Category: Health

Iron deficiency and cardiovascular health in athletes

Iron deficiency and cardiovascular health in athletes

Athletes may also be cardiovasdular risk for Lower cholesterol levels naturally deficiency due to Iron deficiency and cardiovascular health in athletes dietary iron intake. Athletes with low levels of iron are often encouraged to Strategies for long-term blood pressure management iron supplements in athletea to raise their blood iron level, which atthletes increases the amount of oxygen their lungs can absorb known as their VO2max and their anaerobic capacity. Maximum mg iron i. In fact, physically active iron-deficient but non-anemic women who take 8mg of iron supplements twice a day for 6 weeks were able to shave an average of 30 seconds off their 5k time. See Our Editorial Process. Effect of darbepoetin alpha on exercise tolerance in anemic patients with symptomatic chronic heart failure.

Iron deficiency and cardiovascular health in athletes -

Beard, J. Iron status and exercise. American Journal of Clinical Nutrition, 72 2, Suppl. Burke, L. Clinical Sports Nutrition. Australia: McGraw-Hill. Chatard, J. Anemia and iron deficiency in athletes. Sports Medicine, 27 4 , — Eichner, E.

Sports anemia, iron supplements, and blood doping. El-Sayed, M. Haemorheology in exercise and training. Sports Medicine, 35 8 , — Rockwell, M. Understanding iron. Schumacher, Y. Effects of exercise on soluble transferrin receptor and other variables of the iron status.

British Journal of Sports Medicine, 36, — Iron Deficiency Anemia and Exercise. Joanne Adamidou, MS. Apr 30, Updated on: December 1, May, References American Dietetic Association ADA.

Related Articles. Stay On Topic. Are Students Too Buzzed on Energy Drinks? Family Meals Improve Diet Quality. There is now growing evidence that ID is an important therapeutic target for patients with HFrEF, even if they do not have anemia.

Whether this is also true for other HF phenotypes or patients with CVD in general is currently unknown. Randomized trials have shown that intravenous ferric carboxymaltose FCM improved symptoms, health-related quality of life and exercise capacity, and reduced hospitalizations for worsening HF in patients with HFrEF and mildly reduced EF The European Society of Cardiology ESC guidelines on HF provide a Class I, Level of Evidence C recommendation for periodical screening for ID and anemia with a full blood count, serum ferritin concentration, and transferrin saturation for HF patients.

Although there could be a potential role for ID in other CVDs, such as CAD, valvular heart disease, cerebrovascular disease, AF, and pulmonary hypertension, evidence is fragmentary, often conflicting, and the underlying pathophysiological mechanisms are often unknown.

Since ID can be easily treated, future research should aim for a full characterization of patients with CVDs for ID, and to identify those phenotypes or patients who are more likely to benefit from iron supplementation. x You must be logged in to save to your library.

Guidelines JACC Journals on ACC. org JACC JACC: Advances JACC: Asia JACC: Basic to Translational Science JACC: CardioOncology JACC: Cardiovascular Imaging JACC: Cardiovascular Interventions JACC: Case Reports JACC: Clinical Electrophysiology JACC: Heart Failure Membership Current Members Campaign for the Future Become a Member Renew Your Membership Member Benefits and Resources Member Sections Chapters ACC Member Directory About ACC ACC Innovation Program Our Strategic Direction Diversity and Inclusion Our History Our Bylaws and Code of Ethics Leadership and Governance Annual Report Industry Relations Support the ACC Jobs at the ACC Press Releases Social Media Book Our Conference Center.

Clinical Topics Acute Coronary Syndromes Anticoagulation Management Arrhythmias and Clinical EP Cardiac Surgery Cardio-Oncology Chronic Angina Congenital Heart Disease and Pediatric Cardiology COVID Hub Diabetes and Cardiometabolic Disease Dyslipidemia Geriatric Cardiology Heart Failure and Cardiomyopathies Hypertriglyceridemia Invasive Cardiovascular Angiography and Intervention Noninvasive Imaging Pericardial Disease Prevention Pulmonary Hypertension and Venous Thromboembolism Sports and Exercise Cardiology Stable Ischemic Heart Disease Valvular Heart Disease Vascular Medicine.

JACC Quality Improvement for Institutions NCDR CardioSmart MedAxiom Accreditation Services Contact Us Heart House N St.

Secondary defciiency points were analyzed with multiple imputation techniques when Strategies for long-term blood pressure management were unavailable for cardiovascula end point. Carfiovascular values indicate differences across all 4 quartiles using the Wilcoxon rank-sum test. Protocol for the Heart Failure Clinical Research Network: Oral Iron Repletion Effects on Oxygen UpTake in Heart Failure: IRONOUT HF. eTable 2. Multicenter Trials That Evaluated Iron Supplementation for Treatment of Iron Deficiency in Patients With Heart Failure.

Ewa A. Csrdiovascular, Stephan von Haehling, Defficiency D. Anker, Iain C. Iron is a micronutrient essential for cellular energy and metabolism, necessary for maintaining body Iron deficiency and cardiovascular health in athletes.

Iron deficiency is an important co-morbidity in patients with heart failure HF. Andd major factor in the pathogenesis heallth anaemia, it is also a separate condition with serious clinical consequences e.

impaired exercise capacity and poor prognosis Increased awareness state HF patients. Experimental evidence suggests that iron therapy in iron-deficient animals may activate molecular pathways that can be cardio-protective.

Clinical studies have Natural appetite suppressants deficienyc effects of i. iron on xardiovascular functional status, quality of life, and xardiovascular capacity in HF patients. It is hypothesized that i. iron supplementation may become healtb novel therapy Protein smoothie recipes HF patients with iron deficiency.

Iron athletfs ID Natural appetite suppressants the commonest nutritional deficiency Dsficiency, affecting more than one-third of the population.

Importance of iron for functioning and survival Strategies for long-term blood pressure management cardiofascular levels of complexity of living structures.

Inflammation and memory function deficiency is a deficifncy of chronic diseases e.

halth bowel disease, Parkinson's disease, rheumatoid disease, chronic renal failureirrespective of concomitant gealth. In the last cardiovawcular, anaemia was recognized as an important co-morbidity in HF, znd factor limiting physical activity, responsible hwalth a poor quality of life, and a predictor of unfavourable outcomes.

Iron is a hexlth active micronutrient with unique biochemical features. Hewlth Natural appetite suppressants a crucial role Ion Iron deficiency and cardiovascular health in athletes transport haemoglobin componentoxygen storage Ion componentcardiac and defuciency muscle metabolism component of oxidative enzymes and respiratory chain proteinssynthesis, and degradation of proteins, lipids, ribonucleic acids enzyme componentdeficienc332—343738 and mitochondrial function.

Iorn is required for optimal haematopoiesis. erythropoietinand impairs the differentiation and maturation of all types ayhletes haematopoietic cells. In spite of helath unquestionable role for optimal haematopoiesis, iron is indispensable for the maintenance of cellular energy deciciency metabolism of extra-haematopoietic tissues.

skeletal muscles contribute to HF pathophysiology. Iron excess accumulates in cells, and at higher concentrations defiiciency oxidative stress cardioovascular and triggers cardiomyocyte necrosis, 54 whereas at lower concentrations stimulates inducible nitric oxide synthase activity Replenishing skin cells through increased NO production induces signalling pathways promoting cell Strategies for long-term blood pressure management.

Under normal rIon, the same iron amount is lost from skin desquamation, sloughing cardiovvascular epithelial cells, and bleeding. Dietary iron in two forms, inorganic non-haem and organic haemis absorbed using distinct transmembrane transport cardjovascular consisting of Ion elements: cardiofascular specific athlstes protein complex, Defciency enzyme changing the oxidative iron state, and regulatory proteins.

Inorganic dietary iron is absorbed by the apical surface of duodenal enterocytes via the cardiovascukar metal transporter Iron deficiency and cardiovascular health in athletes Diabetic nephropathy self-care and accompanying membrane ferrireductases athlefes ferric to ferrous iron.

There are two major pools of iron, utilized and stored Figure Iron deficiency and cardiovascular health in athletes. Utilized iron consists of circulating and intracellular iron. abd for dietary absorption, macrophages eliminate senescent erythrocytes, hepatocytes release proteins regulating iron athketes hepcidin.

Stored iron is in liver, bone marrow, and spleen cells in deficieny non-toxic form in ferritin shells, defciency is secreted Anxiety relief exercises the extracellular compartment.

Iron pools interact with each cardiovascualr, and cardiofascular can be cardiovzscular between these compartments cardiovasscular tightly regulated mechanisms. Within athlehes homoeostasis, one can distinguish conceptually two dimensions of iron traffic, i.

one related with iron absorption and athldtes transport cardiovasclar tissues in the whole organism systemic iron metabolismand the other related to cadiovascular transport between athlftes within Body composition and diet cell intracellular iron metabolism.

Systemic iron metabolism is controlled by mechanisms involving hepcidin and its receptor ferroportinwhereas intracellular iron metabolism is orchestrated by a complex of iron-regulatory proteins.

Major stimuli decreasing hepcidin expression in the liver and its release into the circulation are: depleted iron stores, hypoxia, and ineffective erythropoiesis, whereas inflammation produces the opposite effect. Two types of ID need to be distinguished: absolute, and functional ID 35769—71 Figure 3.

Absolute ID reflects depleted iron stores, often with intact iron homoeostasis mechanisms and erythropoiesis.

Functional ID reflects inadequate iron supply to meet the demand despite normal or abundant body iron stores, because iron is trapped inside cells of the reticuloendothelial system and is unavailable for cellular metabolism 6970 Figure 3. It is believed to be mainly caused by pro-inflammatory activation with hepcidin overproduction see above.

The gold standard for evaluating iron stores in target tissues is a bone marrow biopsy. The invasiveness of bone marrow biopsy limits its use and can be replaced by the measurement of several blood biomarkers to show iron status indirectly in most clinical scenarios 6970727378 Figure 4.

Absolute ID reflects depleted iron stores, hence its diagnosis is based on the measurement of circulating ferritin, a reliable surrogate of stored iron quantity, which originates from iron-storing cells mainly hepatocytes and reticuloendothelial cells 6970727378 Figure 3. There is a linear relationship between serum ferritin and ferritin expression in iron storage tissues.

In such cases, for the diagnosis of absolute ID, a higher serum ferritin cut-off value is used e. Circulating iron bound to transferrin TIBC, total iron binding capacity—by transferrin reflects the amount of iron available for metabolizing target cells.

Instead, transferrin saturation Tsatthe per cent of transferrin that has iron bound to it ratio of serum iron and TIBC ×is recommended.

Therefore, in chronic diseases, absolute ID is typically diagnosed with higher cut-off ferritin values i. Iron plays a critical role in erythropoiesis, being incorporated into erythroblasts and reticulocytes. iron therapy. The red cell distribution width RDW reflecting MCV heterogeneity quantitative index of anisocytosis, i.

the percentage coefficient of MCV variation can be considered another parameter of ID. Owing to pathophysiological links and overlaps in regulatory mechanisms of erythropoietin and iron metabolism, 26—28 subjects with ID frequently have increased circulating erythropoietin levels, which can be considered another index of iron-restricted erythropoiesis in HF patients, being related to poor outcomes.

Increased soluble transferrin receptor sTfR is another sensitive indicator of ID. Because serum ferritin is a surrogate of iron stores and serum sTfR reflects the tissue iron demand, there is evidence that the combination of these two parameters may describe the iron status more accurately.

A pathophysiology milieu in HF syndrome favours the development of absolute and functional ID. The following mechanisms are presumed to be involved in the development of absolute ID in HF: i insufficient dietary iron supply, 9495 ii poor GI absorption, impaired duodenal iron transport, 96 drug interactions e.

omeprazoleor food reducing absorption, and iii GI blood loss Figure 3. Some studies demonstrate suboptimal dietary iron supply, particularly in patients with advanced HF. Based on a 4-day food diary, Hughes et al. In another study, Lourenço et al. In HF, reduced iron intake may also be a consequence of deranged transport systems in the enterocytes.

Theoretically, reduced expression of membrane proteins importing iron from the intestinal lumen to the enterocyte cytosol and the subsequent iron export to the circulation may result from increased circulating hepcidin levels, analogous to a reported experimental model of chronic kidney disease.

More importantly, the intestinal expression of hypoxia-inducible factor-2α the major regulator of the duodenal iron transportation system 98 was up-regulated in iron-deficient animals without HF, but not in animals with HF.

Heart failure is a state characterized by generalized inflammation with an augmented immune response, overactive immune cells, high circulating levels of pro-inflammatory mediators, and the up-regulation of these molecules within the failing myocardium and peripheral tissues.

renal failure, chronic infections. In this context, hepcidin can be expected to play an important role. Both in rodents and humans, acute myocardial ischaemia is accompanied by increased circulating hepcidin, which subsequently decreases during recovery. Simonis et al.

The role of hepcidin produced locally is unknown. Interestingly, in clinical settings of HF, there was no association between pro-inflammatory activation as evidenced by circulating IL-6 and hepcidin levels.

Clinical evidence on the incidence of ID in HF patients is scarce. Most available studies have presented a traditional view linking ID with anaemia. Ezekowitz et al. Witte et al. Opasich et al. The only study assessing the iron status in HF patients based on the gold standard bone marrow biopsy was reported by Nanas et al.

So far, only two observational studies have reported the incidence of ID in the general HF population. Further studies are warranted. In patients with stable systolic HF, ID was associated with reduced peak oxygen consumption and a high ventilatory response to exercise, also after an adjustment for clinical co-variables.

There is also indirect evidence linking correction of ID with an improvement in exercise capacity in a few interventional studies in HF patients, regardless of baseline anaemia. Iron deficiency carries also a risk of depression in men with systolic HF.

Jankowska et al. Iron deficiency was associated with more severe depression symptoms, irrespective of HF severity, neurohormonal activation, haemoglobin, and inflammation E.

The prognostic impact of ID in HF patients was investigated in only two observational prospective studies. They demonstrated that anaemia accompanied by ID strongly predicted cardiac mortality 33 vs.

Kaplan—Meier curves reflecting 3-year event-free survival rates in patients with systolic heart failure with vs. without iron deficiency. Mechanisms underlying links between ID and poor clinical status, exercise intolerance, and an unfavourable outcome in HF remain unclear.

Dysfunction of both the myocardium and skeletal muscles are at the centre of the pathophysiology of HF. Iron is an element of enzymes and structural proteins in cardiomyocytes, and is stored inside these cells.

Molecular elements controlling iron metabolism are tracked within healthy, failing, ischaemic, and inflamed myocardium. Most available evidence reporting myocardial molecular consequences of ID comes from the experimental model of ID-anaemia. Iron deficiency-anaemic rats develop sympathetic activation with increased cardiac output, 14 left ventricular hypertrophy, 14—17— and finally left ventricular dilatation.

Recently, Maeder et al. They provided experimental evidence that the myocardial expression of TfR 1 was regulated by β-adrenoceptor agonists and aldosterone. The haemoglobin level and iron status are interlinked determinants of exercise capacity and physical fitness.

tissue oxidative capacity and oxygen carrying capacity. The tissue oxidative capacity is impaired proportionally across the whole spectrum of ID also when haemoglobin is normal.

: Iron deficiency and cardiovascular health in athletes

We Care About Your Privacy

Expected hepcidin levels in individuals with iron deficiency and anemia are lower than the values measured in this study.

Other potential mediators of refractoriness to oral iron in heart failure seem less likely to have affected our findings. Use of anticoagulants and antiplatelet agents was prevalent, but the rate of expected loss of iron Therefore, in the absence of overt gastrointenstinal bleeding, which did not occur in any of the participants treated with oral iron during the trial, blood loss would not be expected to account for the observed minimal increases in iron stores with oral iron treatment.

The choice of iron polysaccharide formulation for this study was based on its offering the highest dose of elemental iron among available oral supplements, coupled with its tolerance profile to aid in adherence and minimize risk of unblinding participants.

Polysaccharide iron preparations have been shown to provide comparable iron repletion to iron salts. Hence, even after accounting for limited gastrointestinal iron absorption, the fold increase in oral iron exposure, compared with the recommended daily intake, served to adequately test the hypothesis that oral iron supplementation would improve iron stores and functional capacity in HFrEF.

The selection of change in peak V̇ o 2 for the primary end point, as previously described, 15 was based on the fact that peak V̇ o 2 is the gold standard indicator of functional capacity in heart failure and has been shown to improve with iron repletion in non—heart failure populations.

The lack of treatment effect on quality of life, NT-proBNP, and other physiological end points is consistent with the observed lack of treatment effect on maximal exercise capacity. Submaximum exercise capacity, indicative of endurance and independent of volitional effort, may be more sensitive to subtle changes in iron bioavailability as opposed to peak V̇ o 2.

This trial complements recent studies about intravenous iron treatment in informing the appropriate approach to iron repletion in HFrEF. However, the correlates observed between baseline iron indices and exercise capacity, as well as changes in Tsat being related to improvement in peak V̇ o 2 are consistent with results of recent trials suggesting beneficial effects of intravenous iron on functional capacity in HFrEF.

This study has some important limitations. This study was not powered to detect differences in clinical events or safety end points. There was also no direct comparison between intravenous vs oral iron repletion. Given the relatively short duration of the trial, it is possible that longer duration or higher dose of exposure may have led to more significant improvement in iron stores and increased exercise capacity, particularly among those participants with appropriately low hepcidin levels.

In addition, this study was confined to patients with HFrEF and findings may differ in heart failure with preserved ejection fraction. Among participants with iron deficiency and HFrEF, high-dose oral iron minimally augmented iron stores and did not improve exercise capacity over 16 weeks.

These findings do not support the use of oral iron supplementation to treat iron deficiency in patients with HFrEF. Corresponding Author: Gregory D. Lewis, MD, Pulmonary Critical Care Unit, Cardiology Division, Massachusetts General Hospital, 55 Fruit St, Bigelow , Boston, MA glewis partners.

Correction: This article was corrected for data and typographical errors on May 25, Author Contributions: Drs Lewis and Braunwald had full access to all of the data in the study and take responsibility for the integrity of the data and the accuracy of the data analysis.

Concept and design: Lewis, Malhotra, Hernandez, Felker, Tang, Redfield, Semigran, Givertz, Whellen, Anstrom, Shah, Desvigne-Nickens, Butler, Braunwald. Acquisition, analysis, or interpretation of data: Lewis, Malhotra, Hernandez, McNulty, Smith, Felker, Tang, LaRue, Semigran, Givertz, Van Buren, Whellen, Shah, Desvigne-Nickens, Butler, Braunwald.

Critical revision of the manuscript for important intellectual content: All authors. Administrative, technical, or material support: Hernandez, LaRue, Givertz, Shah, Desvigne-Nickens, Braunwald. Conflict of Interest Disclosures: All authors have completed and submitted the ICMJE Form for Disclosure of Potential Conflicts of Interest.

Dr Lewis reports receipt of grants to the institution from Abbott, Novartis, Shape Systems, and Stealth BioTherapeutics; personal fees for consultancies from Ironwood and Cheetah Medical; and unpaid consultancies from Luitpold and Sonivie. Dr Malhotra reports receipt of personal fees for consultancies from Akros Pharma and Third Pole and for advisory board participation from Mallinckrodt Pharmaceuticals outside the submitted work.

Dr Hernandez reports receipt of grant support from Amgen, AstraZeneca, Bayer, Bristol-Myers Squibb, GlaxoSmithKline, Luitpold, Merck, and Novartis; and personal fees from Amgen, AstraZeneca, Bayer, Bristol-Myers Squibb, Boston Scientific, Luitpold, and Novartis outside the submitted work.

Dr Felker reports receipt of grant support from NHLBI during the conduct of the study , Novartis, Amgen, Merck, Roche Diagnostics, and American Heart Association; and personal fees from Novartis, Amgen, Bristol-Myers Squibb, GlaxoSmithKline, and Myokardia outside the submitted work.

Dr Tang reports receipt of grants from NIH during the conduct of the study and also outside the submitted work. Dr Semigran reports receipt of grant support from NHLBI during the conduct of the study. Dr Van Buren reports receipt of grant support from NIH and personal fees for consultancy services from Medtronic.

Dr Anstrom reports receipt of grant support from the Heart Failure Clinical Research Network during the conduct of the study. Dr Butler reports receipt of grant support from the NHLBI and receipt of personal fees for consultancy services from Luitpold.

Dr Braunwald reports grant support to his institution from Duke University for his role as chair of the NHLBI Heart Failure Network , Merck, AstraZeneca, Novartis, Daiichi Sankyo, and GlaxoSmithKline; personal fees for consultancies from The Medicines Company and Theravance; personal fees for lectures from Medscape, Menarini International, and Daiichi Sankyo; and uncompensated consultancies and lectures for Merck and Novartis.

The NHLBI was not able to prevent manuscript submission. IRONOUT HF Trial Members, Investigators, and Committees: In addition to the Writing Committee, the following individuals participated in the IRONOUT HF study: HFN Member Clinical Centers—Boston VA Healthcare System: N.

Lakdawala, S. Ly, M. Quinn; Brigham and Women's Hospital: S. Anello, K. Brooks; Cleveland Clinic Foundation: T. Fonk, K. Meera; Duke University Medical Center: P. Adams, S. Chavis, A. Mbugua; Emory University Hospital: G. Snell, T. Burns, T. Dickson, N.

Islam; Johns Hopkins Hospital: R. Tedford, A. Bacher; Lancaster General Hospital: T. Nossuli, C. Forney, S. Pointer, H. Testa; Massachusetts General Hospital: D. Cocca-Spofford; Mayo Clinic: S.

Cho, S. Decker, J. Gatzke; Metro Health System: M. Dunlap, J. Nichols, P. Leo; Northwestern Memorial Hospital: S. Shah, H.

Mkrdichian, C. Sanchez; Saint Louis University Hospital: P. Hauptman, M. Lesko, E. Weber; Stony Brook University Medical Center: I. Caikauskaite, N. Nayyar, L. Papadimitriou; Thomas Jefferson University Hospital: S.

Adams, M. Fox, B. Gallagher, M. McCarey, K. Murphy; Tufts Medical Center: G. Huggins, A. Cronkright, G. Jamieson, R. Oliveira, T. Cheutzow; University of Missouri Health System: C.

Danila, S. Collins; University of Pennsylvania Health System: K. Margulies, T. Coppola, T. Wahlen VA Medical Center: S. Drakos, J. Nativi-Nicolau, J. Gibbs, J. Gutierrez; University of Vermont Medical Center: M.

LeWinter, M. Rowen; VA St. Louis Health Care System: I. Halatchev, C. Rowe; Washington University School of Medicine: V. Davila-Roman, J. Flanagan, D. Whitehead; HFN Data and Safety Monitoring Board—D.

Vaughan chair , R. Agarwal, J. Ambrose, D. DeGrazia, K. Kennedy, M. Johnson, J. Parrillo, M. Penn, M. Powers, E. Rose; Protocol Review Committee—W. Abraham chair , R. Cai, D. McNamara, J. Rose, D. Vaughan, R. Virmani; Biomarker Core Lab—University of Vermont: R. Tracy, R. Boyle; CPET Core Lab—L.

Wooster, C. Bailey, A. Dress, D. Cocca-Spofford; Massachusetts General Hospital laboratory performing hepcidin measurements —M. Buswell, G. Shelton, K. This article is also focused on adult athletes and the information discussed may not apply to children.

Iron is a mineral that has several important roles in the body including energy metabolism, oxygen transport, and acid-base balance. Red blood cells transport oxygen throughout the body and are filled with proteins called hemoglobin.

Each hemoglobin molecule contains iron. Oxygen picked up in the lungs binds to the iron inside hemoglobin and then is carried all over the body to supply oxygen to organs and tissues. Iron comes from our diet. Dietary iron can be classified into heme iron and non-heme iron.

Heme iron is found in meat, poultry, and fish. Red meat contains about three times as much iron as both poultry and fish making it one of the richest sources of dietary iron. Heme iron is absorbed by the digestive tract about twice as well as non-heme iron.

Sources of non-heme iron includes fruits, vegetables, and iron fortified foods. Vitamin C assists with the absorption of non-heme iron in the digestive tract so mixing foods rich in vitamin C with non-heme iron containing foods can increase the amount of iron the body absorbs.

Athletes need more iron than the general population. Iron is lost through sweat, skin, urine, the gastrointestinal GI tract, and menstruation.

Athletes lose more iron due to heavy sweating as well as increased blood loss in the urine and GI tract. The mechanical force of a footstrike during endurance running, for example, can increase the destruction of red blood cells in the feet, leading to a shorter red blood cell life span.

Female athletes are at even higher risk for iron deficiency as compared to males due to monthly blood loss associated with menstruation. Athletes may also be at risk for iron deficiency due to insufficient dietary iron intake. Remember, the body is not very effective at absorbing dietary iron. Those following a strict vegetarian or vegan diet can be at even higher risk for iron deficiency due to the decreased absorption of non-heme iron found in plants and fortified foods.

Because iron is necessary for oxygen transport and energy metabolism, both of which are critical for fueling aerobic exercise, endurance athletes can experience a decline in exercise capacity and VO2 max, the maximal amount of oxygen the body can use, with iron deficiency.

As iron deficiency becomes more severe, the body cannot make a sufficient number of red blood cells and anemia, meaning low red blood cells, develops.

Athletes with iron deficiency anemia will generally have more pronounced symptoms than those with iron deficiency alone. A craving for ice chips is actually pretty specific to iron deficiency, so any athletes out there who find themselves wanting to eat a lot of ice should definitely have their iron levels checked.

Iron deficiency is diagnosed through blood tests. The most useful of the typical iron study panel is ferritin, which is a marker of iron stores. In the sports nutrition community, there is no clear ferritin goal for athletes. If a ferritin is dropping significantly during the course of a training cycle, this can also be indicative of developing iron deficiency and the need to intervene, even if the ferritin is within what is generally considered a normal range.

It is also worth mentioning that ferritin levels can quickly increase when the body is under stress so results may be falsely high during periods of active infection or inflammation.

The other traditional iron panel tests can be useful in distinguishing iron deficiency from poor iron utilization states. A complete blood count CBC measures the levels of red blood cell in the body and determines whether or not someone is anemic.

Pregnant women with low iron may be more prone to infection because iron also supports the immune system. It is clear that iron supplements are needed for women who are both pregnant and iron-deficient. However, research is ongoing as to the possibility of recommending additional iron to all pregnant women, even those with normal iron levels.

It is argued that all pregnant women should take 30 to 60 milligrams mg of iron supplements on every day of their pregnancy, regardless of their iron levels.

Insufficient iron in the diet can affect the efficiency with which the body uses energy. Iron carries oxygen to the muscles and brain and is crucial for both mental and physical performance. Low iron levels may result in a lack of focus, increased irritability, and reduced stamina.

Iron deficiency is more common among athletes, especially young female athletes, than in individuals who do not lead an active lifestyle. This appears to be particularly true in female endurance athletes, such as long-distance runners.

Some experts suggest that female endurance athletes should add an additional 10 mg of elemental iron per day to the current RDA for iron intake. Iron deficiency in athletes decreases athletic performance and weakens immune system activity.

For more in-depth resources about vitamins, minerals, and supplements, visit our dedicated hub. Iron has a low bioavailability, meaning that the small intestine does not readily absorb large amounts. This decreases its availability for use and increases the likelihood of deficiency.

There are two types of dietary iron, known as heme and non-heme. Animal sources of food, including meat and seafood, contain heme iron. Heme iron is more easily absorbed by the body. Non-heme iron, the type found in plants, requires that the body take multiple steps to absorb it.

Plant-based sources of iron include beans, nuts, soy, vegetables, and fortified grains. The bioavailability of heme iron from animal sources can be up to 40 percent. Non-heme iron from plant-based sources, however, has a bioavailability of between 2 and 20 percent.

For this reason, the RDA for vegetarians is 1. Consuming vitamin-C-rich foods alongside non-heme sources of iron can dramatically increase iron absorption. When following a vegetarian diet, it is also important to consider components of food and medications that block or reduce iron absorption, such as:.

Calcium can slow both heme and non-heme iron absorption. In most cases, a typical varied, Western-style diet is considered balanced in terms of enhancers and inhibitors of iron absorption. In adults, doses for oral iron supplementation can be as high as 60 to mg of elemental iron per day.

These doses typically apply to women who are pregnant and severely iron-deficient. An upset stomach is a common side effect of iron supplementation, so dividing doses throughout the day may help. Adults with a healthy digestive system have a very low risk of iron overload from dietary sources.

People with a genetic disorder called hemochromatosis are at a high risk of iron overload as they absorb far more iron from food when compared to people without the condition. This can lead to a buildup of iron in the liver and other organs. It can also cause the creation of free radicals that damage cells and tissues, including the liver, heart, and pancreas, as well increasing the risk of certain cancers.

Frequently taking iron supplements that contain more than 20 mg of elemental iron at a time can cause nausea, vomiting, and stomach pain, especially if the supplement is not taken with food. In severe cases, iron overdoses can lead to organ failure, internal bleeding, coma , seizure, and even death.

It is important to keep iron supplements out of reach of children to reduce the risk of fatal overdose. According to Poison Control, accidental ingestion of iron supplements was the most common cause of death from an overdose of medication in children less than 6 years old until the s.

Changes in the manufacture and distribution of iron supplements have helped reduce accidental iron overdoses in children, such as replacing sugar coatings on iron tablets with film coatings, using child-proof bottle caps, and individually packaging high doses of iron.

Share This

Penn, M. Powers, E. Rose; Protocol Review Committee—W. Abraham chair , R. Cai, D. McNamara, J. Rose, D. Vaughan, R.

Virmani; Biomarker Core Lab—University of Vermont: R. Tracy, R. Boyle; CPET Core Lab—L. Wooster, C. Bailey, A. Dress, D. Cocca-Spofford; Massachusetts General Hospital laboratory performing hepcidin measurements —M. Buswell, G. Shelton, K. Allen, D.

Bloch; Coordinating Center—Duke Clinical Research Institute: E. Velazquez, A. Devore, L. Cooper, J. Kelly, P. Monds, M. Sellers, T. Atwood, K. Hwang, T. full text icon Full Text. Download PDF Top of Article Key Points Abstract Introduction Methods Results Discussion Conclusions Article Information References.

Figure 1. Flow of Participants for the IRONOUT HF Study. View Large Download. a Data on patients screened for eligibility were not available. Figure 2. Relationships Between Quartiles of Baseline Plasma Hepcidin Levels and Response in Participants Treated With Iron Polysaccharide.

Table 1. Baseline Characteristics of Participants in the IRONOUT HF Study a. Table 2. Primary, Secondary, and Safety End Points. Table 3. Levels of Iron Metabolism Markers According to Treatment Group. Supplement 1. Supplement 2. IRONOUT HF Inclusion and Exclusion Criteria eTable 1. Serious Adverse Events Listed by Body System for the 2 Treatment Groups eTable 2.

Multicenter Trials That Evaluated Iron Supplementation for Treatment of Iron Deficiency in Patients With Heart Failure eFigure 1. Forest Plot For Prespecified Subgroup Analysis Relative to the Primary End Point of Change in Peak VO2 at Week 16 eFigure 2.

Panel A: Time to First Serious and Nonserious Adverse Event Panel B: Time to Death or Cardiovascular Hospitalization eReferences.

Supplement 3. Statistical Analysis. Pasricha SR. Anemia: a comprehensive global estimate. PubMed Google Scholar Crossref.

Klip IT, Comin-Colet J, Voors AA, et al Iron deficiency in chronic heart failure. Am Heart J. Jankowska EA, Rozentryt P, Witkowska A, et al. Iron deficiency predicts impaired exercise capacity in patients with systolic chronic heart failure.

J Card Fail. Weiss G, Goodnough LT. Anemia of chronic disease. N Engl J Med. Dong F, Zhang X, Culver B, Chew HG Jr, Kelley RO, Ren J. Dietary iron deficiency induces ventricular dilation, mitochondrial ultrastructural aberrations and cytochrome c release.

Clin Sci Lond. Toblli JE, Lombraña A, Duarte P, Di Gennaro F. Intravenous iron reduces NT-pro-brain natriuretic peptide in anemic patients with chronic heart failure and renal insufficiency. J Am Coll Cardiol.

Dunn LL, Suryo Rahmanto Y, Richardson DR. Iron uptake and metabolism in the new millennium. Trends Cell Biol. Haas JD, Brownlie T 4th. Iron deficiency and reduced work capacity. J Nutr. PubMed Google Scholar. Andrews NC. Disorders of iron metabolism.

Melenovsky V, Petrak J, Mracek T, et al. Myocardial iron content and mitochondrial function in human heart failure. Eur J Heart Fail. Georgieva Z, Georgieva M.

Compensatory and adaptive changes in microcirculation and left ventricular function of patients with chronic iron-deficiency anaemia. Clin Hemorheol Microcirc. Jankowska EA, Ponikowski P.

Molecular changes in myocardium in the course of anemia or iron deficiency. Heart Fail Clin. Anker SD, Comin Colet J, Filippatos G, et al; FAIR-HF Trial Investigators. Ferric carboxymaltose in patients with heart failure and iron deficiency.

Ponikowski P, van Veldhuisen DJ, Comin-Colet J, et al; CONFIRM-HF Investigators. Beneficial effects of long-term intravenous iron therapy with ferric carboxymaltose in patients with symptomatic heart failure and iron deficiency.

Eur Heart J. Lewis GD, Semigran MJ, Givertz MM, et al. Oral iron therapy for heart failure with reduced ejection fraction: design and rationale for oral iron repletion effects on oxygen uptake in heart failure.

Circ Heart Fail. Green CP, Porter CB, Bresnahan DR, Spertus JA. Development and evaluation of the Kansas City Cardiomyopathy Questionnaire.

Chatterjee NA, Murphy RM, Malhotra R, et al. Prolonged mean V̇o 2 response time in systolic heart failure. Nemeth E, Valore EV, Territo M, Schiller G, Lichtenstein A, Ganz T. Hepcidin, a putative mediator of anemia of inflammation, is a type II acute-phase protein.

Nicolas G, Chauvet C, Viatte L, et al. The gene encoding the iron regulatory peptide hepcidin is regulated by anemia, hypoxia, and inflammation. J Clin Invest. Ganz T. Hepcidin and iron regulation, 10 years later. Nemeth E, Tuttle MS, Powelson J, et al.

Hepcidin regulates cellular iron efflux by binding to ferroportin and inducing its internalization. Franchini M, Montagnana M, Lippi G.

Hepcidin and iron metabolism. Clin Chim Acta. Swank AM, Horton J, Fleg JL, et al; HF-ACTION Investigators. Modest increase in peak V̇o 2 is related to better clinical outcomes in chronic heart failure patients. Cardiopulmonary exercise testing in heart failure.

JACC Heart Fail. Okonko DO, Grzeslo A, Witkowski T, et al. Effect of intravenous iron sucrose on exercise tolerance in anemic and nonanemic patients with symptomatic chronic heart failure and iron deficiency FERRIC-HF. Beck-da-Silva L, Piardi D, Soder S, et al.

IRON-HF study: a randomized trial to assess the effects of iron in heart failure patients with anemia. Int J Cardiol. van Santen S, van Dongen-Lases EC, de Vegt F, et al.

Hepcidin and hemoglobin content parameters in the diagnosis of iron deficiency in rheumatoid arthritis patients with anemia. Arthritis Rheum. Choi HS, Song SH, Lee JH, Kim HJ, Yang HR. Serum hepcidin levels and iron parameters in children with iron deficiency.

Korean J Hematol. Jacobs P, Fransman D, Coghlan P. Comparative bioavailability of ferric polymaltose and ferrous sulphate in iron-deficient blood donors.

J Clin Apher. Here, look at how to get more iron in the…. Iron deficiency anemia is prevalent among older people. Find out the reasons behind this and learn about the causes, symptoms, and treatment of iron…. Hemochromatosis causes the body to absorb too much iron.

Learn about the causes, symptoms, and treatments here. Hemochromatosis causes people to absorb too much iron from food. In this article, we look at whether and how making dietary changes can help treat….

My podcast changed me Can 'biological race' explain disparities in health? Why Parkinson's research is zooming in on the gut Tools General Health Drugs A-Z Health Hubs Health Tools Find a Doctor BMI Calculators and Charts Blood Pressure Chart: Ranges and Guide Breast Cancer: Self-Examination Guide Sleep Calculator Quizzes RA Myths vs Facts Type 2 Diabetes: Managing Blood Sugar Ankylosing Spondylitis Pain: Fact or Fiction Connect About Medical News Today Who We Are Our Editorial Process Content Integrity Conscious Language Newsletters Sign Up Follow Us.

Medical News Today. Health Conditions Health Products Discover Tools Connect. Everything you need to know about iron. Medically reviewed by Judith Marcin, M. Recommended intake Benefits Foods Risks Iron is a mineral vital to the proper function of hemoglobin, a protein needed to transport oxygen in the blood and perform other various processes.

Fast facts on iron The Recommended Daily Allowance RDA varies between ages, but women who are pregnant require the most. Iron promotes healthy pregnancy, increased energy, and better athletic performance. Iron deficiency is most common in female athletes.

Canned clams, fortified cereals, and white beans are the best sources of dietary iron. Too much iron can increase the risk of liver cancer and diabetes. Was this helpful? Recommended intake. Share on Pinterest Iron is important for maintaining a healthy pregnancy.

Further resources For more in-depth resources about vitamins, minerals, and supplements, visit our dedicated hub. Share on Pinterest Clams are an excellent source of iron.

How we reviewed this article: Sources. Medical News Today has strict sourcing guidelines and draws only from peer-reviewed studies, academic research institutions, and medical journals and associations.

We avoid using tertiary references. We link primary sources — including studies, scientific references, and statistics — within each article and also list them in the resources section at the bottom of our articles.

You can learn more about how we ensure our content is accurate and current by reading our editorial policy. Share this article. Latest news Ovarian tissue freezing may help delay, and even prevent menopause. RSV vaccine errors in babies, pregnant people: Should you be worried?

Scientists discover biological mechanism of hearing loss caused by loud noise — and find a way to prevent it. How gastric bypass surgery can help with type 2 diabetes remission.

Atlantic diet may help prevent metabolic syndrome. Related Coverage. What to know about iron deficiency anemia. Medically reviewed by Alana Biggers, M. Hypoxia-inducible factor 2α HIF2α upregulates the expression of both the brush border machinery DMT1 and DCYTB that uptakes iron from the lumen and the iron exporter FPN at the basolateral membrane by binding hypoxia-responsive elements of these gene promoters.

Macrophages rapidly recycle iron derived from the phagocytosis of senescent red cells Figure 1. However, the absolute amount of iron recycled from hypochromic erythrocytes by heme-oxygenase 1 decreases in parallel with the severity of iron deficiency, because Hb content per cell mean corpuscular Hb [MCH] is reduced.

A novel mechanism related to erythrocyte FPN, which is highly expressed in iron deficiency, may contribute to maintaining circulating iron levels. Cellular iron content is controlled by IRPs that in iron deficiency bind stem-loop sequences IREs in the untranslated regions UTRs of iron genes to posttranscriptionally coordinate proteins of iron absorption, export, use, and storage.

Other IRP-independent mechanisms optimize iron use in low-iron states. mTOR inhibition activates tristetraprolin, which reduces both TFR1 and FPN expression to save iron for tissue metabolic needs. In iron deficiency, ferritin is delivered to autophagosomes for degradation ferritinophagy by nuclear receptor coactivator 4, which in contrast is proteasome degraded in iron-replete cells.

Although serum ferritin is the best biomarker of iron deficiency, the mechanisms of its release as well as its function in the circulation remain mysterious.

Iron restriction limits the expansion of early erythropoiesis and optimizes iron use by terminal erythropoiesis. In vitro iron deprivation blunts the EPO responsiveness of early progenitors through inactivation of iron-dependent aconitase, which suppresses isocitrate production.

The same phenotype, expression of increased EPO sensitivity, is recapitulated by the genetic loss of the EPOR partner TFR2 in mice; this condition mimics iron deficiency, 27 because TFR2 is lost from the membrane when diferric TF is reduced. With the development of anemia and hypoxia, EPO levels increase exponentially, and multiple mediators, such as erythroferrone, 13 GDF15, 29 and PDGF-BB, 30 suppress hepcidin to enhance iron supply.

In this process, a role of soluble TFR sTFR , an accepted biomarker of iron deficiency, 31 although reasonable, remains unproven. Because of the increased number of erythroblasts and limited iron supply, heme content per cell is reduced. Globin translation is also impaired by low heme; the stress sensor heme-regulated inhibitor HRI phosphorylates the elongation initiation factor 2a eIF2A to block translation, concomitantly increasing ATF4, which inhibits the translation regulator mTOR.

The optimization of erythropoiesis might preserve iron for vital functions within a global body economy. However, the mechanism is not fully effective, because even in the absence of anemia, other organs may become iron deficient.

In Western countries, other healthy individuals may be at risk. These include vegetarians, especially vegans, because of diet restriction and blood donors. Females are more affected in all the groups listed here.

Iron deficiency with or without anemia may be isolated or secondary to a causative disorder or occur in the context of multiple pathological conditions eg, in the elderly.

Iron deficiency is usually acquired and exceptionally inherited. In developing countries, iron deficiency anemia is nutritional, resulting from reduced intake of bioavailable iron Table 1 , and often associated with infections causing hemorrhages, such as hookworm infestation or schistosomiasis.

ESA, erythropoiesis-stimulating agent; H 2 antagonists, histamine receptor blockers; IRIDA, iron-refractory iron deficiency anemia; PNH, paroxysmal nocturnal hemoglobinuria.

Rarely resulting from gene mutations other than TMPRSS6. Absolute iron deficiency may be masked by comorbidities eg, in the elderly, and in the setting of renal failure. Anemia in the elderly has multiple causes. Unfortunately, being obscured by comorbidities, it often remains undiagnosed, 38 while even mild anemia worsens the outcome of associated disorders and influences mortality.

A recognized cause of dysregulation of iron metabolism is obesity, which may lead to iron deficiency, especially after bariatric surgery because of global absorption impairment Table 1. Considering the need for balancing iron demand and supply, specific clinical settings are characterized by acute restriction of iron for erythropoiesis.

The best-known example is treatment with erythropoiesis-stimulating agents. Another example is postoperative anemia that follows major surgery.

IRIDA 43 is a rare recessive condition resulting from mutations of TMPRSS6 , 44 , 45 leading to an inability to cleave the BMP coreceptor HJV and inhibit hepcidin.

IRIDA patients are refractory to oral iron supplementation. In adults, especially men, anemia may be less evident than in children, while iron deficiency and microcytosis persist. Populations studies suggest that susceptibility to iron deficiency is in part influenced by genetics.

Studies of blood donors have strengthened the hypothesis that genetic variants of iron genes, especially TMPRSS6 and HFE , reported to influence iron parameters 49 , 50 and hepcidin, 51 may predispose to or protect individuals from iron deficiency.

Iron deficiency anemia characterizes both germinal and intestinal conditional Bpnt 1 knockout mice, establishing a novel link between sulfur and iron homeostasis.

Clinical signs and symptoms of iron deficiency anemia are limited and often neglected. The most important, fatigue, is unspecific. Alterations of epithelial cells such as dry mouth, cheilitis, atrophic glossitis, Plummer-Vinson pharyngeal webs, and hair loss are observed in longstanding deficiency.

Restless leg syndrome reveals iron deficiency in a proportion of cases. For a detailed discussion of symptoms in iron deficiency anemia, readers are referred elsewhere.

A correct diagnosis requires laboratory tests. Low serum ferritin levels are the hallmark of absolute iron deficiency, reflecting exhausted stores. Measuring serum hepcidin may be diagnostic of this atypical iron deficiency, provided that inflammation is excluded. Reticulocyte Hb content may reveal rapid changes in erythropoietic activity.

All tissues are assumed to be iron deficient when ferritin is low. No specific test assesses tissue eg, cardiac or muscle iron deficiency when ferritin is unreliable, such as in inflammation. Perception of this deficiency by patients is highly variable.

Clinical diagnosis relies on deterioration of the specific organ eg, heart function or on unspecific symptoms, the most popular being fatigue. Alternatively, the diagnosis is based on a positive outcome after iron supplementation, such as in heart failure.

The etiological cause of iron deficiency should be addressed in all cases and, whenever possible, eliminated.

Iron treatment should be started immediately, even in the absence of anemia, especially in symptomatic patients. The choice of iron compound and the route of administration are largely dependent on the presence and degree of anemia, reversibility of the underlying cause, clinical status age, sex, longstanding vs recent onset , and in some instances patient preference.

Iron salts such as iron sulfate, fumarate, and gluconate remain a mainstay of therapy in absolute iron deficiency.

Mounting evidence indicates that low doses are more effective and better tolerated than the traditionally recommended to mg of elementary iron per day. Importantly, even a mild increase in serum iron activates hepcidin to limit iron absorption. This physiological response was exploited to design the most appropriate dose and schedule of oral iron administration in iron-deficient nonanemic women.

In short-term studies that used stable iron isotopes, supplementation with iron sulfate mg induced hepcidin increase for up to 48 hours, limiting the absorption of the subsequent doses.

In a study comparing 2 groups of women who were receiving mg of iron sulfate per day either as a single or 2 divided doses, the first group showed smaller serum hepcidin increases.

An ongoing study in women with iron deficiency anemia 70 is assessing whether the alternate-day protocol should also be recommended in the presence of anemia, 71 when hypoxia further increases intestinal iron absorption and fully suppresses hepcidin.

Other adverse effects of unabsorbed iron include alterations in the composition of the gut microbiome, with reduction of beneficial Lactobacillus and Bifidobacterium bacteria, enhancement of potential pathogens Enterobacteriaceae , and increased inflammation and diarrhea, as shown in African children.

The minimal dose used for iron supplementation is 60 mg per day. Lower doses A prophylactic treatment with iron sulfate 60 mg in adults and 30 mg in children has been recommended in world areas characterized by high prevalence of iron deficiency anemia.

Epidemiological 75 and in vitro studies have shown that iron deficiency is an adaptation process protecting from Plasmodium virulence and that its correction may increase infection severity. This would increase erythrocyte iron content, favoring the parasite growth. To avoid the latter effects, a future solution is the development of iron compounds bioavailable only to humans and not to pathogens.

There is great interest in the development of compounds better tolerated than iron salts; numerous compounds have been proposed eg, sucrosomial iron, heme iron polypeptide, iron containing nanoparticles , but studies are limited. In the same condition, the phosphate binder iron ferric citrate simultaneously corrects both hyperphosphatemia and iron deficiency; its double effect is being tested in a clinical trial in CKD.

The natural compound extracted from the bark of the Taiwanese tree hinokitiol restores iron transport in cells lacking transporters, such as DMT1 or FPN. The alternative for patients intolerant or unresponsive to oral compounds is IV iron.

Advantages are the more rapid effect and the negligible gastrointestinal toxicity. IV iron is available in different forms; iron gluconate and iron sucrose require repeated infusions, whereas ferric carboxymaltose, ferumoxytol, low molecular weight iron dextran, and iron isomaltoside may be administered in high doses to rapidly replace the total iron deficit usually However, this decision should be carefully made on an individual basis.

High-dose IV iron may increase Hb or iron stores before surgery predicted to induce heavy bleeding. This is a kind of prevention of acute postoperative anemia and an alternative to blood transfusions, which are associated with several postoperative complications, including infections. Patient blood management programs that limit blood transfusions by perioperative iron use reduce morbidity and negative prognoses in high-risk interventions.

An important issue concerning IV iron is safety. Because iron is a growth factor for several pathogens, iron therapy is contraindicated in infections. The risk of infection after IV iron is still a matter of controversy.

Increased risk was found in a meta-analysis evaluating trials of IV iron to spare transfusions, 93 and caution was suggested in dialysis patients. Hypophosphatemia after ferricarboxymaltose is usually transient and reversible, although rarely, severe cases have been reported after repeated infusions.

The superior efficacy of IV vs oral iron is undisputable and expected; the long-term adverse effects of ROS generation in cases of therapy-induced positive iron balance have been scarcely explored, although overtreatment might occur in functional rather than in absolute iron deficiency.

A recent analysis in CKD concluded that patients seemed to tolerate positive iron balance, because iron that was not used was safely stored in reticule-endothelial cells.

Although advances in understanding iron metabolism and regulation are systematically providing novel insights, additional studies are needed before iron therapy becomes a personalized approach in all cases.

These studies should aim at discovering markers of tissue iron deficiency, investigate novel schedules of iron administration based on iron physiology, provide clearer indications to high-dose IV iron, and contribute long-term evaluations of treatment outcomes.

The author thanks Domenico Girelli for his valuable advice and criticism and Alessia Pagani for help with the figure. Conflict-of-interest disclosure: C. is an advisor for Vifor Iron Core and has received honoraria from Vifor Pharma.

Iron deficiency linked with cardiovascular disease Share on Reddit. Efficacy of oral iron therapy in patients receiving recombinant human erythropoietin. All tissues are assumed to be iron deficient when ferritin is low. It has found that iron deficiency within the smooth muscle cells of the pulmonary arteries is in itself sufficient to cause PAH, even in the absence of anaemia. Owing to pathophysiological links and overlaps in regulatory mechanisms of erythropoietin and iron metabolism, 26—28 subjects with ID frequently have increased circulating erythropoietin levels, which can be considered another index of iron-restricted erythropoiesis in HF patients, being related to poor outcomes. Papadimitriou; Thomas Jefferson University Hospital: S.
Iron Deficiency Anemia and Exercise - IDEA Health & Fitness Association

Sign In. Search Dropdown Menu. header search search input Search input auto suggest. filter your search All Content All Journals Blood. Toggle Menu Menu Issues Current Issue All Issues First edition Abstracts Annual Meeting Late Breaking Annual Meeting Late Breaking Annual Meeting Annual Meeting Late Breaking All Meeting Abstracts Collections Collections Special Collections Multimedia Alerts Author Center Submit Author Guide Style Guide Why Submit to Blood?

About About Blood Editorial Board and Staff Subscriptions Public Access Copyright Alerts Blood Classifieds. Skip Nav Destination Content Menu. Close Abstract. Pathophysiology of iron deficiency. Etiology of iron deficiency. Diagnosing iron deficiency. Advances and controversies in the treatment of iron deficiency.

Future perspectives. Article Navigation. Iron Metabolism and Its Disorders January 3, Clara Camaschella Clara Camaschella. Division of Genetics and Cell Biology, San Raffaele Scientific Institute, Milan, Italy. This Site. Google Scholar. Blood 1 : 30— Article history Submitted:.

Connected Content. A related article has been published: Introduction to a review series on iron metabolism and its disorders. A companion article has been published: The molecular genetics of sideroblastic anemia.

A companion article has been published: Iron metabolism under conditions of ineffective erythropoiesis in β-thalassemia. View more. A companion article has been published: Liver iron sensing and body iron homeostasis.

A companion article has been published: Anemia of inflammation. An erratum has been published: Camaschella C. Iron deficiency. View less. Split-Screen Share Icon Share Facebook Twitter LinkedIn Email Tools Icon Tools Request Permissions.

Cite Icon Cite. toolbar search Search Dropdown Menu. toolbar search search input Search input auto suggest. Figure 1. View large Download PPT. Table 1. Type of cause. Pathophysiologic mechanism. View Large. Table 2. Indication for IV iron therapy. Contribution: C.

conceived, wrote, and reviewed the paper. GBD Disease and Injury Incidence and Prevalence Collaborators. Global, regional, and national incidence, prevalence, and years lived with disability for diseases and injuries for countries, a systematic analysis for the Global Burden of Disease Study Search ADS.

An overview of evidence for a causal relation between iron deficiency during development and deficits in cognitive or behavioral function. The effects of oral iron supplementation on cognition in older children and adults: a systematic review and meta-analysis.

Structure-function analysis of ferroportin defines the binding site and an alternative mechanism of action of hepcidin. Hepcidin regulates cellular iron efflux by binding to ferroportin and inducing its internalization.

The serine protease matriptase-2 TMPRSS6 inhibits hepcidin activation by cleaving membrane hemojuvelin. Regulation of cell surface transferrin receptor-2 by iron-dependent cleavage and release of a soluble form.

Hepcidin is regulated by promoter-associated histone acetylation and HDAC3. Identification of erythroferrone as an erythroid regulator of iron metabolism. The gut in iron homeostasis: role of HIF-2 under normal and pathological conditions.

Erythrocytic ferroportin reduces intracellular iron accumulation, hemolysis, and malaria risk. The physiological functions of iron regulatory proteins in iron homeostasis — an update. Quantitative proteomics identifies NCOA4 as the cargo receptor mediating ferritinophagy. Ferritinophagy via NCOA4 is required for erythropoiesis and is regulated by iron dependent HERC2-mediated proteolysis.

Iron control of erythroid development by a novel aconitase-associated regulatory pathway. Isocitrate ameliorates anemia by suppressing the erythroid iron restriction response.

Deletion of iron regulatory protein 1 causes polycythemia and pulmonary hypertension in mice through translational derepression of HIF2α. The IRP1-HIF-2α axis coordinates iron and oxygen sensing with erythropoiesis and iron absorption.

The second transferrin receptor regulates red blood cell production in mice. Transferrin receptor 2: evidence for ligand-induced stabilization and redirection to a recycling pathway. High levels of GDF15 in thalassemia suppress expression of the iron regulatory protein hepcidin. Hypoxia induced downregulation of hepcidin is mediated by platelet derived growth factor BB.

Quantitative assessment of erythropoiesis and functional classification of anemia based on measurements of serum transferrin receptor and erythropoietin. HRI coordinates translation by eIF2αP and mTORC1 to mitigate ineffective erythropoiesis in mice during iron deficiency. National Heart, Lung, and Blood Institute Recipient Epidemiology and Donor Evaluation Study-III REDS-III.

Effect of iron supplementation on iron stores and total body iron after whole blood donation. Oral versus intravenous iron replacement therapy distinctly alters the gut microbiota and metabolome in patients with IBD. Mutations in TMPRSS6 cause iron-refractory iron deficiency anemia IRIDA.

Membrane-bound serine protease matriptase-2 Tmprss6 is an essential regulator of iron homeostasis. Ineffective erythropoiesis and regulation of iron status in iron loading anaemias. De Falco.

Functional and clinical impact of novel TMPRSS6 variants in iron-refractory iron-deficiency anemia patients and genotype-phenotype studies.

A genome-wide meta-analysis identifies 22 loci associated with eight hematological parameters in the HaemGen consortium. Novel loci affecting iron homeostasis and their effects in individuals at risk for hemochromatosis [published correction appears in Nat Commun.

Association of HFE and TMPRSS6 genetic variants with iron and erythrocyte parameters is only in part dependent on serum hepcidin concentrations.

Genetic factors influencing ferritin levels in 14, blood donors: results from the Danish Blood Donor Study. Modulation of intestinal sulfur assimilation metabolism regulates iron homeostasis. The prevalence and impact of restless legs syndrome on patients with iron deficiency anemia.

Hepcidin is the major predictor of erythrocyte iron incorporation in anemic African children. Laboratory methodologies for indicators of iron status: strengths, limitations, and analytical challenges.

Intravenous iron for the treatment of fatigue in nonanemic, premenopausal women with low serum ferritin concentration. Efficacy of iron supplementation on fatigue and physical capacity in non-anaemic iron-deficient adults: a systematic review of randomised controlled trials.

Ferrous sulfate supplementation causes significant gastrointestinal side-effects in adults: a systematic review and meta-analysis. Oral iron supplements increase hepcidin and decrease iron absorption from daily or twice-daily doses in iron-depleted young women.

Iron absorption from oral iron supplements given on consecutive versus alternate days and as single morning doses versus twice-daily split dosing in iron-depleted women: two open-label, randomised controlled trials. In anaemic women, daily oral iron supplementation increases hepcidin and decreases iron absorption compared to alternate day dosing.

In: Proceedings from the European Iron Club Annual Meeting; February ; Zurich, Switzerland. Iron fortification adversely affects the gut microbiome, increases pathogen abundance and induces intestinal inflammation in Kenyan infants.

World Health Organization. Use of Multiple Micronutrient Powders for Point-of-Use Fortification of Foods Consumed by Infants and Young Children Aged Months and Children Aged Years.

Iron deficiency protects against severe Plasmodium falciparum malaria and death in young children. Effects of routine prophylactic supplementation with iron and folic acid on admission to hospital and mortality in preschool children in a high malaria transmission setting: community-based, randomised, placebo-controlled trial.

Host iron status and iron supplementation mediate susceptibility to erythrocytic stage Plasmodium falciparum. Modern iron replacement therapy: clinical and pathophysiological insights. Effect of oral liposomal iron versus intravenous iron for treatment of iron deficiency anaemia in CKD patients: a randomized trial.

Rationale, design, and characteristics of a trial to evaluate the new phosphate iron-based binder sucroferric oxyhydroxide in dialysis patients with the goal of advancing the practice of E. Ferric maltol is effective in correcting iron deficiency anemia in patients with inflammatory bowel disease: results from a phase-3 clinical trial program.

Restored iron transport by a small molecule promotes absorption and hemoglobinization in animals. FIND-CKD: a randomized trial of intravenous ferric carboxymaltose versus oral iron in patients with chronic kidney disease and iron deficiency anaemia.

Management of iron-deficiency anemia in inflammatory bowel disease: a systematic review. van Rheenen. Iron deficiency in gynecology and obstetrics: clinical implications and management. Intravenous ferric carboxymaltose versus standard care in the management of postoperative anaemia: a prospective, open-label, randomised controlled trial.

Preoperative management of colorectal cancer-induced iron deficiency anemia in clinical practice: data from a large observational cohort. Safety and efficacy of intravenous iron therapy in reducing requirement for allogeneic blood transfusion: systematic review and meta-analysis of randomised clinical trials.

Iron deficiency anemia in chronic kidney disease: uncertainties and cautions. The safety of intravenous iron preparations: systematic review and meta-analysis. Choice of high-dose intravenous iron preparation determines hypophosphatemia risk. Hypersensitivity to intravenous iron: classification, terminology, mechanisms and management.

Hypersensitivity reactions to intravenous iron: guidance for risk minimization and management. Positive iron balance in chronic kidney disease: how much is too much and how to tell?

A new form of IRIDA due to combined heterozygous mutations of TMPRSS6 and ACVR1A encoding the BMP receptor ALK2. Sign in via your Institution. Add comment Close comment form modal. Name Please enter your name.

Affiliations Please enter your affiliations. Comment title Please supply a title for your comment. Comment This field is required.

I agree to the terms and conditions. You must accept the terms and conditions. Read the terms and conditions. Submit Cancel. Thank you for submitting a comment on this article. Your comment will be reviewed and published at the journal's discretion.

Please check for further notifications by email. Comment not saved. Please try again. This feature is available to Subscribers Only Sign In or Create an Account Close Modal. Volume , Issue 1.

Previous Article Next Article. View Metrics. PDF Split View Views. Cite Cite Ewa A. Select Format Select format. ris Mendeley, Papers, Zotero. enw EndNote. bibtex BibTex. txt Medlars, RefWorks Download citation.

Permissions Icon Permissions. Close Navbar Search Filter European Heart Journal This issue ESC Publications Cardiovascular Medicine Books Journals Oxford Academic Enter search term Search. Abstract Iron is a micronutrient essential for cellular energy and metabolism, necessary for maintaining body homoeostasis.

Heart failure , Iron deficiency , Soluble transferrin receptor , Hepcidin , Prognosis , Exercise capacity. Figure 1. Open in new tab Download slide. Figure 2. Major pools of utilized and stored iron in the body. Figure 3. The concept of absolute and functional iron deficiency.

Figure 4. Figure 5. Table 1 Summary of seven studies with intravenous iron therapy administered in patients with heart failure. Studied groups. Iron therapy.

Major results. Inclusion criteria: clinical status. Inclusion criteria: Hb and iron status. Study design. Iron preparation.

Hb and iron status. QoL, HF symptoms. Exercise capacity. CV events. Bolger et al. during 17 days mg i. placebo 20 vs. placebo 24 vs. three times weekly for 3 weeks Maintenance phase: mg iron i. none 27 vs. placebo vs. iron week until repletion dose is achieved Maintenance phase: mg i. Open in new tab.

Figure 6. Iron behaving badly: inappropriate iron chelation as a major contributor to the aetiology of vascular and other progressive inflammatory and degenerative diseases. Google Scholar Crossref. Search ADS. Maternal and child undernutrition: global and regional exposures and health consequences.

Iron deficiency and reduced work capacity: a critical review of the research to determine a causal relationship. Google Scholar PubMed. OpenURL Placeholder Text. Acute reversible heart failure in severe iron-deficiency anemia associated with hookworm infestation in Uganda Africans.

Adaptations to iron deficiency: cardiac functional responsiveness to norepinephrine, arterial remodeling, and the effect of beta-blockade on cardiac hypertrophy. Ultrastructural and cytochemical changes in the heart of iron-deficient rats. Adaptive response of the heart to long-term anemia induced by iron deficiency.

Dietary iron deficiency induces ventricular dilation, mitochondrial ultrastructural aberrations and cytochrome c release: involvement of nitric oxide synthase and protein tyrosine nitration.

The role of correction of anaemia in patients with congestive heart failure: a short review. Anemia and change in hemoglobin over time related to mortality and morbidity in patients with chronic heart failure: results fromVal—Heft.

The impact of new onset anemia on morbidity and mortality in chronic heart failure: results from COMET.

Google Scholar OpenURL Placeholder Text. Anaemia is an independent predictor of poor outcome in patients with chronic heart failure. Anemia is common in heart failure and is associated with poor outcomes: insights from a cohort of 12, patients with new-onset heart failure.

Blunted erythropoietin production and defective iron supply for erythropoiesis as major causes of anaemia in patients with chronic heart failure. Mechanisms of Disease: erythropoietin resistance in patients with both heart and kidney failure.

Randomized double blind trial of darbepoetin alpha treatment in patients with symptomatic heart failure and anemia. Randomized double blind placebo-controlled trial to evaluate the effect of two dosing regimens of darbepoetin alpha in patients with heart failure and anaemia.

Effect of darbepoetin alpha on exercise tolerance in anemic patients with symptomatic chronic heart failure. Google Scholar Google Preview OpenURL Placeholder Text.

Iron biology in immune function, muscle metabolism and neuronal functioning. Mitochondrial mayhem: the mitochondrion as a modulator of iron metabolism and its role in disease. Iron regulatory proteins secure mitochondrial iron sufficiency and function.

Anabolic deficiency in men with chronic heart failure: prevalence and detrimental impact on survival. Energetics and metabolism in the failing heart: important but poorly understood. Towards a unifying, systems biology understanding of large-scale cellular death and destruction caused by poorly liganded iron: Parkinson's, Huntington's, Alzheimer's, prions, bactericides, chemical toxicology and others as examples.

The emerging role of iron dyshomeostasis in the mitochondrial decay of aging. Iron-induced cardiac damage: role of apoptosis and deferasirox intervention.

Iron induces protection and necrosis in cultured cardiomyocytes: role of reactive oxygen species and nitric oxide. Disorders of iron metabolism. Part 1: molecular basis of iron homoeostasis. Ferritin and ferritin isoforms I: structure-function relationships, synthesis, degradation and secretion.

Molecular mechanisms of hepcidin regulation: implications for the anemia of CKD. Modulation of hepcidin production during hypoxia-induced erythropoiesis in humans in vivo: data from the HIGHCARE project. Iron supplementation in the non-dialysis chronic kidney disease ND-CKD patient: oral or intravenous?

Soluble transferrin receptors and iron deficiency, a step beyond ferritin. A systematic review. Serum ferritin concentration and bone marrow iron stores: a prospective study.

Effect of intravenous iron sucrose on exercise tolerance in anemic and nonanemic patients with symptomatic chronic heart failure and iron deficiency FERRIC-HF: a randomized, controlled, observer-blinded trial.

Automated reticulocyte counting: state of the art and clinical applications in the evaluation of erythropoiesis. Evaluation and treatment of iron deficiency anemia: a gastroenterological perspective.

Validation and potential mechanisms of red cell distribution width as a prognostic marker in heart failure.

Red cell distribution width as a novel prognostic marker in heart failure: data from the CHARM Program and the Duke Databank. Red blood cell distribution width and 1-year mortality in acute heart failure.

Adequacy of endogenous erythropoietin levels and mortality in anaemic heart failure patients. Clinical utility of serum soluble transferrin receptor levels and comparison with bone marrow iron stores as an index for iron-deficient erythropoiesis in a heterogeneous group of patients.

Serum transferrin receptor assay in iron deficiency anaemia and anaemia of chronic disease in the elderly. Diagnosis of iron deficiency anemia in the elderly by transferrin receptor-ferritin index. Nutritional status and adequacy of energy and nutrient intakes among heart failure patients.

Impaired expression of duodenal iron transporters in Dahl salt-sensitive heart failure rats. Erythropoietin regulates intestinal iron absorption in a rat model of chronic renal failure. Autonomic imbalance and immune activation in chronic heart failure - pathophysiological links.

Immune modulation: role of the inflammatory cytokine cascade in the failing human heart. Cachexia as a major underestimated and unmet medical need: facts and numbers. An overview of sarcopenia: facts and numbers on prevalence and clinical impact.

Tumour necrosis factor-alpha and the failing heart: pathophysiology and therapeutic implications. The iron-regulatory peptide hepcidin is upregulated in the ischemic and in the remote myocardium after myocardial infarction.

Serum hepcidin is elevated during the acute phase of myocardial infarction. Iron regulatory hormone hepcidin decreases in chronic heart failure patients with anemia. Chronic heart failure leads to an expanded plasma volume and pseudoanaemia, but does not lead to a reduction in the body's red cell volume.

Iron deficiency: an ominous sign in patients with systolic chronic heart failure. Iron deficiency predicts impaired exercise capacity in patients with systolic chronic heart failure. Intravenous iron reduces NT-pro-brain natriuretic peptide in anemic patients with chronic heart failure and renal insufficiency.

Intravenous iron alone for the treatment of anemia in patients with chronic heart failure. Iron deficiency anemia and cardiac mortality in patients with left ventricular systolic dysfunction undergoing coronary stenting. Making the case for skeletal myopathy as the major limitation of exercise capacity in heart failure.

Exercise limitation in chronic heart failure: central role of the periphery. Heart and iron deficiency anaemia in rats with renal insufficiency: the role of hepcidin. The iron regulatory peptide hepcidin is expressed in the heart and regulated by hypoxia and inflammation.

The iron regulatory hormone hepcidin reduces ferroportin 1 content and iron release in H9C2 cardiomyocytes. Expression of the peptide hormone hepcidin increases in cardiomyocytes under myocarditis and myocardial infarction.

Electron microscopic study on the cardiac hypertrophy induced by iron deficiency anaemia in the rat. Capillary growth in anemia-induced ventricular wall remodeling in the rat heart.

Myocyte cellular hypertrophy and hyperplasia contribute to ventricular wall remodeling in anemia-induced cardiac hypertrophy in rats. The effect of iron deficiency on the synthesis of collagenous and non-collagenous proteins in wound granulation tissue and in the heart of rats. Myocardial and systemic iron depletion in heart failure implications for anemia accompanying heart failure.

Strong iron demand during hypoxia-induced erythropoiesis is associated with down-regulation of iron-related proteins and myoglobin in human skeletal muscle. Tissue iron deficiency without anemia impairs adaptation in endurance capacity after aerobic training in previously untrained women.

Marginal iron deficiency without anemia impairs aerobic adaptation among previously untrained women. Iron supplementation improves endurance after training in iron-depleted, nonanemic women.

Iron supplementation improves progressive fatigue resistance during dynamic knee extensor exercise in iron-depleted, nonanemic women. Physiological and biochemical effects of iron deficiency on rat skeletal muscle. Effects of iron deficiency and training on mitochondrial enzymes in skeletal muscle.

Muscle mitochondrial bioenergetics, oxygen supply, and work capacity during dietary iron deficiency and repletion. Iron deficiency in the rat. Physiological and biochemical studies of muscle dysfunction.

Iron supplementation maintains ventilatory threshold and improves energetic efficiency in iron-deficient nonanemic athletes. A pilot evaluation of the long-term effect of combined therapy with intravenous iron sucrose and erythropoietin in elderly patients with advanced chronic heart failure and cardio-renal anemia syndrome: influence on neurohormonal activation and clinical outcomes.

Intravenous iron without erythropoietin for the treatment of iron deficiency anemia in patients with moderate to severe congestive heart failure and chronic kidney insufficiency. Published by Oxford University Press on behalf of European Society of Cardiology. For commercial re-use, please contact journals.

permissions oup. Issue Section:. Download all slides. Views 17, More metrics information. Total Views 17, Email alerts Article activity alert. Advance article alerts. New issue alert.

Receive exclusive offers and updates from Oxford Academic. More on this topic The effect of intravenous ferric carboxymaltose on health-related quality of life in iron-deficient patients with acute heart failure: the results of the AFFIRM-AHF study.

Efficacy of ferric carboxymaltose in heart failure with iron deficiency: an individual patient data meta-analysis. Iron deficiency as a promoter of cardiotoxicity: not only cadmium-induced. Iron deficiency as a promoter of cadmium-induced cardiotoxicity. Related articles in PubMed Grain quality traits within the wheat Triticum spp.

genepool: prospects for improved nutrition through de novo domestication. In situ self-reconstructed hierarchical bimetallic oxyhydroxide nanosheets of metallic sulfides for high-efficiency electrochemical water splitting.

Anemia-related response end points in myelofibrosis clinical trials: current trends and need for renewed consensus. Cost-effectiveness analysis of sodium zirconium cyclosilicate for hyperkalemia among patients with chronic kidney disease or heart failure in Kuwait.

Citing articles via Web of Science Latest Most Read Most Cited Rising stars in cardiology: Floris Heinen, MD. The obesity syndemic in the European community: towards a systems thinking approach for preventive policies.

Cardiac cryptographers: cracking the code of the epitranscriptome. Urgent need to define unmet medical needs in cardiovascular diseases. Advancing cardiovascular research: Gemma Vilahur talks about her pioneering heart health discoveries. More from Oxford Academic.

Cardiovascular Medicine. Clinical Medicine. Medicine and Health. Looking for your next opportunity? Director, Ruth L. and David S. Gottesman Institute for Stem Cell Research and Regenerative Medicine. Assistant Professor. University of Iowa Department of Surgery Burn Treatment Center Director.

Sleep Medicine Neurologist. View all jobs. Twitter YouTube LinkedIn. Online ISSN Print ISSN X Copyright © European Society of Cardiology. About Oxford Academic Publish journals with us University press partners What we publish New features. Authoring Open access Purchasing Institutional account management Rights and permissions.

Get help with access Accessibility Contact us Advertising Media enquiries. Oxford University Press News Oxford Languages University of Oxford. Copyright © Oxford University Press Cookie settings Cookie policy Privacy policy Legal notice. This Feature Is Available To Subscribers Only Sign In or Create an Account.

Maximum mg iron i. iron on Days 15, Okonko et al.

Video

Runner's Anemia: Iron is critical for your running. And other activiites Clara Camaschella; Iron deficiency. Blood Moroccan olive oil 1 : cardiovascualr Total-body absolute iron defifiency is caused by physiologically increased iron requirements in children, Organic pomegranate varieties, young deficuency pregnant women, Athletds reduced iron intake, Natural appetite suppressants by pathological defective absorption or chronic blood Strategies for long-term blood pressure management. Adaptation to iron deficiency at the tissue level is controlled by iron regulatory proteins to increase iron uptake and retention; at the systemic level, suppression of the iron hormone hepcidin increases iron release to plasma by absorptive enterocytes and recycling macrophages. The diagnosis of absolute iron deficiency is easy unless the condition is masked by inflammatory conditions. All cases of iron deficiency should be assessed for treatment and underlying cause. Special attention is needed in areas endemic for malaria and other infections to avoid worsening of infection by iron treatment.

Author: Gakinos

4 thoughts on “Iron deficiency and cardiovascular health in athletes

  1. Sie lassen den Fehler zu. Ich biete es an, zu besprechen. Schreiben Sie mir in PM, wir werden reden.

  2. Ich empfehle Ihnen, die Webseite, mit der riesigen Zahl der Artikel nach dem Sie interessierenden Thema zu besuchen.

Leave a comment

Yours email will be published. Important fields a marked *

Design by ThemesDNA.com