1887

Abstract

Chemokines are important in the immune response against viral infections, and may play a role in human T-lymphotropic virus 1 (HTLV-1)-associated myelopathy/tropical spastic paraparesis (HAM/TSP) pathogenesis. Polymorphisms in the Duffy antigen receptor for chemokines (DARC), such as rs12075 (A>G; ) and rs281477 (−46T>C; GATA-1 box) may influence circulating concentrations of proinflammatory chemokines. We investigate whether Duffy genotypes influence the HTLV-1 proviral load (PVL) level, HTLV-1 infection outcome and chemokine concentrations in HTLV-1 asymptomatic carriers (AC=162), HAM/TSP patients (HAM=135) and seronegative individuals (SN=71).

Quantification of plasmatic IL8, CCL2 and CCL5 were performed by flow cytometry and Duffy genotypes were investigated by real-time PCR. HTLV-1 PVL was quantified in peripheral blood. To control for spurious association, individual ancestry profiles in AC and HAM groups were investigated.

PVL and IL8 level were significantly higher in the HAM group than in the AC group, but were not associated with Duffy genotypes. The highest CCL2 and CCL5 levels were seen in the SN group, and there was no difference when comparing the infected groups. The level of CCL5 was not associated with Duffy genotypes. The polymorphism −46 C/C that abrogates the DARC expression on the erythrocytes was significantly associated with lower levels of CCL2, neutrophil and white blood cell (WBC) counts in HTLV-1-infected individuals.

We conclude that although the Duffy null genotype was associated with leukopenia, neutropenia and lower levels of CCL2, the data do not suggest the influence of Duffy genotypes on the neurologic outcome of HTLV-1 infection, but may be a confounding factor in comparison HTLV-1-infected populations with different ancestries, especially when defining inflammatory biomarkers.

Keyword(s): chemokines , DARC , Duffy , HAM/TSP , HTLV-1 and proviral load
Loading

Article metrics loading...

/content/journal/jmm/10.1099/jmm.0.000539
2017-08-01
2024-04-19
Loading full text...

Full text loading...

/deliver/fulltext/jmm/66/8/1207.html?itemId=/content/journal/jmm/10.1099/jmm.0.000539&mimeType=html&fmt=ahah

References

  1. Uchiyama T. Human T cell leukemia virus type I (HTLV-I) and human diseases. Annu Rev Immunol 1997; 15:15–37 [View Article][PubMed]
    [Google Scholar]
  2. Bangham CR. The immune response to HTLV-I. Curr Opin Immunol 2000; 12:397–402 [View Article][PubMed]
    [Google Scholar]
  3. Bangham CR. HTLV-1 infections. J Clin Pathol 2000; 53:581–586 [View Article][PubMed]
    [Google Scholar]
  4. Nagai M, Usuku K, Matsumoto W, Kodama D, Takenouchi N et al. Analysis of HTLV-I proviral load in 202 HAM/TSP patients and 243 asymptomatic HTLV-I carriers: high proviral load strongly predisposes to HAM/TSP. J Neurovirol 1998; 4:586–593 [View Article][PubMed]
    [Google Scholar]
  5. Olindo S, Lézin A, Cabre P, Merle H, Saint-Vil M et al. HTLV-1 proviral load in peripheral blood mononuclear cells quantified in 100 HAM/TSP patients: a marker of disease progression. J Neurol Sci 2005; 237:53–59 [View Article][PubMed]
    [Google Scholar]
  6. Lezin A, Olindo S, Oliere S, Varrin-Doyer M, Marlin R et al. Human T lymphotropic virus type I (HTLV-I) proviral load in cerebrospinal fluid: a new criterion for the diagnosis of HTLV-I-associated myelopathy/tropical spastic paraparesis?. J Infect Dis 2005; 191:1830–1834 [View Article][PubMed]
    [Google Scholar]
  7. Furtado MS, Andrade RG, Romanelli LC, Ribeiro MA, Ribas JG et al. Monitoring the HTLV-1 proviral load in the peripheral blood of asymptomatic carriers and patients with HTLV-associated myelopathy/tropical spastic paraparesis from a Brazilian cohort: ROC curve analysis to establish the threshold for risk disease. J Med Virol 2012; 84:664–671 [View Article][PubMed]
    [Google Scholar]
  8. Zlotnik A, Yoshie O. Chemokines: a new classification system and their role in immunity. Immunity 2000; 12:121–127[PubMed] [CrossRef]
    [Google Scholar]
  9. Guerreiro JB, Santos SB, Morgan DJ, Porto AF, Muniz AL et al. Levels of serum chemokines discriminate clinical myelopathy associated with human T lymphotropic virus type 1 (HTLV-1)/tropical spastic paraparesis (HAM/TSP) disease from HTLV-1 carrier state. Clin Exp Immunol 2006; 145:296–301 [View Article][PubMed]
    [Google Scholar]
  10. Sato T, Coler-Reilly A, Utsunomiya A, Araya N, Yagishita N et al. CSF CXCL10, CXCL9, and neopterin as candidate prognostic biomarkers for HTLV-1-associated myelopathy/tropical spastic paraparesis. PLoS Negl Trop Dis 2013; 7:e2479 [View Article][PubMed]
    [Google Scholar]
  11. Neote K, Darbonne W, Ogez J, Horuk R, Schall TJ. Identification of a promiscuous inflammatory peptide receptor on the surface of red blood cells. J Biol Chem 1993; 268:12247–12249[PubMed]
    [Google Scholar]
  12. Hadley TJ, Peiper SC. From malaria to chemokine receptor: the emerging physiologic role of the duffy blood group antigen. Blood 1997; 89:3077–3091[PubMed]
    [Google Scholar]
  13. Chaudhuri A, Polyakova J, Zbrzezna V, Williams K, Gulati S et al. 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 1993; 90:10793–10797 [View Article][PubMed]
    [Google Scholar]
  14. Meny GM. The Duffy blood group system: a review. Immunohematology 2010; 26:51–56[PubMed]
    [Google Scholar]
  15. Howes RE, Patil AP, Piel FB, Nyangiri OA, Kabaria CW et al. The global distribution of the Duffy blood group. Nat Commun 2011; 2:266 [View Article][PubMed]
    [Google Scholar]
  16. 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; 10:224–228 [View Article][PubMed]
    [Google Scholar]
  17. Iwamoto S, Li J, Sugimoto N, Okuda H, Kajii E. Characterization of the Duffy gene promoter: evidence for tissue-specific abolishment of expression in Fy(a-b-) of black individuals. Biochem Biophys Res Commun 1996; 222:852–859 [View Article][PubMed]
    [Google Scholar]
  18. Lee JS, Frevert CW, Wurfel MM, Peiper SC, Wong VA 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; 170:5244–5251 [View Article][PubMed]
    [Google Scholar]
  19. Pruenster M, Rot A. Throwing light on DARC. Biochem Soc Trans 2006; 34:1005–1008 [View Article][PubMed]
    [Google Scholar]
  20. Nalls MA, Wilson JG, Patterson NJ, Tandon A, Zmuda JM et al. Admixture mapping of white cell count: genetic locus responsible for lower white blood cell count in the Health ABC and Jackson Heart studies. Am J Hum Genet 2008; 82:81–87 [View Article][PubMed]
    [Google Scholar]
  21. Reich D, Nalls MA, Kao WH, Akylbekova EL, Tandon A 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:e1000360 [View Article][PubMed]
    [Google Scholar]
  22. World Health Organization Report from the scientific group on HTLV-1 infection and its associated diseases, convened by the regional office for the Western Pacific of the World Health Organization in Kagoshima, Japan, 10–15 December 1988. Wkly Epidemiol Rec 1989; 49:382–383
    [Google Scholar]
  23. Andrade RG, Ribeiro MA, Namen-Lopes MS, Silva SM, Basques FV et al. Evaluation of the use of real-time PCR for human T cell lymphotropic virus 1 and 2 as a confirmatory test in screening for blood donors. Rev Soc Bras Med Trop 2010; 43:111–115 [View Article][PubMed]
    [Google Scholar]
  24. Tanaka M, Takahahi J, Hirayama F, Tani Y. High-resolution melting analysis for genotyping Duffy, Kidd and Diego blood group antigens. Leg Med 2011; 13:1–6 [View Article][PubMed]
    [Google Scholar]
  25. Santos HC, Horimoto AV, Tarazona-Santos E, Rodrigues-Soares F, Barreto ML et al. A minimum set of ancestry informative markers for determining admixture proportions in a mixed American population: the Brazilian set. Eur J Hum Genet 2016; 24:725–731 [View Article][PubMed]
    [Google Scholar]
  26. Alexander DH, Novembre J, Lange K. Fast model-based estimation of ancestry in unrelated individuals. Genome Res 2009; 19:1655–1664 [View Article][PubMed]
    [Google Scholar]
  27. Solé X, Guinó E, Valls J, Iniesta R, Moreno V. SNPStats: a web tool for the analysis of association studies. Bioinformatics 2006; 22:1928–1929 [View Article][PubMed]
    [Google Scholar]
  28. Jeffery KJ, Usuku K, Hall SE, Matsumoto W, Taylor GP et al. HLA alleles determine human T-lymphotropic virus-I (HTLV-I) proviral load and the risk of HTLV-I-associated myelopathy. Proc Natl Acad Sci USA 1999; 96:3848–3853 [View Article][PubMed]
    [Google Scholar]
  29. Jeffery KJ, Siddiqui AA, Bunce M, Lloyd AL, Vine AM et al. The influence of HLA class I alleles and heterozygosity on the outcome of human T cell lymphotropic virus type I infection. J Immunol 2000; 165:7278–7284 [View Article][PubMed]
    [Google Scholar]
  30. Darbonne WC, Rice GC, Mohler MA, Apple T, Hébert CA et al. Red blood cells are a sink for interleukin 8, a leukocyte chemotaxin. J Clin Invest 1991; 88:1362–1369 [View Article][PubMed]
    [Google Scholar]
  31. Vine AM, Heaps AG, Kaftantzi L, Mosley A, Asquith B et al. The role of CTLs in persistent viral infection: cytolytic gene expression in CD8+ lymphocytes distinguishes between individuals with a high or low proviral load of human T cell lymphotropic virus type 1. J Immunol 2004; 173:5121–5129 [View Article][PubMed]
    [Google Scholar]
  32. Chaves DG, Sales CC, de Cássia Gonçalves P, da Silva-Malta MC, Romanelli LC et al. Plasmatic proinflammatory chemokines levels are tricky markers to monitoring HTLV-1 carriers. J Med Virol 2016; 88:1438–1447 [View Article][PubMed]
    [Google Scholar]
  33. Schnabel RB, Baumert J, Barbalic M, Dupuis J, Ellinor PT et al. Duffy antigen receptor for chemokines (Darc) polymorphism regulates circulating concentrations of monocyte chemoattractant protein-1 and other inflammatory mediators. Blood 2010; 115:5289–5299 [View Article][PubMed]
    [Google Scholar]
  34. Voruganti VS, Laston S, Haack K, Mehta NR, Smith CW et al. Genome-wide association replicates the association of Duffy antigen receptor for chemokines (DARC) polymorphisms with serum monocyte chemoattractant protein-1 (MCP-1) levels in Hispanic children. Cytokine 2012; 60:634–638 [View Article][PubMed]
    [Google Scholar]
  35. Jilma-Stohlawetz P, Homoncik M, Drucker C, Marsik C, Rot A et al. Fy phenotype and gender determine plasma levels of monocyte chemotactic protein. Transfusion 2001; 41:378–381 [View Article][PubMed]
    [Google Scholar]
  36. Mayr FB, Spiel AO, Leitner JM, Firbas C, Kliegel T et al. Duffy antigen modifies the chemokine response in human endotoxemia. Crit Care Med 2008; 36:159–165 [View Article][PubMed]
    [Google Scholar]
  37. Lee JS, Wurfel MM, Matute-Bello G, Frevert CW, Rosengart MR et al. The Duffy antigen modifies systemic and local tissue chemokine responses following lipopolysaccharide stimulation. J Immunol 2006; 177:8086–8094 [View Article][PubMed]
    [Google Scholar]
  38. Fukuma N, Akimitsu N, Hamamoto H, Kusuhara H, Sugiyama Y et al. A role of the Duffy antigen for the maintenance of plasma chemokine concentrations. Biochem Biophys Res Commun 2003; 303:137–139 [View Article][PubMed]
    [Google Scholar]
  39. Ulvmar MH, Hub E, Rot A. Atypical chemokine receptors. Exp Cell Res 2011; 317:556–568 [View Article][PubMed]
    [Google Scholar]
  40. He W, Neil S, Kulkarni H, Wright E, Agan BK 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; 4:52–62 [View Article][PubMed]
    [Google Scholar]
  41. Kulkarni H, Marconi VC, He W, Landrum ML, Okulicz JF et al. The Duffy-null state is associated with a survival advantage in leukopenic HIV-infected persons of African ancestry. Blood 2009; 114:2783–2792 [View Article][PubMed]
    [Google Scholar]
  42. Ramsuran V, Kulkarni H, He W, Mlisana K, Wright EJ et al. Duffy-null-associated low neutrophil counts influence HIV-1 susceptibility in high-risk South African black women. Clin Infect Dis 2011; 52:1248–1256 [View Article][PubMed]
    [Google Scholar]
  43. Walley NM, Julg B, Dickson SP, Fellay J, Ge D et al. The Duffy antigen receptor for chemokines null promoter variant does not influence HIV-1 acquisition or disease progression. Cell Host Microbe 2009; 5:408–410 [View Article][PubMed]
    [Google Scholar]
  44. Julg B, Reddy S, van der Stok M, Kulkarni S, Qi Y et al. Lack of Duffy antigen receptor for chemokines: no influence on HIV disease progression in an African treatment-naive population. Cell Host Microbe 2009; 5:413–415 [View Article][PubMed]
    [Google Scholar]
  45. Horne KC, Li X, Jacobson LP, Palella F, Jamieson BD et al. Duffy antigen polymorphisms do not alter progression of HIV in African Americans in the MACS cohort. Cell Host Microbe 2009; 5:415–417 [View Article][PubMed]
    [Google Scholar]
  46. Winkler CA, An P, Johnson R, Nelson GW, Kirk G. Expression of Duffy antigen receptor for chemokines (DARC) has no effect on HIV-1 acquisition or progression to AIDS in African Americans. Cell Host Microbe 2009; 5:411–413 [View Article][PubMed]
    [Google Scholar]
  47. Lettow I, Berres ML, Schmitz P, Müller T, Berg T et al. A Duffy antigen receptor for chemokines (DARC) polymorphism that determines pro-fibrotic chemokine serum concentrations is not directly associated with severity of hepatitis C infection. Hum Immunol 2011; 72:273–277 [View Article][PubMed]
    [Google Scholar]
http://instance.metastore.ingenta.com/content/journal/jmm/10.1099/jmm.0.000539
Loading
/content/journal/jmm/10.1099/jmm.0.000539
Loading

Data & Media loading...

This is a required field
Please enter a valid email address
Approval was a Success
Invalid data
An Error Occurred
Approval was partially successful, following selected items could not be processed due to error