Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 136
Filter
1.
J Endocrinol Invest ; 40(4): 347-356, 2017 Apr.
Article in English | MEDLINE | ID: mdl-27995443

ABSTRACT

Pseudohypoparathyroidism (PHP) and pseudopseudohypoparathyroidism (PPHP) are caused by mutations and/or epigenetic changes at the complex GNAS locus on chromosome 20q13.3 that undergoes parent-specific methylation changes at several sites. GNAS encodes the alpha-subunit of the stimulatory G protein (Gsα) and several splice variants thereof. Heterozygous inactivating mutations involving the maternal GNAS exons 1-13 cause PHP type Ia (PHP1A). Because of much reduced paternal Gsα expression in certain tissues, such as the proximal renal tubules, thyroid, and pituitary, there is little or no Gsα protein in the presence of maternal GNAS mutations, thus leading to PTH-resistant hypocalcemia and hyperphosphatemia. When located on the paternal allele, the same or similar GNAS mutations are the cause of PPHP. Besides biochemical abnormalities, patients affected by PHP1A show developmental abnormalities, referred to as Albrights hereditary osteodystrophy (AHO). Some, but not all of these AHO features are encountered also in patients affected by PPHP, who typically show no laboratory abnormalities. Autosomal dominant PHP type Ib (AD-PHP1B) is caused by heterozygous maternal deletions within GNAS or STX16, which are associated with loss-of-methylation (LOM) at exon A/B alone or at all maternally methylated GNAS exons. LOM at exon A/B and the resulting biallelic expression of A/B transcripts reduces Gsα expression, thus leading to hormonal resistance. Epigenetic changes at all differentially methylated GNAS regions are also observed in sporadic PHP1B, the most frequent disease variant, which remains unresolved at the molecular level, except for rare cases with paternal uniparental isodisomy or heterodisomy of chromosome 20q (patUPD20q).


Subject(s)
Epigenesis, Genetic/genetics , GTP-Binding Protein alpha Subunits, Gs/genetics , Pseudohypoparathyroidism/genetics , Animals , Humans
2.
Bone ; 50(5): 1100-6, 2012 May.
Article in English | MEDLINE | ID: mdl-22387237

ABSTRACT

Homozygous and compound heterozygous mutations in SLC34A3, the gene encoding the sodium-dependent co-transporter NaPi-IIc, cause hereditary hypophosphatemic rickets with hypercalciuria (HHRH), a disorder characterized by renal phosphate-wasting resulting in hypophosphatemia, elevated 1,25(OH)(2) vitamin D levels, hypercalciuria, rickets/osteomalacia, and frequently kidney stones or nephrocalcinosis. Similar albeit less severe biochemical changes are also observed in heterozygous carriers, which are furthermore indistinguishable from those encountered in idiopathic hypercalciuria (IH). We now searched for SLC34A3 mutations (exons and introns) in two previously not reported HHRH kindreds, which resulted in the identification of three novel mutations. The affected members of kindred A were compound heterozygous for two different mutations, c.1046_47del and the intronic mutation c.560+23_561-42del, while the index case in kindred B was homozygous for the nonsense SLC34A3 mutation c.1764C>G (p.Y588X). The patient in kindred C was diagnosed with IH because of bilateral medullary nephrocalcinosis, suppressed PTH levels, and hypercalciuria; she was found to have a novel heterozygous c.1571_1880del mutation. The HHRH patients in kindred A were treated for up to 7years with oral phosphate, which led to reversal of hypophosphatemia, hypercalciuria, and prevention or healing of the mild bone abnormalities. PTH levels were normal throughout the observation period, while 1,25(OH)(2) vitamin D levels remained elevated and may thus be helpful for assessing treatment efficacy and patient compliance in HHRH.


Subject(s)
Familial Hypophosphatemic Rickets/complications , Familial Hypophosphatemic Rickets/genetics , Hypercalciuria/complications , Hypercalciuria/genetics , Mutation/genetics , Sodium-Phosphate Cotransporter Proteins, Type IIc/genetics , Child , Child, Preschool , DNA Mutational Analysis , Family , Female , Follow-Up Studies , Humans , Male , Pedigree , Time Factors
3.
Exp Clin Endocrinol Diabetes ; 117(2): 49-56, 2009 Feb.
Article in English | MEDLINE | ID: mdl-18523928

ABSTRACT

Hereditary hypophosphatemic rickets with hypercalciuria (HHRH) is caused by mutations in SLC34A3, the gene encoding the renal sodium-phosphate co-transporter NaPi-IIc. Despite increased urinary calcium excretion, HHRH is typically not associated with kidney stones prior to treatment. However, here we describe two sisters, who displayed nephrolithiasis or nephrocalcinosis upon presentation. The index patient, II-4, presented with short stature, bone pain, and knee X-rays suggestive of mild rickets at age 8.5 years. Laboratory evaluation showed hypophosphatemia, elevated 1,25(OH) (2) vitamin D levels, and hypercalciuria, later also developing vitamin D deficiency. Her sister, II-6, had a low normal serum phosphorous level, biochemically vitamin D deficiency and no evidence for osteomalacia, but had undergone left nephro-ureterectomy at age 17 because of ureteral stricture secondary to renal calculi. Nucleotide sequence analysis of DNA from II-4 and II-6 revealed a homozygous missense mutation c.586G>A (p.G196R) in SLC34A3/NaPi-IIc. Ultrasonographic examinations prior to treatment showed grade I nephrocalcinosis for II-4, while II-6 had grade I-II nephrocalcinosis in her remaining kidney. Four siblings and the mother were heterozygous carriers of the mutation, but showed no biochemical abnormalities. With oral phosphate supplements, hypophosphatemia and hypercalciuria improved in both homozygous individuals. Renal calcifications that are presumably due to increased urinary calcium excretion can be the presenting finding in homozygous carriers of G196R in SLC34A3/NaPi-IIc, and some or all laboratory features of HHRH may be masked by vitamin D deficiency.


Subject(s)
Calcinosis/metabolism , Hypercalciuria/metabolism , Hypophosphatemia/metabolism , Kidney Diseases/metabolism , Rickets/metabolism , Sodium-Phosphate Cotransporter Proteins, Type IIc/metabolism , Adolescent , Adult , Calcinosis/complications , Calcinosis/diagnostic imaging , Calcinosis/genetics , Child , Disease Susceptibility , Female , Humans , Hypercalciuria/complications , Hypercalciuria/diagnostic imaging , Hypercalciuria/genetics , Hypophosphatemia/complications , Hypophosphatemia/diagnostic imaging , Hypophosphatemia/genetics , Kidney Diseases/complications , Kidney Diseases/diagnostic imaging , Kidney Diseases/genetics , Male , Middle Aged , Mutation/genetics , Pedigree , Rickets/complications , Rickets/diagnostic imaging , Rickets/genetics , Sodium-Phosphate Cotransporter Proteins, Type IIc/genetics , Ultrasonography
4.
Kidney Int ; 70(8): 1486-94, 2006 Oct.
Article in English | MEDLINE | ID: mdl-16941023

ABSTRACT

Hypophosphatemia is a common complication of kidney transplantation. Tertiary hyperparathyroidism has long been thought to be the etiology, but hypophosphatemia can occur despite low parathyroid hormone (PTH) levels and can persist after high PTH levels normalize. Furthermore, even in the setting of normal allograft function, hypophosphatemia, and hyperparathyroidism, calcitriol levels remain inappropriately low following transplantation, suggesting that mechanisms other than PTH contribute. Fibroblast growth factor-23 (FGF-23) induces phosphaturia, inhibits calcitriol synthesis, and accumulates in chronic kidney disease. We performed a prospective, longitudinal study of 27 living donor transplant recipients to test the hypotheses that excessive FGF-23 accounts for hypophosphatemia and decreased calcitriol levels following kidney transplantation. Hypophosphatemia <2.5 mg/dl developed in 85% of subjects, including one who had previously undergone parathyroidectomy; 37% developed phosphate < or =1.5 mg/dl. The mean pre-transplant FGF-23 level was 1,218+/-542 RU/ml. Within the first week following transplantation, mean levels decreased to 557+/-579 RU/ml, which were still above normal. FGF-23 was independently associated with serum phosphate (P < 0.01), urinary excretion of phosphate (P < 0.01), and calcitriol levels (P < 0.01); PTH was not independently associated with any of these parameters. We calculated area under the curve for FGF-23 and PTH between the pre- and first post-transplant levels as a summary measure of early exposure to these phosphaturic hormones. An area under the FGF-23 curve greater than the median was associated with a relative risk of developing hypophosphatemia < or =1.5 mg/dl of 5.3 (P = 0.02) compared with lower levels. Increased area under the PTH curve was not associated with greater risk of hypophosphatemia. Excessive FGF-23 exposure in the early post-transplant period appears to be more strongly associated with post-transplant hypophosphatemia than PTH.


Subject(s)
Fibroblast Growth Factors/blood , Hypophosphatemia/blood , Hypophosphatemia/etiology , Kidney Transplantation/adverse effects , Phosphates/blood , Adult , Aged , Aged, 80 and over , Calcitriol/blood , Female , Fibroblast Growth Factor-23 , Humans , Hyperparathyroidism/blood , Hyperparathyroidism/complications , Longitudinal Studies , Male , Middle Aged , Parathyroid Hormone/blood , Phosphates/urine , Phosphorus/blood , Prospective Studies , Vitamin D/blood
5.
Am J Physiol Cell Physiol ; 291(1): C114-21, 2006 Jul.
Article in English | MEDLINE | ID: mdl-16687470

ABSTRACT

Parathyroid hormone (PTH), an 84-amino acid polypeptide, is a major systemic regulator of calcium homeostasis that activates PTH/PTHrP receptors (PTH1Rs) on target cells. Carboxyl fragments of PTH (CPTH), secreted by the parathyroids or generated by PTH proteolysis in the liver, circulate in blood at concentrations much higher than intact PTH-(1-84) but cannot activate PTH1Rs. Receptors specific for CPTH fragments (CPTHRs), distinct from PTH1Rs, are expressed by bone cells, especially osteocytes. Activation of CPTHRs was previously reported to modify intracellular calcium within chondrocytes. To further investigate the mechanism of action of CPTHRs in osteocytes, cytosolic free calcium concentration ([Ca(2+)](i)) was measured in the PTH1R-null osteocytic cell line OC59, which expresses abundant CPTHRs but no PTH1Rs. [Ca(2+)](i) was assessed by single-cell ratiometric microfluorimetry in fura-2-loaded OC59 cells. A rapid and transient increase in [Ca(2+)](i) was observed in OC59 cells in response to the CPTH fragment hPTH-(53-84) (250 nM). No [Ca(2+)](i) signal was observed in COS-7 cells, in which CPTHR binding also cannot be detected. Neither hPTH-(1-34) nor a mutant CPTH analog, [Ala(55-57)]hPTH-(53-84), that does not to bind to CPTHRs, increased [Ca(2+)](i) in OC59 cells. The [Ca(2+)](i) response to hPTH-(53-84) required the presence of extracellular calcium and was blocked by inhibitors of voltage-dependent calcium channels (VDCCs), including nifedipine (100 nM), omega-agatoxin IVA (10 nM), and omega-conotoxin GVIA (100 nM). We conclude that activation of CPTHRs in OC59 osteocytic cells leads to a rapid increase in influx of extracellular calcium, most likely through the opening of VDCCs.


Subject(s)
Calcium Channels/physiology , Parathyroid Hormone/physiology , Peptide Fragments/physiology , Receptors, Parathyroid Hormone/physiology , Calcium/physiology , Calcium Channel Blockers/pharmacology , Cell Line, Transformed , Cells, Cultured , Humans , Osteocytes/drug effects , Osteocytes/physiology , Parathyroid Hormone/pharmacology , Receptors, Parathyroid Hormone/agonists , Signal Transduction/drug effects , Signal Transduction/physiology
6.
Endocrinology ; 146(4): 1863-70, 2005 Apr.
Article in English | MEDLINE | ID: mdl-15625242

ABSTRACT

PTH comprises 84 amino acids of which the first 34 are sufficient for full activation of the classical PTH/PTHrP receptor, the type 1 PTH receptor. It is known that multiple carboxyl (C)-terminal fragments of PTH are present in the blood and that they comprise the majority of circulating PTH. C-PTH fragments, previously regarded as by-products of PTH metabolism, are directly secreted by the parathyroid glands or arise from the peripheral cleavage of the intact hormone. Compelling evidence now strongly suggests that these C-PTH fragments mediate biological effects via activation of a receptor that specifically recognizes the C-terminal portion of intact PTH, and this receptor is therefore named the carboxyl-terminal PTH receptor (CPTHR). We have previously reported that osteocytes abundantly express this novel receptor and that its activation is involved in cell survival and communication. Here we report the characterization of determinants of PTH that are required for high-affinity binding to the CPTHR. Using synthetic PTH peptides harboring alanine substitution or truncations, we showed the existence of discrete binding domains and critical residues within the intact hormone. We have furthermore identified eight amino acids within the PTH sequence that play key roles in optimizing the binding affinity of C-PTH fragments to CPTHRs. These include the tripeptide sequence Arg(25)-Lys(26)-Lys(27), the dibasic sequence Lys(53)-Lys(54), and three additional residues within the PTH (55-84) sequence, Asn(57), Lys(65), and Lys(72). Functional analysis of these residues demonstrated a strong correlation between binding affinity and biological effect and points to a potential role of CPTHR activation in regulating bone cell survival.


Subject(s)
Osteocytes/metabolism , Parathyroid Hormone/metabolism , Peptide Fragments/metabolism , Receptor, Parathyroid Hormone, Type 1/metabolism , Amino Acid Sequence , Binding Sites , Cells, Cultured , Humans , Molecular Sequence Data
7.
Endocrinology ; 143(1): 171-6, 2002 Jan.
Article in English | MEDLINE | ID: mdl-11751606

ABSTRACT

The linear sequence of intact mammalian PTH consists of 84 amino acids, of which only the most amino(N)-terminal portion, i.e. PTH-(1-34), is required for the classical actions of the hormone on mineral ion homeostasis mediated by the type 1 PTH/PTHrP receptor (PTH1R). Like the N-terminus, the carboxyl (C)-terminal sequence of PTH is highly conserved among species, and various circulating PTH C-fragments are generated by peripheral metabolism of intact PTH or are directly secreted, in a calcium-dependent manner, by the parathyroid glands. Certain synthetic PTH C-fragments exert actions on bone and cartilage cells that are not shared by PTH-(1-34), and specific binding of PTH C-peptides has been demonstrated in bone cells in which PTH1R expression was eliminated by gene targeting. The peptide human (h) PTH-(7-84) recently was shown to inhibit the calcemic actions of hPTH-(1-34) or hPTH-(1-84) in parathyroidectomized animals. To determine whether this anticalcemic effect of hPTH-(7-84) in vivo might result from direct actions on bone, we studied its effects on both resorption of intact bone in vitro and formation of osteoclasts in primary cultures of murine bone marrow. Human (h) PTH-(7-84) (300 nM) reduced basal 72-h release of preincorporated (45)Ca from neonatal mouse calvariae by 50% (9.6 +/- 1.9% vs. 17.8 +/- 5.7%; P < 0.001) and similarly inhibited resorption induced by hPTH-(1-84), hPTH-(1-34), 1,25-dihydroxyvitamin D(3) (VitD), PGE(2), or IL-11. In 12-d murine marrow cultures, both hPTH-(7-84) (300 nM) and hPTH-(39-84) (3000 nM) lowered VitD-dependent formation of osteoclast-like cells by 70%. On the contrary, these actions of hPTH-(7-84) were not observed with the PTH1R antagonists hPTH-(3-34)NH(2) and [L(11),D-W(12),W(23),Y(36)]hPTHrP-(7-36)NH(2), which, unlike hPTH-(7-84), did inhibit PTH1R-dependent cAMP accumulation in ROS 17/2.8 cells. We conclude that hPTH-(7-84), acting via receptors distinct from the PTH1R and presumably specific for PTH C-fragments, exerts a direct antiresorptive effect on bone that may be partly due to impaired osteoclast differentiation.


Subject(s)
Bone Resorption/physiopathology , Parathyroid Hormone/pharmacology , Peptide Fragments/pharmacology , Receptors, Parathyroid Hormone/physiology , Animals , Bone Marrow Cells/physiology , Bone Resorption/chemically induced , Calcium/metabolism , Cells, Cultured , Humans , In Vitro Techniques , Male , Mice , Mice, Inbred C57BL , Rats , Receptor, Parathyroid Hormone, Type 1 , Skull/drug effects , Skull/metabolism , Skull/physiopathology
8.
Cell ; 107(4): 513-23, 2001 Nov 16.
Article in English | MEDLINE | ID: mdl-11719191

ABSTRACT

In humans, low peak bone mass is a significant risk factor for osteoporosis. We report that LRP5, encoding the low-density lipoprotein receptor-related protein 5, affects bone mass accrual during growth. Mutations in LRP5 cause the autosomal recessive disorder osteoporosis-pseudoglioma syndrome (OPPG). We find that OPPG carriers have reduced bone mass when compared to age- and gender-matched controls. We demonstrate LRP5 expression by osteoblasts in situ and show that LRP5 can transduce Wnt signaling in vitro via the canonical pathway. We further show that a mutant-secreted form of LRP5 can reduce bone thickness in mouse calvarial explant cultures. These data indicate that Wnt-mediated signaling via LRP5 affects bone accrual during growth and is important for the establishment of peak bone mass.


Subject(s)
Bone Density/genetics , Eye Abnormalities/genetics , Eye/embryology , Osteoblasts/metabolism , Osteoporosis/genetics , Receptors, LDL/physiology , Transforming Growth Factor beta , Zebrafish Proteins , Adaptor Proteins, Signal Transducing , Adult , Animals , Animals, Outbred Strains , Bone Morphogenetic Protein 2 , Bone Morphogenetic Proteins/pharmacology , COS Cells , Child , Child, Preschool , Chlorocebus aethiops , Chromosomes, Human, Pair 11/genetics , Culture Media, Conditioned/pharmacology , DNA, Complementary/genetics , Dishevelled Proteins , Female , Genes, Recessive , Heterozygote , Humans , LDL-Receptor Related Proteins , Low Density Lipoprotein Receptor-Related Protein-5 , Male , Mesoderm/cytology , Mice , Mice, Inbred C57BL , Organ Culture Techniques , Phosphoproteins/genetics , Phosphoproteins/physiology , Proteins/genetics , Proteins/physiology , Proto-Oncogene Proteins/genetics , Proto-Oncogene Proteins/physiology , Receptors, LDL/deficiency , Receptors, LDL/genetics , Recombinant Fusion Proteins/physiology , Recombinant Proteins , Signal Transduction , Skull/cytology , Species Specificity , Stromal Cells/cytology , Stromal Cells/drug effects , Syndrome , Transfection , Wnt Proteins , Wnt-5a Protein , Wnt2 Protein , Wnt3 Protein , Wnt4 Protein
9.
Endocrinology ; 142(12): 5303-10, 2001 Dec.
Article in English | MEDLINE | ID: mdl-11713230

ABSTRACT

The homozygous ablation of the gene encoding the PTH/PTHrP receptor (PPR(-/-)) leads to early lethality and limited developmental defects, including an acceleration of chondrocyte differentiation. In contrast to the findings in homozygous PTHrP-ablated (PTHrP(-/-)) animals, these PPR(-/-) mice show an increase in cortical bone, a decrease in trabecular bone, and a defect in bone mineralization. Opposite observations are made in Jansen's metaphyseal chondrodysplasia, a disorder caused by constitutively active PPR mutants, and in transgenic animals expressing one of these receptor mutants (HKrk-H223R) under control of the type alpha1(I) collagen promoter. Expression of the Jansen transgene under the control of the type alpha1(II) collagen promoter was, furthermore, shown to delay chondrocyte differentiation and to prevent the dramatic acceleration of chondrocyte differentiation in PTHrP(-/-) mice, thus rescuing the early lethality of these animals. In the present study we demonstrated that the type alpha1(II) collagen promoter Jansen transgene restored most of the bone abnormalities in PPR(-/-) mice, but did not prevent their perinatal lethality. These findings suggested that factors other than impaired gas exchange due to an abnormal rib cage contribute to the early death of PPR(-/-) mice.


Subject(s)
Exostoses, Multiple Hereditary/genetics , Gene Expression , Gene Targeting , Receptors, Parathyroid Hormone/deficiency , Transgenes , Animals , Chondrocytes/physiology , Collagen Type II/genetics , Embryo, Mammalian/pathology , Exostoses, Multiple Hereditary/mortality , Exostoses, Multiple Hereditary/pathology , Growth Plate/metabolism , Homozygote , Mice , Mice, Knockout , Mutation , Phenotype , Promoter Regions, Genetic/physiology , Receptor, Parathyroid Hormone, Type 1 , Receptors, Parathyroid Hormone/genetics , Receptors, Parathyroid Hormone/metabolism , Survival Analysis , Tibia/embryology , Tibia/pathology
10.
J Biol Chem ; 276(46): 42692-9, 2001 Nov 16.
Article in English | MEDLINE | ID: mdl-11553625

ABSTRACT

We have investigated receptor structural components responsible for ligand-dependent inverse agonism in a constitutively active mutant of the human parathyroid hormone (PTH)/parathyroid hormone-related peptide (PTHrP) receptor type 1 (hP1R). This mutant receptor, hP1R-H223R (hP1R(CAM-HR)), was originally identified in Jansen's chondrodysplasia and is altered in transmembrane domain (TM) 2. We utilized the PTHrP analog, [Bpa(2),Ile(5),Trp(23),Tyr(36)]PTHrP-(1-36)-amide (Bpa(2)-PTHrP-(1-36)), which has valine 2 replaced by p-benzoyl-l-phenylalanine (Bpa); this substitution renders the peptide a photoreactive inverse agonist at hP1R(CAM-HR). This analog cross-linked to hP1R(CAM-HR) at two contiguous receptor regions as follows: the principal cross-link site (site A) was between receptor residues Pro(415)-Met(441), spanning the TM6/extracellular loop three boundary; the second cross-link site (site B) was within the TM4/TM5 region. Within the site A interval, substitution of Met(425) to Leu converted Bpa(2)-PTHrP-(1-36) from an inverse agonist to a weak partial agonist; this conversion was accompanied by a relative shift of cross-linking from site A to site B. The functional effect of the M425L mutation was specific for Bpa(2)-containing analogs, as inverse agonism of Bpa(2)-PTH-(1-34) was similarly eliminated, whereas inverse agonism of [Leu(11),d-Trp(12)]PTHrP-(5-36) was not affected. Overall, our data indicate that interactions between residue 2 of the ligand and the extracellular end of TM6 of the hP1R play an important role in modulating the conversion between active and inactive receptor states.


Subject(s)
Parathyroid Hormone/agonists , Parathyroid Hormone/chemistry , Receptors, Parathyroid Hormone/agonists , Receptors, Parathyroid Hormone/chemistry , Animals , COS Cells , Cattle , Cross-Linking Reagents/pharmacology , Cyclic AMP/metabolism , DNA Mutational Analysis , Dose-Response Relationship, Drug , Electrophoresis, Polyacrylamide Gel , Humans , Inhibitory Concentration 50 , Leucine/chemistry , Ligands , Mass Spectrometry , Methionine/chemistry , Models, Biological , Mutagenesis, Site-Directed , Mutation , Peptides/chemistry , Plasmids/metabolism , Protein Binding , Protein Structure, Tertiary , Rats , Transfection
11.
Hum Mol Genet ; 10(12): 1231-41, 2001 Jun 01.
Article in English | MEDLINE | ID: mdl-11406605

ABSTRACT

Pseudohypoparathyroidism type Ib (PHP-Ib) is a paternally imprinted disorder which maps to a region on chromosome 20q13.3 that comprises GNAS1 at its telomeric boundary. Exon A/B of this gene was recently shown to display a loss of methylation in several PHP-Ib patients. In nine unrelated PHP-Ib kindreds, in whom haplotype analysis and mode of inheritance provided no evidence against linkage to this chromosomal region, we confirmed lack of exon A/B methylation for affected individuals, while unaffected carriers showed no epigenetic abnormality at this locus. However, affected individuals in one kindred (Y2) displayed additional methylation defects involving exons NESP55, AS and XL, and unaffected carriers in this family showed an abnormal methylation at exon NESP55, but not at other exons. Taken together, current evidence thus suggests that distinct mutations within or close to GNAS1 can lead to PHP-Ib and the associated epigenetic changes. To further delineate the telomeric boundary of the PHP-Ib locus, the previously reported kindred F, in which patient F-V/51 is recombinant within GNAS1, was investigated with several new markers and direct nucleotide sequence analysis. These studies revealed that F-V/51 remains recombinant at a single nucleotide polymorphism (SNP) located 1.2 kb upstream of XL. No heterozygous mutation was identified between exon XL and an SNP approximately 8 kb upstream of NESP55, where this affected individual becomes linked, suggesting that the genetic defect responsible for parathyroid hormone resistance in kindred F, and probably other PHP-Ib patients, is located >or=56 kb centromeric of the abnormally methylated exon A/B. A region upstream of the known coding exons of GNAS1 is therefore predicted to exert, presumably through imprinting of exon A/B, long-range effects on G(s)alpha expression.


Subject(s)
Chromosomes, Human, Pair 20 , GTP-Binding Protein alpha Subunits, Gs/genetics , Genomic Imprinting , Pseudohypoparathyroidism/genetics , Chromosome Mapping , DNA Methylation , Exons , Genetic Linkage , Haplotypes , Humans , Molecular Sequence Data , Mutation , Pedigree , Phenotype , Pseudohypoparathyroidism/physiopathology
12.
Trends Endocrinol Metab ; 12(5): 210-7, 2001 Jul.
Article in English | MEDLINE | ID: mdl-11397646

ABSTRACT

The receptor for parathyroid hormone (PTH) and PTH-related protein (PTHrP) is a G protein-coupled receptor (GPCR) that plays a key role in controlling blood Ca(2+) concentration and endochondral bone formation. This review focuses on the molecular mechanisms by which the receptor recognizes the PTH and PTHrP peptide ligands and transmits their signal across the cell membrane. The available data suggest that there are two principal components to the ligand-receptor interaction. First, a docking interaction between the C-terminal portion of PTH(1-34) and the N-terminal extracellular domain of the receptor; and second, a weaker interaction between the N-terminal portion of the ligand and the juxtamembrane region of the receptor, which induces signal transduction. A full understanding of these processes could lead to new PTH/PTHrP receptor ligands that are effective in controlling diseases of bone and mineral metabolism, such as osteoporosis.


Subject(s)
Receptors, Parathyroid Hormone/chemistry , Receptors, Parathyroid Hormone/metabolism , Amino Acid Sequence , Animals , Binding Sites , Humans , Models, Molecular , Molecular Sequence Data , Neuropeptides/chemistry , Neuropeptides/metabolism , Parathyroid Hormone/metabolism , Parathyroid Hormone-Related Protein , Protein Structure, Tertiary , Proteins/metabolism , Receptor, Parathyroid Hormone, Type 1 , Receptor, Parathyroid Hormone, Type 2 , Signal Transduction
13.
J Biol Chem ; 276(31): 28650-8, 2001 Aug 03.
Article in English | MEDLINE | ID: mdl-11356832

ABSTRACT

The carboxyl-terminal portions of parathyroid hormone (PTH)-(1--34) and PTH-related peptide (PTHrP)-(1-36) are critical for high affinity binding to the PTH/PTHrP receptor (P1R), but the mechanism of receptor interaction for this domain is largely unknown. To identify interaction sites between the carboxyl-terminal region of PTHrP-(1--36) and the P1R, we prepared analogs of [I(5),W(23),Y(36)]PTHrP-(1--36)-amide with individual p-benzoyl-l-phenylalanine (Bpa) substitutions at positions 22--35. When tested with LLC-PK(1) cells stably transfected with human P1R (hP1R), the apparent binding affinity and the EC(50) of agonist-stimulated cAMP accumulation for each analog was, with the exception of the Bpa(24)-substituted analog, similar to that of the parent compound. The radiolabeled Bpa(23)-, Bpa(27)-, Bpa(28)-, and Bpa(33)-substituted compounds affinity-labeled the hP1R sufficiently well to permit subsequent mapping of the cross-linked receptor region. Each of these peptides cross-linked to the amino-terminal extracellular domain of the P1R: [I(5),Bpa(23),Y(36)]PTHrP-(1-36)-amide cross-linked to the extreme end of this domain (residues 33-63); [I(5),W(23),Bpa(27),Y(36)]PTHrP-(1--36)-amide cross-linked to residues 96--102; [I(5),W(23),Bpa(28),Y(36)]PTHrP-(1--36)- amide cross-linked to residues 64--95; and [I(5),W(23), Bpa(33),Y(36)]PTHrP-(1--36)-amide cross-linked to residues 151-172. These data thus predict that residues 23, 27, 28, and 33 of native PTHrP are each near to different regions of the amino-terminal extracellular receptor domain of the P1R. This information helps define sites of proximity between several ligand residues and this large receptor domain, which so far has been largely excluded from models of the hormone-receptor complex.


Subject(s)
Parathyroid Hormone-Related Protein , Parathyroid Hormone/metabolism , Peptide Fragments/metabolism , Peptide Fragments/pharmacology , Proteins/metabolism , Proteins/pharmacology , Receptors, Parathyroid Hormone/chemistry , Receptors, Parathyroid Hormone/metabolism , Affinity Labels/pharmacokinetics , Amino Acid Substitution , Animals , Binding Sites , Cell Line , Cross-Linking Reagents , Cyanogen Bromide , Cyclic AMP/metabolism , Humans , Iodine Radioisotopes , Models, Molecular , Mutagenesis, Site-Directed , Parathyroid Hormone/chemistry , Parathyroid Hormone/pharmacology , Peptide Fragments/chemistry , Phenylalanine/analogs & derivatives , Phenylalanine/pharmacokinetics , Protein Structure, Secondary , Proteins/chemistry , Radioligand Assay , Receptor, Parathyroid Hormone, Type 1 , Receptors, Parathyroid Hormone/drug effects , Recombinant Proteins/chemistry , Recombinant Proteins/metabolism , Structure-Activity Relationship , Transfection
14.
J Endocrinol ; 169(3): 613-20, 2001 Jun.
Article in English | MEDLINE | ID: mdl-11375132

ABSTRACT

In oncogenic osteomalacia (OOM), a tumor produces an unknown substance that inhibits phosphate reabsorption in the proximal tubules. This causes urinary phosphate wasting and, as a consequence, hypophosphatemic osteomalacia. To characterize this poorly understood biological tumor activity we generated aqueous extracts from several OOM tumors. Extracts from three of four tumors inhibited, dose- and time-dependently, (32)P-orthophosphate uptake by opossum kidney (OK) cells; maximum inhibition was about 45% of untreated control. Further characterization revealed that the factor is resistant to heat and several proteases, and that it has a low molecular weight. The tumor extracts also stimulated cAMP accumulation in OK cells, but not in osteoblastic ROS 17/2.8 and UMR106 cells, or in LLC-PK1 kidney cells expressing the parathyroid hormone (PTH)/PTH-related peptide receptor or the PTH-2 receptor. HPLC separation of low molecular weight fractions of the tumor extracts revealed that the flow-through of all three positive tumor extracts inhibited (32)P uptake and stimulated cAMP accumulation in OK cells. Additionally, a second peak with inhibitory activity on phosphate transport, but without cAMP stimulatory activity, was identified in the most potent tumor extract. We have concluded that several low molecular weight molecules with the ability to inhibit phosphate transport in OK cells can be found in extracts from OOM tumors. It remains uncertain, however, whether these are related to the long-sought phosphaturic factor responsible for the phosphate wasting seen in OOM patients.


Subject(s)
Kidney/metabolism , Neoplasms/metabolism , Osteomalacia/metabolism , Phosphates/metabolism , Adult , Animals , Chromatography, High Pressure Liquid , Cyclic AMP/metabolism , Female , Hot Temperature , Humans , Male , Middle Aged , Molecular Weight , Neoplasms/complications , Opossums/metabolism , Osteomalacia/etiology , Paraneoplastic Syndromes/etiology , Paraneoplastic Syndromes/metabolism , Tissue Extracts/chemistry , Tissue Extracts/pharmacology , Tumor Cells, Cultured
15.
Am J Hum Genet ; 68(5): 1283-9, 2001 May.
Article in English | MEDLINE | ID: mdl-11294659

ABSTRACT

Heterozygous inactivating mutations in the GNAS1 exons (20q13.3) that encode the alpha-subunit of the stimulatory G protein (Gsalpha) are found in patients with pseudohypoparathyroidism type Ia (PHP-Ia) and in patients with pseudo-pseudohypoparathyroidism (pPHP). However, because of paternal imprinting, resistance to parathyroid hormone (PTH)-and, sometimes, to other hormones that require Gsalpha signaling-develops only if the defect is inherited from a female carrier of the disease gene. An identical mode of inheritance is observed in kindreds with pseudohypoparathyroidism type Ib (PHP-Ib), which is most likely caused by mutations in regulatory regions of the maternal GNAS1 gene that are predicted to interfere with the parent-specific methylation of this gene. We report a patient with PTH-resistant hypocalcemia and hyperphosphatemia but without evidence for Albright hereditary osteodystrophy who has paternal uniparental isodisomy of chromosome 20q and lacks the maternal-specific methylation pattern within GNAS1. Since studies in the patient's fibroblasts did not reveal any evidence of impaired Gsalpha protein or activity, it appears that the loss of the maternal GNAS1 gene and the resulting epigenetic changes alone can lead to PTH resistance in the proximal renal tubules and thus lead to impaired regulation of mineral-ion homeostasis.


Subject(s)
Aneuploidy , Chromosomes, Human, Pair 20/genetics , DNA Methylation , GTP-Binding Protein alpha Subunits, Gs/genetics , Genomic Imprinting/genetics , Pseudohypoparathyroidism/genetics , Adult , Calcitriol/therapeutic use , Calcium/therapeutic use , Child , Child, Preschool , Cyclic AMP/metabolism , Female , GTP-Binding Protein alpha Subunits, Gs/metabolism , Haplotypes/genetics , Humans , Infant , Infant, Newborn , Male , Models, Genetic , Mutation/genetics , Pedigree , Pseudohypoparathyroidism/drug therapy , Pseudohypoparathyroidism/metabolism , Pseudohypoparathyroidism/physiopathology
16.
J Clin Endocrinol Metab ; 86(2): 497-500, 2001 Feb.
Article in English | MEDLINE | ID: mdl-11157998

ABSTRACT

The gene mutated in autosomal dominant hypophosphatemic rickets (ADHR), a phosphate wasting disorder, has been identified as FGF-23, a protein that shares sequence homology with fibroblast growth factors (FGFs). Patients with ADHR display many of the clinical and laboratory characteristics that are observed in patients with oncogenic hypophosphatemic osteomalacia (OHO), a disorder thought to arise by the secretion of a phosphate wasting factor from different mesenchymal tumors. In the present studies, we therefore investigated whether FGF-23 is a secreted factor and whether it is abundantly expressed in OHO tumors. After transient transfection of OK-E, COS-7, and HEK293 cells with the plasmid encoding full-length FGF-23, all three cell lines efficiently secreted two protein species into the medium that were approximately 32 and 12 kDa upon SDS-PAGE and subsequent Western blot analysis using an affinity-purified polyclonal antibody to FGF-23. Furthermore, Northern blot analysis using total RNA from five different OHO tumors revealed extremely high levels of FGF-23 mRNA, and Western blot analysis of extracts from a sixth tumor detected the 32 kDa FGF-23 protein species. In summary, FGF-23, the gene mutated in ADHR, is a secreted protein and its mRNA is abundantly expressed by several different OHO tumors. Our findings indicate that FGF-23 may be a candidate phosphate wasting factor, previously designated "phosphatonin".


Subject(s)
Fibroblast Growth Factors/genetics , Hypophosphatemia, Familial/genetics , Mesenchymoma/physiopathology , Animals , CHO Cells , Cell Line , Cricetinae , Fibroblast Growth Factor-23 , Humans , Hypophosphatemia, Familial/complications , Hypophosphatemia, Familial/physiopathology , Mesenchymoma/complications , Molecular Sequence Data , Osteomalacia/physiopathology , RNA, Messenger/genetics , Recombinant Proteins/biosynthesis , Transcription, Genetic , Transfection
17.
Endocrinology ; 142(2): 704-9, 2001 Feb.
Article in English | MEDLINE | ID: mdl-11159842

ABSTRACT

The tuberoinfundibular peptide TIP39 [TIP-(1-39)], which exhibits only limited amino acid sequence homology with PTH and PTH-related peptide (PTHrP), stimulates cAMP accumulation in cells expressing the PTH2 receptor (PTH2R), but it is inactive at the PTH/PTHrP receptor (PTH1R). However, when using either (125)I-labeled rat [Nle(8,21),Tyr(34)]PTH-(1-34)amide (rPTH) or (125)I-labeled human [Tyr(36)]PTHrP-(1-36)amide [PTHrP-(1-36)] for radioreceptor studies, TIP-(1-39) bound to LLCPK(1) cells stably expressing the PTH1R (HKrk-B7 cells), albeit with weak apparent affinity (243 +/- 52 and 210 +/- 64 nM, respectively). In comparison to the parent peptide, the apparent binding affinity of TIP-(3-39) was about 3-fold higher, and that of TIP-(9-39) was about 5.5-fold higher. However, despite their improved IC(50) values at the PTH1R, both truncated peptides failed to stimulate cAMP accumulation in HKrk-B7 cells. In contrast, the chimeric peptide PTHrP-(1-20)/TIP-(23-39) bound to HKrk-B7 cells with affinities of 31 +/- 8.2 and 11 +/- 4.0 nM when using radiolabeled rPTH and PTHrP-(1-36), respectively, and it stimulated cAMP accumulation in HKrk-B7 and SaOS-2 cells with potencies (EC(50), 1.40 +/- 0.3 and 0.38 +/- 0.12 nM, respectively) and efficacies (maximum levels, 39 +/- 8 and 31 +/- 3 pmol/well, respectively) similar to those of PTH-(1-34) and PTHrP-(1-36). In both cell lines, TIP(9-39) and, to a lesser extent, TIP-(1-39) inhibited the actions of the three agonists with efficiencies similar to those of [Leu(11),D-Trp(12),Trp(23),Tyr(36)]PTHrP-(7-36)amide, an established PTH1R antagonist. Taken together, the currently available data suggest that the carboxyl-terminal portion of TIP-(1-39) interacts efficiently with the PTH1R, at sites identical to or closely overlapping those used by PTH-(1-34) and PTHrP-(1-36). The amino-terminal residues of TIP-(1-39), however, are unable to interact productively with the PTH1R, thus enabling TIP-(1-39) and some of its truncated analogs to function as an antagonist at this receptor.


Subject(s)
Neuropeptides/physiology , Receptors, Parathyroid Hormone/metabolism , Amino Acid Sequence/genetics , Animals , Binding, Competitive , Cell Line , Chimera , Cyclic AMP/metabolism , LLC-PK1 Cells , Molecular Sequence Data , Mutation/physiology , Neuropeptides/chemistry , Neuropeptides/genetics , Neuropeptides/metabolism , Peptide Fragments/physiology , Receptor, Parathyroid Hormone, Type 1 , Receptors, Parathyroid Hormone/antagonists & inhibitors , Receptors, Parathyroid Hormone/chemistry , Receptors, Parathyroid Hormone/genetics , Swine
18.
Endocrinology ; 142(2): 916-25, 2001 Feb.
Article in English | MEDLINE | ID: mdl-11159865

ABSTRACT

PTH is a potent systemic regulator of cellular differentiation and function in bone. It acts upon cells of the osteoblastic lineage via the G protein-coupled type-1 PTH/PTH-related peptide receptor (PTH1R). Carboxyl fragments of intact PTH(1-84) (C-PTH fragments) are cosecreted with it by the parathyroid glands in a calcium-dependent manner and also are generated via proteolysis of the hormone in peripheral tissues. Receptors that recognize C-PTH fragments (CPTHRs) have been described previously in osteoblastic and chondrocytic cells. To directly study CPTHRs in bone cells, we isolated clonal, conditionally transformed cell lines from fetal calvarial bone of mice that are homozygous for targeted ablation of the PTH1R gene and transgenically express a temperature-sensitive mutant SV40 T antigen. Cells with the highest specific binding of the CPTHR radioligand (125)I-[Tyr(34)]hPTH(19-84) exhibited a stellate, dendritic appearance suggestive of an osteocytic phenotype and expressed 6- to 10-fold more CPTHR sites/cell than did osteoblastic cells previously isolated from the same bones. In these osteocytic (OC) cells, expression of mRNAs for CD44, connexin 43, and osteocalcin was high, whereas that for alkaline phosphatase and cbfa-1/osf-2 was negligible. The CPTHR radioligand was displaced completely by hPTH(1-84), hPTH(19-84) and hPTH(24-84) (IC(50)s = 20-50 nM) and by hPTH(39-84) (IC(50) = 500 nM) but only minimally (24%) by 10,000 nM hPTH(1-34). CPTHR binding was down-regulated dose dependently by hPTH(1-84), an effect mimicked by ionomycin and active phorbol ester. Human PTH(1-84) and hPTH(39-84) altered connexin 43 expression and increased apoptosis in OC cells. Apoptosis induced by PTH(1-84) was blocked by the caspase inhibitor DEVD. We conclude that osteocytes, the most abundant cells in bone, may be principal target cells for unique actions of intact PTH(1-84) and circulating PTH C-fragments that are mediated by CPTHRs.


Subject(s)
Osteocytes/metabolism , Parathyroid Hormone/genetics , Parathyroid Hormone/metabolism , Peptide Fragments/metabolism , Receptors, Parathyroid Hormone/metabolism , Animals , Apoptosis/drug effects , Cells, Cultured , Humans , Ligands , Mice , Mice, Transgenic/genetics , Osteocytes/physiology , Osteocytes/ultrastructure , Parathyroid Hormone/pharmacology , Peptide Fragments/genetics
19.
Endocrinol Metab Clin North Am ; 29(3): 569-89, 2000 Sep.
Article in English | MEDLINE | ID: mdl-11033761

ABSTRACT

The GNAS1 gene (chromosome 20q13.3) encodes the alpha subunit of the stimulatory G protein (Gs alpha) and at least three additional, alternatively spliced transcripts, XL alpha s, NESP55, and the antisense transcript AS. Gs alpha transcripts seem to be derived exclusively, at least in the renal cortex, from the maternal allele. XL alpha s and AS are transcribed only from the paternal allele, and NESP55 is transcribed only from the maternal allele. Numerous GNAS1 mutations have been identified in PHP-Ia and pPHP. Patients with either disorder show skeletal and developmental defects now referred to as AHO. Owing to paternal imprinting, that is, inactivation of the paternal allele, which may be tissue- or cell-specific, resistance toward PTH and, often, other hormones is only observed in patients with PHP-Ia. Patients with PHP-Ib show PTH-resistant hypocalcemia and hyperphosphatemia but no AHO. The abnormal regulation of mineral ion homeostasis is paternally imprinted, such as in PHP-Ia/pPHP kindreds, Gs alpha activity/protein is normal in fibroblasts and blood cells, and no GNAS1 mutations have been identified. Recent linkage studies have mapped the genetic defect responsible for PHP-Ib to chromosome 20q13.3, making it likely that mutations in distinct regions of the GNAS1 gene are the cause of at least three different forms of PHP.


Subject(s)
Pseudohypoparathyroidism/physiopathology , Animals , Female , GTP-Binding Protein alpha Subunits, Gs/genetics , Genomic Imprinting , Heterozygote , Humans , Male , Mutagenesis , Oncogene Proteins/genetics , Parathyroid Hormone/physiology , Pedigree , Phenotype , Pseudohypoparathyroidism/genetics
20.
Mol Cell Probes ; 14(4): 261-4, 2000 Aug.
Article in English | MEDLINE | ID: mdl-10970731

ABSTRACT

GNAS1, which is located in the chromosomal region 20q13.3, gives rise to maternally, paternally or bi-allelically expressed transcripts including the one that encodes the alpha subunit of the stimulatory G protein. Numerous naturally occurring mutations of this gene have been identified in several different disorders including certain forms of pseudohypoparathyroidism, progressive osseous heteroplasia, McCune-Albright syndrome and acromegaly. Polymorphic markers currently employed in the genetic evaluation of these disorders frequently prove uninformative owing to a low heterozygosity value associated with each marker. We searched for potentially polymorphic tandem repeats close to the GNAS1 locus, and identified two new, highly polymorphic loci that are located within a;48-kb region immediately downstream of this gene. These new microsatellite markers, with their high polymorphism information content, may prove to be useful in genetic studies related to GNAS1 as well as to other genes located in the flanking genomic region.


Subject(s)
Chromosomes, Human, Pair 20 , GTP-Binding Protein alpha Subunits, Gs/genetics , Polymorphism, Genetic , Asian People/genetics , Black People/genetics , Genetics, Population , Heterozygote , Humans , Microsatellite Repeats , Repetitive Sequences, Nucleic Acid , White People/genetics
SELECTION OF CITATIONS
SEARCH DETAIL
...