167 results on '"Regina Feederle"'
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. The Nuclear Speckles Protein SRRM2 Is Exposed on the Surface of Cancer Cells
- Author
-
Markus Kellner, Julia Hörmann, Susanne Fackler, Yuanyu Hu, Tielin Zhou, Lin Lu, Ibrahim Ilik, Tugce Aktas, Regina Feederle, Stefanie M. Hauck, Olivier Gires, Kathrin Gärtner, Lietao Li, and Reinhard Zeidler
- Subjects
SRRM2 ,target identification ,cancer target ,therapeutic antibodies ,Cytology ,QH573-671 - Abstract
The membrane composition of extracellular vesicles (EVs) largely reflects that of the plasma membrane of the cell of origin. We therefore hypothesized that EVs could be used for immunizations to generate monoclonal antibodies against well-known tumor antigens but possibly also against hitherto unknown tumor-associated target molecules. From an immunization experiment, we obtained a monoclonal antibody specific for SRRM2, an RNA-binding protein involved in splicing and a major component of nuclear speckles. Here, we used this antibody to demonstrate that SRRM2 is exposed on the surface of most cancer cell lines from various entities and, even more important, on cancer cells in vivo. Moreover, we demonstrated that SRRM2-specific CAR-T cells are functional in vitro and in vivo. Collectively, we identified SRRM2 as a promising new target molecule exposed on the cancer cell surface and showed that our SRRM2-specific antibody can be used as a basis for the development of new targeted cancer therapies.
- Published
- 2024
- Full Text
- View/download PDF
4. The Aurora B-controlled PP1/RepoMan complex determines the spatial and temporal distribution of mitotic H2B S6 phosphorylation
- Author
-
Maximilian Pfisterer, Roman Robert, Vera V. Saul, Amelie Pritz, Markus Seibert, Regina Feederle, and M. Lienhard Schmitz
- Subjects
mitosis ,histone phosphorylation ,phosphatase scaffold ,centromere ,Biology (General) ,QH301-705.5 - Abstract
The precise spatial and temporal control of histone phosphorylations is important for the ordered progression through the different phases of mitosis. The phosphorylation of H2B at S6 (H2B S6ph), which is crucial for chromosome segregation, reaches its maximum level during metaphase and is limited to the inner centromere. We discovered that the temporal and spatial regulation of this modification, as well as its intensity, are governed by the scaffold protein RepoMan and its associated catalytically active phosphatases, PP1α and PP1γ. Phosphatase activity is inhibited at the area of maximal H2B S6 phosphorylation at the inner centromere by site-specific Aurora B-mediated inactivation of the PP1/RepoMan complex. The motor protein Mklp2 contributes to the relocalization of Aurora B from chromatin to the mitotic spindle during anaphase, thus alleviating Aurora B-dependent repression of the PP1/RepoMan complex and enabling dephosphorylation of H2B S6. Accordingly, dysregulation of Mklp2 levels, as commonly observed in tumour cells, leads to the lack of H2B S6 dephosphorylation during early anaphase, which might contribute to chromosomal instability.
- Published
- 2024
- Full Text
- View/download PDF
5. Targeting the glycine-rich domain of TDP-43 with antibodies prevents its aggregation in vitro and reduces neurofilament levels in vivo
- Author
-
Henrick Riemenschneider, Francesca Simonetti, Udit Sheth, Eszter Katona, Stefan Roth, Saskia Hutten, Daniel Farny, Meike Michaelsen, Brigitte Nuscher, Michael K. Schmidt, Andrew Flatley, Aloys Schepers, Lara A. Gruijs da Silva, Qihui Zhou, Thomas Klopstock, Arthur Liesz, Thomas Arzberger, Jochen Herms, Regina Feederle, Tania F. Gendron, Dorothee Dormann, and Dieter Edbauer
- Subjects
Neurodegeneration ,Amyotrophic lateral sclerosis ,Frontotemporal dementia ,TDP-43 ,Immunotherapy ,Phase separation ,Neurology. Diseases of the nervous system ,RC346-429 - Abstract
Abstract Cytoplasmic aggregation and concomitant nuclear clearance of the RNA-binding protein TDP-43 are found in ~ 90% of cases of amyotrophic lateral sclerosis and ~ 45% of patients living with frontotemporal lobar degeneration, but no disease-modifying therapy is available. Antibody therapy targeting other aggregating proteins associated with neurodegenerative disorders has shown beneficial effects in animal models and clinical trials. The most effective epitopes for safe antibody therapy targeting TDP-43 are unknown. Here, we identified safe and effective epitopes in TDP-43 for active and potential future passive immunotherapy. We prescreened 15 peptide antigens covering all regions of TDP-43 to identify the most immunogenic epitopes and to raise novel monoclonal antibodies in wild-type mice. Most peptides induced a considerable antibody response and no antigen triggered obvious side effects. Thus, we immunized mice with rapidly progressing TDP-43 proteinopathy (“rNLS8” model) with the nine most immunogenic peptides in five pools prior to TDP-43ΔNLS transgene induction. Strikingly, combined administration of two N-terminal peptides induced genetic background-specific sudden lethality in several mice and was therefore discontinued. Despite a strong antibody response, no TDP-43 peptide prevented the rapid body weight loss or reduced phospho-TDP-43 levels as well as the profound astrogliosis and microgliosis in rNLS8 mice. However, immunization with a C-terminal peptide containing the disease-associated phospho-serines 409/410 significantly lowered serum neurofilament light chain levels, indicative of reduced neuroaxonal damage. Transcriptomic profiling showed a pronounced neuroinflammatory signature (IL-1β, TNF-α, NfκB) in rNLS8 mice and suggested modest benefits of immunization targeting the glycine-rich region. Several novel monoclonal antibodies targeting the glycine-rich domain potently reduced phase separation and aggregation of TDP-43 in vitro and prevented cellular uptake of preformed aggregates. Our unbiased screen suggests that targeting the RRM2 domain and the C-terminal region of TDP-43 by active or passive immunization may be beneficial in TDP-43 proteinopathies by inhibiting cardinal processes of disease progression. Graphical Abstract
- Published
- 2023
- Full Text
- View/download PDF
6. Translational Studies Using the MALT1 Inhibitor (S)-Mepazine to Induce Treg Fragility and Potentiate Immune Checkpoint Therapy in Cancer
- Author
-
Mauro Di Pilato, Yun Gao, Yi Sun, Amina Fu, Carina Grass, Thomas Seeholzer, Regina Feederle, Irina Mazo, Samuel W. Kazer, Kevin Litchfield, Ulrich H. von Andrian, Thorsten R. Mempel, Russell W. Jenkins, Daniel Krappmann, and Peter Keller
- Subjects
malt1 ,cancer immunotherapy ,regulatory t cells ,fragile tregs ,interferon-γ ,Neoplasms. Tumors. Oncology. Including cancer and carcinogens ,RC254-282 ,Immunologic diseases. Allergy ,RC581-607 - Abstract
Introduction: Regulatory T cells (Tregs) play a critical role in the maintenance of immune homeostasis but also protect tumors from immune-mediated growth control or rejection and pose a significant barrier to effective immunotherapy. Inhibition of MALT1 paracaspase activity can selectively reprogram immune-suppressive Tregs in the tumor microenvironment to adopt a proinflammatory fragile state, which offers an opportunity to impede tumor growth and enhance the efficacy of immune checkpoint therapy (ICT). Methods: We performed preclinical studies with the orally available allosteric MALT1 inhibitor (S)-mepazine as a single-agent and in combination with anti-programmed cell death protein 1 (PD-1) ICT to investigate its pharmacokinetic properties and antitumor effects in several murine tumor models as well as patient-derived organotypic tumor spheroids (PDOTS). Results: (S)-mepazine demonstrated significant antitumor effects and was synergistic with anti-PD-1 therapy in vivo and ex vivo but did not affect circulating Treg frequencies in healthy rats at effective doses. Pharmacokinetic profiling revealed favorable drug accumulation in tumors to concentrations that effectively blocked MALT1 activity, potentially explaining preferential effects on tumor-infiltrating over systemic Tregs. Conclusions: The MALT1 inhibitor (S)-mepazine showed single-agent anticancer activity and presents a promising opportunity for combination with PD-1 pathway-targeted ICT. Activity in syngeneic tumor models and human PDOTS was likely mediated by induction of tumor-associated Treg fragility. This translational study supports ongoing clinical investigations (ClinicalTrials.gov Identifier: NCT04859777) of MPT-0118, (S)-mepazine succinate, in patients with advanced or metastatic treatment-refractory solid tumors.
- Published
- 2023
- Full Text
- View/download PDF
7. Sirtuin-1 sensitive lysine-136 acetylation drives phase separation and pathological aggregation of TDP-43
- Author
-
Jorge Garcia Morato, Friederike Hans, Felix von Zweydorf, Regina Feederle, Simon J. Elsässer, Angelos A. Skodras, Christian Johannes Gloeckner, Emanuele Buratti, Manuela Neumann, and Philipp J. Kahle
- Subjects
Science - Abstract
TDP-43 is a nucleic acid binding protein, whose insoluble aggregates are neuropathological hallmarks of specific subsets of patients with amyotrophic lateral sclerosis and frontotemporal lobar degeneration. Post-translational modifications and acetylation of TDP-43 impact its interaction with RNA, its localization in the cells, and are linked to disease. Using antibodies generated against TDP-43 lysine acetylation sites, sirtuin-1 was found to potently deacetylate amber suppressed [acK136]TDP-43 and reduce its aggregation propensity. Thus, distinct lysine acetylations modulate nuclear import, RNA binding as well as phase separation and aggregation of TDP-43, suggesting regulatory mechanisms for TDP-43 pathogenesis.
- Published
- 2022
- Full Text
- View/download PDF
8. Defining the RBPome of primary T helper cells to elucidate higher-order Roquin-mediated mRNA regulation
- Author
-
Kai P. Hoefig, Alexander Reim, Christian Gallus, Elaine H. Wong, Gesine Behrens, Christine Conrad, Meng Xu, Lisa Kifinger, Taku Ito-Kureha, Kyra A. Y. Defourny, Arie Geerlof, Josef Mautner, Stefanie M. Hauck, Dirk Baumjohann, Regina Feederle, Matthias Mann, Michael Wierer, Elke Glasmacher, and Vigo Heissmeyer
- Subjects
Science - Abstract
An extensive RNA binding protein atlas (RBPome) for primary T cells would be a useful resource. Here the authors use two different methods to characterise the mouse and human T cell RBPome and show regulation of Roquin-1/2 dependent and independent pathways.
- Published
- 2021
- Full Text
- View/download PDF
9. A novel monoclonal IgG1 antibody specific for Galactose-alpha-1,3-galactose questions alpha-Gal epitope expression by bacteria
- Author
-
Luisa Kreft, Aloys Schepers, Miriam Hils, Kyra Swiontek, Andrew Flatley, Robert Janowski, Mohammadali Khan Mirzaei, Michael Dittmar, Neera Chakrapani, Mahesh S. Desai, Stefanie Eyerich, Li Deng, Dierk Niessing, Konrad Fischer, Regina Feederle, Simon Blank, Carsten B. Schmidt-Weber, Christiane Hilger, Tilo Biedermann, and Caspar Ohnmacht
- Subjects
alpha-Gal ,α-Gal ,IgG ,monoclonal antibody ,carbohydrate ,red meat allergy ,Immunologic diseases. Allergy ,RC581-607 - Abstract
The alpha-Gal epitope (α-Gal) with the determining element galactose-α1,3-galactose can lead to clinically relevant allergic reactions and rejections in xenotransplantation. These immune reactions can develop because humans are devoid of this carbohydrate due to evolutionary loss of the enzyme α1,3-galactosyltransferase (GGTA1). In addition, up to 1% of human IgG antibodies are directed against α-Gal, but the stimulus for the induction of anti-α-Gal antibodies is still unclear. Commensal bacteria have been suggested as a causal factor for this induction as α-Gal binding tools such as lectins were found to stain cultivated bacteria isolated from the intestinal tract. Currently available tools for the detection of the definite α-Gal epitope, however, are cross-reactive, or have limited affinity and, hence, offer restricted possibilities for application. In this study, we describe a novel monoclonal IgG1 antibody (27H8) specific for the α-Gal epitope. The 27H8 antibody was generated by immunization of Ggta1 knockout mice and displays a high affinity towards synthetic and naturally occurring α-Gal in various applications. Using this novel tool, we found that intestinal bacteria reported to be α-Gal positive cannot be stained with 27H8 questioning whether commensal bacteria express the native α-Gal epitope at all.
- Published
- 2022
- Full Text
- View/download PDF
10. PAT2 regulates vATPase assembly and lysosomal acidification in brown adipocytes
- Author
-
Jiefu Wang, Yasuhiro Onogi, Martin Krueger, Josef Oeckl, Ruth Karlina, Inderjeet Singh, Stefanie M. Hauck, Regina Feederle, Yongguo Li, and Siegfried Ussar
- Subjects
Brown adipocytes ,Lysosomal acidification ,Proton-coupled amino acid transporter ,Transporter translocation across membranes ,V-ATPase assembly ,Internal medicine ,RC31-1245 - Abstract
Objective: Brown adipocytes play a key role in maintaining body temperature as well as glucose and lipid homeostasis. However, brown adipocytes need to adapt their thermogenic activity and substrate utilization to changes in nutrient availability. Amongst the multiple factors influencing brown adipocyte activity, autophagy is an important regulatory element of thermogenic capacity and activity. Nevertheless, a specific sensing mechanism of extracellular amino acid availability linking autophagy to nutrient availability in brown adipocytes is unknown. Methods: To characterize the role of the amino acid transporter PAT2/SLC36A2 in brown adipocytes, loss or gain of function of PAT2 were studied with respect to differentiation, subcellular localization, lysosomal activity and autophagy. Activity of vATPase was evaluated by quenching of EGFP fused to LC3 or FITC-dextran loaded lysosomes in brown adipocytes upon amino acid starvation, whereas the effect of PAT2 on assembly of the vATPase was investigated by Native-PAGE. Results: We show that PAT2 translocates from the plasma membrane to the lysosome in response to amino acid withdrawal. Loss or overexpression of PAT2 impair lysosomal acidification and starvation-induced S6K re-phosphorylation, as PAT2 facilitates the assembly of the lysosomal vATPase, by recruitment of the cytoplasmic V1 subunit to the lysosome. Conclusions: PAT2 is an important sensor of extracellular amino acids and regulator of lysosomal acidification in brown adipocytes.
- Published
- 2022
- Full Text
- View/download PDF
11. Establishment and Functional Characterization of Murine Monoclonal Antibodies Recognizing Neuritin
- Author
-
Georgia Papadogianni, Inga Ravens, Ahmed Hassan, Andrew Flatley, Regina Feederle, Günter Bernhardt, and Hristo Georgiev
- Subjects
neuritin ,blocking antibody ,flow cytometry ,ELISA ,monoclonal antibody ,Immunologic diseases. Allergy ,RC581-607 - Abstract
Neuritin represents a neurotrophic factor that is not only important in neuronal development and plasticity but also impacts endothelial angiogenesis, cell migration, tumor growth and the production of antibodies by B cells. We established monoclonal mouse anti-mouse neuritin antibodies by immunizing knock-out mice with two different neuritin-derived peptides. Because neuritin is well conserved between species, these new monoclonal antibodies recognize the neuritin of a wide variety of species, including human. Moreover, they not only recognize specifically surface-bound neuritin expressed by murine follicular regulatory T cells but also the block binding of recombinant neuritin to germinal center B cells. This suggests that these newly generated tools will be of great use in studying neuritin expression and function.
- Published
- 2023
- Full Text
- View/download PDF
12. Active site geometry stabilization of a presenilin homolog by the lipid bilayer promotes intramembrane proteolysis
- Author
-
Lukas P Feilen, Shu-Yu Chen, Akio Fukumori, Regina Feederle, Martin Zacharias, and Harald Steiner
- Subjects
amyloid β-peptide ,intramembrane proteolysis ,presenilin ,PSH ,γ-secretase ,Medicine ,Science ,Biology (General) ,QH301-705.5 - Abstract
Cleavage of membrane proteins in the lipid bilayer by intramembrane proteases is crucial for health and disease. Although different lipid environments can potently modulate their activity, how this is linked to their structural dynamics is unclear. Here, we show that the carboxy-peptidase-like activity of the archaeal intramembrane protease PSH, a homolog of the Alzheimer’s disease-associated presenilin/γ-secretase is impaired in micelles and promoted in a lipid bilayer. Comparative molecular dynamics simulations revealed that important elements for substrate binding such as transmembrane domain 6a of PSH are more labile in micelles and stabilized in the lipid bilayer. Moreover, consistent with an enhanced interaction of PSH with a transition-state analog inhibitor, the bilayer promoted the formation of the enzyme’s catalytic active site geometry. Our data indicate that the lipid environment of an intramembrane protease plays a critical role in structural stabilization and active site arrangement of the enzyme-substrate complex thereby promoting intramembrane proteolysis.
- Published
- 2022
- Full Text
- View/download PDF
13. Oligodendrocyte myelin glycoprotein as a novel target for pathogenic autoimmunity in the CNS
- Author
-
Ramona Gerhards, Lena Kristina Pfeffer, Jessica Lorenz, Laura Starost, Luise Nowack, Franziska S. Thaler, Miriam Schlüter, Heike Rübsamen, Caterina Macrini, Stephan Winklmeier, Simone Mader, Mattias Bronge, Hans Grönlund, Regina Feederle, Hung-En Hsia, Stefan F. Lichtenthaler, Juliane Merl-Pham, Stefanie M. Hauck, Tanja Kuhlmann, Isabel J. Bauer, Eduardo Beltran, Lisa Ann Gerdes, Aleksandra Mezydlo, Amit Bar-Or, Brenda Banwell, Mohsen Khademi, Tomas Olsson, Reinhard Hohlfeld, Hans Lassmann, Tania Kümpfel, Naoto Kawakami, and Edgar Meinl
- Subjects
Autoantigen ,Multiple sclerosis ,Neuroinflammation ,Autoimmunity ,Neurology. Diseases of the nervous system ,RC346-429 - Abstract
Abstract Autoimmune disorders of the central nervous system (CNS) comprise a broad spectrum of clinical entities. The stratification of patients based on the recognized autoantigen is of great importance for therapy optimization and for concepts of pathogenicity, but for most of these patients, the actual target of their autoimmune response is unknown. Here we investigated oligodendrocyte myelin glycoprotein (OMGP) as autoimmune target, because OMGP is expressed specifically in the CNS and there on oligodendrocytes and neurons. Using a stringent cell-based assay, we detected autoantibodies to OMGP in serum of 8/352 patients with multiple sclerosis, 1/28 children with acute disseminated encephalomyelitis and unexpectedly, also in one patient with psychosis, but in none of 114 healthy controls. Since OMGP is GPI-anchored, we validated its recognition also in GPI-anchored form. The autoantibodies to OMGP were largely IgG1 with a contribution of IgG4, indicating cognate T cell help. We found high levels of soluble OMGP in human spinal fluid, presumably due to shedding of the GPI-linked OMGP. Analyzing the pathogenic relevance of autoimmunity to OMGP in an animal model, we found that OMGP-specific T cells induce a novel type of experimental autoimmune encephalomyelitis dominated by meningitis above the cortical convexities. This unusual localization may be directed by intrathecal uptake and presentation of OMGP by meningeal phagocytes. Together, OMGP-directed autoimmunity provides a new element of heterogeneity, helping to improve the stratification of patients for diagnostic and therapeutic purposes.
- Published
- 2020
- Full Text
- View/download PDF
14. Butyrophilin-like proteins display combinatorial diversity in selecting and maintaining signature intraepithelial γδ T cell compartments
- Author
-
Anett Jandke, Daisy Melandri, Leticia Monin, Dmitry S. Ushakov, Adam G. Laing, Pierre Vantourout, Philip East, Takeshi Nitta, Tomoya Narita, Hiroshi Takayanagi, Regina Feederle, and Adrian Hayday
- Subjects
Science - Abstract
Butyrophilin-like genes are emerging as central to tissue associated γδ T cell compartments. Here, the authors show that the butyropilin-like gene-products exert their effects as combinatorially diverse heteromers that differentially affect the selection and maintenance of skin-resident and gut-resident intraepithelial γδ T-cell populations.
- Published
- 2020
- Full Text
- View/download PDF
15. Spt6 is a maintenance factor for centromeric CENP-A
- Author
-
Georg O. M. Bobkov, Anming Huang, Sebastiaan J. W. van den Berg, Sreyoshi Mitra, Eduard Anselm, Vasiliki Lazou, Sarah Schunter, Regina Feederle, Axel Imhof, Alexandra Lusser, Lars E. T. Jansen, and Patrick Heun
- Subjects
Science - Abstract
CENP-A is a stable centromere mark, although active transcription poses a potential threat for retaining CENP-A through chromatin remodeling and nucleosome eviction. Here, the authors show that maintenance of the centromeric mark is preserved by Spt6, which recycles CENP-A nucleosomes.
- Published
- 2020
- Full Text
- View/download PDF
16. Enhancing protective microglial activities with a dual function TREM2 antibody to the stalk region
- Author
-
Kai Schlepckow, Kathryn M Monroe, Gernot Kleinberger, Ludovico Cantuti‐Castelvetri, Samira Parhizkar, Dan Xia, Michael Willem, Georg Werner, Nadine Pettkus, Bettina Brunner, Alice Sülzen, Brigitte Nuscher, Heike Hampel, Xianyuan Xiang, Regina Feederle, Sabina Tahirovic, Joshua I Park, Rachel Prorok, Cathal Mahon, Chun‐Chi Liang, Ju Shi, Do Jin Kim, Hanna Sabelström, Fen Huang, Gilbert Di Paolo, Mikael Simons, Joseph W Lewcock, and Christian Haass
- Subjects
Alzheimer's disease ,amyloid β‐peptide ,microglia ,therapeutic antibody ,TREM2 ,Medicine (General) ,R5-920 ,Genetics ,QH426-470 - Abstract
Abstract Triggering receptor expressed on myeloid cells 2 (TREM2) is essential for the transition of homeostatic microglia to a disease‐associated microglial state. To enhance TREM2 activity, we sought to selectively increase the full‐length protein on the cell surface via reducing its proteolytic shedding by A Disintegrin And Metalloproteinase (i.e., α‐secretase) 10/17. We screened a panel of monoclonal antibodies against TREM2, with the aim to selectively compete for α‐secretase‐mediated shedding. Monoclonal antibody 4D9, which has a stalk region epitope close to the cleavage site, demonstrated dual mechanisms of action by stabilizing TREM2 on the cell surface and reducing its shedding, and concomitantly activating phospho‐SYK signaling. 4D9 stimulated survival of macrophages and increased microglial uptake of myelin debris and amyloid β‐peptide in vitro. In vivo target engagement was demonstrated in cerebrospinal fluid, where nearly all soluble TREM2 was 4D9‐bound. Moreover, in a mouse model for Alzheimer's disease‐related pathology, 4D9 reduced amyloidogenesis, enhanced microglial TREM2 expression, and reduced a homeostatic marker, suggesting a protective function by driving microglia toward a disease‐associated state.
- Published
- 2020
- Full Text
- View/download PDF
17. Active poly‐GA vaccination prevents microglia activation and motor deficits in a C9orf72 mouse model
- Author
-
Qihui Zhou, Nikola Mareljic, Meike Michaelsen, Samira Parhizkar, Steffanie Heindl, Brigitte Nuscher, Daniel Farny, Mareike Czuppa, Carina Schludi, Alexander Graf, Stefan Krebs, Helmut Blum, Regina Feederle, Stefan Roth, Christian Haass, Thomas Arzberger, Arthur Liesz, and Dieter Edbauer
- Subjects
amyotrophic lateral sclerosis ,C9orf72 ,frontotemporal dementia ,immunotherapy ,neurodegeneration ,Medicine (General) ,R5-920 ,Genetics ,QH426-470 - Abstract
Abstract The C9orf72 repeat expansion is the most common genetic cause of amyotrophic lateral sclerosis (ALS) and/or frontotemporal dementia (FTD). Non‐canonical translation of the expanded repeat results in abundant poly‐GA inclusion pathology throughout the CNS. (GA)149‐CFP expression in mice triggers motor deficits and neuroinflammation. Since poly‐GA is transmitted between cells, we investigated the therapeutic potential of anti‐GA antibodies by vaccinating (GA)149‐CFP mice. To overcome poor immunogenicity, we compared the antibody response of multivalent ovalbumin‐(GA)10 conjugates and pre‐aggregated carrier‐free (GA)15. Only ovalbumin‐(GA)10 immunization induced a strong anti‐GA response. The resulting antisera detected poly‐GA aggregates in cell culture and patient tissue. Ovalbumin‐(GA)10 immunization largely rescued the motor function in (GA)149‐CFP transgenic mice and reduced poly‐GA inclusions. Transcriptome analysis showed less neuroinflammation in ovalbumin‐(GA)10‐immunized poly‐GA mice, which was corroborated by semiquantitative and morphological analysis of microglia/macrophages. Moreover, cytoplasmic TDP‐43 mislocalization and levels of the neurofilament light chain in the CSF were reduced, suggesting neuroaxonal damage is reduced. Our data suggest that immunotherapy may be a viable primary prevention strategy for ALS/FTD in C9orf72 mutation carriers.
- Published
- 2019
- Full Text
- View/download PDF
18. Beneficial Effect of ACI-24 Vaccination on Aβ Plaque Pathology and Microglial Phenotypes in an Amyloidosis Mouse Model
- Author
-
Jasenka Rudan Njavro, Marija Vukicevic, Emma Fiorini, Lina Dinkel, Stephan A. Müller, Anna Berghofer, Chiara Bordier, Stanislav Kozlov, Annett Halle, Katrin Buschmann, Anja Capell, Camilla Giudici, Michael Willem, Regina Feederle, Stefan F. Lichtenthaler, Chiara Babolin, Paolo Montanari, Andrea Pfeifer, Marie Kosco-Vilbois, and Sabina Tahirovic
- Subjects
Alzheimer’s disease ,immunotherapy ,microglia ,Aβ vaccine ,ACI-24 ,Cytology ,QH573-671 - Abstract
Amyloid-β (Aβ) deposition is an initiating factor in Alzheimer’s disease (AD). Microglia are the brain immune cells that surround and phagocytose Aβ plaques, but their phagocytic capacity declines in AD. This is in agreement with studies that associate AD risk loci with genes regulating the phagocytic function of immune cells. Immunotherapies are currently pursued as strategies against AD and there are increased efforts to understand the role of the immune system in ameliorating AD pathology. Here, we evaluated the effect of the Aβ targeting ACI-24 vaccine in reducing AD pathology in an amyloidosis mouse model. ACI-24 vaccination elicited a robust and sustained antibody response in APPPS1 mice with an accompanying reduction of Aβ plaque load, Aβ plaque-associated ApoE and dystrophic neurites as compared to non-vaccinated controls. Furthermore, an increased number of NLRP3-positive plaque-associated microglia was observed following ACI-24 vaccination. In contrast to this local microglial activation at Aβ plaques, we observed a more ramified morphology of Aβ plaque-distant microglia compared to non-vaccinated controls. Accordingly, bulk transcriptomic analysis revealed a trend towards the reduced expression of several disease-associated microglia (DAM) signatures that is in line with the reduced Aβ plaque load triggered by ACI-24 vaccination. Our study demonstrates that administration of the Aβ targeting vaccine ACI-24 reduces AD pathology, suggesting its use as a safe and cost-effective AD therapeutic intervention.
- Published
- 2022
- Full Text
- View/download PDF
19. Pathological ASXL1 Mutations and Protein Variants Impair Neural Crest Development
- Author
-
Friederike Matheus, Ejona Rusha, Rizwan Rehimi, Lena Molitor, Anna Pertek, Miha Modic, Regina Feederle, Andrew Flatley, Elisabeth Kremmer, Arie Geerlof, Valentyna Rishko, Alvaro Rada-Iglesias, and Micha Drukker
- Subjects
Medicine (General) ,R5-920 ,Biology (General) ,QH301-705.5 - Abstract
Summary: The neural crest (NC) gives rise to a multitude of fetal tissues, and its misregulation is implicated in congenital malformations. Here, we investigated molecular mechanisms pertaining to NC-related symptoms in Bohring-Opitz syndrome (BOS), a developmental disorder linked to mutations in the Polycomb group factor Additional sex combs-like 1 (ASXL1). Genetically edited human pluripotent stem cell lines that were differentiated to NC progenitors and then xenotransplanted into chicken embryos demonstrated an impairment of NC delamination and emigration. Molecular analysis showed that ASXL1 mutations correlated with reduced activation of the transcription factor ZIC1 and the NC gene regulatory network. These findings were supported by differentiation experiments using BOS patient-derived induced pluripotent stem cell lines. Expression of truncated ASXL1 isoforms (amino acids 1–900) recapitulated the NC phenotypes in vitro and in ovo, raising the possibility that truncated ASXL1 variants contribute to BOS pathology. Collectively, we expand the understanding of truncated ASXL1 in BOS and in the human NC. : In this study, Drukker and colleagues developed human pluripotent stem cell models for the rare congenital disorder Bohring-Opitz syndrome, which is caused by mutations in the Polycomb factor ASXL1. In these lines, they found impaired neural crest emigration in vitro and in vivo and link this phenotype to impaired activation of ZIC1. Keywords: ASXL1, neural crest, Bohring-Opitz syndrome, Polycomb, ZIC1
- Published
- 2019
- Full Text
- View/download PDF
20. FK506-Binding Protein 11 Is a Novel Plasma Cell-Specific Antibody Folding Catalyst with Increased Expression in Idiopathic Pulmonary Fibrosis
- Author
-
Stefan Preisendörfer, Yoshihiro Ishikawa, Elisabeth Hennen, Stephan Winklmeier, Jonas C. Schupp, Larissa Knüppel, Isis E. Fernandez, Leonhard Binzenhöfer, Andrew Flatley, Brenda M. Juan-Guardela, Clemens Ruppert, Andreas Guenther, Marion Frankenberger, Rudolf A. Hatz, Nikolaus Kneidinger, Jürgen Behr, Regina Feederle, Aloys Schepers, Anne Hilgendorff, Naftali Kaminski, Edgar Meinl, Hans Peter Bächinger, Oliver Eickelberg, and Claudia A. Staab-Weijnitz
- Subjects
antibody folding ,immunophilin ,lung fibrosis ,interstitial lung disease ,ER stress ,tacrolimus ,Cytology ,QH573-671 - Abstract
Antibodies are central effectors of the adaptive immune response, widespread used therapeutics, but also potentially disease-causing biomolecules. Antibody folding catalysts in the plasma cell are incompletely defined. Idiopathic pulmonary fibrosis (IPF) is a fatal chronic lung disease with increasingly recognized autoimmune features. We found elevated expression of FK506-binding protein 11 (FKBP11) in IPF lungs where FKBP11 specifically localized to antibody-producing plasma cells. Suggesting a general role in plasma cells, plasma cell-specific FKBP11 expression was equally observed in lymphatic tissues, and in vitro B cell to plasma cell differentiation was accompanied by induction of FKBP11 expression. Recombinant human FKBP11 was able to refold IgG antibody in vitro and inhibited by FK506, strongly supporting a function as antibody peptidyl-prolyl cis-trans isomerase. Induction of ER stress in cell lines demonstrated induction of FKBP11 in the context of the unfolded protein response in an X-box-binding protein 1 (XBP1)-dependent manner. While deficiency of FKBP11 increased susceptibility to ER stress-mediated cell death in an alveolar epithelial cell line, FKBP11 knockdown in an antibody-producing hybridoma cell line neither induced cell death nor decreased expression or secretion of IgG antibody. Similarly, antibody secretion by the same hybridoma cell line was not affected by knockdown of the established antibody peptidyl-prolyl isomerase cyclophilin B. The results are consistent with FKBP11 as a novel XBP1-regulated antibody peptidyl-prolyl cis-trans isomerase and indicate significant redundancy in the ER-resident folding machinery of antibody-producing hybridoma cells.
- Published
- 2022
- Full Text
- View/download PDF
21. A Novel Anti-CD73 Antibody That Selectively Inhibits Membrane CD73 Shows Antitumor Activity and Induces Tumor Immune Escape
- Author
-
Markus Kellner, Bettina von Neubeck, Bastian Czogalla, Regina Feederle, Binje Vick, Irmela Jeremias, and Reinhard Zeidler
- Subjects
CD73 ,adenosine ,immune evasion ,extracellular vesicles ,cancer therapy ,therapeutic antibody ,Biology (General) ,QH301-705.5 - Abstract
CD73 catalyzes the conversion of ATP to adenosine, which is involved in various physiological and pathological processes, including tumor immune escape. Because CD73 expression and activity are particularly high on cancer cells and contribute to the immunosuppressive properties of the tumor environment, it is considered an attractive target molecule for specific cancer therapies. In line, several studies demonstrated that CD73 inhibition has a significant antitumor effect. However, complete blocking of CD73 activity can evoke autoimmune phenomena and adverse side effects. We developed a CD73-specific antibody, 22E6, that specifically inhibits the enzymatic activity of membrane-tethered CD73 present in high concentrations on cancer cells and cancer cell-derived extracellular vesicles but has no inhibitory effect on soluble CD73. Inhibition of CD73 on tumor cells with 22E6 resulted in multiple effects on tumor cells in vitro, including increased apoptosis and interference with chemoresistance. Intriguingly, in a xenograft mouse model of acute lymphocytic leukemia (ALL), 22E6 treatment resulted in an initial tumor growth delay in some animals, followed by a complete loss of CD73 expression on ALL cells in all 22E6 treated animals, indicating tumor immune escape. Taken together, 22E6 shows great potential for cancer therapy, favorably in combination with other drugs.
- Published
- 2022
- Full Text
- View/download PDF
22. Novel antibody against low‐n oligomers of tau protein promotes clearance of tau in cells via lysosomes
- Author
-
Ram Reddy Chandupatla, Andrew Flatley, Regina Feederle, Eva‐Maria Mandelkow, and Senthilvelrajan Kaniyappan
- Subjects
aggregation ,antibody ,N2a cells ,screening ,tau ,Neurology. Diseases of the nervous system ,RC346-429 ,Geriatrics ,RC952-954.6 - Abstract
Abstract Introduction Tau, a natively unfolded soluble protein, forms abnormal oligomers and insoluble filaments in several neurodegenerative diseases, including Alzheimer disease (AD). Tau‐induced toxicity is mainly due to oligomers rather than monomers or fibrils. Methods We have developed monoclonal antibodies against purified low‐n tau oligomers of the tau repeat domain as a tool to neutralize tau aggregation and toxicity. In vitro aggregation inhibition was tested by thioflavin S, dynamic light scattering (DLS), and atomic force microscopy (AFM). Using a split‐luciferase complementation assay and fluorescence‐activated cell sorting (FACS), the inhibition of aggregation was analyzed in an N2a cell model of tauopathy. Results Antibodies inhibited tau aggregation in vitro up to ~90% by blocking tau at an oligomeric state. Some antibodies were able to block tau dimerization/oligomerization in cells, as measured by a split‐luciferase complementation assay. Antibodies applied extracellularly were internalized and led to sequestration of tau into lysosomes for degradation. Discussion Novel low‐n tau oligomer specific monoclonal antibody inhibits Tau oligomerization in cells and promotes toxic tau clearance.
- Published
- 2020
- Full Text
- View/download PDF
23. An Antibody-Aptamer-Hybrid Lateral Flow Assay for Detection of CXCL9 in Antibody-Mediated Rejection after Kidney Transplantation
- Author
-
Lisa K. Seiler, Ngoc Linh Phung, Christoph Nikolin, Stephan Immenschuh, Christian Erck, Jessica Kaufeld, Hermann Haller, Christine S. Falk, Rebecca Jonczyk, Patrick Lindner, Stefanie Thoms, Julia Siegl, Günter Mayer, Regina Feederle, and Cornelia A. Blume
- Subjects
biomarkers ,antibody-mediated rejection (AMR) ,neural net analysis ,CXCL9 (MIG) ,aptamer ,antibody ,Medicine (General) ,R5-920 - Abstract
Chronic antibody-mediated rejection (AMR) is a key limiting factor for the clinical outcome of a kidney transplantation (Ktx), where early diagnosis and therapeutic intervention is needed. This study describes the identification of the biomarker CXC-motif chemokine ligand (CXCL) 9 as an indicator for AMR and presents a new aptamer-antibody-hybrid lateral flow assay (hybrid-LFA) for detection in urine. Biomarker evaluation included two independent cohorts of kidney transplant recipients (KTRs) from a protocol biopsy program and used subgroup comparisons according to BANFF-classifications. Plasma, urine and biopsy lysate samples were analyzed with a Luminex-based multiplex assay. The CXCL9-specific hybrid-LFA was developed based upon a specific rat antibody immobilized on a nitrocellulose-membrane and the coupling of a CXCL9-binding aptamer to gold nanoparticles. LFA performance was assessed according to receiver operating characteristic (ROC) analysis. Among 15 high-scored biomarkers according to a neural network analysis, significantly higher levels of CXCL9 were found in plasma and urine and biopsy lysates of KTRs with biopsy-proven AMR. The newly developed hybrid-LFA reached a sensitivity and specificity of 71% and an AUC of 0.79 for CXCL9. This point-of-care-test (POCT) improves early diagnosis-making in AMR after Ktx, especially in KTRs with undetermined status of donor-specific HLA-antibodies.
- Published
- 2022
- Full Text
- View/download PDF
24. Genomic Location of PRMT6-Dependent H3R2 Methylation Is Linked to the Transcriptional Outcome of Associated Genes
- Author
-
Caroline Bouchard, Peeyush Sahu, Marion Meixner, René Reiner Nötzold, Marco B. Rust, Elisabeth Kremmer, Regina Feederle, Gene Hart-Smith, Florian Finkernagel, Marek Bartkuhn, Soni Savai Pullamsetti, Andrea Nist, Thorsten Stiewe, Sjaak Philipsen, and Uta-Maria Bauer
- Subjects
Biology (General) ,QH301-705.5 - Abstract
Summary: Protein arginine methyltransferase 6 (PRMT6) catalyzes asymmetric dimethylation of histone H3 at arginine 2 (H3R2me2a). This mark has been reported to associate with silent genes. Here, we use a cell model of neural differentiation, which upon PRMT6 knockout exhibits proliferation and differentiation defects. Strikingly, we detect PRMT6-dependent H3R2me2a at active genes, both at promoter and enhancer sites. Loss of H3R2me2a from promoter sites leads to enhanced KMT2A binding and H3K4me3 deposition together with increased target gene transcription, supporting a repressive nature of H3R2me2a. At enhancers, H3R2me2a peaks co-localize with the active enhancer marks H3K4me1 and H3K27ac. Here, loss of H3R2me2a results in reduced KMT2D binding and H3K4me1/H3K27ac deposition together with decreased transcription of associated genes, indicating that H3R2me2a also exerts activation functions. Our work suggests that PRMT6 via H3R2me2a interferes with the deposition of adjacent histone marks and modulates the activity of important differentiation-associated genes by opposing transcriptional effects. : Bouchard et al. identify the genome-wide, PRMT6-dependent occurrence of H3R2me2a in a cell model of neural differentiation. H3R2me2a is localized at promoters and enhancers of active genes and influences the chromatin recruitment of histone lysine methyltransferases. Thereby, H3R2me2a modulates the deposition of adjacent histone H3 marks and regulates the transcriptional output of genes relevant for pluripotency and differentiation. Keywords: protein arginine methyltransferases, histone modifications, posttranslational modifications, histone code, histone arginine methylation, chromatin, transcriptional regulation, gene expression, pluripotency, neural differentiation
- Published
- 2018
- Full Text
- View/download PDF
25. Novel antibodies reveal presynaptic localization of C9orf72 protein and reduced protein levels in C9orf72 mutation carriers
- Author
-
Petra Frick, Chantal Sellier, Ian R. A. Mackenzie, Chieh-Yu Cheng, Julie Tahraoui-Bories, Cecile Martinat, R. Jeroen Pasterkamp, Johannes Prudlo, Dieter Edbauer, Mustapha Oulad-Abdelghani, Regina Feederle, Nicolas Charlet-Berguerand, and Manuela Neumann
- Subjects
Frontotemporal dementia ,Frontotemporal lobar degeneration ,Amyotrophic lateral sclerosis ,C9orf72 ,RAB3 ,Synaptic vesicles ,Neurology. Diseases of the nervous system ,RC346-429 - Abstract
Abstract Hexanucleotide repeat expansion in C9orf72 is the most common genetic cause of frontotemporal dementia and amyotrophic lateral sclerosis, but the pathogenic mechanism of this mutation remains unresolved. Haploinsufficiency has been proposed as one potential mechanism. However, insights if and how reduced C9orf72 proteins levels might contribute to disease pathogenesis are still limited because C9orf72 expression, localization and functions in the central nervous system (CNS) are uncertain, in part due to the poor specificity of currently available C9orf72 antibodies. Here, we generated and characterized novel knock-out validated monoclonal rat and mouse antibodies against C9orf72. We found that C9orf72 is a low abundant, cytoplasmic, highly soluble protein with the long 481 amino acid isoform being the predominant, if not exclusively, expressed protein isoform in mouse tissues and human brain. As consequence of the C9orf72 repeat expansion, C9orf72 protein levels in the cerebellum were reduced to 80% in our series of C9orf72 mutation carriers (n = 17) compared to controls (n = 26). However, no associations between cerebellar protein levels and clinical phenotypes were seen. Finally, by utilizing complementary immunohistochemical and biochemical approaches including analysis of human iPSC derived motor neurons, we identified C9orf72, in addition to its association to lysosomes, to be localized to the presynapses and able to interact with all members of the RAB3 protein family, suggestive of a role for C9orf72 in regulating synaptic vesicle functions by potentially acting as guanine nucleotide exchange factor for RAB3 proteins. In conclusion, our findings provide further evidence for haploinsufficiency as potential mechanism in C9orf72 pathogenesis by demonstrating reduced protein levels in C9orf72 mutation carriers and important novel insights into the physiological role of C9orf72 in the CNS. Moreover, the described novel monoclonal C9orf72 antibodies will be useful tools to further dissect the cellular and molecular functions of C9orf72.
- Published
- 2018
- Full Text
- View/download PDF
26. Nucleolar-nucleoplasmic shuttling of TARG1 and its control by DNA damage-induced poly-ADP-ribosylation and by nucleolar transcription
- Author
-
Mareike Bütepage, Christian Preisinger, Alexander von Kriegsheim, Anja Scheufen, Eva Lausberg, Jinyu Li, Ferdinand Kappes, Regina Feederle, Sabrina Ernst, Laura Eckei, Sarah Krieg, Gerhard Müller-Newen, Giulia Rossetti, Karla L. H. Feijs, Patricia Verheugd, and Bernhard Lüscher
- Subjects
Medicine ,Science - Abstract
Abstract Macrodomains are conserved protein folds associated with ADP-ribose binding and turnover. ADP-ribosylation is a posttranslational modification catalyzed primarily by ARTD (aka PARP) enzymes in cells. ARTDs transfer either single or multiple ADP-ribose units to substrates, resulting in mono- or poly-ADP-ribosylation. TARG1/C6orf130 is a macrodomain protein that hydrolyzes mono-ADP-ribosylation and interacts with poly-ADP-ribose chains. Interactome analyses revealed that TARG1 binds strongly to ribosomes and proteins associated with rRNA processing and ribosomal assembly factors. TARG1 localized to transcriptionally active nucleoli, which occurred independently of ADP-ribose binding. TARG1 shuttled continuously between nucleoli and nucleoplasm. In response to DNA damage, which activates ARTD1/2 (PARP1/2) and promotes synthesis of poly-ADP-ribose chains, TARG1 re-localized to the nucleoplasm. This was dependent on the ability of TARG1 to bind to poly-ADP-ribose. These findings are consistent with the observed ability of TARG1 to competitively interact with RNA and PAR chains. We propose a nucleolar role of TARG1 in ribosome assembly or quality control that is stalled when TARG1 is re-located to sites of DNA damage.
- Published
- 2018
- Full Text
- View/download PDF
27. Binding of NUFIP2 to Roquin promotes recognition and regulation of ICOS mRNA
- Author
-
Nina Rehage, Elena Davydova, Christine Conrad, Gesine Behrens, Andreas Maiser, Jenny E. Stehklein, Sven Brenner, Juliane Klein, Aicha Jeridi, Anne Hoffmann, Eunhae Lee, Umberto Dianzani, Rob Willemsen, Regina Feederle, Kristin Reiche, Jörg Hackermüller, Heinrich Leonhardt, Sonia Sharma, Dierk Niessing, and Vigo Heissmeyer
- Subjects
Science - Abstract
The RNA-binding proteins Roquin-1 and Roquin-2 are essential for immune cell function and postnatal survival in mice. Here, the authors identify NUFIP2 as a cofactor of Roquin; Roquin binds and stabilizes NUFIP2 in cells while NUFIP2 regulates Roquin mRNA target recognition.
- Published
- 2018
- Full Text
- View/download PDF
28. An Alzheimer‐associated TREM2 variant occurs at the ADAM cleavage site and affects shedding and phagocytic function
- Author
-
Kai Schlepckow, Gernot Kleinberger, Akio Fukumori, Regina Feederle, Stefan F Lichtenthaler, Harald Steiner, and Christian Haass
- Subjects
Alzheimer's disease ,neurodegeneration ,phagocytosis ,regulated intramembrane proteolysis ,TREM2 ,Medicine (General) ,R5-920 ,Genetics ,QH426-470 - Abstract
Abstract Sequence variations occurring in the gene encoding the triggering receptor expressed on myeloid cells 2 (TREM2) support an essential function of microglia and innate immunity in the pathogenesis of Alzheimer's disease (AD) and other neurodegenerative disorders. TREM2 matures within the secretory pathway, and its ectodomain is shed on the plasma membrane. Missense mutations in the immunoglobulin (Ig)‐like domain such as p.T66M and p.Y38C retain TREM2 within the endoplasmic reticulum and reduce shedding as well as TREM2‐dependent phagocytosis. Using mass spectrometry, we have now determined the cleavage site of TREM2. TREM2 is shed by proteases of the ADAM (a disintegrin and metalloproteinase domain containing protein) family C‐terminal to histidine 157, a position where an AD‐associated coding variant has been discovered (p.H157Y) in the Han Chinese population. Opposite to the characterized mutations within the Ig‐like domain, such as p.T66M and p.Y38C, the p.H157Y variant within the stalk region leads to enhanced shedding of TREM2. Elevated ectodomain shedding reduces cell surface full‐length TREM2 and lowers TREM2‐dependent phagocytosis. Therefore, two seemingly opposite cellular effects of TREM2 variants, namely reduced versus enhanced shedding, result in similar phenotypic outcomes by reducing cell surface TREM2.
- Published
- 2017
- Full Text
- View/download PDF
29. T cell specific Cxcr5 deficiency prevents rheumatoid arthritis
- Author
-
Georgios L. Moschovakis, Anja Bubke, Michaela Friedrichsen, Christine S. Falk, Regina Feederle, and Reinhold Förster
- Subjects
Medicine ,Science - Abstract
Abstract The chemokine receptor CXCR5 is primarily expressed on B cells and Tfh cells and facilitates their migration towards B cell follicles. In the present study we investigated the role of the CXCL13/CXCR5 axis in the pathogenesis of rheumatoid arthritis (RA) and specifically addressed the impact of CXCR5-mediated T and B cell migration in this disease. Employing collagen-induced arthritis (CIA) we identify CXCR5 as an absolutely essential factor for the induction of inflammatory autoimmune arthritis. Cxcr5-deficient mice and mice selectively lacking Cxcr5 on T cells were completely resistant to CIA, showed impaired germinal center responses and failed to mount an IgG1 antibody response to collagen II. Selective ablation of CXCR5 expression in B cells also led to suppression of CIA owing to diminished GC responses in secondary lymphoid organs (SLO) and impaired anti-collagen II antibody production. Chimeric mice harboring Cxcr5-proficient and Cxcr5-deficient immune cells revealed SLO and not the synovial tissue as the compartment where CXCR5-mediated cell migration induces autoimmune inflammation in arthritis. Thus our data demonstrate that CXCR5-mediated co-localization of Tfh cells and B cells in SLOs is absolutely essential for the induction of RA and identify CXCR5 and Tfh cells as promising therapeutic targets for the treatment of RA.
- Published
- 2017
- Full Text
- View/download PDF
30. Poly‐GP in cerebrospinal fluid links C9orf72‐associated dipeptide repeat expression to the asymptomatic phase of ALS/FTD
- Author
-
Carina Lehmer, Patrick Oeckl, Jochen H Weishaupt, Alexander E Volk, Janine Diehl‐Schmid, Matthias L Schroeter, Martin Lauer, Johannes Kornhuber, Johannes Levin, Klaus Fassbender, Bernhard Landwehrmeyer, German Consortium for Frontotemporal Lobar Degeneration, Martin H Schludi, Thomas Arzberger, Elisabeth Kremmer, Andrew Flatley, Regina Feederle, Petra Steinacker, Patrick Weydt, Albert C Ludolph, Dieter Edbauer, Markus Otto, Adrian Danek, Emily Feneberg, Sarah Anderl‐Straub, Christine von Arnim, Holger Jahn, Anja Schneider, Manuel Maler, Maryna Polyakova, Lina Riedl, Jens Wiltfang, and Georg Ziegler
- Subjects
amyotrophic lateral sclerosis ,biomarker ,C9orf72 ,cerebrospinal fluid ,frontotemporal dementia ,Medicine (General) ,R5-920 ,Genetics ,QH426-470 - Abstract
Abstract The C9orf72 GGGGCC repeat expansion is a major cause of amyotrophic lateral sclerosis and frontotemporal dementia (c9ALS/FTD). Non‐conventional repeat translation results in five dipeptide repeat proteins (DPRs), but their clinical utility, overall significance, and temporal course in the pathogenesis of c9ALS/FTD are unclear, although animal models support a gain‐of‐function mechanism. Here, we established a poly‐GP immunoassay from cerebrospinal fluid (CSF) to identify and characterize C9orf72 patients. Significant poly‐GP levels were already detectable in asymptomatic C9orf72 mutation carriers compared to healthy controls and patients with other neurodegenerative diseases. The poly‐GP levels in asymptomatic carriers were similar to symptomatic c9ALS/FTD cases. Poly‐GP levels were not correlated with disease onset, clinical scores, and CSF levels of neurofilaments as a marker for axonal damage. Poly‐GP determination in CSF revealed a C9orf72 mutation carrier in our cohort and may thus be used as a diagnostic marker in addition to genetic testing to screen patients. Presymptomatic expression of poly‐GP and likely other DPR species may contribute to disease onset and thus represents an alluring therapeutic target.
- Published
- 2017
- Full Text
- View/download PDF
31. Antibodies inhibit transmission and aggregation of C9orf72 poly‐GA dipeptide repeat proteins
- Author
-
Qihui Zhou, Carina Lehmer, Meike Michaelsen, Kohji Mori, Dominik Alterauge, Dirk Baumjohann, Martin H Schludi, Johanna Greiling, Daniel Farny, Andrew Flatley, Regina Feederle, Stephanie May, Franziska Schreiber, Thomas Arzberger, Christoph Kuhm, Thomas Klopstock, Andreas Hermann, Christian Haass, and Dieter Edbauer
- Subjects
amyotrophic lateral sclerosis ,C9orf72 ,immunotherapy ,RAN translation ,seeding ,Medicine (General) ,R5-920 ,Genetics ,QH426-470 - Abstract
Abstract Cell‐to‐cell transmission of protein aggregates is an emerging theme in neurodegenerative disease. Here, we analyze the dipeptide repeat (DPR) proteins that form neuronal inclusions in patients with hexanucleotide repeat expansion C9orf72, the most common known cause of amyotrophic lateral sclerosis (ALS) and frontotemporal lobar degeneration (FTLD). Sense and antisense transcripts of the (G4C2)n repeat are translated by repeat‐associated non‐ATG (RAN) translation in all reading frames into five aggregating DPR proteins. We show that the hydrophobic DPR proteins poly‐GA, poly‐GP, and poly‐PA are transmitted between cells using co‐culture assays and cell extracts. Moreover, uptake or expression of poly‐GA induces nuclear RNA foci in (G4C2)80‐expressing cells and patient fibroblasts, suggesting an unexpected positive feedback loop. Exposure to recombinant poly‐GA and cerebellar extracts of C9orf72 patients increases repeat RNA levels and seeds aggregation of all DPR proteins in receiver cells expressing (G4C2)80. Treatment with anti‐GA antibodies inhibits intracellular poly‐GA aggregation and blocks the seeding activity of C9orf72 brain extracts. Poly‐GA‐directed immunotherapy may thus reduce DPR aggregation and disease progression in C9orf72 ALS/FTD.
- Published
- 2017
- Full Text
- View/download PDF
32. Epstein–Barr virus particles induce centrosome amplification and chromosomal instability
- Author
-
Anatoliy Shumilov, Ming-Han Tsai, Yvonne T. Schlosser, Anne-Sophie Kratz, Katharina Bernhardt, Susanne Fink, Tuba Mizani, Xiaochen Lin, Anna Jauch, Josef Mautner, Annette Kopp-Schneider, Regina Feederle, Ingrid Hoffmann, and Henri-Jacques Delecluse
- Subjects
Science - Abstract
Infection with Epstein–Barr virus (EBV) is associated with increased risk of cancer development. Here the authors show that EBV particles, and more specifically the viral protein BNRF1, induce centrosome amplification and chromosomal instability in host cells in the absence of chronic infection.
- Published
- 2017
- Full Text
- View/download PDF
33. MALT1 Phosphorylation Controls Activation of T Lymphocytes and Survival of ABC-DLBCL Tumor Cells
- Author
-
Torben Gehring, Tabea Erdmann, Marco Rahm, Carina Graß, Andrew Flatley, Thomas J. O’Neill, Simone Woods, Isabel Meininger, Ozge Karayel, Kerstin Kutzner, Michael Grau, Hisaaki Shinohara, Katja Lammens, Regina Feederle, Stefanie M. Hauck, Georg Lenz, and Daniel Krappmann
- Subjects
Biology (General) ,QH301-705.5 - Abstract
Summary: The CARMA1/CARD11-BCL10-MALT1 (CBM) complex bridges T and B cell antigen receptor (TCR/BCR) ligation to MALT1 protease activation and canonical nuclear factor κB (NF-κB) signaling. Using unbiased mass spectrometry, we discover multiple serine phosphorylation sites in the MALT1 C terminus after T cell activation. Phospho-specific antibodies reveal that CBM-associated MALT1 is transiently hyper-phosphorylated upon TCR/CD28 co-stimulation. We identify a dual role for CK1α as a kinase that is essential for CBM signalosome assembly as well as MALT1 phosphorylation. Although MALT1 phosphorylation is largely dispensable for protease activity, it fosters canonical NF-κB signaling in Jurkat and murine CD4 T cells. Moreover, constitutive MALT1 phosphorylation promotes survival of activated B cell-type diffuse large B cell lymphoma (ABC-DLBCL) cells addicted to chronic BCR signaling. Thus, MALT1 phosphorylation triggers optimal NF-κB activation in lymphocytes and survival of lymphoma cells. : Gehring et al. identify MALT1 phosphorylation as a process that bridges antigen receptor ligation to downstream signaling pathways in T cells, promotes T lymphocyte activation, and drives survival of B cell lymphoma tumor cells. Keywords: immune response, adaptive immunity, antigen receptor signaling, T cell activation, B cell lymphomas, CBM complex, phosphorylation, NF-kappa B, MALT1, casein kinase 1 alpha
- Published
- 2019
- Full Text
- View/download PDF
34. The Cdk8/19-cyclin C transcription regulator functions in genome replication through metazoan Sld7.
- Author
-
Kerstin Köhler, Luis Sanchez-Pulido, Verena Höfer, Anika Marko, Chris P Ponting, Ambrosius P Snijders, Regina Feederle, Aloys Schepers, and Dominik Boos
- Subjects
Biology (General) ,QH301-705.5 - Abstract
Accurate genome duplication underlies genetic homeostasis. Metazoan Mdm2 binding protein (MTBP) forms a main regulatory platform for origin firing together with Treslin/TICRR and TopBP1 (Topoisomerase II binding protein 1 (TopBP1)-interacting replication stimulating protein/TopBP1-interacting checkpoint and replication regulator). We report the first comprehensive analysis of MTBP and reveal conserved and metazoa-specific MTBP functions in replication. This suggests that metazoa have evolved specific molecular mechanisms to adapt replication principles conserved with yeast to the specific requirements of the more complex metazoan cells. We uncover one such metazoa-specific process: a new replication factor, cyclin-dependent kinase 8/19-cyclinC (Cdk8/19-cyclin C), binds to a central domain of MTBP. This interaction is required for complete genome duplication in human cells. In the absence of MTBP binding to Cdk8/19-cyclin C, cells enter mitosis with incompletely duplicated chromosomes, and subsequent chromosome segregation occurs inaccurately. Using remote homology searches, we identified MTBP as the metazoan orthologue of yeast synthetic lethal with Dpb11 7 (Sld7). This homology finally demonstrates that the set of yeast core factors sufficient for replication initiation in vitro is conserved in metazoa. MTBP and Sld7 contain two homologous domains that are present in no other protein, one each in the N and C termini. In MTBP the conserved termini flank the metazoa-specific Cdk8/19-cyclin C binding region and are required for normal origin firing in human cells. The N termini of MTBP and Sld7 share an essential origin firing function, the interaction with Treslin/TICRR or its yeast orthologue Sld3, respectively. The C termini may function as homodimerisation domains. Our characterisation of broadly conserved and metazoa-specific initiation processes sets the basis for further mechanistic dissection of replication initiation in vertebrates. It is a first step in understanding the distinctions of origin firing in higher eukaryotes.
- Published
- 2019
- Full Text
- View/download PDF
35. Plk1/Polo Phosphorylates Sas-4 at the Onset of Mitosis for an Efficient Recruitment of Pericentriolar Material to Centrosomes
- Author
-
Anand Ramani, Aruljothi Mariappan, Marco Gottardo, Sunit Mandad, Henning Urlaub, Tomer Avidor-Reiss, Maria Riparbelli, Giuliano Callaini, Alain Debec, Regina Feederle, and Jay Gopalakrishnan
- Subjects
Biology (General) ,QH301-705.5 - Abstract
Summary: Centrosomes are the major microtubule-organizing centers, consisting of centrioles surrounded by a pericentriolar material (PCM). Centrosomal PCM is spatiotemporally regulated to be minimal during interphase and expands as cells enter mitosis. It is unclear how PCM expansion is initiated at the onset of mitosis. Here, we identify that, in Drosophila, Plk1/Polo kinase phosphorylates the conserved centrosomal protein Sas-4 in vitro. This phosphorylation appears to occur at the onset of mitosis, enabling Sas-4’s localization to expand outward from meiotic and mitotic centrosomes. The Plk1/Polo kinase site of Sas-4 is then required for an efficient recruitment of Cnn and γ-tubulin, bona fide PCM proteins that are essential for PCM expansion and centrosome maturation. Point mutations at Plk1/Polo sites of Sas-4 affect neither centrosome structure nor centriole duplication but specifically reduce the affinity to bind Cnn and γ-tubulin. These observations identify Plk1/Polo kinase regulation of Sas-4 as essential for efficient PCM expansion. : Ramani et al. show that Plk1/Polo phosphorylates Drosophila Sas-4 at the onset of mitosis. Cell-cycle-specific modification of Sas-4 determines the spatiotemporal localization of Sas-4 in centrosomes, which is required for an efficient recruitment of PCM proteins in mitosis. Keywords: pericentriolar material, centrosomes, Sas-4, Drosophila melanogaster, Plk1, centrosome maturation
- Published
- 2018
- Full Text
- View/download PDF
36. TREM2 deficiency reduces the efficacy of immunotherapeutic amyloid clearance
- Author
-
Xianyuan Xiang, Georg Werner, Bernd Bohrmann, Arthur Liesz, Fargol Mazaheri, Anja Capell, Regina Feederle, Irene Knuesel, Gernot Kleinberger, and Christian Haass
- Subjects
Alzheimer's disease ,immunotherapy ,neurodegeneration ,phagocytosis ,TREM2 ,Medicine (General) ,R5-920 ,Genetics ,QH426-470 - Abstract
Abstract Immunotherapeutic approaches are currently the most advanced treatments for Alzheimer's disease (AD). Antibodies against amyloid β‐peptide (Aβ) bind to amyloid plaques and induce their clearance by microglia via Fc receptor‐mediated phagocytosis. Dysfunctions of microglia may play a pivotal role in AD pathogenesis and could result in reduced efficacy of antibody‐mediated Aβ clearance. Recently, heterozygous mutations in the triggering receptor expressed on myeloid cells 2 (TREM2), a microglial gene involved in phagocytosis, were genetically linked to late onset AD. Loss of TREM2 reduces the ability of microglia to engulf Aβ. We have now investigated whether loss of TREM2 affects the efficacy of immunotherapeutic approaches. We show that anti‐Aβ antibodies stimulate Aβ uptake and amyloid plaque clearance in a dose‐dependent manner in the presence or absence of TREM2. However, TREM2‐deficient N9 microglial cell lines, macrophages as well as primary microglia showed significantly reduced uptake of antibody‐bound Aβ and as a consequence reduced clearance of amyloid plaques. Titration experiments revealed that reduced efficacy of amyloid plaque clearance by Trem2 knockout cells can be compensated by elevating the concentration of therapeutic antibodies.
- Published
- 2016
- Full Text
- View/download PDF
37. Human Natural Killer Cells Prevent Infectious Mononucleosis Features by Targeting Lytic Epstein-Barr Virus Infection
- Author
-
Obinna Chijioke, Anne Müller, Regina Feederle, Mario Henrique M. Barros, Carsten Krieg, Vanessa Emmel, Emanuela Marcenaro, Carol S. Leung, Olga Antsiferova, Vanessa Landtwing, Walter Bossart, Alessandro Moretta, Rocio Hassan, Onur Boyman, Gerald Niedobitek, Henri-Jacques Delecluse, Riccarda Capaul, and Christian Münz
- Subjects
Biology (General) ,QH301-705.5 - Abstract
Primary infection with the human oncogenic Epstein-Barr virus (EBV) can result in infectious mononucleosis (IM), a self-limiting disease caused by massive lymphocyte expansion that predisposes for the development of distinct EBV-associated lymphomas. Why some individuals experience this symptomatic primary EBV infection, whereas the majority acquires the virus asymptomatically, remains unclear. Using a mouse model with reconstituted human immune system components, we show that depletion of human natural killer (NK) cells enhances IM symptoms and promotes EBV-associated tumorigenesis mainly because of a loss of immune control over lytic EBV infection. These data suggest that failure of innate immune control by human NK cells augments symptomatic lytic EBV infection, which drives lymphocyte expansion and predisposes for EBV-associated malignancies.
- Published
- 2013
- Full Text
- View/download PDF
38. Spontaneous Lytic Replication and Epitheliotropism Define an Epstein-Barr Virus Strain Found in Carcinomas
- Author
-
Ming-Han Tsai, Ana Raykova, Olaf Klinke, Katharina Bernhardt, Kathrin Gärtner, Carol S. Leung, Karsten Geletneky, Serkan Sertel, Christian Münz, Regina Feederle, and Henri-Jacques Delecluse
- Subjects
Biology (General) ,QH301-705.5 - Abstract
The Epstein-Barr virus (EBV) is found in a variety of tumors whose incidence greatly varies around the world. A poorly explored hypothesis is that particular EBV strains account for this phenomenon. We report that M81, a virus isolated from a Chinese patient with nasopharyngeal carcinoma (NPC), shows remarkable similarity to other NPC viruses but is divergent from all other known strains. M81 exhibited a reversed tropism relative to common strains with a reduced ability to infect B cells and a high propensity to infect epithelial cells, which is in agreement with its isolation from carcinomas. M81 spontaneously replicated in B cells in vitro and in vivo at unusually high levels, in line with the enhanced viral replication observed in NPC patients. Spontaneous replication and epitheliotropism could be partly ascribed to polymorphisms within viral proteins. We suggest considering M81 and its closely related isolates as an EBV subtype with enhanced pathogenic potential.
- Published
- 2013
- Full Text
- View/download PDF
39. A Viral microRNA Cluster Regulates the Expression of PTEN, p27 and of a bcl-2 Homolog.
- Author
-
Katharina Bernhardt, Janina Haar, Ming-Han Tsai, Remy Poirey, Regina Feederle, and Henri-Jacques Delecluse
- Subjects
Immunologic diseases. Allergy ,RC581-607 ,Biology (General) ,QH301-705.5 - Abstract
The Epstein-Barr virus (EBV) infects and transforms B-lymphocytes with high efficiency. This process requires expression of the viral latent proteins and of the 3 miR-BHRF1 microRNAs. Here we show that B-cells infected by a virus that lacks these non-coding RNAs (Δ123) grew more slowly between day 5 and day 20, relative to wild type controls. This effect could be ascribed to a reduced S phase entry combined with a moderately increased apoptosis rate. Whilst the first phenotypic trait was consistent with an enhanced PTEN expression in B-cells infected with Δ123, the second could be explained by very low BHRF1 protein and RNA levels in the same cells. Indeed, B-cells infected either by a recombinant virus that lacks the BHRF1 protein, a viral bcl-2 homolog, or by Δ123 underwent a similar degree of apoptosis, whereas knockouts of both BHRF1 microRNAs and protein proved transformation-incompetent. We find that that the miR-BHRF1-3 seed regions, and to a lesser extent those of miR-BHRF1-2 mediate these stimulatory effects. After this critical period, B-cells infected with the Δ123 mutant recovered a normal growth rate and became more resistant to provoked apoptosis. This resulted from an enhanced BHRF1 protein expression relative to cells infected with wild type viruses and correlated with decreased p27 expression, two pro-oncogenic events. The upregulation of BHRF1 can be explained by the observation that large BHRF1 mRNAs are the source of BHRF1 protein but are destroyed following BHRF1 microRNA processing, in particular of miR-BHRF1-2. The BHRF1 microRNAs are unlikely to directly target p27 but their absence may facilitate the selection of B-cells that express low levels of this protein. Thus, the BHRF1 microRNAs allowed a time-restricted expression of the BHRF1 protein to innocuously expand the virus B-cell reservoir during the first weeks post-infection without increasing long-term immune pressure.
- Published
- 2016
- Full Text
- View/download PDF
40. The Epstein-Barr Virus BART miRNA Cluster of the M81 Strain Modulates Multiple Functions in Primary B Cells.
- Author
-
Xiaochen Lin, Ming-Han Tsai, Anatoliy Shumilov, Remy Poirey, Helmut Bannert, Jaap M Middeldorp, Regina Feederle, and Henri-Jacques Delecluse
- Subjects
Immunologic diseases. Allergy ,RC581-607 ,Biology (General) ,QH301-705.5 - Abstract
The Epstein-Barr virus (EBV) is a B lymphotropic virus that infects the majority of the human population. All EBV strains transform B lymphocytes, but some strains, such as M81, also induce spontaneous virus replication. EBV encodes 22 microRNAs (miRNAs) that form a cluster within the BART region of the virus and have been previously been found to stimulate tumor cell growth. Here we describe their functions in B cells infected by M81. We found that the BART miRNAs are downregulated in replicating cells, and that exposure of B cells in vitro or in vivo in humanized mice to a BART miRNA knockout virus resulted in an increased proportion of spontaneously replicating cells, relative to wild type virus. The BART miRNAs subcluster 1, and to a lesser extent subcluster 2, prevented expression of BZLF1, the key protein for initiation of lytic replication. Thus, multiple BART miRNAs cooperate to repress lytic replication. The BART miRNAs also downregulated pro- and anti-apoptotic mediators such as caspase 3 and LMP1, and their deletion did not sensitize B-cells to apoptosis. To the contrary, the majority of humanized mice infected with the BART miRNA knockout mutant developed tumors more rapidly, probably due to enhanced LMP1 expression, although deletion of the BART miRNAs did not modify the virus transforming abilities in vitro. This ability to slow cell growth could be confirmed in non-humanized immunocompromized mice. Injection of resting B cells exposed to a virus that lacks the BART miRNAs resulted in accelerated tumor growth, relative to wild type controls. Therefore, we found that the M81 BART miRNAs do not enhance B-cell tumorigenesis but rather repress it. The repressive effects of the BART miRNAs on potentially pathogenic viral functions in infected B cells are likely to facilitate long-term persistence of the virus in the infected host.
- Published
- 2015
- Full Text
- View/download PDF
41. KIT Mutation and Loss of 14q May Be Sufficient for the Development of Clinically Symptomatic Very Low-Risk GIST.
- Author
-
Olaf Karl Klinke, Tuba Mizani, Gouri Baldwin, Brigitte Bancel, Mojgan Devouassoux-Shisheboran, Jean-Yves Scoazec, Pierre-Paul Bringuier, Regina Feederle, Anna Jauch, Katrin Hinderhofer, Philippe Taniere, and Henri-Jacques Delecluse
- Subjects
Medicine ,Science - Abstract
The aim of this study was to determine the minimal set of genetic alterations required for the development of a very low risk clinically symptomatic gastro-intestinal stromal tumour within the stomach wall. We studied the genome of a very low-risk gastric gastro-intestinal stromal tumour by whole-genome sequencing, comparative genomic hybridisation and methylation profiling. The studied tumour harboured two typical genomic lesions: loss of the long arm of chromosome 14 and an activating mutation in exon 11 of KIT. Besides these genetic lesions, only two point mutations that may affect tumour progression were identified: A frame-shift deletion in RNF146 and a missense mutation in a zinc finger of ZNF407. Whilst the frameshift deletion in RNF146 seemed to be restricted to this particular tumour, a similar yet germline mutation in ZNF407 was found in a panel of 52 gastro-intestinal stromal tumours from different anatomical sites and different categories. Germline polymorphisms in the mitotic checkpoint proteins Aurora kinase A and BUB1 kinase B may have furthered tumour growth. The epigenetic profile of the tumour matches that of other KIT-mutant tumours. We have identified mutations in three genes and loss of the long arm of chromosome 14 as the so far minimal set of genetic abnormalities sufficient for the development of a very low risk clinically symptomatic gastric stromal tumour.
- Published
- 2015
- Full Text
- View/download PDF
42. The viral and cellular microRNA targetome in lymphoblastoid cell lines.
- Author
-
Rebecca L Skalsky, David L Corcoran, Eva Gottwein, Christopher L Frank, Dong Kang, Markus Hafner, Jeffrey D Nusbaum, Regina Feederle, Henri-Jacques Delecluse, Micah A Luftig, Thomas Tuschl, Uwe Ohler, and Bryan R Cullen
- Subjects
Immunologic diseases. Allergy ,RC581-607 ,Biology (General) ,QH301-705.5 - Abstract
Epstein-Barr virus (EBV) is a ubiquitous human herpesvirus linked to a number of B cell cancers and lymphoproliferative disorders. During latent infection, EBV expresses 25 viral pre-microRNAs (miRNAs) and induces the expression of specific host miRNAs, such as miR-155 and miR-21, which potentially play a role in viral oncogenesis. To date, only a limited number of EBV miRNA targets have been identified; thus, the role of EBV miRNAs in viral pathogenesis and/or lymphomagenesis is not well defined. Here, we used photoactivatable ribonucleoside-enhanced crosslinking and immunoprecipitation (PAR-CLIP) combined with deep sequencing and computational analysis to comprehensively examine the viral and cellular miRNA targetome in EBV strain B95-8-infected lymphoblastoid cell lines (LCLs). We identified 7,827 miRNA-interaction sites in 3,492 cellular 3'UTRs. 531 of these sites contained seed matches to viral miRNAs. 24 PAR-CLIP-identified miRNA:3'UTR interactions were confirmed by reporter assays. Our results reveal that EBV miRNAs predominantly target cellular transcripts during latent infection, thereby manipulating the host environment. Furthermore, targets of EBV miRNAs are involved in multiple cellular processes that are directly relevant to viral infection, including innate immunity, cell survival, and cell proliferation. Finally, we present evidence that myc-regulated host miRNAs from the miR-17/92 cluster can regulate latent viral gene expression. This comprehensive survey of the miRNA targetome in EBV-infected B cells represents a key step towards defining the functions of EBV-encoded miRNAs, and potentially, identifying novel therapeutic targets for EBV-associated malignancies.
- Published
- 2012
- Full Text
- View/download PDF
43. Epstein-Barr virus infection of naïve B cells in vitro frequently selects clones with mutated immunoglobulin genotypes: implications for virus biology.
- Author
-
Emily Heath, Noelia Begue-Pastor, Sridhar Chaganti, Debbie Croom-Carter, Claire Shannon-Lowe, Dieter Kube, Regina Feederle, Henri-Jacques Delecluse, Alan B Rickinson, and Andrew I Bell
- Subjects
Immunologic diseases. Allergy ,RC581-607 ,Biology (General) ,QH301-705.5 - Abstract
Epstein-Barr virus (EBV), a lymphomagenic human herpesvirus, colonises the host through polyclonal B cell-growth-transforming infections yet establishes persistence only in IgD⁺ CD27⁺ non-switched memory (NSM) and IgD⁻ CD27⁺ switched memory (SM) B cells, not in IgD⁺ CD27⁻ naïve (N) cells. How this selectivity is achieved remains poorly understood. Here we show that purified N, NSM and SM cell preparations are equally transformable in vitro to lymphoblastoid cells lines (LCLs) that, despite upregulating the activation-induced cytidine deaminase (AID) enzyme necessary for Ig isotype switching and Ig gene hypermutation, still retain the surface Ig phenotype of their parental cells. However, both N- and NSM-derived lines remain inducible to Ig isotype switching by surrogate T cell signals. More importantly, IgH gene analysis of N cell infections revealed two features quite distinct from parallel mitogen-activated cultures. Firstly, following 4 weeks of EBV-driven polyclonal proliferation, individual clonotypes then become increasingly dominant; secondly, in around 35% cases these clonotypes carry Ig gene mutations which both resemble AID products and, when analysed in prospectively-harvested cultures, appear to have arisen by sequence diversification in vitro. Thus EBV infection per se can drive at least some naïve B cells to acquire Ig memory genotypes; furthermore, such cells are often favoured during an LCL's evolution to monoclonality. Extrapolating to viral infections in vivo, these findings could help to explain how EBV-infected cells become restricted to memory B cell subsets and why EBV-driven lymphoproliferative lesions, in primary infection and/or immunocompromised settings, so frequently involve clones with memory genotypes.
- Published
- 2012
- Full Text
- View/download PDF
44. A viral microRNA cluster strongly potentiates the transforming properties of a human herpesvirus.
- Author
-
Regina Feederle, Sarah D Linnstaedt, Helmut Bannert, Helge Lips, Maja Bencun, Bryan R Cullen, and Henri-Jacques Delecluse
- Subjects
Immunologic diseases. Allergy ,RC581-607 ,Biology (General) ,QH301-705.5 - Abstract
Epstein-Barr virus (EBV), an oncogenic human herpesvirus, induces cell proliferation after infection of resting B lymphocytes, its reservoir in vivo. The viral latent proteins are necessary for permanent B cell growth, but it is unknown whether they are sufficient. EBV was recently found to encode microRNAs (miRNAs) that are expressed in infected B cells and in some EBV-associated lymphomas. EBV miRNAs are grouped into two clusters located either adjacent to the BHRF1 gene or in introns contained within the viral BART transcripts. To understand the role of the BHRF1 miRNA cluster, we have constructed a virus mutant that lacks all its three members (Δ123) and a revertant virus. Here we show that the B cell transforming capacity of the Δ123 EBV mutant is reduced by more than 20-fold, relative to wild type or revertant viruses. B cells exposed to the knock-out virus displayed slower growth, and exhibited a two-fold reduction in the percentage of cells entering the cell cycle S phase. Furthermore, they displayed higher latent gene expression levels and latent protein production than their wild type counterparts. Therefore, the BHRF1 miRNAs accelerate B cell expansion at lower latent gene expression levels. Thus, this miRNA cluster simultaneously enhances expansion of the virus reservoir and reduces the viral antigenic load, two features that have the potential to facilitate persistence of the virus in the infected host. Thus, the EBV BHRF1 miRNAs may represent new therapeutic targets for the treatment of some EBV-associated lymphomas.
- Published
- 2011
- Full Text
- View/download PDF
45. Expression and processing of a small nucleolar RNA from the Epstein-Barr virus genome.
- Author
-
Roland Hutzinger, Regina Feederle, Jan Mrazek, Natalia Schiefermeier, Piotr J Balwierz, Mihaela Zavolan, Norbert Polacek, Henri-Jacques Delecluse, and Alexander Hüttenhofer
- Subjects
Immunologic diseases. Allergy ,RC581-607 ,Biology (General) ,QH301-705.5 - Abstract
Small nucleolar RNAs (snoRNAs) are localized within the nucleolus, a sub-nuclear compartment, in which they guide ribosomal or spliceosomal RNA modifications, respectively. Up until now, snoRNAs have only been identified in eukaryal and archaeal genomes, but are notably absent in bacteria. By screening B lymphocytes for expression of non-coding RNAs (ncRNAs) induced by the Epstein-Barr virus (EBV), we here report, for the first time, the identification of a snoRNA gene within a viral genome, designated as v-snoRNA1. This genetic element displays all hallmark sequence motifs of a canonical C/D box snoRNA, namely C/C'- as well as D/D'-boxes. The nucleolar localization of v-snoRNA1 was verified by in situ hybridisation of EBV-infected cells. We also confirmed binding of the three canonical snoRNA proteins, fibrillarin, Nop56 and Nop58, to v-snoRNA1. The C-box motif of v-snoRNA1 was shown to be crucial for the stability of the viral snoRNA; its selective deletion in the viral genome led to a complete down-regulation of v-snoRNA1 expression levels within EBV-infected B cells. We further provide evidence that v-snoRNA1 might serve as a miRNA-like precursor, which is processed into 24 nt sized RNA species, designated as v-snoRNA1(24pp). A potential target site of v-snoRNA1(24pp) was identified within the 3'-UTR of BALF5 mRNA which encodes the viral DNA polymerase. V-snoRNA1 was found to be expressed in all investigated EBV-positive cell lines, including lymphoblastoid cell lines (LCL). Interestingly, induction of the lytic cycle markedly up-regulated expression levels of v-snoRNA1 up to 30-fold. By a computational approach, we identified a v-snoRNA1 homolog in the rhesus lymphocryptovirus genome. This evolutionary conservation suggests an important role of v-snoRNA1 during gamma-herpesvirus infection.
- Published
- 2009
- Full Text
- View/download PDF
46. Human Natural Killer Cells Prevent Infectious Mononucleosis Features by Targeting Lytic Epstein-Barr Virus Infection
- Author
-
Obinna Chijioke, Anne Müller, Regina Feederle, Mario Henrique M. Barros, Carsten Krieg, Vanessa Emmel, Emanuela Marcenaro, Carol S. Leung, Olga Antsiferova, Vanessa Landtwing, Walter Bossart, Alessandro Moretta, Rocio Hassan, Onur Boyman, Gerald Niedobitek, Henri-Jacques Delecluse, Riccarda Capaul, and Christian Münz
- Subjects
Biology (General) ,QH301-705.5 - Published
- 2015
- Full Text
- View/download PDF
47. Medin co-aggregates with vascular amyloid-β in Alzheimer’s disease
- Author
-
Jessica Wagner, Karoline Degenhardt, Marleen Veit, Nikolaos Louros, Katerina Konstantoulea, Angelos Skodras, Katleen Wild, Ping Liu, Ulrike Obermüller, Vikas Bansal, Anupriya Dalmia, Lisa M. Häsler, Marius Lambert, Matthias De Vleeschouwer, Hannah A. Davies, Jillian Madine, Deborah Kronenberg-Versteeg, Regina Feederle, Domenico Del Turco, K. Peter R. Nilsson, Tammaryn Lashley, Thomas Deller, Marla Gearing, Lary C. Walker, Peter Heutink, Frederic Rousseau, Joost Schymkowitz, Mathias Jucker, and Jonas J. Neher
- Subjects
PROVIDES ,Medical Biotechnology (with a focus on Cell Biology (including Stem Cell Biology), Molecular Biology, Microbiology, Biochemistry or Biopharmacy) ,metabolism [Amyloid beta-Peptides] ,tau Proteins ,Plaque, Amyloid ,Mice, Transgenic ,Mice ,Amyloid beta-Protein Precursor ,MOUSE MODELS ,Alzheimer Disease ,metabolism [Amyloid beta-Protein Precursor] ,ANGIOPATHY ,Humans ,Animals ,Cognitive Dysfunction ,PEPTIDE ,NETWORK ,BRAIN ,DEPOSITION ,Medicinsk bioteknologi (med inriktning mot cellbiologi (inklusive stamcellsbiologi), molekylärbiologi, mikrobiologi, biokemi eller biofarmaci) ,A-BETA ,Serum Amyloid A Protein ,Amyloid beta-Peptides ,Science & Technology ,Multidisciplinary ,Middle Aged ,metabolism [Plaque, Amyloid] ,metabolism [tau Proteins] ,Multidisciplinary Sciences ,PATHOLOGY ,MICE ,Science & Technology - Other Topics ,ddc:500 ,metabolism [Alzheimer Disease] - Abstract
Aggregates of medin amyloid (a fragment of the protein MFG-E8, also known as lactadherin) are found in the vasculature of almost all humans over 50 years of age1,2, making it the most common amyloid currently known. We recently reported that medin also aggregates in blood vessels of ageing wild-type mice, causing cerebrovascular dysfunction3. Here we demonstrate in amyloid-β precursor protein (APP) transgenic mice and in patients with Alzheimer’s disease that medin co-localizes with vascular amyloid-β deposits, and that in mice, medin deficiency reduces vascular amyloid-β deposition by half. Moreover, in both the mouse and human brain, MFG-E8 is highly enriched in the vasculature and both MFG-E8 and medin levels increase with the severity of vascular amyloid-β burden. Additionally, analysing data from 566 individuals in the ROSMAP cohort, we find that patients with Alzheimer’s disease have higher MFGE8 expression levels, which are attributable to vascular cells and are associated with increased measures of cognitive decline, independent of plaque and tau pathology. Mechanistically, we demonstrate that medin interacts directly with amyloid-β to promote its aggregation, as medin forms heterologous fibrils with amyloid-β, affects amyloid-β fibril structure, and cross-seeds amyloid-β aggregation both in vitro and in vivo. Thus, medin could be a therapeutic target for prevention of vascular damage and cognitive decline resulting from amyloid-β deposition in the blood vessels of the brain. Aggregates of medin amyloid (a fragment of the protein MFG-E8, also known as lactadherin) are found in the vasculature of almost all humans over 50 years of age1,2, making it the most common amyloid currently known. We recently reported that medin also aggregates in blood vessels of ageing wild-type mice, causing cerebrovascular dysfunction3. Here we demonstrate in amyloid-β precursor protein (APP) transgenic mice and in patients with Alzheimer’s disease that medin co-localizes with vascular amyloid-β deposits, and that in mice, medin deficiency reduces vascular amyloid-β deposition by half. Moreover, in both the mouse and human brain, MFG-E8 is highly enriched in the vasculature and both MFG-E8 and medin levels increase with the severity of vascular amyloid-β burden. Additionally, analysing data from 566 individuals in the ROSMAP cohort, we find that patients with Alzheimer’s disease have higher MFGE8 expression levels, which are attributable to vascular cells and are associated with increased measures of cognitive decline, independent of plaque and tau pathology. Mechanistically, we demonstrate that medin interacts directly with amyloid-β to promote its aggregation, as medin forms heterologous fibrils with amyloid-β, affects amyloid-β fibril structure, and cross-seeds amyloid-β aggregation both in vitro and in vivo. Thus, medin could be a therapeutic target for prevention of vascular damage and cognitive decline resulting from amyloid-β deposition in the blood vessels of the brain. ispartof: Nature vol:612 issue:7938 pages:123-131 ispartof: location:England status: published
- Published
- 2022
48. Normality sensing licenses local T cells for innate-like tissue surveillance
- Author
-
Duncan R. McKenzie, Rosie Hart, Nourdine Bah, Dmitry S. Ushakov, Miguel Muñoz-Ruiz, Regina Feederle, and Adrian C. Hayday
- Subjects
Model organisms ,Chemical Biology & High Throughput ,Human Biology & Physiology ,FOS: Clinical medicine ,Genome Integrity & Repair ,Immunology ,Immunology and Allergy ,Tumour Biology ,Genetics & Genomics ,Computational & Systems Biology - Abstract
The increasing implication of lymphocytes in general physiology and immune surveillance outside of infection poses the question of how their antigen receptors might be involved. Here, we show that macromolecular aggregates of intraepidermal γδ T cell antigen receptors (TCRs) in the mouse skin aligned with and depended on Skint1, a butyrophilin-like (BTNL) protein expressed by differentiated keratinocytes (KCs) at steady state. Interruption of TCR-mediated ‘normality sensing’ had no impact on γδ T cell numbers but altered their signature phenotype, while the epidermal barrier function was compromised. In addition to the regulation of steady-state physiology, normality sensing licensed intraepidermal T cells to respond rapidly to subsequent tissue perturbation by using innate tumor necrosis factor (TNF) superfamily receptors. Thus, interfering with Skint1-dependent interactions between local γδ T cells and KCs at steady state increased the susceptibility to ultraviolet B radiation (UVR)-induced DNA damage and inflammation, two cancer-disposing factors.
- Published
- 2022
49. Structures ofArabidopsis thalianaMDL Proteins and Synergistic Effects with the Cytokine MIF on Human Receptors
- Author
-
Lukas Spiller, Ramu Manjula, Franz Leissing, Jerome Basquin, Priscila Bourilhon, Dzmitry Sinitski, Markus Brandhofer, Sophie Levecque, Björn Sabelleck, Regina Feederle, Andrew Flatley, Ralph Panstruga, Jürgen Bernhagen, and Elias Lolis
- Abstract
Vertebrates have developed effective immune mechanisms to fight microbial attacks, relying on a sophisticated network of innate and adaptive responses, a circulatory system, and numerous orchestrating soluble mediators such as cytokines. Mammalian macrophage migration inhibitory factor (MIF) and its paralog D-dopachrome tautomerase (D-DT/MIF-2) are multifunctional inflammatory cytokines with chemokine-like properties that modulate immunity. Plants possess orthologous MIF/D-DT-like (MDL) proteins, whose function is largely unexplored. Driven by the previous discovery of cross-kingdom mimicry of plant (Arabidopsis thaliana) MDL proteins and human MIF receptor signaling, we here characterized the structures of the threeA. thalianaMDLs by X-ray crystallography and explored the mechanism underlying the interplay between plant MDLs, human MIF, and its receptors. We obtained high-resolution structures at 1.56 Å, 1.40 Å, and 2.00 Å resolution for MDL1, MDL2, and MDL3, respectively, revealing a typical trimeric assembly and a high three-dimensional similarity to human MIF. Although residues at the catalytic site of the three MDLs show high identity to human MIF, the proteins showed low tautomerase activity for the substrate 4-hydroxyphenylpyruvate (HPP). Structural differences likely explain the enzymatic inactivity of plant MDLs for HPP. Strikingly, employingin vitro, in vivo,andin plantatest systems, we found that MIF and MDL proteins interact with each other and have the capacity to form hetero-oligomeric complexes. The functional consequences of this interaction were demonstrated applying a yeast-based reporter system specific for the MIF chemokine receptors CXCR2 and CXCR4. MDLs not only triggered receptor signaling on their own, but exhibited pronounced synergism regarding the activation of the CXCR2- and CXCR4-dependent signaling pathways, when co-applied with MIF. These findings were substantiated by the co-administration of pharmacological inhibitors that either disrupt MIF receptor binding or block the catalytic cavity. Moreover, biochemical and biophysical experiments using an allosteric oligomer-specific MIF inhibitor established hexa-oligomer formation between MIF and MDLs as the putative basis for the synergistic effect. Our results are the starting point for a mechanistic understanding of the immunomodulatory activity of a family of highly conserved plant proteins.One Sentence SummaryA. thalianaMDLs and human macrophage inhibitory factor interact with each other and induce synergistic effects in activating CXCR2 and CXCR4.
- Published
- 2023
50. MS4A15 drives ferroptosis resistance through calcium-restricted lipid remodeling
- Author
-
Vanessa A N Kraft, Stefanie M. Hauck, Juliane Merl-Pham, Philippe Schmitt-Kopplin, Xiang Jin, Xuanwen Bao, Constanze Mueller, Susanne Pfeiffer, Shan Xin, Joel A. Schick, and Regina Feederle
- Subjects
Programmed cell death ,Antioxidant ,Biochemical Phenomena ,Endoplasmic reticulum ,medicine.medical_treatment ,chemistry.chemical_element ,Cell Biology ,Calcium ,Cell biology ,Tetraspanin ,chemistry ,Cell culture ,Lipid droplet ,medicine ,Ferroptosis ,lipids (amino acids, peptides, and proteins) ,Lipid Peroxidation ,Oxidation-Reduction ,Molecular Biology ,Phospholipids ,Homeostasis - Abstract
Ferroptosis is an iron-dependent form of cell death driven by biochemical processes that promote oxidation within the lipid compartment. Calcium (Ca2+) is a signaling molecule in diverse cellular processes such as migration, neurotransmission, and cell death. Here, we uncover a crucial link between ferroptosis and Ca2+ through the identification of the novel tetraspanin MS4A15. MS4A15 localizes to the endoplasmic reticulum, where it blocks ferroptosis by depleting luminal Ca2+ stores and reprogramming membrane phospholipids to ferroptosis-resistant species. Specifically, prolonged Ca2+ depletion inhibits lipid elongation and desaturation, driving lipid droplet dispersion and formation of shorter, more saturated ether lipids that protect phospholipids from ferroptotic reactive species. We further demonstrate that increasing luminal Ca2+ levels can preferentially sensitize refractory cancer cell lines. In summary, MS4A15 regulation of anti-ferroptotic lipid reservoirs provides a key resistance mechanism that is distinct from antioxidant and lipid detoxification pathways. Manipulating Ca2+ homeostasis offers a compelling strategy to balance cellular lipids and cell survival in ferroptosis-associated diseases.
- Published
- 2021
Catalog
Discovery Service for Jio Institute Digital Library
For full access to our library's resources, please sign in.