Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 21
Filter
Add more filters










Publication year range
1.
EMBO Rep ; 25(6): 2610-2634, 2024 Jun.
Article in English | MEDLINE | ID: mdl-38698221

ABSTRACT

GABAB receptors (GBRs), the G protein-coupled receptors for GABA, regulate synaptic transmission throughout the brain. A main synaptic function of GBRs is the gating of Cav2.2-type Ca2+ channels. However, the cellular compartment where stable GBR/Cav2.2 signaling complexes form remains unknown. In this study, we demonstrate that the vesicular protein synaptotagmin-11 (Syt11) binds to both the auxiliary GBR subunit KCTD16 and Cav2.2 channels. Through these dual interactions, Syt11 recruits GBRs and Cav2.2 channels to post-Golgi vesicles, thus facilitating assembly of GBR/Cav2.2 signaling complexes. In addition, Syt11 stabilizes GBRs and Cav2.2 channels at the neuronal plasma membrane by inhibiting constitutive internalization. Neurons of Syt11 knockout mice exhibit deficits in presynaptic GBRs and Cav2.2 channels, reduced neurotransmitter release, and decreased GBR-mediated presynaptic inhibition, highlighting the critical role of Syt11 in the assembly and stable expression of GBR/Cav2.2 complexes. These findings support that Syt11 acts as a vesicular scaffold protein, aiding in the assembly of signaling complexes from low-abundance components within transport vesicles. This mechanism enables insertion of pre-assembled functional signaling units into the synaptic membrane.


Subject(s)
Mice, Knockout , Signal Transduction , Synaptotagmins , Animals , Synaptotagmins/metabolism , Synaptotagmins/genetics , Mice , Humans , Neurons/metabolism , Synaptic Transmission , Receptors, GABA-B/metabolism , Receptors, GABA-B/genetics , Presynaptic Terminals/metabolism , Calcium Channels, N-Type/metabolism , Calcium Channels, N-Type/genetics , Golgi Apparatus/metabolism , Protein Binding , HEK293 Cells
2.
Biochem Pharmacol ; : 116176, 2024 Mar 28.
Article in English | MEDLINE | ID: mdl-38555036

ABSTRACT

GABAB receptors (GBRs) are G protein-coupled receptors for GABA, the main inhibitory neurotransmitter in the brain. GBRs regulate fast synaptic transmission by gating Ca2+ and K+ channels via the Gßγ subunits of the activated G protein. It has been demonstrated that auxiliary GBR subunits, the KCTD proteins, shorten onset and rise time and increase desensitization of receptor-induced K+ currents. KCTD proteins increase desensitization of K+ currents by scavenging Gßγ from the channel, yet the mechanism responsible for the rapid activation of K+ currents has remained elusive. In this study, we demonstrate that KCTD proteins preassemble Gßγ at GBRs. The preassembly obviates the need for diffusion-limited G protein recruitment to the receptor, thereby accelerating G protein activation and, as a result, K+ channel activation. Preassembly of Gßγ at the receptor relies on the interaction of KCTD proteins with a loop protruding from the seven-bladed propeller of Gß subunits. The binding site is shared between Gß1 and Gß2, limiting the interaction of KCTD proteins to these particular Gß isoforms. Substituting residues in the KCTD binding site of Gß1 with those from Gß3 hinders the preassembly of Gßγ with GBRs, delays onset and prolongs rise time of receptor-activated K+ currents. The KCTD-Gß interface, therefore, represents a target for pharmacological modulation of channel gating by GBRs.

3.
Elife ; 122023 01 23.
Article in English | MEDLINE | ID: mdl-36688536

ABSTRACT

Amyloid-ß precursor protein (APP) regulates neuronal activity through the release of secreted APP (sAPP) acting at cell surface receptors. APP and sAPP were reported to bind to the extracellular sushi domain 1 (SD1) of GABAB receptors (GBRs). A 17 amino acid peptide (APP17) derived from APP was sufficient for SD1 binding and shown to mimic the inhibitory effect of sAPP on neurotransmitter release and neuronal activity. The functional effects of APP17 and sAPP were similar to those of the GBR agonist baclofen and blocked by a GBR antagonist. These experiments led to the proposal that sAPP activates GBRs to exert its neuronal effects. However, whether APP17 and sAPP influence classical GBR signaling pathways in heterologous cells was not analyzed. Here, we confirm that APP17 binds to GBRs with nanomolar affinity. However, biochemical and electrophysiological experiments indicate that APP17 does not influence GBR activity in heterologous cells. Moreover, APP17 did not regulate synaptic GBR localization, GBR-activated K+ currents, neurotransmitter release, or neuronal activity in vitro or in vivo. Our results show that APP17 is not a functional GBR ligand and indicate that sAPP exerts its neuronal effects through receptors other than GBRs.


Subject(s)
Amyloid beta-Peptides , Amyloid beta-Protein Precursor , Amyloid beta-Peptides/metabolism , Amyloid beta-Protein Precursor/metabolism , Neurons/metabolism , Neurotransmitter Agents/metabolism , gamma-Aminobutyric Acid/metabolism
4.
Curr Top Behav Neurosci ; 52: 19-37, 2022.
Article in English | MEDLINE | ID: mdl-32812202

ABSTRACT

GABAB receptors (GBRs), the G protein-coupled receptors for the inhibitory neurotransmitter γ-aminobutyric acid (GABA), activate Go/i-type G proteins that regulate adenylyl cyclase, Ca2+ channels, and K+ channels. GBR signaling to enzymes and ion channels influences neuronal activity, plasticity processes, and network activity throughout the brain. GBRs are obligatory heterodimers composed of GB1a or GB1b subunits with a GB2 subunit. Heterodimeric GB1a/2 and GB1b/2 receptors represent functional units that associate in a modular fashion with regulatory, trafficking, and effector proteins to generate receptors with distinct physiological functions. This review summarizes current knowledge on the structure, organization, and functions of multi-protein GBR complexes.


Subject(s)
Receptors, GABA-B , Receptors, GABA , Neurons , Signal Transduction , gamma-Aminobutyric Acid
5.
Elife ; 102021 04 29.
Article in English | MEDLINE | ID: mdl-33913808

ABSTRACT

The synaptic connection from medial habenula (MHb) to interpeduncular nucleus (IPN) is critical for emotion-related behaviors and uniquely expresses R-type Ca2+ channels (Cav2.3) and auxiliary GABAB receptor (GBR) subunits, the K+-channel tetramerization domain-containing proteins (KCTDs). Activation of GBRs facilitates or inhibits transmitter release from MHb terminals depending on the IPN subnucleus, but the role of KCTDs is unknown. We therefore examined the localization and function of Cav2.3, GBRs, and KCTDs in this pathway in mice. We show in heterologous cells that KCTD8 and KCTD12b directly bind to Cav2.3 and that KCTD8 potentiates Cav2.3 currents in the absence of GBRs. In the rostral IPN, KCTD8, KCTD12b, and Cav2.3 co-localize at the presynaptic active zone. Genetic deletion indicated a bidirectional modulation of Cav2.3-mediated release by these KCTDs with a compensatory increase of KCTD8 in the active zone in KCTD12b-deficient mice. The interaction of Cav2.3 with KCTDs therefore scales synaptic strength independent of GBR activation.


Subject(s)
Calcium Channels, R-Type/metabolism , Cation Transport Proteins/metabolism , Habenula/metabolism , Presynaptic Terminals/metabolism , Receptors, GABA/metabolism , Animals , Calcium Channels, R-Type/genetics , Cation Transport Proteins/genetics , Humans , Intracellular Signaling Peptides and Proteins , Male , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Receptors, GABA/genetics , Receptors, GABA-B/genetics , Receptors, GABA-B/metabolism , Synapses/genetics , Synapses/metabolism
6.
Neuropharmacology ; 190: 108426, 2021 06 01.
Article in English | MEDLINE | ID: mdl-33279506

ABSTRACT

For a long time metabotropic glutamate receptors (mGluRs) were thought to regulate neuronal functions as obligatory homodimers. Recent reports, however, indicate the existence of heterodimers between group-II and -III mGluRs in the brain, which differ from the homodimers in their signal transduction and sensitivity to negative allosteric modulators (NAMs). Whether the group-I mGluRs, mGlu1 and mGlu5, form functional heterodimers in the brain is still a matter of debate. We now show that mGlu1 and mGlu5 co-purify from brain membranes and hippocampal tissue and co-localize in cultured hippocampal neurons. Complementation assays with mutants deficient in agonist-binding or G protein-coupling reveal that mGlu1/5 heterodimers are functional in heterologous cells and transfected cultured hippocampal neurons. In contrast to heterodimers between group-II and -III mGluRs, mGlu1/5 receptors exhibit a symmetric signal transduction, with both protomers activating G proteins to a similar extent. NAMs of either protomer in mGlu1/5 receptors partially inhibit signaling, showing that both protomers need to be able to reach an active conformation for full receptor activity. Complete heterodimer inhibition is observed when both protomers are locked in their inactive state by a NAM. In summary, our data show that mGlu1/5 heterodimers exhibit a symmetric signal transduction and thus intermediate signaling efficacy and kinetic properties. Our data support the existence of mGlu1/5 heterodimers in neurons and highlight differences in the signaling transduction of heterodimeric mGluRs that influence allosteric modulation.


Subject(s)
Hippocampus/metabolism , Neurons/metabolism , Receptor, Metabotropic Glutamate 5/metabolism , Receptors, Metabotropic Glutamate/metabolism , Allosteric Regulation , Animals , Brain/metabolism , Chromatography, Liquid , Hippocampus/cytology , Mice , Mice, Knockout , Protein Multimerization , Receptor, Metabotropic Glutamate 5/genetics , Receptors, Metabotropic Glutamate/genetics , Signal Transduction , Tandem Mass Spectrometry
7.
Basic Clin Pharmacol Toxicol ; 126 Suppl 6: 25-34, 2020 Jun.
Article in English | MEDLINE | ID: mdl-31033219

ABSTRACT

GABAB receptors (GBRs), the G protein-coupled receptors for the neurotransmitter γ-aminobutyric acid (GABA), regulate synaptic transmission at most synapses in the brain. Proteomic approaches revealed that native GBR complexes assemble from an inventory of ~30 proteins that provide a molecular basis for the functional diversity observed with these receptors. Studies with reconstituted GBR complexes in heterologous cells and complementary knockout studies have allowed to identify cellular and physiological functions for obligate and several non-obligate receptor components. It emerges that modular association of receptor components in space and time generates a variety of multiprotein receptor complexes with different localizations, kinetic properties and effector channels. This article summarizes current knowledge on the organizing principle of GBR complexes. We further discuss unanticipated receptor functions, links to disease and opportunities for drug discovery arising from the identification of novel receptor components.


Subject(s)
Receptors, GABA-B/metabolism , Receptors, GABA-B/physiology , Animals , Brain/metabolism , Cell Membrane , GABA-B Receptor Agonists/pharmacology , GABA-B Receptor Antagonists/pharmacology , Proteomics , Receptors, G-Protein-Coupled , Receptors, GABA-B/chemistry
8.
J Med Chem ; 62(19): 8819-8830, 2019 10 10.
Article in English | MEDLINE | ID: mdl-31509708

ABSTRACT

Targeting multiprotein receptor complexes, rather than receptors directly, is a promising concept in drug discovery. This is particularly relevant to the GABAB receptor complex, which plays a prominent role in many brain functions and diseases. Here, we provide the first studies targeting a key protein-protein interaction of the GABAB receptor complex-the interaction with KCTD proteins. By employing the µSPOT technology, we first defined the GABAB receptor-binding epitope mediating the KCTD interaction. Subsequently, we developed a highly potent peptide-based inhibitor that interferes with the KCTD/GABAB receptor complex and efficiently isolates endogenous KCTD proteins from mouse brain lysates. X-ray crystallography and SEC-MALS revealed inhibitor induced oligomerization of KCTD16 into a distinct hexameric structure. Thus, we provide a template for modulating the GABAB receptor complex, revealing a fundamentally novel approach for targeting GABAB receptor-associated neuropsychiatric disorders.


Subject(s)
Intracellular Signaling Peptides and Proteins/metabolism , Nerve Tissue Proteins/metabolism , Peptides/metabolism , Receptors, GABA-B/metabolism , Amino Acid Sequence , Animals , Binding Sites , Brain/metabolism , Crystallography, X-Ray , Fluorescence Polarization , Humans , Intracellular Signaling Peptides and Proteins/chemistry , Intracellular Signaling Peptides and Proteins/genetics , Mice , Nerve Tissue Proteins/chemistry , Nerve Tissue Proteins/genetics , Peptides/chemistry , Protein Interaction Domains and Motifs , Protein Multimerization , Receptors, GABA-B/chemistry , Recombinant Proteins/biosynthesis , Recombinant Proteins/chemistry , Recombinant Proteins/isolation & purification
9.
Nat Commun ; 10(1): 1331, 2019 03 22.
Article in English | MEDLINE | ID: mdl-30902970

ABSTRACT

GABAB receptors (GBRs) are key regulators of synaptic release but little is known about trafficking mechanisms that control their presynaptic abundance. We now show that sequence-related epitopes in APP, AJAP-1 and PIANP bind with nanomolar affinities to the N-terminal sushi-domain of presynaptic GBRs. Of the three interacting proteins, selectively the genetic loss of APP impaired GBR-mediated presynaptic inhibition and axonal GBR expression. Proteomic and functional analyses revealed that APP associates with JIP and calsyntenin proteins that link the APP/GBR complex in cargo vesicles to the axonal trafficking motor. Complex formation with GBRs stabilizes APP at the cell surface and reduces proteolysis of APP to Aß, a component of senile plaques in Alzheimer's disease patients. Thus, APP/GBR complex formation links presynaptic GBR trafficking to Aß formation. Our findings support that dysfunctional axonal trafficking and reduced GBR expression in Alzheimer's disease increases Aß formation.


Subject(s)
Amyloid beta-Peptides/metabolism , Amyloid/metabolism , Axonal Transport , Receptors, GABA-B/metabolism , Amino Acid Sequence , Amyloid beta-Peptides/chemistry , Animals , Axons/metabolism , Cell Adhesion Molecules/chemistry , Cell Adhesion Molecules/metabolism , Cell Membrane/metabolism , Dendrites/metabolism , Epitopes/metabolism , GTP-Binding Proteins/metabolism , HEK293 Cells , Humans , Kinesins/metabolism , Mice, Inbred C57BL , Nerve Tissue Proteins/chemistry , Nerve Tissue Proteins/metabolism , Protein Binding , Protein Stability , Proteomics , Signal Transduction , Synapses/metabolism
10.
J Neurosci ; 37(5): 1162-1175, 2017 02 01.
Article in English | MEDLINE | ID: mdl-28003345

ABSTRACT

GABAB receptors are the G-protein coupled receptors for the main inhibitory neurotransmitter in the brain, GABA. GABAB receptors were shown to associate with homo-oligomers of auxiliary KCTD8, KCTD12, KCTD12b, and KCTD16 subunits (named after their T1 K+-channel tetramerization domain) that regulate G-protein signaling of the receptor. Here we provide evidence that GABAB receptors also associate with hetero-oligomers of KCTD subunits. Coimmunoprecipitation experiments indicate that two-thirds of the KCTD16 proteins in the hippocampus of adult mice associate with KCTD12. We show that the KCTD proteins hetero-oligomerize through self-interacting T1 and H1 homology domains. Bioluminescence resonance energy transfer measurements in live cells reveal that KCTD12/KCTD16 hetero-oligomers associate with both the receptor and the G-protein. Electrophysiological experiments demonstrate that KCTD12/KCTD16 hetero-oligomers impart unique kinetic properties on G-protein-activated Kir3 currents. During prolonged receptor activation (one min) KCTD12/KCTD16 hetero-oligomers produce moderately desensitizing fast deactivating K+ currents, whereas KCTD12 and KCTD16 homo-oligomers produce strongly desensitizing fast deactivating currents and nondesensitizing slowly deactivating currents, respectively. During short activation (2 s) KCTD12/KCTD16 hetero-oligomers produce nondesensitizing slowly deactivating currents. Electrophysiological recordings from hippocampal neurons of KCTD knock-out mice are consistent with these findings and indicate that KCTD12/KCTD16 hetero-oligomers increase the duration of slow IPSCs. In summary, our data demonstrate that simultaneous assembly of distinct KCTDs at the receptor increases the molecular and functional repertoire of native GABAB receptors and modulates physiologically induced K+ current responses in the hippocampus. SIGNIFICANCE STATEMENT: The KCTD proteins 8, 12, and 16 are auxiliary subunits of GABAB receptors that differentially regulate G-protein signaling of the receptor. The KCTD proteins are generally assumed to function as homo-oligomers. Here we show that the KCTD proteins also assemble hetero-oligomers in all possible dual combinations. Experiments in live cells demonstrate that KCTD hetero-oligomers form at least tetramers and that these tetramers directly interact with the receptor and the G-protein. KCTD12/KCTD16 hetero-oligomers impart unique kinetic properties to GABAB receptor-induced Kir3 currents in heterologous cells. KCTD12/KCTD16 hetero-oligomers are abundant in the hippocampus, where they prolong the duration of slow IPSCs in pyramidal cells. Our data therefore support that KCTD hetero-oligomers modulate physiologically induced K+ current responses in the brain.


Subject(s)
Potassium Channels/genetics , Potassium Channels/metabolism , Receptors, GABA-B/genetics , Receptors, GABA-B/metabolism , Animals , Brain Chemistry/genetics , CHO Cells , Cricetinae , Cricetulus , Electrophysiological Phenomena/genetics , Excitatory Postsynaptic Potentials/genetics , Female , Kinetics , Male , Mice , Mice, Knockout , Patch-Clamp Techniques , Receptors, G-Protein-Coupled/metabolism , Receptors, KIR/metabolism
11.
Nat Neurosci ; 19(2): 233-42, 2016 Feb.
Article in English | MEDLINE | ID: mdl-26691831

ABSTRACT

GABAB receptors, the most abundant inhibitory G protein-coupled receptors in the mammalian brain, display pronounced diversity in functional properties, cellular signaling and subcellular distribution. We used high-resolution functional proteomics to identify the building blocks of these receptors in the rodent brain. Our analyses revealed that native GABAB receptors are macromolecular complexes with defined architecture, but marked diversity in subunit composition: the receptor core is assembled from GABAB1a/b, GABAB2, four KCTD proteins and a distinct set of G-protein subunits, whereas the receptor's periphery is mostly formed by transmembrane proteins of different classes. In particular, the periphery-forming constituents include signaling effectors, such as Cav2 and HCN channels, and the proteins AJAP1 and amyloid-ß A4, both of which tightly associate with the sushi domains of GABAB1a. Our results unravel the molecular diversity of GABAB receptors and their postnatal assembly dynamics and provide a roadmap for studying the cellular signaling of this inhibitory neurotransmitter receptor.


Subject(s)
Proteomics/methods , Receptors, GABA-B/genetics , Amyloid beta-Protein Precursor/genetics , Animals , Caveolin 2/genetics , Cell Membrane/genetics , Cell Membrane/metabolism , Epitopes , Mice , Mice, Inbred BALB C , Mice, Knockout , Rats , Rats, Wistar , Receptors, G-Protein-Coupled , Receptors, GABA-B/metabolism , Signal Transduction/physiology
12.
Neuropharmacology ; 88: 145-54, 2015 Jan.
Article in English | MEDLINE | ID: mdl-25196734

ABSTRACT

GABAB receptors (GABABRs) are considered promising drug targets for the treatment of mental health disorders. GABABRs are obligate heteromers of principal GABAB1 and GABAB2 subunits. GABABRs can additionally associate with auxiliary KCTD8, 12, 12b and 16 subunits, which also bind the G-protein and differentially regulate G-protein signaling. It is unknown whether the KCTDs allosterically influence pharmacological properties of GABABRs. Here we show that KCTD8 and KCTD16 slightly but significantly increase GABA affinity at recombinant receptors. However, KCTDs clearly do not account for the 10-fold higher GABA affinity of native compared to recombinant GABABRs. The positive allosteric modulator (PAM) GS39783, which binds to GABAB2, increases both potency and efficacy of GABA-mediated G-protein activation ([(35)S]GTPγS binding, BRET between G-protein subunits), irrespective of whether KCTDs are present or not. Of note, the increase in efficacy was significantly larger in the presence of KCTD8, which likely is the consequence of a reduced tonic G-protein activation in the combined presence of KCTD8 and GABABRs. We recorded Kir3 currents to study the effects of GS39783 on receptor-activated G-protein ßγ-signaling. In transfected CHO cells and cultured hippocampal neurons GS39783 increased Kir3 current amplitudes activated by 1 µM of baclofen in the absence and presence of KCTDs. Our data show that auxiliary KCTD subunits exert marginal allosteric influences on principal GABABR subunits. PAMs at principal subunits will therefore not be selective for receptor subtypes owing to KCTD subunits. However, PAMs can differentially modulate the responses of receptor subtypes because the KCTDs differentially regulate G-protein signaling.


Subject(s)
Receptors, GABA-B/metabolism , Allosteric Regulation/drug effects , Allosteric Regulation/physiology , Animals , Baclofen/pharmacology , CHO Cells , Cells, Cultured , Cricetulus , Cyclopentanes/pharmacology , G Protein-Coupled Inwardly-Rectifying Potassium Channels/metabolism , GABA Modulators/pharmacology , GABA-B Receptor Agonists/pharmacology , GTP-Binding Proteins/metabolism , HEK293 Cells , Hippocampus/drug effects , Hippocampus/physiology , Humans , Mice , Neurons/drug effects , Neurons/physiology , Potassium/metabolism , Pyrimidines/pharmacology , Rats , Receptors, GABA-B/genetics , Recombinant Proteins/genetics , Recombinant Proteins/metabolism , gamma-Aminobutyric Acid/metabolism
13.
Neuron ; 82(5): 1032-44, 2014 Jun 04.
Article in English | MEDLINE | ID: mdl-24836506

ABSTRACT

Activation of K(+) channels by the G protein ßγ subunits is an important signaling mechanism of G-protein-coupled receptors. Typically, receptor-activated K(+) currents desensitize in the sustained presence of agonists to avoid excessive effects on cellular activity. The auxiliary GABAB receptor subunit KCTD12 induces fast and pronounced desensitization of the K(+) current response. Using proteomic and electrophysiological approaches, we now show that KCTD12-induced desensitization results from a dual interaction with the G protein: constitutive binding stabilizes the heterotrimeric G protein at the receptor, whereas dynamic binding to the receptor-activated Gßγ subunits induces desensitization by uncoupling Gßγ from the effector K(+) channel. While receptor-free KCTD12 desensitizes K(+) currents activated by other GPCRs in vitro, native KCTD12 is exclusively associated with GABAB receptors. Accordingly, genetic ablation of KCTD12 specifically alters GABAB responses in the brain. Our results show that GABAB receptors are endowed with fast and reversible desensitization by harnessing KCTD12 that intercepts Gßγ signaling.


Subject(s)
G Protein-Coupled Inwardly-Rectifying Potassium Channels/metabolism , GTP-Binding Protein beta Subunits/metabolism , GTP-Binding Protein gamma Subunits/metabolism , Receptors, GABA-B/metabolism , Receptors, GABA/metabolism , Animals , Brain/metabolism , CHO Cells , Cricetulus , HEK293 Cells , Humans , Intracellular Signaling Peptides and Proteins/metabolism , Mice , Receptors, GABA-B/chemistry
14.
J Biol Chem ; 288(34): 24848-56, 2013 Aug 23.
Article in English | MEDLINE | ID: mdl-23843457

ABSTRACT

GABA(B) receptors are the G-protein coupled receptors (GPCRs) for GABA, the main inhibitory neurotransmitter in the central nervous system. Native GABA(B) receptors comprise principle and auxiliary subunits that regulate receptor properties in distinct ways. The principle subunits GABA(B1a), GABA(B1b), and GABA(B2) form fully functional heteromeric GABA(B(1a,2)) and GABA(B(1b,2)) receptors. Principal subunits regulate forward trafficking of the receptors from the endoplasmic reticulum to the plasma membrane and control receptor distribution to axons and dendrites. The auxiliary subunits KCTD8, -12, -12b, and -16 are cytosolic proteins that influence agonist potency and G-protein signaling of GABA(B(1a,2)) and GABA(B(1b,2)) receptors. Here, we used transfected cells to study assembly, surface trafficking, and internalization of GABA(B) receptors in the presence of the KCTD12 subunit. Using bimolecular fluorescence complementation and metabolic labeling, we show that GABA(B) receptors associate with KCTD12 while they reside in the endoplasmic reticulum. Glycosylation experiments support that association with KCTD12 does not influence maturation of the receptor complex. Immunoprecipitation and bioluminescence resonance energy transfer experiments demonstrate that KCTD12 remains associated with the receptor during receptor activity and receptor internalization from the cell surface. We further show that KCTD12 reduces constitutive receptor internalization and thereby increases the magnitude of receptor signaling at the cell surface. Accordingly, knock-out or knockdown of KCTD12 in cultured hippocampal neurons reduces the magnitude of the GABA(B) receptor-mediated K(+) current response. In summary, our experiments support that the up-regulation of functional GABA(B) receptors at the neuronal plasma membrane is an additional physiological role of the auxiliary subunit KCTD12.


Subject(s)
Hippocampus/metabolism , Neurons/metabolism , Potassium Channels/metabolism , Potassium/metabolism , Protein Multimerization/physiology , Receptors, GABA-B/metabolism , Signal Transduction/physiology , Animals , COS Cells , Cell Membrane/genetics , Cell Membrane/metabolism , Chlorocebus aethiops , Hippocampus/cytology , Mice , Mice, Knockout , Neurons/cytology , Potassium Channels/genetics , Receptors, GABA-B/genetics
15.
Hepatology ; 57(4): 1607-19, 2013 Apr.
Article in English | MEDLINE | ID: mdl-23175466

ABSTRACT

UNLABELLED: Hepatocellular carcinoma (HCC) and cholangiocarcinoma (CCC) are the most common liver tumors and a leading cause for cancer-related death in men. Notch2 regulates cellular differentiation in the developing and adult liver. Although aberrant Notch signaling is implicated in various cancers, it is still unclear whether Notch2 regulates proliferation and differentiation in liver carcinogenesis and thereby contributes to HCC and CCC formation. Here, we investigated the oncogenic potential of constitutive Notch2 signaling in the liver. We show that liver-specific expression of the intracellular domain of Notch2 (N2ICD) in mice is sufficient to induce HCC formation and biliary hyperplasia. Specifically, constitutive N2ICD signaling in the liver leads to up-regulation of pro-proliferative genes and proliferation of hepatocytes and biliary epithelial cells (BECs). Using the diethylnitrosamine (DEN) HCC carcinogenesis model, we further show that constitutive Notch2 signaling accelerates DEN-induced HCC formation. DEN-induced HCCs with constitutive Notch2 signaling (DEN(N2ICD) HCCs) exhibit a marked increase in size, proliferation, and expression of pro-proliferative genes when compared with HCCs from DEN-induced control mice (DEN(ctrl) HCCs). Moreover, DEN(N2ICD) HCCs exhibit increased Sox9 messenger RNA (mRNA) levels and reduced Albumin and Alpha-fetoprotein mRNA levels, indicating that they are less differentiated than DEN(ctrl) HCCs. Additionally, DEN(N2ICD) mice develop large hepatic cysts, dysplasia of the biliary epithelium, and eventually CCC. CCC formation in patients and DEN(N2ICD) mice is accompanied by re-expression of hepatocyte nuclear factor 4α(HNF4α), possibly indicating dedifferentiation of BECs. CONCLUSION: Our data establish an oncogenic role for constitutive Notch2 signaling in liver cancer development.


Subject(s)
Carcinoma, Hepatocellular/chemically induced , Carcinoma, Hepatocellular/physiopathology , Diethylnitrosamine/adverse effects , Liver Neoplasms/chemically induced , Liver Neoplasms/physiopathology , Receptor, Notch2/physiology , Signal Transduction/physiology , Animals , Bile Duct Neoplasms/physiopathology , Bile Ducts, Intrahepatic , Carcinoma, Hepatocellular/metabolism , Cell Differentiation , Cell Proliferation , Cholangiocarcinoma/physiopathology , Disease Models, Animal , Female , Hepatocyte Nuclear Factor 4/metabolism , Humans , Liver Neoplasms/metabolism , Male , Mice , Mice, Transgenic , Receptor, Notch2/genetics
16.
J Biol Chem ; 287(47): 39869-77, 2012 Nov 16.
Article in English | MEDLINE | ID: mdl-23035119

ABSTRACT

GABA(B) receptors assemble from principle and auxiliary subunits. The principle subunits GABA(B1) and GABA(B2) form functional heteromeric GABA(B(1,2)) receptors that associate with homotetramers of auxiliary KCTD8, -12, -12b, or -16 (named after their K(+) channel tetramerization domain) subunits. These auxiliary subunits constitute receptor subtypes with distinct functional properties. KCTD12 and -12b generate desensitizing receptor responses while KCTD8 and -16 generate largely non-desensitizing receptor responses. The structural elements of the KCTDs underlying these differences in desensitization are unknown. KCTDs are modular proteins comprising a T1 tetramerization domain, which binds to GABA(B2), and a H1 homology domain. KCTD8 and -16 contain an additional C-terminal H2 homology domain that is not sequence-related to the H1 domains. No functions are known for the H1 and H2 domains. Here we addressed which domains and sequence motifs in KCTD proteins regulate desensitization of the receptor response. We found that the H1 domains in KCTD12 and -12b mediate desensitization through a particular sequence motif, T/NFLEQ, which is not present in the H1 domains of KCTD8 and -16. In addition, the H2 domains in KCTD8 and -16 inhibit desensitization when expressed C-terminal to the H1 domains but not when expressed as a separate protein in trans. Intriguingly, the inhibitory effect of the H2 domain is sequence-independent, suggesting that the H2 domain sterically hinders desensitization by the H1 domain. Evolutionary analysis supports that KCTD12 and -12b evolved desensitizing properties by liberating their H1 domains from antagonistic H2 domains and acquisition of the T/NFLEQ motif.


Subject(s)
Evolution, Molecular , Protein Subunits/metabolism , Proteins/metabolism , Receptors, GABA-B/metabolism , Amino Acid Motifs , Animals , CHO Cells , Cricetinae , Cricetulus , HEK293 Cells , Humans , Protein Binding , Protein Structure, Tertiary , Protein Subunits/genetics , Proteins/genetics , Receptors, GABA-B/genetics
17.
Nature ; 465(7295): 231-5, 2010 May 13.
Article in English | MEDLINE | ID: mdl-20400944

ABSTRACT

GABA(B) receptors are the G-protein-coupled receptors for gamma-aminobutyric acid (GABA), the main inhibitory neurotransmitter in the brain. They are expressed in almost all neurons of the brain, where they regulate synaptic transmission and signal propagation by controlling the activity of voltage-gated calcium (Ca(v)) and inward-rectifier potassium (K(ir)) channels. Molecular cloning revealed that functional GABA(B) receptors are formed by the heteromeric assembly of GABA(B1) with GABA(B2) subunits. However, cloned GABA(B(1,2)) receptors failed to reproduce the functional diversity observed with native GABA(B) receptors. Here we show by functional proteomics that GABA(B) receptors in the brain are high-molecular-mass complexes of GABA(B1), GABA(B2) and members of a subfamily of the KCTD (potassium channel tetramerization domain-containing) proteins. KCTD proteins 8, 12, 12b and 16 show distinct expression profiles in the brain and associate tightly with the carboxy terminus of GABA(B2) as tetramers. This co-assembly changes the properties of the GABA(B(1,2)) core receptor: the KCTD proteins increase agonist potency and markedly alter the G-protein signalling of the receptors by accelerating onset and promoting desensitization in a KCTD-subtype-specific manner. Taken together, our results establish the KCTD proteins as auxiliary subunits of GABA(B) receptors that determine the pharmacology and kinetics of the receptor response.


Subject(s)
Multiprotein Complexes/chemistry , Multiprotein Complexes/metabolism , Protein Multimerization , Protein Subunits/chemistry , Protein Subunits/metabolism , Receptors, GABA-B/chemistry , Receptors, GABA-B/metabolism , Animals , CHO Cells , Cricetinae , Cricetulus , Electric Conductivity , GABA-B Receptor Agonists , Heterotrimeric GTP-Binding Proteins/metabolism , Kinetics , Mice , Neurons/metabolism , Oocytes/metabolism , Potassium/metabolism , Potassium Channels/metabolism , Protein Structure, Tertiary , Rats , Rats, Wistar , Signal Transduction , Xenopus
18.
Exp Cell Res ; 315(17): 2888-98, 2009 Oct 15.
Article in English | MEDLINE | ID: mdl-19665017

ABSTRACT

c-Src is a non-receptor tyrosine kinase involved in regulating cell proliferation, cell migration and cell invasion and is tightly controlled by reversible phosphorylation on regulatory sites and through protein-protein interactions. The interaction of c-Src with PDZ proteins was recently identified as novel mechanism to restrict c-Src function. The objective of this study was to identify and characterise PDZ proteins that interact with c-Src to control its activity. By PDZ domain array screen, we identified the interaction of c-Src with the PDZ protein Membrane Protein Palmitoylated 2 (MPP2), a member of the Membrane-Associated Guanylate Kinase (MAGUK) family, to which also the Discs large (Dlg) tumour suppressor protein belongs. The function of MPP2 has not been established and the functional significance of the MPP2 c-Src interaction is not known. We found that in non-transformed breast epithelial MCF-10A cells, endogenous MPP2 associated with the cytoskeleton in filamentous structures, which partially co-localised with microtubules and c-Src. MPP2 and c-Src interacted in cells, where c-Src kinase activity promoted increased interaction of c-Src with MPP2. We furthermore found that MPP2 was able to negatively regulate c-Src kinase activity in cells, suggesting that the functional significance of the MPP2-c-Src interaction is to restrict Src activity. Consequently, the c-Src-dependent disorganisation of the cortical actin cytoskeleton of epithelial cells expressing c-Src was suppressed by MPP2. In conclusion we demonstrate here that MPP2 interacts with c-Src in cells to control c-Src activity and morphological function.


Subject(s)
Forkhead Transcription Factors/genetics , Forkhead Transcription Factors/metabolism , Proto-Oncogene Proteins pp60(c-src)/metabolism , Amino Acid Sequence , Animals , Binding Sites , COS Cells/metabolism , Cell Line , Chlorocebus aethiops , Cloning, Molecular , DNA, Complementary/genetics , Forkhead Box Protein M1 , Forkhead Transcription Factors/chemistry , Humans , Kidney/embryology , Microtubules/metabolism , Protein Binding , Proto-Oncogene Proteins pp60(c-src)/chemistry , Sequence Alignment , Transfection
19.
EMBO J ; 27(9): 1399-410, 2008 May 07.
Article in English | MEDLINE | ID: mdl-18388859

ABSTRACT

We have previously identified a protein, consisting of seven WD-repeats, forming a putative beta-propeller, and an FYVE domain, ProF, which is highly expressed in 3T3-L1 cells, a cell line that can be differentiated into adipocytes. We recently found ProF to interact with the kinases Akt and protein kinase Czeta. Here we demonstrate that ProF is a positive regulator of adipogenesis. Knockdown of ProF by RNA interference leads to decreased adipocyte differentiation. This is shown by reduced lipid accumulation, decreased expression of the differentiation markers PPARgamma and C/EBPalpha, and reduced glucose uptake in differentiated cells. Furthermore, ProF overexpression leads to increased adipogenesis. ProF binds to the transcription factor Foxo1 (Forkhead box O1), a negative regulator of insulin action and adipogenesis, and facilitates the phosphorylation and thus inactivation of Foxo1 by Akt. Additionally, dominant-negative Foxo1 restores adipogenesis in ProF knockdown cells. Thus, ProF modulates Foxo1 phosphorylation by Akt, promoting adipocyte differentiation. Furthermore, ProF might be involved in metabolic disorders such as diabetes.


Subject(s)
Adipogenesis/physiology , Carrier Proteins/physiology , Forkhead Transcription Factors/metabolism , Proto-Oncogene Proteins c-akt/metabolism , 3T3-L1 Cells , Adipocytes/cytology , Adipocytes/metabolism , Adipogenesis/genetics , Animals , Carrier Proteins/genetics , Carrier Proteins/metabolism , Cell Differentiation/genetics , Cell Differentiation/physiology , Cell Line , Forkhead Box Protein O1 , Forkhead Transcription Factors/genetics , Humans , Immunoblotting , Immunoprecipitation , Male , Mice , Mice, Inbred C57BL , Muscle Fibers, Skeletal/cytology , Muscle Fibers, Skeletal/metabolism , Phosphorylation , Protein Binding , Proto-Oncogene Proteins c-akt/genetics , RNA Interference , RNA, Small Interfering/genetics , Reverse Transcriptase Polymerase Chain Reaction , Transfection
20.
FEBS J ; 274(6): 1552-66, 2007 Mar.
Article in English | MEDLINE | ID: mdl-17313651

ABSTRACT

We have recently identified a protein, consisting of seven WD repeats, presumably forming a beta-propeller, and a domain identified in Fab1p, YOTB, VAC1p, and EEA1 (FYVE) domain, ProF. The FYVE domain targets the protein to vesicular membranes, while the WD repeats allow binding of the activated kinases Akt and protein kinase (PK)Czeta. Here, we describe the vesicle-associated membrane protein 2 (VAMP2) as interaction partner of ProF. The interaction is demonstrated with overexpressed and endogenous proteins in mammalian cells. ProF and VAMP2 partially colocalize on vesicular structures with PKCzeta and the proteins form a ternary complex. VAMP2 can be phosphorylated by activated PKCzeta in vitro and the presence of ProF increases the PKCzeta-dependent phosphorylation of VAMP2 in vitro. ProF is an adaptor protein that brings together a kinase with its substrate. VAMP2 is known to regulate docking and fusion of vesicles and to play a role in targeting vesicles to the plasma membrane. The complex may be involved in vesicle cycling in various secretory pathways.


Subject(s)
Carrier Proteins/metabolism , Protein Kinase C/metabolism , Vesicle-Associated Membrane Protein 2/metabolism , 3T3 Cells , Animals , COS Cells , Chlorocebus aethiops , Humans , Immunoprecipitation , Intracellular Signaling Peptides and Proteins , Mice , Microscopy, Confocal , Phosphorylation , Protein Binding , Substrate Specificity , Two-Hybrid System Techniques
SELECTION OF CITATIONS
SEARCH DETAIL
...