Network


Latest external collaboration on country level. Dive into details by clicking on the dots.

Hotspot


Dive into the research topics where Kristin Seré is active.

Publication


Featured researches published by Kristin Seré.


Immunity | 2012

Two Distinct Types of Langerhans Cells Populate the Skin during Steady State and Inflammation

Kristin Seré; Jea-Hyun Baek; Julia L. Ober-Blöbaum; Gerhard Müller-Newen; Frank Tacke; Yoshifumi Yokota; Martin Zenke; Thomas Hieronymus

Langerhans cells (LCs), the dendritic cells (DCs) in skin epidermis, possess an exceptional life cycle and developmental origin. Here we identified two types of LCs, short-term and long-term LCs, which transiently or stably reconstitute the LC compartment, respectively. Short-term LCs developed from Gr-1(hi) monocytes under inflammatory conditions and occurred independently of the transcription factor Id2. Long-term LCs arose from bone marrow in steady state and were critically dependent on Id2. Surface marker and gene expression analysis positioned short-term LCs close to Gr-1(hi) monocytes, which is indicative of their monocytic origin. We also show that LC reconstitution after UV light exposure occurs in two waves: an initial fast and transient wave of Gr-1(hi) monocyte-derived short-term LCs is followed by a second wave of steady-state precursor-derived long-term LCs. Our data demonstrate the presence of two types of LCs that develop through different pathways in inflammation and steady state.


Journal of Immunology | 2010

TGF-β1 Accelerates Dendritic Cell Differentiation from Common Dendritic Cell Progenitors and Directs Subset Specification toward Conventional Dendritic Cells

Piritta Felker; Kristin Seré; Qiong Lin; Christiane Becker; Mihail Hristov; Thomas Hieronymus; Martin Zenke

Dendritic cells (DCs) in lymphoid tissue comprise conventional DCs (cDCs) and plasmacytoid DCs (pDCs) that develop from common DC progenitors (CDPs). CDPs are Flt3+c-kitintM-CSFR+ and reside in bone marrow. In this study, we describe a two-step culture system that recapitulates DC development from c-kithiFlt3−/lo multipotent progenitors (MPPs) into CDPs and further into cDC and pDC subsets. MPPs and CDPs are amplified in vitro with Flt3 ligand, stem cell factor, hyper–IL-6, and insulin-like growth factor-1. The four-factor mixture readily induces self-renewal of MPPs and their progression into CDPs and has no self-renewal activity on CDPs. The amplified CDPs respond to all known DC poietins and generate all lymphoid tissue DCs in vivo and in vitro. Additionally, in vitro CDPs recapitulate the cell surface marker and gene expression profile of in vivo CDPs and possess a DC-primed transcription profile. TGF-β1 impacts on CDPs and directs their differentiation toward cDCs. Genome-wide gene expression profiling of TGF-β1–induced genes identified instructive transcription factors for cDC subset specification, such as IFN regulatory factor-4 and RelB. TGF-β1 also induced the transcription factor inhibitor of differentiation/DNA binding 2 that suppresses pDC development. Thus, TGF-β1 directs CDP differentiation into cDCs by inducing both cDC instructive factors and pDC inhibitory factors.


Immunity | 2015

Distinct Murine Mucosal Langerhans Cell Subsets Develop from Pre-dendritic Cells and Monocytes.

Tal Capucha; Gabriel Mizraji; Hadas Segev; Ronnie Blecher-Gonen; Deborah R. Winter; Abed Khalaileh; Yaara Tabib; Tsipora Attal; Maria Nassar; Katya Zelentsova; Hen Kisos; Martin Zenke; Kristin Seré; Thomas Hieronymus; Tal Burstyn-Cohen; Ido Amit; Asaf Wilensky; Avi-Hai Hovav

Langerhans cells (LCs) populate the mucosal epithelium, a major entry portal for pathogens, yet their ontogeny remains unclear. We found that, in contrast to skin LCs originating from self-renewing radioresistant embryonic precursors, oral mucosal LCs derive from circulating radiosensitive precursors. Mucosal LCs can be segregated into CD103(+)CD11b(lo) (CD103(+)) and CD11b(+)CD103(-) (CD11b(+)) subsets. We further demonstrated that similar to non-lymphoid dendritic cells (DCs), CD103(+) LCs originate from pre-DCs, whereas CD11b(+) LCs differentiate from both pre-DCs and monocytic precursors. Despite this ontogenetic discrepancy between skin and mucosal LCs, the transcriptomic signature and immunological function of oral LCs highly resemble those of skin LCs but not DCs. These findings, along with the epithelial position, morphology, and expression of the LC-associated phenotype strongly suggest that oral mucosal LCs are genuine LCs. Collectively, in a tissue-dependent manner, murine LCs differentiate from at least three distinct precursors (embryonic, pre-DC, and monocytic) in steady state.


European Journal of Immunology | 2007

Transforming growth factor β1 up-regulates interferon regulatory factor 8 during dendritic cell development

Xin-Sheng Ju; David Ruau; Piritta Jäntti; Kristin Seré; Christiane Becker; Eliza Wiercinska; Clemens Bartz; Bettina Erdmann; Steven Dooley; Martin Zenke

Langerhans cells (LC) represent the cutaneous contingent of dendritic cells (DC). Their development critically depends on transforming growth factor β1 (TGF‐β1) as demonstrated by analysis of TGF‐β1–/– mice, which lack LC. Here we used a two‐step culture system and transcriptional profiling by DNA microarrays to search for TGF‐β1 target genes in DC. The study identified interferon regulatory factor 8 (IRF‐8) as a novel target gene of TGF‐β1 signaling in DC. TGF‐β1 effectively induced Smad2/3 phosphorylation and IRF‐8 RNA and protein expression. Blocking the TGF‐β1/Smad pathway by ectopic expression of inhibitory Smad7 and by SB431542 inhibitor abolished TGF‐β1 induced up‐regulation of IRF‐8. Furthermore, TGF‐β1‐dependent induction of IRF‐8 occurred in the absence of protein biosynthesis, suggesting a direct action of TGF‐β1/Smad signaling on IRF‐8 gene activity. TGF‐β1 also induced expression of the chemokine receptor CCR7 and enhanced DC migration towards CCR7 ligand ELC. DC of IRF‐8–/– mice show reduced CCR7 expression and migratory activity, thereby implicating the TGF‐β1/Smad/IRF‐8 signaling pathway in CCR7 regulation. Thus, this study identified a novel TGF‐β1/Smad/IRF‐8 signaling pathway with an impact on DC phenotype and function.


Seminars in Cell & Developmental Biology | 2015

The clash of Langerhans cell homeostasis in skin: Should I stay or should I go?

Thomas Hieronymus; Martin Zenke; Jea-Hyun Baek; Kristin Seré

Langerhans cells (LC), the skin epidermal contingent of dendritic cells (DC), possess an exceptional life cycle and developmental origin. LC, like all mature blood cells, develop from haematopoietic stem cells (HSC) through successive steps of lineage commitment and differentiation. However, LC development is different to that of other DC subsets and not yet fully understood. Haematopoietic cell fate decisions are instructed by specific growth factors and cytokines produced in specialized microenvironments or niches. Upon ligand binding the cognate surface receptors on HSC and further restricted progenitor cells regulate the signalling pathways that eventually leads to the execution of lineage-determining genetic programs. In this review we focus on a specific set of surface receptor kinases that have been identified as critical regulators of LC development using genetically modified mice. Recent studies suggest for some of these kinases to impact on LC/LC progenitor interaction with the local niche by regulating adhesion and/or migration. During embryonic development, in wound healing and aberrantly in tumour invasion the same kinase receptors control a genetic program known as epithelial-to-mesenchymal-transition (EMT). We will discuss how EMT and its reverse program of mesenchymal-to-epithelial-transition (MET) can serve as universal concepts operating also in LC development.


European Journal of Cell Biology | 2012

Dendritic cell lineage commitment is instructed by distinct cytokine signals.

Kristin Seré; Qiong Lin; Piritta Felker; Nina Rehage; Theresa Klisch; Inga Ortseifer; Thomas Hieronymus; Stefan Rose-John; Martin Zenke

Dendritic cells (DC) develop from hematopoietic stem cells, which is guided by instructive signals through cytokines. DC development progresses from multipotent progenitors (MPP) via common DC progenitors (CDP) into DC. Flt3 ligand (Flt3L) signaling via the Flt3/Stat3 pathway is of pivotal importance for DC development under steady state conditions. Additional factors produced during steady state or inflammation, such as TGF-β1 or GM-CSF, also influence the differentiation potential of MPP and CDP. Here, we studied how gp130, GM-CSF and TGF-β1 signaling influence DC lineage commitment from MPP to CDP and further into DC. We observed that activation of gp130 signaling promotes expansion of MPP. Additionally, gp130 signaling inhibited Flt3L-driven DC differentiation, but had little effect on GM-CSF-driven DC development. The inflammatory cytokine GM-CSF induces differentiation of MPP into inflammatory DC and blocks steady state DC development. Global transcriptome analysis revealed a GM-CSF-driven gene expression repertoire that primes MPP for differentiation into inflammatory DC. Finally, TGF-β1 induces expression of DC-lineage affiliated genes in MPP, including Flt3, Irf-4 and Irf-8. Under inflammatory conditions, however, the effect of TGF-β1 is altered: Flt3 is not upregulated, indicating that an inflammatory environment inhibits steady state DC development. Altogether, our data indicate that distinct cytokine signals produced during steady state or inflammation have a different outcome on DC lineage commitment and differentiation.


BMC Systems Biology | 2014

TGF-β stimulation in human and murine cells reveals commonly affected biological processes and pathways at transcription level

Khalid Abnaof; Nikhil Mallela; Gudrun Walenda; Steffen K. Meurer; Kristin Seré; Qiong Lin; Bert Smeets; Kurt Hoffmann; Wolfgang Wagner; Martin Zenke; Ralf Weiskirchen; Holger Fröhlich

BackgroundThe TGF-β signaling pathway is a fundamental pathway in the living cell, which plays a key role in many central cellular processes. The complex and sometimes contradicting mechanisms by which TGF-β yields phenotypic effects are not yet completely understood. In this study we investigated and compared the transcriptional response profile of TGF-β1 stimulation in different cell types. For this purpose, extensive experiments are performed and time-course microarray data are generated in human and mouse parenchymal liver cells, human mesenchymal stromal cells and mouse hematopoietic progenitor cells at different time points. We applied a panel of bioinformatics methods on our data to uncover common patterns in the dynamic gene expression response in respective cells.ResultsOur analysis revealed a quite variable and multifaceted transcriptional response profile of TGF-β1 stimulation, which goes far beyond the well-characterized classical TGF-β1 signaling pathway. Nonetheless, we could identify several commonly affected processes and signaling pathways across cell types and species. In addition our analysis suggested an important role of the transcription factor EGR1, which appeared to have a conserved influence across cell-types and species. Validation via an independent dataset on A549 lung adenocarcinoma cells largely confirmed our findings. Network analysis suggested explanations, how TGF-β1 stimulation could lead to the observed effects.ConclusionsThe analysis of dynamical transcriptional response to TGF-β treatment experiments in different human and murine cell systems revealed commonly affected biological processes and pathways, which could be linked to TGF-β1 via network analysis. This helps to gain insights about TGF-β pathway activities in these cell systems and its conserved interactions between the species and tissue types.


Stem Cells | 2017

Modelling IRF8 Deficient Human Hematopoiesis and Dendritic Cell Development with Engineered iPS Cells

Stephanie Sontag; Malrun Förster; Jie Qin; Paul Wanek; Saskia Mitzka; Herdit M. Schüler; Steffen Koschmieder; Stefan Rose-John; Kristin Seré; Martin Zenke

Human induced pluripotent stem (iPS) cells can differentiate into cells of all three germ layers, including hematopoietic stem cells and their progeny. Interferon regulatory factor 8 (IRF8) is a transcription factor, which acts in hematopoiesis as lineage determining factor for myeloid cells, including dendritic cells (DC). Autosomal recessive or dominant IRF8 mutations occurring in patients cause severe monocytic and DC immunodeficiency. To study IRF8 in human hematopoiesis we generated human IRF8−/− iPS cells and IRF8−/− embryonic stem (ES) cells using RNA guided CRISPR/Cas9n genome editing. Upon induction of hematopoietic differentiation, we demonstrate that IRF8 is dispensable for iPS cell and ES cell differentiation into hemogenic endothelium and for endothelial‐to‐hematopoietic transition, and thus development of hematopoietic progenitors. We differentiated iPS cell and ES cell derived progenitors into CD141+ cross‐presenting cDC1 and CD1c+ classical cDC2 and CD303+ plasmacytoid DC (pDC). We found that IRF8 deficiency compromised cDC1 and pDC development, while cDC2 development was largely unaffected. Additionally, in an unrestricted differentiation regimen, IRF8−/− iPS cells and ES cells exhibited a clear bias toward granulocytes at the expense of monocytes. IRF8−/− DC showed reduced MHC class II expression and were impaired in cytokine responses, migration, and antigen presentation. Taken together, we engineered a human IRF8 knockout model that allows studying molecular mechanisms of human immunodeficiencies in vitro, including the pathophysiology of IRF8 deficient DC. Stem Cells 2017;35:898–908


European Journal of Immunology | 2014

Dendritic cell development requires histone deacetylase activity

Heike Chauvistré; Caroline Küstermann; Nina Rehage; Theresa Klisch; Saskia Mitzka; Piritta Felker; Stefan Rose-John; Martin Zenke; Kristin Seré

DCs develop from multipotent progenitors (MPPs), which commit into DC‐restricted common dendritic cell progenitors (CDPs). CDPs further differentiate into classical DCs (cDCs) and plasmacytoid DCs (pDCs). Here, we studied the impact of histone acetylation on DC development in C57BL/6 mice by interfering with histone acetylation and deacetylation, employing histone deacetylase (HDAC) inhibitors. We observed that commitment of MPPs into CDPs was attenuated by HDAC inhibition and that pDC development was specifically blocked. Gene expression profiling revealed that HDAC inhibition prevents establishment of a DC‐specific gene expression repertoire. Importantly, protein levels of the core DC transcription factor PU.1 were reduced in HDAC inhibitor‐treated cells and consequently PU.1 recruitment at PU.1 target genes Fms‐like tyrosine kinase 3 (Flt3), interferon regulatory factor 8 (IRF8), and PU.1 itself was impaired. Thus, our results demonstrate that attenuation of PU.1 expression by HDAC inhibition causes reduced expression of key DC regulators, which results in attenuation of DC development. We propose that chromatin modifiers, such as HDACs, are required for establishing a DC gene network, where Flt3/STAT3 signaling drives PU.1 and IRF8 expression and DC development. Taken together, our study identifies HDACs as critical regulators of DC lineage commitment and development.


Nucleic Acids Research | 2015

Epigenetic program and transcription factor circuitry of dendritic cell development

Qiong Lin; Heike Chauvistré; Ivan G. Costa; Eduardo G. Gusmao; Saskia Mitzka; Sonja Hänzelmann; Bianka Baying; Theresa Klisch; Richard Moriggl; Benoit Hennuy; H.J.M. Smeets; Kurt Hoffmann; Vladimir Benes; Kristin Seré; Martin Zenke

Dendritic cells (DC) are professional antigen presenting cells that develop from hematopoietic stem cells through successive steps of lineage commitment and differentiation. Multipotent progenitors (MPP) are committed to DC restricted common DC progenitors (CDP), which differentiate into specific DC subsets, classical DC (cDC) and plasmacytoid DC (pDC). To determine epigenetic states and regulatory circuitries during DC differentiation, we measured consecutive changes of genome-wide gene expression, histone modification and transcription factor occupancy during the sequel MPP-CDP-cDC/pDC. Specific histone marks in CDP reveal a DC-primed epigenetic signature, which is maintained and reinforced during DC differentiation. Epigenetic marks and transcription factor PU.1 occupancy increasingly coincide upon DC differentiation. By integrating PU.1 occupancy and gene expression we devised a transcription factor regulatory circuitry for DC commitment and subset specification. The circuitry provides the transcription factor hierarchy that drives the sequel MPP-CDP-cDC/pDC, including Irf4, Irf8, Tcf4, Spib and Stat factors. The circuitry also includes feedback loops inferred for individual or multiple factors, which stabilize distinct stages of DC development and DC subsets. In summary, here we describe the basic regulatory circuitry of transcription factors that drives DC development.

Collaboration


Dive into the Kristin Seré's collaboration.

Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Qiong Lin

RWTH Aachen University

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Researchain Logo
Decentralizing Knowledge