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:

Germinal center B cells selectively oxidize fatty acids for energy while conducting minimal glycolysis

Abstract

Germinal center B cells (GCBCs) are critical for generating long-lived humoral immunity. How GCBCs meet the energetic challenge of rapid proliferation is poorly understood. Dividing lymphocytes typically rely on aerobic glycolysis over oxidative phosphorylation for energy. Here we report that GCBCs are exceptional among proliferating B and T cells, as they actively oxidize fatty acids (FAs) and conduct minimal glycolysis. In vitro, GCBCs had a very low glycolytic extracellular acidification rate but consumed oxygen in response to FAs. [13C6]-glucose feeding revealed that GCBCs generate significantly less phosphorylated glucose and little lactate. Further, GCBCs did not metabolize glucose into tricarboxylic acid (TCA) cycle intermediates. Conversely, [13C16]-palmitic acid labeling demonstrated that GCBCs generate most of their acetyl-CoA and acetylcarnitine from FAs. FA oxidation was functionally important, as drug-mediated and genetic dampening of FA oxidation resulted in a selective reduction of GCBCs. Hence, GCBCs appear to uncouple rapid proliferation from aerobic glycolysis.

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: GCBCs perform OXPHOS but not aerobic glycolysis.
Fig. 2: GCBCs rely on FAO.
Fig. 3: GCBC uptake of glucose and FFA.
Fig. 4: GCBCs upregulate and rely on peroxisomal FAO for oxygen consumption.
Fig. 5: GCBCs are sensitive to dual mitochondrial and peroxisomal FAO in vitro and in vivo.
Fig. 6: 13C carbon tracing of glucose and palmitate in cultured B cells by LC–HRMS.
Fig. 7: Comparison of GCBCs and activated B cells for glycolysis-relevant gene expression.
Fig. 8: Competitive disadvantage of GCBCs after targeted CPT2 mRNA reduction.

Similar content being viewed by others

Data availability

The data that support the findings of this study are available from the corresponding author upon request. RNA-sequencing data have been deposited in the Gene Expression Omnibus with the accession code GSE128710.

References

  1. Herrera, E., Martínez-A, C. & Blasco, M. A. Impaired germinal center reaction in mice with short telomeres. EMBO J. 19, 472–481 (2000).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  2. Raso, F. et al. αv integrins regulate germinal center B cell responses through noncanonical autophagy. J. Clin. Invest. 128, 4163–4178 (2018).

    Article  PubMed  PubMed Central  Google Scholar 

  3. Allen, C. D. C., Okada, T. & Cyster, J. G. Germinal-center organization and cellular dynamics. Immunity 27, 190–202 (2007).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  4. Shlomchik, M. J. & Weisel, F. B cell primary immune responses. Immunol. Rev. 288, 5–9 (2019).

    Article  CAS  PubMed  Google Scholar 

  5. Ganeshan, K. & Chawla, A. Metabolic regulation of immune responses. Annu. Rev. Immunol. 32, 609–634 (2014).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  6. Jung, J., Zeng, H. & Horng, T. Metabolism as a guiding force for immunity. Nat. Cell Biol. 21, 85–93 (2019).

    Article  CAS  PubMed  Google Scholar 

  7. Pearce, E. L. & Pearce, E. J. Metabolic pathways in immune cell activation and quiescence. Immunity 38, 633–643 (2013).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  8. Jang, K.-J. et al. Mitochondrial function provides instructive signals for activation-induced B-cell fates. Nat. Commun. 6, 6750 (2015).

    Article  CAS  PubMed  Google Scholar 

  9. Weinberg, S. E., Sena, L. A. & Chandel, N. S. Mitochondria in the regulation of innate and adaptive immunity. Immunity 42, 406–417 (2015).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  10. Waters, L. R., Ahsan, F. M., Wolf, D. M., Shirihai, O. & Teitell, M. A. Initial B cell activation induces metabolic reprogramming and mitochondrial remodeling. iScience 5, 99–109 (2018).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  11. Vander Heiden, M. G., Cantley, L. C. & Thompson, C. B. Understanding the Warburg effect: the metabolic requirements of cell proliferation. Science 324, 1029–1033 (2009).

    Article  CAS  Google Scholar 

  12. Fox, C. J., Hammerman, P. S. & Thompson, C. B. Fuel feeds function: energy metabolism and the T-cell response. Nat. Rev. Immunol. 5, 844–852 (2005).

    Article  CAS  PubMed  Google Scholar 

  13. Jellusova, J. et al. Gsk3 is a metabolic checkpoint regulator in B cells. Nat. Immunol. 18, 303–312 (2017).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  14. Cho, S. H. et al. Germinal centre hypoxia and regulation of antibody qualities by a hypoxia response system. Nature 537, 234–238 (2016).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  15. Eales, K. L., Hollinshead, K. E. R. & Tennant, D. A. Hypoxia and metabolic adaptation of cancer cells. Oncogenesis 5, e190 (2016).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  16. Shi, W. et al. Transcriptional profiling of mouse B cell terminal differentiation defines a signature for antibody-secreting plasma cells. Nat. Immunol. 16, 663–673 (2015).

    Article  CAS  PubMed  Google Scholar 

  17. Shlomchik, M. J. & Weisel, F. Germinal center selection and the development of memory B and plasma cells. Immunol. Rev. 247, 52–63 (2012).

    Article  PubMed  Google Scholar 

  18. van Nierop, K. et al. Lysosomal destabilization contributes to apoptosis of germinal center B-lymphocytes. J. Histochem. Cytochem. 54, 1425–1435 (2006).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  19. Pound, J. D. & Gordon, J. Maintenance of human germinal center B cells in vitro. Blood 89, 919–928 (1997).

    Article  CAS  PubMed  Google Scholar 

  20. Chang, C.-H. et al. Posttranscriptional control of T cell effector function by aerobic glycolysis. Cell 153, 1239–1251 (2013).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  21. Doughty, C. A. Antigen receptor-mediated changes in glucose metabolism in B lymphocytes: role of phosphatidylinositol 3-kinase signaling in the glycolytic control of growth. Blood 107, 4458–4465 (2006).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  22. Deng, J. et al. Homocysteine activates B cells via regulating PKM2-dependent metabolic reprogramming. J. Immunol. 198, 170–183 (2016).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  23. Wolf, H. P. Possible new therapeutic approach in diabetes mellitus by inhibition of carnitine palmitoyltransferase 1 (CPT1). Horm. Metab. Res. Suppl. 26, 62–67 (1992).

    CAS  PubMed  Google Scholar 

  24. Yao, C.-H. et al. Identifying off-target effects of etomoxir reveals that carnitine palmitoyltransferase I is essential for cancer cell proliferation independent of β-oxidation. PLoS Biol. 16, e2003782 (2018).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  25. Reddy, J. K. & Hashimoto, T. Peroxisomal β-oxidation and peroxisome proliferator-activated receptor ɑ: an adaptive metabolic system. Annu. Rev. Nutr. 21, 193–230 (2001).

    Article  CAS  PubMed  Google Scholar 

  26. Van den Branden, C. & Roels, F. Thioridazine: a selective inhibitor of peroxisomal β-oxidation in vivo. FEBS Lett. 187, 331–333 (1985).

    Article  PubMed  Google Scholar 

  27. 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.e5 (2018).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  28. Sun, L., Shukair, S., Naik, T. J., Moazed, F. & Ardehali, H. Glucose phosphorylation and mitochondrial binding are required for the protective effects of hexokinases I and II. Mol. Cell. Biol. 28, 1007–1017 (2008).

    Article  CAS  PubMed  Google Scholar 

  29. Chaudhry, R. & Varacallo, M. in StatPearls (eds Babak, A. et al.) (StatPearls Publishing, 2018).

  30. Pietrocola, F., Galluzzi, L., Bravo-San Pedro, J. M., Madeo, F. & Kroemer, G. Acetyl coenzyme A: a central metabolite and second messenger. Cell Metab. 21, 805–821 (2015).

    Article  CAS  PubMed  Google Scholar 

  31. Thapa, D. et al. Adropin treatment restores cardiac glucose oxidation in pre-diabetic obese mice. J. Mol. Cell. Cardiol. 129, 174–178 (2019).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  32. Abbott, R. K. et al. Germinal center hypoxia potentiates immunoglobulin class switch recombination. J. Immunol. 197, 4014–4020 (2016).

    Article  CAS  PubMed  Google Scholar 

  33. Schodel, J. et al. High-resolution genome-wide mapping of HIF-binding sites by ChIP-seq. Blood 117, e207–e217 (2011).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  34. Taylor, C. T. & Colgan, S. P. Regulation of immunity and inflammation by hypoxia in immunological niches. Nat. Rev. Immunol. 17, 774–785 (2017).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  35. Weisel, F. J., Zuccarino-Catania, G. V., Chikina, M. & Shlomchik, M. J. A temporal switch in the germinal center determines differential output of memory B and plasma cells. Immunity 44, 116–130 (2016).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  36. Zuccarino-Catania, G. V. et al. CD80 and PD-L2 define functionally distinct memory B cell subsets that are independent of antibody isotype. Nat. Immunol. 15, 631–637 (2014).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

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

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  38. Choi, S.-C. et al. Inhibition of glucose metabolism selectively targets autoreactive follicular helper T cells. Nat. Commun. 9, 4369 (2018).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  39. Konigsberg, M. et al. Effect of oxygen tension on bioenergetics and proteostasis in young and old myoblast precursor cells. Redox Biol. 1, 475–482 (2013).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  40. Jacob, J. & Kelsoe, G. In situ studies of the primary immune response to (4-hydroxy-3-nitrophenyl)acetyl. II. A common clonal origin for periarteriolar lymphoid sheath-associated foci and germinal centers. J. Exp. Med. 176, 679–687 (1992).

    Article  CAS  PubMed  Google Scholar 

  41. Tas, J. M. et al. Visualizing antibody affinity maturation in germinal centers. Science 351, 1048–1054 (2016).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  42. Degn, S. E. et al. Clonal evolution of autoreactive germinal centers. Cell 170, 913–926.e19 (2017).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  43. Martinez-Martin, N. et al. A switch from canonical to noncanonical autophagy shapes B cell responses. Science 355, 641–647 (2017).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  44. Oestreich, K. J. et al. Bcl-6 directly represses the gene program of the glycolysis pathway. Nat. Immunol. 15, 957–964 (2014).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  45. LaPensee, C. R., Lin, G., Dent, A. L. & Schwartz, J. Deficiency of the transcriptional repressor B cell lymphoma 6 (Bcl6) is accompanied by dysregulated lipid metabolism. PLoS ONE 9, e97090 (2014).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  46. Wilhelm, K. et al. FOXO1 couples metabolic activity and growth state in the vascular endothelium. Nature 529, 216–220 (2016).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  47. Dominguez-Sola, D. et al. The FOXO1 transcription factor instructs the germinal center dark zone program. Immunity 43, 1064–1074 (2015).

    Article  CAS  PubMed  Google Scholar 

  48. Sander, S. et al. PI3 kinase and FOXO1 transcription factor activity differentially control B cells in the germinal center light and dark zones. Immunity 43, 1075–1086 (2015).

    Article  CAS  PubMed  Google Scholar 

  49. Dominguez-Sola, D. et al. The proto-oncogene MYC is required for selection in the germinal center and cyclic reentry. Nat. Immunol. 13, 1083–1091 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  50. Ersching, J. et al. Germinal center selection and affinity maturation require dynamic regulation of mTORC1 kinase. Immunity 46, 1045–1058.e6 (2017).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  51. Calado, D. P. et al. The cell-cycle regulator c-Myc is essential for the formation and maintenance of germinal centers. Nat. Immunol. 13, 1092–1100 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  52. Shlomchik, M. J., Luo, W. & Weisel, F. Linking signaling and selection in the germinal center. Immunol. Rev. 288, 49–63 (2019).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  53. Pike, L. S., Smift, A. L., Croteau, N. J., Ferrick, D. A. & Wu, M. Inhibition of fatty acid oxidation by etomoxir impairs NADPH production and increases reactive oxygen species resulting in ATP depletion and cell death in human glioblastoma cells. Biochim. Biophys. Acta 1807, 726–734 (2011).

    Article  CAS  PubMed  Google Scholar 

  54. Scharping, N. E. et al. The tumor microenvironment represses T cell mitochondrial biogenesis to drive intratumoral T cell metabolic insufficiency and dysfunction. Immunity 45, 374–388 (2016).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  55. Alves, T. C. et al. Integrated, step-wise, mass-isotopomeric flux analysis of the TCA cycle. Cell Metab. 22, 936–947 (2015).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  56. Chen, R. et al. In vivo RNA interference screens identify regulators of antiviral CD4+ and CD8+ T cell differentiation. Immunity 41, 325–338 (2014).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

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

    Article  CAS  PubMed  Google Scholar 

  58. 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 

  59. Law, C. W., Chen, Y., Shi, W. & Smyth, G. K. voom: precision weights unlock linear model analysis tools for RNA-seq read counts. Genome Biol. 15, R29 (2014).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  60. Ritchie, M. E. et al. limma powers differential expression analyses for RNA-sequencing and microarray studies. Nucleic Acids Res. 43, e47 (2015).

    PubMed  PubMed Central  Google Scholar 

  61. Haynes, N. M. et al. Role of CXCR5 and CCR7 in follicular Th cell positioning and appearance of a programmed cell death gene-1high germinal center-associated subpopulation. J. Immunol. 179, 5099–5108 (2007).

    Article  CAS  PubMed  Google Scholar 

Download references

Acknowledgements

We thank R. Moreci and M. Carter for supporting experimental procedures, and D. Falkner and A. Yates for cell sorting. This work was funded by NIH grant no. R01 AI-46303 and NIH grant no. R01 AI-105018 to M.J.S.; and by grant no. SKF-015-039, grant no. SU2C-IRG-016-08 and start-up funds to G.M.D. through the Tumor Microenvironment Center at the University of Pittsburgh. T.H.W. was funded by the Deutsche Forschungsgemeinschaft (grant no. TRR130) and S.G.W. through grant no. S10OD023402. This work benefitted from ImageStreamX MARKII grant no. NIH 1S10OD019942-01.

Author information

Authors and Affiliations

Authors

Contributions

M.J.S., F.J.W. and G.M.D. designed the research. F.J.W., S.J.M., W.L., D.W., R.A.E., L.J.C., S.M.J., A.V.M., N.T., M.J.J., T.H.W. and G.M.D. performed experiments and analyzed data. W.F.H. and S.G.W. gave conceptual advice. M.C. and S.S. performed computational analysis. F.J.W. and M.J.S. wrote the manuscript.

Corresponding authors

Correspondence to Florian J. Weisel or Mark J. Shlomchik.

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 Preparation of B cell populations in support of Figs. 1, 2, 4, 5, 6, 7 and 8.

Representative FACS plots are shown to determine frequency of indicated target populations (horizontal patterns). Splenocytes were enriched for indicated target population as described in the methods section. After each purification step (columns) cells were subjected to flow cytometric analysis with indicated surface markers to determine purity. Arrows indicate subsequent gating and numbers percent gated population. (RBC; red blood cell).

Extended Data Fig. 2 GCBC show high viability in culture in support of Figs. 1, 2, 4, 5, 6 and 7.

Indicated cell populations were bead purified and cultured either in RPMI media (ac) or Seahorse XF Cell Mito Stress test media (d, e) for depicted times. Cell viability was assayed flow-cytometric staining for 7-AAD (a, d) at 120 min of culture and their viability was additionally determined at 0, 30 and 60 min utilizing Luna-FlTM automated counting with dual fluorescent microscope optics. Cells were exposed to acridine orange (AO) and propidium iodide (PI) simultaneously (b, c, e). Tabulated data are presented in (a) and (b) of 2 independent experiments depicted in red and blue. (b) shows representative images of the Lina-FlTM counter for data in (a). ns = not significant; *p ≤ 0.05; **p ≤ 0.01; ***p ≤ 0.001; ****p ≤ 0.0001 by unpaired, two-tailed t-test.

Extended Data Fig. 3 GCBC maintain key transcriptional profile during in vitro culture in support of Figs. 1, 2, 4, 5 and 6.

Comparison of transcriptional profile of 259 most differentially regulated GC genes. Genes shown are most differentially regulated between freshly isolated GCBC and in vivo activated B cells (FDR < 0.01; FC > 4 log2) and therefore serve as GCBC identifier geneset. a, correlation of GCBC identifier genes of freshly isolated GCBC and GCBC cultured for 2 h (left; R2 = 0.96) which matches the conditions of seahorse experiments or GCBC cultured for 4 h with aCD40 (right; R2 = 0.86) which matches our 13C tracing studies. b, heatmap of expression levels of 267 GCBC identifier genes in freshly isolated (left column) and cultured GCBC under depicted conditions. R-correlation of freshly isolated GCBC to all culture conditions is depicted below each column and was computed using R “cor” function with “pearson” method.

Extended Data Fig. 4 GCBC only take up minimal amounts of glucose but physiological amounts of FFA in support of Fig. 3.

a, tabulated data of mean 2-NBDG fluorescence normalized to cell size (left) and representative flow histograms of 2-NBDG fluorescence (center left) and forward scatter (center right) of indicated cell populations pulsed in vitro for 30 min with 2-NBDG. Right shows an independent repeat of left. b, independent repeat of data presented in Fig. 3a. c, tabulated data of mean CD36 fluorescence normalized to cell size (left) and mean CD36 fluorescence of indicated cell populations. Shown are combined data of 2 independent experiments. d, independent repeat of data presented in Fig. 3b. e, tabulated data of mean BODIPY fluorescence normalized to cell size (left) and representative flow histograms of BODIPY fluorescence (middle) of indicated cell populations, pulsed in vitro for 30 min with BODIPY. Right panel shows an independent repeat of left panel. f, g, representative Amnis ImageStream images of cells presented in Fig. 3c, d, respectively. h, representative images of adaptive erode function for 100% (total cell, left) and 70% (intracellular, right). Fluorescence intensity is only calculated from areas colored in blue of the same cells shown in left and right panels. Bars represent mean + /- SEM; ns = not significant; *p ≤ 0.05; **p ≤ 0.01; ***p ≤ 0.001; ****p ≤ 0.0001 by unpaired, two-tailed t-test.

Extended Data Fig. 5 Combined inhibition of mitochondrial and peroxisomal FAO with increased in vivo dosage of etomoxir and thioridazine in support of Fig. 5.

Absolute number of live splenic NP+ GCBC (a) and naïve NP- B cells (b) and MFI of 2-NBDG of GCBC after 30 min 2-NBDG in vitro pulse (c) of mice at d14 post NP-CGG immunization and in vivo treatment at d9, d11 and d13 with 22 mg/kg etomoxir and 11 mg/kg thioridazine or vehicle only as in Fig. 5c–e. Every dot represents and individual mouse and graphs are mean + /-SEM; ns = not significant; ****p < 0.0001 by unpaired, two-tailed t-test.

Extended Data Fig. 6 Independent repeat of 13C carbon tracing by LC-HRMS and Hexokinase-2 mRNA expression in support of Fig. 5.

(ag) Bead purified naive, in vivo activated and GC B cells were stimulated with anti-CD40 in glucose and glutamine free RPMI media with the addition of 2 mg/ml [13C6]-glucose for 30 min or 4 h. Cells (ae, g) and supernatants (f) were then subjected to LC-HRMS. Depicted is one representative experiment with n = 6 per sample. Each n represents a pool of 3 individual wells. (H) qRT-PCR for Hexokinase-2 expression from freshly isolated cell populations relative to RPS9. Bars are means + /-SEM. Normalization was performed as described in the methods section; ns = not significant; *p ≤ 0.05; **p ≤ 0.01; ***p ≤ 0.001; ****p ≤ 0.0001 by unpaired, two-tailed t-test.

Extended Data Fig. 7 In vivo 13C tracing in support of Fig. 6.

LC-HRMS analysis of 13C3 lactate generated from 8 h continuous 13C6 glucose infusion using an “insulin clamp”. Mice received a primed (42.5 mU/kg)/continuous (4.5 mU/kg/min) infusion of insulin and a variable infusion of 20% glucose (50% 13C6-glucose: 50% 12C-glucose) to maintain euglycemia for 480 min. Mice were sacrificed and heart and liver were disrupted in liquid nitrogen. B cell populations were isolated as in Extended Data Fig. 1 and all samples were subjected to 13C3-lactate detection by liquid chromatography-high resolution mass spectrometry.

Extended Data Fig. 8 Absence of hypoxia-related gene signatures in the GCBC transcriptome in support of Fig. 7.

Depicted are quantile normalized expression values of HIF1alpha direct target genes that are involved in glycolysis (a) or not involved in glycolysis (b) and control genes that are known to be expressed or absent in GCBC (c). Significant down-regulation of hypoxia-related gene signatures in GCBC transcriptome compared to naïve B cells (left of d) and in vivo activated B cells (right of d). Data were obtained by RNA-sequencing of indicated cell populations as in Extended Data Fig. 1. Shown are averages of 3 independent RNA sequencing reactions per cell population with x-axis showing different cell populations as defined in the text (ac). Genes are connected by lines for easier visualization and bars are means + /-SEM. Gene set enrichment plots illustrating differentially expressed genes in peak GCBCs compared to naive (left of d) and in vivo activated B cells (right of d; n = 3 per group) with respect to genes depicted in a and b. p-values were calculated using the rankSumTestWithCorrelation function in limma with t statistics.

Supplementary information

Rights and permissions

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Weisel, F.J., Mullett, S.J., Elsner, R.A. et al. Germinal center B cells selectively oxidize fatty acids for energy while conducting minimal glycolysis. Nat Immunol 21, 331–342 (2020). https://doi.org/10.1038/s41590-020-0598-4

Download citation

  • Received:

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1038/s41590-020-0598-4

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