1
|
Ryan BJ, Barney DE, McNiff JL, Drummer DJ, Howard EE, Gwin JA, Carrigan CT, Murphy NE, Wilson MA, Pasiakos SM, McClung JP, Margolis LM. Strenuous training combined with erythropoietin induces red cell volume expansion-mediated hypervolemia and alters systemic and skeletal muscle iron homeostasis. Am J Physiol Regul Integr Comp Physiol 2024; 327:R473-R478. [PMID: 39241004 PMCID: PMC11563636 DOI: 10.1152/ajpregu.00164.2024] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2024] [Revised: 08/02/2024] [Accepted: 08/13/2024] [Indexed: 09/08/2024]
Abstract
Strenuous physical training increases total blood volume (BV) through expansion of plasma volume (PV) and red cell volume (RCV). In contrast, exogenous erythropoietin (EPO) treatment increases RCV but decreases PV, rendering BV stable or slightly decreased. This study aimed to determine the combined effects of strenuous training and EPO treatment on BV and markers of systemic and muscle iron homeostasis. In this longitudinal study, eight healthy nonanemic males were treated with EPO (50 IU/kg body mass, three times per week, sc) across 28 days of strenuous training (4 days/wk, exercise energy expenditures of 1,334 ± 24 kcal/day) while consuming a controlled, energy-balanced diet providing 39 ± 4 mg/day iron. Before (PRE) and after (POST) intervention, BV compartments were measured using carbon monoxide rebreathing, and markers of iron homeostasis were assessed in blood and skeletal muscle (vastus lateralis). Training + EPO increased (P < 0.01) RCV (13 ± 6%) and BV (5 ± 4%), whereas PV remained unchanged (P = 0.86). The expansion of RCV was accompanied by a large decrease in whole body iron stores, as indicated by decreased (P < 0.01) ferritin (-77 ± 10%) and hepcidin (-49 ± 23%) concentrations in plasma. Training + EPO decreased (P < 0.01) muscle protein abundance of ferritin (-25 ± 20%) and increased (P < 0.05) transferrin receptor (47 ± 56%). These novel findings illustrate that strenuous training combined with EPO results in both increased total oxygen-carrying capacity and hypervolemia in young healthy males. The decrease in plasma and muscle ferritin suggests that the marked upregulation of erythropoiesis alters systemic and tissue iron homeostasis, resulting in a decline in whole body and skeletal muscle iron stores.NEW & NOTEWORTHY Strenuous exercise training combined with erythropoietin (EPO) treatment increases blood volume, driven exclusively by red cell volume expansion. This hematological adaptation results in increased total oxygen-carrying capacity and hypervolemia. The marked upregulation of erythropoiesis with training + EPO reduces whole body iron stores and circulating hepcidin concentrations. The finding that the abundance of ferritin in muscle decreased after training + EPO suggests that muscle may release iron to support red blood cell production.
Collapse
Affiliation(s)
- Benjamin J Ryan
- Thermal and Mountain Medicine Division, United States Army Research Institute of Environmental Medicine, Natick, Massachusetts, United States
| | - David E Barney
- Military Nutrition Division, United States Army Research Institute of Environmental Medicine, Natick, Massachusetts, United States
- Oak Ridge Institute for Science and Education, Belcamp, Maryland, United States
| | - Julie L McNiff
- Military Nutrition Division, United States Army Research Institute of Environmental Medicine, Natick, Massachusetts, United States
- Combat Feeding Division, United States Army Combat Capabilities Development Command (DEVCOM), Natick, Massachusetts, United States
| | - Devin J Drummer
- Military Nutrition Division, United States Army Research Institute of Environmental Medicine, Natick, Massachusetts, United States
- Oak Ridge Institute for Science and Education, Belcamp, Maryland, United States
| | - Emily E Howard
- Military Nutrition Division, United States Army Research Institute of Environmental Medicine, Natick, Massachusetts, United States
| | - Jess A Gwin
- Military Nutrition Division, United States Army Research Institute of Environmental Medicine, Natick, Massachusetts, United States
| | - Christopher T Carrigan
- Military Nutrition Division, United States Army Research Institute of Environmental Medicine, Natick, Massachusetts, United States
| | - Nancy E Murphy
- Military Nutrition Division, United States Army Research Institute of Environmental Medicine, Natick, Massachusetts, United States
| | - Marques A Wilson
- Military Nutrition Division, United States Army Research Institute of Environmental Medicine, Natick, Massachusetts, United States
| | - Stefan M Pasiakos
- Office of Dietary Supplements, United States Department of Health and Human Services, National Institutes of Health, Bethesda, Maryland, United States
| | - James P McClung
- Military Nutrition Division, United States Army Research Institute of Environmental Medicine, Natick, Massachusetts, United States
| | - Lee M Margolis
- Military Nutrition Division, United States Army Research Institute of Environmental Medicine, Natick, Massachusetts, United States
| |
Collapse
|
2
|
Packer M, Anker SD, Butler J, Cleland JGF, Kalra PR, Mentz RJ, Ponikowski P. Identification of three mechanistic pathways for iron-deficient heart failure. Eur Heart J 2024; 45:2281-2293. [PMID: 38733250 PMCID: PMC11231948 DOI: 10.1093/eurheartj/ehae284] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/13/2023] [Revised: 02/29/2024] [Accepted: 04/23/2024] [Indexed: 05/13/2024] Open
Abstract
Current understanding of iron-deficient heart failure is based on blood tests that are thought to reflect systemic iron stores, but the available evidence suggests greater complexity. The entry and egress of circulating iron is controlled by erythroblasts, which (in severe iron deficiency) will sacrifice erythropoiesis to supply iron to other organs, e.g. the heart. Marked hypoferraemia (typically with anaemia) can drive the depletion of cardiomyocyte iron, impairing contractile performance and explaining why a transferrin saturation < ≈15%-16% predicts the ability of intravenous iron to reduce the risk of major heart failure events in long-term trials (Type 1 iron-deficient heart failure). However, heart failure may be accompanied by intracellular iron depletion within skeletal muscle and cardiomyocytes, which is disproportionate to the findings of systemic iron biomarkers. Inflammation- and deconditioning-mediated skeletal muscle dysfunction-a primary cause of dyspnoea and exercise intolerance in patients with heart failure-is accompanied by intracellular skeletal myocyte iron depletion, which can be exacerbated by even mild hypoferraemia, explaining why symptoms and functional capacity improve following intravenous iron, regardless of baseline haemoglobin or changes in haemoglobin (Type 2 iron-deficient heart failure). Additionally, patients with advanced heart failure show myocardial iron depletion due to both diminished entry into and enhanced egress of iron from the myocardium; the changes in iron proteins in the cardiomyocytes of these patients are opposite to those expected from systemic iron deficiency. Nevertheless, iron supplementation can prevent ventricular remodelling and cardiomyopathy produced by experimental injury in the absence of systemic iron deficiency (Type 3 iron-deficient heart failure). These observations, taken collectively, support the possibility of three different mechanistic pathways for the development of iron-deficient heart failure: one that is driven through systemic iron depletion and impaired erythropoiesis and two that are characterized by disproportionate depletion of intracellular iron in skeletal and cardiac muscle. These mechanisms are not mutually exclusive, and all pathways may be operative at the same time or may occur sequentially in the same patients.
Collapse
Affiliation(s)
- Milton Packer
- Baylor Heart and Vascular Institute, Baylor University Medical Center, 621 North Hall Street, Dallas, TX 75226, USA
- Imperial College, London, UK
| | - Stefan D Anker
- Department of Cardiology of German Heart Center Charité, Institute of Health Center for Regenerative Therapies, German Centre for Cardiovascular Research, partner site Berlin, Charité Universitätsmedizin, Berlin, Germany
| | - Javed Butler
- Baylor Scott and White Research Institute, Baylor University Medical Center, Dallas, TX, USA
- University of Mississippi Medical Center, Jackson, MS, USA
| | - John G F Cleland
- British Heart Foundation Centre of Research Excellence, School of Cardiovascular and Metabolic Health, University of Glasgow, Glasgow, UK
| | - Paul R Kalra
- Department of Cardiology, Portsmouth Hospitals University NHS Trust, Portsmouth, UK
- College of Medical, Veterinary & Life Sciences, University of Glasgow, Glasgow, UK
- Faculty of Science and Health, University of Portsmouth, Portsmouth, UK
| | - Robert J Mentz
- Division of Cardiology, Department of Medicine, Duke University School of Medicine, Durham, NC, USA
- Duke Clinical Research Institute, Durham, NC, USA
| | - Piotr Ponikowski
- Institute of Heart Diseases, Wroclaw Medical University, Wroclaw, Poland
- Institute of Heart Diseases, University Hospital, Wroclaw, Poland
| |
Collapse
|
3
|
Drummer DJ, McNiff JL, Howard EE, Gwin JA, Carrigan CT, Murphy NE, Wilson MA, Michalak J, Ryan BJ, McClung JP, Pasiakos SM, Margolis LM. Exogenous erythropoietin increases hematological status, fat oxidation, and aerobic performance in males following prolonged strenuous training. Physiol Rep 2024; 12:e16038. [PMID: 38757249 PMCID: PMC11099744 DOI: 10.14814/phy2.16038] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2024] [Revised: 04/18/2024] [Accepted: 04/18/2024] [Indexed: 05/18/2024] Open
Abstract
This study investigated the effects of EPO on hemoglobin (Hgb) and hematocrit (Hct), time trial (TT) performance, substrate oxidation, and skeletal muscle phenotype throughout 28 days of strenuous exercise. Eight males completed this longitudinal controlled exercise and feeding study using EPO (50 IU/kg body mass) 3×/week for 28 days. Hgb, Hct, and TT performance were assessed PRE and on Days 7, 14, 21, and 27 of EPO. Rested/fasted muscle obtained PRE and POST EPO were analyzed for gene expression, protein signaling, fiber type, and capillarization. Substrate oxidation and glucose turnover were assessed during 90-min of treadmill load carriage (LC; 30% body mass; 55 ± 5% V̇O2peak) exercise using indirect calorimetry, and 6-6-[2H2]-glucose PRE and POST. Hgb and Hct increased, and TT performance improved on Days 21 and 27 compared to PRE (p < 0.05). Energy expenditure, fat oxidation, and metabolic clearance rate during LC increased (p < 0.05) from PRE to POST. Myofiber type, protein markers of mitochondrial biogenesis, and capillarization were unchanged PRE to POST. Transcriptional regulation of mitochondrial activity and fat metabolism increased from PRE to POST (p < 0.05). These data indicate EPO administration during 28 days of strenuous exercise can enhance aerobic performance through improved oxygen carrying capacity, whole-body and skeletal muscle fat metabolism.
Collapse
Affiliation(s)
- Devin J. Drummer
- Military Nutrition DivisionU.S. Army Research Institute of Environmental MedicineNatickMassachusettsUSA
- Oak Ridge Institute for Science and EducationBelcampMarylandUSA
| | - Julie L. McNiff
- Military Nutrition DivisionU.S. Army Research Institute of Environmental MedicineNatickMassachusettsUSA
- Combat Feeding DivisionU.S. Army Combat Capabilities Development Command (DEVCOM)NatickMassachusettsUSA
| | - Emily E. Howard
- Military Nutrition DivisionU.S. Army Research Institute of Environmental MedicineNatickMassachusettsUSA
| | - Jess A. Gwin
- Military Nutrition DivisionU.S. Army Research Institute of Environmental MedicineNatickMassachusettsUSA
| | - Christopher T. Carrigan
- Military Nutrition DivisionU.S. Army Research Institute of Environmental MedicineNatickMassachusettsUSA
| | - Nancy E. Murphy
- Military Nutrition DivisionU.S. Army Research Institute of Environmental MedicineNatickMassachusettsUSA
| | - Marques A. Wilson
- Military Nutrition DivisionU.S. Army Research Institute of Environmental MedicineNatickMassachusettsUSA
| | - Julia Michalak
- Military Nutrition DivisionU.S. Army Research Institute of Environmental MedicineNatickMassachusettsUSA
- Oak Ridge Institute for Science and EducationBelcampMarylandUSA
| | - Benjamin J. Ryan
- Thermal and Mountain Medicine DivisionU.S. Army Research Institute of Environmental MedicineNatickMassachusettsUSA
| | - James P. McClung
- Military Nutrition DivisionU.S. Army Research Institute of Environmental MedicineNatickMassachusettsUSA
| | - Stefan M. Pasiakos
- Office of Dietary Supplements, National Institutes of HealthU.S. Department of Health and Human ServicesBethesdaMarylandUSA
| | - Lee M. Margolis
- Military Nutrition DivisionU.S. Army Research Institute of Environmental MedicineNatickMassachusettsUSA
| |
Collapse
|
4
|
Breenfeldt Andersen A, Nordsborg NB, Bonne TC, Bejder J. Contemporary blood doping-Performance, mechanism, and detection. Scand J Med Sci Sports 2024; 34:e14243. [PMID: 36229224 DOI: 10.1111/sms.14243] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2022] [Revised: 10/04/2022] [Accepted: 10/09/2022] [Indexed: 10/17/2022]
Abstract
Blood doping is prohibited for athletes but has been a well-described practice within endurance sports throughout the years. With improved direct and indirect detection methods, the practice has allegedly moved towards micro-dosing, that is, reducing the blood doping regime amplitude. This narrative review evaluates whether blood doping, specifically recombinant human erythropoietin (rhEpo) treatment and blood transfusions are performance-enhancing, the responsible mechanism as well as detection possibilities with a special emphasis on micro-dosing. In general, studies evaluating micro-doses of blood doping are limited. However, in randomized, double-blinded, placebo-controlled trials, three studies find that infusing as little as 130 ml red blood cells or injecting 9 IU × kg bw-1 rhEpo three times per week for 4 weeks improve endurance performance ~4%-6%. The responsible mechanism for a performance-enhancing effect following rhEpo or blood transfusions appear to be increased O2 -carrying capacity, which is accompanied by an increased muscular O2 extraction and likely increased blood flow to the working muscles, enabling the ability to sustain a higher exercise intensity for a given period. Blood doping in micro-doses challenges indirect detection by the Athlete Biological Passport, albeit it can identify ~20%-60% of the individuals depending on the sample timing. However, novel biomarkers are emerging, and some may provide additive value for detection of micro blood doping such as the immature reticulocytes or the iron regulatory hormones hepcidin and erythroferrone. Future studies should attempt to validate these biomarkers for implementation in real-world anti-doping efforts and continue the biomarker discovery.
Collapse
Affiliation(s)
- Andreas Breenfeldt Andersen
- Department of Nutrition, Exercise and Sports, University of Copenhagen, Copenhagen, Denmark
- Department of Public Health, Section for Sport Science, Aarhus University, Aarhus, Denmark
| | | | - Thomas Christian Bonne
- Department of Nutrition, Exercise and Sports, University of Copenhagen, Copenhagen, Denmark
| | - Jacob Bejder
- Department of Nutrition, Exercise and Sports, University of Copenhagen, Copenhagen, Denmark
| |
Collapse
|
5
|
Martens K, DeLoughery TG. Sex, lies, and iron deficiency: a call to change ferritin reference ranges. HEMATOLOGY. AMERICAN SOCIETY OF HEMATOLOGY. EDUCATION PROGRAM 2023; 2023:617-621. [PMID: 38066931 PMCID: PMC10727104 DOI: 10.1182/hematology.2023000494] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/18/2023]
Abstract
Iron deficiency is a very common and treatable disorder. Of all the tests available to diagnose iron deficiency, the serum ferritin is the most able to discriminate iron deficiency from other disorders. However, the reference range for ferritin in many laboratories will lead to underdiagnosis of iron deficiency in women. Studies have shown that 30%-50% of healthy women will have no marrow iron stores, so basing ferritin cutoffs on the lowest 2.5% of sampled ferritins is not appropriate. In addition, several lines of evidence suggest the body physiologic ferritin "cutoff" is 50 ng/mL. Work is needed to establish more realistic ferritin ranges to avoid underdiagnosing a readily treatable disorder.
Collapse
Affiliation(s)
- Kylee Martens
- Knight Cancer Institute, Oregon Health & Science University, Portland, OR
| | | |
Collapse
|
6
|
Fighting age-related orthopedic diseases: focusing on ferroptosis. Bone Res 2023; 11:12. [PMID: 36854703 PMCID: PMC9975200 DOI: 10.1038/s41413-023-00247-y] [Citation(s) in RCA: 57] [Impact Index Per Article: 28.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2022] [Revised: 01/03/2023] [Accepted: 01/11/2023] [Indexed: 03/02/2023] Open
Abstract
Ferroptosis, a unique type of cell death, is characterized by iron-dependent accumulation and lipid peroxidation. It is closely related to multiple biological processes, including iron metabolism, polyunsaturated fatty acid metabolism, and the biosynthesis of compounds with antioxidant activities, including glutathione. In the past 10 years, increasing evidence has indicated a potentially strong relationship between ferroptosis and the onset and progression of age-related orthopedic diseases, such as osteoporosis and osteoarthritis. Therefore, in-depth knowledge of the regulatory mechanisms of ferroptosis in age-related orthopedic diseases may help improve disease treatment and prevention. This review provides an overview of recent research on ferroptosis and its influences on bone and cartilage homeostasis. It begins with a brief overview of systemic iron metabolism and ferroptosis, particularly the potential mechanisms of ferroptosis. It presents a discussion on the role of ferroptosis in age-related orthopedic diseases, including promotion of bone loss and cartilage degradation and the inhibition of osteogenesis. Finally, it focuses on the future of targeting ferroptosis to treat age-related orthopedic diseases with the intention of inspiring further clinical research and the development of therapeutic strategies.
Collapse
|
7
|
Song Z, Tang M, Tang G, Fu G, Ou D, Yao F, Hou X, Zhang D. Oral iron supplementation in patients with heart failure: a systematic review and meta-analysis. ESC Heart Fail 2022; 9:2779-2786. [PMID: 35758130 PMCID: PMC9715811 DOI: 10.1002/ehf2.14020] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2021] [Revised: 03/19/2022] [Accepted: 06/03/2022] [Indexed: 11/07/2022] Open
Abstract
AIMS This review aimed to assess whether oral iron supplementation in a chronic heart failure (HF) population with iron deficiency (ID) or mild anaemia is safe and effective according to evidence-based medicine. METHODS We retrieved 1803 records from the PubMed, Embase, and the Cochrane Library databases from 1 January 1991 to 15 September 2021. The clinical outcome of oral iron supplementation for ID anaemia in patients with HF was the primary endpoint. The primary safety measures included adverse events and all-cause mortality, and efficacy measures included transferrin saturation (Tsat), ferritin levels, and the 6-min walk test (6MWT). The rate ratio (RR) was used to pool the efficacy measures. RESULTS Five randomized controlled trials that compared oral iron treatment for patients with the placebo group and included a combined total of 590 participants were analysed. No significant difference was found in all-cause death between oral iron treatment and placebo groups (RR = 0.77; 95% confidence intervals (CI), 0.46-1.29, Z = 0.98; P = 0.33). However, adverse events were not significantly higher in the iron treatment group (RR = 0.83; 95% CI, 0.60-1.16, Z = 1.07; P = 0.28). In addition, ferritin levels and Tsat were slightly increased after iron complex administration in patients with HF but were not statistically significant (ferritin: mean difference [MD] = 2.70, 95% CI, -2.41 to 7.81, Z = 1.04; P = 0.30; Tsat: MD = 27.42, 95% CI, -4.93 to 59.78, Z = 1.66; P = 0.10). No significant difference was found in exercise capacity, as indicated by the 6MWT results (MD = 59.60, 95% CI, -17.89 to 137.08, Z = 1.51; P = 0.13). We also analysed two non-randomized controlled trials with follow-up results showing that oral iron supplementation increased serum iron levels (MD = 28.87, 95% CI, 1.62-56.12, Z = 2.08; P = 0.04). CONCLUSIONS Based on the current findings, oral iron supplementation can increase serum iron levels in patients with HF and ID or mild anaemia but does not improve Tsat and 6MWT. In addition, oral iron supplementation is relatively safe.
Collapse
Affiliation(s)
- Zhiping Song
- Department of Cardiovascular MedicineYuechi County People's HospitalGuang'anChina
| | - Mingyang Tang
- Geriatric Diseases Institute of Chengdu / Cancer Prevention and Treatment Institute of Chengdu, Department of CardiologyChengdu Fifth People’s Hospital (The Second Clinical Medical College, Affiliated Fifth People’s Hospital of Chengdu University of Traditional Chinese Medicine)ChengduChina
| | - Gang Tang
- Geriatric Diseases Institute of Chengdu / Cancer Prevention and Treatment Institute of Chengdu, Department of CardiologyChengdu Fifth People’s Hospital (The Second Clinical Medical College, Affiliated Fifth People’s Hospital of Chengdu University of Traditional Chinese Medicine)ChengduChina
| | - Guoqi Fu
- Geriatric Diseases Institute of Chengdu / Cancer Prevention and Treatment Institute of Chengdu, Department of CardiologyChengdu Fifth People’s Hospital (The Second Clinical Medical College, Affiliated Fifth People’s Hospital of Chengdu University of Traditional Chinese Medicine)ChengduChina
| | - Dengke Ou
- Geriatric Diseases Institute of Chengdu / Cancer Prevention and Treatment Institute of Chengdu, Department of CardiologyChengdu Fifth People’s Hospital (The Second Clinical Medical College, Affiliated Fifth People’s Hospital of Chengdu University of Traditional Chinese Medicine)ChengduChina
| | - Fengyou Yao
- Geriatric Diseases Institute of Chengdu / Cancer Prevention and Treatment Institute of Chengdu, Department of CardiologyChengdu Fifth People’s Hospital (The Second Clinical Medical College, Affiliated Fifth People’s Hospital of Chengdu University of Traditional Chinese Medicine)ChengduChina
| | - Xingzhi Hou
- Department of Cardiovascular MedicineYuechi County People's HospitalGuang'anChina
| | - Denghong Zhang
- Geriatric Diseases Institute of Chengdu / Cancer Prevention and Treatment Institute of Chengdu, Department of CardiologyChengdu Fifth People’s Hospital (The Second Clinical Medical College, Affiliated Fifth People’s Hospital of Chengdu University of Traditional Chinese Medicine)ChengduChina
| |
Collapse
|
8
|
Saugy JJ, Schmoutz T, Botrè F. Altitude and Erythropoietin: Comparative Evaluation of Their Impact on Key Parameters of the Athlete Biological Passport: A Review. Front Sports Act Living 2022; 4:864532. [PMID: 35847455 PMCID: PMC9282833 DOI: 10.3389/fspor.2022.864532] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2022] [Accepted: 05/30/2022] [Indexed: 11/13/2022] Open
Abstract
The hematological module of the Athlete's Biological Passport (ABP) identifies doping methods and/or substances used to increase the blood's capacity to transport or deliver oxygen to the tissues. Recombinant human erythropoietin (rhEPOs) are doping substances known to boost the production of red blood cells and might have an effect on the blood biomarkers of the ABP. However, hypoxic exposure influences these biomarkers similarly to rhEPOs. This analogous impact complicates the ABP profiles' interpretation by antidoping experts. The present study aimed to collect and identify, through a literature search, the physiological effects on ABP blood biomarkers induced by these external factors. A total of 43 studies were selected for this review. A positive correlation (R2 = 0.605, r = 0.778, p < 0.001) was identified between the hypoxic dose and the increase in hemoglobin concentration (HGB) percentage. In addition, the change in the reticulocyte percentage (RET%) has been identified as one of the most sensitive parameters to rhEPO use. The mean effects of rhEPO on blood parameters were greater than those induced by hypoxic exposure (1.7 times higher for HGB and RET% and 4 times higher for hemoglobin mass). However, rhEPO micro-doses have shown effects that are hardly distinguishable from those identified after hypoxic exposure. The results of the literature search allowed to identify temporal and quantitative evolution of blood parameters in connection with different hypoxic exposure doses, as well as different rhEPOs doses. This might be considered to provide justified and well-documented interpretations of physiological changes in blood parameters of the Athlete Biological Passport.
Collapse
Affiliation(s)
- Jonas J. Saugy
- Institute of Sport Sciences, University of Lausanne (ISSUL), Lausanne, Switzerland
- Research and Expertise in anti-Doping Sciences (REDs), University of Lausanne, Lausanne, Switzerland
- *Correspondence: Jonas J. Saugy
| | - Tania Schmoutz
- Institute of Sport Sciences, University of Lausanne (ISSUL), Lausanne, Switzerland
| | - Francesco Botrè
- Institute of Sport Sciences, University of Lausanne (ISSUL), Lausanne, Switzerland
- Research and Expertise in anti-Doping Sciences (REDs), University of Lausanne, Lausanne, Switzerland
| |
Collapse
|
9
|
Correnti M, Gammella E, Cairo G, Recalcati S. Iron Mining for Erythropoiesis. Int J Mol Sci 2022; 23:ijms23105341. [PMID: 35628152 PMCID: PMC9140467 DOI: 10.3390/ijms23105341] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2022] [Revised: 05/05/2022] [Accepted: 05/09/2022] [Indexed: 01/27/2023] Open
Abstract
Iron is necessary for essential processes in every cell of the body, but the erythropoietic compartment is a privileged iron consumer. In fact, as a necessary component of hemoglobin and myoglobin, iron assures oxygen distribution; therefore, a considerable amount of iron is required daily for hemoglobin synthesis and erythroid cell proliferation. Therefore, a tight link exists between iron metabolism and erythropoiesis. The liver-derived hormone hepcidin, which controls iron homeostasis via its interaction with the iron exporter ferroportin, coordinates erythropoietic activity and iron homeostasis. When erythropoiesis is enhanced, iron availability to the erythron is mainly ensured by inhibiting hepcidin expression, thereby increasing ferroportin-mediated iron export from both duodenal absorptive cells and reticuloendothelial cells that process old and/or damaged red blood cells. Erythroferrone, a factor produced and secreted by erythroid precursors in response to erythropoietin, has been identified and characterized as a suppressor of hepcidin synthesis to allow iron mobilization and facilitate erythropoiesis.
Collapse
|
10
|
Joharapurkar AA, Patel VJ, Kshirsagar SG, Patel MS, Savsani HH, Kajavadara C, Valani D, Jain MR. Prolyl hydroxylase inhibitor desidustat improves anemia in erythropoietin hyporesponsive state. CURRENT RESEARCH IN PHARMACOLOGY AND DRUG DISCOVERY 2022; 3:100102. [PMID: 35570856 PMCID: PMC9096675 DOI: 10.1016/j.crphar.2022.100102] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2021] [Revised: 04/14/2022] [Accepted: 04/21/2022] [Indexed: 11/24/2022] Open
Abstract
Many anemic chronic kidney disease (CKD) patients are refractory to erythropoietin (EPO) effects due to inflammation, deranged iron utilization, and generation of EPO antibodies. This work assessed the effect of desidustat, an inhibitor of hypoxia inducible factor (HIF) prolyl hydroxylase (PHD), on EPO-refractory renal anemia. Sprague Dawley rats were made anemic by cisplatin (5 mg/kg, IP, single dose) and turpentine oil (5 mL/kg, SC, once a week). These rats were given recombinant human EPO (rhEPO, 1 μg/kg) and desidustat (15 or 30 mg/kg) for eight weeks. Separately, rhEPO (1-5 μg/kg) was given to anemic rats to sustain the normal hemoglobin levels and desidustat (15 mg/kg) for eight weeks. In another experiment, the anemic rats were treated rhEPO (5 μg/kg) for two weeks and then desidustat (15 mg/kg) for the next two weeks. Dosing of rhEPO was thrice a week, and for desidustat, it was on alternate days. Desidustat inhibited EPO-resistance caused by rhEPO treatment, decreased hepcidin, IL-6, IL-1β, and increased iron and liver ferroportin. Desidustat reduced EPO requirement and anti-EPO antibodies. Desidustat also maintained normal hemoglobin levels after cessation of rhEPO treatment. Thus, novel prolyl hydroxylase inhibitor desidustat can treat EPO resistance via improved iron utilization and decreased inflammation.
Collapse
Affiliation(s)
- Amit A. Joharapurkar
- Department of Pharmacology and Toxicology, Zydus Research Centre, Cadila Healthcare Limited, Sarkhej Bavla NH 8A, Moraiya, Ahmedabad, Gujarat, India
| | - Vishal J. Patel
- Department of Pharmacology and Toxicology, Zydus Research Centre, Cadila Healthcare Limited, Sarkhej Bavla NH 8A, Moraiya, Ahmedabad, Gujarat, India
| | - Samadhan G. Kshirsagar
- Department of Pharmacology and Toxicology, Zydus Research Centre, Cadila Healthcare Limited, Sarkhej Bavla NH 8A, Moraiya, Ahmedabad, Gujarat, India
| | - Maulik S. Patel
- Department of Pharmacology and Toxicology, Zydus Research Centre, Cadila Healthcare Limited, Sarkhej Bavla NH 8A, Moraiya, Ahmedabad, Gujarat, India
| | - Hardikkumar H. Savsani
- Department of Pharmacology and Toxicology, Zydus Research Centre, Cadila Healthcare Limited, Sarkhej Bavla NH 8A, Moraiya, Ahmedabad, Gujarat, India
| | - Chetan Kajavadara
- Department of Pharmacology and Toxicology, Zydus Research Centre, Cadila Healthcare Limited, Sarkhej Bavla NH 8A, Moraiya, Ahmedabad, Gujarat, India
| | - Darshan Valani
- Department of Pharmacology and Toxicology, Zydus Research Centre, Cadila Healthcare Limited, Sarkhej Bavla NH 8A, Moraiya, Ahmedabad, Gujarat, India
| | - Mukul R. Jain
- Department of Pharmacology and Toxicology, Zydus Research Centre, Cadila Healthcare Limited, Sarkhej Bavla NH 8A, Moraiya, Ahmedabad, Gujarat, India
| |
Collapse
|
11
|
Breenfeldt Andersen A, Bonne TC, Bejder J, Jung G, Ganz T, Nemeth E, Olsen NV, Huertas JR, Nordsborg NB. Effects of altitude and recombinant human erythropoietin on iron metabolism: a randomized controlled trial. Am J Physiol Regul Integr Comp Physiol 2021; 321:R152-R161. [PMID: 34160288 DOI: 10.1152/ajpregu.00070.2021] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
Current markers of iron deficiency (ID), such as ferritin and hemoglobin, have shortcomings, and hepcidin and erythroferrone (ERFE) could be of clinical relevance in relation to early assessment of ID. Here, we evaluate whether exposure to altitude-induced hypoxia (2,320 m) alone, or in combination with recombinant human erythropoietin (rHuEPO) treatment, affects hepcidin and ERFE levels before alterations in routine ID biomarkers and stress erythropoiesis manifest. Two interventions were completed, each comprising a 4-wk baseline, a 4-wk intervention at either sea level or altitude, and a 4-wk follow-up. Participants (n = 39) were randomly assigned to 20 IU·kg body wt-1 rHuEPO or placebo injections every second day for 3 wk during the two intervention periods. Venous blood was collected weekly. Altitude increased ERFE (P ≤ 0.001) with no changes in hepcidin or routine iron biomarkers, making ERFE of clinical relevance as an early marker of moderate hypoxia. rHuEPO treatment at sea level induced a similar pattern of changes in ERFE (P < 0.05) and hepcidin levels (P < 0.05), demonstrating the impact of accelerated erythropoiesis and not of other hypoxia-induced mechanisms. Compared with altitude alone, concurrent rHuEPO treatment and altitude exposure induced additive changes in hepcidin (P < 0.05) and ERFE (P ≤ 0.001) parallel with increases in hematocrit (P < 0.001), demonstrating a relevant range of both hepcidin and ERFE. A poor but significant correlation between hepcidin and ERFE was found (R2 = 0.13, P < 0.001). The findings demonstrate that hepcidin and ERFE are more rapid biomarkers of changes in iron demands than routine iron markers. Finally, ERFE and hepcidin may be sensitive markers in an antidoping context.
Collapse
Affiliation(s)
| | - Thomas C Bonne
- Department of Nutrition, Exercise and Sports, University of Copenhagen, Copenhagen, Denmark
| | - Jacob Bejder
- Department of Nutrition, Exercise and Sports, University of Copenhagen, Copenhagen, Denmark
| | - Grace Jung
- Department of Medicine and Pathology, Center for Iron Disorders, David Geffen School of Medicine, University of California Los Angeles, Los Angeles, California
| | - Tomas Ganz
- Department of Medicine and Pathology, Center for Iron Disorders, David Geffen School of Medicine, University of California Los Angeles, Los Angeles, California
| | - Elizabeta Nemeth
- Department of Medicine and Pathology, Center for Iron Disorders, David Geffen School of Medicine, University of California Los Angeles, Los Angeles, California
| | - Niels Vidiendal Olsen
- Department of Neuroscience and Pharmacology, University of Copenhagen, Copenhagen, Denmark
| | - Jesús Rodríguez Huertas
- Department of Physiology, Faculty of Sport Sciences, Institute of Nutrition and Food Technology, Biomedical Research Centre, University of Granada, Armilla, Spain
| | | |
Collapse
|
12
|
Robach P, Gammella E, Recalcati S, Girelli D, Castagna A, Roustit M, Lundby C, Lundby AK, Bouzat P, Vergès S, Séchaud G, Banco P, Uhr M, Cornu C, Sallet P, Cairo G. Induction of erythroferrone in healthy humans by micro-dose recombinant erythropoietin or high-altitude exposure. Haematologica 2021; 106:384-390. [PMID: 31919080 PMCID: PMC7849588 DOI: 10.3324/haematol.2019.233874] [Citation(s) in RCA: 25] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2019] [Accepted: 01/02/2020] [Indexed: 01/22/2023] Open
Abstract
The erythropoietin (Epo)-erythroferrone (ERFE)-hepcidin axis coordinates erythropoiesis and iron homeostasis. While mouse studies have established that Epo-induced ERFE production represses hepcidin synthesis by inhibiting hepatic BMP/SMAD signaling, evidence for the role of ERFE in humans is limited. To investigate the role of ERFE as a physiological erythroid regulator in humans, we conducted two studies. First, 24 males were given six injections of saline (placebo), recombinant Epo (rhEpo) at a dose of 20 IU/kg (micro-dose) or rhEpo at 50 IU/kg (low dose). Second, we quantified ERFE in 22 subjects exposed to high altitude (3800 m) for 15 h. In the first study, total hemoglobin mass (Hbmass) increased after low- but not after micro-dose injections, when compared to the mass after placebo injections. Serum ERFE levels were enhanced by rhEpo, remaining higher than after placebo for 48 h (micro-dose) or 72 h (low-dose) after injections. Conversely, hepcidin levels decreased when Epo and ERFE rose, before any changes in serum iron parameters occurred. In the second study, serum Epo and ERFE increased at high altitude. The present results demonstrate that in healthy humans ERFE responds to slightly increased Epo levels not associated with Hbmass expansion and downregulates hepcidin in an apparently iron-independent way. Notably, ERFE flags micro-dose Epo, thus holding promise as a novel biomarker of doping.
Collapse
Affiliation(s)
- Paul Robach
- National School for Mountains Sports, Chamonix, France
| | - Elena Gammella
- Dept Biomedical Sciences for Health, University of Milan, Italy
| | | | | | | | | | - Carsten Lundby
- Center for Physical Activity Research, University Hospital, Copenhagen, Denmark
| | | | - Pierre Bouzat
- Grenoble Alpes University Hospital, Grenoble, France
| | - Samuel Vergès
- HP2 Laboratory, U1042, Grenoble Alpes University, INSERM, Grenoble, France
| | | | | | - Mario Uhr
- Dept. Hematology Synlab-Suisse, Lugano, Switzerland
| | - Catherine Cornu
- Hospices Civils de Lyon INSERM CIC1407/UMR5558, Hôpital Louis Pradel, Bron, France
| | | | - Gaetano Cairo
- Dept Biomedical Sciences for Health, University of Milan, Italy
| |
Collapse
|
13
|
Hayashi N, Yatsutani H, Mori H, Ito H, Badenhorst CE, Goto K. No effect of supplemented heat stress during an acute endurance exercise session in hypoxia on hepcidin regulation. Eur J Appl Physiol 2020; 120:1331-1340. [PMID: 32303828 DOI: 10.1007/s00421-020-04365-x] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2019] [Accepted: 04/04/2020] [Indexed: 12/21/2022]
Abstract
Hepcidin is a novel factor for iron deficiency in athletes, which is suggested to be regulated by interleukin-6 (IL-6) or erythropoietin (EPO). PURPOSE The purpose of the present study was to compare endurance exercise-induced hepcidin elevation among "normoxia", "hypoxia" and "combined heat and hypoxia". METHODS Twelve males (21.5 ± 0.3 years, 168.1 ± 1.2 cm, 63.6 ± 2.0 kg) participated in the present study. They performed 60 min of cycling at 60% of [Formula: see text] in either "heat and hypoxia" (HHYP; FiO2 14.5%, 32 °C), "hypoxia" (HYP; FiO2 14.5%, 23 °C) or "normoxia" (NOR; FiO2 20.9%, 23 °C). After completing the exercise, participants remained in the prescribed conditions for 3 h post-exercise. Blood samples were collected before, immediately and 3 h after exercise. RESULTS Plasma IL-6 level significantly increased immediately after exercise (P < 0.05), with no significant difference among the trials. A significant elevation in serum EPO was observed 3 h after exercise in hypoxic trials (HHYP and HYP, P < 0.05), with no significant difference between HHYP and HYP. Serum hepcidin level increased 3 h after exercise in all trials (NOR, before 18.3 ± 3.9 and post180 31.2 ± 6.3 ng/mL; HYP, before 13.5 ± 2.5 and post180 23.3 ± 3.6 ng/mL, HHYP; before 15.8 ± 3.3 and post180 31.4 ± 5.3 ng/mL, P < 0.05). However, there was no significant difference among the trials during post-exercise. CONCLUSION Endurance exercise in "combined heat and hypoxia" did not exacerbate exercise-induced hepcidin elevation compared with the same exercise in "hypoxia" or "normoxia".
Collapse
Affiliation(s)
- Nanako Hayashi
- Graduate School of Sport and Health Science, Ritsumeikan University, 1-1-1, Noji-higashi, Kusatsu, Shiga, 525-8577, Japan
| | - Haruka Yatsutani
- Graduate School of Sport and Health Science, Ritsumeikan University, 1-1-1, Noji-higashi, Kusatsu, Shiga, 525-8577, Japan
| | - Hisashi Mori
- School of Human Science and Environment, University of Hyogo, Himeji, Hyogo, Japan
| | - Hiroto Ito
- Graduate School of Sport and Health Science, Ritsumeikan University, 1-1-1, Noji-higashi, Kusatsu, Shiga, 525-8577, Japan
| | - Claire E Badenhorst
- School of Sport, Exercise and Nutrition, Massey University, Auckland, New Zealand
| | - Kazushige Goto
- Graduate School of Sport and Health Science, Ritsumeikan University, 1-1-1, Noji-higashi, Kusatsu, Shiga, 525-8577, Japan.
| |
Collapse
|
14
|
Moretti D, Mettler S, Zeder C, Lundby C, Geurts-Moetspot A, Monnard A, Swinkels DW, Brittenham GM, Zimmermann MB. An intensified training schedule in recreational male runners is associated with increases in erythropoiesis and inflammation and a net reduction in plasma hepcidin. Am J Clin Nutr 2018; 108:1324-1333. [PMID: 30351387 DOI: 10.1093/ajcn/nqy247] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2017] [Accepted: 08/21/2018] [Indexed: 01/24/2023] Open
Abstract
Background Iron status is a determinant of physical performance, but training may induce both low-grade inflammation and erythropoiesis, exerting opposing influences on hepcidin and iron metabolism. To our knowledge, the combined effects on iron absorption and utilization during training have not been examined directly in humans. Objective We hypothesized that 3 wk of exercise training in recreational male runners would decrease oral iron bioavailability by increasing inflammation and hepcidin concentrations. Design In a prospective intervention, nonanemic, iron-sufficient men (n = 10) completed a 34-d study consisting of a 16-d control phase and a 22-d exercise-training phase of 8 km running every second day. We measured oral iron absorption and erythroid iron utilization using oral 57Fe and intravenous 58Fe tracers administered before and during training. We measured hemoglobin mass (mHb) and total red blood cell volume (RCV) by carbon monoxide rebreathing. Iron status, interleukin-6 (IL-6), plasma hepcidin (PHep), erythropoietin (EPO), and erythroferrone were measured before, during, and after training. Results Exercise training induced inflammation, as indicated by an increased mean ± SD IL-6 (0.87 ± 1.1 to 5.17 ± 2.2 pg/mL; P < 0.01), while also enhancing erythropoiesis, as indicated by an increase in mean EPO (0.66 ± 0.42 to 2.06 ± 1.6 IU/L), mHb (10.5 ± 1.6 to 10.8 ± 1.8 g/kg body weight), and mean RCV (30.7 ± 4.3 to 32.7 ± 4.6 mL/kg) (all P < 0.05). Training tended to increase geometric mean iron absorption by 24% (P = 0.083), consistent with a decreased mean ± SD PHep (7.25 ± 2.14 to 5.17 ± 2.24 nM; P < 0.05). The increase in mHb and erythroid iron utilization were associated with the decrease in PHep (P < 0.05). Compartmental modeling indicated that iron for the increase in mHb was obtained predominantly (>80%) from stores mobilization rather than from increased dietary absorption. Conclusions In iron-sufficient men, mild intensification of exercise intensity increases both inflammation and erythropoiesis. The net effect is to decrease hepcidin concentrations and to tend to increase oral iron absorption. This trial was registered at clinicaltrials.gov as NCT01730521.
Collapse
Affiliation(s)
- Diego Moretti
- Laboratory of Human Nutrition, Institute of Food, Nutrition and Health, Swiss Federal Institute of Technology, ETH Zürich, Zurich, Switzerland
| | - Samuel Mettler
- Laboratory of Human Nutrition, Institute of Food, Nutrition and Health, Swiss Federal Institute of Technology, ETH Zürich, Zurich, Switzerland.,Swiss Federal Institute of Sports, Magglingen, Switzerland.,Department of Business, Health, and Social Work, Bern University of Applied Sciences, Bern, Switzerland
| | - Christophe Zeder
- Laboratory of Human Nutrition, Institute of Food, Nutrition and Health, Swiss Federal Institute of Technology, ETH Zürich, Zurich, Switzerland
| | - Carsten Lundby
- Zurich Center for Integrative Human Physiology, Institute of Physiology, University of Zurich, Zurich, Switzerland
| | - Anneke Geurts-Moetspot
- Hepcidinanalysis.com and Department of Laboratory Medicine, Translational Metabolic Laboratory, Radboud University Medical Center, Nijmegen, Netherlands
| | - Arnaud Monnard
- Laboratory of Human Nutrition, Institute of Food, Nutrition and Health, Swiss Federal Institute of Technology, ETH Zürich, Zurich, Switzerland
| | - Dorine W Swinkels
- Hepcidinanalysis.com and Department of Laboratory Medicine, Translational Metabolic Laboratory, Radboud University Medical Center, Nijmegen, Netherlands
| | - Gary M Brittenham
- Laboratory of Human Nutrition, Institute of Food, Nutrition and Health, Swiss Federal Institute of Technology, ETH Zürich, Zurich, Switzerland
| | - Michael B Zimmermann
- Laboratory of Human Nutrition, Institute of Food, Nutrition and Health, Swiss Federal Institute of Technology, ETH Zürich, Zurich, Switzerland
| |
Collapse
|
15
|
Pfeifhofer-Obermair C, Tymoszuk P, Petzer V, Weiss G, Nairz M. Iron in the Tumor Microenvironment-Connecting the Dots. Front Oncol 2018; 8:549. [PMID: 30534534 PMCID: PMC6275298 DOI: 10.3389/fonc.2018.00549] [Citation(s) in RCA: 100] [Impact Index Per Article: 14.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2018] [Accepted: 11/06/2018] [Indexed: 12/18/2022] Open
Abstract
Iron metabolism and tumor biology are intimately linked. Iron facilitates the production of oxygen radicals, which may either result in iron-induced cell death, ferroptosis, or contribute to mutagenicity and malignant transformation. Once transformed, malignant cells require high amounts of iron for proliferation. In addition, iron has multiple regulatory effects on the immune system, thus affecting tumor surveillance by immune cells. For these reasons, inconsiderate iron supplementation in cancer patients has the potential of worsening disease course and outcome. On the other hand, chronic immune activation in the setting of malignancy alters systemic iron homeostasis and directs iron fluxes into myeloid cells. While this response aims at withdrawing iron from tumor cells, it may impair the effector functions of tumor-associated macrophages and will result in iron-restricted erythropoiesis and the development of anemia, subsequently. This review summarizes our current knowledge of the interconnections of iron homeostasis with cancer biology, discusses current clinical controversies in the treatment of anemia of cancer and focuses on the potential roles of iron in the solid tumor microenvironment, also speculating on yet unknown molecular mechanisms.
Collapse
Affiliation(s)
- Christa Pfeifhofer-Obermair
- Department of Internal Medicine II, Infectious Diseases, Immunology, Rheumatology, Pneumology, Medical University of Innsbruck, Innsbruck, Austria
| | - Piotr Tymoszuk
- Department of Internal Medicine II, Infectious Diseases, Immunology, Rheumatology, Pneumology, Medical University of Innsbruck, Innsbruck, Austria
| | - Verena Petzer
- Department of Internal Medicine II, Infectious Diseases, Immunology, Rheumatology, Pneumology, Medical University of Innsbruck, Innsbruck, Austria
| | - Günter Weiss
- Department of Internal Medicine II, Infectious Diseases, Immunology, Rheumatology, Pneumology, Medical University of Innsbruck, Innsbruck, Austria.,Christian Doppler Laboratory for Iron Metabolism and Anemia Research, Medical University of Innsbruck, Innsbruck, Austria
| | - Manfred Nairz
- Department of Internal Medicine II, Infectious Diseases, Immunology, Rheumatology, Pneumology, Medical University of Innsbruck, Innsbruck, Austria
| |
Collapse
|
16
|
Płoszczyca K, Langfort J, Czuba M. The Effects of Altitude Training on Erythropoietic Response and Hematological Variables in Adult Athletes: A Narrative Review. Front Physiol 2018; 9:375. [PMID: 29695978 PMCID: PMC5904371 DOI: 10.3389/fphys.2018.00375] [Citation(s) in RCA: 60] [Impact Index Per Article: 8.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2017] [Accepted: 03/27/2018] [Indexed: 02/02/2023] Open
Abstract
Background: One of the goals of altitude training is to increase blood oxygen-carrying capacity in order to improve sea-level endurance performance in athletes. The elevated erythropoietin (EPO) production in hypoxia is a key factor in the achievement of enhanced hematological variables. The level of the EPO increase and acceleration of erythropoiesis depend on the duration of exposure and degree of hypoxia. Furthermore, many other factors may affect the hematological response to altitude training. Aim: The purpose of this narrative review was to: (1) analyze the kinetics of EPO and hematological variables during and after altitude training; (2) summarize the current state of knowledge about the possible causes of individual or cohort differences in EPO and hematological response to altitude training; (3) formulate practical guidelines for athletes to improve the efficiency of altitude training. Methods: A narrative review was performed following an electronic search of the databases PubMed/MEDLINE and SPORTDiscus via EBSCO for all English-language articles published between 1997 and 2017. Results: Complete unification of results from studies on EPO kinetics was difficult due to different time and frequency of blood sampling by different researchers during and after altitude training, but the data presented in the reviewed literature allowed us to detect certain trends. The results of the reviewed studies were divergent and indicated either increase or no change of hematological variables following altitude training. Factors that may affect the hematological response to altitude training include hypoxic dose, training content, training background of athletes, and/or individual variability of EPO production. Conclusions: Despite the potential benefits arising from altitude training, its effectiveness in improving hematological variables is still debatable. Further research and better understanding of factors influencing the response to altitude, as well as factors affecting the suitable measurement and interpretation of study results, are needed.
Collapse
Affiliation(s)
- Kamila Płoszczyca
- Department of Sports Training, Academy of Physical Education of Katowice, Katowice, Poland
| | - Józef Langfort
- Department of Sports Training, Academy of Physical Education of Katowice, Katowice, Poland
| | - Miłosz Czuba
- Department of Physiology, Institute of Sport, Warsaw, Poland
| |
Collapse
|
17
|
Effects of an Acute Exercise Bout on Serum Hepcidin Levels. Nutrients 2018; 10:nu10020209. [PMID: 29443922 PMCID: PMC5852785 DOI: 10.3390/nu10020209] [Citation(s) in RCA: 38] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/26/2017] [Revised: 01/30/2018] [Accepted: 02/11/2018] [Indexed: 02/07/2023] Open
Abstract
Iron deficiency is a frequent and multifactorial disorder in the career of athletes, particularly in females. Exercise-induced disturbances in iron homeostasis produce deleterious effects on performance and adaptation to training; thus, the identification of strategies that restore or maintain iron homeostasis in athletes is required. Hepcidin is a liver-derived hormone that degrades the ferroportin transport channel, thus reducing the ability of macrophages to recycle damaged iron, and decreasing iron availability. Although it has been suggested that the circulating fraction of hepcidin increases during early post-exercise recovery (~3 h), it remains unknown how an acute exercise bout may modify the circulating expression of hepcidin. Therefore, the current review aims to determine the post-exercise expression of serum hepcidin in response to a single session of exercise. The review was carried out in the Dialnet, Elsevier, Medline, Pubmed, Scielo and SPORTDiscus databases, using hepcidin (and “exercise” or “sport” or “physical activity”) as a strategy of search. A total of 19 articles were included in the review after the application of the inclusion/exclusion criteria. This search found that a single session of endurance exercise (intervallic or continuous) at moderate or vigorous intensity (60–90% VO2peak) stimulates an increase in the circulating levels of hepcidin between 0 h and 6 h after the end of the exercise bout, peaking at ~3 h post-exercise. The magnitude of the response of hepcidin to exercise seems to be dependent on the pre-exercise status of iron (ferritin) and inflammation (IL-6). Moreover, oxygen disturbances and the activation of a hypoxia-induced factor during or after exercise may stimulate a reduction of hepcidin expression. Meanwhile, cranberry flavonoids supplementation promotes an anti-oxidant effect that may facilitate the post-exercise expression of hepcidin. Further studies are required to explore the effect of resistance exercise on hepcidin expression.
Collapse
|
18
|
Pompano LM, Haas JD. Efficacy of iron supplementation may be misinterpreted using conventional measures of iron status in iron-depleted, nonanemic women undergoing aerobic exercise training. Am J Clin Nutr 2017; 106:1529-1538. [PMID: 29092885 DOI: 10.3945/ajcn.117.152777] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2017] [Accepted: 10/05/2017] [Indexed: 11/14/2022] Open
Abstract
Background: Despite its known detrimental effects, iron deficiency remains the most common micronutrient deficiency in the world. Many interventions that aim to improve iron status involve physically active populations. Intense aerobic exercise training negatively affects iron status; however, the impact of regular moderate aerobic exercise on the effectiveness of iron supplementation remains unclear.Objective: This study aimed to determine whether aerobic training modifies the assessment of the effectiveness of iron supplementation in improving conventional iron status measures.Design: Seventy-two iron-depleted, nonanemic Chinese women [serum ferritin (sFer) <25 μg/L and hemoglobin >110 g/L] were included in an 8-wk, partially blinded, randomized controlled trial with a 2 × 2 factorial design including iron supplements (42 mg elemental Fe/d) or placebo and aerobic training (five 25-min sessions/wk at 75-85% of maximum heart rate) or no training. Linear mixed models were used to evaluate the relation between supplement type, training, and changes in iron status over time, measured by sFer, hemoglobin, soluble transferrin receptor (sTfR), and estimated total body iron.Results: After treatment, both the iron-supplemented trained and untrained groups showed significantly improved sFer, sTfR, and body iron values compared with either of the placebo groups. Similarly, trained participants had significantly higher aerobic fitness measures than untrained participants. Training modified the sFer response to supplementation (training by supplement interaction, P = 0.07), with the iron-supplemented trained group having significantly lower sFer than the iron-supplemented untrained group at week 8 (mean ± SD: 31.8 ± 13.5 and 47.6 ± 15.7 μg/L, respectively; P = 0.042), whereas there was no significant difference between the placebo trained and untrained groups (21.3 ± 12.2 and 20.3 ± 7.0 μg/L, respectively; P = 1.00).Conclusions: Regular aerobic training reduces the apparent effectiveness of iron supplementation in improving sFer and calls into question whether conventional measures of iron status accurately reflect iron metabolism in physically active, nonanemic women. This trial was registered at clinicaltrials.gov as NCT03002090.
Collapse
Affiliation(s)
- Laura M Pompano
- Division of Nutritional Sciences, Cornell University, Ithaca, NY
| | - Jere D Haas
- Division of Nutritional Sciences, Cornell University, Ithaca, NY
| |
Collapse
|
19
|
|
20
|
SOUZA JANAINASDE, BRUNETTO ERIKAL, NUNES MARIATEREZA. Iron restriction increases myoglobin gene and protein expression in Soleus muscle of rats. AN ACAD BRAS CIENC 2016; 88:2277-2290. [DOI: 10.1590/0001-3765201620160173] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2016] [Accepted: 07/12/2016] [Indexed: 12/21/2022] Open
Affiliation(s)
- JANAINA S. DE SOUZA
- Universidade de São Paulo, Brazil; Universidade Federal de São Paulo, Brazil
| | | | | |
Collapse
|
21
|
Buratti P, Gammella E, Rybinska I, Cairo G, Recalcati S. Recent Advances in Iron Metabolism: Relevance for Health, Exercise, and Performance. Med Sci Sports Exerc 2016; 47:1596-604. [PMID: 25494391 DOI: 10.1249/mss.0000000000000593] [Citation(s) in RCA: 46] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Iron is necessary for physiological processes essential for athletic performance, such as oxygen transport, energy production, and cell division. However, an excess of "free" iron is toxic because it produces reactive hydroxyl radicals that damage biological molecules, thus leading to cell and tissue injury. Therefore, iron homeostasis is strictly regulated; and in recent years, there have been important advancements in our knowledge of the underlying processes. Hepcidin is the central regulator of systemic iron homeostasis and exerts its function by controlling the presence of the iron exporter ferroportin on the cell membrane. Hepcidin binding induces ferroportin degradation, thus leading to cellular iron retention and decreased levels of circulating iron. As iron is required for hemoglobin synthesis, the tight link between erythropoiesis and iron metabolism is particularly relevant to sports physiology. The iron needed for hemoglobin synthesis is ensured by inhibiting hepcidin to increase ferroportin activity and iron availability and hence to make certain that efficient blood oxygen transport occurs for aerobic exercise. However, hepcidin expression is also affected by exercise-associated conditions, such as iron deficiency, anemia or hypoxia, and, particularly, inflammation, which can play a role in the pathogenesis of sports anemia. Here, we review recent advances showing the relevance of iron for physical exercise and athletic performance. Low body iron levels can cause anemia and thus limit the delivery of oxygen to exercising muscle, but tissue iron deficiency may also affect performance by, for example, hampering muscle oxidative metabolism. Accordingly, a hemoglobin-independent effect of iron on exercise capacity has been demonstrated in animal models and humans. Here, we review recent advances showing the relevance of iron for physical exercise and athletic performance.
Collapse
Affiliation(s)
- Paolo Buratti
- Department of Biomedical Sciences for Health, University of Milan, Milan, ITALY
| | | | | | | | | |
Collapse
|
22
|
Recalcati S, Gammella E, Cairo G. New perspectives on the molecular basis of the interaction between oxygen homeostasis and iron metabolism. HYPOXIA 2015; 3:93-103. [PMID: 27774486 PMCID: PMC5045093 DOI: 10.2147/hp.s83537] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 01/01/2023]
Abstract
Oxygen and iron are two elements closely related from a (bio)chemical point of view. Moreover, they share the characteristic of being indispensable for life, while also being potentially toxic. Therefore, their level is strictly monitored, and sophisticated pathways have evolved to face variations in either element. In addition, the expression of proteins involved in iron and oxygen metabolism is mainly controlled by a complex interplay of proteins that sense both iron levels and oxygen availability (ie, prolyl hydroxylases, hypoxia inducible factors, and iron regulatory proteins), and in turn activate feedback mechanisms to re-establish homeostasis. In this review, we describe how cells and organisms utilize these intricate networks to regulate responses to changes in oxygen and iron levels. We also explore the role of these pathways in some pathophysiological settings.
Collapse
Affiliation(s)
- Stefania Recalcati
- Department of Biomedical Sciences for Health, University of Milan, Milan, Italy
| | - Elena Gammella
- Department of Biomedical Sciences for Health, University of Milan, Milan, Italy
| | - Gaetano Cairo
- Department of Biomedical Sciences for Health, University of Milan, Milan, Italy
| |
Collapse
|
23
|
Altamura S, Bärtsch P, Dehnert C, Maggiorini M, Weiss G, Theurl I, Muckenthaler MU, Mairbäurl H. Increased hepcidin levels in high-altitude pulmonary edema. J Appl Physiol (1985) 2014; 118:292-8. [PMID: 25525212 DOI: 10.1152/japplphysiol.00940.2014] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/17/2023] Open
Abstract
Low iron availability enhances hypoxic pulmonary vasoconstriction (HPV). Considering that reduced serum iron is caused by increased erythropoiesis, insufficient reabsorption, or elevated hepcidin levels, one might speculate that exaggerated HPV in high-altitude pulmonary edema (HAPE) is related to low serum iron. To test this notion we measured serum iron and hepcidin in blood samples obtained in previously published studies at low altitude and during 2 days at 4,559 m (HA1, HA2) from controls, individuals with HAPE, and HAPE-susceptible individuals where prophylactic dexamethasone and tadalafil prevented HAPE. As reported, at 4,559 m pulmonary arterial pressure was increased in healthy volunteers but reached higher levels in HAPE. Serum iron levels were reduced in all groups at HA2. Hepcidin levels were reduced in all groups at HA1 and HA2 except in HAPE, where hepcidin was decreased at HA1 but unexpectedly high at HA2. Elevated hepcidin in HAPE correlated with increased IL-6 at HA2, suggesting that an inflammatory response related to HAPE contributes to increased hepcidin. Likewise, platelet-derived growth factor, a regulator of hepcidin, was increased at HA1 and HA2 in controls but not in HAPE, suggesting that hypoxia-controlled factors that regulate serum iron are inappropriately expressed in HAPE. In summary, we found that HAPE is associated with inappropriate expression of hepcidin without inducing expected changes in serum iron within 2 days at HA, likely due to too short time. Although hepcidin expression is uncoupled from serum iron availability and hypoxia in individuals developing HAPE, our findings indicate that serum iron is not related with exaggerated HPV.
Collapse
Affiliation(s)
- Sandro Altamura
- Pediatric Oncology, Hematology & Immunology, University Hospital Heidelberg, Germany
| | - Peter Bärtsch
- Medical Clinic VII, Sports Medicine, University Hospital Heidelberg, Germany
| | - Christoph Dehnert
- Medical Clinic VII, Sports Medicine, University Hospital Heidelberg, Germany
| | - Marco Maggiorini
- Intensive Care Unit, Internal Medicine, University Hospital Zürich, Switzerland; and
| | - Günter Weiss
- Department of Internal Medicine VI, Clinical Immunology and Infectious Diseases, Medical University of Innsbruck, Austria
| | - Igor Theurl
- Department of Internal Medicine VI, Clinical Immunology and Infectious Diseases, Medical University of Innsbruck, Austria
| | | | - Heimo Mairbäurl
- Medical Clinic VII, Sports Medicine, University Hospital Heidelberg, Germany;
| |
Collapse
|
24
|
Ryan BJ, Wachsmuth NB, Schmidt WF, Byrnes WC, Julian CG, Lovering AT, Subudhi AW, Roach RC. AltitudeOmics: rapid hemoglobin mass alterations with early acclimatization to and de-acclimatization from 5260 m in healthy humans. PLoS One 2014; 9:e108788. [PMID: 25271637 PMCID: PMC4182755 DOI: 10.1371/journal.pone.0108788] [Citation(s) in RCA: 55] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2014] [Accepted: 08/26/2014] [Indexed: 01/09/2023] Open
Abstract
It is classically thought that increases in hemoglobin mass (Hbmass) take several weeks to develop upon ascent to high altitude and are lost gradually following descent. However, the early time course of these erythropoietic adaptations has not been thoroughly investigated and data are lacking at elevations greater than 5000 m, where the hypoxic stimulus is dramatically increased. As part of the AltitudeOmics project, we examined Hbmass in healthy men and women at sea level (SL) and 5260 m following 1, 7, and 16 days of high altitude exposure (ALT1/ALT7/ALT16). Subjects were also studied upon return to 5260 m following descent to 1525 m for either 7 or 21 days. Compared to SL, absolute Hbmass was not different at ALT1 but increased by 3.7±5.8% (mean ± SD; n = 20; p<0.01) at ALT7 and 7.6±6.6% (n = 21; p<0.001) at ALT16. Following descent to 1525 m, Hbmass was reduced compared to ALT16 (−6.0±3.7%; n = 20; p = 0.001) and not different compared to SL, with no difference in the loss in Hbmass between groups that descended for 7 (−6.3±3.0%; n = 13) versus 21 days (−5.7±5.0; n = 7). The loss in Hbmass following 7 days at 1525 m was correlated with an increase in serum ferritin (r = −0.64; n = 13; p<0.05), suggesting increased red blood cell destruction. Our novel findings demonstrate that Hbmass increases within 7 days of ascent to 5260 m but that the altitude-induced Hbmass adaptation is lost within 7 days of descent to 1525 m. The rapid time course of these adaptations contrasts with the classical dogma, suggesting the need to further examine mechanisms responsible for Hbmass adaptations in response to severe hypoxia.
Collapse
Affiliation(s)
- Benjamin J. Ryan
- Department of Integrative Physiology, University of Colorado Boulder, Boulder, Colorado, United States of America
- * E-mail:
| | - Nadine B. Wachsmuth
- Department of Sports Medicine/Sports Physiology, University of Bayreuth, Bayreuth, Germany
| | - Walter F. Schmidt
- Department of Sports Medicine/Sports Physiology, University of Bayreuth, Bayreuth, Germany
| | - William C. Byrnes
- Department of Integrative Physiology, University of Colorado Boulder, Boulder, Colorado, United States of America
| | - Colleen G. Julian
- Altitude Research Center, Department of Emergency Medicine, University of Colorado Anschutz Medical Campus, Aurora, Colorado, United States of America
| | - Andrew T. Lovering
- Department of Human Physiology, University of Oregon, Eugene, Oregon, United States of America
| | - Andrew W. Subudhi
- Altitude Research Center, Department of Emergency Medicine, University of Colorado Anschutz Medical Campus, Aurora, Colorado, United States of America
- Department of Biology, University of Colorado Colorado Springs, Colorado Springs, Colorado, United States of America
| | - Robert C. Roach
- Altitude Research Center, Department of Emergency Medicine, University of Colorado Anschutz Medical Campus, Aurora, Colorado, United States of America
| |
Collapse
|
25
|
Govus AD, Abbiss CR, Garvican-Lewis LA, Swinkels DW, Laarakkers CM, Gore CJ, Peeling P. Acute hypoxic exercise does not alter post-exercise iron metabolism in moderately trained endurance athletes. Eur J Appl Physiol 2014; 114:2183-91. [DOI: 10.1007/s00421-014-2938-2] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2014] [Accepted: 06/12/2014] [Indexed: 12/11/2022]
|
26
|
Epoetin beta pegol (C.E.R.A.) promotes utilization of iron for erythropoiesis through intensive suppression of serum hepcidin levels in mice. Int J Hematol 2014; 99:561-9. [DOI: 10.1007/s12185-014-1554-6] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2013] [Revised: 02/19/2014] [Accepted: 02/20/2014] [Indexed: 02/08/2023]
|
27
|
Badenhorst CE, Dawson B, Goodman C, Sim M, Cox GR, Gore CJ, Tjalsma H, Swinkels DW, Peeling P. Influence of post-exercise hypoxic exposure on hepcidin response in athletes. Eur J Appl Physiol 2014; 114:951-9. [PMID: 24487960 DOI: 10.1007/s00421-014-2829-6] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2013] [Accepted: 01/20/2014] [Indexed: 02/07/2023]
Abstract
PURPOSE To assess the influence of a simulated altitude exposure (~2,900 m above sea level) for a 3 h recovery period following intense interval running on post-exercise inflammation, serum iron, ferritin, erythropoietin, and hepcidin response. METHODS In a cross-over design, ten well-trained male endurance athletes completed two 8 × 3 min interval running sessions at 85 % of their maximal aerobic velocity on a motorized treadmill, before being randomly assigned to either a hypoxic (HYP: F IO2 ~0.1513) or a normoxic (NORM: F IO2 0.2093) 3 h recovery period. Venous blood was collected pre- and immediately post-exercise, and after 3 and 24 h of recovery. Blood was analyzed for interleukin-6, serum iron, ferritin, erythropoietin, and hepcidin. RESULTS Interleukin-6 was significantly elevated (p < 0.01) immediately post-exercise compared to baseline (NORM: 1.08 ± 0.061 to 3.12 ± 1.80) (HYP: 1.32 ± 0.86 to 2.99 ± 2.02), but was not different between conditions. Hepcidin levels were significantly elevated (p < 0.01) at 3 h post-exercise for both conditions when compared to baseline (NORM: 3.25 ± 1.23 to 7.40 ± 4.00) (HYP: 3.24 ± 1.94 to 5.42 ± 3.20), but were significantly lower (p < 0.05) in the HYP trial compared to NORM. No significant differences existed between HYP and NORM for erythropoietin, serum iron, or ferritin. CONCLUSION Simulated altitude exposure (~2,900 m) for 3 h following intense interval running attenuates the peak hepcidin levels recorded at 3 h post-exercise. Consequently, a hypoxic recovery after exercise may be useful for athletes with compromised iron status to potentially increase acute dietary iron absorption.
Collapse
Affiliation(s)
- Claire E Badenhorst
- School of Sport Science, Exercise and Health, The University of Western Australia, M408, 35 Stirling Hwy, Crawley, WA, 6009, Australia
| | | | | | | | | | | | | | | | | |
Collapse
|
28
|
Sasaki Y, Shimonaka Y, Ikuta K, Hosoki T, Sasaki K, Torimoto Y, Kanada H, Moriguchi Y, Kohgo Y. Hepcidin production in response to iron is controlled by monocyte-derived humoral factors. Int J Hematol 2013; 99:12-20. [PMID: 24293278 DOI: 10.1007/s12185-013-1476-8] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2013] [Revised: 11/13/2013] [Accepted: 11/14/2013] [Indexed: 02/08/2023]
Abstract
Hepcidin, which is mainly produced by the liver, is the key regulator in iron homeostasis. Hepcidin expression is up-regulated by iron loading in vivo, but the mechanism underlying this process is not completely understood. In the present study, we investigated the mechanism, following the hypothesis that hepcidin production in response to iron loading is regulated by extra-hepatic iron sensors. We measured serum hepcidin concentrations and iron indices in Wistar rats treated with saccharated ferric oxide (SFO). Human hepatoma-derived HepG2 cells were stimulated using SFO-administered rat sera, and co-cultured with rat spleen cells, human monocyte-derived THP-1 cells, or human monocytes with diferric transferrin (holo-Tf), and hepcidin concentrations in the conditioned media were measured. SFO elevated rat serum hepcidin concentrations. SFO-treated rat sera increased hepcidin production from HepG2 cells, and this induction correlated with serum hepcidin levels, but not with iron indices. Holo-Tf up-regulated hepcidin concentrations in media from HepG2 cells co-cultured with rat spleen cells, THP-1 cells, or human monocytes with or without cell-to-cell contacts, while holo-Tf did not up-regulate hepcidin from HepG2 cells alone. Our results suggest the existence of humoral factors capable of inducing hepcidin production that are secreted by extra-hepatic cells, such as reticuloendothelial monocytes, in response to iron.
Collapse
Affiliation(s)
- Yusuke Sasaki
- Product Research Department, Kamakura Research Labs, Chugai Pharmaceutical Co., Ltd., 200 Kajiwara, Kamakura, Kanagawa, 247-8530, Japan,
| | | | | | | | | | | | | | | | | |
Collapse
|
29
|
Díaz V, Gammella E, Recalcati S, Santambrogio P, Naldi AM, Vogel J, Gassmann M, Cairo G. Liver iron modulates hepcidin expression during chronically elevated erythropoiesis in mice. Hepatology 2013; 58:2122-32. [PMID: 23744538 DOI: 10.1002/hep.26550] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/21/2012] [Accepted: 05/22/2013] [Indexed: 01/01/2023]
Abstract
UNLABELLED The liver-derived peptide hepcidin controls the balance between iron demand and iron supply. By inhibiting the iron export activity of ferroportin, hepcidin modulates iron absorption and delivery from the body's stores. The regulation of hepcidin, however, is not completely understood and includes a variety of different signals. We studied iron metabolism and hepcidin expression in mice constitutively overexpressing erythropoietin (Epo) (Tg6 mice), which leads to excessive erythropoiesis. We observed a very strong down-regulation of hepcidin in Tg6 mice that was accompanied by a strong increase in duodenal expression of ferroportin and divalent metal tranporter-1, as well as enhanced duodenal iron absorption. Despite these compensatory mechanisms, Tg6 mice displayed marked circulating iron deficiency and low levels of iron in liver, spleen, and muscle. To elucidate the primary signal affecting hepcidin expression during chronically elevated erythropoiesis, we increased iron availability by either providing iron (thus further increasing the hematocrit) or reducing erythropoiesis-dependent iron consumption by means of splenectomy. Both treatments increased liver iron and up-regulated hepcidin expression and the BMP6/SMAD pathway despite continuously high plasma Epo levels and sustained erythropoiesis. This suggests that hepcidin expression is not controlled by erythropoietic signals directly in this setting. Rather, these results indicate that iron consumption for erythropoiesis modulates liver iron content, and ultimately BMP6 and hepcidin. Analysis of the BMP6/SMAD pathway targets showed that inhibitor of DNA binding 1 (ID1) and SMAD7, but not transmembrane serine protease 6 (TMPRSS6), were up-regulated by increased iron availability and thus may be involved in setting the upper limit of hepcidin. CONCLUSION We provide evidence that under conditions of excessive and effective erythropoiesis, liver iron regulates hepcidin expression through the BMP6/SMAD pathway.
Collapse
Affiliation(s)
- Víctor Díaz
- Institute of Veterinary Physiology, Vetsuisse Faculty, and Zurich Center for Integrative Human Physiology (ZIHP), and University of Zurich, Switzerland; Department of Health and Human Performance, Faculty of Sports Science, INEF, Technical University of Madrid, Spain
| | | | | | | | | | | | | | | |
Collapse
|
30
|
Makropoulos DA, Achuthanandam R, Avery J, Wilson K, Brosnan K, Miller A, Nesspor T, Chroscinski D, Walker M, Egenolf D, Huang C, Bugelski PJ. CNTO 530 increases expression of HbA and HbF in murine models of β-thalassemia and sickle cell anemia. Curr Pharm Biotechnol 2013; 14:242-8. [PMID: 23157711 PMCID: PMC3613800 DOI: 10.2174/138920113805219449] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2011] [Accepted: 06/16/2011] [Indexed: 11/24/2022]
Abstract
CNTO 530 is an erythropoietin receptor agonist MIMETIBODYTM construct. CNTO 530 has been shown to be active in a number of rodent models of acquired anemia (e.g. renal insufficiency and chemotherapy induced anemia). We investigated the efficacy of CNTO 530 in murine models of β-thalassemia and sickle cell anemia (Berkeley mice). β- thalassemic mice are deficient in expression of α-globin chain and heterozygous mice are characterized by a clinical syndrome similar to the human β-thalassemia intermedia. Berkeley mice are knocked out for murine alpha and beta globin and are transgenic for human alpha, beta (sickle) and gamma globin genes. Berkeley mice thus express human sickle hemoglobin A (HbS) and can also express human fetal hemoglobin. These mice express a severe compensated hypochromic microcytic anemia and display the sickle cell phenotype. To test the effectiveness of CNTO 530, mice from both genotypes received a single subcutaneous (s.c.) dose of CNTO 530 or darbepoetin-α (as a comparator) at 10,000 U/kg, a dose shown to cause a similar increase in reticulocytes and hemoglobin in normal mice. Hematologic parameters were evaluated over time. CNTO 530, but not darbepoetin-α, increased reticulocytes, red blood cells and total hemoglobin in β- thalassemic mice. In Berkeley mice CNTO 530 showed an increase in reticulocytes, red blood cells, F-cells, total hemoglobin and fetal hemoglobin. In conclusion, CNTO 530 is effective in murine models of β-thalassemia and sickle cell anemia. These data suggest that CNTO 530 may have beneficial effects in patients with genetically mediated hemoglobinopathies.
Collapse
Affiliation(s)
- Dorie A Makropoulos
- Biologics Toxicology, Center of Excellence in Biotechnology, Centocor R & D, Radnor, PA 19087, USA.
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
31
|
Swaminathan S, Bose C, Shah SV, Hall KA, Hiatt KM. Gadolinium contrast agent-induced CD163+ ferroportin+ osteogenic cells in nephrogenic systemic fibrosis. THE AMERICAN JOURNAL OF PATHOLOGY 2013; 183:796-807. [PMID: 23867799 DOI: 10.1016/j.ajpath.2013.06.008] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/08/2013] [Revised: 05/29/2013] [Accepted: 06/05/2013] [Indexed: 12/21/2022]
Abstract
Gadolinium-based contrast agents are linked to nephrogenic systemic fibrosis in patients with renal insufficiency. The pathology of nephrogenic systemic fibrosis is characterized by abnormal tissue repair: fibrosis and ectopic ossification. The mechanisms by which gadolinium could induce fibrosis and ossification are not known. We examined in vitro the effect of a gadolinium-based contrast agent on human peripheral blood mononuclear cells for phenotype and function relevant to the pathology of nephrogenic systemic fibrosis using immunofluorescence, flow cytometry, real-time PCR, and osteogenic assays. We also examined tissues from patients with nephrogenic systemic fibrosis, using IHC to identify the presence of cells with phenotype induced by gadolinium. Gadolinium contrast induced differentiation of human peripheral blood mononuclear cells into a unique cellular phenotype--CD163(+) cells expressing proteins involved in fibrosis and bone formation. These cells express fibroblast growth factor (FGF)23, osteoblast transcription factors Runt-related transcription factor 2, and osterix, and show an osteogenic phenotype in in vitro assays. We show in vivo the presence of CD163(+)/procollagen-1(+)/osteocalcin(+) cells in the fibrotic and calcified tissues of nephrogenic systemic fibrosis patients. Gadolinium contrast-induced CD163(+)/ferroportin(+)/FGF23(+) cells with osteogenic potential may play a role in systemic fibrosis and ectopic ossification in nephrogenic systemic fibrosis.
Collapse
Affiliation(s)
- Sundararaman Swaminathan
- Division of Nephrology, Department of Internal Medicine, University of Arkansas for Medical Sciences, Little Rock, Arkansas, USA.
| | | | | | | | | |
Collapse
|
32
|
Robach P, Recalcati S, Girelli D, Campostrini N, Kempf T, Wollert KC, Corbella M, Santambrogio P, Perbellini L, Brasse-Lagnel C, Christensen B, Moutereau S, Lundby C, Cairo G. Serum hepcidin levels and muscle iron proteins in humans injected with low- or high-dose erythropoietin. Eur J Haematol 2013; 91:74-84. [PMID: 23582009 DOI: 10.1111/ejh.12122] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 04/05/2013] [Indexed: 01/05/2023]
Abstract
Inhibition of hepcidin expression by erythropoietic signals is of great physiological importance; however, the inhibitory pathways remain poorly understood. To investigate (i) the direct effect of erythropoietin (Epo) and (ii) the contribution of putative mediators on hepcidin repression, healthy volunteers were injected with a single dose of Epo, either low (63 IU/kg, n = 8) or high (400 IU/kg, n = 6). Low-dose Epo provoked hepcidin down-modulation within 24 h; the effect was not immediate as hepcidin circadian variations were still present following injection. High-dose Epo induced no additional effect on the hepcidin response, that is hepcidin diurnal fluctuations were not abolished in spite of extremely high Epo levels. We did not find significant changes in putative mediators of hepcidin repression, such as transferrin saturation, soluble transferrin receptor, or growth differentiation factor 15. Furthermore, the potential hepcidin inhibitor, soluble hemojuvelin, was found unaltered by Epo stimulation. This finding was consistent with the absence of signs of iron deficiency observed at the level of skeletal muscle tissue. Our data suggest that hepcidin repression by erythropoietic signals in humans may not be controlled directly by Epo, but mediated by a still undefined factor.
Collapse
Affiliation(s)
- Paul Robach
- Département médical, Ecole Nationale de Ski et d'Alpinisme, site de l'Ecole Nationale des Sports de Montagne, Chamonix, France
| | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
33
|
Chiabrando D, Fiorito V, Marro S, Silengo L, Altruda F, Tolosano E. Cell-specific regulation of Ferroportin transcription following experimentally-induced acute anemia in mice. Blood Cells Mol Dis 2013; 50:25-30. [DOI: 10.1016/j.bcmd.2012.08.002] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2012] [Revised: 08/03/2012] [Accepted: 08/03/2012] [Indexed: 12/17/2022]
|
34
|
Lehmberg K, Grosse R, Muckenthaler MU, Altamura S, Nielsen P, Schmid H, Graubner U, Oyen F, Zeller W, Schneppenheim R, Janka GE. Administration of recombinant erythropoietin alone does not improve the phenotype in iron refractory iron deficiency anemia patients. Ann Hematol 2012. [PMID: 23180434 DOI: 10.1007/s00277-012-1618-8] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
Mutations in transmembrane protease, serine 6 (TMPRSS6) cause iron refractory iron deficiency anemia (IRIDA). Parenteral iron administration may slightly improve hemoglobin level but is troublesome for patients. Optimal treatment has yet to be determined. We identified five patients from four independent families displaying the IRIDA picture with truncating biallelic mutations in TMPRSS6, one of which is novel. Liver iron determined by superconducting quantum interference device biosusceptometry ranged from 390 to 720 µg Fe/g wet weight (normal range 100-500; n = 3). Intestinal iron absorption (12 and 32 %, normal range 10-50; n = 2) and 59Fe erythrocyte incorporation after ingestion of 59Fe (57 and 38 %, normal range 70-90; n = 2) were inadequately low for iron-deficient anemic individuals. Baseline serum erythropoietin was elevated or borderline high in four patients. Administration of recombinant human erythropoietin (rhEPO) at up to 273 and 188 U/kg body weight/week alone did not improve anemia or result in a decrease of urinary hepcidin in two individuals. In conclusion, the ability of exogenous rhEPO to increase hemoglobin level appears to be impaired in IRIDA.
Collapse
Affiliation(s)
- Kai Lehmberg
- Department of Pediatric Hematology and Oncology, University Medical Center Eppendorf, Martinistrasse 52, 20246, Hamburg, Germany.
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
35
|
Sasaki Y, Noguchi-Sasaki M, Yasuno H, Yorozu K, Shimonaka Y. Erythropoietin stimulation decreases hepcidin expression through hematopoietic activity on bone marrow cells in mice. Int J Hematol 2012; 96:692-700. [PMID: 23160767 DOI: 10.1007/s12185-012-1217-4] [Citation(s) in RCA: 38] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2012] [Revised: 11/01/2012] [Accepted: 11/01/2012] [Indexed: 02/08/2023]
Abstract
Erythropoiesis-stimulating agents (ESA) are now central to the treatment of renal anemia and are associated with improved clinical outcomes. It is well known that erythropoietin (EPO) is a key regulator of erythropoiesis through its promotion of red blood cell production. In order to investigate the role of ESA on iron metabolism, we analyzed the regulation of the iron regulatory hormone hepcidin by ESA treatment in a bone marrow transplant model in mouse. After treating C57BL/6 mice with continuous erythropoietin receptor activator (C.E.R.A.), recombinant human epoetin-β (rhEPO), or recombinant human carbamylated epoetin-β (rhCEPO), we investigated serum hepcidin concentrations and parameters of erythropoiesis. Serum hepcidin concentrations after rhEPO treatment were analyzed in mice subjected to total body irradiation followed by bone marrow transplantation. C.E.R.A. administration caused long-term downregulation of serum hepcidin levels. Serum hepcidin levels in rhEPO-treated mice decreased significantly, whereas there was no change in rhCEPO-treated mice. The reduction in circulating hepcidin levels after rhEPO administration was not observed in irradiated mice. Finally, bone marrow transplantation recovered the response to rhEPO administration that downregulates hepcidin concentration in irradiated mice. These results indicate that ESA treatment downregulates serum hepcidin concentrations, mainly by indirect mechanisms affecting hematopoietic activity in bone marrow cells.
Collapse
Affiliation(s)
- Yusuke Sasaki
- Product Research Department, Chugai Pharmaceutical Co., Ltd, 200 Kajiwara, Kamakura, Kanagawa, 247-8530, Japan.
| | | | | | | | | |
Collapse
|
36
|
Scarano S, Vestri A, Ermini ML, Minunni M. SPR detection of human hepcidin-25: a critical approach by immuno- and biomimetic-based biosensing. Biosens Bioelectron 2012; 40:135-40. [PMID: 22835525 DOI: 10.1016/j.bios.2012.06.060] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2012] [Revised: 06/22/2012] [Accepted: 06/29/2012] [Indexed: 10/28/2022]
Abstract
The human hepcidin-25 hormone has a key role in iron regulation in blood. The clinical relevance of this hepatic ~2.8 kDa cysteine-rich peptide is rapidly increasing, since altered levels can be associated with inflammatory events and iron dysfunctions, such as hereditary hemochromatosis and iron overload. Moreover, hepcidin has also attracted the anti-doping field for its possible role as indirect marker of erythropoietin blood doping. Methods currently reported are based on immunoassays (ELISA and RIA), or various types of mass spectroscopy (MS)-based protocols, semi-quantitative or quantitative. Despite the great effort in optimizing robust and simple assays measuring hepcidin in real matrices, at present this challenge remains still an open issue. To explore the possibility to face hepcidin detection through the development of affinity-based biosensors, we set up a comparative study by surface plasmon resonance (SPR) technology. An immuno-based, on anti-hepcidin-25 IgG, and a biomimetic-based, on a synthetic peptide corresponding to the hepcidin-binding site on ferroportin (HBD), biosensors were developed. Here we report behaviors and analytical performances of the two systems, discussing limits and potentialities.
Collapse
Affiliation(s)
- S Scarano
- Dipartimento di Chimica Ugo Schiff e CSGI, Università di Firenze, via della Lastruccia 3, 50019 Sesto Fiorentino, Firenze, Italy.
| | | | | | | |
Collapse
|
37
|
Robach P, Boisson RC, Vincent L, Lundby C, Moutereau S, Gergelé L, Michel N, Duthil E, Féasson L, Millet GY. Hemolysis induced by an extreme mountain ultra-marathon is not associated with a decrease in total red blood cell volume. Scand J Med Sci Sports 2012; 24:18-27. [PMID: 22672635 DOI: 10.1111/j.1600-0838.2012.01481.x] [Citation(s) in RCA: 51] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 04/17/2012] [Indexed: 12/14/2022]
Abstract
Prolonged running is known to induce hemolysis. It has been suggested that hemolysis may lead to a significant loss of red blood cells; however, its actual impact on the erythrocyte pool is unknown. Here, we test the hypothesis that prolonged running with high hemolytic potential decreases total red blood cell volume (RCV). Hemolysis (n = 22) and RCV (n = 19) were quantified in ultra-marathon runners before and after a 166-km long mountain ultra-endurance marathon (RUN) with 9500 m of altitude gain/loss. Assessment of total hemoglobin mass (Hbmass) and RCV was performed using a carbon monoxide rebreathing technique. RUN induced a marked acute-phase response and promoted hemolysis, as shown by a decrease in serum haptoglobin (P < 0.05). Elevated serum erythropoietin concentration and reticulocyte count after RUN were indicative of erythropoietic stimulation. Following RUN, runners experienced hemodilution, mediated by a large plasma volume expansion and associated with a large increase in plasma aldosterone. However, neither Hbmass nor RCV were found to be altered after RUN. Our findings indicate that mechanical/physiological stress associated with RUN promotes hemolysis but this has no impact on total erythrocyte volume. We therefore suggest that exercise 'anemia' is entirely due to plasma volume expansion and not to a concomitant decrease in RCV.
Collapse
Affiliation(s)
- P Robach
- Medical Department, National School for Skiing and Mountaineering, Site of the National School for Mountain Sports, Chamonix, France
| | | | | | | | | | | | | | | | | | | |
Collapse
|
38
|
Iron levels in polarized macrophages: regulation of immunity and autoimmunity. Autoimmun Rev 2012; 11:883-9. [PMID: 22449938 DOI: 10.1016/j.autrev.2012.03.003] [Citation(s) in RCA: 104] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2012] [Accepted: 03/08/2012] [Indexed: 02/06/2023]
Abstract
Although the hallmark of autoimmune diseases remains the generation of autoantigen-specific lynfocytic cell response, growing evidence is showing a key role for macrophages in a number of autoimmune diseases. Macrophages are characterized by phenotypical and functional heterogeneity. Different immunological signals, coming from systemic blood circulation or from microenvironment, polarize macrophages to classical (M1) or alternative (M2) phenotypes. Iron accumulation in M1 macrophages is a well known bacteriostatic mechanism and one of the mechanisms at the basis of anemia associated to chronic inflammation. Moreover, some recent data suggest that iron accumulation in macrophages can directly activate macrophages to pro-inflammatory M1 phenotype, highlighting a putative role of macrophage iron retention in the pathogenesis of chronic inflammatory and autoimmune diseases. Conversely, iron content is low in M2 macrophages, principally due to increased iron release, and increased availability of iron in the extracellular milieu supported by M2 macrophages could influence the growth rate of adjacent cell and thus play an important role in tumor growth and tissue remodeling. In this review we summarize the molecular mechanisms sustaining differential iron metabolism in polarized macrophages, discuss the relevance of this metabolic signature in chronic inflammatory and autoimmune diseases, and finally focus on potential therapeutic implications rising from a better understanding of underlying molecular mechanisms.
Collapse
|
39
|
Plenge U, Belhage B, Guadalupe-Grau A, Andersen PR, Lundby C, Dela F, Stride N, Pott FC, Helge JW, Boushel R. Erythropoietin treatment enhances muscle mitochondrial capacity in humans. Front Physiol 2012; 3:50. [PMID: 22419911 PMCID: PMC3299978 DOI: 10.3389/fphys.2012.00050] [Citation(s) in RCA: 33] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2012] [Accepted: 02/23/2012] [Indexed: 11/13/2022] Open
Abstract
Erythropoietin (Epo) treatment has been shown to induce mitochondrial biogenesis in cardiac muscle along with enhanced mitochondrial capacity in mice. We hypothesized that recombinant human Epo (rhEpo) treatment enhances skeletal muscle mitochondrial oxidative phosphorylation (OXPHOS) capacity in humans. In six healthy volunteers rhEpo was administered by sub-cutaneous injection over 8 weeks with oral iron (100 mg) supplementation taken daily. Mitochondrial OXPHOS was quantified by high-resolution respirometry in saponin-permeabilized muscle fibers obtained from biopsies of the vastus lateralis before and after rhEpo treatment. OXPHOS was determined with the mitochondrial complex I substrates malate, glutamate, pyruvate, and complex II substrate succinate in the presence of saturating ADP concentrations, while maximal electron transport capacity (ETS) was assessed by addition of an uncoupler. rhEpo treatment increased OXPHOS (from 92 ± 5 to 113 ± 7 pmol·s−1·mg−1) and ETS (107 ± 4 to 143 ± 14 pmol·s−1·mg−1, p < 0.05), demonstrating that Epo treatment induces an upregulation of OXPHOS and ETS in human skeletal muscle.
Collapse
Affiliation(s)
- Ulla Plenge
- Department of Anaesthesia, Bispebjerg Hospital Copenhagen, Denmark
| | | | | | | | | | | | | | | | | | | |
Collapse
|
40
|
Lundby C, Robach P, Saltin B. The evolving science of detection of 'blood doping'. Br J Pharmacol 2012; 165:1306-15. [PMID: 22225538 PMCID: PMC3372716 DOI: 10.1111/j.1476-5381.2011.01822.x] [Citation(s) in RCA: 35] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2011] [Revised: 11/15/2011] [Accepted: 11/25/2011] [Indexed: 12/21/2022] Open
Abstract
Blood doping practices in sports have been around for at least half a century and will likely remain for several years to come. The main reason for the various forms of blood doping to be common is that they are easy to perform, and the effects on exercise performance are gigantic. Yet another reason for blood doping to be a popular illicit practice is that detection is difficult. For autologous blood transfusions, for example, no direct test exists, and the direct testing of misuse with recombinant human erythropoietin (rhEpo) has proven very difficult despite a test exists. Future blood doping practice will likely include the stabilization of the transcription factor hypoxia-inducible factor which leads to an increased endogenous erythropoietin synthesis. It seems unrealistic to develop specific test against such drugs (and the copies hereof originating from illegal laboratories). In an attempt to detect and limit blood doping, the World Anti-Doping Agency (WADA) has launched the Athlete Biological Passport where indirect markers for all types of blood doping are evaluated on an individual level. The approach seemed promising, but a recent publication demonstrates the system to be incapable of detecting even a single subject as 'suspicious' while treated with rhEpo for 10-12 weeks. Sad to say, the hope that the 2012 London Olympics should be cleaner in regard to blood doping seems faint. We propose that WADA strengthens the quality and capacities of the National Anti-Doping Agencies and that they work more efficiently with the international sports federations in an attempt to limit blood doping.
Collapse
Affiliation(s)
- Carsten Lundby
- Center for Integrative Human Physiology, Institute of Physiology, University of Zurich, Switzerland.
| | | | | |
Collapse
|
41
|
Regulation of iron transport and the role of transferrin. Biochim Biophys Acta Gen Subj 2012; 1820:188-202. [DOI: 10.1016/j.bbagen.2011.10.013] [Citation(s) in RCA: 303] [Impact Index Per Article: 23.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2011] [Revised: 10/27/2011] [Accepted: 10/30/2011] [Indexed: 12/15/2022]
|
42
|
Nairz M, Sonnweber T, Schroll A, Theurl I, Weiss G. The pleiotropic effects of erythropoietin in infection and inflammation. Microbes Infect 2011; 14:238-46. [PMID: 22094132 PMCID: PMC3278592 DOI: 10.1016/j.micinf.2011.10.005] [Citation(s) in RCA: 116] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2011] [Accepted: 10/18/2011] [Indexed: 01/30/2023]
Abstract
Erythropoietin (EPO) is a multi-functional cytokine, which exerts erythropoietic effects but also carries anti-apoptotic and immune-modulatory activities upon binding to two distinct receptors which are expressed on erythroid, parenchymal and immune cells, respectively. Whereas EPO ameliorates hemolytic anemia in malaria or trypanosomiasis and improves the course of autoimmune diseases such as inflammatory bowel disease or autoimmune encephalomyelitis, it deleteriously inhibits macrophage functions in Salmonella infection in animal models. Thus, the specific modulation of extra-erythropoietic EPO activity forms an attractive therapeutic target in infection and inflammation.
Collapse
Affiliation(s)
| | | | | | | | - Günter Weiss
- Corresponding author. Tel.: +43 512 504 23255; fax: +43 512 504 25607.
| |
Collapse
|
43
|
Fania C, Vasso M, Torretta E, Robach P, Cairo G, Lundby C, Gelfi C. Setup for human sera MALDI profiling: the case of rhEPO treatment. Electrophoresis 2011; 32:1715-27. [PMID: 21706496 DOI: 10.1002/elps.201100134] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/04/2023]
Abstract
The implementation of high-throughput technologies based on qualitative and quantitative methodologies for the characterization of complex protein mixtures is increasingly required in clinical laboratories. MALDI profiling is a robust and sensitive technology although the serum high dynamic range imposes a major limitation hampering the identification of less abundant species decreasing the quality of MALDI profiling. A setup to improve these parameters has been performed for recombinant human erythropoietin (rhEPO) monitoring in serum, analyzing the effects of two commercially available columns (MARS Hu7 and Hu14) for immunodepletion, and two matrices (α-cyano-4-hydroxycinnamic acid and 2',4'-dihydroxyacetophenone) for peak quality improvement. The immunodepletion capability of both columns was determined by 2-D DIGE, which precisely revealed the efficacy of Hu14 in protein removal and the serum dynamic range decrement. In addition, the type of matrix, the sample dilution, and the efficacy of optimized parameters were used for serum profiling of ten healthy subjects before and after rhEPO treatment. The principal component analysis indicates that a combination of Hu14 column and 2',4'-dihydroxyacetophenone matrix increases data quality allowing the discrimination between treated and untreated samples, making serum MALDI profiling suitable for clinical monitoring of rhEPO.
Collapse
Affiliation(s)
- Chiara Fania
- Dipartimento di Scienze e Tecnologie Biomediche, Università degli Studi di Milano, Milan, Italy
| | | | | | | | | | | | | |
Collapse
|
44
|
Novel inflammatory mechanisms of accelerated atherosclerosis in kidney disease. Kidney Int 2011; 80:453-63. [DOI: 10.1038/ki.2011.178] [Citation(s) in RCA: 61] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
|
45
|
Lainé F, Laviolle B, Ropert M, Bouguen G, Morcet J, Hamon C, Massart C, Westermann M, Deugnier Y, Loréal O. Early effects of erythropoietin on serum hepcidin and serum iron bioavailability in healthy volunteers. Eur J Appl Physiol 2011; 112:1391-7. [PMID: 21818622 DOI: 10.1007/s00421-011-2097-7] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2010] [Accepted: 07/22/2011] [Indexed: 01/06/2023]
Abstract
Hepcidin regulates plasma iron bioavailability and subsequently iron availability for erythropoiesis. rHuEPO has been reported to decrease hepcidin expression in case of repeated subcutaneous injections. Thus, hepcidin level measurement could be a candidate marker for detection of rHuEPO abuse. However, when used for doping, rHuEPO can be injected intravenously and the scheme of injection is unknown. Our aim was to evaluate the early effects of a single intravenous rHuEPO injection on serum hepcidin levels. Fourteen male healthy volunteers received one intravenous injection of 50 U/Kg of rHuEPO during a placebo-controlled, randomized, double-blind, cross-over study. Serum hepcidin, quantified by a competitive ELISA method and iron parameters was then evaluated for 24 h. Serum levels of hepcidin were significantly increased 4 h after rHuEPO injection when compared with placebo injection (78.3 ± 55.5 vs. 57.5 ± 34.6 ng/ml, respectively; +36%, p < 0.05), whereas iron and transferrin saturation dramatically decreased 12 h after rHuEPO injection when compared with placebo injection (9.2 ± 3.5 vs. 15.8 ± 4.2 μg/l, respectively; -42%, p < 0.05 and 14.8 ± 5.0 vs. 26.3 ± 6.4%, respectively; -44%, p < 0.05). In addition, 12 and 24 h after rHuEPO injection serum hepcidin levels were lower compared with placebo injection (41.6 ± 27.4 vs. 56.6 ± 28.1 ng/ml after 12 h; -27%, p < 0.05 and 26.0 ± 29.6 vs. 81.2 ± 29.4 ng/ml after 24 h; -68%, p < 0.05). Intravenous injection of recombinant EPO induces a precocious and transient increase of serum hepcidin leading to a transient decrease of iron bioavailability. The transitory increase and dynamics of its concentration make difficult the practical use of hepcidin to detect rHuEPO doping.
Collapse
Affiliation(s)
- Fabrice Lainé
- INSERM, Centre d'investigation Clinique 0203, 35033 Rennes, France.
| | | | | | | | | | | | | | | | | | | |
Collapse
|
46
|
Pagani A, Nai A, Corna G, Bosurgi L, Rovere-Querini P, Camaschella C, Silvestri L. Low hepcidin accounts for the proinflammatory status associated with iron deficiency. Blood 2011; 118:736-46. [PMID: 21628413 DOI: 10.1182/blood-2011-02-337212] [Citation(s) in RCA: 109] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022] Open
Abstract
Hepcidin is an antimicrobial peptide that controls systemic iron homeostasis. Hepcidin binding to its receptor ferroportin reduces iron availability, thus controlling microbial growth. In parallel it triggers an anti-inflammatory response in macrophages. Hepcidin is transcriptionally regulated by iron, through the bone morphogenetic protein-son of mothers against decapentaplegic (BMP-SMAD) pathway and by inflammation, through IL6-mediated STAT3 signaling. To investigate the mechanisms linking iron and inflammation, we treated C57BL/6 iron-deficient mice with a sublethal dose of lipopolysaccharide (LPS) and analyzed their inflammatory response in comparison with controls. We show that iron-deprived mice have a proinflammatory condition, exacerbated by LPS treatment leading to increased IL6 and TNFα mRNA in liver and spleen macrophages, and increased serum IL6 (482.29 ± 205.59 pg/mL) versus controls (69.01 ± 17.52 pg/mL; P < .05). Hepcidin was undetectable in iron-deficient mice but pretreatment with hepcidin normalized their response to LPS. Tmprss6(-/-) mice, characterized by iron deficiency and high hepcidin, show a blunted inflammatory response when challenged with LPS. Our data support a model in which the lack of hepcidin is responsible of the high inflammatory response to LPS in iron deficiency. The proinflammatory status associated with chronic iron deficiency could explain the resistance to infection seen in this condition.
Collapse
Affiliation(s)
- Alessia Pagani
- Vita-Salute San Raffaele University and Division of Genetics and Cell Biology, San Raffaele Scientific Institute, Milan, Italy
| | | | | | | | | | | | | |
Collapse
|
47
|
Guidi GC, Lechi Santonastaso C. Advancements in anemias related to chronic conditions. Clin Chem Lab Med 2011; 48:1217-26. [PMID: 20618092 DOI: 10.1515/cclm.2010.264] [Citation(s) in RCA: 25] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023]
Abstract
Anemia of chronic disease (ACD), the most frequent anemia among hospitalized patients, occurs in chronic inflammatory disorders, such as chronic infections, cancer and autoimmune diseases. Different causes contribute to ACD including diversion of iron traffic, diminished erythropoiesis, blunted response to erythropoietin, erythrophagocytosis, hematologic malignancies and solid tumors. A particular case of ACD is represented by anemia of chronic kidney disease (CKD). ACD is characterized by hyposideremia and altered iron transport. Cytokines are implicated in the ACD by reducing erythropoiesis and increasing iron sequestration in the reticuloendothelial system. The regulation of iron absorption across the epithelium of the proximal small intestine is essential for maintaining body iron concentrations within a physiologically defined range. Hepcidin controls cellular iron efflux by binding to the iron export protein ferroportin, causing ferroportin to be phosphorylated and degraded in lysosomes. Finally, hepcidin inhibits iron release from the reticulo-endothelial system. Increased expression of hepcidin leads to decreased iron absorption and iron deficient anemia. Hepcidin, therefore, is a negative regulator of iron transport in plasma. Causes of anemia in patients with CKD are multifactorial, but the most well-known cause is inadequate erythropoietin production. In these patients, anemia increases the risk of either cardiovascular disease or renal failure.
Collapse
Affiliation(s)
- Gian Cesare Guidi
- Laboratory of Clinical Biochemistry, Department of Life and Reproduction Sciences, University of Verona, Verona, Italy.
| | | |
Collapse
|
48
|
Finberg KE. Unraveling mechanisms regulating systemic iron homeostasis. HEMATOLOGY. AMERICAN SOCIETY OF HEMATOLOGY. EDUCATION PROGRAM 2011; 2011:532-537. [PMID: 22160085 PMCID: PMC3648641 DOI: 10.1182/asheducation-2011.1.532] [Citation(s) in RCA: 26] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/31/2023]
Abstract
Systemic iron balance must be tightly regulated to prevent the deleterious effects of iron deficiency and iron overload. Hepcidin, a circulating hormone that is synthesized by the liver, has emerged as a key regulator of systemic iron homeostasis. Hepcidin inhibits the absorption of dietary iron from the intestine and the release of iron derived from red blood cells from macrophages. Therefore, variation in hepcidin levels modifies the total amount of iron stored in the body and the availability of iron for erythropoiesis. The production of hepcidin by the liver is modulated by multiple physiological stimuli, including iron loading, inflammation, and erythropoietic activity. Investigation of the functions of the gene products mutated in inherited iron disorders using tissue-culture systems and animal models has provided valuable insights into the mechanisms by which these hepcidin responses are mediated. This review focuses on recent advances in our understanding of the molecular mechanisms underlying the regulation of systemic iron homeostasis.
Collapse
|
49
|
Modulation of hepcidin production during hypoxia-induced erythropoiesis in humans in vivo: data from the HIGHCARE project. Blood 2010; 117:2953-9. [PMID: 21149632 DOI: 10.1182/blood-2010-08-299859] [Citation(s) in RCA: 103] [Impact Index Per Article: 6.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022] Open
Abstract
Iron is tightly connected to oxygen homeostasis and erythropoiesis. Our aim was to better understand how hypoxia regulates iron acquisition for erythropoiesis in humans, a topic relevant to common hypoxia-related disorders. Forty-seven healthy volunteers participated in the HIGHCARE project. Blood samples were collected at sea level and after acute and chronic exposure to high altitude (3400-5400 m above sea level). We investigated the modifications in hematocrit, serum iron indices, erythropoietin, markers of erythropoietic activity, interleukin-6, and serum hepcidin. Hepcidin decreased within 40 hours after acute hypoxia exposure (P < .05) at 3400 m, reaching the lowest level at 5400 m (80% reduction). Erythropoietin significantly increased (P < .001) within 16 hours after hypoxia exposure followed by a marked erythropoietic response supported by the increased iron supply. Growth differentiation factor-15 progressively increased during the study period. Serum ferritin showed a very rapid decrease, suggesting the existence of hypoxia-dependent mechanism(s) regulating storage iron mobilization. The strong correlation between serum ferritin and hepcidin at each point during the study indicates that iron itself or the kinetics of iron use in response to hypoxia may signal hepcidin down-regulation. The combined and significant changes in other variables probably contribute to the suppression of hepcidin in this setting.
Collapse
|
50
|
Recalcati S, Minotti G, Cairo G. Iron regulatory proteins: from molecular mechanisms to drug development. Antioxid Redox Signal 2010; 13:1593-616. [PMID: 20214491 DOI: 10.1089/ars.2009.2983] [Citation(s) in RCA: 93] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/01/2023]
Abstract
Eukaryotic cells require iron for survival but, as an excess of poorly liganded iron can lead to the catalytic production of toxic radicals that can damage cell structures, regulatory mechanisms have been developed to maintain appropriate cell and body iron levels. The interactions of iron responsive elements (IREs) with iron regulatory proteins (IRPs) coordinately regulate the expression of the genes involved in iron uptake, use, storage, and export at the post-transcriptional level, and represent the main regulatory network controlling cell iron homeostasis. IRP1 and IRP2 are similar (but not identical) proteins with partially overlapping and complementary functions, and control cell iron metabolism by binding to IREs (i.e., conserved RNA stem-loops located in the untranslated regions of a dozen mRNAs directly or indirectly related to iron metabolism). The discovery of the presence of IREs in a number of other mRNAs has extended our knowledge of the influence of the IRE/IRP regulatory network to new metabolic pathways, and it has been recently learned that an increasing number of agents and physiopathological conditions impinge on the IRE/IRP system. This review focuses on recent findings concerning the IRP-mediated regulation of iron homeostasis, its alterations in disease, and new research directions to be explored in the near future.
Collapse
Affiliation(s)
- Stefania Recalcati
- Department of Human Morphology and Biomedical Sciences Città Studi, University of Milan, Milano, Italy
| | | | | |
Collapse
|