m4440090_27_2_11 Immunohematology

48
7/23/2019 m4440090_27_2_11 Immunohematology http://slidepdf.com/reader/full/m444009027211-immunohematology 1/48  V OLUME  27, NUMBER  2, 2011

Transcript of m4440090_27_2_11 Immunohematology

Page 1: m4440090_27_2_11 Immunohematology

7/23/2019 m4440090_27_2_11 Immunohematology

http://slidepdf.com/reader/full/m444009027211-immunohematology 1/48

 V O L U M E 27, NU M B E R   2, 2011

Page 2: m4440090_27_2_11 Immunohematology

7/23/2019 m4440090_27_2_11 Immunohematology

http://slidepdf.com/reader/full/m444009027211-immunohematology 2/48

Page 3: m4440090_27_2_11 Immunohematology

7/23/2019 m4440090_27_2_11 Immunohematology

http://slidepdf.com/reader/full/m444009027211-immunohematology 3/48

75ADVERTISEMENTS

79INSTRUCTIONS FOR AUTHORS

Immunohematology  Volume 27, Number 2, 2011

C O N T E N T S

41REV IEW

Scianna: the lucky 13th blood group system

P.A.R. Brunker and W.A. Flegel

68REV IEW

XG: the forgotten blood group system

N.C. Johnson

58CASE REPORT

Possible suppression of fetal erythropoiesis by the Kell bloodgroup antibody anti-Kpa 

M. Tuson, K. Hue-Roye, K. Koval, S. Imlay, R. Desai, G. Garg, E.

Kazem, D. Stockman, J. Hamilton, and M.E. Reid

61REPORT

Challenging dogma: group A donors as “universal plasma” donorsin massive transfusion protocols

E.J. Isaak, K.M. Tchorz, N. Lang, L. Kalal, C. Slapak, G. Khalife, D. Smith,

and M.C. McCarthy 

66REPORT

Prevalence of RHD*DOL and RHDCE*ce(818T) in two populations

C. Halter Hipsky, D.C. Costa, R. Omoto, A. Zanette, L. Castilho, and M.E. Reid

72COMMUNICAT ION

Erratum

 Vol. 27, No. 1, 2011, pp. 14, 16, and 18

73ANNOUNCEMENTS

Page 4: m4440090_27_2_11 Immunohematology

7/23/2019 m4440090_27_2_11 Immunohematology

http://slidepdf.com/reader/full/m444009027211-immunohematology 4/48

 Immunohematology is published quarterly (March, June, September, and December) by the American Red Cross, National Headquarters, Washington, DC 20006

 Immunohematology is indexed and included in Index Medicus and MEDLINE on theMEDLARS system. The contents are also cited in the EBASE/Excerpta Medica and Elsevier

BIOBASE/Current Awareness in Biological Sciences (CABS) databases

The subscription price is $40.00 (U.S.) and $50.00 (foreign) per year

Subscriptions, Change of Address, and Extra Copies

Immunohematology, P.O. Box 40325Philadelphia, PA 19106

Or call (215) 451-4902

 Web site: www.redcross.org/immunohematology

Copyright 2011 by The American National Red CrossISSN 0894-203X

EDITOR- I N-CHIEF

Sandra Nance, MS, MT(ASCP)SBB

Philadelphia, Pennsylvania

MANAGING EDITOR

Cynthia Flickinger, MT(ASCP)SBB

Philadelphia, Pennsylvania

SENIOR MEDICAL EDITOR

Geralyn M. Meny, MD

Philadelphia, Pennsylvania

 TECHNICAL EDITORS

Christine Lomas-Francis, MSc

New York City, New York 

Dawn M. Rumsey, ART (CSMLT)

Glen Allen, Virginia

ASSOCIATE MEDICAL EDITORS

David Moolten, MD

Philadelphia, Pennsylvania

Ralph R. Vassallo, MD

Philadelphia, Pennsylvania

EDITORIAL ASSISTANT

Sheetal Patel

COPY EDITOR

Mary L. Tod

PROOFREADER

Lucy Oppenheim

PRODUCTION ASSISTANT

Marge Manigly 

ELECTRONIC PUBLISHER

Paul Duquette

EDITORIAL BOARD

Patricia Arndt, MT(ASCP)SBBPomona, California

James P. AuBuchon, MDSeattle, Washington

Martha R. Combs, MT(ASCP)SBBDurham, North Carolina

Geoffrey Daniels, PhDBristol, United Kingdom

 Anne F. Eder, MD Washington, Distr ict of Columbia

George Garratty, PhD, FRCPathPomona, California

Brenda J. Grossman, MDSt. Louis, Missouri

Christine Lomas-Francis, MScNew York City, New York 

Gary Moroff, PhDRockville, Maryland

Paul M. Ness, MDBaltimore, Maryland

Joyce Poole, FIBMSBristol, United Kingdom

Mark Popovsky, MDBraintree, Massachusetts

Marion E. Reid, PhD, FIBMSNew York City, New York 

S. Gerald Sandler, MD Washington, Distr ict of Columbia

Jill R. Storry, PhDLund, Sweden

David F. Stroncek, MDBethesda, Maryland

EMERITUS EDITOR

Delores Mallory, MT(ASCP) SBBSupply, North Carolina

ON OUR COVER

Ophelia by John William Waterhouse

Ophelia is a favorite subject of the pre-Raphaelites and, in particular, John William Waterhouse. The

painting, completed in 1910, is one of his more famous. It depicts the doomed daughter of Polonius pickingowers near the river in the moments before she drowns. The red and white blossoms represent both the

 vitality and the transience of life, as well the curse and fate of lost innocence. The scene is pastoral, but th

darker hues and the intensity in her gaze and in how tightly she clutches the owers and her dress betray

her true state of mind. Ophelia mirrors Hamlet in her brooding over the moral collapse around her but is

unable to resist the descent into madness.

 Like sweet bells jangled, out of tune and harsh;

That unmatch’d form and feature of blown youth

 Blasted with ecstasy: O, woe is me,

To have seen what I have seen, see what I see!

The XG blood group, reviewed in this issue, has been investigated in genetic marker studies suggesting

possible linkage with manic-depressive illness.

  D AVI D MOOLTEN, MD

Page 5: m4440090_27_2_11 Immunohematology

7/23/2019 m4440090_27_2_11 Immunohematology

http://slidepdf.com/reader/full/m444009027211-immunohematology 5/48IMMUNOHEMATOLOGY, Volume 27, Number 2, 2011 41

Scianna: the lucky 13th blood group systemP.A.R. Brunker and W.A. Flegel

The Scianna system was named in 1974 when it was appreciatedthat two antibodies described in 1962 in fact identied antitheticalantigens. However, it was not until 2003 that the protein on which antigens of this system are found and the rst molecular

 variants were described. Scianna was the last previouslyserologically dened, protein-based blood group system to becharacterized at the molecular level, marking the end of an erain immunohematology. This story highlights the critical rolethat availability of laboratory reagents for serologic testing hasplayed in the initial characterization of a blood group and sets thestage for the development of new reagents, such as recombinantproteins, to assist in this process. The central role that genetics

has played, both by classical pedigree analysis and by moleculartechniques, in the discovery and characterization of this bloodgroup is reviewed.  Immunohematology 2011;27:41–57.

The high- and low-prevalence antigens that constitute

the Scianna (SC) blood group system are caused by

 variants in the erythroid membrane–associated protein

(ERMAP).1 SC was initially identied by serologic methods;

the clinical signicance of antibodies specic to SC is

uncertain, although case reports demonstrating rare cases

of hemolytic disease attributed to SC variants exist. Genetic

analyses in both the classical and molecular approaches,

have been central to the discovery and elaboration of theSC system. This article reviews the story of the SC blood

group from a genetic point of view, emphasizing the way it

has been brought into focus thanks to genetic tools ranging

from pedigree analysis to physical mapping.

History

Nomenclature: Sc1, Sc2, Sc3, and Sc4

The story of the 13th International Society of Blood

Transfusion (ISBT) blood group system began in 1962,

 when a new high-prevalence antigen was reported alongside

a coexisting anti-D in a 25-year-old, multiparous woman

of Italian descent in Miami, Florida, who experienced

several fetal deaths as a result of hemolytic disease of the

fetus and newborn (HDFN).2 She came to clinical attention

 because of difculty obtaining compatible blood. Her ABO

and Rh typings were O ccddee, and her husband’s were O

CCDee. After an unremarkable rst pregnancy and birth,

she experienced three subsequent and progressively earlier

fetal demises—at term, at 7, and at 6 months’ gestation—

in the late 1950s. After her second fetal death, her anti-D

titer was demonstrated at 256, and the new antibody to a

high-prevalence antigen, originally named anti-Sm, was

demonstrated at a titer of 16. An informative family study

revealed three antigen-negative siblings with a likely

autosomal dominant mode of antigen inheritance, and no

unrelated antigen-negative specimens were identied in a

population survey of 600 D– random individuals. A clue

to the genetic position of the responsible locus was present

even in this dening family: based on the pedigree, it could

not be determined whether the new antigen was part of the

Rh system as it was in linkage disequilibrium with cc inthat kindred.

In spite of this very dramatic introduction, the clinica

importance of the new antigen was uncertain, as the

concurrent anti-D clearly could account for the proband’s

unfortunate obstetric history. While the work of the Miam

group was in the pipeline for publication, the Winnipeg Rh

Laboratory, in Manitoba, reported an antibody to a new

low-prevalence antigen arising in a 50-year-old man with

stomach cancer.3  In this patient, the antibody originally

named anti-Bua, found in serum Char., was identied during

a routine pretransfusion crossmatch. As the patient had

 been transfused with three units of blood 14 days earlierthis delayed serologic transfusion reaction was investigated

 which revealed that although his serum was crossmatch

compatible with all three donor samples before transfusion

it reacted with one of the three samples after transfusion

 A follow-up survey of 18 panel red blood cells (RBCs

demonstrated one reactive cell, suggesting a relatively high

prevalence for this new antigen; however, this proved not to

 be the case, as only one of the next 1,000 donors was positive

The families of all three of these probands took part in

pedigree analysis, one of which was extremely informative

 with a kindred of both parents and nine offspring. These

studies in classical genetics demonstrated that the new

locus segregated independently from ABO, MNSs, P, Rh

Kell, Kidd, Duffy, and X-chromosome.

Genetics and Inheritance

It did not take long for the relationship between the Sm

and Bua to be postulated, tested, and proven. In 1964, the

anti-Bua serum was used to type the available members of

the index Sm family (Fig. 1). The importance of using this

REVIEW

Page 6: m4440090_27_2_11 Immunohematology

7/23/2019 m4440090_27_2_11 Immunohematology

http://slidepdf.com/reader/full/m444009027211-immunohematology 6/4842  IMMU NOHEM ATOLOGY, Volum e 27, Numb er 2, 2011

P.A.R. Brunker and W.A. Flegel

serum as a typing reagent is underscored by

the fact that it was required to demonstratethat the parent generation consists of

a mating of two Sm/Bua  heterozygotes

(parents PM Sr. and RM): the F1 generation

consists of four Sm– homozygotes, one

Sm/Bua  heterozygote (individual AM),

and one Bua/Bua  homozygote (individual

CS). Without it, the zygosities of AM

and CS could not be determined. This is

the only outbred family in the Sm/Bua 

literature in which both parents are Sm/

Bua heterozygotes.

Concurrent with their suggestion

that Sm and Bua  were the result of a

 biallelic polymorphism, the Winnipeg Rh

Laboratory also reported an extensive

study of a Mennonite population in whom

the Bua antigen had a considerably higher

frequency of 5 in 348 samples than the

1 in 1,000 prevalence observed in other

Caucasian populations.4,5  Although they

tested 145 Caucasian families for Bua  and

rigorously examined 19 based on the presence of Bua, it was

not until additional reagent serum was available in 1966

that denitive proof of this relationship was found.

Further examples of anti-Bua  were discovered in a

group of individuals from Poland and the United Kingdom

(particularly the strongest serum Soch.) who had produced

anti-Bua after having been articially immunized with D+

cells to stimulate the production of Rh antibodies.6 Through

a careful study of the donors used for these stimulations

three additional subjects who were also stimulated with

these cells were found to have created an anti-Bua, although

it was much weaker than that in the  Soch.  serum. The

prevalence of the Bua allele was determined as 0.88 percent

in a Warsaw and 0.67 percent in a London cohort.

Using this reagent after adsorption to remove the

iatrogenically generated anti-D also present in the serum

the Winnipeg Rh Laboratory identied a large, six-

generation Mennonite kindred that included two cousinmatings of Sm/Bua  heterozygotes, which produced 20

offspring.7  Through a methodical review of the serologic

data from samples from the kindred, which showed the

effects of antibody dosage on antigen expression, this report

conrmed and expanded the denition of the new system

to exclude all blood groups reported before 1962 (except

Diego, Yt, and Auberger) and Doa  and Csa. Independence

from Yt was established in two British families.8,9 Finally

the currently accepted nomenclature (Table 1) was proposed

in 1974 to reect the surname of the index family in the

Table 1. Alleles, encoded antigens, and ISBT terminology of the Scianna blood groupsystem

Wild-type Variant

Antigen Trivial n ame Allele Genotype

High-prevalence

antigen Allele Genotype

Low-prevalence

antigen

Sc1 Sca (Sm)   SC*01 169g57GlySc1

Sc2 Scb (Bua)   SC*02 169a57ArgSc2

Sc3 Sc null   SC*03N.01† 307D2Frameshift:

null

Sc3 Sc null 994c 332Arg   SC*03N.02† 994t 332Stop:null

Sc4 Rd 178c60ProRd–

  SC*04 178g60AlaRd+

Sc5 STAR 139g47GluSTAR+

  SC*05 139a47LysSTAR–

Sc6 SCER 242g81Arg

SCER+  SC*06  242a

81GlnSCER–

Sc7 SCAN 103g35Gly

SCAN+  SC*07 103a

35SerSCAN–

†Provisional name suggested by the International Society of Blood Transfusion (ISBT) WorkingParty on Red Cell Immunogenetics and Blood Group Terminology

Fig. 1  Index family in the characterization of the Sm antigen anddemonstration of the antithetical relationship between Sm and Bu a antigens. The proband (patient Ms. Scianna) is indicated by thearrow. Solid color represents Sm+ (Sc:1+) antigen test. Striped fillrepresents Bua+ (Sc:2+) antigen test. Inferred genotype is shownbelow the symbol for each family member. The proband’s parentswere deceased at the time of Bua  testing, so are inferred to beheterozygotes, indicated by the interrupted stripes and parenthesesin their genotype designation. The combination of Sm and Bua typingconfirms that subject AM is a heterozygote, which was suggestedby observations of dosage effects in serologic testing. (Redrawnwith data from references 2 and 4.)

Page 7: m4440090_27_2_11 Immunohematology

7/23/2019 m4440090_27_2_11 Immunohematology

http://slidepdf.com/reader/full/m444009027211-immunohematology 7/48IMMUNOHEMATOLOGY, Volume 27, Number 2, 2011 43

Scianna blood group system: a review

discovery of anti-Sm, Scianna,10 such that Sm was renamed

Sc1 and Bua was renamed Sc2.

The  SC  Gene Lies on Chromosome 1

The focus then shifted from denition of the system

to characterization of the locus responsible for the antigen

at a chromosomal level. Much of this seminal work was

performed in the Winnipeg Rh Laboratory. So it is not

surprising that their detailed investigations of the genetic

mapping of chromosome 1 contributed signicantly to

the progress on SC. Their unique access to informative

kindreds certainly hastened this process, and in a series

of reports from 1976 to 1978, linkage between  RH  and  SC  

 was established, showing a logarithm of the odds ratio

(LOD score) of 5.34 at a recombination fraction, θ = 0.10 if

paternally segregated,11 and the relative position of SC  was

determined12–14 and rened.15

 A Third Antigen in the System: Sc3 or Scianna Null

 Alleles

The appearance of “minus-minus” phenotypes, i.e.,

individuals whose RBCs tested negative for both Sc1 and the

antithetical Sc2 antigen (Sc:–1,–2), was rst documented

in 1973 and demonstrated the existence of apparent  SC  

null alleles.16 However, the term Sc3 was not coined until

1980 when an antibody from an Sc:–1,–2 individual

demonstrated no evidence of a separable anti-Sc1 or anti-

Sc2.17  The index patient examined in 1973 was a female

surgical patient from the Likiep Atoll in the Marshall

Islands, who had been transfused 7 months earlier withoutcrossmatching difculty. Her cells phenotyped as Sc:–1,–

2 as did those from one cousin, but both women’s RBCs

could still adsorb anti-Sc2. Despite four pregnancies with

an Sc:1,–2 husband, the proband’s cousin had a negative

antibody screen. Because these RBCs reduced the titer of

anti-Sc2, but not anti-Sc1, they may have had some weak

Sc2 expression, and a new antigen was not denitively

characterized at that time.

However, in 1980, a 67-year-old man in New York being

treated with chemoradiotherapy for metastasis to the throat

of a carcinoma of unknown origin required a preoperative

transfusion, and demonstrated a positive antibody screen.17 

He had been transfused 4 years earlier without event.

Unlike the Likiepian Sc:–1,–2 erythrocytes, RBCs from this

patient did not reduce the titer of either anti-Sc1 or anti-

Sc2. When tested against the Likiepian Sc:–1,–2 cells, the

New York serum did not agglutinate the RBCs. No other

Sc:–1,–2 cells were found in a family study. No separable

anti-Sc1 was present in this patient’s serum, which is

somewhat unusual because patients who completely lack all

known antigens for a blood group and have been transfused

typically make a polyclonal mixture of antibodies directed

at the common epitopes of that system.18

The next report of a null phenotype for SC was described

in 1986, in another Pacic Islander.19  This patient was a

4-year-old girl from Papua New Guinea with thalassemia

major who had been transfused many times. Anti-Sc3 was

demonstrated. Erythrocytes from the patient’s mother

 were compatible, and both mother and daughter were

found to have the Sc:–1,–2 phenotype. In this community

the Sc:–1,–2 phenotype was surprisingly common: A survey

of 29 family members and apparently unrelated villagers

revealed 6 others (2 of whom had no obvious relationship

to the patient) who were also Sc:–1,–2. This very high

phenotype prevalence (20.6%) makes heterozygosity for

a putative recessive  Sc3 allele in the patient’s father quite

likely (although his RBC phenotype is not reported in the

short abstract), as well as explaining the homozygosity for

the same allele observed in her mother. Additional Sc:–1,–2 individuals have been reported

(including a patient who experienced an apparent delayed

hemolytic transfusion reaction), some of whom have also

lacked several other high-prevalence antigens.18  Evidence

that each patient in their series produced antibodies with

different specicities is discussed in Antibodies section.

Radin: A “New” Low-Prevalence Antigen Is Linked to

Scianna (via RH)

 When Radin was rst described, in 1967,20  the

relationship between Radin and Scianna was not

appreciated. Although the clinical signicance of antibodiesgenerated to antigens within the SC system in genera

is controversial, the analysis of the antibodies that led to

the discovery of the Radin antigen is based on its clinical

importance, with the initial description encompassing

ve cases of mild to moderate hemolytic disease of the

newborn, of which one required exchange transfusion and

one appeared in the rst pregnancy.20  Additional cases

and conrmation of the low general frequency (1 in 205)

appeared shortly thereafter.21

Once again, it was the unique analysis performed by

the Winnipeg Rh Laboratory that elucidated the putative

connection between Radin and SC. Linkage analysis

of eight propositi in ten nuclear families demonstrated

linkage between  Rd   and  RH.22  Similar to the  Sc1/Sc2

linkage analysis in the present dataset, the Winnipeg

analysis demonstrated heterochiasmy, with the paterna

LOD score exceeding 3 at a recombination fraction, θ  =

0.10, whereas the corresponding maternal LOD neither

suggests nor refutes linkage (LOD = 0.41 at θ  = 0.10 )

This nding was only sufcient to propose the connection

 between SC  and Rd, as heterozygotes at both of these (i.e.

Page 8: m4440090_27_2_11 Immunohematology

7/23/2019 m4440090_27_2_11 Immunohematology

http://slidepdf.com/reader/full/m444009027211-immunohematology 8/4844  IMMU NOHEM ATOLOGY, Volum e 27, Numb er 2, 2011

P.A.R. Brunker and W.A. Flegel

double recombinants) had not been found. Incorporation of

the gene encoding the Rd antigen into the bigger picture of

chromosome 1 mapping placed it so close to  SC  that it was

proposed even at that time that the gene encoding  Rd   “is

either very closely linked to or identical with SC.”23

There were few additional reports on the SC blood group

system for more than 20 years until its molecular basis was

nally resolved in 2003.1  Since then, molecular analysis

has identied the three additional alleles underpinning

the three antibodies described by Devine and coworkers in

1988.18

Molecular Basis

The ERMAP Expresses the SC Antigens

The molecular basis of the SC blood group system was

identied in 2003 by merging the genetic mapping data

 with protein chemistry showing that the SC antigens wereon a single glycoprotein of approximately 60 to 68 kDa

that must be expressed by RBCs.24,25 The SC  gene had been

mapped to chromosome 1, and based on its linkage to the

 RH   genes, the chromosomal location had been further

rened to 1p34 to 1p36. This led to the identication of a

strong candidate gene.1

The ERMAP is a 475-amino acid, type 1 single-pass

membrane glycoprotein that is a member of the butyrophilin

(BTN) family and is encoded by the  ERMAP   gene.26,27  It

consists of a predicted signal sequence of 29 amino acids

at the NH3  terminus, an extracellular immunoglobulin

 V domain (amino acids 50–126), a short transmembranedomain spanning amino acids 157 through 176, and an

intracellular carboxyl terminus encompassing a B30.2

domain from amino acids 238 through 395.1

The nomenclature for the transcripts of  ERMAP   is

complicated and has been revised many times. At least

two transcript variants have been described, which

result from use of an alternative upstream promoter

and alternative splicing.28  The longer variant is 3,423

 bp and is designated as transcript 1, or as transcript b

 by GenBank curators,29  as it was the second transcript

 variant identied, and it includes an additional upstream

exon (GenBank NM_001017922.1). The shorter variant is

designated as transcript 2 or as transcript a, is 3,369 bp,

excludes this upstream exon (GenBank NM_018538.3),

 begins transcription 45 bp upstream from the start of exon

2, and was the rst variant discovered; thus, early reports

describe this gene as consisting of 11 exons. However, the

revised current nomenclature for this gene is based on the

presence of 12 exons spanning approximately 28 kb. The

relative production of these two mRNAs is not known, nor

has it been determined whether the use of either promoter

is favored in some tissue types or physiologic conditions

Both transcripts share the common ATG start codon in

exon 3; thus, there is no predicted difference in the protein

product of the two transcripts. They differ in both the

transcript initiation site and the DNA stretch of exon 2 that

 becomes part of the nal transcript; consequently, there are

two alternative exon 2s, named exon 2a and exon 2b. The

longer transcript 1 comprises exon 1, exon 2b, and exons

3 through 12, whereas the shorter transcript 2 starts with

exon 2a and also includes exons 3 through 12 (Fig. 2).

The Ensembl genome browser curators have delineated

ve transcripts in ERMAP,30,31  three of which are protein

coding and two of which generate a processed transcript only

Two of the Ensembl transcripts correspond to GenBank’s

transcripts 1 and 2; however, unique to the Ensemb

database is a 3,949-bp transcript (named ERMAP-002),3

 which is reported as protein coding and generates a short

protein encoding 385 amino acids rather than the usual475. However, only the two transcripts that encode the

475–amino acid protein are included in the Consensus

Coding Sequence Project (CCDS; both with identication

number CCDS475), so the biological signicance of the

3,949-bp transcript is unclear.

In addition to explaining the  Sc1/Sc2  biallelic

single nucleotide polymorphism (SNP) at Gly57Arg, a

 binucleotide GA deletion was identied at nt307 (now

known as  SC*03N.01,  provisional name suggested by

the ISBT Working Party on Red Cell Immunogenetics

and Blood Group Terminology). This deletion causes a

frameshift and premature stop codon, which explain theSC null phenotype.1  The Rd antigen (Sc4) was dened as

the Pro60Ala variation, and two other variants in the

presumed leader signal peptide (54C>T and 76C>T) were

described in the original study elucidating the molecular

 basis by Wagner and colleagues1 (Table 2).

Understanding the Variants Discovered in the Post-

ERMAP  Era

The knowledge of the gene responsible for the antigens

of this blood group protein opened the oodgates through

 which variant descriptions of unknown, unresolved sero-

logic cases promptly ow. This was clearly the case for SC, for

 which three new variants were discovered within just a few

 years after the characterization of ERMAP  as the SC  gene

Careful molecular follow-up of the three patients described

 by Devine and coworkers18  in 1988 revealed three distinct

 ERMAP/SC   variants, demonstrating the importance of

molecular testing in the resolution of serologic SC mysteries

The success of these investigations depended not only on

the cooperation among an international collaborative, but

also on the supportive participation of two patients and

Page 9: m4440090_27_2_11 Immunohematology

7/23/2019 m4440090_27_2_11 Immunohematology

http://slidepdf.com/reader/full/m444009027211-immunohematology 9/48IMMUNOHEMATOLOGY, Volume 27, Number 2, 2011 45

a family who kindly released two autologous RBC units.

It was appreciated in the initial case reports in 1988 thatthe SC protein carried multiple high-prevalence antigens

other than Sc1/2.18 Sera or eluates from these three Sc:1,–2

patients who had developed high-prevalence antibodies did

not react with the Sc:–1,–2 null RBCs, but the samples were

not mutually compatible.

Sc5 (STAR)

In 1982, a 65-year-old man with a history of transfusion

of three units of crossmatch-compatible whole blood

 before presentation underwent routine preoperative blood

 bank testing, demonstrating anti-C and anti-e. He was

transfused with another three units of C–e– RBCs, and 1

 week later, a new anti-Jk  b and an antibody against a high-

prevalence antigen were detected. Because his serum

reacted with all cells except his own (phenotyped as Sc:1,

–2), those of a sibling, and Sc:–1,–2 RBCs, it was suspected

that he had developed an antibody to another antigen on

the SC protein.35 Blood needs for the patient were met with

autologous units. Blood samples from the proband and 16

family members had been frozen. Sequencing of the exons

encoding the extracellular and transmembrane domains

demonstrated homozygosity at a new nonsynonymous

polymorphism at amino acid 47 (glutamic acid to lysine) which is in the N-terminal domain.36  This variation is

catalogued as rs56047316 in the SNP database (dbSNP), and

it results from the guanine–adenine transition at nucleotide

139 in the complementary DNA (cDNA). Frequencies of this

allele in population studies have not been reported, nor

have additional cases of this antibody.

Sc6 (SCER) and Sc7 (SCAN)

The remaining two antibody cases described by Devine

and colleagues18  were concomitantly resolved using the

same approach as for the Sc5 antigen.37 These investigators

sequenced all 11 exons of  ERMAP   known at the time

 which encompasses all cDNA. They also solicited others to

submit suspected SC variants and the eight other orphan

low-prevalence antigens By, Toa, Pta, Rea, Jea, Lia, SARA

and Sk a. The proband for case 1 reported by Devine and

colleagues18  demonstrated homozygosity for a guanine–

adenine transition at cDNA nucleotide 242, predicted to

cause an amino acid change at position 81 (from arginine

to glutamine), which is in the immunoglobulin V loop.1

The case 2 proband demonstrated a new homozygous

Scianna blood group system: a review

Fig. 2 Transcripts, exon structure, and variants of erythroid membrane-associated protein (ERMAP ). Codon numbers are indicated aboveamino acids involved in described variants. The immunoglobulin variable domain is underlined. Exon 4 is expanded to the nucleotide levein the gray box to show the locations of most ISBT-recognized Scianna polymorphisms. The wild-type reference sequence is under thecorresponding amino acid and is the chromosome 1 reference sequence GenBank NC_000001.10. * = nucleotide deletion.

Page 10: m4440090_27_2_11 Immunohematology

7/23/2019 m4440090_27_2_11 Immunohematology

http://slidepdf.com/reader/full/m444009027211-immunohematology 10/4846  IMMU NOHEM ATOLOGY, Volum e 27, Numb er 2, 2011

P.A.R. Brunker and W.A. Flegel

Table 2. Erythroid membrane–associated protein polymorphisms in the coding region and global population frequencies

Sc name Descriptive name dbSNP ID: cDNA nt mRNA* Wild-type (WT) Variant Protein effectAmino acid (aa)

codon

rs35757049 11 281 c t nonsynonymous substitution 4

rs33950227 54 324 c t silent 18

rs33953680 76 346 c t nonsynonymous substitution 26

Sc7 SCAN 103 373 g a nonsynonymous substitution 35

Sc5 STAR rs56047316 139 409 g a nonsynonymous substitution 47

Sc1 vs. Sc2 rs56025238 169 439 g (Sc1)a

(Sc2)nonsynonymous substitution 57

Sc4 Rd rs56136737 178 448 c g nonsynonymous substitution 60

rs33954154 219 489 c t silent 73

Sc6 SCER 242 512 g a nonsynonymous substitution 81

Sc null (D ga together) rs55695242 307 577 g del –1 frameshift 103

Sc null (D ga together) rs56151267 308 578 a del –1 frameshift 103

rs35147822 775 1045 t c nonsynonymous substitution 259

rs34441268 788 1058 g a nonsynonymous substitution 263

rs35972628 888 1158 g a silent 296

Sc null 994 1264 c t nonsense (STOP) 332

rs55773259 1227 1497 a g silent 409

rs55872827 1324 1594 t c nonsynonymous substitution 442

rs56405033 1355 1625 t c nonsynonymous substitution 452

rs55677363 1356 1626 g t silent 452

*NCBI Reference Sequence Position: NM_001017922.1†Minor allele frequencies for some populations are determined by reports of serologic phenotype and assuming individuals positive for a rare antigen areheterozygotes.

cDNA = complementary DNA; dbSNP = single nucleotide polymorphism database; IgV = immunoglobulin V; mRNA = messenger RNA; nt = nucleotide.

nonsynonymous variant at cDNA nucleotide position 103

(again a guanine–adenine transition), corresponding to

a glycine to serine change at amino acid 35 in the NH3 

terminus. This specimen also demonstrated two other SNPs

(at cDNA nucleotides 54C>T and 76C>T), which had been

previously reported, are common in Caucasian populations,

and are in tight linkage disequilibrium (LD).1 Because these

 variants are both in the predicted leader sequence (and the

54C>T is a silent mutation), they are not predicted to directly

impact on ERMAP structure. This report also excluded the

eight orphan low-prevalence antigens from the SC system

 because the only variants found were in the leader sequence

or in introns.

Molecular Basis of SC Null Phenotypes

Central to the set of specimens originally reported with

the discovery of ERMAP as SC was an Sc:–1,–2 sample

from a Saudi Arabian pedigree,1  which demonstrated

homozygosity at three SNPs: the common 54C>T and

76C>T (described in an earlier section) as well as a 2-bp

deletion starting at cDNA nucleotide 307 (307Δga) that

is predicted to cause a frameshift mutation and early

termination codon after 113 amino acids. The nt54 and nt76

SNPs were genotyped in an additional 111 European blood

donors and were found in tight LD. The frameshift would

account for a complete lack of an intact ERMAP protein in

the cell membrane.

Two Sc:–1,–2 individuals from northern and southern

California were then sequenced in a follow-up study,37 and

 both shared a new  SC   null allele formed by a nonsense

mutation at codon 332. This is the rst report of a variant

in the B30.2 intracellular domain in a patient immunized

to SC. It is not, however, the only variant in the B30.2

domain (Table 2), because three others have been described

Page 11: m4440090_27_2_11 Immunohematology

7/23/2019 m4440090_27_2_11 Immunohematology

http://slidepdf.com/reader/full/m444009027211-immunohematology 11/48IMMUNOHEMATOLOGY, Volume 27, Number 2, 2011 47

in dbSNP, two of which are nonsynonymous amino acid

changes (C259R and G263E) and one of which is silent

(E296E). This result suggests that an early translation

termination, even as late as at codon 332 of the expected

475, sufciently interferes with correct protein trafcking,

membrane insertion, or stability so as to render the

individual susceptible to alloimmunization at the Sc1/2 site

(codon 57).

Other Variants in ERMAP : Using dbSNP and HapMap

 Variants in the coding region, particularly those in the

extracellular domain of transmembrane proteins, are of

particular clinical interest in transfusion medicine, as their

location provides an obvious mechanism for putative clinical

signicance by alloimmunization. However, much variation

in the human genome is found outside of coding regions,

and ERMAP  is no exception. Studies of this variation are a

critical tool in the investigation of human disease and basic

sciences. Several collaborative projects in human genomics

focus on gathering, sharing, and interpreting human

genetic variation, and although a full cataloguing of these is

 beyond the scope of this review, we will present variants in

 ERMAP  that are part of two such projects: dbSNP38 and the

International HapMap Project.39 Table 2 integrates some of

these variants (identied by their rs numbers) with those

described in the transfusion medicine literature. These

data allow informative LD studies of ERMAP  (Fig. 3). This

LD map shows two distinct LD blocks that correspond to

the exons encoding the extracellular immunoglobulin-like

domains and the intracellular B30.2 domains of ERMAP

The importance of this pattern of LD is that it reveals a

possible evolutionary vestige of the modular nature of the

 butyrophilin-like (BTNL) protein family and is even more

pronounced in the Nigerian population. Long stretches of

Scianna blood group system: a review

WT aa Variant aa Exon Protein domain Minor allele frequency in blood donors†

Ala Val 3 Leader 0.054 (dbSNP)

Leu Leu 3 Leader 0.28 (German1), 0.1 (dbSNP)

His Tyr 3 Leader0.33 (German1), 0.25 (Caucasian North American32), 0.1 (dbSNP),

0.05 (African-American32)

Gly Ser 4 NH3-terminus

Glu Lys 4 NH3-terminus

Gly Arg 4 IgV loop0.01 (Caucasian North American,33 Brazilian34), 0.008 (German1),

0.005 (Hispanic American33), 0.002 (African-American33), 0 (Asian American33)

Pro Ala 4 IgV loop0.003 (Slavs33), 0.002 (Caucasian North American,33 Danish,21 German,1 

New York Jewish20)

Arg Arg 4 IgV loop 0.017 (dbSNP)

Arg Gln 4 IgV loop

Asp frameshift 4 IgV loop

Asp frameshift 4 IgV loop

Cys Arg 12 B30.2 domain 0.022 (dbSNP)

Gly Glu 12 B30.2 domain 0.011 (dbSNP)

Glu Glu 12 B30.2 domain 0.011 (dbSNP)

Arg STOP 12 B30.2 domain

Leu Leu 12   C-terminus

Ser Pro 12   C-terminus

Leu Pro 12  C-terminus at a 3’ dileucine (LL)

phosphorylation motif

Leu Leu 12  C-terminus at a 3’ dileucine (LL)

phosphorylation motif0.054 (dbSNP)

Page 12: m4440090_27_2_11 Immunohematology

7/23/2019 m4440090_27_2_11 Immunohematology

http://slidepdf.com/reader/full/m444009027211-immunohematology 12/4848  IMMU NOHEM ATOLOGY, Volum e 27, Numb er 2, 2011

P.A.R. Brunker and W.A. Flegel

LD such as that illustrated in the extracellular domain of

 ERMAP, where all of the SC antigens are encoded, raise the

possibility that a selective advantage by genetic hitchhiking

may be operating at ERMAP.40

Global Variation

Some ethnographic trends have already been

historically appreciated with SC variants during the early

 years of their investigation (Table 2). For instance, Sc4 has

 been identied most often in Ashkenazi Jews and Slavic

populations, and the few Sc:–1,–2 phenotypes have been

reported particularly in populations from Oceania. Sc2

appears to be more common in a consanguineous Canadian

Mennonite population (derived from a small region in

Eastern Europe),5 but it remains very rare in or absent from

populations of African5,33  or Native American10  descent.

However, it is not sufcient to rely on case reports to

propose generalizations regarding population frequencies.

Fortunately, recent technologies are available to rapidly

genotype  ERMAP  variants using an automated genotype-

calling platform, and additional population studies wil

continue to be reported.33

Biochemistry and Physiology

ERMAP Is a Member of the Butyrophilin-like Family of

the Immunoglobulin Superfamily

By virtue of its extracellular immunoglobulin V and

the intracellular B30.2 domains, ERMAP is a member

of the BTNL protein family, which is a subset of the

immunoglobulin superfamily.41  The compact, globular

110–amino acid immunoglobulin domain denes this

superfamily and is found in three operational domain

subclasses: variable (immunoglobulin V), intermediate

(immunoglobulin I), or constant (immunoglobulin C).42,4

These proteins are central to many immunologic processes

especially cell adhesion, costimulation, and signalling.44,4

The immunoglobulin superfamily is one of the largest in

all eukaryotic organisms.46  Other members of this family

 well known to the transfusion medicine community

Fig. 3  Pairwise linkage disequilibrium (LD) heat plot of the logarithmic odds ratio (LOD) score for single nucleotide polymorphisms inerythroid membrane–associated protein (ERMAP) in a Nigerian population. Darker shades (black) = areas of high LD, lighter shades (lightgray) = areas of lower LD. The areas of tightest LD correspond to the extracellular domains of ERMAP, on the left-hand side of the figure.Increased LD is also seen in the intracellular domains, at the right side. This segmental pattern of LD reveals a possible evolutionary vestigeof the modular nature of the butyrophilin-like protein family. YRI = Yoruba in Ibadan, Nigeria. (Source: Haploview™ software run at http://

hapmap.ncbi.nlm.nih.gov/cgi-perl/gbrowse/hapmap3r2_B36/#search; accessed April 27, 2010.)

Page 13: m4440090_27_2_11 Immunohematology

7/23/2019 m4440090_27_2_11 Immunohematology

http://slidepdf.com/reader/full/m444009027211-immunohematology 13/48IMMUNOHEMATOLOGY, Volume 27, Number 2, 2011 49

Scianna blood group system: a review

include the Lutheran, LW, OK, JMH, and Indian blood

groups, which are found on the B-CAM, ICAM4, CD147,

CD108, and CD44 molecules, respectively.47 BTN proteins

are within the B7-CD28-like branch of this superfamily 48 

and have been extensively studied in cows.49 The prototypeof the human BTN family of proteins is BTN1A1, which is

primarily expressed in the lactating breast, where it makes

up 20 percent of the protein in the membrane of milk

fat globules. The name derives from the Greek butyros 

and  philos,  meaning “having an afnity for butterfat.”

 Although  ERMAP   is carried on chromosome 1, the genes

for many members of this family are located in the major

histocompatibility complex (MHC) on chromosome 6.50

The lactational and immunologic functions of BTNs

as a part of the secretory granule-to-plasma membrane

zipper complex and as an inhibitor of T-cell activation are

only recently described,51,52  and even less is known about

the BTNL proteins. There are six known human BTNL

proteins, which are dened by their homology to BTN

proteins (Fig. 4). The most extensively studied of these is

BTNL2, the gene for which is located in the MHC class II

cluster at 6p21.3. A truncating splice site mutation in BTNL2 

has been associated with sarcoidosis,53  and although this

SNP has been investigated in many other infectious and

autoimmune diseases, some of these associations have

 been attributable to LD with the MHC.54  BTNL2 inhibits

T-cell activation by inhibiting proliferation in response to

the stimulatory T-cell receptor signal.55  This inability for

BTNL2 to propagate a cell signal has led some investigators

to propose its role as “decoy receptor” because it lacks the

B30.2 intracellular domain present in most other BTN andBTNL proteins.54

The B30.2 Cytoplasmic Domain: A Role in

Erythropoiesis?

The B30.2 (or PRYSPRY) domain was described in 1993

 with the discovery of an exon in the MHC class I region

showing similarity to other mammalian and amphibian

proteins, which have since been termed the tripartite

motif family.56,57  Such a domain was demonstrated in

 bovine BTN58  and in human BTN proteins.59  The B30.2

domain is proposed to be a recent evolutionary adaptation

in the immune system of mammals as a fusion of two

ancient domains (PRY and SPRY), which are found in

all eukaryotes.50  The genes for proteins in this family

have a modular structure, suggesting that they arose

 by duplication.56  Mutations in the B30.2 domain in the

tripartite motif–branch of the B30.2 proteins have been

associated with Opitz syndrome (MID1 protein) and

familial Mediterranean fever (pyrin protein). However

human syndromes have not been associated with mutations

in the B30.2 domain of the BTN or BTNL families.

Fig. 4  The organization of the butyrophilin-like (BTNL) family incomparison to the butyrophilin (BTN) and B7 families. The B7branch of the immunoglobulin superfamily is characterized byvariable (IgV) and constant (IgC) immunoglobulin domains. TheBTN subfamily includes the B30.2 intracellular domain. The BTNLproteins exhibit various combinations of these features. MOG =myelin oligodendrocyte glycoprotein, an autoantigen implicated inmultiple sclerosis. (Modified from reference 52.)

Fig.5 On the left, blood from a fish of the genus Notothenia  (withhemoglobin-containing erythrocytes) and on the right is bloodfrom Chaenocephalus aceratus.  Icefish transport oxygen strictly inphysical solution, without a carrier molecule. The recent discoverythat bloodthirsty (bty),  a B30.2-domain–containing protein likeERMAP, has dramatically decreased expression in hemoglobinlessicefish provides a fascinating link between this protein domain anderythropoiesis. Courtesy of Professor Guillaume Lecointre.

Page 14: m4440090_27_2_11 Immunohematology

7/23/2019 m4440090_27_2_11 Immunohematology

http://slidepdf.com/reader/full/m444009027211-immunohematology 14/4850  IMMU NOHEM ATOLOGY, Volum e 27, Numb er 2, 2011

P.A.R. Brunker and W.A. Flegel

The crystal structure of the B30.2 domain of pyrin was

recently elucidated. A β-barrel consisting of two antiparallel

β-sheets has been described, forming a central cavity.60 The

ligand(s) that interact with the B30.2 domain have yet to be

described, although binding analyses suggest that it is the

site of protein–protein interactions.61 The crystal structure

of one other B30.2 domain interacting with a peptide has

shown that there is a conformationally r igid peptide binding

pocket (consisting of a core β-sandwich around variable

loops) binding to a short-sequence motif, which may allow

multiple intracellular targets to bind.62  Recently, BTN1A1

has been shown to bind xanthine oxidoreductase via B30.2,

apparently stabilizing the milk fat globule membrane in

mammary tissue, but it is also hypothesized to function

as a novel signaling pathway in nonmammary tissues or

perhaps in the innate immune system via generation of

reactive oxygen species.63

 A role for B30.2 domains in erythropoiesis has beenrevealed through study of an unlikely model organism:

 Antarctic icesh. These animals are the only vertebrate

taxon that fails to produce RBCs (Fig. 5), and as such

have been studied in a hunt for genes important in

erythropoiesis and cardiovascular biology.64  Using

this approach, a new B30.2-containing protein, called

bloodthirsty (bty), was identied in the pronephric kidney

of a red-blooded Antarctic rockcod ( Notothenia coriiceps),

 which was present at levels tenfold higher than those in

an icesh (Chaenocephalus aceratus).65  Interestingly,

disruption of bty  synthesis in zebrash suppressed both

erythrocyte production and hemoglobin synthesis, andalthough interactions between bty and ERMAP  have been

hypothesized, they have not been empirically demonstrated

(H. William Detrich, personal communication, January

20, 2010).

Functional Studies of ERMAP 

The function of  ERMAP   itself, however, is not well

understood. The murine homolog, Ermap, was described

rst and named as such because it was found to be produced

exclusively in erythroid cells.66  The human homolog was

characterized shortly thereafter, and Northern blots

demonstrated high expression in hematopoietic tissues,

such as fetal liver and bone marrow, with weaker expression

in peripheral blood leukocytes, thymus, lymph node,

and spleen.26  Sc1 has been demonstrated on phagocytic

leukocytes using an antibody absorption technique.67 

Recent RNA array expression analysis supports this

nding, having detected low levels of  ERMAP   transcripts

in leukocytes, especially monocytes; however, quantitative

protein expression remains to be assessed.54  An in silico

analysis of nucleotide database searches of human

expressed-sequence tags using the  ERMAP   transcript 1

(GenBank NM_001017922.1) as the probe detected the

transcript in cDNA libraries from hematogenous tissues

such as bone marrow, a chronic myeloid leukemia cell line

peripheral blood pool, thymus, spleen, and fetal liver, as wel

as in neural tissues.68 However, it is difcult to denitively

ascribe transcript production to the nonerythroid cells in

the neural tissues, as contamination from the vasculature

cannot be completely eliminated.  ERMAP  mRNA reaches

peak levels in fetal liver in the 18th through 20th weeks

and it is present from the 15th through 32nd weeks in feta

 bone marrow, suggesting that ERMAP may be related

to the migration of erythroid cells to these sites during

hematopoietic development.69

 Additional evidence that ERMAP may be involved in

erythroid differentiation has been put forward, although

only abstracts of these reports are available in the English

literature. Using uorescent quantitative polymerasechain reaction,70  ERMAP expression has been found in

the K562 cell line, which is derived from chronic myeloid

leukemia and is of undifferentiated granulocytic lineage.7

To test the degree of erythroid-specicity of ERMAP

in hematopoiesis, this group used cytarabine (Ara-C)

to induce these cells toward erythroid differentiation

and 12-O-tetradecanoylphorbol-13-acetate to induce

development toward the macrophage lineage, but found

an increase in  ERMAP  mRNA after an Ara-C stimulation

only.72 This nding suggests that although ERMAP may be

present on cells of the monocyte/macrophage lineage, its

functional importance may be limited in these cell typesRNA silencing experiments using an ERMAP shRNA/K562

cell line also reported by this group showed decreased

 ERMAP   expression and morphologic features, including

the relative amounts of surface erythrocyte maturation

markers, leading the authors to conclude that ERMAP

shRNA inhibited Ara-C–induced erythroid differentiation.7

 A second model of erythroid differentiation using umbilical

cord blood with two naturally occurring hormones (namely

stem cell factor and IL-3) and erythropoietin to provoke

differentiation also showed a concurrent rise in  ERMAP

mRNA with erythrocyte maturation.74

Many structural features of ERMAP point toward a

putative role in immunity, perhaps by adhering to other

cells or pathogens via its extracellular immunoglobulin V

domain, or by immunoregulatory mechanisms such as the

modulation of cellular activation signals via B30.2 as is

observed in its BTN family members.52 Much work is needed

in this area to better characterize the proteins that interact

 with ERMAP, both extracellularly and intracellularly

and to determine whether ERMAP has a central role in

erythropoiesis itself.

Page 15: m4440090_27_2_11 Immunohematology

7/23/2019 m4440090_27_2_11 Immunohematology

http://slidepdf.com/reader/full/m444009027211-immunohematology 15/48IMMUNOHEMATOLOGY, Volume 27, Number 2, 2011 51

Scianna blood group system: a review

 Antibodies in the System

The discovery of the SC blood group system began

 with and has relied on the detection and investigation of

alloantibodies. The effects of enzymes and chemicals and

the in vitro characteristics of SC antibodies were recently

reported elsewhere.75 In general, SC antigens are resistant

to cin plus papain, trypsin, α-chymotrypsin, sialidase,

and 50 mM dithiothreitol (DTT), and they are sensitive to

pronase and 200 mM DTT. The exception to this trend is the

 variable sensitivity of Sc4 to trypsin and α-chymotrypsin.

Enzymatic properties of anti-SCER and anti-SCAN are only

described in the initial case report as showing no change

in the strength of antibody activity when tested with RBCs

that had been treated with cin, papain, trypsin, ZZAP

(mixture of 0.1 M DTT plus 0.1% cysteine-activated papain),

or chloroquine diphosphate.18  Anti-STAR demonstrated

enhanced reactivity with enzyme-treated RBCs.36

 Patientsor donors in whom an antibody to an SC antigen has been

reported (Table 3) have been thoroughly reviewed recently.76 

Thirteen of the 19 reports of alloantibodies to SC system

antigens listed in Table 3 have occurred in cases in which

they do not appear to manifest a major clinical signicance.

 All six of the reports with clinical signicance occurred in

cases of HDFN. All six of the reported autoantibodies to SC

system antigens in patients were in the context of clinically

signicant autoimmune hemolytic anemia, although ve

of these cases were presented as an abstract only, without

the full clinical details. Alleles  Sc4–7   were not tested in

any of these cases. Such an investigation is warranted because heterozygosity for alleles with weak expression

in the Rh blood group system has been associated with D

autoantibodies.86

Clinically important reactions to an erythrocyte

antibody typically result from signicant RBC destruction

and usually manifest in the patient as a hemolytic anemia or

HDFN. In the SC system, signicant HDFN dened the blood

group in a patient named Ms. Scianna; however, she also

had anti-D to account for her severe obstetric complications

in addition to the anti-Sc1.2 Anti-Sc2 has been reported in

a case of HDFN requiring simple transfusion in a 20-day-

old infant.83 Importantly, the search for antibodies to low-

prevalence antigens in this patient was only performed

 because the neonate and the mother were ABO-compatible:

had they been ABO-incompatible, the HDFN would likely

have been attributed to this, and the anti-Sc2 antibody may

not have been detected.

The recent availability of recombinant reagents to

assist in the detection of SC antibodies87  may bring this

ability into the wider immunohematology arena. This novel

technique will allow one to appraise the relevance of SC

antibodies in conjunction with other antibodies and tease

out their relative clinical importance, which may have been

previously underestimated. The ERMAP protein has been

expressed in its native form in a eukaryotic system,87 and

so although the initial report describes the utility of this

reagent for detecting the high-prevalence SC antigens, a

screening protein for low-prevalence antigens Sc2 and Sc4

 would also be feasible.

Some SC alloantibodies (Table 3) resulted in delayed

serologic transfusion reactions without associated

hemolysis, including a so-called naturally occurring anti-

Sc4 described in a 55-year-old man without a transfusion

history.21  Many of these patients were transfused with

crossmatch-compatible blood without incident. However

some surgeries were delayed owing to lack of availability

of crossmatch-compatible units, and other patients

 were transfused with autologous products in nonurgent

scenarios.18

The remaining reports of important reactions involving

SC antibodies are in cases of autoimmune hemolytic

anemia. Auto-anti-Sc1 and -Sc3 have been eluted from

direct antiglobulin test–positive RBCs of these patients. In

many cases, these antibodies were transient and associated

 with decreased expression of SC antigens.79,84  Severa

patients underwent splenectomy in addition to treatment

 with many types of immunosuppressant medications. One

patient with erythroid hypoplasia and diagnosed with an

Evans-like syndrome even underwent a total of 20 plasma

exchanges over the course of 11 weeks.82  After the ninth

exchange, the antibody was no longer detectable, althoughit reappeared 1 week later. He was nally transfused after 16

exchanges, despite the continued presence of the antibody

 with a resultant rise in his hematocrit from 14% to 30%.

Clinical Significance

Is It Worth Phenotype or Genotype Matching?

Of the 23 cases of SC alloantibodies catalogued here

(Table 3), one case of HDFN83 and one delayed hemolytic

transfusion reaction (case 2)18 relate sufcient information

in their reports to convincingly attribute clinical relevance

to SC antibodies. The HDFN cases described in the initial

report of the Radin antigen also speak to the potentia

importance of antibodies to Sc4 (especially the case that

required exchange transfusion); however, these cases are

not reported in sufcient detail to denitively implicate

anti-Sc4 to the exclusion of all other causes.20  Even the

 very dramatic presentation of Ms. Scianna’s antibody does

not incriminate anti-Sc1 as the cause of her poor obstetric

outcomes, as an anti-D of a higher titer was also found.

The fundamental question of when and whether antibodies

Page 16: m4440090_27_2_11 Immunohematology

7/23/2019 m4440090_27_2_11 Immunohematology

http://slidepdf.com/reader/full/m444009027211-immunohematology 16/4852  IMMU NOHEM ATOLOGY, Volum e 27, Numb er 2, 2011

P.A.R. Brunker and W.A. Flegel

Table 3. Published reports of antibodies to antigens in the Scianna blood group system

AntibodyAllo orauto? Authors Year

Caseidentifier

Sciannaphenotype

Age(years) Sex Ethnicity

Importantreactions?

Otherantibodies

Transfusion/pregnancyhistory

anti-Sc1 Allo Schmidt et al.2 1962 Mrs. N.S. Sc:–1,2 25 F Caucasian HDFN anti-D Multiparous, after 1st baby withHDFN.

Kaye et al.77 1990 Sc:–1,2 28 F Indian No HDFN Excluded all exceptE and Lu(a)

G2, 1st was uncomplicated withnegative Ab screen.

Au to Tregellas et al.78 1979 Sc:1, –2 49

McDowell et al.79 1986 Case 1 Sc:1,–2 Hemolytic anemia

McDowell et al.79 1986 Case 2 Sc:1,–2(1+, but 3+ 2years later)

Hemolytic anemia 4 units RBC transfused earlier.

Owen et al.80 1992 Sc:1,–2 10 months F West Indies Hemolytic anemia

Ramsey andWilliams81

2010 Sc:1 20 F Caucasian Hemolytic anemia,acute hemolysis

15 units RBC 3 years earlierduring chemotherapy, previously

negative Ab screen.

Auto vs. Allonot resolved

Steane et al.82 1982 Case 2: ptC.D.

22 M Caucasian Not reported.

anti-Sc2 Allo Anderson et al.3 1963 Mr. Char. Sc:1,–2 50 M Caucasian DSTR vs. DHTR 3 units RBC transfused 2 weeksearlier.

Seyfried et al.6 1966 Four donors

DeMarco et al.83 1995 Sc:1,–2 29 F Caucasian HDFN Negative maternalAb screen

G0, no transfusion history.

anti-Sc3 Allo McCreary et al.16 1973 Sc:–1,–2 F Likiep Atoll, MarshallIslands

Transfused 7 months earlierwithout difficulty.

Nason et al.17 1980 Sc:–1,–2 67 M Caucasian DSTR Transfused 4 years earlierwithout incident.

Woodfield et al.19 1986 Sc:–1,–2 4 F Papua New Guinea Many transfusions.

Auto Peloquin et al.84 1989 Case 1 weak Sc1 andSc3

64 Hemolytic anemia

Peloquin et al.84

1989 Case 2 weak Sc1 andSc3 54 Hemolytic anemia 4 units RBCs transfused earlier.

anti-Sc4(anti-Rd)

Allo Rausen et al.20 1967 Family 1 –Rd Sc:–4 (Rd+) F Russian Jewish HDFN “Newborn with hemolyticdisease” in 1st and 3rd children.

Rausen et al.20 1967 Family 2 –Fl Sc:–4 (Rd+) F African American HDFN severe “Newborn with hemolyticdisease” in 7th and 9th children

Rausen et al.20 1967 Family 3 –Ha Sc:–4 (Rd+) F Northern European HDFN “Newborn with hemolyticdisease” in 4th child.

Rausen et al.20 1967 Family 4 –We Sc:–4 (Rd+) F Native American HDFN

Rausen et al.20 1967 Family –5 Gr Sc:–4 (Rd+) F German Jewish orScotch-Irish

HDFN

Lundsgaard andJensen21

1968 Mrs. J.P. Sc:–4 (Rd+) 47 F DSTR: No clinicalevidence ofhemolysis

Had a 9-year-old child.2 units blood transfused during

gynecologic operation.

Lundsgaard and

Jensen21

1968 Mr. L.C. Sc:–4 (Rd+) 55 M Never transfused or hospitalized

Winn et al.85 1976 19 M Caucasian DSTR: No clinicalevidence ofhemolysis

ant i-Vw Negat ive DAT, negat ive Ab screebefore transfusion.

anti-Sc5(anti-STAR)

Allo Devine et al.18 and Skradski

et al.35

1988 Case 3 Sc:1,–2 65 M Irish and English DSTR anti-Canti-e

anti-Jk b

3 units RBC 3 years earlier.

anti-Sc6(anti-SCER)

Allo Devine et al.18 1988 Case 1 Sc:1,–2 76 M German DSTR 3 units RBC 12 years earlier; 3units RBC 6 years earlier.

anti-Sc7(anti-SCAN)

Allo Devine et al.18 1988 Case 2 Sc:1,–2 50 M German, English, andNative American

DHTR anti-D 3 units RBC 8 years earlier;2 units RBC 3 years earlier.

Ab = antibody; ADCC = antibody-dependent cell-mediated cytotoxicity; AHG = antihuman globulin; AIHA = autoimmune hemolytic anemia; CHF = congestiveheart failure; DAT = direct antiglobulin test; DHTR = delayed hemolytic transfusion reaction; DOL = day of life; DSTR = delayed serologic transfusion reaction;

Page 17: m4440090_27_2_11 Immunohematology

7/23/2019 m4440090_27_2_11 Immunohematology

http://slidepdf.com/reader/full/m444009027211-immunohematology 17/48IMMUNOHEMATOLOGY, Volume 27, Number 2, 2011 53

Scianna blood group system: a Review

Clinical synopsis Other laboratory data

Impossible to distinguish role of anti-Sc1 in presence of anti-D.

Neonate with 3+ DAT, eluted anti-Sc1, but Hgb = 13.5 g/dL and no HDFN. Mother B, R1r;infant B, rr; husband Sc:1,–2.

IgG3 subclass; steady rise in ADCC despite const ant titer throughout pregnancy; 9 weeks postpar tum ADCCincreased ×4 and titer doubled (16 to 32).

Healthy blood donor. Antibody demonstrated in serum, not in plasma, suggesting autoantibody is only seen when it reacts with acoagulation factor; IgG3; very weak DAT (both IgG and C3b), weak anti-Sc1 eluted.

3+ DAT and anti-Sc1 in IAT. Eluates by heat, ether, and glycine acid were nonreactive, but xylene eluate showedanti-Sc1.

Transient Sc antigen weakening: serum from initial presentation reacted 3+ with patient’s RBCs 2 years later.

Sudden-onset jaundice, pallor, fever with hepatomegaly, Hgb = 4.1 g/dL. AIHA diagnosed,treated with steroids and IVIG , transfusion, urgent splenectomy. Transfusion-dependent for4 weeks, then steroids stopped 7 months after splenectomy.

DAT+ (IgG and C3d), elevated unconjugated bilirubin, reduced haptoglobin, hemoglobinuria. DAT negative withfollow-up. Serum and eluate demonstrated anti-Sc1.

Hodgkin disease in remission, presented with 3 weeks of fatigue, 2-day Hgb decrea se (9.1to 5.7 g/dL). Gross hematuria and T bili = 5.5 mg/dL during 3 units RBC transfusion.

Ab screen 1–2+ in all cells (anti-Sc1 identified). DAT– (hospital), w+ ( ref. lab; negative eluate). Posttran sfusionHgb = 9.2 g/dL. A nti-Sc1 reacted w+ to patient’s cells. Genotyped SC1/SC1. Patient group O–.

Evans-like syndrome with anemia during a lung infection, 2 years of hospitalizations,steroids, splenectomy, immune suppressants. Transfused safely af ter 16 plasma exchanges.

Reported DAT+ , but “presence of an antibody in his plasma which reacted with all ery throcytes tested except hisown.” Antibody undetectable after the 9th exchange, but it r eappeared, then disappeared after 4 more exchanges.

Carcinoma of the stomach, pretransfusion antibody screen positive. Hemolysis could be thereason for the 2nd blood request, but this is not stated.

Whether the patient was hemolyzing and thus required the pretransfu sion testing is not stated; therefore, we cannodistinguish a DSTR from a DHTR as described in this paper.

Healthy donors iatrogenically immunized for anti-D production.

Jaundice in infant (Tbili = 14.3 mg/dL) on DOL2, postphototherapy discharged Hct = 45%,DOL20 Tbili = 6.5 mg/dL , Hct 17.3% with pallor, tachypnea, tachycardia; transfused 45 mL;Hct 4 months later = 36.3% Mother B +; infant B-.

Maternal serum 3+ against paternal RBCs ( titer of 64) and Sc: –1,2 RBCs; Paternal RBCs Sc:1,2. Neonate cord bloodDAT macroscopically + IgG; Neonate peripheral venous blood 2+; Readmission on DOL20 DAT 2+ (polyspec, IgG, andC3); Eluate + on paternal and S c:1,–2 RBCs.

Preoperative testing. Antibody dropped to below detectable levels shortly after discovery.

Preoperative positive antibody screen for metastatic carcinoma surgery. Not transfusedowing to deteriorating clinical condition and lack of compatible blood.

Negative DAT, did not adsorb anti-Sc1 or anti-Sc2.

Thalassemic with new IgG Ab, mother used as only available donor (also S c:–1,–2). Patientunderwent splenectomy.

Antibody was undetectable after splenectomy and not stimulated by use of XM-compatible blood.

Lymphoma patient with severe anemia; 5 units incompatible RBCs transfused withoutdifficulty.

DAT weak with IgG and C3d, eluates negative. Antibody disappeared within 70 days.

Hodgkin disease, CHF, moderate anemia; 6 units incompatible blood tr ansfused withoutdifficulty. DAT 3+ with C3d, negative with IgG , eluate negative. Serum antibody weaker after transfusion ; additional follow-upnot possible.

2 chi ldren Rd+ , both had HDFN; 1 chi ld Rd– , d id not have HDFN. Presumably very m ild HDFN, as case detected by “ rout ine ant i-globul in t es ting o f cord b lood .”

10 children in pedigree: 5 Rd– with no HDFN; 1st 3 Rd+ without HDFN, last 2 Rd+ withHDFN (1st of these requiring exchange transfusion).

4 children in family: 1 Rd– with no HDFN; 1st 2 Rd+ without HDFN, last 1 Rd+ with HDFN. In the same pedigree, Rd– grandmother of HDN proband had 2 children, 1st Rd+ and 2nd Rd–, neither had HDFN.

4 children in family: 1 Rd– with no HDFN; 1st 2 Rd+ without HDFN, last 1 Rd+ with HDFN.

5 children in family: 3 Rd– with no HDFN; 1 stillborn (3rd in birth order) followed by 1 Rd+with HDFN.

No description of pathology of the stillborn fetus.

Moderately strong DAT (polyspecific and IgG-eluate showed an ti-Rd), weakened with time.Negative Ab screen preoperatively.

Patient is O+; 2 weeks after transfusion, strongly r eactive antibody screen at new hospital. Her child’s RBCs do notreact with patient’s posttransfusion plasma.

Preoperative positive antibody screen for atoxic goiter surgery. Considered as a “naturally

occurring antibody.”11 units RBC on 3 dates for gunshot wound surgery (7/5 , 7/17, and 8/5). 1 unit foundincompatible on crossmatch (RT, 37°C, and AHG) but screening RBCs s till negative.

anti-Vw accounts for incompatible RT crossmatch.

1 week after transfusion of 3 units RBC, A b screen showed new anti-Jk b and a high-frequency antigen. DAT–, patient eventually diagnosed with lymphoma and transfused withautologous units.

Serum reacted with all cells except Sc: –1,–2, autologous, and the patient’s sibling (phenotype Sc:1,–2). Identit y ofantigen discovered with molecular testing years afterward.

Preoperative positive Ab screen for revision of right to tal hip arthroplasty, proceduredelayed until autologous units could be collected.

Identity of antigen discovered with molecular testing years after ward.

12 days after transfusion of 2 units RBC after or thopedic surgery, patient had decrease inhematocrit, pretransfusion Ab screen positive, weakly DAT+ (IgG and C3).

Eluate reacted with all cells except Sc: –1,–2. Antibody no longer detected within 9 months. Identity of antigendiscovered with molecular testing years afterward.

Hct = hematocrit; HDFN = hemolytic disease of the fetus and newborn; Hgb = hemoglobin; IAT = indirect antiglobulin test; IgG = immunoglobulin G; IVIG =intravenous immunoglobulin; RBCs = red blood cells; RT = room temperature; Tbili = total bilirubin; XM = crossmatch.

Page 18: m4440090_27_2_11 Immunohematology

7/23/2019 m4440090_27_2_11 Immunohematology

http://slidepdf.com/reader/full/m444009027211-immunohematology 18/4854  IMMU NOHEM ATOLOGY, Volum e 27, Numb er 2, 2011

P.A.R. Brunker and W.A. Flegel

to SC system antigens are of clinical importance remains

unresolved because these antibodies are not routinely

characterized, especially if they are detected along with

other, better-understood alloantibodies that can account

for a clinical presentation.

On the Detection of SC System Antibodies in Routine

Pretransfusion Testing

The most important obstacle to understanding the

clinical consequences of transfusing across SC antigens is

that serologic reagents to simply dene these antigens in

 both patients and test RBCs have not been widely available.

 As they do for the Dombrock blood group system,88 

molecular approaches help resolve these situations. It

has been estimated that approximately 13 percent of the

transfused population are immunologic responders capable

of creating alloantibodies,89 but unless RBC reagents with

SC antigens are included in the routine laboratory workup,these antibodies may go unnoticed. It is also problematic

if an antibody to one of the high-prevalence SC antigens is

in fact detected because the mere presence of the antibody

does not necessarily correspond to clinical importance.

Such a qualitative assay is insufcient. The quantitative

characterization of the antibody, such as by titer, has

only been performed in a few SC antibody cases, and we

recommend that this parameter be more routinely evaluated

in cases of possible hemolysis caused by antibodies in the SC

system and in most other blood group systems. Especially

 because HDFN has been reported, establishing a better

understanding of antibody potency as detected by the titer would help inform practice guidelines.

 An economic and rational approach would be

the routine use of recombinant SC protein during

pretransfusion testing before titration to effectively screen

only for antibodies present above a threshold titer.87  In

this way, nuisance antibodies (akin to the low-titer cold

autoantibodies detected before routine testing at higher

temperatures) would fall under the radar as desired and

precious laboratory resources would not be spent working

up these most likely incidental ndings. Only higher titer

antibodies would be detected, focusing the laboratory

investigation on cases more likely to be of consequence to

the patient. If these laboratory techniques do lead to the

increased identication of SC system alloantibodies, the

medical importance of respecting them in a particular

patient’s transfusion recommendation remains debatable.

Should We Genotype for SC Alleles?

The integration of molecular diagnostics with trans-

fusion medicine has been a slow, but steady, process, with

applications ranging from individual patient prenatal

diagnosis to routine high-throughput donor testing.90,9

 Although the rate of implementation of these technologies

 varies among nations92 and local transfusion services,93  it is

expanding.94 The transfusion community should move these

procedures from potentially inaccessible research laboratories

to standard clinical practice.95  For optimal performance of

such an approach toward personalized medicine, ideally al

donors and all recipients should be evaluated at a molecular

level. Economic barriers to this strategy are falling as high-

throughput and multiplexed assays are achieved, because

the incremental cost of adding a handful of additional SNPs

 when designing a DNA microarray is negligible. Consequently

molecular testing strategies are shifting from decisions abou

 which variants to include in a genotyping system that force the

prioritization of well-studied variants already known to have

medical importance, to nding effective platforms to analyze

data derived from genotyping as many known variants as

possible, regardless of their allele frequencies or uncertainclinical effects.

The most compelling reason to genotype SC variants in

greater depth, both by increasing the number of donors and

patients and also by including more variants regardless of

their prevalence, is that such data are necessary to clarify

the presently ambiguous role of SC in clinical transfusion

practice. At the same time, valuable data for research into

ERMAP biology are accrued without added costs. Inclusion

of the  SC*01  to  SC*07   alleles in high-throughput assays

 would be a move in the right direction, and in cases o

unresolved serology, a referral to the molecular laboratory

for an in-depth investigation may be desirable. By usinga high-throughput genetic test to determine the potential

for alloimmunization by a variant homozygote, we can

collect such genetic data almost for free. The pursuit of

alloantibodies in such variant homozygous patients is

a feasible strategy to achieve complete resolution of al

alloantibodies in a posttransfusion sample submitted as a

transfusion reaction investigation. Consequently, knowing

these genetic data gives us the opportunity to better dene

 which antibodies are of clinical importance.

Conclusion

The SC story is an excellent example of the essential

roles that classical genetics played in the original physical

mapping of a blood group system and is an exceptional

illustration of transfusion laboratories that were in the right

place at the right time to nally identify the responsible

locus in the era of whole genome analysis. Parsing out the

meaning of global polymorphism frequency distributions

in light of founder effects or selection pressures would be

a useful adjunct to in vitro studies of transcript utilization

Page 19: m4440090_27_2_11 Immunohematology

7/23/2019 m4440090_27_2_11 Immunohematology

http://slidepdf.com/reader/full/m444009027211-immunohematology 19/48IMMUNOHEMATOLOGY, Volume 27, Number 2, 2011 55

Scianna blood group system: a review

and protein expression and function. The continued

study of ERMAP homologues, such as the BTNL family of

proteins in general, and their physiologic interactions in

other species, such as the B30.2-containing bloodthirsty 

gene in Antarctic iceshes, is a promising avenue to explore

fundamental insights into erythropoiesis. Investigatorsin transfusion medicine are uniquely poised not only to

carry out this work but also to lead the way toward a more

comprehensive understanding of the “lucky 13th” blood

group, SC, making us the lucky ones indeed.

 Acknowledgment

 We gratefully acknowledge Professor Guillaume

Lecointre for kindly providing the photograph of blood

from the Antarctic icesh (Fig. 5).

References

  1. Wagner FF, Poole J, Flegel WA. Scianna antigens includingRd are expressed by ERMAP. Blood 2003;101:752–7.

  2. Schmidt RP, Griftts JJ, Northman FF. A new antibody,anti-Sm, reacting with a high incidence antigen. Transfusion1962;2:338–40.

  3. Anderson C, Hunter J, Zipursky A, Lewis M, Chown B. An antibody dening a new blood group antigen, Bu-a.Transfusion 1963;3:30–3.

  4. Lewis M, Chown B, Schmidt RP, Griftts JJ. A possiblerelationship between the blood group antigens Sm andBu-a. Am J Hum Genet 1964;16:254–5.

  5. Lewis M, Chown B, Kaita H, Philipps S. Furtherobservations on the blood group antigen Bu-a. Am J HumGenet 1964;16:256–60.

  6. Seyfried H, Frankowska K, Giles CM. Further examplesof anti-bu-a found in immunized donors. Vox Sang 1966;11:512–16.

  7. Lewis M, Chown B, Kaita H. On the blood group antigensBua and Sm. Transfusion 1967;7:92–4.

  8. Giles CM, Bevan B, Hughes RM. A family showingindependent segregation of Bua and Ytb. Vox Sang 1970;18:265–6.

  9. Rowe GP. A second family showing independent segregationof Sc 2(Bua) and Ytb. Vox Sang 1986;50:191.

  10. Lewis M, Kaita H, Chown B. Scianna blood group system. Vox Sang 1974;27:261–4.

  11. Lewis M, Kaita H, Chown B. Genetic linkage between thehuman blood group loci Rh and Sc (Scianna). Am J HumGenet 1976;28:619–20.

  12. Lewis M, Kaita H, Côté GB, Chown B, Giblett ER, AndersonJA. Chromosome 1: lods on linkage among eight loci: Do,ENO1, Fy, PGM1, Rh, UMPK, Sc, and PGD. Birth DefectsOrig Artic Ser 1976;12:322–5.

  13. Lewis M, Kaita H, Chown B. Relative positions ofchromosome 1 loci Fy, PGM1, Sc, UMPK, Rh, PGD andENO1 in man. Can J Genet Cytol 1977;19:695–709.

  14. Lewis M, Kaita H, Giblett ER, Anderson JE. Data on

chromosome 1 loci Fy, PGM1, Sc, UMPK, Rh, PGD, and

ENO1: two-point lods, R:NR counts, multipoint informationand map. Cytogenet Cell Genet 1978;22:392–5.

  15. Noades JE, Corney G, Cook PJ, et al. The Scianna bloodgroup lies distal to uridine monophosphate kinase on

chromosome 1p. Ann Hum Genet 1979;43:121–32.

  16. McCreary J, Vogler AL, Sabo B, Eckstein EG, Smith TR

 Another minus-minus phenotype: Bu(a-)Sm-, two examples

in one family [abstract]. Transfusion 1973;13:350.

  17. Nason SG, Vengelen-Tyler V, Cohen N, Best M, Quirk J. A

high incidence antibody (anti-Sc3) in the serum of a Sc:-1,-2patient. Transfusion 1980;20:531–5.

  18. Devine P, Dawson FE, Motschman TL, et al. Serologicevidence that Scianna null (Sc:-1,-2) red cells lack multiple

high-frequency antigens. Transfusion 1988;28:346–9.

  19. Woodeld DG, Giles C, Poole J, Oraka R, Tolanu T. A further

null phenotype (Sc-1-2) in Papua New Guinea [Abstract]

21st Congress of the Int Soc Haem and 19th Congress of theISBT, Sydney, Australia, May 1986; 651.

  20. Rausen AR, Roseneld RE, Alter AA, et al. A “new” infrequen

red cell antigen, Rd (radin). Transfusion 1967;7:336–42.

  21. Lundsgaard A, Jensen KG. Two new examples of anti-Rd

 A preliminary report on the frequency of the Rd (Radinantigen in the Danish population. Vox Sang 1968;14:452–7.

  22. Lewis M, Kaita H. Genetic linkage between the Radin andRh blood group loci. Vox Sang 1979;37:286–9.

  23. Lewis M, Kaita H, Philipps S, et al. The position of the Radin blood group locus in relation to other chromosome l loci

 Ann Hum Genet 1980;44(Pt 2):179–84.

  24. Spring FA, Herron R, Rowe G. An erythrocyte glycoprotein

of apparent Mr 60,000 expresses the Sc1 and Sc2 antigens

 Vox Sang 1990;58:122–5.

  25. Spring FA. Characterization of blood-group-active

erythrocyte membrane glycoproteins with human antiserasTransfus Med 1993;3:167–78.

  26. Su YY, Gordon CT, Ye TZ, Perkins AC, Chui DH. HumanERMAP: an erythroid adhesion/receptor transmembrane

protein. Blood Cells Mol Dis 2001;27:938–49.

  27. Xu H, Foltz L, Sha Y, et al. Cloning and characterization o

human erythroid membrane-associated protein, human

ERMAP. Genomics 2001;76:2–4.

  28. Ensembl entry for ERMAP, number ENSG00000164010

2010. http://www.ensembl.org/Homo_sapiens/Location/

 View?db=core;r=1:43276635-43326634. Accessed July 142010.

  29. Entrez Gene entry for ERMAP. http://www.ncbi.nlm.nihgov/gene/114625. Accessed July 14, 2010.

  30. Havana entry for ERMAP, transcr ipt numbersOTTHUMT00000020180 through OTTHUMT

00000020184 2010. http://vega.sanger.ac.uk/Homo_

sapiens/Gene/Summary?g=OTTHUMG00000007619. Accessed July 14, 2010.

  31. Ensembl Transcript View, ERMAP’s 5 listed transcr ipts2010. http://www.ensembl.org/Homo_sapiens/Gene/Su

mmary?db=core;g=ENSG00000164010;r=1:43276635

43326634;t=ENST00000470938. Accessed July 14, 2010.

Page 20: m4440090_27_2_11 Immunohematology

7/23/2019 m4440090_27_2_11 Immunohematology

http://slidepdf.com/reader/full/m444009027211-immunohematology 20/4856  IMMU NOHEM ATOLOGY, Volum e 27, Numb er 2, 2011

  32. Fuchisawa A, Lomas-Francis C, Hue-Roye K, Reid ME. The

polymorphism nt 76 in exon 2 of SC is more frequent in whites than in blacks. Immunohematology 2009;25:18–19.

  33. Hashmi G, Sharif f T, Zhang Y, et al. Determination of 24

minor red blood cell antigens for more than 2000 blooddonors by high-throughput DNA analysis [published

correction appears in Transfusion 2007;47:952].

Transfusion 2007;47:736–47.

  34. Ribeiro KR, Guarnieri MH, da Costa DC, Costa FF,

Pellegrino J Jr, Castilho L. DNA array analysis for red blood cell antigens facilitates the transfusion support with

antigen-matched blood in patients with sickle cell disease.

 Vox Sang 2009;97:147–52.

  35. Skradski KJ, McCreary J, Sabo B, Polesky HF. An antibody

against a high frequency antigen absent on red cells of

the Scianna:-2,-2 phenotype [abstract]. Transfusion 1982(22):406.

  36. Hue-Roye K, Chaudhuri A, Velliquette RW, et al. STAR: anovel high-prevalence antigen in the Scianna blood group

system. Transfusion 2005;45:245–7.

  37. Flegel WA, Chen Q, Reid ME, et al. SCER and SCAN: twonovel high-prevalence antigens in the Scianna blood group

system. Transfusion 2005;45:1940–4.

  38. Smigielski EM, Sirotkin K, Ward M, Sherry ST. dbSNP: a

database of single nucleotide polymorphisms. Nucleic AcidsRes 2000;28:352–5.

  39. The International HapMap Project Homepage 2010. http://hapmap.ncbi.nlm.nih.gov/. Accessed July 14, 2010.

  40. Stephan W. Genetic hitchhiking versus backgroundselection: the controversy and its implications. Philos Trans

R Soc Lond B Biol Sci 2010;365:1245–53.

  41. Williams AF, Barclay AN. The immunoglobulin super-

family—domains for cell surface recognition. Annu Rev

Immunol 1988;6:381–405.

  42. Cannon JP. Plasticity of the immunoglobulin domain in the

evolution of immunity. Integr Comp Biol 2009;49:187–96.

  43. Harpaz Y, Chothia C. Many of the immunoglobulin

superfamily domains in cell adhesion molecules and surface

receptors belong to a new structural set which is close tothat containing variable domains. J Mol Biol 1994;238: 

528–39.

  44. Aricescu AR, Jones EY. Immunoglobulin superfamily cell

adhesion molecules: zippers and signals. Curr Opin Cell Biol

2007;19:543–50.

  45. Peggs KS, Allison JP. Co-stimulatory pathways in

lymphocyte regulation: the immunoglobulin superfamily.Br J Haematol 2005;130:809–24.

  46. Vogel C, Chothia C. Protein family expansions and biologicalcomplexity. PLoS Comput Biol 2006;2:e48.

  47. Simon TL, Dzik WH, Snyder EL, Stowell CP, Strauss RG,eds. Rossi’s Principles of Transfusion Medicine. 3rd ed.

Philadelphia, PA: Lippincott Williams & Wilkins, 2002.

  48. Sharpe AH, Freeman GJ. The B7-CD28 superfamily. Nat

Rev Immunol 2002;2:116–26.

  49. Mather IH, Jack LJ. A review of the molecular and cellular

 biology of butyrophilin, the major protein of bovine milk fat

globule membrane. J Dairy Sci 1993;76:3832–50.

  50. Rhodes DA, Stammers M, Malcherek G, Beck S, TrowsdaleJ. The cluster of  BTN   genes in the extended majorhistocompatibility complex. Genomics 2001;71:351–62.

  51. Robenek H, Hofnagel O, Buers I, et al. Butyrophilincontrols milk fat globule secretion. Proc Natl Acad Sci U S A2006;103:10385–90.

  52. Smith IA, Knezevic BR, Ammann JU, et al. BTN1A1, the

mammary gland butyrophilin, and BTN2A2 are bothinhibitors of T cell activation. J Immunol 2010;1843514–25.

  53. Valentonyte R, Hampe J, Huse K, et al. Sarcoidosis isassociated with a truncating splice site mutation in BTNL2[published correction appears in Nat Genet 2005;37:652]Nat Genet 2005;37:357–64.

  54. Arnett HA, Escobar SS, Viney JL. Regulation ocostimulation in the era of butyrophilins. Cytokine 200946:370–5.

  55. Nguyen T, Liu XK, Zhang Y, Dong C. BTNL2, a butyrophilin-like molecule that functions to inhibit T cell activation. JImmunol 2006;176:7354–60.

  56. Meyer M, Gaudieri S, Rhodes DA, Trowsdale J. Cluster oTRIM genes in the human MHC class I region sharing theB30.2 domain. Tissue Antigens 2003;61:63–71.

  57. Vernet C, Boretto J, Mattei MG, et al. Evolutionary study omultigenic families mapping close to the human MHC classI region. J Mol Evol 1993;37:600–12.

  58. Jack LJ, Mather IH. Cloning and analysis of cDNA encoding bovine butyrophilin, an apical glycoprotein expressed inmammary tissue and secreted in association with the milk-fat globule membrane during lactation. J Biol Chem 1990265:14481–6.

  59. Tazi-Ahnini R, Henry J, Offer C, Bouissou-BouchouataC, Mather IH, Pontarotti P. Cloning, localization, andstructure of new members of the butyrophilin gene family inthe juxta-telomeric region of the major histocompatibilitycomplex. Immunogenetics 1997;47:55–63.

  60. Weinert C, Grütter C, Roschitzki-Voser H, Mittl PRGrütter MG. The crystal structure of human pyrin b30.2domain: implications for mutations associated with familiaMediterranean fever. J Mol Biol 2009;394:226–36.

  61. Woo JS, Imm JH, Min CK, Kim KJ, Cha SS, Oh BHStructural and functional insights into the B30.2/SPRYdomain. EMBO J 2006;25:1353–63.

  62. Woo JS, Suh HY, Park SY, Oh BH. Structural basis forprotein recognition by B30.2/SPRY domains. Mol Cel2006;24:967–76.

  63. Jeong J, Rao AU, Xu J, et al. The PRY/SPRY/B30.2

domain of butyrophilin 1A1 (BTN1A1) binds to xanthineoxidoreductase: implications for the function of BTN1A1 inthe mammary gland and other tissues. J Biol Chem 2009284:22444–56.

  64. Garofalo F, Pellegrino D, Amelio D, Tota B. The Antarctichemoglobinless icesh, fty ve years later: a uniquecardiocirculatory interplay of disaptation and phenotypicplasticity. Comp Biochem Physiol A Mol Integr Physiol2009;154:10–28.

  65. Yergeau DA, Cornell CN, Parker SK, Zhou Y, Detrich HW3rd. bloodthirsty,  an RBCC/TRIM gene required forerythropoiesis in zebrash. Dev Biol 2005;283:97–112.

P.A.R. Brunker and W.A. Flegel

Page 21: m4440090_27_2_11 Immunohematology

7/23/2019 m4440090_27_2_11 Immunohematology

http://slidepdf.com/reader/full/m444009027211-immunohematology 21/48IMMUNOHEMATOLOGY, Volume 27, Number 2, 2011 57

  66. Ye TZ, Gordon CT, Lai YH, et al. Ermap, a gene coding fora novel erythroid specic adhesion/receptor membraneprotein. Gene 2000;242:337–45.

  67. Kuriyan MA, Oyen RE, Marsh WL. Demonstration of Diego(Dib) and Scianna (Scl) antigens on phagocytic leukocytesof the blood. Transfusion 1978;18:361–4.

  68. Rojewski MT, Schrezenmeier H, Flegel WA. Tissue

distribution of blood group membrane proteins beyond redcells: evidence from cDNA libraries. Transfus Apher Sci2006;35:71–82.

  69. He XR, He YY, Chen Y, Ye TZ. Expression of human ERMAP  gene in fetal tissues [in Chinese]. Zhongguo Shi Yan Xue YeXue Za Zhi 2006;14:972–5.

  70. Zhang XH, Ye TZ, Hu B, Si WZ. Quantication of human ERMAP  by using real-time FQ-PCR [in Chinese]. ZhongguoShi Yan Xue Ye Xue Za Zhi 2005;13:154–7.

  71. He YY, Zhang XH, Ye TZ, Wu ZL. Study on the expressionof human  ERMAP  gene in erythropoietic and macrophagedifferentiation of K562 cells [in Chinese]. Zhongguo Shi YanXue Ye Xue Za Zhi 2005;13:553–6.

  72. He YY, Zhang XH, Ye TZ, Wu ZL. Expression of human ERMAP  gene in different cell lines [in Chinese]. ZhongguoShi Yan Xue Ye Xue Za Zhi 2005;13:819–22.

  73. Liang JF, Chen Y, Ye TZ, et al. Effects of human ERMAP-siRNA on erythroid differentiation of K562 cells induced by Ara-C [in Chinese]. Zhongguo Shi Yan Xue Ye Xue Za Zhi2009;17:49–53.

  74. Lin LD, He XR, Ye TZ, et al. Expression of human ERMAP  gene in umbilical cord blood mononuclear cells duringdifferentiation and development towards erythroidlineage [in Chinese]. Zhongguo Shi Yan Xue Ye Xue Za Zhi2008;16:328–32.

  75. Reid ME, Lomas-Francis C. The blood group antigenfactsbook. 2nd ed. San Diego, CA: Academic Press; 2004.

  76. Velliquette RW. Review: the Scianna blood group system.Immunohematology 2005;21:70–6.

  77. Kaye T, Williams EM, Garner SF, Leak MR, Lumley H. Anti-Sc1 in pregnancy. Transfusion 1990;30:439–40.

  78. Tregellas WM, Holub MP, Moulds JJ, Lacey PA. An exampleof autoanti-Sc1 demonstrable in serum but not in plasma[abstract]. Transfusion 1979;19:650.

  79. McDowell MA, Stocker I, Nance S, Garratty G. Auto anti-Sc1associated with autoimmune hemolytic anemia [abstract].Transfusion 1986;26:578.

  80. Owen I, Chowdhury V, Reid ME, Poole J, Marsh JC, HowsJM. Autoimmune hemolytic anemia associated with anti-Sc1. Transfusion 1992;32:173–6.

  81. Ramsey G, Williams LM. Autoimmune hemolytic

anemia with auto-anti-Sc1, weakened Sc:1 antigen, andsuperimposed transfusion-associated acute hemolysis[abstract]. Transfusion 2010;50(2S):156A.

  82. Steane EA, Sheehan RG, Brooks BD, Frenkel EPTherapeutic plasmapheresis in patients with antibodies tohigh-frequency red cell antigens. Prog Clin Biol Res 1982106:347–53.

  83. DeMarco M, Uhl L, Fields L, Pacini D, Gorlin JB, KruskalMS. Hemolytic disease of the newborn due to the Sciannaantibody, anti-Sc2. Transfusion 1995;35:58–60.

  84. Peloquin P, Moulds M, Keenan J, Kennedy M. Anti-Sc3as an apparent autoantibody in two patients [abstract]Transfusion 1989;29(Suppl):49S.

  85. Winn LC, Eska PL, Grindon AJ. Anti-Rd (Radin) followingthe transfusion of a Radin positive unit of blood. Transfusion1976;16:351–2.

  86. Wagner FF, Frohmajer A, Ladewig B, et al. Weak D allelesexpress distinct phenotypes. Blood 2000;95:2699–708.

  87. Seltsam A, Grueger D, Blasczyk R, Flegel WA. Easyidentication of antibodies to high-prevalence Sciannaantigens and detection of admixed alloantibodies usingsoluble recombinant Scianna protein. Transfusion 200949:2090–6.

  88. Reid ME. Complexities of the Dombrock blood group systemrevealed. Transfusion 2005;45(2 Suppl):92S–9S.

  89. Higgins JM, Sloan SR. Stochastic modeling of human RBCalloimmunization: evidence for a distinct population oimmunologic responders. Blood 2008;112:2546–53.

  90. Reid ME. Transfusion in the age of molecular diagnosticsHematology Am Soc Hematol Educ Program 2009:171–7.

  91. Veldhuisen B, van der Schoot CE, de Haas M. Bloodgroup genotyping: from patient to high-throughput donorscreening. Vox Sang 2009;97:198–206.

  92. Flegel WA. Blood group genotyping in Germany. Transfusion2007;47(1 Suppl):47S–53S.

  93. Hillyer CD, Shaz BH, Winkler AM, Reid M. Integratingmolecular technologies for red blood cell typing and

compatibility testing into blood centers and transfusionservices. Transfus Med Rev 2008;22:117–32.

  94. Garraty G, Reid M, Westhoff C, eds. A Workshop onMethods in Molecular Immunohematology. Transfusion2010;47:1S–100S.

  95. Denomme GA, Flegel WA. Applying molecular immuno-hematology discoveries to standards of practice in blood banks: now is the time. Transfusion 2008;48:2461–75.

 Patricia A.R. Brunker, MD, DPhil, and Willy A. Flegel

 MD (corresponding author), Laboratory Services Section

 Department of Transfusion Medicine, Clinical Center, Nationa

 Institutes of Health, Bethesda, MD 20892.

Scianna blood group system: a review

Important Notice About Manuscripts for

Immunohematology 

Please e-mail all manuscripts to [email protected]

Immunohematology  is on the Web!

www.redcross.org/immunohematology

For more information, send an e-mail to

[email protected]

Page 22: m4440090_27_2_11 Immunohematology

7/23/2019 m4440090_27_2_11 Immunohematology

http://slidepdf.com/reader/full/m444009027211-immunohematology 22/4858  IMMU NOHEM ATOLOGY, Volum e 27, Numb er 2, 2011

 Antibodies to antigens in the Kell blood group system are usuallyimmunoglobulin G, and, notoriously, anti-K, anti-k, and anti-Kpa can cause severe hemolytic transfusion reactions, as well as severehemolytic disease of the fetus and newborn (HDFN). It has beenshown that the titer of anti-K does not correlate with the severityof HDFN because, in addition to immune destruction of red

 blood cells (RBCs), anti-K causes suppression of erythropoiesisin the fetus, which can result in severe anemia. We report a caseinvolving anti-Kpa  in which one twin was anemic and the other was not. Standard hemagglutination and polymerase chainreaction (PCR)-based tests were used. At delivery, anti-Kpa was

identied in serum from the mother and twin A, and in the eluateprepared from the baby’s RBCs. PCR-based assays showed twin A (boy) was  KEL*841T/C (KEL*03/KEL*04),  which is predictedto encode Kp(a+b+). Twin B (girl) was  KEL*841C/C (KEL*04/  KEL*04),  which is predicted to encode Kp(a–b+). We describethe rst reported case of probable suppression of erythropoiesisattributable to anti-Kpa. One twin born to a woman whose serumcontained anti-Kpa  experienced HDFN while the other did not.Based on DNA analysis, the predicted blood type of the affectedtwin was Kp(a+b+) and that of the unaffected twin was Kp(a–b+).The laboratory ndings and clinical course of the affected twin were consistent with suppression of erythropoiesis in addition

to immune RBC destruction.  Immunohematology  2011;27:58–60.

Key words:  erythropoiesis—suppression, Kell blood

group system, hemolytic disease of the newborn, Kpa 

The rst antigen in the Kell blood group system, K, was

discovered in 1946 as the result of hemolytic disease of the

newborn. The system now includes 31 antigens. Antibodies

to K, k, Kpa, Kp b, Kpc, Jsa, and Js b  are the most clinically

important.1  Next to anti-A, anti-B, and anti-D, anti-K is

the most common immune RBC antibody.2  Antibodies to

antigens in the Kell blood group system are usually IgG,

and, notoriously, anti-K, anti-k, and anti-Kpa cause severe

hemolytic transfusion reactions, as well as severe hemolytic

disease of the fetus and newborn (HDFN).3,4

It is well known that the titer of anti-K does not correlate

 with the severity of HDFN; indeed, severe HDFN caused

 by anti-K has been associated with lower antibody titers,

 bilirubin levels, and reticulocytosis than has hemolytic

disease of the newborn caused by anti-D.2,3  The anemia

seen in these infants is caused by a combination of immune

destruction and suppression of erythropoiesis in the

fetus.5,6  Vaughan et al. demonstrated experimentally that

proliferation of K+ erythroid progenitors was inhibited by

anti-K, and suggested that the K glycoprotein plays a role in

erythropoiesis.5

Cases of HDFN involving antibodies to other antigens

in the Kell system of antibodies are rare. We retrospectively

report a case of HDFN in twins of a mother whose serum

contained anti-Kpa. The Kp(a+) twin was anemic, and the

Kp(a–) twin was not.

Case History

 A 31-year-old gravida 3, para 2 woman with a history

of a negative antibody screen delivered ABO-identical, Rh-

compatible fraternal twins at full term (37 weeks’ gestation)

The mother had two previous pregnancies delivered by

cesarean section; no problems were noted in the medica

record. At delivery, the cord hemoglobin (Hb) for twin A

(male) was 10.2 g/dL and for twin B (female) was 17.0 g/dL

Unfortunately, there were no cord bilirubin or reticulocyte

counts done for either twin.

Because of anemia, shortly after birth twin A received

a single-aliquot RBC transfusion that was compatible withthe maternal serum. On day 1, he received an additional

single-aliquot transfusion with RBCs and was discharged

on day 5 with a Hb of 15.1 g/dL and an absolute reticulocyte

count of 0.16 × 109/L (normal range of 0.125–0.325 ×

109/L). Transfusion was again required on day 25 when the

Hb reached a nadir of 7.5 g/dL with a decreased absolute

reticulocyte count of 0.007 m/μL. Additional transfusions

 were given on days 30 and 39 to maintain a Hb level above

9.0 g/dL. After transfusion of two aliquots on day 39, the

Hb of twin A was 9.6 g/dL and on day 43 and 52 it was 9.3

g/dL and 9.5 g/dL, respectively. Two weeks later the Hb was

10.3 g/dL, and 6 months later it was 12.6 g/dL. The infant’s

 bilirubin peaked at 5 mg/dL on day 1 and fell steadily during

the monitoring period. White blood cell and platelet counts

remained normal.

Materials and Methods

Hemagglutination

Standard hemagglutination tests were used throughout

The cord blood samples were tested by tube method for

Possible suppression of fetal erythropoiesis bythe Kell blood group antibody anti-Kpa

M. Tuson, K. Hue-Roye, K. Koval, S. Imlay, R. Desai, G. Garg, E. Kazem, D. Stockman, J . Hamilton, and M.E. Reid

CASE REPORT

Page 23: m4440090_27_2_11 Immunohematology

7/23/2019 m4440090_27_2_11 Immunohematology

http://slidepdf.com/reader/full/m444009027211-immunohematology 23/48IMMUNOHEMATOLOGY, Volume 27, Number 2, 2011 59

Suppression of fetal erythropoiesis by anti-Kpa

 ABO, Rh, and direct antiglobulin test (DAT). The maternal

and paternal samples were tested by MTS gel cards (Ortho

Clinical Diagnostics, Raritan, NJ).

Polymerase Chain Reaction–Restriction Fragment

Length Polymorphism Analysis for  KEL*03/KEL*04

Genomic DNA was isolated from epithelial cells

captured by the buccal swabs obtained from the twins

using a DNA kit (QIAamp DNA Blood Mini Kit, QIAGEN,

Inc., Valencia, CA). Exon 8 of  KEL  was amplied using

a forward oligonucleotide primer (Life Technologies,

Inc., Gaithersburg, MD) designed within exon 8

(TACCTGACTTACCTGAATCAGCTGGGAACC) and a

reverse oligonucleotide primer designed at the 3′ end of

exon 8 and into intron 8 (tcttctggcccccagttccaggcacCATGA).

Five microliters of DNA per reaction was amplied by 5

units of Taq DNA polymerase (HotStarTaq, QIAGEN Inc.,

 Valencia, CA) in a 50-μL reaction mixture containing2.5 mM MgCl2, 1 × PCR buffer, 0.2 mM dNTPs, and 100

ng of forward and reverse primer. The PCR amplication

 was achieved in 35 cycles with a nal extension time of 10

minutes, using 62°C as the annealing temperature. The

PCR 202-bp products were digested using the restriction

enzyme NlaIII. A restriction enzyme site for this restriction

enzyme is present in the KEL*03 variant (841T) but not in

the wild-type (841C).

Results

Hemagglutination At the time of delivery, anti-Kpa  was identied in the

maternal serum. No other unexpected antibodies were

present in the maternal serum. Tests using the serum

from twin A and an eluate prepared from his RBCs also

contained anti-Kpa (Table 1). Antibody testing on days 25,

30, and 39 demonstrated the persistence of anti-Kpa in the

 baby’s serum.

Polymerase Chain Reaction–Restriction Fragment

Length Polymorphism Analysis for  KEL*03/KEL*04

Twin A (boy) was  KEL*841T/C (KEL*03/KEL*04), 

 which is predicted to encode Kp(a+b+). Twin B (girl) was

 KEL*841C/C (KEL*04/KEL*04),  which is predicted to

encode Kp(a–b+).

Conclusions

 We describe the rst reported case of possible

suppression of erythropoiesis attributable to anti-Kpa. One

twin born to a woman whose serum contained anti-Kpa 

experienced HDFN and anemia, while the other did not.

Based on DNA analysis, the predicted blood type of the

affected twin was Kp(a+b+) and that of the unaffected twin

 was Kp(a–b+). The laboratory ndings and clinical course

of the affected twin were consistent with suppression of

erythropoiesis in addition to immune RBC destruction. Thesteadily declining bilirubin levels concurrent with a declining

hemoglobin level and decreased absolute reticulocyte

count provide supportive evidence for this interpretation

It is possible that the change in the reticulocyte count was

induced, or exacerbated, by transfusion. Although this was

a retrospective study and not all data were available to us, it

is worth documenting, especially because one twin was an

antigen-negative twin and served as a control.

 Antigens in the Kell blood group system appear on

erythroid progenitor cells early in erythropoiesis.7,8 Because

erythroid progenitor cells do not contain hemoglobin

immune destruction of these cells by anti-Kpa  couldexplain the absence of hyperbilirubinemia in our patient

The restriction of Kell messenger RNA and protein to

erythroid precursors explains why white blood cell and

platelet counts remained normal in the affected twin.9,10

The anemia of the affected twin resolved after 3 months

and his Hb has remained within the normal range. Twin B

had no postdelivery complications and provides a biologic

control for factors other than maternal alloantibody as a

cause of the anemia. Although anti-K is most commonly

associated with erythropoietic suppression, twin A shows

that antibodies to other antigens in the Kell blood group

system can also have this capability.

 Acknowledgments

 We thank Yaddanapudi Ravindranath, MD, for follow

up information after the twins were discharged from

the hospital and Robert Ratner for help in preparing the

manuscript.

Table 1. Results of hemagglutination tests

Mother Twin A Twin B Father

ABO/Rh A + A + A + A +

DAT Not tested 4+ 0 Not tested

IAT withmaternal serum

Not tested Not tes ted Compatible Incompatib le

Other studies Anti-Kpa Anti-Kpa inserum and

eluate

Kp(a+)

DNA frombuccal smear

Not tested   KEL*03/ KEL*04

KEL*04/ KEL*04

Not tested

DAT = direct antiglobulin test; IAT = indirect antiglobulin test.

Page 24: m4440090_27_2_11 Immunohematology

7/23/2019 m4440090_27_2_11 Immunohematology

http://slidepdf.com/reader/full/m444009027211-immunohematology 24/4860  IMMU NOHEM ATOLOGY, Volum e 27, Numb er 2, 2011

M. Tuson et al.

References

  1. Reid ME, Lomas-Francis C. Blood Group Antigens and Antibodies: A guide to clinical relevance and technical tips.New York, NY: Star Bright Books, 2007.

  2. Klein HG, Anstee DJ. Mollison’s blood transfusion in clinicalmedicine. 11th ed. Oxford: Wiley-Blackwell, 2006.

  3. Roback JD, Combs MR, Grossman BJ, et al. Technicalmanual. 16th ed. Bethesda, MD: American Association ofBlood Banks, 2008.

  4. Costamagna L, Barbarini M, Viarengo GL, Pagani A, IserniaD, Salvaneschi L. A case of hemolytic disease of the newborndue to anti-Kpa. Immunohematology 1997;13:61–2.

  5. Vaughan JI, Manning M, Warwick RM, Letsky EA, MurrayNA, Roberts IA. Inhibition of erythroid progenitor cells byanti-Kell antibodies in fetal alloimmune anemia. N Engl JMed 1998;338:798–803.

  6. Daniels G, Hadley A, Green CA. Causes of fetal anemia inhemolytic disease due to anti-K. Transfusion 2003;43:115–16.

  7. Southcott MJG, Tanner MJ, Anstee DJ. The expression ofhuman blood group antigens during erythropoiesis in a cellculture system. Blood 1999;93:4425–35.

  8. Daniels G, Green C. Expression of red cell surface antigens

during erythropoiesis. Vox Sang 2000;78(Suppl 2):149–53.

  9. Jaber A, Loirat MJ, Willem C, Bloy C, Cartron JP, BlanchardD. Characterization of murine monoclonal antibodiesdirected against the Kell blood group glycoprotein. Br JHaematol 1991;79:311–15.

  10. McGinniss MH, Dean A. Expression of red cell antigens byK562 human leukemia cells before and after induction ohemoglobin synthesis by hemin. Transfusion 1985;25:105–9.

 Michelle Tuson, MT(ASCP)SBB, Trinity Health, Farmington Hills

 MI; Kim Hue-Roye, BSc, Laboratory of Immunochemistry, New

York Blood Center, New York, NY; Karen Koval, MT, Sherwin

 Imlay, MD, Rajendra Desai, MD, Gayatri Garg, MD, and Esam

 Kazem, MD, St. Joseph Mercy Hospital—Oakland, Pontiac, MI

 Diane Stockman, MT(ASCP), and Janis S. Hamilton, MT(ASCP)

 SBB, American Red Cross, Southeastern Michigan Region

 Detroit, MI; and Marion E. Reid, PhD (corresponding author)

 Laboratory of Immunochemistry, New York Blood Center, 310 East 67th Street, New York, NY 10065.

 Attention:

State Blood Bank Meeting Organizers

If you are planning a state meeting and would like copies

of Immunohematology  for distribution, please send request,

4 months in advance, to [email protected]

Notice to Readers

All articles published, including communications and bookreviews, reect the opinions of the authors and do not

necessarily reect the ofcial policy of the American RedCross.

For information concerning the National Reference

Laboratory for Blood Group Serology, including the American

Rare Donor Program, contact Sandra Nance, by phone at

(215) 451-4362, by fax at (215) 451-2538, or by e-mail at

[email protected]

 Attention: SBB and BB Students

You are eligible for a free 1-year subscription to

Immunohematology.

Ask your education supervisor to submit the name and

complete address for each student and the inclusive dates

of the training period to [email protected]

Page 25: m4440090_27_2_11 Immunohematology

7/23/2019 m4440090_27_2_11 Immunohematology

http://slidepdf.com/reader/full/m444009027211-immunohematology 25/48IMMUNOHEMATOLOGY, Volume 27, Number 2, 2011 61

The use of massive transfusion protocols (MTPs) in

trauma patients presenting with near-exsanguinating

injuries has increased in the past several years. This is

largely a result of the increasing evidence suggesting that

transfusion of blood components in ratios more closely

approximating the composition of whole blood may reduce

mortality.1–4

  Inherent in the design of these protocolsis the necessity for the timely transfusion of plasma.

Unfortunately, the need for plasma often precedes the

determination of blood group.

Group AB plasma is commonly used in MTP when

the patient’s blood type has not yet been determined. The

foundation for this practice is rooted in the theory that

group AB plasma is “universal” because of the absence of

anti-A and anti-B hemagglutinins. Likewise, it is analogous

to the belief that group O red blood cells (RBCs) are

universal and may be transfused to all patients, regardless

of blood type. This universal blood component practice

continues to this day, based on concerns that infusion ofincompatible plasma may produce hemolysis, a potentially

fatal complication. Unfortunately, in the United States the

availability of group AB plasma is limited because only 4

percent of all blood donors in North America are group

 AB.5 Therefore, plasma from group AB donors may not be

available in sufcient quantities to satisfy the requirements

for an MTP in all trauma settings.

Historically, this risk of hemolysis has outweighed

concerns about the potential risks associated with

compatible but nonidentical plasma. Although experience

in populations receiving apheresis platelets has proved that

the concern about intravascular hemolysis is warranted,6–18

the belief that group AB plasma is universal plasma has

 been repeatedly challenged in several patient populations,

including those undergoing cardiac surgery,19  receiving

treatment for hematologic malignancies,20  or receiving

stem cell transplants.21 For patients in whom a hemolytic

reaction may occur, there are three important observations

to consider. First, minor incompatibilities appear to be

rare events, with one study suggesting an incidence of 1 in

9000.22  Second, in these anecdotal case reports in which

hemolytic reactions occurred, the suspected components

contained plasma from group O donors.8,23,24 After carefu

review of these case reports, no patient belonging to group

B or group AB was found to have experienced a hemolytic

event after transfusion with group A plasma. Third, and

most interestingly, it appears that correlations between

these hemolytic episodes and agglutinin titers exist.17,25–27

If the level of anti-A and anti-B agglutinin titers could

 be quantied in compatible but nonidentical plasma units

perhaps a novel approach to a plasma substitution could

 be made for MTPs. For example, if group AB plasma is

not available or is in short supply from local blood bank

sources, it may be feasible and preferable to use group A

plasma. Because approximately 15 percent of the North

 American population are group B or group AB, the use o

group A plasma is already acceptable for approximately 85

percent of the general population. Published experience

from different institutions indicates that transfusion of 300

to 500 mL of incompatible plasma in 1 day and transfusionof 1 L of incompatible plasma in 1 week have been practiced

 without signicant clinical sequelae in adult patients. In

addition, many institutions use a critical titer of 64 or higher

as the cutoff when saline solution is used for labeling a unit

as “high-titer.”24 Therefore, it would be logical to conclude

that it may be safe to transfuse 1 L of group A plasma to a

group B or group AB patient as long as the anti-B agglutinin

titer is less than 16.

The goal of this study is to identify acceptable group A

plasma units that could theoretically be safely transfused as

universal plasma to any patient during MTP. To determine

concept feasibility, we propose classifying group A plasma

donors into those with low titers (LT) of anti-B agglutinins

(≤ 64 but > 16) and those with very low titers (VLT) of anti-B

agglutinins (≤ 16). In the trauma setting, while a blood

group determination is being performed, it may be possible

to transfuse more than 1 unit of group A plasma during the

initial MTP shipments if group AB plasma is unavailable

Group A plasma meeting these specications could be

used to supplement the group AB plasma inventory in the

treatment of trauma patients requiring MTP. The solution

Challenging dogma: group A donors as“universal plasma” donors in massive

transfusion protocolsE.J. Isaak, K.M. Tchorz, N. Lang, L. Kalal, C. Slapak, G. Khalife, D. Smith, and M.C. McCarthy

REPORT

Page 26: m4440090_27_2_11 Immunohematology

7/23/2019 m4440090_27_2_11 Immunohematology

http://slidepdf.com/reader/full/m444009027211-immunohematology 26/4862  IMMU NOHEM ATOLOGY, Volum e 27, Numb er 2, 2011

E.J. Isaak et al.

proposed in this pilot study assumes established criteria

supported by the currently documented literature.

Materials and Methods

Fresh plasma (less than 48 hours old) was obtained

from the pilot tubes of 100 consecutive group A blood

donors during December 2008. All donors consented to

research-related activities as part of the general consent

procedure at the Community Blood Center/Community

Tissue Services (CBC/CTS), Dayton, Ohio, and all were

eligible donors based on an approved donor health

questionnaire. These documents were reviewed by the

Food and Drug Administration and are on le at the Center

for Biologics Evaluation and Research as required by the

Code of Federal Regulations. Demographic data including

sex, age, and history of previous pregnancy were recorded

from the information on the questionnaire.The plasma from each of these donors was diluted with

0.85 percent saline solution (Blood Bank Saline, Fisher

Scientic USA, Pittsburgh, PA) to prepare dilutions of 1:16

(1 part plasma and 15 parts saline) and 1:64 (1 part plasma

and 63 parts saline) in sufcient quantities for testing. A

freshly collected random group B donor unit was selected

as the testing RBCs. The group B RBCs were washed and

prepared as a 4% concentration in saline. One drop of the

4% group B RBCs was added to 0.2 mL of each of the 1:16

dilutions of the group A plasmas and mixed well at room

temperature (27°C). The mixtures were centrifuged and

read macroscopically for agglutination; reactions wererecorded as either negative (no agglutination observed)

or positive (any macroscopic agglutination observed,

regardless of strength). All tubes were then transferred

to a 37°C dry heat incubator and were allowed to incubate

for 30 minutes. The tubes were again centrifuged and read

macroscopically for agglutination with the same criteria

used for recording as negative or positive.

Similarly, one drop of the 4% group B RBCs was

added to 0.2 mL of each of the 1:64 dilutions of the group

 A plasmas and mixed well at room temperature. These

mixtures were also centrifuged and read macroscopically

for agglutination, and recorded as either negative or

positive using the criteria previously established. All tubes

 were then transferred to a 37°C dry heat incubator and

 were allowed to incubate for 30 minutes. The tubes were

centrifuged and read macroscopically for agglutination,

and recorded as either negative or positive using the criteria

previously established.

 All dilutions and testing were performed by one of

two experienced CBC/CTS immunohematology reference

laboratory technologists. The resulting patterns of

reactivity were analyzed for correlation with age, sex, and

history of pregnancy. Statistical analysis with the Pearson

chi-square test was then used to determine any correlation

 between the patterns and age. Signicance was determined

at α = 0.05.

Results

 Among the 100 plasma samples, seven different

patterns of reactivity were noted. Sixty three percent of

donor samples were negative at a titer of 64 both at room

temperature and at 37°C. These donor samples can be

classied as LT. Fifteen percent of donor samples were

negative at titers of 64 and 16 both at room temperature

and at 37°C and can be classied as VLT (Table 1).

 Although low titers and very low titers occur in both

sexes and across all age ranges, some general patterns do

emerge. The three most commonly observed patterns of

reactivity are (1) positivity at titers of 16 and 64 at room

temperature and 37°C (pattern A), (2) positivity at a titer o

16 at room temperature and 37°C but negativity at a titer of

64 at both temperatures (pattern B), and (3) negativity at

 both titers at both temperatures (pattern C).

The ratio of men to women within pattern A is 4:19;

the ratio is more evenly balanced within patterns B and C

The relationship of these three patterns of reactivity with

regard to age is noted in Table 2. There is a statistically

signicant difference (p < 0.029) in the prevalence of

patterns A and C in donors younger than 50 years of age

compared with donors who are older than 50 years. Pattern

 A is more common in younger donors, whereas pattern C is

more common in older donors.

Table 1. Donor sample demographic data and agglutinin titer

NMean age,y (range)

Male:f emal e Pregnancies R T(VLT ) RT( LT ) 37 °C (V LT ) 37 °C (LT )

23 39(17–75)

4:19 13/19 + + + +

11 47(19–71)

7:4 4/4 + – + +

39 51(20–72)

19:20 15/20 + – + –

5 48(43–51)

2:3 3/3 – – + –

3 53(50–56)

0:3 3/3 + + + –

4 53(46–59)

4:0 0/0 + – – –

15 54(38–77)

8:7 7/7 – – – –

LT = low titer ≤ 64; RT = room temperature 27°C; VLT = very low titer ≤ 16.

Page 27: m4440090_27_2_11 Immunohematology

7/23/2019 m4440090_27_2_11 Immunohematology

http://slidepdf.com/reader/full/m444009027211-immunohematology 27/48IMMUNOHEMATOLOGY, Volume 27, Number 2, 2011 63

Group A as universal plasma donors in massive transfusion protocols

Discussion

The results of this laboratory study demonstrate that

evaluation of anti-B agglutinin titers in group A plasma as

 VLT may serve to identify acceptable group A plasma unitsthat may be substituted in MTP when blood type has not yet

 been determined.

 As the demand for plasma resuscitation increases,

transfusion services are developing algorithms for

maintaining an inventory of thawed plasma. Because of the

relative scarcity of group AB plasma, in general, it is simply

not practical to maintain thawed group AB plasma. This

translates into a delay in plasma distribution because the

process of obtaining a blood bank specimen, determining

a blood type, and issuing the plasma can be challenging.

In addition, group AB plasma may involve additional risks

 because of the formation of circulating immune complexes,28 especially when transfused to group O patients. In fact, in

one large study mortality was increased whenever type-

compatible but not type-specic plasma was transfused.

However, statistical signicance was reached only when

group AB plasma was given to group O patients, and in

this case when more than ve units of group AB plasma

 were transfused to a group O patient, the relative risk of

increased mortality was 1.26 (p = 0.004).29  Thus, even if

group AB plasma were available in sufcient amounts, it

might not be ideal. A possible solution to this dilemma is

to identify a type of plasma that is present in abundance

and can be safely transfused to any trauma patient, at least

up to a certain volume, thus allowing sufcient time for a

 blood group determination to be made. Group A plasma

is relatively abundant, and because nearly 85 percent of

trauma patients are group A or group O, they can safely

receive group A plasma. For the 15 percent of patients for

 whom the transfusion of group A plasma creates a minor

incompatibility, published experience has demonstrated

that reactions caused by minor incompatibilities are

extremely rare.

Multiple reviews in the published literature have

provided details about the anecdotal cases in which a

minor incompatibility has resulted in a serious adverse

outcome. The overwhelming majority of these cases involve

group O plasma transfused to a group A patient, although

there are sporadic instances in which group O plasma was

transfused to a group B patient.8,23,24  The reports warn

that the issue may be underreported and that physicians

should be vigilant when transfusing incompatible plasma

Nonetheless, the preponderance of the published clinica

experience seems to indicate that the transfusion of

incompatible group A plasma appears to be safe.

Despite the lack of a reported problem with the

transfusion of group A plasma to either a group B or a

group AB patient, additional factors support using group A

plasma. There appears to be a correlation between adverse

clinical consequences and high ABO agglutinin titers, which

provides an avenue for interventions to deal with the issue othe rare but potentially signicant minor incompatibilities

Some transfusion services are identifying agglutinin titers

 before issuing platelet components containing incompatible

plasma.23,26 Evaluation of anti-B agglutinin titers in group

 A plasma may serve to identify acceptable group A plasma

units that could theoretically be safely transfused to any

patient. In determining these titers, the laboratory method

is important, and various laboratories have used tube

agglutination, gel technology, or microtiter plates in these

determinations.30  In our pilot study, agglutination titers

 were determined using tube agglutination, and the titers

 we used as thresholds were based on relatively conservative values from reports in the platelet literature in which

tube agglutination was used. A recent review containing

an extensive summary of hemolytic reactions associated

 with minor incompatibility in patients transfused with

platelets indicated that the range of titers fell between 128

as a minimum in saline up to 32,000 using antiglobulin

reagent.24 The rationale for the use of titers in our study was

to base our proposed solution on conservative assumptions

 with the use of available platelet transfusion data. It may

turn out that these assumptions are overly conservative

 Another element that is important in the laboratory

method is the choice of RBCs used to perform the titer.

 We used RBCs from a random group B whole blood donor

Because of variation in antigen sites in group A RBCs, titers

are best performed using blended pools of group A RBCs

it is not clear that the same issue exists in group B RBCs

although further investigation of this open question may be

 warranted.

In the preceding paragraph, the conservative

assumptions are as follows: (1) group A plasma will be

statistically compatible with 85 percent of patients, (2)

Table 2. Donor sample age group with agglutinin titer patternassociation

Pattern

Age Group A B C Total

< 50 years Count 17 18 5 40

% within age group 42.5% 45.0% 12.5% 100.0%

≥ 50 years Count 6 21 10 37

% within age group 16.2% 56.8% 27.0% 100.0%

Total Count 23 39 15 77

% within age group 29.9% 50.6% 19.5% 100.0%

Page 28: m4440090_27_2_11 Immunohematology

7/23/2019 m4440090_27_2_11 Immunohematology

http://slidepdf.com/reader/full/m444009027211-immunohematology 28/4864  IMMU NOHEM ATOLOGY, Volum e 27, Numb er 2, 2011

E.J. Isaak et al.

minor incompatibilities are extremely rare, (3) there has

 been no reported case of a minor reaction with group A

plasma in either a group B or group AB patient, (4) serious

adverse reactions can be associated with certain threshold

agglutinin levels, and (5) conservative values from evidence-

 based publications were used to set an acceptable agglutinin

titer threshold level. Based on these assumptions, our study

showed that 63 percent of our donors could safely donate

one unit of incompatible plasma and that 15 percent could

safely donate up to 1 L of incompatible plasma to any patient.

Furthermore, we found that the VLT donors are more

common in donors older than 50 years of age. Therefore,

a possible recruitment strategy in our donor population

 would involve screening donors older than 50 years of age

to establish a dedicated group of VLT donors for MTP.

 With regard to the limitations of this pilot study, there

are several. First, the donor sample size is only 100. This

number was selected because the purpose of the study was merely to test for feasibility of the proposed approach.

Second, the donor population is a midwest population,

and therefore these results may not extrapolate to other

donor populations. The determinants of agglutinin

levels appear to be largely determined by geography and

environmental factors,31,32 and one study of populations in

 Asia found higher agglutinin levels in donors older than

50 years of age.33 Although it is well known that immune

function deteriorates with increasing age, this population

had increased agglutinin titers. Our results of a midwest

North American population demonstrated the opposite.

Surprisingly, our population showed titer levels that appearto be independent of history of pregnancy. The signicance

of these ndings remains to be determined.

Because populations of blood donors will have different

patterns of agglutinin titers based on ethnicity and age,

select blood centers could develop specic recruitment

strategies to provide these VLT universal plasma products

to meet local and regional needs. Finally, and most

importantly, this new concept of VLT group A plasma as

universal plasma in MTP has not been studied in trauma

patients requiring MTP and will require thoughtful study

design to test the hypothesis.

Conclusions

The results of this study suggest a fresh perspective

on resuscitation in trauma patients. When a massively

injured trauma patient arrives at the trauma center, it is

theoretically possible to initiate MTP with thawed VLT

group A plasma instead of thawed group AB plasma.

Because MTP suggests a ratio of one RBC unit to one

plasma unit during acute hemorrhage, the time required to

transfuse the initial MTP shipment of four RBC units and

four plasma units (essentially equivalent to 1 L of plasma)

 would provide transfusion services with ample time to

determine blood type and then issue the appropriate type-

specic blood components. When handled in this fashion

group AB plasma may remain frozen and only be thawed

for use in those patients with group AB blood.

References

  1. Borgman MA, Spinella PC, Perkins JG, et al. The ratio o blood products transfused affects mortality in patientsreceiving massive transfusions at a combat support hospitalJ Trauma 2007;63:805–13.

  2. Bormanis J. Development of a massive transfusion protocolTransfus Apher Sci 2008;38:57–63.

  3. Gonzalez EA, Moore FA, Holcomb JB, et al. Fresh frozenplasma should be given earlier to patients requiring massivetransfusion. J Trauma 2007;62:112–19.

  4. Huber-Wagner S, Qvick M, Mussack T, et al. Massive bloodtransfusion and outcome in 1062 polytrauma patients: aprospective study based on the Trauma Registry of theGerman Trauma Society. Vox Sang 2007;92:69–78.

  5. Roback JD, Combs MR, Grossman BJ, et al (eds). Technicamanual. 16th ed. Arlington, VA: American Association oBlood Banks, 2008.

  6. McLeod BC, Sassetti RJ, Weens JH, Vaithianathan THaemolytic transfusion reaction due to ABO incompatibleplasma in a platelet concentrate. Scand J Haematol 1982;28:193–6.

  7. Larsson LG, Welsh VJ, Ladd DJ. Acute intravascularhemolysis secondary to out-of-group platelet transfusionTransfusion 2000;40:902–6.

  8. McManigal S, Sims KL. Intravascular hemolysis secondaryto ABO incompatible platelet products. An underrecognizedtransfusion reaction. Am J Clin Pathol 1999;111:202–6.

  9. Murphy MF, Hook S, Waters AH, et al. Acute haemolysisafter ABO-incompatible platelet transfusions. Lancet 1990335:974–5.

  10. Pierce RN, Reich LM, Mayer K. Hemolysis following plateletransfusions from ABO-incompatible donors. Transfusion1985;25:60–2.

  11. Sadani DT, Urbaniak SJ, Bruce M, Tighe JE. Repeat ABO-incompatible platelet transfusions leading to haemolytictransfusion reaction. Transfus Med 2006;16:375–9.

  12. Sapatnekar S, Sharma G, Downes KA, Wiersma S, McGrath

C, Yomtovían R. Acute hemolytic transfusion reactionin a pediatric patient following transfusion of apheresisplatelets. J Clin Apher 2005;20:225–9.

  13. Oztürk A, Türken O, Sayan O, Atasoyu EM. Acuteintravascular hemolysis due to ABO-incompatible platelettransfusion. Acta Haematol 2003;110:211–12.

  14. Barjas-Castro ML, Locatelli MF, Carvalho MA, Gilli SOCastro V. Severe immune haemolysis in a group A recipientof a group O red blood cell unit. Transfus Med 2003;13239–41.

Page 29: m4440090_27_2_11 Immunohematology

7/23/2019 m4440090_27_2_11 Immunohematology

http://slidepdf.com/reader/full/m444009027211-immunohematology 29/48IMMUNOHEMATOLOGY, Volume 27, Number 2, 2011 65

Group A as universal plasma donors in massive transfusion protocols

  15. Chow MP, Yung CH, Hu HY, Tzeng CH. Hemolysis after ABO-incompatible platelet transfusions. Zhonghua Yi XueZa Zhi (Taipei) 1991;48:131–4.

  16. Ferguson DJ. Acute intravascular hemolysis after a platelettransfusion. CMAJ 1988;138:523–4.

  17. Harris SB, Josephson CD, Kost CB, Hillyer CD. Nonfatalintravascular hemolysis in a pediatric patient after

transfusion of a platelet unit with high-titer anti-A.Transfusion 2007;47:1412–17.

  18. Valbonesi M, De Luigi MC, Lercari G, et al. Acuteintravascular hemolysis in two patients transfused withdry-platelet units obtained from the same ABO incompatibledonor. Int J Artif Organs 2000;23:642–6.

  19. Blumberg N, Heal JM, Hicks GL Jr, Risher WH. Associationof ABO-mismatched platelet transfusions with morbidityand mortality in cardiac surgery. Transfusion 2001;41: 790–3.

  20. Heal JM, Kenmotsu N, Rowe JM, Blumberg N. A possiblesurvival advantage in adults with acute leukemia receiving ABO-identical platelet transfusions. Am J Hematol 1994;45:189–90.

  21. Heal JM, Liesveld JL, Phillips GL, Blumberg N. What wouldKarl Landsteiner do? the ABO blood group and stem celltransplantation. Bone Marrow Transplant 2005;36:747–55.

  22. Mair B, Benson K. Evaluation of changes in hemoglobinlevels associated with ABO-incompatible plasma inapheresis platelets. Transfusion 1998;38:51–5.

  23. Fung MK, Downes KA, Shulman IA. Transfusion ofplatelets containing ABO-incompatible plasma: a survey of3156 North American laboratories. Arch Pathol Lab Med2007;131:909–16.

  24. Cooling L. ABO and platelet transfusion therapy.Immunohematology 2007;23:20–33.

  25. Eder AF, Chambers LA. Noninfectious complications of

 blood transfusion. Arch Pathol Lab Med 2007;131:708–18.  26. Josephson CD, Mullis NC, Van Demark C, Hillyer CD.

Signicant numbers of apheresis-derived group O plateletunits have “high-titer” anti-A/A,B: implications fortransfusion policy. Transfusion 2004;44:805–8.

  27. Reesink HW, van der Hart M, van Loghem JJ. Evaluation ofa simple method for determination of IgG titre anti-A or -Bin cases of possible ABO blood group incompatibility. VoxSang 1972;22:397–407.

  28. Heal JM, Masel D, Rowe JM, Blumberg N. Circulatingimmune complexes involving the ABO system after platelettransfusion. Br J Haematol 1993;85:566–72.

  29. Shanwell A, Andersson TM, Rostgaard K, et al. Post-transfusion mortality among recipients of ABO-compatible but non-identical plasma. Vox Sang 2009;96:316–23.

  30. Cooling LL, Downs TA, Butch SH, Davenport RD. Anti-A

and anti-B titers in pooled group O platelets are comparableto apheresis platelets. Transfusion 2008;48:2106–13.

  31. Redman M, Malde R, Contreras M. Comparison of IgMand IgG anti-A and anti-B levels in Asian, Caucasian andNegro donors in the North West Thames Region. Vox Sang1990;59:89–91.

  32. Toy PT, Reid ME, Papenfus L, Yeap HH, Black D. Prevalenceof ABO maternal-infant incompatibility in Asians, BlacksHispanics and Caucasians. Vox Sang 1988;54:181–3.

  33. Mazda T, Yabe R, NaThalang O, Thammavong T, TadokoroK. Differences in ABO antibody levels among blood donorsa comparison between past and present Japanese, Laotianand Thai populations. Immunohematology 2007;23:38–41

 Edahn J. Isaak, MD, FACP, Director of Transfusion Medicine

 Staten Island University Hospital, Staten Island, NY; Kathryn M

Tchorz, MD, FACS (corresponding author), Associate Professor

 Department of Surgery-Division of Trauma/Critical Care

Wright State University-Boonshoft School of Medicine, Miam

Valley Hospital, One Wyoming Street, 7th Floor CHE Building

 Dayton, OH 45409; Nancy Lang, SBB, Immunohematology

 Reference Technologist, Lawrence Kalal, MT, Immunohematol

ogy Reference Technologist, Colleen Slapak, MS, Transfusion

 Safety Director, Ghada Khalife, MD, Medical Director, David

 Smith, MD, CEO, Community Blood Center/Community Tissue

 Services, Dayton, OH; and Mary C. McCarthy, MD, FACS

 Professor, Department of Surgery, Chief, Trauma/Critical CareWright State University-Boonshoft School of Medicine, Miam

Valley Hospital, Dayton, OH.

The editorial staff of Immunohematology  welcomes manuscriptspertaining to blood group serology and education for considerationfor publication. We are especially interested in case reports,papers on platelet and white cell serology, scientic articlescovering original investigations, and papers on new methods foruse in the blood bank. Deadlines for receipt of manuscripts forconsideration for the March, June, September, and December

issues are the rst weeks in November, February, May, andAugust, respectively. For instructions for scientic articles, casereports, and review articles, see Instructions for Authors in everyissue of Immunohematology  or on the Web at www.redcross.org/immunohematology. Include fax and phone numbers and

e-mail address with all manuscripts and correspondence. E-mail all manuscripts to [email protected]

Manuscripts

Page 30: m4440090_27_2_11 Immunohematology

7/23/2019 m4440090_27_2_11 Immunohematology

http://slidepdf.com/reader/full/m444009027211-immunohematology 30/4866  IMMU NOHEM ATOLOGY, Volum e 27, Numb er 2, 2011

The alleles  RHCE*ceBI (RHCE*ce 48C, 712G, 818T, 1132G) and  RHCE*ceSM (RHCE*ce 48C, 712G, 818T)  encode the low-prevalence Rh antigen STEM. These alleles frequently travel incis with RHD*DOL. To estimate the frequency of these alleles, wetested a total of more than 700 samples in two populations. Bloodsamples were obtained from patients with sickle cell diseaseand from blood donors of African descent. DNA extractions andanalyses were performed by standard methods. In the UnitedStates, none of 70 patient samples had the RHCE*818 nucleotidechange. Two of 220 donors (frequency of 0.009) were heterozygousfor  RHCE*818C/T (RHCE*ceBI).  One of these samples had RHD/RHD*DOL and the other had RHD/RHD*DOL-2.  In these290 samples, no other  RHD*DOL  alleles were found. In Brazil,

1 of 244 patients with sickle cell disease (frequency of 0.004)and 1 of 171 donors (frequency of 0.006) were heterozygous for RHCE*818C/T (RHCE*ceBI). Testing of more than 500 additionalsamples from people of African descent, selected because theyhad a diverse range of common and variant  RHCE   alleles, didnot reveal a sample with RHD*DOL or  RHD/RHD*DOL-2 in theabsence of  RHCE*ce(818T).  Although the numbers are small,our study shows that in the United States, the frequency of RHCE*818T  is 0.007 (2 in 290 samples) and in Brazil it is 0.004

(2 in 515 samples). The four RHCE*818T  alleles were RHCE*ceBI.  Immunohematology 2011;27:66–67.

Key words:  blood groups, Rh alleles, low-prevalence

antigens, population study 

The low-prevalence Rh antigen STEM (RH49) was

reported in 1993. The antigen was serologically shown

to be associated with an altered e phenotype because

approximately 65 percent of hrS– and 30 percent of hrB–

red blood cells (RBCs) from South African donors were

reported to be STEM+. STEM has a variable expression,

 which is an inherited characteristic. Anti-STEM has

induced mild hemolytic disease of the fetus and newborn.1 

Owing to the paucity of anti-STEM and typed RBCs, little

further work has been done and no other serologic reports

have appeared in the literature.

The STEM antigen is encoded by two RHCE alleles:

 RHCE*ceBI (RHCE*ce 48C, 712G, 818T, 1132G)  and

 RHCE*ceSM (RHCE*ce 48C, 712G, 818T)  (Fig. 1A). The

nucleotide (nt) 818C>T change is believed to be associated

 with expression of STEM, because the other nucleotide

changes occur in other alleles.2  In analyzing DNA from

samples known to be STEM+ or DAK+, it was revealed

that these variant RHCE  are usually in cis to RHD*DOL or

 RHD*DOL-2 (Fig. 1B).3 We tested more than 700 samples

in New York and in Brazil to estimate the frequency of

 RHCE*ce818C>T  and to investigate RHD in these samples.

Prevalence of RHD*DOL and RHCE*ce(818T) in two populations

C. Halter Hipsky, D.C. da Costa, R. Omoto, A. Zanette, L. Castilho, and M.E. Reid

REPORT

Figure 1. Diagram of selected RHCE  ( A) and RHD (B) alleles. The10 exons are numbered. Nucleotide changes are indicated in italicsand amino acids are given in parentheses below the exon in whichthey are found. Changes from the wild-type nucleotides and aminoacids are written in bold.

B. RHD alleles

 A. RHCE alleles

Page 31: m4440090_27_2_11 Immunohematology

7/23/2019 m4440090_27_2_11 Immunohematology

http://slidepdf.com/reader/full/m444009027211-immunohematology 31/48IMMUNOHEMATOLOGY, Volume 27, Number 2, 2011 67

Prevalence of RHD*DOL and RHDCE*ce(818T)

Materials and Methods

Blood samples were obtained from patients with sickle

cell disease and from blood donors who self-identied as

 being of African descent. Genomic DNA was isolated from

the whole blood samples using the QIAamp DNA Blood

Mini Kit (QIAGEN, Inc., Valencia, CA). Initial screening of

samples was carried out by sequencing  RHCE- and RHD- 

reverse transcriptase–polymerase chain reaction (PCR)

products, as described previously,4 or analyzing RHCE*818 

and RHCE*1132 by standard PCR products generated with

genomic DNA using  RHCE-specic primers, followed by

restriction fragment length polymorphism (RFLP) using,

respectively,  MwoI and Tsp45 I restriction enzymes.

Genomic DNA was amplied using  RHD-specic primers

anking exon 4 and exon 8, and products were submitted

for sequencing to determine the presence of  RHD*DOL (nt

509T>C in exon 4) and RHD*DOL-2 (nt 509T>C in exon 4and nt 1132C>G in exon 8). PCR products were sequenced

in both directions.

Results

In the United States, none of 70 patient samples had

the  RHCE*818  nucleotide change. Two of 220 donors

(frequency of 0.009) were heterozygous for RHCE*818C/T. 

Both samples had the change at  RHCE*ce1132C>G   and,

thus, are  RHCE*ceBI.  One sample had  RHD/RHD*DOL 

and the other had  RHD/RHD*DOL-2.  No homozygous

 RHCE*818T   sample was found. In these 290 samples, noother RHD*DOL alleles were found.

In Brazil, 1 of 244 patients with sickle cell disease

(frequency of 0.004) and 1 of 171 donors (frequency of

0.006) were heterozygous for  RHCE*818C/T   and for

 RHCE*ce1132C/G   and, thus, are also  RHCE*ceBI.  The

samples were not analyzed for RHD.

Taking the combined data, 4 samples in a total of 705

tested had  RHCE*818C/T   (all 4 were  RHCE*ceBI ), which

gives an allele frequency of 0.006.

Conclusions

 Although the numbers are small, our study shows

that in the United States, the frequency of  RHCE*818T

(RHCE*ceBI)  is 0.007 (2 in 290 samples; 580 alleles)

and in Brazil it is 0.004 (2 in 415 samples; 830 alleles)

Our ndings show that  RHCE*818T,  which encodes the

STEM antigen, is not as rare as previously thought. This

raises the question as to whether anti-STEM is also more

prevalent but not identied. In the cohort of samples tested

the RHD*DOL or  RHD*DOL-2 alleles were in cis with the

 RHCE*ce(818T) allele; no  RHD*DOL or  RHD*DOL-2 was

found in the absence of RHCE*ce(818T).

 Acknowledgments

This study was supported in part by grant NIH

HL091030 (CHH, MER) and INCTS-CNPQ/MCT/FAPESP

(LC). We thank Robert Ratner for help in preparing this

manuscript.

References

  1. Marais I, Moores P, Smart E, Martell R. STEM, a newlow-frequency Rh antigen associated with the e-varianphenotypes hrS-(Rh: -18, -19) and hrB-(Rh: -31, -34)Transfus Med 1993;3:35–41.

  2. Halter-Hipsky C, Hue-Roye K, Coghlan G, Lomas-FrancisC, Reid ME. Two alleles with RHCE*nt818C>T changeencode the low prevalence Rh antigen STEM [abstract]Blood 2009;114(Suppl):1226–7.

  3. Halter Hipsky C, Hue-Roye K, Lomas-Francis C, Reid MERHD*DOL travels with  RHCE*ce(818C>T),  which encodesa partial e, hrS–, hrB+, STEM+ phenotype [abstract]Transfusion 2010;50(Suppl):48A.

  4. Halter Hipsky C, Lomas-Francis C, Fuchisawa A, et al RHCE*ceCF  encodes partial c and partial e but not CELO, anantigen antithetical to Crawford. Transfusion 2011;51:25-3

Christine Halter Hipsky, Laboratory of Immunochemistry, New

York Blood Center, New York, NY; Daiane Cobianchi da Costa,

 MS, Ricardo Omoto, MS, Angela Zanette, MD, Lilian Castilho

 PhD, Hemocentro-Unicamp-Campinas-Brazil, Campinas

 SP, Brazil; and Marion E. Reid, PhD (corresponding author)

 Laboratory of Immunochemistry, New York Blood Center, 310

 East 67th Street, New York, NY 10065.

Notice to Readers

 Immunohematology is printed on acid-free paper.

For information concerning Immunohematology  or theImmunohematology Methods and Procedures manual,contact us by e-mail at [email protected]

Page 32: m4440090_27_2_11 Immunohematology

7/23/2019 m4440090_27_2_11 Immunohematology

http://slidepdf.com/reader/full/m444009027211-immunohematology 32/4868  IMMU NOHEM ATOLOGY, Volum e 27, Numb er 2, 2011

The XG blood group system is best known for its contributionsto the elds of genetics and chromosome mapping. This systemcomprises two antigens, Xga and CD99, that are not antithetical

 but that demonstrate a unique phenotypic relationship.  XG   islocated on the tip of the short arm of the X chromosome withexons 1 to 3 present in the pseudoautosomal region of the X(and Y) chromosome(s) and exons 4 to 10 located only on theX chromosome. Xga  demonstrates a clear X-linked pattern ofinheritance. MIC2, the gene encoding the CD99 antigen, is foundin the pseudoautosomal region of both the X and Y chromosomes. Anti-Xga  is comparatively rare, and only two examples of anti-CD99 have ever been identied. Alloanti-Xga  is considered

clinically insignicant; only one example of autoanti-Xga has beenreported, but it resulted in severe hemolytic anemia. Insufcientdata exist to determine the clinical signicance of anti-CD99.Linkage of  XG   to several X-borne genes encoding inheriteddisorders has been demonstrated. CD99 is an adhesion molecule,and high levels are associated with some types of cancer. Immunohematology 2011;27:68–71.

Key Words: Xga, X-linked, CD99, XG, MIC2, PBDX, 12E7,

Lyonization, qualitative polymorphism, pseudoautosomal

History

The XG blood group system is not revered for itsantigens and antibodies, but is instead respected for its

contributions to the elds of genetics and chromosome

mapping. XG is the rst, and so far the only, blood group

system to be assigned to the X chromosome.1  There are

only two antigens in the system: Xga and CD99. Xga may be

expressed only on red blood cells (RBCs), whereas CD99 is

expressed on many tissue cells. Anti-Xga can be naturally

occurring and is not considered clinically signicant. Only

two examples of alloanti-CD99 have ever been reported; its

clinical signicance is unknown.2

The rst example of anti-Xga  was reported in 1961 in

the serum of a man who had been multiply transfused for

severe nosebleeds as a result of familial telangiectasia.3 

Shortly thereafter, in 1962, Mann et al. characterized Xga 

as originating from a locus on the X chromosome.1 Xga  is

named after the X chromosome and Grand Rapids, where

the rst-reported patient to have the antibody (Mr. And) was

treated.4 It was later discovered that PBDX, a gene found to

span the pseudoautosomal boundary on the X chromosome,

is the gene that encodes Xga (see Molecular Basis section).5 

The function of the Xga protein is not known.6

Two decades after the rst example of anti-Xga  was

reported, Goodfellow and Tippett observed a mouse

monoclonal antibody, 12E7, which recognized another

determinant that appeared to be controlled by a gene also

found on the X chromosome.7 This gene,  MIC2,  is located

in the pseudoautosomal region of both the X and the Y

chromosomes and encodes CD99. CD99 is an adhesion

molecule,5,8  and high levels are associated with some

types of cancer. Xga and CD99 enjoy a unique phenotypic

relationship, which will be discussed in further detail in

the Genetics and Inheritance section. Fourteen years laterin 1995, Uchikawa et al. reported the only examples of

alloanti-CD99, detected in two unrelated, healthy Japanese

 blood donors.2 CD99, became part of the XG blood group

system ofcially in 2000 when it was conrmed that  MIC2

and XG  are adjacent, homologous genes.4

Because of its location in the pseudoautosomal boundary

region of the sex chromosome, study of the inheritance of

Xga  has helped to dene the mechanisms responsible for

Turner, Klinefelter, and other syndromes resulting from an

abnormal number of sex chromosomes. Sex chromosome

aneuploidies are commonly associated with infertility.

Nomenclature

The XG blood group system has been assigned the

ISBT symbol and number XG and 012, respectively. The

ISBT terminology for the two antigens in the system, Xg

and CD99, is listed in Table 1. No antithetical counterpart

to the Xga  antigen has been discovered, so the failure of

RBCs to react with anti-Xga denes the silent allele, Xg, or

an absence of Xga.

Genetics and Inheritance

Xga  is inherited as an X-linked, dominant trait, and

the pattern of inheritance is as expected for an X-borne

characteristic.1,8 If  Xga is present, it is expressed on RBCs

XG: the forgotten blood group systemN.C. Johnson

REVIEW

Table 1. Nomenclature

Xga CD99

ISBT symbol XG1 XG2

ISBT number 012001 012002

Page 33: m4440090_27_2_11 Immunohematology

7/23/2019 m4440090_27_2_11 Immunohematology

http://slidepdf.com/reader/full/m444009027211-immunohematology 33/48IMMUNOHEMATOLOGY, Volume 27, Number 2, 2011 69

 XG blood group system: a review

The Xg(a+) phenotype is found in 88.7 percent of females

and 65.6 percent of males. (Table 2.) Females can have a

single (Xga/Xg) or a double dose (Xga/Xga) of Xga. Having

only one X chromosome, males can be hemizygous for Xga 

( Xga /Y ) or be Xg/Y  and not express the antigen at all.6

Lyonization is a process by which one X chromosome

is inactivated early in somatic cell development in XX

females.9  The process is random in each cell, ensuring

that all genes on the two X chromosomes are found at the

phenotypic level. By studying RBCs of Xga/Xg women it can

 be shown that all cells carry Xga, proving that Xga  is not

subject to inactivation or Lyonization.  XG   was the rst X

chromosome locus proven to escape this process.10  It was

later shown that MIC2 is not subject to Lyonization either.11

New Guineans, Australian aborigines, Navajo Indians,

and Sardinians are reported to have the highest prevalenceof Xga. On the other hand, native Taiwanese; Chinese in

Singapore, Hong Kong, Taiwan, and Hakka; and Malays in

Singapore are observed to have the lowest incidence.3,12–14 

Interestingly, Asian populations with a higher prevalence of

Xg(a–) phenotypes do not have a higher incidence of anti-

Xga.6

Using monoclonal antibodies, it has been shown that

CD99 is present on all tissue cells tested15; however, the

expression of the adhesion molecule on RBCs is unique.

CD99 is expressed at different levels on RBCs, and this

expression is directly related to the presence or absence of

Xga .16 In females, the Xg(a–) phenotype is always associated

 with low expression of CD99. (Table 3.) Curiously, among

Xg(a–) males, 74 percent are high expressors of CD99.17 

To explain this quantitative polymorphism, Goodfellow

and Tippett proposed the existence of a locus on the Y

chromosome, YG, analogous to XG  on the X chromosome.

Like XG, YG  would have two alleles, Yga and Yg. In Xg(a–)

males, the presence of Yga would result in high expression

of CD99, and the absence of Yga  (Yg) would result in low

expression of CD99.7,16

The presence of YG   has been substantiated by family

studies,17  but to date, there has been no Yga  antigen

identied. It is important to note that  MIC2 is responsible

for expression of CD99 on all tissue cells, but  XG  and YG

create the CD99 quantitative polymorphism observed on

RBCs.8

Molecular Basis

Sex chromosomes have two genetically distinct regions

sex chromosome–specic sequences and pseudoautosoma

sequences. Pseudoautosomal refers to a structurally

homologous region on sex chromosomes, the function

of which is to facilitate pairing during male meiosis.8  XG

spans the pseudoautosomal boundary between the two

regions of the X chromosome at Xp22.3; exons 1 to 3 are

located in the pseudoautosomal region (PAR) and exons 4

to 10 are found in the sex chromosome–specic region. 4–6

 MIC2, the closest neighbor to XG, is located in the PAR at

position Xp22.2. An identical copy of MIC2 is found in the

PAR region of the Y chromosome at Yp11.2.4 Almost half(48%) of the predicted amino acid sequences of  XG   and

 MIC2 are identical.18

Biochemistry

Immunoblotting of immunoprecipitates conrmed that

CD99 and Xga antigens are located on different structures

later conrmed to be sialoglycoproteins.18,19 CD99 and Xg

are associated in the membrane, possibly in the form of a

heterodimer.7,18

The RBC membrane molecule expressing Xga was rst

identied in 1989 by Herron and Smith, by immunoblotting

and electropheresis.19 Several years later, Petty and Tippett

used immunoblotting and immunoprecipitation to show tha

Xga is carried on a broad band of apparent molecular weight

24.5 to 29.5 kDa, consisting of a darkly stained component

at 24.5 kDa and a more diffusely stained component

 between 26.5 and 29.5 kDa.18 No bands are observed with

protease-treated cells, and the apparent molecular weight

of Xga  is decreased after treatment with neuraminidase

suggesting that Xga resides on a sialoglycoprotein.

Table 2. Phenotype prevalence and genotype frequencies

Male (%) Female (%)

Phenotype

Xg(a+) 65.6 88.7

Xg(a–) 34.4 11.3

Genotype

 Xga/Xga 0.434

 Xga/Xg 0.450

 Xg/Xg 0.116

 Xga/Xg 0.656

 Xg/Xg 0.344

Table 3. Relationship of Xga, Yga, and CD99

Females Xg(a+) CD99 high

Xg(a+w) CD99 high

Xg(a–) CD99 low

Males Xg(a+) CD99 high

Xg(a–) Yga CD99 high

Xg(a–) Yg CD99 low

Page 34: m4440090_27_2_11 Immunohematology

7/23/2019 m4440090_27_2_11 Immunohematology

http://slidepdf.com/reader/full/m444009027211-immunohematology 34/4870  IMMU NOHEM ATOLOGY, Volum e 27, Numb er 2, 2011

N.C. Johnson

Xga is sensitive to treatment with cin, papain, trypsin,

 bromelin, α-chymotrypsin, and pronase, and is resistant to

sialidase, 0.2 M dithiothreitol (DTT), neuraminidase, and

2-aminoethylisothiouronium bromide.4,13,20  The effects of

acid are unknown. The expression of the antigen decreases

as the RBC ages and it has been shown by indirect

radioimmunoassay to have an in vivo half-life of 47 days.21

Early experiments using a microcomplement xation

method and indirect radioimmunoassay suggested that

Xga might be present on other cell lines and tissues,22 but

subsequent testing was unable to duplicate these results.

 XG   is expressed on hematopoietic tissues8 and is strongly

expressed as mRNA in broblasts.20

 Apart from humans, the only animals found to have

Xga on their RBCs are one species of gibbons. Of 52 gibbons

tested, 30 percent of males and 53 percent of females were

Xg(a+). Chimpanzees (67); gorillas (2); orangutans (20);

another species of gibbons (5); various monkeys, including60 baboons; and a few nonprimates, including mice and

dogs, were all Xg(a–).3,4

Monoclonal anti-CD99 identify a protein of apparent

molecular weight of 32 kDa.5,8  Immunoblotting and

immunoprecipitate studies suggest the CD99 protein is

composed of two polypeptides, a 32-kDa surface compo-

nent, and a 30-kDa intracellular polypeptide.18  CD99

is sensitive to treatment with trypsin, α-chymotrypsin,

cin, papain, and pronase15  and, like Xga, is carried on a

sialoglycoprotein. CD99 is resistant to 0.2 M DTT and is

usually resistant to neuraminidase treatment; results vary

 with sialidase treatment.CD99 is present on all human cell types and tissues

tested.15 High levels of CD99 are a tumor marker in Ewing

sarcoma, some neuroectodermal tumors, lymphoblastic

lymphoma, and acute lymphoblastic leukemia.8 CD99 was

not detected on the RBCs or peripheral blood lymphocytes

of 10 gibbons, regardless of their Xga  phenotype. CD99

 was detected on RBCs and broblasts of chimpanzees and

gorillas, but not of orangutans or any of the other mammals

tested.23

 Antibodies in the System

 Anti-Xga  is comparatively rare; most immunohema-

tologists will never see anti-Xga  in their careers. When

identied, it is usually found as a lone antibody specicity.

Some examples of anti-Xga  are naturally occurring, yet

are more frequently IgG than IgM.4  Optimal methods for

identication include room temperature incubation (even

 when they are IgG in composition3), indirect antiglobulin

test (IAT), and capillary testing.4  One IAT-reactive anti-

Xga was shown to have IgG1 and IgG2 subclasses.24 Some

examples of anti-Xga have been shown to x complement.

The only reported example of autoanti-Xga  was found in

a pregnant woman. The antibody appears to have caused

signicant hemolytic anemia, but the possibility that the

hemolytic anemia was exacerbated by the pregnancy was

not ruled out.25

 As a result of the sex-related frequency differences o

Xga, it is expected that men would make more examples of

anti-Xga. Although this is true, it is striking that greater

than 85 percent of antibody makers are men when the

incidence of the Xg(a–) phenotype is only 23 percent higher

in men than in women. It has been postulated, although

not studied, that this higher than expected prevalence

of anti-Xga  in men may be associated with the expression

of CD99.6

The rst example of anti-CD99 ever made was a mouse

monoclonal antibody called 12E7, which was generated to

screen for antigen differences in human acute lymphocyticleukemia.26  The only two examples of anti-CD99 ever

detected were found in two unrelated, healthy Japanese

 blood donors. One antibody was reported to react with

 variable strength with different RBC samples. The second

 blood donor had a history of pregnancy. One of the two

siblings of the second blood donor was determined to be

CD99 negative also.2  The antibodies were determined to

 be IgG in nature and reacted optimally by the IAT.5  It is

not known whether the antibodies were capable of binding

complement.

Clinical Significance

 Alloanti-Xga  has never been reported to cause

hemolytic disease of the fetus or newborn or a hemolytic

transfusion reaction.4  One patient received six units of

antigen-positive blood with no signs of a reaction.27  A

chromium survival study on another patient showed norma

survival of Xg(a+) RBCs and no posttransfusion increase

in antibody strength.28  One male Japanese patient had a

slight temperature elevation and exhibited urticaria when

transfused with Xg(a+) blood. Subsequent transfusions

 with Xg(a–) blood were tolerated with no symptoms.29

The one example of autoanti-Xga  reported was

identied in a pregnant female. The antibody was IgG in

nature, activated complement, and caused severe hemolytic

anemia. The infant was Xg(a+) and demonstrated a strongly

positive direct antiglobulin test at birth, and anti-Xga was

eluted from the infant’s cells. The clinical course was

remarkable only for a mild rise in bilirubin after birth.25

Because the only two examples of anti-CD99 were

found in healthy blood donors, information on the clinical

signicance of this antibody is not known.

Page 35: m4440090_27_2_11 Immunohematology

7/23/2019 m4440090_27_2_11 Immunohematology

http://slidepdf.com/reader/full/m444009027211-immunohematology 35/48IMMUNOHEMATOLOGY, Volume 27, Number 2, 2011 71

 XG blood group system: a review

 XG   is linked to genes responsible for several genetic

disorders.3,4  Ichthyosis is a disorder in which the skin

resembles scales on a sh. Ocular albinism is unique in that

only the eyes lack pigment. Retinoschisis is a sex-linked

form of macular degeneration affecting only men. High

levels of CD99 have been reported in Ewing sarcoma, some

neuroectodermal tumors, lymphoblastic lymphoma, and

acute lymphoblastic leukemia.8

Summary and Future Perspectives

The XG blood group system is most important for the

insight it has provided into the study and understanding of

meiotic errors that lead to sex chromosome aneuploidies. As

the roles of Xga and CD99 become more clearly delineated,

knowledge of the forgotten blood group system may aid in

the development of cures for certain genetic disorders and

types of cancers.

References

  1. Mann JD, Cahan A, Gelb AG, et al. A sex-linked blood group.Lancet 1962;1:8–10.

  2. Uchikawa M, Tsuneyama H, Tadokoro K, Juji T, YamadaM, Maeda Y. An alloantibody to 12E7 antigen detected in 2healthy donors [abstract]. Transfusion 1995;35:23S.

  3. Race RR, Sanger R. The Xg blood groups. In: Blood groupsin man. 6th ed. Oxford, PA: Blackwell Scientic Publication,1975:578–635.

  4. Reid ME, Lomas-Francis C. The blood group antigenfactsbook. 2nd ed. San Diego, CA: Academic Press, 2004:338–43.

  5. Ellis N, Tippett P, Petty A, et al. PBDX  is the XG  blood groupgene. Nat Genet 1994;8:285–90.

  6. Issitt PD, Anstee DJ. Applied blood group serology. 4th ed.Durham, NC: Montgomery Scientic Publications, 1998.

  7. Daniels G. Xg blood group system. In: Human blood groups.2nd ed. Malden, MA: Blackwell Science, 2002:374–86.

  8. Tippett P, Ellis NA. The Xg blood group system: a review.Transfus Med Rev 1998;12:233–57.

  9. Lyon MF. Gene action in the X-chromosome of the mouse( Mus musculus L.). Nature 1961;190:372–3.

  10. Lawler SD, Sanger R. Xg blood-groups and clonal-origintheory of chronic myeloid leukaemia. Lancet 1970;1:584–5.

  11. Goodfellow P, Pym B, Mohandas T, Shapiro LJ. The cellsurface antigen locus, MIC2X, escapes X-inactivation. Am J

Hum Genet 1984;36:777–82.  12. Dewey WJ, Mann JD. Xg blood group frequencies in some

further populations. J Med Genet 1967;4:12–15.

  13. Siniscalco M, Filippi G, Latte B, et al. Failure to deteclinkage between Xg and other  X -borne loci in Sardinians Ann Hum Genet 1966;29:231–52.

  14. Simmons RT. X-linked blood groups, Xg, in Australian Aborigines and New Guineans. Nature 1970;227:1363.

  15. Goodfellow P. Expression of the 12E7 antigen is controlledindependently by genes on the human X and Y chromosomes

Differentiation 1983;23(Suppl):S35–9.  16. Goodfellow PN, Tippett P. A human quantitative polymorphism related to Xg blood groups. Nature 1981;289404–5.

  17. Tippett P, Shaw M-A, Green CA, Daniels GL. The 12E7 redcell quantitative polymorphism: control by the Y-bornelocus, Yg. Ann Hum Genet 1986;50(Pt 4):339–47.

  18. Petty AC, Tippett P. Investigation of the biochemicarelationship of the blood group antigens Xga  and CD99(12E7 antigen) on red cells. Vox Sang 1995;69:231–5.

  19. Herron R, Smith GA. Identication and immunochemicacharacterization of the human erythrocyte membraneglycoproteins that carry the Xga antigen. Biochem J 1989262:369–71.

  20. Ellis NA, Ye T-Z, Patton S, German J, Goodfellow PN, Welle

P. Cloning of PBDX, and MIC2-related gene that spans thepseudoautosomal boundary on chromosome Xp. Nat Genet1994;6:394–400.

  21. Campana T, Szabo P, Piomelli S, Siniscalco M. The Xgantigen on red cells and broblasts. Cytogenet Cell Genet1978;22:524–6.

  22. Fellous M, Bengtsson B, Finnegan D, Bodmer WF. Expressionof the Xga antigen on cells in culture and its segregation insomatic cell hybrids. Ann Hum Genet 1974;37:421–30.

  23. Shaw MA, Tippet P, Delhanty JD, Andrews M, GoodfellowP. Expression of Xg and the 12E7 antigen in primates. JImmunogenet 1985;12:115–18.

  24. Devenish A, Burslem MF, Morris R, Contreras M. Serologiccharacteristics of a further example of anti-Xga  in North

London blood donors. Transfusion 1986;26:426–7.  25. Yokoyama M, McCoy JE Jr. Further studies on auto-anti-Xga antibody. Vox Sang 1967;13:15–17.

  26. Levy R, Dilley J, Fox RI, Warnke R. A human thymus-leukemia antigen dened by hybridoma monoclonaantibodies. Proc Natl Acad Sci U S A 1979;76:6552–6.

  27. Cook IA, Polley MJ, Mollison PL. A second example of anti-Xga. Lancet 1963;1:857–9.

  28. Sausais L, Krevans JR, Townes AS. Characteristics of a thirdexample of anti-Xga [abstract]. Transfusion 1964;4:312.

  29. Azar PM, Saji H, Yamanaka R, Hosoi T. Anti-Xga suspectedof causing a transfusion reaction. Transfusion 1982;22340–1.

 Nanette C. Johnson, MT(ASCP)SBB, Director, Immunohematol

ogy Reference Laboratory, Greater Chesapeake and Potomac

 Region, 4700 Mt. Hope Drive, Baltimore, MD 21215.

Page 36: m4440090_27_2_11 Immunohematology

7/23/2019 m4440090_27_2_11 Immunohematology

http://slidepdf.com/reader/full/m444009027211-immunohematology 36/4872  IMMU NOHEM ATOLOGY, Volum e 27, Numb er 2, 2011

Erratum V OL . 27, NO. 1, 2011, PP. 14, 16, AND 18

Red blood cell phenotype matching for various ethnic groups

The author has informed the editors of Immunohematology that there is an error on pages 14, 16, and 18. The running head

should appear as K.S.W. Badjie et al.

COMMUNICATION

Free Classified Ads and Announcements

Immunohematology  will publish classified ads and announcements (SBB schools, meetings, symposia, etc.) without charge. 

Deadlines for receipt of these items are as follows:

Deadlines

  1st week in January for the March issue 1st week in July for the September issue

  1st week in April for the June issue 1st week in October for the December issue

E-mail or fax information to [email protected] or phone (215) 451-2538

Page 37: m4440090_27_2_11 Immunohematology

7/23/2019 m4440090_27_2_11 Immunohematology

http://slidepdf.com/reader/full/m444009027211-immunohematology 37/48IMMUNOHEMATOLOGY, Volume 27, Number 2, 2011 73

Monoclonal antibodies available at no charge:

The New York Blood Center has developed a wide range of

monoclonal antibodies (both murine and humanized) that

are useful for donor screening and for typing RBCs with a

positive DAT. These include anti-A 1, -M, -s, -U, -D, -Rh17,

-K, -k, -Kpa, -Js b, -Fy a, -Fy3, -Fy6, -Wr b, -Xga, -CD99, -Do b,

-H, -Ge2, -Ge3, -CD55 (both SCR2/3 and SCR4), -Ok a, -I,

and anti-CD59. Most of the antibodies are murine IgG and

require the use of anti-mouse IgG for detection (Anti-K,

-k, and -Kpa). Some are directly agglutinating (Anti-A 1, -M,

-Wr b, and -Rh17) and a few have been humanized into the

IgM isoform (Anti-Js b). The antibodies are available at no

charge to anyone who requests them. Please visit our Web

site for a complete list of available monoclonal antibodies

and the procedure for obtaining them.

For additional information, contact:  Gregory Halverson,

New York Blood Center, 310 East 67th Street, New York,

NY 10021/e-mail: [email protected], phone:

(212) 570-3026, FAX: (212) 737-4935, or visit the Web

site at www.nybloodcenter.org >research >laboratories

>immunochemistry >current list of monoclonal antibodies

available.

Specialist in Blood Bank (SBB) Program

The Department of Transfusion Medicine, National

Institutes of Health, is accepting applications for its 1-year

Specialist in Blood Bank Technology Program. Students are

federal employees who work 32 hours/week. This program

introduces students to all areas of transfusion medicine

including reference serology, cell processing, HLA, and

infectious disease testing. Students also design and

conduct a research project. NIH is an Equal Opportunity

Organization. Application deadline is December 31, 2011

for the July 2012 class. See www.cc.nih.gov/dtm > education

for brochure and application. For further information

contact  Karen M. Byrne at (301) 451-8645 or KByrne@

mail.cc.nih.gov 

ANNOUNCEMENTS

Page 38: m4440090_27_2_11 Immunohematology

7/23/2019 m4440090_27_2_11 Immunohematology

http://slidepdf.com/reader/full/m444009027211-immunohematology 38/4874  IMMU NOHEM ATOLOGY, Volum e 27, Numb er 2, 2011

Announcements, cont.

Page 39: m4440090_27_2_11 Immunohematology

7/23/2019 m4440090_27_2_11 Immunohematology

http://slidepdf.com/reader/full/m444009027211-immunohematology 39/48IMMUNOHEMATOLOGY, Volume 27, Number 2, 2011 75

ADVERTISEMENTS

Masters (MSc) in Transfusion and Transplantation Sciences

at

The University of Bristol, England

 Applications are invited from medical or science graduates for the Master of Science (MSc) degree

in Transfusion and Transplantation Sciences at the University of Bristol. The course starts in

October 2011 and will last for 1 year. A part-time option lasting 2 or 3 years is also available. Theremay also be opportunities to continue studies for PhD or MD following the MSc. The syllabus is

organized jointly by The Bristol Institute for Transfusion Sciences and the University of Bristol,

Department of Pathology and Microbiology. It includes:

• Scientic principles of transfusion and transplantation

• Clinical applications of these principles

• Practical techniques in transfusion and transplantation

• Principles of study design and biostatistics

• An original research project

 Application can also be made for Diploma in Transfusion and Transplantation Science or a

Certicate in Transfusion and Transplantation Science.

The course is accredited by the Institute of Biomedical Sciences.

Further information can be obtained from the Web site:

http://ibgrl.blood.co.uk/MSc/MscHome.htm

For further details and application forms please contact:

Dr Patricia Denning-Kendall

University of Bristol

Paul O’Gorman Lifeline Centre

Department of Pathology and Microbiology 

Southmead Hospital

 Westbury-on-Trym, Bristol BS10 5NB, England

Fax +44 1179 595 342, Telephone +44 1779 595 455, e-mail: [email protected].

Page 40: m4440090_27_2_11 Immunohematology

7/23/2019 m4440090_27_2_11 Immunohematology

http://slidepdf.com/reader/full/m444009027211-immunohematology 40/4876  IMMU NOHEM ATOLOGY, Volum e 27, Numb er 2, 2011

Advertisements, cont.

Diagnostic testing for:• Neonatal alloimmune thrombocytopenia (NAIT)• Posttransfusion purpura (PTP)• Refractoriness to platelet transfusion• Heparin-induced thrombocytopenia (HIT)

• Alloimmune idiopathic thrombocytopenia purpura (AITP)Medical consultation available

Test methods:• GTI systems tests

— detection of glycoprotein-specic platelet antibodies  — detection of heparin-induced antibodies (PF4 ELISA)• Platelet suspension immunouorescence test (PSIFT)• Solid phase red cell adherence (SPRCA) assay• Monoclonal immobilization of platelet antigens (MAIPA)• Molecular analysis for HPA-1a/1b

FOR FURTHER INFORMATION, CONTACT:

Platelet Serology Laboratory (215) 451-4205

Janet Demcoe (215) 451-4914 [email protected]

Sandra Nance (215) [email protected]

American Red Cross Biomedical Services

Musser Blood Center

700 Spring Garden Street

Philadelphia, PA 19123-3594

National Platelet Serology Reference Laboratory

CLIA licensed

Our laboratory specializes in granulocyte antibody detection

and granulocyte antigen typing.

Indications for granulocyte serology testing

include:

• Alloimmune neonatal neutropenia (ANN)• Autoimmune neutropenia (AIN)• Transfusion-related acute lung injury (TRALI)

Methodologies employed:

• Granulocyte agglutination (GA)• Granulocyte immunouorescence by ow cytometry (GIF)• Monoclonal antibody immobilization of neutrophil antigens

(MAINA)

TRALI investigations also include:

• HLA (PRA) Class I and Class II antibody detection

FOR FURTHER INFORMATION, CONTACT:

Neutrophil Serology Laboratory (651) 291-6797Randy Schuller (651) 291-6758 

[email protected]

American Red Cross Biomedical Services

Neutrophil Serology Laboratory

100 South Robert Street

St. Paul, MN 55107

National Neutrophil Serology Reference Laboratory

CLIA licensed

Page 41: m4440090_27_2_11 Immunohematology

7/23/2019 m4440090_27_2_11 Immunohematology

http://slidepdf.com/reader/full/m444009027211-immunohematology 41/48IMMUNOHEMATOLOGY, Volume 27, Number 2, 2011 77

Advertisements, cont

 Antibody identication and problem resolution

HLA-A, B, C, and DR typing

HLA-disease association typing

Paternity testing/DNA

FOR INFORMATION, CONTACT:

Mehdizadeh Kashi

at (503) 280-0210

or write to:

Tissue Typing Laboratory

American Red Cross Biomedical Services

Pacifc Northwest Region

3131 North Vancouver

Portland, OR 97227

Reference and Consultation Services

CLIA licensed, ASHI accredited

IgA and anti-IgA testing are available to do the

following:

• Identify IgA-decient patients

• Investigate anaphylactic reactions

• Conrm IgA-decient donors

Our ELISA for IgA detects protein to 0.05 mg/dL.

FOR ADDITIONAL INFORMATION CONTACT:

Janet Demcoe at (215) 451-4914,

or e-mail:

[email protected],

or write to:

American Red Cross Biomedical Services

Musser Blood Center

700 Spring Garden Street

Philadelphia, PA 19123-3594

ATTN: Janet Demcoe

IgA/Anti-IgA Testing

CLIA licensed

• Effective tool for screening large volumes of donors

• Gel diffusion test that has a 15-year proven track record:

Approximately 90 percent of all donors identied as

IgA decient by this method are conrmed by the more

sensitive testing methods

FOR ADDITIONAL INFORMATION :

Kathy Kaherl

at (860) 678-2764

e-mail:

[email protected]

or write to:

Reference Laboratory

American Red Cross Biomedical Services

Connecticut Region

209 Farmington Ave.

Farmington, CT 06032

Donor IgA Screening

CLIA licensed

Immunohematology Reference Laboratory

AABB, ARC, New York State, and CLIA licensed24-hour phone number:

(215) 451-4901Fax:

(215) 451-2538

 American Rare Donor Program

24-hour phone number:(215) 451-4900

Fax:(215) 451-2538

[email protected]

Immunohematology 

Phone, business hours:(215) 451-4902

Fax:(215) 451-2538

[email protected]

Quality Control of Cryoprecipitated–AHF

Phone, business hours:(215) 451-4903

Fax:(215) 451-2538

National Reference Laboratory

for Blood Group Serology

Page 42: m4440090_27_2_11 Immunohematology

7/23/2019 m4440090_27_2_11 Immunohematology

http://slidepdf.com/reader/full/m444009027211-immunohematology 42/4878  IMMU NOHEM ATOLOGY, Volum e 27, Numb er 2, 2011

Advertisements, cont.

What is a certied Specialist in Blood Banking (SBB)?

• Someone with educational and work experience qualications that successfully passes the American Society for Clinical Pathology (ASCP)

board of certication (BOC) examination for the Specialist in Blood Banking.

• This person will have advanced knowledge, skills, and abilities in the eld of transfusion medicine and blood banking.

Individuals who have an SBB certication serve in many areas of transfusion medicine:

• Serve as regulatory, technical, procedural, and research advisors

• Perform and direct administrative functions

• Develop, validate, implement, and perform laboratory procedures

• Analyze quality issues preparing and implementing corrective actions to prevent and document nonconformances

• Design and present educational programs

• Provide technical and scientic training in transfusion medicine

• Conduct research in transfusion medicine

Who are SBBs?

Supervisors of Transfusion Services Executives and Managers of Blood Centers LIS Coordinators Educators

Supervisors of Reference Laboratories Research Scientists Consumer Safety Ofcers

Quality Assurance Ofcers Technical Representatives Reference Lab Specialists

Why become an SBB?

Professional growth Job placement Job satisfaction Career advancement

How does one become an SBB?

CAAHEP-accredited SBB Technology program or grandfather the exam based on ASCP education and experience criteria.

Fact:  In recent years, a greater percentage of individuals who graduate from CAAHEP-accredited programs pass the SBB exam compared

to individuals who grandfather the exam. The BEST route for obtaining an SBB certication is to attend a CAAHEP-accredited Specialist in

Blood Bank Technology Program.

Which approach are you more compatible with?

Contact the following programs for more information:

Additional information can be found by visiting the following Web sites: www.ascp.org, www.caahep.org, and www.aabb.org

Program Contact Name Phone Contact Email Contact WebsiteOnsite orOnline Program

Walter Reed Army Medical Center William Turcan 202-782-6210 [email protected] www.militaryblood.dod.mil/fellow Onsite

American Red Cross, Southern California Region Michael Coover 909-859-7496 [email protected] none Onsite

ARC-Central OH Region Joanne Kosanke 614-253-2740 x 2270 [email protected] none Onsite

Blood Center of Wisconsin Lynne LeMense 414-937-6403 [email protected] www.bcw.edu Onsite

Community Blood Center/CTS Dayton, Ohio Nancy Lang 937-461-3293 [email protected] www.cbccts.org/education/sbb.html Online

Gulf Coast School of Blood Bank Technology Clare Wong 713-791-6201 [email protected] http://giveblood.org/index.php?page =sbb-distance-program Online

Hoxworth Blood Center/Universit y of Cincinnati Susan Wilkinson 513-558-1275 Pamela.inglish@ [email protected]

www.hoxworth.org Onsite

Indiana Blood Center Jayanna Slayten 317-916-5186 [email protected] www.indianablood.org Online

Johns Hopkins Hospital Lorraine Blagg 410-502-9584 [email protected] http://pathology.jhu.edu/department/divisions/tranfusion/sbb2.cfm

Onsite

Medical Center of Louisiana Karen Kirkley 504-903-3954 [email protected] none Onsite

NIH Clinical Center Department of Transfusion Medicine Karen Byrne 301-496-8335 [email protected] www.cc.nih.gov/dtm/research/sbb.html Onsite

Rush University Veronica Lewis 312-942-2402 [email protected] www.rushu.rush.edu/catalog/acadprograms/chs/cls/clssbbcert.html

Online

Transfusion Medicine Academic Center at Florida BloodServices

Marjorie Doty 727-568-5433 x 1514 [email protected] www.fbsblood.org Online

University Health System and Affiliates, San Antonio Linda Myers 210-731-5526 [email protected] www.sbbofsa.org Onsite

University of Texas Medical Branch at Galveston Janet Vincent 409-772-3055 [email protected] www.utmb.edu/sbb Online

University of Texas SW Medical Center LeAnne Hutson 214-648-1780 [email protected] http://utsouthwestern.edu/mls Online

Becoming a Specialist in Blood Banking (SBB)

Revised August 2009

Page 43: m4440090_27_2_11 Immunohematology

7/23/2019 m4440090_27_2_11 Immunohematology

http://slidepdf.com/reader/full/m444009027211-immunohematology 43/48IMMUNOHEMATOLOGY, Volume 27, Number 2, 2011 79

I. GENERAL INSTRUCTIONSBefore submitting a manuscript, consult current issues of

Immunohematology  for st yle. Double-space throughout the manuscript.

Number the pages consecutively in the upper right-hand corner, beginning

with the title page.

II. SCIENTIFIC ARTICLE, REVIEW, OR CASE REPORT WITH

LITERATURE REVIEW 

 A. Each component of the manuscript must star t on a new page in the

following order:

1. Title page

2. Abstract 

3. Text 

4. Acknowledgments

5. References

6. Author information

7. Tables

8. Figures

B. Preparation of manuscript

1. Title page

a. Full title of manuscript with only first letter of first word

capitalized (bold title)

b. Initials and last name of each author (no degrees; all CAPS), e.g.,

M.T. JONES, J.H. BROWN, AND S.R. SMITH

c. Running title of ≤40 characters, including spaces

d. Three to ten key words

2. Abstract 

a. One paragraph, no longer than 300 words

b. Purpose, methods, findings, and conclusion of study

3. Key words

a. List under abstract 

4. Text (serial pages): Most manuscripts can usually, but not

necessarily, be divided into sections (as described below). Survey

results and review papers may need individualized sections

a. Introduction — Purpose and rationale for study, including

pertinent background references

b. Case Report (if indicated by study) — Clinical and/or hematologic

data and background serology/molecular 

c. Materials and Methods — Selection and number of subjects,

samples, items, etc. studied and description of appropriate

controls, procedures, methods, equipment, reagents, etc.

Equipment and reagents should be identified in parentheses by

model or lot and manufacturer’s name, city, and state. Do not

use patient’s names or hospital numbers.

d. Results — Presentation of concise and sequential results,

referring to pertinent tables and/or figures, if applicable

e. Discussion — Implication and limitations of the study, links to

other studies; if appropriate, link conclusions to purpose of study

as stated in introduction

5. Acknowledgments: Acknowledge those who have made substantialcontributions to the study, including secretarial assistance; list any

grants.

6. References

a. In text, use superscript, Arabic numbers.

b. Number references consecutively in the order they occur in the

text.

7. Tables

a. Head each with a brief title; capitalize the first letter of first word

(e.g., Table 1. Results of . . .) use no punctuation at the end of

the title.

b. Use short headings for each column needed and capitalize first

letter of first word. Omit vertical lines.

c. Place explanation in footnotes (sequence: *, †, ‡, §, ¶, **, ††).

8. Figures

a. Figures can be submitted either by e-mail or as photographs

(5ʺ×7ʺ glossy).

b. Place caption for a figure on a separate page (e.g. Fig. 1 Results

of...), ending with a period. If figure is submitted as a glossy,

place first author’s name and figure number on back of each

glossy submitted.

c. When plotting points on a figure, use the following symbols if

possible:llssnn.

9. Author information

a. List first name, middle initial, last name, highest degree,

position held, institution and department, and complete address

(including ZIP code) for all authors. List country when applicable.

III. EDUCATIONAL FORUM

 A. All submitted manuscripts should be approx imately 2000 to 2500 words

 with per tinent references. Submissions may include:1. An immunohematologic case that illustrates a sound investigative

approach with clinical correlation, reflec ting appropriate

collaboration to sharpen problem solving skills

2. Annotated conference proceedings

B. Preparation of manuscript

1. Title page

a. Capitalize first word of title.

b. Initials and last name of each author (no degrees; all CAPs)

2. Text 

a. Case should be written as progressive disclosure and may

include the following headings, as appropriate

i. Clinical Case Presentation: Clinical informat ion and

differential diagnosis

ii. Immunohematologic Evaluation and Results: Serology and

molecular testing 

iii. Interpretation: Include interpretation of laboratory results,

correlating with clinical findings

iv. Recommended Therapy: Include both transfusion and

nontransfusion-based therapies

v. Discussion: Brief review of literature with unique features of

this case

vi. Reference: Limited to those directly pertinent 

vii. Author information (see II.B.9.)

viii. Tables (see II.B.7.)

IV. LETTER TO THE EDITOR

 A. Preparation

1. Heading (To the Editor)

2. Title (first word capitalized)

3. Text (written in letter [paragraph] format)

4. Author(s) (type flush right; for first author: name, degree, institution,

address [including city, state, Zip code and country]; for other

authors: name, degree, institution, city and state)

5. References (limited to ten)

6. Table or figure (limited to one)

Send all manuscripts by e-mail to [email protected] 

Immunohematology Journal of Blood Group Serology and Education

Instructions for Authors

Page 44: m4440090_27_2_11 Immunohematology

7/23/2019 m4440090_27_2_11 Immunohematology

http://slidepdf.com/reader/full/m444009027211-immunohematology 44/48

Page 45: m4440090_27_2_11 Immunohematology

7/23/2019 m4440090_27_2_11 Immunohematology

http://slidepdf.com/reader/full/m444009027211-immunohematology 45/48

Page 46: m4440090_27_2_11 Immunohematology

7/23/2019 m4440090_27_2_11 Immunohematology

http://slidepdf.com/reader/full/m444009027211-immunohematology 46/48

Page 47: m4440090_27_2_11 Immunohematology

7/23/2019 m4440090_27_2_11 Immunohematology

http://slidepdf.com/reader/full/m444009027211-immunohematology 47/48

Ordering Information

The book, which costs $25, can beordered in two ways:

Order online from the publisher•

at: www.sbbpocketbook.com

Order from the authors, who•

 will sign the book. Send a check,made payable to “New York BloodCenter” and indicate “Pocket-book” on the memo line, to:

Marion ReidLaboratory of ImmunochemistryNew York Blood Center310 East 67th StreetNew York, NY 10065

Please include the recipient’scomplete mailing address.

Blood Group

 Antigens & Antibodies A guide to clinical relevance& technical tips

by Marion E. Reid & Christine Lomas-Francis

Pocketbook Education FundThe authors are using royalties generated from the sale of

 this pocketbook for educational purposes to mentor peoplein the joys of immunohematology as a career. They willaccomplish this in the following ways:

Sponsor workshops, seminars, and lectures•

Sponsor students to attend a meeting•

Provide copies of the pocketbook •

(See www.sbbpocketbook.com for details to apply for funds)

This compact “pocketbook” from the authors of the Blood

Group Antigen FactsBook is a must for anyone who is involvedin the laboratory or bedside care of patients with blood group alloantibodies.

The book contains clinical and technical information about the nearly 300 ISBT recognized blood group antigens and their corresponding antibodies. The information is listed inalphabetical order for ease of finding—even in the middle of the night. Included in the book is information relating to:

Clinical significance of antibodies in transfusion and HDN.•

Number of compatible donors that would be expected•

 to be found in testing 100 donors. Variations in differentethnic groups are given.

Characteristics of the antibodies and optimal technique(s)•

for their detection.

Technical tips to aid their identification.•

 Whether the antibody has been found as an autoantibody.•

Page 48: m4440090_27_2_11 Immunohematology

7/23/2019 m4440090_27_2_11 Immunohematology

http://slidepdf.com/reader/full/m444009027211-immunohematology 48/48

FIRST CLASS

U.S. POSTAGE

PAIDSOUTHERN MD

PERMIT NO. 4144Musser Blood Center

700 Spring Garden Street

Philadelphia, PA 19123-3594

(Place Label Here)