Network


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

Hotspot


Dive into the research topics where Carolin Daniel is active.

Publication


Featured researches published by Carolin Daniel.


Journal of Experimental Medicine | 2009

Retinoic acid can enhance conversion of naive into regulatory T cells independently of secreted cytokines

Jens Nolting; Carolin Daniel; Sabine Reuter; Christina Stuelten; Peng Li; Henry M. Sucov; Byung-Gyu Kim; John J. Letterio; Karsten Kretschmer; Hye Jung Kim; Harald von Boehmer

It has been reported that retinoic acid (RA) enhances regulatory T (T reg) cell conversion by inhibiting the secretion of cytokines that interfere with conversion. This report shows that these conclusions provide a partial explanation at best. First, RA not only interfered with cytokine secretion but also with the ability of these cytokines to inhibit T reg cell conversion of naive T cells. Furthermore, RA enhanced conversion even in the absence of inhibitory cytokines. The latter effect depended on the RA receptor α (RARα) but did not require Smad3, despite the fact that RA enhanced Smad3 expression. The RARα1 isoform was not essential for RA-dependent enhancement of transforming growth factor β–driven conversion, suggesting that conversion can also be mediated by RARα2. Interleukin (IL)-6 strongly reduced RARα expression levels such that a deficiency of the predominant RARα1 isoform leaves too little RARα2 for RA to inhibit the generation of Th17 cells in the presence of IL-6.


Nature Reviews Drug Discovery | 2013

Therapeutic opportunities for manipulating TReg cells in autoimmunity and cancer

Harald von Boehmer; Carolin Daniel

Forkhead box P3 (FOXP3)-expressing regulatory T (TReg) cells have a pivotal role in the regulation of immune responses and in the maintenance of immunological self-tolerance. These cells have emerged as attractive targets for strategies that allow the steering of immune responses in desired directions — arming the immune system to destroy infected cells and cancer cells or downregulating it to limit tissue destruction in autoimmunity. Efforts to understand the generation, activation and function of TReg cells should permit the development of therapeutics for reprogramming the immune system. In this Review, we discuss insights into the generation of TReg cells, their involvement in disease and the molecular basis of the dominant tolerance exerted by FOXP3+ TReg cells that could permit their safe and specific manipulation in humans.


Journal of Experimental Medicine | 2011

Prevention of type 1 diabetes in mice by tolerogenic vaccination with a strong agonist insulin mimetope

Carolin Daniel; Benno Weigmann; Roderick T. Bronson; Harald von Boehmer

Subimmunogenic vaccination with an agonist mimetope of insulin converts naive T cells into regulatory T cells and prevents type 1 diabetes in NOD mice.


Proceedings of the National Academy of Sciences of the United States of America | 2010

Enhancement of antigen-specific Treg vaccination in vivo

Carolin Daniel; Kerstin Wennhold; Hye-Jung Kim; Harald von Boehmer

The conversion of naive T cells into Treg can be achieved in vivo by delivery of antigen under subimmunogenic conditions. Here we have examined several drugs for their ability to enhance the conversion process in vivo and have found that the rapamycin analog everolimus potently enhances Treg conversion by interfering with T-cell costimulation, reducing cell division and thereby activation of DNA methyltransferase 1 as well as by reducing T-cell activation through the ATP-gated P2×7 receptor controlling Ca2+ influx. The resulting Tregs exhibit increased stability of Foxp3 expression even when generated in TGFβ-containing media in vitro. Thus the mammalian target of rapamycin (mTOR) inhibitor everolimus in addition to inhibiting immune responses enhances Treg conversion by several distinct pathways. The converted Tregs can be further expanded by injection of IL-2/IL-2ab complexes. These complexes also increase the number of CD25+Foxp3− cells that, however, do not represent cytokine secreting effector cells but anergic cells, some of which can secrete IL-10 and can themselves be considered regulatory T cells as well. The combined use of everolimus and IL-2/IL-2ab complexes in vivo makes it feasible to achieve highly effective antigen-driven conversion of naive T cells into Treg and their expansion in vivo and thereby the described protocols constitute important tools to achieve immunological tolerance by Treg vaccination.


Cancer Research | 2012

Transcription factor NFATc2 controls the emergence of colon cancer associated with IL-6-dependent colitis

Katharina Gerlach; Carolin Daniel; Hans A. Lehr; Alexei Nikolaev; Thomas Gerlach; Raja Atreya; Stefan Rose-John; Markus F. Neurath; Benno Weigmann

NFAT transcription factors control T-cell activation and function. Specifically, the transcription factor NFATc2 affects the regulation of cell differentiation and growth and plays a critical role in the development of colonic inflammation. Here, we used an experimental model of colitis-associated colorectal carcinoma to investigate the contribution of NFATc2 to the promotion of colonic tumors. Compared with wild-type animals that readily presented with multiple colon tumors, NFATc2-deficient mice were protected from tumor development. This observed decrease in colonic tumor progression was associated with reduced endoscopic inflammation, increased apoptosis of lamina propria T lymphocytes, and significantly reduced levels of the critical proinflammatory cytokines interleukin (IL)-21 and IL-6. Administration of hyper IL-6 abrogated protection from tumor progression in NFATc2-knockout mice and restored tumor incidence to control levels. Taken together, our findings highlight a pivotal role for NFATc2 in the establishment of inflammation-associated colorectal tumors mediated by control of IL-6 expression.


Methods of Molecular Biology | 2011

Antigen-Specific Induction of Regulatory T Cells In Vivo and In Vitro

Carolin Daniel; Hidde L. Ploegh; Harald von Boehmer

The peripheral induction of Foxp3-expressing regulatory T cells outside the thymus is required in order to maintain local homeostasis in distinct microenvironments such as the gut. Extrathymic induction of Treg may also be exploited to prevent unwanted immune responses. Here, we discuss the methodology allowing for the stable de novo generation of Tregs specific for foreign antigens in peripheral lymphoid tissue via subimmunogenic peptide delivery using either peptide contained in fusion antibodies directed against the DEC205 endocytotic receptor on steady-state dendritic cells or the implantation of peptide-delivering osmotic mini-pumps. Furthermore, we also address methods in order to achieve TGFβ-dependent Treg conversion in vitro, thereby mainly focusing on the role of retinoic acid (RA) to enhance TGFβ-dependent conversion into Tregs.


Advances in Immunology | 2011

Extrathymic Generation of Regulatory T Cells—Chances and Challenges for Prevention of Autoimmune Disease

Carolin Daniel; Harald von Boehmer

Fopx3(+) expressing regulatory T cells (Tregs) function as an indispensable cellular constituent of the immune system by establishing and maintaining immunological self-tolerance. T cell receptor (TCR) ligands of high agonist activity, when applied in vivo under subimmunogenic conditions, convert naive but not activated T cells into stable Tregs expressing Foxp3. Tolerogenic vaccination with strong-agonist mimetopes of self-antigens may function as a safe and highly specific instrument in the prevention of autoimmune disease by promoting self-antigen-specific tolerance. In this review, we address the requirements for generation of dominant tolerance exerted by Foxp3(+) Tregs in autoimmune disease with special focus on type 1 diabetes (T1D). Further understanding of differentiation of T cells into Tregs at the cellular and molecular level will facilitate development of additional tolerogenic vaccination strategies that can be used in prevention as well as therapeutically to combat unwanted immunity.


Seminars in Immunology | 2011

Extra-thymically induced regulatory T cells: do they have potential in disease prevention?

Carolin Daniel; Harald von Boehmer

Fopx3(+) Treg safeguard against autoimmune diseases and immune pathology. The extrathymic conversion of naïve T cells into Foxp3(+) regulatory T cells can be achieved in vivo by the delivery of strong-agonist ligands under subimmunogenic conditions. Tolerogenic vaccination with strong-agonist mimetopes of self-antigen to promote self-antigen specific tolerance may represent the most specific and safest means of preventing autoimmunity. This review discusses the requirements for induction of dominant tolerance exerted by Foxp3(+) Tregs in autoimmunity with special emphasis on their impact to interfere with T1D. The future goals are the understanding of self-non-self discrimination at the cellular and molecular level, which should then enable investigators to develop clinical vaccination protocols that specifically interfere with unwanted immune responses.


Proceedings of the National Academy of Sciences of the United States of America | 2016

miRNA92a targets KLF2 and the phosphatase PTEN signaling to promote human T follicular helper precursors in T1D islet autoimmunity

Isabelle Serr; Rainer W. Fürst; Verena B. Ott; Martin G. Scherm; Alexei Nikolaev; Füsun Gökmen; Stefanie Kälin; Stephanie Zillmer; Melanie Bunk; Benno Weigmann; Nicole Kunschke; Brigitta Loretz; Claus-Michael Lehr; Benedikt Kirchner; Bettina Haase; Michael W. Pfaffl; Ari Waisman; Richard A. Willis; Anette-G. Ziegler; Carolin Daniel

Significance The onset of type 1 diabetes autoimmunity is indicated by the development of multiple islet autoantibodies, produced by B cells with the help of T follicular helper (TFH) cells. MicroRNAs (miRNAs) are small noncoding RNAs that regulate cellular states, as immune activation, making them suitable targets for disease intervention. Here, we show an enrichment of insulin-specific C-X-C chemokine receptor type 5 (CXCR5)+CD4+ TFH precursors correlating with high miRNA92a abundance during onset of autoimmunity and identify Krueppel-like factor 2 (KLF2) as a target for miRNA92a. We demonstrate that miRNA92a inhibition blocks TFH induction and reduces murine islet autoimmunity in vivo. Therefore, we propose miRNA92a and the phosphatase and tension homolog-phosphoinositol-3-kinase-KLF2 signaling network as possible innovative precision medicine targets to interfere with aberrant immune activation in islet autoimmunity. Aberrant immune activation mediated by T effector cell populations is pivotal in the onset of autoimmunity in type 1 diabetes (T1D). T follicular helper (TFH) cells are essential in the induction of high-affinity antibodies, and their precursor memory compartment circulates in the blood. The role of TFH precursors in the onset of islet autoimmunity and signaling pathways regulating their differentiation is incompletely understood. Here, we provide direct evidence that during onset of islet autoimmunity, the insulin-specific target T-cell population is enriched with a C-X-C chemokine receptor type 5 (CXCR5)+CD4+ TFH precursor phenotype. During onset of islet autoimmunity, the frequency of TFH precursors was controlled by high expression of microRNA92a (miRNA92a). miRNA92a-mediated TFH precursor induction was regulated by phosphatase and tension homolog (PTEN) - phosphoinositol-3-kinase (PI3K) signaling involving PTEN and forkhead box protein O1 (Foxo1), supporting autoantibody generation and triggering the onset of islet autoimmunity. Moreover, we identify Krueppel-like factor 2 (KLF2) as a target of miRNA92a in regulating human TFH precursor induction. Importantly, a miRNA92a antagomir completely blocked induction of human TFH precursors in vitro. More importantly, in vivo application of a miRNA92a antagomir to nonobese diabetic (NOD) mice with ongoing islet autoimmunity resulted in a significant reduction of TFH precursors in peripheral blood and pancreatic lymph nodes. Moreover, miRNA92a antagomir application reduced immune infiltration and activation in pancreata of NOD mice as well as humanized NOD Scid IL2 receptor gamma chain knockout (NSG) human leucocyte antigen (HLA)-DQ8 transgenic animals. We therefore propose that miRNA92a and the PTEN-PI3K-KLF2 signaling network could function as targets for innovative precision medicines to reduce T1D islet autoimmunity.


BioDrugs | 2014

Treg vaccination in autoimmune type 1 diabetes.

Isabelle Serr; Benno Weigmann; Randi Kristina Franke; Carolin Daniel

Foxp3+ regulatory T (Treg) cells are critical contributors to the establishment and maintenance of immunological self-tolerance. Autoimmune type 1 diabetes (T1D) is characterized by the loss of self-tolerance to the insulin-producing β cells in the pancreas and the destruction of β cells, resulting in the development of chronic hyperglycemia at diagnosis. The application of strong-agonistic T-cell receptor ligands provided under subimmunogenic conditions functions as a critical means for the efficient de novo conversion of naive CD4+ T cells into Foxp3+ Treg cells. The specific induction of Treg cells upon supply of strong-agonistic variants of certain self-antigens could therefore function as a critical instrument in order to achieve safe and specific prevention of autoimmunity such as T1D via the restoration of self-tolerance. Such immunotherapeutic strategies are being developed, and in the case of T1D aim to restrict autoimmunity and β-cell destruction. In this review, we discuss the requirements and opportunities for Treg-based tolerance approaches with the goal of interfering with autoimmune T1D.

Collaboration


Dive into the Carolin Daniel's collaboration.

Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Katharina Gerlach

University of Erlangen-Nuremberg

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Markus F. Neurath

University of Erlangen-Nuremberg

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Raja Atreya

University of Erlangen-Nuremberg

View shared research outputs
Researchain Logo
Decentralizing Knowledge