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










Publication year range
1.
Proc Natl Acad Sci U S A ; 120(9): e2214921120, 2023 02 28.
Article in English | MEDLINE | ID: mdl-36812200

ABSTRACT

Mutant KRAS is a major driver of oncogenesis in a multitude of cancers but remains a challenging target for classical small molecule drugs, motivating the exploration of alternative approaches. Here, we show that aggregation-prone regions (APRs) in the primary sequence of the oncoprotein constitute intrinsic vulnerabilities that can be exploited to misfold KRAS into protein aggregates. Conveniently, this propensity that is present in wild-type KRAS is increased in the common oncogenic mutations at positions 12 and 13. We show that synthetic peptides (Pept-ins™) derived from two distinct KRAS APRs could induce the misfolding and subsequent loss of function of oncogenic KRAS, both of recombinantly produced protein in solution, during cell-free translation and in cancer cells. The Pept-ins exerted antiproliferative activity against a range of mutant KRAS cell lines and abrogated tumor growth in a syngeneic lung adenocarcinoma mouse model driven by mutant KRAS G12V. These findings provide proof-of-concept that the intrinsic misfolding propensity of the KRAS oncoprotein can be exploited to cause its functional inactivation.


Subject(s)
Lung Neoplasms , Proto-Oncogene Proteins p21(ras) , Animals , Mice , Cell Line, Tumor , Lung Neoplasms/genetics , Mutation , Oncogene Proteins/metabolism , Proto-Oncogene Proteins p21(ras)/genetics , Protein Folding
2.
Proc Natl Acad Sci U S A ; 114(38): E7997-E8006, 2017 09 19.
Article in English | MEDLINE | ID: mdl-28874589

ABSTRACT

G protein-coupled receptors (GPCRs) are classically characterized as cell-surface receptors transmitting extracellular signals into cells. Here we show that central components of a GPCR signaling system comprised of the melatonin type 1 receptor (MT1), its associated G protein, and ß-arrestins are on and within neuronal mitochondria. We discovered that the ligand melatonin is exclusively synthesized in the mitochondrial matrix and released by the organelle activating the mitochondrial MT1 signal-transduction pathway inhibiting stress-mediated cytochrome c release and caspase activation. These findings coupled with our observation that mitochondrial MT1 overexpression reduces ischemic brain injury in mice delineate a mitochondrial GPCR mechanism contributing to the neuroprotective action of melatonin. We propose a new term, "automitocrine," analogous to "autocrine" when a similar phenomenon occurs at the cellular level, to describe this unexpected intracellular organelle ligand-receptor pathway that opens a new research avenue investigating mitochondrial GPCR biology.


Subject(s)
Brain Injuries/metabolism , Brain Ischemia/metabolism , Melatonin/biosynthesis , Mitochondria/metabolism , Receptor, Melatonin, MT1/metabolism , Signal Transduction , Animals , Brain Injuries/genetics , Brain Ischemia/genetics , Cytochromes c/genetics , Cytochromes c/metabolism , Male , Melatonin/genetics , Mice , Mitochondria/genetics , Receptor, Melatonin, MT1/genetics
3.
Nat Chem Biol ; 13(3): 259-261, 2017 03.
Article in English | MEDLINE | ID: mdl-28024151

ABSTRACT

Cells express several G-protein-coupled receptors (GPCRs) at their surfaces, transmitting simultaneous extracellular hormonal and chemical signals into cells. A comprehensive understanding of mechanisms underlying the integrated signaling response induced by distinct GPCRs is thus required. Here we found that the ß2-adrenergic receptor, which induces a short cAMP response, prolongs nuclear cAMP and protein kinase A (PKA) activation by promoting endosomal cAMP production in parathyroid hormone (PTH) receptor signaling through the stimulatory action of G protein Gßγ subunits on adenylate cyclase type 2.


Subject(s)
Endosomes/metabolism , GTP-Binding Protein beta Subunits/metabolism , GTP-Binding Protein gamma Subunits/metabolism , Receptor, Parathyroid Hormone, Type 1/metabolism , Receptors, Adrenergic, beta-2/metabolism , Signal Transduction , Cells, Cultured , HEK293 Cells , Humans , Protein Subunits/metabolism
4.
Front Immunol ; 7: 222, 2016.
Article in English | MEDLINE | ID: mdl-27375620

ABSTRACT

The cyclic AMP/protein kinase A (cAMP/PKA) pathway is one of the most common and versatile signal pathways in eukaryotic cells. A-kinase anchoring proteins (AKAPs) target PKA to specific substrates and distinct subcellular compartments providing spatial and temporal specificity for mediation of biological effects channeled through the cAMP/PKA pathway. In the immune system, cAMP is a potent negative regulator of T cell receptor-mediated activation of effector T cells (Teff) acting through a proximal PKA/Csk/Lck pathway anchored via a scaffold consisting of the AKAP Ezrin holding PKA, the linker protein EBP50, and the anchoring protein phosphoprotein associated with glycosphingolipid-enriched microdomains holding Csk. As PKA activates Csk and Csk inhibits Lck, this pathway in response to cAMP shuts down proximal T cell activation. This immunomodulating pathway in Teff mediates clinically important responses to regulatory T cell (Treg) suppression and inflammatory mediators, such as prostaglandins (PGs), adrenergic stimuli, adenosine, and a number of other ligands. A major inducer of T cell cAMP levels is PG E2 (PGE2) acting through EP2 and EP4 prostanoid receptors. PGE2 plays a crucial role in the normal physiological control of immune homeostasis as well as in inflammation and cancer immune evasion. Peripherally induced Tregs express cyclooxygenase-2, secrete PGE2, and elicit the immunosuppressive cAMP pathway in Teff as one tumor immune evasion mechanism. Moreover, a cAMP increase can also be induced by indirect mechanisms, such as intercellular transfer between T cells. Indeed, Treg, known to have elevated levels of intracellular cAMP, may mediate their suppressive function by transferring cAMP to Teff through gap junctions, which we speculate could also be regulated by PKA/AKAP complexes. In this review, we present an updated overview on the influence of cAMP-mediated immunoregulatory mechanisms acting through localized cAMP signaling and the therapeutical increasing prospects of AKAPs disruptors in T-cell immune function.

5.
J Biol Chem ; 288(51): 36426-36, 2013 Dec 20.
Article in English | MEDLINE | ID: mdl-24196963

ABSTRACT

The interaction between vascular cells and macrophages is critical during vascular remodeling. Here we report that the scaffolding protein, ezrin-binding phosphoprotein 50 (EBP50), is a central regulator of macrophage and vascular smooth muscle cells (VSMC) function. EBP50 is up-regulated in intimal VSMC following endoluminal injury and promotes neointima formation. However, the mechanisms underlying these effects are not fully understood. Because of the fundamental role that inflammation plays in vascular diseases, we hypothesized that EBP50 mediates macrophage activation and the response of vessels to inflammation. Indeed, EBP50 expression increased in primary macrophages and VSMC, and in the aorta of mice, upon treatment with LPS or TNFα. This increase was nuclear factor-κB (NF-κB)-dependent. Conversely, activation of NF-κB was impaired in EBP50-null VSMC and macrophages. We found that inflammatory stimuli promote the formation of an EBP50-PKCζ complex at the cell membrane that induces NF-κB signaling. Macrophage activation and vascular inflammation after acute LPS treatment were reduced in EBP50-null cells and mice as compared with WT. Furthermore, macrophage recruitment to vascular lesions was significantly reduced in EBP50 knock-out mice. Thus, EBP50 and NF-κB participate in a feed-forward loop leading to increased macrophage activation and enhanced response of vascular cells to inflammation.


Subject(s)
Feedback, Physiological , NF-kappa B/metabolism , Phosphoproteins/metabolism , Sodium-Hydrogen Exchangers/metabolism , Vasculitis/metabolism , Animals , Aorta/metabolism , Human Umbilical Vein Endothelial Cells/drug effects , Human Umbilical Vein Endothelial Cells/metabolism , Human Umbilical Vein Endothelial Cells/physiology , Humans , Lipopolysaccharides/pharmacology , Macrophages/metabolism , Macrophages/physiology , Mice , Mice, Inbred C57BL , Myocytes, Smooth Muscle/drug effects , Myocytes, Smooth Muscle/metabolism , Myocytes, Smooth Muscle/physiology , Phosphoproteins/genetics , Protein Kinase C/metabolism , Sodium-Hydrogen Exchangers/genetics , Tumor Necrosis Factor-alpha/pharmacology , Vasculitis/etiology
6.
J Biol Chem ; 288(39): 27849-60, 2013 Sep 27.
Article in English | MEDLINE | ID: mdl-23935101

ABSTRACT

The vasopressin type 2 receptor (V2R) is a critical G protein-coupled receptor (GPCR) for vertebrate physiology, including the balance of water and sodium ions. It is unclear how its two native hormones, vasopressin (VP) and oxytocin (OT), both stimulate the same cAMP/PKA pathway yet produce divergent antinatriuretic and antidiuretic effects that are either strong (VP) or weak (OT). Here, we present a new mechanism that differentiates the action of VP and OT on V2R signaling. We found that vasopressin, as opposed to OT, continued to generate cAMP and promote PKA activation for prolonged periods after ligand washout and receptor internalization in endosomes. Contrary to the classical model of arrestin-mediated GPCR desensitization, arrestins bind the VP-V2R complex yet extend rather than shorten the generation of cAMP. Signaling is instead turned off by the endosomal retromer complex. We propose that this mechanism explains how VP sustains water and Na(+) transport in renal collecting duct cells. Together with recent work on the parathyroid hormone receptor, these data support the existence of a novel "noncanonical" regulatory pathway for GPCR activation and response termination, via the sequential action of ß-arrestin and the retromer complex.


Subject(s)
Arrestins/metabolism , Gene Expression Regulation , Receptors, Vasopressin/metabolism , Signal Transduction , Animals , Antidiuretic Agents/pharmacology , Aquaporin 2/metabolism , Cell Membrane/metabolism , Cyclic AMP/metabolism , Dogs , Endosomes/metabolism , HEK293 Cells , Humans , Kidney/metabolism , Ligands , Madin Darby Canine Kidney Cells , Oxytocin/chemistry , Phosphorylation , Receptors, G-Protein-Coupled/metabolism , Sodium/metabolism , beta-Arrestins
7.
Methods Enzymol ; 522: 337-63, 2013.
Article in English | MEDLINE | ID: mdl-23374192

ABSTRACT

We describe optical and microscopy methods based on Förster resonance energy transfer, fluorescence recovery after photobleaching, and imaging cross-correlation spectroscopy that permit to determine kinetic and dynamic properties of key reactions involved G protein-coupled receptor (GPCR) signaling from the initial ligand binding step to the generation of the second messenger, cAMP. Well suited to determine rate-limiting reactions taking place along a GPCR signaling cascade in live cells, these techniques have also uncovered new concepts in GPCR signaling as well as many interesting mechanistic subtleties by which GPCRs transmit neurotransmitter and hormone signals into cells.


Subject(s)
Cyclic AMP/metabolism , Fluorescence Resonance Energy Transfer/methods , Heterotrimeric GTP-Binding Proteins/metabolism , Parathyroid Hormone/metabolism , Receptors, Parathyroid Hormone/metabolism , Signal Transduction/genetics , Arrestins/genetics , Arrestins/metabolism , Gene Expression , HEK293 Cells , Heterotrimeric GTP-Binding Proteins/genetics , Humans , Kinetics , Ligands , Microscopy, Confocal , Microscopy, Fluorescence , Photobleaching , Protein Binding , Protein Stability , Receptors, Parathyroid Hormone/genetics
8.
Proc Natl Acad Sci U S A ; 110(4): 1530-5, 2013 Jan 22.
Article in English | MEDLINE | ID: mdl-23297229

ABSTRACT

G protein-coupled receptors (GPCRs) participate in ubiquitous transmembrane signal transduction processes by activating heterotrimeric G proteins. In the current "canonical" model of GPCR signaling, arrestins terminate receptor signaling by impairing receptor-G-protein coupling and promoting receptor internalization. However, parathyroid hormone receptor type 1 (PTHR), an essential GPCR involved in bone and mineral metabolism, does not follow this conventional desensitization paradigm. ß-Arrestins prolong G protein (G(S))-mediated cAMP generation triggered by PTH, a process that correlates with the persistence of arrestin-PTHR complexes on endosomes and which is thought to be associated with prolonged physiological calcemic and phosphate responses. This presents an inescapable paradox for the current model of arrestin-mediated receptor-G-protein decoupling. Here we show that PTHR forms a ternary complex that includes arrestin and the Gßγ dimer in response to PTH stimulation, which in turn causes an accelerated rate of G(S) activation and increases the steady-state levels of activated G(S), leading to prolonged generation of cAMP. This work provides the mechanistic basis for an alternative model of GPCR signaling in which arrestins contribute to sustaining the effect of an agonist hormone on the receptor.


Subject(s)
Arrestins/metabolism , GTP-Binding Protein beta Subunits/metabolism , GTP-Binding Protein gamma Subunits/metabolism , Receptor, Parathyroid Hormone, Type 1/metabolism , Receptors, G-Protein-Coupled/metabolism , Arrestins/chemistry , Cyclic AMP/biosynthesis , Fluorescence Resonance Energy Transfer , GTP-Binding Protein beta Subunits/chemistry , GTP-Binding Protein gamma Subunits/chemistry , HEK293 Cells , Humans , Kinetics , Models, Biological , Multiprotein Complexes/chemistry , Multiprotein Complexes/metabolism , Parathyroid Hormone/metabolism , Parathyroid Hormone/pharmacology , Receptor, Parathyroid Hormone, Type 1/chemistry , Receptors, G-Protein-Coupled/chemistry , Recombinant Fusion Proteins/chemistry , Recombinant Fusion Proteins/metabolism , Signal Transduction , beta-Arrestins
9.
Trends Pharmacol Sci ; 33(8): 423-31, 2012 Aug.
Article in English | MEDLINE | ID: mdl-22709554

ABSTRACT

The classical model of arrestin-mediated desensitization of cell-surface G-protein-coupled receptors (GPCRs) is thought to be universal. However, this paradigm is incompatible with recent reports that the parathyroid hormone (PTH) receptor (PTHR), a crucial GPCR for bone and mineral ion metabolism, sustains G(S) activity and continues to generate cAMP for prolonged periods after ligand washout; during these periods the receptor is observed mainly in endosomes, associated with the bound ligand, G(S) and ß-arrestins. In this review we discuss possible molecular mechanisms underlying sustained signaling by the PTHR, including modes of signal generation and attenuation within endosomes, as well as the biological relevance of such non-canonical signaling.


Subject(s)
Receptors, Parathyroid Hormone/chemistry , Signal Transduction , Animals , Arrestins/metabolism , Cyclic AMP/metabolism , Endosomes/metabolism , GTP-Binding Proteins/metabolism , Humans , Protein Conformation , Receptors, Parathyroid Hormone/metabolism , beta-Arrestins
10.
Proc Natl Acad Sci U S A ; 109(19): 7433-8, 2012 May 08.
Article in English | MEDLINE | ID: mdl-22538810

ABSTRACT

Parathyroid hormone (PTH), the major calcium-regulating hormone, and norepinephrine (NE), the principal neurotransmitter of sympathetic nerves, regulate bone remodeling by activating distinct cell-surface G protein-coupled receptors in osteoblasts: the parathyroid hormone type 1 receptor (PTHR) and the ß(2)-adrenergic receptor (ß(2)AR), respectively. These receptors activate a common cAMP/PKA signal transduction pathway mediated through the stimulatory heterotrimeric G protein. Activation of ß(2)AR via the sympathetic nervous system decreases bone formation and increases bone resorption. Conversely, daily injection of PTH (1-34), a regimen known as intermittent (i)PTH treatment, increases bone mass through the stimulation of trabecular and cortical bone formation and decreases fracture incidences in severe cases of osteoporosis. Here, we show that iPTH has no osteoanabolic activity in mice lacking the ß(2)AR. ß(2)AR deficiency suppressed both iPTH-induced increase in bone formation and resorption. We showed that the lack of ß(2)AR blocks expression of iPTH-target genes involved in bone formation and resorption that are regulated by the cAMP/PKA pathway. These data implicate an unexpected functional interaction between PTHR and ß(2)AR, two G protein-coupled receptors from distinct families, which control bone formation and PTH anabolism.


Subject(s)
Bone and Bones/drug effects , Parathyroid Hormone/pharmacology , Receptor, Parathyroid Hormone, Type 1/metabolism , Receptors, Adrenergic, beta-2/metabolism , Absorptiometry, Photon , Anabolic Agents/metabolism , Anabolic Agents/pharmacology , Animals , Bone Density/drug effects , Bone and Bones/diagnostic imaging , Bone and Bones/metabolism , Female , Femur/drug effects , Femur/metabolism , Fluoresceins , Gene Expression Regulation/drug effects , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Mice, Transgenic , Osteogenesis/drug effects , Osteogenesis/genetics , Parathyroid Hormone/metabolism , Receptor, Parathyroid Hormone, Type 1/genetics , Receptors, Adrenergic, beta-2/genetics , Reverse Transcriptase Polymerase Chain Reaction , X-Ray Microtomography
11.
J Biol Chem ; 286(44): 38558-38569, 2011 Nov 04.
Article in English | MEDLINE | ID: mdl-21890629

ABSTRACT

Murine models indicate that Gαs and its extra-long variant XLαs, both of which are derived from GNAS, markedly differ regarding their cellular actions, but these differences are unknown. Here we investigated activation-induced trafficking of Gαs and XLαs, using immunofluorescence microscopy, cell fractionation, and total internal reflection fluorescence microscopy. In transfected cells, XLαs remained localized to the plasma membrane, whereas Gαs redistributed to the cytosol after activation by GTPase-inhibiting mutations, cholera toxin treatment, or G protein-coupled receptor agonists (isoproterenol or parathyroid hormone (PTH)(1-34)). Cholera toxin treatment or agonist (isoproterenol or pituitary adenylate cyclase activating peptide-27) stimulation of PC12 cells expressing Gαs and XLαs endogenously led to an increased abundance of Gαs, but not XLαs, in the soluble fraction. Mutational analyses revealed two conserved cysteines and the highly charged domain as being critically involved in the plasma membrane anchoring of XLαs. The cAMP response induced by M-PTH(1-14), a parathyroid hormone analog, terminated quickly in HEK293 cells stably expressing the type 1 PTH/PTH-related peptide receptor, whereas the response remained maximal for at least 6 min in cells that co-expressed the PTH receptor and XLαs. Although isoproterenol-induced cAMP response was not prolonged by XLαs expression, a GTPase-deficient XLαs mutant found in certain tumors and patients with fibrous dysplasia of bone and McCune-Albright syndrome generated more basal cAMP accumulation in HEK293 cells and caused more severe impairment of osteoblastic differentiation of MC3T3-E1 cells than the cognate Gαs mutant (gsp oncogene). Thus, activated XLαs and Gαs traffic differently, and this may form the basis for the differences in their cellular actions.


Subject(s)
GTP-Binding Protein alpha Subunits, Gs/chemistry , 3T3 Cells , Animals , Cell Differentiation , Cell Membrane/metabolism , Cholera Toxin/chemistry , Chromogranins , Cyclic AMP/metabolism , Fluorescence Resonance Energy Transfer , HEK293 Cells , Humans , Mice , Osteoblasts/cytology , PC12 Cells , Rats , Receptor, Parathyroid Hormone, Type 1/metabolism , Signal Transduction
12.
Nat Chem Biol ; 7(5): 278-84, 2011 May.
Article in English | MEDLINE | ID: mdl-21445058

ABSTRACT

The generation of cAMP by G protein-coupled receptors (GPCRs) and its termination are currently thought to occur exclusively at the plasma membrane of cells. Under existing models of receptor regulation, this signal is primarily restricted by desensitization of the receptors through their binding to ß-arrestins. However, this paradigm is not consistent with recent observations that the parathyroid hormone receptor type 1 (PTHR) continues to stimulate cAMP production even after receptor internalization, as ß-arrestins are known to rapidly bind and internalize activated PTHR. Here we show that binding to ß-arrestin1 prolongs rather than terminates the generation of cAMP by PTHR, and that cAMP generation correlates with the persistence of arrestin-receptor complexes on endosomes. PTHR signaling is instead turned off by the retromer complex, which regulates the movement of internalized receptor from endosomes to the Golgi apparatus. Thus, binding by the retromer complex regulates the sustained generation of cAMP triggered by an internalized GPCR.


Subject(s)
Cyclic AMP/metabolism , Endosomes/metabolism , Golgi Apparatus/metabolism , Receptor, Parathyroid Hormone, Type 1/metabolism , Arrestins/metabolism , Fluorescence Resonance Energy Transfer , HEK293 Cells , Humans , Models, Biological , Receptor, Parathyroid Hormone, Type 1/genetics , Signal Transduction , Time Factors , beta-Arrestins
SELECTION OF CITATIONS
SEARCH DETAIL
...