1
|
Javier AJS, Kennedy FM, Yi X, Wehling-Henricks M, Tidball JG, White KE, Witczak CA, Kuro-O M, Welc SS. Klotho Is Cardioprotective in the mdx Mouse Model of Duchenne Muscular Dystrophy. THE AMERICAN JOURNAL OF PATHOLOGY 2025; 195:923-940. [PMID: 39889824 PMCID: PMC12016860 DOI: 10.1016/j.ajpath.2024.12.017] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/11/2024] [Revised: 12/18/2024] [Accepted: 12/30/2024] [Indexed: 02/03/2025]
Abstract
Duchenne muscular dystrophy (DMD) is a lethal, progressive skeletal and cardiac myopathy. Cardiomyopathy is the leading cause of death in patients with DMD, but the molecular basis for heart failure is incompletely understood. As with humans, in the mdx mouse model of DMD, cardiac function is impaired after the onset of skeletal muscle pathology. Dysregulation of Klotho gene regulation in dystrophic skeletal muscles occurs at disease onset, affecting pathogenesis. Whether Klotho is protective against dystrophin-deficient cardiomyopathy is unknown. This study found that expression of a Klotho transgene prevented deficits in left ventricular ejection fraction and fractional shortening in mdx mice. Improvements in cardiac performance were associated with reductions in adverse cardiac remodeling, cardiac myocyte hypertrophy, and fibrosis. In addition, mdx mice expressed high concentrations of plasma fibroblast growth factor 23 (FGF23), and expression was increased locally in hearts. The cardioprotective effects of Klotho were not associated with differences in renal function or serum biochemistries, but transgene expression prevented increased expression of plasma FGF23 and cardiac Fgf23 mRNA expression. Cardiac reactive oxygen species, oxidative damage, mitochondrial damage, and apoptosis were reduced in transgenic hearts. FGF23 stimulated hypertrophic growth in dystrophic neonatal mouse ventricular myocytes in vitro, which was inhibited by co-stimulation with soluble Klotho. Taken together, these results show that Klotho prevented dystrophic cardiac remodeling and improved function.
Collapse
MESH Headings
- Animals
- Klotho Proteins
- Muscular Dystrophy, Duchenne/metabolism
- Muscular Dystrophy, Duchenne/physiopathology
- Muscular Dystrophy, Duchenne/pathology
- Muscular Dystrophy, Duchenne/complications
- Muscular Dystrophy, Duchenne/genetics
- Glucuronidase/metabolism
- Glucuronidase/genetics
- Fibroblast Growth Factor-23
- Fibroblast Growth Factors/metabolism
- Fibroblast Growth Factors/blood
- Fibroblast Growth Factors/genetics
- Mice, Inbred mdx
- Disease Models, Animal
- Mice
- Cardiomyopathies/pathology
- Cardiomyopathies/metabolism
- Myocytes, Cardiac/metabolism
- Myocytes, Cardiac/pathology
- Mice, Transgenic
- Mice, Inbred C57BL
- Male
- Apoptosis
- Reactive Oxygen Species/metabolism
- Fibrosis
- Humans
- Myocardium/pathology
- Myocardium/metabolism
Collapse
Affiliation(s)
- Areli Jannes S Javier
- Indiana Center for Musculoskeletal Health, Indiana University School of Medicine, Indianapolis, Indiana
| | - Felicia M Kennedy
- Department of Anatomy, Cell Biology & Physiology, Indiana University School of Medicine, Indianapolis, Indiana
| | - Xin Yi
- Department of Anatomy, Cell Biology & Physiology, Indiana University School of Medicine, Indianapolis, Indiana
| | | | - James G Tidball
- Department of Integrative Biology and Physiology, University of California, Los Angeles, California; Department of Pathology and Laboratory Medicine, University of California, Los Angeles, California
| | - Kenneth E White
- Department of Medical and Molecular Genetics, Indiana University School of Medicine, Indianapolis, Indiana
| | - Carol A Witczak
- Indiana Center for Musculoskeletal Health, Indiana University School of Medicine, Indianapolis, Indiana; Department of Anatomy, Cell Biology & Physiology, Indiana University School of Medicine, Indianapolis, Indiana
| | - Makoto Kuro-O
- Division of Anti-Aging Medicine, Center for Molecular Medicine, Jichi Medical University, Shimotsuke, Japan
| | - Steven S Welc
- Indiana Center for Musculoskeletal Health, Indiana University School of Medicine, Indianapolis, Indiana; Department of Anatomy, Cell Biology & Physiology, Indiana University School of Medicine, Indianapolis, Indiana.
| |
Collapse
|
2
|
Carullo N, Sorbo D, Faga T, Pugliese S, Zicarelli MT, Costa D, Ielapi N, Battaglia Y, Pisani A, Coppolino G, Bolignano D, Michael A, Serra R, Andreucci M. Anemia and Mineral Bone Disorder in Kidney Disease Patients: The Role of FGF-23 and Other Related Factors. Int J Mol Sci 2024; 25:12838. [PMID: 39684548 DOI: 10.3390/ijms252312838] [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: 10/27/2024] [Revised: 11/20/2024] [Accepted: 11/26/2024] [Indexed: 12/18/2024] Open
Abstract
Anemia and mineral and bone disorder (MBD) are significant complications of chronic kidney disease (CKD). The erythropoietin (Epo) pathway plays a key role in both of these processes in CKD. Another molecule that plays an important role in CKD-MBD is fibroblast growth factor (FGF)-23, whose main role is to maintain serum phosphate levels in the normal range, acting via its co-receptor Klotho; however, its activity may also be related to anemia and inflammation. In this review, the regulation of Epo and FGF-23 and the molecular mechanisms of their action are outlined. Furthermore, the complex interaction between EPO and FGF-23 is discussed, as well as their association with other anemia-related factors and processes such as Klotho, vitamin D, and iron deficiency. Together, these may be part of a "kidney-bone marrow-bone axis" that promotes CKD-MBD.
Collapse
Affiliation(s)
- Nazareno Carullo
- "G. Jazzolino" Hospital, A.S.P. Vibo Valentia, I89900 Vibo Valentia, Italy
| | - David Sorbo
- San Bortolo Hospital, ULSS 8 Berica, I36100 Vicenza, Italy
| | - Teresa Faga
- Department of Health Sciences, "Magna Graecia" University, I88100 Catanzaro, Italy
| | - Sara Pugliese
- Department of Health Sciences, "Magna Graecia" University, I88100 Catanzaro, Italy
| | - Maria Teresa Zicarelli
- Amantea Outpatient Clinic, A.S.P. Cosenza, I87032 Amantea, Italy
- Department of Medical and Surgical Sciences, "Magna Graecia" University, I88100 Catanzaro, Italy
| | - Davide Costa
- Department of Medical and Surgical Sciences, "Magna Graecia" University, I88100 Catanzaro, Italy
- Interuniversity Center of Phlebolymphology (CIFL), "Magna Graecia" University, I88100 Catanzaro, Italy
| | - Nicola Ielapi
- Interuniversity Center of Phlebolymphology (CIFL), "Magna Graecia" University, I88100 Catanzaro, Italy
- Department of Public Health and Infectious Disease, "Sapienza" University of Rome, I00185 Rome, Italy
| | - Yuri Battaglia
- Department of Medicine, University of Verona, I37129 Verona, Italy
| | - Antonio Pisani
- Department of Public Health, University of Naples Federico II, I80131 Naples, Italy
| | - Giuseppe Coppolino
- Department of Health Sciences, "Magna Graecia" University, I88100 Catanzaro, Italy
| | - Davide Bolignano
- Department of Medical and Surgical Sciences, "Magna Graecia" University, I88100 Catanzaro, Italy
| | - Ashour Michael
- Department of Health Sciences, "Magna Graecia" University, I88100 Catanzaro, Italy
| | - Raffaele Serra
- Department of Medical and Surgical Sciences, "Magna Graecia" University, I88100 Catanzaro, Italy
- Interuniversity Center of Phlebolymphology (CIFL), "Magna Graecia" University, I88100 Catanzaro, Italy
| | - Michele Andreucci
- Department of Health Sciences, "Magna Graecia" University, I88100 Catanzaro, Italy
| |
Collapse
|
3
|
Ito K, Akizawa T, Arita K, Mitobe Y, Komatsu N. Effect of ferric citrate hydrate on fibroblast growth factor 23 and platelets in non-dialysis-dependent chronic kidney disease and non-chronic kidney disease patients with iron deficiency anemia. Clin Exp Nephrol 2024; 28:636-646. [PMID: 38402503 PMCID: PMC11189996 DOI: 10.1007/s10157-023-02455-6] [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: 10/04/2023] [Accepted: 12/27/2023] [Indexed: 02/26/2024]
Abstract
BACKGROUND Iron deficiency anemia (IDA) increases levels of C-terminal fibroblast growth factor 23 (cFGF23) and platelet count (PLT), each of which is associated with cardiovascular events. Therefore, we hypothesized that iron replacement with ferric citrate hydrate (FC) would decrease cFGF23 levels and PLT in patients with IDA. METHODS In a randomized, open-label, multicenter, 24-week clinical trial, patients with non-dialysis-dependent chronic kidney disease (CKD) and non-CKD complicated by IDA (8.0 ≤ hemoglobin < 11.0 g/dL; and serum ferritin < 50 ng/mL [CKD]; < 12 ng/mL [non-CKD]) were randomized 1:1 to FC-low (500 mg: approximately 120 mg elemental iron/day) or FC-high (1000 mg: approximately 240 mg elemental iron/day). If sufficient iron replacement had been achieved after week 8, further treatment was discontinued. RESULTS Seventy-three patients were allocated to FC-low (CKD n = 21, non-CKD n = 15) and FC-high (CKD n = 21, non-CKD n = 16). Regardless of CKD status, FC increased serum ferritin and transferrin saturation, did not change intact FGF23 or serum phosphorus, but decreased cFGF23. In FC-low group, median changes in cFGF23 from baseline to week 8 were -58.00 RU/mL in CKD and -725.00 RU/mL in non-CKD; in FC-high group, the median changes were -66.00 RU/mL in CKD and -649.50 RU/mL in non-CKD. By week 8, FC treatment normalized PLT in all patients with high PLT at baseline (>35.2 × 104/µL; FC-low: 1 CKD, 8 non-CKD; FC-high: 3 CKD, 8 non-CKD). CONCLUSION Regardless of CKD status, iron replacement with FC decreased elevated cFGF23 levels and normalized elevated PLT in patients with IDA. CLINICAL TRIAL REGISTRATION NUMBER jRCT2080223943.
Collapse
Affiliation(s)
- Kyoko Ito
- Medical Affairs Department, Torii Pharmaceutical Co., Ltd., 3-4-1, Nihonbashi-Honcho, Chuo-Ku, Tokyo, 103-8439, Japan
- Doctoral Program in Life Science Innovation (Disease Mechanism), Degree Programs in Comprehensive Human Sciences, Graduate School of Comprehensive Human Sciences, University of Tsukuba, 1-1-1 Tennodai, Tsukuba, Ibaraki, 305-8577, Japan
| | - Tadao Akizawa
- Division of Nephrology, Department of Medicine, Showa University School of Medicine, Namics Shinagawa 301, 4-24-51 Takanawa, Minato-Ku, Tokyo, 108-0074, Japan.
| | - Kojo Arita
- Clinical Development Department, Pharmaceutical Division, Japan Tobacco Inc., 3-4-1, Nihonbashi-Honcho, Chuo-Ku, Tokyo, 103-0023, Japan
| | - Yuko Mitobe
- Medical Affairs Department, Torii Pharmaceutical Co., Ltd., 3-4-1, Nihonbashi-Honcho, Chuo-Ku, Tokyo, 103-8439, Japan
| | - Norio Komatsu
- Department of Hematology, Juntendo University School of Medicine, 2-1-1, Hongo, Bunkyo-Ku, Tokyo, 113-8421, Japan
| |
Collapse
|
4
|
Hamid AK, Pastor Arroyo EM, Calvet C, Hewitson TD, Muscalu ML, Schnitzbauer U, Smith ER, Wagner CA, Egli-Spichtig D. Phosphate Restriction Prevents Metabolic Acidosis and Curbs Rise in FGF23 and Mortality in Murine Folic Acid-Induced AKI. J Am Soc Nephrol 2024; 35:261-280. [PMID: 38189228 PMCID: PMC10914210 DOI: 10.1681/asn.0000000000000291] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2023] [Accepted: 12/02/2023] [Indexed: 01/09/2024] Open
Abstract
SIGNIFICANCE STATEMENT Patients with AKI suffer a staggering mortality rate of approximately 30%. Fibroblast growth factor 23 (FGF23) and phosphate (P i ) rise rapidly after the onset of AKI and have both been independently associated with ensuing morbidity and mortality. This study demonstrates that dietary P i restriction markedly diminished the early rise in plasma FGF23 and prevented the rise in plasma P i , parathyroid hormone, and calcitriol in mice with folic acid-induced AKI (FA-AKI). Furthermore, the study provides evidence for P i -sensitive osseous Fgf23 mRNA expression and reveals that P i restriction mitigated calciprotein particles (CPPs) formation, inflammation, acidosis, cardiac electrical disturbances, and mortality in mice with FA-AKI. These findings suggest that P i restriction may have a prophylactic potential in patients at risk for AKI. BACKGROUND In AKI, plasma FGF23 and P i rise rapidly and are independently associated with disease severity and outcome. METHODS The effects of normal (NP) and low (LP) dietary P i were investigated in mice with FA-AKI after 3, 24, and 48 hours and 14 days. RESULTS After 24 hours of AKI, the LP diet curbed the rise in plasma FGF23 and prevented that of parathyroid hormone and calcitriol as well as of osseous but not splenic or thymic Fgf23 mRNA expression. The absence of Pth prevented the rise in calcitriol and reduced the elevation of FGF23 in FA-AKI with the NP diet. Furthermore, the LP diet attenuated the rise in renal and plasma IL-6 and mitigated the decline in renal α -Klotho. After 48 hours, the LP diet further dampened renal IL-6 expression and resulted in lower urinary neutrophil gelatinase-associated lipocalin. In addition, the LP diet prevented the increased formation of CPPs. Fourteen days after AKI induction, the LP diet group maintained less elevated plasma FGF23 levels and had greater survival than the NP diet group. This was associated with prevention of metabolic acidosis, hypocalcemia, hyperkalemia, and cardiac electrical disturbances. CONCLUSIONS This study reveals P i -sensitive FGF23 expression in the bone but not in the thymus or spleen in FA-AKI and demonstrates that P i restriction mitigates CPP formation, inflammation, acidosis, and mortality in this model. These results suggest that dietary P i restriction could have prophylactic potential in patients at risk for AKI.
Collapse
Affiliation(s)
- Ahmad Kamal Hamid
- Institute of Physiology, University of Zurich, Zurich, Switzerland
- Swiss National Centre of Competence in Research (NCCR) Kidney.CH, Zurich, Switzerland
| | - Eva Maria Pastor Arroyo
- Institute of Physiology, University of Zurich, Zurich, Switzerland
- Swiss National Centre of Competence in Research (NCCR) Kidney.CH, Zurich, Switzerland
| | - Charlotte Calvet
- Institute of Physiology, University of Zurich, Zurich, Switzerland
- Zurich Integrative Rodent Physiology (ZIRP), University of Zurich, Zurich, Switzerland
| | - Timothy D. Hewitson
- Department of Nephrology, The Royal Melbourne Hospital, Melbourne, Australia
- Department of Medicine, University of Melbourne, Melbourne Australia
| | - Maria Lavinia Muscalu
- Institute of Physiology, University of Zurich, Zurich, Switzerland
- Swiss National Centre of Competence in Research (NCCR) Kidney.CH, Zurich, Switzerland
| | - Udo Schnitzbauer
- Institute of Physiology, University of Zurich, Zurich, Switzerland
| | - Edward R. Smith
- Department of Nephrology, The Royal Melbourne Hospital, Melbourne, Australia
- Department of Medicine, University of Melbourne, Melbourne Australia
| | - Carsten Alexander Wagner
- Institute of Physiology, University of Zurich, Zurich, Switzerland
- Swiss National Centre of Competence in Research (NCCR) Kidney.CH, Zurich, Switzerland
| | - Daniela Egli-Spichtig
- Institute of Physiology, University of Zurich, Zurich, Switzerland
- Swiss National Centre of Competence in Research (NCCR) Kidney.CH, Zurich, Switzerland
| |
Collapse
|
5
|
Edmonston D, Grabner A, Wolf M. FGF23 and klotho at the intersection of kidney and cardiovascular disease. Nat Rev Cardiol 2024; 21:11-24. [PMID: 37443358 DOI: 10.1038/s41569-023-00903-0] [Citation(s) in RCA: 47] [Impact Index Per Article: 47.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 06/13/2023] [Indexed: 07/15/2023]
Abstract
Cardiovascular disease is the leading cause of death in patients with chronic kidney disease (CKD). As CKD progresses, CKD-specific risk factors, such as disordered mineral homeostasis, amplify traditional cardiovascular risk factors. Fibroblast growth factor 23 (FGF23) regulates mineral homeostasis by activating complexes of FGF receptors and transmembrane klotho co-receptors. A soluble form of klotho also acts as a 'portable' FGF23 co-receptor in tissues that do not express klotho. In progressive CKD, rising circulating FGF23 levels in combination with decreasing kidney expression of klotho results in klotho-independent effects of FGF23 on the heart that promote left ventricular hypertrophy, heart failure, atrial fibrillation and death. Emerging data suggest that soluble klotho might mitigate some of these effects via several candidate mechanisms. More research is needed to investigate FGF23 excess and klotho deficiency in specific cardiovascular complications of CKD, but the pathophysiological primacy of FGF23 excess versus klotho deficiency might never be precisely resolved, given the entangled feedback loops that they share. Therefore, randomized trials should prioritize clinical practicality over scientific certainty by targeting disordered mineral homeostasis holistically in an effort to improve cardiovascular outcomes in patients with CKD.
Collapse
Affiliation(s)
- Daniel Edmonston
- Division of Nephrology, Department of Medicine, Duke University School of Medicine, Durham, NC, USA
- Duke Clinical Research Institute, Duke University School of Medicine, Durham, NC, USA
| | - Alexander Grabner
- Division of Nephrology, Department of Medicine, Duke University School of Medicine, Durham, NC, USA
| | - Myles Wolf
- Division of Nephrology, Department of Medicine, Duke University School of Medicine, Durham, NC, USA.
- Duke Clinical Research Institute, Duke University School of Medicine, Durham, NC, USA.
| |
Collapse
|
6
|
Leidner AS, Cai X, Zelnick LR, Lee J, Bansal N, Pasch A, Kansal M, Chen J, Anderson AH, Sondheimer JH, Lash JP, Townsend RR, Go AS, Feldman HI, Shah SJ, Wolf M, Isakova T, Mehta RC. Fibroblast Growth Factor 23 and Risk of Heart Failure Subtype: The CRIC (Chronic Renal Insufficiency Cohort) Study. Kidney Med 2023; 5:100723. [PMID: 37915961 PMCID: PMC10616385 DOI: 10.1016/j.xkme.2023.100723] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/03/2023] Open
Abstract
Rationale & Objective Heart failure (HF) is an important cause of morbidity and mortality among individuals with chronic kidney disease (CKD). A large body of evidence from preclinical and clinical studies implicates excess levels of fibroblast growth factor 23 (FGF23) in HF pathogenesis in CKD. It remains unclear whether the relationship between elevated FGF23 levels and HF risk among individuals with CKD varies by HF subtype. Study Design Prospective cohort study. Settings & Participants A total of 3,502 participants were selected in the Chronic Renal Insufficiency Cohort study. Exposure Baseline plasma FGF23. Outcomes Incident HF by subtype and total rate of HF hospitalization. HF was categorized as HF with preserved ejection fraction (HFpEF, ejection fraction [EF] ≥ 50%), HF with reduced EF (HFrEF, EF < 50%) and HF with unknown EF (HFuEF). Analytical Approach Multivariable-adjusted cause-specific Cox proportional hazards models were used to investigate associations between FGF23 and incident hospitalizations for HF by subtype. The Lunn-McNeil method was used to compare hazard ratios across HF subtypes. Poisson regression models were used to evaluate the total rate of HF. Results During a median follow-up time of 10.8 years, 295 HFpEF, 242 HFrEF, and 156 HFuEF hospitalizations occurred. In multivariable-adjusted cause-specific Cox proportional hazards models, FGF23 was significantly associated with the incidence of HFpEF (HR, 1.41; 95% CI, 1.21-1.64), HFrEF (HR, 1.27; 95% CI, 1.05-1.53), and HFuEF (HR, 1.40; 95% CI, 1.13-1.73) per 1 standard deviation (SD) increase in the natural log of FGF23. The Lunn-McNeil method determined that the risk association was consistent across all subtypes. The rate ratio of total HF events increased with FGF23 quartile. In multivariable-adjusted models, compared with quartile 1, FGF23 quartile 4 had a rate ratio of 1.81 (95% CI, 1.28-2.57) for total HF events. Limitations Self-report of HF hospitalizations and possible lack of an echocardiogram at time of hospitalization. Conclusions In this large multicenter prospective cohort study, elevated FGF23 levels were associated with increased risks for all HF subtypes. Plain-Language Summary Heart failure (HF) is a prominent cause of morbidity and mortality in individuals with chronic kidney disease (CKD). Identifying potential pathways in the development of HF is essential in developing therapies to prevent and treat HF. In a large cohort of individuals with CKD, the Chronic Renal Insufficiency Cohort (N = 3,502), baseline fibroblast growth factor-23 (FGF23), a hormone that regulates phosphorous, was evaluated in relation to the development of incident and recurrent HF with reduced, preserved, and unknown ejection fraction. In this large multicenter prospective cohort study, elevated FGF23 levels were associated with increased risk of all HF subtypes. These findings demonstrate the need for further research into FGF23 as a target in preventing the development of HF in individuals with CKD.
Collapse
Affiliation(s)
| | - Xuan Cai
- Northwestern University Feinberg School of Medicine, Chicago, IL
| | | | - Jungwha Lee
- Northwestern University Feinberg School of Medicine, Chicago, IL
| | | | | | | | | | | | | | | | | | | | | | - Sanjiv J. Shah
- Northwestern University Feinberg School of Medicine, Chicago, IL
| | - Myles Wolf
- Duke University School of Medicine, Durham, North Carolina
| | - Tamara Isakova
- Northwestern University Feinberg School of Medicine, Chicago, IL
| | - Rupal C. Mehta
- Northwestern University Feinberg School of Medicine, Chicago, IL
| |
Collapse
|
7
|
Egli-Spichtig D, Hamid AK, Arroyo EMP, Ketteler M, Wiecek A, Rosenkranz AR, Pasch A, Lorenz H, Hellmann B, Karus M, Ammer R, Rubio-Aliaga I, Wagner CA. Intact FGF23 predicts serum phosphate improvement after combined nicotinamide and phosphate binder treatment in hemodialysis patients. Clin Kidney J 2023; 16:1622-1633. [PMID: 37779856 PMCID: PMC10539220 DOI: 10.1093/ckj/sfad040] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2022] [Indexed: 10/03/2023] Open
Abstract
Background Hyperphosphatemia is associated with increased mortality and cardiovascular morbidity of end-stage kidney failure (ESKF) patients. Managing serum phosphate in ESKF patients is challenging and mostly based on limiting intestinal phosphate absorption with low phosphate diets and phosphate binders (PB). In a multi-centric, double-blinded, placebo-controlled study cohort of maintenance hemodialysis patients with hyperphosphatemia, we demonstrated the efficacy of nicotinamide modified release (NAMR) formulation treatment in addition to standard PB therapy in decreasing serum phosphate. Here we aimed to assess the relationship between phosphate, FGF23, inflammation and iron metabolism in this cohort. Methods We measured the plasma concentrations of intact fibroblast growth factor 23 (iFGF23) and selected proinflammatory cytokines at baseline and Week 12 after initiating treatment. Results We observed a strong correlation between iFGF23 and cFGF23 (C-terminal fragment plus iFGF23). We identified iFGF23 as a better predictor of changes in serum phosphate induced by NAMR and PB treatment compared with cFGF23. Recursive partitioning revealed at baseline and Week 12, that iFGF23 and cFGF23 together with T50 propensity were the most important predictors of serum phosphate, whereas intact parathyroid hormone (iPTH) played a minor role in this model. Furthermore, we found serum phosphate and iPTH as the best predictors of iFGF23 and cFGF23. Sex, age, body mass index, and markers of inflammation and iron metabolism had only a minor impact in predicting FGF23. Conclusion Lowering serum phosphate in ESKF patients may depend highly on iFGF23 which is correlated to cFGF23 levels. Serum phosphate was the most important predictor of plasma FGF23 in this ESKF cohort.
Collapse
Affiliation(s)
- Daniela Egli-Spichtig
- Institute of Physiology, University of Zurich, 8057 Zurich, Switzerland, and National Center of Competence in Research NCCR Kidney.CH
| | - Ahmad Kamal Hamid
- Institute of Physiology, University of Zurich, 8057 Zurich, Switzerland, and National Center of Competence in Research NCCR Kidney.CH
| | - Eva Maria Pastor Arroyo
- Institute of Physiology, University of Zurich, 8057 Zurich, Switzerland, and National Center of Competence in Research NCCR Kidney.CH
| | - Markus Ketteler
- Robert Bosch Hospital, Department of General Internal Medicine and Nephrology, Stuttgart, Germany
| | - Andrzej Wiecek
- Department of Nephrology, Transplantation and Internal Medicine, Medical University of Silesia, Katowice, Poland
| | - Alexander R Rosenkranz
- Division of Nephrology, Department of Internal Medicine, Medical University of Graz, Graz, Austria
| | - Andreas Pasch
- Calciscon AG, 2503 Biel, Switzerland
- Department of Nephrology, Lindenhofspital, 3012 Bern, Switzerland
- Department of Physiology and Pathophysiology, Johannes Kepler University Linz, Linz, Austria
| | - Horst Lorenz
- Buero fuer Biometrie und Statistik, Neuberg, Germany
| | | | - Michael Karus
- MEDICE Arzneimittel Pütter GmbH & Co KG, Iserlohn, Germany
| | - Richard Ammer
- MEDICE Arzneimittel Pütter GmbH & Co KG, Iserlohn, Germany
- Department of Medicine D, Division of General Internal Medicine, Nephrology and Rheumatology, University Hospital of Münster, Münster, Germany
| | - Isabel Rubio-Aliaga
- Institute of Physiology, University of Zurich, 8057 Zurich, Switzerland, and National Center of Competence in Research NCCR Kidney.CH
| | - Carsten A Wagner
- Institute of Physiology, University of Zurich, 8057 Zurich, Switzerland, and National Center of Competence in Research NCCR Kidney.CH
| |
Collapse
|
8
|
Kim KJ, Song JE, Kim JH, Hong N, Kim SG, Lee J, Rhee Y. Elevated morbidity and mortality in patients with chronic idiopathic hypophosphatemia: a nationwide cohort study. Front Endocrinol (Lausanne) 2023; 14:1229750. [PMID: 37635983 PMCID: PMC10448510 DOI: 10.3389/fendo.2023.1229750] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/26/2023] [Accepted: 07/26/2023] [Indexed: 08/29/2023] Open
Abstract
Background Chronic idiopathic hypophosphatemia (CIH) induced by X-linked hypophosphatemic rickets or tumor-induced osteomalacia is a rare inherited or acquired disorder. However, due to its rarity, little is known about the epidemiology and natural course of CIH. Therefore, we aimed to identify the prevalence and long-term health outcomes of CIH patients. Methods Using the Korean Health Insurance Review and Assessment claims database, we evaluated the incidence of hypophosphatemia initially diagnosed from 2003 to 2018. After excluding secondary conditions that could change serum phosphorus levels, we identified 154 patients (76 men and 78 women) with non-secondary and non-renal hypophosphatemia. These hypophosphatemic patients were compared at a ratio of 1:10 with age-, sex-, and index-year-matched controls (n = 1,540). Results In the distribution of age at diagnosis, a large peak was observed in patients aged 1-4 years and small peaks were observed in ages from 40-70 years. The age-standardized incidence rate showed non-statistically significant trend from 0.24 per 1,000,000 persons in 2003 to 0.30 in 2018. Hypophosphatemic patients had a higher risk of any complication (adjusted hazard ratio [aHR], 2.17; 95% confidence interval [CI], 1.67-2.69) including cardiovascular outcomes, chronic kidney disease, hyperparathyroidism, osteoporotic fractures, periodontitis, and depression. Hypophosphatemic patients also had higher risks of mortality and hospitalization than the controls (aHR, 3.26; 95% CI, 1.83-5.81; and aHR, 2.49; 95% CI, 1.97-3.16, respectively). Conclusion This first nationwide study of CIH in South Korea found a bimodal age distribution and no sex differences among patients. Hypophosphatemic patients had higher risks of complications, mortality, and hospitalization compared to age- and sex-matched controls.
Collapse
Affiliation(s)
- Kyoung Jin Kim
- Division of Endocrinology and Metabolism, Department of Internal Medicine, Korea University College of Medicine, Seoul, Republic of Korea
| | - Ji Eun Song
- Department of Biostatistics, Korea University College of Medicine, Seoul, Republic of Korea
| | - Ji Hyun Kim
- Department of Biostatistics, Korea University College of Medicine, Seoul, Republic of Korea
| | - Namki Hong
- Department of Internal Medicine, Endocrine Research Institute, Yonsei University College of Medicine, Seoul, Republic of Korea
| | - Sin Gon Kim
- Division of Endocrinology and Metabolism, Department of Internal Medicine, Korea University College of Medicine, Seoul, Republic of Korea
| | - Juneyoung Lee
- Department of Biostatistics, Korea University College of Medicine, Seoul, Republic of Korea
| | - Yumie Rhee
- Department of Internal Medicine, Endocrine Research Institute, Yonsei University College of Medicine, Seoul, Republic of Korea
| |
Collapse
|
9
|
Schmitz T, Wein B, Heier M, Peters A, Meisinger C, Linseisen J. Baseline fibroblast growth factor 23 is associated with long-term mortality in ST-elevation myocardial infarction-results from the augsburg myocardial infarction registry. Front Cardiovasc Med 2023; 10:1173281. [PMID: 37600039 PMCID: PMC10436601 DOI: 10.3389/fcvm.2023.1173281] [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: 02/24/2023] [Accepted: 07/21/2023] [Indexed: 08/22/2023] Open
Abstract
Background The aim of this study was to investigate the association between inflammatory plasma protein concentrations and long-term mortality in patients with ST-elevation myocardial infarction (STEMI). Methods For 343 STEMI patients recorded between 2009 and 2013 by the population-based Myocardial Infarction Registry Augsburg, 92 inflammatory plasma proteins were measured at the index event using the OLINK inflammation panel. In multivariable-adjusted Cox regression models, the association between each plasma protein and all-cause long-term mortality was investigated. Median follow-up time was 7.6 (IQR: 2.4) years. For plasma protein that showed a strong association with long-term mortality, a 5-year survival ROC analysis was performed. Results One plasma protein, namely Fibroblast Growth Factor 23 (FGF-23), was particularly well associated with long-term mortality in the multivariable-adjusted Cox model with an FDR-adjusted p-value of <0.001 and a Hazard Ratio (HR) of 1.57 [95% CI: 1.29-1.91]. In the 5-years ROC analysis, an AUC of 0.6903 [95% CI: 0.594-0.781] was estimated for FGF-23. All other plasma protein didńt show strong associations, each marker with FDR-adjusted p-values >0.05 in the multivariable-adjusted Cox models. Conclusions FGF-23 is independently associated with long-term mortality after STEMI and might play an important role in the response to myocardial injury. The results suggest FGF-23 to be a useful marker in the long-term treatment of STEMI patients and a potential target for drug development.
Collapse
Affiliation(s)
- Timo Schmitz
- Epidemiology, Medical Faculty, University of Augsburg, Augsburg, Germany
| | - Bastian Wein
- Department of Cardiology, Respiratory Medicine and Intensive Care, University Hospital Augsburg, Augsburg, Germany
| | - Margit Heier
- KORA Study Centre, University Hospital of Augsburg,Augsburg, Germany
- Helmholtz Zentrum München, Institute for Epidemiology, Neuherberg, Germany
| | - Annette Peters
- Helmholtz Zentrum München, Institute for Epidemiology, Neuherberg, Germany
- Chair of Epidemiology, Institute for Medical Information Processing, Biometry and Epidemiology, Medical Faculty, Ludwig-Maximilians-Universität München, Munich, Germany
- German Center for Diabetes Research (DZD), Neuherberg, Germany
- German Research Center for Cardiovascular Research (DZHK), Partner Site Munich Heart Alliance, Munich, Germany
| | - Christa Meisinger
- Epidemiology, Medical Faculty, University of Augsburg, Augsburg, Germany
| | - Jakob Linseisen
- Epidemiology, Medical Faculty, University of Augsburg, Augsburg, Germany
| |
Collapse
|
10
|
Clinkenbeard E. Fibroblast Growth Factor 23 Bone Regulation and Downstream Hormonal Activity. Calcif Tissue Int 2023; 113:4-20. [PMID: 37306735 DOI: 10.1007/s00223-023-01092-1] [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] [Received: 03/05/2023] [Accepted: 05/01/2023] [Indexed: 06/13/2023]
Abstract
Mineral homeostasis of calcium and phosphate levels is one critical component to the maintenance of bone mineral density (BMD) and strength. Diseases that disrupt calcium and phosphate balanced have highlighted not only the role these minerals play in overall bone homeostasis, but also the factors, hormones and downstream transporters, responsible for mineral metabolism. The key phosphaturic hormone elucidated from studying rare heritable disorders of hypophosphatemia is Fibroblast Growth Factor 23 (FGF23). FGF23 is predominantly secreted from bone cells in an effort to maintain phosphate balance by directly controlling renal reabsorption and indirectly affecting intestinal uptake of this mineral. Multiple factors have been shown to enhance bone mRNA expression; however, FGF23 can also undergo proteolytic cleavage to control secretion of the biologically active form of the hormone. The review focuses specifically on the regulation of FGF23 and its secretion from bone as well as its hormonal actions under physiological and disease conditions.
Collapse
Affiliation(s)
- Erica Clinkenbeard
- Department of Medical and Molecular Genetics, School of Medicine, Indiana University, 635 Barnhill Drive MS 5023, Indianapolis, IN, 46202, USA.
| |
Collapse
|
11
|
Nakano T, Kishimoto H, Tokumoto M. Direct and indirect effects of fibroblast growth factor 23 on the heart. Front Endocrinol (Lausanne) 2023; 14:1059179. [PMID: 36909314 PMCID: PMC9999118 DOI: 10.3389/fendo.2023.1059179] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/01/2022] [Accepted: 01/30/2023] [Indexed: 03/14/2023] Open
Abstract
Fibroblast growth factor (FGF)23 is a bone-derived phosphotropic hormone that regulates phosphate and mineral homeostasis. Recent studies have provided evidence that a high plasma concentration of FGF23 is associated with cardiac disease, including left ventricular hypertrophy (LVH), heart failure, atrial fibrillation, and cardiac death. Experimental studies have shown that FGF23 activates fibroblast growth factor receptor 4 (FGFR4)/phospholipase Cγ/calcineurin/nuclear factor of activated T-cells signaling in cardiomyocytes and induces cardiac hypertrophy in rodents. Activation of FGFR4 by FGF23 normally requires the co-receptor α-klotho, and klotho-independent signaling occurs only under conditions characterized by extremely high FGF23 concentrations. Recent studies have demonstrated that FGF23 activates the renin-angiotensin-aldosterone system (RAAS) and induces LVH, at least in part as a result of lower vitamin D activation. Moreover, crosstalk between FGF23 and RAAS results in the induction of cardiac hypertrophy and fibrosis. In this review, we summarize the results of studies regarding the relationships between FGF23 and cardiac events, and describe the potential direct and indirect mechanisms whereby FGF23 induces LVH.
Collapse
Affiliation(s)
- Toshiaki Nakano
- Center for Cohort Studies, Graduate School of Medical Sciences, Kyushu University, Fukuoka, Japan
- Department of Medicine and Clinical Science, Graduate School of Medical Sciences, Kyushu University, Fukuoka, Japan
- *Correspondence: Toshiaki Nakano,
| | - Hiroshi Kishimoto
- Department of Medicine and Clinical Science, Graduate School of Medical Sciences, Kyushu University, Fukuoka, Japan
| | - Masanori Tokumoto
- Department of Nephrology, Fukuoka Red Cross Hospital, Fukuoka, Japan
| |
Collapse
|
12
|
Liu M, Xia P, Tan Z, Song T, Mei K, Wang J, Ma J, Jiang Y, Zhang J, Zhao Y, Yu P, Liu X. Fibroblast growth factor-23 and the risk of cardiovascular diseases and mortality in the general population: A systematic review and dose-response meta-analysis. Front Cardiovasc Med 2022; 9:989574. [PMID: 36407457 PMCID: PMC9669381 DOI: 10.3389/fcvm.2022.989574] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2022] [Accepted: 10/06/2022] [Indexed: 11/12/2023] Open
Abstract
BACKGROUND In the past decade, fibroblast growth factor 23 (FGF23) has been recognized as an important biomarker of cardiovascular diseases. This study aimed to assess the relationship between FGF23 and the risk of cardiovascular diseases (CVDs) in general populations. METHODS The protocol was registered prospectively in PROSPERO (CRD42021281837) and two authors independently searched for relevant studies in the PubMed, EMBASE, and Cochrane Library databases. The random effects model was applied. RESULTS In total, 29 prospective studies involving 135,576 participants were included. In the general population, the category analysis revealed that elevated FGF23 levels were related to increased risks of myocardial infarction (MI) (RR: 1.40, 95%CI: 1.03-1.89), stroke (RR: 1.20, 95%CI: 1.02-1.43), heart failure (HF) (RR: 1.37, 95%CI: 1.23-1.52), CVD events (RR: 1.22, 95%CI: 0.99-1.51), cardiovascular mortality (RR: 1.46, 95%CI: 1.29-1.65), and all-cause mortality (RR: 1.50, 95%CI: 1.29-1.74). In the continuous analysis, per doubling of FGF23 was associated with increased risks of MI (RR: 1.08, 95%CI: 0.94-1.25), stroke (RR: 1.21, 95%CI: 0.99-1.48), HF (RR: 1.24, 95%CI: 1.14-1.35), CVD events (RR: 1.12, 95%CI: 0.99-1.27), cardiovascular mortality (RR: 1.43, 95%CI: 1.09-1.88), all-cause mortality (RR: 1.37, 95%CI: 1.15-1.62). Furthermore, the dose-response analysis demonstrated a potentially non-linear relationship between FGF23 and stroke, HF, and all-cause mortality. In contrast, a potentially linear relationship between FGF23 and cardiovascular mortality was observed (p for non-linearity = 0.73). CONCLUSION The present study suggests that increased serum FGF23 levels are positively related to CVD events and mortality in the general population. The clinical application of FGF23 levels to predict CVD risk requires further research.
Collapse
Affiliation(s)
- Menglu Liu
- Department of Cardiology, Seventh People’s Hospital of Zhengzhou, Zhengzhou, China
| | - Panpan Xia
- Department of Endocrine, The Second Affiliated Hospital of Nanchang University, Nanchang, China
| | - Ziqi Tan
- Department of Endocrine, The Second Affiliated Hospital of Nanchang University, Nanchang, China
| | - Tiangang Song
- Department of Endocrine, The Second Affiliated Hospital of Nanchang University, Nanchang, China
| | - Kaibo Mei
- Department of Anesthesiology, People’s Hospital of Shangrao, Shangrao, China
| | - Jingfeng Wang
- Department of Cardiology, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, Guangdong, China
| | - Jianyong Ma
- Department of Pharmacology and Systems Physiology, University of Cincinnati College of Medicine, Cincinnati, OH, United Status
| | - Yuan Jiang
- Department of Cardiology, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, Guangdong, China
| | - Jing Zhang
- Department of Anesthesiology, The Second Affiliated Hospital of Nanchang University, Nanchang, China
| | - Yujie Zhao
- Department of Cardiology, Seventh People’s Hospital of Zhengzhou, Zhengzhou, China
| | - Peng Yu
- Department of Endocrine, The Second Affiliated Hospital of Nanchang University, Nanchang, China
| | - Xiao Liu
- Department of Cardiology, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, Guangdong, China
| |
Collapse
|
13
|
Vergara N, de Mier MVPR, Rodelo-Haad C, Revilla-González G, Membrives C, Díaz-Tocados JM, Martínez-Moreno JM, Torralbo AI, Herencia C, Rodríguez-Ortiz ME, López-Baltanás R, Richards WG, Felsenfeld A, Almadén Y, Martin-Malo A, Ureña J, Santamaría R, Soriano S, Rodríguez M, Muñoz-Castañeda JR. The direct effect of fibroblast growth factor 23 on vascular smooth muscle cell phenotype and function. Nephrol Dial Transplant 2022; 38:322-343. [PMID: 35867864 PMCID: PMC9923714 DOI: 10.1093/ndt/gfac220] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2022] [Indexed: 11/13/2022] Open
Abstract
BACKGROUND In chronic kidney disease (CKD) patients, increased levels of fibroblast growth factor 23 (FGF23) are associated with cardiovascular mortality. The relationship between FGF23 and heart hypertrophy has been documented, however, it is not known whether FGF23 has an effect on vasculature. Vascular smooth muscle cells VSMCs may exhibit different phenotypes; our hypothesis is that FGF23 favours a switch from a contractile to synthetic phenotype that may cause vascular dysfunction. Our objective was to determine whether FGF23 may directly control a change in VSMC phenotype. METHODS This study includes in vitro, in vivo and ex vivo experiments and evaluation of patients with CKD stages 2-3 studying a relationship between FGF23 and vascular dysfunction. RESULTS In vitro studies show that high levels of FGF23, by acting on its specific receptor FGFR1 and Erk1/2, causes a change in the phenotype of VSMCs from contractile to synthetic. This change is mediated by a downregulation of miR-221/222, which augments the expression of MAP3K2 and PAK1. miR-221/222 transfections recovered the contractile phenotype of VSMCs. Infusion of recombinant FGF23 to rats increased vascular wall thickness, with VSMCs showing a synthetic phenotype with a reduction of miR-221 expression. Ex-vivo studies on aortic rings demonstrate also that high FGF23 increases arterial stiffening. In CKD 2-3 patients, elevation of FGF23 was associated with increased pulse wave velocity and reduced plasma levels of miR-221/222. CONCLUSION In VSMCs, high levels of FGF23, through the downregulation of miR-221/222, causes a change to a synthetic phenotype. This change in VSMCs increases arterial stiffening and impairs vascular function, which might ultimately worsen cardiovascular disease.
Collapse
Affiliation(s)
| | | | | | - Gonzalo Revilla-González
- Instituto de Biomedicina de Sevilla, Hospital Universitario Virgen del Rocío/CSIC/Universidad de Sevilla, Departemento de Fisiología Médica y Biofísica, Sevilla, Spain
| | - Cristina Membrives
- Maimonides Institute for Biomedical Research of Cordoba, Cordoba, Spain,University of Cordoba, Spain
| | - Juan M Díaz-Tocados
- Maimonides Institute for Biomedical Research of Cordoba, Cordoba, Spain,University of Cordoba, Spain
| | - Julio M Martínez-Moreno
- Maimonides Institute for Biomedical Research of Cordoba, Cordoba, Spain,University of Cordoba, Spain
| | - Ana I Torralbo
- Maimonides Institute for Biomedical Research of Cordoba, Cordoba, Spain,University of Cordoba, Spain
| | - Carmen Herencia
- Maimonides Institute for Biomedical Research of Cordoba, Cordoba, Spain,University of Cordoba, Spain
| | | | - Rodrigo López-Baltanás
- Maimonides Institute for Biomedical Research of Cordoba, Cordoba, Spain,University of Cordoba, Spain
| | | | - Arnold Felsenfeld
- Department of Medicine, Veterans Affairs Greater Los Angeles Healthcare System and the David Geffen School of Medicine, University of California, Los Angeles, CA, USA
| | - Yolanda Almadén
- Maimonides Institute for Biomedical Research of Cordoba, Cordoba, Spain,Internal Medicine Service, Reina Sofia University Hospital, Cordoba, Spain,Spanish Biomedical Research Networking Centre consortium for the area of Physiopathology of Obesity and Nutrition, Institute of Health Carlos III, Madrid, Spain
| | - Alejandro Martin-Malo
- Maimonides Institute for Biomedical Research of Cordoba, Cordoba, Spain,University of Cordoba, Spain,Nephrology Service, Reina Sofia University Hospital, Cordoba, Spain,Spanish Renal Research Network (REDinREN), Institute of Health Carlos III, Madrid, Spain, and the European Uremic Toxins group
| | - Juan Ureña
- Instituto de Biomedicina de Sevilla, Hospital Universitario Virgen del Rocío/CSIC/Universidad de Sevilla, Departemento de Fisiología Médica y Biofísica, Sevilla, Spain
| | | | - Sagrario Soriano
- Maimonides Institute for Biomedical Research of Cordoba, Cordoba, Spain,University of Cordoba, Spain,Nephrology Service, Reina Sofia University Hospital, Cordoba, Spain,Spanish Renal Research Network (REDinREN), Institute of Health Carlos III, Madrid, Spain, and the European Uremic Toxins group
| | | | | |
Collapse
|
14
|
Foroni MZ, Cendoroglo MS, Costa AG, Marin-Mio RV, do Prado Moreira PF, Maeda SS, Bilezikian JP, Lazaretti-Castro M. FGF23 levels as a marker of physical performance and falls in community-dwelling very old individuals. ARCHIVES OF ENDOCRINOLOGY AND METABOLISM 2022; 66:333-344. [PMID: 35612845 PMCID: PMC9832858 DOI: 10.20945/2359-3997000000488] [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: 05/13/2021] [Accepted: 03/07/2022] [Indexed: 11/23/2022]
Abstract
Objective The fibroblast growth factor 23 (FGF23) has been related to biological aging, but data in elderly individuals are scant. We determined the profile of serum FGF23 levels in a population of very-old individuals and studied their correlations with parameters of bone metabolism and health markers, as functional performance. Methods This cross-sectional study was performed on 182 community dwellers aged ≥ 80 years. Serum levels of FGF23, PTH, calcium, albumin, phosphorus, creatinine, bone markers, and bone mineral density data were analyzed. Physical performance was evaluated with the stationary march (Step), Flamingo, and functional reach tests, along with questionnaires to assess falls and fractures in the previous year, energy expenditure (MET), and the Charlson index (CI). Physical activity was evaluated with the International Physical Activity Questionnaire (IPAQ). Results Most participants (75%) had FGF23 levels between 30-120 RU/mL (range: 6.0-3,170.0 RU/mL). FGF23 levels correlated with estimated glomerular filtration rate (eGFR; r = -0.335; p = 0.001) and PTH (r = 0.318; p < 0.0001). Individuals with FGF23 in the highest tertile had more falls in the previous year (p = 0.032), worse performance in the Flamingo (p = 0.009) and Step (p < 0.001) tests, worse CI (p = 0.009) and a trend toward sedentary lifestyle (p = 0.056). On multiple regression, FGF23 tertiles remained significant, independently of eGFR, for falls in the previous year, performance in the Flamingo and stationary march tests, lean mass index, and IPAQ classification. Conclusion In a population of very elderly individuals, FGF23 levels were inversely associated with neuromuscular and functional performances. Higher concentrations were related to more falls, lower muscle strength and aerobic capacity, and poorer balance, regardless of renal function, suggesting a potentially deleterious role of high FGF23 concentrations in musculoskeletal health.
Collapse
Affiliation(s)
- Mariana Zuccolotto Foroni
- Divisão de Endocrinologia, Escola Paulista de Medicina - Universidade Federal de São Paulo (Unifesp), São Paulo, SP, Brasil,
| | - Maysa Seabra Cendoroglo
- Divisão de Geriatria, Escola Paulista de Medicina - Universidade Federal de São Paulo (Unifesp), São Paulo, SP, Brasil
| | - Aline Granja Costa
- Divisão de Endocrinologia, Escola Paulista de Medicina - Universidade Federal de São Paulo (Unifesp), São Paulo, SP, Brasil
| | - Rosangela Villa Marin-Mio
- Divisão de Endocrinologia, Escola Paulista de Medicina - Universidade Federal de São Paulo (Unifesp), São Paulo, SP, Brasil
| | | | - Sergio Setsuo Maeda
- Divisão de Endocrinologia, Escola Paulista de Medicina - Universidade Federal de São Paulo (Unifesp), São Paulo, SP, Brasil
| | - John P Bilezikian
- Department of Medicine, Division of Endocrinology, College of Physicians and Surgeons, Columbia University, New York, New York, USA
| | - Marise Lazaretti-Castro
- Divisão de Endocrinologia, Escola Paulista de Medicina - Universidade Federal de São Paulo (Unifesp), São Paulo, SP, Brasil
| |
Collapse
|
15
|
Tanaka SI, Fujioka Y, Tsujino T, Ishida T, Hirata KI. Association between urinary N-acetyl-β-glucosaminidase activity–urinary creatinine concentration ratio and risk of disability and all-cause mortality. PLoS One 2022; 17:e0265637. [PMID: 35333903 PMCID: PMC8956177 DOI: 10.1371/journal.pone.0265637] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2021] [Accepted: 03/07/2022] [Indexed: 12/03/2022] Open
Abstract
Background Recent studies have suggested that chronic kidney disease is associated with cardiovascular disease, dementia, and frailty, all of which cause disability and early death. We investigated whether increased activity of urinary N-acetyl-β-glucosaminidase (NAG), a marker of kidney injury, is associated with risk of disability or all-cause mortality in a general population. Methods Follow-up data from the Hidaka Cohort Study, a population-based cohort study of members of a Japanese rural community, were obtained via questionnaires completed by participants or their relatives. Multivariable analyses were used to investigate relations between urinary NAG activity–urinary creatinine concentration ratio and risk of disability or all-cause mortality. Results A total of 1182 participants were followed up for a median of 12.4 years. The endpoints were receipt of support under the public long-term care insurance program, and all-cause mortality. A total of 122 participants (10.3%) were reported to be receiving long-term care and 230 (19.5%) had died. After adjustment for cardiovascular risk factors along with physical activity, and using the quartile 1 results as a reference, the odds ratio (OR) for disability was 2.12 [95% confidence interval (95% confidence interval [CI]), 1.04–4.33; p = 0.038) and the hazard ratio (HR) for all-cause mortality was 1.65 (95% CI, 1.05–2.62; p = 0.031) in participants with urinary NAG/creatinine ratio in quartile 4. Similar results were obtained in participants without proteinuria: OR for disability, 2.46 (95% CI, 1.18–5.16; p = 0.017); and HR for all-cause mortality, 1.62 (95% CI, 1.00–2.63; p = 0.049). Conclusions Increased urinary NAG/creatinine ratio was associated with risk of disability or all-cause mortality in a general population.
Collapse
Affiliation(s)
- Shin-ichiro Tanaka
- Department of Internal Medicine, Toyooka Hospital Hidaka Medical Center, Toyooka, Hyogo, Japan
- * E-mail:
| | - Yoshio Fujioka
- Division of Clinical Nutrition, Faculty of Nutrition, Kobe Gakuin University, Kobe, Japan
| | - Takeshi Tsujino
- Department of Pharmacy, School of Pharmacy, Hyogo University of Health Sciences, Kobe, Japan
| | - Tatsuro Ishida
- Division of Cardiovascular Medicine, Kobe University Graduate School of Medicine, Kobe, Japan
| | - Ken-ichi Hirata
- Division of Cardiovascular Medicine, Kobe University Graduate School of Medicine, Kobe, Japan
| |
Collapse
|
16
|
Fibroblast Growth Factor 23 as Regulator of Vitamin D Metabolism. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2022; 1362:47-54. [DOI: 10.1007/978-3-030-91623-7_6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/18/2022]
|
17
|
Meshkini F, Soltani S, Clark CCT, Tam V, Meyre D, Toupchian O, Saraf-Bank S, Abdollahi S. The effect of vitamin D supplementation on serum levels of fibroblast growth factor- 23: A systematic review and meta-analysis of randomized controlled trials. J Steroid Biochem Mol Biol 2022; 215:106012. [PMID: 34710560 DOI: 10.1016/j.jsbmb.2021.106012] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/27/2021] [Revised: 09/18/2021] [Accepted: 10/21/2021] [Indexed: 12/29/2022]
Abstract
Previous studies of the effect of vtamin D on serum levels of fibroblast growth factor- 23 (FGF-23) have yeilded an inconsistent findings. This systematic review and meta-analysis of randomized controlled trials (RCTs) sought to investigate the effect of vitamin D supplementation on serum levels of FGF-23. PubMed, Scopus, ISI Web of Science, and the Cochrane Library were searched, from database inception to November 2020, for RCTs that evaluated the effects of native or active vitamin D supplementation on serum levels of FGF-23 in adults. Weighted mean difference (WMD) were calculated and random effects meta-analysis was used to estimate the overall effects. Twenty-seven trials were included in the meta-analysis. Supplementation with native vitamin D (23 studies, n = 2247 participants; weighted mean difference [WMD] = 0.5 pg/mL, 95 % CI: -0.52 to 1.51, P = 0.33; I2 = 29.9 %), and active vitamin D (5 studies, n = 342 participants, WMD = 29.45 pg/mL, 95 % CI: -3.9 to 62.81, P = 0.08; I2 = 99.3%) had no significant effects on serum FGF-23 concentration. In subgroup analyses, supplementation with ergocalciferol (3 studies, n = 205 participants; WMD = 18.27 pg/mL, 95 % CI: 5.36-31.17, P = 0.006), and daily dosing regimens (9 studies, n = 1374 participants; WMD = 0.41 pg/mL, 95 % CI: 0.22 to 0.59, P < 0.001) increased serum FGF-23 levels compared to control. Overall, our findings revealed no significan effect of vitamin D supplementation on serum FGF-23 concentration. However, further high quality, large-scale studies are needed to better elucidate this relationship.
Collapse
Affiliation(s)
- Fatemeh Meshkini
- Department of Biochemistry, School of Medicine, Shahid Sadoughi University of Medical Sciences, Yazd, Iran; Student Research Committee, Shahid Sadoughi University of Medical Sciences, Yazd, Iran
| | - Sepideh Soltani
- Yazd Cardiovascular Research Center, Shahid Sadoughi University of Medical Sciences, Yazd, Iran
| | - Cain C T Clark
- Centre for Intelligent Healthcare, Coventry University, Coventry, CV1 5FB, UK
| | - Vivian Tam
- Department of Health Research Methods, Evidence, and Impact, McMaster University, Hamilton, Canada
| | - David Meyre
- Department of Health Research Methods, Evidence, and Impact, McMaster University, Hamilton, Canada; Department of Pathology and Molecular Medicine, McMaster University, Hamilton, Canada
| | - Omid Toupchian
- Department of Nutrition, School of Public Health, North Khorasan University of Medical Sciences, Bojnurd, Iran
| | - Sahar Saraf-Bank
- Food Security Research Center, Department of Community Nutrition, Isfahan University of Medical Sciences, Isfahan, Iran
| | - Shima Abdollahi
- Department of Nutrition, School of Public Health, North Khorasan University of Medical Sciences, Bojnurd, Iran.
| |
Collapse
|
18
|
Alderson HV, Chinnadurai R, Ibrahim ST, Asar O, Ritchie JP, Middleton R, Larsson A, Diggle PJ, Larsson TE, Kalra PA. Longitudinal change in c-terminal fibroblast growth factor 23 and outcomes in patients with advanced chronic kidney disease. BMC Nephrol 2021; 22:329. [PMID: 34600515 PMCID: PMC8487581 DOI: 10.1186/s12882-021-02528-2] [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: 10/16/2020] [Accepted: 09/10/2021] [Indexed: 11/10/2022] Open
Abstract
BACKGROUND Fibroblast growth factor23 (FGF23) is elevated in CKD and has been associated with outcomes such as death, cardiovascular (CV) events and progression to Renal Replacement therapy (RRT). The majority of studies have been unable to account for change in FGF23 over time and those which have demonstrate conflicting results. We performed a survival analysis looking at change in c-terminal FGF23 (cFGF23) over time to assess the relative contribution of cFGF23 to these outcomes. METHODS We measured cFGF23 on plasma samples from 388 patients with CKD 3-5 who had serial measurements of cFGF23, with a mean of 4.2 samples per individual. We used linear regression analysis to assess the annual rate of change in cFGF23 and assessed the relationship between time-varying cFGF23 and the outcomes in a cox-regression analysis. RESULTS Across our population, median baseline eGFR was 32.3mls/min/1.73m2, median baseline cFGF23 was 162 relative units/ml (RU/ml) (IQR 101-244 RU/mL). Over 70 months (IQR 53-97) median follow-up, 76 (19.6%) patients progressed to RRT, 86 (22.2%) died, and 52 (13.4%) suffered a major non-fatal CV event. On multivariate analysis, longitudinal change in cFGF23 was significantly associated with risk for death and progression to RRT but not non-fatal cardiovascular events. CONCLUSION In our study, increasing cFGF23 was significantly associated with risk for death and RRT.
Collapse
Affiliation(s)
- Helen V Alderson
- Vascular Research Group, Manchester Academic Health Sciences Centre, University of Manchester, Salford Royal NHS Foundation Trust, Salford, UK. .,Department of Renal Medicine, Salford Royal Hospital, Level 2, Hope Building, Stott Lane, Salford, M6 8HD, UK.
| | - Rajkumar Chinnadurai
- Vascular Research Group, Manchester Academic Health Sciences Centre, University of Manchester, Salford Royal NHS Foundation Trust, Salford, UK
| | - Sara T Ibrahim
- Vascular Research Group, Manchester Academic Health Sciences Centre, University of Manchester, Salford Royal NHS Foundation Trust, Salford, UK.,Department of Internal Medicine and Nephrology, Faculty of Medicine, Alexandria University, Alexandria, Egypt
| | - Ozgur Asar
- Department of Biostatistics and Medical Informatics, Acibadem Mehmet Ali Aydinlar University, Istanbul, Turkey
| | - James P Ritchie
- Vascular Research Group, Manchester Academic Health Sciences Centre, University of Manchester, Salford Royal NHS Foundation Trust, Salford, UK
| | - Rachel Middleton
- Vascular Research Group, Manchester Academic Health Sciences Centre, University of Manchester, Salford Royal NHS Foundation Trust, Salford, UK
| | - Anders Larsson
- Section of Clinical Chemistry, Department of Medical Sciences, Uppsala University, Uppsala, Sweden
| | - Peter J Diggle
- CHICAS, Lancaster Medical School, Lancaster University, Lancaster, UK
| | - Tobias E Larsson
- Department of Clinical Science, Intervention and Technology, Renal Unit, Karolinska Institute, Stockholm, Sweden
| | - Philip A Kalra
- Vascular Research Group, Manchester Academic Health Sciences Centre, University of Manchester, Salford Royal NHS Foundation Trust, Salford, UK
| |
Collapse
|
19
|
Zhang R, Wang SY, Yang F, Ma S, Lu X, Kan C, Zhang JB. Crosstalk of fibroblast growth factor 23 and anemia-related factors during the development and progression of CKD (Review). Exp Ther Med 2021; 22:1159. [PMID: 34504604 PMCID: PMC8393509 DOI: 10.3892/etm.2021.10593] [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: 06/19/2020] [Accepted: 03/08/2021] [Indexed: 11/06/2022] Open
Abstract
Fibroblast growth factor 23 (FGF23) plays an important role in the development of chronic kidney disease-mineral bone disorder (CKD-MBD). Abnormally elevated levels of 1,25-dihydroxyvitamin D cause osteocytes to secrete FGF23, which subsequently induces phosphaturia. Recent studies have reported that iron deficiency, erythropoietin (EPO) and hypoxia regulate the pathways responsible for FGF23 production. However, the molecular mechanisms underlying the interactions between FGF23 and anemia-related factors are not yet fully understood. The present review discusses the associations between FGF23, iron, EPO and hypoxia-inducible factors (HIFs), and their impact on FGF23 bioactivity, focusing on recent studies. Collectively, these findings propose interactions between FGF23 gene expression and anemia-related factors, including iron deficiency, EPO and HIFs. Taken together, these results suggest that FGF23 bioactivity is closely associated with the occurrence of CKD-related anemia and CKD-MBD.
Collapse
Affiliation(s)
- Rui Zhang
- Department of Nephrology, Jilin Province People's Hospital, Changchun, Jilin 130021, P.R. China
| | - Song-Yan Wang
- Department of Nephrology, Jilin Province People's Hospital, Changchun, Jilin 130021, P.R. China
| | - Fan Yang
- Department of Nephrology, Jilin Province People's Hospital, Changchun, Jilin 130021, P.R. China
| | - Shuang Ma
- Department of Nephrology, Jilin Province People's Hospital, Changchun, Jilin 130021, P.R. China
| | - Xu Lu
- Department of Clinical Medicine, Changchun University of Chinese Medicine, Changchun, Jilin 130000, P.R. China
| | - Chao Kan
- Department of Clinical Medicine, Changchun University of Chinese Medicine, Changchun, Jilin 130000, P.R. China
| | - Jing-Bin Zhang
- Department of Nephrology, Jilin Province People's Hospital, Changchun, Jilin 130021, P.R. China
| |
Collapse
|
20
|
Hampson G, Elder GJ, Cohen-Solal M, Abrahamsen B. A review and perspective on the assessment, management and prevention of fragility fractures in patients with osteoporosis and chronic kidney disease. Endocrine 2021; 73:509-529. [PMID: 33974225 PMCID: PMC8325650 DOI: 10.1007/s12020-021-02735-9] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/19/2021] [Accepted: 04/19/2021] [Indexed: 01/01/2023]
Abstract
This article aims to review the methods used for the assessment of fracture risk and the use of osteoporosis medications for fracture prevention in the population with CKD, and highlights the difficulties faced by clinicians in the management of these patients and the latest recommendations and guidelines. Chronic kidney disease (CKD) and osteoporosis often co-exist in older adults, and they present a major healthcare challenge. CKD mineral and bone disorder (CKD-MBD) occurs as renal function declines and this syndrome affects most patients in CKD stages 4 and 5. The biochemical abnormalities of CKD-MBD, renal bone disease and risk factors associated with age-related bone loss and osteoporosis lead to a cumulative effect on fracture risk and mortality. There is a need for routine evaluation of fracture risk and fracture prevention in this population. Measurement of bone mineral density (BMD) and the use of the FRAX tool have predictive value for incident fractures in the general population and in CKD. This enables physicians to identify CKD patients most at risk of sustaining a fragility fracture and allows a more targeted approach to fracture prevention. Data analysis from the pivotal trials of therapeutic agents used in osteoporosis show that these drugs can be considered in mild and moderate CKD (stages 1-3 CKD). Off-label drug use in patients with CKD-MBD and more severe renal impairment (CKD stages 4 and 5) could offer significant benefits to sub-groups of patients when carefully tailored to each individual's bone turnover and calcium and phosphate balance. However, this requires a selective approach and treatment decisions based on inference from pathophysiology while we await further trials. Guidelines advocate the correction and/or reduction of the biochemical abnormalities of CKD-MBD before initiation of treatment with osteoporosis drugs and close monitoring during treatment.
Collapse
Affiliation(s)
- Geeta Hampson
- Department of Chemical Pathology and Metabolic Medicine, St Thomas' Hospital, London, UK.
- Metabolic Bone Clinic, Department of Rheumatology, Guy's Hospital, London, UK.
| | - Grahame J Elder
- Department of Renal Medicine, Westmead Hospital, Sydney, New South Wales, Australia
- Osteoporosis and Bone Biology Program, Garvan Institute of Medical Research, Sydney, New South Wales, Australia
- Faculty of Medicine, University of Notre Dame Australia, Level 2, 88-90 Water Street, Auburn, New South Wales, 2144, Australia
| | - Martine Cohen-Solal
- Bioscar Inserm U1132 and Université de Paris, Hôpital Lariboisière, F-75010, Paris, France
| | - Bo Abrahamsen
- Department of Medicine, Holbæk Hospital, Holbæk, Denmark
- Department of Clinical Research, Open Data Explorative Network, University of Southern Denmark, Odense, Denmark
| |
Collapse
|
21
|
Inflammation: a putative link between phosphate metabolism and cardiovascular disease. Clin Sci (Lond) 2021; 135:201-227. [PMID: 33416083 PMCID: PMC7796315 DOI: 10.1042/cs20190895] [Citation(s) in RCA: 42] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2020] [Revised: 12/11/2020] [Accepted: 12/15/2020] [Indexed: 02/06/2023]
Abstract
Dietary habits in the western world lead to increasing phosphate intake. Under physiological conditions, extraosseous precipitation of phosphate with calcium is prevented by a mineral buffering system composed of calcification inhibitors and tight control of serum phosphate levels. The coordinated hormonal regulation of serum phosphate involves fibroblast growth factor 23 (FGF23), αKlotho, parathyroid hormone (PTH) and calcitriol. A severe derangement of phosphate homeostasis is observed in patients with chronic kidney disease (CKD), a patient collective with extremely high risk of cardiovascular morbidity and mortality. Higher phosphate levels in serum have been associated with increased risk for cardiovascular disease (CVD) in CKD patients, but also in the general population. The causal connections between phosphate and CVD are currently incompletely understood. An assumed link between phosphate and cardiovascular risk is the development of medial vascular calcification, a process actively promoted and regulated by a complex mechanistic interplay involving activation of pro-inflammatory signalling. Emerging evidence indicates a link between disturbances in phosphate homeostasis and inflammation. The present review focuses on critical interactions of phosphate homeostasis, inflammation, vascular calcification and CVD. Especially, pro-inflammatory responses mediating hyperphosphatemia-related development of vascular calcification as well as FGF23 as a critical factor in the interplay between inflammation and cardiovascular alterations, beyond its phosphaturic effects, are addressed.
Collapse
|
22
|
McCarty MF, Lerner A, DiNicolantonio JJ, Iloki-Assanga SB. High Intakes of Bioavailable Phosphate May Promote Systemic Oxidative Stress and Vascular Calcification by Boosting Mitochondrial Membrane Potential-Is Good Magnesium Status an Antidote? Cells 2021; 10:1744. [PMID: 34359914 PMCID: PMC8303439 DOI: 10.3390/cells10071744] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2021] [Revised: 06/23/2021] [Accepted: 06/29/2021] [Indexed: 12/23/2022] Open
Abstract
Chronic kidney disease is characterized by markedly increased risk for cardiovascular mortality, vascular calcification, and ventricular hypertrophy, and is associated with increased systemic oxidative stress. Hyperphosphatemia, reflecting diminished glomerular phosphate (Pi) clearance, coupled with a compensatory increase in fibroblast growth factor 23 (FGF23) secretion are thought to be key mediators of this risk. Elevated serum and dietary Pi and elevated plasma FGF23 are associated with increased cardiovascular and total mortality in people with normal baseline renal function. FGF23 may mediate some of this risk by promoting cardiac hypertrophy via activation of fibroblast growth factor receptor 4 on cardiomyocytes. Elevated serum Pi can also cause a profound increase in systemic oxidative stress, and this may reflect the ability of Pi to act directly on mitochondria to boost membrane potential and thereby increase respiratory chain superoxide production. Moreover, elevated FGF23 likewise induces oxidative stress in vascular endothelium via activation of NADPH oxidase complexes. In vitro exposure of vascular smooth muscle cells to elevated Pi provokes an osteoblastic phenotypic transition that is mediated by increased mitochondrial oxidant production; this is offset dose-dependently by increased exposure to magnesium (Mg). In vivo, dietary Mg is protective in rodent models of vascular calcification. It is proposed that increased intracellular Mg opposes Pi's ability to increase mitochondrial membrane potential; this model could explain its utility for prevention of vascular calcification and predicts that Mg may have a more global protective impact with regard to the direct pathogenic effects of hyperphosphatemia.
Collapse
Affiliation(s)
- Mark F. McCarty
- Catalytic Longevity Foundation, 811 B Nahant Ct., San Diego, CA 92109, USA;
| | - Aaron Lerner
- Chaim Sheba Medical Center, The Zabludowicz Research Center for Autoimmune Diseases, Tel Hashomer 5262000, Israel
| | | | - Simon B. Iloki-Assanga
- Department of Research and Postgraduate in Food Science, Universidad de Sonora, Hermosillo 83000, Mexico;
| |
Collapse
|
23
|
FGF23: A Review of Its Role in Mineral Metabolism and Renal and Cardiovascular Disease. DISEASE MARKERS 2021; 2021:8821292. [PMID: 34055103 PMCID: PMC8149241 DOI: 10.1155/2021/8821292] [Citation(s) in RCA: 23] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/18/2020] [Revised: 11/01/2020] [Accepted: 05/04/2021] [Indexed: 01/03/2023]
Abstract
FGF23 is a hormone secreted mainly by osteocytes and osteoblasts in bone. Its pivotal role concerns the maintenance of mineral ion homeostasis. It has been confirmed that phosphate and vitamin D metabolisms are related to the effect of FGF23 and its excess or deficiency leads to various hereditary diseases. Multiple studies have shown that FGF23 level increases in the very early stages of chronic kidney disease (CKD), and its concentration may also be highly associated with cardiac complications. The present review is limited to some of the most important aspects of calcium and phosphate metabolism. It discusses the role of FGF23, which is considered an early and sensitive marker for CKD-related bone disease but also as a novel and potent cardiovascular risk factor. Furthermore, this review gives particular attention to the reliability of FGF23 measurement and various confounding factors that may impact on the clinical utility of FGF23. Finally, this review elaborates on the clinical usefulness of FGF23 and evaluates whether FGF23 may be considered a therapeutic target.
Collapse
|
24
|
Vázquez-Sánchez S, Poveda J, Navarro-García JA, González-Lafuente L, Rodríguez-Sánchez E, Ruilope LM, Ruiz-Hurtado G. An Overview of FGF-23 as a Novel Candidate Biomarker of Cardiovascular Risk. Front Physiol 2021; 12:632260. [PMID: 33767635 PMCID: PMC7985069 DOI: 10.3389/fphys.2021.632260] [Citation(s) in RCA: 42] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2020] [Accepted: 02/15/2021] [Indexed: 12/12/2022] Open
Abstract
Fibroblast growth factor-23 (FGF)-23 is a phosphaturic hormone involved in mineral bone metabolism that helps control phosphate homeostasis and reduces 1,25-dihydroxyvitamin D synthesis. Recent data have highlighted the relevant direct FGF-23 effects on the myocardium, and high plasma levels of FGF-23 have been associated with adverse cardiovascular outcomes in humans, such as heart failure and arrhythmias. Therefore, FGF-23 has emerged as a novel biomarker of cardiovascular risk in the last decade. Indeed, experimental data suggest FGF-23 as a direct mediator of cardiac hypertrophy development, cardiac fibrosis and cardiac dysfunction via specific myocardial FGF receptor (FGFR) activation. Therefore, the FGF-23/FGFR pathway might be a suitable therapeutic target for reducing the deleterious effects of FGF-23 on the cardiovascular system. More research is needed to fully understand the intracellular FGF-23-dependent mechanisms, clarify the downstream pathways and identify which could be the most appropriate targets for better therapeutic intervention. This review updates the current knowledge on both clinical and experimental studies and highlights the evidence linking FGF-23 to cardiovascular events. The aim of this review is to establish the specific role of FGF-23 in the heart, its detrimental effects on cardiac tissue and the possible new therapeutic opportunities to block these effects.
Collapse
Affiliation(s)
- Sara Vázquez-Sánchez
- Cardiorenal Translational Laboratory, Institute of Research i+12, Hospital Universitario 12 de Octubre, Madrid, Spain
| | - Jonay Poveda
- Cardiorenal Translational Laboratory, Institute of Research i+12, Hospital Universitario 12 de Octubre, Madrid, Spain
| | - José Alberto Navarro-García
- Cardiorenal Translational Laboratory, Institute of Research i+12, Hospital Universitario 12 de Octubre, Madrid, Spain
| | - Laura González-Lafuente
- Cardiorenal Translational Laboratory, Institute of Research i+12, Hospital Universitario 12 de Octubre, Madrid, Spain
| | - Elena Rodríguez-Sánchez
- Cardiorenal Translational Laboratory, Institute of Research i+12, Hospital Universitario 12 de Octubre, Madrid, Spain
| | - Luis M. Ruilope
- Cardiorenal Translational Laboratory, Institute of Research i+12, Hospital Universitario 12 de Octubre, Madrid, Spain
- CIBER-CV, Hospital Universitario 12 de Octubre, Madrid, Spain
- School of Doctoral Studies and Research, European University of Madrid, Madrid, Spain
| | - Gema Ruiz-Hurtado
- Cardiorenal Translational Laboratory, Institute of Research i+12, Hospital Universitario 12 de Octubre, Madrid, Spain
- CIBER-CV, Hospital Universitario 12 de Octubre, Madrid, Spain
| |
Collapse
|
25
|
Mirna M, Lauten A, Jirak P, Rezar R, Wernly B, Paar V, Felder TK, Hoppe UC, Motloch LJ, Jung C, Alushi B, Lichtenauer M, Salmhofer H. Serum levels of C-terminal FGF23 (cFGF23) are associated with 1-year-mortality in patients undergoing transcatheter aortic valve replacement (TAVR). Eur J Intern Med 2021; 85:98-107. [PMID: 33191056 DOI: 10.1016/j.ejim.2020.09.022] [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: 05/12/2020] [Revised: 08/23/2020] [Accepted: 09/23/2020] [Indexed: 11/22/2022]
Abstract
INTRODUCTION Serum levels of FGF23 have been associated with adverse outcomes in cardiovascular diseases in patients with and without impaired renal function. Hence, this study aimed to explore the prognostic relevance of intact FGF23 (iFGF23) and its derivate C-terminal FGF23 (cFGF23) in patients undergoing transcatheter aortic valve replacement (TAVR) with regard to renal function. METHODS A total of 274 patients undergoing transfemoral TAVR were enrolled in this study. Blood samples were obtained preinterventionally and analyzed for iFGF23 and cFGF23 by means of enzyme linked immunosorbent assay (ELISA). Follow-up was obtained for 12 months. RESULTS Serum levels of cFGF23 and iFGF23 both correlated positively with serum creatinine and inversely with estimated glomerular filtration rate (eGFR). Cox regression analysis revealed a significant association of cFGF23 with 1-year-mortality in patients with eGFR ≥45ml/min/1.73m², but not in patients with an eGFR <45ml/min/1.73m². A cut-off was calculated for cFGF23 (6.82 pmol/l) and patients with eGFR ≥45ml/min/1.73m² were retrospectively divided into two groups (above/below cut-off). Patients above the cut-off had a significantly worse 1-year-mortality than patients below the cut-off (33.3% vs. 19.6%; OR 2.05 (95%CI 1.03-4.07), p= 0.038). The association of cFGF23 with 1-year-mortality in patients with eGFR ≥45ml/min/1.73m² remained statistically significant even after correction for possible confounders in a multivariate Cox regression analysis. CONCLUSION cFGF23 could be an individual risk factor for mortality in patients undergoing TAVR with an eGFR ≥45ml/min/1.73m².
Collapse
Affiliation(s)
- Moritz Mirna
- Department of Internal Medicine II, Division of Cardiology, Paracelsus Medical University of Salzburg, Müllner Hauptstraße 48, 5020 Salzburg, Austria.
| | - Alexander Lauten
- Department of General and Interventional Cardiology and Rhythmology, Helios Clinic, Erfurt, Germany
| | - Peter Jirak
- Department of Internal Medicine II, Division of Cardiology, Paracelsus Medical University of Salzburg, Müllner Hauptstraße 48, 5020 Salzburg, Austria
| | - Richard Rezar
- Department of Internal Medicine II, Division of Cardiology, Paracelsus Medical University of Salzburg, Müllner Hauptstraße 48, 5020 Salzburg, Austria
| | - Bernhard Wernly
- Department of Internal Medicine II, Division of Cardiology, Paracelsus Medical University of Salzburg, Müllner Hauptstraße 48, 5020 Salzburg, Austria
| | - Vera Paar
- Department of Internal Medicine II, Division of Cardiology, Paracelsus Medical University of Salzburg, Müllner Hauptstraße 48, 5020 Salzburg, Austria
| | - Thomas K Felder
- Department of Laboratory Medicine, Paracelsus Medical University of Salzburg, Salzburg, Austria
| | - Uta C Hoppe
- Department of Internal Medicine II, Division of Cardiology, Paracelsus Medical University of Salzburg, Müllner Hauptstraße 48, 5020 Salzburg, Austria
| | - Lukas J Motloch
- Department of Internal Medicine II, Division of Cardiology, Paracelsus Medical University of Salzburg, Müllner Hauptstraße 48, 5020 Salzburg, Austria
| | - Christian Jung
- Division of Cardiology, Pulmonology, and Vascular Medicine, Medical Faculty, University Duesseldorf, Germany
| | - Brunilda Alushi
- Department of General and Interventional Cardiology and Rhythmology, Helios Clinic, Erfurt, Germany
| | - Michael Lichtenauer
- Department of Internal Medicine II, Division of Cardiology, Paracelsus Medical University of Salzburg, Müllner Hauptstraße 48, 5020 Salzburg, Austria
| | - Hermann Salmhofer
- Department of Internal Medicine I, Division of Gastroenterology and Nephrology, Paracelsus Medical University of Salzburg, Austria
| |
Collapse
|
26
|
Braithwaite VS, Mwangi MN, Jones KS, Demir AY, Prentice A, Prentice AM, Andang'o PEA, Verhoef H. Antenatal iron supplementation, FGF23, and bone metabolism in Kenyan women and their offspring: secondary analysis of a randomized controlled trial. Am J Clin Nutr 2021; 113:1104-1114. [PMID: 33675347 PMCID: PMC8106766 DOI: 10.1093/ajcn/nqaa417] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2020] [Accepted: 12/10/2020] [Indexed: 12/13/2022] Open
Abstract
BACKGROUND Fibroblast growth factor-23 (FGF23) regulates body phosphate homeostasis primarily by increasing phosphaturia. It also acts as a vitamin D-regulating hormone. Maternal iron deficiency is associated with perturbed expression and/or regulation of FGF23 and hence might be implicated in the pathogenesis of hypophosphatemia-driven rickets in their offspring. OBJECTIVES We aimed to determine the effect of antenatal oral iron supplementation on FGF23 concentration and maternal and infant markers of bone-mineral regulation. METHODS We performed a secondary analysis of a trial in which 470 rural Kenyan women with singleton pregnancies and hemoglobin concentrations ≥ 90 g/L were randomly allocated to daily, supervised supplementation with 60 mg elemental iron as ferrous fumarate or placebo from 13-23 weeks of gestation until 1 mo postpartum. As previously reported, iron supplementation improved iron status in mothers and neonates. For the present study, we reanalyzed all available plasma samples collected in mothers and neonates at birth, with primary outcomes being concentrations of FGF23, measured by 2 assays: 1 that detects intact hormone and C-terminal cleavage products (total-FGF23) and another that detects the intact hormone only (intact-FGF23). RESULTS Analysis was performed on 433 women (n = 216, iron group; n = 217, placebo group) and 414 neonates (n = 207, iron group; n = 207, placebo group). Antenatal iron supplementation reduced geometric mean total-FGF23 concentrations in mothers and neonates by 62.6% (95% CI: 53.0%, 70.3%) and 15.2% (95% CI: -0.3%, 28.4%, P = 0.06), respectively. In addition, it increased geometric mean neonatal intact-FGF23 concentrations by 21.6% (95% CI: 1.2%, 46.1%), increased geometric mean maternal hepcidin concentrations by 136.4% (95% CI: 86.1%, 200.3%), and decreased mean maternal 25-hydroxyvitamin D concentrations by 6.1 nmol/L (95% CI: -11.0, -1.2 nmol/L). CONCLUSIONS Analysis of this randomized trial confirms that iron supplementation can reverse elevated FGF23 production caused by iron deficiency in iron-deficient mothers and their neonates. Further investigations are warranted to assess to what extent iron supplementation can prevent FGF23-mediated hypophosphatemic rickets or osteomalacia.
Collapse
Affiliation(s)
| | - Martin N Mwangi
- Wageningen University, Division of Human Nutrition and Health, P.O. Box 17, 6700 AA Wageningen, The Netherlands,University of Malawi, College of Medicine, Training and Research Unit of Excellence, Private Bag 360, BT 3, Chichiri, Blantyre, Malawi
| | - Kerry S Jones
- National Institute for Health Research (NIHR) Biomedical Research Centre Nutritional Biomarker Laboratory, MRC Epidemiology Unit, University of Cambridge, Cambridge Biomedical Campus, Cambridge, CB2 0AH, United Kingdom
| | - Ayşe Y Demir
- Meander Medical Centre, Laboratory for Clinical Chemistry and Hematology, P.O. Box 1502, 3800 BM Amersfoort, The Netherlands
| | - Ann Prentice
- Medical Research Council (MRC) Nutrition and Bone Health Research Group, Clifford Allbutt Building, University of Cambridge, Cambridge Biomedical Campus, Cambridge, CB2 0AH, United Kingdom (formerly the MRC Elsie Widdowson Laboratory, 120 Fulbourn Road, Cambridge, CB1 9NL, United Kingdom),Medical Research Council (MRC) Unit The Gambia at London School of Hygiene & Tropical Medicine, Atlantic Boulevard, Fajara, Banjul, The Gambia
| | - Andrew M Prentice
- Medical Research Council (MRC) Unit The Gambia at London School of Hygiene & Tropical Medicine, Atlantic Boulevard, Fajara, Banjul, The Gambia
| | - Pauline E A Andang'o
- Maseno University, School of Public Health and Community Development, Maseno, Kenya
| | - Hans Verhoef
- Wageningen University, Division of Human Nutrition and Health, P.O. Box 17, 6700 AA Wageningen, The Netherlands,Wageningen University, Cell Biology and Immunology Group, P.O. Box 338, 6708 WD Wageningen, The Netherlands
| |
Collapse
|
27
|
Sharma S, Katz R, Dubin RF, Drew DA, Gutierrez OM, Shlipak MG, Sarnak MJ, Ix JH. FGF23 and Cause-Specific Mortality in Community-Living Individuals-The Health, Aging, and Body Composition Study. J Am Geriatr Soc 2020; 69:711-717. [PMID: 33170519 DOI: 10.1111/jgs.16910] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2020] [Revised: 09/15/2020] [Accepted: 10/12/2020] [Indexed: 11/29/2022]
Abstract
OBJECTIVES Fibroblast growth factor (FGF)-23 is a key regulator of mineral metabolism and has been linked with left ventricular hypertrophy in animal models. Most existing epidemiologic studies evaluated a C-terminal FGF23 assay which measures both the intact (active) hormone and inactive fragments. The relationship of intact FGF23 with cause-specific mortality is unknown. DESIGN Prospective analyses of data from Health, Aging, & Body Composition (HABC) study. SETTING Community-living adults aged 70 to 79 years with longitudinal follow up.
Collapse
Affiliation(s)
- Shilpa Sharma
- Division of Nephrology, David Geffen School of Medicine at UCLA and VA Greater Los Angeles Healthcare System, Los Angeles, California, USA
| | - Ronit Katz
- Department of Obstetrics and Gynecology, University of Washington, Seattle, Washington, USA
| | - Ruth F Dubin
- Kidney Health Research Collaborative, University of California San Francisco, San Francisco, California, USA
| | - David A Drew
- Division of Nephrology, Department of Medicine, Tufts Medical Center, Boston, Massachusetts, USA
| | - Orlando M Gutierrez
- Departments of Medicine and Epidemiology, University of Alabama at Birmingham, Birmingham, Alabama, USA
| | - Michael G Shlipak
- Kidney Health Research Collaborative, University of California San Francisco, San Francisco, California, USA
| | - Mark J Sarnak
- Division of Nephrology, Department of Medicine, Tufts Medical Center, Boston, Massachusetts, USA
| | - Joachim H Ix
- Division of Nephrology-Hypertension, University of California San Diego and Veteran Affairs San Diego Healthcare System, San Diego, California, USA
| |
Collapse
|
28
|
Komaba H, Fuller DS, Taniguchi M, Yamamoto S, Nomura T, Zhao J, Bieber BA, Robinson BM, Pisoni RL, Fukagawa M. Fibroblast Growth Factor 23 and Mortality Among Prevalent Hemodialysis Patients in the Japan Dialysis Outcomes and Practice Patterns Study. Kidney Int Rep 2020; 5:1956-1964. [PMID: 33163716 PMCID: PMC7609896 DOI: 10.1016/j.ekir.2020.08.013] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2020] [Revised: 08/04/2020] [Accepted: 08/11/2020] [Indexed: 12/13/2022] Open
Abstract
INTRODUCTION Elevated fibroblast growth factor 23 (FGF23) levels have been strongly associated with mortality in the predialysis and incident hemodialysis populations, but few studies have examined this relationship in a large cohort of prevalent hemodialysis patients and in particular among persons with high dialysis vintage. To address this, we analyzed data from the Japan Dialysis Outcomes and Practice Patterns Study (J-DOPPS). METHODS We included 1122 prevalent hemodialysis patients from the J-DOPPS phase 5 (2012-2015) who had FGF23 measurements. We evaluated the association of FGF23 levels with all-cause mortality and cardiovascular composite outcome using Cox regression adjusted for potential confounders. RESULTS At study enrollment, median dialysis vintage was 5.8 years (interquartile range, 2.7-12.4 years) and median FGF23 level was 2113 pg/ml (interquartile range, 583-6880 pg/ml). During 3-year follow-up, 154 of the 1122 participants died. In adjusted analyses, higher FGF23 was associated with a greater hazard of death (hazard ratio per doubling of FGF23, 1.12; 95% confidence interval, 1.03-1.21); however, the association became weaker as the dialysis vintage increased and finally disappeared in the highest tertile (>9.4 years). Similar patterns of effect modification by dialysis vintage were observed for cardiovascular composite outcome and in time-dependent models. CONCLUSION Elevated FGF23 was associated with mortality and cardiovascular events in prevalent hemodialysis patients, but the association was attenuated at longer dialysis vintages. This novel finding suggests that long-term hemodialysis patients may be less susceptible to the detrimental effects of FGF23 or correlated biological processes, and additional studies are needed to gain understanding of these possibilities.
Collapse
Affiliation(s)
- Hirotaka Komaba
- Division of Nephrology, Endocrinology, and Metabolism, Tokai University School of Medicine, Isehara, Japan
- The Institute of Medical Sciences, Tokai University, Isehara, Japan
| | | | | | - Suguru Yamamoto
- Division of Clinical Nephrology and Rheumatology, Niigata University Graduate School of Medical and Dental Sciences, Niigata, Japan
| | - Takanobu Nomura
- Medical Affairs Department, Kyowa Kirin Co., Ltd., Tokyo, Japan
| | - Junhui Zhao
- Arbor Research Collaborative for Health, Ann Arbor, Michigan, USA
| | - Brian A. Bieber
- Arbor Research Collaborative for Health, Ann Arbor, Michigan, USA
| | | | - Ronald L. Pisoni
- Arbor Research Collaborative for Health, Ann Arbor, Michigan, USA
| | - Masafumi Fukagawa
- Division of Nephrology, Endocrinology, and Metabolism, Tokai University School of Medicine, Isehara, Japan
| |
Collapse
|
29
|
|
30
|
Manou E, Thodis E, Arsos G, Pasadakis P, Panagoutsos S, Papadopoulou D, Papagianni A. Fibroblast Growth Factor 23 and α-Klotho Protein Are Associated with Adverse Clinical Outcomes in Non-Dialysis CKD Patients. Kidney Blood Press Res 2020; 45:900-915. [PMID: 33040068 DOI: 10.1159/000510351] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2019] [Accepted: 07/22/2020] [Indexed: 12/20/2022] Open
Abstract
BACKGROUND Fibroblast growth factor 23 (FGF-23) and α-Klotho protein appear to have an important role in the pathogenesis of CKD-mineral and bone disorders. The aim of this study was to investigate the association of FGF-23 and α-Klotho levels with adverse clinical outcomes in patients with non-dialysis CKD. MATERIALS AND METHODS We conducted a prospective cohort study, enrolling participants with non-dialysis CKD from a single center in Greece. At enrollment, glomerular filtration rate (GFR) was measured (mGFR) and plasma levels of carboxyl terminal FGF-23 (cFGF-23) and soluble α-Klotho (sKlotho) were determined by enzyme-linked immunoassay. Participants were followed for up to 5 years or until the occurrence of the primary endpoint of initiation of renal replacement therapy or death. Multivariate regression tree analysis was used to identify informative baseline parameters in order to categorize participants. Also, using median values of cFGF-23 and sKlotho, participants were categorized into 4 groups, in whom survival was compared using Kaplan-Meier and Cox regression analysis. RESULTS 128 participants were enrolled with a median mGFR of 41.5 mL/min/1.73 m2 (IQR = 28.2). Baseline mGFR correlated with cFGF-23 and sKlotho (r = -0.54 and r = 0.49, respectively; p < 0.0001 for both). cFGF-23 and sKlotho levels correlated negatively (r = -0.24, p = 0.006). Multivariate regression tree analysis resulted in 3 groups defined by cutoff values of mGFR (60.9 mL/min/1.73 m2) and phosphate (3.7 mg/dL). These groups correlated with CKD stage, cFGF-23, and sKlotho (p < 0.0001 for all). During a median follow-up of 36 months (IQR = 22), 40 (31.2%) participants reached the primary endpoint (31 initiated renal replacement therapy, 9 died). Survival to primary endpoint differed among the 4 groups formed using median values of both biomarkers, with the low FGF-23/high Klotho and high FGF-23/low Klotho having the longest and shortest survival, respectively. High FGF-23/low Klotho group, compared to the opposite one, had a significantly elevated risk of the primary outcome (HR, 6.8; 95% CI, 2.3-19.6; p = 0.0004). CONCLUSIONS In patients with CKD stages 1-5, the combination of higher cFGF-23 and lower sKlotho levels along with mGFR and serum phosphate was associated with adverse clinical outcomes. The utility of combinations of traditional and novel biomarkers to predict outcomes warrants further study.
Collapse
Affiliation(s)
- Eleni Manou
- Department of Nephrology, Papageorgiou Hospital, Thessaloniki, Greece,
| | - Elias Thodis
- Department of Nephrology, Democritus University of Thrace, Alexandroupolis, Greece
| | - Georgios Arsos
- 3rd Department of Nuclear Medicine, Aristotle University of Thessaloniki, Papageorgiou Hospital, Thessaloniki, Greece
| | - Ploumis Pasadakis
- Department of Nephrology, Democritus University of Thrace, Alexandroupolis, Greece
| | | | | | - Aikaterini Papagianni
- Department of Nephrology, Aristotle University of Thessaloniki, Hippokration Hospital, Thessaloniki, Greece
| |
Collapse
|
31
|
Kwon YE, Choi HY, Oh HJ, Ahn SY, Ryu DR, Kwon YJ. Vertebral fracture is associated with myocardial infarction in incident hemodialysis patients: a Korean nationwide population-based study. Osteoporos Int 2020; 31:1965-1973. [PMID: 32394062 DOI: 10.1007/s00198-020-05423-y] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/21/2019] [Accepted: 04/14/2020] [Indexed: 10/24/2022]
Abstract
UNLABELLED Chronic kidney disease (CKD)-mineral and bone disorder suggests that fragile bone and vascular disorder might be connected closely in CKD patients. In this study, fracture event was significantly associated with myocardial infarction (MI) in end-stage renal disease patients on hemodialysis (HD), especially for vertebral fractures. INTRODUCTION CKD-mineral and bone disorder is characterized by biochemical abnormalities, bone disorders, and vascular calcification. We aimed to verify the association between fracture and MI in CKD patients. METHODS Records for incident CKD stage 3 to 5 patients and patients who initiated HD between July 2014 and June 2018 were retrieved from the Korean Health Insurance Review & Assessment Service Database. Fractures were defined using diagnostic codes and were classified into vertebral, femoral, and other site fractures. MI was defined using a combination of MI diagnostic codes and related procedure codes. Multiple logistic regressions and 1:1 propensity score matching analysis were conducted. RESULTS A total of 38,935 patients (HD, 11,379; pre-dialysis CKD, 27,556) were included in this study. A total of 5,057 (13.0%) patients experienced fracture, and 1,431 (3.7%) patients had MI. Multiple logistic regression analysis showed that fracture was significantly associated with MI in the HD group (odds ratio (OR) 1.34, P = 0.024), but not in the pre-dialysis CKD group (OR 1.04, P = 0.701). After propensity score matching for age, gender, and diabetes mellitus between patients with and without fracture, fracture still significantly correlated with MI in HD patients (OR 1.47, P = 0.034) but not in patients with pre-dialysis CKD (OR 1.04, P = 0.751). Subgroup analysis by fracture site found that vertebral fracture was associated with MI in HD patients (OR 2.11, P = 0.024), but femoral or other site fractures were not. CONCLUSION In HD patients, fracture was significantly associated with MI, especially for vertebral fractures patients.
Collapse
Affiliation(s)
- Y E Kwon
- Department of Internal Medicine, Hanyang University College of Medicine, Myongji Hospital, Goyang-si, South Korea
| | - H Y Choi
- The Korean Society of Nephrology, Seoul, South Korea
| | - H J Oh
- Ewha Institute of Convergence Medicine and Research Institute for Human Health Information, Ewha Womans University, Seoul, South Korea
| | - S Y Ahn
- Department of Internal Medicine, Korea University College of Medicine, Seoul, South Korea
| | - D-R Ryu
- Department of Internal Medicine, Ewha Womans University College of Medicine, Seoul, South Korea.
| | - Y J Kwon
- Department of Internal Medicine, Korea University College of Medicine, Seoul, South Korea.
| |
Collapse
|
32
|
Humalda JK, Yeung SMH, Geleijnse JM, Gijsbers L, Riphagen IJ, Hoorn EJ, Rotmans JI, Vogt L, Navis G, Bakker SJL, de Borst MH. Effects of Potassium or Sodium Supplementation on Mineral Homeostasis: A Controlled Dietary Intervention Study. J Clin Endocrinol Metab 2020; 105:5854371. [PMID: 32506135 PMCID: PMC7365698 DOI: 10.1210/clinem/dgaa359] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/02/2020] [Accepted: 06/03/2020] [Indexed: 11/19/2022]
Abstract
CONTEXT Although dietary potassium and sodium intake may influence calcium-phosphate metabolism and bone health, the effects on bone mineral parameters, including fibroblast growth factor 23 (FGF23), are unclear. OBJECTIVE Here, we investigated the effects of potassium or sodium supplementation on bone mineral parameters. DESIGN, SETTING, PARTICIPANTS We performed a post hoc analysis of a dietary controlled randomized, blinded, placebo-controlled crossover trial. Prehypertensive individuals not using antihypertensive medication (n = 36) received capsules containing potassium chloride (3 g/d), sodium chloride (3 g/d), or placebo. Linear mixed-effect models were used to estimate treatment effects. RESULTS Potassium supplementation increased plasma phosphate (from 1.10 ± 0.19 to 1.15 ± 0.19 mmol/L, P = 0.004), in line with an increase in tubular maximum of phosphate reabsorption (from 0.93 ± 0.21 to 1.01 ± 0.20 mmol/L, P < 0.001). FGF23 decreased (114.3 [96.8-135.0] to 108.5 [93.5-125.9] RU/mL, P = 0.01), without change in parathyroid hormone and 25-hydroxy vitamin D3. Fractional calcium excretion decreased (from 1.25 ± 0.50 to 1.11 ± 0.46 %, P = 0.03) without change in plasma calcium. Sodium supplementation decreased both plasma phosphate (from 1.10 ± 0.19 to 1.06 ± 0.21 mmol/L, P = 0.03) and FGF23 (from 114.3 [96.8-135.0] to 108.7 [92.3-128.1] RU/mL, P = 0.02). Urinary and fractional calcium excretion increased (from 4.28 ± 1.91 to 5.45 ± 2.51 mmol/24 hours, P < 0.001, and from 1.25 ± 0.50 to 1.44 ± 0.54 %, P = 0.004, respectively). CONCLUSIONS Potassium supplementation led to a decrease in FGF23, which was accompanied by increase in plasma phosphate and decreased calcium excretion. Sodium supplementation reduced FGF23, but this was accompanied by decrease in phosphate and increase in fractional calcium excretion. Our results indicate distinct effects of potassium and sodium intake on bone mineral parameters, including FGF23. CLINICAL TRIAL REGISTRATION NUMBER NCT01575041.
Collapse
Affiliation(s)
- Jelmer K Humalda
- Department of Internal Medicine, Division of Nephrology, University of Groningen, University Medical Center Groningen, RB Groningen, the Netherlands
| | - Stanley M H Yeung
- Department of Internal Medicine, Division of Nephrology, University of Groningen, University Medical Center Groningen, RB Groningen, the Netherlands
| | - Johanna M Geleijnse
- Division of Human Nutrition and Health, Wageningen University, HB Wageningen, the Netherlands
| | - Lieke Gijsbers
- Division of Human Nutrition and Health, Wageningen University, HB Wageningen, the Netherlands
| | - Ineke J Riphagen
- Department of Laboratory Medicine, University of Groningen, University Medical Center Groningen, RB Groningen, the Netherlands
| | - Ewout J Hoorn
- Department of Internal Medicine, Division of Nephrology & Transplantation, Erasmus Medical Center, University Medical Center Rotterdam, CA Rotterdam, The Netherlands
| | - Joris I Rotmans
- Department of Internal Medicine, Leiden University Medical Center, RC Leiden, the Netherlands
| | - Liffert Vogt
- Department of Internal Medicine, Section of Nephrology, Amsterdam Cardiovascular Sciences, Amsterdam University Medical Centers, University of Amsterdam, DD Amsterdam Zuidoost, the Netherlands
| | - Gerjan Navis
- Department of Internal Medicine, Division of Nephrology, University of Groningen, University Medical Center Groningen, RB Groningen, the Netherlands
| | - Stephan J L Bakker
- Department of Internal Medicine, Division of Nephrology, University of Groningen, University Medical Center Groningen, RB Groningen, the Netherlands
| | - Martin H de Borst
- Department of Internal Medicine, Division of Nephrology, University of Groningen, University Medical Center Groningen, RB Groningen, the Netherlands
- Correspondence and Reprint Requests: Martin H. de Borst, MD, PhD, Department of Internal Medicine, Division of Nephrology, University of Groningen, University Medical Center Groningen, P.O. Box 30.001, 9700 RB Groningen, the Netherlands. E-mail:
| |
Collapse
|
33
|
Bouma-de Krijger A, Vervloet MG. Fibroblast growth factor 23: are we ready to use it in clinical practice? J Nephrol 2020; 33:509-527. [PMID: 32130720 PMCID: PMC7220896 DOI: 10.1007/s40620-020-00715-2] [Citation(s) in RCA: 25] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2020] [Accepted: 02/15/2020] [Indexed: 12/15/2022]
Abstract
Patients with chronic kidney disease (CKD) have a greatly enhanced risk of cardiovascular morbidity and mortality. Over the past decade it has come clear that a disturbed calcium-phosphate metabolism, with Fibroblast Growth Factor-23 as a key hormone, is partly accountable for this enhanced risk. Numerous studies have been performed unravelling FGF23s actions and its association with clinical conditions. As FGF23 is strongly associated with adverse outcome it may be a promising biomarker for risk prediction or, even more important, targeting FGF23 may be a strategy to improve patient outcome. This review elaborates on the clinical usefulness of FGF23 measurement. Firstly it discusses the reliability of the FGF23 measurement. Secondly, it evaluates whether FGF23 measurement may lead to improved patient risk classification. Finally, and possibly most importantly, this review evaluates if lowering of FGF23 should be a target for therapy. For this, the review discusses the current evidence indicating that FGF23 may be in the causal pathway to cardiovascular pathology, provides an overview of strategies to lower FGF23 levels and discusses the current evidence concerning the benefit of lowering FGF23.
Collapse
Affiliation(s)
- Annet Bouma-de Krijger
- Department of Nephrology, Amsterdam Cardiovascular Science, Amsterdam University Medical Center, De Boelelaan 1117, 1081 HV Amsterdam, The Netherlands
| | - Marc G. Vervloet
- Department of Nephrology, Amsterdam Cardiovascular Science, Amsterdam University Medical Center, De Boelelaan 1117, 1081 HV Amsterdam, The Netherlands
| |
Collapse
|
34
|
Gateva A, Tsakova A, Hristova J, Kamenov Z. Fibroblast growth factor 23 and 25(OH)D levels are related to abdominal obesity and cardiovascular risk in patients with polycystic ovarian syndrome. Gynecol Endocrinol 2020; 36:402-405. [PMID: 31709849 DOI: 10.1080/09513590.2019.1689550] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 10/25/2022] Open
Abstract
Fibroblast growth factor 23 (FGF23) and Klotho are extensively studied in relation to bone metabolism and progression of chronic kidney disease. There is very limited information about their role in polycystic ovarian syndrome (PCOS). The aim of the present study was to investigate some bone markers in women with PCOS in relation to obesity and cardiovascular risk. In the study were included 80 patients, divided into three age-matched groups -Non-obese PCOS (n = 40); Obese PCOS (n = 20) and Obese control group (n = 20). Bone marker levels were measured by an enzyme-linked immunosorbent assay. Obese PCOS patients had higher levels of FGF23 and sRANKL, lower levels of 25(OH)D and higher prevalence of vitamin D deficiency compared to non-obese subjects. Patients with abdominal obesity (waist circumference >80 cm) independently of PCOS status had significantly higher levels of FGF23 (112.5 ± 86.5 vs. 73.4 ± 37.9 pg/ml; p = .023) and lower of 25(OH)D (35.8 ± 21.4 vs 47.8 ± 26.5 nmol/l; p = .034). Patients with PCOS at risk of cardiovascular diseases according to AE-PCOS consensus also had increased levels of FGF23 (111.6 ± 84.5 vs. 66.5 ± 35.1 pg/ml; p = .031) and decreased levels of 25(OH)D (31.9 ± 16.8 vs. 47.1 vs 28.4 nmol/l; p = .017) compared to those not at risk. There was no correlation between bone markers and blood glucose levels, insulin resistance or hormonal levels.
Collapse
Affiliation(s)
- Antoaneta Gateva
- Department of Internal Medicine, Medical University-Sofia, Sofia, Bulgaria
- Clinic of Endocrinology, University Hospital "Alexandrovska", Sofia, Bulgaria
| | - Adelina Tsakova
- Department of Clinical Laboratory and Clinical Immunology, Medical University-Sofia, Sofia, Bulgaria
- Clinical Laboratory and Clinical Pharmacology, University Hospital "Alexandrovska", Sofia, Bulgaria
| | - Julieta Hristova
- Department of Clinical Laboratory and Clinical Immunology, Medical University-Sofia, Sofia, Bulgaria
- Clinical Laboratory and Clinical Pharmacology, University Hospital "Alexandrovska", Sofia, Bulgaria
| | - Zdravko Kamenov
- Department of Internal Medicine, Medical University-Sofia, Sofia, Bulgaria
- Clinic of Endocrinology, University Hospital "Alexandrovska", Sofia, Bulgaria
| |
Collapse
|
35
|
Mazzaferro S, De Martini N, Rotondi S, Tartaglione L, Ureña-Torres P, Bover J, Pasquali M. Bone, inflammation and chronic kidney disease. Clin Chim Acta 2020; 506:236-240. [PMID: 32275989 DOI: 10.1016/j.cca.2020.03.040] [Citation(s) in RCA: 23] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2020] [Revised: 03/27/2020] [Accepted: 03/27/2020] [Indexed: 01/04/2023]
Abstract
Increasing knowledge on inflammatory mediators and bone metabolism highlights the relationship between inflammation and bone disease. During acute illness, inflammatory cells and cytokines modulate bone cells activity so as to mobilize calcium seemingly to supply the metabolic requirements for immune response. In case of long lasting, chronic inflammatory states a condition of maladaptive, smouldering inflammation is realized and negatively affects calcium bone balance. Aging, now nicknamed inflammaging, is regarded as a chronic inflammatory condition, characterized by increased circulating inflammatory cytokines, that contributes to the development of osteoporosis, cardiovascular diseases and chronic kidney disease. In patients with renal insufficiency, the development of bone and mineral disorders (so called CKD-MBD "syndrome") is now a recognized pathogenic factor for the seemingly accelerated process of aging and for the increased risk of cardiovascular death in these patients. The adaptive changes in mineral and bone metabolism developing in the early stages of chronic kidney disease could represent a hypothetical model of accelerated aging, osteoporosis and cardiovascular disease.
Collapse
Affiliation(s)
- Sandro Mazzaferro
- Department of Translational and Precision Medicine, Sapienza University, Rome, Italy.
| | - Natalia De Martini
- Department of Translational and Precision Medicine, Sapienza University, Rome, Italy.
| | | | - Lida Tartaglione
- Department of Translational and Precision Medicine, Sapienza University, Rome, Italy.
| | - Pablo Ureña-Torres
- AURA Nord Saint Ouen, Saint Ouen, France; Department of Renal Physiology, Necker Hospital, University of Paris Descartes, Paris, France.
| | - Jordi Bover
- Fundació Puigvert, Department of Nephrology, IIB Sant Pau, Universitat Autònoma, RedinRen, Barcelona, Catalonia, Spain.
| | - Marzia Pasquali
- Azienda Ospedaliero-Universitaria Policlinico Umberto I, Roma, Italy.
| | | |
Collapse
|
36
|
Law JP, Price AM, Pickup L, Radhakrishnan A, Weston C, Jones AM, McGettrick HM, Chua W, Steeds RP, Fabritz L, Kirchhof P, Pavlovic D, Townend JN, Ferro CJ. Clinical Potential of Targeting Fibroblast Growth Factor-23 and αKlotho in the Treatment of Uremic Cardiomyopathy. J Am Heart Assoc 2020; 9:e016041. [PMID: 32212912 PMCID: PMC7428638 DOI: 10.1161/jaha.120.016041] [Citation(s) in RCA: 21] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
Chronic kidney disease is highly prevalent, affecting 10% to 15% of the adult population worldwide and is associated with increased cardiovascular morbidity and mortality. As chronic kidney disease worsens, a unique cardiovascular phenotype develops characterized by heart muscle disease, increased arterial stiffness, atherosclerosis, and hypertension. Cardiovascular risk is multifaceted, but most cardiovascular deaths in patients with advanced chronic kidney disease are caused by heart failure and sudden cardiac death. While the exact drivers of these deaths are unknown, they are believed to be caused by uremic cardiomyopathy: a specific pattern of myocardial hypertrophy, fibrosis, with both diastolic and systolic dysfunction. Although the pathogenesis of uremic cardiomyopathy is likely to be multifactorial, accumulating evidence suggests increased production of fibroblast growth factor-23 and αKlotho deficiency as potential major drivers of cardiac remodeling in patients with uremic cardiomyopathy. In this article we review the increasing understanding of the physiology and clinical aspects of uremic cardiomyopathy and the rapidly increasing knowledge of the biology of both fibroblast growth factor-23 and αKlotho. Finally, we discuss how dissection of these pathological processes is aiding the development of therapeutic options, including small molecules and antibodies, directly aimed at improving the cardiovascular outcomes of patients with chronic kidney disease and end-stage renal disease.
Collapse
Affiliation(s)
- Jonathan P. Law
- Birmingham Cardio‐Renal GroupUniversity Hospitals BirminghamUniversity of BirminghamUnited Kingdom
- Institute of Cardiovascular SciencesUniversity of BirminghamUnited Kingdom
- Department of NephrologyUniversity Hospitals Birmingham NHS Foundation TrustBirminghamUnited Kingdom
| | - Anna M. Price
- Birmingham Cardio‐Renal GroupUniversity Hospitals BirminghamUniversity of BirminghamUnited Kingdom
- Institute of Cardiovascular SciencesUniversity of BirminghamUnited Kingdom
- Department of NephrologyUniversity Hospitals Birmingham NHS Foundation TrustBirminghamUnited Kingdom
| | - Luke Pickup
- Birmingham Cardio‐Renal GroupUniversity Hospitals BirminghamUniversity of BirminghamUnited Kingdom
- Institute of Cardiovascular SciencesUniversity of BirminghamUnited Kingdom
| | - Ashwin Radhakrishnan
- Birmingham Cardio‐Renal GroupUniversity Hospitals BirminghamUniversity of BirminghamUnited Kingdom
| | - Chris Weston
- Institute of Immunology and ImmunotherapyUniversity of BirminghamUnited Kingdom
- NIHR Birmingham Biomedical Research CentreUniversity Hospitals Birmingham NHS Foundation Trust and University of BirminghamUnited Kingdom
| | - Alan M. Jones
- School of PharmacyUniversity of BirminghamUnited Kingdom
| | | | - Winnie Chua
- Birmingham Cardio‐Renal GroupUniversity Hospitals BirminghamUniversity of BirminghamUnited Kingdom
- Institute of Cardiovascular SciencesUniversity of BirminghamUnited Kingdom
| | - Richard P. Steeds
- Birmingham Cardio‐Renal GroupUniversity Hospitals BirminghamUniversity of BirminghamUnited Kingdom
- Institute of Cardiovascular SciencesUniversity of BirminghamUnited Kingdom
- Department of CardiologyUniversity Hospitals Birmingham NHS Foundation TrustBirminghamUnited Kingdom
| | - Larissa Fabritz
- Birmingham Cardio‐Renal GroupUniversity Hospitals BirminghamUniversity of BirminghamUnited Kingdom
- Institute of Cardiovascular SciencesUniversity of BirminghamUnited Kingdom
- Department of CardiologyUniversity Hospitals Birmingham NHS Foundation TrustBirminghamUnited Kingdom
| | - Paulus Kirchhof
- Birmingham Cardio‐Renal GroupUniversity Hospitals BirminghamUniversity of BirminghamUnited Kingdom
- Institute of Cardiovascular SciencesUniversity of BirminghamUnited Kingdom
| | - Davor Pavlovic
- Birmingham Cardio‐Renal GroupUniversity Hospitals BirminghamUniversity of BirminghamUnited Kingdom
- Institute of Cardiovascular SciencesUniversity of BirminghamUnited Kingdom
| | - Jonathan N. Townend
- Birmingham Cardio‐Renal GroupUniversity Hospitals BirminghamUniversity of BirminghamUnited Kingdom
- Institute of Cardiovascular SciencesUniversity of BirminghamUnited Kingdom
- Department of CardiologyUniversity Hospitals Birmingham NHS Foundation TrustBirminghamUnited Kingdom
| | - Charles J. Ferro
- Birmingham Cardio‐Renal GroupUniversity Hospitals BirminghamUniversity of BirminghamUnited Kingdom
- Institute of Cardiovascular SciencesUniversity of BirminghamUnited Kingdom
- Department of NephrologyUniversity Hospitals Birmingham NHS Foundation TrustBirminghamUnited Kingdom
| |
Collapse
|
37
|
Hawley S, Shaw NJ, Delmestri A, Prieto-Alhambra D, Cooper C, Pinedo-Villanueva R, Javaid MK. Prevalence and Mortality of Individuals With X-Linked Hypophosphatemia: A United Kingdom Real-World Data Analysis. J Clin Endocrinol Metab 2020; 105:5626435. [PMID: 31730177 PMCID: PMC7025948 DOI: 10.1210/clinem/dgz203] [Citation(s) in RCA: 41] [Impact Index Per Article: 8.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/15/2019] [Accepted: 11/14/2019] [Indexed: 12/11/2022]
Abstract
BACKGROUND X-linked hypophosphatemia (XLH) is a rare multisystemic disease with a prominent musculoskeletal phenotype. We aim here to improve understanding of the prevalence of XLH across the life course and of overall survival among people with XLH. METHODS This was a population-based cohort study using a large primary care database in the United Kingdom (UK) from 1995 to 2016. XLH cases were matched by age, gender, and practice to up to 4 controls. Trends in prevalence over the study period were estimated (stratified by age) and survival among cases was compared with that of controls. FINDINGS From 522 potential cases, 122 (23.4%) were scored as at least possible XLH, while 62 (11.9%) were classified as highly likely or likely (conservative definition). In main analyses, prevalence (95% CI) increased from 3.1 (1.5-6.7) per million in 1995-1999 to 14.0 (10.8-18.1) per million in 2012-2016. Corresponding estimates using the conservative definition were 3.0 (1.4-6.5) to 8.1 (5.8-11.4). Nine (7.4%) of the possible cases died during follow-up, at median age of 64 years. Fourteen (2.9%) of the controls died at median age of 72.5 years. Mortality was significantly increased in those with possible XLH compared with controls (hazard ratio [HR] 2.93; 95% CI, 1.24-6.91). Likewise, among those with likely or highly likely XLH (HR 6.65; 1.44-30.72). CONCLUSIONS We provide conservative estimates of the prevalence of XLH in children and adults within the UK. There was an unexpected increase in mortality in later life, which may have implications for other fibroblast growth factor 23-related disorders.
Collapse
Affiliation(s)
- Samuel Hawley
- Nuffield Department of Orthopaedics, Rheumatology and Musculoskeletal Sciences, University of Oxford, Oxford, UK
| | - Nick J Shaw
- Birmingham Women’s and Children’s Hospital NHS Foundation Trust, Birmingham, UK
- Institute of Metabolism & Systems Research, University of Birmingham, Birmingham, UK
| | - Antonella Delmestri
- Nuffield Department of Orthopaedics, Rheumatology and Musculoskeletal Sciences, University of Oxford, Oxford, UK
| | - Daniel Prieto-Alhambra
- Nuffield Department of Orthopaedics, Rheumatology and Musculoskeletal Sciences, University of Oxford, Oxford, UK
- GREMPAL Research Group, Idiap Jordi Gol and CIBERFes, Universitat Autònoma de Barcelona and Instituto de Salud Carlos III, Barcelona, Spain
| | - Cyrus Cooper
- MRC Lifecourse Epidemiology Unit, University of Southampton, Southampton, UK
| | - Rafael Pinedo-Villanueva
- Nuffield Department of Orthopaedics, Rheumatology and Musculoskeletal Sciences, University of Oxford, Oxford, UK
| | - M Kassim Javaid
- Nuffield Department of Orthopaedics, Rheumatology and Musculoskeletal Sciences, University of Oxford, Oxford, UK
- MRC Lifecourse Epidemiology Unit, University of Southampton, Southampton, UK
- Correspondence: Muhammad Kassim Javaid, MBBS, BMedSci, FRCP. PhD, The Botnar Research Centre, NDORMS, University of Oxford, Windmill Road, Oxford, OX3 7YD, UK. E-mail:
| |
Collapse
|
38
|
Han X, Cai C, Xiao Z, Quarles LD. FGF23 induced left ventricular hypertrophy mediated by FGFR4 signaling in the myocardium is attenuated by soluble Klotho in mice. J Mol Cell Cardiol 2020; 138:66-74. [PMID: 31758962 PMCID: PMC7195870 DOI: 10.1016/j.yjmcc.2019.11.149] [Citation(s) in RCA: 49] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/31/2019] [Revised: 11/12/2019] [Accepted: 11/14/2019] [Indexed: 12/15/2022]
Abstract
There is controversy regarding whether excess FGF23 causes left ventricular hypertrophy (LVH) directly through activation of fibroblast growth factor receptor 4 (FGFR4) in cardiomyocytes or indirectly through reductions in soluble Klotho (sK). We investigated the respective roles of myocardial FGFR4 and sKL in mediating FGF23-induced LVH using mouse genetic and pharmacological approaches. To investigate a direct role of myocardial FGFR4 in mediating the cardiotoxic effects of excess circulating FGF23, we administered rFGF23 to mice with cardiac-specific loss of FGFR4 (FGFR4 heart-cKO). We tested a model of sKL deficiency, hypertension and LVH created by the conditional deletion of FGFR1 in the renal distal tubule (FGFR1DT cKO mice). The cardioprotective effects of sKL in both mouse models was assessed by the systemic administration of recombinant sKL. We confirmed that FGF23 treatment activates PLCγ in the heart and induces LVH in the absence of membrane α-Klotho. Conditional deletion of FGFR4 in the myocardium prevented rFGF23-induced LVH in mice, establishing direct cardiotoxicity of FGF23 through activation of FGFR4. Recombinant sKL administration prevented LVH, but not HTN, in FGFR1DT cKO mice, consistent with direct cardioprotective effects. Co-administration of recombinant sKL with FGF23 in culture inhibited rFGF23-induced p-PLCγ signaling. Thus, FGF23 ability to include LVH represents a balance between FGF23 direct cardiac activation of FGFR4 and the modulating effects of circulating sKL to alter FGF23-dependent myocardial signaling pathways.
Collapse
Affiliation(s)
- Xiaobin Han
- Department of Medicine, University of Tennessee Health Science Center, Memphis, TN 38163, United States of America
| | - Chun Cai
- Department of Medicine, University of Tennessee Health Science Center, Memphis, TN 38163, United States of America
| | - Zhousheng Xiao
- Department of Medicine, University of Tennessee Health Science Center, Memphis, TN 38163, United States of America
| | - L Darryl Quarles
- Department of Medicine, University of Tennessee Health Science Center, Memphis, TN 38163, United States of America.
| |
Collapse
|
39
|
Xiao Z, King G, Mancarella S, Munkhsaikhan U, Cao L, Cai C, Quarles LD. FGF23 expression is stimulated in transgenic α-Klotho longevity mouse model. JCI Insight 2019; 4:132820. [PMID: 31801907 PMCID: PMC6962016 DOI: 10.1172/jci.insight.132820] [Citation(s) in RCA: 34] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2019] [Accepted: 10/23/2019] [Indexed: 12/21/2022] Open
Abstract
Observations in transgenic α-Klotho (Kl) mice (KlTg) defined the antiaging role of soluble Klotho (sKL130). A genetic translocation that elevates sKL levels in humans is paradoxically associated with increased circulating fibroblast growth factor 23 (FGF23) levels and the potential of both membrane KL (mKL135) and sKL130 to act as coreceptors for FGF23 activation of fibroblast growth factor receptors (FGFRs). Neither FGF23 expression nor the contributions of FGF23, mKL135, and sKL130 codependent and independent functions have been investigated in KlTg mice. In the current study, we examined the effects of Kl overexpression on FGF23 levels and functions in KlTg mice. We found that mKL135 but not sKL130 stimulated FGF23 expression in osteoblasts, leading to elevated Fgf23 bone expression and circulating levels in KlTg mice. Elevated FGF23 suppressed 1,25(OH)2D and parathyroid hormone levels but did not cause hypophosphatemic rickets in KlTg mice. KlTg mice developed low aldosterone-associated hypertension but not left ventricular hypertrophy. Mechanistically, we found that mKL135 and sKL130 are essential cofactors for FGF23-mediated ERK activation but that they inhibited FGF23 stimulation of PLC-γ and PI3K/AKT signaling. Thus, increased longevity in KlTg mice occurs in the presence of excess FGF23 that interacts with mKL and sKL to bias FGFR pathways.
Collapse
Affiliation(s)
- Zhousheng Xiao
- Department of Medicine, University of Tennessee Health Science Center, Memphis, Tennessee, USA
| | - Gwendalyn King
- Department of Biology, Creighton University, Omaha, Nebraska, USA
| | | | - Undral Munkhsaikhan
- Department of Pediatrics, University of Tennessee Health Science Center, Memphis, Tennessee, USA
| | - Li Cao
- Department of Medicine, University of Tennessee Health Science Center, Memphis, Tennessee, USA
| | - Chun Cai
- Department of Medicine, University of Tennessee Health Science Center, Memphis, Tennessee, USA
| | - Leigh Darryl Quarles
- Department of Medicine, University of Tennessee Health Science Center, Memphis, Tennessee, USA
| |
Collapse
|
40
|
Wagner CA, Rubio-Aliaga I, Egli-Spichtig D. Fibroblast growth factor 23 in chronic kidney disease: what is its role in cardiovascular disease? Nephrol Dial Transplant 2019; 34:1986-1990. [PMID: 30903187 DOI: 10.1093/ndt/gfz044] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2018] [Accepted: 02/10/2019] [Indexed: 11/14/2022] Open
Affiliation(s)
- Carsten A Wagner
- National Center of Competence in Research Kidney, CH, Institute of Physiology, University of Zurich, Zurich, Switzerland
| | - Isabel Rubio-Aliaga
- National Center of Competence in Research Kidney, CH, Institute of Physiology, University of Zurich, Zurich, Switzerland
| | - Daniela Egli-Spichtig
- National Center of Competence in Research Kidney, CH, Institute of Physiology, University of Zurich, Zurich, Switzerland
| |
Collapse
|
41
|
Pickup L, Radhakrishnan A, Townend JN, Ferro CJ. Arterial stiffness in chronic kidney disease. Curr Opin Nephrol Hypertens 2019; 28:527-536. [DOI: 10.1097/mnh.0000000000000535] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
|
42
|
Eisenga MF, Emans ME, van der Putten K, Cramer MJ, Diepenbroek A, Velthuis BK, Doevendans PA, Verhaar MC, Joles JA, Bakker SJL, Nolte IM, Braam B, Gaillard CAJM. Epoetin Beta and C-Terminal Fibroblast Growth Factor 23 in Patients With Chronic Heart Failure and Chronic Kidney Disease. J Am Heart Assoc 2019; 8:e011130. [PMID: 31423921 PMCID: PMC6759901 DOI: 10.1161/jaha.118.011130] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
Background In patients with chronic heart failure and chronic kidney disease, correction of anemia with erythropoietin-stimulating agents targeting normal hemoglobin levels is associated with an increased risk of cardiovascular morbidity and mortality. Emerging data suggest a direct effect of erythropoietin on fibroblast growth factor 23 (FGF23), elevated levels of which have been associated with adverse outcomes. We investigate effects of erythropoietin-stimulating agents in patients with both chronic heart failure and chronic kidney disease focusing on FGF23. Methods and Results In the EPOCARES (Erythropoietin in CardioRenal Syndrome) study, we randomized 56 anemic patients (median age 74 [interquartile range 69-80] years, 66% male) with both chronic heart failure and chronic kidney disease into 3 groups, of which 2 received epoetin beta 50 IU/kg per week for 50 weeks, and the third group served as control. Measurements were performed at baseline and after 2, 26, and 50 weeks. Data were analyzed using linear mixed-model analysis. After 50 weeks of erythropoietin-stimulating agent treatment, hematocrit and hemoglobin levels increased. Similarly, C-terminal FGF23 levels, in contrast to intact FGF23 levels, rose significantly due to erythropoietin-stimulating agents as compared with the controls. During median follow-up for 5.7 (2.0-5.7) years, baseline C-terminal FGF23 levels were independently associated with increased risk of mortality (hazard ratio 2.20; 95% CI, 1.35-3.59; P=0.002). Conclusions Exogenous erythropoietin increases C-terminal FGF23 levels markedly over a period of 50 weeks, elevated levels of which, even at baseline, are significantly associated with an increased risk of mortality. The current results, in a randomized trial setting, underline the strong relationship between erythropoietin and FGF23 physiology in patients with chronic heart failure and chronic kidney disease. Clinical Trial Registration URL: http://www.clinicaltrials.gov. Unique identifier: NCT00356733.
Collapse
Affiliation(s)
- Michele F Eisenga
- Division of Nephrology Department of Internal Medicine University of Groningen University Medical Center Groningen Groningen the Netherlands
| | - Mireille E Emans
- Department of Cardiology Ikazia Hospital Rotterdam the Netherlands
| | | | - Maarten J Cramer
- Department of Cardiology University of Utrecht University Medical Center Utrecht Utrecht the Netherlands
| | - Adry Diepenbroek
- Division of Nephrology Department of Internal Medicine University of Groningen University Medical Center Groningen Groningen the Netherlands
| | - Birgitta K Velthuis
- Department of Radiology University of Utrecht University Medical Center Utrecht Utrecht the Netherlands
| | - Pieter A Doevendans
- Department of Cardiology University of Utrecht University Medical Center Utrecht Utrecht the Netherlands
| | - Marianne C Verhaar
- Department of Nephrology and Hypertension University of Utrecht University Medical Center Utrecht Utrecht the Netherlands
| | - Jaap A Joles
- Department of Nephrology and Hypertension University of Utrecht University Medical Center Utrecht Utrecht the Netherlands
| | - Stephan J L Bakker
- Division of Nephrology Department of Internal Medicine University of Groningen University Medical Center Groningen Groningen the Netherlands
| | - Ilja M Nolte
- Department of Epidemiology University of Groningen University Medical Center Groningen Groningen the Netherlands
| | - Branko Braam
- Division of Nephrology and Immunology Department of Medicine University of Alberta Edmonton Canada
| | - Carlo A J M Gaillard
- Department of Internal Medicine and Dermatology University of Utrecht University Medical Center Utrecht Utrecht the Netherlands
| |
Collapse
|
43
|
Eisenga MF, De Jong MA, Van der Meer P, Leaf DE, Huls G, Nolte IM, Gaillard CAJM, Bakker SJL, De Borst MH. Iron deficiency, elevated erythropoietin, fibroblast growth factor 23, and mortality in the general population of the Netherlands: A cohort study. PLoS Med 2019; 16:e1002818. [PMID: 31170159 PMCID: PMC6553711 DOI: 10.1371/journal.pmed.1002818] [Citation(s) in RCA: 20] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/04/2019] [Accepted: 05/02/2019] [Indexed: 01/12/2023] Open
Abstract
BACKGROUND Emerging data in chronic kidney disease (CKD) patients suggest that iron deficiency and higher circulating levels of erythropoietin (EPO) stimulate the expression and concomitant cleavage of the osteocyte-derived, phosphate-regulating hormone fibroblast growth factor 23 (FGF23), a risk factor for premature mortality. To date, clinical implications of iron deficiency and high EPO levels in the general population, and the potential downstream role of FGF23, are unclear. Therefore, we aimed to determine the associations between iron deficiency and higher EPO levels with mortality, and the potential mediating role of FGF23, in a cohort of community-dwelling subjects. METHODS AND FINDINGS We analyzed 6,544 community-dwelling subjects (age 53 ± 12 years; 50% males) who participated in the Prevention of Renal and Vascular End-Stage Disease (PREVEND) study-a prospective population-based cohort study, of which we used the second survey (2001-2003)-and follow-up was performed for a median of 8 years. We measured circulating parameters of iron status, EPO levels, and plasma total FGF23 levels. Our primary outcome was all-cause mortality. In multivariable linear regression analyses, ferritin (ß = -0.43), transferrin saturation (TSAT) (ß = -0.17), hepcidin (ß = -0.36), soluble transferrin receptor (sTfR; ß = 0.33), and EPO (ß = 0.28) were associated with FGF23 level, independent of potential confounders. During median (interquartile range [IQR]) follow-up of 8.2 (7.7-8.8) years, 379 (6%) subjects died. In multivariable Cox regression analyses, lower levels of TSAT (hazard ratio [HR] per 1 standard deviation [SD], 0.84; 95% confidence interval [CI], 0.75-0.95; P = 0.004) and higher levels of sTfR (HR, 1.15; 95% CI 1.03-1.28; P = 0.01), EPO (HR, 1.17; 95% CI 1.05-1.29; P = 0.004), and FGF23 (HR, 1.20; 95% CI 1.10-1.32; P < 0.001) were each significantly associated with an increased risk of death, independent of potential confounders. Adjustment for FGF23 levels markedly attenuated the associations of TSAT (HR, 0.89; 95% CI 0.78-1.01; P = 0.06), sTfR (HR, 1.08; 95% CI 0.96-1.20; P = 0.19), and EPO (HR, 1.10; 95% CI 0.99-1.22; P = 0.08) with mortality. FGF23 remained associated with mortality (HR, 1.15; 95% CI 1.04-1.27; P = 0.008) after adjustment for TSAT, sTfR, and EPO levels. Mediation analysis indicated that FGF23 explained 31% of the association between TSAT and mortality; similarly, FGF23 explained 32% of the association between sTfR and mortality and 48% of the association between EPO and mortality (indirect effect P < 0.05 for all analyses). The main limitations of this study were the observational study design and the absence of data on intact FGF23 (iFGF23), precluding us from discerning whether the current results are attributable to an increase in iFGF23 or in C-terminal FGF23 fragments. CONCLUSIONS AND RELEVANCE In this study, we found that functional iron deficiency and higher EPO levels were each associated with an increased risk of death in the general population. Our findings suggest that FGF23 could be involved in the association between functional iron deficiency and increased EPO levels and death. Investigation of strategies aimed at correcting iron deficiency and reducing FGF23 levels is warranted.
Collapse
Affiliation(s)
- Michele F. Eisenga
- Division of Nephrology, Department of Internal Medicine, University of Groningen, University Medical Center Groningen, Groningen, the Netherlands
- * E-mail:
| | - Maarten A. De Jong
- Division of Nephrology, Department of Internal Medicine, University of Groningen, University Medical Center Groningen, Groningen, the Netherlands
| | - Peter Van der Meer
- Department of Cardiology, University of Groningen, University Medical Center Groningen, Groningen, the Netherlands
| | - David E. Leaf
- Division of Renal Medicine, Brigham and Women’s Hospital, Harvard Medical School, Boston, Massachusetts, United States of America
| | - Gerwin Huls
- Division of Hematology, Department of Internal Medicine, University of Groningen, University Medical Center Groningen, Groningen, the Netherlands
| | - Ilja M. Nolte
- Department of Epidemiology, University of Groningen, University Medical Center Groningen, Groningen, the Netherlands
| | - Carlo A. J. M. Gaillard
- Department of Internal Medicine and Dermatology, University of Utrecht, University Medical Center Utrecht, Utrecht, the Netherlands
| | - Stephan J. L. Bakker
- Division of Nephrology, Department of Internal Medicine, University of Groningen, University Medical Center Groningen, Groningen, the Netherlands
| | - Martin H. De Borst
- Division of Nephrology, Department of Internal Medicine, University of Groningen, University Medical Center Groningen, Groningen, the Netherlands
| |
Collapse
|
44
|
Associations of plasma fibroblast growth factor 23 and other markers of chronic kidney disease-Mineral and bone disorder with all-cause mortality in South African patients on maintenance dialysis: A 3-year prospective cohort study. PLoS One 2019; 14:e0216656. [PMID: 31107896 PMCID: PMC6527219 DOI: 10.1371/journal.pone.0216656] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2018] [Accepted: 04/25/2019] [Indexed: 12/30/2022] Open
Abstract
INTRODUCTION Few studies have linked high levels of plasma C-terminal fibroblast growth factor 23 (FGF23) with poor clinical outcomes in patients on maintenance haemodialysis (MHD), while the association between intact FGF23 and mortality in this group of patients remains inconclusive. Therefore, the aim of this study was to evaluate the association between plasma levels of intact FGF23 and mortality in dialysis patients. METHODS A prospective multicenter study involving patients undergoing dialysis at three dialysis centers in Johannesburg was undertaken between 1st October 2014 and 31st December 2017. RESULTS The study comprised 165 chronic dialysis patients (111 blacks, 54 whites) with a mean age of 46.6 ±14.2 years. During a three year follow up period, there were 46 deaths (1.03 per 100 person-years). The median plasma FGF 23 level was 382 pg/ml (interquartile range [IQR], 145-2977). In adjusted multivariable analyses, there was a non-statistically significant increase in the risk of mortality with higher quartiles of FGF 23 levels: the adjusted hazard ratios (HR) for the second, third and fourth quantiles were HR 3.20 (95% CI, 0.99-10.35; P = 0.052), HR 2.43(95% CI,0.65-9.09; P = 0.19), and HR 2.09 (95% CI, 0.66-7.32; P = 0.25),respectively. Corrected serum calcium 2.38-2.5 mmol/l [HR 2.98 (95% CI, 1.07-8.29; P = 0.04] and > 2.50 mmol/l [HR 5.50 (95% CI, 1.84-16.48; P = 0.002] were independently associated with increased risk of death. Likewise, patients with intact parathyroid hormone > 600 pg/ml had a 3.46-fold higher risk of death (HR 3.46, 95% CI, 1.22-9.82 P = 0.019). These findings persisted in time -dependent analyses. CONCLUSION Higher levels of intact FGF 23 appear not to be independently associated with all-cause mortality in our dialysis patients, while hypercalcaemia and severe hyperparathyroidism were found to be independent predictors of mortality in this cohort of patients.
Collapse
|
45
|
Egli-Spichtig D, Imenez Silva PH, Glaudemans B, Gehring N, Bettoni C, Zhang MYH, Pastor-Arroyo EM, Schönenberger D, Rajski M, Hoogewijs D, Knauf F, Misselwitz B, Frey-Wagner I, Rogler G, Ackermann D, Ponte B, Pruijm M, Leichtle A, Fiedler GM, Bochud M, Ballotta V, Hofmann S, Perwad F, Föller M, Lang F, Wenger RH, Frew I, Wagner CA. Tumor necrosis factor stimulates fibroblast growth factor 23 levels in chronic kidney disease and non-renal inflammation. Kidney Int 2019; 96:890-905. [PMID: 31301888 DOI: 10.1016/j.kint.2019.04.009] [Citation(s) in RCA: 57] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2018] [Revised: 03/11/2019] [Accepted: 04/05/2019] [Indexed: 01/03/2023]
Abstract
Fibroblast growth factor 23 (FGF23) regulates phosphate homeostasis, and its early rise in patients with chronic kidney disease is independently associated with all-cause mortality. Since inflammation is characteristic of chronic kidney disease and associates with increased plasma FGF23 we examined whether inflammation directly stimulates FGF23. In a population-based cohort, plasma tumor necrosis factor (TNF) was the only inflammatory cytokine that independently and positively correlated with plasma FGF23. Mouse models of chronic kidney disease showed signs of renal inflammation, renal FGF23 expression and elevated systemic FGF23 levels. Renal FGF23 expression coincided with expression of the orphan nuclear receptor Nurr1 regulating FGF23 in other organs. Antibody-mediated neutralization of TNF normalized plasma FGF23 and suppressed ectopic renal Fgf23 expression. Conversely, TNF administration to control mice increased plasma FGF23 without altering plasma phosphate. Moreover, in Il10-deficient mice with inflammatory bowel disease and normal kidney function, plasma FGF23 was elevated and normalized upon TNF neutralization. Thus, the inflammatory cytokine TNF contributes to elevated systemic FGF23 levels and also triggers ectopic renal Fgf23 expression in animal models of chronic kidney disease.
Collapse
Affiliation(s)
- Daniela Egli-Spichtig
- Institute of Physiology, University of Zurich, Zurich, Switzerland; Swiss National Center of Competence in Research NCCR-Kidney.CH, University of Zurich, Zurich, Switzerland; Department of Pediatrics, Division of Nephrology, University of California, San Francisco, San Francisco, California, USA
| | - Pedro Henrique Imenez Silva
- Institute of Physiology, University of Zurich, Zurich, Switzerland; Swiss National Center of Competence in Research NCCR-Kidney.CH, University of Zurich, Zurich, Switzerland
| | - Bob Glaudemans
- Institute of Physiology, University of Zurich, Zurich, Switzerland; Swiss National Center of Competence in Research NCCR-Kidney.CH, University of Zurich, Zurich, Switzerland
| | - Nicole Gehring
- Institute of Physiology, University of Zurich, Zurich, Switzerland; Swiss National Center of Competence in Research NCCR-Kidney.CH, University of Zurich, Zurich, Switzerland
| | - Carla Bettoni
- Institute of Physiology, University of Zurich, Zurich, Switzerland; Swiss National Center of Competence in Research NCCR-Kidney.CH, University of Zurich, Zurich, Switzerland
| | - Martin Y H Zhang
- Department of Pediatrics, Division of Nephrology, University of California, San Francisco, San Francisco, California, USA
| | - Eva M Pastor-Arroyo
- Institute of Physiology, University of Zurich, Zurich, Switzerland; Swiss National Center of Competence in Research NCCR-Kidney.CH, University of Zurich, Zurich, Switzerland
| | - Désirée Schönenberger
- Institute of Physiology, University of Zurich, Zurich, Switzerland; Swiss National Center of Competence in Research NCCR-Kidney.CH, University of Zurich, Zurich, Switzerland
| | - Michal Rajski
- Institute of Physiology, University of Zurich, Zurich, Switzerland; Swiss National Center of Competence in Research NCCR-Kidney.CH, University of Zurich, Zurich, Switzerland
| | - David Hoogewijs
- Institute of Physiology, University of Zurich, Zurich, Switzerland; Swiss National Center of Competence in Research NCCR-Kidney.CH, University of Zurich, Zurich, Switzerland
| | - Felix Knauf
- Division of Nephrology, Charité - Universitätsmedizin Berlin, Berlin, Germany
| | - Benjamin Misselwitz
- University Hospital Zurich, Clinic for Gastroenterology and Hepatology, Zurich, Switzerland
| | - Isabelle Frey-Wagner
- University Hospital Zurich, Clinic for Gastroenterology and Hepatology, Zurich, Switzerland
| | - Gerhard Rogler
- University Hospital Zurich, Clinic for Gastroenterology and Hepatology, Zurich, Switzerland
| | - Daniel Ackermann
- Department of Nephrology and Hypertension, Inselspital, Bern University Hospital, University of Bern, Bern, Switzerland
| | - Belen Ponte
- Department of Nephrology, University Hospital of Geneva (HUG), Geneva, Switzerland
| | - Menno Pruijm
- Department of Nephrology, Lausanne University Hospital (CHUV), Lausanne, Switzerland
| | - Alexander Leichtle
- Institute of Clinical Chemistry, Inselspital, Bern University Hospital, University of Bern, Bern, Switzerland
| | - Georg-Martin Fiedler
- Institute of Clinical Chemistry, Inselspital, Bern University Hospital, University of Bern, Bern, Switzerland
| | - Murielle Bochud
- Swiss National Center of Competence in Research NCCR-Kidney.CH, University of Zurich, Zurich, Switzerland; Institute of Social and Preventive Medicine (IUMSP), Lausanne University Hospital (CHUV), Lausanne, Switzerland
| | - Virginia Ballotta
- Department of Biomedical Engineering and Institute for Complex Molecular Systems, Eindhoven University of Technology, Eindhoven, The Netherlands
| | - Sandra Hofmann
- Department of Biomedical Engineering and Institute for Complex Molecular Systems, Eindhoven University of Technology, Eindhoven, The Netherlands
| | - Farzana Perwad
- Department of Pediatrics, Division of Nephrology, University of California, San Francisco, San Francisco, California, USA
| | - Michael Föller
- Institute of Physiology, University of Hohenheim, Stuttgart, Germany
| | - Florian Lang
- Institute of Physiology I, University of Tübingen, Tübingen, Germany
| | - Roland H Wenger
- Institute of Physiology, University of Zurich, Zurich, Switzerland; Swiss National Center of Competence in Research NCCR-Kidney.CH, University of Zurich, Zurich, Switzerland
| | - Ian Frew
- Institute of Physiology, University of Zurich, Zurich, Switzerland; Swiss National Center of Competence in Research NCCR-Kidney.CH, University of Zurich, Zurich, Switzerland
| | - Carsten A Wagner
- Institute of Physiology, University of Zurich, Zurich, Switzerland; Swiss National Center of Competence in Research NCCR-Kidney.CH, University of Zurich, Zurich, Switzerland.
| |
Collapse
|
46
|
Kim SH, Kim JW, Hwang IG, Jang JS, Hong S, Kim TY, Baek JY, Shin SH, Sun DS, Hong DS, Kim HJ, Hong YS, Woo IS, Lee JH, Kim JH. Serum biomarkers for predicting overall survival and early mortality in older patients with metastatic solid tumors. J Geriatr Oncol 2019; 10:749-756. [PMID: 30952517 DOI: 10.1016/j.jgo.2019.03.015] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2018] [Revised: 12/20/2018] [Accepted: 03/22/2019] [Indexed: 01/07/2023]
Abstract
OBJECTIVES We aimed to explore serum biomarkers for predicting survival of older patients with metastatic solid tumors who received first line palliative chemotherapy. MATERIALS AND METHODS Serum samples were prospectively collected before first-line chemotherapy at 11 academic centers in Korea. All patients were participants in a prospective cohort study of older patients with metastatic solid tumors. Serum levels of C-reactive protein (CRP), CXCL10, SIRT1, VEGF-A, activin A, C-terminal telopeptide of type I collagen (CTx), total 25-hydroxyvitamin D were measured by ELISA and interleukin-6 (IL-6), myostatin, irisin, FGF-19, FGF-21, FGF-23 by Luminex multiplex assay. Overall survival (OS) was determined. RESULTS Serum samples from 138 patients (median age: 75 years, range: 70-92 years) were collected from February 2014 to December 2016. During a median follow up time of 13.8 months, 73 (52.9%) patients died. Among 13 serum markers, CRP (log-rank, P = 0.009), activin A (P = 0.007), and myostatin (P = 0.047) were significantly correlated with OS in univariate analyses. Activin A (hazard ratio [HR] 2.22, 95% confidence interval [CI] 1.32-3.72; P = 0.003) and myostatin (HR 3.02, 95% CI 1.39-6.57; P = 0.005) were significantly associated with OS after adjustment for other clinical factors. In predicting early (6-month) mortality, two inflammatory markers, IL-6 and CRP, were included in the decision-tree model. CONCLUSION In older patients with cancer, high serum concentrations of activin A and myostatin were predictive of poor OS. IL-6 and CRP might be useful to select older patients at risk of early mortality. These markers could be incorporated into predictive tools for clinical decision-making and warrant further investigation.
Collapse
Affiliation(s)
- Se Hyun Kim
- Department of Internal Medicine, Seoul National University Bundang Hospital, Seongnam, Republic of Korea
| | - Jin Won Kim
- Department of Internal Medicine, Seoul National University Bundang Hospital, Seongnam, Republic of Korea
| | - In Gyu Hwang
- Diveision of Hematology/Oncology, Department of Internal Medicine, Chung-Ang University Hospital, Chung-Ang University College of Medicine, Seoul, Republic of Korea
| | - Joung Soon Jang
- Diveision of Hematology/Oncology, Department of Internal Medicine, Chung-Ang University Hospital, Chung-Ang University College of Medicine, Seoul, Republic of Korea
| | - Soojung Hong
- Departmenet of Internal Medicine, National Health Insurance Service Ilsan Hospital, Goyang, Republic of Korea
| | - Tae-Yong Kim
- Department of Internal Medicine, Seoul National University Hospital, Seoul, Republic of Korea
| | - Ji Yeon Baek
- Center for Colorectal Cancer, National Cancer Center, Goyang, Republic of Korea
| | - Seong Hoon Shin
- Department of Internal Medicine, Kosin University Gospel Hospital, Busan, Republic of Korea
| | - Der Sheng Sun
- Department of Internal Medicine, Uijeongbu St. Mary's Hospital, The Catholic University of Korea, Republic of Korea
| | - Dae-Sik Hong
- Department of Internal Medicine, Soonchunhyang University Hospital, Bucheon, Republic of Korea
| | - Hyo Jung Kim
- Department of Internal Medicine, Hallym University Sacred Heart Hospital, Anyang, Republic of Korea
| | - Yong Sang Hong
- Department of Oncology, Asan Medical Center, Seoul, Republic of Korea
| | - In Sook Woo
- Department of Internal Medicine, Yeouido St.Mary's Hospital, College of Medicine, The Catholic University of Korea, Seoul, Republic of Korea
| | - Ju-Hyun Lee
- Department of Internal Medicine, Seoul National University Bundang Hospital, Seongnam, Republic of Korea
| | - Jee Hyun Kim
- Department of Internal Medicine, Seoul National University Bundang Hospital, Seongnam, Republic of Korea.
| |
Collapse
|
47
|
von Jeinsen B, Sopova K, Palapies L, Leistner DM, Fichtlscherer S, Seeger FH, Honold J, Dimmeler S, Aßmus B, Zeiher AM, Keller T. Bone marrow and plasma FGF-23 in heart failure patients: novel insights into the heart-bone axis. ESC Heart Fail 2019; 6:536-544. [PMID: 30912310 PMCID: PMC6487718 DOI: 10.1002/ehf2.12416] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2018] [Accepted: 01/17/2019] [Indexed: 01/13/2023] Open
Abstract
Aims Fibroblast growth factor 23 (FGF‐23) is known to be elevated in patients with congestive heart failure (CHF). As FGF‐23 is expressed in the bone but can also be expressed in the myocardium, the origin of serum FGF‐23 in CHF remains unclear. It is also unclear if FGF‐23 expressed in the bone is associated with outcome in CHF. The aim of the present study was to investigate FGF‐23 levels measured in bone marrow plasma (FGF‐23‐BM) and in peripheral blood (FGF‐23‐P) in CHF patients to gain further insights into the heart–bone axis of FGF‐23 expression. We also investigated possible associations between FGF‐23‐BM as well as FGF‐23‐P and outcome in CHF patients. Methods and results We determined FGF‐23‐P and FGF‐23‐BM levels in 203 CHF patients (85% male, mean age 61.3 years) with a left ventricular ejection fraction (LVEF) ≤45% and compared them with those of 48 healthy controls (48% male, mean age 39.2 years). We investigated the association between FGF‐23‐BM and FGF‐23‐P with all‐cause mortality in CHF patients, 32 events, median follow‐up 1673 days, interquartile range [923, 1828]. FGF‐23‐P (median 60.3 vs. 22.0 RU/mL, P < 0.001) and FGF‐23‐BM (median 130.7 vs. 93.1 RU/mL, P < 0.001) levels were higher in CHF patients compared with healthy controls. FGF‐23‐BM levels were significantly higher than FGF‐23‐P levels in both CHF patients and in healthy controls (P < 0.001). FGF‐23‐P and FGF‐23‐BM correlated significantly with LVEF (r = −0.37 and r = −0.33, respectively), N terminal pro brain natriuretic peptide levels (r = 0.57 and r = 0.6, respectively), New York Heart Association status (r = 0.28 and r = 0.25, respectively), and estimated glomerular filtration rate (r = −0.43 and r = −0.41, respectively) (P for all <0.001) and were independently associated with all‐cause mortality in CHF patients after adjustment for LVEF, estimated glomerular filtration rate, New York Heart Association status, and N terminal pro brain natriuretic peptide, hazard ratio 2.71 [confidence interval: 1.18–6.20], P = 0.018, and hazard ratio 2.80 [confidence interval: 1.19–6.57], P = 0.018, respectively. Conclusions In CHF patients, FGF‐23 is elevated in bone marrow plasma and is independently associated with heart failure severity and all‐cause mortality. The failing heart seems to interact via FGF‐23 within a heart–bone axis.
Collapse
Affiliation(s)
- Beatrice von Jeinsen
- Division of Cardiology, Department of Internal Medicine III, Goethe University Frankfurt, Frankfurt, Germany.,Department of Cardiology, Kerckhoff Heart and Thorax Center, Benekestr. 2-8, 61231, Bad Nauheim, Germany.,German Centre for Cardiovascular Research (DZHK), partner site RheinMain, Frankfurt, Germany
| | - Kateryna Sopova
- Division of Cardiology, Department of Internal Medicine III, Goethe University Frankfurt, Frankfurt, Germany.,German Centre for Cardiovascular Research (DZHK), partner site RheinMain, Frankfurt, Germany
| | - Lars Palapies
- Division of Cardiology, Department of Internal Medicine III, Goethe University Frankfurt, Frankfurt, Germany
| | - David M Leistner
- Division of Cardiology, Department of Internal Medicine III, Goethe University Frankfurt, Frankfurt, Germany.,Campus Benjamin Franklin, Department of Cardiology, Charité - Universitaetsmedizin Berlin, Berlin, Germany.,German Centre for Cardiovascular Research (DZHK), partner site Berlin, Berlin, Germany
| | - Stephan Fichtlscherer
- Division of Cardiology, Department of Internal Medicine III, Goethe University Frankfurt, Frankfurt, Germany.,German Centre for Cardiovascular Research (DZHK), partner site RheinMain, Frankfurt, Germany
| | - Florian H Seeger
- Division of Cardiology, Department of Internal Medicine III, Goethe University Frankfurt, Frankfurt, Germany.,German Centre for Cardiovascular Research (DZHK), partner site RheinMain, Frankfurt, Germany
| | - Jörg Honold
- Division of Cardiology, Department of Internal Medicine III, Goethe University Frankfurt, Frankfurt, Germany.,German Centre for Cardiovascular Research (DZHK), partner site RheinMain, Frankfurt, Germany
| | - Stefanie Dimmeler
- Institute for Cardiovascular Regeneration, Goethe University Frankfurt, Frankfurt, Germany.,German Centre for Cardiovascular Research (DZHK), partner site RheinMain, Frankfurt, Germany
| | - Birgit Aßmus
- Division of Cardiology, Department of Internal Medicine III, Goethe University Frankfurt, Frankfurt, Germany.,German Centre for Cardiovascular Research (DZHK), partner site RheinMain, Frankfurt, Germany
| | - Andreas M Zeiher
- Division of Cardiology, Department of Internal Medicine III, Goethe University Frankfurt, Frankfurt, Germany.,German Centre for Cardiovascular Research (DZHK), partner site RheinMain, Frankfurt, Germany
| | - Till Keller
- Division of Cardiology, Department of Internal Medicine III, Goethe University Frankfurt, Frankfurt, Germany.,Department of Cardiology, Kerckhoff Heart and Thorax Center, Benekestr. 2-8, 61231, Bad Nauheim, Germany.,German Centre for Cardiovascular Research (DZHK), partner site RheinMain, Frankfurt, Germany
| |
Collapse
|
48
|
Mesinovic J, Mousa A, Wilson K, Scragg R, Plebanski M, de Courten M, Scott D, Naderpoor N, de Courten B. Effect of 16-weeks vitamin D replacement on calcium-phosphate homeostasis in overweight and obese adults. J Steroid Biochem Mol Biol 2019; 186:169-175. [PMID: 30367939 DOI: 10.1016/j.jsbmb.2018.10.011] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/24/2018] [Revised: 10/03/2018] [Accepted: 10/23/2018] [Indexed: 12/21/2022]
Abstract
This randomised placebo-controlled trial aimed to determine the effect of 16-weeks cholecalciferol supplementation on calcium-phosphate homeostasis and bone mineral density (BMD) in overweight and obese adults. Fifty-four vitamin D-deficient (25OHD<50 nmol/L), overweight and obese adults (mean age 32 ± 8.5 years) were included in the trial. Participants were randomly assigned to receive either a bolus oral dose of 100,000 IU cholecalciferol followed by 4000 IU cholecalciferol/d or a matching placebo for 16 weeks. Before and after the intervention, serum calcium, phosphate, 25-hydroxyvitamin D [25(OH)D], intact parathyroid hormone (iPTH) and C-terminal plasma fibroblast growth factor-23 (cFGF-23) concentrations were measured. Whole-body BMD was assessed using dual-energy X-ray absorptiometry (DXA) and diet and sun exposure were assessed using self-administered questionnaires. There were no significant differences in baseline characteristics between the vitamin D and placebo group. After 16-weeks of vitamin D supplementation, mean changes in 25(OH)D concentration were higher in the vitamin D group (57 nmol/L 95% CI 49, 65) compared with placebo (2 nmol/L 95% CI -4, 8), P < 0.001. Additionally, iPTH concentrations declined in the vitamin D group (-1.19 pmol/L 95% CI -1.9, -0.47) compared with placebo (0.14 pmol/L 95% CI -0.49, 0.77), P = 0.006. There were no significant differences in calcium, phosphate, iPTH and cFGF-23 concentrations and whole-body BMD between vitamin D and placebo at follow-up. Inverse correlations were observed between mean change in serum iPTH and cFGF-23 in the vitamin D group only (r=-0.41, P = 0.029). In individuals with greater vitamin D deficiency at baseline (25(OH)D < 30 nmol/L), there was a significant increase in mean whole-body BMD (0.01 g/cm2, 95% CI 0.001, 0.025) however, the mean change in BMD was not different between vitamin D and placebo groups in this sub-group analysis. We conclude that cholecalciferol supplementation for 16 weeks increases serum 25(OH)D concentrations and reduces iPTH concentrations in overweight and obese, but otherwise healthy adults with vitamin D deficiency, and has no effect on calcium, phosphate and iFGF-23 concentrations and whole-body BMD.
Collapse
Affiliation(s)
- Jakub Mesinovic
- Department of Medicine, School of Clinical Sciences at Monash Health, Faculty of Medicine, Nursing and Health Sciences, Monash University, Clayton, Australia
| | - Aya Mousa
- Monash Centre for Health Research and Implementation, School of Public Health and Preventive Medicine, Monash University, Clayton, Australia
| | - Kirsty Wilson
- Department of Immunology and Pathology, Monash University, Australia
| | | | - Magdalena Plebanski
- Department of Immunology and Pathology, Monash University, Australia; School of Health and Biomedical Sciences, RMIT University, Australia
| | | | - David Scott
- Department of Medicine, School of Clinical Sciences at Monash Health, Faculty of Medicine, Nursing and Health Sciences, Monash University, Clayton, Australia
| | - Negar Naderpoor
- Monash Centre for Health Research and Implementation, School of Public Health and Preventive Medicine, Monash University, Clayton, Australia; Diabetes and Vascular Unit, Monash Health, Australia
| | - Barbora de Courten
- Monash Centre for Health Research and Implementation, School of Public Health and Preventive Medicine, Monash University, Clayton, Australia; Diabetes and Vascular Unit, Monash Health, Australia.
| |
Collapse
|
49
|
Affiliation(s)
- Clyde W Yancy
- Department of Medicine, Northwestern University Feinberg School of Medicine, Chicago, Illinois
| |
Collapse
|
50
|
Abstract
Chronic kidney disease mineral and bone disorder (MBD) encompasses changes in mineral ion and vitamin D metabolism that are widespread in the setting of chronic kidney disease and end-stage renal disease. MBD components associate with cardiovascular disease in many epidemiologic studies. Through impacts on hypertension, activation of the renin-angiotensin-aldosterone system, vascular calcification, endothelial function, and cardiac remodeling and conduction, MBD may be a direct and targetable cause of cardiovascular disease. However, assessment and treatment of MBD is rife with challenges owing to biological tensions between its many components, such as calcium and phosphorus with their regulatory hormones fibroblast growth factor 23 and parathyroid hormone; fibroblast growth factor 23 with its co-receptor klotho; and vitamin D with control of calcium and phosphorus. These complex interactions between MBD components hinder the simple translation to clinical trials, which ultimately are needed to prove the benefits of treating MBD. Deeper investigation using precision medicine tools and principles, including genomics and individualized risk assessment and therapy, may help move the field closer toward clinical applications. This review provides a high-level overview of conventional and precision epidemiology in MBD, potential mechanisms of cardiovascular disease pathogenesis, and guiding therapeutic principles for established and emerging treatments.
Collapse
Affiliation(s)
- Joseph Lunyera
- Department of Medicine, Duke University School of Medicine, Durham, NC
| | - Julia J Scialla
- Department of Medicine, Duke University School of Medicine, Durham, NC; Duke Clinical Research Institute, Duke University School of Medicine, Durham, NC; Department of Medicine, Durham Veterans Affairs Medical Center, Durham, NC.
| |
Collapse
|