Abstract
CD8+ T cells are major mediators of antiviral and antitumor immunity. During persistent antigen stimulation as in chronic infection and cancer, however, they differentiate into exhausted T cells that display impaired functionality. Precursors of exhausted T (TPEX) cells exhibit stem-like properties, including high proliferative, self-renewal and developmental potential, and are responsible for long-term CD8+ T cell responses against persistent antigens. Here we identify the chromatin organizer and transcriptional regulator SATB1 as a major regulator of exhausted CD8+ T cell differentiation. SATB1 was specifically expressed in TPEX cells where it limited population expansion and effector differentiation while preserving functionality of CD8+ T cells. SATB1 downregulation was required for TPEX cell-to-effector cell differentiation in chronic infection and contributed to coordinated effector and memory differentiation in acute viral infection. DNA binding of SATB1 regulated gene expression both dependent and independent of chromatin accessibility. Finally, SATB1 limited antitumor CD8+ and chimeric antigen receptor T cell immunity. Overall, our results identify SATB1 as a central regulator of precursor fate and effector differentiation of CD8+ T cells both in infection and in cancer.
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 /Â 30Â days
cancel any time
Subscribe to this journal
Receive 12 print issues and online access
$259.00 per year
only $21.58 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
References
McLane, L. M., Abdel-Hakeem, M. S. & Wherry, E. J. CD8 T cell exhaustion during chronic viral infection and cancer. Annu. Rev. Immunol. 37, 457â495 (2019).
Blank, C. U. et al. Defining âT cell exhaustionâ. Nat. Rev. Immunol. 19, 665â674 (2019).
Kallies, A., Zehn, D. & Utzschneider, D. T. Precursor exhausted T cells: key to successful immunotherapy?. Nat. Rev. Immunol. 20, 128â136 (2020).
Zehn, D., Thimme, R., Lugli, E., de Almeida, G. P. & Oxenius, A. âStem-likeâ precursors are the fount to sustain persistent CD8+ T cell responses. Nat. Immunol. 23, 836â847 (2022).
Rausch, L. & Kallies, A. Molecular mechanisms governing CD8 T cell differentiation and checkpoint inhibitor response in cancer. Annu. Rev. Immunol. 43, 515â543 (2025).
Thommen, D. S. & Schumacher, T. N. T cell dysfunction in cancer. Cancer Cell 33, 547â562 (2018).
Ribas, A. & Wolchok, J. D. Cancer immunotherapy using checkpoint blockade. Science 359, 1350â1355 (2018).
Albelda, S. M. CAR T cell therapy for patients with solid tumours: key lessons to learn and unlearn. Nat. Rev. Clin. Oncol. 21, 47â66 (2024).
Chan, J. D. et al. Cellular networks controlling T cell persistence in adoptive cell therapy. Nat. Rev. Immunol. 21, 769â784 (2021).
Martinez, G. J. et al. The transcription factor NFAT promotes exhaustion of activated CD8+ T cells. Immunity 42, 265â278 (2015).
Man, K. et al. Transcription factor IRF4 promotes CD8+ T cell exhaustion and limits the development of memory-like T cells during chronic infection. Immunity 47, 1129â1141 (2017).
Alfei, F. et al. TOX reinforces the phenotype and longevity of exhausted T cells in chronic viral infection. Nature 571, 265â269 (2019).
Scott, A. C. et al. TOX is a critical regulator of tumour-specific T cell differentiation. Nature 571, 270â274 (2019).
Liu, X. et al. Genome-wide analysis identifies NR4A1 as a key mediator of T cell dysfunction. Nature 567, 525â529 (2019).
Khan, O. et al. TOX transcriptionally and epigenetically programs CD8+ T cell exhaustion. Nature 571, 211â218 (2019).
Seo, H. et al. TOX and TOX2 transcription factors cooperate with NR4A transcription factors to impose CD8+ T cell exhaustion. Proc. Natl Acad. Sci. USA 116, 12410â12415 (2019).
He, R. et al. Follicular CXCR5-expressing CD8+ T cells curtail chronic viral infection. Nature 537, 412â428 (2016).
Im, S. J. et al. Defining CD8+ T cells that provide the proliferative burst after PD-1 therapy. Nature 537, 417â421 (2016).
Utzschneider, D. T. et al. T cell factor 1-expressing memory-like CD8+ T cells sustain the immune response to chronic viral infections. Immunity 45, 415â427 (2016).
Leong, Y. A. et al. CXCR5+ follicular cytotoxic T cells control viral infection in B cell follicles. Nat. Immunol. 17, 1187â1196 (2016).
Wu, T. et al. The TCF1âBCL6 axis counteracts type I interferon to repress exhaustion and maintain T cell stemness. Sci. Immunol. 1, eaai8593 (2016).
Miller, B. C. et al. Subsets of exhausted CD8+ T cells differentially mediate tumor control and respond to checkpoint blockade. Nat. Immunol. 20, 326â336 (2019).
Utzschneider, D. T. et al. Early precursor T cells establish and propagate T cell exhaustion in chronic infection. Nat. Immunol. 21, 1256â1266 (2020).
Siddiqui, I. et al. Intratumoral TCF1+PD-1+CD8+ T cells with stem-like properties promote tumor control in response to vaccination and checkpoint blockade immunotherapy. Immunity 50, 195â211 (2019).
Tsui, C. et al. MYB orchestrates T cell exhaustion and response to checkpoint inhibition. Nature 609, 354â360 (2022).
Wijesinghe, S. K. M. et al. Lymph-node-derived stem-like but not tumor-tissue-resident CD8+ T cells fuel anticancer immunity. Nat. Immunol. 26, 1367â1383 (2025).
Hudson, W. H. et al. Proliferating transitory T cells with an effector-like transcriptional signature emerge from PD-1+ stem-like CD8+ T cells during chronic infection. Immunity 51, 1043â1058 (2019).
Zander, R. et al. CD4+ T cell help is required for the formation of a cytolytic CD8+ T cell subset that protects against chronic infection and cancer. Immunity 51, 1028â1042 (2019).
Rahim, M. K. et al. Dynamic CD8+ T cell responses to cancer immunotherapy in human regional lymph nodes are disrupted in metastatic lymph nodes. Cell 186, 1127â1143 (2023).
Tsui, C. et al. Lymph nodes fuel KLF2-dependent effector CD8+ T cell differentiation during chronic infection and checkpoint blockade. Nat. Immunol. 26, 1752â1765 (2025).
Yamauchi, T. et al. T-cell CX3CR1 expression as a dynamic blood-based biomarker of response to immune checkpoint inhibitors. Nat. Commun. 12, 1402 (2021).
Sen, D. R. et al. The epigenetic landscape of T cell exhaustion. Science 354, 1165â1169 (2016).
Philip, M. et al. Chromatin states define tumour-specific T cell dysfunction and reprogramming. Nature 545, 452â456 (2017).
Ghoneim, H. E. et al. De novo epigenetic programs inhibit PD-1 blockade-mediated T cell rejuvenation. Cell 170, 142â157 (2017).
Hao, B. et al. An anti-silencer- and SATB1-dependent chromatin hub regulates Rag1 and Rag2 gene expression during thymocyte development. J. Exp. Med. 212, 809â824 (2015).
Kakugawa, K. et al. Essential roles of SATB1 in specifying T lymphocyte subsets. Cell Rep. 19, 1176â1188 (2017).
Chaurio, R. A. et al. TGF-β-mediated silencing of genomic organizer SATB1 promotes TFH cell differentiation and formation of intra-tumoral tertiary lymphoid structures. Immunity 55, 115â128 (2022).
Yasuda, K. et al. SATB1 regulates the effector program of encephalitogenic tissue TH17 cells in chronic inflammation. Nat. Commun. 10, 549 (2019).
Kitagawa, Y. et al. Guidance of regulatory T cell development by SATB1-dependent super-enhancer establishment. Nat. Immunol. 18, 173â183 (2017).
Nussing, S. et al. SATB1 ensures appropriate transcriptional programs within naive CD8+ T cells. Immunol. Cell Biol. 100, 636â652 (2022).
Stephen, T. L. et al. SATB1 expression governs epigenetic repression of PD-1 in tumor-reactive T cells. Immunity 46, 51â64 (2017).
Nussing, S. et al. Divergent SATB1 expression across human life span and tissue compartments. Immunol. Cell Biol. 97, 498â511 (2019).
Maekawa, Y. et al. NOTCH2 integrates signaling by the transcription factors RBP-J and CREB1 to promote T cell cytotoxicity. Nat. Immunol. 9, 1140â1147 (2008).
Sommer, D. et al. Efficient genome engineering by targeted homologous recombination in mouse embryos using transcription activator-like effector nucleases. Nat. Commun. 5, 3045 (2014).
Kohne, M. et al. SATB1 directs the differentiation of TH17 cells through suppression of IL-2 expression. Cell Rep. 44, 115866 (2025).
Gottimukkala, K. P. et al. Regulation of SATB1 during thymocyte development by TCR signaling. Mol. Immunol. 77, 34â43 (2016).
Gabriel, S. S. et al. Transforming growth factor-β-regulated mTOR activity preserves cellular metabolism to maintain long-term T cell responses in chronic infection. Immunity 54, 1698â1714 (2021).
Jadhav, R. R. et al. Epigenetic signature of PD-1+ TCF1+ CD8 T cells that act as resource cells during chronic viral infection and respond to PD-1 blockade. Proc. Natl Acad. Sci. USA 116, 14113â14118 (2019).
Kallies, A., Xin, A., Belz, G. T. & Nutt, S. L. Blimp-1 transcription factor is required for the differentiation of effector CD8+ T cells and memory responses. Immunity 31, 283â295 (2009).
Shin, H. et al. A role for the transcriptional repressor Blimp-1 in CD8+ T cell exhaustion during chronic viral infection. Immunity 31, 309â320 (2009).
Fransen, M. F. et al. Tumor-draining lymph nodes are pivotal in PD-1/PD-L1 checkpoint therapy. JCI Insight 3, e124507 (2018).
Huang, Q. et al. The primordial differentiation of tumor-specific memory CD8+ T cells as bona fide responders to PD-1/PD-L1 blockade in draining lymph nodes. Cell 185, 4049â4066 (2022).
Dammeijer, F. et al. The PD-1/PD-L1-checkpoint restrains T cell immunity in tumor-draining lymph nodes. Cancer Cell 38, 685â700 (2020).
Schenkel, J. M. et al. Conventional type I dendritic cells maintain a reservoir of proliferative tumor-antigen specific TCF-1+ CD8+ T cells in tumor-draining lymph nodes. Immunity 54, 2338â2353 (2021).
Sade-Feldman, M. et al. Defining T cell states associated with response to checkpoint immunotherapy in melanoma. Cell 176, 404 (2019).
Gago da Graca, C. et al. Stem-like memory and precursors of exhausted T cells share a common progenitor defined by ID3 expression. Sci. Immunol. 10, eadn1945 (2025).
Cai, S., Lee, C. C. & Kohwi-Shigematsu, T. SATB1 packages densely looped, transcriptionally active chromatin for coordinated expression of cytokine genes. Nat. Genet. 38, 1278â1288 (2006).
Ahlfors, H. et al. SATB1 dictates expression of multiple genes including IL-5 involved in human T helper cell differentiation. Blood 116, 1443â1453 (2010).
Wang, B., Ji, L. & Bian, Q. SATB1 regulates 3D genome architecture in T cells by constraining chromatin interactions surrounding CTCF-binding sites. Cell Rep. 42, 112323 (2023).
Russ, B. E. et al. Active maintenance of CD8+ T cell naivety through regulation of global genome architecture. Cell Rep. 42, 113301 (2023).
Leveen, P. et al. Induced disruption of the transforming growth factor β type II receptor gene in mice causes a lethal inflammatory disorder that is transplantable. Blood 100, 560â568 (2002).
Utzschneider, D. T. et al. T cells maintain an exhausted phenotype after antigen withdrawal and population reexpansion. Nat. Immunol. 14, 603â610 (2013).
Battegay, M. et al. Quantification of lymphocytic choriomeningitis virus with an immunological focus assay in 24- or 96-well plates. J. Virol. Methods 33, 191â198 (1991).
Giuffrida, L. et al. CRISPR/Cas9 mediated deletion of the adenosine A2A receptor enhances CAR T cell efficacy. Nat. Commun. 12, 3236 (2021).
Dobin, A. et al. STAR: ultrafast universal RNA-seq aligner. Bioinformatics 29, 15â21 (2013).
Ritchie, M. E. et al. limma powers differential expression analyses for RNA-sequencing and microarray studies. Nucleic Acids Res. 43, e47 (2015).
Yu, G., Wang, L. G., Han, Y. & He, Q. Y. clusterProfiler: an R package for comparing biological themes among gene clusters. OMICS 16, 284â287 (2012).
Harris, M. A. et al. The Gene Ontology (GO) database and informatics resource. Nucleic Acids Res. 32, D258âD261 (2004).
Kanehisa, M. & Goto, S. KEGG: Kyoto Encyclopedia of Genes and Genomes. Nucleic Acids Res. 28, 27â30 (2000).
Subramanian, A. et al. Gene set enrichment analysis: a knowledge-based approach for interpreting genome-wide expression profiles. Proc. Natl Acad. Sci. USA 102, 15545â15550 (2005).
Mootha, V. K. et al. PGC-1α-responsive genes involved in oxidative phosphorylation are coordinately downregulated in human diabetes. Nat. Genet. 34, 267â273 (2003).
Grandi, F. C., Modi, H., Kampman, L. & Corces, M. R. Chromatin accessibility profiling by ATACâseq. Nat. Protoc. 17, 1518â1552 (2022).
Martin, M. Cutadapt removes adapter sequences from high-throughput sequencing reads. EMBnet. J. 17, 10â12 (2011).
Danecek, P. et al. Twelve years of SAMtools and BCFtools. Gigascience 10, giab008 (2021).
Ramirez, F. et al. deepTools2: a next generation web server for deep-sequencing data analysis. Nucleic Acids Res. 44, W160âW165 (2016).
Zhang, Y. et al. Model-based analysis of ChIPâseq (MACS). Genome Biol. 9, R137 (2008).
Liao, Y., Smyth, G. K. & Shi, W. featureCounts: an efficient general purpose program for assigning sequence reads to genomic features. Bioinformatics 30, 923â930 (2014).
Ewels, P. A. et al. The nf-core framework for community-curated bioinformatics pipelines. Nat. Biotechnol. 38, 276â278 (2020).
Patel, H. et al. nf-core/atacseq: nf-core/atacseq v1.2.1 - Iron Centipede (1.2.1). Zenodo https://doi.org/10.5281/zenodo.2634132 (2020).
Heinz, S. et al. Simple combinations of lineage-determining transcription factors prime cis-regulatory elements required for macrophage and B cell identities. Mol. Cell 38, 576â589 (2010).
Carlson, M. org.Mm.eg.db: genome wide annotation for mouse. Bioconductor https://bioconductor.org/packages/release/data/annotation/html/org.Mm.eg.db.html (2000).
Acknowledgements
This study was supported by a National Health and Medical Research Council (NHMRC) Investigator Grant (2017420 to A.K.), NHMRC Ideas and Project Grants (1147409 to A.K., 2028921 to L.R. and C.T., 1185346 to A.K. and 1003131 to S.J.T.), an Australian Research Council grant (DE240100827 to C.T.), a Cancer Council grant (to A.K.), the Melbourne Research Scholarship (L.H., S.K.M.W., S.L., M.H.S., C.G.G. and N.T.H.-A.), the Deutsche Forschungsgemeinschaft (DFG) via a Postdoctoral Fellowship (L.R.), IRTG2168 272482170 (Z.A., H.H. and M.D.B.), DFG EXC2151 (390873048 to Z.A. and M.D.B.), CRC237/B15/B17 (369799452 to Z.A.), CRC1454 (432325352) (to Z.A. and M.D.B) and a CSL Centenary Fellowship (D.T.U.). Furthermore, M.D.B. is supported by the EU-funded project NEUROCOV, the RIA HORIZON Research and Innovation under GA 101057775 and the ElseâKrönerâFresenius Foundation (2018_A158). We thank the NIH Tetramer Facility for providing us with tetramers. We thank the Melbourne Cytometry Platform for providing flow cytometry services and the Doherty Institute Biological Resource Facility for animal husbandry and management.
Author information
Authors and Affiliations
Contributions
L.H. and A.K. conceived the study, designed the experiments, interpreted the results and wrote the paper. L.H. performed the majority of the experiments with help from M.H.S., S.K.M.W., S.L., N.T.H.-A., C.G.G. and H.H. L.R. performed and analyzed the tumor experiments. Z.A., S.J.T., M.D.B. and D.T.U. contributed to conceptualization and supervision of the study. L.D., A.F., J.S. and M.D.B. performed bioinformatic analyses. M.L.M. and L.H. performed the bulk RNA-seq and ATAC-seq experiments. C.T. performed the scRNA-seq experiment. M.D.B., M.K., D.S. and F.T.W. created the Satb1 transgenic and flexed mouse models. C.M.S., M.J.E., J.T., O.H. and P.K.D. performed the CAR T cell experiments. S.L.P., B.v.S. and L.K.M. created the B16-GP33 tumor cell line. S.J.T. provided key reagents.
Corresponding author
Ethics declarations
Competing interests
A.K. receives research support from Pfizer. The other authors declare no competing interests.
Peer review
Peer review information
Nature Immunology thanks Hazem Ghoneim, Charalampos Spilianakis and Tuoqi Wu for their contribution to the peer review of this work. Peer reviewer reports are available. Primary Handling Editor: Laurie A. Dempsey, in collaboration with the Nature Immunology team.
Additional information
Publisherâs note Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional affiliations.
Extended data
Extended Data Fig. 1 SATB1 limits TEX cell differentiation.
(a) Histogram and quantification showing the expression of SATB1 in polyclonal PD-1+ TPEX and TEX cells compared to naïve CD8 T cells and B cells (nâ=â10). (b-e) Naïve SATB1-deficient mice (Satb1flex/flex/Cd8Cre, SATB1-KO) or control mice (Satb1flex/flex) were analyzed using flow cytometry. (b) Flow cytometry plots displaying GFP expression upon Cre-mediated Satb1 gene flip in CD8+ T cells of KO mice compared to control mice (nâ=â5). (c) Quantification of SATB1 expression in CD4+ and CD8+ T cells (nâ=â5). (d) Numbers of splenic CD4+ and CD8+ T cells in SATB1-KO and control mice (nâ=â5). (e) Flow cytometry plots and quantification showing CD44 vs CD62L expression in SATB1-KO and control mice and quantification of naïve and memory phenotype CD8+ T cells (nâ=â5). (f-i) Naïve CD45.2+ SATB1-KO P14 cells or control cells P14 cells were adoptively transferred into naïve wildtype CD45.1+ mice and subsequently infected with LCMV-Docile. On day 12, 21, and 28 p.i. spleens were analyzed using flow cytometry. (f) Flow cytometry plots showing frequencies and numbers of SATB1-KO or control TPEX and TEX cells. (g) Flow cytometry plots and quantification on days 12 (upper) (nâ=â8) and 21 (lower) (nâ=â14) p.i. showing frequencies and numbers of SATB1-KO or control CD62L+ TPEX cells. (h) Flow cytometry plots and quantification on day 28 p.i showing the frequencies and numbers of SATB1-KO or control P14 cells (nâ=â5). (i) Flow cytometry plots and quantification on day 28 p.i showing the numbers of SATB1-KO and control TPEX, total TEX, CX3CR1+ TEX and CD101+ TEX cells (nâ=â5). Flow cytometry plots are representative. Dots in graphs represent individual mice; horizontal lines and error bars of bar graphs indicate meansâ±âSEM, respectively. Data are pooled (a,f,g) or representative (b-e, h,i) from two (day 12, day 28 p.i.) or three (day 21 p.i.) independent experiments. P values are from two-tailed unpaired studentâs t test.
Extended Data Fig. 2 SATB1 promotes functionality of exhausted T cells.
(a-h) Naïve CD45.2+ SATB1-deficient P14 cells (Satb1flex/flex/Cd8Cre, SATB1-KO) or control cells (Satb1flex/flex) P14 cells were adoptively transferred into naïve wildtype CD45.1+ mice and subsequently infected with LCMV-Docile. On days 12 and 21 p.i. spleens were analyzed using flow cytometry. (a) Flow cytometry plots and quantification of SATB1-KO or control P14 cells producing IFN-γ and TNF and quantification of IFN-γ and TNF expression after incubation with Gp33 peptide ex vivo on day 12 p.i. (nâ=â5). (b,c) Flow cytometry plots and quantification of SATB1-KO or control TPEX cells (b) and TEX cells. (c) producing IFN-γ or co-producing IFN-γ and TNF after incubation with Gp33 peptide ex vivo on days 12 (nâ=â5) and 21 (nâ=â4) p.i. (d,e) Histogram and quantification showing expression of GzmB (d) and PD-1 (e) in SATB1-KO or control TEX cells on day 12 p.i (nâ=â8). (f) Expression of PD-1 on SATB1-KO and control P14 cells after 66âh of in vitro stimulation (nâ=â10). (g) Expression of TOX in SATB1-KO or control P14 cells from day 12 of LCMV-Docile infection (nâ=â8). (h) Viral load in the lungs and kidneys of SATB1-KO or control mice infected with LCMV-Docile on day 31 p.i. (nâ=â3). Flow cytometry plots are representative. Dots in graphs represent biological replicates; horizontal lines and error bars of bar graphs indicate meansâ±âSEM (a-g) or SD (h), respectively. Data are representative (a-c, h) or pooled from two (d-g) independent experiments. P values are from two-tailed unpaired studentâs t test. PFU, Plaque forming units.
Extended Data Fig. 3 SATB1 downregulation is required for TEX cell differentiation.
(a) Schematic illustrating the generation of Rosa26Satb1 locus allele. A lox-STOP-lox cassette followed by an Satb1-IRES-GFP sequence was inserted downstream of the CAG promoter region on the Rosa26 gene locus. Cre-mediates deletion of the STOP-codon leads to continuously expression of Satb1-GFP. (b-g) Mixed bone marrow chimeric mice containing Satb1Tg (CD45.2+) and control T cells (CD45.1/2+) were infected with LCMV-Docile. Spleens were harvested on day 12 and 21 p.i. and analyzed using flow cytometry. (b) Flow cytometry plots and quantification on day 12 p.i. showing frequencies of activated CD44+ and exhausted PD-1+ Satb1Tg or control cells (nâ=â14). (c) Quantification on day 12 p.i. of CD44 (nâ=â9) and PD-1 (nâ=â14) expression among PD-1+ Satb1Tg or control cells. (d) Normalized numbers of Satb1Tg or control TPEX and TEX cells on day 21 p.i (nâ=â9). (e) Flow cytometry plots and quantification of Satb1Tg or control TPEX and TEX cells from Day 12 p.i (nâ=â14). (f,g) Expression of TCF1 (nâ=â10) and Ly108 (nâ=â15) (f) and PD-1 (nâ=â14), TOX (nâ=â9), and CD44 (nâ=â13) (g) in Satb1Tg or control TPEX cells from day 21 p.i. (h) Flow cytometry plots and quantification on days 12 (nâ=â9) and 21 (nâ=â14) p.i. showing frequencies of Satb1Tg and control CD62L+ TPEX cells. Flow cytometry plots are representative. Dots in graphs represent individual mice; horizontal lines and error bars of bar graphs indicate meansâ±âSEM, respectively. Data are pooled from two (c) or three (b,d-g) independent experiments. P values are from two-tailed paired studentâs t test.
Extended Data Fig. 4 SATB1 expression is required for efficient cytokine production.
Mixed bone marrow chimeric mice containing Satb1Tg (CD45.2+) and control T cells (CD45.1/2+) were infected with LCMV-Docile. On days 12 and 21 p.i. spleens were analyzed using flow cytometry. (a) Quantification of PD-1 expression on day 12 p.i. (nâ=â9) and 21 p.i. (nâ=â8), and of TOX (nâ=â4) expression on day 21 in Gp33+ Satb1Tg or control cells. (b) Flow cytometry plots and quantification on day 21 p.i. showing frequencies of Satb1Tg or control TPEX and TEX cells among Gp33+ cells (nâ=â9). (c-e) Flow cytometry plots and quantification of IFN-γ (nâ=â13) and TNF (nâ=â11) production from TPEX (c) and TEX (nâ=â13) (d,e) cells after incubation of Satb1Tg or control CD8 T cells with Gp33 peptide on day 21 p.i. (f) Flow cytometry plots and quantification of IFN-γ and TNF production from Satb1Tg or control PD-1+ CD8 T cells after incubation with PMA/Ionomycin (nâ=â8). (g-l) CTV-labelled SATB1-KO and control P14 cells were adoptively transferred to recipient mice prior to infection with chronic LCMV-Clone13 before analysis on day 3 p.i. (g) Outline of experimental scheme. (h) Flow cytometry plot of control cells and quantification of SATB1 expression per cell division (nâ=â5). (i) Histogram and quantification of frequencies of TCF1+ SATB1-KO and control P14 cells per cell division (nâ=â5). (j) Histogram and quantification of frequencies of CD62L+ SATB1-KO and control P14 cells per cell division (nâ=â5). (k) Quantification of PD-1 expression on SATB1-KO and control P14 cells per cell division (nâ=â5). (l) Flow cytometry plots and quantification of KLRG1 expression on SATB1-KO and control P14 cells in divisions 6 and >7 (nâ=â5). Flow cytometry plots are representative. Data are representative (h-l) or pooled (a-c) from two or three (d-f) independent experiments. Dots in graphs represent individual mice; horizontal lines and error bars represent indicate meansâ±âSEM, respectively. P values in (a-f) are from two-tailed paired studentâs t test.
Extended Data Fig. 5 SATB1 expression is repressed by TGF-β and limits differentiation of short-lived effect CD8+ T cells.
(a) Histogram and quantification of SATB1 expression in wild-type CD8+ T cells that were stimulated in vitro in the presence or absence of titrated doses of TGF-β (nâ=â8). (b-d) Mixed bone marrow chimeric mice containing TGF-βR2 deficient and control CD8+ T cells were infected with LCMV-Docile. Spleens were analyzed on day 21 p.i. by flow cytometry. (b) Schematic of experimental set up. (c) SATB1 expression in TGF-βR2 deficient and control Gp33-specific CD8+ T cells (nâ=â10). (d) SATB1 expression in TGF-βR2 deficient and control Gp33-specific CD8+ T cell subsets (nâ=â10). (e,f) Naïve CD45.1+ P14 cells were adoptively transferred into naïve wildtype CD45.2+ mice and infected with LCMV-Armstrong. Spleens were harvested on days 7 or 28 p.i. and analyzed using flow cytometry. (e) Quantification showing the expression of SATB1 in MPC and SLEC cells compared to naïve CD8+ T cells and B cells (nâ=â10). (f) Histograms and quantification showing the expression of SATB1 in TCM and TEM cells compared to naïve CD8+ T cells and B cells (nâ=â9). (g-n) Naïve CD45.2+ SATB1-deficient P14 cells (Satb1flex/flex/Cd8Cre, SATB1-KO) or control cells (Satb1flex/flex) P14 cells were adoptively transferred into naïve wildtype CD45.1+ mice and subsequently infected with LCMV-Armstrong. Mice were analyzed on day 7 p.i. (g) Schematic of experimental set up. (h,i) Flow cytometry plots (h) and quantification (i) of SATB1-KO and control P14 frequencies and numbers per spleen (nâ=â10). (j) Flow cytometry plots and quantification showing TIM-3 and PD-1 expression in SATB1-KO and control splenic P14 cells (nâ=â10). (k) Quantification of SATB1-KO and control SLEC and MPC numbers per spleen (nâ=â10). (l) Flow cytometry plots of IFN-γ and TNF production from SATB1-KO or control SLEC and MPC cells after incubation with Gp33 peptide (nâ=â10). (m) Quantification of frequencies of IFN-γ and TNF producing SATB1-KO and control SLEC and MPC (nâ=â10). (n) Quantification of IFN-γ and TNF expression in SATB1-KO and control SLEC and MPC (nâ=â10). Flow cytometry plots are representative. Dots in graphs represent biological replicates; horizontal lines and error bars of graphs indicate means and SD respectively (a). Data are pooled from at least two experiments. Dots in graphs represent individual mice (c-n); horizontal lines and error bars of bar graphs indicate meansâ±âSEM, respectively. P values are from unpaired, two-way ANOVA (e,f), or two-tailed paired studentâs t-tests (c,d). P values are from unpaired studentâs t test (a,i-n).
Extended Data Fig. 6 SATB1 downregulation is required for proper effector differentiation in acute viral infection.
(a-c) Naïve CD45.1+ P14 cells were adoptively transferred into naïve wildtype CD45.2+ mice and infected with LCMV-Armstrong. Spleens were harvested on days 7 or 28 p.i. and analyzed using flow cytometry. (a) Flow cytometry plots and quantification of SATB1-KO and control CX3CR1 and KLRG1 SLEC populations on day 7 p.i. (nâ=â10). (b) Histogram and quantification of GzmB producing SATB1-KO or control SLEC on day 7 p.i. (nâ=â10). (c) Quantification of SATB1-KO (nâ=â9) and control (nâ=â10) TCM and TEM cell numbers per spleen on day 28 p.i. (d-g) Mixed bone marrow chimeric mice containing Satb1Tg (CD45.2+) and control T cells (CD45.1/2+) were infected with LCMV-Armstrong. On day 7 p.i. spleens were analyzed using flow cytometry. (d) Flow cytometry plots and quantification of activated CD44+ Satb1Tg and control cells (nâ=â10). (e) Quantification of PD-1 expression in PD-1+ and Gp33+ Satb1Tg and control cells (nâ=â10). (f) Flow cytometry plots and quantification of CX3CR1 and KLRG1 Gp33+ Satb1Tg and control populations (nâ=â10). (g) Quantification of cytokine expression Satb1Tg and control cells after Gp33 peptide restimulation (nâ=â10). Flow cytometry plots are representative. Dots in graphs represent individual mice; horizontal lines and error bars of bar graphs indicate meansâ±âSEM, respectively. Data are pooled from two independent experiments. P values are from two-tailed paired studentâs t test.
Extended Data Fig. 7 Loss of SATB1 does not impact exhausted T cell subset specific chromatin accessibility.
Naïve CD45.2+ SATB1-deficient P14 cells (Satb1flex/flex/Cd8Cre, SATB1-KO) or control cells (Satb1flex/flex) P14 cells were adoptively transferred into naïve wildtype CD45.1+ mice which were subsequently infected with LCMV-Docile. On 21 p.i., CD62L+ and CD62Lâ TPEX and CX3CR1+ and CD101+ TEX cells were sorted from either SATB1-KO or control spleens and used for RNA and ATAC-sequencing. (a) Comparison of gene accessibility in control CD62L+ TPEX, CD62Lâ TPEX, CX3CR1+ TEX and CD101+ TEX cells. TCF7, Irf4, Slamf1 and Tgfbr2 are shown as examples. (b) Representation of the first two components of a PCA dimensionality reduction based on rlog-transformed counts for all accessible peaks. First component explains 56% and second component explains 19% of total variability. Cell type and genotypes are color coded. (c) K-means clustered heatmap showing normalized counts of all peaks identified using the nextflow pipeline nf-core/atacseq v1.2.1. TPEX and TEX cell subsets, genotype and replicates are color coded and shown as bars at the top of the heatmap. (d) Proportion of open chromatin peaks identified by ATAC-seq that overlap with SATB1 binding sites as identified by ChIP-seq. (e) Volcano plots showing genes associated with differentially accessible regions (DARs). DARs were identified using the nextflow pipeline nf-core/atacseq v1.2.1, which passed the False Discover Rate (FDR)â<â0.05. Genes closest to the respective DAR were identified using Homer v4.11. Each volcano plot shows a SATB1-KO vs Ctrl comparison for each TPEX and TEX cell subset. Dashed lines: reciprocal adjusted p-valueâ<â0.01 (padjâ1 vertical) and log2 fold change (log2FC)â>â1.5 (horizontal). Statistical significance is color coded by passing log2FC and padjâ1 cut-offs (red), passing only log2FC- (green) or padjâ1 cut-off (blue). Top genes were labelled with the gene name. (f) Volcano plot showing accessible regions in the Ifng, Tnf and Gzmb gene loci. The Gzmb transcriptional start site is highlighted in red. (g) GOEA analysis of genes associated with differentially accessible regions. DARs were used for Gene Ontology Enrichment Analysis (GOEA) using the Hallmark gene sets from the GSEA-MSigDB database as reference. Ontology terms were filtered for an adjusted p-valueâ<â0.05. Each Hallmark term is indicated as the main bubble surrounded by genes belonging to the term. Number of genes belonging to a term is indicated by bubble size. Comparison to identify DARs is indicated by color. Shared terms between comparisons are colored by multiple colors.
Extended Data Fig. 8 SATB1 binds to genomic regions that show differential accessibility during exhausted T cell differentiation.
(a-c) Naïve CD45.2+ SATB1-deficient P14 cells (Satb1flex/flex/Cd8Cre, SATB1-KO) or control cells (Satb1flex/flex) P14 cells were adoptively transferred into naïve wildtype CD45.1+ mice which were subsequently infected with LCMV-Docile. On 21 p.i., CD62L+ and CD62Lâ TPEX and CX3CR1+ and CD101+ TEX cells were sorted from either SATB1-KO or control spleens and used for RNA and ATAC-sequencing. Shown are SATB1 ChIP binding and gene accessibility in indicated cell types. (a) Ifng, (b) Slamf1, Slamf6, and (c) Arid5a, Ccr5 and Prdm1. (d-i) Mixed bone marrow chimeric mice containing SATB1-DNA-binding mutant (Satb1m1Anu/m1Anu) (CD45.2+) and control T cells (CD45.1/2+) were infected with LCMV-Docile. On day 21 p.i. spleens were analyzed using flow cytometry. (d) Flow cytometry plots and quantification showing frequencies of activated Gp33+ Satb1m1Anu/m1Anu or control cells. (e) Flow cytometry plots and quantification showing frequencies of Satb1m1Anu/m1Anu or control TPEX and TEX cells. (f) Expression of TCF1 and Ly108 in Gp33+ Satb1m1Anu/m1Anu or control TPEX cells. (g) Flow cytometry plots and quantification showing frequencies of Satb1m1Anu/m1Anu or control CD62L+ TPEX cells. (h) Flow cytometry plots and quantification showing frequencies of Satb1m1Anu/m1Anu or control CX3CR1+ and CD101+ TEX cells. (i) Flow cytometry plots and quantification showing frequencies of IFN-γ and TNF producing Satb1m1Anu/m1Anu or control cells after stimulation with Gp33 peptide ex vivo. Flow cytometry plots are representative. Dots in graphs represent individual mice; horizontal lines and error bars of bar graphs indicate meansâ±âSEM, respectively. Data are pooled from two independent experiments. P values are from two-tailed unpaired studentâs t test.
Extended Data Fig. 9 SATB1 limits tumor control and response to checkpoint inhibition.
Naïve CD45.2+ SATB1-deficient P14 cells (Satb1flex/flex/Cd8Cre, SATB1-KO) or control cells (Satb1flex/flex) P14 cells were adoptively transferred into naïve wildtype CD45.1+ mice 3 days after inoculation with B16/Gp33+ Tumor cells. Mice were treated with immune checkpoint blockade (ICB, anti-PDL-1 + anti-CTLA4) or PBS. (a,b) Tumor growth (a) and survival (b) of mice transferred with either SATB1-KO or control P14 cells treated with or without ICB (nâ=â8). Data are pooled from two independent experiments. (c,d) Representative flow cytometry plots and quantification showing the frequencies of IFN-γ and TNF producing SATB1-KO and Control P14 cells in the tumor (c) and tdLNs (d) treated with or without ICB after ex vivo restimulation with Gp33 peptide (untreated nâ=â8, treated KO nâ=â5, treated WT nâ=â7). Dots in graphs represent individual mice; horizontal lines and error bars of bar graphs indicate meansâ±âSEM, respectively. Data are pooled from two independent experiments. P values are from two-way ANOVA (b) or unpaired studentâs t test (c,d).
Extended Data Fig. 10 SATB1 limits effector differentiation of CD8+ T cells in tumor draining lymph nodes.
Naïve CD45.2+ SATB1-deficient P14 cells (Satb1flex/flex/Cd8Cre, SATB1-KO) or control cells (Satb1flex/flex) P14 cells were adoptively transferred into naïve wildtype CD45.1+ mice 3 days after inoculation with B16/Gp33 tumor cells. Mice were treated with immune checkpoint blockade (ICB, anti-PDL-1 + anti-CTLA4) or PBS on indicated days. Tumor draining LN (tdLNs) were harvested on day 16 after inoculation and analyzed using flow cytometry. (a) Representative flow cytometry plots and quantification showing frequencies and numbers of SATB1-KO or control P14 cells (untreated nâ=â8, treated KO nâ=â6, treated WT nâ=â7). (b) Representative flow cytometry plots and quantification showing frequencies and numbers SATB1-KO or control CX3CR1+ TEX cells (untreated nâ=â8, treated KO nâ=â6, treated WT nâ=â7). (c) Representative flow cytometry plots and quantification showing frequencies and numbers of GzmB producing SATB1-KO or control P14 cells (untreated nâ=â8, treated KO nâ=â6, treated WT nâ=â7). Dots in graphs represent individual mice; horizontal lines and error bars of bar graphs indicate meansâ±âSEM, respectively. Data are representative of two independent experiments. P values are from unpaired studentâs t test.
Supplementary information
Supplementary Information
Supplementary Figs. 1â3.
Supplementary Table 1
ATAC DARs, cell-type specific, FDR_0_05_LFC_1_5.
Supplementary Table 2
ATAC DARs, genotype specific, KOvsWT_FDR_0_1.
Supplementary Table 3
Venn diagram related to Fig. 6f.
Supplementary Table 4
Differentially expressed genes between wild-type and Satb1 KO.
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
Heyden, L., Rausch, L., Shannon, M.H. et al. Transcriptional regulator SATB1 limits CD8+ T cell population expansion and effector differentiation in chronic infection and cancer. Nat Immunol (2025). https://doi.org/10.1038/s41590-025-02316-2
Received:
Accepted:
Published:
Version of record:
DOI: https://doi.org/10.1038/s41590-025-02316-2


