Skip to main content

Thank you for visiting nature.com. You are using a browser version with limited support for CSS. To obtain the best experience, we recommend you use a more up to date browser (or turn off compatibility mode in Internet Explorer). In the meantime, to ensure continued support, we are displaying the site without styles and JavaScript.

  • Article
  • Published:

Infection-induced plasmablasts are a nutrient sink that impairs humoral immunity to malaria

Abstract

Plasmodium parasite–specific antibodies are critical for protection against malaria, yet the development of long-lived and effective humoral immunity against Plasmodium takes many years and multiple rounds of infection and cure. Here, we report that the rapid development of short-lived plasmablasts during experimental malaria unexpectedly hindered parasite control by impeding germinal center responses. Metabolic hyperactivity of plasmablasts resulted in nutrient deprivation of the germinal center reaction, limiting the generation of memory B cell and long-lived plasma cell responses. Therapeutic administration of a single amino acid to experimentally infected mice was sufficient to overcome the metabolic constraints imposed by plasmablasts and enhanced parasite clearance and the formation of protective humoral immune memory responses. Thus, our studies not only challenge the current model describing the role and function of blood-stage Plasmodium-induced plasmablasts but they also reveal new targets and strategies to improve anti-Plasmodium humoral immunity.

This is a preview of subscription content, access via your institution

Access options

Buy this article

Prices may be subject to local taxes which are calculated during checkout

Fig. 1: Preferential expansion of plasmablasts during experimental malaria.
Fig. 2: Blocking plasmablast development improves humoral immunity.
Fig. 3: Deletion of plasmablasts enhances humoral immunity.
Fig. 4: Plasmablasts are metabolically hyperactive.
Fig. 5: l-glutamine supplementation enhances GC responses.
Fig. 6: Plasmablast deletion and l-glutamine are functionally redundant.
Fig. 7: Plasmablast deletion and l-glutamine enhance humoral immune memory.
Fig. 8: Positive correlation between parasite burden and plasmablasts in human malaria.

Similar content being viewed by others

Data availability

Source data for Figs. 18 and Extended Data Figs. 13, 5 and 7 are presented with the paper. The data that support the findings of this study are available from the Gene Expression Omnibus (accession code GSE134548). All data are available from the corresponding author upon request.

References

  1. Nutt, S. L., Hodgkin, P. D., Tarlinton, D. M. & Corcoran, L. M. The generation of antibody-secreting plasma cells. Nat. Rev. Immunol. 15, 160–171 (2015).

    Article  CAS  PubMed  Google Scholar 

  2. Inoue, T., Moran, I., Shinnakasu, R., Phan, T. G. & Kurosaki, T. Generation of memory B cells and their reactivation. Immunol. Rev. 283, 138–149 (2018).

    Article  CAS  PubMed  Google Scholar 

  3. Martin, F., Oliver, A. M. & Kearney, J. F. Marginal zone and B1 B cells unite in the early response against T-independent blood-borne particulate antigens. Immunity 14, 617–629 (2001).

    Article  CAS  PubMed  Google Scholar 

  4. Racine, R., Chatterjee, M. & Winslow, G. M. CD11c expression identifies a population of extrafollicular antigen-specific splenic plasmablasts responsible for CD4 T-independent antibody responses during intracellular bacterial infection. J. Immunol. 181, 1375–1385 (2008).

    Article  CAS  PubMed  Google Scholar 

  5. Slifka, M. K., Antia, R., Whitmire, J. K. & Ahmed, R. Humoral immunity due to long-lived plasma cells. Immunity 8, 363–372 (1998).

    Article  CAS  PubMed  Google Scholar 

  6. World Malaria Report 2018 (World Health Organization, 2018); https://www.who.int/malaria/publications/world-malaria-report-2018/en/

  7. Cohen, S., McGregor, I. A. & Carrington, S. Gamma-globulin and acquired immunity to human malaria. Nature 192, 733–737 (1961).

    Article  CAS  PubMed  Google Scholar 

  8. Changrob, S. et al. Persistence of long-lived memory B cells specific to Duffy binding protein in individuals exposed to Plasmodium vivax. Sci. Rep. 8, 8347 (2018).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  9. Longley, R. J. et al. Naturally acquired antibody responses to more than 300 Plasmodium vivax proteins in three geographic regions. PLoS Negl. Trop. Dis. 11, e0005888 (2017).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  10. Ndungu, F. M. et al. Long-lived Plasmodium falciparum specific memory B cells in naturally exposed Swedish travelers. Eur. J. Immunol. 43, 2919–2929 (2013).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  11. Crompton, P. D. et al. A prospective analysis of the Ab response to Plasmodium falciparum before and after a malaria season by protein microarray. Proc. Natl Acad. Sci. USA 107, 6958–6963 (2010).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  12. Tran, T. M. et al. An intensive longitudinal cohort study of Malian children and adults reveals no evidence of acquired immunity to Plasmodium falciparum infection. Clin. Infect. Dis. 57, 40–47 (2013).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  13. Obeng-Adjei, N. et al. Circulating Th1-cell-type Tfh cells that exhibit impaired B cell help are preferentially activated during acute malaria in children. Cell Rep. 13, 425–439 (2015).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  14. Kurup, S. P. et al. Regulatory T cells impede acute and long-term immunity to blood-stage malaria through CTLA-4. Nat. Med. 23, 1220–1225 (2017).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  15. Walther, M. et al. Upregulation of TGF-β, FOXP3, and CD4+CD25+ regulatory T cells correlates with more rapid parasite growth in human malaria infection. Immunity 23, 287–296 (2005).

    Article  CAS  PubMed  Google Scholar 

  16. Weiss, G. E. et al. Atypical memory B cells are greatly expanded in individuals living in a malaria-endemic area. J. Immunol. 183, 2176–2182 (2009).

    Article  CAS  PubMed  Google Scholar 

  17. Obeng-Adjei, N. et al. Malaria-induced interferon-γ drives the expansion of Tbethi atypical memory B cells. PLoS Pathog. 13, e1006576 (2017).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  18. Portugal, S. et al. Malaria-associated atypical memory B cells exhibit markedly reduced B cell receptor signaling and effector function. Elife 4, e07218 (2015).

    Article  PubMed Central  CAS  Google Scholar 

  19. Sullivan, R. T. et al. FCRL5 delineates functionally impaired memory B cells associated with Plasmodium falciparum exposure. PLoS Pathog. 11, e1004894 (2015).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  20. Ryg-Cornejo, V. et al. Severe malaria infections impair germinal center responses by inhibiting T follicular helper cell differentiation. Cell Rep. 14, 68–81 (2016).

    Article  CAS  PubMed  Google Scholar 

  21. Keitany, G. J. et al. Blood stage malaria disrupts humoral immunity to the pre-erythrocytic stage circumsporozoite protein. Cell Rep. 17, 3193–3205 (2016).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  22. Butler, N. S. et al. Therapeutic blockade of PD-L1 and LAG-3 rapidly clears established blood-stage Plasmodium infection. Nat. Immunol. 13, 188–195 (2011).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  23. Horne-Debets, J. M. et al. PD-1 dependent exhaustion of CD8+ T cells drives chronic malaria. Cell Rep. 5, 1204–1213 (2013).

    Article  CAS  PubMed  Google Scholar 

  24. Guthmiller, J. J., Graham, A. C., Zander, R. A., Pope, R. L. & Butler, N. S. Cutting edge: IL-10 is essential for the generation of germinal center B cell responses and anti-Plasmodium humoral immunity. J. Immunol. 198, 617–622 (2017).

    Article  CAS  PubMed  Google Scholar 

  25. Krishnamurty, A. T. et al. Somatically hypermutated plasmodium-specific IgM+ memory B cells are rapid, plastic, early responders upon malaria rechallenge. Immunity 45, 402–414 (2016).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  26. Shaffer, A. L. et al. Blimp-1 orchestrates plasma cell differentiation by extinguishing the mature B cell gene expression program. Immunity 17, 51–62 (2002).

    Article  CAS  PubMed  Google Scholar 

  27. William, J., Euler, C. & Shlomchik, M. J. Short-lived plasmablasts dominate the early spontaneous rheumatoid factor response: differentiation pathways, hypermutating cell types, and affinity maturation outside the germinal center. J. Immunol. 174, 6879–6887 (2005).

    Article  CAS  PubMed  Google Scholar 

  28. Tellier, J. et al. Blimp-1 controls plasma cell function through the regulation of immunoglobulin secretion and the unfolded protein response. Nat. Immunol. 17, 323–330 (2016).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  29. Meding, S. J., Cheng, S. C., Simon-Haarhaus, B. & Langhorne, J. Role of gamma interferon during infection with Plasmodium chabaudi chabaudi. Infect. Immun. 58, 3671–3678 (1990).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  30. Su, Z. & Stevenson, M. M. Central role of endogenous gamma interferon in protective immunity against blood-stage Plasmodium chabaudi AS infection. Infect. Immun. 68, 4399–4406 (2000).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  31. Reimold, A. M. et al. Plasma cell differentiation requires the transcription factor XBP-1. Nature 412, 300–307 (2001).

    Article  CAS  PubMed  Google Scholar 

  32. Boothby, M. & Rickert, R. C. Metabolic regulation of the immune humoral response. Immunity 46, 743–755 (2017).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  33. Cantor, J. et al. CD98hc facilitates B cell proliferation and adaptive humoral immunity. Nat. Immunol. 10, 412–419 (2009).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  34. Cordy, R. J. et al. Distinct amino acid and lipid perturbations characterize acute versus chronic malaria. JCI Insight 4, e125156 (2019).

    Article  PubMed Central  Google Scholar 

  35. Suan, D. et al. CCR6 defines memory B cell precursors in mouse and human germinal centers, revealing light-zone location and predominant low antigen affinity. Immunity 47, 1142–1153.e4 (2017).

    Article  CAS  PubMed  Google Scholar 

  36. Alugupalli, K. R. et al. B1b lymphocytes confer T cell-independent long-lasting immunity. Immunity 21, 379–390 (2004).

    Article  CAS  PubMed  Google Scholar 

  37. Haas, K. M., Poe, J. C., Steeber, D. A. & Tedder, T. F. B-1a and B-1b cells exhibit distinct developmental requirements and have unique functional roles in innate and adaptive immunity to S. pneumoniae. Immunity 23, 7–18 (2005).

    Article  CAS  PubMed  Google Scholar 

  38. Pelletier, N. et al. Plasma cells negatively regulate the follicular helper T cell program. Nat. Immunol. 11, 1110–1118 (2010).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  39. Khan, A. R. et al. PD-L1hi B cells are critical regulators of humoral immunity. Nat. Commun. 6, 5997 (2015).

    Article  CAS  PubMed  Google Scholar 

  40. Matsumoto, M. et al. Interleukin-10-producing plasmablasts exert regulatory function in autoimmune inflammation. Immunity 41, 1040–1051 (2014).

    Article  CAS  PubMed  Google Scholar 

  41. Clark, E. H. et al. Plasmodium falciparum malaria in the Peruvian Amazon, a region of low transmission, is associated with immunologic memory. Infect. Immun. 80, 1583–1592 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  42. Labak, C. M. et al. Glucose transport: meeting the metabolic demands of cancer, and applications in glioblastoma treatment. Am. J. Cancer Res. 6, 1599–1608 (2016).

    CAS  PubMed  PubMed Central  Google Scholar 

  43. Lam, W. Y. et al. Metabolic and transcriptional modules independently diversify plasma cell lifespan and function. Cell Rep. 24, 2479–2492.e6 (2018).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  44. Chapuis, N., Poulain, L., Birsen, R., Tamburini, J. & Bouscary, D. Rationale for targeting deregulated metabolic pathways as a therapeutic strategy in acute myeloid leukemia. Front. Oncol. 9, 405 (2019).

    Article  PubMed  PubMed Central  Google Scholar 

  45. Zhu, X., Pan, Y., Li, Y., Cui, L. & Cao, Y. Supplement of l-arg improves protective immunity during early-stage Plasmodium yoelii 17XL infection. Parasite Immunol. 34, 412–420 (2012).

    Article  CAS  PubMed  Google Scholar 

  46. Gordon, E. B. et al. Targeting glutamine metabolism rescues mice from late-stage cerebral malaria. Proc. Natl Acad. Sci. USA 112, 13075–13080 (2015).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  47. Miyakoda, M. et al. Malaria-specific and nonspecific activation of CD8+ T cells during blood stage of Plasmodium berghei infection. J. Immunol. 181, 1420–1428 (2008).

    Article  CAS  PubMed  Google Scholar 

  48. McCarthy, J. S. et al. A pilot randomised trial of induced blood-stage Plasmodium falciparum infections in healthy volunteers for testing efficacy of new antimalarial drugs. PLoS ONE 6, e21914 (2011).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  49. Rockett, R. J. et al. A real-time, quantitative PCR method using hydrolysis probes for the monitoring of Plasmodium falciparum load in experimentally infected human volunteers. Malar. J. 10, 48 (2011).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  50. Collins, K. A. et al. A controlled human malaria infection model enabling evaluation of transmission-blocking interventions. J. Clin. Invest. 128, 1551–1562 (2018).

    Article  PubMed  PubMed Central  Google Scholar 

  51. Montes de Oca, M. et al. Type I interferons regulate immune responses in humans with blood-stage Plasmodium falciparum infection. Cell Rep. 17, 399–412 (2016).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  52. Malleret, B. et al. A rapid and robust tri-color flow cytometry assay for monitoring malaria parasite development. Sci. Rep. 1, 118 (2011).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  53. Villarino, N. F. et al. Composition of the gut microbiota modulates the severity of malaria. Proc. Natl Acad. Sci. USA 113, 2235–2240 (2016).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  54. Dobin, A. et al. STAR: ultrafast universal RNA-Seq aligner. Bioinformatics 29, 15–21 (2013).

    Article  CAS  PubMed  Google Scholar 

  55. Liao, Y., Smyth, G. K. & Shi, W. featureCounts: an efficient general purpose program for assigning sequence reads to genomic features. Bioinformatics 30, 923–930 (2014).

    Article  CAS  PubMed  Google Scholar 

  56. Gray, L. R. et al. Hepatic mitochondrial pyruvate carrier 1 is required for efficient regulation of gluconeogenesis and whole-body glucose homeostasis. Cell Metab. 22, 669–681 (2015).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

Download references

Acknowledgements

We thank members of the University of Iowa Butler laboratory for assistance and helpful discussions, F. Lund (University of Alabama, Birmingham) for the μS–/– mice, T. Honjo (Kyoto University) for the Aicda–/– mice, T. Waldschmidt (University of Iowa) for the clone MR-1 antibody, J. Harty and S. Perlman (University of Iowa) for critical reading of the manuscript and helpful discussions, A. Pewa, E. Taylor and A. Rauckhorst (University of Iowa) for assistance with the metabolic measurements, G. Beuttener and B. Wagner (University of Iowa) for the metabolic flux assays, and members of the University of Iowa Flow Cytometry Facility for cell sorting. The research reported in this publication was supported by the NCI (grant number P30CA086862) and the National Center for Research Resources of the NIH (grant number S10OD016199). J.J.G. was supported by a Predoctoral Fellowship from the American Heart Association (grant number 16PRE27660002). F.A.S. was supported by the NIH (grant number T32 AI007485). K.J.R. was supported by the NIH (T32 GM067795). W.J.M. was supported by the NIH (grant numbers AI134733 and AI139902). H.-H.X. was supported by the NIH (grant numbers AI121080 and AI139874) and the Veteran Affairs BLR&D Merit Review Program (BX002903). C.R.E. was supported by an NHMRC Senior Research Fellowship (grant number 1154265) and an NHMRC Program grant (grant number 1132975). J.S.M. was supported by an NHMRC Program grant (grant number 1132975) and an NHMRC Practitioner Fellowship (grant number 1135955). M.J.B. was supported by an NHMRC Career Development Fellowship (grant number 1141632) and an NHMRC Project Grant (grant number 1141278). N.S.B. was supported by the NIH (grant numbers AI125446, AI127481 and AI139902). We thank the participants involved in the malaria VISs, Q-Pharm staff and the Medicine for Malaria Venture for funding the clinical trials.

Author information

Authors and Affiliations

Authors

Contributions

R.V., J.J.G. and N.S.B. conceptualized the project. J.J.G. performed the foundational studies and characterized the phenotype of plasmablasts in mice. R.V. characterized the transcriptome of the plasmablasts and conceived and executed the adoptive transfer studies, chimeric studies, metabolism analyses and glutamine supplementation experiments. W.J.M., K.J.R., F.L. and H.-H.X. provided critical reagents and technical assistance. C.R.E., J.S.M. and M.J.B. supervised the clinical studies. N.S.B. supervised the experimental rodent studies. R.V., J.J.G., H.-H.X., C.R.E., J.S.M., M.J.B. and N.S.B. designed the experiments. R.V., J.J.G., A.J.S., F.A.S., R.L.P., D.A., J.-A.C. and F.d.L.R. performed the experiments. R.V., J.J.G., A.J.S., F.A.S., R.R.S., J.-A.C., F.d.L.R., L.W., C.R.E., J.S.M., M.J.B. and N.S.B. analyzed the data. R.V. and R.R.S. performed the bioinformatics analyses. R.V. and J.J.G. made the figures. R.V. wrote the original draft. J.J.G., A.J.S., C.R.E., J.S.M., M.J.B. and N.S.B. reviewed and edited the manuscript and figures.

Corresponding author

Correspondence to Noah S. Butler.

Ethics declarations

Competing interests

The authors declare no competing interests.

Additional information

Peer review information L. A. Dempsey was the primary editor on this article and managed its editorial process and peer review in collaboration with the rest of the editorial team.

Publisher’s note Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Extended data

Extended Data Fig. 1 Experimental malaria expands extrafollicular plasmablast populations.

a, Gating strategy for identifying plasmablasts (CD138hiIgDneg), activated (CD138loIgDneg) and resting B cells (CD138loIgDhi). b, kinetics of PB in blood. Data are means ± s.d, representative of n = 2 biologically independent experiments with similar results using n = 3 mice/time point. c, CD138hiIgDneg plasmablasts were sort-purified from Py-infected mice on day 10 p.i., cultured for 20 hours and parasite lysate-specific IgM and IgG secreting ASCs were detected. Representative wells of ELISPOT assay. d, Relative CD19 expression by CD138hiIgDneg plasmablasts on days 7, 10, and 14 post Py infection. Data are representative of n > 5 experiments with similar results. e, BrdU incorporation in CD138hiIgDneg plasmablasts was assessed on day 10 p.i. Histogram represents BrdU staining, solid gray histogram is isotype (mouse IgG1). Data are representative of n = 2 experiments with similar results using n = 8 mice. f, Forward scatter and side scatter of CD138hiIgDneg plasmablasts examined on day 10 p.i. Data are representative of n > 5 experiments with similar results. g, Blimp-1/eYFP reporter mice were infected with Py. CD138hi Blimp-1/eYFP+ cells in bone marrow from naïve (left panel) or day 21 infected mice (right panel). Data are representative of n = 2 independent experiments with similar results using n = 4 mice/group. h,i, CD21 and CD23 expression by B cells in a naïve mouse (h) and day 10 Py-infected mouse (i) showing plasmablasts (green box) activated (blue box) and resting (red box) B cells. Data are representative of n > 5 experiments with similar results using n = 3 mice/group. j, Representative plots of the frequency and total numbers of GC B cells (GL7+CD95+) among plasmablasts, activated and resting B cell populations on day 10 p.i. Data are means ± s.e.m., representative of n = 3 experiments with similar results using n = 5 (Day 0 Total B cells) and n = 4 mice (each remaining group).

Source data

Extended Data Fig. 2 Developmental abrogation of blood stage Plasmodium infection-induced plasmablast responses.

a, Experimental design for adoptive transfers. b, Experimental design for Rosa26-ERT2/Cre × Prdm1fl/fl (CD45.2) : μMT bone marrow chimeric system. Eight weeks after engraftment, mice were infected with 1 × 106 Py and then treated with either corn oil or tamoxifen on days 4, 5, and 6 p.i. c, Gating strategy of TFH cells. d, Kinetics of parasite burden in Py-infected wild-type mice treated with tamoxifen or corn oil on days 4, 5, and 6 p.i. Data are means ± s.e.m., pooled from n = 2 biologically independent experiments with n = 6 mice/group. e, Experimental design for the three-way mixed bone marrow chimera. f, Eight weeks after engraftment, mice were infected with Py, treated with either corn oil or tamoxifen on days 4, 5, and 6 p.i. and the relative proportions (pie diagram) of Prdm1fl/fl (CD45.2) and wild-type (CD45.1) cells in the GC B cell compartment was analyzed. Data are means ± s.e.m., pooled from n = 2 biologically independent experiments with n = 4 mice/group. g, Evaluation of TH1 responses in PBS and tamoxifen-treated mixed bone marrow chimeric mice (as shown in b). Data are means ± s.d. representative of n = 2 biologically independent experiments with similar results using n = 5 (corn oil) and n = 4 mice (tamoxifen). Data in f,g were analyzed using two-tailed Mann-Whitney. Symbols and symbols represent individual mice.

Source data

Extended Data Fig. 3 Deletion of blood stage Plasmodium infection-induced plasmablast responses.

a, Experimental design for generating CD138-DTR chimeras. Eight weeks after engraftment, mice were infected with 1 × 106 Py and on days 5 and 7 p.i. treated with either DTx or PBS to delete plasmablasts. Data are means ± s.d., representative of n = 2 independent experiments with similar results using n = 4 mice/group. b-d, Py-infected wild-type mice were treated with either DTx or PBS on days 5 and 7 p.i. Kinetics of parasite burden (b), representative plots and summary data of GC B cells (c) and GC-Tfh cells (d) on 21 p.i. Data are means ± s.d., representative of n = 2 biologically independent experiments with similar results using n = 4 (PBS) and n = 3 mice (DTx). e-g, CD138-DTR chimeric mice were infected with 1 × 106 Py, plasmablasts were deleted with DTx and mice were subsequently treated with either MR-1 (anti-CD40L) or hamster IgG on days 8–11 p.i. Representative plots and summary data of GC B (e) and GC-TFH (f) cells as measured on day 21 p.i. and kinetics of parasite burden (g). Data are means ± s.e.m., pooled from n = 2 biologically independent experiments with n = 6 mice/group. Symbols in c-f represent individual mice and data were analyzed using two-tailed Mann-Whitney.

Source data

Extended Data Fig. 4 Differentially expressed genes among splenic B cell populations.

a, Venn diagram showing differentially expressed genes assessed by RNA-seq among the three splenic B cell populations on day 10 p.i. Respective cell types were sort-purified from n = 4 Py-infected mice. Data were obtained from one RNA-Seq experiment. Two-tailed ANOVA was used for identifying differentially expressed genes. b, Heat map showing the relative expression of all annotated genes assessed using RNA-Seq. c, Heat map showing the relative expression of genes involved in the unfolded protein response pathway (UPR).

Extended Data Fig. 5 L-glutamine enhances GC responses during experimental malaria.

a, L-glut concentrations in the spleens of naive and Py-infected wild-type mice on day 5 p.i. Data are means ± s.d. representative of n = 2 biologically independent experiments with similar results using n = 3 mice/group analyzed by a two-tailed unpaired t test (DF = 4; t = 5.933). b, c, Kinetics of parasite burden in mice treated with either L-alanine (b, H2O + L-ala), L-valine (c, H2O + L-val) or water starting on day 0 p.i. Data are means ± s.d., representative of n = 2 independent experiments with similar results using n = 3 mice/group. df, Py-infected wild-type mice were treated with L-glutamine (H2O + L-glut) or water starting day 0 p.i. and subsequently treated with MR-1 (anti-CD40L) or Hamster IgG antibody on days 8–11 p.i. Kinetics of parasite burden (d) and frequency of GC B cells (e) and GC-Tfh cells (f) on day 21 p.i. Data in d are means ± s.e.m, pooled from n = 2 independent experiments with n = 7 mice/group. Data in e,f are representative of n = 2 biologically independent experiments with similar results using n = 3 mice/group. g-m, Py-infected wild-type mice were treated with L-glutamine (H2O + L-glut) or water starting day 0 p.i. Kinetics of GC B cells (g), class-switched GC B cells (h), plasmablasts (i), and GC-TFH-like cells (j). Data in g-j are means ± s.d., representative of n = 2 biologically independent experiments with similar results using n = 6 mice/group analyzed by two-tailed Mann-Whitney. k, gMFI of BCL6 on GC-TFH-like cells. Data are means ± s.d., representative of n = 2 biologically independent experiments with similar results using n = 3 mice/group analyzed by two tailed unpaired t test (DF = 4; t = 1.257). Number of TH1 cells (l, Ly6C+CXCR3+IFNg+) and gMFI of CD80 and CD86 expression on splenic dendritic cells (m, MHCII+CD11c+) on day 10 p.i. Data in l,m are means ± s.e.m, pooled from n = 2 biologically independent experiments with n = 6 (H2O) and n = 7 mice (H2O + L-glut) analyzed by two-tailed Mann-Whitney. n,o, Kinetics of parasite burden (n) and area under curve as a measure of total parasite biomass (o) in Py-infected wild-type mice treated with L-glutamine (H2O + L-glut) starting on day 6 p.i. Data in n are means ± s.d., representative of n = 2 biologically independent experiments with similar results using n = 4 mice/group. Data in n analyzed using two-way ANOVA with Sidak’s multiple comparison (DF = 5; F = 5.728). Data in o analyzed with two-tailed Mann Whitney. p,q, Kinetics of parasite burden (p) and area under curve (q) as a measure of total parasite biomass in Py-infected wild-type mice treated with L-glutamine (H2O + L-glut) starting day on 10 p.i. Data in p,q are means ± s.d., representative of n = 2 independent experiments with similar results using n = 4 mice/group analyzed by two-tailed Mann Whitney. Symbols in a, l-q represent individual mice.

Source data

Extended Data Fig. 6 Experimental design for treating CD138-DTR: WT chimeras with L-glutamine.

Eight weeks after engraftment, mice were infected with Py and treated with L-glut or water starting on day 0 p.i. Mice were subsequently treated with either DTx or PBS on days 5 and 7 p.i. to delete plasmablasts.

Extended Data Fig. 7 Post-GC administration of L-glutamine does not appreciably enhance LLPC and MBC responses.

a, Gating strategy for long lived plasma cells (LLPCs) in the bone marrow of Py-infected mice. b, Numbers of LLPCs in the bone marrow on day 60 p.i. from Py-infected mice treated with either L-glutamine (H2O + L-glut) or water (H2O). Data are means ± s.e.m., pooled from n = 2 biologically independent experiments with n = 7 mice/group analyzed by two-tailed Mann Whitney. c,d, Py-infected wild-type mice were treated with either L-glutamine (H2O + L-glut) or water (H2O) at indicated time points p.i. and analyzed on day 30 p.i. Numbers of splenic memory B cells (c, CCR6+CD38+) and representative ELISPOT wells and summary data (d) demonstrating number of antibody-secreting LLPCs in the bone marrow. Data in c,d are means ± s.e.m, pooled from n = 2 biologically independent experiments with n = 5 (d, days 6–10) and n = 6 mice (remaining groups) analyzed by two-tailed Mann Whitney. Symbols represent individual mice.

Source data

Extended Data Fig. 8 Experimental design for the volunteer infection study.

Two thousand eight hundred viable P. falciparum infected RBCs were intravenously into malaria naïve healthy volunteers (n = 36 men, n = 4 women). On day 8 p.i., volunteers received anti-malarial drug treatment. On days 0, 8 (treatment day), 14/15 and 27/28/36 (end-of-study, EOS), blood samples were collected and plasmablasts were assessed by flow cytometry.

Supplementary information

Reporting Summary

Supplementary Tables

Supplementary Tables 1–6.

Source data

Source Data Fig. 1

Statistical Source Data.

Source Data Fig. 2

Statistical Source Data.

Source Data Fig. 3

Statistical Source Data.

Source Data Fig. 4

Statistical Source Data.

Source Data Fig. 5

Statistical Source Data.

Source Data Fig. 6

Statistical Source Data.

Source Data Fig. 7

Statistical Source Data.

Source Data Fig. 8

Statistical Source Data.

Source Data Extended Data Fig. 1

Statistical Source Data.

Source Data Extended Data Fig. 2

Statistical Source Data.

Source Data Extended Data Fig. 3

Statistical Source Data.

Source Data Extended Data Fig. 5

Statistical Source Data.

Source Data Extended Data Fig. 7

Statistical Source Data.

Rights and permissions

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Vijay, R., Guthmiller, J.J., Sturtz, A.J. et al. Infection-induced plasmablasts are a nutrient sink that impairs humoral immunity to malaria. Nat Immunol 21, 790–801 (2020). https://doi.org/10.1038/s41590-020-0678-5

Download citation

  • Received:

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1038/s41590-020-0678-5

This article is cited by

Search

Quick links

Nature Briefing

Sign up for the Nature Briefing newsletter — what matters in science, free to your inbox daily.

Get the most important science stories of the day, free in your inbox. Sign up for Nature Briefing