Go to JCI Insight
  • About
  • Editors
  • Consulting Editors
  • For authors
  • Publication ethics
  • Publication alerts by email
  • Advertising
  • Job board
  • Contact
  • Clinical Research and Public Health
  • Current issue
  • Past issues
  • By specialty
    • COVID-19
    • Cardiology
    • Gastroenterology
    • Immunology
    • Metabolism
    • Nephrology
    • Neuroscience
    • Oncology
    • Pulmonology
    • Vascular biology
    • All ...
  • Videos
    • Conversations with Giants in Medicine
    • Video Abstracts
  • Reviews
    • View all reviews ...
    • Pancreatic Cancer (Jul 2025)
    • Complement Biology and Therapeutics (May 2025)
    • Evolving insights into MASLD and MASH pathogenesis and treatment (Apr 2025)
    • Microbiome in Health and Disease (Feb 2025)
    • Substance Use Disorders (Oct 2024)
    • Clonal Hematopoiesis (Oct 2024)
    • Sex Differences in Medicine (Sep 2024)
    • View all review series ...
  • Viewpoint
  • Collections
    • In-Press Preview
    • Clinical Research and Public Health
    • Research Letters
    • Letters to the Editor
    • Editorials
    • Commentaries
    • Editor's notes
    • Reviews
    • Viewpoints
    • 100th anniversary
    • Top read articles

  • Current issue
  • Past issues
  • Specialties
  • Reviews
  • Review series
  • Conversations with Giants in Medicine
  • Video Abstracts
  • In-Press Preview
  • Clinical Research and Public Health
  • Research Letters
  • Letters to the Editor
  • Editorials
  • Commentaries
  • Editor's notes
  • Reviews
  • Viewpoints
  • 100th anniversary
  • Top read articles
  • About
  • Editors
  • Consulting Editors
  • For authors
  • Publication ethics
  • Publication alerts by email
  • Advertising
  • Job board
  • Contact
Top
  • View PDF
  • Download citation information
  • Send a comment
  • Terms of use
  • Standard abbreviations
  • Need help? Email the journal
  • Top
  • Supplemental material
  • Footnotes
  • References
  • Version history
  • Article usage
  • Citations to this article

Advertisement

Research LetterCardiologyInflammation Open Access | 10.1172/JCI192811

Type 1 classical dendritic cells govern long-term cardiac allograft acceptance

Macee C. Owen,1 Vinay R. Penna,1 Hao Dun,2 Wenjun Li,3 Benjamin J. Kopecky,3 Kenneth M. Murphy,4 Daniel Kreisel,3,4 and Kory J. Lavine1,4,5

1Department of Medicine, Washington University School of Medicine, St. Louis, Missouri, USA.

2Department of Medicine, University of Colorado Anschutz Medical Campus, Aurora, Colorado, USA.

3Department of Surgery,

4Department of Pathology & Immunology, and

5Department of Developmental Biology, Washington University School of Medicine, St. Louis, Missouri, USA.

Address correspondence to: Kory J. Lavine, Washington University School of Medicine, Campus Box 8086, St. Louis, Missouri 63110, USA. Phone: 314.362.1171; Email: klavine@wustl.edu.

Find articles by Owen, M. in: PubMed | Google Scholar

1Department of Medicine, Washington University School of Medicine, St. Louis, Missouri, USA.

2Department of Medicine, University of Colorado Anschutz Medical Campus, Aurora, Colorado, USA.

3Department of Surgery,

4Department of Pathology & Immunology, and

5Department of Developmental Biology, Washington University School of Medicine, St. Louis, Missouri, USA.

Address correspondence to: Kory J. Lavine, Washington University School of Medicine, Campus Box 8086, St. Louis, Missouri 63110, USA. Phone: 314.362.1171; Email: klavine@wustl.edu.

Find articles by Penna, V. in: PubMed | Google Scholar

1Department of Medicine, Washington University School of Medicine, St. Louis, Missouri, USA.

2Department of Medicine, University of Colorado Anschutz Medical Campus, Aurora, Colorado, USA.

3Department of Surgery,

4Department of Pathology & Immunology, and

5Department of Developmental Biology, Washington University School of Medicine, St. Louis, Missouri, USA.

Address correspondence to: Kory J. Lavine, Washington University School of Medicine, Campus Box 8086, St. Louis, Missouri 63110, USA. Phone: 314.362.1171; Email: klavine@wustl.edu.

Find articles by Dun, H. in: PubMed | Google Scholar

1Department of Medicine, Washington University School of Medicine, St. Louis, Missouri, USA.

2Department of Medicine, University of Colorado Anschutz Medical Campus, Aurora, Colorado, USA.

3Department of Surgery,

4Department of Pathology & Immunology, and

5Department of Developmental Biology, Washington University School of Medicine, St. Louis, Missouri, USA.

Address correspondence to: Kory J. Lavine, Washington University School of Medicine, Campus Box 8086, St. Louis, Missouri 63110, USA. Phone: 314.362.1171; Email: klavine@wustl.edu.

Find articles by Li, W. in: PubMed | Google Scholar

1Department of Medicine, Washington University School of Medicine, St. Louis, Missouri, USA.

2Department of Medicine, University of Colorado Anschutz Medical Campus, Aurora, Colorado, USA.

3Department of Surgery,

4Department of Pathology & Immunology, and

5Department of Developmental Biology, Washington University School of Medicine, St. Louis, Missouri, USA.

Address correspondence to: Kory J. Lavine, Washington University School of Medicine, Campus Box 8086, St. Louis, Missouri 63110, USA. Phone: 314.362.1171; Email: klavine@wustl.edu.

Find articles by Kopecky, B. in: PubMed | Google Scholar

1Department of Medicine, Washington University School of Medicine, St. Louis, Missouri, USA.

2Department of Medicine, University of Colorado Anschutz Medical Campus, Aurora, Colorado, USA.

3Department of Surgery,

4Department of Pathology & Immunology, and

5Department of Developmental Biology, Washington University School of Medicine, St. Louis, Missouri, USA.

Address correspondence to: Kory J. Lavine, Washington University School of Medicine, Campus Box 8086, St. Louis, Missouri 63110, USA. Phone: 314.362.1171; Email: klavine@wustl.edu.

Find articles by Murphy, K. in: PubMed | Google Scholar

1Department of Medicine, Washington University School of Medicine, St. Louis, Missouri, USA.

2Department of Medicine, University of Colorado Anschutz Medical Campus, Aurora, Colorado, USA.

3Department of Surgery,

4Department of Pathology & Immunology, and

5Department of Developmental Biology, Washington University School of Medicine, St. Louis, Missouri, USA.

Address correspondence to: Kory J. Lavine, Washington University School of Medicine, Campus Box 8086, St. Louis, Missouri 63110, USA. Phone: 314.362.1171; Email: klavine@wustl.edu.

Find articles by Kreisel, D. in: PubMed | Google Scholar

1Department of Medicine, Washington University School of Medicine, St. Louis, Missouri, USA.

2Department of Medicine, University of Colorado Anschutz Medical Campus, Aurora, Colorado, USA.

3Department of Surgery,

4Department of Pathology & Immunology, and

5Department of Developmental Biology, Washington University School of Medicine, St. Louis, Missouri, USA.

Address correspondence to: Kory J. Lavine, Washington University School of Medicine, Campus Box 8086, St. Louis, Missouri 63110, USA. Phone: 314.362.1171; Email: klavine@wustl.edu.

Find articles by Lavine, K. in: PubMed | Google Scholar

Published July 8, 2025 - More info

Published in Volume 135, Issue 18 on September 16, 2025
J Clin Invest. 2025;135(18):e192811. https://doi.org/10.1172/JCI192811.
© 2025 Owen et al. This work is licensed under the Creative Commons Attribution 4.0 International License. To view a copy of this license, visit http://creativecommons.org/licenses/by/4.0/.
Published July 8, 2025 - Version history
View PDF

Related article:

Treg activation during allograft tolerance induction requires mitochondrion-induced TGF-β1 in type 1 conventional dendritic cells
Samantha L. Schroth, … , Maria-Luisa Alegre, Edward B. Thorp
Samantha L. Schroth, … , Maria-Luisa Alegre, Edward B. Thorp
This study newly implicates a dendritic cell subset and an underlying immunometabolic mechansiom during strategies for allograft tolerance
Research Article Immunology

Treg activation during allograft tolerance induction requires mitochondrion-induced TGF-β1 in type 1 conventional dendritic cells

  • Text
  • PDF
Abstract

The role of conventional type 1 DCs (cDC1s) in tolerance induction to solid organ allografts is unknown and important for strategies that seek to prolong allograft viability. Using a murine model deficient in cDC1s, we report cDC1s are required for donor antigen and costimulation blockade (DST + CoB) tolerance induction and survival of cardiac allografts. cDC1 deficiency led to decreases in CD4+CD25+FoxP3+ T cells within allograft and spleen tissue of transplant recipients, and this was found to be antigen specific. Donor antigen stimulation induced TGF-β1 expression in both in vivo cDC1s and in vitro Flt3L-derived cDC1s. Genetic deletion of TGF-β1 in cDC1s prevented induction of antigen-specific CD4+CD25+FoxP3+ T cells and was associated with cardiac allograft rejection. In parallel, single-cell RNA sequencing and metabolic analysis revealed upregulation of cDC1 mitochondrial metabolic signatures after in vivo exposure to DST + CoB. Genetic inactivation of cDC1 mitochondrial metabolism reduced expression of cDC1 TGF-β1, decreased antigen-specific Treg populations, and impaired allograft tolerance. Taken together, our findings implicate cDC1s in strategies to preserve solid organ allografts and also implicate mitochondrial metabolism of cDC1s as a molecular mechanism to enhance the generation of antigen-specific CD4+CD25+FoxP3+ T cells through TGF-β1.

Authors

Samantha L. Schroth, Lei Zhang, Rebecca T.L. Jones, Kristofor Glinton, Nikita L. Mani, Hiroyasu Inui, Jesse T. Davidson, Samuel E. Weinberg, Navdeep S. Chandel, Maria-Luisa Alegre, Edward B. Thorp

×

To the Editor: Cardiac transplantation is a lifesaving procedure for patients with complex congenital heart diseases and end-stage heart failure. Unfortunately, rejection remains common owing to limitations in current immunosuppressive strategies and alternative therapies are needed. Among proposed strategies, costimulation blockade (CSB) represents a promising approach, promoting tolerance rather than suppressing alloimmune responses. CSB with CTLA4-Ig and anti-CD40L antibodies is efficacious in experimental models and early clinical studies in islet and kidney transplantation (1, 2). How CSB modulates recipient immune responses remains incompletely understood.

Costimulation pathways signal bidirectionally, influencing both antigen-presenting cells (APCs) and T cells. While CSB’s effect on T cells is well studied, less is known about its effects on APCs. To investigate how CSB and a conventional immunosuppressant (cyclosporine [CSA]) influence APCs in cardiac allografts, we performed single-cell RNA sequencing (scRNA-Seq) on murine hearts 7 days after transplant. BALB/c donor hearts were transplanted into B6 Zbtb46gfp/+ recipients treated with either CSB (anti-CD40L and CTLA4-Ig) or CSA. Histologically, CSA-treated grafts exhibited increased cellular infiltration compared with CSB-treated counterparts (Supplemental Figure 1A; supplemental material available online with this article; https://doi.org/10.1172/JCI192811DS1). Flow cytometry–isolated mononuclear phagocytes were used for 10X Genomics scRNA-seq, yielding 14,524 high-quality cells (Supplemental Figure 1, B and C), including monocyte, macrophage, and classical DC (cDC) subsets (Figure 1A and Supplemental Figure 1, D and E). CSB-treated grafts were enriched for recipient cDCs (GFP+), whereas CSA-treated grafts had increased monocytes and macrophages (Figure 1B). Reference mapping of naive hearts, syngeneic grafts, and a second model of allograft rejection (low-dose CTLA4-Ig) highlighted that cDC enrichment was CSB specific (Supplemental Figure 1F). Differential gene expression analysis revealed upregulation of genes involved in cDC activation, antigen presentation, and immunoregulation in CSB samples (Supplemental Figure 1, G and H). Flow cytometry and immunostaining confirmed increased frequencies of GFP+ cDCs in CSB-treated allografts, with a shift toward higher proportions of type 1 cDCs (cDC1s) (Figure 1, C–E, and Supplemental Figure 2A). Moreover, cDCs in CSB-treated allografts expressed PDL1 at a higher frequency than cDCs in CSA-treated allografts (Figure 1, D and E).

Type 1 classical DCs are necessary for cardiac allograft acceptance and modFigure 1

Type 1 classical DCs are necessary for cardiac allograft acceptance and modulate T cell phenotypes. (A) scRNA-Seq UMAP and (B) composition plot of mononuclear phagocytes sorted from allografts 7 days after transplantation of BALB/c hearts into CSB- (n = 3) or CSA-treated (n = 3) B6 WT mice. (C) Flow cytometry quantification of cDC1s within allografts of CSB- (n = 3) and CSA-treated (n = 3) B6 WT recipients at 7 days after transplant. (D) Immunostaining of graft-infiltrating cDCs (GFP+) and PDL1 in CSB- (n = 6) and CSA-treated (n = 5) B6 Zbtb46gfp/+ recipients of BALB/c hearts at 7 days after transplant. Scale bar: 50 μm. (E) Quantification of GFP+ and PDL1+ cells in CSB- (n = 6) and CSA-treated (n = 5) allografts at 7 days after transplant into B6 Zbtb46gfp/+ mice. (F) Kaplan-Meier survival curves of BALB/c hearts after transplantation into CSB-treated B6 WT, Δ1+2+3, and Irf8+32–/– mice (n = 10 per condition). (G) Histology (H&E, Verhoeff-Van Gieson elastin stain) of allografts from B6 WT, Δ1+2+3, and Irf8+32–/– recipients at 60 days after transplant (WT, Δ1+2+3) or time of rejection (Irf8+32–/–). Fractions in the top right corner indicate the number of samples with histology matching the representative image out of the total samples in each cohort. Scale bar: 50 μm (top); 20 μm (bottom). (H) Immunostaining of CD4+ and CD8+ T cells in allografts of B6 Irf8+32–/– (n = 3) and WT (n = 3) recipients at 14 days after transplant. Scale bar: 50 μm. (I) scRNA-Seq UMAP and (J) composition plot of subclustered T cells B6 WT (n = 3) and Irf8+32–/– (n = 3) recipients. (K) Pathway analysis of genes differentially expressed in T cells.

We next set out to define the requirement for recipient cDC1s and cDC2s in CSB-mediated long-term cardiac allograft acceptance. BALB/c hearts were transplanted into WT, Δ1+2+3 (cDC2-deficient), and Irf8+32–/– (cDC1-deficient) B6 CSB-treated recipients (Supplemental Figure 2B) (3, 4). While WT and cDC2-deficient recipients accepted cardiac allografts long-term, cDC1-deficient recipients rejected the transplanted hearts (Figure 1, F and G). Irf8+32–/– recipients exhibited intragraft infiltration of CD4+ and CD8+ T cells at day 14 after transplant and the time of rejection. WT and Δ1+2+3 recipients had significantly fewer T cells at both time points (Figure 1H and Supplemental Figure 2C). We also observed increased Foxp3+CD4+ T cells in allografts transplanted into WT versus Irf8+32–/– recipients, suggesting that cDC1s recruit regulatory T cells (Supplemental Figure 2D).

To examine if cDC1 deficiency impacts the composition and transcriptional state of intragraft T cells, BALB/c donor hearts were transplanted into CSB-treated B6 WT or Irf8+32–/– recipients. Extravascular immune cells were isolated from allografts 14 days after transplant by flow cytometry, and scRNA-Seq was performed, yielding 12,580 high-quality cells (Supplemental Figure 3, A and B). Allografts transplanted into Irf8+32–/– recipients exhibited shifts in CD4+ and CD8+ T cell phenotype (Figure 1, I and J, and Supplemental Figure 3, C–E). We observed an increase in Rora+CD4+ effector T cells in allografts from Irf8+32–/– recipients. Rora, a key regulator of Th17 cells, has been implicated in colitis, in which it drives T cell infiltration, activation, and prevention of apoptosis (5). Moreover, we observed marked reduction in a CD8+ T cell subset expressing Tcf7, Xcl1, and immunoregulatory genes (Cd200, Cd160, Lag3) in allografts from Irf8+32–/– recipients. Xcl1 is secreted by CD8+ T cells and is a ligand for Xcr1, a cDC1-specific receptor that regulates antigen presentation, regulatory T cell activation, and prevents intestinal inflammation (6). Pathway analysis revealed upregulation of IL-1, IL-5, TNF, and CD40L signaling in T cells from allografts transplanted into Irf8+32–/– recipients and enhanced immunoregulatory responses and T cell apoptosis in WT recipients (Figure 1K).

Collectively, we demonstrate that cDC1s expand in response to CSB and are essential for long-term allograft acceptance. CSB facilitates recruitment of immunoregulatory cDC1s, which modulate T cell phenotypes. CSB represents a tractable approach to achieve organ transplant tolerance in the clinical setting. Unlike other tolerance protocols, CSB does not necessitate exposure of the recipient to donor cells or tissues and instead only involves perioperative treatment with CTLA4-Ig and anti-CD40L antibodies. Identification of cDC1s as a key cell involved in cardiac allograft acceptance provides a critical clue regarding underlying mechanisms. Future studies dissecting tolerogenic cDC1 effector mechanisms may lead to improved CSB regimens, methodologies to measure CSB efficacy, and platforms to predict posttransplant outcomes.

For detailed methods, information regarding sex as a biological variable, statistics, study approval, data availability, author contributions, and acknowledgments, see the Supplemental Methods.

Supplemental material

View Supplemental data

View Supporting data values

Footnotes

Conflict of interest: The authors have declared that no conflict of interest exists.

Copyright: © 2025, Owen et al. This is an open access article published under the terms of the Creative Commons Attribution 4.0 International License.

Reference information: J Clin Invest. 2025;135(18):e192811. https://doi.org/10.1172/JCI192811.

See the related article at Treg activation during allograft tolerance induction requires mitochondrion-induced TGF-β1 in type 1 conventional dendritic cells.

References
  1. Larsen CP, et al. Long-term acceptance of skin and cardiac allografts after blocking CD40 and CD28 pathways. Nature. 1996;381(6581):434–438.
    View this article via: CrossRef PubMed Google Scholar
  2. Anwar IJ, et al. The anti-CD40L monoclonal antibody AT-1501 promotes islet and kidney allograft survival and function in nonhuman primates. Sci Transl Med. 2023;15(711):eadf6376.
    View this article via: CrossRef PubMed Google Scholar
  3. Liu TT, et al. Ablation of cDC2 development by triple mutations within the Zeb2 enhancer. Nature. 2022;607(7917):142–148.
    View this article via: CrossRef PubMed Google Scholar
  4. Durai V, et al. Cryptic activation of an Irf8 enhancer governs cDC1 fate specification. Nat Immunol. 2019;20(9):1161–1173.
    View this article via: CrossRef PubMed Google Scholar
  5. Chi X, et al. RORα is critical for mTORC1 activity in T cell-mediated colitis. Cell Rep. 2021;36(11):109682.
    View this article via: CrossRef PubMed Google Scholar
  6. Moreira TG, et al. PD-L1+ and XCR1+ dendritic cells are region-specific regulators of gut homeostasis. Nat Commun. 2021;12(1):4907.
    View this article via: CrossRef PubMed Google Scholar
Version history
  • Version 1 (July 8, 2025): In-Press Preview
  • Version 2 (September 16, 2025): Electronic publication

Article tools

  • View PDF
  • Download citation information
  • Send a comment
  • Terms of use
  • Standard abbreviations
  • Need help? Email the journal

Metrics

  • Article usage
  • Citations to this article

Go to

  • Top
  • Supplemental material
  • Footnotes
  • References
  • Version history
Advertisement
Advertisement

Copyright © 2025 American Society for Clinical Investigation
ISSN: 0021-9738 (print), 1558-8238 (online)

Sign up for email alerts