Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 31
Filter
1.
Vet Sci ; 10(2)2023 Feb 05.
Article in English | MEDLINE | ID: mdl-36851425

ABSTRACT

Red blood cell (RBC) transfusion is associated with recipient inflammation and infection, which may be triggered by excessive circulating iron. Iron chelation following transfusion may reduce these risks. The aim of this study was to evaluate the effect of deferoxamine on circulating iron and inflammation biomarkers over time and in vitro growth of Escherichia coli (E. coli) following RBC transfusion in dogs with atraumatic hemorrhage. Anesthetized dogs were subject to atraumatic hemorrhage and transfusion of RBCs, then randomized to receive either deferoxamine or saline placebo of equivalent volume (n = 10 per group) in a blinded fashion. Blood was sampled before hemorrhage and then 2, 4, and 6 h later. Following hemorrhage and RBC transfusion, free iron increased in all dogs over time (both p < 0.001). Inflammation biomarkers interleukin-6 (IL6), CXC motif chemokine-8 (CXCL8), interleukin-10 (IL10), and keratinocyte-derived chemokine (KC) increased in all dogs over time (all p < 0.001). Logarithmic growth of E. coli clones within blood collected 6 h post-transfusion was not different between groups. Only total iron-binding capacity was different between groups over time, being significantly increased in the deferoxamine group at 2 and 4 h post-transfusion (both p < 0.001). In summary, while free iron and inflammation biomarkers increased post-RBC transfusion, deferoxamine administration did not impact circulating free iron, inflammation biomarkers, or in vitro growth of E. coli when compared with placebo.

2.
Int J Mol Sci ; 23(3)2022 Jan 25.
Article in English | MEDLINE | ID: mdl-35163276

ABSTRACT

Hemolysis is known to cause acute kidney injury (AKI). The iron regulatory hormone hepcidin, produced by renal distal tubules, is suggested to exert a renoprotective role during this pathology. We aimed to elucidate the molecular mechanisms of renal hepcidin synthesis and its protection against hemoglobin-induced AKI. In contrast to known hepatic hepcidin induction, incubation of mouse cortical collecting duct (mCCDcl1) cells with IL-6 or LPS did not induce Hamp1 mRNA expression, whereas iron (FeS) and hemin significantly induced hepcidin synthesis (p < 0.05). Moreover, iron/heme-mediated hepcidin induction in mCCDcl1 cells was caused by the nuclear factor erythroid 2-related factor 2 (Nrf2) pathway, as indicated by increased nuclear Nrf2 translocation and induced expression of Nrf2 downstream targets GCLM (p < 0.001), NQO1 (p < 0.001), and TXNRD1 (p < 0.005), which could be prevented by the known Nrf2 inhibitor trigonelline. Newly created inducible kidney-specific hepcidin KO mice demonstrated a significant reduction in renal Hamp1 mRNA expression. Phenylhydrazine (PHZ)-induced hemolysis caused renal iron loading and oxidative stress in both wildtype (Wt) and KO mice. PHZ treatment in Wt induced inflammatory markers (IL-6, TNFα) but not Hamp1. However, since PHZ treatment also significantly reduced systemic hepcidin levels in both Wt and KO mice (both p < 0.001), a dissection between the roles of systemic and renal hepcidin could not be made. Combined, the results of our study indicate that there are kidney-specific mechanisms in hepcidin regulation, as indicated by the dominant role of iron and not inflammation as an inducer of renal hepcidin, but also emphasize the complex interplay of various iron regulatory mechanisms during AKI on a local and systemic level.


Subject(s)
Acute Kidney Injury/metabolism , Hepcidins/metabolism , Acute Kidney Injury/chemically induced , Acute Kidney Injury/physiopathology , Animals , Hemin/metabolism , Hemoglobins/metabolism , Hemolysis/physiology , Hepcidins/physiology , Iron/metabolism , Kidney/metabolism , Kidney/pathology , Kidney Tubules, Distal/metabolism , Mice , Mice, Knockout , Oxidative Stress
3.
Sci Rep ; 12(1): 1199, 2022 01 24.
Article in English | MEDLINE | ID: mdl-35075227

ABSTRACT

Kidney iron deposition may play a role in the progression of tubulointerstitial injury during chronic kidney disease. Here, we studied the molecular mechanisms of kidney iron loading in experimental focal segmental glomerulosclerosis (FSGS) and investigated the effect of iron-reducing interventions on disease progression. Thy-1.1 mice were injected with anti-Thy-1.1 monoclonal antibody (mAb) to induce proteinuria. Urine, blood and tissue were collected at day (D)1, D5, D8, D15 and D22 after mAb injection. Thy-1.1 mice were subjected to captopril (CA), iron-deficient (ID) diet or iron chelation (deferoxamine; DFO). MAb injection resulted in significant albuminuria at all time points (p < 0.01). Kidney iron loading, predominantly in distal tubules, increased in time, along with urinary kidney injury molecule-1 and 24p3 concentration, as well as kidney mRNA expression of Interleukin-6 (Il-6) and Heme oxygenase-1 (Ho-1). Treatment with CA, ID diet or DFO significantly reduced kidney iron deposition at D8 and D22 (p < 0.001) and fibrosis at D22 (p < 0.05), but not kidney Il-6. ID treatment increased kidney Ho-1 (p < 0.001). In conclusion, kidney iron accumulation coincides with progression of tubulointerstitial injury in this model of FSGS. Reduction of iron loading halts disease progression. However, targeted approaches to prevent excessive kidney iron loading are warranted to maintain the delicate systemic and cellular iron balance.


Subject(s)
Glomerulosclerosis, Focal Segmental/metabolism , Iron/metabolism , Kidney Tubules, Distal/metabolism , Angiotensin-Converting Enzyme Inhibitors/therapeutic use , Animals , Captopril/therapeutic use , Deferoxamine/therapeutic use , Disease Models, Animal , Female , Glomerulosclerosis, Focal Segmental/diet therapy , Glomerulosclerosis, Focal Segmental/drug therapy , Male , Mice , Receptors, Cell Surface/metabolism , Siderophores/therapeutic use
4.
PLoS One ; 16(7): e0254851, 2021.
Article in English | MEDLINE | ID: mdl-34283879

ABSTRACT

Erythroferrone (ERFE), the erythroid regulator of iron metabolism, inhibits hepcidin to increase iron availability for erythropoiesis. ERFE plays a pathological role during ineffective erythropoiesis as occurs in X-linked sideroblastic anemia (XLSA) and ß-thalassemia. Its measurement might serve as an indicator of severity for these diseases. However, for reliable quantification of ERFE analytical characterization is indispensable to determine the assay's limitations and define proper methodology. We developed a sandwich ELISA for human serum ERFE using polyclonal antibodies and report its extensive analytical validation. This new assay showed, for the first time, the differentiation of XLSA and ß-thalassemia major patients from healthy controls (p = 0.03) and from each other (p<0.01), showing the assay provides biological plausible results. Despite poor dilution linearity, parallelism and recovery in patient serum matrix, which indicated presence of a matrix effect and/or different immunoreactivity of the antibodies to the recombinant standard and the endogenous analyte, our assay correlated well with two other existing ERFE ELISAs (both R2 = 0.83). Nevertheless, employment of one optimal dilution of all serum samples is warranted to obtain reliable results. When adequately performed, the assay can be used to further unravel the human erythropoiesis-hepcidin-iron axis in various disorders and assess the added diagnostic value of ERFE.


Subject(s)
Anemia, Sideroblastic/diagnosis , Peptide Hormones/analysis , beta-Thalassemia/diagnosis , Adult , Aged , Anemia/blood , Anemia/diagnosis , Anemia, Sideroblastic/blood , Cytokines/metabolism , Enzyme-Linked Immunosorbent Assay , Erythropoiesis , Female , Hepcidins/blood , Humans , Iron/metabolism , Iron Overload/diagnosis , Iron Overload/metabolism , Male , Middle Aged , Peptide Hormones/blood , beta-Thalassemia/blood
5.
Arch Toxicol ; 95(8): 2719-2735, 2021 08.
Article in English | MEDLINE | ID: mdl-34181029

ABSTRACT

The liver hormone hepcidin regulates systemic iron homeostasis. Hepcidin is also expressed by the kidney, but exclusively in distal nephron segments. Several studies suggest hepcidin protects against kidney damage involving Fe2+ overload. The nephrotoxic non-essential metal ion Cd2+ can displace Fe2+ from cellular biomolecules, causing oxidative stress and cell death. The role of hepcidin in Fe2+ and Cd2+ toxicity was assessed in mouse renal cortical [mCCD(cl.1)] and inner medullary [mIMCD3] collecting duct cell lines. Cells were exposed to equipotent Cd2+ (0.5-5 µmol/l) and/or Fe2+ (50-100 µmol/l) for 4-24 h. Hepcidin (Hamp1) was transiently silenced by RNAi or overexpressed by plasmid transfection. Hepcidin or catalase expression were evaluated by RT-PCR, qPCR, immunoblotting or immunofluorescence microscopy, and cell fate by MTT, apoptosis and necrosis assays. Reactive oxygen species (ROS) were detected using CellROX™ Green and catalase activity by fluorometry. Hepcidin upregulation protected against Fe2+-induced mIMCD3 cell death by increasing catalase activity and reducing ROS, but exacerbated Cd2+-induced catalase dysfunction, increasing ROS and cell death. Opposite effects were observed with Hamp1 siRNA. Similar to Hamp1 silencing, increased intracellular Fe2+ prevented Cd2+ damage, ROS formation and catalase disruption whereas chelation of intracellular Fe2+ with desferrioxamine augmented Cd2+ damage, corresponding to hepcidin upregulation. Comparable effects were observed in mCCD(cl.1) cells, indicating equivalent functions of renal hepcidin in different collecting duct segments. In conclusion, hepcidin likely binds Fe2+, but not Cd2+. Because Fe2+ and Cd2+ compete for functional binding sites in proteins, hepcidin affects their free metal ion pools and differentially impacts downstream processes and cell fate.


Subject(s)
Cadmium/toxicity , Hepcidins/genetics , Iron/toxicity , Oxidative Stress/drug effects , Animals , Apoptosis/drug effects , Binding Sites , Binding, Competitive , Cadmium/administration & dosage , Cell Death/drug effects , Cell Line , Cells, Cultured , Deferoxamine/pharmacology , Female , Gene Silencing , Iron/administration & dosage , Kidney Tubules, Collecting/cytology , Kidney Tubules, Collecting/drug effects , Male , Mice , Mice, Inbred C57BL , Reactive Oxygen Species/metabolism
6.
Int J Sport Nutr Exerc Metab ; 31(2): 101-108, 2021 03 01.
Article in English | MEDLINE | ID: mdl-33383570

ABSTRACT

This study implemented a 2-week high carbohydrate (CHO) diet intended to maximize CHO oxidation rates and examined the iron-regulatory response to a 26-km race walking effort. Twenty international-level, male race walkers were assigned to either a novel high CHO diet (MAX = 10 g/kg body mass CHO daily) inclusive of gut-training strategies, or a moderate CHO control diet (CON = 6 g/kg body mass CHO daily) for a 2-week training period. The athletes completed a 26-km race walking test protocol before and after the dietary intervention. Venous blood samples were collected pre-, post-, and 3 hr postexercise and measured for serum ferritin, interleukin-6, and hepcidin-25 concentrations. Similar decreases in serum ferritin (17-23%) occurred postintervention in MAX and CON. At the baseline, CON had a greater postexercise increase in interleukin-6 levels after 26 km of walking (20.1-fold, 95% CI [9.2, 35.7]) compared with MAX (10.2-fold, 95% CI [3.7, 18.7]). A similar finding was evident for hepcidin levels 3 hr postexercise (CON = 10.8-fold, 95% CI [4.8, 21.2]; MAX = 8.8-fold, 95% CI [3.9, 16.4]). Postintervention, there were no substantial differences in the interleukin-6 response (CON = 13.6-fold, 95% CI [9.2, 20.5]; MAX = 11.2-fold, 95% CI [6.5, 21.3]) or hepcidin levels (CON = 7.1-fold, 95% CI [2.1, 15.4]; MAX = 6.3-fold, 95% CI [1.8, 14.6]) between the dietary groups. Higher resting serum ferritin (p = .004) and hotter trial ambient temperatures (p = .014) were associated with greater hepcidin levels 3 hr postexercise. Very high CHO diets employed by endurance athletes to increase CHO oxidation have little impact on iron regulation in elite athletes. It appears that variations in serum ferritin concentration and ambient temperature, rather than dietary CHO, are associated with increased hepcidin concentrations 3 hr postexercise.


Subject(s)
Dietary Carbohydrates/administration & dosage , Iron/blood , Physical Endurance/physiology , Sports/physiology , Walking/physiology , Adult , Dietary Carbohydrates/metabolism , Ferritins/blood , Hepcidins/blood , Humans , Interleukin-6/blood , Male , Oxidation-Reduction , Physical Conditioning, Human/physiology , Temperature
7.
Pediatr Hematol Oncol ; 38(2): 108-123, 2021 Mar.
Article in English | MEDLINE | ID: mdl-33026897

ABSTRACT

Chronic low-grade inflammation in type 1 diabetes (T1D) might increase hepcidin synthesis, possibly resulting in functional iron deficiency (FID). We hypothesized that in T1D children with FID, hepcidin concentrations are increased compared to those with normal iron status and those with absolute iron deficiency (AID). We evaluated hepcidin concentrations in T1D children in relation to iron status, and investigated whether hepcidin is useful in assessing FID. A cross-sectional study was conducted. FID was defined as elevated zinc protoporphyrin/heme ratio and/or red blood cell distribution width, and AID as low serum ferritin concentration. Post-hoc analyses with different definitions of FID were performed, using transferrin saturation and reticulocyte hemoglobin content. Serum hepcidin concentrations were measured using mass-spectrometry. The IRODIAB-study is registered at www.trialregister.nl (NTR4642). This study included 215 T1D children with a median age of 13.7 years (Q1-Q3: 10.1-16.3). The median (Q1-Q3) hepcidin concentration in patients with normal iron status was 1.8 nmol/l (0.9-3.3), in AID-patients, 0.4 nmol/l (0.4-0.4) and in FID-patients, 1.6 nmol/l (0.7-3.5). Hepcidin concentrations in FID-patients were significantly higher than in AID-patients (p < 0.001). Irrespective of FID-definition used, hepcidin concentrations did not differ between FID-patients and patients with normal iron status. This might be explained by the influence of various factors on hepcidin concentrations, and/or by differences in response of iron parameters over time. Single hepcidin measurements do not seem useful in assessing FID in T1D children. Multiple hepcidin measurements over time in future studies, however, might prove to be more useful in assessing FID in children with T1D.


Subject(s)
Anemia, Iron-Deficiency/blood , Anti-Infective Agents/blood , Diabetes Mellitus, Type 1/blood , Hepcidins/blood , Iron/blood , Adolescent , Cross-Sectional Studies , Female , Humans , Male
8.
Nat Rev Nephrol ; 16(2): 77-98, 2020 02.
Article in English | MEDLINE | ID: mdl-31554933

ABSTRACT

Iron is an essential element that is indispensable for life. The delicate physiological body iron balance is maintained by both systemic and cellular regulatory mechanisms. The iron-regulatory hormone hepcidin assures maintenance of adequate systemic iron levels and is regulated by circulating and stored iron levels, inflammation and erythropoiesis. The kidney has an important role in preventing iron loss from the body by means of reabsorption. Cellular iron levels are dependent on iron import, storage, utilization and export, which are mainly regulated by the iron response element-iron regulatory protein (IRE-IRP) system. In the kidney, iron transport mechanisms independent of the IRE-IRP system have been identified, suggesting additional mechanisms for iron handling in this organ. Yet, knowledge gaps on renal iron handling remain in terms of redundancy in transport mechanisms, the roles of the different tubular segments and related regulatory processes. Disturbances in cellular and systemic iron balance are recognized as causes and consequences of kidney injury. Consequently, iron metabolism has become a focus for novel therapeutic interventions for acute kidney injury and chronic kidney disease, which has fuelled interest in the molecular mechanisms of renal iron handling and renal injury, as well as the complex dynamics between systemic and local cellular iron regulation.


Subject(s)
Acute Kidney Injury/metabolism , Iron-Regulatory Proteins/metabolism , Iron/metabolism , Kidney/metabolism , Renal Insufficiency, Chronic/metabolism , Erythropoiesis , Erythropoietin/metabolism , Homeostasis , Humans , Inflammation/metabolism , Kidney Tubules, Distal/metabolism , Kidney Tubules, Proximal/metabolism , Mitochondria/metabolism , Nephrons/metabolism , Oxidative Stress
9.
Int J Sport Nutr Exerc Metab ; 30(1): 34-41, 2020 Jan 01.
Article in English | MEDLINE | ID: mdl-31629353

ABSTRACT

Sleeping with low carbohydrate (CHO) availability is a dietary strategy that may enhance training adaptation. However, the impact on an athlete's health is unclear. This study quantified the effect of a short-term "sleep-low" dietary intervention on markers of iron regulation and immune function in athletes. In a randomized, repeated-measures design, 11 elite triathletes completed two 4-day mixed cycle run training blocks. Key training sessions were structured such that a high-intensity training session was performed in the field on the afternoon of Days 1 and 3, and a low-intensity training (LIT) session was performed on the following morning in the laboratory (Days 2 and 4). The ingestion of CHO was either divided evenly across the day (HIGH) or restricted between the high-intensity training and LIT sessions, so that the LIT session was performed with low CHO availability (LOW). Venous blood and saliva samples were collected prior to and following each LIT session and analyzed for interleukin-6, hepcidin 25, and salivary immunoglobulin-A. Concentrations of interleukin-6 increased acutely after exercise (p < .001), but did not differ between dietary conditions or days. Hepcidin 25 increased 3-hr postexercise (p < .001), with the greatest increase evident after the LOW trial on Day 2 (2.5 ± 0.9 fold increase ±90% confidence limit). The salivary immunoglobulin-A secretion rate did not change in response to exercise; however, it was highest during the LOW condition on Day 4 (p = .046). There appears to be minimal impact to markers of immune function and iron regulation when acute exposure to low CHO availability is undertaken with expert nutrition and coaching input.


Subject(s)
Dietary Carbohydrates/administration & dosage , Interleukin-6/metabolism , Iron/metabolism , Physical Conditioning, Human/physiology , Sleep/physiology , Bicycling/physiology , Biomarkers/blood , Biomarkers/metabolism , Cross-Over Studies , Female , Hepcidins/blood , Hepcidins/metabolism , High-Intensity Interval Training , Humans , Immunoglobulin A, Secretory/metabolism , Interleukin-6/blood , Male , Physical Conditioning, Human/methods , Resistance Training , Running/physiology , Saliva/immunology , Saliva/metabolism , Swimming/physiology , Young Adult
10.
Pharmaceuticals (Basel) ; 12(3)2019 Aug 23.
Article in English | MEDLINE | ID: mdl-31450766

ABSTRACT

Peptide hormone hepcidin regulates systemic iron metabolism and has been described to be partially bound to α2-macroglobulin and albumin in blood. However, the reported degree of hepcidin protein binding varies between <3% and ≈89%. Since protein-binding may influence hormone function and quantification, better insight into the degree of hepcidin protein binding is essential to fully understand the biological behavior of hepcidin and interpretation of its measurement in patients. Here, we used peritoneal dialysis to assess human hepcidin protein binding in a functional human setting for the first time. We measured freely circulating solutes in blood and peritoneal fluid of 14 patients with end-stage renal disease undergoing a peritoneal equilibration test to establish a curve describing the relation between molecular weight and peritoneal clearance. Calculated binding percentages of total cortisol and testosterone confirmed our model. The protein-bound fraction of hepcidin was calculated to be 40% (±23%). We, therefore, conclude that a substantial proportion of hepcidin is freely circulating. Although a large inter-individual variation in hepcidin clearance, besides patient-specific peritoneal transport characteristics, may have affected the accuracy of the determined binding percentage, we describe an important step towards unraveling human hepcidin plasma protein binding in vivo including the caveats that need further research.

11.
PLoS One ; 14(3): e0212023, 2019.
Article in English | MEDLINE | ID: mdl-30917125

ABSTRACT

In chronic kidney disease both renal insufficiency and chronic inflammation trigger elevated hepcidin levels, which impairs iron uptake, availability. and erythropoiesis. Here we report the two first-in-human phase 1 trials of PRS-080#22, a novel, rationally engineered Anticalin protein that targets and antagonizes hepcidin. A single intravenous infusion of placebo or PRS-080#22 was administered to 48 healthy volunteers (phase 1a) and 24 patients with end stage chronic kidney disease (CKD) on hemodialysis (phase 1b) at different doses (0.08-16mg/kg for the phase 1a study and 2-8mg/kg for the phase 1b study) in successive dosing cohorts. The primary endpoint for both randomized, double-blind, phase 1 trials was safety and tolerability. Following treatment, all subjects were evaluable, with none experiencing dose limiting toxicities. Most adverse events were mild. One serious adverse event occurred in the phase 1b (CKD patient) study. There were no clinically significant changes in safety laboratory values or vital signs. PRS-080#22 showed dose-proportional pharmacokinetics (PK), with a terminal half-life of approximately three days in healthy volunteers and 10 to 12 days in CKD patients. Serum hepcidin levels were suppressed in a dose dependent manner and remained low for up to 48 hours after dosing. PRS-080#22 dose-dependently mobilized serum iron with increases in both serum iron concentration and transferrin saturation. No consistent changes were observed with regard to ferritin, reticulocytes, hemoglobin, and reticulocyte hemoglobin. Low titer anti-drug-antibodies were detected in five healthy volunteers but in none of the CKD patients. PRS-080#22, a novel Anticalin protein with picomolar affinity for hepcidin, was safe and well-tolerated when administered to healthy volunteers and CKD patients at all doses tested. The drug exhibited linear pharmacokinetics, longer half-life in CKD patients in comparison to healthy volunteers as well as expected pharmacodynamic effects which hold promise for further clinical studies.


Subject(s)
Hepcidins/antagonists & inhibitors , Lipocalins/pharmacology , Renal Insufficiency, Chronic/drug therapy , Adult , Double-Blind Method , Female , Half-Life , Healthy Volunteers , Humans , Infusions, Intravenous , Lipocalins/pharmacokinetics , Male , Middle Aged , Renal Dialysis/methods
12.
J Sci Med Sport ; 22(6): 635-640, 2019 Jun.
Article in English | MEDLINE | ID: mdl-30630742

ABSTRACT

OBJECTIVES: Adhering to a low carbohydrate (CHO) high fat (LCHF) diet can alter markers of iron metabolism in endurance athletes. This investigation examined the re-introduction of CHO prior to, and during exercise on the iron-regulatory response to exercise in a homogenous (in regard to serum ferritin concentration) group of athletes adapted to a LCHF diet. DESIGN: Parallel groups design. METHODS: Three weeks prior to the exercise trials, twenty-three elite race walkers adhered to either a CHO-rich (n=14) or LCHF diet (n=9). A standardised 19-25km race walk was performed while athletes were still adhering to their allocated dietary intervention (Adapt). A second test was performed three days later, where all athletes were placed on a high CHO diet (CHO Restoration). Venous blood samples were collected pre-, post- and 3h post-exercise and measured for interleukin-6 (IL-6) and hepcidin-25. RESULTS: The post-exercise IL-6 increase was greater in LCHF (p<0.001) during both the Adapt (LCHF: 13.1-fold increase; 95% CI: 5.6-23.0, CHO: 8.0-fold increase; 5.1-11.1) and CHO Restoration trials (LCHF: 18.5-fold increase; 10.9-28.9, CHO: 6.3-fold increase; 3.9-9.5); outcomes were not different between trials (p=0.84). Hepcidin-25 concentrations increased 3h post-exercise (p<0.001), however, they did not differ between trials (p=0.46) or diets (p=0.84). CONCLUSIONS: The elevated IL-6 response in athletes adapted to a LCHF diet was not attenuated by an acute increase in exogenous CHO availability. Despite diet-induced differences in IL-6 response to exercise, post-exercise hepcidin levels were similar between diets and trials, indicating CHO availability has minimal influence on post-exercise iron metabolism.


Subject(s)
Diet, Carbohydrate-Restricted , Dietary Carbohydrates/administration & dosage , Hepcidins/blood , Iron/metabolism , Sports Nutritional Physiological Phenomena , Walking/physiology , Adult , Athletes , Female , Humans , Interleukin-6/blood , Male , Young Adult
13.
Am J Physiol Renal Physiol ; 316(3): F606-F614, 2019 03 01.
Article in English | MEDLINE | ID: mdl-30623722

ABSTRACT

In physiological conditions, circulating iron can be filtered by the glomerulus and is almost completely reabsorbed by the tubular epithelium to prevent urinary iron wasting. Increased urinary iron concentrations have been associated with renal injury. However, it is not clear whether increased urinary iron concentrations in patients are the result of increased glomerular iron filtration and/or insufficient tubular iron reabsorption and if these processes contribute to renal injury. We measured plasma and urine iron parameters and urinary tubular injury markers in healthy human subjects ( n = 20), patients with systemic iron overload ( n = 20), and patients with renal tubular dysfunction ( n = 18). Urinary iron excretion parameters were increased in both patients with systemic iron overload and tubular dysfunction, whereas plasma iron parameters were only increased in patients with systemic iron overload. In patients with systemic iron overload, increased urinary iron levels were associated with elevated circulating iron, as indicated by transferrin saturation (TSAT), and increased body iron, as suggested by plasma ferritin concentrations. In patients with tubular dysfunction, enhanced urinary iron and transferrin excretion were associated with distal tubular injury as indicated by increased urinary glutathione S-transferase pi 1-1 (GSTP1-1) excretion. In systemic iron overload, elevated urinary iron and transferrin levels were associated with increased injury to proximal tubules, indicated by increased urinary kidney injury marker 1 (KIM-1) excretion. Our explorative study demonstrates that both glomerular filtration of elevated plasma iron levels and insufficient tubular iron reabsorption could increase urinary iron excretion and cause renal injury.


Subject(s)
Glomerular Filtration Rate/physiology , Iron Overload/metabolism , Iron/urine , Kidney/metabolism , Adult , Female , Humans , Iron Overload/urine , Kidney/physiopathology , Kidney Tubules, Proximal/metabolism , Kidney Tubules, Proximal/physiopathology , Male
14.
Clin Chem Lab Med ; 57(6): 864-872, 2019 05 27.
Article in English | MEDLINE | ID: mdl-30485171

ABSTRACT

Background Hepcidin concentrations measured by various methods differ considerably, complicating interpretation. Here, a previously identified plasma-based candidate secondary reference material (csRM) was modified into a serum-based two-leveled sRM. We validated its functionality to increase the equivalence between methods for international standardization. Methods We applied technical procedures developed by the International Consortium for Harmonization of Clinical Laboratory Results. The sRM, consisting of lyophilized serum with cryolyoprotectant, appeared commutable among nine different measurement procedures using 16 native human serum samples in a first round robin (RR1). Harmonization potential of the sRM was simulated in RR1 and evaluated in practice in RR2 among 11 measurement procedures using three native human plasma samples. Comprehensive purity analysis of a candidate primary RM (cpRM) was performed by state of the art procedures. The sRM was value assigned with an isotope dilution mass spectrometry-based candidate reference method calibrated using the certified pRM. Results The inter-assay CV without harmonization was 42.1% and 52.8% in RR1 and RR2, respectively. In RR1, simulation of harmonization with sRM resulted in an inter-assay CV of 11.0%, whereas in RR2 calibration with the material resulted in an inter-assay CV of 19.1%. Both the sRM and pRM passed international homogeneity criteria and showed long-term stability. We assigned values to the low (0.95±0.11 nmol/L) and middle concentration (3.75±0.17 nmol/L) calibrators of the sRM. Conclusions Standardization of hepcidin is possible with our sRM, which value is assigned by a pRM. We propose the implementation of this material as an international calibrator for hepcidin.


Subject(s)
Enzyme-Linked Immunosorbent Assay/methods , Hepcidins/blood , Tandem Mass Spectrometry , Calibration , Chromatography, High Pressure Liquid/standards , Enzyme-Linked Immunosorbent Assay/standards , Hepcidins/standards , Humans , Isotope Labeling , Reference Standards , Tandem Mass Spectrometry/standards
15.
Sci Rep ; 8(1): 13390, 2018 Sep 03.
Article in English | MEDLINE | ID: mdl-30177718

ABSTRACT

A correction to this article has been published and is linked from the HTML and PDF versions of this paper. The error has been fixed in the paper.

16.
Sci Rep ; 8(1): 9353, 2018 06 19.
Article in English | MEDLINE | ID: mdl-29921869

ABSTRACT

Iron is suggested to play a detrimental role in the progression of chronic kidney disease (CKD). The kidney recycles iron back into the circulation. However, the localization of proteins relevant for physiological tubular iron handling and their potential role in CKD remain unclear. We examined associations between iron deposition, expression of iron handling proteins and tubular injury in kidney biopsies from CKD patients and healthy controls using immunohistochemistry. Iron was deposited in proximal (PT) and distal tubules (DT) in 33% of CKD biopsies, predominantly in pathologies with glomerular dysfunction, but absent in controls. In healthy kidney, PT contained proteins required for iron recycling including putative iron importers ZIP8, ZIP14, DMT1, iron storage proteins L- and H-ferritin and iron exporter ferroportin, while DT only contained ZIP8, ZIP14, and DMT1. In CKD, iron deposition associated with increased intensity of iron importers (ZIP14, ZIP8), storage proteins (L-, H-ferritin), and/or decreased ferroportin abundance. This demonstrates that tubular iron accumulation may result from increased iron uptake and/or inadequate iron export. Iron deposition associated with oxidative injury as indicated by heme oxygenase-1 abundance. In conclusion, iron deposition is relatively common in CKD, and may result from altered molecular iron handling and may contribute to renal injury.


Subject(s)
Iron/metabolism , Kidney/metabolism , Renal Insufficiency, Chronic/metabolism , Apoferritins/metabolism , Biopsy , Cation Transport Proteins/metabolism , Female , Heme Oxygenase-1/metabolism , Humans , Immunohistochemistry , Male , Prevalence
17.
Cell Death Dis ; 9(5): 550, 2018 05 01.
Article in English | MEDLINE | ID: mdl-29749404

ABSTRACT

Hemoglobinuria is associated with kidney injury in various hemolytic pathologies. Currently, there is no treatment available and its pathophysiology is not completely understood. Here we studied the potential detrimental effects of hemoglobin (Hb) exposure to the distal nephron (DN). Involvement of the DN in Hb kidney injury was suggested by the induction of renal hepcidin synthesis (p < 0.001) in mice repeatedly injected with intravenous Hb. Moreover, the hepcidin induction was associated with a decline in urinary kidney injury markers 24p3/NGAL and KIM1, suggesting a role for hepcidin in protection against Hb kidney injury. We demonstrated that uptake of Hb in the mouse cortical collecting duct cells (mCCDcl1) is mediated by multi-protein ligand receptor 24p3R, as indicated by a significant 90% reduction in Hb uptake (p < 0.001) after 24p3R silencing. Moreover, incubation of mCCDcl1 cells with Hb or hemin for 4 or 24 h resulted in hepcidin synthesis and increased mRNA expression of markers for oxidative, inflammatory and ER stress, but no cell death as indicated by apoptosis staining. A protective role for cellular hepcidin against Hb-induced injury was demonstrated by aggravation of oxidative, inflammatory and ER stress after 4 h Hb or hemin incubation in hepcidin silenced mCCDcl1 cells. Hepcidin silencing potentiated hemin-mediated cell death that could be diminished by co-incubation of Nec-1, suggesting that endogenous hepcidin prevents necroptosis. Combined, these results demonstrate that renal hepcidin synthesis protects the DN against hemin and hemoglobin-mediated injury.


Subject(s)
Hemin/metabolism , Hemoglobins/metabolism , Hemoglobinuria/metabolism , Hepcidins/biosynthesis , Kidney Diseases/metabolism , Kidney Tubules, Distal/metabolism , Animals , Hemoglobinuria/pathology , Kidney Diseases/pathology , Kidney Tubules, Distal/pathology , Male , Mice , Necrosis
18.
Br J Pharmacol ; 175(7): 1054-1065, 2018 04.
Article in English | MEDLINE | ID: mdl-29329501

ABSTRACT

BACKGROUND AND PURPOSE: Anaemia of chronic disease (ACD) has been linked to iron-restricted erythropoiesis imposed by high circulating levels of hepcidin, a 25 amino acid hepatocyte-derived peptide that controls systemic iron homeostasis. Here, we report the engineering of the human lipocalin-derived, small protein-based anticalin PRS-080 hepcidin antagonist with high affinity and selectivity. EXPERIMENTAL APPROACH: Anticalin- and hepcidin-specific pharmacokinetic (PK)/pharmacodynamic modelling (PD) was used to design and select the suitable drug candidate based on t1/2 extension and duration of hepcidin suppression. The development of a novel free hepcidin assay enabled accurate analysis of bioactive hepcidin suppression and elucidation of the observed plasma iron levels after PRS-080-PEG30 administration in vivo. KEY RESULTS: PRS-080 had a hepcidin-binding affinity of 0.07 nM and, after coupling to 30 kD PEG (PRS-080-PEG30), a t1/2 of 43 h in cynomolgus monkeys. Dose-dependent iron mobilization and hepcidin suppression were observed after a single i.v. dose of PRS-080-PEG30 in cynomolgus monkeys. Importantly, in these animals, suppression of free hepcidin and subsequent plasma iron elevation were sustained during repeated s.c. dosing. After repeated dosing and followed by a treatment-free interval, all iron parameters returned to pre-dose values. CONCLUSIONS AND IMPLICATIONS: In conclusion, we developed a dose-dependent and safe approach for the direct suppression of hepcidin, resulting in prolonged iron mobilization to alleviate iron-restricted erythropoiesis that can address the root cause of ACD. PRS-080-PEG30 is currently in early clinical development.


Subject(s)
Hepcidins/antagonists & inhibitors , Hepcidins/blood , Iron/blood , Animals , Female , Macaca fascicularis , Male , Models, Biological
19.
PLoS One ; 12(7): e0181117, 2017.
Article in English | MEDLINE | ID: mdl-28704474

ABSTRACT

Heme is an efficient source of iron in the diet, and heme preparations are used to prevent and cure iron deficiency anemia in humans and animals. However, the molecular mechanisms responsible for heme absorption remain only partially characterized. Here, we employed young iron-deficient piglets as a convenient animal model to determine the efficacy of oral heme iron supplementation and investigate the pathways of heme iron absorption. The use of bovine hemoglobin as a dietary source of heme iron was found to efficiently counteract the development of iron deficiency anemia in piglets, although it did not fully rebalance their iron status. Our results revealed a concerted increase in the expression of genes responsible for apical and basolateral heme transport in the duodenum of piglets fed a heme-enriched diet. In these animals the catalytic activity of heme oxygenase 1 contributed to the release of elemental iron from the protoporphyrin ring of heme within enterocytes, which may then be transported by the strongly expressed ferroportin across the basolateral membrane to the circulation. We hypothesize that the well-recognized high bioavailability of heme iron may depend on a split pathway mediating the transport of heme-derived elemental iron and intact heme from the interior of duodenal enterocytes to the bloodstream.


Subject(s)
Anemia, Iron-Deficiency/diet therapy , Duodenum/metabolism , Gene Expression Profiling/methods , Heme Oxygenase-1/genetics , Heme/administration & dosage , Administration, Oral , Anemia, Iron-Deficiency/genetics , Anemia, Iron-Deficiency/metabolism , Animals , Disease Models, Animal , Gene Expression Regulation/drug effects , Gene Regulatory Networks/drug effects , Heme/therapeutic use , Heme Oxygenase-1/chemistry , Humans , Swine
20.
Am J Nephrol ; 44(5): 368-378, 2016.
Article in English | MEDLINE | ID: mdl-27771699

ABSTRACT

BACKGROUND: Hepcidin regulates systemic iron homeostasis by downregulating the iron exporter ferroportin. Circulating hepcidin is mainly derived from the liver but hepcidin is also produced in the heart. We studied the differential and local regulation of hepcidin gene expression in response to myocardial infarction (MI) and/or chronic kidney disease (CKD). We hypothesized that cardiac hepcidin gene expression is induced by and regulated to severity of cardiac injury, either through direct (MI) or remote (CKD) stimuli, as well as through increased local iron content. METHODS: Nine weeks after subtotal nephrectomy (SNX) or sham surgery (CON), rats were subjected to coronary ligation (CL) or sham surgery to realize 4 groups: CON, SNX, CL and SNX + CL. In week 16, the gene expression of hepcidin, iron and damage markers in cardiac and liver tissues was assessed by quantitative polymerase chain reaction and ferritin protein expression was studied by immunohistochemistry. RESULTS: Cardiac hepcidin messenger RNA (mRNA) expression was increased 2-fold in CL (p = 0.03) and 3-fold in SNX (p = 0.01). Cardiac ferritin staining was not different among groups. Cardiac hepcidin mRNA expression correlated with mRNA expression levels of brain natriuretic peptide (ß = 0.734, p < 0.001) and connective tissue growth factor (ß = 0.431, p = 0.02). In contrast, liver hepcidin expression was unaffected by SNX and CL alone, while it had decreased 50% in SNX + CL (p < 0.05). Hepatic ferritin immunostaining was not different among groups. CONCLUSIONS: Our data indicate differences in hepcidin regulation in liver and heart and suggest a role for injury rather than iron as the driving force for cardiac hepcidin expression in renocardiac failure.


Subject(s)
Hepcidins/metabolism , Iron/metabolism , Liver/metabolism , Myocardial Infarction/metabolism , Myocardium/metabolism , Renal Insufficiency, Chronic/metabolism , Animals , Bone Morphogenetic Protein 6/metabolism , CCAAT-Enhancer-Binding Protein-alpha/metabolism , Cation Transport Proteins/metabolism , Connective Tissue Growth Factor/metabolism , Cytokines/metabolism , Gene Expression Regulation , Heme Oxygenase (Decyclizing)/metabolism , Male , Natriuretic Peptide, Brain/metabolism , Rats, Inbred Lew
SELECTION OF CITATIONS
SEARCH DETAIL
...