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










Publication year range
1.
Cold Spring Harb Perspect Med ; 5(1): a020909, 2014 Oct 03.
Article in English | MEDLINE | ID: mdl-25280901

ABSTRACT

Patents directed to naturally occurring genetic material, such as DNA, RNA, chromosomes, and genes, in an isolated or purified form have been granted in Australia for many years. This review provides scientists with a summary of the gene patent debate from an Australian perspective and specifically reviews how the various levels of the legal system as they apply to patents-the Australian Patent Office, Australian courts, and Australian government-have dealt with the issue of whether genetic material is proper subject matter for a patent.


Subject(s)
Genetics/legislation & jurisprudence , Patents as Topic/legislation & jurisprudence , Australia , Biotechnology/legislation & jurisprudence , Genetic Techniques , Genetic Testing/legislation & jurisprudence , Jurisprudence
2.
Curr Top Microbiol Immunol ; 347: 79-104, 2010.
Article in English | MEDLINE | ID: mdl-20582532

ABSTRACT

The catalytic and regulatory subunits of class I phosphoinositide 3-kinase (PI3K) have oncogenic potential. The catalytic subunit p110α and the regulatory subunit p85 undergo cancer-specific gain-of-function mutations that lead to enhanced enzymatic activity, ability to signal constitutively, and oncogenicity. The ß, γ, and δ isoforms of p110 are cell-transforming as overexpressed wild-type proteins. Class I PI3Ks have the unique ability to generate phosphoinositide 3,4,5 trisphosphate (PIP(3)). Class II and class III PI3Ks lack this ability. Genetic and cell biological evidence suggests that PIP(3) is essential for PI3K-mediated oncogenicity, explaining why class II and class III enzymes have not been linked to cancer. Mutational analysis reveals the existence of at least two distinct molecular mechanisms for the gain of function seen with cancer-specific mutations in p110α; one causing independence from upstream receptor tyrosine kinases, the other inducing independence from Ras. An essential component of the oncogenic signal that is initiated by PI3K is the TOR (target of rapamycin) kinase. TOR is an integrator of growth and of metabolic inputs. In complex with the raptor protein (TORC1), it controls cap-dependent translation, and this function is essential for PI3K-initiated oncogenesis.


Subject(s)
Neoplasms/etiology , Phosphatidylinositol 3-Kinases/physiology , Animals , Cell Transformation, Neoplastic , Humans , Isoenzymes/physiology , Mutation , Neoplasms/genetics , Phosphatidylinositol 3-Kinases/genetics
3.
Mol Cancer Res ; 7(7): 1132-8, 2009 Jul.
Article in English | MEDLINE | ID: mdl-19584261

ABSTRACT

Phosphatidylinositol 3-kinases (PI3K) are divided into three classes, which differ in their substrates and products. Class I generates the inositol phospholipids PI(3)P, PI(3,4)P2, and PI(3,4,5)P3 referred as PIP, PIP2, and PIP3, respectively. Class II produces PIP and PIP2, and class III generates only PIP. Substrate and product differences of the three classes are determined by the activation loops of their catalytic domains. Substitution of the class I activation loop with either class II or III activation loop results in a corresponding change of substrate preference and product restriction. We have evaluated such activation loop substitutions to show that oncogenic activity of class I PI3K is linked to the ability to produce PIP3. We further show that reduction of cellular PIP3 levels by the 5'-phosphatase PIPP interferes with PI3K-induced oncogenic transformation. PIPP also attenuates signaling through Akt and target of rapamycin. Class III PI3K fails to induce oncogenic transformation. Likewise, a constitutively membrane-bound class I PI3K mutant retaining only the protein kinase is unable to induce transformation. We conclude that PIP3 is an essential component of PI3K-mediated oncogenesis and that inability to generate PIP3 abolishes oncogenic potential.


Subject(s)
Cell Transformation, Neoplastic/metabolism , Phosphatidylinositol 3-Kinases/metabolism , Phosphatidylinositol Phosphates/metabolism , Animals , Catalytic Domain , Cell Line , Chick Embryo , Fibroblasts/metabolism , Inositol Polyphosphate 5-Phosphatases , Phosphoric Monoester Hydrolases/metabolism , Proto-Oncogene Proteins c-akt/metabolism , Signal Transduction
4.
Methods Enzymol ; 438: 291-305, 2008.
Article in English | MEDLINE | ID: mdl-18413256

ABSTRACT

Signaling by class I phosphatidylinositol 3-kinase (PI3K) controls cell growth, replication, motility, and metabolism. The PI3K pathway commonly shows gain of function in cancer. Two small GTPases, Rheb (Ras homolog enriched in brain) and Ras (rat sarcoma viral oncogene), play important roles in PI3K signaling. Rheb activates the TOR (target of rapamycin) kinase in a GTP-dependent manner; it links TOR to upstream signaling components, including the tuberous sclerosis complex (TSC) and Akt (homolog of the Akt8 murine lymphoma viral oncoprotein). Constitutively active, GTP-bound Rheb is oncogenic in cell culture, and activity that requires farnesylation. Ras activates PI3K by recruitment to the plasma membrane and possibly by inducing a conformational change in the catalytic subunit p110 of PI3K. In return, Ras signaling through the MAP kinase (MAPK) pathway is activated by PIP(3), the product of PI3K. Loss of Ras function can interfere with PI3K signaling. Various lines of evidence suggest complementary roles for PI3K and MAPK signaling in oncogenesis.


Subject(s)
Neoplasms/physiopathology , Phosphatidylinositol 3-Kinases/genetics , Animals , Cell Transformation, Neoplastic , Class I Phosphatidylinositol 3-Kinases , Mice , Monomeric GTP-Binding Proteins/physiology , Neoplasms/genetics , Neuropeptides/physiology , Phosphatidylinositol 3-Kinases/physiology , Phosphatidylinositol Phosphates/physiology , Ras Homolog Enriched in Brain Protein , Signal Transduction
5.
EMBO J ; 27(1): 265-76, 2008 Jan 09.
Article in English | MEDLINE | ID: mdl-18046459

ABSTRACT

Embryonic development and normal growth require exquisite control of insulin-like growth factors (IGFs). In mammals the extracellular region of the cation-independent mannose-6-phosphate receptor has gained an IGF-II-binding function and is termed type II IGF receptor (IGF2R). IGF2R sequesters IGF-II; imbalances occur in cancers and IGF2R is implicated in tumour suppression. We report crystal structures of IGF2R domains 11-12, 11-12-13-14 and domains 11-12-13/IGF-II complex. A distinctive juxtaposition of these domains provides the IGF-II-binding unit, with domain 11 directly interacting with IGF-II and domain 13 modulating binding site flexibility. Our complex shows that Phe19 and Leu53 of IGF-II lock into a hydrophobic pocket unique to domain 11 of mammalian IGF2Rs. Mutagenesis analyses confirm this IGF-II 'binding-hotspot', revealing that IGF-binding proteins and IGF2R have converged on the same high-affinity site.


Subject(s)
Insulin-Like Growth Factor II/chemistry , Insulin-Like Growth Factor II/physiology , Receptor, IGF Type 2/chemistry , Receptor, IGF Type 2/physiology , Animals , CHO Cells , Cell Line , Cricetinae , Cricetulus , Crystallography, X-Ray , Humans , Insulin-Like Growth Factor II/genetics , Mutagenesis , Protein Binding/physiology , Protein Structure, Tertiary , Receptor, IGF Type 2/genetics , Structure-Activity Relationship
6.
Mol Cell Biol ; 27(10): 3569-77, 2007 May.
Article in English | MEDLINE | ID: mdl-17325037

ABSTRACT

The insulin-like growth factors (insulin-like growth factor I [IGF-I] and IGF-II) exert important effects on growth, development, and differentiation through the IGF-I receptor (IGF-IR) transmembrane tyrosine kinase. The insulin receptor (IR) is structurally related to the IGF-IR, and at high concentrations, the IGFs can also activate the IR, in spite of their generally low affinity for the latter. Two mechanisms that facilitate cross talk between the IGF ligands and the IR at physiological concentrations have been described. The first of these is the existence of an alternatively spliced IR variant that exhibits high affinity for IGF-II as well as for insulin. A second phenomenon is the ability of hybrid receptors comprised of IGF-IR and IR hemireceptors to bind IGFs, but not insulin. To date, however, direct activation of an IR holoreceptor by IGF-I at physiological levels has not been demonstrated. We have now found that IGF-I can function through both splice variants of the IR, in spite of low affinity, to specifically activate IRS-2 to levels similar to those seen with equivalent concentrations of insulin or IGF-II. The specific activation of IRS-2 by IGF-I through the IR does not result in activation of the extracellular signal-regulated kinase pathway but does induce delayed low-level activation of the phosphatidylinositol 3-kinase pathway and biological effects such as enhanced cell viability and protection from apoptosis. These findings suggest that IGF-I can function directly through the IR and that the observed effects of IGF-I on insulin sensitivity may be the result of direct facilitation of insulin action by IGF-I costimulation of the IR in insulin target tissues.


Subject(s)
Insulin-Like Growth Factor II/metabolism , Insulin-Like Growth Factor I/metabolism , Intracellular Signaling Peptides and Proteins/metabolism , Phosphoproteins/metabolism , Receptor, IGF Type 1/metabolism , Receptor, Insulin/metabolism , Alternative Splicing , Animals , Cell Line , Cell Survival , Humans , Insulin/metabolism , Insulin Receptor Substrate Proteins , Insulin-Like Growth Factor Binding Proteins/genetics , Insulin-Like Growth Factor Binding Proteins/metabolism , Intracellular Signaling Peptides and Proteins/genetics , Mice , Phosphoproteins/genetics , RNA, Small Interfering/genetics , RNA, Small Interfering/metabolism , Receptor, IGF Type 1/genetics , Receptor, Insulin/genetics , Recombinant Fusion Proteins/genetics , Recombinant Fusion Proteins/metabolism , Signal Transduction/physiology
7.
Proteins ; 64(3): 758-68, 2006 Aug 15.
Article in English | MEDLINE | ID: mdl-16741994

ABSTRACT

Insulin-like growth factors (IGFs) are key regulators of cell proliferation, differentiation, and transformation, and are thus pivotal in cancer, especially breast, prostate, and colon neoplasm. Their potent mitogenic and anti-apoptotic actions depend primarily on their availability to bind to the signaling IGF cell surface receptors. One mechanism by which IGF-II availability is thought to be modulated is by binding to the nonsignaling IGF-II receptor (IGF2R). This binding is essentially mediated by domain 11 in the multidomain IGF2R extracellular region. The crystal structure of domain 11 of the human IGF-II receptor (IGF2R-d11) has identified a putative IGF-II binding site, and a nuclear magnetic resonance (NMR) solution structure for the IGF-II ligand has also been characterized. These structures have now been used to model in silico the protein-protein interaction between IGF-II and IGF2R-d11 using the program 3D-Dock. Because the IGF-II data comprise an ensemble of 20 structures, all of which satisfy the NMR constraints, the docking procedure was applied to each member of the ensemble. Only those models in which residue Ile1572 of IGF2R-d11, known to be essential for the binding of IGF-II, was at the interface were considered further. These plausible complexes were then critically assessed using an array of analysis techniques including consideration of additional mutagenesis data. One model was strongly supported by these analyses and is discussed here in detail. Furthermore, we demonstrate in vitro experimental support for this model by studying the binding of chimeras of IGF-I and IGF-II to IGF2R fragments.


Subject(s)
Insulin-Like Growth Factor II/chemistry , Mutation/genetics , Receptor, IGF Type 2/chemistry , Surface Plasmon Resonance/methods , Binding Sites/genetics , Humans , Insulin-Like Growth Factor I/chemistry , Insulin-Like Growth Factor I/genetics , Insulin-Like Growth Factor I/metabolism , Insulin-Like Growth Factor II/genetics , Insulin-Like Growth Factor II/metabolism , Magnetic Resonance Spectroscopy , Models, Molecular , Protein Binding , Protein Structure, Tertiary , Receptor, IGF Type 2/genetics , Receptor, IGF Type 2/metabolism , Structure-Activity Relationship
8.
Proc Natl Acad Sci U S A ; 103(5): 1289-94, 2006 Jan 31.
Article in English | MEDLINE | ID: mdl-16432180

ABSTRACT

Class I phosphoinositide 3-kinase contains four isoforms of the catalytic subunit, p110alpha, -beta, -gamma, and -delta. At physiological levels of expression, the wild-type p110alpha isoform lacks oncogenic potential, but gain-of-function mutations and overexpression of p110alpha are correlated with oncogenicity. The p110beta, -gamma, and -delta isoforms induce transformation of cultured cells as wild-type proteins. This oncogenic potential requires kinase activity and can be suppressed by the target of rapamycin inhibitor rapamycin. The p110delta isoform constitutively activates the Akt signaling pathway; p110gamma activates Akt only in the presence of serum. The isoforms differ in their requirements for upstream signaling. The transforming activity of the p110gamma isoform depends on rat sarcoma viral oncogene homolog (Ras) binding; preliminary data suggest the same for p110beta and indicate Ras-independent oncogenic potential of p110delta. The surprising oncogenic potential of the wild-type non-alpha isoforms of class I phosphoinositide 3-kinase may explain the dearth of cancer-specific mutations in these proteins, because these non-alpha isoforms could contribute to the oncogenic phenotype of the cell by differential expression.


Subject(s)
Cell Transformation, Neoplastic , Phosphatidylinositol 3-Kinases/chemistry , Animals , Antibiotics, Antineoplastic/pharmacology , Blotting, Western , Cell Differentiation , Cell Line, Tumor , Cells, Cultured , Chick Embryo , Class I Phosphatidylinositol 3-Kinases , Culture Media, Serum-Free/pharmacology , Humans , Mutagenesis, Site-Directed , Mutation , Phosphatidylinositol 3-Kinases/metabolism , Plasmids/metabolism , Point Mutation , Protein Binding , Protein Isoforms , Protein Kinases/metabolism , Signal Transduction , Sirolimus/pharmacology , TOR Serine-Threonine Kinases , Time Factors , Transfection , ras Proteins/metabolism
9.
Endocrinology ; 147(2): 1029-36, 2006 Feb.
Article in English | MEDLINE | ID: mdl-16239300

ABSTRACT

The actions of IGF-I and IGF-II are thought to be largely due to their activation of the IGF-I receptor. However, IGF-II can also bind with high affinity to, and effectively activate, an isoform of the insulin receptor (IR-A) that lacks a sequence at the carboxyl-terminal end of the extracellular alpha subunit due to the alternative splicing of exon 11. This isoform is poorly activated by IGF-I. Here, we show that IGF-II, but not IGF-I, induces potent autophosphorylation of residues Y1158, Y1162, and Y1163 in the activation loop of the kinase domain and tyrosine 960 in the juxtamembrane region of both IR-A and IR-B (exon 11+) isoforms. We have also found, by using IGF chimeras, that the C domain of IGF-II completely accounts for the ability of IGF-II to stimulate IR autophosphorylation compared with IGF-I. We further show that the C domains are responsible for the differential abilities of IGF-II and IGF-I to activate phosphorylation of insulin receptor substrate-1 and Akt, as well as their ability to induce migration and cell survival via the IR-A. Finally, we show for the first time that IGF signaling through the IR-A can protect cells from butyrate-induced apoptosis. In summary, our studies define the structural determinants that allow potent IGF-II signaling and regulation of cellular functions through the IR-A and provide novel insights into IGF signaling via the IR.


Subject(s)
Adipocytes/metabolism , Insulin-Like Growth Factor II/metabolism , Insulin-Like Growth Factor I/metabolism , Receptor, Insulin/metabolism , 3T3 Cells , Adipocytes/cytology , Animals , Gene Expression Regulation , Insulin-Like Growth Factor I/chemistry , Insulin-Like Growth Factor II/chemistry , Mice , Phosphorylation , Protein Isoforms , Protein Structure, Tertiary/physiology , Receptor, Insulin/chemistry
10.
Cytokine Growth Factor Rev ; 16(4-5): 421-39, 2005.
Article in English | MEDLINE | ID: mdl-15936977

ABSTRACT

The insulin-like growth factor (IGF) system is a complex network of two soluble ligands; several cell surface transmembrane receptors and six soluble high-affinity binding-proteins. The IGF system is essential for normal embryonic and postnatal growth, and plays an important role in the function of a healthy immune system, lymphopoiesis, myogenesis and bone growth among other physiological functions. Deregulation of the IGF system leads to stimulation of cancer cell growth and survival. In order to manipulate the IGF system in the treatment of certain disorders, we must understand the protein-protein interactions at a molecular level. The complex molecular interactions of the ligands and receptors of the IGF system underlie all the biological actions mentioned above and will be the focus of this review.


Subject(s)
Insulin-Like Growth Factor Binding Proteins/metabolism , Insulin-Like Growth Factor II/metabolism , Insulin-Like Growth Factor I/metabolism , Receptors, Somatomedin/metabolism , Amino Acid Sequence , Animals , Humans , Models, Biological , Models, Molecular , Molecular Sequence Data , Protein Binding
11.
Mol Endocrinol ; 19(3): 711-21, 2005 Mar.
Article in English | MEDLINE | ID: mdl-15576456

ABSTRACT

We have previously described the phenotype resulting from a missense mutation in the IGF-I gene, which leads to expression of IGF-I with a methionine instead of a valine at position 44 (Val44Met IGF-I). This mutation caused severe growth and mental retardation as well as deafness evident at birth and growth retardation in childhood, but is relatively well tolerated in adulthood. We have conducted a biochemical and structural analysis of Val44Met IGF-I to provide a molecular basis for the phenotype observed. Val44Met IGF-I exhibits a 90-fold decrease in type 1 IGF receptor (IGF-1R) binding compared with wild-type human IGF-I and only poorly stimulates autophosphorylation of the IGF-1R. The ability of Val44Met IGF-I to signal via the extracellular signal-regulated kinase 1/2 and Akt/protein kinase B pathways and to stimulate DNA synthesis is correspondingly poorer. Binding or activation of both insulin receptor isoforms is not detectable even at micromolar concentrations. However, Val44Met IGF-I binds IGF-binding protein-2 (IGFBP-2), IGFBP-3, and IGFBP-6 with equal affinity to IGF-I, suggesting the maintenance of overall structure, particularly in the IGFBP binding domain. Structural analysis by nuclear magnetic resonance confirms retention of near-native structure with only local side-chain disruptions despite the significant loss of function. To our knowledge, our results provide the first structural study of a naturally occurring mutant human IGF-I associated with growth and developmental abnormalities and identifies Val44 as an essential residue involved in the IGF-IGF-1R interaction.


Subject(s)
Insulin-Like Growth Factor I/genetics , Methionine/chemistry , Mutation, Missense , Valine/chemistry , Binding Sites , Binding, Competitive , Biological Assay , Blotting, Western , Cell Proliferation , DNA/metabolism , DNA Mutational Analysis , Dose-Response Relationship, Drug , Fibroblasts/metabolism , Humans , Immunoprecipitation , Inhibitory Concentration 50 , Insulin/metabolism , Insulin-Like Growth Factor I/metabolism , Ligands , Magnetic Resonance Spectroscopy , Mitogen-Activated Protein Kinase 1/metabolism , Mitogen-Activated Protein Kinase 3/metabolism , Models, Molecular , Mutation , Phenotype , Phosphorylation , Plasmids/metabolism , Protein Binding , Protein Isoforms , Protein Serine-Threonine Kinases/metabolism , Proto-Oncogene Proteins/metabolism , Proto-Oncogene Proteins c-akt , Receptor, IGF Type 1/metabolism , Recombinant Proteins/chemistry , Signal Transduction , Surface Plasmon Resonance , Thymidine/chemistry , Time Factors
12.
Mol Endocrinol ; 18(10): 2502-12, 2004 Oct.
Article in English | MEDLINE | ID: mdl-15205474

ABSTRACT

The insulin receptor (IR) lacking the alternatively spliced exon 11 (IR-A) is preferentially expressed in fetal and cancer cells. The IR-A has been identified as a high-affinity receptor for insulin and IGF-II but not IGF-I, which it binds with substantially lower affinity. Several cancer cell types that express the IR-A also overexpress IGF-II, suggesting a possible autocrine proliferative loop. To determine the regions of IGF-I and IGF-II responsible for this differential affinity, chimeras were made where the C and D domains were exchanged between IGF-I and IGF-II either singly or together. The abilities of these chimeras to bind to, and activate, the IR-A were investigated. We also investigated the ability of these chimeras to bind and activate the IR exon 11+ isoform (IR-B) and as a positive control, the IGF-I receptor (IGF-1R). We show that the C domain and, to a lesser extent, the D domains represent the principal determinants of the binding differences between IGF-I and IGF-II to IR-A. The C and D domains of IGF-II promote higher affinity binding to the IR-A than the equivalent domains of IGF-I, resulting in an affinity close to that of insulin for the IR-A. The C and D domains also regulate the IR-B binding specificity of the IGFs in a similar manner, although the level of binding for all IGF ligands to IR-B is lower than to IR-A. In contrast, the C and D domains of IGF-I allow higher affinity binding to the IGF-1R than the analogous domains of IGF-II. Activation of IGF-1R by the chimeras reflected their binding affinities whereas the phosphorylation of the two IR isoforms was more complex.


Subject(s)
Exons/genetics , Insulin-Like Growth Factor II/metabolism , Receptor, Insulin/genetics , 3T3 Cells , Alternative Splicing , Amino Acid Sequence , Animals , Humans , Insulin/chemistry , Insulin/genetics , Insulin-Like Growth Factor I/genetics , Insulin-Like Growth Factor I/metabolism , Insulin-Like Growth Factor II/chemistry , Insulin-Like Growth Factor II/genetics , Mice , Mice, Inbred BALB C , Molecular Sequence Data , Protein Isoforms/chemistry , Protein Isoforms/genetics , Recombinant Fusion Proteins/chemistry , Recombinant Fusion Proteins/metabolism , Restriction Mapping , Sequence Deletion
SELECTION OF CITATIONS
SEARCH DETAIL
...