Abstract
The primary driver of type I diabetes is the autoimmune T cells that destroy insulin-producing β-cells within the islets of Langerhans in the pancreas1. Pancreatic islet macrophages have also been variably linked to disease onset and progression. As macrophage-mediated removal of dying cells through efferocytosis regulates tissue homeostasis and immune responses2, here we investigated how efferocytosis by intra-islet macrophages influences the immune environment of pancreatic islets. Using a series of complementary omics-based and functional approaches, we identify a subset of anti-inflammatory intra-islet efferocytic macrophages (e-Mac) within the pancreas of mice and humans. When limited β-cell apoptosis is induced in vivo in wild-type C57BL/6 mice and diabetic-prone NOD mice, islet macrophages adopt this e-Mac phenotype without an apparent increase in the total numbers of intra-islet macrophages. Such limited β-cell apoptosis and increase in e-Mac numbers led to long-term suppression of autoimmune diabetes in NOD mice. This e-Mac phenotype could also be recapitulated ex vivo by co-culturing macrophages with apoptotic β-cells. Mechanistically, the e-Mac-enriched populations imparted an anergic-like state on CD4+ T cells ex vivo and promoted accumulation of such anergic-like CD4+ T cells in vivo within the islets. Analysing macrophage–T cell interactions within pancreatic islets using NicheNet and targeted experimental validation, we identify the IGF-1–IGF1R axis as a contributor to the anergic-like T cell phenotype in the islets. Collectively, these data advance a concept that efferocytosis-associated reprogramming of the islet macrophages and the subsequent influence on the adaptive immune response could be beneficial in modulating diabetic autoimmunity.
This is a preview of subscription content, access via your institution
Access options
Access Nature and 54 other Nature Portfolio journals
Get Nature+, our best-value online-access subscription
$32.99 / 30 days
cancel any time
Subscribe to this journal
Receive 51 print issues and online access
$199.00 per year
only $3.90 per issue
Buy this article
- Purchase on SpringerLink
- Instant access to full article PDF
Prices may be subject to local taxes which are calculated during checkout





Similar content being viewed by others
Data availability
All transcriptional data generated in the current study were deposited at the GEO and are publicly available: GSE304628 (scRNA-seq mouse islets), GSE304620 (scRNA-seq human donor islets) and GSE304616 (bulk RNA-seq CD4+ T cells, mouse). Publicly available datasets reanalysed in this study include the following: GSE244278 (anergic CD4+ T cell RNA-seq) and GSE143739 (anergic CD4+ T cell microarray). For comparing the single-cell transcriptional profile of islet macrophages with transcriptomics of macrophages from other tissues, the GSM7049631 (mouse microglia)36 and GSE128518 (macrophages in adipose tissue of mice fed a high-fat diet)37 datasets were used. Source data are provided with this paper.
References
Herold, K. C., Vignali, D. A. A., Cooke, A. & Bluestone, J. A. Type 1 diabetes: translating mechanistic observations into effective clinical outcomes. Nat. Rev. Immunol. 13, 243–256 (2013).
Doran, A. C., Yurdagul, A. & Tabas, I. Efferocytosis in health and disease. Nat. Rev. Immunol. 20, 254–267 (2020).
Lavin, Y. et al. Tissue-resident macrophage enhancer landscapes are shaped by the local microenvironment. Cell 159, 1312–1326 (2014).
Ginhoux, F. & Guilliams, M. Tissue-resident macrophage ontogeny and homeostasis. Immunity 44, 439–449 (2016).
Park, M. D., Silvin, A., Ginhoux, F. & Merad, M. Macrophages in health and disease. Cell 185, 4259–4279 (2022).
Calderon, B. et al. The pancreas anatomy conditions the origin and properties of resident macrophages. J. Exp. Med. 212, 1497–1512 (2015).
Brissova, M. et al. Islet microenvironment, modulated by vascular endothelial growth factor-A signaling, promotes β cell regeneration. Cell Metab. 19, 498–511 (2014).
Riley, K. G. et al. Macrophages are essential for CTGF-mediated adult β-cell proliferation after injury. Mol. Metab. 4, 584–591 (2015).
Thai, L. M. et al. β-Cell function is regulated by metabolic and epigenetic programming of islet-associated macrophages, involving Axl, Mertk, and TGFβ receptor signaling. iScience 26, 106477 (2023).
Banaei-Bouchareb, L. et al. Insulin cell mass is altered in Csf1op/Csf1op macrophage-deficient mice. J. Leukoc. Biol. 76, 359–367 (2004).
Oschilewski, U., Kiesel, U. & Kolb, H. Administration of silica prevents diabetes in BB-rats. Diabetes 34, 197–199 (1985).
Carrero, J. A. et al. Resident macrophages of pancreatic islets have a seminal role in the initiation of autoimmune diabetes of NOD mice. Proc. Natl Acad. Sci. USA 114, E10418–E10427 (2017).
Chen, D., Thayer, T. C., Wen, L. & Wong, F. S. Mouse models of autoimmune diabetes: the nonobese diabetic (NOD) mouse. Methods Mol. Biol. 2128, 87–92 (2020).
Calderon, B., Carrero, J. A. & Unanue, E. R. The central role of antigen presentation in islets of Langerhans in autoimmune diabetes. Curr. Opin. Immunol. 26, 32–40 (2014).
Mohan, J. F. et al. Imaging the emergence and natural progression of spontaneous autoimmune diabetes. Proc. Natl Acad. Sci. USA 114, E7776–E7785 (2017).
Zakharov, P. N., Hu, H., Wan, X. & Unanue, E. R. Single-cell RNA sequencing of murine islets shows high cellular complexity at all stages of autoimmune diabetes. J. Exp. Med. 217, e20192362 (2020).
Pugliese, A. Autoreactive T cells in type 1 diabetes. J. Clin. Invest. 127, 2881–2891 (2017).
Delong, T. et al. Pathogenic CD4 T cells in type 1 diabetes recognize epitopes formed by peptide fusion. Science 351, 711–714 (2016).
Wan, X. et al. Pancreatic islets communicate with lymphoid tissues via exocytosis of insulin peptides. Nature 560, 107–111 (2018).
DiLorenzo, T. P. & Serreze, D. V. The good turned ugly: immunopathogenic basis for diabetogenic CD8+ T cells in NOD mice. Immunol. Rev. 204, 250–263 (2005).
Noble, J. A. & Erlich, H. A. Genetics of type 1 diabetes. Cold Spring Harb. Perspect. Med. 2, a007732 (2012).
Wang, Y. J. et al. Multiplexed in situ imaging mass cytometry analysis of the human endocrine pancreas and immune system in type 1 diabetes. Cell Metab. 29, 769–783 (2019).
Fadok, V. A. et al. Macrophages that have ingested apoptotic cells in vitro inhibit proinflammatory cytokine production through autocrine/paracrine mechanisms involving TGF-beta, PGE2, and PAF. J. Clin. Invest. 101, 890–898 (1998).
Morioka, S. et al. Efferocytosis induces a novel SLC program to promote glucose uptake and lactate release. Nature 563, 714–718 (2018).
Tufan, T. et al. Rapid unleashing of macrophage efferocytic capacity via transcriptional pause release. Nature 628, 408–415 (2024).
Hugues, S. et al. Tolerance to islet antigens and prevention from diabetes induced by limited apoptosis of pancreatic beta cells. Immunity 16, 169–181 (2002).
Boada-Romero, E., Martinez, J., Heckmann, B. L. & Green, D. R. The clearance of dead cells by efferocytosis. Nat. Rev. Mol. Cell Biol. 21, 398–414 (2020).
Turley, S., Poirot, L., Hattori, M., Benoist, C. & Mathis, D. Physiological β cell death triggers priming of self-reactive T cells by dendritic cells in a type-1 diabetes model. J. Exp. Med. 198, 1527–1537 (2003).
Lenzen, S. The mechanisms of alloxan- and streptozotocin-induced diabetes. Diabetologia 51, 216–226 (2008).
Furman, B. L. Streptozotocin‐induced diabetic models in mice and rats. Curr. Protoc. 1, e78 (2021).
Ferris, S. T. et al. The islet-resident macrophage is in an inflammatory state and senses microbial products in blood. J. Exp. Med. 214, 2369–2385 (2017).
Brosseau, C., Colas, L., Magnan, A. & Brouard, S. CD9 tetraspanin: a new pathway for the regulation of inflammation? Front. Immunol. 9, 2316 (2018).
Sano, H. et al. Critical role of galectin-3 in phagocytosis by macrophages. J. Clin. Invest. 112, 389–397 (2003).
Lemke, G. & Rothlin, C. V. Immunobiology of the TAM receptors. Nat. Rev. Immunol. 8, 327–336 (2008).
Lindsay, R. S. et al. MERTK on mononuclear phagocytes regulates T cell antigen recognition at autoimmune and tumor sites. J. Exp. Med. 218, e20200464 (2021).
Millet, A., Ledo, J. H. & Tavazoie, S. F. An exhausted-like microglial population accumulates in aged and APOE4 genotype Alzheimer’s brains. Immunity 57, 153–170 (2024).
Jaitin, D. A. et al. Lipid-associated macrophages control metabolic homeostasis in a Trem2-dependent manner. Cell 178, 686–698 (2019).
Keren-Shaul, H. et al. A unique microglia type associated with restricting development of Alzheimer’s disease. Cell 169, 1276–1290 (2017).
Carrero, J. A., Calderon, B., Towfic, F., Artyomov, M. N. & Unanue, E. R. Defining the transcriptional and cellular landscape of type 1 diabetes in the NOD mouse. PLoS ONE 8, e59701 (2013).
Calderon, B., Carrero, J. A., Miller, M. J. & Unanue, E. R. Cellular and molecular events in the localization of diabetogenic T cells to islets of Langerhans. Proc. Natl Acad. Sci. USA 108, 1561–1566 (2011).
Katz, J. D., Wang, B., Haskins, K., Benoist, C. & Mathis, D. Following a diabetogenic T cell from genesis through pathogenesis. Cell 74, 1089–1100 (1993).
Gonzalez, A. et al. Genetic control of diabetes progression. Immunity 7, 873–883 (1997).
Trefzer, A. et al. Dynamic adoption of anergy by antigen-exhausted CD4+ T cells. Cell Rep. 34, 108748 (2021).
Titcombe, P. J., Silva Morales, M., Zhang, N. & Mueller, D. L. BATF represses BIM to sustain tolerant T cells in the periphery. J. Exp. Med. 220, e20230183 (2023).
Kalekar, L. A. et al. CD4+ T cell anergy prevents autoimmunity and generates regulatory T cell precursors. Nat. Immunol. 17, 304–314 (2016).
Price, J. D., Hotta-Iwamura, C., Zhao, Y., Beauchamp, N. M. & Tarbell, K. V. DCIR2+ cDC2 DCs and Zbtb32 restore CD4+ T-cell tolerance and inhibit diabetes. Diabetes 64, 3521–3531 (2015).
Shin, H. M. et al. Transient expression of ZBTB32 in anti-viral CD8+ T cells limits the magnitude of the effector response and the generation of memory. PLoS Pathog. 13, e1006544 (2017).
Alroy, I., Towers, T. L. & Freedman, L. P. Transcriptional repression of the interleukin-2 gene by vitamin D3: direct inhibition of NFATp/AP-1 complex formation by a nuclear hormone receptor. Mol. Cell. Biol. 15, 5789–5799 (1995).
Martinez, R. J. et al. Arthritogenic self-reactive CD4+ T cells acquire an FR4hiCD73hi anergic state in the presence of Foxp3+ regulatory T cells. J. Immunol. 188, 170–181 (2012).
Browaeys, R., Saelens, W. & Saeys, Y. NicheNet: modeling intercellular communication by linking ligands to target genes. Nat. Methods 17, 159–162 (2020).
Nackiewicz, D. et al. Islet macrophages shift to a reparative state following pancreatic beta-cell death and are a major source of islet insulin-like growth factor-1. iScience 23, 100775 (2020).
Shapiro, M. R. et al. Insulin-like growth factor dysregulation both preceding and following type 1 diabetes diagnosis. Diabetes 69, 413–423 (2020).
Han, C. Z. et al. Macrophages redirect phagocytosis by non-professional phagocytes and influence inflammation. Nature 539, 570–574 (2016).
Mallol, C. et al. AAV-mediated pancreatic overexpression of Igf1 counteracts progression to autoimmune diabetes in mice. Mol. Metab. 6, 664–680 (2017).
Finegood, D. T., Scaglia, L. & Bonner-Weir, S. Dynamics of beta-cell mass in the growing rat pancreas. Estimation with a simple mathematical model. Diabetes 44, 249–256 (1995).
Scaglia, L., Cahill, C. J., Finegood, D. T. & Bonner-Weir, S. Apoptosis participates in the remodeling of the endocrine pancreas in the neonatal rat. Endocrinology 138, 1736–1741 (1997).
Trudeau, J. D. et al. Neonatal beta-cell apoptosis: a trigger for autoimmune diabetes? Diabetes 49, 1–7 (2000).
Kassem, S. A., Ariel, I., Thornton, P. S., Scheimberg, I. & Glaser, B. Beta-cell proliferation and apoptosis in the developing normal human pancreas and in hyperinsulinism of infancy. Diabetes 49, 1325–1333 (2000).
Ciecko, A. E. et al. Heterogeneity of islet-infiltrating IL-21+ CD4 T cells in a mouse model of type 1 diabetes. J. Immunol. 210, 935–946 (2023).
Foda, B. M. et al. The CD137 ligand is important for type 1 diabetes development but dispensable for the homeostasis of disease-suppressive CD137+FOXP3+ regulatory CD4 T cells. J. Immunol. 204, 2887–2899 (2020).
Mohan, J. F., Calderon, B., Anderson, M. S. & Unanue, E. R. Pathogenic CD4+ T cells recognizing an unstable peptide of insulin are directly recruited into islets bypassing local lymph nodes. J. Exp. Med. 210, 2403–2414 (2013).
Goudy, K. S. et al. Systemic overexpression of IL-10 induces CD4+CD25+ cell populations in vivo and ameliorates type 1 diabetes in nonobese diabetic mice in a dose-dependent fashion. J. Immunol. 171, 2270–2278 (2003).
Kolberg, L., Raudvere, U., Kuzmin, I., Vilo, J. & Peterson, H. gprofiler2—an R package for gene list functional enrichment analysis and namespace conversion toolset g:Profiler. F1000Research 9, ELIXIR-709 (2020).
Wu, T. et al. clusterProfiler 4.0: a universal enrichment tool for interpreting omics data. Innovation 2, 100141 (2021).
Zinselmeyer, B. H. et al. The resident macrophages in murine pancreatic islets are constantly probing their local environment, capturing beta cell granules and blood particles. Diabetologia 61, 1374–1383 (2018).
Love, M. I., Huber, W. & Anders, S. Moderated estimation of fold change and dispersion for RNA-seq data with DESeq2. Genome Biol. 15, 550 (2014).
Zhu, A., Ibrahim, J. G. & Love, M. I. Heavy-tailed prior distributions for sequence count data: removing the noise and preserving large differences. Bioinformatics 35, 2084–2092 (2019).
Blighe, K. et al. EnhancedVolcano: publication-ready volcano plots with enhanced colouring and labeling. https://doi.org/10.18129/B9.BIOC.ENHANCEDVOLCANO (Bioconductor, 2018).
Zhang, Y., Parmigiani, G. & Johnson, W. E. ComBat-seq: batch effect adjustment for RNA-seq count data. NAR Genom. Bioinform. 2, lqaa078 (2020).
Ritchie, M. E. et al. limma powers differential expression analyses for RNA-sequencing and microarray studies. Nucleic Acids Res. 43, e47 (2015).
Acknowledgements
We thank the members of the K.S.R. and X.W. laboratories for inputs and suggestions; J. I. Etchegaray, S. Arandjelovic, C. Maueröder, N. Srivastava, P. Mehrotra, M. Colonna and the members of the Colonna laboratory for inputs; C. Rothlin for providing the Mertkfl/fl mice; R. Birge for providing some reagents; the staff at the following Washington University School of Medicine cores: the Flow Cytometry Core, WUCCI Imaging Core and the Genome Technology Access Center at the McDonnell Genome Institute for RNA-seq library preparation and sequencing; A. Srivastav for help with some mouse studies and W. Beatty for help with electron microscopy imaging. K.S.R. is supported by supported by R01AI159551 (NIAID) and BJC Investigator Funds from the Washington University School of Medicine; X.W. is supported by R01AI162591 (NIAID); E.K. is supported by 1R01CA245277-01A1 (NCI), 5R01AR075959-02 (NIAMS) and a National Psoriasis Foundation Grant. Graphics in Figs. 1h,l,m, 2b, 3a,b, 4d,h, 5a,e,f and Extended Data Figs. 4a,b, 5a, 6a, 8f and 10d contain schematics created using a paid subscription of BioRender.
Author information
Authors and Affiliations
Contributions
P.N.Z., K.S.R., X.W. and E.R.U. designed the experiments and interpreted the data. K.S.R. and P.N.Z. wrote the manuscript with input from all of the authors. X.W. provided critical experimental inputs and reagents. P.N.Z. carried out most of the experiments, analysed the data and performed bioinformatics analysis of scRNA-seq and bulk RNA-seq data. C.S.C., P.N.Z. and O.J.P. carried out confocal microscopy. C.S.C. and P.N.Z. carried out image processing. C.S.C. performed 3D image rendering. P.N.Z. and O.J.P. performed pancreatic islet isolation. L.G. and E.K. provided scRNA-seq data for human donor C. B.B. carried out annexin V purification. A.N.V. assisted in several experiments.
Corresponding authors
Ethics declarations
Competing interests
The authors declare no competing interests.
Peer review
Peer review information
Nature thanks Raymond Birge, Edward Thorp, and the other, anonymous, reviewer(s) for their contribution to the peer review of this work.
Additional information
Publisher’s note Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional affiliations.
Extended data figures and tables
Extended Data Fig. 1 Efferocytosis by islet macrophages following STZ1-low treatment in C57BL/6 mice.
a. Immunofluorescent confocal microscopy imaging showing confocal planes of the pancreatic islets of C57BL/6 mice 12 h after STZ1-low or control vehicle treatment. Green – apoptotic cells (cleaved caspase-3, CC3), blue – nuclei (Hoechst). Scale bar 30 µm. b. Quantification of fraction of apoptotic cells (CC3+) relative to the total cell number in the islets (Hoechst+). n = 9 islets in both conditions. c. Confocal images of islets taken 12 h after STZ1-low treatment showing X-Y, Y-Z, and X-Z views of efferocytic events. Red – insulin, white – macrophages (F4/80), green – apoptotic cells (CC3), blue – nuclei (Hoechst). The CC3/insulin double positive areas inside the macrophages are considered as efferocytic events. The top row of images corresponds to (Fig. 1b). Scale bar – 30 µm. d. Quantification of efferocytic events via Imaris software automatic counting (see Methods). n = 9(Vehicle), n = 8(STZ1-low). e. Percentage of islet macrophages (Live, CD45+, F4/80+, CD11c+) relative to all islet cells based on flow cytometric analysis 10 days after treatment with STZ1-low or vehicle (C57BL/6 mice). n = 8(Vehicle), n = 11(STZ1-low) mice. Statistics - two-tailed unpaired t-test, data shown as mean ± SEM.
Extended Data Fig. 2 scRNASeq of islet leukocytes following limited β-cell death.
a. The UMAP plot showing scRNASeq analysis of CD45+ cells from the islets of C57BL/6 J mice 10 days after STZ1-low or vehicle treatment. b. Phenotypic markers expressed by cells from (a). c. Relative fractions of cells identified on (a) and (b).
Extended Data Fig. 3 Islet e-Mac shows similarity with pathology-associated macrophages from other tissues.
a. UMAP plots show scRNASeq of islet macrophages (C57BL/6 mice) and two publicly available scRNASeq datasets: i) microglia in the brain of an Alzheimer’s disease mouse model (GSM7049631 (ref. 36)), and ii) macrophages in adipose tissue of mice fed a high-fat-diet (GSE128518 (ref. 37)). Expression of some marker genes are shown on the right. b. Venn diagram illustrates overlaps among gene signatures from macrophage subsets in (a).
Extended Data Fig. 4 Gal-3 upregulation in islet and peritoneal macrophages following apoptotic β-cell uptake ex vivo.
a. Isolation of islet macrophages using CD11c+ magnetic beads (MACS) (non-autoimmune C57BL/6 J mice). Flow cytometric analysis on dispersed islet cells before enrichment (left), and MACS-enriched Live, CD45+ F4/80+ CD11c+ macrophages (right). b. Schematic of the assay when primary islet macrophages co-cultured with live or apoptotic β-cells (UV-irradiated Min6 cells). c. Flow cytometric evaluation of e-Mac frequencies in the co-culture experiment (b). Quantification is shown on the right. n = 4(UT), n = 2(Live), n = 8(Apo) biological replicates over N = 4 independent experiments for “UT” and “Apo” and N = 2 for “Live”. Two-tailed unpaired t-test with Welch’s correction. d. Flow cytometric analysis showing peritoneal macrophages. Peritoneal lavage cells were collected from C57BL/6 mice, plated for 2 h, washed to remove unbound cells, and analysed by flow cytometry 24 h after the beginning of the incubation. e. Flow cytometric analysis showing engulfment of apoptotic β-cells by peritoneal macrophages. Apoptotic β-cells (Min6 cell line) were labelled with pHrodo red dye and co-cultured with pMacs for 2 h; unbound apoptotic cells were then washed away, and incubation continued. The macrophage-to-β-cell ratio was 1:3. Flow cytometry was done 16 h after beginning of the incubation. f. Flow cytometric analysis of peritoneal macrophages after co-culture with live or apoptotic β-cells (Min6). Two negative controls were used: cytochalasin D (CytoD, 1 µM) – inhibitor of cytoskeletal reorganization that prevents corpse uptake; and annexin V (20 µg/ml) that binds phosphatidylserine (PtdSer) and masks it from scavenger receptors on the phagocytic cells. g. Quantification of (f). n = 6 (No β-cell), n = 6 (Live β-cell), n = 6 (Apo β-cell, alone), n = 3 (Apo β-cell, +CytoD), n = 3 (Apo β-cell, +Ann V) biological replicates; N = 2. Statistics - two-tailed unpaired t-test. Data are representative of N = 3 independent experiments (a, b). The diagrams in a and b were created in BioRender. Ravichandran, K. (2025) https://BioRender.com/oc6ssn7; Ravichandran, K. (2025) https://BioRender.com/laier1m.
Extended Data Fig. 5 Both e-Mac and non-e-Mac islet macrophages can uptake apoptotic β-cells.
a. Representative confocal images of the pancreatic islets of NOD mice 12 h after STZ1-low or control vehicle treatment. Green – apoptotic cells (cleaved caspase-3, CC3), blue – nuclei (Hoechst), red – Gal-3, white – F4/80, yellow – insulin. Scale bars: 20 µm (left, whole-islet view); 2 µm (right, zoomed-in view). b. Expression of Mer-TK by islet macrophages in Mertkfl/fl/Cx3cr1cre/wt (Mertk cKO) and Mertkfl/fl/Cx3cr1wt/wt, C57BL/6 mice. Data are representative of N = 3 independent experiments. The diagrams in a were created in BioRender. Ravichandran, K. (2025) https://BioRender.com/lwltf25.
Extended Data Fig. 6 Intra-islet macrophages are the primary cells engulfing apoptotic β-cells.
a. Representative confocal images of the pancreatic islets of NOD mice 12 h after STZ1-low or control vehicle treatment. Green – apoptotic cells (cleaved caspase-3, CC3), blue – nuclei (Hoechst), red – CD11c, white – F4/80, yellow – insulin. Scale bars: 20 µm (left, whole-islet view); 2 µm (right, zoomed-in view). b. Quantification of the fraction of MФ and DC among all islet cells (top), and absolute number of each cell type per islet (bottom). c. Quantification of the fraction of MФ and DC containing apoptotic remnants (CC3+insulin+) among all islet cells (top), and absolute number of MФ and DC with internalized apoptotic material (bottom). Scale bars: 20 µm (left, whole-islet view); 2 µm (right, zoomed-in view). Statistics - paired t-test; N = 2. The diagram in a was created in BioRender. Ravichandran, K. (2025) https://BioRender.com/lwltf25.
Extended Data Fig. 7 Leukocytes from islets of NOD mice following limited β-cell death.
a. The UMAP plot showing CD45+ cells from the islets of NOD mice 10 days after inducing limited β-cell death with STZ1-low treatment. b. Differentially expressed genes defining cell types from (a).
Extended Data Fig. 8 Efferocytosis induces an anti-inflammatory program in islet macrophages.
a. Heatmap showing differentially expressed genes defining macrophage subsets in the islets of NOD mice at 5.5 weeks of age (10 days after treatment). b. Overrepresentation pathway analysis based on the gene signatures of e-Mac, Mac-2, and Mac-3 subsets (hypergeometric test). c. Venn diagrams comparing gene signature of e-Mac with those of other macrophage subsets. Note that gene signatures of Mac-1 and e-Mac have overlap. d. Examples of islet infiltration scores based on hematoxylin and eosin (H&E) staining of pancreas sections of 30-week-old NOD female mice treated at 4 weeks with either STZ1-low or vehicle. Scale bar – 20 µm. N = 2. e. Flow cytometric chart of CD45+ cells from pancreatic islets isolated from NOD.Rag1−/− mice (aged 4–6 weeks; pooled from 5 mice). Representative of N = 4 independent experiments. f. Flow cytometric charts of experiment in which naive CD4+ T cells were activated by islet macrophages or by conventional dendritic cells (cDC) from spleens and lymph nodes. Islet macrophages from either STZ1-low- or vehicle-treated NOD.Rag1−/− mice were co-cultured with CD4+ T cells without adding cognate antigen for 96 h. When conventional dendritic cells were used as antigen-presenting cells, 0.5 µM BDC mimotope was added when indicated. Conventional dendritic cells were isolated from spleen and lymph nodes of NOD.Rag1−/− mice. (See also Fig. 3b). The diagram in f was created in BioRender. Ravichandran, K. (2025) https://BioRender.com/mrggrwq.
Extended Data Fig. 9 CD4+ T cells acquire partial anergic-like phenotype after activation by efferocytosis-associated islet macrophages both ex vivo and in vivo.
a. Peritoneal macrophages isolated from NOD mouse using magnetic-activated cell sorting (MACS). CD11bpos macrophages are enriched into Tim-4hi large peritoneal macrophages (LPM, expressing low level of MHC-II) and Tim-4low small peritoneal macrophages (SPM, MHC-IIhi). Representative of N = 3 independent experiments. b. Antigen presentation assay in which naive TCR transgenic BDC2.5 CD4+ T cells were co-cultured for 3 days with SPM or LPM at indicated concentrations of antigenic peptide; N = 2. c. Gene set enrichment analysis (GSEA) plot showing enrichment of gene signature of CD4+ T cells activated by “e-Mac enriched” macrophages interrogated against publicly available transcriptional dataset comparing anergic and naive CD4+ T cells (GSE14373943). d. Summary plot of GSEA using CD4+ T cells gene signature induced by islet macrophages from STZ1-low- or vehicle- treated mice (“e-Mac enriched” and “Vehicle” correspondingly). Differential expression comparisons between anergic cells and either naive or antigen-experienced CD4+ T cells were used as reference 43 (GSE143739). The CD4+ T cells in the reference dataset were exposed to different levels of antigen. e. GSEA pathways comparing gene expression pathways upregulated in CD4+ T cells activated by islet macrophages from STZ1-low- versus vehicle- treated mice. Gene Ontology MSigDB (GO:BP). f. Heatmap showing differentially expressed genes among CD4+ T cell subsets in the islets of NOD mice 10 days after the treatment with either STZ1-low- or control vehicle. g. GSEA plot showing islet anergic-like CD4+ T cells gene signature interrogated against bulk transcriptional dataset43 (GSE14373943) comparing anergic and naive cells. NES, normalized enrichment score. Statistics: weighted Kolmogorov-Smirnov test (c, e, g).
Extended Data Fig. 10 Macrophages from 12-week-old NOD mice and human islet leukocytes analysed by scRNASeq.
a. scRNASeq analysis (UMAP plots) showing macrophage subsets from pancreatic islets at 12 weeks of age (late pre-diabetic) from NOD mice treated with STZ1-low or vehicle at 4 weeks of age (at early pre-diabetic stage). b. Markers of macrophage subsets from (a). c. Reanalysis of a publicly available dataset showing Igf1r expression across CD4+ T cell subsets from spleen and lymph node (GSE24427844). (Wald statistics, DESeq2). Data are presented as mean values ± SEM, 3 biological replicates over N = 1 RNASeq experiment. d. Flow cytometric analysis of human donor pancreatic islet cells. Islet macrophages were identified as Live, CD45+ CX3CR1+ CD11c+ cells. Representative of N = 2. e. Human islet leukocytes analysed by scRNASeq, 3 individual donors. CD45+ cells were FACS-purified as shown on (d), left. Transcriptional data was integrated between the donors and subjected to clustering. f,g. Markers differentially expressed between the identified immune cell types in human pancreatic islets. The diagram in d was created in BioRender. Ravichandran, K. (2025) https://BioRender.com/ddu5zzj.
Supplementary information
Supplementary Videos 1–3
Representative videos showing an independent event of macrophage efferocytosis in the islets taken 12 h after STZ1-low treatment. Red, insulin; white, macrophages (F4/80); green, apoptotic cells (cleaved caspase 3, CC3); blue, nuclei (Hoechst). The CC3/insulin double-positive areas inside the macrophages are considered to be efferocytic events. Scale bar, 2 µm.
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.
About this article
Cite this article
Zakharov, P.N., Chowdhury, C.S., Peterson, O.J. et al. Efferocytic remodelling of pancreatic islet macrophages by limited β-cell death. Nature (2025). https://doi.org/10.1038/s41586-025-09560-4
Received:
Accepted:
Published:
DOI: https://doi.org/10.1038/s41586-025-09560-4