Você está na página 1de 16

From bloodjournal.hematologylibrary.org by guest on June 12, 2013. For personal use only.

1997 89: 3077-3091

From Malaria to Chemokine Receptor: The Emerging Physiologic Role of the Duffy Blood Group Antigen
Terence J. Hadley and Stephen C. Peiper

Updated information and services can be found at: http://bloodjournal.hematologylibrary.org/content/89/9/3077.full.html Articles on similar topics can be found in the following Blood collections Review Articles (449 articles) Information about reproducing this article in parts or in its entirety may be found online at: http://bloodjournal.hematologylibrary.org/site/misc/rights.xhtml#repub_requests Information about ordering reprints may be found online at: http://bloodjournal.hematologylibrary.org/site/misc/rights.xhtml#reprints Information about subscriptions and ASH membership may be found online at: http://bloodjournal.hematologylibrary.org/site/subscriptions/index.xhtml

Blood (print ISSN 0006-4971, online ISSN 1528-0020), is published weekly by the American Society of Hematology, 2021 L St, NW, Suite 900, Washington DC 20036. Copyright 2011 by The American Society of Hematology; all rights reserved.

From bloodjournal.hematologylibrary.org by guest on June 12, 2013. For personal use only.

BLOOD
VOL 89, NO 9
REVIEW ARTICLE

The Journal of The American Society of Hematology


MAY 1, 1997

From Malaria to Chemokine Receptor: The Emerging Physiologic Role of the Duffy Blood Group Antigen
By Terence J. Hadley and Stephen C. Peiper

NE OF THE MOST notable achievements of biomedical research in the rst half of this century was the identication of red blood cell (RBC) antigens and the recognition of their importance to transfusion medicine and hemolytic disease of the newborn.1,2 These discoveries gave rise to an era of descriptive RBC serology during which a complex array of RBC membrane antigens were dened by their reactivity with specic alloantibodies.3,4 In recent years, RBC immunohematology has progressed from descriptive serology into an era of structural and functional analysis of blood group antigens, many of which are expressed in both erythroid and nonerythroid tissue.5-8 Here we will focus on the Duffy blood group antigen, a structure that has been of particular interest because it serves as a receptor on the RBC for the malarial parasite, Plasmodium vivax (P vivax ).8-10 We will attempt to highlight recent advances in our understanding of the molecular basis for the interaction of the P vivax malaria parasite with the Duffy blood group antigen and indicate how this information also contributed to the identication of an important gene family for cytoadherence proteins of P falciparum. In the context of normal physiology, the Duffy blood group antigen has been shown to be receptor for chemoattractant cytokines, or chemokines, and to be expressed by endothelial cells of postcapillary venules and by Purkinje cells of the cerebellum.11,12 We will attempt to review this important area of chemokine receptor research and discuss the potential relevance of the Duffy chemokine receptor to immunology and neurobiology. Duffy Blood Group Serology The Duffy blood group system rst came to light in a report by Cutbush et al13 describing an alloantibody against an antigen denoted as Fya in a patient with hemophilia who had received multiple transfusions. The antithetical antigen, Fyb, was described 1 year later.14 Three phenotypes were identied in whites using anti-Fya and anti-Fyb antisera: Fy(a/b0), Fy(a0b/), and Fy(a/b/).15 These phenotypes are the products of codominant alleles comprising genotypes FY*A/FY*A, FY*B/FY*B, and FY*A/FY*B, respectively. Some whites express a weak or quantitatively reduced Fyb, the genetic basis of which remains uncertain.16 Most West Africans and 68% of African Americans do not express Fya or Fyb on their RBCs.17,18 The absence of Fya and Fyb on erythrocytes is designated the Duffy negative
Blood, Vol 89, No 9 (May 1), 1997: pp 3077-3091

phenotype and denoted as Fy(a0b0). When Duffy-negative individuals of African descent develop anti-Duffy alloantibodies, they are almost always anti-Fya rather than antiFyb.19,20 The genotype designated FY/FY, which gives rise to the Fy(a0b0) erythroid phenotype, contains a coding sequence identical to that of FY*B.21 In individuals of the Fy(a0b0) phenotype, this sequence remains silent in erythroid cells but is transcribed and expressed on endothelial cells of postcapillary venules.22 Reactivity with anti-Fya and anti-Fyb alloantibodies is abolished after chymotrypsin or papain treatment of intact RBCs.23,24 Albrey et al25 described an anti-Duffy alloantibody, denoted anti-Fy3, that reacts with chymotrypsinand papain-treated RBCs. Anti-Fy3 reacts with both Fy(a/b0) and Fy(a0b/) RBCs, but not Fy(a0b0) RBCs. Adsorption and elution experiments showed that anti-Fy3 reacts with a site common to both Fy(a/b0) and Fy(a0b/) RBCs. Although initially described in the serum of a rare Fy(a0b0) white woman (AZ) with a history of pregnancy and blood transfusions, anti-Fy3 can also occur in Duffy-negative individuals of African descent, often in conjunction with anti-Fya.18 Other Duffy-related epitopes have been dened by rare antisera, anti-Fy4 and anti-Fy5.26,27 Anti-Fy4 reacts only with Fy(a0b0) RBCs from individuals of African descent.26 Anti-Fy5 reacts with all human RBCs except those that are Fy(a0b0) and Rh null or express a variant of the e antigen of the Rh system.27 The rarity of these antisera has impeded progress in characterizing their epitopes.

From the Departments of Medicine, Pathology, and Biochemistry, Henry Vogt Cancer Research Institute, James Graham Brown Cancer Center, University of Louisville; and the Department of Veterans Affairs Medical Center, Louisville, KY. Submitted July 17, 1996; accepted December 13, 1996. Supported by a Merit Review Grant from the Ofce of Research and Development, Medical Research Service, Department of Veterans Affairs, by the Agnes Brown Duggan Endowment for Oncologic Research, and the the Humana Endowment for Excellence. Address reprint requests to Terence J. Hadley, MD, James Graham Brown Cancer Center, University of Louisville, 529 S Jackson St, Louisville, KY 40292. 1997 by The American Society of Hematology. 0006-4971/97/8909-0030$3.00/0
3077

From bloodjournal.hematologylibrary.org by guest on June 12, 2013. For personal use only.
3078 HADLEY AND PEIPER

THE DUFFY ANTIGEN IS A GLYCOPROTEIN

Moore et al28 showed that when surface-radioiodinated Fy(a/b0) RBCs were incubated with anti-Fya before solubilization in detergent, a protein of 35 to 43 kD was specically immunoprecipitated. Coprecipitation of a radiolabeled protein other than the Duffy antigen was not formally excluded. More conclusive Western blotting experiments conrmed that a 35- to 43-kD protein carries the Fya antigenic determinant.29 Desialylation of RBC membranes with Vibrio cholera neuraminidase resulted in an altered electrophoretic mobility of the 35- to 43-kD Fya protein, suggesting that the Duffy antigen is a glycoprotein.30 Treatment of RBC membranes with N-glycanase caused a shift in the apparent molecular weight of the Duffy protein to around 30 kD.30,31 Although treatment of intact RBCs with trypsin did not proteolytically cleave the Duffy antigen, treatment of partially puried Duffy antigen with trypsin resulted in a 28-kD fragment.32 After incremental digestion of this 28-kD Duffy tryptic peptide with N-glycanase, Western blotting resolved multiple species, indicating that this glycoprotein has two or three asparagine-linked oligosaccharide chains.33 Parallel digestion with O-glycanase had no effect on the electrophoretic mobility of the glycoprotein, signifying the absence of oligosaccharide chains linked to serine and threonine residues.
MONOCLONAL ANTIBODIES (MoAbs) TO THE DUFFY ANTIGEN

In 1987, Nichols et al34 reported on the rst MoAb (NYBC-BG6) against the Duffy antigen. This MoAb dened a new epitope, denoted Fy6, which is common to Fya and Fyb, but absent from Fy(a0b0) erythrocytes. Treatment of intact RBCs with chymotrypsin destroyed the Fy6 epitope as well as Fya and Fyb.35 An anti-Fy6 MoAb (i3A) similar to NYBC-BG6 was produced by Riwom et al36 and a MoAb (CBC-512) with a specicity similar to that of anti-Fy3 (reactivity not destroyed by treatment of RBCs with chymotrypsin or papain) was produced by Dr M. Uchikawa at the Japanese Red Cross (see Acknowledgment) (Table 1).
DUFFY IS INVOLVED IN P VIVAX PATHOPHYSIOLOGY

As a background to discussing the Duffy antigen in relation to malaria, a brief overview of the malarial life cycle will be presented. Malaria is transmitted by female anophiline mosquitos. When an infected mosquito takes a blood meal, sporozoites enter the blood and invade hepatocytes where they divide into thousands of merozoites. This part of the life cycle produces no symptoms in the host. Merozoites emerge from the liver and enter the the blood where they

invade erythrocytes. This stage of the life cycle is associated with the clinical illness. Inside erythrocytes, merozoites develop sequentially into ring forms, trophozoites, and schizonts; mature schizonts give rise to many new merozoites. Infected erythrocytes ultimately lyse (rupture) and emerging merozoites invade other erythrocytes. Some ring forms develop into gametocytes instead of schizonts. Gametocytes are responsible for the transmission of malaria back to the mosquito when the mosquito takes a blood meal. There are four species of human malaria: P falciparum, P vivax, P ovale, and P malariae. Of these, P falciparum and P vivax are the most prevalent. P falciparum causes the most mortality; P vivax is second to P falcipaurm as a cause of malaria-related morbidity. P knowlesi, a primate malaria related to P vivax and capable of invading human RBCs, has been used as a laboratory model to investigate how malaria parasites invade RBCs. In 1975, Miller et al reported that human erythrocytes of the Fy(a0b0), or Duffy-negative phenotype, were resistant to invasion by merozoites (the RBC invasive form) of P knowlesi.37,38 Human erythrocytes of all other phenotypes were invaded. Furthermore, anti-Fya and anti-Fyb specically inhibited invasion of P knowlesi merozoites into human RBCs of the Fy(a/b0) and Fy(a0b/) phenotype, respectively.37 Cytochalasin Btreated merozoites were used to study attachment independent of invasion.39 Cytochalasin B treated merozoites of P knowlesi attached equally well to both Duffy-positive and Duffy-negative erythrocytes. Ultrastructural analysis of Duffy positive erythrocytes with adherent merozoites showed an electron dense structure beneath the RBC lipid bilayer in the area of apposition between RBC and merozoite.39 Previous studies had demonstrated that this structure, termed a tight junction, is of crucial importance for parasite invasion.40 The tight junction was not observed with merozoites adherent to Duffy-negative erythrocytes. The nding that P knowlesi requires the Duffy antigen to form a tight junction and to invade human erythrocytes suggested an explanation for two observations related to the human malaria, P vivax. First, P vivax, which is prevalent in most tropical and subtropical areas of the world, is specically absent from West Africa, a geographic area where greater than 95% of individuals are Fy(a0b0). Second, during treatment of neurosyphilitic patients with therapeutically induced malaria, many African Americans were noted to be innately resistant to P vivax.41 Miller et al42 postulated that the innate resistance of some African Americans to P vivax malaria might be related to the Duffy-negative phenotype. A number of studies conrmed this hypothesis and demon-

Table 1. Available Human Polyclonal and Murine Monoclonal (MoAb) Anti-Duffy Antibodies
Antibody Reactive RBCs Nonreactive RBCs Sensitive (S) or Resistant (R) to Chymotrypsin

Anti-Fya (human) Anti-Fyb (human) Anti-Fy3 (human; MoAb) Anti-Fy6 (MoAb)

Fy(a/b0), Fy(a0b/), Fy(a/b0), Fy(a/b0),

Fy(a/b/) Fy(a/b/) Fy(a0b/) Fy(a/b/) Fy(a0b/) Fy(a/b/)

Fy(a0b/), Fy(a0b0) Fy(a/b0), Fy(a0b0) Fy(a0b0) Fy(a0b0)

S S R S

From bloodjournal.hematologylibrary.org by guest on June 12, 2013. For personal use only.
DUFFY ANTIGEN RECEPTOR FOR CHEMOKINES 3079

strated a correlation between the Fy(a0b0) phenotype and resistance to P vivax infection.42-44 It was subsequently shown that Fy(a0b0) erythrocytes cannot be invaded by P vivax in vitro and that the anti-Fy6 MoAb, NYBC-BG6, can block invasion of Duffy-positive erythrocytes by P vivax.34,35 One of the distinguishing features of P vivax is its preference for reticulocytes.45 Because the Duffy antigen is expressed on mature erythrocytes as well as on reticulocytes, it has been postulated that in addition to the Duffy glycoprotein, P vivax parasites require a second RBC receptor for invasion that is specic for reticulocytes. Although the reticulocyte-specic RBC receptor for invasion by P vivax has not yet been identied, a P vivax ligand that binds specically to reticulocytes has been identied and cloned.46
DUFFY BINDING LIGANDS OF P KNOWLESI AND P VIVAX

Haynes et al47 identied a parasite protein (Duffy binding ligand) from P knowlesi that binds specically to Duffypositive erythrocytes but not to Duffy-negative erythrocytes. The 135-kD Duffy binding ligand of P knowlesi also binds to rhesus RBCs, which serologically react as Fy(a0b/) and which express an Fyb-related antigen that is highly homologous, but not identical to, human Fyb.21,47 A similar Duffy binding ligand of 140 kD was isolated by Wertheimer and Barnwell48 from P vivax. Unlike the 135-kD Duffy binding protein of P knowlesi, the 140-kD Duffy binding protein of P vivax does not bind to rhesus RBCs, a cell type that cannot be invaded by P vivax.48 Recent molecular analyses of the Duffy binding ligands of P knowlesi, P vivax, and the erythrocyte binding ligand of P falciparum (which does not bind to Duffy but to a sialic aciddependent domain on glycophorin A) indicate regions of homology among these erythrocyte binding proteins.49,50 Antibodies prepared against the 135-kD Duffy binding ligand of P knowlesi were used to screen expression complementary DNA (cDNA) libraries prepared from P knowlesi asexual erythrocyte-stage mRNA and cDNA encoding the 135-kD Duffy binding protein was cloned.51 The highly related 140-kD Duffy binding protein of P vivax was identied from cDNA clones obtained by cross-hybridization with the P knowlesi cDNA probe.52 Immunohistochemical studies localized the Duffy binding protein of P knowlesi to the micronemes, specialized organelles at the apical end of the parasite (merozoite) that appear to function in the process of invasion. This further supported the conclusion that Duffy binding proteins are indeed parasite ligands that bind to the Duffy receptor during invasion.51 How these proteins are translocated from micronemes to the apical surface of the merozoite to interact with the Duffy receptor is unknown. The extracellular regions of the 140-kD Duffy binding protein of P vivax and the 135-kD Duffy binding protein of P knowlesi were classied into six domains based on amino acid sequence similarities.51,53 COS 7 cells were transfected with constructs encoding region I, region II, regions III-V, and region VI, each designed to yield cell-surface expression.53 Cells expressing the cysteine-rich region II specically formed rosettes with Duffy-positive, but not Duffynegative, erythrocytes, indicating that this domain is responsible

for interactions with erythrocytes mediated by the Duffy glycoprotein.53 Although the 140-kD Duffy binding ligand of P vivax did not bind to rhesus RBCs in the rosetting assay, binding did occur if rhesus RBCs were rst treated with N-glycanase.54 Synthetic peptides consisting of the rst 35 amino acids from the N-terminus of the human Duffy protein and the rst 34 amino acids from the N-terminus of the rhesus Duffy homolog both blocked rosetting of Duffy-positive human erythrocytes with COS cells expressing the 140-kD Duffy binding protein of P vivax.54 These data support the conclusion that the Duffy homolog on rhesus RBCs contains the peptide binding site for the P vivax Duffy binding protein. However, it appears that on the intact rhesus RBCs, this site is altered or obscured by the presence of one or more N-linked sugar side chains. Thus, it appears that differences in glycosylation may contribute to species specicity of malaria parasite-host cell interactions.
DUFFY BINDING LIGAND MOTIFS IN THE VAR GENES OF P FALCIPARUM

Identication of the Duffy binding proteins of P knowlesi and P vivax provided a clue to the identication of another important malarial gene family, termed the var genes.55-58 Howard et al59-61 had shown that malaria parasites, including P falciparum, insert parasite-derived proteins into the membranes of infected RBCs. Biologic data indicated that one or more of these proteins function in cytoadherence of P falciparum infected RBCs to endothelial cells.62,63 Cytoadherence of P falciparum infected RBCs to endothelial cells of postcapillary venules accounts for the sequestration of mature parasite-infected RBCs in the deep vasculature and their consequent absence on diagnostic blood lms.64,65 Ultrastructural studies of sequestered parasites showed that the points of contact between infected RBCs and endothelial cells consist of electron dense knobs on the RBC membranes.66,67 Sequestration allows parasites to grow within the vasculature of the host without circulating through the spleen, an organ known to have anti-malarial properties.67,68 Cytoadherence of infected RBCs to endothelial cells of cerebral vasculature also contributes to the pathogenesis of cerebral malaria.69 David et al70 demonstrated that treatment of P falciparum infected monkeys with hyperimmune serum reversed sequestration in a strain-specic fashion and led to the appearance of crisis forms (morphologically deteriorating parasites within erythrocytes). Serologic and immunochemical evidence indicated that the endothelial binding proteins on the surface of parasite-infected RBCs undergo antigenic variation, a phenomenon of obvious importance to the parasite as it affords a mechanism of circumventing the effects of specic antibodies that might otherwise block cytoadherence.71-75 Understanding the molecular basis for endothelial cytoadherence and antigenic variation was thwarted for years by difculties purifying sufcient quantities of the malarial endothelial cytoadherence protein for microsequencing. Unexpectedly, analysis of Duffy binding ligands provided a clue to the indentication of genes encoding variant endothelial cytoadherence proteins of P falciparum.55,56 While sequencing DNA from regions of parasite chromo-

From bloodjournal.hematologylibrary.org by guest on June 12, 2013. For personal use only.
3080 HADLEY AND PEIPER

some 7, Peterson et al55 and Su et al56 identied several gene sequences that contain domains similar to region II of the erythrocyte binding proteins of P knowlesi, P vivax, and P falciparum. This family of related sequences possessed properties predicted for genes encoding cytoadherence proteins: they were polymorphic, they contained large open reading frames consistent with the size of putative cytoadherence proteins, and their predicted protein products included multiple domains with regions homologous to Duffy binding proteins, which had already been shown to have cytoadhesive properties. Taking advantage of homologies within these Duffy binding-like domains, Smith et al57 used degenerate oligonucleotide primers to study messenger RNA from these genes (designated variant or var genes) expressed in cloned parasites with serologically distinct surface antigens and cytoadherence properties. They showed that the expression of distinct cytoadhesive properties and antigenic reactivity at the surface of infected erythrocytes correlated with expression of distinct var genes.57 Baruch et al58 cloned cDNA from an expression library for the putative cytoadherence protein, PfEMP1 (P falciparum erythrocyte membrane protein 1), and demonstrated by immunochemical methods that peptides derived from the cDNA had antigenic properties predicted for a cytoadherence protein. These investigators, together with Su et al, showed that cDNA for PfEMP1 is a member of the var gene family.57,58 The studies by Su et al,56 Smith et al,57 and Baruch et al58 were published simultaneously and conrmed that var genes encode related, but antigenically variant, cytoadherence proteins. Identication of var genes of P falciparum represents a major advance in malaria research because it provides the molecular basis for understanding sequestration, antigenic variation, and the chronicity of malaria. There is evidence that each parasite of P falciparum contains a large repertoire of var genes (50 to 150 copies) scattered throughout the malarial genome.57 These genes, which encode highly homologous but antigenically distinct cytoadherence proteins, account for 2% to 6% of the haploid parasite genome.57 Despite antigenic differences among these variant cytoadherence proteins of P falciparum, their capacity to bind to endothelial cells is conserved.56 The molecular basis for this conservation of function in the face of antigenic variation remains to be elucidated, although it is clear that a number of endothelial receptors, including CD36, ICAM, VCAM, E-selectin, and thrombospondin may be involved in cytoadherence.76-78

THE IMPORTANCE OF MALARIAL CYTOADHERENCE TO VACCINE RESEARCH

Cytoadherence of all species of malarial merozoites to host RBCs, followed by invasion, is crucial for the life cycle of these obligate intracellular parasites. Cytoadherence of P falciparum infected RBCs to endothelial cells of postcapillary venules appears crucial to the survival of this particular parasite. The parasite proteins discussed above are part of a newly described superfamily of malarial cytoadherence molecules (Table 2). If these molecules could be used as immunogens to induce antibodies that block cytoadherence, protection against malaria might be achieved (Fig 1). Even if the protection were partial, it would potentially be benecial because many of the pathologic sequelae of malarial infections in the partially immune host are related to the level of parasitemia.79 Because the endothelial cytoadherence molecule of P falciparum is highly variant, it will present a major challenge to development of a malaria vaccine. The parasite ligands that bind to RBC receptors appear much less variant, although there is evidence that P falciparum can invade by more than one pathway and can switch between at least two alternative pathways for invasion.80 Although there is some variation within the adhesion domain of the P vivax Duffy binding protein, this parasite appears to be absolutely dependent on the Duffy pathway for invasion.81 P vivax provides a model for testing the efcacy of an erythrocyte binding proteinbased vaccine, especially now that inhibition of ligand-receptor interaction by immune sera can be tested in vitro using the transfected COS cell-RBC rosetting assay established by Chitnis et al.53,54
THE DUFFY ANTIGEN IS A NOVEL RECEPTOR FOR CHEMOATTRACTANT CYTOKINES

Thus far we have discussed the function of the Duffy antigen in the pathogenesis of malaria. We have attempted to describe how knowledge of the Duffy antigen and its relationship to P knowlesi and P vivax malaria led to identication of malarial Duffy binding ligands, and this in turn provided clues to the identication of endothelial binding ligands of P falciparum. But clearly the Duffy glycoprotein exists for purposes other than malarial invasion of RBCs. Unexpectedly, research on chemoattractant cytokines, abbreviated chemokines, and their receptors converged with investigation on the Duffy blood group antigen, providing insight into a potential normal physiologic role for this allelic glycoprotein.10,11 To put this research into proper perspective, it

Table 2. Malarial Receptor/Adhesion Proteins: Potential Targets of Receptor/Adhesion Blocking Immunity


Parasite Receptor/Adhesion Protein Location Host Counter Receptor

P knowlesi P vivax P falciparum P falciparum

135-kD Duffy binding protein (for invasion) 140-kD Duffy binding protein (for invasion) 170-kD Erythrocyte binding protein (for invasion) var gene products (for sequestration)

Merozoite* Merozoites Merozoites Membrane of infected RBCs

Duffy glycoprotein (DARC) on RBCs Duffy glycoprotein (DARC) on RBCs Sialic Acid-glycophorin A on RBCs CD36, ICAM, VCAM, E-selectin (? thrombospondin) on endothelial cells

* 135-kD Duffy binding protein has been localized to apical organelles (micronemes) of the merozoite. The 170-kD erythrocyte binding protein and the var gene products of P falciparum contain Duffy binding-like domains.

From bloodjournal.hematologylibrary.org by guest on June 12, 2013. For personal use only.
DUFFY ANTIGEN RECEPTOR FOR CHEMOKINES 3081

Fig 1. (A) Two receptors for apical attachment of P vivax merozoites to RBCs have been cloned: the Duffy binding protein and the reticulocyte binding protein.46,52 One strategy for vaccine development is to use these proteins as immunogens to induce antibodies that might block invasion of the parasite into the RBC. (B) Variant cytoadherence proteins of P falciparum have also been cloned.55-57 Antibodies against this protein can block cytoadherence of P falciparum infected RBCs to endothelial cells, forcing the parasite to circulate through the spleen, where immunologic mechanisms (which have not been clearly dened) are brought into play against P falciparum parasites. The cytoadherence antigen is variant and antigenic variation is used by the parasite to escape antibody mediated blockade of cytoadherence. Another strategy for vaccine development is to identify domains that are shared among different variants of the cytoadherece protein of P falciparum; antibodies against shared domains could block cytoadherence in a variant-independent fashion.

is necessary to briey review the biologic activities of chemokines. Chemokines are members of a superfamily of small, secreted proteins (8 to 13 kD), numbering over 20, that recruit leukocytes to sites of inammation.82 This superfamily has two major branches, encoded in separate gene clusters, that preferentially promote acute and chronic inammatory processes.83-85 In addition to the functional differences between the two families, they are also distinguished by structural characteristics and at the level of genomic organization.83-85 Chemokines of the C-X-C family, in which the two amino proximal cysteine residues are separated by one amino acid, function primarily as chemoattractants for neutrophils, whereas chemokines of the C-C family, in which these cysteines are juxtaposed, function primarily as chemoattractants for lymphocytes and monocytes. A newly discovered chemokine, lymphotactin, has a single cysteine, or Cmotif rather than a C-C or C-X-C motif.86 The effects of chemokines are mediated through highafnity receptors that have seven hydrophobic membrane-

spanning helices and signal through coupling with G-proteins.87-92 In general, each chemokine has a specic receptor to which it binds with high afnity and another receptor to which it binds with lower afnity and with which it shares binding with another chemokine of the same family. In 1991, Darbonne et al93 discovered a novel chemokine receptor on RBCs. Analysis of the repertoire of ligands bound by this novel receptor showed that it binds selected members of both the C-X-C and C-C families with high afnity (Kd 5 nM).94,95 These include interleukin-8 (IL-8) and melanoma growth stimulatory activity (MGSA) from the C-X-C family and RANTES (regulated upon activation, normally T-cell expressed) and monocyte chemotactic peptide (MCP-1) from the C-C family.94,95 The RBC chemokine receptor does not bind the C chemokine, lymphotactin.96 The multispecic erythrocyte chemokine binding activity is preserved in other mammalian species, including mice, and in avian species.11,21,96 Further studies on the multispecic chemokine receptor on human RBCs led to the observation that approximately

From bloodjournal.hematologylibrary.org by guest on June 12, 2013. For personal use only.
3082 HADLEY AND PEIPER

when detected by immunoblotting with anti-Fy6. MGSA, IL-8, and a mutant MGSA, designated MGSA-E6A, which evinces high-afnity binding only to the RBC chemokine receptor, were shown to block invasion of human RBCs by P knowlesi, a parasite known to use the Duffy antigen for invasion.97,98 MGSA was also shown to block binding of the 135-kD Duffy binding protein of P knowlesi to Duffy-positive human RBCs.97 The conclusion drawn from this evidence that the human RBC multispecic chemokine receptor and the Duffy blood group antigen are identical was subsequently conrmed by transfection experiments. K562 and 293 cells (both of which normally lack the Duffy antigen and are devoid of a multispecic chemokine receptor) were transfected with cloned Duffy cDNA and consequently displayed concomitant Duffy antigenic and multispecic chemokine receptor activity on their surfaces.99,100
DUFFY ANTIGEN RECEPTOR FOR CHEMOKINES (DARC) IS EXPRESSED BY ENDOTHELIAL CELLS OF POSTCAPILLARY VENULES

Fig 2. Inhibition of chemokines binding to the DARC by the antiDuffy MoAb, anti-Fy6. (A) Inhibition of specic 125I-labeled IL-8 binding to Duffy-positive RBCs by increasing concentrations of anti-Fy6. The closed circles represent binding of 125I-labeled IL-8 to cells expressing type A IL-8 receptor. Anti-Fy6 specically inhibits binding of IL-8 to DARC in a dose-dependent fashion. (B) Inhibition of specic binding of radiolabeled IL-8, MGSA, MCP-1, and RANTES to Duffy-positive RBCs by anti-Fy6. Anti-Fy6 inhibits the binding of both C-X-C chemokines (IL-8, MGSA) and C-C chemokines (MCP-1, RANTES) to DARC. (Reprinted with permission from Science, Horuk R, Chitnis CE, Darbonne WC, Colby TJ, Rybicki A, Hadley TJ, Miller LH: A receptor for the malarial parasite Plasmodium vivax: The erythrocyte chemokine receptor. 261:1182, 1993. Copyright 1993 American Association for the Advancement of Science.)97

two thirds of African Americans lack this receptor activity, a frequency that was intriguingly similar to that of the Duffynegative phenotype. Subsequent investigation led to the conclusion that the RBC chemokine receptor and the Duffy blood group antigen are identical.97 There was an absolute correlation between chemokine binding activity and Duffy blood group antigen expression. Anti-Fy6 specically blocked binding of chemokines to Duffy positive RBCs (Fig 2). Both the RBC chemokine receptor and the Duffy blood group antigen were destroyed by chymotrypsin treatment, but not trypsin treatment, of intact RBCs. Cross-linking experiments with 125I-labeled MGSA indicated that the RBC chemokine receptor is a 35- 43-kD protein with a similar appearance on sodium dodecyl sulfate-polyacrylamide gel electrophoresis (SDS-PAGE) as that of the Duffy antigen

The physiologic signicance of the novel multispecic RBC chemokine receptor, designated the Duffy antigen receptor for chemokines (DARC), initially appeared questionable because no pathologic or inammatory consequences have been associated with the Duffy-negative phenotype. However, immunohistochemical staining of human tissues with anti-Fy6 provided additional insight into a potential physiologic function for DARC.22,101 Specic and intense staining with anti-Fy6 was observed with endothelial cells lining postcapillary venules throughout the body. Staining was not observed on endothelial cells lining capillaries or larger vessels, including venules, veins, arterioles, and arteries.101a Littoral cells, specialized endothelial cells that line the sinusoids in the red pulp of the spleen, were also strongly positive (Fig 3) as were the endothelial cells that line the bone marrow sinusoids and choroid plexus. Parallel chemokine binding experiments conrmed the presence of a highafnity receptor having a promiscuous chemokine binding repertoire in membrane fractions from kidney and spleen.22,100 Endothelial cells lining the hepatic sinusoids lacked immunoreactivity. Tissue from individuals of the Fy(a0b0) erythroid phenotype were also examined and found to react with anti-Fy6 in a fashion identical to tissue from Duffy-positive individuals.22 Interestingly, endothelial cells of larger vessels were observed to react with anti-Fy6 under conditions of inammation (eg, temporal arteritis, thrombophlebitis, omphalitis), suggesting upregulation of DARC under these conditions.101a As noted previously, DARC-like molecules are expressed on RBCs of rodents and other mammalian and avian species.11,21,96 Because many model systems for the actions of chemokines on leukocyte trafcking, angiogenesis, and microvascular leak syndromes have been developed in rodents, experiments were performed to determine whether DARC is expressed by endothelial cells of postcapillary venules in rats. Because immunologic reagents for the rodent homolog of DARC are not yet available, intravital microscopy was performed on rats infused with uorescent microspheres covalently coated with IL-8 or MCP-1 into the cremaster ves-

From bloodjournal.hematologylibrary.org by guest on June 12, 2013. For personal use only.
DUFFY ANTIGEN RECEPTOR FOR CHEMOKINES 3083

Fig 3. Immunohistochemistry of spleen using the antiDuffy MoAb, anti-Fy6. Analysis was done on both freshly obtained and archival specimens of spleen. Anti-Fy6 reacts with specialized endothelial cells (littoral cells) lining the sinusoids in the red pulp. Endothelial cells lining arterioles, venules, arteries, and veins did not stain with anti-Fy6. Similar staining was observed with endothelial cells of postcapillary venules in every organ examined thus far. Immunohistochemical staining of endothelial cells in tissue obtained from Duffy-negative individuals (by RBC typing) was identical to that observed in tissue obtained from Duffy-positive individuals. In contrast to the sinusoids of the spleen, endothelial cells lining hepatic sinusoids did not stain.

sels.101a Monitoring of uorescent microsphere circulation revealed attachment to endothelial cells lining small venules, but not to larger vessels. This binding of IL-8coated microspheres was inhibited by previous infusion of IL-8, as well as MCP-1. Control microspheres did not adhere to endothelial cells. These experiments provide indirect functional data that the rodent homolog of DARC is expressed by subsets of endothelium according to a program that mimics that observed in humans. It is of note that the subset of endothelial cells that

express DARC is similar to the anatomic site of leukocyte trafcking and are targets for both adhesion and diapedesis.102-104 In addition, endothelial cells in this anatomic distribution are targets for vascular leak syndromes induced by pathologic actions of IL-8. Thus, in addition to expression on erythrocytes, which is not essential, DARC is expressed by endothelial cells at a dynamic interphase of leukocyte trafcking, a process that is highly regulated by the actions of chemokines that bind to DARC. Whether or not DARC actually plays a role in leukocyte trafcking remains to be determined.

Fig 6. Purkinje cells of the cerebellum express the DARC. Immunohistochemical staining of archival specimens of human cerebellum with the anti-DARC MoAb, anti-Fy6, showed highlevel expression of DARC by Purkinje neurons. This staining was inhibited by a recombinant fusion protein in which the amino terminal extracellular domain of DARC was expressed in continuous translational frame with glutathione-S-transferase. Cross-linking experiments with 125I-labeled MGSA and immunoblots with anti-Fy6 showed that MGSA and anti-Fy6 react with a protein component of cerebellar membranes with the same size and appearance on SDS-PAGE as RBC and endothelial cell DARC (data not shown).

From bloodjournal.hematologylibrary.org by guest on June 12, 2013. For personal use only.
3084 HADLEY AND PEIPER

THE MOLECULAR BIOLOGY OF DARC

Complementary DNA (cDNA) encoding DARC was molecularly cloned by Chaudhuri et al105 at the New York Blood Center in 1993. Sequences of various peptides from the Duffy glycoprotein, which were puried by immunoafnity chromatography with Fy6 MoAbs, were used to design oligonucleotide primers that were employed in DNA amplication reactions.105,106 A 72-bp probe generated by reverse transcription followed by polymerase chain reaction was used to screen a cDNA library prepared from bone marrow cells of a Duffy-positive individual. Nucleotide sequence analysis of overlapping cDNA clones revealed an open reading frame (ORF) of 1,014 bp, which predicted a protein product of 338 amino acids, with a theoretical molecular weight of 35 kD. In situ hybridization conrmed previous linkage analysis that the Duffy locus is on chromosome 1q22 r q23.107,108 The Duffy cDNA was noted to share signicant homology with the human and rabbit type B IL-8 receptor, which also binds MGSA at high afnity.105 Surprisingly, DARC also shares a similar level of homology to the multispecic receptor for endothelins, vaso-active proteins with 21 amino acid residues and two internal disulde bonds, which are also expressed by endothelial cells and have a profound effect on vascular biology. Whereas the similarity

Fig 4. Proposed seven transmembrane spanning topology of DARC. (A) Molecular modeling predicts that DARC has topologic features similar to other members of the chemokine receptor family. (Modied and reprinted with permission.113) The amino terminal extracellular domain contains the binding site for anti-Fy6 MoAbs and the polymorphism resulting in the Fya and Fyb blood groups. This domain has also been found to contain sequences necessary for multi-specic chemokine binding. Sequences required for binding a MoAb with anti-Fy3 specicity have been tentatively localized to the third predicted extracellular loop. (B) The deduced amino acid sequence of DARC; the rst seven N-terminal amino acids shown are those predicted from the exon identied by Iwamoto111 and are thought to be present in the major DARC transcript; the N-terminal amino acids shown in parentheses and the rest of the amino acid sequence are derived from the single DARC exon as originally cloned by Chaudhuri et al.105 (Modied and reprinted with permission from Horuk R: Molecular properties of the chemokine receptor family. Trends in Pharmacological Sciences, vol 15, p 159, 1994.88).

From bloodjournal.hematologylibrary.org by guest on June 12, 2013. For personal use only.
DUFFY ANTIGEN RECEPTOR FOR CHEMOKINES 3085

Fig 5. Schematic representation of the DARC genomic locus. DARC is encoded by an ORF contained in two exons separated by one intron. Primer extension and 5RACE analysis of both human DARC and mouse DARC homolog (Wang Z.X., Lu A.H., Peiper S.C., unpublished observations, September 1995) reveal the presence of an upstream exon containing a methionine translation initiation codon and codons for six additional amino acid residues. An internal initiation codon that may be used in the human gene is not conserved in the mouse gene. The polymorphism responsible for the Fya and Fyb phenotypes is encoded at codon 44.

between DARC and the IL-8RB is more prominent in the amino-terminal and carboxyl-terminal regions, its similarity to the multispecic endothelin receptor (type B) is evident in the loops (both extracellular and cytoplasmic) and transmembrane-spanning helices. Unlike virtually all other heptahelical receptors, which are almost universally linked to Gproteins, DARC lacks a highly conserved DRY motif in the second cytoplasmic loop. Although molecular modeling approaches described in the original cloning report predicted the presence of nine hydrophobic helices that could serve as transmembrane spanning domains, subsequent molecular modeling analyses predicted the presence of seven hydrophobic a-helices, analogous to receptors for all of the other known chemoattractants.11,99,109 Although DARC undergoes cotranslational addition of asparagine-linked oligosaccharide chains and posttranslational remodeling of carbohydrates in the Golgi, the predicted primary structure lacks an N-terminal series of hydrophobic amino acid residues that could function as a signal peptide. This characteristic is common to all of the cloned chemokine receptors. It is presumed that these proteins are targeted to membrane-bound polyribosomes and the Golgi apparatus via the hydrophobic helices, which are predicted to serve as transmembrane anchors. The presence of seven hydrophobic helices suggests a topology in which there are an amino-terminal extracellular domain (approximately 62 residues), three extracellular loops, three cytoplasmic loops, and a carboxly-terminal cytoplasmic tail (28 residues) (Fig 4). Alignment with the primary structures of other known chemokine receptors shows that DARC contains cysteine residues in each extracellular loop that line up with those present in the other receptors, as well as the presence of other highly conserved residues. The N-terminal extracellular domain contains at least two, and possibly three, sites for addition of oligosaccharide chains to asparagine residues. The N-terminal extracellular domain is rich in acidic amino acid residues, as might be expected for a domain involved in binding ligands with a basic isoelectric point (pI). As characteristic of other chemokines receptors, the cytoplasmic tail is composed of approximately 50% serine and threonine residues, which are presumed to be targets for phosphorylation, as has been observed for the type B IL-8 receptor. The organization of the gene encoding DARC has not yet

been completely elucidated. Using primer extension analysis, Chaudhuri et al105 found the RNA cap site to be approximately 175 bp upstream from the (presumed) translation initiation codon. Amplication of the ORF with primers from the 5 and 3 untranslated regions by polymerase chain reaction yielded DNA fragments of identical size when genomic DNA and cDNA reverse transcribed from mRNA served as the source of templates. Nucleotide sequence analysis conrmed that the ORF was contained within a single, uninterrupted exon. Constructs prepared from this ORF were capable of encoding a protein of the predicted size that bound anti-Duffy MoAbs and the predicted panel of chemokines at the appropriate afnity when expressed in K562 cells and human embryonic kidney cells.99,100 However, because the amino-terminal peptide sequence of the Duffy antigen has not been determined, it has not been conrmed that the translation initiation codon proposed by Chaudhuri et al is the predominant one used in vivo.105 Analysis by other groups provide evidence that for an mRNA cap site different from that initially reported. Using the method of rapid amplication of cDNA ends (5RACE), Tournamille et al110 reported that the mRNA initiation site was 495 bp upstream of the translation initiation codon. Similar studies described by Iwamoto et al111 localized the cap site to 0550 bp. The latter report also described a novel upstream exon that contained an alternative ATG codon. Direct comparison showed that this exon, which encodes a methionine and six additional amino acid residues, is preferentially utilized over the downstream cap site described by Chaudhuri et al. Analysis of the mouse gene encoding the homolog of DARC performed in our laboratory supports the ndings of Iwamoto et al.111 The downstream methionine residue is not conserved in the mouse gene. 5RACE of mRNA from mouse spleen conrmed the presence of an upstream exon encoding seven amino acids, led by a methionine residue. This exon, which is followed by a consensus splice donor sequence, is separated from the exon containing the majority of the ORF by an intron of approximately 450 bp. Together, these ndings suggest that both human DARC and the mouse homolog are encoded by a gene composed of two exons and one intron (Fig 5). The importance of DARC is evidenced by the conservation of this gene across species. We have cloned genes encoding DARC homologs from six nonhuman primates, cow,

From bloodjournal.hematologylibrary.org by guest on June 12, 2013. For personal use only.
3086 HADLEY AND PEIPER

pig, rabbit, and mouse. There is over 95% conservation of primary structure between the DARC homologs in humans and great apes. There is some divergence in macaques. The nucleotide sequence of the rhesus homolog predicts six amino acid differences compared with humans and a deletion of codon 24 which encodes an aspartic acid residue in the amino-terminal extracellular domain.21 Rhesus RBCs react with anti-Fyb but not with anti-Fy6. There is divergence in the predicted amino acid sequence of the murine homolog to a level of approximately 65% identity with the human receptor protein.
PRESERVATION OF DARC EXPRESSION IN ENDOTHELIAL CELLS OF DUFFY-NEGATIVE INDIVIDUALS

Despite the conservation of DARC function and primary structure across species, the fact that a large population of individuals of African ancestry lacks expression of this receptor on their RBCs without any detectable adverse consequences represented a problem for those wishing to assign physiologic signicance to DARC. Whereas Southern blotting experiments failed to show evidence of a gross rearrangement or deletion involving the DARC gene in DNA from Duffy-negative individuals, Northern blot analysis failed to detect mRNA transcripts encoding DARC in bone marrow (erythroid) cells.103 However, Northern blot analysis indicated that mRNA transcripts that annealed to a DARC probe under high stringency conditions were present in the spleen and other tissues of Duffy-negative individuals.21,22 As noted above, immunohistochemical staining revealed reactivity of anti-Fy6 with endothelial cells lining splenic sinusoids, bone marrow sinusoids, choroid plexus, and postcapillary venules in Duffy-negative as well as Duffy-positive individuals. Parallel biochemical studies demonstrated the presence of a high-afnity chemokine receptor (Kd 5 nM) with a binding specicity that included members of both CX-C and C-C families, identical to that of DARC from the spleen of a Duffy individual.22 Nucleotide sequence analysis of the ORF from these patients failed to show evidence of a mutation in the coding sequences of the gene. As was observed by Chaudhuri et al, the genotype of the Duffynegative African Americans tested was FY*B/FY*B.21 This was consistent with previous observations that Duffy-negative individuals of African descent produce anti-Fya antibodies, but not anti-Fyb.19,20 Based on the presence of an intact gene and a tissuespecic expression defect, it was postulated that the loss of expression in erythroid cells of Duffy-negative individuals could be due to a promoter/enhancer defect.20,108-112 Tournamille et al showed that Duffy-negative individuals have a point mutation in a consensus binding site for GATA-1, a transcription factor active in erythroid cells.110 This point mutation, located at 046, abolishes erythroid promoter activity in reporter gene assays.110 Further work is required to understand the molecular basis for maintenance of expression of DARC on endothelial cells and Purkinje cells, even in individuals who are Duffy negative. The immunohistochemical and molecular genetic evidence suggest an important physiologic role for DARC. Although the selective pressure from malaria led to loss of

DARC expression on RBCs (affording protection from P vivax malaria), natures experiment selected a mechanism whereby expression of DARC was preserved on endothelial cells. This evolutionary outcome suggests that the function of DARC on endothelial cells is less dispensable than its function on RBCs. Recently, the Duffy genotype of a rare Fy(a0b0) white individual was reported.113 This was the same individual (AZ) who produced anti-Fy3 described by Albrey et al.25 Analysis of the nucleotide sequence of amplication products from the DARC gene disclosed the presence of a 14-bp deletion that resulted in a frameshift mutation beginning at codon 98. AZ is apparently homozygous for this rare deletion. The mutation predicted the translation of a 118-amino acid residue polypeptide that would include the amino terminal extracellular domain, the rst transmembrane spanning a-helix, and the rst cytoplasmic loop. However, RBCs from this individual are devoid of serologically detectable Duffy antigen and presumably DARC is missing in other tissues as well. These studies were performed on blood cells that had been previously frozen, and neither current clinical information nor immunohistochemical information from nonerythroid tissue in this individual were obtainable. However, in the report by Albrey et al describing anti-Fy3 during AZs third pregnancy, there is no mention of ill health. This suggests that a DARC-nullizygous phenotype may not be associated with adverse biologic consequences. Of course, biologic redundancy or compensatory mechanisms could obscure a defect in such individuals. It is hoped that a mouse nullizygous for the DARC homolog can be produced by targeted gene disruption to further explore the function of DARC. This was part of our rationale for cloning the murine DARChomolog.
STRUCTURE-FUNCTION ANALYSIS OF DARC

Nucleotide sequence analysis of molecular clones of DARC genes from Fy(a/b0), Fy(a0b/), and Fy(a/b/) individuals in several laboratories showed that the Fya and Fyb alleles differ by a single base substitution in the second position of codon 44 (Fig 4) that encodes a glycine residue in Fya and an aspartic acid residue in Fyb.21,100,113-115 This polymorphism does not appear to have any biologic consequences. Anti-Fy6 was shown to inhibit chemokine binding to DARC in a dose-dependent fashion whereas anti-Fy3 reagent did not alter binding at concentrations up to 100 mmol/L.116 At higher concentrations of anti-Fy3, some inhibition of chemokine binding to DARC was obtained.96,117 IL-8 did not inhibit the binding of anti-Fy6 to DARC on RBCs, indicating that the IL-8 binding site and the Fy6 epitope are distinct.118 The Fy6 and Fy3 epitopes were mapped using chimeric receptor proteins composed of complementary portions of DARC and IL-8RB, the chemoattractant receptor most closely related to DARC.116 Only chimeric receptors containing the amino terminal extracellular domain of DARC reacted with anti-Fy6 MoAbs, whether stably expressed in human cell lines or in insect cells using baculovirus. The Fy6 epitope was further localized by Wasniowska et al119 using a combinatorial peptide approach and site-directed mu-

From bloodjournal.hematologylibrary.org by guest on June 12, 2013. For personal use only.
DUFFY ANTIGEN RECEPTOR FOR CHEMOKINES 3087

tagenesis. These investigators showed that two adjacent acidic amino acid residues, encoded by codons 25 (aspartic acid) and 26 (glutamic acid), are critical for anti-Fy6 binding, whereas the asparagine residues encoded by codons 18 and 29 are not involved (Fig 4). Furthermore, since mutation of these two residues eliminates two sites for the addition of oligosaccharide chains, it is likely that the Fy6 epitope is composed solely of peptide residues. The epitope recognized by anti-Fy3 has also been mapped, although not to the same degree of molecular detail as Fy6.116 Immunouorescent analysis of insect cells expressing chimeric receptors showed that the anti-Fy3 MoAb bound to constructs containing the three predicted extracellular loops of DARC, but not to constructs containing only the amino terminal extracellular domain.119 Anti-Fy3 did not bind to receptor chimeras that included the N-terminal extracellular domain and the rst and second predicted extracellular loops. It was concluded that sequences necessary for the binding of anti-Fy3 are present in the third predicted extracellular loop of DARC (Fig 4). Parallel studies localized the regions of the receptor involved in the binding of chemokines and the malarial Duffy binding ligands to this receptor.119 Analysis of a chimera expressing the N-terminal extracellular domain of DARC with the remainder of the heptahelical structure being that of IL-8RB (DARCe1/IL-RB) showed that it bound not only IL-8 and MGSA at high afnity, but also RANTES, a C-C chemokine that binds to DARC, but not IL-8RB. Moreover, the DARCe1/ IL-8RB chimera bound the DARC-specic MGSA-E6A mutant with a Kd of 12 nM, mirroring the activity of intact DARC. Preliminary studies suggest that the 140-kD Duffy binding protein of P vivax also binds to the DARCe1/IL-8RB chimera (C. Chitnis, L.H. Miller, S. Peiper, personal communication, June 1995). Taken together, these ndings highlight the signicance of the amino terminal extracellular domain in the various biologic activities of DARC. Recent studies using DARC variants created by in situ mutagenesis indicate chemokine binding to DARC is dependent on the disulde bond between a cysteine on the N-terminal extracellular domain and domain and a cysteine on the third extracellular loop.117
DARC IS EXPRESSED BY PURKINJE CELLS OF THE CEREBELLUM

one of the predicted size, 1.2 kb, which was also detected in many other tissues tested; and one of 8.5 kb, which thus far has only been observed in brain. Although the nature of the 8.5-kb transcript has not yet been elucidated, the ndings of DARC transcripts in the brain prompted our investigation of brain by immunohistochemistry.11,123 Anti-Fy6 was found to react specically with Purkinje neurons of the cerebellum (Fig 6, see page 3083). Parallel experiments were done with an MoAb against IL-8 receptor B (IL-8RB) which reacted specically with subsets of neurons in diverse regions of the brain and spinal cord.123 These included the hippocampus, dentate nucleus, pontine nuclei, locus coeruleus, and paraventricular nucleus in the brain and the anterior horn, interomediolateral cell column, and Clarkes column of the spinal cord. Like Purkinje neurons of the cerebellum that express DARC, the neurons in other areas of the brain and spinal cord that express the IL-8RB are projection neurons, bearing long axons that connect one neuronal region with another. Interneurons, which connect neurons within a region, did not react with either anti-Fy6 or the antiIL-8RB antibody. Further experiments were performed to conrm that antiFy6 reactivity with Purkinje cells was indeed caused by the presence of DARC. Membranes were isolated from cerebellar tissue at postmortem examination and these were analyzed for the presence of DARC by chemokine binding assays, chemokine cross-linking experiments, and immunblotting. Results obtained by each of these methods supported the immunohistochemical data and indicated that DARC is indeed expressed on Purkinje neurons of the cerebellum. What function DARC serves on Purkinje neurons and how this relates to its expression on RBCs and on endothelial cells remains to be determined.
THE RIDDLE OF DARC FUNCTION

Emerging evidence suggests that chemokines are involved in normal homeostatic processes in the central nervous system. Experiments using in situ hybridization localized rat mRNA that hybridizes to a human IL-8 probe in specic areas of the brain, including hippocampus and cerebellum.120 Human IL-8 has also been shown to enhance survival of rat hippocampal neurons in vitro.121 It is thought that IL-8 is synthesized and secreted by astrocytes.122 There are approximately 10-fold more astrocytes in the central nervous system than neurons and one of the functions of astrocytes is to nurture neurons by the secretion of neurotrophic factors. Until recently there were no reports on neurons expressing chemokine receptors. Chaudhuri et al105 demonstrated the presence of mRNA in human brain that specically hybridized with DARC cDNA. Interestingly, there were two species of mRNA observed:

Despite knowledge of structure-function relationships and tissue localization of DARC, the precise role of this receptor in normal and pathologic physiology remains uncertain. DARC does not appear to present chemokines in an active form to leukocyte receptors. Once bound to DARC expressed on the surface of erythrocytes, IL-8 did not induce effects in neutrophils associated with chemokine stimulation (K. Neote, S. Peiper, unpublished observations, September 1994).93 Human erythroleukemia (HEL) cells express DARC, but its function on these cells has not been elucidated.124 Although DARC on RBCs does not internalize ligand, there is evidence that DARC-transfectants can internalize ligands.22 It is possible that endothelial DARC internalizes ligands, thereby generating the chemotactic gradient essential for leukocyte attraction. Based on what is known of other heptahelical membrane receptors, it seems likely that DARC transmits a signal across the membrane upon chemokine binding. However, DARC-dependent signal transduction has not yet been demonstrated. DARC does not appear to be coupled to a guanosine triphosphate binding protein (G-protein). It does not stimulate GTPase activity nor mediate calcium ux upon ligand binding; furthermore, DARC lacks a DRY motif in the second cytoplasmic loop that is characteristic of G-protein coupled seven-membranespanning receptors.

From bloodjournal.hematologylibrary.org by guest on June 12, 2013. For personal use only.
3088 HADLEY AND PEIPER

To date, only a few other heptahelical receptors that lack DRY motifs have been cloned. They include the cAMP receptor in Dictostelium, bride of Sevenless (bos ) in drosophila, and the two seven-transmembranespanning proteins that harbor mutations in familial cases of Alzheimers disease.125-128 The yeast homolog of the heptahelical protein mutated in Alzheimers disease has been shown to be involved in signaling via the notch pathway.129 Thus, there is precedent for heptahelical receptors that signal through pathways independent of G-coupled proteins. It is possible that DARC interacts or cross-talks with other membrane components (which could differ depending on cell type) and thereby elicit tissue specic responses to ligand binding.
SUMMARY

cells of glomeruli, capillaries, vasa recta, and epithelial cells of collecting tubules in the kidney; reactivity was also observed with capillaries in the thyroid, and capillaries, large venules, and type I alveolar squamous cells of lung. Whether or not the carbohydrate epitope recognized by polyclonal rabbit antibody 6615 is also present on molecules other than the Duffy antigen remains to be determined.
ACKNOWLEDGMENT

The glycoprotein expressed on surface of erythrocytes initially known as the Duffy blood group antigen has been shown to be a receptor for the invasion of these cells by P vivax parasites. The parasite ligand that binds to the Duffy antigen has now been cloned and characterized. The region on the Duffy binding ligand of the parasite responsible for interaction with the Duffy antigen has also been identied. It is hoped that this molecule will useful as an immunogen to induce antibodies capable of blocking invasion of the parasite into the erythrocyte. Research on the Duffy binding ligand also provided a clue (a shared motif) to the identication of the long-sought family of variant endothelial binding ligands that mediate attachment of P falciparum infected erythrocytes to endothelial cells of postcapillary venules. If this attachment could be disrupted during the course of a malarial infection, it might lead to spleen-mediated parasite death. It might also lead to amelioration of cerebral malaria, a condition which in part is due to the blockade of cerebral venules by sequestered adherent parasites. Recently, research on the Duffy antigen has taken on a new dimension. The Duffy antigen has been shown to be a multispecic heptahelical receptor for chemokines, expressed on RBCs, endothelial cells of postcapillary venules, and Purkinje cells of the cerebellum. The challenge now is to determine its function, both in immunobiology and neurobiology. Its capacity to bind chemoattractant cytokines and its expression on endothelial cells lining postcapillary venules are highly conserved across species, suggesting that this receptor subserves a critical function. This is supported by natures experiment, the Duffy blood group negative phenotype, in which the genetic mechanism selected to remove expression on erythroid cells to protect against malarial infection, preserved expression on endothelial cells of postcapillary venules and splenic sinusoids. Although we have signicant insight into structure-function relationships for the Duffy antigen/receptor for chemokines, its mechanism of signaling and its biologic function remain to be elucidated.
NOTE ADDED IN PROOF

We acknowledge the investigators in our laboratories including Zi-xuan Wang, Zhaohai Lu, and Haihong Guo. Collaborators at the University of Louisville include Alvin Martin and Victor Fingar and Stephen Slone. We also acknowledge Richard Horuk (Berlex, Richmond, CA), who has been a major collaborator, and Domanique Blanchard (Centre Regional de Transfusion Sanguine, Nantes, France) and Makoto Uchikawa (Japanese Red Cross Central Blood Center, Tokyo, Japan) for providing anti-Fy6 and anti-Fy3, respectively. Anti-Fy3 was obtained through Peter Byrne at the National Reference Laboratory (Rockville, MD). We also thank Beverly Kirkpatrick and Abby Carden for assistance in preparing the manuscript for this review.
REFERENCES

Chaudhuri et al (Blood 89:701, 1997) recently described results of immunohistochemical staining with a polyclonal rabbit antibody (6615) that reacts with carbohydrate on the Duffy antigen. Reactivity was observed with endothelial

1. Landsteiner K: Individual differences in human blood. Science 73:405, 1931 2. Levine P, Burnham L, Katzin EM, Vogel P: Role of iso-immunization in pathogenesis of erythroblastosis fetalis. Am J Obstet Gynecol 42:925, 1941 3. Anstee DJ: Blood group-active surface molecules of the human red blood cell. Vox Sang 58:1, 1990 4. Issitt PD, Issitt CH: Applied Blood Group Serology. Miami, FL, Montgomery Scientic, 1985 5. Anstee DJ: Blood group antigens dened by the amino acid sequences of red cell surface proteins. Transf Med 5:1, 1995 6. Agre PC, Catron J-P (eds): Blood Group Antigens of the Human Red Cell. Structure, Function, and Clinical Signicance. Baltimore, MD, Johns Hopkins University Press, 1992 7. Cartron, J-P, Rouger P (eds): Molecular Basis of Major Blood Group Antigens, Blood Cell Biochemistry, vol 8. New York, NY, Plenum, 1995 8. Telen MJ: Erythrocyte blood group antigens: Not so simple after all. Blood 85:299, 1995 9. Barnwell JW, Galinski MR: Plasmodium vivax: A glimpse into the unique and shared biology of the merozoite. Ann Trop Med Parasitol 89:113, 1995 10. Pogo AO, Chaudhuri A: Duffy and receptors for P. vivax and chemokine peptides. Transf Clin Biol 2:269, 1995 11. Horuk R, Peiper SC: The Duffy antigen receptor for chemokines, in Horuk R (ed): Chemoattractant Ligands and Their Receptors. New York, NY, CRC, 1996 12. Horuk R, Martin A, Hesselgesser J, Hadley T, Lu ZH, Wang ZX, Peiper SC: The Duffy antigen receptor for chemokines: Structural analysis and expression in the brain. J Leukoc Biol 59:29, 1996 13. Cutbush M, Mollison PL, Parkin DM: A new human blood group. Nature 165:188, 1950 14. Ikin EW, Mourant AE, Pettenkoffer JH, Blumenthal G: Discovery of the expected haemagglutinin anti-Fyb. Nature 168:1077, 1951 15. Chown B, Lewis M, Kaita H: The Duffy blood group in caucasians: Evidence for a new allele. Am J Hum Genet 17:384, 1965 16. Lewis M, Kaita H, Chown B: The Duffy blood group system in caucasians: A further population sample. Vox Sang 23:523, 1972 17. Mourant AE, Kopec AC, Domaniewska-Sobczak K: The Dis-

From bloodjournal.hematologylibrary.org by guest on June 12, 2013. For personal use only.
DUFFY ANTIGEN RECEPTOR FOR CHEMOKINES 3089

tribution of Human Blood Groups and Other Polymorphisms (ed 2). London, UK, Oxford University Press, 1976 18. Sanger R, Race RR, Jack J: The Duffy blood groups of New York Negroes: The phenotype Fy(a0b0). Br J Haematol 1:370, 1955 19. Vengelen-Tyler V: Anti-Fya preceding anti-Fy3 or -Fy5: A study of ve cases. Transfusion Abstracts S150, 1985 (suppl) 20. Le Pennec PY, Rouger P, Klein MT, Robert N, Salmon C: Study of anti-Fya in ve black Fy(a0b0) patients. Vox Sang 52:246, 1987 21. Chaudhuri A, Polyakova J, Zbrzezna V, Pogo O: The coding sequence of Duffy blood group gene in humans and simians: Restriction fragment length polymorphism, antibody and malarial parasite specicities and expression in nonerythroid tissues in Duffy negative individuals. Blood 85:615, 1995 22. Peiper SC, Wang ZX, Neote K, Martin AW, Showell HJ, Conklyn MJ, Ogborne K, Hadley TJ, Lu ZH, Hesselgesser J, Horuk R: The Duffy antigen/receptor for chemokines (DARC) is expressed in endothelial cells of Duffy negative individuals who lack the erythrocyte receptor. J Exp Med 181:1311, 1995 23. Morton JA: Some observations on the action of blood-group antibodies on red cells treated with proteolytic enzymes. Br J Haematol 8:134, 1962 24. Judson PA, Anstee DJ: Comparative effect of trypsin and chymotrypsin on blood group antigens. Med Lab Sci 34:1, 1977 25. Albrey JA, Vincent EE Jr, Hutchinson J, Marsh WL, Allen FH, Gavin J, Sanger R: A new antibody, anti-Fy3, in the Duffy blood group system. Vox Sang 20:29, 1971 26. Behzad O, Lee CL, Gavin J, Marsh WL: A new anti-erythrocyte antibody in the Duffy system: Anti-Fy4. Vox Sang 24:337, 1973 27. Colledge KI, Pezzulich M, Marsh WL: Anti-Fy5: An antibody disclosing a probable association between the Rhesus and Duffy glood group genes. Vox Sang 24:193, 1973 28. Moore S, Woodrow CF, McClelland DB: Isolation of membrane components associated with human red cell antigens Rh(D), (c), (E) and Fy. Nature 295:529, 1982 29. Hadley TJ, David PH, McGinniss MH, Miller LH: Identication of an erythrocyte component carrying the Duffy blood group Fya antigen. Science 223:597, 1984 30. Tanner MJ, Anstee DJ, Mallison G, Ridgewell K, Martin PG, Avent ND, Parsons SF: Effect of endoglycosidase F-peptidyl Nglycosidase F preparations on the surface components of the human erythrocyte. Carbohydrate Res 178:203, 1988 31. Chaudhuri A, Pogo OA: The Duffy is a glycoprotein, in Cartron J-P, Rouger P (eds): Molecular Basis of Major Human Blood Group Antigens, Blood Cell Biochemistry, vol 8. New York, NY, Plenum, 1995 32. Wasniowska K, Eichenberger P, Kugele F, Hadley TJ: Purication of a 28 kD non-aggregating tryptic peptide of the Duffy blood group protein. Biochem Biophys Res Commun 192:366, 1993 33. Wasniowska K, Hadley TJ: The Duffy blood group antigen: An update. Transf Med Rev 8:281, 1994 34. Nichols ME, Rubinstein P, Barnwell J, Rodriguez de Cordoba S, Roseneld RE: A new human Duffy blood group specicity dened by a murine monoclonal antibody. Immunogenetics and association with susceptibility to Plasmodium vivax. J Exp Med 166:776, 1987 35. Barnwell JW, Nichols ME, Rubenstein P: In vitro evaluation of the role of the Duffy blood group in erythrocyte invasion by Plasmodium vivax. J Exp Med 169:162, 1989 36. Riwom S, Janvier D, Navenot JM, Benbunan M, Muller JY, Blanchard D: Production of a new murine monoclonal antibody with Fy6 spcicity and characterization of the immunopuried Nglycosylated Duffy-active molecule. Vox Sang 66:61, 1994 37. Miller LH, Mason SJ, Dvorak JA, McGinniss MH, Rothman IK: Erythrocyte receptors for (Plasmodium knowlesi ) malaria: Duffy blood group determinants. Science 189:561, 1975 38. Mason SJ, Miller LH, Shiroishi T, Dvorak JA, McGinniss

MH: The Duffy blood group determinants: Their role in the susceptibility of human and animal erythrocytes to Plasmodium knowlesi malaria. Br J Haematol 36:327, 1977 39. Miller LH, Aikawa M, Johnson JG, Shiroishi T: Interaction between cytochalasin B-treated malarial parasites and erythrocytes. Attachment and junction formation. J Exp Med 149:172, 1979 40. Aikawa M, Miller LH, Johnson J, Rabbege J: Erythrocyte entry by malarial parasites: A moving junction between erythrocyte and parasite. J Cell Biol 77:72, 1978 41. Young MD, Eyles DE, Burgess RW, Jeffrey GM: Experimental testing of the immunity of Negroes to Plasmodium vivax. J Parasitol 41:315, 1955 42. Miller LH, Mason SJ, Clyde DF, McGinniss MH: The resistance factor to Plsmodium vivax in blacks: The Duffy blood group genotype, FyFy. N Engl J Med 295:302, 1976 43. Miller LH, McGinniss MH, Holland PV, Sigmon P: The Duffy blood group phenotype in American blacks with Plasmodium vivax in Vietnam. Am J Trop Med Hyg 27:1069, 1978 44. Spencer HC, Miller LH, Collins WE, Knud-Hansen C, McGinnis MH, Shiroishi T, Lobos RA, Feldman RA: The Duffy blood group and resistance to Plasmodium vivax in Honduras. Am J Trop Med Hyg 27:664, 1978 45. Kitchen SF: The infection of reticulocytes by Plasmodium vivax. Am J Trop Med 18:347, 1938 46. Galinski MR, Medina CC, Ingravallo P, Barnwell JW: A reticulocyte-binding protein complex of Plasmodium vivax merozoites. Cell 69:1213, 1992 47. Haynes JD, Dalton JP, Klotz FW, McGinnis MH, Hadley TJ, Hudson DE, Miller LH: Receptor-like specicity of a Plasmodium knowlesi malarial protein that binds to Duffy antigen ligands on erythrocytes. J Exp Med 167:1873, 1988 48. Wertheimer SP, Barnwell JW: Plasmodium vivax interaction with the human Duffy blood group glycoprotein: Identication of a parasite receptor-like protein. Exp Parasitol 69:340, 1989 49. Sim BKL, Chitnis CE, Wasniowska K, Hadley TJ, Miller LH: Receptor and ligand domains for invasion of erythrocytes by Plasmodium falciparum malaria. Science 264:1941, 1994 50. Adams JH, Sim BK, Dolan SA, Fang X, Kaslow DC, Miller LH: A family of erythrocyte binding proteins of malaria parasites. Proc Natl Acad Sci USA 89:7085, 1992 51. Adams JH, Hudson DE, Torii M, Ward GE, Wellems TE, Aikawa M, Miller LH: The Duffy receptor family of Plasmodium knowlesi is located within the micronemes of invasive Malaria merozoites. Cell 63:141, 1990 52. Fang XD, Kaslow DC, Adams JH, Miller LH: Cloning of the Plasmodium vivax Duffy receptor. Mol Biochem Parasitol 44:125, 1992 53. Chitnis CE, Miller LH: Identication of the erythrocyte binding domains of Plasmodium vivax and Plasmodium knowlesi proteins involved in erythrocyte invasion. J Exp Med 180:497, 1994 54. Chitnis CE, Chaudhuri A, Horuk R, Pogo O, Miller LH: The domain on the Duffy blood group antigen for binding Plasmodium vivax and P. knowlesi malarial parasites to erythroytes. J Exp Med 184:1531, 1996 55. Peterson DS, Miller LH, Wellems TE: Isolation of multiple sequences from the Plasmodium falciparum genome that encode conserved domains homologous to those in erythrocyte-binding proteins. Proc Natl Acad Sci USA 92:7100, 1995 56. Su XZ, Heatwole VM, Wertheimer SP, Guinet F, Herrfeldt JA, Peterson DS, Ravetch JA, Wellems TE. The large diverse gene family var encodes proteins involved in cytoadherence and antigenic variation of Plasmodium falciparum-infected erythrocytes. Cell 82:89, 1995 57. Smith JD, Chitnis CE, Craig AG, Roberts DJ, Hudson-Taylor DE, Peterson DS, Pinches R, Newbold CI, Miller LH: Switches in expression of Plamodium falciparum var genes correlate with

From bloodjournal.hematologylibrary.org by guest on June 12, 2013. For personal use only.
3090 HADLEY AND PEIPER

changes in antigenic and cytoadherent phenotypes of infected erythrocytes. Cell 82:101, 1995 58. Baruch DI, Pasloske BL, Singh HB, Bi X, Ma XC, Feldman M, Taraschi TF, Howard RJ: Cloning the P. falciparum gene encoding PfEMP1, a malarial variant antigen and adherence receptor on the surface of parasitized human erythrocytes. Cell 82:77, 1995 59. Leech JH, Barnwell JW, Miller LH, Howard RJ: Identication of a strain-specic malarial antigen exposed on the surface of Plasmodium falciparum-infected erythrocytes. J Exp Med 159:1567, 1984 60. Howard RJ, Barnwell JW, Rock EP, Neequaye J, Ofori-Adjei D, Maloy WL, Lyon JA, Saul A: Two approximately 300 kilodalton Plasmodium falciparum proteins at the surface membrane of infected erythrocytes. Mol Biochem Parasitol 27:207, 1988 61. van Shravendijk MR, Rock EP, Marsh K, Ito Y, Aikawa M, Neequaye J, Ofori-Adjei D, Rodriquez R, Patarroyo ME, Howard RJ: Characterization and localization of Plasmodium falciparum surface antigens on incated erythrocytes from West Africa. Blood 78:226, 1991 62. Udeinya IJ, Schmidt JA, Aikawa M, Miller LH, Green I: Falciparum malaria-infected erythrocytes specically bind to cultured human endothelial cells Science 213:555, 1981 63. Pasloske BL, Howard RJ: Malaria, the red cell and the endothelium. Annu Rev Med 45:283, 1994 64. Raventos-Suarez C, Kaul DK, Macaluso F, Nagel RL: Membrane knobs are required for the microcirculatory obstruction induced by Plasmodium falciparum-infected erythrocytes. Proc Natl Acad Sci USA 82:3829, 1985 65. Pongponratn E, Riganti M, Punpoowong B, Aikawa M: Microvascular sequestration of parasitized erythrocytes in human falciparum malaria: A pathologic study. Am J Trop Med Hyg 44:168, 1991 66. Luse SA, Miller LH: Plasmodium falciparum malaria: Ultrastructure of parasitized erythrocytes in cardiac vessels. Am J Trop Med Hyg 20:655, 1971 67. Langreth SG, Peterson E: Pathogenicity, stability and immunogenicity of a knobless clone of Plasmodium falciparum in Columbian owl monkeys. Infect Immun 47:760, 1985 68. Oster CN, Koontz LC, Wyler DJ: Malaria in asplenic mice: Effects of splenectomy, congenital asplenia, and splenic reconstitution on the course of infection. Am J Trop Med Hyg 29:1138, 1980 69. Howard RJ, Gilladoga AD: Molecular studies related to the pathogenesis of cerebral malaria. Blood 74:2603, 1989 70. David PH, Hommel M, Miller LH, Udeinya IJ, Oligino LD: Parasite sequestration in Plasmodium falciparum malaria: Spleen and antibody modulation of cytoadherence of infected erythrocytes. Proc Natl Acad Sci USA 80:5075, 1983 71. Udeinya IJ, Miller LH, McGregor IA, Jensen JB: Plasmodium falciparum strain-specic antibody blocks binding of infected erythrocytes to amelanotic melanoma cells. Nature 303:429, 1983 72. Marsh K, Howard RJ: Antigens induced on erythrocytes by P. falciparum: Expression of diverse and conserved determinants. Science 231:150 1986 73. Magowan C, Wollish W, Anderson L, Leech J: Cytoadherence by Plasmodium falciparum-infected erythrocytes is correlated with the expression of a family of variable proteins on infected erythrocytes. J Exp Med 168:1307, 1988 74. Roberts DJ, Craig AG, Berendt AR, Pinches R, Nash G, Marsh K, Newbold CI: Rapid switching to multiple antigenic and adhesive phenotypes in malaria. Nature 357:689, 1992 75. Biggs BP, Anders RF, Dillon HE, Davern KM, Martin M, Peterson C, Brown GV: Adherence of infected erythrocytes to venular endothelium selects for antigenic variants of Plasmodium falciparum. J Immunol 149:2047, 1992 76. Ockenhouse CF, Ho M, Tandon NN, Van Seventer GA, Shaw S, White NJ, Jamieson GA, Chulay JD, Webster HK: Molecular basis of sequestration in severe and uncomplicated Plasmodium fal-

ciparum malaria: Differential adhesion of infected erythrocytes to CD36 and ICAM-1. J Infect Dis 164:163, 1991 77. Ockenhouse CF, Tegoshi T, Maeno Y, Benjamin C, Ho M, Kan KE, Thway Y, Win K Aikawa M, Lobb RR: Human vascular endothelial cell adhesion receptors for Plasmodium-infected erythrocytes: Roles for endothelial leukocyte adhesion molecule 1 and vascular cell adhesion molecule 1. J Exp Med 176:1183, 1992 78. Roberts DD, Sherwood JA, Spitalnik SL, Panton LJ, Howard RJ, Dixit VM, Frazier WA, Miller LH, Ginsburg V: Thrombospondin binds falciparum malaria parasitized erythrocytes and may mediate cytoadherence. Nature 318:64, 1985 79. Miller LH, Good MF, Milon G: Malaria pathogenesis. Science 264:1878, 1994 80. Dolan SA, Miller LH, Wellems TE: Evidence for a switching mechanism in the invasion of erythrocytes by Plasmodium falciparum. J Clin Invest 86:618, 1990 81. Tsuboi T, Kappe SH, al-Yaman F, Prickett MD, Alpers M, Adams JH: Natural variation within the principal adhesion domain of the Plasmodium vivax Duffy binding protein. Infect Immun 62:5581, 1994 82. Oppenheim JJ, Zachariae CO, Mukaida N, Matsushima K: Properties of the novel proinammatory supergene intercrine cytokine family. Annu Rev Immunol 9:617, 1991 83. Baggiolini M, Dewald B, Moser B: Interleukin-8 and related chemotactic cytokinesCXC and CC chemokines. Adv Immunol 55:97, 1994 84. Schall T: The chemokines, in Thomson AW (ed): The Cytokine Handbook (ed 2). New York, NY, Academic, 1994, p 419 85. Schall TJ: Biology of the RANTES/SIS cytokine family. Cytokine 3:165, 1991 86. Kelner GS, Kennedy J, Bacon KB, Kleyensteuber S, Largaespada DA, Jenkins NA, Copeland NG, Bazan JF, Moore KW, Schall TJ, Zlotnik A: Lymphotactin: A cytokine that represents a new class of chemokine. Science 266:1395, 1994 87. Murphy PM: The molecular biology of leukocyte chemoattractant receptors. Annu Rev Immunol 12:593, 1994 88. Horuk R: Molecular properties of the chemokine receptor family. Trends Pharmacol Sci 15:159, 1994 89. Horuk R: Cytokine receptors, in Peroutka SJ (ed): Handbook of Receptors and Channels: G Protein-Coupled Receptors. Boca Raton, FL, CRC, 1993, p 87 90. Neote K, DiGregorio D, Mak JY, Horuk R, Schall TJ: Molecular cloning, functional expression, and signaling characteristics of a C-C chemokine receptor. Cell 72:415, 1993 91. Holmes WE, Lee J, Kuang WJ, Rice GC, Wood WI: Structure and functional expression of a human interleukin-8 receptor. Science 253:1278, 1991 92. Hebert CA, Chuntharapai A, Smith M, Colby T, Kim J, Horuk R: Partial functional mapping of the human interleukin-8 type A receptor. Identication of a major ligand binding domain. J Biol Chem 268:18549, 1993 93. Darbonne WC, Rice GC, Mohler MA, Apple T, Hegert CA, Valente AJ, Baker JB: Red blood cells are a sink for interleukin 8, a leukocyte chemotaxin. J Clin Invest 88:1362, 1991 94. Neote K, Darbonne W, Ogez J, Horuk R, Schall TJ: Identication of a promiscuous inammatory peptide receptor on the surface of red blood cells. J Biol Chem 268:12247, 1993 95. Horuk R, Colby TJ, Darbonne WC, Schall TJ, Neote K: The human erythrocyte inammatory peptide (chemokine) receptor. Biochemical characterization, solubilization, and development of a binding assay for the soluble receptor. Biochemistry 32:5733, 1993 96. Szabo MC, Soo KS, Zlotnik A, Schall TJ: Chemokine class differences in binding to the Duffy antigen/erythrocyte receptor. J Biol Chem 270:25348, 1995 97. Horuk R, Chitnis CE, Darbonne WC, Colby TJ, Rybicki A, Hadley TJ, Miller LH: A receptor for the malarial parasite Plasmo-

From bloodjournal.hematologylibrary.org by guest on June 12, 2013. For personal use only.
DUFFY ANTIGEN RECEPTOR FOR CHEMOKINES 3091

dium vivax: The erythrocyte chemokine receptor. Science 261:1182, 1993 98. Hesselgesser J, Chitnis CE, Miller LH, Yansura DG, Simmons LC, Fairbrother WJ, Kotts C, Wirth C, Gillece-Castro BL, Horuk R: A mutant of melanoma growth stimulating activity does not activate neutrophils but blocks erythrocyte invasion by malaria. J Biol Chem 270:11472, 1995 99. Chaudhuri A, Zbrzezna V, Polykova J, Pogo AO, Hesselgesser J, Horuk R: Expression of the Duffy antigen in K562 cells. Evidence that it is the human erythrocyte chemokine receptor. J Biol Chem 269:7835, 1994 100. Neote K, Mak JY, Kolakowski LF, Schall TJ: Functional and biochemical analysis of the cloned Duffy antigen: Identity with the red blood cell chemokine receptor. Blood 84:44, 1994 101. Hadley TJ, Lu Z-h, Wasniowska K, Martin AW, Peiper SC, Hesselgesser J, Horuk R: Postcapillary venule endothelial cells in kidney express a multispecc chemokine receptor that is structurally and functionally identical to the erythroid isoform which is the Duffy blood group antigen. J Clin Invest 94:985, 1994 101a. Guo H, Slone S, Fingar V, Blanchard D, Peiper S, Hadley T: Tissue distribution and upregulation by inammation. Blood 88:513a, 1996 (abstr, suppl 1) 102. Rot A: Endothelial cell binding of NAP-1/IL-8: Role in neutrophil emigration. Immunol Today 13:291, 1992 103. Hechtman DH, Cybulsky MI, Fuchs HJ, Baker JB, Gimbrone MA: Intravascular IL-8: Inhibitor of polymorphonuclear leukocyte accumulation at sites of acute inammation. J Immunol 147:883, 1991 104. Lawrence LB, Springer TA: Leukocytes roll on a selectin at physiologic ow rates: Distinction from and prerequisite for adhesion through integrins. Cell 65:859, 1991 105. Chaudhuri A, Polyakova J, Zbrezna V, Williams K, Gulati S, Pogo AO: Cloning of glycoprotein D cDNA which encodes the major subunit of the Duffy blood group system and the receptor for the Plasmodium vivax malaria parasite. Proc Natl Acad Sci USA 90:10793, 1993 106. Chaudhuri A, Zbrzezna V, Johnson C, Nichols M, Rubinstein P, Marsh WL, Pogo AO: Purication and characterization of an erythrocyte membrane protein complex carrying Duffy blood group antigenicity. Possible receptor for plasmodium vivax and plasmodium Knowlesi malaria parasite. J Biol Chem 264:13770, 1989 107. Jacobs PA, Brunton M, Fackiewicz A, Newton M, Cook PJ, Robson EB: Studies on a family with three cytogenetic markers. Ann Hum Genet 33:325, 1970 108. Mathew S, Chaudhuri A, Murty VV, Pogo AO: Conrmation of Duffy blood group antigen locus (FY) at 1q22 r q23 by ourescence in situ hybridization. Cytogenet Cell Genet 67:68, 1994 109. Eisenberg D: Three-dimensional structure of membrane and surface proteins. Annu Rev Biochem 53:595, 1984 110. Tournamille C, Colin Y, Cartron JP, Le Van Kim C: Disruption of a GATA motif in the Duffy gene promoter abolishes erythroid gene expression in Duffy-negative individuals. Nature Genet 10:224, 1995 111. Iwamoto S, Li J, Omi T, Ikemoto S, Kajii E: Identication of a novel exon and spliced form of Duffy mRNA that is the predominant transcript in both erythroid and postcapillary endothelium. Blood 87:378, 1996 112. Iwamoto S, Li J, Sugimoto N, Okuda H, Kajii E: Characterization of the Duffy gene promoter: Evidence for tissue specic abolishment of expression in Fy(a0b0) of black individuals. Biochem Biophys Res Commun 222:852, 1996 113. Mallinson G, Soo KS, Schall TJ, Pisacka M, Anstee DJ: Mutations in the erythrocyte chemokine receptor (Duffy) gene: The molecular basis of the Fya/Fyb antigens and identication of a deletion in the Duffy gene of an apparently healthy individual with the Fy(a0b0) phenotype. Br J Haematol 90:823, 1995

114. Iwamoto S, Omi T, Kajii E, Ikemoto S: Genomic organization of the glycoprotein D gene: Duffy blood group Fya/Fyb alloantigen system is associated with a polymorphism at the 44-amino acid residue. Blood 85:662, 1995 115. Tournamille C, Le Van Kim C, Gane P, Cartron JP, Colin Y: Molecular basis and PCR-DNA typing of the Fya/Fyb blood group polymorphism. Hum Genet 95:407, 1995 116. Lu Z-h, Wang Z-x, Horuk R, Hesselgesser J, Lou YC, Hadley TJ, Peiper SC: The promiscuous chemokine binding prole of the Duffy antigen/recptor for chemokines (DARC) is primarily localized to sequences in the amino terminal domain. J Biol Chem 270:26239, 1995 117. Tournamille C, Le Van Kim C, Gane P, Proudfoot A, Cartron JP, Colin Y: Epitopes of the erythrocyte receptor for chemokines (Duffy glycoprotein) involved in IL8 binding (abstr). Trans Clin Biol 3:54s, 1996 (suppl) 118. Hausman E, Dzik W, Blanchard D: The red cell chemokine receptor is distinct from the Fy6 epitope. Transfusion 36:421, 1996 119. Wasniowska K, Blanchard D, Jauvier D, Wang ZX, Peiper SC, Hadley TJ, Lisowska E: Identication of the Fy6 epitope recognized by two monoclonal antibodies in the N-terminal extracellular portion of the Duffy antigen receptor for chemokines. Mol Immunol 33:917, 1996 120. Rothwell NJ, Hardwick AJ, Lindley I: Central actions of interleukin-8 in the rat are independent of prostaglandins. Horm Metab Res 22:595, 1990 121. Araujo DM, Cotman CW: Trophic effects of interleukin-4, -7 and -8 on hippocampal neuronal cultures: Potential involvement of glial-derived factors. Brain Res 600:49, 1993 122. Van Meir E, Ceska M, Effenberger F, Walz A, Grouzmann E, Desbaillets I, Frei K, Fontana A, de Tribolet N: Interleukin-8 is produced in neoplastic and infectious diseases of the human central nervous system. Cancer Res 52:4297, 1992 123. Horuk R, Martin AW, Wang ZY, Schweitzer L, Gerassimides A, Guo H, Lu ZH, Hesselgesser J, Perez HD, Kim J, Parker J, Hadley TJ, Peiper SC: Expression of chemokine receptors by subset of neurons in the central nervous system. J Immunol 158:2882, 1997 124. Horuk R, Wang ZX, Peiper SC, Hesselgesser J: Identication and characterization of a promiscuous chemokine binding protein in a human erythroleukemia cell line. J Biol Chem 269:17730, 1994 125. Klein PS, Sun TJ, Saxe CL, Kimmel AR, Johnson RL, Devreotes PN: A chemoattractant receptor controls development in Dictyostelium discoideum. Science 241:1467, 1988 126. Hart AC, Kramer H, Van Vactor DL, Paidhungat M, Zipursky SL: Induction of cell fate in the Drosophila retina: The bride of sevenless protein is predicted to contain a large extracellular domain and seven transmembrane segments. Genes Dev 4:1835, 1990 127. Rogaev EI, Sherrington R, Rogaeva EA, Levesque G, Ikeda M, Liang Y, Chi H, Lin C, Holman K, Tsuda T, Mar L, Sorbi S, Nacmias B, Placentini S, Amaducci L, Chumakov I, Cohen D, Lannfelt L, Fraser PE, Rommens JM, St George-Hyslop PH: Familial Alheimers disease in kindreds with missense mutations in a gene on chromosome 1 related to the Alzhelmers disease type 3 gene. Nature 376:775, 1995 128. Sherrington R, Rogaev EI, Liang Y, Rogaeva EA, Levesque G, Ikeda M, Chi H, Lin C, Li G, Holman K, Tsuda T, Mar L, Foncin J-F, Bruni AC, Montesi, M.P., Sorbi S, Rainero I, Pinessi L, Nee L, Chumakov I, Pollen D, Brookes A, Sanseau P, Pollinsky R J, Wasco W, Da Silva HAR, Haines JL, Pericak-Vance MA, Tanzi, RE, Roses AD, Fraser PE, Rommens JM, St George-Hyslop PH: Cloning of a gene bearing missense mutations in early onset familial Alzheimers disease. Nature 375:754, 1995 129. Levitan D, Greenwald I: Facilitation of lin-12-mediated signalling be sel-12, a Caenorhabditis elegans S182 Alzheimers disease gene. Nature 377:351, 1995

Você também pode gostar