PHYTOACTIVE AS CHELATORS OF IRON WITH THE POTENTIAL TO MITIGATE ITS SIDE EFFECTS AND ENHANCE IRON ABSORPTION
HTML Full TextPHYTOACTIVE AS CHELATORS OF IRON WITH THE POTENTIAL TO MITIGATE ITS SIDE EFFECTS AND ENHANCE IRON ABSORPTION
Shobha A. Udipi *, Jayashree Joshi, Gurmeet Singh, Sujata Sawarkar, Aashish Phadke, Arati Prabhu, Neerja Rastogi and Ashok B. Vaidya
Hon Director Integrative Nutrition and Ayurceuticals, Kasturba Health Society-Medical Research Centre, Vile Parle(W), Mumbai, Maharashtra, India.
ABSTRACT: Iron deficiency anaemia afflicts a large proportion of the human population, compromising productivity and quality of life. Iron supplements are widely prescribed for treating anaemia. However, poor compliance due to side effects, particularly gastrointestinal side effects, remain a hurdle in alleviating the problem, leading to unabated high prevalence. Thus, a fresh look at novel approaches to supplementation is essential. Iron toxicity is attributed to the Fenton Reaction and generation of reactive oxygen species leading to oxidative stress. Several studies suggest that the FR and ROS caused by excess iron, results in chronic inflammation, leading to metabolic syndrome, diabetes mellitus, atherosclerosis, cancer, neurodegenerative disorders. Iron chelates have been developed as treatment modalities to reduce free radical generation, reduce GI side effects, and enhance absorption. Classical Fe chelators, developed to counteract iron overload/ toxicity, cannot be used in IDA, due to severe side effects. In Ayurveda, iron formulations are prepared using phytomolecules that chelate iron and/or reduce OS. These phytomolecules are metabolized by the gut microbiota, which plays an important role in oxidative stress, release of inflammatory cytokines and GI side effects. The antioxidant, anti-inflammatory and antimicrobial actions of bio-active phytomolecules of Ayurvedic plants highlight their potential for reducing possible oxidant damage and side effects associated with iron supplementation. The present communication summarises the role of reactive oxygen species, potential of oxidant damage in causing side effects and microbial dysbiosis due to iron. Management of ‘Pandu’ an anaemia - like condition described in Ayurveda and use of selected medicinal phytoactives are reviewed.
Keywords: Anemia, Iron deficiency, Bhasmas, Phytoactives, Polyphenols
INTRODUCTION:
Prevalence of IDA, Causes of Stagnation in Incidence of Anaemia: Anaemia is a highly prevalent persistent problem with potentially serious consequences Fig. 1 1, 2.
The principal cause of anaemia is Iron Deficiency (ID) and some vitamins and protein most commonly. Functional and economic consequences of ID are lower work capacity and productivity, reduced immunity and increased susceptibility to infections.
In children, ID retards growth, cognitive development and endocrine maturation. These consequences impact strongly on the health and economic status of a nation 1, 3. Globally, anaemia is prevalent in more than 20%of the population in 132 of 159 WHO-member countries 3.
ID is not restricted to middle- and low- income countries, as global statistics, indicate that anaemia prevails in industrialized and developed nations 4. Globally, approximately 1.6 billion is anaemic and with the huge numbers of those affected, the disease puts an immense burden on global health 1, 5. Across nations, in various sub-populations and socio-economic groups, the majority of those affected are women of reproductive age and children. Anaemia during pregnancy can lead to complications, contributing substantially to maternal and neonatal morbidity and mortality 6. Since most cases of anaemia are due to nutritional iron (Fe) deficiency, efforts have been made to control and prevent ID using iron supplements (IS). IDA is considered as one of the most expensive diseases due to lost productivity, and the large numbers affected; 4, 7 its effect on overall health and earning potential at family level, productivity and impact on the economy at community and country level. Since the consequences of anaemia for an individual and an entire country can be devastating, prevention and control of anaemia by 2025 is one of the global targets 6, 7, 8, 9. IDA results from not only Fe deficiency but also from haemorrhagic blood loss or diarrhoeas Fig. 1. Therefore, IS is needed to replenish body Fe and ensure adequate stores, whatever the cause. Towards the end of the 20th century, it was evident that Fe deficiency, with or without anaemia, needed to be addressed seriously as a major health problem. Despite constant efforts to overcome this serious but surmountable health problem, its prevalence has not reduced significantly in decades. In India, too, National anaemia prophylactic programs and Fe supplementation programs have been implemented by the Government since 1970, yet anaemia persists as a public health problem 10, 11. According to the IVth National Family Health Survey, more than 50% of women and children and about one-fifth of men were anaemic 4.
FIG. 1: CAUSES AND CONSEQUENCES OF IRON DEFICIENCY ANAEMIA
Prevalence has hardly decreased, as per the NHFS-5 report and the report of the Ministry of Health and Family Welfare (2019) 10. The Comprehensive Nutrition Survey 11 indicated that 41% of pre-schoolers, 24% of school-age children, and 28% of adolescents were also anaemic, with a higher prevalence among female adolescents (40%) than their male counterparts (18%).
Major Reasons for Persistence of Anaemia as a Public Health Problem: The major reasons why anaemia persists as a public health problem are: a) population growth with unequal distribution of resources- poverty- Fe deficient nutrition, b) relative illiteracy in women, poor knowledge about adequate nutrition in the whole population, c) inaccessibility of health resources, d) side effects of Fe and discontinuation of IS, as continuation rates for therapy may be less than 50 % 12 and e) failure in some cases to identify metabolic defects in Fe metabolism and treatment of all cases of anaemia as ‘Nutritional /IDA’.
While the first three causes are a function of policy makers and the social distribution systems, the fourth factor warrants analysis and can be remedied within the context of current scientific advances. The fifth factor is very difficult to detect on a mass scale with the current technology but may play some role in IDA management.
Side Effects of Iron of IS/Fe Therapy: First-line treatment of IDA is oral therapy with ferrous Fe salts, but a substantial proportion of patients suffers from gastro-intestinal (GI) side effects, resulting in non-adherence and treatment failure. Common side effects include constipation, dark stools, stomach pain, nausea, and vomiting, whereas diarrhoea, heartburn, urine discoloration, and teeth staining are less common 13, 14.
Side effects of oral IS, mainly related to the gastrointestinal (GI) system, are particularly troublesome during early pregnancy, which is inherently associated with nausea and vomiting. Hence, pregnant women are generally not given oral IS during the first three months unless the anaemia is severe.
The National policy prescribes routine oral IS for at least 3 months during pregnancy, given the high prevalence among pregnant women in India. However, a majority of pregnant women probably do not complete the treatment due to the side effects of IS and almost 35% of them could be noncompliant 8, 15, 16.
To overcome noncompliance, intermittent vs. daily Fe supplementation has been tried, and the side effects with the latter were less 17, 18. A meta-analysis of 2133 full texts by Tolkien 19 indicated that even in women without prior GI disease, different oral Fepreparations /IS can cause side effects (constipation, nausea, and diarrhoea) in 32-47% of women.
Side effects are reported in children also 20. In India, too, programs have not succeeded in addressing this huge problem.
Even the minor side effects of Fe interfere with the daily life of more than a third of the population treated with oral Fe. Some of these side effects can be counteracted through modifications in formulation development.
The most common approaches have been oral supplements with organic or inorganic Fe compounds to increase bioavailability and to reduce side effects. But some of these may be expensive or not very effective in reducing the side effects and have mostly not been made available in public programs.
This highlights the need to focus attention on side effects' cause (s) and investigate more acceptable alternative formulations that are also effective substitute (s) for current oral IS.
Iron, an Essential Mineral in All Body Functions: Iron is a constituent of at least 27 essential proteins and enzyme complexes in the human body that are important for cellular function Fig. 2 21, 22.
The plasma Fe pool of 3-4 mg is recycled manifold to meet the body’s daily demand of about 20-25 mg for several functions, including erythropoiesis 23. About 30% Fe is the storage compartment, with 20% being present in ferritin.
Fe is commonly present in the divalent ferrous (Fe2+) and the trivalent ferric (Fe3+) states in the body. Fe contributes to haemoglobin (Hb) and myohemoglobin (Myb) which are vital for circulatory and locomotive functions. Fe is essential for the central nervous system. Fe-containing proteins that contain about 5% of body Fe are needed for various processes Table 1. Besides Hb and Myb, major Fe-related protein are catalase and peroxidase participate in oxygen metabolism; cytochromes are involved in electron transport and mitochondrial respiration.
FIG. 2: ROLE OF IRON IN CELLULAR FUNCTION
Several non-heme proteins are essential for DNA synthesis, cell proliferation and differentiation, gene regulation, drug metabolism and steroid synthesis. Transferrintransports Fe for erythropoiesis, to muscle cells and T- and B-lymphocytes. Fe-sulfur clusters are non-heme essential cofactors with a role in electron transfer, substrate binding/activation, Fe/sulfur storage, regulation of gene expression, and enzyme activity 24. Three other enzymes - hephaestin, ferroportin, and hepcidin, do not contain Fe but are involved in Fe transport, uptake or release. A metabolic defect in any of these enzymes may cause Fe insufficiency even when dietary intake is adequate. IS in these cases may correct the anaemia but may lead to undesired Fenton reaction (FR) and side effects. This has been reported for Iron Resistant IDA that is refractory to oral IS 12, 26.
TABLE 1: IRON-CONTAINING PROTEINS IN THE BODY ACCORDING TO THE LOCATION OF THE IRON-CONTAINING PROSTHETIC GROUP
Iron-containing prosthetic group | Names |
Heme | Hemoglobin, myoglobin, cytochromes, cytochrome P450, Tryptophan 2,3-dioxygenase, peroxidases, catalases, cyclooxygenase (8) |
Amino acids | Ribonucleotide reductase, proline hydroxylase, Phenylalanine hydroxylase,Homogentisic acid 2,3-dioxygenase, lipoxygenase, cyclooxygenase, transferrin, lactoferrin (8) |
Iron-sulphide containing Fe-S complexes | Adrenodoxin, aconitase, succinate dehydrogenase, NADH dehydrogenase, xanthene oxidase, aldehyde oxidase (6) |
Oxyhydroxide, Phosphate Fe | Ferritin, hemosiderin (2) |
Adapted from Konteghiorghes (2020) 22
Iron Absorption: Since, the human body lacks physiologic regulatory mechanisms for Fe excretion, maintenance of Fe homeostasis is almost entirely regulated via Fe absorption. Systemic Fe stores are regulated largely in the GI through Fe absorption, homeostasis at systemic and cellular levels, and storage. The daily loss of1-2 mg Fe through desquamation of intestinal cells, skin, minor blood losses is generally restored via intestinal absorption that is influenced by: (i) body Fe stores, (ii) hypoxia, and (ii) the erythropoietic rate. Dietary Fe is present as haeme and/or non-haeme Fe, with their sources and absorption mechanisms being different Fig. 3A and B.
Fe absorption from oral supplements consumed with and without food ranges from 2- 13% and 5-28%, respectively, in subjects with IDA 26 Fe absorption and its transport across the gut membrane is tightly controlled by hepcidin [Hcp] 27.
Hcp inhibits the Fe transporter/exporter protein, ferroportin (FPN1), which mediates the transport of gut Fe through enterocytes into the bloodstream. Daily administration of Fe supplements acutely increased plasma Hcp levels 28-29 and significantly reduced absorption of supplemental Fe from the intestinal lumen. In contrast, dose reduction and alternate-day dosing favoured better Fe absorption 30 Fig. 3A and B. Absorption is influenced by the body’s requirements Fig. 3B. If a dietary Fe bolus is available and intracellular Fe levels are higher, the gut signal makes absorptive enterocytes resistant to acquiring Fe.
Thus, DMT1 expression is suppressed. Low Fe stores send a signal to stimulate and enhance absorption. Under normal conditions, once the Fe stores are replenished, the uptake returns to basal levels. Erythropoiesis modulates Fe absorption.
FIG. 3A: FIVE-STEP PROCESS OF IRON ABSORPTION IN INTESTINES
Storage and Recycling of Iron: Senescent/ damaged erythrocytes are phagocytosed by macrophages, and the haeme Fe is recovered byhaeme oxygenase for utilization, storage/ recycling. FPN1 has specific functions in Fe homeostasis is expressed in high amounts by enterocytes, macrophages, hepatocytes and trophoblasts. In macrophages, FPN1acts with ceruloplasmin / haepahaestin to release the ferric Fe to transferrin.
FPN1 is upregulated by haeme but down-regulated by inflammatory cytokines for sequestering Fe. Fe represses FPN1 translation.
FPN1 deletion resulted in haemolytic anaemia because of the toxic effects of Fe resulting from its oxidation in Hb, particularly because erythrocytes do not have much antioxidant capacity 31.
DMT1 is required for transport from vacuoles into the cytoplasm. In macrophages, Nramp1 plays an important role 32. Delivery of Fe to cytosolicferritin, needs poly (RC)-binding protein 1 32. That can canstore up to 4500 Fe atomsin its mineral core, protecting against oxidative damage.
In the gastrointestinal tract (GIT), it can regulate Fe absorption, can convert the Fe3+ to the Fe2+ form, and prevent Fe overload.
Management of IDA: Conventional Supplements and Fortification:
Biomarkers for Assessment: Hb is generally used to diagnose anaemia, but Hb levels tend to be normal in subclinical Fe deficiency. Hb reflects IDA only in the later stages 33. Also, it does not respond rapidly to Fe therapy/IS.
WHO 33 recommends the use of soluble transferrin receptors and serum ferritin. However, inflammation or Fe overload elevate ferritin levels because it reflects macrophage ferritin content 21, 35.
Therefore, inflammatory biomarkers need to be measured along with serum ferritin, to assess Fe status of when Fe bioavailability or toxicity are studied in-vivo or in-vitro using cell lines 36, 39.
FIG. 3B: SCHEMATIC DIAGRAM OF ABSORPTION AND TRANSPORT OF IRON. Refs34-37, (STEAPs) -Six Transmembrane Epithelial Antigen of the Prostate Proteins
Iron formulations for Supplementation: Commercial Fe formulations are generally in two salt forms-ferric (Fe3+) and ferrous (Fe2+), or as aminoacid chelates, carbonyl Fe, polysaccharide-Fe complex, as single or combination and extended-release products Table 2. The objective of treating IDA is to restore Hb levels, RBCs indices and Fe stores. For effective absorption and optimum bioavailability, Fe3+ has to be reduced to the Fe2+ form that is absorbed three times more readily than Fe3+. FeSO4 (200mg twice daily) is the simplest and cheapest choice for initiating therapy. However, dose adjustment may be required for persons who cannot tolerate this dose. Instead of FeSO4, better-tolerated salts like ferrous fumarate, ferrous gluconate, and ferrous ascorbate may be used, the latter being best tolerated 40. Once the deficiency is corrected, oral Fe therapy should be continued to replenish Fe stores. Ascorbic acid (250-500mg) may be used to enhance Fe absorption; however, its effectiveness in improving Hb is not yet established 40-42. Patients who are unresponsive or intolerant to oral Fe therapy can be administered parenteral formulations like Fe3+carboxymaltose, Fe sucrose, Fe3+ hydroxide dextran either intravenously or by deep gluteal intramuscular injections. Fe sucrose is reasonably well tolerated compared to other salts when given by IV route, with a low incidence of adverse events. IV iron dextran can replenish Fe stores and increase Hb levels in one infusion, but serious adverse events, including anaphylactic reactions, may be fatal in some cases 43. Also, this approach is not suitable for community programs as hospitalization is required. Parenteral Fe can increase oxidative stress (OS) and lead to cardiovascular complications in rare cases 44. Oral IS results in GI side effects (vide supra) that include erratic Fe absorption due to the inability to localize Fe at the site of its maximum absorption. There is a need to design suitable forms of Fe or delivery system(s) that can deliver and help restore Hb with minimal side effects. Controlled release/ enteric preparations deliver Fe past the duodenum and jejunum, but they are poorly absorbed, and there is no substantial evidence that they fully alleviate side effects 45.
Fortification of Foods with Iron: Selecting a suitablefortificant and a food vehicle that will not diminish Febioavailability is important, as is minimizing undesirable organoleptic changes since some compounds tend to interact strongly with food components. Fe availability differs for different for toxicants such as those listed in Table 2, and others likeFe2+lactate,Fe3+pyrophosphate, NaFe EDTA, monosodium Fe3+EDTA, Fe2+bisglycinate, Fe2+succinate, Fe3+ ortho / pyrophosphate and elemental Fe powders.
The amount used will vary with the compound. If Fe3+ pyrophosphate or electrolytic Fe are used, the amount needed is about twice the amount of FeSO4, whereas a comparatively smaller amount of NaFe EDTA is needed, as it has higher relative bioavailability 40, 42, 45, 46, 48. Worldwide, several foods, e.g., cereal flours, bread, pasta, salt, infant foods, milk, and yogurt, are fortified. For commercial foods, the amount of bioavailable Fe per serving generally provides 15-30% of the recommended daily allowance, or 7-8 mg Fe/day, with no risk of exceeding the Upper Limit of daily Fe intake 49. Fe absorption from water-soluble Fe compounds and those completely soluble in gastric juice is similar to dietary Fe.
However, from the haematological perspective, universal Fe fortification of food may be problematic, notably for individuals with haemochromatosis and other Fe loading diseases. Also, its detrimental effects on gut microbiota need to be seriously considered 49, 50.
TABLE 2: COMMON FORMS OF CONVENTIONAL IRON SUPPLEMENTS, THEIR ADVANTAGES, AND DISADVANTAGES
Iron supplement | Elemental
Fe (%w/w) |
Dosage
Forms |
Advantages | Disadvantages |
Carbonyl iron | 100 | Tablets,
Chewable tablets, Suspension |
Highest amount of elemental Fe
Good solubility in gastric fluid, toxicity index of this form is less |
Sometimes: diarrhea, constipation, nausea, vomiting, stomach pain, tooth discoloration |
Fe3+ ammonium citrate | 18 | Capsules | Most commonly used Fe supplement
Toxicity is low |
Bioavailability is less, has to be reduced to Fe2+form before absorption |
Ferrous sulfate
(FeSO4) Is the oral soln a suspension or ?? |
20
|
Oral solution (su, tablets, Enteric-coated tablets, Film-coated tablets | Most commonly used
Easily available Most economical |
Tolerance is poor, Gastrointestinalside effects common |
Fe2+fumarate + Fe2+asparto glycinate – chelated iron(2) | 33
|
Tablets,
Chewable tablets |
Better palatability as it is tasteless
Efficacy equivalent to FeSO4, well tolerated |
Sparingly soluble in water
|
Ferrous gluconate | 12 | Tablets, Intravenous | Efficacy equivalent to FeSO4, Well tolerated | Causes gastrointestinal distress to some extent. |
Polysaccharide -Fe complex – Fe Carbohydrate complex,Fe3+Carboxymaltose, Low mol. wt Fe -dextran complex, Fe polymaltose, Sodium Fe3+gluconate Fe sucrose, Fe3+ Carboxymaltose, Ferumoxytol polyglucose sorbitol carboxymethyl ether, Feisomaltoside | 100 | Capsules, Solution,
Film-coated tablets, Intravenous |
Ferric iron is complexed to
hydrolyzed starch, making it tasteless and odorless. Bioavailability comparable to FeSO4 |
Safety and efficacy of different Fe –carbohydrate complex administered intravenously depends on different carbohydrate coatings, and their pharmacokinetic profile (3) |
Sources 44-47 |
Consumers' use of multiple fortified foods can theoretically result in over ingestion of Fe with possible side effects 51. Hence, food fortification and distribution may need to be supervised.
Possible Causes of Side Effects of Iron: Oxidative Stress and Inflammation: Until recently, GI side effects were thought to be simply a unique function of a particular drug or host response. Eliminating these did not receive much attention, perhaps because they were considered unavoidable, and the benefits likely outweighed the side effects. However, studies on diseases of Fe overload e.g., hemochromatosis orhaemoly-ticanaemias, usually of genetic origin, have identified the specific toxicity caused by Fe. High circulating Fe levels in these diseases were associated with Fe deposition in various tissues, including the central nervous system, kidney and liver and high morbidity and mortality due to these complications. The site of Fenton reaction (FR) and Fe toxicity, resulting from the excess of free Fe, is located at the tissue level.
The treatment requires parenteral administration of strong Fe chelators. However, these chelators are associated with side effects and complications; hence the treatment needs to be done in special centers under medical supervision. Also, this does not apply to IS side effects encountered in public health programs. The unabsorbed, excess free Fe in the GI tract is highly reactive and can directly mediate the formation of reactive oxygen species (ROS), generated through the Fenton and Haber–Weiss reactions 52, 53. The FR and the resultant ROS are major sources of oxidative stress (OS). Some OS is needed for several essential biological enzymatic reactions, but a balance must be maintained because excessive free radicals can damage tissues. The FR and Fe toxicity occur at the intestinal luminal level and the tissue level, particularly in the liver, heart, pancreas, and brain, which have high levels of oxidative metabolism and are susceptible to ROS damage.
Mechanisms and Consequences of FR - Oxidative Stress and Inflammation: In 1894, HJH Fenton showed that metals like Fe and copper react with hydrogen peroxide (H2O2). They catalyze enzymatic reactions to produce highly reactive free radicals like superoxide(O2.-), and hydroxyl radicals (OH.), which can immediately react with other macro- and micromolecules they come in contact with, ultimately damaging cell function and structure. ROS are by-products of mitochondrial metabolism through the electron transport chain and the cytochrome P450 system Fig. 4. NADPH oxidases in phagocytes and endothelial cells 54 are intricately involved in the genesis of the inflammatory response 55 and are responsible for ROS generation. The major ROS include O2.-, H2O2, hydroxyl (OH-) anions, OH., peroxynitrite (ONOO-), and hypochlorous acid (HOCl). One-electron reduction of oxygen O2, mediated by NADPH oxidase/xanthine oxidase (XO,) uncoupled endothelial nitric oxide synthase (eNOS), or mitochondrial electron transport chain (ETC); results in O2. - Formation 55-57 Fig. 3. The reaction starts with the donation of an electron and the conversion of the ferrous to the ferric form. The ferric salt is reconverted to the ferrous form with the Haber–Weiss reaction resulting in a chain reaction, wherein the reaction of H2O2 and superoxide ion (O 2 • −) catalyzed by Fe, generates hydroxyl and hydroxide ions.
FIG. 4: MECHANISM OF FENTON REACTION, INTRACELLULAR & INTRALUMINAR ROS RELEASE IN INTESTINE
Intracellularly, ascorbate can replace O2•- and reduce Fe 3+ to Fe2+.Superoxide O2.- a short-lived species can spontaneously dismutate to H2O2, a reaction accelerated by superoxide dismutase 55, 56. It can also react with Fe3+ in the Haber-Weiss reaction to produce again Fe2+, thereby affecting redox cycling. H2O2 is scavenged by catalase, glutathione peroxidase or peroxiredoxins and converted to H2O and O2.At inflammatory sites where polymorph nuclear neutrophils (PMN)are abundant, PMN-specific enzyme myeloperoxidase mediates the reaction between H2O2 and chloride and generates the highly reactive HOCl58. O2.- rapidly reacts with nitric oxide(NO) to form the highly reactive ONOO-. This reaction itself does not cause OS toxicity, but it paves the way for the FR, where Fe2+ triggers the conversion of H2O2 to the highly reactive OH-radical and OH. Anions that are potent oxidizing species of biological membrane proteins and lipids 55, 58-60. They interact with the surrounding biomolecules, in four ways: addition, hydrogen abstraction, electron transfer and radical interaction, consequent to which a cascade of reactions occur, with Fe-induced oxidative damage. The GIT is no exception to these effects of the unabsorbed/free Fe.
As intracellular Fe is in close proximity to DNA and other organelles, these highly reactive intermediates interact with DNA, alter its structure and function, cause polymerisation and deletions, translocations, or irreparable mutation. Modifications of various protein or sugar moieties, denaturation of proteins and proteolipid molecules, and abnormal cross-linkages, lead to cell death, apoptosis or autophagy. Proteolipid precipitation can form insoluble structures e,g, lipofuscin that can deposit as plaques 59-61. Also, the intracellular events can damage microsomes, and activate Nuclear Factor Kappa-β, genes, and several other intracellular proteins with cross-linkages, oxidation, and proteolysis. Membrane damage can occur, leading to an imbalance in calcium homoeostasis, activation of Ca - dependent proteases, end nucleases, lipases, ATP depletion, NADP (H) and ultimately cell destruction 62. Lipid peroxidation can occur, damaging the bilayer cell membrane and membrane-bound structures. The highly reactive lipid peroxidation products can cause cross-linkages and inactivate several physiological enzymes 19, 61, 62.
Further, transmembrane proteins, such as voltage-dependent anion channels (VDAC) in mitochondria, permit the transmembrane passage of the negatively charged superoxide O2.-, although it has limited permeability across membranes. H2O2 can cross biological membranes facilitated by aquaporin channels such as AQP3 and AQP8. Thus, superoxide and H2O2 can spread through cells that are in contact and the ROS-generating oxidative pathways in tissues distant from their point of origin 63-65.
Iron Supplementation and ROS: In the past, the FR was considered significant in pathological complications associated with Fe overload and Fe storage diseases like haemolytic anaemias, and liver and spleen diseases. However, subsequent reports indicated toxicity accompanying the use of therapeutic Fe load or oral IS used for treating IDA. The observations that a significant proportion of IDA patients discontinued the treatment due to GI side effects led to the suspicion and later confirmation of the possible role of Fe during Fe replacement therapy.
The excess Fe from the diet or supplements makes the GIT vulnerable to ROS that is also produced by intestinal immune cells and intestinal flora. The GIT has the highest concentration of xanthine oxidase, and numerous phagocytic cells generate considerable amounts of O2·− 66. Daily Fe supplementation in mildly anaemic women led to significant increase in OS 67, 68. A 40% increase in free radicals was detected in the faeces of healthy human volunteers who consumed Fe supplements (98 mg of Fe as FeSO4/day for 70 days) 64, 69. Ethane exhalation in breath also increased, and plasma malonaldehyde levels were correlated with the increase in plasma Fe and ferritin. Even a single oral dose of 80 mg FeSO4significantly increased lipid peroxidation in healthy volunteers with a high probability of oxidative damage in the small intestine 70. In 72 healthy adults and children who exercised daily, Fe status, creatine kinase activity(CK), and redox status were assessed at baseline, before, and 72 h after exercise. The experimental group was treated with 35 mg of elemental Fe for 3 weeks before assessment. ROS increased in adults and in children, while CK activity increased only in adults. Fe concentration and transferrin saturation increased after Fe therapy. The authors proposed that higher doses or longer duration of IS may further increase the ROS that may lead to side effects 71
Oxidative Stress, Iron Deficiency Anemia and Chronic Diseases: The conditions that give rise to Fe deficiency also promote OS 72. When there is infection and/or inflammation, the body tends to absorb less Fe to withhold it from the pathogens that require Fe 73. OS occurs in many chronic diseases in which Fe deficiency occurs, wherein FPN1 is negatively regulated by hepcidin, possibly leading to Fe deficiency 74. Additionally, the hypoxiainduced by anaemia may worsen OS, via pro-oxidant changes leading to increased free radical production, altered cellular metabolism, increased catecholamine metabolism, and leukocyte activation 75. IS can help in Hb production and build up iron stores; however, it is important to manage any underlying condition that could negate the benefits of IS therapy. In this respect, antioxidant therapy can ameliorate anemia-related oxidative damage 76. Even a small proportion of excess Fe can activate the FR and ROS-induced damage. Even if it is asymptomatic, there can be risk of chronic diseases over time, resulting from the direct toxic effect of Fe from the FR and secondly due to dysbiosis and other causes. Chronic diseases include premature aging 77, metabolic syndrome, diabetes mellitus 78 Alzheimer’s disease 79, Parkinson’s disease 80, inflammatory bowel disease 81 non-alcoholic fatty liver disease 82, cancer 83 cardiovascular disease 84 and arthritis 85. Ferroptosis i.e. cell death and necrosis resulting from Fe toxicity has been reported 21, 22, 86, 87.
Inflammation: Enzymes with Fe complexes are vulnerable to disintegration when there is Fe - generated ROS. Disruption of the antioxidant/ oxidant balance and excess ROS lead to changes in membrane permeability, activation of NF-kB, production of many proinflammatory cytokines e.g., IL-6, Tumor Necrosis Factor (TNF)-α, extracellular kinases that are mitogen activators, can activate protein kinase C, generate other stress proteins and disrupt multiple pathways. Ultimately, Fe excess can lead to cell death and inflammation. Aseptic inflammation of intestinal luminal cells occurs, resulting in the production of toxic products responsible for the side effects 86, 87.
In rodent models, Fe overload-induced ROS caused inflammation and several histopathological alterations in the duodenum and ileum, impaired cellular function, cell turnover, shortening of microvillus height, and partial to complete erosion of the duodenal microvilli. Even a single oral dose of 8 mg Fe, led to apoptosis of GI mucosal cells and necrosis of the GIT absorptive surface 88.
Fe toxicity increased infection, inflammation, and Fe deposition in the duodenal villi tips 89. In rats, administration of ferric citrate (2.5, 5 or 10 mg/day) for 16 weeks resulted in body Fe overload, decreased jejunal villus height, the ratio of villus height to crypt depth, the number of intraepithelial lymphocytes and goblet cells 90. Proinflammatory cytokine levels increased while anti-inflammatory cytokines and sIgA declined. Malondialdehyde, protein carbonyl, and serum lactate increased, while tight junction proteins like claudin-1, occludin, ZO-1, MUC-2, TFF3, and glutathione and antioxidant enzymes like superoxide dismutase, glutathione peroxidase decreased. These alterations reflected intestinal immune and barrier function impairment attributable to ferric citrate-caused OS 91.
However, different Fe compounds may vary in their effects. Liquid ferrous gluconate was found to be comparatively safe, whereas Fe polysaccharide was associated with diarrhea 90, 91. In children, effects of Fe2+or Fe3+ administration over 6 months were compared on erythrocyte malondialdehyde, urine 8-isoprostane, oxidized LDL, superoxide dismutase, catalase, and glutathione peroxidase activities. In the Fe3+ group, superoxide dismutase levels and oxidized LDL levels were higher at the end of the 1stand 6th months, respectively 92.
Since, dietary Fe is poorlyabsorbed–maximum of 20% among meat eaters and only 10% in vegetarians; oral Fe preparations contain more than five times the amount actually absorbed. Daily supplemental doses of up to 200 mg have been used, although the estimated average requirement for adults is only 11-32 mg. With 200 mg, side effects like abdominal cramps, diarrhoea or constipation, and an increase in C-reactive protein (CRP) were seen. Even with a lower dose of 100 mg, side effects have been reported 93, because the unabsorbed Fe (80-90% of a single dose) is highly reactive and mediates ROS formation through the Fenton and Haber–Weiss reactions 93, 94, causing damage and inflammation during its transit in the GIT. Also, this unabsorbed Fe promotes the growth of pathogenic intestinal microbes 95, 96, resulting in intestinal inflammation, leading to a double assault.
Inter play between Iron and the Gut Microbiome: The alteration of the intestinal microbiome by excess Fe and its effects warrant attention. The commensal organisms in the human GIT protect against opportunistic pathogens, synthesize nutrients like vitamins B12, K, metabolize undigested substances, particularly using prebiotics as substrates, and produce short-chain fatty acids (SCFA) that provide energy to the intestinal cells. Also, they contribute to the development of the intestinal structure and functioning of the immune system 97, 98.
Gut microflora plays an important role in Fe metabolism. Numerous intestinal organisms need Fe for their replication, development, and metabolism. Colonic microbiota can shift the valence state of Fe and sequester Fe through siderophore production to meet their needs 66. The viability of gut microbiota was favored by both ferrous and ferric forms of Fe 66. Since, only about 10-15% of dietary Fe is absorbed in the duodenum, much of the unabsorbed Fe, particularly from supplements, is available to the resident colonic microflora and can cause changes in the intestinal microenvironment. The organisms can produce harmful metabolites that may damage the lumen or be absorbed into the circulation, reach various tissues and increase the risk of chronic diseases 98, 99.
In germ-free mice, in the presence of gut microbiota, the intestinal cells acquired an ability to store Fe in ferritin, and Fe transport was favoured by increased FPN1expression 100. Propionic acid, a SCFA produced from substrates like prebiotics in the colon has been shown to enhance luminal Fe absorption101. Lactic acid-producing bacteria enhance Fe absorption whereas other types of bacteria do not, suggesting that the type of organism may influence the risk of IDA. Fe can increase the replication and virulence of gut pathogens like Salmonella, Shigella, Campylobacter, because it is important for their gene expression 102-104. Fe status may influence the survival of Salmonella in host intestinal epithelial cells105. A similar phenomenon has been observed in the infectivity of different organisms residing in macrophages 104, 105. Thus, Fe can influence the infectivity of pathogens and modify the gut microbiota through metabolic changes in the colonic lumen 66, 98. Parmanand et al., 98 reported that Fe availability reduced through chelation led to significantly reduced growth of E. coli, S. typhimurium, B. thetaiotaomicron and B. longum. In contrast, a decrease in the beneficial barrier commensal gut bacteria e.g., Bifidobacteria and Lactobacilli have been observed in Fe supplemental states. This is further worsened by an increase in enterobacteria, including enteropathogenic Escherichia coli 95, 96.
The host immune system responds to harmful pathogenic bacteria by inducing varying levels of gut inflammation. This is often manifested as diarrhoea, in many adults, but especially in vulnerable populations such as infants and children are given Fe supplements 106, 107. Young children in the first two years are vulnerable to the adverse effects of IDA on cognitive, motor, social-emotional, and neurophysiologic development 107, 108. IS benefits cognitive performance in school-aged children 108, 109. WHO 110 recommends Fe supplementation as a public health preventive measure, for infants and children aged 6-59 months and for school children living in settings where anaemia is highly prevalent. However, in supplemented children, Fe supplements have been associated with hospitalization and mortality 96.
In a placebo-controlled clinical trial, African children (1-35 months old) who were at risk of complications of anaemia in addition to malaria were treated with prophylactic Fe+ folic acid supplements and followed up for hospital admissions and mortality 111. This study was discontinued because the interim analysis showed that hospital admissions and deaths were more common in the group receiving Fe + folic acid supplements and indicated the adverse effect of Fe in promoting inflammation of the bowel and other organs. Therefore, children with IDA may benefit from IS, but those with adequate Fe intakes may be affected by Fe toxicity even in the absence of Hemoglobinopathy or liver metabolic disorders e.g., Hepatosplenomegaly- Haemochromatosis. Increased morbidity and altered gut microflora in Fe - replete infants and young children have been noted 111, 112. A large meta-analysis found that although oral IS may contribute to general health, stamina, cognition, and brain health, it did not promote linear growth, in contrast to a positive effect seen with zinc supplements 112.
Sachdev et al., 114 and Pasricha et al., 115 in their systematic reviews on the effect of IS on physical growth in children, indicated that there was no significant, positive effect on anthropometric indices and physical growth, although the daily Fe supplement reduced anaemia. Children who received Fe had slightly lower length and weight gain and their risk of vomiting and fever was likely to be more 115, 116.
Such possibly differential effects of IS on Fe-replete, well-nourished children and on malnourished children warrant closer inquiry. Nchito et al.116 showed that African schoolchildren givenoral Fe supplements had increased small intestinal permeability that may allow easier translocation of pathogenic bacteria across the gut wall, with severe, bloody diarrhea in some cases. In a 10-year follow up study of Chilean children who were fed Fe-fortified (12.7 mg/l) or low-Fe formula (2.3 mg/l), from 6 -12 months of age, the Fe- supplemented children were found to have lower scores for IQ, arithmetic, visual perception and motor coordination. In Cambodian primary school children, consumption of micronutrient fortified rice (including Fe) was associated with a higher risk of hookworm infection 117.
Thus, the addition of Fe to the diet or Fe supplementation in malaria-endemic areas has to be weighed against the risk of increased incidence and severity of malaria, as the parasite requires Fe for its growth 117. Similarly, other intestinal pathogenic bacteria also show overgrowth in the presence of excess Fe, possibly leading to direct intestinal bacterial infections. Overgrowth of pathogenic bacteria can also cause OS and produce undesirable toxic products like sulphur dioxide, which can irritate the luminal cells and result in abdominal colic or diarrhoea118, 119. The human microbiome is altered with age, sex, diet, antibiotic treatment, hormonal dysfunction, and obesity. An altered microbiome has emerged as an important determinant of metabolic syndrome that predisposes many individuals to cardiovascular diseases 120, 121 hence, the role of Fe in dysbiosis bears scrutiny. In this context, plant components/ bioactive could be potentially useful. Those with antimicrobial properties could help in modulating the gut microbiome. Also, they could serve as anti-oxidants, chelate Fe, and help to attenuate the FR, reducing ROS. The following section discusses the role of plant/plant-based products and phytoactives.
Concepts in Traditional System of Ayurved: ‘Panduroga’has been described in ancient Ayurvedic texts (5 - 6 BC) 122‘Pandu’ is characterized by yellowish-white skin colour of anaemic patients. Ayurvedic texts describe numerous symptoms like generalized weakness, fatigue, lethargy, loss of normal complexion, loss of appetite, giddiness, body ache, fever, breathlessness, palpitations, irritability, ringing in the ears, leg cramps etc., that are akin to those described for ID in modern times. ‘Panduroga’ was treated with different ‘bhasmas’that are essentially metallic/mineral preparations made by special pharmaceutical processing and other measures. These formulations have been used extensively for centuries by Ayurvedic physicians and are still used presently by Vaidyas to manage Panduroga (IDA) 122. Different pharmaceutical processes and medicinal plants are used for preparing different bhasmas. Various texts describe numerous formulations to be used according to the type of patient, cause of anaemia, associated conditions, and complications 123. In case of Fe, there are more than 300 preparations, about 290 are based on iron oxide nanoparticles i.e., Lauhabhasma, approximately 80 formulations are based on iron pyrite, ferrous sulphate, and ochre 123. Four preparations are quite commonly used: Loha/Lauha Bhasma, 124 ManduraBhasma 125 (Ferri proxy rubram), Kasis126 (Iron sulphate), Suvarna MakshikBhasma (Iron pyrite) 124-,127; or Ayaskruti 128.
Preparation of IS in Ayurveda: All Ayurvedic metallic drugs undergo various purification/cleansing processes, termed ‘Shodhana’for removal of external/internal impurities in the ore and to enhance the therapeutic action of the final formulation.
Shodhana consists of Samanya Shodhana’(general purification) and ‘VisheshShodhana’(specific purification). ‘Marana’follows‘Shodhana’, wherein the iron ore is ignited repeatedly in a ‘Puta’ (dried cow-dungfurnace) and triturated until fine powders /Bhasmasare finally obtained Fig. 5. Lohabhasma was found to exhibit irregular aggregates of various sizes and shapes with nanostructures on the surface (100 to 500 nm) 128, 129 which has been confirmed with scanning electron micrographs 130, 131.
In some studies, the bhasmas contained nanoparticles that may easily penetrate the human intestinal epithelium and interact at the subcellular level 131.Whether this will enhance Fe absorption and what will be their effects on ROS and inflammation needs to be studied. Many medicinal plants are used for Shodhana and Marana i.e. Bhasmikaran. Table 3 lists the seven most commonly used plants and their properties. Many of these plant materials /phytoactive have pleiotropic action. Although numerous phytoactive have been investigated for several important pharmacological activities, e.g., anticancer, hypoglycemic, cardioprotective, wound healing, etc., these are not included. The review primarily deals with the anti-oxidant and/or anti-oxidant or anti-inflammatory activities that are likely to counter the side effects of Fe therapy.
The seven Ayurvedic plants used traditionally in the four bhasmas mentioned above and listed in Table 3 have both antioxidant and anti-inflammatory effects.
FIG. 5: TYPICAL PROCESS OF BHASMA PREPARATION
These properties may reduce the side effects of Fe, and some may also increase Fe bioavailability. The use of iron oxide specially heated, powdered, and processed with medicinal plants (Lauhabhasma) has been described, and different plants have been used for different types of anaemias 124, 125. Most of the plants used in bhasma preparation contain bioactive constituents and are pharmacologically active. Curcumin in turmeric, ellagic acid in Amlaki / Emblica offincinalis, and zingiberene in ginger possess antioxidant and anti-inflammatory properties, as do all the plants listed in Table 3.
These phytoactives also chelate Fe1 153, 154 and might help to reduce gut dysbiosis that can result from the unabsorbed Fe. Several clinical studies conducted on Fe preparations used in Ayurveda to treat Pandu (IDA) have shown encouraging results without exhibiting the common side effects as seen with conventional modern Fe preparations 143-148, 154-163.
Table 4 lists some plants used in other traditional systems of medicine. Most of the phytoactivesin the plants listed in Tables 3 and 4 are polyphenols that are well-known antioxidants.
TABLE 3: BENEFITS OF SELECTED AYURVEDIC MEDICINAL PLANTS USED IN SHODHANAAND MARANA OF LOHA & MANDUR ORE
Part of Plant | Botanical name | Sanskrit and Common) name | Phytoactive | Other pharmacological indications
|
Refs |
Seeds | Sesamum indicum | Til
(Til) |
Sesamin, sesamolin, sesaminol, sesamolinol, pinoresinol, etc | anticancer, antihyperlipidemic, hepatoprotective, | 132,133 |
Grains | Oryza sativa | Shali
(Rice) |
ϒ-Oryzanol, ferulic acid, β-sitosterol,* stigmasterol*, capesterol*, etc | antiaging, melanogenesis stimulating activity | 134,135,136 |
Roots | Raphanus sativus | Mulak
( Radish) |
Glucoraphanin*, glucoraphanin*, | Gut stimulatory, antihypertensive, antidiabetic, antiobesity & anti tussive, | 137 |
Grains | Mycrotylora uniflorum / Dolichos biflorum | Kulthha
( Horsegram) |
Quercetin, kaempferol, myricetin, daidzein, genistein, etc | Cytotoxic , analgesic, diuretic | 138, 139 |
Fruit | Phyllanthus emblica / Emblica officinalis | Amalki
(Amla) |
Cinnamic acid, gallic acid, quercetin, β-Daucosterol*, ellagic cid, 5-hydroxymethylfurfural*, | Antimicrobial, analgesic and antipyretic, hepatoprotective, antitumor and antiulcerogenic, antidiabetic, hypolipidemic, antibacterial activities | 140 |
Fruit | Terminalia chebula | Haritaki
( Harda ) |
Gallic acid, ellagic acid, ethyl gallate, vanillic acid, terchebulin, chebulagic acid, chebulanin, etc | Anticarcinogenic, Antimutagenic, radioprotective and chemopreventive, Hepatoprotective, Cardioprotective, Cytoprotective, Antidiabetic and renoprotective, Antibacterial, Antifungal, Antiviral, etc | 141,142 |
Fruit | Terminalia bellerica | Bibhitaki
(Behda) |
Bellericanin, ellagic acid, gallic acid, termilignan, thannilignan, ellargic acid, ethyl gallate, galloyl glucose,chebulaginic acid, phenyllemblin, α- sitosterol* | Antitussive, analgesic, hepatoprotective, antibacterial, anticancer and immune-modulatory activities | 143 |
Rhizome | Curcuma longa | Nisha (Haldi) | Curcumin; demethoxycurcumin; bisdemethoxycurcumin; 1,7-bis (4-hydroxyphenyl)-1, curcumadiol
4, 6-heptatrien-3-one; α-turmerone; β- turmerone; terpinolene; α-phellandrene; |
Anti-tumor, anticancer, anti-mutagenic, antifungal, antidiabetic, antifibrogenic, wound healing, immumodulatory | 144 |
*-compounds are not polyphenols
TABLE 4: PHYTOACTIVES IN SELECTED PLANT MATERIALS WITH ANTIOXIDANT AND ANTI-INFLAMMATORY ACTIVITY
S. no. | Plant source | Botanical name | Common name | Phytoactive | References |
1 | Leaves | Camellia sinensis | Tea | Epicatechin gallate, epigallocatechin gallate, epicatechin, epigallocatechin, carbohydrates*, caffeine, adenine, gallic acids, tannins, gallotannins, quercetin glycosides, carotenoids, tocopherols, vitamins (A, K, B, C)*, small amounts of aminophylline* | 145,146 |
2 | Leaves | Mimosa pudica | Touch-me-not | C-glycosylflavones, including 7, 8, 3',4'-tetrahydroxyl-6-C-[α-L-rhamnopyranosyl-(1→2)]-β-D-glucopyranosyl flavone; 5, 7, 4'-trihydroxyl-8-C-[α-L-rhamnopyranosyl-(→2)]-β-D-glucopyranosyl flavone; and 5, 7,3',4'-tetrahydroxyl-6-C-[α-L-rhamnopyranosyl-(1→2)]-β-D-glucopyranosyl flavone; 5,7,3',4'-tetrahydroxy-6-C-[β-D-apiose-(1→4)]-β-D-glucopyranosyl flavones; 5,7,4'-trihydroxyl-8-C-β-D-glu-copyranosyl flavones; 5,7,3',4'-teteahydroxy-6-C-[β-D-apiose-(1→4)]-β-D-glucopyranosyl flavones [3]; and 5,7,4'-trihydroxyl-8-C-β-D-glucopyranosyl flavones | 147 |
3 | Seed | Garcinia kola | Bitter kola | Kolaviron, garcinia hydroxybiflavanonol | 148 |
4 | Root | Scutellariabaicalensis Georgi | Chinese skull cap | Polyhydroxyflavonoids, namely baicalein, oroxylin A and wogonin |
149 |
5 | Root | Ligusticumwallichi | Szechuan lovage | Tetramethylpyrazine, (±)-3-methoxysedanenolide, sedanenolide, Z-ligustilide, Z-butylidenephthalide, methyl ferulate, trans-ferulic and trans-isoferulic acids, falcarindiol, -sitosterol, daucosterol* | 150,151 |
6 | Fruit | Vaccinium macrocarpon | Cranberry | Proanthocynadin Dimer A-type, Proanthocynadin Trimer B-type, Proanthocynadin Trimer A-type, Chlorogenic acid, Malonylcaffeoylquinic acid, Quercetin, Quercetin-3-rut, Quercetin-3-gal/glc, Quercetin-3-arab, Myricetin-3-gal/glu, Myricetin-3-ara, Syringetin-3-O-glu, Syringetin-ara, Laricitringlu, laricitrinara | 152 |
*-compounds are not polyphenols
Iron Chelation: Chelation of transition metals can directly reduce the rate of the FR and prevent OS caused by highly reactive ·OH radicals 164, 165. Fe chelators are chemically quite diverse they typically contain oxygen, nitrogen or sulfur (S)-donor atoms that form coordinate bonds with bound Fe. The donor atoms of the ligand affect the preference of the chelator for either the Fe (II) or Fe (III) oxidation states 165. Chelators that prefer Fe (II) contain ‘soft’ donor atoms, such as nitrogen and S, and consequently retain a relatively high affinity for other biologically important divalent metals such as Cu2+, Zn2+ 166. Fe can coordinate six ligands in an octahedral arrangement; thus, Fe chelators with the highest affinity will normally be hexadentate, binding Fe in a 1:1 ratio (chelator: iron). By contrast, bidentate (2:1 ratio) or tridentate (3:1 ratio) chelators, which bind to only two or three of the available Fe chelation sites, can potentially participate in the redox cycle and thereby promote free radical generation 167. Kontoghiorghe, Kolnagou & Kontoghiorghes 86 pointed out that unlike drugs or chelators used to treat Fe overload, substances/compounds explored as possible candidates for treatment of IDA, “should be able to increase Fe absorption and increase Hb production to normal physiological levels”. Effective Fe chelators must efficiently compete with the biological ligands that normally bind Fe; therefore, the affinity of chelators for Fe and their stoichiometry of Fe binding will greatly impact their activity as therapeutic agents 166, 169. Several synthetic compounds like deferoxamine mesylate, desferrithiocin, deferiprone are siderophores and synthetic chelators like 8-OH hydroxyquinoline derivatives, deferasirox, dexrazone, tachypyridine, thiosemicarbazones, pyridoxal isonicotinoyl hydrazone analogs are in focus. These compounds not only selectively chelate Fe3+ but also have antineoplastic activity 167. Plants or plant products, including those consumed as food by humans, contain low molecular weight organic molecules that are candidates as Fe chelators. They could protect against the toxic effects of ROS and reactive nitrogen species by inhibiting or mitigating the effects 86. Several compounds including polyphenols have great potential. Carboxylic acid derivatives like citrate, ascorbic acid, fulvic acid, humic acid, inositol phosphates, particularly the hexaphosphate, are also potential candidate phytomolecules 86, 87.
Antioxidant Activity of Plants / Phytochemicals: Polyphenols are among the most diverse and ubiquitous groups of plant secondary metabolites/ biomolecules. Although the anti-oxidant property of polyphenols has been known for a long time, the focus on their role as Fe chelators is recent 87, 168. Metal chelation by polyphenols has implications for several important processes in nature and biological systems 163, 164. They are part of the ‘plant defense’ against pathogens by chelating Fe and other essential minerals and thus limit the growth of invasive microorganisms by causing severe mineral depletion 169. These compounds are heterogeneous molecules, differing in their chemical structures 169-171. Also, they may be present as glycosides with different sugar units / acylated sugars at different positions of the polyphenol skeletons or bound to organic acids or with one another, creating additional diversity 172. Naturally occurring phenolic compounds can be divided into three classes: i) shortly distributed, e.g., simple phenols, pyrocatechol, hydroquinone, aldehydes derived from benzoic acids that are components of essential oils, e.g., vanillin, ii) widely distributed i.e., flavonoids and their derivatives, coumarins and iii) phenolic acids, e.g., benzoic/cinnamic acid and their derivatives) and polymers (tannins and lignins) 173.
Based on location in the plant (free in the soluble fraction of cell or bound to compounds of the cell wall), together with their chemical structures, phenolic compounds may also be: simple phenols (flavonoids and tannins of low and medium molecular weight not bound to membranes’ compounds) and essentially constituted phenols (condensed tannins, phenolic acids, other phenolic compounds of low-molecular-weight bound to cell wall polysaccharides or proteins forming insoluble stable complexes). This classification is relevant in terms of the metabolic fate in the GIT, and the physiological effects of each group will depend largely on their solubility characteristics. Insoluble phenolic compounds are not digested and may be partially or fully recovered quantitatively in the faeces, while a part of the soluble ones can cross the intestinal barrier and be found in the blood, unchanged or as metabolites 174.
Several polyphenols with a 1,2-dihydroxy, a α-hydroxyketo, or β-hydroxyketo substitution efficiently chelate trace metal ions, like Al3+ and Fe3+, and Cu+, that play an important role in oxygen metabolism and free radical formation 172-174. Polyphenols with gallol and catechol groups are the most potent antioxidants because of the large Fe-binding stability constants for these groups 171-173. Amongst polyphenols that are free radical scavengers and metal chelators, flavonoids have been well studied for their ability to chelate metals. The Fe chelation activity of flavonoids depends on their chemical structures and the position and degree of hydroxylation appear particularly important for their biochemical and pharmacological actions 171-176. Additionally, flavonoids have been shown to chelate Fe more efficiently when the metal ion is in bivalent form, meaning that the flavonoid needs to reduce Fe3+ to Fe2+ before association that depends upon the number of hydroxyl groups and pH of medium. The proposed binding site for trace metal ions to flavonoids is the 3′, 4′-di-OH moiety in the B ring. In addition, C-3 and C-5 OH groups and the 4-carbonyl group also contribute to metal ion chelation.
At pH 5.5, the flavones myricetin and quercetin reduced Fe3+, rutin, catechin, and taxifolin were moderately active but kaempherol and luteolin were poor reductants. This is because the simultaneous presence of the catechol group in the B-ring and 3-hydroxyl group in the C-ring plays a crucial role in reducing the potential of flavonoids. The presence of 2, 3-double bond in conjugation with the 4-oxo group in C ring is also important for Fe3+ reducing capacity. The 2,3-double bond apparently increases the planarity of the molecule, confers higher rigidity to the ring, and holds the A and C rings in a more coplanar position, allowing the 3-hydroxyl/4-oxo groups and 5-hydroxyl/4-oxo groups to be closer. Thus, polyphenols with gallol and catechol groups are the most potent antioxidants because of the large Fe-binding stability constants for these groups 171-176. The catechol moiety of polyphenols is not just important for metal chelation. It also confers antioxidant properties to curcumin, caffeic acid, catechin, and protocatechuic acid, gallic acid, epicatechin gallate, ellagic acid, tannins. Flavonoids like quercetin, kaempferol, myricetin, green tea catechins and black tea theaflavins, the isoflavones genistein and mangiferin all have Fe-chelating ability. Overall, all gallic acid containing compounds could be potential Fe chelators 176-178. Another potential advantage besides binding Fe and functioning as antioxidants86 is that compounds like flavan-3-ols and their condensation products. i.e. proanthocyanidins have anti-microbial properties 171, 176, 178.
The strong Fe binding affinities of polyphenol-rich plant materials that help to suppress Fenton chemistry 178, suggest their potential for use as Fe chelators in two new but important areas – (i) developing Fe supplements with low side effects and (ii) food supplements for diseases characterized by OS. In a way, both these ideas are not new. Ayurveda uses elemental Fe (Lohabhasma) for the treatment of anaemia. Many of the IS formulations contain botanicals such as amla (Indian gooseberry), dadima (pomegranate), draksha (raisins), and all these botanicals are rich in polyphenols. One potential reason for their inclusion in the formulations could have been to control the gut's free radical generation, but this needs to be studied.
The health benefits of dietary fruits and vegetables, especially those related to OS e.g., cardiovascular disease, atherosclerosis, neurodegenerative diseases, and cancer are well recognized. These health benefits maybe partly due to the Fe chelating activity of polyphenols and flavonoids and mitigating OS through free-radical sequestration 85,169, 174-183. While excess Fe is not regarded as the underlying cause of these diseases, it may play an important role in disease progression either through promotion of cellular growth and proliferation or through participation in redox reactions that catalyze ROS formation and increase OS.
The most well-absorbed flavonoids by humans, such as isoflavones, gallic acid, catechins, flavanones, and quercetin glucosides176, may offer opportunities to design novel Fe-chelating food supplements and herbo-mineral Fe supplements with low side-effects. Even the non-absorbed flavonoids may play an important role in designing ‘side-effect’-free IS, as that chemistry happens in the gut. The bioactivity related to Fe chelation for select foods from Ayurveda and modern diets that are polyphenol-rich are summarized herein:
Fruits: Grape seed extract (GSE) contains various polyphenols like gallic acid, catechin, epigallocatechin gallate (EGCG) and proanthocyanidins. GSE possesses antioxidant activity183-185, as these constituents scavenge free radicals and chelate metals such as Fe 153. However, the effect of these compounds on uptake of dietary Fe and the uptake of Fe–polyphenol complexes needs to be assessed. Procyanidins extracted from Vitis vinifera, formed a complex with Fe3+with a procyanidin–Fe ratio of 1:2. The stability constant of the procyanidin–Fe complex was comparable to another strong Fe chelating agent, nitrilotriacetate 185, 186. The mechanism of this complex interaction between radical species requires further investigation; furthermore, in-vivo studies are necessary to identify the precise compounds that eventually exert biological activity.
Quercetin, a flavonol is the major phenolic compound in cranberries. It was found to bind Fe2+ more strongly than ferrozine-a widely used Fe2+ chelator 179, 184, 185. Quercetin completely suppressed Fe-promoted Fenton chemistry at micromolar levels, even in the presence of the major cellular Fe chelators -ATP or citrate.
These data indicate that it can completely inhibit the Fenton chemistry, although it provides only partial protection against Fenton chemistry-mediated damage 179. Flavonols, including quercetin, are rapidly absorbed and reach maximum plasma concentration within a few hours. Their elimination (e.g., quercetin metabolites) is quite slow, with reported elimination half-lives ranging from 11 to 28 h. Circulating flavonoids (and their metabolites) are further delivered into various organs, including the liver, skin, and brain. The link of Fe chelation ability of bioavailable polyphenols of Cranberry extracts to various biological activities e.g., LDL oxidation 179, 183, oxidative and inflammatory damage to the vascular endothelium 187, 188, inhibition of proliferation of several human tumor cell lines 189, 190, 191 needs to be explored.
Green tea catechins & black tea theaflavins: The most abundant polyphenolic compoundsin green tea are catechins 177.
Besides their radical scavenging action, green tea catechins possess well-established metal-chelating properties. The structurally important features defining their chelating potential are the 3′,4′-dihydroxyl group in the B ring and the gallate group 192-196. Polyphenols from green and black tea have shown promise in protective, chemotherapeutic and chemopreventive activity in-vitro. Dried and purified tea polyphenols from black and green teas had a profound protective effect on red blood cells challenged with exogenous oxidants via forming a redox-inactive complex with Fe 195. Their chemotherapeutic ability has been evaluated for inducing cell death in several human cancer cells e.g. human breast (MCF-7), colon (HT-29), hepatoma (liver-HepG2), prostate (PC-3) and lung (HEL299) cells 197 and in rodent models of breast carcinogenesis 198-200. The role of Fe chelation as a mechanism for these protective, chemotherapeutic and chemo preventive activities of tea polyphenols needs to be studied.
Green tea catechin polyphenols act as multimodal acting molecules that direct numerous cellular neuroprotection/ neurorescue mechanisms involving Fe chelation, scavenging oxygen and nitrogen radical species, activation of protein kinase C signaling pathway and prosurvival genes 199. Their nontoxic, lipophilic (and thus, brain-permeable) nature is advocated for Fe removal from those brain areas where it preferentially accumulates in neurodegenerative diseases 200. EGCG has been shown to improve age-related cognitive decline and to protect against cerebral IRIs 201, 202, brain inflammation, and neuronal damage in experimental autoimmune encephalomyelitis 202. Long-term administration of green tea catechins or EGCG improved spatial cognition and learning ability in rats and reduced cerebral amyloidosis in Alzheimer’s transgenic mice 203, 204.
Curcumin: Curcumin, the active ingredient in the traditional herbal remedy and dietary spice turmeric, is a free radical scavenger and hydrogen donor, with both pro- and anti-oxidant activity 170. It is remarkably nontoxic and has limited bioavailability. Curcumin has great promise as a therapeutic agent and is currently in human clinical trials for varied conditions, e.g., multiple myeloma, pancreatic cancer, myelodysplastic syndromes, colon cancer, psoriasis, and AD. Curcumin inhibits cancer development and progression via its ability to target multiple steps in the pathway(s) to malignancy 206.
chelates Fe in-vivo, particularly in mild Fe deficiency 205, 206. Under these conditions, dietary curcumin exerted profound effects on systemic Fe, inducing a decline in hematocrit, Hb, serum Fe, transferrin saturation, hypochromic RBC appearance, and decreases in spleen and liver Fe content, besides repressing hepcidin synthesis. Since Fe is critical to catalyzing redox cycling, Fe chelation therapy should be considered a valuable strategy for treating neurodegenerative diseases 206, 207. Fe chelation may contribute to curcumin’santi-cancer activity 205,208, as its chemical properties are consistent with Fe-chelator activity 209, 210.
Other neuroprotective phytoactives are apocyanin present in the rhizome of Picrorhizakurroa, a well-known herb in traditional Ayurvedic medicine 211-213. Apocyanin exhibited potent antioxidative properties by scavenging free radicals and acted on specific signaling pathways that regulate inflammatory responses in cultured human eosinophils and neutrophils 213. Mucuna pruriens, another naturally occurring antioxidant used in traditional Ayurvedic Indian medicine, has also been shown to slow the progression of PD symptoms, without having the side effects of the current pharmaceutical L-dopa. Mucuna pruiens inhibited the oxidation of lipids and deoxyribose sugars and exhibited divalent Fe-chelating activity 214.
As stated by Guo et al., 215 phenolic compounds with an ‘Fe-binding motif’ are strong Fe-chelating agents that may modulate the bioactivity and bioavailability of Fe in the body and merit closer scientific scrutiny.
Future Perspectives: The recognition that a number of plant polyphenols function as Fe chelators is encouraging. The chelating activity of dietary polyphenols may partially contribute to the widely acknowledged health benefits of dietary fruits and vegetables 183, 210. The recognition that dietary constituents are long known to have beneficial health effects contain constituents with strong binding affinities for Fe that can suppress Fenton chemistry opens the prospect of extending the use of Fechelators for designing herbo-mineral Fe supplements that have lesser side effects than current Fe salts as well as polyphenol supplements for OS-related diseases. Optimization of the use of such polyphenols will require a careful balancing of overall nutritional demands. Nevertheless, prospects for the use of Fechelators in the treatment and prevention of human disease appear greater than ever.
ACKNOWLEDGEMENT: The authors acknowledge the Rural India Support Trust for supporting Gurmeet Singh's research at the University of Trans-Disciplinary Health Sciences & Technology. The authors wish to acknowledge the Rajiv Gandhi Science and Technology Commission, Government of Maharashtra.
CONFLICTS OF INTEREST: The authors have no conflicts to declare.
REFERENCES:
- World Health Organization: Iron Deficiency Anaemia. Assessment, Prevention, and Control. A Guide for Programme Managers. World Health Organization, United Nations University 2001.
- Mayo Clinic: Anemia, Symptoms and Causes. https://www.mayoclinic.org/d-2020 accessed on August 1, 2021
- Gardner W and Kassebaum N: Global, regional, and national prevalence of anemia and its causes in 204 countries and territories, 1990–2019, global, regional, and national prevalence of anemia and its causes in 204 countries and territories, 1990–2019. Curr Dev Nutr 2020; 4(2): 830. https://doi.org/10.1093/cdn/nzaa053_035:
- International Institute of Population Sciences (IIPS) and ICF: National Family Health Survey (NFHS-4), 2015-16: India. IIPS 2017
- Sunuwar DR, Singh DR, Chaudhary NK, Pradhan PMN, Rai P and Tiwari K: Prevalence and factors associated with anemia among women of reproductive age in seven South and Southeast Asian countries: Evidence from nationally representative surveys. PLoS One 2020; 15(8): 0236449.
- Daru J, Zamora J, Fernandéz-Félix BM, Vogel J, Oladapo OT and Morisaki N: Risk of maternal mortality in women with severe anemia during pregnancy and postpartum: a multilevel analysis. The Lancet-Global Health 2018; 6(5): 548-554.
- World Health Organization. Global Nutrition Targets 2025: Policy Brief Series WHO/NMH/NMD/14.2). Geneva: World Health Organization.
- Rai S and Kar AC: A review of role of psychological factors in the etiopathogenesis of Pandu roga with reference to iron deficiency anemia. Ayu 2016; 31(1): 18-21.
- National Family Health Survey (NFHS-5) http://rchiips.org › nfhs › factsheet_NFHS-5. Accessed on August 15, 2021
- Ministry of Health and Family Welfare. Health and Family Welfare Statistics in India 2019-20, Govt of India.
- Ministry of Health and Family Welfare. Comprehensive National Nutrition Survey. Government of India 2016-18.
- De Falco L, Sanchez M, Silvestri L, Kannengiesser C, Muckenthaler MU, Iolascon A, Gouya L, Camaschella C, and Beaumont C: Iron refractory iron deficiency anemia. Haematologica 2013; 98(6): 845-853.
- Bloor SR, Schutte R, and Hobson AR: Oral iron supplementation gastrointestinal side effects and the impact on the gut microbiota. Microbiol Res 2021; 12: 491–502.
- Mayo Clinic: Drugs and Supplements: Iron Supplement- Oral route, Parenteral route. https://www.mayoclinic.org/drugs-supplements/iron-supplement-oral-route-parenteral-route/side-effects/drg-20070148 accessed on August 15, 2021.
- Dubey S, Suri V, Aggarwal N and Das R: Is it safe to use intravenous iron sucrose during pregnancy? A randomized controlled trial. Int J Reprod Contracept Obstet Gynecol 2013; 2(4): 544-549.
- Mithra P, Unnikrishnan B, Rekha T, Nithin K, Mohan K, Kulkarni V, Kulkarni Vand Agarwal D: Compliance with iron-folic acid (IFA) therapy among pregnant women in an urban area of south India. Afr Health Sci 2013; 13(4): 880-885.
- Deshmukh PR, Garg BS and Bharambe MS: Effectiveness of weekly supplementation of iron to control anaemia among adolescent girls of Nashik, Maharashtra, India. J Health PopulNutr 2008; 26(1): 74-78.
- Pēna-Rosas JP, De-Regil LM, Malave HG, Flores-Urrutia MC, and Dowsell T: Intermittent oral iron supplementation during pregnancy. Cochrane Database Syst Rev 2015; 2015(10): CD009997.
- Tolkien Z, Stecher L, Mander AP, Pereira DIA and Powell JJ: Ferrous Sulfate Supplementation Causes Significant Gastrointestinal Side-Effects in Adults: A Systematic Review and Meta-Analysis. PLoS ONE 2015; 10(2): 0117383.
- Yasa B, Agoglu L and Unuvar E: Efficacy, tolerability, and acceptability of iron hydroxide polymaltose complex versus ferrous sulfate: A randomized trial in pediatric patients with iron deficiency Anemia. Int J of Pediatrics 2011; 2011: 524520.
- Birben E, Sahiner UM, Sackesen C, Erzurum S and Kalayci O: Oxidative stress and antioxidant defense. World Allergy Organ J 2012; 5(1): 9-19.
- Kontoghiorghes GJ and Kontoghiorghe CN: Iron and chelation in Biochemistry and Medicine: New Approaches to Controlling Iron Metabolism and Treating Related Diseases. Cells 2020; 9(6): 1456.
- Camaschella C, Nai A and Silvestri L: Iron metabolism and iron disorders revisited in the hepcidin era. B247 2020; 105(2): 260-272.
- Johnson DC, Dean DR, Smith AD and Johnson MK: Structure, function, and formation of biological iron-sulfur clusters. Ann Rev Biochem 2005; 74: 247-281.
- Heeney MM and Finberg KE: Iron-refractory iron deficiency anemia (IRIDA). Hematology/ Oncology Clinics of North America 2014; 28(4): 637-652.
- Cook JD and Reddy MB: Efficacy of weekly compared with daily iron supplementation. Am J Clin Nutr 1995; 62(1): 117-20.
- Ganz T and Nemeth E: Hepcidin and iron homeostasis. BiochimBiophys Acta 2012; 1823(9): 1434-1443.
- Nemeth E, Rivera S, Gabayan V, Keller C, Taudorf S, Pedersen BK and Ganz TL: IL-6 mediates hypoferremia of inflammation by inducing the synthesis of the iron regulatory hormone hepcidin. J Clin Investig 2004;113(9):1271-1276.
- Lin L, Valore E V, Nemeth E, Goodnough JB, Gabayan V and Ganz T: Iron transferrin regulates hepcidin synthesis in primary hepatocyte culture through hemojuvelin and BMP2/4. Blood 2007; 110(6): 2182-2189.
- Moretti D, Goede JS, Zeder C, Jiskra M, Chatzinakou V, Tjalsma H, Melse-Boonstra A, Brittenham G, Swinkels DW and Zimmerman MB: Oral iron supplements increase hepcidin and decrease iron absorption from daily or twice-daily doses in iron-depleted young women. Blood 2015; 126(17): 1981-1989.
- Waldvogel-Abramowskia S, Waeberb W, Gassnerc C, Buserd A, Beat M, Bernard F and Favrate JD: Physiology of iron metabolism. Transfusion Medicine and Hemotherapy 2014; 41: 213-221.
- Soe-Lin S, Apte SS, Andriopoulos B, Andrews MC, Schranzhofer M, Kahawita T, Garcia-Santos D and Ponka P: Nramp1 promotes efficient macrophage recycling of iron following erythrophagocytosis in-vivo. PNAS 2009; 106(14): 5960-5965.
- Shi H, Bencze KZ, Stemmler TL and Philpott CC: A cytosolic iron chaperone that delivers iron to ferritin. Science 2008; 320(5880): 1207-1210.
- Knutson MD: Steap proteins: implications for iron and copper metabolism. Nutr Rev 2007; 65: 335-340.
- Lonnerdal B, Georgieff MK, and Hernell O: Developmental physiology of iron absorption, homeostasis, and metabolism in the healthy term infant. J Pediatr 2016; 167(4): 8-14.
- Shayeghi M, Latunde-Dada GO, Oakhill JS, Laftah AH, Takeuchi K, Halliday N, Khan Y, Warley A, McCann FE, Hider RC, Frazer DM, Anderson GJ, Vulpe CD, Simpson RJ, and McKie AT. Identification of an intestinal heme transporter. Cell 2005; 122: 789–801.
- Le Blanc S, Garrick MD and Arredondo M: Heme carrier protein 1 transports heme and is involved in heme-Fe metabolism. Am J Physiol: Cell Physiol 2012; 302: 1780–1785.
- World Health Organization Haemoglobin concentrations for the diagnosis of anaemia and assessment of severity. VMNIS | Vitamin and Mineral Nutrition Information SystemWHO/NMH/NHD/MNM/11.1 2007
- Pfeiffer CM and Looker AC: Laboratory methodologies for indicators of iron status: strengths, limitations, and analytical challenges. Am J Clin Nutr 2017; 106(6):1606-1614.
- Adsul BB, Qayum M, Khandeparkar P and Naik M: An efficacy, safety and tolerability study of ferrous ascorbate and folic acid (Phosfomin-XT) in iron deficiency anemia. Ind J Clin Prac 2012; 22(11): 565-570.
- Li N, Zhao G, Wu W, Zhang M, Liu W, Chen Q and Wang X: The efficacy and safety of vitamin C for iron supplementation in adult Patients With iron deficiency anemia. A randomized clinical trial. JAMA Netw Open 2020; 3(11): 2023644.
- Auerbach M and Ballard H: Clinical use of intravenous iron: administration, efficacy and safety. Hematology Am Soc Hematol Educ Program 2010; 2010: 338-47.
- Nielsen OH, Ainsworth M, Coskun M and Weiss G: Management of iron-deficiency anemia in inflammatory bowel disease. A systematic review. Medicine (Baltimore) 2015; 94(23): 963.
- Auerbach M, Coyne D, and Ballard H. Intravenous iron: from anathema to standard of care. Am J Hematol 2008; 83(7): 580-588.
- Pharmacist’s Letter / Prescriber’s Letter. August 2008-Volume 24- Number 240811
- Gutiérrez OM: Treatment of Iron Deficiency Anemia in CKD and End-Stage Kidney Disease. Kidney International Reports 2021; 6(9): 2261-2269.
- Nikravesh N, Borchard G, Hofmann H, Philipp E, Fluhmann B, and Wick P: Factors influencing safety and efficacy of intravenous iron-carbohydrate nanomedicines: From production to clinical practice. Nanomedicine. 2020; 26: 102178.
- Thankachan P, Bose B, Subramanian R, Koneri R and Kurpad AV: Fractional iron absorption from enteric-coated ferrous sulphate tablet. Ind J Med Res 2020; 151: 371-374.
- Hurrell RF: Iron Fortification Practices and implications for iron addition to salt. J Nutr 2021; 151: 3-14.
- Allen L, de Benoist B, Dary O and Hurrell R: (eds) Guidelines on Food Fortification with Micronutrients. World Health Organization and Food and Agricultural Organization of the United Nations 2006
- Kurpad AV, Ghosh S, Thomas T, Bandyopadhyay S, Goswami R, Gupta A, Gupta P, John AT, Kapil U, Kulkarni B, Kuriyan R, Madan J, Makkar S, Nair KM, Pullakhandam R, Reddy GB, Shah D and Sachdev HS: Perspective: When the cure might become the malady: the layering of multiple interventions with mandatory micronutrient fortification of foods in India. Am J Clin Nutr 2021; 114: 1261-1266.
- Fodor I and Marx JJM: Lipid peroxidation of rabbit small intestinal microvillus membrane vesicles by iron complexes. Biochim Biophys Acta 1988; 961(1): 96-102.
- Srigiridhar K and Nair MK: Iron-deficient intestine is more susceptible to peroxidative damage during iron supplementation in rats. Free Radic Biol Med 1998; 25(6): 660-665.
- Pendyala S and Natarajan V: Redox regulation of Nox proteins. Respir Physiol Neurobiol 2010; 174: 265–271.
- Thannickal VJ and Fanburg BL: Reactive oxygen species in cell signaling. Am J Physiol Lung Cell Mol Physiol 2000; 279: 1005–1028.
- Lambeth JD: NOX enzymes and the biology of reactive oxygen. Nat Rev Immunol 2004; 4(3): 181-189.
- Handy DE and Loscalzo J: Redox regulation of mitochondrial function. Antioxid Redox Signal 2012; 16: 1323-1367.
- Graf E, Mahoney JR, Bryant RG and Eaton JW: Iron catalyzed hydroxyl radical formation. Stringent requirement for free iron coordination site. J Biol Chem 1984; 259(6): 3620-3624.
- Kell DB: Iron behaving badly: inappropriate iron chelation as a major contributor to the aetiology of vascular and other progressive inflammatory and degenerative diseases. BMC Med Genomics 2009; 2: 2.
- Navarro-Yepes J, Burns M, Anandhan A, Khalimonchuk O, del Razo LM, Quintanilla- Vega B, Pappa A, Panayiotidis MI and Franco MR: Oxidative stress, redox signaling, and autophagy: cell death versus survival. Antioxid Redox Signal 2014; 21(1): 66-85.
- Huang X and Groves JT: Oxygen Activation and Radical Transformations in Heme Proteins and Metalloporphyrins. Chem Rev 2018; 118(5): 2491-2553.
- Stohs SJ and Bagchi D: Oxidative mechanisms in the toxicity of metal ions. RBM 1995; 18(2): 321-336.
- Bienert GP, Moller AL, Kristiansen KA, Schulz A, Moller IM, Schjoerring JK and Jahn TP: Specific aquaporins facilitate the diffusion of hydrogen peroxide across membranes. J Biol Chem 2007; 282: 1183-1192.
- Miller EW, Dickinson BC and Chang CJ: Aquaporin-3 mediates hydrogen peroxide uptake to regulate downstream intracellular signalling. PNAS 2010; 107: 15681-15686.
- Hara-Chikuma M, Chikuma S, Sugiyama Y, Kabashima K, Verkman AS, Inoue S and Miyachi Y: Chemokine-dependent T cell migration requires aquaporin-3-mediated hydrogen peroxide uptake. J Exp Biol Med 2012; 209: 1743-1752.
- Bhattacharyya A, Chattopadhyay R, Mitra S, and Crowe SE: Oxidative Stress: An Essential Factor in the Pathogenesis of Gastrointestinal Mucosal Diseases. Physiol Rev 2014; 94(2): 329-54.
- Tiwari AKM, Mahdi AA, Chandyan S, Zahra F, Godbole MM, Jaiswar SP, Srivastava VK, and Negi MPS. Oral iron supplementation leads to oxidative imbalance in anemic women: a prospective study. Clin Nutr 2011; 30(2): 188-193.
- Shastri L, Pammal RS, Mani I, Thomas T and Kurpad AV: Oxidative stress during early pregnancy and birth outcomes. Public Health Nutr 2016; 9(17): 3210–3215.
- Lund EK, Wharf SG, Fairweather-Tait SJ and Johnson IT: Oral ferrous sulfate supplements increase the free radical-generating capacity of feces from healthy volunteers. Am J Clin Nutr 1999; 69(2): 250-255.
- Troost FJ, Saris WHM, Haenen GRMM, Bast A and Brummer R-JM: New method to study oxidative damage and antioxidants in the human small bowel: effects of iron application. Am J PhysiolGastrointest Liver Physiol 2003; 285(2): 354-359.
- Deli CK, Fatouros IG, Paschalis V, Tsiokanos A, Georgakouli K, Zalavras A, Avloniti A, Koutedakis Y, and Jamurtas AZ: Iron supplementation effects on redox status following aseptic skeletal muscle trauma in adults and children. Oxid Med Cell Longev 2017; 2017: 4120421.
- Imam MU, Zhang S, Ma J, Wang H, and Wang F: Antioxidants mediate both iron homeostasis and oxidative stress. Nutrients 2017; 9(7): 671.
- Hurrell RF: Influence of inflammatory disorders and infection on iron absorption and efficacy of iron-fortified foods. Nestle Nutrition Institute Workshop Series. 2012; 70: 107-16.
- Schmidt PJ: Regulation of iron metabolism by hepcidin under conditions of inflammation. J Biol Chem 2015; 290: 18975-18983.
- Cassat JE and Skaar EP: Iron in infection and immunity. Cell Host Microbe 2013; 13: 509-519.
- Grune T, Sommerburg O and Siems WG: Oxidative stress in anemia. Clin Nephrol 2000; 53: 18-22.
- Eid HM, Wright ML, Kumar NVA, Qawasmeh A, Hassan STS, Mocan A, Nabavi SM, Rastrelli L and Haddad PS: Significance of microbiota in obesity and metabolic diseases and the modulatory potential by medicinal plant and food ingredients. Front Pharmacol. 2017; 8: 387.
- Botta A, Barra NG, Lam NH, Chow S, Pantopoulos K, Schertzer JD and Sweeney G: Iron reshapes the gut microbiome and host metabolism. J Lipid Atheroscler 2021; 10(2): 160-183.
- Liu S, Gao J, Zhu M, Liu K and Zhang H-L: Gut microbiota and dysbiosis in Alzheimer’s disease: Implications for pathogenesis and treatment. Mol Neurobiol 2020; 57: 5026-5043.
- van Vuuren MJ, Nell TA, Carr JA, Kell DB and Preteorius E: Iron dysregulation and inflammagens related to oral and gut health are central to the development of Parkinson’s disease. Biomol 2021; 11(1): 30.
- Mirsepasi-Lauridsen HC, Vrankx K, Engberg J, Friis-Møller A, Brynskov J, Nordgaard-Lassen I, Petersen A, and Krogfelt KA: Disease-specific enteric microbiome dysbiosis in inflammatory bowel disease. Front Med 2018; 5: 304.
- Mayneris-Perxachs J, Cardellini M, Hoyles L, Latorre J, Davato F, Moreno-Navarrete J-M, Arnoriaga-Rodríguez M, Serino M, Abbott J, Barton RH, Puig J, Fernández-Real X, Ricart W, Tomlinson C, Holmes E, Nicholson JK, Pérez-Brocal V, Moya A, Mc Clain D, Burcelin R, Dumas M-E, Federic M and Fernández-Real J-M: Iron status influences non-alcoholic fatty liver disease in obesity through the gut microbiome. Microbiome 2021; 9: 104.
- Vivarelli S, Salemi R, Candido S, Falzone L, Santagati M, Stefani S, Torino F, Banna GL, Tonini G and Libra M: Gut microbiota and cancer: From pathogenesis to therapy. Cancers (Basel) 2019; 11(1): 38.
- Serino M, Blasco-Baque V, Nicolas S, and Burcelin R: Far from the Eyes, Close to the heart: Dysbiosis of gut microbiota and cardiovascular consequences. Curr Cardiol Rep 2014; 16(11): 540.
- Kell DB and Pretorius E: No effects without causes: The iron dysregulation and dormant microbes hypothesis for chronic, inflammatory diseases. Biol Rev Camb Philos Soc 2018; 93(3): 1518-1557.
- Kontoghiorghe CN, Kolnagou A and Kontoghiorghes GJ: Phytochelators intended for clinical use in iron overload, other diseases of iron imbalance and free radical pathology. Mol 2015;20(11):20841-20872.
- Kontoghiorghes, GJ and Kontoghiorghes CN: Prospects for the introduction of targeted antioxidant drugs for the prevention and treatment of diseases related to free radical pathology. Expert OpinInvestig Drugs 2019; 28(7): 593-603.
- Srigiridhar K, Nair KM, Subramanian R, and Singotamu L: Oral repletion of iron induces free radical mediated alterations in the gastrointestinal tract of rat. Mol Cell Biochem 2001; 219(1-2): 91-98.
- El-Sheikh AA, Ameen SA and Abdel-Fatah SS: Ameliorating Iron Overload in Intestinal Tissue of Adult Male Rats: Quercetin vs J Toxicol 2018; 2018: 8023840.
- Luo Q, Lao C, Huang C, Xia Y, Ma W, Liu W and Chen Z: Iron Overload Resulting from the Chronic Oral Administration of Ferric Citrate Impairs Intestinal Immune and Barrier in Mice. Biol Trace Elem Res 2021; 199(3): 1027-1036.
- Powers JM, Buchanan GR, and Adix L: Effect of low-dose ferrous sulfate vs iron polysaccharide complex on hemoglobin concentration in young children with nutritional iron-deficiency anemia. A randomized clinical trial. J Am Med Assoc 2017; 317(22): 2297-2304.
- Casparis D, Carlo PD, Branconi F, Grossi A, Merante D, and Gafforio L: Effectiveness and tolerability of oral liquid ferrous gluconate in iron-deficiency anemia in pregnancy and in the immediate post-partum period: comparison with other liquid or solid formulations containing bivalent or trivalent iron. Minerva Obstet Gynecol 1996; 48(11): 511-518.
- Kavakli K, Yilmaz D, Çetinkaya B, Balkan C, Sözman EY and Săgin FG: Safety profiles of Fe2+ and Fe3+ oral preparations in the treatment of iron deficiency anemia in children. PediatrHematol Oncol 2004; 21(5):403-410.
- Stoffel NU, Zeder C, Brittenham GM, Moretti D and Zimmerman MB: Iron absorption from supplements is greater with alternate day than with consecutive day dosing in iron-deficient anemic women. Haematologica 2020; 105(5): 1232-1239.
- Srigiridhar K and Nair KM: Iron-deficient intestine is more susceptible to peroxidative damage during iron supplementation in rats. Free Radic Biol Med 1998; 25(6): 660-665.
- Jaeggi T, Kortman GA, Moretti D, Chassard C, Holding P, Dostal A, Boekhorst J, Timmerman HM, Swinkels DW, Tjalsma H, Njenga J, Mwangi A, Kvalsvig J, Lacroix C, and Zimmermann MB: Iron fortification adversely affects the gut microbiome, increases pathogen abundance and induces intestinal inflammation in Kenyan infants. Gut 2015; 64(5): 731-42.
- Paganini D and Zimmermann MB: The effects of iron fortification and supplementation on the gut microbiome and diarrhea in infants and children: a review. Am J Clin Nutr 2017; 106: 1688-1693.
- Parmanand BA, Kellingray L, Gall GL, Basit AW, Fairweather-Tait S and Narbad A: A decrease in iron availability to human gut microbiome reduces the growth of potentially pathogenic gut bacteria; an in vitro colonic fermentation study. J Nutri Biochem 2019; 67:20-27.
- Wilson AS, Koller KR, Ramaboli MC, Nesengani LT, Ocvirk S, Chen C, Flanagan CA, Sapp FR, Merritt ZT, Bhatti F, Thomas TK and O'Keefe SJD: Diet and the human gut microbiome: An international review. Dig Dis Sci 2020; 65(3): 723-740.
- Yilmaz B, Mooser C, Keller I, Zimmermann J, Bosshard L, Fuhrer T, de Agüero MG, Trigo NF, Tschanz-Lischer H, Limenitakis JP, Hardt W-D, McCoy KD, Stecher B, Excoffier L, Ganal-Vonarburg SC and Macpherson AJ: Long-term evolution and short-term adaptation of microbiota strains and sub-strains in mice. Cell Host Microbe 2021; 29(4): 650-663. 9.
- Finlayson-Trick ECL, Fischer JAJ, Goldfarb DM, and Karakochuk D: The effects of iron supplementation and fortification on the gut microbiota: A review. Gastrointest Disord 2020; 2(4): 327-340.
- Bouglé D, Vaghefi-Vaezzadeh N, Roland N and Bouvard G: Influence of short-chain fatty acids on iron absorption by proximal colon. Scand J Gastroenterol 2002; 37(9): 008-1011.
- Zughaier SM and Cornelis P. Editorial: Role of iron in bacterial pathogenesis. Front Cell Infe Micro 2018; 8: 344.
- Sritharan M. Iron and bacterial virulence. Ind J Med Microbiol 2006; 24(3): 163-164.
- Kortman GAM, Boleij A, Swinkels DW and Tjalsma H: Iron availability increases the pathogenic potential of Salmonella typhimurium and other enteric pathogens at the intestinal epithelial interface. PLOS One 2012; 7(1): 29968.
- Nairz M and Weiss G: Iron in infection and immunity. Mol Aspects Med 2020; 75: 100764.
- Gera T and Sachdev HPS: Effect of iron supplementation on incidence of infectious illness in children: Systematic Review. BMJ 2002; 325(7373): 1142-1152.
- Larson LM, Phiri KS and Pasricha SR: Iron and Cognitive Development: What Is the Evidence? Ann Nutr Metab 2017: 71(3): 25-38.
- Hare DJ, Braat S, Cardoso BR, Morgan C, Szymlek-Gay EA, and Biggs BA: Health outcomes of iron supplementation and/or food fortification in iron-replete children aged 4-24 months: protocol for a systematic review and meta-analysis. Syst Rev 2019; 8(1): 253.
- World Health Organization. Guideline: Daily Iron Supplementation in Infants and Children. World Health Organization, Geneva 2016.
- Sazawal S, Black RE, Ramsan M, Chwaya HM, Stoltzfus RJ, Dutta A, Dhingra U, Kabole I, Deb S, Othman MK, and Kabole FM: 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. Lancet 2006; 367(9505): 133-143.
- Lönnerdal B: Excess iron intake as a factor in growth, infections and development of infants and young children. Am J Clin Nutr 2017; 106(l6): 1681-1687.
- Roberts JL and Stein AD: The impact of nutritional interventions beyond the first 2 years of life on linear growth: A systematic review and meta-analysis. Adv Nutr 2017; 8(2): 323-336.
- Sachdev HPS, Gera T and Nestel P: Effect of iron supplementation on physical growth in children: systematic review of randomised controlled trials. Public Health Nutr 2006; 9(7): 904-920.
- Pasricha S-R, Hayes E, Kalumba K, and Biggs B-A: Effect of daily iron supplementation on health in children aged 4-23 months: a systematic review and meta-analysis of randomised controlled trials. Lancet Glob Health 2013; 1(2):77-86.
- Nchito M, Friis H, Michaelsen KF, Mubila F and Olsen A: Iron supplementation increases small intestine permeability in primary schoolchildren in Lusaka, Zambia. Trans R Soc Trop Med Hy 2006; 100(8): 791-794.
- De Gier B, Ponce MC, Perignon M, Florentino M, Khov K, Chamnan C, de Boer MR, Parker ME, Burja K, Dijkhuizen MA, Berger J, Polman K, and Wieringa FT: Micronutrient-Fortified Rice Can Increase Hookworm Infection Risk: A Cluster Randomized Trial. PLoS One 2016; 11(1): 0145351.
- Clark MA, Goheen MM, Fulford A, Prentice AM, Elnagheeb MA, Patel J, Fisher N, Taylor SM, Kasthuri RS, and Cerami C: Host iron status and iron supplementation mediate susceptibility to erythrocytic stage Plasmodium falciparum. Nature Comm 2014; 25(5):4446.
- Pizzorno J: Toxins from the gut. Integr Med (Encinitas) 2014; 13(6): 8-11.
- Buret AG, Allain T, Motta J-P, and Wallace JL: Effects of hydrogen sulfide on the microbiome: from toxicity to therapy. Antioxid Redox Signal 2021; Online ahead of Print.
- Ahmad AF, Dwivedi G, O’Gara F, Caparros-Martin J, and Ward NC: The gut microbiome and cardiovascular disease: current knowledge and clinical potential. Am J Physiol Heart Circ Physiol 2019;317: 923-938.
- Dahanukar SA and Thatte UM: Ayurveda Revisited - Ayurveda in the Light of Contemporary Medicine, Chapter 2: Historical Survey of the Evolution of Ayurveda, Popular Prakashan, Bombay,1st Edition pp.10,1989.
- Prajapati S and Acharya R: Pandu (anaemia): an Ayurvedic literature review. International J Res Ayur Pharma 2017; 8(5): 140-145.
- Panda AK, Mishra S, and Mohapatra SK: Iron in Ayurvedic Medicine. International J Adv Dev Res 2011; 2: 287-293.
- Shastri K and Sharma PSS:LohaVirachitRasatarangini, 20th Tarang. Motilal Banarasidas, Edition 11th reprint 487 -514, 1982
- Nadkarni KN:Mandura - Feroso -Ferric Oxide (FerriPeroxidum Rubrum), Popular Prakashan, Bombay, 2nd Reprint of 3rd revised Edition 1989;pp.62 – 66.
- Pandey PS: An attempt to standardize Swarna Makshika - A mineral drug of Ayurveda. International J Ayur Med. 2019;10(4):333-337.
- Sri Vagbhatacharya:Rasaratnasamuchhaya edited with the Suratnojjvalahindi commentary by Ambikadatta Shastri, Varanasi, ChaukhambaAmarabharatiPrakashana, 2010.
- Singh N and Reddy KRC: Particle size estimation and elemental analysis of Lauhabhasma. International J Res Ayur Pharma 2011;2(1):30-35.
- Jha CB, Bhattacharya B, and Narang KK:Bhasmas as natural nanorobots: The biorelevant metal complex. Journal of Tradt Nat Med. 2015;1(1):2-9.
- Pareek A and Bhatnagar N: Revisiting ancient therapeutic potential of Ayurvedic Bhasma. Int J Pharm Sci Res 2018;9(8):3150-3165.
- Miraj S and Kiani S: Bioactivity of Sesamum indicum: A review study. Der Pharmacia Lettre. 2016;8(6):328-334.
- Wu M-S, Aquino LBB, Barbaza MYU, Chieh-Lun Hsieh C-L, De Castro-Cruz KA, Yang L-L, and Tsai P-W: Anti-Inflammatory and Anticancer Properties of Bioactive Compounds from Sesamum indicum L.—A Review. Mol. 2019;24(24):4426.
- Widowati W, Fauziah N, Herdiman H, Afni M, Afifah E, Sari H, Kusuma W, Nufus H, Arumwardana S, and Rihibiha DD: Antioxidant and Anti-Aging Assays of Oryza Sativa Extracts, Vanillin and Coumaric Acid. J Nat Remedies. 2016;16(3):88-99.
- Palungwachira P, Tancharoen S, Phruksaniyom C, S Klungsaeng, Srichan R, Kikuchi K, and Nararatwanchai T: Antioxidant and Anti-Inflammatory Properties of Anthocyanins Extracted from Oryza sativa L. in Primary Dermal Fibroblasts. Oxid Med Cell Longev. 2019;2089817:18 pages.
- Soradech S, Reungpatthanaphong P, Tangsatirapakdee S, Panaphong K, Thammachat T, Manchun S and Thubthimthed S: Radical scavenging, antioxidant and melanogenesis stimulating activities of diiferent species of rice (Oryza sativa L.) extracts for hair treatment formulation. Thai J Pharma Sci 2016; 40: 92-95.
- Sham T-T, Yuen AC-Y, Ng Y-F, Chan C-O, DK-W Mok, and Chan SW: A Review of the Phytochemistry and Pharmacological Activities of Raphani Semen. Evidence-Based Comp Alt Med. 2013;636194:16 pages
- Chakraborty G, Manna K, Debnath B, Singh WS, Goswami S, and Maiti D: Phytochemical Analysis, Anti-oxidant and Cytotoxic Activity of Seed Coat of Macrotyloma uniflorum in Different Solvents. Nat Pdts Chem Res 2018;6(5):1-7.
- Ashraf J, Baig SG, Ahmed S, and Hasan MM: Analgesic, anti-inflammatory and diuretic activities of Macrotyloma uniflorum (Lam.) Verdc. Pakistan J Pharma Sci 2018;31(5):1859-1863.
- Gaire BP and Subedi L: Phytochemistry, pharmacology and medicinal properties of Phyllanthus emblica Linn. Chinese J Integr Med 2014;2014:828.
- Meher SK, Panda P, Das B, Bhuyan GC, and Rath KK: Pharmacological Profile of Terminalia chebula Retz. and Willd. (Haritaki) in Ayurveda with Evidences. Res J PharmacolPharmacody 2018;10(3):115-124.
- Sireeratawong S, Jaijoy K, Khonsung P, and Soonthornchareonnon N: Analgesic and anti-inflammatory activities of the water extract from Terminalia chebulaRezt. Af J Tradt Comp Alt Med 2014;11(6):77-82.
- Gupta A, Kumar R, Ganguly R, Singh AK, Rana HK, and Pandey AK: Antioxidant, anti-inflammatory and hepatoprotective activities of Terminalia bellirica and its bioactive component ellagic acid against diclofenac induced oxidative stress and hepatotoxicity. Toxicol Rep 2021; 8:44-52.
- Niranjan A and Prakash D: Chemical constituents and biological activities of turmeric (Curcuma longa L.) - A review. J Food Sci Technol 2008; 45(2): 109-116.
- De Mejia EG, Ramirez-Mares MV, and Puangpraphant S: Bioactive components of tea: Cancer, inflammation and behavior. Brain, Behavior, and Immunity 2009; 23(6): 721-731.
- Thitimuta S, Pithayanukul P, Nithitanakool S, Bavovada R, Leanpolchareanchai J and Saparpakorn P: Camellia sinensis Extract and Its Potential Beneficial Effects in Antioxidant, Anti-Inflammatory, Anti-Hepatotoxic, and Anti-Tyrosinase Activities. Mol 2017; 22: 401-414.
- Muhammad G, Hussain MA, Jantan I and Bukhari SNA: Mimosa pudica L., a High-Value Medicinal Plant as a Source of Bioactives for Pharmaceuticals. Comprehensive Rev Food Sci Food Safety 2016; 15: 303-315.
- Nwaehujor CO, Udegbunam RI, Ode JO and Udegbunam SO: Analgesic anti-inflammatory anti-pyretic activities of Garcinia hydroxybiflavanonol (GB1) from Garcinia kola. J Korean Soc Appl Biol Chem 2015; 58(1): 91-96.
- Huang WH, An-Rong L and Ching-Huey Y: Antioxidative and Anti-Inflammatory Activities of Polyhydroxy flavonoids of Scutellaria baicalensis Biosci Biotech Biochem 2006; 70(10): 2371-2380.
- Giap TH, Dung NA, Thoa HT, Dang NH, Dat NT, Hang NTM, Cuong PV, Hung NV, Minh CV, Mishchenko NP, Fedoreev SA and Thanh LN: Phthalides and Other Metabolites from Roots of Ligusticumwallichii. Chem Nat Comp 2018; 54(1): 34-37.
- Liu J, Zhang F, Ravikanth V, Olajide OA, Li C and Wei L-X: Chemical Compositions of Metals in Bhasmas and Tibetan Zuotai Are a Major Determinant of Their Therapeutic Effects and Toxicity. Evidence-Based Comp Alt Med 2019; 1697804: 13.
- Nardi GM, Januario AGF, Freire CG, Megiolaro F, Schneider K, Perazzoli MRA, Nascimento SRD, Gon AC, Mariano LNB, Wagner G, Niero R, and Locatelli C: Anti-inflammatory Activity of Berry Fruits in Mice Model of Inflammation is based on Oxidative Stress Modulation. Pharmacognosy Res 2016; 8(1): 42-S49.
- Rainey NE, Moustapha A, Saric A, Nicolas G, Sureau F, and Petit PX: Iron chelation by curcumin suppresses both curcumin-induced autophagy and cell death together with iron overload neoplastic transformation. Cell Death Discovery 2019; 5: 150.
- Kumar A and Garai AK: A clinical study on Pandu Roga, iron deficiency anemia, with Trikatrayadi Lauha suspension in children. J Ayur Int Med 2012; 3(4): 215-22.
- Khot BM, Patil AJ, and Kakad AC: Comparative clinical study of Dhatri Lauha and Navayasa Lauha in Garbhini Panduroga with reference to anemia in pregnancy. IOSR J Dental Med Sci 2013; 11(1): 28-33.
- Kumari P, Sastry R, Babu V, and Ravindar K: Effect of Kayyonyadichurna in the management of Pandu roga (Anaemia). International J Ayur Med 2013; 41(1): 1-3.
- Tekam V, Pushpalatha B, Bharathi K, and Kadam S: management of Garbhini Pandu with Draksha Ghrita and Lakshmana Lauha: a comparative clinical study. Ayushdhara 2020; 7(4): 2781-2790.
- Tubaki BR, Benni JM, Rao N and Prasad UNR: Effect of Ayurveda medications (KasīsaBhasma and DhātrīAvaleha) on iron deficiency anaemia: A randomized controlled study. Ancient Science of Life 2016; 36(1): 48-55.
- Sannd R, Meena H, Das B, Yadav B, Dua P, Khanduri S Rana RK, Singhal R, Bhuyan GC, Sarada O, Srikanth N, Padhi MM and Singh K: Clinical efficacy and safety of Punarnavadimandura and Dadimadighrita in the management of iron deficiency anemia: A prospective open-label multicenter study. J Res Ayur Sci 2017; 1: 1-8.
- Yadav B, Meena BR, Singh H and Sharma H: Clinical Efficacy and Safety of NavayasaChurna in the Management of Iron Deficiency Anemia. J Res Ayur Sci 2017; 1(2): 117-124.
- Kumar S, Chaudhary M and Chaudhary V: Clinical study to determine the role of Guda Haritaki and Punarnava Mandura in the management of Pandu Rogaw.s.r. to iron deficiency anaemia among children. International J Ayur Pharma Res 2019; 7(9): 8-18.
- Dhakad R, Gowswami B, and Prasad M: A clinical case study on the Ayurvedic management of Pandu Roga (anaemia). Inter Res J Ayur Yoga 2021; 4(1): 100-107.
- Perron NR and Brumaghim JL: A review of the antioxidant mechanisms of polyphenol compounds related to iron binding. Cell Biochem Biophy 2009; 53: 75-100.
- Kalinowski DS and Richardson D: The evolution of iron chelators for the treatment of iron overload disease and cancer. Pharmacol Rev 2005; 57: 547-583.
- Liu ZD and Hider RC: Design of iron chelators with therapeutic application. Coordination Chem Rev 2002; 232(1-2): 151-171.
- Zhou T, Winkelmann G, Dai ZY and Hider RC: Design of clinically useful macromolecular iron chelators. J Pharm Pharmacol 2011; 63(7): 893-903.
- Buss JL, Torti FM and Torti SV: The role of iron chelation in cancer therapy. Curr Medi Chem 2003; 10: 1021-1034.
- Buss JL, Arduini E, Shepard KC and Ponka P: Lipophilicity of analogs of pyridoxal isonicotinoyl hydrazone (PIH) determines the efflux of iron complexes and toxicity in K562 cells. Biochem Pharmacol 2003; 65(3): 349-60.
- Hatcher HC, Singh RN, Torti FM and Torti SV: Synthetic and natural iron chelators: therapeutic potential and clinical use. Future Medi Chem 2009; 1(9): 1643-1670.
- Hatcher H, Planalp R, Cho J, Torti FM and Torti SV: Curcumin: from ancient medicine to current clinical trials. Cell Mol Life Sci 2008; 65(11): 1631-1652.
- Dangles O and Dufour C: Flavonoid–protein interactions. In: Andersen, O and Markham K: (Eds.), Flavonoids: Chemistry, Biochemistry & Applications. CRC Press, Boca Raton, USA 2006; 443-469.
- Aron PM and Kennedy JA: Flavan-3-ols: Nature, occurrence and biological activity. Mol Nutr Food Res 2008; 52(1): 79-104.
- Manach C, Scalbert A, Morand C, Remesy C, and Jimenez L: Polyphenols: food sources and bioavailability. Am J Clin Nutr 2004; 79(5): 727-747.
- Bravo L: Polyphenols: chemistry, dietary sources, metabolism and nutritional significance. Nutr Rev 1998: 56; 317-333.
- Hu JP, Calomme M, Lasure A, De Bruyne T, Pieters L, Viletinck A and Beghe DAV: Structure-activity relationship of flavonoids with superoxide scavenging activity. Biological Trace Element Res 1995; 47: 327-331.
- Korkina LG and Afanas'ev IB: Antioxidant and chelating properties of flavonoids. Adv Pharma 1997; 38: 151-163.
- Srichairatanakool S, Ounjaijean S, Thephinlap C, Khansuwan U, Phisalaphong C and Fucharoen S: Iron-Chelating and Free-Radical Scavenging Activities of Microwave-Processed Green Tea in Iron Overload. Hemoglobin 2006; 30(2): 311-327.
- Nkhili E, Loonis M, Mihai S, Hajji HE, and Dangles O: Reactivity of food phenols with iron and copper ions: binding, dioxygen activation and oxidation mechanisms. Food and Function 2014. 5(6): 1186-1202.
- Guo M, Perez C, Wei Y, Rapoza E, Su G, Bou-Abdallah F and Chasteen ND: Iron-binding properties of plant phenolics and cranberry's bioeffects. Dalton Transactions 2007; (43): 4951-4961.
- Halliwell B: Dietary Polyphenols: Good, Bad, or Indifferent for Your Health? Cardiovasc Res 2007; 73: 341-347.
- Hider RC, Liu ZD and Khodr HH: Metal chelation of polyphenols. Methods in Enzymol 2001; 335: 190-203.
- Manach C, Williamson G, Morand C, Scalbert A and Rémésy C: Bioavailability and bioefficacy of polyphenols in humans. I. Review of 97 bioavailability studies. Am J Clin Nutr 2005; 81(1): 230-242.
- Williamson G and Manach C: Bioavailability and bioefficacy of polyphenols in humans. II. Review of 93 intervention studies. Am J Clin Nutr 2005; 81(1): 243-255.
- Frederiksen H, Mortensen A, Schroder M, Frandsen H, Bysted A, Knuthsen P and Rasmussen SE: Effects of red grape skin and seed extract supplementation on atherosclerosis in Watanabe heritable hyperlipidemic rabbits. Mol Nutr Food Res 2007; 51: 564-571.
- Bagchi D, Swaroop A, Preuss HG and Bagchi M: Free radical scavenging. Antioxidant and cancer chemoprevention by grape seed proanthocyanidin: an overview. Mutation Res Fundamental Mol Mechanisms Mutagenesis 2014; 768: 69-73.
- Sato M, Maulik G, Ray PS, Bagchi D and Das DK: Cardioprotective effects of grape seed proanthocyanidin against ischemic reperfusion injury. J Mol Cell Cardiol 1999; 31: 1289-1297.
- Maffei FR, Carini M, Aldini G, Berti F, Rossoni G, Bombardelli E and Morazzoni P: Procyanidines from Vitis vinifera seeds protect rabbit heart from ischemia/reperfusion injury: antioxidant intervention and/or iron and copper sequestering ability. Planta Med 1996; 62: 495-502.
- Spencer JPE, Abd-el-Mohsen M, and Rice-Evans C: Cellular uptake and metabolism of flavonoids and their metabolites: implications for their bioactivity. Arch Biochem Biophy 2004; 423(1): 148-61.
- Porter ML, Krueger CG, Wiebe DA, Cunningham DG and Reed JD: Cranberry proanthocyanidins associate with low-density lipoprotein and inhibit in vitro Cu2+-induced oxidation. JSci Food Agri 2001; 81(14): 1306-1313.
- Yan X, Murphy BT, Hammond GB, Vinson JA and Neto CC: Antioxidant activities and antitumor screening of extracts from cranberry fruit (Vaccinium macrocarpon). JAgri Food Chem 2002; 50(21): 5844-5849.
- Sreeram NP, Adams LS, Hardy ML and Heber D: Total cranberry extract versus its phytochemical constituents: antiproliferative and synergistic effects against human tumor cell lines. J. Agric Food Chem 2004; 52: 2512-2517.
- Mandel S, Weinreb O, Reznichenko L, Kalfon L and Amit T: Green tea catechins as brainpermeable, non toxic iron chelators to “iron out iron” from the brain. J Neural Trans Suppl 2006; 71: 249-257.
- Guo Q, Zhao B, Li M, Shen S and Xin W: Studies on protective mechanisms of four components of green tea polyphenols against lipid peroxidation in synaptosomes. BiochimBiophy Acta 1996; 1304: 210-222.
- Kumamoto M, Sonda T, Nagayama K and Tabata M: Effects of pH and metal ions on antioxidative activities of catechins. BiosciBiotechnolBiochem 2001; 65: 126-132.
- Grinberg LN. Newmark H, Kitrossky N, Rahamim E, Chevion M and Rachemilewitz EA: Protective effects of tea polyphenols against oxidative damage to red blood cells. BiochemPharmacol 1997; 54(9): 973-978.
- Friedman M, Mackey BE, Kim HJ, Lee IS, Lee KR, Lee SU, Kozukue E and Kozukue N: Structure-activity relationships of tea compounds against human cancer cells. JAgri Food Chem 2007: 55(2): 243-53.
- Kaur S, Greaves P, Cooke DN, Edwards R, Steward WP, Gescher AJ and Marczylo TH: Breast cancer prevention by green tea catechins and black tea theaflavins in the C3(1) SV40 T,t antigen transgenic mouse model is accompanied by increased apoptosis and a decrease in oxidative DNA adducts. JAgri Food Chem 2007; 55: 3378-3385.
- Weisburger JH, Rivenson A, Garr K and Aliaga C: Tea, or tea and milk, inhibit mammary gland and colon carcinogenesis in rats. Cancer Lett 1997; 114: 323-327.
- Zhou JR, Yu L, Mai Z and Blackburn GL: Combined inhibition of estrogen-dependent human breast carcinoma by soy and tea bioactive components in mice. Int J Cancer 2004; 108: 8-14.
- Mandel SA, Avramovich-Tirosh Y, Reznichenko L, Zheng H, Weinreb O, Amit T and Youdim MBH: Multifunctional activities of green tea catechins in neuroprotection. Modulation of cell survival genes, iron-dependent oxidative stress and PKC signaling pathway. Neurosignals 2005; 14: 46-60.
- Sutherland BA, Shaw OM, Clarkson AN, Jackson DN, Sammut IA and Appleton I: Neuroprotective effects of (−)-epigallocatechin gallate following hypoxia-ischemia-induced brain damage: novel mechanisms of action. The FASEB J 2005; 19: 258-260.
- Aktas O, Prozorovski T, Smorodchenko A, Savaskan NE, Lauster R, Kloetzel P-M, Infante-Duarte C, Brocke S, and Zipp F: Green tea epigallocatechin-3-gallate mediates T cellular NF-κ B inhibition and exerts neuroprotection in autoimmune encephalomyelitis. J Immunol 2004; 173: 5794-5800.
- Haque AM, Hashimoto M, Katakura M, Tanabe Y, Hara Y and Shido O: Long-term administration of green tea catechins improves spatial cognition learning ability in rats. J Nutr 2006; 136: 1043-1047.
- Rezai-Zadeh K, Shytle D, Sun N, Mori T, Hou H, Jeanninton D, Ehrhart J, Townsend K, Zeng J, Morgan D, Hardy J, Town T and Tan J: Green tea epigallocatechin-3-gallate (EGCG) modulates amyloid precursor protein cleavage and reduces cerebral amyloidosis in Alzheimer transgenic mice. J Neurosci 2005; 25: 8807-8814.
- Jiao Y, Wilkinson J, Christine Pietsch E, Buss J, Wang W, Planalp R, Torti FM and Torti SV: Iron chelation in the biological activity of curcumin. Free Radic Biol Med 2006; 40: 1152-1160.
- Jiao Y, Wilkinson J, Di X, Wang W, Hatcher H, Kook ND, D’Agostino R, Knovich MA, Torti FM and Torti SV: Curcumin, a cancer chemopreventive and chemotherapeutic agent, is a biologically active iron chelator. Blood 2009; 113: 462-469.
- Mandel SA, Amit T, Kalfon L, Reznichenko L, and Youdim MB: Targeting multiple neurodegenerative diseases etiologies with multimodal-acting green tea catechins. J Nutr 2008; 138:1578-1583.
- Weinreb O, Amit T, Mandel S and Youdim MBH: Neuroprotective molecular mechanisms of (2)-epigallocatechin3-gallate: a reflective outcome of its antioxidant, iron chelating and neuritogenic properties. Genes Nutr 2009; 4: 283-296.
- Bernabe-Pineda M, Ramirez-Silva MT, Romero-Romo MA, Gonzalez-Vergara E and Rojas-Hernandez A: Spectrophotometric and electrochemical determination of the formation constants of the complexes Curcumin–Fe(III)–water and Curcumin– Fe(II)–water. Spectrochimica Acta Part A: Mol Biomol Spectr 2004; 60: 1105-1113.
- Sun AY, Wang Q, Simonyi A, and Sun GY: Botanical phenolics and brain health. Neuromol Med 2008; 10: 259-274.
- Sriram N, Kalayarasan S, Ashokkumar P, Sureshkumar A and Sudhandiran G: Diallyl sulfide induces apoptosis in Colo 320 DM human colon cancer cells: involvement of caspase-3, NF-kappaB, and ERK-2. Mol Cell Biochem 2008; 311: 157-165.
- Tang LL, Ye K, Yang XF and Zheng JS: Apocynin attenuates cerebral infarction after transient focal ischaemia in rats. JInternational Med Res 2007; 35: 517-522.
- Wang Q, Tompkins KD, Simonyi A, Korthuis RJ, Sun AY and Sun GY: Apocynin protects against global cerebral ischemia–reperfusion-induced oxidative stress and injury in the gerbil hippocampus. Brain Res 2006; 1090: 182-189.
- Dhanasekaran M, Tharakan B and Manyam BV: AntiParkinson drug – Mucuna pruriens shows antioxidant and metal chelating activity. Phytother Res 2008; 22:6-11.
- Guo M, Perez C, Wei Y, Rapoza E, Su G, Bou-Abdallah F and Chasteen ND: Iron-binding properties of plant phenolics and cranberry’s bioeffects. Dalton Transactions 2007; 21(43): 4951-4961.
How to cite this article:
Udipi SA, Joshi J, Singh G, Sawarkar S, Phadke A, Prabhu A, Rastogi N and Vaidya AB: Phytoactives as chelators of iron with potential to mitigate its side effects and enhance iron absorption. Int J Pharm Sci & Res 2022; 13(8): 3078-02. doi: 10.13040/IJPSR.0975-8232.13(8).3078-02.
All © 2022 are reserved by International Journal of Pharmaceutical Sciences and Research. This Journal licensed under a Creative Commons Attribution-NonCommercial-ShareAlike 3.0 Unported License.
Article Information
12
3078-3102
1336 KB
477
English
IJPSR
Shobha A. Udipi *, Jayashree Joshi, Gurmeet Singh, Sujata Sawarkar, Aashish Phadke, Arati Prabhu, Neerja Rastogi and Ashok B. Vaidya
Hon Director Integrative Nutrition and Ayurceuticals, Kasturba Health Society-Medical Research Centre, Vile Parle(W), Mumbai, Maharashtra, India.
drshobhaudipi@gmail.com
08 December 2021
17 January 2022
28 April 2022
10.13040/IJPSR.0975-8232.13(8).3078-02
01 August 2022