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 Mar 18;135(10):e178975.
doi: 10.1172/JCI178975. eCollection 2025 May 15.

Maintenance of graft tissue-resident Foxp3+ cells is necessary for lung transplant tolerance in mice

Affiliations

Maintenance of graft tissue-resident Foxp3+ cells is necessary for lung transplant tolerance in mice

Wenjun Li et al. J Clin Invest. .

Abstract

Mechanisms that mediate allograft tolerance differ between organs. We have previously shown that Foxp3+ T cell-enriched bronchus-associated lymphoid tissue (BALT) is induced in tolerant murine lung allografts and that these Foxp3+ cells suppress alloimmune responses locally and systemically. Here, we demonstrated that Foxp3+ cells that reside in tolerant lung allografts differed phenotypically and transcriptionally from those in the periphery and were clonally expanded. Using a mouse lung retransplant model, we showed that recipient Foxp3+ cells were continuously recruited to the BALT within tolerant allografts. We identified distinguishing features of graft-resident and newly recruited Foxp3+ cells and showed that graft-infiltrating Foxp3+ cells acquired transcriptional profiles resembling those of graft-resident Foxp3+ cells over time. Allografts underwent combined antibody-mediated rejection and acute cellular rejection when recruitment of recipient Foxp3+ cells was prevented. Finally, we showed that local administration of IL-33 could expand and activate allograft-resident Foxp3+ cells, providing a platform for the design of tolerogenic therapies for lung transplant recipients. Our findings establish graft-resident Foxp3+ cells as critical orchestrators of lung transplant tolerance and highlight the need to develop lung-specific immunosuppression.

Keywords: Immunology; Tolerance; Transplantation.

PubMed Disclaimer

Figures

Figure 1
Figure 1. Allograft-resident and spleen Foxp3+ cells are phenotypically and transcriptionally distinct in tolerant lung transplant recipients.
(AC) Gross image (n = 6) (A), H&E staining (n = 6) (B), and CCSP immunofluorescence staining (n = 2) (C) of left lung transplant from BALB/c donor into CSB-treated B6 recipient, at least 30 days after engraftment. Scale bars: 100 μm. (D) Two-photon intravital microscopy of BALB/c lung at least 30 days after transplantation into CSB-treated B6 CD11c-EYFP/Foxp3-GFP recipient (n = 3). CD11c+ cells are yellow, Foxp3+ cells are green, collagen appears blue owing to second-harmonic generation (SHG), and vessels are red following i.v. injection of quantum dots. Scale bar: 20 μm. (EI) Representative flow cytometry plots and quantification of abundance of Foxp3-expressing CD45+CD90.2+CD4+CD8 T cells (E), CD25-expressing CD45+CD90.2+CD4+CD8Foxp3+ T cells (CD25 expression determined based on isotype control staining) (F), effector memory (CD44hiCD62Llo), central memory (CD44hiCD62Lhi), and naive (CD44loCD62Lhi) CD45+CD90.2+CD4+CD8Foxp3+ T cells (G), CD69-expressing CD45+CD90.2+CD4+CD8Foxp3+ T cells (H), and (I) PD-1–expressing CD45+CD90.2+CD4+CD8Foxp3+ T cells in BALB/c lung and recipient spleen at least 30 days after transplantation into CSB-treated B6 recipients (n = 4). At least 30 days after transplantation of BALB/c lungs into CSB-treated B6 Foxp3-GFP mice, CD45+CD90.2+CD4+CD8GFP+ cells were sorted and processed for TCR and genome sequencing (n = 2). (J) Gini coefficient (0 represents maximal diversity) comparison of clonal expansion (left) and number of cells in the top 11 shared clonotypes between Foxp3+ cells in tolerant lung allografts and recipient spleens (right) (triangles and circles denote individual mice). (K) Heatmap of most highly differentially expressed genes in Foxp3+ cells between tolerant lung allografts and recipient spleens (2 pooled lungs and spleens). (LN) Gross image (n = 4) (L), H&E staining (n = 4) (M), and CCSP immunofluorescence staining (n = 2) (N) of BALB/c lungs 30 days after transplantation into CSB-treated B6 Foxp3-YFP-Cre Aregfl/fl mice. Scale bars: 100 μm. Results expressed as mean ± SEM. **P < 0.01, ***P < 0.001. d30, day 30; q-dot, quantum dot; Tcm, T central memory; Tem, T effector memory; Treg, regulatory Foxp3+ T cell; TX, transplanted lung.
Figure 2
Figure 2. Graft-infiltrating Foxp3+ cells are derived from the thymus rather than from peripheral conversion of Foxp3 T cells.
(AC) Representative flow cytometric plots (A and B) and quantification (C) of Helios and neuropilin-1 expression by CD45+CD90.2+CD4+CD8Foxp3+ T cells in BALB/c lungs at least 30 days after transplantation into CSB-treated B6 recipients (n = 4). Thirty days after transplantation of BALB/c (CD45.2) lungs into CSB-treated B6 (CD45.2) recipients, CD90.2+CD4+GFP T cells, isolated from secondary lymphoid organs of B6 Foxp3-GFP (CD45.1) reporter mice, were injected i.v. Lungs were analyzed 7 days later. (D and E) Representative flow cytometric plot and analysis of GFP expression in adoptively transferred cells (CD45.1+) in lung allografts (n = 3). Results expressed as mean ± SEM.
Figure 3
Figure 3. Newly recruited Foxp3+ cells infiltrate BALT within tolerant lung allografts and differ transcriptionally from graft-resident Foxp3+ cells.
(A) BALB/c lungs initially transplanted into CSB-treated B6 CD11c-EYFP mice and then at least 30 days later retransplanted into non-immunosuppressed B6 Foxp3-GFP hosts were imaged with intravital 2-photon microscopy 3 days after retransplantation (n = 3). White arrows point to contacts between Foxp3+ (blue) and CD11c+ (green) cells within the BALT of lung allografts. Rhodamine-dextran labels vessels red. (B) BALB/c lungs initially transplanted into CSB-treated B6 Foxp3-GFP mice and then at least 30 days later retransplanted into non-immunosuppressed B6 Foxp3-RFP recipients were imaged with intravital 2-photon microscopy 3 days after retransplantation (n = 3). White arrows point to contacts between graft-resident (green) and graft-infiltrating (red) Foxp3+ cells within lung allografts. (C and D) BALB/c (CD45.2) lungs were transplanted into CSB-treated B6 (CD45.2) recipients and at least 30 days later retransplanted into non-immunosuppressed B6 (CD45.1) mice. Seven and 21 days after retransplantation, graft-resident (CD45.2) (7 days) and extravasated graft-infiltrating (CD45.1) (7 and 21 days) T cells were sorted from the lung allografts (samples were collected from 4 retransplant recipients and pooled) and processed for single-cell RNA sequencing. (C) Uniform manifold approximation and projection (UMAP) embedding plot of regulatory T cell subpopulations. (D) Heatmap of statistically significant (log2 fold change > 0.25, adjusted P value < 0.05) differentially expressed genes between graft-resident CD45.2 (day 7) and extravasated graft-infiltrating CD45.1 (days 7 and 21) regulatory T cells grouped by condition. (EI) Representative flow cytometry plots and quantification of abundance of effector memory (CD44hiCD62Llo), central memory (CD44hiCD62Lhi), and naive (CD44loCD62Lhi) graft-resident CD45.2+ versus graft-infiltrating CD45.1+CD90.2+CD4+CD8Foxp3+ T cells on day 7 (n = 4). Results expressed as mean ± SEM. Scale bars: 20 μm. **P < 0.01, ***P < 0.001. RFP, red fluorescent protein; Tcm, T central memory; Tem, T effector memory.
Figure 4
Figure 4. Depletion of recipient Foxp3+ cells results in loss of allograft tolerance.
(A) Schematic depicting left lung from BALB/c (CD45.2) donor initially transplanted into CSB-treated B6 (CD45.2) primary recipient and then at least 30 days later retransplanted into non-immunosuppressed B6 (CD45.1) or B6 Foxp3-DTR (CD45.1) secondary recipient, with diphtheria toxin (DT) administration and analysis 7 days after retransplantation. (B and C) Gross image (first panel, n = 4), H&E histology (second panel, n = 4), CCSP immunofluorescence staining (green, third panel, n = 2), and immunohistochemical staining for complement 4d (C4d) (brown, fourth panel; arrows point to alveolar endothelial C4d staining; n = 2) in DT-treated B6 (B) and B6 Foxp3-DTR (C) secondary recipients. (DF) Representative flow cytometric plots and quantification of abundance of CD69 (D), Fas (E), and IgM/IgD (F) expression in graft-infiltrating CD45.2CD45.1+CD19+B220+ B cells in control (left panels) and recipient Foxp3-depleted (right panels) retransplants (n ≥ 4). (G and H) Flow cytometric analysis of serum donor-specific IgM antibody titers (expressed as mean fluorescence intensity) (G) and International Society for Heart and Lung Transplantation (ISHLT) A rejection grades (H) in control and recipient Foxp3-depleted retransplants (n = 4). Results expressed as mean ± SEM. Scale bars: 100 μm. *P < 0.05, **P < 0.01, ***P < 0.001. DT, diphtheria toxin; DTR, diphtheria toxin receptor; TX, transplanted lung.
Figure 5
Figure 5. Rejection after recipient Foxp3+ cell depletion is dependent on B cells.
(A) BALB/c lungs were initially transplanted into CSB-treated B6 CD11c-EYFP mice and then retransplanted into non-immunosuppressed B6 Foxp3-GFP hosts at least 30 days later. Recipient-matched B6 B cells labeled with the rhodamine-based red cell dye CMTMR were injected into recipients 2 days after retransplantation, and allografts were imaged with intravital 2-photon microscopy the following day (n = 3). White arrows point to contacts between Foxp3+ (green) and B cells (red). CD11c+ cells (yellow) mark the BALT within tolerant lung allografts (n = 4). Scale bar: 20 μm. (B) Gross (top) and histological (H&E; bottom) images of BALB/c lungs that were initially transplanted into CSB-treated B6 mice and then at least 30 days later retransplanted into Foxp3-depleted non-immunosuppressed B6 recipients that received anti-CD20 (right) or isotype control antibodies (left). Scale bars: 100 μm. (C and D) Flow cytometric analysis of serum IgM DSA titers (expressed as mean fluorescence intensity) (C) and ISHLT A rejection grades (D) in retransplant recipients described in B (n ≥ 4). DT, diphtheria toxin; DTR, diphtheria toxin receptor; TX, transplanted lung.
Figure 6
Figure 6. Lung allograft tolerance is maintained in the global absence of CD4+ T cells.
(A and B) Gross (A) and histological (H&E) (B) images of BALB/c lungs that were initially transplanted into CSB-treated B6 mice and then at least 30 days later retransplanted into non-immunosuppressed B6 CD4 knockout recipients. Scale bar: 100 μm. (C and D) ISHLT A rejection grades (C) and flow cytometric analysis of serum IgM DSA titers (expressed as mean fluorescence intensity) (D) in retransplants depicted in A and B compared with tolerant BALB/c lung allografts that were retransplanted into non-immunosuppressed B6 mice (n ≥ 3). Mice were examined 7 days after retransplantation. TX, transplanted lung.
Figure 7
Figure 7. IL-33 promotes the expansion and activation of lung allograft–resident Foxp3+ cells.
BALB/c lungs were transplanted into CSB-treated B6 recipients. At least 30 days after transplantation, recipients were treated with intratracheal IL-33 or PBS, and grafts were analyzed 7 days later. (AE) Representative flow cytometry plots and quantification of abundance of Foxp3-expressing CD45+CD90.2+CD4+CD8 T cells (A), CD25-expressing CD45+CD90.2+CD4+CD8Foxp3+ T cells (B), effector memory (CD44hiCD62Llo), central memory (CD44hiCD62Lhi), and naive (CD44loCD62Lhi) CD45+CD90.2+CD4+CD8Foxp3+ T cells (C), CD69-expressing CD45+CD90.2+CD4+CD8Foxp3+ T cells (D), and (E) PD-1–expressing CD45+CD90.2+CD4+CD8Foxp3+ T cells in lung allografts after treatment with IL-33 or PBS (n = 4). (F) Gross (top) and histological (H&E; bottom) appearances of left BALB/c lungs that were initially transplanted into CSB-treated B6 recipients and then 30 days later retransplanted into DT-treated B6 Foxp3-DTR secondary recipients that received PBS (left; n = 4) or IL-33 (right; n = 9) intratracheally. Grafts were examined 7 days after retransplantation. Scale bars: 100 μm. (G and H) ISHLT A rejection grades (G) and flow cytometric analysis of serum IgM DSA titers (expressed as mean fluorescence intensity) (H) in recipients depicted in F (PBS, n = 4; IL-33, n = 9). Results expressed as mean ± SEM. *P < 0.05, **P < 0.01. DT, diphtheria toxin; DTR, diphtheria toxin receptor; Tcm, T central memory; Tem, T effector memory; TX, transplanted lung.

References

    1. Balsara KR, et al. A single-center experience of 1500 lung transplant patients. J Thorac Cardiovasc Surg. 2018;156(2):894–905. doi: 10.1016/j.jtcvs.2018.03.112. - DOI - PMC - PubMed
    1. Witt CA, et al. Lung transplant immunosuppression — time for a new approach? Expert Rev Clin Immunol. 2014;10(11):1419–1421. doi: 10.1586/1744666X.2014.959499. - DOI - PMC - PubMed
    1. Krupnick AS, et al. Central memory CD8+ T lymphocytes mediate lung allograft acceptance. J Clin Invest. 2014;124(3):1130–1143. doi: 10.1172/JCI71359. - DOI - PMC - PubMed
    1. Onyema OO, et al. Eosinophils promote inducible NOS-mediated lung allograft acceptance. JCI Insight. 2017;2(24):e96455. doi: 10.1172/jci.insight.96455. - DOI - PMC - PubMed
    1. Onyema OO, et al. Eosinophils downregulate lung alloimmunity by decreasing TCR signal transduction. JCI Insight. 2019;4(11):e128241. doi: 10.1172/jci.insight.128241. - DOI - PMC - PubMed