Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 16 de 16
Filter
1.
J Biol Chem ; 294(12): 4621-4633, 2019 03 22.
Article in English | MEDLINE | ID: mdl-30659094

ABSTRACT

Phosphoinositide 3-kinase ß (PI3Kß) is regulated by receptor tyrosine kinases (RTKs), G protein-coupled receptors (GPCRs), and small GTPases such as Rac1 and Rab5. Our lab previously identified two residues (Gln596 and Ile597) in the helical domain of the catalytic subunit (p110ß) of PI3Kß whose mutation disrupts binding to Rab5. To better define the Rab5-p110ß interface, we performed alanine-scanning mutagenesis and analyzed Rab5 binding with an in vitro pulldown assay with GST-Rab5GTP Of the 35 p110ß helical domain mutants assayed, 11 disrupted binding to Rab5 without affecting Rac1 binding, basal lipid kinase activity, or Gßγ-stimulated kinase activity. These mutants defined the Rab5-binding interface within p110ß as consisting of two perpendicular α-helices in the helical domain that are adjacent to the initially identified Gln596 and Ile597 residues. Analysis of the Rab5-PI3Kß interaction by hydrogen-deuterium exchange MS identified p110ß peptides that overlap with these helices; no interactions were detected between Rab5 and other regions of p110ß or p85α. Similarly, the binding of Rab5 to isolated p85α could not be detected, and mutations in the Ras-binding domain (RBD) of p110ß had no effect on Rab5 binding. Whereas soluble Rab5 did not affect PI3Kß activity in vitro, the interaction of these two proteins was critical for chemotaxis, invasion, and gelatin degradation by breast cancer cells. Our results define a single, discrete Rab5-binding site in the p110ß helical domain, which may be useful for generating inhibitors to better define the physiological role of Rab5-PI3Kß coupling in vivo.


Subject(s)
Breast Neoplasms/pathology , Neoplasm Invasiveness , Phosphatidylinositol 3-Kinase/metabolism , rab5 GTP-Binding Proteins/metabolism , Binding Sites , Breast Neoplasms/metabolism , Cell Line, Tumor , Chemotaxis , Gelatin/metabolism , HEK293 Cells , Humans , Mass Spectrometry/methods , Mutation , Phosphatidylinositol 3-Kinase/genetics , Protein Binding
2.
PLoS Pathog ; 14(8): e1007248, 2018 08.
Article in English | MEDLINE | ID: mdl-30102745

ABSTRACT

Salmonella enterica serotype Typhimurium (S. Typhimurium) is one of the most frequent causes of food-borne illness in humans and usually associated with acute self-limiting gastroenteritis. However, in immunocompromised patients, the pathogen can disseminate and lead to severe systemic diseases. S. Typhimurium are facultative intracellular bacteria. For uptake and intracellular life, Salmonella translocate numerous effector proteins into host cells using two type-III secretion systems (T3SS), which are encoded within Salmonella pathogenicity islands 1 (SPI-1) and 2 (SPI-2). While SPI-1 effectors mainly promote initial invasion, SPI-2 effectors control intracellular survival and proliferation. Here, we elucidate the mode of action of Salmonella SPI-2 effector SseI, which is involved in control of systemic dissemination of S. Typhimurium. SseI deamidates a specific glutamine residue of heterotrimeric G proteins of the Gαi family, resulting in persistent activation of the G protein. Gi activation inhibits cAMP production and stimulates PI3-kinase γ by Gαi-released Gßγ subunits, resulting in activation of survival pathways by phosphorylation of Akt and mTOR. Moreover, SseI-induced deamidation leads to non-polarized activation of Gαi and, thereby, to loss of directed migration of dendritic cells.


Subject(s)
Bacterial Proteins/physiology , Chemotaxis , GTP-Binding Protein alpha Subunits, Gi-Go/metabolism , Salmonella typhimurium , Type III Secretion Systems/physiology , Animals , Bacterial Proteins/genetics , Cell Survival/genetics , Chemotaxis/genetics , Deamination/genetics , Female , GTP-Binding Protein alpha Subunits, Gi-Go/chemistry , HEK293 Cells , HeLa Cells , Host-Pathogen Interactions/genetics , Humans , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Protein Multimerization/genetics , Protein Processing, Post-Translational/genetics , RAW 264.7 Cells , Salmonella Infections/metabolism , Salmonella Infections/pathology , Salmonella typhimurium/genetics , Salmonella typhimurium/metabolism , Type III Secretion Systems/genetics , Type III Secretion Systems/metabolism
3.
Biochem J ; 474(23): 3903-3914, 2017 11 16.
Article in English | MEDLINE | ID: mdl-29046393

ABSTRACT

Phosphoinositide 3-kinases (PI 3-kinases) are regulated by a diverse range of upstream activators, including receptor tyrosine kinases (RTKs), G-protein-coupled receptors (GPCRs), and small GTPases from the Ras, Rho and Rab families. For the Class IA PI 3-kinase PI3Kß, two mechanisms for GPCR-mediated regulation have been described: direct binding of Gßγ subunits to the C2-helical domain linker of p110ß, and Dock180/Elmo1-mediated activation of Rac1, which binds to the Ras-Binding Domain of p110ß. We now show that the integration of these dual pathways is unexpectedly complex. In breast cancer cells, expression of constitutively activated Rac1 (CA-Rac1) along with either GPCR stimulation or expression of Gßγ led to an additive PI3Kß-dependent activation of Akt. Whereas CA-Rac1-mediated activation of Akt was blocked in cells expressing a mutated PI3Kß that cannot bind Gßγ, Gßγ and GPCR-mediated activation of Akt was preserved when Rac1 binding to PI3Kß was blocked. Surprisingly, PI3Kß-dependent CA-Rac1 signaling to Akt was still seen in cells expressing a mutant p110ß that cannot bind Rac1. Instead of directly binding to PI3Kß, CA-Rac1 acts by enhancing Gßγ coupling to PI3Kß, as CA-Rac1-mediated Akt activation was blocked by inhibitors of Gßγ. Cells expressing CA-Rac1 exhibited a robust induction of macropinocytosis, and inhibitors of macropinocytosis blocked the activation of Akt by CA-Rac1 or lysophosphatidic acid. Our data suggest that Rac1 can potentiate the activation of PI3Kß by GPCRs through an indirect mechanism, by driving the formation of macropinosomes that serve as signaling platforms for Gßγ coupling to PI3Kß.


Subject(s)
Class Ia Phosphatidylinositol 3-Kinase/metabolism , GTP-Binding Protein beta Subunits/metabolism , GTP-Binding Protein gamma Subunits/metabolism , Pinocytosis/physiology , Signal Transduction/physiology , rac1 GTP-Binding Protein/metabolism , Adaptor Proteins, Signal Transducing/genetics , Adaptor Proteins, Signal Transducing/metabolism , Cell Line, Tumor , Class Ia Phosphatidylinositol 3-Kinase/genetics , Enzyme Activation/genetics , GTP-Binding Protein beta Subunits/genetics , GTP-Binding Protein gamma Subunits/genetics , HEK293 Cells , Humans , Lysophospholipids/genetics , Lysophospholipids/metabolism , Protein Binding , Proto-Oncogene Proteins c-akt/genetics , Proto-Oncogene Proteins c-akt/metabolism , rac GTP-Binding Proteins/genetics , rac GTP-Binding Proteins/metabolism , rac1 GTP-Binding Protein/genetics
4.
Angew Chem Int Ed Engl ; 56(21): 5750-5754, 2017 05 15.
Article in English | MEDLINE | ID: mdl-28429411

ABSTRACT

G-protein-coupled-receptors (GPCRs) are of fundamental importance for signal transduction through cell membranes. This makes them important drug targets, but structure-based drug design (SBDD) is still hampered by the limitations for structure determination of unmodified GPCRs. We show that the interligand NOEs for pharmacophore mapping (INPHARMA) method can provide valuable information on ligand poses inside the binding site of the unmodified human A2A adenosine receptor reconstituted in nanodiscs. By comparing experimental INPHARMA spectra with back-calculated spectra based on ligand poses obtained from molecular dynamics simulations, a complex structure for A2A R with the low-affinity ligand 3-pyrrolidin-1-ylquinoxalin-2-amine was determined based on the X-ray structure of ligand ZM-241,358 in complex with a modified A2A R.


Subject(s)
Receptor, Adenosine A2A/chemistry , Receptors, G-Protein-Coupled/chemistry , Binding Sites , Humans , Ligands , Lipids , Magnetic Resonance Spectroscopy , Molecular Structure , Protein Binding , Protein Domains
5.
J Biol Chem ; 290(48): 28915-31, 2015 Nov 27.
Article in English | MEDLINE | ID: mdl-26438819

ABSTRACT

Phosphatidylinositol 3,4,5-trisphosphate (PIP3)-dependent Rac exchanger 2 (PREX2) is a guanine nucleotide exchange factor (GEF) for the Ras-related C3 botulinum toxin substrate 1 (Rac1) GTPase, facilitating the exchange of GDP for GTP on Rac1. GTP-bound Rac1 then activates its downstream effectors, including p21-activated kinases (PAKs). PREX2 and Rac1 are frequently mutated in cancer and have key roles within the insulin-signaling pathway. Rac1 can be inactivated by multiple mechanisms; however, negative regulation by insulin is not well understood. Here, we show that in response to being activated after insulin stimulation, Rac1 initiates its own inactivation by decreasing PREX2 GEF activity. Following PREX2-mediated activation of Rac1 by the second messengers PIP3 or Gßγ, we found that PREX2 was phosphorylated through a PAK-dependent mechanism. PAK-mediated phosphorylation of PREX2 reduced GEF activity toward Rac1 by inhibiting PREX2 binding to PIP3 and Gßγ. Cell fractionation experiments also revealed that phosphorylation prevented PREX2 from localizing to the cellular membrane. Furthermore, the onset of insulin-induced phosphorylation of PREX2 was delayed compared with AKT. Altogether, we propose that second messengers activate the Rac1 signal, which sets in motion a cascade whereby PAKs phosphorylate and negatively regulate PREX2 to decrease Rac1 activation. This type of regulation would allow for transient activation of the PREX2-Rac1 signal and may be relevant in multiple physiological processes, including diseases such as diabetes and cancer when insulin signaling is chronically activated.


Subject(s)
Guanine Nucleotide Exchange Factors/metabolism , Second Messenger Systems/physiology , p21-Activated Kinases/metabolism , rac1 GTP-Binding Protein/metabolism , Guanine Nucleotide Exchange Factors/genetics , HEK293 Cells , Humans , Phosphorylation/physiology , p21-Activated Kinases/genetics , rac1 GTP-Binding Protein/genetics
6.
Biochem J ; 469(1): 59-69, 2015 Jul 01.
Article in English | MEDLINE | ID: mdl-26173259

ABSTRACT

Class IB phosphoinositide 3-kinases γ (PI3Kγ) are second-messenger-generating enzymes downstream of signalling cascades triggered by G-protein-coupled receptors (GPCRs). PI3Kγ variants have one catalytic p110γ subunit that can form two different heterodimers by binding to one of a pair of non-catalytic subunits, p87 or p101. Growing experimental data argue for a different regulation of p87-p110γ and p101-p110γ allowing integration into distinct signalling pathways. Pharmacological tools enabling distinct modulation of the two variants are missing. The ability of an anti-p110γ monoclonal antibody [mAb(A)p110γ] to block PI3Kγ enzymatic activity attracted us to characterize this tool in detail using purified proteins. In order to get insight into the antibody-p110γ interface, hydrogen-deuterium exchange coupled to MS (HDX-MS) measurements were performed demonstrating binding of the monoclonal antibody to the C2 domain in p110γ, which was accompanied by conformational changes in the helical domain harbouring the Gßγ-binding site. We then studied the modulation of phospholipid vesicles association of PI3Kγ by the antibody. p87-p110γ showed a significantly reduced Gßγ-mediated phospholipid recruitment as compared with p101-p110γ. Concomitantly, in the presence of mAb(A)p110γ, Gßγ did not bind to p87-p110γ. These data correlated with the ability of the antibody to block Gßγ-stimulated lipid kinase activity of p87-p110γ 30-fold more potently than p101-p110γ. Our data argue for differential regulatory functions of the non-catalytic subunits and a specific Gßγ-dependent regulation of p101 in PI3Kγ activation. In this scenario, we consider the antibody as a valuable tool to dissect the distinct roles of the two PI3Kγ variants downstream of GPCRs.


Subject(s)
Antibodies, Monoclonal, Murine-Derived/chemistry , Class Ib Phosphatidylinositol 3-Kinase , GTP-Binding Protein beta Subunits , GTP-Binding Protein gamma Subunits , Animals , Class Ib Phosphatidylinositol 3-Kinase/chemistry , Class Ib Phosphatidylinositol 3-Kinase/genetics , Class Ib Phosphatidylinositol 3-Kinase/metabolism , Deuterium Exchange Measurement , GTP-Binding Protein beta Subunits/chemistry , GTP-Binding Protein beta Subunits/genetics , GTP-Binding Protein beta Subunits/metabolism , GTP-Binding Protein gamma Subunits/chemistry , GTP-Binding Protein gamma Subunits/genetics , GTP-Binding Protein gamma Subunits/metabolism , HEK293 Cells , Humans , Sf9 Cells , Spodoptera
7.
J Med Chem ; 58(1): 212-21, 2015 Jan 08.
Article in English | MEDLINE | ID: mdl-24983663

ABSTRACT

The importance of complete elucidation of the biological functions of phosphoinositide 3-kinases (PI3K) was realized years ago. They generate 3-phosphoinositides, which are known to function as important second messengers in many inter- and intracellular signaling pathways. However, the functional role of class II PI3Ks is still unclear. Herein, we describe the synthesis of a panel of compounds that were tested against all eight mammalian PI3K-isoforms. We found inhibitors with some selectivity for class II PI3K-C2γ and also compounds with preferred inhibition of class II PI3K-C2ß, providing structural leads to develop selective tool compounds.


Subject(s)
Phosphoinositide-3 Kinase Inhibitors , Protein Kinase Inhibitors/chemistry , Protein Kinase Inhibitors/pharmacology , Animals , Class II Phosphatidylinositol 3-Kinases/metabolism , Dose-Response Relationship, Drug , Drug Discovery , Humans , Models, Chemical , Molecular Structure , Protein Kinase Inhibitors/chemical synthesis , Pyrazines/chemistry , Pyrazines/pharmacology , Sulfonamides/chemistry , Sulfonamides/pharmacology , Benzenesulfonamides
8.
PLoS One ; 9(7): e102425, 2014.
Article in English | MEDLINE | ID: mdl-25014207

ABSTRACT

Small Rho GTPases are well known to regulate a variety of cellular processes by acting as molecular switches. The regulatory function of Rho GTPases is critically dependent on their posttranslational modification at the carboxyl terminus by isoprenylation and association with proper cellular membranes. Despite numerous studies, the mechanisms of recycling and functional integration of Rho GTPases at the biological membranes are largely unclear. In this study, prenylated human Rac1, a prominent member of the Rho family, was purified in large amount from baculovirus-infected Spodoptera frugiperda insect cells using a systematic detergent screening. In contrast to non-prenylated human Rac1 purified from Escherichia coli, prenylated Rac1 from insect cells was able to associate with synthetic liposomes and to bind Rho-specific guanine nucleotide dissociation inhibitor 1 (GDI1). Subsequent liposome reconstitution experiments revealed that GDI1 efficiently extracts Rac1 from liposomes preferentially in the inactive GDP-bound state. The extraction was prevented when Rac1 was activated to its GTP-bound state by Rac-specific guanine nucleotide exchange factors (GEFs), such as Vav2, Dbl, Tiam1, P-Rex1 and TrioN, and bound by the downstream effector Pak1. We found that dissociation of Rac1-GDP from its complex with GDI1 strongly correlated with two distinct activities of especially Dbl and Tiam1, including liposome association and the GDP/GTP exchange. Taken together, our results provided first detailed insights into the advantages of the in vitro liposome-based reconstitution system to study both the integration of the signal transducing protein complexes and the mechanisms of regulation and signaling of small GTPases at biological membranes.


Subject(s)
Guanine Nucleotide Dissociation Inhibitors/chemistry , Guanine Nucleotide Exchange Factors/chemistry , Liposomes/chemistry , Protein Processing, Post-Translational , p21-Activated Kinases/chemistry , rac1 GTP-Binding Protein/chemistry , Animals , Baculoviridae/genetics , Escherichia coli/genetics , Escherichia coli/metabolism , Gene Expression , Guanine Nucleotide Dissociation Inhibitors/genetics , Guanine Nucleotide Dissociation Inhibitors/metabolism , Guanine Nucleotide Exchange Factors/genetics , Guanine Nucleotide Exchange Factors/metabolism , Humans , Liposomes/metabolism , Models, Biological , Protein Prenylation , Recombinant Proteins/chemistry , Recombinant Proteins/genetics , Recombinant Proteins/metabolism , Spodoptera , p21-Activated Kinases/genetics , p21-Activated Kinases/metabolism , rac1 GTP-Binding Protein/genetics , rac1 GTP-Binding Protein/metabolism
9.
Proc Natl Acad Sci U S A ; 110(47): 18862-7, 2013 Nov 19.
Article in English | MEDLINE | ID: mdl-24190998

ABSTRACT

Phosphoinositide 3-kinase gamma (PI3Kγ) has profound roles downstream of G-protein-coupled receptors in inflammation, cardiac function, and tumor progression. To gain insight into how the enzyme's activity is shaped by association with its p101 adaptor subunit, lipid membranes, and Gßγ heterodimers, we mapped these regulatory interactions using hydrogen-deuterium exchange mass spectrometry. We identify residues in both the p110γ and p101 subunits that contribute critical interactions with Gßγ heterodimers, leading to PI3Kγ activation. Mutating Gßγ-interaction sites of either p110γ or p101 ablates G-protein-coupled receptor-mediated signaling to p110γ/p101 in cells and severely affects chemotaxis and cell transformation induced by PI3Kγ overexpression. Hydrogen-deuterium exchange mass spectrometry shows that association with the p101 regulatory subunit causes substantial protection of the RBD-C2 linker as well as the helical domain of p110γ. Lipid interaction massively exposes that same helical site, which is then stabilized by Gßγ. Membrane-elicited conformational change of the helical domain could help prepare the enzyme for Gßγ binding. Our studies and others identify the helical domain of the class I PI3Ks as a hub for diverse regulatory interactions that include the p101, p87 (also known as p84), and p85 adaptor subunits; Rab5 and Gßγ heterodimers; and the ß-adrenergic receptor kinase.


Subject(s)
Class Ib Phosphatidylinositol 3-Kinase/chemistry , Class Ib Phosphatidylinositol 3-Kinase/metabolism , Models, Molecular , Phosphatidylinositol 3-Kinases/metabolism , Protein Conformation , Receptors, G-Protein-Coupled/metabolism , Signal Transduction/physiology , Animals , Chemotaxis , Class Ib Phosphatidylinositol 3-Kinase/genetics , Deuterium Exchange Measurement , Enzyme Activation , HEK293 Cells , Humans , Mass Spectrometry , Mice , Microscopy, Confocal , NIH 3T3 Cells , Receptors, G-Protein-Coupled/agonists , Signal Transduction/genetics , ras Proteins/metabolism
10.
J Biol Chem ; 288(43): 31059-68, 2013 Oct 25.
Article in English | MEDLINE | ID: mdl-24014027

ABSTRACT

Class IB phosphoinositide 3-kinase γ (PI3Kγ) comprises a single catalytic p110γ subunit, which binds to two non-catalytic subunits, p87 or p101, and controls a plethora of fundamental cellular responses. The non-catalytic subunits are assumed to be redundant adaptors for Gßγ enabling G-protein-coupled receptor-mediated regulation of PI3Kγ. Growing experimental data provide contradictory evidence. To elucidate the roles of the non-catalytic subunits in determining the specificity of PI3Kγ, we tested the impact of p87 and p101 in heterodimeric p87-p110γ and p101-p110γ complexes on the modulation of PI3Kγ activity in vitro and in living cells. RT-PCR, biochemical, and imaging data provide four lines of evidence: (i) specific expression patterns of p87 and p101, (ii) up-regulation of p101, providing the basis to consider p87 as a protein forming a constitutively and p101 as a protein forming an inducibly expressed PI3Kγ, (iii) differences in basal and stimulated enzymatic activities, and (iv) differences in complex stability, all indicating apparent diversity within class IB PI3Kγ. In conclusion, expression and activities of PI3Kγ are modified differently by p87 and p101 in vitro and in living cells, arguing for specific regulatory roles of the non-catalytic subunits in the differentiation of PI3Kγ signaling pathways.


Subject(s)
Class Ib Phosphatidylinositol 3-Kinase/metabolism , Gene Expression Regulation, Enzymologic/physiology , Protein Multimerization/physiology , Signal Transduction/physiology , Animals , Class Ib Phosphatidylinositol 3-Kinase/genetics , Female , HEK293 Cells , Humans , Male , Sf9 Cells , Spodoptera , Substrate Specificity/physiology
11.
PLoS One ; 8(5): e63833, 2013.
Article in English | MEDLINE | ID: mdl-23734178

ABSTRACT

The PI3-kinase pathway is commonly activated in tumors, most often by loss of PTEN lipid phosphatase activity or the amplification or mutation of p110α. Oncogenic mutants have commonly been found in p110α, but rarely in any of the other catalytic subunits of class I PI3-kinases. We here characterize a p110ß helical domain mutation, E633K, first identified in a Her2-positive breast cancer. The mutation increases basal p110ß activity, but does not affect activation of p85/p110ß dimers by phosphopeptides or Gßγ. Expression of the mutant causes increases in Akt and S6K1 activation, transformation, chemotaxis, proliferation and survival in low serum. E633 is conserved among class I PI3 Ks, and its mutation in p110ß is also activating. Interestingly, the E633K mutant occurs near a region that interacts with membranes in activated PI 3-kinases, and its mutation abrogates the requirement for an intact Ras-binding domain in p110ß-mediated transformation. We propose that the E633K mutant activates p110ß by enhancing its basal association with membranes. This study presents the first analysis of an activating oncogenic mutation of p110ß.


Subject(s)
Amino Acid Substitution , Breast Neoplasms/genetics , Class Ia Phosphatidylinositol 3-Kinase/genetics , Mutation , Amino Acid Sequence , Animals , Blotting, Western , Breast Neoplasms/enzymology , Cell Membrane/metabolism , Cell Movement/genetics , Cell Proliferation , Cell Survival/genetics , Cell Transformation, Neoplastic/genetics , Class Ia Phosphatidylinositol 3-Kinase/metabolism , Enzyme Activation/genetics , Female , HEK293 Cells , Humans , Liposomes/metabolism , Mice , NIH 3T3 Cells , Phosphotransferases/metabolism , Protein Binding , Proto-Oncogene Proteins c-akt/metabolism , Ribosomal Protein S6 Kinases, 70-kDa/metabolism , Sequence Homology, Amino Acid , Sf9 Cells
12.
Sci Signal ; 5(253): ra89, 2012 Dec 04.
Article in English | MEDLINE | ID: mdl-23211529

ABSTRACT

Synergistic activation by heterotrimeric guanine nucleotide-binding protein (G protein)-coupled receptors (GPCRs) and receptor tyrosine kinases distinguishes p110ß from other class IA phosphoinositide 3-kinases (PI3Ks). Activation of p110ß is specifically implicated in various physiological and pathophysiological processes, such as the growth of tumors deficient in phosphatase and tensin homolog deleted from chromosome 10 (PTEN). To determine the specific contribution of GPCR signaling to p110ß-dependent functions, we identified the site in p110ß that binds to the Gßγ subunit of G proteins. Mutation of this site eliminated Gßγ-dependent activation of PI3Kß (a dimer of p110ß and the p85 regulatory subunit) in vitro and in cells, without affecting basal activity or phosphotyrosine peptide-mediated activation. Disrupting the p110ß-Gßγ interaction by mutation or with a cell-permeable peptide inhibitor blocked the transforming capacity of PI3Kß in fibroblasts and reduced the proliferation, chemotaxis, and invasiveness of PTEN-null tumor cells in culture. Our data suggest that specifically targeting GPCR signaling to PI3Kß could provide a therapeutic approach for tumors that depend on p110ß for growth and metastasis.


Subject(s)
Cell Transformation, Neoplastic/metabolism , Fibroblasts/metabolism , GTP-Binding Protein beta Subunits/metabolism , GTP-Binding Protein gamma Subunits/metabolism , Neoplasm Proteins/metabolism , Neoplasms/metabolism , Phosphatidylinositol 3-Kinases/metabolism , Receptors, G-Protein-Coupled/metabolism , Cell Line , Cell Transformation, Neoplastic/genetics , Cell Transformation, Neoplastic/pathology , Class I Phosphatidylinositol 3-Kinases , Fibroblasts/pathology , GTP-Binding Protein beta Subunits/genetics , GTP-Binding Protein gamma Subunits/genetics , Humans , Neoplasm Invasiveness , Neoplasm Metastasis , Neoplasm Proteins/genetics , Neoplasms/genetics , Neoplasms/pathology , Neoplasms/therapy , Phosphatidylinositol 3-Kinases/genetics , Receptors, G-Protein-Coupled/genetics , Signal Transduction/genetics
13.
J Cell Biochem ; 113(3): 934-45, 2012 Mar.
Article in English | MEDLINE | ID: mdl-22371973

ABSTRACT

Zonula occludens protein 1 (ZO-1) is a ubiquitous scaffolding protein, but it is unknown why it functions in very different cellular contacts. We hypothesized that a specific segment, the unique hinge region, can be bound by very different regulatory proteins. Using surface plasmon resonance spectroscopy and binding assays to peptide libraries, we show, for the first time, that the hinge region directly interacts with disparate signal elements such as G-proteins alpha 12 and alpha i2, the regulator of G-protein signaling 5, multifunctional signaling protein ahnak1, and L-type Ca2+-channel beta-2-subunit. The novel binding proteins specifically bound to a coiled coil-helix predicted in the hinge region of ZO-. The interactions were modulated by phosphorylation in the hinge helix. Activation of the G-proteins influenced their association to ZO-1. In colon cells, G alpha i2 and ZO-1 were associated, as shown by coimmunoprecipitation. After cotransfection in kidney cells, G alpha i2 barely colocalized with ZO-1; the colocalization coefficient was significantly increased when epinephrine activated G-protein signaling. In conclusion, proteins with different regulatory potential adhere to and influence cellular functions of ZO-proteins, and the interactions can be modulated via its hinge region and/or the binding proteins.


Subject(s)
Intracellular Signaling Peptides and Proteins/metabolism , Membrane Proteins/metabolism , Phosphoproteins/metabolism , Caco-2 Cells , Cell Membrane/chemistry , Epithelial Cells/chemistry , Epithelial Cells/metabolism , GTP-Binding Protein alpha Subunit, Gi2/metabolism , HEK293 Cells , Humans , Membrane Proteins/analysis , Membrane Proteins/chemistry , Phosphoproteins/analysis , Phosphoproteins/chemistry , RGS Proteins/metabolism , Zonula Occludens-1 Protein
14.
Biochem J ; 441(3): 851-8, 2012 Feb 01.
Article in English | MEDLINE | ID: mdl-22054284

ABSTRACT

G-protein-regulated PI3Kγ (phosphoinositide 3-kinase γ) plays a crucial role in inflammatory and allergic processes. PI3Kγ, a dimeric protein formed by the non-catalytic p101 and catalytic p110γ subunits, is stimulated by receptor-released Gßγ complexes. We have demonstrated previously that Gßγ stimulates both monomeric p110γ and dimeric p110γ/p101 lipid kinase activity in vitro. In order to identify the Gß residues responsible for the Gßγ-PI3Kγ interaction, we examined Gß1 mutants for their ability to stimulate lipid and protein kinase activities and to recruit PI3Kγ to lipid vesicles. Our findings revealed different interaction profiles of Gß residues interacting with p110γ or p110γ/p101. Moreover, p101 was able to rescue the stimulatory activity of Gß1 mutants incapable of modulating monomeric p110γ. In addition to the known adaptor function of p101, in the present paper we show a novel regulatory role of p101 in the activation of PI3Kγ.


Subject(s)
Class Ib Phosphatidylinositol 3-Kinase/physiology , GTP-Binding Protein beta Subunits/metabolism , Animals , Cells, Cultured , Class Ib Phosphatidylinositol 3-Kinase/chemistry , Class Ib Phosphatidylinositol 3-Kinase/genetics , Class Ib Phosphatidylinositol 3-Kinase/metabolism , Drug Resistance/genetics , Drug Resistance/physiology , Enzyme Activation/genetics , GTP-Binding Protein beta Subunits/chemistry , GTP-Binding Protein beta Subunits/genetics , Humans , Isoenzymes/chemistry , Isoenzymes/genetics , Isoenzymes/metabolism , Isoenzymes/physiology , Lipid Metabolism/genetics , Lipid Metabolism/physiology , Models, Molecular , Mutant Proteins/genetics , Mutant Proteins/metabolism , Proteolysis/drug effects , Spodoptera , Transfection , Trypsin/metabolism , Trypsin/pharmacology
15.
Proc Natl Acad Sci U S A ; 106(48): 20312-7, 2009 Dec 01.
Article in English | MEDLINE | ID: mdl-19906996

ABSTRACT

Class I(B) phosphoinositide 3-kinase gamma (PI3Kgamma) elicits various immunologic and cardiovascular responses; however, the molecular basis for this signal heterogeneity is unclear. PI3Kgamma consists of a catalytic p110gamma and a regulatory p87(PIKAP) (p87, also p84) or p101 subunit. Hitherto p87 and p101 are generally assumed to exhibit redundant functions in receptor-induced and G protein betagamma (Gbetagamma)-mediated PI3Kgamma regulation. Here we investigated the molecular mechanism for receptor-dependent p87/p110gamma activation. By analyzing GFP-tagged proteins expressed in HEK293 cells, PI3Kgamma-complemented bone marrow-derived mast cells (BMMCs) from p110gamma(-/-) mice, and purified recombinant proteins reconstituted to lipid vesicles, we elucidated a novel pathway of p87-dependent, G protein-coupled receptor (GPCR)-induced PI3Kgamma activation. Although p101 strongly interacted with Gbetagamma, thereby mediating PI3Kgamma membrane recruitment and stimulation, p87 exhibited only a weak interaction, resulting in modest kinase activation and lack of membrane recruitment. Surprisingly, Ras-GTP substituted the missing Gbetagamma-dependent membrane recruitment of p87/p110gamma by direct interaction with p110gamma, suggesting the indispensability of Ras for activation of p87/p110gamma. Consequently, interference with Ras signaling indeed selectively blocked p87/p110gamma, but not p101/p110gamma, kinase activity in HEK293 and BMMC cells, revealing an important crosstalk between monomeric and trimeric G proteins for p87/p110gamma activation. Our data display distinct signaling requirements of p87 and p101, conferring signaling specificity to PI3Kgamma that could open up new possibilities for therapeutic intervention.


Subject(s)
Enzyme Activation/physiology , Models, Molecular , Phosphatidylinositol 3-Kinases/metabolism , Receptors, G-Protein-Coupled/metabolism , Signal Transduction/physiology , ras Proteins/metabolism , Animals , Cell Line , Fluorescent Antibody Technique , Green Fluorescent Proteins , Humans , Mast Cells/metabolism , Mice , Microscopy, Confocal
16.
Protein Pept Lett ; 16(9): 1053-6, 2009.
Article in English | MEDLINE | ID: mdl-19508220

ABSTRACT

G-protein betagamma dimers are prime regulators transmitting extracellular signals to wide-ranging cellular effectors including phosphoinositide 3-kinase (PI3K) isoforms beta and gamma. Recombinant Gbetagamma purified from Sf9 cells via metal-affinity and anion exchange chromatography exhibited a wortmannin-insensitive phospholipid kinase activity that copurified from the insect cells. To exclude false-positive results of Gbetagamma-dependent lipid kinase activity, the elimination of insect phospholipid kinase from Gbetagamma protein samples is necessary to avoid interference with the intrinsic lipid kinase activity of PI3K isoforms in reconstitution experiments. Here we describe an improved procedure of Gbeta(1)gamma(2) purification from cell membranes that separates the contaminating phospholipid kinase.


Subject(s)
GTP-Binding Protein beta Subunits/isolation & purification , GTP-Binding Protein gamma Subunits/isolation & purification , Phosphatidylinositol 3-Kinases/metabolism , Androstadienes/pharmacology , Animals , Baculoviridae/genetics , Chromatography, Gel , GTP-Binding Protein beta Subunits/metabolism , GTP-Binding Protein gamma Subunits/metabolism , Phosphatidylinositol 3-Kinases/isolation & purification , Phosphotransferases/isolation & purification , Recombinant Proteins/isolation & purification , Spodoptera/metabolism , Wortmannin
SELECTION OF CITATIONS
SEARCH DETAIL
...