Elsevier

Bone

Volume 43, Issue 6, December 2008, Pages 1022-1030
Bone

Differential effects of intermittent PTH(1–34) and PTH(7–34) on bone microarchitecture and aortic calcification in experimental renal failure

https://doi.org/10.1016/j.bone.2008.07.250Get rights and content

Abstract

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

Introduction

Disturbances of bone and mineral metabolism are well recognized features of chronic kidney disease (CKD) [1], [2]. Vascular calcification and ectopic calcification of other soft tissues are commonly observed. These actions on mineral metabolism in patients with CKD are accompanied by increased synthesis, secretion and circulating levels of parathyroid hormone (PTH) and PTH fragments, which lead to the development of secondary hyperparathyroidism (SHPT) [3], [4]. The factors involved in the pathogenesis of SHPT in CKD, include phosphate retention, low 1,25(OH)2D3 and vitamin D receptors, decreased serum free calcium and parathyroid calcium-sensing receptors [5], [6], downregulation of PTH receptors [7], [8], [9], [10], and the accumulation of uremic toxins and acidosis [9], [11] that contribute to skeletal resistance to the actions of PTH and a spectrum of bone diseases collectively called osteodystrophy. Although the mechanism of resistance is incompletely understood, uremic patients require PTH levels 2–3 times normal to maintain bone turnover [56]. Excessive suppression of PTH secretion, however, may lead to low bone turnover, so-called adynamic bone disease [12], [13].

The type I PTH receptor (PTH1R) is the predominant form expressed in bone and kidney and is a member of class B of the superfamily of G protein-coupled receptors (GPCRs) [14], [15]. It mediates the actions of PTH and of PTH fragments containing an intact amino terminus. In the kidney, the PTH1R mediates the regulation of PTH on the renal transport of phosphate and calcium [16]. In renal tubular epithelial cells, signal and expression of PTH1R appears to be modulated in a cell-specific manner. Mounting evidence supports the view that in renal failure serum PTH(7–84) accumulates to high levels [17], [18] that may approach those of PTH(1–84) [18], [19], [20]. It has been suggested that the competitive inhibition of PTH1R binding by amino-truncated PTH fragments contributes to the PTH resistance of uremia [21]. However, as indicated above, PTH1R downregulation has been described in the same pathological setting [7], [22], which is inconsistent with the view that PTH(7–34) acts exclusively as an antagonist.

Recent work shows that PTH(7–84) and its synthetic analog, PTH(7–34), internalize [23], [24] and downregulate [25] the PTH1R without antecedent or concomitant receptor activation, whereas PTH(1–34) promotes synchronized PTH1R activation and internalization. This phenomenon occurs in a cell-specific manner that depends on the expression of the scaffolding protein Na/H exchange regulatory factor (NHERF1) [23], [26], a cytoplasmic adaptor that interacts with the carboxy-terminus of the PTH1R and is implicated in protein targeting and in the assembly of protein complexes [26], [27]. These findings provide a plausible alternative explanation for PTH1R downregulation and resistance in renal failure. According to this hypothesis, PTH(7–84) may contribute to hormone resistance by inducing PTH1R internalization and downregulation without accompanying activation. Reducing the number of plasma membrane-delimited PTH receptors would diminish the action of full-length or amino-terminal PTH fragments.

Other studies addressed competitive interactions between amino-truncated PTH fragments and PTH(1–34) or PTH(1–84) [28], [29]. PTH(7–34) induces PTH1R internalization on its own and is only a competitive inhibitor at higher concentrations [23]. In the present study, we used exogenous PTH(1–34) and the amino-truncated peptide PTH(7–34) in animal models of CKD without secondary hyperparathyroidism to test the hypothesis that amino-truncated PTH fragments downregulate the PTH1R, thereby reducing bone demineralization and vascular calcification.

Section snippets

Animals and experimental protocol

All experiment protocols were approved by the University of Pittsburgh Institutional Animal Care and Use Committee (IACUC).

Nine-week old male Wistar rats weighing 250–300 g, were purchased from Charles River Laboratories. Animals were fed standard diet (LabDiet, PMI Nutrition LLC, MO, US) containing 1.0% calcium, 0.75% phosphorus and 22.5% protein. The animals were allowed free access to food and water and housed in individual cages at constant room temperature with a 12-h light and dark cycle.

Results

All animals gained weight from the time of entry in the protocol. Rats undergoing PTX gained as much weight as Sham animals. However, rats subjected to NPX or PTX/NPX gained significantly less weight (83% and 73% compared to Sham; P < 0.05, P < 0.01, respectively).

Discussion

The present studies were designed to analyze and compare the effects of intermittent PTH(1–34) or PTH(7‑34) treatment on bone microarchitecture, vascular calcification, and PTH1R expression in an animal model of experimental renal failure. It has been reported that the administration of PTH prevents osteoporotic fractures in postmenopausal women, improves fracture healing and implant fixation, and osteogenesis in model animals [37], [38], [39], [40]. We show here that in an animal model of

Acknowledgments

This work was supported by grant DK 54171 (PAF) and by a Minority Supplement (EMS) from the National Institutes of Health, an internal award from the Office of the Senior Vice Chancellor for the Health Sciences, University of Pittsburgh (EMS) and The Carl L. Nelson Chair of Orthopaedic Surgery (LJS). The authors thank Dr. Nathalie Taesch, who helped with vascular histology and staining.

References (63)

  • M. Cozzolino et al.

    Pathogenesis of vascular calcification in chronic kidney disease

    Kidney Int.

    (2005)
  • P. D'Amour et al.

    Structure of non-(1–84) PTH fragments secreted by parathyroid glands in primary and secondary hyperparathyroidism

    Kidney Int.

    (2005)
  • C.A. Frolik et al.

    Anabolic and catabolic bone effects of human parathyroid hormone (1–34) are predicted by duration of hormone exposure

    Bone

    (2003)
  • D.J. Rickard et al.

    Intermittent treatment with parathyroid hormone (PTH) as well as a non-peptide small molecule agonist of the PTH1 receptor inhibits adipocyte differentiation in human bone marrow stromal cells

    Bone

    (2006)
  • M.A. Miller et al.

    Disparate effects of mild, moderate, and severe secondary hyperparathyroidism on cancellous and cortical bone in rats with chronic renal insufficiency

    Bone

    (1998)
  • P. Magnusson et al.

    Effect of chronic renal failure on bone turnover and bone alkaline phosphatase isoforms

    Kidney Int.

    (2001)
  • M. Fukagawa et al.

    Skeletal resistance to PTH as a basic abnormality underlying uremic bone diseases

    Am. J. Kidney Dis.

    (2001)
  • P. Ureña et al.

    Parathyroidectomy does not prevent the renal PTH/PTHrP receptor down-regulation in uremic rats

    Kidney Int.

    (1995)
  • S. Disthabanchong et al.

    Regulation of PTH1 receptor expression by uremic ultrafiltrate in UMR 106-01 osteoblast-like cells

    Kidney Int.

    (2004)
  • W.G. Goodman et al.

    Vascular calcification in chronic kidney disease

    Am. J. Kidney Dis.

    (2004)
  • K. Lomashvili et al.

    Chemical and hormonal determinants of vascular calcification in vitro

    Kidney Int.

    (2006)
  • G.A. Block et al.

    Re-evaluation of risks associated with hyperphosphatemia and hyperparathyroidism in dialysis patients: recommendations for a change in management

    Am. J. Kidney Dis.

    (2000)
  • K.J. Martin et al.

    Metabolic bone disease in chronic kidney disease

    J. Am. Soc. Nephrol.

    (2007)
  • M. Fukagawa et al.

    Regulation of parathyroid function in chronic kidney disease (CKD)

    Clin. Exp. Nephrol.

    (2006)
  • M. Cozzolino et al.

    Pathogenesis of parathyroid hyperplasia in renal failure

    J. Nephrol.

    (2005)
  • J. Chen et al.

    Down-regulation of PTH/PTHrP receptor in the kidney of patients with renal impairment

    Chin. Med. J.

    (1998)
  • R.M. Edwards et al.

    Parathyroid hormone-1 receptor down-regulation in kidneys from rats with chronic renal failure

    Pharmacology

    (2001)
  • T.B. Drüeke

    Abnormal skeletal response to parathyroid hormone and the expression of its receptor in chronic uremia

    Pediatr. Nephrol.

    (1996)
  • P. Urena

    Use of calcimimetics in uremic patients with secondary hyperparathyroidism: review

    Artif. Organs

    (2003)
  • A.J. Brown et al.

    Drug Insight: vitamin D analogs in the treatment of secondary hyperparathyroidism in patients with chronic kidney disease

    Nat. Clin. Pract. Endocrinol. Metab.

    (2007)
  • A.J. Harmar

    Family-B G-protein-coupled receptors

    Genome Biology

    (2001)
  • Cited by (50)

    • Postmenopausal osteoporosis coexisting with other metabolic diseases: Treatment considerations

      2021, Maturitas
      Citation Excerpt :

      In this context, it has been hypothesized that anti-osteoporotic medications could exert beneficial effects on the progression of atherosclerosis. Bisphosphonates and denosumab were shown to inhibit arterial calcifications in animal models [79,80], while teriparatide did not affect preexisting cardiovascular calcifications [81]. Preclinical studies with anti-sclerostin antibodies [82] did not report a greater risk of vascular calcifications, despite the fact that Wnt signaling is implicated in cardiovascular remodeling and sclerostin is expressed in bone and vascular smooth muscle cells of aorta [82] and atherosclerotic plaques [83].

    • Translating in vitro ligand bias into in vivo efficacy

      2018, Cellular Signalling
      Citation Excerpt :

      How can one account for the paradoxical response to [D-Trp12,Tyr34]-bPTH(7-34) in vivo if the effects of PTH on both bone formation and resorption are solely dependent upon activation of Gs-cAMP signaling? The neutral PTH1R antagonist PTH(7-34) has no anabolic activity in a renal failure model of rats subjected to prior parathyroidectomy [70], raising the question of whether endogenous PTH is necessary for a PTH antagonist to stimulate bone formation. With intact parathyroid glands, intermittent exposure to a PTH1R antagonist might produce periodic ‘dips’ or even rebound ‘spikes’ in endogenous PTH action that might stimulate bone formation in a manner analogous to a conventional ligand, i.e. by producing cyclic modulation of PTH1R-Gs signaling.

    • Bone loss and vascular calcification: A bi-directional interplay?

      2016, Vascular Pharmacology
      Citation Excerpt :

      PTH and 1,25-dihydroxyvitamin D are essential key factors to regulate circulating calcium, phosphate levels, and matrix mineralization. The role of PTH is complex, because although high PTH levels are generally associated to aortic stiffness and VC, the PTH 1–34 fragment may inhibit calcification [17–20]. However, experimental data in a model of rat uricemia showed that hyperphosphataemia and high PTH, in isolation or in conjunction, plays a role in the pathogenesis of VC, because they are both associated with overexpression of Runx2 [21].

    View all citing articles on Scopus
    View full text