Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 10 de 10
Filter
Add more filters










Publication year range
1.
Clin Chem ; 53(8): 1470-6, 2007 Aug.
Article in English | MEDLINE | ID: mdl-17599957

ABSTRACT

BACKGROUND: A new parathyroid hormone (PTH) species, the N-terminal PTH form (N-PTH), is distinct from intact human PTH of 84 amino acid residues [hPTH(1-84)] and is recognized in a 3rd-generation assay of "whole" PTH (wPTH; the 1-2 epitope) but not in a 2nd-generation assay of "total" PTH (tPTH; the 12-18 epitope). N-PTH usually represents <15% of wPTH but can be overproduced in severe primary hyperparathyroidism (PHPT) and secondary hyperparathyroidism. We investigated whether N-PTH is also overproduced in parathyroid cancer and whether N-PTH concentration is influenced by calcimimetic therapy. METHODS: We studied 8 patients with parathyroid carcinoma before and at week 16 of cinacalcet therapy, 6 patients with PHPT, and 6 control individuals. We fractionated sera with HPLC and analyzed fractions with the 2 assays to quantify hPTH(1-84), N-PTH, and non-(1-84) PTH fragments. RESULTS: Half of parathyroid carcinoma patients had an increased wPTH:tPTH ratio [mean (SD), 1.35 (0.29)]; the others had a typical ratio [0.72 (0.12)]. HPLC fractionation of sera from 2 high-ratio patients confirmed N-PTH overproduction [65% (12%) of wPTH]. The N-PTH fraction was <15% of wPTH in PHPT and healthy individuals. Calcimimetic therapy appreciably reduced calcium concentrations in parathyroid carcinoma patients but had little influence on PTH concentration, the wPTH:tPTH ratio, or the PTH HPLC profile. CONCLUSION: N-PTH is overproduced in some parathyroid cancer patients, but calcimimetic therapy does not influence its production. The clinical implications of this finding in parathyroid carcinoma await additional studies with an emphasis on N-PTH's biological activity and with larger numbers of patients.


Subject(s)
Biomarkers, Tumor/biosynthesis , Parathyroid Hormone/biosynthesis , Parathyroid Neoplasms/metabolism , Peptide Fragments/biosynthesis , Adult , Aged , Chromatography, High Pressure Liquid , Cinacalcet , Female , Humans , Hyperparathyroidism, Primary/drug therapy , Hyperparathyroidism, Primary/metabolism , Male , Middle Aged , Naphthalenes/therapeutic use , Parathyroid Hormone/blood , Parathyroid Neoplasms/drug therapy , Peptide Fragments/blood
3.
J Clin Endocrinol Metab ; 91(1): 283-9, 2006 Jan.
Article in English | MEDLINE | ID: mdl-16219713

ABSTRACT

CONTEXT: The quantitative evaluation of circulating PTH peaks revealed by PTH assays after HPLC separation constitutes the best way to study the behavior of PTH molecular forms, but it is also impractical. OBJECTIVE: The objective of the study was to investigate the regulation of circulating PTH molecular forms by calcium through the use of PTH fragments/PTH (1-84) ratios derived from PTH assays with different specificities before and after HPLC separation of circulating PTH. DESIGN: CaCl2 and Na citrate were infused in eight volunteers. PTH was measured in serum and HPLC fractions at different calcium concentrations in PTH assays reacting with regions 1-2 (CA), 12-18 (T), and 65-69 (C) of the PTH structure. RESULTS: From hypo- to hypercalcemia, the C/CA ratio had the highest range (1.92 to 9.75; P < 0.001), and the C/T ratio had a higher range (1.69 to 6.11; P < 0.01) than the T/CA ratio (1.15 to 1.86). Human (h) PTH (1-84) represented 32.7 and 4.3% of circulating PTH in hypo- and hypercalcemic HPLC profiles, respectively. These numbers were 5 and 0.9% for amino-terminal (N)-PTH, an amino-terminal form of PTH distinct from hPTH (1-84), 7.3 and 6.8% for non-(1-84) PTH or large C-PTH fragments with a partially preserved N structure, and 54.9 and 88.1% for C-PTH fragments missing a N structure. The HPLC C-PTH fragments to hPTH (1-84) ratio had the most extensive range (1.67 to 20.58). Despite their quantitative differences, all ratios identified identical behavior of PTH fragments relative to PTH (1-84). CONCLUSIONS: PTH assay ratios are an adequate tool to investigate the modulation of PTH molecular forms, even if all PTH assays show some undesirable cross-reactivity with certain circulating forms of PTH.


Subject(s)
Calcium/physiology , Parathyroid Hormone/metabolism , Adult , Algorithms , Calcium/blood , Calcium/pharmacology , Calcium Chloride/pharmacology , Chromatography, High Pressure Liquid , Citrates/pharmacology , Female , Humans , Hypercalcemia/blood , Hypocalcemia/blood , Male , Parathyroid Glands/physiology , Parathyroid Hormone/chemistry , Peptide Fragments/blood , Peptide Fragments/metabolism , Sodium Citrate
4.
Clin J Am Soc Nephrol ; 1(3): 525-31, 2006 May.
Article in English | MEDLINE | ID: mdl-17699255

ABSTRACT

Measurement of bioactive parathyroid hormone (PTH) is essential for optimal management of bone abnormalities in dialysis patients. This can be accomplished by PTH measurements using third-generation PTH assays, which detect more or less of the first six amino acids of the PTH structure. Such assays do not detect non-(1-84) PTH fragments, such as human PTH (7-84), which are recognized by the second-generation PTH assays that use a detection antibody that recognizes an epitope within the 13-34 region of the PTH structure. Therefore, third-generation PTH results are expected to be lower than those that are obtained with second-generation PTH assays. Rare exceptions to this rule have been reported for patients with severe primary hyperparathyroidism or parathyroid cancer. Sera and gland extracts were analyzed from a dialysis patient with high bone turnover disease and with surprising higher PTH levels by a third-generation assay than by a second-generation assay. This finding normalized after the surgical removal of an enlarged gland with a single nodule, an advanced type of nodular hyperplasia. HPLC fractionation of sera and gland extracts revealed the overproduction and secretion of a PTH molecule with an intact amino-terminus structure distinct from (1-84) PTH. This form of PTH was readily detectable by third-generation PTH assays but was poorly reactive in second-generation PTH assays. Therefore, parathyroid glands with advanced uremic nodular hyperplasia may overproduce and secrete a novel, biologically active form of PTH with an intact 1-6 region but a presumably modified 12-18 region required for the detection in second-generation PTH assays.


Subject(s)
Parathyroid Glands/metabolism , Parathyroid Glands/pathology , Parathyroid Hormone/biosynthesis , Parathyroid Hormone/metabolism , Uremia/metabolism , Female , Humans , Hyperplasia , Middle Aged , Severity of Illness Index
5.
Kidney Int ; 68(3): 998-1007, 2005 Sep.
Article in English | MEDLINE | ID: mdl-16105030

ABSTRACT

BACKGROUND: Non-(1-84) parathyroid hormone (PTH) fragments are large circulating carboxyl-terminal (C) fragments with a partially preserved amino-terminal (N) structure. hPTH (7-84), a synthetic surrogate, has been demonstrated to exert biologic effects in vivo and in vitro which are opposite to those of hPTH (1-34) on the PTH/PTHrP type I receptor through a C-PTH receptor. We wanted to determine the N structure of non-(1-84) PTH fragments. METHODS: Parathyroid cells isolated from glands obtained at surgery from three patients with primary hyperparathyroidism and three patients with secondary hyperparathyroidism were incubated with 35S-methionine to internally label their secretion products. Incubations were performed for 8 hours at the patient-ionized calcium concentration and in the presence of various protease inhibitors. The supernatant was fractionated by high-performance liquid chromatography (HPLC) and fractions were analyzed with PTH assays having (1 to 4) and (12 to 23) epitopes, respectively. The serum of each patient was similarly analyzed. Peaks of immunoreactivity identified were submitted to sequence analysis to recover the 35S-methionine residues in positions 8 and 18. RESULTS: Three regions of interest were identified with PTH assays. They corresponded to non-(1-84) PTH fragments (further divided in regions 3 and 4), a peak of N-PTH migrating in front of hPTH (1-84) (region 2) and a peak of immunoreactivity corresponding to the elution position of hPTH (1-84) (region 1). The last corresponded to a single sequence starting at position 1. Region 2 gave similar results in all cases (a major signal starting at position 1) but also sometimes minor sequences starting at position 4 or 7. Regions 3 and 4 always identified a major sequence starting at positions 7 and minor sequences starting at positions 8, 10, and 15. Surprisingly, a major signal starting at position 1 was also present in region 3. The HPLC profile obtained from a given patient's parathyroid cells was qualitatively similar to the one obtained with his/her serum in each case. CONCLUSION: These results indicate that non-(1-84) PTH fragments are composed of a family of fragments which may be generated by specific or progressive cleavage at the N region. The longest fragment starts at position 4 and the shortest at position 15. A peptide starting at position 7 appears as the major component of non-(1-84) PTH fragments. The generation process is similar to the one described for smaller C-PTH fragments a number of years ago, suggesting a similar production mechanism and source for all C-PTH fragments.


Subject(s)
Hyperparathyroidism, Primary/metabolism , Hyperparathyroidism, Secondary/metabolism , Parathyroid Glands/metabolism , Parathyroid Hormone/metabolism , Peptide Fragments/metabolism , Amino Acid Sequence , Chromatography, High Pressure Liquid , Female , Humans , Male , Molecular Sequence Data , Parathyroid Hormone/chemistry , Peptide Fragments/chemistry
6.
Curr Opin Nephrol Hypertens ; 14(4): 330-6, 2005 Jul.
Article in English | MEDLINE | ID: mdl-15931000

ABSTRACT

PURPOSE OF REVIEW: Carboxyl-terminal parathyroid hormone (C-PTH) fragments constitute 80% of circulating PTH. Since the first 34 amino acids of the PTH structure are sufficient to explain PTH classical biological effects on the type I PTH/PTHrP receptor and since C-PTH fragments do not bind to this receptor, they have long been considered inactive. Recent data suggest the existence of a C-PTH receptor through which C-PTH fragments exert biological effects opposite to those of human PTH(1-84) on the type I PTH/PTHrP receptor. This is why a lot of attention has been paid to these fragments recently. RECENT FINDINGS: In vivo, synthetic C-PTH fragments are able to decrease calcium concentration, to antagonize the calcemic response to human PTH(1-34) and human PTH(1-84) and to decrease the high bone turnover rate induced by human PTH(1-84). In vitro, they inhibit bone resorption, promote osteocyte apoptosis and exert a variety of effects on bone and cartilaginous cells. These effects are opposite to those of human PTH(1-84) on the PTH/PTHrP type I receptor. This suggests that the molecular forms of circulating PTH may control bone participation in calcium homeostasis via two different receptors. Clinically, the accumulation of C-PTH fragments in renal failure patients may cause PTH resistance and may be associated with adynamic bone disease. Rare parathyroid tumors, without a set point error, overproduce C-PTH fragments. The implication of C-PTH fragments in osteoporosis is still to be explored. SUMMARY: C-PTH fragments represent a new field of investigation in PTH biology. More studies are necessary to disclose their real importance in calcium and bone homeostasis in health and disease.


Subject(s)
Hyperparathyroidism, Secondary/blood , Hyperparathyroidism/blood , Osteoporosis/blood , Parathyroid Hormone/blood , Peptide Fragments/blood , Animals , Humans
7.
Clin Endocrinol (Oxf) ; 62(6): 721-7, 2005 Jun.
Article in English | MEDLINE | ID: mdl-15943835

ABSTRACT

OBJECTIVE: Rare patients with severe primary hyperparathyroidism present with large parathyroid tumours, severe hypercalcaemia, very high PTH levels and osteitis fibrosa cystica. Some of these patients display a large amount of C-PTH fragments in circulation and present with a higher C-PTH/I-PTH ratio than seen in less severe cases of primary hyperparathyroidism. We wanted to determine how PTH levels and circulating PTH high-performance liquid chromatography (HPLC) profiles analysed with PTH assays having different epitopes could be affected by medical and surgical treatment in such patients. DESIGN: A 55-year-old man with severe hypercalcaemia (Ca(2+): 2.01 mmol/l), very high PTH levels (CA-PTH 82.1 and T-PTH 72 pmol/l) caused by a large parathyroid tumour (7.35 g) and accompanied by significant bone involvement (alkaline phosphatase of 185 UI/l and subperiostal bone resorption of hands) was referred to us. Blood was obtained at various time points during his medical treatment, before and after surgery, to measure parameters of calcium and phosphorus metabolism, and of bone turnover. HPLC separations of circulating PTH molecular forms were performed and analysed with PTH assays having 1-4 (CA), 12-18 (T), 26-32 (E) and 65-84 (C) epitopes. RESULTS: Before surgery, serum Ca2+ was nearly normalized with hydratation, intravenous (IV) pamidronate and oral vitamin D administration. Despite a decrease in Ca2+ to 1.31 mmol/l, CA-PTH and T-PTH levels decreased by half in relation to a threefold increase in basal 1,25-dihydroxyvitamin D [1,25(OH)2D] level (94 to 337 pmol/l). After this initial positive response, hypercalcaemia and elevated CA- and T-PTH levels recurred even if 1,25(OH)2D levels remained elevated. The tumour was removed surgically and proved to be poorly differentiated with nuclear atypia and mitosis. After surgery, the Ca2+ level and PTH secretion normalized. The higher CA-PTH level relative to the T-PTH level observed before surgery in this patient was related to the oversecretion of an amino-terminal (N) form of PTH recognized by PTH assays with (1-4) or (26-32) epitopes but not by the T-PTH assay with a (12-18) epitope. This molecular form represented 50% of CA-PTH measured in this patient, but only 7% in less severe cases of primary hyperparathyroidism. It was unaffected by medical therapy and disappeared after surgery. CONCLUSION: The relationship between the overexpression of this N-PTH molecular form and severe primary hyperparathyroidism remains unclear. Further studies will be required in these rare patients to see whether N-PTH is a marker of less well differentiated parathyroid tumours and/or relates to the overproduction of C-PTH fragments in the presence of severe hypercalcaemia.


Subject(s)
Adenoma/blood , Hyperparathyroidism/blood , Mediastinal Neoplasms/blood , Parathyroid Hormone/blood , Adenoma/drug therapy , Adenoma/surgery , Antineoplastic Agents/therapeutic use , Calcium/blood , Chromatography, High Pressure Liquid/methods , Diphosphonates/therapeutic use , Fluid Therapy , Humans , Hyperparathyroidism/drug therapy , Hyperparathyroidism/surgery , Male , Mediastinal Neoplasms/drug therapy , Mediastinal Neoplasms/surgery , Middle Aged , Pamidronate , Parathyroid Hormone/chemistry , Vitamin D/therapeutic use
8.
Clin Chem ; 51(1): 169-76, 2005 Jan.
Article in English | MEDLINE | ID: mdl-15514090

ABSTRACT

BACKGROUND: Non-(1-84) parathyroid hormone (PTH) fragments are large C-terminal fragments of PTH with a partially preserved N-terminal structure. They differ from other C-terminal PTH fragments, which do not have an N-terminal structure and do not react in intact PTH assays. We aimed to identify the minimal N-terminal structure common to all non-(1-84) PTH fragments. METHODS: Sera obtained from six healthy individuals and six patients with primary hyperparathyroidism, and six serum pools from dialysis patients with different PTH concentrations were fractionated by HPLC and analyzed by four different PTH assays. Each assay was characterized by saturation analysis of its detection antibody and capacity to react with different PTH fragments. Human PTH(1-84) [hPTH(1-84)] calibrators were normalized to an in-house hPTH(1-84) calibrator. RESULTS: The cyclase-activating PTH (CA-PTH) assay had an early (1, 2,) epitope and reacted only with hPTH(1-84). The other assays had epitopes in region (13-34). Total and intact PTH assays had epitopes proximal to position 18 and reacted equally well with hPTH(1-84) and hPTH(7-84), and the Elecsys PTH assay had an epitope distal to position 19, being saturable by hPTH(18-48) and also reacting with [Tyr(34)]hPTH(19-84). The HPLC profiles obtained with these assays showed that non-(1-84) PTH fragments did not react in the CA-PTH assay, as expected. The amount of non-(1-84) PTH detected by the other three assays was similar when the assay results were normalized to a common calibrator. CONCLUSIONS: The results suggest that the amount of non-(1-84) PTH detected by epitopes proximal or distal to position 19 of the PTH structure is identical, indicating a common minimum structure starting before position 19. This in turn points to a probable high-affinity interaction with the C-PTH receptor, as observed previously with [Tyr(34)]hPTH(19-84) in various cell lines and in mouse osteocytes with PTH/PTHrP type I receptor ablation.


Subject(s)
Parathyroid Hormone/chemistry , Peptide Fragments/chemistry , Calibration , Epitopes , Humans , Hyperparathyroidism/blood , Immunoassay , Parathyroid Hormone/blood , Parathyroid Hormone/immunology , Peptide Fragments/blood , Peptide Fragments/immunology
9.
Clin Chem ; 49(12): 2037-44, 2003 Dec.
Article in English | MEDLINE | ID: mdl-14633875

ABSTRACT

BACKGROUND: To separate non-(1-84)parathyroid hormone [non-(1-84)PTH] from PTH(1-84), we developed new HPLC gradients and observed that the peak coeluting with hPTH(1-84) could be separated into two entities recognized by a cyclase-activating PTH (CA-PTH) assay that reacts with the first four amino acids of the PTH structure. METHODS: Sera from six healthy individuals and five patients with primary hyperparathyroidism, and eight pools of sera from patients in renal failure were fractionated by HPLC. A total (T)-PTH assay reacting with the (15-20) region, the CA-PTH assay, and a COOH-terminal (C)-PTH assay with a (65-84) structure requirement were used to measure basal and fractionated PTH values. RESULTS: T-PTH was higher than CA-PTH in all healthy controls [mean (SD), 3.13 (0.37) vs 2.29 (0.33) pmol/L; P <0.01] and in renal failure patients [47 (35.1) vs 33.4 (26.1) pmol/L; P <0.01]. By contrast, CA-PTH concentrations were similar to or higher than T-PTH in three of five patients with primary hyperparathyroidism [25.7 (26.1) vs 23.1 (24.2) pmol/L; not significant]. The CA-PTH assay reacted with the hPTH(1-84) peak and with a minor peak different from the non-(1-84) peak recognized by the T-PTH assay. This minor peak was not recognized by the T-PTH assay. It represented 8 (2)% of CA-PTH in controls, 25 (23)% in patients with primary hyperparathyroidism, and 22 (7)% in renal failure patients, assuming equimolar reactivity to hPTH(1-84) in the CA-PTH assay. It was not oxidized hPTH(1-84), which migrated differently on HPLC and reacted similarly in the CA and T-PTH assays. CONCLUSIONS: This new molecular form of PTH has structural integrity of the (1-4) region but presumably is modified in the region (15-20), which is usually recognized by the T-PTH assay. Its clinical implications remain to be defined.


Subject(s)
Hyperparathyroidism/metabolism , Parathyroid Hormone/biosynthesis , Chromatography, High Pressure Liquid , Humans , Immunoassay/methods , Parathyroid Hormone/blood , Peptide Fragments/blood , Renal Insufficiency/metabolism
10.
Semin Dial ; 15(3): 196-201, 2002.
Article in English | MEDLINE | ID: mdl-12100458

ABSTRACT

Non-(1-84) parathyroid hormones (PTHs) are large circulating carboxyl-terminal PTH (C-PTH) fragments with a partially preserved amino-terminal structure. They were discovered during high-performance liquid chromatography (HPLC) analysis of circulating PTH molecular forms detected by an intact PTH (I-PTH) assay. Like other C-PTH fragments, they accumulate in blood in renal failure and account for up to 50% of I-PTH. They are secreted by the parathyroid glands in humans, and are generated by the peripheral metabolism of hPTH(1-84) in rats. The exact structure of non-(1-84)PTH fragments is not known. To study the possible role of non-(1-84) in PTH biology, hPTH(7-84) has been used as a surrogate, being the only large C fragment available on the market. In anesthetized, thyroparathyroidectomized rats, hPTH(7-84) caused hypocalcemia beyond that induced by surgery. It also blocked the calcemic response to hPTH(1-84) or hPTH(1-34). Other smaller C-PTH fragments, such as hPTH(39-84) and hPTH(53-84), were synergistic to hPTH(7-84) effects. hPTH(7-84) did not bind to the PTH/PTHrP receptor, but only to the C-PTH receptor in ROS 17/2.8 clonal cells, and did not stimulate cyclic adenosine monophosphate (cAMP) production by the same cells, suggesting that its hypocalcemic action was mediated via a receptor different from the PTH/PTHrP receptor, and that the calcium concentration resulted from the sum of the positive effect of hPTH(1-84) on the PTH/PTHrP receptor and of the negative effect of hPTH(7-84) and of C-PTH fragments on the C-PTH receptor. These data will change our understanding of circulating calcium regulation, which must now be viewed as the end result of opposite actions on two PTH receptors. PTH immunoheterogeneity, a highly regulated phenomenon, contributes to this dual biological effect, generating an agonist for the two different receptors. Clinically these results could have some implications in our knowledge of the PTH resistance of renal failure, of renal osteodystrophy, and of certain aspects of the uremic syndrome.


Subject(s)
Parathyroid Hormone/metabolism , Renal Insufficiency/metabolism , Animals , Calcium/blood , Humans , Parathyroid Hormone/chemistry , Rats , Receptors, Parathyroid Hormone/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...