#PAGE_PARAMS# #ADS_HEAD_SCRIPTS# #MICRODATA#

FCRL5 Delineates Functionally Impaired Memory B Cells Associated with Exposure


A subset of “atypical” memory B cells found in individuals with high exposure to P. falciparum has been hypothesized to be dysfunctional, based on phenotypic similarities to analogous cells found in HIV-infected individuals. However, the functional capabilities of these cells have been poorly characterized in the setting of malaria exposure, and previous reports have been controversial regarding whether these cells produce antibody. In our study, we analyze the molecular programming of atypical memory B cells, find that they are dysfunctional in a manner similar to that observed in B cells from HIV-infected individuals, and present data that may reconcile previously conflicting studies. By delineating the transcriptional landscape of atMBCs and identifying expression of FCRL5 as a key marker of dysfunction, we provide a foundation for improving our understanding of the role of these cells in immunity to malaria.


Vyšlo v časopise: FCRL5 Delineates Functionally Impaired Memory B Cells Associated with Exposure. PLoS Pathog 11(5): e32767. doi:10.1371/journal.ppat.1004894
Kategorie: Research Article
prolekare.web.journal.doi_sk: https://doi.org/10.1371/journal.ppat.1004894

Souhrn

A subset of “atypical” memory B cells found in individuals with high exposure to P. falciparum has been hypothesized to be dysfunctional, based on phenotypic similarities to analogous cells found in HIV-infected individuals. However, the functional capabilities of these cells have been poorly characterized in the setting of malaria exposure, and previous reports have been controversial regarding whether these cells produce antibody. In our study, we analyze the molecular programming of atypical memory B cells, find that they are dysfunctional in a manner similar to that observed in B cells from HIV-infected individuals, and present data that may reconcile previously conflicting studies. By delineating the transcriptional landscape of atMBCs and identifying expression of FCRL5 as a key marker of dysfunction, we provide a foundation for improving our understanding of the role of these cells in immunity to malaria.


Zdroje

1. Cohen S, McGregor IA, Carrington S. Gamma-globulin and acquired immunity to human malaria. Nature. 1961;192: 733–737. 13880318

2. Bejon P, Warimwe G, Mackintosh CL, Mackinnon MJ, Kinyanjui SM, Musyoki JN, et al. Analysis of immunity to febrile malaria in children that distinguishes immunity from lack of exposure. Infect Immun. 2009;77: 1917–1923. doi: 10.1128/IAI.01358-08 19223480

3. Sabchareon A, Burnouf T, Ouattara D, Attanath P, Bouharoun-Tayoun H, Chantavanich P, et al. Parasitologic and clinical human response to immunoglobulin administration in falciparum malaria. Am J Trop Med Hyg. 1991;45: 297–308. 1928564

4. WHO | World Malaria Report 2013. In: WHO [Internet]. [cited 5 Jan 2015]. http://www.who.int/malaria/publications/world_malaria_report_2013/en/

5. Beadle C, McElroy PD, Oster CN, Beier JC, Oloo AJ, Onyango FK, et al. Impact of transmission intensity and age on Plasmodium falciparum density and associated fever: implications for malaria vaccine trial design. J Infect Dis. 1995;172: 1047–1054. 7561179

6. Doolan DL, Dobaño C, Baird JK. Acquired Immunity to Malaria. Clin Microbiol Rev. 2009;22: 13–36. doi: 10.1128/CMR.00025-08 19136431

7. Murray CJL, Rosenfeld LC, Lim SS, Andrews KG, Foreman KJ, Haring D, et al. Global malaria mortality between 1980 and 2010: a systematic analysis. Lancet. 2012;379: 413–431. doi: 10.1016/S0140-6736(12)60034-8 22305225

8. Struik SS, Riley EM. Does malaria suffer from lack of memory? Immunol Rev. 2004;201: 268–290. 15361247

9. Langhorne J, Ndungu FM, Sponaas A-M, Marsh K. Immunity to malaria: more questions than answers. Nat Immunol. 2008;9: 725–732. doi: 10.1038/ni.f.205 18563083

10. Illingworth J, Butler NS, Roetynck S, Mwacharo J, Pierce SK, Bejon P, et al. Chronic Exposure to Plasmodium falciparum Is Associated with Phenotypic Evidence of B and T Cell Exhaustion. J Immunol. 2013;190: 1038–1047. doi: 10.4049/jimmunol.1202438 23264654

11. Weiss GE, Crompton PD, Li S, Walsh LA, Moir S, Traore B, et al. Atypical memory B cells are greatly expanded in individuals living in a malaria-endemic area. J Immunol Baltim Md 1950. 2009;183: 2176–2182. doi: 10.4049/jimmunol.0901297 19592645

12. Weiss GE, Clark EH, Li S, Traore B, Kayentao K, Ongoiba A, et al. A Positive Correlation between Atypical Memory B Cells and Plasmodium falciparum Transmission Intensity in Cross-Sectional Studies in Peru and Mali. PLoS ONE. 2011;6: e15983. doi: 10.1371/journal.pone.0015983 21264245

13. Weiss GE, Traore B, Kayentao K, Ongoiba A, Doumbo S, Doumtabe D, et al. The Plasmodium falciparum-Specific Human Memory B Cell Compartment Expands Gradually with Repeated Malaria Infections. PLoS Pathog. 2010;6: e1000912. doi: 10.1371/journal.ppat.1000912 20502681

14. Kano FS, Lima BA, Tang ML, Costa PA, Fontes CJ, Sanchez BM, et al. Plasmodium vivax infection: atypical memory B cells are expanded and associated with the persistence of Duffy binding protein II (DBPII) antibody response. Malar J. 2014;13: P52.

15. Nogaro SI, Hafalla JC, Walther B, Remarque EJ, Tetteh KKA, Conway DJ, et al. The Breadth, but Not the Magnitude, of Circulating Memory B Cell Responses to P. falciparum Increases with Age/Exposure in an Area of Low Transmission. PLoS ONE. 2011;6: e25582. doi: 10.1371/journal.pone.0025582 21991321

16. Ayieko C, Maue AC, Jura WGZO, Noland GS, Ayodo G, Rochford R, et al. Changes in B Cell Populations and Merozoite Surface Protein-1-Specific Memory B Cell Responses after Prolonged Absence of Detectable P. falciparum Infection. PLoS ONE. 2013;8: e67230. 23826242

17. Ampomah P, Stevenson L, Ofori MF, Barfod L, Hviid L. Kinetics of B Cell Responses to Plasmodium falciparum Erythrocyte Membrane Protein 1 in Ghanaian Women Naturally Exposed to Malaria Parasites. J Immunol Author Choice. 2014;192: 5236–5244. doi: 10.4049/jimmunol.1400325 24760153

18. Kardava L, Moir S, Shah N, Wang W, Wilson R, Buckner CM, et al. Abnormal B cell memory subsets dominate HIV-specific responses in infected individuals. J Clin Invest. 2014;124: 3252–3262. doi: 10.1172/JCI74351 24892810

19. Kardava L, Moir S, Wang W, Ho J, Buckner CM, Posada JG, et al. Attenuation of HIV-associated human B cell exhaustion by siRNA downregulation of inhibitory receptors. J Clin Invest. 2011;121: 2614–2624. doi: 10.1172/JCI45685 21633172

20. Moir S, Ho J, Malaspina A, Wang W, DiPoto AC, O’Shea MA, et al. Evidence for HIV-associated B cell exhaustion in a dysfunctional memory B cell compartment in HIV-infected viremic individuals. J Exp Med. 2008;205: 1797–1805. doi: 10.1084/jem.20072683 18625747

21. Jacobi AM, Reiter K, Mackay M, Aranow C, Hiepe F, Radbruch A, et al. Activated memory B cell subsets correlate with disease activity in systemic lupus erythematosus: delineation by expression of CD27, IgD, and CD95. Arthritis Rheum. 2008;58: 1762–1773. doi: 10.1002/art.23498 18512812

22. Rakhmanov M, Keller B, Gutenberger S, Foerster C, Hoenig M, Driessen G, et al. Circulating CD21low B cells in common variable immunodeficiency resemble tissue homing, innate-like B cells. Proc Natl Acad Sci U S A. 2009;106: 13451–13456. doi: 10.1073/pnas.0901984106 19666505

23. Isnardi I, Ng Y-S, Menard L, Meyers G, Saadoun D, Srdanovic I, et al. Complement receptor 2/CD21- human naive B cells contain mostly autoreactive unresponsive clones. Blood. 2010;115: 5026–5036. doi: 10.1182/blood-2009-09-243071 20231422

24. Wei C, Anolik J, Cappione A, Zheng B, Pugh-Bernard A, Brooks J, et al. A new population of cells lacking expression of CD27 represents a notable component of the B cell memory compartment in systemic lupus erythematosus. J Immunol Baltim Md 1950. 2007;178: 6624–6633. 17475894

25. Becker AM, Dao KH, Han BK, Kornu R, Lakhanpal S, Mobley AB, et al. SLE peripheral blood B cell, T cell and myeloid cell transcriptomes display unique profiles and each subset contributes to the interferon signature. PloS One. 2013;8: e67003. doi: 10.1371/journal.pone.0067003 23826184

26. Charles ED, Brunetti C, Marukian S, Ritola KD, Talal AH, Marks K, et al. Clonal B cells in patients with hepatitis C virus-associated mixed cryoglobulinemia contain an expanded anergic CD21low B-cell subset. Blood. 2011;117: 5425–5437. doi: 10.1182/blood-2010-10-312942 21421840

27. Ehrhardt GRA, Hsu JT, Gartland L, Leu C-M, Zhang S, Davis RS, et al. Expression of the immunoregulatory molecule FcRH4 defines a distinctive tissue-based population of memory B cells. J Exp Med. 2005;202: 783–791. 16157685

28. Ehrhardt GRA, Hijikata A, Kitamura H, Ohara O, Wang J-Y, Cooper MD. Discriminating gene expression profiles of memory B cell subpopulations. J Exp Med. 2008;205: 1807–1817. doi: 10.1084/jem.20072682 18625746

29. Day CL, Kaufmann DE, Kiepiela P, Brown JA, Moodley ES, Reddy S, et al. PD-1 expression on HIV-specific T cells is associated with T-cell exhaustion and disease progression. Nature. 2006;443: 350–354. 16921384

30. Barber DL, Wherry EJ, Masopust D, Zhu B, Allison JP, Sharpe AH, et al. Restoring function in exhausted CD8 T cells during chronic viral infection. Nature. 2006;439: 682–687. 16382236

31. Muellenbeck MF, Ueberheide B, Amulic B, Epp A, Fenyo D, Busse CE, et al. Atypical and classical memory B cells produce Plasmodium falciparum neutralizing antibodies. J Exp Med. 2013;210: 389–399. doi: 10.1084/jem.20121970 23319701

32. Jagannathan P, Kim CC, Greenhouse B, Nankya F, Bowen K, Eccles-James I, et al. Loss and dysfunction of Vδ2+ γδ T cells are associated with clinical tolerance to malaria. Sci Transl Med. 2014;6: 251ra117. doi: 10.1126/scitranslmed.3009793 25163477

33. Kim CC, Nelson CS, Wilson EB, Hou B, DeFranco AL, DeRisi JL. Splenic red pulp macrophages produce type I interferons as early sentinels of malaria infection but are dispensable for control. PloS One. 2012;7: e48126. doi: 10.1371/journal.pone.0048126 23144737

34. De Nicola F, Catena V, Rinaldo C, Bruno T, Iezzi S, Sorino C, et al. HIPK2 sustains apoptotic response by phosphorylating Che-1/AATF and promoting its degradation. Cell Death Dis. 2014;5: e1414. doi: 10.1038/cddis.2014.381 25210797

35. D’Orazi G, Cecchinelli B, Bruno T, Manni I, Higashimoto Y, Saito S, et al. Homeodomain-interacting protein kinase-2 phosphorylates p53 at Ser 46 and mediates apoptosis. Nat Cell Biol. 2002;4: 11–19. 11780126

36. Hofmann TG, Möller A, Sirma H, Zentgraf H, Taya Y, Dröge W, et al. Regulation of p53 activity by its interaction with homeodomain-interacting protein kinase-2. Nat Cell Biol. 2002;4: 1–10. 11740489

37. Shahbazi J, Lock R, Liu T. Tumor protein 53-induced nuclear protein 1 enhances p53 function and represses tumorigenesis. Cancer Genet. 2013;4: 80.

38. Kuribayashi K, Krigsfeld G, Wang W, Xu J, Mayes PA, Dicker DT, et al. TNFSF10 (TRAIL), a p53 target gene that mediates p53-dependent cell death. Cancer Biol Ther. 2008;7: 2034–2038. 19106633

39. Ihrie RA, Reczek E, Horner JS, Khachatrian L, Sage J, Jacks T, et al. Perp is a mediator of p53-dependent apoptosis in diverse cell types. Curr Biol CB. 2003;13: 1985–1990. 14614825

40. Bodmer JL, Burns K, Schneider P, Hofmann K, Steiner V, Thome M, et al. TRAMP, a novel apoptosis-mediating receptor with sequence homology to tumor necrosis factor receptor 1 and Fas(Apo-1/CD95). Immunity. 1997;6: 79–88. 9052839

41. Munroe ME, Bishop GA. Role of Tumor Necrosis Factor (TNF) Receptor-associated Factor 2 (TRAF2) in Distinct and Overlapping CD40 and TNF Receptor 2/CD120b-mediated B Lymphocyte Activation. J Biol Chem. 2004;279: 53222–53231. 15485859

42. Miscia S, Marchisio M, Grilli A, Di Valerio V, Centurione L, Sabatino G, et al. Tumor necrosis factor alpha (TNF-alpha) activates Jak1/Stat3-Stat5B signaling through TNFR-1 in human B cells. Cell Growth Differ Mol Biol J Am Assoc Cancer Res. 2002;13: 13–18.

43. Ettinger R, Sims GP, Fairhurst A-M, Robbins R, da Silva YS, Spolski R, et al. IL-21 induces differentiation of human naive and memory B cells into antibody-secreting plasma cells. J Immunol Baltim Md 1950. 2005;175: 7867–7879. 16339522

44. Ozaki K, Spolski R, Ettinger R, Kim H-P, Wang G, Qi C-F, et al. Regulation of B cell differentiation and plasma cell generation by IL-21, a novel inducer of Blimp-1 and Bcl-6. J Immunol Baltim Md 1950. 2004;173: 5361–5371. 15494482

45. Moir S, Malaspina A, Pickeral OK, Donoghue ET, Vasquez J, Miller NJ, et al. Decreased survival of B cells of HIV-viremic patients mediated by altered expression of receptors of the TNF superfamily. J Exp Med. 2004;200: 587–599. 15508184

46. Doi H, Tanoue S, Kaplan DE. Peripheral CD27-CD21- B-cells represent an exhausted lymphocyte population in hepatitis C cirrhosis. Clin Immunol Orlando Fla. 2014;150: 184–191. doi: 10.1016/j.clim.2013.12.001 24434272

47. Patel N, Brinkman-Van der Linden EC, Altmann SW, Gish K, Balasubramanian S, Timans JC, et al. OB-BP1/Siglec-6. a leptin- and sialic acid-binding protein of the immunoglobulin superfamily. J Biol Chem. 1999;274: 22729–22738. 10428856

48. Shaffer AL, Yu X, He Y, Boldrick J, Chan EP, Staudt LM. BCL-6 represses genes that function in lymphocyte differentiation, inflammation, and cell cycle control. Immunity. 2000;13: 199–212. 10981963

49. Nicholas KJ, Zern EK, Barnett L, Smith RM, Lorey SL, Copeland CA, et al. B cell responses to HIV antigen are a potent correlate of viremia in HIV-1 infection and improve with PD-1 blockade. PloS One. 2013;8: e84185. doi: 10.1371/journal.pone.0084185 24358343

50. Tarte K, Zhan F, De Vos J, Klein B, Shaughnessy J. Gene expression profiling of plasma cells and plasmablasts: toward a better understanding of the late stages of B-cell differentiation. Blood. 2003;102: 592–600. 12663452

51. Requena P, Campo JJ, Umbers AJ, Ome M, Wangnapi R, Barrios D, et al. Pregnancy and malaria exposure are associated with changes in the B cell pool and in plasma eotaxin levels. J Immunol Baltim Md 1950. 2014;193: 2971–2983. doi: 10.4049/jimmunol.1401037 25135831

52. Davis RS, Wang YH, Kubagawa H, Cooper MD. Identification of a family of Fc receptor homologs with preferential B cell expression. Proc Natl Acad Sci U S A. 2001;98: 9772–9777. 11493702

53. Polson AG, Zheng B, Elkins K, Chang W, Du C, Dowd P, et al. Expression pattern of the human FcRH/IRTA receptors in normal tissue and in B-chronic lymphocytic leukemia. Int Immunol. 2006;18: 1363–1373. 16849395

54. Kilama M, Smith DL, Hutchinson R, Kigozi R, Yeka A, Lavoy G, et al. Estimating the annual entomological inoculation rate for Plasmodium falciparum transmitted by Anopheles gambiae s.l. using three sampling methods in three sites in Uganda. Malar J. 2014;13: 111. doi: 10.1186/1475-2875-13-111 24656206

55. Kamya, Moses, Arinaitwe, Emmanuel, Wanzira, Humphrey, Katureebe, Agaba, Barusya, Chris, Kiqozi, Simon, et al. Malaria Transmission, Infection and Disease at Three Sites with Varied Transmission Intensity in Uganda: Implications for Malaria Control. Am J Trop Med Hyg. In Press.

56. Haga CL, Ehrhardt GRA, Boohaker RJ, Davis RS, Cooper MD. Fc receptor-like 5 inhibits B cell activation via SHP-1 tyrosine phosphatase recruitment. Proc Natl Acad Sci. 2007;104: 9770–9775. 17522256

57. Jelicic K, Cimbro R, Nawaz F, Huang DW, Zheng X, Yang J, et al. HIV-1 gp120 impairs B cell proliferation by inducing TGF-β1 production and FcRL4 expression. Nat Immunol. 2013;14. doi: 10.1038/ni.2489 23238752

58. Franco A, Damdinsuren B, Ise T, Dement-Brown J, Li H, Nagata S, et al. Human Fc receptor-like 5 binds intact IgG via mechanisms distinct from those of Fc receptors. J Immunol Baltim Md 1950. 2013;190: 5739–5746. doi: 10.4049/jimmunol.1202860 23616577

59. Wilson TJ, Fuchs A, Colonna M. Cutting edge: human FcRL4 and FcRL5 are receptors for IgA and IgG. J Immunol Baltim Md 1950. 2012;188: 4741–4745. doi: 10.4049/jimmunol.1102651 22491254

60. Zinöcker S, Schindler CE, Skinner J, Rogosch T, Waisberg M, Schickel J- N, et al. The V Gene Repertoires of Classical and Atypical Memory B Cells in Malaria-Susceptible West African Children. J Immunol. 2015;194: 929–939. doi: 10.4049/jimmunol.1402168 25556245

61. Portugal S, Moebius J, Skinner J, Doumbo S, Doumtabe D, Kone Y, et al. Exposure-dependent control of malaria-induced inflammation in children. PLoS Pathog. 2014;10: e1004079. doi: 10.1371/journal.ppat.1004079 24743880

62. Ladeia-Andrade S, Ferreira MU, de Carvalho ME, Curado I, Coura JR. Age-dependent acquisition of protective immunity to malaria in riverine populations of the Amazon Basin of Brazil. Am J Trop Med Hyg. 2009;80: 452–459. 19270298

63. Smyth GK, Speed T. Normalization of cDNA microarray data. Methods San Diego Calif. 2003;31: 265–273. 14597310

64. Tusher VG, Tibshirani R, Chu G. Significance analysis of microarrays applied to the ionizing radiation response. Proc Natl Acad Sci U S A. 2001;98: 5116–5121. 11309499

65. Huang DW, Sherman BT, Lempicki RA. Systematic and integrative analysis of large gene lists using DAVID bioinformatics resources. Nat Protoc. 2009;4: 44–57. doi: 10.1038/nprot.2008.211 19131956

66. Jeanty C, Longrois D, Mertes P-M, Wagner DR, Devaux Y. An optimized protocol for microarray validation by quantitative PCR using amplified amino allyl labeled RNA. BMC Genomics. 2010;11: 542. doi: 10.1186/1471-2164-11-542 20929564

Štítky
Hygiena a epidemiológia Infekčné lekárstvo Laboratórium

Článok vyšiel v časopise

PLOS Pathogens


2015 Číslo 5
Najčítanejšie tento týždeň
Najčítanejšie v tomto čísle
Kurzy

Zvýšte si kvalifikáciu online z pohodlia domova

Získaná hemofilie - Povědomí o nemoci a její diagnostika
nový kurz

Eozinofilní granulomatóza s polyangiitidou
Autori: doc. MUDr. Martina Doubková, Ph.D.

Všetky kurzy
Prihlásenie
Zabudnuté heslo

Zadajte e-mailovú adresu, s ktorou ste vytvárali účet. Budú Vám na ňu zasielané informácie k nastaveniu nového hesla.

Prihlásenie

Nemáte účet?  Registrujte sa

#ADS_BOTTOM_SCRIPTS#