Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 3.667
Filtrar
Mais filtros

Intervalo de ano de publicação
1.
Kidney Int ; 105(2): 338-346, 2024 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-37918791

RESUMO

Precise determination of circulating parathyroid hormone (PTH) concentration is crucial to diagnose and manage various disease conditions, including the chronic kidney disease-mineral and bone disorder. However, the lack of standardization in PTH assays is challenging for clinicians, potentially leading to medical errors because the different assays do not provide equivalent results and use different reference ranges. Here, we aimed to evaluate the impact of recalibrating PTH immunoassays by means of a recently developed LC-MS/MS method as the reference. Utilizing a large panel of pooled plasma samples with PTH concentrations determined by the LC-MS/MS method calibrated with the World Health Organization (WHO) 95/646 International Standard, five PTH immunoassays were recalibrated. The robustness of this standardization was evaluated over time using different sets of samples. The recalibration successfully reduced inter-assay variability with harmonization of PTH measurements across different assays. By recalibrating the assays based on the WHO 95/646 International Standard, we demonstrated the feasibility for standardizing PTH measurement results and adopting common reference ranges for PTH assays, facilitating a more consistent interpretation of PTH values. The recalibration process aligns PTH results obtained from various immunoassays with the LC-MS/MS method, providing more consistent and reliable measurements. Thus, establishing true standardization across all PTH assays is crucial to ensure consistent interpretation and clinical decision-making.


Assuntos
Distúrbio Mineral e Ósseo na Doença Renal Crônica , Insuficiência Renal Crônica , Humanos , Cromatografia Líquida/métodos , Espectrometria de Massas em Tandem , Hormônio Paratireóideo , Insuficiência Renal Crônica/diagnóstico
2.
Curr Opin Nephrol Hypertens ; 33(2): 226-230, 2024 Mar 01.
Artigo em Inglês | MEDLINE | ID: mdl-38088374

RESUMO

PURPOSE OF REVIEW: The aim of this review is to highlight recent evidence on the role of the gastrointestinal tract and gut microbiome on chronic kidney disease-mineral bone disorder (CKD-MBD) outcomes, including intestinal phosphorus absorption and sensing, and the effect of gut-oriented therapies. RECENT FINDINGS: Recent evidence has revealed a complex interplay among mineral metabolism and novel gut-related factors, including paracellular intestinal phosphate absorption, the gut microbiome, and the immune system, prompting a reevaluation of treatment approaches for CKD-MBD. The inhibition of NHE3 limits phosphate transport in the intestine and may lead to changes in the gut microbiome. A study in rats with CKD showed that the supplementation of the fermentable dietary inulin delayed CKD-MBD, lowering circulating phosphorus and parathyroid hormone, reducing bone remodeling and improving cortical parameters, and lowering cardiovascular calcifications. In non-CKD preclinical studies, probiotics and prebiotics improved bone formation mediated through the effect of butyrate facilitating the differentiation of T cells into Tregs, and Tregs stimulating the osteogenic Wnt10b, and butyrate was also necessary for the parathyroid hormone (PTH) bone effects. SUMMARY: Recent findings support multiple possible roles for gut-oriented therapies in addressing CKD-MBD prevention and management that should be further explored through clinical and translational studies.


Assuntos
Distúrbio Mineral e Ósseo na Doença Renal Crônica , Microbioma Gastrointestinal , Insuficiência Renal Crônica , Humanos , Ratos , Animais , Distúrbio Mineral e Ósseo na Doença Renal Crônica/tratamento farmacológico , Hormônio Paratireóideo , Fósforo , Fosfatos , Minerais , Butiratos , Trato Gastrointestinal
3.
Curr Opin Nephrol Hypertens ; 33(4): 383-390, 2024 07 01.
Artigo em Inglês | MEDLINE | ID: mdl-38651491

RESUMO

PURPOSE OF REVIEW: Defining the optimal parathyroid hormone (PTH) target in chronic kidney disease (CKD) is challenging, especially for bone outcomes, due to the substantial variability in the skeleton's response to PTH. Although PTH hyporesponsiveness is as integral a component of CKD-mineral bone disorder as elevated PTH levels, clinical awareness of this condition is limited. In this review, we will discuss factors and mechanisms contributing to PTH hyporesponsiveness in CKD. This knowledge may provide clues towards a personalized approach to treating secondary hyperparathyroidism in CKD. RECENT FINDINGS: Indicates a link between disturbed phosphate metabolism and impaired skeletal calcium sensing receptor signaling as an important mediator of PTH hyporesponsiveness in CKD. Further, cohort studies with diverse populations point towards differences in mineral metabolism control, rather than genetic or environmental factors, as drivers of the variability of PTH responsiveness. IN SUMMARY: Skeletal PTH hyporesponsiveness in CKD has a multifactorial origin, shows important interindividual variability, and is challenging to estimate in clinical practice. The variability in skeletal responsiveness compromises PTH as a biomarker of bone turnover, especially when considering populations that are heterogeneous in ethnicity, demography, kidney function, primary kidney disease and mineral metabolism control, and in patients treated with bone targeting drugs.


Assuntos
Hiperparatireoidismo Secundário , Hormônio Paratireóideo , Insuficiência Renal Crônica , Humanos , Hormônio Paratireóideo/metabolismo , Hormônio Paratireóideo/sangue , Insuficiência Renal Crônica/tratamento farmacológico , Insuficiência Renal Crônica/metabolismo , Hiperparatireoidismo Secundário/tratamento farmacológico , Hiperparatireoidismo Secundário/etiologia , Hiperparatireoidismo Secundário/metabolismo , Remodelação Óssea/efeitos dos fármacos , Animais , Osso e Ossos/metabolismo , Osso e Ossos/efeitos dos fármacos , Distúrbio Mineral e Ósseo na Doença Renal Crônica/tratamento farmacológico , Distúrbio Mineral e Ósseo na Doença Renal Crônica/metabolismo
4.
Nephrol Dial Transplant ; 39(2): 341-366, 2024 Jan 31.
Artigo em Inglês | MEDLINE | ID: mdl-37697718

RESUMO

Mineral and bone disorders (MBD) are common in patients with chronic kidney disease (CKD), contributing to significant morbidity and mortality. For several decades, the first-line approach to controlling hyperparathyroidism in CKD was by exogenous calcium loading. Since the turn of the millennium, however, a growing awareness of vascular calcification risk has led to a paradigm shift in management and a move away from calcium-based phosphate binders. As a consequence, contemporary CKD patients may be at risk of a negative calcium balance, which, in turn, may compromise bone health, contributing to renal bone disease and increased fracture risk. A calcium intake below a certain threshold may be as problematic as a high intake, worsening the MBD syndrome of CKD, but is not addressed in current clinical practice guidelines. The CKD-MBD and European Renal Nutrition working groups of the European Renal Association (ERA), together with the CKD-MBD and Dialysis working groups of the European Society for Pediatric Nephrology (ESPN), developed key evidence points and clinical practice points on calcium management in children and adults with CKD across stages of disease. These were reviewed by a Delphi panel consisting of ERA and ESPN working groups members. The main clinical practice points include a suggested total calcium intake from diet and medications of 800-1000 mg/day and not exceeding 1500 mg/day to maintain a neutral calcium balance in adults with CKD. In children with CKD, total calcium intake should be kept within the age-appropriate normal range. These statements provide information and may assist in decision-making, but in the absence of high-level evidence must be carefully considered and adapted to individual patient needs.


Assuntos
Doenças Ósseas , Fosfatos de Cálcio , Distúrbio Mineral e Ósseo na Doença Renal Crônica , Insuficiência Renal Crônica , Adulto , Criança , Humanos , Distúrbio Mineral e Ósseo na Doença Renal Crônica/etiologia , Distúrbio Mineral e Ósseo na Doença Renal Crônica/terapia , Cálcio , Diálise Renal , Insuficiência Renal Crônica/terapia , Insuficiência Renal Crônica/tratamento farmacológico , Rim
5.
Pediatr Nephrol ; 39(3): 723-739, 2024 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-37624528

RESUMO

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.


Assuntos
Doenças Ósseas Metabólicas , Distúrbio Mineral e Ósseo na Doença Renal Crônica , Insuficiência Renal Crônica , Calcificação Vascular , Criança , Humanos , Idoso , Adulto Jovem , Adulto , Cálcio , Insuficiência Renal Crônica/complicações , Calcificação Vascular/etiologia , Minerais , Distúrbio Mineral e Ósseo na Doença Renal Crônica/complicações
6.
Curr Osteoporos Rep ; 22(1): 69-79, 2024 02.
Artigo em Inglês | MEDLINE | ID: mdl-38195897

RESUMO

PURPOSE OF REVIEW: This review aims to describe the pathogenic factors involved in bone-vessel anomalies in CKD which are the object of numerous experimental and clinical research. RECENT FINDINGS: Knowledge on the pathophysiological mechanisms involved in the regulation of vascular calcification and mineral-bone disorders is evolving. Specific bone turnover anomalies influence the vascular health while recent studies demonstrate that factors released by the calcified vessels also contribute to bone deterioration in CKD. Current therapies used to control mineral dysregulations will impact both the vessels and bone metabolism. Available anti-osteoporotic treatments used in non-CKD population may negatively or positively affect vascular health in the context of CKD. It is essential to study the bone effects of the new therapeutic options that are currently under investigation to reduce vascular calcification. Our paper highlights the complexity of the bone-vascular axis and discusses how current therapies may affect both organs in CKD.


Assuntos
Distúrbio Mineral e Ósseo na Doença Renal Crônica , Insuficiência Renal Crônica , Calcificação Vascular , Humanos , Distúrbio Mineral e Ósseo na Doença Renal Crônica/terapia , Insuficiência Renal Crônica/complicações , Insuficiência Renal Crônica/metabolismo , Osso e Ossos/metabolismo , Minerais
7.
BMC Med Inform Decis Mak ; 24(1): 36, 2024 Feb 05.
Artigo em Inglês | MEDLINE | ID: mdl-38317140

RESUMO

INTRODUCTION: Chronic kidney disease-mineral and bone disorder (CKD-MBD) is characterized by bone abnormalities, vascular calcification, and some other complications. Although there are diagnostic criteria for CKD-MBD, in situations when conducting target feature examining are unavailable, there is a need to investigate and discover alternative biochemical criteria that are easy to obtain. Moreover, studying the correlations between the newly discovered biomarkers and the existing ones may provide insights into the underlying molecular mechanisms of CKD-MBD. METHODS: We collected a cohort of 116 individuals, consisting of three subtypes of CKD-MBD: calcium abnormality, phosphorus abnormality, and PTH abnormality. To identify the best biomarker panel for discrimination, we conducted six machine learning prediction methods and employed a sequential forward feature selection approach for each subtype. Additionally, we collected a separate prospective cohort of 114 samples to validate the discriminative power of the trained prediction models. RESULTS: Using machine learning under cross validation setting, the feature selection method selected a concise biomarker panel for each CKD-MBD subtype as well as for the general one. Using the consensus of these features, best area under ROC curve reached up to 0.95 for the training dataset and 0.74 for the perspective dataset, respectively. DISCUSSION/CONCLUSION: For the first time, we utilized machine learning methods to analyze biochemical criteria associated with CKD-MBD. Our aim was to identify alternative biomarkers that could serve not only as early detection indicators for CKD-MBD, but also as potential candidates for studying the underlying molecular mechanisms of the condition.


Assuntos
Distúrbio Mineral e Ósseo na Doença Renal Crônica , Insuficiência Renal Crônica , Humanos , Distúrbio Mineral e Ósseo na Doença Renal Crônica/diagnóstico , Estudos Prospectivos , Biomarcadores , Cálcio , Insuficiência Renal Crônica/diagnóstico
8.
J Am Soc Nephrol ; 34(4): 668-681, 2023 04 01.
Artigo em Inglês | MEDLINE | ID: mdl-36749125

RESUMO

SIGNIFICANCE STATEMENT: Renal osteodystrophy (ROD) contributes substantially to morbidity in CKD, including increased fracture risk. Metabolic acidosis (MA) contributes to the development of ROD, but an up-to-date skeletal phenotype in CKD-associated acidosis has not been described. We comprehensively studied associations between acidosis and bone in patients with CKD using advanced methods to image the skeleton and analyze bone-tissue, along with biochemical testing. Cross-sectionally, acidosis was associated with higher markers of bone remodeling and female-specific impairments in cortical and trabecular bone quality. Prospectively, acidosis was associated with cortical expansion and trabecular microarchitectural deterioration. At the bone-tissue level, acidosis was associated with deficits in bone mineral content. Future work investigating acidosis correction on bone quality is warranted. BACKGROUND: Renal osteodystrophy is a state of impaired bone quality and strength. Metabolic acidosis (MA) is associated with alterations in bone quality including remodeling, microarchitecture, and mineralization. No studies in patients with CKD have provided a comprehensive multimodal skeletal phenotype of MA. We aim to describe the structure and makeup of bone in patients with MA in the setting of CKD using biochemistry, noninvasive imaging, and histomorphometry. METHODS: The retrospective cross-sectional analyses included 180 patients with CKD. MA was defined as bicarbonate ≤22 mEq/L. We evaluated circulating bone turnover markers and skeletal imaging with dual energy x-ray absorptiometry and high-resolution peripheral computed tomography. A subset of 54 participants had follow-up. We assessed associations between baseline and change in bicarbonate with change in bone outcomes. Histomorphometry, microCT, and quantitative backscatter electron microscopy assessed bone biopsy outcomes in 22 participants. RESULTS: The mean age was 68±10 years, 54% of participants were male, and 55% were White. At baseline, acidotic subjects had higher markers of bone turnover, lower areal bone mineral density at the radius by dual energy x-ray absorptiometry, and lower cortical and trabecular volumetric bone mineral density and impaired trabecular microarchitecture. Over time, acidosis was associated with opposing cortical and trabecular effects: cortical expansion but trabecular deterioration. Bone-tissue analyses showed reduced tissue mineral density with increased heterogeneity of calcium distribution in acidotic participants. CONCLUSIONS: MA is associated with multiple impairments in bone quality. Future work should examine whether correction of acidosis improves bone quality and strength in patients with CKD.


Assuntos
Acidose , Distúrbio Mineral e Ósseo na Doença Renal Crônica , Insuficiência Renal Crônica , Masculino , Feminino , Humanos , Distúrbio Mineral e Ósseo na Doença Renal Crônica/etiologia , Estudos Transversais , Estudos Retrospectivos , Bicarbonatos , Densidade Óssea , Rádio (Anatomia) , Insuficiência Renal Crônica/complicações , Insuficiência Renal Crônica/patologia , Acidose/complicações
9.
Int J Mol Sci ; 25(3)2024 Feb 03.
Artigo em Inglês | MEDLINE | ID: mdl-38339137

RESUMO

Kidney transplantation is the preferred gold standard modality of treatment for kidney failure. Bone disease after kidney transplantation is highly prevalent in patients living with a kidney transplant and is associated with high rates of hip fractures. Fractures are associated with increased healthcare costs, morbidity and mortality. Post-transplant bone disease (PTBD) includes renal osteodystrophy, osteoporosis, osteonecrosis and bone fractures. PTBD is complex as it encompasses pre-existing chronic kidney disease-mineral bone disease and compounding factors after transplantation, including the use of immunosuppression and the development of de novo bone disease. After transplantation, the persistence of secondary and tertiary hyperparathyroidism, renal osteodystrophy, relative vitamin D deficiency and high levels of fibroblast growth factor-23 contribute to post-transplant bone disease. Risk assessment includes identifying both general risk factors and kidney-specific risk factors. Diagnosis is complex as the gold standard bone biopsy with double-tetracycline labelling to diagnose the PTBD subtype is not always readily available. Therefore, alternative diagnostic tools may be used to aid its diagnosis. Both non-pharmacological and pharmacological therapy can be employed to treat PTBD. In this review, we will discuss pathophysiology, risk assessment, diagnosis and management strategies to manage PTBD after kidney transplantation.


Assuntos
Distúrbio Mineral e Ósseo na Doença Renal Crônica , Fraturas Ósseas , Transplante de Rim , Osteoporose , Deficiência de Vitamina D , Humanos , Transplante de Rim/efeitos adversos , Distúrbio Mineral e Ósseo na Doença Renal Crônica/diagnóstico , Distúrbio Mineral e Ósseo na Doença Renal Crônica/etiologia , Distúrbio Mineral e Ósseo na Doença Renal Crônica/terapia , Osteoporose/etiologia , Fraturas Ósseas/etiologia , Deficiência de Vitamina D/complicações , Densidade Óssea/fisiologia
10.
Int J Mol Sci ; 25(3)2024 Feb 03.
Artigo em Inglês | MEDLINE | ID: mdl-38339121

RESUMO

Shortly after the discovery of Klotho, interest grew in its potential role in chronic kidney disease (CKD). There are three isoforms of the Klotho protein: αKlotho, ßKlotho and γKlotho. This review will focus on αKlotho due to its relevance as a biomarker in CKD. αKlotho is synthesized mainly in the kidneys, but it can be released into the bloodstream and urine as soluble Klotho (sKlotho), which undertakes systemic actions, independently or in combination with FGF23. It is usually accepted that sKlotho levels are reduced early in CKD and that lower levels of sKlotho might be associated with the main chronic kidney disease-mineral bone disorders (CKD-MBDs): cardiovascular and bone disease. However, as results are inconsistent, the applicability of sKlotho as a CKD-MBD biomarker is still a matter of controversy. Much of the inconsistency can be explained due to low sample numbers, the low quality of clinical studies, the lack of standardized assays to assess sKlotho and a lack of consensus on sample processing, especially in urine. In recent decades, because of our longer life expectancies, the prevalence of accelerated-ageing diseases, such as CKD, has increased. Exercise, social interaction and caloric restriction are considered key factors for healthy ageing. While exercise and social interaction seem to be related to higher serum sKlotho levels, it is not clear whether serum sKlotho might be influenced by caloric restriction. This review focuses on the possible role of sKlotho as a biomarker in CKD-MBD, highlighting the difference between solid knowledge and areas requiring further research, including the role of sKlotho in healthy ageing.


Assuntos
Distúrbio Mineral e Ósseo na Doença Renal Crônica , Envelhecimento Saudável , Proteínas Klotho , Humanos , Biomarcadores , Distúrbio Mineral e Ósseo na Doença Renal Crônica/diagnóstico , Fatores de Crescimento de Fibroblastos , Glucuronidase , Envelhecimento Saudável/metabolismo , Minerais , Insuficiência Renal Crônica/complicações , Proteínas Klotho/sangue , Proteínas Klotho/metabolismo
11.
Curr Opin Nephrol Hypertens ; 32(6): 559-564, 2023 11 01.
Artigo em Inglês | MEDLINE | ID: mdl-37753646

RESUMO

PURPOSE OF REVIEW: Renal osteodystrophy (ROD) is a complex disorder of bone metabolism that affects virtually all adults and children with chronic kidney disease (CKD). ROD is associated with adverse clinical outcomes including bone loss, mineralization and turnover abnormalities, skeletal deformities, fractures, cardiovascular events, and death. Despite current therapies, fracture incidence is 2-fold to 100-fold higher in adults and 2-fold to 3-fold higher in children when compared to without CKD. Limited knowledge of ROD pathogenesis, due to the lack of patient-derived large-scale multimodal datasets, impedes development of therapeutics aimed at reducing morbidity and mortality of CKD patients. The purpose of the review is to define the much needed infrastructure for the advancement of RDO treatment. RECENT FINDINGS: Recently, we created a large-scale data and tissue biorepository integrating clinical, bone quality, transcriptomic, and epigenomic data along with stored urine, blood, and bone samples. This database will provide the underpinnings for future research endeavors leading to the elucidation and characterization of the pathogenesis of ROD in CKD patients with and without dialysis. SUMMARY: The availability of an open-access NIH-funded resource that shares bone-tissue-based information obtained from patients with ROD with the broad scientific community represents a critical step in the process of discovering new information regarding unrecognized bone changes that have severe clinical complications. This will facilitate future high-impact hypothesis-driven research to redefine our understanding of ROD pathogenesis and pathophysiology and inform the development of disease-modifying and prevention strategies.


Assuntos
Doenças Ósseas Metabólicas , Calcinose , Distúrbio Mineral e Ósseo na Doença Renal Crônica , Fraturas Ósseas , Adulto , Criança , Humanos , Distúrbio Mineral e Ósseo na Doença Renal Crônica/epidemiologia , Distúrbio Mineral e Ósseo na Doença Renal Crônica/terapia , Diálise Renal , Osso e Ossos
12.
Am J Kidney Dis ; 81(4): 466-474, 2023 04.
Artigo em Inglês | MEDLINE | ID: mdl-36410592

RESUMO

Development of clinical guidelines and recommendations to address the care of pediatric patients with chronic kidney disease (CKD) has rarely included the perspectives of providers from a variety of health care disciplines or the patients and parents themselves. Accordingly, the National Kidney Foundation hosted an in-person, one and a half-day workshop that convened a multidisciplinary group of physicians, allied health care professionals, and pediatric patients with CKD and their parents, with the goal of developing key clinical recommendations regarding best practices for the clinical management of pediatric patients living with CKD. The key clinical recommendations pertained to 5 broad topics: addressing the needs of patients and parents/caregivers; modifying the progression of CKD; clinical management of CKD-mineral and bone disorder and growth retardation; clinical management of anemia, cardiovascular disease, and hypertension; and transition and transfer of pediatric patients to adult nephrology care. This report describes the recommendations generated by the participants who attended the workshop.


Assuntos
Distúrbio Mineral e Ósseo na Doença Renal Crônica , Nefrologia , Médicos , Insuficiência Renal Crônica , Adulto , Humanos , Criança , Insuficiência Renal Crônica/terapia , Rim
13.
Osteoporos Int ; 34(12): 2003-2012, 2023 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-37658999

RESUMO

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.


Assuntos
Doenças Ósseas Metabólicas , Reabsorção Óssea , Distúrbio Mineral e Ósseo na Doença Renal Crônica , Osteomalacia , Insuficiência Renal Crônica , Humanos , Osteomalacia/complicações , Estudos Prospectivos , Osso e Ossos , Insuficiência Renal Crônica/complicações , Doenças Ósseas Metabólicas/complicações , Distúrbio Mineral e Ósseo na Doença Renal Crônica/complicações
14.
Calcif Tissue Int ; 113(4): 416-425, 2023 10.
Artigo em Inglês | MEDLINE | ID: mdl-37665403

RESUMO

BACKGROUND: Vascular calcification, a component of chronic kidney disease-mineral and bone disorder (CKD-MBD), is prevalent in patients with end-stage kidney disease (ESKD) and contributes to high mortality. However, the association between the blood level of total osteocalcin (OC) and vascular calcification and mortality remains inconclusive. We, therefore, investigated whether different OC fractions can serve as biomarkers of vascular calcification and mortality in the ESKD population. METHODS: This observational cohort study enrolled patients on maintenance hemodialysis. Plasma carboxylated OC (cOC), uncarboxylated OC (ucOC), and intact parathyroid hormone (PTH) were measured. The percentage of carboxylated OC (%cOC) was calculated as dividing cOC by total OC. The vascular calcification severity was defined by an aortic calcification grade. The patients were followed for three years and one month. RESULTS: A total of 184 patients were enrolled. In the multivariable logistic regression, plasma %cOC, but not cOC or ucOC, was independently associated with the severity of vascular calcification (OR 1.019, p = 0.036). A significant U-shaped correlation was found between plasma %cOC and PTH (p = 0.002). In the multivariable Cox regression, patients with higher plasma %cOC had a higher risk of mortality (quartiles Q4 versus Q1-Q3, HR 1.991 [95% CI: 1.036-3.824], p = 0.039). CONCLUSIONS: In patients undergoing chronic hemodialysis, plasma %cOC positively correlated with vascular calcification and exhibited a U-shaped correlation with PTH. Furthermore, a higher plasma %cOC was associated with increased mortality. These findings suggest that plasma %cOC may serve as a biomarker for CKD-MBD and a predictor of clinical outcomes in chronic hemodialysis patients.


Assuntos
Distúrbio Mineral e Ósseo na Doença Renal Crônica , Calcificação Vascular , Humanos , Osteocalcina , Diálise Renal , Ácidos Carboxílicos
15.
Nephrol Dial Transplant ; 38(4): 932-938, 2023 03 31.
Artigo em Inglês | MEDLINE | ID: mdl-35790138

RESUMO

BACKGROUND: Biomarkers of chronic kidney disease-mineral and bone disorder (CKD-MBD) have been implicated in CKD progression in follow-up studies focusing on single measurements of individual biomarkers made at baseline only. The simultaneous relationship between the time trend of these biomarkers over the course of CKD and renal outcomes has never been tested. METHODS: We applied the joint model (JM) to investigate the longitudinal relationship between repeated measurements of CKD-MBD biomarkers and a combined renal endpoint (estimated glomerular filtration rate reduction >30%, dialysis or transplantation) in 729 stage 2-5 CKD patients over a 36-month follow-up. RESULTS: In the survival submodel of the JM, the longitudinal series of parathyroid hormone (PTH) values was directly and independently related to the risk of renal events [hazard ratio (HR) (1 ln increase in parathyroid hormone (PTH) 2.0 (range 1.5-2.8), P < .001)] and this was also true for repeated measurements of serum phosphate [HR (1 mg/dl) 1.3924 (range 1.1459-1.6918), P = .001], serum calcium [HR (1 mg/dl) 0.7487 (range 0.5843-0.9593), P = .022], baseline fibroblast growth factor 23 [HR (1 pg/ml) 1.001 (range 1.00-1.002), P = .045] and 1,25-dihydroxyvitamin D [HR (1 pg/ml) 0.9796 (range 0.9652-0.9942), P = .006]. CONCLUSION: Repeated measurements of serum PTH, calcium and phosphate as well as baseline FGF23 and 1,25-dihydroxyvitamin D are independently related with the progression to kidney failure in a cohort of stage 2-5 CKD patients. This longitudinal study generates the hypothesis that interventions at multiple levels on MBD biomarkers can mitigate renal function loss in this population.


Assuntos
Distúrbio Mineral e Ósseo na Doença Renal Crônica , Insuficiência Renal Crônica , Humanos , Distúrbio Mineral e Ósseo na Doença Renal Crônica/diagnóstico , Distúrbio Mineral e Ósseo na Doença Renal Crônica/etiologia , Cálcio , Estudos Longitudinais , Diálise Renal , Hormônio Paratireóideo , Biomarcadores , Fosfatos , Insuficiência Renal Crônica/complicações , Fatores de Crescimento de Fibroblastos
16.
Nephrol Dial Transplant ; 38(6): 1397-1404, 2023 05 31.
Artigo em Inglês | MEDLINE | ID: mdl-35977397

RESUMO

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.


Assuntos
Distúrbio Mineral e Ósseo na Doença Renal Crônica , Hiperparatireoidismo Secundário , Insuficiência Renal Crônica , Humanos , Diálise Renal/efeitos adversos , Hiperparatireoidismo Secundário/tratamento farmacológico , Hiperparatireoidismo Secundário/etiologia , Insuficiência Renal Crônica/terapia , Insuficiência Renal Crônica/tratamento farmacológico , Vitamina D/uso terapêutico , Distúrbio Mineral e Ósseo na Doença Renal Crônica/terapia , Distúrbio Mineral e Ósseo na Doença Renal Crônica/complicações , Hormônio Paratireóideo/uso terapêutico
17.
Nephrol Dial Transplant ; 38(11): 2562-2575, 2023 Oct 31.
Artigo em Inglês | MEDLINE | ID: mdl-37230954

RESUMO

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.


Assuntos
Distúrbio Mineral e Ósseo na Doença Renal Crônica , Insuficiência Renal Crônica , Humanos , Idoso , Distúrbio Mineral e Ósseo na Doença Renal Crônica/complicações , Cálcio , Hormônio Paratireóideo , Fosfatos , Cálcio da Dieta , Biomarcadores , Insuficiência Renal Crônica/complicações , Insuficiência Renal Crônica/terapia , Diálise Renal
18.
Nephrol Dial Transplant ; 38(8): 1823-1835, 2023 07 31.
Artigo em Inglês | MEDLINE | ID: mdl-36869794

RESUMO

BACKGROUND: This trial aimed to evaluate oral cinacalcet versus total parathyroidectomy (PTx) with forearm autografting on cardiovascular surrogate outcomes and health-related quality of life (HRQOL) measures in dialysis patients with advanced secondary hyperparathyroidism (SHPT). DESIGN: In this pilot prospective randomized trial conducted in two university-affiliated hospitals, 65 adult peritoneal dialysis patients with advanced SHPT were randomized to receive either oral cinacalcet or PTx. Primary endpoints were changes in left ventricular (LV) mass index by cardiac magnetic resonance imaging and coronary artery calcium scores (CACS) over 12 months. Secondary endpoints included changes in heart valves calcium scores, aortic stiffness, biochemical parameters of chronic kidney disease-mineral bone disease (CKD-MBD) and HRQOL measures over 12 months. RESULTS: Changes in LV mass index, CACS, heart valves calcium score, aortic pulse wave velocity and HRQOL did not differ between groups or within groups, despite significant reductions in plasma calcium, phosphorus and intact parathyroid hormone in both groups. Cinacalcet-treated patients experienced more cardiovascular-related hospitalizations than those who underwent PTx (P = .008) but the difference became insignificant after adjusting for baseline difference in heart failure (P = .43). With the same monitoring frequency, cinacalcet-treated patients had fewer hospitalizations due to hypercalcemia (1.8%) than patients who underwent PTx (16.7%) (P = .005). No significant changes were observed in HRQOL measures in either group. CONCLUSIONS: Both cinacalcet and PTx effectively improved various biochemical abnormalities of CKD-MBD and stabilized but did not reduce LV mass, coronary artery and heart valves calcification, or arterial stiffness, or improve patient-centered HRQOL measures in PD patients with advanced SHPT. Cinacalcet may be used in place of PTx for treating advanced SHPT. Long-term and powered studies are required to evaluate PTx versus cinacalcet on hard cardiovascular outcomes in dialysis patients. Trial registration: ClinicalTrials.gov identifier: NCT01447368.


Assuntos
Distúrbio Mineral e Ósseo na Doença Renal Crônica , Cinacalcete , Hiperparatireoidismo Secundário , Falência Renal Crônica , Paratireoidectomia , Diálise Peritoneal , Insuficiência Renal Crônica , Adulto , Humanos , Distúrbio Mineral e Ósseo na Doença Renal Crônica/etiologia , Cinacalcete/administração & dosagem , Cinacalcete/uso terapêutico , Hiperparatireoidismo Secundário/tratamento farmacológico , Hiperparatireoidismo Secundário/cirurgia , Falência Renal Crônica/complicações , Falência Renal Crônica/terapia , Paratireoidectomia/efeitos adversos , Diálise Peritoneal/efeitos adversos , Estudos Prospectivos , Qualidade de Vida , Diálise Renal/efeitos adversos , Insuficiência Renal Crônica/complicações , Insuficiência Renal Crônica/terapia
19.
Semin Dial ; 36(6): 462-467, 2023.
Artigo em Inglês | MEDLINE | ID: mdl-37170692

RESUMO

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.


Assuntos
Distúrbio Mineral e Ósseo na Doença Renal Crônica , Fraturas Ósseas , Hiperparatireoidismo Secundário , Falência Renal Crônica , Traumatismos dos Tendões , Humanos , Incidência , Diálise Renal/efeitos adversos , Hiperparatireoidismo Secundário/etiologia , Falência Renal Crônica/complicações , Falência Renal Crônica/terapia , Ruptura Espontânea/epidemiologia , Ruptura Espontânea/complicações , Traumatismos dos Tendões/etiologia , Traumatismos dos Tendões/complicações , Distúrbio Mineral e Ósseo na Doença Renal Crônica/complicações , Tendões , Hormônio Paratireóideo
20.
Kidney Blood Press Res ; 48(1): 231-240, 2023.
Artigo em Inglês | MEDLINE | ID: mdl-36882038

RESUMO

INTRODUCTION: Chronic kidney disease-mineral and bone disorder (CKD-MBD) is frequently observed in maintenance hemodialysis (MHD) patients and is associated with fracture, muscle weakness, malnutrition, etc.; however, relationships of CKD-MBD markers and fatigue are not well established. METHODS: This was a cross-sectional study including 244 MHD patients (89 elders) from July to September 2021 in the First Affiliated Hospital of Shandong First Medical University. CKD-MBD markers and other clinical data were collected from medical records. Fatigue in the past week was measured by Standardized Outcomes in Nephrology-Hemodialysis (SONG-HD) fatigue measure; fatigue at the end of hemodialysis was measured by numeric rating scale (NRS). Spearman correlation, linear regression, and robust linear regression were. RESULTS: In all MHD patients, lg[25(OH)D] (nmol/L) was negatively correlated with SONG-HD score (ß = -1.503, 95% CI: -2.826 to 0.18, p = 0.026) and NRS score (ß = -1.532, p = 0.04) in multiple regression models adjusting for sex, age, and all CKD-MBD characters; but no correlations were found on univariate regression or in other multiple regression models. Interaction effects between age ≥65 years and lg(25[OH]D [nmol/L]) in terms of fatigue scores were significant based on multiple linear regressions (SONG-HD score ß = -3.613, p for interaction = 0.006; NRS score ß = -3.943, p for interaction = 0.008). Compared with non-elderly patients, elderly patients were with higher ACCI scores (7 [6, 8] vs. 4 [3, 5], p < 0.001), higher SONG-HD scores (3 [2, 6] vs. 2 [1, 3], p < 0.001), higher NRS score (4 [2, 7] vs. 3 [1, 5], p < 0.001), lower serum phosphate levels (1.65 [1.29, 2.10] vs. 1.87 [1.55, 2.26] mmol/L, p = 0.002), and lower serum iPTH levels (160.6 [90.46, 306.45] vs. 282.2 [139, 445.7] pg/mL, p < 0.001). There were no differences in serum calcium, alkaline serum, or 25(OH)D levels between the two groups. In elderly patients, lg[25(OH)D] was negatively correlated with SONG-HD score (ß = -3.323, p = 0.010) and NRS score (ß = -3.521, p = 0.006) on univariate linear regressions. Following adjustment for sex, age, and all CKD-MBD characters, lg[25(OH)D] was negatively correlated with SONG-HD scores (multiple linear regression ß = -4.012, p = 0.004; multiple robust regression ß = -4.012, p = 0.003) or NRS scores (multiple linear regression ß = -4.104, p = 0.002; multiple robust regression ß = -4.104, p = 0.001). There were no significant correlations between fatigue scores and other CKD-MBD markers (calcium, phosphate, lgiPTH, alkaline phosphatase) in elderly MHD patients, on either univariate linear regressions or multiple regressions. CONCLUSION: Serum 25(OH)D level is negatively associated with fatigue in elderly MHD patients.


Assuntos
Distúrbio Mineral e Ósseo na Doença Renal Crônica , Humanos , Idoso , Pessoa de Meia-Idade , Cálcio , Estudos Transversais , Diálise Renal , Fadiga/etiologia , Fosfatos , Hormônio Paratireóideo
SELEÇÃO DE REFERÊNCIAS
Detalhe da pesquisa