You are on page 1of 11

REVIEW

Advances in alloimmune thrombocytopenia: perspectives on current


concepts of human platelet antigens, antibody detection strategies,
and genotyping

Tomoya Hayashi, Fumiya Hirayama

Japanese Red Cross Kinki Block Blood Centre, Osaka, Japan

Abstract HPA antibodies were detected after a peripheral blood


Alloimmunisation to platelets leads to the production stem cell transplant between sisters with identical
of antibodies against platelet antigens and consequently HLA-A, -B, -DR, -DQ, -DP and ABO phenotypes
to thrombocytopenia. Numerous molecules located on which was followed by persistent, severe isolated
the platelet surface are antigenic and induce immune- thrombocytopenia resistant to platelet transfusions15.
mediated platelet destruction with symptoms that The detection of these antibodies is required to diagnose,
can be serious. Human platelet antigens (HPA) cause treat, and prevent these disorders. It is also important

l
thrombocytopenias, such as neonatal alloimmune to distinguish between thrombocytopenia induced by

Sr
thrombocytopenia, post-transfusion purpura, and platelet alloantibodies from drug-induced disease16.
transfusion refractoriness. Thirty-four HPA are classified The platelet surface membrane contains antigenic
into 28 systems. Assays to identify HPA and anti-HPA molecules other than HPA, including ABO blood type

i
antibodies are critically important for preventing antigens17, human leucocyte antigens (HLA)18, and the
iz
and treating thrombocytopenia caused by anti-HPA Naka antigen localised on CD3619. HLA alloantibodies
antibodies. Significant progress in furthering our are the most important cause of PTR18; therefore, careful
v
understanding of HPA has been made in the last decade: screening for HLA antibodies is required even when
er
new HPA have been discovered, antibody-detection other antibodies are identified. Because CD36 deficiency
methods have improved, and new genotyping methods is rare in Caucasians but frequent in Asians and Africans,
S

have been developed. We review these advances and Nak a antibodies are important for diagnosing and
discuss issues that remain to be resolved as well as treating thrombocytopenia in the latter populations.
TI

future prospects for preventing and treating immune Early diagnosis is essential to prevent severe disease in
thrombocytopenia. a timely and effective manner20.
New HPA were discovered in the last decade, and they
M

Introduction have been implicated in immune thrombocytopenia11;


Immune and non-immune mechanisms can strategies for detecting antibodies and genotyping are
SI

decrease the number of platelets, leading to bleeding improving. The primary purpose of this review is to
manifestations that range from petechiae and simple describe the molecular properties of HPA and related
bruising to intracranial haemorrhage and death1. Platelets molecules (other than HLA and ABO) and the antibody-
©

interact with coagulation factors to arrest haemorrhage detection systems that have been developed to enhance
and support vascular integrity2,3. At sites of vascular the sensitivity and specificity of HPA detection. We
injury, platelets change shape, adhere to the site, and consider genotyping methods as well, which can be
secrete cytokines that are essential for tissue repair1,4,5. performed quickly using multiple analytical techniques.
Platelets are indispensable for maintaining vascular We conclude by discussing important issues that must
integrity. Molecules expressed by platelets that are be resolved and offer our perspectives for the future
involved in arresting haemorrhage include integrin regarding the prevention and treatment of immune
αIIbβ3, GPIb/IX, and integrin α2β16-10. thrombocytopenia.
The antigens that are recognised by alloantibodies
have been categorised by the Platelet Nomenclature Polymorphisms and functions of human platelet
Committee, and 34 human platelet antigens (HPA) antigens and CD36
have been defined11,12. Antibodies against HPA are According to the Immuno Polymorphism Database
regarded as the principal cause of various reactions (IPD - www.ebi.ac.uk/ipd/hpa/), the number of HPA has
elicited by platelet transfusion, such as platelet reached 28 systems11. The variety of HPA is generated
transfusion refractoriness (PTR) and post-transfusion by the substitution of a single amino acid residue and
purpura (PTP)1,13. Antibodies against HPA cause foetal/ by deletion of one amino acid residue from platelet
neonatal alloimmune thrombocytopenia (FNAIT)14. glycoproteins (Figure 1)11,12,21-23. Six of the systems

Blood Transfus 2015; 13; 380-90 DOI 10.2450/2015.0275-14


© SIMTI Servizi Srl
380

All rights reserved - For personal use only


No other uses without permission
Current concepts of human platelet antigens

l
Figure 1 - Human platelet antigens (HPA) polymorphisms.

Sr
This figure is constructed using data from the IPD web site (http://www.ebi.ac.uk/ipd/hpa/table2.html) and United States National
Center for Biotechnology Information (NCBI). HPA are located on the following six molecules, GPIIIa (integrin β3), GPIIb (integrin
α2b), GPIa (integrin α2), CD109, GPIbα, and GPIbβ, and are encoded by ITGB3, ITGA2B, ITGA2, CD109, GP1BA, and GP1BB,
respectively. The gene symbols are those approved by the Human Genome Organisation (HUGO) Gene Nomenclature Committee

i
(http://www.genenames.org). Sequences encoding HPA within the same exons are indicated by the same colours. Nucleotide variations:
iz
nucleotide numbers are taken from the reference sequence in the NCBI database, which may differ from those given in the original
publication describing the mutation. Nucleotide and protein variations are shown as changes from the more common (a) to the less
common (b) forms. The number of amino acid variations is derived from the precursor protein, and the numbers in parenthesis indicate
v
amino acid residue number in the mature protein. The definitions of symbols (#, s, §, , ‡, *, ¶ and †) are described under each residue.
er

are biallelic (HPA-1, -2, -3, -4, -5, and -15), and are present in patients with Glanzmann's thrombasthenia,
S

others include 34 platelet-specific alloantigens defined which is an autosomal recessive disorder caused by
according to the rules of the ISBT Platelet Working mutation of the genes encoding GPIIb or GPIIIa or is
Party. The allelic frequencies of HPA differ among them, acquired as an autoimmune disorder that leads to an
TI

and racial differences are numerous. For the six biallelic increased tendency to bleed26.
systems, the most frequent HPA are defined as "a" alleles Most HPA are localised to GPIIb and GPIIIa
M

or "wild-type." All alloantigen systems comprise genes (Figure 1). GPIIIa and GPIIb comprise 16 and seven
encoding only six proteins. Among them, GPIIb [integrin HPA systems, respectively11,12. There are several genetic
SI

alpha subunit IIb (IIb), CD41] and GPIIIa [integrin beta hot-spots in which the mutations that encode two to four
subunit 3 (β3), CD61] form the GPIIb/GPIIIa (GPIIbIIIa, HPA are present in the same exons (Figure 1).
integrin IIbβ3) complex and GPIb GPIb and GPIX Although all HPA present on GPIIbIIIa are associated
©

and GPV form the GPIb-IX-V complex. GPIIIa also with FNAIT, HPA-1a is strongly associated with FNAIT,
forms a complex with αV and is expressed by platelets particularly in Caucasians , and antibodies against HPA-
and other cells including osteoclasts and endothelium 4b and HPA-21b are present in Asians27-29. Antibodies
cells24. GPIa (α2) and GPIIa (β) form the GPIaGPIIa against HPA-1a, HPA-1b, and HPA-3b cause PTR, and
(GPIaIIa, integrin α2β) complex. antibodies against HPA-1a, HPA-1b, HPA-3a, HPA-
3b, and HPA-4a cause PTP30-33. PTR can be caused by
GPIIb/GPIIIa (integrin αIIbβ3, the fibrinogen isoantibodies against GPIIbIIIa in patients with type 1
receptor) Glanzmann's thrombasthenia34.
GPIIb and GPIIIa form the GPIIbIIIa complex,
although GPIIIa forms a complex with αV as well24. GPIb
The GPIIbIIIa complex is the most abundant molecule GPIbα (CD42b) and GPIbβ (CD42c) bind covalently
on the surface of platelets. At the site of endothelial to form a complex and associate non-covalently with
damage, GPIIbIIIa is activated and plays a central role GPIX (CD42a). Dimers of these three molecules further
in the formation of an occlusive thrombus; it binds associate with the GPV (CD42d) molecule to form
to fibrinogen, von Willebrand factor, fibronectin, and GPIbIXV (CD42). CD42 (approximately 25,000 copies
vitronectin, which are required for haemostasis25,26. of GPIb/IX and 12,000 copies of GPV molecules per
Functional abnormalities and deficiency of GPIIbIIIa cell) is the second most abundant molecule on the platelet

Blood Transfus 2015; 13; 380-90 DOI 10.2450/2015.0275-14


381

All rights reserved - For personal use only


No other uses without permission
Hayashi T, Hirayama F

surface. The levels of the glycoprotein Ib-IX-V complex CD109 is expressed as a 205-kDa glycoprotein, which
are higher in neonatal cord blood than in adult blood. is cleaved by furin (furinase) in the Golgi apparatus into
The binding of GPIbIXV to von Willebrand 180-kDa and 25-kDa subunits46. CD109 is associated
factor mediates platelet adhesion under fast flow with the growth of carcinomas and keratinocytes
conditions and plays an important role in maintaining through the regulation of TGF- signaling46. CD109 may
haemostasis at sites of vascular injury. GPIbIXV binds play a role in the interactions of T cells with antigen-
to the complement receptor of macrophages Mac-1 presenting cells or in T- and B-cell interactions46. The
(macrophage-1 antigen or integrin αMβ2), which amounts of CD109 on the platelet surface differ by as
comprises CD11b (integrin αM) and CD18 (integrin much as a factor of >100 among individuals. However,
β2), and mediates the phagocytosis of platelets, the expression levels of CD109 are generally lower
indicating that GPIbIXV plays an important role in than those of HPA-associated glycoproteins47. Surface
platelet turnover35. Quantitative or qualitative defects of expression of CD109 on platelets is decreased by cooling
GPIbIXV expressed by platelets occur in patients with and freezing47. Although the precise function of platelet
Bernard-Soulier syndrome, which is characterised by no CD109 is unknown, its identification as a member
platelet agglutination in response to ristocetin, prolonged of thioester-containing proteins suggests that it may
bleeding and abnormal platelet size and turnover36. mediate covalent binding of cells to substrates as well as
The HPA-2 and HPA-12 systems are located on intercellular interactions46. Further studies are required
the alpha and beta chains of GPIb, respectively11,12. to define the function of platelet CD109 in detail.

l
Antibodies against HPA-2b and HPA-12bw are CD109 is a component of only HPA-1511,12. The

Sr
associated with FNAIT37,38, and the former are sometimes detection of antibodies against CD109 is hampered,
detected in the patients with PTR39. Combinations because platelets express low levels of CD109 on
of antibodies against HPA-1b and HPA-2a induced their surface and CD109 is unstable when platelets are

i
moderate thrombocytopenia in a patient with FNAIT40.
Isoantibodies against GPIb can cause PTR in patients
v iz
cooled or frozen. Panels of platelets must, therefore,
be carefully selected according to CD109 levels and
with Bernard-Soulier syndrome. stored under appropriate conditions. The development
of new methods for detecting anti-HPA antibodies may
er

GPIa accelerate our understanding of their importance48,49.


GPIa (CD49b) associates with GPIIa (CD29) to form This subject is considered in the section "Assays for
S

integrin α2β1 (GPIaIIa) complex, known as very late detecting HPA antibodies". Anti-HPA-15 antibodies are
antigen (VLA-2). GPIaIIa is expressed by monocytes, detected in patients receiving multiple transfusions and
TI

T cells, B cells, NK cells, and platelets41. Platelet mothers with FNAIT48,49. Although antibodies against
GPIaIIa is involved in adhesion at low flow rates by HPA-15a and HPA-15b are associated with FNAIT,
M

binding to collagen and participates in cell surface- they are usually detected together with anti-HLA
mediated signalling leading to GPIIbIIIa activation. antibodies49. The incidence of HPA-15 alloimmunisation
Approximately 800-2,800 GPIaIIa molecules are present is significantly higher in patients who undergo multiple
SI

on the platelet surface. transfusions than in mothers with FNAIT48.


Four HPA systems (HPA-5, HPA-13, HPA-18,
HPA-25) are located on GPIa 11,12. The HPA-13bw CD36
©

mutation is unusual in that it alters the function of GPIa, CD36 (also called GPIIIB, PAS IV, PAS-4, fatty acid
and platelets from HPA-13bw-positive individuals translocase, glycoprotein IIIb, platelet glycoprotein 4,
have a reduced response to collagen, as revealed by platelet glycoprotein IV, or Naka antigen) is expressed
aggregation studies, and a reduced ability to spread on on various cells, including platelets, and is not an
a collagen surface. Antibodies against HPA-5a, HPA-5b, HPA component. Because CD36 mediates immune
HPA-13bw, HPA-18bw, and HPA-25bw cause FNAIT, thrombocytopenia50, it is relevant to this review. CD36
and antibodies against HPA-5a are not limited to patients is one of four major glycoproteins on the platelet surface
of a particular race42-44. Antibodies against HPA-5a and and serves as a receptor in cells other than platelets51.
HPA-5b are present in patients with PTR, and the former CD36 binds diverse molecules, including collagen,
causes PTP as well. anionic phospholipids, oxidized low density lipoproteins,
and thrombospondin. It directly mediates cytoadherence
CD109 of Plasmodium falciparum to erythrocytes, binds long-
GPI-anchored alpha 2 macroglobulin-related protein chain fatty acids, and may regulate or directly mediate
(CD109, 150 kDa TGF--1-binding protein, C3 and PZP- the transport of fatty acids. CD36 is, therefore, attracting
like alpha-2-macroglobulin domain-containing protein considerable attention from researchers in the fields of
7) links to the platelet surface through a GPI anchor45. obesity and diabetes.

Blood Transfus 2015; 13; 380-90 DOI 10.2450/2015.0275-14


382

All rights reserved - For personal use only


No other uses without permission
Current concepts of human platelet antigens

The expression or lack of expression of CD36 a well-characterised panel of platelets is necessary for
among cell types, particularly by blood cells, is detecting antibodies against HPA. Unfortunately, it is
noteworthy (Table I)52. Briefly, in patients with type I difficult to prepare such a panel. Second, the serum
CD36 deficiency, CD36 is not expressed by platelets or antibodies and the monoclonal antibody may compete67.
monocytes, whereas in patients with type II deficiency This risk can be limited to some extent by using multiple
only platelets fail to express CD36. Type II deficiency mouse monoclonal antibodies reactive with different
comprises types 2a and 2b, and CD36 deficiency epitopes; however, developing monoclonal antibodies
restricted to platelets is designated type 2a. When CD36 is expensive. To overcome these problems, new assay
is absent from erythroblasts, the phenotype is classified systems were developed that substitute target platelets
as type 2b53. Multiple alternatively spliced transcripts with transfected cell lines or recombinant peptides
encoding CD36 are present in various tissues and may (Table III).
account for its complex pattern of expression.
Certain mutations in CD36 cause type I deficiency; Panel of transfected cell lines
therefore, the antibody against CD36 is defined as an Techniques using cells transfected with cDNA
isoantibody, not as an alloantibody. CD36 is expressed encoding specific HPA typically employ CHO and 293T
in almost 100% of white Europeans and is not detectably cells and serve as alternatives to unavailable platelet
expressed by 2% of sub-Saharan Africans and 10% of panels. Recently, we established various K562 cell lines
Asians54-56. Because anti-CD36 antibodies target diverse such as Hayashi's platelet-associated (HP) cells that

l
tissues, patients display a broad range of symptoms. express various HPA, do not express HLA, HNA, or

Sr
For example, antibodies that react with platelets may HPA, and show low non-specific reactivity with human
lead to immune thrombocytopenia, those that react sera. These test systems are highly specific and sensitive
with platelets and monocytes lead to transfusion-related for detecting antibodies against HPA68-72. Briefly, the cell

i
acute lung injury as well as life-threatening transfusion
reactions, and those that react with erythrocytes lead to
v iz
lines express one of the molecules as follows: CD36,
wild-type GPIIb/GPIIIa (HPA-1a) as well as HPA-1b,
hydrops foetalis57. -3b, -4b, -5b, -6b, -7b, -7 variant, -13b, -15a, -15b,
-18b, or HPA-21b. We recently established cell lines
er

Assays for detecting antibodies against human expressing wild-type GPIb and GPIb (HPA-2a and
platelet antigens HPA-12a) and HPA-2b (unpublished data).
S

Several methods that use platelets as target cells These cell lines overcome the difficulty of preparing
are available for the detection of antibodies against a platelet panel. They also detect anti-HPA antibodies in
TI

HPA, such as the monoclonal antibody-specific the presence of anti-HLA antibodies. More recently, we
immobilisation of platelet antigens (MAIPA) assay58, the developed transfected cell lines that employ an antigen-
M

platelet-antigen capture (PAC) assay59, the mixed passive capture assay system for detecting antibodies against
haemagglutination test (MPHA)60, flow cytometric CD36 without using monoclonal antibodies against
analysis61, a modified antigen-capture enzyme-linked CD3673. Since this cell line-dependent system does not
SI

immunosorbent assay (ELISA) 62 and a Luminex need mouse antibodies, it can avoid binding competition
bead assay63,64. The properties of these methods are between human and mouse antibodies. Moreover, the
summarised in Table II. receiver operating characteristic curve is superior to
©

Anti-HLA antibodies create problems for "whole that of the MAIPA system, and the transfectants allow
platelet" antibody detection methods such as MPHA monoclonal antibodies against CD36 to be omitted.
and flow cytometric analysis but not for glycoprotein- We provided HP-15a and HP-15b cells to the ISBT
specific assays such as MAIPA. Because the MAIPA Platelet Immunology Working Party. They recently
assay is highly sensitive and specific, it is considered reported the usefulness of paraformaldehyde-fixed
the gold standard65,66. These tests, including the MAIPA, HP-15 cells at the meeting of the ISBT held in June
do have some disadvantages. First, the preparation of 2014, Seoul, Korea. In addition, we have provided some

Table I - Types and frequencies of CD36 deficiency.

Deficiency Expression Frequency of deficiency


type
Platelets Monocytes RBC progenitors Endothelial cells Caucasian Sub-Sahara Asian
African

I absent absent absent absent <0.3% 2.50% 0.5-1%

II absent # 3-10%

#depending on the presence or absence on RBC progenitors; the deficiency is classified into type II-a and type II-b, respectively.

Blood Transfus 2015; 13; 380-90 DOI 10.2450/2015.0275-14


383

All rights reserved - For personal use only


No other uses without permission
Hayashi T, Hirayama F

Table II - Summary of the properties of antibody detection assays.

Whole platelet methods

Method Principle Advantage Disadvantage Additional remarks

MPHA The test serum is added onto a microplate coated with platelet Simple procedure Possible insufficient High-titre HLA
SPRCA extracts and indicator sheep red blood cells coated with anti- Low cost sensitivity and antibodies hamper
human IgG are added. The presence of the human antibody is Detector not required specificity the detection of HPA
judged by agglutination patterns. antibodies.
PSIFT The patient's serum is added to a platelet suspension and an Simple procedure Possible insufficient HLA antibodies
anti-human IgG conjugated to a fluorescent dye is added. NonHPA platelet sensitivity and hamper the detection
Fluorescence emission is analysed using a flow cytometer or antibodies are specificity of HPA antibodies.
fluorescence plate reader. detectable as well instrumentation
(FACS) is expensive

Glycoprotein specific methods

Method Principle Advantage Disadvantage Additional remarks

MAIPA Immune complexes composed of the target HPA, human-specific High sensitivity and High cost Need to be careful
PAC antibody, and mouse monoclonal antibody are captured by anti- high specificity Complicated procedure about the antibody
mouse IgG coated on a plate. The human antibody is detected competition
using enzyme-linked anti-human IgG and a colorimetric detector.

l
Sr
MACE Monoclonal antibodies coated onto a solid phase are used to capture High specificity High cost Need to be careful
the target HPA. The human antibody is detected using enzyme- Complicated procedure about the antibody
linked anti-human IgG and a colorimetric detector. competition
ICFA Microarray beads are separately coupled with recombinant GP High specificity High cost Need to be careful

i
fragments or monoclonal antibodies specific for HPA and HLA. Simultaneous analysis
iz Expensive about the antibody
Luminex xMAP technology adapts MAIPA onto the Luminex of HLA and HPA instrumentation competition when
platform by using a secondary antibody conjugated to a fluorescent antibodies in one well (FACS or Luminex) using antibody-
v
dye. coupled microspheres.
SPR Measures binding of antibody onto an antigen-coated planar metal High sensitivity. Requires highly Since the sensitivity is
er
surface. Bound molecules change the local index of refraction, Detects low-avidity purified antigens and so high that the clinical
which is generated by the antigen-antibody interaction that changes antibodies. Washing immunoglobulins. significance of the
S

the resonance of the surface plasmon waves. and labelling are not Requires specialised detected antibodies are
required equipment. addressed by in-vivo
experiments, such as
TI

those using the NOD/


SCID mouse.
HP-IPA Immune complexes composed of the target HPA, human specific High sensitivity High cost
M

antibody, and enzyme-linked mouse-anti-human IgG are captured Eliminates antibody Complicated procedure
by anti-mouse IgG coated on a plate, and the human antibody is competition Requires a cell panel
detected after adding the substrate.
SI

MPHA: mixed passive haemagglutination61; SPRCA: solid phase red cell adherence61; PSIFT: platelet suspension immuno-fluorescence test58; MAIPA:
monoclonal antibody-specific immobilisation of platelet antigens; PAC: platelet antigen capture immunoassays60; MACE: modified antigen capture ELISA62;
ICFA: immuno-complex capture fluorescence analysis63,64; SPR: surface plasmon resonance75,76; HP-IPA: HP cell-based monoclonal antibody-independent
Immobilisation of Platelet Antigen73.
©

Table III - Platelet-independent methods for detecting anti-HPA antibodies.


Sources of antigens Glycoproteins Established molecules (peptides) Detected antibodies Applicable methods
(reported)
HP cell lines GPIIb and GPIIIa Wild type, HPA-1b, HPA-3b, HPA- HPA-1a, 3a*, 4b, 6b, MAIPA, MACE, IFT,
(integrin αIIb and β3) 4b, HPA-6b, HPA-7b, HPA-7variant, 7variant, 21b HP-IPA, ICFA (xMAP)
(CD41 and CD61) HPA-21b
GPIa HPA-5b, HPA-13b, HPA-18b HPA-5a, 5b MAIPA, MACE, IFT, ICFA
(integrin α2, CD49b) (xMAP)
GPIbα (CD42b) HPA-2a, HPA-2b HPA-2b MAIPA, MACE, IFT
CD109 HPA-15a, HPA-15b HPA-15a, 15b MAIPA, MACE, IFT, ICFA
(xMAP)
CD36 CD36 Naka MAIPA, MACE, IFT,
HP-IPA, ICFA (xMAP)
Recombinant peptide GPIIIa ∆β3 peptide (wild type), Super rare HPA-1a, 1b, 4a, 4b, 6bw, ICFA (xMAP), SPR
peptide, 1a, 1b, 4b, 8b 7bw, 8bw, 11bw*, 16bw
Aptamer non glycoprotein Specific for HPA-1a antibody HPA-1a MPHA, SPRCA, ELISA
*Some antibodies are not detected. MAIPA: monoclonal-specific immobilization of platelet antigens; MACE: modified antigen capture ELISA; IFT: immuno
fluorescence test; HP-IPA: HP cell-based monoclonal antibody-independent Immobilisation of Platelet Antigen; ICFA (xMAP): immuno-complex capture
fluorescence analysis; SPR: surface plasmon resonance; MPHA:mixed passive hemagglutination; SPRCA: solid-phase red cell adherence assay.

Blood Transfus 2015; 13; 380-90 DOI 10.2450/2015.0275-14


384

All rights reserved - For personal use only


No other uses without permission
Current concepts of human platelet antigens

HP cell lines, including HP-15a and HP-15b, to several HPA-3a antibodies were detected only using MPHA,
blood centres. but not with recombinant GPIIbIIIa expressed by K562
cells71. Furthermore, Leon et al. reported that certain
Recombinant peptide techniques and highly sensitive anti-HPA-3a antibodies were not detected using the
methods HPA-3a peptide81. Taken together, these findings indicate
Another approach that does not require platelet that the methods for detecting antibodies against HPA-3a
panels involves the use of HPA peptides. Stafford vary in sensitivity and that using only one method may
et al.22 published unique methods to detect integrin increase the risk of not detecting them.
β3-associated anti-HPA antibodies using recombinant Allen et al. indicated that detection of anti-HPA
integrin β3 peptides that contained seven rare HPA, antibodies is influenced by cation-chelating compounds
termed SuperRare peptides that are suitable for detecting such as EDTA, and suggested that the results reported
anti-HPA antibodies. However, because the antigens by different laboratories might depend on the buffer,
used in peptide techniques do not have a natural three- antibody concentrations, or both 82. We, therefore,
dimensional structure or carbohydrate chains, this suggest that it is important to consider the advantages
system still requires careful evaluation using larger and disadvantages of each method and to use them in
numbers of positive control samples in the future74. combination.
Although peptides are often used in surface plasmon
resonance assays for detecting antibodies, purified Causal relationship between anti-human platelet

l
glycoproteins have been used to detect anti-HPA antigen antibodies and thrombocytopenia.

Sr
antibodies. Peterson et al.75 and Blackhoul et al.76 At present, low affinity anti-HPA antibodies can
reported highly sensitive systems that detect low-avidity be detected using highly sensitive methods such as
anti-HPA-1a antibodies using GPIIbIIIa purified from surface plasmon resonance. However, detection of

i
platelets.
A peptide aptamer, which mimics the HPA-1a
v iz
anti-HPA antibodies does not necessarily indicate that
the antibodies cause thrombocytopenia. Clinically, it is
antigen, is recognised by HPA-1a alloantibodies with obviously important to determine whether the antibodies
very high sensitivity77. Because aptamers are usually cause thrombocytopenia. Some fascinating reports
er

designed according to structure or affinity, their specific describe that a very low titre of antibodies can decrease
antigenicity limits applying them to each HPA. Future platelet counts in animal models75. Indeed, there is
S

studies are, therefore, required to develop assays using evidence that antibodies may cause thrombocytopenia
aptamers. even if the antibody level is lower than the detection
TI

limit of conventional methods; however, only a few


Limitations of serological systems for detecting laboratories are capable of performing highly sensitive
M

antibodies against human platelet antigens assays.


Standard control sera Because none of the current tests is perfect,
Although positive control antibodies are important depending on the circumstances of each laboratory,
SI

for every method, the availability of antisera is limiting. the tests should be performed together according to
Methods are being developed using a patient's B cells to their characteristics summarized in Table II. Moreover,
continuously produce specific anti-HPA antibodies. In genotype information would be useful for generating
©

the meantime, collaboration among laboratories expert reliable data, as described in the next section.
in platelet immunology is required to share the limited
amount of control antisera. Importance of genotyping for patients, their
parents, and the platelet panel
Anti-HPA-3a antibodies Genotyping patients, their parents, or both
Certain detection systems fail to detect anti-HPA-3a support antibody testing to diagnose alloimmune
antibodies. Harrison et al. reported that some anti-HPA- thrombocytopenia. Although HPA phenotyping is
3a antibodies are detected only in whole platelets but not important, reference sera are precious and limited in
when the platelet lysate is used78. Kataoka et al. reported number, so that HPA phenotyping is sometimes difficult.
that one of the anti-HPA-3b antibodies was detected In contrast, genetic technologies provide powerful tools
only using fresh but not fixed platelets79. Socher et al. to identify HPA. Techniques using genomic DNA for
reported that some anti-HPA-3a antibodies were detected HPA genotyping are relatively easy to perform, because
only using fresh but not stored platelets, and other genomic DNA can be extracted from any cell type,
anti-HPA-3a antibodies were not detected by western including leucocytes, except from erythrocytes and
blotting analysis of recombinant GPIIb and GPIIIa platelets. Moreover, commercial materials and reagents
expressed by CHO cells80. We found that some anti- are available for the analysis of single nucleotide

Blood Transfus 2015; 13; 380-90 DOI 10.2450/2015.0275-14


385

All rights reserved - For personal use only


No other uses without permission
Hayashi T, Hirayama F

polymorphisms. Thus, technological innovation paves with A-T base pairs. HRM is, therefore, useful for
the way to platelet typing of the foetus in suspected cases distinguishing HPA-b alleles from wild-type ones.
of FNAIT and enables matched platelets to be found to Furthermore, because the HRM method simultaneously
prevent PTR. Although determining the specificities detects one to four polymorphisms, it is advantageous
of antibodies is important in either case, the requisites for determining the frequency of rare HPA86; however
vary slightly between them. In the former case, platelet an expensive detector is required. The fluoPCR-reverse
typing predicts disease and indicates the requirement sequence-specific oligonucleotide probe technique,
for medical attention, while in the latter case it is used using a fluorescent hybridisation probe, simultaneously
to provide matched platelet products for treatment. determines several polymorphisms90. Many multiplex
Further, allelic frequency is important to predict the PCR techniques are commercially available.
incidence of incompatibility between an infant and A limitation of genotyping techniques is that if the
mother as well as between patients and platelet donors. target sequence is located in a genetic hot-spot, the
Various genotyping methods work in these situations. results may be greatly affected and it might be difficult to
Genotyping technologies are summarized in Table IV. design an appropriate primer and probe91. Thus, none of
Numerous techniques available for determining the methods described above is perfect, and the method
single nucleotide polymorphisms can be applied of choice will depend on the purpose of the analysis
to HPA genotyping83,84. HPA genotypes are widely and the identity of the HPA. Use of two genotyping
determined using sequence-specific primer-polymerase techniques using different primer sets can limit this risk.

l
chain reaction (PCR-SSP), denaturing gradient gel

Sr
electrophoresis (PCR-DGGE)85, and restriction fragment Pitfalls of genotyping
length polymorphism (PCR-RFLP) techniques. Although genotyping is a powerful tool for definitive
However, these methods are generally specialised to diagnosis of FNAIT, genotype does not always correlate

i
determine one polymorphism in a single test and are
therefore applicable in the analysis of patients with
v iz
with phenotype, as in a propositus heterozygous for type
I Glanzmann's thrombathenia or Bernard-syndromes.
FNAIT. In contrast, a high-throughput method is A propositus can be genotypically heterozygous for
required for donor screening and frequency analysis. a particular HPA but phenotypically homozygous,
er

For example, the high-resolution melting (HRM) because one of the allele is not expressed on the platelet
method is cost-effective. Several groups reported that surface36,92.
S

HRM is useful for HPA genotyping of a large donor


pool, for determining genotype frequency, or both86-89. Future study
TI

HRM determines the melting point of DNA, which is Predicting the severity of thrombocytopenia is likely
influenced by the abundance of C-G base pairs. Most important for efficacious treatment of patients with
M

HPA polymorphisms are generated by replacing C-G FNAIT, autoimmune thrombocytopenia, or alloimmune

Table IV - Genotyping methods.


SI

Method Principle Advantage Disadvantage Additional


Remarks
PCR-SSP83 Sequence-specific primers amplify allele-specific DNA. Low cost Poor resolution
©

of amplicons
PCR-DGGE 84 The target sequence is amplified and separated using Low cost Lengthy procedure Ambiguous data
denaturing polyacrylamide gel electrophoresis. It
distinguishes mobility shifts of type a and b caused by a
change in the conformation of single-stranded amplicons.
PCR-RFLP83,85 Digestion using restriction enzymes High specificity and Lengthy procedure Occasionally leaves
low cost ambiguities
HRM86,88,89 The melting temperatures of DNA depend on their base Simple and quick. Expensive detector Occasionally
composition. PCR is performed using DNA intercalating Applicable for high- amplicons with
dyes, such as SYBR® green. As the sample is heated and the throughput analysis different sequences
two strands of the DNA separate (melt), the concentration melt at the same
of double-stranded DNA decreases, reducing fluorescence. temperature
The instrument generates a melting curve.
Real-time Uses allele-specific primers to amplify allele-specific DNA Simple and quick. Expensive detector
PCR-SSP85,87,88,89 and intercalating agents such as SYBR® green are used. Applicable for high-
throughput analysis
PCR-SSP: sequence specific primer; PCR-DGGE: denaturing gradient gel electrophoresis; PCR-RFLP: restriction fragment length polymorphism;
HRM: high-resolution melting method; SYBR® green: N',N'-dimethyl-N-[4-[(E)-(3-methyl-1,3-benzothiazol-2-ylidene)methyl]-1-phenylquinolin-1-ium-
2-yl]-N-propylpropane-1,3-diamine: SYBR Green is a cyanine dye and preferentially binds to double-stranded DNA.

Blood Transfus 2015; 13; 380-90 DOI 10.2450/2015.0275-14


386

All rights reserved - For personal use only


No other uses without permission
Current concepts of human platelet antigens

thrombocytopenia93. Alloantibodies destroy platelets inhibits the binding of maternal anti-HPA-1a antibodies
and eliminate them from the circulation through harvested from patients with FNAIT to a patient's
antibody-induced immune-mediated phagocytosis and platelets and abrogates monocyte responses to anti-HPA-
activation of complement59,93-100. The characteristics of 1a-coated platelets in vitro. Furthermore, the authors
the alloantibodies and the titre of IgG may, therefore, reported that in mice and humans, B2G1Δnab prevents
contribute to the severity of symptoms. the removal from the circulation of platelets that express
HPA-1a by destructive HPA-1a antibodies99,100.
Immunoglobulin subclasses
The ability to activate complement and the binding Other antigen systems
affinity for HPA antibodies of the Fc receptor on Although many high-sensitivity methods for
phagocytic cells differ depending on the subclasses of detecting antibodies are available, as described in
antibodies. Data are available on the immunoglobulin this review, there are patients with thrombocytopenia
subclasses of HPA antibodies59,93. Although evidence with no detectable anti-HPA antibodies. For example,
indicates that IgG2 is not as effective as IgG1 and approximately 50% of patients with FNAIT in
IgG3 for inducing phagocytosis and complement- Japan do not have detectable anti-HPA antibodies.
mediated cell lysis, Kelsch pointed out that IgG2 Alloantibody-dependent and alloantibody-independent
causes platelet dysfunction and leads to clinically thrombocytopenia may occur, including drug-induced
significant bleeding even when platelet counts are and autoantibody-induced thrombocytopenia. In patients

l
normal 59 . The IgG3 subclass contributes to the with antibody-dependent thrombocytopenia, there

Sr
pathogenesis of PTP93. Because multiple subclasses should be antibodies against already-known epitopes
are often detected in patients with thrombocytopenia93 present on the six molecules GPIIb, GPIIIa, GPIb,
and the causality of immunisation seems to have a GPIb, GPIa, or CD109. Otherwise, antibodies against

i
low impact on the repartition of subclasses, subclass
analysis is not considered clinically relevant94; however,
v iz
new epitopes on the six molecules or against molecules
other than these six molecules should be present. A large
immunoglobulin subclasses should be kept in mind. number of molecules are expressed by platelets as well
as by other blood cells. Antibodies to these molecules
er

Glycosylation pattern of immunoglobulin heavy chain may cause thrombocytopenia. We should, therefore,
Immunoglobulin molecules comprise heavy and pay attention to non-HPA molecules that are expressed
S

light chains. The Fab and Fc domains of antibodies are by platelets. When thombocytopenias of non-immune
important for recognition of antigen and binding to Fc origin are discarded by clinical or other diagnosis, we
TI

receptors, respectively. Furthermore, the glycosylation must assume that a causative antibody is present that
pattern of the Fc domain affects its binding affinity for eludes detection by the available analytical techniques.
M

Fc receptors. Structural studies of Fc domains show


that N297 glycans stabilise an "open" Fc conformation Conclusion
recognised by Fc receptors95. Okazaki showed that Improvements in technology help to identify the
SI

depleting fucose from the oligosaccharide moiety of antibodies and antigens that mediate alloimmune
human IgG1 increases the moiety's affinity for the Fc thrombocytopenia, and management strategies are
receptor. Other studies show that modulation of core- under development. However, illuminating the
©

fucosylation of IgG may exert a profound effect on detailed mechanisms of this disease is a daunting
disease severity and prognosis of rheumatic diseases96 task. Success in meeting this challenge is essential
as well as the antibody-dependent cytotoxicity of for the health of patients and to use an increasingly
alloantibodies against HPA97. scarce resource effectively. The underlying complex
This concept has been exploited in the manufacture problems described here should be overcome in the
of recombinant medical products and antibody future; however, strenuous efforts are required to
drugs. Although still at the level of basic research, improve patients' care. Given the common goals but
in the field of platelet transfusion, Bakchoul et al. limited resources and considering how difficult it is
reported that a deglycosylated monoclonal mouse anti- for a single laboratory to obtain a large number of
HPA-1a antibody prevented anti-HPA-1a-mediated specimens, international collaboration is required to
platelet destruction in a mouse model 98. Further, meet these challenges.
Ghevaert et al. constructed a recombinant high-affinity
anti-HPA-1a IgG antibody, B2G1Δnab, comprising Acknowledgements
the variable region of a high-affinity antibody against We thank the staff of the Japanese Red Cross
HPA-1a and constant region modified to minimise Fc Kinki Block Blood Centre for invigorating discussions
receptor-dependent platelet destruction. B2G1Δnab about platelet alloimmunisation. We also thank Drs.

Blood Transfus 2015; 13; 380-90 DOI 10.2450/2015.0275-14


387

All rights reserved - For personal use only


No other uses without permission
Hayashi T, Hirayama F

E. Amakishi, K. Yasui, N. Matsuyama, R.A. Furuta, 21) Santoso S, Kiefel V, Richter IG, et al. A functional platelet
and S. Tanaka for their valuable comments during the fibrinogen receptor with a deletion in the cysteine-rich
repeat region of the beta(3) integrin: the Oe(a) alloantigen
preparation of this review. in neonatal alloimmune thrombocytopenia. Blood 2002;
99: 1205-14.
Keywords: human platelet antigen, antibody detection, 22) Stafford P, Garner SF, Huiskes E, et al. Three novel beta3
genotyping. domain-deletion peptides for the sensitive and specific
detection of HPA-4 and six low frequency beta3-HPA
antibodies. J Thromb Haemost 2008; 6: 376-83.
The Authors declare no conflict of interest. 23) Koh Y, Taniue A, Ishii H, et al. Neonatal alloimmune
thrombocytopenia caused by an antibody specific for a newly
References identified allele of human platelet antigen-7. Transfusion
1) Hod E, Schwartz J. Platelet transfusion refractoriness. Br J 2010; 50: 1276-84.
Haematol 2008; 142: 348-60. 24) Sajid M, Stouffer GA. The role of alpha(v)beta3 integrins
2) Hanson SR, Slichter SJ. Platelet kinetics in patients with bone in vascular healing. Thromb Haemost 2002; 87: 187-93.
marrow hypoplasia: evidence for a fixed platelet requirement. 25) Calvete JJ. On the structure and function of platelet integrin
Blood 1985; 66: 1105-9. alpha IIb beta 3, the fibrinogen receptor. Proc Soc Exp Biol
3) Munnix IC, Cosemans JM, Auger JM, Heemskerk JW. Med 1995; 208: 346-60.
Platelet response heterogeneity in thrombus formation. 26) Bellucci S, Caen J. Molecular basis of Glanzmann's
Thromb Haemost 2009; 102: 1149-56. thrombasthenia and current strategies in treatment. Blood
4) Ross R. Platelet-derived growth factor. Annu Rev Med Rev 2002; 16: 193-202.
1987; 38: 71-9. 27) Ohto H. Neonatal alloimmune thrombocytopenia. Nippon

l
5) Hoffman R. Hematology: Basic Principles and Practice. 4th Rinsho 1997; 55: 2310-4. [In Japanese].

Sr
ed. London: Elsevier; 2005. 28) Pappalardo PA, Secord AR, Quitevis P, et al. Platelet
6) Rivera J, Lozano ML, Navarro-Nunez L, Vicente V. Platelet transfusion refractoriness associated with HPA-1a (Pl(A1))
receptors and signaling in the dynamics of thrombus alloantibody without coexistent HLA antibodies successfully
treated with antigen-negative platelet transfusions.

i
formation. Haematologica 2009; 94: 700-11.
7) Jackson SP, Nesbitt WS, Kulkarni S. Signaling events izTransfusion 2001; 41: 984-7.
underlying thrombus formation. J Thromb Haemost 2003; 29) Kunishima S, Hayakawa A, Fujita K, Saito H. Transient
1: 1602-12. macrothrombocytopenia associated with maternal-neonatal
v
8) Reininger AJ, Heijnen HF, Schumann H, et al. Mechanism of HPA-21bw incompatibility. Thromb Res 2013; 131: e286-8.
30) Kickler TS, Herman JH, Furihata K, et al. Identification
er
platelet adhesion to von Willebrand factor and microparticle
formation under high shear stress. Blood 2006; 107: 3537-45. of Bakb, a new platelet-specific antigen associated with
9) Estavillo D, Ritchie A, Diacovo TG, Cruz MA. Functional posttransfusion purpura. Blood 1988; 71: 894-8.
S

analysis of a recombinant glycoprotein Ia/IIa (Integrin 31) Engelfriet CP, van der H, van LJ Jr. The use of chicken
alpha(2)beta(1)) I domain that inhibits platelet adhesion to red cells in the Coombs consumption test as a specific and
collagen and endothelial matrix under flow conditions. J Biol simple method for the detection of the L.E. factor. Vox Sang
TI

Chem 1999; 274: 35921-6. 1959; 4: 485-92.


10) Ruggeri ZM, Mendolicchio GL. Adhesion mechanisms in 32) Simon TL, Collins J, Kunicki TJ, et al. Posttransfusion
platelet function. Circ Res 2007; 100: 1673-85. purpura associated with alloantibody specific for the platelet
M

11) Metcalfe P, Watkins NA, Ouwehand WH, et al. Nomenclature antigen, Pen(a). Am J Hematol 1988; 29: 38-40.
of human platelet antigens. Vox Sang 2003; 85: 240-5. 33) Taaning E, Killmann SA, Morling N, et al. Post-transfusion
12) Robinson J, Marsh SG. IPD: the Immuno Polymorphism purpura (PTP) due to anti-Zwb (-PlA2): the significance of
SI

Database. Methods Mol Biol 2007; 409: 61-74. IgG3 antibodies in PTP. Br J Haematol 1986; 64: 217-25.
13) Warkentin TE, Smith JW. The alloimmune thrombocytopenic 34) McMillan R. The pathogenesis of chronic immune
syndromes. Transfus Med Rev 1997; 11: 296-307. thrombocytopenic purpura. Semin Hematol 2007; 44: S3-
S11.
©

14) Kaplan C. Foetal and neonatal alloimmune thrombocytopaenia.


Orphanet J Rare Dis 2006; 1: 39. 35) Hoffmeister KM, Felbinger TW, Falet H, et al. The clearance
15) Lucas G, Culliford S, Green F, et al. Recipient-derived mechanism of chilled blood platelets. Cell 2003; 112: 87-97.
HPA-1a antibodies: a cause of prolonged thrombocytopenia 36) Kunishima S, Imai T, Kobayashi R, et al. Bernard-Soulier
after unrelated donor stem cell transplantation. Transfusion syndrome caused by a hemizygous GPIbß mutation and
2010; 50: 334-9. 22q11.2 deletion. Pediatr Int 2013; 55: 434-7.
16) A s t e r R H , B o u g i e D W. D r u g - i n d u c e d i m m u n e 37) Kuijpers RW, Faber NM, Cuypers HT, et al. NH2-terminal
thrombocytopenia. N Engl J Med 2007; 357: 580-7. globular domain of human platelet glycoprotein Ib alpha has
17) Curtis BR, Edwards JT, Hessner MJ, et al. Blood group A a methionine 145/threonine145 amino acid polymorphism,
and B antigens are strongly expressed on platelets of some which is associated with the HPA-2 (Ko) alloantigens. J Clin
individuals. Blood 2000; 96: 1574-81. Invest 1992; 89: 381-4.
18) Vassallo RR. Recognition and management of antibodies 38) Sachs UJ, Kiefel V, Bohringer M, et al. Single amino
to human platelet antigens in platelet transfusion-refractory acid substitution in human platelet glycoprotein Ibbeta
patients. Immunohematology 2009; 25: 119-24. is responsible for the formation of the platelet-specific
19) Kashiwagi H, Honda S, Take H, et al. Presence of the entire alloantigen Iy(a). Blood 2000; 95: 1849-55.
coding region of GP IV mRNA in Nak(a)-negative platelets. 39) Tsubaki K. Anti-platelet antibody by alloimmunization after
Int J Hematol 1993; 57: 153-61. frequent blood transfusion in patients with hematological
20) Arinsburg SA, Shaz BH, Westhoff C, Cushing MM. disorders. [Rinsho ketsueki] Jpn J Clin Hematol 1993; 34:
Determination of human platelet antigen typing by molecular 588-92. [In Japanese] .
methods: importance in diagnosis and early treatment of neonatal 40) Kuijpers RW, van den Anker JN, Baerts W, von dem Borne
alloimmune thrombocytopenia. Am J Hematol 2012; 87: 525-8. AE. A case of severe neonatal thrombocytopenia with

Blood Transfus 2015; 13; 380-90 DOI 10.2450/2015.0275-14


388

All rights reserved - For personal use only


No other uses without permission
Current concepts of human platelet antigens

schizencephaly associated with anti-HPA-1b and anti-HPA- 61) von dem Borne AE, Verheugt FW, Oosterhof F, et al. A
2a. Br J Haematol 1994; 87: 576-9. simple immunofluorescence test for the detection of platelet
41) Kunicki TJ, Kritzik M, Annis DS, Nugent DJ. Hereditary antibodies. Br J Haematol 1978; 39: 195-207.
variation in platelet integrin alpha 2 beta 1 density is 62) Ishida F, Saji H, Maruya E, Furihata K. Human platelet-
associated with two silent polymorphisms in the alpha 2 gene specific antigen, Siba, is associated with the molecular
coding sequence. Blood 1997; 89: 1939-43. weight polymorphism of glycoprotein Ib alpha. Blood 1991;
42) Ohto H, Miura S, Ariga H, et al. The natural history of 78: 1722-9.
maternal immunization against foetal platelet alloantigens. 63) Porcelijn L, Huiskes E, Comijs-van Osselen I, et al. A new
Transfus Med 2004; 14: 399-408. bead-based human platelet antigen antibodies detection assay
43) Skouri H, Gandouz R, Kraiem I, et al. Platelet-specific versus the monoclonal antibody immobilization of platelet
alloantigens and antibodies in Tunisian women after three or antigens assay. Transfusion 2014; 54: 1486-92.
more pregnancies. Transfus Med 2009; 19: 269-73. 64) Fujiwara K, Shimano K, Tanaka H, et al. Application of
44) Kekomaki S, Koskela S, Laes M, et al. Neonatal bead array technology to simultaneous detection of human
thrombocytopenia in two of six human platelet alloantigen leucocyte antigen and human platelet antigen antibodies. Vox
(HPA) 5a-positive children of an HPA-5a-immunized mother. Sang 2009; 96: 244-51.
Transfus Med 2000; 10: 81-5. 65) Campbell K, Rishi K, Howkins G, et al. A modified rapid
45) Lin M, Sutherland DR, Horsfall W, et al. Cell surface antigen monoclonal antibody-specific immobilization of platelet
CD109 is a novel member of the alpha(2) macroglobulin/ antigen assay14th for the detection of human platelet antigen
C3, C4, C5 family of thioester-containing proteins. Blood (HPA) antibodies: a multicentre evaluation. Vox Sang 2007;
2002; 99: 1683-91. 93: 289-97.
46) Hagiwara S, Murakumo Y, Mii S, et al. Processing of CD109 66) Wu GG, Kaplan C, Curtis BR, Pearson HA. Report on the
by furin and its role in the regulation of TGF-beta signaling. 14th International Society of Blood Transfusion. Vox Sang

l
Oncogene 2010; 29: 2181-91. 2010; 99: 375-81.

Sr
47) Berry JE, Murphy CM, Smith GA, et al. Detection of Gov 67) Morel-Kopp MC, Daviet L, McGregor J, Kaplan C. Drawbacks
system antibodies by MAIPA reveals an immunogenicity of the MAIPA technique in characterising human antiplatelet
similar to the HPA-5 alloantigens. Br J Haematol 2000; antibodies. Blood Coagul Fibrinolysis 1996; 7: 144-6.
110: 735-42. 68) Hayashi T, Yasui K, Matsuyama N, et al. Establishment of a

i
48) Ertel K, Al-Tawil M, Santoso S, Kroll H. Relevance of the novel method for detecting Nak antibodies by using a panel
iz
HPA-15 (Gov) polymorphism on CD109 in alloimmune cell line. Transfusion 2009; 49: 390-2.
thrombocytopenic syndromes. Transfusion 2005; 45: 366-73. 69) Hayashi T, Amakishi E, Matsuyama N, et al. Detection of
v
49) Matsuhashi M, Tsuno NH, Kawabata M, et al. The first antibodies against human platelet antigens 15a and 15b by
case of alloantibody against human platelet antigen-15b in using a cell line panel. Br J Haematol 2010; 151: 402-4.
er
Japan: possible alloimmunization by a hydatidiform mole. 70) Hayashi T, Amakishi E, Inoue M, Hirayama F. Establishment
Transfusion 2010; 50: 1126-30. of a cell line panel for the detection of antibodies against
S

50) Yamamoto N, Ikeda H, Tandon NN, et al. A platelet human platelet antigen 4b. Int J Hematol 2011; 93: 170-5.
membrane glycoprotein (GP) deficiency in healthy blood 71) Hayashi T, Amakishi E, Matsuyama N, et al. Establishment of
donors: Naka- platelets lack detectable GPIV (CD36). Blood a cell line panel as an alternative source of platelet antigens
TI

1990; 76: 1698-703. for a screening assay of anti-human platelet antibodies.


51) Rac ME, Safranow K, Poncyljusz W. Molecular basis of Transfus Med 2011; 21: 199-204.
human CD36 gene mutations. Mol Med 2007; 13: 288-96. 72) Hayashi T, Amakishi E, Matsuyama N, et al. Detection of
M

52) Greenwalt DE, Lipsky RH, Ockenhouse CF, et al. Membrane anti-human platelet antibodies against integrin alpha2beta1
glycoprotein CD36: a review of its roles in adherence, signal using cell lines. Blood Transfus 2014; 12: s273-80.
transduction, and transfusion medicine. Blood 1992; 80: 73) Amakishi E, Hayashi T, Koh Y, et al. A new transfectant
SI

1105-15. panel cell line-based MoAb-independent antigen capture


53) Toba K, Hanawa H, Watanabe K, et al. Erythroid involvement assay system for detection of CD36 antibody. Vox Sang
in CD36 deficiency. Exp Hematol 2001; 29: 1194-200. 2014; 106: 368-71.
54) Yanai H, Chiba H, Fujiwara H, et al. Phenotype-genotype 74) Santoso S, Kiefel V. Human platelet alloantigens. Wien Klin
©

correlation in CD36 deficiency types I and II. Thromb Wochenschr 2001; 113: 806-13.
Haemost 2000; 84: 436-41. 75) Peterson JA, Kanack A, Nayak D, et al. Prevalence and
55) Lee K, Godeau B, Fromont P, et al. CD36 deficiency is clinical significance of low-avidity HPA-1a antibodies in
frequent and can cause platelet immunization in Africans. women exposed to HPA-1a during pregnancy. Transfusion
Transfusion 1999; 39: 873-9. 2013; 53: 1309-18.
56) Curtis BR, Aster RH. Incidence of the Nak(a)-negative 76) Bakchoul T, Bertrand G, Krautwurst A, et al. The implementation
platelet phenotype in African Americans is similar to that of of surface plasmon resonance technique in monitoring
Asians. Transfusion 1996; 36: 331-4. pregnancies with expected fetal and neonatal alloimmune
57) Okajima S, Cho K, Chiba H, et al. Two sibling cases thrombocytopenia. Transfusion 2013; 53: 2078-85.
of hydrops fetalis due to alloimmune anti-CD36 (Naka) 77) Thibaut J, Merieux Y, Rigal D, Gillet G. A novel assay for the
antibody. Thromb Haemost 2006; 95: 267-71. detection of anti-human platelet antigen antibodies (HPA-1a)
58) Kiefel V. The MAIPA assay and its applications in based on peptide aptamer technology. Haematologica 2012;
immunohaematology. Transfus Med 1992; 2: 181-8. 97: 696-704.
59) K e l s c h R , H u t t K , C a s s e n s U , S i b r o w s k i W. 78) Harrison CR, Curtis BR, McFarland JG, et al. Severe neonatal
Semiquantitative measurement of IgG subclasses and alloimmune thrombocytopenia caused by antibodies to
IgM of platelet-specific antibodies in a glycoprotein- human platelet antigen 3a (Baka) detectable only in whole
specific platelet-antigen capture assay. Br J Haematol platelet assays. Transfusion 2003; 43: 1398-402.
2002; 117: 141-50. 79) Kataoka S, Kobayashi H, Chiba K, et al. Neonatal
60) Shibata Y, Juji T, Nishizawa Y, et al. Detection of platelet alloimmune thrombocytopenia due to an antibody against
antibodies by a newly developed mixed agglutination with a labile component of human platelet antigen-3b (Bak b).
platelets. Vox Sang 1981; 41: 25-31. Transfus Med 2004; 14: 419-23.

Blood Transfus 2015; 13; 380-90 DOI 10.2450/2015.0275-14


389

All rights reserved - For personal use only


No other uses without permission
Hayashi T, Hirayama F

80) Socher I, Zwingel C, Santoso S, Kroll H. Heterogeneity 93) Proulx C, Filion M, Goldman M, et al. Analysis of
of HPA-3 alloantibodies: consequences for the diagnosis immunoglobulin class, IgG subclass and titre of HPA-1a
of alloimmune thrombocytopenic syndromes. Transfusion antibodies in alloimmunized mothers giving birth to babies
2008; 48: 463-72. with or without neonatal alloimmune thrombocytopenia. Br
81) de Leon EJ, Yuan FF, Pearson H, et al. Evidence of J Haematol 1994; 87: 813-7.
heterogeneity in the antibody response against the platelet 94) Prigent A, Maillard N, Absi L, et al. From donor to recipient:
antigen 3a; recognition of an 11-mer peptide carrying the current questions relating to humoral alloimmunization.
HPA-3a polymorphic determinant. Vox Sang 2009; 96: 252-5. Antibodies 2014; 3: 130-52.
82) Allen DL, Metcalfe P, Kaplan C, et al. Sensitivity of 95) Borrok MJ, Jung ST, Kang TH, et al. Revisiting the role of
assays for the detection of HPA-1a antibodies: results of an glycosylation in the structure of human IgG Fc. ACS Chem
international workshop demonstrating the impact of cation Biol 2012; 7: 1596-602.
chelation from integrin alphaIIbbeta3 on three widely used 96) Axford JS. Glycosylation and rheumatic disease. Biochim
assays. Vox Sang 2013; 105: 167-73. Biophys Acta 1999; 1455: 219-29.
83) Peterson JA, Gitter M, Bougie DW, et al. Low-frequency 97) Wuhrer M, Porcelijn L, Kapur R, et al. Regulated
human platelet antigens as triggers for neonatal alloimmune glycosylation patterns of IgG during alloimmune responses
thrombocytopenia. Transfusion 2014; 54: 1286-93. against human platelet antigens. J Proteome Res 2009; 8:
84) Nai S, Ghosh K, Shetty S, Mohanty D. Mutations in GPIIIa 450-6.
molecule as a cause for Glanzmann thrombasthenia in Indian 98) Bakchoul T, Greinacher A, Sachs UJ, et al. Inhibition
patients. J Thromb Haemost 2005; 3: 482-8. of HPA-1a alloantibody-mediated platelet destruction
85) Curtis BR. Genotyping for human platelet alloantigen by a deglycosylated anti-HPA-1a monoclonal antibody
polymorphisms: applications in the diagnosis of alloimmune in mice: toward targeted treatment of fetal-alloimmune
platelet disorders. Semin Thromb Hemost 2008; 34: 539-48. thrombocytopenia. Blood 2013; 122: 321-7.

l
86) Hayashi T, Ishii H, Tanaka M, et al. High-resolution melting 99) Ghevaert C, Wilcox DA, Fang J, et al. Developing

Sr
method for genotyping human platelet antigens on ITGB3 recombinant HPA-1a-specific antibodies with abrogated
exon 11. Transfusion 2012; 52: 1837-8. Fcgamma receptor binding for the treatment of fetomaternal
87) Le Toriellec E, Chenet C, Kaplan C. Safe fetal platelet alloimmune thrombocytopenia. J Clin Invest 2008; 118:
genotyping: new developments. Transfusion 2013; 53: 2929-38.

i
1755-62. 100)
iz Ghevaert C, Herbert N, Hawkins L, et al. Recombinant HPA-
88) Liew M, Nelson L, Margraf R, et al. Genotyping of human 1a antibody therapy for treatment of fetomaternal alloimmune
platelet antigens 1 to 6 and 15 by high-resolution amplicon thrombocytopenia: proof of principle in human volunteers.
v
melting and conventional hybridization probes. J Mol Diagn Blood 2013; 122: 313-20.
2006; 8: 97-104.
er
89) Zhou SH, Liu M, An WX, et al. Genotyping of human platelet
antigen-15 by single closed-tube Tm-shift method. Int J Lab
S

Hematol 2012; 34: 41-6.


90) Fujiwara K, Isa K, Oka T, et al. Large-scale DNA typing
for human platelet alloantigens by PCR-PHFA (preferential
TI

homoduplex formation assay). Br J Haematol 1996; 95:


198-203.
91) Bertrand G, Jallu V, Beranger T, et al. HPA-5 typing
M

discrepancy reveals an Ile503Leu substitution in platelet


Arrived: 29 October 2014 - Revision accepted: 15 February 2015
GPIa (alpha2 integrin). Vox Sang 2013; 105: 73-6.
Correspondence: Tomoya Hayashi
92) Ambo H, Kamata T, Handa M, et al. Three novel integrin Japanese Red Cross
SI

beta3 subunit missense mutations (H280P, C560F, and Kinki Block Blood Center
G579S) in thrombasthenia, including one (H280P) prevalent 7-5-17 Saito Asagi
in Japanese patients. Biochem Biophys Res Commun 1998; Ibaraki, Osaka, Japan
251: 763-8. e-mail: to-hayashi@kk.bbc.jrc.or.jp
©

Blood Transfus 2015; 13; 380-90 DOI 10.2450/2015.0275-14


390

All rights reserved - For personal use only


No other uses without permission

You might also like