Dr. Christian Schmidl
Head of Research Group | Epigenetic Immuno-oncology
The Epigenetic Immuno-oncology Research Group has a particular focus on the non-coding genome. We investigate this using high-throughput (single-cell) sequencing approaches to study immune cells and their interactions with friend and foe. We also collaborate closely with other clinical partners to address the topics of cancer and inflammation. Our goal is to translate our findings for the patient’s well-being.
Specifically, we focus on CD8+ T cells that can detect and destroy malignant cells. In this context, we observe that their effectiveness can sometimes be compromised in the tumour itself and that they can become dysfunctional as a result. On the cellular level, chronic antigen exposure and the suppressive microenvironment drive the process of dysfunction, which is accompanied by a remodeling of the T cells’ chromatin landscapes. Our Research Group dissects the heterogeneous chromatin and transcriptional patterns of human tumor-infiltrating T cells to obtain deeper insights into the underlying molecular processes that shape T-cell function and dysfunction. Based on this data, we seek to learn how specific signals sent during the immune response direct gene expression in T cells.
Part of our remit is to explore the potential to delete T cell-exhaustion driver genes in immunotherapeutic products such as chimeric antigen receptor (CAR) T cells, so as to improve their function. With our technical focus on CRISPR gene manipulation and epigenome profiling, our work extends to research fields such as cancer metastasis and stem-cell transplantation in leukemia patients.
Quote from Dr. Christian Schmidl
My team and I are fascinated by the molecular mechanisms of gene regulation.
Head of Research Group Epigenetic Immuno-oncology
Biography
Academic background and qualifications
Dr. Christian Schmidl studied Biology and obtained his PhD in 2013 at the University of Regensburg under the supervision of Michael Rehli. Here, he studied DNA methylation patterns in lymphocytes. During this time, Dr. Schmidl was also involved in the international Functional Annotation of the Human Genome (FANTOM) project, where he got increasingly interested in how different cells in the human body regulate expression of their genes. This led him to study cell type-specific gene expression in regulatory and conventional T cells.
Professional career
Dr. Schmidl joined CeMM, Vienna Austria, as an Alexander von Humboldt postdoctoral fellow. He worked in Christoph Bock’s lab, where he applied his knowledge to better understand molecular regulation of malignant immune cells with a focus on chronic lymphocytic leukaemia. For this purpose, Dr. Schmidl developed new chromatin-mapping methods (ChIPmentation) that allow epigenome profiling from scarce patient material. In 2017, Dr. Schmidl established his own research group at the Leibniz Institute for Immunotherapy, where he is studying gene-regulatory mechanisms in T cells fighting cancer.
Honors
In 2013 Dr. Schmidl received a PhD thesis award from the Faculty of Biology and Preclinical Medicine from the University of Regensburg, Germany. From 2014 until 2017 he was awarded the Feodor Lynen Postdoc scholarship by the Alexander von Humboldt Foundation. In 2023 he received the Regensburg Oncology Award.
Explore our Research Group in greater depth
Get to know our team and find out more about our pioneering research.
Visit the complete publications list on Google Scholar:
https://scholar.google.de/citations?user=qFeznaMAAAAJ&hl=de
Here is a selection of the most important publications from the last few years:
- Riegel D*, Romero-Fernandez E*, Simon M*, Adenugba AR, Singer K, Mayr R, Weber F, Kleemann M, Imbusch CD, Kreutz M, Brors B, Ugele I, Werner JM, Siska PJ, Schmidl C. Integrated single-cell profiling dissects cell-state-specific enhancer landscapes of human tumor-infiltrating CD8(+) T cells. Mol Cell. 2023 Feb 16. 83:622-636 e610. doi:10.1016/j.molcel.2022.12.029. PMID: 36657444
- Babl N, Decking SM, Voll F, Althammer M, Sala-Hojman A, Ferretti R, Korf C, Schmidl C, Schmidleithner L, Nerb B, Matos C, Koehl GE, Siska P, Bruss C, Kellermeier F, Dettmer K, Oefner PJ, Wichland M, Ugele I, Bohr C, Herr W, Ramaswamy S, Heinrich T, Herhaus C, Kreutz M et al. MCT4 blockade increases the efficacy of immune checkpoint blockade. J Immunother Cancer. 2023 Oct. 11doi:10.1136/jitc-2023-007349. PMID: 37880183
- Rendeiro AF, Krausgruber T, Fortelny N, Zhao F, Penz T, Farlik M, Schuster LC, Nemc A, Tasnady S, Reti M, Matrai Z, Alpar D#, Bodor C#, Schmidl C#, Bock C#. Chromatin mapping and single-cell immune profiling define the temporal dynamics of ibrutinib response in CLL. Nat Commun. 2020 Jan 29. 11:577. doi:10.1038/s41467-019-14081-6. PMID: 31996669
- Feldker N, Ferrazzi F, Schuhwerk H, Widholz SA, Guenther K, Frisch I, Jakob K, Kleemann J, Riegel D, Bonisch U, Lukassen S, Eccles RL, Schmidl C, Stemmler MP, Brabletz T, Brabletz S. Genome-wide cooperation of EMT transcription factor ZEB1 with YAP and AP-1 in breast cancer. EMBO J. 2020 Sep 1. 39:e103209. doi:10.15252/embj.2019103209. PMID: 32692442
- Delacher M, Imbusch CD, Hotz-Wagenblatt A, Mallm JP, Bauer K, Simon M, Riegel D, Rendeiro AF, Bittner S, Sanderink L, Pant A, Schmidleithner L, Braband KL, Echtenachter B, Fischer A, Giunchiglia V, Hoffmann P, Edinger M, Bock C, Rehli M, Brors B, Schmidl C#, Feuerer M#. Precursors for Nonlymphoid-Tissue Treg Cells Reside in Secondary Lymphoid Organs and Are Programmed by the Transcription Factor BATF. Immunity. 2020 Feb 18. 52:295-312 e211. doi:10.1016/j.immuni.2019.12.002. PMID: 31924477
- Schmidl C*, Vladimer GI*, Rendeiro AF*, Schnabl S*, Krausgruber T, Taubert C, Krall N, Pemovska T, Araghi M, Snijder B, Hubmann R, Ringler A, Runggatscher K, Demirtas D, de la Fuente OL, Hilgarth M, Skrabs C, Porpaczy E, Gruber M, Hoermann G, Kubicek S, Staber PB, Shehata M, Superti-Furga G, Jager U et al. Combined chemosensitivity and chromatin profiling prioritizes drug combinations in CLL. Nat Chem Biol. 2019 Mar. 15:232-240. doi:10.1038/s41589-018-0205-2. PMID: 30692684
- Datlinger P, Rendeiro AF, Schmidl C, Krausgruber T, Traxler P, Klughammer J, Schuster LC, Kuchler A, Alpar D, Bock C. Pooled CRISPR screening with single-cell transcriptome readout. Nat Methods. 2017 Mar. 14:297-301. doi:10.1038/nmeth.4177. PMID: 28099430
- Rendeiro AF*, Schmidl C*, Strefford JC*, Walewska R, Davis Z, Farlik M, Oscier D, Bock C. Chromatin accessibility maps of chronic lymphocytic leukaemia identify subtype-specific epigenome signatures and transcription regulatory networks. Nat Commun. 2016 Jun 27. 7:11938. doi:10.1038/ncomms11938. PMID: 27346425
- Schmidl C*, Rendeiro AF*, Sheffield NC, Bock C. ChIPmentation: fast, robust, low-input ChIP-seq for histones and transcription factors. Nat Methods. 2015 Oct. 12:963-965. doi:10.1038/nmeth.3542. PMID: 26280331
- Schmidl C, Renner K, Peter K, Eder R, Lassmann T, Balwierz PJ, Itoh M, Nagao-Sato S, Kawaji H, Carninci P, Suzuki H, Hayashizaki Y, Andreesen R, Hume DA, Hoffmann P, Forrest AR, Kreutz MP, Edinger M, Rehli M, and the FANTOM consortium. Transcription and enhancer profiling in human monocyte subsets. Blood. 2014 Apr 24. 123:e90-99. doi:10.1182/blood-2013-02-484188. PMID: 24671955
- Andersson R, Gebhard C, Miguel-Escalada I, Hoof I, Bornholdt J, Boyd M, Chen Y, Zhao X, Schmidl C, Suzuki T, Ntini E, Arner E, Valen E, Li K, Schwarzfischer L, Glatz D, Raithel J, Lilje B, Rapin N, Bagger FO, Jorgensen M, Andersen PR, Bertin N, Rackham O, Burroughs AM et al. An atlas of active enhancers across human cell types and tissues. Nature. 2014 Mar 27. 507:455-461. doi:10.1038/nature12787. PMID: 24670763
- Schmidl C, Klug M, Boeld TJ, Andreesen R, Hoffmann P, Edinger M, Rehli M. Lineage-specific DNA methylation in T cells correlates with histone methylation and enhancer activity. Genome Res. 2009 Jul. 19:1165-1174. doi:10.1101/gr.091470.109. PMID: 19494038
Many thanks to the funding agencies who support our work:
2023–2025: German Cancer Aid (DKH)
Enhancement of CAR T-cell function with CRISPR activation and interference.
2022–2025: SFB TRR 221
Timed targeting of cGAS/STING to improve tissue regeneration and antitumor responses following allo-HSCT
2021–2024: SFB TRR 305
EMT-dependent transcriptional enhancers important for metastatic colonization
2017–2022: German Research Foundation (DFG)
Tumor microenvironment-directed epigenome remodeling of tumor-infiltrating T cells
2020: 10x Genomics
High-throughput dissection of gene-regulatory element manipulations in T cells using single-cell RNA sequencing
Here are some of the partners with whom the Research Group has recently collaborated:
cGAS/STING in allogeneic stem-cell transplantations
Allogeneic hematopoietic stem cell transplantations (allo-HSCT) represent an important strategy to treat patients with hematologic malignancies, as donor immune cells mediate a graft-versus-leukemia (GvL) effect that eliminates residual malignant cells and thus prevents relapse. However, this strategy comes with the disadvantage that alloreactive T cells can attack the patient’s tissues and lead to life-threatening graft-versus-host disease (GVHD).
Embedded in the SFB221, and in collaboration with Hendrik Poeck (Internal Medicine III, University Hospital Regensburg), we study how manipulation of the innate nucleic acid receptor pathways cGAS/STING can shape the outcome of allo-HSCT. Endogenous and external activation of cGAS/STING prior to tissue injury—induced by allo-HSCT—results in protective IFN-I signaling and reduces GVHD by strengthening gut epithelial barrier integrity, and potentially increasing GvL T cell activity by inducing a stem-like memory phenotype. In this project, we therefore investigate how a selective and timed cGAS/STING activation or inhibition during the course of allo-HSCT can improve anti-tumor responses following allo-HSCT, while minimizing the immune-mediated side-effects of these treatment approaches such as GVHD.
Gene regulation in cancer metastasis
Cancer is a leading cause of death worldwide. And yet it is not the primary tumor but the metastases that are responsible for 90% of cancer-related deaths. In collaboration with the Brabletz lab at the Friedrich-Alexander University of Erlangen-Nürnberg, and embedded in the SFB305, we investigate gene-regulatory mechanisms involved in the metastatic colonization process. Specifically, we focus on the Epithelial-Mesenchymal Transition (EMT) process that governs cellular changes when cancer cells leave the primary tumor and advance to form metastasis.
Of significance here, is the fact that EMT is activated by a core set of transcription factors such as members of Snail, Twist, and ZEB families. Among these EMT-TFs, ZEB1 has been widely studied because it is a crucial modulator of the metastatic process. This is because it triggers an invasive behaviour which correlates to a poor prognosis in different cancer histotypes including Pancreatic Ductal Adenocarcinoma (PDAC)—one of the most aggressive human cancers and the fourth leading cause of cancer-related deaths worldwide. We have recently shown that Zeb1 can act as a transcriptional enhancer and therefore our main focus is around dissecting how Zeb1 shapes the EMT process and the metastasis formation. By using chromatin profiling and CRISPR tools, we map and manipulate Zeb1-dependent non-coding gene regulatory element activity during EMT to better understand the molecular mechanisms that underlie the process.
Dr. Christian Schmidl
Tel: +49 941 944–18176
Email: christian.schmidl@ukr.de