1,626 results on '"Catalytic Domain"'
Search Results
2. SHIP-mediated inhibition of K562 erythroid differentiation requires an intact catalytic domain and Shc binding site
- Author
-
Siegel, Josie, Li, Yun, and Whyte, Peter
- Published
- 1999
- Full Text
- View/download PDF
3. The catalytic domain of PKC-ε, in reciprocal PKC-δ and -ε chimeras, is responsible for conferring tumorgenicity to NIH3T3 cells, whereas both regulatory and catalytic domains of PKC-ε contribute to in vitro transformation
- Author
-
Wang, Qiming J, Acs, Peter, Goodnight, JoAnne, Blumberg, Peter M, Mischak, Harald, and Mushinski, J Frederic
- Published
- 1998
- Full Text
- View/download PDF
4. The c-Src tyrosine kinase associates with the catalytic domain of ErbB-2: implications for ErbB-2 mediated signaling and transformation
- Author
-
Dongmei Zuo, Richard Chan, Monica Najoukas, Morag Park, David Dankort, Harold Kim, and William J. Muller
- Subjects
Cancer Research ,Receptor, ErbB-2 ,Cellular differentiation ,Recombinant Fusion Proteins ,Kidney ,Receptor tyrosine kinase ,Cell Line ,Focal adhesion ,CSK Tyrosine-Protein Kinase ,Dogs ,ErbB ,Catalytic Domain ,Cell polarity ,Genetics ,Animals ,Humans ,Receptor ,Molecular Biology ,Epithelial polarity ,Focal Adhesions ,Binding Sites ,biology ,Cell Polarity ,Cell Differentiation ,Protein-Tyrosine Kinases ,Rats ,ErbB Receptors ,Cell Transformation, Neoplastic ,src-Family Kinases ,Mutation ,biology.protein ,Cancer research ,Mutagenesis, Site-Directed ,Signal transduction ,Signal Transduction - Abstract
c-Src associates with and is activated by the ErbB-2 receptor tyrosine kinase, but is unable to bind the EGFR. Although c-Src has been found to interact directly and specifically with the ErbB-2 receptor, the significance of this interaction is unclear. Using both chimeric receptor and site-directed mutagenesis approaches, the region of interaction of c-Src on ErbB-2 was identified. Significantly, EGFR could be converted into a receptor capable of binding c-Src by replacement of a catalytic domain of ErbB-2. We further demonstrated that MDCK cells that express mutant EGFR that are competent in c-Src recruitment lose epithelial polarity in organoid cultures, whereas cells overexpressing the wild-type EGFR retain a polarized phenotype. ErbB-2-dependent activation of c-Src results in disruption of epithelial cell-cell contacts leading to cell dispersal that correlates with the re-localization of phospho-MAPK to focal adhesions. Taken together, these observations suggest that recruitment of c-Src to these closely related EGFR family members plays a critical role in modulating cell polarity.
- Published
- 2005
5. SHIP-mediated inhibition of K562 erythroid differentiation requires an intact catalytic domain and Shc binding site
- Author
-
Peter Whyte, Josie Siegel, and Yun Li
- Subjects
Cancer Research ,Phosphoric monoester hydrolases ,Cellular differentiation ,Biology ,environment and public health ,Catalysis ,src Homology Domains ,hemic and lymphatic diseases ,Catalytic Domain ,Genetics ,medicine ,Humans ,Binding site ,Phosphorylation ,Molecular Biology ,Adaptor Proteins, Signal Transducing ,DNA Primers ,GRB2 Adaptor Protein ,chemistry.chemical_classification ,Binding Sites ,Base Sequence ,technology, industry, and agriculture ,Proteins ,Cell Differentiation ,Phosphoric Monoester Hydrolases ,Cell biology ,Red blood cell ,medicine.anatomical_structure ,Enzyme ,chemistry ,Biochemistry ,Cell culture ,Phosphatidylinositol-3,4,5-Trisphosphate 5-Phosphatases ,Hemin ,Tyrosine ,biological phenomena, cell phenomena, and immunity ,K562 Cells ,hormones, hormone substitutes, and hormone antagonists ,K562 cells ,Protein Binding - Abstract
Growing evidence supports a role for the SHIP inositol 5'-phosphatase in the negative regulation of a variety of receptor-mediated signaling pathways in hematopoietic cells. SHIP expression among cultured cell lines was examined and found to be restricted to cells of hematopoietic origin, with the exception of the K562 erythroleukemia cell line, in which SHIP protein and mRNA were undetectable. The absence of endogenous SHIP in K562 cells provided a useful system to study the role of SHIP in growth and differentiation. When stably expressed in K562 cells, SHIP was found to be constitutively tyrosine phosphorylated and associated with endogenous Shc and Grb-2. Stable expression of SHIP did not affect growth of the cells but resulted in decreased synthesis of hemoglobin protein and epsilon-globin mRNA in response to hemin, an inducer of erythroid differentiation. This effect was not due to increased cell death in the SHIP-expressing lines following hemin stimulation, but was likely the result of an impaired differentiation program in these cells. Mutational analysis indicated that SHIP must retain both an intact catalytic domain and Shc binding site to efficiently inhibit K562 erythroid differentiation.
- Published
- 1999
6. Nuclear translocation of the catalytic subunit of protein kinase A induced by an antisense oligonucleotide directed against the RIalpha regulatory subunit.
- Author
-
Neary CL and Cho-Chung YS
- Subjects
- Active Transport, Cell Nucleus drug effects, Cell Nucleus drug effects, Cell Nucleus enzymology, Colforsin pharmacology, Cyclic AMP-Dependent Protein Kinase RIalpha Subunit, Cyclic AMP-Dependent Protein Kinases genetics, Humans, Male, Oligonucleotides, Antisense genetics, Protein Subunits, Substrate Specificity, Time Factors, Tumor Cells, Cultured, Catalytic Domain, Cell Nucleus metabolism, Cyclic AMP-Dependent Protein Kinases chemistry, Cyclic AMP-Dependent Protein Kinases metabolism, Oligonucleotides, Antisense pharmacology
- Abstract
The regulatory (R) subunits of cAMP-dependent protein kinase (PKA) are implicated in the regulation of cell proliferation and differentiation. There are two isoforms of PKA that are distinguished by two types of R subunit, RI and RII. Evidence suggests that RI is associated with proliferation and RII is associated with cell differentiation. Previous work in this laboratory has demonstrated that depletion of the RIalpha subunit by treatment with an antisense oligonucleotide (ODN) induces differentiation in leukemia cells and growth arrest and apoptosis in epithelial cancer cells. Using the prostate cancer cell line PC3M as a model system, we have developed a cell line that overexpresses a retroviral vector construct containing the RIalpha antisense gene. This cell line has been characterized and the effectiveness of the construct determined. In the work presented here, we demonstrate by immunocytochemistry that treatment with RIalpha antisense ODN induces translocation of the Calpha subunit of PKA to the nucleus of PC3M prostate cancer cells. The translocation of Calpha triggered by exogenous antisense ODN treatment mirrors that observed in cells endogenously overexpressing the antisense gene. Triggering the nuclear translocation of the Calpha subunit of PKA in the cell may be an important mechanism of action of RIalpha antisense that regulates cell growth independent of adenylate cyclase and cellular cAMP levels. The nuclear localization of the Calpha subunit of PKA may be an essential step in revealing the mechanism whereby this critical kinase regulates cell growth.
- Published
- 2001
- Full Text
- View/download PDF
7. Transamidase site-targeted agents alter the conformation of the transglutaminase cancer stem cell survival protein to reduce GTP binding activity and cancer stem cell survival.
- Author
-
Kerr C, Szmacinski H, Fisher ML, Nance B, Lakowicz JR, Akbar A, Keillor JW, Lok Wong T, Godoy-Ruiz R, Toth EA, Weber DJ, and Eckert RL
- Subjects
- Aminoacyltransferases chemistry, Binding Sites drug effects, Catalytic Domain drug effects, Catalytic Domain genetics, Cell Survival drug effects, Cell Survival genetics, Cells, Cultured, GTP-Binding Proteins genetics, GTP-Binding Proteins metabolism, Gene Knockout Techniques, Humans, Molecular Targeted Therapy, Protein Binding drug effects, Protein Conformation drug effects, Protein Glutamine gamma Glutamyltransferase 2, Transglutaminases genetics, Transglutaminases metabolism, Aminoacyltransferases antagonists & inhibitors, Antineoplastic Agents pharmacology, GTP-Binding Proteins antagonists & inhibitors, GTP-Binding Proteins chemistry, Guanosine Triphosphate metabolism, Neoplastic Stem Cells drug effects, Neoplastic Stem Cells physiology, Transglutaminases antagonists & inhibitors, Transglutaminases chemistry
- Abstract
Type 2 transglutaminase (TG2) is an important cancer stem cell survival protein that exists in open and closed conformations. The major intracellular form is the closed conformation that functions as a GTP-binding GTPase and is required for cancer stem cell survival. However, at a finite rate, TG2 transitions to an open conformation that exposes the transamidase catalytic site involved in protein-protein crosslinking. The activities are mutually exclusive, as the closed conformation has GTP binding/GTPase activity, and the open conformation transamidase activity. We recently showed that GTP binding, but not transamidase activity, is required for TG2-dependent cancer stem cell invasion, migration and tumour formation. However, we were surprised that transamidase site-specific inhibitors reduce cancer stem cell survival. We now show that compounds NC9, VA4 and VA5, which react exclusively at the TG2 transamidase site, inhibit both transamidase and GTP-binding activities. Transamidase activity is inhibited by direct inhibitor binding at the transamidase site, and GTP binding is blocked because inhibitor interaction at the transamidase site locks the protein in the extended/open conformation to disorganize/inactivate the GTP binding/GTPase site. These findings suggest that transamidase site-specific inhibitors can inhibit GTP binding/signalling by driving a conformation change that disorganizes the TG2 GTP binding to reduce TG2-dependent signalling, and that drugs designed to target this site may be potent anti-cancer agents.
- Published
- 2017
- Full Text
- View/download PDF
8. WNK kinases, a novel protein kinase subfamily in multi-cellular organisms
- Author
-
Veríssimo, Fátima and Jordan, Peter
- Published
- 2001
- Full Text
- View/download PDF
9. NPM-ALK phosphorylates WASp Y102 and contributes to oncogenesis of anaplastic large cell lymphoma.
- Author
-
Murga-Zamalloa CA, Mendoza-Reinoso V, Sahasrabuddhe AA, Rolland D, Hwang SR, McDonnell SR, Sciallis AP, Wilcox RA, Bashur V, Elenitoba-Johnson K, and Lim MS
- Subjects
- Animals, Carcinogenesis, Catalytic Domain, Cell Line, Tumor, Gene Knockdown Techniques, Heterografts, Humans, Lymphoma, Large-Cell, Anaplastic genetics, Lymphoma, Large-Cell, Anaplastic pathology, Mice, Mice, SCID, Phosphorylation, Wiskott-Aldrich Syndrome Protein genetics, Lymphoma, Large-Cell, Anaplastic metabolism, Protein-Tyrosine Kinases metabolism, Wiskott-Aldrich Syndrome Protein metabolism
- Abstract
Mechanisms by which NPM-ALK signaling regulates cell migration, invasion and contributes to the oncogenesis of anaplastic large cell lymphoma (ALCL) are not completely understood. In an attempt to identify novel actin signaling pathways regulated by NPM-ALK, a comprehensive phosphoproteome analysis of ALCL cell lines was performed in the presence or absence of NPM-ALK activity. Numerous phosphoproteins involved in actin dynamics including Wiskott-Aldrich syndrome protein (WASp) were regulated by NPM-ALK. Network analysis revealed that WASp is a central component of the NPM-ALK-dependent actin signaling pathway. Here we show that NPM-ALK phosphorylates WASp at its known activation site (Y290) as well as at a novel residue (Y102). Phosphorylation of WASp at Y102 negatively regulates its interaction with Wiskott-Aldrich interacting protein and decreases its protein stability. Phosphorylation of WASp at Y102 enhances anchorage-independent growth and tumor growth in an in vivo xenograft model and enhances invasive properties of ALCL. We show that knock-down of WASp or expression of Y102F mutant of WASp decreases colony formation and in vivo tumor growth. Our results show that WASp is a novel substrate of ALK and has a critical role in regulating invasiveness and oncogenesis of ALCL.
- Published
- 2017
- Full Text
- View/download PDF
10. Preventing E-cadherin aberrant N-glycosylation at Asn-554 improves its critical function in gastric cancer.
- Author
-
Carvalho S, Catarino TA, Dias AM, Kato M, Almeida A, Hessling B, Figueiredo J, Gärtner F, Sanches JM, Ruppert T, Miyoshi E, Pierce M, Carneiro F, Kolarich D, Seruca R, Yamaguchi Y, Taniguchi N, Reis CA, and Pinho SS
- Subjects
- Amino Acid Sequence, Animals, Asparagine genetics, Cadherins chemistry, Cadherins genetics, Cadherins physiology, Catalytic Domain genetics, Cell Line, Tumor, Dogs, Gastric Mucosa metabolism, Gastric Mucosa pathology, Glycosylation, HT29 Cells, Humans, Madin Darby Canine Kidney Cells, Mice, Mice, Inbred C57BL, Mice, Knockout, Molecular Sequence Data, Mutagenesis, Site-Directed, N-Acetylglucosaminyltransferases antagonists & inhibitors, N-Acetylglucosaminyltransferases genetics, Sequence Homology, Amino Acid, Stomach Neoplasms genetics, Stomach Neoplasms pathology, Cadherins metabolism, N-Acetylglucosaminyltransferases metabolism, Stomach Neoplasms metabolism
- Abstract
E-cadherin is a central molecule in the process of gastric carcinogenesis and its posttranslational modifications by N-glycosylation have been described to induce a deleterious effect on cell adhesion associated with tumor cell invasion. However, the role that site-specific glycosylation of E-cadherin has in its defective function in gastric cancer cells needs to be determined. Using transgenic mice models and human clinical samples, we demonstrated that N-acetylglucosaminyltransferase V (GnT-V)-mediated glycosylation causes an abnormal pattern of E-cadherin expression in the gastric mucosa. In vitro models further indicated that, among the four potential N-glycosylation sites of E-cadherin, Asn-554 is the key site that is selectively modified with β1,6 GlcNAc-branched N-glycans catalyzed by GnT-V. This aberrant glycan modification on this specific asparagine site of E-cadherin was demonstrated to affect its critical functions in gastric cancer cells by affecting E-cadherin cellular localization, cis-dimer formation, molecular assembly and stability of the adherens junctions and cell-cell aggregation, which was further observed in human gastric carcinomas. Interestingly, manipulating this site-specific glycosylation, by preventing Asn-554 from receiving the deleterious branched structures, either by a mutation or by silencing GnT-V, resulted in a protective effect on E-cadherin, precluding its functional dysregulation and contributing to tumor suppression.
- Published
- 2016
- Full Text
- View/download PDF
11. A novel LKB1 isoform enhances AMPK metabolic activity and displays oncogenic properties.
- Author
-
Dahmani R, Just PA, Delay A, Canal F, Finzi L, Prip-Buus C, Lambert M, Sujobert P, Buchet-Poyau K, Miller E, Cavard C, Marmier S, Terris B, Billaud M, and Perret C
- Subjects
- AMP-Activated Protein Kinase Kinases, Alternative Splicing, Animals, Catalytic Domain, Cell Line, Tumor, Humans, Isoenzymes chemistry, Isoenzymes metabolism, Mice, Mice, Nude, Muscle, Skeletal metabolism, Myocardium metabolism, Neoplasm Transplantation, Neoplasms, Experimental pathology, Protein Serine-Threonine Kinases chemistry, AMP-Activated Protein Kinases metabolism, Neoplasms, Experimental metabolism, Protein Serine-Threonine Kinases metabolism
- Abstract
The LKB1 tumor suppressor gene encodes a master kinase that coordinates the regulation of energetic metabolism and cell polarity. We now report the identification of a novel isoform of LKB1 (named ΔN-LKB1) that is generated through alternative transcription and internal initiation of translation of the LKB1 mRNA. The ΔN-LKB1 protein lacks the N-terminal region and a portion of the kinase domain. Although ΔN-LKB1 is catalytically inactive, it potentiates the stimulating effect of LKB1 on the AMP-activated protein kinase (AMPK) metabolic sensor through a direct interaction with the regulatory autoinhibitory domain of AMPK. In contrast, ΔN-LKB1 negatively interferes with the LKB1 polarizing activity. Finally, combining in vitro and in vivo approaches, we showed that ΔN-LKB1 has an intrinsic oncogenic property. ΔN-LKB1 is expressed solely in the lung cancer cell line, NCI-H460. Silencing of ΔN-LKB1 decreased the survival of NCI-H460 cells and inhibited their tumorigenicity when engrafted in nude mice. In conclusion, we have identified a novel LKB1 isoform that enhances the LKB1-controlled AMPK metabolic activity but inhibits LKB1-induced polarizing activity. Both the LKB1 tumor suppressor gene and the oncogene ΔN-LKB1 are expressed from the same locus and this may account for some of the paradoxical effects of LKB1 during tumorigenesis.
- Published
- 2015
- Full Text
- View/download PDF
12. Preventing E-cadherin aberrant N-glycosylation at Asn-554 improves its critical function in gastric cancer
- Author
-
Y Yamaguchi, Thomas Ruppert, Eiji Miyoshi, Michael Pierce, Telmo Catarino, A. S. Almeida, Naoyuki Taniguchi, Celso A. Reis, Bernd Hessling, Ana M. Dias, Daniel Kolarich, Salomé S. Pinho, Fátima Gärtner, Joao Sanches, Joana Figueiredo, Fátima Carneiro, Raquel Seruca, Sandra Carvalho, M Kato, and Instituto de Investigação e Inovação em Saúde
- Subjects
0301 basic medicine ,Cancer Research ,Glycosylation ,Cadherins/metabolism ,Stomach Neoplasms/genetics ,Stomach Neoplasms/metabolism ,Madin Darby Canine Kidney Cells ,Mice ,chemistry.chemical_compound ,N-linked glycosylation ,Catalytic Domain ,Asparagine/genetics ,Gastric Mucosa/metabolism ,Cellular localization ,Mice, Knockout ,Stomach Neoplasms/pathology ,N-Acetylglucosaminyltransferases/genetics ,Cadherins ,3. Good health ,Cell biology ,medicine.anatomical_structure ,Cadherins/chemistry ,Asparagine ,HT29 Cells ,N-Acetylglucosaminyltransferases/antagonists & inhibitors ,Molecular Sequence Data ,Gastric Mucosa/pathology ,N-Acetylglucosaminyltransferases/metabolism ,Biology ,N-Acetylglucosaminyltransferases ,Article ,Adherens junction ,03 medical and health sciences ,Catalytic Domain/genetics ,Dogs ,Stomach Neoplasms ,Cell Line, Tumor ,Genetics ,Gastric mucosa ,medicine ,Animals ,Humans ,Amino Acid Sequence ,Cell adhesion ,Molecular Biology ,Sequence Homology, Amino Acid ,Cadherin ,Cadherins/genetics ,Mice, Inbred C57BL ,carbohydrates (lipids) ,030104 developmental biology ,chemistry ,Gastric Mucosa ,Cancer cell ,Immunology ,Mutagenesis, Site-Directed ,Cadherins/physiology - Abstract
E-cadherin is a central molecule in the process of gastric carcinogenesis and its posttranslational modifications by N-glycosylation have been described to induce a deleterious effect on cell adhesion associated with tumor cell invasion. However, the role that site-specific glycosylation of E-cadherin has in its defective function in gastric cancer cells needs to be determined. Using transgenic mice models and human clinical samples, we demonstrated that N-acetylglucosaminyltransferase V (GnT-V)-mediated glycosylation causes an abnormal pattern of E-cadherin expression in the gastric mucosa. In vitro models further indicated that, among the four potential N-glycosylation sites of E-cadherin, Asn-554 is the key site that is selectively modified with ß1,6 GlcNAc-branched N-glycans catalyzed by GnT-V. This aberrant glycan modification on this specific asparagine site of E-cadherin was demonstrated to affect its critical functions in gastric cancer cells by affecting E-cadherin cellular localization, cis-dimer formation, molecular assembly and stability of the adherens junctions and cell-cell aggregation, which was further observed in human gastric carcinomas. Interestingly, manipulating this site-specific glycosylation, by preventing Asn-554 from receiving the deleterious branched structures, either by a mutation or by silencing GnT-V, resulted in a protective effect on E-cadherin, precluding its functional dysregulation and contributing to tumor suppression. IPATIMUP integrates the I3S Research Unit, which is partially supported by FCT, the Portuguese Foundation for Science and Technology. This work is funded by FEDER funds through the Operational Programme for Competitiveness Factors—COMPETE— and National Funds through the FCT—Foundation for Science and Technology, under the projects: PTDC/CVT/111358/2009; EXPL/BIM-MEC/0149/2012; and PTDC/BBB-EBI/0786/2012. SC (SFRH/BD/77386/2011), AMD (SFRH/BI/52380/2013) BiotechHealth Doctoral Programme, and SSP (SFRH/BPD/63094/2009) thank FCT and the Luso-American Foundation (FLAD) for funding. JMS acknowledges FCT (UID/EEA/50009/2013). DK acknowledges support by the Max Planck Society and European Union (Seventh Framework Programme 'Glycoproteomics', grant number PCIG09-GA-2011-293847. CAR and DK acknowledge GastricGlycoExplorer project, grant number 316929). We thank Ola Soderberg and Gaëlle Cane (Department of Genetics and Pathology, University of Uppsala, Uppsala, Sweden) for providing the Streptavidin PLA probe and to Márcia Pereira and Sara Campos for the support in PLA technique and statistical analyses.
- Published
- 2015
- Full Text
- View/download PDF
13. Regulation of lipid binding underlies the activation mechanism of class IA PI3-kinases.
- Author
-
Hon WC, Berndt A, and Williams RL
- Subjects
- Amino Acid Sequence, Amino Acid Substitution, Aniline Compounds chemistry, Animals, Catalytic Domain, Cholesterol chemistry, Chromones chemistry, Class I Phosphatidylinositol 3-Kinases genetics, Class Ia Phosphatidylinositol 3-Kinase genetics, Crystallography, X-Ray, Enzyme Activation, Enzyme Activators chemistry, Humans, Hydrophobic and Hydrophilic Interactions, Liposomes chemistry, Mice, Models, Molecular, Molecular Sequence Data, Phosphatidylcholines chemistry, Phosphatidylethanolamines chemistry, Phosphatidylserines chemistry, Phosphoinositide-3 Kinase Inhibitors, Phosphopeptides chemistry, Protein Binding, Protein Structure, Quaternary, Protein Structure, Secondary, Sequence Deletion, Class I Phosphatidylinositol 3-Kinases chemistry, Class Ia Phosphatidylinositol 3-Kinase chemistry
- Abstract
Somatic missense mutations in PIK3CA, which encodes the p110α catalytic subunit of phosphoinositide 3-kinases, occur frequently in human cancers. Activating mutations spread across multiple domains, some of which are located at inhibitory contact sites formed with the regulatory subunit p85α. PIK3R1, which encodes p85α, also has activating somatic mutations. We find a strong correlation between lipid kinase and lipid-binding activities for both wild-type (WT) and a representative set of oncogenic mutant complexes of p110α/p85α. Lipid binding involves both electrostatic and hydrophobic interactions. Activation caused by a phosphorylated receptor tyrosine kinase (RTK) peptide binding to the p85α N-terminal SH2 domain (nSH2) induces lipid binding. This depends on the polybasic activation loop as well as a conserved hydrophobic motif in the C-terminal region of the kinase domain. The hotspot E545K mutant largely mimics the activated WT p110α. It shows the highest basal activity and lipid binding, and is not significantly activated by an RTK phosphopeptide. Both the hotspot H1047R mutant and rare mutations (C420R, M1043I, H1047L, G1049R and p85α-N564D) also show increased basal kinase activities and lipid binding. However, their activities are further enhanced by an RTK phosphopeptide to levels markedly exceeding that of activated WT p110α. Phosphopeptide binding to p110β/p85α and p110δ/p85α complexes also induces their lipid binding. We present a crystal structure of WT p110α complexed with the p85α inter-SH2 domain and the inhibitor PIK-108. Additional to the ATP-binding pocket, an unexpected, second PIK-108 binding site is observed in the kinase C-lobe. We show a global conformational change in p110α consistent with allosteric regulation of the kinase domain by nSH2. These findings broaden our understanding of the differential biological outputs exhibited by distinct types of mutations regarding growth factor dependence, and suggest a two-tier classification scheme relating p110α and p85α mutations with signalling potential.
- Published
- 2012
- Full Text
- View/download PDF
14. Differential roles for the p101 and p84 regulatory subunits of PI3Kγ in tumor growth and metastasis.
- Author
-
Brazzatti JA, Klingler-Hoffmann M, Haylock-Jacobs S, Harata-Lee Y, Niu M, Higgins MD, Kochetkova M, Hoffmann P, and McColl SR
- Subjects
- Animals, Breast Neoplasms enzymology, Breast Neoplasms pathology, Catalytic Domain genetics, Cell Line, Tumor, Cell Movement genetics, Female, Gene Expression Regulation, Enzymologic, Gene Expression Regulation, Neoplastic, Humans, Lung Neoplasms enzymology, Lung Neoplasms secondary, Male, Mice, Mice, SCID, Neoplasm Invasiveness genetics, Neoplasm Transplantation, Class Ib Phosphatidylinositol 3-Kinase metabolism, Neoplasm Metastasis
- Abstract
Phosphoinositide 3-kinase γ (PI3Kγ) consists of a catalytic subunit p110γ, which forms mutually exclusive dimers with one of the regulatory subunits called p101 and p84/p87(PIKAP). Recently, PI3Kγ emerged as being a potential oncogene because overexpression of the catalytic subunit p110γ or the regulatory subunit p101 leads to oncogenic cellular transformation and malignancy. However, the contribution of the individual subunits to tumor growth and metastasis and the mechanisms involved are not understood. We therefore individually knocked down the PI3Kγ subunits (p84, p101 and p110γ) in MDA-MB-231 cells, which reduced in vitro migration of the cell lines. Knockdown of p110γ or p101 inhibited apoptosis, Akt phosphorylation and lung colonization in SCID mice. Similarly, the knockdown of p110γ and p101 in murine epithelial carcinoma 4T1.2 cells inhibited primary tumor growth and spontaneous metastasis, as well as lung colonization. In contrast, knockdown of p84 in MDA-MB-231 cells enhanced Akt phosphorylation and lung colonization. These findings are the first to implicate differential functions of the two PI3Kγ regulatory subunits in the process of oncogenesis, and indicate that loss of p101 is sufficient to reduce in vivo tumor growth and metastasis to the same extent as that of p110γ.
- Published
- 2012
- Full Text
- View/download PDF
15. Integrin-linked kinase: not so 'pseudo' after all.
- Author
-
Hannigan GE, McDonald PC, Walsh MP, and Dedhar S
- Subjects
- Catalytic Domain, Humans, In Vitro Techniques, Mutation, Neoplasms metabolism, Phosphorylation, Protein Serine-Threonine Kinases chemistry, Protein Serine-Threonine Kinases genetics, Protein Serine-Threonine Kinases physiology
- Abstract
Integrin-linked kinase (ILK) is a highly evolutionarily conserved intracellular protein that was originally identified as an integrin-interacting protein, and extensive genetic and biochemical studies have shown that ILK expression is vital during both embryonic development and tissue homeostasis. At the cellular and tissue levels, ILK regulates signaling pathways for cell adhesion-mediated cell survival (anoikis), apoptosis, proliferation and mitosis, migration, invasion, and vascularization and tumor angiogenesis. ILK also has central roles in cardiac and smooth-muscle contractility, and ILK dysregulation causes cardiomyopathies in humans. ILK protein levels are increased in several human cancers and often the expression level predicts poor patient outcome. Abundant evidence has accumulated suggesting that, of the diverse functions of ILK, some may require kinase activity whereas others depend on protein-protein interactions and are, therefore, independent of kinase activity. However, the past several years have seen an ongoing debate about whether ILK indeed functions as a protein serine/threonine kinase. This debate centers on the atypical protein kinase domain of ILK, which lacks some amino-acid residues thought to be essential for phosphotransferase activity. However, similar deficiencies are present in the catalytic domains of other kinases now known to possess protein kinase activity. Numerous studies have shown that ILK phosphorylates peptide substrates in vitro, corresponding to ILK-mediated phosphorylations in intact cells, and a recent report characterizing in vitro phosphotransferase activity of highly purified, full-length ILK, accompanied by detailed enzyme kinetic analyses, shows that, at least in vitro, ILK is a bona fide protein kinase. However, several genetic studies suggest that, not all biological functions of ILK require kinase activity, and that it can function as an adaptor/scaffold protein. Here, we review evidence for and against ILK being an active kinase, and provide a framework for strategies to further analyze the kinase and adaptor functions of ILK in different cellular contexts.
- Published
- 2011
- Full Text
- View/download PDF
16. Regulation of DNA-dependent protein kinase by protein kinase CK2 in human glioblastoma cells.
- Author
-
Olsen BB, Issinger OG, and Guerra B
- Subjects
- Brain Neoplasms metabolism, Casein Kinase II genetics, Catalytic Domain genetics, Cell Death, Cell Line, Tumor, DNA-Activated Protein Kinase genetics, Down-Regulation, Gene Expression Regulation, Neoplastic, Gene Knockdown Techniques, Humans, Phosphorylation, Calcium-Binding Proteins metabolism, Casein Kinase II metabolism, DNA Damage, DNA-Activated Protein Kinase metabolism, Glioblastoma genetics
- Abstract
The DNA-dependent protein kinase (DNA-PK) is a nuclear serine/threonine protein kinase composed of a large catalytic subunit (DNA-PKcs) and a heterodimeric DNA-targeting subunit Ku. DNA-PK is a major component of the nonhomologous end-joining pathway of DNA double-strand breaks repair. Although DNA-PK has been biochemically characterized in vitro, relatively little is known about its functions in the context of DNA repair and how its kinase activity is precisely regulated in vivo. Here, we report that cellular depletion of the individual catalytic subunits of protein kinase CK2 by RNA interference leads to significant cell death in M059K human glioblastoma cells expressing DNA-PKcs, but not in their isogenic counterpart, that is M059J cells, devoid of DNA-PKcs. The lack of CK2 results in enhanced DNA-PKcs activity and strongly inhibits DNA damage-induced autophosphorylation of DNA-PKcs at S2056 as well as repair of DNA double-strand breaks. By the application of the in situ proximity ligation assay, we show that CK2 interacts with DNA-PKcs in normal growing cells and that the association increases upon DNA damage. These results indicate that CK2 has an important role in the modulation of DNA-PKcs activity and its phosphorylation status providing important insights into the mechanisms by which DNA-PKcs is regulated in vivo.
- Published
- 2010
- Full Text
- View/download PDF
17. MdmX is a substrate for the deubiquitinating enzyme USP2a.
- Author
-
Allende-Vega N, Sparks A, Lane DP, and Saville MK
- Subjects
- Antineoplastic Agents pharmacology, Blotting, Western, Catalytic Domain genetics, Cell Cycle Proteins, Cell Line, Tumor, Cisplatin pharmacology, Down-Regulation drug effects, Endopeptidases genetics, Humans, Immunoprecipitation, Mutation, Nuclear Proteins genetics, Protein Binding, Proto-Oncogene Proteins genetics, Proto-Oncogene Proteins c-mdm2 genetics, RNA Interference, Reverse Transcriptase Polymerase Chain Reaction, Substrate Specificity, Transfection, Ubiquitin Thiolesterase, Ubiquitination, Endopeptidases metabolism, Nuclear Proteins metabolism, Proto-Oncogene Proteins metabolism, Proto-Oncogene Proteins c-mdm2 metabolism
- Abstract
It has previously been shown that ubiquitin-specific protease 2a (USP2a) is a regulator of the Mdm2/p53 pathway. USP2a binds to Mdm2 and can deubiquitinate Mdm2 without reversing Mdm2-mediated p53 ubiquitination. Overexpression of USP2a causes accumulation of Mdm2 and promotes p53 degradation. We now show that MdmX is also a substrate for USP2a. MdmX associates with USP2a independently of Mdm2. Ectopic expression of wild-type USP2a but not a catalytic mutant prevents Mdm2-mediated degradation of MdmX. This correlates with the ability of wild-type USP2a to deubiquitinate MdmX. siRNA-mediated knockdown of USP2a in NTERA-2 testicular embryonal carcinoma cells and MCF7 breast cancer cells causes destabilization of MdmX and results in a decrease in MdmX protein levels, showing that endogenous USP2a participates in the regulation of MdmX stability. The therapeutic drug, cisplatin decreases MdmX protein expression. USP2a mRNA and protein levels were also reduced after cisplatin exposure. The magnitude and time course of USP2a downregulation suggests that the reduction in USP2a levels could contribute to the decrease in MdmX expression following treatment with cisplatin. Knockdown of USP2a increases the sensitivity of NTERA-2 cells to cisplatin, raising the possibility that suppression of USP2a in combination with cisplatin may be an approach for cancer therapy.
- Published
- 2010
- Full Text
- View/download PDF
18. The Ste20-like kinase SLK is required for ErbB2-driven breast cancer cell motility.
- Author
-
Roovers K, Wagner S, Storbeck CJ, O'Reilly P, Lo V, Northey JJ, Chmielecki J, Muller WJ, Siegel PM, and Sabourin LA
- Subjects
- Animals, Blotting, Western, Breast Neoplasms genetics, Breast Neoplasms metabolism, Breast Neoplasms pathology, Catalytic Domain genetics, Cell Line, Cell Line, Tumor, Cell Movement drug effects, Cell Movement genetics, Chemotaxis drug effects, Focal Adhesion Kinase 1 genetics, Focal Adhesion Kinase 1 metabolism, HeLa Cells, Humans, Immunoprecipitation, Mitogen-Activated Protein Kinase Kinases antagonists & inhibitors, Mitogen-Activated Protein Kinase Kinases metabolism, Mutation, Neuregulin-1 pharmacology, Phosphatidylinositol 3-Kinases metabolism, Phosphoinositide-3 Kinase Inhibitors, Phospholipase C gamma antagonists & inhibitors, Phospholipase C gamma metabolism, Phosphorylation, Protein Binding, Protein Serine-Threonine Kinases genetics, RNA, Small Interfering genetics, Receptor, ErbB-2 genetics, Signal Transduction drug effects, Transfection, Tyrosine metabolism, Cell Movement physiology, Protein Serine-Threonine Kinases metabolism, Receptor, ErbB-2 metabolism
- Abstract
The Ste20-like kinase, SLK, is involved in the control of cell motility through its effects on actin reorganization and focal adhesion turnover. Here we investigated the role of SLK in chemotaxis downstream of the tyrosine kinase receptor, HER2/ErbB2/Neu, which is frequently overexpressed in human breast cancers. Our results show that SLK is required for the efficient cell migration of human and mouse mammary epithelial cell lines in the presence of the Neu activator, heregulin, as a chemoattractant. SLK activity is stimulated by heregulin treatment or by overexpression of activated Neu. Phosphorylation of tyrosine 1201 or tyrosines 1226/7 on Neu is a key event for SLK activation and cell migration, and cancer cell invasion mediated by these tyrosines is inhibited by kinase-inactive SLK. Signaling pathway inhibitors show that Neu-mediated SLK activation is dependent on MEK, PI3K, PLCgamma and Shc signaling. Furthermore, heregulin-stimulated SLK activity requires signals from the focal adhesion proteins, FAK and src. Finally, phospho-FAK analysis shows that SLK is required for Neu-dependent focal adhesion turnover. Together, these studies define an interaction between Neu and SLK signaling in the regulation of cancer cell motility.
- Published
- 2009
- Full Text
- View/download PDF
19. p120Ras-GAP binds the DLC1 Rho-GAP tumor suppressor protein and inhibits its RhoA GTPase and growth-suppressing activities.
- Author
-
Yang XY, Guan M, Vigil D, Der CJ, Lowy DR, and Popescu NC
- Subjects
- Catalytic Domain, Cell Line, Tumor, Cell Proliferation, GTPase-Activating Proteins, Humans, Tumor Suppressor Proteins analysis, Tumor Suppressor Proteins antagonists & inhibitors, Tumor Suppressor Proteins chemistry, p120 GTPase Activating Protein analysis, p120 GTPase Activating Protein chemistry, src Homology Domains, Neoplasms pathology, Tumor Suppressor Proteins physiology, p120 GTPase Activating Protein physiology, rhoA GTP-Binding Protein metabolism
- Abstract
DLC1 (deleted in liver cancer 1), which encodes a Rho GTPase-activating protein (Rho-GAP), is a potent tumor suppressor gene that is frequently inactivated in several human cancers. DLC1 is a multidomain protein that has been shown previously to bind members of the tensin gene family. Here we show that p120Ras-GAP (Ras-GAP; also known as RASA1) interacts and extensively colocalizes with DLC1 in focal adhesions. The binding was mapped to the SH3 domain located in the N terminus of Ras-GAP and to the Rho-GAP catalytic domain located in the C terminus of the DLC1. In vitro analyses with purified proteins determined that the isolated Ras-GAP SH3 domain inhibits DLC1 Rho-GAP activity, suggesting that Ras-GAP is a negative regulator of DLC1 Rho-GAP activity. Consistent with this possibility, we found that ectopic overexpression of Ras-GAP in a Ras-GAP-insensitive tumor line impaired the growth-suppressing activity of DLC1 and increased RhoA activity in vivo. Our observations expand the complexity of proteins that regulate DLC1 function and define a novel mechanism of the cross talk between Ras and Rho GTPases.1R01CA129610
- Published
- 2009
- Full Text
- View/download PDF
20. Transamidase site-targeted agents alter the conformation of the transglutaminase cancer stem cell survival protein to reduce GTP binding activity and cancer stem cell survival
- Author
-
David J. Weber, Abdullah Akbar, B Nance, Richard L. Eckert, Candace L. Kerr, Jeffrey W. Keillor, Henryk Szmacinski, Raquel Godoy-Ruiz, Eric A. Toth, Matthew L. Fisher, T Lok Wong, and Joseph R. Lakowicz
- Subjects
0301 basic medicine ,cancer stem cells ,squamous cell carcinoma ,Cancer Research ,GTP' ,Tissue transglutaminase ,Cell Survival ,Protein Conformation ,Antineoplastic Agents ,GTPase ,Biology ,Molecular oncology ,Article ,03 medical and health sciences ,Gene Knockout Techniques ,0302 clinical medicine ,Growth factor receptor ,Cancer stem cell ,GTP-Binding Proteins ,Catalytic Domain ,Genetics ,CP4d ,Humans ,cancer ,Protein Glutamine gamma Glutamyltransferase 2 ,Molecular Targeted Therapy ,Molecular Biology ,Cells, Cultured ,NC9 ,Binding Sites ,Transglutaminases ,VA4 ,VA5 ,Cell cycle ,Aminoacyltransferases ,Cell biology ,Transglutaminase 2 ,030104 developmental biology ,Biochemistry ,030220 oncology & carcinogenesis ,biology.protein ,Neoplastic Stem Cells ,Guanosine Triphosphate ,Intracellular ,Protein Binding - Abstract
Type 2 transglutaminase (TG2) is an important cancer stem cell survival protein that exists in open and closed conformations. The major intracellular form is the closed conformation that functions as a GTP-binding GTPase and is required for cancer stem cell survival. However, at a finite rate, TG2 transitions to an open conformation that exposes the transamidase catalytic site involved in protein-protein crosslinking. The activities are mutually exclusive, as the closed conformation has GTP binding/GTPase activity, and the open conformation transamidase activity. We recently showed that GTP binding, but not transamidase activity, is required for TG2-dependent cancer stem cell invasion, migration and tumour formation. However, we were surprised that transamidase site-specific inhibitors reduce cancer stem cell survival. We now show that compounds NC9, VA4 and VA5, which react exclusively at the TG2 transamidase site, inhibit both transamidase and GTP-binding activities. Transamidase activity is inhibited by direct inhibitor binding at the transamidase site, and GTP binding is blocked because inhibitor interaction at the transamidase site locks the protein in the extended/open conformation to disorganize/inactivate the GTP binding/GTPase site. These findings suggest that transamidase site-specific inhibitors can inhibit GTP binding/signalling by driving a conformation change that disorganizes the TG2 GTP binding to reduce TG2-dependent signalling, and that drugs designed to target this site may be potent anti-cancer agents.
- Published
- 2016
21. Novel evidences for a tumor suppressor role of Rev3, the catalytic subunit of Pol zeta.
- Author
-
Brondello JM, Pillaire MJ, Rodriguez C, Gourraud PA, Selves J, Cazaux C, and Piette J
- Subjects
- Catalytic Domain, Cells, Cultured, Checkpoint Kinase 2, Colonic Neoplasms metabolism, DNA Breaks, Double-Stranded, DNA Repair, DNA Replication, DNA-Binding Proteins analysis, DNA-Binding Proteins genetics, DNA-Directed DNA Polymerase analysis, DNA-Directed DNA Polymerase genetics, Gene Expression Regulation, Neoplastic, Histones metabolism, Humans, Phosphorylation, Protein Serine-Threonine Kinases physiology, RNA, Messenger analysis, S Phase, DNA-Binding Proteins physiology, DNA-Directed DNA Polymerase physiology, Tumor Suppressor Proteins physiology
- Abstract
Cell cycle checkpoints and DNA repair act in concert to ensure DNA integrity during perturbation of normal replication or in response to genotoxic agents. Deficiencies in these protective mechanisms can lead to cellular transformation and ultimately tumorigenesis. Here we focused on Rev3, the catalytic subunit of the low-fidelity DNA repair polymerase zeta. Rev3 is believed to play a role in double-strand break (DSB)-induced DNA repair by homologous recombination. In line with this hypothesis, we show the accumulation of chromatin-bound Rev3 protein in late S-G2 of untreated cells and in response to clastogenic DNA damage as well as an gamma-H2AX accumulation in Rev3-depleted cells. Moreover, serine 995 of Rev3 is in vitro phosphorylated by the DSB-inducible checkpoint kinase, Chk2. Our data also disclose a significant reduction of rev3 gene expression in 74 colon carcinomas when compared to the normal adjacent tissues. This reduced expression is independent of the carcinoma stages, suggesting that the downregulation of rev3 might have occurred early during tumorigenesis.
- Published
- 2008
- Full Text
- View/download PDF
22. Protein kinase CK2 interacts with the splicing factor hPrp3p.
- Author
-
Lehnert S, Götz C, Kartarius S, Schäfer B, and Montenarh M
- Subjects
- Amino Acid Sequence, Casein Kinase II genetics, Catalytic Domain, Cell Cycle, Databases, Protein, HeLa Cells, Holoenzymes metabolism, Humans, Molecular Sequence Data, Nuclear Proteins chemistry, Nuclear Proteins genetics, Phosphorylation, Protein Binding, RNA Splicing genetics, Ribonucleoprotein, U4-U6 Small Nuclear chemistry, Ribonucleoprotein, U4-U6 Small Nuclear genetics, Spliceosomes metabolism, Casein Kinase II metabolism, Nuclear Proteins metabolism, Ribonucleoprotein, U4-U6 Small Nuclear metabolism
- Abstract
Numerous signalling pathways in cells are influenced by the ubiquitous Ser/Thr protein kinase CK2. Protein kinase CK2 is composed of two regulatory beta-subunits and two catalytic alpha- or alpha'-subunits. Several of the known CK2 substrates are proteins known to regulate transcriptional events. Here, we describe that protein kinase CK2 interacts with the splicing factor hPrp3p, which is important for the assembly of the spliceosome. In a two-hybrid screen hPrp3p is exclusively bound to the catalytic alpha- or alpha'-subunits of CK2 but not to the regulatory beta-subunit. The interaction was confirmed by coimmunoprecipitation experiments in vitro and in vivo. Moreover, both proteins colocalized in nuclear speckles which is typical for splicing factor compartments within the nucleus. Phosphorylation experiments revealed that hPrp3p is also a substrate of protein kinase CK2. The main phosphorylation site was mapped to C-terminal residues. In vitro and in vivo splicing assays showed that the splicing activity is significantly influenced by the CK2-hPrp3p interaction. Thus, these data showed that CK2 is involved in the regulation of RNA processing.
- Published
- 2008
- Full Text
- View/download PDF
23. Disruption of the retinoblastoma pathway by small interfering RNA and ectopic expression of the catalytic subunit of telomerase lead to immortalization of human ovarian surface epithelial cells.
- Author
-
Yang G, Rosen DG, Colacino JA, Mercado-Uribe I, and Liu J
- Subjects
- Base Sequence, Catalytic Domain, Epithelial Cells, Female, Humans, Karyotyping, Models, Biological, Molecular Sequence Data, Ovarian Neoplasms drug therapy, Ovarian Neoplasms metabolism, Retinoblastoma Protein genetics, Cell Line, Transformed, Ovary, RNA, Small Interfering pharmacology, Retinoblastoma Protein metabolism, Telomerase metabolism
- Abstract
The risk of developing ovarian cancer is about 1% over a lifetime, but it is the most deadly gynecologic cancer, in part due to lack of diagnostic markers for early-stage disease and cell model system for studying early neoplastic changes. Most existing immortal human ovarian surface epithelial cells were achieved by using viral protein such as SV40 T/t antigen or E6/E7, which inactivate multiple cellular pathways. In the current study, we used a small interfering RNA (siRNA) against the retinoblastoma gene (pRb) and ectopic expression of human telomerase reverse transcriptase (hTERT) to immortalize the primary ovarian epithelial cell line OSE137 and two additional human ovarian surface epithelial cells. The immortalized OSE137 showed increased telomerase activity, lengthened telomeres, increased G2/M phase, altered cell-cycle regulatory proteins but nontumorigenic. As both Rb and hTERT pathways are commonly altered in human ovarian cancer and these genetic changes are faithfully modeled in these cells without using viral protein, these immortal cells represent an authentic in vitro model system with which to study the initiation and progression of human ovarian cancer.
- Published
- 2007
- Full Text
- View/download PDF
24. hTERT: a novel endogenous inhibitor of the mitochondrial cell death pathway.
- Author
-
Massard C, Zermati Y, Pauleau AL, Larochette N, Métivier D, Sabatier L, Kroemer G, and Soria JC
- Subjects
- Antineoplastic Agents pharmacology, Catalytic Domain, Cell Line, Tumor, Cell Survival, Cisplatin pharmacology, Colonic Neoplasms pathology, DNA-Binding Proteins metabolism, Etoposide pharmacology, Female, Humans, Mitomycin pharmacology, Reactive Oxygen Species, Telomerase metabolism, Uterine Cervical Neoplasms pathology, bcl-2-Associated X Protein metabolism, fas Receptor biosynthesis, Apoptosis, DNA-Binding Proteins physiology, Mitochondria metabolism, Telomerase physiology
- Abstract
hTERT is the catalytic subunit of the telomerase and is hence required for telomerase maintenance activity and cancer cell immortalization. Here, we show that acute hTERT depletion has no adverse effects on the viability or proliferation of cervical and colon carcinoma cell lines, as evaluated within 72 h after transfection with hTERT-specific small interfering RNAs (siRNAs). Within the same time frame, hTERT depletion facilitated the induction of apoptotic cell death by cisplatin, etoposide, mitomycin C and reactive oxygen species, yet failed to sensitize cells to death induction via the CD95 death receptor. Experiments performed with p53 knockout cells or chemical p53 inhibitors revealed that p53 was not involved in the chemosensitizing effect of hTERT knockdown. However, the proapoptotic Bcl-2 family protein Bax was involved in cell death induction by hTERT siRNAs. Depletion of hTERT facilitated the conformational activation of Bax induced by genotoxic agents. Moreover, Bax knockout abolished the chemosensitizing effect of hTERT siRNAs. Inhibition of mitochondrial membrane permeabilization by overexpression of Bcl-2 or expression of the cytomegalovirus-encoded protein vMIA (viral mitochondrial inhibitor of apoptosis), which acts as a specific Bax inhibitor, prevented the induction of cell death by the combination of hTERT depletion and chemotherapeutic agents. Altogether, our data indicate that hTERT inhibition may constitute a promising strategy for facilitating the induction of the mitochondrial pathway of apoptosis.
- Published
- 2006
- Full Text
- View/download PDF
25. Smad6 is a protein kinase X phosphorylation substrate and is required for HL-60 cell differentiation.
- Author
-
Glesne D and Huberman E
- Subjects
- Active Transport, Cell Nucleus drug effects, Carcinogens pharmacology, Catalytic Domain, Cell Adhesion drug effects, Cell Nucleus metabolism, Cyclic AMP-Dependent Protein Kinases, Gene Expression Regulation drug effects, HL-60 Cells, Humans, Phosphorylation drug effects, Promoter Regions, Genetic, Protein Binding, RNA Interference, Tetradecanoylphorbol Acetate pharmacology, Two-Hybrid System Techniques, Cell Differentiation drug effects, Macrophages enzymology, Protein Processing, Post-Translational drug effects, Protein Serine-Threonine Kinases metabolism, Signal Transduction drug effects, Smad6 Protein metabolism
- Abstract
To gain insight into the function of human protein kinase X (PrKX), a signal-transduction protein required for macrophage differentiation, we identified regulatory subunit I alpha of protein kinase A, T54 and Smad6 as partners for this protein using a yeast two-hybrid interaction screen. Interactions between PrKX and these proteins were substantiated by co-immunoprecipitation. Interaction between Smad6 and PrKX was also confirmed in human myeloid HL-60 cells following their phorbol 12-myristate 13-acetate (PMA)-induced differentiation into macrophages. In vitro phosphorylation assays demonstrated that PrKX phosphorylates Smad6 at a serine residue. Mutagenesis of this site resulted in abrogation of PrKX phosphorylation. Both PrKX and Smad6 were shown to be co-localized to the nuclear compartment of HL-60 cells during their macrophage differentiation where PrKX levels are induced and Smad6 protein levels remain relatively constant while levels of serine phosphorylation of Smad6 increase. By using in vitro electrophoretic mobility shift assays and in vivo chromatin immunoprecipitation, we also demonstrate that during macrophage differentiation Smad6 displays an increased binding to the human osteopontin, Id2, and Hex gene promoters, which correlates to an observed increased expression of these genes. Finally, vector-based RNA interference experiments established that both Smad6 and PrKX proteins are required for PMA-induced cell attachment and spreading.
- Published
- 2006
- Full Text
- View/download PDF
26. Unique and overlapping substrate specificities of caspase-8 and caspase-10.
- Author
-
Fischer U, Stroh C, and Schulze-Osthoff K
- Subjects
- Amino Acid Sequence, Apoptosis, BH3 Interacting Domain Death Agonist Protein metabolism, Caspase 10, Caspase 8, Catalytic Domain, Cell Line, Tumor, Humans, Jurkat Cells, Models, Genetic, Molecular Sequence Data, Protein Binding, Reverse Transcriptase Polymerase Chain Reaction, Signal Transduction, Substrate Specificity, Caspases chemistry, Caspases metabolism, Gene Expression Regulation, Enzymologic, Gene Expression Regulation, Neoplastic
- Abstract
Although caspase-8 has an established role as an initiator of death receptor-mediated apoptosis, the function of its closest homolog, caspase-10, is almost completely unknown. To gain a closer insight into the physiological function of caspase-10, we compared the cleavage of known caspase-8 substrates by both initiator caspases. We demonstrate that caspase-10 and -8 have overlapping cleavage preferences for several substrates such as the kinases RIP and PAK2. Interestingly, in other substrates, such as the Bcl-2 protein Bid, we found additional and distinct cleavage sites for both caspases, which might have important consequences for mitochondrial targeting and propagation of the death signal. Caspase-8 and -10 also caused different interchain cleavage patterns of their enzyme precursors. Together, these results suggest that caspase-8 and -10, despite having overlapping functions, also have selective substrate cleavage specificities and might thereby exert nonredundant roles in apoptosis signaling.
- Published
- 2006
- Full Text
- View/download PDF
27. The 'regulatory' beta-subunit of protein kinase CK2 negatively influences p53-mediated allosteric effects on Chk2 activation.
- Author
-
Bjørling-Poulsen M, Siehler S, Wiesmüller L, Meek D, Niefind K, and Issinger OG
- Subjects
- Allosteric Regulation, Animals, COS Cells, Catalytic Domain, Checkpoint Kinase 2, Chlorocebus aethiops, Enzyme Activation, Humans, Phosphorylation, Protein Binding, Transfection, Casein Kinase II metabolism, Casein Kinase II physiology, Genes, p53, Protein Serine-Threonine Kinases metabolism, Tumor Suppressor Protein p53 metabolism
- Abstract
The 'regulatory' beta-subunit of protein kinase CK2 has previously been shown to interact with protein kinases such as A-Raf, c-Mos, Lyn and Chk1 in addition to the catalytic subunit of CK2. Sequence alignments suggest that these interactions have a structural basis, and hence other protein kinases harboring corresponding sequences may be potential interaction partners for CK2beta. We show here that Chk2 specifically interacts with CK2beta in vitro and in cultured cells, and that activation of Chk2 leads to a reduction of this interaction. Additionally, we show that the presence of the CK2beta-subunit significantly reduces the Chk2-catalysed phosphorylation of p53 in vitro. These findings support the notion that CK2beta can act as a general modulator of remote docking sites in protein kinase--substrate interactions.
- Published
- 2005
- Full Text
- View/download PDF
28. Disease associated mutations at valine 804 in the RET receptor tyrosine kinase confer resistance to selective kinase inhibitors.
- Author
-
Carlomagno F, Guida T, Anaganti S, Vecchio G, Fusco A, Ryan AJ, Billaud M, and Santoro M
- Subjects
- Catalytic Domain, Cell Line, Drug Resistance, Mitogens pharmacology, Piperidines pharmacology, Proto-Oncogene Proteins c-ret, Pyrazoles pharmacology, Pyrimidines pharmacology, Quinazolines pharmacology, Signal Transduction, Valine genetics, Carcinoma, Medullary genetics, Enzyme Inhibitors pharmacology, Point Mutation, Proto-Oncogene Proteins antagonists & inhibitors, Proto-Oncogene Proteins genetics, Receptor Protein-Tyrosine Kinases antagonists & inhibitors, Receptor Protein-Tyrosine Kinases genetics, Thyroid Neoplasms genetics
- Abstract
We have recently demonstrated that the pyrazolopyrimidines PP1 and PP2 and the 4-anilinoquinazoline ZD6474 display a strong inhibitory activity (IC(50)< or =100 nM) towards constitutively active oncogenic RET kinases. Here, we show that most oncogenic MEN2-associated RET kinase mutants are highly susceptible to PP1, PP2 and ZD6474 inhibition. In contrast, MEN2-associated swap of bulky hydrophobic leucine or methionine residues for valine 804 in the RET kinase domain causes resistance to the three compounds. Substitution of valine 804 with the small amino- acid glycine renders the RET kinase even more susceptible to inhibition (ZD6474 IC(50): 20 nM) than the wild-type kinase. Our data identify valine 804 of RET as a structural determinant mediating resistance to pyrazolopyrimidines and 4-anilinoquinazolines.
- Published
- 2004
- Full Text
- View/download PDF
29. Role of the Brk SH3 domain in substrate recognition.
- Author
-
Qiu H and Miller WT
- Subjects
- Adaptor Proteins, Signal Transducing, Baculoviridae genetics, Binding Sites, Catalytic Domain physiology, Cell Line, DNA-Binding Proteins, Gene Expression Regulation, Neoplastic, Humans, Kidney cytology, Kidney embryology, Kinetics, Ligands, Mutagenesis, Site-Directed, Neoplasm Proteins, Peptides chemistry, Peptides metabolism, Phosphorylation, Protein Structure, Tertiary, Protein-Tyrosine Kinases genetics, RNA-Binding Proteins metabolism, Substrate Specificity, Protein-Tyrosine Kinases chemistry, Protein-Tyrosine Kinases metabolism, src Homology Domains physiology
- Abstract
Breast tumor kinase (Brk) is a nonreceptor tyrosine kinase that is overexpressed in a high percentage of breast carcinomas. Brk contains SH3, SH2, and tyrosine kinase catalytic domains in a similar arrangement as Src family kinases. In this study, we explored the roles of the SH3 and SH2 domains in Brk regulation and substrate binding. We introduced a series of mutations into Brk that were predicted to disrupt the intramolecular interactions involving the SH3 and SH2 domains. These mutant forms of Brk displayed higher activity than wild-type Brk when expressed in human embryonic kidney HEK293 cells. These studies also allowed us to pinpoint the intramolecular binding site for the SH3 domain. To examine substrate binding, we compared binding and phosphorylation of Sam68, a physiological substrate of Brk. These experiments showed that the SH3 domain plays a particularly important role in substrate recognition. We confirmed this conclusion using a series of synthetic peptides in which a substrate sequence was coupled to an SH3 or SH2 ligand. The SH3-binding substrate had a significantly lower K(m) than a control, while no difference was observed between an SH2-binding substrate and a control. Taken together, our data suggest that SH3 interactions will govern phosphorylation of many substrates by Brk.
- Published
- 2004
- Full Text
- View/download PDF
30. Phosphorylation of p53 at serine 37 is important for transcriptional activity and regulation in response to DNA damage.
- Author
-
Dohoney KM, Guillerm C, Whiteford C, Elbi C, Lambert PF, Hager GL, and Brady JN
- Subjects
- Catalytic Domain, Cell Line, Gamma Rays, Humans, Okadaic Acid pharmacology, Phosphoprotein Phosphatases chemistry, Phosphoprotein Phosphatases physiology, Phosphorylation, Protein Phosphatase 2, Serine, Tumor Suppressor Protein p53 chemistry, DNA Damage, Transcription, Genetic, Tumor Suppressor Protein p53 metabolism
- Abstract
The p53 tumor suppressor protein plays a critical role in mediating cellular response to stress. Upon DNA damage, post-translational modifications stabilize and activate this nuclear phosphoprotein. To determine the effect of phosphorylation site mutants in the context of the whole p53 protein, we performed reporter assays in p53 and MDM2 knockout mouse embryonic fibroblasts transfected with full-length p53 constructs. We show that mutation of S37 causes a decrease in p53 transcriptional activity compared to wild-type p53. Our data further suggest that the dephosphorylation of p53 at S37 is a regulated event involving protein phosphatase 2A (PP2A). Coimmunoprecipitation and immunofluorescence microscopy studies demonstrate that PP2A and p53 associate with one another in vivo following gamma-irradiation. Consistent with these observations, phosphorylated S37 accumulates in cell extracts prepared from gamma-irradiated Molt-4 cells in the presence of okadaic acid. Furthermore, in vitro phosphatase assays show that PP2A dephosphorylates p53 at S37. These results suggest that dephosphorylation of p53 at S37 plays a role in the transcriptional regulation of the p53 protein in response to DNA damage.
- Published
- 2004
- Full Text
- View/download PDF
31. Modulation of human checkpoint kinase Chk1 by the regulatory beta-subunit of protein kinase CK2.
- Author
-
Guerra B, Issinger OG, and Wang JY
- Subjects
- 3T3 Cells, Animals, COS Cells, Casein Kinase II, Catalytic Domain physiology, Checkpoint Kinase 1, Enzyme Activation physiology, Humans, Mice, Phosphorylation, Protein Binding, Protein Conformation, Protein Kinases chemistry, Protein Serine-Threonine Kinases chemistry, Cell Cycle physiology, Protein Kinases metabolism, Protein Serine-Threonine Kinases metabolism
- Abstract
Protein kinase CK2 is a serine/threonine protein kinase involved in various aspects of cellular regulation. The regulatory beta-subunit of CK2 exerts a central role not only in mediating formation of tetrameric CK2 complexes but also as a docking partner for several protein kinases. In this study, CK2beta is found to interact with the human cell cycle checkpoint kinase Chk1. The Chk1-interacting region of CK2beta is localized at the C-terminus and the complex between CK2beta and Chk1 is devoid of the catalytic CK2alpha-subunit. The interaction between CK2beta and Chk1 leads to an increase in the Cdc25C phosphorylation activity of Chk1. The screening of several cell lines has revealed that the association between CK2beta and Chk1 also occurs in vivo at a different degree. Collectively, these studies confirm the implication of the regulatory beta-subunit of protein kinase CK2 in cell cycle regulation and identify a novel mechanism for the activation of Chk1 protein kinase.
- Published
- 2003
- Full Text
- View/download PDF
32. Cysteine residues in the C-terminal lobe of Src: their role in the suppression of the Src kinase.
- Author
-
Oo ML, Senga T, Thant AA, Amin AR, Huang P, Mon NN, and Hamaguchi M
- Subjects
- Alkylating Agents pharmacology, Allosteric Regulation, Amino Acid Sequence, Animals, Avian Sarcoma Viruses enzymology, Avian Sarcoma Viruses genetics, COS Cells, CSK Tyrosine-Protein Kinase, Catalysis, Catalytic Domain, Chlorocebus aethiops, Codon, Drug Resistance, Enzyme Inhibitors pharmacology, Maleimides pharmacology, Molecular Sequence Data, Oncogene Protein pp60(v-src) antagonists & inhibitors, Phosphotyrosine chemistry, Protein-Tyrosine Kinases antagonists & inhibitors, Recombinant Fusion Proteins chemistry, Sequence Alignment, Sequence Homology, Amino Acid, Structure-Activity Relationship, src Homology Domains, src-Family Kinases antagonists & inhibitors, src-Family Kinases chemistry, Cysteine chemistry, Oncogene Protein pp60(v-src) chemistry, Protein-Tyrosine Kinases chemistry
- Abstract
To evaluate the function of cysteine residues of the Src kinase, we constructed a series of Src mutants in which some of cysteines were replaced to alanines. With these mutants, we studied the effect of SH-alkylating agents, N-[p-(2-benzimidazolyl)phenyl] maleimide (BIPM) and N-(9-acridinyl) maleimide (NAM), on their kinase activity. Of 10 cysteine residues scattered over v-Src, either a single mutation at Cys520 or multiple mutations at the four clustered cyteines, Cys483, Cys487, Cys496 and Cys498, yielded clear resistance to the treatment with 10 microM BIPM or 1 microM NAM. In contrast, other cysteines including those in the SH2 domain and those in the catalytic cleft of the kinase domain were dispensable for the inactivation by BIPM and NAM. Similarly, deletion of SH2 and SH3 did not confer the resistance to v-Src, suggesting the inactivation by the SH-alkylating agents is SH2/SH3-independent. Although Cys520-mutated v-Src was resistant to 1 microM NAM, it was inactivated by 5 microM NAM. However, combined mutation including all of Cys483, Cys487, Cys496, Cys498 and Cys520 yielded clear resistance to 5 microM NAM. Among these mutants, those with double mutations in the four clustered cysteines yielded a temperature sensitive phenotype in the transfected cells, whereas Cys520 did not, suggesting that Cys520 has, at least in part, a discrete function. In contrast to v-Src, c-Src, which lacks cysteine at position 520, was resistant to 1 microM NAM but sensitive to 5 microM NAM. While replacement of Phe520 of c-Src to cysteine made it sensitive to 1 microM NAM, double mutation in clustered cysteines again yielded resistance to 5 microM NAM. Taken together, our results strongly suggest that the multiple cysteine residues clustered at the end of the C-terminal lobe are critical for the inhibition by the SH-alkylating agents and, thereby, have an allosteric repressor effect on the catalytic activity of Src in a SH2-phosphoTyr527 independent manner.
- Published
- 2003
- Full Text
- View/download PDF
33. Effect of tyrosine kinase inhibitor STI571 on the kinase activity of wild-type and various mutated c-kit receptors found in mast cell neoplasms.
- Author
-
Zermati Y, De Sepulveda P, Féger F, Létard S, Kersual J, Castéran N, Gorochov G, Dy M, Ribadeau Dumas A, Dorgham K, Parizot C, Bieche Y, Vidaud M, Lortholary O, Arock M, Hermine O, and Dubreuil P
- Subjects
- Benzamides, Catalytic Domain genetics, Cell Line, Humans, Imatinib Mesylate, Mastocytosis, Systemic genetics, Protein-Tyrosine Kinases antagonists & inhibitors, Structure-Activity Relationship, Antineoplastic Agents pharmacology, Enzyme Inhibitors pharmacology, Mastocytosis, Systemic drug therapy, Piperazines pharmacology, Proto-Oncogene Proteins c-kit drug effects, Proto-Oncogene Proteins c-kit genetics, Pyrimidines pharmacology
- Abstract
Systemic mastocytosis (SM) is a rare disease caused by an abnormal mast cell accumulation in various tissues. Two classes of constitutive activating c-kit mutations are found in SM. The most frequent class occurs in the catalytic pocket coding region with substitutions at codon 816 and the other in the intracellular juxtamembrane coding region. Therefore, kinase inhibitors that block mutated c-kit activity might be used as therapeutic agents in SM. Here, we show that STI571 inhibits both wild-type and juxtamembrane mutant c-kit kinase activity, but has no effect on the activity of the D816 V mutant. Accordingly, STI571 selectively decreases the survival of normal mast cell and of mast cell lines either with juxtamembrane c-kit mutations, but not that of tumoral mast cell from patient with SM or of mast cell lines with the D816 V mutation. Therefore, STI571 is not a good candidate to treat SM and specific kinase inhibitors should be designed to inhibit constitutive activating mutations at codon 816.
- Published
- 2003
- Full Text
- View/download PDF
34. Uracil in DNA--occurrence, consequences and repair.
- Author
-
Krokan HE, Drabløs F, and Slupphaug G
- Subjects
- Base Pair Mismatch, Catalytic Domain, DNA chemistry, Endodeoxyribonucleases genetics, Evolution, Molecular, Humans, N-Glycosyl Hydrolases chemistry, N-Glycosyl Hydrolases genetics, Uracil chemistry, Uracil-DNA Glycosidase, DNA physiology, DNA Glycosylases, DNA Repair physiology, Endodeoxyribonucleases metabolism, N-Glycosyl Hydrolases metabolism, Thymine DNA Glycosylase, Uracil physiology
- Abstract
Uracil in DNA results from deamination of cytosine, resulting in mutagenic U : G mispairs, and misincorporation of dUMP, which gives a less harmful U : A pair. At least four different human DNA glycosylases may remove uracil and thus generate an abasic site, which is itself cytotoxic and potentially mutagenic. These enzymes are UNG, SMUG1, TDG and MBD4. The base excision repair process is completed either by a short patch- or long patch pathway, which largely use different proteins. UNG2 is a major nuclear uracil-DNA glycosylase central in removal of misincorporated dUMP in replication foci, but recent evidence also indicates an important role in repair of U : G mispairs and possibly U in single-stranded DNA. SMUG1 has broader specificity than UNG2 and may serve as a relatively efficient backup for UNG in repair of U : G mismatches and single-stranded DNA. TDG and MBD4 may have specialized roles in the repair of U and T in mismatches in CpG contexts. Recently, a role for UNG2, together with activation induced deaminase (AID) which generates uracil, has been demonstrated in immunoglobulin diversification. Studies are now underway to examine whether mice deficient in Ung develop lymphoproliferative malignancies and have a different life span.
- Published
- 2002
- Full Text
- View/download PDF
35. The distinct capacity of Fyn and Lck to phosphorylate Sam68 in T cells is essentially governed by SH3/SH2-catalytic domain linker interactions.
- Author
-
Feuillet V, Semichon M, Restouin A, Harriague J, Janzen J, Magee A, Collette Y, and Bismuth G
- Subjects
- Adaptor Proteins, Signal Transducing, Catalytic Domain, DNA-Binding Proteins, Humans, Jurkat Cells, Phosphorylation, Proto-Oncogene Proteins c-fyn, Substrate Specificity, src Homology Domains, Lymphocyte Specific Protein Tyrosine Kinase p56(lck) metabolism, Proto-Oncogene Proteins metabolism, RNA-Binding Proteins metabolism, T-Lymphocytes metabolism
- Abstract
Sam68 phosphorylation correlates with Fyn but not Lck expression in T cells. This substrate has been used here to explore the possible basis of the specificity of Fyn versus Lck. We show that this specificity is not based on a spatial segregation of the two kinases, since a chimeric Lck molecule containing the membrane anchoring domain of Fyn does not phosphorylate Sam68. Moreover, a Sam68 molecule targeted to the plasma membrane by the farnesylation signal of c-Ha-Ras remains poorly phosphorylated by Lck. In T cells, Fyn appears to be the active Src kinase in rafts, but Sam68 is not expressed in rafts, and its distinct phosphorylation by Fyn and Lck is not affected by raft dispersion. The Fyn/Lck specificity does not reflect a higher kinase activity of Fyn in general, as both Fyn and Lck are similarly recognized by an anti-active Src antibody. Both also strongly phosphorylate another Src substrate in vivo. Mainly, Lck phosphorylates Sam68 when the interaction between the SH3 domain and the SH2-catalytic domain linker is altered in heterologous Src molecules or after mutating key residues in the linker that increase the accessibility of the SH3 domain. Thus, the distinct potential of Fyn and Lck to phosphorylate Sam68 is likely controlled by the interaction of the kinase SH3 domain with the linker and Sam68, possibly on a competitive binding basis.
- Published
- 2002
- Full Text
- View/download PDF
36. Negative regulation of urokinase-type plasminogen activator production through FGF-2-mediated activation of phosphoinositide 3-kinase.
- Author
-
Mochizuki Y, Tsuda S, Kanetake H, and Kanda S
- Subjects
- Animals, Catalytic Domain, Enzyme Activation, Mice, Mitogen-Activated Protein Kinases physiology, Phosphatidylinositol 3-Kinases chemistry, Phosphoinositide-3 Kinase Inhibitors, ras Proteins physiology, Fibroblast Growth Factor 2 physiology, Phosphatidylinositol 3-Kinases physiology, Urokinase-Type Plasminogen Activator biosynthesis
- Abstract
Activation of phosphoinositide 3-kinase (PI3-kinase) is involved in many cellular responses. FGF-2 is one of the potent inducers of urokinase-type plasminogen activator (uPA) production in endothelial cells. However, little is known about the molecular mechanisms underlying FGF-2-mediated uPA production. Here we examined the signal transduction pathways involved in the regulation of uPA production by FGF-2-treatment. FGF-2 potently upregulated uPA production in murine brain capillary endothelial cells (IBE cells), as well as porcine aortic endothelial (PAE) cells and L6 myoblasts ectopically expressing FGFR1. PI3-kinase inhibitors, wortmannin and LY294002, both enhanced FGF-2-dependent uPA production by these cells. Stable expression of activated mutant p110alpha catalytic subunit of PI3-kinase into IBE cells decreased FGF-2-mediated uPA production, suggesting that PI3-kinase exhibited the negative regulatory effect on uPA production. No increase in FGF-2-induced PI3-kinase activity was observed in proteins immunoprecipitated by anti-phosphotyrosine antibody. Although stable expression of deleted mutant p85alpha regulatory subunit, which lacks association with p110 catalytic subunit, in IBE cells showed no dominant negative effect, transient expression of dominant negative Ras inhibited FGF-2-mediated PI3-kinase activation. These results suggest that only activated Ras contributed the FGF-2-mediated PI3-kinase activation. In cells stably expressing mutant p85alpha subunit, FGF-2 efficiently induced uPA production. Taken together, activation of PI3-kinase by FGF-2 is Ras-dependent and results in down-regulation of uPA production.
- Published
- 2002
- Full Text
- View/download PDF
37. Motif analysis of the tumor suppressor gene MMAC/PTEN identifies tyrosines critical for tumor suppression and lipid phosphatase activity.
- Author
-
Koul D, Jasser SA, Lu Y, Davies MA, Shen R, Shi Y, Mills GB, and Yung WK
- Subjects
- Amino Acid Motifs, Animals, Binding Sites, Catalytic Domain, Cell Division, Central Nervous System Neoplasms enzymology, Central Nervous System Neoplasms pathology, Genes, Tumor Suppressor, Glioma enzymology, Glioma pathology, Humans, Kinetics, Mice, Mice, Nude, Mutation, Neoplasms pathology, PTEN Phosphohydrolase, Phosphoric Monoester Hydrolases genetics, Protein Structure, Tertiary, Sequence Analysis, Protein, Tumor Cells, Cultured, Tumor Suppressor Proteins genetics, Neoplasms enzymology, Phosphoric Monoester Hydrolases chemistry, Phosphoric Monoester Hydrolases metabolism, Phosphotyrosine metabolism, Tumor Suppressor Proteins chemistry, Tumor Suppressor Proteins metabolism
- Abstract
The tumor suppressor gene, MMAC/PTEN, has phosphatase, C2, and PDZ-binding domains as well as potential sites of regulation by phosphorylation, including tyrosine phosphorylation, which may contribute to its ability to modulate cell growth and viability. Several obvious and significant motifs were found in MMAC/PTEN, including most notably, a catalytic domain of tyrosine phosphatase (IHCxxGxxRS/T) and several potential tyrosine phosphorylation sites. To examine the functional significance of tyrosine phosphorylation of MMAC/PTEN, retroviral constructs were generated with mutations at two putative tyrosine phosphorylation sites (Y240A/Y240F and Y315A/Y315F). Stable expression of wild-type MMAC/PTEN in U251 human glioma cells (which do not normally produce a functional MMAC/PTEN gene product) resulted in a significant reduction of tumor growth in nude mice, decreased growth rate, saturation density, and colony formation in vitro, as well as dephosphorylation of D3-phosphorylated phosphatidylinositols (PtdIns) in vitro. Mutation of Y240 or Y315 to either alanine or phenylalanine abrogated the ability of MMAC/PTEN to alter growth rate, saturation density, and colony formation in vitro. The ability of MMAC/PTEN to limit tumor growth in nude mice was markedly decreased but not abrogated by mutation of Y240 or Y315 to alanine. Thus, Y240 and Y315 are required for MMAC/PTEN to decrease tumor growth in vitro and in vivo. In contrast to wild-type MMAC/PTEN, mutant MMAC/PTEN containing Y240A or Y315A was unable to dephosphorylate D3-phosphorylated PtdIns in vitro. Thus, Y240A and Y315A are involved in the ability of MMAC/PTEN to dephosphorylate PtdIns and regulate tumor cell growth in vitro and in vivo.
- Published
- 2002
- Full Text
- View/download PDF
38. Comparison of human mammary epithelial cells immortalized by simian virus 40 T-Antigen or by the telomerase catalytic subunit.
- Author
-
Toouli CD, Huschtscha LI, Neumann AA, Noble JR, Colgin LM, Hukku B, and Reddel RR
- Subjects
- Adult, Aneuploidy, Antigens, Polyomavirus Transforming genetics, Catalytic Domain, Cell Differentiation, Cell Line, Transformed, Cell Survival, Chromosome Aberrations, Chromosomes, Human ultrastructure, Cyclin-Dependent Kinase Inhibitor p16 deficiency, Cyclin-Dependent Kinase Inhibitor p16 physiology, DNA drug effects, DNA Damage, Dactinomycin pharmacology, Epithelial Cells enzymology, Epithelial Cells pathology, Epithelial Cells virology, Female, Genes, p16, Humans, Intercalating Agents pharmacology, Karyotyping, Protein Subunits, Recombinant Fusion Proteins chemistry, Recombinant Fusion Proteins physiology, Simian virus 40 genetics, Telomerase chemistry, Telomerase genetics, Telomere ultrastructure, Transfection, Antigens, Polyomavirus Transforming physiology, Breast cytology, Cell Transformation, Neoplastic, Cell Transformation, Viral, Simian virus 40 physiology, Telomerase physiology
- Abstract
We directly compared two methods of immortalizing human mammary epithelial cells (HMECs). Cells were transfected with an expression plasmid either for hTERT, the catalytic subunit of telomerase, or for the simian virus 40 (SV40) early region genes. Under standard culture conditions, HMECs were not immortalized by hTERT unless they had spontaneously ceased expression of the p16(INK4a) tumor suppressor gene. Untransfected HMECs had low levels of telomerase expression, and immortalization by both methods was associated with an increase in telomerase activity and prevention of telomere shortening. SV40-induced immortalization was accompanied by aberrant differentiation, loss of DNA damage response, karyotypic instability and, in some cases, tumorigenicity. hTERT-immortalized cells had fewer karyotypic changes, but had intact DNA damage responses, and features of normal differentiation. Although SV40-immortalized cells are useful for studies of carcinogenesis, hTERT-immortalized cells retain more properties of normal cells.
- Published
- 2002
- Full Text
- View/download PDF
39. Cdc25B activity is regulated by 14-3-3.
- Author
-
Forrest A and Gabrielli B
- Subjects
- 14-3-3 Proteins, Binding Sites, Catalytic Domain, Cell Cycle Proteins chemistry, Cell Cycle Proteins genetics, Cyclin-Dependent Kinases metabolism, Cyclins metabolism, G2 Phase, HeLa Cells, Humans, Mutation, Protein Structure, Tertiary, Serine genetics, cdc25 Phosphatases chemistry, cdc25 Phosphatases genetics, Cell Cycle Proteins metabolism, Tyrosine 3-Monooxygenase physiology, cdc25 Phosphatases metabolism
- Abstract
In the G2 phase cell cycle checkpoint arrest, the cdc25-dependent activation of cyclin B/cdc2, a critical step in regulating entry into mitosis, is blocked. Studies in yeast have demonstrated that the inhibition of cdc25 function involves 14-3-3 binding to cdc25. In humans, two cdc25 isoforms have roles in G2/M progression, cdc25B and cdc25C, both bind 14-3-3. Abrogating 14-3-3 binding to cdc25C attenuates the G2 checkpoint arrest, but the contribution of 14-3-3 binding to the regulation of cdc25B function is unknown. Here we demonstrate that high level over-expression of cdc25B in G2 checkpoint arrested cells can activate cyclin B/cdc2 and overcome the checkpoint arrest. Mutation of the major 14-3-3 binding site, S323, or removal of the N-terminal regulatory domain are strong activating mutations, increasing the efficiency with which the mutant forms of cdc25B not only overcome the arrest, but also initiate aberrant mitosis. We also demonstrate that 14-3-3 binding to the S323 site on cdc25B blocks access of the substrate cyclin/cdks to the catalytic site of the enzyme, thereby directly inhibiting the activity of cdc25B. This provides direct mechanistic evidence that 14-3-3 binding to cdc25B can regulate its activity, thereby controlling progression into mitosis.
- Published
- 2001
- Full Text
- View/download PDF
40. Variations in Prkdc encoding the catalytic subunit of DNA-dependent protein kinase (DNA-PKcs) and susceptibility to radiation-induced apoptosis and lymphomagenesis.
- Author
-
Mori N, Matsumoto Y, Okumoto M, Suzuki N, and Yamate J
- Subjects
- Alleles, Animals, Catalytic Domain, Cell Transformation, Neoplastic genetics, Chromosome Mapping, DNA-Activated Protein Kinase, Female, Genetic Predisposition to Disease, Mice, Mice, Inbred BALB C, Mice, Inbred C57BL, Nuclear Proteins, Radiation Tolerance genetics, Recombination, Genetic, Apoptosis genetics, DNA-Binding Proteins, Genetic Variation, Lymphoma genetics, Protein Serine-Threonine Kinases genetics
- Abstract
DNA double-strand breaks (DSBs) induced by ionizing radiation enforce cells to die, if unrepaired; while if misrepaired, DSBs may cause malignant transformation. The DSB repair system predominant in mammals requires DNA-dependent protein kinase (DNA-PK). Previously, we identified the apoptosis susceptibility gene Radiation-induced apoptosis 1 (Rapop1) on mouse chromosome 16. The STS/A (STS) allele at Rapop1 leads to decreased sensitivity to apoptosis in the BALB/cHeA (BALB/c) background. In the present study, we established Rapop1 congenic strains C.S-R1 and C.S-R1L, which contain the STS genome in a 0.45 cM interval critical for Rapop1 in common in the BALB/c background. Within the segment critical for Rapop1, Prkdc encoding the catalytic subunit of DNA-PK (DNA-PKcs) was assigned. Two variations T6,418C and G11,530A, which induce amino acid substitutions C2,140R downstream from the putative leucine zipper motif and V3,844M near the kinase domain, respectively, were found between BALB/c and STS for Prkdc. The majority of inbred strains such as C57BL/6J carried the STS allele at Prkdc; a few strains including 129/SvJ and C.B17 carried the BALB/c allele. DNA-PK activity as well as DNA-PKcs expression was profoundly diminished in BALB/c and 129/SvJ mice as compared with C57BL/6 and C.S-R1 mice. In the crosses (C.S-R1 x BALB/c)F(1) x 129/SvJ and (C.S-R1 x BALB/c)F(1) x C.B17, enhanced apoptosis occurred in the absence of the wild-type allele at Prkdc. C.S-R1 and C.S-R1L were both less sensitive to radiation lymphomagenesis than BALB/c. Our study provides strong evidence for Prkdc as a candidate for Rapop1 and a susceptibility gene for radiation lymphomagenesis as well.
- Published
- 2001
- Full Text
- View/download PDF
41. A role for protein kinase CK2 in cell proliferation: evidence using a kinase-inactive mutant of CK2 catalytic subunit alpha.
- Author
-
Lebrin F, Chambaz EM, and Bianchini L
- Subjects
- 3T3 Cells cytology, 3T3 Cells enzymology, Animals, Casein Kinase II, Catalytic Domain, Cricetinae, Mice, Mutation, Phosphorylation, Precipitin Tests, Protein Serine-Threonine Kinases genetics, Protein Serine-Threonine Kinases metabolism, Transfection, Cell Division physiology, Protein Serine-Threonine Kinases physiology
- Abstract
Protein kinase CK2 is an ubiquitous and pleiotropic Ser/Thr protein kinase composed of two catalytic (alpha and/or alpha') and two regulatory (beta) subunits generally combined to form alpha(2)beta(2), alphaalpha'beta(2), or alpha'(2)beta(2) heterotetramers. To gain more insight into the role of CK2 in the control of proliferation in mammalian cells, overexpression of isolated CK2 subunits alpha, alpha', or beta was carried out in two fibroblast cell lines: NIH3T3 and CCL39. To interfere with CK2 cellular functions, cells were also transfected with a kinase-inactive mutant of CK2alpha catalytic subunit: CK2alpha-K68A. In NIH3T3 cells, overexpression of either wild-type subunit (alpha, alpha' or beta) had no effect on cell proliferation. In contrast, overexpression of the CK2alpha kinase-deficient mutant induced a marked inhibition of cell proliferation. This resulted from a defect in G1/S progression as demonstrated in transient transfection experiments in both NIH3T3 and CCL39 cells using BrdU incorporation measurements and in CCL39 clones stably overexpressing the CK2alpha-K68A mutant by growth curve analysis. We demonstrated that the kinase-negative mutant has the capacity to integrate the endogenous CK2 subunit pool both as an isolated kinase-inactive alpha subunit and as associated to the beta subunit in a kinase-inactive tetramer. Finally we showed that expression of the kinase-inactive mutant interferes with phosphorylation of an endogenous CK2 substrate; we speculate that optimal phosphorylation of target proteins by CK2 is required to achieve optimal cell cycle progression.
- Published
- 2001
- Full Text
- View/download PDF
42. The catalytic activity of dsRNA-dependent protein kinase, PKR, is required for NF-kappaB activation.
- Author
-
Gil J, Rullas J, García MA, Alcamí J, and Esteban M
- Subjects
- Animals, Catalytic Domain, Cells, Cultured, Enzyme Activation, Eukaryotic Initiation Factor-2 metabolism, Genes, Dominant, Humans, I-kappa B Kinase, Mice, Phosphorylation, Point Mutation, Protein Serine-Threonine Kinases metabolism, Recombinant Proteins genetics, Recombinant Proteins metabolism, Vaccinia virus genetics, eIF-2 Kinase genetics, NF-kappa B metabolism, eIF-2 Kinase metabolism
- Abstract
The double stranded RNA-dependent protein kinase (PKR), in addition to its role as a translational controlling factor, is a key transcriptional regulator exerting antiviral and antitumoral activities. We have previously shown that induction of NF-kappaB by PKR is involved in apoptosis commitment and this process is mediated through activation of the IKK complex. To gain insights into the mechanism of activation of NF-kappaB by PKR, we have analysed the domains of PKR involved in IKK activation and subsequent NF-kappaB induction. In PKR(0/0) cells infected with a collection of vaccinia virus (VV) recombinants expressing different mutant forms of PKR, we found that only PKR forms conserving the catalytic activity are able to activate NF-kappaB. An inactive PKR mutant (K296R), was unable to induce NF-kappaB activation despite full expression of the protein in a wide range of concentrations, as defined by Western blot, EMSA, IKK kinase activity and NF-kappaB transactivation assays. Moreover, the mutant PKR (K296R) acts as a dominant negative of PKR-induced eIF-2alpha phosphorylation and NF-kappaB activation. However, PKR mutants unable to activate NF-kappaB still retain their ability to associate with the IKK complex, as confirmed by immunoprecipitation analysis. We conclude that the catalytic activity of PKR and not only a protein-protein interaction with the IKK complex, is needed for activation of the transcription factor NF-kappaB.
- Published
- 2001
- Full Text
- View/download PDF
43. Development of inhibitors for protein tyrosine kinases.
- Author
-
Al-Obeidi FA and Lam KS
- Subjects
- Catalytic Domain, Combinatorial Chemistry Techniques, Enzyme Inhibitors chemical synthesis, Protein-Tyrosine Kinases metabolism, Enzyme Inhibitors pharmacology, Protein-Tyrosine Kinases antagonists & inhibitors
- Abstract
In the last 5 years, through combinatorial chemistry, high-throughput screening, computational chemistry, and traditional medicinal chemistry, numerous inhibitors for various protein tyrosine kinases (PTKs) have been developed. The majority of these compounds are small molecules that compete at the ATP binding site of the catalytic domain of the enzymes. Some compounds such as pseudosubstrate-based peptide inhibitor binds to the peptide/protein substrate site of the catalytic domain. Some inhibitors, primarily monoclonal antibodies, bind to the extracellular domain of receptor tyrosine kinases. Some of these inhibitors are highly potent and selective. Several are currently undergoing clinical trials for a number of diseases such as cancer.
- Published
- 2000
- Full Text
- View/download PDF
44. The effector loop and prenylation site of R-Ras are involved in the regulation of integrin function.
- Author
-
Oertli B, Han J, Marte BM, Sethi T, Downward J, Ginsberg M, and Hughes PE
- Subjects
- Amino Acid Sequence, Animals, Binding Sites, CHO Cells metabolism, Catalytic Domain, Cell Membrane metabolism, Conserved Sequence, Cricetinae, Enzyme Activation, GTP Phosphohydrolases genetics, GTP Phosphohydrolases metabolism, Molecular Sequence Data, Mutagenesis, Site-Directed, Phosphatidylinositol 3-Kinases metabolism, Protein Binding, Protein Prenylation, Proto-Oncogene Proteins c-raf metabolism, ral Guanine Nucleotide Exchange Factor metabolism, ras Proteins genetics, ras Proteins metabolism, GTP Phosphohydrolases physiology, Integrins physiology, Signal Transduction physiology, ras Proteins physiology
- Abstract
The closely related small GTP-binding proteins H-Ras and R-Ras have opposing effects on the regulation of integrin cell adhesion receptors. To gain insight into the properties of R-Ras with respect to the regulation of integrin function and interactions with downstream effectors we performed an analysis of R-Ras variants containing mutations in the effector binding domain and C-terminal prenylation site. We found that the activation of the downstream effector PI 3-kinase was sensitive to mutations in the effector binding domain, as was the binding to the effectors, Ral-GDS, Raf-1 and the novel effector Nore1. Furthermore, specific mutations in the effector binding loop and C-terminal prenylation motif impaired the ability of R-Ras to regulate integrin function in CHO cells. However, the ability of the R-Ras effector loop mutants to bind, and activate known effectors did not correlate with their ability to regulate integrin function. Thus, the known R-Ras effectors are not critical for regulating integrin activation, at least in CHO cells. Consequently, these studies provide insight into the structural basis of the interactions between R-Ras and its candidate effectors and suggest the existence of novel mechanisms through which this GTPase could regulate cell adhesion.
- Published
- 2000
- Full Text
- View/download PDF
45. The mitotic serine/threonine kinase Aurora2/AIK is regulated by phosphorylation and degradation.
- Author
-
Walter AO, Seghezzi W, Korver W, Sheung J, and Lees E
- Subjects
- Amino Acid Sequence, Aurora Kinases, Catalytic Domain, Cell Cycle, Cysteine Endopeptidases metabolism, Enzyme Activation drug effects, HeLa Cells drug effects, HeLa Cells enzymology, Humans, Mitosis, Molecular Sequence Data, Multienzyme Complexes metabolism, Mutagenesis, Site-Directed, Neoplasm Proteins metabolism, Okadaic Acid pharmacology, Phosphorylation drug effects, Phosphothreonine metabolism, Proteasome Endopeptidase Complex, Protein Phosphatase 1, Protein Serine-Threonine Kinases chemistry, Protein Serine-Threonine Kinases genetics, Recombinant Fusion Proteins metabolism, Sequence Alignment, Sequence Homology, Amino Acid, Ubiquitins metabolism, Cyclic AMP-Dependent Protein Kinases metabolism, Phosphoprotein Phosphatases metabolism, Protein Processing, Post-Translational drug effects, Protein Serine-Threonine Kinases metabolism
- Abstract
Aurora2 is a cell cycle regulated serine/threonine protein kinase which is overexpressed in many tumor cell lines. We demonstrate that Aurora2 is regulated by phosphorylation in a cell cycle dependent manner. This phosphorylation occurs on a conserved residue, Threonine 288, within the activation loop of the catalytic domain of the kinase and results in a significant increase in the enzymatic activity. Threonine 288 resides within a consensus motif for the cAMP dependent kinase and can be phosphorylated by PKA in vitro. The protein phosphatase 1 is shown to dephosphorylate this site in vitro, and in vivo the phosphorylation of T288 is induced by okadaic acid treatment. Furthermore, we show that the Aurora2 kinase is regulated by proteasome dependent degradation and that Aurora2 phosphorylated on T288 may be targeted for degradation during mitosis. Our experiments suggest that phosphorylation of T288 is important for regulation of the Aurora2 kinase both for its activity and its stability.
- Published
- 2000
- Full Text
- View/download PDF
46. The Src/Csk regulatory circuit arose early in metazoan evolution.
- Author
-
Miller MA, Malik IA, Shenk MA, and Steele RE
- Subjects
- Animals, CSK Tyrosine-Protein Kinase, Catalytic Domain, Cell Division genetics, Gene Expression Regulation, Phosphorylation, Phylogeny, Polymerase Chain Reaction, Protein-Tyrosine Kinases metabolism, Receptor Protein-Tyrosine Kinases metabolism, Receptors, Cell Surface metabolism, Yeasts genetics, Yeasts growth & development, src-Family Kinases metabolism, Biological Evolution, Hydra physiology, Protein-Tyrosine Kinases genetics, Receptor Protein-Tyrosine Kinases genetics, Receptors, Cell Surface genetics, src-Family Kinases genetics
- Abstract
We have identified a gene encoding a member of the Csk family of non-receptor protein-tyrosine kinases (PTKs) in the early-diverging metazoan Hydra. In situ hybridization analysis of the distribution of RNA from the Hydra Csk gene indicates that it is expressed in most of the epithelial cells of the adult polyp and in gametogenic cells. Comparison of the expression pattern of Hydra Csk with that of STK, the Hydra Src gene orthologue, reveals that the two genes are largely co-expressed. Such co-expression is consistent with a role for Hydra Csk in regulation of STK activity. This possibility was tested directly by coexpressing Hydra Csk with STK in yeast. Co-expression suppressed the growth inhibition seen when STK alone is expressed in yeast. Suppression was dependent on the presence of the putative regulatory tyrosine in the carboxyl-terminal tail of STK. Phosphotyrosine immunoblot analysis confirmed that expression of Csk resulted in suppression of STK kinase activity. Taken together these data indicate that the regulatory circuit involving Src and Csk PTKs was established prior to the divergence of the phylum Cnidaria from the rest of the metazoans.
- Published
- 2000
- Full Text
- View/download PDF
47. Adenovirus E4orf4 protein interacts with both Balpha and B' subunits of protein phosphatase 2A, but E4orf4-induced apoptosis is mediated only by the interaction with Balpha.
- Author
-
Shtrichman R, Sharf R, and Kleinberger T
- Subjects
- Adenovirus E4 Proteins genetics, Adenoviruses, Human genetics, Binding Sites, Catalytic Domain, Cell Line, Transformed, Humans, Mutagenesis, Phosphoprotein Phosphatases genetics, Protein Phosphatase 2, Protein Structure, Tertiary, Tumor Cells, Cultured, Viral Proteins genetics, Adenovirus E4 Proteins metabolism, Adenoviruses, Human metabolism, Apoptosis, Fungal Proteins, Phosphoprotein Phosphatases metabolism, Plant Proteins genetics, Viral Proteins metabolism
- Abstract
Adenovirus E4orf4 protein is a multifunctional viral regulator, which is involved in down regulation of virally-modulated signal transduction, in control of alternative splicing of viral mRNAs, and in induction of apoptosis in transformed cells. It has been previously shown that E4orf4 interacts with protein phosphatase 2A through the phosphatase Balpha subunit. It was further shown that PP2A is required for performing the various E4orf4 functions. We report here that E4orf4 interacts with multiple isoforms of the PP2A-B' subunit, as well as with Balpha. We map the interaction sites of the B subunits on E4orf4 and show that they overlap but are not identical. We identify a dominant negative E4orf4 mutant, which disrupts the PP2A holoenzyme. We show that induction of apoptosis by E4orf4, which we previously reported to require the interaction with Balpha, is not affected by the interaction with B'. Our results suggest that the interaction of E4orf4 with various PP2A subpopulations may mediate the different E4orf4 functions.
- Published
- 2000
- Full Text
- View/download PDF
48. The human vaccinia-related kinase 1 (VRK1) phosphorylates threonine-18 within the mdm-2 binding site of the p53 tumour suppressor protein.
- Author
-
Lopez-Borges S and Lazo PA
- Subjects
- 3T3 Cells, Amino Acid Sequence, Animals, Base Sequence, Binding Sites, Casein Kinases, Catalytic Domain, Cell Nucleus metabolism, HeLa Cells, Humans, Intracellular Signaling Peptides and Proteins, Mice, Molecular Sequence Data, Nuclear Proteins classification, Nuclear Proteins genetics, Phosphorylation, Protein Kinases classification, Protein Kinases metabolism, Protein Serine-Threonine Kinases classification, Protein Serine-Threonine Kinases genetics, Proto-Oncogene Proteins c-mdm2, Recombinant Fusion Proteins classification, Recombinant Fusion Proteins genetics, Recombinant Fusion Proteins metabolism, Serine metabolism, Substrate Specificity, Threonine genetics, Transcription Factors metabolism, Tumor Suppressor Protein p53 genetics, Viral Proteins classification, Viral Proteins genetics, Nuclear Proteins metabolism, Protein Serine-Threonine Kinases metabolism, Proteins, Proto-Oncogene Proteins metabolism, Threonine metabolism, Tumor Suppressor Protein p53 metabolism, Viral Proteins metabolism
- Abstract
The tumour suppressor p53 protein integrates multiple signals regulating cell cycle progression and apoptosis. This regulation is mediated by several kinases that phosphorylate specific residues in the different functional domains of the p53 molecule. The human VRK1 protein is a new kinase related to a poxvirus kinase, and more distantly to the casein kinase 1 family. We have characterized the biochemical properties of human VRK1 from HeLa cells. VRK1 has a strong autophosphorylating activity in several Ser and Thr residues. VRK-1 phosphorylates acidic proteins, such as phosvitin and casein, and basic proteins such as histone 2b and myelin basic protein. Because some transcription factors are regulated by phosphorylation, we tested as substrates the N-transactivation domains of p53 and c-Jun fused to GST. Human c-Jun is not phosphorylated by VRK1. VRK1 phosphorylates murine p53 in threonine 18. This threonine is within the p53 hydrophobic loop (residues 13-23) required for the interaction of p53 with the cleft of its inhibitor mdm-2. The VRK1 C-terminus domain (residues 268-396) that contains a nuclear localization signal targets the protein to the nucleus, as determined by using fusion proteins with the green fluorescent protein. We conclude that VRK1 is an upstream regulator of p53 that belongs to a new signalling pathway.
- Published
- 2000
- Full Text
- View/download PDF
49. Demonstration in vivo that stromelysin-3 functions through its proteolytic activity.
- Author
-
Noël A, Boulay A, Kebers F, Kannan R, Hajitou A, Calberg-Bacq CM, Basset P, Rio MC, and Foidart JM
- Subjects
- Animals, Base Sequence, Carcinogenicity Tests, Catalytic Domain, Female, Humans, Mammary Neoplasms, Experimental genetics, Mammary Neoplasms, Experimental pathology, Matrix Metalloproteinase 11, Mice, Mice, Nude, Molecular Sequence Data, Mutagenesis, Site-Directed, Retroviridae genetics, Tissue Inhibitor of Metalloproteinase-2 genetics, Tissue Inhibitor of Metalloproteinase-2 metabolism, Tumor Cells, Cultured, Adenocarcinoma genetics, Breast Neoplasms genetics, Metalloendopeptidases genetics, Metalloendopeptidases metabolism
- Abstract
Stromelysin-3 (ST3), a matrix metalloproteinase (MMP) expressed in aggressive carcinomas, has been shown to promote tumor development in different in vivo experimental models. However, the inability of its mature form to degrade extracellular matrix components casts doubt on whether ST3 functions in vivo as a protease. In this study, we evaluated whether the ST3 tumor-promoting effect could be ascribed to its proteolytic activity and whether this putative protease could be targeted with MMP inhibitors. Catalytically inactive mutant cDNA of human (h) ST3 or mouse (m) ST3 were generated and transfected into MCF7 cells. When injected into nude mice in the presence of matrigel, the mutant-bearing cells did not exhibit the enhanced tumorigenicity elicited by MCF7 cells transfected with wild-type ST3 cDNA. In a second approach, TIMP2 overproduction in MCF7 cells expressing hST3 was induced by retroviral infection. The co-expression of ST3 and TIMP2 failed to enhance the tumorigenicity of MCF7 cells. Notably, matrigel depleted of low-molecular-weight proteins and growth factors failed to promote the tumorigenicity of ST3-expressing MCF7 cells. These findings provide the first in vivo evidence that ST3 is indeed a protease that can modulate cancer progression by remodeling extracellular matrix and probably by inducing it to release the necessary microenvironmental factors. Thus, ST3 represents an interesting target for specific MMP inhibition.
- Published
- 2000
- Full Text
- View/download PDF
50. Activity of MDM2, a ubiquitin ligase, toward p53 or itself is dependent on the RING finger domain of the ligase.
- Author
-
Honda R and Yasuda H
- Subjects
- Amino Acid Sequence, Animals, Catalytic Domain genetics, Ligases physiology, Mice, Molecular Sequence Data, Protein Structure, Tertiary genetics, Proto-Oncogene Proteins genetics, Proto-Oncogene Proteins c-mdm2, Ubiquitin-Protein Ligases, Ligases metabolism, Nuclear Proteins, Proto-Oncogene Proteins metabolism, Ubiquitins metabolism, Zinc Fingers physiology
- Abstract
We previously showed that oncoprotein MDM2 has ubiquitin ligase activity toward tumor suppressor p53. In that paper, we showed very weak homology in the carboxyl terminal portion between MDM2 and E6AP (HECT domain). We mutated the cysteine residue (C464) corresponding to the residue essential for the ubiquitin ligase activity of E6AP and this mutation diminished the ligase activity of MDM2. The cysteine residue described above is also one of the cysteine residues that form the RING finger domain of MDM2. We tried to find out whether the diminishing of the activity by the mutation is attributable to the disruption of the RING finger domain or not. When the ring finger domain of MDM2 was deleted, the truncation mutant did not have the ubiquitin ligase activity. When we mutated the seven cysteine residues of RING finger domain of MDM2 in the carboxyl terminus, the disruption of each residue in the RING finger completely diminished the ubiquitin ligase activity of MDM2 toward MDM2 itself and toward tumor suppressor p53. These data indicate that the RING finger domain in MDM2 is essential for its ubiquitin ligase activity toward p53 and itself.
- Published
- 2000
- Full Text
- View/download PDF
Catalog
Discovery Service for Jio Institute Digital Library
For full access to our library's resources, please sign in.