Your browser doesn't support javascript.
loading
: 20 | 50 | 100
1 - 20 de 522
1.
Nutrients ; 16(5)2024 Feb 22.
Article En | MEDLINE | ID: mdl-38474734

Bone represents a metabolically active tissue subject to continuous remodeling orchestrated by the dynamic interplay between osteoblasts and osteoclasts. These cellular processes are modulated by a complex interplay of biochemical and mechanical factors, which are instrumental in assessing bone remodeling. This comprehensive evaluation aids in detecting disorders arising from imbalances between bone formation and reabsorption. Osteoporosis, characterized by a reduction in bone mass and strength leading to heightened bone fragility and susceptibility to fractures, is one of the more prevalent chronic diseases. Some epidemiological studies, especially in patients with chronic kidney disease (CKD), have identified an association between osteoporosis and vascular calcification. Notably, low bone mineral density has been linked to an increased incidence of aortic calcification, with shared molecules, mechanisms, and pathways between the two processes. Certain molecules emerging from these shared pathways can serve as biomarkers for bone and mineral metabolism. Detecting and evaluating these alterations early is crucial, requiring the identification of biomarkers that are reliable for early intervention. While traditional biomarkers for bone remodeling and vascular calcification exist, they suffer from limitations such as low specificity, low sensitivity, and conflicting results across studies. In response, efforts are underway to explore new, more specific biomarkers that can detect alterations at earlier stages. The aim of this review is to comprehensively examine some of the emerging biomarkers in mineral metabolism and their correlation with bone mineral density, fracture risk, and vascular calcification as well as their potential use in clinical practice.


Chronic Kidney Disease-Mineral and Bone Disorder , Fractures, Bone , Osteoporosis , Renal Insufficiency, Chronic , Vascular Calcification , Humans , Chronic Kidney Disease-Mineral and Bone Disorder/complications , Osteoporosis/etiology , Bone Density/physiology , Renal Insufficiency, Chronic/complications , Fractures, Bone/etiology , Vascular Calcification/complications , Biomarkers , Minerals
2.
Int Urol Nephrol ; 56(5): 1669-1676, 2024 May.
Article En | MEDLINE | ID: mdl-37964112

BACKGROUND: Mineral and bone disease in children with chronic kidney disease can cause abnormalities in calcium, phosphorus, parathyroid hormone, and vitamin D and when left untreated can result in impaired growth, bone deformities, fractures, and vascular calcification. Cinacalcet is a calcimimetic widely used as a therapy to reduce parathyroid hormone levels in the adult population, with hypocalcemia among its side effects. The analysis of safety in the pediatric population is questioned due to the scarcity of randomized clinical trials in this group. OBJECTIVE: To assess the onset of symptomatic hypocalcemia or other adverse events (serious or non-serious) with the use of cinacalcet in children and adolescents with mineral and bone disorder in chronic kidney disease. DATA SOURCES AND STUDY ELIGIBILITY CRITERIA: The bibliographic search identified 2699 references from 1927 to August/2023 (57 LILACS, 44 Web of Science, 686 PubMed, 131 Cochrane, 1246 Scopus, 535 Embase). Four references were added from the bibliography of articles found and 12 references from the gray literature (Clinical Trials). Of the 77 studies analyzed in full, 68 were excluded because they did not meet the following criteria: population, types of studies, medication, publication types and 1 article that did not present results (gray literature). PARTICIPANTS AND INTERVENTIONS: There were 149 patients aged 0-18 years old with Chronic Kidney Disease and mineral bone disorder who received cinacalcet. STUDY APPRAISAL AND SYNTHESIS METHODS: Nine eligible studies were examined for study type, size, intervention, and reported outcomes. RESULTS: There was an incidence of 0.2% of fatal adverse events and 16% of serious adverse events (p < 0.01 and I2 = 69%), in addition to 10.7% of hypocalcemia, totaling 45.7% of total adverse events. LIMITATIONS: There was a bias in demographic information and clinical characteristics of patients in about 50% of the studies and the majority of the studies were case series. CONCLUSIONS AND IMPLICATIONS OF KEY FINDINGS: If used in the pediatric population, the calcimimetic cinacalcet should be carefully monitored for serum calcium levels and attention to possible adverse events, especially in children under 50 months. SYSTEMATIC REVIEW REGISTRATION NUMBER (PROSPERO REGISTER): CRD42019132809.


Bone Diseases , Chronic Kidney Disease-Mineral and Bone Disorder , Hyperparathyroidism, Secondary , Hypocalcemia , Renal Insufficiency, Chronic , Child , Adult , Humans , Adolescent , Infant, Newborn , Infant , Child, Preschool , Cinacalcet/adverse effects , Calcium , Calcimimetic Agents/adverse effects , Hypocalcemia/etiology , Renal Insufficiency, Chronic/therapy , Parathyroid Hormone , Minerals/therapeutic use , Chronic Kidney Disease-Mineral and Bone Disorder/etiology , Chronic Kidney Disease-Mineral and Bone Disorder/complications , Hyperparathyroidism, Secondary/drug therapy , Hyperparathyroidism, Secondary/etiology , Renal Dialysis/adverse effects
3.
Rom J Intern Med ; 62(1): 52-60, 2024 Mar 01.
Article En | MEDLINE | ID: mdl-37991505

INTRODUCTION: Chronic kidney disease (CKD) has always been a complicated global challenge, ranking as the 12th leading cause of death worldwide. Hemodialysis, being one of the most opted renal replacement therapies (RRTs) for patients with end-stage renal disease (ESRD), still possesses some limitations in preventing complications, such as malnutrition and mineral bone disease (CKD-MBD). While efforts have focused on controlling CKD-MBD parameters like calcium and phosphate, less attention has been given to dietary interventions. Moreover, the adoption of low-phosphorus diets for hemodialysis patients is very complex due to potential conflicts with the guideline-recommended high-protein dietary approach. This study sought to investigate the relationship between dietary phosphorus intake and nutritional status in CKD patients undergoing regular hemodialysis. METHOD: This non-randomized cross-sectional study comprising 88 patients was conducted at the Hemodialysis Unit, RSUD Dr. Soetomo, Surabaya, East Java, using a three-day dietary record in March 2022. Relationships between variables were analyzed using Spearman and ANOVA tests. RESULT: No significant positive association was found between dietary calcium with corrected calcium levels (p = 0.988; rs = -0.002) and between dietary phosphorus with plasma phosphate levels (p = 0.082; rs = 0.187). However, Spearman's analysis revealed a weak but positive correlation between dietary phosphorus and nutritional status (p = 0.022; rs = 0.215*). CONCLUSION: Our study highlights a positive relationship between dietary phosphorus and nutritional status among hemodialysis patients, offering insights into potential strategies for optimizing patient care and outcomes.


Chronic Kidney Disease-Mineral and Bone Disorder , Phosphorus, Dietary , Renal Insufficiency, Chronic , Humans , Calcium , Renal Dialysis , Chronic Kidney Disease-Mineral and Bone Disorder/complications , Cross-Sectional Studies , Hemodialysis Units, Hospital , Renal Insufficiency, Chronic/complications , Renal Insufficiency, Chronic/therapy , Phosphates
4.
Pediatr Nephrol ; 39(3): 723-739, 2024 Mar.
Article En | MEDLINE | ID: mdl-37624528

Chronic kidney disease (CKD) mineral and bone disorder (MBD) comprises a triad of biochemical abnormalities (of calcium, phosphate, parathyroid hormone and vitamin D), bone abnormalities (turnover, mineralization and growth) and extra-skeletal calcification. Mineral dysregulation leads to bone demineralization causing bone pain and an increased fracture risk compared to healthy peers. Vascular calcification, with hydroxyapatite deposition in the vessel wall, is a part of the CKD-MBD spectrum and, in turn, leads to vascular stiffness, left ventricular hypertrophy and a very high cardiovascular mortality risk. While the growing bone requires calcium, excess calcium can deposit in the vessels, such that the intake of calcium, calcium- containing medications and high calcium dialysate need to be carefully regulated. Normal physiological bone mineralization continues into the third decade of life, many years beyond the rapid growth in childhood and adolescence, implying that skeletal calcium requirements are much higher in younger people compared to the elderly. Much of the research into the link between bone (de)mineralization and vascular calcification in CKD has been performed in older adults and these data must not be extrapolated to children or younger adults. In this article, we explore the physiological changes in bone turnover and mineralization in children and young adults, the pathophysiology of mineral bone disease in CKD and a potential link between bone demineralization and vascular calcification.


Bone Diseases, Metabolic , Chronic Kidney Disease-Mineral and Bone Disorder , Renal Insufficiency, Chronic , Vascular Calcification , Child , Humans , Aged , Young Adult , Adult , Calcium , Renal Insufficiency, Chronic/complications , Vascular Calcification/etiology , Minerals , Chronic Kidney Disease-Mineral and Bone Disorder/complications
5.
Indian Pediatr ; 60(12): 1039-1040, 2023 Dec 15.
Article En | MEDLINE | ID: mdl-38087789

We report clinical and etiological profile of 19 children (10 males) with renal rickets managed in the years 2021-2022. Median (IQR) age of presentation was 60 (18-96) months. The commonest cause was renal tubular acidosis (n=8). Genetic analysis revealed the diagnosis in 83% subjects (5 out of 6 tested).


Acidosis, Renal Tubular , Chronic Kidney Disease-Mineral and Bone Disorder , Rickets , Male , Child , Humans , Child, Preschool , Rickets/diagnosis , Rickets/complications , Chronic Kidney Disease-Mineral and Bone Disorder/complications , Acidosis, Renal Tubular/diagnosis , Acidosis, Renal Tubular/genetics
6.
Nephrol Ther ; 19(6): 532-541, 2023 11 02.
Article Fr | MEDLINE | ID: mdl-37915196

Patients with advanced chronic kidney disease and those already on dialysis have an increased prevalence of cardiovascular calcifications. They are the cause of severe complications and are associated with a reduced life expectancy in these patients. Recommendations and imaging scores have been developed to detect and assess their importance, to guide and improve the management of cardiovascular risk. However, despite these recommendations, current practice teaches us that they are only partially applied. The prevention and treatment of cardiovascular calcifications go through the correction of classic risk factors associated with atherosclerosis, mineral and bone metabolism disorders and by optimizing the dose and the efficiency of dialysis. New therapeutic strategies are beginning to emerge, others are being evaluated, such as sodium thiosulfate, rheopheresis, vitamin K, magnesium supplementation, and SNF-472.


Les patients atteints de maladie rénale chronique (MRC) avancée et ceux déjà traités par dialyse présentent une prévalence accrue de calcifications cardiovasculaires. Elles sont à l'origine des complications sévères et s'associent à une diminution de l'espérance de vie chez ces patients. Des recommandations et des scores radiographiques ont été développés pour dépister et évaluer leur importance, afin d'orienter et améliorer la prise en charge du risque cardiovasculaire. Cependant, en dépit de ces recommandations, la pratique courante nous enseigne qu'elles ne sont que partiellement appliquées. La prévention et le traitement de calcifications cardiovasculaires passent par la correction des facteurs de risque classiques associés à l'athérosclérose, des troubles du métabolisme minéral et osseux et en optimisant la dose et l'efficacité de la dialyse. Des nouvelles stratégies thérapeutiques commencent à voir le jour, d'autres sont en cours d'évaluation, comme le thiosulfate de sodium, la rhéophérèse, la vitamine K, la supplémentation en magnésium et le SNF-472.


Chronic Kidney Disease-Mineral and Bone Disorder , Renal Insufficiency, Chronic , Humans , Renal Dialysis/adverse effects , Renal Insufficiency, Chronic/complications , Renal Insufficiency, Chronic/therapy , Renal Insufficiency, Chronic/metabolism , Chronic Kidney Disease-Mineral and Bone Disorder/complications
7.
Endokrynol Pol ; 74(5): 490-498, 2023.
Article En | MEDLINE | ID: mdl-37902013

Secondary hyperparathyroidism (SHPT) is one of the most common metabolic complications resulting from chronic kidney disease (CKD). The complexity of calcium and phosphate disorders associated with CKD is defined by the Kidney Disease Improvement Global Outcomes (KDIGO) working group as CKD-related mineral and bone disorders (CKD-MBD). The last update of the KDIGO guidelines on the conduct in CKD-MBD was published in 2017. The treatment of SHPT is based on 2 strategies: counteracting hyperphosphataemia and suppressing parathyroid hormone (PTH) secretion. Therapy should be based on optimally selected drugs, taking into account additional effects to reduce the risk of chronic complications and side effects. The creation of new drugs with a better safety profile, significant reduction of side effects, and greater efficiency in achieving target serum phosphorus and PTH values forces the gradual replacement of existing treatment with new pharmacotherapies. The aim of this study is to discuss the latest issues (in connection with the latest KDIGO guidelines) regarding the pathomechanism of secondary hyperparathyroidism and the current directions of the therapy in these disorders.


Chronic Kidney Disease-Mineral and Bone Disorder , Hyperparathyroidism, Secondary , Renal Insufficiency, Chronic , Humans , Chronic Kidney Disease-Mineral and Bone Disorder/complications , Parathyroid Hormone/therapeutic use , Hyperparathyroidism, Secondary/drug therapy , Hyperparathyroidism, Secondary/etiology , Renal Insufficiency, Chronic/complications , Renal Insufficiency, Chronic/therapy , Calcium
8.
Osteoporos Int ; 34(12): 2003-2012, 2023 Dec.
Article En | MEDLINE | ID: mdl-37658999

Renal osteodystrophy (ROD) starts early and progresses with further loss of kidney function in patients with chronic kidney disease (CKD). There are four distinct types of ROD based on undecalcified bone biopsy results. Adynamic bone disease and osteomalacia are the predominant forms of low bone turnover, while hyperparathyroid bone disease and mixed uremic osteodystrophy (MUO) are typically associated with high bone turnover. MUO is a prevalent but poorly described pathology that demonstrates evidence of osteomalacia on top of the high bone formation/resorption. The prevalence of MUO ranges from 5 to 63% among different studies. The pathogenesis of MUO is multi-factorial. Altered phosphate homeostasis, hypocalcemia, vitamin D deficiency, increased FGF-23, interleukins 1 and 6, TNF-α, amyloid, and heavy metal accumulation are the main inducers of MUO. The clinical findings of MUO are usually non-specific. The use of non-invasive testing such as bone turnover markers and imaging techniques might help to suspect MUO. However, it is usually impossible to precisely diagnose this condition without performing bone biopsy. The principal management of MUO is to control the maladaptive hyperparathyroidism along with correcting any nutritional mineral deficiencies that may induce mineralization defect. MUO is a common but still poorly understood bone pathology category; it demonstrates the complexity and difficulty in understanding ROD. A large prospective bone biopsy-based studies are needed for better identification as proper diagnosis and management would improve the outcome of patients with MUO.


Bone Diseases, Metabolic , Bone Resorption , Chronic Kidney Disease-Mineral and Bone Disorder , Osteomalacia , Renal Insufficiency, Chronic , Humans , Osteomalacia/complications , Prospective Studies , Bone and Bones , Renal Insufficiency, Chronic/complications , Bone Diseases, Metabolic/complications , Chronic Kidney Disease-Mineral and Bone Disorder/complications
9.
Medicine (Baltimore) ; 102(25): e34044, 2023 Jun 23.
Article En | MEDLINE | ID: mdl-37352066

BACKGROUND: Correction of calcium, phosphorus, and parathyroid hormone disorders is the standard of treatment in nondialysis patients with chronic kidney disease-mineral and bone disorder (CKD-MBD). However, the side effects and adverse reactions are still the main problems. Moreover, the lack of protection of kidney function in the treatment dramatically affects patients' health. Although Traditional Chinese Medicine, specifically tonifying kidney and strengthen bone (TKSB) therapy, is wildly applied to patients with CKD-MBD in China, the evidence of TKSB therapy in the treatment of CKD-MBD is limited. Thus, we conducted this meta-analysis to evaluate the efficacy and safety of TKSB therapy combined with Western medicine (WM) for nondialysis patients with CKD-MBD. METHODS: Two investigators conducted systematic research of randomized controlled trials of TKSB therapy for CKD-MBD from 7 electronic databases. Methodological quality evaluations were performed using the Cochrane collaboration tool, and data analysis was conducted by RevMan v5.3 software and STATA v15.0. RESULTS: In total, 8 randomized controlled trials involving 310 patients met the criteria of meta-analysis. The complete results showed that compared with WM alone, TKSB treatment could improve the clinical efficacy rate (risk ratio = 4.49, 95% confidence interval [CI]: [2.64, 7.61], P  .00001), calcium (weighted mean difference [WMD] = 0.11, 95% CI: [0.08, 0.14], P < .00001), serum creatinine (WMD = 45.58, 95% CI: [32.35, 58.8], P < .00001) phosphorus (WMD = 0.11, 95% CI: [0.08, 0.13], P < .00001), parathyroid hormone (WMD = 16.72, 95% CI: [12.89, 20.55], P < .00001), blood urea nitrogen levels (WMD = 0.95, 95% CI: [0.26, 1.64], P = .007) on nondialysis patients with CKD-MBD, which was beneficial to improve the patients' bone metabolic state and renal function. In addition, evidence shows that, compared with WM alone, TKSB treatment is safe and does not increase side effects. CONCLUSION: The systematic review found that TKSB therapy combined with WM has a positive effect on improving renal function and correcting bone metabolism disorder in nondialysis patients with CKD-MBD, which shows that Traditional Chinese Medicine is effective and safe in treating CKD-MBD. However, more high-quality, large-sample, multicenter clinical trials should be conducted to assess the safety and efficacy of TKSB therapy in treating nondialysis patients with CKD-MBD.Systematic review registration: INPLASY2020120086.


Chronic Kidney Disease-Mineral and Bone Disorder , Drug-Related Side Effects and Adverse Reactions , Humans , Calcium , Calcium, Dietary , Chronic Kidney Disease-Mineral and Bone Disorder/complications , Chronic Kidney Disease-Mineral and Bone Disorder/therapy , Kidney , Multicenter Studies as Topic , Parathyroid Hormone , Randomized Controlled Trials as Topic
10.
Nefrologia (Engl Ed) ; 43 Suppl 1: 1-36, 2023 06.
Article En | MEDLINE | ID: mdl-37202281

As in 2011, when the Spanish Society of Nephrology (SEN) published the Spanish adaptation to the Kidney Disease: Improving Global Outcomes (KDIGO) universal Guideline on Chronic Kidney Disease-Mineral and Bone Disorder (CKD-MBD), this document contains an update and an adaptation of the 2017 KDIGO guidelines to our setting. In this field, as in many other areas of nephrology, it has been impossible to irrefutably answer many questions, which remain pending. However, there is no doubt that the close relationship between the CKD-MBD/cardiovascular disease/morbidity and mortality complex and new randomised clinical trials in some areas and the development of new drugs have yielded significant advances in this field and created the need for this update. We would therefore highlight the slight divergences that we propose in the ideal objectives for biochemical abnormalities in the CKD-MBD complex compared to the KDIGO suggestions (for example, in relation to parathyroid hormone or phosphate), the role of native vitamin D and analogues in the control of secondary hyperparathyroidism and the contribution of new phosphate binders and calcimimetics. Attention should also be drawn to the adoption of important new developments in the diagnosis of bone abnormalities in patients with kidney disease and to the need to be more proactive in treating them. In any event, the current speed at which innovations are taking place, while perhaps slower than we might like, globally drives the need for more frequent updates (for example, through Nefrología al día).


Bone Diseases, Metabolic , Chronic Kidney Disease-Mineral and Bone Disorder , Nephrology , Renal Insufficiency, Chronic , Humans , Chronic Kidney Disease-Mineral and Bone Disorder/therapy , Chronic Kidney Disease-Mineral and Bone Disorder/complications , Renal Insufficiency, Chronic/complications , Renal Insufficiency, Chronic/therapy , Renal Insufficiency, Chronic/diagnosis , Bone Diseases, Metabolic/drug therapy , Bone Diseases, Metabolic/etiology , Bone Diseases, Metabolic/diagnosis , Minerals/therapeutic use , Phosphates
11.
J Atheroscler Thromb ; 30(8): 835-850, 2023 Aug 01.
Article En | MEDLINE | ID: mdl-37258233

Cardiovascular disease (CVD) is the leading cause of death in patients with chronic kidney disease (CKD). Multiple factors account for the increased incidence of cardiovascular morbidity and mortality in patients with CKD. Traditional risk factors for atherosclerosis and arteriosclerosis, including age, hypertension, dyslipidemia, diabetes mellitus, and smoking, are also risk factors for CKD. Non-traditional risk factors specific for CKD are also involved in CVD pathogenesis in patients with CKD. Recently, CKD-mineral and bone disorder (CKD-MBD) has emerged as a key player in CVD pathogenesis in the context of CKD. CKD-MBD manifests as hypocalcemia and hyperphosphatemia in the later stages of CKD; however, it initially develops much earlier in disease course. The initial step in CKD-MBD involves decreased phosphate excretion in the urine, followed by increased circulating concentrations of fibroblast growth factor 23 (FGF23) and parathyroid hormone (PTH), which increase urinary phosphate excretion. Simultaneously, the serum calcitriol concentration decreases as a result of FGF23 elevation. Importantly, FGF23 and PTH cause left ventricular hypertrophy, arrhythmia, and cardiovascular calcification. More recently, calciprotein particles, which are nanoparticles composed of calcium, phosphate, and fetuin-A, among other components, have been reported to cause inflammation, cardiovascular calcification, and other clinically relevant outcomes. CKD-MBD has become one of the critical therapeutic targets for the prevention of cardiovascular events and is another link between cardiology and nephrology. In this review, we describe the role of CKD-MBD in the pathogenesis of cardiovascular disorders and present the current treatment strategies for CKD-MBD.


Atherosclerosis , Cardiovascular Diseases , Chronic Kidney Disease-Mineral and Bone Disorder , Renal Insufficiency, Chronic , Humans , Chronic Kidney Disease-Mineral and Bone Disorder/complications , Cardiovascular Diseases/etiology , Cardiovascular Diseases/metabolism , Renal Insufficiency, Chronic/complications , Renal Insufficiency, Chronic/metabolism , Parathyroid Hormone , Phosphates , Atherosclerosis/pathology , Disease Progression , Minerals
12.
Nephrol Dial Transplant ; 38(11): 2562-2575, 2023 Oct 31.
Article En | MEDLINE | ID: mdl-37230954

BACKGROUND: Chronic kidney disease-mineral and bone disorder (CKD-MBD) is a common complication of CKD; it is associated with higher mortality in dialysis patients, while its impact in non-dialysis patients remains mostly unknown. We investigated the associations between parathyroid hormone (PTH), phosphate and calcium (and their interactions), and all-cause, cardiovascular (CV) and non-CV mortality in older non-dialysis patients with advanced CKD. METHODS: We used data from the European Quality study, which includes patients aged ≥65 years with estimated glomerular filtration rate ≤20 mL/min/1.73 m2 from six European countries. Sequentially adjusted Cox models were used to assess the association between baseline and time-dependent CKD-MBD biomarkers and all-cause, CV and non-CV mortality. Effect modification between biomarkers was also assessed. RESULTS: In 1294 patients, the prevalence of CKD-MBD at baseline was 94%. Both PTH [adjusted hazard ratio (aHR) 1.12, 95% confidence interval (CI) 1.03-1.23, P = .01] and phosphate (aHR 1.35, 95% CI 1.00-1.84, P = .05), but not calcium (aHR 1.11, 95% CI 0.57-2.17, P = .76), were associated with all-cause mortality. Calcium was not independently associated with mortality, but modified the effect of phosphate, with the highest mortality risk found in patients with both hypercalcemia and hyperphosphatemia. PTH level was associated with CV mortality, but not with non-CV mortality, whereas phosphate was associated with both CV and non-CV mortality in most models. CONCLUSIONS: CKD-MBD is very common in older non-dialysis patients with advanced CKD. PTH and phosphate are independently associated with all-cause mortality in this population. While PTH level is only associated with CV mortality, phosphate seems to be associated with both CV and non-CV mortality.


Chronic Kidney Disease-Mineral and Bone Disorder , Renal Insufficiency, Chronic , Humans , Aged , Chronic Kidney Disease-Mineral and Bone Disorder/complications , Calcium , Parathyroid Hormone , Phosphates , Calcium, Dietary , Biomarkers , Renal Insufficiency, Chronic/complications , Renal Insufficiency, Chronic/therapy , Renal Dialysis
13.
Semin Dial ; 36(6): 462-467, 2023.
Article En | MEDLINE | ID: mdl-37170692

INTRODUCTION: Spontaneous tendon rupture (STR) is one of the complications related to hyperparathyroidism. This study aims to verify this and to elucidate the feasibility of combined incidence rate of STR and bony fracture to assess clinical management of renal bone disease in dialysis cohort. METHOD: This is a clinical audit of cases of STR and fracture with 5504 patient-year dialysis vintage over 10 years. In order to verify the risk factor, comparison of cases of tendon rupture, the gender, and dialysis vintage matched patients without tendon rupture were done, followed by comparison with post-parathyroidectomy patients. RESULT: Six cases of STR involving eight tendons were identified, including a case of concurrent tendon rupture and bony fracture. These include two cases of double tendons ruptures. During this time, there were 15 cases of bony fracture without tendon rupture. The overall incidence rate for STR and fracture was of 0.0011 and 0.0029 incidence per year of dialysis vintage or one case per 917 and 344 patient-year dialysis vintage, respectively. For patients with PTH ≥ 600 pg/mL, the incidence rate of tendon rupture and fracture was 0.0199 and 0.0430 incidence per person-years or one case per 50 and 23 person-years, respectively. For patients with PTH < 600 pg/mL, the respective rate was 0 and 0.0006 incidence per person-years or one case per >5202 and 1734 person-years. There was significant difference for incidence rates of tendon rupture and fracture between these two groups, with six incidences of tendon rupture per 302 patient-dialysis-years of PTH ≥ 600 pg/mL versus 0 incidence per 5202 patient-year dialysis vintage of PTH < 600 pg/mL (p < 0.001). In similar comparison, there was also significant difference for incidence rate of fracture with 13/302 versus 2/5202 (p < 0.001). In the 5 yearly audit over 10 years, the incidence rates of tendon rupture and fracture have dropped from 0.0052 to 0.0028 or one incidence per 192 to 356 person-years. Among 35 patients post-parathyroidectomy, there was an incidence of tendon rupture in a patient with recurrence of hyperparathyroidism, 5 years after surgery. Comparing six survived patients with tendon rupture versus 12 age-gender-dialysis vintage matched patients, hyperparathyroidism has been shown to be most important risk factor. And subsequently, comparing them with six post-parathyroidectomy patients, reduction of alkaline phosphatase (ALP) has been shown to be associated with lower risk of tendon rupture. Median survival in patients with history of tendon rupture and those with history of fracture was 5.9 and 2.2 years, respectively (p = 0.192). CONCLUSION: Although rare, end-stage renal failure patients on dialysis with PTH > 600 pg/mL had high risk of tendon rupture and bony fracture. Parathyroidectomy might reduce the risk of tendon rupture and fracture with lowering ALP signifying reduction in bone turn over. Combined incidence rate of tendon rupture and fracture could be used to assess the control of hyperparathyroidism related issues in dialysis center.


Chronic Kidney Disease-Mineral and Bone Disorder , Fractures, Bone , Hyperparathyroidism, Secondary , Kidney Failure, Chronic , Tendon Injuries , Humans , Incidence , Renal Dialysis/adverse effects , Hyperparathyroidism, Secondary/etiology , Kidney Failure, Chronic/complications , Kidney Failure, Chronic/therapy , Rupture, Spontaneous/epidemiology , Rupture, Spontaneous/complications , Tendon Injuries/etiology , Tendon Injuries/complications , Chronic Kidney Disease-Mineral and Bone Disorder/complications , Tendons , Parathyroid Hormone
14.
J Bone Miner Res ; 38(8): 1192-1207, 2023 08.
Article En | MEDLINE | ID: mdl-37191192

Chronic kidney disease (CKD) is characterized by kidney damage and loss of renal function. CKD mineral and bone disorder (CKD-MBD) describes the dysregulation of mineral homeostasis, including hyperphosphatemia and elevated parathyroid hormone (PTH) secretion, skeletal abnormalities, and vascular calcification. CKD-MBD impacts the oral cavity, with effects including salivary gland dysfunction, enamel hypoplasia and damage, increased dentin formation, decreased pulp volume, pulp calcifications, and altered jaw bones, contributing to clinical manifestations of periodontal disease and tooth loss. Underlying mechanisms are not fully understood, and CKD mouse models commonly require invasive procedures with high rates of infection and mortality. We aimed to characterize the dentoalveolar effects of an adenine diet (AD)-induced CKD (AD-CKD) mouse model. Eight-week-old C57BL/6J mice were provided either a normal phosphorus diet control (CTR) or adenine and high-phosphorus diet CKD to induce kidney failure. Mice were euthanized at 15 weeks old, and mandibles were collected for micro-computed tomography and histology. CKD mice exhibited kidney failure, hyperphosphatemia, and hyperparathyroidism in association with porous cortical bone in femurs. CKD mice showed a 30% decrease in molar enamel volume compared to CTR mice. Enamel wear was associated with reduced ductal components, ectopic calcifications, and altered osteopontin (OPN) deposition in submandibular salivary glands of CKD mice. Molar cusps in CKD mice were flattened, exposing dentin. Molar dentin/cementum volume increased 7% in CKD mice and pulp volume decreased. Histology revealed excessive reactionary dentin and altered pulp-dentin extracellular matrix proteins, including increased OPN. Mandibular bone volume fraction decreased 12% and bone mineral density decreased 9% in CKD versus CTR mice. Alveolar bone in CKD mice exhibited increased tissue-nonspecific alkaline phosphatase localization, OPN deposition, and greater osteoclast numbers. AD-CKD recapitulated key aspects reported in CKD patients and revealed new insights into CKD-associated oral defects. This model has potential for studying mechanisms of dentoalveolar defects or therapeutic interventions. © 2023 The Authors. Journal of Bone and Mineral Research published by Wiley Periodicals LLC on behalf of American Society for Bone and Mineral Research (ASBMR).


Chronic Kidney Disease-Mineral and Bone Disorder , Hyperphosphatemia , Renal Insufficiency, Chronic , Mice , Animals , Chronic Kidney Disease-Mineral and Bone Disorder/complications , Adenine , X-Ray Microtomography , Hyperphosphatemia/complications , Mice, Inbred C57BL , Renal Insufficiency, Chronic/complications , Phosphorus
15.
J Ren Nutr ; 33(6): 717-730, 2023 Nov.
Article En | MEDLINE | ID: mdl-37116624

INTRODUCTION: Plant-based protein is of growing interest for dietary management of chronic kidney disease (CKD) and is hypothesized to preserve kidney function and reduce CKD-mineral bone disorder (MBD) complications, among other benefits. This systematic review aimed to summarize the available clinical trial evidence for the effect of plant-based protein on kidney function and CKD-MBD outcomes in adults with stage 3-5 CKD not on dialysis. METHODS: Searches of Medline, Embase, Agricola, CAB abstracts, Web of Science, Scopus, and hand searching were performed. Clinical trials with ≥8 participants ≥18 years of age with an estimated glomerular filtration rate <60 mL/min/1.73 m2 but not on dialysis were included. Additionally, only clinical trials with ≥1-week interventions with ≥50% dietary protein from plant-based sources and reported at least one outcome for both kidney function and CKD-MBD outcomes were included. Of the 10,962 identified abstracts, 32 met inclusion criteria and were assessed for risk of bias. RESULTS: Results for kidney function and CKD-MBD outcomes were heterogenous, with most studies having suboptimal methodological quality. In most of the studies (27/32), protein source was altered only secondarily to low-protein diet interventions. Thus, data synthesis and interpretation were focused on a subset of five studies that investigated a change in protein source only (i.e., animal vs. plant). Of this subset, four studies reported no change in kidney function, while one study reported a decrease. Three studies reported no change in serum phosphorus, and one study reported lower serum phosphorus following a vegetarian diet. Further, limited data and inconclusive results were observed for phosphaturic hormones, parathyroid hormone, and fibroblast growth factor-23. CONCLUSION: Current clinical trial evidence on plant-based protein interventions for preserving kidney function and preventing CKD-MBD is limited to inform clinical guidelines at this time. This systematic review emphasizes the ongoing need to research the effects of plant-based protein on kidney function and CKD-MBD outcomes.


Chronic Kidney Disease-Mineral and Bone Disorder , Kidney Failure, Chronic , Renal Insufficiency, Chronic , Adult , Humans , Chronic Kidney Disease-Mineral and Bone Disorder/complications , Plant Proteins , Renal Insufficiency, Chronic/complications , Parathyroid Hormone , Minerals , Phosphorus , Dietary Proteins , Kidney
16.
Curr Osteoporos Rep ; 21(2): 193-204, 2023 04.
Article En | MEDLINE | ID: mdl-36848027

PURPOSES OF REVIEW: With chronic kidney disease (CKD) progression, secondary hyperparathyroidism (sHPT) and mineral and bone metabolism disease (MBD) almost inevitably develop and result in renal osteodystrophy and cardiovascular disease (CVD). Together with active vitamin D, calcimimetics are the main therapy for sHPT in CKD. This review provides an overview of the therapeutic effects of oral cinacalcet and intravenous etelcalcetide on CKD-MBD and vascular disease, with a focus on pediatric dialysis patients. RECENT FINDINGS: Randomized controlled trials in adults and children demonstrate efficient lowering of parathyroid hormone (PTH) by the calcimimetics together with a reduction in serum calcium and phosphate when combined with low-dose active vitamin D, while therapy with active vitamin D analogs alone increases serum calcium and phosphate. Cinacalcet and etelcalcetide both improve bone formation and correct adynamic bone, i.e., have a direct bone anabolic effect. They decrease serum calciprotein particles, which are involved in endothelial dysfunction, atherogenesis, and vascular calcification. Clinical trials in adults suggest a modest slowing of the progression of cardiovascular calcification with cinacalcet. Calcimimetic agents represent a major pharmacological tool for improved control of CKD-MBD, by efficiently counteracting sHPT and allowing for better control of calcium/phosphate and bone homeostasis. Albeit definite evidence is lacking, the beneficial effects of calcimimetics on CVD are promising. Routine use of cinacalcet has been suggested in children.


Cardiovascular Diseases , Chronic Kidney Disease-Mineral and Bone Disorder , Hyperparathyroidism, Secondary , Renal Insufficiency, Chronic , Adult , Humans , Child , Cinacalcet/therapeutic use , Renal Dialysis , Calcium/therapeutic use , Chronic Kidney Disease-Mineral and Bone Disorder/drug therapy , Chronic Kidney Disease-Mineral and Bone Disorder/complications , Hyperparathyroidism, Secondary/drug therapy , Hyperparathyroidism, Secondary/etiology , Calcimimetic Agents/therapeutic use , Parathyroid Hormone , Vitamin D/therapeutic use , Renal Insufficiency, Chronic/complications , Renal Insufficiency, Chronic/therapy , Minerals , Phosphates/metabolism
17.
Curr Med Chem ; 30(7): 841-856, 2023.
Article En | MEDLINE | ID: mdl-35761503

Fibroblast growth factor 23 (FGF23) is a new endocrine product discovered in the past decade. In addition to being related to bone diseases, it has also been found to be related to kidney metabolism and parathyroid metabolism, especially as a biomarker and a key factor to be used in kidney diseases. FGF23 is upregulated as early as the second and third stages of chronic kidney disease (CKD) in response to relative phosphorus overload. The early rise of FGF23 has a protective effect on the body and is essential for maintaining phosphate balance. However, with the decline in renal function, eGFR (estimated glomerular filtration rate) declines, and the phosphorus excretion effect caused by FGF23 is weakened. It eventually leads to a variety of complications, such as bone disease (Chronic Kidney Disease-Mineral and Bone Metabolism Disorder), vascular calcification (VC), and more. Monoclonal antibodies against FGF23 are currently used to treat genetic diseases with increased FGF23. CKD is also a state of increased FGF23. This article reviews the current role of FGF23 in CKD and discusses the crosstalk between various organs under CKD conditions and FGF23. Studying the effect of hyperphosphatemia on different organs of CKD is important. The prospect of FGF23 for therapy is also discussed.


Bone Diseases, Metabolic , Chronic Kidney Disease-Mineral and Bone Disorder , Fibroblast Growth Factor-23 , Renal Insufficiency, Chronic , Humans , Bone Diseases, Metabolic/complications , Bone Diseases, Metabolic/metabolism , Chronic Kidney Disease-Mineral and Bone Disorder/complications , Phosphates , Phosphorus , Renal Insufficiency, Chronic/metabolism , Fibroblast Growth Factor-23/metabolism
18.
Clin Exp Nephrol ; 27(2): 179-187, 2023 Feb.
Article En | MEDLINE | ID: mdl-36303046

BACKGROUND: It is necessary to re-examine the optimal phosphate (P) and calcium (Ca) target values in the contemporary management of chronic kidney disease-mineral and bone disorder to reduce the risks of cardiovascular events in patients receiving hemodialysis. METHODS: We performed a post-hoc analysis of the LANDMARK study. The outcomes were defined as cardiovascular events and all-cause death. Data from 2135 patients receiving hemodialysis at risk of vascular calcification were analyzed using a time-dependent Cox proportional hazard model adjusted for background factors. RESULTS: On the hazard ratio (HR) curve, the ranges where the lower 95% confidence interval (CI) were below the minimum of HR (= 1.00) were as follows: P = 3.5-5.5 mg/dL; albumin-adjusted Ca < 9.1 mg/dL for cardiovascular events; and P = 3.6-5.3 mg/dL; albumin-adjusted Ca < 9.1 mg/dL for all-cause mortality. In stratified analysis, the HRs for cardiovascular events in P < 3.5 mg/dL and P ≥ 5.5 mg/dL were similar to that of P = 3.5-5.5 mg/dL (P ≥ 0.05), and albumin-adjusted Ca ≥ 9.1 mg/dL had higher HR than values < 9.1 mg/dL [1.30 (95% CI 1.00-1.68; P = 0.046)]. For all-cause mortality, the HR in P < 3.6 mg/dL was higher than that in P = 3.6-5.3 mg/dL [1.76 (95% CI 1.25-2.48; P = 0.001)], while the HRs between P ≥ 5.3 mg/dL and P = 3.6-5.3 mg/dL as well as those between albumin-adjusted Ca ≥ 9.1 and < 9.1 mg/dL were not significantly different (P ≥ 0.05). CONCLUSIONS: Managing albumin-adjusted Ca < 9.1 mg/dL may reduce the cardiovascular risk among patients undergoing hemodialysis. Hypophosphatemia < 3.6 mg/dL may be associated with mortality.


Chronic Kidney Disease-Mineral and Bone Disorder , Renal Dialysis , Humans , Albumins , Calcium/blood , Calcium/chemistry , Cardiovascular Diseases/etiology , Cardiovascular Diseases/prevention & control , Phosphates/blood , Phosphates/chemistry , Renal Dialysis/adverse effects , Renal Dialysis/standards , Renal Insufficiency, Chronic/complications , Renal Insufficiency, Chronic/therapy , Chronic Kidney Disease-Mineral and Bone Disorder/complications , Chronic Kidney Disease-Mineral and Bone Disorder/therapy , Hypophosphatemia/etiology
19.
Endocrine ; 79(1): 180-189, 2023 01.
Article En | MEDLINE | ID: mdl-36184719

PURPOSE: Renal osteodystrophy (ROD), a component of chronic kidney disease-mineral and bone disorder (CKD-MBD) can lead to bone loss increasing fracture risks in CKD patients. Therefore, it is important to prevent and treat ROD. Activation of hypoxia-inducible factor-1α (HIF-1α) signaling was reported to prevent osteoporotic bone loss. Roxadustat, which is used to treat renal anemia in the clinic, is a novel HIF stabilizer. In our study, we aimed to investigate the effects of roxadustat on ROD. METHODS: We established an adenine-induced CKD rat model. Roxadustat was administered intragastrically to normal and CKD rats for 4 weeks. Hemoglobin concentrations and serum biochemical parameters were tested, and bone histomorphometric analysis was performed. RESULTS: CKD rats exhibited impaired renal function with anemia, secondary hyperparathyroidism and high-turnover ROD-induced significant bone loss. Roxadustat ameliorated renal anemia and attenuated the extreme increase in intact parathyroid hormone (iPTH) and fibroblast growth factor 23 (FGF23) in CKD rats. Bone histomorphometric analysis showed that roxadustat significantly alleviated bone loss and bone microarchitecture deterioration in CKD rats by increasing osteoblast activity and inhibiting osteoclast activity. We did not find that roxadustat had significant effects on bone metabolism in normal rats. CONCLUSION: Roxadustat can improve ROD via dual regulation of bone remodeling. The use of roxadustat may be a promising strategy to treat osteoporotic bone disorders, such as ROD.


Anemia , Chronic Kidney Disease-Mineral and Bone Disorder , Osteoporosis , Renal Insufficiency, Chronic , Rats , Animals , Chronic Kidney Disease-Mineral and Bone Disorder/drug therapy , Chronic Kidney Disease-Mineral and Bone Disorder/complications , Bone Remodeling , Anemia/complications , Renal Insufficiency, Chronic/complications , Renal Insufficiency, Chronic/drug therapy , Osteoporosis/complications
20.
Nephrol Dial Transplant ; 38(6): 1397-1404, 2023 05 31.
Article En | MEDLINE | ID: mdl-35977397

The situation of secondary hyperparathyroidism (SHPT) in chronic kidney disease patients not on dialysis (ND-CKD) is probably best characterised by the Kidney Disease: Improving Global Outcomes Chronic Kidney Disease-Mineral and Bone Disorder Update 2017 guideline 4.2.1 stating that the optimal parathyroid hormone levels are not known in these stages. Furthermore, new caution became recommended with regard to the routine use of active vitamin D analogues in early CKD stages and moderate SHPT phenotypes, due to their potential risks for hypercalcaemia and hyperphosphataemia aggravation. Nevertheless, there is still a substantial clinical need to prevent the development of parathyroid gland autonomy, with its associated consequences of bone and vascular damage, including fracture risks and cardiovascular events. Therefore we now attempt to review the current guideline-based and clinical practice management of SHPT in ND-CKD, including their strengths and weaknesses, favouring individualised approaches respecting calcium and phosphate homeostasis. We further comment on extended-release calcifediol (ERC) as a new differential therapeutic option now also available in Europe and on a potentially novel understanding of a required vitamin D saturation in more advanced CKD stages. There is no doubt, however, that knowledge gaps will remain unless powerful randomised controlled trials with hard and meaningful endpoints are performed.


Chronic Kidney Disease-Mineral and Bone Disorder , Hyperparathyroidism, Secondary , Renal Insufficiency, Chronic , Humans , Renal Dialysis/adverse effects , Hyperparathyroidism, Secondary/drug therapy , Hyperparathyroidism, Secondary/etiology , Renal Insufficiency, Chronic/therapy , Renal Insufficiency, Chronic/drug therapy , Vitamin D/therapeutic use , Chronic Kidney Disease-Mineral and Bone Disorder/therapy , Chronic Kidney Disease-Mineral and Bone Disorder/complications , Parathyroid Hormone/therapeutic use
...