Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 41
Filter
Add more filters










Publication year range
1.
Oncotarget ; 7(35): 56944-56957, 2016 08 30.
Article in English | MEDLINE | ID: mdl-27486970

ABSTRACT

Hepatocellular carcinoma (HCC) is one of the most common cancers and a leading cause of cancer mortality. Prognosis of this disease largely depends on its stage. An Enlarged liver, due to dysplasia, may be a critical point in the multi-step progression to HCC. The mechanism underlying hepatomegaly in human and mouse models are poorly understood. We previously reported we observed enlarged liver in hepatitis B virus X protein (HBx) expressing mice (HBx mice). Here we identify the critical role of HBx induced IGF-II in hepatomegaly in mice and abnormal cell growth in human hepatoma cells. We found that HBx induced IGF-II is essential to induce epithelial-mesenchymal transition (EMT) through loss of E-cadherin. In mouse liver, loss of E-cadherin was mediated by post-translational regulation, at least in part, by protease and SUMOylation not by transcriptional regulation. In contrast, in hepatoma cell line (HepG2 cells) Akt signal pathway controls the mRNA expression level of EMT-related transcription factors, especially Twist, in addition to post- translational modification through SUMOylation. Thus, IGF-II-mediated loss of E-cadherin is central in developing hepatomegaly in mice and abnormal cell growth in the hepatoma cell line. HBx induced IGF-II represents a potential biomarker, which is also a therapeutic target in HCC.


Subject(s)
Cadherins/metabolism , Gene Expression Regulation, Neoplastic , Insulin-Like Growth Factor II/metabolism , SUMO-1 Protein/metabolism , Trans-Activators/metabolism , Animals , Antigens, CD , Biomarkers, Tumor/metabolism , Carcinoma, Hepatocellular/metabolism , Cdh1 Proteins/metabolism , Epithelial-Mesenchymal Transition , Hep G2 Cells , Hepatocytes/metabolism , Hepatomegaly/metabolism , Homozygote , Humans , Liver/metabolism , Mice , Mice, Inbred C57BL , Mice, Transgenic , Protein Processing, Post-Translational , Viral Regulatory and Accessory Proteins
2.
Anticancer Res ; 35(6): 3235-43, 2015 Jun.
Article in English | MEDLINE | ID: mdl-26026083

ABSTRACT

BACKGROUND/AIM: The current study investigated the mechanisms underlying the antitumor activity of SB265610, a cysteine-amino acid-cysteine (CXC) chemokines receptor 2 (CXCR2) antagonist. MATERIALS AND METHODS: Cell-cycle progression and regulatory molecules were assessed by flow cytometry, immunoblotting, real-time PCR and immunoprecipitation. Target validation was achieved via RNA interference. RESULTS: G1 arrest induced by SB265610 occurred at concentrations lacking CXCR2 selectivity, persisted upon interleukin 8 (IL8) challenge, and did not affect IL8 downstream target expression. Profiling of G1 regulators revealed cyclin-dependent kinase 2 (CDK2) (Thr160) hypophosphorylation, cyclin D3 gene down-regulation, and p21 post-translational induction. However, only cyclin D3 and CDK2 contributed towards G1 arrest. Furthermore, SB265610 induced a sustained phosphorylation of the p38MAPK. Pharmacological interference with p38MAPK significantly abrogated SB265610-induced G1 arrest and normalized the expression of cyclin D3, with restoration of its exclusive binding to CDK6, but with weak recovery of CDK2 (Thr160) hypo-phosphorylation. CONCLUSION: The present study described the mechanisms for the anti-proliferative activity of SB265610 which may be of value in IL8-rich tumor microenvironments.


Subject(s)
Cyclin D3/biosynthesis , Cyclin-Dependent Kinase 2/biosynthesis , Phenylurea Compounds/administration & dosage , Prostatic Neoplasms/genetics , Receptors, Interleukin-8B/biosynthesis , Triazoles/administration & dosage , Cell Cycle Checkpoints/drug effects , Cyclin D3/genetics , Cyclin-Dependent Kinase 2/genetics , Cyclin-Dependent Kinase Inhibitor p21/genetics , G1 Phase/drug effects , Gene Expression Regulation, Neoplastic/drug effects , Humans , Interleukin-8/genetics , Male , Phosphorylation , Prostatic Neoplasms/drug therapy , Prostatic Neoplasms/pathology , Receptors, Interleukin-8B/genetics , Tumor Microenvironment/drug effects , p38 Mitogen-Activated Protein Kinases/genetics
3.
Chem Biol Drug Des ; 85(5): 638-44, 2015 May.
Article in English | MEDLINE | ID: mdl-25323822

ABSTRACT

In this study, we have synthesized novel water soluble derivatives of natural compound aloe emodin 4(a-j) by coupling with various amino acid esters and substituted aromatic amines, in an attempt to improve the anticancer activity and to explore the structure-activity relationships. The structures of the compounds were determined by (1) H NMR and mass spectroscopy. Cell growth inhibition assays revealed that the aloe emodin derivatives 4d, 4f, and 4i effectively decreased the growth of HepG2 (human liver cancer cells) and NCI-H460 (human lung cancer cells) and some of the derivatives exhibited comparable antitumor activity against HeLa (Human epithelial carcinoma cells) and PC3 (prostate cancer cells) cell lines compared to that of the parent aloe emodin at low micromolar concentrations.


Subject(s)
Anthraquinones/chemistry , Anthraquinones/pharmacology , Antineoplastic Agents/chemical synthesis , Anthraquinones/chemical synthesis , Antineoplastic Agents/chemistry , Antineoplastic Agents/pharmacology , Cell Line, Tumor , Cell Proliferation/drug effects , HeLa Cells , Hep G2 Cells , Humans , Structure-Activity Relationship
4.
Mol Oncol ; 9(1): 309-22, 2015 Jan.
Article in English | MEDLINE | ID: mdl-25245324

ABSTRACT

Novel combinations aiming at maximizing the efficacy of bortezomib are highly valued in the clinic. Therefore the current study investigated the outcomes of combining bortezomib with dipyridamole, a well-known antiplatelet. The co-treatment exerted a synergistic lethality in a panel of human leukemia/lymphoma cell lines of different origin. Mechanistically, dipyridamole did not modulate the proteasome inhibitory activity of bortezomib. However, dipyridamole triggered an endoplasmic reticulum (ER) stress, and co-treatment with bortezomib resulted in higher levels of ER stress than either monotherapies. Relieving ER stress with the protein translation inhibitor, cycloheximide suppressed cell death. Moreover, the enhanced ER stress by the co-treatment was associated with an aggravation of reactive oxygen species (ROS) generation and glutathione (GSH) depletion. Replenishing GSH pools significantly scavenged ROS and rescued the cells. Importantly, the cytotoxicity of the co-treatment was executed mainly via the mitochondrial apoptotic pathway with an efficient suppression of the key anti-apoptotic regulators, Mcl-1, Bcl-xl, Bcl-2 and XIAP, driving the independence of the co-treatment-induced apoptosis of a single apoptotic trigger. Furthermore, the intrinsic potential of bortezomib to inhibit important pro-survival pathways was enhanced by dipyridamole in a GSH/ROS-dependent manner. Interestingly, dipyridamole abrogated JAK2 phosphorylation indirectly and selectively in cancer cells, and the co-treatment-induced cytotoxicity was preserved in a model of stromal-mediated chemoresistance. In nude mice, the antitumor activity of the co-treatment surpassed that of bortezomib monotherapy despite that synergy was lacking. In summary, findings of the present study provided a preclinical rationale which warrants further clinical evaluation of bortezomib/dipyridamole novel combination in hematologic malignancies.


Subject(s)
Antineoplastic Combined Chemotherapy Protocols/pharmacology , Hematologic Neoplasms/drug therapy , Animals , Bortezomib/pharmacology , Dipyridamole/pharmacology , HL-60 Cells , Hematologic Neoplasms/metabolism , Hematologic Neoplasms/pathology , Humans , Jurkat Cells , K562 Cells , Mice , Mice, Nude , Neoplasm Proteins/antagonists & inhibitors , Neoplasm Proteins/metabolism , U937 Cells , Xenograft Model Antitumor Assays
5.
Nat Struct Mol Biol ; 21(8): 696-703, 2014 Aug.
Article in English | MEDLINE | ID: mdl-24997597

ABSTRACT

Polo-like kinase 4 (Plk4) is a key regulator of centriole duplication, an event critical for the maintenance of genomic integrity. We show that Plk4 relocalizes from the inner Cep192 ring to the outer Cep152 ring as newly recruited Cep152 assembles around the Cep192-encircled daughter centriole. Crystal-structure analyses revealed that Cep192- and Cep152-derived peptides bind the cryptic polo box (CPB) of Plk4 in opposite orientations and in a mutually exclusive manner. The Cep152 peptide bound to the CPB markedly better than did the Cep192 peptide and effectively 'snatched' the CPB away from a preformed CPB-Cep192 peptide complex. A cancer-associated Cep152 mutation impairing the Plk4 interaction induced defects in procentriole assembly and chromosome segregation. Thus, Plk4 is intricately regulated in time and space through ordered interactions with two distinct scaffolds, Cep192 and Cep152, and a failure in this process may lead to human cancer.


Subject(s)
Cell Cycle Proteins/chemistry , Centrioles/metabolism , Chromosomal Proteins, Non-Histone/chemistry , Protein Serine-Threonine Kinases/chemistry , Amino Acid Sequence , Cell Cycle Proteins/genetics , Cell Cycle Proteins/metabolism , Chromosomal Proteins, Non-Histone/metabolism , Crystallography, X-Ray , HEK293 Cells , HeLa Cells , Humans , Hydrogen Bonding , Models, Molecular , Molecular Sequence Data , Mutation, Missense , Neoplasms/genetics , Protein Binding , Protein Interaction Domains and Motifs , Protein Serine-Threonine Kinases/metabolism , Protein Structure, Quaternary , Protein Structure, Secondary
6.
Mutat Res Rev Mutat Res ; 761: 31-39, 2014.
Article in English | MEDLINE | ID: mdl-24630986

ABSTRACT

Despite advances in treatment, prognosis for many types of carcinoma remains poor. Polo-like kinase 1 (Plk1) has been explored as a target for the development of anticancer drugs. As a mitotic master Ser/Thr kinase, Plk1 is involved in centrosomal maturation, microtubule nucleation, chromosomal segregation, and cytokinesis. Additional functions in interphase and in response to DNA damage have been revealed. The multiple locations of Plk1 correspond to distinct functions, mediated by phosphorylation of multiple substrates. Since it is highly expressed in several carcinomas, and expression of Plk1 is inversely correlated with the survival rate of patients in non-small cell lung, head and neck, and esophageal cancer, Plk1 is recognized as a valid prognostic marker. Connections between Plk1 and p53 or KRAS in carcinoma provide a rationale and several possible routes to the development of therapies. Tumors with both p53-deficiency and high Plk1 expression may be particularly sensitive to Plk1 inhibitors, although some controversial data exist. In KRAS-mutant cancers, on the other hand, Plk1 may be essential for tumor cell survival, but detailed studies as to whether Plk1 inhibitors are more effective in KRAS-mutant cancers must be performed in order to determine whether this is the case. Here, we present evidence for Plk1 as a prognostic marker and potentially effective target for the treatment of patients with carcinoma, to demonstrate the value of Plk1 as a target for the development of cancer treatment, especially for patients with solid tumors. In addition, the effects of Plk1 inhibition in p53- or KRAS-mutated cancer are discussed with respect to clinical implications. Structural specifics of Plk1 are presented, as well as current strategies for discovering new Plk1 inhibitors by targeting the conserved ATP binding site or polo-box domain of Plk1, in order to develop Plk1-specific anticancer drugs.

7.
Cancer Prev Res (Phila) ; 7(1): 74-85, 2014 Jan.
Article in English | MEDLINE | ID: mdl-24169959

ABSTRACT

Mitogen- and stress-activated kinase 1 (MSK1) is a nuclear serine/threonine protein kinase that acts downstream of both extracellular signal-regulated kinases and p38 mitogen-activated protein kinase in response to stress or mitogenic extracellular stimuli. Increasing evidence has shown that MSK1 is closely associated with malignant transformation and cancer development. MSK1 should be an effective target for cancer chemoprevention and chemotherapy. However, very few MSK1 inhibitors, especially natural compounds, have been reported. We used virtual screening of a natural products database and the active conformation of the C-terminal kinase domain of MSK1 (PDB id 3KN) as the receptor structure to identify chrysin and its derivative, compound 69407, as inhibitors of MSK1. Compared with chrysin, compound 69407 more strongly inhibited proliferation and 12-O-tetradecanoylphorbol-13-acetate (TPA)-induced neoplastic transformation of JB6 P+ cells with lower cytotoxicity. Western blot data demonstrated that compound 69407 suppressed phosphorylation of the MSK1 downstream effector histone H3 in intact cells. Knocking down the expression of MSK1 effectively reduced the sensitivity of JB6 P+ cells to compound 69407. Moreover, topical treatment with compound 69407 before TPA application significantly reduced papilloma development in terms of number and size in a two-stage mouse skin carcinogenesis model. The reduction in papilloma development was accompanied by the inhibition of histone H3 phosphorylation at Ser10 in tumors extracted from mouse skin. The results indicated that compound 69407 exerts inhibitory effects on skin tumorigenesis by directly binding with MSK1 and attenuates the MSK1/histone H3 signaling pathway, which makes it an ideal chemopreventive agent against skin cancer.


Subject(s)
Flavones/pharmacology , Flavonoids/pharmacology , Phenethylamines/pharmacology , Ribosomal Protein S6 Kinases, 90-kDa/metabolism , Skin Neoplasms/prevention & control , Animals , Anticarcinogenic Agents/pharmacology , Cell Cycle/drug effects , Cell Proliferation , Cell Transformation, Neoplastic , Histones/metabolism , Male , Mice , Papilloma/prevention & control , Phosphorylation , Protein Structure, Tertiary , Ribosomal Protein S6 Kinases, 90-kDa/antagonists & inhibitors , Signal Transduction/drug effects , Skin/drug effects , Tetradecanoylphorbol Acetate , p38 Mitogen-Activated Protein Kinases/metabolism
8.
Proc Natl Acad Sci U S A ; 110(50): E4849-57, 2013 Dec 10.
Article in English | MEDLINE | ID: mdl-24277814

ABSTRACT

Centrosomes play an important role in various cellular processes, including spindle formation and chromosome segregation. They are composed of two orthogonally arranged centrioles, whose duplication occurs only once per cell cycle. Accurate control of centriole numbers is essential for the maintenance of genomic integrity. Although it is well appreciated that polo-like kinase 4 (Plk4) plays a central role in centriole biogenesis, how it is recruited to centrosomes and whether this step is necessary for centriole biogenesis remain largely elusive. Here we showed that Plk4 localizes to distinct subcentrosomal regions in a temporally and spatially regulated manner, and that Cep192 and Cep152 serve as two distinct scaffolds that recruit Plk4 to centrosomes in a hierarchical order. Interestingly, Cep192 and Cep152 competitively interacted with the cryptic polo box of Plk4 through their homologous N-terminal sequences containing acidic-α-helix and N/Q-rich motifs. Consistent with these observations, the expression of either one of these N-terminal fragments was sufficient to delocalize Plk4 from centrosomes. Furthermore, loss of the Cep192- or Cep152-dependent interaction with Plk4 resulted in impaired centriole duplication that led to delayed cell proliferation. Thus, the spatiotemporal regulation of Plk4 localization by two hierarchical scaffolds, Cep192 and Cep152, is critical for centriole biogenesis.


Subject(s)
Cell Cycle Proteins/metabolism , Centrioles/physiology , Centrosome/metabolism , Chromosomal Proteins, Non-Histone/metabolism , Protein Serine-Threonine Kinases/metabolism , Cell Cycle Proteins/genetics , Centrioles/metabolism , Chromosomal Proteins, Non-Histone/genetics , Cloning, Molecular , Computational Biology , DNA, Complementary/genetics , Fluorescent Antibody Technique, Indirect , Immunoblotting , Immunoprecipitation , Lentivirus , Mutagenesis , Oligonucleotides/genetics , Reverse Transcriptase Polymerase Chain Reaction , Sequence Analysis, DNA
9.
Acta Crystallogr D Biol Crystallogr ; 69(Pt 8): 1522-9, 2013 Aug.
Article in English | MEDLINE | ID: mdl-23897475

ABSTRACT

Unlike other classical protein tyrosine phosphatases (PTPs), PTPRQ (PTP receptor type Q) has dephosphorylating activity towards phosphatidylinositide (PI) substrates. Here, the structure of the catalytic domain of PTPRQ was solved at 1.56 Šresolution. Overall, PTPRQ adopts a tertiary fold typical of other classical PTPs. However, the disordered M6 loop of PTPRQ surrounding the catalytic core and the concomitant absence of interactions of this loop with residues in the PTP loop results in a flat active-site pocket. On the basis of structural and biochemical analyses, it is proposed that this structural feature might facilitate the accommodation of large substrates, making it suitable for the dephosphorylation of PI substrates. Moreover, subsequent kinetic experiments showed that PTPRQ has a strong preferences for PI(3,4,5)P3 over other PI substrates, suggesting that its regulation of cell survival and proliferation reflects downregulation of Akt signalling.


Subject(s)
Receptor-Like Protein Tyrosine Phosphatases, Class 3/chemistry , Receptor-Like Protein Tyrosine Phosphatases, Class 3/metabolism , Catalytic Domain , Crystallography, X-Ray , Humans , Kinetics , Models, Molecular , Mutation , Phosphatidylinositol Phosphates/chemistry , Phosphatidylinositol Phosphates/metabolism , Phosphorylation , Protein Conformation , Receptor-Like Protein Tyrosine Phosphatases, Class 3/genetics , Substrate Specificity
10.
J Biol Chem ; 288(36): 25924-25937, 2013 Sep 06.
Article in English | MEDLINE | ID: mdl-23888052

ABSTRACT

Chrysin (5,7-dihydroxyflavone), a natural flavonoid widely distributed in plants, reportedly has chemopreventive properties against various cancers. However, the anticancer activity of chrysin observed in in vivo studies has been disappointing. Here, we report that a chrysin derivative, referred to as compound 69407, more strongly inhibited EGF-induced neoplastic transformation of JB6 P(+) cells compared with chrysin. It attenuated cell cycle progression of EGF-stimulated cells at the G1 phase and inhibited the G1/S transition. It caused loss of retinoblastoma phosphorylation at both Ser-795 and Ser-807/811, the preferred sites phosphorylated by Cdk4/6 and Cdk2, respectively. It also suppressed anchorage-dependent and -independent growth of A431 human epidermoid carcinoma cells. Compound 69407 reduced tumor growth in the A431 mouse xenograft model and retinoblastoma phosphorylation at Ser-795 and Ser-807/811. Immunoprecipitation kinase assay results showed that compound 69407 attenuated endogenous Cdk4 and Cdk2 kinase activities in EGF-stimulated JB6 P(+) cells. Pulldown and in vitro kinase assay results indicated that compound 69407 directly binds with Cdk2 and Cdk4 in an ATP-independent manner and inhibited their kinase activities. A binding model between compound 69407 and a crystal structure of Cdk2 predicted that compound 69407 was located inside the Cdk2 allosteric binding site. The binding was further verified by a point mutation binding assay. Overall results indicated that compound 69407 is an ATP-noncompetitive cyclin-dependent kinase inhibitor with anti-tumor effects, which acts by binding inside the Cdk2 allosteric pocket. This study provides new insights for creating a general pharmacophore model to design and develop novel ATP-noncompetitive agents with chemopreventive or chemotherapeutic potency.


Subject(s)
Carcinoma, Squamous Cell/drug therapy , Cyclin-Dependent Kinases/antagonists & inhibitors , Flavonoids/pharmacology , Protein Kinase Inhibitors/pharmacology , Skin Neoplasms/drug therapy , Allosteric Regulation/drug effects , Animals , Binding Sites , Carcinoma, Squamous Cell/enzymology , Carcinoma, Squamous Cell/genetics , Carcinoma, Squamous Cell/pathology , Cell Line, Tumor , Crystallography, X-Ray , Cyclin-Dependent Kinases/genetics , Cyclin-Dependent Kinases/metabolism , Epidermal Growth Factor/genetics , Epidermal Growth Factor/metabolism , Flavonoids/chemistry , G1 Phase/drug effects , G1 Phase/genetics , Humans , Mice , Mice, Inbred BALB C , Mice, Nude , Models, Molecular , Neoplasm Transplantation , Protein Kinase Inhibitors/chemistry , Retinoblastoma Protein/genetics , Retinoblastoma Protein/metabolism , S Phase/drug effects , S Phase/genetics , Skin Neoplasms/enzymology , Skin Neoplasms/genetics , Skin Neoplasms/pathology
11.
Cell Cycle ; 12(4): 655-62, 2013 Feb 15.
Article in English | MEDLINE | ID: mdl-23343771

ABSTRACT

Primary cilia are microtubule-based solitary sensing structures on the cell surface that play crucial roles in cell signaling and development. Abnormal ciliary function leads to various human genetic disorders, collectively known as ciliopathies. Outer dense fiber protein 2 (Odf2) was initially isolated as a major component of sperm-tail fibers. Subsequent studies have demonstrated the existence of many splicing variants of Odf2, including Cenexin1 (Odf2 isoform 9), which bears an unusual C-terminal extension. Strikingly, Odf2 localizes along the axoneme of primary cilia, whereas Cenexin1 localizes to basal bodies in cultured mammalian cells. Whether Odf2 and Cenexin1 contribute to primary cilia assembly by carrying out either concerted or distinct functions is unknown. By taking advantage of odf2-/- cells lacking endogenous Odf2 and Cenexin1, but exogenously expressing one or both of these proteins, we showed that Cenexin1, but not Odf2, was necessary and sufficient to induce ciliogenesis. Furthermore, the Cenexin1-dependent primary cilia assembly pathway appeared to function independently of Odf2. Consistently, Cenexin1, but not Odf2, interacted with GTP-loaded Rab8a, localized to the distal/subdistal appendages of basal bodies, and facilitated the recruitment of Chibby, a centriolar component that is important for proper ciliogenesis. Taken together, our results suggest that Cenexin1 plays a critical role in ciliogenesis through its C-terminal extension that confers a unique ability to mediate primary cilia assembly. The presence of multiple splicing variants hints that the function of Odf2 is diversified in such a way that each variant has a distinct role in the complex cellular and developmental processes.


Subject(s)
Axoneme/genetics , Centrioles/genetics , Cilia/genetics , Gene Expression Regulation , Heat-Shock Proteins/genetics , Alternative Splicing , Animals , Axoneme/metabolism , Axoneme/ultrastructure , Carrier Proteins/genetics , Carrier Proteins/metabolism , Cell Line, Tumor , Centrioles/metabolism , Centrioles/ultrastructure , Cilia/metabolism , Cilia/ultrastructure , HEK293 Cells , Heat-Shock Proteins/metabolism , Humans , Mice , Nuclear Proteins/genetics , Nuclear Proteins/metabolism , Protein Binding , Protein Isoforms/genetics , Protein Isoforms/metabolism , Protein Structure, Tertiary , Signal Transduction , Transfection , rab GTP-Binding Proteins/genetics , rab GTP-Binding Proteins/metabolism
12.
Mol Cell ; 49(1): 172-85, 2013 Jan 10.
Article in English | MEDLINE | ID: mdl-23142078

ABSTRACT

The metabolism of glucose and glutamine, primary carbon sources utilized by mitochondria to generate energy and macromolecules for cell growth, is directly regulated by mTORC1. We show that glucose and glutamine, by supplying carbons to the TCA cycle to produce ATP, positively feed back to mTORC1 through an AMPK-, TSC1/2-, and Rag-independent mechanism by regulating mTORC1 assembly and its lysosomal localization. We discovered that the ATP-dependent TTT-RUVBL1/2 complex was disassembled and repressed by energy depletion, resulting in its decreased interaction with mTOR. The TTT-RUVBL complex was necessary for the interaction between mTORC1 and Rag and formation of mTORC1 obligate dimers. In cancer tissues, TTT-RUVBL complex mRNAs were elevated and positively correlated with transcripts encoding proteins of anabolic metabolism and mitochondrial function-all mTORC1-regulated processes. Thus, the TTT-RUVBL1/2 complex responds to the cell's metabolic state, directly regulating the functional assembly of mTORC1 and indirectly controlling the nutrient signal from Rags to mTORC1.


Subject(s)
Energy Metabolism , Lysosomes/metabolism , Proteins/metabolism , Stress, Physiological , ATPases Associated with Diverse Cellular Activities , Adenosine Triphosphate/metabolism , Adenylate Kinase/metabolism , Animals , Breast Neoplasms/genetics , Breast Neoplasms/metabolism , Carcinoma/genetics , Carcinoma/metabolism , Carrier Proteins/genetics , Carrier Proteins/metabolism , Cells, Cultured , Citric Acid Cycle , DNA Helicases/genetics , DNA Helicases/metabolism , Female , Glucose/deficiency , Glutamine/deficiency , Humans , Intracellular Signaling Peptides and Proteins , Mechanistic Target of Rapamycin Complex 1 , Mice , Mice, Knockout , Monomeric GTP-Binding Proteins/metabolism , Multiprotein Complexes , Protein Binding , Protein Multimerization , Protein Transport , Ribosomal Protein S6 Kinases, 90-kDa/metabolism , Signal Transduction , Statistics, Nonparametric , TOR Serine-Threonine Kinases , Telomere-Binding Proteins/genetics , Telomere-Binding Proteins/metabolism , Tuberous Sclerosis Complex 1 Protein , Tuberous Sclerosis Complex 2 Protein , Tumor Suppressor Proteins/genetics , Tumor Suppressor Proteins/metabolism
13.
Biosci Rep ; 32(5): 455-63, 2012 Oct.
Article in English | MEDLINE | ID: mdl-22724525

ABSTRACT

RNase H (retroviral ribonuclease H) cleaves the phosphate backbone of the RNA template within an RNA/DNA hybrid to complete the synthesis of double-stranded viral DNA. In the present study we have determined the complete structure of the RNase H domain from XMRV (xenotropic murine leukaemia virus-related virus) RT (reverse transcriptase). The basic protrusion motif of the XMRV RNase H domain is folded as a short helix and an adjacent highly bent loop. Structural superposition and subsequent mutagenesis experiments suggest that the basic protrusion motif plays a role in direct binding to the major groove in RNA/DNA hybrid, as well as in establishing the co-ordination among modules in RT necessary for proper function.


Subject(s)
Ribonuclease H/chemistry , Xenotropic murine leukemia virus-related virus/enzymology , Amino Acid Sequence , Catalytic Domain , Conserved Sequence , Crystallography, X-Ray , Models, Molecular , Molecular Sequence Data , Mutation , Protein Conformation , Protein Structure, Tertiary , RNA-Directed DNA Polymerase/chemistry , RNA-Directed DNA Polymerase/metabolism , Ribonuclease H/genetics , Ribonuclease H/metabolism
14.
Bioorg Med Chem Lett ; 22(5): 2084-8, 2012 Mar 01.
Article in English | MEDLINE | ID: mdl-22326397

ABSTRACT

The serine phosphatase SerB653 plays a crucial role in the infection of Porphyromonas gingivalis, which contributes to the pathogenesis of periodontitis, an inflammatory disease of teeth-supporting tissues. Because functional loss of SerB653 eliminates the virulence of P. gingivalis, SerB653 inhibitors are considered potential periodontitis therapeutic or preventive agents. To identify SerB653 inhibitors with potent anti-periodontitis activity, we conducted a high-throughput screen of a representative 6800-compound subset of a synthetic chemical library of the Korea Chemical Bank (KCB) for compounds with activity against SerB653. The primary screening yielded 150 hits, and subsequent confirmatory studies identified eight compounds, mainly within a single cluster of 3-acyl-2-phenylamino-1,4-dihydroquinolin-4-one derivatives, that showed greater than 50% inhibition of SerB653 activity at a concentration of 50µM. A second screening with a focused library identified 10 compounds with IC(50) values less than 10µM. In antibacterial tests, three of these compounds showed a minimum inhibitory concentration against P. gingivalis growth of 5-50nM.


Subject(s)
Enzyme Inhibitors/pharmacology , Periodontitis/microbiology , Phosphoric Monoester Hydrolases/antagonists & inhibitors , Porphyromonas gingivalis/drug effects , Porphyromonas gingivalis/enzymology , Quinolines/pharmacology , Bacteroidaceae Infections/drug therapy , Bacteroidaceae Infections/enzymology , Enzyme Inhibitors/chemistry , Humans , Models, Molecular , Periodontitis/drug therapy , Phosphoric Monoester Hydrolases/metabolism , Porphyromonas gingivalis/growth & development , Quinolines/chemistry , Structure-Activity Relationship
16.
Proc Natl Acad Sci U S A ; 108(28): 11446-51, 2011 Jul 12.
Article in English | MEDLINE | ID: mdl-21690413

ABSTRACT

Bipolar spindle formation is pivotal for accurate segregation of mitotic chromosomes during cell division. A growing body of evidence suggests that, in addition to centrosome- and chromatin-based microtubule (MT) nucleation, MT-based MT nucleation plays an important role for proper bipolar spindle formation in various eukaryotic organisms. Although a recently discovered Augmin complex appears to play a central role in this event, how Augmin is regulated remains unknown. Here we provide evidence that a mammalian polo-like kinase 1 (Plk1) localizes to mitotic spindles and promotes MT-based MT nucleation by directly regulating Augmin. Mechanistically, we demonstrated that Cdc2-dependent phosphorylation on a γ-tubulin ring complex (γ-TuRC) recruitment protein, Nedd1/GCP-WD, at the previously uncharacterized S460 residue induces the Nedd1-Plk1 interaction. This step appeared to be critical to allow Plk1 to phosphorylate the Hice1 subunit of the Augmin complex to promote the Augmin-MT interaction and MT-based MT nucleation from within the spindle. Loss of either the Nedd1 S460 function or the Plk1-dependent Hice1 phosphorylation impaired both the Augmin-MT interaction and γ-tubulin recruitment to the spindles, thus resulting in improper bipolar spindle formation that ultimately leads to mitotic arrest and apoptotic cell death. Thus, via the formation of the Nedd1-Plk1 complex and subsequent Augmin phosphorylation, Plk1 regulates spindle MT-based MT nucleation to accomplish normal bipolar spindle formation and mitotic progression.


Subject(s)
Cell Cycle Proteins/metabolism , Microtubules/metabolism , Protein Serine-Threonine Kinases/metabolism , Proto-Oncogene Proteins/metabolism , Amino Acid Sequence , Animals , Base Sequence , Binding Sites/genetics , CDC2 Protein Kinase , Cell Cycle Proteins/genetics , Cyclin B/metabolism , Cyclin-Dependent Kinases , DNA Primers/genetics , HeLa Cells , Humans , Microtubule-Associated Proteins/chemistry , Microtubule-Associated Proteins/genetics , Microtubule-Associated Proteins/metabolism , Microtubule-Organizing Center/metabolism , Models, Biological , Molecular Sequence Data , Multiprotein Complexes/metabolism , Phosphorylation , Protein Serine-Threonine Kinases/genetics , Proto-Oncogene Proteins/genetics , RNA Interference , Recombinant Proteins/chemistry , Recombinant Proteins/genetics , Recombinant Proteins/metabolism , Spindle Apparatus/metabolism , Tubulin/metabolism , Polo-Like Kinase 1
17.
Cell Cycle ; 10(9): 1374-7, 2011 May 01.
Article in English | MEDLINE | ID: mdl-21519187

ABSTRACT

The accurate division of duplicated DNA is essential for maintenance of genomic stability in proliferating eukaryotic cells. Errors in DNA replication and chromosomal segregation may lead to cell death or genomic mutations that lead to oncogenic properties. Thus, tight regulation of DNA replication and mitosis is essential for maintaining genomic integrity. Cell division cycle 6 (Cdc6) is an essential factor for initiating DNA replication. Recent work shows that phosphorylation of Cdc6 by polo-like kinase 1 (Plk1), one of the essential mitotic kinases, regulates mitotic exit mediated by Cdk1 and separase. Here we discuss how pre-replicative complex factors are connected with Plk1 and affect mitotic exit.


Subject(s)
Cell Cycle Proteins/physiology , Cell Division/physiology , DNA Replication/physiology , Mitosis/physiology , Protein Serine-Threonine Kinases/physiology , Proto-Oncogene Proteins/physiology , Animals , Cell Cycle Proteins/metabolism , Humans , Membrane Proteins/physiology , Nuclear Proteins/metabolism , Phosphoproteins/physiology , Polo-Like Kinase 1
18.
Proc Natl Acad Sci U S A ; 108(20): 8200-5, 2011 May 17.
Article in English | MEDLINE | ID: mdl-21525413

ABSTRACT

The feed-forward mechanism is observed in some of the intracellular events, such as metabolic and transcriptional regulatory networks, but not in dynamic mitotic processes. Mammalian polo-like kinase 1 (Plk1) rapidly accumulates at centrosomes and kinetochores as cells enter mitosis. Plk1 function is spatially regulated through the targeting activity of the polo-box domain (PBD) that binds to a phosphoepitope generated by either cyclin dependent kinase 1 (Cdk1) (non-self-priming) or Plk1 itself (self-priming). "Non-self-priming and binding" is thought to ensure the orderly execution of cell cycle events. The physiological significance of the "self-priming and binding" is unknown. Using a pair of ELISA, here we demonstrated that mutations of the self-priming site of a kinetochore component, PBIP1/MLF1IP/KLIP1/CENP-50/CENP-U (PBIP1), to a Cdk1-dependent non-self-priming site abolished product-activated cooperativity in the formation of the Plk1-PBIP1 complex. Both PBD-dependent "two-dimensional" interaction with surface-restricted PBIP1 and subsequent phosphorylation of PBIP1 by anchored Plk1 were crucial to cooperatively generate the Plk1-PBIP1 complex. Highlighting the importance of this mechanism, failure in this process resulted in improper Plk1 recruitment to kinetochores, mitotic arrest, chromosome missegregation, and apoptosis. Thus, Plk1 PBD-dependent biochemical cooperativity is tightly coupled to mitotic events at the kinetochore plate through a product-activated, feed-forward mechanism. Given the critical role of self-priming and binding in the recruitment of Plk1 to surface-confined structures, such as centrosomes, kinetochores, and midbody, we propose that the observed feed-forward mechanism serves as a fundamental biochemical process that ensures dynamic nature of Plk1 localization to and delocalization from these subcellular locations.


Subject(s)
Kinetochores , Mitosis , CDC2 Protein Kinase/metabolism , Cell Cycle Proteins/metabolism , HeLa Cells , Humans , Protein Serine-Threonine Kinases/metabolism , Protein Transport , Proto-Oncogene Proteins/metabolism , Polo-Like Kinase 1
19.
FEBS Lett ; 585(4): 664-70, 2011 Feb 18.
Article in English | MEDLINE | ID: mdl-21266175

ABSTRACT

Heat shock protein 33 (Hsp33) from Escherichia coli is a redox-regulated molecular chaperone that protects cells from oxidative stress. To understand the molecular basis for the monomer-dimer switch in the functional regulation of E. coli Hsp33, we generated a constitutively monomeric Hsp33 by introducing the Q151E mutation in the dimeric interface and determined its crystal structure. The overall scaffold of the monomeric Hsp33(1-235) (Q151E) mutant is virtually the same as that of the dimeric form, except that there is no domain swapping. The measurement of chaperone activity to thermally denatured luciferase showed that the constitutively monomeric Hsp33 mutant still retains chaperone activity similar to that of wild-type Hsp33(1-235), suggesting that a Hsp33 monomer is sufficient to interact with slowly unfolded substrate.


Subject(s)
Escherichia coli Proteins/chemistry , Escherichia coli Proteins/metabolism , Escherichia coli/metabolism , Heat-Shock Proteins/chemistry , Heat-Shock Proteins/metabolism , Molecular Chaperones/chemistry , Molecular Chaperones/metabolism , Mutant Proteins/chemistry , Mutant Proteins/metabolism , Amino Acid Substitution , Dimerization , Escherichia coli Proteins/genetics , Heat-Shock Proteins/genetics , Hot Temperature/adverse effects , Kinetics , Luciferases, Firefly/chemistry , Models, Molecular , Molecular Chaperones/genetics , Mutant Proteins/genetics , Oxidation-Reduction , Oxidative Stress , Peptide Fragments/chemistry , Peptide Fragments/genetics , Peptide Fragments/metabolism , Protein Conformation , Protein Interaction Domains and Motifs , Recombinant Proteins/chemistry , Recombinant Proteins/metabolism
20.
Proc Natl Acad Sci U S A ; 107(46): 19742-7, 2010 Nov 16.
Article in English | MEDLINE | ID: mdl-21041660

ABSTRACT

Defining the links between cell division and DNA replication is essential for understanding normal cell cycle progression and tumorigenesis. In this report we explore the effect of phosphorylation of cell division cycle 6 (Cdc6), a DNA replication initiation factor, by polo-like kinase 1 (Plk1) on the regulation of chromosomal segregation. In mitosis, the phosphorylation of Cdc6 was highly increased, in correlation with the level of Plk1, and conversely, Cdc6 is hypophosphorylated in Plk1-depleted cells, although cyclin A- and cyclin B1-dependent kinases are active. Binding between Cdc6 and Plk1 occurs through the polo-box domain of Plk1, and Cdc6 is phosphorylated by Plk1 on T37. Immunohistochemistry studies reveal that Cdc6 and Plk1 colocalize to the central spindle in anaphase. Expression of T37V mutant of Cdc6 (Cdc6-TV) induces binucleated cells and incompletely separated nuclei. Wild-type Cdc6 but not Cdc6-TV binds cyclin-dependent kinase 1 (Cdk1). Expression of wild-type Plk1 but not kinase-defective mutant promotes the binding of Cdc6 to Cdk1. Cells expressing wild-type Cdc6 display lower Cdk1 activity and higher separase activity than cells expressing Cdc6-TV. These results suggest that Plk1-mediated phosphorylation of Cdc6 promotes the interaction of Cdc6 and Cdk1, leading to the attenuation of Cdk1 activity, release of separase, and subsequent anaphase progression.


Subject(s)
CDC2 Protein Kinase/metabolism , Cell Cycle Proteins/metabolism , Chromosome Segregation , Endopeptidases/metabolism , Mitosis , Nuclear Proteins/metabolism , Protein Serine-Threonine Kinases/metabolism , Proto-Oncogene Proteins/metabolism , Cell Cycle Proteins/chemistry , Cyclin A/metabolism , Cyclin B1/metabolism , DNA Replication , Enzyme Activation , Giant Cells/cytology , Giant Cells/metabolism , HEK293 Cells , HeLa Cells , Humans , Models, Biological , Mutant Proteins/metabolism , Nuclear Proteins/deficiency , Phosphorylation , Phosphothreonine/metabolism , Protein Binding , Protein Serine-Threonine Kinases/chemistry , Protein Serine-Threonine Kinases/deficiency , Protein Structure, Tertiary , Proto-Oncogene Proteins/chemistry , Proto-Oncogene Proteins/deficiency , Separase , Substrate Specificity , Polo-Like Kinase 1
SELECTION OF CITATIONS
SEARCH DETAIL
...