Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 347
Filter
1.
Endocrinology ; 163(7)2022 07 01.
Article in English | MEDLINE | ID: mdl-35460406

ABSTRACT

Rodent models are commonly used to evaluate parathyroid hormone (PTH) and PTH-related protein (PTHrP) ligands and analogues for their pharmacologic activities and potential therapeutic utility toward diseases of bone and mineral ion metabolism. Divergence, however, in the amino acid sequences of rodent and human PTH receptors (rat and mouse PTH1Rs are 91% identical to the human PTH1R) can lead to differences in receptor-binding and signaling potencies for such ligands when assessed on rodent vs human PTH1Rs, as shown by cell-based assays in vitro. This introduces an element of uncertainty in the accuracy of rodent models for performing such preclinical evaluations. To overcome this potential uncertainty, we used a homologous recombination-based knockin (KI) approach to generate a mouse (in-host strain C57Bl/6N) in which complementary DNA encoding the human PTH1R replaces a segment (exon 4) of the murine PTH1R gene so that the human and not the mouse PTH1R protein is expressed. Expression is directed by the endogenous mouse promoter and hence occurs in all biologically relevant cells and tissues and at appropriate levels. The resulting homozygous hPTH1R-KI (humanized) mice were healthy over at least 10 generations and showed functional responses to injected PTH analog peptides that are consistent with a fully functional human PTH1R in target bone and kidney cells. The initial evaluation of these mice and their potential utility for predicting behavior of PTH analogues in humans is reported here.


Subject(s)
Parathyroid Hormone-Related Protein , Parathyroid Hormone , Receptor, Parathyroid Hormone, Type 1 , Amino Acid Sequence , Animals , Ligands , Mice , Mice, Inbred C57BL , Mice, Transgenic , Parathyroid Hormone/genetics , Parathyroid Hormone/metabolism , Parathyroid Hormone-Related Protein/genetics , Parathyroid Hormone-Related Protein/metabolism , Rats , Receptor, Parathyroid Hormone, Type 1/genetics , Receptor, Parathyroid Hormone, Type 1/metabolism , Receptors, Parathyroid Hormone/genetics , Receptors, Parathyroid Hormone/metabolism , Signal Transduction
2.
Cytokine ; 148: 155685, 2021 12.
Article in English | MEDLINE | ID: mdl-34411988

ABSTRACT

The progression of chronic kidney disease (CKD) in children is associated with deregulated parathyroid hormone (PTH), growth retardation, and low bone accrual. PTH can cause both catabolic and anabolic impact on bone, and the activating transcription factor 4 (ATF4), a downstream target gene of PTH, is related to its anabolic effect. Osteoprotegerin (OPG) and receptor activator of NF-κB ligand (RANKL) are PTH-dependent cytokines, which may play an important role in the regulation of bone remodeling. This study aimed to evaluate the impact of endogenous PTH and the bone RANKL/OPG system on bone growth, cross-sectional geometry and strength utilizing young, nephrectomized rats. The parameters of cross-sectional geometry were significantly elevated in rats with CKD during the three-month experimental period compared with the controls, and they were strongly associated with serum PTH levels and the expression of parathyroid hormone 1 receptor (PTH1R)/ATF4 genes in bone. Low bone soluble RANKL (sRANKL) levels and sRANKL/OPG ratios were also positively correlated with cross-sectional bone geometry and femoral length. Moreover, the analyzed geometric parameters were strongly related to the biomechanical properties of femoral diaphysis. In summary, the mild increase in endogenous PTH, its anabolic PTH1R/ATF4 axis and PTH-dependent alterations in the bone RANKL/OPG system may be one of the possible mechanisms responsible for the favorable impact on bone growth, cross-sectional geometry and strength in young rats with experimental CKD.


Subject(s)
Activating Transcription Factor 4/metabolism , Bone Development , Bone and Bones/pathology , Osteoprotegerin/metabolism , Parathyroid Hormone/blood , RANK Ligand/metabolism , Receptors, Parathyroid Hormone/metabolism , Renal Insufficiency, Chronic/blood , Activating Transcription Factor 4/genetics , Animals , Biomechanical Phenomena , Bone and Bones/metabolism , Femur/pathology , Femur/physiopathology , Gene Expression Regulation , Parathyroid Hormone/genetics , Rats , Receptors, Parathyroid Hormone/genetics , Renal Insufficiency, Chronic/genetics , Renal Insufficiency, Chronic/pathology , Renal Insufficiency, Chronic/physiopathology , Solubility
3.
PLoS Genet ; 16(5): e1008772, 2020 05.
Article in English | MEDLINE | ID: mdl-32365064

ABSTRACT

In the postgenomics era, comparative genomics have advanced the understanding of evolutionary processes of neuropeptidergic signaling systems. The evolutionary origin of many neuropeptidergic signaling systems can be traced date back to early metazoan evolution based on the conserved sequences. Insect parathyroid hormone receptor (iPTHR) was previously described as an ortholog of vertebrate PTHR that has a well-known function in controlling bone remodeling. However, there was no sequence homologous to PTH sequence in insect genomes, leaving the iPTHR as an orphan receptor. Here, we identified the authentic ligand insect PTH (iPTH) for the iPTHR. The taxonomic distribution of iPTHR, which is lacking in Diptera and Lepidoptera, provided a lead for identifying the authentic ligand. We found that a previously described orphan ligand known as PXXXamide (where X is any amino acid) described in the cuttlefish Sepia officinalis has a similar taxonomic distribution pattern as iPTHR. Tests of this peptide, iPTH, in functional reporter assays confirmed the interaction of the ligand-receptor pair. Study of a model beetle, Tribolium castaneum, was used to investigate the function of the iPTH signaling system by RNA interference followed by RNA sequencing and phenotyping. The results suggested that the iPTH system is likely involved in the regulation of cuticle formation that culminates with a phenotype of defects in wing exoskeleton maturation at the time of adult eclosion. Moreover, RNAi of iPTHRs also led to significant reductions in egg numbers and hatching rates after parental RNAi.


Subject(s)
Neuropeptides/metabolism , Parathyroid Hormone/metabolism , Receptors, Parathyroid Hormone/genetics , Tribolium/anatomy & histology , Animals , Evolution, Molecular , Insect Proteins/genetics , Insect Proteins/metabolism , Phenotype , Phylogeny , Receptors, Parathyroid Hormone/metabolism , Sequence Analysis, RNA , Tribolium/genetics , Tribolium/metabolism , Wings, Animal/anatomy & histology
4.
Ann Endocrinol (Paris) ; 76(2): 98-100, 2015 May.
Article in English | MEDLINE | ID: mdl-25913526

ABSTRACT

Molecular alterations of cAMP-mediated signaling affect primarily the signaling of the PTH/PTHrp receptor, and, with different severities the signaling of other hormones, including TSH. The identification of PTH and other hormonal resistances implies to look for the genetic disorder supporting the metabolic disorder.


Subject(s)
Parathyroid Diseases/therapy , Parathyroid Hormone/physiology , Humans , Parathyroid Diseases/diagnosis , Parathyroid Diseases/physiopathology , Pseudohypoparathyroidism/genetics , Pseudohypoparathyroidism/therapy , Receptors, Parathyroid Hormone/genetics , Receptors, Parathyroid Hormone/metabolism , Thyrotropin/physiology
5.
Br J Nutr ; 113(6): 909-22, 2015 Mar 28.
Article in English | MEDLINE | ID: mdl-25744000

ABSTRACT

In the present study, we investigated whether high dietary Ca and exogenous parathyroid hormone 1-34 fragments (PTH 1-34) have synergistic effects on bone formation in adult mice, and explored the related mechanisms. Adult male mice were fed a normal diet, a high-Ca diet, a PTH-treated diet, or a high-Ca diet combined with subcutaneously injected PTH 1-34 (80 µg/kg per d) for 4 weeks. Bone mineral density, trabecular bone volume, osteoblast number, alkaline phosphatase (ALP)- and type I collagen-positive areas, and the expression levels of osteoblastic bone formation-related genes and proteins were increased significantly in mice fed the high-Ca diet, the PTH-treated diet, and, even more dramatically, the high-Ca diet combined with PTH. Osteoclast number and surface and the ratio of receptor activator for nuclear factor-κB ligand (RANKL):osteoprotegerin (OPG) were decreased in the high-Ca diet treatment group, increased in the PTH treatment group, but not in the combined treatment group. Furthermore, third-passage osteoblasts were treated with high Ca (5 mM), PTH 1-34 (10⁻8 M) or high Ca combined with PTH 1-34. Osteoblast viability and ALP activity were increased in either the high Ca-treated or PTH-treated cultures and, even more dramatically, in the cultures treated with high Ca plus PTH, with consistent up-regulation of the expression levels of osteoblast proliferation and differentiation-related genes and proteins. These results indicate that dietary Ca and PTH play synergistic roles in promoting osteoblastic bone formation by stimulating osteoblast proliferation and differentiation.


Subject(s)
Bone Density Conservation Agents/therapeutic use , Bone Resorption/prevention & control , Calcium, Dietary/therapeutic use , Food-Drug Interactions , Osteoblasts/drug effects , Osteogenesis/drug effects , Parathyroid Hormone/analogs & derivatives , Parathyroid Hormone/therapeutic use , Animals , Bone Density/drug effects , Bone Density Conservation Agents/pharmacology , Bone Resorption/metabolism , Bone Resorption/pathology , Bone and Bones/drug effects , Bone and Bones/metabolism , Bone and Bones/pathology , Calcium Channels/genetics , Calcium Channels/metabolism , Calcium, Dietary/metabolism , Cell Proliferation/drug effects , Cells, Cultured , Combined Modality Therapy , Male , Mice, Inbred C57BL , Osteoblasts/metabolism , Osteoblasts/pathology , Parathyroid Hormone/pharmacology , Peptide Fragments/pharmacology , Peptide Fragments/therapeutic use , Receptors, Calcium-Sensing/agonists , Receptors, Calcium-Sensing/genetics , Receptors, Calcium-Sensing/metabolism , Receptors, Parathyroid Hormone/agonists , Receptors, Parathyroid Hormone/genetics , Receptors, Parathyroid Hormone/metabolism , TRPV Cation Channels/agonists , TRPV Cation Channels/genetics , TRPV Cation Channels/metabolism , Up-Regulation/drug effects
6.
Int J Mol Sci ; 16(2): 2663-77, 2015 01 23.
Article in English | MEDLINE | ID: mdl-25625518

ABSTRACT

Osteoporosis is a bone disease that commonly results in a 30% incidence of fracture in hens used to produce eggs for human consumption. One of the causes of osteoporosis is the lack of mechanical strain placed on weight-bearing bones. In conventionally-caged hens, there is inadequate space for chickens to exercise and induce mechanical strain on their bones. One approach is to encourage mechanical stress on bones by the addition of perches to conventional cages. Our study focuses on the molecular mechanism of bone remodeling in end-of-lay hens (71 weeks) with access to perches. We examined bone-specific transcripts that are actively involved during development and remodeling. Using real-time quantitative PCR, we examined seven transcripts (COL2A1 (collagen, type II, alpha 1), RANKL (receptor activator of nuclear factor kappa-B ligand), OPG (osteoprotegerin), PTHLH (PTH-like hormone), PTH1R (PTH/PTHLH type-1 receptor), PTH3R (PTH/PTHLH type-3 receptor), and SOX9 (Sry-related high mobility group box)) in phalange, tibia and femur. Our results indicate that the only significant effect was a difference among bones for COL2A1 (femur > phalange). Therefore, we conclude that access to a perch did not alter transcript expression. Furthermore, because hens have been used as a model for human bone metabolism and osteoporosis, the results indicate that bone remodeling due to mechanical loading in chickens may be a product of different pathways than those involved in the mammalian model.


Subject(s)
Bone Remodeling/genetics , Femur/metabolism , Tibia/metabolism , Aging , Animals , Chickens , Collagen Type II/genetics , Collagen Type II/metabolism , Female , Osteoprotegerin/genetics , Osteoprotegerin/metabolism , Parathyroid Hormone-Related Protein/genetics , Parathyroid Hormone-Related Protein/metabolism , RANK Ligand/genetics , RANK Ligand/metabolism , Receptors, Parathyroid Hormone/genetics , Receptors, Parathyroid Hormone/metabolism , SOX9 Transcription Factor/genetics , SOX9 Transcription Factor/metabolism , Up-Regulation
7.
Nan Fang Yi Ke Da Xue Xue Bao ; 33(7): 956-61, 2013 Jul.
Article in Chinese | MEDLINE | ID: mdl-23895832

ABSTRACT

OBJECTIVE: To establish HEK293 cell lines with stable expression of human parathyroid hormone (PTH) receptors. METHODS: The purified gene fragments of PTH-related peptide receptor (PTHR) and its mutant form (DSEL) were cloned separately into pcDNA3.1(+) vector after digestion with EcoR I and Not I, and the resulted pcDNA3.1(+)-PTHR and pcDNA3.1(+)-DSEL plasmids were verified by restriction enzyme digestion and DNA sequencing. HEK293 cells were transfected with these plasmids and the expression of PTHR and DSEL in the cells were examined by RT-PCR and ELSIA. RESULTS: Sequencing and restriction enzyme digestion analysis showed that PTHR and DSEL cDNAs were correctly cloned into pcDNA3.1(+)vector. After a 48-h transfection of HEK293 cells with the recombinant plasmids and G418 selection, the positive cell clones stably expressing the constructs were obtained, which showed expressions of PTHR and DSEL mRNAs detected by RT-PCR. These positive cells showed high levels of PLC and aAMP production in response to PTH stimulation. CONCLUSION: The HEK293 cell lines with stable expression of PTH1R or DSEL gene established in this study provide useful cell models for studying the physiological functions of PTH peptides.


Subject(s)
Gene Expression , HEK293 Cells , Receptors, Parathyroid Hormone/genetics , Transfection , Genetic Vectors , Humans , Plasmids , Receptors, Parathyroid Hormone/metabolism , Sequence Analysis, DNA , Signal Transduction/genetics
8.
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
9.
Development ; 139(22): 4239-49, 2012 Nov.
Article in English | MEDLINE | ID: mdl-23034629

ABSTRACT

Parathyroid hormone-related protein (PTHrP) regulates cell fate and specifies the mammary mesenchyme during embryonic development. Loss of PTHrP or its receptor (Pthr1) abolishes the expression of mammary mesenchyme markers and allows mammary bud cells to revert to an epidermal fate. By contrast, overexpression of PTHrP in basal keratinocytes induces inappropriate differentiation of the ventral epidermis into nipple-like skin and is accompanied by ectopic expression of Lef1, ß-catenin and other markers of the mammary mesenchyme. In this study, we document that PTHrP modulates Wnt/ß-catenin signaling in the mammary mesenchyme using a Wnt signaling reporter, TOPGAL-C. Reporter expression is completely abolished by loss of PTHrP signaling and ectopic reporter activity is induced by overexpression of PTHrP. We also demonstrate that loss of Lef1, a key component of the Wnt pathway, attenuates the PTHrP-induced abnormal differentiation of the ventral skin. To characterize further the contribution of canonical Wnt signaling to embryonic mammary development, we deleted ß-catenin specifically in the mammary mesenchyme. Loss of mesenchymal ß-catenin abolished expression of the TOPGAL-C reporter and resulted in mammary buds with reduced expression of mammary mesenchyme markers and impaired sexual dimorphism. It also prevented the ectopic, ventral expression of mammary mesenchyme markers caused by overexpression of PTHrP in basal keratinocytes. Therefore, we conclude that a mesenchymal, canonical Wnt pathway mediates the PTHrP-dependent specification of the mammary mesenchyme.


Subject(s)
Mammary Glands, Animal/embryology , Mesoderm/embryology , Parathyroid Hormone-Related Protein/metabolism , Wnt Signaling Pathway , Animals , Cell Differentiation , Female , Gene Expression Regulation, Developmental , Keratinocytes/metabolism , Lymphoid Enhancer-Binding Factor 1/biosynthesis , Mammary Glands, Animal/metabolism , Mesoderm/cytology , Mesoderm/metabolism , Mice , Mice, Knockout , Parathyroid Hormone-Related Protein/deficiency , Parathyroid Hormone-Related Protein/genetics , Receptors, Parathyroid Hormone/biosynthesis , Receptors, Parathyroid Hormone/deficiency , Receptors, Parathyroid Hormone/genetics , Thrombospondins/metabolism , Wnt Proteins/biosynthesis , Wnt Proteins/metabolism , beta Catenin/biosynthesis , beta Catenin/metabolism
10.
BMC Evol Biol ; 12: 110, 2012 Jul 06.
Article in English | MEDLINE | ID: mdl-22768871

ABSTRACT

BACKGROUND: The parathyroid hormone (PTH)-family consists of a group of structurally related factors that regulate calcium and bone homeostasis and are also involved in development of organs such as the heart, mammary gland and immune system. They interact with specific members of family 2 B1 G-protein coupled receptors (GPCRs), which have been characterised in teleosts and mammals. Two PTH/PTHrP receptors, PTH1R and PTH2R exist in mammals and in teleost fish a further receptor PTH3R has also been identified. Recently in chicken, PTH-family members involved in calcium transport were characterized and specific PTHRs are suggested to exist although they have not yet been isolated or functionally characterized. The aim of this study is to further explore the evolution and function of the vertebrate PTH/PTHrP system through the isolation, phylogenetic analysis and functional characterization of the chicken receptors. RESULTS: Two PTHRs were isolated in chicken and sequence comparison and phylogenetic analysis indicate that the chicken receptors correspond to PTH1R and PTH3R, which emerged prior to the teleost/tetrapod divergence since they are present in cartilaginous fish. The vertebrate PTH2R receptor and its ligand TIP39 have been lost from bird genomes. Chicken PTH1R and PTH3R have a divergent and widespread tissue expression and are also evident in very early embryonic stages of development. Receptor stimulation studies using HEK293 cells stably expressing the chicken PTH1R and PTH3R and monitoring cAMP production revealed they are activated by chicken 1-34 N-terminal PTH-family peptides in a dose dependent manner. PTH-L and PTHrP were the most effective peptides in activating PTH1R (EC(50) = 7.7 nM and EC(50) = 22.7 nM, respectively). In contrast, PTH-L (100 nM) produced a small cAMP accumulation on activation of PTH3R but PTHrP and PTH (EC(50) = 2.5 nM and EC(50) = 22.1 nM, respectively) readily activated the receptor. PTHrP also stimulated intracellular Ca(2+) accumulation on activation of PTH1R but not PTH3R. CONCLUSION: Two PTHR homologues of the vertebrate PTH1R and PTH3R were isolated and functionally characterized in chicken. Their distinct pattern of expression during embryo development and in adult tissues, together with their ligand preference, suggests that they have acquired specific functions, which have contributed to their maintenance in the genome. PTH2R and its activating ligand, TIP39, are absent from bird genomes. Nonetheless identification of putative PTH2R and TIP39 in the genome of an ancient agnathan, lamprey, suggests the PTH/PTHrP ligand and receptor family was already present in an early basal paraphyletic group of vertebrates and during the vertebrate radiation diverged via gene/genome duplication and deletion events. Knowledge of the role PTH/PTHrP system in early vertebrates will help to establish evolution of function.


Subject(s)
Chickens/genetics , Phylogeny , Receptors, Parathyroid Hormone/genetics , Amino Acid Sequence , Animals , Calcium/metabolism , Cloning, Molecular , Cyclic AMP/metabolism , Evolution, Molecular , Expressed Sequence Tags , Genetic Linkage , HEK293 Cells , Humans , Molecular Sequence Data , Parathyroid Hormone/metabolism , Parathyroid Hormone-Related Protein/metabolism , Sequence Alignment
11.
J Clin Endocrinol Metab ; 97(9): 2947-56, 2012 Sep.
Article in English | MEDLINE | ID: mdl-22745236

ABSTRACT

PTHrP was identified as a cause of hypercalcemia in cancer patients 25 yr ago. In the intervening years, we have learned that PTHrP and PTH are encoded by related genes that are part of a larger "PTH gene family." This evolutionary relationship permits them to bind to the same type 1 PTH/PTHrP receptor, which explains why humoral hypercalcemia of malignancy resembles hyperparathyroidism. This review will outline basic facts about PTHrP biology and its normal physiological functions, with an emphasis on new findings of the past 5-10 yr. The medical and research communities first became aware of PTHrP because of its involvement in a common paraneoplastic syndrome. Now, research into the basic biology of PTHrP has suggested previously unrecognized connections to a variety of disease states such as osteoporosis, osteoarthritis, and breast cancer and has highlighted how PTHrP itself might be used in therapy for osteoporosis and diabetes. Therefore, the story of this remarkable protein is a paradigm for translational research, having gone from bedside to bench and now back to bedside.


Subject(s)
Parathyroid Hormone-Related Protein/physiology , Animals , Cell Nucleus/metabolism , Humans , Parathyroid Diseases/genetics , Parathyroid Diseases/physiopathology , Parathyroid Hormone/genetics , Parathyroid Hormone-Related Protein/genetics , Protein Conformation , Receptors, Parathyroid Hormone/genetics
12.
Bone ; 48(5): 1186-93, 2011 May 01.
Article in English | MEDLINE | ID: mdl-21334472

ABSTRACT

The effect of fugu parathyroid hormone 1 (fugu PTH1) on osteoblasts and osteoclasts in teleosts was examined with an assay system using teleost scale and the following markers: alkaline phosphatase (ALP) for osteoblasts and tartrate-resistant acid phosphatase (TRAP) for osteoclasts. Synthetic fugu PTH1 (1-34) (100pg/ml-10ng/ml) significantly increased ALP activity at 6h of incubation. High-dose (10ng/ml) fugu PTH1 significantly increased ALP activity even after 18h of incubation. In the case of TRAP activity, fugu PTH1 did not change at 6h of incubation, but fugu PTH1 (100pg/ml-10ng/ml) significantly increased TRAP activity at 18h. Similar results were obtained for human PTH (1-34), but there was an even greater response with fugu PTH1 than with human PTH. In vitro, we demonstrated that both the receptor activator of the NF-κB ligand in osteoblasts and the receptor activator NF-κB mRNA expression in osteoclasts increased significantly by fugu PTH1 treatment. In an in vivo experiment, fugu PTH1 induced hypercalcemia resulted from the increase of both osteoblastic and osteoclastic activities in the scale as well as the decrease of scale calcium contents after fugu PTH1 injection. In addition, an in vitro experiment with intramuscular autotransplanted scale indicated that the ratio of multinucleated osteoclasts/mononucleated osteoclasts in PTH-treated scales was significantly higher than that in the control scales. Thus, we concluded that PTH acts on osteoblasts and osteoclasts in the scales and regulates calcium metabolism in goldfish.


Subject(s)
Animal Structures/drug effects , Calcium/metabolism , Goldfish/metabolism , Parathyroid Hormone/pharmacology , Acid Phosphatase/metabolism , Alkaline Phosphatase/metabolism , Amino Acid Sequence , Animal Structures/enzymology , Animal Structures/transplantation , Animal Structures/ultrastructure , Animals , Calcium/blood , Cell Differentiation/drug effects , Cells, Cultured , Gene Expression Regulation/drug effects , Giant Cells/cytology , Giant Cells/drug effects , Goldfish/blood , Humans , Isoenzymes/metabolism , Muscles/drug effects , Muscles/transplantation , Osteoclasts/cytology , Osteoclasts/drug effects , Osteoclasts/ultrastructure , RANK Ligand/genetics , RANK Ligand/metabolism , RNA, Messenger/genetics , RNA, Messenger/metabolism , Receptor Activator of Nuclear Factor-kappa B/genetics , Receptor Activator of Nuclear Factor-kappa B/metabolism , Receptors, Parathyroid Hormone/chemistry , Receptors, Parathyroid Hormone/genetics , Receptors, Parathyroid Hormone/metabolism , Takifugu , Tartrate-Resistant Acid Phosphatase , Transplantation, Autologous
14.
Bone ; 43(6): 1022-30, 2008 Dec.
Article in English | MEDLINE | ID: mdl-18761112

ABSTRACT

PTH(1-84) and PTH(7-84) are elevated in chronic kidney disease (CKD). These peptides, as their shorter analogs PTH(1-34) and PTH(7-34) both promote PTH receptor (PTH1R) internalization but only PTH(1-34) and PTH(1-84) activate the receptor. Here, we examined the effects of intermittent administration of PTH(1-34) and PTH(7-34) on mineral ion metabolism, bone architecture, and vascular calcification in rats with experimental CKD. CKD with or without parathyroidectomy (PTX) was established by 5/6 nephrectomy (NPX) in rats. Animals were divided into 4 groups: Sham PTX+ sham NPX (Sham); PTX+ sham NPX (PTX); Sham PTX+NPX (NPX); PTX+NPX (PTX/NPX). Rats were treated with single daily doses of 40 microg/kg PTH(1-34), PTH(7-34), or vehicle. Creatinine was higher in NPX and Ca lower in PTX and PTX/NPX groups than in Sham or NPX rats. Plasma phosphate was higher in PTX, NPX and PTX/NPX than in Sham rats. PTH(1-34) was more hypercalcemic than PTH(7-34) in PTX rats. Fractional bone volume in rats treated with PTH(1-34) increased significantly in all groups compared to that of vehicle treatment. In addition, trabecular number, thickness and volumetric bone density increased in rats treated with PTH(1-34). In contrast, PTH(1-34) diminished vascular calcification. Bone and renal PTH1R mRNA expression was reduced as much or more in PTX/NPX rats as in NPX alone, whereas PTH(7-34) had no effect on PTH1R expression. Renal but not bone PTH1R mRNA increased in response to PTH(1-34). These findings suggest that PTH(1-34) exerts greater hypercalcemic and anabolic effects in parathyroidectomized and/or nephrectomized rats than does PTH(7-34). There was no evidence for significant bone or vascular actions of PTH(7-34). We conclude that PTH(1-34) protects against vascular calcification and bone demineralization in experimental renal failure.


Subject(s)
Aorta/drug effects , Bone and Bones/drug effects , Calcinosis , Parathyroid Hormone/pharmacology , Peptide Fragments/pharmacology , Renal Insufficiency/physiopathology , Animals , Aorta/pathology , Base Sequence , Bone and Bones/metabolism , DNA Primers , Kidney/metabolism , Male , RNA, Messenger/genetics , Rats , Rats, Wistar , Receptors, Parathyroid Hormone/genetics , Receptors, Parathyroid Hormone/metabolism , Renal Insufficiency/metabolism , Reverse Transcriptase Polymerase Chain Reaction , Tomography, X-Ray Computed
15.
Mol Endocrinol ; 22(1): 156-66, 2008 Jan.
Article in English | MEDLINE | ID: mdl-17872377

ABSTRACT

PTH and PTHrP use the same G protein-coupled receptor, the PTH/PTHrP receptor (PTHR), to mediate their distinct biological actions. The extent to which the mechanisms by which the two ligands bind to the PTHR differ is unclear. We examined this question using several pharmacological and biophysical approaches. Kinetic dissociation and equilibrium binding assays revealed that the binding of [(125)I]PTHrP(1-36) to the PTHR was more sensitive to GTPgammaS (added to functionally uncouple PTHR-G protein complexes) than was the binding of [(125)I]PTH(1-34) ( approximately 75% maximal inhibition vs. approximately 20%). Fluorescence resonance energy transfer-based kinetic analyses revealed that PTHrP(1-36) bound to the PTHR more slowly and dissociated from it more rapidly than did PTH(1-34). The cAMP signaling response capacity of PTHrP(1-36) in cells decayed more rapidly than did that of PTH(1-34) (t(1/2) = approximately 1 vs. approximately 2 h). Divergent residue 5 in the ligand, Ile in PTH and His in PTHrP, was identified as a key determinant of the altered receptor-interaction responses exhibited by the two peptides. We conclude that whereas PTH and PTHrP bind similarly to the G protein-coupled PTHR conformation (RG), PTH has a greater capacity to bind to the G protein-uncoupled conformation (R(0)) and, hence, can produce cumulatively greater signaling responses (via R(0)-->RG isomerization) than can PTHrP. Such conformational selectivity may relate to the distinct modes by which PTH and PTHrP act biologically, endocrine vs. paracrine, and may help explain reported differences in the effects that the ligands have on calcium and bone metabolism when administered to humans.


Subject(s)
Parathyroid Hormone-Related Protein/metabolism , Parathyroid Hormone/metabolism , Receptors, Parathyroid Hormone/metabolism , Animals , COS Cells , Cell Line , Cell Line, Tumor , Chlorocebus aethiops , Cyclic AMP/metabolism , Fluorescence Resonance Energy Transfer , Humans , Kinetics , Luminescent Proteins/genetics , Luminescent Proteins/metabolism , Parathyroid Hormone/chemistry , Parathyroid Hormone-Related Protein/chemistry , Parathyroid Hormone-Related Protein/genetics , Protein Binding , Protein Conformation , Receptors, Parathyroid Hormone/chemistry , Receptors, Parathyroid Hormone/genetics , Recombinant Fusion Proteins/chemistry , Recombinant Fusion Proteins/genetics , Recombinant Fusion Proteins/metabolism , Transfection
16.
J Biol Chem ; 282(35): 25509-16, 2007 Aug 31.
Article in English | MEDLINE | ID: mdl-17500070

ABSTRACT

Multiple signaling pathways participate in the regulation of bone remodeling, and pathological negative balance in the regulation results in osteoporosis. However, interactions of signaling pathways that act comprehensively in concert to maintain bone mass are not fully understood. We investigated roles of parathyroid hormone receptor (PTH/PTHrP receptor) signaling in osteoblasts in unloading-induced bone loss using transgenic mice. Hind limb unloading by tail suspension reduced bone mass in wild-type mice. In contrast, signaling by constitutively active PTH/PTHrP receptor (caPPR), whose expression was regulated by the osteoblast-specific Col1a1 promoter (Col1a1-caPPR), suppressed unloading-induced reduction in bone mass in these transgenic mice. In Col1a1-caPPR transgenic (Tg) mice, hind limb unloading suppressed bone formation parameters in vivo and mineralized nodule formation in vitro similarly to those observed in wild-type mice. In addition, serum osteocalcin levels and mRNA expression levels of type I collagen, Runx2 and Osterix in bone were suppressed by unloading in both wild-type mice and Tg mice. However, in contrast to unloading-induced enhancement of bone resorption parameters in wild-type mice, Col1a1-caPPR signaling suppressed, rather than enhanced, osteoclast number and osteoclast surface as well as urinary deoxypyridinoline excretion upon unloading. Col1a1-caPPR signaling also suppressed mRNA expression levels of RANK and c-fms in bone upon unloading. Although the M-CSF and monocyte chemoattractant protein 1 (MCP-1) mRNA levels were enhanced in control Tg mice, these levels were suppressed in unloaded Tg mice. These results indicated that constitutive activation of PTH/PTHrP receptor signaling in osteoblastic cells suppresses unloading-induced bone loss specifically through the regulation of osteoclastic activity.


Subject(s)
Bone Resorption/metabolism , Gene Expression Regulation , Osteoblasts/metabolism , Receptors, Parathyroid Hormone/biosynthesis , Signal Transduction , Animals , Bone Resorption/genetics , Bone Resorption/pathology , Chemokine CCL2/biosynthesis , Chemokine CCL2/genetics , Collagen Type I/biosynthesis , Collagen Type I/genetics , Collagen Type I/metabolism , Collagen Type I, alpha 1 Chain , Core Binding Factor Alpha 1 Subunit/biosynthesis , Core Binding Factor Alpha 1 Subunit/genetics , Female , Gene Expression Regulation/genetics , Hindlimb Suspension , Humans , Macrophage Colony-Stimulating Factor/biosynthesis , Macrophage Colony-Stimulating Factor/genetics , Male , Mice , Mice, Transgenic , Osteoblasts/pathology , Osteocalcin/genetics , Osteocalcin/metabolism , Osteoclasts/metabolism , Osteoclasts/pathology , Osteogenesis/genetics , Osteoporosis/genetics , Osteoporosis/metabolism , Osteoporosis/pathology , Parathyroid Hormone/genetics , Parathyroid Hormone/metabolism , Promoter Regions, Genetic/genetics , RNA, Messenger/biosynthesis , RNA, Messenger/genetics , Receptors, Parathyroid Hormone/genetics , Signal Transduction/genetics , Sp7 Transcription Factor , Transcription Factors/biosynthesis , Transcription Factors/genetics
17.
Eur J Cell Biol ; 85(5): 423-31, 2006 May.
Article in English | MEDLINE | ID: mdl-16487626

ABSTRACT

We recently reported that forced expression of basic helix-loop-helix transcription factor Dec1 accelerated chondrogenic differentiation of mesenchymal stem cells (MSC) in pellet cultures (Shen, M., Yoshida, E., Yan, W., Kawamoto, T., Suardita, K., Koyano, Y., Fujimoto, K., Noshiro, M., Kato, Y., 2002. Basic helix-loop-helix protein DEC1 promotes chondrocyte differentiation at the early and terminal stages. J. Biol. Chem. 277, 50112-50120). Since MSC have multilineage differentiation potential, we investigated the roles of Dec1 in osteogenic and adipogenic differentiation of human bone marrow-derived MSC. After osteogenic induction of MSC in medium containing dexamethasone, beta-glycerophosphate, and ascorbic acid, Dec1 expression gradually increased from day 5 to day 14, while expression levels of Dec1 mRNA markedly decreased on days 3 and 7 after adipogenic induction. Infection with adenovirus expressing Dec1 raised mRNA levels of several bone characteristic molecules such as osteopontin, PTH receptor, and alkaline phosphatase, even in the absence of the osteogenic induction medium, although it had little effect on Runx2 expression or calcification. In the osteogenic induction medium, Dec1 overexpression enhanced the expression of osteopontin and alkaline phosphatase and induced matrix calcification. Knockdown of Dec1 with siRNA suppressed the expression of osteoblastic phenotype by the induced MSC. Using MSC cultures, we also confirmed that forced expression of Dec1 suppressed adipogenic differentiation. These findings suggest that Dec1 modulates osteogenic differentiation of MSC by inducing the expression of several, but not all, bone-related genes.


Subject(s)
Adipogenesis/physiology , Basic Helix-Loop-Helix Transcription Factors/metabolism , Cell Differentiation/physiology , Homeodomain Proteins/metabolism , Mesenchymal Stem Cells/physiology , Osteogenesis/physiology , Animals , Basic Helix-Loop-Helix Transcription Factors/genetics , Bone Marrow Cells/cytology , Bone Marrow Cells/physiology , Cells, Cultured , Core Binding Factor Alpha 1 Subunit/genetics , Core Binding Factor Alpha 1 Subunit/metabolism , Homeodomain Proteins/genetics , Humans , Mesenchymal Stem Cells/cytology , Osteopontin/genetics , Osteopontin/metabolism , RNA Interference , RNA, Small Interfering/genetics , RNA, Small Interfering/metabolism , Receptors, Parathyroid Hormone/genetics , Receptors, Parathyroid Hormone/metabolism
18.
Med Sci (Paris) ; 21(11): 954-61, 2005 Nov.
Article in French | MEDLINE | ID: mdl-16274647

ABSTRACT

Skeletal development is a highly sophisticated process involving, as a first step, migration and condensation of mesenchymal cells into osteoprogenitor cells. These cells further differentiate into chondrocytes and osteoblasts through multiple differentiation stages requiring a set of specific transcriptional factors. Defective endochondral ossification in human is associated with a large number of inherited skeletal dysplasias caused by mutations in genes encoding extracellular matrix components, growth factors and their receptors, signaling molecules and transcription factors. This review summarizes some of the recent findings on a series of chondrodysplasias caused by mutations in FGFR3 and PTHR1, two receptors expressed in the cartilage growth plate and mediating two main signaling pathways. Data from human diseases and relevant animal models provide new clues for understanding how signaling molecules and their interaction with key transcription factors control and regulate the development and growth of long bones.


Subject(s)
Chondrogenesis/physiology , Osteochondrodysplasias/genetics , Osteogenesis/physiology , Receptor, Fibroblast Growth Factor, Type 3/physiology , Receptors, Parathyroid Hormone/physiology , Animals , Cell Differentiation/genetics , Cell Differentiation/physiology , Chondrogenesis/genetics , Fibroblast Growth Factor 3/physiology , Growth Plate/pathology , Humans , Mesoderm/cytology , Models, Animal , Models, Genetic , Mutation , Osteochondrodysplasias/physiopathology , Osteogenesis/genetics , Parathyroid Hormone/physiology , Receptor, Fibroblast Growth Factor, Type 3/deficiency , Receptor, Fibroblast Growth Factor, Type 3/genetics , Receptor, Parathyroid Hormone, Type 1 , Receptors, Parathyroid Hormone/deficiency , Receptors, Parathyroid Hormone/genetics , Signal Transduction/genetics , Signal Transduction/physiology
19.
Endocrinology ; 146(11): 4597-608, 2005 Nov.
Article in English | MEDLINE | ID: mdl-16099862

ABSTRACT

An adequate supply of Ca2+ is critical for normal growth plate development. Previous studies suggest that changes in extracellular [Ca2+] ([Ca2+]e) modulate the function of chondrocytes with high [Ca2+]e promoting cell differentiation. In contrast, signal transduction by the PTH/PTHrP type I receptor (PTH1R) slows down chondrocyte differentiation. This study addressed whether changes in [Ca2+]e modulate the differentiation of mouse growth plate chondrocytes by interacting with PTHrP/PTH1Rs. Raising [Ca2+]e from 0.5-3.0 mM dose-dependently promoted the development of mouse growth plate chondrocytes as indicated by decreases in proteoglycan accumulation and in the expression of early differentiation marker genes and by increases in mineral deposition and in the expression of markers of terminal differentiation. The effects of high [Ca2+]e on gene expression and matrix synthesis were blunted by incubating cells with PTHrP and vice versa. High [Ca2+]e also suppressed the expression of PTH1Rs. Chronic stimulation of PTHrP/PTH1R signaling by adenoviral expression of constitutively active human PTH1Rs (223hPTH1Rs) reduced the effects of high [Ca2+]e on proteoglycan synthesis and gene expression. Similar results were seen when we treated cells with forskolin or 8-bromo-cAMP. Taken together, these data support the idea that the pace of chondrocyte differentiation depends on a balance of interactions between PTHrP/PTH1R and extracellular Ca2+ signaling and that high [Ca2+]e promote cell differentiation potentially by reducing the availability of PTH1Rs and the level of cAMP-dependent signal transduction.


Subject(s)
Calcium/metabolism , Cell Differentiation/physiology , Chondrocytes/cytology , Extracellular Space/metabolism , Growth Plate/cytology , Parathyroid Hormone-Related Protein/metabolism , Signal Transduction/physiology , Animals , Cell Division , Cells, Cultured , Cyclic AMP/metabolism , Gene Transfer Techniques , Humans , Mice , Osmolar Concentration , Parathyroid Hormone-Related Protein/pharmacology , Peptide Fragments/pharmacology , Receptor, Parathyroid Hormone, Type 1 , Receptors, Parathyroid Hormone/genetics , Receptors, Parathyroid Hormone/metabolism
20.
Biochemistry ; 44(15): 5749-54, 2005 Apr 19.
Article in English | MEDLINE | ID: mdl-15823033

ABSTRACT

The calcitonin receptor-like receptor (CLR) requires the associated receptor activity-modifying protein (RAMP)1 to reveal a calcitonin gene-related peptide (CGRP) receptor. Here, the subdomain of the CLR that associates with RAMP1 has been identified in chimeras between the CLR and the parathyroid hormone (PTH) receptor 1 (PTHR). The PTHR alone does not interact with RAMP1. RAMP1 requires the CLR for its transport to the cell surface. Thus, receptor-dependent RAMP1 delivery to the plasma membrane and coimmunoprecipitation from the cell surface were used as measures for receptor/RAMP1 interaction. Several chimeric CLR-PTHR included the N-terminal amino acids 23-60 of the CLR transported RAMP1 to the surface of COS-7 cells much like the intact CLR. Moreover, RAMP1 coimmunoprecipitated with these receptors from the cell surface. A CLR deletion mutant, consisting of the N-terminal extracellular domain, the first transmembrane domain, and the C-terminal intracellular region, revealed the same results. Cyclic AMP was stimulated by CGRP in CLR/RAMP1 expressing cells (58 +/- 19-fold, EC(50) = 0.12 +/- 0.03 nM) and by PTH-related protein in cells expressing the PTHR (50 +/- 10-fold, EC(50) = 0.25 +/- 0.03 nM) or a PTHR with the N-terminal amino acids 23-60 of the CLR (23 +/- 5-fold, EC(50) > 1000 nM). Other chimeric CLR-PTHR were inactive. In conclusion, structural elements in the extreme N-terminus of the CLR between amino acids 23-60 are required and sufficient for CLR/RAMP1 cotransport to the plasma membrane and heterodimerization.


Subject(s)
Membrane Proteins/chemistry , Membrane Proteins/metabolism , Receptors, Calcitonin/chemistry , Receptors, Calcitonin/metabolism , Receptors, Parathyroid Hormone/chemistry , Receptors, Parathyroid Hormone/metabolism , Amino Acid Sequence , Animals , Biological Transport, Active , COS Cells , Calcitonin Receptor-Like Protein , Cell Membrane/metabolism , Chickens , In Vitro Techniques , Intracellular Signaling Peptides and Proteins , Membrane Proteins/genetics , Mice , Molecular Sequence Data , Protein Structure, Tertiary , Rats , Receptor Activity-Modifying Protein 1 , Receptor Activity-Modifying Proteins , Receptors, Calcitonin/genetics , Receptors, Parathyroid Hormone/genetics , Recombinant Fusion Proteins/chemistry , Recombinant Fusion Proteins/genetics , Recombinant Fusion Proteins/metabolism , Sequence Deletion , Sequence Homology, Amino Acid
SELECTION OF CITATIONS
SEARCH DETAIL
...