genetic basis of blood group diversity - storry - 2004 - british journal of haematology - wiley...

Upload: hasna-f-bidayah

Post on 02-Jun-2018

220 views

Category:

Documents


0 download

TRANSCRIPT

  • 8/9/2019 Genetic Basis of Blood Group Diversity - Storry - 2004 - British Journal of Haematology - Wiley Online Library

    1/13

    Genetic basis of blood group diversity

    Jill R. Storry and Martin L. Olsson

    Blood Centre, University Hospital and Department of Transfusion Medicine, Institute of Laboratory Medicine, Lund, Sweden

    Summary

    In the last 18 years the genes that encode all but one of the 29

    blood group systems present on red blood cells (RBCs) have

    been identified. This body of knowledge has permitted the

    application of molecular techniques to characterize the com-

    mon blood group antigens and to elucidate the background for

    some of the variant phenotypes. Just as the RBC was used as a

    model for the biochemical characterization of cell membranes,

    so the genes encoding blood groups provide a readily accessible

    model for the study of gene expression and diversity. The

    application of genotyping techniques to identify fetuses at risk

    of haemolytic disease of the newborn is now the standard of

    care, and the expansion of nucleic acid testing platforms to

    include both disease testing and blood typing in the blood

    centre is on the horizon. This review summarizes the

    molecular basis of blood groups and illustrates the mechanisms

    that generate diversity through specific examples.

    Keywords: blood group, allele, molecular techniques, geno-

    typing, genetic diversity.

    For well over a century, blood group antigens have been

    recognized as differences between the red blood cells (RBCs) of

    one person and another. Antigens have been defined by human

    antibodies, immune and naturally occurring, as well as those

    deliberately stimulated in animals. Assignment of blood group

    antigen status requires the novel factor to be inherited from

    one generation to the next, thus demonstrating that blood

    group antigens were the products of genes. Many of the

    cellular components that carry blood group antigens have been

    identified and characterized and indeed, the RBC has provided

    a useful model for the study of cell membranes. Concurrently,

    the genes encoding the blood group proteins have been

    mapped to different chromosomes throughout the genome.

    The development of DNA sequencing techniques, and then the

    polymerase chain reaction (PCR) has paved the way for the

    rapid molecular characterization of the genes encoding blood

    group antigens, such that, in the last 18 years, all but one of

    the 29 blood group genes have been characterized. This

    knowledge, combined with continual improvements in gene

    analysis are changing the way in which testing can be

    performed.

    Blood group antigens are part of carbohydrate or protein

    structures exposed on the extracellular surface of the RBC

    membrane. In blood group nomenclature, antigens encoded by

    the same gene, or cluster of genes, are assigned to the same

    blood group system. Each system may consist of one or more

    antigens. Proteins that are glycosylated by N-linked glycans,

    e.g. band 3 also carry carbohydrate antigens, like ABH and I

    antigens. Currently, 29 blood group systems, which include a

    total of 240 antigens, have been established by the Interna-

    tional Society of Blood Transfusion (ISBT) Committee on

    Terminology for Red Cell Surface Antigens. In addition, 38

    antigens not yet fulfilling the requirements for classification

    into a system have been gathered in collections or series of

    high- and low-frequency antigens (Daniels et al, 2003). These

    numbers are not static and new blood group antigens are

    identified by unusual serological findings each year.

    This review will discuss the genes and polymorphisms

    underlying the expression of human blood group systems. A

    comprehensive summary of information detailing the genesand carrier molecule(s) for each blood group system has been

    collated in Table I and the single nucleotide polymorphisms

    (SNPs) associated with some clinically important antigens are

    shown in Table II.

    References describing the cloning of the genes and the

    identification of the specific polymorphisms are given in the

    appropriate table. In addition, a selection of relevant references

    is given throughout the text, mostly the ones that serve as

    illustrative examples of different genetic principles. Additional

    papers describing the elucidation of the polymorphisms

    responsible for the blood group antigens within each system

    can be retrieved via the GenBank accession numbers given in

    Table I and at the Blood Group Antigen Database (http://

    www.bioc.aecom.yu.edu/bgmut/index.htm). The interested

    reader is also referred to current textbooks and reviews, a

    few examples of which can be found in the reference list (Issitt

    & Anstee, 1998; Reid & Yazdanbakhsh, 1998; Daniels, 2002;

    Reid & Lomas-Francis, 2003).

    In general, antigens belonging to blood group systems are

    better characterized at the molecular level than those antigens

    assigned to a collection or series. In most instances, the paucity

    of genetic data regarding the latter antigens prevents assign-

    Correspondence: Martin L. Olsson, Blood Centre, University Hospital

    and Department of Transfusion Medicine, Institute of Laboratory

    Medicine, Lund, Sweden. E-mail: [email protected]

    review

    2004 Blackwell Publishing Ltd, British Journal of Haematology, 126, 759771 doi:10.1111/j.1365-2141.2004.05065.x

  • 8/9/2019 Genetic Basis of Blood Group Diversity - Storry - 2004 - British Journal of Haematology - Wiley Online Library

    2/13

    Table

    I.

    Summaryofinformationon

    genesandgeneproductsinthecurrentlyacknowledgedbloodgroupsystems.

    ISBT

    Name

    ISBT

    number

    ISBT

    system

    symbol

    Ch

    romosome

    loc

    ation

    ISGN

    gene

    symbol

    Genbank

    account

    number*

    Gene

    product

    Protein

    type

    Apparent

    mass

    (kDa)

    Number

    ofamino

    acids

    CD

    number

    Numberof

    antigens

    Cloni

    ngreference

    ABO

    1

    ABO

    9q342

    ABO

    NM_

    020469

    a3GalNAcT,a3GalT

    II

    n.a.

    354

    4

    Yama

    motoetal(1990b))

    MNS

    2

    MNS

    4q282-q311

    GYPA

    NM_

    002099

    Glyco

    phorinA

    I

    36

    131

    CD235a

    43

    SiebertandFukuda(1986)

    GYPB

    NM_

    002100

    Glyco

    phorinB

    I

    24

    72

    CD235b

    SiebertandFukuda(1987)

    P

    3

    P1

    22q112-ter

    P1

    a4GalT?

    -

    n.a.

    1

    Rh

    4

    RH

    1p3613-p343

    RHD

    RHCE

    NM_

    016225

    NM_

    138618

    RhD

    RhCE

    M-12

    M-12

    3032

    417

    417

    CD240D

    CD240CE

    48

    Aventetal(1990),

    Cheri

    f-Zaharetal(1990),

    LeVa

    nKimetal(1992),

    Arceetal(1993),

    Kajii

    etal(1993)

    Lutheran

    5

    LU

    19q132

    LU

    NM_

    005581

    Luthe

    ranglycoprotein(IgSF)

    I

    7885

    597

    CD239

    19

    Parso

    nsetal(1995)

    B-CA

    M

    (IgSF)

    I

    557

    Kell

    6

    KEL

    7q33

    KEL

    NM_

    000420

    Kellg

    lycoprotein

    II

    93

    732

    CD258

    24

    Leeetal(1991)

    Lewis

    7

    LE

    19p133

    FUT3

    NM_

    000149

    a3/4F

    ucT

    II

    n.a.

    361

    6

    Kukowska-Latalloetal(1990)

    Duffy

    8

    FY

    1q21-q25

    DARC

    NM_

    002036

    DARC,

    Duffyglycoprotein

    M-7

    3545

    338

    CD234

    6

    Chaudhurietal(1993)

    Kidd

    9

    JK

    18q11-q12

    SLC14A1

    NM_

    015865

    HUT,

    Kiddglycoprotein

    M-10

    50

    389

    3

    Olivesetal(1994)

    Diego

    10

    DI

    17q12-q21

    SLC4A1

    NM_

    000342

    AE-1,

    Band3

    M-14

    90

    911

    CD233

    21

    Tanneretal(1988),

    Luxe

    tal(1989)

    Yt

    11

    YT

    7q22

    ACHE

    NM_

    015831

    Acety

    lcholinesterase

    GPI

    160

    557

    2

    Lietal(1991)

    Xg

    12

    XG

    Xp

    2232

    XG

    NM_

    175569

    Xgglycoprotein

    I

    2229

    180

    CD99

    2

    Darlingetal(1986),

    Ellisetal(1994)

    Scianna

    13

    SC

    1p34

    ERMAP

    NM_

    018538

    ERMAP(IgSF)

    I

    60

    475

    4

    Yeet

    al(2000)

    Dombrock

    14

    DO

    12p132-p121

    ART4

    NM_

    021071

    ADP-

    ribosyltransferase4

    GPI

    5457

    314

    5

    Gubinetal(2000)

    Dombrockglycoprotein

    Colton

    15

    CO

    7p14

    AQP1

    NM_

    000385

    CHIP

    ,Aquaporin-1

    M-6

    28or50

    269

    3

    PrestonandAgre(1991)

    Landsteiner-

    Wiener

    16

    LW

    19p132-cen

    ICAM4

    NM_

    022377

    ICAM

    -4

    LWg

    lycoprotein(IgSF)

    I

    42

    241

    CD242

    3

    Bailly

    etal(1994)

    Chido-

    Rodgers

    17

    CH/RG

    6p213

    C4A

    C4B

    NM_

    007293

    NM_

    000592

    Complementfactor4A

    Complementfactor4B

    S S

    n.a.

    1741

    1744

    9

    Yuet

    al(1986),

    Yu(1991)

    Hh

    18

    H

    19q133

    FUT1

    NM_

    000148

    a2FucT

    II

    n.a.

    365

    CD173

    1

    Kelly

    etal(1994)

    Kx

    19

    XK

    Xp

    211

    XK

    NM_

    021083

    Xkglycoprotein

    M-10

    37

    444

    1

    Hoetal(1994)

    Gerbich

    20

    GE

    2q14-q21

    GYPC

    NM_

    002101

    GPC

    I

    32

    128

    CD236R

    7

    Colin

    etal(1986)

    GPD

    I

    23

    107

    CD236

    Cromer

    21

    CROM

    1q32

    DAF

    NM_

    000574

    DAF

    GPI

    70

    347

    CD55

    12

    Caras

    etal(1987),

    Medo

    fetal(1987)

    Knops

    22

    KN

    1q32

    CR1

    NM_

    000573

    CR1

    I

    170280

    1998

    CD35

    8

    Wongetal(1989)

    Indian

    23

    IN

    11p13

    CD44

    NM_

    000610

    Herm

    esantigen

    I

    80

    341

    CD44

    2

    GoldsteinandButcher(1990),

    Harn

    etal(1991)

    Review

    760 2004 Blackwell Publishing Ltd, British Journal of Haematology, 126, 759771

  • 8/9/2019 Genetic Basis of Blood Group Diversity - Storry - 2004 - British Journal of Haematology - Wiley Online Library

    3/13

    Table

    1.

    (continued)

    ISBT

    Name

    ISBT

    number

    ISBT

    system

    symbol

    Ch

    romosome

    loc

    ation

    ISGN

    gene

    symbol

    Genbank

    account

    number*

    Geneproduct

    Protein

    type

    Apparent

    mass

    (kDa

    )

    Number

    ofamino

    acids

    CD

    number

    Numberof

    antigens

    Cloningreference

    Ok

    24

    OK

    19p133

    BSG

    NM_

    001728

    Neurothelin,

    basigin

    I

    356

    9

    248

    CD147

    1

    Guoetal(1998)

    Raph

    25

    RAPH

    11p155

    MER2

    NM_

    004357

    MER2

    M-4

    40

    253

    CD151

    1

    Hasegawaetal(1996)

    JMH

    26

    JMH

    15q223-q23

    SEMA7A

    NM_

    003612

    H-Sema-L

    GPI

    758

    0

    656

    CD108

    1

    Yamadaetal(1999)

    I

    27

    I

    6p24

    GCNT2

    NM_

    145649

    b6GlcNAcT

    II

    n.a.

    400

    1

    Bierhuizenetal(1993)

    Globoside

    28

    GLOB

    3q25

    B3GALT3**

    NM_

    033169

    b3GalNAcT1,Psynthase

    II

    n.a.

    331

    1

    Amadoetal(1998)

    GIL

    29

    GIL

    9p13

    AQP3

    NM_

    004925

    Aquaporin-3

    M-6

    45

    292

    1

    Inaseetal(1995)

    *SeveraldifferentGenBankentriesmay

    existforeachsystem;n.a.,notapplicable.Mostaccessionnumbersgivenwereretrievedfrom

    http://www.gene.ucl.ac.uk/cgi-bin/nomenclature/searchgenes.p

    linJuly

    2003.

    TypeIandIIaresinglemembranepa

    ssmoleculeswiththeiramino-orcarboxyterm

    inalsoutsidethecell(insidetheGolgiforgly

    cosyltransferases)respectively.

    M-nisamultimembranepassmolecule

    thattraversesthemembranentimes;G

    PIisamoleculeanchoredtotheRBCmemb

    raneviaaglycosylphosphatidylinositollink;S

    isamoleculethatisfoundinitssolubleform

    inplasmabuthasbeen

    adsorbedtotheRBCmembraneandc

    ovalentlyboundtolysineresidues.

    Insomeinstancesthenumberofaminoacidsgivenmayvarybetweendifferentva

    riants/formsofthemolecule.

    Whilsttheprimarygeneproductistheglycosyltransferasegiven,thebloodgroup

    antigensarecarriedbycarbohydratestructuresonglycoproteinsand/orglycolipids.

    ThesizegivencorrespondstothefulllengthofC4.ObservethattheC4dfragmen

    tonlyisadsorbedandboundtotheRBCandcarriestheCH/RGantigens.

    **Currentgenenamebasedontheerr

    oneousassignmentofthegeneproductasab3-galactosyltransferase.

    Review

    2004 Blackwell Publishing Ltd, British Journal of Haematology, 126, 759771 761

  • 8/9/2019 Genetic Basis of Blood Group Diversity - Storry - 2004 - British Journal of Haematology - Wiley Online Library

    4/13

    ment of these antigens to a specific blood group system.

    However, there are exceptions in both cases. For example, the

    gene responsible for expression of P1 antigen in the P blood

    group system has not been identified. Despite this, it has been

    acknowledged as a system of its own since it was declared

    independent of all other blood group systems. The opposite is

    true for the Pk blood group antigen, currently residing in

    collection 209. The locus responsible for Pk blood group

    antigen expression was unambiguously shown to be a 4-a-

    galactosyltransferase gene (A4GALT) on the long arm of

    chromosome 22 (Steffensen et al, 2000). However, since its

    relation to the P blood group system (also coded for by a locus

    on the long arm of chromosome 22 according to family

    studies) is unclear, it remains in the GLOB collection until this

    issue has been resolved.

    Molecular mechanisms that generate bloodgroup diversity

    Diversity in the human genome arises through a number of

    different mechanisms (Table III) but the most common is the

    SNP. SNPs are predicted to account for much of the diversity

    observed between subjects of the same or even closely related

    species. The 142 million SNPs originally reported in a genomic

    map of human genetic variation was just a first hint of the true

    number of SNPs (Sachidanandamet al, 2001). Since the initial

    sequencing and mapping of the human genome, the number of

    SNPs reported has grown exponentially and over 2 million

    SNPs have been identified and validated (http://www.ncbi.-

    nih.gov/SNP/index.html).

    SNPs in exon sequences

    The SNPs can be silent or affect the translated gene product,

    either as missense mutations, or non-sense mutations. Single

    amino acid substitutions resulting from missense mutations in

    exon sequences are common. Accordingly, it is not surprising

    that it has been estimated that two thirds of all blood group

    antigens are defined by missense SNPs in blood group genes

    (Reid & Yazdanbakhsh, 1998). SNPs associated with some

    important pairs of antithetical antigens are listed in Table II.

    Non-sense SNPs are those that cause an immediate (and

    premature) stop codon e.g., a T>A mutation in the FY gene

    occurring at different points in three unrelated people of the

    Fy(ab) phenotype resulted in the substitution of tryptophan

    by a premature stop codon (Rios et al, 2000a). The mutationsat nucleotide 287, 407 or 408 demonstrate the effect of

    premature stop codons on protein synthesis, since there was no

    detectable Duffy protein present on the RBCs.

    SNPs cannot only alter the antigen expressed by a certain

    blood group molecule but also modify the number of copies

    expressed in the RBC membrane. This is well illustrated by the

    many RHD alleles in which a SNP results in weakened

    expression of the D antigen. Indeed, a single nucleotide change

    can have a profound effect on the amount of D antigen

    expressed at the RBC surface, reducing the amount by as much

    Table II. Single nucleotide polymorphisms (SNPs) associated with selected antigens in some important blood group systems.

    System symbol Antige n 1 Critical SNP (amino acid change) Antigen 2 Reference

    ABO A C796A, G803C* (Leu266Met, Gly168Ala) B Yamamoto et al(1990a)

    MNS M C59T, G71A, T72G (Ser1Leu, Gly5Glu) N Siebert and Fukuda (1986)

    S T143C (Met29Thr) s Siebert and Fukuda (1987)

    RH C T307C* (Ser103Pro) c Mouro et al (1993), Simseket al(1994)

    E C676G (Pro226Ala) e Mouro et al (1993), Simseket al(1994)

    LU Lua A252G (His77Arg) Lub El Nemer et al (1997), Parsons et al(1997)

    Aua A1637G (Thr539Ala) Aub Parsons et al(1997)

    KEL K1 T698C (Met193Thr) K2 Lee et al(1995)

    Kpa T961C (Trp281Arg) Kpb Lee et al(1996)

    FY Fy a G125A (Gly42Asp) Fy b Chaudhuri et al (1995), Iwamoto et al(1995),

    Mallinson et al (1995), Tournamille et al(1995b)

    JK Jk a G838A Asp280Asn Jk b Olives et al(1997)

    DI Dia C2561T (Leu854Pro) Dib Bruce et al(1994)

    Wra G1972A Glu658Lys Wrb Bruce et al(1995)

    YT Yta C1057A (His353Asn) Ytb Bartels et al(1993)

    SC Sc1 G169A (Gly57Arg) Sc2 Wagner et al(2003)

    DO Doa A793G (Asn265Asp) Dob Gubin et al(2000)

    CO Co

    a

    C134T (Ala45Val) Co

    b

    Smith et al(1994)LW LWa A308G (Gln70Arg) LWb Hermand et al(1995)

    CROM Tca G155T (Arg18Leu) Tcb Lublin et al(2000)

    IN Ina C252G (Pro46Arg) Inb Telen et al(1996)

    *Additional missense mutations that differ between these alleles can occur.

    Another missense mutation at this position encodes a blood group antigen of very low prevalence that is antithetical to antigen 1 and antigen 2.

    Review

    762 2004 Blackwell Publishing Ltd, British Journal of Haematology, 126, 759771

  • 8/9/2019 Genetic Basis of Blood Group Diversity - Storry - 2004 - British Journal of Haematology - Wiley Online Library

    5/13

    as 100-fold (Wagner et al, 1999, 2000). Another interesting

    example is the Fyx phenotype, in which a missense mutation

    that encodes an amino acid change in an intracellular loop of

    the molecule results in less Duffy glycoprotein on the cell

    surface (Olsson et al, 1998; Parasol et al, 1998; Tournamille

    et al, 1998). The SNP that encodes the Kpa polymorphism not

    only results in the altered antigen specificity but also affects

    trafficking of the Kell glycoprotein to the RBC surface so that

    there is a reduced expression of Kell in the cell membrane

    whilst increased amounts can be found intracellularly (Ya-

    zdanbakhshet al, 1999).

    Evolutionary pressure from various pathogens is generally

    thought to be responsible for the generation of genetic

    variants, the host effects of which determine whether or not

    they will survive over time. This has been discussed with a

    main focus on microbial pathogens, e.g. relating to the

    differences in carbohydrates expressed on cell surfaces

    (Gagneux & Varki, 1999) but there is also strong evidence

    for the role of malaria on the genetic variants of some of the

    integral RBC membrane proteins, such as the Duffy glycopro-

    tein and the RBC anion exchanger (AE1; band 3) (Milleret al,1976; Bruce & Tanner, 1999).

    SNPs in introns and regulatory regions

    The splice sites are crucial for exon fusion when the introns are

    removed by the splicing machinery of the cell during the

    maturation of hnRNA to mRNA. Mutations that affect the

    invariant GT at +1 and +2 of the 5-donor splice site or

    the invariant AG at 1 and 2 of the 3-receptor splice site

    will cause skipping of the preceding or succeeding exons

    respectively. Mutations of the less conserved nucleotides of the

    splice site recognition sequence can also affect exon processing

    and cause exon skipping. The Jknull phenotype in Polynesians

    (Irshaidet al, 2000) and some K0phenotypes (Lee et al, 2001)

    are examples of how this phenomenon can alter the RBC

    phenotype. Individuals whose RBCs carry null phenotypes,

    such as the Jknull and K0 phenotypes, are at risk of immun-

    ization by blood transfusion or pregnancy and once immun-

    ized, will require the provision of rare blood for any further

    transfusion therapy. The SsU+w phenotype in African-

    Americans also arises from exon-skipping events due either to

    a mutation in the intron 5 splice site or to mutations in exon 5

    that activate a cryptic splice site (Storryet al, 2003). The SNP

    responsible for the Dr(a) phenotype in the Cromer blood

    group system, also creates a cryptic splice site in the DAFgene

    that is used preferentially (Lublin et al, 1991). The product of

    the alternative splicing is not found on the RBC surface and

    consequently, Dr(a) RBCs express only 40% of normal levels

    of DAF.

    The SNPs that occur in the regulatory elements, such as the

    promoter or enhancer regions of blood group genes, canmodify or abolish antigen expression. A well-known example

    of such modification is the altered tissue distribution of the

    Duffy blood group antigens commonly found in individuals of

    African origin. A disruption of the GATA-1 motif in the

    promoter region of the FY*B gene by a single nucleotide

    substitution abolishes erythroid expression whilst the molecule

    is expressed normally in other tissues (Tournamille et al,

    1995a). This mutation is a perfect illustration of evolutionary

    pressure exerted by a pathogen since the Duffy protein is the

    exclusive receptor on mature RBCs for the malarial parasite,

    Table III. Molecular mechanisms that generate diversity in blood group genes.

    Type of change Molecular mechanism Example of gene event Phenotypic consequence

    Antithetical antigen Missense SNP KEL 698C>T K2fiK1 antigen

    Novel antigen Missense SNP GYPB161G>A Mit+

    Equal crossover between homologous genes GYP(B-A) Ss+wU, Dantu+

    DNA conversion between homologous genes RH(D-CE-D) DVI, BARC+

    Exon duplication GYPC.Lsa Ls(a+)

    Reduced amount of

    expected antigen

    Missense SNP ABO646T>A Ax

    FY298C>T Fy x

    CROM596C>T Dr(a)

    Splice site mutation GYPB intron 5 +5g>t SsU+w

    XK intron 2 +5g>a McLeod phenotype:

    weakened Kell antigens

    No protein product Non-se nse SNP DO 442T>C Gy(a)

    Nucleotide deletion RHAG 1086delA RhnullCO 232delG Co(ab)

    Mutation in transcription motif FY46T>C Fy(ab)

    Splice site mutation DO intron 1 2a>g Gy(a)

    Gene deletion DRHD D

    D

    GYPA En(a)Modifying gene In(LU) Lu(ab)

    *SNP, single nucleotide polymorphism; D, deletion.

    Review

    2004 Blackwell Publishing Ltd, British Journal of Haematology, 126, 759771 763

  • 8/9/2019 Genetic Basis of Blood Group Diversity - Storry - 2004 - British Journal of Haematology - Wiley Online Library

    6/13

    Plasmodium vivax (Miller et al, 1976). The Duffy protein is

    absent from the RBCs of up to 100% of native West Africans

    and consequently, these individuals are protected fromP. vivax

    infection. Furthermore, the same evolutionary pressure is

    proposed to have driven the identical mutation in the GATA-1

    sequences of the FY*A allele in a Papua New Guinean

    population but, interestingly, the mutation is thought to be a

    much more recent and unlinked event (Zimmerman et al,

    1999).

    Other mechanisms that contribute to blood group diversity

    While single nucleotide changes can have far-reaching conse-

    quences on gene product expression and function, there are

    also other mechanisms that contribute to diversity. Gene

    rearrangements due to recombination or gene conversions

    between homologous genes, such as those encoding the Rh and

    MNS blood group systems, can affect blood group expression

    in many different ways (Blumenfeld & Huang, 1997; Avent,

    2001). Surprisingly, the same is true for ABO where only a

    single gene locus results in multiple hybrid alleles, giving rise to

    unexpected phenotypes (Fig 1) (Olsson & Chester, 2001).

    Recombination between the two homologous genes in the Rhand MNS blood group systems is common and can lead to

    many different kinds of phenotype. Examples of exchange

    between homologous genes in trans are common in the MNS

    system where hybrids of GPA and GPB are created by unequal

    crossover or gene conversion events (Fig 2A) (Blumenfeld &

    Huang, 1997). New antigens arise as a result of the novel

    amino acid sequences generated by the hybrid genes. The

    hybrid molecules that carry unusual phenotypes in the Rh

    blood group system are thought to be generated by crossover

    between the RHD and RHCE genes in cis (Fig 2B) (Wagner

    et al, 2001), that may alter or abolish the expression of

    expected antigens and create novel antigens/phenotypes. For

    example, the partial D phenotype, DVI type I, results from a

    RH(D-CE-D) hybrid in which exons 4 and 5 of RHD are

    replaced by the corresponding exons of an RHcEallele (Avent

    et al, 1997; Huang, 1997). The hybrid protein expresses a

    qualitatively and quantitatively altered D antigen. A similar

    exchange of RHD with exons 4, 5 and 6 of an RHCe allele

    produces a hybrid protein with a qualitatively identical D

    antigen, as determined by monoclonal antibody studies;

    however, more copies of the D antigen are present and these

    RBCs also express the low incidence antigen, BARC (Mouro

    et al, 1994; Tippett et al, 1996).

    Non-sense mutations, such as nucleotide deletions or

    insertions, often abolish or decrease blood group expression

    by causing a shift in the open reading frame of the sequence

    such that the amino acids encoded after the mutation are

    completely different. For example, in the ABO system, two

    different single nucleotide deletions in the consensus A1

    sequence have been shown to account for the common O and

    A2 blood groups. These are illustrated in Fig 3 (Yamamoto

    et al, 1990a, 1992).

    Not surprisingly, the deletion of whole genes or parts ofgenes can result in loss of blood group antigen expression as

    exemplified by the following reports concerning the MNS

    (Huanget al, 1987; Rahuelet al, 1988), RH (Wagner & Flegel,

    2000), JK (Irshaid et al, 2002), H (Koda et al, 1997) and GE

    (Colinet al, 1989) blood group systems.

    Duplication of genetic material can result in the formation of

    a novel antigen, for example, the nucleotide sequence created

    by the exon 3-exon 3 duplication in the GYPC.Lsa variant gene

    encodes the Lsa antigen (Reid et al, 1994). Conversely, a

    duplication event may result in the loss of an existing antigen.

    Fig 1. Hybrid genes arising from crossover events between ABO genes have been shown to give rise to more or less unexpected phenotypes. In this

    example, a crossover in intron 6 between an O1v allele and aBallele to generate aBO1v hybrid was shown to be one molecular mechanism behind the

    Ax

    phenotype (Olsson & Chester, 1998). Weak A antigen expression occurs because exon 7 of the O1v allele encodes A transferase activity that is

    normally silenced by the presence of 261delG in exon 6. Since exon 6 is derived from the Ballele without this deletion in the hybrid, enzyme activity is

    restored. The corresponding product of the crossover would be expected to encode a non-functional protein since the 261delG mutation is present in

    exon 6. Indeed, such an allele was reported to be common in Brazilian blacks (Olsson et al, 1997) and has since been found in Blacks of several

    different geographic origins (unpublished observations). Note that the introns are not drawn to scale.

    Review

    764 2004 Blackwell Publishing Ltd, British Journal of Haematology, 126, 759771

  • 8/9/2019 Genetic Basis of Blood Group Diversity - Storry - 2004 - British Journal of Haematology - Wiley Online Library

    7/13

    The best example of this is the 37 bp nucleotide duplication

    that occurs at the intron 3/exon 4 border in the RHD

    pseudogene (Singleton et al, 2000). The duplication results in

    an alteration of the reading frame and an eventual premature

    stop codon so that no RhD protein is found on the RBCs.

    In theABOgene, a 43 bp minisatellite motif 4 kbp upstreamfrom exon 1, with the ability to bind the transcription factor

    CBF/NF-Y has been suggested to govern transcription in an

    enhancer-like way (Kominato et al, 1997). Allele-related vari-

    ation in the number of repeats was observed in samples of

    different ABO genotypes: A1 and O2 alleles having one copy

    only while A2, B, O1 and O1v alleles had four copies (Irshaid

    et al, 1999). In an experimental model, four repeats (associated

    with the common A2, B and O1/O1v alleles) produced

    approximately 100 times more mRNA than a single repeat

    (found in A1 andO2 alleles) (Yu et al, 2000).

    Blood group genes that control carbohydrate antigens

    All genes encode proteins but not all blood group genes encode

    blood-group-carrying proteins. The reason, of course, is that

    not all blood groups antigens are of protein nature. This

    apparent anomaly was indeed confusing to the pioneers whohad elucidated the biochemical basis of blood groups like

    ABO, H and Lewis. On one hand, DNA was supposed to code

    for proteins but, on the other hand, these carbohydrate blood

    groups were definitely inherited characters. The solution came

    when Watkins hypothesized that the genes encoded blood-

    group-specific glycosyltransferases (Watkins, 1974). This hypo-

    thesis held true, although today researchers still struggle to

    clarify the genetic heterogeneity underlying variant carbohy-

    drate blood groups. In theory, any mutation that changes the

    enzymatic properties of the primary gene product including

    Fig 2. Mechanisms that generate hybrid genes

    in the MNS and Rh blood group systems. (A)

    DNA exchange between the GYPA and GYPB

    genes intrans, either by unequal crossover or by

    gene conversion, creates novel sequences that

    are recognized as blood group antigens. (B)Crossover of the RHgenes in cis creates hybrid

    genes that have altered expression of expected

    antigens and may create novel antigens. SMPis

    an unrelated gene. Genes are illustrated in a

    53 direction unless noted otherwise. Novel

    sequences are indicated by the parenthesis.

    Review

    2004 Blackwell Publishing Ltd, British Journal of Haematology, 126, 759771 765

  • 8/9/2019 Genetic Basis of Blood Group Diversity - Storry - 2004 - British Journal of Haematology - Wiley Online Library

    8/13

    activity, substrate and acceptor preference may cause, e.g. a

    weak A or B subgroup (Chester & Olsson, 2001). The null

    phenotypes O, p, or Bombay result from the inheritance of two

    inactive alleles of the respective glycosyltransferase gene. In all

    three genes, inactivating mutations have been found through-

    out the coding sequence. For ABO, however, a single-

    nucleotide deletion is the predominant cause of the blood

    group O phenotype whilst other causes are infrequent or rare.

    Currently, the number of alleles at the ABO locus approaches a

    hundred and reaches a degree of complexity due to point

    mutations and hybrid allele formation that is only matched by

    theRHand MNS loci.

    Heterogeneity of null phenotypes

    By comparison with theABO locus, most of the protein-based

    systems are relatively simple and a polymorphism defined at

    the genetic level can almost always be correlated with the

    expression of a certain blood group antigen. However, in

    genomic DNA-based analysis, the interpretation of results can

    be confounded by the existence of null phenotypes. Clearly,

    any mutation in the gene that results in the failure of the

    antigen-carrying molecule to be expressed at the RBC surface

    will, in fact, be a null mutation. Because some of these

    mutations occur relatively commonly, it is important toinclude their detection together with assays for common

    phenotype prediction in order to avoid false positive results.

    For example, it is necessary to test for the GATA-1 mutation in

    the FY gene described above, when determining Fya and Fyb

    antigen status but not for the other very rare mutations that

    result in the Fy(ab) phenotype. Similarly, in the Nordic

    population, it should be considered to include detection of

    the 871T>C mutation in the JKgene that is the basis for the

    Jk(ab) phenotype in the Finnish people (Irshaid et al, 2000).

    The heterogeneity of the molecular bases of these phenotypes is

    a major problem for all DNA-based prediction of blood

    groups.

    Most null blood group phenotypes are the result of

    molecular changes in the gene that encodes the carrier

    molecule. However, there are important interactions between

    proteins at the cell surface and with the cytoskeleton and

    therefore mutations that change the expression of an interact-

    ive protein can affect the proteins around it. Mutations in

    RHAG that stop the expression of the Rh-associated glyco-

    protein (RhAG) also prevent the expression of the RhD and

    RhCE proteins the so-called regulator type of Rhnullphenotype (reviewed in Daniels, 2002). Similarly, mutations

    leading to the absence of the XK protein in the red cell

    membrane results in the weakened expression of Kell blood

    group antigens (Daneket al, 2001; Russo et al, 2002).

    Future perspectives

    Today, almost all of the genes underlying expression of the

    human blood group systems have been cloned and the

    polymorphisms responsible for the phenotypes encountered

    in different individuals and populations clarified. A challenge

    that remains for the future is to investigate and understand the

    genetics of antigens in the blood group collections and series

    since our experience tells us that they are likely to be carried onfunctional molecules.

    The increasing knowledge of the genes encoding blood

    group antigens has relevance in the clinical laboratory.

    Genomic typing assays for fetal RBC phenotype prediction to

    determine the risk to a foetus of haemolytic disease of the

    newborn are now standard of care. Much of this testing is

    performed on DNA isolated from amniotic fluid; however, in

    the last few years, more sensitive quantitative PCR techniques

    have permitted the identification of fetal RHD alleles from

    DNA isolated from maternal plasma (Lo et al, 1998; Lo, 2001;

    Fig 3. A single nucleotide deletion in the coding region of a gene can alter the open reading frame. For example, in the ABO blood group system, the

    deletion of 261G in the consensus sequence (A1 allele) results in a frameshift and a subsequent introduction of a premature stop codon (O1 allele).

    The truncated protein that is encoded is inactive. Conversely, the A2 allele results from the deletion of 1061C, and instead, the open reading frame is

    elongated. The glycosyltransferase encoded by this gene consists of 21 additional amino acids and is less efficient, as determined by the presence of

    fewer A antigens on the RBCs of group A2 people. In addition, it appears to be unable to synthesize the A1 antigen. Asterisks (*) represent stop

    codons. The grey shading indicates nucleotides that are transcribed normally; the white boxes indicate a nucleotide sequence that is not transcribed as

    a result of the nucleotide deletion at position 261; the hatched box represents the additional nucleotide sequence that is transcribed as a result of the

    nucleotide deletion at 1061.

    Review

    766 2004 Blackwell Publishing Ltd, British Journal of Haematology, 126, 759771

  • 8/9/2019 Genetic Basis of Blood Group Diversity - Storry - 2004 - British Journal of Haematology - Wiley Online Library

    9/13

    van der Schoot et al, 2003). Although there are some current

    limitations to the technique, the advantages of this non-

    invasive method are obvious. Other applications of blood

    group genotyping have included testing samples from multiply

    transfused patients that are either immunized, or at risk of

    being immunized, to one or more blood group antigens (Reid

    et al, 2000). Patient groups, such as those with Sickle Cell

    Disease or other transfusion-dependent haemoglobinopathies,

    can benefit from better antigen-matched blood (Reed &

    Vichinsky, 2001). Genotyping is also useful in other serological

    situations where the RBC phenotype cannot be accurately

    determined (Rios et al, 2000b).

    The potential for testing blood donor samples on a large

    scale is clear to all in the field, but current techniques are both

    laborious and expensive. However, automation of SNP detec-

    tion, as a faster and easier way to type blood donors, is a

    much-discussed issue and there are several techniques being

    evaluated.

    Lastly, the knowledge gained from the identification of

    blood group genes leads to a better understanding of therelationship between blood group differences and subtle

    functional differences in the molecules that carry the antigens.

    The molecular genetics of blood groups may also help us to

    better understand the functionality of the RBC at large, e.g.

    moderation of the intracellular environment for invading

    parasites, plasticity of the RBC membrane in the circulation

    during stress, or the role of the circulating RBC in haemostasis.

    References

    Amado, M., Almeida, R., Carneiro, F., Levery, S.B., Holmes, E.H.,

    Nomoto, M., Hollingsworth, M.A., Hassan, H., Schwientek, T.,

    Nielsen, P.A., Bennett, E.P. & Clausen, H. (1998) A family of human

    beta3-galactosyltransferases. Characterization of four members of a

    UDP-galactose: beta-N-acetyl-glucosamine/beta-nacetyl-galactosa-

    mine beta-1,3-galactosyltransferase family. Journal of Biological

    Chemistry, 273, 1277012778.

    Arce, M.A., Thompson, E.S., Wagner, S., Coyne, K.E., Ferdman, B.A. &

    Lublin, D.M. (1993) Molecular cloning of RhD cDNA derived from

    a gene present in RhD-positive, but not RhD-negative individuals.

    Blood, 82, 651655.

    Avent, N.D. (2001) Molecular biology of the Rh blood group system.

    Journal of Pediatric Hematology/Oncology, 23, 394402.

    Avent, N.D., Ridgwell, K., Tanner, M.J. & Anstee, D.J. (1990) cDNA

    cloning of a 30 kDa erythrocyte membrane protein associated with

    Rh (Rhesus)-blood-group-antigen expression. Biochemical Journal,271,821825.

    Avent, N.D., Liu, W., Jones, J.W., Scott, M.L., Voak, D., Pisacka, M.,

    Watt, J. & Fletcher, A. (1997) Molecular analysis of Rh transcripts

    and polypeptides from individuals expressing the DVI variant

    phenotype: an RHD gene deletion event does not generate All

    DVIccEe phenotypes. Blood, 89, 17791786.

    Bailly, P., Hermand, P., Callebaut, I., Sonneborn, H.H., Khamlichi, S.,

    Mornon, J.P. & Cartron, J.P. (1994) The LW blood group glyco-

    protein is homologous to intercellular adhesion molecules. Pro-

    ceedings of the National Academy of Sciences of the United States of

    America, 91, 53065310.

    Bartels, C.F., Zelinski, T. & Lockridge, O. (1993) Mutation at codon

    322 in the human acetylcholinesterase (ACHE) gene accounts for YT

    blood group polymorphism. American Journal of Human Genetics,

    52, 928936.

    Bierhuizen, M.F., Mattei, M.G. & Fukuda, M. (1993) Expression of the

    developmental I antigen by a cloned human cDNA encoding a

    member of a beta-1,6-N-acetylglucosaminyltransferase gene family.

    Genes and Development, 7, 468478.Blumenfeld, O.O. & Huang, C.H. (1997) Molecular genetics of gly-

    cophorin MNS variants. Transfusion Clinique et Biologique, 4, 357

    365.

    Bruce, L.J. & Tanner, M.J. (1999) Erythroid band 3 variants and dis-

    ease. Baillieres Best Practice and Research Clinical Haematology, 12,

    637654.

    Bruce, L.J., Anstee, D.J., Spring, F.A. & Tanner, M.J. (1994) Band 3

    Memphis variant II. Altered stilbene disulfonate binding and the

    Diego (Dia) blood group antigen are associated with the human

    erythrocyte band 3 mutation Pro854>Leu. Journal of Biological

    Chemistry, 269, 1615516158.

    Bruce, L.J., Ring, S.M., Anstee, D.J., Reid, M.E., Wilkinson, S. &

    Tanner, M.J. (1995) Changes in the blood group Wright antigens are

    associated with a mutation at amino acid 658 in human erythrocyteband 3: a site of interaction between band 3 and glycophorin A

    under certain conditions. Blood, 85, 541547.

    Caras, I.W., Davitz, M.A., Rhee, L., Weddell, G., Martin, Jr., D.W. &

    Nussenzweig, V. (1987) Cloning of decay-accelerating factor sug-

    gests novel use of splicing to generate two proteins. Nature, 325,

    545549.

    Chaudhuri, A., Polyakova, J., Zbrzezna, V., Williams, K., Gulati, S. &

    Pogo, A.O. (1993) Cloning of glycoprotein D cDNA, which encodes

    the major subunit of the Duffy blood group system and the receptor

    for the Plasmodium vivax malaria parasite. Proceedings of the

    National Academy of Sciences of the United States of America, 90,

    1079310797.

    Chaudhuri, A., Polyakova, J., Zbrzezna, V. & Pogo, A.O. (1995) The

    coding sequence of Duffy blood group gene in humans and simians:

    restriction fragment length polymorphism, antibody and malarial

    parasite specificities, and expression in nonerythroid tissues in

    Duffy-negative individuals. Blood, 85, 615621.

    Cherif-Zahar, B., Bloy, C., Le Van, K.C., Blanchard, D., Bailly, P.,

    Hermand, P., Salmon, C., Cartron, J.P. & Colin, Y. (1990) Molecular

    cloning and protein structure of a human blood group Rh poly-

    peptide.Proceedings of the National Academy of Sciences of the United

    States of America, 87, 62436247.

    Chester, M.A. & Olsson, M.L. (2001) The ABO blood group gene A

    locus of considerable genetic diversity. Transfusion Medicine Reviews,

    11, 295313.

    Colin, Y., Rahuel, C., London, J., Romeo, P.H., dAuriol, L., Galibert,

    F. & Cartron, J.P. (1986) Isolation of cDNA clones and completeamino acid sequence of human erythrocyte glycophorin C.Journal of

    Biological Chemistry, 261, 229233.

    Colin, Y., Le Van, K.C., Tsapis, A., Clerget, M., dAuriol, L., London, J.,

    Galibert, F. & Cartron, J.P. (1989) Human erythrocyte glycophorin

    C. Gene structure and rearrangement in genetic variants. Journal of

    Biological Chemistry, 264, 37733780.

    Danek, A., Rubio, J.P., Rampoldi, L., Ho, M., Dobson-Stone, C., Tison, F.,

    Symmans, W.A., Oechsner, M., Kalckreuth, W., Watt, J.M., Corbett,

    A.J., Hamdalla, H.H., Marshall, A.G., Sutton, I., Dotti, M.T., Ma-

    landrini, A., Walker, R.H., Daniels, G. & Monaco, A.P. (2001)

    Review

    2004 Blackwell Publishing Ltd, British Journal of Haematology, 126, 759771 767

  • 8/9/2019 Genetic Basis of Blood Group Diversity - Storry - 2004 - British Journal of Haematology - Wiley Online Library

    10/13

    McLeod neuroacanthocytosis: genotype and phenotype. Annals of

    Neurology, 50, 755764.

    Daniels, G. (2002)Human Blood Groups, 2nd edn, Blackwell Scientific,

    Oxford, UK.

    Daniels, G.L., Cartron, J.P., Fletcher, A., Garratty, G., Henry, S.,

    Jorgensen, J., Judd, W.J., Levene, C., Lin, M., Lomas-Francis, C.,

    Moulds, J.J., Moulds, J.M., Moulds, M., Overbeeke, M., Reid, M.E.,

    Rouger, P., Scott, M., Sistonen, P., Smart, E., Tani, Y., Wendel, S. &Zelinski, T. (2003) International Society of Blood Transfusion

    Committee on terminology for red cell surface antigens:Vancouver

    Report. Vox Sanguinis, 84, 244247.

    Darling, S.M., Banting, G.S., Pym, B., Wolfe, J. & Goodfellow, P.N.

    (1986) Cloning an expressed gene shared by the human sex chro-

    mosomes. Proceedings of the National Academy of Sciences of the

    United States of America, 83, 135139.

    El Nemer, W., Rahuel, C., Colin, Y., Gane, P., Cartron, J.P. & Le Van,

    K.C. (1997) Organization of the human LU gene and molecular

    basis of the Lu(a)/Lu(b) blood group polymorphism. Blood, 89,

    46084616.

    Ellis, N.A., Tippett, P., Petty, A., Reid, M., Weller, P.A., Ye, T.Z.,

    German, J., Goodfellow, P.N., Thomas, S. & Banting, G. (1994)

    PBDX is the XG blood group gene. Nature Genetics, 8, 285290.Gagneux, P. & Varki, A. (1999) Evolutionary considerations in relating

    oligosaccharide diversity to biological function. Glycobiology,9,747

    755.

    Goldstein, L.A. & Butcher, E.C. (1990) Identification of mRNA that

    encodes an alternative form of H-CAM(CD44) in lymphoid and

    nonlymphoid tissues. Immunogenetics, 32, 389397.

    Gubin, A.N., Njoroge, J.M., Wojda, U., Pack, S.D., Rios, M., Reid, M.E.

    & Miller, J.L. (2000) Identification of the dombrock blood group

    glycoprotein as a polymorphic member of the ADP-ribosyl-

    transferase gene family. Blood, 96, 26212627.

    Guo, H., Majmudar, G., Jensen, T.C., Biswas, C., Toole, B.P. & Gor-

    don, M.K. (1998) Characterization of the gene for human EMM-

    PRIN, a tumor cell surface inducer of matrix metalloproteinases.

    Gene, 220, 99108.

    Harn, H.J., Isola, N. & Cooper, D.L. (1991) The multispecific cell

    adhesion molecule CD44 is represented in reticulocyte cDNA.Bio-

    chemical and Biophysical Research Communications,178,11271134.

    Hasegawa, H., Utsunomiya, Y., Kishimoto, K., Yanagisawa, K. & Fujita,

    S. (1996) SFA-1, a novel cellular gene induced by human T-cell

    leukemia virus type 1, is a member of the transmembrane 4 su-

    perfamily. Journal of Virology, 70, 32583263.

    Hermand, P., Gane, P., Mattei, M.G., Sistonen, P., Cartron, J.P. &

    Bailly, P. (1995) Molecular basis and expression of the LWa/LWb

    blood group polymorphism. Blood, 86, 15901594.

    Ho, M., Chelly, J., Carter, N., Danek, A., Crocker, P. & Monaco, A.P.

    (1994) Isolation of the gene for McLeod syndrome that encodes a

    novel membrane transport protein. Cell, 77, 869880.Huang, C.H. (1997) Human DVI category erythrocytes: correlation of

    the phenotype with a novel hybrid RhD-CE-D gene but not an

    internally deleted RhD gene. Blood, 89, 18341835.

    Huang, C.H., Johe, K., Moulds, J.J., Siebert, P.D., Fukuda, M. &

    Blumenfeld, O.O. (1987) Delta glycophorin (glycophorin B) gene

    deletion in two individuals homozygous for the SsU blood group

    phenotype. Blood, 70, 18301835.

    Inase, N., Fushimi, K., Ishibashi, K., Uchida, S., Ichioka, M., Sasaki, S.

    & Marumo, F. (1995) Isolation of human aquaporin 3 gene.Journal

    of Biological Chemistry, 270, 1791317916.

    Irshaid, N.M., Chester, M.A. & Olsson, M.L. (1999) Allele-related

    variation in minisatellite repeats involved in the transcription of the

    blood group ABO gene. Transfusion Medicine, 9, 219226.

    Irshaid, N.M., Henry, S.M. & Olsson, M.L. (2000) Genomic char-

    acterization of the Kidd blood group gene: different molecular basis

    of the Jk(a-b-) phenotype in Polynesians and Finns. Transfusion,40,

    6974.

    Irshaid, N.M., Eicher, N.I., Hustinx, H., Poole, J. & Olsson, M.L.(2002) Novel alleles at the JK blood group locus explain the absence

    of the erythrocyte urea transporter in European families. British

    Journal of Haematology, 116, 445453.

    Issitt, P. & Anstee, D.J. (1998) Applied Blood Group Serology, 4th edn,

    Montgomery Scientific Publications, Miami, FL, USA.

    Iwamoto, S., Omi, T., Kajii, E. & Ikemoto, S. (1995) Genomic orga-

    nization of the glycoprotein D gene: Duffy blood group Fya/Fyb

    alloantigen system is associated with a polymorphism at the 44-

    amino acid residue. Blood, 85, 622626.

    Kajii, E., Umenishi, F., Iwamoto, S. & Ikemoto, S. (1993) Isolation of a

    new cDNA clone encoding an Rh polypeptide associated with the Rh

    blood group system. Human Genetics, 91, 157162.

    Kelly, R.J., Ernst, L.K., Larsen, R.D., Bryant, J.G., Robinson, J.S. &

    Lowe, J.B. (1994) Molecular basis for H blood group deficiency inBombay (Oh) and para-Bombay individuals. Proceedings of the

    National Academy of Sciences of the United State of America, 91,

    58435847.

    Koda, Y., Soejima, M., Johnson, P.H., Smart, E. & Kimura, H. (1997)

    Missense mutation of FUT1 and deletion of FUT2 are responsible

    for Indian Bombay phenotype of ABO blood group system. Bio-

    chemical and Biophysical Research Communications, 238, 2125.

    Kominato, Y., Tsuchiya, T., Hata, N., Takizawa, H. & Yamamoto, F.

    (1997) Transcription of human ABO histo-blood group genes is

    dependent upon binding of transcription factor CBF/NF-Y to min-

    isatellite sequence.Journal of Biological Chemistry,272,2589025898.

    Kukowska-Latallo, J.F., Larsen, R.D., Nair, R.P. & Lowe, J.B. (1990) A

    cloned human cDNA determines expression of a mouse stage-spe-

    cific embryonic antigen and the Lewis blood group alpha(1,3/

    1,4)fucosyltransferase. Genes and Development, 4, 12881303.

    Le Van Kim, C., Mouro, I., Cherif-Zahar, B., Raynal, V., Cherrier, C.,

    Cartron, J.P. & Colin, Y. (1992) Molecular cloning and primary

    structure of the human blood group RhD polypeptide.Proceedings of

    the National Academy of Sciences of the United State of America , 89,

    1092510929.

    Lee, S., Zambas, E.D., Marsh, W.L. & Redman, C.M. (1991) Molecular

    cloning and primary structure of Kell blood group protein.

    Proceedings of the National Academy of Sciences of the United State of

    America, 88, 63536357.

    Lee, S., Wu, X., Reid, M., Zelinski, T. & Redman, C. (1995) Molecular

    basis of the Kell (K1) phenotype. Blood, 85, 912916.

    Lee, S., Wu, X., Son, S., Naime, D., Reid, M., Okubo, Y., Sistonen, P. &Redman, C. (1996) Point mutations characterize KEL10, the KEL3,

    KEL4, and KEL21 alleles, and the KEL17 and KEL11 alleles. Trans-

    fusion, 36, 490494.

    Lee, S., Russo, D.C., Reiner, A.P., Lee, J.H., Sy, M.Y., Telen, M.J., Judd,

    W.J., Simon, P., Rodrigues, M.J., Chabert, T., Poole, J., Jovanovic-

    Srzentic, S., Levene, C., Yahalom, V. & Redman, C.M. (2001) Mo-

    lecular defects underlying the Kell null phenotype. Journal of Bio-

    logical Chemistry, 276, 2728127289.

    Li, Y., Camp, S., Rachinsky, T.L., Getman, D. & Taylor, P. (1991) Gene

    structure of mammalian acetylcholinesterase. Alternative exons

    Review

    768 2004 Blackwell Publishing Ltd, British Journal of Haematology, 126, 759771

  • 8/9/2019 Genetic Basis of Blood Group Diversity - Storry - 2004 - British Journal of Haematology - Wiley Online Library

    11/13

    dictate tissue-specific expression. Journal of Biological Chemistry,

    266,2308323090.

    Lo, Y.M. (2001) Fetal DNA in maternal plasma: application to non-

    invasive blood group genotyping of the fetus. Transfusion Clinique et

    Biologique, 8, 306310.

    Lo, Y.M., Hjelm, N.M., Fidler, C., Sargent, I.L., Murphy, M.F.,

    Chamberlain, P.F., Poon, P.M., Redman, C.W. & Wainscoat, J.S.

    (1998) Prenatal diagnosis of fetal RhD status by molecular analysisof maternal plasma. New England Journal of Medicine, 339, 1734

    1738.

    Lublin, D.M., Thompson, E.S., Green, A.M., Levene, C. & Telen, M.J.

    (1991) Dr(a-) polymorphism of decay accelerating factor. Bio-

    chemical, functional, and molecular characterization and produc-

    tion of allele-specific transfectants. Journal of Clinical Investigation,

    87, 19451952.

    Lublin, D.M., Kompelli, S., Storry, J.R. & Reid, M.E. (2000) Molecular

    basis of Cromer blood group antigens. Transfusion, 40, 208213.

    Lux, S.E., John, K.M., Kopito, R.R. & Lodish, H.F. (1989) Cloning and

    characterization of band 3, the human erythrocyte anion-exchange

    protein (AE1).Proceedings of the National Academy of Sciences of the

    United States of America, 86, 90899093.

    Mallinson, G., Soo, K.S., Schall, T.J., Pisacka, M. & Anstee, D.J. (1995)Mutations in the erythrocyte chemokine receptor (Duffy) gene: the

    molecular basis of the Fya/Fyb antigens and identification of a de-

    letion in the Duffy gene of an apparently healthy individual with the

    Fy(a-b-) phenotype. British Journal of Haematology, 90, 823829.

    Medof, M.E., Lublin, D.M., Holers, V.M., Ayers, D.J., Getty, R.R.,

    Leykam, J.F., Atkinson, J.P. & Tykocinski, M.L. (1987) Cloning and

    characterization of cDNAs encoding the complete sequence of de-

    cay-accelerating factor of human complement. Proceedings of the

    National Academy of Sciences of the United States of America, 84,

    20072011.

    Miller, L.H., Mason, S.J., Clyde, D.F. & McGinniss, M.H. (1976) The

    Resistance factor to Plasmodium vivax in blacks. The Duffy-blood-

    group genotype. New England Journal of Medicine, 295, 302304.

    Mouro, I., Colin, Y., Cherif-Zahar, B., Cartron, J.P. & Le Van, K.C.

    (1993) Molecular genetic basis of the human Rhesus blood group

    system. Nature Genetics, 5, 6265.

    Mouro, I., Le Van, K.C., Rouillac, C., van Rhenen, D.J., Le Pennec,

    P.Y., Bailly, P., Cartron, J.P. & Colin, Y. (1994) Rearrangements of

    the blood group RhD gene associated with the DVI category phe-

    notype. Blood, 83, 11291135.

    Olives, B., Neau, P., Bailly, P., Hediger, M.A., Rousselet, G., Cartron,

    J.P. & Ripoche, P. (1994) Cloning and functional expression of a

    urea transporter from human bone marrow cells. Journal of Biolo-

    gical Chemistry, 269, 3164931652.

    Olives, B., Merriman, M., Bailly, P., Bain, S., Barnett, A., Todd, J.,

    Cartron, J.P. & Merriman, T. (1997) The molecular basis of the

    Kidd blood group polymorphism and its lack of association withtype 1 diabetes susceptibility. Human Molecular Genetics, 6, 1017

    1020.

    Olsson, M.L. & Chester, M.A. (1998) Heterogeneity of the blood group

    Ax allele: genetic recombination of common alleles can result in the

    Axphenotype. Transfusion Medicine, 8, 231238.

    Olsson, M.L. & Chester, M.A. (2001) Polymorphism and recombina-

    tion events at the ABO locus: a major challenge for genomic ABO

    blood grouping strategies. Transfusion Medicine, 11, 295313.

    Olsson, M.L., Guerreiro, J.F., Zago, M.A. & Chester, M.A. (1997)

    Molecular analysis of the O alleles at the blood group ABO locus in

    populations of different ethnic origin reveals novel crossing-over

    events and point mutations. Biochemical and Biophysical Research

    Communications, 234, 779782.

    Olsson, M.L., Smythe, J.S., Hansson, C., Poole, J., Mallinson, G., Jones,

    J., Avent, N.D. & Daniels, G. (1998) The Fy(x) phenotype is asso-

    ciated with a missense mutation in the Fy(b) allele predicting

    Arg89Cys in the Duffy glycoprotein. British Journal of Haematology,

    103,11841191.Parasol, N., Reid, M., Rios, M., Castilho, L., Harari, I. & Kosower, N.S.

    (1998) A novel mutation in the coding sequence of the FY*B allele of

    the Duffy chemokine receptor gene is associated with an altered

    erythrocyte phenotype. Blood, 92, 22372243.

    Parsons, S.F., Mallinson, G., Holmes, C.H., Houlihan, J.M., Simpson,

    K.L., Mawby, W.J., Spurr, N.K., Warne, D., Barclay, A.N. & Anstee,

    D.J. (1995) The Lutheran blood group glycoprotein, another

    member of the immunoglobulin superfamily, is widely expressed in

    human tissues and is developmentally regulated in human liver.

    Proceedings of the National Academy of Sciences of the United States of

    America, 92, 54965500.

    Parsons, S.F., Mallinson, G., Daniels, G.L., Green, C.A., Smythe, J.S. &

    Anstee, D.J. (1997) Use of domain-deletion mutants to locate Lu-

    theran blood group antigens to each of the five immunoglobulinsuperfamily domains of the Lutheran glycoprotein: elucidation of

    the molecular basis of the Lu(a)/Lu(b) and the Au(a)/Au(b) poly-

    morphisms. Blood, 89, 42194225.

    Preston, G.M. & Agre, P. (1991) Isolation of the cDNA for erythrocyte

    integral membrane protein of 28 kilodaltons: member of an ancient

    channel family.Proceedings of the National Academy of Sciences of the

    United States of America, 88, 1111011114.

    Rahuel, C., London, J., Vignal, A., Cherif-Zahar, B., Colin, Y., Siebert,

    P., Fukuda, M. & Cartron, J.P. (1988) Alteration of the genes for

    glycophorin A and B in glycophorin-A-deficient individuals. Eur-

    opean Journal of Biochemistry, 177, 605614.

    Reed, W. & Vichinsky, E.P. (2001) Transfusion therapy: a coming-of-

    age treatment for patients with sickle cell disease. Journal of Pediatric

    Hematology/Oncology, 23, 197202.

    Reid, M.E. & Lomas-Francis, C. (2003) The Blood Group Antigen

    FactsBook, 2nd edn, Academic Press, San Diego.

    Reid, M.E. & Yazdanbakhsh, K. (1998) Molecular insights into blood

    groups and implications for blood transfusion.Current Opinions in

    Hematology, 5, 93102.

    Reid, M.E., Mawby, W., King, M.J. & Sistonen, P. (1994) Duplication

    of exon 3 in the glycophorin C gene gives rise to the Lsa blood group

    antigen. Transfusion, 34, 966969.

    Reid, M.E., Rios, M., Powell, V.I., Charles-Pierre, D. & Malavade, V.

    (2000) DNA from blood samples can be used to genotype patients

    who have recently received a transfusion. Transfusion, 40, 4853.

    Rios, M., Chaudhuri, A., Mallinson, G., Sausais, L., Gomensoro-Gar-

    cia, A.E., Hannon, J., Rosenberger, S., Poole, J., Burgess, G., Pogo, O.& Reid, M. (2000a) New genotypes in Fy(a-b-) individuals: nonsense

    mutations (Trp to stop) in the coding sequence of either FY A or FY

    B. British Journal of Haematology, 108, 448454.

    Rios, M., Hue-Roye, K., Storry, J.R. & Reiss, R.F. (2000b) Cell typing

    the sensitized transfusion-dependent patient. Annals of Clinical La-

    boratory Science, 30, 379386.

    Russo, D.C., Lee, S., Reid, M.E. & Redman, C.M. (2002) Point mu-

    tations causing the McLeod phenotype. Transfusion, 42, 287293.

    Sachidanandam, R., Weissman, D., Schmidt, S.C., Kakol, J.M., Stein,

    L.D., Marth, G., Sherry, S., Mullikin, J.C., Mortimore, B.J., Willey,

    Review

    2004 Blackwell Publishing Ltd, British Journal of Haematology, 126, 759771 769

  • 8/9/2019 Genetic Basis of Blood Group Diversity - Storry - 2004 - British Journal of Haematology - Wiley Online Library

    12/13

    D.L., Hunt,S.E.,Cole,C.G.,Coggill, P.C., Rice, C.M., Ning,Z., Rogers,

    J., Bentley, D.R., Kwok, P.Y., Mardis, E.R., Yeh, R.T., Schultz, B.,

    Cook, L., Davenport, R., Dante, M., Fulton, L., Hillier, L., Waterston,

    R.H., McPherson, J.D., Gilman, B., Schaffner, S., Van Etten, W.J.,

    Reich, D., Higgins, J., Daly, M.J., Blumenstiel, B., Baldwin, J., Stange-

    Thomann, N., Zody, M.C., Linton, L., Lander, E.S. & Altshuler, D.

    (2001) A map of human genome sequence variation containing 1.42

    million single nucleotide polymorphisms. Nature,409,928933.van der Schoot, C.E., Tax, G.H., Rijnders, R.J., de Haas, M. & Chris-

    tiaens, G.C. (2003) Prenatal typing of Rh and Kell blood group

    system antigens: the edge of a watershed. Transfusion Medicine Re-

    views, 17, 3144.

    Siebert, P.D. & Fukuda, M. (1986) Isolation and characterization of

    human glycophorin A cDNA clones by a synthetic oligonucleotide

    approach: nucleotide sequence and mRNA structure. Proceedings of

    the National Academy of Sciences of the United States of America, 83,

    16651669.

    Siebert, P.D. & Fukuda, M. (1987) Molecular cloning of a human

    glycophorin B cDNA:nucleotide sequence and genomic relationship

    to glycophorin A. Proceedings of the National Academy of Sciences of

    the United States of America, 84, 67356739.

    Simsek, S., de Jong, C.A., Cuijpers, H.T., Bleeker, P.M., Westers, T.M.,Overbeeke, M.A., Goldschmeding, R., van der Schoot, C.E. & Dem

    Borne, A.E. (1994) Sequence analysis of cDNA derived from

    reticulocyte mRNAs coding for Rh polypeptides and demonstration

    of E/e and C/c polymorphisms. Vox Sanguinis, 67, 203209.

    Singleton, B.K., Green, C.A., Avent, N.D., Martin, P.G., Smart, E.,

    Daka, A., Narter-Olaga, E.G., Hawthorne, L.M. & Daniels, G. (2000)

    The presence of an RHD pseudogene containing a 37 base pair

    duplication and a nonsense mutation in africans with the Rh D-

    negative blood group phenotype. Blood, 95, 1218.

    Smith, B.L., Preston, G.M., Spring, F.A., Anstee, D.J. & Agre, P. (1994)

    Human red cell aquaporin CHIP. I. Molecular characterization of

    ABH and Colton blood group antigens. Journal of Clinical

    Investigation, 94, 10431049.

    Steffensen, R., Carlier, K., Wiels, J., Levery, S.B., Stroud, M., Ceder-

    gren, B., Nilsson, S.B., Bennett, E.P., Jersild, C. & Clausen, H. (2000)

    Cloning and expression of the histo-blood group Pk UDP-galactose:

    Ga1beta-4G1cbeta1-cer alpha1, 4-galactosyltransferase. Molecular

    genetic basis of the p phenotype. Journal of Biological Chemistry,275,

    1672316729.

    Storry, J.R., Reid, M.E., Fetics, S. & Huang, C.-H. (2003) Mutations

    in GYPB exon 5 drive the SsU+var phenotype in persons of

    African descent: implications for transfusion. Transfusion, 43,

    17381747.

    Tanner, M.J., Martin, P.G. & High, S. (1988) The complete amino acid

    sequence of the human erythrocyte membrane anion-transport

    protein deduced from the cDNA sequence. Biochemistry Journal,

    256,703712.Telen, M.J., Udani, M., Washington, M.K., Levesque, M.C., Lloyd, E. &

    Rao, N. (1996) A blood group-related polymorphism of CD44

    abolishes a hyaluronan-binding consensus sequence without pre-

    venting hyaluronan binding. Journal of Biological Chemistry, 271,

    71477153.

    Tippett, P., Lomas-Francis, C. & Wallace, M. (1996) The Rh antigen D:

    partial D antigens and associated low incidence antigens. Vox San-

    guinis, 70, 123131.

    Tournamille, C., Colin, Y., Cartron, J.P. & Le Van, K.C. (1995a)

    Disruption of a GATA motif in the Duffy gene promoter abolishes

    erythroid gene expression in Duffy-negative individuals. Nature

    Genetics, 10, 224228.

    Tournamille, C., Le Van, K.C., Gane, P., Cartron, J.P. & Colin, Y.

    (1995b) Molecular basis and PCR-DNA typing of the Fya/fyb blood

    group polymorphism. Human Genetics, 95, 407410.

    Tournamille, C., Le Van, K.C., Gane, P., Le Pennec, P.Y., Roubinet, F.,

    Babinet, J., Cartron, J.P. & Colin, Y. (1998) Arg89Cys substitution

    results in very low membrane expression of the Duffy antigen/re-ceptor for chemokines in Fy(x) individuals.Blood, 92, 21472156.

    Wagner, F.F. & Flegel, W.A. (2000) RHD gene deletion occurred in the

    Rhesus box. Blood, 95, 36623668.

    Wagner, F.F., Gassner, C., Muller, T.H., Schonitzer, D., Schunter, F. &

    Flegel, W.A. (1999) Molecular basis of weak D phenotypes. Blood,

    93,385393.

    Wagner, F.F., Frohmajer, A., Ladewig, B., Eicher, N.I., Lonicer, C.B.,

    Muller, T.H., Siegel, M.H. & Flegel, W.A. (2000) Weak D alleles

    express distinct phenotypes. Blood, 95, 26992708.

    Wagner, F.F., Frohmajer, A. & Flegel, W.A. (2001) RHD positive

    haplotypes in D negative Europeans. BMC Genetics, 2, 10.

    Wagner, F.F., Poole, J. & Flegel, W.A. (2003) Scianna antigens in-

    cluding Rd are expressed by ERMAP. Blood, 101, 752757.

    Watkins, W.M. (1974) Genetic regulation of the structure of blood-group-specific glycoproteins.Biochemical Society Symposia, 40, 125

    146.

    Wong, W.W., Cahill, J.M., Rosen, M.D., Kennedy, C.A., Bonaccio,

    E.T., Morris, M.J., Wilson, J.G., Klickstein, L.B. & Fearon, D.T.

    (1989) Structure of the human CR1 gene. Molecular basis of the

    structural and quantitative polymorphisms and identification of a

    new CR1-like allele.Journal of Experimental Medicine,169,847863.

    Yamada, A., Kubo, K., Takeshita, T., Harashima, N., Kawano, K.,

    Mine, T., Sagawa, K., Sugamura, K. & Itoh, K. (1999) Molecular

    cloning of a glycosylphosphatidylinositol-anchored molecule

    CDw108. Journal of Immunology, 162, 40944100.

    Yamamoto, F., Clausen, H., White, T., Marken, J. & Hakomori, S.

    (1990a) Molecular genetic basis of the histo-blood group ABO

    system. Nature, 345, 229233.

    Yamamoto, F., Marken, J., Tsuji, T., White, T., Clausen, H. & Hako-

    mori, S. (1990b) Cloning and characterization of DNA com-

    plementary to human UDP-GalNAc: Fuc-alpha1-2Gal alpha1-

    3GalNAc transferase (histo-blood group A transferase) mRNA.

    Journal of Biological Chemistry, 265, 11461151.

    Yamamoto, F., McNeill, P.D. & Hakomori, S. (1992) Human histo-

    blood group A2 transferase coded by A2 allele, one of the A subtypes,

    is characterized by a single base deletion in the coding sequence,

    which results in an additional domain at the carboxyl terminal.

    Biochemical and Biophysical Research Communications,187,366374.

    Yazdanbakhsh, K., Lee, S., Yu, Q. & Reid, M.E. (1999) Identification of

    a defect in the intracellular trafficking of a Kell blood group variant.

    Blood, 94, 310318.Ye, T.Z., Gordon, C.T., Lai, Y.H., Fujiwara, Y., Peters, L.L., Perkins,

    A.C. & Chui, D.H. (2000) Ermap, a gene coding for a novel ery-

    throid specific adhesion/receptor membrane protein. Gene, 242,

    337345.

    Yu, C.Y. (1991) The complete exon-intron structure of a human

    complement component C4A gene. DNA sequences, polymorphism,

    and linkage to the 21-hydroxylase gene. Journal of Immunology,146,

    10571066.

    Yu, C.Y., Belt, K.T., Giles, C.M., Campbell, R.D. & Porter, R.R. (1986)

    Structural basis of the polymorphism of human complement com-

    Review

    770 2004 Blackwell Publishing Ltd, British Journal of Haematology, 126, 759771

  • 8/9/2019 Genetic Basis of Blood Group Diversity - Storry - 2004 - British Journal of Haematology - Wiley Online Library

    13/13

    ponents C4A and C4B: gene size, reactivity and antigenicity. The

    EMBO Journal, 5, 28732881.

    Yu, L.C., Chang, C.Y., Twu, Y.C. & Lin, M. (2000) Human histo-blood

    group ABO glycosyltransferase genes: different enhancer structures

    with different transcriptional activities. Biochemical and Biophysical

    Research Communications, 273, 459466.

    Zimmerman, P.A., Woolley, I., Masinde, G.L., Miller, S.M., McNa-

    mara, D.T., Hazlett, F., Mgone, C.S., Alpers, M.P., Genton, B., Bo-

    atin, B.A. & Kazura, J.W. (1999) Emergence of FY*A(null) in a

    Plasmodium vivax-endemic region of Papua New Guinea .Proceedings

    of the National Academy of Sciences of the United State of America,

    96, 1397313977.

    Review

    2004 Blackwell Publishing Ltd, British Journal of Haematology, 126, 759771 771