Skip to main page content
U.S. flag

An official website of the United States government

Dot gov

The .gov means it’s official.
Federal government websites often end in .gov or .mil. Before sharing sensitive information, make sure you’re on a federal government site.

Https

The site is secure.
The https:// ensures that you are connecting to the official website and that any information you provide is encrypted and transmitted securely.

Access keys NCBI Homepage MyNCBI Homepage Main Content Main Navigation
. 2025 Feb 11;9(3):520-532.
doi: 10.1182/bloodadvances.2024013267.

Congenital T-cell activation impairs transitional-to-follicular B-cell maturation in humans

Affiliations

Congenital T-cell activation impairs transitional-to-follicular B-cell maturation in humans

Hugues Allard-Chamard et al. Blood Adv. .

Abstract

Patients with cytotoxic T-lymphocyte-associated protein 4 (CTLA4) deficiency exhibit profound humoral immune dysfunction, yet the basis for the B-cell defect is not known. We observed a marked reduction in transitional-to-follicular (FO) B-cell development in patients with CTLA4 deficiency, correlating with decreased CTLA4 function in regulatory T cells, increased CD40L levels in effector CD4+ T cells, and increased mammalian target of rapamycin complex 1 (mTORC1) signaling in transitional B cells (TrBs). Treatment of TrBs with CD40L was sufficient to induce mTORC1 signaling and inhibit FO B-cell maturation in vitro. Frequent cell-to-cell contacts between CD40L+ T cells and immunoglobulin D-positive CD27- B cells were observed in patient lymph nodes. FO B-cell maturation in patients with CTLA4 deficiency was partially rescued after CTLA4 replacement therapy in vivo. We conclude that functional regulatory T cells and the containment of excessive T-cell activation may be required for human TrBs to mature and attain metabolic quiescence at the FO B-cell stage.

PubMed Disclaimer

Conflict of interest statement

Conflict-of-interest disclosure: J.R.F. was supported by an investigator-initiated research grant from Bristol Myers Squibb. A.K.S. is a founder and serves on the scientific advisory board of Honeycomb Biotechnologies, which is developing Seq-Well arrays for commercial use. S.P. serves on the scientific advisory boards of Abpro Inc, Paratus, and BE Biopharma Inc. H.A.-C. was supported by investigator-initiated research grants from Pfizer, AstraZeneca, Fresenius Kabi, and Boehringer Ingelheim. The remaining authors declare no competing financial interests.

The current affiliation for K.H. is Division of Pediatric Hematology-Oncology, Department of Pediatrics, Hassenfeld Children's Hospital at New York University Langone Health, New York University Grossman School of Medicine, New York, NY.

The current affiliation for J.R.F. is Clinical Immunodeficiency Program of Beth Israel Lahey Health, Division of Allergy and Immunology, Lahey Hospital & Medical Center, Burlington, MA.

Figures

None
Graphical abstract
Figure 1.
Figure 1.
Increased frequency of circulating TrBs and decreased frequency of circulating FO B cells in patients with CTLA4 deficiency. Flow cytometric analysis of peripheral blood B-cell populations in patients with CTLA4 deficiency (n = 11) and healthy controls (n = 16). (A) Representative plots from a healthy control (left) and a patient with CTLA4 deficiency (right, homozygous CTLA4 3′ UTR mutation carrier), shown at 5% of events with gating strategy outlined (full description of B-cell gating strategy in supplemental Figure 1A). (B) Quantification of major B-cell subsets in the peripheral blood, including naïve, marginal zone (MZ), SWM, and DN populations as subset from total CD19+ B cells by the markers IgD and CD27. (C) Quantification of naïve (IgD+CD27) B-cell subsets in the peripheral blood, including T1/2, T3a, and T3b TrBs, resting mature FO B cells, aN B cells, and MZP B cells as further subsets by the indicated markers. For the CD45RB by CD21 and count by CD73 B-cell subset plots, final IgD+CD27 B-cell frequency was derived as a percentage of the indicated parent gate. Symbols represent unique individuals; bars represent means (± standard deviation [SD]) of all data. ∗P < .05; ∗∗P < .01; ∗∗∗P < .001; ∗∗∗∗P < .0001 or as listed by 2-tailed Mann-Whitney U test. MZP, MZ precursor.
Figure 2.
Figure 2.
Reduced frequency of circulating FO B cells correlates with degree of congenital Treg dysfunction in CTLA4 deficiency. CHO-CD80-GFP cells were cocultured with activated T cells from the peripheral blood of patients with CTLA4 deficiency (n = 10) and healthy controls (n = 7). Gated CD4+FOXP3+ Tregs that had acquired GFP from CHO expressing CD80-GFP cells were scored positive for intact CTLA4-mediated transendocytosis. (A) Representative contour plots from a healthy control (left) and a patient with CTLA4 deficiency (right, heterozygous CTLA4 c.410C>T, p.P137L mutation carrier), shown at 2% of events with gating strategy outlined. Quantification of (B) total cellular CTLA4 levels by intracellular staining or (C) CTLA4 function by CD80-GFP transendocytosis as percent GFP+CTLA4+ in stimulated peripheral blood CD4+FOXP3+ Tregs from patients with CTLA4 deficiency compared with healthy controls (left) and compared by CTLA4 mutation type (right). Symbols represent unique individuals and bars represent means (± SD) of all data. ∗∗P < .01 by 2-tailed Mann-Whitney U test. (D-E) Correlations between FO B-cell frequency in the peripheral blood and severity of the underlying CTLA4 mutation type. Y-axes are FO B-cell frequency as percent IgD+CD27 B cells in the peripheral blood (data from Figure 1C, here shown as means ± standard error of the mean for all analyzed patients with the indicated CTLA4 mutation type or healthy control state, respectively). (D) X-axis is total cellular CTLA4 levels as MFI by intracellular staining in stimulated CD4+FOXP3+ Tregs (data from panel B, here consolidated as geometric mean per CTLA4 mutation type or healthy control state, respectively). (E) X-axis is CTLA4 function as CD80-GFP transendocytosis (percent GFP+CTLA4+) in stimulated CD4+FOXP3+ Tregs (data from panel C, here consolidated as geometric mean per CTLA4 mutation type or healthy control state, respectively). Simple linear regression with correlation coefficient (R2) and P value shown. Homo, homozygous; het, heterozygous.
Figure 3.
Figure 3.
Increased CD40L levels in effector CD4+ T cells in CTLA4 deficiency correlate with a decrease in the frequency of circulating FO B cells. (A) Extracellular flow cytometric analysis of peripheral blood T-cell populations in patients with CTLA4 deficiency (n = 11) and healthy controls (n = 10). Representative contour plots from a healthy control (left) and 2 patients with CTLA4 deficiency (right, heterozygous CTLA4 c.410C>T, p.P137L mutation carriers) shown at 5% of events (full description of T-cell gating strategy in supplemental Figure 1B). (B) Quantification of the major CD4+ T-cell subsets defined by the extracellular markers CXCR5, CCR7, and CD45RA, including TFH, naïve, CM, TEMRA, and EM populations. Symbols represent unique individuals; bars represent means (± SD) of all data. ∗P < .05; ∗∗P < .01 by 2-tailed Mann-Whitney U test. (C) Correlation between FO B-cell frequency as percent IgD+CD27 B cells in the peripheral blood (data from Figure 1C; x-axis) and naïve CD4+ T-cell frequency as percent CD4+CXCR5 T cells in the peripheral blood (data from Panel B; y-axis). Simple linear regression with correlation coefficient (R2), P value, and 95% confidence interval (CI) shown. (D) Intracellular flow cytometric analysis of peripheral blood T-cell populations in patients with CTLA4 deficiency (n = 8) and healthy controls (n = 10). Representative overlaid histograms from a healthy control (blue) and a patient with CTLA4 deficiency (red, heterozygous CTLA4 c.173G>C p.C58S mutation carrier) shown. (E) Quantification of total cellular CD40L levels (MFI) by intracellular staining in the major CD4+ T-cell subsets described above in patients with CTLA4 deficiency and healthy controls. Symbols represent unique individuals; bars represent means (± SD) of all data. ∗P < .05; ∗∗P < .01 or as listed by 2-tailed Mann-Whitney U test. (F) Correlation between FO B-cell frequency as percent IgD+CD27 B cells in the peripheral blood (data from Figure 1C; x-axis) and total cellular CD40L levels as MFI in TEMRA CD4+ T cells (data from panel E; y-axis). Simple linear regression with correlation coefficient (R2), P value, and 95% CI shown. CM, central memory; EM, effector memory; TFH, T follicular helper.
Figure 4.
Figure 4.
Increased mTORC1 signaling due to abundance of transitional and ABC-like aN B-cell clusters in patients with CTLA4 deficiency. (A) Volcano plot of shared differential gene expression analysis between bulk RNA-seq samples of flow-sorted resting FO and TrB populations (T1/2, T3; n = 3 healthy controls; significant differentially expressed genes defined by adjusted P value ≤ .01). (B) Adjusted P value of ingenuity pathway analysis pathway overlaps with significantly differentially expressed genes in TrB vs resting FO B cells. (C) Uniform manifold approximation and projection (UMAP) projection of total naïve (IgD+CD27) B cells profiled by scRNA-seq from patients with CTLA4 deficiency (n = 4) and healthy controls (n = 3), annotated by naïve B-cell subtype. (D) Dot plot showing the marker gene expression of FO, TrB, and aN B-cell types by cell type annotation; color represents average normalized gene expression and size represents the percent cells with non-zero gene expression. (E) Heat map of scaled gene expression of top 30 differentially expressed genes between annotated cell types. (F) UMAP projection of total naïve (IgD+CD27) B cells profiled by scRNA-seq, annotated by control or patients with CTLA4 deficiency. (G) Fraction representation of each annotated cell type within each donor scRNA-seq sample. (H) Average log2-fold change of significant differentially expressed genes (adjusted P value ≤ .05) between distinguishing CD38+ TrBs vs CCR7+ FO B cells plotted on the x-axis and average log2-fold change of significant differentially expressed genes between ITGAX+ ABC-like aN B cells vs CCR7+ FO B cells plotted on the y-axis, showing correlation in differential gene expression distinguishing both TrBs and ABC-like aN B cells from FO B cells. Differentially expressed genes that overlap with significant differentially expressed genes from the bulk RNA-seq analysis in panel A are outlined. (I) Single-cell module scores defined by TrB and ABC-like aN cells shared differentially expressed genes in both TrB and ABC-like aN cells plotted on the x-axis (TrB coexpression score), vs module scores defined by shared FO differentially expressed genes in FO cells on the y-axis (FO B score). Single cells from healthy controls are plotted separately from single cells from patients with CTLA4 deficiency to demonstrate consistent trends in cell type population scores. Pearson correlation rho values (−0.313 for healthy controls, −0.316 for patients with CTLA4 deficiency) were both significant (P < 2E−16). (J) Violin plots of module scores over msigDB hallmark oxidative phosphorylation and mTORC1 signaling pathways (see “Methods”) in healthy controls and patients with CTLA4 deficiency. Violin plots of single-cell module scores shown and evaluated for significance separately for healthy controls and patients with CTLA4 deficiency; ∗∗P < .001; ∗∗∗P < .0001. DEG, differentialy expressed gene; ERK, extracellular signal-regulated kinase; IL4, interleukin 4; msigDB, molecular signatures database; NAD, nicotinamide adenine dinucleotide.
Figure 4.
Figure 4.
Increased mTORC1 signaling due to abundance of transitional and ABC-like aN B-cell clusters in patients with CTLA4 deficiency. (A) Volcano plot of shared differential gene expression analysis between bulk RNA-seq samples of flow-sorted resting FO and TrB populations (T1/2, T3; n = 3 healthy controls; significant differentially expressed genes defined by adjusted P value ≤ .01). (B) Adjusted P value of ingenuity pathway analysis pathway overlaps with significantly differentially expressed genes in TrB vs resting FO B cells. (C) Uniform manifold approximation and projection (UMAP) projection of total naïve (IgD+CD27) B cells profiled by scRNA-seq from patients with CTLA4 deficiency (n = 4) and healthy controls (n = 3), annotated by naïve B-cell subtype. (D) Dot plot showing the marker gene expression of FO, TrB, and aN B-cell types by cell type annotation; color represents average normalized gene expression and size represents the percent cells with non-zero gene expression. (E) Heat map of scaled gene expression of top 30 differentially expressed genes between annotated cell types. (F) UMAP projection of total naïve (IgD+CD27) B cells profiled by scRNA-seq, annotated by control or patients with CTLA4 deficiency. (G) Fraction representation of each annotated cell type within each donor scRNA-seq sample. (H) Average log2-fold change of significant differentially expressed genes (adjusted P value ≤ .05) between distinguishing CD38+ TrBs vs CCR7+ FO B cells plotted on the x-axis and average log2-fold change of significant differentially expressed genes between ITGAX+ ABC-like aN B cells vs CCR7+ FO B cells plotted on the y-axis, showing correlation in differential gene expression distinguishing both TrBs and ABC-like aN B cells from FO B cells. Differentially expressed genes that overlap with significant differentially expressed genes from the bulk RNA-seq analysis in panel A are outlined. (I) Single-cell module scores defined by TrB and ABC-like aN cells shared differentially expressed genes in both TrB and ABC-like aN cells plotted on the x-axis (TrB coexpression score), vs module scores defined by shared FO differentially expressed genes in FO cells on the y-axis (FO B score). Single cells from healthy controls are plotted separately from single cells from patients with CTLA4 deficiency to demonstrate consistent trends in cell type population scores. Pearson correlation rho values (−0.313 for healthy controls, −0.316 for patients with CTLA4 deficiency) were both significant (P < 2E−16). (J) Violin plots of module scores over msigDB hallmark oxidative phosphorylation and mTORC1 signaling pathways (see “Methods”) in healthy controls and patients with CTLA4 deficiency. Violin plots of single-cell module scores shown and evaluated for significance separately for healthy controls and patients with CTLA4 deficiency; ∗∗P < .001; ∗∗∗P < .0001. DEG, differentialy expressed gene; ERK, extracellular signal-regulated kinase; IL4, interleukin 4; msigDB, molecular signatures database; NAD, nicotinamide adenine dinucleotide.
Figure 5.
Figure 5.
CD40L induces mTORC signaling and arrests FO B-cell maturation in vitro. (A) Levels of (p)-S6 by intracellular flow cytometry in transitional (T) and FO B-cell subsets, sorted and cultured for 24 hours in vitro with CD40L treatment (+CD40L) compared with without (untx) as indicated. Histograms are representative of 3 independent experiments. Quantified data, from 3 independent experiments, are means (± SD) of all data. ∗∗P < .01 by unpaired Student t test. (B) Differentiation by extracellular flow cytometry of T3a and T3b B cells, sorted and cultured for 24 hours in vitro with, compared with, without CD40L treatment or AICAR treatment to inhibit mTORC signaling as a control. Histograms are representative of 3 independent experiments, here showing differences in T3a B-cell levels of MitoTracker (MTG) and CD73 at 24 hours in culture by treatment condition as indicated. Differentiation to the FO B-cell stage was scored by the acquisition of the CD73+MTG phenotype. Quantified data, from 2 independent experiments, are means (± SD) of all data. ∗∗P < .01; ∗∗∗∗P < .0001 by unpaired Student t test. AICAR, 5-aminoimidazole-4-carboxamide ribonucleotide; untx, untreated.
Figure 6.
Figure 6.
Treatment with abatacept (CTLA4 immunoglobulin) restores FO B-cell maturation coincident with decreased CD40L levels on effector T cells in CTLA4 deficiency. (A-B) Flow cytometric analysis of peripheral blood B-cell populations in patients with CTLA4 deficiency before (n = 11) and after abatacept (n = 8) therapy (6 patients captured at paired intervals before/after abatacept therapy) and healthy controls (n = 16). Representative contour plots and histograms from a healthy control (left) and the same patient with CTLA4 deficiency (middle and right) before/after abatacept therapy (homozygous CTLA4 3′ UTR mutation carrier in panel A, and heterozygous CTLA4 c.410C>T, p.P137L mutation carrier in panel B) shown at 10% and 5% of events, respectively. Quantitation of B-cell subset frequency as percent IgD+CD27 B cells in the peripheral blood. Symbols represent unique individuals; bars represent means (± SD) of all data; dotted lines represent paired pretreatment to posttreatment time points. ∗P < .05; ∗∗P < .01; ∗∗∗P < .001; ∗∗∗∗P < .0001, or as listed by analysis of variance (ANOVA) for multiple comparisons or by Wilcoxon matched-pairs signed-rank test, as indicated. (C) scRNA-seq of total IgD+CD27 B cells (from heterozygous CTLA4 c.410C>T, p.P137L mutation carrier) before and after abatacept therapy. UMAP projection of cells is shown by cell type clustering and state of abatacept treatment. Fraction representation of each clustered scRNA-seq cell type. (D) Flow cytometric analysis of peripheral blood T-cell populations in patients with CTLA4 deficiency before (n = 8) and after abatacept (n = 6) therapy (4 patients captured at paired intervals before/after abatacept therapy) and healthy controls (n = 10). Representative overlaid histograms from the same patient with CTLA4 deficiency before/after abatacept therapy (heterozygous CTLA4 c.173G>C p.C58S mutation carrier). Quantitation of total cellular CD40L by intracellular staining in CD4+ T-cell subsets. Symbols represent unique individuals; bars represent means (± SD) of all data; dotted lines represent paired before to after treated unique patients. ∗P < .05 or as listed by ANOVA for multiple comparisons or by Wilcoxon matched-pairs signed-rank test, as indicated. (E) Correlation between FO B-cell frequency as percent IgD+CD27 B cells in the peripheral blood (data from panel A; x-axis) and total cellular CD40L levels as MFI in TEMRA CD4+ T cells (data from panel D; y-axis). Data are from untreated patients with CTLA4 deficiency, treated patients with CTLA4 deficiency, and healthy controls, as indicated. Simple linear regression with correlation coefficient (R2), P value, and 95% CI shown. ns, not significant.

References

    1. Farmer JR, Allard-Chamard H, Sun N, et al. Induction of metabolic quiescence defines the transitional to follicular B cell switch. Sci Signal. 2019;12(604) - PMC - PubMed
    1. Schubert D, Bode C, Kenefeck R, et al. Autosomal dominant immune dysregulation syndrome in humans with CTLA4 mutations. Nat Med. 2014;20(12):1410–1416. - PMC - PubMed
    1. Kuehn HS, Ouyang W, Lo B, et al. Immune dysregulation in human subjects with heterozygous germline mutations in CTLA4. Science. 2014;345(6204):1623–1627. - PMC - PubMed
    1. Schwab C, Gabrysch A, Olbrich P, et al. Phenotype, penetrance, and treatment of 133 cytotoxic T-lymphocyte antigen 4-insufficient subjects. J Allergy Clin Immunol. 2018;142(6):1932–1946. - PMC - PubMed
    1. Tivol EA, Borriello F, Schweitzer AN, Lynch WP, Bluestone JA, Sharpe AH. Loss of CTLA-4 leads to massive lymphoproliferation and fatal multiorgan tissue destruction, revealing a critical negative regulatory role of CTLA-4. Immunity. 1995;3(5):541–547. - PubMed

MeSH terms