Open access peer-reviewed chapter

Diet-Related Thalassemia Associated with Iron Overload

Written By

Somdet Srichairatanakool, Pimpisid Koonyosying and Suthat Fucharoen

Reviewed: 05 March 2020 Published: 14 October 2020

DOI: 10.5772/intechopen.91998

From the Monograph

Personalized Medicine, in Relation to Redox State, Diet and Lifestyle

Edited by Faik Atroshi

Chapter metrics overview

856 Chapter Downloads

View Full Metrics

Abstract

Thalassemia is an inherited disease caused by the genetic disorder of α- and β-globin genes, resulting in ineffective erythropoiesis and chronic anemia. Transfusion-dependent β-thalassemia patients require red cell transfusion to maintain their blood hemoglobin level in the normal range, whereas non-transfusion-dependent thalassemia patients increase duodenal absorption of dietary iron in an attempt to accelerate erythropoiesis. These changes give rise to iron overload, oxidative stress, organ dysfunction, and other complications. Effective iron chelators are necessary to achieve negative iron balance and to relieve such complications associated with iron overload. Some pharmaceuticals such as hydroxyurea, N-acetylcysteine, ascorbic acid, vitamin E, and glutathione are also given to thalassemia patients in order to overcome oxidative cell and tissue damage and to generate a better quality of life. Interestingly, functional natural products (such as mango, tea, caffeine, and curcumin), vegetables, and cereal (e.g., rice) are helpful for their health-providing properties by supplementing the endogenous antioxidant defensive power in the body. Natural products exhibit many pharmacological activities, but they are safer if used in the traditional manner.

Keywords

  • thalassemia
  • personalized medicine
  • antioxidant
  • green tea
  • functional fruits
  • iron

1. Introduction

Thailand is one of the countries located in Southeast Asia (SEA) with an ongoing thalassemia endemic and has been affected by this inherited disease for a long time. In 2012, we had an official meeting for reviewing progression in the field to develop a good clinical practice guideline (CPG) for thalassemia management in Thailand.

Advertisement

2. Etiology of thalassemia

Thalassemia is an inherited autosomal recessive disorder of hemoglobin molecules (ineffective erythropoiesis) that is characterized by an imbalanced α- and β-globin chain synthesis. The accumulation of unbound α-globin chains in erythroid cells is the major cause of pathology in β-thalassemia. Stimulation of γ-globin chain synthesis can relieve disease severity because it combines with the α-globin chain to form a fetal hemoglobin (Hb F). The disease occurs prevalently from Southeast Asia to the Mediterranean.

2.1. α-Thalassemia

α-Thalassemia is due to an impaired production of α-globin chains from 1, 2, 3, or all 4 of the α-globin genes, leading to a relative excess of β-globin chains. The severity of the disease is based on how many genes are affected. Four clinical conditions of increased severity are recognized: two carrier states, α+-thalassemia caused by the deletion or dysfunction of one of the four α-globin genes, and α0-thalassemia resulting from deletion or dysfunction of two α-globin genes in cis. The two clinically relevant forms are Hb Bart’s hydrops fetalis syndrome and Hb H disease. Patients with Hb Bart’s hydrops fetalis syndrome (homozygous α-thalassemia) have nonfunctioning α-globin genes (genotype α-thal 1/α-thal 1 or − −/− −) and mostly die before birth. Mothers usually suffer hypertension, edema, and toxic pregnancy. Hb H disease patients carry only one functioning α-globin gene (genotype α-thal 1/α-thal 2 or − −/− α, and α-thal 1/Hb Constant Spring (CS) or −/αCSα) and mostly suffer mild-to-severe anemia, jaundice, febrile, and splenomegaly and hepatomegaly. α-Thalassemia is prevalent in tropical and subtropical regions similar to other common globin gene disorders such as β-thalassemia and sickle cell anemia where malaria was and still is an epidemic. As a consequence of massive population migrations, α-thalassemia has become a relatively common clinical problem in North America, Europe, and Australia [1, 2, 3].

In northeast Thailand, thalassemia patients suffered with Hb H disease mostly due to the interaction of α-thalassemia 1 (SEA type) with the Hb CS, the deletion of three α-globin genes with the SEA type α-thalassemia 1 and the 3.7- or 4.2-kb deletion of α-thalassemia 2, and the interaction of the SEA α-thalassemia 1 with the Hb Pakse [4]. In Cambodia, α-globin gene mutation was mostly caused by the α-(3.7) (rightward) deletion (frequency 0.098–0.255), α-thal-1 (– –(SEA)) (frequency 0.008–0.011), and α-thal-2 [-alpha(4.2) (leftward deletion)] (frequency 0.003–0.008) [5].

2.2. β-Thalassemia

Human β-thalassemia is characterized by the deficient production of the β-globin chains of adult hemoglobin (Hb A), typically due to mutations of the β-globin gene. Over 200 mutations have been identified in this gene, and the type of mutation can influence the severity of the disease. There are three main types of β-thalassemia, listed in order of decreasing severity: homozygous β-thalassemia major (TM) (genotype β00) caused by mutations in both alleles, β-thalassemia intermedia (TI) (genotype β0+, β++, and β+E) caused by diverse mutations, and heterozygous β-thalassemia minor caused by single mutation, including hereditary persistent fetal hemoglobin (HPFH). TI patients usually become mildly anemic (baseline Hb level 7–10 g/dl) and have widely varying severity. Some patients require blood transfusion and chelation to promote their growth in childhood and prevent bone deformities in adults and sometimes get splenectomy due to hypersplenism and mechanical encumbrance. Enhancing Hb F synthesis is useful in some patients, and anti-oxidative compounds were found not to improve blood Hb levels. Stem cell transplantation and gene therapy are possible in well-developed countries but limited in developing countries and in some severe cases. Many complications such as pulmonary hypertension, thrombosis, hypercoagulability, pseudoxanthoma elasticum, and osteoporosis are reported in TI patients and can affect their treatment [6].

β-Thalassemia hemoglobin E (Hb E) (genotype β0E or β +E) is most prevalent in SEA countries including Thailand where the carrier frequency is around 50%. The interaction of thalassemia Hb E and β-thalassemia results in a clinical spectrum ranging from a condition indistinguishable from TM to a mild form of TI. Three categories can be identified depending on symptoms as followed: asymptomatic (normal Hb level), mild (baseline Hb level <9.0 g/dl), moderate (baseline Hb level 7–9 g/dl), and severe (baseline Hb level <7.0 g/dl). In transgenic mice, homozygous beta-knockout (BKO) thalassemia shows many clinical features of red blood cells (RBC) indices, in particular mild anemia similar to human TI. The abnormalities include decreased blood Hb concentration, hematocrit (Hct), numbers and osmotic fragility of RBC, and the increase of reticulocyte count. Additionally, Perl’s staining and colorimetric assays shows deposition of iron in the spleen, liver, and kidneys but not in the heart [7].

Advertisement

3. Anemia in thalassemia

The accumulation of excess unbound α-globin chains in erythroid cells of β-thalassemia patients can result in RBC hemolysis and anemia; nevertheless, stimulation of γ-globin gene to produce γ-globin chain which can combine with the α-globin to form Hb F is a therapeutic approach. Like cell apoptosis, eryptosis is a programmed cell death or suicidal death of erythrocytes which is characterized by shrinkage, membrane bleb, activation of proteases (e.g., caspase and calpain) after oxidative stress, and phosphatidylserine (PS) exposure at the outer plasma membrane leaflet of the affected RBC. Eryptosis can be triggered by osmotic shock, energy depletion, hyperthermia, curcumin, ceramide, prostaglandin E2, platelet-activating factor, valinomycin, amyloid peptide, hemolysin, chlorpromazine, cyclosporine, paclitaxel, stressors-induced injury, and iron-induced oxidative stress. In contrast, it is inhibited by erythropoietin (EPO), catecholamines, and nitric oxide (NO). Eryptosis is probably a useful mechanism to get rid of defective RBC and infectious agents. Nonetheless, excessive eryptosis found in iron deficiency, intoxication of metals (such as Al, Cu, Pb, and Hg), xenobiotics, β-thalassemia, sickle cell disease (SCD), glucose-6-phosphate dehydrogenase (G6PD) deficiency, hereditary spherocytosis, paroxysmal nocturnal hemoglobinuria, myelodysplastic syndrome (MDS), phosphate depletion, sepsis, hemolytic uremic syndrome, renal insufficiency, diabetes, pathogenic infection (e.g., malaria, mycoplasma, and hemolysin-producing bacteria), and Wilson’s disease can result in short lifespan and microvesicles of the RBC, consequently leading to anemia and impaired microcirculation [8, 9, 10]. Synthetic compounds and natural products of interests need to be investigated to elucidate their therapeutic potential of inhibitors of excessive eryptosis in β-thalassemia with chronic anemia.

Advertisement

4. Iron overload in thalassemia

4.1. Pathophysiology and complications

Iron overload in thalassemia is assessed with an increase of plasma iron and transferrin saturation, the presence of redox iron as non-transferrin-bound iron (NTBI) and labile plasma iron (LPI), and a high deposition of tissue iron in the forms of hemosiderin, ferritin, and labile iron pools (LIP). Excessive iron accumulation in the vital organs is the cause of liver diseases (e.g., hepatitis, hepatic fibrosis, and hepatocellular carcinoma), cardiomyopathies (e.g., cardiac arrhythmia and heart failure), and endocrinopathies (e.g., diabetes, growth retardation, defective puberty, hypopituitarism, hypogonadism, and hypoparathyroidism) [11, 12]. Iron overload can be caused by an increase of dietary iron absorption due to chronic anemia and by multiple blood transfusions to maintain normal blood Hb level. Under incomplete or partial synthesis of β-chains of Hb in β-thalassemia patients, the remaining excessive α-globin chains are unstable and eventually precipitate, causing RBC membrane damage [13]. The affected RBCs are prematurely hemolyzed in the bone marrow and spleen, resulting in increased RBC turnover, ineffective erythropoiesis, and severe anemia, so patients require regular blood transfusions to prevent the anemia and ischemia. Though thalassemia patients do not receive transfusions, abnormal iron absorption produces an increase in the body iron burden evaluated at 2–5 g per year [14]. Regular blood transfusions (420 ml/unit of donor blood equivalent to 200 mg of iron) lead to double this iron accumulation. Consequently, iron accumulation introduces progressive damage in the liver, heart, and in endocrine glands. Circulating NTBI as well as LPI is detected whenever the capacity of transferrin to incorporate iron derived from either gastrointestinal tract or reticuloendothelial (RE) cells becomes a limiting factor. Both forms of toxic iron appear primarily in transfused patients where the total iron-binding capacity (TIBC) has been surpassed [15]. Pathologically, the NTBI fraction seems to be translocated across cell membrane irregularly, while the LPI is redox active and susceptible to chelation [16].

4.2. Redox iron catalysis

In enzymatic reactions as shown in Figure 1, superoxide (O2−•) which is one of the reactive oxygen species (ROS) is normally produced by NADH:ubiquinone oxidoreductase catalysis at the complex I (I) in oxidative phosphorylation and will be converted to hydrogen peroxide (H2O2) by superoxide dismutase (SOD) catalysis (II). Hydrogen peroxide (H2O2) which is another ROS is produced by xanthine oxidase (XO) catalysis of hypoxanthine to xanthine (III) and xanthine to uric acid (IV) in purine catabolic pathway. Finally, hydrogen peroxide will be degraded or detoxified by peroxidase (POD) and catalase (CAT) to water and oxygen (V).

Figure 1.

Enzymatic production of ROS.

In Haber-Weiss/Fenton nonenzymatic reactions, iron can participate in the oxidation-reduction process known to generate ROS including hydrogen peroxide reacts to form hydroxyl radical (OH) and hydroxide anion (OH) [17] (Figure 2).

Figure 2.

Iron-catalyzed redox reactions of biological importance.

ROS can induce cell death through initiating a series of chemical reactions with many significant biomolecules, resulting in DNA oxidation, protein damage, and membrane lipids peroxidation [18, 19]. Among these ROS, hydroxyl radicals might be the most harmful to lipid and protein membrane components. The OH-induced membrane damage can be related directly to a membrane-associated Fenton reagent [20]. Oxidative cell damage has been attributed to the emergence of excessive levels of LPI that promote the production of ROS exceeding the cellular defensive capacity [21]. Cellular LIP is a source of chelatable and redox-active iron, which is transitory and serves as a crossroad of cell iron metabolism. The nature of the LIP has been revealed by its capacity to promote ROS generation in its “rise-and-fall” patterns. LIP plays a role as a self-regulatory pool that is sensed by cytosolic iron-regulatory proteins (IRPs) and its feedback regulated by an IRP-dependent expression of iron import and storage. LIP is influenced by a range of biochemical reactions that are capable of overriding the IRP regulatory loops. Excess labile iron can react with unsaturated lipids [22]. Such redox reactions lead to the damage of cells, tissues, and organs as demonstrated as the iron overload associated with β-thalassemia.

4.3. Tissue iron deposition

The spleen contains macrophages that digests hemoglobin and stores the resulting iron in ferritin. The number of blood transfusions in β-TM patients correlates with their splenic hemosiderosis and weight [23]. Hemosiderin deposition was found to be greater in the iron-overloaded livers than in the iron-overloaded spleens. Ferritin and hemosiderin increased in hepatocytes and splenic RE cells [24]. Splenectomy is one of the clinical complications of hypertransfused TM patients to reduce hyperactivity of RE macrophage; nevertheless, it may increase the iron overload.

The liver is one of the main storage organs for iron. Iron overload is considered when the ferritin level consistently exceeds 1,000 ng/ml (normal range 20–200 ng/ml). Excess free radicals can cause progressive tissue injury and eventually cirrhosis or hepatocellular carcinoma in iron overload patients whose iron is sequestrated predominantly in ferritin or hemosiderin [25]. When plasma transferrin becomes highly saturated, NTBI is detectable and rapidly transported across the hepatocyte membrane via a specific pathway. Likely, ferroportin 1 is the only protein that mediates the transport of iron out of hepatocytes and is then oxidized by ceruloplasmin and bound to transferrin [26]. Iron deposition affects hepatic parenchymal cells (hepatocytes and bile duct cells) and mesenchymal cells (endothelial cells, macrophage, and Kupffer cells) and often distributes differently from one area to another [27].

The heart is one of the most mitochondrial-rich tissues in the body, making the iron of particular importance to cardiac function. Iron as iron-sulfur cluster and cytochromes plays a key role for oxidative phosphorylation and superoxide production in the mitochondria. Iron deposition in the heart cells can lead to cellular oxidative stress and damage and an alteration of myocardial function. Heart failure is the leading cause of death among hemosiderosis β-thalassemia patients, of whom around 60% die from cardiac failure. Harmful effects of iron overload on the heart of TM patients can be monitored efficiently by using noninvasive techniques as described below, whereas invasive techniques such as Perl’s stained in biopsied heart tissue are rather impossible. Treatment with effective iron chelators can protect these patients from iron-loaded cardiomyopathy [28, 29].

Bone marrow iron deposition (186 μg/g wet weight) increases in proportion to the total body iron store in dietary iron overload of African Bantu people and Caucasian idiopathic hemochromatosis patients [30]. MDS patients who are a heterogeneous group of clonal hematopoietic stem cell malignancies show bone marrow hemosiderosis and may develop systemic iron overload.

Though hematological care is improved in homozygous transfusion-dependent β-thalassemia (TDT) patients, multi-endocrine dysfunction is still a common complication. Thyroid dysfunction is defined as overt hypothyroidism, subclinical hypothyroidism, and an exaggerated thyroid-stimulating hormone response was reported in β-thalassemia patients [31]. Possibly, growth retardation, secondary hypogonadotropic hypogonadism and hypothyroidism are originated from pituitary damage primarily caused by iron overload and oxidative stress [32, 33]. Approximately half of patients’ pituitary gland dysfunction associated with iron overload is irreversible [34].

The redox irons in TDT patients with TM and TE are catalytically harmful to adrenal glands and can cause adrenal insufficiency [35]. Though all TM patients were nondiabetic, some of them decreased in the oral glucose tolerance test. They showed normal response of cortisol to insulin and adrenocorticotropic hormone stimulation. Moreover, the β-cell pancreatic function and adrenal cortical function were depressed in the severely iron-loaded. Recently, Koonyosying and colleagues have demonstrated green tea extract could reduce cellular the levels of iron and ROS and increase insulin secretion in concentration-dependent manner in iron-loaded pancreatic cell line (RINm5F), indicating the amelioration of oxidative stress and endocrinal improvement of pancreatic β-cells [36]. They also found that eltrombopag, which is a thrombopoietin receptor agonist and potent metal ion-chelating agent, efficiently decreased cellular levels of iron and ROS from cultured HuH7, H9C2, and RINm5F cells and restored insulin secretion from iron-loaded RINm5F cells [37].

4.4. Assessment of tissue iron content

Serum ferritin level has been used as a surrogate biochemical marker to correlate closely with liver iron concentration for a long time and would be a valuable alternative to assess visceral iron overload in heavily iron-loaded TM patients [38]. Sophisticated noninvasive magnetic resonance imaging, magnetic iron detector susceptometry, superconducting quantum interference device, and nuclear resonance scattering techniques can also be used to assess iron status in tissues. Alternatively, invasive tissue biopsied needle aspiration associated with ferrozine colorimetry or graphite-furnace atomic absorption spectrometry is routinely quantitated for nonheme iron in tissues (e.g., myocardium, liver, pancreas, adrenal glands, anterior pituitary gland, and skin) [39, 40, 41]. These methods are all valuable when evaluating iron load in the tissues and monitoring the response of different organs to chelation therapy.

Advertisement

5. Thrombotic events in thalassemia

Heart failure and arrhythmia are the main causes of death in TM patients with cardiac siderosis, pulmonary hypertension, and thrombosis and also the major cardiovascular complications in TI patients possibly due to pro-atherogenic biochemical factors (e.g., iron status and lipid profile) [42, 43]. Hypercoagulable pulmonary microthromboembolism in Thai pediatric TE patient was previously investigated [44]. After splenectomy TI patients mostly had thrombosis, thrombocytosis, and lower levels of anticoagulation inhibitors (e.g., protein S, protein C, and antithrombin III) [45]. Splenectomy promotes portal vein thrombosis in TM patients [46]. Ineffective erythropoiesis, chronic anemia, iron overload, and polycythemia by erythrocytosis and thrombosis are coincidently occurring in β-thalassemia patients. Signs of cerebrovascular accident (brain ischemia, hemorrhage, and infarct) and heart disease (congestive heart failure and atrial fibrillation) were described in chronically hypercoagulable thromboembolic thalassemia patients, so anticoagulant and/or antiplatelet therapy is recommended. Hypoxia and iron overload are the two major mechanisms of ROS overproduction [47]. The levels of plasma hemostatic and thrombotic markers were significantly higher in splenectomized TE patients than non-splenectomized ones, implying splenectomy increases platelet hyperactivity, blood hypercoagulability, and risk of thrombosis. ROS-induced activation of vascular endothelial cells can cause vasculitis and thrombosis, showing increased levels of many soluble adhesion molecules and von Willebrand factor (vWF) in thalassemia blood [48]. Procoagulant activity of circulating RBC microvesicles or microparticles (MPs) may contribute to thrombotic events in thalassemia hypercoagulability [49]. Carotid artery thrombus is usually associated with severe cardiovascular diseases (CVD), iron deficiency anemia, and thrombocytosis. Thromboembolic complications are documented in thalassemia patients, possibly due to aggregation of abnormal RBC and high amounts of RBC membrane-derived MPs [50]. Antioxidant treatment of β-thalassemia HbE patients can improve oxidative stress and hypercoagulable state [51]. Iron overload, in particular NTBI level, would be one of the risk factors in pulmonary thrombosis and hypertension in splenectomized non-transfusion-dependent thalassemia (NTDT) patients [52]. Iron chelators are useful and effective in the amelioration of iron overload and oxidative stress in thalassemia mice, possibly in the prevention of pulmonary thrombosis [53]. Nitric oxide (NO) synthesized from L-arginine by catalysis of nitric oxide synthase (NOS) species is a free-radical, physiologic vasodilator, and potent inhibitor of platelet function. Excessive iron-liberated heme degradation contributes to hypercoagulability [54]. Low arginine bioavailability in β-thalassemia patients can cause pulmonary hypertension and cardiopulmonary dysfunctions [55]. Splenectomy, thrombocytosis, RBC, and platelet MPs may be residual hypercoagulable/thrombotic risks in TDT patients [56, 57]. Liver inflammation and cirrhosis can involve in hypercoagulability, thrombosis, and reduced fibrinolysis [58, 59].

Advertisement

6. Treatment and implements

Strategy and approach have been suggested for the treatment and support of thalassemia patients to have better quality of life and well-being [60]. These approaches include occasional/regular blood transfusions, iron chelation therapy, antioxidant supplement, Hb F switching agents, anti-allergic drugs, antibiotics (such as antibacterial, antiviral, antifungal, and antimalarial drugs), splenectomy (in the past), dental care, and hemopoietic stem cell transplantation.

6.1. Iron chelation therapy

Iron chelation therapy aims to prevent the accumulation of toxic iron and eliminate the excess iron in TDT patients. Effective chelation and good management of the patients have been correlated with a decline in early deaths and complications [61]. Reduction of plasma and cellular chelatable iron such as NTBI, LPI, and LIP is a slow process and requires aggressive chelation therapy. The chelation will maintain the iron balance at safe levels to prevent high iron accumulation and oxidative tissue injury. Such non-iron and iron-overloaded models as RBC, cell cultures (e.g., hepatocytes, HepG2 cells, and cardiomyocytes), animals (e.g., mice, gerbils, rats, and transgenic BKO mice), and even human thalassemia patients are experimentally investigated and clinically tested to assess the safety and efficacy of iron chelators. At present, three standard iron chelators including desferrioxamine (DFO), deferiprone (DFP), and deferasirox (DFX) are widely used for the treatment of β-thalassemia patients with iron overload to prevent oxidative stress-induced organ dysfunctions and such complications (Figure 3). Combined DFO/DFP and DFP/DFX treatments can reverse endocrine complications by improving glucose intolerance and gonadal dysfunction in TDT patients [62].

Figure 3.

Chemical structures of DFO, DFP, and DFX.

Under continuous chelation therapy, many TDT patients with moderate-to-severe pituitary iron overload had normal volume and function of the pituitary gland, representing a potential therapeutic window, while some hypogonadal patients preserved their pituitary volumes and functions. Thai clinicians have reported that DFO chelation therapy for 1.5 years largely decreased serum ferritin level and improved secretion of prolactin (PRL) and growth hormone (GH) but not other pituitary hormones [63].

6.2. Supplementation of antioxidants

Compounds such as vitamins A, C, E, β-carotene, reduced glutathione (GSH), and N-acetylcysteine (NAC) and enzymes such as SOD, CAT, glutathione peroxidase (GPx), and glutathione reductase (GR) can remove free radicals by enzymatic and nonenzymatic antioxidant systems in the body (Figure 4). Since β-thalassemia patients have a higher oxidative stress level than normal people, effective antioxidants would be a complementary treatment of choice in these patients. Ideas for using drug antioxidants to eliminate oxidative tissue damage and empower antioxidant systems in thalassemia patients have been applicable for a long time [64]. Commercially available compounds included vitamin C, vitamin E, NAC, coenzyme Q10, and hydroxyurea (HU) which were used for the treatment, with vitamin E being the most popular [65, 66, 67, 68, 69, 70, 71, 72, 73, 74, 75, 76, 77, 78, 79]. Importantly, treatment with vitamin E significantly lowered the levels of plasma lipid peroxidation products and adenosine diphosphate (ADP)-challenged platelet activity in non-splenectomized and splenectomized HbE/β-thalassemia patients [80]. Regarding other anti-oxidative natural products, silymarin restored glutathione level in thalassemia patients [81]. Fermented papaya preparation (FPP) increased glutathione levels in blood cells and platelets and decreased membrane lipid peroxidation products in β-thalassemia patients [82]. Treatment with a cocktail of DFP, NAC, vitamin E, and curcumin for 1 year improved antioxidant capacity in HbE/β-thalassemia patients [80, 83]. The levels of serum vitamins A and E, Zn, Se, and Cu were lower in young thalassemia patients than in controls, whereas serum ferritin and iron levels were inversely correlated with serum retinol and selenium levels (p < 0.05). Interestingly, vitamin E and polyphenols can abolish increased oxidative stress in thalassemia patients; if given along with iron chelators, then they may provide a substantial improvement in chronic anemia and complications [84].

Figure 4.

Structures of antioxidants.

6.3. Vitamin C

Ascorbic acid or vitamin C is a simple water-soluble vitamin which cannot be enzymatically synthesized in the human body. The substance normally functions as a cofactor of proline and lysine hydroxylase in collagen synthesis. The levels of leukocyte and urinary AA are decreased in idiopathic hemochromatosis patients, TDT patients, and Bantu people [85]. Platelet vitamin C level is lower in thalassemia patients with iron overload than normal people [86]. When TM patients are treated with vitamin C, their levels of serum iron, transferrin saturation, and ferritin are increased [87]; possibly vitamin C would be involved in the mobilization of storage iron from tissues and increase oxidative damage in the patients. However, vitamin C plus vitamin E supplementation for β-thalassemia patients has benefits more than vitamin E alone in promoting their antioxidant activity [66].

6.4. Vitamin E

Vitamin E (α-tocopherol) is considered to be the most important lipid-soluble exogenous antioxidant in humans. Low serum level of vitamin E is found in homozygous TM and TE patients. Oral administration of high doses of vitamin E effectively decreased plasma lipid peroxidation in β-thalassemia patients and prolonged RBC survival in some patients [71, 88]. A therapeutic trial with vitamin E was carried out in TM and TI patients with 750–1000 IU/day for an average period of 16 months. The treated patients showed fourfold increase in both serum and RBC vitamin E levels and a reduced level of malonyldialdehyde (MDA) when compared with the untreated group [89]. Daily vitamin E supplementation for 3 months significantly increased plasma α-tocopherol levels and reduced plasma oxidant levels in splenectomized TE patients [80].

6.5. Glutathione

Glutathione (γ-glutamylcysteinylglycine) is a tripeptide synthesized by the catalysis of γ-glutamyl cysteine synthetase and glutathione synthetase in cells and indicated as a very important endogenous free-radical scavenger due to the presence of cysteine sulfhydryl group in the molecule. In addition, GR, GPx, and glutathione-S-transferase (GST) work as antioxidants to get rid of harmful free radicals mostly in the cells. Physiologically, GR together with reduced nicotinamide adenine dinucleotide phosphate functions to recycle oxidized glutathione (GSSG) back to GSH to scavenge ROS, and GPx converts hydrogen peroxide to water and oxygen. GSH is approximately 80% present in the liver. GR activity was slightly decreased in TDT patients, whereas GPx activity was not different when compared with healthy persons [90]. Blood GSH levels of α-, β-, and HbE/β-thalassemia patients with iron overload were significantly lower than those of the healthy controls [91, 92, 93, 94, 95]. Importantly, treatment with flavonoid silymarin restored a decreased GSH content in T cells of β-thalassemia major patients [81]. Though endogenous GSH content is unable to be filled up with direct consumption due to digestive peptidase activity, oral administration of some antioxidants such as vitamin E (10 mg/kg/day), commercially available FFP, silymarin tablet (140 mg three times a day), HU (10–20 mg/kg/day), NAC (2,400 mg/day), and curcumin (500 mg/day)/vitamin E cocktail can increase/restore intracellular GSH content in thalassemia patients instead.

6.6. Hydroxyurea

HU (alternatively hydroxycarbamide) is a drug of choice used for enhancing γ-globin gene expression and modifying γ-globin chain production, as a consequence of Hb F production in SCD and β-thalassemia patients. In controversy, the compound is toxic and suspected to the pathogenesis of colonic ulcerative [96]. Indeed, HU effectively increases Hb F production in patients with SCD, SCD with α-thalassemia, and TI and results in a decrease in the number of blood transfusions required [97, 98, 99]. A current clinical study in TI patients has shown HU decreased serum ferritin (50 vs. 33%), LIP (20 vs. 13%), apoptotic event (62 vs. 15%), and ROS (60 vs. 50%) levels and increased GSH level (66 vs. 25%) in the responders compared to the nonresponders [100]. In addition to the increase in Hb F synthesis, treatment with HU (30 mg/day) in β-thalassemia patients with Hb E for 3 months decreased SOD activity and MDA concentration of the RBC, probably due to inhibition of cytosolic superoxide radical and membrane lipid peroxidation [101, 102].

6.7. N-acetylcysteine (NAC)

NAC, an anti-oxidative thiol-containing compound, is able to trap ROS and reactive nitrogen species (RNS) and therefore protect cells from such free-radical-mediated damage. After crossing the cell membrane, the compound will be hydrolyzed to cysteine used for the synthesis of GSH. Importantly, NAC can protect the RBC of SCD patients and of normal subjects from oxidative stress condition [65, 103]. In vitro treatment of blood cells including RBC, platelets (PLT), and polymorphonuclear leukocytes of β-thalassemia patients with N-acetylcysteine amide increased GSH content and reduced ROS level in these cells, possibly resulting in a significant reduction in the sensitivity of thalassemia RBC to hemolysis and phagocytosis by macrophages [65]. They also showed that the intraperitoneal injection of AD4 to β-thalassemia mice (150 mg/kg) significantly reduced all parameters of oxidative stress. One β-thalassemia with hemoglobin sickle (Hb S) who received NAC (2400 mg/day) for 6 weeks showed an increase in whole-blood GSH levels and a decrease in the RBC membrane PS exposure [104]. Consistently, TDT patients who received NAC (10 mg/kg/day) for 3 months showed a decrease in total oxidative stress and total oxidative stress index and an increase in total antioxidant capacity and blood Hb level [105]. Our group has reported that treatment of β-thalassemia HbE with a cocktail of DFP, NAC, and either vitamin E or cucumin for 12 months significantly decreased levels of iron overload (e.g., NTBI, LPI, erythrocyte membrane nonheme iron) and oxidative stress (e.g., MDA and erythrocyte ROS) parameters and increased levels of blood Hb and antioxidant indicators (e.g., CAT, SOD, and GSH), suggesting an effective antioxidant property [51].

Advertisement

7. Supplementation of functional food

7.1. Curcuminoids

Curcumin (diferuloylmethane) is one of the major phytochemicals (70–80%, w/w) from the golden spice turmeric Curcuma longa Linn (family Zingiberaceae). The three main constituents of curcuminoids are curcumin, demethoxycurcumin, and bis-demethoxycurcumin, of which the important molecular structure for biological activity is diketone moiety (Figure 5).

Figure 5.

Structures of ingredients in curcuminoids.

Apparently, curcumin and its metabolites including di-, tetra-, and hexa-hydrocurcumin exhibit strong antioxidant, free-radical scavenging, anti-lipid peroxidative, antithrombotic, and anti-inflammatory activities. Many clinical investigations have addressed pharmacokinetics, safety (maximum dose 12 g/day over 3 months), and efficacy of this attractive nutraceutical against several human diseases including β-thalassemia. Many formulations of curcumin including nanoparticles, liposomal encapsulation, emulsions, capsules, tablets, and powder are available for a single and adjunctive treatment [106]. Curcumin is claimed to be a potential hexadentate iron chelator and found to remove NTBI in thalassemia serum and also suppress the ROS generation and lipid peroxidation in thalassemia RBC [83, 107, 108, 109, 110, 111]. Curcuminoids (particularly bis-demethoxycurcumin) and its metabolite (hexahydrobisdemethoxycurcumin) potentially enhanced the upregulation of γ-globin gene and synthesis of Hb F in human erythroid leukemia (K562) and primary erythroid precursor cells [112]. Curcumin is reported one of the triggers of eryptosis to allow defective RBC to escape hemolysis [8]. The oxidative stress condition in circulating RBC of TE patients is reduced after treatment with a curcumin cocktail, leading to improvement in their quality of life [83]. Curcumin markedly decreased iron deposition and lipid peroxidation product as MDA in the liver and spleen and the liver of iron-loaded rats [113].

7.2. Green tea

Tea (Camellia sinensis L., Theaceae family) is one of the most popular beverages in the world in which the products, depending on duration of fermentation, can be classified into green tea (GT), oolong tea, white tea, yellow tea, black tea, pu-erh tea, and Miang tea. GT (C. sinensis L. var japonica) is produced without any fermentation (oxidation), so the major persisting catechins are not destroyed by naturally occurring polyphenol oxidase (PPO) in fresh tea leaves. Oolong tea (C. sinensis var sinensis) is processed from tea leaves under semi-fermentation, in which β-glycosidic aroma precursors including 8-hydroxygeranyl β-d-primeveroside, trans- and cis-linalool 3,6-oxide 6-O-β-d-xylopyranosyl-β-d-glucopyranosides, and (2R,3S,4S,4aS,11bS)-3,4,11-trihydroxy-2-(hydroxymethyl)-8-(4-hydroxyphenyl)-3,4,4α, 11β-tetrahydro-2H,10H-pyrano[2′,3′:4,5]furo[3,2-g]chromen-10-one are the main volatile constituents besides the catechin derivatives. Black tea (long fermentation) and Miang tea (C. sinensis L. kuntze var assamica) require very long fermentation times depending on the manufacturing process. Miang (a northern Thai word) is a chewing tea and commonly used for gum chewing in elderly people, relief of skin burn and inflammation, and as an antidiarrheal remedy.

In industry, GT is produced from steaming or roasting fresh tea leaves at high temperatures, consequentially drying and inactivating the PPO enzymes and leaving polyphenols known as flavonols or catechins at 30–40% by weight of dry tea leaves. It contains at least four major catechin derivatives including (−)-epigallocatechin-3-gallate (EGCG), (−)-epigallocatechin (EGC), (−)-epicatechin-3-gallate (ECG), and (−)-epicatechin (EC), of which the lipophilic permeable EGCG exhibits anti-oxidative and iron-chelating activities (Figure 6) [114]. Additionally, other phenolic acids including chlorogenic acid (CGA), caffeic acid (CA), and gallic acid (GA) and flavonols including kaempferol, myricetin, and quercetin are present in green tea [115]. Green tea extract (GTE) and EGCG, which show iron-chelating and antioxidant properties [116, 117] decrease labile iron (e.g., NTBI and LPI) level and consequently deplete lipid peroxidation as well as oxidative stress in both iron-loaded rats and thalassemia mice [118, 119]. The compounds were effective in the inhibition of RBC hemolysis, resulting in a prolonged RBC lifespan and decreased iron deposition and oxidative damage in the liver [119].

Figure 6.

Structures of catechins in green tea.

TI showed higher intestinal nonheme iron absorption than TM, while tea produced 41–90% inhibition of iron absorption in these patients, suggesting that tea consumption would be recommended to thalassemia patients, particularly TI patients [120]. Logically, anti-oxidative GTE interferes duodenal absorption of dietary iron and iron-chelating properties in vitro and in vivo [116, 117, 118, 119]. The preparation also showed inhibitory effect on catecholamine secretion from isolated rabbit adrenal glands, possibly by blocking L-type calcium channels in the adrenal medullary glands [121]. Therefore, GTE might be helpful to decrease iron deposition, reduce ROS levels, and ameliorate functions of targeted endocrine glands (e.g., pancreas and adrenal cortex) in β-thalassemia models. In controversy, a study reports development of thrombotic thrombocytopenic purpura in a person after consuming a weight-loss product containing green tea [122]. Most importantly, green tea showed antithrombosis ex vivo and inhibition of cyclooxygenase 1 activity [123, 124].

In our recent study, we have produced a functional GT-CUR concentrate (Figure 7) for investigating its effects in Thai adult TDT patents. We found that the drink did not affect white blood cell and platelet numbers, Hb, and Hct but increased RBC numbers following daily consumption for 2 months. The levels of blood urea, serum alanine aminotransferase, aspartate aminotransferase, and alkaline phosphatase activity tended to decrease but neither significantly nor dose dependently. In month 1 and 2 of the treatments, there were a decrease of serum MDA (−0.07 ± 2.95 and −0.87 ± 1.68 μM, respectively), NTBI (−1.20 ± 8.03 and −3.93 ± 3.83 μM, respectively), and LPI (1.91 ± 4.99 and −1.10 ± 2.94 μM, respectively) and increase of serum antioxidant activity (5.08 ± 8.86 and 0.28 ± 13.39 mg trolox equivalent/ml, respectively). These findings suggest GT-CUR drink would increase erythropoiesis, improve liver and kidney function, and diminish oxidative stress and iron overload in thalassemia patients [125]. Surprisingly, we demonstrate that treatment of GTE (1–10 μM EGCG equivalent) decreased cellular iron approximately 45% and ROS level in a concentration-dependent manner in iron-loaded pancreatic cell line (RINm5F) when compared with control cells. Secretory insulin level was nearly 2.5-fold times the highest safe concentration of the GTE [36]. The results imply that catechin-rich GT would indeed be an effective drink to remove iron, decrease ROS, and improve pancreatic cell function thereby increasing insulin production, leading to the amelioration of diabetic complications in thalassemia patients with iron overload.

Figure 7.

GT-CUR concentration: from field to nutraceutical product.

Evidently, green tea is abundant with phytonutrient and enriched with active phytochemicals that exhibit many biological and pharmacological activities and it can be utilized for a functional drink and health benefits. Up to now, many green tea products are being marketed worldwide for many purposes in different population ages. We are eager to use our multifunctional cocktail containing green tea extract, DFP, and vitamin E to examine if the product could diminish hypercoagulability and excessive platelet activity in thalassemia patients and thrombosis-related diseases, besides iron chelation.

7.3. Coffee

Coffee is also one of the most widely consumed beverages in the world because they contain many active ingredients that are a benefit for human health. Coffee (Coffea arabica L., Coffea canephora L. family Rubiaceae) is an original crop that will be further processed to roast coffee, coffee powder, coffee brew, coffee biscuit, and coffee candy for commercial purposes. Coffee is widely naturalized in many parts of the world including Africa, Latin America, the Pacific and Caribbean Ocean, Southeast Asia, and China. In Thailand, coffee is usually cultivated on the highlands at Doi Chang and Huay Nam Khun of Chiang Rai, Doi Saket District of Chiang Mai, and Kraburi District of Ranong (Figure 8).

Figure 8.

Coffee crop in Thailand.

Caffeine (1,3,7-trimethylxanthine) is a predominant ingredient persisting in tea and coffee, which is the most widely used pharmacologic substance showing prooxidant and antioxidant and hydroxyl radical scavenger [126, 127, 128]. Coffee contains many kinds of monosaccharide including sucrose, polysaccharides, d-arabinose, d-mannose, d-glucose, d-galactose, d-rhamnose, and d-xylose in nearly equal amounts. The amounts of caffeine and CGA are slightly higher in raw arabica coffee (0.9–1.2% and 1.6–2.4% w/w, respectively) than in raw robusta coffee (5.5–8.0% and 7.0–10.0% w/w, respectively) [129]. Interestingly, only arginine and cysteine are much more abundant in the green coffee (3.61% and 2.89% for arabica 2.28% and 3.87% for robusta) when compared with the roast coffee (0% and 0.76% for arabica 0% and 0.14% for robusta). Phenolic compounds including mono- and di-caffeoylquinic acids, CA, ferulic acid, p-coumaric acid, sinapic acid, 4-hydroxybenzoic acid, and CGA were detected in spent coffee by-product [130, 131] (Figure 9). Phenolic compounds, in particular CGA in coffee was able to chelate metal ion such as Zn [132]. In controversy for CVD incidence, one report supports the administration of caffeine augments endothelium-dependent vasodilation in young healthy volunteers through an increase in NO production [133]. Approximately one third of CGA and almost all of the CA are absorbed in the small intestine of humans, so the two antioxidants might have preventive effect of CVD [134]. The 10-kDa or less fractions but no other common components (e.g., CGA, CA, caffeine, quinic acid, trigonelline hydrochloride, and 5-(hydroxymethyl)-2-furfural) in hot-water extract of coffee had antithrombin and antiplatelet activity [135]. CGA protected oxidative damage and dose dependently increased the production of NO of human aortic endothelial cells [136].

Figure 9.

Some major constituents in coffee.

Caffeine increases intracellular calcium-stimulating endothelial NOS to accelerate the production of NO which will be diffused to vascular smooth muscle cell to produce vasodilation [137]. Tocopherols are found in coffee bean oil [138]. Caffeine (300 mg, equivalent to two to three cups) is metabolized in the human body to theophylline (170 ng/ml plasma) 7 hours post-administration [139]. Tea and coffee dose dependently inhibited absorption of nonheme iron of either animal or plant food [140]. Dihydrocaffeic acid, a metabolite of CA detected in human plasma following coffee ingestion, was able to decrease ROS and increase NOS activity in human-derived EA.hy926 endothelial cells [141]. Ingestion of green coffee extract for 4 months led to the decrease in plasma level of homocysteine and improvement of human vessel reactivity [142]. Coffee ground residual has higher phenolic contents than roast coffee bean and shows inhibitory effects on the production of NO and pro-inflammatory cytokines in the macrophage [143]. Surprisingly, healthy volunteers who consumed coffee for 2 months (420 and 780 mg CGA equivalent/day) showed increase of plasma total antioxidant capacity [144]. A recent study has demonstrated coffee would counteract cerebral arterial constriction via endothelial NOS induction and smooth muscle dilation [145]. Two catechols, particularly CGA and CA which is abundant in coffee, could potentially scavenge free radicals and subsequently inhibit the production of pro-inflammatory cytokines as interleukin-8 in intestinal epithelial cells [146]. Consistent with our previous study, healthy adults consuming CGA-enriched coffee showed a significant increase of plasma antioxidant capacity when compared with the control group [144, 147]. Additionally, CGA-enriched green and roast coffee can protect oxidative damage of biomolecules in human consumers [148].

Nowadays, there are varieties of coffee products including green coffee powder, green coffee capsules, green coffee extracts, green coffee cleans detox, roast coffee, roast coffee, coffee brew, and herbal coffee that are commercially available for all-level consumers. In socioeconomics, the coffee beverage business is very popular and a growing industry in Thailand. We are applying the wonderful properties of coffee for health benefits in thalassemia patients regarding anti-oxidation, metal chelation, and antithrombosis.

Advertisement

8. Fruits and vegetables

Epicarp extracts of bergamot (Citrus bergamia Risso) containing “citropten” and “bergapten” powerfully induced the expression and differentiation of γ-globin gene in human erythroid cells (K562) and consequently the production of Hb F, suggesting a potential therapy application in β-thalassemia and sickle cell anemia [149]. Fermented papaya preparation (FPP) increased the glutathione levels in blood cells and platelets and also decreased erythrocyte ROS level and membrane lipid peroxidation product levels such as MDA and phosphatidylserine in β-thalassemia patients [82, 150]. Mango (Mangifera indica L., family Anacardiaceae) is a tropical edible fruit cultivated globally and is annually produced from March to May. The seasonal fruit gives a high yield in Thailand and can be consumed in the forms of green and yellow fruits. It was found that aqueous extracts of the stem barks and peel from Vimang mango displayed potent antioxidant, free-radical scavenging and divalent metal ion-chelating properties due to the presence of a major polyphenol “mangiferin” [151]. Consistently, our group demonstrated that aqueous and ethanolic extracts of Thai mango (M. indica var Mahachanok and M. indica var Kaew) lowered plasma levels of glucose and triglyceride in streptozotocin-induced rats. Obviously, the extract showed analgesic, anti-gastric ulcerative, and chemical-induced hepatoprotective effects in rats. In addition, the extracts increased plasma antioxidant capacity in rats and humans [152]. The results suggest fresh and fermented mangoes would be a potential functional and therapeutic food against the deleterious action of ROS generated during iron overload (e.g., β-thalassemia, Friedreich’s ataxia, hemochromatosis, and inflammation).

Rice (Oryza sativa L.) is the chief economic crop cultivated in every region of Thailand. One study demonstrated that consumption of wheatgrass juice and tablets decreased the requirement of RBC transfusions in Indian β-thalassemia patients by 25% or more [153, 154]. It was possible that pheophytin compound in the wheatgrass would increase hemoglobin synthesis. In controversy, another study showed that the juice therapy did not affect the production of hemoglobin [155]. Pancytopenia such as leukocytopenia and thrombocytopenia is observed in the chelation treatment of thalassemia; however, herbs like wheatgrass, papaya leaves, and garlic would be effective in treating single lineage cytopenias [156]. We found that ethanol extract of neem (Azadirachta indica var siamensis Valeton) leaves displayed free-radical scavenging and iron-binding activities in vitro, and the study of the extract will be extended to β-thalassemia mice with iron overload [157].

Advertisement

9. Conclusions

Regular iron chelation therapy with high dietary intake of antioxidants effectively lowers the harmfulness of iron overload-mediated oxidative tissue damage and organ dysfunctions in thalassemia patients. The supplementation with single nutrients, like antioxidants, is generally not effective in ameliorating such iron overload conditions or in slowing the progression of the disease. It is recommended that these nutrients should be consumed as part of a healthy diet/functional fruits in daily meals. Nutritional and herbal strategies for modifying the pathological and clinical courses of thalassemia disease should consider the major active ingredients, nutraceuticals, biological activities, and hematological efficacy. Moreover, pre-implant/prenatal detections of thalassemia in the fetus using sensitive and specific molecular-biological and ultrasonic techniques could block new cases and problematic carriers of hemoglobinopathies. Understanding the genetics underlying the heritable subphenotypes of thalassemia would be prognostically useful and inform us further about personalized therapeutics as well as help the discovery and development of new pharmacogenomics. An effective medical regime, adjunctive supplementation of synthetic and natural antioxidants, and caregiver education could also be important factors to prevent or treat symptoms/complications in thalassemia. Selected protocols using single or combined chelators could be designed for personalized iron chelation therapy in TDT and NTDT patients, which would effectively and safely remove all the excess toxic iron (e.g., NTBI, LPI, and LIP) and prevent cardiac, liver, and other organ damage. Finally, a reliable approach based on genomics and proteomics may be effective to build a rational personalized medicine framework that can be applied in the preclinical, clinical, and therapeutic settings of hypercoagulability in thalassemia.

Advertisement

Acknowledgments

We appreciate the Global Society for Nutrition, Environment and Health (GSNEH) for their support. We thank Emeritus Professor Robert C. Hider, PhD., King’s College London, and Mr. Michael Creswell from the University of Manchester, Manchester, UK, for English proofreading.

Symbols and abbreviations

α

alpha

β

beta

γ

gamma

ADP

adenosine diphosphate

BKO

beta-knockout

CA

caffeic acid

CAT

catalase

CGA

chlorogenic acid

CS

Constant Spring

CVD

cardiovascular diseases

DFO

desferrioxamine

DFP

deferiprone

DFX

deferasirox

EC

(−)-epicatechin

ECG

(−)-epicatechin-3-gallate

EGC

(−)-epigallocatechin

EGCG

(−)-epigallocatechin-3-gallate

EPO

erythropoietin

FPP

fermented papaya preparation

G6PD

glucose-6-phosphate dehydrogenase

GA

gallic acid

GPx

glutathione peroxidase

GR

glutathione reductase

GSH

reduced glutathione

GST

glutathione-S-transferase

GT

green tea

GTE

green tea extract

Hb

hemoglobin

Hb A

adult hemoglobin

Hb E

hemoglobin E

Hb F

fetal hemoglobin

Hb S

hemoglobin sickle

Hct

hematocrit

HPFH

hereditary persistent fetal hemoglobin

HU

hydroxyurea

IRPs

iron-regulatory proteins

LIP

labile iron pools

LPI

labile plasma iron

MDA

malonyldialdehyde

MDS

myelodysplastic syndrome

MPs

microparticles

NAC

N-acetylcysteine

NO

nitric oxide

NOS

nitric oxide synthase

NTBI

non-transferrin-bound iron

NTDT

non-transfusion-dependent thalassemia

POD

peroxidase

PPO

polyphenol oxidase

PS

phosphatidylserine

RBC

red blood cells

RE

reticuloendothelial

ROS

reactive oxygen species

SAE

Southeast Asia

SCD

sickle cell disease

SOD

superoxide dismutase

TDT

transfusion-dependent β-thalassemia

thal

thalassemia

TI

β-thalassemia intermedia

TIBC

total iron-binding capacity

TM

β-thalassemia major

vWF

von Willebrand factor

XO

xanthine oxidase

References

  1. 1. Fucharoen G, Fucharoen S, Wanhakit C, Srithong W. Molecular basis of alpha (0)-thalassemia in northeast of Thailand. The Southeast Asian Journal of Tropical Medicine and Public Health. 1995;26(Suppl 1):249-251
  2. 2. Fucharoen S, Viprakasit V. Hb H disease: Clinical course and disease modifiers. Hematology-American Society of Hematology Education Program. 2009;2009(1):26-34
  3. 3. Fucharoen S, Winichagoon P, Siritanaratkul N, Chowthaworn J, Pootrakul P. Alpha- and beta-thalassemia in Thailand. Annals of the New York Academy of Sciences. 1998;850:412-414
  4. 4. Boonsa S, Sanchaisuriya K, Fucharoen G, Wiangnon S, Jetsrisuparb A, Fucharoen S. The diverse molecular basis and hematological features of Hb H and AE Bart's diseases in Northeast Thailand. Acta Haematologica. 2004;111:149-154
  5. 5. Munkongdee T, Tanakulmas J, Butthep P, Winichagoon P, Main B, Yiannakis M, et al. Molecular epidemiology of hemoglobinopathies in Cambodia. Hemoglobin. 2016;40:163-167
  6. 6. Borgna-Pignatti C. Modern treatment of thalassaemia intermedia. British Journal of Haematology. 2007;138:291-304
  7. 7. Garrick LM, Strano-Paul LA, Hoke JE, Kirdani-Ryan LA, Alberico RA, Everett MM, et al. Tissue iron deposition in untransfused beta-thalassemic mice. Experimental Hematology. 1989;17:423-428
  8. 8. Foller M, Huber SM, Lang F. Erythrocyte programmed cell death. IUBMB Life. 2008;60:661-668
  9. 9. Lang F, Abed M, Lang E, Foller M. Oxidative stress and suicidal erythrocyte death. Antioxidants & Redox Signaling. 2013;21:138-153
  10. 10. Lang F, Qadri SM. Mechanisms and significance of eryptosis, the suicidal death of erythrocytes. Blood Purification. 2012;33:125-130
  11. 11. Kushner JP, Porter JP, Olivieri NF. Secondary iron overload. Hematology-American Society of Hematology Education Program. 2001;2001(1):47-61
  12. 12. Weatherall DJ. Pathophysiology of thalassaemia. Baillière's Clinical Haematology. 1998;11:127-146
  13. 13. Weatherall DJ, Pressley L, Wood WG, Higgs DR, Clegg JB. Molecular basis for mild forms of homozygous beta-thalassaemia. Lancet. 1981;1:527-529
  14. 14. Hershko C, Konijn AM, Link G. Iron chelators for thalassaemia. British Journal of Haematology. 1998;101:399-406
  15. 15. Breuer W, Hershko C, Cabantchik ZI. The importance of non-transferrin bound iron in disorders of iron metabolism. Transfusion Science. 2000;23:185-192
  16. 16. Esposito BP, Breuer W, Sirankapracha P, Pootrakul P, Hershko C, Cabantchik ZI. Labile plasma iron in iron overload: Redox activity and susceptibility to chelation. Blood. 2003;102:2670-2677
  17. 17. Jomova K, Valko M. Importance of iron chelation in free radical-induced oxidative stress and human disease. Current Pharmaceutical Design. 2011;17:3460-3473
  18. 18. Cairo G, Recalcati S, Pietrangelo A, Minotti G. The iron regulatory proteins: Targets and modulators of free radical reactions and oxidative damage. Free Radical Biology & Medicine. 2002;32:1237-1243
  19. 19. Yurkova I, Kisel M, Arnhold J, Shadyro O. Iron-mediated free-radical formation of signaling lipids in a model system. Chemistry and Physics of Lipids. 2005;137:29-37
  20. 20. Hebbel RP. Auto-oxidation and a membrane-associated 'Fenton reagent': A possible explanation for development of membrane lesions in sickle erythrocytes. Clinics in Haematology. 1985;14:129-140
  21. 21. Hershko CM, Link GM, Konijn AM, Cabantchik ZI. Iron chelation therapy. Current Hematology Reports. 2005;4:110-116
  22. 22. Schafer FQ, Qian SY, Buettner GR. Iron and free radical oxidations in cell membranes. Cellular and Molecular Biology (Noisy-le-Grand, France). 2000;46:657-662
  23. 23. Okon E, Levij IS, Rachmilewitz EA. Splenectomy, iron overload and liver cirrhosis in beta-thalassemia major. Acta Haematologica. 1976;56:142-150
  24. 24. Matsuno T, Mori M, Awai M. Distribution of ferritin and hemosiderin in the liver, spleen and bone marrow of normal, phlebotomized and iron overloaded rats. Acta Medica Okayama. 1985;39:347-360
  25. 25. Jensen PD. Evaluation of iron overload. British Journal of Haematology. 2004;124:697-711
  26. 26. Andrews NC. Disorders of iron metabolism. The New England Journal of Medicine. 1999;341:1986-1995
  27. 27. Deugnier Y, Turlin B. Pathology of hepatic iron overload. Seminars in Liver Disease. 2011;31:260-271
  28. 28. Freeman AP, Giles RW, Berdoukas VA, Talley PA, Murray IP. Sustained normalization of cardiac function by chelation therapy in thalassaemia major. Clinical and Laboratory Haematology. 1989;11:299-307
  29. 29. Wolfe L, Olivieri N, Sallan D, Colan S, Rose V, Propper R, et al. Prevention of cardiac disease by subcutaneous deferoxamine in patients with thalassemia major. The New England Journal of Medicine. 1985;312:1600-1603
  30. 30. Brink B, Disler P, Lynch S, Jacobs P, Charlton R, Bothwell T. Patterns of iron storage in dietary iron overload and idiopathic hemochromatosis. The Journal of Laboratory and Clinical Medicine. 1976;88:725-731
  31. 31. Zervas A, Katopodi A, Protonotariou A, Livadas S, Karagiorga M, Politis C, et al. Assessment of thyroid function in two hundred patients with beta-thalassemia major. Thyroid. 2002;12:151-154
  32. 32. Soliman AT, elZalabany MM, Mazloum Y, Bedair SM, Ragab MS, Rogol AD, et al. Spontaneous and provoked growth hormone (GH) secretion and insulin-like growth factor I (IGF-I) concentration in patients with beta thalassaemia and delayed growth. Journal of Tropical Pediatrics. 1999;45:327-337
  33. 33. Tato L, Lahlou N, Zamboni G, De Sanctis V, De Luca F, Arrigo T, et al. Impaired response of free alpha-subunits after luteinizing hormone-releasing hormone and thyrotropin-releasing hormone stimulations in beta-thalassemia major. Hormone Research. 1993;39:213-217
  34. 34. Wood JC, Noetzl L, Hyderi A, Joukar M, Coates T, Mittelman S. Predicting pituitary iron and endocrine dysfunction. Annals of the New York Academy of Sciences. 2010;1202:123-128
  35. 35. Nakavachara P, Viprakasit V. Adrenal insufficiency is prevalent in HbE/beta-thalassaemia paediatric patients irrespective of their clinical severity and transfusion requirement. Clinical Endocrinology. 2013;79:776-783
  36. 36. Koonyosying P, Uthaipibull C, Fucharoen S, Koumoutsea EV, Porter JB, Srichairatanakool S. Decrement in cellular iron and reactive oxygen species, and improvement of insulin secretion in a pancreatic cell line using green tea extract. Pancreas. 2019;48(5):636-643
  37. 37. Vlachodimitropoulou E, Chen YL, Garbowski M, Koonyosying P, Psaila B, Sola-Visner M, et al. Eltrombopag: A powerful chelator of cellular or extracellular iron(III) alone or combined with a second chelator. Blood. 2017;130:1923-1933
  38. 38. Letsky EA, Miller F, Worwood M, Flynn DM. Serum ferritin in children with thalassaemia regularly transfused. Journal of Clinical Pathology. 1974;27:652-655
  39. 39. Butensky E, Fischer R, Hudes M, Schumacher L, Williams R, Moyer TP, et al. Variability in hepatic iron concentration in percutaneous needle biopsy specimens from patients with transfusional hemosiderosis. American Journal of Clinical Pathology. 2005;123:146-152
  40. 40. Mavrogeni SI, Markussis V, Kaklamanis L, Tsiapras D, Paraskevaidis I, Karavolias G, et al. A comparison of magnetic resonance imaging and cardiac biopsy in the evaluation of heart iron overload in patients with beta-thalassemia major. European Journal of Haematology. 2005;75:241-247
  41. 41. Youssry I, Mohsen NA, Shaker OG, El-Hennawy A, Fawzy R, Abu-Zeid NM, et al. Skin iron concentration: A simple, highly sensitive method for iron stores evaluation in thalassemia patients. Hemoglobin. 2007;31:357-365
  42. 42. Cohen AR, Galanello R, Pennell DJ, Cunningham MJ, Vichinsky E. Thalassemia. Hematology-American Society of Hematology Education Program. 2004;2004(1):14-34
  43. 43. Sirachainan N. Thalassemia and the hypercoagulable state. Thrombosis Research. 2013;132:637-641
  44. 44. Chuansumrit A, Hathirat P, Isarangkura P, Pintadit P, Mahaphan W. Thrombotic risk of children with thalassemia. Journal of the Medical Association of Thailand. 1993;76(Suppl 2):80-84
  45. 45. Bhattacharyya M, Kannan M, Chaudhry VP, Mahapatra M, Pati H, Saxena R. Hypercoagulable state in five thalassemia intermedia patients. Clinical and Applied Thrombosis/Hemostasis. 2007;13:422-427
  46. 46. Al-Hawsawi ZM, Haouimi AS, Hassan RA, Tarawah AM. Portal vein thrombosis after splenectomy for beta-thalassemia major. Saudi Medical Journal. 2004;25:225-228
  47. 47. Tyan PI, Radwan AH, Eid A, Haddad AG, Wehbe D, Taher AT. Novel approach to reactive oxygen species in nontransfusion-dependent thalassemia. BioMed Research International. 2014;2014:350432
  48. 48. Butthep P, Bunyaratvej A, Funahara Y, Kitaguchi H, Fucharoen S, Sato S, et al. Possible evidence of endothelial cell activation and disturbance in thalassemia: An in vitro study. The Southeast Asian Journal of Tropical Medicine and Public Health. 1997;28(Suppl 3):141-148A
  49. 49. Cappellini MD, Grespi E, Cassinerio E, Bignamini D, Fiorelli G. Coagulation and splenectomy: An overview. Annals of the New York Academy of Sciences. 2005;1054:317-324
  50. 50. Pattanapanyasat K, Gonwong S, Chaichompoo P, Noulsri E, Lerdwana S, Sukapirom K, et al. Activated platelet-derived microparticles in thalassaemia. British Journal of Haematology. 2007;136:462-471
  51. 51. Yanpanitch OU, Hatairaktham S, Charoensakdi R, Panichkul N, Fucharoen S, Srichairatanakool S, et al. Treatment of beta-thalassemia/hemoglobin E with antioxidant cocktails results in decreased oxidative stress, increased hemoglobin concentration, and improvement of the hypercoagulable state. Oxidative Medicine and Cellular Longevity. 2015;2015:537954
  52. 52. Inthawong K, Charoenkwan P, Silvilairat S, Tantiworawit A, Phrommintikul A, Choeyprasert W, et al. Pulmonary hypertension in non-transfusion-dependent thalassemia: Correlation with clinical parameters, liver iron concentration, and non-transferrin-bound iron. Hematology. 2015;20:610-617
  53. 53. Yatmark P, Morales NP, Chaisri U, Wichaiyo S, Hemstapat W, Srichairatanakool S, et al. Effects of iron chelators on pulmonary iron overload and oxidative stress in beta-thalassemic mice. Pharmacology. 2015;96:192-199
  54. 54. Nielsen VG, Pretorius E. Iron and carbon monoxide enhance coagulation and attenuate fibrinolysis by different mechanisms. Blood Coagulation & Fibrinolysis. 2014;25:695-702
  55. 55. Morris CR, Kim HY, Klings ES, Wood J, Porter JB, Trachtenberg F, et al. Dysregulated arginine metabolism and cardiopulmonary dysfunction in patients with thalassaemia. British Journal of Haematology. 2015;169:887-898
  56. 56. Cappellini MD, Musallam KM, Poggiali E, Taher AT. Hypercoagulability in non-transfusion-dependent thalassemia. Blood Reviews. 2012;26(Suppl 1):S20-S23
  57. 57. Musallam KM, Taher AT, Karimi M, Rachmilewitz EA. Cerebral infarction in beta-thalassemia intermedia: Breaking the silence. Thrombosis Research. 2012;130:695-702
  58. 58. Gonzalez-Reimers E, Quintero-Platt G, Martin-Gonzalez C, Perez-Hernandez O, Romero-Acevedo L, Santolaria-Fernandez F. Thrombin activation and liver inflammation in advanced hepatitis C virus infection. World Journal of Gastroenterology. 2016;22:4427-4437
  59. 59. Kell DB, Pretorius E. Serum ferritin is an important inflammatory disease marker, as it is mainly a leakage product from damaged cells. Metallomics. 2014;6:748-773
  60. 60. Fucharoen S, Weatherall DJ. Progress toward the control and management of the thalassemias. Hematology/Oncology Clinics of North America. 2016;30:359-371
  61. 61. Porter JB. Optimizing iron chelation strategies in beta-thalassaemia major. Blood Reviews. 2009;23(Suppl 1):S3-S7
  62. 62. Farmaki K, Tzoumari I, Pappa C. Oral chelators in transfusion-dependent thalassemia major patients may prevent or reverse iron overload complications. Blood Cells, Molecules & Diseases. 2011;47:33-40
  63. 63. Vannasaeng S, Fucharoen S, Pootrakul P, Ploybutr S, Yansukon P. Pituitary function in thalassemic patients and the effect of chelation therapy. Acta Endocrinologica. 1991;124:23-30
  64. 64. Halliwell B. Drug antioxidant effects. A basis for drug selection? Drugs. 1991;42:569-605
  65. 65. Amer J, Atlas D, Fibach E. N-acetylcysteine amide (AD4) attenuates oxidative stress in beta-thalassemia blood cells. Biochimica et Biophysica Acta. 2008;1780:249-255
  66. 66. Dissayabutra T, Tosukhowong P, Seksan P. The benefits of vitamin C and vitamin E in children with beta-thalassemia with high oxidative stress. Journal of the Medical Association of Thailand. 2005;88(Suppl 4):S317-S321
  67. 67. Giardini O, Cantani A, Donfrancesco A. Vitamin E therapy in homozygous beta-thalassemia. The New England Journal of Medicine. 1981;305:644
  68. 68. Giardini O, Cantani A, Donfrancesco A, Martino F, Mannarino O, D'Eufemia P, et al. Biochemical and clinical effects of vitamin E administration in homozygous beta-thalassemia. Acta Vitaminologica et Enzymologica. 1985;7:55-60
  69. 69. Hyman CB, Landing B, Alfin-Slater R, Kozak L, Weitzman J, Ortega JA. Dl-alpha-tocopherol, iron, and lipofuscin in thalassemia. Annals of the New York Academy of Sciences. 1974;232:211-220
  70. 70. Kalpravidh RW, Wichit A, Siritanaratkul N, Fucharoen S. Effect of coenzyme Q10 as an antioxidant in beta-thalassemia/Hb E patients. BioFactors. 2005;25:225-234
  71. 71. Miniero R, Canducci E, Ghigo D, Saracco P, Vullo C. Vitamin E in beta-thalassemia. Acta Vitaminologica et Enzymologica. 1982;4:21-25
  72. 72. Miniero R, David O, Ghigo D, Luzzatto L, Ramenghi U, Saracco P, et al. Administration of vitamin E in heterozygous beta-thalassaemia: The effect on red blood cell survival. Panminerva Medica. 1984;26:283-286
  73. 73. Miniero R, Piga A, Luzzatto L, Gabutti V. Vitamin E and beta-thalassaemia. Haemato-logica. 1983;68:562-563
  74. 74. O'Brien RT. Ascorbic acid enhancement of desferrioxamine-induced urinary iron excretion in thalassemia major. Annals of the New York Academy of Sciences. 1974;232:221-225
  75. 75. Ozsoylu S, Gurgey A. Vitamin E treatment in triplicated alpha-globin gene-heterozygous beta-thalassemia. American Journal of Hematology. 1991;38:335-336
  76. 76. Palasuwan A, Soogarun S, Wiwanitkit V, Luechapudiporn R, Pradniwat P, Lertlum T. Preliminary study of the effect of vitamin E supplementation on the antioxidant status of hemoglobin-E carriers. The Southeast Asian Journal of Tropical Medicine and Public Health. 2006;37(Suppl 3):184-189
  77. 77. Pfeifer WP, Degasperi GR, Almeida MT, Vercesi AE, Costa FF, Saad ST. Vitamin E supplementation reduces oxidative stress in beta thalassaemia intermedia. Acta Haematologica. 2008;120:225-231
  78. 78. Tesoriere L, D'Arpa D, Butera D, Allegra M, Renda D, Maggio A, et al. Oral supplements of vitamin E improve measures of oxidative stress in plasma and reduce oxidative damage to LDL and erythrocytes in beta-thalassemia intermedia patients. Free Radical Research. 2001;34:529-540
  79. 79. Vatanavicharn S, Yenchitsomanus P, Siddhikol C. Vitamin E in beta-thalassaemia and alpha-thalassaemia (HbH) diseases. Acta Haematologica. 1985;73:183
  80. 80. Unchern S, Laoharuangpanya N, Phumala N, Sipankapracha P, Pootrakul P, Fucharoen S, et al. The effects of vitamin E on platelet activity in beta-thalassaemia patients. British Journal of Haematology. 2003;123:738-744
  81. 81. Alidoost F, Gharagozloo M, Bagherpour B, Jafarian A, Sajjadi SE, Hourfar H, et al. Effects of silymarin on the proliferation and glutathione levels of peripheral blood mononuclear cells from beta-thalassemia major patients. International Immuno-pharmacology. 2006;6:1305-1310
  82. 82. Amer J, Goldfarb A, Rachmilewitz EA, Fibach E. Fermented papaya preparation as redox regulator in blood cells of beta-thalassemic mice and patients. Phytotherapy Research. 2008;22:820-828
  83. 83. Kalpravidh RW, Siritanaratkul N, Insain P, Charoensakdi R, Panichkul N, Hatairaktham S, et al. Improvement in oxidative stress and antioxidant parameters in beta-thalassemia/Hb E patients treated with curcuminoids. Clinical Biochemistry. 2009;43:424-429
  84. 84. Fibach E, Rachmilewitz EA. The role of antioxidants and iron chelators in the treatment of oxidative stress in thalassemia. Annals of the New York Academy of Sciences. 2010;1202:10-16
  85. 85. Wapnick AA, Lynch SR, Krawitz P, Seftel HC, Charlton RW, Bothwell TH. Effects of iron overload on ascorbic acid metabolism. British Medical Journal. 1968;3:704-707
  86. 86. Chatterjea B, Maitra A, Banerjee DK, Basu AK. Status of ascorbic acid in iron deficiency anaemia and thalassaemia. Acta Haematologica. 1980;64:271-275
  87. 87. Chapman RW, Hussain MA, Gorman A, Laulicht M, Politis D, Flynn DM, et al. Effect of ascorbic acid deficiency on serum ferritin concentration in patients with beta-thalassaemia major and iron overload. Journal of Clinical Pathology. 1982;35:487-491
  88. 88. Livrea MA, Tesoriere L, Pintaudi AM, Calabrese A, Maggio A, Freisleben HJ, et al. Oxidative stress and antioxidant status in beta-thalassemia major: Iron overload and depletion of lipid-soluble antioxidants. Blood. 1996;88:3608-3614
  89. 89. Vatanavicharn S, Anuwatanakulchai M, Yenchitsomanus P, Siddhikol C. Relationship of serum vitamin E, erythrocyte nonheme iron, and malonyldialdehyde (lipid membrane peroxidation product) in thalassemia. Birth Defects Original Article Series. 1987;23:207-211
  90. 90. Selden C, Seymour CA, Peters TJ. Activities of some free-radical scavenging enzymes and glutathione concentrations in human and rat liver and their relationship to the pathogenesis of tissue damage in iron overload. Clinical Science (London, England). 1980;58:211-219
  91. 91. Amer J, Fibach E. Oxidative status of platelets in normal and thalassemic blood. Thrombosis and Haemostasis. 2004;92:1052-1059
  92. 92. Aphinives C, Kukongviriyapan U, Jetsrisuparb A, Kukongviriyapan V, Somparn N. Impaired endothelial function in pediatric hemoglobin E/beta-thalassemia patients with iron overload. The Southeast Asian Journal of Tropical Medicine and Public Health. 2015;45:1454-1463
  93. 93. Chakraborty D, Bhattacharyya M. Antioxidant defense status of red blood cells of patients with beta-thalassemia and E/beta-thalassemia. Clinica Chimica Acta. 2001;305:123-129
  94. 94. Cheng ML, Ho HY, Tseng HC, Lee CH, Shih LY, Chiu DT. Antioxidant deficit and enhanced susceptibility to oxidative damage in individuals with different forms of alpha-thalassaemia. British Journal of Haematology. 2005;128:119-127
  95. 95. Chiou SS, Tsao CJ, Tsai SM, Wu YR, Liao YM, Lin PC, et al. Metabolic pathways related to oxidative stress in patients with hemoglobin H disease and iron overload. Journal of Clinical Laboratory Analysis. 2014;28:261-268
  96. 96. Boonyawat K, Wongwaisayawan S, Nitiyanant P, Atichartakarn V. Hydroxyurea and colonic ulcers: A case report. BMC Gastroenterology. 2014;14:134
  97. 97. Bradai M, Abad MT, Pissard S, Lamraoui F, Skopinski L, de Montalembert M. Hydro-xyurea can eliminate transfusion requirements in children with severe beta-thalassemia. Blood. 2003;102:1529-1530
  98. 98. Bradai M, Pissard S, Abad MT, Dechartres A, Ribeil JA, Landais P, et al. Decreased transfusion needs associated with hydroxyurea therapy in Algerian patients with thalassemia major or intermedia. Transfusion. 2007;47:1830-1836
  99. 99. Darbari DS, Nouraie M, Taylor JG, Brugnara C, Castro O, Ballas SK. Alpha-thalassaemia and response to hydroxyurea in sickle cell anaemia. European Journal of Haematology. 2013;92:341-345
  100. 100. Italia K, Chandrakala S, Ghosh K, Colah R. Can hydroxyurea serve as a free radical scavenger and reduce iron overload in beta-thalassemia patients? Free Radical Research. 2016;50:959-965
  101. 101. Ajanta H, Chakraborty S, Madhusnata D, Bhattacharya DK, Manisha D. The activity of superoxide dismutase in hydroxyurea-treated E beta thalassemia. The Journal of the Association of Physicians of India. 2002;50:1034-1035
  102. 102. De M, Banerjee N, Talukdar G, Bhattacharya DK. Lipid peroxidation and spectrin of RBC membrane in hydroxyurea treated E/beta thalassaemia. Indian Journal of Clinical Biochemistry. 2000;15:56-59
  103. 103. Udupi V, Rice-Evans C. Thiol compounds as protective agents in erythrocytes under oxidative stress. Free Radical Research Communications. 1992;16:315-323
  104. 104. Nur E, Brandjes DP, Teerlink T, Otten HM, Oude Elferink RP, Muskiet F, et al. N-acetylcysteine reduces oxidative stress in sickle cell patients. Annals of Hematology. 2012;91:1097-1105
  105. 105. Ozdemir ZC, Koc A, Aycicek A, Kocyigit A. N-Acetylcysteine supplementation reduces oxidative stress and DNA damage in children with beta-thalassemia. Hemoglobin. 2014;38:359-364
  106. 106. Gupta SC, Patchva S, Aggarwal BB. Therapeutic roles of curcumin: Lessons learned from clinical trials. The AAPS Journal. 2012;15:195-218
  107. 107. Iqbal M, Okazaki Y, Okada S. In vitro curcumin modulates ferric nitrilotriacetate (Fe-NTA) and hydrogen peroxide (H2O2)-induced peroxidation of microsomal membrane lipids and DNA damage. Teratogenesis Carcinogenesis and Mutagenesis. 2003;Suppl 1:151-160
  108. 108. Sharma OP. Antioxidant activity of curcumin and related compounds. Biochemical Pharmacology. 1976;25:1811-1812
  109. 109. Srichairatanakool S, Thephinlap C, Phisalaphong C, Porter JB, Fucharoen S. Curcumin contributes to in vitro removal of non-transferrin bound iron by deferiprone and desferrioxamine in thalassemic plasma. Medicinal Chemistry. 2007;3:469-474
  110. 110. Thephinlap C, Phisalaphong C, Fucharoen S, Porter JB, Srichairatanakool S. Efficacy of curcuminoids in alleviation of iron overload and lipid peroxidation in thalassemic mice. Medicinal Chemistry. 2009;5:474-482
  111. 111. Thephinlap C, Phisalaphong C, Lailerd N, Chattipakorn N, Winichagoon P, Vadolas J, et al. Reversal of cardiac iron loading and dysfunction in thalassemic mice by curcuminoids. Medicinal Chemistry. 2011;7:62-69
  112. 112. Chaneiam N, Changtam C, Mungkongdee T, Suthatvoravut U, Winichagoon P, Vadolas J, et al. A reduced curcuminoid analog as a novel inducer of fetal hemoglobin. Annals of Hematology. 2012;92:379-386
  113. 113. Badria FA, Ibrahim AS, Badria AF, Elmarakby AA. Curcumin attenuates iron accumulation and oxidative stress in the liver and spleen of chronic iron-overloaded rats. PLoS ONE. 2015;10:e0134156
  114. 114. McKay DL, Blumberg JB. The role of tea in human health: An update. Journal of the American College of Nutrition. 2002;21:1-13
  115. 115. Cabrera C, Artacho R, Gimenez R. Beneficial effects of green tea—A review. Journal of the American College of Nutrition. 2006;25:79-99
  116. 116. Srichairatanakool S, Ounjaijean S, Thephinlap C, Khansuwan U, Phisalpong C, Fucharoen S. Iron-chelating and free-radical scavenging activities of microwave-processed green tea in iron overload. Hemoglobin. 2006;30:311-327
  117. 117. Thephinlap C, Ounjaijean S, Khansuwan U, Fucharoen S, Porter JB, Srichairatanakool S. Epigallocatechin-3-gallate and epicatechin-3-gallate from green tea decrease plasma non-transferrin bound iron and erythrocyte oxidative stress. Medicinal Chemistry. 2007;3:289-296
  118. 118. Ounjaijean S, Thephinlap C, Khansuwan U, Phisalapong C, Fucharoen S, Porter JB, et al. Effect of green tea on iron status and oxidative stress in iron-loaded rats. Medicinal Chemistry. 2008;4:365-370
  119. 119. Saewong T, Ounjaijean S, Mundee Y, Pattanapanyasat K, Fucharoen S, Porter JB, et al. Effects of green tea on iron accumulation and oxidative stress in livers of iron-challenged thalassemic mice. Medicinal Chemistry. 2010;6:57-64
  120. 120. de Alarcon PA, Donovan ME, Forbes GB, Landaw SA, Stockman JA 3rd. Iron absorption in the thalassemia syndromes and its inhibition by tea. The New England Journal of Medicine. 1979;300:5-8
  121. 121. Lim DY. Comparison of green tea extract and epigallocatechin gallate on secretion of catecholamines from the rabbit adrenal medulla. Archives of Pharmacal Research. 2005;28:914-922
  122. 122. Liatsos GD, Moulakakis A, Ketikoglou I, Klonari S. Possible green tea-induced thrombotic thrombocytopenic purpura. American Journal of Health-System Pharmacy. 2010;67:531-534
  123. 123. Kang WS, Lim IH, Yuk DY, Chung KH, Park JB, Yoo HS, et al. Antithrombotic activities of green tea catechins and (−)-epigallocatechin gallate. Thrombosis Research. 1999;96:229-237
  124. 124. Lee DH, Kim YJ, Kim HH, Cho HJ, Ryu JH, Rhee MH, et al. Inhibitory effects of epigallocatechin-3-gallate on microsomal cyclooxygenase-1 activity in platelets. Biomolecules & Therapeutics. 2013;21:54-59
  125. 125. Koonyosying P, Tantiworawit A, Hantrakool S, Utama-Ang, N, Cresswell M, Fucharoen S, et al. Consumption of a green tea extract-curcumin drink decreases blood urea nitrogen and redox iron in β-thalassemia patients. Food & Functions. 2020;11(1):932-943
  126. 126. Devasagayam TP, Kamat JP, Mohan H, Kesavan PC. Caffeine as an antioxidant: Inhibition of lipid peroxidation induced by reactive oxygen species. Biochimica et Biophysica Acta. 1996;1282:63-70
  127. 127. Shi X, Dalal NS, Jain AC. Antioxidant behaviour of caffeine: Efficient scavenging of hydroxyl radicals. Food and Chemical Toxicology. 1991;29:1-6
  128. 128. Stadler RH, Turesky RJ, Muller O, Markovic J, Leong-Morgenthaler PM. The inhibitory effects of coffee on radical-mediated oxidation and mutagenicity. Mutation Research. 1994;308:177-190
  129. 129. Chu YF, Farah A. Coffee constituents. In: Coffee. New Jersey, USA: John Wiley & Sons Inc. and the Institute of Food Technologists; 2012. pp. 22-58 (Chapter 2)
  130. 130. Monente C, Ludwig IA, Irigoyen A, De Pena MP, Cid C. Assessment of total (free and bound) phenolic compounds in spent coffee extracts. Journal of Agricultural and Food Chemistry. 2015;63:4327-4334
  131. 131. Monente C, Ludwig IA, Stalmach A, de Pena MP, Cid C, Crozier A. In vitro studies on the stability in the proximal gastrointestinal tract and bioaccessibility in Caco-2 cells of chlorogenic acids from spent coffee grounds. International Journal of Food Sciences and Nutrition. 2015;66:657-664
  132. 132. Takenaka M, Sato N, Asakawa H, Wen X, Murata M, Homma S. Characterization of a metal-chelating substance in coffee. Bioscience, Biotechnology, and Biochemistry. 2005;69:26-30
  133. 133. Umemura T, Ueda K, Nishioka K, Hidaka T, Takemoto H, Nakamura S, et al. Effects of acute administration of caffeine on vascular function. The American Journal of Cardiology. 2006;98:1538-1541
  134. 134. Olthof MR, Hollman PC, Katan MB. Chlorogenic acid and caffeic acid are absorbed in humans. The Journal of Nutrition. 2001;131:66-71
  135. 135. Naito S, Yatagai C, Maruyama M, Sumi H. Effect of coffee extracts on plasma fibrinolysis and platelet aggregation. Nihon Arukōru Yakubutsu Igakkai Zasshi. 2011;46:260-269
  136. 136. Jiang R, Hodgson JM, Mas E, Croft KD, Ward NC. Chlorogenic acid improves ex vivo vessel function and protects endothelial cells against HOCl-induced oxidative damage, via increased production of nitric oxide and induction of Hmox-1. The Journal of Nutritional Biochemistry. 2015;27:53-60
  137. 137. Echeverri D, Montes FR, Cabrera M, Galan A, Prieto A. Caffeine's vascular mechanisms of action. International Journal of Vascular Medicine. 2010;2010:834060
  138. 138. Folstar P, van der Plas HC, Pilnik W, de Heus JG. Tocopherols in the unsaponifiable matter of coffee bean oil. Journal of Agricultural and Food Chemistry. 1977;25:283-285
  139. 139. Sved S, Hossie RD, McGilveray IJ. The human metabolism of caffeine to theophylline. Research Communications in Chemical Pathology and Pharmacology. 1976;13:185-192
  140. 140. Morris ER. An overview of current information on bioavailability of dietary iron to humans. Federation Proceedings. 1983;42:1716-1720
  141. 141. Huang J, de Paulis T, May JM. Antioxidant effects of dihydrocaffeic acid in human EA.hy926 endothelial cells. The Journal of Nutritional Biochemistry. 2004;15:722-729
  142. 142. Ochiai R, Jokura H, Suzuki A, Tokimitsu I, Ohishi M, Komai N, et al. Green coffee bean extract improves human vasoreactivity. Hypertension Research. 2004;27:731-737
  143. 143. Lopez-Barrera DM, Vazquez-Sanchez K, Loarca-Pina MG, Campos-Vega R. Spent coffee grounds, an innovative source of colonic fermentable compounds, inhibit inflammatory mediators in vitro. Food Chemistry. 2016;212:282-290
  144. 144. Agudelo-Ochoa GM, Pulgarin-Zapata IC, Velasquez-Rodriguez CM, Duque-Ramirez M, Naranjo-Cano M, Quintero-Ortiz MM, et al. Coffee consumption increases the antioxidant capacity of plasma and has no effect on the lipid profile or vascular function in healthy adults in a randomized controlled trial. The Journal of Nutrition. 2016;146:524-531
  145. 145. Chang J, Fedinec AL, Kuntamallappanavar G, Leffler CW, Bukiya AN, Dopico AM. Endothelial nitric oxide mediates caffeine antagonism of alcohol-induced cerebral artery constriction. The Journal of Pharmacology and Experimental Therapeutics. 2015;356:106-115
  146. 146. Shin HS, Satsu H, Bae MJ, Totsuka M, Shimizu M. Catechol groups enable reactive oxygen species scavenging-mediated suppression of PKD-NF kappaB-IL-8 signaling pathway by chlorogenic and caffeic acids in human intestinal cells. Nutrients. 2017;9:E165. DOI: 10.3390/nu9020165
  147. 147. Bakuradze T, Boehm N, Janzowski C, Lang R, Hofmann T, Stockis JP, et al. Antioxidant-rich coffee reduces DNA damage, elevates glutathione status and contributes to weight control: Results from an intervention study. Molecular Nutrition & Food Research. 2011;55:793-797
  148. 148. Hoelzl C, Knasmuller S, Wagner KH, Elbling L, Huber W, Kager N, et al. Instant coffee with high chlorogenic acid levels protects humans against oxidative damage of macromolecules. Molecular Nutrition & Food Research. 2010;54:1722-1733
  149. 149. Guerrini A, Lampronti I, Bianchi N, Zuccato C, Breveglieri G, Salvatori F, et al. Bergamot (Citrus bergamia Risso) fruit extracts as gamma-globin gene expression inducers: Phytochemical and functional perspectives. Journal of Agricultural and Food Chemistry. 2009;57:4103-4111
  150. 150. Fibach E, Tan ES, Jamuar S, Ng I, Amer J, Rachmilewitz EA. Amelioration of oxidative stress in red blood cells from patients with beta-thalassemia major and intermedia and E-beta-thalassemia following administration of a fermented papaya preparation. Phytotherapy Research. 2010;24:1334-1338
  151. 151. Pardo-Andreu GL, Sanchez-Baldoquin C, Avila-Gonzalez R, Yamamoto ET, Revilla A, Uyemura SA, et al. Interaction of Vimang (Mangifera indica L. extract) with Fe(III) improves its antioxidant and cytoprotecting activity. Pharmacological Research. 2006;54:389-395
  152. 152. Srichairatanakool S et al. Nutritional values and pharmacological activities of Mangifera indica Linn. cv Mahachanok. In: A Final Report of Thailand Research Fund. 2013
  153. 153. Marawaha RK, Bansal D, Kaur S, Trehan A. Wheat grass juice reduces transfusion requirement in patients with thalassemia major: A pilot study. Indian Pediatrics. 2004;41:716-720
  154. 154. Singh K, Pannu MS, Singh P, Singh J. Effect of wheat grass tablets on the frequency of blood transfusions in thalassemia major. Indian Journal of Pediatrics. 2010;77:90-91
  155. 155. Choudhary DR, Naithani R, Panigrahi I, Kumar R, Mahapatra M, Pati HP, et al. Effect of wheat grass therapy on transfusion requirement in beta-thalassemia major. Indian Journal of Pediatrics. 2009;76:375-376
  156. 156. Bagwe SM, Kale PP, Bhatt LK, Prabhavalkar KS. Herbal approach in the treatment of pancytopenia. Journal of Complementary and Integrative Medicine. 2017;14(1):1-11
  157. 157. Pangjit K, Tantiphaipunwong P, Sajjapong W, Srichairatanakool S. Iron-chelating, free radical scavenging and anti-proliferative activities of Azadirachta indica. Journal of the Medical Association of Thailand. 2014;97(Suppl 4):S36-S43

Written By

Somdet Srichairatanakool, Pimpisid Koonyosying and Suthat Fucharoen

Reviewed: 05 March 2020 Published: 14 October 2020