Skip to main content
Erschienen in: BMC Nephrology 1/2023

Open Access 01.12.2023 | Research

A potential link between fibroblast growth factor-23 and the progression of AKI to CKD

Erschienen in: BMC Nephrology | Ausgabe 1/2023

Abstract

Background

Patients who recover from acute kidney injury (AKI) have a 25% increase in the risk of chronic kidney disease (CKD) and a 50% increase in mortality after a follow-up of approximately 10 years. Circulating FGF-23 increases significantly early in the development of AKI, is significantly elevated in patients with CKD and has become a major biomarker of poor clinical prognosis in CKD. However, the potential link between fibroblast growth factor-23 levels and the progression of AKI to CKD remains unclear.

Method

Serum FGF-23 levels in AKI patients and ischaemia‒reperfusion injury (IRI) mice were detected with ELISA. Cultured HK2 cells were incubated with FGF-23 and PD173074, a blocker of FGFR, and then TGFβ/Smad and Wnt/β-catenin were examined with immunofluorescence and immunoblotting. Quantitative real-time polymerase chain reaction was used to detect the expression of COL1A1 and COL4A1. Histologic staining confirmed renal fibrosis.

Results

The level of serum FGF-23 was significantly different between AKI patients and healthy controls (P < 0.01). Moreover, serum FGF-23 levels in the CKD progression group were significantly higher than those in the non-CKD progression group of AKI patients (P < 0.01). In the AKI-CKD mouse model, serum FGF-23 levels were increased, and renal fibrosis occurred; moreover, the protein expression of β-catenin and p-Smad3 was upregulated. PD173074 downregulated the expression of β-catenin and p-Smad3 and reduced fibrosis in both mice and HK2 cells.

Conclusion

The increase in FGF-23 may be associated with the progression of AKI to CKD and may mediate renal fibrosis via TGF-β and Wnt/β-catenin activation.
Hinweise

Supplementary Information

The online version contains supplementary material available at https://​doi.​org/​10.​1186/​s12882-023-03125-1.
Yinghui Lu and Shutian Xu contributed equally to this work.

Publisher’s Note

Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Background

Acute kidney injury (AKI) is a common clinical problem in hospitalized patients worldwide, and it is one of the most common acute and critical illnesses in clinical departments. AKI not only affects the patient survival rate, long-term prognosis and quality of life but also places a huge economic burden on the patient’s family and society. AKI confers formidable morbidity and mortality in its acute phase, and among survivors of AKI, the long-term outcomes are far from benign. Patients who recover from AKI have a 25% increase in the risk of chronic kidney disease (CKD) and a 50% increase in mortality after a follow-up of approximately 10 years [13]. Although clinical observations describe a clear association, the underlying mechanism of AKI progression to CKD needs to be explored.
Bone-derived fibroblast growth factor-23 (FGF-23) is an important endocrine regulator of mineral homeostasis, and its effects are transduced by cognate FGF receptor (FGFR)1-α-Klotho complexes [3, 4]. Circulating FGF-23 levels increase precipitously in patients with kidney diseases and indicate worse renal and cardiovascular outcomes [5, 6]. FGF-23 levels increase early in CKD [7], predominantly as intact protein, and generally precede changes in other mineral metabolites [8]. Similarly, circulating FGF-23 concentrations increase rapidly in AKI, also preceding changes in other mineral markers and conventional measures of renal function [9].
Renal fibrosis is the pathological basis of CKD [10]. The canonical TGF-β1/smad3 signalling pathway mediates the transdifferentiation of renal tubular epithelial cells and plays an important role in the progression of renal fibrosis [11]. Animal studies of ureteral obstruction (UUO) have shown that tubule-derived FGF-23 can enhance the activity of myofibroblasts during AKI, possibly promoting the signalling cascade of renal fibrosis by activating TGF-β channels. [12, 13] The Wnt/β-catenin signalling pathway is closely related to the occurrence and development of renal interstitial fibrosis. Persistent activation of the Wnt/β-catenin pathway plays an important role in promoting the development of AKI to CKD [14].
In this study, we examined whether the increase in circulating FGF-23 was associated with CKD development after AKI. Then, we explored the potential molecular mechanism of FGF-23 in the progression of AKI to CKD. Our study aimed to uncover a novel mechanism of the progression of AKI to CKD and provides a potential therapeutic target for preventing and improving the prognosis of AKI.

Materials and methods

AKI patients

We selected 94 AKI patients who were admitted to the Nephrology Intensive Care Unit (ICU) of the National Kidney Disease Clinical Research Center and included 60 healthy humans in the same period as the control group. AKI diagnosis and staging criteria were based on the AKI guidelines of the Kidney Disease Improvement Global Prognosis Organization (KDIGO) [15]. The diagnostic criteria for CKD were a basal eGFR of less than 60 ml/(min 1.73 m2) and a duration of more than 3 months.

Animals

C57BL/6 mice (male, 12 weeks old) were purchased from Gem Pharmatech at Nanjing. The mice were housed individually at 23 °C with a 12:12-h light-dark cycle and were maintained on water and food.

Surgery protocols

Briefly, the mice were anaesthetized with pentobarbital (50 mg/ml, i.p.), and the body temperature was maintained at 36.8–37.5 °C during surgery with a temperature-controlled operating table. The renal pedicle was carefully dissected and clamped with a silver clip for 35 min. After releasing the clip, the wound was sutured. Sham-operated animals without clamping served as controls.

Treatment with PD173074

The pan-FGF receptor blocker PD173074 (Sigma‒Aldrich, Inc.) was dissolved in PBS. The mice (sham and IRI, twelve-week-old male C57BL/6 mice) were intraperitoneally injected with PBS or PD173074 (1 mg/kg) once daily for the same duration. After 2 weeks, the animals were sacrificed, and blood and kidney tissues were collected.

Serum biochemistry

Blood samples (~ 200 µl/each) were centrifuged at 3,000 rpm for 10 min at 4 °C to separate the serum (~ 100 µl/each). Blood urea nitrogen and creatinine were measured by Servicebio (https://​www.​servicebio.​cn/​).

FGF23 concentration assay

The active intact FGF23 (iFGF23) enzyme-linked immunosorbent assay (ELISA) kit (catalog no. CY-4000; Kainos Laboratories, Tokyo, Japan) was used. It is a two-point ELISA kit for the determination of iFGF23 in serum.

Morphological analysis of mouse tissues

After blood collection, the animals were sacrificed. The kidneys were isolated and prepared for molecular and histological analyses. Kidneys tissues were stained with Masson dye to observe fibrosis.

HK2 cell culture and treatments

The HK2 cell line was purchased from ATCC, and the cells were cultured in F12 (Gibco, Inc.) supplemented with 10% foetal bovine serum (Gibco, Inc.) in an atmosphere of 5% (v/v) CO2 in air at 37 °C. The cells were treated with 25 ng/ml FGF-23 (Research & Diagnostics Systems, Inc.).

Immunofluorescence

After being treated, cultured HK2 cells were fixed in 4% paraformaldehyde and permeabilized with 0.1% Triton X-100 in PBS, followed by blocking with 5% goat serum in PBS. Rabbit monoclonal antibodies against p-smad3 (catalogue C25A9; Cell Signalling Technologies, Inc.) were used at 1:1000. A rabbit monoclonal antibody against β-catenin (catalogue 610,154; BD Biosciences, Inc.) was used at 1:2000. Cy3-conjugated goat anti-mouse (catalogue A0521; Beyotime Biotechnology, Inc.) was used as a secondary antibody at 1:500. To visualize nuclei, the fixed cells were incubated with DAPI (400 ng/mL in PBS) for 10 min. Immunofluorescence images were taken with a DM5000B microscope (Leica). The myocyte cross-sectional area was measured by ImageJ software (http://​rsbweb.​nih.​gov/​ij/​).

RNA extraction and quantification.

Cultured HK2 cells and mouse kidney tissues were subjected to total RNA extraction with an isolation kit (Thermo Fisher Scientific, Inc.). In brief, this extraction method is based on the ability of glass fibers to bind nucleic acids in concentrated chaotropic salt solutions. Samples are disrupted in a typical high concentration guanidinium salt solution that simultaneously lyses cells and inactivates endogenous RNases. The lysate is diluted with an ethanol solution to make the RNA competent for binding to the glass fiber filter in the RNAqueous Filter Cartridge. This solution is passed through the filter pad where RNA binds and most other cellular contents flow through. The Filter Cartridge is washed 3 times to remove contaminants, and the RNA is eluted in a very low ionic strength solution. Reverse transcription of 300 ng of RNA to cDNA using the Reverse Transcription Kit (Takara Biomedical Technology (Beijing) Co., Ltd.). 2 reverse transcription primers, Random 6 mers and Oligo dT Primer, are included in the kit to synthesize cDNA suitable for Real Time PCR. mRNA sample was quantified by qPCR using a kit from TaKaRa (TaKaRa Bio, Inc.). The primers used are summarized in Table 1. For Q-PCR experiments, annealing temperature is set at 55–60 °C. Relative mRNA expression was evaluated with the 2–ΔΔCT method using 18 S for normalization.
Table 1
The primers used in this study
 
Forward sequence (5’→3’)
Reverse sequence (5’→3’)
COL1A1
GCTCCTCTTAGGGGCCACT
ATTGGGGACCCTTAGGCCAT
COL4A1
TCCGGGAGAGATTGGTTTCC
CTGGCCTATAAGCCCTGGT
18s
TTTCTCGATTCCGTGGGTGG
AGCATGCCAGAGTCTCGTTC

Protein extraction and Western blot analysis

Proteins were extracted from kidney tissues with RIPA buffer (Beyotime Biotechnology, Inc.) according to the manufacturer’s instructions. Antibodies against GAPDH (catalogue BS65529; Bioworld Technology, Inc.), α-SMA (catalogue ab5694; Abcam, Inc.), β-catenin (catalogue 610,154; BD Biosciences, Inc.) and p-smad3 (catalogue C25A9; Cell Signalling Technologies, Inc.), E-cad (catalogue 20874-1-AP; Proteintech, Inc.), and NGAL (catalogue ab216462; Abcam, Inc.) were used as primary antibodies, and horseradish peroxidase-conjugated goat anti-rabbit or anti-mouse (Beyotime Biotechnology, Inc.) secondary antibodies were used. Image analysis with image J software.

Statistical analysis

The data were tested for normal distribution. If the measures were normally distributed, they were expressed as mean ± SD, and the t-test was used for comparison between two groups, and Pearson correlation was used for correlation analysis. The Kruskal-Wallis test was used for comparison between two or more groups, and the Spearman’s rank correlation test was used for correlation analysis. The chi-square test was used for comparison of the sex and diabetes ratio. The correlation between AKI patients and FGF-23 was analysed by logistic regression analysis. P < 0.05 was considered statistically significant.

Results

FGF-23 is expressed at increased levels in AKI patients progressing to CKD

There were 94 AKI patients were enrolled. The baseline characteristics of the patients are shown in Tables 2 and 3. The levels of serum FGF-23 were significantly different between AKI patients and healthy controls (152.65 (92.18, 293.90) versus 37.62 (26.40, 64.01) pg/ml pg/ml, P < 0.01, Fig. 1a). These AKI patients were followed up for three months after discharge to observe their progression to CKD. Eight of these patients lost follow-up at the three months. These AKI patients were divided into a CKD progression group (45 cases) and a non-CKD progression group (41 cases). Serum FGF-23 levels in the CKD progression group were significantly higher than those in the non-CKD progression group (189.05 (151.59, 282.87) pg/ml versus 230.60 (66.58, 152.68) pg/ml, P < 0.05, Fig. 1b). Collectively, these results indicated that AKI patients had a high level of circulating FGF-23, and higher FGF-23 levels were found in those patients that progressed from AKI to CKD.
Table 2
Characteristics of the AKI patients and healthy controls
 
AKI
(n = 94)
Controls
(n = 60)
P
Sex (male/female)
52/42
32/28
> 0.05
Age
47.00 ± 16.90
46.65 ± 13.15
> 0.05
eGFR
8.44(5.23–12.49)
105.76(94.58-113.85)
< 0.001
Note: P < 0.05 was considered statistically significant. eGFR: estimated glomerular filtration rate
Table 3
Characteristics of the AKI patients at baseline
 
AKI
(n = 94)
male n (%)
52 (55.3)
Age
47.00 ± 16.90
Body Mass Index
23.20 (20.00, 25.20)
Systolic blood pressure
140.61 ± 21.87
 
Diastolic blood pressure
83.27 ± 14.10
 
Diabetes n (%)
10(10.6)
 
C-reactive protein (mg/L)
21.60 (7.80, 94.00)
 
IL-6 (ng/L)
22.01 (11.90, 42.87)
 
Serum creatinine (mg/dl)
6.17 (4.69, 9.88)
 
Blood urea nitrogen (mg/dl)
68.38 ± 31.13
 
P < 0.05 was considered statistically significant. eGFR: estimated glomerular filtration rate.
Patients were divided into two groups based on FGF-23 levels (< 53.8 pg/ml and ≥ 53.8 pg/ml), and we found that the higher group had poorer renal recovery at discharge than the lower group (P < 0.05, Table 4). We performed logistic regression analysis and found that serum levels of FGF-23 were associated with AKI (OR = 1.044, P < 0.001) (Supplementary Table 1), also related with AKI progression to CKD (OR = 1.018, P = 0.018) (Supplementary Table 2).
Table 4
The correlation between FGF-23 level and AKI post CKD
FGF-23(pg/ml)
non-CKD-p(n = 41)
CKD-p(n = 45)
P
< 53.8(n = 15)
11(73.4%)
4(26.7%)
0.029
≥ 53.8(n = 71)
30(42.2%)
41(57.3%)
 
Note: non-CKD-p: non-CKD progression. CKD-p: CKD progression. AKI post CKD: AKI progresses to CKD. P < 0.05 was considered statistically significant

FGF-23 exhibits a sustained increase in the AKI-CKD mouse model

We generated a bilateral ischaemia‒reperfusion injury (Bi-IRI) mouse model. The bilateral kidneys of the mice were reperfused after 35 min of ischaemia. Kidney histologic staining revealed increasing pathologic changes including acute inflammatory infiltration, brush edge detachment, tubular atrophy and interstitial fibrosis 14 days after IRI (Fig. 2a). The epithelial-mesenchymal transition marker α-smooth muscle actin (α-SMA) was markedly induced at 14 days after IRI, as evidenced by immunofluorescent staining (Fig. 2b). At 14 days post-AKI, IRI caused obvious kidney shrinkage (Fig. 2c). The serum levels of FGF-23 were increased after 3 days of IRI, followed by sustained elevation until 14 days of IRI (Fig. 2d). Moreover, we found that COL1A1 and COL4A1 levels in kidney tissues were upregulated in IRI mice (Fig. 2e). These results suggested that two weeks after surgery, these mice showed markedly high expression of FGF-23 and obvious fibrosis. Consistent with the previous literature [16], renal fibrosis at 14 days post-AKI may be the precursor of CKD. In the next experiment, we used a mouse model 14 days after Bi-IRI.

FGF-23 promotes profibrotic cellular signalling in HK2 cells

To explore the molecular mechanism by which the increase in FGF-23 promotes renal fibrosis, we used renal tubular HK2 cells to observe the changes in response to FGF-23 and FGFR inhibitors. FGF-23 markedly reduced the epithelial marker E-cadherin (E-cad) and increased the myofibroblast marker α-SMA (Fig. 3a, b). Then, we examined the TGFβ/Smad and Wnt/β-catenin signalling pathways, which are the major profibrogenic pathways that are causatively related to renal fibrogenesis (Fig. 3a, b). FGF-23 activated Smad3 phosphorylation and β-catenin and led to their nuclear translocation in HK2 cells (Fig. 3c). In contrast, the pan-FGFR inhibitor PD173074 reduced Smad3 phosphorylation and β-catenin expression, as well as their nuclear translocation (Fig. 3c). Thus, we speculate that FGF-23 may promote renal fibrosis via TGFβ/Smad and Wnt/β-catenin signalling.

FGFR inhibition promotes kidney recovery and reduces renal fibrosis in AKI-CKD mice

To further determine how FGF-23 promotes renal fibrosis, we treated IRI mice daily with the pan-FGFR inhibitor PD173074 for 2 weeks beginning on the first day after surgery. The serum levels of FGF-23 were increased after IRI, and there was no significant difference between IRI and mice treated with PD173074 (Fig. 4d). The kidney histologic examinations showed that PD173074 mitigated renal fibrosis induced by IRI (Fig. 4a). PD173074 mitigated the abnormal expression of neutrophil gelatinase-associated lipocalin (NGAL), β-catenin, E-cadherin, α-SMA, and phosphorylated Smad3, indicating that PD173074 can protect the kidney from previous damage and functional abnormalities induced by IRI (Fig. 4b, c). PD173074 treatment attenuates creatinine and urea nitrogen in IRI mice (Supplementary Fig. 1).

Discussion

AKI is an increasing health burden with high morbidity and mortality rates worldwide. AKI is a risk factor for CKD development. The understanding of AKI developing into CKD was derived from retrospective clinical observations. AKI is now considered to be an independent risk factor for the development of CKD [17]. However, there are limited experimental data directly addressing the AKI-to-CKD transition. Thus, we explored the potential profibrotic role of FGF-23 after AKI and described its molecular mechanism.
Bone is the major source of circulating FGF-23. FGF-23 acts on the kidneys through fibroblast growth factor receptors (FGFRs) and the coreceptor Klotho to promote phosphaturia by downregulating phosphate transporters, as well as controlling vitamin D metabolizing enzymes to reduce blood 1,25-dihydroxyvitamin D [18]. Intact FGF-23 (iFGF-23) can be cleaved into N-terminal FGF-23 and C-terminal FGF-23 (cFGF-23) by Furin or plasminogen activators [19]. According to the literature, circulating FGF-23 is nearly all iFGF-23 in haemodialysis patients due to impaired FGF-23 cleavage [20], and approximately 80% of circulating FGF-23 is iFGF-23 in predialysis patients [21]. In this study, we selected iFGF-23 concentration detection in human and mouse serum. Several studies have shown that when present in excess, FGF-23 can produce off-target effects beyond classical endocrine mineral processing. This effect is thought to occur in the hearts of CKD patients, possibly driving hypertrophic and fibrotic signalling programs through the inappropriate activation of cells that do not express their physiological coreceptor Klotho [22]. In our previous study, we demonstrated that FGF-23 was elevated in the serum of CKD rats, which led to myocardial hypertrophy and promoted cardiomyocyte fibrosis [23]. Most notably, it has been shown that FGF-23 enhances the profibrotic signalling cascade in injury-induced renal fibroblasts by activating FGFR4 and upregulating the calcium transporter transient receptor potential cation channel 6 [2426].
In this study, we found that circulating FGF-23 levels were significantly increased in AKI patients, and FGF-23 levels in AKI patients were closely related to the recovery of renal function; in other words, the higher the FGF-23 level was, the higher the possibility of progression to CKD (Fig. 1). Moreover, after ischaemia‒reperfusion, continuous increases in serum FGF-23 were observed in IRI mice (Fig. 2). Based on these results, we hypothesized that FGF-23 could mediate CKD progression.
Several pathways are thought to be involved in FGF-23-mediated promotion of CKD. For instance, hyperphosphatemia is closely related to the degree of tubulointerstitial damage [26]. FGF23 is associated with the inflammatory response [27], endothelial injury [22], and sympathetic and RAAS activation [28]. Most notably, renal fibrosis is the pathological basis of CKD. In recent years, studies have revealed that the canonical TGF-β1/smad3 signalling pathway plays an important role in the progression of renal fibrosis, and persistent activation of the Wnt/β-catenin pathway is involved in promoting the development of AKI to CKD. Coincidently, our previous study showed that TGF-β and Wnt/β-catenin pathway inhibition could reverse renal tubular fibrosis [29]. According to these results, we hypothesized that FGF-23 accelerated AKI-CKD via the TGF-β and Wnt/β-catenin pathways. Through in vivo and in vitro experiments, we suggest that FGF-23 could not only lead to renal tubular fibrosis directly but could also activate the fibrotic TGFβ/Smad and Wnt/β-catenin pathways (Figs. 2 and 3). In addition, PD173074, a blocker of FGFR, inhibited this effect both in vivo and in vitro. (Figures 2 and 4). On the other hand, myofibroblasts are the main source of extracellular matrix (ECM) during kidney fibrosis [30], and genetic fate-tracing data in mice and histological analyses of human tissue suggested that epithelial, endothelial, haematopoietic and resident mesenchymal cells all contribute to fibrosis [31]. Does FGF-23 lead to myofibroblast activation and eventually cause renal fibrosis by damaging renal tubular epithelial cells? This needs to be confirmed by further studies.
In summary, we extended our understanding of FGF-23 in AKI-CKD. We showed a sustained increase in circulating FGF-23 in AKI patients and IRI mice, which may be related to AKI progression to CKD. We further confirmed that the upregulation of FGF-23 facilitates the activation of TGF-β and Wnt/β-catenin signalling. Moreover, FGF-23 upregulation may result in renal fibrosis via TGF-β and Wnt/β-catenin activation. We believe these data not only uncover a novel mechanism of AKI-CKD but also hint at a potential therapeutic target for AKI treatment.

Acknowledgements

Blood samples of patients and healthy controls were obtained from Renal Biobank of National Clinical Research Center of Kidney Diseases, Jiangsu Provincial Science and Technology Resources Coordination Service Platform.

Declarations

Competing interests

The authors declare that they have no competing interests.
All procedures performed in studies involving human participants were in accordance with the ethical standards of the institutional research committee of Jinling Hospital (healthy subjects IRB NO. 2014ZFYJ-021-01 and AKI patients IRB NO. 2017NZKY-013-01) and with the 1964 Helsinki declaration and its later amendments or comparable ethical standards.
Informed consentwas obtained from all individual participants included in the study. The animal in this study were treated in accordance with the standard ethical guidelines. All methods are reported in accordance with ARRIVE guidelines. All animal experiments in the present study were approved by the Animal Care and Use Committee of Jinling Hospital (NO. 2022DZGKJDWLS-00121).
Not applicable.
Open AccessThis article is licensed under a Creative Commons Attribution 4.0 International License, which permits use, sharing, adaptation, distribution and reproduction in any medium or format, as long as you give appropriate credit to the original author(s) and the source, provide a link to the Creative Commons licence, and indicate if changes were made. The images or other third party material in this article are included in the article's Creative Commons licence, unless indicated otherwise in a credit line to the material. If material is not included in the article's Creative Commons licence and your intended use is not permitted by statutory regulation or exceeds the permitted use, you will need to obtain permission directly from the copyright holder. To view a copy of this licence, visit http://​creativecommons.​org/​licenses/​by/​4.​0/​. The Creative Commons Public Domain Dedication waiver (http://​creativecommons.​org/​publicdomain/​zero/​1.​0/​) applies to the data made available in this article, unless otherwise stated in a credit line to the data.

Publisher’s Note

Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional affiliations.
Anhänge

Electronic supplementary material

Below is the link to the electronic supplementary material.
Literatur
1.
Zurück zum Zitat Zhang L, Wang F, Wang L, et al. Prevalence of chronic kidney disease in China: a cross-sectional survey. Lancet. 2012;379(9818):815–22.CrossRefPubMed Zhang L, Wang F, Wang L, et al. Prevalence of chronic kidney disease in China: a cross-sectional survey. Lancet. 2012;379(9818):815–22.CrossRefPubMed
2.
Zurück zum Zitat Ali T, Khan I, Simpson W, Prescott G, et al. Incidence and outcomes in acute kidney injury: a comprehensive population-based study. J Am Soc Nephrol. 2007;18:1292–8.CrossRefPubMed Ali T, Khan I, Simpson W, Prescott G, et al. Incidence and outcomes in acute kidney injury: a comprehensive population-based study. J Am Soc Nephrol. 2007;18:1292–8.CrossRefPubMed
3.
Zurück zum Zitat Grams ME, Estrella MM, Coresh J, et al. National Heart, Lung, and Blood Institute Acute Respiratory Distress Syndrome Network: fluid balance, diuretic use, and mortality in acute kidney injury. Clin J Am Soc Nephrol. 2011;6:966–73.CrossRefPubMedPubMedCentral Grams ME, Estrella MM, Coresh J, et al. National Heart, Lung, and Blood Institute Acute Respiratory Distress Syndrome Network: fluid balance, diuretic use, and mortality in acute kidney injury. Clin J Am Soc Nephrol. 2011;6:966–73.CrossRefPubMedPubMedCentral
4.
Zurück zum Zitat Yu XJ, Ibrahimi OA, Goetz R, et al. Analysis of the biochemical mechanisms for the endocrine actions of fibroblast growth factor-23 [J]. Endocrinology. 2005;146(11):4647–56.CrossRefPubMed Yu XJ, Ibrahimi OA, Goetz R, et al. Analysis of the biochemical mechanisms for the endocrine actions of fibroblast growth factor-23 [J]. Endocrinology. 2005;146(11):4647–56.CrossRefPubMed
5.
Zurück zum Zitat Goetz R, Ohnishi M, Kir S, et al. Conversion of a paracrine fibroblast growth factor into an endocrine fibroblast growth factor [J]. J Biol Chem. 2012;287(34):29134–46.CrossRefPubMedPubMedCentral Goetz R, Ohnishi M, Kir S, et al. Conversion of a paracrine fibroblast growth factor into an endocrine fibroblast growth factor [J]. J Biol Chem. 2012;287(34):29134–46.CrossRefPubMedPubMedCentral
7.
Zurück zum Zitat Isakova T, Wahl P, Vargas GS, et al. Fibroblast growth factor 23 is elevated before parathyroid hormone and phosphate in chronic kidney disease. Kidney Int. 2011;79:1370–8.CrossRefPubMedPubMedCentral Isakova T, Wahl P, Vargas GS, et al. Fibroblast growth factor 23 is elevated before parathyroid hormone and phosphate in chronic kidney disease. Kidney Int. 2011;79:1370–8.CrossRefPubMedPubMedCentral
8.
Zurück zum Zitat Smith ER, Cai MM, McMahon LP, et al. Biological variability of plasma intact and C-terminal FGF-23 measurements. J Clin Endocrinol Metab. 2012;97:3357–65.CrossRefPubMed Smith ER, Cai MM, McMahon LP, et al. Biological variability of plasma intact and C-terminal FGF-23 measurements. J Clin Endocrinol Metab. 2012;97:3357–65.CrossRefPubMed
10.
Zurück zum Zitat Grande MT, Pérez-Barriocanal F, López-Novoa JM. Role of inflammation in túbulo-interstitial damage associated to obstructive nephropathy. J Inflamm (Lond). 2010;7:19.CrossRefPubMed Grande MT, Pérez-Barriocanal F, López-Novoa JM. Role of inflammation in túbulo-interstitial damage associated to obstructive nephropathy. J Inflamm (Lond). 2010;7:19.CrossRefPubMed
11.
Zurück zum Zitat Iwano M, Neilson EG. Mechanisms of tubulointerstitial fibrosis. Curr Opin Nephrol Hypertens. 2004;13(3):279–84.CrossRefPubMed Iwano M, Neilson EG. Mechanisms of tubulointerstitial fibrosis. Curr Opin Nephrol Hypertens. 2004;13(3):279–84.CrossRefPubMed
12.
Zurück zum Zitat Xiao L, Zhou D, Tan RJ, et al. Sustained activation of Wnt/β-catenin signaling drives AKI to CKD progression. J Am Soc Nephrol. 2016;27(6):1727–40.CrossRefPubMed Xiao L, Zhou D, Tan RJ, et al. Sustained activation of Wnt/β-catenin signaling drives AKI to CKD progression. J Am Soc Nephrol. 2016;27(6):1727–40.CrossRefPubMed
13.
Zurück zum Zitat Smith ER, Tan SJ, Holt SG, et al. FGF-23 is synthesised locally by renal tubules and activates injury-primed fibroblasts. Sci Rep. 2017;7(1):3345.CrossRefPubMedPubMedCentral Smith ER, Tan SJ, Holt SG, et al. FGF-23 is synthesised locally by renal tubules and activates injury-primed fibroblasts. Sci Rep. 2017;7(1):3345.CrossRefPubMedPubMedCentral
14.
Zurück zum Zitat Zhou D, Li Y, Lin L, et al. Tubule-specific ablation of endogenous β-catenin aggravates acute kidney injury in mice. Kidney Int. 2012;82(5):537–47.CrossRefPubMedPubMedCentral Zhou D, Li Y, Lin L, et al. Tubule-specific ablation of endogenous β-catenin aggravates acute kidney injury in mice. Kidney Int. 2012;82(5):537–47.CrossRefPubMedPubMedCentral
15.
Zurück zum Zitat KDIGO AKI Work Group. KDIGO clinical practice guideline for acute kidney injury. Kidney Int Suppl. 2012;2:1–138. KDIGO AKI Work Group. KDIGO clinical practice guideline for acute kidney injury. Kidney Int Suppl. 2012;2:1–138.
16.
Zurück zum Zitat Shi M, Flores B, Gillings N, et al. αKlotho mitigates progression of AKI to CKD through activation of Autophagy. J Am Soc Nephrol. 2016;27(8):2331–45.CrossRefPubMed Shi M, Flores B, Gillings N, et al. αKlotho mitigates progression of AKI to CKD through activation of Autophagy. J Am Soc Nephrol. 2016;27(8):2331–45.CrossRefPubMed
17.
Zurück zum Zitat Wu J, Zheng C, Wang X et al. MicroRNA-30 family members regulate calcium/calcineurin signaling in podocytes.J Clin Invest. 2015;125(11). Wu J, Zheng C, Wang X et al. MicroRNA-30 family members regulate calcium/calcineurin signaling in podocytes.J Clin Invest. 2015;125(11).
18.
Zurück zum Zitat Agoro R, Ni P, Noonan ML, White KE. Osteocytic FGF23 and its kidney function. Front Endocrinol (Lausanne). 2020;11:592.CrossRefPubMed Agoro R, Ni P, Noonan ML, White KE. Osteocytic FGF23 and its kidney function. Front Endocrinol (Lausanne). 2020;11:592.CrossRefPubMed
19.
Zurück zum Zitat Wolf M, et al. Coupling fibroblast growth factor 23 production and cleavage: iron deficiency, rickets, and kidney disease. Curr Opin Nephrol Hypertens. 2014;23(4):411–9.CrossRefPubMedPubMedCentral Wolf M, et al. Coupling fibroblast growth factor 23 production and cleavage: iron deficiency, rickets, and kidney disease. Curr Opin Nephrol Hypertens. 2014;23(4):411–9.CrossRefPubMedPubMedCentral
20.
Zurück zum Zitat Shimada T, et al. Circulating fibroblast growth factor 23 in patients with end-stage renal disease treated by peritoneal dialysis is intact and biologically active. J Clin Endocrinol Metab. 2010;95(2):578–85.CrossRefPubMed Shimada T, et al. Circulating fibroblast growth factor 23 in patients with end-stage renal disease treated by peritoneal dialysis is intact and biologically active. J Clin Endocrinol Metab. 2010;95(2):578–85.CrossRefPubMed
21.
Zurück zum Zitat Smith ER, et al. Biological variability of plasma intact and C-terminal FGF23 measurements. J Clin Endocrinol Metab. 2012;97(9):3357–65.CrossRefPubMed Smith ER, et al. Biological variability of plasma intact and C-terminal FGF23 measurements. J Clin Endocrinol Metab. 2012;97(9):3357–65.CrossRefPubMed
22.
Zurück zum Zitat David V, Martin A, Isakova T, et al. Inflammation and functional iron deficiency regulate fibroblast growth factor 23 production. Kidney Int. 2016;89:135–46.CrossRefPubMedPubMedCentral David V, Martin A, Isakova T, et al. Inflammation and functional iron deficiency regulate fibroblast growth factor 23 production. Kidney Int. 2016;89:135–46.CrossRefPubMedPubMedCentral
23.
24.
Zurück zum Zitat Smith ER, Holt SG, Hewitson TD. FGF-23 activates injury-primed renal fibroblasts via FGFR4-dependent signalling and enhancement of TGF-β autoinduction. Int J Biochem Cell Biol. 2017;92:63–78.CrossRefPubMed Smith ER, Holt SG, Hewitson TD. FGF-23 activates injury-primed renal fibroblasts via FGFR4-dependent signalling and enhancement of TGF-β autoinduction. Int J Biochem Cell Biol. 2017;92:63–78.CrossRefPubMed
25.
Zurück zum Zitat Smith ER, McMahon LP, Holt SG. Fibroblast growth factor 23. Ann Clin Biochem. 2014;51:203–27.CrossRefPubMed Smith ER, McMahon LP, Holt SG. Fibroblast growth factor 23. Ann Clin Biochem. 2014;51:203–27.CrossRefPubMed
26.
Zurück zum Zitat Vervloet M. Renal and extrarenal effects of fibroblast growth factor 23. Nat Rev Nephrol. 2019; Feb; 15(2):109–120. Vervloet M. Renal and extrarenal effects of fibroblast growth factor 23. Nat Rev Nephrol. 2019; Feb; 15(2):109–120.
27.
Zurück zum Zitat Zanchi C, Locatelli M, Benigni A, et al. Renal expression of FGF23 in progressive renal disease of diabetes and the effect of ACE inhibitor. PLoS ONE. 2013;8:e70775.CrossRefPubMedPubMedCentral Zanchi C, Locatelli M, Benigni A, et al. Renal expression of FGF23 in progressive renal disease of diabetes and the effect of ACE inhibitor. PLoS ONE. 2013;8:e70775.CrossRefPubMedPubMedCentral
28.
Zurück zum Zitat de Oliveira Neves FM, Araújo CB, de Freitas DF, et al. Fibroblast growth factor 23, endothelium biomarkers and acute kidney injury in critically-ill patients. J Transl Med. 2019;17(1):121.CrossRefPubMedPubMedCentral de Oliveira Neves FM, Araújo CB, de Freitas DF, et al. Fibroblast growth factor 23, endothelium biomarkers and acute kidney injury in critically-ill patients. J Transl Med. 2019;17(1):121.CrossRefPubMedPubMedCentral
29.
Zurück zum Zitat Zhang Q, Yin S, Liu L, et al. Rhein reversal of DNA hypermethylation-associated Klotho suppression ameliorates renal fibrosis in mice. Sci Rep. 2016;6:34597.CrossRefPubMedPubMedCentral Zhang Q, Yin S, Liu L, et al. Rhein reversal of DNA hypermethylation-associated Klotho suppression ameliorates renal fibrosis in mice. Sci Rep. 2016;6:34597.CrossRefPubMedPubMedCentral
30.
Zurück zum Zitat Kuppe C, Ibrahim MM, Kranz J, et al. Decoding myofibroblast origins in human kidney fibrosis. Nature. 2021;589(7841):281–6.CrossRefPubMed Kuppe C, Ibrahim MM, Kranz J, et al. Decoding myofibroblast origins in human kidney fibrosis. Nature. 2021;589(7841):281–6.CrossRefPubMed
Metadaten
Titel
A potential link between fibroblast growth factor-23 and the progression of AKI to CKD
Publikationsdatum
01.12.2023
Erschienen in
BMC Nephrology / Ausgabe 1/2023
Elektronische ISSN: 1471-2369
DOI
https://doi.org/10.1186/s12882-023-03125-1

Weitere Artikel der Ausgabe 1/2023

BMC Nephrology 1/2023 Zur Ausgabe

Leitlinien kompakt für die Innere Medizin

Mit medbee Pocketcards sicher entscheiden.

Seit 2022 gehört die medbee GmbH zum Springer Medizin Verlag

Echinokokkose medikamentös behandeln oder operieren?

06.05.2024 DCK 2024 Kongressbericht

Die Therapie von Echinokokkosen sollte immer in spezialisierten Zentren erfolgen. Eine symptomlose Echinokokkose kann – egal ob von Hunde- oder Fuchsbandwurm ausgelöst – konservativ erfolgen. Wenn eine Op. nötig ist, kann es sinnvoll sein, vorher Zysten zu leeren und zu desinfizieren. 

Wo hapert es noch bei der Umsetzung der POMGAT-Leitlinie?

03.05.2024 DCK 2024 Kongressbericht

Seit November 2023 gibt es evidenzbasierte Empfehlungen zum perioperativen Management bei gastrointestinalen Tumoren (POMGAT) auf S3-Niveau. Vieles wird schon entsprechend der Empfehlungen durchgeführt. Wo es im Alltag noch hapert, zeigt eine Umfrage in einem Klinikverbund.

Das Risiko für Vorhofflimmern in der Bevölkerung steigt

02.05.2024 Vorhofflimmern Nachrichten

Das Risiko, im Lauf des Lebens an Vorhofflimmern zu erkranken, ist in den vergangenen 20 Jahren gestiegen: Laut dänischen Zahlen wird es drei von zehn Personen treffen. Das hat Folgen weit über die Schlaganfallgefährdung hinaus.

VHF-Ablation nützt wohl nur bei reduzierter Auswurfleistung

02.05.2024 Ablationstherapie Nachrichten

Ob die Katheterablation von Vorhofflimmern bei Patienten mit Herzinsuffizienz die Komplikationsraten senkt, scheint davon abzuhängen, ob die Auswurfleistung erhalten ist oder nicht. Das legen die Ergebnisse einer Metaanalyse nahe.

Update Innere Medizin

Bestellen Sie unseren Fach-Newsletter und bleiben Sie gut informiert.