Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 36
Filter
1.
Infect Genet Evol ; 119: 105577, 2024 Apr.
Article in English | MEDLINE | ID: mdl-38403035

ABSTRACT

In January 2021, the monitoring of circulating variants of SARS-CoV-2 was initiated in Germany under the Corona Surveillance Act, which was discontinued after July 2023. This initiative aimed to enhance pandemic containment, as specific amino acid changes, particularly in the spike protein, were associated with increased transmission and reduced vaccine efficacy. Our group conducted whole genome sequencing using the ARTIC protocol (currently V4) on Illumina's NextSeq 500 platform (and, starting in May 2023, on the MiSeq DX platform) for SARS-CoV-2 positive specimen from patients at Heidelberg University Hospital, associated hospitals, and the public health office in the Rhine-Neckar/Heidelberg region. In total, we sequenced 26,795 SARS-CoV-2-positive samples between January 2021 and July 2023. Valid sequences, meeting the requirements for upload to the German electronic sequencing data hub (DESH) operated by the Robert Koch Institute (RKI), were determined for 24,852 samples, and the lineage/clade could be identified for 25,912 samples. The year 2021 witnessed significant dynamics in the circulating variants in the Rhine-Neckar/Heidelberg region, including A.27.RN, followed by the emergence of B.1.1.7 (Alpha), subsequently displaced by B.1.617.2 (Delta), and the initial occurrences of B.1.1.529 (Omicron). By January 2022, B.1.1.529 had superseded B.1.617.2, dominating with over 90%. The years 2022 and 2023 were then characterized by the dominance of B.1.1.529 and its sublineages, particularly BA.5 and BA.2, and more recently, the emergence of recombinant variants like XBB.1.5. Since the global dominance of B.1.617.2, the identified variant distribution in our local study, apart from a time delay in the spread of new variants, can be considered largely representative of the global distribution. om a time delay in the spread of new variants, can be considered largely representative of the global distribution.


Subject(s)
COVID-19 , SARS-CoV-2 , Humans , SARS-CoV-2/genetics , COVID-19/epidemiology , Germany/epidemiology , Hospitals, University
2.
Nat Commun ; 15(1): 269, 2024 Jan 08.
Article in English | MEDLINE | ID: mdl-38191550

ABSTRACT

Medulloblastomas with extensive nodularity are cerebellar tumors characterized by two distinct compartments and variable disease progression. The mechanisms governing the balance between proliferation and differentiation in MBEN remain poorly understood. Here, we employ a multi-modal single cell transcriptome analysis to dissect this process. In the internodular compartment, we identify proliferating cerebellar granular neuronal precursor-like malignant cells, along with stromal, vascular, and immune cells. In contrast, the nodular compartment comprises postmitotic, neuronally differentiated malignant cells. Both compartments are connected through an intermediate cell stage resembling actively migrating CGNPs. Notably, we also discover astrocytic-like malignant cells, found in proximity to migrating and differentiated cells at the transition zone between the two compartments. Our study sheds light on the spatial tissue organization and its link to the developmental trajectory, resulting in a more benign tumor phenotype. This integrative approach holds promise to explore intercompartmental interactions in other cancers with varying histology.


Subject(s)
Cerebellar Neoplasms , Medulloblastoma , Humans , Medulloblastoma/genetics , Cell Differentiation , Cerebellar Neoplasms/genetics , Disease Progression , Histological Techniques
3.
Nat Commun ; 14(1): 5011, 2023 08 17.
Article in English | MEDLINE | ID: mdl-37591845

ABSTRACT

In multiple myeloma spatial differences in the subclonal architecture, molecular signatures and composition of the microenvironment remain poorly characterized. To address this shortcoming, we perform multi-region sequencing on paired random bone marrow and focal lesion samples from 17 newly diagnosed patients. Using single-cell RNA- and ATAC-seq we find a median of 6 tumor subclones per patient and unique subclones in focal lesions. Genetically identical subclones display different levels of spatial transcriptional plasticity, including nearly identical profiles and pronounced heterogeneity at different sites, which can include differential expression of immunotherapy targets, such as CD20 and CD38. Macrophages are significantly depleted in the microenvironment of focal lesions. We observe proportional changes in the T-cell repertoire but no site-specific expansion of T-cell clones in intramedullary lesions. In conclusion, our results demonstrate the relevance of considering spatial heterogeneity in multiple myeloma with potential implications for models of cell-cell interactions and disease progression.


Subject(s)
Multiple Myeloma , Humans , Multiple Myeloma/genetics , Cell Communication , Chromatin Immunoprecipitation Sequencing , Clone Cells , Disease Progression , Tumor Microenvironment/genetics
4.
Blood ; 142(19): 1633-1646, 2023 11 09.
Article in English | MEDLINE | ID: mdl-37390336

ABSTRACT

Intratumor heterogeneity as a clinical challenge becomes most evident after several treatment lines, when multidrug-resistant subclones accumulate. To address this challenge, the characterization of resistance mechanisms at the subclonal level is key to identify common vulnerabilities. In this study, we integrate whole-genome sequencing, single-cell (sc) transcriptomics (scRNA sequencing), and chromatin accessibility (scATAC sequencing) together with mitochondrial DNA mutations to define subclonal architecture and evolution for longitudinal samples from 15 patients with relapsed or refractory multiple myeloma. We assess transcriptomic and epigenomic changes to resolve the multifactorial nature of therapy resistance and relate it to the parallel occurrence of different mechanisms: (1) preexisting epigenetic profiles of subclones associated with survival advantages, (2) converging phenotypic adaptation of genetically distinct subclones, and (3) subclone-specific interactions of myeloma and bone marrow microenvironment cells. Our study showcases how an integrative multiomics analysis can be applied to track and characterize distinct multidrug-resistant subclones over time for the identification of molecular targets against them.


Subject(s)
Multiple Myeloma , Humans , Multiple Myeloma/drug therapy , Multiple Myeloma/genetics , Multiomics , Mutation , Transcriptome , Tumor Microenvironment/genetics
5.
Nat Commun ; 12(1): 6960, 2021 11 29.
Article in English | MEDLINE | ID: mdl-34845188

ABSTRACT

Virtually all patients with multiple myeloma become unresponsive to treatment over time. Relapsed/refractory multiple myeloma (RRMM) is accompanied by the clonal evolution of myeloma cells with heterogeneous genomic aberrations and profound changes of the bone marrow microenvironment (BME). However, the molecular mechanisms that drive drug resistance remain elusive. Here, we analyze the heterogeneous tumor cell population and its complex interaction network with the BME of 20 RRMM patients by single cell RNA-sequencing before/after treatment. Subclones with chromosome 1q-gain express a specific transcriptomic signature and frequently expand during treatment. Furthermore, RRMM cells shape an immune suppressive BME by upregulation of inflammatory cytokines and close interaction with the myeloid compartment. It is characterized by the accumulation of PD1+ γδ T-cells and tumor-associated macrophages as well as the depletion of hematopoietic progenitors. Thus, our study resolves transcriptional features of subclones in RRMM and mechanisms of microenvironmental reprogramming with implications for clinical decision-making.


Subject(s)
Drug Resistance, Neoplasm/genetics , Gene Expression Regulation, Neoplastic , Multiple Myeloma/genetics , Transcriptome , Tumor Microenvironment/genetics , Antineoplastic Agents/therapeutic use , Bone Marrow/drug effects , Bone Marrow/immunology , Bone Marrow/pathology , Cytokines/genetics , Cytokines/immunology , Drug Resistance, Neoplasm/immunology , Gene Expression Profiling , Gene Regulatory Networks , Hematopoietic Stem Cells/immunology , Hematopoietic Stem Cells/pathology , Humans , Intraepithelial Lymphocytes/immunology , Intraepithelial Lymphocytes/pathology , Multiple Myeloma/drug therapy , Multiple Myeloma/immunology , Multiple Myeloma/pathology , Programmed Cell Death 1 Receptor/genetics , Programmed Cell Death 1 Receptor/immunology , Receptors, Antigen, T-Cell, gamma-delta/genetics , Receptors, Antigen, T-Cell, gamma-delta/immunology , Recurrence , Sequence Analysis, RNA , Signal Transduction , Single-Cell Analysis , Tumor Microenvironment/drug effects , Tumor Microenvironment/immunology
6.
Polymers (Basel) ; 13(17)2021 Sep 04.
Article in English | MEDLINE | ID: mdl-34503040

ABSTRACT

The aim of this work was to evaluate the potential of cold plasma polymerization as a simple, fast and versatile technique for deposition of protective hydrophobic and oleophobic polymer layers on hydrophilic biopolymer aerogels. Polymerization of different fluorinated monomers (octafluorocyclobutane C4F8 and perfluoro-acrylates PFAC-6 and PFAC-8) on aerogel monoliths derived from alginate, cellulose, whey protein isolate (WPI) and potato protein isolate (PPI) resulted in fast and significant surface hydrophobization after short process times of 5 min and led to superhydrophobic surfaces with static water contact angles up to 154° after application of poly-C4F8 coatings. Simultaneous introduction of hydro- and oleophobicity was possible by deposition of perfluoro-acrylates. While the porous structure of aerogels stayed intact during the process, polymerization inside the aerogels pores led to the generation of new porous moieties and resulted therefore in significant increase in the specific surface area. The magnitude of the effect depended on the individual process settings and on the overall porosity of the substrates. A maximization of specific surface area increase (+179 m2/g) was obtained by applying a pulsed wave mode in the C4F8-coating of alginate aerogels.

7.
Front Immunol ; 12: 712948, 2021.
Article in English | MEDLINE | ID: mdl-34566968

ABSTRACT

Infection and inflammation can augment local Na+ abundance. These increases in local Na+ levels boost proinflammatory and antimicrobial macrophage activity and can favor polarization of T cells towards a proinflammatory Th17 phenotype. Although neutrophils play an important role in fighting intruding invaders, the impact of increased Na+ on the antimicrobial activity of neutrophils remains elusive. Here we show that, in neutrophils, increases in Na+ (high salt, HS) impair the ability of human and murine neutrophils to eliminate Escherichia coli and Staphylococcus aureus. High salt caused reduced spontaneous movement, degranulation and impaired production of reactive oxygen species (ROS) while leaving neutrophil viability unchanged. High salt enhanced the activity of the p38 mitogen-activated protein kinase (p38/MAPK) and increased the interleukin (IL)-8 release in a p38/MAPK-dependent manner. Whereas inhibition of p38/MAPK did not result in improved neutrophil defense, pharmacological blockade of the phagocyte oxidase (PHOX) or its genetic ablation mimicked the impaired antimicrobial activity detected under high salt conditions. Stimulation of neutrophils with phorbol-12-myristate-13-acetate (PMA) overcame high salt-induced impairment in ROS production and restored antimicrobial activity of neutrophils. Hence, we conclude that high salt-impaired PHOX activity results in diminished antimicrobial activity. Our findings suggest that increases in local Na+ represent an ionic checkpoint that prevents excessive ROS production of neutrophils, which decreases their antimicrobial potential and could potentially curtail ROS-mediated tissue damage.


Subject(s)
Bacterial Infections/metabolism , Bacterial Infections/microbiology , Cellular Microenvironment , Neutrophils/physiology , Oxidoreductases/metabolism , Phagocytes/physiology , Sodium/metabolism , Animals , Bacterial Infections/immunology , Disease Resistance , Disease Susceptibility , Escherichia coli Infections/immunology , Escherichia coli Infections/metabolism , Escherichia coli Infections/microbiology , Host-Pathogen Interactions , Humans , Mice , NADPH Oxidases/metabolism , Reactive Oxygen Species/metabolism
8.
Angew Chem Int Ed Engl ; 60(37): 20178-20183, 2021 09 06.
Article in English | MEDLINE | ID: mdl-34081840

ABSTRACT

Covalent kinase inhibitors account for some of the most successful drugs that have recently entered the clinic and many others are in preclinical development. A common strategy is to target cysteines in the vicinity of the ATP binding site using an acrylamide electrophile. To increase the tissue selectivity of kinase inhibitors, it could be advantageous to control the reactivity of these electrophiles with light. Here, we introduce covalent inhibitors of the kinase JNK3 that function as photoswitchable affinity labels (PALs). Our lead compounds contain a diazocine photoswitch, are poor non-covalent inhibitors in the dark, and become effective covalent inhibitors after irradiation with visible light. Our proposed mode of action is supported by X-ray structures that explain why these compounds are unreactive in the dark and undergo proximity-based covalent attachment following exposure to light.


Subject(s)
Light , Protein Kinase Inhibitors/pharmacology , Protein Kinases/metabolism , Humans , Protein Kinase Inhibitors/chemistry
12.
Immunity ; 52(2): 295-312.e11, 2020 02 18.
Article in English | MEDLINE | ID: mdl-31924477

ABSTRACT

Specialized regulatory T (Treg) cells accumulate and perform homeostatic and regenerative functions in nonlymphoid tissues. Whether common precursors for nonlymphoid-tissue Treg cells exist and how they differentiate remain elusive. Using transcription factor nuclear factor, interleukin 3 regulated (Nfil3) reporter mice and single-cell RNA-sequencing (scRNA-seq), we identified two precursor stages of interleukin 33 (IL-33) receptor ST2-expressing nonlymphoid tissue Treg cells, which resided in the spleen and lymph nodes. Global chromatin profiling of nonlymphoid tissue Treg cells and the two precursor stages revealed a stepwise acquisition of chromatin accessibility and reprogramming toward the nonlymphoid-tissue Treg cell phenotype. Mechanistically, we identified and validated the transcription factor Batf as the driver of the molecular tissue program in the precursors. Understanding this tissue development program will help to harness regenerative properties of tissue Treg cells for therapy.


Subject(s)
Basic-Leucine Zipper Transcription Factors/metabolism , Lymph Nodes/immunology , Spleen/immunology , T-Lymphocytes, Regulatory/cytology , Adoptive Transfer , Animals , Basic-Leucine Zipper Transcription Factors/deficiency , Basic-Leucine Zipper Transcription Factors/genetics , Cell Differentiation/genetics , Chromatin/metabolism , GATA3 Transcription Factor/genetics , GATA3 Transcription Factor/metabolism , Gene Expression Profiling , Gene Expression Regulation/immunology , Interleukin-1 Receptor-Like 1 Protein/metabolism , Lectins, C-Type/genetics , Lectins, C-Type/metabolism , Mice , Organ Specificity/immunology , Receptors, Immunologic/genetics , Receptors, Immunologic/metabolism , T-Lymphocytes, Regulatory/metabolism
13.
Cell Death Dis ; 10(12): 944, 2019 12 10.
Article in English | MEDLINE | ID: mdl-31822659

ABSTRACT

Ecotropic virus integration site 1 (EVI1), whose overexpression characterizes a particularly aggressive subtype of acute myeloid leukemia (AML), enhanced anti-leukemic activities of all-trans retinoic acid (atRA) in cell lines and patient samples. However, the drivers of leukemia formation, therapy resistance, and relapse are leukemic stem cells (LSCs), whose properties were hardly reflected in these experimental setups. The present study was designed to address the effects of, and interactions between, EVI1 and retinoids in AML LSCs. We report that Evi1 reduced the maturation of leukemic cells and promoted the abundance, quiescence, and activity of LSCs in an MLL-AF9-driven mouse model of AML. atRA further augmented these effects in an Evi1 dependent manner. EVI1 also strongly enhanced atRA regulated gene transcription in LSC enriched cells. One of their jointly regulated targets, Notch4, was an important mediator of their effects on leukemic stemness. In vitro exposure of leukemic cells to a pan-RAR antagonist caused effects opposite to those of atRA. In vivo antagonist treatment delayed leukemogenesis and reduced LSC abundance, quiescence, and activity in Evi1high AML. Key results were confirmed in human myeloid cell lines retaining some stem cell characteristics as well as in primary human AML samples. In summary, our study is the first to report the importance of EVI1 for key properties of AML LSCs. Furthermore, it shows that atRA enhances, and a pan-RAR antagonist counteracts, the effects of EVI1 on AML stemness, thus raising the possibility of using RAR antagonists in the therapy of EVI1high AML.


Subject(s)
Leukemia, Myeloid, Acute/genetics , MDS1 and EVI1 Complex Locus Protein/genetics , Receptor, Notch4/genetics , Tretinoin/metabolism , Animals , Apoptosis/drug effects , Carcinogenesis/drug effects , Cell Proliferation/drug effects , Disease Models, Animal , Gene Expression Regulation, Leukemic/drug effects , Humans , Leukemia, Myeloid, Acute/pathology , Mice , Myeloid Cells/drug effects , Neoplastic Stem Cells/metabolism
14.
Acta Neuropathol Commun ; 7(1): 201, 2019 12 05.
Article in English | MEDLINE | ID: mdl-31806013

ABSTRACT

The presence of genome-wide DNA hypermethylation is a hallmark of lower grade gliomas (LGG) with isocitrate dehydrogenase (IDH) mutations. Further molecular classification of IDH mutant gliomas is defined by the presence (IDHmut-codel) or absence (IDHmut-noncodel) of hemizygous codeletion of chromosome arms 1p and 19q. Despite the DNA hypermethylation seen in bulk tumors, intra-tumoral heterogeneity at the epigenetic level has not been thoroughly analyzed. To address this question, we performed the first epigenetic profiling of single cells in a cohort of 5 gliomas with IDH1 mutation using single nucleus Assay for Transposase-Accessible Chromatin with high-throughput sequencing (snATAC-seq). Using the Fluidigm HT IFC microfluidics platform, we generated chromatin accessibility maps from 336 individual nuclei, and identified variable promoter accessibility of non-coding RNAs in LGGs. Interestingly, local chromatin structures of several non-coding RNAs are significant factors that contribute to heterogeneity, and show increased promoter accessibility in IDHmut-noncodel samples. As an example for clinical significance of this result, we identify CYTOR as a poor prognosis factor in gliomas with IDH mutation. Open chromatin assay points to differential accessibility of non-coding RNAs as an important source of epigenetic heterogeneity within individual tumors and between molecular subgroups. Rare populations of nuclei that resemble either IDH mutant molecular group co-exist within IDHmut-noncodel and IDHmut-codel groups, and along with non-coding RNAs may be an important issue to consider for future studies, as they may help guide predict treatment response and relapse.A web-based explorer for the data is available at shiny.turcanlab.org.


Subject(s)
Brain Neoplasms/genetics , Cell Nucleus/genetics , Chromatin/genetics , Epigenesis, Genetic/genetics , Glioma/genetics , Isocitrate Dehydrogenase/genetics , Brain Neoplasms/pathology , Cell Nucleus/pathology , Chromatin/pathology , Cohort Studies , Glioma/pathology , Humans , Mutation/genetics , Sequence Analysis, RNA/methods
15.
Sci Rep ; 9(1): 9139, 2019 06 24.
Article in English | MEDLINE | ID: mdl-31235852

ABSTRACT

Acute myeloid leukemia (AML) is a heterogeneous disease with respect to its genetic and molecular basis and to patients´ outcome. Clinical, cytogenetic, and mutational data are used to classify patients into risk groups with different survival, however, within-group heterogeneity is still an issue. Here, we used a robust likelihood-based survival modeling approach and publicly available gene expression data to identify a minimal number of genes whose combined expression values were prognostic of overall survival. The resulting gene expression signature (4-GES) consisted of 4 genes (SOCS2, IL2RA, NPDC1, PHGDH), predicted patient survival as an independent prognostic parameter in several cohorts of AML patients (total, 1272 patients), and further refined prognostication based on the European Leukemia Net classification. An oncogenic role of the top scoring gene in this signature, SOCS2, was investigated using MLL-AF9 and Flt3-ITD/NPM1c driven mouse models of AML. SOCS2 promoted leukemogenesis as well as the abundance, quiescence, and activity of AML stem cells. Overall, the 4-GES represents a highly discriminating prognostic parameter in AML, whose clinical applicability is greatly enhanced by its small number of genes. The newly established role of SOCS2 in leukemia aggressiveness and stemness raises the possibility that the signature might even be exploitable therapeutically.


Subject(s)
Leukemia, Myeloid, Acute/genetics , Leukemia, Myeloid, Acute/pathology , Suppressor of Cytokine Signaling Proteins/genetics , Animals , Cell Line, Tumor , Gene Expression Regulation, Neoplastic , Humans , Leukemia, Myeloid, Acute/diagnosis , Mice , Neoplastic Stem Cells/pathology , Prognosis
16.
Cell ; 176(6): 1407-1419.e14, 2019 03 07.
Article in English | MEDLINE | ID: mdl-30827680

ABSTRACT

The function of somatic stem cells declines with age. Understanding the molecular underpinnings of this decline is key to counteract age-related disease. Here, we report a dramatic drop in the neural stem cells (NSCs) number in the aging murine brain. We find that this smaller stem cell reservoir is protected from full depletion by an increase in quiescence that makes old NSCs more resistant to regenerate the injured brain. Once activated, however, young and old NSCs show similar proliferation and differentiation capacity. Single-cell transcriptomics of NSCs indicate that aging changes NSCs minimally. In the aging brain, niche-derived inflammatory signals and the Wnt antagonist sFRP5 induce quiescence. Indeed, intervention to neutralize them increases activation of old NSCs during homeostasis and following injury. Our study identifies quiescence as a key feature of old NSCs imposed by the niche and uncovers ways to activate NSCs to repair the aging brain.


Subject(s)
Brain/physiology , Age Factors , Animals , Brain/cytology , Cell Differentiation/physiology , Cell Division/physiology , Cell Proliferation/physiology , Cellular Senescence/physiology , Homeostasis , Male , Mice , Mice, Inbred C57BL , Nerve Regeneration , Neural Stem Cells/cytology , Neural Stem Cells/physiology , Neurogenesis , Stem Cell Niche
17.
Proc Natl Acad Sci U S A ; 115(26): E5980-E5989, 2018 06 26.
Article in English | MEDLINE | ID: mdl-29895693

ABSTRACT

CD8+ T cells are considered prototypical cells of adaptive immunity. Here, we uncovered a distinct CD8+ T cell population expressing the activating natural killer (NK) receptor NKp30 in the peripheral blood of healthy individuals. We revealed that IL-15 could de novo induce NKp30 expression in a population of CD8+ T cells and drive their differentiation toward a broad innate transcriptional landscape. The adaptor FcεRIγ was concomitantly induced and was shown to be crucial to enable NKp30 cell-surface expression and function in CD8+ T cells. FcεRIγ de novo expression required promoter demethylation and was accompanied by acquisition of the signaling molecule Syk and the "innate" transcription factor PLZF. IL-15-induced NKp30+CD8+ T cells exhibited high NK-like antitumor activity in vitro and were able to synergize with T cell receptor signaling. Importantly, this population potently controlled tumor growth in a preclinical xenograft mouse model. Our study, while blurring the borders between innate and adaptive immunity, reveals a unique NKp30+FcεRIγ+CD8+ T cell population with high antitumor therapeutic potential.


Subject(s)
CD8-Positive T-Lymphocytes/immunology , Immunity, Cellular , Killer Cells, Natural/immunology , Natural Cytotoxicity Triggering Receptor 3/immunology , Neoplasms/immunology , Receptors, Fc/immunology , CD8-Positive T-Lymphocytes/pathology , Cell Line, Tumor , Female , HEK293 Cells , Humans , Killer Cells, Natural/pathology , Male , Neoplasms/pathology
18.
Behav Sci (Basel) ; 8(2)2018 Feb 22.
Article in English | MEDLINE | ID: mdl-29470435

ABSTRACT

Stress is one of the world's largest health problems, leading to exhaustion, burnout, anxiety, a weak immune system, or even organ damage. In Germany, stress-induced work absenteeism costs about 20 billion Euros per year. Therefore, it is not surprising that the Central Federal Association of the public Health Insurance Funds in Germany ascribes particular importance to stress prevention and stress management as well as health enhancing measures. Building on current integrative and embodied stress theories, Creative Arts Therapies (CATs) or arts interventions are an innovative way to prevent stress and improve stress management. CATs encompass art, music, dance/movement, and drama therapy as their four major modalities. In order to obtain an overview of CATs and arts interventions' efficacy in the context of stress reduction and management, we conducted a systematic review with a search in the following data bases: Academic Search Complete, ERIC, Medline, Psyndex, PsycINFO and SocINDEX. Studies were included employing the PICOS principle and rated according to their evidence level. We included 37 studies, 73% of which were randomized controlled trials. 81.1% of the included studies reported a significant reduction of stress in the participants due to interventions of one of the four arts modalities.

20.
Nat Immunol ; 18(10): 1160-1172, 2017 Oct.
Article in English | MEDLINE | ID: mdl-28783152

ABSTRACT

Regulatory T cells (Treg cells) perform two distinct functions: they maintain self-tolerance, and they support organ homeostasis by differentiating into specialized tissue Treg cells. We found that epigenetic modifications defined the molecular characteristics of tissue Treg cells. Tagmentation-based whole-genome bisulfite sequencing revealed more than 11,000 regions that were methylated differentially in pairwise comparisons of tissue Treg cell populations and lymphoid T cells. Similarities in the epigenetic landscape led to the identification of a common tissue Treg cell population that was present in many organs and was characterized by gain and loss of DNA methylation that included many gene sites associated with the TH2 subset of helper T cells, such as the gene encoding cytokine IL-33 receptor ST2, as well as the production of tissue-regenerative factors. Furthermore, the ST2-expressing population was dependent on the transcriptional regulator BATF and could be expanded by IL-33. Thus, tissue Treg cells integrate multiple waves of epigenetic reprogramming that define their tissue-restricted specialization.


Subject(s)
DNA Methylation , Genome-Wide Association Study , T-Lymphocytes, Regulatory/metabolism , Animals , Biomarkers , Cluster Analysis , Computational Biology/methods , CpG Islands , Epigenesis, Genetic , Gene Expression Profiling , Gene Expression Regulation , Gene Ontology , High-Throughput Nucleotide Sequencing , Immunophenotyping , Mice , Mice, Transgenic , Molecular Sequence Annotation , Organ Specificity/genetics , Organ Specificity/immunology , Promoter Regions, Genetic , Th2 Cells/metabolism , Transcription Initiation Site , Transcriptome
SELECTION OF CITATIONS
SEARCH DETAIL