164
Views
0
CrossRef citations to date
0
Altmetric
Review

The role of the human Duffy antigen receptor for chemokines in malaria susceptibility: current opinions and future treatment prospects

, , &
Pages 1-11 | Published online: 26 Sep 2016

References

  • Cutbush M, Mollison PL. The Duffy blood group system. Heredity (Edinb). 1950;4(3):383–389.
  • Miller LH, Mason SJ, Clyde DF, McGinniss MH. The resistance factor to Plasmodium vivax in blacks. The Duffy-blood-group genotype, FyFy. N Engl J Med. 1976;295(6):302–304.
  • Miller LH, Mason SJ, Dvorak JA, McGinniss MH, Rothman IK. Erythrocyte receptors for (Plasmodium knowlesi) malaria: Duffy blood group determinants. Science. 1975;189(4202):561–563.
  • Chaudhuri A, Polyakova J, Zbrzezna 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 U S A. 1993;90(22):10793–10797.
  • He W, Neil S, Kulkarni H, et al. Duffy antigen receptor for chemokines mediates trans-infection of HIV-1 from red blood cells to target cells and affects HIV-AIDS susceptibility. Cell Host Microbe. 2008;4(1):52–62.
  • de Brevern AG, Wong H, Tournamille C, Colin Y, Le Van Kim C, Etchebest C. A structural model of a seven-transmembrane helix receptor: the Duffy antigen/receptor for chemokine (DARC). Biochim Biophys Acta. 2005;1724(3):288–306.
  • 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. 1995;85(3):622–626.
  • Behzad O, Lee CL, Gavin J, Marsh WL. A new anti-erythrocyte antibody in the Duffy system: anti-Fy4. Vox Sang. 1973;24(4):337–342.
  • Colledge KI, Pezzulich M, Marsh WL. Anti-Fy5, and antibody disclosing a probable association between the Rhesus and Duffy blood group genes. Vox Sang. 1973;24(3):193–199.
  • 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. Nat Genet. 1995;10(2):224–228.
  • Marsh WL, Ehrich CC. The Duffy blood group system: a review of recent developments. Infusionsther Klin Ernahr. 1975;2(4):280–289.
  • Meny GM. The Duffy blood group system: a review. Immunohematology. 2010;26(2):51–56.
  • Sanger R, Race RR, Jack J. The Duffy blood groups of New York negroes: the phenotype Fy (a-b-). Br J Haematol. 1955;1(4):370–374.
  • Parasol N, Reid M, Rios M, Castilho L, Harari I, Kosower NS. A novel mutation in the coding sequence of the FY*B allele of the Duffy chemokine receptor gene is associated with an altered erythrocyte phenotype. Blood. 1998;92(7):2237–2243.
  • Horuk R, Chitnis CE, Darbonne WC, et al. A receptor for the malarial parasite Plasmodium vivax: the erythrocyte chemokine receptor. Science. 1993;261(5125):1182–1184.
  • Neote K, Mak JY, Kolakowski LF Jr, Schall TJ. Functional and biochemical analysis of the cloned Duffy antigen: identity with the red blood cell chemokine receptor. Blood. 1994;84(1):44–52.
  • Rot A, von Andrian UH. Chemokines in innate and adaptive host defense: basic chemokinese grammar for immune cells. Annu Rev Immunol. 2004;22:891–928.
  • Demogines A, Truong KA, Sawyer SL. Species-specific features of DARC, the primate receptor for Plasmodium vivax and Plasmodium knowlesi. Mol Biol Evol. 2012;29(2):445–449.
  • Horuk R, Colby TJ, Darbonne WC, Schall TJ, Neote K. The human erythrocyte inflammatory peptide (chemokine) receptor. Biochemical characterization, solubilization, and development of a binding assay for the soluble receptor. Biochemistry. 1993;32(22):5733–5738.
  • Darbonne WC, Rice GC, Mohler MA, et al. Red blood cells are a sink for interleukin 8, a leukocyte chemotaxin. J Clin Invest. 1991;88(4):1362–1369.
  • Waśniowska K, Czerwinski M, Jachymek W, Lisowska E. Expression and binding properties of a soluble chimeric protein containing the N-terminal domain of the Duffy antigen. Biochem Biophys Res Commun. 2000;273(2):705–711.
  • McMorran BJ, Wieczorski L, Drysdale KE, et al. Platelet factor 4 and Duffy antigen required for platelet killing of Plasmodium falciparum. Science. 2012;338(6112):1348–1351.
  • Tournamille C, Filipe A, Wasniowska K, et al. Structure-function analysis of the extracellular domains of the Duffy antigen/receptor for chemokines: characterization of antibody and chemokine binding sites. Br J Haematol. 2003;122(6):1014–1023.
  • Peiper SC, Wang ZX, Neote K, et al. The Duffy antigen/receptor for chemokines (DARC) is expressed in endothelial cells of Duffy negative individuals who lack the erythrocyte receptor. J Exp Med. 1995;181(4):1311–1317.
  • Horuk R, Martin A, Hesselgesser J, et al. The Duffy antigen receptor for chemokines: structural analysis and expression in the brain. J Leukoc Biol. 1996;59(1):29–38.
  • Hadley TJ, Lu ZH, Wasniowska K, et al. Postcapillary venule endothelial cells in kidney express a multispecific chemokine receptor that is structurally and functionally identical to the erythroid isoform, which is the Duffy blood group antigen. J Clin Invest. 1994;94(3):985–991.
  • Lee JS, Frevert CW, Wurfel MM, et al. Duffy antigen facilitates movement of chemokine across the endothelium in vitro and promotes neutrophil transmigration in vitro and in vivo. J Immunol. 2003;170(10):5244–5251.
  • Williams MR, Azcutia V, Newton G, Alcaide P, Luscinskas FW. Emerging mechanisms of neutrophil recruitment across endothelium. Trends Immunol. 2011;32(10):461–469.
  • Pruenster M, Mudde L, Bombosi P, et al. The Duffy antigen receptor for chemokines transports chemokines and supports their promigratory activity. Nat Immunol. 2009;10(1):101–108.
  • Horne K, Woolley IJ. Shedding light on DARC: the role of the Duffy antigen/receptor for chemokines in inflammation, infection and malignancy. Inflamm Res. 2009;58(8):431–435.
  • Moore BB, Arenberg DA, Stoy K, et al. Distinct CXC chemokines mediate tumorigenicity of prostate cancer cells. Am J Pathol. 1999;154(5):1503–1512.
  • Shen H, Schuster R, Stringer KF, Waltz SE, Lentsch AB. The Duffy antigen/receptor for chemokines (DARC) regulates prostate tumor growth. FASEB J. 2006;20(1):59–64.
  • Lentsch AB. The Duffy antigen/receptor for chemokines (DARC) and prostate cancer. A role as clear as black and white? FASEB J. 2002;16(9):1093–1095.
  • Nemesure B, Wu SY, Hennis A, Leske MC. Distribution of Duffy antigen receptor for chemokines (DARC) and risk of prostate cancer in Barbados, West Indies. J Immigr Minor Health. 2015;17(3):679–683.
  • Zijlstra A, Quigley JP. The DARC side of metastasis: shining a light on KAI1-mediated metastasis suppression in the vascular tunnel. Cancer Cell. 2006;10(3):177–178.
  • Wang J, Ou ZL, Hou YF, et al. Enhanced expression of Duffy antigen receptor for chemokines by breast cancer cells attenuates growth and metastasis potential. Oncogene. 2006;25(54):7201–7211.
  • Mayr FB, Spiel AO, Leitner JM, et al. Racial differences in endotoxin-induced tissue factor-triggered coagulation. J Thromb Haemost. 2009;7(4):634–640.
  • Watorek E, Boratynska M, Halon A, Klinger M. Anti-Fya antibodies as the cause of an unfortunate post-transplant course in renal transplant recipient. Ann Transplant. 2008;13(1):48–52.
  • Afenyi-Annan A, Kail M, Combs MR, Orringer EP, Ashley-Koch A, Telen MJ. Lack of Duffy antigen expression is associated with organ damage in patients with sickle cell disease. Transfusion. 2008;48(5):917–924.
  • Nebor D, Durpes MC, Mougenel D, et al. Association between Duffy antigen receptor for chemokines expression and levels of inflammation markers in sickle cell anemia patients. Clin Immunol. 2010;136(1):116–122.
  • Kulkarni H, Marconi VC, He W, et al. The Duffy-null state is associated with a survival advantage in leukopenic HIV-infected persons of African ancestry. Blood. 2009;114(13):2783–2792.
  • Reich D, Nalls MA, Kao WH, et al. Reduced neutrophil count in people of African descent is due to a regulatory variant in the Duffy antigen receptor for chemokines gene. PLoS Genet. 2009;5(1):e1000360.
  • Walton RT, Rowland–Jones SL. HIV and chemokine binding to red blood cells–DARC matters. Cell Host Microbe. 2008;4(1):3–5.
  • Vergara C, Tsai YJ, Grant AV, et al. Gene encoding Duffy antigen/receptor for chemokines is associated with asthma and IgE in three populations. Am J Respir Crit Care Med. 2008;178(10):1017–1022.
  • Woolley IJ, Hutchinson P, Reeder JC, Kazura JW, Cortes A. Southeast Asian ovalocytosis is associated with increased expression of Duffy antigen receptor for chemokines (DARC). Immunohematology. 2009;25(2):63–66.
  • Geleff S, Draganovici D, Jaksch P, Segerer S. The role of chemokine receptors in acute lung allograft rejection. Eur Respir J. 2010;35(1):167–175.
  • Barnwell JW, Nichols ME, Rubinstein P. In vitro evaluation of the role of the Duffy blood group in erythrocyte invasion by Plasmodium vivax. J Exp Med. 1989;169(5):1795–1802.
  • Wertheimer SP, Barnwell JW. Plasmodium vivax interaction with the human Duffy blood group glycoprotein: identification of a parasite receptor-like protein. Exp Parasitol. 1989;69(4):340–350.
  • Xu L, Niu J, Chaudhuri A. The domain on the mouse Duffy protein for Plasmodium yoelii binding and invasion to mouse erythrocytes. Mol Biochem Parasitol. 2006;146(2):142–150.
  • Swardson-Olver CJ, Dawson TC, Burnett RC, Peiper SC, Maeda N, Avery AC. Plasmodium yoelii uses the murine Duffy antigen receptor for chemokines as a receptor for normocyte invasion and an alternative receptor for reticulocyte invasion. Blood. 2002;99(8):2677–2684.
  • Cox-Singh J, Singh B. Knowlesi malaria: newly emergent and of public health importance? Trends Parasitol. 2008;24(9):406–410.
  • Price RN, Tjitra E, Guerra CA, Yeung S, White NJ, Anstey NM. Vivax malaria: neglected and not benign. Am J Trop Med Hyg. 2007;77(Suppl 6):79–87.
  • Mendis K, Sina BJ, Marchesini P, Carter R. The neglected burden of Plasmodium vivax malaria. Am J Trop Med Hyg. 2001;64(Suppl 1–2):97–106.
  • Galinski MR, Barnwell JW. Plasmodium vivax: who cares? Malar J. 2008;7(Suppl 1):S9.
  • Kochar DK, Das A, Kochar SK, et al. Severe Plasmodium vivax malaria: a report on serial cases from Bikaner in northwestern India. Am J Trop Med Hyg. 2009;80(2):194–198.
  • Sharma A, Khanduri U. How benign is benign tertian malaria? J Vector Borne Dis. 2009;46(2):141–144.
  • Alexandre MA, Ferreira CO, Siqueira AM, et al. Severe Plasmodium vivax malaria, Brazilian Amazon. Emerg Infect Dis. 2010;16(10):1611–1614.
  • Rijken MJ, Boel ME, Russell B, et al. Chloroquine resistant vivax malaria in a pregnant woman on the western border of Thailand. Malar J. 2011;10:113.
  • Ketema T, Bacha K, Birhanu T, Petros B. Chloroquine-resistant Plasmodium vivax malaria in Serbo town, Jimma zone, south-west Ethiopia. Malar J. 2009;8:177.
  • Mohan K, Maithani MM. Congenital malaria due to chloroquine-resistant Plasmodium vivax: a case report. J Trop Pediatr. 2010;56(6):454–455.
  • Cowman AF, Crabb BS. Invasion of red blood cells by malaria parasites. Cell. 2006;124(4):755–766.
  • Adams JH, Hudson DE, Torii M, et al. The Duffy receptor family of Plasmodium knowlesi is located within the micronemes of invasive malaria merozoites. Cell. 1990;63(1):141–153.
  • 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 U S A. 1992;89(15):7085–7089.
  • Guerra CA, Howes RE, Patil AP, et al. The international limits and population at risk of Plasmodium vivax transmission in 2009. PLoS Negl Trop Dis. 2010;4(8):e774.
  • Miller LH, McGinniss MH, Holland PV, Sigmon P. The Duffy blood group phenotype in American blacks infected with Plasmodium vivax in Vietnam. Am J Trop Med Hyg. 1978;27(6):1069–1072.
  • King CL, Adams JH, Xianli J, et al. Fy(a)/Fy(b) antigen polymorphism in human erythrocyte Duffy antigen affects susceptibility to Plasmodium vivax malaria. Proc Natl Acad Sci U S A. 2011;108(50):20113–20118.
  • Michon P, Woolley I, Wood EM, Kastens W, Zimmerman PA, Adams JH. Duffy-null promoter heterozygosity reduces DARC expression and abrogates adhesion of the P. vivax ligand required for blood-stage infection. FEBS Lett. 2001;495(1–2):111–114.
  • Howes RE, Patil AP, Piel FB, et al. The global distribution of the Duffy blood group. Nat Commun. 2011;2:266.
  • Ryan JR, Stoute JA, Amon J, et al. Evidence for transmission of Plasmodium vivax among a Duffy antigen negative population in Western Kenya. Am J Trop Med Hyg. 2006;75(4):575–581.
  • Menard D, Barnadas C, Bouchier C, et al. Plasmodium vivax clinical malaria is commonly observed in Duffy-negative Malagasy people. Proc Natl Acad Sci U S A. 2010;107(13):5967–5971.
  • Cavasini CE, Mattos LC, Couto AA, et al. Plasmodium vivax infection among Duffy antigen-negative individuals from the Brazilian Amazon region: an exception? Trans R Soc Trop Med Hyg. 2007;101(10):1042–1044.
  • Wurtz N, Mint Lekweiry K, Bogreau H, et al. Vivax malaria in Mauritania includes infection of a Duffy-negative individual. Malar J. 2011;10:336.
  • Fru-Cho J, Bumah VV, Safeukui I, Nkuo-Akenji T, Titanji VP, Haldar K. Molecular typing reveals substantial Plasmodium vivax infection in asymptomatic adults in a rural area of Cameroon. Malar J. 2014;13:170.
  • Hester J, Chan ER, Menard D, et al. De novo assembly of a field isolate genome reveals novel Plasmodium vivax erythrocyte invasion genes. PLoS Negl Trop Dis. 2013;7(12):e2569.
  • Menard D, Chan ER, Benedet C, et al. Whole genome sequencing of field isolates reveals a common duplication of the Duffy binding protein gene in Malagasy Plasmodium vivax strains. PLoS Negl Trop Dis. 2013;7(11):e2489.
  • Gunalan K, Lo E, Hostetler JB, et al. Role of Plasmodium vivax Duffy-binding protein 1 in invasion of Duffy-null Africans. Proc Natl Acad Sci U S A. 2016;113(22):6271–6276.
  • VanBuskirk KM, Sevova E, Adams JH. Conserved residues in the Plasmodium vivax Duffy-binding protein ligand domain are critical for erythrocyte receptor recognition. Proc Natl Acad Sci U S A. 2004;101(44):15754–15759.
  • Ranjan A, Chitnis CE. Mapping regions containing binding residues within functional domains of Plasmodium vivax and Plasmodium knowlesi erythrocyte-binding proteins. Proc Natl Acad Sci U S A. 1999;96(24):14067–14072.
  • Chitnis CE, Miller LH. Identification of the erythrocyte binding domains of Plasmodium vivax and Plasmodium knowlesi proteins involved in erythrocyte invasion. J Exp Med. 1994;180(2):497–506.
  • Hans D, Pattnaik P, Bhattacharyya A, et al. Mapping binding residues in the Plasmodium vivax domain that binds Duffy antigen during red cell invasion. Mol Microbiol. 2005;55(5):1423–1434.
  • Chitnis CE, Chaudhuri A, Horuk R, Pogo AO, Miller LH. The domain on the Duffy blood group antigen for binding Plasmodium vivax and P. knowlesi malarial parasites to erythrocytes. J Exp Med. 1996;184(4):1531–1536.
  • Choe H, Moore MJ, Owens CM, et al. Sulphated tyrosines mediate association of chemokines and Plasmodium vivax Duffy binding protein with the Duffy antigen/receptor for chemokines (DARC). Mol Microbiol. 2005;55(5):1413–1422.
  • Batchelor JD, Malpede BM, Omattage NS, DeKoster GT, Henzler-Wildman KA, Tolia NH. Red blood cell invasion by Plasmodium vivax: structural basis for DBP engagement of DARC. PLoS Pathog. 2014;10(1):e1003869.
  • Farzan M, Mirzabekov T, Kolchinsky P, et al. Tyrosine sulfation of the amino terminus of CCR5 facilitates HIV-1 entry. Cell. 1999;96(5):667–676.
  • Cimbro R, Gallant TR, Dolan MA, et al. Tyrosine sulfation in the second variable loop (V2) of HIV-1 gp120 stabilizes V2-V3 interaction and modulates neutralization sensitivity. Proc Natl Acad Sci U S A. 2014;111(8):3152–3157.
  • Farzan M, Vasilieva N, Schnitzler CE, et al. A tyrosine-sulfated peptide based on the N terminus of CCR5 interacts with a CD4-enhanced epitope of the HIV-1 gp120 envelope glycoprotein and inhibits HIV-1 entry. J Biol Chem. 2000;275(43):33516–33521.
  • Batchelor JD, Zahm JA, Tolia NH. Dimerization of Plasmodium vivax DBP is induced upon receptor binding and drives recognition of DARC. Nat Struct Mol Biol. 2011;18(8):908–914.
  • Tolia NH, Enemark EJ, Sim BK, Joshua-Tor L. Structural basis for the EBA-175 erythrocyte invasion pathway of the malaria parasite Plasmodium falciparum. Cell. 2005;122(2):183–193.
  • Srivastava A, Gangnard S, Round A, et al. Full-length extracellular region of the var2CSA variant of PfEMP1 is required for specific, high-affinity binding to CSA. Proc Natl Acad Sci U S A. 2010;107(11):4884–4889.
  • Rucker J, Edinger AL, Sharron M, et al. Utilization of chemokine receptors, orphan receptors, and herpesvirus-encoded receptors by diverse human and simian immunodeficiency viruses. J Virol. 1997;71(12):8999–9007.
  • Horuk R. The interleukin-8-receptor family: from chemokines to malaria. Immunol Today. 1994;15(4):169–174.
  • Berger EA, Murphy PM, Farber JM. Chemokine receptors as HIV-1 coreceptors: roles in viral entry, tropism, and disease. Annu Rev Immunol. 1999;17:657–700.
  • Michon P, Fraser T, Adams JH. Naturally acquired and vaccine-elicited antibodies block erythrocyte cytoadherence of the Plasmodium vivax Duffy binding protein. Infect Immun. 2000;68(6):3164–3171.
  • Grimberg BT, Udomsangpetch R, Xainli J, et al. Plasmodium vivax invasion of human erythrocytes inhibited by antibodies directed against the Duffy binding protein. PLoS Med. 2007;4(12):e337.
  • Ceravolo IP, Sanchez BA, Sousa TN, et al. Naturally acquired inhibitory antibodies to Plasmodium vivax Duffy binding protein are short-lived and allele-specific following a single malaria infection. Clin Exp Immunol. 2009;156(3):502–510.
  • King CL, Michon P, Shakri AR, et al. Naturally acquired Duffy-binding protein-specific binding inhibitory antibodies confer protection from blood-stage Plasmodium vivax infection. Proc Natl Acad Sci U S A. 2008;105(24):8363–8368.
  • Chootong P, Ntumngia FB, VanBuskirk KM, et al. Mapping epitopes of the Plasmodium vivax Duffy binding protein with naturally acquired inhibitory antibodies. Infect Immun. 2010;78(3):1089–1095.
  • Kocken CH, Dubbeld MA, Van Der Wel A, et al. High-level expression of Plasmodium vivax apical membrane antigen 1 (AMA-1) in Pichia pastoris: strong immunogenicity in Macaca mulatta immunized with P. vivax AMA-1 and adjuvant SBAS2. Infect Immun. 1999;67(1):43–49.
  • Singh S, Pandey K, Chattopadhayay R, et al. Biochemical, biophysical, and functional characterization of bacterially expressed and refolded receptor binding domain of Plasmodium vivax Duffy-binding protein. J Biol Chem. 2001;276(20):17111–17116.
  • Xainli J, Cole-Tobian JL, Baisor M, et al. Epitope-specific humoral immunity to Plasmodium vivax Duffy binding protein. Infect Immun. 2003;71(5):2508–2515.
  • Galinski MR, Medina CC, Ingravallo P, Barnwell JW. A reticulocyte-binding protein complex of Plasmodium vivax merozoites. Cell. 1992;69(7):1213–1226.
  • Galinski MR, Xu M, Barnwell JW. Plasmodium vivax reticulocyte binding protein-2 (PvRBP-2) shares structural features with PvRBP-1 and the Plasmodium yoelii 235 kDa rhoptry protein family. Mol Biochem Parasitol. 2000;108(2):257–262.
  • Galinski MR, Ingravallo P, Corredor-Medina C, Al-Khedery B, Povoa M, Barnwell JW. Plasmodium vivax merozoite surface proteins-3beta and-3gamma share structural similarities with P. vivax merozoite surface protein-3alpha and define a new gene family. Mol Biochem Parasitol. 2001;115(1):41–53.
  • Lima-Junior JC, Tran TM, Meyer EV, et al. Naturally acquired humoral and cellular immune responses to Plasmodium vivax merozoite surface protein 9 in Northwestern Amazon individuals. Vaccine. 2008;26(51):6645–6654.
  • Dutta S, Ware LA, Barbosa A, Ockenhouse CF, Lanar DE. Purification, characterization, and immunogenicity of a disulfide cross-linked Plasmodium vivax vaccine candidate antigen, merozoite surface protein 1, expressed in Escherichia coli. Infect Immun. 2001;69(9):5464–5470.
  • Arevalo-Herrera M, Chitnis C, Herrera S. Current status of Plasmodium vivax vaccine. Hum Vaccin. 2010;6(1):124–132.
  • Valencia SH, Rodriguez DC, Acero DL, Ocampo V, Arevalo-Herrera M. Platform for Plasmodium vivax vaccine discovery and development. Mem Inst Oswaldo Cruz. 2011;106(Suppl 1):179–192.
  • Ntumngia FB, Adams JH. Design and immunogenicity of a novel synthetic antigen based on the ligand domain of the Plasmodium vivax Duffy binding protein. Clin Vaccine Immunol. 2012;19(1):30–36.
  • Ntumngia FB, Schloegel J, McHenry AM, et al. Immunogenicity of single versus mixed allele vaccines of Plasmodium vivax Duffy binding protein region II. Vaccine. 2013;31(40):4382–4388.
  • Cole-Tobian JL, Michon P, Biasor M, et al. Strain-specific Duffy binding protein antibodies correlate with protection against infection with homologous compared to heterologous Plasmodium vivax strains in Papua New Guinean children. Infect Immun. 2009;77(9):4009–4017.
  • Chen E, Salinas ND, Ntumngia FB, Adams JH, Tolia NH. Structural analysis of the synthetic Duffy binding protein (DBP) antigen DEKnull relevant for Plasmodium vivax malaria vaccine design. PLoS Negl Trop Dis. 2015;9(3):e0003644.
  • Dean M, Carrington M, Winkler C, et al. Genetic restriction of HIV-1 infection and progression to AIDS by a deletion allele of the CKR5 structural gene. Hemophilia Growth and Development Study, Multicenter AIDS Cohort Study, Multicenter Hemophilia Cohort Study, San Francisco City Cohort, ALIVE Study. Science. 1996;273(5283):1856–1862.
  • Cong L, Ran FA, Cox D, et al. Multiplex genome engineering using CRISPR/Cas systems. Science. 2013;339(6121):819–823.
  • Hu W, Kaminski R, Yang F, et al. RNA-directed gene editing specifically eradicates latent and prevents new HIV-1 infection. Proc Natl Acad Sci U S A. 2014;111(31):11461–11466.
  • Alic N, Hoddinott MP, Foley A, Slack C, Piper MD, Partridge L. Detrimental effects of RNAi: a cautionary note on its use in Drosophila ageing studies. PLoS One. 2012;7(9):e45367.
  • Dragic T, Trkola A, Thompson DA, et al. A binding pocket for a small molecule inhibitor of HIV-1 entry within the transmembrane helices of CCR5. Proc Natl Acad Sci U S A. 2000;97(10):5639–5644.
  • Baba M, Nishimura O, Kanzaki N, et al. A small-molecule, nonpeptide CCR5 antagonist with highly potent and selective anti-HIV-1 activity. Proc Natl Acad Sci U S A. 1999;96(10):5698–5703.
  • Perez EE, Wang J, Miller JC, et al. Establishment of HIV-1 resistance in CD4+ T cells by genome editing using zinc-finger nucleases. Nat Biotechnol. 2008;26(7):808–816.
  • Wilen CB, Wang J, Tilton JC, et al. Engineering HIV-resistant human CD4+ T cells with CXCR4-specific zinc-finger nucleases. PLoS Pathog. 2011;7(4):e1002020.
  • Kajumbula H, Byarugaba W, Wayengera M. Targeting wild-type erythrocyte receptors for Plasmodium falciparum and vivax merozoites by zinc finger nucleases in- silico: towards a genetic vaccine against malaria. Genet Vaccines Ther. 2012;10(1):8.
  • Mussolino C, Morbitzer R, Lutge F, Dannemann N, Lahaye T, Cathomen T. A novel TALE nuclease scaffold enables high genome editing activity in combination with low toxicity. Nucleic Acids Res. 2011;39(21):9283–9293.
  • Clark-Lewis I, Schumacher C, Baggiolini M, Moser B. Structure-activity relationships of interleukin-8 determined using chemically synthesized analogs. Critical role of NH2-terminal residues and evidence for uncoupling of neutrophil chemotaxis, exocytosis, and receptor binding activities. J Biol Chem. 1991;266(34):23128–23134.
  • Gong JH, Clark-Lewis I. Antagonists of monocyte chemoattractant protein 1 identified by modification of functionally critical NH2-terminal residues. J Exp Med. 1995;181(2):631–640.
  • Proudfoot AE, Power CA, Hoogewerf AJ, et al. Extension of recombinant human RANTES by the retention of the initiating methionine produces a potent antagonist. J Biol Chem. 1996;271(5):2599–2603.
  • Horuk R. Chemokine receptor antagonists: overcoming developmental hurdles. Nat Rev Drug Discov. 2009;8(1):23–33.
  • Dorr P, Westby M, Dobbs S, et al. Maraviroc (UK-427,857), a potent, orally bioavailable, and selective small-molecule inhibitor of chemokine receptor CCR5 with broad-spectrum anti-human immunodeficiency virus type 1 activity. Antimicrob Agents Chemother. 2005;49(11):4721–4732.
  • Ng HP, May K, Bauman JG, et al. Discovery of novel non-peptide CCR1 receptor antagonists. J Med Chem. 1999;42(22):4680–4694.
  • Liang M, Mallari C, Rosser M, et al. Identification and characterization of a potent, selective, and orally active antagonist of the CC chemokine receptor-1. J Biol Chem. 2000;275(25):19000–19008.
  • Rodriguez-Frade JM, Vila-Coro AJ, de Ana AM, Albar JP, Martinez-A C, Mellado M. The chemokine monocyte chemoattractant protein-1 induces functional responses through dimerization of its receptor CCR2. Proc Natl Acad Sci U S A. 1999;96(7):3628–3633.
  • Rodriguez-Frade JM, Vila-Coro AJ, Martin A, et al. Similarities and differences in RANTES- and (AOP)-RANTES-triggered signals: implications for chemotaxis. J Cell Biol. 1999;144(4):755–765.
  • Russell B, Suwanarusk R, Borlon C, et al. A reliable ex vivo invasion assay of human reticulocytes by Plasmodium vivax. Blood. 2011;118(13):e74–e81.
  • Tran TM, Moreno A, Yazdani SS, Chitnis CE, Barnwell JW, Galinski MR. Detection of a Plasmodium vivax erythrocyte binding protein by flow cytometry. Cytometry A. 2005;63(1):59–66.
  • Hesselgesser J, Chitnis CE, Miller LH, et al. A mutant of melanoma growth stimulating activity does not activate neutrophils but blocks erythrocyte invasion by malaria. J Biol Chem. 1995;270(19):11472–11476.
  • Chitnis CE. Molecular insights into receptors used by malaria parasites for erythrocyte invasion. Curr Opin Hematol. 2001;8(2):85–91.
  • Chitnis CE, Sharma A. Targeting the Plasmodium vivax Duffy-binding protein. Trends Parasitol. 2008;24(1):29–34.
  • Schwartz TW, Rosenkilde MM. Is there a ‘lock’ for all agonist ‘keys’ in 7TM receptors? Trends Pharmacol Sci. 1996;17(6):213–216.
  • Handel TM, Horuk R. Duffy antigen inhibitors: useful therapeutics for malaria? Trends Parasitol. 2010;26(7):329–333.