Next Article in Journal
Genetic Cause of Hybrid Lethality Observed in Reciprocal Interspecific Crosses between Nicotiana simulans and N. tabacum
Next Article in Special Issue
High Ferritin Is Not Needed in Hemodialysis Patients: A Retrospective Study of Total Body Iron and Oral Iron Replacement Therapy
Previous Article in Journal
Changes in Photosynthetic Characteristics between Green-Leaf Poplar Linn. “2025” and Its Bud-Sporting Colored-Leaf Cultivars
Previous Article in Special Issue
Changes in Metabolomic Profiles Induced by Switching from an Erythropoiesis-Stimulating Agent to a Hypoxia-Inducible Factor Prolyl Hydroxylase Inhibitor in Hemodialysis Patients: A Pilot Study
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Article

Tubular Endogenous Erythropoietin Protects Renal Function against Ischemic Reperfusion Injury

1
Department of Physiology, Kitasato University School of Medicine, 1-15-1 Kitasato, Minami-ku, Sagamihara 252-0374, Kanagawa, Japan
2
Department of Nephrology, Kumamoto University Graduate School of Medical Sciences, 1-1-1 Honjo, Chuo-ku, Kumamoto 860-8556, Kumamoto, Japan
3
Division of Biomedical Research, Kitasato University Medical Center, 6-100 Arai, Kitamoto 364-8501, Saitama, Japan
4
Division of Kidney and Dialysis, Department of Internal Medicine, Hyogo Medical University, 1-1 Mukogawa-cho, Nishinomiya 663-8501, Hyogo, Japan
5
Division of Internal Medicine, Kitasato University Medical Center, 6-100 Arai, Kitamoto 364-8501, Saitama, Japan
6
Renal Division, Department of Medicine, Emory University School of Medicine, 1639 Pierce Drive, WMB Room 3313, Atlanta, GA 30322, USA
*
Author to whom correspondence should be addressed.
Int. J. Mol. Sci. 2024, 25(2), 1223; https://doi.org/10.3390/ijms25021223
Submission received: 24 November 2023 / Revised: 16 January 2024 / Accepted: 17 January 2024 / Published: 19 January 2024
(This article belongs to the Special Issue Updates on Erythropoietin)

Abstract

:
Many large-scale studies show that exogenous erythropoietin, erythropoiesis-stimulating agents, lack any renoprotective effects. We investigated the effects of endogenous erythropoietin on renal function in kidney ischemic reperfusion injury (IRI) using the prolyl hydroxylase domain (PHD) inhibitor, Roxadustat (ROX). Four h of hypoxia (7% O2) and 4 h treatment by ROX prior to IRI did not improve renal function. In contrast, 24–72 h pretreatment by ROX significantly improved the decline of renal function caused by IRI. Hypoxia and 4 h ROX increased interstitial cells-derived Epo production by 75- and 6-fold, respectively, before IRI, and worked similarly to exogenous Epo. ROX treatment for 24–72 h increased Epo production during IRI by 9-fold. Immunohistochemistry revealed that 24 h ROX treatment induced Epo production in proximal and distal tubules and worked similarly to endogenous Epo. Our data show that tubular endogenous Epo production induced by 24–72 h ROX treatment results in renoprotection but peritubular exogenous Epo production by interstitial cells induced by hypoxia and 4 h ROX treatment did not. Stimulation of tubular, but not peritubular, Epo production may link to renoprotection.

1. Introduction

Erythropoietin (Epo) is found in urine of anemic patients [1,2]. Erythropoiesis-stimulating agents (ESAs) changed the treatment of renal anemia [3,4,5,6]. Many large-scale studies investigated the effects of ESAs for the progression of chronic renal failure and found that ESAs have no renoprotective effects [5,7,8,9]. Prolyl hydroxylase domain (PHD) inhibitors have been used for the treatment of renal anemia instead of ESAs due to the ease of oral intake [6,10,11,12,13,14,15]. Epo produced by ESAs works similarly to exogenous Epo. In contrast, PHD inhibitors stimulate endogenous Epo production. PHD inhibitors inhibit PHD activity and increase HIF2α expression to increase endogenous Epo production [16,17,18]. Production of endogenous Epo has additional effects beyond Epo itself. Endogenous Epo is known to decrease interleukin 6 and monocyte chemoattractant protein-1 production by macrophages [19]. The difference in the physiological effect between exogenous and endogenous Epo is not clear yet. Epo pretreatment has been used as a substitute for preconditioning to ischemia [20,21,22,23,24,25,26]. Some reports show a significant improvement of ischemic reperfusion injury (IRI) by Epo pretreatment due to the reduction in inflammation or oxidative stress, but some did not. Inflammation is known to affect anemia [27]. Epo pretreatment examined the effects of exogenous Epo on renal function. We have invented the deglycosylation-coupled Western blotting method for Epo in blood, urine, and tissue [28,29,30]. Our method improved the sensitivity and specificity of Epo detection, especially in tissue. We also investigated tubular Epo production by immunohistochemistry (IHC) [28,31,32,33]. Our IHC helps to distinguish the site of Epo production and exogenous/endogenous Epo.
There are two sites of Epo production in the kidney: interstitial cells and nephrons [28,31,32,33,34,35,36,37,38,39]. Epo production by the interstitial cells is not seen in control conditions and is stimulated to extreme levels only in severe hypoxia and anemia [16,40,41]. In contrast, Epo production by the nephron is observed in the control condition and is stimulated several fold by activation of the renin–angiotensin–aldosterone system (RAS) [42,43,44,45,46]. In our study, exogenous Epo means peritubular production of Epo by the interstitial cells. Epo is also produced by the nephron and acts similarly to endogenous Epo. Thus, exogenous Epo means peritubular Epo and endogenous Epo is tubular Epo in our study. We investigated the effects of endogenous Epo on renal function in IRI, a model of acute kidney injury (AKI).

2. Results

2.1. Serum Creatinine Levels Pre- and Post-IRI

IRI caused an increase in serum creatinine from 0.53 ± 0.02 to 2.52 ± 0.29 mg/dL, the same as previous reports [21,22,23,24,25,26]. Hematoxylin–eosin (HE) staining of the kidney showed damage to proximal tubules but not in the glomerulus (Supplementary Figure S1). Severe hypoxia and 4 h ROX (R4) treatment did not decrease the serum creatinine level. In contrast, 24 h ROX (R24) and 72 h ROX (R72) significantly decreased creatinine levels (2.52   ±   0.29 ,   1.69   ±   0.64 , 1.40   ±   0.26, 0.76   ±   0.05 **, and 0.97   ±   0.19 ** mg/dL by control, hypoxia, R4, R24, and R72, respectively, ** p < 0.01 by ANOVA and Scheffe’s multiple comparison, Figure 1).

2.2. Plasma Epo Concentration before and after IRI

Plasma Epo concentration was examined before and after IRI. A high plasma Epo concentration was observed in the hypoxia and R4 groups (Figure 2). Only the R4 group showed a high plasma Epo concentration after IRI. This high plasma Epo concentration in the R4 group was thought to be caused by Epo production before IRI.

2.3. Western Blot Analysis of Epo Production during IRI

To reveal the amount of Epo production during IRI, Western blot analysis was performed using the kidney after IRI. We first examined Epo detection both in the absence and presence of Peptide-N-Glycosidase F (PNGase) (Figure 3a,b). It is difficult to detect glycosylated Epo at 35–38 kDa (left lanes in Figure 3a,b). It is easy to detect deglycosylated Epo at 22 kDa (right lanes in Figure 3a–c). Deglycosylated Epo production was examined at pre- and post-IRI. Hypoxia caused high Epo production before IRI and R4 showed a slight increase in Epo production (75− and 6−fold increase compared with control, Figure 3c). Epo production by the R4 group is lower than in our previous report. Our previous reports examined Epo production at 6 h after the injection of ROX. Therefore, the maximum increase in Epo production was thought to be caused during the formation of IRI in the R4 group. In contrast, Epo production after IRI was highest in R24 group (9-fold increase compared with control, Figure 3c).

2.4. Epo mRNA Expression before and after IRI

Epo mRNA expression was very high in the hypoxia and R4 groups and very low in the R24 and R72 groups before IRI (Figure 4). Epo mRNA after IRI was highest in the R4 group and very low in the hypoxia group. The R24 and R72 groups showed slight increases in Epo mRNA expression.

2.5. Immunohistochemistry (IHC) of Epo Production by the Kidney

Since Epo production after IRI in the kidney was stimulated by R24, we examined the site of Epo production by IHC. The R24 group showed production of Epo by the proximal convoluted tubules and the distal tubules (brown staining in Figure 5a). Proximal tubules showed higher production than distal tubules (Figure 5b,c). Interstitial cells showed no production of Epo by R24.

2.6. Western Blot Analysis of Autophagy and Apoptosis

We examined the expression of LC3-II and p62 in the kidney to determine whether autophagy is involved in the renoprotective effects of Epo. LC3-II and p62 expressions were not changed by hypoxia and ROX (Figure 6a). Next, BAX and Bcl-2 expressions in the kidney were examined to determine the participation of apoptosis in the renoprotective effects of R24. Bcl-2 expression was lower in the kidney cortex than medulla. BAX and Bcl-2 expressions were not changed by hypoxia or ROX (Figure 6b).

2.7. Tubular Protein and mRNA Expressions in the Kidney during IRI

The expressions of Hif2α and Hif1α during IRI were not changed by hypoxia and ROX except for the decline of Phd2 mRNA expression in R24-72 (Table 1). PEPCK (Pck-1) is mainly present in the proximal convoluted tubules. We examined phosphoenolpyruvate carboxykinase (PEPCK) expression after IRI. R24-72 showed higher expression than hypoxia and R4 groups (Figure 7a). Gr (Nr3c1) is present in proximal convoluted tubules and early distal tubules. Rhcg is present in distal convoluted tubules and collecting ducts. The mRNA expression of Gr and Rhcg were increased in the R24 group (Figure 7b).

3. Discussion

Our present study showed that administration of ROX 24–72 h prior to the induction of IRI protected against the decline of renal function by stimulating tubular endogenous Epo production. Stimulation of Epo production before IRI by 4 h hypoxia and during IRI by R4 largely increased interstitial cells-derived Epo production and served as exogenous Epo but did not protect against the decline of renal function. In our IRI model, hypoxia and R4 stimulated Epo production by the interstitial cells judging from the high plasma Epo concentration. Epo production by the nephron during IRI by R24-72 was small and did not increase the plasma level. Our deglycosylated Western blot and immunohistochemistry studies revealed Epo production during IRI.
Renoprotective effects of PHD inhibitors have been reported but the results are controversial [21,22,23,24,25,26,47,48,49,50,51]. Many mechanisms such as the reduction in inflammation, oxidative stress, or apoptosis have been suggested. We have employed deglycosylation-coupled Western blotting and immunohistochemistry for the detection of Epo protein in the kidney. We found that exogenous Epo has no renoprotective effects as previously revealed by many large-scale studies [7,8]. The Hir2α and Hif1α during IRI was not changed by four groups except for the decline of Phd2 mRNA expression in R24-72, suggesting that R24-72 caused endogenous Epo production by some parts of proximal and distal tubules. IRI is known to induce damage to proximal tubules. The increased expression of PEPCK and Gr mRNA by R24-72 revealed the recovery of proximal tubules. The increase in Rhcg mRNA expression showed the recovery of collecting ducts. Therefore, R24-72 seemed to recover not only proximal but also distal tubular function.
We investigated the mechanisms of renoprotection by Epo production. Autophagy and reduction in apoptosis can induce renoprotection [52,53]. However, Western blot analysis of LC3-II and p62 expression was not changed by 24–72 h ROX. The expression of BAX and Bcl-2 were also unchanged. These data suggest the lack of autophagy and apoptosis induced by ROX.
The Epo receptor has some role in Epo effects on tubular function [53]. Epo receptors in the kidney exist in the inner medullary collecting ducts [54]. Inner medullary collecting ducts are the terminal portion of the nephron and are an important site for urine concentration. Therefore, the renoprotective effect of tubule-produced Epo is not thought to be caused by activation of the Epo receptor.
It is interesting that tubules are the site of Epo production in this IRI model. Renal Epo producing (REP) cells have been known to produce large amounts of Epo, but a small production of Epo by the nephron was enough to protect against renal failure by IRI. We and others have shown that the RAS regulates Epo production by the collecting duct [31,33,40,42,43,44,45,46]. Taken together, endogenous Epo production by the tubules is thought to play an important role in the maintenance of kidney function, even with the lower capacity for Epo production. We have shown that aldosterone and angiotensin-II stimulate Epo production by the collecting ducts, especially by the intercalated cells [31,33]. Endogenous Epo production by the proximal tubules is larger than by the distal tubules in this IRI model. This is probably because IRI causes a larger amount of damage to the proximal tubules than distal ones, suggesting that proximal and distal tubules can produce Epo in response to local hypoxia. We have distinguished peritubular Epo production by REP cells from tubular endogenous Epo production. Our data clearly show that a small amount of tubular endogenous Epo production has a renoprotective effect, while the large amount of REP cell-derived peritubular Epo production has no effect on tubular function. Large-scale studies showed the lack of renoprotective effect by ESAs, the same as with 4 h hypoxia and R4 in our study [5,7,8,9]. Considering the renoprotection caused by Epo production, methods to induce tubular Epo production would be useful. One of the keys to finding useful tools for renoprotection is that the RAS has some interaction with tubular Epo production [31,33,40,42,43,44,45,46]. Our model is close to the situation of kidney transplantation. The use of PHD inhibitors 24 h before the transplant may largely decrease the damage that occurs after transplantation.
In summary, 24–72 h ROX caused renoproctive effects by stimulating tubular endogenous Epo production, but peritubular interstitial cells-derived exogenous Epo production induced by 4 h hypoxia and 4 h ROX did not repair tubular dysfunction. Methods to induce tubular endogenous Epo production might have a crucial role for renoprotection.

4. Materials and Methods

4.1. Materials and Animals

Male Sprague Dawley rats (200–250 g, Japan SLC, Hamamatsu, Japan) were used in our study. The rats were randomly assigned to two groups with the absence and presence of IRI: w/o IRI group; control and ROX group; w/IRI group were divided into 5 groups, control, 4 h hypoxia, 4 h ROX, 24 h ROX, and 72 h ROX groups. ROX (FG-4592; MedChemExpress, Monmouth Junction, NJ, USA) were dissolved in 5% glucose with 32.5 mmol/L NaOH. Rats in the ROX and ROX–IRI groups were pretreated intraperitoneal injection of ROX (50 mg/kg body weight). Control and IRI groups were injected with the same dose of 5% glucose with 32.5 mmol/L NaOH. Hypoxia group: rats were exposed to 7% O2 and 93% N2 for 4 h. After 4, 24, or 72 h pretreatment by ROX or 4 h hypoxia, all rats were anesthetized with 1.5% isoflurane in 30% O2 (a mixture of 100% O2 and air), and the left kidney was removed. IRI and ROX-IRI groups: right renal pedicle was clamped using non-traumatic vascular clips for 45 min and, then, the clamps were released to allow reperfusion to the kidney. Control and ROX groups: rats underwent the same procedure without clamping of the renal pedicles. After 24 h of I/R, all rats were anesthetized and blood samples were collected from the abdominal aorta. Kidneys were fixed in 4% PFA, embedded in paraffin, and stored in liquid nitrogen. Our protocols were checked and approved by the Ethics Committee at Kitasato University Medical Center (H24-011 and 2022-12) and Kitasato University School of Medicine (2022-140 and 2023-077).
The hypoxia group was expected to increase plasma Epo concentration at the starting time of the IRI operation and the R4 group was expected to produce Epo during the procedure of IRI [28,32]. We have shown that ROX caused high plasma Epo levels 6 h after administration [32]. The 24 h and 72 h ROX groups were expected to increase Epo production after the IRI operation.

4.2. Serum Creatinine and Plasma Epo Concentration Measurements

Blood samples were centrifuged at 3000× g for 10 min. Supernatants were collected to measure serum creatinine levels using StatSensor (Nova Biomedical, Waltham, MA, USA). Plasma Epo concentrations were measured by CLEIA (SRL, Tokyo, Japan).

4.3. Western Blot Analysis

Deglycosylation-coupled Western blot analysis was performed as described previously [26,31,32]. Protein was extracted from the kidney using CelLytic MT (C-3228; Sigma-Aldrich, Burlington, MA, USA) plus protease inhibitor (05892970001, Roche, Basel, Switzerland) and was used for Western blotting. Samples were deglycosylated using Peptide-N-Glycosidase F (PNGase, 4450; Takara Bio, Kusatsu, Japan). A total of 1 μL of 10% SDS was added to 10 μL samples and boiled for 3 min. Then, 11 μL of 2× stabilizing buffer was added. After the addition of 1 μL of PBS (to measure glycosylated Epo) or PNGase (to measure deglycosylated Epo), samples were incubated in a water bath for 17–20 h at 37 °C. After the incubation, samples were spun down and the supernatant was used for SDS-PAGE (10–20% gradient gel, 414893; Cosmo Bio, Tokyo, Japan). The 2× stabilizing buffer contained 125 mM Tris-HCl (pH 8.6), 48 mM EDTA, 4% Nonidet P-40 and 8% 2-mercaptoethanol. Recombinant rat Epo (rRatEpo, 592302; BioLegend, San Diego, USA) was used as a positive control both in glycosylated and deglycosylated samples. After SDS-PAGE, proteins were transferred to a PVDF membrane (Immobilon-P, IPVH00010; Merck Millipore, Burlington, MA, USA) for 60–90 min at 120 mA. The membrane was blocked with 5% skim milk (Morinaga, Tokyo, Japan) for 60 min and incubated with the antibody against Epo (sc-5290, 1:500; Santa Cruz) for 60 min at room temperature. After washing, the membrane was incubated with a secondary antibody (goat anti-mouse IgG (H+L) (115-035-166, 1:5000; Jackson ImmunoResearch Laboratories, West Grove, PA, USA) for 60 min. Bands were visualized by the ECL Select Western Blotting Detection System (RPN2235; GE Healthcare Bio-Science AB, Uppsala, Sweden) and LAS 4000 (Fujifilm, Tokyo, Japan). After measuring Epo protein expression, the membrane was stripped (stripping solution, Wako, RR39LR, Tokyo, Japan) and reprobed with the antibody against β-actin (MBL, M177-3, Tokyo, Japan), GAPDH (Santa Cruz, sc-32233) or α-tubulin (Santa Cruz, sc-69969) for the normalization of the band. Western blotting of LC3-II (Cell Signaling, D3U4C) [55], p62 (MBL, PM066), Bcl-2 (Santa Cruz, sc-23960), BAX (GeneTex, 127309), and PEPCK (Cayman, 10004943) was also performed in the same way as for Epo.

4.4. Real-Time Quantitative RT-PCR

RNA was extracted from kidney and liver using Qiacube and the RNeasy Mini Kit (74106; Quiagen, Venlo, The Netherlands) as described previously [28,33]. cDNA was synthesized using a Takara PrimeScript II 1st strand cDNA Synthesis Kit (6210; Takara Bio). Real-time PCR was performed using probes form Applied Biosystems, Waltham, MA, USA (β-actin Rn00667869_m1, Epo Rn00667869_m1, Hif2α Rn00576515_m1, Hif1α Rn01472831_m1, Phd2 Rn00710295_m1, Gr (Nr3c1) Rn00561369_m1, Rhcg Rn00788284_m1), and Premix Ex Taq (RP39LR; Takara Bio). mRNA expressions in control and IRI rats were compared by relative gene expression data using real-time quantitative PCR and the 2−ΔΔCT.

4.5. Immunohistochemistry

Kidney sections were immuno-stained as described previously [28,31,32,33]. In brief, the sections were blocked with 5% normal goat serum and reacted with rabbit polyclonal anti-human Epo antibody (sc-7956, 1:10; Santa Cruz Biotechnology, Santa Cruz, CA, USA), followed by Histofine simple stain MAX-PO (414341F; Nichirei Bioscience, Tokyo, Japan). Sections were stained using DAB liquid system (BSB 0016; Bio SB, Santa Barbara, CA, USA) and counterstained with Mayer’s haematoxylin (30002; Muto Pure Chemicals, Tokyo, Japan).
Images were obtained using an optical microscope (Axio Imager M2; Carl Zeiss, Oberkochen, Germany) with a digital camera (AxioCam 506, Carl Zeiss). Captured images were analysed using an image analysing system (ZEN 2, Carl Zeiss).

4.6. Statistical Analyses

Data are expressed as mean ± SEM. Statistical significance was performed using Excel Statics (BellCurve, Tokyo, Japan). Statistical significance was analyzed using ANOVA and multiple comparison of Scheffe. p < 0.05 was considered statistically significant.

Supplementary Materials

The following supporting information can be downloaded at: https://www.mdpi.com/article/10.3390/ijms25021223/s1.

Author Contributions

Y.Y., Y.I. and H.N. designed the research; T.F., T.O., N.K. and H.N. performed deglycosylation; Y.Y., T.Y., T.U. and H.N. performed Western blot analysis; Y.Y., T.O. and K.K. performed IHC; M.N., Y.S., Y.N. and H.N. performed statistical analyses; M.M., J.M.S., N.T. and K.K. advised on the experimental design and data interpretation; Y.Y., Y.I., J.M.S. and H.N. wrote the manuscript. All authors have read and agreed to the published version of the manuscript.

Funding

This study was supported by a grant to aid scientific research from the Ministry of Education, Culture, Sports, Sciences, and Technology of Japan (18K08247(YI) and 19K09226(TF)) and the Science Research Promotion Fund from the Promotion Department at the Kitasato University Medical Center (H25-003 (TF) and H24-011 (HN)).

Institutional Review Board Statement

The study was conducted according to the guidelines of the Declaration of Helsinki and approved by the Institutional Ethics Committee of the Kitasato University Medical Center: protocol code 2018032 (date of approval 22 October 2018) and 2019029 (date of approval 16 January 2021) and the Kitasato University School of Medicine: protocol code 2019141 (date of approval 2 August 2019).

Informed Consent Statement

Not applicable for studies not involving humans.

Data Availability Statement

The data presented in this study are openly available. Samples of the compounds are available from the authors.

Conflicts of Interest

The authors declare no conflicts of interest.

References

  1. Miyake, T.; Kung, C.K.; Goldwasser, E. Purification of human erythropoietin. J. Biol. Chem. 1977, 252, 5558–5564. [Google Scholar] [CrossRef] [PubMed]
  2. Jacobs, K.; Shoemaker, C.; Rudersdorf, R.; Neill, S.D.; Kaufman, R.J.; Mufson, A.; Seehra, J.; Jones, S.S.; Hewick, R.; Fritsch, E.F.; et al. Isolation and characterization of genomic and cDNA clones of human erythropoietin. Nature 1985, 313, 806–810. [Google Scholar] [CrossRef] [PubMed]
  3. Fishbane, S.; Schiller, B.; Locatelli, F.; Covic, A.C.; Provenzano, R.; Wiecek, A.; Levin, N.W.; Kaplan, M.; Macdougall, I.C.; Francisco, C.; et al. Peginesatide in patients with anemia undergoing hemodialysis. N. Engl. J. Med. 2013, 368, 307–319. [Google Scholar] [CrossRef] [PubMed]
  4. Jörg, D.J.; Fuertinger, D.H.; Kotanko, P. Mechanisms of hemoglobin cycling in anemia patients treated with erythropoiesis-stimulating agents. PLoS Comput. Biol. 2023, 19, e1010850. [Google Scholar] [CrossRef] [PubMed]
  5. Macdougall, I.C.; Provenzano, R.; Sharma, A.; Spinowitz, B.S.; Schmidt, R.J.; Pergola, P.E.; Zabaneh, R.I.; Tong-Starksen, S.; Mayo, M.R.; Tang, H.; et al. Peginesatide for anemia in patients with chronic kidney disease not receiving dialysis. N. Engl. J. Med. 2013, 368, 320–332. [Google Scholar] [CrossRef] [PubMed]
  6. Babitt, J.L.; Lin, H.Y. Mechanisms of anemia in CKD. J. Am. Soc. Nephrol. 2012, 23, 1631–1634. [Google Scholar] [CrossRef]
  7. Elliott, S.; Tomita, D.; Endre, Z. Erythropoiesis stimulating agents and reno-protection: A meta-analysis. BMC Nephrol. 2017, 18, 14. [Google Scholar] [CrossRef]
  8. Pfeffer, M.A.; Burdmann, E.A.; Chen, C.Y.; Cooper, M.E.; de Zeeuw, D.; Eckardt, K.U.; Feyzi, J.M.; Ivanovich, P.; Kewalramani, R.; Levey, A.S.; et al. A trial of darbepoetin alfa in type 2 diabetes and chronic kidney disease. N. Engl. J. Med. 2009, 361, 2019–2032. [Google Scholar] [CrossRef]
  9. Solomon, S.D.; Uno, H.; Lewis, E.F.; Eckardt, K.U.; Lin, J.; Burdmann, E.A.; de Zeeuw, D.; Ivanovich, P.; Levey, A.S.; Parfrey, P.; et al. Erythropoietic response and outcomes in kidney disease and type 2 diabetes. N. Engl. J. Med. 2010, 363, 1146–1155. [Google Scholar] [CrossRef]
  10. Akizawa, T.; Iwasaki, M.; Otsuka, T.; Reusch, M.; Misumi, T. Roxadustat Treatment of Chronic Kidney Disease-Associated Anemia in Japanese Patients Not on Dialysis: A Phase 2, Randomized, Double-Blind, Placebo-Controlled Trial. Adv. Ther. 2019, 36, 1438–1454. [Google Scholar] [CrossRef]
  11. Besarab, A.; Provenzano, R.; Hertel, J.; Zabaneh, R.; Klaus, S.J.; Lee, T.; Leong, R.; Hemmerich, S.; Yu, K.H.; Neff, T.B. Randomized placebo-controlled dose-ranging and pharmacodynamics study of roxadustat (FG-4592) to treat anemia in nondialysis-dependent chronic kidney disease (NDD-CKD) patients. Nephrol. Dial. Transplant. 2015, 30, 1665–1673. [Google Scholar] [CrossRef] [PubMed]
  12. Gupta, N.; Wish, J.B. Hypoxia-Inducible Factor Prolyl Hydroxylase Inhibitors: A Potential New Treatment for Anemia in Patients With CKD. Am. J. Kidney Dis. 2017, 69, 815–826. [Google Scholar] [CrossRef] [PubMed]
  13. Ogawa, C.; Tsuchiya, K.; Maeda, K. Hypoxia-Inducible Factor Prolyl Hydroxylase Inhibitors and Iron Metabolism. Int. J. Mol. Sci. 2023, 24, 3037. [Google Scholar] [CrossRef]
  14. Mima, A. Hypoxia-inducible factor-prolyl hydroxylase inhibitors for renal anemia in chronic kidney disease: Advantages and disadvantages. Eur. J. Pharmacol. 2021, 912, 174583. [Google Scholar] [CrossRef] [PubMed]
  15. Singh, A.K.; Carroll, K.; Perkovic, V.; Solomon, S.; Jha, V.; Johansen, K.L.; Lopes, R.D.; Macdougall, I.C.; Obrador, G.T.; Waikar, S.S.; et al. Daprodustat for the Treatment of Anemia in Patients Undergoing Dialysis. N. Engl. J. Med. 2021, 385, 2325–2335. [Google Scholar] [CrossRef]
  16. Haase, V.H. Regulation of erythropoiesis by hypoxia-inducible factors. Blood Rev. 2013, 27, 41–53. [Google Scholar] [CrossRef]
  17. Rosenberger, C.; Mandriota, S.; Jürgensen, J.S.; Wiesener, M.S.; Hörstrup, J.H.; Frei, U.; Ratcliffe, P.J.; Maxwell, P.H.; Bachmann, S.; Eckardt, K.-U. Expression of Hypoxia-Inducible Factor-1α and -2α in Hypoxic and Ischemic Rat Kidneys. J. Am. Soc. Nephrol. 2002, 13, 1721–1732. [Google Scholar] [CrossRef]
  18. Locatelli, F.; Del Vecchio, L. Hypoxia-Inducible Factor-Prolyl Hydroxyl Domain Inhibitors: From Theoretical Superiority to Clinical Noninferiority Compared with Current ESAs? J. Am. Soc. Nephrol. 2022, 33, 1966–1979. [Google Scholar] [CrossRef]
  19. Bin, S.; Cantarelli, C.; Horwitz, J.K.; Gentile, M.; Podestà, M.A.; La Manna, G.; Heeger, P.S.; Cravedi, P. Endogenous erythropoietin has immunoregulatory functions that limit the expression of autoimmune kidney disease in mice. Front. Immunol. 2023, 14, 1195662. [Google Scholar] [CrossRef]
  20. Murry, C.E.; Jennings, R.B.; Reimer, K.A. Preconditioning with ischemia: A delay of lethal cell injury in ischemic myocardium. Circulation 1986, 74, 1124–1136. [Google Scholar] [CrossRef]
  21. Liu, Q.S.; Cheng, Z.W.; Xiong, J.G.; Cheng, S.; He, X.F.; Li, X.C. Erythropoietin pretreatment exerts anti-inflammatory effects in hepatic ischemia/reperfusion-injured rats via suppression of the TLR2/NF-κB pathway. Transplant. Proc. 2015, 47, 283–289. [Google Scholar] [CrossRef]
  22. Zhang, J.; Zou, Y.R.; Zhong, X.; Deng, H.D.; Pu, L.; Peng, K.; Wang, L. Erythropoietin pretreatment ameliorates renal ischaemia-reperfusion injury by activating PI3K/Akt signalling. Nephrology 2015, 20, 266–272. [Google Scholar] [CrossRef] [PubMed]
  23. Elshiekh, M.; Kadkhodaee, M.; Seifi, B.; Ranjbaran, M.; Ahghari, P. Ameliorative Effect of Recombinant Human Erythropoietin and Ischemic Preconditioning on Renal Ischemia Reperfusion Injury in Rats. Nephrourol. Mon. 2015, 7, e31152. [Google Scholar] [CrossRef] [PubMed]
  24. Chou, Y.H.; Liao, F.L.; Chen, Y.T.; Yeh, P.Y.; Liu, C.H.; Shih, H.M.; Chang, F.C.; Chiang, W.C.; Chu, T.S.; Lin, S.L. Erythropoietin modulates macrophages but not post-ischemic acute kidney injury in mice. J. Formos. Med. Assoc. 2019, 118, 494–503. [Google Scholar] [CrossRef] [PubMed]
  25. Miao, A.F.; Liang, J.X.; Yao, L.; Han, J.L.; Zhou, L.J. Hypoxia-inducible factor prolyl hydroxylase inhibitor roxadustat (FG-4592) protects against renal ischemia/reperfusion injury by inhibiting inflammation. Ren. Fail. 2021, 43, 803–810. [Google Scholar] [CrossRef] [PubMed]
  26. Zhang, M.; Dong, R.; Yuan, J.; Da, J.; Zha, Y.; Long, Y. Roxadustat (FG-4592) protects against ischaemia/reperfusion-induced acute kidney injury through inhibiting the mitochondrial damage pathway in mice. Clin. Exp. Pharmacol. Physiol. 2022, 49, 311–318. [Google Scholar] [CrossRef]
  27. Weiss, G.; Ganz, T.; Goodnough, L.T. Anemia of inflammation. Blood 2019, 133, 40–50. [Google Scholar] [CrossRef]
  28. Yasuoka, Y.; Fukuyama, T.; Izumi, Y.; Nakayama, Y.; Inoue, H.; Yanagita, K.; Oshima, T.; Yamazaki, T.; Uematsu, T.; Kobayashi, N.; et al. Erythropoietin production by the kidney and the liver in response to severe hypoxia evaluated by Western blotting with deglycosylation. Physiol. Rep. 2020, 8, e14485. [Google Scholar] [CrossRef]
  29. Yasuoka, Y.; Fukuyama, T.; Izumi, Y.; Yamashita, T.; Nakayama, Y.; Inoue, H.; Yanagita, K.; Oshima, T.; Yamazaki, T.; Uematsu, T.; et al. Differentiation of endogenous erythropoietin and exogenous ESAs by Western blotting. Heliyon 2020, 6, e05389. [Google Scholar] [CrossRef]
  30. Yasuoka, Y.; Izumi, Y.; Sands, J.M.; Kawahara, K.; Nonoguchi, H. Progress in the Detection of Erythropoietin in Blood, Urine, and Tissue. Molecules 2023, 28, 4446. [Google Scholar] [CrossRef]
  31. Yasuoka, Y.; Izumi, Y.; Fukuyama, T.; Inoue, H.; Oshima, T.; Yamazaki, T.; Uematsu, T.; Kobayashi, N.; Shimada, Y.; Nagaba, Y.; et al. Effects of Angiotensin II on Erythropoietin Production in the Kidney and Liver. Molecules 2021, 26, 5399. [Google Scholar] [CrossRef] [PubMed]
  32. Yasuoka, Y.; Izumi, Y.; Fukuyama, T.; Omiya, H.; Pham, T.D.; Inoue, H.; Oshima, T.; Yamazaki, T.; Uematsu, T.; Kobayashi, N.; et al. Effects of Roxadustat on Erythropoietin Production in the Rat Body. Molecules 2022, 27, 1119. [Google Scholar] [CrossRef] [PubMed]
  33. Yasuoka, Y.; Izumi, Y.; Nagai, T.; Fukuyama, T.; Nakayama, Y.; Inoue, H.; Horikawa, K.; Kimura, M.; Nanami, M.; Yanagita, K.; et al. Fludrocortisone stimulates erythropoietin production in the intercalated cells of the collecting ducts. Biochem. Biophys. Res. Commun. 2018, 503, 3121–3127. [Google Scholar] [CrossRef] [PubMed]
  34. Obara, N.; Suzuki, N.; Kim, K.; Nagasawa, T.; Imagawa, S.; Yamamoto, M. Repression via the GATA box is essential for tissue-specific erythropoietin gene expression. Blood 2008, 111, 5223–5232. [Google Scholar] [CrossRef] [PubMed]
  35. Sato, K.; Hirano, I.; Sekine, H.; Miyauchi, K.; Nakai, T.; Kato, K.; Ito, S.; Yamamoto, M.; Suzuki, N. An immortalized cell line derived from renal erythropoietin-producing (REP) cells demonstrates their potential to transform into myofibroblasts. Sci. Rep. 2019, 9, 11254. [Google Scholar] [CrossRef] [PubMed]
  36. Suzuki, N.; Yamamoto, M. Roles of renal erythropoietin-producing (REP) cells in the maintenance of systemic oxygen homeostasis. Pflugers Arch. 2016, 468, 3–12. [Google Scholar] [CrossRef] [PubMed]
  37. Bapst, A.M.; Knöpfel, T.; Nolan, K.A.; Imeri, F.; Schuh, C.D.; Hall, A.M.; Guo, J.; Katschinski, D.M.; Wenger, R.H. Neurogenic and pericytic plasticity of conditionally immortalized cells derived from renal erythropoietin-producing cells. J. Cell Physiol. 2022, 237, 2420–2433. [Google Scholar] [CrossRef]
  38. Kobayashi, H.; Davidoff, O.; Pujari-Palmer, S.; Drevin, M.; Haase, V.H. EPO synthesis induced by HIF-PHD inhibition is dependent on myofibroblast transdifferentiation and colocalizes with non-injured nephron segments in murine kidney fibrosis. Acta Physiol. 2022, 235, e13826. [Google Scholar] [CrossRef]
  39. Nagai, T.; Yasuoka, Y.; Izumi, Y.; Horikawa, K.; Kimura, M.; Nakayama, Y.; Uematsu, T.; Fukuyama, T.; Yamazaki, T.; Kohda, Y.; et al. Reevaluation of erythropoietin production by the nephron. Biochem. Biophys. Res. Commun. 2014, 449, 222–228. [Google Scholar] [CrossRef]
  40. Jelkmann, W. Regulation of erythropoietin production. J. Physiol. 2011, 589, 1251–1258. [Google Scholar] [CrossRef]
  41. Koury, M.J.; Haase, V.H. Anaemia in kidney disease: Harnessing hypoxia responses for therapy. Nat. Rev. Nephrol. 2015, 11, 394–410. [Google Scholar] [CrossRef] [PubMed]
  42. Kato, H.; Ishida, J.; Imagawa, S.; Saito, T.; Suzuki, N.; Matsuoka, T.; Sugaya, T.; Tanimoto, K.; Yokoo, T.; Ohneda, O.; et al. Enhanced erythropoiesis mediated by activation of the renin-angiotensin system via angiotensin II type 1a receptor. Faseb. J. 2005, 19, 2023–2025. [Google Scholar] [CrossRef]
  43. Kato, H.; Ishida, J.; Matsusaka, T.; Ishimaru, T.; Tanimoto, K.; Sugiyama, F.; Yagami, K.; Nangaku, M.; Fukamizu, A. Erythropoiesis and Blood Pressure Are Regulated via AT1 Receptor by Distinctive Pathways. PLoS ONE 2015, 10, e0129484. [Google Scholar] [CrossRef]
  44. Kim, Y.C.; Mungunsukh, O.; Day, R.M. Erythropoietin Regulation by Angiotensin II. Vitam. Horm. 2017, 105, 57–77. [Google Scholar] [CrossRef] [PubMed]
  45. Kim, Y.C.; Mungunsukh, O.; McCart, E.A.; Roehrich, P.J.; Yee, D.K.; Day, R.M. Mechanism of erythropoietin regulation by angiotensin II. Mol. Pharmacol. 2014, 85, 898–908. [Google Scholar] [CrossRef]
  46. Gossmann, J.; Burkhardt, R.; Harder, S.; Lenz, T.; Sedlmeyer, A.; Klinkhardt, U.; Geiger, H.; Scheuermann, E.H. Angiotensin II infusion increases plasma erythropoietin levels via an angiotensin II type 1 receptor-dependent pathway. Kidney Int. 2001, 60, 83–86. [Google Scholar] [CrossRef] [PubMed]
  47. Ito, M.; Tanaka, T.; Ishii, T.; Wakashima, T.; Fukui, K.; Nangaku, M. Prolyl hydroxylase inhibition protects the kidneys from ischemia via upregulation of glycogen storage. Kidney Int. 2020, 97, 687–701. [Google Scholar] [CrossRef]
  48. Joharapurkar, A.A.; Patel, V.J.; Kshirsagar, S.G.; Patel, M.S.; Savsani, H.H.; Jain, M.R. Prolyl hydroxylase inhibitor desidustat protects against acute and chronic kidney injury by reducing inflammatory cytokines and oxidative stress. Drug Dev. Res. 2021, 82, 852–860. [Google Scholar] [CrossRef]
  49. Bernhardt, W.M.; Câmpean, V.; Kany, S.; Jürgensen, J.S.; Weidemann, A.; Warnecke, C.; Arend, M.; Klaus, S.; Günzler, V.; Amann, K.; et al. Preconditional activation of hypoxia-inducible factors ameliorates ischemic acute renal failure. J. Am. Soc. Nephrol. 2006, 17, 1970–1978. [Google Scholar] [CrossRef]
  50. Kapitsinou, P.P.; Jaffe, J.; Michael, M.; Swan, C.E.; Duffy, K.J.; Erickson-Miller, C.L.; Haase, V.H. Preischemic targeting of HIF prolyl hydroxylation inhibits fibrosis associated with acute kidney injury. Am. J. Physiol. Renal. Physiol. 2012, 302, F1172–F1179. [Google Scholar] [CrossRef]
  51. Wang, Z.; Schley, G.; Türkoglu, G.; Burzlaff, N.; Amann, K.U.; Willam, C.; Eckardt, K.U.; Bernhardt, W.M. The protective effect of prolyl-hydroxylase inhibition against renal ischaemia requires application prior to ischaemia but is superior to EPO treatment. Nephrol. Dial. Transplant. 2012, 27, 929–936. [Google Scholar] [CrossRef] [PubMed]
  52. Arcasoy, M.O. The non-haematopoietic biological effects of erythropoietin. Br. J. Haematol. 2008, 141, 14–31. [Google Scholar] [CrossRef] [PubMed]
  53. Shi, M.; Flores, B.; Li, P.; Gillings, N.; McMillan, K.L.; Ye, J.; Huang, L.J.; Sidhu, S.S.; Zhong, Y.P.; Grompe, M.T.; et al. Effects of erythropoietin receptor activity on angiogenesis, tubular injury, and fibrosis in acute kidney injury: A “U-shaped” relationship. Am. J. Physiol. Renal. Physiol. 2018, 314, F501–F516. [Google Scholar] [CrossRef] [PubMed]
  54. Lee, J.W.; Chou, C.L.; Knepper, M.A. Deep Sequencing in Microdissected Renal Tubules Identifies Nephron Segment-Specific Transcriptomes. J. Am. Soc. Nephrol. 2015, 26, 2669–2677. [Google Scholar] [CrossRef]
  55. Mizushima, N.; Yoshimori, T. How to interpret LC3 immunoblotting. Autophagy 2007, 3, 542–545. [Google Scholar] [CrossRef]
Figure 1. Changes in renal function by IRI. Serum creatinine levels by IRI and Epo stimulation. IRI caused an increase in serum creatinine from 0.53 to 2.52 mg/dL. Hypoxia and R4 did not decrease serum creatinine levels. In contrast, R24 and R72 decreased serum creatinine levels to 0.76 and 0.97, respectively. ** p < 0.01, *** p < 0.001 by ANOVA and Scheffe’s multiple comparison. C, control; H, hypoxia; R, ROX.
Figure 1. Changes in renal function by IRI. Serum creatinine levels by IRI and Epo stimulation. IRI caused an increase in serum creatinine from 0.53 to 2.52 mg/dL. Hypoxia and R4 did not decrease serum creatinine levels. In contrast, R24 and R72 decreased serum creatinine levels to 0.76 and 0.97, respectively. ** p < 0.01, *** p < 0.001 by ANOVA and Scheffe’s multiple comparison. C, control; H, hypoxia; R, ROX.
Ijms 25 01223 g001
Figure 2. Plasma Epo levels after IRI. Plasma levels of Epo in each group. Hypoxia caused a large increase in plasma Epo level. R4 caused a slight increase in plasma Epo level. After IRI, R4 caused an increase in plasma Epo level but hypoxia and R24−72 did not change the levels. The numbers in parenthesis shows the number of samples. * p < 0.05, ** p < 0.01 by ANOVA and Scheffe’s multiple comparison. C, control; H, hypoxia; R, ROX.
Figure 2. Plasma Epo levels after IRI. Plasma levels of Epo in each group. Hypoxia caused a large increase in plasma Epo level. R4 caused a slight increase in plasma Epo level. After IRI, R4 caused an increase in plasma Epo level but hypoxia and R24−72 did not change the levels. The numbers in parenthesis shows the number of samples. * p < 0.05, ** p < 0.01 by ANOVA and Scheffe’s multiple comparison. C, control; H, hypoxia; R, ROX.
Ijms 25 01223 g002
Figure 3. (a). Western blot analysis of Epo protein before IRI in the absence and presence of PNGase. Epo production by the kidney before IRI. A typical gel of pre-IRI is shown. Highest production is observed by 4 h hypoxia. C, control; H, hypoxia; R, ROX. (b). Western blot analysis of Epo protein after IRI in the absence and presence of PNGase. Epo production by the kidney after IRI. A typical gel of post-IRI is shown. Highest production is observed by 24 h ROX. C, control; H, hypoxia; R, ROX. (c). Epo production by the kidney before and after IRI. Western blot analysis of Epo production by the renal cortex. Hypoxia induced a large amount of Epo production before IRI. After IRI, R24 caused a slight increase in Epo production. R4 did not induce Epo production after IRI, suggesting that the high plasma Epo level is caused by Epo production before IRI. Deglycosylated Epo expression at 22 kDa (degEpo shown by the left arrow head) was corrected by β-actin expression (degEpo/β-actin), * p < 0.05 by ANOVA and Scheffe’s multiple comparison. n = 4–5. C, control; H, hypoxia; R, ROX.
Figure 3. (a). Western blot analysis of Epo protein before IRI in the absence and presence of PNGase. Epo production by the kidney before IRI. A typical gel of pre-IRI is shown. Highest production is observed by 4 h hypoxia. C, control; H, hypoxia; R, ROX. (b). Western blot analysis of Epo protein after IRI in the absence and presence of PNGase. Epo production by the kidney after IRI. A typical gel of post-IRI is shown. Highest production is observed by 24 h ROX. C, control; H, hypoxia; R, ROX. (c). Epo production by the kidney before and after IRI. Western blot analysis of Epo production by the renal cortex. Hypoxia induced a large amount of Epo production before IRI. After IRI, R24 caused a slight increase in Epo production. R4 did not induce Epo production after IRI, suggesting that the high plasma Epo level is caused by Epo production before IRI. Deglycosylated Epo expression at 22 kDa (degEpo shown by the left arrow head) was corrected by β-actin expression (degEpo/β-actin), * p < 0.05 by ANOVA and Scheffe’s multiple comparison. n = 4–5. C, control; H, hypoxia; R, ROX.
Ijms 25 01223 g003aIjms 25 01223 g003b
Figure 4. Epo mRNA expression before and after IRI. (a): Hypoxia and R4 induced large increases in Epo mRNA expressions before IRI. (b): A slight stimulation of Epo mRNA expression was observed by R4, R24, and R72 after IRI. n = 3–7. H, hypoxia; R, ROX. * p < 0.05 by ANOVA and Dunnett’s multiple comparison.
Figure 4. Epo mRNA expression before and after IRI. (a): Hypoxia and R4 induced large increases in Epo mRNA expressions before IRI. (b): A slight stimulation of Epo mRNA expression was observed by R4, R24, and R72 after IRI. n = 3–7. H, hypoxia; R, ROX. * p < 0.05 by ANOVA and Dunnett’s multiple comparison.
Ijms 25 01223 g004
Figure 5. Immunohistochemical study of Epo production sites by kidney in IRI. (a). Epo staining was not observed in control and ROX without IRI. Epo staining is detected in 24 h ROX but not in control after IRI (upper panel). Strong vesicular staining was detected only in the proximal tubules (middle panel) and weak staining was detected in the distal tubules (distal convoluted tubules and cortical collecting ducts) (lower panel) after IRI with 24 h ROX. (b,c). DAB staining intensity of proximal tubules and distal tubules. ** p < 0.001 by ANOVA and Scheffe’s multiple comparison, n = 4–8. PT, proximal tubule; DT, distal tubule.
Figure 5. Immunohistochemical study of Epo production sites by kidney in IRI. (a). Epo staining was not observed in control and ROX without IRI. Epo staining is detected in 24 h ROX but not in control after IRI (upper panel). Strong vesicular staining was detected only in the proximal tubules (middle panel) and weak staining was detected in the distal tubules (distal convoluted tubules and cortical collecting ducts) (lower panel) after IRI with 24 h ROX. (b,c). DAB staining intensity of proximal tubules and distal tubules. ** p < 0.001 by ANOVA and Scheffe’s multiple comparison, n = 4–8. PT, proximal tubule; DT, distal tubule.
Ijms 25 01223 g005
Figure 6. (a). Effects of hypoxia and ROX on autophagy. The effect of hypoxia and ROX on autophagy was examined. LC3-II (lower bands) and p62 (upper bands) expression was not changed by hypoxia and ROX. n = 4–17. C, control; H, hypoxia; R, ROX. (b). Effects of hypoxia and ROX on apoptosis. The effect of hypoxia and ROX on BAX and Bcl-2 expression. Hypoxia and ROX did not change BAX and Bcl-2 expression in renal cortex and medulla. C, control; H, hypoxia; R, ROX.
Figure 6. (a). Effects of hypoxia and ROX on autophagy. The effect of hypoxia and ROX on autophagy was examined. LC3-II (lower bands) and p62 (upper bands) expression was not changed by hypoxia and ROX. n = 4–17. C, control; H, hypoxia; R, ROX. (b). Effects of hypoxia and ROX on apoptosis. The effect of hypoxia and ROX on BAX and Bcl-2 expression. Hypoxia and ROX did not change BAX and Bcl-2 expression in renal cortex and medulla. C, control; H, hypoxia; R, ROX.
Ijms 25 01223 g006
Figure 7. (a). Effects of hypoxia and ROX on nephron-specific protein expression after IRI. PEPCK expression was increased by R24-72. * p < 0.05 by ANOVA and Dunnett’s multiple comparison, n = 4–5. C, control; H, hypoxia; R, ROX. (b). Effects of hypoxia and ROX on nephron-specific mRNA expressions after IRI. Gr (Nr3c1) mRNA and Rhcg mRNA expressions were increased in R24 after IRI. * p < 0.05 by ANOVA and Scheffe’s multiple comparison. n = 3–7. C, control; H, hypoxia; R, ROX.
Figure 7. (a). Effects of hypoxia and ROX on nephron-specific protein expression after IRI. PEPCK expression was increased by R24-72. * p < 0.05 by ANOVA and Dunnett’s multiple comparison, n = 4–5. C, control; H, hypoxia; R, ROX. (b). Effects of hypoxia and ROX on nephron-specific mRNA expressions after IRI. Gr (Nr3c1) mRNA and Rhcg mRNA expressions were increased in R24 after IRI. * p < 0.05 by ANOVA and Scheffe’s multiple comparison. n = 3–7. C, control; H, hypoxia; R, ROX.
Ijms 25 01223 g007
Table 1. The mRNA expression of Hif2α, Hif1α, and Phd2 in the renal cortex before and after IRI. The mRNA expression of Hif2α, Hif1α, and Phd2 was examined in the renal cortex before and after IRI. The decrease in Phd2 mRNA expression was observed in the R24 and R72 groups after IRI. Data are expressed as mean ± SD. n = 3–7. * p < 0.05 by Student’s t-test.
Table 1. The mRNA expression of Hif2α, Hif1α, and Phd2 in the renal cortex before and after IRI. The mRNA expression of Hif2α, Hif1α, and Phd2 was examined in the renal cortex before and after IRI. The decrease in Phd2 mRNA expression was observed in the R24 and R72 groups after IRI. Data are expressed as mean ± SD. n = 3–7. * p < 0.05 by Student’s t-test.
HIF2αHIF1αPHD2
Pre-IRIH1.16 ± 0.160.81 ± 0.110.91 ± 0.52
R40.71 ± 0.749.37 ± 11.660.91 ± 0.53
R241.27 ± 0.532.81 ± 3.281.01 ± 0.82
R720.91 ± 0.640.56 ± 0.021.26 ± 1.05
Post-IRIH1.12 ± 0.241.14 ± 0.820.76 ± 0.63
R41.25 ± 0.910.77 ± 0.471.65 ± 1.72
R241.04 ± 0.231.54 ± 1.870.54 ± 0.29 *
R721.20 ± 0.911.05 ± 0.210.48 ± 0.30 *
Disclaimer/Publisher’s Note: The statements, opinions and data contained in all publications are solely those of the individual author(s) and contributor(s) and not of MDPI and/or the editor(s). MDPI and/or the editor(s) disclaim responsibility for any injury to people or property resulting from any ideas, methods, instructions or products referred to in the content.

Share and Cite

MDPI and ACS Style

Yasuoka, Y.; Izumi, Y.; Fukuyama, T.; Oshima, T.; Yamazaki, T.; Uematsu, T.; Kobayashi, N.; Nanami, M.; Shimada, Y.; Nagaba, Y.; et al. Tubular Endogenous Erythropoietin Protects Renal Function against Ischemic Reperfusion Injury. Int. J. Mol. Sci. 2024, 25, 1223. https://doi.org/10.3390/ijms25021223

AMA Style

Yasuoka Y, Izumi Y, Fukuyama T, Oshima T, Yamazaki T, Uematsu T, Kobayashi N, Nanami M, Shimada Y, Nagaba Y, et al. Tubular Endogenous Erythropoietin Protects Renal Function against Ischemic Reperfusion Injury. International Journal of Molecular Sciences. 2024; 25(2):1223. https://doi.org/10.3390/ijms25021223

Chicago/Turabian Style

Yasuoka, Yukiko, Yuichiro Izumi, Takashi Fukuyama, Tomomi Oshima, Taiga Yamazaki, Takayuki Uematsu, Noritada Kobayashi, Masayoshi Nanami, Yoshitaka Shimada, Yasushi Nagaba, and et al. 2024. "Tubular Endogenous Erythropoietin Protects Renal Function against Ischemic Reperfusion Injury" International Journal of Molecular Sciences 25, no. 2: 1223. https://doi.org/10.3390/ijms25021223

Note that from the first issue of 2016, this journal uses article numbers instead of page numbers. See further details here.

Article Metrics

Back to TopTop