Next Article in Journal
Nocturnal Hemodialysis Leads to Improvement in Physical Performance in Comparison with Conventional Hemodialysis
Next Article in Special Issue
Nutritional Assessment and Support in Children with Chronic Kidney Disease: The Benefits of Working with a Registered Dietitian
Previous Article in Journal
Food Literacy Scale: Validation through Exploratory and Confirmatory Factor Analysis in a Sample of Portuguese University Students
Previous Article in Special Issue
Adipose and Plasma microRNAs miR-221 and 222 Associate with Obesity, Insulin Resistance, and New Onset Diabetes after Peritoneal Dialysis
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Bone Disease in Chronic Kidney Disease and Kidney Transplant

by
Ezequiel Bellorin-Font
1,*,
Eudocia Rojas
2 and
Kevin J. Martin
1
1
Division of Nephrology and Hypertension, Saint Louis University, Saint Louis, MO 63103, USA
2
Division of Nephrology and Kidney Transplantation, University Hospital of Caracas, Caracas 1053, Venezuela
*
Author to whom correspondence should be addressed.
Nutrients 2023, 15(1), 167; https://doi.org/10.3390/nu15010167
Submission received: 11 October 2022 / Revised: 19 December 2022 / Accepted: 19 December 2022 / Published: 29 December 2022
(This article belongs to the Special Issue Relevant Nutritional, Biochemical and Molecular Disorders in CKD)

Abstract

:
Chronic Kidney Disease–Mineral and Bone Disorder (CKD-MBD) comprises alterations in calcium, phosphorus, parathyroid hormone (PTH), Vitamin D, and fibroblast growth factor-23 (FGF-23) metabolism, abnormalities in bone turnover, mineralization, volume, linear growth or strength, and vascular calcification leading to an increase in bone fractures and vascular disease, which ultimately result in high morbidity and mortality. The bone component of CKD-MBD, referred to as renal osteodystrophy, starts early during the course of CKD as a result of the effects of progressive reduction in kidney function which modify the tight interaction between mineral, hormonal, and other biochemical mediators of cell function that ultimately lead to bone disease. In addition, other factors, such as osteoporosis not apparently dependent on the typical pathophysiologic abnormalities resulting from altered kidney function, may accompany the different varieties of renal osteodystrophy leading to an increment in the risk of bone fracture. After kidney transplantation, these bone alterations and others directly associated or not with changes in kidney function may persist, progress or transform into a different entity due to new pathogenetic mechanisms. With time, these alterations may improve or worsen depending to a large extent on the restoration of kidney function and correction of the metabolic abnormalities developed during the course of CKD. In this paper, we review the bone lesions that occur during both CKD progression and after kidney transplant and analyze the factors involved in their pathogenesis as a means to raise awareness of their complexity and interrelationship.

1. Introduction

Chronic kidney disease (CKD) is associated with multiple abnormalities that lead to a complex disorder comprising biochemical alterations in the metabolism of calcium, phosphorus, parathyroid hormone (PTH), Vitamin D, and fibroblast growth factor-23 (FGF-23), abnormalities in bone turnover, mineralization, volume, linear growth or strength, and vascular calcification leading to an increase in bone fractures and vascular disease, which ultimately result in high morbidity and mortality [1,2]. These alterations have been defined by KDIGO (Kidney Disease Improving Global Outcomes) as the Chronic Kidney Disease Mineral and Bone Disorder (CKD-MBD) [2]. Regarding the bone, it has been well established that patients with CKD stages 3a to 5D have increased fracture rates compared with the general population [3,4,5] and that incident hip fractures are associated with substantial morbidity and mortality [6,7,8,9,10]. Kidney transplantation is the treatment of choice for many patients with advanced CKD. However, the improvement of kidney function by means of a kidney transplant, unfortunately does not solve these complex situations completely. Instead, bone disease may persist, progress or transform into a different entity independently of the kidney function due to new pathogenetic mechanisms [11,12,13,14]. In this paper, we will review the bone disease seen in CKD-MBD, also known as renal osteodystrophy, and the pathogenetic mechanisms involved in its development.

2. Bone Disease in CKD

The abnormalities of bone in CKD are defined based on bone histology with histomorphometric analysis. Classically, renal osteodystrophy has been classified in different subtypes encompassing a wide spectrum from high turnover to low turnover. At one end of the spectrum, high PTH levels has been considered a surrogate for high-turnover bone disease, known as hyperparathyroid bone disease or osteitis fibrosa, and characterized by elevated bone turnover, increased number and activity of osteoclasts and osteoblasts, variable alterations in osteoid deposition, usually with a woven pattern, and variable amounts of peritrabecular fibrosis (Figure 1B). At the other end of the spectrum, the distinctive pattern is an adynamic bone disease in which typically, bone turnover is decreased with normal mineralization, a paucity of osteoid, bone cells, and a marked decrease in active remodeling sites (Figure 1C). Osteomalacia, another lesion with low turnover, is characterized by increased osteoid seam width, increase in the trabecular surface covered with osteoid, and decrease in mineralization as assessed by tetracycline labeling (Figure 1D). Mixed uremic osteodystrophy is a complex disorder in which elevated bone turnover coexists with features of osteomalacia [15,16,17] (Figure 1E). The frequency of the two last lesions has decreased consistently in recent decades [18]. More recently, it has been shown that osteoporosis is a frequent feature in patients with CKD-MBD [19,20,21] that may complicate their outcome. This disorder, characterized by a decreased bone mass, strength and quality predisposing individuals to bone fracture, is frequent in the CKD population and is likely associated with the elevated risk of fracture not only in the aging groups but also in younger strata [22]. The definition of osteoporosis has been based mainly on bone densitometry. The World Health Organization (WHO) defines osteoporosis as a disease characterized by low bone mineral density and microarchitectural deterioration leading to low bone strength and increase in fracture risk [23]. In the normal population, osteoporosis is defined as a DEXA T score ≤2.5 DS below the normal range for the peak obtained in young persons. In the non-CKD population, both cortical and cancellous bone are substantially reduced [24]. However, in CKD patients, histologic changes are more difficult to interpret as osteoporosis has been seen coexisting with other types of renal osteodystrophy [25] with no difference in prevalence, among them.
As a mean to standardize the definition of bone disease in CKD, KDIGO proposed a new classification based on bone turnover (T), mineralization (M) and volume (V) or TMV, which highlights the most significant bone alterations relevant for clinical evaluation and therapeutic implications [26,27]. However, the classical definition of renal osteodystrophy described above is still used coexisting with the new classification based on TMV.
Most of the early studies describing the CKD associated bone disease were performed in patients with advanced CKD or ESKD (Table 1). In many of them, high-turnover bone disease was described as the predominant form of renal osteodystrophy [17,28,29,30]. More recently, low-turnover bone disease has been increasingly reported in patients with ESKD [31,32]. In some studies, an important number of patients showed normal bone turnover [17,33,34]. In two large series on bone biopsy in patient on dialysis, comprising 630 patients from USA and Europe [34] and 492 patients from different countries (Brazil, Portugal, Turkey, and Venezuela) [35], low-turnover bone disease was observed in 58% and 52% of the patients, respectively (Table 1). Malluche et al. [34] examined the possible role of race in the type of bone lesions occurring in CKD. Biopsies were analyzed using the parameters of TMV as recommended by KDIGO [2]. As a whole, low turnover was observed in 58% of the cases. This type of lesion was more prevalent in white patients, while in black patients, high turnover was observed in 68% of cases. All patients with high bone turnover were younger. Mineralization defects were seen in only 3% of the cases. Regarding bone volume, biopsies in white patients revealed a similar proportion of normal, high or low bone volume, whereas black patients showed a higher proportion of high volume. No differences were observed regarding diabetes, gender, and treatment with Vitamin D. The differences observed in this study suggest that race may be another factor that may influence the type of bone lesion observed in CKD [34].
In the studies summarized in Table 1, patients are heterogeneous with regard to time of the study, gender, age, treatment with vitamin analogs and phosphate binders, calcimimetics, and other drugs prior to the inclusion in the study, as well as the percent of diabetic patients, among others. The use of aluminum containing phosphate binders was relatively common in earlier studies but less frequent in more recent studies. These aspects are important as they may influence bone turnover among other properties of the bone.
Table 2 summarizes the findings of bone turnover at different stages of CKD not-on dialysis, in studies published from 1976 to 2022. In 1976, Malluche et al. [45] examined bone histology in 50 patients with different stages of kidney disease. It was observed histological evidence of PTH excess, particularly osteoclastic surface resorption, empty osteoclastic lacunae, and woven osteoid in more than 50% of patients with a GFR of 40 mL/1.73 mL/min, whereas endosteal fibrosis was seen when GFR fell below 30 mL/min suggesting that hyperparathyroid bone disease was present since early stages and progressed with advanced CKD. In contrast, in more recent studies, low-turnover bone disease has been increasingly reported predominantly in patients with CKD stages 2 to 4 [46,47,48], with a prevalence that in some cases reached 80 to 100% of the patients. In the study by El-Husseini [48], in patients with a mean eGFR of 44 ± 16 mL/min/1.73 m2, low turnover was observed in 84% of the patients. Of interest, most of them had vascular calcifications which correlated positively with levels of phosphorus, FGF-23 and activin, and negatively with bone turnover as has been also reported previously, whereas others have found less prevalence of vascular calcification in lower CKD stages. Other observation of relatively recent studies is the finding of osteoporosis. Nevertheless, the results are not uniform as the prevalence of low-turnover bone varies among studies likely reflecting the different population examined, degree of kidney function, age, ethnicity, geographic distribution, medications including corticosteroids, immunosuppressants, Vitamin D, and type of renal disease leading to CKD, which may play a role in the type and degree of turnover alteration. Indeed, in preliminary studies (Abstract ASN 2011) performed in the laboratory of two of the authors in a group of 46 patients with CKD stages 3 to 5, and patients with ESKD on hemodialysis, low bone turnover was observed in 7 (15.2%) of the patients with CKD stages 3 to 5 not on dialysis, whereas high bone turnover was seen in 20 patients (43.5%). Bone alterations consistent with osteoporosis [24,25,34] was found in 12 patients (26.1%). Of interest, this finding was more frequently observed in diabetics (33.5%) compared with non-diabetics (27.3%), female gender, and older age. Normal bone histology was seen in 15.2% of the patients. In contrast, in ESKD, 40 (80%) patients had high bone turnover and only 10 patients (20%) had low bone turnover. In this group, we did not observe changes consistent with predominant osteoporosis or normal bone turnover. There is no clear explanation for the different types of bone turnover predominating at CKD stages as well as the apparent increase in the incidence of low bone turnover lesions along the years reported by different authors. It has been considered that medications, particularly those associated with a possible effect in bone metabolism could explain the differences. Vitamin D analogs and cinacalcet have been shown to affect bone metabolism and particularly bone turnover [49,50,51]. Indeed, in prospective studies, it has been shown that cinacalcet diminishes bone turnover after one year of treatment [51,52]. Thus, it has been argued that oversuppression of PTH by these drugs may result in adynamic bone disease. Our patients with CKD stages 3 to 5 not on dialysis were not receiving treatment with medications that may affect mineral metabolism such as Vitamin Derivatives, calcimimetics, steroids, bisphosphonates, etc. Thus, use of these medications cannot explain the findings. This is similar to that reported in many of studies referred in Table 2. Several other possible factors have been proposed as possible causes of the increase in incidence of low-turnover bone disease. Thus, it has been shown evidence of an indirect relationship between eGFR and turnover, which remains after adjustment for age and the presence of diabetes [48,53]. Likewise, resistance to PTH and an increase in PTH fragments that may have an antagonistic effect [54] is another possibility. Increasing evidence suggest that sclerostin [55,56,57], a factor that increases early during the course of CKD and that is associated with low bone turnover could be one of the main causes responsible for these alterations. Further studies with follow up of patients may be required to obtain an explanation to the apparently changing bone turnover as CKD progresses.
In summary, renal osteodystrophy starts early and progresses along the spectrum of CKD. The histologic pattern has changed along the years towards an increase in low-turnover bone disease in all ranges of CKD. Several factors, including age, gender, race, concurrent disease, use of corticosteroids, diuretics, heparin during hemodialysis, and Vitamin D receptor activators may play a role in the type of bone lesions and vascular disease in patients on hemodialysis. Indeed, it has been shown that Vitamin D agonists influence VDR and osteocalcin expression in circulating in endothelial progenitor cells [61]. More recently, osteoporosis has been considered an important component of bone disease in patients with CKD.

3. Pathogenesis of Bone Disease in CKD-MBD

Figure 2 summarizes the mechanisms involved in the pathophysiology of CKD-MBD. The pathogenesis of CKD-MBD is directly associated with alterations in the regulation of the metabolism of calcium and phosphorus by PTH, 1,25(OH)2D3, and FGF-23, which closely interact to maintain the homeostasis.
Extracellular calcium concentrations are tightly controlled within a narrow physiological range optimal for proper cellular functions. Calcium absorption in the intestine is regulated by calcitriol. Calcium acts directly on the parathyroid cell through its specific receptor, the calcium sensing receptor (CaSR), that detects subtle decreases in extracellular calcium, leading to an immediate release of PTH. Conversely, an increase in extracellular calcium suppresses PTH secretion. Numerous studies assign the pathogenesis of the bone alterations of CKD-MBD to the early changes in the metabolism of phosphorus, calcium, FGF-23, and calcitriol that occur as kidney function declines [62,63,64,65,66]. These alterations manifest as an elevation in the levels of FGF-23 and PTH to increase renal phosphate excretion. Studies suggest that FGF-23 increases early in CKD, even prior to a measurable elevation of PTH [67]. It rises phosphate excretion through binding to its klotho co-receptor activating FGFR-1 and FGF-3 receptors leading to a decrease in NaPi2a and NaPi2c cotransporters expression which ultimately will increase phosphate excretion. PTH directly decreases phosphate reabsorption through similar mechanisms. FGF-23 and PTH can maintain phosphate balance until GFR approaches stage 4. Thereafter, neither PTH or FGF-23 are capable of completely maintaining phosphate balance and hyperphosphatemia ensues. The 1,25-(OH)2 Vitamin D (calcitriol) synthesis in the kidney is reduced due to the inhibitory effects of elevated FGF-23 and phosphate on 1-alfa- Hydroxylase [32]. Calcitriol deficiency decreases intestinal calcium absorption and diminishes tissue levels of VDR, resulting in resistance to calcitriol-mediated regulation of PTH secretion. The concurrent decrease in the expression of CaSr in the parathyroid cells stimulates PTH secretion [68,69]. In addition, elevated serum phosphorus increases PTH secretion by mechanisms that include a direct action on the CaSr [70]. All these factors in concert lead to the development of secondary hyperparathyroidism. Elevated PTH activate osteoblasts and osteoclasts via the receptor activator of nuclear factor-kappaB (RANK-L) and osteoprotegerin signaling pathway lading to an increase in bone turnover resulting in a bone structure with lower strength and increased fragility, which contributes to an elevation of bone fracture risk and alterations in vascular metabolism resulting in vascular and valvular calcifications [64,65,71,72,73]. Of interest, heparin, an agent to which ESRD patients on hemodialysis are frequently exposed, has been shown to increase osteoprotegerin intra-and postdialytic levels, thus suggesting that this could be an additional factor in the pathogenesis of bone and vascular disease in hemodialysis patients [74]. Additional factors and mechanisms, including inhibition of the canonical Wnt/B catenin signaling pathway, and accumulation of uremic toxins such as indoxyl sulfate have also been proposed as factors that may lead to disruption of the normal regulation of bone turnover leading to renal osteodystrophy and vascular disease [59,75,76,77].
The direct role of FGF-23 on bone metabolism is not clear. Studies have shown that in CKD, osteocytes exhibit an increased synthesis of FGF-23. A relationship between FGF-23 and bone abnormalities occurs through its effects on phosphate excretion and suppression of calcitriol synthesis [65,78]. However, FGF-23 levels have been shown to be elevated even before phosphate levels are increased [67]. In addition, it has been suggested an association between FGF-23 with alterations in bone mineralization [79], and FGF-23 and alfa-klotho, the coreceptor for FGF-23, have been shown to stimulate osteoblastic-like cell proliferation and inhibit mineralization [47,78]. Thus, a study in adult dialysis patients found that patients with high bone turnover had higher serum levels of FGF-23 compared with patients with low bone turnover. Likewise, patients with high FGF-23 had normal mineralization, whereas delayed mineralization correlated negatively with FGF-3 levels. Using regression analysis, FGF-23 was the only independent predictor for mineralization lag time [60]. Similarly, in pediatric patients with high bone turnover renal osteodystrophy, it has been shown an association between high serum levels of FGF-23 and improved mineralization, although a correlation between FGF-23 and bone formation rates was not observed [80]. Furthermore, studies examining the expression of proteins in bone tissue of patients with CKD stages 2 to stage 5 on dialysis and healthy individuals have shown that as serum calcium declines, serum alkaline phosphatase, FGF-23, PTH, and osteoprotegerin increase with progression of CKD [47]. These alterations occurred while there was an increase in bone resorption, decreased bone formation and impairment in bone mineralization. Of interest, sclerostin and PTH-receptor-1 expression in the bone was higher in early stages of CKD whereas FGF-23 was elevated in late stages. FGF-23 expression was observed mainly in early osteocytes, whereas sclerostin, which is considered a marker of mature osteocytes, was expressed in cells deeply embedded in the mineralization matrix. These proteins did not co-localize in the same cells. In other studies, high bone turnover was associated with high FGF-23, whereas low bone turnover was observed with lower FGF-23 [81]. Thus, FGF-23 seems to play a direct role in bone metabolism and may be a predictor of bone mineralization in patient with CKD on dialysis and a marker to predict alterations in bone metabolism.
Studies have demonstrated a relationship between PTH and FGF-23 in the bone. In osteoblast-like UMR106 cells, PTH increases FGF-23 mRNA levels and inhibits sclerostin mRNA messenger, which is an inhibitor of the Wnt/beta-catenin pathway. These studies directly associate the effects of PTH and FGF-23 likely via stimulation of the Wnt pathway. Sclerostin levels vary with renal function. Thus, the expression of sclerostin in jck mouse, a model of progressive kidney disease occurs at early stages of CKD, even before PTH and FGF-23 increase [57], suggesting that sclerostin may play an early role in the development of renal osteodystrophy. Conversely, sclerostin has been associated with adynamic bone disease and vascular calcification [82]. Increased sclerostin has been observed in early CKD but the mechanisms are not clear. It has been suggested that it may relate to partial calcitonin exposure, disturbed phosphate metabolism and extraskeletal sources [55,83].
Serum levels of sclerostin and PTH correlate negatively in patients with CKD stage 5D [57]. In addition, in unadjusted and adjusted analysis, sclerostin correlated negatively with bone turnover, osteoblast number and function, and was superior to PTH for the positive prediction of high bone turnover and osteoblast number. On the other hand, PTH was superior to sclerostin for negative prediction of low bone turnover [53]. These findings agree with studies in mice showing that PTH directly inhibits sclerostin transcription in vivo [84,85], suggesting that sclerostin may be useful as a marker of high bone turnover. This negative correlation is in agreement with the negative regulatory function of sclerostin in the intracellular transduction of the PTH signal described in vitro and in vivo [84]. Moreover, studies in patients with renal osteodystrophy subjected to bone biopsy have shown an inverse correlation of sclerostin and PTH levels, as well as a negative association with bone turnover [47,57]. Receiver operator curves analysis showed that PTH and FGF-23 were able to predict high bone turnover, whereas sclerostin was a good predictor of low bone turnover. Sclerostin expression in bone was higher at early stages of CKD and has shown association with lower bone formation rate and greater mineralization defect. These findings favor the notion that sclerostin may play a role in the development of adynamic bone disease in CKD, and hence on fractures [86].
Although elevation of PTH is an early finding in CKD, low bone turnover has been increasingly described in patients with CKD [31,32]. The mechanisms behind these apparently incongruent findings are complex and include maladaptation to the pathophysiologic mechanisms described above, inappropriate PTH signaling and hyporesponsiveness of the PTH receptor, repressed Wnt/B-catenin signaling [55], and elevation of sclerostin in early CKD [47]. Other factors may include the use of medication to prevent or control secondary hyperparathyroidism, oxidative modification of PTH [87], nutritional determinants, age, and underlying diseases. Another factor that has been associated with the early events in the pathogenesis of renal osteodystrophy is indoxyl sulfate. This compound increases in early CKD, even before FGF-23, and has been associated with resistance to PTH [48].
In summary, the mechanisms responsible for the development of renal osteodystrophy are complex and multiple. Based on the sequence of pathophysiologic events that occur during the development of CKD-MBD, high bone turnover is expected to be the most frequent histologic alteration observed in early CKD as PTH starts to increase when GFR drops below 60 mL/min/1.73 m2 [10,67,88,89]. Although some studies have confirmed this notion [90], others have shown a high prevalence of adynamic bone disease in the early stages of CKD and even a change of pattern as CKD progresses. Thus, it seems that a single mechanism does not fit all the phenotypes described at the different stages of CKD and that other factors associated with bone disease may also be determinant to the increased bone fragility and fracture in patients with CKD. The interpretation of these findings together is difficult as there is a vast variability in the populations studied, including age, ethnicity, geographic and social background, among others. In evaluating the causes of bone fragility and fracture in CKD, osteoporosis has been increasingly described in patient with CKD and considered to play a major role in bone fracture, particularly given the fact that a high proportion of patients with CKD bone disease have also the usual risk factors for osteoporosis [91].

4. Bone Fracture in CKD

Bone fracture is a frequent complication of CKD. Table 3 summarizes a number of studies examining the incidence of fractures in patients with CKD. Patients with CKD stages 3a to 5D have higher rate of bone fractures compared with the general population [5,6,7,8,9,10,23,92]. Most studies show that the incidence of fracture increases as GFR decreases, as well as an association between fracture and age in patients with CKD which is superior to that in patients of similar age in the general population. Likewise, the risk of mortality is superior in CKD patients with fracture compared with the general dialysis population. The causes are diverse. Thus, in addition to the pathophysiologic mechanisms leading to renal osteodystrophy, many other factors that include nutrition, medications, concurrent diseases such as diabetes, cardiovascular disease, sarcopenia, increased propensity to fall, among others, may per se increase the risk of fracture in patients with CKD [93]. The estimated prevalence of CKD varies greatly between countries but may reach between 9% and 13.4% of the global population [94,95]. Therefore, the expected prevalence of bone fracture associated with CKD is also high [96]. In most patients, CKD is thought to progress slowly and the bone and the mineral derangement that start in early stages progress relatively silent until it reaches advanced stages in which fracture incidence increases and is more evident. As shown in Table 3, there is an increase in fractures, particularly of the hip [6,7,8,97] but is also seen in other bones. Although this is frequently associated with older age, evidence shows that in general, the incidence of fractures in patients with CKD is elevated. Indeed, as shown in Table 3, several studies have revealed that patients with CKD stages 3 to 5, and those undergoing dialysis have an increased incidence of fractures compared with age-matched subjects without CKD [23]. In addition, the study by Klawansky et al. [98], based on the NHANES III population, a strong trend was noted in which lower bone mineral density (BMD) as determined by dual energy X-ray absorptiometry (DEXA) was strongly associated with reduced creatinine clearance as estimate by the Cockcroft-Gault (CCr) equation. The percentage of women with CCr < 35 mL/min, increased from 0.3% for women with BMD in the normal range to 4% for women with osteopenia to 24% for those with osteoporosis. Similar trend was observed in men, albeit to a lesser extent. Likewise, in another population-based study the prevalence of osteoporosis was 31.8% in patients with CKD stages 3–5 [9]. Thus, the combination of CKD, osteoporosis, and age, results in an elevation of fracture risk.
Osteoporosis is frequent in the CKD population and is likely associated with the elevated risk of fracture not only in the aging groups but also in younger strata [22]. The prevalence of osteoporosis increases with age, a condition that parallels CKD which is also more prevalent in the aging population. Thus, it has been argued that the higher rates of fracture in CKD patients may be a consequence of an increase in the prevalence of age-related osteoporosis. In the younger population, the data are more conflicting. In a recent population based prospective study performed in 19,391 individuals 40 to 69 years old from Canada, with CKD stages 2 and 3 followed for 70 months, it was found that, compared with the median eGFR of 90 mL/min/1.73 m2, those with eGFR of ≤60 had an increased risk of fracture in unadjusted and adjusted models [9]. This effect was more evident in younger individual. Hence, there is clear evidence of an increase in fracture risk at all stages of CKD. However, the studies are difficult to compare because most of them followed different methodology and the population studied are heterogeneous.
The complex alterations that express as low bone mineral density bone mineral density BMD in DEXA studies constitute important components of the bone abnormalities observed in CKD patients and may manifest as osteoporosis and an increased risk for fracture, particularly in those with advanced CKD [104,105,106]. However, in reality, the mechanical properties of the bone express a combination of factors that include aspects related to the specific changes of bone quality associated with renal osteodystrophy [21,107,108] and those due to osteoporosis itself. Thus, low turnover is associated with microstructural abnormalities, whereas bone with high turnover is associated with alteration of material and mechanical property [104]. Some have suggested the combination of the bone alterations described in renal osteodystrophy and the addition of bone fragility osteoporosis be described as “uremic osteoporosis” [65,109].
As previously discussed, bone quality in patients with CKD is best characterized by analysis of bone remodeling, mineralizing and volume properties by means of a bone biopsy with histomorphometry. However, this is an invasive method that requires technical and financial resources that are not available for clinical purposes in most centers. In contrast, DEXA, a method widely available, allows the evaluation of bone quantity. Several studies analyzing DEXA in patients with different stages of CKD have provided abundant information indicating an association between CKD-MBD and osteoporosis. In the KDIGO CKD-MBD guideline of 2009 [1], routinely evaluation of BMD testing was not suggested to be performed in patients with CKD, because based on the evidence available at that time, BMD would not predict fracture risk as it does in the general population and would not predict the type of renal osteodystrophy. However, in the updated KDIGO CKD-MBD guideline of 2017 [110], after new prospective studies documented that lower DXA BMD predicts incident of fractures in patients with CKD G3a–G5D [102,111,112,113], it is suggested that in patients with CKD stages G3a–G5D with evidence of CKD-MBD and/or risk factors for osteoporosis, BMD testing to assess fracture risk be performed if results will impact treatment decisions. In one of those studies, BMD by DEXA measured yearly in 485 hemodialysis patients was useful to predict any type of fracture for females with low PTH or to discriminate spine fracture for any patient [111]. Of note, a significant greater risk for fractures was observed with PTH levels either below 150 or above 300 pg-ml. Likewise, bone alkaline phosphatase was a useful surrogate marker for any type of fracture [111,112]. Another study by Naylor et al. [113] has shown that the Fracture Risk Assessment Tool (http://www.shef.ac.uk/FRAX/index.aspx.) with or without BMD measurement is also useful to predict major osteoporotic fracture. Over a period of 5 years, the risk for fracture was not different in individuals with eGFR <60 mL/min/1.73 m2 compared with those with an eGFR > 60 mL/min/1.73 m2. The trabecular bone score (TBS) is a DEXA-derived algorithm for the evaluation of bone microarchitecture whose utility in patient with osteoporosis has been largely demonstrated [48,114,115]. Studies have shown that TBS may also be useful to measure bone quality in patients with CKD-MBD [114,116]. Indeed, in CKD patient, TBS was significantly associated with the histologic parameters of trabecular bone volume and trabecular spacing but not with dynamic parameters, suggesting that TBS reflects trabecular microarchitecture and cortical width [117]. However, in another study, there was no significant relationship between TBS and bone histomorphometric parameters. TBS has also been shown to correlate inversely with coronary calcification and aortic calcification. Thus, TBS is an important parameter to consider in the evaluation of CKD bone and cardiovascular disease. The fact that TBS may be obtained with DEXA analysis opens an opportunity for further evaluation of bone disease in CKD patients using non-invasive methods. However, further studies are needed to demonstrate whether TBS may predict clinical fractures in patients with CKD-MBD.
Given the limitations of BMD to ascertain the type of bone disease in patients with CKD and the difficulties in performing a bone biopsy as a method to evaluate bone abnormalities in large number of patients, studies have analyzed the possible benefit of other methods than can provide more details of the bone structure. High resolution peripheral quantitative computed tomography (HR-pQCT) can detect microarchitectural changes in patients with CKD [44,118]. A recent study in CKD patients, evaluating bone biomarkers, bone histomorphometry, and HR-pQCT revealed lower BMD, mostly due to trabecular bone impairment compared to controls. It was found that radius BMD and microarchitecture were negatively associated with bone turnover in advanced CKD [44]. HR-pQCT was able to discriminate low bone turnover from non-low bone turnover, whereas there was no difference in DXA BMD between different bone turnover classes. A similar trend has been shown on HR-pQCT and bone turnover as determined by biomarkers in women on dialysis [119].
Although several serum biomarkers have been used to assess bone activity in the general population, their use in patients with advanced CKD and those on treatment with dialysis is limited as many of them are affected by renal function. However, recent studies have shown that some biomarkers, are not affected by the kidney function and thus, can be useful to discriminate the type of bone turnover alteration in patients with CKD. As shown in Table 4, PTH, bone specific alkaline phosphatase (BSAP), N-terminal propeptide of collagen type 1 (P1NP), and tartrate-resistant acid phosphatase 5B (TRAP-5B), which are not affected by kidney function, may correlate with bone histomorphometry findings. BSAP is produced by osteoblasts during bone formation. Thus, it is considered a marker of bone formation and is associated with fracture and all cause cardiovascular mortality [120,121]. Studies have shown a positive predictive value of BSAP for low bone turnover [35,36,58,122]. In the study by Sprague et al. [35] evaluating the accuracy of bone turnover markers to discriminate bone turnover in 492 ESRD subjected to bone biopsy, PTH (iPTH) and bone specific alkaline phosphatase (BSAP) were able to discriminate low from non-low and high from non-high bone turnover, whereas P1NP, another marker of bone formation, that has been shown to be reliable in CKD in some studies, did not improve the diagnostic accuracy. Similar results, for iPTH and BSAP, have been shown by other studies (Table 4). In contrast, it has also been found that BALP, intact P1NP, TRAP5B, and radius HR-pQCT, but not PTH can discriminate low bone turnover [44]. Other biomarkers such as osteocalcin and FGF-23 albeit promising, require further studies. Therefore, differences still exist as to which biomarkers are useful in the diagnosis of bone turnover. In this regard, Evenepoel et al. [122], in an analysis of several studies on the use of different bone biomarkers, concluded that there is not consistent evidence to replace bone histomorphometry for the diagnosis of bone turnover. It is possible that differences in biomarker assays and the populations evaluated may influence the different results across studies. Meanwhile, the KDIGO CKD-MBD guideline update of 2017 suggests that measurements of serum PTH or BSAP can be used to evaluate bone disease in CKD stages 3–5D because markedly high or low values predict underlying bone turnover stage 3 to 5 [109].
In summary, renal osteodystrophy starts early during the course of CKD and is determined by complex pathophysiologic mechanisms that result in alteration of bone turnover and to a lower extent, mineralization and volume. The pattern of renal osteodystrophy has changed over the years towards an increase in the prevalence of low-turnover bone disease. Osteoporosis is a frequent feature observed in patients with renal osteodystrophy which may play an important role in the elevated risk of fracture in these patients. CKD stage, age, race, and probably other factors may have a role in the type and severity of the bone alterations seen in CKD. PTH and alkaline phosphatase are good predictors to differentiate high and low bone turnover disease, whereas DEXA does not discriminate between the histologic and architectural defects that define renal osteodystrophy but seems important in evaluating the risk of fracture.

5. Bone Disease after Kidney Transplantation

For many patients with ESRD, kidney transplantation is the treatment of choice as it improves the quality of life, the metabolic derangements resulting from CKD, and patient survival compared with dialysis. However, numerous studies have demonstrated that after transplantation, bone disease may persist, progress, or evolve into a different phenotype of CKD-MBD [129,130,131,132]. Thus, the risk of fracture in transplant patients increases about 30% over that in CKD patients during the first 3 years after transplantation [99], and up to 25% of transplant receptors may suffer a fracture in the 5 years that follow a kidney transplant. Studies have found evidence of osteoporosis in kidney transplant patients depending on the gender or the DEXA measured site in up to 44% of patients [133]. An interesting aspect to be considered is the relationship between BMD and fracture risk in transplant patients. Although a low BMD is a potent fracture risk factor, many transplant patients with low BMD do not have fractures [134], and an overlap in the BMD measurements between patients with and without fractures has also been shown [135].
Several conditions have been implicated as risk factors for fracture after kidney transplantation. Studies have shown that the risk is increased in white women over 65 years of age, deceased donor kidney recipients, increased HLA mismatches, previous diabetes mellitus, long time on dialysis prior to transplant, the type and severity of pretransplant bone disease, aggressive immunosuppression induction, glucocorticoids, calcineurin inhibitors and alterations in the metabolism of calcium, phosphorus, Vitamin D, and PTH [11,14,132]. Hyperparathyroidism is frequent in CKD as amply discussed previously. PTH levels decrease during the first 3 to 6 months after transplantation [136] due to the improvement of many abnormalities involved in its pathogenesis. Persistent hyperparathyroidism has been considered a maladaptive response of the parathyroid gland resulting from pre-exiting secondary hyperparathyroidism developed during the course of CKD. PTH may remain high particularly within the first year after transplantation in up to 60% of patients [11,14,137,138,139]. The presence of hyperparathyroidism before transplantation, the duration of dialysis, and the development of nodular and monoclonal hyperplasia of the parathyroid gland are the most important mechanisms in the persistence of hyperparathyroidism after transplantation. Therefore, persistent hyperparathyroidism may be a cause of bone disease after transplantation. De novo hyperparathyroidism (compensatory adaptive response) in kidney transplant patients results from elevated PTH levels as a consequence of deteriorating transplant function to maintain phosphate levels and calcium metabolism [140] Therefore, both, persistent hyperparathyroidism and de novo hyperparathyroidism may be a cause of bone disease after transplantation. Hypophosphatemia is a frequent finding after transplantation and has been associated with alterations in bone histology. Mechanisms of hypophosphatemia are diverse, but elevated FGF-23 may play a role. Alterations of Vitamin D metabolism is another important factor that may be associated with posttransplant bone disease as many patients arrive to the transplant period with low levels of Vitamin D [141]. The elevated FGF-23 levels seen in ESRD patients on dialysis decrease after transplantation but may persist high to counteract hyperphosphatemia favored by reduced kidney function. However, at the same time, FGF-23 inhibits inhibit 1-alfa-hydroxylase in the kidney resulting in lower levels of 1,25(OH)2Vitamin D. These alterations, together with elevated PTH favor the development or maintenance of the bone lesions observed in kidney transplant recipients.
Age has been shown to be a strong risk factor for fracture. Thus, for each decade of life, the risk of hip fracture is estimated to be 55% higher. Fortunately, the role of glucocorticoid use, an important risk factor for bone fractures, seems to be decreasing in transplant patients after new protocols with lower doses or shorter term are increasingly common [142,143,144,145].
Initial studies showed that the changes in BMD differ in early and late posttransplant periods. Julian et al. [136] showed that BMD in the lumbar spine decreases sharply by almost 7% during the first year after transplantation with a somewhat less pronounced decrease thereafter, reaching around 9% at 18 months. Milkus et al. [146] in a prospective study showed a loss of BMD in lumbar spine within 6 months with no significant loss in femoral neck, while other authors have reported a decrease in BMD in femoral neck at 3 months after transplantation [147]. The bone loss seen after a kidney transplant may persist for years but ultimately tends to improve in patients with preserved kidney function. Carlini et al. [11,12,148] demonstrated in a group of long-term renal transplant patients that BMD progressively improved as time after transplantation increased, approaching normal values after 10 years. Other studies analyzing BMD in patients between 6 and 20 years after renal transplantation showed a mean annual decrease in lumbar T scores of −0.6 ± 1.9% [134], a value relatively similar to that observed in the general population with aging [149]. In an evaluation of 3992 first kidney transplants from the Swedish National Renal Registry, 279 fractures occurred (7% of all patients), of which 139 were in the forearm, 69 in the hip, 45 in the humerus and 26 in the spine. The multivariate-adjusted fracture incidence rates were highest during the first 6 months after transplantation, and 86% higher in women than in men. High age, female gender, diabetes nephropathy, history of previous fracture, dialysis, and acute transplant rejection were associated with an increased risk of fracture [150], whereas pre-emptive kidney transplant was associated with a lower risk of fracture. In patients with major fractures followed by a median time of 3 years, mortality associated with hip fracture was the highest (148/1000 patient-years) followed by spine fracture (87/1000 patient year). In adjusted Cox proportional multivariate analysis hip fracture and spine fractures had the worst clinical outcome, whereas forearm fractures were not associated with increased mortality risk [150].
Recently, Evenepoel et al. [145], examined prospectively BMD and bone turnover markers in transplant recipients. BMD was determined by DEXA within 2 weeks after transplantation. During the average follow-up of 5.2 years, 38 patients (7.3%) sustained a fragility fracture, corresponding to 14.2 fractures per 1000 person-year. The median time from transplantation to the first fracture was 17.1 months, and the prevalence of osteoporosis ranged between 10% and 35%. In more detail, osteopenia and osteoporosis were 34.5% and 39.6% at the distal radius, and 22.1% and 55% at the femoral neck, respectively. Patient with lower T-scores were women, older, had higher PTH and lower BMI and sclerostin. Of interest, bone turnover markers were inversely correlated with BMD at all skeletal sites, suggesting that bone markers may increase the ability to evaluate fracture risk in this patient population. Of note, 30 patients from a total of 518 evaluated, had a previous history of fragility fractures at the time of transplantation, and osteoporosis or osteopenia was observed in 77% of them, indicating that they had a severe bone disease prior to transplantation. In a more recent prospective study by the same group performed in 97 subjects evaluated before or at the time of kidney transplantation, and re-evaluated at 12 months posttransplant, it was observed that changes in BMD were highly variable ranging from −18% to + 17% per year. Significant bone loss was observed at the distal third radius and ultradistal radius, with no overall changes in BMD at the spine or hip. Cumulative steroid dose was related with bone loss at the hip, while resolution of hyperparathyroidism related to bone gain at spine and hip [151]. It is important to consider that the incidence and type of fracture varies between different studies, which may reflect the heterogeneity of the populations analyzed. It seems that fractures at peripheral sites rather than central skeleton are more frequent [150].
In conclusion, decreased BMD is frequent and more pronounced during the first months after kidney transplantation but varies individually and with the population studied. Age, sex, time in dialysis before transplantation, bone disease before transplantation, steroids use, and mineral metabolism, among others are important risk factors for the loss of BMD observed, and ultimately for fracture and mortality.

6. Alterations of Bone Histology after Transplantation

In early reports, posttransplant bone disease was mainly considered a consequence of glucocorticoids. However, it has become evident that it comprises a variety of metabolic alterations in the pretransplant and posttransplant periods. The pre-existing renal osteodystrophy plays a central role as this is an almost universal complication in patients with CKD. Thus, as previously described, different alterations in bone turnover, volume, and to a lesser extent, mineralization are the dominant features of altered bone histology in CKD patients. In addition, many patients with CKD arrive to transplant with different degrees of osteoporosis and alterations of bone strength. Although after transplantation, the rapid improvement of kidney function helps controlling many of the pathophysiologic mechanisms involved in the development of renal osteodystrophy, in many cases kidney function does not reach normality or decreases after some time, thus maintaining to a certain degree part of the known mechanisms responsible for bone alterations in CKD. In other cases, bone alterations such as osteoporosis may occur or progress adding to the basic defective bone metabolism.
The histology of the bone alterations described after transplantation help understand its pathogenesis and evolution along the years. The initial studies by Julian et al. [136], showed that as early as 6 months after transplantation, patients showed a decrease in bone formation and bone apposition rate, and reduced cortical thickness. These findings were interpreted as adynamic bone disease. The factors involved in these changes may include a rapid decrease in PTH levels in patients with relatively mild bone disease, and the effect of high doses of glucocorticoids used in early months after transplantation. Similarly, in patients undergoing bone biopsy around 6 years after transplant, low bone turnover, low bone volume and focal or generalized osteomalacia were frequent histologic features [152]. In patients with long-term renal transplantation and relatively preserved GFR the results are discrepant. We have described a mixed histologic pattern with increased bone resorption, low bone formation, and prolonged mineralization lag time in patients with a mean of 7.5 years after transplantation and relatively preserved kidney function. These lesions were more severe in patients with less time after transplantation but approached normal values in patients with more than 10 years after transplant [148,153]. Similar results have been published by others [13]. Other studies have confirmed that the pattern of histologic changes after transplantation is heterogeneous although a decrease in bone formation and mineralization tend to be a relatively frequent finding [154,155,156,157]. Prospective studies performed at different times posttransplant have helped to clarify the changes in bone histomorphometry. Rojas et al. [158] studied the early alterations in bone remodeling in patients subjected to a bone biopsy performed on the day of transplantation and repeated within 21 to 120 days later. The main histologic alterations were a decrease in osteoid and osteoblast surfaces, and a decrease in bone formation rate with a prolongation of mineralization lag time. In addition, almost half of the patients, most of them with adynamic bone disease and mixed bone disease prior to transplant, showed early osteoblast apoptosis and a decrease in osteoblast surface and number. Of interest, lower levels of serum phosphate were observed in those patients whose biopsies showed osteoblast apoptosis, and there was a positive correlation between posttransplant serum phosphate and osteoblasts number. It was suggested that impaired osteoblastogenesis and early osteoblast apoptosis may play a role in posttransplant bone disease. Hypophosphatemia has been described in the posttransplant period, but it is not clear to what extent it may be associated with mineralization defects [155]. In a recent prospective longitudinal study, Jorgensen et al. [151] showed that phosphate levels and nadir after transplantation correlated inversely with changes in mineralization, suggesting that hypophosphatemia may negatively affect bone mineralization. PTH levels, glucocorticoids, and phosphatonins have been suggested as possible causes of hypophosphatemia in these patients [137].
Analysis of bone biopsies utilizing the TMV system have helped understand and define the histologic changes that occur in the bone after transplantation. In a recent prospective study, Keronen et al. [157] showed that the proportion of patients with high bone turnover declined from 63% to 19% at two years posttransplant, while the presence of low bone turnover increased from 8% to 38% and mineralization defects increased from 33 to 44%. Trabecular bone volume showed little changes after transplantation. In a prospective study involving 97 patients undergoing a bone biopsy before or at the time of transplantation and a repeat biopsy 12 months after transplantation [151], bone turnover remained normal or improved in the majority of patients. Bone resorption as well as fibrosis decreased while delayed mineralization was present in 15% of the patients after transplantation. Hypophosphatemia was an important finding, particularly during the first 3 months. However, at 12 months 13% of the patients still had hypophosphatemia (serum phosphate less than 2.3 mg/dL). Interestingly, delayed mineralization was observed in a subset of patients after transplantation and associated with the duration and severity of hypophosphatemia. In densitometry analysis, the cumulative dose of steroids was related to bone loss at the hip whereas resolution of hyperparathyroidism was related to bone gain.
Thus, bone disease after transplantation is a complex disorder that relates to the pretransplant renal osteodystrophy, changes in mineral metabolism after transplantation, ethnicity, age at the time of transplantation, prior and de novo acquired hypoparathyroidism, osteoporosis, use of glucocorticoids, and CNI inhibitors among other factors. Hence, in analyzing bone disease after kidney transplantation, it is important to consider that not all patients arrive to transplant with a uniform bone histologic pattern. High bone turnover has been observed in many of them, whereas in other studies, low bone turnover has been the most frequent finding. Interestingly, however, since the early studies a decrease in bone turnover after transplantation seems to be the most frequent observation. Similarly, decreased mineralization has also been reported in several studies. The mechanisms responsible for this change are not completely clear. It is possible that impaired phosphate metabolism plays a role in this bone defect, at least in part, given the association of impaired mineralization with low phosphate levels observed in a subset of transplanted patients.
An additional consideration should be given to discrepancies that may occur in the interpretation of the findings of the bone biopsy as the reference ranges used by different laboratories for histomorphometric parameters may vary. Indeed, in a recent reanalysis of two set of data of bone histomorphometry in kidney transplant patients using reference ranges from different laboratories, there were disagreements in the categorization of bone turnover on the basis of the cutoff applied, which at the end may affect the diagnostic precision with potential clinical impact [159].
In summary, CKD-MBD [2] comprises mineral and hormonal alterations, abnormalities in bone turnover, mineralization, volume, linear growth, leading, and vascular calcification leading to an increase in bone fractures and vascular disease, which ultimately result in high morbidity and mortality. The bone alterations occurring during the course of CKD, typically described as renal osteodystrophy may extend or evolve to a different pattern after kidney transplantation by mechanisms that include but are not limited to those leading to the CKD. Osteoporosis is a frequent finding in patients with CKD-MBD that can aggravate the bone disease and increase the risk of fracture (Figure 3).

Author Contributions

E.B.-F., E.R. and K.J.M. contributed equally to the preparation of the manuscript. All authors have read and agreed to the published version of the manuscript.

Funding

This research received no external funding.

Institutional Review Board Statement

Not applicable.

Informed Consent Statement

Not applicable.

Data Availability Statement

Not applicable.

Conflicts of Interest

EBF and ER declare no conflict. KJM is a consultant for Amgen, Ardelyx, and Applied Therapeutics.

References

  1. Kidney Disease: Improving Global Outcomes CKDMBDWG. KDIGO clinical practice guideline for the diagnosis, evaluation, prevention, and treatment of Chronic Kidney Disease-Mineral and Bone Disorder (CKD-MBD). Kidney Int. Suppl. 2009, S1, 130. [Google Scholar]
  2. Moe, S.M.; Drueke, T.; Lameire, N.; Eknoyan, G. Chronic kidney disease-mineral-bone disorder: A new paradigm. Adv. Chronic Kidney Dis. 2007, 14, 3–12. [Google Scholar] [CrossRef]
  3. Ball, A.M.; Gillen, D.L.; Sherrard, D.; Weiss, N.S.; Emerson, S.S.; Seliger, S.L.; Kestenbaum, B.R.; Stehman-Breen, C. Risk of hip fracture among dialysis and renal transplant recipients. JAMA 2002, 288, 3014–3018. [Google Scholar] [CrossRef] [Green Version]
  4. Alem, A.M.; Sherrard, D.J.; Gillen, D.L.; Weiss, N.S.; Beresford, S.A.; Heckbert, S.R.; Wong, C.; Stehman-Breen, C. Increased risk of hip fracture among patients with end-stage renal disease. Kidney Int. 2000, 58, 396–399. [Google Scholar] [CrossRef] [Green Version]
  5. Nickolas, T.L.; McMahon, D.J.; Shane, E. Relationship between moderate to severe kidney disease and hip fracture in the United States. J. Am. Soc. Nephrol. 2006, 17, 3223–3232. [Google Scholar] [CrossRef]
  6. Coco, M.; Rush, H. Increased incidence of hip fractures in dialysis patients with low serum parathyroid hormone. Am. J. Kidney Dis. 2000, 36, 1115–1121. [Google Scholar] [CrossRef] [PubMed]
  7. Hung, L.W.; Hwang, Y.T.; Huang, G.S.; Liang, C.C.; Lin, J. The influence of renal dialysis and hip fracture sites on the 10-year mortality of elderly hip fracture patients: A nationwide population-based observational study. Medicine 2017, 96, e7618. [Google Scholar] [CrossRef] [PubMed]
  8. Lin, Z.Z.; Wang, J.J.; Chung, C.R.; Huang, P.C.; Su, B.A.; Cheng, K.C.; Chio, C.C.; Chien, C.C. Epidemiology and mortality of hip fracture among patients on dialysis: Taiwan National Cohort Study. Bone 2014, 64, 235–239. [Google Scholar] [PubMed]
  9. Desbiens, L.C.; Goupil, R.; Madore, F.; Mac-Way, F. Incidence of fractures in middle-aged individuals with early chronic kidney disease: A population-based analysis of CARTaGENE. Nephrol. Dial. Transplant. 2020, 35, 1712–1721. [Google Scholar] [CrossRef] [PubMed]
  10. Pimentel, A.; Urena-Torres, P.; Zillikens, M.C.; Bover, J.; Cohen-Solal, M. Fractures in patients with CKD-diagnosis, treatment, and prevention: A review by members of the European Calcified Tissue Society and the European Renal Association of Nephrology Dialysis and Transplantation. Kidney Int. 2017, 92, 1343–1355. [Google Scholar] [CrossRef] [Green Version]
  11. Bellorin-Font, E.; Rojas, E.; Carlini, R.G.; Suniaga, O.; Weisinger, J.R. Bone remodeling after renal transplantation. Kidney Int. Suppl. 2003, 63, S125–S128. [Google Scholar] [CrossRef]
  12. Weisinger, J.R.; Carlini, R.G.; Rojas, E.; Bellorin-Font, E. Bone disease after renal transplantation. Clin. J. Am. Soc. Nephrol. 2006, 1, 1300–1313. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  13. Cueto-Manzano, A.M.; Konel, S.; Hutchison, A.J.; Crowley, V.; France, M.W.; Freemont, A.J.; Adams, J.E.; Mawer, B.; Gokal, R. Bone loss in long-term renal transplantation: Histopathology and densitometry analysis. Kidney Int. 1999, 55, 2021–2029. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  14. Evenepoel, P. Recovery versus persistence of disordered mineral metabolism in kidney transplant recipients. Semin. Nephrol. 2013, 33, 191–203. [Google Scholar] [CrossRef]
  15. Monier-Faugere, M.C.; Malluche, H.H. Trends in renal osteodystrophy: A survey from 1983 to 1995 in a total of 2248 patients. Nephrol. Dial. Transplant. 1996, 11 (Suppl. 3), 111–120. [Google Scholar] [CrossRef] [Green Version]
  16. Sprague, S.M. Renal bone disease. Curr. Opin. Endocrinol. Diabetes Obes. 2010, 17, 535–539. [Google Scholar] [CrossRef] [PubMed]
  17. Salusky, I.B.; Coburn, J.W.; Brill, J.; Foley, J.; Slatopolsky, E.; Fine, R.N.; Goodman, W.G. Bone disease in pediatric patients undergoing dialysis with CAPD or CCPD. Kidney Int. 1988, 33, 975–982. [Google Scholar] [CrossRef] [Green Version]
  18. Malluche, H.H.; Mawad, H.; Monier-Faugere, M.C. The importance of bone health in end-stage renal disease: Out of the frying pan, into the fire? Nephrol. Dial. Transplant. 2004, 19 (Suppl. 1), i9–i13. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  19. Hsu, C.Y.; Chen, L.R.; Chen, K.H. Osteoporosis in Patients with Chronic Kidney Diseases: A Systemic Review. Int. J. Mol. Sci. 2020, 21, 6846. [Google Scholar] [CrossRef]
  20. Cannata-Andia, J.B.; Rodriguez Garcia, M.; Gomez Alonso, C. Osteoporosis and adynamic bone in chronic kidney disease. J. Nephrol. 2013, 26, 73–80. [Google Scholar] [CrossRef]
  21. Bover, J.; Urena-Torres, P.; Torregrosa, J.V.; Rodriguez-Garcia, M.; Castro-Alonso, C.; Gorriz, J.L.; Laiz Alonso, A.M.; Cigarrán, S.; Benito, S.; López-Báez, V.; et al. Osteoporosis, bone mineral density and CKD-MBD complex (I): Diagnostic considerations. Nefrologia 2018, 38, 476–490. [Google Scholar] [CrossRef] [PubMed]
  22. Cunningham, J. Posttransplantation bone disease. Transplantation 2005, 79, 629–634. [Google Scholar] [CrossRef] [PubMed]
  23. Cunningham, J.; Sprague, S.M.; Cannata-Andia, J.; Coco, M.; Cohen-Solal, M.; Fitzpatrick, L.; Goltzmann, D.; Lafage-Proust, M.-H.; Leonard, M.; Ott, S.; et al. Osteoporosis in chronic kidney disease. Am. J. Kidney Dis. 2004, 43, 566–571. [Google Scholar] [CrossRef]
  24. Barger-Lux, M.J.; Recker, R.R. Bone microstructure in osteoporosis: Transilial biopsy and histomorphometry. Top Magn. Reason. Imaging 2002, 13, 297–305. [Google Scholar] [CrossRef] [PubMed]
  25. Carbonara, C.E.M.; Dos Reis, L.M.; Quadros, K.R.D.S.; Roza, N.A.V.; Sano, R.; Carvalho, A.B.; Jorgetti, V.; De Oliveira, R.B. Renal osteodystrophy and clinical outcomes: Data from the Brazilian Registry of Bone Biopsies—REBRABO. J. Bras. Nefrol. 2020, 42, 138–146. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  26. Moe, S.; Drüeke, T.; Cunningham, J.; Goodman, W.; Martin, K.; Olgaard, K.; Ott, S.; Sprague, S.; Lameire, N.; Eknoyan, G. Definition, evaluation, and classification of renal osteodystrophy: A position statement from Kidney Disease: Improving Global Outcomes (KDIGO). Kidney Int. 2006, 69, 1945–1953. [Google Scholar] [CrossRef] [Green Version]
  27. Massry, S.G.; Coburn, J.W.; Chertow, G.M.; Hruska, K.; Langman, C.; Malluche, H.; Martin, K.; McCann, L.M.; McCarthy, J.T.; Moe, S.; et al. K/DOQI clinical practice guidelines for bone metabolism and disease in chronic kidney disease. Am. J. Kidney Dis. 2003, 42 (Suppl. 3), S1–S201. [Google Scholar]
  28. Sherrard, D.J.; Baylink, D.J.; Wergedal, J.E.; Maloney, N.A. Quantitative histological studies on the pathogenesis of uremic bone disease. J. Clin. Endocrinol. Metab. 1974, 39, 119–135. [Google Scholar] [CrossRef]
  29. Ellis, H.A.; Pierides, A.M.; Feest, T.G.; Ward, M.K.; Kerr, D.N. Histopathology of renal osteodystrophy with particular reference to the effects of 1alpha-hydroxyVitamin D3 in patients treated by long-term haemodialysis. Clin. Endocrinol. 1977, 7 (Suppl. 3), 1–8. [Google Scholar] [CrossRef]
  30. Feest, T.G.; Ward, M.K.; Ellis, H.A.; Conceicao, S.; Pierides, A.M.; Aird, E.; Simpson, W.; Cook, D.B.; Kerr, D.N. Renal bone disease--what is it and why does it happen? Clin. Endocrinol. 1977, 7, 19–23. [Google Scholar] [CrossRef]
  31. Spasovski, G.B.; Bervoets, A.R.; Behets, G.J.; Ivanovski, N.; Sikole, A.; Dams, G.; Couttenye, M.; De Broe, M.E.; D’Haese, P.C. Spectrum of renal bone disease in end-stage renal failure patients not yet on dialysis. Nephrol. Dial. Transplant. 2003, 18, 1159–1166. [Google Scholar] [CrossRef] [PubMed]
  32. Drueke, T.B.; Massy, Z.A. Changing bone patterns with progression of chronic kidney disease. Kidney Int. 2016, 89, 289–302. [Google Scholar] [CrossRef]
  33. Spasovski, G.B. Bone biopsy in the diagnosis of renal osteodystrophy. Prilozi 2004, 25, 83–93. [Google Scholar] [PubMed]
  34. Malluche, H.H.; Mawad, H.W.; Monier-Faugere, M.C. Renal osteodystrophy in the first decade of the new millennium: Analysis of 630 bone biopsies in black and white patients. J. Bone Miner. Res. 2010, 26, 1368–1376. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  35. Sprague, S.M.; Bellorin-Font, E.; Jorgetti, V.; Carvalho, A.B.; Malluche, H.H.; Ferreira, A.; D’Haese, P.C.; Drüeke, T.B.; Du, H.; Manley, T.; et al. Diagnostic Accuracy of Bone Turnover Markers and Bone Histology in Patients with CKD Treated by Dialysis. Am. J. Kidney Dis. 2016, 67, 559–566. [Google Scholar] [CrossRef] [PubMed]
  36. Couttenye, M.M.; D’Haese, P.C.; Van Hoof, V.O.; Lemoniatou, E.; Goodman, W.; Verpooten, G.A.; De Broe, M.E. Low serum levels of alkaline phosphatase of bone origin: A good marker of adynamic bone disease in haemodialysis patients. Nephrol. Dial. Transplant. 1996, 11, 1065–1072. [Google Scholar] [CrossRef] [PubMed]
  37. Rodriguez-Perez, J.C.; Plaza, C.; Torres, A.; Vega, N.; Anabitarte, A.; Fernandez, A.; Lorenzo, A.; Hortal, L.; Palop, L. Low turnover bone disease is the more common form of bone disease in CAPD patients. Adv. Perit. Dial. 1992, 8, 376–380. [Google Scholar]
  38. Sherrard, D.J.; Hercz, G.; Pei, Y.; Maloney, N.A.; Greenwood, C.; Manuel, A.; Saiphoo, C.; Fenton, S.S.; Segre, G.V. The spectrum of bone disease in end-stage renal failure--an evolving disorder. Kidney Int. 1993, 43, 436–442. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  39. Torres, A.; Lorenzo, V.; Hernandez, D.; Rodriguez, J.C.; Concepcion, M.T.; Rodriguez, A.P.; Hernández, A.; de Bonis, E.; Darias, E.; González-Posada, J.M.; et al. Bone disease in predialysis, hemodialysis, and CAPD patients: Evidence of a better bone response to PTH. Kidney Int. 1995, 47, 1434–1442. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  40. Hamdy, N.A.; Kanis, J.A.; Beneton, M.N.; Brown, C.B.; Juttmann, J.R.; Jordans, J.G.; Josse, S.; Meyrier, A.; Lins, R.L.; Fairey, I.T. Effect of alfacalcidol on natural course of renal bone disease in mild to moderate renal failure. BMJ 1995, 310, 358–363. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  41. Coen, G.; Ballanti, P.; Bonucci, E.; Calabria, S.; Costantini, S.; Ferrannini, M.; Giustini, M.; Giordano, R.; Nicolai, G.; Manni, M.; et al. Renal Osteodystrophy in Predialysis and Hemodialysis Patients: Comparison of Histologic Patterns and Diagnostic Predictivity of Intact PTH. Nephron 2002, 91, 103–111. [Google Scholar] [CrossRef]
  42. Coen, G.; Ballanti, P.; Bonucci, E.; Calabria, S.; Centorrino, M.; Fassino, V.; Manni, M.; Mantella, D.; Mazzaferro, S.; Napoletano, I.; et al. Bone markers in the diagnosis of low turnover osteodystrophy in haemodialysis patients. Nephrol. Dial. Transpl. 1998, 13, 2294–2302. [Google Scholar] [CrossRef] [Green Version]
  43. Ferreira, A.; Frazao, J.M.; Monier-Faugere, M.C.; Gil, C.; Galvao, J.; Oliveira, C.; Baldaia, J.; Rodrigues, I.; Santos, C.; Ribeiro, S. Effects of sevelamer hydrochloride and calcium carbonate on renal osteodystrophy in hemodialysis patients. J. Am. Soc. Nephrol. 2008, 19, 405–412. [Google Scholar] [CrossRef] [Green Version]
  44. Salam, S.; Gallagher, O.; Gossiel, F.; Paggiosi, M.; Khwaja, A.; Eastell, R. Diagnostic Accuracy of Biomarkers and Imaging for Bone Turnover in Renal Osteodystrophy. J. Am. Soc. Nephrol. 2018, 29, 1557–1565. [Google Scholar] [CrossRef] [Green Version]
  45. Malluche, H.; Ritz, E.; Lange, H. Bone histology in incipient and advanced renal failure. Kidney Int. 1976, 9, 355–362. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  46. Tomiyama, C.; Carvalho, A.B.; Higa, A.; Jorgetti, V.; Draibe, S.A.; Canziani, M.E. Coronary calcification is associated with lower bone formation rate in CKD patients not yet in dialysis treatment. J. Bone Miner. Res. 2010, 25, 499–504. [Google Scholar] [CrossRef]
  47. Graciolli, F.G.; Neves, K.R.; Barreto, F.; Barreto, D.V.; Dos Reis, L.; Canziani, M.E.; Sabbagh, Y.; Carvalho, A.B.; Jorgetti, V.; Elias, R.M.; et al. The complexity of chronic kidney disease-mineral and bone disorder across stages of chronic kidney disease. Kidney Int. 2017, 91, 1436–1446. [Google Scholar] [CrossRef] [PubMed]
  48. El-Husseini, A.; Abdalbary, M.; Lima, F.; Issa, M.; Ahmed, M.-T.; Winkler, M.; Srour, H.; Davenport, D.; Wang, G.; Faugere, M.-C.; et al. Low Turnover Renal Osteodystrophy with Abnormal Bone Quality and Vascular Calcification in Patients With Mild-to-Moderate CKD. Kidney Int. Rep. 2022, 7, 1016–1026. [Google Scholar] [CrossRef]
  49. Coen, G.; Mazzaferro, S.; Bonucci, E.; Ballanti, P.; Massimetti, C.; Donato, G.; Landi, A.; Smacchi, A.; Della Rocca, C.; Cinotti, G.A.; et al. Treatment of secondary hyperparathyroidism of predialysis chronic renal failure with low doses of 1,25(OH)2D3: Humoral and histomorphometric results. Miner. Electrolyte Metab. 1986, 12, 375–382. [Google Scholar] [PubMed]
  50. Sperschneider, H.; Humbsch, K.; Abendroth, K. Oral calcitriol pulse therapy in hemodialysis patients. Effects on histomorphometry of bone in renal hyperparathyroidism. Med. Klin. 1997, 92, 597–603. [Google Scholar] [CrossRef]
  51. Behets, G.J.; Spasovski, G.; Sterling, L.R.; Goodman, W.G.; Spiegel, D.M.; De Broe, M.E.; D’Haese, P.C. Bone histomorphometry before and after long-term treatment with cinacalcet in dialysis patients with secondary hyperparathyroidism. Kidney Int. 2015, 87, 846–856. [Google Scholar] [CrossRef] [Green Version]
  52. Malluche, H.; Monier-Faugere, M.-C.; Wang, G.; O, J.M.F.; Charytan, C.; Coburn, J.; Coyne, D.; Kaplan, M.; Baker, N.; McCary, L.; et al. An assessment of cinacalcet HCl effects on bone histology in dialysis patients with secondary hyperparathyroidism. Clin. Nephrol. 2008, 69, 269–278. [Google Scholar] [CrossRef]
  53. Cejka, D.; Herberth, J.; Branscum, A.J.; Fardo, D.W.; Monier-Faugere, M.C.; Diarra, D.; Haas, M.; Malluche, H.H. Sclerostin and Dickkopf-1 in renal osteodystrophy. Clin. J. Am. Soc. Nephrol. 2011, 6, 877–882. [Google Scholar] [CrossRef] [PubMed]
  54. Slatopolsky, E.; Finch, J.; Clay, P.; Martin, D.; Sicard, G.; Singer, G.; Gao, P.; Cantor, T.; Dusso, A. A novel mechanism for skeletal resistance in uremia. Kidney Int. 2000, 58, 753–761. [Google Scholar] [CrossRef] [Green Version]
  55. Pelletier, S.; Dubourg, L.; Carlier, M.C.; Hadj-Aissa, A.; Fouque, D. The relation between renal function and serum sclerostin in adult patients with CKD. Clin. J. Am. Soc. Nephrol. 2013, 8, 819–823. [Google Scholar] [CrossRef] [Green Version]
  56. Evenepoel, P.; D’Haese, P.; Brandenburg, V. Sclerostin and DKK1: New players in renal bone and vascular disease. Kidney Int. 2015, 88, 235–240. [Google Scholar] [CrossRef]
  57. Sabbagh, Y.; Graciolli, F.G.; O’Brien, S.; Tang, W.; dos Reis, L.M.; Ryan, S.; Phillips, L.; Boulanger, J.; Song, W.; Bracken, C.; et al. Repression of osteocyte Wnt/beta-catenin signaling is an early event in the progression of renal osteodystrophy. J. Bone Miner. Res. 2012, 27, 1757–1772. [Google Scholar] [CrossRef] [PubMed]
  58. Bervoets, A.R.; Spasovski, G.B.; Behets, G.J.; Dams, G.; Polenakovic, M.H.; Zafirovska, K.; Van Hoof, V.O.; De Broe, M.E.; D’Haese, P.C. Useful biochemical markers for diagnosing renal osteodystrophy in predialysis end-stage renal failure patients. Am. J. Kidney Dis. 2003, 41, 997–1007. [Google Scholar] [CrossRef] [PubMed]
  59. Barreto, F.C.; Barreto, D.V.; Canziani, M.E.; Tomiyama, C.; Higa, A.; Mozar, A.; Glorieux, G.; Vanholder, R.; A Massy, Z.; De Carvalho, A.B. Association between indoxyl sulfate and bone histomorphometry in pre-dialysis chronic kidney disease patients. J. Bras. Nefrol. 2014, 36, 289–296. [Google Scholar] [CrossRef] [Green Version]
  60. Lima, F.; El-Husseini, A.; Monier-Faugere, M.C.; David, V.; Mawad, H.; Quarles, D.; Malluche, H.H. FGF-23 serum levels and bone histomorphometric results in adult patients with chronic kidney disease on dialysis. Clin. Nephrol. 2014, 82, 287. [Google Scholar] [CrossRef] [Green Version]
  61. Cianciolo, G.; La Manna, G.; Della Bella, E.; Cappuccilli, M.L.; Angelini, M.L.; Dormi, A.; Capelli, I.; Laterza, C.; Costa, R.; Alviano, F.; et al. Effect of Vitamin D receptor activator therapy on Vitamin D receptor and osteocalcin expression in circulating endothelial progenitor cells of hemodialysis patients. Blood Purif. 2013, 35, 187–195. [Google Scholar] [CrossRef] [PubMed]
  62. Slatopolsky, E.; Gonzalez, E.; Martin, K. Pathogenesis and treatment of renal osteodystrophy. Blood Purif. 2003, 21, 318–326. [Google Scholar] [CrossRef]
  63. Slatopolsky, E. The role of calcium, phosphorus and Vitamin D metabolism in the development of secondary hyperparathyroidism. Nephrol. Dial Transplant. 1998, 13, 3–8. [Google Scholar] [CrossRef]
  64. Hruska, K.A.; Sugatani, T.; Agapova, O.; Fang, Y. The chronic kidney disease—Mineral bone disorder (CKD-MBD): Advances in pathophysiology. Bone 2017, 100, 80–86. [Google Scholar] [CrossRef]
  65. Cannata-Andia, J.B.; Martin-Carro, B.; Martin-Virgala, J.; Rodriguez-Carrio, J.; Bande-Fernandez, J.J.; Alonso-Montes, C.; Carrillo-López, N. Chronic Kidney Disease-Mineral and Bone Disorders: Pathogenesis and Management. Calcif. Tissue Int. 2021, 108, 410–422. [Google Scholar] [CrossRef] [PubMed]
  66. Rodriguez-Ortiz, M.E.; Rodriguez, M. Recent advances in understanding and managing secondary hyperparathyroidism in chronic kidney disease. F1000Res 2020, 9, F1000. [Google Scholar] [CrossRef] [PubMed]
  67. Isakova, T.; on behalf of the Chronic Renal Insufficiency Cohort (CRIC) Study Group; Wahl, P.; Vargas, G.S.; Gutiérrez, O.M.; Scialla, J.; Xie, H.; Appleby, D.; Nessel, L.; Bellovich, K.; et al. Fibroblast growth factor 23 is elevated before parathyroid hormone and phosphate in chronic kidney disease. Kidney Int. 2011, 79, 1370–1378. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  68. Gogusev, J.; Duchambon, P.; Hory, B.; Giovannini, M.; Goureau, Y.; Sarfati, E.; Drüeke, T.B. Depressed expression of calcium receptor in parathyroid gland tissue of patients with hyperparathyroidism. Kidney Int. 1997, 51, 328–336. [Google Scholar] [CrossRef] [Green Version]
  69. Rodriguez, M.; Nemeth, E.; Martin, D. The calcium-sensing receptor: A key factor in the pathogenesis of secondary hyperparathyroidism. Am. J. Physiol. Renal. Physiol. 2005, 288, F253–F264. [Google Scholar] [CrossRef] [Green Version]
  70. Centeno, P.P.; Herberger, A.; Mun, H.-C.; Tu, C.; Nemeth, E.F.; Chang, W.; Conigrave, A.D.; Ward, D.T. Phosphate acts directly on the calcium-sensing receptor to stimulate parathyroid hormone secretion. Nat. Commun. 2019, 10, 4693. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  71. Slatopolsky, E.; Brown, A.; Dusso, A. Pathogenesis of secondary hyperparathyroidism. Kidney Int. Suppl. 1999, 73, S14–S19. [Google Scholar] [CrossRef] [Green Version]
  72. Slatopolsky, E.; Brown, A.; Dusso, A. Role of phosphorus in the pathogenesis of secondary hyperparathyroidism. Am. J. Kidney Dis. 2001, 37, S54–S57. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  73. Martin, K.J.; Floege, J.; Ketteler, M. Bone and Mineral Disorders in Chronic Kidney Disease. In Comprehensive Clinical Nephrology, 6th ed.; Feehally, J., Ed.; Springer: Berlin/Heidelberg, Germany, 2019. [Google Scholar]
  74. Cianciolo, G.; La Manna, G.; Donati, G.; Dormi, A.; Cappuccilli, M.L.; Cuna, V.; Legnani, C.; Palareti, G.; Colì, L.; Stefoni, S. Effects of unfractioned heparin and low-molecular-weight heparin on osteoprotegerin and RANKL plasma levels in haemodialysis patients. Nephrol. Dial. Transpl. 2011, 26, 646–652. [Google Scholar] [CrossRef] [PubMed]
  75. Asci, G.; Ok, E.; Savas, R.; Ozkahya, M.; Duman, S.; Toz, H.; Kayikcioglu, M.; Branscum, A.J.; Monier-Faugere, M.-C.; Herberth, J.; et al. The link between bone and coronary calcifications in CKD-5 patients on haemodialysis. Nephrol. Dial Transpl. 2011, 26, 1010–1015. [Google Scholar] [CrossRef] [Green Version]
  76. Fang, Y.; Ginsberg, C.; Seifert, M.; Agapova, O.; Sugatani, T.; Register, T.C.; Freedman, B.I.; Monier-Faugere, M.-C.; Malluche, H.; Hruska, K.A. CKD-induced wingless/integration1 inhibitors and phosphorus cause the CKD-mineral and bone disorder. J. Am. Soc. Nephrol. 2014, 25, 1760–1773. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  77. Case records of the Massachusetts General Hospital. Weekly clinicopathological exercises. Case 44-1986. An 80-year-old woman with Paget’s disease and severe hypercalcemia after a recent fracture. N. Engl. J. Med. 1986, 315, 1209–1219. [Google Scholar] [CrossRef]
  78. Elias, R.M.; Dalboni, M.A.; Coelho, A.C.E.; Moyses, R.M.A. CKD-MBD: From the Pathogenesis to the Identification and Development of Potential Novel Therapeutic Targets. Curr. Osteoporos. Rep. 2018, 16, 693–702. [Google Scholar] [CrossRef] [PubMed]
  79. Pereira, R.C.; Juppner, H.; Azucena-Serrano, C.E.; Yadin, O.; Salusky, I.B.; Wesseling-Perry, K. Patterns of FGF-23, DMP1, and MEPE expression in patients with chronic kidney disease. Bone 2009, 45, 1161–1168. [Google Scholar] [CrossRef] [Green Version]
  80. Wesseling-Perry, K.; Pereira, R.C.; Wang, H.; Elashoff, R.M.; Sahney, S.; Gales, B.; Juppner, H.; Salusky, I.B. Relationship between plasma fibroblast growth factor-23 concentration and bone mineralization in children with renal failure on peritoneal dialysis. J. Clin. Endocrinol. Metab. 2009, 94, 511–517. [Google Scholar] [CrossRef]
  81. Lavi-Moshayoff, V.; Wasserman, G.; Meir, T.; Silver, J.; Naveh-Many, T. PTH increases FGF23 gene expression and mediates the high-FGF23 levels of experimental kidney failure: A bone parathyroid feedback loop. Am. J. Physiol. Renal Physiol. 2010, 299, F882–F889. [Google Scholar] [CrossRef] [Green Version]
  82. Ferreira, J.C.; Ferrari, G.O.; Neves, K.R.; Cavallari, R.T.; Dominguez, W.V.; Dos Reis, L.M.; Graciolli, F.G.; Oliveira, E.C.; Liu, S.; Sabbagh, Y.; et al. Effects of dietary phosphate on adynamic bone disease in rats with chronic kidney disease—Role of sclerostin? PLoS ONE 2013, 8, e79721. [Google Scholar] [CrossRef] [Green Version]
  83. Roforth, M.M.; Fujita, K.; McGregor, U.I.; Kirmani, S.; McCready, L.K.; Peterson, J.M.; Drake, M.T.; Monroe, D.G.; Khosla, S. Effects of age on bone mRNA levels of sclerostin and other genes relevant to bone metabolism in humans. Bone 2014, 59, 1–6. [Google Scholar] [CrossRef] [Green Version]
  84. Keller, H.; Kneissel, M. SOST is a target gene for PTH in bone. Bone 2005, 37, 148–158. [Google Scholar] [CrossRef]
  85. Kulkarni, N.H.; Halladay, D.L.; Miles, R.R.; Gilbert, L.M.; Frolik, C.A.; Galvin, R.J.; Gillespie, M.; Onyia, J. Effects of parathyroid hormone on Wnt signaling pathway in bone. J. Cell Biochem. 2005, 95, 1178–1190. [Google Scholar] [CrossRef]
  86. Santos, M.F.P.; Hernandez, M.J.; de Oliveira, I.B.; Siqueira, F.R.; Dominguez, W.V.; dos Reis, L.M.; Carvalho, A.B.; Moysés, R.M.A.; Jorgetti, V. Comparison of clinical, biochemical and histomorphometric analysis of bone biopsies in dialysis patients with and without fractures. J. Bone Miner. Metab. 2019, 37, 125–133. [Google Scholar] [CrossRef] [PubMed]
  87. Haarhaus, M.; Evenepoel, P.; European Renal Osteodystrophy workgroup; Chronic Kidney Disease Mineral and Bone Disorder (CKD-MBD) working group of the European Renal Association–European Dialysis and Transplant Association (ERA-EDTA). Differentiating the causes of adynamic bone in advanced chronic kidney disease informs osteoporosis treatment. Kidney Int. 2021, 100, 546–558. [Google Scholar] [CrossRef] [PubMed]
  88. Levin, A.; Bakris, G.L.; Molitch, M.; Smulders, M.; Tian, J.; Williams, L.A.; Andress, D.L. Prevalence of abnormal serum vitamin, D.; PTH, calcium, and phosphorus in patients with chronic kidney disease: Results of the study to evaluate early kidney disease. Kidney Int. 2007, 71, 31–38. [Google Scholar] [CrossRef] [Green Version]
  89. Moranne, O.; Froissart, M.; Rossert, J.; Gauci, C.; Boffa, J.J.; Haymann, J.P.; Ben M’Rad, M.; Jacquot, C.; Houillier, P.; Stengel, B.; et al. Timing of onset of CKD-related metabolic complications. J. Am. Soc. Nephrol. 2009, 20, 164–171. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  90. Lehmann, G.; Ott, U.; Kaemmerer, D.; Schuetze, J.; Wolf, G. Bone histomorphometry and biochemical markers of bone turnover in patients with chronic kidney disease Stages 3–5. Clin. Nephrol. 2008, 70, 296–305. [Google Scholar] [CrossRef] [PubMed]
  91. Beaubrun, A.C.; Kilpatrick, R.D.; Freburger, J.K.; Bradbury, B.D.; Wang, L.; Brookhart, M.A. Temporal trends in fracture rates and postdischarge outcomes among hemodialysis patients. J. Am. Soc. Nephrol. 2013, 24, 1461–1469. [Google Scholar] [CrossRef] [Green Version]
  92. Moe, S.M.; Nickolas, T.L. Fractures in Patients with CKD: Time for Action. Clin. J. Am. Soc. Nephrol. 2016, 11, 1929–1931. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  93. Bover, J.; Urena-Torres, P.; Cozzolino, M.; Rodriguez-Garcia, M.; Gomez-Alonso, C. The Non-invasive Diagnosis of Bone Disorders in CKD. Calcif. Tissue Int. 2021, 108, 512–527. [Google Scholar] [CrossRef]
  94. Lv, J.C.; Zhang, L.X. Prevalence and Disease Burden of Chronic Kidney Disease. Adv. Exp. Med. Biol. 2019, 1165, 3–15. [Google Scholar]
  95. Bello, A.K.; Alrukhaimi, M.; Ashuntantang, G.E.; Bellorin-Font, E.; Benghanem Gharbi, M.; Braam, B.; Feehally, J.; Harris, D.C.; Jha, V.; Jindal, K.; et al. Global overview of health systems oversight and financing for kidney care. Kidney Int. Suppl. 2018, 8, 41–51. [Google Scholar] [CrossRef] [PubMed]
  96. Tentori, F.; McCullough, K.; Kilpatrick, R.D.; Bradbury, B.D.; Robinson, B.M.; Kerr, P.G.; Pisoni, R.L. High rates of death and hospitalization follow bone fracture among hemodialysis patients. Kidney Int. 2014, 85, 166–173. [Google Scholar] [CrossRef] [Green Version]
  97. Naylor, K.L.; McArthur, E.; Leslie, W.D.; Fraser, L.A.; Jamal, S.A.; Cadarette, S.M.; Pouget, J.G.; Lok, C.E.; Hodsman, A.B.; Adachi, J.D.; et al. The three-year incidence of fracture in chronic kidney disease. Kidney Int. 2014, 86, 810–818. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  98. Klawansky, S.; Komaroff, E.; Cavanaugh, P.F.; Mitchell, D.Y., Jr.; Gordon, M.J.; Connelly, J.E.; Ross, S.D. Relationship between age, renal function and bone mineral density in the US population. Osteoporos. Int. 2003, 14, 570–576. [Google Scholar] [CrossRef] [PubMed]
  99. Jadoul, M.; Albert, J.M.; Akiba, T.; Akizawa, T.; Arab, L.; Bragg-Gresham, J.L.; Mason, N.; Prutz, K.G.; Young, E.W.; Pisoni, R.L. Incidence and risk factors for hip or other bone fractures among hemodialysis patients in the Dialysis Outcomes and Practice Patterns Study. Kidney Int. 2006, 70, 1358–1366. [Google Scholar] [CrossRef] [Green Version]
  100. Danese, M.D.; Kim, J.; Doan, Q.V.; Dylan, M.; Griffiths, R.; Chertow, G.M. PTH and the risks for hip, vertebral, and pelvic fractures among patients on dialysis. Am. J. Kidney Dis. 2006, 47, 149–156. [Google Scholar] [CrossRef]
  101. Dukas, L.; Schacht, E.; Stahelin, H.B. In elderly men and women treated for osteoporosis a low creatinine clearance of <65 ml/min is a risk factor for falls and fractures. Osteoporos Int. 2005, 16, 1683–1690. [Google Scholar]
  102. Naylor, K.L.; Leslie, W.D.; Hodsman, A.B.; Rush, D.N.; Garg, A.X. FRAX predicts fracture risk in kidney transplant recipients. Transplantation 2014, 97, 940–945. [Google Scholar] [CrossRef]
  103. Yamamoto, S.; Kido, R.; Onishi, Y.; Fukuma, S.; Akizawa, T.; Fukagawa, M.; Kazama, J.J.; Narita, I.; Fukuhara, S. Use of renin-angiotensin system inhibitors is associated with reduction of fracture risk in hemodialysis patients. PLoS ONE 2015, 10, e0122691. [Google Scholar] [CrossRef] [Green Version]
  104. Malluche, H.H.; Porter, D.S.; Monier-Faugere, M.C.; Mawad, H.; Pienkowski, D. Differences in bone quality in low- and high-turnover renal osteodystrophy. J. Am. Soc. Nephrol. 2012, 23, 525–532. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  105. Ginsberg, C.; Ix, J.H. Diagnosis and Management of Osteoporosis in Advanced Kidney Disease: A Review. Am. J. Kidney Dis. 2022, 79, 427–436. [Google Scholar] [CrossRef]
  106. Tentori, F.; Albert, J.M.; Young, E.W.; Blayney, M.J.; Robinson, B.M.; Pisoni, R.L.; Akiba, T.; Greenwood, R.N.; Kimata, N.; Levin, N.W.; et al. The survival advantage for haemodialysis patients taking Vitamin D is questioned: Findings from the Dialysis Outcomes and Practice Patterns Study. Nephrol. Dial. Transpl. 2009, 24, 963–972. [Google Scholar] [CrossRef] [PubMed]
  107. Urena, P.; Bernard-Poenaru, O.; Ostertag, A.; Baudoin, C.; Cohen-Solal, M.; Cantor, T.; De Vernejoul, M.C. Bone mineral density, biochemical markers and skeletal fractures in haemodialysis patients. Nephrol. Dial. Transpl. 2003, 18, 2325–2331. [Google Scholar] [CrossRef] [Green Version]
  108. Malluche, H.H.; Davenport, D.L.; Cantor, T.; Monier-Faugere, M.C. Bone mineral density and serum biochemical predictors of bone loss in patients with CKD on dialysis. Clin. J. Am. Soc. Nephrol. 2014, 9, 1254–1262. [Google Scholar] [CrossRef] [Green Version]
  109. Ketteler, M.; Block, G.A.; Evenepoel, P.; Fukagawa, M.; Herzog, C.A.; McCann, L.; Moe, S.M.; Shroff, R.; Tonelli, M.A.; Toussaint, N.D.; et al. Executive summary of the 2017 KDIGO Chronic Kidney Disease-Mineral and Bone Disorder (CKD-MBD) Guideline Update: What’s changed and why it matters. Kidney Int. 2017, 92, 26–36. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  110. Kidney Disease: Improving Global Outcomes CKDMBDUWG. KDIGO 2017 Clinical Practice Guideline Update for the Diagnosis, Evaluation, Prevention, and Treatment of Chronic Kidney Disease-Mineral and Bone Disorder (CKD-MBD). Kidney Int. Suppl. 2017, 7, 1–59. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  111. Iimori, S.; Mori, Y.; Akita, W.; Kuyama, T.; Takada, S.; Asai, T.; Kuwahara, M.; Sasaki, S.; Tsukamoto, Y. Diagnostic usefulness of bone mineral density and biochemical markers of bone turnover in predicting fracture in CKD stage 5D patients—A single-center cohort study. Nephrol. Dial. Transpl. 2012, 27, 345–351. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  112. Yenchek, R.H.; Ix, J.H.; Shlipak, M.G.; Bauer, D.C.; Rianon, N.J.; Kritchevsky, S.; Harris, T.B.; Netwman, A.B.; A Cauley, J.; Fried, L.F. Bone Mineral Density and Fracture Risk in Older Individuals with CKD. Clin. J. Am. Soc. Nephrol. 2012, 7, 1130–1136. [Google Scholar] [CrossRef] [Green Version]
  113. Naylor, K.L.; Garg, A.X.; Zou, G.; Langsetmo, L.; Leslie, W.D.; Fraser, L.A.; Adachi, J.D.; Morin, S.; Goltzman, D.; Lentle, B.; et al. Comparison of fracture risk prediction among individuals with reduced and normal kidney function. Clin. J. Am. Soc. Nephrol. 2015, 10, 646–653. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  114. Poiana, C.; Dusceac, R.; Niculescu, D.A. Utility of Trabecular Bone Score (TBS) in Bone Quality and Fracture Risk Assessment in Patients on Maintenance Dialysis. Front. Med. 2021, 8, 782837. [Google Scholar] [CrossRef] [PubMed]
  115. Silva, B.C.; Leslie, W.D.; Resch, H.; Lamy, O.; Lesnyak, O.; Binkley, N.; McCloskey, E.V.; Kanis, J.A.; Bilezikian, J.P. Trabecular bone score: A noninvasive analytical method based upon the DXA image. J. Bone Miner. Res. 2014, 29, 518–530. [Google Scholar] [CrossRef]
  116. Abdalbary, M.; Sobh, M.; Elnagar, S.; Elhadedy, M.A.; Elshabrawy, N.; Abdelsalam, M.; Asadipooya, K.; Sabry, A.; Halawa, A.; El-Husseini, A. Management of osteoporosis in patients with chronic kidney disease. Osteoporos. Int. 2022, 33, 2259–2274. [Google Scholar] [CrossRef] [PubMed]
  117. Ramalho, J.; Marques, I.D.B.; Hans, D.; Dempster, D.; Zhou, H.; Patel, P.; Pereira, R.M.R.; Jorgetti, V.; Moyses, R.M.A.; Nickolas, T.L. The trabecular bone score: Relationships with trabecular and cortical microarchitecture measured by HR-pQCT and histomorphometry in patients with chronic kidney disease. Bone 2018, 116, 215–220. [Google Scholar] [CrossRef] [PubMed]
  118. Nickolas, T.L.; Stein, E.M.; Dworakowski, E.; Nishiyama, K.K.; Komandah-Kosseh, M.; Zhang, C.A.; McMahon, D.J.; Liu, X.S.; Boutroy, S.; Cremers, S.; et al. Rapid cortical bone loss in patients with chronic kidney disease. J. Bone Miner. Res. 2013, 28, 1811–1820. [Google Scholar] [CrossRef] [PubMed]
  119. Negri, A.L.; Del Valle, E.E.; Zanchetta, M.B.; Nobaru, M.; Silveira, F.; Puddu, M.; Barone, R.; Bogado, C.E.; Zanchetta, J.R. Evaluation of bone microarchitecture by high-resolution peripheral quantitative computed tomography (HR-pQCT) in hemodialysis patients. Osteoporos. Int. 2012, 23, 2543–2550. [Google Scholar] [CrossRef]
  120. Drechsler, C.; Verduijn, M.; Pilz, S.; Krediet, R.T.; Dekker, F.W.; Wanner, C.; Ketteler, M.; Boeschoten, E.W.; Brandenburg, V.; NECOSAD Study Group. Bone alkaline phosphatase and mortality in dialysis patients. Clin. J. Am. Soc. Nephrol. 2011, 6, 1752–1759. [Google Scholar] [CrossRef] [Green Version]
  121. Kobayashi, I.; Shidara, K.; Okuno, S.; Yamada, S.; Imanishi, Y.; Mori, K.; Ishimura, E.; Shoji, S.; Yamakawa, T.; Inaba, M. Higher serum bone alkaline phosphatase as a predictor of mortality in male hemodialysis patients. Life Sci. 2012, 90, 212–218. [Google Scholar] [CrossRef]
  122. Evenepoel, P.; Cavalier, E.; D’Haese, P.C. Biomarkers Predicting Bone Turnover in the Setting of CKD. Curr. Osteoporos. Rep. 2017, 15, 178–186. [Google Scholar] [CrossRef] [PubMed]
  123. Fletcher, S.; Jones, R.G.; Rayner, H.C.; Harnden, P.; Hordon, L.D.; Aaron, J.E.; Oldroyd, B.; Brownjohn, A.M.; Turney, J.H.; Smith, M.A. Assessment of renal osteodystrophy in dialysis patients: Use of bone alkaline phosphatase, bone mineral density and parathyroid ultrasound in comparison with bone histology. Nephron 1997, 75, 412–419. [Google Scholar] [CrossRef]
  124. Coen, G.; Ballanti, P.; Balducci, A.; Calabria, S.; Fischer, M.S.; Jankovic, L.; Manni, M.; Morosetti, M.; Moscaritolo, E.; Sardella, D.; et al. Serum osteoprotegerin and renal osteodystrophy. Nephrol. Dial. Transpl. 2002, 17, 233–238. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  125. Barreto, F.C.; Barreto, D.V.; Moyses, R.M.; Neves, K.R.; Canziani, M.E.; Draibe, S.A.; Jorgetti, V.; Carvalho, A.B. K/DOQI-recommended intact PTH levels do not prevent low-turnover bone disease in hemodialysis patients. Kidney Int. 2008, 73, 771–777. [Google Scholar] [CrossRef]
  126. Lehmann, G.; Stein, G.; Huller, M.; Schemer, R.; Ramakrishnan, K.; Goodman, W.G. Specific measurement of PTH (1-84) in various forms of renal osteodystrophy (ROD) as assessed by bone histomorphometry. Kidney Int. 2005, 68, 1206–1214. [Google Scholar] [CrossRef] [PubMed]
  127. Haarhaus, M.; Monier-Faugere, M.C.; Magnusson, P.; Malluche, H.H. Bone alkaline phosphatase isoforms in hemodialysis patients with low versus non-low bone turnover: A diagnostic test study. Am. J. Kidney Dis. 2015, 66, 99–105. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  128. Marques, I.D.; Araujo, M.J.; Graciolli, F.G.; Reis, L.M.; Pereira, R.M.; Custodio, M.R.; Jorgetti, V.; Elias, R.M.; David-Neto, E.; Moysés, R.M.A. Biopsy vs. peripheral computed tomography to assess bone disease in CKD patients on dialysis: Differences and similarities. Osteoporos. Int. 2017, 28, 1675–1683. [Google Scholar] [CrossRef]
  129. Grotz, W.H.; Mundinger, F.A.; Gugel, B.; Exner, V.; Kirste, G.; Schollmeyer, P.J. Bone fracture and osteodensitometry with dual energy X-ray absorptiometry in kidney transplant recipients. Transplantation 1994, 58, 912–915. [Google Scholar] [CrossRef]
  130. Nikkel, L.E.; Hollenbeak, C.S.; Fox, E.J.; Uemura, T.; Ghahramani, N. Risk of fractures after renal transplantation in the United States. Transplantation 2009, 87, 1846–1851. [Google Scholar] [CrossRef]
  131. O’Shaughnessy, E.A.; Dahl, D.C.; Smith, C.L.; Kasiske, B.L. Risk factors for fractures in kidney transplantation. Transplantation 2002, 74, 362–366. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  132. Khairallah, P.; Nickolas, T.L. Bone and Mineral Disease in Kidney Transplant Recipients. Clin. J. Am. Soc. Nephrol. 2022, 17, 121–130. [Google Scholar] [CrossRef]
  133. Patel, S.; Kwan, J.T.; McCloskey, E.; McGee, G.; Thomas, G.; Johnson, D.; Wills, R.; Ogunremi, L.; Barron, J. Prevalence and causes of low bone density and fractures in kidney transplant patients. J. Bone Miner. Res. 2001, 16, 1863–1870. [Google Scholar] [CrossRef] [PubMed]
  134. Grotz, W.H.; Mundinger, F.A.; Gugel, B.; Exner, V.M.; Kirste, G.; Schollmeyer, P.J. Bone mineral density after kidney transplantation. A cross-sectional study in 190 graft recipients up to 20 years after transplantation. Transplantation 1995, 59, 982–986. [Google Scholar] [CrossRef] [PubMed]
  135. Durieux, S.; Mercadal, L.; Orcel, P.; Dao, H.; Rioux, C.; Bernard, M.; Rozenberg, S.; Barrou, B.; Bourgeois, P.; Deray, G.; et al. Bone mineral density and fracture prevalence in long-term kidney graft recipients. Transplantation 2002, 74, 496–500. [Google Scholar] [CrossRef]
  136. Julian, B.A.; Laskow, D.A.; Dubovsky, J.; Dubovsky, E.V.; Curtis, J.J.; Quarles, L.D. Rapid loss of vertebral mineral density after renal transplantation. N. Engl. J. Med. 1991, 325, 544–550. [Google Scholar] [CrossRef] [PubMed]
  137. Evenepoel, P.; Meijers, B.K.; de Jonge, H.; Naesens, M.; Bammens, B.; Claes, K.; Kuypers, D.; Vanrenterghem, Y. Recovery of hyperphosphatoninism and renal phosphorus wasting one year after successful renal transplantation. Clin. J. Am. Soc. Nephrol. 2008, 3, 1829–1836. [Google Scholar] [CrossRef] [Green Version]
  138. Perrin, P.; Caillard, S.; Javier, R.M.; Braun, L.; Heibel, F.; Borni-Duval, C.; Muller, C.; Olagne, J.; Moulin, B. Persistent hyperparathyroidism is a major risk factor for fractures in the five years after kidney transplantation. Am. J. Transplant. 2013, 13, 2653–2663. [Google Scholar] [CrossRef] [PubMed]
  139. Lou, I.; Foley, D.; Odorico, S.K.; Leverson, G.; Schneider, D.F.; Sippel, R.; Chen, H. How Well Does Renal Transplantation Cure Hyperparathyroidism? Ann. Surg. 2015, 262, 653–659. [Google Scholar] [CrossRef] [Green Version]
  140. Cianciolo, G.; Cozzolino, M. FGF23 in kidney transplant: The strange case of Doctor Jekyll and Mister Hyde. Clin. Kidney J. 2016, 9, 665–668. [Google Scholar] [CrossRef] [Green Version]
  141. Lobo, P.I.; Cortez, M.S.; Stevenson, W.; Pruett, T.L. Normocalcemic hyperparathyroidism associated with relatively low 1:25 Vitamin D levels post-renal transplant can be successfully treated with oral calcitriol. Clin. Transplant. 1995, 9, 277–281. [Google Scholar]
  142. Iyer, S.P.; Nikkel, L.E.; Nishiyama, K.K.; Dworakowski, E.; Cremers, S.; Zhang, C.; McMahon, D.J.; Boutroy, S.; Liu, X.S.; Ratner, L.E.; et al. Kidney transplantation with early corticosteroid withdrawal: Paradoxical effects at the central and peripheral skeleton. J. Am. Soc. Nephrol. 2014, 25, 1331–1341. [Google Scholar] [CrossRef] [Green Version]
  143. Batteux, B.; Gras-Champel, V.; Lando, M.; Brazier, F.; Mentaverri, R.; Desailly-Henry, I.; Rey, A.; Bennis, Y.; Masmoudi, K.; Choukroun, G.; et al. Early steroid withdrawal has a positive effect on bone in kidney transplant recipients: A propensity score study with inverse probability-of-treatment weighting. Ther. Adv. Musculoskelet. Dis. 2020, 12, 1759720X20953357. [Google Scholar] [CrossRef]
  144. Nikkel, L.E.; Mohan, S.; Zhang, A.; McMahon, D.J.; Boutroy, S.; Dube, G.; Ratner, L.; Hollenbeak, C.S.; Leonard, M.B.; Shane, E.; et al. Reduced fracture risk with early corticosteroid withdrawal after kidney transplant. Am. J. Transplant. 2012, 12, 649–659. [Google Scholar] [CrossRef] [Green Version]
  145. Evenepoel, P.; Claes, K.; Meijers, B.; Laurent, M.R.; Bammens, B.; Naesens, M.; Sprangers, B.; Pottel, H.; Cavalier, E.; Kuypers, D. Bone mineral density, bone turnover markers, and incident fractures in de novo kidney transplant recipients. Kidney Int. 2019, 95, 1461–1470. [Google Scholar] [CrossRef] [PubMed]
  146. Mikuls, T.R.; Julian, B.A.; Bartolucci, A.; Saag, K.G. Bone mineral density changes within six months of renal transplantation. Transplantation 2003, 75, 49–54. [Google Scholar] [CrossRef] [PubMed]
  147. Almond, M.K.; Kwan, J.T.; Evans, K.; Cunningham, J. Loss of regional bone mineral density in the first 12 months following renal transplantation. Nephron 1994, 66, 52–57. [Google Scholar] [CrossRef] [PubMed]
  148. Carlini, R.G.; Rojas, E.; Weisinger, J.R.; Lopez, M.; Martinis, R.; Arminio, A.; Bellorin-Font, E. Bone disease in patients with long-term renal transplantation and normal renal function. Am. J. Kidney Dis. 2000, 36, 160–166. [Google Scholar] [CrossRef] [PubMed]
  149. Brandenburg, V.M.; Ketteler, M.; Heussen, N.; Politt, D.; Frank, R.D.; Westenfeld, R.; Ittel, T.H.; Floege, J. Lumbar bone mineral density in very long-term renal transplant recipients: Impact of circulating sex hormones. Osteoporos. Int. 2005, 16, 1611–1620. [Google Scholar] [CrossRef] [PubMed]
  150. Iseri, K.; Carrero, J.J.; Evans, M.; Fellander-Tsai, L.; Berg, H.E.; Runesson, B.; Stenvinkel, P.; Lindholm, B.; Qureshi, A.R. Fractures after kidney transplantation: Incidence, predictors, and association with mortality. Bone 2020, 140, 115554. [Google Scholar] [CrossRef]
  151. Jørgensen, H.S.; Behets, G.; Bammens, B.; Claes, K.; Meijers, B.; Naesens, M.; Sprangers, B.; Kuypers, D.R.J.; Cavalier, E.; D’Haese, P.; et al. Natural History of Bone Disease following Kidney Transplantation. J. Am. Soc. Nephrol. 2022, 33, 638–652. [Google Scholar] [CrossRef]
  152. Monier-Faugere, M.C.; Mawad, H.; Qi, Q.; Friedler, R.M.; Malluche, H.H. High prevalence of low bone turnover and occurrence of osteomalacia after kidney transplantation. J. Am. Soc. Nephrol. 2000, 11, 1093–1099. [Google Scholar] [CrossRef] [PubMed]
  153. Carlini, R.G.; Rojas, E.; Arminio, A.; Weisinger, J.R.; Bellorin-Font, E. What are the bone lesions in patients with more than four years of a functioning renal transplant? Nephrol. Dial. Transpl. 1998, 13, 103–104. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  154. Evenepoel, P.; Behets, G.J.; Viaene, L.; D’Haese, P.C. Bone histomorphometry in de novo renal transplant recipients indicates a further decline in bone resorption 1 year posttransplantation. Kidney Int. 2017, 91, 469–476. [Google Scholar] [CrossRef]
  155. Neves, C.L.; dos Reis, L.M.; Batista, D.G.; Custodio, M.R.; Graciolli, F.G.; Martin Rde, C.; Neves, K.R.; Dominguez, W.V.; Moyses, R.M.; Jorgetti, V. Persistence of bone and mineral disorders 2 years after successful kidney transplantation. Transplantation 2013, 96, 290–296. [Google Scholar] [CrossRef] [PubMed]
  156. Cruz, E.A.; Lugon, J.R.; Jorgetti, V.; Draibe, S.A.; Carvalho, A.B. Histologic evolution of bone disease 6 months after successful kidney transplantation. Am. J. Kidney Dis. 2004, 44, 747–756. [Google Scholar] [CrossRef]
  157. Keronen, S.; Martola, L.; Finne, P.; Burton, I.S.; Kroger, H.; Honkanen, E. Changes in Bone Histomorphometry after Kidney Transplantation. Clin. J. Am. Soc. Nephrol. 2019, 14, 894–903. [Google Scholar] [CrossRef]
  158. Rojas, E.; Carlini, R.G.; Clesca, P.; Arminio, A.; Suniaga, O.; De Elguezabal, K.; Weisinger, J.R.; Hruska, K.A.; Bellorin-Font, E. The pathogenesis of osteodystrophy after renal transplantation as detected by early alterations in bone remodeling. Kidney Int. 2003, 63, 1915–1923. [Google Scholar] [CrossRef] [Green Version]
  159. Jorgensen, H.S.; Ferreira, A.C.; D’Haese, P.; Haarhaus, M.; Vervloet, M.; Lafage-Proust, M.H.; Ferreira, A.; Evenepoel, P.; European Renal Osteodystrophy (EUROD) workgroup; Chronic Kidney Disease—Mineral and Bone Disorder Working Group (CKD-MBD WG). Bone histomorphometry for the diagnosis of renal osteodystrophy: A call for harmonization of reference ranges. Kidney Int. 2022, 102, 431–434. [Google Scholar] [CrossRef]
Figure 1. Examples of different types of bone disease in CKD Bone histology in CKD. (A) Panoramic view of normal bone histology. Magnification 4×. Toluidine blue staining. Figure shows interconnected bone trabeculae (purple). Clear areas between trabeculae represent medullary space. (B) Hyperparathyroid bone disease (high bone turnover). Magnification 20×. Goldner trichrome staining. Figure shows a resorption area with multinucleated osteoclast in the periphery. Area in blue corresponds to a calcified trabecula. (C) Adynamic bone disease (low bone turnover). Magnification 20×. Toluidine blue staining. Figure shows a bone section without cellular activity. Trabeculae are thin and disconnected. (D) Osteomalacia (low bone turnover). Magnification 20×. Toluidine blue staining. Figure shows abundant osteoid matrix (light blue) covering the mineralized bone trabecula. (E). Mixed uremic osteodystrophy (combines features of high turnover and osteoid matrix). Magnification 20×. Goldner trichrome staining. Picture shows an area of osteoclast resorption with abundant osteoid covering the bone trabecula.
Figure 1. Examples of different types of bone disease in CKD Bone histology in CKD. (A) Panoramic view of normal bone histology. Magnification 4×. Toluidine blue staining. Figure shows interconnected bone trabeculae (purple). Clear areas between trabeculae represent medullary space. (B) Hyperparathyroid bone disease (high bone turnover). Magnification 20×. Goldner trichrome staining. Figure shows a resorption area with multinucleated osteoclast in the periphery. Area in blue corresponds to a calcified trabecula. (C) Adynamic bone disease (low bone turnover). Magnification 20×. Toluidine blue staining. Figure shows a bone section without cellular activity. Trabeculae are thin and disconnected. (D) Osteomalacia (low bone turnover). Magnification 20×. Toluidine blue staining. Figure shows abundant osteoid matrix (light blue) covering the mineralized bone trabecula. (E). Mixed uremic osteodystrophy (combines features of high turnover and osteoid matrix). Magnification 20×. Goldner trichrome staining. Picture shows an area of osteoclast resorption with abundant osteoid covering the bone trabecula.
Nutrients 15 00167 g001
Figure 2. Summary of the pathophysiology of CKD-MBD. The progressive loss of kidney function results in a decrease in renal phosphate excretion. FGF-23 produced by osteocytes and osteoblasts increases early in CKD decreasing NaPi2 and 2c cotransporters expression in the kidney leading to phosphaturia. PTH decreases phosphate reabsorption by similar mechanisms. At some point during the progression of CKD, when GFR approaches stage 4, both mechanisms are insufficient to maintain phosphaturia efficiently and serum phosphorus rises. In addition, FGF-23 inhibits 1-alfa-Hydroxylase synthesis of 1,25-(OH)2 Vitamin D (calcitriol) in the kidney, which results in a decrease in intestinal calcium absorption, serum calcium concentration, and reduction in tissue VDR, resulting in resistance to calcitriol-mediated regulation of PTH secretion. With time, hyperplasia of the parathyroid glands ensues. If conditions persist, hyperplastic glands transform into nodular hyperplasia and lately, into single nodular glands. These glands have a decreased expression of CaSr and consequently, poor response to calcium and calcimimetics. All these factors in concert, lead to the development and progression of secondary hyperparathyroidism. PTH activates osteoblasts and osteoclasts leading to an increase in bone remodeling. Typically, bone turnover in patients with CKD may be elevated as a consequence of the action of PTH in the bone or decreased due to mechanisms not completely understood that include among others, elevated sclerostin, FGF-23, uremic toxins, low calcitriol levels, and hyporesponsiveness of the cells to PTH. In addition to the typical lesions classically described in CKD, osteoporosis has been increasingly described in CKD and seems to play an important role in the elevated risk of fracture in these patients.
Figure 2. Summary of the pathophysiology of CKD-MBD. The progressive loss of kidney function results in a decrease in renal phosphate excretion. FGF-23 produced by osteocytes and osteoblasts increases early in CKD decreasing NaPi2 and 2c cotransporters expression in the kidney leading to phosphaturia. PTH decreases phosphate reabsorption by similar mechanisms. At some point during the progression of CKD, when GFR approaches stage 4, both mechanisms are insufficient to maintain phosphaturia efficiently and serum phosphorus rises. In addition, FGF-23 inhibits 1-alfa-Hydroxylase synthesis of 1,25-(OH)2 Vitamin D (calcitriol) in the kidney, which results in a decrease in intestinal calcium absorption, serum calcium concentration, and reduction in tissue VDR, resulting in resistance to calcitriol-mediated regulation of PTH secretion. With time, hyperplasia of the parathyroid glands ensues. If conditions persist, hyperplastic glands transform into nodular hyperplasia and lately, into single nodular glands. These glands have a decreased expression of CaSr and consequently, poor response to calcium and calcimimetics. All these factors in concert, lead to the development and progression of secondary hyperparathyroidism. PTH activates osteoblasts and osteoclasts leading to an increase in bone remodeling. Typically, bone turnover in patients with CKD may be elevated as a consequence of the action of PTH in the bone or decreased due to mechanisms not completely understood that include among others, elevated sclerostin, FGF-23, uremic toxins, low calcitriol levels, and hyporesponsiveness of the cells to PTH. In addition to the typical lesions classically described in CKD, osteoporosis has been increasingly described in CKD and seems to play an important role in the elevated risk of fracture in these patients.
Nutrients 15 00167 g002
Figure 3. The bone alterations in CKD-MBD, typically described as renal osteodystrophy, may extend or evolve to a different pattern after kidney transplantation by mechanisms that include but are not limited to those leading to the CKD. Osteoporosis is a frequent finding in patients with CKD-MBD that can aggravate the bone disease and increase the risk of fracture.
Figure 3. The bone alterations in CKD-MBD, typically described as renal osteodystrophy, may extend or evolve to a different pattern after kidney transplantation by mechanisms that include but are not limited to those leading to the CKD. Osteoporosis is a frequent finding in patients with CKD-MBD that can aggravate the bone disease and increase the risk of fracture.
Nutrients 15 00167 g003
Table 1. Type of CKD-associated alteration of bone turnover (BTO) in patients with advanced CKD (ESKD or pre-dialysis).
Table 1. Type of CKD-associated alteration of bone turnover (BTO) in patients with advanced CKD (ESKD or pre-dialysis).
ReferenceCountryPatients NumberAge
Years
Kidney Function Low BTO
% Patients
High BTO
% Patients
Normal BTO
% Patients
Comments
Couttenye [36]
1996
Belgium, Greece, Egypt, Argentina, Slovakia, Luxemburg103
Female 53
59.7 ± 1.3ESKD46.740.812.6P P-binder: 89 pts (CaCO3, 33, Al (OH)3 31, both 31). Vit D: 42 pts.
Al overload: 12 pts PTX: 8 pts. Previous kidney transplant: 8 pts
Salusky [17]
1988
USA44
Fem 22
11.8 ± 5.8ESKD
CAPD
206416P-binder: 45% of pts on Al (OH)3. CaCO3 54.5% of pts. All pts receiving PO calcitriol
Bone surface AL in 10 out 20 pts receiving Al (OH)3.
Rodriguez-Perez [37] 1992Spain26NAESKD5743NANA
Sherard [38]
1993
USA 239
Canada 20 pts
259
Female 63
PD: 60 ± 12
Female 63,
HD: 52 ± 1.4
Female 39
ESKD66
38
44
62
NRP-binder: Al (OH)3 predominantly early in the study. CaCO3 predominantly later. Vitamin D: 12.7% of the pts). Surface bone Al 25% in 31% of the pts with low BTO. Serum Al higher in HD.
Low BTO more frequent in PD. Diabetes: 30% of pts on PD and 19% of pts on HD
PTX: 4% of pts on PD, 6% of pts on HD. Dialysis fluid Ca concentration 3.25 meq/L
Torres [39]
1995
Spain
(Canary Islands)
119
Female 80
47 ± 16Predialysis 38
HD: 49
CAPD: 32
60
44
59
40
56
61
NRP-binder: CaCaO3 was primary. Al (OH)3 added to 40% of predialysis pts and 56% of dialysis pts.).
Vitamin D (PO alfa-calcifediol): HD: 6, CAPD 12, predialysis none
Hamdy [40]
1995
Belgium, France, Netherlands, United Kingdom.176 * 18–81CrCl 15–50 mL/min4
7
91
93
NA* Placebo 87
Alfacalcidol 89
Monier-Faugere [15]
1996
USA2248
Female 1144
Female 54 ± 0.4 Male: 50 ± 0.5ESKD
HD: 81.4%
CAPD 18.6%
22.677.4NAP-binder: Al containing more used between 1983 and 1987. Ca-based binders use increased progressively. Vitamin D: Calcitriol: 20% of pts; use of IV progressively increased in time. Al staining at 30% of trabecular surface in 85% of pts. Trend decreased progressively. BTO type varied within the study period. Proportion of use of DFO decreased along the survey.
Coen [41]
2002
ItalyPredialysis 79
HD 107
51 ± 14
51.1 ± 12
S. creatinine
5.2 ± 2.5
ESKD
Predialysis
21.5
HD 14
Predialysis
65.8
HD 86
Predialysis
12.6
NA
P-binder or Vitamin D: Predialysis patients: No. HD pts: CaCO3 Ca
None of the pts were on Vitamin D.
Bone Al higher in HD pts.
Coen [42]Italy 4151.9 ± 12.4ESKD HD2278 P-binder: Ca-based in most patients. Al agents in 39% of pts for short periods.
Calcitriol in a minority of pts.
Spazovsky [31] 2003Macedonia84
Female 40
54.2 ± 12CCl < 5
Non-dialysis
352738P-binder: CaCO3 in 70% of pts). Vitamin D analog: None of the pts.
Ferreira 2008 [43]Portugal68
Female
55. ± 15.4 *
53.9 ± 13.7 **
ESKD* 67 > 56
** 60 > 54
33 > 46
40 > 46
NRP-binders: * Sevelamer N = 33. ** Ca-based N = 35. Pts underwent bone biopsy prior to and at the end of treatment with either P-binder. BTO is shown at both periods.
Malluche [34]
2011
USA
Europe
630
Black 87
White 543
55 ± 1
Black 50.7 ± 1.4
White 56.3 ± 0.6
ESKD
HD: 600
PD: 30
582418Black patients were younger, had less time on dialysis, more treatment with vit D analog and non-Ca/non-Al containing P-binders. Diabetes prevalence: 25.3% black vs. 20.4% white pts, respectively.
P-binder: CaCO3 429 pts. Active Vitamin D analogs: 109 pts.
Dialysate Ca: 2.5 meq/l in 371 pts, Ca 3.3 meq/l in 259 pts.
Sprague [35]
2016
Brazil Portugal, Turkey Venezuela492
Female 218
49.5±ESKD5248MR P-binders: Ca-based 379; Al salts 52, Sevelamer 42 pts, respectively. Vit D 143 pts. Corticosteroids 24, immunosuppressives 20 pts.
Previous kidney transplant 46. Parathyroidectomy 5.
Salam [44]
2018
UK43 59 ± 12CKD 4–5
65% predialysis
264034Diabetes: 28% of pts vs. 0 in controls. Previous fragility fracture 22% of pts vs. 7% in controls. By HR-pQCT, CKD pts had lower BMD, trabecular thickness, and trabecular bone volume at distal radius and distal tibia compared with controls.
Carbonara [25] 2020Brazil26051 ± 12 21.676.61.8Osteoporosis in 35% of pts irrespective of ROD type.
Al staining in 38% of the biopsies.
BTO: bone turnover; ESKD: end stage kidney disease; P-binder: phosphate binder; PO: oral; HD: hemodialysis; PD: peritoneal dialysis; CAPD: continuous ambulatory peritoneal dialysis; CCl: creatinine clearance; pts: patients, VC: vascular calcification; * refer to placebo. ** refers to alfacalcdol.
Table 2. Distribution of types of CKD-associated alteration of bone turnover (BTO) in patients at different CKD stages (not on dialysis).
Table 2. Distribution of types of CKD-associated alteration of bone turnover (BTO) in patients at different CKD stages (not on dialysis).
ReferenceCountryPatients
Number
Age
Years
Kidney Function CKD Stage or GFR
ml/min/1.73 m2
Low BTO
% Patients
High BTO
% Patients
Normal BTO
% Patients
Comments
Malluche 1976 [45]Germany50
Female 31
43.2. ± 116–80NAMost patientsNot reportedBTO increased with decreasing GFR. None of the pts were receiving vit D analogs, PO phosphate binders or Ca supplements.
Bervoets [58]
2003
84 Predialysis ESKD352738Active Vitamin D in 30.8% of the pts across CKD stages, mostly CKD stages 4 to 5D. Calcium containing P-binder in 21.2% of pts, mostly in CKD stage 4 to 5D
Tomiyama [46]
201
Brazil50
Female 66
GFR 15–90CKD 2: 100
CKD 3: 88
CKD 4: 78
0
0
2
0
12
20
No treatment with P-binders or vit D analogs.
High prevalence of hypertension, diabetes, overweight/obesity dyslipidemia.
CAC detected in 66% of pts correlated with bone turnover.
Barreto [59] 2014; Drueke [32] * 2016Brazil49 Female
32
52CKD stage 2–5
GFR 36 ± 17
CKD 2–3: Predominant low BTOCKD 4–5:
Predominant high BTO
Not reportedNo P-binder or vit D treatment
Association of indoxyl sulfate with osteoblast surface and bone fibrosis
* Reanalysis of the same data
Lima [60]
2019
USA104
Female 75
59 ± 15CKD stage 2: 22 pts
CKD stage. 3: 29 pts
CKD stage 4–5: 19 pts
ESKD HD: 34 pts
553313Treatment with active Vitamin D in 30.8% of the patients across the different stages of CKD, mostly in CKD st 4 to 5D
Calcium containing P-binder in 21.2% of patients, mostly in CKD stage 4 to 5D
Graciolli [47]
2017
Brazil148
Female 51
50–54CKD stage 2–5
CKD stage 2–3
CKD stage 4
CKD stage 5
83
94
83
81
17
6
17
19
Not reportedP-binder or Vitamin D: Predialysis patients: No. HD pts: CaCO3 Ca
None of the pts were on Vitamin D.
Bone Al higher in HD pts. Predictive value of iPTH is higher in HD in HD.
El-Husseini
[48]
2022
USA3261 ± 1144 ± 168416Not reportedCalcium supplement 2 pts, Vitamin D none. Diuretics 15 pts.
In white pts, eGFR correlated negatively with BTO. Most pts had VC >80%. VC correlated positively with serum P, FGF-23, and activin.
TBS correlated negatively with coronary calcification
BTO: bone turnover; ESKD: end stage kidney disease; BALP: bone alkaline phosphatase; P-binder: phosphate binder; CAC: coronary artery calcification; ALP: alkaline phosphatase; TBS trabecular bone score.
Table 3. Incidence of fractures in CKD.
Table 3. Incidence of fractures in CKD.
ReferenceCohort
Number of pts (N)
CKD Stage/CCl ml/min or eGFR ml/min/1.73 m2Fracture Type and NumberFracture
Incidence
1000 Person-Years
Comments
Alem [5]
2000
USRDS data base N: 326,464 person-yearESKDHip 6542Men 7.45
Women 13.63
Relative risk highest in younger people. Added incidence of fracture increased with age and was greater for women than for men.
Coco [6]
2000
N: 4039 person-year, N: 1272 pts treated ESKD HDHip 56 Men 11.7
Women 24.1
The one-year mortality rate from hip fracture was ~2.5 times higher in dialysis pts compared with general population.
Jadoul [99]
2006
DOPPS: HD pts. 12,782ESKD HDHip 174
Any 498
8.9 for hip
25.6 for any new fracture
Older age, female sex, prior kidney transplant and low serum albumin were predictive of new fracture. PTH > 900 was associated with risk of new fracture
Danese [100] 2006DMMS data base
N: 9007 pts
NAHip and vertebral580/1000 vs. 217/1000 in the general dialysis population Age and sex-adjusted mortality rate after fracture 2.7 times greater than the dialysis population. Pts with lower PTH were more likely to sustain a hip fracture than those with higher PTH.
Dukas [101]
2005
Cross sectional
N: Women 5313 N: Men 3238
CrCl ml/min
60.9% < 65
39.1% ≥ 65
Not reportedNot reported CCl < 65 increased risk of experiencing falls and risk for hip fracture (OR 1.57, 95%CI 1.18–2.09, p = 0.002), and for vertebral fracture
Lin [8]
2014
Taiwan NHIRD
N: 51,473 incident dialysis patients
ESKD
Dialysis
Hip 19038.92
Men 7.54
Female 10.12
HD pts had a 31% higher incidence of hip fracture than PD patients (HR 1.31, 95% CI: 1.01–1.70). Patients ≥65 years old had more than 13 times the risk of a hip fracture than those 18–44 years old (HR: 13.65; 95% CI: 10.12–18.40)
Tentori [90]
2014
International
DOPPS N: 34,579
ESKD/HDNAJapan 1
Belgium 45
Fracture pts had 3.7-fold higher rates of death compared to DOPPS population.
In most countries, mortality rates exceeded 500 per 1000 patient-year
Naylor [97]
2014
Data base from Ontario, Canada N: 679,114 eGFR
ml/min/1.73 m2:
≥60; 45–59; 30–44; 15–29; <15
Hip
Forearm
Pelvis
Humerus
Not available in women ≥ 60Fracture rate in women ≥ 65 years old at different eGFR (ml/min/1.73 m2):
>60: 4.3%
45–59: 43%: 5.8%
30–44: 47.9%: 6.5%
15–29: 54.4%: 7.8%
<15.54.2%: 9.6
Naylor [102]
2015
2107
320 individuals with eGFR < 60 mL/min/1.73m2
1787 individuals with eGFR ≥ 60 mL/min/m2
eGFR ml/min/1.73 m2:
≥60; 45–59; 30–44
15–29; <15
64 (3%) over 4.8 yearsNot availableThe 5-year observed major osteoporotic fracture risk was 5.3% in individuals with eGFR < 60 mL/min/1.73m2 was 5.3%, comparable to the FRAX predicted fracture risk.
No difference in the AUC values for FRAX in individuals with eGFR < 60 mL/min/1.73 m2 vs. those with eGFR ≥ 60 mL/min/m2
Yamamoto
[103] 2015
3276 * 1.48
** 2.33
Mortality was lower in pts * using ACEI/ARB than those ** not using ACEI/ARB 13.6% vs. 16.8%
Hung [7]
2017
Taiwan’s NHIRD
Total of 61,346 first fragility hip fracture nationwide.
997 dialysis hip fracture patients were matched to 4985 non-dialysis hip fracture subjects
ESKD DialysisHip 997Not available Higher proportion of femoral neck fractures in the dialysis group compared to the non-dialysis group (51% and 42%, respectively; p < 0.001)
The mortality rate was significantly higher for patients in the dialysis group, with a mortality rate of 91% compared to 71% for those in the non-dialysis group ( p < 0.001).
Desbiens [9]
2020
CARTaGENE data base (CAG)
N: 679,114
19,391pts with CKD included
Non-CKD: 9521
CKD: 2: 9114
CKD 3: 756
829
Various type
Non-CKD: 6.9
CKD 2: 7.6
CKD 3: 11.3
Compared with the median eGFR of 90 mL/min/1.73 m2, eGFRs of 60 mL/min/1.73 m2 were associated with increased fracture incidence [adjusted hazard ratio (HR) ¼ 1.25 (95% confidence interval 1.05–1.49) for 60 mL/min/1.73 m2; 1.65 (1.14–2.37) for 45 mL/min/1.73 m2]. The effect of decreased eGFR on fracture incidence was higher in younger individuals [HR 2.45 (1.28–4.67) at 45 years; 1.11 (0.73–1.67) at 65 years and in men.
URDS: United States Renal Data System; DOPPS: Dialysis Outcomes and Practice Patterns Study; Taiwan NHIRD: Taiguan’s National Health Insurance; HD: hemodialysis; CCl: creatinine clearance. FRAX: Fracture Risk Assessment Tool.
Table 4. Relationship between bone markers and discrimination of the type of bone turnover alteration in CKD.
Table 4. Relationship between bone markers and discrimination of the type of bone turnover alteration in CKD.
ReferenceCountryNumber of PatientsAge
Years
Kidney FunctionComments
Coutteneye [36]
1996
Belgium, Greece, Egypt, Argentina, Slovakia, Luxemburg 103
Female 53
59.7 ± 1.3ESKDCut-off: BALP ≤ 27 U/l, osteocalcin 1≤; PTH: ≤ 150 pg/mL had the best specificity and sensitivity for detection of adynamic bone disease.
Salusky [17]
1988
USA44
Female 22
11.8 ± 5.8ESKD on CAPDBone formation rate correlated with serum PTH.
Sherard [38]
1993
USA and Canada259
Female 63
PD: 60 ± 12
HD: 52 ± 1.4
ESKD iPTH correlated directly with bone formation: higher in high bone turnover lesions, intermediate in mild lesions with normal bone formation
Torres [39]
1995
Spain
(Canary Islands)
119
Female 80
47 ± 16Predialysis 38
ESKD HD: 49 pts
ESKD CAPD: 32 pts
iPTH level 120–250 pg/mL needed to avoid low bone turnover and HPT bone disease.
Monier-Faugere [15]
1996
USA2248
Female 1144
Female 54 ± 0.4
Male: 50 ± 0.5
ESKD HD 81.4%
CAPD18.6%
Serum ALP significantly greater in patients with HPT, and in low BTO.
iPTH greater in pts with HPT than in those with MUO, and low BTO,, respectively.
Fletcher [123]
1997
USA 73 DialysisPTH > 100 pg/mL sensitivity of 81%, specificity of 66% for high BTO
Coen 2002 [124]Italy186 Predialysis: 79 pts
HD: 107 pts
In dialysis pts, iPTH level of 150 pg/mL had a negative predictive value for low BTO of 96.4%, and a Youden index of 0.69.
In predialysis pts. The Youden index was 0.57.
AP had a Youden Index of 0.64.
Bervoets [58]
2003
84 For ABD, osteocalcin 41mcg/L, sensitivity of 83% and specificity of 67%. PPV 47%. Combination with BALP 23 U/L or less increased sensitivity, specificity, and PPV to 72%, 89% and 77%, respectively
Barreto [125]
2008
Brazil97 ESRD on dialysisFor low turnover: PTH < 150 pg/mL, sensitivity of 50%, specificity of 85%, PPV 83%
For high bone turnover: PTH >300 pg/mL, sensitivity 0.69, specificity 0.75, PPV 0.62
Lehman [126]
2005
USA132 CKD 3–5Patients with CKD stage 3–4 and low BTO had BI-PTH (biointact PTH) and iPTH (intact PTH) levels (in pg/mL ± SD) of 35 ± 34 and 59 ± 63. For high BTO, BI-PTH and iPTH 141 ± 60 and 221 ± 106, respectively.
In CKD stage 5 and low BTO BI-PTH 51 ± 38 and iPTH 90 ± 60 pg/mL. For high BTO BI-PTH and iPTH levels of 237 ± 214 and 461 ± 437 pg/mL, respectively.
Areas under the ROC curves for distinguishing low BTO from high BTO were 0.94 for BI-PTH and 0.91 for iPTH, respectively in stages 3–4.
For CKD stage 5, values were 0.86 and 0.85, respectively.
Malluche [34]
2011
USA
Europe
630
Female 301
55 ± 1
ESKD
HD: 600 PD: 30
PTH not a significant predictor of bone turnover in black dialysis patients. PTH was a predictor of low bone turnover in white patients on dialysis
Haarhaus [127]
2015
PTH, BALP, Bix have similar diagnostic accuracy in distinguishing low from non-low BTO. BALP (AUC, 0.89) and PTH (AUC, 0.85) are useful for the diagnosis of non-low BTO. B1x can be used for the diagnosis of low BTO (area under the curve (0.83)
Sprague [35]
2016
Brazil 156
Portugal 89
Turkey 133
Venezuela 114
492
Female 218
49.5±ESKD iPTH and BALP allowed discrimination of low from non-low and high from non-high BTO. Optimal iPTH to discriminate low from non-low BTO: 104/pg/mL, and for high vs. non-high bone turnover: 323 pg/mL.
Optimal BALP: 33.1 U/L, better for diagnosing low BTO. Combination of iPTH and BALP did not improve discrimination between low and high BTO
Marques [128]
2017
Brazil 31
Female 19
41 ± 11ESKD
HD 27, PD 3
No dialysis 1
Patients with low BTO: low PTH, BAP, and TRAP5b.
BAP was the best predictor of BTO status. Best cut-off 66.6 U/L. Sensitivity 85, specificity 82.
Graciolli [47]
2017
Brazil148
Female 51
50–54 CKD stage 2 -5 ROC PTH and FGF-23 curves were able to predict high BTO and low BTO.
None of the markers was able to predict bone mineralization
Salam [44]
2018
UK43 59 ± 12CKD 4–5
including HD
BALP, intact PINP, and TRAP 5b better than iPTH for discriminating low from non-low BTO. iPTH can discriminate high BTO from non-high BTO
Lima [60]
2019
USA104
Female 75
59 ± 15CKD stage 2: 22
CKD stage 3: 29
CKD stage 4–519
ESKD on HD: 34
Serum activin increased with declining eGFR
Activin showed similar AUC results, specificity, and sensitivity in predicting high turnover as iPTH, BSAP, and FGF-23 for discrimination of high vs. non-high bone turnover.
AUC: area under the curve; BALP bone alkaline phosphatase; B1x: isoform of BALP found in sermon of some patients with CKD; BBTO: bone turnover iPTH: intact parathyroid hormone; BI-PTH: biointact PTH; PPV: predictive positive value. ABD: adynamic bone disease. TRAP5b: tartrate-resistant acid phosphatase 5b.
Disclaimer/Publisher’s Note: The statements, opinions and data contained in all publications are solely those of the individual author(s) and contributor(s) and not of MDPI and/or the editor(s). MDPI and/or the editor(s) disclaim responsibility for any injury to people or property resulting from any ideas, methods, instructions or products referred to in the content.

Share and Cite

MDPI and ACS Style

Bellorin-Font, E.; Rojas, E.; Martin, K.J. Bone Disease in Chronic Kidney Disease and Kidney Transplant. Nutrients 2023, 15, 167. https://doi.org/10.3390/nu15010167

AMA Style

Bellorin-Font E, Rojas E, Martin KJ. Bone Disease in Chronic Kidney Disease and Kidney Transplant. Nutrients. 2023; 15(1):167. https://doi.org/10.3390/nu15010167

Chicago/Turabian Style

Bellorin-Font, Ezequiel, Eudocia Rojas, and Kevin J. Martin. 2023. "Bone Disease in Chronic Kidney Disease and Kidney Transplant" Nutrients 15, no. 1: 167. https://doi.org/10.3390/nu15010167

Note that from the first issue of 2016, this journal uses article numbers instead of page numbers. See further details here.

Article Metrics

Back to TopTop