Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 16 de 16
Filter
1.
Anticancer Drugs ; 32(4): 365-375, 2021 04 01.
Article in English | MEDLINE | ID: mdl-33595947

ABSTRACT

Patients with advanced breast cancer often develop bone metastases. Treatment is limited to palliative care. Parathyroid hormone (PTH)/parathyroid hormone-related peptide (PTHrP) antagonists for bone metastases failed clinically due to short half-life and inadequate concentration in bone. We synthesized two novel PTHrP antagonists fused to an inert bacterial collagen binding domain (CBD) that directs drugs to bone. PTH(7-33)-CBD is an N-terminal truncated PTHrP antagonist. [W2]PTH(1-33)-CBD is an PTHrP inverse-agonist. The aim of this study was to assess PTH(7-33)-CBD to reduce breast cancer bone metastases and prevent osteolytic destruction in mice and to assess both drugs for apoptosis of breast cancer cells in vitro and inhibition of PTH receptor (PTHR1). PTH(7-33)-CBD (1000 µg/kg, subcutaneous) or vehicle was administered 24 h prior to MDA-MB-231 breast cancer cell inoculation into the tibia marrow. Weekly tumor burden and bone density were measured. Pharmacokinetic analysis of PTH(7-33)-CBD in rat serum was evaluated. Drug effect on cAMP accumulation in SaOS-2 osteosarcoma cells and apoptosis of MDA-MB-231 cells was assessed. PTH(7-33)-CBD reduced MDA-MB-231 tumor burden and osteolytic destruction in mice 4-5 weeks post-treatment. PTH(7-33)-CBD (1000 µg/kg i.v. and subcutaneous) in rats was rapidly absorbed with peak concentration 5-min and terminal half-life 3-h. Bioavailability by the subcutaneous route was 43% relative to the i.v. route. PTH(7-33)-CBD was detected only on rat periosteal bone surfaces that stained positive for collagen-1. PTH(7-33)-CBD and [W2]PTH(1-33)-CBD (10-8M) blocked basal and PTH agonist-induced cAMP accumulation in SaOS-2 osteosarcoma cells. Both drugs induced PTHR1-dependent apoptosis of MDA-MB-231 cells in vitro. Novel bone-targeted PTHrP antagonists represent a new paradigm for treatment of breast cancer bone metastases.


Subject(s)
Bone Neoplasms/prevention & control , Bone Neoplasms/secondary , Breast Neoplasms/drug therapy , Breast Neoplasms/pathology , Parathyroid Hormone/antagonists & inhibitors , Peptide Fragments/pharmacology , Animals , Bone Density/drug effects , Cell Line, Tumor , Female , Mice , Mice, Inbred BALB C , Mice, Nude , Parathyroid Hormone-Related Protein/antagonists & inhibitors , Xenograft Model Antitumor Assays
2.
Horm Res Paediatr ; 90(3): 151-160, 2018.
Article in English | MEDLINE | ID: mdl-30261514

ABSTRACT

BACKGROUND/AIMS: Patients with short stature (SS)/growth hormone deficiency (GHD) and precocious puberty (PP) undergo brain MRI to evaluate for structural brain abnormalities or pituitary lesions, and pituitary microadenomas are a common finding. Theoretically, a mass effect from these lesions could cause GHD and growth hormone treatment could cause them to enlarge, but they should not cause PP, at least in females. METHODS: We investigated if pituitary microadenomas cause GHD by comparing their incidence in patients with SS/GHD to that in females with PP. We performed a retrospective chart review of patients with these disorders who had a brain MRI between 2000 and 2013. RESULTS: The incidence of microadenoma was high in both groups, 18.5% for SS (n = 346) and 21.1% for PP females (n = 194), but did not differ between groups (p = 0.46). In patients with microadenomas, repeat imaging showed resolution in 58% (SS, n = 33) and 67% (PP females, n = 21). Importantly, none of the lesions grew, even in patients treated with growth hormone. CONCLUSIONS: Pituitary microadenomas are common in children with GHD/SS and PP, but it does not appear that they are a cause of GHD. They appear to be of limited clinical significance and should not be considered a contraindication to growth hormone therapy.


Subject(s)
Adenoma/diagnosis , Adenoma/epidemiology , Dwarfism, Pituitary/diagnosis , Dwarfism, Pituitary/epidemiology , Human Growth Hormone/deficiency , Pituitary Neoplasms/diagnosis , Pituitary Neoplasms/epidemiology , Adenoma/complications , Adenoma/drug therapy , Adolescent , Child , Dwarfism, Pituitary/drug therapy , Dwarfism, Pituitary/etiology , Female , Follow-Up Studies , Human Growth Hormone/therapeutic use , Humans , Incidence , Magnetic Resonance Imaging , Male , Pituitary Neoplasms/complications , Pituitary Neoplasms/drug therapy , Prognosis , Retrospective Studies , Tumor Burden
3.
J Investig Dermatol Symp Proc ; 17(2): 13-5, 2015 Nov.
Article in English | MEDLINE | ID: mdl-26551937

ABSTRACT

UNLABELLED: Alopecia areata is a common disorder in which autoimmune destruction of hair follicles results in patchy hair loss. Currently there is no adequate therapy, although immune modulator therapies are currently in development. Parathyroid hormone (PTH) is a hair cycle stimulator which shows promise in treating various forms of alopecia, although its short half-life limits its clinical use. PTH-CBD is a PTH analog which binds collagen, prolonging retention in skin. We tested effects of PTH-CBD in C3H/HeJ-engrafted mice, the animal model for alopecia areata, on hair growth and found that a significant proportion of animals had reduced hair loss (PTH-CBD: 13/21, 62% vs. CONTROL: 3/10, 30%; P<0.01). Histological analysis showed no change in immune response, but there was increased number of anagen hair follicles and increased production of beta-catenin, a factor which initiates the anagen phase of the hair cycle. PTH-CBD thus shows promise as a therapy for alopecia areata, either alone or in conjunction with immune modulation therapy.


Subject(s)
Alopecia Areata/drug therapy , Hair Follicle/drug effects , Parathyroid Hormone/agonists , Recombinant Fusion Proteins/therapeutic use , Alopecia Areata/immunology , Alopecia Areata/pathology , Animals , Disease Models, Animal , Hair/growth & development , Hair Follicle/pathology , Mice , beta Catenin/metabolism
4.
Exp Dermatol ; 23(12): 865-7, 2014 Dec.
Article in English | MEDLINE | ID: mdl-25041375

ABSTRACT

While the effects of PTHrP have been studied for almost 20 years, most of these studies have focused on effects on the termination of the anagen phase, giving an incomplete picture of the overall effect of PTHrP on the hair cycle. PTHrP was determined in several experimental models to promote transition of hair follicles from anagen to catagen phase, which by itself would suggest that PTHrP blockade might prolong the anagen phase and promote hair growth. However, clinical trials with topically applied PTHrP antagonists have been disappointing, leading to a reconsideration of this model. Additional studies performed in mouse models where hair follicles are damaged (alopecia areata, chemotherapy-induced alopecia) suggest that PTHrP has effects early in the hair cycle as well, promoting hair follicles' entry into anagen phase and initiates the hair cycle. While the mechanism of this has yet to be elucidated, it may involve activation of the Wnt pathway. Thus, the overall effect of PTHrP is to stimulate and accelerate the hair cycle, and in the more clinically relevant models of hair loss where hair follicles have been damaged or become quiescent, it is the agonists, not the antagonists, which would be expected to promote hair growth.


Subject(s)
Hair/growth & development , Parathyroid Hormone-Related Protein/physiology , Alopecia/drug therapy , Alopecia/pathology , Animals , Disease Models, Animal , Hair/drug effects , Hair Follicle/drug effects , Hair Follicle/pathology , Humans , Mice , Parathyroid Hormone-Related Protein/agonists , Parathyroid Hormone-Related Protein/antagonists & inhibitors
5.
J Investig Dermatol Symp Proc ; 16(1): S58, 2013 Dec.
Article in English | MEDLINE | ID: mdl-24326561

ABSTRACT

Reported is a patient with severe alopecia areata, multiple autoimmune diseases (chronic lymphocytic thyroidis, primary ovarian failure), and Down syndrome. She had a poor response to topical treatment with glucocorticoids and minoxidil, but showed some improvement with glucocorticoid injections. At the time of evaluation, she had hair loss on 85-90% of her scalp. She was treated initially with oral prednisone 50 mg per day for 2 weeks, followed by a 3-month course of prednisone 10 mg per day and cyclosporine 125 mg (4 mg kg(-1)) two times per day. She responded well with excellent regrowth of hair on the scalp, and prednisone was tapered and ultimately discontinued. Importantly, her parents noted marked improvement in sense of well-being. Several months after discontinuing treatment, she developed hyperpigmentation on the trunk consistent with confluent and reticulated papillomatosis; she has several known risk factors for this disorder, but it is not clear if this is related to her previous treatment.


Subject(s)
Alopecia Areata/drug therapy , Anti-Inflammatory Agents/therapeutic use , Cyclosporine/therapeutic use , Immunosuppressive Agents/therapeutic use , Prednisone/therapeutic use , Adolescent , Alopecia Areata/complications , Down Syndrome/complications , Drug Therapy, Combination , Female , Hashimoto Disease/complications , Humans , Papilloma/complications , Primary Ovarian Insufficiency/complications , Skin Neoplasms/complications
6.
Calcif Tissue Int ; 91(3): 196-203, 2012 Sep.
Article in English | MEDLINE | ID: mdl-22806683

ABSTRACT

Parathyroid hormone (PTH) is the most effective osteoporosis treatment, but it is only effective if administered by daily injections. We fused PTH(1-33) to a collagen binding domain (PTH-CBD) to extend its activity, and have shown an anabolic bone effect with monthly dosing. We tested the duration of action of this compound with different routes of administration. Normal young C57BL/6J mice received a single intraperitoneal injection of PTH-CBD (320 µg/kg). PTH-CBD treated mice showed a 22.2 % increase in bone mineral density (BMD) at 6 months and 12.8 % increase at 12 months. When administered by subcutaneous injection, PTH-CBD again caused increases in BMD, 15.2 % at 6 months and 14.3 % at 12 months. Radiolabeled PTH-CBD was concentrated in bone and skin after either route of administration. We further investigated skin effects of PTH-CBD, and histological analysis revealed an apparent increase in anagen VI hair follicles. A single dose of PTH-CBD caused sustained increases in BMD by >10 % for 1 year in normal mice, regardless of the route of administration, thus showing promise as a potential osteoporosis therapy.


Subject(s)
Anabolic Agents/administration & dosage , Bacterial Proteins/genetics , Bone Density/drug effects , Collagen/metabolism , Collagenases/genetics , Parathyroid Hormone/genetics , Recombinant Fusion Proteins/administration & dosage , Anabolic Agents/pharmacology , Animals , Female , Injections, Subcutaneous , Mice , Mice, Inbred BALB C , Recombinant Fusion Proteins/genetics , Recombinant Fusion Proteins/pharmacology
7.
Int J Cancer ; 131(5): E813-21, 2012 Sep 01.
Article in English | MEDLINE | ID: mdl-22130912

ABSTRACT

Parathyroid hormone (PTH) agonists and antagonists have been shown to improve hair growth after chemotherapy; however, rapid clearance and systemic side-effects complicate their usage. To facilitate delivery and retention to skin, we fused PTH agonists and antagonists to the collagen binding domain (CBD) of Clostridium histolyticum collagenase. in-vitro studies showed that the agonist fusion protein, PTH-CBD, bound collagen and activated the PTH/parathyroid hormone-related peptide receptor in SaOS-2 cells. The antagonist fusion proteins, PTH(7-33)-CBD and PTH([-1]-33)-CBD, also bound collagen and antagonized PTH(1-34) effect in SaOS-2 cells; however, PTH(7-33)-CBD had lower intrinsic activity. Distribution studies confirmed uptake of PTH-CBD to the skin at 1 and 12 hr after subcutaneous injection. We assessed in vivo efficacy of PTH-CBD and PTH(7-33)-CBD in C57BL/6J mice. Animals were depilated to synchronize the hair follicles; treated on Day 7 with agonist, antagonist, or vehicle; treated on Day 9 with cyclophosphamide (150 mg/kg i.p.) or vehicle; and sacrificed on Day 39. Normal mice (no chemo and no treatment) showed rapid regrowth of hair and normal histology. Chemo+Vehicle mice showed reduced hair regrowth and decreased pigmentation; histology revealed reduced number and dystrophic anagen/catagen follicles. Chemo+Antagonist mice were grossly and histologically indistinguishable from Chemo+Vehicle mice. Chemo+Agonist mice showed more rapid regrowth and repigmentation of hair; histologically, there was a normal number of hair follicles, most of which were in the anagen phase. Overall, the agonist PTH-CBD had prominent effects in reducing chemotherapy-induced damage of hair follicles and may show promise as a therapy for chemotherapy-induced alopecia.


Subject(s)
Alopecia/drug therapy , Collagen/metabolism , Cyclophosphamide/adverse effects , Hormone Antagonists/pharmacology , Parathyroid Hormone/agonists , Parathyroid Hormone/antagonists & inhibitors , Peptide Fragments/pharmacology , Alopecia/chemically induced , Alopecia/metabolism , Amino Acid Sequence , Animals , Bone Density/drug effects , Female , Immunosuppressive Agents/adverse effects , Mice , Mice, Inbred C57BL , Molecular Sequence Data , Protein Binding , Receptor, Parathyroid Hormone, Type 1/metabolism
8.
Toxicol Mech Methods ; 19(3): 225-31, 2009 Mar.
Article in English | MEDLINE | ID: mdl-19750018

ABSTRACT

Dual Energy X-ray Absorptiometry (DXA) is effective in measuring bone mineral density (BMD) in mice for early detection of osteoporosis. However, scanners designed for use with small animals (i.e. PIXImus) are very expensive. Used human DXA machines are cheaper to obtain, but analysis of scans from these instruments is operator-dependent. Obtaining reliable data depends on having a single operator analyze the scans in a blinded fashion. Scan quality is improved by excising the bone prior to scanning, which does not allow serial measurements. This study describes a novel method of analyzing lumbar spine BMD in mice using whole body DXA. This non-invasive technique has a high degree of precision and reproducibility, with good correlation between multiple observers. Inter-observer variability (0.063 +/- 0.00317 g/cm(2) [mean +/- SD], 5.05 [% coefficient of variation (CV)], repeat scan variability (0.063 +/- 0.00364 g/cm(2) [mean +/- SD], 5.94 [%CV]) were very low compared to variability between different animals (0.063 +/- 0.00588 g/cm(2) [mean +/- SD], 9.64 [%CV]) and variability seen in same animal over time (0.011 +/- 0.00885 g/cm(2) [mean +/- SD], 80.68 [%CV]). The measurement error is thus smaller than the biological variation. Accuracy was determined by comparing average peak BMD from two scans per mouse in-vivo (0.066 g/cm(2)) versus excised spine (0.065 g/cm(2)). Furthermore, correlation between bone ash weights and whole body lumbar spine BMD measurements (p < 0.0001) was highly significant. This technique thus shows a high degree of precision and accuracy, even with multiple observers, for measuring BMD in mice using a DXA machine designed for clinical use.


Subject(s)
Absorptiometry, Photon/methods , Bone Density , Lumbar Vertebrae , Animals , Female , Mice , Mice, Inbred C57BL , Osteoporosis/diagnostic imaging , Reproducibility of Results
9.
J Clin Invest ; 115(5): 1250-7, 2005 May.
Article in English | MEDLINE | ID: mdl-15864348

ABSTRACT

Infantile cortical hyperostosis (Caffey disease) is characterized by spontaneous episodes of subperiosteal new bone formation along 1 or more bones commencing within the first 5 months of life. A genome-wide screen for genetic linkage in a large family with an autosomal dominant form of Caffey disease (ADC) revealed a locus on chromosome 17q21 (LOD score, 6.78). Affected individuals and obligate carriers were heterozygous for a missense mutation (3040Ctwo head right arrowT) in exon 41 of the gene encoding the alpha1(I) chain of type I collagen (COL1A1), altering residue 836 (R836C) in the triple-helical domain of this chain. The same mutation was identified in affected members of 2 unrelated, smaller families with ADC, but not in 2 prenatal cases and not in more than 300 chromosomes from healthy individuals. Fibroblast cultures from an affected individual produced abnormal disulfide-bonded dimeric alpha1(I) chains. Dermal collagen fibrils of the same individual were larger, more variable in shape and size, and less densely packed than those in control samples. Individuals bearing the mutation, whether they had experienced an episode of cortical hyperostosis or not, had joint hyperlaxity, hyperextensible skin, and inguinal hernias resembling symptoms of a mild form of Ehlers-Danlos syndrome type III. These findings extend the spectrum of COL1A1-related diseases to include a hyperostotic disorder.


Subject(s)
Bone and Bones/physiopathology , Collagen Type I/metabolism , Hyperostosis, Cortical, Congenital/physiopathology , Chromosome Mapping , Chromosomes, Human, Pair 17 , Collagen Type I/genetics , Collagen Type I, alpha 1 Chain , Dermis/pathology , Dermis/ultrastructure , Female , Fibula/diagnostic imaging , Haplotypes , Humans , Hyperostosis, Cortical, Congenital/genetics , Infant , Male , Mutation , Pedigree , Radiography , Tibia/diagnostic imaging
10.
Am J Hum Genet ; 76(5): 804-14, 2005 May.
Article in English | MEDLINE | ID: mdl-15800843

ABSTRACT

A unique heterozygous 3-kb microdeletion within STX16, a closely linked gene centromeric of GNAS, was previously identified in multiple unrelated kindreds as a cause of autosomal dominant pseudohypoparathyroidism type Ib (AD-PHP-Ib). We now report a novel heterozygous 4.4-kb microdeletion in a large kindred with AD-PHP-Ib. Affected individuals from this kindred share an epigenetic defect that is indistinguishable from that observed in patients with AD-PHP-Ib who carry the 3-kb microdeletion in the STX16 region (i.e., an isolated loss of methylation at GNAS exon A/B). The novel 4.4-kb microdeletion overlaps with the previously identified deletion by 1,286 bp and, similar to the latter deletion, removes several exons of STX16 (encoding syntaxin-16). Because these microdeletions lead to AD-PHP-Ib only after maternal transmission, we analyzed expression of this gene in lymphoblastoid cells of affected individuals with the 3-kb or the 4.4-kb microdeletion, an individual with a NESP55 deletion, and a healthy control. We found that STX16 mRNA was expressed in all cases from both parental alleles. Thus, STX16 is apparently not imprinted, and a loss-of-function mutation in one allele is therefore unlikely to be responsible for this disorder. Instead, the region of overlap between the two microdeletions likely harbors a cis-acting imprinting control element that is necessary for establishing and/or maintaining methylation at GNAS exon A/B, thus allowing normal G alpha(s) expression in the proximal renal tubules. In the presence of either of the two microdeletions, parathyroid hormone resistance appears to develop over time, as documented in an affected individual who was diagnosed at birth with the 4.4-kb deletion of STX16 and who had normal serum parathyroid hormone levels until the age of 21 mo.


Subject(s)
GTP-Binding Protein alpha Subunits, Gs/genetics , Pseudohypoparathyroidism/genetics , Vesicular Transport Proteins/genetics , Chromogranins , DNA Methylation , Female , Gene Deletion , Genes, Dominant , Genomic Imprinting , Humans , Male , Microsatellite Repeats , Pedigree , Syntaxin 16
11.
Biochem Biophys Res Commun ; 328(3): 666-78, 2005 Mar 18.
Article in English | MEDLINE | ID: mdl-15694400

ABSTRACT

Parathyroid hormone (PTH) has a central role in the regulation of serum calcium and phosphate, while parathyroid hormone-related peptide (PTHrP) has important developmental roles. Both peptides signal through the same receptor, the PTH/PTHrP receptor (a class B G-protein-coupled receptor). The different biological effects of these ligands result from their modes of regulation and secretion, endocrine vs. paracrine/autocrine. The importance of PTH and PTHrP is evident by the variety of clinical syndromes caused by deficiency or excess production of either peptide, and the demonstration that intermittent injection of PTH increases bone mass, and thus provides a means to treat osteoporosis. This, in turn, has triggered increased interest in understanding the mechanisms of PTH/PTHrP receptor action and the search for smaller peptide or non-peptide agonists that have efficacy at this receptor when administered non-parenterally.


Subject(s)
Bone and Bones/embryology , Bone and Bones/metabolism , Parathyroid Hormone-Related Protein/metabolism , Parathyroid Hormone/metabolism , Receptor, Parathyroid Hormone, Type 1/metabolism , Receptors, Parathyroid Hormone/metabolism , Animals , Humans , Models, Biological , Models, Chemical , Parathyroid Hormone/chemistry , Parathyroid Hormone-Related Protein/chemistry , Receptor, Parathyroid Hormone, Type 1/chemistry , Receptors, Parathyroid Hormone/chemistry , Signal Transduction/physiology , Structure-Activity Relationship
12.
Mol Cell Probes ; 18(5): 353-7, 2004 Oct.
Article in English | MEDLINE | ID: mdl-15294324

ABSTRACT

The parathyroid hormone (PTH)/PTH-related peptide receptor is a critical component in the control of mineral ion metabolism and in bone development. This receptor is encoded by a single gene (PTHR1) on chromosome 3p21.1-p24.2, and mutations in this gene have been found in several clinical disorders of bone and mineral metabolism. To facilitate future genetic studies of this important gene, we determined haplotype frequencies and performed linkage disequilibrium (LD) analysis of four different polymorphisms at the PTHR1 locus. Combined analysis of Caucasian, African-American and Asian individuals indicated that LD exists between all but one pair of the four polymorphisms. However, the pattern of LD differed substantially among the three subpopulations; for example, LD between two closely spaced (154-bp apart) single nucleotide polymorphisms appeared to be present only in Asians. Depending on the population under study, genetic association studies may need to test even more closely spaced polymorphic markers when screening the PTHR1 locus. These findings may thus affect the design and interpretation of future genetic studies involving PTHR1.


Subject(s)
Haplotypes/genetics , Linkage Disequilibrium/genetics , Polymorphism, Single Nucleotide/genetics , Receptor, Parathyroid Hormone, Type 1/genetics , Black or African American/genetics , Asian People/genetics , Humans , White People/genetics
13.
Endocrinology ; 145(11): 5294-304, 2004 Nov.
Article in English | MEDLINE | ID: mdl-15297442

ABSTRACT

Although the PTH type 2 receptor (PTH2R) has been isolated from mammals and zebrafish, only its mammalian agonist, tuberoinfundibular peptide 39 (TIP39), has been characterized thus far. To determine whether zebrafish TIP39 (zTIP39) functions similarly with the zebrafish PTHR (zPTH2R) and human PTH2Rs and to determine its tissue-specific expression, fugu (Takifugu rubripes) and zebrafish (Danio rerio) genomic databases were screened with human TIP39 (hTIP39) sequences. A single TIP39 gene was identified for each fish species, which showed significant homology to mammalian TIP39. Using standard molecular techniques, we isolated cDNA sequences encoding zTIP39. The fugu TIP39 precursor was encoded by a gene comprising at least three exons. It contained a hydrophobic signal sequence and a predicted prosequence with a dibasic cleavage site, similar to that found in mammalian TIP39 ligands. Phylogenetic analyses suggested that TIP39 forms the basal group from which PTH and PTHrP have been derived. Functionally, subtle differences in potency could be discerned between hTIP39 and zTIP39. The human PTH2R and zPTH2R were stimulated slightly better by both hTIP39 and zTIP39, whereas zTIP39 had a higher potency at a previously isolated zPTH2R splice variant. Whole-mount in situ hybridization of zebrafish revealed strong zTIP39 expression in the region of the hypothalamus and in the heart of 24- and 48-h-old embryos. Similarly, zPTH2R expression was highly expressed throughout the brain of 48- and 72-h-old embryos. Because the mammalian PTH2R was also most abundantly expressed in these tissues, the TIP39-PTH2R system may serve conserved physiological roles in mammals and fishes.


Subject(s)
Neuropeptides/genetics , Takifugu/genetics , Zebrafish/genetics , Amino Acid Sequence , Animals , Base Sequence , COS Cells , Cloning, Molecular , Cyclic AMP/metabolism , DNA, Complementary , Gene Expression Regulation, Developmental , Molecular Sequence Data , Neuropeptides/metabolism , Parathyroid Hormone/genetics , Parathyroid Hormone-Related Protein/genetics , Phylogeny
14.
Endocrinology ; 145(4): 1634-9, 2004 Apr.
Article in English | MEDLINE | ID: mdl-14684608

ABSTRACT

Zebrafish (Danio rerio) have receptors homologous to the human PTH (hPTH)/PTHrP receptor (PTH1R) and PTH-2 receptor (PTH2R) and an additional receptor (PTH3R) with high homology to the PTH1R. To find natural ligands for zPTH1R and zPTH3R, we searched the zebrafish genomic database and discovered two distinct regions that, when translated (zPTH1 and zPTH2), showed high homology to hPTH. Isolation of cDNAs and determination of the intron/exon boundaries revealed genomic structures which were similar to known PTHs. Peptides consisting of the first 34 amino acids after the pre- and prosequences of the zebrafish PTHs (zPTHs) were synthesized and were shown to be fully active at the hPTH1R. zPTH2(1-34) was, however, approximately 30-fold less potent at the zPTH1R than hPTH(1-34), hPTHrP(1-36), and zPTH1(1-34). When tested with zPTH3R, zPTH1(1-34) and hPTHrP(1-36) showed similar potencies, whereas the potency of zPTH2(1-34) was moderately (3-fold) reduced. To determine whether other fishes have multiple PTHs, we searched the genomic database of the Japanese pufferfish (Takifugu rubripes) and identified zPTH1 and zPTH2 homologs. Phylogenetic analysis showed that PTHs from zebrafish and pufferfish are more closely related to each other than to known mammalian PTH homologs or to PTHrP and tuberoinfundibular peptide of 39 residues. This is consistent with evolution of two teleost PTH-like peptides occurring after the evolutionary divergence between fishes and mammals. Overall, the PTH system appears more complex in fishes than in mammals, providing evidence of continued evolution in nontetrapod species. The availability of multiple forms of fish PTH and their receptors provide additional tools for PTH ligand/receptor structure-function studies.


Subject(s)
Parathyroid Hormone/genetics , Parathyroid Hormone/metabolism , Peptide Fragments/genetics , Peptide Fragments/metabolism , Zebrafish/genetics , Zebrafish/metabolism , Amino Acid Sequence , Animals , DNA, Complementary , Evolution, Molecular , Fishes/genetics , Genome , Humans , Molecular Sequence Data , Phylogeny , Receptor, Parathyroid Hormone, Type 1/metabolism , Receptors, Parathyroid Hormone/metabolism
15.
Mol Endocrinol ; 17(12): 2647-58, 2003 Dec.
Article in English | MEDLINE | ID: mdl-12947048

ABSTRACT

Recent functional studies have suggested that position 19 in PTH interacts with the portion of the PTH-1 receptor (P1R) that contains the extracellular loops and seven transmembrance helices (TMs) (the J domain). We tested this hypothesis using the photoaffinity cross-linking approach. A PTHrP(1-36) analog and a conformationally constrained PTH(1-21) analog, each containing para-benzoyl-l-phenylalanine (Bpa) at position 19, each cross-linked efficiently to the P1R expressed in COS-7 cells, and digestive mapping analysis localized the cross-linked site to the interval (Leu232-Lys240) at the extracellular end of TM2. Point mutation analysis identified Ala234, Val235, and Lys240 as determinants of cross-linking efficiency, and the Lys240-->Ala mutation selectively impaired the binding of PTH(1-21) and PTH(1-19) analogs, relative to that of PTH(1-15) analogs. The findings support the hypothesis that residue 19 of the receptor-bound ligand contacts, or is close to, the P1R J domain-specifically, Lys240 at the extracellular end of TM2. The findings also support a molecular model in which the 1-21 region of PTH binds to the extracellular face of the P1R J domain as an alpha-helix.


Subject(s)
Parathyroid Hormone-Related Protein/chemistry , Parathyroid Hormone/chemistry , Receptors, Parathyroid Hormone/chemistry , Amino Acid Sequence , Amino Acid Substitution , Animals , Binding Sites , COS Cells , Chlorocebus aethiops , Humans , Models, Molecular , Molecular Sequence Data , Mutagenesis, Site-Directed , Peptide Fragments/chemistry , Protein Conformation , Rats , Receptor, Parathyroid Hormone, Type 1 , Receptors, Parathyroid Hormone/genetics , Receptors, Parathyroid Hormone/metabolism , Transfection
16.
Nat Genet ; 30(3): 306-10, 2002 Mar.
Article in English | MEDLINE | ID: mdl-11850620

ABSTRACT

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.


Subject(s)
Bone Neoplasms/physiopathology , Enchondromatosis/physiopathology , Mutation , Receptors, Parathyroid Hormone/physiology , Animals , Bone Neoplasms/genetics , COS Cells , Enchondromatosis/genetics , Hedgehog Proteins , Humans , Mice , Mice, Transgenic , Mutagenesis, Site-Directed , Receptor, Parathyroid Hormone, Type 1 , Receptors, Parathyroid Hormone/genetics , Receptors, Parathyroid Hormone/metabolism , Second Messenger Systems , Signal Transduction , Trans-Activators/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...