The Plasmodium falciparum-specific human memory B cell compartment expands gradually with repeated malaria infections

Greta E Weiss, Boubacar Traore, Kassoum Kayentao, Aissata Ongoiba, Safiatou Doumbo, Didier Doumtabe, Younoussou Kone, Seydou Dia, Agnes Guindo, Abdramane Traore, Chiung-Yu Huang, Kazutoyo Miura, Marko Mircetic, Shanping Li, Amy Baughman, David L Narum, Louis H Miller, Ogobara K Doumbo, Susan K Pierce, Peter D Crompton, Greta E Weiss, Boubacar Traore, Kassoum Kayentao, Aissata Ongoiba, Safiatou Doumbo, Didier Doumtabe, Younoussou Kone, Seydou Dia, Agnes Guindo, Abdramane Traore, Chiung-Yu Huang, Kazutoyo Miura, Marko Mircetic, Shanping Li, Amy Baughman, David L Narum, Louis H Miller, Ogobara K Doumbo, Susan K Pierce, Peter D Crompton

Abstract

Immunity to Plasmodium falciparum (Pf) malaria is only acquired after years of repeated infections and wanes rapidly without ongoing parasite exposure. Antibodies are central to malaria immunity, yet little is known about the B-cell biology that underlies the inefficient acquisition of Pf-specific humoral immunity. This year-long prospective study in Mali of 185 individuals aged 2 to 25 years shows that Pf-specific memory B-cells and antibodies are acquired gradually in a stepwise fashion over years of repeated Pf exposure. Both Pf-specific memory B cells and antibody titers increased after acute malaria and then, after six months of decreased Pf exposure, contracted to a point slightly higher than pre-infection levels. This inefficient, stepwise expansion of both the Pf-specific memory B-cell and long-lived antibody compartments depends on Pf exposure rather than age, based on the comparator response to tetanus vaccination that was efficient and stable. These observations lend new insights into the cellular basis of the delayed acquisition of malaria immunity.

Conflict of interest statement

The authors have declared that no competing interests exist.

Figures

Figure 1. Malaria immunity is acquired gradually…
Figure 1. Malaria immunity is acquired gradually despite intense annual exposure to the Pf parasite.
(A) Number of malaria episodes per day during the study period. Over a one-year surveillance period 185 individuals experienced 219 malaria episodes during a sharply-demarcated six-month malaria season. Malaria episodes were defined as fever ≥37.5°C and Pf asexual parasitemia ≥5000/µL blood. To track the B-cell response to acute malaria, and after a period of reduced Pf exposure, PBMCs and plasma were collected at points indicated by the arrows: before the malaria season, two weeks after the first malaria episode (arrow with asterisk indicates the mean time to first malaria episode, 132 days from enrollment), and six months after the end of the malaria season. (B) Kaplan-Meier estimates of the cumulative probability of malaria over the study period, according to age category. The number of individuals at risk over the study period is shown below the graph. The P value was obtained using the log rank test.
Figure 2. The Pf -specific MBC and…
Figure 2. The Pf-specific MBC and long-lived antibody compartments expand gradually with age.
Shown are the MBC frequencies (bars, left axis) and antibody levels (lines, right axis) specific for AMA1 (A) and MSP1 (B) by age category; and TT (C) by age category and gender; before the malaria season in Pf-uninfected individuals. The frequency of AMA1- and MSP1-specific MBCs increased with age (P<0.001 for both trends), as did the level of AMA1- and MSP1-specific antibodies (P<0.001 for both trends). There were no significant differences by gender for the AMA1- and MSP1-specific responses (not shown). To determine if the expansion of Pf-specific MBCs with age was driven by exposure to antigen or simply a function of age, we measured the TT-specific MBC and antibody response with age. In Mali, infants are vaccinated with TT, and females receive a TT booster around the age of 15 years to prevent neonatal tetanus. In contrast to AMA1 and MSP1, the frequency of TT-specific MBCs and the level of TT-specific antibodies for males did not change significantly from age 2 to 25 years (P = 0.80 and P = 0.44, respectively). However, the frequency of TT-specific MBCs and the level of TT-specific antibodies was higher in female adults compared to female children (P<0.001 for both comparisons). MBC frequencies were determined by ELISPOT and are expressed per million PBMC. The number of individual samples assayed and the percent of individual samples that exceeded the limit of detection (i.e. those considered positive) is indicated below the graph. The discrepancy in the sample size for ELISA data among 2–4 year olds is due to technical error during the performance of the ELISA. P values were obtained by the Kruskal-Wallis test. Data are shown as mean ± s.e.m.
Figure 3. The size of total IgG…
Figure 3. The size of total IgG+ MBC compartment expands gradually with age.
The frequency of IgG+ MBCs per million PBMCs measured before the malaria season increased with age (P<0.001). The number of individuals in each age category is indicated. The P value was obtained by the Kruskal-Wallis test. Data are shown as mean ± s.e.m.
Figure 4. Longitudinal analysis of the Pf…
Figure 4. Longitudinal analysis of the Pf- and TT-specific MBC and antibody response.
Compared to month zero, the MBC frequencies and antibody levels specific for AMA1 (A) and MSP1 (B) increased two weeks after the first episode of malaria and then contracted to a point slightly higher than pre-infection levels after a six-month period of decreased Pf exposure. Compared to month zero, there was a small but statistically significant increase in TT-specific MBC two weeks after the first episode of malaria (C), whereas the level of TT-specific antibodies did not change. The number of individuals in each age category is indicated. Only statistically significant P values are shown. P values were obtained by the Wilcoxon matched-pairs signed-rank test. Data are shown as mean ± s.e.m.
Figure 5. Profile of B-cell subsets before…
Figure 5. Profile of B-cell subsets before the malaria season in children and adults.
(A) By flow cytometry the following B cell subsets were quantified from samples collected before the malaria season: immature B cells CD19+ CD10+, naïve B cells CD19+ CD27− CD21+ CD10−, atypical MBCs CD19+ CD27− CD21− CD10−, classical MBCs CD19+ CD27+ CD21+, activated MBCs CD19+ CD21− CD27+ CD20+, and PCs CD19+ CD21− CD20− CD10−. As a percentage of CD19+ B cells, immature B cells (P<0.001) and naïve B cells (P = 0.047) decreased with age, while atypical MBCs (P = 0.002), IgG+ atypical MBCs (P<0.001), IgG+ classical MBCs (P<0.001)), activated MBCs (P = 0.001), IgG+ activated MBCs (P<0.001) and PCs (P = 0.046) increased with age. P values were obtained by the Kruskal-Wallis test. (B) As a percentage of CD19+ B cells, atypical MBCs decreased 14 days after acute malaria in children aged 2–10 years compared to the percentage before the malaria season. The P value was obtained by the Wilcoxon matched-pairs signed-rank test. Data are shown as mean ± s.e.m.

References

    1. Plotkin SA. Vaccines: correlates of vaccine-induced immunity. Clin Infect Dis. 2008;47:401–409.
    1. Vekemans J, Ballou WR. Plasmodium falciparum malaria vaccines in development. Expert Rev Vaccines. 2008;7:223–240.
    1. Marsh K, Kinyanjui S. Immune effector mechanisms in malaria. Parasite Immunol. 2006;28:51–60.
    1. Cohen S, Mc GI, Carrington S. Gamma-globulin and acquired immunity to human malaria. Nature. 1961;192:733–737.
    1. Langhorne J, Ndungu FM, Sponaas AM, Marsh K. Immunity to malaria: more questions than answers. Nat Immunol. 2008;9:725–732.
    1. Gourley TS, Wherry EJ, Masopust D, Ahmed R. Generation and maintenance of immunological memory. Semin Immunol. 2004;16:323–333.
    1. Tarlinton DM. Evolution in miniature: selection, survival and distribution of antigen reactive cells in the germinal centre. Immunol Cell Biol. 2008;86:133–138.
    1. Manz RA, Thiel A, Radbruch A. Lifetime of plasma cells in the bone marrow. Nature. 1997;388:133–134.
    1. Zinkernagel RM, Bachmann MF, Kundig TM, Oehen S, Pirchet H, et al. On immunological memory. Annu Rev Immunol. 1996;14:333–367.
    1. Bernasconi NL, Traggiai E, Lanzavecchia A. Maintenance of serological memory by polyclonal activation of human memory B cells. Science. 2002;298:2199–2202.
    1. Macallan DC, Wallace DL, Zhang Y, Ghattas H, Asquith B, et al. B-cell kinetics in humans: rapid turnover of peripheral blood memory cells. Blood. 2005;105:3633–3640.
    1. Greenwood BM, Vick RM. Evidence for a malaria mitogen in human malaria. Nature. 1975;257:592–594.
    1. Donati D, Zhang LP, Chene A, Chen Q, Flick K, et al. Identification of a polyclonal B-cell activator in Plasmodium falciparum. Infect Immun. 2004;72:5412–5418.
    1. Crompton PD, Traore B, Kayentao K, Doumbo S, Ongoiba A, et al. Sickle Cell Trait Is Associated with a Delayed Onset of Malaria: Implications for Time-to-Event Analysis in Clinical Studies of Malaria. J Infect Dis. 2008;198:1265–1275.
    1. Severe falciparum malaria. World Health Organization, Communicable Diseases Cluster. Trans R Soc Trop Med Hyg. 2000;94(Suppl 1):S1–90.
    1. Crompton PD, Mircetic M, Weiss G, Baughman A, Huang CY, et al. The TLR9 ligand CpG promotes the acquisition of Plasmodium falciparum-specific memory B cells in malaria-naive individuals. J Immunol. 2009;182:3318–3326.
    1. Dicko A, Klion AD, Thera MA, Sagara I, Yalcouye D, et al. The etiology of severe anemia in a village and a periurban area in Mali. Blood. 2004;104:1198–1200.
    1. Weiss GE, Crompton PD, Li S, Walsh LA, Moir S, et al. Atypical memory B cells are greatly expanded in individuals living in a malaria-endemic area. J Immunol. 2009;183:2176–2182.
    1. Moir S, Ho J, Malaspina A, Wang W, DiPoto AC, 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.
    1. Sagara I, Dicko A, Ellis RD, Fay MP, Diawara SI, et al. A randomized controlled phase 2 trial of the blood stage AMA1-C1/Alhydrogel malaria vaccine in children in Mali. Vaccine. 2009;27:3090–3098.
    1. Ogutu BR, Apollo OJ, McKinney D, Okoth W, Siangla J, et al. Blood stage malaria vaccine eliciting high antigen-specific antibody concentrations confers no protection to young children in Western Kenya. PLoS One. 2009;4:e4708.
    1. Wipasa J, Suphavilai C, Okell LC, Cook J, Corran PH, et al. Long-lived antibody and B Cell memory responses to the human malaria parasites, Plasmodium falciparum and Plasmodium vivax. PLoS Pathog. 6:e1000770.
    1. Dogan I, Bertocci B, Vilmont V, Delbos F, Megret J, et al. Multiple layers of B cell memory with different effector functions. Nat Immunol. 2009;10:1292–1299.
    1. Anderson SM, Tomayko MM, Ahuja A, Haberman AM, Shlomchik MJ. New markers for murine memory B cells that define mutated and unmutated subsets. J Exp Med. 2007;204:2103–2114.
    1. Baer J, Santiago F, Yang H, Wu H, Holden-Wiltse J, et al. B cell responses to H5 influenza HA in human subjects vaccinated with a drifted variant. Vaccine. 28:907–915.
    1. Wrammert J, Smith K, Miller J, Langley WA, Kokko K, et al. Rapid cloning of high-affinity human monoclonal antibodies against influenza virus. Nature. 2008;453:667–671.
    1. Miller JD, van der Most RG, Akondy RS, Glidewell JT, Albott S, et al. Human effector and memory CD8+ T cell responses to smallpox and yellow fever vaccines. Immunity. 2008;28:710–722.
    1. Harris AM, Bhuiyan MS, Chowdhury F, Khan AI, Hossain A, et al. Antigen-specific memory B-cell responses to Vibrio cholerae O1 infection in Bangladesh. Infect Immun. 2009;77:3850–3856.
    1. Morell A, Terry WD, Waldmann TA. Metabolic properties of IgG subclasses in man. J Clin Invest. 1970;49:673–680.
    1. Cavanagh DR, Elhassan IM, Roper C, Robinson VJ, Giha H, et al. A longitudinal study of type-specific antibody responses to Plasmodium falciparum merozoite surface protein-1 in an area of unstable malaria in Sudan. J Immunol. 1998;161:347–359.
    1. Kinyanjui SM, Conway DJ, Lanar DE, Marsh K. IgG antibody responses to Plasmodium falciparum merozoite antigens in Kenyan children have a short half-life. Malar J. 2007;6:82.
    1. Amanna IJ, Carlson NE, Slifka MK. Duration of humoral immunity to common viral and vaccine antigens. N Engl J Med. 2007;357:1903–1915.
    1. Crompton PD, Kayala MA, Traore B, Kayentao K, Ongoiba A, et al. A prospective analysis of the antibody response to Plasmodium falciparum before and after a malaria season by protein microarray. Proc Natl Acad Sci U S A. In press
    1. Crotty S, Felgner P, Davies H, Glidewell J, Villarreal L, et al. Cutting edge: long-term B cell memory in humans after smallpox vaccination. J Immunol. 2003;171:4969–4973.
    1. Dorfman JR, Bejon P, Ndungu FM, Langhorne J, Kortok MM, et al. B cell memory to 3 Plasmodium falciparum blood-stage antigens in a malaria-endemic area. J Infect Dis. 2005;191:1623–1630.
    1. Scherf A, Lopez-Rubio JJ, Riviere L. Antigenic variation in Plasmodium falciparum. Annu Rev Microbiol. 2008;62:445–470.
    1. Takala SL, Plowe CV. Genetic diversity and malaria vaccine design, testing and efficacy: preventing and overcoming ‘vaccine resistant malaria’. Parasite Immunol. 2009;31:560–573.
    1. Ruben FL, Smith EA, Foster SO, Casey HL, Pifer JM, et al. Simultaneous administration of smallpox, measles, yellow fever, and diphtheria-pertussis-tetanus antigens to Nigerian children. Bull World Health Organ. 1973;48:175–181.
    1. Delgado MF, Coviello S, Monsalvo AC, Melendi GA, Hernandez JZ, et al. Lack of antibody affinity maturation due to poor Toll-like receptor stimulation leads to enhanced respiratory syncytial virus disease. Nat Med. 2009;15:34–41.
    1. Traore B, Kone Y, Doumbo S, Doumtabe D, Traore A, et al. The TLR9 agonist CpG fails to enhance the acquisition of Plasmodium falciparum-specific memory B cells in semi-immune adults in Mali. Vaccine 2009
    1. Wykes MN, Zhou YH, Liu XQ, Good MF. Plasmodium yoelii can ablate vaccine-induced long-term protection in mice. J Immunol. 2005;175:2510–2516.
    1. Zellweger RM, Hangartner L, Weber J, Zinkernagel RM, Hengartner H. Parameters governing exhaustion of rare T cell-independent neutralizing IgM-producing B cells after LCMV infection. Eur J Immunol. 2006;36:3175–3185.
    1. Carvalho LJ, Ferreira-da-Cruz MF, Daniel-Ribeiro CT, Pelajo-Machado M, Lenzi HL. Germinal center architecture disturbance during Plasmodium berghei ANKA infection in CBA mice. Malar J. 2007;6:59.
    1. Achtman AH, Khan M, MacLennan IC, Langhorne J. Plasmodium chabaudi chabaudi infection in mice induces strong B cell responses and striking but temporary changes in splenic cell distribution. J Immunol. 2003;171:317–324.
    1. Schmidlin H, Diehl SA, Blom B. New insights into the regulation of human B-cell differentiation. Trends Immunol. 2009;30:277–285.
    1. O'Connor BP, Raman VS, Erickson LD, Cook WJ, Weaver LK, et al. BCMA is essential for the survival of long-lived bone marrow plasma cells. J Exp Med. 2004;199:91–98.
    1. Weill JC, Weller S, Reynaud CA. Human marginal zone B cells. Annu Rev Immunol. 2009;27:267–285.
    1. Querec TD, Akondy RS, Lee EK, Cao W, Nakaya HI, et al. Systems biology approach predicts immunogenicity of the yellow fever vaccine in humans. Nat Immunol. 2009;10:116–125.
    1. Ehrhardt GR, Hsu JT, Gartland L, Leu CM, Zhang S, 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.
    1. Simpson JA, Aarons L, Collins WE, Jeffery GM, White NJ. Population dynamics of untreated Plasmodium falciparum malaria within the adult human host during the expansion phase of the infection. Parasitology. 2002;124:247–263.
    1. West DJ, Calandra GB. Vaccine induced immunologic memory for hepatitis B surface antigen: implications for policy on booster vaccination. Vaccine. 1996;14:1019–1027.
    1. Curtain CC, Kidson C, Champness DL, Gorman JG. Malaria Antibody Content of Gamma 2-7s Globulin in Tropical Populations. Nature. 1964;203:1366–1367.
    1. Di Genova G, Roddick J, McNicholl F, Stevenson FK. Vaccination of human subjects expands both specific and bystander memory T cells but antibody production remains vaccine specific. Blood. 2006;107:2806–2813.
    1. Ahuja A, Anderson SM, Khalil A, Shlomchik MJ. Maintenance of the plasma cell pool is independent of memory B cells. Proc Natl Acad Sci U S A. 2008;105:4802–4807.
    1. Richard K, Pierce SK, Song W. The agonists of TLR4 and 9 are sufficient to activate memory B cells to differentiate into plasma cells in vitro but not in vivo. J Immunol. 2008;181:1746–1752.
    1. Florens L, Washburn MP, Raine JD, Anthony RM, Grainger M, et al. A proteomic view of the Plasmodium falciparum life cycle. Nature. 2002;419:520–526.
    1. Daniel-Ribeiro CT, Zanini G. Autoimmunity and malaria: what are they doing together? Acta Trop. 2000;76:205–221.
    1. Zanini GM, De Moura Carvalho LJ, Brahimi K, De Souza-Passos LF, Guimaraes SJ, et al. Sera of patients with systemic lupus erythematosus react with plasmodial antigens and can inhibit the in vitro growth of Plasmodium falciparum. Autoimmunity. 2009;42:545–552.
    1. Benson MJ, Elgueta R, Schpero W, Molloy M, Zhang W, et al. Distinction of the memory B cell response to cognate antigen versus bystander inflammatory signals. J Exp Med. 2009;206:2013–2025.
    1. Donati D, Mok B, Chene A, Xu H, Thangarajh M, et al. Increased B cell survival and preferential activation of the memory compartment by a malaria polyclonal B cell activator. J Immunol. 2006;177:3035–3044.
    1. Krishnegowda G, Hajjar AM, Zhu J, Douglass EJ, Uematsu S, et al. Induction of proinflammatory responses in macrophages by the glycosylphosphatidylinositols of Plasmodium falciparum: cell signaling receptors, glycosylphosphatidylinositol (GPI) structural requirement, and regulation of GPI activity. J Biol Chem. 2005;280:8606–8616.
    1. Parroche P, Lauw FN, Goutagny N, Latz E, Monks BG, et al. Malaria hemozoin is immunologically inert but radically enhances innate responses by presenting malaria DNA to Toll-like receptor 9. Proc Natl Acad Sci U S A. 2007;104:1919–1924.
    1. Jones KR, Hickling JK, Targett GA, Playfair JH. Polyclonal in vitro proliferative responses from nonimmune donors to Plasmodium falciparum malaria antigens require UCHL1+ (memory) T cells. Eur J Immunol. 1990;20:307–315.
    1. Ho M, Tongtawe P, Kriangkum J, Wimonwattrawatee T, Pattanapanyasat K, et al. Polyclonal expansion of peripheral gamma delta T cells in human Plasmodium falciparum malaria. Infect Immun. 1994;62:855–862.
    1. Hviid L, Kurtzhals JA, Adabayeri V, Loizon S, Kemp K, et al. Perturbation and proinflammatory type activation of V delta 1(+) gamma delta T cells in African children with Plasmodium falciparum malaria. Infect Immun. 2001;69:3190–3196.
    1. Good KL, Avery DT, Tangye SG. Resting human memory B cells are intrinsically programmed for enhanced survival and responsiveness to diverse stimuli compared to naive B cells. J Immunol. 2009;182:890–901.
    1. Wykes MN, Good MF. What have we learnt from mouse models for the study of malaria? Eur J Immunol. 2009;39:2004–2007.
    1. Mestas J, Hughes CC. Of mice and not men: differences between mouse and human immunology. J Immunol. 2004;172:2731–2738.
    1. Davis MM. A prescription for human immunology. Immunity. 2008;29:835–838.
    1. Miura K OA, Muratova OV, Miller LH, Saul A, Long CA. Development and characterization of a standardized ELISA including a reference serum on each plate to detect antibodies induced by experimental malaria vaccines. Vaccine. 2008;26:193–200.
    1. Crotty S, Aubert RD, Glidewell J, Ahmed R. Tracking human antigen-specific memory B cells: a sensitive and generalized ELISPOT system. J Immunol Methods. 2004;286:111–122.

Source: PubMed

3
Subskrybuj