Journal Information
Vol. 31. Issue. 5.September 2011
Pages 0-626
Vol. 31. Issue. 5.September 2011
Pages 0-626
Full text access
Chronic kidney disease-mineral and bone disorder: a complex scenario
El complejo escenario de las alteraciones de metabolismo óseo y mineral en la enfermedad renal crónica
Visits
12813
Natalia Mejiaa, N.. Mejíab, Pablo Roman-Garciac, P.. Roman-Garcíad, Ana Belen Miare, A.B.. Miarf, Beatriz Tavirag, B.. Tavirah, Jorge B Cannata-Andíac, J.B.. Cannata-Andíad
a Servicio de Nefrología Pediátrica, Hospital Universitario Central de Asturias, Oviedo, Asturias, Spain,
b Servicio de Nefrología Pediátrica, Hospital Universitario Central de Asturias, Oviedo, Asturias,
c Servicio de Metabolismo Óseo y Mineral, Hospital Universitario Central de Asturias. Instituto Reina Sofía de Investigación. REDinREN del ISCIII. Universidad de Oviedo, Oviedo, Asturias, Spain,
d Servicio de Metabolismo Óseo y Mineral, Hospital Universitario Central de Asturias. Instituto Reina Sofía de Investigación. REDinREN del ISCIII. Universidad de Oviedo, Oviedo, Asturias,
e Departamente de Biología Funcional y Celular, Universidad de Oviedo, Oviedo, Asturias, Spain,
f Departamente de Biología Funcional y Celular, Universidad de Oviedo, Oviedo, Asturias,
g Laboratorio de Genética Molecular, Hospital Universitario Central de Asturias. (REDinREN, Red de investigación renal del Instituto de Salud Carlos III, Fondos FEDER), Oviedo, Asturias, Spain,
h Laboratorio de Genética Molecular, Hospital Universitario Central de Asturias. (REDinREN, Red de investigación renal del Instituto de Salud Carlos III, Fondos FEDER), Oviedo, Asturias,
This item has received
Article information
Abstract
Full Text
Bibliography
Download PDF
Statistics

Las alteraciones del metabolismo óseo en el escenario de la enfermedad renal crónica (CKD-MBD) constituyen un dinámico campo de estudio. Al conjunto de reguladores clásicos del metabolismo óseo tales como calcio, fósforo, hormona paratiroidea (PTH) y calcitriol se ha añadido el factor de crecimiento fibroblástico 23 (FGF-23). La calcificación vascular, una de las complicaciones más importantes de la enfermedad renal crónica, está sujeta a una compleja regulación en la que intervienen factores promotores e inhibidores del proceso de mineralización. La asociación entre calcificación vascular, desmineralización ósea y mortalidad y la existencia de factores y vías de señalización comunes está siendo objeto de interesantes investigaciones.

           

Palabras clave:
Hiperparatiroidismo secundario
Palabras clave:
CKD-MBD
Palabras clave:
Calcificación vascular
Palabras clave:
FGF-23
Palabras clave:
Desmineralización ósea

The chronic kidney disease-bone and mineral disorders (CKD-MBD) represents a dinamic area of research. Recently, new factors such as FGF-23 have been added to the classic list of regulators of bone metabolism, which include calcium, phosphorus, PTH and calcitriol. Vascular calcification, one of the most important complication of CKD-MBD is regulated by a complex variety of  promoters and inhibitors. The relationship between vascular calcification, bone loss and mortality, together with the existence of likely common signaling pathways are subject of interesting investigations.

Keywords:
Secondary hyperparathyroidism
Keywords:
CKD-MBD
Keywords:
Vascular calcification
Keywords:
FGF-23
Keywords:
Bone demineralization
Full Text

INTRODUCTION

In healthy individuals kidneys regulate calcium and phosphorus homeostasis through tubular reabsorption mechanisms. Patients with chronic kidney disease have seriously compromised homeostatic mechanisms, giving rise to different adaptive changes in calcium (Ca), phosphorus (P), parathyroid hormone (PTH), vitamin D and fibroblastic growth factor (FGF-23) levels.

Among all the CKD disorders, those related to bone and mineral metabolism have a significant impact on morbidity and mortality. Traditionally, these abnormalities are included within the term ‘renal osteodystrophy’.1 Recently, the collection of biochemical abnormalities such as calcium, phosphorus, vitamin D and PTH disorders, changes in bone morphology such as variation in turnover, volume and bone mineralisation, and vascular or other soft-tissue calcifications, have been included under the definition of “chronic kidney disease mineral and bone disorders” (CKD-MBD).2-4 There are various clinical signs, although secondary hyperparathyroidism (SHPT), fractures, bone pain, vascular calcification and cardiovascular events are highlighted as causing lower quality of life with a high morbidity and mortality.5

This review pays special attention to the Ca-P-PTH-vitamin D-FGF-23 axis, SHPT, vascular calcifications and their relationship with bone-mineral density (BMD) in CKD-MBD.

 

CA-P-PTH-VITAMIN D-FGF-23 AXIS

The most important factors that regulate bone and mineral metabolism are PTH, Ca, P, FGF-23 and the vitamin D related compounds. These factors are also totally interrelated and their effects depend on the target organ studied. The progression of CKD is associated with an early increase in FGF-23 and with a reduced functioning kidney mass; both factors favour the decrease in alpha-1-hydroxylase, the enzyme responsible for calcitriol synthesis, a physiologically active form of vitamin D. The final result is that this hormone’s levels are found frecuently decreased.6 The decrease in calcitriol significantly affects the intestinal Ca absorption, favouring the serum calcium to decrease and stimulating PTH.7 In the bone, PTH stimulates Ca and P release, whilst in the kidney, it stimulates Ca reabsorption and inhibits phosphate reabsorption. Furthermore, PTH increases alpha-1-hydroxylase expression, therefore favouring calcitriol synthesis which increases intestinal Ca and P absorption.8 As a result of these changes, serum Ca increases and P decreases.9

Reduced renal function also directly affects P reabsorption. The kidney is not capable of filtering enough P, and its high level in blood directly stimulates the parathyroid gland which, in turn, stimulates FGF-23 synthesis and secretion by the osteocytes.10-13 In principle, FGF-23 was described to inhibit renal P reabsorption and calcitriol production; these effects are synergic and contrary to those of PTH, respectively.14 However, previous studies have shown that FGF-23 not only has an effect on renal tissue but also on the parathyroid gland, which under normal conditions activates the mitogen-activated protein kinase (MAPK) pathway, reducing PTH synthesis and secretion.15 To do so, it needs to bind to its FGFR-1 and 3 receptors, and its co-receptor Klotho. The Klotho gene codes for a transmembrane protein whose deficiency in rats results in a premature ageing-like phenotype, characterised by atherosclerosis, osteoporosis, hyperphospatemia, calcifications in the vascular medial layer, cardiac muscle and other tissues.16 Klotho expression determines the tissue-specificity of FGF-23 and is increased by FGF-23 in a feedback mechanism.

 

SECONDARY HYPERPARATHYROIDISM

The parathyroid gland is the main organ responsible for Ca homeostasis in the organism, it senses serum Ca concentration via the Ca receptor (CaR), and also has vitamin D receptors (VDR). Its main action is PTH production.17 Extracellular ionic Ca is the main parathyroid regulator; low levels stimulate PTH secretion in a matter of minutes, whilst high levels inhibit hormone release and, furthermore, favour its degradation within the parathyroid cells.18-20 The result is a sigmoidal response in the parathyroid gland, in which small extracellular ionic Ca changes provoke large variations in PTH, achieving maximum inhibition in hypercalcaemia. The effects of Ca on PTH are mediated by its specific receptor, CaR,21 a receptor belonging to the family of G protein-coupled receptors, which are found at the cell membrane.

Calcitriol acts on the parathyroid gland via its specific receptor, VDR, a high affinity and specificity receptor that belongs to the family of steroid/thyroid receptors.22 When calcitriol is bound to a receptor, the calcitriol/VDR complex is translocated to the cell nucleus, forming a heterodimer with retinoid-X receptor (RXR). The calcitriol/VDR/RXR complex is bound to vitamin D responsive elements (VDR-E) present in the PTH gene’s promoter region, blocking its transcription. Furthermore, calcitriol is able to indirectly inhibit PTH secretion, increasing intestinal Ca absorption, and at the same time, stimulating the reabsorption of Ca deposits.23-25

In CKD, the incorrect control of PTH secretion has partly been attributed to the reduced VDR and CaR expression which occurs in parallel to parathyroid gland growth. Parathyroid gland hyperplasia and the consequent increase in PTH secretion are responsible for SHPT observed in CKD. In mild and moderate forms of SHPT, the parathyroid gland is still able to respond to its main regulators, such as Ca, P and FGF-23.26,27 On the other hand, in more advanced stages, especially in tertiary HPT, an irreversible form of HPT which is common in patients undergoing dialysis for a long time and kidney transplant patients, the parathyroid gland has a scarce or null response to usual stimuli and presents a high degree of autonomy.28

FGF-23/Klotho and Ca/CaR act on the parathyroid gland via mechanisms similar to the MAPK pathway,15 with the common objective of reducing PTH synthesis and secretion. Under normal conditions, FGF-23 reduces PTH levels, although in chronic renal failure FGF-23 and PTH levels increase. This is due to a reduction in Klotho/FGFR in uremic parathyroid glands so that FGF-23 fails to exert its inhibitory effect likely due to a decrease in MAPK activation.29

 

VASCULAR CALCIFICATIONS

Soft-tissue calcification occurs in a high percentage of CKD patients and it has been reported since the 19th century. Tissues that are frequently affected are blood vessels, lungs, kidneys, myocardium, coronary arteries, the central nervous system and gastric mucosa. Calcifications are associated with many factors such as excessive doses of calcitriol, hyperphosphatemia, tobacco use, arterial hypertension, increase in CaxP product, calcium overload, diabetes and male gender.30 In CKD, lesions are mainly observed in the medial layer, but also in the intima layer and may affect the flow and vascular stiffness, with the consequent increase in arterial pressure and pulse wave velocity.

Vascular calcifications in the artery intima layer are often associated with previous atherosclerotic plaques. They affect the medial layer of medium calibre arteries, the aorta, and coronary arteries, with concentric Ca precipitates in vascular smooth muscle cells, causing stiffness and atherosclerosis. A phenotypic differentiation from vascular to bone cells is produced in these calcifications, critically reducing the contractile ability of muscle cells. Vascular complications often precede bone alterations, which occur very late and insidiously.31 Although all histological forms of renal osteodystrophy have been associated with a greater prevalence of vascular calcifications, the one with the most impact is that observed in low turnover renal osteodystrophy.

There is a large number of promoters and inhibitors involved in vascular calcification. Promoters favour vessel calcification but, under normal conditions, there are more vascular calcification inhibitors circulating in the blood. With older age and CKD, this process is reversed and calcification inhibitors are down-regulated, while promoters are up-regulated.

Phosphorus is the most significant and probably the most studied vascular calcification promoter and its action is facilitated by the Na/P cotransporter, called PiT-1. Ca, osteopontin, osteocalcin, bone morphogenetic proteins (BMP-2 and BMP4), bone sialoprotein, type I collagen, and alkaline phosphatase (ALP) and many transcription factors (CbfaI/RUNX2 and MSX-2) have been described as downsctream vascular calcification promoters.32-37 Several types of proteins are vascular calcification inhibitors. Fetuin-A is a circulating molecule that inhibits the formation of hydroxyapatite crystals, and there is a correlation between decreased fetuin-A levels and an increased mortality in patients on haemodialysis and patients with heart diseases.38 Osteoprotegerin (OPG) also participates in inhibiting vascular calcification. Studies on KO mice for OPG showed calcifications in the aorta, renal arteries and osteoporosis.39 Studies on KO mice for bone Gla protein (BGP) found calcification in the medial layer and the cartilage.40 Among the bone morphogenetic proteins (BMP), BMP-7 is associated with vascular calcification inhibition. In KO mice studies for low density lipoprotein (LDL), it was shown that the expression of vascular osteocalcin is down-regulated in animals treated with BMP-7.41

 

RELATIONSHIP BETWEEN DEMINERALISATION AND VASCULAR CALCIFICATION

Although this relationship was first described 20 years ago,42 the relationship between osteoporosis and vascular calcification has been underestimated, possibly because demineralisation, osteoporosis and vascular calcification have always been considered as disorders secondary to ageing. Age can not be excluded as a favouring factor, but evidence of a frequent association between bone fragility and vascular calcification further suggests that there may be a causal relationship between the two. The pathogenic factors involved in both processes are still to a large extent unknown.43

 

KEY CONCEPTS

1.            FGF-23 has been added to the list of bone metabolism regulators in chronic kidney disease, given its effects on the kidney, parathyroid gland and vitamin D metabolism.  

2.            Vascular calcification, an important chronic kidney disease complication, is subject to a complex regulation in which mineralisation promoters and inhibitors intervene.  

3.            Epidemiological, clinical and experimental studies have shown a relationship between vascular calcification progres­sion and bone demineralisation, suggesting that between both processes there is an interrelation, involving com­mon mediators, which goes beyond the known effect that age has on both alterations.

 

Bibliography
[1]
Torregrosa JV, Bover Sanjuán J, Cannata Andía J. Recomendaciones de la Sociedad Española de Nefrologia para el manejo de las alteraciones del metabolismo óseo-mineral en los pacientes con enfermedad renal crónica. Nefrologia 2011;31:3-32. [Pubmed]
[2]
Moe S, Drueke T, Cunningham J, Goodman W, Martin K, Olgaard K, et al. Definition, evaluation, and classification of renal osteodystrophy: a position statement from Kidney Disease: Improving Global Outcomes (KDIGO). Kidney Int 2006;69:1945-53.
[3]
Moe SM. Disorders involving calcium, phosphorus, and magnesium. Prim Care 2008;35:215-237. [Pubmed]
[4]
Groothoff JW, Lilien MR, Van de Kar NC, Wolff ED, Davin JC. Cardiovascular disease as a late complication of end-stage renal disease in children. Pediatr Nephrol 2005;20:374-9. [Pubmed]
[5]
KBS. Calcium and Phosphorus Homeostasis. In: Avner ENP (ed.). Pediatric Nephrology, 5 ed. Philadelphia: Williams & Wilkins; 2005. p. 1347-73.
[6]
Jüppner HB. Parathyroid Hormone. Philadelphia: Lippincot: Williams & Wilkins; 1999.
[7]
Koch Nogueira PC, David L, Cochat P. Evolution of secondary hyperparathyroidism after renal transplantation. Pediatr Nephrol 2000;14:342-6. [Pubmed]
[8]
Drueke TB. Cell biology of parathyroid gland hyperplasia in chronic renal failure. J Am Soc Nephrol 2000;11:1141-52. [Pubmed]
[9]
Shimada T, Kakitani M, Yamazaki Y. Targeted ablation of Fgf23 demonstrates an essential physiological role of FGF23 in phosphate and vitamin D metabolism. J Clin Invest 2004;113:561-8. [Pubmed]
[10]
Fukagawa M, Kazama JJ. FGF23: its role in renal bone disease. Pediatr Nephrol 2006;21:1802-6. [Pubmed]
[11]
Liu S, Tang W, Zhou J. Fibroblast growth factor 23 is a counter-regulatory phosphaturic hormone for vitamin D. J Am Soc Nephrol 2006;17:1305-15. [Pubmed]
[12]
Stubbs JR, Liu S, Tang W. Role of hyperphosphatemia and 1,25-dihydroxyvitamin D in vascular calcification and mortality in fibroblastic growth factor 23 null mice. J Am Soc Nephrol 2007;18:2116-24. [Pubmed]
[13]
Komaba H, Fukagawa M. FGF23: a key player in mineral and bone disorder in CKD. Nefrologia 2009;29:392-6. [Pubmed]
[14]
Ben-Dov IZ, Galitzer H, Lavi-Moshayoff V. The parathyroid is a target organ for FGF23 in rats. J Clin Invest 2007;117:4003-8. [Pubmed]
[15]
Kuro-o M, Matsumura Y, Aizawa H. Mutation of the mouse klotho gene leads to a syndrome resembling ageing. Nature 1997;390:45-51. [Pubmed]
[16]
Naveh-Many T, Silver J. Regulation of parathyroid hormone gene expression by hypocalcemia, hypercalcemia, and vitamin D in the rat. J Clin Invest 1990;86:1313-9. [Pubmed]
[17]
Durán CE, Torregrosa JV, Almadén Y. Dynamics of calcium-regulated PTH secretion in secondary hyperparathyroidism: comparison between "in vivo" vs. "in vitro" responses. Nefrologia 2010;30:73-7. [Pubmed]
[18]
Silver J, Yalcindag C, Sela-Brown A. Regulation of the parathyroid hormone gene by vitamin D, calcium and phosphate. Kidney Int Suppl 1999;73:S2-7. [Pubmed]
[19]
Silver J, Levi R. Regulation of PTH synthesis and secretion relevant to the management of secondary hyperparathyroidism in chronic kidney disease. Kidney Int Suppl 2005;S8-12.
[20]
Brown EM, Pollak M, Seidman CE. Calcium-ion-sensing cell-surface receptors. N Engl J Med 1995;333:234-40. [Pubmed]
[21]
Brown AJ, Dusso A, Slatopolsky E. Vitamin D. Am J Physiol 1999;277:F157-175.
[22]
Baker AR, McDonnell DP, Hughes M. Cloning and expression of full-length cDNA encoding human vitamin D receptor. Proc Natl Acad Sci USA 1988;85:3294-8. [Pubmed]
[23]
Garfia B, Canadillas S, Canalejo A. Regulation of parathyroid vitamin D receptor expression by extracellular calcium. J Am Soc Nephrol 2002;13:2945-52. [Pubmed]
[24]
Naveh-Many T, Marx R, Keshet E. Regulation of 1,25-dihydroxyvitamin D3 receptor gene expression by 1,25-dihydroxyvitamin D3 in the parathyroid in vivo. J Clin Invest 1990;86:1968-75. [Pubmed]
[25]
Canadillas S, Canalejo A, Santamaría R, Rodríguez ME, Estepa J, Martín-Malo A, et al. Calcium-sensing receptor expression and parathyroid hormone secretion in hyperplastic parathyroid glands from humans. J Am Soc Nephrol 2005;16:2190-7. [Pubmed]
[26]
Rodríguez M, Canadillas S, López I. Regulation of parathyroid function in chronic renal failure. J Bone Miner Metab 2006;24:164-8. [Pubmed]
[27]
Locatelli F, Cannata-Andia JB, Drueke TB. Management of disturbances of calcium and phosphate metabolism in chronic renal insufficiency, with emphasis on the control of hyperphosphataemia. Nephrol Dial Transplant 2002;17:723-31. [Pubmed]
[28]
Canalejo R, Canalejo A, Martínez-Moreno JM, Rodríguez-Ortiz ME, Estepa JC, Mendoza FJ, et al. FGF23 fails to inhibit uremic parathyroid glands. J Am Soc Nephrol 2010;21:1125-35. [Pubmed]
[29]
Milliner DS, Zinsmeister AR, Lieberman E, Landing B. Soft tissue calcification in pediatric patients with end-stage renal disease. Kidney Int 1990;38:931-6. [Pubmed]
[30]
Hruska KA, Choi ET, Memon I, Davis TK, Mathew S. Cardiovascular risk in chronic kidney disease (CKD): the CKD-mineral bone disorder (CKD-MBD). Pediatr Nephrol 2010;25:769-78. [Pubmed]
[31]
Goodman WG, Goldin J, Kuizon BD, Yoon C, Gales B, Sider D, et al. Coronary-artery calcification in young adults with end-stage renal disease who are undergoing dialysis. N Engl J Med 2000;342:1478-83. [Pubmed]
[32]
Tyson KL, Reynolds JL, McNair R, Zhang Q, Weissberg PL, Shanahan CM. Osteo/chondrocytic transcription factors and their target genes exhibit distinct patterns of expression in human arterial calcification. Arterioscler Thromb Vasc Biol 2003;23:489-94. [Pubmed]
[33]
Giachelli CM, Speer MY, Li X, Rajachar RM, Yang H. Regulation of vascular calcification: roles of phosphate and osteopontin. Circ Res 2005;96:717-22. [Pubmed]
[34]
Moe SM, O'Neill KD, Duan D, Ahmed S, Chen NX, Leapman SB, et al. Medial artery calcification in ESRD patients is associated with deposition of bone matrix proteins. Kidney Int 2002;61:638-47. [Pubmed]
[35]
Bostrom K, Watson KE, Horn S, Wortham C, Herman IM, Demer LL. Bone morphogenetic protein expression in human atherosclerotic lesions. J Clin Invest 1993;91:1800-9. [Pubmed]
[36]
Moe SM, Chen NX. Mechanisms of vascular calcification in chronic kidney disease. J Am Soc Nephrol 2008;19:213-6. [Pubmed]
[37]
Ketteler M, Bongartz P, Westenfeld R, Wildberger JE, Mahnken AH, Böhm R, et al. Association of low fetuin-A (AHSG) concentrations in serum with cardiovascular mortality in patients on dialysis: a cross-sectional study. Lancet 2003;361:827-33. [Pubmed]
[38]
Simonet WS, Lacey DL, Dunstan CR, Kelley M, Chang M-S, Luethy R, et al. Osteoprotegerin: a novel secreted protein involved in the regulation of bone density. Cell 1997;89:309-19. [Pubmed]
[39]
Luo G, Ducy P, McKee MD, Pinero GJ, Loyer E, Behringer RR, et al. Spontaneous calcification of arteries and cartilage in mice lacking matrix GLA protein. Nature 1997;386:78-81. [Pubmed]
[40]
Davies MR, Lund RJ, Hruska KA. BMP-7 is an efficacious treatment of vascular calcification in a murine model of atherosclerosis and chronic renal failure. J Am Soc Nephrol 2003;14:1559-67. [Pubmed]
[41]
Frye MA, Melton LJ, 3rd, Bryant SC, Fitzpa- trick LA, Wahner HW, Schwartz RS, et al. Osteoporosis and calcification of the aorta. Bone Miner 1992;19:185-94. [Pubmed]
[42]
Schulz E, Arfai K, Liu X, Sayre J, Gilsanz V. Aortic calcification and the risk of osteoporosis and fractures. J Clin Endocrinol Metab 2004;89:4246-53. [Pubmed]
[43]
Naves M, Rodríguez-García M, Díaz-López JB, Gómez C, Rodríguez-Rebollar A, Rodríguez-García M, et al. Progression of vascular calcifications is associated with greater bone loss and increased bone fractures. Osteoporos Int 2008;19:1161-6. [Pubmed]
[44]
Rodríguez-García M, Gómez-Alonso C, Naves-Díaz M, Díaz-López JB, Díaz-Corte C. Vascular calcifications, vertebral fractures and mortality in haemodialysis patients. Nephrol Dial Transplant 2009;24:239-46. [Pubmed]
[45]
London GM, Marty C, Marchais SJ. Arterial calcifications and bone histomorphometry in end-stage renal disease. J Am Soc Nephrol 2004;15:1943-51. [Pubmed]
[46]
Matias PJ, Ferreira C, Jorge C, Borges M, Aires I, Amaral T, et al. 25-Hydroxyvitamin D3, arterial calcifications and cardiovascular risk markers in haemodialysis patients. Nephrol Dial Transplant 2009;24:611-8.
[47]
London GM, Marchais SJ, Guerin AP, Boutouyrie P, Metivier F, De Vernejoul MC. Association of bone activity, calcium load, aortic stiffness, and calcifications in ESRD. J Am Soc Nephrol 2008;19:1827-35. [Pubmed]
[48]
Bucay N, Sarosi I, Dunstan CR, Morony S, Tarpley J, Capparelli C, et al. Osteoprotegerin-deficient mice develop early onset osteoporosis and arterial calcification. Genes Dev 1998;12:1260-8. [Pubmed]
[49]
Collin-Osdoby P. Regulation of vascular calcification by osteoclast regulatory factors RANKL and osteoprotegerin. Circ Res 2004;95:1046-57. [Pubmed]
[50]
Panizo S, Cardús A, Encinas M. RANKL increases vascular smooth muscle cell calcification through a RANK-BMP4-dependent pathway. Circ Res 2009;104:1041-8. [Pubmed]
[51]
Torres PU, Prie D, Molina-Bletry V,Beck L, Silve C, Friedlander G. Klotho: an antiaging protein involved in mineral and vitamin D metabolism. Kidney Int 2007;71:730-7. [Pubmed]
[52]
Wang Y, Sun Z. Current understanding of klotho. Ageing Res Rev 2009;8:43-51. [Pubmed]
[53]
Wong YF, Xu Q. Ablation of klotho and premature aging: is 1,25-dihydroxyvitamin D the key middleman? Kidney Int 2009;75:1137-9.
[54]
Kuro-o M. Klotho as a regulator of fibroblast growth factor signaling and phosphate/calcium metabolism. Curr Opin Nephrol Hypertens 2006;15:437-41. [Pubmed]
Idiomas
Nefrología (English Edition)
Article options
Tools
es en

¿Es usted profesional sanitario apto para prescribir o dispensar medicamentos?

Are you a health professional able to prescribe or dispense drugs?