomycin, and SN-38, and synergy was observed in combination with gemcitabine, docetaxel, or vincristine. The most profound antiproliferative effects were observed with sequential administration of SNS-314 followed by docetaxel or vincristine. In vivo, SNS-314 potentiated the antitumor activity of docetaxel in xenografts. Both the in vitro synergies observed between SNS-314 and agents that target the mitotic spindle and the potentiation seen with docetaxel in vivo are consistent with a mechanism of action in which Aurora inhibition bypasses the mitotic spindle assembly checkpoint and prevents cytokinesis, augmenting subsequent spindle toxin-mediated mitotic catastrophe and cell death.


Subject(s)
Antineoplastic Agents/pharmacology , Colonic Neoplasms/drug therapy , Disease Models, Animal , Enzyme Inhibitors/pharmacology , Microtubules/drug effects , Phenylurea Compounds/pharmacology , Protein Serine-Threonine Kinases/antagonists & inhibitors , Thiazoles/pharmacology , Animals , Aurora Kinases , Cell Proliferation/drug effects , Cell Survival/drug effects , Drug Synergism , Drug Therapy, Combination , Female , Humans , Mice , Spindle Apparatus/drug effects , Xenograft Model Antitumor Assays
5.
Biochemistry ; 48(21): 4488-96, 2009 Jun 02.
Article in English | MEDLINE | ID: mdl-19284778

ABSTRACT

BACE-1 (beta-site amyloid precursor protein cleaving enzyme), a prominent target in Alzheimer's disease drug discovery efforts, was surveyed using Tethering technology to discover small molecule fragment ligands that bind to the enzyme active site. Screens of a library of >15000 thiol-containing fragments versus a panel of BACE-1 active site cysteine mutants under redox-controlled conditions revealed several novel amine-containing fragments that could be selectively captured by subsets of the tethering sites. For one such hit class, defined by a central aminobenzylpiperidine (ABP) moiety, X-ray crystal structures of BACE mutant-disulfide conjugates revealed that the fragment bound by engaging both catalytic aspartates with hydrogen bonds. The affinities of ABP fragments were improved by structure-guided chemistry, first for conjugation as thiol-containing fragments and then for stand-alone, noncovalent inhibition of wild-type (WT) BACE-1 activity. Crystallography confirmed that the inhibitors bound in exactly the same mode as the disulfide-conjugated fragments that were originally selected from the screen. The ABP ligands represent a new type of nonpeptidic BACE-1 inhibitor motif that has not been described in the aspartyl protease literature and may serve as a starting point for the development of BACE-1-directed Alzheimer's disease therapeutics.


Subject(s)
Amyloid Precursor Protein Secretases/antagonists & inhibitors , Drug Discovery/methods , Enzyme Inhibitors/chemistry , Enzyme Inhibitors/pharmacology , Amyloid Precursor Protein Secretases/chemistry , Amyloid Precursor Protein Secretases/genetics , Amyloid Precursor Protein Secretases/metabolism , Biocatalysis , Catalytic Domain , Cysteine , Drug Evaluation, Preclinical , Enzyme Inhibitors/metabolism , Humans , Ligands , Models, Molecular , Molecular Conformation , Mutation , Peptides/chemistry , Piperidines/chemistry , Piperidines/metabolism , Structure-Activity Relationship
6.
Bioorg Med Chem Lett ; 18(20): 5648-52, 2008 Oct 15.
Article in English | MEDLINE | ID: mdl-18793847

ABSTRACT

A series of 2-amino-pyrazolopyridines was designed and synthesized as Polo-like kinase (Plk) inhibitors based on a low micromolar hit. The SAR was developed to provide compounds exhibiting low nanomolar inhibitory activity of Plk1; the phenotype of treated cells is consistent with Plk1 inhibition. A co-crystal structure of one of these compounds with zPlk1 confirms an ATP-competitive binding mode.


Subject(s)
Cell Cycle Proteins/antagonists & inhibitors , Chemistry, Pharmaceutical/methods , Protein Serine-Threonine Kinases/antagonists & inhibitors , Proto-Oncogene Proteins/antagonists & inhibitors , Pyrazoles/chemical synthesis , Pyridines/chemical synthesis , Adenosine Triphosphate/chemistry , Amino Acid Motifs , Cell Cycle , Crystallography, X-Ray , Drug Design , Humans , Inhibitory Concentration 50 , Models, Chemical , Molecular Conformation , Phenotype , Pyrazoles/chemistry , Pyrazoles/pharmacology , Pyridines/chemistry , Pyridines/pharmacology , Structure-Activity Relationship , Polo-Like Kinase 1
8.
Acta Crystallogr D Biol Crystallogr ; 64(Pt 9): 909-18, 2008 Sep.
Article in English | MEDLINE | ID: mdl-18703838

ABSTRACT

Polo-like kinase 1 (Plk1) is a member of a family of serine/threonine kinases involved in the regulation of cell-cycle progression and cytokinesis and is an attractive target for the development of anticancer therapeutics. A zebrafish homolog of the human Plk1 (hPlk1) kinase domain (KD) was identified that can be expressed in large quantities in bacteria and crystallizes readily, whether in a wild-type form or as a variant containing the activating Thr196-->Asp substitution, in one space group and under similar conditions both in the absence and presence of active-site compounds. This construct was validated by testing a panel of hPlk1 inhibitors against human and zebrafish proteins and it was shown that the selected small molecules inhibited the homologs with a high degree of correlation. Crystal structures of ligand-free wild-type and activated zebrafish Plk1 (zPlk1) KDs revealed the organization of the secondary structural elements around the active site and demonstrated that the activation segment was disordered in the activated form of the domain but possessed a well defined secondary structure in the wild-type enzyme. The cocrystal structure of wild-type zPlk1 KD with ADP documented the hydrolysis of ATP and revealed the phosphorylation site. The cocrystal structure of the activated KD with wortmannin, a covalent inhibitor of Plk1 and PI3 kinases, showed the binding mode of the small molecule to the enzyme and may facilitate the design of more potent Plk1 inhibitors. The work presented in this study establishes the zPlk1 KD as a useful tool for rapid low- and high-throughput structure-based screening and drug discovery of compounds specific for this mitotic target.


Subject(s)
Catalytic Domain , Cell Cycle Proteins/chemistry , Protein Serine-Threonine Kinases/chemistry , Proto-Oncogene Proteins/chemistry , Zebrafish Proteins/chemistry , Adenosine Diphosphate/chemistry , Amino Acid Sequence , Amino Acid Substitution , Androstadienes/chemistry , Animals , Binding Sites , Cell Cycle Proteins/antagonists & inhibitors , Cell Cycle Proteins/genetics , Crystallization , Crystallography, X-Ray , Humans , Kinetics , Ligands , Models, Molecular , Protein Conformation , Protein Serine-Threonine Kinases/antagonists & inhibitors , Protein Serine-Threonine Kinases/genetics , Proto-Oncogene Proteins/antagonists & inhibitors , Proto-Oncogene Proteins/genetics , Wortmannin , Zebrafish , Polo-Like Kinase 1
9.
Article in English | MEDLINE | ID: mdl-18678933

ABSTRACT

Polo-like kinase 1 (Plk1) is a member of the Polo-like kinase family of serine/threonine kinases involved in the regulation of cell-cycle progression and cytokinesis and is an attractive target for the development of anticancer therapeutics. The catalytic domain of this enzyme shares significant primary amino-acid homology and structural similarity with another mitotic kinase, Aurora A. While screening an Aurora A library of ATP-competitive compounds, a urea-containing inhibitor with low affinity for mouse Aurora A but with submicromolar potency for human and zebrafish Plk1 (hPlk1 and zPlk1, respectively) was identified. A crystal structure of the zebrafish Plk1 kinase domain-inhibitor complex reveals that the small molecule occupies the purine pocket and extends past the catalytic lysine into the adaptive region of the active site. Analysis of the structures of this protein-inhibitor complex and of similar small molecules cocrystallized with other kinases facilitates understanding of the specificity of the inhibitor for Plk1 and documents for the first time that Plk1 can accommodate extended ATP-competitive compounds that project toward the adaptive pocket and help the enzyme order its activation segment.


Subject(s)
Cell Cycle Proteins/chemistry , Protein Serine-Threonine Kinases/chemistry , Proto-Oncogene Proteins/chemistry , Zebrafish Proteins/chemistry , Zebrafish/metabolism , Animals , Base Sequence , Catalytic Domain , Cell Cycle Proteins/antagonists & inhibitors , Cell Cycle Proteins/genetics , Crystallography, X-Ray , DNA Primers , Models, Molecular , Mutagenesis, Site-Directed , Protein Conformation , Protein Serine-Threonine Kinases/antagonists & inhibitors , Protein Serine-Threonine Kinases/genetics , Proto-Oncogene Proteins/antagonists & inhibitors , Proto-Oncogene Proteins/genetics , Substrate Specificity , Zebrafish Proteins/antagonists & inhibitors , Zebrafish Proteins/genetics , Polo-Like Kinase 1
10.
J Med Chem ; 49(3): 839-42, 2006 Feb 09.
Article in English | MEDLINE | ID: mdl-16451048

ABSTRACT

A series of novel beta-site amyloid precursor protein cleaving enzyme (BACE-1) inhibitors containing an aminoethylene (AE) tetrahedral intermediate isostere were synthesized and evaluated in comparison to corresponding hydroxyethylene (HE) compounds. Enzymatic inhibitory values were similar for both isosteres, as were structure-activity relationships with respect to stereochemical preference and substituent variation (P2/P3, P1, and P2'); however, the AE compounds were markedly more potent in a cell-based assay for reduction of beta-secretase activity. The incorporation of preferred P2/P3, P1, and P2' substituents into the AE pharmacophore yielded compound 7, which possessed enzymatic and cell assay IC(50)s of 26 nM and 180 nM, respectively. A three-dimensional crystal structure of 7 in complex with BACE-1 revealed that the amino group of the inhibitor core engages the catalytic aspartates in a manner analogous to hydroxyl groups in HE inhibitors. The AE isostere class represents a promising advance in the development of BACE-1 inhibitors.


Subject(s)
Endopeptidases/chemistry , Ethylamines/chemical synthesis , Protease Inhibitors/chemical synthesis , Amyloid Precursor Protein Secretases , Aspartic Acid Endopeptidases , Binding Sites , Cell Line , Crystallography, X-Ray , Dipeptides/chemical synthesis , Dipeptides/chemistry , Dipeptides/pharmacology , Ethylamines/chemistry , Ethylamines/pharmacology , Humans , Models, Molecular , Molecular Structure , Protease Inhibitors/chemistry , Protease Inhibitors/pharmacology , Structure-Activity Relationship
11.
J Cell Biol ; 169(3): 383-9, 2005 May 09.
Article in English | MEDLINE | ID: mdl-15866890

ABSTRACT

Cytoskeletal dynamics at the Golgi apparatus are regulated in part through a binding interaction between the Golgi-vesicle coat protein, coatomer, and the regulatory GTP-binding protein Cdc42 (Wu, W.J., J.W. Erickson, R. Lin, and R.A. Cerione. 2000. Nature. 405:800-804; Fucini, R.V., J.L. Chen, C. Sharma, M.M. Kessels, and M. Stamnes. 2002. Mol. Biol. Cell. 13:621-631). The precise role of this complex has not been determined. We have analyzed the protein composition of Golgi-derived coat protomer I (COPI)-coated vesicles after activating or inhibiting signaling through coatomer-bound Cdc42. We show that Cdc42 has profound effects on the recruitment of dynein to COPI vesicles. Cdc42, when bound to coatomer, inhibits dynein binding to COPI vesicles whereas preventing the coatomer-Cdc42 interaction stimulates dynein binding. Dynein recruitment was found to involve actin dynamics and dynactin. Reclustering of nocodazole-dispersed Golgi stacks and microtubule/dynein-dependent ER-to-Golgi transport are both sensitive to disrupting Cdc42 mediated signaling. By contrast, dynein-independent transport to the Golgi complex is insensitive to mutant Cdc42. We propose a model for how proper temporal regulation of motor-based vesicle translocation could be coupled to the completion of vesicle formation.


Subject(s)
COP-Coated Vesicles/metabolism , Coatomer Protein/metabolism , Dyneins/metabolism , cdc42 GTP-Binding Protein/metabolism , Actins/metabolism , Animals , COP-Coated Vesicles/ultrastructure , Cattle , Chlorocebus aethiops , Dynactin Complex , Endoplasmic Reticulum/metabolism , Endoplasmic Reticulum/ultrastructure , Feedback, Physiological/physiology , Golgi Apparatus/metabolism , Golgi Apparatus/ultrastructure , Microscopy, Electron, Transmission , Microtubule-Associated Proteins/metabolism , Microtubules/metabolism , Microtubules/ultrastructure , Mutation/physiology , Protein Binding/physiology , Protein Transport/physiology , Rats , Subcellular Fractions , Vero Cells , cdc42 GTP-Binding Protein/genetics
12.
Mol Biol Cell ; 13(2): 621-31, 2002 Feb.
Article in English | MEDLINE | ID: mdl-11854417

ABSTRACT

Recent studies indicate that regulation of the actin cytoskeleton is important for protein trafficking, but its precise role is unclear. We have characterized the ARF1-dependent assembly of actin on the Golgi apparatus. Actin recruitment involves Cdc42/Rac and requires the activation of the Arp2/3 complex. Although the actin-binding proteins mAbp1 (SH3p7) and drebrin share sequence homology, they are differentially segregated into two distinct ARF-dependent actin complexes. The binding of Cdc42 and mAbp1, which localize to the Golgi apparatus, but not drebrin, is blocked by occupation of the p23 cargo-protein-binding site on coatomer. Exogenously expressed mAbp1 is mislocalized and inhibits Golgi transport in whole cells. The ability of ARF, vesicle-coat proteins, and cargo to direct the assembly of cytoskeletal structures helps explain how only a handful of vesicle types can mediate the numerous trafficking steps in the cell.


Subject(s)
Actins/physiology , Golgi Apparatus/physiology , Membrane Proteins/physiology , Microfilament Proteins/physiology , src Homology Domains , ADP-Ribosylation Factor 1/physiology , Animals , Cattle , Cytoskeleton/physiology , Macromolecular Substances , Rats , Secretory Vesicles/physiology , cdc42 GTP-Binding Protein/physiology , rho GTP-Binding Proteins/physiology
SELECTION OF CITATIONS
SEARCH DETAIL
...