1
|
Duque EJ, Ferreira GF, Oliveira IB, Dominguez W, Agena F, Jorgetti V, Lemos F, Wolf M, David-Neto E, Moysés RMA. Short and long-term effects of kidney donation on mineral and bone metabolism. BMC Nephrol 2024; 25:381. [PMID: 39462348 PMCID: PMC11514742 DOI: 10.1186/s12882-024-03827-0] [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: 08/05/2024] [Accepted: 10/17/2024] [Indexed: 10/29/2024] Open
Abstract
BACKGROUND Living kidney donors (LKD) experience an abrupt decline in glomerular filtration rate (GFR) resulting in abnormalities of mineral and bone metabolism (MBD), and this may have implications for skeletal health. We prospectively studied acute and long term MBD adaptation of LKD from two kidney transplant centers (São Paulo, Brazil and Miami, USA). METHODS Renal function and MBD parameters longitudinally after kidney donation (baseline - D0, day 1, 14, 180 and 360 post-operatively) were measured in 74 patients (40 y, 73% female, 54% Brazilian). A subset of 20 donors from Brazil were reassessed after 10 years of nephrectomy. RESULTS At baseline, Brazilian donors presented lower intact FGF23 (20.8 vs. 80.1 pg/mL, P < 0.01) and higher PTH (47.4 vs. 40.1, P = 0.04) than their US counterparts. GFR decreased to 63% of its baseline levels just after donation but improved 10% during the first year. PTH levels increased on D1, returning to baseline levels on D14, while FGF23 remained higher than baseline over the first year. LKD had a significant reduction of serum phosphate on D1, which returned to baseline levels on D180. A higher fractional excretion of phosphate (FEP) was noted since D14. After 10 years of donation, 20 LKD presented a sustained reduction in GFR (74.8 ± 14mL/min). There was a return to baseline in serum FGF23 [21.8 (18-30) pg/mL] and FEP, accompanied by an increase in serum calcium. PTH remained elevated (57.9 ± 18 pg/mL), whereas serum calcitriol and Klotho were lower than before the donation. CONCLUSIONS The abrupt decline in kidney mass is associated with an increase in PTH and FGF23 that is not explained by phosphate retention. In a long-term evaluation, LKD showed a sustained drop in GFR, with lower serum calcitriol and Klotho, and higher PTH. The effects of these changes should be investigated in further studies.
Collapse
Affiliation(s)
- Eduardo Jorge Duque
- Urology Division, Kidney Transplant Service - Hospital das Clínicas da Faculdade de Medicina da Universidade de São Paulo (HCFMUSP), São Paulo, Brazil.
- Nephrology Division, Laboratório de Fisiopatologia Renal (LIM16) - Hospital das Clínicas da Faculdade de Medicina da Universidade de São Paulo (HCFMUSP), São Paulo, Brazil.
| | - Gustavo Fernandes Ferreira
- Urology Division, Kidney Transplant Service - Hospital das Clínicas da Faculdade de Medicina da Universidade de São Paulo (HCFMUSP), São Paulo, Brazil
| | - Ivone Braga Oliveira
- Nephrology Division, Laboratório de Fisiopatologia Renal (LIM16) - Hospital das Clínicas da Faculdade de Medicina da Universidade de São Paulo (HCFMUSP), São Paulo, Brazil
| | - Wagner Dominguez
- Nephrology Division, Laboratório de Fisiopatologia Renal (LIM16) - Hospital das Clínicas da Faculdade de Medicina da Universidade de São Paulo (HCFMUSP), São Paulo, Brazil
| | - Fabiana Agena
- Urology Division, Kidney Transplant Service - Hospital das Clínicas da Faculdade de Medicina da Universidade de São Paulo (HCFMUSP), São Paulo, Brazil
| | - Vanda Jorgetti
- Nephrology Division, Laboratório de Fisiopatologia Renal (LIM16) - Hospital das Clínicas da Faculdade de Medicina da Universidade de São Paulo (HCFMUSP), São Paulo, Brazil
| | - Francine Lemos
- Urology Division, Kidney Transplant Service - Hospital das Clínicas da Faculdade de Medicina da Universidade de São Paulo (HCFMUSP), São Paulo, Brazil
| | - Myles Wolf
- Nephrology Division, Department of Medicine, Duke Clinical Research Institute, Duke University School of Medicine, Durham, NC, USA
| | - Elias David-Neto
- Urology Division, Kidney Transplant Service - Hospital das Clínicas da Faculdade de Medicina da Universidade de São Paulo (HCFMUSP), São Paulo, Brazil
| | - Rosa Maria A Moysés
- Nephrology Division, Laboratório de Fisiopatologia Renal (LIM16) - Hospital das Clínicas da Faculdade de Medicina da Universidade de São Paulo (HCFMUSP), São Paulo, Brazil
| |
Collapse
|
2
|
Wu W, Fu Y, Li H, Xiang Y, Zeng Y, Cai J, Dong Z. GALNT3 in Ischemia-Reperfusion Injury of the Kidney. J Am Soc Nephrol 2024:00001751-990000000-00463. [PMID: 39446490 DOI: 10.1681/asn.0000000530] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2024] [Accepted: 10/16/2024] [Indexed: 10/26/2024] Open
Abstract
Key Points
N-acetylgalactosaminyltransferase-3 (GALNT3) was downregulated in both ischemic AKI and cisplatin nephrotoxicity.GALNT3 played a protective role in renal tubular cells, and its downregulation contributed to AKI.Mechanistically, GALNT3 protected kidney tubular cells at least partially through O-glycosylation of EGF receptor.
Background
Damages to subcellular organelles, such as mitochondria and endoplasmic reticulum, are well recognized in tubular cell injury and death in AKI. However, the changes and involvement of Golgi apparatus are much less known. In this study, we report the regulation and role of N-acetylgalactosaminyltransferase-3 (GALNT3), a key enzyme for protein glycosylation in Golgi apparatus, in AKI.
Methods
AKI was induced in mice by renal ischemia–reperfusion injury or cisplatin. In vitro, rat kidney proximal tubular cells were subjected to hypoxia/reoxygenation (H/R) injury. To determine the role of GALNT3, its specific inhibitor T3inh-1 was tested in mice, and the effects of GALNT3 overexpression as well as knockdown were examined in the rat renal proximal tubular cells. EGF receptor (EGFR) activation was induced by recombinant EGF or by overexpressing EGFR.
Results
GALNT3 was significantly decreased in both in vivo and in vitro models of AKI induced by renal ischemia–reperfusion injury and cisplatin. T3Inh-1, a specific GALNT3 inhibitor, exacerbated ischemic AKI and suppressed tubular cell proliferation in mice. Moreover, knockdown of GALNT3 increased apoptosis during H/R treatment in rat renal proximal tubular cells, whereas overexpression of GALNT3 attenuated H/R-induced apoptosis, further supporting a protective role of GALNT3. Mechanistically, GALNT3 contributed to O-glycosylation of EGFR and associated EGFR signaling. Activation or overexpression of EGFR suppressed the proapoptotic effect of GALNT3 knockdown in H/R-treated rat renal proximal tubular cells.
Conclusions
GALNT3 protected kidney tubular cells in AKI at least partially through O-glycosylation of EGFR.
Collapse
Affiliation(s)
- Wenwen Wu
- Department of Nephrology, Hunan Key Laboratory of Kidney Disease and Blood Purification, Second Xiangya Hospital at Central South University, Changsha, China
| | - Ying Fu
- Department of Nephrology, Hunan Key Laboratory of Kidney Disease and Blood Purification, Second Xiangya Hospital at Central South University, Changsha, China
| | - Honglin Li
- Department of Nephrology, Hunan Key Laboratory of Kidney Disease and Blood Purification, Second Xiangya Hospital at Central South University, Changsha, China
| | - Yu Xiang
- Department of Nephrology, Hunan Key Laboratory of Kidney Disease and Blood Purification, Second Xiangya Hospital at Central South University, Changsha, China
| | - Yuqing Zeng
- Department of Nephrology, Hunan Key Laboratory of Kidney Disease and Blood Purification, Second Xiangya Hospital at Central South University, Changsha, China
| | - Juan Cai
- Department of Nephrology, Hunan Key Laboratory of Kidney Disease and Blood Purification, Second Xiangya Hospital at Central South University, Changsha, China
| | - Zheng Dong
- Department of Nephrology, Hunan Key Laboratory of Kidney Disease and Blood Purification, Second Xiangya Hospital at Central South University, Changsha, China
- Department of Cellular Biology and Anatomy, Medical College of Georgia at Augusta University, Augusta, Georgia
- Research Department, Charlie Norwood VA Medical Center, Augusta, Georgia
| |
Collapse
|
3
|
Valdes AM, Ikram A, Taylor LA, Zheng A, Kouraki A, Kelly A, Ashraf W, Vijay A, Miller S, Nightingale J, Selby NM, Ollivere BJ. Preoperative inflammatory biomarkers reveal renal involvement in postsurgical mortality in hip fracture patients: an exploratory study. Front Immunol 2024; 15:1372079. [PMID: 38919625 PMCID: PMC11197399 DOI: 10.3389/fimmu.2024.1372079] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2024] [Accepted: 05/20/2024] [Indexed: 06/27/2024] Open
Abstract
Background Hip fractures in frail patients result in excess mortality not accounted for by age or comorbidities. The mechanisms behind the high risk of mortality remain undetermined but are hypothesized to be related to the inflammatory status of frail patients. Methods In a prospective observational exploratory cohort study of hospitalized frail hip fracture patients, 92 inflammatory markers were tested in pre-operative serum samples and markers were tested against 6-month survival post-hip fracture surgery and incidence of acute kidney injury (AKI). After correcting for multiple testing, adjustments for comorbidities and demographics were performed on the statistically significant markers. Results Of the 92 markers tested, circulating levels of fibroblast growth factor 23 (FGF-23) and interleukin-15 receptor alpha (IL15RA), both involved in renal disease, were significantly correlated with 6-month mortality (27.5% overall) after correcting for multiple testing. The incidence of postoperative AKI (25.4%) was strongly associated with 6-month mortality, odds ratio = 10.57; 95% CI [2.76-40.51], and with both markers plus estimated glomerular filtration rate (eGFR)- cystatin C (CYSC) but not eGFR-CRE. The effect of these markers on mortality was significantly mediated by their effect on postoperative AKI. Conclusion High postoperative mortality in frail hip fracture patients is highly correlated with preoperative biomarkers of renal function in this pilot study. The effect of preoperative circulating levels of FGF-23, IL15RA, and eGFR-CYSC on 6-month mortality is in part mediated by their effect on postoperative AKI. Creatinine-derived preoperative renal function measures were very poorly correlated with postoperative outcomes in this group.
Collapse
Affiliation(s)
- Ana M. Valdes
- Injury, Recovery and Inflammation Sciences, School of Medicine, University of Nottingham, Nottingham, United Kingdom
- National Institute for Health and Care Research (NIHR) Nottingham Biomedical Research Centre, Nottingham, United Kingdom
| | - Adeel Ikram
- Injury, Recovery and Inflammation Sciences, School of Medicine, University of Nottingham, Nottingham, United Kingdom
- National Institute for Health and Care Research (NIHR) Nottingham Biomedical Research Centre, Nottingham, United Kingdom
| | - Lauren A. Taylor
- Injury, Recovery and Inflammation Sciences, School of Medicine, University of Nottingham, Nottingham, United Kingdom
- National Institute for Health and Care Research (NIHR) Nottingham Biomedical Research Centre, Nottingham, United Kingdom
| | - Amy Zheng
- Injury, Recovery and Inflammation Sciences, School of Medicine, University of Nottingham, Nottingham, United Kingdom
- National Institute for Health and Care Research (NIHR) Nottingham Biomedical Research Centre, Nottingham, United Kingdom
| | - Afroditi Kouraki
- Injury, Recovery and Inflammation Sciences, School of Medicine, University of Nottingham, Nottingham, United Kingdom
- National Institute for Health and Care Research (NIHR) Nottingham Biomedical Research Centre, Nottingham, United Kingdom
| | - Anthony Kelly
- Injury, Recovery and Inflammation Sciences, School of Medicine, University of Nottingham, Nottingham, United Kingdom
- National Institute for Health and Care Research (NIHR) Nottingham Biomedical Research Centre, Nottingham, United Kingdom
| | - Waheed Ashraf
- Injury, Recovery and Inflammation Sciences, School of Medicine, University of Nottingham, Nottingham, United Kingdom
- National Institute for Health and Care Research (NIHR) Nottingham Biomedical Research Centre, Nottingham, United Kingdom
| | - Amrita Vijay
- Injury, Recovery and Inflammation Sciences, School of Medicine, University of Nottingham, Nottingham, United Kingdom
- National Institute for Health and Care Research (NIHR) Nottingham Biomedical Research Centre, Nottingham, United Kingdom
| | - Suzanne Miller
- Injury, Recovery and Inflammation Sciences, School of Medicine, University of Nottingham, Nottingham, United Kingdom
- National Institute for Health and Care Research (NIHR) Nottingham Biomedical Research Centre, Nottingham, United Kingdom
| | - Jessica Nightingale
- Injury, Recovery and Inflammation Sciences, School of Medicine, University of Nottingham, Nottingham, United Kingdom
- National Institute for Health and Care Research (NIHR) Nottingham Biomedical Research Centre, Nottingham, United Kingdom
| | - Nicholas M. Selby
- Centre for Kidney Research and Innovation, Academic Unit for Translational Medical Sciences, School of Medicine, University of Nottingham, Nottingham, United Kingdom
| | - Benjamin J. Ollivere
- Injury, Recovery and Inflammation Sciences, School of Medicine, University of Nottingham, Nottingham, United Kingdom
- National Institute for Health and Care Research (NIHR) Nottingham Biomedical Research Centre, Nottingham, United Kingdom
| |
Collapse
|
4
|
Martínez-Heredia L, Canelo-Moreno JM, García-Fontana B, Muñoz-Torres M. Non-Classical Effects of FGF23: Molecular and Clinical Features. Int J Mol Sci 2024; 25:4875. [PMID: 38732094 PMCID: PMC11084844 DOI: 10.3390/ijms25094875] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2024] [Revised: 04/21/2024] [Accepted: 04/26/2024] [Indexed: 05/13/2024] Open
Abstract
This article reviews the role of fibroblast growth factor 23 (FGF23) protein in phosphate metabolism, highlighting its regulation of vitamin D, parathyroid hormone, and bone metabolism. Although it was traditionally thought that phosphate-calcium homeostasis was controlled exclusively by parathyroid hormone (PTH) and calcitriol, pathophysiological studies revealed the influence of FGF23. This protein, expressed mainly in bone, inhibits the renal reabsorption of phosphate and calcitriol formation, mediated by the α-klotho co-receptor. In addition to its role in phosphate metabolism, FGF23 exhibits pleiotropic effects in non-renal systems such as the cardiovascular, immune, and metabolic systems, including the regulation of gene expression and cardiac fibrosis. Although it has been proposed as a biomarker and therapeutic target, the inhibition of FGF23 poses challenges due to its potential side effects. However, the approval of drugs such as burosumab represents a milestone in the treatment of FGF23-related diseases.
Collapse
Affiliation(s)
- Luis Martínez-Heredia
- Instituto de Investigación Biosanitaria de Granada, 18014 Granada, Spain;
- Biomedical Research Network in Fragility and Healthy Aging (CIBERFES), Instituto de Salud Carlos III, 28029 Madrid, Spain
| | | | - Beatriz García-Fontana
- Instituto de Investigación Biosanitaria de Granada, 18014 Granada, Spain;
- Biomedical Research Network in Fragility and Healthy Aging (CIBERFES), Instituto de Salud Carlos III, 28029 Madrid, Spain
- Endocrinology and Nutrition Unit, University Hospital Clínico San Cecilio, 18016 Granada, Spain
- Department of Cell Biology, University of Granada, 18016 Granada, Spain
| | - Manuel Muñoz-Torres
- Instituto de Investigación Biosanitaria de Granada, 18014 Granada, Spain;
- Biomedical Research Network in Fragility and Healthy Aging (CIBERFES), Instituto de Salud Carlos III, 28029 Madrid, Spain
- Endocrinology and Nutrition Unit, University Hospital Clínico San Cecilio, 18016 Granada, Spain
- Department of Medicine, University of Granada, 18016 Granada, Spain
| |
Collapse
|
5
|
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: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2023] [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
|
6
|
Hamid AK, Pastor Arroyo EM, Lee SS, Wagner CA, Egli-Spichtig D. A novel method for automated crystal visualization and quantification in murine folic acid-induced acute kidney injury. Am J Physiol Renal Physiol 2024; 326:F105-F117. [PMID: 37881875 PMCID: PMC11194050 DOI: 10.1152/ajprenal.00140.2023] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2023] [Revised: 09/14/2023] [Accepted: 10/06/2023] [Indexed: 10/27/2023] Open
Abstract
Folic acid (FA)-induced acute kidney injury (FA-AKI) is an increasingly prevalent rodent disease model involving the injection of a high dose of FA that culminates in renal FA crystal deposition and injury. However, the literature characterizing the FA-AKI model is sparse and dated in part due to the absence of a well-described methodology for the visualization and quantification of renal FA crystals. Using widely available materials and tools, we developed a straightforward and crystal-preserving histological protocol that can be coupled with automated imaging for renal FA crystal visualization and generated an automated macro for downstream crystal content quantification. The applicability of the method was demonstrated by characterizing the model in male and female C57BL6/JRj mice after 3 and 30 h of FA treatment. Kidneys from both sexes and timepoints showed a bimodal distribution of FA crystal deposition in the cortical and medullary regions while, compared with males, females exhibited higher renal FA crystal content at the 30-h timepoint accompanied by greater kidney weight and higher plasma urea. Despite comparable plasma phosphate concentrations, FA-AKI resulted in a substantially more elevated plasma intact fibroblast growth factor 23 (FGF23) in females, reflected by a similar pattern in osseous Fgf23 mRNA expression. Therefore, the presented method constitutes a valuable tool for the quantification of renal FA crystals, which can aid the mechanistic characterization of the FA-AKI model and serves as a means to control for confounding changes in FA crystallization when using the model for investigating early and prophylactic AKI therapeutic interventions.NEW & NOTEWORTHY Here, we describe a novel method for the visualization and quantification of renal folic acid (FA) crystals in the rodent FA-induced acute kidney injury (FA-AKI) model. The protocol involves a straightforward histological approach followed by fully automated imaging and quantification steps. Applicability was confirmed by showing that the FA-AKI model is sex-dependent. The method can serve as a tool to aid in characterizing FA-AKI and to control for studies investigating prophylactic therapeutic avenues using FA-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
| | - Sung Sik Lee
- Scientific Center for Optical and Electron Microscopy, ETH Zurich, Zurich, Switzerland
- Institute of Biochemistry, Department of Biology, ETH Zurich, Zurich, Switzerland
| | - 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
|
7
|
Borucki JP, Woods R, Fielding A, Webb LA, Hernon JM, Lines SW, Stearns AT. Postoperative decline in renal function after rectal resection and all-cause mortality: a retrospective cohort study. Colorectal Dis 2023; 25:2225-2232. [PMID: 37803491 DOI: 10.1111/codi.16768] [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: 05/10/2023] [Revised: 07/20/2023] [Accepted: 08/14/2023] [Indexed: 10/08/2023]
Abstract
AIM Fluid loss, dehydration and resultant kidney injury are common when a diverting ileostomy is formed during rectal cancer surgery, the consequences of which are unknown. The aim of this retrospective single-site cohort study is to evaluate the impact of sustained postoperative renal dysfunction after rectal resection on long-term renal impairment and survival. METHOD All patients with rectal adenocarcinoma undergoing resection between January 2003 and March 2017 were included, with follow-up to June 2020. The primary outcome was impact on long-term mortality attributed to a 25% or greater drop in estimated glomerular filtration rate (eGFR) following rectal resection. Secondary outcomes were the long-term effect on renal function resulting from the same drop in eGFR and the effect on long-term mortality and renal function of a 50% drop in eGFR. We also calculated the effect on mortality of a 1% drop in eGFR. RESULTS A total of 1159 patients were identified. Postoperative reductions in eGFR of 25% and 50% were associated with long-term overall mortality with adjusted hazard ratios of 1.84 (1.22-2.77) (p = 0.004) and 2.88 (1.45-5.71) (p = 0.002). The median survival of these groups was 86.0 (64.0-108.0) months and 53.3 (7.8-98.8) months compared with 144.5 (128.1-160.9) months for controls. Long-term effects on renal function were demonstrated, with those who sustained a >25% drop in renal function having a 38.8% mean decline in eGFR at 10 years compared with 10.2% in controls. CONCLUSION Persistent postoperative declines in renal function may be linked to long-term mortality. Further research is needed to assess causal relationships and prevention.
Collapse
Affiliation(s)
- Joseph P Borucki
- Department of General Surgery, James Paget University Hospitals NHS Foundation Trust, Great Yarmouth, UK
- Sir Thomas Browne Academic Colorectal Unit, Norfolk and Norwich University Hospitals NHS Foundation Trust, Norwich, UK
- Norwich Medical School, University of East Anglia, Norwich, UK
- Norwich Surgical Training and Research Academy, Level 3 Centre, Norfolk and Norwich University Hospital, Norwich, UK
| | - Rebecca Woods
- Sir Thomas Browne Academic Colorectal Unit, Norfolk and Norwich University Hospitals NHS Foundation Trust, Norwich, UK
- Norwich Surgical Training and Research Academy, Level 3 Centre, Norfolk and Norwich University Hospital, Norwich, UK
| | - Alexandra Fielding
- Sir Thomas Browne Academic Colorectal Unit, Norfolk and Norwich University Hospitals NHS Foundation Trust, Norwich, UK
- Norwich Surgical Training and Research Academy, Level 3 Centre, Norfolk and Norwich University Hospital, Norwich, UK
| | - Lucy-Ann Webb
- Sir Thomas Browne Academic Colorectal Unit, Norfolk and Norwich University Hospitals NHS Foundation Trust, Norwich, UK
- Norwich Surgical Training and Research Academy, Level 3 Centre, Norfolk and Norwich University Hospital, Norwich, UK
| | - James M Hernon
- Sir Thomas Browne Academic Colorectal Unit, Norfolk and Norwich University Hospitals NHS Foundation Trust, Norwich, UK
- Norwich Medical School, University of East Anglia, Norwich, UK
- Norwich Surgical Training and Research Academy, Level 3 Centre, Norfolk and Norwich University Hospital, Norwich, UK
| | - Simon W Lines
- Department of Nephrology, St Bernard's Hospital, Gibraltar, Gibraltar
| | - Adam T Stearns
- Sir Thomas Browne Academic Colorectal Unit, Norfolk and Norwich University Hospitals NHS Foundation Trust, Norwich, UK
- Norwich Medical School, University of East Anglia, Norwich, UK
- Norwich Surgical Training and Research Academy, Level 3 Centre, Norfolk and Norwich University Hospital, Norwich, UK
| |
Collapse
|
8
|
Toro L, Rojas V, Conejeros C, Ayala P, Parra-Lucares A, Ahumada F, Almeida P, Silva MF, Bravo K, Pumarino C, Tong AM, Pinto ME, Romero C, Michea L. A Combined Biomarker That Includes Plasma Fibroblast Growth Factor 23, Erythropoietin, and Klotho Predicts Short- and Long-Term Morbimortality and Development of Chronic Kidney Disease in Critical Care Patients with Sepsis: A Prospective Cohort. Biomolecules 2023; 13:1481. [PMID: 37892163 PMCID: PMC10604443 DOI: 10.3390/biom13101481] [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: 06/01/2023] [Revised: 08/23/2023] [Accepted: 08/28/2023] [Indexed: 10/29/2023] Open
Abstract
Acute Kidney Injury (AKI) is a frequent complication in intensive care unit (ICU) patients that increases mortality and chronic kidney disease (CKD) development. AKI is associated with elevated plasma fibroblast growth factor 23 (FGF23), which can be modulated by erythropoietin (EPO) and Klotho. We aimed to evaluate whether a combined biomarker that includes these molecules predicted short-/long-term outcomes. We performed a prospective cohort of ICU patients with sepsis and previously normal renal function. They were followed during their inpatient stay and for one year after admission. We measured plasma FGF23, EPO, and Klotho levels at admission and calculated a combined biomarker (FEK). A total of 164 patients were recruited. Of these, 50 (30.5%) had AKI at admission, and 55 (33.5%) developed AKI within 48 h. Patients with AKI at admission and those who developed AKI within 48 h had 12- and 5-fold higher FEK values than non-AKI patients, respectively. Additionally, patients with higher FEK values had increased 1-year mortality (41.9% vs. 18.6%, p = 0.003) and CKD progression (26.2% vs. 8.3%, p = 0.023). Our data suggest that the FEK indicator predicts the risk of AKI, short-/long-term mortality, and CKD progression in ICU patients with sepsis. This new indicator can improve clinical outcome prediction and guide early therapeutic strategies.
Collapse
Affiliation(s)
- Luis Toro
- Division of Nephrology, Department of Medicine, Hospital Clínico Universidad de Chile, Santiago 8380456, Chile; (L.T.)
- Centro de Investigación Clínica Avanzada, Hospital Clínico Universidad de Chile, Santiago 8380456, Chile
- Centro de Pacientes Críticos, Clinica Las Condes, Santiago 7591047, Chile
| | - Verónica Rojas
- Centro de Investigación Clínica Avanzada, Hospital Clínico Universidad de Chile, Santiago 8380456, Chile
- Unidad de Pacientes Críticos, Departamento de Medicina, Hospital Clínico Universidad de Chile, Santiago 8380456, Chile
| | - Carolina Conejeros
- Division of Nephrology, Department of Medicine, Hospital Clínico Universidad de Chile, Santiago 8380456, Chile; (L.T.)
| | - Patricia Ayala
- Centro de Investigación Clínica Avanzada, Hospital Clínico Universidad de Chile, Santiago 8380456, Chile
| | - Alfredo Parra-Lucares
- Instituto de Ciencias Biomédicas, Facultad de Medicina, Universidad de Chile, Santiago 8380456, Chile
| | - Francisca Ahumada
- Instituto de Ciencias Biomédicas, Facultad de Medicina, Universidad de Chile, Santiago 8380456, Chile
| | - Paula Almeida
- Instituto de Ciencias Biomédicas, Facultad de Medicina, Universidad de Chile, Santiago 8380456, Chile
| | - María Fernanda Silva
- Instituto de Ciencias Biomédicas, Facultad de Medicina, Universidad de Chile, Santiago 8380456, Chile
| | - Karin Bravo
- Instituto de Ciencias Biomédicas, Facultad de Medicina, Universidad de Chile, Santiago 8380456, Chile
| | - Catalina Pumarino
- Instituto de Ciencias Biomédicas, Facultad de Medicina, Universidad de Chile, Santiago 8380456, Chile
| | - Ana María Tong
- Clinical Laboratory, Hospital Clínico Universidad de Chile, Santiago 8380456, Chile
| | - María Eugenia Pinto
- Clinical Laboratory, Hospital Clínico Universidad de Chile, Santiago 8380456, Chile
| | - Carlos Romero
- Unidad de Pacientes Críticos, Departamento de Medicina, Hospital Clínico Universidad de Chile, Santiago 8380456, Chile
| | - Luis Michea
- Division of Nephrology, Department of Medicine, Hospital Clínico Universidad de Chile, Santiago 8380456, Chile; (L.T.)
- Instituto de Ciencias Biomédicas, Facultad de Medicina, Universidad de Chile, Santiago 8380456, Chile
- Laboratorio de Fisiologia Integrativa, Facultad de Medicina Universidad de Chile, Santiago 8380456, Chile
| |
Collapse
|
9
|
Mattinzoli D, Molinari P, Romero-González G, Bover J, Cicero E, Pesce F, Abinti M, Conti C, Castellano G, Alfieri C. Is there a role in acute kidney injury for FGF23 and Klotho? Clin Kidney J 2023; 16:1555-1562. [PMID: 37779849 PMCID: PMC10539225 DOI: 10.1093/ckj/sfad093] [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] [Received: 01/02/2023] [Indexed: 10/03/2023] Open
Abstract
Cardio-renal syndrome is a clinical condition that has recently been well defined. In acute kidney disease, this interaction might trigger chronic processes determining the onset of cardiovascular events and the progression of chronic kidney disease. Moreover, the high mortality rate of acute kidney injury (AKI) is also linked to the fact that this condition is often complicated by dysfunctions of other organs such as lungs or heart, or is associated with septic episodes. In this context the role and the potential link between bone, heart and kidney is becoming an important topic of research. The aim of this review is to describe the cardiac alterations in the presence of AKI (cardiorenal syndrome type 3) and explore how bone can interact with heart and kidney in determining and influencing the trend of AKI in the short and long term. The main anomalies of mineral metabolism in patients with AKI will be reported, with specific reference to the alterations of fibroblast growth factor 23 and Klotho as a link between the bone-kidney-heart axis.
Collapse
Affiliation(s)
- Deborah Mattinzoli
- Department of Nephrology, Dialysis and Renal Transplantation, Fondazione IRCCS Ca’ Granda Ospedale Policlinico, Milan, Italy
| | - Paolo Molinari
- Department of Nephrology, Dialysis and Renal Transplantation, Fondazione IRCCS Ca’ Granda Ospedale Policlinico, Milan, Italy
- Post-Graduate School of Specialization in Nephrology, University of Milan, Milan, Italy
| | - Gregorio Romero-González
- Department of Nephrology, Germans Trias i Pujol University Hospital, Research Group on Renal Diseases (REMAR), Germans Trias i Pujol Research Institute, Badalona, Spain
| | - Jordi Bover
- Department of Nephrology, Germans Trias i Pujol University Hospital, Research Group on Renal Diseases (REMAR), Germans Trias i Pujol Research Institute, Badalona, Spain
| | - Elisa Cicero
- Department of Nephrology, Dialysis and Renal Transplantation, Fondazione IRCCS Ca’ Granda Ospedale Policlinico, Milan, Italy
- Post-Graduate School of Specialization in Nephrology, University of Milan, Milan, Italy
| | - Francesco Pesce
- Nephrology, Dialysis and Transplantation Unit Department of Precision and Regenerative Medicine and Ionian Area (DiMePre-J) University of Bari “Aldo Moro”
| | - Matteo Abinti
- Department of Nephrology, Dialysis and Renal Transplantation, Fondazione IRCCS Ca’ Granda Ospedale Policlinico, Milan, Italy
- Post-Graduate School of Specialization in Nephrology, University of Milan, Milan, Italy
| | - Costanza Conti
- Department of Nephrology, Dialysis and Renal Transplantation, Fondazione IRCCS Ca’ Granda Ospedale Policlinico, Milan, Italy
- Post-Graduate School of Specialization in Nephrology, University of Milan, Milan, Italy
| | - Giuseppe Castellano
- Department of Nephrology, Dialysis and Renal Transplantation, Fondazione IRCCS Ca’ Granda Ospedale Policlinico, Milan, Italy
- Department of Clinical Sciences and Community Health, University of Milan, Milan, Italy
| | - Carlo Alfieri
- Department of Nephrology, Dialysis and Renal Transplantation, Fondazione IRCCS Ca’ Granda Ospedale Policlinico, Milan, Italy
- Department of Clinical Sciences and Community Health, University of Milan, Milan, Italy
| |
Collapse
|
10
|
Neyra JA, Moe OW. Bone Dysregulation in Acute Kidney Injury. Nephron Clin Pract 2023; 147:747-753. [PMID: 37757785 DOI: 10.1159/000534228] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2023] [Accepted: 09/05/2023] [Indexed: 09/29/2023] Open
Abstract
Acute kidney injury (AKI) is a highly prevalent condition with multiple acute and chronic consequences. Survivors of AKI are at risk of AKI-to-chronic kidney disease (CKD) transition, which carries significant morbidity and mortality. One retrospective analysis showed increased risk of bone fracture post-AKI in humans, which was independent of CKD development. While there are several theoretical reasons for late disturbances of bone health post-AKI, no definitive data are available to date. An important question is whether there are bone sequelae from AKI that are independent of CKD, meaning bone disease prior to the onset, or in the absence of CKD - a form of "post-AKI osteopathy." While preclinical studies examining bone health after acute stressors have focused mostly on sepsis models, multiple experimental AKI models are readily available for longitudinal bone health interrogation. Future research should be tailored to define whether AKI is a risk factor, independent of CKD, for bone disease and if present, the time course and type of bone disease. This review summarizes a fraction of the existing data to provide some guidance in future research efforts.
Collapse
Affiliation(s)
- Javier A Neyra
- Department of Medicine, University of Alabama, Birmingham, Alabama, USA
- Charles and Jane Pak Center for Mineral Metabolism and Clinical Research, University of Texas Southwestern Medical Center, Dallas, Texas, USA
| | - Orson W Moe
- Charles and Jane Pak Center for Mineral Metabolism and Clinical Research, University of Texas Southwestern Medical Center, Dallas, Texas, USA
- Department of Internal Medicine, University of Texas Southwestern Medical Center, Dallas, Texas, USA
- Departments of Internal Medicine and Physiology, University of Texas Southwestern Medical Center, Dallas, Texas, USA
| |
Collapse
|
11
|
McCoy IE, Hsu JY, Zhang X, Diamantidis CJ, Taliercio J, Go AS, Liu KD, Drawz P, Srivastava A, Horwitz EJ, He J, Chen J, Lash JP, Weir MR, Hsu CY. Probing the Association between Acute Kidney Injury and Cardiovascular Outcomes. Clin J Am Soc Nephrol 2023; 18:850-857. [PMID: 37116457 PMCID: PMC10356151 DOI: 10.2215/cjn.0000000000000163] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2023] [Accepted: 04/17/2023] [Indexed: 04/30/2023]
Abstract
BACKGROUND Patients hospitalized with AKI have higher subsequent risks of heart failure, atherosclerotic cardiovascular events, and mortality than their counterparts without AKI, but these higher risks may be due to differences in prehospitalization patient characteristics, including the baseline level of estimated glomerular filtration rate (eGFR), the rate of prior eGFR decline, and the proteinuria level, rather than AKI itself. METHODS Among 2177 adult participants in the Chronic Renal Insufficiency Cohort study who were hospitalized in 2013-2019, we compared subsequent risks of heart failure, atherosclerotic cardiovascular events, and mortality between those with serum creatinine-based AKI (495 patients) and those without AKI (1682 patients). We report both crude associations and associations sequentially adjusted for prehospitalization characteristics including eGFR, eGFR slope, and urine protein-creatinine ratio (UPCR). RESULTS Compared with patients hospitalized without AKI, those with hospitalized AKI had lower eGFR prehospitalization (42 versus 49 ml/min per 1.73 m 2 ), faster chronic loss of eGFR prehospitalization (-0.84 versus -0.51 ml/min per 1.73 m 2 per year), and more proteinuria prehospitalization (UPCR 0.28 versus 0.16 g/g); they also had higher prehospitalization systolic BP (130 versus 127 mm Hg; P < 0.01 for all comparisons). Adjustment for prehospitalization patient characteristics attenuated associations between AKI and all three outcomes, but AKI remained an independent risk factor. Attenuation of risk was similar after adjustment for absolute eGFR, eGFR slope, or proteinuria, individually or in combination. CONCLUSIONS Prehospitalization variables including eGFR, eGFR slope, and proteinuria confounded associations between AKI and adverse cardiovascular outcomes, but these associations remained significant after adjusting for prehospitalization variables.
Collapse
Affiliation(s)
- Ian E. McCoy
- Division of Nephrology, University of California San Francisco, San Francisco, California
| | - Jesse Y. Hsu
- Department of Biostatistics, Epidemiology, and Informatics, University of Pennsylvania, Philadelphia, Pennsylvania
| | - Xiaoming Zhang
- Department of Biostatistics, Epidemiology, and Informatics, University of Pennsylvania, Philadelphia, Pennsylvania
| | | | - Jonathan Taliercio
- Department of Kidney Medicine, Cleveland Clinic Foundation, Cleveland, Ohio
| | - Alan S. Go
- Division of Research, Kaiser Permanente Northern California, Oakland, California
| | - Kathleen D. Liu
- Division of Nephrology, University of California San Francisco, San Francisco, California
| | - Paul Drawz
- Division of Nephrology and Hypertension, University of Minnesota, Minneapolis, Minnesota
| | - Anand Srivastava
- Division of Nephrology, Department of Medicine, University of Illinois Chicago, Chicago, Illinois
| | - Edward J. Horwitz
- Division of Nephrology, MetroHealth Medical Center, Case Western Reserve University, Cleveland, Ohio
| | - Jiang He
- Department of Epidemiology, Tulane University, New Orleans, Louisiana
| | - Jing Chen
- Department of Epidemiology, Tulane University, New Orleans, Louisiana
- Division of Nephrology, Tulane University, New Orleans, Louisiana
| | - James P. Lash
- Department of Medicine, University of Illinois College of Medicine at Chicago, Chicago, Illinois
| | - Matthew R. Weir
- Division of Nephrology, University of Maryland School of Medicine, Baltimore, Maryland
| | - Chi-yuan Hsu
- Division of Nephrology, University of California San Francisco, San Francisco, California
- Division of Research, Kaiser Permanente Northern California, Oakland, California
| |
Collapse
|
12
|
Matsuura R, Doi K, Rabb H. Acute kidney injury and distant organ dysfunction-network system analysis. Kidney Int 2023; 103:1041-1055. [PMID: 37030663 DOI: 10.1016/j.kint.2023.03.025] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2022] [Revised: 03/24/2023] [Accepted: 03/24/2023] [Indexed: 04/10/2023]
Abstract
Acute kidney injury (AKI) occurs in about half of critically ill patients and associates with high in-hospital mortality, increased long-term mortality post-discharge and subsequent progression to chronic kidney disease. Numerous clinical studies have shown that AKI is often complicated by dysfunction of distant organs, which is a cause of the high mortality associated with AKI. Experimental studies have elucidated many mechanisms of AKI-induced distant organ injury, which include inflammatory cytokines, oxidative stress and immune responses. This review will provide an update on evidence of organ crosstalk and potential therapeutics for AKI-induced organ injuries, and present the new concept of a systemic organ network to balance homeostasis and inflammation that goes beyond kidney-crosstalk with a single distant organ.
Collapse
Affiliation(s)
- Ryo Matsuura
- Department of Nephrology and Endocrinology, the University of Tokyo Hospital
| | - Kent Doi
- Department of Emergency and Critical Care Medicine, the University of Tokyo Hospital.
| | - Hamid Rabb
- Division of Nephrology, Department of Medicine, Johns Hopkins University School of Medicine
| |
Collapse
|
13
|
Copur S, Demiray A, Basile C, Kanbay M. Endocrinological disorders in acute kidney injury: an often overlooked field of clinical research. J Nephrol 2023; 36:885-893. [PMID: 36652168 DOI: 10.1007/s40620-022-01554-z] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2022] [Accepted: 12/05/2022] [Indexed: 01/19/2023]
Abstract
Acute kidney injury (AKI) is a common comorbidity, affecting approximately one in five hospitalized adults. The kidney is the site for the production, metabolism or excretion of most hormones, including the production of erythropoietin (EPO), the active form of vitamin D, renin, thrombopoietin, and the excretion of insulin, catecholamines, gastrin and many other hormones. Therefore, it is reasonable to say that AKI can have a considerable impact on the endocrine system. Although the effects of AKI on various parameters, including cardiovascular parameters, serum electrolytes and acid-base disorders, neuro-humoral mechanisms and neurological outcomes have been extensively studied, the endocrinological consequences of AKI are understudied. Thyroid dysfunction, mainly euthyroid sick syndrome, hypo/hyperglycemia, bone mineral disorders, changes in EPO and atrial natriuretic peptide (ANP) levels are commonly found in AKI. EPO, thyroxine and ANP administration have been evaluated as potential tools to prevent or treat AKI with varying success, while the effects of AKI on some key hormones, including cortisol and insulin, have never been studied. Aim of this narrative review is to illustrate what is known and what is not known about the endocrinological outcomes of AKI. Few clinical trials are ongoing: however, there is a clear need for large-scale randomized controlled trials investigating the endocrinological consequences of AKI.
Collapse
Affiliation(s)
- Sidar Copur
- Department of Medicine, Koc University School of Medicine, Istanbul, Turkey
| | - Atalay Demiray
- Department of Medicine, Koc University School of Medicine, Istanbul, Turkey
| | - Carlo Basile
- Associazione Nefrologica Gabriella Sebastio, Martina Franca, Italy.
| | - Mehmet Kanbay
- Department of Medicine, Division of Nephrology, Koc University School of Medicine, Istanbul, Turkey
| |
Collapse
|
14
|
Thein OS, Ali NA, Mahida RY, Dancer RCA, Ostermann M, Amrein K, Martucci G, Scott A, Thickett DR, Parekh D. Raised FGF23 Correlates to Increased Mortality in Critical Illness, Independent of Vitamin D. BIOLOGY 2023; 12:biology12020309. [PMID: 36829583 PMCID: PMC9953634 DOI: 10.3390/biology12020309] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 01/18/2023] [Revised: 02/09/2023] [Accepted: 02/09/2023] [Indexed: 02/17/2023]
Abstract
BACKGROUND Fibroblast Growth Factor (FGF23) is an endocrine hormone classically associated with the homeostasis of vitamin D, phosphate, and calcium. Elevated serum FGF23 is a known independent risk factor for mortality in chronic kidney disease (CKD) patients. We aimed to determine if there was a similar relationship between FGF23 levels and mortality in critically ill patients. METHODS Plasma FGF23 levels were measured by ELISA in two separate cohorts of patients receiving vitamin D supplementation: critical illness patients (VITdAL-ICU trial, n = 475) and elective oesophagectomy patients (VINDALOO trial, n = 76). Mortality data were recorded at 30 and 180 days or at two years, respectively. FGF23 levels in a healthy control cohort were also measured (n = 27). RESULTS Elevated FGF23 (quartile 4 vs. quartiles 1-3) was associated with increased short-term (30 and 180 day) mortality in critical illness patients (p < 0.001) and long-term (two-year) mortality in oesophagectomy patients (p = 0.0149). Patients who died had significantly higher FGF23 levels than those who survived: In the critical illness cohort, those who died had 1194.6 pg/mL (range 0-14,000), while those who survived had 120.4 pg/mL (range = 15-14,000) (p = 0.0462). In the oesophagectomy cohort, those who died had 1304 pg/mL (range = 154-77,800), while those who survived had 644 pg/mL (range = 179-54,894) (p < 0.001). This was found to be independent of vitamin D or CKD status (critical illness p = 0.3507; oesophagectomy p = 0.3800). FGF23 levels in healthy controls were similar to those seen in oesophagectomy patients (p = 0.4802). CONCLUSIONS Elevated baseline serum FGF23 is correlated with increased mortality in both the post-oesophagectomy cohort and the cohort of patients with critical illness requiring intensive care admission. This was independent of vitamin D status, supplementation, or CKD status, which suggests the presence of vitamin D-independent mechanisms of FGF23 action during the acute and convalescent stages of critical illness, warranting further investigation.
Collapse
Affiliation(s)
- Onn Shaun Thein
- Birmingham Acute Care Research Group, Institute of Inflammation and Ageing, University of Birmingham, Level 2 Queen Elizabeth Hospital Birmingham, Birmingham B15 2TH, UK
- Correspondence:
| | - Naeman Akbar Ali
- Birmingham Acute Care Research Group, Institute of Inflammation and Ageing, University of Birmingham, Level 2 Queen Elizabeth Hospital Birmingham, Birmingham B15 2TH, UK
| | - Rahul Y. Mahida
- Birmingham Acute Care Research Group, Institute of Inflammation and Ageing, University of Birmingham, Level 2 Queen Elizabeth Hospital Birmingham, Birmingham B15 2TH, UK
| | | | - Marlies Ostermann
- King’s College London, Guy’s & St Thomas’ Hospital, London SE1 7EH, UK
| | - Karin Amrein
- Division of Endocrinology and Diabetology, Medical University of Graz, 8036 Graz, Austria
| | - Gennaro Martucci
- Department of Anesthesia and Intensive Care, Istituto Mediterraneo per i Trapianti e Terapie ad Alta Specializzazione (IRCCS-ISMETT), 90133 Palermo, Italy
| | - Aaron Scott
- Birmingham Acute Care Research Group, Institute of Inflammation and Ageing, University of Birmingham, Level 2 Queen Elizabeth Hospital Birmingham, Birmingham B15 2TH, UK
| | - David R. Thickett
- Birmingham Acute Care Research Group, Institute of Inflammation and Ageing, University of Birmingham, Level 2 Queen Elizabeth Hospital Birmingham, Birmingham B15 2TH, UK
| | - Dhruv Parekh
- Birmingham Acute Care Research Group, Institute of Inflammation and Ageing, University of Birmingham, Level 2 Queen Elizabeth Hospital Birmingham, Birmingham B15 2TH, UK
| |
Collapse
|
15
|
Wolf M. FGF23 AND ALTERED MINERAL HOMEOSTASIS IN KIDNEY DISEASE AND FOLLOWING INTRAVENOUS IRON. TRANSACTIONS OF THE AMERICAN CLINICAL AND CLIMATOLOGICAL ASSOCIATION 2023; 133:262-273. [PMID: 37701608 PMCID: PMC10493719] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Subscribe] [Scholar Register] [Indexed: 09/14/2023]
Abstract
Fibroblast growth factor 23 (FGF23) is an endocrine hormone that stimulates renal phosphate excretion and suppresses circulating concentrations of 1,25-dihydroxyvitamin D (1,25D). These effects of FGF23 are most evident in rare diseases that are characterized by FGF23-mediated hypophosphatemic rickets-osteomalacia. More commonly, elevated FGF23 is a ubiquitous, early consequence of chronic kidney disease (CKD) in which it helps to maintain normal serum phosphate levels but causes secondary hyperparathyroidism by suppressing 1,25D, and directly promotes cardiovascular disease and death. Elevated FGF23 is also a common complication of intravenous administration of ferric carboxymaltose (FCM), which is widely used to treat iron deficiency anemia. Among patients with normal kidney function who receive FCM, the resulting increase in FGF23 and subsequent FGF23-mediated reduction of 1,25D and secondary hyperparathyroidism promote hypophosphatemia that can be symptomatic, severe, and associated with musculoskeletal complications. Ongoing research is needed to design novel therapeutic approaches to mitigate FGF23-related illnesses.
Collapse
|
16
|
Zhang L, Qin W. Research progress of fibroblast growth factor 23 in acute kidney injury. Pediatr Nephrol 2022:10.1007/s00467-022-05791-z. [PMID: 36416954 DOI: 10.1007/s00467-022-05791-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/11/2022] [Revised: 09/19/2022] [Accepted: 10/11/2022] [Indexed: 11/24/2022]
Abstract
Fibroblast growth factor 23 (FGF23) is primarily produced in bones and mainly regulates calcium and phosphorus metabolism. The level of circulating FGF23 increases rapidly in the early stage of acute kidney injury (AKI). Recent studies have shown that FGF23 may serve as a biomarker for the diagnosis and poor prognosis of AKI. The mechanism of increased FGF23 in AKI may include increased production of FGF23, decreased renal clearance of FGF23, and some new regulatory factors, such as inflammation and glycerol 3-phosphate. However, the biological effects of elevated FGF23 in AKI are still unclear. It is also not known whether reducing the level of circulating FGF23 could alleviate AKI or its poor prognosis. Here, we review the pathophysiological mechanism and possible regulation of FGF23 in AKI and discuss the possibility of using FGF23 as a therapeutic target.
Collapse
Affiliation(s)
- Lina Zhang
- Division of Nephrology, Department of Medicine, West China Hospital, Sichuan University, 37 Guoxue Lane, Chengdu, 610041, Sichuan, China.,Division of Nephrology, Henan Key Laboratory for Kidney Disease and Immunology, Henan Provincial People's Hospital, Zhengzhou, Henan, China
| | - Wei Qin
- Division of Nephrology, Department of Medicine, West China Hospital, Sichuan University, 37 Guoxue Lane, Chengdu, 610041, Sichuan, China.
| |
Collapse
|
17
|
Portales-Castillo I, Simic P. PTH, FGF-23, Klotho and Vitamin D as regulators of calcium and phosphorus: Genetics, epigenetics and beyond. Front Endocrinol (Lausanne) 2022; 13:992666. [PMID: 36246903 PMCID: PMC9558279 DOI: 10.3389/fendo.2022.992666] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/12/2022] [Accepted: 09/12/2022] [Indexed: 11/30/2022] Open
Abstract
The actions of several bone-mineral ion regulators, namely PTH, FGF23, Klotho and 1,25(OH)2 vitamin D (1,25(OH)2D), control calcium and phosphate metabolism, and each of these molecules has additional biological effects related to cell signaling, metabolism and ultimately survival. Therefore, these factors are tightly regulated at various levels - genetic, epigenetic, protein secretion and cleavage. We review the main determinants of mineral homeostasis including well-established genetic and post-translational regulators and bring attention to the epigenetic mechanisms that affect the function of PTH, FGF23/Klotho and 1,25(OH)2D. Clinically relevant epigenetic mechanisms include methylation of cytosine at CpG-rich islands, histone deacetylation and micro-RNA interference. For example, sporadic pseudohypoparathyroidism type 1B (PHP1B), a disease characterized by resistance to PTH actions due to blunted intracellular cAMP signaling at the PTH/PTHrP receptor, is associated with abnormal methylation at the GNAS locus, thereby leading to reduced expression of the stimulatory G protein α-subunit (Gsα). Post-translational regulation is critical for the function of FGF-23 and such modifications include glycosylation and phosphorylation, which regulate the cleavage of FGF-23 and hence the proportion of available FGF-23 that is biologically active. While there is extensive data on how 1,25(OH)2D and the vitamin D receptor (VDR) regulate other genes, much more needs to be learned about their regulation. Reduced VDR expression or VDR mutations are the cause of rickets and are thought to contribute to different disorders. Epigenetic changes, such as increased methylation of the VDR resulting in decreased expression are associated with several cancers and infections. Genetic and epigenetic determinants play crucial roles in the function of mineral factors and their disorders lead to different diseases related to bone and beyond.
Collapse
Affiliation(s)
- Ignacio Portales-Castillo
- Department of Medicine, Division of Nephrology, Massachusetts General Hospital and Harvard Medical School, Boston, MA, United States
- Endocrine Unit, Massachusetts General Hospital and Harvard Medical School, Boston, MA, United States
| | - Petra Simic
- Department of Medicine, Division of Nephrology, Massachusetts General Hospital and Harvard Medical School, Boston, MA, United States
- Endocrine Unit, Massachusetts General Hospital and Harvard Medical School, Boston, MA, United States
| |
Collapse
|
18
|
Wang W, Zhang L, Yang T, Ma S, Zhang Q, Shi P, Ding F. Combined serum free light chain predicts prognosis in acute kidney injury following cardiovascular surgery. Ren Fail 2022; 44:1-10. [PMID: 35086423 PMCID: PMC8797736 DOI: 10.1080/0886022x.2021.2013886] [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] [Indexed: 11/16/2022] Open
Abstract
Objectives Increased polyclonal free light chains (FLCs) are found in inflammatory conditions. Inflammation is recognized in the progression of acute kidney injury (AKI). This study was aimed to determine whether polyclonal combined FLC (cFLC) was associated with prognosis of AKI patients. Methods This prospective cohort included 145 adults with hospital-acquired AKI following cardiovascular surgery between 2014 and 2016, according to the KDIGO creatinine criteria. The primary end point of the study was all-cause death during follow-up. Results The median of serum cFLC concentration in the cohort was 42.0 (31.9–60.3 mg/L) and levels of cFLC in patients with AKI stage 3 were higher than those in AKI stage 1 and stage 2. cFLC levels correlated significantly with renal function biomarkers, high sensitivity C-reactive protein (hsCRP), and sequential organ failure assessment (SOFA) score. Patients were organized into the following two groups: the low-cFLC group (cFLC <43.3 mg/L) and the high-cFLC group (cFLC ≥ 43.3 mg/L). A total of 17 (11.0%) patient deaths occurred within 90 d, 13 (18.8%) in the high-cFLC group. Kaplan–Meier analysis revealed that the two groups differed significantly with respect to 90-d survival (log-rank p = .012), and Cox regression analysis showed that an cFLC level ≥43.3 mg/L was significantly associated with a 5.0-fold increased risk of death (adjusted hazard ratio [HR], 5.95; 95% confidence interval [CI], 1.04– 33.91; p = .045) compared with an cFLC level <43.3 mg/L. Conclusions Serum cFLC levels were significantly elevated and might be an independent predictor of mortality in patients with AKI following cardiovascular surgery.
Collapse
Affiliation(s)
- Wenji Wang
- School of Medicine, Division of Nephrology, Shanghai Ninth People's Hospital, Shanghai Jiaotong University, Shanghai, PR China
| | - Lulu Zhang
- School of Medicine, Division of Nephrology, Shanghai Ninth People's Hospital, Shanghai Jiaotong University, Shanghai, PR China
| | - Tianye Yang
- School of Medicine, Division of Nephrology, Shanghai Ninth People's Hospital, Shanghai Jiaotong University, Shanghai, PR China
| | - Shaojun Ma
- School of Medicine, Division of Nephrology, Shanghai Ninth People's Hospital, Shanghai Jiaotong University, Shanghai, PR China
| | - Qi Zhang
- School of Medicine, Division of Nephrology, Shanghai Ninth People's Hospital, Shanghai Jiaotong University, Shanghai, PR China
| | - Peng Shi
- Department of Medical Statistics, Children's Hospital; Center for Evidence-based Medicine, Fudan University, Shanghai, PR China
| | - Feng Ding
- School of Medicine, Division of Nephrology, Shanghai Ninth People's Hospital, Shanghai Jiaotong University, Shanghai, PR China
| |
Collapse
|
19
|
Batte A, Kasirye P, Baluku R, Kiguli S, Kalyesubula R, John CC, Schwaderer AL, Imel EA, Conroy AL. Mineral bone disorders and kidney disease in hospitalized children with sickle cell anemia. Front Pediatr 2022; 10:1078853. [PMID: 36819194 PMCID: PMC9932899 DOI: 10.3389/fped.2022.1078853] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/24/2022] [Accepted: 12/19/2022] [Indexed: 02/05/2023] Open
Abstract
BACKGROUND Mineral bone disorders (MBD) are common in sickle cell anemia (SCA). Frequent vaso-occlusive crises (VOC) further impact MBD in children with SCA. We evaluated the prevalence of markers of SCA-related MBD (sMBD) in hospitalized children and assessed the relationship between sMBD and individual mineral abnormalities with kidney disease. METHODS We prospectively recruited 185 children with SCA hospitalized with a VOC. Serum measures of mineral bone metabolism (calcium, phosphate, parathyroid hormone, 25-hydroxy vitamin D, FGF23, osteopontin) were measured at enrollment. The primary outcome was markers of sMBD defined as a composite of hypocalcemia, hyperphosphatemia, hyperparathyroidism, or deficiency in 25-OH vitamin D. Secondary outcomes included individual abnormalities in mineral metabolism. The Kidney Disease: Improving Global Outcomes (KDIGO) guidelines were used to define SCA-associated acute kidney injury (AKI). AKI was further assessed using urine NGAL as a marker of tubular injury. Acute kidney disease (AKD) was defined as a composite of AKI, an eGFR < 90 ml/min per 1.73 m2 using the Cystatin C GFR equation, or evidence of structural injury (positive biomarker test or albuminuria). RESULTS The mean age of children was 8.9 years and 41.6% were female. The prevalence of sMBD was 47.6%, with hypocalcemia the most frequent abnormality (29.9%, 55/184) followed by hyperphosphatemia (20.7%, 38/184), hyperparathyroidism (8.7%, 16/185), and vitamin D deficiency (5.4%, 10/185). There was no association between sMBD and sKDIGO-defined AKI using serial changes in creatinine or when incorporating biomarkers to define AKI. However, the presence of AKD was associated with a 2.01-fold increased odds of sMBD (95% CI 1.05 to 3.83) and was driven by a decrease in eGFR (OR, 2.90 95% CI: 1.59 to 5.29). When evaluating individual mineral abnormalities, hypocalcemia was associated with AKD and low eGFR while hyperparathyroidism was associated with low eGFR, AKI and structural injury. Vitamin D deficiency was associated with structural kidney injury. Vitamin D deficiency, hyperparathryoidism, and increases in FGF23 and osteopontin predicted mortality (p < 0.05 for all). CONCLUSION MBD is common among children with SCA hospitalized with VOC. Biomarkers of kidney injury and bone health may help risk stratify children at risk of sMBD. Routine evaluation of sMBD in children with SCA may improve long-term bone health.
Collapse
Affiliation(s)
- Anthony Batte
- Child Health and Development Centre, Makerere University College of Health Sciences, Kampala, Uganda
| | - Philip Kasirye
- Department of Paediatrics and Child Health, Makerere University College of Health Sciences, Kampala, Uganda
| | - Reagan Baluku
- Department of Paediatrics and Child Health, Makerere University College of Health Sciences, Kampala, Uganda
| | - Sarah Kiguli
- Department of Paediatrics and Child Health, Makerere University College of Health Sciences, Kampala, Uganda
| | - Robert Kalyesubula
- Department of Physiology, Makerere University College of Health Sciences, Kampala, Uganda
| | - Chandy C John
- Department of Pediatrics, Indiana University School of Medicine, Indianapolis, IN, United States.,Ryan White Center for Pediatric Infectious Diseases and Global Health, Indianapolis, IN, United States
| | - Andrew L Schwaderer
- Department of Pediatrics, Indiana University School of Medicine, Indianapolis, IN, United States
| | - Erik A Imel
- Department of Pediatrics, Indiana University School of Medicine, Indianapolis, IN, United States
| | - Andrea L Conroy
- Department of Pediatrics, Indiana University School of Medicine, Indianapolis, IN, United States.,Ryan White Center for Pediatric Infectious Diseases and Global Health, Indianapolis, IN, United States
| |
Collapse
|
20
|
Abstract
PURPOSE OF REVIEW Fibroblast growth factor 23 (FGF23) is a bone- and bone marrow-derived hormone that is critical to maintain phosphate homeostasis. The principal actions of FGF23 are to reduce serum phosphate levels by decreasing kidney phosphate reabsorption and 1,25-dihydroxyvitamin D synthesis. FGF23 deficiency causes hyperphosphatemia and ectopic calcifications, while FGF23 excess causes hypophosphatemia and skeletal defects. Excess FGF23 also correlates with kidney disease, where it is associated with increased morbidity and mortality. Accordingly, FGF23 levels are tightly regulated, but the mechanisms remain incompletely understood. RECENT FINDINGS In addition to bone mineral factors, additional factors including iron, erythropoietin, inflammation, energy, and metabolism regulate FGF23. All these factors affect Fgf23 expression, while some also regulate FGF23 protein cleavage. Conversely, FGF23 may have a functional role in regulating these biologic processes. Understanding the bi-directional relationship between FGF23 and non-bone mineral factors is providing new insights into FGF23 regulation and function.
Collapse
Affiliation(s)
- Petra Simic
- Nephrology Division, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA.
- Endocrine Unit, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA.
| | - Jodie L Babitt
- Nephrology Division, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA.
- Program in Membrane Biology, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA.
| |
Collapse
|
21
|
Noe KM, Ngo JP, Martin A, Zhu MZL, Cochrane AD, Smith JA, Thrift AG, Singh H, Evans RG. Intra-operative and early post-operative prediction of cardiac surgery-associated acute kidney injury: Urinary oxygen tension compared with plasma and urinary biomarkers. Clin Exp Pharmacol Physiol 2021; 49:228-241. [PMID: 34674291 DOI: 10.1111/1440-1681.13603] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2021] [Revised: 10/11/2021] [Accepted: 10/19/2021] [Indexed: 12/29/2022]
Abstract
Acute kidney injury (AKI) is a common and serious post-operative complication of cardiac surgery. The value of a predictive biomarker is determined not only by its predictive efficacy, but also by how early this prediction can be made. For a biomarker of cardiac surgery-associated AKI, this is ideally during the intra-operative period. Therefore, in 82 adult patients undergoing cardiac surgery requiring cardiopulmonary bypass (CPB), we prospectively compared the predictive efficacy of various blood and urinary biomarkers with that of continuous measurement of urinary oxygen tension (UPO2 ) at pre-determined intra- and post-operative time-points. None of the blood or urine biomarkers we studied showed predictive efficacy for post-operative AKI when measured intra-operatively. When treated as a binary variable (≤ or > median for the whole cohort), the earliest excess risk of AKI was predicted by an increase in urinary neutrophil gelatinase-associated lipocalin (NGAL) at 3 h after entry into the intensive care unit (odds ratio [95% confidence limits], 2.86 [1.14-7.21], p = 0.03). Corresponding time-points were 6 h for serum creatinine (3.59 [1.40-9.20], p = 0.008), and 24 h for plasma NGAL (4.54 [1.73-11.90], p = 0.002) and serum cystatin C (6.38 [2.35-17.27], p = 0.001). In contrast, indices of intra-operative urinary hypoxia predicted AKI after weaning from CPB, and in the case of a fall in UPO2 to ≤10 mmHg, during the rewarming phase of CPB (3.00 [1.19-7.56], p = 0.02). We conclude that continuous measurement of UPO2 predicts AKI earlier than plasma or urinary NGAL, serum cystatin C, or early post-operative changes in serum creatinine.
Collapse
Affiliation(s)
- Khin M Noe
- Cardiovascular Disease Program, Biomedicine Discovery Institute and Department of Physiology, Monash University, Melbourne, Victoria, Australia.,Department of Surgery, School of Clinical Sciences at Monash Health, Monash University, Melbourne, Victoria, Australia
| | - Jennifer P Ngo
- Cardiovascular Disease Program, Biomedicine Discovery Institute and Department of Physiology, Monash University, Melbourne, Victoria, Australia.,Department of Surgery, School of Clinical Sciences at Monash Health, Monash University, Melbourne, Victoria, Australia.,Department of Cardiac Physiology, National Cerebral and Cardiovascular Center Research Institute, Osaka, Japan
| | - Andrew Martin
- Cardiovascular Disease Program, Biomedicine Discovery Institute and Department of Physiology, Monash University, Melbourne, Victoria, Australia.,Department of Surgery, School of Clinical Sciences at Monash Health, Monash University, Melbourne, Victoria, Australia.,Department of Cardiothoracic Surgery, Monash Health, Monash University, Melbourne, Victoria, Australia
| | - Michael Z L Zhu
- Cardiovascular Disease Program, Biomedicine Discovery Institute and Department of Physiology, Monash University, Melbourne, Victoria, Australia.,Department of Surgery, School of Clinical Sciences at Monash Health, Monash University, Melbourne, Victoria, Australia.,Department of Cardiothoracic Surgery, Monash Health, Monash University, Melbourne, Victoria, Australia
| | - Andrew D Cochrane
- Department of Surgery, School of Clinical Sciences at Monash Health, Monash University, Melbourne, Victoria, Australia.,Department of Cardiothoracic Surgery, Monash Health, Monash University, Melbourne, Victoria, Australia
| | - Julian A Smith
- Department of Surgery, School of Clinical Sciences at Monash Health, Monash University, Melbourne, Victoria, Australia.,Department of Cardiothoracic Surgery, Monash Health, Monash University, Melbourne, Victoria, Australia
| | - Amanda G Thrift
- Department of Medicine, School of Clinical Sciences at Monash Health, Monash University, Melbourne, Victoria, Australia
| | - Harshil Singh
- Cardiovascular Disease Program, Biomedicine Discovery Institute and Department of Physiology, Monash University, Melbourne, Victoria, Australia
| | - Roger G Evans
- Cardiovascular Disease Program, Biomedicine Discovery Institute and Department of Physiology, Monash University, Melbourne, Victoria, Australia.,Department of Surgery, School of Clinical Sciences at Monash Health, Monash University, Melbourne, Victoria, Australia.,Pre-clinical Critical Care Unit, Florey Institute of Neuroscience and Mental Health, University of Melbourne, Melbourne, Victoria, Australia
| |
Collapse
|
22
|
Huang SS, Huang PH, Leu HB, Wu TC, Chen JW, Lin SJ. Significance of serum FGF-23 for risk assessment of contrast-associated acute kidney injury and clinical outcomes in patients undergoing coronary angiography. PLoS One 2021; 16:e0254835. [PMID: 34297744 PMCID: PMC8301629 DOI: 10.1371/journal.pone.0254835] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2021] [Accepted: 07/05/2021] [Indexed: 11/26/2022] Open
Abstract
Background Fibroblast growth factor (FGF)-23 levels rise as kidney function declines. Whether elevated FGF-23 levels are associated with an increased risk for contrast-associated acute kidney injury (CA-AKI) and major adverse cardiovascular events (MACE) in patients undergoing coronary angiography remain uncertain. Methods In total, 492 patients receiving coronary angiography were enrolled. Their serum FGF-23 levels were measured before administration of contrast media. The occurrence of CA-AKI was defined as a rise in serum creatinine of 0.5 mg/dL or a 25% increase from the baseline value within 48 h after the procedure. All patients were followed up for at least 1 year or until the occurrence of MACE including death, nonfatal myocardial infarction (MI), and ischemic stroke. Results Overall, CA-AKI occurred in 41 (8.3%) patients. During a median follow-up of 2.6 years, there were 24 deaths, 3 nonfatal MIs, and 7 ischemic strokes. Compared with those in the lowest FGF-23 tertile, individuals in the highest FGF-23 tertile had a significantly higher incidence of CA-AKI (P < 0.001) and lower incidence of MACE-free survival (P = 0.001). In multivariate regression analysis, higher FGF-23 level was found to be independently associated with a graded risk for CA-AKI (OR per doubling, 1.90; 95% CI 1.48–2.44) and MACE (HR per doubling, 1.25; 95% CI 1.02–1.52). Conclusions Elevated FGF-23 levels were associated with an increased risk for CA-AKI and future MACE among patients undergoing coronary angiography. FGF-23 may play a role in early diagnosis of CA-AKI and predicting clinical outcomes after coronary angiography.
Collapse
Affiliation(s)
- Shao-Sung Huang
- Division of Cardiology, Department of Internal Medicine, Taipei Veterans General Hospital, Taipei, Taiwan
- Healthcare and Management Center, Taipei Veterans General Hospital, Taipei, Taiwan
- School of Medicine, National Yang Ming Chiao Tung University, Tainan City, Taiwan
| | - Po-Hsun Huang
- Division of Cardiology, Department of Internal Medicine, Taipei Veterans General Hospital, Taipei, Taiwan
- Department of Critical Care Medicine, Taipei Veterans General Hospital, Taipei, Taiwan
- Institute of Clinical Medicine, Cardiovascular Research Center, National Yang Ming Chiao Tung University, Tainan City, Taiwan
- * E-mail:
| | - Hsin-Bang Leu
- Division of Cardiology, Department of Internal Medicine, Taipei Veterans General Hospital, Taipei, Taiwan
- Healthcare and Management Center, Taipei Veterans General Hospital, Taipei, Taiwan
- Institute of Clinical Medicine, Cardiovascular Research Center, National Yang Ming Chiao Tung University, Tainan City, Taiwan
| | - Tao-Cheng Wu
- Division of Cardiology, Department of Internal Medicine, Taipei Veterans General Hospital, Taipei, Taiwan
- Institute of Clinical Medicine, Cardiovascular Research Center, National Yang Ming Chiao Tung University, Tainan City, Taiwan
| | - Jaw-Wen Chen
- Division of Cardiology, Department of Internal Medicine, Taipei Veterans General Hospital, Taipei, Taiwan
- Healthcare and Management Center, Taipei Veterans General Hospital, Taipei, Taiwan
- School of Medicine, National Yang Ming Chiao Tung University, Tainan City, Taiwan
| | - Shing-Jong Lin
- Division of Cardiology, Department of Internal Medicine, Taipei Veterans General Hospital, Taipei, Taiwan
- Healthcare and Management Center, Taipei Veterans General Hospital, Taipei, Taiwan
- Institute of Clinical Medicine, Cardiovascular Research Center, National Yang Ming Chiao Tung University, Tainan City, Taiwan
| |
Collapse
|
23
|
Trivedi N, Kumar D. Fibroblast growth factor and kidney disease: Updates for emerging novel therapeutics. J Cell Physiol 2021; 236:7909-7925. [PMID: 34196395 DOI: 10.1002/jcp.30497] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2021] [Revised: 05/04/2021] [Accepted: 05/28/2021] [Indexed: 01/01/2023]
Abstract
The discovery of fibroblast growth factors (FGFs) and fibroblast growth factor receptors (FGFRs) provided a profound new insight into physiological and metabolic functions. FGF has a large family by having divergent structural elements and enable functional divergence and specification. FGF and FGFRs are highly expressed during kidney development. Signals from the ureteric bud regulate morphogenesis, nephrogenesis, and nephron progenitor survival. Thus, FGF signaling plays an important role in kidney progenitor cell aggregation at the sites of new nephron formation. This review will summarize the current knowledge about functions of FGF signaling in kidney development and their ability to promote regeneration in injured kidneys and its use as a biomarker and therapeutic target in kidney diseases. Further studies are essential to determine the predictive significance of the various FGF/FGFR deviations and to integrate them into clinical algorithms.
Collapse
Affiliation(s)
- Neerja Trivedi
- Department of Neurological Sciences, University of Nebraska Medical Center, Omaha, Nebraska, USA
| | - Devendra Kumar
- Department of Pharmaceutical Sciences, College of Pharmacy, University of Nebraska Medical Center, Omaha, Nebraska, USA
| |
Collapse
|
24
|
Glycerol-3-phosphate and fibroblast growth factor 23 regulation. Curr Opin Nephrol Hypertens 2021; 30:397-403. [PMID: 33901058 PMCID: PMC8312345 DOI: 10.1097/mnh.0000000000000715] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
Abstract
PURPOSE OF REVIEW Both classical and nonclassical factors regulate fibroblast growth factor 23 (FGF23), with impacts on gene expression and proteolytic cleavage. Here, we review recent publications that extend current knowledge on these factors. RECENT FINDINGS Emerging nonclassical FGF23 regulators such as erythropoietin cause a balanced increase in FGF23 expression and cleavage, with minimal or no increase in biologically active intact FGF23 (iFGF23) in blood. However, circulating FGF23 profiles may not reflect the bone marrow microenvironment. For example, granulocyte colony-stimulating factor increases local marrow iFGF23 levels without impacting circulating iFGF23 levels. The view that phosphate does not increase bone FGF23 production also warrants reconsideration, as phosphate can reduce iFGF23 cleavage and phosphate-containing calciprotein particles increase FGF23 expression. Finally, a screen of renal venous plasma identifies glycerol-3-phosphate as a kidney-derived molecule that circulates to bone and bone marrow, where it is converted to lysophosphatidic acid and signals through a G-protein coupled receptor to increase FGF23 synthesis. SUMMARY FGF23 regulation is complex, requiring consideration of known and emerging stimuli, expression and cleavage, and circulating and local levels. Recent work identifies glycerol-3-phosphate as an FGF23 regulator derived from the injured kidney; whether it participates in FGF23 production downstream of classical or nonclassical factors requires further study.
Collapse
|
25
|
Jiang S, Huang L, Zhang W, Zhang H. Vitamin D/VDR in Acute Kidney Injury: A Potential Therapeutic Target. Curr Med Chem 2021; 28:3865-3876. [PMID: 33213307 DOI: 10.2174/0929867327666201118155625] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2020] [Revised: 09/29/2020] [Accepted: 10/07/2020] [Indexed: 11/22/2022]
Abstract
Despite many strategies and parameters used in clinical practice, the incidence and mortality of acute kidney injury (AKI) are still high with poor prognosis. With the development of molecular biology, the role of vitamin D and vitamin D receptor (VDR) in AKI is drawing increasing attention. Accumulated researches have suggested that Vitamin D deficiency is a risk factor of both clinical and experimental AKI, and vitamin D/VDR could be a promising therapeutic target against AKI. However, more qualitative clinical researches are needed to provide stronger evidence for the clinical application of vitamin D and VDR agonists in the future. Issues like the route and dosage of administration also await more attention. The present review aims to summarize the current works on the role of vitamin D/VDR in AKI and provides some new insight on its therapeutic potential.
Collapse
Affiliation(s)
- Siqing Jiang
- Department of Nephrology, Third Xiangya Hospital, Central South University, 138 Tongzipo Rd, Changsha, Hunan 410013, China
| | - Lihua Huang
- Center for Medical Experiments, Third Xiangya Hospital, Central South University, Changsha, Hunan 410013, China
| | - Wei Zhang
- Department of Nephrology, Third Xiangya Hospital, Central South University, 138 Tongzipo Rd, Changsha, Hunan 410013, China
| | - Hao Zhang
- Department of Nephrology, Third Xiangya Hospital, Central South University, 138 Tongzipo Rd, Changsha, Hunan 410013, China
| |
Collapse
|
26
|
FGF23 ameliorates ischemia-reperfusion induced acute kidney injury via modulation of endothelial progenitor cells: targeting SDF-1/CXCR4 signaling. Cell Death Dis 2021; 12:409. [PMID: 33866326 PMCID: PMC8053200 DOI: 10.1038/s41419-021-03693-w] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/25/2020] [Revised: 03/30/2021] [Accepted: 04/01/2021] [Indexed: 01/09/2023]
Abstract
The levels of fibroblast growth factor 23 (FGF23) rapidly increases after acute kidney injury (AKI). However, the role of FGF23 in AKI is still unclear. Here, we observe that pretreatment with FGF23 protein into ischemia-reperfusion induced AKI mice ameliorates kidney injury by promoting renal tubular regeneration, proliferation, vascular repair, and attenuating tubular damage. In vitro assays demonstrate that SDF-1 induces upregulation of its receptor CXCR4 in endothelial progenitor cells (EPCs) via a non-canonical NF-κB signaling pathway. FGF23 crosstalks with the SDF-1/CXCR4 signaling and abrogates SDF-1-induced EPC senescence and migration, but not angiogenesis, in a Klotho-independent manner. The downregulated pro-angiogenic IL-6, IL-8, and VEGF-A expressions after SDF-1 infusion are rescued after adding FGF23. Diminished therapeutic ability of SDF-1-treated EPCs is counteracted by FGF23 in a SCID mouse in vivo AKI model. Together, these data highlight a revolutionary and important role that FGF23 plays in the nephroprotection of IR-AKI.
Collapse
|
27
|
Hu PP, Bao JF, Li A. Roles for fibroblast growth factor-23 and α-Klotho in acute kidney injury. Metabolism 2021; 116:154435. [PMID: 33220250 DOI: 10.1016/j.metabol.2020.154435] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/29/2020] [Revised: 11/08/2020] [Accepted: 11/13/2020] [Indexed: 12/21/2022]
Abstract
Acute kidney injury is a global disease with high morbidity and mortality. Recent studies have revealed that the fibroblast growth factor-23-α-Klotho axis is closely related to chronic kidney disease, and has multiple biological functions beyond bone-mineral metabolism. However, although dysregulation of fibroblast growth factor-23-α-Klotho has been observed in acute kidney injury, the role of fibroblast growth factor-23-α-Klotho in the pathophysiology of acute kidney injury remains largely unknown. In this review, we describe recent findings regarding fibroblast growth factor-23-α-Klotho, which is mainly involved in inflammation, oxidative stress, and hemodynamic disorders. Further, based on these recent results, we put forth novel insights regarding the relationship between the fibroblast growth factor-23-α-Klotho axis and acute kidney injury, which may provide new therapeutic targets for treating acute kidney injury.
Collapse
Affiliation(s)
- Pan-Pan Hu
- State Key Laboratory of Organ Failure Research, National Clinical Research Center for Kidney Disease, Nanfang Hospital, Southern Medical University, 510515 Guangzhou, China; Guangdong Provincial Key Laboratory of Renal Failure Research, Guangzhou Regenerative Medicine and Health Guangdong Laboratory, 510005 Guangzhou, China
| | - Jing-Fu Bao
- State Key Laboratory of Organ Failure Research, National Clinical Research Center for Kidney Disease, Nanfang Hospital, Southern Medical University, 510515 Guangzhou, China; Guangdong Provincial Key Laboratory of Renal Failure Research, Guangzhou Regenerative Medicine and Health Guangdong Laboratory, 510005 Guangzhou, China
| | - Aiqing Li
- State Key Laboratory of Organ Failure Research, National Clinical Research Center for Kidney Disease, Nanfang Hospital, Southern Medical University, 510515 Guangzhou, China; Guangdong Provincial Key Laboratory of Renal Failure Research, Guangzhou Regenerative Medicine and Health Guangdong Laboratory, 510005 Guangzhou, China.
| |
Collapse
|
28
|
Sun S, Liu Z, Chen C, Wang Z, Jin H, Meng X, Dai B, Zhang L, Zhou C, Xue C, Li X. Serum fibroblast growth factor 23 for early detection of acute kidney injury in critical illness. Am J Transl Res 2021; 13:12141-12151. [PMID: 34956442 PMCID: PMC8661170] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2021] [Accepted: 10/12/2021] [Indexed: 03/18/2023]
Abstract
BACKGROUND Serum fibroblast growth factor 23 (FGF23) is associated with acute kidney injury (AKI) and mortality in patients with critical illnesses. However, the accurate predictive performance of FGF23 on AKI remains inconclusive. METHODS Meta-analysis was performed using data sources including PubMed, Web of Science, EMBASE, and Cochrane (until June 1, 2021). Cohort or observational studies including patients with AKI and serum FGF23 level as the index test were included. The primary outcome was the AKI detective accuracy. This study has been registered in PROSPERO (CRD42021249930). RESULTS Eleven studies with 1946 patients in seven countries were included. Across all settings, the sensitivity and specificity for serum FGF23 levels to predict AKI were 82% (95% CI, 66-91%) and 77% (95% CI, 67-85%), respectively. The diagnostic odds ratio of FGF23 was 15.51 (95% CI, 4.89-49.19), with the pooled positive likelihood ratio of 3.62 (95% CI, 2.25-5.83) and a negative likelihood ratio of 0.23 (95% CI, 0.11-0.50). The area under the receiver operating characteristic curve to detect AKI was 0.86 (95% CI, 0.82-0.88). C-terminal FGF23 had a better performance than intact FGF23. CONCLUSIONS Plasma FGF23 is a valuable biomarker for incident AKI in critically ill patients. Comparisons of FGF23 with other biomarkers in AKI still need more studies to prove.
Collapse
Affiliation(s)
- Shu Sun
- Department of Urology, The 8th Medical Center of Chinese PLA General Hospital Beijing 100091, China
| | - Zhijia Liu
- Department of Urology, The 8th Medical Center of Chinese PLA General Hospital Beijing 100091, China
| | - Changqing Chen
- Department of Urology, The 8th Medical Center of Chinese PLA General Hospital Beijing 100091, China
| | - Zhisong Wang
- Department of Urology, The 8th Medical Center of Chinese PLA General Hospital Beijing 100091, China
| | - Hailong Jin
- Organ Transplantation Center, The 3rd Medical Center of Chinese PLA General Hospital Beijing 100039, China
| | - Xiaoyun Meng
- Department of Urology, The 8th Medical Center of Chinese PLA General Hospital Beijing 100091, China
| | - Bing Dai
- Division of Nephrology, Changzheng Hospital, Second Military Medical University Shanghai 200003, China
| | - Liming Zhang
- Department of Nephrology, Zhabei Central Hospital of Jingan District of Shanghai Shanghai 200000, China
| | - Chenchen Zhou
- Outpatient Department, Yangpu Third Military Retreat Shanghai 200000, China.,Department of Nephrology, Yueyang Hospital Shanghai 200000, China
| | - Cheng Xue
- Department of Urology, The 8th Medical Center of Chinese PLA General Hospital Beijing 100091, China.,Division of Nephrology, Changzheng Hospital, Second Military Medical University Shanghai 200003, China
| | - Xiang Li
- Department of Urology, The 8th Medical Center of Chinese PLA General Hospital Beijing 100091, China
| |
Collapse
|
29
|
Pramong N, Gojaseni P, Suttipongkeat S, Kiattisunthorn K, Chittinandana A. Diagnostic accuracy of fibroblast growth factor 23 for predicting acute kidney injury in patients with acute decompensated heart failure. Nephrology (Carlton) 2020; 26:126-133. [PMID: 32902010 DOI: 10.1111/nep.13780] [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: 06/02/2020] [Revised: 08/27/2020] [Accepted: 08/30/2020] [Indexed: 11/30/2022]
Abstract
BACKGROUND Elevated plasma levels of fibroblast growth factor 23 (FGF23) have emerged as a predictor for the development of acute kidney injury (AKI) in patients undergoing cardiac surgery and those with critical illnesses. However, accurate data in cases involving acute decompensated heart failure (ADHF) remains limited. METHODS Single centre cohort study was performed in patients admitted for ADHF. Plasma c-terminal FGF23 (c-FGF23) was measured at baseline and 24 hours after being diagnosed with ADHF. AKI was defined by KDIGO 2012 criteria. RESULTS The study enrolled 62 patients diagnosed with ADHF. The incidence of AKI was 45% and significantly increased the risk of death. Patients developing AKI had significantly higher levels of plasma c-FGF23 at baseline in comparison with those not developing AKI [median value 1258.5 (57.2, 15 850) vs 230.2 (68.515 850) RU/mL, P = .005]. During the first 24 hours, plasma c-FGF23 levels decreased in both groups, and the levels of c-FGF23 at 24 hours were consistent with the baseline [861.8 (75.7, 15 850) vs 226.3 (56, 5450.8) RU/mL, P = .006]. Receiver operating characteristic analysis of both first time and second time for plasma c-FGF23 collection yielded an area under curve of 0.71 for the prediction of AKI incidence. With the cut-off point at 450 RU/mL, the sensitivity and specificity of plasma c-FGF23 at the baseline for predicting AKI were 71.4% and 61.8% respectively. CONCLUSION Plasma c-FGF23 may serve as a novel biomarker for development of AKI in patients with ADHF. These results should be revalidated in larger-scale, cohort studies.
Collapse
Affiliation(s)
- Nattha Pramong
- Division of Nephrology, Department of Medicine, Bhumibol Adulyadej Hospital, Bangkok, Thailand
| | - Pongsathorn Gojaseni
- Division of Nephrology, Department of Medicine, Bhumibol Adulyadej Hospital, Bangkok, Thailand
| | - Sarunyoo Suttipongkeat
- Division of Cardiology, Department of Medicine, Bhumibol Adulyadej Hospital, Bangkok, Thailand
| | - Kraiwiporn Kiattisunthorn
- Division of Nephrology, Department of Medicine, Faculty of Medicine, Siriraj Medical School and Hospital, Mahidol University, Bangkok, Thailand
| | - Anutra Chittinandana
- Division of Nephrology, Department of Medicine, Bhumibol Adulyadej Hospital, Bangkok, Thailand
| |
Collapse
|
30
|
Xie Y, Su N, Yang J, Tan Q, Huang S, Jin M, Ni Z, Zhang B, Zhang D, Luo F, Chen H, Sun X, Feng JQ, Qi H, Chen L. FGF/FGFR signaling in health and disease. Signal Transduct Target Ther 2020; 5:181. [PMID: 32879300 PMCID: PMC7468161 DOI: 10.1038/s41392-020-00222-7] [Citation(s) in RCA: 410] [Impact Index Per Article: 82.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2020] [Revised: 05/28/2020] [Accepted: 06/15/2020] [Indexed: 12/13/2022] Open
Abstract
Growing evidences suggest that the fibroblast growth factor/FGF receptor (FGF/FGFR) signaling has crucial roles in a multitude of processes during embryonic development and adult homeostasis by regulating cellular lineage commitment, differentiation, proliferation, and apoptosis of various types of cells. In this review, we provide a comprehensive overview of the current understanding of FGF signaling and its roles in organ development, injury repair, and the pathophysiology of spectrum of diseases, which is a consequence of FGF signaling dysregulation, including cancers and chronic kidney disease (CKD). In this context, the agonists and antagonists for FGF-FGFRs might have therapeutic benefits in multiple systems.
Collapse
Affiliation(s)
- Yangli Xie
- Department of Wound Repair and Rehabilitation Medicine, State Key Laboratory of Trauma, Burns and Combined Injury, Trauma Center, Research Institute of Surgery, Daping Hospital, Army Medical University, Chongqing, China.
| | - Nan Su
- Department of Wound Repair and Rehabilitation Medicine, State Key Laboratory of Trauma, Burns and Combined Injury, Trauma Center, Research Institute of Surgery, Daping Hospital, Army Medical University, Chongqing, China
| | - Jing Yang
- Department of Wound Repair and Rehabilitation Medicine, State Key Laboratory of Trauma, Burns and Combined Injury, Trauma Center, Research Institute of Surgery, Daping Hospital, Army Medical University, Chongqing, China
| | - Qiaoyan Tan
- Department of Wound Repair and Rehabilitation Medicine, State Key Laboratory of Trauma, Burns and Combined Injury, Trauma Center, Research Institute of Surgery, Daping Hospital, Army Medical University, Chongqing, China
| | - Shuo Huang
- Department of Wound Repair and Rehabilitation Medicine, State Key Laboratory of Trauma, Burns and Combined Injury, Trauma Center, Research Institute of Surgery, Daping Hospital, Army Medical University, Chongqing, China
| | - Min Jin
- Department of Wound Repair and Rehabilitation Medicine, State Key Laboratory of Trauma, Burns and Combined Injury, Trauma Center, Research Institute of Surgery, Daping Hospital, Army Medical University, Chongqing, China
| | - Zhenhong Ni
- Department of Wound Repair and Rehabilitation Medicine, State Key Laboratory of Trauma, Burns and Combined Injury, Trauma Center, Research Institute of Surgery, Daping Hospital, Army Medical University, Chongqing, China
| | - Bin Zhang
- Department of Wound Repair and Rehabilitation Medicine, State Key Laboratory of Trauma, Burns and Combined Injury, Trauma Center, Research Institute of Surgery, Daping Hospital, Army Medical University, Chongqing, China
| | - Dali Zhang
- Department of Wound Repair and Rehabilitation Medicine, State Key Laboratory of Trauma, Burns and Combined Injury, Trauma Center, Research Institute of Surgery, Daping Hospital, Army Medical University, Chongqing, China
| | - Fengtao Luo
- Department of Wound Repair and Rehabilitation Medicine, State Key Laboratory of Trauma, Burns and Combined Injury, Trauma Center, Research Institute of Surgery, Daping Hospital, Army Medical University, Chongqing, China
| | - Hangang Chen
- Department of Wound Repair and Rehabilitation Medicine, State Key Laboratory of Trauma, Burns and Combined Injury, Trauma Center, Research Institute of Surgery, Daping Hospital, Army Medical University, Chongqing, China
| | - Xianding Sun
- Department of Wound Repair and Rehabilitation Medicine, State Key Laboratory of Trauma, Burns and Combined Injury, Trauma Center, Research Institute of Surgery, Daping Hospital, Army Medical University, Chongqing, China
| | - Jian Q Feng
- Department of Biomedical Sciences, Texas A&M University College of Dentistry, Dallas, TX, 75246, USA
| | - Huabing Qi
- Department of Wound Repair and Rehabilitation Medicine, State Key Laboratory of Trauma, Burns and Combined Injury, Trauma Center, Research Institute of Surgery, Daping Hospital, Army Medical University, Chongqing, China.
| | - Lin Chen
- Department of Wound Repair and Rehabilitation Medicine, State Key Laboratory of Trauma, Burns and Combined Injury, Trauma Center, Research Institute of Surgery, Daping Hospital, Army Medical University, Chongqing, China.
| |
Collapse
|
31
|
Neyra JA, Hu MC, Moe OW. Fibroblast Growth Factor 23 and αKlotho in Acute Kidney Injury: Current Status in Diagnostic and Therapeutic Applications. Nephron Clin Pract 2020; 144:665-672. [PMID: 32841947 DOI: 10.1159/000509856] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2020] [Accepted: 06/30/2020] [Indexed: 12/25/2022] Open
Abstract
Fibroblast growth factor (FGF) 23 and αKlotho are circulating mineral regulatory substances that also have a very diverse range of actions. Acute kidney injury (AKI) is a state of high FGF23 and low αKlotho. Clinical association data for FGF23 are strong, but the basic pathobiology of FGF23 in AKI is rather sparse. Conversely, preclinical data supporting a pathogenic role of αKlotho in AKI are strong, but the human data are still being generated. This pair of substances can potentially serve as diagnostic and prognostic biomarkers. FGF23 blockade and αKlotho restoration can have prophylactic and therapeutic utility in AKI. The literature to date is briefly reviewed in this article.
Collapse
Affiliation(s)
- Javier A Neyra
- Charles and Jane Pak Center for Mineral Metabolism and Clinical Research, Dallas, Texas, USA.,Department of Internal Medicine, University of Texas Southwestern Medical Center, Dallas, Texas, USA.,Department of Medicine, Division of Nephrology, Bone and Mineral Metabolism, University of Kentucky, Lexington, Kentucky, USA
| | - Ming Chang Hu
- Charles and Jane Pak Center for Mineral Metabolism and Clinical Research, Dallas, Texas, USA.,Department of Internal Medicine, University of Texas Southwestern Medical Center, Dallas, Texas, USA
| | - Orson W Moe
- Charles and Jane Pak Center for Mineral Metabolism and Clinical Research, Dallas, Texas, USA, .,Department of Internal Medicine, University of Texas Southwestern Medical Center, Dallas, Texas, USA, .,Department of Physiology, University of Texas Southwestern Medical Center, Dallas, Texas, USA,
| |
Collapse
|
32
|
Inflammation both increases and causes resistance to FGF23 in normal and uremic rats. Clin Sci (Lond) 2020; 134:15-32. [PMID: 31860056 DOI: 10.1042/cs20190779] [Citation(s) in RCA: 20] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2019] [Revised: 12/03/2019] [Accepted: 12/20/2019] [Indexed: 12/19/2022]
Abstract
Fibroblast growth factor 23 (FGF23) increases phosphorus excretion and decreases calcitriol (1,25(OH)2D) levels. FGF23 increases from early stages of renal failure. We evaluated whether strict control of phosphorus intake in renal failure prevents the increase in FGF23 and to what extent inflammation impairs regulation of FGF23. The study was performed in 5/6 nephrectomized (Nx) Wistar rats fed diets containing 0.2-1.2% phosphorus for 3 or 15 days. FGF23 levels significantly increased in all Nx groups in the short-term (3-day) experiment. However, at 15 days, FGF23 increased in all Nx rats except in those fed 0.2% phosphorus. In a second experiment, Nx rats fed low phosphorus diets (0.2 and 0.4%) for 15 days received daily intraperitoneal lipopolysaccharide (LPS) injections to induce inflammation. In these rats, FGF23 increased despite the low phosphorus diets. Thus, higher FGF23 levels were needed to maintain phosphaturia and normal serum phosphorus values. Renal Klotho expression was preserved in Nx rats on a 0.2% phosphorus diet, reduced on a 0.4% phosphorus diet, and markedly reduced in Nx rats receiving LPS. In ex vivo experiments, high phosphorus and LPS increased nuclear β-catenin and p65-NFκB and decreased Klotho. Inhibition of inflammation and Wnt signaling activation resulted in decreased FGF23 levels and increased renal Klotho. In conclusion, strict control of phosphorus intake prevented the increase in FGF23 in renal failure, whereas inflammation independently increased FGF23 values. Decreased Klotho may explain the renal resistance to FGF23 in inflammation. These effects are likely mediated by the activation of NFkB and Wnt/β-catenin signaling.
Collapse
|
33
|
Erythropoietin, Fibroblast Growth Factor 23, and Death After Kidney Transplantation. J Clin Med 2020; 9:jcm9061737. [PMID: 32512806 PMCID: PMC7356141 DOI: 10.3390/jcm9061737] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2020] [Revised: 06/01/2020] [Accepted: 06/03/2020] [Indexed: 12/02/2022] Open
Abstract
Elevated levels of erythropoietin (EPO) are associated with an increased risk of death in renal transplant recipients (RTRs), but the underlying mechanisms remain unclear. Emerging data suggest that EPO stimulates production of the phosphaturic hormone fibroblast growth factor 23 (FGF23), another strong risk factor for death in RTRs. We hypothesized that the hitherto unexplained association between EPO levels and adverse outcomes may be attributable to increased levels of FGF23. We included 579 RTRs (age 51 ± 12 years, 55% males) from the TransplantLines Insulin Resistance and Inflammation Cohort study (NCT03272854). During a follow-up of 7.0 years, 121 RTRs died, of which 62 were due to cardiovascular cause. In multivariable Cox regression analysis, EPO was independently associated with all-cause (HR, 1.66; 95% CI 1.16–2.36; P = 0.005) and cardiovascular death (HR, 1.87; 95% CI 1.14–3.06; P = 0.01). However, the associations were abrogated following adjustment for FGF23 (HR, 1.28; 95% CI 0.87–1.88; P = 0.20, and HR, 1.45; 95% CI 0.84–2.48; P = 0.18, respectively). In subsequent mediation analysis, FGF23 mediated 72% and 50% of the association between EPO and all-cause and cardiovascular death, respectively. Our results underline the strong relationship between EPO and FGF23 physiology, and provide a potential mechanism underlying the relationship between increased EPO levels and adverse outcomes in RTRs.
Collapse
|
34
|
Fayed A, Radwan WA, Amin M, Gamal A. Prediction of mortality and need for renal replacement therapy in patients of acute kidney injury using fibroblast growth factor 23. SAUDI JOURNAL OF KIDNEY DISEASES AND TRANSPLANTATION 2020; 30:1044-1051. [PMID: 31696842 DOI: 10.4103/1319-2442.270259] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/04/2022] Open
Abstract
Acute kidney injury (AKI) occurs in up to 50% of patients admitted to the intensive care units. Fibroblast growth factor 23 (FGF23), which plays an important role in regulating phosphate, rises early in AKI. Few studies were conducted to correlate the level of FGF23 and adverse outcomes in AKI. The study was conducted on 30 participants with AKI, which was defined according to AKI network criteria, admitted to the Critical Care Department, Kasr El Aini Hospital, Cairo University between July 2016 and May 2017, and serum FGF23 was measured within 24 h of AKI onset to correlate the level of FGF23 with mortality and need for renal replacement therapy (RRT). Enrollment FGF23 levels were significantly higher among patients who died than in the survival group (mean level: 544.2 vs. 59.3 pg/mL, P = 0.004). Furthermore, FGF23 levels were significantly higher in patients who needed RRT than in other participants (mean level: 529.5 vs. 285.11 pg/mL, P = 0.04). There was a statistically significant positive relationship between FGF23 level and sequential organ failure assessment score (P = 0.03). In patients with AKI, higher FGF23 levels are associated with increased risk of mortality and need for RRT.
Collapse
Affiliation(s)
- Ahmed Fayed
- Department of Internal Medicine, Nephrology Unit, Kasr Al Aini School of Medicine, Cairo University, Cairo, Egypt
| | - Waheed Ahmed Radwan
- Department of Critical Care, Kasr El Aini School of Medicine, Cairo University, Cairo, Egypt
| | - Mohamed Amin
- Department of Critical Care, Kasr El Aini School of Medicine, Cairo University, Cairo, Egypt
| | - Ahmed Gamal
- Department of Critical Care, Kasr El Aini School of Medicine, Cairo University, Cairo, Egypt
| |
Collapse
|
35
|
Simic P, Kim W, Zhou W, Pierce KA, Chang W, Sykes DB, Aziz NB, Elmariah S, Ngo D, Pajevic PD, Govea N, Kestenbaum BR, de Boer IH, Cheng Z, Christov M, Chun J, Leaf DE, Waikar SS, Tager AM, Gerszten RE, Thadhani RI, Clish CB, Jüppner H, Wein MN, Rhee EP. Glycerol-3-phosphate is an FGF23 regulator derived from the injured kidney. J Clin Invest 2020; 130:1513-1526. [PMID: 32065590 PMCID: PMC7269595 DOI: 10.1172/jci131190] [Citation(s) in RCA: 80] [Impact Index Per Article: 16.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2019] [Accepted: 12/11/2019] [Indexed: 12/24/2022] Open
Abstract
Fibroblast growth factor 23 (FGF23) is a bone-derived hormone that controls blood phosphate levels by increasing renal phosphate excretion and reducing 1,25-dihydroxyvitamin D3 [1,25(OH)2D] production. Disorders of FGF23 homeostasis are associated with significant morbidity and mortality, but a fundamental understanding of what regulates FGF23 production is lacking. Because the kidney is the major end organ of FGF23 action, we hypothesized that it releases a factor that regulates FGF23 synthesis. Using aptamer-based proteomics and liquid chromatography-mass spectrometry-based (LC-MS-based) metabolomics, we profiled more than 1600 molecules in renal venous plasma obtained from human subjects. Renal vein glycerol-3-phosphate (G-3-P) had the strongest correlation with circulating FGF23. In mice, exogenous G-3-P stimulated bone and bone marrow FGF23 production through local G-3-P acyltransferase-mediated (GPAT-mediated) lysophosphatidic acid (LPA) synthesis. Further, the stimulatory effect of G-3-P and LPA on FGF23 required LPA receptor 1 (LPAR1). Acute kidney injury (AKI), which increases FGF23 levels, rapidly increased circulating G-3-P in humans and mice, and the effect of AKI on FGF23 was abrogated by GPAT inhibition or Lpar1 deletion. Together, our findings establish a role for kidney-derived G-3-P in mineral metabolism and outline potential targets to modulate FGF23 production during kidney injury.
Collapse
Affiliation(s)
- Petra Simic
- Nephrology Division and.,Endocrine Unit, Endocrinology Division, Department of Medicine, Massachusetts General Hospital, Boston, Massachusetts, USA
| | - Wondong Kim
- Nephrology Division and.,Endocrine Unit, Endocrinology Division, Department of Medicine, Massachusetts General Hospital, Boston, Massachusetts, USA
| | - Wen Zhou
- Nephrology Division and.,Endocrine Unit, Endocrinology Division, Department of Medicine, Massachusetts General Hospital, Boston, Massachusetts, USA
| | - Kerry A Pierce
- Broad Institute, Massachusetts Institute of Technology and Harvard University, Cambridge, Massachusetts, USA
| | - Wenhan Chang
- Endocrine Research Unit, San Francisco Veterans Affairs Medical Center, UCSF, San Francisco, California, USA
| | | | | | - Sammy Elmariah
- Cardiology Division, Department of Medicine, Massachusetts General Hospital, Boston, Massachusetts, USA
| | - Debby Ngo
- Cardiovascular Research Center, Division of Cardiovascular Medicine, Department of Medicine, Beth Israel Deaconess Medical Center, Boston, Massachusetts, USA
| | - Paola Divieti Pajevic
- Department of Molecular and Cell Biology, Henry M. Goldman School of Dental Medicine, Boston University, Boston, Massachusetts, USA
| | - Nicolas Govea
- Endocrine Unit, Endocrinology Division, Department of Medicine, Massachusetts General Hospital, Boston, Massachusetts, USA
| | - Bryan R Kestenbaum
- Kidney Research Institute, University of Washington Medicine and Northwest Kidney Centers, Seattle, Washington, USA
| | - Ian H de Boer
- Kidney Research Institute, University of Washington Medicine and Northwest Kidney Centers, Seattle, Washington, USA
| | - Zhiqiang Cheng
- Endocrine Research Unit, San Francisco Veterans Affairs Medical Center, UCSF, San Francisco, California, USA
| | - Marta Christov
- Department of Medicine, New York Medical College, Touro College, Valhalla, New York, USA
| | - Jerold Chun
- Sanford Burnham Prebys Medical Discovery Institute, La Jolla, California, USA
| | - David E Leaf
- Division of Renal (Kidney) Medicine, Department of Medicine, Brigham and Women's Hospital, Boston, Massachusetts, USA
| | - Sushrut S Waikar
- Renal Section, Department of Medicine, Boston University Medical Center, Boston, Massachusetts, USA
| | - Andrew M Tager
- Division of Pulmonary and Critical Care Medicine, Department of Medicine, and
| | - Robert E Gerszten
- Broad Institute, Massachusetts Institute of Technology and Harvard University, Cambridge, Massachusetts, USA.,Cardiovascular Research Center, Division of Cardiovascular Medicine, Department of Medicine, Beth Israel Deaconess Medical Center, Boston, Massachusetts, USA
| | | | - Clary B Clish
- Broad Institute, Massachusetts Institute of Technology and Harvard University, Cambridge, Massachusetts, USA
| | - Harald Jüppner
- Endocrine Unit, Endocrinology Division, Department of Medicine, Massachusetts General Hospital, Boston, Massachusetts, USA.,Pediatric Nephrology and Hypertension Program, Mass General for Children, Massachusetts General Hospital, Boston, Massachusetts, USA
| | - Marc N Wein
- Endocrine Unit, Endocrinology Division, Department of Medicine, Massachusetts General Hospital, Boston, Massachusetts, USA
| | - Eugene P Rhee
- Nephrology Division and.,Endocrine Unit, Endocrinology Division, Department of Medicine, Massachusetts General Hospital, Boston, Massachusetts, USA.,Broad Institute, Massachusetts Institute of Technology and Harvard University, Cambridge, Massachusetts, USA
| |
Collapse
|
36
|
Gao L, Zhong X, Jin J, Li J, Meng XM. Potential targeted therapy and diagnosis based on novel insight into growth factors, receptors, and downstream effectors in acute kidney injury and acute kidney injury-chronic kidney disease progression. Signal Transduct Target Ther 2020; 5:9. [PMID: 32296020 PMCID: PMC7018831 DOI: 10.1038/s41392-020-0106-1] [Citation(s) in RCA: 66] [Impact Index Per Article: 13.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2019] [Revised: 12/01/2019] [Accepted: 12/17/2019] [Indexed: 02/08/2023] Open
Abstract
Acute kidney injury (AKI) is defined as a rapid decline in renal function and is characterized by excessive renal inflammation and programmed death of resident cells. AKI shows high morbidity and mortality, and severe or repeated AKI can transition to chronic kidney disease (CKD) or even end-stage renal disease (ESRD); however, very few effective and specific therapies are available, except for supportive treatment. Growth factors, such as epidermal growth factor (EGF), insulin-like growth factor (IGF), and transforming growth factor-β (TGF-β), are significantly altered in AKI models and have been suggested to play critical roles in the repair process of AKI because of their roles in cell regeneration and renal repair. In recent years, a series of studies have shown evidence that growth factors, receptors, and downstream effectors may be highly involved in the mechanism of AKI and may function in the early stage of AKI in response to stimuli by regulating inflammation and programmed cell death. Moreover, certain growth factors or correlated proteins act as biomarkers for AKI due to their sensitivity and specificity. Furthermore, growth factors originating from mesenchymal stem cells (MSCs) via paracrine signaling or extracellular vesicles recruit leukocytes or repair intrinsic cells and may participate in AKI repair or the AKI-CKD transition. In addition, growth factor-modified MSCs show superior therapeutic potential compared to that of unmodified controls. In this review, we summarized the current therapeutic and diagnostic strategies targeting growth factors to treat AKI in clinical trials. We also evaluated the possibilities of other growth factor-correlated molecules as therapeutic targets in the treatment of AKI and the AKI-CKD transition.
Collapse
Affiliation(s)
- Li Gao
- The Key Laboratory of Major Autoimmune Diseases, Anhui Institute of Innovative Drugs, School of Pharmacy, Anhui Medical University, 230032, Hefei, China
| | - Xiang Zhong
- Department of Nephrology, Sichuan Academy of Medical Sciences & Sichuan Provincial People's Hospital, School of Medicine, University of Electronic Science and Technology of China, 610072, Chengdu, Sichuan, China
| | - Juan Jin
- Department of Pharmacology, Key Laboratory of Anti-inflammatory and Immunopharmacology, Ministry of Education, Anhui Medical University, 230032, Hefei, China
| | - Jun Li
- The Key Laboratory of Major Autoimmune Diseases, Anhui Institute of Innovative Drugs, School of Pharmacy, Anhui Medical University, 230032, Hefei, China
| | - Xiao-Ming Meng
- The Key Laboratory of Major Autoimmune Diseases, Anhui Institute of Innovative Drugs, School of Pharmacy, Anhui Medical University, 230032, Hefei, China.
| |
Collapse
|
37
|
Hegde A, Denburg MR, Glenn DA. Acute Kidney Injury and Pediatric Bone Health. Front Pediatr 2020; 8:635628. [PMID: 33634055 PMCID: PMC7900149 DOI: 10.3389/fped.2020.635628] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/30/2020] [Accepted: 12/30/2020] [Indexed: 12/29/2022] Open
Abstract
Acute kidney injury (AKI) has been associated with deleterious impacts on a variety of body systems. While AKI is often accompanied by dysregulation of mineral metabolism-including alterations in calcium, phosphate, vitamin D, parathyroid hormone, fibroblast growth factor 23, and klotho-its direct effects on the skeletal system of children and adolescents remain largely unexplored. In this review, the pathophysiology of dysregulated mineral metabolism in AKI and its potential effects on skeletal health are discussed, including data associating AKI with fracture risk.
Collapse
Affiliation(s)
- Anisha Hegde
- Department of Pediatrics, University of North Carolina Hospitals, Chapel Hill, NC, United States
| | - Michelle R Denburg
- Division of Nephrology, The Children's Hospital of Philadelphia, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, United States
| | - Dorey A Glenn
- Division of Nephrology, University of North Carolina School of Medicine, Chapel Hill, NC, United States
| |
Collapse
|
38
|
Chen LS, Singh RJ. Utilities of traditional and novel biomarkers in the management of acute kidney injury. Crit Rev Clin Lab Sci 2019. [DOI: 10.1080/10408363.2019.1689916] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/25/2022]
Affiliation(s)
- Li-Sheng Chen
- Research and Development Directorate (J-9), Defense Health Agency, Silver Spring, MD, USA
| | | |
Collapse
|
39
|
Abstract
Fibroblast growth factor 23 (FGF23) is a hormone with a central role in the regulation of phosphate homeostasis. This regulation is accomplished by the coordinated modulation of renal phosphate handling, vitamin D metabolism and parathyroid hormone secretion. Patients with kidney disease have increased circulating levels of FGF23 and in other patient populations and in healthy individuals, FGF23 levels also rise following an increase in dietary phosphate intake. Maladaptive increases in FGF23 have a detrimental effect on several organs and tissues and, importantly, these pathological changes most likely contribute to increased morbidity and mortality. For example, in the context of heart disease, FGF23 is involved in the development of pathological hypertrophy that can lead to congestive heart failure. Increased FGF23 concentrations can also lead to microcirculatory changes, in particular reduced vasodilatory capacity, and collectively these cardiovascular changes can compromise tissue perfusion. In addition, FGF23 is associated with inflammation and an increased risk of infection; other potentially detrimental effects of FGF23 are likely to emerge in the future. Most importantly, recent insights demonstrate that FGF23 can be therapeutically targeted, which holds promise for the treatment of many patients in a variety of clinical settings.
Collapse
|
40
|
Abstract
Chronic kidney disease (CKD) is a global health epidemic that accelerates cardiovascular disease, increases risk of infection, and causes anemia and bone disease, among other complications that collectively increase risk of premature death. Alterations in calcium and phosphate homeostasis have long been considered nontraditional risk factors for many of the most morbid outcomes of CKD. The discovery of fibroblast growth factor 23 (FGF23), which revolutionized the diagnosis and treatment of rare hereditary disorders of FGF23 excess that cause hypophosphatemic rickets, has also driven major paradigm shifts in our understanding of the pathophysiology and downstream end-organ complications of disordered mineral metabolism in CKD. As research of FGF23 in CKD has rapidly advanced, major new questions about its regulation and effects continuously emerge. These are promoting exciting innovations in laboratory, patient-oriented, and epidemiological research and stimulating clinical trials of new therapies and repurposing of existing ones to target FGF23.
Collapse
Affiliation(s)
- John Musgrove
- Division of Nephrology, Department of Medicine, Duke University School of Medicine, Durham, North Carolina 27710, USA;
| | - Myles Wolf
- Division of Nephrology, Department of Medicine, Duke University School of Medicine, Durham, North Carolina 27710, USA; .,Duke Clinical Research Institute, Duke University School of Medicine, Durham, North Carolina 27710, USA
| |
Collapse
|
41
|
Association between mineral and bone disorder in patients with acute kidney injury following cardiac surgery and adverse outcomes. BMC Nephrol 2019; 20:369. [PMID: 31615432 PMCID: PMC6794865 DOI: 10.1186/s12882-019-1572-y] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2018] [Accepted: 09/30/2019] [Indexed: 12/29/2022] Open
Abstract
BACKGROUND Numerous studies have evaluated the prevalence and importance of mineral and bone disorders among patients with chronic kidney disease (CKD) and end-stage renal disease (ESRD). However, little is known about dysregulated mineral and bone metabolism in acute kidney injury (AKI). METHODS We evaluated the association between mineral and bone metabolites and clinical outcomes in 158 patients who underwent cardiac surgery and developed AKI between June 2014 and January 2016. The baseline characteristics of the patients were recorded, and the levels of mineral and bone metabolites, including calcium, phosphate, intact parathyroid hormone (iPTH), 25-hydroxyvitamin D (25D), bone-specific alkaline phosphatase (BAP), tartrate-resistant acid phosphatase 5b (TRACP-5b) and C-terminal fibroblast growth factor 23 (cFGF23) were measured within 12 h after establishing the clinical diagnosis. RESULTS The serum phosphate, iPTH and cFGF23 levels were significantly associated with the 28-day mortality (phosphate: Hazard Ratio [HR] =2.620, 95% CI: 1.083 to 6.338, p = 0.035; iPTH: HR = 1.044, 95% CI: 1.001 to 1.090, p = 0.046; cFGF23: HR = 1.367, 95% CI: 1.168 to 1.599, p < 0.001). Moreover, higher serum cFGF23 and BAP levels were independently associated with an increased risk of adverse outcomes. Additionally, we found that the serum cFGF23 levels rose most significantly and were associated with the severity of AKI (P < 0.001). CONCLUSIONS Mineral and bone metabolites are dysregulated and are associated with adverse clinical outcomes among patients with AKI. TRIAL REGISTRATION www.clinicaltrials.gov NCT00953992. Registered 6 August 2009.
Collapse
|
42
|
Jacob KA, Leaf DE. Prevention of Cardiac Surgery-Associated Acute Kidney Injury: A Review of Current Strategies. Anesthesiol Clin 2019; 37:729-749. [PMID: 31677688 PMCID: PMC7644277 DOI: 10.1016/j.anclin.2019.08.007] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
Acute kidney injury is a common and often severe postoperative complication after cardiac surgery, and is associated with poor short-term and long-term outcomes. Numerous randomized controlled trials have been conducted to investigate various strategies for prevention of cardiac surgery-associated acute kidney injury. Unfortunately, most trials that have been conducted to date have been negative. However, encouraging results have been demonstrated with preoperative administration of corticosteroids, leukocyte filtration, and administration of inhaled nitric oxide intraoperatively, and implementation of a Kidney Disease: Improving Global Outcomes bundle of care approach postoperatively. These findings require validation in large, multicenter trials.
Collapse
Affiliation(s)
- Kirolos A Jacob
- Department of Cardiothoracic Surgery, University Medical Center Utrecht, Mail Stop E03.511, PO Box 85500, Utrecht 3508 GA, the Netherlands.
| | - David E Leaf
- Division of Renal Medicine, Brigham and Women's Hospital, 75 Francis Street, Medial Research Building Room MR416B, Boston, MA 02115, USA
| |
Collapse
|
43
|
Hanudel MR, Zinter MS, Chen L, Gala K, Lim M, Guglielmo M, Deshmukh T, Vangala S, Matthay M, Sapru A. Plasma total fibroblast growth factor 23 levels are associated with acute kidney injury and mortality in children with acute respiratory distress syndrome. PLoS One 2019; 14:e0222065. [PMID: 31487315 PMCID: PMC6728039 DOI: 10.1371/journal.pone.0222065] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2019] [Accepted: 08/21/2019] [Indexed: 11/24/2022] Open
Abstract
Acute respiratory distress syndrome (ARDS) has high rates of mortality and multisystem morbidity. Pre-clinical data suggest that fibroblast growth factor 23 (FGF23) may contribute to pulmonary pathology, and FGF23 is associated with mortality and morbidity, including acute kidney injury (AKI), in non-ARDS cohorts. Here, we assess whether FGF23 is associated with AKI and/or mortality in a cohort of 161 pediatric ARDS patients. Plasma total (intact + C-terminal) FGF23 and intact FGF23 concentrations were measured within 24 hours of ARDS diagnosis (Day 1), and associations with Day 3 AKI and 60-day mortality were evaluated. 35 patients (22%) developed AKI by 3 days post-ARDS diagnosis, and 25 (16%) died by 60 days post-ARDS diagnosis. In unadjusted models, higher Day 1 total FGF23 was associated with Day 3 AKI (odds ratio (OR) 2.22 [95% confidence interval (CI) 1.62, 3.03], p<0.001), but Day 1 intact FGF23 was not. In a model adjusted for demographics and disease severity, total FGF23 remained associated with AKI (OR 1.52 [95% CI 1.02, 2.26], p = 0.039). In unadjusted models, both higher Day 1 total and intact FGF23 were associated with 60-day mortality (OR 1.43 [95% CI 1.07, 1.91], p = 0.014; and OR 1.44 [95% CI 1.02, 2.05], p = 0.039, respectively). In the adjusted model, only total FGF23 remained associated with 60-day mortality (OR 1.62 [95% CI 1.07, 2.45], p = 0.023). In a subgroup analysis of patients with Day 1 plasma IL-6 concentrations available, inflammation partially mediated the association between total FGF23 and AKI. Our data suggest both inflammation-dependent and inflammation-independent associations between total FGF23 and clinical outcomes in pediatric ARDS patients.
Collapse
Affiliation(s)
- Mark R. Hanudel
- Department of Pediatrics, David Geffen School of Medicine at UCLA, Los Angeles, CA, United States of America
- * E-mail:
| | - Matthew S. Zinter
- Department of Pediatrics, UCSF School of Medicine, San Francisco, CA, United States of America
| | - Lucia Chen
- Department of Medicine, David Geffen School of Medicine at UCLA, Los Angeles, CA, United States of America
| | - Kinisha Gala
- Department of Pediatrics, David Geffen School of Medicine at UCLA, Los Angeles, CA, United States of America
| | - Michelle Lim
- Department of Pediatrics, David Geffen School of Medicine at UCLA, Los Angeles, CA, United States of America
| | - Mona Guglielmo
- Department of Pediatrics, David Geffen School of Medicine at UCLA, Los Angeles, CA, United States of America
| | - Tanaya Deshmukh
- Department of Pediatrics, David Geffen School of Medicine at UCLA, Los Angeles, CA, United States of America
| | - Sitaram Vangala
- Department of Medicine, David Geffen School of Medicine at UCLA, Los Angeles, CA, United States of America
| | - Michael Matthay
- Department of Medicine, UCSF School of Medicine, San Francisco, CA, United States of America
| | - Anil Sapru
- Department of Pediatrics, David Geffen School of Medicine at UCLA, Los Angeles, CA, United States of America
| |
Collapse
|
44
|
Bär L, Stournaras C, Lang F, Föller M. Regulation of fibroblast growth factor 23 (FGF23) in health and disease. FEBS Lett 2019; 593:1879-1900. [PMID: 31199502 DOI: 10.1002/1873-3468.13494] [Citation(s) in RCA: 52] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2019] [Revised: 03/26/2019] [Accepted: 03/27/2019] [Indexed: 12/19/2022]
Abstract
Fibroblast growth factor 23 (FGF23) is mainly produced in the bone and, upon secretion, forms a complex with a FGF receptor and coreceptor αKlotho. FGF23 can exert several endocrine functions, such as inhibiting renal phosphate reabsorption and 1,25-dihydroxyvitamin D3 production. Moreover, it has paracrine activities on several cell types, including neutrophils and hepatocytes. Klotho and Fgf23 deficiencies result in pathologies otherwise encountered in age-associated diseases, mainly as a result of hyperphosphataemia-dependent calcification. FGF23 levels are also perturbed in the plasma of patients with several disorders, including kidney or cardiovascular diseases. Here, we review mechanisms controlling FGF23 production and discuss how FGF23 regulation is perturbed in disease.
Collapse
Affiliation(s)
- Ludmilla Bär
- Institute of Agricultural and Nutritional Sciences, Martin Luther University Halle-Wittenberg, Halle (Saale), Germany
| | - Christos Stournaras
- Institute of Biochemistry, University of Crete Medical School, Heraklion, Greece
| | - Florian Lang
- Institute of Physiology, University of Tübingen, Germany
| | - Michael Föller
- Institute of Physiology, University of Hohenheim, Stuttgart, Germany
| |
Collapse
|
45
|
Affiliation(s)
- David E Leaf
- Division of Renal Medicine, Brigham and Women's Hospital, Boston, MA
| |
Collapse
|
46
|
Abstract
Acute kidney injury (AKI) is associated with many of the same mineral metabolite abnormalities that are observed in chronic kidney disease. These include increased circulating levels of the osteocyte-derived, vitamin D-regulating hormone, fibroblast growth factor 23 (FGF23), and decreased renal expression of klotho, the co-receptor for FGF23. Recent data have indicated that increased FGF23 and decreased klotho levels in the blood and urine could serve as novel predictive biomarkers of incident AKI, or as novel prognostic biomarkers of adverse outcomes in patients with established AKI. In addition, because FGF23 and klotho exert numerous classic as well as off-target effects on a variety of organ systems, targeting their dysregulation in AKI may represent a unique opportunity for therapeutic intervention. We review the pathophysiology, kinetics, and regulation of FGF23 and klotho in animal and human studies of AKI, and we discuss the challenges and opportunities involved in targeting FGF23 and klotho therapeutically.
Collapse
Affiliation(s)
- Marta Christov
- Department of Medicine, New York Medical College, Valhalla, NY.
| | - Javier A Neyra
- Division of Nephrology, Bone and Mineral Metabolism, Department of Internal Medicine, University of Kentucky, Lexington, KY; Division of Nephrology, Department of Internal Medicine, University of Texas Southwestern, Dallas, TX
| | - Sanjeev Gupta
- Department of Medicine, New York Medical College, Valhalla, NY
| | - David E Leaf
- Division of Renal Medicine, Brigham and Women's Hospital, Boston, MA
| |
Collapse
|
47
|
Urine Klotho Is Lower in Critically Ill Patients With Versus Without Acute Kidney Injury and Associates With Major Adverse Kidney Events. Crit Care Explor 2019; 1. [PMID: 32123869 PMCID: PMC7051168 DOI: 10.1097/cce.0000000000000016] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022] Open
Abstract
Supplemental Digital Content is available in the text. Klotho and fibroblast growth factor-23 were recently postulated as candidate biomarkers and/or therapeutic targets in acute kidney injury. We examined whether urine Klotho and serum intact fibroblast growth factor-23 levels were differentially and independently associated with major adverse kidney events in critically ill patients with and without acute kidney injury.
Collapse
|
48
|
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: 19] [Impact Index Per Article: 3.2] [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
|
49
|
de Oliveira Neves FM, Araújo CB, de Freitas DF, Arruda BFT, de Macêdo Filho LJM, Salles VB, Meneses GC, Martins AMC, Libório AB. Fibroblast growth factor 23, endothelium biomarkers and acute kidney injury in critically-ill patients. J Transl Med 2019; 17:121. [PMID: 30971270 PMCID: PMC6458699 DOI: 10.1186/s12967-019-1875-6] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2019] [Accepted: 04/05/2019] [Indexed: 02/06/2023] Open
Abstract
BACKGROUND Fibroblast growth factor 23 (FGF23) and endothelium-related biomarkers have been related to AKI in critically-ill patients. Also, FGF23 is associated with endothelial dysfunction. In this study, we investigated if elevated FGF23 association with severe AKI is mediated by several endothelial/glycocalyx-related biomarkers. METHODS Prospective cohort study with critically-ill patients. Blood samples were collected within the first 24 h after ICU admission. Severe AKI (defined according to KDIGO stage 2/3) was the analyzed outcome. RESULTS 265 patients were enrolled and 82 (30.9%) developed severe AKI-defined according to KDIGO stage 2/3. Blood samples to biomarkers measurement were collected within the first 24 h after ICU admission. After adjustment for several variables, FGF23, vascular cell adhesion protein 1 (VCAM-1), angiopoietin 2 (AGPT2), syndecan-1 and intercellular adhesion molecule-1 (ICAM-1) were associated with severe AKI. The individual indirect effects of VCAM-1, AGPT2 and syndecan-1 explained 23%, 31%, and 32% of the total observed effect of FGF23 on severe AKI, respectively. ICAM-1 showed no statistically significant mediation. When all three endothelium-related biomarkers were included in a directed acyclic graph (DAG), the Bayesian network learning suggested the following causal association pathway FGF-23 → syndecan-1 → VCAM-1 → AGPT2 → severe AKI. CONCLUSIONS The association between FGF23 and AKI are mediated by endothelium-related biomarkers, mainly VCAM-1, AGPT2 and syndecan-1. Moreover, the statistical models show that syndecan-1, a biomarker of endothelial glycocalyx dysfunction, seems to be the initial mediator between FGF23 and severe AKI.
Collapse
Affiliation(s)
- Fernanda Macedo de Oliveira Neves
- Medical Sciences Postgraduate Program, Department of Clinical Medicine, Universidade Federal do Ceará, Avenida Abolição, 4043 Ap 1203, Fortaleza, Ceará, CEP 60165-082, Brazil
| | - Camila Barbosa Araújo
- Medical Sciences Postgraduate Program, Universidade de Fortaleza-UNIFOR, Fortaleza, Ceara, Brazil
| | | | | | | | | | - Gdayllon Cavalcante Meneses
- Medical Sciences Postgraduate Program, Department of Clinical Medicine, Universidade Federal do Ceará, Avenida Abolição, 4043 Ap 1203, Fortaleza, Ceará, CEP 60165-082, Brazil
| | - Alice Maria Costa Martins
- Department of Clinical and Toxicological Analysis, Faculty of Pharmacy, Federal University of Ceara, Fortaleza, Ceara, Brazil
| | - Alexandre Braga Libório
- Medical Sciences Postgraduate Program, Department of Clinical Medicine, Universidade Federal do Ceará, Avenida Abolição, 4043 Ap 1203, Fortaleza, Ceará, CEP 60165-082, Brazil. .,Medical Sciences Postgraduate Program, Universidade de Fortaleza-UNIFOR, Fortaleza, Ceara, Brazil.
| |
Collapse
|
50
|
|