1
|
Nogueira de Sa P, Narayanan M, Lim MAC. Electrolyte and Acid-Base Abnormalities After Kidney Transplantation. ADVANCES IN KIDNEY DISEASE AND HEALTH 2024; 31:450-457. [PMID: 39232615 DOI: 10.1053/j.akdh.2024.03.007] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/18/2023] [Revised: 03/14/2024] [Accepted: 03/25/2024] [Indexed: 09/06/2024]
Abstract
Kidney transplantation is the optimal therapeutic approach for individuals with end-stage kidney disease. The Scientific Registry of Transplant Recipients has reported a continuous rise in the total number of kidney transplants performed in the United States, with 25,500 new kidney recipients in 2022 alone. Despite an improved glomerular filtration rate, the post-transplant period introduces a unique set of electrolyte abnormalities that differ from those encountered in chronic kidney disease. A variety of factors contribute to the high prevalence of hypomagnesemia, hyperkalemia, metabolic acidosis, hypercalcemia, and hypophosphatemia seen after kidney transplantation. These include the degree of allograft function, immunosuppressive medications and their diverse mechanisms of action, and metabolic changes after transplant. This article aims to provide a comprehensive review of the key aspects surrounding the most commonly encountered electrolyte and acid-base abnormalities in the post-transplant setting.
Collapse
Affiliation(s)
- Patricia Nogueira de Sa
- Renal-Electrolyte and Hypertension Division, University of Pennsylvania, Perelman School of Medicine, Philadelphia, PA.
| | - Mohanram Narayanan
- Division of Nephrology and Hypertension, Department of Medicine, Baylor Scott & White, Medical Center, Temple, TX
| | - Mary Ann C Lim
- Renal-Electrolyte and Hypertension Division, University of Pennsylvania, Perelman School of Medicine, Philadelphia, PA
| |
Collapse
|
2
|
Jaikaransingh V. Bone health and fracture prevention after kidney transplantation. J Clin Transl Endocrinol 2024; 36:100345. [PMID: 38737624 PMCID: PMC11081796 DOI: 10.1016/j.jcte.2024.100345] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2024] [Revised: 04/21/2024] [Accepted: 04/24/2024] [Indexed: 05/14/2024] Open
Abstract
Changes in bone health and strength are common after kidney transplantation and can lead to an increased risk of fracture. This has implications for morbidity, mortality and renal allograft survival. This review will focus on the changes that occur in bone health and fracture risk after kidney transplantation and examine the evidence available to guide diagnostic and therapeutic decisions with the aim of fracture prevention.
Collapse
Affiliation(s)
- Vishal Jaikaransingh
- University of Florida College of Medicine – Jacksonville, Department of Medicine, Division of Nephrology, United States
| |
Collapse
|
3
|
Ito N, Hidaka N, Kato H. Acquired Forms of Fibroblast Growth Factor 23-Related Hypophosphatemic Osteomalacia. Endocrinol Metab (Seoul) 2024; 39:255-261. [PMID: 38467164 PMCID: PMC11066443 DOI: 10.3803/enm.2023.1908] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/23/2023] [Revised: 01/25/2024] [Accepted: 01/29/2024] [Indexed: 03/13/2024] Open
Abstract
Fibroblast growth factor 23 (FGF23) is a pivotal humoral factor for the regulation of serum phosphate levels and was first identified in patients with autosomal dominant hypophosphatemic rickets and tumor-induced osteomalacia (TIO), the most common form of acquired FGF23-related hypophosphatemic rickets/osteomalacia (FGF23rHR). After the identification of FGF23, many other inherited and acquired forms of FGF23rHR were reported. In this review article, the detailed features of each acquired FGF23rHR are discussed, including TIO, ectopic FGF23 syndrome with malignancy, fibrous dysplasia/McCune-Albright syndrome, Schimmelpenning-Feuerstein-Mims syndrome/cutaneous skeletal hypophosphatemia syndrome, intravenous iron preparation-induced FGF23rHR, alcohol consumption-induced FGF23rHR, and post-kidney transplantation hypophosphatemia. Then, an approach for the differential diagnosis and therapeutic options for each disorder are concisely introduced. Currently, the majority of endocrinologists might only consider TIO when encountering patients with acquired FGF23rHR; an adequate differential diagnosis can reduce medical costs and invasive procedures such as positron emission tomography/computed tomography and venous sampling to identify FGF23-producing tumors. Furthermore, some acquired FGF23rHRs, such as intravenous iron preparation/alcohol consumption-induced FGF23rHR, require only cessation of drugs or alcohol to achieve full recovery from osteomalacia.
Collapse
Affiliation(s)
- Nobuaki Ito
- Division of Nephrology and Endocrinology, The University of Tokyo Hospital, Tokyo, Japan
- Osteoporosis Center, The University of Tokyo Hospital, Tokyo, Japan
| | - Naoko Hidaka
- Division of Nephrology and Endocrinology, The University of Tokyo Hospital, Tokyo, Japan
- Osteoporosis Center, The University of Tokyo Hospital, Tokyo, Japan
| | - Hajime Kato
- Division of Nephrology and Endocrinology, The University of Tokyo Hospital, Tokyo, Japan
- Osteoporosis Center, The University of Tokyo Hospital, Tokyo, Japan
| |
Collapse
|
4
|
Ito N, Hidaka N, Kato H. The pathophysiology of hypophosphatemia. Best Pract Res Clin Endocrinol Metab 2024; 38:101851. [PMID: 38087658 DOI: 10.1016/j.beem.2023.101851] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/27/2024]
Abstract
After identification of fibroblast growth factor (FGF) 23 as the pivotal regulator of chronic serum inorganic phosphate (Pi) levels, the etiology of disorders causing hypophosphatemic rickets/osteomalacia has been clarified, and measurement of intact FGF23 serves as a potent tool for differential diagnosis of chronic hypophosphatemia. Additionally, measurement of bone-specific alkaline phosphatase (BAP) is recommended to differentiate acute and subacute hypophosphatemia from chronic hypophosphatemia. This article divides the etiology of chronic hypophosphatemia into 4 groups: A. FGF23 related, B. primary tubular dysfunction, C. disturbance of vitamin D metabolism, and D. parathyroid hormone 1 receptor (PTH1R) mediated. Each group is further divided into its inherited form and acquired form. Topics for each group are described, including "ectopic FGF23 syndrome," "alcohol consumption-induced FGF23-related hypophosphatemia," "anti-mitochondrial antibody associated hypophosphatemia," and "vitamin D-dependent rickets type 3." Finally, a flowchart for differential diagnosis of chronic hypophosphatemia is introduced.
Collapse
Affiliation(s)
- Nobuaki Ito
- Division of Nephrology and Endocrinology, The University of Tokyo Hospital, Tokyo, Japan; Osteoporosis Center, The University of Tokyo Hospital, Tokyo, Japan.
| | - Naoko Hidaka
- Division of Nephrology and Endocrinology, The University of Tokyo Hospital, Tokyo, Japan; Osteoporosis Center, The University of Tokyo Hospital, Tokyo, Japan.
| | - Hajime Kato
- Division of Nephrology and Endocrinology, The University of Tokyo Hospital, Tokyo, Japan; Osteoporosis Center, The University of Tokyo Hospital, Tokyo, Japan.
| |
Collapse
|
5
|
Ryu JH, Jeon HJ, Han R, Jung HY, Kim MG, Huh KH, Park JB, Kang KP, Han S, Yang J. High pretransplant FGF23 level is associated with persistent vitamin D insufficiency and poor graft survival in kidney transplant patients. Sci Rep 2023; 13:19640. [PMID: 37949967 PMCID: PMC10638428 DOI: 10.1038/s41598-023-46889-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2023] [Accepted: 11/06/2023] [Indexed: 11/12/2023] Open
Abstract
Vitamin D3 (25[OH]D3) insufficiency and fibroblast growth factor 23 (FGF23) elevation are usually attenuated after kidney transplantation (KT). However, elevated FGF23 may be associated with poor graft outcomes and vitamin D insufficiency after KT. This study investigated the effect of pretransplant FGF23 levels on post-KT 25(OH)D3 status and graft outcomes. Serum FGF23 levels from 400 participants of the KoreaN Cohort Study for Outcome in Patients With Kidney Transplantation were measured. Annual serum 25(OH)D3 levels, all-cause mortality, cardiovascular event, and graft survival were assessed according to baseline FGF23 levels. Serum 25(OH)D3 levels were initially increased 1 year after KT (12.6 ± 7.4 vs. 22.6 ± 6.4 ng/mL). However, the prevalence of post-KT vitamin D deficiency increased again after post-KT 3 years (79.1% at baseline, 30.8% and 37.8% at 3 and 6 years, respectively). Serum FGF23 level was decreased 3 years post-KT. When participants were categorized into tertiles according to baseline FGF23 level (low, middle, high), 25(OH)D3 level in the low FGF23 group was persistently low at a median follow-up of 8.3 years. Furthermore, high baseline FGF23 level was a risk factor for poor graft survival (HR 5.882, 95% C.I.; 1.443-23.976, P = 0.013). Elevated FGF23 levels are associated with persistently low post-transplant vitamin D levels and poor graft survival.
Collapse
Affiliation(s)
- Jung-Hwa Ryu
- Department of Internal Medicine, Ewha Womans University Seoul Hospital, Seoul, Republic of Korea
| | - Hee Jung Jeon
- Department of Internal Medicine, Hallym University College of Medicine, Seoul, Republic of Korea
| | - Ro Han
- Department of Internal Medicine, Gachon University Gil Medical Center, Incheon, Republic of Korea
| | - Hee-Yeon Jung
- Department of Internal Medicine, Kyungpook National University Hospital, Daegu, Republic of Korea
| | - Myung-Gyu Kim
- Department of Internal Medicine, Korea University College of Medicine, Seoul, Republic of Korea
| | - Kyu Ha Huh
- Department of Surgery, Yonsei University College of Medicine, Seoul, Republic of Korea
| | - Jae Berm Park
- Department of Surgery, Seoul Samsung Medical Center, Sungkyunkwan University, Seoul, Republic of Korea
| | - Kyung Pyo Kang
- Department of Internal Medicine, Jeonbuk National University Hospital, Jeonju, Republic of Korea
| | - Seungyeup Han
- Department of Internal Medicine, Dongsan Medical Center, Keimyung University, Daegu, Republic of Korea
| | - Jaeseok Yang
- Department of Internal Medicine, Yonsei University College of Medicine, 50-1 Yonsei-ro, Seodaemun-gu, Seoul, 03722, Republic of Korea.
| |
Collapse
|
6
|
Kotwal N, Bansal N, Muthukrishnan J, Verma V. Transplant endocrinology. Med J Armed Forces India 2023; 79:651-656. [PMID: 37981924 PMCID: PMC10654368 DOI: 10.1016/j.mjafi.2023.08.017] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2023] [Accepted: 08/27/2023] [Indexed: 11/21/2023] Open
Abstract
Solid organ transplants and stem cell transplants are becoming more common but a significant proportion of patients are still on waiting lists, awaiting transplants. When endocrinologists treat transplant recipients who have underlying endocrine problems, which might include endocrine emergencies, there are special clinical care considerations to be aware of. The stage of the transplant (pre-transplant, early post-transplant, and chronic post-transplant) must be taken into account. Additionally, it's crucial to be knowledgeable about immunosuppressive medications, their typical adverse effects and drug interactions. The review article addresses a number of endocrine and metabolic abnormalities that are reported after transplantation.
Collapse
Affiliation(s)
- Narendra Kotwal
- Director & Commandant, Armed Forces Medical College, Pune, India
| | - Naresh Bansal
- Senior Adviser (Medicine) & Endocrinologist, Command Hospital (Southern Command), Pune, India
| | - J. Muthukrishnan
- Professor & Head, Department of Internal Medicine, Armed Forces Medical College, Pune, India
| | - Vishesh Verma
- Professor, Department of Internal Medicine, Armed Forces Medical College, Pune, India
| |
Collapse
|
7
|
Vinke JS, Eisenga MF, Sanders JSF, Berger SP, Spikman JM, Abdulahad WH, Bakker SJ, Gaillard CAJM, van Zuilen AD, van der Meer P, de Borst MH. Effect of Intravenous Ferric Carboxymaltose on Exercise Capacity After Kidney Transplantation (EFFECT-KTx): rationale and study protocol for a double-blind, randomised, placebo-controlled trial. BMJ Open 2023; 13:e065423. [PMID: 36948568 PMCID: PMC10040026 DOI: 10.1136/bmjopen-2022-065423] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 03/24/2023] Open
Abstract
INTRODUCTION Iron deficiency (ID) is common and has been associated with an excess mortality risk in kidney transplant recipients (KTRs). In patients with chronic heart failure and ID, intravenous iron improves exercise capacity and quality of life. Whether these beneficial effects also occur in KTRs is unknown. The main objective of this trial is to address whether intravenous iron improves exercise tolerance in iron-deficient KTRs. METHODS AND ANALYSIS The Effect of Ferric Carboxymaltose on Exercise Capacity after Kidney Transplantation study is a multicentre, double-blind, randomised, placebo-controlled clinical trial that will include 158 iron-deficient KTRs. ID is defined as plasma ferritin <100 µg/L or plasma ferritin 100-299 µg/L with transferrin saturation <20%. Patients are randomised to receive 10 mL of ferric carboxymaltose (50 mg Fe3+/mL, intravenously) or placebo (0.9% sodium chloride solution) every 6 weeks, four dosages in total. The primary endpoint is change in exercise capacity, as quantified by the 6 min walk test, between the first study visit and the end of follow-up, 24 weeks later. Secondary endpoints include changes in haemoglobin levels and iron status, quality of life, systolic and diastolic heart function, skeletal muscle strength, bone and mineral parameters, neurocognitive function and safety endpoints. Tertiary (explorative) outcomes are changes in gut microbiota and lymphocyte proliferation and function. ETHICS AND DISSEMINATION The protocol of this study has been approved by the medical ethical committee of the University Medical Centre Groningen (METc 2018/482;) and is being conducted in accordance with the principles of the Declaration of Helsinki, the Standard Protocol Items: Recommendations for Interventional Trials checklist and the Good Clinical Practice guidelines provided by the International Council for Harmonization of Technical Requirements for Pharmaceuticals for Human Use. Study results will be disseminated through publications in peer-reviewed journals and conference presentations. TRIAL REGISTRATION NUMBER NCT03769441.
Collapse
Affiliation(s)
- Joanna Sj Vinke
- Department of Nephrology, University Medical Centre Groningen, University of Groningen, Groningen, Netherlands
| | - Michele F Eisenga
- Department of Nephrology, University Medical Centre Groningen, University of Groningen, Groningen, Netherlands
| | - Jan-Stephan F Sanders
- Department of Nephrology, University Medical Centre Groningen, University of Groningen, Groningen, Netherlands
| | - Stefan P Berger
- Department of Nephrology, University Medical Centre Groningen, University of Groningen, Groningen, Netherlands
| | - Jacoba M Spikman
- Department of Neuropsychology, University Medical Centre Groningen, University of Groningen, Groningen, Netherlands
| | - Wayel H Abdulahad
- Department of Immunology, University Medical Centre Groningen, University of Groningen, Groningen, Netherlands
| | - Stephan Jl Bakker
- Department of Nephrology, University Medical Centre Groningen, University of Groningen, Groningen, Netherlands
| | - Carlo A J M Gaillard
- Department of Nephrology, University Medical Centre Utrecht, Utrecht, The Netherlands
| | - Arjan D van Zuilen
- Department of Nephrology, University Medical Centre Utrecht, Utrecht, The Netherlands
| | - P van der Meer
- Department of Cardiology, University Medical Centre Groningen, University of Groningen, Groningen, The Netherlands
| | - Martin H de Borst
- Department of Nephrology, University Medical Centre Groningen, University of Groningen, Groningen, Netherlands
| |
Collapse
|
8
|
Courbebaisse M, Bourmaud A, Souberbielle JC, Sberro-Soussan R, Moal V, Le Meur Y, Kamar N, Albano L, Thierry A, Dantal J, Danthu C, Moreau K, Morelon E, Heng AE, Bertrand D, Arzouk N, Perrin P, Morin MP, Rieu P, Presne C, Grimbert P, Ducloux D, Büchler M, Le Quintrec M, Ouali N, Pernin V, Bouvier N, Durrbach A, Alamartine E, Randoux C, Besson V, Hazzan M, Pages J, Colas S, Piketty ML, Friedlander G, Prié D, Alberti C, Thervet E. Nonskeletal and skeletal effects of high doses versus low doses of vitamin D 3 in renal transplant recipients: Results of the VITALE (VITamin D supplementation in renAL transplant recipients) study, a randomized clinical trial. Am J Transplant 2023; 23:366-376. [PMID: 36695682 DOI: 10.1016/j.ajt.2022.12.007] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2022] [Revised: 12/02/2022] [Accepted: 12/02/2022] [Indexed: 01/11/2023]
Abstract
Vitamin D sufficiency is associated with a reduced risk of fractures, diabetes mellitus, cardiovascular events, and cancers, which are frequent complications after renal transplantation. The VITALE (VITamin D supplementation in renAL transplant recipients) study is a multicenter double-blind randomized trial, including nondiabetic adult renal transplant recipients with serum 25-hydroxy vitamin D (25(OH) vitamin D) levels of <30 ng/mL, which is randomized 12 to 48 months after transplantation to receive high (100 000 IU) or low doses (12 000 IU) of cholecalciferol every 2 weeks for 2 months and then monthly for 22 months. The primary outcome was a composite endpoint, including diabetes mellitus, major cardiovascular events, cancer, and death. Of 536 inclusions (50.8 [13.7] years, 335 men), 269 and 267 inclusions were in the high-dose and low-dose groups, respectively. The serum 25(OH) vitamin D levels increased by 23 versus 6 ng/mL in the high-dose and low-dose groups, respectively (P < .0001). In the intent-to-treat analysis, 15% versus 16% of the patients in the high-dose and low-dose groups, respectively, experienced a first event of the composite endpoint (hazard ratio, 0.94 [0.60-1.48]; P = .78), whereas 1% and 4% of patients in the high-dose and low-dose groups, respectively, experienced an incident symptomatic fracture (odds ratio, 0.24 [0.07-0.86], P = .03). The incidence of adverse events was similar between the groups. After renal transplantation, high doses of cholecalciferol are safe but do not reduce extraskeletal complications (trial registration: ClinicalTrials.gov; identifier: NCT01431430).
Collapse
Affiliation(s)
- Marie Courbebaisse
- Paris University; Physiology Department, European Georges-Pompidou Hospital, APHP; INSERM U1151. Paris, France.
| | - Aurelie Bourmaud
- APHP.Nord-Université de Paris, Hôpital Universitaire Robert Debré, Unité d'Epidémiologie Clinique, Inserm, CIC 1426, F-75019 Paris, France; Université de Paris, ECEVE UMR 1123, INSERM. F-75010 Paris, France
| | - Jean-Claude Souberbielle
- Service des explorations fonctionnelles hôpital Necker-Enfants Malades, DMU Biophygen, GHU Centre Université de Paris APHP. Paris, France
| | - Rebecca Sberro-Soussan
- Université de Paris; Service de Transplantation Rénale et Néphrologie, Hôpital Necker Enfant Malades, APHP. Paris, France
| | - Valérie Moal
- Aix-Marseille Université - AP-HM - Hôpital Conception - Centre de Néphrologie et Transplantation Rénale. Marseille, France
| | - Yannick Le Meur
- Department of Nephrology, CHU de Brest; UMR1227, Lymphocytes B et Autoimmunité, Université de Brest, Inserm, Labex IGO. Brest, France
| | - Nassim Kamar
- Department of Nephrology, Dialysis and Organ Transplantation, CHU Rangueil, INSERM U1043, IFR -BMT, University Paul Sabatier. Toulouse, France
| | - Laetitia Albano
- Service de Transplantation Rénale, CHU de Nice. Nice, France
| | - Antoine Thierry
- Service de Néphrologie, INSERM U1082 et Fédération Hospitalo-Universitaire BIOSUPORT. Poitiers, France
| | - Jacques Dantal
- CRTI (Centre de Recherche en Transplantation et Immunologie) INSERM UMR1064, Université de Nantes, Centre Hospitalier Universitaire de Nantes. Nantes, France
| | - Clément Danthu
- Department of Nephrology, Limoges University hospital; UMR INSERM 1092, RESINFIT, Limoges University hospital 2. Limoges, France
| | - Karine Moreau
- Unité de transplantation rénale, Hôpital Pellegrin, CHU de Bordeaux. Bordeaux, France
| | - Emmanuel Morelon
- Service de transplantation, néphrologie et immunologie clinique, Hôpital Edouard Herriot, Hospices Civils de Lyon; INSERM U1111, Université Claude Bernard Lyon1. Lyon, France
| | - Anne-Elisabeth Heng
- Service de Néphrologie, Dialyse, Transplantation, CHU de Clermont-Ferrand; Université Clermont Auvergne. F-63000 Clermont-Ferrand, France
| | - Dominique Bertrand
- Nephrology, dialysis and kidney transplantation, Rouen University Hospital. Rouen, France
| | - Nadia Arzouk
- Service de Transplantation Rénale, hôpital La Pitié-Salpétrière, APHP. Paris, France
| | - Peggy Perrin
- Department of Nephrology and Transplantation, Strasbourg University Hospital; Fédération de Médecine Translationnelle, INSERM U1109, LabEx TRANSPLANTEX. Strasbourg, France
| | | | - Philippe Rieu
- Division of Nephrology, Reims university hospital, CRNS UMR 7369 MEDyC laboratory. Reims, France
| | - Claire Presne
- Nephrology Internal Medicine Dialysis Transplantation Department, Amiens University Hospital. Amiens France
| | - Philippe Grimbert
- Nephrology and Transplant Department, CHU Henri-Mondor, APHP; Université Paris Est Créteil, INSERM U955. Paris, France
| | - Didier Ducloux
- Department of Nephrology, CHU Besançon. Besançon, France
| | - Matthias Büchler
- Department of Nephrology and Transplantation, CHU Tours; University of Tours, EA4245 Transplantation, Immunology, Inflammation; FHU SUPORT. Tours, France
| | | | - Nacéra Ouali
- Nephrology department, SINRA, Hôpital Tenon. Paris, France
| | - Vincent Pernin
- University of Montpellier, Department of Nephrology, Dialysis and Transplantation, Montpellier University hospital; Institute for Regenerative Medicine & Biotherapy (IRMB), INSERM U1183. Montpellier, France
| | - Nicolas Bouvier
- Service de Néphrologie-Dialyse-Transplantation, CHU Caen Normandie; Université de Caen Normandie. Caen, France
| | - Antoine Durrbach
- Université Paris Saclay, France; INSERM UMR 1186, Gustave Roussy. Villejuif, France; Nephrology Department, Bicêtre Hospital APHP. Le Kremlin-Bicêtre, France
| | - Eric Alamartine
- CHU de Saint Etienne et CIRI - INSERM U1111 - CNRS UMR5308 - ENS Lyon/UCBL1/Université St Etienne. Saint Etienne, France
| | - Christine Randoux
- Service de Néphrologie, CHU Bichat Claude Bernard, APHP.Nord. Paris, France
| | - Virginie Besson
- Service de Néphrologie-Dialyse-transplantation, CHU d'Angers. Angers, France
| | - Marc Hazzan
- Université de Lille, INSERM, CHU Lille, U1286 - Infinite - Institute for Translational Research in Inflammation. F-59000 Lille, France
| | - Justine Pages
- APHP.Nord-Université de Paris, Hôpital Universitaire Robert Debré, Unité d'Epidémiologie Clinique, INSERM, CIC 1426. F-75019 Paris, France
| | - Sandra Colas
- Unité de Recherche Clinique Necker-Cochin, APHP. Paris, France
| | - Marie-Liesse Piketty
- Service des explorations fonctionnelles hôpital Necker-Enfants Malades, DMU Biophygen, GHU Centre Université de Paris APHP. Paris, France
| | | | - Dominique Prié
- Université de Paris; INSERM U1151, service des explorations fonctionnelles hôpital Necker-Enfants Malades, DMU Biophygen, GHU Centre Université de Paris APHP. Paris, France
| | - Corinne Alberti
- APHP.Nord-Université de Paris, Hôpital Universitaire Robert Debré, Unité d'Epidémiologie Clinique, Inserm, CIC 1426, F-75019 Paris, France; Université de Paris, ECEVE UMR 1123, INSERM. F-75010 Paris, France
| | - Eric Thervet
- Paris University; Nephrology Department, European Georges-Pompidou Hospital, APHP; INSERM UMR 970, Paris Cardiovascular Research Center. Paris, France
| |
Collapse
|
9
|
Kubota M, Hamasaki Y, Hashimoto J, Aoki Y, Kawamura T, Saito A, Yuasa R, Muramatsu M, Komaba H, Toyoda M, Fukagawa M, Shishido S, Sakai K. Fibroblast growth factor 23-Klotho and mineral metabolism in the first year after pediatric kidney transplantation: A single-center prospective study. Pediatr Transplant 2023; 27:e14440. [PMID: 36471536 DOI: 10.1111/petr.14440] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/27/2022] [Revised: 10/27/2022] [Accepted: 10/28/2022] [Indexed: 12/12/2022]
Abstract
BACKGROUND The role of fibroblast growth factor 23 (FGF23) levels in mineral metabolism before and after kidney transplantation in pediatric patients is poorly understood. METHODS We prospectively evaluated 24 patients under 18 years of age (4.5 [3.3-9.8] years) who underwent living kidney transplantation between July 2016 and March 2018, and measured intact FGF23 and serum αKlotho levels, and other parameters of mineral metabolism before and after transplantation (Day 7, 1 and 4 months, and 1 year). Relationships between parameters were examined by linear analysis. RESULTS FGF23 level was 440.8 [63.4-5916.3] pg/ml pre-transplant and decreased significantly to 37.1 [16.0-71.5] pg/ml at Day 7 post-transplant (-91.6%, p < .001). Thereafter, it remained at normal levels until 1 year. αKlotho level was 785 [568-1292] pg/ml pre-transplant and remained low at Day 7 and 1 month post-transplant, with an increasing trend at 4 months. Post-transplant phosphorus levels were significantly decreased compared with pre-transplant, with a lowest level of 1.7 [1.3-2.9] mg/dl, -5.7 [-6.8, -3.8] SD at Day 4, followed by gradual recovery. Phosphorus levels and the ratio of tubular maximum phosphate reabsorption were significantly and negatively associated with pre-transplant FGF23 until 4 months of post-transplant. Pre-transplant αKlotho was negatively associated with pre-transplant FGF23 but not FGF23 or other parameters after transplantation. CONCLUSION FGF23 in pediatric kidney transplant patients decreased rapidly after transplantation and associated with post-transplant hypophosphatemia and increased phosphorus excretion. Post-transplant αKlotho was low early post-transplant but tended to increase subsequently. Post-transplant αKlotho was unaffected by pre-transplant FGF23 or other factors, suggesting pre-transplant chronic kidney disease status has no effect.
Collapse
Affiliation(s)
- Mai Kubota
- Department of Nephrology, Toho University, Faculty of Medicine, Tokyo, Japan
| | - Yuko Hamasaki
- Department of Nephrology, Toho University, Faculty of Medicine, Tokyo, Japan
| | - Junya Hashimoto
- Department of Nephrology, Toho University, Faculty of Medicine, Tokyo, Japan
| | - Yujiro Aoki
- Department of Nephrology, Toho University, Faculty of Medicine, Tokyo, Japan
| | - Takeshi Kawamura
- Department of Nephrology, Toho University, Faculty of Medicine, Tokyo, Japan
| | - Akinobu Saito
- Department of Nephrology, Toho University, Faculty of Medicine, Tokyo, Japan
| | - Rena Yuasa
- Department of Nephrology, Toho University, Faculty of Medicine, Tokyo, Japan
| | - Masaki Muramatsu
- Department of Nephrology, Toho University, Faculty of Medicine, Tokyo, Japan
| | - Hirotaka Komaba
- Division of Nephrology, Endocrinology and Metabolism, Tokai University School of Medicine, Isehara, Japan
| | - Masao Toyoda
- Division of Nephrology, Endocrinology and Metabolism, Tokai University School of Medicine, Isehara, Japan
| | - Masafumi Fukagawa
- Division of Nephrology, Endocrinology and Metabolism, Tokai University School of Medicine, Isehara, Japan
| | - Seichiro Shishido
- Department of Pediatric Nephrology, Toho University, Faculty of Medicine, Tokyo, Japan
| | - Ken Sakai
- Department of Nephrology, Toho University, Faculty of Medicine, Tokyo, Japan
| |
Collapse
|
10
|
Bosman A, Palermo A, Vanderhulst J, De Beur SMJ, Fukumoto S, Minisola S, Xia W, Body JJ, Zillikens MC. Tumor-Induced Osteomalacia: A Systematic Clinical Review of 895 Cases. Calcif Tissue Int 2022; 111:367-379. [PMID: 35857061 PMCID: PMC9474374 DOI: 10.1007/s00223-022-01005-8] [Citation(s) in RCA: 29] [Impact Index Per Article: 14.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/05/2022] [Accepted: 07/01/2022] [Indexed: 11/24/2022]
Abstract
Tumor-induced osteomalacia (TIO) is a rare and largely underdiagnosed paraneoplastic condition. Previous reviews often reported incomplete data on clinical aspects, diagnosis or prognosis. The aim of this study was to present a systematic clinical review of all published cases of TIO. A search was conducted in Pubmed, Embase, Web of Science from inception until April 23rd, 2020. We selected case reports and case series of patients diagnosed with TIO, with information on tumor localization and serum phosphate concentration. Two reviewers independently extracted data on biochemical and clinical characteristics including bone involvement, tumor localization and treatment. 468 articles with 895 unique TIO cases were included. Median age was 46 years (range 9 months-90 years) and 58.3% were males. Hypophosphatemia and inappropriately low or normal 1,25-dihydroxyvitamin D levels, characteristic for TIO, were present in 98% of cases. Median tumor size was 2.7 cm (range 0.5 to 25.0 cm). Serum fibroblast growth factor 23 was related to tumor size (r = 0.344, P < 0.001). In 32% of the cases the tumor was detected by physical examination. Data on bone phenotype confirmed skeletal involvement: 62% of cases with BMD data had a T-score of the lumbar spine ≤ - 2.5 (n = 61/99) and a fracture was reported in at least 39% of all cases (n = 346/895). Diagnostic delay was longer than 2 years in more than 80% of cases. 10% were reported to be malignant at histology. In conclusion, TIO is a debilitating disease characterized by a long diagnostic delay leading to metabolic disturbances and skeletal impairment. Increasing awareness of TIO should decrease its diagnostic delay and the clinical consequences.
Collapse
Affiliation(s)
- Ariadne Bosman
- Department of Internal Medicine, Erasmus MC, University Medical Center Rotterdam, Rotterdam, The Netherlands
| | - Andrea Palermo
- Unit of Metabolic Bone and Thyroid Disorders, Fondazione Policlinico Universitario Campus Bio-Medico, Rome, Italy
| | - Julien Vanderhulst
- Department of Medicine, CHU Brugmann, Université Libre de Bruxelles (ULB), Brussels, Belgium
| | | | - Seiji Fukumoto
- Fujii Memorial Institute of Medical Sciences, Institute of Advanced Medical Sciences, Tokushima University, Tokushima, Japan
| | - Salvatore Minisola
- Department of Clinical, Internal, Anesthesiological and Cardiological Sciences, "Sapienza" Rome University, 00161, Rome, Italy
| | - Weibo Xia
- Department of Endocrinology, Key Laboratory of Endocrinology, The National Commission of Health, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences, Beijing, China
| | - Jean-Jacques Body
- Department of Medicine, CHU Brugmann, Université Libre de Bruxelles (ULB), Brussels, Belgium
| | - M Carola Zillikens
- Department of Internal Medicine, Erasmus MC, University Medical Center Rotterdam, Rotterdam, The Netherlands.
| |
Collapse
|
11
|
Halim A, Burney HN, Li X, Li Y, Tomkins C, Siedlecki AM, Lu TS, Kalim S, Thadhani R, Moe S, Ting SM, Zehnder D, Hiemstra TF, Lim K. FGF23 and Cardiovascular Structure and Function in Advanced Chronic Kidney Disease. KIDNEY360 2022; 3:1529-1541. [PMID: 36245643 PMCID: PMC9528374 DOI: 10.34067/kid.0002192022] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/23/2022] [Accepted: 06/29/2022] [Indexed: 11/27/2022]
Abstract
Background Fibroblast growth factor 23 (FGF23) is a bone-derived phosphatonin that is elevated in chronic kidney disease (CKD) and has been implicated in the development of cardiovascular disease. It is unknown whether elevated FGF23 in CKD is associated with impaired cardiovascular functional capacity, as assessed by maximum exercise oxygen consumption (VO2Max). We sought to determine whether FGF23 is associated with cardiovascular functional capacity in patients with advanced CKD and after improvement of VO2Max by kidney transplantation. Methods We performed secondary analysis of 235 patients from the Cardiopulmonary Exercise Testing in Renal Failure and After Kidney Transplantation (CAPER) cohort, which recruited patients with stage 5 CKD who underwent kidney transplantation or were waitlisted and hypertensive controls. All patients underwent cardiopulmonary exercise testing (CPET) and echocardiography and were followed longitudinally for 1 year after study enrollment. Results Patients across FGF23 quartiles differed in BMI (P=0.004) and mean arterial pressure (P<0.001) but did not significantly differ in sex (P=0.5) or age (P=0.08) compared with patients with lower levels of FGF23. Patients with higher FGF23 levels had impaired VO2Max (Q1: 24.2±4.8 ml/min per kilogram; Q4: 18.6±5.2 ml/min per kilogram; P<0.001), greater left ventricular mass index (LVMI; P<0.001), reduced HR at peak exercise (P<0.001), and maximal workload (P<0.001). Kidney transplantation conferred a significant decline in FGF23 at 2 months (P<0.001) before improvement in VO2Max at 1 year (P=0.008). Multivariable regression modeling revealed that changes in FGF23 was significantly associated with VO2Max in advanced CKD (P<0.001) and after improvement after kidney transplantation (P=0.006). FGF23 was associated with LVMI before kidney transplantation (P=0.003), however this association was lost after adjustment for dialysis status (P=0.4). FGF23 was not associated with LVMI after kidney transplantation in all models. Conclusions FGF23 levels are associated with alterations in cardiovascular functional capacity in advanced CKD and after kidney transplantation. FGF23 is only associated with structural cardiac adaptations in advanced CKD but this was modified by dialysis status, and was not associated after kidney transplantation.
Collapse
Affiliation(s)
- Arvin Halim
- Division of Nephrology and Hypertension, Department of Medicine, Indiana University School of Medicine, Indianapolis, Indiana
| | - Heather N. Burney
- Department of Biostatistics and Health Data Science, Indiana University School of Medicine, Indianapolis, Indiana
| | - Xiaochun Li
- Department of Biostatistics and Health Data Science, Indiana University School of Medicine, Indianapolis, Indiana
| | - Yang Li
- Department of Biostatistics and Health Data Science, Indiana University School of Medicine, Indianapolis, Indiana
| | - Claudia Tomkins
- Biochemistry Department, Kettering General Hospital NHS Foundation Trust, Kettering, United Kingdom
| | - Andrew M. Siedlecki
- Division of Renal Medicine, Brigham and Women’s Hospital, Harvard Medical School, Boston, Massachusetts
| | - Tzong-shi Lu
- Division of Renal Medicine, Brigham and Women’s Hospital, Harvard Medical School, Boston, Massachusetts
| | - Sahir Kalim
- Nephrology Division, Massachusetts General Hospital, Boston, Massachusetts
| | - Ravi Thadhani
- Mass General Brigham, Harvard Medical School, Massachusetts
| | - Sharon Moe
- Division of Nephrology and Hypertension, Department of Medicine, Indiana University School of Medicine, Indianapolis, Indiana
| | - Stephen M.S. Ting
- Department of Medicine, University Hospitals Birmingham NHS Foundation Trust, Birmingham, United Kingdom
| | - Daniel Zehnder
- Department of Nephrology and Department of Acute Medicine, North Cumbria University Hospital NHS Trust, Carlisle, United Kingdom
| | - Thomas F. Hiemstra
- School of Clinical Medicine, University of Cambridge; Clinical Trials Unit (CTU), Addenbrooke’s Hospital, Cambridge, United Kingdom
| | - Kenneth Lim
- Division of Nephrology and Hypertension, Department of Medicine, Indiana University School of Medicine, Indianapolis, Indiana
| |
Collapse
|
12
|
Hsieh MC, Hsiao PJ, Liao MT, Hou YC, Chang YC, Chiang WF, Wu KL, Chan JS, Lu KC. The Role of Vitamin D in SARS-CoV-2 Infection and Acute Kidney Injury. Int J Mol Sci 2022; 23:7368. [PMID: 35806377 PMCID: PMC9266309 DOI: 10.3390/ijms23137368] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2022] [Revised: 06/30/2022] [Accepted: 06/30/2022] [Indexed: 02/04/2023] Open
Abstract
Vitamin D has been described as an essential nutrient and hormone, which can cause nuclear, non-genomic, and mitochondrial effects. Vitamin D not only controls the transcription of thousands of genes, directly or indirectly through the modulation of calcium fluxes, but it also influences the cell metabolism and maintenance specific nuclear programs. Given its broad spectrum of activity and multiple molecular targets, a deficiency of vitamin D can be involved in many pathologies. Vitamin D deficiency also influences mortality and multiple outcomes in chronic kidney disease (CKD). Active and native vitamin D serum levels are also decreased in critically ill patients and are associated with acute kidney injury (AKI) and in-hospital mortality. In addition to regulating calcium and phosphate homeostasis, vitamin D-related mechanisms regulate adaptive and innate immunity. Severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) infections have a role in excessive proinflammatory cell recruitment and cytokine release, which contribute to alveolar and full-body endothelial damage. AKI is one of the most common extrapulmonary manifestations of severe coronavirus disease 2019 (COVID-19). There are also some correlations between the vitamin D level and COVID-19 severity via several pathways. Proper vitamin D supplementation may be an attractive therapeutic strategy for AKI and has the benefits of low cost and low risk of toxicity and side effects.
Collapse
Affiliation(s)
- Ming-Chun Hsieh
- Department of Internal Medicine, Taoyuan Armed Forces General Hospital, Taoyuan 235, Taiwan;
| | - Po-Jen Hsiao
- Division of Nephrology, Department of Internal Medicine, Taoyuan Armed Forces General Hospital, Taoyuan 235, Taiwan; (Y.-C.C.); (W.-F.C.); (K.-L.W.); (J.-S.C.)
- Division of Nephrology, Department of Internal Medicine, Tri-Service General Hospital, National Defense Medical Center, Taipei 114, Taiwan
- Department of Life Sciences, National Central University, Taoyuan 320, Taiwan
- School of Medicine, Fu-Jen Catholic University, New Taipei City 242, Taiwan;
- Institute of Molecular and Cellular Biology, National Tsing Hua University, Hsinchu 300, Taiwan
| | - Min-Tser Liao
- School of Medicine, Fu-Jen Catholic University, New Taipei City 242, Taiwan;
- Department of Pediatrics, Taoyuan Armed Forces General Hospital, Taoyuan 325, Taiwan
- Department of Pediatrics, Tri-Service General Hospital, National Defense Medical Center, Taipei 114, Taiwan
| | - Yi-Chou Hou
- Department of Medicine, Cardinal Tien Hospital, School of Medicine, Fu Jen Catholic University, New Taipei City 242, Taiwan;
| | - Ya-Chieh Chang
- Division of Nephrology, Department of Internal Medicine, Taoyuan Armed Forces General Hospital, Taoyuan 235, Taiwan; (Y.-C.C.); (W.-F.C.); (K.-L.W.); (J.-S.C.)
- Division of Nephrology, Department of Internal Medicine, Tri-Service General Hospital, National Defense Medical Center, Taipei 114, Taiwan
| | - Wen-Fang Chiang
- Division of Nephrology, Department of Internal Medicine, Taoyuan Armed Forces General Hospital, Taoyuan 235, Taiwan; (Y.-C.C.); (W.-F.C.); (K.-L.W.); (J.-S.C.)
- Division of Nephrology, Department of Internal Medicine, Tri-Service General Hospital, National Defense Medical Center, Taipei 114, Taiwan
| | - Kun-Lin Wu
- Division of Nephrology, Department of Internal Medicine, Taoyuan Armed Forces General Hospital, Taoyuan 235, Taiwan; (Y.-C.C.); (W.-F.C.); (K.-L.W.); (J.-S.C.)
- Division of Nephrology, Department of Internal Medicine, Tri-Service General Hospital, National Defense Medical Center, Taipei 114, Taiwan
| | - Jenq-Shyong Chan
- Division of Nephrology, Department of Internal Medicine, Taoyuan Armed Forces General Hospital, Taoyuan 235, Taiwan; (Y.-C.C.); (W.-F.C.); (K.-L.W.); (J.-S.C.)
- Division of Nephrology, Department of Internal Medicine, Tri-Service General Hospital, National Defense Medical Center, Taipei 114, Taiwan
| | - Kuo-Cheng Lu
- Division of Nephrology, Department of Internal Medicine, Tri-Service General Hospital, National Defense Medical Center, Taipei 114, Taiwan
- Division of Nephrology, Department of Medicine, Taipei Tzu Chi Hospital, Buddhist Tzu Chi Medical Foundation, New Taipei City 231, Taiwan
- Division of Nephrology, Department of Medicine, Fu-Jen Catholic University Hospital, School of Medicine, Fu-Jen Catholic University, New Taipei City 242, Taiwan
| |
Collapse
|
13
|
Kamel MH, Ahmed DH, Mikhael ES, Abdalla MS, Sadek KM, ElNahid MS. Serum Phosphorus, Parathyroid Hormone, and Serum Fibroblast Growth Factor-23 in Egyptian Patients Six Months after Undergoing Living-donor Kidney Transplantation. SAUDI JOURNAL OF KIDNEY DISEASES AND TRANSPLANTATION 2022; 33:353-360. [PMID: 37843136 DOI: 10.4103/1319-2442.385958] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/17/2023] Open
Abstract
End-stage renal disease is a major health problem with many complications. Previous studies emphasized the relationship of cardiovascular disease and mortality among these patients to dysregulated phosphate homeostasis. Even after successful renal transplantation, the risk is not eliminated. Several factors seem to interplay to regulate serum phosphorus levels after renal transplantation. Fibroblast growth factor-23 (FGF-23) is a hormone with the major function of inhibiting the reabsorption of phosphate by the renal tubules. Parathormone reduces the reabsorption of phosphate from the proximal tubule of the kidney. The aim of our study was to explore the changes that occurred in FGF-23 and intact parathyroid hormone (iPTH) levels in a cohort of Egyptian patients undergoing renal transplantation and to examine the effect of these factors on posttransplant serum phosphorus levels. The study was carried out prospectively on 37 candidates for live-donor renal transplantation. Serum levels of calcium, phosphorus, iPTH, and FGF-23 were measured before and 6 months after renal transplantation. Statistically significant differences were detected in serum calcium, phosphorus, FGF-23, and iPTH before and 6 months after transplantation (P < 0.001, P < 0.001, P < 0.001, and P < 0.001, respectively). The results also showed a statistically significant correlation between FGF-23 levels and phosphorus levels before transplantation. The interplay between FGF-23 and iPTH has an impact on posttransplant serum phosphorus levels.
Collapse
Affiliation(s)
- Mai Hamed Kamel
- Department of Clinical and Chemical Pathology, Faculty of Medicine, Cairo University, Cairo, Egypt
| | - Dina Hesham Ahmed
- Department of Clinical and Chemical Pathology, Faculty of Medicine, Cairo University, Cairo, Egypt
| | - Emily Samir Mikhael
- Department of Clinical and Chemical Pathology, Faculty of Medicine, Cairo University, Cairo, Egypt
| | - Mohamed Shehata Abdalla
- Department of Clinical and Chemical Pathology, Faculty of Medicine, Cairo University, Cairo, Egypt
| | - Khaled Marzouk Sadek
- Department of Internal Medicine and Nephrology, Faculty of Medicine, Cairo University, Cairo, Egypt
| | - Maggie Said ElNahid
- Department of Internal Medicine and Nephrology, Faculty of Medicine, Cairo University, Cairo, Egypt
| |
Collapse
|
14
|
Ammar YA, Maharem DA, Mohamed AH, Khalil GI, Shams-Eldin RS, Dwedar FI. Fibroblast growth factor-23 rs7955866 polymorphism and risk of chronic kidney disease. EGYPTIAN JOURNAL OF MEDICAL HUMAN GENETICS 2022. [DOI: 10.1186/s43042-022-00289-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/10/2022] Open
Abstract
Abstract
Background
A missense gain-of-function fibroblast growth factor-23 (FGF23) gene single nucleotide polymorphism (SNP) (rs7955866) has been associated with FGF23 hypersecretion, phosphaturia, and bone disease. Excess circulating FGF23 was linked with atherosclerosis, hypertension, initiation, and progression of chronic kidney disease (CKD).
Methods
The study included 72 CKD stage 2/3 Egyptian patients (27–71 years old, 37 females) and 26 healthy controls matching in age and sex. Repeated measures of blood pressure were used to quantify hypertension on a semiquantitative scale (grades 0 to 5). Fasting serum urea, creatinine, uric acid, total proteins, albumin, calcium, phosphorus, vitamin D3, intact parathyroid hormone (iPTH), and intact FGF23 (iFGF23) were measured. DNA extracted from peripheral blood leucocytes was used for genotyping of FGF23 rs7955866 SNP using the TaqMan SNP genotyping allelic discrimination method.
Results
Major causes of CKD were hypertension, diabetic kidney disease, and CKD of unknown etiology. There was no significant difference in minor allele (A) frequency between the studied groups (0.333 in GI and 0.308 in GII). Median (IQR) serum iFGF23 was significantly higher in GI [729.2 (531.9–972.3)] than in GII [126.1 (88.5–152.4)] pg/mL, P < 0.001. Within GI, the minor allele (A) frequency load, coded for codominant inheritance, had a significant positive correlation with both hypertension grade (r = 0.385, P = 0.001) and serum iFGF23 (r = 0.259, P = 0.028). Hypertension grade had a significant positive correlation with serum phosphorus and iFGF23.
Conclusions
For the first time in an Egyptian cohort, we report a relatively high frequency of the rs7955866 SNP. It may remain dormant or become upregulated in response to some environmental triggers, notably dietary phosphorus excess, leading to increased circulating iFGF23 with ensuing hypertension and/or renal impairment. Subjects with this SNP, particularly in the homozygous form, are at increased risk for CKD of presumably “unknown” etiology, with a tendency for early onset hypertension and increased circulating iFGF23 out of proportion with the degree of renal impairment. Large-scale population studies are needed to confirm these findings and explore the role of blockers of the renin–angiotensin–aldosterone system and sodium chloride cotransporters in mitigating hypertension associated with FGF23 excess.
Collapse
|
15
|
Abstract
After kidney transplantation, mineral and bone disorders are associated with higher risk of fractures and consequent morbidity and mortality. Disorders of calcium and phosphorus, vitamin D deficiency, and hyperparathyroidism are also common. The epidemiology of bone disease has evolved over the past several decades due to changes in immunosuppressive regimens, mainly glucocorticoid minimization or avoidance. The assessment of bone disease in kidney transplant recipients relies on risk factor recognition and bone mineral density assessment. Several drugs have been trialed for the treatment of post-transplant mineral and bone disorders. This review will focus on the epidemiology, effect, and treatment of metabolic and skeletal derangements in the transplant recipient.
Collapse
Affiliation(s)
- Pascale Khairallah
- Section of Nephrology and Selzman Institute for Kidney Health, Baylor College of Medicine, Houston, Texas
| | - Thomas L. Nickolas
- Division of Nephrology, Columbia University Irving Medical Center, New York, New York
| |
Collapse
|
16
|
Ralston MR, Stevenson KS, Mark PB, Geddes CC. Clinical factors associated with severe hypophosphataemia after kidney transplant. BMC Nephrol 2021; 22:407. [PMID: 34886802 PMCID: PMC8656060 DOI: 10.1186/s12882-021-02624-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2021] [Accepted: 11/24/2021] [Indexed: 11/10/2022] Open
Abstract
BACKGROUND The mechanism by which hypophosphataemia develops following kidney transplantation remains debated, and limited research is available regarding risk factors. This study aimed to assess the association between recipient and donor variables, and the severity of post-transplantation hypophosphataemia. METHODS We performed a single-centre retrospective observational study. We assessed the association between demographic, clinical and biochemical variables and the development of hypophosphataemia. We used linear regression analysis to assess association between these variables and phosphate nadir. RESULTS 87.6% of patients developed hypophosphataemia. Patients developing hypophosphataemia were younger, had a shorter time on renal replacement therapy, were less likely to have had a parathyroidectomy or to experience delayed graft function, were more likely to have received a living donor transplant, from a younger donor. They had higher pre-transplantation calcium levels, and lower alkaline phosphatase levels. Receipt of a living donor transplant, lower donor age, not having had a parathyroidectomy, receiving a transplant during the era of tacrolimus-based immunosuppression, not having delayed graft function, higher pre-transplantation calcium, and higher pre-transplantation phosphate were associated with lower phosphate nadir by multiple linear regression. CONCLUSIONS This analysis demonstrates an association between variables relating to better graft function and hypophosphataemia. The links with biochemical measures of mineral-bone disease remain less clear.
Collapse
Affiliation(s)
- Maximilian R Ralston
- Glasgow Renal & Transplant Unit, Queen Elizabeth University Hospital, 1345 Govan Road, Glasgow, G51 4TF, UK.
| | - Karen S Stevenson
- Glasgow Renal & Transplant Unit, Queen Elizabeth University Hospital, 1345 Govan Road, Glasgow, G51 4TF, UK
| | - Patrick B Mark
- Glasgow Renal & Transplant Unit, Queen Elizabeth University Hospital, 1345 Govan Road, Glasgow, G51 4TF, UK.,Institute of Cardiovascular & Medical Sciences, University of Glasgow, Glasgow, UK
| | - Colin C Geddes
- Glasgow Renal & Transplant Unit, Queen Elizabeth University Hospital, 1345 Govan Road, Glasgow, G51 4TF, UK
| |
Collapse
|
17
|
Xi Y, Ma Y, Xie B, Di A, Xu S, Luo X, Wang C, Dai H, Yan G, Qi Z. Vitamin D3 combined with antibody agents suppresses alloreactive memory T-cell responses to induce heart allograft long-term survival. Transpl Immunol 2021; 66:101374. [PMID: 33592299 DOI: 10.1016/j.trim.2021.101374] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2021] [Revised: 02/11/2021] [Accepted: 02/11/2021] [Indexed: 10/22/2022]
Abstract
BACKGROUND The pre-stored memory T cells in organ transplant patient carry a high risk of allograft rejection. The current study aimed to determine whether the allogenic response of adoptively transferred memory T cells in mice was suppressed by vitamin D3 monotherapy alone or in combination with monoclonal antibody treatment. METHODS Prior to vascularized heterotopic heart transplantation, naïve C57BL/6 mice were primed with memory T cells. Recipient mice were administered vitamin D3 alone or in combination with monoclonal antibodies (anti-CD40L/ anti-LFA-1). Memory T cells and CD4+ forkhead box P3+ T cells in recipient spleens were measured using flow cytometry. Additionally, the expression of cytokines was measured by ELISA and quantitative PCR. Inflammatory factors in the grafts were identified by hematoxylin and eosin staining. RESULTS Vitamin D3 in conjunction with anti-CD40L/ anti-LFA-1 antibodies were administered according to the median survival time from 6.5 to 80 days. The results revealed that grafts were protected through the prevention of inflammatory cell infiltration. Combined treatment decreased the mRNA levels of IL-2, IFN-γ and IL-10 and increased the mRNA levels of IL-4, Foxp3 and TGF-β in the allograft. Rejection was suppressed by a reduction of CD4+CD44high CD62L+ and CD8+ CD44high CD62L+ memory T cells, the induction of regulatory T cells in the recipient spleen and a reduction of serum IL-2, IFN-γ and IL-10 levels. CONCLUSION Vitamin D3 efficiently protected allografts from memory T-cell allo-responses when combined with anti-CD40L/anti-LFA-1 antibodies therapy.
Collapse
Affiliation(s)
- Yanfeng Xi
- Fujian Provincial Key Laboratory of Organ and Tissue Regeneration, School of Medicine, Xiamen University, Xiamen, Fujian, China; The tumor hospital of Chang Zhou, Chang Zhou, Jiangsu, China
| | - Yunhan Ma
- Fujian Provincial Key Laboratory of Organ and Tissue Regeneration, School of Medicine, Xiamen University, Xiamen, Fujian, China
| | - Baiyi Xie
- Department of Urology Surgery, Ruikang Hospital affiliated to Guangxi University of Chinese Medicine, Nanning, Guangxi, China
| | - Anjie Di
- Basic Medical Department of Medical College, School of Medicine, Xiamen University, Xiamen, Fujian, China
| | - Shuangyue Xu
- Fujian Provincial Key Laboratory of Organ and Tissue Regeneration, School of Medicine, Xiamen University, Xiamen, Fujian, China
| | - Xuewei Luo
- Medicinal College, Guangxi University, Nanning, Guangxi, China
| | - Chenxi Wang
- Basic Medical Department of Medical College, School of Medicine, Xiamen University, Xiamen, Fujian, China
| | - Helong Dai
- Department of Kidney Transplantation, The Second Xiangya Hospital of Central South University, Changsha, Hunan, China; Clinical Research Center for Organ Transplantation in Hunan Province, Changsha, Hunan, China; Clinical Immunology Center, Central South University, Changsha, Hunan 410000, China.
| | - Guoliang Yan
- Fujian Provincial Key Laboratory of Organ and Tissue Regeneration, School of Medicine, Xiamen University, Xiamen, Fujian, China; Basic Medical Department of Medical College, School of Medicine, Xiamen University, Xiamen, Fujian, China.
| | - Zhongquan Qi
- Fujian Provincial Key Laboratory of Organ and Tissue Regeneration, School of Medicine, Xiamen University, Xiamen, Fujian, China; Medicinal College, Guangxi University, Nanning, Guangxi, China.
| |
Collapse
|
18
|
Isakov O, Ghinea R, Beckerman P, Mor E, Riella LV, Hod T. Early persistent hyperparathyroidism post-renal transplantation as a predictor of worse graft function and mortality after transplantation. Clin Transplant 2020; 34:e14085. [PMID: 32949044 DOI: 10.1111/ctr.14085] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2020] [Revised: 08/19/2020] [Accepted: 08/27/2020] [Indexed: 12/28/2022]
Abstract
BACKGROUND Persistent hyperparathyroidism (pHPT) is frequently seen after transplantation contributing to post-transplant complications. METHODS We conducted a retrospective single center analysis to explore the relationship of early pHPT and long-term allograft outcome. Patients were divided into high (N = 153) and low (N = 252) PTH groups based on serum parathyroid hormone (PTH) level 3 months post-transplant (PTH ≥ 150 and < 150 pg/mL, respectively). RESULTS High PTH was found to be an independent predictor for reduced kidney allograft function up to 3 years post-transplant. eGFR decreased by 11.4 mL/min (P < .001) and the odds of having an eGFR < 60 mL/min 3 years post-transplant were sixfold higher (P < .01) in the high compared to the low PTH group. Subgroup analysis based on eGFR 1 year post-transplant, presence of slow graft function (SGF), and transplant type revealed similar results. High PTH three months post-transplant was also independently associated with an increased risk for overall mortality and for death with a functioning graft (P < .05). CONCLUSIONS pHPT three months post-renal transplantation is an independent predictor for a worse allograft function up to 3 years post-transplant and a risk factor for mortality. This relationship remains statistically significant after accounting for baseline allograft function, presence of SGF and serum mineral levels abnormalities.
Collapse
Affiliation(s)
- Ofer Isakov
- Department of Internal Medicine "T", Tel Aviv Souraski Medical Center, Tel Aviv University, Tel Aviv, Israel
| | - Ronen Ghinea
- Department of Surgery, Sheba Medical Center, Tel Aviv University, Tel Aviv, Israel.,Transplant Nephrology Center, Sheba Medical Center, Tel Aviv University, Tel Aviv, Israel
| | - Pazit Beckerman
- Department of Nephrology, Sheba Medical Center, Tel Aviv University, Tel Aviv, Israel
| | - Eytan Mor
- Transplant Nephrology Center, Sheba Medical Center, Tel Aviv University, Tel Aviv, Israel
| | - Leonardo V Riella
- Renal Division, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
| | - Tammy Hod
- Transplant Nephrology Center, Sheba Medical Center, Tel Aviv University, Tel Aviv, Israel.,Department of Nephrology, Sheba Medical Center, Tel Aviv University, Tel Aviv, Israel
| |
Collapse
|
19
|
Vinke JSJ, Francke MI, Eisenga MF, Hesselink DA, de Borst MH. Iron deficiency after kidney transplantation. Nephrol Dial Transplant 2020; 36:1976-1985. [PMID: 32910168 PMCID: PMC8577626 DOI: 10.1093/ndt/gfaa123] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2019] [Indexed: 12/30/2022] Open
Abstract
Iron deficiency (ID) is highly prevalent in kidney transplant recipients (KTRs) and has been independently associated with an excess mortality risk in this population. Several causes lead to ID in KTRs, including inflammation, medication and an increased iron need after transplantation. Although many studies in other populations indicate a pivotal role for iron as a regulator of the immune system, little is known about the impact of ID on the immune system in KTRs. Moreover, clinical trials in patients with chronic kidney disease or heart failure have shown that correction of ID, with or without anaemia, improves exercise capacity and quality of life, and may improve survival. ID could therefore be a modifiable risk factor to improve graft and patient outcomes in KTRs; prospective studies are warranted to substantiate this hypothesis.
Collapse
Affiliation(s)
- Joanna Sophia J Vinke
- Department of Medicine, Division of Nephrology, University of Groningen, University Medical Center Groningen, Groningen, The Netherlands
| | - Marith I Francke
- Department of Internal Medicine, Division of Nephrology and Transplantation, Erasmus MC, University Medical Center Rotterdam, Rotterdam, The Netherlands
| | - Michele F Eisenga
- Department of Medicine, Division of Nephrology, University of Groningen, University Medical Center Groningen, Groningen, The Netherlands
| | - Dennis A Hesselink
- Department of Internal Medicine, Division of Nephrology and Transplantation, Erasmus MC, University Medical Center Rotterdam, Rotterdam, The Netherlands
| | - Martin H de Borst
- Department of Medicine, Division of Nephrology, University of Groningen, University Medical Center Groningen, Groningen, The Netherlands
| |
Collapse
|
20
|
Ghorbani M, Ossareh S. Early postkidney transplantation hypophosphatemia. JOURNAL OF RESEARCH IN MEDICAL SCIENCES 2020; 25:36. [PMID: 32582342 PMCID: PMC7306234 DOI: 10.4103/jrms.jrms_452_19] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Received: 08/06/2019] [Revised: 10/11/2019] [Accepted: 12/25/2019] [Indexed: 11/17/2022]
Abstract
Background: As hypophosphatemia is a common multifactorial problem of kidney transplantation (Tx), this research aimed at studying the frequency of posttransparent hypophosphatemia in the early postkidney Tx period and investigating the risk components associated with the situation. Materials and Methods: In this study, 50 renal transplant recipients on the day before (−1) and on days 10 (+10) and 30 (+30) days after kidney Tx were examined for the levels of serum phosphate (Pi). Levels of serum creatinine (Cr), Pi, 25-hydroxyvitamin D (25[OH] D), intact parathyroid hormone (iPTH) and fibroblast growth factor 23 (FGF-23), the 24 h urinary excretion of Pi and Cr, estimated glomerular filtration rate (eGFR), and the ratio of transport maximum of Pi (TMP) to eGFR (TMP/GFR) were evaluated on the same days. Results: Hypophosphatemia (serum Pi <2.5 mg/dl) was seen in 0%, 40%, and 42% of the patients on days −1, +10, and +30, respectively. The levels of 25(OH)D and iPTH were not significantly different in patients with and without hypophosphatemia on days +10 and +30. Compared to those with normophosphatemia, pre-Tx FGF-23 level was significantly higher in patients with hypophosphatemia on days +10 and +30, respectively. The regression coefficient of TMP/GFR and Cr was positive on days −1, +10, and +30. The coefficient of pre-Tx FGF-23 on post-Tx serum Pi was negative on days +10 (P < 0.03) and +30 (P < 0.003), and the coefficient of post-Tx FGF-23 was negative just on day +10 with serum Pi (P < 0.008). Conclusion: The main causes of post-Tx hypophosphatemia in the multivariate linear analysis were pre-Tx FGF-23 and post-Tx FGF-23 levels on days +10, post-Tx Cr, and TMP/GFR.
Collapse
Affiliation(s)
- Maryam Ghorbani
- Department of Internal Medicine, Ziaeian Hospital, Tehran University of Medical Sciences, Tehran, Iran
| | - Shahrzad Ossareh
- Department of Medicine, Hasheminejad Kidney Center, Iran University of Medical Sciences, Tehran, Iran
| |
Collapse
|
21
|
Kawabata C, Komaba H, Ishida H, Nakagawa Y, Hamano N, Koizumi M, Kanai G, Wada T, Nakamura M, Fukagawa M. Changes in Fibroblast Growth Factor 23 and Soluble Klotho Levels After Hemodialysis Initiation. Kidney Med 2019; 2:59-67. [PMID: 33015612 PMCID: PMC7525799 DOI: 10.1016/j.xkme.2019.09.007] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/01/2022] Open
Abstract
Rationale & Objective Patients with chronic kidney failure have markedly elevated fibroblast growth factor 23 (FGF-23) levels and decreased soluble Klotho levels. However, no studies have examined the effects of hemodialysis initiation on the levels of these hormones and other parameters of mineral metabolism. Study Design Prospective single-arm study. Setting & Participants 20 individuals with incident kidney failure initiating hemodialysis. Exposure Initiation of hemodialysis. Dose adjustments of phosphate binders and vitamin D receptor activators and use of calcimimetics, erythropoiesis-stimulating agents, and intravenous iron were prohibited. Outcomes Changes in serum levels of FGF-23, soluble Klotho, and other biochemical parameters of mineral metabolism, measured before and after each hemodialysis session, for a total of 4 sessions over 5 days. Analytical Approach Repeated-measures analysis of variance. Results At baseline, participants had 18-fold higher median FGF-23 levels and 1.6-fold lower mean soluble Klotho levels compared with age- and sex-matched healthy individuals. Initiation of hemodialysis led to progressive reductions in serum phosphorus, intact parathyroid hormone, and FGF-23 levels, with dialysis-related fluctuations. No reductions were observed in levels of α1-microglobulin, which has molecular weight comparable to FGF-23. The magnitude of the FGF-23 level reductions was strongly associated with concomitant changes in serum phosphorus levels but not with the changes in intact parathyroid hormone levels. Soluble Klotho levels did not change after the initiation of hemodialysis. Limitations Single-arm design, small sample size, short follow-up period. Conclusions Initiation of hemodialysis in patients with chronic kidney failure led to progressive reductions in FGF-23 levels in association with reductions in serum phosphorus levels. These results suggest that phosphorus is a strong inducer of FGF-23 production and that regulation of FGF-23 production is a rapid process.
Collapse
Affiliation(s)
- Chiaki Kawabata
- Division of Nephrology, Endocrinology and Metabolism, Tokai University School of Medicine, Isehara, Japan
| | - Hirotaka Komaba
- Division of Nephrology, Endocrinology and Metabolism, Tokai University School of Medicine, Isehara, Japan.,The Institute of Medical Sciences, Tokai University, Isehara, Japan
| | - Hiroaki Ishida
- Department of Transplant Surgery, Tokai University School of Medicine, Isehara, Japan
| | - Yosuke Nakagawa
- Division of Nephrology, Endocrinology and Metabolism, Tokai University School of Medicine, Isehara, Japan
| | - Naoto Hamano
- Division of Nephrology, Endocrinology and Metabolism, Tokai University School of Medicine, Isehara, Japan
| | - Masahiro Koizumi
- Division of Nephrology, Endocrinology and Metabolism, Tokai University School of Medicine, Isehara, Japan
| | - Genta Kanai
- Division of Nephrology, Endocrinology and Metabolism, Tokai University School of Medicine, Isehara, Japan
| | - Takehiko Wada
- Division of Nephrology, Endocrinology and Metabolism, Tokai University School of Medicine, Isehara, Japan
| | - Michio Nakamura
- Department of Transplant Surgery, Tokai University School of Medicine, Isehara, Japan
| | - Masafumi Fukagawa
- Division of Nephrology, Endocrinology and Metabolism, Tokai University School of Medicine, Isehara, Japan
| |
Collapse
|
22
|
Abstract
Chronic kidney disease (CKD) is a global health epidemic that accelerates cardiovascular disease, increases risk of infection, and causes anemia and bone disease, among other complications that collectively increase risk of premature death. Alterations in calcium and phosphate homeostasis have long been considered nontraditional risk factors for many of the most morbid outcomes of CKD. The discovery of fibroblast growth factor 23 (FGF23), which revolutionized the diagnosis and treatment of rare hereditary disorders of FGF23 excess that cause hypophosphatemic rickets, has also driven major paradigm shifts in our understanding of the pathophysiology and downstream end-organ complications of disordered mineral metabolism in CKD. As research of FGF23 in CKD has rapidly advanced, major new questions about its regulation and effects continuously emerge. These are promoting exciting innovations in laboratory, patient-oriented, and epidemiological research and stimulating clinical trials of new therapies and repurposing of existing ones to target FGF23.
Collapse
Affiliation(s)
- John Musgrove
- Division of Nephrology, Department of Medicine, Duke University School of Medicine, Durham, North Carolina 27710, USA;
| | - Myles Wolf
- Division of Nephrology, Department of Medicine, Duke University School of Medicine, Durham, North Carolina 27710, USA; .,Duke Clinical Research Institute, Duke University School of Medicine, Durham, North Carolina 27710, USA
| |
Collapse
|
23
|
Bone biomarkers in de novo renal transplant recipients. Clin Chim Acta 2019; 501:179-185. [PMID: 31734147 DOI: 10.1016/j.cca.2019.10.035] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2019] [Accepted: 10/24/2019] [Indexed: 12/30/2022]
Abstract
Successful kidney transplantation (partly) corrects the physiologic and metabolic abnormalities driving chronic kidney disease - mineral and bone disorders. At the same time, renal transplant recipients are exposed to immunosuppressive agents that may affect bone metabolism. Bone biomarkers have been suggested as surrogates of or adjuncts to bone biopsy and imaging techniques to assess bone health and to classify risk of bone loss and fractures. Bone biomarkers may be classified as circulating factors that affect bone metabolism (commonly referred to as bone metabolism markers) or that reflect bone cell number and/or activity (commonly referred to as bone turnover markers). A growing body of evidence shows that successful renal transplantation has a major impact on both bone metabolism and bone turnover. Analytical issues, including the cross-reactivity with fragments, complicate the interpretation of bone biomarkers, especially in the setting of a rapid changing kidney function, as is the case after successful renal transplantation. Overall, bone turnover seems to decline following renal transplantation, but inter-individual variability is substantial. Preliminary evidence indicates that bone biomarkers may be useful in guiding mineral and bone therapy in renal transplant recipients.
Collapse
|
24
|
Pathogenesis and treatment of electrolyte problems post transplant. Curr Opin Pediatr 2019; 31:213-218. [PMID: 30585865 DOI: 10.1097/mop.0000000000000715] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/25/2022]
Abstract
PURPOSE OF REVIEW Electrolyte abnormalities posttransplant are common occurrences that can have significant short-term and long-term effects on graft outcome and patient quality of life. Understanding the pathophysiology of these electrolyte derangements can help guide management to optimize bone health and minimize cardiovascular disease. This review explores the pathogenesis of the most common postrenal transplant electrolytes abnormalities as well as current treatment options. RECENT FINDINGS Clarifications of the role of FGF-23 has improved our understanding of posttransplant bone disease in addition to the known roles of hyperparathyroidism and vitamin D. The mechanisms of renal electrolyte wasting by immunosuppressive agents give insight into potential treatment options for hyperkalemia and hypomagnesemia. SUMMARY Understanding the pathogenesis of the common electrolyte abnormalities found post renal transplant may lead to targeted treatment options that in turn may improve transplant complications. Further studies are required to evaluate the effects on long-term outcomes of renal allografts.
Collapse
|
25
|
Uremic Toxins and Clinical Outcomes: The Impact of Kidney Transplantation. Toxins (Basel) 2018; 10:toxins10060229. [PMID: 29874852 PMCID: PMC6024850 DOI: 10.3390/toxins10060229] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2018] [Revised: 05/31/2018] [Accepted: 06/01/2018] [Indexed: 12/13/2022] Open
Abstract
Non-transplanted and transplanted patients with chronic kidney disease (CKD) differ in terms of mortality and the risk of clinical events. This difference is probably due to the difference of both traditional and non-traditional risk factors. Uremic retention solutes may constitute important non-traditional risk factors in this population. In the present review, we selected a set of uremic toxins that have been associated with harmful effects, and are an appealing target for adjuvant therapy in CKD. For each toxin reviewed here, relevant studies were selected and the relationship with hard clinical outcomes of uremic toxins were compared between non-transplanted CKD patients and transplanted patients taking into account the level of glomerular filtration rate in these two situations.
Collapse
|
26
|
Tiryaki O, Usalan C, Tarakcioglu M, Coban S. Calcitriol Reduces Albuminuria and Urinary Angiotensinogen Level in Renal Transplant Recipients. Transplant Proc 2018; 50:1342-1347. [DOI: 10.1016/j.transproceed.2018.01.055] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2017] [Accepted: 01/23/2018] [Indexed: 12/22/2022]
|
27
|
Piotti G, Gandolfini I, Palmisano A, Maggiore U. Metabolic risk profile in kidney transplant candidates and recipients. Nephrol Dial Transplant 2018; 34:388-400. [DOI: 10.1093/ndt/gfy151] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2018] [Accepted: 05/01/2018] [Indexed: 12/30/2022] Open
Affiliation(s)
- Giovanni Piotti
- Department of Nephrology, Azienda Ospedaliero-Universitaria di Parma, Parma, Italy
| | - Ilaria Gandolfini
- Department of Nephrology, Azienda Ospedaliero-Universitaria di Parma, Parma, Italy
| | - Alessandra Palmisano
- Department of Nephrology, Azienda Ospedaliero-Universitaria di Parma, Parma, Italy
| | - Umberto Maggiore
- Department of Nephrology, Azienda Ospedaliero-Universitaria di Parma, Parma, Italy
| |
Collapse
|
28
|
Affiliation(s)
- S. Bandini
- U. O. Nefrologia, Dialisi e Trapianto, Azienda Ospedaliera Careggi, Firenze
| |
Collapse
|
29
|
Araujo MJCLN, Ramalho JAM, Elias RM, Jorgetti V, Nahas W, Custodio M, Moysés RMA, David-Neto E. Persistent hyperparathyroidism as a risk factor for long-term graft failure: the need to discuss indication for parathyroidectomy. Surgery 2018; 163:1144-1150. [PMID: 29331397 DOI: 10.1016/j.surg.2017.12.010] [Citation(s) in RCA: 39] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2017] [Revised: 11/28/2017] [Accepted: 12/05/2017] [Indexed: 02/02/2023]
Abstract
BACKGROUND Although a successful kidney transplant (KTx) improves most of the mineral and bone disorders (MBD) produced by chronic kidney disease (CKD), hyperparathyroidism may persist (pHPT). Current guidelines recommend parathyroidectomy if serum parathormone is persistently elevated 1 year after KTx, because pHPT has been recently associated with poor graft outcomes. However, whether patients with pHPT and adequate renal function are at risk for long-term graft failure is unknown. METHODS Longitudinal follow-up of 911 adults submitted to KTx between January 2005 and December 2014, with estimated glomerular filtration rate (eGFR) ≥ 30 mL/min 1 year after surgery. Clinical and laboratory data were collected from electronic database. Graft failure was defined as return to dialysis. RESULTS Overall, 62% of the patients were classified as having pHPT 1 year after KTx. After a mean follow-up time of 47 months, there were 59 graft failures (49 in pHPT and 10 in non-pHPT group, P = .003). At last follow-up, death-censored graft survival was lower in the pHPT group (P = .009), even after adjustment for age at KTx, donor age, donor type, acute rejection, parathyroidectomy, and eGFR at 1 year after transplantation (odds ratio [OR] 1.99; 1.004-3.971; P = .049). A PTH of 150 pg/mL at 6 months was the best cutoff to predict pHPT at 1 year (specificity = 92.1%). CONCLUSION Having pHPT after a successful KTx increases the long-term risk of death-censored graft failure. This result highlights the need for better recognition and management of CKD-MBD before and during the first year after KTx, and opens a discussion on the more appropriate timing to perform parathyroidectomy.
Collapse
Affiliation(s)
- Maria Júlia Correia Lima Nepomuceno Araujo
- Renal Transplantation Service, São Paulo University School of Medicine, São Paulo, Brazil; Nephrology Division, São Paulo University School of Medicine, São Paulo, Brazil
| | - Janaina Almeida Mota Ramalho
- Renal Transplantation Service, São Paulo University School of Medicine, São Paulo, Brazil; Nephrology Division, São Paulo University School of Medicine, São Paulo, Brazil
| | | | - Vanda Jorgetti
- Nephrology Division, São Paulo University School of Medicine, São Paulo, Brazil
| | - William Nahas
- Renal Transplantation Service, São Paulo University School of Medicine, São Paulo, Brazil
| | - Melani Custodio
- Nephrology Division, São Paulo University School of Medicine, São Paulo, Brazil
| | - Rosa M A Moysés
- Nephrology Division, São Paulo University School of Medicine, São Paulo, Brazil.
| | - Elias David-Neto
- Renal Transplantation Service, São Paulo University School of Medicine, São Paulo, Brazil; Nephrology Division, São Paulo University School of Medicine, São Paulo, Brazil
| |
Collapse
|
30
|
Tsagalis G, Psimenou E, Manios E, Laggouranis A. Fibroblast Growth Factor 23 (FGF23) and the kidney. Int J Artif Organs 2018; 32:232-9. [DOI: 10.1177/039139880903200407] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
Phosphate homeostasis in humans is a complex phenomenon involving the interplay of several different organs and circulating hormones. Among the latter, parathyroid hormone (PTH), and vitamin D3 (Vit D3) were thought to be the main regulators of serum phosphate concentration since they mediated the intestinal, renal and bone responses that follow fluctuations in serum phosphate levels. The study of three rare disorders – tumor-induced osteomalacia (TIO), autosomal dominant hypophosphatemic rickets (ADHR) and X-linked hypophosphatemic rickets (XLH) – has offered a completely new insight into phosphate metabolism by unraveling the role of a group of peptides that can directly affect serum phosphate concentration by increasing urinary phosphate excretion. Fibroblast growth factor-23 (FGF-23) is the most extensively studied “phosphatonin”. The production, mechanism of action, effects in various target tissues, and its role in common clinical disorders are the focus of this review.
Collapse
Affiliation(s)
| | | | - Efstathios Manios
- Department of Clinical Therapeutics, Alexandra Hospital, Athens - Greece
| | | |
Collapse
|
31
|
Cruzado JM, Lauzurica R, Pascual J, Marcen R, Moreso F, Gutierrez-Dalmau A, Andrés A, Hernández D, Torres A, Beneyto MI, Melilli E, Manonelles A, Arias M, Praga M. Paricalcitol Versus Calcifediol for Treating Hyperparathyroidism in Kidney Transplant Recipients. Kidney Int Rep 2018; 3:122-132. [PMID: 29340322 PMCID: PMC5762965 DOI: 10.1016/j.ekir.2017.08.016] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2017] [Revised: 08/23/2017] [Accepted: 08/29/2017] [Indexed: 02/06/2023] Open
Abstract
INTRODUCTION Secondary hyperparathyroidism (SHPT) and vitamin D deficiency are common at kidney transplantation and are associated with some early and late complications. This study was designed to evaluate whether paricalcitol was more effective than nutritional vitamin D for controlling SHPT in de novo kidney allograft recipients. METHODS This was a 6-month, investigator-initiated, multicenter, open-label, randomized clinical trial. Patients with pretransplantation iPTH between 250 and 600 pg/ml and calcium <10 mg/dl were randomized to paricalcitol (PAR) or calcifediol (CAL). The intention-to-treat population (PAR: n = 46; CAL: n = 47) was used for the analysis. The primary endpoint was the percentage of patients with serum iPTH >110 pg/ml at 6 months. Secondary endpoints were bone mineral metabolism, renal function, and allograft protocol biopsies. RESULTS The primary outcome occurred in 19.6% of patients in the PAR group and 36.2% of patients in the CAL group (P = 0.07). However, there was a higher percentage of patients with iPTH <70 pg/ml in the PAR group than in the CAL group (63.4% vs. 37.2%; P = 0.03). No differences were observed in bone turnover biomarkers and bone mineral density. The estimated glomerular filtration rate was significantly higher in the CAL group than in the PAR group without differences in albuminuria. In protocol biopsies, interstitial fibrosis and tubular atrophy tended to be higher in the PAR group than in the CAL group (48% vs. 23.8%; P = 0.09). Both medications were well tolerated. CONCLUSION Both PAR and CAL reduced iPTH, but PAR was associated with a higher proportion of patients with iPTH <70 pg/ml. These results do not support the use of PAR to treat posttransplantation hyperparathyroidism.
Collapse
Affiliation(s)
- Josep M. Cruzado
- Nephrology Department, Hospital Universitari de Bellvitge, University of Barcelona, IDIBELL, L’Hospitalet de Llobregat, Barcelona, Spain
| | - Ricardo Lauzurica
- Department of Nephrology, Hospital Universitari Germans Trias i Pujol, Badalona, Spain
| | - Julio Pascual
- Department of Nephrology, Hospital del Mar, Barcelona, Spain
| | - Roberto Marcen
- Department of Nephrology, Hospital Universitario Ramón y Cajal, Madrid, Spain
| | - Francesc Moreso
- Hospital Universitari Vall d'Hebron, Nephrology, Barcelona, Spain
| | | | - Amado Andrés
- Department of Nephrology, Hospital 12 de Octubre, Madrid, Spain
| | | | - Armando Torres
- Department of Nephrology, Hospital Universitario de Canarias, Tenerife, Spain
| | | | - Edoardo Melilli
- Nephrology Department, Hospital Universitari de Bellvitge, University of Barcelona, IDIBELL, L’Hospitalet de Llobregat, Barcelona, Spain
| | - Anna Manonelles
- Nephrology Department, Hospital Universitari de Bellvitge, University of Barcelona, IDIBELL, L’Hospitalet de Llobregat, Barcelona, Spain
| | - Manuel Arias
- Nephrology Department, Hospital Universitario Marqués de Valdecilla, Santander, Spain
| | - Manuel Praga
- Department of Nephrology, Hospital 12 de Octubre, Madrid, Spain
| |
Collapse
|
32
|
Boyle SM, Malat G, Harhay MN, Lee DH, Pang L, Talluri S, Sharma A, Bias TE, Ranganna K, Doyle AM. Association of tenofovir disoproxil fumarate with primary allograft survival in HIV-positive kidney transplant recipients. Transpl Infect Dis 2017; 19. [PMID: 28520146 DOI: 10.1111/tid.12727] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2016] [Revised: 03/09/2017] [Accepted: 03/18/2017] [Indexed: 12/29/2022]
Abstract
BACKGROUND Tenofovir disoproxil fumarate (TDF) is an antiretroviral agent frequently used to treat human immunodeficiency virus (HIV). There are concerns regarding its potential to cause acute kidney injury, chronic kidney disease, and proximal tubulopathy. Although TDF can effectively suppress HIV after kidney transplantation, it is unknown whether use of TDF-based antiretroviral therapy (ART) after kidney transplantation adversely affects allograft survival. METHODS We examined 104 HIV+ kidney transplant (KT) recipients at our center between 2001 and 2014. We generated a propensity score for TDF treatment using recipient and donor characteristics. We then fit Cox proportional hazards models to investigate the association between TDF treatment and 3-year, death-censored primary allograft failure, adjusting for the propensity score and delayed graft function (DGF). RESULTS Of the 104 HIV+ KT candidates who underwent transplantation during the study period, 23 (22%) were maintained on TDF-based ART at the time of transplantation, and 81 (78%) were on non-TDF-based ART. Median age of the cohort was 48 years; 87% were male; 88% were black; and median CD4 count at transplantation was 450 cells/mm3 . Median kidney donor risk index was 1.2. At 3 years post transplantation, primary allograft failure occurred in 26% of patients on TDF-based ART and in 28% of patients on non-TDF-based ART (P=.5). TDF treatment was not associated with primary allograft failure at 3 years post transplant after adjusting for DGF and a propensity score for TDF use (hazard ratio 2.12, 95% confidence interval 0.41-10.9). CONCLUSIONS In a large single-center experience of HIV+ kidney transplantation, TDF use following kidney transplantation was not significantly associated with primary allograft failure. These results may help inform management for HIV+ KT recipients in need of TDF therapy for adequate viral suppression.
Collapse
Affiliation(s)
- Suzanne M Boyle
- Division of Nephrology, Drexel University College of Medicine, Philadelphia, PA, USA
| | - Gregory Malat
- Department of Pharmacy, Hahnemann University Hospital, Philadelphia, PA, USA
| | - Meera N Harhay
- Division of Nephrology, Drexel University College of Medicine, Philadelphia, PA, USA
| | - Dong H Lee
- Division of Infectious Disease, Drexel University College of Medicine, Philadelphia, PA, USA
| | - Lisa Pang
- Jefferson College of Pharmacy, Thomas Jefferson University, Philadelphia, PA, USA
| | - Sindhura Talluri
- Division of Nephrology, Drexel University College of Medicine, Philadelphia, PA, USA
| | - Akshay Sharma
- Division of Nephrology, Drexel University College of Medicine, Philadelphia, PA, USA
| | - Tiffany E Bias
- Department of Pharmacy, Hahnemann University Hospital, Philadelphia, PA, USA
| | - Karthik Ranganna
- Division of Nephrology, Drexel University College of Medicine, Philadelphia, PA, USA
| | - Alden M Doyle
- Division of Nephrology, University of Virginia School of Medicine, Charlottesville, VA, USA
| |
Collapse
|
33
|
Coskun Y, Paydas S, Balal M, Soyupak S, Kara E. Bone Disease and Serum Fibroblast Growth Factor-23 Levels in Renal Transplant Recipients. Transplant Proc 2017; 48:2040-5. [PMID: 27569941 DOI: 10.1016/j.transproceed.2016.05.012] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2015] [Revised: 04/19/2016] [Accepted: 05/04/2016] [Indexed: 11/16/2022]
Abstract
BACKGROUND Posttransplantation bone disease develops commonly and results in important complications. In this study, we aimed to investigate the relationship between bone diseases and serum fibroblast growth factor-23 (FGF-23) in renal transplant recipients. METHODS This study was conducted in 106 kidney transplant recipients (KTrs; group G1) and 30 patients with chronic kidney disease (group G2). Patients with fever, heart failure, angina pectoris, acute renal failure, malignant disease, or any gastrointestinal disease were excluded. KTrs were treated with triple immunosuppressive drugs including glucocorticoids. Complete blood count (CBC), blood urea nitrogen (BUN), creatinine, glomerular filtration rate (GFR, Modification of Diet in Renal Disease [MDRD] formula), lipid profile, calcium (Ca), phosphorous (P), parathormone (PTH), 25OHD3, serum levels of tacrolimus/cyclosporine, and intact FGF-23 were measured. Bone mineral density (BMD) was measured with dual energy X-ray absorptiometry. RESULTS The mean patient age was 40.1 ± 11.1 years and 39.2 ± 11.3 years in G1 and G2, respectively (P > .05). In G1 and G2, 76 and 15 patients were male, respectively. Compared with the G2 patients, G1 patients had lower body mass index (BMI), serum glucose levels, P, Mg, and Ca·P (P < .05 for all). T scores of the lumbar vertebrae/femur were -1.82 ± 0.99/-1.34 ± 0.89 and -1.13 ± 1.34/-0.51 ± 1.18 in G1 and G2 patients, respectively (P < .05 for all). The incidences of osteopenia/osteoporosis in the lumbar spine and femur were 50.9%/27.4% and 57.5%/10.4% in G1 and 16.6%/23.3%, and 40%/3.3% in G2. There were positive correlations between BMD and BMI, the time elapsed after renal transplantation, and GFR. In our study, a statistically significant relationship was found between lipid parameters and BMD, PTH, and 250HD3 levels, as well as use of corticosteroid and calcineurin inhibitors (P < .05 for all). In G1 and G2, BMD of the lumbar spine in patients with serum creatinine >1.5 mg/dL was lower than that in patients with serum creatinine <1.5 mg/dL. CONCLUSION The association between age and BMD was found only in the femur of KTrs. No relationship was observed between serum FGF-23 levels and BMD values. In both groups, the BMD T score of the lumbar spine was lower compared to the BMD T score of the femur and in patients with serum creatinine >1.5 mg/dL. In long-term follow-up of renal transplantation by as much as 58 months, the incidence of bone disease such as osteoporosis/osteopenia was as high as 67% and was also higher than that of nontransplant patients with similar GFR. In addition to decreased renal function, dyslipidemia, inflammation, and continuing hypophosphatemia were also accompanied by decreased BMD as in cardiovascular disease in KTrs.
Collapse
Affiliation(s)
- Y Coskun
- Department of Internal Medicine and Nephrology, Cukurova University Faculty of Medicine, Adana, Turkey
| | - S Paydas
- Department of Internal Medicine and Nephrology, Cukurova University Faculty of Medicine, Adana, Turkey.
| | - M Balal
- Department of Internal Medicine and Nephrology, Cukurova University Faculty of Medicine, Adana, Turkey
| | - S Soyupak
- Department of Radiology, Cukurova University Faculty of Medicine, Adana, Turkey
| | - E Kara
- Department of Public Health, Cukurova University Faculty of Medicine, Adana, Turkey
| |
Collapse
|
34
|
Kuczera P, Adamczak M, Wiecek A. Fibroblast Growth Factor-23-A Potential Uremic Toxin. Toxins (Basel) 2016; 8:toxins8120369. [PMID: 27941640 PMCID: PMC5198563 DOI: 10.3390/toxins8120369] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2016] [Revised: 11/30/2016] [Accepted: 12/01/2016] [Indexed: 12/25/2022] Open
Abstract
Fibroblast growth factor-23 (FGF23) is a circulating member of the FGF family produced mainly by the osteocytes and osteoblasts that can act as a hormone. The main action of FGF23 is to lower phosphatemia via the reduction of urinary phosphate reabsorption and the decrease of 1,25(OH)2-D generation in the kidney. In the course of chronic kidney disease (CKD), plasma FGF23 concentration rises early, most probably to compensate the inability of the deteriorating kidneys to excrete an adequate amount of phosphate. However, this comes at the cost of FGF23-related target organ toxicity. Results of clinical studies suggest that elevated plasma FGF23 concentration is independently associated with the increased risk of CKD progression, occurrence of cardio-vascular complications, and mortality in different stages of CKD. FGF23 also contributes to cardiomyocyte hypertrophy, vascular calcification, and endothelial dysfunction. The impact of FGF23 on heart muscle is not dependent on Klotho, but rather on the PLCγ–calcineurin–NFAT (nuclear factor of activated T-cells) pathway. Among the factors increasing plasma FGF23 concentration, active vitamin D analogues play a significant role. Additionally, inflammation and iron deficiency can contribute to the increase of plasma FGF23. Among the factors decreasing plasma FGF23, dietary phosphate restriction, some intestinal phosphate binders, cinacalcet (and other calcimimetics), and nicotinamide can be enumerated. Anti-FGF23 antibodies have also recently been developed to inhibit the action of FGF23 in target organs. Still, the best way to normalize plasma FGF23 in maintenance hemodialysis patients is restoring kidney function by successful kidney transplantation.
Collapse
Affiliation(s)
- Piotr Kuczera
- Department of Nephrology, Transplantation and Internal Medicine, Medical University of Silesia, Katowice 40-027, Poland.
| | - Marcin Adamczak
- Department of Nephrology, Transplantation and Internal Medicine, Medical University of Silesia, Katowice 40-027, Poland.
| | - Andrzej Wiecek
- Department of Nephrology, Transplantation and Internal Medicine, Medical University of Silesia, Katowice 40-027, Poland.
| |
Collapse
|
35
|
Prasad N, Jaiswal A, Agarwal V, Kumar S, Chaturvedi S, Yadav S, Gupta A, Sharma RK, Bhadauria D, Kaul A. FGF23 is associated with early post-transplant hypophosphataemia and normalizes faster than iPTH in living donor renal transplant recipients: a longitudinal follow-up study. Clin Kidney J 2016; 9:669-76. [PMID: 27679713 PMCID: PMC5036900 DOI: 10.1093/ckj/sfw065] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2016] [Accepted: 06/03/2016] [Indexed: 01/28/2023] Open
Abstract
Background We aimed to longitudinally analyse changes in the levels of serum fibroblast growth factor 23 (FGF23), intact parathyroid hormone (iPTH) and associated minerals in patients undergoing renal transplantation. Methods Sixty-three patients with end-stage renal disease (ESRD) who underwent living donor transplantation were recruited. Serum FGF23, iPTH, uric acid, inorganic phosphorous (iP), blood urea nitrogen and serum creatinine were measured pre-transplant and at 1 (M1), 3 (M3) and 12 months (M12) post-transplantation. Results FGF23 levels were decreased at M1, M3 and M12 by 93.81, 96.74 and 97.53%, respectively. iPTH levels were decreased by 67.95, 74.95 and 84.9%, respectively. The prevalence of hyperparathyroidism at M1, M3 and M12 post-transplantation was 63.5, 42.9 and 11.1%, respectively. FGF23 and iP levels remained above the normal range in 23 (36.5%) and 17 (27%) patients at M1, 10 (15.9%) and 5 (8%) at M3 and in none at M12 post-transplantation, respectively. A multivariate regression model revealed that, pre-transplant, iP was positively associated with iPTH (P = 0.016) but not with FGF 23; however, post-transplant, iP level was negatively associated with FGF23 (P < 0.001) but not with iPTH. Conclusions Post-transplant FGF23 levels settle faster than those of iPTH. However, 11% of patients continued to have hyperparathyroidism even after 12 months.
Collapse
Affiliation(s)
- Narayan Prasad
- Department of Nephrology and Renal Transplantation , Sanjay Gandhi Postgraduate Institute of Medical Sciences , Lucknow , Uttar Pradesh, India
| | - Akhilesh Jaiswal
- Department of Nephrology and Renal Transplantation , Sanjay Gandhi Postgraduate Institute of Medical Sciences , Lucknow , Uttar Pradesh, India
| | - Vikas Agarwal
- Clinical Immunology , Sanjay Gandhi Postgraduate Institute of Medical Sciences , Lucknow , Uttar Pradesh, India
| | - Shashi Kumar
- Department of Nephrology and Renal Transplantation , Sanjay Gandhi Postgraduate Institute of Medical Sciences , Lucknow , Uttar Pradesh, India
| | - Saurabh Chaturvedi
- Clinical Immunology , Sanjay Gandhi Postgraduate Institute of Medical Sciences , Lucknow , Uttar Pradesh, India
| | - Subhash Yadav
- Endocrinology , Sanjay Gandhi Postgraduate Institute of Medical Sciences , Lucknow , Uttar Pradesh, India
| | - Amit Gupta
- Department of Nephrology and Renal Transplantation , Sanjay Gandhi Postgraduate Institute of Medical Sciences , Lucknow , Uttar Pradesh, India
| | - Raj K Sharma
- Department of Nephrology and Renal Transplantation , Sanjay Gandhi Postgraduate Institute of Medical Sciences , Lucknow , Uttar Pradesh, India
| | - Dharmendra Bhadauria
- Department of Nephrology and Renal Transplantation , Sanjay Gandhi Postgraduate Institute of Medical Sciences , Lucknow , Uttar Pradesh, India
| | - Anupama Kaul
- Department of Nephrology and Renal Transplantation , Sanjay Gandhi Postgraduate Institute of Medical Sciences , Lucknow , Uttar Pradesh, India
| |
Collapse
|
36
|
Abstract
The last 25 years have been characterized by dramatic improvements in short-term patient and allograft survival after kidney transplantation. Long-term patient and allograft survival remains limited by cardiovascular disease and chronic allograft injury, among other factors. Cardiovascular disease remains a significant contributor to mortality in native chronic kidney disease as well as cardiovascular mortality in chronic kidney disease more than doubles that of the general population. The chronic kidney disease (CKD)-mineral bone disorder (MBD) is a syndrome recently coined to embody the biochemical, skeletal, and cardiovascular pathophysiology that results from disrupting the complex systems biology between the kidney, skeleton, and cardiovascular system in native and transplant kidney disease. The CKD-MBD is a unique kidney disease-specific syndrome containing novel cardiovascular risk factors, with an impact reaching far beyond traditional notions of renal osteodystrophy and hyperparathyroidism. This overview reviews current knowledge of the pathophysiology of the CKD-MBD, including emerging concepts surrounding the importance of circulating pathogenic factors released from the injured kidney that directly cause cardiovascular disease in native and transplant chronic kidney disease, with potential application to mechanisms of chronic allograft injury and vasculopathy.
Collapse
|
37
|
Zhang J, Li Y. Therapeutic uses of FGFs. Semin Cell Dev Biol 2016; 53:144-54. [DOI: 10.1016/j.semcdb.2015.09.007] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2015] [Accepted: 09/07/2015] [Indexed: 01/23/2023]
|
38
|
Wolf M, Weir MR, Kopyt N, Mannon RB, Von Visger J, Deng H, Yue S, Vincenti F. A Prospective Cohort Study of Mineral Metabolism After Kidney Transplantation. Transplantation 2016; 100:184-93. [PMID: 26177089 PMCID: PMC4683035 DOI: 10.1097/tp.0000000000000823] [Citation(s) in RCA: 95] [Impact Index Per Article: 11.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/16/2023]
Abstract
Kidney transplantation corrects or improves many complications of chronic kidney disease, but its impact on disordered mineral metabolism is incompletely understood. The prevalence of posttransplant hyperparathyroidism was 86% at 12 months (PTH >65 pg/ml) but only 40% (PTH >130 mg/dL) in the absence of cinacalcet, vitamin D sterols, or parathyroidectomy. Intact fibroblast growth factor 23 decreased rapidly to G40 pg/ml by 3 months posttransplant. Supplemental digital content is available in the text.
Collapse
Affiliation(s)
- Myles Wolf
- 1 Division of Nephrology and Hypertension, Department of Medicine, Northwestern University Feinberg School of Medicine, Chicago, IL. 2 Division of Nephrology, Department of Medicine, University of Maryland School of Medicine, Baltimore, MD. 3 Lehigh Valley Hospital, Allentown, PA. 4 University of Alabama at Birmingham, Birmingham, AL. 5 The Ohio State University Medical Center, Columbus, OH. 6 Amgen Inc., Thousand Oaks, CA. 7 Kidney Transplant Service, University of California San Francisco, San Francisco, CA
| | | | | | | | | | | | | | | |
Collapse
|
39
|
Taweesedt PT, Disthabanchong S. Mineral and bone disorder after kidney transplantation. World J Transplant 2015; 5:231-242. [PMID: 26722650 PMCID: PMC4689933 DOI: 10.5500/wjt.v5.i4.231] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/25/2015] [Revised: 09/11/2015] [Accepted: 10/27/2015] [Indexed: 02/05/2023] Open
Abstract
After successful kidney transplantation, accumulated waste products and electrolytes are excreted and regulatory hormones return to normal levels. Despite the improvement in mineral metabolites and mineral regulating hormones after kidney transplantation, abnormal bone and mineral metabolism continues to present in most patients. During the first 3 mo, fibroblast growth factor-23 (FGF-23) and parathyroid hormone levels decrease rapidly in association with an increase in 1,25-dihydroxyvitamin D production. Renal phosphate excretion resumes and serum calcium, if elevated before, returns toward normal levels. FGF-23 excess during the first 3-12 mo results in exaggerated renal phosphate loss and hypophosphatemia occurs in some patients. After 1 year, FGF-23 and serum phosphate return to normal levels but persistent hyperparathyroidism remains in some patients. The progression of vascular calcification also attenuates. High dose corticosteroid and persistent hyperparathyroidism are the most important factors influencing abnormal bone and mineral metabolism in long-term kidney transplant (KT) recipients. Bone loss occurs at a highest rate during the first 6-12 mo after transplantation. Measurement of bone mineral density is recommended in patients with estimated glomerular filtration rate > 30 mL/min. The use of active vitamin D with or without bisphosphonate is effective in preventing early post-transplant bone loss. Steroid withdrawal regimen is also beneficial in preservation of bone mass in long-term. Calcimimetic is an alternative therapy to parathyroidectomy in KT recipients with persistent hyperparathyroidism. If parathyroidectomy is required, subtotal to near total parathyroidectomy is recommended. Performing parathyroidectomy during the waiting period prior to transplantation is also preferred in patients with severe hyperparathyroidism associated with hypercalcemia.
Collapse
|
40
|
Cruzado JM, Moreno P, Torregrosa JV, Taco O, Mast R, Gómez-Vaquero C, Polo C, Revuelta I, Francos J, Torras J, García-Barrasa A, Bestard O, Grinyó JM. A Randomized Study Comparing Parathyroidectomy with Cinacalcet for Treating Hypercalcemia in Kidney Allograft Recipients with Hyperparathyroidism. J Am Soc Nephrol 2015; 27:2487-94. [PMID: 26647424 DOI: 10.1681/asn.2015060622] [Citation(s) in RCA: 108] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2015] [Accepted: 11/01/2015] [Indexed: 12/16/2022] Open
Abstract
Tertiary hyperparathyroidism is a common cause of hypercalcemia after kidney transplant. We designed this 12-month, prospective, multicenter, open-label, randomized study to evaluate whether subtotal parathyroidectomy is more effective than cinacalcet for controlling hypercalcemia caused by persistent hyperparathyroidism after kidney transplant. Kidney allograft recipients with hypercalcemia and elevated intact parathyroid hormone (iPTH) concentration were eligible if they had received a transplant ≥6 months before the study and had an eGFR>30 ml/min per 1.73 m(2) The primary end point was the proportion of patients with normocalcemia at 12 months. Secondary end points were serum iPTH concentration, serum phosphate concentration, bone mineral density, vascular calcification, renal function, patient and graft survival, and economic cost. In total, 30 patients were randomized to receive cinacalcet (n=15) or subtotal parathyroidectomy (n=15). At 12 months, ten of 15 patients in the cinacalcet group and 15 of 15 patients in the parathyroidectomy group (P=0.04) achieved normocalcemia. Normalization of serum phosphate concentration occurred in almost all patients. Subtotal parathyroidectomy induced greater reduction of iPTH and associated with a significant increase in femoral neck bone mineral density; vascular calcification remained unchanged in both groups. The most frequent adverse events were digestive intolerance in the cinacalcet group and hypocalcemia in the parathyroidectomy group. Surgery would be more cost effective than cinacalcet if cinacalcet duration reached 14 months. All patients were alive with a functioning graft at the end of follow-up. In conclusion, subtotal parathyroidectomy was superior to cinacalcet in controlling hypercalcemia in these patients with kidney transplants and persistent hyperparathyroidism.
Collapse
Affiliation(s)
- Josep M Cruzado
- Nephrology Department, Hospital Universitari de Bellvitge, University of Barcelona, Institut d'Investigació Biomèdica de Bellvitge (IDIBELL), L'Hospitalet de Llobregat, Barcelona, Spain;
| | | | - José V Torregrosa
- Nephrology and Renal Transplant Service, Hospital Clínic, Barcelona, Spain
| | - Omar Taco
- Nephrology Department, Hospital Universitari de Bellvitge, University of Barcelona, Institut d'Investigació Biomèdica de Bellvitge (IDIBELL), L'Hospitalet de Llobregat, Barcelona, Spain
| | | | - Carmen Gómez-Vaquero
- Rheumatology, Hospital Universitari de Bellvitge, L'Hospitalet de Llobregat, Barcelona, Spain; and
| | - Carolina Polo
- Nephrology Department, Hospital Universitari de Bellvitge, University of Barcelona, Institut d'Investigació Biomèdica de Bellvitge (IDIBELL), L'Hospitalet de Llobregat, Barcelona, Spain
| | - Ignacio Revuelta
- Nephrology and Renal Transplant Service, Hospital Clínic, Barcelona, Spain
| | | | - Joan Torras
- Nephrology Department, Hospital Universitari de Bellvitge, University of Barcelona, Institut d'Investigació Biomèdica de Bellvitge (IDIBELL), L'Hospitalet de Llobregat, Barcelona, Spain
| | | | - Oriol Bestard
- Nephrology Department, Hospital Universitari de Bellvitge, University of Barcelona, Institut d'Investigació Biomèdica de Bellvitge (IDIBELL), L'Hospitalet de Llobregat, Barcelona, Spain
| | - Josep M Grinyó
- Nephrology Department, Hospital Universitari de Bellvitge, University of Barcelona, Institut d'Investigació Biomèdica de Bellvitge (IDIBELL), L'Hospitalet de Llobregat, Barcelona, Spain
| |
Collapse
|
41
|
Hirukawa T, Kakuta T, Nakamura M, Fukagawa M. Mineral and bone disorders in kidney transplant recipients: reversible, irreversible, and de novo abnormalities. Clin Exp Nephrol 2015; 19:543-55. [DOI: 10.1007/s10157-015-1117-z] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2014] [Accepted: 04/12/2015] [Indexed: 12/18/2022]
|
42
|
Barros X, Fuster D, Paschoalin R, Oppenheimer F, Rubello D, Perlaza P, Pons F, Torregrosa JV. Changes in bone mineral metabolism parameters, including FGF23, after discontinuing cinacalcet at kidney transplantation. Endocrine 2015; 49:267-73. [PMID: 25154517 DOI: 10.1007/s12020-014-0400-1] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/20/2014] [Accepted: 08/18/2014] [Indexed: 12/11/2022]
Abstract
Little is known about the effects of the administration of cinacalcet in dialytic patients who are scheduled for kidney transplantation, and in particular about the changes in FGF23 and other mineral metabolism parameters after surgery compared with recipients not on cinacalcet at kidney transplantation. We performed a prospective observational cohort study with recruitment of consecutive kidney transplant recipients at our institution. Patients were classified according to whether they were under treatment with cinacalcet before transplantation. Bone mineral metabolism parameters, including C-terminal FGF23, were measured at baseline, on day 15, and at 1, 3, and 6 months after transplantation. In previously cinacalcet-treated patients, cinacalcet therapy was discontinued on the day of surgery and was not restarted after transplantation. A total of 48 kidney transplant recipients, 20 on cinacalcet at surgery and 28 cinacalcet non-treated patients, completed the follow-up. Serum phosphate declined significantly in the first 15 days after transplantation with no differences between the two groups, whereas cinacalcet-treated patients showed higher FGF23 levels, although not significant. After transplantation, PTH and serum calcium were significantly higher in cinacalcet-treated patients. We conclude that patients receiving cinacalcet on dialysis presented similar serum phosphate levels but higher PTH and serum calcium levels during the initial six months after kidney transplantation than cinacalcet non-treated patients. The group previously treated with cinacalcet before transplantation showed higher FGF23 levels without significant differences, so further studies should investigate its relevance in the management of these patients.
Collapse
Affiliation(s)
- Xoana Barros
- Nephrology and Renal Transplant Department, Hospital Clinic, Barcelona, Spain
| | | | | | | | | | | | | | | |
Collapse
|
43
|
Guo YC, Yuan Q. Fibroblast growth factor 23 and bone mineralisation. Int J Oral Sci 2015; 7:8-13. [PMID: 25655009 PMCID: PMC4817534 DOI: 10.1038/ijos.2015.1] [Citation(s) in RCA: 61] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 12/23/2014] [Indexed: 02/07/2023] Open
Abstract
Fibroblast growth factor 23 (FGF23) is a hormone that is mainly secreted by osteocytes and osteoblasts in bone. The critical role of FGF23 in mineral ion homeostasis was first identified in human genetic and acquired rachitic diseases and has been further characterised in animal models. Recent studies have revealed that the levels of FGF23 increase significantly at the very early stages of chronic kidney disease (CKD) and may play a critical role in mineral ion disorders and bone metabolism in these patients. Our recent publications have also shown that FGF23 and its cofactor, Klotho, may play an independent role in directly regulating bone mineralisation instead of producing a systematic effect. In this review, we will discuss the new role of FGF23 in bone mineralisation and the pathophysiology of CKD-related bone disorders.
Collapse
Affiliation(s)
- Yu-Chen Guo
- State Key Laboratory of Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, China
| | - Quan Yuan
- State Key Laboratory of Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, China
| |
Collapse
|
44
|
Sawires HK, Essam RM, Morgan MF, Mahmoud RA. Serum klotho: relation to fibroblast growth factor-23 and other regulators of phosphate metabolism in children with chronic kidney disease. Nephron Clin Pract 2015; 129:293-9. [PMID: 25766835 DOI: 10.1159/000377633] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2014] [Accepted: 01/31/2015] [Indexed: 11/19/2022] Open
Abstract
FGF23 and Klotho synergize to regulate phosphate homeostasis by promoting renal phosphate excretion. Chronic kidney disease (CKD) may be viewed as a state of FGF23 resistance caused by Klotho deficiency. This viewpoint explains several observations on phosphate metabolism in CKD that lack mechanistic insights. Our objectives were to correlate serum klotho and FGF-23 with other variables that regulate phosphate metabolism. We studied 40 patients with CKD on conservative treatment (group A), 44 patients with end-stage renal disease (ESRD) on regular hemodialysis (group B), 40 kidney transplant recipients (KTR) (group C) and 40 healthy controls for measuring serum klotho and FGF-23. Blood samples were withdrawn for measuring the levels of serum Calcium (Ca), Phosphorus (P), alkaline phosphatase (ALP), 1,25 (OH)2 D3, intact parathyroid hormone (PTH), FGF-23 and α klotho. The mean levels of FGF-23 and α klotho in control group were 225.78 ± 111.05 pg/ml (range: 102.4, 418.5) and 6.78 ± 1.90 ng/ml (range: 4, 11), respectively. The mean levels of FGF-23 in the 3 studied groups were 1,034.2 ± 84.6, 1,288.7 ± 131.4 and 1,008.7 ± 117.6 pg/ml, respectively. The median levels of s-klotho in the 3 studied groups were 3.15, 2.3 and 2.95, respectively. It was found that FGF-23 was significantly increased and α klotho was significantly decreased in all patients when compared with those in the control group (p < 0.001, <0.001, respectively). We found that there was a significant inverse correlation between serum Ca and α klotho in the studied groups. There was no significant correlation between FGF-23 and α klotho in the studied groups (p > 0.05). We have shown that circulating s-klotho was not related to FGF-23 in CKD, dialysis and KTR patients. In addition, we demonstrated a novel association between serum Ca and s-klotho that needs to be further studied.
Collapse
|
45
|
Courbebaisse M, Alberti C, Colas S, Prié D, Souberbielle JC, Treluyer JM, Thervet E. VITamin D supplementation in renAL transplant recipients (VITALE): a prospective, multicentre, double-blind, randomized trial of vitamin D estimating the benefit and safety of vitamin D3 treatment at a dose of 100,000 UI compared with a dose of 12,000 UI in renal transplant recipients: study protocol for a double-blind, randomized, controlled trial. Trials 2014; 15:430. [PMID: 25376735 PMCID: PMC4233037 DOI: 10.1186/1745-6215-15-430] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2014] [Accepted: 10/15/2014] [Indexed: 12/21/2022] Open
Abstract
Background In addition to their effects on bone health, high doses of cholecalciferol may have beneficial non-classic effects including the reduction of incidence of type 2 diabetes mellitus, cardiovascular disease, and cancer. These pleiotropic effects have been documented in observational and experimental studies or in small intervention trials. Vitamin D insufficiency is a frequent finding in renal transplant recipients (RTRs), and this population is at risk of the previously cited complications. Methods/design The VITALE study is a prospective, multicentre, double-blind, randomized, controlled trial with two parallel groups that will include a total of 640 RTRs. RTRs with vitamin D insufficiency, defined as circulating 25-hydroxyvitamin D levels of less than 30 ng/ml (or 75 nmol/l), will be randomized between 12 and 48 months after transplantation to blinded groups to receive vitamin D3 (cholecalciferol) either at high or low dose (respectively, 100,000 UI or 12,000 UI every 2 weeks for 2 months then monthly for 22 months) with a follow-up of 2 years. The primary objective of the study is to evaluate the benefit/risk ratio of high-dose versus low-dose cholecalciferol on a composite endpoint consisting of de novo diabetes mellitus; major cardiovascular events; de novo cancer; and patient death. Secondary endpoints will include blood pressure (BP) control; echocardiography findings; the incidences of infection and acute rejection episodes; renal allograft function using estimated glomerular filtration rate; proteinuria; graft survival; bone mineral density; the incidence of fractures; and biological relevant parameters of mineral metabolism. Discussion We previously reported that the intensive cholecalciferol treatment (100 000 IU every 2 weeks for 2 months) was safe in RTR. Using a pharmacokinetic approach, we showed that cholecalciferol 100,000 IU monthly should maintain serum 25-hydroxyvitamin D at above 30 ng/ml but below 80 ng/ml after renal transplantation. Taken together, these results are reassuring regarding the safety of the cholecalciferol doses that will be used in the VITALE study. Analysis of data collected during the VITALE study will demonstrate whether high or low-dose cholecalciferol is beneficial in RTRs with vitamin D insufficiency. Trial registration ClinicalTrials.gov Identifier: NCT01431430. Electronic supplementary material The online version of this article (doi:10.1186/1745-6215-15-430) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Marie Courbebaisse
- Department of Physiology, Assistance Publique-hôpitaux de Paris, Hôpital Européen Georges Pompidou, F-75015 Paris, France.
| | | | | | | | | | | | | |
Collapse
|
46
|
Tomei P, Zaza G, Granata S, Gatti D, Fraccarollo C, Gesualdo L, Boschiero L, Lupo A. Sclerostin and Dickkopf-1 in Post-menopausal Renal Allograft Recipients. Transplant Proc 2014; 46:2241-6. [DOI: 10.1016/j.transproceed.2014.07.024] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/03/2023]
|
47
|
Tsujita M, Inaguma D, Goto N, Yamamoto T, Hiramitsu T, Katayama A, Takeda A, Kobayashi T, Morozumi K, Uchida K, Narumi S, Watarai Y, Tominaga Y. Beneficial effects of preemptive kidney transplantation on calcium and phosphorus disorders in early post-transplant recipients. Clin Exp Nephrol 2014; 19:319-24. [PMID: 24706030 DOI: 10.1007/s10157-014-0967-0] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2013] [Accepted: 03/13/2014] [Indexed: 11/26/2022]
Abstract
BACKGROUND Recently, preemptive kidney transplantation (PKT) has increased in Japan; however, the effects of PKT on calcium (Ca) and phosphorus (Pi) metabolism are poorly understood. METHODS Thirty-two consecutive patients were enrolled in this study at Nagoya Daini Red Cross Hospital. Fifteen patients were in the PKT group and 17 patients were in the non-PKT group. Parameters of Ca and Pi metabolism, including fibroblast growth factor (FGF) 23 and intact parathyroid hormone, were measured before transplantation and 1, 3, and 24 weeks after transplantation. RESULTS FGF 23 decreased dramatically in both groups after transplantation; however, FGF 23 before transplantation and at 1 and 3 weeks after transplantation was significantly lower in the PKT group than in the non-PKT group (p < 0.05). Although iPTH levels were higher in the PKT group than in the non-PKT group before transplantation, these levels were lower in the PKT group at 24 weeks after transplantation (p < 0.05). Corrected Ca was lower at 24 weeks in the PKT group (p < 0.05), whereas Pi was lower in the non-PKT group at 1 and 3 weeks (p < 0.05), but not significantly different at 24 weeks. Multivariate linear regression analysis revealed that FGF 23 before transplantation was the strongest predictor of Ca and Pi disorders in early post-transplant recipients. CONCLUSIONS This study suggests that PKT has beneficial effects on Ca and Pi metabolism and pre-transplant FGF 23 levels are a good marker of post-transplant Ca and Pi metabolism disorders.
Collapse
Affiliation(s)
- Makoto Tsujita
- Department of the Kidney Disease Center, Nagoya Daini Red Cross Hospital, 2-9 Myoken-cho, Showa-ku, Nagoya, 466-8650, Japan,
| | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
48
|
[Mineral and bone disorders in renal transplantation]. Nephrol Ther 2013; 9:461-70. [PMID: 24176653 DOI: 10.1016/j.nephro.2013.07.372] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2013] [Revised: 07/23/2013] [Accepted: 07/26/2013] [Indexed: 11/22/2022]
Abstract
The deregulation of bone and mineral metabolism during chronic kidney disease (CKD) is a daily challenge for physicians, its management aiming at decreasing the risk of both fractures and vascular calcifications. Renal transplantation in the context of CKD, with pre-existing renal osteodystrophy as well as nutritional impairment, chronic inflammation, hypogonadism and corticosteroids exposure, represents a major risk factor for bone impairment in the post-transplant period. The aim of this review is therefore to provide an update on the pathophysiology of mineral and bone disorders after renal transplantation.
Collapse
|
49
|
FGF23 and mineral metabolism in the early post-renal transplantation period. Pediatr Nephrol 2013; 28:2207-15. [PMID: 23852336 PMCID: PMC3796035 DOI: 10.1007/s00467-013-2547-z] [Citation(s) in RCA: 30] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/18/2012] [Revised: 05/08/2013] [Accepted: 06/07/2013] [Indexed: 01/31/2023]
Abstract
BACKGROUND The relationship between fibroblast growth factor 23 (FGF23) and vitamin D production and catabolism post-renal transplantation has not been characterized. METHODS Circulating creatinine, calcium, phosphorus, albumin, parathyroid hormone, FGF23, and 1,25(OH)2 vitamin D (calcitriol) values were obtained pre-transplantation, daily post-operatively for 5 days, and at 6 months post-transplantation in 44 patients aged 16.4 ± 0.4 years undergoing renal transplantation at UCLA from 1 August 2005 through to 30 April 2007. 25(OH) Vitamin D and 24,25(OH)2 vitamin D concentrations were obtained at baseline and on post-operative days 5 and 180, and urinary concentrations of creatinine, phosphorus, and FGF23 were measured on post-operative days 1, 3, 5, and 180. RESULTS Circulating phosphate concentrations declined more rapidly and the fractional excretion of phosphorus was higher in the first week post-transplantation in subjects with higher FGF23 values. Fractional excretion of FGF23 was low at all time-points. Circulating 1,25(OH)2 vitamin D levels rose more rapidly and were consistently higher in patients with lower FGF23 values; however, 25(OH) vitamin D and 24,25(OH)2 vitamin D values were unrelated to FGF23 concentrations. CONCLUSIONS Inhibition of renal 1α-hydroxylase, rather than stimulation of 24-hydroxylase, may primarily contribute to the relationship between FGF23 values and calcitriol. The rapid decline in FGF23 levels post-transplantation in our patient cohort was not mediated solely by the filtration of intact FGF23 by the new kidney.
Collapse
|
50
|
Recovery versus persistence of disordered mineral metabolism in kidney transplant recipients. Semin Nephrol 2013; 33:191-203. [PMID: 23465505 DOI: 10.1016/j.semnephrol.2012.12.019] [Citation(s) in RCA: 72] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
In patients with end-stage renal disease, successful renal transplantation improves the quality of life and increases survival, as compared with long-term dialysis treatment. Although it long has been believed that successful kidney transplantation to a large extent solves the problem of chronic kidney disease-mineral and bone disorders (CKD-MBD), increasing evidence indicates that it only changes the phenotype of CKD-MBD. Posttransplant CKD-MBD reflects the effects of immunosuppression, previous CKD-MBD persisting after transplantation, and de novo CKD-MBD. A major and often-underestimated problem after successful renal transplantation is persistent hyperparathyroidism. Besides contributing to posttransplant hypercalcemia and hypophosphatemia, persistent hyperparathyroidism may be involved in the pathogenesis of allograft dysfunction (nephrocalcinosis), progression of vascular calcification, and bone disease (uncoupling of bone formation and bone resorption and bone mineral density loss) in renal transplant recipients. Similar to nontransplanted patients, CKD-MBD has a detrimental impact on (cardiovascular) mortality and morbidity. Additional studies urgently are needed to get more insights into the pathophysiology of posttransplant CKD-MBD. These new insights will allow for a more targeted and causal therapeutic approach.
Collapse
|