Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 346
Filtrar
Más filtros

Bases de datos
País/Región como asunto
Tipo del documento
Intervalo de año de publicación
1.
PLoS Genet ; 16(5): e1008772, 2020 05.
Artículo en Inglés | MEDLINE | ID: mdl-32365064

RESUMEN

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.


Asunto(s)
Neuropéptidos/metabolismo , Hormona Paratiroidea/metabolismo , Receptores de Hormona Paratiroidea/genética , Tribolium/anatomía & histología , Animales , Evolución Molecular , Proteínas de Insectos/genética , Proteínas de Insectos/metabolismo , Fenotipo , Filogenia , Receptores de Hormona Paratiroidea/metabolismo , Análisis de Secuencia de ARN , Tribolium/genética , Tribolium/metabolismo , Alas de Animales/anatomía & histología
2.
Cytokine ; 148: 155685, 2021 12.
Artículo en Inglés | MEDLINE | ID: mdl-34411988

RESUMEN

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.


Asunto(s)
Factor de Transcripción Activador 4/metabolismo , Desarrollo Óseo , Huesos/patología , Osteoprotegerina/metabolismo , Hormona Paratiroidea/sangre , Ligando RANK/metabolismo , Receptores de Hormona Paratiroidea/metabolismo , Insuficiencia Renal Crónica/sangre , Factor de Transcripción Activador 4/genética , Animales , Fenómenos Biomecánicos , Huesos/metabolismo , Fémur/patología , Fémur/fisiopatología , Regulación de la Expresión Génica , Hormona Paratiroidea/genética , Ratas , Receptores de Hormona Paratiroidea/genética , Insuficiencia Renal Crónica/genética , Insuficiencia Renal Crónica/patología , Insuficiencia Renal Crónica/fisiopatología , Solubilidad
3.
Development ; 139(22): 4239-49, 2012 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-23034629

RESUMEN

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.


Asunto(s)
Glándulas Mamarias Animales/embriología , Mesodermo/embriología , Proteína Relacionada con la Hormona Paratiroidea/metabolismo , Vía de Señalización Wnt , Animales , Diferenciación Celular , Femenino , Regulación del Desarrollo de la Expresión Génica , Queratinocitos/metabolismo , Factor de Unión 1 al Potenciador Linfoide/biosíntesis , Glándulas Mamarias Animales/metabolismo , Mesodermo/citología , Mesodermo/metabolismo , Ratones , Ratones Noqueados , Proteína Relacionada con la Hormona Paratiroidea/deficiencia , Proteína Relacionada con la Hormona Paratiroidea/genética , Receptores de Hormona Paratiroidea/biosíntesis , Receptores de Hormona Paratiroidea/deficiencia , Receptores de Hormona Paratiroidea/genética , Trombospondinas/metabolismo , Proteínas Wnt/biosíntesis , Proteínas Wnt/metabolismo , beta Catenina/biosíntesis , beta Catenina/metabolismo
4.
Br J Nutr ; 113(6): 909-22, 2015 Mar 28.
Artículo en Inglés | MEDLINE | ID: mdl-25744000

RESUMEN

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.


Asunto(s)
Conservadores de la Densidad Ósea/uso terapéutico , Resorción Ósea/prevención & control , Calcio de la Dieta/uso terapéutico , Interacciones Alimento-Droga , Osteoblastos/efectos de los fármacos , Osteogénesis/efectos de los fármacos , Hormona Paratiroidea/análogos & derivados , Hormona Paratiroidea/uso terapéutico , Animales , Densidad Ósea/efectos de los fármacos , Conservadores de la Densidad Ósea/farmacología , Resorción Ósea/metabolismo , Resorción Ósea/patología , Huesos/efectos de los fármacos , Huesos/metabolismo , Huesos/patología , Canales de Calcio/genética , Canales de Calcio/metabolismo , Calcio de la Dieta/metabolismo , Proliferación Celular/efectos de los fármacos , Células Cultivadas , Terapia Combinada , Masculino , Ratones Endogámicos C57BL , Osteoblastos/metabolismo , Osteoblastos/patología , Hormona Paratiroidea/farmacología , Fragmentos de Péptidos/farmacología , Fragmentos de Péptidos/uso terapéutico , Receptores Sensibles al Calcio/agonistas , Receptores Sensibles al Calcio/genética , Receptores Sensibles al Calcio/metabolismo , Receptores de Hormona Paratiroidea/agonistas , Receptores de Hormona Paratiroidea/genética , Receptores de Hormona Paratiroidea/metabolismo , Canales Catiónicos TRPV/agonistas , Canales Catiónicos TRPV/genética , Canales Catiónicos TRPV/metabolismo , Regulación hacia Arriba/efectos de los fármacos
5.
Int J Mol Sci ; 16(2): 2663-77, 2015 01 23.
Artículo en Inglés | MEDLINE | ID: mdl-25625518

RESUMEN

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.


Asunto(s)
Remodelación Ósea/genética , Fémur/metabolismo , Tibia/metabolismo , Envejecimiento , Animales , Pollos , Colágeno Tipo II/genética , Colágeno Tipo II/metabolismo , Femenino , Osteoprotegerina/genética , Osteoprotegerina/metabolismo , Proteína Relacionada con la Hormona Paratiroidea/genética , Proteína Relacionada con la Hormona Paratiroidea/metabolismo , Ligando RANK/genética , Ligando RANK/metabolismo , Receptores de Hormona Paratiroidea/genética , Receptores de Hormona Paratiroidea/metabolismo , Factor de Transcripción SOX9/genética , Factor de Transcripción SOX9/metabolismo , Regulación hacia Arriba
6.
Nat Genet ; 30(3): 306-10, 2002 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-11850620

RESUMEN

Enchondromas are common benign cartilage tumors of bone. They can occur as solitary lesions or as multiple lesions in enchondromatosis (Ollier and Maffucci diseases). Clinical problems caused by enchondromas include skeletal deformity and the potential for malignant change to chondrosarcoma. The extent of skeletal involvement is variable in enchondromatosis and may include dysplasia that is not directly attributable to enchondromas. Enchondromatosis is rare, obvious inheritance of the condition is unusual and no candidate loci have been identified. Enchondromas are usually in close proximity to, or in continuity with, growth-plate cartilage. Consequently, they may result from abnormal regulation of proliferation and terminal differentiation of chondrocytes in the adjoining growth plate. In normal growth plates, differentiation of proliferative chondrocytes to post-mitotic hypertrophic chondrocytes is regulated in part by a tightly coupled signaling relay involving parathyroid hormone related protein (PTHrP) and Indian hedgehog (IHH). PTHrP delays the hypertrophic differentiation of proliferating chondrocytes, whereas IHH promotes chondrocyte proliferation. We identified a mutant PTH/PTHrP type I receptor (PTHR1) in human enchondromatosis that signals abnormally in vitro and causes enchondroma-like lesions in transgenic mice. The mutant receptor constitutively activates Hedgehog signaling, and excessive Hedgehog signaling is sufficient to cause formation of enchondroma-like lesions.


Asunto(s)
Neoplasias Óseas/fisiopatología , Encondromatosis/fisiopatología , Mutación , Receptores de Hormona Paratiroidea/fisiología , Animales , Neoplasias Óseas/genética , Células COS , Encondromatosis/genética , Proteínas Hedgehog , Humanos , Ratones , Ratones Transgénicos , Mutagénesis Sitio-Dirigida , Receptor de Hormona Paratiroídea Tipo 1 , Receptores de Hormona Paratiroidea/genética , Receptores de Hormona Paratiroidea/metabolismo , Sistemas de Mensajero Secundario , Transducción de Señal , Transactivadores/metabolismo
7.
BMC Evol Biol ; 12: 110, 2012 Jul 06.
Artículo en Inglés | MEDLINE | ID: mdl-22768871

RESUMEN

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.


Asunto(s)
Pollos/genética , Filogenia , Receptores de Hormona Paratiroidea/genética , Secuencia de Aminoácidos , Animales , Calcio/metabolismo , Clonación Molecular , AMP Cíclico/metabolismo , Evolución Molecular , Etiquetas de Secuencia Expresada , Ligamiento Genético , Células HEK293 , Humanos , Datos de Secuencia Molecular , Hormona Paratiroidea/metabolismo , Proteína Relacionada con la Hormona Paratiroidea/metabolismo , Alineación de Secuencia
8.
Endocrinology ; 163(7)2022 07 01.
Artículo en Inglés | MEDLINE | ID: mdl-35460406

RESUMEN

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.


Asunto(s)
Proteína Relacionada con la Hormona Paratiroidea , Hormona Paratiroidea , Receptor de Hormona Paratiroídea Tipo 1 , Secuencia de Aminoácidos , Animales , Ligandos , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Hormona Paratiroidea/genética , Hormona Paratiroidea/metabolismo , Proteína Relacionada con la Hormona Paratiroidea/genética , Proteína Relacionada con la Hormona Paratiroidea/metabolismo , Ratas , Receptor de Hormona Paratiroídea Tipo 1/genética , Receptor de Hormona Paratiroídea Tipo 1/metabolismo , Receptores de Hormona Paratiroidea/genética , Receptores de Hormona Paratiroidea/metabolismo , Transducción de Señal
9.
Dev Cell ; 3(2): 183-94, 2002 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-12194850

RESUMEN

One G protein-coupled receptor (GPCR) can activate more than one G protein, but the physiologic importance of such activation has not been demonstrated in vivo. We have generated mice expressing exclusively a mutant form of the PTH/PTHrP receptor (DSEL) that activates adenylyl cyclase normally but not phospholipase C (PLC). DSEL mutant mice exhibit abnormalities in embryonic endochondral bone development, including delayed ossification and increased chondrocyte proliferation. Analysis of the differentiation of embryonic metatarsals in vitro shows that PTH(1-34) and forskolin inhibit, whereas active phorbol ester stimulates, hypertrophic differentiation. Thus, PLC signaling via the PTH/PTHrP receptor normally slows the proliferation and hastens the differentiation of chondrocytes, actions that oppose the dominant effects of PTH/PTHrP receptors and that involve cAMP-dependent signaling pathways.


Asunto(s)
Huesos/anomalías , Diferenciación Celular/genética , Condrocitos/metabolismo , AMP Cíclico/metabolismo , Hipertrofia/genética , Receptores de Hormona Paratiroidea/deficiencia , Transducción de Señal/genética , Teriparatido/análogos & derivados , Fosfolipasas de Tipo C/metabolismo , Animales , Huesos/metabolismo , Huesos/patología , Diferenciación Celular/efectos de los fármacos , División Celular/genética , Condrocitos/patología , Colforsina/farmacología , Femenino , Hipertrofia/metabolismo , Hipertrofia/patología , Masculino , Ratones , Ratones Noqueados , Mutación/genética , Hormona Paratiroidea/farmacología , Fragmentos de Péptidos/farmacología , Fenotipo , Ésteres del Forbol/farmacología , ARN Mensajero/metabolismo , Receptor de Hormona Paratiroídea Tipo 1 , Receptores de Hormona Paratiroidea/genética , Transducción de Señal/efectos de los fármacos , Teriparatido/farmacología , Factores de Tiempo , Fosfolipasas de Tipo C/efectos de los fármacos
10.
Endocr Rev ; 26(1): 78-113, 2005 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-15689574

RESUMEN

PTH is a major systemic regulator of the concentrations of calcium, phosphate, and active vitamin D metabolites in blood and of cellular activity in bone. Intermittently administered PTH and amino-terminal PTH peptide fragments or analogs also augment bone mass and currently are being introduced into clinical practice as therapies for osteoporosis. The amino-terminal region of PTH is known to be both necessary and sufficient for full activity at PTH/PTHrP receptors (PTH1Rs), which mediate the classical biological actions of the hormone. It is well known that multiple carboxyl-terminal fragments of PTH are present in blood, where they comprise the major form(s) of circulating hormone, but these fragments have long been regarded as inert by-products of PTH metabolism because they neither bind to nor activate PTH1Rs. New in vitro and in vivo evidence, together with older observations extending over the past 20 yr, now points strongly to the existence of novel large carboxyl-terminal PTH fragments in blood and to receptors for these fragments that appear to mediate unique biological actions in bone. This review traces the development of this field in the context of the evolution of our understanding of the "classical" receptor for amino-terminal PTH and the now convincing evidence for these receptors for carboxyl-terminal PTH. The review summarizes current knowledge of the structure, secretion, and metabolism of PTH and its circulating fragments, details available information concerning the pharmacology and actions of carboxyl-terminal PTH receptors, and frames their likely biological and clinical significance. It seems likely that physiological parathyroid regulation of calcium and bone metabolism may involve receptors for circulating carboxy-terminal PTH ligands as well as the action of amino-terminal determinants within the PTH molecule on the classical PTH1R.


Asunto(s)
Hormona Paratiroidea/metabolismo , Fragmentos de Péptidos/metabolismo , Fragmentos de Péptidos/farmacología , Receptores de Hormona Paratiroidea/fisiología , Secuencia de Aminoácidos , Animales , Sitios de Unión , Secuencia Conservada , Humanos , Intestinos/efectos de los fármacos , Datos de Secuencia Molecular , Osteoclastos/efectos de los fármacos , Hormona Paratiroidea/química , Hormona Paratiroidea/farmacología , Fragmentos de Péptidos/química , Receptores de Hormona Paratiroidea/genética
11.
J Cell Biol ; 133(2): 457-68, 1996 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-8609176

RESUMEN

The regulatory role of parathyroid hormone (PTH)/PTH-related peptide (PTHrP) signaling has been implicated in embryonic skeletal development. Here, we studied chondrogenic differentiation of the mouse embryonal carcinoma-derived clonal cell line ATDC5 as a model of chondrogenesis in the early stages of endochondral bone development. ATDC5 cells retain the properties of chondroprogenitor cells, and rapidly proliferate in the presence of 5% FBS. Insulin (10 micrograms/ml) induced chondrogenic differentiation of the cells in a postconfluent phase through a cellular condensation process, resulting in the formation of cartilage nodules, as evidenced by expression of type II collagen and aggrecan genes. We found that differentiated cultures of ATDC5 cells abundantly expressed the high affinity receptor for PTH (Mr approximately 80 kD; Kd = 3.9 nM; 3.2 x 10(5) sites/cell). The receptors on differentiated cells were functionally active, as evidenced by a PTH-dependent activation of adenylate cyclase. Specific binding of PTH to cells markedly increased with the formation of cartilage nodules, while undifferentiated cells failed to show specific binding of PTH. Northern blot analysis indicated that expression of the PTH/PTHrP receptor gene became detectable at the early stage of chondrogenesis of ATDC5 cells, preceding induction of aggrecan gene expression. Expression of the PTH/PTHrP receptor gene was undetectable in undifferentiated cells. The level of PTH/PTHrP receptor mRNA was markedly elevated parallel to that of type II collagen mRNA. These lines of evidence suggest that the expression of functional PTH/PTHrP receptor is associated with the onset of chondrogenesis. In addition, activation of the receptor by exogenous PTH or PTHrP significantly interfered with cellular condensation and the subsequent formation of cartilage nodules, suggesting a novel site of PTHrP action.


Asunto(s)
Cartílago/citología , Proteínas de la Matriz Extracelular , Regulación del Desarrollo de la Expresión Génica/fisiología , Células Madre Neoplásicas/citología , Hormona Paratiroidea/metabolismo , Receptores de Hormona Paratiroidea/genética , Adenilil Ciclasas/metabolismo , Agrecanos , Animales , Secuencia de Bases , Cartílago/metabolismo , Diferenciación Celular , División Celular , Células Clonales , Colágeno/genética , Células Madre de Carcinoma Embrionario , Insulina/farmacología , Lectinas Tipo C , Ratones , Datos de Secuencia Molecular , Osteogénesis , Hormona Paratiroidea/genética , Hormona Paratiroidea/farmacología , Proteína Relacionada con la Hormona Paratiroidea , Proteínas/genética , Proteínas/metabolismo , Proteínas/farmacología , Proteoglicanos/genética , ARN Mensajero/análisis , Receptor de Hormona Paratiroídea Tipo 1 , Receptores de Hormona Paratiroidea/fisiología , Transducción de Señal/fisiología
12.
Science ; 268(5207): 98-100, 1995 Apr 07.
Artículo en Inglés | MEDLINE | ID: mdl-7701349

RESUMEN

A single heterozygous nucleotide exchange in exon M2 of the gene encoding the parathyroid hormone-parathyroid hormone-related peptide (PTH-PTHrP) receptor was identified in a patient with Jansen-type metaphyseal chondrodysplasia, which changes a strictly conserved histidine residue at position 223 in the receptor's first intracellular loop to arginine. Constitutive, ligand-independent adenosine 3',5'-monophosphate accumulation was observed in COS-7 cells expressing the mutant PTH-PTHrP receptor but not in cells expressing the wild-type receptor. This finding explains the severe ligand-independent hypercalcemia and hypophosphatemia, and most likely the abnormal formation of endochondral bone, in this rare form of short-limbed dwarfism.


Asunto(s)
Enanismo/genética , Osteocondrodisplasias/genética , Mutación Puntual , Receptores de Hormona Paratiroidea/genética , Secuencia de Aminoácidos , Línea Celular , AMP Cíclico/metabolismo , Análisis Mutacional de ADN , Femenino , Humanos , Fosfatos de Inositol/metabolismo , Masculino , Datos de Secuencia Molecular , Receptor de Hormona Paratiroídea Tipo 1 , Receptores de Hormona Paratiroidea/biosíntesis , Receptores de Hormona Paratiroidea/fisiología , Proteínas Recombinantes/biosíntesis , Transfección
13.
Science ; 273(5275): 663-6, 1996 Aug 02.
Artículo en Inglés | MEDLINE | ID: mdl-8662561

RESUMEN

The PTH/PTHrP receptor binds to two ligands with distinct functions: the calcium-regulating hormone, parathyroid hormone (PTH), and the paracrine factor, PTH-related protein (PTHrP). Each ligand, in turn, is likely to activate more than one receptor. The functions of the PTH/PTHrP receptor were investigated by deletion of the murine gene by homologous recombination. Most PTH/PTHrP receptor (-/-) mutant mice died in mid-gestation, a phenotype not observed in PTHrP (-/-) mice, perhaps because of the effects of maternal PTHrP. Mice that survived exhibited accelerated differentiation of chondrocytes in bone, and their bones, grown in explant culture, were resistant to the effects of PTHrP and Sonic hedgehog. These results suggest that the PTH/PTHrP receptor mediates the effects of Indian Hedgehog and PTHrP on chondrocyte differentiation.


Asunto(s)
Desarrollo Óseo , Cartílago/citología , Placa de Crecimiento/citología , Osteogénesis , Receptores de Hormona Paratiroidea/fisiología , Transactivadores , Animales , Cartílago/metabolismo , Diferenciación Celular , División Celular , Clonación Molecular , Técnicas de Cultivo , Retroalimentación , Eliminación de Gen , Marcación de Gen , Placa de Crecimiento/metabolismo , Proteínas Hedgehog , Ratones , Ratones Endogámicos C57BL , Osteoblastos/citología , Hormona Paratiroidea , Proteína Relacionada con la Hormona Paratiroidea , Biosíntesis de Proteínas , Proteínas/farmacología , Proteínas/fisiología , Receptor de Hormona Paratiroídea Tipo 1 , Receptores de Hormona Paratiroidea/genética , Células Madre
14.
Mol Endocrinol ; 22(1): 156-66, 2008 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-17872377

RESUMEN

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.


Asunto(s)
Proteína Relacionada con la Hormona Paratiroidea/metabolismo , Hormona Paratiroidea/metabolismo , Receptores de Hormona Paratiroidea/metabolismo , Animales , Células COS , Línea Celular , Línea Celular Tumoral , Chlorocebus aethiops , AMP Cíclico/metabolismo , Transferencia Resonante de Energía de Fluorescencia , Humanos , Cinética , Proteínas Luminiscentes/genética , Proteínas Luminiscentes/metabolismo , Hormona Paratiroidea/química , Proteína Relacionada con la Hormona Paratiroidea/química , Proteína Relacionada con la Hormona Paratiroidea/genética , Unión Proteica , Conformación Proteica , Receptores de Hormona Paratiroidea/química , Receptores de Hormona Paratiroidea/genética , Proteínas Recombinantes de Fusión/química , Proteínas Recombinantes de Fusión/genética , Proteínas Recombinantes de Fusión/metabolismo , Transfección
15.
Endocr Rev ; 15(1): 40-60, 1994 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-8156938

RESUMEN

Since the elucidation of the structures of the three human PRHrP isoforms in 1987, information has rapidly accured which indicates that the role of PTHrP in normal physiology will prove to be crucial as well as exceedingly complex. The importance of the role of PTHrP in normal physiology is underscored by its broad tissue expression, by its intense evolutionary conservation, by its extremely early expression after fertilization of the ovum, and by the lethal consequences of PTHrP gene disruption. The complexity of the role of PTHrP in normal physiology increases almost monthly. This complexity is reflected in the broad tissue distribution of the peptide, its complex transcriptional regulation and mRNA instability motifs, and its multiple transcripts and isoforms. It is now clear that additional complexity exists at the level of posttranslational processing. Expression of the PTHrP gene leads to the tissue-specific processing and secretion of an increasingly complex family of derivative peptides, each with its own repertoire of cognate receptors, signal transduction pathways, and physiological consequences. Further elucidation of the posttranslational processing pathways and mechanisms can be anticipated in the coming years, coupled with a corresponding elucidation of multiple PTHrP receptors, their specific signal transduction pathways, and their unique physiological roles. The role of PTHrP in causing HHM is now clearly established. Work in the coming decade will focus on the normal physiological roles played by PTHrP.


Asunto(s)
Procesamiento Proteico-Postraduccional/fisiología , Proteínas/fisiología , Receptores de Hormona Paratiroidea/fisiología , Secuencia de Aminoácidos , Animales , Clonación Molecular , Humanos , Hipercalcemia/fisiopatología , Queratinocitos/fisiología , Datos de Secuencia Molecular , Proteína Relacionada con la Hormona Paratiroidea , Proteínas/genética , Proteínas/aislamiento & purificación , Proteínas/metabolismo , Receptores de Hormona Paratiroidea/genética
16.
J Clin Invest ; 104(4): 399-407, 1999 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-10449432

RESUMEN

Parathyroid hormone (PTH) and parathyroid hormone-related peptide (PTHrP) bind to and activate the same PTH/PTHrP receptor. Deletion of either the PTHrP gene or the PTH/PTHrP receptor gene leads to acceleration of differentiation of growth plate chondrocytes. To explore further the functional relationships of PTHrP and the PTH/PTHrP receptor, bones of knockout mice were analyzed early in development, and the phenotypes of double-knockout mice were characterized. One early phenotype is shared by both knockouts. Normally, the first chondrocytes to become hypertrophic are located in the centers of long bones; this polarity is greatly diminished in both these knockouts. The PTH/PTHrP receptor-deficient (PTH/PTHrP-R(-/-)) mice exhibited 2 unique phenotypes not shared by the PTHrP(-/-) mice. During intramembranous bone formation in the shafts of long bones, only the PTH/PTHrP-R(-/-) bones exhibit a striking increase in osteoblast number and matrix accumulation. Furthermore, the PTH/PTHrP-R(-/-) mice showed a dramatic decrease in trabecular bone formation in the primary spongiosa and a delay in vascular invasion of the early cartilage model. In the double-homozygous knockout mice, the delay in vascular invasion did not occur. Thus, PTHrP must slow vascular invasion by a mechanism independent of the PTH/PTHrP receptor.


Asunto(s)
Desarrollo Óseo/genética , Desarrollo Óseo/fisiología , Huesos/anomalías , Proteínas/genética , Proteínas/fisiología , Receptores de Hormona Paratiroidea/genética , Receptores de Hormona Paratiroidea/fisiología , Animales , Huesos/irrigación sanguínea , Femenino , Masculino , Ratones , Ratones Noqueados , Osteoblastos/patología , Proteína Relacionada con la Hormona Paratiroidea , Fenotipo , Embarazo , Receptor de Hormona Paratiroídea Tipo 1
17.
J Clin Invest ; 103(3): 373-81, 1999 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-9927498

RESUMEN

Transcription of the mouse parathyroid hormone (PTH)/PTH-related peptide (PTHrP) receptor (PTHR) gene is controlled by promoters P1 and P2. We performed transcript-specific in situ hybridization and found that P2 is the predominant promoter controlling PTHR gene expression in bone and cartilage. Treatment with 1alpha, 25-dihydroxyvitamin D3 (D3) in vivo specifically downregulated P2-specific transcripts in osteoblasts, but not in chondrocytes, under conditions where it enhanced bone resorption. Treatment of the osteoblastic cell line MC3T3-E1 with D3 in vitro reduced expression of both P2-specific transcripts and PTHR protein. This effect was not blocked by cycloheximide, indicating that D3 inhibits PTHR expression by downregulating transcription of the P2 promoter. A similar inhibitory effect of D3 was not observed in the chondrocytic cell line CFK2. Gene-transfer experiments showed that P2, but not P1, is active in both MC3T3-E1 and CFK2 cells, and that D3 specifically inhibited P2 promoter activity in MC3T3-E1, but not in CFK2 cells. Inhibition of P2 activity by D3 required promoter sequences lying more that 1.6 kb upstream of the P2 transcription start site. Thus, the P2 promoter controls PTHR gene expression in both osteoblasts and chondrocytes. D3 downregulates PTHR gene transcription in a cell-specific manner by inhibiting P2 promoter activity in osteoblasts, but not in chondrocytes.


Asunto(s)
Huesos/metabolismo , Calcitriol/farmacología , Agonistas de los Canales de Calcio/farmacología , Cartílago/metabolismo , Regulación de la Expresión Génica/efectos de los fármacos , Receptores de Hormona Paratiroidea/genética , Animales , Técnicas de Transferencia de Gen , Hibridación in Situ , Ratones , Hormona Paratiroidea/metabolismo , Regiones Promotoras Genéticas/genética , Receptor de Hormona Paratiroídea Tipo 1 , Receptores de Hormona Paratiroidea/biosíntesis
18.
J Clin Invest ; 107(8): 1007-15, 2001 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-11306604

RESUMEN

We used Hoxa3 knockout mice and other mouse models to study the role of the fetal parathyroids in fetal calcium homeostasis. Hoxa3-null fetuses lack parathyroid glands, and absence of parathyroid hormone (PTH) was confirmed with a rodent PTH immunoradiometric assay. The ionized calcium level of Hoxa3-null fetuses was significantly lower than that of wild-type or heterozygous littermates or of the mother. Both the rate of placental calcium transfer and the plasma PTHrP level were normal in Hoxa3 mutants and their heterozygous siblings. Because we had previously observed an increase in placental calcium transfer in PTH/PTHrP receptor 1-null (Pthr1-null) fetuses, we assayed plasma PTHrP in those mice. Pthr1-null fetuses had plasma PTHrP levels 11-fold higher than those of their littermates. Northern analysis, immunohistochemical, and in situ hybridization studies of Pthr1-null fetuses indicated that liver and placenta had increased expression of PTHRP: In summary, loss of fetal parathyroids in Hoxa3-null fetuses caused marked hypocalcemia but did not alter placental calcium transfer or the circulating PTHrP level. The findings in the Pthr1-null fetuses indicate that several tissues may contribute to the circulating PTHrP level in fetal mice.


Asunto(s)
Calcio/metabolismo , Glándulas Paratiroides/fisiología , Hormona Paratiroidea/metabolismo , Placenta/metabolismo , Animales , Transporte Biológico , Calcitonina/metabolismo , Proteínas de Homeodominio/genética , Ratones , Ratones Noqueados , Glándulas Paratiroides/metabolismo , Proteína Relacionada con la Hormona Paratiroidea , Proteínas/genética , Proteínas/metabolismo , Receptor de Hormona Paratiroídea Tipo 1 , Receptores Sensibles al Calcio , Receptores de Superficie Celular/genética , Receptores de Superficie Celular/metabolismo , Receptores de Hormona Paratiroidea/genética , Receptores de Hormona Paratiroidea/metabolismo , Distribución Tisular
19.
J Clin Invest ; 99(12): 2961-70, 1997 Jun 15.
Artículo en Inglés | MEDLINE | ID: mdl-9185520

RESUMEN

It has been reported that PTH exerts bone-forming effects in vivo when administered intermittently. In the present study, the anabolic effects of PTH(1-34) on osteoblast differentiation were examined in vitro. Osteoblastic cells isolated from newborn rat calvaria were cyclically treated with PTH(1-34) for the first few hours of each 48-h incubation cycle. When osteoblastic cells were intermittently exposed to PTH only for the first hour of each 48-h incubation cycle and cultured for the remainder of the cycle without the hormone, osteoblast differentiation was inhibited by suppressing alkaline phosphatase activity, bone nodule formation, and mRNA expression of alkaline phosphatase, osteocalcin, and PTH/PTHrP receptor. Experiments using inhibitors and stimulators of cAMP/protein kinase A (PKA) and Ca2+/PKC demonstrated that cAMP/PKA was the major signal transduction system in the inhibitory action of PTH. In contrast, the intermittent exposure to PTH for the first 6 h of each 48-h cycle stimulated osteoblast differentiation. Both cAMP/ PKA and Ca2+/PKC systems appeared to be involved cooperatively in this anabolic effect. Continuous exposure to PTH during the 48-h incubation cycle strongly inhibited osteoblast differentiation. Although both cAMP/PKA and Ca2+/PKC were involved in the effect of continuous exposure to PTH, they appeared to act independently. A neutralizing antibody against IGF-I blocked the stimulatory effect on alkaline phosphatase activity and the expression of osteocalcin mRNA induced by the 6-h intermittent exposure. The inhibitory effect induced by the 1-h intermittent exposure was not affected by anti-IGF-I antibody. These results suggest that PTH has diverse effects on osteoblast differentiation depending on the exposure time in vitro mediated through different signal transduction systems. These in vitro findings explain at least in part the in vivo action of PTH that varies with the mode of administration.


Asunto(s)
Diferenciación Celular/efectos de los fármacos , Osteoblastos/citología , Hormona Paratiroidea/farmacología , Fosfatasa Alcalina/antagonistas & inhibidores , Fosfatasa Alcalina/genética , Fosfatasa Alcalina/metabolismo , Animales , Ciclo Celular , Células Cultivadas , Colforsina/farmacología , Inhibidores Enzimáticos/farmacología , Expresión Génica , Factor I del Crecimiento Similar a la Insulina/antagonistas & inhibidores , Factor I del Crecimiento Similar a la Insulina/fisiología , Osteoblastos/efectos de los fármacos , Osteocalcina/genética , ARN Mensajero/metabolismo , Ratas , Ratas Sprague-Dawley , Receptor de Hormona Paratiroídea Tipo 1 , Receptores de Hormona Paratiroidea/genética , Transducción de Señal
20.
J Clin Invest ; 107(3): 277-86, 2001 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-11160151

RESUMEN

Parathyroid hormone (PTH), an important regulator of calcium homeostasis, targets most of its complex actions in bone to cells of the osteoblast lineage. Furthermore, PTH is known to stimulate osteoclastogenesis indirectly through activation of osteoblastic cells. To assess the role of the PTH/PTH-related protein receptor (PPR) in mediating the diverse actions of PTH on bone in vivo, we generated mice that express, in cells of the osteoblastic lineage, one of the constitutively active receptors described in Jansen's metaphyseal chondrodysplasia. In these transgenic mice, osteoblastic function was increased in the trabecular and endosteal compartments, whereas it was decreased in the periosteum. In trabecular bone of the transgenic mice, there was an increase in osteoblast precursors, as well as in mature osteoblasts. Osteoblastic expression of the constitutively active PPR induced a dramatic increase in osteoclast number in both trabecular and compact bone in transgenic animals. The net effect of these actions was a substantial increase in trabecular bone volume and a decrease in cortical bone thickness of the long bones. These findings, for the first time to our knowledge, identify the PPR as a crucial mediator of both bone-forming and bone-resorbing actions of PTH, and they underline the complexity and heterogeneity of the osteoblast population and/or their regulatory microenvironment.


Asunto(s)
Remodelación Ósea , Huesos/metabolismo , Osteoblastos/metabolismo , Hormona Paratiroidea/fisiología , Receptores de Hormona Paratiroidea/genética , Factores de Edad , Animales , Huesos/citología , Huesos/efectos de los fármacos , Hibridación in Situ , Ratones , Ratones Transgénicos , Mutación , Osteoblastos/efectos de los fármacos , Receptor de Hormona Paratiroídea Tipo 1 , Receptores de Hormona Paratiroidea/biosíntesis , Transducción de Señal , Tibia/citología , Tibia/efectos de los fármacos , Tibia/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA