Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 33
Filter
1.
Glia ; 68(9): 1794-1809, 2020 09.
Article in English | MEDLINE | ID: mdl-32077526

ABSTRACT

Finding causative genetic mutations is important in the diagnosis and treatment of hereditary peripheral neuropathies. This study was conducted to find new genes involved in the pathophysiology of hereditary peripheral neuropathy. We identified a new mutation in the EBP50 gene, which is co-segregated with neuropathic phenotypes, including motor and sensory deficit in a family with Charcot-Marie-Tooth disease. EBP50 is known to be important for the formation of microvilli in epithelial cells, and the discovery of this gene mutation allowed us to study the function of EBP50 in the nervous system. EBP50 was strongly expressed in the nodal and paranodal regions of sciatic nerve fibers, where Schwann cell microvilli contact the axolemma, and at the growth tips of primary Schwann cells. In addition, EBP50 expression was decreased in mouse models of peripheral neuropathy. Knockout mice were used to study EBP50 function in the peripheral nervous system. Interestingly motor function deficit and abnormal histology of nerve fibers were observed in EBP50+/- heterozygous mice at 12 months of age, but not 3 months. in vitro studies using Schwann cells showed that NRG1-induced AKT activation and migration were significantly reduced in cells overexpressing the I325V mutant of EBP50 or cells with knocked-down EBP50 expression. In conclusion, we show for the first time that loss of function due to EBP50 gene deficiency or mutation can cause peripheral neuropathy.


Subject(s)
Charcot-Marie-Tooth Disease , Animals , Charcot-Marie-Tooth Disease/genetics , Mice , Mice, Knockout , Mutation , Peripheral Nerves , Peripheral Nervous System
2.
J Biol Chem ; 293(15): 5556-5571, 2018 04 13.
Article in English | MEDLINE | ID: mdl-29444827

ABSTRACT

G protein-coupled receptor (GPCR) signaling and trafficking are essential for cellular function and regulated by phosphorylation, ß-arrestin, and ubiquitination. The GPCR parathyroid hormone receptor (PTHR) exhibits time-dependent reversible ubiquitination. The exact ubiquitination sites in PTHR are unknown, but they extend upstream of its intracellular tail. Here, using tandem MS, we identified Lys388 in the third loop and Lys484 in the C-terminal tail as primary ubiquitination sites in PTHR. We found that PTHR ubiquitination requires ß-arrestin and does not display a preference for ß-arrestin1 or -2. PTH stimulated PTHR phosphorylation at Thr387/Thr392 and within the Ser489-Ser493 region. Such phosphorylation events may recruit ß-arrestin, and we observed that chemically or genetically blocking PTHR phosphorylation inhibits its ubiquitination. Specifically, Ala replacement at Thr387/Thr392 suppressed ß-arrestin binding and inhibited PTHR ubiquitination, suggesting that PTHR phosphorylation and ubiquitination are interdependent. Of note, Lys-deficient PTHR mutants promoted normal cAMP formation, but exhibited differential mitogen-activated protein kinase (MAPK) signaling. Lys-deficient PTHR triggered early onset and delayed ERK1/2 signaling compared with wildtype PTHR. Moreover, ubiquitination of Lys388 and Lys484 in wildtype PTHR strongly decreased p38 signaling, whereas Lys-deficient PTHR retained signaling comparable to unstimulated wildtype PTHR. Lys-deficient, ubiquitination-refractory PTHR reduced cell proliferation and increased apoptosis. However, elimination of all 11 Lys residues in PTHR did not affect its internalization and recycling. These results pinpoint the ubiquitinated Lys residues in PTHR controlling MAPK signaling and cell proliferation and survival. Our findings suggest new opportunities for targeting PTHR ubiquitination to regulate MAPK signaling or manage PTHR-related disorders.


Subject(s)
Cell Proliferation , MAP Kinase Signaling System , Mutation, Missense , Receptor, Parathyroid Hormone, Type 1/metabolism , Ubiquitination , Amino Acid Substitution , Animals , Cell Survival/genetics , HEK293 Cells , Humans , Mice , Receptor, Parathyroid Hormone, Type 1/genetics , p38 Mitogen-Activated Protein Kinases/genetics , p38 Mitogen-Activated Protein Kinases/metabolism
3.
Proc Natl Acad Sci U S A ; 113(36): E5308-17, 2016 09 06.
Article in English | MEDLINE | ID: mdl-27540115

ABSTRACT

Despite numerous reports implicating NADPH oxidases (Nox) in the pathogenesis of many diseases, precise regulation of this family of professional reactive oxygen species (ROS) producers remains unclear. A unique member of this family, Nox1 oxidase, functions as either a canonical or hybrid system using Nox organizing subunit 1 (NoxO1) or p47(phox), respectively, the latter of which is functional in vascular smooth muscle cells (VSMC). In this manuscript, we identify critical requirement of ezrin-radixin-moesin-binding phosphoprotein 50 (EBP50; aka NHERF1) for Nox1 activation and downstream responses. Superoxide (O2 (•-)) production induced by angiotensin II (AngII) was absent in mouse EBP50 KO VSMC vs. WT. Moreover, ex vivo incubation of aortas with AngII showed a significant increase in O2 (•-) in WT but not EBP50 or Nox1 nulls. Similarly, lipopolysaccharide (LPS)-induced oxidative stress was attenuated in femoral arteries from EBP50 KO vs. WT. In silico analyses confirmed by confocal microscopy, immunoprecipitation, proximity ligation assay, FRET, and gain-/loss-of-function mutagenesis revealed binding of EBP50, via its PDZ domains, to a specific motif in p47(phox) Functional studies revealed AngII-induced hypertrophy was absent in EBP50 KOs, and in VSMC overexpressing EBP50, Nox1 gene silencing abolished VSMC hypertrophy. Finally, ex vivo measurement of lumen diameter in mouse resistance arteries exhibited attenuated AngII-induced vasoconstriction in EBP50 KO vs. WT. Taken together, our data identify EBP50 as a previously unidentified regulator of Nox1 and support that it promotes Nox1 activity by binding p47(phox) This interaction is pivotal for agonist-induced smooth muscle ROS, hypertrophy, and vasoconstriction and has implications for ROS-mediated physiological and pathophysiological processes.


Subject(s)
ATPases Associated with Diverse Cellular Activities/metabolism , DNA Helicases/metabolism , Hypertrophy/metabolism , NADPH Oxidase 1/genetics , Phosphoproteins/metabolism , Proteins/metabolism , Sodium-Hydrogen Exchangers/metabolism , ATPases Associated with Diverse Cellular Activities/genetics , Adaptor Proteins, Signal Transducing , Angiotensin II/administration & dosage , Angiotensin II/adverse effects , Animals , DNA Helicases/genetics , Femoral Artery/drug effects , Femoral Artery/metabolism , Femoral Artery/pathology , Humans , Hypertrophy/chemically induced , Hypertrophy/pathology , Lipopolysaccharides/toxicity , Mice , Mice, Knockout , Muscle, Smooth, Vascular/metabolism , Muscle, Smooth, Vascular/pathology , NADPH Oxidase 1/metabolism , Oxidative Stress/drug effects , Phosphoproteins/genetics , Proteins/genetics , Reactive Oxygen Species/metabolism , Sodium-Hydrogen Exchangers/genetics , Superoxides/metabolism , Vasoconstriction/drug effects , Vasoconstriction/genetics
4.
Biochemistry ; 56(20): 2584-2593, 2017 05 23.
Article in English | MEDLINE | ID: mdl-28376304

ABSTRACT

Na+/H+ exchanger regulatory factor-1 (NHERF1) is a scaffolding protein containing two PSD95/discs large protein/ZO1 (PDZ) domains that modifies the signaling, trafficking, and function of the parathyroid hormone receptor (PTHR), a family B G-protein-coupled receptor. PTHR and NHERF1 bind through a PDZ-ligand-recognition mechanism. We show that PTH elicits phosphorylation of Thr591 in the canonical -ETVM binding motif of PTHR. Conservative substitution of Thr591 with Cys does not affect PTH(1-34)-induced cAMP production or binding of PTHR to NHERF1. The findings suggested the presence of additional sites upstream of the PDZ-ligand motif through which the two proteins interact. Structural determinants outside the canonical NHERF1 PDZ-PTHR interface that influence binding have not been characterized. We used molecular dynamics (MD) simulation to predict residues involved in these interactions. Simulation data demonstrate that the negatively charged Glu side chains at positions -3, -5, and -6 upstream of the PDZ binding motif are involved in PDZ-PTHR recognition. Engineered mutant peptides representing the PTHR C-terminal region were used to measure the binding affinity with NHERF1 PDZ domains. Comparable micromolar affinities for peptides of different length were confirmed by fluorescence polarization, isothermal titration calorimetry, and surface plasmon resonance. Binding affinities measured for Ala variants validate MD simulations. The linear relation between the change in enthalpy and entropy following Ala substitutions at upstream positions -3, -5, and -6 of the PTHR peptide provides a clear example of the thermodynamic compensation rule. Overall, our data highlight sequences in PTHR that contribute to NHERF1 interaction and can be altered to prevent phosphorylation-mediated inhibition.


Subject(s)
Computational Biology , PDZ Domains , Phosphoproteins/metabolism , Receptor, Parathyroid Hormone, Type 1/metabolism , Sodium-Hydrogen Exchangers/metabolism , Amino Acid Sequence , Calorimetry , Cyclic AMP/biosynthesis , Fluorescence Polarization , HEK293 Cells , Humans , Molecular Dynamics Simulation , Phosphoproteins/chemistry , Phosphorylation , Sodium-Hydrogen Exchangers/chemistry , Spectrometry, Mass, Electrospray Ionization , Surface Plasmon Resonance
5.
J Biol Chem ; 291(21): 10986-1002, 2016 May 20.
Article in English | MEDLINE | ID: mdl-27008860

ABSTRACT

The G protein-coupled parathyroid hormone receptor (PTHR) regulates mineral-ion homeostasis and bone remodeling. Upon parathyroid hormone (PTH) stimulation, the PTHR internalizes into early endosomes and subsequently traffics to the retromer complex, a sorting platform on early endosomes that promotes recycling of surface receptors. The C terminus of the PTHR contains a type I PDZ ligand that binds PDZ domain-containing proteins. Mass spectrometry identified sorting nexin 27 (SNX27) in isolated endosomes as a PTHR binding partner. PTH treatment enriched endosomal PTHR. SNX27 contains a PDZ domain and serves as a cargo selector for the retromer complex. VPS26, VPS29, and VPS35 retromer subunits were isolated with PTHR in endosomes from cells stimulated with PTH. Molecular dynamics and protein binding studies establish that PTHR and SNX27 interactions depend on the PDZ recognition motif in PTHR and the PDZ domain of SNX27. Depletion of either SNX27 or VPS35 or actin depolymerization decreased the rate of PTHR recycling following agonist stimulation. Mutating the PDZ ligand of PTHR abolished the interaction with SNX27 but did not affect the overall rate of recycling, suggesting that PTHR may directly engage the retromer complex. Coimmunoprecipitation and overlay experiments show that both intact and mutated PTHR bind retromer through the VPS26 protomer and sequentially assemble a ternary complex with PTHR and SNX27. SNX27-independent recycling may involve N-ethylmaleimide-sensitive factor, which binds both PDZ intact and mutant PTHRs. We conclude that PTHR recycles rapidly through at least two pathways, one involving the ASRT complex of actin, SNX27, and retromer and another possibly involving N-ethylmaleimide-sensitive factor.


Subject(s)
Actins/metabolism , Receptor, Parathyroid Hormone, Type 1/metabolism , Sorting Nexins/metabolism , Actins/chemistry , Animals , CHO Cells , Cricetulus , Endosomes/metabolism , HEK293 Cells , Humans , Metabolic Networks and Pathways , Molecular Dynamics Simulation , Multiprotein Complexes/chemistry , Multiprotein Complexes/genetics , Multiprotein Complexes/metabolism , N-Ethylmaleimide-Sensitive Proteins/metabolism , PDZ Domains , Protein Binding , Protein Subunits , Protein Transport , Proteolysis , Receptor, Parathyroid Hormone, Type 1/chemistry , Receptor, Parathyroid Hormone, Type 1/genetics , Recombinant Proteins/chemistry , Recombinant Proteins/genetics , Recombinant Proteins/metabolism , Sorting Nexins/chemistry , Sorting Nexins/genetics
6.
J Biol Chem ; 290(5): 2879-87, 2015 Jan 30.
Article in English | MEDLINE | ID: mdl-25492869

ABSTRACT

The regulation of the cell cycle by the ubiquitin-proteasome system is dependent on the activity of E3 ligases. Skp2 (S-phase kinase associated protein-2) is the substrate recognition subunit of the E3 ligase that ubiquitylates the cell cycle inhibitors p21(cip1) and p27(kip1) thus promoting cell cycle progression. Increased expression of Skp2 is frequently observed in diseases characterized by excessive cell proliferation, such as cancer and neointima hyperplasia. The stability and cellular localization of Skp2 are regulated by Akt, but the molecular mechanisms underlying these effects remain only partly understood. The scaffolding protein Ezrin-Binding Phosphoprotein of 50 kDa (EBP50) contains two PDZ domains and plays a critical role in the development of neointimal hyperplasia. Here we report that EBP50 directly binds Skp2 via its first PDZ domain. Moreover, EBP50 is phosphorylated by Akt on Thr-156 within the second PDZ domain, an event that allosterically promotes binding to Skp2. The interaction with EBP50 causes cytoplasmic localization of Skp2, increases Skp2 stability and promotes proliferation of primary vascular smooth muscle cells. Collectively, these studies define a novel regulatory mechanism contributing to aberrant cell growth and highlight the importance of scaffolding function of EBP50 in Akt-dependent cell proliferation.


Subject(s)
Phosphoproteins/metabolism , Proto-Oncogene Proteins c-akt/metabolism , S-Phase Kinase-Associated Proteins/metabolism , Sodium-Hydrogen Exchangers/metabolism , Animals , Cell Proliferation , Cells, Cultured , Humans , Mice , Phosphoproteins/chemistry , Phosphorylation , Protein Binding , Protein Stability , Proto-Oncogene Proteins c-akt/chemistry , S-Phase Kinase-Associated Proteins/chemistry , Sodium-Hydrogen Exchangers/chemistry
7.
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
8.
J Am Chem Soc ; 136(37): 12848-51, 2014 Sep 17.
Article in English | MEDLINE | ID: mdl-25191938

ABSTRACT

Glucagon-like peptide-1 (GLP-1) is a natural agonist for GLP-1R, a G protein-coupled receptor (GPCR) on the surface of pancreatic ß cells. GLP-1R agoinsts are attractive for treatment of type 2 diabetes, but GLP-1 itself is rapidly degraded by peptidases in vivo. We describe a design strategy for retaining GLP-1-like activity while engendering prolonged activity in vivo, based on strategic replacement of native α residues with conformationally constrained ß-amino acid residues. This backbone-modification approach may be useful for developing stabilized analogues of other peptide hormones.


Subject(s)
Glucagon-Like Peptide 1/analogs & derivatives , Glucagon-Like Peptide 1/pharmacology , Receptors, Glucagon/agonists , Amino Acid Sequence , Animals , Cells, Cultured , Diabetes Mellitus, Type 2/drug therapy , Glucagon-Like Peptide 1/chemistry , Glucagon-Like Peptide 1/metabolism , Glucagon-Like Peptide-1 Receptor , Humans , Mice , Molecular Sequence Data , Protein Stability
9.
J Biol Chem ; 287(29): 24148-63, 2012 Jul 13.
Article in English | MEDLINE | ID: mdl-22628548

ABSTRACT

Congenital defects in the Na/H exchanger regulatory factor-1 (NHERF1) are linked to disordered phosphate homeostasis and skeletal abnormalities in humans. In the kidney, these mutations interrupt parathyroid hormone (PTH)-responsive sequestration of the renal phosphate transporter, Npt2a, with ensuing urinary phosphate wasting. We now report that NHERF1, a modular PDZ domain scaffolding protein, coordinates the assembly of an obligate ternary complex with Npt2a and the PKA-anchoring protein ezrin to facilitate PTH-responsive cAMP signaling events. Activation of ezrin-anchored PKA initiates NHERF1 phosphorylation to disassemble the ternary complex, release Npt2a, and thereby inhibit phosphate transport. Loss-of-function mutations stabilize an inactive NHERF1 conformation that we show is refractory to PKA phosphorylation and impairs assembly of the ternary complex. Compensatory mutations introduced in mutant NHERF1 re-establish the integrity of the ternary complex to permit phosphorylation of NHERF1 and rescue PTH action. These findings offer new insights into a novel macromolecular mechanism for the physiological action of a critical ternary complex, where anchored PKA coordinates the assembly and turnover of the Npt2a-NHERF1-ezrin complex.


Subject(s)
Cyclic AMP-Dependent Protein Kinases/metabolism , Cytoskeletal Proteins/metabolism , Phosphates/metabolism , Phosphoproteins/metabolism , Sodium-Hydrogen Exchangers/metabolism , Animals , Biological Transport/physiology , Cell Line , Humans , Immunoblotting , Immunoprecipitation , Phosphoproteins/chemistry , Phosphorylation , Sodium-Hydrogen Exchangers/chemistry , Sodium-Phosphate Cotransporter Proteins, Type IIa/metabolism
10.
Arterioscler Thromb Vasc Biol ; 32(1): 33-41, 2012 Jan.
Article in English | MEDLINE | ID: mdl-22034511

ABSTRACT

OBJECTIVE: The Ezrin-radixin-moesin-binding phosphoprotein 50 (EBP50) is a scaffolding protein known to regulate ion homeostasis in the kidney and intestine. Previous work showed that EBP50 expression increases after balloon injury in rat carotids. This study was designed to determine the role of EBP50 on vascular smooth muscle cells (VSMC) proliferation and the development of neointimal hyperplasia. METHODS AND RESULTS: Wire injury was performed in wild type (WT) and EBP50 knockout (KO) mice. Two weeks after injury, neointima formation was 80% lower in KO than in WT mice. Proliferation of KO VSMC was significantly lower than WT cells and overexpression of EBP50 increased VSMC proliferation. Akt activity and expression of S-phase kinase protein2 decreased in KO cells resulting in the stabilization of the cyclin-dependent kinase inhibitor, p21(cip1). Consequently, KO cells were arrested in G(0)/G(1) phase. Consistent with these observations, p21(cip1) was detected in injured femoral arteries of KO but not WT mice. No differences in apoptosis between WT and KO were observed. CONCLUSIONS: EBP50 is critical for neointima formation and induces VSMC proliferation by decreasing S-phase kinase protein2 stability, thereby accelerating the degradation of the cell cycle inhibitor p21(cip1).


Subject(s)
Cyclin-Dependent Kinase Inhibitor p21/physiology , Myocytes, Smooth Muscle/cytology , Myocytes, Smooth Muscle/physiology , Neointima/etiology , Phosphoproteins/physiology , S-Phase Kinase-Associated Proteins/physiology , Sodium-Hydrogen Exchangers/physiology , Animals , Cell Proliferation , Femoral Artery/injuries , Femoral Artery/pathology , Femoral Artery/physiopathology , Mice , Mice, Inbred C57BL , Mice, Knockout , Neointima/pathology , Neointima/physiopathology , Phosphoproteins/deficiency , Phosphoproteins/genetics , Proto-Oncogene Proteins c-akt/metabolism , Rats , Sodium-Hydrogen Exchangers/genetics
11.
Nat Med ; 12(4): 425-32, 2006 Apr.
Article in English | MEDLINE | ID: mdl-16582918

ABSTRACT

The mechanisms underlying leptin resistance are still being defined. We report here the presence in human blood of several serum leptin-interacting proteins (SLIPs), isolated by leptin-affinity chromatography and identified by mass spectrometry and immunochemical analysis. We confirmed that one of the major SLIPs is C-reactive protein (CRP). In vitro, human CRP directly inhibits the binding of leptin to its receptors and blocks its ability to signal in cultured cells. In vivo, infusion of human CRP into ob/ob mice blocked the effects of leptin upon satiety and weight reduction. In mice that express a transgene encoding human CRP, the actions of human leptin were completely blunted. We also found that physiological concentrations of leptin can stimulate expression of CRP in human primary hepatocytes. Recently, human CRP has been correlated with increased adiposity and plasma leptin. Thus, our results suggest a potential mechanism contributing to leptin resistance, by which circulating CRP binds to leptin and attenuates its physiological functions.


Subject(s)
C-Reactive Protein/metabolism , Leptin/metabolism , Animals , Blotting, Western , Body Weight/drug effects , C-Reactive Protein/pharmacology , Carrier Proteins/isolation & purification , Carrier Proteins/metabolism , Cells, Cultured , Dose-Response Relationship, Drug , Drug Interactions , Female , Hepatocytes/drug effects , Hepatocytes/metabolism , Humans , Inhibitory Concentration 50 , Interleukin-6/pharmacology , Leptin/blood , Leptin/pharmacology , Mice , Mice, Obese , Mice, Transgenic , Precipitin Tests , RNA-Binding Proteins , Rats , Transgenes
12.
J Mol Cell Cardiol ; 53(6): 809-19, 2012 Dec.
Article in English | MEDLINE | ID: mdl-22974528

ABSTRACT

The ezrin-radixin-moesin-binding phosphoprotein 50 (EBP50) is a PDZ-containing scaffolding protein that regulates a variety of physiological functions. In the vasculature, EBP50 promotes neointima formation following arterial injury. In this study the role of EBP50 on vascular smooth muscle cell (VSMC) migration was characterized. The spreading and motility of primary VSMC isolated from EBP50 knockout (KO) mice were significantly reduced compared to wild-type (WT) cells. EBP50-null VSMC had fewer and larger focal adhesions than wild-type cells. Assembly and disassembly of focal adhesion-assessed by live-cell total internal reflection fluorescence imaging-in response to epidermal growth factor (EGF) were significantly reduced in KO cells. Immunoprecipitation experiments showed that EBP50 interacts with EGF receptor via the PDZ2 domain and with focal adhesion kinase (FAK) via the C-terminal ERM domain. EBP50 promoted the formation of a complex containing both EGF receptor and FAK. Phosphorylation of Tyr-925 of FAK in response to EGF was significantly reduced in KO cell compared to WT cells. The residence time of FAK in focal adhesions-determined by fluorescence recovery after photobleaching-was increased in WT cells. Collectively, these studies indicate that EBP50, by scaffolding EGF receptor and FAK, facilitates activation of FAK, focal adhesion turnover, and migration of VSMC.


Subject(s)
Blood Vessels/metabolism , Cell Movement , Focal Adhesions/metabolism , Myocytes, Smooth Muscle/metabolism , Phosphoproteins/metabolism , Sodium-Hydrogen Exchangers/metabolism , Animals , Cell Line , Cell Movement/drug effects , Cell Movement/genetics , Epidermal Growth Factor/pharmacology , ErbB Receptors/metabolism , Focal Adhesion Kinase 1/metabolism , Focal Adhesions/drug effects , Focal Adhesions/genetics , Mice , Mice, Knockout , Myocytes, Smooth Muscle/drug effects , Phosphoproteins/genetics , Phosphorylation/drug effects , Protein Binding , Sodium-Hydrogen Exchangers/genetics
13.
J Mol Cell Cardiol ; 49(6): 1012-21, 2010 Dec.
Article in English | MEDLINE | ID: mdl-20843475

ABSTRACT

Parathyroid hormone-related protein (PTHrP) and the parathyroid hormone type 1 receptor (PTH1R) are important regulators of vascular remodeling. PTHrP expression is associated to increased proliferation of vascular smooth muscle cells (VSMC). In contrast, signaling via the PTH1R inhibits cell growth. The mechanisms regulating the dual effect of PTHrP and PTH1R on VSMC proliferation are only partially understood. In this study we examined the role of the adaptor protein ezrin-radixin-moesin-binding phosphoprotein (EBP50) on PTH1R expression, trafficking, signaling and control of A10 cell proliferation. In normal rat vascular tissues, EBP50 was restricted to the endothelium with little expression in VSMC. EBP50 expression significantly increased in VSMC following angioplasty in parallel with PTHrP. Interestingly, PTHrP was able to induce EBP50 expression. In the clonal rat aortic smooth muscle cell line A10, EBP50 increased the recruitment of PTH1R to the cell membrane and delayed its internalization in response to PTHrP(1-36). This effect required an intact C-terminal motif in the PTH1R. In naïve A10 cells, PTHrP(1-36) stimulated cAMP production but not intracellular calcium release. In contrast, PTHrP(1-36) induced both cAMP and calcium signaling in A10 cells over-expressing EBP50. Finally, EBP50 attenuated the induction of p27(kip1) and the anti-proliferative effect of PTHrP(1-36). In summary, this study demonstrates the dynamic expression of EBP50 in vessels following injury and the effects of EBP50 on PTH1R function in VSMC. These findings highlight one of the mechanisms leading to increased VSMC proliferation and have important implication in the understanding of the molecular events leading to restenosis.


Subject(s)
Carrier Proteins/metabolism , Mitogens/metabolism , Muscle, Smooth, Vascular/metabolism , Myocytes, Smooth Muscle/cytology , Phosphoproteins/metabolism , Receptor, Parathyroid Hormone, Type 1/metabolism , Angioplasty , Animals , Carotid Arteries/metabolism , Carotid Arteries/surgery , Cell Proliferation , Endocytosis , HEK293 Cells , Humans , Male , Models, Biological , Neointima/metabolism , Neointima/pathology , Protein Transport , Rats , Rats, Sprague-Dawley , Signal Transduction , Sodium-Hydrogen Exchangers , Up-Regulation
14.
Mol Endocrinol ; 22(5): 1163-70, 2008 May.
Article in English | MEDLINE | ID: mdl-18202147

ABSTRACT

The effects of the expression of the Na+/H+ exchanger regulatory factor-1 (NHERF1) on the distribution, dynamics, and signaling properties of the PTH type 1 receptor (PTH1R) were studied in rat osteosarcoma cells ROS 17/2.8. NHERF1 had a dramatic effect on the subcellular distribution of PTH1R, promoting a substantial relocation of the receptor to regions of the plasma membrane located in very close proximity to cytoskeletal fibers. Direct interactions of NHERF1 with the PTH1R and the cytoskeleton were required for these effects, because they were abolished by 1) PTH1R mutations that impair NHERF1 binding, and 2) NHERF1 mutations that impair binding to the PTH1R or the cytoskeleton. NHERF1 reduced significantly the diffusion of the PTH1R by a mechanism that was also dependent on a direct association of NHERF1 with the PTH1R and the cytoskeleton. NHERF1 increased ligand-dependent production of cAMP and induced ligand-dependent rises in intracellular calcium. These effects on calcium were due to increased calcium uptake, as they were blocked by calcium channel inhibitors and by the addition of EGTA to the medium. These calcium effects were abolished by protein kinase A inhibition but phospholipase C inhibition was without effect. Based on these analyses, we propose that, in ROS cells, the presence of NHERF1 induces PTH-dependent calcium signaling by a cAMP-mediated mechanism that involves local protein kinase A-dependent activation of calcium channels.


Subject(s)
Phosphoproteins/physiology , Receptor, Parathyroid Hormone, Type 1/physiology , Signal Transduction , Sodium-Hydrogen Exchangers/physiology , Animals , Biological Transport , Calcium/metabolism , Cell Line, Tumor , Humans , Osteosarcoma/genetics , Osteosarcoma/metabolism , Osteosarcoma/pathology , Phosphoproteins/genetics , Phosphoproteins/metabolism , Rats , Receptor, Parathyroid Hormone, Type 1/genetics , Receptor, Parathyroid Hormone, Type 1/metabolism , Sodium-Hydrogen Exchangers/genetics , Sodium-Hydrogen Exchangers/metabolism
15.
Circ Res ; 99(9): 933-42, 2006 Oct 27.
Article in English | MEDLINE | ID: mdl-17023675

ABSTRACT

Parathyroid hormone-related protein (PTHrP) is present in vascular smooth muscle (VSM), is markedly upregulated in response to arterial injury, is essential for normal VSM proliferation, and also markedly accentuates neointima formation following rat carotid angioplasty. PTHrP contains a nuclear localization signal (NLS) through which it enters the nucleus and leads to marked increases in retinoblastoma protein (pRb) phosphorylation and cell cycle progression. Our goal was to define key cell cycle molecules upstream of pRb that mediate cell cycle acceleration induced by PTHrP. The cyclin D/cdk-4,-6 system and its upstream regulators, the inhibitory kinases (INKs), are not appreciably influenced by PTHrP. In striking contrast, cyclin E/cdk-2 kinase activity is markedly increased by PTHrP, and this is a result of a specific, marked, PTHrP-induced proteasomal degradation of p27(kip1). Adenoviral restoration of p27(kip1) fully reverses PTHrP-induced cell cycle progression, indicating that PTHrP mediates its cell cycle acceleration in VSM via p27(kip1). In confirmation, adenoviral delivery of PTHrP to murine primary vascular smooth muscle cells (VSMCs) significantly decreases p27(kip1) expression and accelerates cell cycle progression. p27(kip1) is well known to be a central cell cycle regulatory molecule involved in both normal and pathological VSM proliferation and is a target of widely used drug-eluting stents. The current observations define a novel "PTHrP/p27(kip1) pathway" in the arterial wall and suggest that this pathway is important in normal arterial biology and a potential target for therapeutic manipulation of the arterial response to injury.


Subject(s)
Arteries/metabolism , Cell Cycle , Cyclin-Dependent Kinase Inhibitor p27/metabolism , Muscle, Smooth, Vascular/metabolism , Parathyroid Hormone-Related Protein/metabolism , Animals , Arteries/cytology , Cell Cycle Proteins/metabolism , Cell Line , Cell Proliferation , Cells, Cultured , Down-Regulation , G1 Phase , Mice , Muscle, Smooth, Vascular/cytology , Nuclear Proteins/metabolism , Proteasome Endopeptidase Complex/metabolism , Proteome/metabolism , Rats , S Phase , Signal Transduction
16.
Mol Endocrinol ; 20(12): 3400-11, 2006 Dec.
Article in English | MEDLINE | ID: mdl-16931572

ABSTRACT

The glucagon-like peptide 1 receptor (GLP-1R) mediates important effects on beta-cell function and glucose homeostasis and is one of the most promising therapeutic targets for type 2, and possibly type 1, diabetes. Yet, little is known regarding the molecular and cellular mechanisms that regulate its function. Therefore, we examined the cellular trafficking of the GLP-1R and the relation between receptor localization and signaling activity. In resting human embryonic kidney 293 and insulinoma MIN6 cells, a fully functional green fluorescent protein-tagged GLP-1R was localized both at the cell membrane and in highly mobile intracellular compartments. Real-time confocal fluorescence microscopy allowed direct visualization of constitutive cycling of the receptor. Overexpression of K44A-dynamin increased the number of functional receptors at the cell membrane. Immunoprecipitation, sucrose sedimentation, and microscopy observations demonstrated that the GLP-1R localizes in lipid rafts and interacts with caveolin-1. This interaction is necessary for membrane localization of the GLP-1R, because overexpression of a dominant-negative form of caveolin-1 (P132L-cav1) or specific mutations within the putative GLP-1R's caveolin-1 binding domain completely inhibited GLP-1 binding and activity. Upon agonist stimulation, the GLP-1R underwent rapid and extensive endocytosis independently from arrestins but in association with caveolin-1. Finally, GLP-1R-stimulated activation of ERK1/2, which involves transactivation of epidermal growth factor receptors, required lipid raft integrity. In summary, the interaction of the GLP-1R with caveolin-1 regulates subcellular localization, trafficking, and signaling activity. This study provides further evidence of the key role of accessory proteins in specifying the cellular behavior of G protein-coupled receptors.


Subject(s)
Caveolin 1/metabolism , Cell Membrane/metabolism , Receptors, Glucagon/metabolism , Animals , Caveolin 1/genetics , Cell Membrane/chemistry , Cells, Cultured , Glucagon-Like Peptide-1 Receptor , Humans , Membrane Microdomains/chemistry , Membrane Microdomains/metabolism , Membrane Microdomains/ultrastructure , Mice , Mitogen-Activated Protein Kinase 1/metabolism , Mitogen-Activated Protein Kinase 3/metabolism , Mutation , Protein Transport , Receptors, Glucagon/analysis , Receptors, Glucagon/genetics
17.
J Bone Miner Res ; 20(10): 1792-803, 2005 Oct.
Article in English | MEDLINE | ID: mdl-16160737

ABSTRACT

UNLABELLED: Osteoblast activity and plasma 1,25(OH)2 vitamin D are increased in HPT but suppressed in HHM. To model HPT and HHM, we directly compared multiday continuous infusions of PTH versus PTHrP in humans. Continuous infusion of both PTH and PTHrP results in marked and prolonged suppression of bone formation; renal 1,25(OH)2D synthesis was stimulated effectively by PTH but poorly by PTHrP. INTRODUCTION: PTH and PTH-related protein (PTHrP) cause primary hyperparathyroidism (HPT) and humoral hypercalcemia of malignancy (HHM), respectively. Whereas HHM and HPT resemble one another in many respects, osteoblastic bone formation and plasma 1,25(OH)2 vitamin D are increased in HPT but reduced in HHM. MATERIALS AND METHODS: We performed 2- to 4-day continuous infusions of escalating doses of PTH and PTHrP in 61 healthy young adults, comparing the effects on serum calcium and phosphorus, renal calcium and phosphorus handling, 1,25(OH)2 vitamin D, endogenous PTH(1-84) concentrations, and plasma IGF-1 and markers of bone turnover. RESULTS: PTH and PTHrP induced comparable effects on renal calcium and phosphorus handling, and both stimulated IGF-1 and bone resorption similarly. Surprisingly, PTH was consistently more calcemic, reflecting a selectively greater increase in renal 1,25(OH)2 vitamin D production by PTH. Equally surprisingly, continuous infusion of both peptides markedly, continuously, and equivalently suppressed bone formation. CONCLUSIONS: PTHrP and PTH produce markedly different effects on 1,25(OH)2 vitamin D homeostasis in humans, leading to different calcemic responses. Moreover, both peptides produce profound suppression of bone formation over multiple days, contrasting with events in HPT, but mimicking HHM. These findings underscore the facts that the mechanisms underlying the anabolic skeletal response to PTH and PTHrP in humans is poorly understood, as are the signal transduction mechanisms that link the renal PTH receptor to 1,25(OH)2 vitamin D synthesis. These studies emphasize that much remains to be learned regarding the normal regulation of vitamin D metabolism and bone formation in response to PTH and PTHrP in humans.


Subject(s)
Bone Density Conservation Agents/administration & dosage , Calcitriol/blood , Hypercalcemia/blood , Osteogenesis/drug effects , Parathyroid Hormone-Related Protein/administration & dosage , Teriparatide/administration & dosage , Adult , Calcium/blood , Female , Humans , Hypercalcemia/chemically induced , Male , Osteoblasts/metabolism
18.
Mol Endocrinol ; 18(6): 1486-98, 2004 Jun.
Article in English | MEDLINE | ID: mdl-14988434

ABSTRACT

The human PTH receptor type 2 (PTH2R) is activated by PTH and tuberoinfundibular peptide of 39 residues (TIP39), resulting in cAMP and intracellular Ca signaling. We now report that, despite these similarities, PTH and TIP39 elicit distinct responses from PTH2R. First, TIP39 induced beta-arrestin and protein kinase Cbeta mobilization and receptor internalization, whereas PTH did not. However, PTH stimulated trafficking of these molecules for a chimeric PTH2R containing the N terminus and third extracellular loop of PTH receptor type 1 (PTH1R). Second, whereas PTH-stimulated cAMP activity was brief and rapidly resensitized, the response to TIP39 was sustained and partly desensitized for a prolonged period. PTH2R desensitization was mediated by beta-arrestin interaction with the C terminus (amino acids 426-457) of PTH2R, whereas beta-arrestin mobilization had a minor influence on PTH2R internalization in response to TIP39, as shown with C terminus deletion mutants and/or dominant negative forms of beta-arrestin and dynamin. These data contrast with PTH1R, at which these dominant negative mutants markedly inhibited receptor internalization. Collectively, these results further highlight how specific interactions within the ligand-receptor bimolecular complex mediate distinct postactivation responses of class II G protein- coupled receptors and provide novel insights into the physiological regulation of PTH2R activity.


Subject(s)
Parathyroid Hormone/metabolism , Receptor, Parathyroid Hormone, Type 2/metabolism , Amino Acid Sequence , Animals , Arrestins/metabolism , Calcium/metabolism , Cell Line , Cloning, Molecular , Cyclic AMP/metabolism , Dynamins/metabolism , Endocytosis , Genes, Dominant , Green Fluorescent Proteins/metabolism , Humans , Kinetics , Ligands , Molecular Sequence Data , Mutation , Peptides/chemistry , Plasmids/metabolism , Protein Binding , Protein Kinase C/metabolism , Protein Kinase C beta , Protein Structure, Tertiary , Rats , Receptor, Parathyroid Hormone, Type 2/chemistry , Recombinant Fusion Proteins/chemistry , Sequence Homology, Amino Acid , Signal Transduction , Temperature , Time Factors , beta-Arrestins
19.
PLoS One ; 10(6): e0129554, 2015.
Article in English | MEDLINE | ID: mdl-26070212

ABSTRACT

Na+/H+ Exchanger Regulatory Factor-1 (NHERF1) is a scaffolding protein containing 2 PDZ domains that coordinates the assembly and trafficking of transmembrane receptors and ion channels. Most target proteins harboring a C-terminus recognition motif bind more-or-less equivalently to the either PDZ domain, which contain identical core-binding motifs. However some substrates such as the type II sodium-dependent phosphate co-transporter (NPT2A), uniquely bind only one PDZ domain. We sought to define the structural determinants responsible for the specificity of interaction between NHERF1 PDZ domains and NPT2A. By performing all-atom/explicit-solvent molecular dynamics (MD) simulations in combination with biological mutagenesis, fluorescent polarization (FP) binding assays, and isothermal titration calorimetry (ITC), we found that in addition to canonical interactions of residues at 0 and -2 positions, Arg at the -1 position of NPT2A plays a critical role in association with Glu43 and His27 of PDZ1 that are absent in PDZ2. Experimentally introduced mutation in PDZ1 (Glu43Asp and His27Asn) decreased binding to NPT2A. Conversely, introduction of Asp183Glu and Asn167His mutations in PDZ2 promoted the formation of favorable interactions yielding micromolar KDs. The results describe novel determinants within both the PDZ domain and outside the canonical PDZ-recognition motif that are responsible for discrimination of NPT2A between two PDZ domains. The results challenge general paradigms for PDZ recognition and suggest new targets for drug development.


Subject(s)
Binding Sites , PDZ Domains , Phosphoproteins/chemistry , Phosphoproteins/metabolism , Sodium-Hydrogen Exchangers/chemistry , Sodium-Hydrogen Exchangers/metabolism , Sodium-Phosphate Cotransporter Proteins, Type IIa/chemistry , Sodium-Phosphate Cotransporter Proteins, Type IIa/metabolism , Humans , Kinetics , Models, Molecular , Mutation , PDZ Domains/genetics , Peptides/chemistry , Peptides/metabolism , Protein Binding , Protein Conformation , Structure-Activity Relationship
20.
Endocrinology ; 145(6): 2815-23, 2004 Jun.
Article in English | MEDLINE | ID: mdl-15016722

ABSTRACT

G protein-coupled receptors (GPCRs) mediate the action of many hormones, cytokines, and sensory and chemical signals. It is generally thought that receptor desensitization and internalization require occupancy and activation of the GPCR. PTH and PTHrP receptor (PTH1R) belongs to GPCR class B and is the major regulator of extracellular calcium homeostasis. Using kidney distal convoluted tubule cells transfected with a human PTH1R/enhanced green fluorescent protein fusion protein, quantitative, real-time fluorescence microscopy was used to analyze receptor internalization. In these cells, which are the target of the calcium-sparing action of PTH, PTH(1-34) activated adenylyl cyclase (AC) and phospholipase C (PLC) and PTH1R endocytosis. PTH(1-31), however, stimulated AC and PLC but not PTH1R endocytosis. Conversely, PTH(7-34) rapidly stimulated PTH1R internalization without activating AC or PLC. PTH(2-34) and (3-34) caused PTH1R internalization intermediate between PTH(1-34) and (7-34). PTH1R sequestration occurred in a dynamin- and clathrin-dependent manner. Directly activating AC inhibited PTH1R internalization in response to PTH(7-34). PTH1R endocytosis was sensitive to protein kinase C inhibition. PTH(1-34), (7-34), and (1-31) evoked PTH1R phosphorylation. Removal of most of the C terminus of the PTH1R eliminated receptor phosphorylation and the cAMP/protein kinase C sensitivity of internalization. PTH(1-34) and (7-34) internalized the truncated PTH1R with identical kinetics, and the response was unaffected by forskolin. Thus, the PTH1R C terminus contains regulatory sequences that are involved in, but not required for, PTH1R internalization. The results demonstrate that receptor activation and internalization can be selectively dissociated.


Subject(s)
Endocytosis , Receptor, Parathyroid Hormone, Type 1/physiology , Animals , Caveolae/physiology , Cells, Cultured , Clathrin/physiology , Endocytosis/drug effects , Endocytosis/physiology , Humans , Intracellular Membranes/metabolism , Kidney Tubules, Distal/cytology , Kidney Tubules, Distal/metabolism , Ligands , Mice , Parathyroid Hormone/chemistry , Peptide Fragments/pharmacology , Phosphorylation , Phosphotransferases/physiology , Receptor, Parathyroid Hormone, Type 1/metabolism , Receptors, Parathyroid Hormone/metabolism , Receptors, Parathyroid Hormone/physiology , Second Messenger Systems/physiology , Signal Transduction
SELECTION OF CITATIONS
SEARCH DETAIL