56 results on '"Carolin Daniel"'
Search Results
2. The thymocyte-specific RNA-binding protein Arpp21 provides TCR repertoire diversity by binding to the 3’-UTR and promoting Rag1 mRNA expression
- Author
-
Meng Xu, Taku Ito-Kureha, Hyun-Seo Kang, Aleksandar Chernev, Timsse Raj, Kai P. Hoefig, Christine Hohn, Florian Giesert, Yinhu Wang, Wenliang Pan, Natalia Ziętara, Tobias Straub, Regina Feederle, Carolin Daniel, Barbara Adler, Julian König, Stefan Feske, George C. Tsokos, Wolfgang Wurst, Henning Urlaub, Michael Sattler, Jan Kisielow, F. Gregory Wulczyn, Marcin Łyszkiewicz, and Vigo Heissmeyer
- Subjects
Science - Abstract
Abstract The regulation of thymocyte development by RNA-binding proteins (RBPs) is largely unexplored. We identify 642 RBPs in the thymus and focus on Arpp21, which shows selective and dynamic expression in early thymocytes. Arpp21 is downregulated in response to T cell receptor (TCR) and Ca2+ signals. Downregulation requires Stim1/Stim2 and CaMK4 expression and involves Arpp21 protein phosphorylation, polyubiquitination and proteasomal degradation. Arpp21 directly binds RNA through its R3H domain, with a preference for uridine-rich motifs, promoting the expression of target mRNAs. Analysis of the Arpp21–bound transcriptome reveals strong interactions with the Rag1 3′-UTR. Arpp21–deficient thymocytes show reduced Rag1 expression, delayed TCR rearrangement and a less diverse TCR repertoire. This phenotype is recapitulated in Rag1 3′-UTR mutant mice harboring a deletion of the Arpp21 response region. These findings show how thymocyte-specific Arpp21 promotes Rag1 expression to enable TCR repertoire diversity until signals from the TCR terminate Arpp21 and Rag1 activities.
- Published
- 2024
- Full Text
- View/download PDF
3. Beta cell and immune cell interactions in autoimmune type 1 diabetes: How they meet and talk to each other
- Author
-
Martin G. Scherm, Rebecca C. Wyatt, Isabelle Serr, David Anz, Sarah J. Richardson, and Carolin Daniel
- Subjects
Type 1 diabetes ,Autoimmunity ,Autoimmune diabetes ,Beta cells ,Immune cells ,Immune Regulation ,Internal medicine ,RC31-1245 - Abstract
Background: The highly complex pathogenesis of Type 1 Diabetes is driven by several immune cell types with both effector and regulatory characteristics, which ultimately ends in the destruction of the insulin-producing beta cells. There are multiple layers of interaction between these immune cell populations and the pancreatic islets. Scope of review: In this review article, we aim to discuss important recent insights into the multiple layers of interaction between immune cell populations and the pancreatic islets. Specifically, we discuss the environment where immune and beta cell interactions occur, the key cell types and molecules involved, and the outcomes of these interactions. Major conclusions: Most of the molecular mechanisms underlying aberrant immune cell activation and impaired immune tolerance remain insufficiently understood, which hinders the development of efficient prevention and treatment strategies. In order to overcome this knowledge gap, a better understanding of the complex interactions of immune cells and beta cells, including both the underlying protective and pathogenic mechanisms is urgently required.
- Published
- 2022
- Full Text
- View/download PDF
4. Antigen-Specific Treg Therapy in Type 1 Diabetes – Challenges and Opportunities
- Author
-
Isabelle Serr, Felix Drost, Benjamin Schubert, and Carolin Daniel
- Subjects
antigen-specific Treg therapy ,autoimmunity ,T1D ,microRNAs ,tissue Tregs ,single-cell multi-omics integration ,Immunologic diseases. Allergy ,RC581-607 - Abstract
Regulatory T cells (Tregs) are key mediators of peripheral self-tolerance and alterations in their frequencies, stability, and function have been linked to autoimmunity. The antigen-specific induction of Tregs is a long-envisioned goal for the treatment of autoimmune diseases given reduced side effects compared to general immunosuppressive therapies. However, the translation of antigen-specific Treg inducing therapies for the treatment or prevention of autoimmune diseases into the clinic remains challenging. In this mini review, we will discuss promising results for antigen-specific Treg therapies in allergy and specific challenges for such therapies in autoimmune diseases, with a focus on type 1 diabetes (T1D). We will furthermore discuss opportunities for antigen-specific Treg therapies in T1D, including combinatorial strategies and tissue-specific Treg targeting. Specifically, we will highlight recent advances in miRNA-targeting as a means to foster Tregs in autoimmunity. Additionally, we will discuss advances and perspectives of computational strategies for the detailed analysis of tissue-specific Tregs on the single-cell level.
- Published
- 2021
- Full Text
- View/download PDF
5. miRNA142-3p targets Tet2 and impairs Treg differentiation and stability in models of type 1 diabetes
- Author
-
Martin G. Scherm, Isabelle Serr, Adam M. Zahm, Jonathan Schug, Saverio Bellusci, Rossella Manfredini, Victoria K. Salb, Katharina Gerlach, Benno Weigmann, Anette-Gabriele Ziegler, Klaus H. Kaestner, and Carolin Daniel
- Subjects
Science - Abstract
miRNA142-3p and Tet2 are separately known to regulate Treg. Here the authors show that miRNA142-3p targets Tet2 and by this opposes Treg differentiation in autoimmune diabetes.
- Published
- 2019
- Full Text
- View/download PDF
6. Short-term cold exposure supports human Treg induction in vivo
- Author
-
Maike Becker, Isabelle Serr, Victoria K. Salb, Verena B. Ott, Laura Mengel, Matthias Blüher, Benno Weigmann, Hans Hauner, Matthias H. Tschöp, and Carolin Daniel
- Subjects
Internal medicine ,RC31-1245 - Abstract
Objective: Obesity and type-2 diabetes (T2D) are metabolic diseases that represent a critical health problem worldwide. Metabolic disease is differentially associated with fat distribution, while visceral white adipose tissue (VAT) is particularly prone to obesity-associated inflammation. Next to their canonical function of immune suppression, regulatory T cells (Tregs) are key in controlling adipose tissue homeostasis. Towards understanding the molecular underpinnings of metabolic disease, we focus on how environmental-metabolic stimuli impinge on the functional interplay between Tregs and adipose tissue. Here, cold exposure or beta3-adrenergic signaling are a promising tool to increase energy expenditure by activating brown adipose tissue, as well as by reducing local inflammation within fat depots by supporting immunosuppressive Tregs. However, in humans, the underlying mechanisms that enable the environmental-immune crosstalk in the periphery and in the respective tissue remain currently unknown. Methods: We used combinatorial approaches of next generation humanized mouse models and in vitro and in vivo experiments together with beta3-adrenergic stimulation to dissect the underlying mechanisms of human Treg induction exposed to environmental stimuli such as cold. To test the translational relevance of our findings, we analyzed samples from the FREECE study in which human subjects were exposed to individualized cooling protocols. Samples were analyzed ex vivo and after in vitro Treg induction using qRT-PCR, immunofluorescence, as well as with multicolor flow cytometry and cell sorting. Results: In vivo application of the beta3-adrenergic receptor agonist mirabegron in humanized mice induced thermogenesis and improved the Treg induction capacity of naïve T cells isolated from these animals. Using samples from the human FREECE study, we demonstrate that a short-term cold stimulus supports human Treg induction in vitro and in vivo. Mechanistically, we identify BORCS6 encoding the Ragulator-interacting protein C17orf59 to be significantly induced in human CD4+ T cells upon short-term cold exposure. Strong mTOR signaling is known to limit successful Treg induction and thus likely by interfering with mTOR activation at lysosomal surfaces, C17orf59 improves the Treg induction capacity of human naïve T cells upon cold exposure. Conclusions: These novel insights into the molecular underpinnings of human Treg induction suggest an important role of Tregs in linking environmental stimuli with adipose tissue function and metabolic diseases. Moreover, these discoveries shed new light on potential approaches towards tailored anti-inflammatory concepts that support human adipose tissue homeostasis by enabling Tregs. Keywords: Regulatory T cell, Human adipose tissue, Beta3-adrenergic stimulation, Mirabegron, Immunometabolism, Humanized mice
- Published
- 2019
- Full Text
- View/download PDF
7. Advances in Human Immune System Mouse Models for Personalized Treg-Based Immunotherapies
- Author
-
Isabelle Serr, Maria Kral, Martin G. Scherm, and Carolin Daniel
- Subjects
HIS mice ,Treg ,cancer immunotherapy ,personalized medicine ,iPSC-derived HSCs ,microRNA ,Immunologic diseases. Allergy ,RC581-607 - Abstract
Immunodeficient mice engrafted with a functional human immune system [Human immune system (HIS) mice] have paved the way to major advances for personalized medicine and translation of immune-based therapies. One prerequisite for advancing personalized medicine is modeling the immune system of individuals or disease groups in a preclinical setting. HIS mice engrafted with peripheral blood mononuclear cells have provided fundamental insights in underlying mechanisms guiding immune activation vs. regulation in several diseases including cancer. However, the development of Graft-vs.-host disease restrains relevant long-term studies in HIS mice. Alternatively, engraftment with hematopoietic stem cells (HSCs) enables mimicking different disease stages, however, low frequencies of HSCs in peripheral blood of adults impede engraftment efficacy. One possibility to overcome those limitations is the use of patient-derived induced pluripotent stem cells (iPSCs) reprogrammed into HSCs, a challenging process which has recently seen major advances. Personalized HIS mice bridge research in mice and human diseases thereby facilitating the translation of immunomodulatory therapies. Regulatory T cells (Tregs) are important mediators of immune suppression and thereby contribute to tumor immune evasion, which has made them a central target for cancer immunotherapies. Importantly, studying Tregs in the human immune system in vivo in HIS mice will help to determine requirements for efficient Treg-targeting. In this review article, we discuss advances on personalized HIS models using reprogrammed iPSCs and review the use of HIS mice to study requirements for efficient targeting of human Tregs for personalized cancer immunotherapies.
- Published
- 2021
- Full Text
- View/download PDF
8. miRNA-Mediated Immune Regulation in Islet Autoimmunity and Type 1 Diabetes
- Author
-
Martin G. Scherm and Carolin Daniel
- Subjects
immune regulation ,islet autoimmunity ,type 1 diabetes ,miRNA ,regulatory T cell ,biomarker ,Diseases of the endocrine glands. Clinical endocrinology ,RC648-665 - Abstract
The important role of microRNAs as major modulators of various physiological processes, including immune regulation and homeostasis, has been increasingly recognized. Consequently, aberrant miRNA expression contributes to the defective regulation of T cell development, differentiation, and function. This can result in immune activation and impaired tolerance mechanisms, which exert a cardinal function for the onset of islet autoimmunity and the progression to T1D. The specific impact of miRNAs for immune regulation and how miRNAs and their downstream targets are involved in the pathogenesis of islet autoimmunity and T1D has been investigated recently. These studies revealed that increased expression of individual miRNAs is involved in several layers of tolerance impairments, such as inefficient Treg induction and Treg instability. The targeted modulation of miRNAs using specific inhibitors, resulting in improved immune homeostasis, as well as improved methods for the targeting of miRNAs, suggest that miRNAs, especially in T cells, are a promising target for the reestablishment of immune tolerance.
- Published
- 2020
- Full Text
- View/download PDF
9. The role of T cell miRNAs for regulatory T cell induction in islet autoimmunity
- Author
-
Martin G. Scherm, Isabelle Serr, Klaus H. Kaestner, and Carolin Daniel
- Subjects
Internal medicine ,RC31-1245 - Abstract
Background: microRNAs (miRNAs) have emerged as critical contributors to immune regulation and homeostasis, and their dysregulation is involved in the aberrant differentiation and function of T cell subsets. In type 1 diabetes (T1D), the clinically overt disease is preceded by a presymptomatic phase which is marked by the presence of islet autoantibodies while the individual is still normoglycemic. Recent analyses revealed impaired regulatory T (Treg) cell induction from naive CD4+ T cells during this early phase of autoimmunity. Scope of the review: In this review article, we aim to discuss important recent insights into miRNA regulation of immune homeostasis and activation. Specifically, we highlight the role of miRNAs as biomarkers in autoimmunity and T1D as well as the contribution of specific miRNAs and their downstream pathways to the onset and progression of islet immunity. Furthermore, we focus on critical next steps required to establish miRNAs as biomarkers to predict disease onset and progression and as novel targets of future prevention and treatment strategies to control autoimmunity. Major conclusions: Several recent studies have provided considerable insight into the miRNA regulation of immune homeostasis and how dysregulated miRNAs contribute to onset and progression of islet autoimmunity. Specifically, high levels of individual miRNAs such as miR92a and miR181a are involved in impaired Treg induction during the onset of islet autoimmunity, thereby contributing to disease pathogenesis. The recent advancements in the field suggest miRNAs as potential biomarkers for islet autoimmunity and their direct targeting, especially in a T cell-specific manner, could contribute to the reestablishment of immune homeostasis and ultimately interfere with the onset of islet autoimmunity. Keywords: Immune regulation, Islet autoimmunity, Type 1 diabetes, Regulatory T cell, miRNA, Biomarker
- Published
- 2019
- Full Text
- View/download PDF
10. Type 1 diabetes vaccine candidates promote human Foxp3+Treg induction in humanized mice
- Author
-
Isabelle Serr, Rainer W. Fürst, Peter Achenbach, Martin G. Scherm, Füsun Gökmen, Florian Haupt, Eva-Maria Sedlmeier, Annette Knopff, Leonard Shultz, Richard A. Willis, Anette-Gabriele Ziegler, and Carolin Daniel
- Subjects
Science - Abstract
Type 1 diabetes is associated with the loss of self-tolerance to the insulin-producing β-cells in the pancreas. Here the authors show that vaccination with insulin mimetopes can induce human insulin-specific regulatory T cells to mediate tolerance in a humanized mouse model.
- Published
- 2016
- Full Text
- View/download PDF
11. Regulation of T Follicular Helper Cells in Islet Autoimmunity
- Author
-
Isabelle Serr and Carolin Daniel
- Subjects
T follicular helper cells ,islet autoimmunity ,microRNA92a ,krueppel-like factor 2 ,type 1 diabetes ,Immunologic diseases. Allergy ,RC581-607 - Abstract
T follicular helper (TFH) cells are an integral part of humoral immunity by providing help to B cells to produce high-affinity antibodies. The TFH precursor compartment circulates in the blood and TFH cell dysregulation is implied in various autoimmune diseases including type 1 diabetes (T1D). Symptomatic T1D is preceded by a preclinical phase (indicated by the presence of islet autoantibodies) with a highly variable progression time to the symptomatic disease. This heterogeneity points toward differences in immune activation in children with a fast versus slow progressor phenotype. In the context of T1D, previous studies on TFH cells have mainly focused on the clinically active state of the disease. In this review article, we aim to specifically discuss recent insights on TFH cells in human islet autoimmunity before the onset of symptomatic T1D. Furthermore, we will highlight advances in the field of TFH differentiation and function during human islet autoimmunity. Specifically, we will focus on the regulation of TFH cells by microRNAs (miRNAs), as well as on the potential use of miRNAs as biomarkers to predict disease progression time and as future drug targets to interfere with autoimmune activation.
- Published
- 2018
- Full Text
- View/download PDF
12. Supplementary Figure 6 from Transcription Factor NFATc2 Controls the Emergence of Colon Cancer Associated with IL-6–Dependent Colitis
- Author
-
Benno Weigmann, Markus F. Neurath, Stefan Rose-John, Raja Atreya, Thomas Gerlach, Alexei Nikolaev, Hans A. Lehr, Carolin Daniel, and Katharina Gerlach
- Abstract
PDF file - 112K, expression of cox-2 and VEGF
- Published
- 2023
- Full Text
- View/download PDF
13. Supplementary Figure 1 from Transcription Factor NFATc2 Controls the Emergence of Colon Cancer Associated with IL-6–Dependent Colitis
- Author
-
Benno Weigmann, Markus F. Neurath, Stefan Rose-John, Raja Atreya, Thomas Gerlach, Alexei Nikolaev, Hans A. Lehr, Carolin Daniel, and Katharina Gerlach
- Abstract
PDF file - 547K, DSS Colitis
- Published
- 2023
- Full Text
- View/download PDF
14. Supplementary Table 1 from Transcription Factor NFATc2 Controls the Emergence of Colon Cancer Associated with IL-6–Dependent Colitis
- Author
-
Benno Weigmann, Markus F. Neurath, Stefan Rose-John, Raja Atreya, Thomas Gerlach, Alexei Nikolaev, Hans A. Lehr, Carolin Daniel, and Katharina Gerlach
- Abstract
PDF file - 419K, table of patient data for supplementary figures
- Published
- 2023
- Full Text
- View/download PDF
15. Supplementary Figure Legends 1-7, Methods from Transcription Factor NFATc2 Controls the Emergence of Colon Cancer Associated with IL-6–Dependent Colitis
- Author
-
Benno Weigmann, Markus F. Neurath, Stefan Rose-John, Raja Atreya, Thomas Gerlach, Alexei Nikolaev, Hans A. Lehr, Carolin Daniel, and Katharina Gerlach
- Abstract
PDF file - 106K
- Published
- 2023
- Full Text
- View/download PDF
16. Supplementary Figure 3 from Transcription Factor NFATc2 Controls the Emergence of Colon Cancer Associated with IL-6–Dependent Colitis
- Author
-
Benno Weigmann, Markus F. Neurath, Stefan Rose-John, Raja Atreya, Thomas Gerlach, Alexei Nikolaev, Hans A. Lehr, Carolin Daniel, and Katharina Gerlach
- Abstract
PDF file - 559K, expression of IL-11 and IL-22
- Published
- 2023
- Full Text
- View/download PDF
17. Supplementary Figure 7 from Transcription Factor NFATc2 Controls the Emergence of Colon Cancer Associated with IL-6–Dependent Colitis
- Author
-
Benno Weigmann, Markus F. Neurath, Stefan Rose-John, Raja Atreya, Thomas Gerlach, Alexei Nikolaev, Hans A. Lehr, Carolin Daniel, and Katharina Gerlach
- Abstract
PDF file - 330K, proliferation staining KI-67 in cryosection
- Published
- 2023
- Full Text
- View/download PDF
18. Supplementary Figure 4 from Transcription Factor NFATc2 Controls the Emergence of Colon Cancer Associated with IL-6–Dependent Colitis
- Author
-
Benno Weigmann, Markus F. Neurath, Stefan Rose-John, Raja Atreya, Thomas Gerlach, Alexei Nikolaev, Hans A. Lehr, Carolin Daniel, and Katharina Gerlach
- Abstract
PDF file - 547K, cytokineproduction of spleen cells and LPMC
- Published
- 2023
- Full Text
- View/download PDF
19. Supplementary Figure 2 from Transcription Factor NFATc2 Controls the Emergence of Colon Cancer Associated with IL-6–Dependent Colitis
- Author
-
Benno Weigmann, Markus F. Neurath, Stefan Rose-John, Raja Atreya, Thomas Gerlach, Alexei Nikolaev, Hans A. Lehr, Carolin Daniel, and Katharina Gerlach
- Abstract
PDF file - 589K, expresion of IL-1b and IFN-g
- Published
- 2023
- Full Text
- View/download PDF
20. Supplementary Figure 5 from Transcription Factor NFATc2 Controls the Emergence of Colon Cancer Associated with IL-6–Dependent Colitis
- Author
-
Benno Weigmann, Markus F. Neurath, Stefan Rose-John, Raja Atreya, Thomas Gerlach, Alexei Nikolaev, Hans A. Lehr, Carolin Daniel, and Katharina Gerlach
- Abstract
PDF file - 660K, staining of EpCAM and IL-6 in cytospin preparation
- Published
- 2023
- Full Text
- View/download PDF
21. Droplet-based forward genetic screening of astrocyte–microglia cross-talk
- Author
-
Michael A. Wheeler, Iain C. Clark, Hong-Gyun Lee, Zhaorong Li, Mathias Linnerbauer, Joseph M. Rone, Manon Blain, Camilo Faust Akl, Gavin Piester, Federico Giovannoni, Marc Charabati, Joon-Hyuk Lee, Yoon-Chul Kye, Joshua Choi, Liliana M. Sanmarco, Lena Srun, Elizabeth N. Chung, Lucas E. Flausino, Brian M. Andersen, Veit Rothhammer, Hiroshi Yano, Tomer Illouz, Stephanie E. J. Zandee, Carolin Daniel, David Artis, Marco Prinz, Adam R. Abate, Vijay K. Kuchroo, Jack P. Antel, Alexandre Prat, and Francisco J. Quintana
- Subjects
Multidisciplinary ,Article - Abstract
Cell–cell interactions in the central nervous system play important roles in neurologic diseases. However, little is known about the specific molecular pathways involved, and methods for their systematic identification are limited. Here, we developed a forward genetic screening platform that combines CRISPR-Cas9 perturbations, cell coculture in picoliter droplets, and microfluidic-based fluorescence-activated droplet sorting to identify mechanisms of cell–cell communication. We used SPEAC-seq (systematic perturbation of encapsulated associated cells followed by sequencing), in combination with in vivo genetic perturbations, to identify microglia-produced amphiregulin as a suppressor of disease-promoting astrocyte responses in multiple sclerosis preclinical models and clinical samples. Thus, SPEAC-seq enables the high-throughput systematic identification of cell–cell communication mechanisms.
- Published
- 2023
22. Autoimmunerkrankung Typ-1-Diabetes
- Author
-
Carolin Daniel and Martin G. Scherm
- Subjects
0303 health sciences ,03 medical and health sciences ,0302 clinical medicine ,Endocrinology, Diabetes and Metabolism ,Internal Medicine ,030209 endocrinology & metabolism ,030304 developmental biology - Abstract
ZUSAMMENFASSUNGDie umfassende Erforschung der Immunologie des Typ-1-Diabetes kann entscheidend zu unserem Verständnis der Krankheit beitragen. Hierbei ist insbesondere auch die Identifizierung der zugrundeliegenden Signalwege, die zur fehlerhaften Immuntoleranz sowie der Aktivierung und dem Fortschreiten der Inselautoimmunität beitragen, von entscheidender Bedeutung für die Entwicklung zukünftiger Interventionsstrategien, um die Entstehung von Typ-1-Diabetes zu verlangsamen oder sogar ganz zu verhindern 1. So bieten kürzlich identifizierte miRNAs, die zur Entstehung von Inselautoimmunität beitragen, einen vielversprechenden Ansatz, um Treg-vermittelte Toleranzdefekte mittels gezielter miRNA-Modulation zu reduzieren.
- Published
- 2021
- Full Text
- View/download PDF
23. Mechanismen der Immuntoleranz bei Typ-1-Diabetes im Kindesalter – Ferdinand-Bertram-Preis 2021 – eine Kurzübersicht der Preisträgerin Carolin Daniel
- Author
-
Carolin Daniel and Martin Scherm
- Subjects
Endocrinology, Diabetes and Metabolism - Published
- 2021
- Full Text
- View/download PDF
24. Aging Aggravates Cachexia in Tumor-Bearing Mice
- Author
-
Julia Geppert, Alina Walth, Raúl Terrón Expósito, Doris Kaltenecker, Pauline Morigny, Juliano Machado, Maike Becker, Estefania Simoes, Joanna Lima, Carolin Daniel, Mauricio Berriel Diaz, Stephan Herzig, Marilia Seelaender, and Maria Rohm
- Subjects
Cancer Research ,Oncology ,aging ,Aging ,Cachexia ,Cancer ,Mouse Models ,cachexia ,cancer ,mouse models ,Neoplasms. Tumors. Oncology. Including cancer and carcinogens ,RC254-282 ,Article ,ddc - Abstract
Simple Summary Cachexia is a deadly disease that accompanies many different types of cancers. Animal studies on cachexia have so far mostly been conducted using young mice, while cancer in humans is a disease of high age. Mouse models used to date may therefore not be suitable to study cachexia with relevance to patients. By comparing young and old mice of three different strains and two different tumor types, we here show that the age of mice has a substantial effect on cachexia progression (specifically body weight, tissue weight, fiber size, molecular markers) that is dependent on the mouse strain studied. This is independent of glucose tolerance. The cachexia markers IL6 and GDF15 differ between ages in both mice and patients. Future studies on cachexia should consider the age and strain of mice. Abstract Background: Cancer is primarily a disease of high age in humans, yet most mouse studies on cancer cachexia are conducted using young adolescent mice. Given that metabolism and muscle function change with age, we hypothesized that aging may affect cachexia progression in mouse models. Methods: We compare tumor and cachexia development in young and old mice of three different strains (C57BL/6J, C57BL/6N, BALB/c) and with two different tumor cell lines (Lewis Lung Cancer, Colon26). Tumor size, body and organ weights, fiber cross-sectional area, circulating cachexia biomarkers, and molecular markers of muscle atrophy and adipose tissue wasting are shown. We correlate inflammatory markers and body weight dependent on age in patients with cancer. Results: We note fundamental differences between mouse strains. Aging aggravates weight loss in LLC-injected C57BL/6J mice, drives it in C57BL/6N mice, and does not influence weight loss in C26-injected BALB/c mice. Glucose tolerance is unchanged in cachectic young and old mice. The stress marker GDF15 is elevated in cachectic BALB/c mice independent of age and increased in old C57BL/6N and J mice. Inflammatory markers correlate significantly with weight loss only in young mice and patients. Conclusions: Aging affects cachexia development and progression in mice in a strain-dependent manner and influences the inflammatory profile in both mice and patients. Age is an important factor to consider for future cachexia studies.
- Published
- 2021
- Full Text
- View/download PDF
25. The role of T cell miRNAs for regulatory T cell induction in islet autoimmunity
- Author
-
Isabelle Serr, Klaus H. Kaestner, Martin G. Scherm, and Carolin Daniel
- Subjects
0301 basic medicine ,lcsh:Internal medicine ,Immune regulation ,Regulatory T cell ,T cell ,Autoimmunity ,030209 endocrinology & metabolism ,Review ,Disease ,Biology ,medicine.disease_cause ,T-Lymphocytes, Regulatory ,Islets of Langerhans ,03 medical and health sciences ,0302 clinical medicine ,Immunity ,Biomarker ,Immune Regulation ,Islet Autoimmunity ,Regulatory T Cell ,Type 1 Diabetes ,Mirna ,microRNA ,medicine ,Animals ,Humans ,lcsh:RC31-1245 ,Molecular Biology ,Islet autoimmunity ,miRNA ,geography ,geography.geographical_feature_category ,Cell Biology ,Islet ,Review article ,MicroRNAs ,Diabetes Mellitus, Type 1 ,Type 1 diabetes ,030104 developmental biology ,medicine.anatomical_structure ,Immunology - Abstract
Background: microRNAs (miRNAs) have emerged as critical contributors to immune regulation and homeostasis, and their dysregulation is involved in the aberrant differentiation and function of T cell subsets. In type 1 diabetes (T1D), the clinically overt disease is preceded by a presymptomatic phase which is marked by the presence of islet autoantibodies while the individual is still normoglycemic. Recent analyses revealed impaired regulatory T (Treg) cell induction from naive CD4+ T cells during this early phase of autoimmunity. Scope of the review: In this review article, we aim to discuss important recent insights into miRNA regulation of immune homeostasis and activation. Specifically, we highlight the role of miRNAs as biomarkers in autoimmunity and T1D as well as the contribution of specific miRNAs and their downstream pathways to the onset and progression of islet immunity. Furthermore, we focus on critical next steps required to establish miRNAs as biomarkers to predict disease onset and progression and as novel targets of future prevention and treatment strategies to control autoimmunity. Major conclusions: Several recent studies have provided considerable insight into the miRNA regulation of immune homeostasis and how dysregulated miRNAs contribute to onset and progression of islet autoimmunity. Specifically, high levels of individual miRNAs such as miR92a and miR181a are involved in impaired Treg induction during the onset of islet autoimmunity, thereby contributing to disease pathogenesis. The recent advancements in the field suggest miRNAs as potential biomarkers for islet autoimmunity and their direct targeting, especially in a T cell-specific manner, could contribute to the reestablishment of immune homeostasis and ultimately interfere with the onset of islet autoimmunity. Keywords: Immune regulation, Islet autoimmunity, Type 1 diabetes, Regulatory T cell, miRNA, Biomarker
- Published
- 2019
26. Type 1 diabetes vaccine candidates promote human Foxp3+Treg induction in humanized mice
- Author
-
Leonard Shultz, Anette Gabriele Ziegler, Carolin Daniel, Isabelle Serr, Peter Achenbach, F Haupt, A. Knopff, Martin G. Scherm, Eva Maria Sedlmeier, Füsun Gökmen, Richard A. Willis, and Rainer W. Fürst
- Subjects
0301 basic medicine ,Male ,General Physics and Astronomy ,Autoimmunity ,medicine.disease_cause ,Lymphocyte Activation ,Autoantigens ,T-Lymphocytes, Regulatory ,Immune tolerance ,Mice ,Medicine ,Insulin ,CTLA-4 Antigen ,Receptors, Immunologic ,Child ,Vaccines ,Multidisciplinary ,geography.geographical_feature_category ,Effector ,Hematopoietic Stem Cell Transplantation ,FOXP3 ,hemic and immune systems ,Forkhead Transcription Factors ,Islet ,3. Good health ,ddc ,Vaccination ,Haematopoiesis ,Self Tolerance ,Child, Preschool ,Female ,Adult ,Adolescent ,Science ,chemical and pharmacologic phenomena ,Mice, Transgenic ,General Biochemistry, Genetics and Molecular Biology ,Article ,03 medical and health sciences ,Young Adult ,TIGIT ,HLA-DQ Antigens ,Immune Tolerance ,Animals ,Humans ,geography ,business.industry ,Interleukin-2 Receptor alpha Subunit ,General Chemistry ,030104 developmental biology ,Diabetes Mellitus, Type 1 ,Gene Expression Regulation ,Immunology ,business - Abstract
Immune tolerance is executed partly by Foxp3+regulatory T (Treg) cells, which suppress autoreactive T cells. In autoimmune type 1 diabetes (T1D) impaired tolerance promotes destruction of insulin-producing β-cells. The development of autoantigen-specific vaccination strategies for Foxp3+Treg-induction and prevention of islet autoimmunity in patients is still in its infancy. Here, using human haematopoietic stem cell-engrafted NSG-HLA-DQ8 transgenic mice, we provide direct evidence for human autoantigen-specific Foxp3+Treg-induction in vivo. We identify HLA-DQ8-restricted insulin-specific CD4+T cells and demonstrate efficient human insulin-specific Foxp3+Treg-induction upon subimmunogenic vaccination with strong agonistic insulin mimetopes in vivo. Induced human Tregs are stable, show increased expression of Treg signature genes such as Foxp3, CTLA4, IL-2Rα and TIGIT and can efficiently suppress effector T cells. Such Foxp3+Treg-induction does not trigger any effector T cells. These T1D vaccine candidates could therefore represent an expedient improvement in the challenge to induce human Foxp3+Tregs and to develop novel precision medicines for prevention of islet autoimmunity in children at risk of T1D., Type 1 diabetes is associated with the loss of self-tolerance to the insulin-producing β-cells in the pancreas. Here the authors show that vaccination with insulin mimetopes can induce human insulin-specific regulatory T cells to mediate tolerance in a humanized mouse model.
- Published
- 2016
27. Immunometabolism and atherosclerosis: perspectives and clinical significance: a position paper from the Working Group on Atherosclerosis and Vascular Biology of the European Society of Cardiology
- Author
-
Peter Libby, Luke A. J. O'Neill, Christian Weber, Ingrid E. Dumitriu, Carolin Daniel, Jan Van den Bossche, Daniel F. J. Ketelhuth, Imo E. Hoefer, Christoph J. Binder, Esther Lutgens, Magnus Bäck, Paul C. Evans, Molecular cell biology and Immunology, Amsterdam Gastroenterology Endocrinology Metabolism, Medical Biochemistry, ACS - Atherosclerosis & ischemic syndromes, AII - Inflammatory diseases, and AGEM - Amsterdam Gastroenterology Endocrinology Metabolism
- Subjects
0301 basic medicine ,Physiology ,MACROPHAGE IMMUNOMETABOLISM ,T-Lymphocytes ,Macrophages/immunology ,Disease ,030204 cardiovascular system & hematology ,Bioinformatics ,Monocytes ,0302 clinical medicine ,IMMUNE-RESPONSE ,INDUCTION ,Vascular biology ,Position Paper from European Society of Cardiology Working Group ,Arteries ,Inflammation/immunology ,Plaque, Atherosclerotic ,3. Good health ,TRIMETHYLAMINE N-OXIDE ,medicine.symptom ,Inflammation Mediators ,Cardiology and Cardiovascular Medicine ,Atherosclerosis/immunology ,medicine.drug ,Signal Transduction ,Consensus ,IMAGING ATHEROSCLEROSIS ,Inflammation ,METABOLISM ,DENDRITIC CELLS ,Immunomodulation ,03 medical and health sciences ,Immune system ,Immunity ,Vascular ,Physiology (medical) ,medicine ,Animals ,Humans ,Inflammation Mediators/immunology ,Clinical significance ,REGULATORY T-CELLS ,business.industry ,Macrophages ,Arteries/immunology ,GLUTAMINE UPTAKE ,Monocytes/immunology ,T-Lymphocytes/immunology ,Atherosclerosis ,Canakinumab ,030104 developmental biology ,Immune System ,Position paper ,Energy Metabolism/immunology ,Immune System/immunology ,business ,Energy Metabolism - Abstract
Inflammation is an important driver of atherosclerosis, and the favourable outcomes of the Canakinumab Anti-inflammatory Thrombosis Outcome Study (CANTOS) trial revealed the large potential of anti-inflammatory drugs for the treatment of cardiovascular disease, especially in patients with a pro-inflammatory constitution. However, the complex immune reactions driving inflammation in the vascular wall in response to an atherosclerotic microenvironment are still being unravelled. Novel insights into the cellular processes driving immunity and inflammation revealed that alterations in intracellular metabolic pathways are strong drivers of survival, growth, and function of immune cells. Therefore, this position paper presents a brief overview of the recent developments in the immunometabolism field, focusing on its role in atherosclerosis. We will also highlight the potential impact of immunometabolic markers and targets in clinical cardiovascular medicine.
- Published
- 2018
- Full Text
- View/download PDF
28. A miRNA181a/NFAT5 axis links impaired T cell tolerance induction with autoimmune type 1 diabetes
- Author
-
Ari Waisman, Maike Becker, James H. Segars, Richard A. Willis, Markus Hippich, Anette Gabriele Ziegler, Martin G. Scherm, Katharina Gerlach, Adam M. Zahm, Benedikt Kirchner, Stefanie Kälin, Victoria K. Flynn, Jonathan Schug, Benno Weigmann, Christoph Küper, Claus-Michael Lehr, Nicole Liebsch, Bettina Haase, Klaus H. Kaestner, Alexei Nikolaev, Isabelle Serr, Ralf Palmisano, Brigitta Loretz, Peter Achenbach, Wan-Uk Kim, Carolin Daniel, Melanie Spornraft, and HIPS, Helmholtz-Institute für pharmazeutische Forschung Saarland, Universitätscampus E8.1, 66123 Saarbrücken, Germany.
- Subjects
CD4-Positive T-Lymphocytes ,0301 basic medicine ,Regulatory T cell ,Biology ,medicine.disease_cause ,Autoimmunity ,Mice ,03 medical and health sciences ,NFAT5 ,microRNA ,Immunogenetics ,medicine ,Animals ,Humans ,PI3K/AKT/mTOR pathway ,geography ,geography.geographical_feature_category ,NFATC Transcription Factors ,Antagomirs ,FOXP3 ,Forkhead Transcription Factors ,General Medicine ,Islet ,Mice, Mutant Strains ,MicroRNAs ,Tolerance induction ,Diabetes Mellitus, Type 1 ,030104 developmental biology ,medicine.anatomical_structure ,Cancer research ,Female - Abstract
Molecular checkpoints that trigger the onset of islet autoimmunity or progression to human type 1 diabetes (T1D) are incompletely understood. Using T cells from children at an early stage of islet autoimmunity without clinical T1D, we find that a microRNA181a (miRNA181a)-mediated increase in signal strength of stimulation and costimulation links nuclear factor of activated T cells 5 (NFAT5) with impaired tolerance induction and autoimmune activation. We show that enhancing miRNA181a activity increases NFAT5 expression while inhibiting FOXP3+ regulatory T cell (Treg) induction in vitro. Accordingly, Treg induction is improved using T cells from NFAT5 knockout (NFAT5ko) animals, whereas altering miRNA181a activity does not affect Treg induction in NFAT5ko T cells. Moreover, high costimulatory signals result in phosphoinositide 3-kinase (PI3K)-mediated NFAT5, which interferes with FoxP3+ Treg induction. Blocking miRNA181a or NFAT5 increases Treg induction in murine and humanized models and reduces murine islet autoimmunity in vivo. These findings suggest targeting miRNA181a and/or NFAT5 signaling for the development of innovative personalized medicines to limit islet autoimmunity.
- Published
- 2018
29. Adipose-tissue regulatory T cells: Critical players in adipose-immune crosstalk
- Author
-
Maike Becker, Megan K. Levings, and Carolin Daniel
- Subjects
0301 basic medicine ,Immunology ,Adipose tissue ,Inflammation ,chemical and pharmacologic phenomena ,Type 2 diabetes ,Biology ,T-Lymphocytes, Regulatory ,Immune tolerance ,03 medical and health sciences ,Mice ,0302 clinical medicine ,Immune system ,Adipose Tissue Function ,Foxp3 ,Il-33 ,Immune-adipose Crosstalk ,Regulatory T Cells (tregs) ,T Cell Tolerance ,Type 2 Diabetes ,medicine ,Immunology and Allergy ,Animals ,Humans ,Obesity ,FOXP3 ,Receptor Cross-Talk ,medicine.disease ,Interleukin 33 ,Crosstalk (biology) ,030104 developmental biology ,Adipose Tissue ,Diabetes Mellitus, Type 2 ,medicine.symptom ,030215 immunology - Abstract
Obesity and type-2 diabetes (T2D) are associated with metabolic defects and inflammatory processes in fat depots. FoxP3 + regulatory T cells (Tregs) control immune tolerance, and have an important role in controlling tissue-specific inflammation. In this mini-review we will discuss current insights into how cross-talk between T cells and adipose tissue shapes the inflammatory environment in obesity-associated metabolic diseases, focusing on the role of CD4 + T cells and Tregs. We will also highlight potential opportunities for how the immunoregulatory properties of Tregs could be harnessed to control inflammation in obesity and T2D and emphasize the critical need for more research on humans to establish mechanisms that are conserved in both mice and humans.
- Published
- 2017
30. Treg Vaccination in Autoimmune Type 1 Diabetes
- Author
-
Isabelle Serr, Benno Weigmann, Carolin Daniel, and Randi Kristina Franke
- Subjects
CD4-Positive T-Lymphocytes ,medicine.disease_cause ,T-Lymphocytes, Regulatory ,Autoimmunity ,Mice ,Diabetes mellitus ,medicine ,Animals ,Humans ,Pharmacology (medical) ,Antigens ,Receptor ,Pharmacology ,Type 1 diabetes ,biology ,business.industry ,Vaccination ,FOXP3 ,Forkhead Transcription Factors ,General Medicine ,medicine.disease ,Molecular medicine ,Disease Models, Animal ,Diabetes Mellitus, Type 1 ,Self Tolerance ,medicine.anatomical_structure ,Immunology ,biology.protein ,Antibody ,Pancreas ,business ,Biotechnology - Abstract
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.
- Published
- 2013
- Full Text
- View/download PDF
31. Nuclear factor of activated T cells-A transcription factor family as critical regulator in lung and colon cancer
- Author
-
Benno Weigmann, Carolin Daniel, Katharina Gerlach, Martin Väth, and Markus F. Neurath
- Subjects
Cancer Research ,Colorectal cancer ,T cell ,Regulator ,Cancer ,NFAT ,Biology ,medicine.disease ,NFATC Transcription Factors ,medicine.anatomical_structure ,Immune system ,Oncology ,Immunology ,cardiovascular system ,medicine ,Cancer research ,Transcription factor - Abstract
Nuclear factor of activated T cells (NFAT) was first identified as a transcription factor which is activated upon T cell stimulation. Subsequent studies uncovered that a whole family of individual NFAT proteins exists with pleiotropic functions not only in immune but also in nonimmune cells. However, dysregulation of NFAT thereby favors malignant growth and cancer. Summarizing the recent advances in understanding how individual NFAT factors regulate the immune system, this review gives new insights into the critical role of NFAT in cancer development with special focus on inflammation-associated colorectal cancer.
- Published
- 2013
- Full Text
- View/download PDF
32. Follicular Helper T Cells in Autoimmunity
- Author
-
Carolin Daniel, Verena B. Ott, and Martin G. Scherm
- Subjects
0301 basic medicine ,Endocrinology, Diabetes and Metabolism ,Cellular differentiation ,Autoimmunity ,Autoantibodies ,Biomarker ,Tfh Cells ,Type 1 Diabetes ,medicine.disease_cause ,03 medical and health sciences ,Immune system ,Internal Medicine ,medicine ,Homeostasis ,Humans ,B-cell lymphoma ,business.industry ,Autoantibody ,Germinal center ,Cell Differentiation ,T-Lymphocytes, Helper-Inducer ,medicine.disease ,BCL6 ,030104 developmental biology ,Diabetes Mellitus, Type 1 ,Immunology ,Humoral immunity ,business ,Biomarkers - Abstract
The development of multiple disease-relevant autoantibodies is a hallmark of autoimmune diseases. In autoimmune type 1 diabetes (T1D), a variable time frame of autoimmunity precedes the clinically overt disease. The relevance of T follicular helper (TFH) cells for the immune system is increasingly recognized. Their pivotal contribution to antibody production by providing help to germinal center (GC) B cells facilitates the development of a long-lived humoral immunity. Their complex differentiation process, involving various stages and factors like B cell lymphoma 6 (Bcl6), is strictly controlled, as anomalous regulation of TFH cells is connected with immunopathologies. While the adverse effects of a TFH cell-related insufficient humoral immunity are obvious, the role of increased TFH frequencies in autoimmune diseases like T1D is currently highlighted. High levels of autoantigen trigger an excessive induction of TFH cells, consequently resulting in the production of autoantibodies. Therefore, TFH cells might provide promising approaches for novel therapeutic strategies.
- Published
- 2016
33. miRNA92a targets KLF2 and the phosphatase PTEN signaling to promote human T follicular helper precursors in T1D islet autoimmunity
- Author
-
Benedikt Kirchner, Carolin Daniel, Stefanie Kälin, Isabelle Serr, Nicole Kunschke, Michael W. Pfaffl, Anette-G. Ziegler, Ari Waisman, Martin G. Scherm, Stephanie Zillmer, Benno Weigmann, Richard A. Willis, Füsun Gökmen, Alexei Nikolaev, Melanie Bunk, Bettina Haase, Brigitta Loretz, Rainer W. Fürst, Verena B. Ott, Claus-Michael Lehr, and Helmholtz-Institut für pharmazeutische Forschung Saarland, Universitätscampus E8.1, 66123 Saarbrücken, Germany.
- Subjects
0301 basic medicine ,Male ,Receptors, CXCR5 ,endocrine system ,Adolescent ,Population ,Primary Cell Culture ,Kruppel-Like Transcription Factors ,Autoimmunity ,Mice, Transgenic ,Nod ,Biology ,medicine.disease_cause ,CXCR5 ,03 medical and health sciences ,Islets of Langerhans ,Mice ,Phosphatidylinositol 3-Kinases ,0302 clinical medicine ,Mice, Inbred NOD ,medicine ,Animals ,Humans ,IL-2 receptor ,Klf2 ,Pten-pi3k Signaling ,T Follicular Helper Cells ,Mirna92a ,Type 1 Diabetes ,education ,Child ,PI3K/AKT/mTOR pathway ,NOD mice ,Autoantibodies ,geography ,education.field_of_study ,Multidisciplinary ,geography.geographical_feature_category ,Forkhead Box Protein O1 ,PTEN Phosphohydrolase ,Antagomirs ,T-Lymphocytes, Helper-Inducer ,Islet ,MicroRNAs ,030104 developmental biology ,Diabetes Mellitus, Type 1 ,Gene Expression Regulation ,Immunology ,Cancer research ,Female ,030215 immunology ,Signal Transduction - Abstract
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.
- Published
- 2016
34. Transcription Factor NFATc2 Controls the Emergence of Colon Cancer Associated with IL-6–Dependent Colitis
- Author
-
Stefan Rose-John, Markus F. Neurath, Alexei Nikolaev, Carolin Daniel, Katharina Gerlach, Thomas Gerlach, Hans A. Lehr, Raja Atreya, and Benno Weigmann
- Subjects
CD4-Positive T-Lymphocytes ,Cancer Research ,Colorectal cancer ,Apoptosis ,Proinflammatory cytokine ,Mice ,medicine ,Animals ,Humans ,Colitis ,Transcription factor ,Mice, Knockout ,Mice, Inbred BALB C ,NFATC Transcription Factors ,Interleukin-6 ,business.industry ,Interleukin ,NFAT ,Inflammatory Bowel Diseases ,medicine.disease ,Gene Expression Regulation, Neoplastic ,Cell Transformation, Neoplastic ,Oncology ,Tumor progression ,Colonic Neoplasms ,Immunology ,business - Abstract
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. Cancer Res; 72(17); 4340–50. ©2012 AACR.
- Published
- 2012
- Full Text
- View/download PDF
35. Treg Vaccination with a Strong-Agonistic Insulin Mimetope
- Author
-
Benno Weigmann and Carolin Daniel
- Subjects
Endocrinology, Diabetes and Metabolism ,Insulin ,medicine.medical_treatment ,T-cell receptor ,FOXP3 ,Autoimmunity ,Forkhead Transcription Factors ,hemic and immune systems ,chemical and pharmacologic phenomena ,Inflammation ,Biology ,medicine.disease_cause ,T-Lymphocytes, Regulatory ,Vaccination ,Diabetes Mellitus, Type 1 ,Antigen ,Immunology ,Internal Medicine ,medicine ,Animals ,Humans ,medicine.symptom ,Transcription factor - Abstract
Foxp3(+) regulatory T (Treg) cells serve as a vital mechanism of negative regulation to maintain immunological self-tolerance thereby suppressing immune-mediated inflammation. The identification of the transcription factor Foxp3 as the specification factor for the Treg cell lineage facilitated our understanding in the biology of Treg generation and function. In the past, we carefully studied the extrathymic conversion of naive CD4(+) T cells into Foxp3(+) expressing Treg cells and found that this process is most efficient upon subimmunogenic supply of strong-agonistic T cell receptor (TCR) ligands avoiding activation of antigen-presenting and T cells. In contrast, weak-agonistic antigens fail to efficiently induce stable Foxp3(+) Treg cells irrespective of the applied dose. Here, we discuss the specific requirements for the establishment of Treg vaccination protocols to interfere with autoimmunity such as Type 1 diabetes.
- Published
- 2012
- Full Text
- View/download PDF
36. Immunotherapy in Autoimmune Type 1 Diabetes
- Author
-
Randi Kristina Franke, Benno Weigmann, and Carolin Daniel
- Subjects
Endocrinology, Diabetes and Metabolism ,medicine.medical_treatment ,Review ,Antigen ,Autoimmunity ,Conversion ,Foxp3 ,Immunotherapy ,Insulin ,Mimetope ,Regulatory T cells ,Suppression ,Tolerance ,Type 1 diabetes ,Disease ,medicine.disease_cause ,Endocrinology ,Immunity ,Insulin-Secreting Cells ,Cyclosporin a ,Internal Medicine ,medicine ,Animals ,Humans ,Autoimmune disease ,business.industry ,FOXP3 ,medicine.disease ,Disease Models, Animal ,Diabetes Mellitus, Type 1 ,Immunology ,business - Abstract
Type 1 diabetes (T1D) is a chronic autoimmune disease affecting millions of people worldwide. The disease is characterized by the loss of self-tolerance to the insulin-producing β-cells in the pancreas, the destruction of β-cells, and finally the development of chronic hyperglycemia at diagnosis of T1D. Its incidence and prevalence are rising dramatically, highlighting the need for immunotherapeutic strategies able to prevent or treat the disease in a safe and specific manner. Immunotherapeutic strategies are being developed, and aim to restore immunological self-tolerance, thereby limiting unwanted immunity and β-cell destruction. Foxp3+ regulatory T (Treg) cells exert essential functions to maintain and restore immunological self-tolerance. The identification of the transcription factor Foxp3 as the specification factor for the Treg cell lineage facilitated our understanding in the biology of Treg generation and function. This review highlights the current understanding of immunotherapeutic approaches as preventative and curative measures for autoimmune T1D. It includes an overview on early immunointervention studies, which made use of general immunosuppressive agents such as cyclosporin A, followed by a discussion on newly emerging clinical trials. Besides non-antigen-specific therapies, particular attention is given to antigen-specific generation of Foxp3+ Treg cells and their potential use to limit autoimmunity such as T1D.
- Published
- 2012
- Full Text
- View/download PDF
37. Extra-thymically induced regulatory T cells: Do they have potential in disease prevention?
- Author
-
Carolin Daniel and Harald von Boehmer
- Subjects
Autoimmune disease ,Cellular differentiation ,Vaccination ,Immunology ,FOXP3 ,Cell Differentiation ,chemical and pharmacologic phenomena ,Thymus Gland ,Biology ,medicine.disease ,medicine.disease_cause ,T-Lymphocytes, Regulatory ,Article ,Autoimmune Diseases ,Autoimmunity ,Immune tolerance ,Immune system ,Immune Tolerance ,medicine ,Animals ,Humans ,Immunology and Allergy ,Disease prevention - Abstract
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.
- Published
- 2011
- Full Text
- View/download PDF
38. Retinoic acid can enhance conversion of naive into regulatory T cells independently of secreted cytokines
- Author
-
Harald von Boehmer, Jens Nolting, Hye-Jung Kim, Henry M. Sucov, Carolin Daniel, Byung-Gyu Kim, Peng Li, Karsten Kretschmer, Sabine Reuter, Christina H. Stuelten, and John J. Letterio
- Subjects
medicine.medical_specialty ,Receptors, Retinoic Acid ,medicine.medical_treatment ,Immunology ,Retinoic acid ,Tretinoin ,Lymphocyte Activation ,T-Lymphocytes, Regulatory ,03 medical and health sciences ,chemistry.chemical_compound ,Mice ,0302 clinical medicine ,Transforming Growth Factor beta ,Internal medicine ,medicine ,Immunology and Allergy ,Animals ,Smad3 Protein ,030304 developmental biology ,Smad4 Protein ,0303 health sciences ,biology ,Retinoic Acid Receptor alpha ,Brief Definitive Report ,Interleukin ,Transforming growth factor beta ,Cell biology ,Mice, Inbred C57BL ,Retinoic acid receptor ,Endocrinology ,Cytokine ,Hyaluronan Receptors ,chemistry ,Retinoic acid receptor alpha ,biology.protein ,Cytokines ,Cytokine secretion ,030215 immunology ,medicine.drug - Abstract
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 alpha (RAR alpha) but did not require Smad3, despite the fact that RA enhanced Smad3 expression. The RAR alpha 1 isoform was not essential for RA-dependent enhancement of transforming growth factor beta-driven conversion, suggesting that conversion can also be mediated by RAR alpha 2. Interleukin (IL)-6 strongly reduced RAR alpha expression levels such that a deficiency of the predominant RAR alpha 1 isoform leaves too little RAR alpha 2 for RA to inhibit the generation of Th17 cells in the presence of IL-6.
- Published
- 2009
39. Salmon calcitonin – a potent inhibitor of food intake in states of impaired leptin signalling in laboratory rodents
- Author
-
Alexandra Steinbrueck, Ingrid Schmidt, Eckhart Simon, Sandra Eiden, and Carolin Daniel
- Subjects
Calcitonin ,Leptin ,Male ,Food intake ,medicine.medical_specialty ,Physiology ,Mice, Obese ,Body Mass Index ,Eating ,Mice ,Inbred strain ,Internal medicine ,medicine ,Animals ,Insulin ,Receptor ,Mice, Knockout ,Leptin Deficiency ,Chemistry ,digestive, oral, and skin physiology ,Rats, Inbred Strains ,Original Articles ,medicine.disease ,Obesity ,Diet ,Rats ,Mice, Inbred C57BL ,Endocrinology ,Receptors, Corticotropin ,Anorectic ,Receptor, Melanocortin, Type 4 ,Receptors, Leptin ,Female ,Melanocortin ,Energy Intake ,Signal Transduction - Abstract
To compare the anorectic effectiveness of leptin and the amylin analogue salmon calcitonin (sCT), rodents were treated on 1 day with subcutaneous injections. In chow-fed C57Bl/6J mice, leptin and sCT reduced energy intake and acted additively. After C57Bl/6J mice had become leptin-resistant on being fed chocolate as a palatable high-caloric supplement to chow, their sCT-induced decrease in energy intake was more pronounced than in chow-fed mice with differential changes in the intake of chocolate (strong reduction) and chow (slight increase). Dose-response relationships for sCT-induced reductions in energy intake were analysed in chow-fed C57Bl/6J mice and two obese strains, ob/ob mice and melanocortin-4 receptor knockout (MC4-r-KO) mice, as well as in wild-type and fatty (fa/fa) rats. Compared to C57Bl/6J mice, reduction in food intake induced by sCT was attenuated in MC4-r-KO mice, and nearly absent in ob/ob mice, over the dose range investigated. Compared to C57Bl/6J mice, wild-type rats responded more sensitively to sCT and its efficiency was only slightly reduced in fatty (fa/fa) rats. Thus, while genetically induced failures of leptin signalling reduce the action of sCT, it effectively inhibits the intake of a palatable, high fat-high sugar diet even in states of diet-induced obesity with functional leptin resistance.
- Published
- 2002
- Full Text
- View/download PDF
40. A Stat6/Pten Axis Links Regulatory T Cells with Adipose Tissue Function
- Author
-
Françoise Rohner-Jeanrenaud, Ralf Palmisano, Martin Jastroch, Carolin Daniel, Anette-Gabriele Ziegler, Maike Becker, Martin G. Scherm, Mohammad M.H. Mollah, Benno Weigmann, Isabelle Serr, Victoria K. Flynn, Philipp Tripal, Tobias Bopp, Manuel Serrano, Stephen C. Woods, Lucas F. Nascimento, Daniel Lamp, Fabian Hosp, Matthias H. Tschöp, Sarah Dietzen, Katharina Gerlach, Matthias Blüher, Christian Wolfrum, Mark H. Kaplan, Matthias Mann, Vanessa Popp, Verena B. Ott, Purna Krishnamurthy, Stefanie Kälin, Susanne Keipert, and Rohner-Jeanrenaud, Françoise
- Subjects
0301 basic medicine ,PTEN ,Proteome ,Physiology ,Adipose tissue ,Stimulation ,mTORC1 ,Diet induced thermogenesis ,Borcs6 ,C17orf59 ,Foxp3 ,Pten ,Stat6 ,T Cells ,Tregs ,Adipose Tissue Function ,Cold Exposure ,Metabolic Function ,Metabolism ,Regulatory T cells ,T-Lymphocytes, Regulatory ,chemistry.chemical_compound ,0302 clinical medicine ,Adipose Tissue, Brown ,Adipocyte ,Uncoupling Protein 1 ,Tissue homeostasis ,STAT6 ,ddc:616 ,Mice, Inbred BALB C ,FOXP3 ,Forkhead Transcription Factors ,hemic and immune systems ,Cell biology ,Cold Temperature ,Female ,Metabolic function ,medicine.symptom ,Signal Transduction ,Adipose Tissue, White ,Cold exposure ,T cells ,chemical and pharmacologic phenomena ,Inflammation ,Biology ,Article ,03 medical and health sciences ,Receptors, Adrenergic, beta ,Adipose tissue function ,medicine ,Animals ,Molecular Biology ,PTEN Phosphohydrolase ,Cell Biology ,030104 developmental biology ,chemistry ,Immunology ,STAT6 Transcription Factor ,030217 neurology & neurosurgery - Abstract
Obesity and type 2 diabetes are associated with metabolic defects and adipose tissue inflammation. Foxp3(+) regulatory Tcells (Tregs) control tissue homeostasis by counteracting local inflammation. However, if and how Tcells interlink environmental influences with adipocyte function remains unknown. Here, we report that enhancing sympathetic tone by cold exposure, beta3-adrenergic receptor (ADRB3) stimulation or a short-term high-calorie diet enhances Treg induction invitro and invivo. CD4(+) Tcell proteomes revealed higher expression of Foxp3 regulatory networks in response to cold or ADRB3 stimulation invivo reflecting Treg induction. Specifically, Ragulator-interacting protein C17orf59, which limits mTORC1 activity, was upregulated in CD4(+) Tcells by either ADRB3 stimulation or cold exposure, suggesting contribution to Treg induction. By loss- and gain-of-function studies, including Treg depletion and transfers invivo, we demonstrated that a Tcell-specific Stat6/Pten axis links cold exposure or ADRB3 stimulation with Foxp3(+) Treg induction and adipose tissue function. Our findings offer a new mechanistic model in which tissue-specific Tregs maintain adipose tissue function.
- Published
- 2017
- Full Text
- View/download PDF
41. Nuclear factor of activated T cells - a transcription factor family as critical regulator in lung and colon cancer
- Author
-
Carolin, Daniel, Katharina, Gerlach, Martin, Väth, Markus F, Neurath, and Benno, Weigmann
- Subjects
Enzyme Activation ,Inflammation ,Cell Transformation, Neoplastic ,Lung Neoplasms ,NFATC Transcription Factors ,T-Lymphocytes ,Nuclear Factor of activated T celss ,Cancer ,Transcription factor ,Colonic Neoplasms ,cardiovascular system ,Humans ,Signal Transduction - Abstract
Nuclear factor of activated T cells (NFAT) was first identified as a transcription factor which is activated upon T cell stimulation. Subsequent studies uncovered that a whole family of individual NFAT proteins exists with pleiotropic functions not only in immune but also in nonimmune cells. However, dysregulation of NFAT thereby favors malignant growth and cancer. Summarizing the recent advances in understanding how individual NFAT factors regulate the immune system, this review gives new insights into the critical role of NFAT in cancer development with special focus on inflammation-associated colorectal cancer.
- Published
- 2014
42. Cyclosporine A regulates pro-inflammatory cytokine production in ulcerative colitis
- Author
-
Anika Fischer, Carolin Daniel, Imke Atreya, Stefanie Steiner, Benno Weigmann, Maximilian J. Waldner, Raja Atreya, Markus F. Neurath, Helmut Neumann, and Simon Hirschmann
- Subjects
Adult ,CD4-Positive T-Lymphocytes ,Adolescent ,Cell Survival ,medicine.medical_treatment ,T cell ,T-Lymphocytes ,Immunology ,Apoptosis ,Peripheral blood mononuclear cell ,Inflammatory bowel disease ,Young Adult ,Crohn Disease ,Cyclosporin a ,Immunology and Allergy ,Medicine ,Humans ,Colitis ,Aged ,Inflammation ,Caspase 8 ,Interleukin-13 ,business.industry ,General Medicine ,Middle Aged ,medicine.disease ,Flow Cytometry ,Ulcerative colitis ,Cytokine ,medicine.anatomical_structure ,Gene Expression Regulation ,Microscopy, Fluorescence ,Case-Control Studies ,Interleukin 13 ,Cyclosporine ,Leukocytes, Mononuclear ,Cytokines ,Colitis, Ulcerative ,business - Abstract
Crohn's disease (CD) and ulcerative colitis (UC) are the two major forms of inflammatory bowel diseases (IBD), which are defined as relapsing inflammations of the gastrointestinal tract. Cyclosporine A (CsA) is a potential rescue treatment to avoid colectomy in severe steroid-refractory UC patients. The molecular mechanism of action of CsA in UC is nevertheless still not well understood. The aim of this study was to investigate the effect of CsA on a possible modulation of cytokine production by peripheral blood mononuclear cells (PBMCs) of controls and patients with UC or CD. Upon CsA treatment, analyses of cytokine levels revealed a significant reduction of IL-13 expression in PBMCs from patients with UC, whereas other cytokine expression levels remained unaffected. To address the question whether CsA treatment impinges on the induction of cell death, apoptosis assays were performed using CD4(+) T cells from peripheral blood of patients suffering from either UC or CD. It became clear that CsA treatment resulted in a specific induction of apoptosis in samples from controls and patients with UC but not with CD. Apoptosis induction was not mediated via the mitochondrial apoptosis pathway. The present data support the concept that CsA treatment modulates pro-inflammatory cytokine production and T cell survival in UC via the induction of apoptosis and might therefore help to explain the clinical efficacy of CsA in patients with UC.
- Published
- 2013
43. Therapeutic opportunities for manipulating T(Reg) cells in autoimmunity and cancer
- Author
-
Harald von Boehmer and Carolin Daniel
- Subjects
chemical and pharmacologic phenomena ,Autoimmunity ,Disease ,Biology ,medicine.disease_cause ,T-Lymphocytes, Regulatory ,Autoimmune Diseases ,Immune system ,Neoplasms ,Drug Discovery ,medicine ,Animals ,Humans ,Pharmacology ,FOXP3 ,Cancer ,hemic and immune systems ,Forkhead Transcription Factors ,General Medicine ,medicine.disease ,Self Tolerance ,Cancer cell ,Immunology ,Cancer research ,Immunotherapy ,Reprogramming ,Function (biology) - Abstract
Forkhead box P3 (FOXP3)-expressing regulatory T (T(Reg)) 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 T(Reg) cells should permit the development of therapeutics for reprogramming the immune system. In this Review, we discuss insights into the generation of T(Reg) cells, their involvement in disease and the molecular basis of the dominant tolerance exerted by FOXP3(+) T(Reg) cells that could permit their safe and specific manipulation in humans.
- Published
- 2013
44. Extrathymic generation of regulatory T cells--chances and challenges for prevention of autoimmune disease
- Author
-
Carolin, Daniel and Harald, von Boehmer
- Subjects
Peripheral Tolerance ,Receptors, Antigen, T-Cell ,Epitopes, T-Lymphocyte ,Immunotherapy, Active ,Cell Differentiation ,Forkhead Transcription Factors ,Autoantigens ,T-Lymphocytes, Regulatory ,Autoimmune Diseases ,Immunomodulation ,Diabetes Mellitus, Type 1 ,Biomimetic Materials ,Animals ,Humans - Abstract
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.
- Published
- 2011
45. Prevention of type 1 diabetes in mice by tolerogenic vaccination with a strong agonist insulin mimetope
- Author
-
Roderick T. Bronson, Carolin Daniel, Benno Weigmann, and Harald von Boehmer
- Subjects
medicine.medical_specialty ,T cell ,medicine.medical_treatment ,Insulin Antibodies ,Immunology ,Mice, Transgenic ,Nod ,Biology ,T-Lymphocytes, Regulatory ,Epitope ,Article ,Immune tolerance ,03 medical and health sciences ,Epitopes ,Mice ,0302 clinical medicine ,Mice, Inbred NOD ,Internal medicine ,medicine ,Immune Tolerance ,Immunology and Allergy ,Animals ,Humans ,Insulin ,Amino Acid Sequence ,Pancreas ,030304 developmental biology ,NOD mice ,Autoantibodies ,0303 health sciences ,Type 1 diabetes ,Vaccines ,Vaccination ,FOXP3 ,Forkhead Transcription Factors ,medicine.disease ,3. Good health ,Endocrinology ,medicine.anatomical_structure ,Diabetes Mellitus, Type 1 ,Female ,030215 immunology - Abstract
Subimmunogenic vaccination with an agonist mimetope of insulin converts naive T cells into regulatory T cells and prevents type 1 diabetes in NOD mice., Type 1 diabetes (T1D) results from the destruction of insulin-secreting pancreatic β cells by autoreactive T cells. Insulin is an essential target of the autoimmune attack. Insulin epitopes recognized by diabetogenic T cell clones bind poorly to the class II I-Ag7 molecules of nonobese diabetic (NOD) mice, which results in weak agonistic activity of the peptide MHC complex. Here, we describe a strongly agonistic insulin mimetope that effectively converts naive T cells into Foxp3+ regulatory T cells in vivo, thereby completely preventing T1D in NOD mice. In contrast, natural insulin epitopes are ineffective. Subimmunogenic vaccination with strongly agonistic insulin mimetopes might represent a novel strategy to prevent T1D in humans at risk for the disease.
- Published
- 2011
46. Antigen-specific induction of regulatory T cells in vivo and in vitro
- Author
-
Carolin, Daniel, Hidde, Ploegh, and Harald, von Boehmer
- Subjects
Minor Histocompatibility Antigens ,Antigens, CD ,Transforming Growth Factor beta ,Immunologic Techniques ,Animals ,Lectins, C-Type ,Receptors, Cell Surface ,Tretinoin ,Antigens ,T-Lymphocytes, Regulatory - Abstract
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.
- Published
- 2011
47. Antigen-Specific Induction of Regulatory T Cells In Vivo and In Vitro
- Author
-
Carolin Daniel, Harald von Boehmer, and Hidde L. Ploegh
- Subjects
biology ,Retinoic acid ,chemical and pharmacologic phenomena ,In vitro ,Cell biology ,TCIRG1 ,chemistry.chemical_compound ,Immune system ,chemistry ,Antigen ,biology.protein ,IL-2 receptor ,Antibody ,Receptor - Abstract
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.
- Published
- 2011
- Full Text
- View/download PDF
48. Extrathymic Generation of Regulatory T Cells—Chances and Challenges for Prevention of Autoimmune Disease
- Author
-
Harald von Boehmer and Carolin Daniel
- Subjects
Autoimmune disease ,Agonist ,Type 1 diabetes ,business.industry ,medicine.drug_class ,T-cell receptor ,FOXP3 ,hemic and immune systems ,chemical and pharmacologic phenomena ,medicine.disease ,Vaccination ,Immune system ,Immunity ,Immunology ,medicine ,business - Abstract
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.
- Published
- 2011
- Full Text
- View/download PDF
49. Enhancement of antigen-specific Treg vaccination in vivo
- Author
-
Kerstin Wennhold, Hye-Jung Kim, Harald von Boehmer, and Carolin Daniel
- Subjects
Interleukin 2 ,medicine.medical_treatment ,chemical and pharmacologic phenomena ,Biology ,Lymphocyte Activation ,T-Lymphocytes, Regulatory ,Mice ,Immune system ,Antigen ,In vivo ,medicine ,Animals ,IL-2 receptor ,Everolimus ,Antigens ,Cell Proliferation ,Sirolimus ,Multidisciplinary ,Receptors, Purinergic P2 ,Vaccination ,Interleukin-2 Receptor alpha Subunit ,FOXP3 ,hemic and immune systems ,Forkhead Transcription Factors ,Biological Sciences ,Cell biology ,Interleukin-10 ,Mice, Inbred C57BL ,Interleukin 10 ,Cytokine ,Immunology ,Interleukin-2 ,Female ,Receptors, Purinergic P2X7 ,Immunosuppressive Agents ,medicine.drug - Abstract
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.
- Published
- 2010
50. Effects of periodic intake of a high-caloric diet on body mass and leptin resistance
- Author
-
Ingrid Schmidt, Sandra Eiden, Carolin Daniel, Alexandra Steinbrück, and Mauricio Berriel Díaz
- Subjects
Leptin ,medicine.medical_specialty ,Periodicity ,Drug Resistance ,Adipokine ,Experimental and Cognitive Psychology ,Biology ,Body Mass Index ,Behavioral Neuroscience ,Eating ,Mice ,Inbred strain ,Internal medicine ,medicine ,Animals ,Leptin resistance ,Behavior, Animal ,Body Weight ,Age Factors ,Caloric theory ,medicine.disease ,Obesity ,Dietary Fats ,Mice, Inbred C57BL ,Endocrinology ,Dietary history ,Anorectic ,Female ,Energy Intake - Abstract
The effects of continuous or intermittent access to a high-caloric (HC) diet, always offered in addition to standard chow, on body mass and leptin resistance were analyzed in female C57BL/6J mice. Susceptibility for diet-induced obesity (DIO) was apparent from the marked preference for the HC diet. Continuous HC diet feeding of mice at 4 weeks of age induced leptin resistance within 2 weeks and massive gains in body mass, although with increasing inter-individual variability in the inbred strain considered to be isogenic. In adult mice receiving HC diet for the first time, leptin treatment failed to reduce energy intake first after 11 days of HC diet feeding, but became effective again within 3 days after HC diet withdrawal. In mice with a history of several preceding periods of access to the HC diet totalling >30 days, supplementary HC diet abolished the anorectic effect of leptin treatment within only 3 days and it reappeared not earlier than 11 days after HC diet withdrawal. Thus, in the investigated DIO-prone mouse strain both, the loss of responsiveness to leptin under HC diet and its recovery after HC diet withdrawal strongly depended on the dietary history. Recovery from leptin resistance during periods of intermittent chow feeding was associated with losses of body mass that did not completely compensate for the obesity-inducing effect of the preceding HC diet.
- Published
- 2005
Catalog
Discovery Service for Jio Institute Digital Library
For full access to our library's resources, please sign in.