Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 87
Filter
1.
Horm Metab Res ; 48(11): 689-699, 2016 Nov.
Article in English | MEDLINE | ID: mdl-27643449

ABSTRACT

G-protein coupled receptors (GPCRs) and their ligands are critical for normal osteoblast formation and function. GPCRs mediate a wide variety of biological processes and are activated by multiple types of extracellular signals, ranging from photons to small molecules to peptides. GPCRs signal through a select number of canonical pathways: the Gs and Gi pathways increase or decrease intracellular cAMP levels, respectively, by acting on adenylate cyclase, while the Gq pathway increases intracellular calcium by activating phospholipase C. In addition, non-canonical GPCR pathways such as ß-arrestin activation are important for osteoblast function. Since many cells express multiple GPCRs, and each individual GPCR may activate multiple signaling pathways, the resulting combinatorial signal provides a mechanism for regulating complex biological processes and effector functions. However, the wide variety of GPCRs, the possibility of multiple receptors acting with signaling redundancy, and the possibility of an individual GPCR activating multiple signaling pathways, also pose challenges for elucidating the role of a particular GPCR. Here, we briefly review the roles of Gs and Gi GPCR signaling in osteoblast function. We describe the successful application of a strategy for directly manipulating the Gs and Gi pathways using engineered receptors. These powerful tools will allow further elucidation of the roles of GPCR signaling in specific lineages of osteoblastic cells, as well as in non-osteoblast cells, all of which remain critical areas of active research.


Subject(s)
Bone and Bones/cytology , Cell Differentiation , GTP-Binding Protein alpha Subunits, Gi-Go/metabolism , GTP-Binding Protein alpha Subunits, Gs/metabolism , Receptors, G-Protein-Coupled/metabolism , Animals , Humans , Signal Transduction
2.
Calcif Tissue Int ; 89(1): 10-20, 2011 Jul.
Article in English | MEDLINE | ID: mdl-21526395

ABSTRACT

Activation of the G(s) G protein-coupled receptor Rs1 in osteoblasts increases bone mineral density by 5- to 15-fold in mice and recapitulates histologic aspects of fibrous dysplasia of the bone. However, the effects of constitutive G(s) signaling on bone tissue quality are not known. The goal of this study was to determine bone tissue quality in mice resulting from osteoblast-specific constitutive G(s) activation, by the complementary techniques of FTIR spectroscopy and synchrotron radiation micro-computed tomography (SRµCT). Col1(2.3)-tTA/TetO-Rs1 double transgenic (DT) mice, which showed osteoblast-specific constitutive G(s) signaling activity by the Rs1 receptor, were created. Femora and calvariae of DT and wild-type (WT) mice (6 and 15 weeks old) were analyzed by FTIR spectroscopy. WT and DT femora (3 and 9 weeks old) were imaged by SRµCT. Mineral-to-matrix ratio was 25% lower (P = 0.010), carbonate-to-phosphate ratio was 20% higher (P = 0.025), crystallinity was 4% lower (P = 0.004), and cross-link ratio was 11% lower (P = 0.025) in 6-week DT bone. Differences persisted in 15-week animals. Quantitative SRµCT analysis revealed substantial differences in mean values and heterogeneity of tissue mineral density (TMD). TMD values were 1,156 ± 100 and 711 ± 251 mg/cm(3) (mean ± SD) in WT and DT femoral diaphyses, respectively, at 3 weeks. Similar differences were found in 9-week animals. These results demonstrate that continuous G(s) activation in murine osteoblasts leads to deposition of immature bone tissue with reduced mineralization. Our findings suggest that bone tissue quality may be an important contributor to increased fracture risk in fibrous dysplasia patients.


Subject(s)
Bone Density , Bone and Bones/chemistry , GTP-Binding Protein alpha Subunits, Gs/metabolism , Osteoblasts/metabolism , Animals , Bone and Bones/metabolism , Femur/metabolism , Mice , Mice, Transgenic , Receptors, G-Protein-Coupled/metabolism , Signal Transduction , Spectroscopy, Fourier Transform Infrared , Synchrotrons , Tomography, X-Ray Computed
3.
Endocrinology ; 149(3): 1329-37, 2008 Mar.
Article in English | MEDLINE | ID: mdl-18048501

ABSTRACT

G protein-coupled receptors (GPCRs) coupled to activation of Gs, such as the PTH1 receptor (PTH1R), have long been known to regulate skeletal function and homeostasis. However, the role of GPCRs coupled to other G proteins such as Gi is not well established. We used the tet-off system to regulate the expression of an activated Gi-coupled GPCR (Ro1) in osteoblasts in vivo. Skeletal phenotypes were assessed in mice expressing Ro1 from conception, from late stages of embryogenesis, and after weaning. Long bones were assessed histologically and by microcomputed tomography. Expression of Ro1 from conception resulted in neonatal lethality that was associated with reduced bone mineralization. Expression of Ro1 starting at late embryogenesis resulted in a severe trabecular bone deficit at 12 wk of age (>51% reduction in trabecular bone volume fraction in the proximal tibia compared with sex-matched control littermates; n = 11; P < 0.01). Ro1 expression for 8 wk beginning at 4 wk of age resulted in a more than 20% reduction in trabecular bone volume fraction compared with sex-matched control littermates (n = 16; P < 0.01). Bone histomorphometry revealed that Ro1 expression is associated with reduced rates of bone formation and mineral apposition without a significant change in osteoblast or osteoclast surface. Our results indicate that signaling by a Gi-coupled GPCR in osteoblasts leads to osteopenia resulting from a reduction in trabecular bone formation. The severity of the phenotype is related to the timing and duration of Ro1 expression during growth and development. The skeletal phenotype in Ro1 mice bears some similarity to that produced by knockout of Gs-alpha expression in osteoblasts and thus may be due at least in part to Gi-mediated inhibition of adenylyl cyclase.


Subject(s)
Bone Diseases, Metabolic/metabolism , Osteoblasts/metabolism , Receptors, G-Protein-Coupled/metabolism , Animals , Bone Density/physiology , Bone Development/physiology , Bone Diseases, Metabolic/pathology , Bone and Bones/embryology , Bone and Bones/metabolism , Cells, Cultured , Disease Models, Animal , Female , GTP-Binding Protein alpha Subunits, Gi-Go/metabolism , Gene Expression Regulation, Developmental/physiology , Male , Mice , Mice, Transgenic , Osteoblasts/pathology , Receptors, G-Protein-Coupled/genetics , Receptors, Opioid, kappa/genetics , Receptors, Opioid, kappa/metabolism , Signal Transduction/physiology
4.
Mol Endocrinol ; 15(7): 1186-99, 2001 Jul.
Article in English | MEDLINE | ID: mdl-11435617

ABSTRACT

The type 1 parathyroid hormore receptor (PTH1r) belongs to the class II family of G protein-coupled receptors. To delineate the sites in the PTH1r's N-terminal region, and the carboxy-core domain (transmembrane segments + extracellular loops) involved in PTH binding, we have evaluated the functional properties of 27 PTH1-secretin chimeras receptors stably expressed in HEK-293 cells. The wild type and chimeric receptors were analyzed for cell surface expression, binding for PTH and secretin, and functional responsiveness (cAMP induction) toward secretin and PTH. The expression levels of the chimeric receptors were comparable to that of the PTH1r (60-100%). The N-terminal region of PTH1r was divided into three segments that were replaced either singly or in various combinations with the homologous region of the secretin receptor (SECr). Substitution of the carboxy-terminal half (residues 105-186) of the N-terminal region of PTH1r for a SECr homologous segment did not reduced affinity for PTH but abolished signaling in response to PTH. This data indicate that receptor activation is dissociable from high affinity hormone binding in the PTH1r, and that the N-terminal region might play a critical role in the activation process. Further segment replacements in the N-termini focus on residues 105-186 and particularly residues 146-186 of PTH1r as providing critical segments for receptor activation. The data obtained suggest the existence of two distinct PTH binding sites in the PTH1r's N-terminal region: one site in the amino-terminal half (residues 1-62) (site 1) that participates in high-affinity PTH binding; and a second site of lower affinity constituted by amino acid residues scattered throughout the carboxy-terminal half (residues 105-186) (site 2). In the absence of PTH binding to site 1, higher concentrations of hormone are required to promote receptor activation. In addition, elimination of the interaction of PTH with site 2 results in a loss of signal transduction without loss of high-affinity PTH binding. Divers substitutions of the extracellular loops of the PTH1r highlight the differential role of the first- and third extracellular loop in the process of PTH1r activation after hormone binding. A chimera containing the entire extracellular domains of the PTH1r and the transmembrane + cytoplasmic domains of SECr had very low PTH binding affinity and did not signal in response to PTH. Further substitution of helix 5 of PTH1r in this chimera increased affinity for PTH that is close to the PTH affinity for the wild-type PTH1r but surprisingly, did not mediate signaling response. Additional substitutions of PTH1r's helices in various combinations emphasize the fundamental role of helix 3 and helix 6 on the activation process of the PTH1r. Overall, our studies demonstrated that several PTH1r domains contribute differentially to PTH binding affinity and signal transduction mechanism and highlight the role of the N-terminal domain and helix 3 and helix 6 on receptor activation.


Subject(s)
Parathyroid Hormone/metabolism , Receptors, Gastrointestinal Hormone/chemistry , Receptors, Parathyroid Hormone/chemistry , Receptors, Parathyroid Hormone/physiology , Recombinant Fusion Proteins/chemistry , Amino Acid Sequence , Animals , Binding Sites , Cell Line , Embryo, Mammalian , Gene Expression , Humans , Kidney , Molecular Sequence Data , Opossums , Peptide Fragments/chemistry , Peptide Fragments/genetics , Protein Structure, Secondary , Rats , Receptor, Parathyroid Hormone, Type 1 , Receptors, G-Protein-Coupled , Receptors, Gastrointestinal Hormone/genetics , Receptors, Gastrointestinal Hormone/physiology , Receptors, Parathyroid Hormone/genetics , Recombinant Fusion Proteins/genetics , Recombinant Fusion Proteins/physiology , Signal Transduction , Structure-Activity Relationship , Transfection
5.
J Biol Chem ; 276(36): 33435-43, 2001 Sep 07.
Article in English | MEDLINE | ID: mdl-11387315

ABSTRACT

After stimulation with agonist, G protein-coupled receptors (GPCRs) activate G proteins and become phosphorylated by G protein-coupled receptor kinases (GRKs), and most of them translocate cytosolic arrestin proteins to the cytoplasmic membrane. Agonist-activated GPCRs are specifically phosphorylated by GRKs and are targeted for endocytosis by arrestin proteins, suggesting a connection between GPCR conformational changes and interaction with GRKs and arrestins. Previously, we showed that by substitution of histidine for residues at the cytoplasmic side of helix 3 (H3) and helix 6 (H6) of the parathyroid hormone (PTH) receptor (PTHR), a zinc metal ion-binding site is engineered that prevents PTH-stimulated G(s) activation (Sheikh, S. P., Vilardaga, J.-P., Baranski, T. J., Lichtarge, O., Iiri, T., Meng, E. C., Nissenson, R. A., and Bourne, H. R. (1999) J. Biol. Chem. 274, 17033-17041). These data suggest that relative movements between H3 and H6 are critical for G(s) activation. Does this molecular event play a similar role in activation of GRK and arrestin and in PTHR-mediated G(q) activation? To answer this question, we utilized the two previously described mutant forms of PTHR, H401 and H402, which contain a naturally present histidine residue at position 301 in H3 and a second substituted histidine residue at positions 401 and 402 in H6, respectively. Both mutant receptors showed inhibition of PTH-stimulated inositol phosphate and cAMP generation in the presence of increasing concentrations of Zn(II). However, the mutants showed no Zn(II)-dependent impairment of phosphorylation by GRK-2. Likewise, the mutants were indistinguishable from wild-type PTHR in the ability to translocate beta-arrestins/green fluorescent protein to the cell membrane and were also not affected by sensitivity to Zn(II). These results suggest that agonist-mediated phosphorylation and internalization of PTHR require conformational switches of the receptor distinct from the cAMP and inositol phosphate signaling state. Furthermore, PTHR sequestration does not appear to require G protein activation.


Subject(s)
Arrestin/metabolism , GTP-Binding Proteins/metabolism , Protein Serine-Threonine Kinases/metabolism , Receptors, Parathyroid Hormone/metabolism , Animals , Arrestins/metabolism , Binding Sites , Binding, Competitive , COS Cells , Cell Membrane/metabolism , Chlorides/pharmacology , Cyclic AMP/metabolism , Dose-Response Relationship, Drug , Enzyme Activation , Histidine/chemistry , Immunoblotting , Inhibitory Concentration 50 , Inositol Phosphates/metabolism , Ions/metabolism , Kinetics , Ligands , Mutation , Phosphorylation , Protein Binding , Protein Conformation , Receptor, Parathyroid Hormone, Type 1 , Signal Transduction , Time Factors , Zinc/metabolism , Zinc Compounds/pharmacology , beta-Arrestins
6.
J Cell Biochem ; 81(4): 594-603, 2001.
Article in English | MEDLINE | ID: mdl-11329614

ABSTRACT

In osteogenic and other cells the mitogen-activated protein (MAP) kinases have a key role in regulating proliferation and differentiated functions. The osteogenic growth peptide (OGP) is a 14 mer mitogen of osteogenic and fibroblastic cells that regulates bone turnover, fracture healing, and hematopoiesis, including the engraftment of bone marrow transplants. It is present in the serum and extracellular fluid either free or complexed to OGP-binding proteins (OGPBPs). The free immunoreactive OGP consists of the full length peptide and its C-terminal pentapeptide OGP(10-14). In the present study, designed to probe the signaling pathways triggered by OGP, we demonstrate in osteogenic MC3T3 E1 cells that mitogenic doses of OGP(10-14), but not OGP, enhance MAP kinase activity in a time-dependent manner. The OGP(10-14)-induced stimulation of both MAP kinase activity and DNA synthesis were abrogated by pertusis toxin, a G(i) protein inhibitor. These data offer direct evidence for the occurrence in osteogenic cells of a peptide-activated, mitogenic Gi protein-MAP kinase-signaling cascade. Forskolin and dBu(2)-cAMP abrogated the OGP(10-14)-stimulated proliferation, but induced only 50% inhibition of the OGP(10-14)-mediated MAP kinase activation, suggesting additional MAP kinase-dependent, OGP(10-14)-regulated, cellular functions. Finally, it is demonstrated that OGP(10-14) is the active form of OGP, apparently generated proteolytically in the extracellular milieu upon dissociation of OGP-OGPBP complexes.


Subject(s)
Cyclic AMP/metabolism , Growth Substances/metabolism , Intercellular Signaling Peptides and Proteins , Mitogen-Activated Protein Kinases/metabolism , Osteoblasts/metabolism , Peptides/metabolism , Amino Acid Motifs/physiology , Animals , Cell Division/drug effects , Cell Division/physiology , Cell Line/metabolism , Cyclic AMP/pharmacology , Growth Substances/pharmacology , Histones , Osteoblasts/cytology , Peptides/pharmacology , Signal Transduction/physiology
7.
Mol Endocrinol ; 14(2): 241-54, 2000 Feb.
Article in English | MEDLINE | ID: mdl-10674397

ABSTRACT

The present studies were carried out to evaluate the mechanisms by which PTH/PTHrP receptor (PTHR) activation influences cell viability. In 293 cells expressing recombinant PTHRs, PTH treatment markedly reduced the number of viable cells. This effect was associated with a marked apoptotic response including DNA fragmentation and the appearance of apoptotic nuclei. Similar effects were evidenced in response to serum withdrawal or to the addition of tumor necrosis factor (TNFalpha). Addition of caspase inhibitors or overexpression of bcl-2 partially abrogated apoptosis induced by serum withdrawal. Caspase inhibitors also protected cells from PTH-induced apoptosis, but overexpression of bcl-2 did not. The effects of PTH on cell number and apoptosis were neither mimicked by activators of the cAMP pathway (forskolin, isoproterenol) nor blocked by an inhibitor (H-89). However, elevation of Ca(i)2+ by addition of thapsigargin induced rapid apoptosis, and suppression of Ca(i)2+ by overexpression of the calcium- binding protein, calbindin D28k, inhibited PTH-induced apoptosis. The protein kinase C inhibitor GF 109203X partially inhibited PTH-induced apoptosis. Regulator of G protein signaling 4 (RGS4) (an inhibitor of the activity of the alpha-subunit of Gq) suppressed apoptotic signaling by the PTHR, whereas the C-terminal fragment of GRK2 (an inhibitor of the activity of the beta(gamma)-subunits of G proteins) was without effect. Chemical mutagenesis allowed selection of a series of 293 cell lines resistant to the apoptotic actions of PTH; a subset of these were also resistant to TNFalpha. These results suggest that 1) apoptosis produced by PTHR and TNF receptor signaling involve converging pathways; and 2) Gq-mediated phospholipase C/Ca2+ signaling, rather than Gs-mediated cAMP signaling, is required for the apoptotic effects of PTHR activation.


Subject(s)
Apoptosis/physiology , Cyclic AMP-Dependent Protein Kinases/metabolism , Receptors, Parathyroid Hormone/metabolism , Sulfonamides , Adenylyl Cyclases/metabolism , Amino Acid Chloromethyl Ketones/pharmacology , Apoptosis/drug effects , Calbindin 1 , Calbindins , Caspase Inhibitors , Cell Line/drug effects , Culture Media, Serum-Free , Cyclic AMP/metabolism , Cyclic AMP-Dependent Protein Kinases/antagonists & inhibitors , Cysteine Proteinase Inhibitors/pharmacology , DNA Fragmentation , Humans , Indoles/pharmacology , Isoquinolines/pharmacology , Maleimides/pharmacology , Oligopeptides/pharmacology , Parathyroid Hormone/metabolism , Parathyroid Hormone/pharmacology , Proto-Oncogene Proteins c-bcl-2/drug effects , Proto-Oncogene Proteins c-bcl-2/metabolism , Receptor, Parathyroid Hormone, Type 1 , Receptors, Parathyroid Hormone/antagonists & inhibitors , Receptors, Parathyroid Hormone/genetics , Recombinant Proteins/genetics , Recombinant Proteins/metabolism , S100 Calcium Binding Protein G/metabolism , Signal Transduction , Type C Phospholipases/metabolism , beta-Adrenergic Receptor Kinases
9.
J Biol Chem ; 274(24): 17033-41, 1999 Jun 11.
Article in English | MEDLINE | ID: mdl-10358054

ABSTRACT

The seven transmembrane helices of serpentine receptors comprise a conserved switch that relays signals from extracellular stimuli to heterotrimeric G proteins on the cytoplasmic face of the membrane. By substituting histidines for residues at the cytoplasmic ends of helices III and VI in retinal rhodopsin, we engineered a metal-binding site whose occupancy by Zn(II) prevented the receptor from activating a retinal G protein, Gt (Sheikh, S. P., Zvyaga, T. A. , Lichtarge, O., Sakmar, T. P., and Bourne, H. R. (1996) Nature 383, 347-350). Now we report engineering of metal-binding sites bridging the cytoplasmic ends of these two helices in two other serpentine receptors, the beta2-adrenoreceptor and the parathyroid hormone receptor; occupancy of the metal-binding site by Zn(II) markedly impairs the ability of each receptor to mediate ligand-dependent activation of Gs, the stimulatory regulator of adenylyl cyclase. We infer that these two receptors share with rhodopsin a common three-dimensional architecture and an activation switch that requires movement, relative to one another, of helices III and VI; these inferences are surprising in the case of the parathyroid hormone receptor, a receptor that contains seven stretches of hydrophobic sequence but whose amino acid sequence otherwise shows no apparent similarity to those of receptors in the rhodopsin family. These findings highlight the evolutionary conservation of the switch mechanism of serpentine receptors and help to constrain models of how the switch works.


Subject(s)
Evolution, Molecular , Receptors, Adrenergic, beta-2/metabolism , Receptors, Parathyroid Hormone/metabolism , Zinc/pharmacology , Animals , Binding, Competitive , GTP-Binding Protein alpha Subunits, Gs/metabolism , Guanosine 5'-O-(3-Thiotriphosphate)/metabolism , Histidine/genetics , Humans , Isoproterenol/metabolism , Models, Molecular , Mutation , Pindolol/analogs & derivatives , Pindolol/metabolism , Protein Engineering , Protein Structure, Secondary , Receptors, Adrenergic, beta-2/chemistry , Receptors, Adrenergic, beta-2/drug effects , Receptors, Adrenergic, beta-2/genetics , Receptors, Parathyroid Hormone/chemistry , Receptors, Parathyroid Hormone/drug effects , Receptors, Parathyroid Hormone/genetics , Rod Opsins , Secretin , Sequence Alignment , Signal Transduction
10.
Endocrinology ; 140(3): 1294-300, 1999 Mar.
Article in English | MEDLINE | ID: mdl-10067856

ABSTRACT

For G protein-coupled receptors, limited information is available on the role of agonist binding or of the second-messenger products of receptor signaling on receptor endocytosis. We explored this problem using the opossum PTH/PTH-related protein (PTHrP) receptor, a prototypical Class II G protein-coupled receptor, as a model. In one approach, we evaluated the endocytic properties of mutated forms of the opossum PTH/PTHrP receptor that we had previously shown to be impaired in their ability to initiate agonist-induced signaling when expressed in COS-7 cells. A point mutation in the third cytoplasmic loop (K382A) that severely impairs PTH/PTHrP receptor signaling significantly reduced internalization, whereas two mutant receptors that displayed only partial defects in signaling were internalized normally. To explore more directly the role of second-messenger pathways, we used a cleavable biotinylation method to assess endocytosis of the wild-type receptor stably expressed in human embryonic kidney (HEK) 293 cells. A low rate of constitutive internalization was detected (<5% over a 30-min incubation at 37 C); the rate of receptor internalization was enhanced about 10-fold by the receptor agonists PTH(1-34) or PTHrP(1-34), whereas the receptor antagonist PTH(7-34) had no effect. Forskolin treatment produced a minimal increase in constitutive receptor endocytosis, and the protein kinase (PK)-A inhibitor H-89 failed to block agonist-stimulated endocytosis. Similarly, activation of PK-C, by treatment with phorbol 12-myristate 13-acetate, elicited only a minimal increase in constitutive receptor endocytosis; and blockade of the PK-C pathway, by treatment with a bisindolylmaleimide, failed to inhibit agonist-induced receptor endocytosis. Immunofluorescence confocal microscopic studies of PTH/PTHrP receptor internalization confirmed the results using receptor biotinylation. These findings suggest that: 1) agonist binding is required for the efficient endocytosis of the PTH/PTHrP receptor; 2) receptor activation (agonist-induced receptor conformational change) and/or coupling to G proteins plays a critical role in receptor internalization; and 3) activation of PK-A and PK-C is neither necessary nor sufficient for agonist-stimulated receptor internalization.


Subject(s)
GTP-Binding Proteins/metabolism , Parathyroid Hormone/metabolism , Proteins/metabolism , Receptors, Parathyroid Hormone/agonists , Signal Transduction/physiology , Animals , COS Cells , Cell Line , Endocytosis/physiology , Humans , Parathyroid Hormone-Related Protein , Protein Conformation , Receptor, Parathyroid Hormone, Type 1 , Second Messenger Systems/physiology
12.
Mol Endocrinol ; 12(12): 1846-56, 1998 Dec.
Article in English | MEDLINE | ID: mdl-9849959

ABSTRACT

In some G protein-coupled receptors (GPCRs), agonist-dependent phosphorylation by specific GPCR kinases (GRKs) is an important mediator of receptor desensitization and endocytosis. Phosphorylation and the subsequent events that it triggers, such as arrestin binding, have been suggested to be regulatory mechanisms for a wide variety of GPCRs. In the present study, we investigated whether agonist-induced phosphorylation of the PTH receptor, a class II GPCR, also regulates receptor internalization. Upon agonist stimulation, the PTH receptor was exclusively phosphorylated on serine residues. Phosphoamino acid analysis of a number of receptor mutants in which individual serine residues had been replaced by threonine identified serine residues in positions 485, 486, and 489 of the cytoplasmic tail as sites of phosphorylation after agonist treatment. When serine residues at positions 483, 485, 486, 489, 495, and 498 were simultaneously replaced by alanine residues, the PTH receptor was no longer phosphorylated either basally or in response to PTH. The substitution of these serine residues by alanine affected neither the number of receptors expressed on the cell surface nor the ability of the receptor to signal via Gs. Overexpression of GRK2, but not GRK3, enhanced PTH-stimulated receptor phosphorylation, and this phosphorylation was abolished by alanine mutagenesis of residues 483, 485, 486, 489, 495, and 498. Thus, phosphorylation of the PTH receptor by the endogenous kinase in HEK-293 cells occurs on the same residues targeted by overexpressed GRK2. Strikingly, the rate and extent of PTH-stimulated internalization of mutated PTH receptors lacking phosphorylation sites were identical to that observed for the wild-type PTH receptor. Moreover, overexpressed GRK2, while enhancing the phosphorylation of the wild-type PTH receptor, had no affect on the rate or extent of receptor internalization in response to PTH. Thus, the agonist-occupied PTH receptor is phosphorylated by a kinase similar or identical to GRK2 in HEK-293 cells, but this phosphorylation is not requisite for efficient receptor endocytosis.


Subject(s)
GTP-Binding Proteins , Protein Serine-Threonine Kinases , Receptors, Parathyroid Hormone/chemistry , Receptors, Parathyroid Hormone/metabolism , Animals , Arrestin/metabolism , Binding Sites , Cattle , Cell Line , Cyclic AMP-Dependent Protein Kinases/genetics , Cyclic AMP-Dependent Protein Kinases/metabolism , Endocytosis , G-Protein-Coupled Receptor Kinase 3 , Gene Expression , Humans , Immunosorbent Techniques , Mutagenesis, Site-Directed , Opossums , Parathyroid Hormone/pharmacology , Phosphorylation , Phosphoserine/analysis , Phosphoserine/metabolism , Receptor Protein-Tyrosine Kinases/genetics , Receptor Protein-Tyrosine Kinases/metabolism , Receptors, Parathyroid Hormone/genetics , beta-Adrenergic Receptor Kinases
13.
J Biol Chem ; 273(7): 3830-7, 1998 Feb 13.
Article in English | MEDLINE | ID: mdl-9461563

ABSTRACT

The mechanisms of ligand binding and receptor activation for G-protein-coupled receptors in the secretin/parathyroid hormone (PTH) receptor subfamily are not understood. The PTH1 receptor (PTH1R) signals in response to both PTH and parathyroid hormone-related peptide (PTHrP), whereas the PTH2 receptor (PTH2R) responds only to PTH, not to PTHrP. To locate PTHrP discriminatory domains in the PTH2R, we generated PTH1R/PTH2R chimeras in which the extracellular amino-terminal domains were exchanged. Production of cAMP in response to 1 microM PTHrP or PTH was identical in cells expressing the PTH1R with the PTH2R amino terminus and in cells expressing the PTH2R with the PTH1R amino terminus. The ability of the chimeric receptor with the PTH2R amino terminus to respond fully to PTHrP showed that the body of the PTH2R must contain sites that limit the response to PTHrP. Mutations to PTH1R sequence were therefore made in each of the seven transmembrane domains of the PTH2R. Mutations in transmembrane domains 3 and 7 resulted in receptors able to respond to PTHrP. Thus, residues in more than one domain form a barrier or filter, allowing the receptor to discriminate between different ligands.


Subject(s)
Parathyroid Hormone/metabolism , Peptide Fragments/metabolism , Proteins , Receptors, Parathyroid Hormone/chemistry , Recombinant Fusion Proteins/genetics , Amino Acid Sequence , Animals , Cell Line , Cloning, Molecular , Cyclic AMP/metabolism , Humans , Membrane Proteins/chemistry , Membrane Proteins/metabolism , Models, Molecular , Molecular Sequence Data , Mutagenesis, Site-Directed/genetics , Opossums , Parathyroid Hormone-Related Protein , Receptor, Parathyroid Hormone, Type 2 , Receptors, Parathyroid Hormone/metabolism , Transfection/genetics
14.
Mol Endocrinol ; 11(7): 851-8, 1997 Jun.
Article in English | MEDLINE | ID: mdl-9178745

ABSTRACT

Two different activating PTH/PTH-related peptide (PTHrP) receptor mutations, H223R and T410P, were recently identified as the most likely cause of Jansen's metaphyseal chondrodysplasia. To assess the functional importance of either amino acid position in the human PTH/PTHrP receptor, H223 and T410 were individually replaced by all other amino acids. At position 223, only arginine and lysine led to agonist-independent cAMP accumulation; all other amino acid substitutions resulted in receptor mutants that lacked constitutive activity or were uninformative due to poor cell surface expression. In contrast, most amino acid substitutions at position 410 conferred constitutive cAMP accumulation and affected PTH/PTHrP receptor expression not at all or only mildly. Mutations corresponding to the H223R or T410P exchange in the human PTH/PTHrP receptor also led to constitutive activity when introduced into the opossum receptor homolog, but showed little or no change in basal cAMP accumulation when introduced into the rat PTH/PTHrP receptor. The PTH/PTHrP receptor residues mutated in Jansen's disease are conserved in all mammalian members of this family of G protein-coupled receptors. However, when the equivalent of either the H223R or the T410P mutation was introduced into several other related receptors, including the PTH2 receptor and the receptors for calcitonin, secretin, GH-releasing hormone, glucagon-like peptide I, and CRH, the resulting mutants failed to induce constitutive activity. These studies suggest that two residues in the human PTH/PTHrP receptor, 223 and 410, have critical roles in signal transduction, but with different sequence constrains.


Subject(s)
Cyclic AMP/metabolism , Gene Expression Regulation/genetics , Osteochondrodysplasias/genetics , Point Mutation/genetics , Receptors, Parathyroid Hormone/genetics , Amino Acid Sequence , Animals , COS Cells , DNA/genetics , Dose-Response Relationship, Drug , Humans , Immune Sera/immunology , Molecular Sequence Data , Rabbits , Rats , Receptors, Parathyroid Hormone/biosynthesis , Receptors, Parathyroid Hormone/immunology , Recombinant Proteins/biosynthesis , Recombinant Proteins/genetics , Recombinant Proteins/immunology , Signal Transduction/physiology
15.
J Biol Chem ; 271(52): 33382-9, 1996 Dec 27.
Article in English | MEDLINE | ID: mdl-8969199

ABSTRACT

Structural determinants within the parathyroid hormone (PTH)/PTH-related peptide (PTHrP) receptor that mediate G-protein activation of adenylate cyclase and phospholipase C are unknown. We investigated the role of the N-terminal region of the third intracellular loop of the opossum PTH/PTHrP receptor in coupling to two signal transduction pathways. We mutated residues in this region by tandem-alanine scanning and expressed these mutant receptors in COS-7 cells and/or Xenopus oocytes. All mutant receptors retained high affinity PTH binding in COS-7 cells, indistinguishable from wild-type receptors. Receptors with tandem-alanine substitutions in two N-terminal segments (377RVL379 and 381TKLR384) demonstrated impaired adenylate cyclase and phospholipase C activation. Receptor mutants with single-alanine substitutions scanning these two segments showed three different signaling defects in COS-7 cells. 1) Two mutant receptors (V378A and L379A) had reduced inositol phosphate (IP), but normal cAMP responses to PTH. 2) Mutant receptor T381A showed reduced cAMP, but wild-type IP responses to PTH. 3) Mutant receptor K382A demonstrated both markedly reduced cAMP and IP production due to PTH. In oocytes, mutants T381A and K382A showed decreased PTH-stimulated cAMP accumulation and intracellular Ca2+ mobilization. Thus, the N-terminal region of the third intracellular loop of this receptor plays a critical role in coupling to both Gs- and Gq-mediated second-messenger generation.


Subject(s)
Calcium/metabolism , Cyclic AMP/metabolism , Inositol Phosphates/metabolism , Receptors, Parathyroid Hormone/metabolism , Signal Transduction , Animals , COS Cells , Female , Kinetics , Models, Molecular , Oocytes/metabolism , Parathyroid Hormone/metabolism , Receptor, Parathyroid Hormone, Type 1 , Xenopus
16.
Endocrinology ; 137(11): 5109-18, 1996 Nov.
Article in English | MEDLINE | ID: mdl-8895385

ABSTRACT

To test the hypothesis that PTH-related peptide (PTHrP) is a paracrine regulator of endochondral bone development, we localized PTHrP and its cognate receptor during normal skeletal development at both messenger RNA (mRNA) and protein levels and compared the growth plate phenotypes of PTHrP-deficient [(PTHrP(-/-)] mice to those of normal littermates [PTHrP(+/+]. PTHrP mRNA was expressed adjacent to uncavitated joints, in the perichondrium of long bones and to a lower level in proliferating chondrocytes. In contrast, PTHrP protein was most evident at the interface of proliferating and hypertrophic zones, where it colocalized with PTH/PTHrP receptor mRNA and protein. Most strikingly, the proliferating zone was dramatically shorter in PTHrP(-/-) cartilage, although the percentage of cells in S-phase of the cell cycle in the proliferating zone was indistinguishable between PTHrP(+/+) and PTHrP(-/-) mice. Terminal differentiation of chondrocytes, which was characterized by cell hypertrophy, apoptosis (DNA fragmentation and decreased bcl-2 mRNA expression), and matrix mineralization, was more advanced in growth cartilage of PTHrP(-/-), compared with PTHrP(+/+) animals. These data demonstrate that PTHrP acts principally as a paracrine factor, which promotes elongation of endochondral bone by restraining or delaying the pace of chondrocytic development and terminal differentiation of growth-plate chondrocytes.


Subject(s)
Bone Development/drug effects , Cartilage, Articular/physiology , Gene Expression Regulation, Developmental , Proteins/physiology , Transcription, Genetic , Animals , Cartilage, Articular/cytology , Cartilage, Articular/embryology , Cell Differentiation/drug effects , Embryonic and Fetal Development , Fetus , Genes, bcl-2 , Mice , Mice, Mutant Strains , Molecular Sequence Data , Parathyroid Hormone/physiology , Parathyroid Hormone-Related Protein , Phenotype , Protein Biosynthesis , Proteins/genetics , RNA, Antisense , RNA, Messenger/biosynthesis
17.
Mol Endocrinol ; 10(9): 1066-76, 1996 Sep.
Article in English | MEDLINE | ID: mdl-8885241

ABSTRACT

Using a PCR-based strategy, two variants of the PTH/PTH-related peptide (PTH-rp) receptor mRNA were identified in human kidney, SaOS-2 human osteoblast cells, and rat bone that are produced by alternative splicing of exons coding for the N-terminal portion of the receptor. In the S-N3-E2 isoform, the exon coding the signal peptide (S) is spliced to an alternative 3'-acceptor site, producing a product respecting the reading frame, but in which the E1 exon is replaced by 12 amino acids derived from the N3 intron. In the S-E2 isoform, in which the E1 exon is deleted by cassette exclusion, the reading frame is changed, but a truncated receptor may be produced by reinitiation of translation at an overlapping stop/start codon. After transfection of COS and Chinese hamster ovary cells with the originally described S-E1-E2 isoform and the two splice variants, active transcription of PTH/PTH-rp receptor mRNA was detected by RT-PCR in all cases. Cell lines transfected with the S-E1-E2 and S-N3-E2 isoforms displayed a 15- to 25-fold and 2- to 3-fold increase, respectively, in cAMP content after stimulation with 2.4 x 10(-7) M human PTH(1-34), whereas cells transfected with the S-E2 isoform did not respond. PTH elicited an increase in intracellular calcium only in cells transfected with the S-E1-E2 isoform. Studies evaluating the surface expression of receptors using anti-human PTH/PTH-rp receptor antibodies and the ability of transfected cells to bind [125I]PTH-rp indicated that the low or absent responses to PTH stimulation resulted, at least in part, from low surface expression of the S-N3-E2 and S-E2 isoforms. These studies support the conclusion that exon E1 is extremely important in promoting surface expression of the PTH/PTH-rp receptor but indicate that isoforms lacking this exon can retain the ability to recognize PTH. The possible intracellular expression of these splice variants, which account for 15-20% of total PTH/PTH-rp receptor mRNA, needs to be evaluated.


Subject(s)
Alternative Splicing , Bone and Bones/metabolism , Kidney/metabolism , Receptors, Parathyroid Hormone/genetics , Receptors, Parathyroid Hormone/metabolism , Amino Acid Sequence , Animals , Base Sequence , CHO Cells/metabolism , COS Cells/metabolism , Calcium/metabolism , Cell Membrane/metabolism , Cloning, Molecular , Cricetinae , Cyclic AMP/metabolism , DNA Primers , DNA, Complementary/chemistry , Humans , Iodine Radioisotopes , Molecular Sequence Data , Parathyroid Hormone/metabolism , Peptide Fragments/genetics , Peptide Fragments/metabolism , Polymerase Chain Reaction/methods , RNA, Messenger/genetics , Rabbits , Rats , Receptor, Parathyroid Hormone, Type 1 , Receptors, Cell Surface/genetics , Receptors, Cell Surface/metabolism , Receptors, Parathyroid Hormone/immunology , Sequence Analysis, DNA , Transfection
18.
J Bone Miner Res ; 11(5): 578-86, 1996 May.
Article in English | MEDLINE | ID: mdl-9157772

ABSTRACT

Activation of the G protein-coupled receptor for parathyroid hormone (PTH)/PTH-related protein (PTHrP) produces homologous desensitization of receptor signaling. We have shown recently that the opossum PTH/PTHrP receptor stably expressed in human embryonic kidney (HEK) 293 cells is phosphorylated upon agonist binding and upon activation of serine/threonine protein kinases (PKA and PKC), an event which for some G protein-coupled receptors has been linked to desensitization. To locate the sites of phosphorylation, mutated forms of the opossum PTH/PTHrP receptor were stably expressed in HEK 293 cells, and ligand-stimulated receptor phosphorylation was evaluated. The five serine and threonine residues of the third cytoplasmic loop of the receptor were not required for receptor phosphorylation. Basal and ligand-induced phosphorylation were, however, completely abolished upon deletion of all but the 16 juxtamembrane residues of the cytoplasmic C-terminal tail of the receptor, even though this truncated receptor resembled the wild-type receptor in its level of expression based on Western blotting and radioligand binding. To identify further the phosphorylation sites, the 129 amino acid C-terminal tail of the rat PTH/PTHrP receptor was expressed in E. coli as a recombinant glutathione S-transferase fusion protein. Elimination of a single PKA consensus site in the tail (serine 491) resulted in > or = 90% loss of PKA-mediated phosphorylation, identifying this as the preferential site for PKA, with two other sites (serine 473 and/or 475) being minor sites. Phosphorylation by PKC occurred largely in the proximal portion of the tail, whereas beta-adrenergic receptor kinase 1 (beta ARK1) phosphorylated more distally in the tail. The ability of these kinases to phosphorylate the PTH/PTHrP receptor at distinct sites on the cytoplasmic tail may allow differential regulation of receptor signaling and trafficking.


Subject(s)
Proteins/metabolism , Receptors, Parathyroid Hormone/metabolism , Signal Transduction , Amino Acid Sequence , Animals , Cell Line , Humans , Molecular Sequence Data , Parathyroid Hormone-Related Protein , Phosphorylation , Rats , Receptor, Parathyroid Hormone, Type 1 , Receptors, Parathyroid Hormone/genetics , Recombinant Fusion Proteins/genetics , Recombinant Fusion Proteins/metabolism
19.
J Bone Miner Res ; 11(5): 614-25, 1996 May.
Article in English | MEDLINE | ID: mdl-9157776

ABSTRACT

Intermittent parathyroid hormone (PTH) therapy increases bone mass. The purpose of this study was to determine if analogs of human PTH(1-34) (hPTH[1-34]), which differ from the native sequence in their receptor-activating properties, could promote bone formation in an ovariectomized (OVX) osteopenic rat model. We synthesized two hPTH(1-34) analogs with single substitutions for serine in the 3-position that in vitro are partial agonists in kidney. In the renal cell line OK, maximal cyclic adenosine monophosphate (cAMP) activation by [His(3)]hPTH(134) was 50%, and maximal cAMP activation by [Leu(3)]hPTH(1-34) was 20% of that produced by hPTH(1-34). Both analogs were full agonists in UMR-106 rat osteosarcoma cells and other bone-derived systems, but both had reduced potency compared with hPTh(1-34). Six-month-old retired breeder Sprague-Dawley rats were ovariectomized, and five animals underwent sham operation. On day 56 post-OVX, five sham-operated and five pre-PTH treatment OVX animals were sacrificed, and the remaining animals were randomized into 10 groups of six animals each. All other animals were injected with one of the hPTH analogs or hPTH(1-34) at 0, 4, 40, or 400 mu g/kg of body weight (BW)/day and were killed on day 84. Histomorphometry of the proximal tibia metaphysis and biochemical markers of bone turnover (osteocalcin and pyridinoline cross-links) were the primary endpoints. The cancellous bone volume was significantly lower at day 56 post-OVX (pretreatment) and at day 84 post-OVX (post-vehicle treatment) than at baseline. None of the compounds significantly increased the cancellous bone volume. Trabecular number declined after OVX and did not change with hPTH treatment. In contrast, the trabecular thickness declined after OVX but was higher after treatment with 40 mu g/kg of BW/day or 400 mu g/kg of BW/day of hPTH(1-34). In OVX rats, the mineralizing surface was higher than baseline at day 56 and fell toward control levels by day 84. All three peptides produced marked dose-related increases in the mineralizing surface and bone formation rates, but the two analogs were less potent than hPTH(1-34). Likewise, all peptides produced significant dose-related increases in the serum osteocalcin level. The osteoclast surface was not affected by OVX but was decreased with medium and high doses of hPTH(1-34). Pyridinoline cross-link excretion was not significantly affected by treatment with hPTH(1-34) but responded with a dose-dependent decrease to treatment with [His3]hPTH(1-34). These data suggest that bone selective analogs of hPTH(1-34) maintain the ability to induce bone formation but are less potent than hPTH(1-34).


Subject(s)
Bone Remodeling/drug effects , Bone and Bones/physiopathology , Teriparatide/analogs & derivatives , Animals , Bone Density/drug effects , Bone and Bones/pathology , Cells, Cultured , Female , Humans , Ovariectomy , Rats , Rats, Sprague-Dawley , Teriparatide/pharmacology
20.
J Biol Chem ; 271(16): 9205-8, 1996 Apr 19.
Article in English | MEDLINE | ID: mdl-8621578

ABSTRACT

The seven transmembrane segments (TMs) of many G-protein-coupled receptors (GPCRs) are thought to form a cavity into which cognate ligands insert, leading to receptor activation. Residues lining the cavity are often essential for optimal ligand binding and/or signal transduction. The present studies evaluated whether residues lining the cavity also contribute to specificity, using GPCRs for the polypeptides parathyroid hormone (PTH) and secretin as models. These ligands display no sequence homology with one another, and neither ligand cross-reacts with the other's receptor. However, mutation of a single amino acid in the second TM of the secretin receptor to the corresponding residue in the PTH receptor (N192I) resulted in a receptor that binds and signals in response to PTH. The reciprocal mutation in the PTH receptor (I234N) likewise unmasked responsiveness to secretin. Neither mutation significantly altered the response of the receptors to their own ligands. The results suggest a model of specificity wherein TM residues near the extracellular surface of the receptor function as a selectivity filter that restricts access of inappropriate ligands to an activation site in the transmembrane cavity.


Subject(s)
GTP-Binding Proteins/metabolism , Parathyroid Hormone/pharmacology , Receptors, Gastrointestinal Hormone/chemistry , Receptors, Gastrointestinal Hormone/metabolism , Receptors, Parathyroid Hormone/chemistry , Receptors, Parathyroid Hormone/metabolism , Secretin/pharmacology , Amino Acid Sequence , Animals , Binding Sites , Cell Line , Cell Membrane/metabolism , Chlorocebus aethiops , Cyclic AMP/metabolism , Kinetics , Molecular Sequence Data , Mutagenesis, Site-Directed , Opossums , Parathyroid Hormone/metabolism , Protein Conformation , Receptors, G-Protein-Coupled , Receptors, Gastrointestinal Hormone/biosynthesis , Receptors, Parathyroid Hormone/biosynthesis , Recombinant Fusion Proteins/biosynthesis , Recombinant Fusion Proteins/chemistry , Recombinant Fusion Proteins/metabolism , Secretin/metabolism , Substrate Specificity
SELECTION OF CITATIONS
SEARCH DETAIL
...