Open access peer-reviewed chapter

Overview of Beta-Thalassemia

Written By

Kenneth Oshiokhayamhe Iyevhobu, Omolumen Lucky E., Tobechukwu Joseph Okobi, Edidiong Raphael Usoro, A. Airefetalor Ivie, Benedicta A. Ken-Iyevhobu and O. Omokpo Victoria

Submitted: 20 October 2022 Reviewed: 25 April 2023 Published: 22 November 2023

DOI: 10.5772/intechopen.111682

From the Edited Volume

Thalassemia Syndromes - New Insights and Transfusion Modalities

Edited by Marwa Zakaria, Tamer Hassan, Laila Sherief and Osaro Erhabor

Chapter metrics overview

98 Chapter Downloads

View Full Metrics

Abstract

Beta-thalassemias are a group of hereditary blood disorders characterized by anomalies in the synthesis of the beta chains of hemoglobin resulting in variable phenotypes ranging from severe anemia to clinically asymptomatic individuals. Three main forms have been described: thalassemia major, thalassemia intermedia, and thalassemia minor. Individuals with thalassemia major usually present within the first 2 years of life with severe anemia, requiring regular red blood cell (RBC) transfusions. Patients with thalassemia intermedia present later in life with moderate anemia and do not require regular transfusions. Thalassemia minor is clinically asymptomatic, but some subjects may have moderate anemia. Beta-thalassemias are caused by point mutations or, more rarely, deletions in the beta-globin gene on chromosome 11, leading to reduced (beta+) or absent (beta0) synthesis of the beta chains of hemoglobin (Hb). Transmission is autosomal recessive; however, dominant mutations have also been reported. Diagnosis of thalassemia is based on hematologic and molecular genetic testing. Laboratory tests that are conventionally performed to diagnose the β-thalassemia and HbE are classified into two groups, based on the purposes, including the screening tests and confirmatory tests.

Keywords

  • beta
  • thalassemia
  • hemoglobin
  • anemia
  • globin

1. Introduction

The term thalassemia is deduced from the Greek, namely thalassa (ocean) and haima (blood). Thalassemia is among the most common heritable diseases in the world [1, 2, 3]. The thalassemias are conditions caused by dropped expression of one of the two globin chains of the hemoglobin patch, namely α (HBA) and β (HBB). Inherited through an autosomal sheepish pathway, point mutations and elisions on the genes that decode for the globin chains beget dropped hemoglobin (Hb) product, leading to severe anemia [4]. Thalassemia cases depend on lifelong medical care, entering routine blood transfusions and supplemental curatives [5]. Thus, timely opinion and forestallment is essential, especially in regions with high frequency of this complaint [6, 7, 8]. Beta-thalassemia characterized by reduced or absent β-globin chain conflation is one of the most common inherited blood diseases in the world and hence a major interference to public health. Beta-thalassemia is a blood complaint that reduces the product of hemoglobin [9]. The protein in red blood cells called hemoglobin, which contains iron, transports oxygen to every cell in the body. Low hemoglobin levels in beta-thalassemia patients cause an oxygen shortage in a number of bodily passageways [10]. A lack of red blood cells in affected people (anemia) can result in pale skin, weakness, weariness, and more severe problems. Beta-thalassemia patients are more likely to get irregular blood clots [11]. Beta-thalassemia runs are a group of heritable blood diseases characterized by reduced or absent beta-globin chain conflation, performing in reduced Hb in red blood cells (RBC), dropped RBC product and anemia. Beta-thalassemia major was first described in the medical literature in 1925 by an American croaker-Thomas Cooley. Beta-thalassemia includes three main forms, namely thalassemia major, perfectly appertained to as “Cooley’s Anemia” and “Mediterranean Anemia”, thalassemia intermedia, and thalassemia minor also called “beta-thalassemia carrier”, “beta-thalassemia particularity”, or “heterozygous beta-thalassemia” [10, 11]. Piecemeal from the rare dominant forms, subjects with thalassemia major are homozygotes or emulsion heterozygotes for beta0 or beta genes, subjects with thalassemia intermedia are substantially homozygotes or emulsion heterozygotes, and subjects with thalassemia minor are substantially heterozygotes. Although wide, the major at- threat populations are substantially from Mediterranean, Middle East, and Southeast Asian countries [12, 13]. The thalassemia category of hemoglobin conflation illnesses differs from the others in that there is either little or no-globin chain conflation. The main forms of β-thalassemia are two. The two types of thalassemia are βO-thalassemia, in which no-globin chain is made, and -thalassemia, in which some globin is produced but at a lower level than usual [14]. Microcytosis, often known as thalassemia minor clinically, affects heterozygotes for either type of allele. A more severe phenotype known as β-thalassemia intermedia, which involves anemia, hemolysis, iron loading, and the sporadic need for transfusion, is present in homozygous heterozygotes for two thalassemia alleles or one and one β0 allele. The most severe form of the condition, known asthalassemia major, is present in people who have two β0-thalassemia alleles and results in transfusion-dependent anemia, severe transfusional iron overload, reduced life expectancy, and chelation therapy. A severe form of thalassemia with no globin chain product is called β0-thalassemia. Point mutations in the rendering region (exon) or exon-intron junction of the β-globin gene, which result in an unseasonable stop codon or the production of aberrant β-globin mRNA, are the primary cause. Absence of the β-globin chain product is the result of these anomalies [14]. The frequency of thalassemia is growing mainly in non-indigenous regions, similar as Northern Europe, North America, and Australia, due to increased mobility and migration overflows of populations in recent decades [12, 15, 16, 17, 18]. The global burden of hemoglobinopathies necessitates perpetration of public health interventions, similar as webbing programs and antenatal opinion, indeed in non-indigenous countries with high rates of immigration [15]. The genes that render for globin proteins are located on β-and α-globin gene clusters on chromosome 11 and 16, independently [4]. Expression of each globin gene varies throughout the embryonic and fetal development, which is why the Hb patterns of babe and grown-ups differ from each other [19, 20].

Advertisement

2. Epidemiology of beta-thalassemia

Beta-thalassemia is an in actuality normal blood inconvenience round the field. A huge amount of babies are considered with beta-thalassemia consistently. Beta-thalassemia happens limit of the time in people from Mediterranean nations, North Africa, the Center East, Central Asia, Southeast Asia, India, southern China, and the Far East, notwithstanding nations close by the northern coast of Africa and within side the South America. The most extreme exceptional carrier repeat is situated in Cyprus (14%), Sardinia (10.3%), and Southeast Asia [21]. High-incredible repeat of beta-thalassemia in those districts might be extremely probably to be perceived with the special strain of wilderness fever Plasmodium falciparum [21]. Populace resettlement and intermarriage among unique ethnic congregations have incited thalassemia in virtually all nations of the field, along with northern Europe, wherein thalassemia in the past did now presently do not exist. It has been normal that round 1.5% of the field’s general population (80 to 90 million people) are organizations of beta-thalassemia, with cycle 60,000 characteristic individuals transforming into pregnant every year, the especially gigantic a piece of the scene. Without a doubt the yearly cost of interesting people is normal at 1 out of 100,000 worldwide and 1 out of 10,000 within side the European Affiliation [10]. Regardless, there might be a deficiency of explicit records on delivery costs in several people gatherings, for the most part in areas of the field which can be perceived or anticipated to be emphatically impacted [12, 21]. As indicated by the Thalassemia Overall Association, around 200,000 victims stay with thalassemia dominating and are enrolled for standard cure worldwide [21]. The excellent perceived total of beta-thalassemia with phenomenal Hb or number one Hb form with thalassemic homes is HbE/beta-thalassemia, that is typically huge in Southeast Asia, wherein carrier.

2.1 Types and clinical description of beta-thalassemia

The aggregates of homozygous or hereditary heterozygous compound beta-thalassemias incorporate Cooley’s frailty and thalassemia intermedia [10]. People with Cooley’s frailty ordinarily come to clinical consideration inside the essential 2 years of life and need standard RBC bondings to endure. Those introducing later do not need bonding and get an analysis of thalassemia intermedia [22]. Thalassemia intermedia incorporates patients who present later and do not need customary bonding. Besides inside the uncommon predominant structures, heterozygous beta-thalassemia prompts the clinically quiet transporter state. HbE/beta-thalassemia and HbC/beta-thalassemia display a superb home as far as variety of aggregates and range of seriousness [11].

2.1.1 Beta-thalassemia major (Cooley’s anemia)

Beta-thalassemia significant alludes to a serious clinical aggregate that happens when patients are homozygous or compound heterozygous for more extreme beta chain transformations (for example serious B+/B+ changes, B+/B0, B0/B0) [10, 11].

Clinical show of thalassemia major happens somewhere in the range of 6 and 2 years. Youngsters foster dangerous weakness. They do not put on weight and develop at the normal rate (inability to flourish) and may create yellowing of the skin and whites of the eyes (jaundice) and become logically pale. Influenced people might have an amplified spleen, liver, and heart, and their bones might be distorted. Taking care of issues, looseness of the bowels, peevishness, repetitive episodes of fever, and reformist amplification of the midsection brought about by spleen and liver augmentation might happen. The clinical picture of thalassemia major is described by development impediment, paleness, jaundice, helpless musculature, genu valgum, hepatosplenomegaly, leg ulcers, improvement of masses from extramedullary hematopoiesis, and skeletal changes brought on by extension of the bone marrow in some developing nations where patients are untreated or ineffectively bonded due to a lack of resources. Skeletal modifications include frequent craniofacial changes and deformations of the long bones of the legs (bossing of the skull, conspicuous malar greatness, gloom of the extension of the nose, inclination to a mongoloid inclination of the eye, and hypertrophy of the maxillae, which will in general uncover the upper teeth). A few youths with thalassemia significant experience deferred adolescence. Many individuals with thalassemia major have such extreme manifestations that they need incessant blood bondings to recharge their red platelet supply. Over the long haul, a convergence of iron-containing hemoglobin from ongoing blood bondings can prompt a development of iron in the body, bringing about liver, heart, and chemical issues.

On the off chance that a customary bonding program that keeps a base Hb centralization of 9.5 to 10.5 g/dL is started, development and advancement will in general be ordinary up to 10 to 12 years [15]. Bonded patients might foster difficulties identified with iron over-burden. Entanglements of iron over-burden in youngsters incorporate development hindrance and disappointment or postponement of sexual development. Later iron over-burden-related inconveniences incorporate inclusion of the heart (widened myocardiopathy or once in a while arrythmias), liver (fibrosis and cirrhosis), and endocrine organs (diabetes mellitus, hypogonadism, and inadequacy of the parathyroid, thyroid, pituitary, and, less regularly, adrenal organs) [23]. Consistence with iron chelation treatment essentially impacts recurrence and seriousness of the iron over-burden-related complexities [24].

2.1.2 Beta-thalassemia intermedia

Beta-thalassemia intermedia is in the middle of clinical aggregate with heterogeneous hereditary changes that actually consider some beta chain creation (e.g., B+/B0, B+/B+). Some uncommon cases likewise exist in which both beta and alpha transformations exist together [11, 12].

Individuals with thalassemia intermedia present later than thalassemia major, have milder anemia and by definition do not require or only occasionally require transfusion. At the severe end of the clinical spectrum, patients present between the ages of 2 and 6 years and although they are capable of surviving without regular blood transfusion, growth and development are retarded. At the opposite finish of the range are patients who are totally asymptomatic until grown-up existence with just gentle frailty. The signs and side effects of thalassemia intermedia show up in youth or sometime down the road. Influenced people are gentle to direct sickliness and may likewise have slow development and bone anomalies. Hypertrophy of erythroid marrow with the chance of extramedullary erythropoiesis, a compensatory system of bone marrow to beat ongoing iron deficiency, is normal. It leads to common facial and bone deformities, osteoporosis with pathologic breaks in long bones, and the growth of erythropoietic masses that have a significant impact on the spleen, liver, lymph nodes, chest, and spine. The spleen’s important role in clearing damaged red blood cells from the circulatory system contributes to its growth. Extramedullary erythropoiesis may result in neurological problems, such as intrathoracic masses and spinal rope pressure with paraplegia. Gallstones may develop in thalassemia intermedia individuals more frequently than in thalassemia major because to ineffective erythropoiesis and fringe hemolysis [25]. Patients with thalassemia intermedia often foster leg ulcers and have an expanded inclination to apoplexy when contrasted with thalassemia major, particularly if splenectomised. Such occasions incorporate profound vein apoplexy, entrance vein apoplexy, stroke, and aspiratory embolism [26].

In spite of the fact that people with thalassemia intermedia are in danger of iron over-burden optional to expanded digestive iron retention, hypogonadism, hypothyroidism, and diabetes are not normal [27]. Ladies might have effective unconstrained pregnancies. Nonetheless, if blood bondings are fundamental during pregnancy, those never or negligibly bonded are in danger of creating hemolytic alloantibodies and erythrocyte autoantibodies. Intrauterine development hindrance, in spite of a customary bonding routine, has been accounted for [28]. Cardiovascular inclusion in thalassemia intermedia results chiefly from a high-yield state and aspiratory hypertension, while systolic left ventricle work is generally protected [29]. Pseudoxantoma elasticum, a diffuse connective tissue issue with vascular indication brought about by degeneration of the versatile lamina of the blood vessel divider and calcium statement, has been portrayed in such patients [30].

2.1.3 Beta-thalassemia minor (Beta-thalassemia carrier/trait)

Beta-thalassemia minor is a gentle clinical aggregate when one typical duplicate of the beta globulin quality is available (e.g., B+/B, B0/B). Transporters of thalassemia minor are normally clinically asymptomatic, however, here, and there have a gentle weakness. At the point when the two guardians are transporters, there is a 25% danger at every pregnancy of having kids with homozygous thalassemia [10, 11].

2.1.4 Dominant beta-thalassemia

Conversely, with the old style latent types of beta-thalassemia, which lead to a diminished creation of ordinary beta-globin chains, some uncommon transformations bring about the union of incredibly unsteady beta-globin variations which hasten in erythroid forerunners causing insufficient erythropoiesis [11]. These transformations are related with a clinically perceptible thalassemia aggregate in the heterozygote and are in this way alluded to as prevailing beta-thalassemias. The presence of hyper-temperamental Hb ought to be suspected in any person with thalassemia intermedia when the two guardians are hematologically ordinary, or in families with an example of autosomal prevailing transmission of the thalassemia intermedia aggregate. Beta-globin quality sequencing sets up the conclusion [6].

Most people who are heterozygous for a beta-thalassemia change have clinicopathological highlights depicted as “thalassemia minor”; for example, the blood count and film are strange yet there are no unusual actual discoveries or indications. Notwithstanding, a few transformations produce clinically obvious anomalies in heterozygotes, mostly splenomegaly, frailty, jaundice, and an expanded occurrence of gallstones. This is alluded to as predominant beta-thalassemia [6]. Predominant beta-thalassemia is uncommon; however, cases are found dissipated all through the world. The clinicopathological highlights are those of thalassemia intermedia. Red cell endurance is not exactly in run of the mill beta-thalassemia attribute and the reticulocyte count is expanded. Patients might require incidental blood bondings. There is extramedullary hematopoiesis, and iron over-burden might happen. The blood film is typically exceptionally unusual with conspicuous basophilic texturing and circling nucleated red cells. The bone marrow shows erythroid hyperplasia and dyserythropoiesis [6].

2.1.5 Beta-thalassemia associated with other Hb anomalies

The participation of HbE and beta-thalassemia achieves thalassemia totals going from a condition unclear from thalassemia major to a delicate sort of thalassemia intermedia. Dependent upon the earnestness of signs, three characterizations may be perceived [31]:

  • Mild HbE/beta-thalassemia: It is seen in around 15% of all cases in Southeast Asia. This get-together of patients stays aware of Hb levels some place in the scope of 9 and 12 g/dl and by and large does not cultivate clinically basic issues. No treatment is required.

  • Moderately genuine HbE/beta-thalassemia: The vast majority of HbE/beta-thalassemia cases fall into this class. The Hb levels stay at 6–7 g/dl, and the clinical signs resemble thalassemia intermedia. Bondings are not required aside from if sicknesses support further fragility. Iron over-weight may occur.

  • Severe HbE/beta-thalassemia: The Hb level can be basically just about as low as 4–5 g/dl. Patients in this social event show signs like thalassemia major and are treated as thalassemia critical patients [10].

Patients with HbC/beta-thalassemia may live freed from signs and be examined during routine tests. Exactly when present, clinical appearances are iron lack and improvement of the spleen. Blood bondings are just every so often required. Microcytosis and hypochromia are found for every circumstance. The blood film shows specific Hb C valuable stones with straight equivalent edges, target cells, and irregularly contracted cells with components of thalassemia like microcytosis [11]. The relationship of acquired ingenuity of fetal Hb (HPFH) with beta-thalassemia mitigates the clinical appearances which change from normal to thalassemia intermedia. Individuals with HbS/beta-thalassemia have a clinical course like that of Hb SS [10].

2.1.6 Beta-thalassemia associated with other features

Rarely, the beta-globin quality group does not contain the beta-thalassemia defect. The sub-atomic damage has been discovered in the quality encoding the record factor TFIIH (beta-thalassemia attribute related with tricothiodystrophy) or in the X-connected record factor GATA-1 (X-connected thrombocytopenia with thalassemia) in cases where the beta-thalassemia characteristic is related with different elements [32, 33, 34].

2.2 Signs and symptoms of beta-thalassemia

The majority of people with beta-thalassemia quality do not show any symptoms. Depending on the type of disorientation gained, different people will experience different side effects. Children with beta-thalassemia intermedia or major may not display any symptoms at all, although they usually develop them during the first 2 years of life. Beginning with one person and progressing to the next, the symptoms and severity of beta-thalassemia vary dramatically [10].

The most serious kind of beta-thalassemia is beta-thalassemia major. Children that are born with this type of personality will show signs early on in life, such as

  • Pale skin

  • Tiredness

  • Shortness of breath

  • Fussy

  • Having a poor appetite

  • Having many infections.

Over time more symptoms will appear, including:

  • Slowed growth

  • A fast heartbeat

  • Moodiness

  • Belly (abdominal) swelling

  • Yellowish skin and eyes (jaundice).

Individuals with beta-thalassemia or intermedia typically have a development of iron within the body, either from the particular illness or from the rehashed blood bondings. Abundance iron will hurt the center, liver, and endocrine framework. While not treatment, the spleen, liver, and heart become broadened. Bones will likewise end up to be meager, weak, and twisted. People with this condition would force continuous blood bondings and will not stick with it with a typical lifespan. Iron develops within the heart and totally different organs from blood bondings. This may cause vast breakdown as right time because the teenagers or middle 20s people with beta-thalassemia might need different real medical problems, including:

Thalassemia minima: this type often causes no symptoms but may have a mild anemia. Many individuals with beta thalassemia minor go through life never knowing they carry an altered gene for the disorder.

Thalassemia intermedia: folks determined to own beta monogenic disorder intermedia have a typically shifted articulation of the difficulty. Creditably extreme weakness is traditional, and influenced folks may need intermittent blood bondings. Each individual case is one among a form. This type will create aspect effects of moderate serious sickliness including:

  • Pallor

  • Extreme tiredness (fatigue)

  • Pale skin

  • Jaundice

  • Leg ulcers

  • Gallstones (cholelithiasis)

  • Slow or delayed growth

  • Weak bones

  • Abnormal enlargement of the liver and spleen

  • Moderate to severe skeletal malformations.

Predominant Beta-Thalassemia: Prevailing beta-thalassemia is a very uncommon structure where people who have one changed HBB quality foster specific manifestations related with beta-thalassemia. Influenced people might create gentle to direct sickliness, jaundice, and a strangely broadened spleen (splenomegaly).

2.3 Etiology of beta-thalassemia

Hemoglobin is made of two alpha proteins and two beta proteins. A quality change (transformation) in the alpha proteins causes alpha thalassemia. A quality change in the beta proteins causes beta-thalassemia. Most beta-thalassemia cases are brought about by a transformation in the HBB quality. In incredibly uncommon cases, a deficiency of hereditary material (erasure) that incorporates the HBB quality causes the issue [11]. In beta-thalassemia, the quality change causes an irregularity of hemoglobin proteins. The irregularity causes sickliness in light of the fact that [10]:

  • Red platelets separate quicker than ordinary.

  • Fewer RBCs are made.

  • Less hemoglobin is made.

The awkwardness additionally prompts clinical issues during the bones, bone marrow, and different organs. In excess of 200 changes have been so far announced; the larger part is point transformations in practically significant districts of the beta-globin quality [35]. Erasures of the beta-globin quality are remarkable. The beta-globin quality transformations cause a diminished or missing creation of beta-globin chains. Transformations in the HBB quality reason beta-thalassemia. The HBB quality gives guidelines to making a protein called beta-globin. Beta-globin is a part (subunit) of hemoglobin. Hemoglobin comprises four protein subunits, ordinarily two subunits of beta-globin and two subunits of another protein called alpha-globin [35].

The development of any beta-globin is prevented by a few modifications to the HBB quality. The term beta-zero (β0) thalassemia refers to a lack of beta-globin. Additional HBB quality alterations allow for the creation of some beta-globin, but in smaller amounts. Beta in addition to (β+) thalassemia is characterized by a decreased level of beta-globin. Possessing either a β0 or β+ thalassemia does not necessarily indicate how bad your condition will be; people with these two types have been found to have thalassemia major and thalassemia intermedia, respectively.

An absence of beta-globin prompts a diminished measure of practical hemoglobin. Without adequate hemoglobin, red platelets do not grow regularly, causing a deficiency of mature red platelets. People with beta-thalassemia have pallor and other related medical problems due to the low quantity of developed red platelets. Beta-thalassemia is the result of damaged or absent components. There are two distinct traits present. There are various varieties of this problem:

Cooley’s weakness (beta-thalassemia major). There are two damaged characteristics. The most severe form of this issue is this. Those with this ailment will need further blood bondings. They might not live out a typical lifespan.

Beta-thalassemia minor or thalassemia attribute. Just a single quality is harmed. This causes less extreme pallor. Individuals with this kind have a half shot at passing the quality to their kids. On the off chance that the other parent is not influenced, their kids will likewise have this type of the problem. This sort is additionally isolated into

  • Thalassemia minima: There are few or no symptoms.

  • Thalassemia intermedia: This causes moderate to severe anemia.

Many individuals with this problem are given iron substitution unintentionally. This happens when an absence of iron is accepted to cause their pallor. An excessive amount of iron can be hurtful. So, get the right determination.

2.3.1 Genetic modifiers

The modifying qualities are characterized by being hereditary variations that give rise to contrasts in the aggregate of the infection. In homozygous beta-thalassemia, the essential inherited modifiers that affect the clinical severity of the infection contain inherited variations that are ready to reduce the irregularity of the globin chain, creating a milder form of thalassemia [11]. These elements are the presence of quiet or mild beta-thalassemia alleles associated with a high residual yield of beta-globin, the co-inheritance of alpha-thalassemia, and, in addition, hereditary determinants ready for the incessant production of gamma-globin chains of support (HbF) in adult life [36]. Some beta-thalassemia transformations (e.g., deleting and not canceling delta-beta-thalassemia, local 5′ deletions of beta-globin quality) “essentially” increase the performance of gamma-globin quality [10]. Several transformations that extend HbF production are associated with deletional and non-deletional HPFH associated with the beta-globin quality package. Recently, the genome-wide affiliation approach, which specifically focuses on quantitative quality loci (QTL) that cause elevated HbF levels, has inherited components (e.g., changing the severity of homozygous beta-zero thalassemia [37].

The clinical aggregate of homozygous beta-thalassemia may also be altered by the interaction of other planned heritable variations outside the globin groups. These additional heritable modifiers have a profound effect in confounding the aggregate of thalassemia [11]. Some additional heritable modifiers have been recognized in recent years. The appearance of polymorphism (TA) 7 in the uridine diphosphate glucuronosyltransferase quality-reporting site, which in the homozygous state is associated with Gilbert’s disease, is a risk factor for the progression of cholelithiasis in patients with thalassemia major and intermedia [38]. Other competing qualities to alter the aggregate of thalassemia are the apolipoprotein E 4 allele and some HLA haplotypes, which appear to be inherited risk factors for left ventricular disappointment in homozygous beta-thalassemia [10, 39, 40]. For qualities related to iron digestion (e.g., C282Y and H63D-HFE quality transformations), less stable information was considered, probably since its effects on iron deposition are obscured by treatment (e.g., B. iron deposits auxiliary to the binding of red blood cells and iron chelation) and for qualities related to bone digestion [41, 42, 43]. Recently, a polymorphism in the quality of glutathione transferase M1 has been associated with an increased risk of heart iron overload in thalassemia major [44].

In certain cases, heterozygous beta-thalassemia could trigger the intermediate aggregation of thalassemia instead of the asymptomatic transporter state [10]. Most of these patients have an abundance of practical alpha globin qualities (triple or quadruple alpha quality), which increases the asymmetry in the ratio of the combination of alpha/non-alpha globin chains [36, 45].

2.3.2 Pathophysiology

A general oversupply of unbound alpha globin chains that speed in erythroid precursors in the bone marrow cause their unexpected passing and consequently lead to insufficient erythropoiesis. This is caused by a decreased amount (beta+) or nonexistence (beta0) of beta-globin chains. The concept of the transformation at the beta-globin quality located on chromosome 11 controls the degree of globin chain decline. As opposed to thalassemia intermedia, fringe hemolysis, which exacerbates illness, occurs when insoluble alpha globin attaches rapid layer damage to the fringe erythrocytes. Paleness animates the creation of erythropoietin with resulting concentrated yet inadequate development of the bone marrow (up 25 to multiple times ordinary), which thusly causes the regular recently portrayed bone disfigurements. Delayed and serious paleness and expanded erythropoietic drive additionally result in hepatosplenomegaly and extramedullary erythropoiesis [31].

2.3.3 Hereditary transmission

The beta-thalassemias square measures nonheritable in AN chromosome latent approach. The guardians of AN influenced juvenile square measure commit heterozygotes and convey a solitary duplicate of illness inflicting beta simple protein quality amendment. Mediterranean anemia intermedia square measures non-heritable in an chromosome passive example, which means the 2 duplicates of the HBB quality in each cell have changes. The parents of an individual with an autosomal recessive condition each carry one copy of the mutated gene, but they typically do not show signs and symptoms of the condition [11]. At times, in any case, people with just one HBB quality amendment in each cell foster light paleness. These somewhat influenced people square measure same to own Mediterranean anemia minor. At origination, each offspring of heterozygotes guardians has 25th shot at being influenced, shot at being AN symptomless transporter, and 25th shot at being unaffected and not transporter. The guardians of the proband have 25% hazard of getting to boot influenced youngsters in every gestation [31].

Predominant varieties of beta-thalassemia, connected with transformations that outcome within the creation of deeply unsound transferrin variations and prompting a clinically showing combination of beta-thalassemia in heterozygotes, are talked regarding on top of within the clinical portrayal section. In an exceedingly very little level of families, the HBB quality transformation is nonheritable in AN chromosome predominant approach. In these cases, one duplicate of the changed quality in each cell is up to cause the signs and manifestations of beta Mediterranean anemia [10].

Advertisement

3. Diagnosis beta-thalassemia

Beta-thalassemia is regularly found in individuals who are from Greek, Italian, African, or Asian beginning. The determination is frequently made somewhere in the range of 6 and 12 years of age. Thalassemia major is normally suspected in a newborn child more youthful than 2 years old with extreme microcytic frailty, gentle jaundice, and hepatosplenomegaly. Thalassemia intermedia presents at a later age with comparative yet milder clinical discoveries. Transporters are typically asymptomatic, however, at times might have gentle paleness. People with thalassemias have more modest measured red platelets than unaffected individuals just as low red platelet counts (pallor). Thalassemia major and thalassemia minor would now be able to be analyzed (and recognized from each other) by ordinary clinical and blood testing, yet additionally by atomic and hereditary tests. These tests license exact finding to be made whenever, even before birth (indeed, a long time before the beta chains are even incorporated) [33].

3.1 Hematologic diagnosis

RBC lists show microcytic iron deficiency. Thalassemia major is portrayed by diminished Hb level (50 < 70 fl and mean corpuscolar Hb (MCH) > 12 < 20 pg. Thalassemia intermedia is portrayed by Hb level somewhere in the range of 7 and 10 g/dl, MCV somewhere in the range of 50 and 80 fl and MCH somewhere in the range of 16 and 24 pg. Thalassemia minor is portrayed by diminished MCV and MCH, with expanded Hb A2 level [31].

Complete blood count (CBC): This test really looks at the size, number, and development of various platelets in a set volume of blood.

3.2 Smear of peripheral blood

Affected individuals have RBC morphologic alterations, including nucleated RBC, microcytosis, hypochromia, anisocytosis, and poikilocytosis (spiculated tear-drop and extended cells) (i.e., erythroblasts). After splenectomy, the number of erythroblasts significantly increases and is correlated with the degree of frailty. Compared to influenced people, transporters exhibit less severe RBC morphologic alterations. Usually, erythroblasts are not visible [22].

3.3 HPLC/electrophoresis

The Hb design in beta-thalassemia changes as indicated by beta-thalassemia type. In beta0 thalassemia, homozygotes HbA is absent and HbF constitutes the 92-95% of the total Hb. In beta+ thalassemia homozygotes and beta+/ beta0 genetic compounds HbA levels are between 10 and 30% and HbF between 70-90%. HbA2 is variable in beta-thalassemia homozygotes, and it is upgraded in beta-thalassemia minor [22]. Hb electrophoresis and HPLC likewise identify different hemoglobinopathies (S, C, E, OArab, Lepore) that might cooperate with beta-thalassemia [11].

Hemoglobin electrophoresis with hemoglobin F and A2 quantitation: A lab test that separates the kinds of hemoglobin [10]. Subjective and quantitative Hb investigation distinguishes the sum and kind of Hb present [10].

3.4 Molecular genetic analysis

The pervasiveness of a predetermined number of changes in every populace has incredibly worked with sub-atomic hereditary testing. Generally happening transformations of the beta-globin quality are recognized by PCR-based systems [33, 46]. The most generally utilized techniques are converse dab smudge examination or groundwork explicit enhancement, with a bunch of tests or preliminaries corresponding to the most well-known transformations in the populace from which the influenced individual started. Whenever designated change examination neglects to recognize the transformation, beta-globin quality succession investigation can be utilized to distinguish changes in the beta-globin quality [12].

3.5 Differential diagnosis

Barely any conditions share similitudes with homozygous beta-thalassemia [11]:

  • Not really settled sideroblastic anemias are effortlessly separated in view of ring sideroblasts in the bone marrow and dynamically raised serum grouping of erythrocyte protoporphyrin. Most sideroblastic anemias are related with abandons in the heme biosynthetic pathway, particularly delta-aminolevulinic corrosive synthase.

  • Congenital dyserythropoietic anemias do not have high HbF and do have other unmistakable components, like multinuclearity of the red platelet forerunners.

  • A few obtained conditions related with high HbF (adolescent ongoing myelomonocytic leukemia with ordinary karyotype, aplastic weakness both inherent and procured during the recuperation stage) might be confused with beta-thalassemia, despite the fact that they have exceptionally trademark clinical and hematological provisions [10].

Normal beta-thalassemia transporters are distinguished by examination of RBC files, which shows microcytosis (low MCV) and diminished substance of Hb per red cell (low MCH), and by subjective and quantitative Hb investigation, which shows the increment of HbA2 [11]. These examinations should be possible from a solitary blood test. In a pregnant lady, the child is analyzed utilizing CVS (chorionic villus inspecting) or amniocentesis.

Entanglements in transporter distinguishing proof by hematologic testing are:

  • Coinheritance of alpha-thalassemia, which might standardize the RBC records. Nonetheless, in alpha/beta twofold heterozygotes, the HbA2 fixation stays in the beta-thalassemia transporter reach and consequently is indicative. Accordingly, HbA2 assurance ought to consistently be performed for beta-thalassemia transporter ID.

  • Coinheritance of delta-thalassemia, which might diminish to ordinary the expanded Hb A2 levels common of the beta-thalassemia transporter state. Twofold heterozygosity for delta-and beta-thalassemia can be recognized from the most well-known alpha-thalassemia transporter state by globin chain blend or globin quality examination [10].

  • Silent transformations, i.e., extremely gentle changes related with reliable remaining yield of Hb beta chains and with ordinary RBC files and typical or fringe HbA2. The above detailed gatherings of transporters are alluded to as abnormal transporters.

At the point when the hematologic investigation is unusual, atomic hereditary testing of beta-globin quality is performed to distinguish the infection causing change [46].

3.6 Genetic counseling and prenatal diagnosis

Anticipation of beta-thalassemia depends on transporter distinguishing proof, hereditary advising, and pre-birth analysis [47]. Transporter identification has been recently portrayed. Hereditary guiding gives data to people and in danger couples (e.g., the two transporters) with respect to the method of legacy, the hereditary danger of having influenced kids and the regular history of the sickness including the accessible therapy and treatments being scrutinized. Pre-birth detection for pregnancies with a higher risk of complications is possible by analyzing the DNA of fetal cells obtained by amniocentesis, which is typically done at roughly 15 to 18 weeks of gestation, or by chorionic villi inspection at 11 weeks of development. Before pre-birth testing can be carried out, both alleles that cause the illnesses must be identified. Currently, fetal DNA in maternal plasma and fetal cells in maternal blood are being examined for the presence of the father’s transformation [48]. Families with the identified disease-causing mutations may be eligible for preimplantation hereditary conclusion.

3.7 Management of beta-thalassemia

3.7.1 Management of beta-thalassemia major

Babies with thalassemia major are well upon entering the world due to a unique type of hemoglobin present in the hatchling and infant [49]. At last, notwithstanding, this hemoglobin is supplanted by deficient hemoglobin. Manifestations arise late in the main year of life. The youngster creates fair skin, crabbiness, development impediment, expanding of the midsection because of extension of the liver and spleen (hepatosplenomegaly) with jaundice. This is related with serious iron deficiency with burst of the red platelets (hemolytic weakness). The kid with thalassemia major becomes reliant upon blood bondings and, despite the fact that they do help, they make further issues including iron over-burden. Folic corrosive supplementation is frequently given. Right now, the essential medicines are aimed at diminishing manifestations of the sickness. Chosen patients might fit the bill for bone marrow or undifferentiated cell transfers. Quality treatment stays a likely treatment for what is to come. The drawn-out trust is that thalassemia significant will be restored by addition of the ordinary beta-chain quality through quality treatment or by one more methodology of atomic medication [33, 49].

3.7.2 Transfusions

The objectives of bonding treatment are amendment of pallor, concealment of erythropoiesis, and hindrance of gastrointestinal iron retention, which happens in non-bonded patients as an outcome of expanded, albeit insufficient, erythropoiesis. The choice to begin bonding in patients with affirmed determination of thalassemia ought to be founded on the presence of serious weakness (Hb < 7 g/dl for over about 14 days, barring other contributory causes like contaminations). Nonetheless, additionally in patients with Hb > 7 g/dl, different variables ought to be thought of, including facial changes, helpless development, proof of hard extension and expanding splenomegaly. Whenever the situation allows, the choice to begin ordinary bondings ought not be postponed until after the 2nd-3rd year, because of the danger of fostering various red cell antibodies and resulting trouble in discovering reasonable blood givers. A few diverse transfusional regimens have been proposed throughout the long term, yet the most broadly acknowledged focuses on a pretransfusional Hb level of 9 to 10 g/dl and a post-bonding level of 13 to 14 g/dl. This forestalls development hindrance, organ harm and bone deformations, permitting ordinary action and personal satisfaction [23].

3.7.3 Management of thalassemia intermedia

Treatment of people with thalassemia intermedia is indicative [23, 50]. As hypersplenism might cause demolishing sickliness, hindered development and mechanical unsettling influence from the huge spleen, splenectomy is an applicable part of the administration of thalassemia intermedia. Dangers related with splenectomy incorporate an expanded helplessness to contaminations fundamentally from exemplified microbes (Streptococcus Pneumoniae, Haemophilus Influenzae, and Neisseria Meningitidis) and an increment in thromboembolic occasions [49]. Sepsis after splenectomy can be prevented through vaccination against the aforementioned microbes, anti-infection prophylaxis, and early anti-toxin therapy for fever and agitation. The gallbladder should be examined during splenectomy and removed if necessary to treat or prevent gallstones due to the increased prevalence of cholelithiasis and the risks of cholecystitis in splenectomized patients. Radiation therapy with hydroxycarbamide is used to treat extramedullary erythropoietic masses that are detected by attractive reverberation imaging. Managing a leg ulcer after it has developed is extremely difficult. Zinc supplementation and pentoxifylline, and the utilization of an oxygen chamber have been proposed for ulcer treatment. Hydroxycarbamide additionally has some advantage, either alone or with erythropoietin. As of late encouraging outcomes have been gotten with platelet inferred development factor. Since patients with thalassemia intermedia have a high danger of apoplexy, exacerbated by splenectomy, know about thrombotic inconveniences. Suggested treatment choices incorporate appropriate anticoagulation before careful or other high danger systems, platelet hostile to totaling specialists if there should arise an occurrence of thrombocytosis (platelet count higher than 700,000/mm3) and low sub-atomic weight heparin in patients with recorded apoplexy [33]. Since people with thalassemia intermedia may foster iron over-burden from expanded gastrointestinal assimilation of iron or from incidental bondings, chelation treatment is begun when the serum ferritin fixation surpasses 300 ng/ml or when iron over-burden is exhibited by immediate or circuitous techniques [51]. Beneficial folic corrosive can be recommended to patients with thalassemia intermedia to keep inadequacy from hyperactive bone marrow.

3.7.4 Improving ineffective erythropoiesis (IE) in thalassemia

Several various therapy modalities are currently being researched throughout the world thanks to recent developments in our understanding of the pathogenic mechanism behind thalassaemia. The investigational drug products that have recently entered the clinical phase are discussed in the section that follows, with an emphasis on their present and potential future relevance for clinical practice. The current standard of care for IE in thalassaemia, which includes blood transfusions and stem cell transplants as well as novel therapeutic approaches based on gene therapies, is not covered in this study and is available elsewhere.

The US Food and Drug Administration (FDA) in 2019 and the European Medicines Agency (EMA) in 2020 have both approved the first treatment for thalassemia, luspatercept, for TDT patients.

Mitapivat is a small-molecule, oral allosteric activator of RBC pyruvate kinase (PK-R), a crucial enzyme to control the synthesis of ATP through glycolysis (Figure 1).

Figure 1.

Distinction between healthy and erythropoiesis that is unsuccessful due to β-thalassemia [52, 53]. Erythropoiesis, which involves a number of proliferative and differentiation phases, is the process that converts hematopoietic stem cells into adult RBCs. The production of cell surface proteins, cell size reduction, progressive hemoglobinization, nuclear condensation, and nuclear extrusion are all temporally regulated processes that occur in conjunction with erythroid differentiation. The growth of extremely early erythroid precursors (proerythroblasts and earlier stages) and subsequent inefficient erythropoiesis are characteristics of β-thalassemia dyserythropoiesis in humans. A proliferating pool of immature erythroblasts is subject to ineffective erythropoiesis, which is characterized by (1) rapid erythroid differentiation, (2) maturation inhibition during the polychromatophilic stage, and (3) death of erythroid precursors [54, 55, 56].

3.7.5 Hematopoietic stem cell transplantation for thalassemia

Hemoglobinopathies are treated by allogeneic HSCT; following conditioning to get beyond the immune barrier, allogeneic stem cells are employed as vehicles to rectify the fundamental genetic flaw by re-inserting genes required for healthy hematopoiesis. Allogeneic HSCT is essentially allogeneic stem cell gene therapy in the treatment of various disorders. Autologous stem cells altered by the insertion of healthy genes may 1 day be used as vectors, but there is currently little sign that this strategy will be available in clinics anytime soon.

Thalassemia treatment with allogeneic hematopoietic stem cell transplantation (HSCT) has been a key component in the growth of HSCT. The allogeneic HSC with successful erythropoiesis is substituted for the thalassemic HSC having deficient erythropoiesis in order to treat thalassemia. This cellular replacement therapy results in the replacement of the entire hematopoietic system rather than just the damaged erythropoietic component. Nonetheless, it is a useful method to achieve a long-lasting, possibly permanent, clinically successful correction of hemolytic anemia, eliminating the need for transfusions and the difficulties that go along with them (i.e., iron overload).

3.8 Risk factors and complications of beta-thalassemia

Family ancestry and heritage are factors that increment the danger of beta-thalassemia. Contingent upon family ancestry, in case an individual’s folks or grandparents had beta-thalassemia major or intermedia, there is a 75% (3 out of 4) likelihood of the transformed quality being acquired by a posterity. Regardless of whether a youngster does not have beta-thalassemia major or intermedia, they can in any case be a transporter, potentially bringing about people in the future of their posterity having beta-thalassemia. Another danger factor is lineage. Beta-thalassemia happens regularly in individuals of Italian, Greek, Center Eastern, Southern Asian, and African heritage [57].

Complexities of beta-thalassemia change contingent upon the kind:

  • Thalassemia minima is gentle and causes no issues. However, the singular will be a transporter of the issue.

  • Thalassemia intermedia can cause issues dependent on the seriousness of the weakness. These issues incorporate postponed development, feeble bones, and augmented spleen.

  • Beta-thalassemia significant messes major up and can bring about early passing. Entanglements might incorporate postponed development, bone issues causing facial changes, liver and nerve bladder issues, expanded spleen, augmented kidneys, diabetes, hypothyroidism, and heart issues.

3.9 Prevention of beta-thalassemia

Beta-thalassemia is an innate sickness taking into consideration a safeguard therapy via transporter screening and pre-birth conclusion. It very well may be forestalled in the event that one parent has ordinary qualities, leading to screenings that engage transporters to choose accomplices with typical hemoglobin [58]. This screening technique demonstrated obtuse in populaces of West African family line in light of the markers has high pervasiveness of alpha thalassemia. Nations have programs dispersing data about the regenerative dangers related with transporters of haemoglobinopathies. Thalassemia transporter screening programs have instructive projects in schools, military, and through broad communications just as giving guiding to transporters and transporter couples [12]. Screening has shown diminished rate; by 1995 the commonness in Italy decreased from 1:250 to 1:4000, and a 95% abatement around there. The reduction in occurrence has helped those influenced with thalassemia, as the interest for blood has diminished, accordingly working on the stock of treatment.

Advertisement

4. Conclusions

Thalassemia is a perplexing condition that needs in-depth approach for lab testing and analysis. High-throughput testing algorithms and procedures are needed for population screening in high-prevalence areas, which increases the difficulty of the diagnosis. Despite this, current best-practice guidelines and protocols are capable of the successful regulation of complicated laboratory procedures, coupled with essential EQA programs. Many novel methods are now being developed that have the potential to improve the accuracy, throughput, and efficacy of laboratory diagnostics of thalassemia and other hemoglobinopathies.

The majority of families cannot afford the estimated US $ 3200 per child per year expense of treating serious thalassemia disease. Thalassemia management is not only distressing for the family but also has a significant socioeconomic impact on the nation, making its prevention and control a top priority. So, the first step toward easing the disease’s burden is to prevent the birth of any afflicted fetuses.

Prenatal diagnosis (PND), genetic counseling, carrier screening, and termination of the afflicted fetus are all parts of prevention. This strategy is affordable and reducing the prevalence of thalassemia in many nations with remarkable results. Understanding the range and distribution of thalassemia mutations in a given population is a requirement for an efficient and quick prenatal diagnosis/genetic counseling.

References

  1. 1. De Sanctis V, Kattamis C, Canatan D, Soliman AT, Elsedfy H, Karimi M, et al. β-Thalassemia distribution in the old world: An ancient disease seen from a historical standpoint. Mediterranean Journal of Hematology and Infectious Diseases. 2017;9:e2017018
  2. 2. Modell B, Darlison M. Global epidemiology of haemoglobin disorders and derived service indicators. Bulletin of the World Health Organization. 2008;86:480-487
  3. 3. Williams TN, Weatherall DJ. World distribution, population genetics, and health burden of the hemoglobinopathies. Cold Spring Harbor Perspectives in Medicine. 2012;2:a011692
  4. 4. Thein SL. The molecular basis of β-thalassemia. Cold Spring Harbor Perspectives in Medicine. 2013;3:a011700
  5. 5. Taher AT, Weatherall DJ, Cappellini MD. Thalassaemia. Lancet. 2018;391:155-167
  6. 6. Bain BJ. The alpha, beta, delta and gamma thalassaemias and related conditions. In: Haemoglobinopathy Diagnosis. 2nd ed. Oxford: Wiley-Blackwell; 2006. pp. 63-138
  7. 7. Cao A, Kan YW. The prevention of thalassemia. Cold Spring Harbor Perspectives in Medicine. 2013;3:a011775
  8. 8. Angastiniotis M, Eleftheriou A, Galanello R, Harteveld CL, Petrou M, Traeger-Synodinos J, et al. Prevention of Thalassaemias and Other Haemoglobin Disorders: Volume 1: Principles [Internet]. Old J, editor. 2nd ed. Nicosia (Cyprus): Thalassaemia International Federation; 2013. PMID: 24672827
  9. 9. Flint J, Harding RM, Boyce AJ, Clegg JB. The population genetics of the haemoglobinopathies. Baillière's Clinical Haematology. 1998;11(1):1-51
  10. 10. Origa R. Beta-Thalassemia. 2000 Sep 28 [updated 2018 Jan 25]. In: Adam MP, Ardinger HH, Pagon RA, Wallace SE, LJH B, Stephens K, Amemiya A, editors. GeneReviews® [Internet]. Seattle; 1993-2020: Seattle (WA): University of Washington; 2020 Available from: http://www.ncbi.nlm.nih.gov/books/NBK1426/CitationonPubMed
  11. 11. Rund D, Rachmilewitz E. Beta-thalassemia. The New England Journal of Medicine. 2005;353(11):1135-1146
  12. 12. Akhavan-Niaki H, Derakhshandeh-Peykar P, Banihashemi A, Mostafazadeh A, Asghari B, Ahmadifard MR, et al. A comprehensive molecular characterization of beta thalassemia in a highly heterogeneous population. Blood Cells, Molecules & Diseases. 2011;47(1):29-32
  13. 13. Islam MT, Sarkar SK, Sultana N, Begum MN, Bhuyan GS, Talukder S, et al. High resolution melting curve analysis targeting the HBB gene mutational hot-spot offers a reliable screening approach for all common as well as most of the rare beta-globin gene mutations in Bangladesh. BMC Genetics. 2018;19(1):1
  14. 14. Thein SL. Genetic modifiers of beta-thalassemia. Haematologica. 2005;90(5):649-660
  15. 15. Aguilar Martinez P, Angastiniotis M, Eleftheriou A, Gulbis B, Manú Pereira M, Del Petrova-Benedict R, et al. Haemoglobinopathies in Europe: Health & migration policy perspectives. Orphanet Journal of Rare Diseases. 2014;9:97
  16. 16. Angastiniotis M, Vives Corrons JL, Soteriades ES, Eleftheriou A. The impact of migrations on the health services for rare diseases in Europe: The example of haemoglobin disorders. Scientific World Journal. 2013;2013:727905
  17. 17. Modell B, Darlison M, Birgens H, Cario H, Faustino P, Giordano PC, et al. Epidemiology of haemoglobin disorders in Europe: An overview. Scandinavian Journal of Clinical and Laboratory Investigation. 2007;67:39-70
  18. 18. Piel FB, Tatem AJ, Huang Z, Gupta S, Williams TN, Weatherall DJ. Global migration and the changing distribution of sickle haemoglobin: A quantitative study of temporal trends between 1960 and 2000. The Lancet Global Health. 2014;2:e80-e89
  19. 19. Sankaran VG, Orkin SH. The switch from fetal to adult hemoglobin. Cold Spring Harbor Perspectives in Medicine. 2013;3:a011643
  20. 20. Wang X, Thein SL. Switching from fetal to adult hemoglobin. Nature Genetics. Apr 2018;50(4):478-480. DOI: 10.1038/s41588-018-0094-z. PMID: 29610477; PMCID: PMC6419756
  21. 21. Vichinsky EP. Changing patterns of thalassemia worldwide. Annals of the New York Academy of Sciences. 2005;2005(1054):18-24
  22. 22. Cao A, Galanello R. Beta-thalassemia. Genetics in Medicine. 2010;12:61-76
  23. 23. Borgna-Pignatti C, Galanello R. Thalassemias and related disorders: Quantitative disorders of hemoglobin synthesis. In: Wintrobe's Clinical Hematology. 11th ed. Vol. 42. Philadelphia: Lippincott Williams & Wilkins; 2004. pp. 1319-1365
  24. 24. Borgna-Pignatti C, Cappellini MD, De Stefano P, Del Vecchio GC, Forni GL, Gamberini MR, et al. Survival and complications in thalassemia. Annals of the New York Academy of Sciences. 2005;1054:40-47
  25. 25. Galanello R, Piras S, Barella S, Leoni GB, Cipollina MD, Perseu L, et al. Cholelithiasis and Gilbert's syndrome in homozygous betathalassemia. British Journal of Haematology. 2001;115:926-928
  26. 26. Taher AT, Otrock ZK, Uthman I, Cappellini MD. Thalassemia and hypercoagulability. Blood Reviews. 2008;22:283-292
  27. 27. De Sanctis V, Tangerini A, Testa MR, Lauriola AL, Gamberini MR, Cavallini AR, et al. Final height and endocrine function in thalassemia intermedia. Journal of Pediatric Endocrinology & Metabolism. 1998;11:965-971
  28. 28. Nassar AH, Naja M, Cesaretti C, Eprassi B, Cappellini MD, Taher A. Pregnancy outcome in patients with beta-thalassemia intermedia at two tertiary care centers, in Beirut and Milan. Haematologica. 2008;93:1586-1587
  29. 29. Aessopos A, Farmakis D, Deftereos S, Tsironi M, Tassiopoulos S, Moyssakis I, et al. Thalassemia heart disease: A comparative evaluation of thalassemia major and thalassemia intermedia. Chest. 2005;127:1523-1530
  30. 30. Aessopos A, Farmakis D, Loukopoulos D. Elastic tissue abnormalities resembling pseudoxanthoma elasticum in beta thalassemia and the sickling syndromes. Blood. 2002;99:30-35
  31. 31. Galanello R, Origa R. Beta-thalassemia. Orphanet Journal of Rare Diseases. 2010;5:11. DOI: 10.1186/1750-1172-5-11
  32. 32. Freson K, Matthijs G, Thys C, Marien P, Hoylaerts MF, Vermylen J, et al. Different substitutions at residue D218 of the X-linked transcription factor GATA1 lead to altered clinical severity of macrothrombocytopenia and anemia and are associated with variable skewed X inactivation. Human Molecular Genetics. 2002;11:147-152
  33. 33. Jarjour RA, Murad H, Moasses F, Al-Achkar W. Molecular update of β-thalassemia mutations in the Syrian population: Identification of rare β-thalassemia mutations. Hemoglobin. 2014;38(4):272-276
  34. 34. Viprakasit V, Gibbons RJ, Broughton BC, Tolmie JL, Brown D, Lunt P, et al. Mutations in the general transcription factor TFIIH result in beta- thalassemia in individuals with trichothiodystrophy. Human Molecular Genetics. 2001;10:2797-2802
  35. 35. Giardine B, van Baal S, Kaimakis P, Riemer C, Miller W, Samara M, et al. HbVar database of human hemoglobin variants and thalassemia mutations: 2007 update. Human Mutation. 2007;28:206
  36. 36. Galanello R, Cao A. Relationship between genotype and phenotype. Thalassemia intermedia. Annals of the New York Academy of Sciences. 1998;850:325-333
  37. 37. Uda M, Galanello R, Sanna S, Lettre G, Sankaran VG, Chen W, et al. Genome-wide association study shows BCL11A associated with peristent fetal hemoglobin and amelioration of the phenotype of beta-thalassemia. Proceedings of the National Academy of Sciences of the United States of America. 2008;105:1620-1625
  38. 38. Origa R, Galanello R, Perseu L, Tavazzi D, Domenica Cappellini M, Terenzani L, et al. Cholelithiasis in thalassemia major. European Journal of Haematology. 2009;82:22-25
  39. 39. Economou-Petersen E, Aessopos A, Kladi A, Flevari P, Karabatsos F, Fragodimitri C, et al. Apolipoprotein E epsilon4 allele as a genetic risk factor for left ventricular failure in homozygous betathalassemia. Blood. 1998;92:3455-3459
  40. 40. Kremastinos DT, Flevari P, Spyropoulou M, Vrettou H, Tsiapras D, Stavropoulos-Giokas CG. Association of heart failure in homozygous beta-thalassemia with the major histocompatibility complex. Circulation. 1999;100:2074-2078
  41. 41. Dresner Pollack R, Rachmilewitz E, Blumenfeld A, Idelson M, Goldfarb AW. Bone mineral metabolism in adults with β-thalassemia major and intermedia. British Journal of Haematology. 2000;111:902-907
  42. 42. Longo F, Zecchina G, Sbaiz L, Fischer R, Piga A, Camaschella C. The influence of hemochromatosis mutations on iron overload of thalassemia major. Haematologica. 1999;84:799-803
  43. 43. Perrotta S, Cappellini MD, Bertoldo F, Servedio V, Iolascon G, D'Agruma L, et al. Osteoporosis in β-thalassemia major patients: Analysis of the genetic background. British Journal of Haematology. 2000;111:461-466
  44. 44. Origa R, Satta S, Matta G, Galanello R. Glutathione S-transferase gene polymorphism and cardiac iron overload in thalassemia major. British Journal of Haematology. 2008;142:143-145
  45. 45. Sollaino MC, Paglietti ME, Perseu L, Giagu N, Loi D, Galanello R. Association of alpha globin gene quadruplication and heterozygous beta thalassemia in patients with thalassemia intermedia. Haematologica. 2009;94:1445-1448
  46. 46. Vrettou C, Traeger-Synodinos J, Tzetis M, Malamis G, Kanavakis E. Rapid screening of multiple betaglobin gene mutations by real-time PCR on the LightCycler: Application to carrier screening and prenatal diagnosis of thalassemia syndromes. Clinical Chemistry. 2003;49:769-776
  47. 47. Cao A, Galanello R, Rosatelli MC. Prenatal diagnosis and screening of the haemoglobinopathies. Baillière's Clinical Haematology. 1998;11:215-238
  48. 48. Mavrou A, Kouvidi E, Antsaklis A, Souka A, Kitsiou Tzeli S, Kolialexi A. Identification of nucleated red blood cells in maternal circulation: A second step in screening for fetal aneuploidies and pregnancy complications. Prenatal Diagnosis. 2007;27:150-153
  49. 49. Wonke B. Clinical management of beta-thalassemia major. Seminars in Hematology. 2001;38(4):350-359
  50. 50. Borgna-Pignatti C. Modern treatment of thalassaemia intermedia. British Journal of Haematology. 2007;138:291-304
  51. 51. Origa R, Galanello R, Ganz T, Giagu N, Maccioni L, Faa G, et al. Liver iron concentrations and urinary hepcidin in beta-thalassemia. Haematologica. 2007;92:583-588
  52. 52. Centis F, Tabellini L, Lucarelli G. The importance of erythroid expansion in determining the extent of apoptosis in erythroid precursors in patients with 𝛽-thalassemia major. Blood. 2000;96(10):3624-3629
  53. 53. Yuan J, Angelucci E, Lucarelli G. Accelerated programmed cell death (apoptosis) in erythroid precursors of patients with severe 𝛽-thalassemia (Cooley’s anemia). Blood. 1993;82(2):374-377
  54. 54. de Franceschi L, Bertoldi M, de Falco L. Oxidative stress modulates heme synthesis and induces peroxiredoxin-2 as a novel cytoprotective response in 𝛽-thalassemic erythropoiesis. Haematologica. 2011;96(11):1595-1604
  55. 55. Leecharoenkiat A, Wannatung T, Lithanatudom P. Increased oxidative metabolism is associated with erythroid precursor expansion in 𝛽0-thalassaemia/Hb E disease. Blood Cells, Molecules and Diseases. 2011;47(3):143-157
  56. 56. Mathias LA, Fisher TC, Zeng L. Ineffective erythropoiesis in 𝛽-thalassemia major is due to apoptosis at the polychromatophilic normoblast stage. Experimental Hematology. 2000;28(12):1343-1353
  57. 57. Mayo Clinic. Risk factors. 2017. Available from: http://www.mayoclinic.org/diseases-conditions/thalassemia/symptoms-causes/dxc-20261829). Retrieved 14 Oct 2020
  58. 58. Cousens NE, Gaff CL, Metcalfe SA, Delatycki MB. Carrier screening for Betathalassaemia: A review of international practice. European Journal of Human Genetics. 2010;18(10):1077-1083

Written By

Kenneth Oshiokhayamhe Iyevhobu, Omolumen Lucky E., Tobechukwu Joseph Okobi, Edidiong Raphael Usoro, A. Airefetalor Ivie, Benedicta A. Ken-Iyevhobu and O. Omokpo Victoria

Submitted: 20 October 2022 Reviewed: 25 April 2023 Published: 22 November 2023