Sunteți pe pagina 1din 11

review

Genetic insights into the clinical diversity of b thalassaemia

The identification of defective genes underlying inherited


b globin gene – structure, function and
diseases has made it clear that patients with the same genotype
expression
2 can have very variable patterns of clinical expression. The
profound phenotypic variability of the b thalassaemias, b globin is encoded by a structural gene found in a cluster with
including age of onset, varying involvement of different organs the other b like genes on the short arm of chromosome 11. The
and transfusion needs, is prototypical of how the wide cluster contains five functional genes, 5¢--Gc-Ac-wb-d-b-3¢,
spectrum in disease severity of a monogenic disorder can be which are arranged in the order of their developmental
generated. Relating phenotype to genotype is complicated not expression. Upstream of the entire b globin complex is the
only by the complex interaction of the environment with the locus control region (LCR), which consists of five DNase 1
different allelic variants, but interaction with other genetic hypersensitive (HS) sites (designated HS 1–5) distributed
factors at the secondary and tertiary levels is also involved. between 6 and 20 kb 5¢ of the  gene. The LCR plays a critical
Evidence for these modifier genes comes from the range of role in b globin gene expression by maintaining an open
phenotypes within families sharing the same genotypes. This chromatin state and acting as a powerful enhancer of globin
article presents an overview of the b globin gene structure, gene transcription; in its absence, the level of b globin gene
function and expression, followed by a short description of the expression is low. Four of the sites (HS 1–4) are erythroid-
clinical and haematological diversity encountered in b thalas- specific, encompassing binding sequences for erythroid-restric-
saemia and the underlying pathophysiology. A discussion of ted transcription factors (GATA-1 and NF-E2), while HS5 is
the genetic basis of the disease is presented with an overview of ubiquitous. There is one other hypersensitive site approxi-
the various genetic loci (modifier genes) that modulate the mately 20 kb 3¢ to the b gene. The two extreme HS sites
effects of its clinical expression on different organs. flanking the b complex have been suggested to mark the
boundaries of the b globin gene domain. The b globin complex
is embedded in a cluster of olfactory receptor genes (ORG),
Introduction
part of the family of approximately 1000 genes that are widely
b thalassaemia can be broadly defined as a syndrome of distributed throughout the genome, and expressed in the
inherited haemoglobin disorders characterized by a quanti- olfactory epithelium (Bulger et al, 2000).
tative deficiency of functional b globin chains. Although it is The general structure of the b globin gene is typical of the
defined as a reduction in the synthesis of b globin, some forms other globin loci (Efstratiadis et al, 1980). The genomic
result from structural haemoglobin variants that are ineffec- sequence, which codes for 146 amino acids, spans 1600 bp;
tively synthesized or are so unstable that they result in a the transcribed region is contained in three exons separated by
functional deficiency of the b chains and a thalassaemia two introns or intervening sequences (IVSs). Exon 2 encodes
phenotype (Weatherall & Clegg, 2001). The most common the residues involved in haem binding and ab dimer forma-
forms are those that are prevalent in the malarial tropical and tion, while exons 1 and 3 encode for the non-haem-binding
subtropical regions, where a few mutations have reached high regions of the b globin chain. Many of the amino acids
gene frequencies because of the protection they provide against involved in globin subunit interactions required for the Bohr
malaria. In these countries where b thalassaemia is prevalent, a effect, and 2,3-diphosphoglycerate binding, are found in exon
limited number of alleles (four to six) account for more than 3. Conserved sequences important for b globin gene expression
90% of the b thalassaemia (Flint et al, 1998), allowing a are found in the 5¢ promoter region, at the exon–intron
targeted molecular diagnostic approach to be undertaken. In junctions, and in the 3¢ untranslated region (3¢-UTR) at the
other countries such as the UK, where there is an ethnic mix end of the mRNA sequences. The b globin gene promoter
because of recent population migrations, a screening approach includes three positive cis-acting elements: a TATA box
may be more appropriate and effective. (positions )28 to )31), a CCAAT box (positions )2 to )76)
and duplicated CACCC motifs (proximal at positions )86 to
)90, and distal at position )101 to )105). While the CCAAT
and TATA elements are found in many eukaryotic promoters,
Correspondence: Professor Swee Lay Thein, Department of
the CACCC sequence is found predominantly in erythroid cell-
Haematological Medicine Guy’s, King’s & St Thomas’ School of
specific promoters. Binding of the erythroid Krüppel like
Medicine, Denmark Hill Campus, Bessemer Road, London SE5 9PJ.
factor (EKLF) to the CACCC motif appears to be crucial for
E-mail: sl.thein@kcl.ac.uk

264 ª 2004 Blackwell Publishing Ltd, British Journal of Haematology, 124, 264–274
Review

normal adult b globin expression. In addition to these motifs, gene is up-regulated by promoter mutations, as illustrated by
the region upstream of the b globin promoter contains two the non-deletional hereditary persistence of fetal haemoglobin
binding motifs for the erythroid transcription factor GATA-1. (HPFH) (Wood, 2001). In addition, mutations that affect the
The importance of these various 5¢–flanking sequences for b promoter, which remove competition for the b-LCR, tend to
normal gene expression is underscored by b thalassaemia be associated with variable increases in the c and d gene
arising from point mutations in these sequences specifically in expression (Huisman, 1997; Thein, 1998).
and around the TATA box and the CACCC motifs in the )80
to )100 region. An enhancer is also found in intron 2
Pathophysiology and clinical diversity
(Antoniou et al, 1988) and 3¢ of the globin gene, 600–900 bp
of b thalassaemia
downstream of the poly (A) site (Trudel & Costantini, 1987).
The 5¢-UTR occupies a region of 50 nucleotides between the The underlying pathophysiology of b thalassaemia relates to a
CAP site, the start of transcription, and the initiation (ATG) quantifiable deficiency of functional b globin chains, which
codon. There are two prominently conserved sequences in the leads to an imbalanced globin chain production and an excess
5¢-UTR of the various globin genes (both a and b) (Collins of a globin chains (Weatherall & Clegg, 2001; Schrier, 2002)
& Weissman, 1984). One is the CTTCTG hexanucleotide found (see Fig 1). The latter aggregate in red cell precursors, forming
8–13 nucleotides downstream from the CAP site, i.e. at positions inclusion bodies that cause mechanical damage and their
+8 to +13. The second conserved sequence is CACCATG, in premature destruction in the bone marrow, i.e. ineffective
which the last three nucleotides form the initiation codon erythropoiesis. Red cells that survive to reach the peripheral
(ATG). Again, the importance of these sequences in the circulation are prematurely destroyed in the spleen. Anaemia
regulation of the b gene expression is exemplified by several in b thalassaemia thus results from a combination of
mutations in the 5¢-UTR causing b thalassaemia. ineffective erythropoiesis, peripheral haemolysis and an overall
The 3¢-UTR constitutes the region of 132 nucleotides reduction in haemoglobin synthesis. Factors which reduce the
between the termination codon (TAA) and the poly (A) tail degree of chain imbalance and the magnitude of a chain excess
with one conserved sequence, AATAAA, located 20 nucleotides in the red cell precursors, have an ameliorating effect on the b
upstream of the poly (A) tail. Several mutations affecting the thalassaemia phenotype.
AATAAA sequence and other sequences in the 3¢-UTR causing A direct effect of the anaemia is the increased production of
b thalassaemia have been described. erythropoietin, which leads to intense proliferation and
The developmental regulation of the globin genes reflect expansion of the bone marrow with the resulting skeletal
their sequential activation in a 5¢)3¢ direction. Transcription deformities. These secondary complications of bone disease,
of the  gene in the embryonic yolk gene switches after the splenomegaly, endocrine and cardiac damage can be related to
sixth week of gestation to the transcription of the two c genes the severity of anaemia and the iron loading that results from
in the fetal liver, and then around the prenatal period, to that the increased gastrointestinal absorption and the blood
of the d (minor adult) and b (major adult) genes. At 6 months transfusions. Recently, it has become apparent that these
after birth, Hb F comprises <5% of the total haemoglobin and complications of b thalassaemia may also be genetically
continues to fall reaching the adult level of <1% at 2 years of modified by variability at other loci (tertiary modifiers).
age. It is at this stage that mutations affecting the b gene The clinical manifestations of b thalassaemia are extremely
become clinically apparent. The ‘switch’ from fetal (c) to adult diverse, spanning a broad spectrum from the transfusion-
(b) haemoglobin production is not complete, and small dependent state of thalassaemia major to the asymptomatic
amounts of c expression persist in adult life. All adults have state of thalassaemia trait. The most severe end of the spectrum
residual amounts of fetal haemoglobin (Hb F, a2c2), present in is characterized by the complete absence of b globin production
a subset of erythrocytes called F cells which also contain adult and results from the inheritance of two b thalassaemia alleles,
(a2b2) haemoglobin (Boyer et al, 1975). These levels of Hb F homozygous or compound heterozygous states. This combi-
and F cells in adults vary considerably, and are largely nation of genotype usually results in b thalassaemia major and,
genetically controlled (Garner et al, 2000a). at their worst, the patients present within 6 months of life, and
The developmental expression of the individual globin genes if not treated with regular blood transfusions, die within their
relies on two mechanisms, gene silencing and gene competi- first 2 years. Conversely, many patients who have inherited two
tion, governed by direct physical interactions between the b thalassaemia alleles may have a milder disease, ranging from a
globin promoters and the b-LCR (Carter et al, 2002; Tolhuis condition that is only slightly less severe than transfusion
et al, 2002), which are dependent on the transcription dependence through a spectrum of decreasing severity to one
environment in embryonic, fetal and adult cells. While the that is asymptomatic and often mistaken as b thalassaemia trait.
- and c globin genes are autonomously silenced at the This diverse collection of phenotypes between the two extremes
appropriate developmental stage, expression of the adult b of thalassaemia major and thalassaemia trait constitute the
globin gene depends on the lack of competition from the c clinical syndrome of thalassaemia intermedia. In a large
gene for the LCR sequences. This is supported by the number of patients with thalassaemia intermedia, the reduced
concomitant down-regulation of the cis b gene when the c disease severity can be explained by the inheritance of the

ª 2004 Blackwell Publishing Ltd, British Journal of Haematology, 124, 264–274 265
Review

Fig 1. Pathophysiology of b thalassaemia. Factors that modify the b thalassaemia phenotype act at three levels: (1) primary – generally these refer to
the nature of the mutation affecting the b globin gene itself, (2) secondary – loci that affect the a/b globin chain imbalance and include the a and c
globin genes. However, loci that affect the stability and amount of the a chain and loci that affect expression of the c globin should also be included;
(3) tertiary – loci that are not involved in globin imbalance but modify the complications of the disease in different ways. These include jaundice and
gallstones (UGTA1), iron loading (HFE) and bone disease (VDR, OesR, COL1A1).

milder b thalassaemia alleles (b++ and ‘silent’) that allow the cases, the b thalassaemia allele can be phenotypically ‘silent’,
production of a significant proportion of b globin chains. A with no anaemia or any haematological abnormalities. In
substantial number, however, have b thalassaemia, and in such others, the heterozygous state causes a phenotype almost as
cases, the absence of b globin chains is compensated by an severe as the major forms, that is, the b thalassaemia allele is
inherent ability to produce fetal haemoglobin (Hb F a2c2). Yet dominantly inherited.
other thalassaemia patients have inherited only one b thalas- Although definition of the two extremes of the clinical
saemia allele. Most cases of unusually severe heterozygous b spectrum of b thalassaemia is easy, assigning the severity of the
thalassaemia are due to the co-inheritance of extra a globin intermediate form can be problematical. Criteria such as age
genes (see below) while others are due to the nature of the and level of haemoglobin at presentation, transfusion history
underlying b thalassaemia mutation itself (see ‘Dominantly and the requirements for intermittent blood transfusion have
inherited b thalassaemia’). Hence, the underlying genotypes of been used, but these have their inherent limitations and are
b thalassaemia intermedia are extremely heterogeneous; the highly subjective and clinician dependent.
genetic basis can be the inheritance of one or two b
thalassaemia alleles interacting with other genetic variables.
Mechanisms underlying phenotypic diversity of
Carriers for b thalassaemia are typically clinically asympto-
b thalassaemia
matic; they may have a mild anaemia with characteristic
hypochromic microcytic red blood cells, elevated levels of Progress in our understanding of the mechanisms underlying
HbA2 and variable levels of Hb F. However, even the the remarkable phenotypic variability of b thalassaemia has
heterozygous states for b thalassaemia show a phenotypic been made possible by a combination of the analysis of the
diversity comparable with that of thalassaemia major. In some molecular basis of the different forms of thalassaemia, family

266 ª 2004 Blackwell Publishing Ltd, British Journal of Haematology, 124, 264–274
Review

studies and analysis of the genotype/phenotype relationship of found in exon 1 of the b globin gene, spanning codons 24–27.
the thalassaemia intermedia. Three mutations within this region activate this cryptic site,
which acts as an alternative donor site in RNA processing. The
mutations in codon 26 (GAG fi AAG) that gives rise to HbE
Primary modifiers – heterogeneity and variable severity of
(b 26 Gln fi Lys) is one such mutation that activates this
b thalassaemia alleles
cryptic splice site, with a reduction of the normal splicing that
With the exception of a few deletions, the vast majority of b produces the HbE variant. As HbE production is also
thalassaemias are caused by point mutations within the gene or quantifiably reduced, the compound heterozygous state,
its immediate flanking sequences. A few b thalassaemia HbE/ b thalassaemia results in a clinical picture closely
mutations that segregate independently of the b globin cluster resembling homozygous b thalassaemia, ranging from severe
have been described in several families (Thein, 1998); in such anaemia and transfusion-dependency to thalassaemia inter-
cases, trans-acting regulatory factors have been implicated. media. Other RNA processing mutants affect the polyadeny-
Examples of reduced b globin production caused by loci lation signal (AATAAA) and the 3¢-UTR. These are generally
outside the b globin complex include those mutations arising mild b+ thalassaemia alleles.
in the general transcription factor TF11H (Viprakasit et al, Mutations that abrogate mRNA translation either at the
2001) and the erythroid-specific transcription factor GATA1 initiation or extension phases of globin synthesis are all
(Yu et al, 2002). associated with a b phenotype. Approximately half of the b
thalassaemia alleles are characterized by premature termin-
b versus b+ thalassaemia alleles. Functionally, the b ation of b chain extension. They result from the introduction
thalassaemia alleles can be classified as b or b+ reflecting the of premature termination codons due to frameshifts or
resulting phenotype: b thalassaemia in which there is a nonsense mutations and nearly all terminate within exons 1
complete absence of b globin production and the most severe and 2. These mutations are associated with minimal steady-
possible, and b+ thalassaemia in which there is some, albeit state levels of mutant b mRNA in erythroid cells. In
reduced b globin product. Mild b thalassaemia, sometimes heterozygotes for such cases, no b chain is produced from
referred to as b++, alleles allow a moderate amount of b globin the mutant allele and only half the normal b globin is present,
to be produced, while in the ‘silent’ b thalassaemia, the deficit resulting in a typical asymptomatic phenotype.
in b chain production is minimal and carriers have minimally The ‘silent’ mutations are normally identified in the
reduced or normal red cell indices and their HbA2 levels are compound heterozygous states with a severe b thalassaemia
normal. allele, which results in thalassaemia intermedia, or in homo-
The point mutations causing b thalassaemia result from zygotes who have a typical phenotype of b thalassaemia trait.
single base substitutions, minor insertions or deletions of a few The ‘silent’ b thalassaemia alleles are not common, except for
bases within the gene or its immediate flanking sequences the )101 C–T, which accounts for a large number of the milder
(Thein, 1998), and may affect any level of genetic regulation. forms of b thalassaemia in the Mediterranean (Maragoudaki
Mutations affecting transcription can either involve the et al, 1999).
conserved DNA sequences that form the b globin promoter The mild b thalassaemia (b++) alleles are associated with
or the stretch of 50 nucleotides in the 5¢-UTR. Generally they clearly defined changes in heterozygotes and result in disorders
result in a mild to minimal deficit of b globin output that of intermediate severity in homozygotes. Interactions with the
reflects the relatively mild phenotype of these b+ thalassaemias. severe alleles are less predictable because of the wider range of
The C–T mutation at position )101 to the b globin gene b globin output, and extend from transfusion dependence to
appears to cause an extremely mild deficit of b globin, such intermediate forms of b thalassaemia at the mild end of the
that it is ‘silent’ in heterozygotes who have normal HbA2 levels spectrum (Camaschella et al, 1995; Ho et al, 1998).
and normal red cell indices (Gonzalez-Redondo et al, 1989; Deletions causing b thalassaemia result in a complete
Maragoudaki et al, 1999). Several mutations in the 5¢-UTR, absence of b globin product and should cause a severe
e.g. CAP + 1A-C, also have a ‘silent’ phenotype (Wong et al, phenotype. However, it is now apparent that these rare b
1987). thalassaemia alleles are associated with unusually high Hb F
Mutations that affect RNA processing can involve either of and HbA2 levels in heterozygotes. These deletions differ widely
the invariant dinucleotides (GT at 5¢ and AG at 3¢) in the splice in size, and apart from the Indian 619 bp deletion, commonly
junction in which case normal splicing is completely abolished remove a region (from positions )125 to +78 relative to the
with the resulting phenotype of b thalassaemia. Mutations mRNA cap site) in the 5¢-b promoter, which includes the
within the consensus sequences at the splice junctions reduce CACCC, CCAAT and TATA elements. The mechanism for
the efficiency of normal splicing to varying degrees and the unusually high levels of HbA2 and Hb F in heterozygotes
produce a b+ phenotype that ranges from mild to severe. for these deletions appears to be related to the removal of
Mutations within introns or exons might also affect the competition for the upstream LCR and limiting transcription
splicing pattern of the pre-mRNAs. For example, a cryptic factors, resulting in increased interaction with the c and d
splice site that contains the sequence GT GGT GAG G has been genes in cis, enhancing their expression. Although the increases

ª 2004 Blackwell Publishing Ltd, British Journal of Haematology, 124, 264–274 267
Review

in Hb F are variable and moderate in heterozygotes, they are of two mutant alleles – as homozygotes or compound
adequate to compensate for the complete absence of b globin heterozygotes – is required to produce clinical disease. Some
in homozygotes for these deletions (Schokker et al, 1966; Craig forms of b thalassaemia, however, are dominantly inherited, in
et al, 1992). This mechanism may also explain the unusually that inheritance of a single b thalassaemia allele in the presence
high HbA2 levels that accompany point mutations affecting the of a normal a globin genotype, results in a clinically detectable
b promoter region. disease (Thein, 1999; Thein, 2001). Heterozygotes have a
Transposable elements may occasionally disrupt human thalassaemia intermedia phenotype with moderate anaemia,
genes and result in their inactivation. The insertion of such an splenomegaly and a thalassaemic blood picture. Apart from the
element, a retrotransposon of the family called L1, has been usual features of heterozygous b thalassaemia, such as
reported with the phenotype of b+ thalassaemia. Despite the increased levels of HbA2 and the imbalanced a/b globin
insertion of 6–7 kb DNA into its IVS2, the affected gene biosynthesis, large inclusion bodies similar to those seen in
expresses full length b globin transcripts at a level corres- thalassaemia major, are often observed in the red cell
ponding to approximately 15% of normal b globin mRNA precursors, hence the original term of ‘inclusion body b
(Divoky et al, 1996). thalassaemia’ (Fei et al, 1989).
The variable severity of the different b thalassaemia alleles is This unusual form of b thalassaemia was probably first
reflected in their phenotypic effect in heterozygotes, in the described in an Irish family in 1973 (Weatherall et al, 1973);
degree of hypochromia and microcytosis as indicated by the several members of the family spanning three generations had
mean cell haemoglobin and mean cell volume (MCV) values a thalassaemia intermedia phenotype that was clearly inherited
respectively. Rund et al (1991) showed that the b thalassaemia as a Mendelian dominant. Since the first description, more
alleles which are associated with the most severe phenotype, than 30 dominantly inherited b thalassaemia alleles have now
demonstrated a fairly tight range of MCVs (63Æ1 fl; SD ¼ 3Æ4) been described (Thein, 1992, 1999); they include missense
while the b+ alleles were associated with a wider range of mutations, minor deletions leading to the loss of intact codons,
MCVs (69Æ3 fl; SD ¼ 5Æ6). The broader range of MCV in b+ frameshifts arising from minor insertions and deletions
thalassaemia when compared with b thalassaemia, is not resulting in elongated b variants with abnormal carboxy
surprising given the broad range in the deficit of b globin terminal ends, and truncated b variants resulting from
production, from barely detectable levels at the severe end, to nonsense mutations. The common denominator of these
moderately reduced (b++) and to just a little less than normal mutations is the predicted synthesis of highly unstable b chain
in the ‘silent’ b thalassaemia alleles. variants, so unstable that in many cases, they are not detectable
A more recent study has taken the correlation between the and only implicated from the DNA sequence. The predicted
severity of b thalassaemia alleles with haematological param- synthesis is supported by the presence of substantial amounts
eters to a finer level. Skarmoutsou et al (2003) measured a of abnormal b mRNA in the peripheral reticulocytes (Hall &
series of haematological parameters, including reticulocyte Thein, 1994), comparable in amounts with that produced from
haemoglobin content (CHr), soluble transferrin receptor the normal b allele. Indeed, the large intra-erythroblastic
(sTfR), reticulocytes and HbA2 and F levels in 57 iron-replete inclusions, which are so characteristic of this form of b
individuals with heterozygous b thalassaemia. sTfR values, a thalassaemia, have subsequently been shown to be composed
reliable quantifiable assessment of the erythropoietic activity, of both a and b globin chains (Ho et al, 1997). In contrast, the
were lowest in the very mild b thalassaemia (bsilent), and inclusion bodies in homozygous b thalassaemia consisted only
highest in b thalassaemia heterozygotes. CHr, a product of of precipitated a globin.
reticulocyte haemoglobin and volume, was lower in bsilent It should be noted that some of these dominantly inherited
thalassaemia compared with normals but the difference was b thalassaemia alleles arise from premature termination
not statistically significant. However, the CHr values between mutations similar to those that are recessively inherited.
the bsilent and the severe groups (b+ and b thalassaemia alleles) How is it that some premature termination mutations cause
was significant, being much higher (27Æ0–32Æ0 pg) in the thalassaemia intermedia while others are clinically asympto-
former, compared with 19Æ5 to 25Æ3 pg in the latter group. matic in the heterozygous state? The answer appears to lie in
Furthermore, while sTfR values showed a positive correlation the differential effects of these in-phase termination mutants
with HbA2, there was a significant negative correlation between on the accumulation of mutant mRNA. The stop codons and
CHr and HbA2 levels. This study confirms that all heterozy- frameshift mutations that are recessively inherited terminate in
gous b thalassaemias have some degree of ineffective erythro- exon 1 or 2, while those that are dominantly inherited,
poiesis that varies with the severity of the b thalassaemia terminate much later in the sequence of the b globin gene, in
mutation. exon 3 or beyond. Premature stop codons near the 3¢ end of
the gene, in exon 3 of the b gene, are less likely to trigger the
Dominantly inherited b thalassaemia. The common b surveillance mechanism of nonsense mediated decay, leading
thalassaemia alleles that are prevalent in the malarious to an accumulation of the mutant b mRNA and to the
regions are inherited typically as Mendelian recessives; synthesis of truncated b chain variants (Maquat, 1995; Hentze
heterozygotes are clinically asymptomatic and the inheritance & Kulozik, 1999). These in-phase termination mutations

268 ª 2004 Blackwell Publishing Ltd, British Journal of Haematology, 124, 264–274
Review

exemplify how shifting the position of a termination codon depends on the severity of the b thalassaemia alleles and the
can alter the phenotype of recessive inheritance caused by number of functional a globin genes (Camaschella et al, 1995;
haplo-insufficiency, to a dominant negative effect because of Ho et al, 1998). Co-inheritance of a single a gene deletion has
the synthesis of an abnormal and deleterious protein. very little effect on the phenotype of b thalassaemia while
The pathophysiology of these b chain variants relates to individuals with two a gene deletions and homozygous b+
their hyperinstability (Thein, 2001). The molecular mecha- thalassaemia may have a mild form of thalassaemia intermedia.
nisms include substitution of the critical amino acids in the At the other extreme, patients who have co-inherited HbH
hydrophobic haem pocket displacing haem, leading to aggre- (equivalent of only one functioning a gene) and homozygous
gation of the globin variant; disruption of secondary structure b thalassaemia, also have thalassaemia intermedia.
because of replacement of critical amino acids; substitution or Just as co-inheritance of a thalassaemia can reduce the
deletion of amino acids involved in ab dimer formation; and clinical severity of homozygous b thalassaemia, the presence of
elongation of subunits by a hydrophobic tail (Bunn & Forget, increased a globin product in b thalassaemia heterozygotes tips
1986). Again, a spectrum in phenotypic severity of this class of the globin chain imbalance further and crosses the threshold,
b thalassaemia variants is observed, which can be related to converting a typically clinical asymptomatic state to that of
variation in the degree of instability of the b globin products thalassaemia intermedia. In the majority of cases, this is related
causing different degrees of ineffective erythropoiesis. to the co-inheritance of triplicated a globin genes. Triplicated a
genes (aaa/) occur in most populations at a low frequency. The
Mosaicism due to somatic deletion of b globin gene. This novel co-inheritance of two extra a globin genes (aaa/aaa) or (aaaa/
mechanism was recently described in an individual who had aa) with heterozygous b thalassaemia results in the thalas-
moderately severe thalassaemia intermedia despite being saemia intermedia (Galanello et al, 1983; Thein et al, 1984).
constitutionally heterozygous for b thalassaemia with a However, the phenotype of a single extra a gene (aaa /aa) with
normal a genotype (Badens et al, 2002). Subsequent heterozygous b thalassaemia is more variable and depends on
investigations revealed that he had a somatic deletion of a the severity of the b thalassaemia allele (Traeger-Synodinos
region of chromosome 11p15 including the b globin complex et al, 1996; Camaschella et al, 1997). There appears to be a
giving rise to a mosaic of cells, 50% with one and 50% without critical threshold of globin chain imbalance in each individual,
any b globin gene. The sum total of the b globin product is above which clinical symptoms appear. This may be related to
approximately 25% less than the normally asymptomatic b the efficiency of the proteolytic mechanism of the erythroid
thalassaemia trait. precursors and/or perhaps to the level of a-haemoglobin-
This unusual case once again illustrates that the severity of stabilizing protein, a chaperone of a globin (Kihm et al, 2002).
anaemia of b thalassaemia reflects the defective b globin chain
production. Furthermore, with respect to potential gene Variation in fetal haemoglobin production. The role of
therapy, expression of a single b globin gene in a proportion increased Hb F response as an ameliorating factor becomes
of the red blood cells appears to be sufficient to redress the evident in the group of homozygous b thalassaemia patients
chain imbalance to produce a condition mild enough not to who are not able to produce any haemoglobin A (a2b2) but yet
need major medical intervention. have a mild disease with a reasonable level of haemoglobin, all
of which is Hb F. Production of fetal haemoglobin after the
neonatal period in b thalassaemia is an extremely complex
Secondary modifiers
process and still poorly understood. There appears to be a
The severity of anaemia in b thalassaemia reflects the degree of genuine increase in c chain synthesis, presumably reflecting the
globin chain imbalance and the excess of a globin chains with expansion of the ineffective erythroid mass. The effect is
all their deleterious effects on the red cell precursors. This augmented by the selective survival of the erythroid precursors
globin chain imbalance can be genetically modified by two that synthesize relatively more c chains. Hence all b
factors – variation in the amount of a globin production and thalassaemias, heterozygous or homozygous, have variable
variation in fetal haemoglobin response. increases in their levels of Hb F. Against this background, there
are undoubtedly genetic factors involved. Studies have shown
a globin genotype. In many populations in which b that approximately 90% of the variation in the level of Hb F
thalassaemia is prevalent, a thalassaemia also occurs at a and F cells (subset of erythrocytes that contain Hb F) is
high frequency and hence it is not uncommon to co-inherit genetically controlled (Garner et al, 2000a). About one-third of
both conditions. Homozygotes or compound heterozygotes for the genetic variance is the result of a common genetic variant,
b thalassaemia who co-inherit a thalassaemia will have less C-T at position )158 of the Gc globin gene, also referred to as
redundant a globin and tend to have a less severe condition. As the Xmn1-Gc polymorphism, but more than 50% of the
with b thalassaemia, the different a thalassaemias that genetic variance in F-cell levels is caused by factors not linked
predominate in different racial groups display a wide range to the b chromosome (Garner et al, 2000b).
of severity. This interaction alone provides the basis for The Xmn1-Gc site is common in all population groups and
considerable clinical heterogeneity; the degree of amelioration is present at a frequency of 0Æ32–0Æ35 (Garner et al, 2000b).

ª 2004 Blackwell Publishing Ltd, British Journal of Haematology, 124, 264–274 269
Review

Unlike the rare mutations in the c globin promoter that are complications have become apparent and evidence suggests
consistently associated with large discrete effects of increased that they may be affected by genetic variants.
Hb F levels of 10–35% in heterozygotes, the so-called Hyperbilirubinaemia and a propensity to gallstone forma-
pancellular HPFH (Wood, 2001), the change at Gc )158 does tion is a common complication of b thalassaemia and is
not always raise the Hb F levels in otherwise normal attributed to the rapid turnover of the red blood cells, bilirubin
individuals. The rare non-deletion HPFH mutations occur in being a breakdown product of haemoglobin. Varying degrees
transcription factor binding motifs in the c promoters, of jaundice have often been observed in the thalassaemia
clustered in three regions, around positions )114 to )117, at syndromes – from thalassaemia trait through to thalassaemia
)175, and from )195 to )202, while the sequence in the )158 major (Galanello et al, 1997; Sampietro et al, 1997; Galanello
region is not a recognized binding motif for any of the known et al, 2001). Studies have shown that the levels of bilirubin and
transcription factors. Nonetheless, although it has little effect the incidence of gallstones in b thalassaemia is related to a
in normal individuals, clinical studies have shown that, under polymorphic variant (seven TA repeats) in the promoter of the
conditions of haemopoietic stress, for example in homozygous uridine diphosphate-glucoronyltransferase IA (UGTIA) gene,
b thalassaemia and sickle cell disease, presence of the Xmn1-Gc also referred to as Gilbert’s syndrome. In vitro studies indicate
site favour a higher Hb F response (Labie et al, 1985; Thein the variant causes a reduced expression of the UGTIA gene
et al, 1987). This could explain why the same mutations on (Bosma et al, 1995). Normal individuals who are homozygous
different b chromosomal backgrounds (some with and others for the [TA]7 variant instead of the usual six, tend to have
without the Xmn1-Gc site) are associated with different higher levels of bilirubin (Bosma et al, 1995). The [TA]7
clinical severity. The increased Hb F output observed in variant has also been shown to be associated with increased
deletions or mutations that involve the promoter sequence of bilirubin levels in sickle cell disease and other haemolytic
the b globin gene reflect the competition between the c and b anaemia (Passon et al, 2001).
globin gene promoters for interaction with the LCR or rate- A common complication of b thalassaemia involves organ
limiting transcription factors. Hence, although such deletions damage from iron overload, not just from blood transfusions
cause a complete absence of b globin product, the severity of but also from increased absorption. While the C282Y mutation
the phenotype is offset by the concomitant increase in Hb F in the HFE1 gene, which causes hereditary haemochromatosis,
(Craig et al, 1992). predisposes to iron loading in thalassaemia intermedia (Rees
Although presence of the cis Xmn1-Gc site is a modulating et al, 1997), the co-existence of b thalassaemia trait aggravates
factor, clearly there are some patients who have enhanced Hb F and accentuates iron loading in C282Y HFE homozygotes
response despite being Xmn1-Gc)/) (Galanello et al, 1989; (Piperno et al, 2000). As the C282Y mutation is rare in
Ho et al, 1998). In many cases, family studies have shown that populations in which b thalassaemia is common it has a
there is an inherent capacity for producing Hb F and that the limited role in iron loading among these patients (Merry-
genetic determinant is not linked to the b globin cluster weather-Clarke et al, 1997). Much more common is the HFE
(Gianni et al, 1983; Thein & Weatherall, 1989). This is in gene polymorphism, H63D, whose functional role is still being
keeping with our genetic studies, which showed that >50% of investigated. Nonetheless, a recent study showed that b
the F-cell variance in the general population is accounted for thalassaemia carriers who are homozygotes for H63D have
by trans-acting factors. Indeed, linkage studies have mapped higher serum ferritin levels than carriers without the poly-
loci controlling Hb F and F-cell levels to three regions of the morphism, suggesting that the H63D polymorphism may have
genome – chromosome 6q23 (Craig et al, 1996), Xp22 (Dover a modulating effect on iron absorption (Melis et al, 2002). As
et al, 1992) and 8q11 (Garner et al, 2002). The effects of the 8q other genes in iron homeostasis become uncovered, it is likely
locus are conditional on the Xmn1-Gc site (Garner et al, there will be genetic variants in these loci that influence the
2002). As the genetic basis of the propensity to produce Hb F different degrees of iron loading in b thalassaemia (Andrews,
becomes unravelled it is becoming clear that the conglomer- 2000, 2002).
ation of the Xmn1-Gc polymorphism, the quantitative trait Progressive osteoporosis and osteopenia is another increas-
loci on 6q, Xp and 8q and others, linked and unlinked to the b ingly common complication encountered in young adults with
globin complex, contribute to the quantitative trait of Hb F b thalassaemia (Wonke, 1998). Several studies suggest that the
that constitute the loosely defined syndrome of heterocellular prevalence of bone disease in b thalassaemia is higher in men
HPFH (Thein & Craig, 1998). Until the different entities than women, and that it is more severe in the spine than
become better defined, detection of an inherent capacity for femoral neck (Jensen et al, 1998; Dresner Pollack et al, 2000;
increased Hb F production is, at present, difficult and usually Perrotta et al, 2000). It is manifested by diffuse bone pain,
inferred from family studies. particularly in the lower back, vertebral fractures, cord
compression, spontaneous fractures and femoral head necrosis.
Bone disease is defined by reduced bone mineral density
Tertiary modifiers
(BMD), as measured by dual-energy X-ray absorptiometry
With the increasing lifespan of the b thalassaemia patients, (DXA). Osteoporosis is present when the BMD is <2Æ5 SD of
subtle variations in the phenotype with regard to some of the the mean value in young adults (T score <)2Æ5), while

270 ª 2004 Blackwell Publishing Ltd, British Journal of Haematology, 124, 264–274
Review

osteopenia is defined by a T score between )1 and )2Æ5. Bone imbalance and the a globin excess. Much of the variation can
mass is determined by a combination of genetic and environ- be explained by heterogeneity of the molecular lesions affecting
mental factors. Anaemia and bone marrow expansion, which is the b globin gene itself but it is also clear that variability at the
prevalent in b thalassaemia, is a major contributor in two loci – a and c globin genes – is important in determining
inadequately treated patients. The osteoporosis that occurs in the phenotype, which is extremely encouraging for genetic
other patients who are reasonably well transfused but in which counselling. However, while genotyping at the b globin and a
there is severe iron loading, may be related to hypogonadism globin loci is relatively easy to incorporate into the prenatal
(Anapliotou et al, 1995). However, it has become apparent diagnosis and counselling programme, detecting an inherent
from recent studies that bone disease is increasingly common ability to increase Hb F in response to haemopoietic stress is
even among the well-transfused and iron-chelated patients still difficult. The presence of such heterocellular HPFH
(Wonke, 1998) and may be related to desferrioxamine toxicity. determinants is usually implicated from studies of family
Studies have suggested that, all things being equal, some members who are often not available. Until the quantitative
patients may be more susceptible to bone disease than others. trait loci for Hb F are better defined, it would appear that it is
Bone mass is another quantitative trait known to be under still not possible to consistently predict phenotype from
strong genetic control involving multiple loci (Pocock et al, genotype apart from the two categories of extra a globin genes
1987; Slemenda et al, 1991; Soroko et al, 1994). The genetic with heterozygous b thalassaemia, and the inheritance of
loci implicated include oestrogen receptor gene, vitamin D mild b+ thalassaemia alleles. Ethnicity and environment are
receptor (VDR), collagen type a1 (COL1A1) and COL1A2 important factors in the analysis of genotype/phenotype
genes, and transforming growth factor b1 (TGFb1). Poly- relationships. Studies have shown that all three categories of
morphism in TGFb1 has been associated with severe osteo- genetic modifiers – primary, secondary and tertiary – are
porosis and increased bone turnover in women (Bertoldo et al, population-specific. The tertiary loci include the many
2000), but a study of TGFb1 polymorphism failed to different genetic polymorphisms that form the background
demonstrate a statistical difference in b thalassaemia patients genes, some of which have been co-selected with the
with different BMDs. The VDR gene polymorphism in intron 8 thalassaemias.
(involving the Bsm1 site) was associated with osteopenia in
3 thalassaemia (Dresner Pollack et al, 2000). A G fi T poly-
Acknowledgments
morphism involving an Sp1 binding site in the COL1A1 gene
was strongly associated with reduced bone mass and osteo- I thank Claire Steward for help in preparation of the
porosis (Grant et al, 1996). This same polymorphism in the manuscript and Dr Barnaby Clark for critical reading of the
COL1A1 gene has also been shown to be strongly associated manuscript.
with reduced BMD and osteoporosis in b thalassaemia
Swee Lay Thein
(Perrotta et al, 2000).
Cardiac complications are the main cause of death in b Department of Haematological Medicine Guy’s, King’s and St Thomas’
thalassaemia. Many aspects of cardiac complications are still School of Medicine, Denmark Hill Campus, Bessemer Road, London SE5
poorly understood, and again it is clear that cardiac disease in 9PJ, UK
b thalassaemia is multifactorial, reflecting the chronic anaemia,
iron overload and pulmonary hypertension. The situation is
References
further confounded by the unpredictability of the severity of
cardiac iron deposition. A risk factor for left ventricular failure Anapliotou, M.L., Kastanias, I.T., Psara, P., Evangelou, E.A., Liparaki,
in thalassaemia is decreased antioxidant activity of apolipo- M. & Dimitriou, P. (1995) The contribution of hypogonadism to the
protein E4, related to the frequency of the apolipoprotein E4 development of osteoporosis in thalassaemia major: new therapeutic
approaches. Clinical Endocrinology, 42, 279–287.
allele (Economou-Peterson et al, 1998).
Andrews, N. (2000) Iron homeostasis: insights from genetics and
Genetic variants implicated in other complications of b
animal models. Nature Reviews Genetics, 1, 208–216.
thalassaemia include specific human leucocyte antigen alleles Andrews, N.C. (2002) A genetic view of iron homeostasis. Seminars in
in the tendency to hepatitis and liver cirrhosis; genetic variants Hematology, 39, 227–234.
in Factor V, prothrombin and methylenetetrahydrofolate Antoniou, M., deBoer, E., Habets, G. & Grosveld, F. (1988) The human
reductase, and the tendency to thrombosis. For a superb b-globin gene contains multiple regulatory regions: identification of
review of the hypercoagulable state in thalassaemia and the one promoter and two downstream enhancers. EMBO Journal, 7,
possible underlying genetic and environmental contribution, 377–384.
the reader should consult Eldor and Rachmilewitz (2002). Badens, C., Mattei, M.G., Imbert, A.M., Lapoumérouliee, C., Martini,
N., Michel, G. & Lena-Russo, D. (2002) A novel mechanism for
thalassaemia intermedia. The Lancet, 359, 132–133.
Conclusion Bertoldo, F., D’Agruma, L., Furlan, F., Colapietro, F., Lorenzi, M.T.,
Maiorano, N., Iolascon, A., Zelante, L., Locascio, V. & Gasparini, P.
There is a spectrum of phenotypes in b thalassaemia, the
(2000) Transforming growth factor-beta1 gene polymorphism, bone
severity of which relates directly to the degree of chain

ª 2004 Blackwell Publishing Ltd, British Journal of Haematology, 124, 264–274 271
Review

turnover, and bone mass in Italian postmenopausal women. Journal for left ventricular failure in homozygous beta-thalassemia. Blood,
of Bone and Mineral Research, 15, 634–639. 92, 3455–3459.
Bosma, P.J., Chowdhury, J.R., Bakker, C., Gantla, S., De Boer, A., Efstratiadis, A., Posakony, J.W., Maniatis, T., Lawn, R.M., O’Connell,
Oostra, B.A., Lindhout, D., Tytgat, G.N.J., Jansen, P.L.M., Elferink, C., Spritz, R.A., DeRiel, J.K., Forget, B.G., Weissman, S.M., Sligh-
R.P.J.O. & Chowdhury, N.R. (1995) The genetic basis of the tom, J.L., Blechl, A.E., Smithies, O., Baralle, F.E., Shoulders, C.C. &
reduced expression of bilirubin UCP-glucuronosultransferase 1 in Proudfoot, N.J. (1980) The structure and evolution of the human
Gilbert’s syndrome. New England Journal of Medicine, 333, 1171– beta-globin gene family. Cell, 21, 653–668.
1175. Eldor, A. & Rachmilewitz, E.A. (2002) The hypercoagulable state in
Boyer, S.H., Belding, T.K., Margolet, L. & Noyes, A.N. (1975) Fetal thalassemia. Blood, 99, 36–43.
hemoglobin restriction to a few erythrocytes (F cells) in normal Fei, Y.J., Stoming, T.A., Kutlar, A., Huisman, T.H.J. & Stama-
human adults. Science, 188, 361–363. toyannopoulos, G. (1989) One form of inclusion body ß-thalassemia
Bulger, M., Bender, M.A., van Doorninck, J.H., Wertman, B., Farrell, is due to a GAA->TAA mutation at codon 121 of the ß chain. Blood,
C.M., Felsenfeld, G., Groudine, M. & Hardison, R. (2000) Com- 73, 1075–1077.
parative structural and functional analysis of the olfactory receptor Flint, J., Harding, R.M., Boyce, A.J. & Clegg, J.B. (1998) The popula-
genes flanking the human and mouse beta-globin gene clusters. tion genetics of the haemoglobinopathies. In: Bailliére’s Clinical
PNAS, 97, 14560–14565. Haematology (ed. by G.P. Rodgers), Vol. 11, pp. 1–52. Bailliere
Bunn, H.F. & Forget, B.G. (1986) Hemoglobin: Molecular, Genetic and Tindall, London.
Clinical Aspects. W.B. Saunders Company, Philadelphia, PA. Galanello, R., Ruggeri, R., Paglietti, E., Addis, M., Melis, M.A. & Cao,
Camaschella, C., Maza, U., Roetto, A., Gottardi, E., Parziale, A., Travi, A. (1983) A family with segregating triplicated alpha globin loci and
M., Fattore, S., Bacchiega, D., Fiorelli, G. & Cappellini, M.D. (1995) ß thalassaemia. Blood, 62, 1035–1040.
Genetic interactions in thalassemia intermedia: analysis of beta- Galanello, R., Dessi, E., Melis, M.A., Addis, M., Sanna, M.A., Rosatelli,
mutations, alpha-genotype, gamma-promoters, and beta-LCR C., Argiolu, F., Giagu, N., Turco, M.P., Cacace, E., Pirastu, M. &
hypersensitive sites 2 and 4 in Italian patients. American Journal of Cao, A. (1989) Molecular analysis of beta zero-thalassemia inter-
Hematology, 48, 82–87. media in Sardinia. Blood, 74, 823–827.
Camaschella, C., Kattamis, A.C., Petroni, D., Roetto, A., Sivera, P., Galanello, R., Perseu, L., Melis, M.A., Cipollina, L., Barella, S., Giagu,
Sbaiz, L., Cohen, A., Ohene-Frempong, K., Trifillis, P., Surrey, S. & N., Turco, M.P., Maccioni, O. & Cao, A. (1997) Hyper-
Fortina, P. (1997) Different hematological phenotypes caused by the bilirubinaemia in heterozygous beta-thalassaemia is related to
interaction of triplicated alpha-globin genes and heterozygous beta- co-inherited Gilbert’s syndrome. British Journal of Haematology, 99,
thalassemia. American Journal of Hematology, 55, 83–88. 433–436.
Carter, D., Chakalova, L., Osborne, C.S., Dai, Y.F. & Fraser, P. (2002) Galanello, R., Piras, S., Barella, S., Leoni, G.B., Cipollina, M.D., Perseu,
Long-range chromatin regulatory interactions in vivo. Nature L. & Cao, A. (2001) Cholelithiasis and Gilbert’s syndrome in
Genetics, 32, 623–626. homozygous beta-thalassaemia. British Journal of Haematology, 115,
Collins, F.S. & Weissman, S.M. (1984) The molecular Genetics of 926–928.
human hemoglobin. Progress in Nucleic Acid Research and Molecular Garner, C., Tatu, T., Reittie, J.E., Littlewood, T., Darley, J., Cervino, S.,
Biology, 31, 315–465. Farrall, M., Kelly, P., Spector, T.D. & Thein, S.L. (2000a) Genetic
Craig, J.E., Kelly, S.J., Barnetson, R. & Thein, S.L. (1992) Molecular influences on F cells and other hematological variables: a twin
characterization of a novel 10.3 kb deletion causing beta-thalas- heritability study. Blood, 95, 342–346.
saemia with unusually high Hb A2. British Journal of Haematology, Garner, C., Tatu, T., Game, L., Cardon, L.R., Spector, T.D., Farrall, M.
82, 735–744. & Thein, S.L. (2000b) A candidate gene study of F cell levels in
Craig, J.E., Rochette, J., Fisher, C.A., Weatherall, D.J., Marc, S., sibling pairs using a joint linkage and association analysis. GeneSc-
Lathrop, G.M., Demenais, F. & Thein, S.L. (1996) Dissecting the loci reen, 1, 9–14.
controlling fetal haemoglobin production on chromosomes 11p and Garner, C.P., Tatu, T., Best, S., Creary, L. & Thein, S.L. (2002) Evi-
6q by the regressive approach. Nature Genetics, 12, 58–64. dence for Genetic Interaction between the beta-globin complex and
Divoky, V., Indrak, K., Mrug, M., Brabec, V., Huisman, T.H.J. & chromosome 8q in the expression of fetal hemoglobin. American
Prchal, J.T. (1996) A novel mechanism of ß thalassemia: The Journal of Human Genetics, 70, 793–799.
insertion of L1 retrotransposable element into ß globin IVS II. Blood, Gianni, A.M., Bregni, M., Cappellini, M.D., Fiorelli, G., Taramelli, R.,
88, 148a. Giglioni, B., Comi, P. & Ottolenghi, S. (1983) A gene controlling
Dover, G.J., Smith, K.D., Chang, Y.C., Purvis, S., Mays, A., Meyers, fetal hemoglobin expression in adults is not linked to the non-alpha
D.A., Sheils, C. & Serjeant, G. (1992) Fetal hemoglobin levels in globin cluster. EMBO Journal, 2, 921–925.
sickle cell disease and normal individuals are partially controlled by Gonzalez-Redondo, J.M., Stoming, T.A., Kutlar, A., Kutlar, F., Lanclos,
an X-linked gene located at Xp22.2. Blood, 80, 816–824. K.D., Howard, E.F., Fei, Y.J., Aksoy, M., Altay, C., Gurgey, A., Basak,
Dresner Pollack, R., Rachmilewitz, E., Blumenfeld, A., Idelson, M. & A.N., Efremov, G.D., Petkov, G. & Huisman, T.H.J. (1989) A
Goldfarb, A.W. (2000) Bone mineral metabolism in adults with C fi T substitution at nt-101 in a conserved DNA sequence of the
beta-thalassaemia major and intermedia. British Journal of Haema- promoter region of the ß-globin gene is associated with ‘silent’ ß-
tology, 111, 902–907. thalassemia. Blood, 73, 1705–1711.
Economou-Peterson, E., Aesspopos, A., Kladi, A., Flevari, P., Kar- Grant, S.F., Reid, D.M., Blake, G., Herd, R., Fogelman, I. & Ralston,
abatsos, F., Fragodimitri, C., Nicolaidis, P., Vrettou, H., Vassilo- S.H. (1996) Reduced bone density and osteoporosis associated with
poulos, D., Karagiorga-Lagana, M., kremastinos, D.T. & Petersen, a polymorphic Sp1 binding site in the collagen type I alpha 1 gene.
M.B. (1998) Apolipoprotein E epsilon4 allele as a genetic risk factor Nature Genetics, 14, 203–205.

272 ª 2004 Blackwell Publishing Ltd, British Journal of Haematology, 124, 264–274
Review

Hall, G.W. & Thein, S.L. (1994) Nonsense codon mutations in the Pocock, N.A., Eisman, J.A., Hopper, J.L., Yeates, M.G., Sambrook, P.N.
terminal exon of the ß-globin gene are not associated with a re- & Eberl, S. (1987) Genetic determinants of bone mass in adults. A
duction in ß-mRNA accumulation: a mechanism for the phenotype twin study. Journal of Clinical Investigation, 80, 706–710.
of dominant ß-thalassemia. Blood, 83, 2031–2037. Rees, D.C., Luo, L.Y., Thein, S.L., Sing, B.M. & Wickramasinghe, S.
Hentze, M.W. & Kulozik, A.E. (1999) A perfect message: RNA sur- (1997) Nontransfusional iron overload in thalassemia: Association
veillance and nonsense-mediated decay. Cell, 96, 307–310. with hereditary hemochromatosis. Blood, 90, 3234–3236.
Ho, P.J., Wickramasinghe, S.N., Rees, D.C., Lee, M.J., Eden, A. & Rund, D., Filon, D., Strauss, N., Rachmilewitz, E.A. & Oppenheim, A.
Thein, S.L. (1997) Erythroblastic inclusions in dominantly inherited (1991) Mean corpuscular volume of heterozygotes for ß-thalassemia
ß thalassaemias. Blood, 89, 322–328. correlates with the severity of mutations. Blood, 79, 238–243.
Ho, P.J., Hall, G.W., Luo, L.Y., Weatherall, D.J. & Thein, S.L. (1998) Sampietro, M., Lupica, L., Perrero, L., Comino, A. & Martinez di
Beta thalassemia intermedia: is it possible to consistently predict Montemuros, F. (1997) The expression of uridine diphosphate
phenotype from genotype? British Journal of Haematology, 100, 70– glucuronosyltransferase gene is a major determinant of bilirubin
78. level in heterozygous beta-thalassaemia and in glucose-6-phosphate.
Huisman, T.H.J. (1997) Levels of Hb A2 in heterozygotes and homo- British Journal of Haematology, 99, 437–439.
zygotes for beta-thalassemia mutations: Influence of mutations in Schokker, R.C., Went, L.N. & Bok, J. (1966) A new genetic variant of
the CACCC and ATAAA motifs of the beta-globin gene promoter. ß-thalassaemia. Nature, 209, 44–46.
Acta Haematologica, 98, 187–194. Schrier, S.L. (2002) Pathophysiology of thalassemia. Current Opinion
Jensen, C.E., Tuck, S.M., Agnew, J.E., Koneru, S., Morris, R.W., in Hematology, 9, 123–126.
Yardumian, A., Prescott, E., Hoffbrand, A.V. & Wonke, B. (1998) Skarmoutsou, C., Papassotiriou, I., Traeger-Synodinos, J., Stamou, H.,
High prevalence of low bone mass in thalassaemia major. British Ladis, V., Metaxotou-Mavrommati, A., Stamoulakatou, A. & Ka-
Journal of Haematology, 103, 911–915. navakis, E. (2003) Erythroid bone marrow activity and red cell he-
Kihm, A.J., Kong, Y., Hong, W., Russell, J.E., Rouda, S., Adachi, K., moglobinization in iron sufficient beta-thalassemia heterozygotes as
Simon, M.C., Blobel, G.A. & Weiss, M.J. (2002) An abundant ery- reflected by soluble transferrin receptor and reticulocyte hemoglobin
throid protein that stabilizes free alpha-haemoglobin. Nature, 417, in content. Correlation with genotypes and Hb A2 levels. Haema-
758–763. tologica, 88, 631–636.
Labie, D., Pagnier, J., Lapoumeroulie, C., Rouabhi, F., Dunda-Bel- Slemenda, C.W., Christian, J.C., Williams, C.J., Norton, J.A. & John-
khodja, O., Chardin, P., Beldjord, C., Wajcman, H., Fabry, M.E. ston Jr, C.C. (1991) Genetic determinants of bone mass in adult
& Nagel, R.L. (1985) Common haplotype dependency of high G women: a re-evaluation of the twin model and the potential im-
gamma-globin gene expression and high Hb F levels in beta- portance of gene interaction on heritability estimates. Journal of
thalassemia and sickle cell anemia patients. Proceedings of the National Bone and Mineral Research, 6, 561–567.
Academy of Sciences of the United States of America, 82, 2111–2114. Soroko, S.B., Barrett-Connor, E., Edelstein, S.L. & Kritz-Silverstein, D.
Maquat, L.E. (1995) When cells stop making sense: effects of nonsense (1994) Family history of osteoporosis and bone mineral density at
codons on RNA metabolism in vertebrate cells. RNA, 1, 453–465. the axial skeleton: the Rancho Bernardo Study. Journal of Bone and
Maragoudaki, E., Kanavakis, E., Trager-Synodinos, J., Vrettou, C., Mineral Research, 9, 761–769.
Tzetis, M., Metxotou-Mavrommati, A. & Kattamis, C. (1999) Mo- Thein, S.L. (1992) Dominant beta thalassaemia: molecular basis and
lecular, haematological and clinical studies of the -101 C fi T pathophysiology. British Journal of Haematology, 80, 273–277.
substitution in the b-globin gene promoter in 25 b-thalassaemia Thein, S.L. (1998) Beta thalassaemia. In: Baillière’s Clinical Haema-
intermedia patients and 45 heterozygotes. British Journal of Hae- tology, Sickle Cell Disease and Thalassaemia (ed. by G.P. Rodgers),
matology, 107, 699–706. Vol. 11:1, pp. 91–126. Baillière Tindall, London.
Melis, M.A., Cau, M., Deidda, F., Barella, S., Cao, A. & Galanello, R. Thein, S.L. (1999) Is it dominantly inherited beta thalassaemia or just a
(2002) H63D mutation in the HFE gene increases iron overload in beta-chain variant that is highly unstable? British Journal of Hae-
beta-thalassemia carriers. Haematologica, 87, 242–245. matology, 107, 12–21.
Merryweather-Clarke, A.T., Pointon, J.J., Shearman, J.D. & Robson, Thein, S.L. (2001) Structural variants with a beta-thalassemia pheno-
K.J.H. (1997) Global prevalence of putative haemochromatosis type. In: Disorders of Hemoglobin: Genetics, Pathophysiology, and
mutations. Journal of Medical Genetics, 34, 275–278. Clinical Management (ed. by M.H. Steinberg, B.G. Forget, D.R.
Passon, R.G., Howard, T.A., Zimmerman, S.A., Schultz, W.H. & Ware, Higgs & R.L. Nagel), pp. 342–355. Cambridge University Press,
R.E. (2001) Influence of bilirubin uridine diphosphate-glucur- Cambridge, UK.
onosyltransferase 1A promoter polymorphisms on serum bilirubin Thein, S.L. & Craig, J.E. (1998) Genetics of Hb F/F cell variance in
levels and cholelithiasis in children with sickle cell anemia. American adults and heterocellular hereditary persistence of fetal hemoglobin.
Journal of Pediatric Hematology and Oncology, 23, 448–451. Hemoglobin, 22, 401–414.
Perrotta, S., Cappellini, M.D., Bertoldo, F., Servedio, V., Iolascon, G., Thein, S.L. & Weatherall, D.J. (1989) A non-deletion hereditary per-
D’Agruma, L., Gasparini, P., Siciliani, M.C. & Iolascon, A. (2000) sistance of fetal hemoglobin (HPFH) determinant not linked to the
Osteoporosis in beta-thalassaemia major patients: analysis of the beta-globin gene complex. In: Hemoglobin Switching, Part B: Cellular
genetic background. British Journal of Haematology, 111, 461–466. and Molecular Mechanisms (ed. by G. Stamatoyannopoulos & A.W.
Piperno, A., Mariani, R., Arosio, C., Vergani, A., Bosio, S., Fargion, S., Nienhuis), pp. 97–111. Alan R Liss, Inc., New York, NY.
Sampietro, M., Girelli, D., Fraquelli, M., Conte, D., Fiorelli, G. & Thein, S.L., Al-Hakim, I. & Hoffbrand, A.V. (1984) Thalassaemia
Camaschella, C. (2000) Haemochromatosis in patients with beta- intermedia: a new molecular basis. British Journal of Haematology,
thalassaemia trait. British Journal of Haematology, 111, 908–914. 56, 333–337.

ª 2004 Blackwell Publishing Ltd, British Journal of Haematology, 124, 264–274 273
Review

Thein, S.L., Wainscoat, J.S., Sampietro, M., Old, J.M., Cappellini, D., Weatherall, D.J., Clegg, J.B., Knox-Macaulay, H.H.M., Bunch, C.,
Fiorelli, G., Modell, B. & Weatherall, D.J. (1987) Association of Hopkins, C.R. & Temperley, I.J. (1973) A genetically determined
thalassaemia intermedia with a beta-globin gene haplotype. British disorder with features both of thalassaemia and congenital dysery-
Journal of Haematology, 65, 367–373. thropoietic anaemia. British Journal of Haematology, 24, 681–702.
Tolhuis, B., Palstra, R.J., Splinter, E., Grosveld, F. & de Laat, W. (2002) Wong, C., Dowling, C.E., Saiki, R.K., Higuchi, R.G., Erlich, H.A. &
Looping and interaction between hypersensitive sites in the active Kazazian, H.H.J. (1987) Characterization of beta-thalassaemia
beta-globin locus. Molecular Cell, 10, 1453–1465. mutations using direct genomic sequencing of amplified single copy
Traeger-Synodinos, J., Kanavakis, E., Vrettou, C., Maragoudaki, E., DNA. Nature, 330, 384–386.
Michael, T. & Metaxotou-Mavromati, A. (1996) The triplicated al- Wonke, B. (1998) Bone disease in beta-thalassaemia major. British
pha-globin gene locus in beta-thalassaemia heterozygotes: clinical, Journal of Haematology, 103, 897–901.
haematological, biosynthetic and molecular studies. British Journal Wood, W.G. (2001) Hereditary persistence of fetal hemoglobin and
of Haematology, 95, 467–471. delta beta thalassemia. In: Disorders of Hemoglobin: Genetics, Path-
Trudel, M. & Costantini, F. (1987) A 3¢enhancer contributes to the ophysiology, and Clinical Management (ed. by M.H. Steinberg, B.G.
stage-specific expression of the human b-globin gene. Genes and Forget, D.R. Higgs & R.L. Nagel), pp. 356–388. Cambridge Uni-
Development, 1, 954–961. versity Press, Cambridge, UK.
Viprakasit, V., Gibbons, R.J., Broughton, B.C., Tolmie, J.L., Brown, D., Yu, C., Niakan, K.K., Matsushita, M., Stamatoyannopoulos, G., Orkin,
Lunt, P., Winter, R.M., Marinoni, S., Stefanini, M., Brueton, L., S.H. & Raskind, W.H. (2002) X-linked thrombocytopenia with
Lehmann, A.R. & Higgs, D.R. (2001) Mutations in the general thalassemia from a mutation in the amino finger of GATA-1 af-
transcription factor TFIIH result in beta-thalassaemia in individuals fecting DNA binding rather than FOG-1 interaction. Blood, 100,
with trichothiodystrophy. Human Molecular Genetics, 10, 2797–2802. 2040–2045.
Weatherall, D.J. & Clegg, J.B. (2001) The Thalassaemia Syndromes, 4th
edn. Blackwell Science, Oxford. Keywords: genotype-phenotype correlation, b thalassaemia.

274 ª 2004 Blackwell Publishing Ltd, British Journal of Haematology, 124, 264–274

S-ar putea să vă placă și