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:

PI3Kγ in B cells promotes antibody responses and generation of antibody-secreting cells

Abstract

The differentiation of naive and memory B cells into antibody-secreting cells (ASCs) is a key feature of adaptive immunity. The requirement for phosphoinositide 3-kinase-delta (PI3Kδ) to support B cell biology has been investigated intensively; however, specific functions of the related phosphoinositide 3-kinase-gamma (PI3Kγ) complex in B lineage cells have not. In the present study, we report that PI3Kγ promotes robust antibody responses induced by T cell-dependent antigens. The inborn error of immunity caused by human deficiency in PI3Kγ results in broad humoral defects, prompting our investigation of roles for this kinase in antibody responses. Using mouse immunization models, we found that PI3Kγ functions cell intrinsically within activated B cells in a kinase activity-dependent manner to transduce signals required for the transcriptional program supporting differentiation of ASCs. Furthermore, ASC fate choice coincides with upregulation of PIK3CG expression and is impaired in the context of PI3Kγ disruption in naive B cells on in vitro CD40-/cytokine-driven activation, in memory B cells on toll-like receptor activation, or in human tonsillar organoids. Taken together, our study uncovers a fundamental role for PI3Kγ in supporting humoral immunity by integrating signals instructing commitment to the ASC fate.

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: PI3Kγ is required for T cell-dependent antibody responses.
Fig. 2: PI3Kγ is dispensable for Ig class switching and somatic mutation frequency but required for GC B cells to adopt ASC gene signatures.
Fig. 3: Loss of B cell-intrinsic PI3Kγ is sufficient to impair specific IgG responses with no effect on overall IgA production.
Fig. 4: PI3Kγ functions in activated GC B cells to promote generation of ASCs.
Fig. 5: Human B cells require PI3Kγ for PIP3-dependent primary and secondary ASC formation via IRF4 induction.

Similar content being viewed by others

Data availability

Bulk RNA-seq and scRNA-seq and BCR-seq data are available via the Gene Expression Omnibus (GEO) and were analyzed using mm10 reference genome. The bulk and scRNA-seq data used in the present study are available through the GEO database under accession nos. GSE269303 and GSE269300, respectively. Source data are provided with this paper.

Code availability

Customized scripts in R v.4.3.1 for scBCR analysis were deposited under bitbucket.org/kleinstein/projects (Lanahan2023 folder).

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. Brandtzaeg, P. et al. Immunobiology and immunopathology of human gut mucosa: humoral immunity and intraepithelial lymphocytes. Gastroenterology 97, 1562–1584 (1989).

    Article  CAS  PubMed  Google Scholar 

  3. van der Heijden, P. J., Stok, W. & Bianchi, A. T. Contribution of immunoglobulin-secreting cells in the murine small intestine to the total ‘background’ immunoglobulin production. Immunology 62, 551–555 (1987).

    PubMed  PubMed Central  Google Scholar 

  4. Gaudette, B. T. et al. Resting innate-like B cells leverage sustained Notch2/mTORC1 signaling to achieve rapid and mitosis-independent plasma cell differentiation. J. Clin. Invest. 131, e151975 (2021).

  5. Skrzypczynska, K. M., Zhu, J. W. & Weiss, A. Positive regulation of Lyn kinase by CD148 is required for B cell receptor signaling in B1 but not B2 B cells. Immunity 45, 1232–1244 (2016).

  6. Gaudette, B. T., Jones, D. D., Bortnick, A., Argon, Y. & Allman, D. mTORC1 coordinates an immediate unfolded protein response-related transcriptome in activated B cells preceding antibody secretion. Nat. Commun. 11, 723 (2020).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  7. Tumang, J. R., Frances, R., Yeo, S. G. & Rothstein, T. L. Spontaneously Ig-secreting B-1 cells violate the accepted paradigm for expression of differentiation-associated transcription factors. J. Immunol. 174, 3173–3177 (2005).

    Article  CAS  PubMed  Google Scholar 

  8. 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 

  9. Klein, U. et al. Transcription factor IRF4 controls plasma cell differentiation and class-switch recombination. Nat. Immunol. 7, 773–782 (2006).

    Article  CAS  PubMed  Google Scholar 

  10. 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 

  11. Carotta, S. et al. The transcription factors IRF8 and PU.1 negatively regulate plasma cell differentiation. J. Exp. Med. 211, 2169–2181 (2014).

    Article  PubMed  PubMed Central  Google Scholar 

  12. Scharer, C. D. et al. Antibody-secreting cell destiny emerges during the initial stages of B-cell activation. Nat. Commun. 11, 3989 (2020).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  13. Jones, D. D. et al. mTOR has distinct functions in generating versus sustaining humoral immunity. J. Clin. Invest. 126, 4250–4261 (2016).

    Article  PubMed  PubMed Central  Google Scholar 

  14. Luo, W. et al. IL-21R signal reprogramming cooperates with CD40 and BCR signals to select and differentiate germinal center B cells. Sci. Immunol. 8, eadd1823 (2023).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  15. Luo, W., Weisel, F. & Shlomchik, M. J. B cell receptor and CD40 signaling are rewired for synergistic induction of the c-Myc transcription factor in germinal center B cells. Immunity 48, 313–326.e315 (2018).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  16. Clayton, E. et al. A crucial role for the p110delta subunit of phosphatidylinositol 3-kinase in B cell development and activation. J. Exp. Med. 196, 753–763 (2002).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  17. Conley, M. E. et al. Agammaglobulinemia and absent B lineage cells in a patient lacking the p85alpha subunit of PI3K. J. Exp. Med. 209, 463–470 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  18. Coulter, T. I. et al. Clinical spectrum and features of activated phosphoinositide 3-kinase delta syndrome: A large patient cohort study. J. Allergy Clin. Immunol. 139, 597–606.e594 (2017).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  19. Bader, A. G., Kang, S., Zhao, L. & Vogt, P. K. Oncogenic PI3K deregulates transcription and translation. Nat. Rev. Cancer 5, 921–929 (2005).

    Article  CAS  PubMed  Google Scholar 

  20. Vanhaesebroeck, B., Stephens, L. & Hawkins, P. PI3K signalling: the path to discovery and understanding. Nat. Rev. Mol. Cell Biol. 13, 195–203 (2012).

    Article  CAS  PubMed  Google Scholar 

  21. Burke, J. E. & Williams, R. L. Synergy in activating class I PI3Ks. Trends Biochem. Sci. 40, 88–100 (2015).

    Article  CAS  PubMed  Google Scholar 

  22. Rommel, C., Camps, M. & Ji, H. PI3Kδ and PI3Kγ: partners in crime in inflammation in rheumatoid arthritis and beyond? Nat. Rev. Immunol. 7, 191–201 (2007).

    Article  CAS  PubMed  Google Scholar 

  23. Hirsch, E. et al. Central role for G protein-coupled phosphoinositide 3-kinase gamma in inflammation. Science 287, 1049–1053 (2000).

    Article  CAS  PubMed  Google Scholar 

  24. Sasaki, T. et al. Function of PI3Kgamma in thymocyte development, T cell activation, and neutrophil migration. Science 287, 1040–1046 (2000).

  25. Beer-Hammer, S. et al. The catalytic PI3K isoforms p110gamma and p110delta contribute to B cell development and maintenance, transformation, and proliferation. J. Leukoc. Biol. 87, 1083–1095 (2010).

    Article  CAS  PubMed  Google Scholar 

  26. Reif, K. et al. Cutting edge: differential roles for phosphoinositide 3-kinases, p110gamma and p110delta, in lymphocyte chemotaxis and homing. J. Immunol. 173, 2236–2240 (2004).

  27. Takeda, A. J. et al. Human PI3Kγ deficiency and its microbiota-dependent mouse model reveal immunodeficiency and tissue immunopathology. Nat. Commun. 10, 4364 (2019).

    Article  PubMed  PubMed Central  Google Scholar 

  28. Suire, S. et al. p84, a new Gbetagamma-activated regulatory subunit of the type IB phosphoinositide 3-kinase p110gamma. Curr. Biol. 15, 566–570 (2005).

    Article  CAS  PubMed  Google Scholar 

  29. Stephens, L. R. et al. The G beta gamma sensitivity of a PI3K is dependent upon a tightly associated adaptor, p101. Cell 89, 105–114 (1997).

    Article  CAS  PubMed  Google Scholar 

  30. Allen, D., Simon, T., Sablitzky, F., Rajewsky, K. & Cumano, A. Antibody engineering for the analysis of affinity maturation of an anti-hapten response. EMBO J. 7, 1995–2001 (1988).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  31. Elsner, R. A. & Shlomchik, M. J. Germinal center and extrafollicular B cell responses in vaccination, immunity, and autoimmunity. Immunity 53, 1136–1150 (2020).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  32. Casola, S. et al. Tracking germinal center B cells expressing germ-line immunoglobulin gamma1 transcripts by conditional gene targeting. Proc. Natl Acad. Sci. USA 103, 7396–7401 (2006).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  33. Breasson, L. et al. PI3Kγ activity in leukocytes promotes adipose tissue inflammation and early-onset insulin resistance during obesity. Sci. Signal. 10, eaaf2969 (2017).

  34. Nojima, T. et al. In-vitro derived germinal centre B cells differentially generate memory B or plasma cells in vivo. Nat. Commun. 2, 465 (2011).

    Article  PubMed  Google Scholar 

  35. Monaco, G. et al. RNA-seq signatures normalized by mRNA abundance allow absolute deconvolution of human immune cell types. Cell Rep. 26, 1627–1640 e1627 (2019).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  36. Lanahan, S. M., Wymann, M. P. & Lucas, C. L. The role of PI3Kγ in the immune system: new insights and translational implications. Nat. Rev. Immunol. 22, 687–700 (2022).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  37. Heng, T. S., Painter, M. W. & Immunological Genome Project The Immunological Genome Project: networks of gene expression in immune cells. Nat. Immunol. 9, 1091–1094 (2008).

    Article  CAS  PubMed  Google Scholar 

  38. Pinto, D. et al. A functional BCR in human IgA and IgM plasma cells. Blood 121, 4110–4114 (2013).

    Article  CAS  PubMed  Google Scholar 

  39. Wagar, L. E. et al. Modeling human adaptive immune responses with tonsil organoids. Nat. Med. 27, 125–135 (2021).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  40. Le Coz, C. et al. Human T follicular helper clones seed the germinal center-resident regulatory pool. Sci. Immunol. 8, eade8162 (2023).

    Article  PubMed  PubMed Central  Google Scholar 

  41. Bekeredjian-Ding, I. & Jego, G. Toll-like receptors—sentries in the B-cell response. Immunology 128, 311–323 (2009).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  42. Gong, G. Q. et al. A small-molecule PI3Kα activator for cardioprotection and neuroregeneration. Nature 618, 159–168 (2023).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  43. Donahue, A. C. & Fruman, D. A. Distinct signaling mechanisms activate the target of rapamycin in response to different B-cell stimuli. Eur. J. Immunol. 37, 2923–2936 (2007).

    Article  CAS  PubMed  Google Scholar 

  44. Thian, M. et al. Germline biallelic PIK3CG mutations in a multifaceted immunodeficiency with immune dysregulation. Haematologica 105, e448–e492 (2020).

    PubMed Central  Google Scholar 

  45. Camps, M. et al. Blockade of PI3Kgamma suppresses joint inflammation and damage in mouse models of rheumatoid arthritis. Nat. Med. 11, 936–943 (2005).

    Article  CAS  PubMed  Google Scholar 

  46. Barber, D. F. et al. PI3Kgamma inhibition blocks glomerulonephritis and extends lifespan in a mouse model of systemic lupus. Nat. Med. 11, 933–935 (2005).

    Article  CAS  PubMed  Google Scholar 

  47. Del Prete, A. et al. Defective dendritic cell migration and activation of adaptive immunity in PI3Kγ-deficient mice. EMBO J. 23, 3505–3515 (2004).

    Article  PubMed  PubMed Central  Google Scholar 

  48. Nobs, S. P. et al. PI3Kγ is critical for dendritic cell-mediated CD8+ T cell priming and viral clearance during influenza virus infection. PLoS Pathog. 12, e1005508 (2016).

    Article  PubMed  PubMed Central  Google Scholar 

  49. Laidlaw, B. J. et al. The Eph-related tyrosine kinase ligand Ephrin-B1 marks germinal center and memory precursor B cells. J. Exp. Med. 214, 639–649 (2017).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  50. Rossbacher, J. & Shlomchik, M. J. The B cell receptor itself can activate complement to provide the complement receptor 1/2 ligand required to enhance B cell immune responses in vivo. J. Exp. Med. 198, 591–602 (2003).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  51. Luo, W. et al. SREBP signaling is essential for effective B cell responses. Nat. Immunol. 24, 337–348 (2023).

    Article  CAS  PubMed  Google Scholar 

  52. Study to Evaluate the Efficacy/Safety of IPI-549 in Combination With Nivolumab in Patients With Advanced Urothelial Carcinoma (MARIO-275). NIH https://clinicaltrials.gov/ct2/show/NCT03980041 (2019).

  53. Evaluation of IPI-549 Combined With Front-line Treatments in Pts. With Triple-Negative Breast Cancer or Renal Cell Carcinoma (MARIO-3). NIH https://clinicaltrials.gov/ct2/show/NCT03961698 (2019).

  54. Evans, C. A. et al. Discovery of a selective phosphoinositide-3-kinase (PI3K)-γ inhibitor (IPI-549) as an immuno-oncology clinical candidate. ACS Med. Chem. Lett. 7, 862–867 (2016).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  55. Haniuda, K. & Kitamura, D. Induced germinal center B cell culture system. Bio Protoc. 9, e3163 (2019).

    Article  PubMed  PubMed Central  Google Scholar 

  56. Pahl, M. C. et al. Implicating effector genes at COVID-19 GWAS loci using promoter-focused Capture-C in disease-relevant immune cell types. Genome Biol. 23, 125 (2022).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  57. Ramaswamy, A. et al. Immune dysregulation and autoreactivity correlate with disease severity in SARS-CoV-2-associated multisystem inflammatory syndrome in children. Immunity 54, 1083–1095.e1087 (2021).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  58. Barmada, A. et al. Cytokinopathy with aberrant cytotoxic lymphocytes and profibrotic myeloid response in SARS-CoV-2 mRNA vaccine-associated myocarditis. Sci. Immunol. 8, eadh3455 (2023).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  59. Lopez, R., Regier, J., Cole, M. B., Jordan, M. I. & Yosef, N. Deep generative modeling for single-cell transcriptomics. Nat. Methods 15, 1053–1058 (2018).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  60. Wolf, F. A., Angerer, P. & Theis, F. J. SCANPY: large-scale single-cell gene expression data analysis. Genome Biol. 19, 15 (2018).

    Article  PubMed  PubMed Central  Google Scholar 

  61. Ye, J., Ma, N., Madden, T. L. & Ostell, J. M. IgBLAST: an immunoglobulin variable domain sequence analysis tool. Nucleic Acids Res. 41, W34–40 (2013).

    Article  PubMed  PubMed Central  Google Scholar 

  62. Lefranc, M. P. et al. IMGT, the international ImMunoGeneTics information system. Nucleic Acids Res. 37, D1006–1012 (2009).

    Article  CAS  PubMed  Google Scholar 

  63. Alsoussi, W. B. et al. A potently neutralizing antibody protects mice against SARS-CoV-2 infection. J. Immunol. 205, 915–922 (2020).

    Article  CAS  PubMed  Google Scholar 

  64. Gupta, N. T. et al. Change-O: a toolkit for analyzing large-scale B cell immunoglobulin repertoire sequencing data. Bioinformatics 31, 3356–3358 (2015).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  65. Nouri, N. & Kleinstein, S. H. A spectral clustering-based method for identifying clones from high-throughput B cell repertoire sequencing data. Bioinformatics 34, i341–i349 (2018).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  66. Stern, J. N. et al. B cells populating the multiple sclerosis brain mature in the draining cervical lymph nodes. Sci. Transl. Med. 6, 248ra107 (2014).

    Article  PubMed  PubMed Central  Google Scholar 

Download references

Acknowledgements

We thank the patients and their families for participation in research and all clinical care staff for their contributions. We thank L. Wirth for technical assistance, J.-M. Carpier for critical feedback and training, E. Fagerberg and J. Attanasio for contributions to RNA-seq experimental design and A. Rice for his contributions. We acknowledge D. Kitamura, Tokyo University of Science, Organization for Research Advancement, Research Institute for Biomedical Sciences for the 40LB feeder cell line used in Extended Data Fig. 3. C.L.L. received funding for this work from the Colton Center for Autoimmunity at Yale, NIH/NIAID (grant no. R21AI144315) and Yale University. M.P.W. was funded by the Swiss National Science Foundation (SNF; grant no. 310030_189065). N.R. is funded by the NIH, NIAID (grant no. AI146026 to N.R.), the Chan Zuckerberg Initiative Pediatric Human Cell Atlas and the Jeffrey Modell Foundation. BioRender.com was used for schematics in Extended Data Fig. 2h and Extended Data Fig. 6.

Author information

Authors and Affiliations

Authors

Contributions

S.M.L. was involved in idea generation, performed experiments, analyzed data and wrote the manuscript. L.Y. and K.M.J. performed experiments, analyzed data and wrote methods for their work. Z.Q., D.B.U.K., L.X. and P.S. conducted experiments and analyses. E.C.C., L.Y.C. and N.R. conducted and analyzed the tonsillar organoid experiments. A.R., A.B., G.G. and S.H.K. performed analysis of single-cell transcriptome and BCR-seq. K.R., P.M. and R.A. provided patient care and analyses relating to patient A.1’s samples. M.P.W. provided KO and floxed mice and input. C.L.L. supervised overall research and data analysis, was involved in idea generation and wrote and edited the manuscript. All authors discussed and reviewed the manuscript.

Corresponding author

Correspondence to Carrie L. Lucas.

Ethics declarations

Competing interests

C.L.L. reports an advisory/consulting role for Pharming Healthcare Inc. and unrelated funding support from Ono Pharma. S.H.K. receives consulting fees from Peraton. R.S.A. discloses support from ClinGen, Journal of Immunology and Clinical Laboratory Standards Institute. The remaining authors declare no competing interests.

Peer review

Peer review information

Nature Immunology thanks Sidonia Fagarasan and the other, anonymous, reviewer(s) for their contribution to the peer review of this work. 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.

Additional information

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

Extended data

Extended Data Fig. 1 PI3Kγ is required for T-dependent antibody responses.

(a-b) Serum IgM (a) and IgA (b) levels in Patient A.1. (c-e) Mice were immunized (as in Fig. 1b, c) and antigen-specific IgM (day 5) (c), primary (non-boosted) IgG1 (day 12) (d), or IgG2c (e) was measured via high conjugation ratio NP-BSA ELISA. (f) Mice were immunized (as in Fig. 1d, e) and IgG2b was measured via NP-BSA ELISA. Data are from 2-3 independent experiments and are presented as box-and-whisker plots showing median, min, and max. Data points represent different biological samples. Statistical analysis was performed using non-parametric two-tailed unpaired T-tests. (c) n = 12 for control, n = 12 for KO. (d) n = 15 for control, n = 14 for KO. (e) n = 12 for control, n = 11 for KO. (f) n = 5 for control, n = 6 for KO.

Source data

Extended Data Fig. 2 PI3Kγ is required for GC B cells to adopt ASC gene signatures.

(a) Stacked bar plots showing proportions of cell subsets within Naïve or GC B cells used for CSR/SHM analysis. (b) Percentage of high-affinity W38L mutations from the total number of sequences with IGHV1-72 gene and lambda light chains. Statistical analysis was Wilcoxon Rank Sum (c-d) Quantification of marginal zone B cells (c) and follicular B cells (d) from the spleens of indicated mice assessed by flow cytometry. Data are representative of 2 independent experiments (n = 5 for each condition). (e) Volcano plot showing gene expression from RNA sequencing in sorted naïve B cells (IgD+ B220+) from WT versus PI3Kγ KO mice 28 days after SRBC i.p. immunization. Data are from one experiment. (f) Volcano plot of differentially expressed genes from bulk RNA sequencing of sorted GC B cells at day 28 post immunization, as described for Fig. 2e. (g) Expression of ‘Activated B cell’ UPR program genes in sorted splenic GC B cells from Pik3cg heterozygous or KO mice 28 days after SRBC immunization. Transcriptional program is defined by Gaudette, et. al 2020. Data are from one experiment (n = 3 for het, n = 4 for KO). (h) Diagram of germinal center B cells that are beginning to adopt expression of antibody secreting cell surface markers (CD138). Diagram was created with BioRender.com. (c-d) Each dot represents different biological samples and data are presented as box-and-whisker plots showing median, min, and max. Statistical analysis was performed using non-parametric two-tailed unpaired T-tests.

Source data

Extended Data Fig. 3 B cells lacking PI3Kγ activate normally and are capable of forming germinal centers and normal IgA responses.

(a) Littermate control or conditional Pik3cg KO mice were immunized and boosted with NP-OVA precipitated in alum. Antigen-specific IgG2c (day 28) was measured via NP-BSA ELISA, and arbitrary units were defined using pooled immunized sera as a standard. (b) Immunofluorescence imaging of spleens from indicated mice 28 days after SRBC immunization. Data are representative of two independent repeats (n = 6 for each genotype). (c-h) Murine naïve B cells were stimulated using TI stimulation with TLR agonists (CpG or LPS) or TD stimulation with anti-CD40, anti-IgM F(ab’)2 and assessed for proliferation using cell trace violet (gating on live cells after 4 days) and activation by staining for CD69 or CD86 (gating on live cells after 2 days). Data (n = 3 for each condition) are representative of two independent experiments. (i-n) Quantification of B cell subsets from Peyer’s patches of mice assessed by flow cytometry. Data are representative of three independent experiments (n = 13 for control and 14 for KO). (o) Quantification of fecal IgA in control versus B cell-specific PI3Kγ KO mice (n = 6 per group). (p) Quantification of surface IgG-expressing memory and GC B cells, normalized to control animals, in control versus B cell-specific PI3Kγ KO mice (n = 3 for control and 5 for KO). (q-r) In-vitro generation of GC B cells using 40LB feeder cells, as in Nojima et al. 2011. Data are from two independent experiments (n = 5 for each condition). Data are presented as a bar graph (median ± SD) or box-and-whisker plots showing median, min, and max. Each dot represents different biological samples and statistical analysis was performed using non-parametric two-tailed unpaired T-tests, except for panels q-r which uses non-parametric paired T-test.

Source data

Extended Data Fig. 4 Functional PI3Kγ is required for optimal ASC differentiation in vitro and in vivo despite being dispensable for innate-like B cell IgM responses.

(a) Expression of PI3K genes in human/mouse immune cells derived from Immgen3. (b) Expression of Pik3cg in mouse B cells comparing splenic follicular and germinal center B cells to innate-like marginal zone and B1a B cells. (c-d) Mice were immunized i.p. with TI antigens and serum was taken on day 5 to measure antigen-specific IgM via ELISA. (e-f) Antigen-specific ASCs were measured from splenocytes via ELISPOT in WT mice treated as described for Fig. 4e, f. (g-h) Assessment of in-vitro differentiation of naïve mouse B cells into ASCs (anti-CD40, IL-4, IL-5 gating on CD138+ after 4 days) in utilizing B cells from Blimp1-YFP mice treated in vitro with DMSO vehicle or the PI3Kγ inhibitor IPI-549. Data are from 2-4 independent experiments. Data presented as box-and-whisker plots show median, min, and max. (c) n = 11 for controls, n = 12 for KOs. (d) n = 12 for both groups. (e) n = 15 for both groups. (f) n = 13 for both. (g-h) n = 6 for each group. Each dot represents different biological samples and statistical analysis was performed using non-parametric two-tailed unpaired T-tests, with the exception of panel H which used non-parametric paired T-test.

Source data

Extended Data Fig. 5 PI3Kγ is required for robust human ASC differentiation.

(a-h) Cells from human tonsillar organoid model were assessed via flow cytometry (n = 5). Graphs depict percentage of each subset or percentage of live cells in each subset. (i) Assessment of in-vitro differentiation of primary healthy human peripheral blood pan-B cells into IRF4+ cells with 100 nM dose of PI3Kγi IPI-549 versus DMSO (shown as a proportion) after 5 days in the presence of DMSO control or PI3Kα activator (UCL-TRO-1938). (j-k) Overnight IPI-549 treatment of isolated primary human CD138+ ASCs from peripheral blood. ELISPOTs and quantification are from 2 independent experiments (n = 6). Each dot represents different biological samples and data are presented as mean ± SD. Statistical analysis was performed using non-parametric two-tailed unpaired T-tests and paired T-test for (i).

Source data

Extended Data Fig. 6 Model summarizing the function of PI3Kγ in antibody responses.

B cell-intrinsic PI3Kγ supports the transition to the ASC transcriptional program and ASC differentiation in activated B cells. Diagram was created with BioRender.com.

Supplementary information

Supplementary Information

Patient A.1 antibody titers elicited by indicated vaccines.

Reporting Summary

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 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. 4

Statistical source data.

Source Data Extended Data Fig. 5

Statistical source data.

Rights and permissions

Springer Nature or its licensor (e.g. a society or other partner) holds exclusive rights to this article under a publishing agreement with the author(s) or other rightsholder(s); author self-archiving of the accepted manuscript version of this article is solely governed by the terms of such publishing agreement and applicable law.

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Lanahan, S.M., Yang, L., Jones, K.M. et al. PI3Kγ in B cells promotes antibody responses and generation of antibody-secreting cells. Nat Immunol (2024). https://doi.org/10.1038/s41590-024-01890-1

Download citation

  • Received:

  • Accepted:

  • Published:

  • DOI: https://doi.org/10.1038/s41590-024-01890-1

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