61 results on '"Stephan Hailfinger"'
Search Results
2. Intratumor heterogeneity and T cell exhaustion in primary CNS lymphoma
- Author
-
Michael Heming, Svea Haessner, Jolien Wolbert, I-Na Lu, Xiaolin Li, Benjamin Brokinkel, Michael Müther, Markus Holling, Walter Stummer, Christian Thomas, Andreas Schulte-Mecklenbeck, Flavia de Faria, Marlon Stoeckius, Stephan Hailfinger, Georg Lenz, Kornelius Kerl, Heinz Wiendl, Gerd Meyer zu Hörste, and Oliver M. Grauer
- Subjects
Primary central nervous system lymphoma ,Single-cell RNA sequencing ,Intratumoral heterogeneity ,T cell exhaustion ,Spatial transcriptomics ,Flow cytometry ,Medicine ,Genetics ,QH426-470 - Abstract
Abstract Background Primary central nervous system lymphoma (PCNSL) is a rare lymphoma of the central nervous system, usually of diffuse large B cell phenotype. Stereotactic biopsy followed by histopathology is the diagnostic standard. However, limited material is available from CNS biopsies, thus impeding an in-depth characterization of PCNSL. Methods We performed flow cytometry, single-cell RNA sequencing, and B cell receptor sequencing of PCNSL cells released from biopsy material, blood, and cerebrospinal fluid (CSF), and spatial transcriptomics of biopsy samples. Results PCNSL-released cells were predominantly activated CD19+CD20+CD38+CD27+ B cells. In single-cell RNA sequencing, PCNSL cells were transcriptionally heterogeneous, forming multiple malignant B cell clusters. Hyperexpanded B cell clones were shared between biopsy- and CSF- but not blood-derived cells. T cells in the tumor microenvironment upregulated immune checkpoint molecules, thereby recognizing immune evasion signals from PCNSL cells. Spatial transcriptomics revealed heterogeneous spatial organization of malignant B cell clusters, mirroring their transcriptional heterogeneity across patients, and pronounced expression of T cell exhaustion markers, co-localizing with a highly malignant B cell cluster. Conclusions Malignant B cells in PCNSL show transcriptional and spatial intratumor heterogeneity. T cell exhaustion is frequent in the PCNSL microenvironment, co-localizes with malignant cells, and highlights the potential of personalized treatments.
- Published
- 2022
- Full Text
- View/download PDF
3. Human invariant natural killer T cells promote tolerance by preferential apoptosis induction of conventional dendritic cells
- Author
-
Hannes Schmid, Emmanuelle M. Ribeiro, Kathy-Ann Secker, Silke Duerr-Stoerzer, Hildegard Keppeler, Ruoyun Dong, Timo Munz, Klaus Schulze-Osthoff, Stephan Hailfinger, Corina Schneidawind, and Dominik Schneidawind
- Subjects
Diseases of the blood and blood-forming organs ,RC633-647.5 - Abstract
Graft-versus-host disease (GvHD) is a major cause of morbidity and mortality after allogeneic hematopoietic cell transplantation. We recently showed in murine studies and in vitro human models that adoptively transferred invariant natural killer T (iNKT) cells protect from GvHD and promote graft-versus-leukemia effects. The cellular mechanisms underlying GvHD prevention by iNKT cells in humans, however, remain unknown. In order to study relevant cellular interactions, dendritic cells (DC) were either generated from monocytes or isolated directly from blood of healthy donors or GvHD patients and co-cultured in a mixed lymphocyte reaction (MLR) with T cells obtained from healthy donors or transplantation bags. Addition of culture-expanded iNKT cells to the MLR-induced DC apoptosis in a cell contact-dependent manner, thereby preventing T-cell activation and proliferation. Annexin V/propidium iodide staining and image stream assays showed that CD4+CD8–, CD4–CD8+ and double negative iNKT cells are similarly able to induce DC apoptosis. Further MLR assays revealed that conventional DC (cDC) but not plasmacytoid DC (pDC) could induce alloreactive T-cell activation and proliferation. Interestingly, cDC were also more susceptible to apoptosis induced by iNKT cells, which correlates with their higher CD1d expression, leading to a bias in favor of pDC. Remarkably, these results could also be observed in GvHD patients. We propose a new mechanism how ex vivo expanded human iNKT cells prevent alloreactivity of T cells. iNKT cells modulate T-cell responses by selective apoptosis of DC subsets, resulting in suppression of T-cell activation and proliferation while enabling beneficial immune responses through pDC.
- Published
- 2021
- Full Text
- View/download PDF
4. The Paracaspase MALT1 in Cancer
- Author
-
Beatriz Gomez Solsona, Anja Schmitt, Klaus Schulze-Osthoff, and Stephan Hailfinger
- Subjects
MALT1 ,BCL10 ,CARD11 ,CARD10 ,CBM complex ,NF-κB ,Biology (General) ,QH301-705.5 - Abstract
Almost twenty years ago, the importance of the paracaspase MALT1 in antigen receptor-induced NF-κB activation was first described. Since then, several other immune receptors, G-protein-coupled receptors, and receptor tyrosine kinases were identified as relying on MALT1 to induce NF-κB activation. In various hematological malignancies and solid tumors, MALT1 is constitutively activated and drives chronic NF-κB target gene expression. Deregulated MALT1 activity in cancer thus promotes tumor cell survival, proliferation, and metastasis. Since the molecular function of MALT1 partially requires its protease activity, pharmacological targeting of MALT1 may represent a promising anti-cancer strategy. Here, we review the molecular features of MALT1 activation and function as well as the therapeutic potential of MALT1 inhibition in hematological malignancies and solid tumors.
- Published
- 2022
- Full Text
- View/download PDF
5. Release of Immunomodulatory Ebola Virus Glycoprotein-Containing Microvesicles Is Suppressed by Tetherin in a Species-Specific Manner
- Author
-
Julia Nehls, Ramona Businger, Markus Hoffmann, Constantin Brinkmann, Birgit Fehrenbacher, Martin Schaller, Brigitte Maurer, Caroline Schönfeld, Daniela Kramer, Stephan Hailfinger, Stefan Pöhlmann, and Michael Schindler
- Subjects
Biology (General) ,QH301-705.5 - Abstract
Summary: The Ebola virus glycoprotein (EBOV-GP) forms GP-containing microvesicles, so-called virosomes, which are secreted from GP-expressing cells. However, determinants of GP-virosome release and their functionality are poorly understood. We characterized GP-mediated virosome formation and delineated the role of the antiviral factor tetherin (BST2, CD317) in this process. Residues in the EBOV-GP receptor-binding domain (RBD) promote GP-virosome secretion, while tetherin suppresses GP-virosomes by interactions involving the GP-transmembrane domain. Tetherin from multiple species interfered with GP-virosome release, and tetherin from the natural fruit bat reservoir showed the highest inhibitory activity. Moreover, analyses of GP from various ebolavirus strains, including the EBOV responsible for the West African epidemic, revealed the most efficient GP-virosome formation by highly pathogenic ebolaviruses. Finally, EBOV-GP-virosomes were immunomodulatory and acted as decoys for EBOV-neutralizing antibodies. Our results indicate that GP-virosome formation might be a determinant of EBOV immune evasion and pathogenicity, which is suppressed by tetherin. : Nehls et al. demonstrate that the glycoprotein of the highly pathogenic Ebola virus is incorporated into secretory vesicles, called GP-virosomes, to dampen the immune response and capture neutralizing antibodies. The lack of replication competence and the incorporation of antigenically intact GP might qualify GP-virosomes as safe vaccine candidates. Keywords: Ebola virus, glycoprotein, microvesicles, virosome, exosome, tetherin, immune modulation, immune evasion, antiviral immune response, neutralizing antibody
- Published
- 2019
- Full Text
- View/download PDF
6. NF-κB Activation in Lymphoid Malignancies: Genetics, Signaling, and Targeted Therapy
- Author
-
Paula Grondona, Philip Bucher, Klaus Schulze-Osthoff, Stephan Hailfinger, and Anja Schmitt
- Subjects
NF-κB ,lymphoma ,leukemia ,CARMA1 ,CARD11 ,CD79 ,MyD88 ,Biology (General) ,QH301-705.5 - Abstract
The NF-κB transcription factor family plays a crucial role in lymphocyte proliferation and survival. Consequently, aberrant NF-κB activation has been described in a variety of lymphoid malignancies, including diffuse large B-cell lymphoma, Hodgkin lymphoma, and adult T-cell leukemia. Several factors, such as persistent infections (e.g., with Helicobacter pylori), the pro-inflammatory microenvironment of the cancer, self-reactive immune receptors as well as genetic lesions altering the function of key signaling effectors, contribute to constitutive NF-κB activity in these malignancies. In this review, we will discuss the molecular consequences of recurrent genetic lesions affecting key regulators of NF-κB signaling. We will particularly focus on the oncogenic mechanisms by which these alterations drive deregulated NF-κB activity and thus promote the growth and survival of the malignant cells. As the concept of a targeted therapy based on the mutational status of the malignancy has been supported by several recent preclinical and clinical studies, further insight in the function of NF-κB modulators and in the molecular mechanisms governing aberrant NF-κB activation observed in lymphoid malignancies might lead to the development of additional treatment strategies and thus improve lymphoma therapy.
- Published
- 2018
- Full Text
- View/download PDF
7. Monoubiquitination and activity of the paracaspase MALT1 requires glutamate 549 in the dimerization interface.
- Author
-
Katrin Cabalzar, Christiane Pelzer, Annette Wolf, Georg Lenz, Justyna Iwaszkiewicz, Vincent Zoete, Stephan Hailfinger, and Margot Thome
- Subjects
Medicine ,Science - Abstract
The mucosa-associated lymphoid tissue protein-1 (MALT1, also known as paracaspase) is a protease whose activity is essential for the activation of lymphocytes and the growth of cells derived from human diffuse large B-cell lymphomas of the activated B-cell subtype (ABC DLBCL). Crystallographic approaches have shown that MALT1 can form dimers via its protease domain, but why dimerization is relevant for the biological activity of MALT1 remains largely unknown. Using a molecular modeling approach, we predicted Glu 549 (E549) to be localized within the MALT1 dimer interface and thus potentially relevant. Experimental mutation of this residue into alanine (E549A) led to a complete impairment of MALT1 proteolytic activity. This correlated with an impaired capacity of the mutant to form dimers of the protease domain in vitro, and a reduced capacity to promote NF-κB activation and transcription of the growth-promoting cytokine interleukin-2 in antigen receptor-stimulated lymphocytes. Moreover, this mutant could not rescue the growth of ABC DLBCL cell lines upon MALT1 silencing. Interestingly, the MALT1 mutant E549A was unable to undergo monoubiquitination, which we identified previously as a critical step in MALT1 activation. Collectively, these findings suggest a model in which E549 at the dimerization interface is required for the formation of the enzymatically active, monoubiquitinated form of MALT1.
- Published
- 2013
- Full Text
- View/download PDF
8. Plasmablastic lymphoma: from genetics to treatment
- Author
-
Fabian, Frontzek, Stephan, Hailfinger, and Georg, Lenz
- Subjects
Cancer Research ,Oncology ,Hematology - Abstract
Plasmablastic lymphoma (PBL) represents a rare distinct lymphoma entity with plasmablastic morphology and plasmacytic immunophenotype that is characterized by an aggressive clinical course. Standard chemotherapeutic regimens often remain insufficient to cure affected patients. Recently, comprehensive molecular analyses of large cohorts of primary PBL samples have revealed the mutational landscape as well as the pattern of copy number alterations of this rare lymphoma subtype. Identification of recurrent aberrations affecting the JAK-STAT, RAS-RAF, NOTCH, IRF4, and MYC signaling pathways drive the molecular pathogenesis of PBL and hold great potential for novel targeted therapeutic approaches.
- Published
- 2022
- Full Text
- View/download PDF
9. Combination Treatment Targeting mTOR and MAPK Pathways Has Synergistic Activity in Multiple Myeloma
- Author
-
Kaiyan Sun, Ling Jin, Jana Karolová, Jan Vorwerk, Stephan Hailfinger, Bertram Opalka, Myroslav Zapukhlyak, Georg Lenz, and Cyrus Khandanpour
- Subjects
multiple myeloma ,mTOR ,temsirolimus ,MEK ,trametinib ,targeted therapy ,Cancer Research ,Oncology ,Medizin - Abstract
Multiple myeloma (MM) is an incurable, malignant B cell disorder characterized by frequent relapses and a poor prognosis. Thus, new therapeutic approaches are warranted. The phosphatidylinositol-3-kinase (PI3K) pathway plays a key role in many critical cellular processes, including cell proliferation and survival. Activated PI3K/AKT (protein kinases B)/mTOR (mammalian target of rapamycin) signaling has been identified in MM primary patient samples and cell lines. In this study, the efficacy of PI3K and mTOR inhibitors in various MM cell lines representing three different prognostic subtypes was tested. Whereas MM cell lines were rather resistant to PI3K inhibition, treatment with the mTOR inhibitor temsirolimus decreases the phosphorylation of key molecules in the PI3K pathway in MM cell lines, leading to G0/G1 cell cycle arrest and thus reduced proliferation. Strikingly, the efficacy of temsirolimus was amplified by combining the treatment with the Mitogen-activated protein kinase kinase (MEK) inhibitor trametinib. Our findings provide a scientific rationale for the simultaneous inhibition of mTOR and MEK as a novel strategy for the treatment of MM.
- Published
- 2023
10. Molecular profiling of EBV associated diffuse large B-cell lymphoma
- Author
-
Fabian Frontzek, Annette M. Staiger, Ramona Wullenkord, Michael Grau, Myroslav Zapukhlyak, Katrin S. Kurz, Heike Horn, Tabea Erdmann, Falko Fend, Julia Richter, Wolfram Klapper, Peter Lenz, Stephan Hailfinger, Anna Tasidou, Marcel Trautmann, Wolfgang Hartmann, Andreas Rosenwald, Leticia Quintanilla-Martinez, German Ott, Ioannis Anagnostopoulos, and Georg Lenz
- Subjects
Cancer Research ,Oncology ,Hematology - Abstract
Epstein-Barr virus (EBV) associated diffuse large B-cell lymphoma (DLBCL) represents a rare aggressive B-cell lymphoma subtype characterized by an adverse clinical outcome. EBV infection of lymphoma cells has been associated with different lymphoma subtypes while the precise role of EBV in lymphomagenesis and specific molecular characteristics of these lymphomas remain elusive. To further unravel the biology of EBV associated DLBCL, we present a comprehensive molecular analysis of overall 60 primary EBV positive (EBV+) DLBCLs using targeted sequencing of cancer candidate genes (CCGs) and genome-wide determination of recurrent somatic copy number alterations (SCNAs) in 46 cases, respectively. Applying the LymphGen classifier 2.0, we found that less than 20% of primary EBV + DLBCLs correspond to one of the established molecular DLBCL subtypes underscoring the unique biology of this entity. We have identified recurrent mutations activating the oncogenic JAK-STAT and NOTCH pathways as well as frequent amplifications of 9p24.1 contributing to immune escape by PD-L1 overexpression. Our findings enable further functional preclinical and clinical studies exploring the therapeutic potential of targeting these aberrations in patients with EBV + DLBCL to improve outcome.
- Published
- 2022
11. Impaired Autophagy in Psoriasis and Atopic Dermatitis: A New Therapeutic Target?
- Author
-
Stephan Hailfinger and Klaus Schulze-Osthoff
- Subjects
Keratinocytes ,Cathepsin ,business.industry ,Inflammatory skin disease ,Autophagy ,Treatment options ,Inflammation ,Cell Biology ,Dermatology ,Atopic dermatitis ,medicine.disease ,Biochemistry ,Dermatitis, Atopic ,body regions ,Psoriasis ,Immunology ,Humans ,Medicine ,Keratinocyte differentiation ,medicine.symptom ,business ,Molecular Biology ,Skin - Abstract
Dysfunctional autophagy is linked to various diseases, including psoriasis and atopic dermatitis. Recent evidence suggests that exposure of keratinocytes to TNF-α results in impaired autophagy and lysosomal function. The skin of patients with psoriasis and atopic dermatitis reveals a decreased expression of lysosomal cathepsins. Impaired autophagy is presumably involved in inflammation and disturbed keratinocyte differentiation, whereas stimulating autophagy might be a treatment option in inflammatory skin disease.
- Published
- 2021
- Full Text
- View/download PDF
12. Dimethyl fumarate induces ferroptosis and impairs NF-κB/STAT3 signaling in DLBCL
- Author
-
Pavel Klener, Klaus Schulze-Osthoff, Karsten Boldt, Philipp Berning, Martina Konantz, Michael Grau, Georg Lenz, Caroline Schönfeld, Wendan Xu, Philip Bucher, Petra Vockova, Mohamed Ali Jarboui, Anja Schmitt, Andreas Rosenwald, German Ott, Melanie Grimm, Claudia Lengerke, Marc Brändle, Heike Horn, Stephan Hailfinger, and Myroslav Zapukhlyak
- Subjects
Programmed cell death ,Dimethyl fumarate ,biology ,Chemistry ,Immunology ,Germinal center ,Cell Biology ,Hematology ,GPX4 ,Biochemistry ,chemistry.chemical_compound ,medicine.anatomical_structure ,immune system diseases ,hemic and lymphatic diseases ,medicine ,biology.protein ,Cancer research ,Signal transduction ,Janus kinase ,STAT3 ,B cell - Abstract
Despite the development of novel targeted drugs, the molecular heterogeneity of diffuse large B-cell lymphoma (DLBCL) still poses a substantial therapeutic challenge. DLBCL can be classified into at least 2 major subtypes (germinal center B cell [GCB]-like and activated B cell [ABC]-like DLBCL), each characterized by specific gene expression profiles and mutation patterns. Here we demonstrate a broad antitumor effect of dimethyl fumarate (DMF) on both DLBCL subtypes, which is mediated by the induction of ferroptosis, a form of cell death driven by the peroxidation of phospholipids. As a result of the high expression of arachidonate 5-lipoxygenase in concert with low glutathione and glutathione peroxidase 4 levels, DMF induces lipid peroxidation and thus ferroptosis, particularly in GCB DLBCL. In ABC DLBCL cells, which are addicted to NF-κB and STAT3 survival signaling, DMF treatment efficiently inhibits the activity of the IKK complex and Janus kinases. Interestingly, the BCL-2–specific BH3 mimetic ABT-199 and an inhibitor of ferroptosis suppressor protein 1 synergize with DMF in inducing cell death in DLBCL. Collectively, our findings identify the clinically approved drug DMF as a promising novel therapeutic option in the treatment of both GCB and ABC DLBCLs.
- Published
- 2021
- Full Text
- View/download PDF
13. mTOR inhibition amplifies the anti-lymphoma effect of PI3Kβ/δ blockage in diffuse large B-cell lymphoma
- Author
-
Wendan Xu, Philipp Berning, Tabea Erdmann, Michael Grau, Nardjas Bettazová, Myroslav Zapukhlyak, Fabian Frontzek, Corinna Kosnopfel, Peter Lenz, Michael Grondine, Brandon Willis, James T. Lynch, Pavel Klener, Stephan Hailfinger, Simon T. Barry, and Georg Lenz
- Subjects
Cancer Research ,Oncology ,Hematology - Abstract
Diffuse large B-cell lymphoma (DLBCL) is an aggressive disease that exhibits constitutive activation of phosphoinositide 3-kinase (PI3K) driven by chronic B-cell receptor signaling or PTEN deficiency. Since pan-PI3K inhibitors cause severe side effects, we investigated the anti-lymphoma efficacy of the specific PI3Kβ/δ inhibitor AZD8186. We identified a subset of DLBCL models within activated B-cell–like (ABC) and germinal center B-cell–like (GCB) DLBCL that were sensitive to AZD8186 treatment. On the molecular level, PI3Kβ/δ inhibition decreased the pro-survival NF-κB and AP-1 activity or led to downregulation of the oncogenic transcription factor MYC. In AZD8186-resistant models, we detected a feedback activation of the PI3K/AKT/mTOR pathway following PI3Kβ/δ inhibition, which limited AZD8186 efficacy. The combined treatment with AZD8186 and the mTOR inhibitor AZD2014 overcame resistance to PI3Kβ/δ inhibition and completely prevented outgrowth of lymphoma cells in vivo in cell line- and patient-derived xenograft mouse models. Collectively, our study reveals that subsets of DLBCLs are addicted to PI3Kβ/δ signaling and thus identifies a previously unappreciated role of the PI3Kβ isoform in DLBCL survival. Furthermore, our data demonstrate that combined targeting of PI3Kβ/δ and mTOR is effective in all major DLBCL subtypes supporting the evaluation of this strategy in a clinical trial setting.
- Published
- 2022
14. Human invariant natural killer T cells promote tolerance by preferential apoptosis induction of conventional dendritic cells
- Author
-
Klaus Schulze-Osthoff, Ruoyun Dong, Timo Munz, Silke Duerr-Stoerzer, Hannes Schmid, Hildegard Keppeler, Dominik Schneidawind, Kathy-Ann Secker, Corina Schneidawind, Stephan Hailfinger, and Emmanuelle M Ribeiro
- Subjects
Cell ,Graft vs Host Disease ,Apoptosis ,Lymphocyte Activation ,Mice ,03 medical and health sciences ,0302 clinical medicine ,Immune system ,medicine ,Animals ,Humans ,030304 developmental biology ,0303 health sciences ,biology ,Chemistry ,Dendritic Cells ,Hematology ,Mixed lymphocyte reaction ,Transplantation ,medicine.anatomical_structure ,CD1D ,biology.protein ,Cancer research ,Natural Killer T-Cells ,CD8 ,Ex vivo ,030215 immunology - Abstract
Graft-versus-host disease (GvHD) is a major cause of morbidity and mortality after allogeneic hematopoietic cell transplantation. We recently showed in murine studies and in vitro human models that adoptively transferred invariant natural killer T (iNKT) cells protect from GvHD and promote graft-versus-leukemia effects. The cellular mechanisms underlying GvHD prevention by iNKT cells in humans, however, remain unknown. In order to study relevant cellular interactions, dendritic cells (DC) were either generated from monocytes or isolated directly from blood of healthy donors or GvHD patients and co-cultured in a mixed lymphocyte reaction (MLR) with T cells obtained from healthy donors or transplantation bags. Addition of culture-expanded iNKT cells to the MLR-induced DC apoptosis in a cell contact-dependent manner, thereby preventing T-cell activation and proliferation. Annexin V/propidium iodide staining and image stream assays showed that CD4+CD8–, CD4–CD8+ and double negative iNKT cells are similarly able to induce DC apoptosis. Further MLR assays revealed that conventional DC (cDC) but not plasmacytoid DC (pDC) could induce alloreactive T-cell activation and proliferation. Interestingly, cDC were also more susceptible to apoptosis induced by iNKT cells, which correlates with their higher CD1d expression, leading to a bias in favor of pDC. Remarkably, these results could also be observed in GvHD patients. We propose a new mechanism how ex vivo expanded human iNKT cells prevent alloreactivity of T cells. iNKT cells modulate T-cell responses by selective apoptosis of DC subsets, resulting in suppression of T-cell activation and proliferation while enabling beneficial immune responses through pDC.
- Published
- 2021
- Full Text
- View/download PDF
15. Ferroptotic pores induce Ca2+ fluxes and ESCRT-III activation to modulate cell death kinetics
- Author
-
Uris Ros, Stephan Hailfinger, Jenny Stroh, Silvia von Karstedt, Rafael A. Espiritu, Lohans Pedrera, Josephine Weber, Anja Schmitt, and Ana J. García-Sáez
- Subjects
Programmed cell death ,Necrosis ,Cell ,Cell Biology ,ESCRT ,Cell biology ,Lipid peroxidation ,Cytosol ,chemistry.chemical_compound ,medicine.anatomical_structure ,Membrane ,chemistry ,Lytic cycle ,medicine ,medicine.symptom ,Molecular Biology - Abstract
Ferroptosis is an iron-dependent form of regulated necrosis associated with lipid peroxidation. Despite its key role in the inflammatory outcome of ferroptosis, little is known about the molecular events leading to the disruption of the plasma membrane during this type of cell death. Here we show that a sustained increase in cytosolic Ca2+ is a hallmark of ferroptosis that precedes complete bursting of the cell. We report that plasma membrane damage leading to ferroptosis is associated with membrane nanopores of a few nanometers in radius and that ferroptosis, but not lipid peroxidation, can be delayed by osmoprotectants. Importantly, Ca2+ fluxes during ferroptosis induce the activation of the ESCRT-III-dependent membrane repair machinery, which counterbalances the kinetics of cell death and modulates the immunological signature of ferroptosis. Our findings with ferroptosis provide a unifying concept that sustained increase of cytosolic Ca2+ prior to plasma membrane rupture is a common feature of regulated types of necrosis and position ESCRT-III activation as a general protective mechanism in these lytic cell death pathways.
- Published
- 2020
- Full Text
- View/download PDF
16. The CDK4/6-EZH2 pathway is a potential therapeutic target for psoriasis
- Author
-
Matthias Dobbelstein, Antje Dickmanns, Anne Müller, Daniela Kramer, Knut Schäkel, Stephan Hailfinger, Klaus Schulze-Osthoff, and Claudia Resch
- Subjects
Keratinocytes ,0301 basic medicine ,macromolecular substances ,Proinflammatory cytokine ,Mice ,03 medical and health sciences ,0302 clinical medicine ,Psoriasis ,medicine ,Animals ,Humans ,Enhancer of Zeste Homolog 2 Protein ,STAT3 ,Transcription factor ,integumentary system ,biology ,business.industry ,Kinase ,Cyclin-Dependent Kinase 4 ,Cyclin-Dependent Kinase 6 ,General Medicine ,medicine.disease ,3. Good health ,Disease Models, Animal ,HEK293 Cells ,030104 developmental biology ,medicine.anatomical_structure ,030220 oncology & carcinogenesis ,Cancer research ,biology.protein ,Cyclin-dependent kinase 6 ,Signal transduction ,business ,Keratinocyte ,Signal Transduction ,Research Article - Abstract
Psoriasis is a frequent, inflammatory skin disease characterized by keratinocyte hyperproliferation and a disease-related infiltration of immune cells. Here, we identified a novel proinflammatory signaling pathway driven by cyclin-dependent kinase 4 (CDK4) and CDK6 and the methyltransferase EZH2 as a valid target for psoriasis therapy. Delineation of the pathway revealed that CDK4/6 phosphorylated EZH2 in keratinocytes, thereby triggering a methylation-induced activation of STAT3. Subsequently, active STAT3 resulted in the induction of IκBζ, which is a key proinflammatory transcription factor required for cytokine synthesis in psoriasis. Pharmacological or genetic inhibition of CDK4/6 or EZH2 abrogated psoriasis-related proinflammatory gene expression by suppressing IκBζ induction in keratinocytes. Importantly, topical application of CDK4/6 or EZH2 inhibitors on the skin was sufficient to fully prevent the development of psoriasis in various mouse models by suppressing STAT3-mediated IκBζ expression. Moreover, we found a hyperactivation of the CDK4/6-EZH2 pathway in human and mouse psoriatic skin lesions. Thus, this study not only identifies a novel psoriasis-relevant proinflammatory pathway, but also proposes the repurposing of CDK4/6 or EZH2 inhibitors as a new therapeutic option for patients with psoriasis.
- Published
- 2020
- Full Text
- View/download PDF
17. The Paracaspase MALT1 in Cancer
- Author
-
Beatriz, Gomez Solsona, Anja, Schmitt, Klaus, Schulze-Osthoff, and Stephan, Hailfinger
- Abstract
Almost twenty years ago, the importance of the paracaspase MALT1 in antigen receptor-induced NF-κB activation was first described. Since then, several other immune receptors, G-protein-coupled receptors, and receptor tyrosine kinases were identified as relying on MALT1 to induce NF-κB activation. In various hematological malignancies and solid tumors, MALT1 is constitutively activated and drives chronic NF-κB target gene expression. Deregulated MALT1 activity in cancer thus promotes tumor cell survival, proliferation, and metastasis. Since the molecular function of MALT1 partially requires its protease activity, pharmacological targeting of MALT1 may represent a promising anti-cancer strategy. Here, we review the molecular features of MALT1 activation and function as well as the therapeutic potential of MALT1 inhibition in hematological malignancies and solid tumors.
- Published
- 2021
18. Intratumor heterogeneity and T cell exhaustion in primary CNS lymphoma
- Author
-
Michael, Heming, Svea, Haessner, Jolien, Wolbert, I-Na, Lu, Xiaolin, Li, Benjamin, Brokinkel, Michael, Müther, Markus, Holling, Walter, Stummer, Christian, Thomas, Andreas, Schulte-Mecklenbeck, Flavia, de Faria, Marlon, Stoeckius, Stephan, Hailfinger, Georg, Lenz, Kornelius, Kerl, Heinz, Wiendl, Gerd, Meyer Zu Hörste, and Oliver M, Grauer
- Subjects
Central Nervous System Neoplasms ,Lymphoma ,T-Lymphocytes ,Tumor Microenvironment ,Humans ,Receptors, Antigen, B-Cell ,Immune Checkpoint Proteins - Abstract
Primary central nervous system lymphoma (PCNSL) is a rare lymphoma of the central nervous system, usually of diffuse large B cell phenotype. Stereotactic biopsy followed by histopathology is the diagnostic standard. However, limited material is available from CNS biopsies, thus impeding an in-depth characterization of PCNSL.We performed flow cytometry, single-cell RNA sequencing, and B cell receptor sequencing of PCNSL cells released from biopsy material, blood, and cerebrospinal fluid (CSF), and spatial transcriptomics of biopsy samples.PCNSL-released cells were predominantly activated CD19sup+/supCD20sup+/supCD38sup+/supCD27sup+/supB cells. In single-cell RNA sequencing, PCNSL cells were transcriptionally heterogeneous, forming multiple malignant B cell clusters. Hyperexpanded B cell clones were shared between biopsy- and CSF- but not blood-derived cells. T cells in the tumor microenvironment upregulated immune checkpoint molecules, thereby recognizing immune evasion signals from PCNSL cells. Spatial transcriptomics revealed heterogeneous spatial organization of malignant B cell clusters, mirroring their transcriptional heterogeneity across patients, and pronounced expression of T cell exhaustion markers, co-localizing with a highly malignant B cell cluster.Malignant B cells in PCNSL show transcriptional and spatial intratumor heterogeneity. T cell exhaustion is frequent in the PCNSL microenvironment, co-localizes with malignant cells, and highlights the potential of personalized treatments.
- Published
- 2021
19. Temporal Dynamics of Reactive Oxygen and Nitrogen Species and NF-κB Activation During Acute and Chronic T Cell–Driven Inflammation
- Author
-
Kamran Ghoreschi, Wolfgang M. Thaiss, Harald Carlsen, Roman Mehling, Stephan Hailfinger, Hasan Halit Öz, Bernd J. Pichler, Martin Röcken, Manfred Kneilling, Johannes Schwenck, Klaus Schulze-Osthoff, Leticia Quintanilla-Martinez, Daniela Kramer, Dominik Hartl, and Irene Gonzalez Menendez
- Subjects
Cancer Research ,Contact allergy ,T-Lymphocytes ,T cell ,CD3 ,Mice, Transgenic ,Inflammation ,Picryl Chloride ,Pharmacology ,NF-κB ,030218 nuclear medicine & medical imaging ,Mice ,03 medical and health sciences ,0302 clinical medicine ,In vivo ,L-012 ,medicine ,Animals ,Radiology, Nuclear Medicine and imaging ,biology ,Chemistry ,Anti-inflammatory effect ,Optical Imaging ,NF-kappa B ,Delayed-type hypersensitivity reaction ,Free Radical Scavengers ,Reactive Nitrogen Species ,N-acetylcysteine ,Acetylcysteine ,Mice, Inbred C57BL ,Disease Models, Animal ,Real-time polymerase chain reaction ,medicine.anatomical_structure ,Oncology ,Myeloperoxidase ,biology.protein ,Contact hypersensitivity reaction ,Immunohistochemistry ,Female ,medicine.symptom ,Reactive Oxygen Species ,Ex vivo ,Research Article ,Signal Transduction - Abstract
Purpose Reactive oxygen and nitrogen species (ROS/RNS) production and the NF-κB activation are critically involved in inflammatory responses, but knowledge about the temporal dynamics during acute and chronic inflammation is limited. Here, we present a comparative longitudinal in vivo study of both parameters in an experimental model of acute and chronic T cell–driven delayed-type hypersensitivity reaction (DTHR) using noninvasive optical imaging. Procedures Trinitrochlorobenzene (TNCB)-sensitized NF-κB-luciferase-reporter and wild-type mice were TNCB challenged on the right ear to elicit acute DTHR and then repetitively challenged (up to five times) to induce chronic DTHR. Mice were treated with the ROS-scavenging and NF-κB inhibiting molecule N-acetylcysteine (NAC) or underwent sham treatment. ROS/RNS production was noninvasively analyzed in vivo using the ROS-/RNS-sensitive chemiluminescent probe L-012, and NF-κB activation was measured using NF-κB-luciferase-reporter mice. H&E staining, CD3 and myeloperoxidase (MPO) immunohistochemistry (IHC), and quantitative PCR (qPCR) analyses were employed to investigate immune cell infiltration and expression of NF-κB- and ROS-/RNS-driven genes. Results In acute DTHR, we found strongly elevated ROS/RNS production and NF-κB activation 12 h after the 1st TNCB ear challenge, peaking at 24 h after the challenge. In chronic DTHR, ROS production peaked as early as 4 h after the 5th TNCB challenge, whereas NF-κB activity peaked after 12 h. The increase in ROS/RNS production in acute DTHR was higher than the increase in NF-κB activity but the relationship was inverse in chronic DTHR. Treatment with the ROS scavenger NAC had differential effects on ROS/RNS production and NF-κB activation during acute and chronic DTHR. Ex vivo cross-validation by histopathology and qPCR analysis correlated closely with the in vivo imaging results. Conclusions Noninvasive in vivo imaging is capable of assessing the temporal dynamics of ROS/RNS production and NF-κB activation during progression from acute to chronic DTHR and enables monitoring of anti-inflammatory treatment responses. Electronic supplementary material The online version of this article (10.1007/s11307-019-01412-8) contains supplementary material, which is available to authorized users.
- Published
- 2019
- Full Text
- View/download PDF
20. The paracaspase MALT1 in psoriasis
- Author
-
Klaus Schulze-Osthoff and Stephan Hailfinger
- Subjects
0301 basic medicine ,medicine.medical_treatment ,Clinical Biochemistry ,Biochemistry ,03 medical and health sciences ,chemistry.chemical_compound ,0302 clinical medicine ,Immune system ,Psoriasis ,medicine ,Humans ,Molecular Biology ,Transcription factor ,business.industry ,Membrane Proteins ,NF-κB ,Paracaspase ,medicine.disease ,BCL10 ,CARD Signaling Adaptor Proteins ,MALT1 ,030104 developmental biology ,Cytokine ,chemistry ,Guanylate Cyclase ,Mucosa-Associated Lymphoid Tissue Lymphoma Translocation 1 Protein ,030220 oncology & carcinogenesis ,Cancer research ,business - Abstract
Psoriasis is a frequent autoimmune-related skin disease, which involves various cell types such as T cells, keratinocytes and dendritic cells. Genetic variations, such as mutations of CARD14, can promote the development of the disease. CARD14 mutations as well as the stimulation of immune and cytokine receptors activate the paracaspase MALT1, a potent activator of the transcription factors NF-κB and AP-1. The disease-promoting role of MALT1 for psoriasis is mediated by both its protease activity as well as its molecular scaffold function. Here, we review the importance of MALT1-mediated signaling and its therapeutic implications in psoriasis.
- Published
- 2021
21. Abstract 2008: The oncogenic fusion protein DNAJB1-PRKACA can be actively targeted by peptide-based immunotherapy in fibrolamellar hepatocellular carcinoma
- Author
-
Jens Bauer, Natalie Köhler, Yacine Maringer, Philip Bucher, Tatjana Bilich, Melissa Zwick, Severin Dicks, Annika Nelde, Marissa Dubbelaar, Jonas Scheid, Marcel Wacker, Jonas J. Heitmann, Sarah Schroeder, Jonas Rieth, Monika Denk, Marion Richter, Reinhild Klein, Irina Bonzheim, Julia Luibrand, Ursula Holzer, Martin Ebinger, Ines B. Brecht, Michael Bitzer, Melanie Boerries, Helmut R. Salih, Hans-Georg Rammensee, Stephan Hailfinger, and Juliane S. Walz
- Subjects
Cancer Research ,Oncology - Abstract
Fibrolamellar hepatocellular carcinoma (FL-HCC) is a rare tumor disease, which affects children and adolescents without history of primary liver disease. Beside surgical resection established treatment options are lacking for FL-HCC. Recently, the DNAJB1-PRKACA fusion transcript was identified as the oncogenic driver of tumor pathogenesis in 100% of FL-HCC patients. Here, we investigated the role of the DNAJB1-PRKACA fusion protein as a source for immunogenic neoepitopes and showed first immunotherapeutic application of these antigens in a FL-HCC patient.HLA class I- and class II-presented neoantigens derived from the DNAJB1-PRKACA fusion protein were predicted in silico using NetMHCpan 4.1 and SYFPEITHI 1.0, or NetMHCIIpan 4.0, respectively. With this workflow nine binding cores of nine amino acid length for a total of 1290 different HLA class II alleles, as well as 13 HLA class I ligands for the 20 most frequent HLA class I allotypes (European population, iedb.org) were identified. Cellular processing and HLA presentation of DNAJB1-PRKACA-derived peptides was proven by liquid chromatography-coupled tandem mass spectrometry (LC-MS/MS) of DNAJB1-PRKACA-transduced HCC cell lines. Immunogenicity of DNAJB1-PRKACA-derived peptides was assessed for the HLA class II peptide (PII-1) and the HLA-A*24 peptide (PA*24) by in vitro priming experiments which showed an induction of multifunctional peptide-specific CD4+ and CD8+ T cells, respectively, with expression of CD107a, IFNγ, and TNF upon peptide-pulsing. Furthermore, PA*24-specific T cells showed antigen-specific lysis of autologous peptide-loaded target cells and single-cell next-generation sequencing (10x Genomics) of PA*24-specific CD8+ T cells further enabled the identification of DNAJB1-PRKACA-reactive T cell receptors. Based on these preclinical data we applied a peptide vaccine, consisting of three HLA class I ligands (PA*02, PB*44, and PC*05) and PII-1 spanning the DNAJB1-PRKACA fusion region, to a 15-year old patient with histologically confirmed FL-HCC, who experienced multiple tumor relapses after early liver transplant due to unresectable FL-HCC not responsive to chemotherapy. After two vaccinations in vivo induction of multifunctional CD4+ T cells targeting PII-1 and PB*44 was observed by IFNγ ELISPOT. Single-cell RNA sequencing of vaccine-induced CD4+ T cells revealed distinct gene expression clusters of T cell activation and high TCR clonality. DNAJB1-PRKACA-specific T cells persisted in peripheral blood and were accompanied by relapse free survival of the patient until now, more than one year post vaccination. These findings identified the DNAJB1-PRKACA fusion transcript as novel prime source for broadly applicable neoepitopes and corresponding TCRs and provide first evidence for their application in cancer immunotherapy of FL-HCC. Citation Format: Jens Bauer, Natalie Köhler, Yacine Maringer, Philip Bucher, Tatjana Bilich, Melissa Zwick, Severin Dicks, Annika Nelde, Marissa Dubbelaar, Jonas Scheid, Marcel Wacker, Jonas J. Heitmann, Sarah Schroeder, Jonas Rieth, Monika Denk, Marion Richter, Reinhild Klein, Irina Bonzheim, Julia Luibrand, Ursula Holzer, Martin Ebinger, Ines B. Brecht, Michael Bitzer, Melanie Boerries, Helmut R. Salih, Hans-Georg Rammensee, Stephan Hailfinger, Juliane S. Walz. The oncogenic fusion protein DNAJB1-PRKACA can be actively targeted by peptide-based immunotherapy in fibrolamellar hepatocellular carcinoma [abstract]. In: Proceedings of the American Association for Cancer Research Annual Meeting 2022; 2022 Apr 8-13. Philadelphia (PA): AACR; Cancer Res 2022;82(12_Suppl):Abstract nr 2008.
- Published
- 2022
- Full Text
- View/download PDF
22. Classification and nomenclature of metacaspases and paracaspases: no more confusion with caspases
- Author
-
Christiane Funk, Vanina E. Alvarez, Heinz D. Osiewacz, Juan José Cazzulo, Simon Stael, Boris Zhivotovsky, Chang Jae Choi, Frank Madeo, Jens Staal, Kailash C. Pandey, Lynn A. Megeney, Yigong Shi, Magali Casanova, Andrei Smertenko, Maurício F.M. Machado, Eric Lam, Renier A. L. van der Hoorn, Juergen Ruland, Ilana Berman-Frank, Panagiotis N. Moschou, Peter V. Bozhkov, Jeremy C. Mottram, Kay D. Bidle, Jerry Ståhlberg, Rudi Beyaert, Christopher M. Overall, Frédéric Bornancin, Kris Gevaert, Margot Thome, Assaf Vardi, Núria S. Coll, Patrick Gallois, Frank Van Breusegem, Thomas Nyström, Vishva M. Dixit, Marko Dolinar, Maria F. Suarez, Stephan Hailfinger, Nicolas Fasel, Emilio Gutierrez-Beltran, John A. Berges, Anna Linusson, Hannele Tuominen, Daniel Krappmann, Guy S. Salvesen, Marina Klemenčič, Elena A. Minina, Eugene V. Koonin, Canaan, Stephane, Swedish University of Agricultural Sciences (SLU), Universiteit Gent = Ghent University (UGENT), Universidad Nacional de San Martin (UNSAM), University of Wisconsin - Milwaukee, University of Haifa [Haifa], Rutgers, The State University of New Jersey [New Brunswick] (RU), Rutgers University System (Rutgers), Novartis Institutes for BioMedical Research (NIBR), Laboratoire d'ingénierie des systèmes macromoléculaires (LISM), Aix Marseille Université (AMU)-Institut National de la Santé et de la Recherche Médicale (INSERM)-Centre National de la Recherche Scientifique (CNRS), University of Texas at Austin [Austin], Centre for Research in Agricultural Genomics (CRAG), Genentech, Inc., Genentech, Inc. [San Francisco], University of Ljubljana, Université de Lausanne = University of Lausanne (UNIL), Umeå University, Universidade de Mogi das Cruces = University of Mogi das Cruzes (UMC), Karl-Franzens-Universität Graz, University of Ottawa [Ottawa], Institute of Molecular Biology and Biotechnology (IMBB-FORTH), Foundation for Research and Technology - Hellas (FORTH), University of York [York, UK], University of Gothenburg (GU), Goethe-University Frankfurt am Main, University of British Columbia (UBC), National Institute of Malaria Research [New Dehli, Inde] (NIMR), Indian Council of Medical Research [New Dehli] (ICMR), Technische Universität Munchen - Université Technique de Munich [Munich, Allemagne] (TUM), Sanford Burnham Prebys Medical Discovery Institute, Westlake University [Zhejiang], Washington State University (WSU), Universidad de Málaga [Málaga] = University of Málaga [Málaga], University of Oxford, Weizmann Institute of Science [Rehovot, Israël], Lomonosov Moscow State University (MSU), Knut and Alice Wallenberg Foundation, Swedish University of Agricultural Sciences and Linnean Center for Plant Biology, Universiteit Gent = Ghent University [Belgium] (UGENT), Centre National de la Recherche Scientifique (CNRS)-Institut National de la Santé et de la Recherche Médicale (INSERM)-Aix Marseille Université (AMU), University of Lausanne (UNIL), Karl-Franzens-Universität [Graz, Autriche], University of Oxford [Oxford], University of Graz, and Technical University of Munich (TUM)
- Subjects
Consensus ,METACASPASES ,Protein Conformation ,[SDV]Life Sciences [q-bio] ,Computational biology ,Article ,purl.org/becyt/ford/1 [https] ,Structure-Activity Relationship ,03 medical and health sciences ,0302 clinical medicine ,Terminology as Topic ,medicine ,Animals ,Humans ,CRYSTAL-STRUCTURE ,purl.org/becyt/ford/1.6 [https] ,SPECIFICITY ,Molecular Biology ,Nomenclature ,Caspase ,PARACASPASES ,ComputingMilieux_MISCELLANEOUS ,Plant Proteins ,030304 developmental biology ,Confusion ,0303 health sciences ,biology ,MALT1 ,Biology and Life Sciences ,Cell Biology ,3. Good health ,PROTEASES ,[SDV] Life Sciences [q-bio] ,KEY ,Mucosa-Associated Lymphoid Tissue Lymphoma Translocation 1 Protein ,Caspases ,biology.protein ,CLAN CD ,medicine.symptom ,030217 neurology & neurosurgery - Abstract
Metacaspases and paracaspases are proteases that were first identified as containing a caspase-like structural fold (Uren et al., 2000). Like caspases, metacaspases and paracaspases are multifunctional proteins regulating diverse biological phenomena, such as aging, immunity, proteostasis, and programmed cell death. The broad phylogenetic distribution of metacaspases and paracaspases across all kingdoms of life and large variation of their biochemical and structural features complicate classification and annotation of the rapidly growing number of identified homologs. Establishment of an adequate classification and unified nomenclature of metacaspases and paracaspases is especially important to avoid frequent confusion of these proteases with caspases—a tenacious misnomer that unfortunately does not appear to decline with time. This Letter represents a consensus opinion of researchers studying different aspects of caspases, metacaspases, and paracaspases in various organisms, ranging from microbes to plants and animals., This work was supported by the Knut and Alice Wallenberg Foundation.
- Published
- 2020
- Full Text
- View/download PDF
23. IκBζ is a key transcriptional regulator of IL-36–driven psoriasis-related gene expression in keratinocytes
- Author
-
Klaus Schulze-Osthoff, Stephan Hailfinger, Daniela Kramer, André Hennig, Paula Grondona, Anne Müller, and Sebastian Lorscheid
- Subjects
STAT3 Transcription Factor ,keratinocytes ,0301 basic medicine ,IκBζ ,Proinflammatory cytokine ,Mice ,03 medical and health sciences ,IL36G ,Immunology and Inflammation ,0302 clinical medicine ,Psoriasis ,Gene expression ,Transcriptional regulation ,medicine ,Animals ,Humans ,STAT3 ,Adaptor Proteins, Signal Transducing ,Cell Proliferation ,Mice, Knockout ,Multidisciplinary ,biology ,NF-kappa B ,NFKBIZ ,Nuclear Proteins ,psoriasis ,Biological Sciences ,medicine.disease ,IL-36 ,030104 developmental biology ,medicine.anatomical_structure ,Gene Expression Regulation ,Cancer research ,biology.protein ,Tumor necrosis factor alpha ,Keratinocyte ,Interleukin-1 ,Signal Transduction ,030215 immunology - Abstract
Significance Psoriasis is an autoinflammatory disease characterized by cytokine-driven keratinocyte proliferation and infiltration of immune cells. While IL-17A and TNFα are established targets in psoriasis therapy, IL-36 is emerging as an important cytokine in this disease. The mechanisms of IL-36–driven proinflammatory responses are largely unknown. Here we identified IκBζ, a transcriptional regulator of selective NF-κB target genes, as a crucial mediator of IL-36 action. In keratinocytes, IκBζ was required for the expression of several psoriasis-related cytokines and chemokines. Moreover, genetic deletion of IκBζ prevented IL-36–mediated dermatitis induction in mice. Since IκBζ is essential not only for IL-36 but also for IL-17 signaling, our results suggest that inhibition of IκBζ function could be a future strategy in psoriasis therapy., Proinflammatory cytokine signaling in keratinocytes plays a crucial role in the pathogenesis of psoriasis, a skin disease characterized by hyperproliferation and abnormal differentiation of keratinocytes and infiltration of inflammatory cells. Although IL-17A and TNFα are effective therapeutic targets in psoriasis, IL-36 has recently emerged as a proinflammatory cytokine. However, little is known about IL-36 signaling and its downstream transcriptional responses. Here, we found that exposure of keratinocytes to IL-36 induced the expression of IκBζ, an atypical IκB member and a specific transcriptional regulator of selective NF-κB target genes. Induction of IκBζ by IL-36 was mediated by NF-κB and STAT3. In agreement, IL-36–mediated induction of IκBζ was found to be required for the expression of various psoriasis-related genes involved in inflammatory signaling, neutrophil chemotaxis, and leukocyte activation. Importantly, IκBζ-knockout mice were protected against IL-36–mediated dermatitis, accompanied by reduced proinflammatory gene expression, decreased immune cell infiltration, and a lack of keratinocyte hyperproliferation. Moreover, expression of IκBζ mRNA was highly up-regulated in biopsies of psoriasis patients where it coincided with IL36G levels. Thus our results uncover an important role for IκBζ in IL-36 signaling and validate IκBζ as an attractive target for psoriasis therapy.
- Published
- 2018
- Full Text
- View/download PDF
24. YB-1 Expression and Phosphorylation Regulate Tumorigenicity and Invasiveness in Melanoma by Influencing EMT
- Author
-
Peter R. Mertens, Birgit Sauer, Birgit Schittek, Heike Niessner, Tobias Sinnberg, Stephan Forchhammer, Cornelia Grimmel, Sandra E. Dunn, Corinna Kosnopfel, Christian Busch, Claus Garbe, Stephan Hailfinger, and Anja Schmitt
- Subjects
0301 basic medicine ,Cancer Research ,Epithelial-Mesenchymal Transition ,Skin Neoplasms ,Carcinogenesis ,Cell ,Chick Embryo ,Biology ,Mice ,03 medical and health sciences ,0302 clinical medicine ,Cell Movement ,Cell Line, Tumor ,medicine ,Animals ,Humans ,Neoplasm Invasiveness ,Translation factor ,Melanoma ,Molecular Biology ,Cell Proliferation ,Regulation of gene expression ,Neural crest ,medicine.disease ,Xenograft Model Antitumor Assays ,Gene Expression Regulation, Neoplastic ,030104 developmental biology ,medicine.anatomical_structure ,Oncology ,Cell culture ,030220 oncology & carcinogenesis ,Cutaneous melanoma ,Cancer cell ,Cancer research ,Y-Box-Binding Protein 1 - Abstract
Cutaneous melanoma represents one of the most aggressive human tumor entities possessing a high tendency to metastasize. Cancer cells frequently exploit a highly conserved developmental program, the epithelial-to-mesenchymal transition (EMT), to gain migratory and invasive properties promoting their metastatic spread. Cytoplasmic localization of the oncogenic transcription and translation factor Y-box binding protein 1 (YB-1) is a powerful inducer of EMT in breast carcinoma cells. Interestingly, EMT-like processes have also been observed in cutaneous melanoma despite its neural crest origin. Here, increased expression of YB-1 negatively affects patient survival in malignant melanoma and promotes melanoma cell tumorigenicity both in vitro and in vivo. Intriguingly, this effect seems to be mainly mediated by cytoplasmic YB-1 that does not exhibit phosphorylation at serine-102 (S102). Moreover, S102 unphosphorylated YB-1 enhances the migratory and invasive potential of human melanoma cells in two-dimensional (2D) and three-dimensional (3D) culture systems and facilitates acquisition of a mesenchymal-like invasive phenotype in the chick embryo model. Collectively, these data demonstrate that the cytoplasmic activity of YB-1 stimulates tumorigenicity and metastatic potential of melanoma cells by promoting EMT-like properties. Implications: This study reveals for the first time that YB-1 efficiently drives tumorigenicity and invasiveness of melanoma cells in its S102 unphosphorylated cytoplasmic state and that YB-1 expression represents a negative prognostic factor in primary melanoma patients. Mol Cancer Res; 16(7); 1149–60. ©2018 AACR.
- Published
- 2018
- Full Text
- View/download PDF
25. Human melanoma cells resistant to MAPK inhibitors can be effectively targeted by inhibition of the p90 ribosomal S6 kinase
- Author
-
Birgit Schittek, Heike Niessner, Claus Garbe, Tobias Sinnberg, Corinna Kosnopfel, Andrea Forschner, Birgit Sauer, Stephan Hailfinger, Anja Schmitt, and Elena Makino
- Subjects
Proto-Oncogene Proteins B-raf ,0301 basic medicine ,MAPK/ERK pathway ,Indoles ,therapy resistance ,Cell Survival ,Antineoplastic Agents ,YB-1 ,Ribosomal Protein S6 Kinases, 90-kDa ,03 medical and health sciences ,0302 clinical medicine ,Downregulation and upregulation ,Cell Line, Tumor ,melanoma ,medicine ,Humans ,Vemurafenib ,Protein Kinase Inhibitors ,Trametinib ,Sulfonamides ,business.industry ,Effector ,MEK inhibitor ,Melanoma ,medicine.disease ,G2 Phase Cell Cycle Checkpoints ,MAPK inhibition ,030104 developmental biology ,Oncology ,Drug Resistance, Neoplasm ,030220 oncology & carcinogenesis ,Immunology ,Cancer research ,Y-Box-Binding Protein 1 ,Mitogen-Activated Protein Kinases ,Signal transduction ,business ,Research Paper ,p90 ribosomal S6 kinase ,Signal Transduction ,medicine.drug - Abstract
The clinical availability of small molecule inhibitors specifically targeting mutated BRAF marked a significant breakthrough in melanoma therapy. Despite a dramatic anti-tumour activity and improved patient survival, rapidly emerging resistance, however, greatly limits the clinical benefit. The majority of the already described resistance mechanisms involve a reactivation of the MAPK signalling pathway. The p90 ribosomal S6 kinase (RSK), a downstream effector of the MAPK signalling cascade, has been reported to enhance survival of melanoma cells in response to chemotherapy. Here, we can show that RSK activity is significantly increased in human melanoma cells with acquired resistance to the BRAFV600E/K inhibitor vemurafenib. Interestingly, inhibition of RSK signalling markedly impairs the viability of vemurafenib resistant melanoma cells and is effective both in two-dimensional and in three-dimensional culture systems, especially in a chronic, long-term application. The effect of RSK inhibition can be partly replicated by downregulation of the well-known RSK target, Y-box binding protein 1 (YB-1). Intriguingly, RSK inhibition also retains its efficacy in melanoma cells with combined resistance to vemurafenib and the MEK inhibitor trametinib. These data suggest that active RSK signalling might be an attractive novel therapeutic target in melanoma with acquired resistance to MAPK pathway inhibitors.
- Published
- 2017
- Full Text
- View/download PDF
26. Ferroptotic pores induce Ca2+ fluxes and ESCRT-III activation to modulate cell death kinetics
- Author
-
Lohans Pedrera, Stephan Hailfinger, Anja Schmitt, Rafael A. Espiritu, Ana J. García-Sáez, and Uris Ros
- Subjects
Lipid peroxidation ,Cytosol ,Bursting ,Programmed cell death ,chemistry.chemical_compound ,Membrane ,medicine.anatomical_structure ,chemistry ,Kinetics ,Cell ,medicine ,ESCRT ,Cell biology - Abstract
SummaryFerroptosis is an iron-dependent form of regulated necrosis associated with lipid peroxidation. Despite its key role in the inflammatory outcome of ferroptosis, little is known about the molecular events leading to the disruption of the plasma membrane during this type of cell death. Here we show that a sustained increase in cytosolic Ca2+ is a hallmark of ferroptosis that precedes complete bursting of the cell. We report that plasma membrane damage leading to ferroptosis is associated with membrane nanopores of few nanometers in radius and that ferroptosis, but not lipid peroxidation, can be delayed by osmoprotectants. Importantly, Ca2+ fluxes during ferroptosis correlate with the activation of ESCRT-III-mediated membrane repair, which counterbalances the kinetics of cell death and modulates the inflammatory signature of ferroptosis. Our findings with ferroptosis provide a unifying concept that sustained high levels of cytosolic Ca2+ prior to plasma membrane disruption are a common feature of regulated necrosis and position ESCRT-III as a general protective mechanism in these inflammatory cell death pathways.
- Published
- 2019
- Full Text
- View/download PDF
27. Keratinocyte-derived IκBζ drives psoriasis and associated systemic inflammation
- Author
-
Jessica Löffler, Knut Schäkel, Daniela Kramer, Anne Müller, Claudia Resch, Stephan Hailfinger, Ari Waisman, Florian C. Kurschus, Sebastian Lorscheid, Philip Bucher, and Klaus Schulze-Osthoff
- Subjects
0301 basic medicine ,Keratinocytes ,Male ,Autoimmune diseases ,Inflammation ,Mice, Transgenic ,Autoimmunity ,Dermatology ,Systemic inflammation ,medicine.disease_cause ,Proinflammatory cytokine ,03 medical and health sciences ,Mice ,0302 clinical medicine ,Psoriasis ,medicine ,Animals ,Cells, Cultured ,Adaptor Proteins, Signal Transducing ,Skin ,Innate immunity ,Innate immune system ,business.industry ,Interleukin-17 ,General Medicine ,medicine.disease ,CXCL2 ,030104 developmental biology ,medicine.anatomical_structure ,030220 oncology & carcinogenesis ,Cancer research ,Female ,medicine.symptom ,Keratinocyte ,business ,Research Article - Abstract
The transcriptional activator IκBζ is a key regulator of psoriasis, but which cells mediate its pathogenic effect remains unknown. Here we found that IκBζ expression in keratinocytes triggers not only skin lesions but also systemic inflammation in mouse psoriasis models. Specific depletion of IκBζ in keratinocytes was sufficient to suppress the induction of imiquimod- or IL-36–mediated psoriasis. Moreover, IκBζ ablation in keratinocytes prevented the onset of psoriatic lesions and systemic inflammation in keratinocyte-specific IL-17A–transgenic mice. Mechanistically, this psoriasis protection was mediated by IκBζ deficiency in keratinocytes abrogating the induction of specific proinflammatory target genes, including Cxcl5, Cxcl2, Csf2, and Csf3, in response to IL-17A or IL-36. These IκBζ-dependent genes trigger the generation and recruitment of neutrophils and monocytes that are needed for skin inflammation. Consequently, our data uncover a surprisingly pivotal role of keratinocytes and keratinocyte-derived IκBζ as key mediators of psoriasis and psoriasis-related systemic inflammation., Deletion of IκBζ in keratinocytes is sufficient to abrogate psoriasis induction in mouse models due to changes in transcription of keratinocyte-derived chemo- and cytokines.
- Published
- 2019
28. Targeting chronic NFAT activation with calcineurin inhibitors in diffuse large B-cell lymphoma
- Author
-
Daniela Kramer, Paula Grondona, Pavel Klener, Wendan Xu, Tabea Erdmann, Michael Grau, Anja Schmitt, Christoph Schürch, Edgar Serfling, Caroline Schönfeld, Klaus Schulze-Osthoff, Stephan Hailfinger, Philip Bucher, Claudia Lengerke, Georg Lenz, and Myroslav Zapukhlyak
- Subjects
Immunology ,Calcineurin Inhibitors ,Aggressive lymphoma ,Biochemistry ,immune system diseases ,Cyclosporin a ,hemic and lymphatic diseases ,Tumor Cells, Cultured ,Medicine ,Humans ,Transcription factor ,Cell Proliferation ,NFATC Transcription Factors ,business.industry ,Calcineurin ,Germinal center ,NFAT ,Cell Biology ,Hematology ,medicine.disease ,Lymphoma ,Proto-Oncogene Proteins c-bcl-2 ,Cancer research ,Myeloid Cell Leukemia Sequence 1 Protein ,Calcium ,Lymphoma, Large B-Cell, Diffuse ,business ,Diffuse large B-cell lymphoma ,BLOOD Commentary - Abstract
Diffuse large B-cell lymphoma (DLBCL) represents the most common adult lymphoma and can be divided into 2 major molecular subtypes: the germinal center B-cell-like and the aggressive activated B-cell-like (ABC) DLBCL. Previous studies suggested that chronic B-cell receptor signaling and increased NF-κB activation contribute to ABC DLBCL survival. Here we show that the activity of the transcription factor NFAT is chronically elevated in both DLBCL subtypes. Surprisingly, NFAT activation is independent of B-cell receptor signaling, but mediated by an increased calcium flux and calcineurin-mediated dephosphorylation of NFAT. Intriguingly, although NFAT is activated in both DLBCL subtypes, long-term calcineurin inhibition with cyclosporin A or FK506, both clinically approved drugs, triggers potent cytotoxicity specifically in ABC DLBCL cells. The antitumor effects of calcineurin inhibitors are associated with the reduced expression of c-Jun, interleukin-6, and interleukin-10, which were identified as NFAT target genes that are particularly important for the survival of ABC DLBCL. Furthermore, calcineurin blockade synergized with BCL-2 and MCL-1 inhibitors in killing ABC DLBCL cells. Collectively, these findings identify constitutive NFAT signaling as a crucial functional driver of ABC DLBCL and highlight calcineurin inhibition as a novel strategy for the treatment of this aggressive lymphoma subtype.
- Published
- 2019
29. Threonine Phosphorylation of IκBζ Mediates Inhibition of Selective Proinflammatory Target Genes
- Author
-
Caroline Schönfeld, Klaus Schulze-Osthoff, Shabaz Mohammed, Sabrina Liberatori, Barbara Streibl, André Hennig, Daniela Kramer, Philip Bucher, Stephan Hailfinger, Frank Essmann, Paula Grondona, Anne Müller, and Anja Schmitt
- Subjects
0301 basic medicine ,Keratinocytes ,Threonine ,Transcription, Genetic ,Primary Cell Culture ,Histone Deacetylase 1 ,Dermatology ,Biology ,Biochemistry ,Proinflammatory cytokine ,03 medical and health sciences ,0302 clinical medicine ,Transcription (biology) ,Coactivator ,Humans ,Phosphorylation ,Promoter Regions, Genetic ,Molecular Biology ,Transcription factor ,Cells, Cultured ,Adaptor Proteins, Signal Transducing ,Skin ,Zymosan ,Promoter ,Fungal Polysaccharides ,Cell Biology ,HDAC1 ,Cell biology ,030104 developmental biology ,Gene Expression Regulation ,030220 oncology & carcinogenesis ,Host-Pathogen Interactions ,Inflammation Mediators ,Mitogen-Activated Protein Kinases ,Protein Processing, Post-Translational - Abstract
Transcription factors of the NF-κB family play a crucial role for immune responses by activating the expression of chemokines, cytokines, and antimicrobial peptides involved in pathogen clearance. IκBζ, an atypical nuclear IκB protein and selective coactivator of particular NF-κB target genes, has recently been identified as an essential regulator for skin immunity. This study discovered that IκBζ is strongly induced in keratinocytes that sense the fungal glucan zymosan A. Additionally, IκBζ is essential for the optimal expression of proinflammatory genes, such as IL6, CXCL5, IL1B, or S100A9. Moreover, this study found that IκBζ was not solely regulated on the transcriptional level but also by phosphorylation events. This study identified several IκBζ phosphorylation sites, including a conserved cluster of threonine residues located in the N-terminus of the protein, which can be phosphorylated by MAPKs. Surprisingly, IκBζ phosphorylation at this threonine cluster promoted the recruitment of histone deacetylase 1 to specific target gene promoters and, thus, negatively controlled transcription. Taken together, this study proposes a model of how an antifungal response translates to the expression of proinflammatory cytokines and highlights an additional layer of complexity in the regulation of the NF-κB responses in keratinocytes.
- Published
- 2019
30. The Atypical Inhibitor of NF-κB, IκBζ, Controls Macrophage Interleukin-10 Expression
- Author
-
Frank Essmann, Sebastian Hörber, Klaus Schulze-Osthoff, Wolfgang S. Lieb, Sebastian Lorscheid, Stephan Hailfinger, and Dominic G. Hildebrand
- Subjects
0301 basic medicine ,medicine.medical_treatment ,Immunology ,Immunoblotting ,Gene Expression ,Inflammation ,Biology ,Biochemistry ,Cell Line ,03 medical and health sciences ,chemistry.chemical_compound ,Gene expression ,medicine ,Animals ,Promoter Regions, Genetic ,Molecular Biology ,Cells, Cultured ,Adaptor Proteins, Signal Transducing ,Mice, Knockout ,Reverse Transcriptase Polymerase Chain Reaction ,Macrophages ,NF-kappa B ,Nuclear Proteins ,Interleukin ,NF-κB ,Cell Biology ,Fibroblasts ,Embryo, Mammalian ,NFKB1 ,Interleukin-10 ,Mice, Inbred C57BL ,Interleukin 10 ,030104 developmental biology ,Cytokine ,chemistry ,Macrophages, Peritoneal ,Cancer research ,Female ,Ectopic expression ,Inflammation Mediators ,medicine.symptom - Abstract
Macrophages constitute a first line of pathogen defense by triggering a number of inflammatory responses and the secretion of various pro-inflammatory cytokines. Recently, we and others found that IκBζ, an atypical IκB family member and transcriptional coactivator of selected NF-κB target genes, is essential for macrophage expression of a subset of pro-inflammatory cytokines, such as IL-6, IL-12, and CCL2. Despite defective pro-inflammatory cytokine expression, however, IκBζ-deficient mice develop symptoms of chronic inflammation. To elucidate this discrepancy, we analyzed a regulatory role of IκBζ for the expression of anti-inflammatory cytokines and identified IκBζ as an essential activator of IL-10 expression. LPS-challenged peritoneal and bone marrow-derived macrophages from IκBζ-deficient mice revealed strongly decreased transcription and secretion of IL-10 compared with wild-type mice. Moreover, ectopic expression of IκBζ was sufficient to stimulate Il10 transcription. On the molecular level, IκBζ directly activated the Il10 promoter at a proximal κB site and was required for the transcription-enhancing trimethylation of histone 3 at lysine 4. Together, our findings show for the first time the IκBζ-dependent expression of an anti-inflammatory cytokine that is crucial in controlling immune responses.
- Published
- 2016
- Full Text
- View/download PDF
31. Release of Immunomodulatory Ebola Virus Glycoprotein-Containing Microvesicles Is Suppressed by Tetherin in a Species-Specific Manner
- Author
-
Martin Schaller, Julia Nehls, Daniela Kramer, Michael Schindler, Birgit Fehrenbacher, Markus Hoffmann, Caroline Schönfeld, Brigitte Maurer, Ramona Businger, Stefan Pöhlmann, Stephan Hailfinger, and Constantin Brinkmann
- Subjects
0301 basic medicine ,Ebola Virus ,Antiviral Immune Response ,Exosome ,Glycoprotein ,Immune Evasion ,Immune Modulation ,Microvesicles ,Neutralizing Antibody ,Tetherin ,Virosome ,Biology ,medicine.disease_cause ,General Biochemistry, Genetics and Molecular Biology ,Immunomodulation ,03 medical and health sciences ,0302 clinical medicine ,Immune system ,medicine ,Humans ,Secretion ,lcsh:QH301-705.5 ,Virus Release ,Glycoproteins ,chemistry.chemical_classification ,Ebolavirus ,Ebola virus ,Bone Marrow Stromal Antigen 2 ,Virology ,030104 developmental biology ,lcsh:Biology (General) ,chemistry ,030217 neurology & neurosurgery - Abstract
Summary: The Ebola virus glycoprotein (EBOV-GP) forms GP-containing microvesicles, so-called virosomes, which are secreted from GP-expressing cells. However, determinants of GP-virosome release and their functionality are poorly understood. We characterized GP-mediated virosome formation and delineated the role of the antiviral factor tetherin (BST2, CD317) in this process. Residues in the EBOV-GP receptor-binding domain (RBD) promote GP-virosome secretion, while tetherin suppresses GP-virosomes by interactions involving the GP-transmembrane domain. Tetherin from multiple species interfered with GP-virosome release, and tetherin from the natural fruit bat reservoir showed the highest inhibitory activity. Moreover, analyses of GP from various ebolavirus strains, including the EBOV responsible for the West African epidemic, revealed the most efficient GP-virosome formation by highly pathogenic ebolaviruses. Finally, EBOV-GP-virosomes were immunomodulatory and acted as decoys for EBOV-neutralizing antibodies. Our results indicate that GP-virosome formation might be a determinant of EBOV immune evasion and pathogenicity, which is suppressed by tetherin. : Nehls et al. demonstrate that the glycoprotein of the highly pathogenic Ebola virus is incorporated into secretory vesicles, called GP-virosomes, to dampen the immune response and capture neutralizing antibodies. The lack of replication competence and the incorporation of antigenically intact GP might qualify GP-virosomes as safe vaccine candidates. Keywords: Ebola virus, glycoprotein, microvesicles, virosome, exosome, tetherin, immune modulation, immune evasion, antiviral immune response, neutralizing antibody
- Published
- 2019
- Full Text
- View/download PDF
32. The paracaspase MALT1 dampens NF-κB signalling by cleaving the LUBAC subunit HOIL-1
- Author
-
Stephan Hailfinger, Anja Schmitt, and Klaus Schulze-Osthoff
- Subjects
0301 basic medicine ,Ubiquitin-Protein Ligases ,Protein subunit ,Biochemistry ,03 medical and health sciences ,0302 clinical medicine ,Humans ,Lymphocytes ,Molecular Biology ,Transcription factor ,Caspase ,biology ,NF-kappa B ,Cell Biology ,Paracaspase ,NFKB1 ,Molecular biology ,Neoplasm Proteins ,Ubiquitin ligase ,Protein Subunits ,MALT1 ,030104 developmental biology ,Mucosa-Associated Lymphoid Tissue Lymphoma Translocation 1 Protein ,Caspases ,030220 oncology & carcinogenesis ,biology.protein ,Signal transduction ,Signal Transduction ,Transcription Factors - Published
- 2016
- Full Text
- View/download PDF
33. Targeting B-cell lymphomas with inhibitors of the MALT1 paracaspase
- Author
-
Georg Lenz, Stephan Hailfinger, and Margot Thome
- Subjects
Lymphoma, B-Cell ,Protease ,medicine.medical_treatment ,Point mutation ,Lymphocyte proliferation ,Paracaspase ,Biology ,Biochemistry ,Small molecule ,Neoplasm Proteins ,3. Good health ,Analytical Chemistry ,MALT1 ,medicine.anatomical_structure ,Mucosa-Associated Lymphoid Tissue Lymphoma Translocation 1 Protein ,Caspases ,medicine ,Cancer research ,Animals ,Humans ,Enzyme Inhibitors ,B cell ,Function (biology) - Abstract
The paracaspase MALT1 is an Arg-specific protease that cleaves multiple substrates to promote lymphocyte proliferation and survival. The catalytic activity of MALT1 is normally tightly regulated by antigen receptor triggering, which promotes MALT1 activation by its inducible monoubiquitination-dependent dimerization. Constitutive MALT1 activity is a hallmark of specific subsets of B-cell lymphomas, which are characterized by chromosomal translocations or point mutations that activate MALT1 or its upstream regulators. Recent findings suggest that such lymphomas may be sensitive to treatment with MALT1 inhibitors. Here we review recent progress in the understanding of MALT1 function and regulation, and the development of small molecule MALT1 inhibitors for therapeutic applications.
- Published
- 2014
- Full Text
- View/download PDF
34. Human neutrophils release bioactive inflammasome complexes; relevance for cystic fibrosis
- Author
-
Hartl Dominik, Gassen Nils, Julie Laval, Hector Andreas, Martina Bakele, and Stephan Hailfinger
- Subjects
medicine.diagnostic_test ,Inflammation ,Inflammasome ,Biology ,Peripheral blood mononuclear cell ,Paracrine signalling ,Cytosol ,Western blot ,Immunology ,medicine ,Extracellular ,medicine.symptom ,Autocrine signalling ,medicine.drug - Abstract
Rationale. Massive neutrophil recruitment and their secreted products are hallmarks of cystic fibrosis (CF) airway disease. We previously described that healthy neutrophils store key inflammasome components and release Il-1s, found at high levels in CF airways (Bakele et al, 2014). Moreover, a recent study identified that inflammasomes, usually thought to be cytosolic multi-protein complexes, could be secreted and act as extracellular danger signal to amplify inflammation (Baroja-Mazo et al, 2014). Aims. We attempt to characterize neutrophil inflammasome components and their release to evaluate the potential impact in CF lung disease. Methods. We activated inflammasomes in isolated neutrophils and PBMCs (LPS +/- ATP or Nigericin) and also performed autocrine/paracrine stimulation to evaluate the impact on neutrophil/PBMC activity. Collected supernatants were analysed by western blot, enzymatic assays and ELISA to detect secreted inflammasome proteins and caspases9 activity and Il-1s levels. Finally, we assessed inflammasome activity in CF neutrophils and fluids, which we compared to healthy control samples. Results. We demonstrated that human neutrophils release distinct oligomeric inflammasome-associated particles. These secreted complexes remained enzymatically active, interacted physically with caspase-1 and amplified the inflammatory response. In addition, we described inflammasome modulation in CF samples. Conclusions. These findings support a model where neutrophils release inflammasome components as an endogenous danger signal and therefore amplify the inflammatory response, suggesting potential anti-inflammatory therapeutic strategies in CF.
- Published
- 2016
- Full Text
- View/download PDF
35. CARD14-Mediated Activation of Paracaspase MALT1 in Keratinocytes: Implications for Psoriasis
- Author
-
Rudi Beyaert, Inna S. Afonina, Stephan Hailfinger, Elien Van Nuffel, Klaus Schulze-Osthoff, and Anja Schmitt
- Subjects
0301 basic medicine ,Scaffold protein ,Keratinocytes ,Dermatology ,medicine.disease_cause ,Biochemistry ,03 medical and health sciences ,Mice ,medicine ,Animals ,Humans ,Psoriasis ,Molecular Biology ,Caspase ,Adaptor Proteins, Signal Transducing ,Mutation ,biology ,Membrane Proteins ,Cell Biology ,Paracaspase ,medicine.disease ,B-Cell CLL-Lymphoma 10 Protein ,BCL10 ,Cell biology ,Neoplasm Proteins ,CARD Signaling Adaptor Proteins ,MALT1 ,030104 developmental biology ,Guanylate Cyclase ,Mucosa-Associated Lymphoid Tissue Lymphoma Translocation 1 Protein ,Caspases ,Cancer research ,biology.protein ,Generalized pustular psoriasis ,Signal transduction ,Signal Transduction - Abstract
Mutations in caspase recruitment domain-containing protein 14 (CARD14) have been linked to susceptibility to psoriasis. CARD14 is an intracellular scaffold protein that regulates proinflammatory gene expression. Recent studies have offered novel insights into the mechanisms of CARD14-mediated signaling in keratinocytes and the molecular impact of psoriasis-associated CARD14 mutations. CARD14 forms a signaling complex with BCL10 and the paracaspase MALT1, and this process is enhanced upon pathogenic CARD14 mutation, culminating in the activation of MALT1 protease activity and psoriasis-associated gene expression. This review summarizes the current knowledge of CARD14/MALT1-mediated signaling in keratinocytes and its therapeutic implications in psoriasis.
- Published
- 2016
36. MCL1 is deregulated in subgroups of diffuse large B-cell lymphoma
- Author
-
George Deeb, Francisco J. Hernandez-Ilizaliturri, Alexandar Tzankov, Cory Mavis, Margot Thome, Sören-Sebastian Wenzel, Michael Grau, Hannelore Madle, Annette Wolf, Bernd Dörken, Stephan Hailfinger, Georg Lenz, Stefan Dirnhofer, and Peter Lenz
- Subjects
Cancer Research ,Myeloid ,Antineoplastic Agents ,Biology ,Real-Time Polymerase Chain Reaction ,03 medical and health sciences ,chemistry.chemical_compound ,0302 clinical medicine ,hemic and lymphatic diseases ,medicine ,Humans ,MCL1 ,030304 developmental biology ,Comparative Genomic Hybridization ,0303 health sciences ,Gene Expression Profiling ,Germinal center ,Hematology ,medicine.disease ,Immunohistochemistry ,3. Good health ,Lymphoma ,Myeloid Cell Leukemia Sequence 1 Protein ,Gene expression profiling ,medicine.anatomical_structure ,Proto-Oncogene Proteins c-bcl-2 ,Oncology ,chemistry ,030220 oncology & carcinogenesis ,Cancer research ,Lymphoma, Large B-Cell, Diffuse ,Diffuse large B-cell lymphoma ,Obatoclax - Abstract
Myeloid cell leukemia 1 (MCL1) is an anti apoptotic member of the BCL2 family that is deregulated in various solid and hematological malignancies. However its role in the molecular pathogenesis of diffuse large B cell lymphoma (DLBCL) is unclear. We analyzed gene expression profiling data from 350 DLBCL patient samples and detected that activated B cell like (ABC) DLBCLs express MCL1 at significantly higher levels compared with germinal center B cell like DLBCL patient samples (P=2.7 × 10( 10)). Immunohistochemistry confirmed high MCL1 protein expression predominantly in ABC DLBCL in an independent patient cohort (n=249; P=0.001). To elucidate molecular mechanisms leading to aberrant MCL1 expression we analyzed array comparative genomic hybridization data of 203 DLBCL samples and identified recurrent chromosomal gains/amplifications of the MCL1 locus that occurred in 26 of ABC DLBCLs. In addition aberrant STAT3 signaling contributed to high MCL1 expression in this subtype. Knockdown of MCL1 as well as treatment with the BH3 mimetic obatoclax induced apoptotic cell death in MCL1 positive DLBCL cell lines. In summary MCL1 is deregulated in a significant fraction of ABC DLBCLs and contributes to therapy resistance. These data suggest that specific inhibition of MCL1 might be utilized therapeutically in a subset of DLBCLs.Leukemia advance online publication 18 January 2013; doi:10.1038/leu.2012.367.
- Published
- 2012
- Full Text
- View/download PDF
37. Malt1-dependent RelB cleavage promotes canonical NF-κB activation in lymphocytes and lymphoma cell lines
- Author
-
Georg Lenz, Christiane Pelzer, Montserrat Guzzardi, Chantal Décaillet, Bernd Dörken, Margot Thome, Stephan Hailfinger, Maike Jaworski, Peter Lenz, Jean-Enno Charton, Hendrik Nogai, Michael Grau, and Katrin Cabalzar
- Subjects
Multidisciplinary ,RELA ,biology ,RELB ,Transcription Factor RelB ,Transcription Factor RelA ,NF-kappa B ,Biological Sciences ,Paracaspase ,Lymphocyte Activation ,NFKB1 ,Neoplasm Proteins ,MALT1 ,Mucosa-Associated Lymphoid Tissue Lymphoma Translocation 1 Protein ,Caspases ,Cell Line, Tumor ,Cancer research ,biology.protein ,Humans ,Lymphocytes ,Lymphoma, Large B-Cell, Diffuse ,Caspase - Abstract
The protease activity of the paracaspase Malt1 contributes to antigen receptor-mediated lymphocyte activation and lymphomagenesis. Malt1 activity is required for optimal NF-κB activation, but little is known about the responsible substrate(s). Here we report that Malt1 cleaved the NF-κB family member RelB after Arg-85. RelB cleavage induced its proteasomal degradation and specifically controlled DNA binding of RelA- or c-Rel–containing NF-κB complexes. Overexpression of RelB inhibited expression of canonical NF-κB target genes and led to impaired survival of diffuse large B-cell lymphoma cell lines characterized by constitutive Malt1 activity. These findings identify a central role for Malt1-dependent RelB cleavage in canonical NF-κB activation and thereby provide a rationale for the targeting of Malt1 in immunomodulation and cancer treatment.
- Published
- 2011
- Full Text
- View/download PDF
38. CARMA1- and MyD88-dependent activation of Jun/ATF-type AP-1 complexes is a hallmark of ABC diffuse large B-cell lymphomas
- Author
-
Alexandar Tzankov, Georg Lenz, Urban Novak, Yara Banz, Mélanie Juilland, Stephan Hailfinger, Tabea Erdmann, Montserrat Gonzalez, Michael Grau, Zala Jevnikar, and Margot Thome
- Subjects
0301 basic medicine ,JUNB ,Proto-Oncogene Proteins c-jun ,Immunology ,Activating transcription factor ,Biology ,Biochemistry ,03 medical and health sciences ,Jurkat Cells ,immune system diseases ,hemic and lymphatic diseases ,medicine ,Humans ,610 Medicine & health ,Transcription factor ,B cell ,Regulation of gene expression ,Activating Transcription Factor 3 ,Activating Transcription Factor 2 ,Activating Transcription Factor 2/genetics ,Activating Transcription Factor 2/metabolism ,Activating Transcription Factor 3/genetics ,Activating Transcription Factor 3/metabolism ,Activating Transcription Factors/genetics ,Activating Transcription Factors/metabolism ,CARD Signaling Adaptor Proteins/genetics ,CARD Signaling Adaptor Proteins/metabolism ,Gene Expression Regulation, Neoplastic ,Germinal Center/metabolism ,Germinal Center/pathology ,Guanylate Cyclase/genetics ,Guanylate Cyclase/metabolism ,Lymphoma, Large B-Cell, Diffuse/genetics ,Lymphoma, Large B-Cell, Diffuse/metabolism ,Myeloid Differentiation Factor 88/genetics ,Myeloid Differentiation Factor 88/metabolism ,Proto-Oncogene Proteins c-jun/genetics ,Proto-Oncogene Proteins c-jun/metabolism ,Transcription Factor AP-1/genetics ,Transcription Factor AP-1/metabolism ,Transcription Factors/genetics ,Transcription Factors/metabolism ,Germinal center ,Cell Biology ,Hematology ,Germinal Center ,Activating transcription factor 2 ,Activating Transcription Factors ,CARD Signaling Adaptor Proteins ,Transcription Factor AP-1 ,030104 developmental biology ,medicine.anatomical_structure ,Guanylate Cyclase ,Myeloid Differentiation Factor 88 ,Cancer research ,biology.protein ,570 Life sciences ,biology ,Lymphoma, Large B-Cell, Diffuse ,Transcription Factors - Abstract
A hallmark of the diffuse large B-cell lymphoma (DLBCL) of the activated B-cell (ABC) type, a molecular subtype characterized by adverse outcome, is constitutive activation of the transcription factor nuclear factor-κB (NF-κB), which controls expression of genes promoting cellular survival and proliferation. Much less, however, is known about the role of the transcription factor activator protein-1 (AP-1) in ABC DLBCL. Here, we show that AP-1, like NF-κB, was controlled by constitutive activation of the B-cell receptor signaling component caspase recruitment domain-containing membrane-associated guanylate kinase 1 (CARMA1) and/or the Toll-like receptor signaling component myeloid differentiation primary response gene 88 (MyD88) in ABC DLBCL cell lines. In contrast to germinal center (GC) B-cell (GCB) DLBCL, ABC DLBCL cell lines expressed high levels of the AP-1 family members c-Jun, JunB, and JunD, which formed heterodimeric complexes with the AP-1 family members activating transcription factor (ATF) 2, ATF3, and ATF7. Inhibition of these complexes by a dominant-negative approach led to impaired growth of a majority of ABC DLBCL cell lines. Individual silencing of c-Jun, ATF2, or ATF3 decreased cellular survival and revealed c-Jun/ATF2-dependent control of ATF3 expression. As a consequence, ATF3 expression was much higher in ABC vs GCB DLBCL cell lines. Samples derived from DLBCL patients showed a clear trend toward high and nuclear ATF3 expression in nodal DLBCL of the non-GC or ABC subtype. These findings identify the activation of AP-1 complexes of the Jun/ATF-type as an important element controlling the growth of ABC DLBCL.
- Published
- 2016
39. Regulation of P53 stability in p53 mutated human and mouse hepatoma cells
- Author
-
Maike Jaworski, Philip Marx-Stoelting, Michael Schwarz, Stephan Hailfinger, and Ines Wanke
- Subjects
Cyclin-Dependent Kinase Inhibitor p21 ,Cancer Research ,Carcinoma, Hepatocellular ,Tumor suppressor gene ,Ultraviolet Rays ,Ratón ,Blotting, Western ,Apoptosis ,Biology ,medicine.disease_cause ,law.invention ,Mice ,law ,Tumor Cells, Cultured ,medicine ,Animals ,Humans ,Immunoprecipitation ,RNA, Messenger ,chemistry.chemical_classification ,Messenger RNA ,Mutation ,Reactive oxygen species ,Reverse Transcriptase Polymerase Chain Reaction ,Liver Neoplasms ,Proto-Oncogene Proteins c-mdm2 ,Molecular biology ,Gene Expression Regulation, Neoplastic ,Oncology ,chemistry ,biology.protein ,Cancer research ,Suppressor ,Mdm2 ,Tumor Suppressor Protein p53 - Abstract
The tumor suppressor p53 is frequently mutated in cancer. We have investigated the regulation of P53 in p53 wild type mouse hepatoma cells (line 55.1c), in p53 heterozygeously mutated cells (56.1b) and in p53 defective cells (lines 56.1d, 70.4 and HUH7) under various experimental settings. The basal levels of P53 were low in 55.1c cells, but nuclear accumulation occurred upon UV-irradiation. Similarly, UV-exposure induced stabilization of P53 in the heterozygeously p53 mutated 56.1b hepatoma cells. By contrast, the 3 hepatoma lines, which lack transcriptionally active P53, demonstrated high basal nuclear concentrations of P53 protein and, unexpectedly, showed loss of P53 upon UV-irradiation. Expression of p53 mRNA was also decreased in p53 defective cells after 24 hr post UV-irradiation, which may be linked to induction of apoptosis of the irradiated cells under these conditions. Other stressors like H2O2 also mediated a decrease in P53 concentration in p53 defective cells. This effect occurred at very low concentrations and was already detectable 1–2 hr after exposure of cells. There were no signs of apoptosis of H2O2-exposed cells at this time point and no significant changes in p53 mRNA or MDM2 level. These unexpected findings indicate a new aspect related to regulation of P53 stability in cells with a defect in the tumor suppressor protein. © 2006 Wiley-Liss, Inc.
- Published
- 2007
- Full Text
- View/download PDF
40. IκBζ is a key driver in the development of psoriasis
- Author
-
Claus Johansen, Lars Iversen, Trine Bertelsen, Sebastian Lorscheid, Pernille Ommen, Maike Mose, Hanne Vinter, Stephan Hailfinger, and Klaus Schulze-Osthoff
- Subjects
Keratinocytes ,Regulator ,Inflammation ,Biology ,Animals, Genetically Modified ,Pathogenesis ,Mice ,Psoriasis ,Coactivator ,medicine ,Animals ,Humans ,Intradermal injection ,Gene ,Adaptor Proteins, Signal Transducing ,Imiquimod ,Multidisciplinary ,Interleukin-17 ,Nuclear Proteins ,medicine.disease ,Disease Models, Animal ,PNAS Plus ,Immunology ,Aminoquinolines ,Cytokines ,I-kappa B Proteins ,Tumor necrosis factor alpha ,medicine.symptom ,Signal Transduction - Abstract
Psoriasis is a common immune-mediated, chronic, inflammatory skin disease characterized by hyperproliferation and abnormal differentiation of keratinocytes and infiltration of inflammatory cells. Although TNFα- and IL-17A-targeting drugs have recently proven to be highly effective, the molecular mechanism underlying the pathogenesis of psoriasis remains poorly understood. We found that expression of the atypical IκB member IκB (inhibitor of NF-κB) ζ, a selective coactivator of particular NF-κB target genes, was strongly increased in skin of patients with psoriasis. Moreover, in human keratinocytes IκBζ was identified as a direct transcriptional activator of TNFα/IL-17A-inducible psoriasis-associated proteins. Using genetically modified mice, we found that imiquimod-induced psoriasis-like skin inflammation was completely absent in IκBζ-deficient mice, whereas skin inflammation was still inducible in IL-17A- and TNFα-deficient mice. IκBζ deficiency also conferred resistance against IL-23-induced psoriasis. In addition, local abrogation of IκBζ function by intradermal injection of IκBζ siRNA abolished psoriasis-like skin inflammation. Taken together, we identify IκBζ as a hitherto unknown key regulator of IL-17A-driven effects in psoriasis. Thus, targeting IκBζ could be a future strategy for treatment of psoriasis, and other inflammatory diseases for which IL-17 antagonists are currently tested in clinical trials.
- Published
- 2015
- Full Text
- View/download PDF
41. MALT1 Protease Activity Controls the Expression of Inflammatory Genes in Keratinocytes upon Zymosan Stimulation
- Author
-
Franziska C. Eberle, Paula Grondona, Anja Schmitt, Stephan Hailfinger, Birgit Schittek, Klaus Schulze-Osthoff, Amir S. Yazdi, Günter Jäger, Tabea Maier, Corinna Kosnopfel, Caroline Schönfeld, and Marc Brändle
- Subjects
0301 basic medicine ,Keratinocytes ,beta-Defensins ,medicine.medical_treatment ,Biochemistry ,chemistry.chemical_compound ,0302 clinical medicine ,Anti-Infective Agents ,Protein Isoforms ,Caspase ,Protein Kinase C ,Oligonucleotide Array Sequence Analysis ,biology ,Interleukin-17 ,Paracaspase ,BCL10 ,Cell biology ,Neoplasm Proteins ,src-Family Kinases ,030220 oncology & carcinogenesis ,Caspases ,Ionomycin ,Staphylococcus aureus ,Dermatology ,03 medical and health sciences ,medicine ,Gene silencing ,Humans ,Gene Silencing ,Molecular Biology ,Adaptor Proteins, Signal Transducing ,Inflammation ,Protease ,Tumor Necrosis Factor-alpha ,Gene Expression Profiling ,Zymosan ,Interleukin-8 ,Membrane Proteins ,NF-kappa B p50 Subunit ,Cell Biology ,B-Cell CLL-Lymphoma 10 Protein ,CARD Signaling Adaptor Proteins ,MALT1 ,030104 developmental biology ,chemistry ,Guanylate Cyclase ,Mucosa-Associated Lymphoid Tissue Lymphoma Translocation 1 Protein ,biology.protein ,Cancer research - Abstract
The protease activity of the paracaspase mucosa-associated lymphoid tissue lymphoma translocation gene 1 (MALT1) plays an important role in antigen receptor-mediated lymphocyte activation by controlling the activity of the transcription factor nuclear factor-κB and is thus essential for the expression of inflammatory target genes. MALT1 is not only present in cells of the hematopoietic lineage, but is ubiquitously expressed. Here we report that stimulation with zymosan or Staphylococcus aureus induced MALT1 protease activity in human primary keratinocytes. Inhibition of the Src family of kinases or novel protein kinase C isoforms as well as silencing of CARMA2 or BCL10 interfered with activation of MALT1 protease. Silencing or inhibition of MALT1 protease strongly decreased the expression of important inflammatory genes such as TNFα, IL-17C, CXCL8 and HBD-2. MALT1-inhibited cells were unable to mount an antimicrobial response upon zymosan stimulation or phorbolester/ionomycin treatment, demonstrating a central role of MALT1 protease activity in keratinocyte immunity and suggesting MALT1 as a potential target in inflammatory skin diseases.
- Published
- 2015
42. Differential gene expression in periportal and perivenous mouse hepatocytes
- Author
-
Stephan Hailfinger, Albrecht Buchmann, Christoph Köhle, Michael Bonin, Michael Schwarz, Carina Ittrich, and Albert Braeuning
- Subjects
Male ,Protein Denaturation ,Hepatic Veins ,Biology ,Models, Biological ,Biochemistry ,Gene Expression Regulation, Enzymologic ,Xenobiotics ,Mice ,Ammonia ,Gene expression ,Animals ,Lobules of liver ,Glycolysis ,Amino Acids ,Molecular Biology ,Gene ,Regulation of gene expression ,Mice, Inbred C3H ,Portal Vein ,Microarray analysis techniques ,Gene Expression Profiling ,Fatty Acids ,Gluconeogenesis ,Cell Biology ,Molecular biology ,Gene expression profiling ,Cholesterol ,Gene Expression Regulation ,Liver ,Hepatocytes - Abstract
Hepatocytes located in the periportal and perivenous zones of the liver lobule show remarkable differences in the levels and activities of various enzymes and other proteins. To analyze global gene expression patterns of periportal and perivenous hepatocytes, enriched populations of the two cell types were isolated by combined collagenase/digitonin perfusion from mouse liver and used for microarray analysis. In total, 198 genes and expressed sequences were identified that demonstrated a >/= 2-fold difference in expression between hepatocytes from the two different zones of the liver. A subset of 20 genes was additionally analyzed by real-time RT-PCR, validating the results obtained by the microarray analysis. Several of the differentially expressed genes encoded key enzymes of intermediary metabolism, including those involved in glycolysis and gluconeogenesis, fatty acid degradation, cholesterol and bile acid metabolism, amino acid degradation and ammonia utilization. In addition, several enzymes of phase I and phase II of xenobiotic metabolism were differentially expressed in periportal and perivenous hepatocytes. Our results confirm previous findings on metabolic zonation in liver, and extend our knowledge of the regulatory mechanisms at the transcriptional level.
- Published
- 2006
- Full Text
- View/download PDF
43. Zonal gene expression in murine liver: Lessons from tumors
- Author
-
Michael Schwarz, Stephan Hailfinger, Albrecht Buchmann, Maike Jaworski, and Albert Braeuning
- Subjects
Pathology ,medicine.medical_specialty ,Cell signaling ,Ratón ,Gene Expression ,Biology ,Mice ,Glutamate-Ammonia Ligase ,Gene expression ,medicine ,Animals ,Lobules of liver ,Gene ,beta Catenin ,chemistry.chemical_classification ,Hepatology ,Liver Neoplasms ,Cadherins ,Cell biology ,Disease Models, Animal ,Genes, ras ,Enzyme ,Liver ,chemistry ,Models, Animal ,Hepatocytes ,Murine liver ,Signal transduction ,Signal Transduction - Abstract
Gene expression in hepatocytes within the liver lobule is differentially regulated along the portal to central axis; however, the mechanisms governing the processes of zonation within the lobule are unknown. A model for zonal heterogeneity in normal liver is proposed, based on observations of differential expression of genes in liver tumors from mice that harbor activating mutations in either Catnb (which codes for beta-catenin) or Ha-ras. According to the model, the regulatory control consists of two opposing signals, one delivered by endothelial cells of the central veins activating a beta-catenin-dependent pathway (retrograde signal), the other by blood-borne molecules activating Ras-dependent downstream cascades (anterograde signal). In conclusion, gradients of opposing signaling molecules along the portocentral axis determine the pattern of enzymes and other proteins expressed in hepatocytes of the periportal and pericentral domains of the liver lobule.
- Published
- 2006
- Full Text
- View/download PDF
44. Human p53 knock-in ( hupki ) mice do not differ in liver tumor response from their counterparts with murine p53
- Author
-
Albrecht Buchmann, Maike Jaworski, Monica Hollstein, Manfred Hergenhahn, Michael Schwarz, Carina Ittrich, and Stephan Hailfinger
- Subjects
Cancer Research ,Liver tumor ,Genotype ,Tumor suppressor gene ,Mice, Transgenic ,Biology ,medicine.disease_cause ,Risk Assessment ,Mice ,Glutamate-Ammonia Ligase ,Gene knockin ,medicine ,Animals ,Humans ,Allele ,Gene ,Mutation ,Genetic Carrier Screening ,Liver Neoplasms ,General Medicine ,Genes, p53 ,medicine.disease ,Cancer research ,Liver cancer - Abstract
Mouse models are important tools in toxicologic research. Differences between species in pathways contributing to tumor development, however, raise the question in how far mouse models are valid for human risk assessment. One striking difference relates to the frequency of spontaneous liver cancer which is high in certain mouse strains but rather low in humans. Similarly, mutation frequencies in cancer genes are characteristically different, i.e. P53 mutations are frequent in human but very rare in murine liver tumors, whereas Ras genes are often mutated in mouse liver tumors but hardly ever in human liver cancers. Since P53 has been shown to control oncogenic RAS in human cells, we hypothesized that this function of the tumor suppressor could differ in mouse hepatocytes. To test this hypothesis, we used hupki (human p53 knock-in) mice which carry a partly humanized P53 sequence (P53KI). In this study, we report the results of the first hepatocarcinogenesis experiment with this strain of mice. Mice of the genotypes P53KI/KI, P53WT/KI and P53WT/WT were treated with N-nitrosodiethylamine at 2 weeks of age and killed 35 weeks later. The frequency of liver tumors and glucose-6-phosphatase-altered liver lesions was almost identical in all three P53 genotypes and approximately 40-50% of liver tumors showed activating mutations in codon 61 of the Ha-Ras gene independent of genotype. Moreover, only very few P53-positive lesions were observed but without nuclear localization of the protein, suggesting the absence of P53 mutations. These data suggest that the hupki allele behaves like its murine ortholog in mouse hepatocarcinogenesis.
- Published
- 2005
- Full Text
- View/download PDF
45. Detection and measurement of paracaspase MALT1 activity
- Author
-
Stephan, Hailfinger, Christiane, Pelzer, and Margot, Thome
- Subjects
Jurkat Cells ,Mucosa-Associated Lymphoid Tissue Lymphoma Translocation 1 Protein ,Caspases ,NF-kappa B ,Ubiquitination ,Humans ,Lymphocytes ,Lymphocyte Activation ,Molecular Biology ,Neoplasm Proteins - Abstract
The paracaspase MALT1 is a Cys-dependent, Arg-specific protease that plays an essential role in the activation and proliferation of lymphocytes during the immune response. Oncogenic activation of MALT1 is associated with the development of specific forms of B-cell lymphomas. Through specific cleavage of its substrates, MALT1 controls various aspects of lymphocyte activation, including the activation of transcriptional pathways, the stabilization of mRNAs, and an increase in cellular adhesion. In lymphocytes, the activity of MALT1 is tightly controlled by its inducible monoubiquitination, which promotes the dimerization of MALT1. Here, we describe both in vitro and in vivo assays that have been developed to assess MALT1 activity.
- Published
- 2014
46. Detection and Measurement of Paracaspase MALT1 Activity
- Author
-
Margot Thome, Christiane Pelzer, and Stephan Hailfinger
- Subjects
MALT1 ,Immune system ,Ubiquitin ,biology ,In vivo ,Chemistry ,biology.protein ,Monoubiquitination ,Paracaspase ,Cell adhesion ,Jurkat cells ,3. Good health ,Cell biology - Abstract
The paracaspase MALT1 is a Cys-dependent, Arg-specific protease that plays an essential role in the activation and proliferation of lymphocytes during the immune response. Oncogenic activation of MALT1 is associated with the development of specific forms of B-cell lymphomas. Through specific cleavage of its substrates, MALT1 controls various aspects of lymphocyte activation, including the activation of transcriptional pathways, the stabilization of mRNAs, and an increase in cellular adhesion. In lymphocytes, the activity of MALT1 is tightly controlled by its inducible monoubiquitination, which promotes the dimerization of MALT1. Here, we describe both in vitro and in vivo assays that have been developed to assess MALT1 activity.
- Published
- 2014
- Full Text
- View/download PDF
47. Paracaspase
- Author
-
Margot Thome and Stephan Hailfinger
- Subjects
Chemistry ,Paracaspase ,Neuroscience - Published
- 2013
- Full Text
- View/download PDF
48. Contributors
- Author
-
Catherine Anne Abbott, Carmela R. Abraham, Hideki Adachi, Osao Adachi, Zach Adam, Michael W.W. Adams, Michael J. Adang, Ibrahim M. Adham, Patrizia Aducci, David A. Agard, Alexey A. Agranovsky, Tetsuya Akamatsu, Yoshinori Akiyama, Reidar Albrechtsen, Alí Alejo, Sean M. Amberg, Alexander Y. Amerik, Piti Amparyup, Felipe Andrade, Germán Andrés, Daniel M. Andrews, Robert K. Andrews, Toni M. Antalis, Colin S. Anthony, Naoya Aoki, Suneel S. Apte, Kazunari Arima, Gérard Arlaud, Raghuvir Krishnaswamy Arni, Pascal Arnoux, Nathan N. Aronson, Michel Arthur, Yasuhisa Asano, Paolo Ascenzi, Marina T. Assakura, David S. Auld, Veridiana de Melo Rodrigues Ávila, Francesc X. Avilés, William M. Awad, Anand K. Bachhawat, Shan Bai, Teaster T. Baird, S. Paul Bajaj, Susan C. Baker, Agnieszka Banbula, Alan J. Barrett, Jemima Barrowman, John D. Bartlett, Jörg W. Bartsch, Nikola Baschuk, Isolda P. Baskova, Jyotsna Batra, Karl Bauer, Ulrich Baumann, Wolfgang Baumeister, Cédric Bauvois, Alex Bayés, Anne Beauvais, Christoph Becker-Pauly, Tadhg P. Begley, Miklós Békés, Robert Belas, Daniah Beleford, Teruhiko Beppu, Ernst M. Bergmann, Bruno A. Bernard, Dominique Bernard, Michael C. Berndt, Giovanna Berruti, Colin Berry, Greg P. Bertenshaw, Christian Betzel, Chetana Bhaskarla, Manoj Bhosale, Gabriele Bierbaum, B. Bjarnason Jón, Michael Blaber, Michael J. Blackman, Alexander Blinkovsky, Jef D. Boeke, Matthew Bogyo, Stefan Bohn, Guy Boileau, Mike Boland, Tové C. Bolken, Judith S. Bond, Jan Bondeson, Javier Bordallo, Claudia Borelli, Tiago O. Botelho, Richard R. Bott, David G. Bourne, Niels Bovenschen, Ralph A. Bradshaw, Klaus Breddam, Keith Brew, Paul J. Brindley, Diane L. Brinkman, Collette Britton, Jeff R. Broadbent, Anne Broadhurst, Dieter Brómme, Murray Broom, Jeremy S. Brown, Mark A. Brown, Iris Bruchhaus, Barbara A. Burleigh, Kristin E. Burns, James F. Burrows, Michael J. Butler, David J. Buttle, Chelsea M. Byrd, Tony Byun, Sandrine Cadel, Conor R. Caffrey, Santiago Cal, Javier Caldentey, Thomas Candela, Clemente Capasso, Daniel R. Capriogilio, Vincenzo Carginale, Adriana Karaoglanovic Carmona, Vern B. Carruthers, Francis J. Castellino, Joseph J. Catanese, Bruce Caterson, George H. Caughey, Naimh X. Cawley, Tim E. Cawston, Juan José Cazzulo, Jijie Chai, Karl X. Chai, Olga Meiri Chaim, L.S. Chang, Julie Chao, Marie-Pierre Chapot-Chartier, Jean-Louis Charli, Paulette Charlier, Karen J. Chave, Jian-Min Chen, Jinq-May Chen, Li-Mei Chen, Ya-Wen Chen, Yu-Yen Chen, Bernard Chevrier, Jean-François Chich, Jeremy Chien, Suneeta Chimalapati, Ki Joon Cho, Kwan Yong Choi, Woei-Jer Chuang, Chin Ha Chung, Ivy Yeuk Wah Chung, Christine Clamagirand, Ian M. Clark, Adrian K. Clarke, Nicola E. Clarke, Steven Gerard Clarke, Philippe Clauziat, Judith A. Clements, Catherine Coffinier, Paul Cohen, Alain Colige, Anne Collignon, Sean D. Colloms, Andreas Conzelmann, Graham H. Coombs, Jakki C. Cooney, Jonathan B. Cooper, Max D. Cooper, Nikki A. Copeland, Graeme S. Cottrell, Joseph T. Coyle, Charles S. Craik, John W.M. Creemers, Daniela Cretu, Jenifer Croce, Keith J. Cross, Rosario Cueva, Sheng Cui, Luis Cunha, Simon Cutting, Christophe d’Enfert, Hugues D’Orchymont, Björn Dahlbäck, Shujia Dai, Ross E. Dalbey, John P. Dalton, Pam M. Dando, R.M. Daniel, Sergei M. Danilov, Donna E. Davies, Heloisa S. De Araujo, Teresa De los Santos, Viviana de Luca, Ingrid De Meester, Ana Karina de Oliveira, Eduardo Brandt de Oliveira, Pedro Lagerblad De Oliveira, Sarah de Vos, Jeroen Declercq, Wim Declercq, Ala-Eddine Deghmane, Niek Dekker, Sonia Del Prete, Marina Del Rosal, Bernard Delmas, Robert DeLotto, Ilya V. Demidyuk, Mark R. Denison, Jan M. Deussing, Lakshmi A. Devi, Eleftherios P. Diamandis, Isabel Diaz, Araceli Díaz-Perales, Bauke W. Dijkstra, Yan Ding, Jack E. Dixon, Johannes Dodt, Terje Dokland, Iztok Dolenc, Ningzheng Dong, Tran Cat Dong, Ying Dong, Mitesh Dongre, Mark Donovan, Timothy M. Dore, Loretta Dorstyn, Hong Dou, Zhicheng Dou, Annette M. Dougall, Marcin Drag, Edward G. Dudley, Ben M. Dunn, Bruno Dupuy, Maria Conceicāo Duque-Magalhāes, M. Asunción Durá, Yves Eeckhout, Vincent Eijsink, Arthur Z. Eisen, Azza Eissa, Sandra Eklund, Ziad M. Eletr, Vincent Ellis, Wolfgang Engel, Ervin G. Erdös, Teresa Escalante, David A. Estell, Michael Etscheid, Herbert J. Evans, Roger D. Everett, Alex C. Faesen, Falk Fahrenholz, Miriam Fanjul-Fernández, Christopher J. Farady, Georges Feller, Hong Feng, Kurt M. Fenster, Claude Férec, Silvia Ferrari, Barbara Fingleton, Jed F. Fisher, Paula M. Fives-Taylor, Loren G. Fong, F. Forneris, Brian M. Forster, Friedrich Forster, Simon J. Foster, Thierry Foulon, Stephen I. Foundling, Jay William Fox, Bruno Franzetti, Alejandra P. Frasch, Hudson H. Freeze, Jean-Marie Frère, Teryl K. Frey, Beate Fricke, Lloyd D. Fricker, Rafael Fridman, Christopher J. Froelich, Camilla Fröhlich, Hsueh-Liang Fu, Cynthia N. Fuhrmann, Satoshi Fujimura, Hiroshi Fujiwara, Jun Fukushima, Keiichi Fukuyama, Robert S. Fuller, Martin Fusek, Christine Gaboriaud, Christian Gache, Oleksandr Gakh, Peter Gal, Junjun Gao, Adolfo García-Sastre, Donald L. Gardiner, John A. Gatehouse, G.M. Gaucher, Francis Gauthier, Jean-Marie Ghuysen, Wade Gibson, Jennifer Gillies, Elzbieta Glaser, Fabian Glaser, Michael H. Glickman, Peter Goettig, Colette Goffin, Eiichi Gohda, Alfred L. Goldberg, Daniel E. Goldberg, Gregory I. Goldberg, Nathan E. Goldfarb, F. Xavier Gomis-Rüth, B. Gopal, Alexander E. Gorbalenya, Stuart G. Gordon, Mark D. Gorrell, Friedrich Götz, Theodoros Goulas, Cécile Gouzy-Darmon, K. Govind, Lászlo Gráf, Robert R. Granados, Melissa Ann Gräwert, Douglas A. Gray, Thomas P. Graycar, Jonathan A. Green, Luiza Helena Gremski, Michael Groll, Tania Yu Gromova, P. Gros, Marvin J. Grubman, Amy M. Grunden, Ágústa Gudmundsdóttir, Micheline Guinand, Djamel Gully, Alla Gustchina, José María Gutiérrez, Byung Hak Ha, Jesper Z. Haeggström, James H. Hageman, Johanna Haiko, Stephan Hailfinger, Hans Michael Haitchi, Ji Seon Han, Chantal Hanquez, Minoru Harada, Ikuko Hara-Nishimura, Marianne Harboe, Torleif Härd, David A. Harris, Ulrich Hassiepen, Shoji Hata, Akira Hattori, Rong-Qiao He, Albert J.R. Heck, Dirk F. Hendricks, Bernhard Henrich, Patrick Henriet, Andrés Hernández-Arana, Irma Herrera-Camacho, Gerhard Heussipp, Toshihiko Hibino, P.M. Hicks, Bradley I. Hillman, B. Yukihiro Hiraoka, Jun Hiratake, Yohei Hizukuri, Heng-Chien Ho, Ngo Thi Hoa, Mark Hochstrasser, Kathryn M. Hodge, Theo Hofmann, Thomas Hohn, John R. Hoidal, Joachim-Volker Höltje, Koichi J. Homma, John F. Honek, Vivian Y.H. Hook, John D. Hooper, Nigel M. Hooper, Kazuo Hosoi, Christopher J. Howe, Dennis E. Hruby, James J.-D. Hseih, Chun-Chieh Hsu, Tony T. Huang, Tur-Fu Huang, Yoann Huet, Clare Hughes, Jean-Emmanuel Hugonnet, Adrienne L. Huston, Oumaïma Ibrahim-Granet, Eiji Ichishima, Yukio Ikehara, Tadashi Inagami, Jessica Ingram, R.E. Isaac, Grazia Isaya, Clara E. Isaza, Shin-ichi Ishii, Amandine Isnard, Kiyoshi Ito, Koreaki Ito, Yoshifumi Itoh, Xavier Iturrioz, Sadaaki Iwanaga, Ralph W. Jack, Mel C. Jackson, Michael N.G. James, Jiří Janata, Claire Janoir, Hanna Janska, Ken F. Jarrell, Mariusz Jaskolski, Sheila S. Jaswal, Ying Y. Jean, Dieter E. Jenne, Young Joo Jeon, Ping Jiang, John E. Johnson, Michael D. Johnson, James A. Johnston, Amanda Jones, Elizabeth W. Jones, Carine Joudiou, Luiz Juliano, Hea-Jin Jung, Ray Jupp, Todd F. Kagawa, Hubert Kalbacher, Yayoi Kamata, Shuichi Kaminogawa, Yoshiyuki Kamio, Makoto Kaneda, Sung Gyun Kang, Sung Hwan Kang, Mary Kania, Tomasz Kantyka, Nobuyuki Kanzawa, Abdulkarim Y. Karim, Takafumi Kasumi, Hiroaki Kataoka, Hardeep Kaur, Shun-Ichiro Kawabata, Mari Kawaguchi, John Kay, Murat Kaynar, Kenneth C. Keiler, R.M. Kelly, Nathaniel T. Kenton, Michael A. Kerr, Kristof Kersse, Jukka Kervinen, Benedikt M. Kessler, Efrat Kessler, Timo K. Khoronen, Simon Kidd, Marjolein Kikkert, Mogens Kilian, Do-Hyung Kim, Doyoun Kim, Eunice EunKyeong Kim, In Seop Kim, Jung-Gun Kim, Kyeong Kyu Kim, Kyung Hyun Kim, Matthew S. Kimber, Yukio Kimura, Heidrun Kirschke, Yoshiaki Kiso, Colin Kleanthous, Jürgen R. Klein, Michael Klemba, Beata Kmiec, Hideyuki Kobayashi, Hiroyuki Kodama, Gerald Koelsch, Jan Kok, P.E. Kolattukody, Fabrice A. Kolb, Harald Kolmar, Yumiko Komori, Jan Konvalinka, Brice Korkmaz, Sergey V. Kostrov, Hans-Georg Kräusslich, Gabi Krczal, Lawrence F. Kress, Magnüs Már Kristjánsson, Tomáš Kučera, Sayali S. Kukday, Hidehiko Kumagai, Sharad Kumar, Malika Kumarasiri, Takashi Kumazaki, Beate M. Kümmerer, Kouji Kuno, Markku Kurkinen, Eva Kutejová, Marie Kveiborg, Agnieszka Kwarciak, Liisa Laakkonen, Nikolaos E. Labrou, Gavin D. Laing, Gayle Lamppa, Thomas Langer, Richard A. Laursen, Richard A. Lawrenson, Matthew D. Layne, Bernard F. Le Bonniec, María C. Leal, Ronald M. Lechan, David H. Lee, Irene Lee, Jae Lee, Kye Joon Lee, Soohee Lee, Xiaobo Lei, Jonathan Leis, Ellen K. LeMosy, Thierry Lepage, Stephen H. Leppla, Adam Lesner, Ivan A.D. Lessard, Guy Lhomond, Huilin Li, Shu-Ming Li, Weiguo Li, Ta-Hsiu Liao, Robert C. Liddington, Toby Lieber, H.R. Lijnen, Christopher D. Lima, Chen-Yong Lin, Gang Lin, Ming T. Lin, Xinli Lin, Yee-Shin Lin, L.L. Lindsay, William N. Lipscomb, John W. Little, Ching-Chuan Liu, Chuan-ju Liu, Mark O. Lively, Nurit Livnat-Levanon, Per O. Ljungdahl, Catherine Llorens-Cortes, Peter Lobel, Y. Peng Loh, Jouko Lohi, G.P. Lomonossoff, Yvan Looze, Carlos López-Otin, Landys Lopez-Quezada, Alex Loukas, Long-Sheng Lu, Áke Lundwall, Liu-Ying Luo, Andrei Lupas, Dawn S. Luthe, Nicholas J. Lynch, Peter J. Lyons, Vivian L. MacKay, Jesica M. Levingston Macleod, Viktor Magdolen, Jean-Luc Mainardi, Kauko K. Mäkinen, Jeremy P. Mallari, Surya P. Manandhar, Fajga R. Mandelbaum, Anne M. Manicone, Johanna Mansfeld, Joseph Marcotrigiano, Michael Mares, Gemma Marfany, Francis S. Markland, Judith Marokházi, Hélène Marquis, Robert A. Marr, Enzo Martegani, Erik W. Martin, Manuel Martinez, L. Miguel Martins, Masato Maruyama, Masugi Maruyama, Sususmu Maruyama, Takeharu Masaki, Ramin Massoumi, Rency T. Mathew, Lynn M. Matrisian, Yoshihiro Matsuda, Osamu Matsushita, Marco Matuschek, Anna Matušková, Krisztina Matúz, Cornelia Mauch, Michael R. Maurizi, Lorenz M. Mayr, Dewey G. McCafferty, J. Ken McDonald, James H. McKerrow, David McMillan, Robert P. Mecham, Darshini P. Mehta, Chris Meisinger, Alan Mellors, Roger G. Melton, Jeffrey A. Melvin, Robert Ménard, Luis Menéndez-Arias, Milene C. Menezes, Andrew Mesecar, Stéphane Mesnage, Diane H. Meyer, Gregor Meyers, Susan Michaelis, Karolina Michalska, Wojciech P. Mielicki, Igor Mierau, Galina V. Mikoulinskaia, Charles G. Miller, Lydia K. Miller, John Mills, Kenneth V. Mills, Jinrong Min, Michel-Yves Mistou, Yoshio Misumi, Shin-ichi Miyoshi, Shigehiko Mizutani, Shahriar Mobashery, Satsuki Mochizuki, William L. Mock, Frank Möhrlen, Nathalie Moiré, Paul E. Monahan, Angela Moncada-Pazos, Véronique Monnet, Michel Monod, Cesare Montecucco, Laura Morelli, Sumiko Mori, Takashi Morita, James H. Morrissey, Richard J. Morse, John S. Mort, Uffe H. Mortensen, Rory E. Morty, Joel Moss, Hidemasa Motoshima, Jeremy C. Mottram, Ana M. Moura-da-Silva, Mary Beth Mudgett, Egbert Mundt, Kazuo Murakami, Mario Tyago Murakami, Kimiko MurakamiMurofoshi, Sawao Murao, Gillian Murphy, M.R.N. Murthy, Tatsushi Muta, Elmarie Myburgh, Nino Mzhavia, A.H.M. Nurun Nabi, Hideaki Nagase, Michael W. Nagle, Dorit K. Nägler, Rajesh R. Naik, Divya B. Nair, Toshiki Nakai, Yoshitaka Nakajima, Yukio Nakamura, Hitoshi Nakatogawa, Toru Nakayama, Natalia N. Nalivaeva, Dipankar Nandi, Maria Clara Leal Nascimento-Silva, Kim Nasmyth, Carl F. Nathan, Fernando Navarro-García, Dayane Lorena Naves, Danny D. Nedialkova, Keir C. Neuman, Jeffrey-Tri Nguyen, Ky-Anh Nguyen, Gabriela T. Niemirowicz, Toshiaki Nikai, Eiichiro Nishi, Wataru Nishii, Makoto Nishiyama, Yasuhiro Nishiyama, Masatoshi Noda, Seiji Nomura, Shigemi Norioka, Desire M.M. Nsangou, Amornrat O’Brien, Michael B. O’Connor, Kohei Oda, Irina V. Odinokova, Joyce Oetjen, Teru Ogura, Dennis E Ohman, Yoshinori Ohsumi, Mukti Ojha, Akinobu Okabe, Yasunori Okada, Keinosuke Okamoto, Kenji Okuda, Nobuaki Okumura, Takashi Okuno, Kjeld Oleson, Priscila Oliveira de Giuseppe, Martin Olivier, Yasuko Ono, Stephen Oroszlan, Nobuyuki Ota, Michael Ovadia, Jiyang O-Wang, Claus Oxvig, Jeremy C.L. Packer, Sergio Padilla-López, Mark Paetzel, Michael J. Page, Andrea Page-McCaw, Mark J.I. Paine, Byoung Chul Park, Eunyong Park, John E. Park, Pyong Woo Park, Sung Goo Park, Kirk L. Parkin, William C Parks, Thaysa Paschoalin, Annalisa Pastore, Alexander Nikolich Patananan, Sudhir Paul, Henry L. Paulson, Ulrich von Pawel-Rammingen, David A. Pearce, Mark S. Pearson, Duanqing Pei, Gunnar Pejler, Alan D. Pemberton, Jianhao Peng, Julien Pernier, Jan-Michael Peters, Thorsten Pfirrmann, Viet-Laï Pham, Iva Pichová, Darren Pickering, Christophe Piesse, David Pignol, Robert N. Pike, Lothaire Pinck, Hubert Pirkle, Henry C. Pitot, Andrew G. Plaut, Hidde Ploegh, László Polgár, Corrine Porter, Rolf Postina, Jan Potempa, Knud Poulsen, Scott D. Power, Rex. F. Pratt, Gerd Prehna, Gilles Prévost, Alexey V. Pshezhetsky, Mohammad A. Qasim, Feng Qian, Jiazhou Qiu, Víctor Quesada, Evette S. Radisky, Stephen D. Rader, Kavita Raman, Andrew J. Ramsay, Derrick E. Rancourt, Najju Ranjit, Narayanam V. Rao, Kiira Ratia, Neil D. Rawlings, Robert B. Rawson, Vijay Reddy, Colvin M. Redman, Maria Elena Regonesi, Andreas S. Reichert, Antonia P. Reichl, Han Remaut, S. James Remington, Martin Renatus, David Reverter, Eric C. Reynolds, Mohamed Rholam, Charles M. Rice, Todd W. Ridky, Howard Riezman, D.C. Rijken, Marie-Christine Rio, Alison Ritchie, Janine Robert-Baudouy, Mark W. Robinson, Michael Robinson, Adela Rodriguez-Romero, Renata Santos Rodriques, John C. Rogers, Camilo Rojas, Floyd E. Romesberg, David J. Roper, Nora Rosas-Murrieta, A.M. Rose, Philip J. Rosenthal, J. Rosing, Ornella Rossetto, Véronique Rossi, Richard A. Roth, Hanspeter Rottensteiner, Andrew D. Rowan, Mikhail Rozanov, Alexandra Rucavado, Andrea Ruecker, Françoise Rul, Till Rümenapf, Ilaria Russo, Martin D. Ryan, Elena Sacco, J. Evan Sadler, W. Saenger, Hans-Georg Sahl, Mohammed Sajid, Masayoshi Sakaguchi, Fumio Sakiyama, Maria L. Salas, Maria Cristina O. Salgado, Guy S. Salvesen, Edith Sánchez, Eladio F. Sanchez, Qing-Xiang Amy Sang, Krishnan Sankaran, Susanta K. Sarkar, Michael P. Sarras, Yoshikiyo Sasagawa, Araki Satohiko, Eric Sauvage, Loredana Saveanu, H.S. Savithri, Hitoshi Sawada, R. Gary Sawers, Isobel A. Scarisbrick, Andreas Schaller, Justin M. Scheer, Friedrich Scheiflinger, Cordelia Schiene-Fischer, Uwe Schlomann, Manfred Schlösser, Alvin H. Schmaier, Walter K. Schmidt, Anette Schneemann, Rick G. Schnellmann, Henning Scholze, Lutz Schomburg, Wilhelm J. Schwaeble, Christopher J. Scott, Rosaria Scudiero, Atsuko Sehara-Fujisawa, Nabil G. Seidah, Motoharu Seiki, Junichi Sekiguchi, Andrea Senff-Ribeiro, Ihn Sik Seong, Mihaela Serpe, Solange M.T. Serrano, Peter Setlow, Tina Shahian, M. Shanks, Feng Shao, Steven D. Shapiro, Navneet Sharma, Lindsey N. Shaw, Aimee Shen, Lei Shen, Roger F. Sherwood, Yun-Bo Shi, Hitoshi Shimoi, Yoichiro Shimura, A.D. Shirras, Viji Shridhar, Jinal K. Shukla, Ene Siigur, Jüri Siigur, Natalie C. Silmon de Monerri, Robert B. Sim, James P. Simmer, William H. Simmons, Jaspreet Singh, Alison Singleton, Tatiana D. Sirakova, Titia K. Sixma, Tim Skern, Randal A. Skidgel, Jeffrey Slack, David E. Sleat, Barbara S. Slusher, Janet L. Smith, Matthew A. Smith, Mark J. Smyth, Erik J. Snijder, Solmaz Sobhanifar, Kenneth Söderhaäll, Istvan Sohar, Peter Sonderegger, Marcos Henrique Ferreira Sorgine, Hiroyuki Sorimachi, Karen E. Soukhodolets, Tatiana de Arruda Campos Brasil de Souza, Tamás Sperka, Shiranee Sriskandan, Joseph W. St. Geme, Raymond J. St. Leger, Peter Staib, James L. Steele, Bjarki Stefansson, Christian Steinkühler, Leisa M. Stenberg, Johan Stenflo, Henning R. Stennicke, Valentin M. Stepanov, Olga A. Stepnaya, Frank Steven, Richard L. Stevens, Kenneth J. Stevenson, Mathieu St-Louis, Christopher C. Stobart, Walter Stöcker, Andrew C. Storer, Norbert Sträter, Ellen G. Strauss, James H. Strauss, Kvido Stříšovský, Natalie C.J. Strynadka, Edward D. Sturrock, Dan Su, Xiao-Dong Su, Paz Suárez-Rendueles, Traian Sulea, Venkatesh Sundararajan, Ryoji Suno, Carolyn K. Suzuki, Fumiaki Suzuki, Hideyuki Suzuki, Nobuhiro Suzuki, Stephen Swenson, Rose L. Szabady, Pal Bela Szecsi, Lászlo Szilágyi, Muhamed-Kheir Taha, Eizo Takahashi, Kenji Takahashi, Toshiro Takai, Atsushi Takeda, Soichi Takeda, Jeremy J.R.H. Tame, Tomohiro Tamura, Fulong Tan, Keiji Tanaka, Carmen Tanase, Jordan Tang, Martha M. Tanizaki, Egbert Tannich, Guido Tans, Anthony L. Tarentino, Anchalee Tassanakajon, Hiroki Tatsumi, Norbert Tautz, Erin Bassford Taylor, Pedro Filipe Teixeira, Bhanu Prakash V.L. Telugu, Markus F. Templin, Shigeyuki Terada, Uchikoba Tetsuya, C. Thacker, Maulik Thaker, Heinz-Jürgen Thiel, Nicole Thielens, Gonzales Thierry, Karine Thivierge, Mark D. Thomas, Margot Thome, Mary K. Thorsness, Peter E. Thorsness, Natalie J. Tigue, Sokol V. Todi, Birgitta Tomkinson, Fiorella Tonello, Liang Tong, H.S. Toogood, Paolo Tortora, József Tözsèr, Luiz Rodolpho Travassos, James Travis, Dilza Trevisan-Silva, Francesca Trinchella, Neil N. Trivedi, Carol M. Troy, Harald Tschesche, Yu-Lun Tseng, Masafumi Tsujimoto, Anthony T. Tu, Kathleen E. Tumelty, Boris Turk, Dusan Turk, Vito Turk, Anthony J. Turner, Tetsuya Uchikoba, Takayuki Ueno, Alejandro P. Ugalde, Veli-Jukka Uitto, Sinisa Urban, Olivier Valdenaire, Adrian Valli, Jozef Van Beeumen, Bertus Van den Burg, Renier A.L. Van der Hoorn, Jan Maarten van Dijl, Peter Van Endert, Bram J. Van Raam, Harold E. Van Wart, Tom Vanden Berghe, Peter Vandenabeele, Margo Vanoni, Silvio Sanches Veiga, William H. Velander, Gloria Velasco, Josep Vendrell, I. István Venekei, Vaclav Vetvicka, F.-Nora Vögtle, Waldemar Vollmer, Kei Wada, Fred W. Wagner, Sun Nyunt Wai, Timothy Wai, Shane Wainwright, Kenneth W. Walker, Stephen J. Walker, Jean Wallach, Linda L. Walling, Peter N. Walsh, Hai-Yan Wang, Hengbin Wang, Jianwei Wang, Peng Wang, Ping Wang, Michael Wassenegger, Kunihiko Watanabe, Helen Webb, Joseph M. Weber, Niklas Weber, Daniel R. Webster, Shuo Wei, Rodney A. Welch, James A. Wells, Herbert Wenzel, Ingrid E. Wertz, Ulla W. Wewer, Alison R. Whyteside, Sherwin Wilk, Jean-Marc Wilkin, Claudia Wilmes, Jakob R. Winther, David S. Wishart, Alexander Wlodawer, J. Fred Woessner, Michael S. Wolfe, Wilson Wong, Roger Woodgate, Gerry Wright, Jiunn-Jong Wu, Qingyu Wu, Magdalena Wysocka, Chao Xu, Zhenghong Xu, Kinnosuke Yahori, Shoji Yamada, Nozomi Yamaguchi, Shinji Yamaguchi, Yoshio Yamakawa, Hiroki Yamamoto, Ikao Yana, Maozhou Yang, Na Yang, Chenjuan Yao, Tingting Yao, Noriko Yasuda, Toshimasa Yasuhara, Shigeki Yasumasu, Edward T.H. Yeh, Irene Yiallouros, Jiang Yin, Hiroo Yonezawa, Soon Ji Yoo, Tadashi Yoshimoto, Michael W. Young, Stephen G. Young, Nousheen Zaidi, Ludmila L. Zavalova, Peter Zavodszky, Aidong Zhang, Xianming Zhang, Yi-Zheng Zhang, Dominick Zheng, Guangming Zhong, Rong Zhong, Yuan Zhou, Zhaohui Sunny Zhou, Michael Zick, Paola Zigrino, and Andrei A. Zimin
- Published
- 2013
- Full Text
- View/download PDF
49. Cathepsin X cleavage of the beta2 integrin regulates talin-binding and LFA-1 affinity in T cells
- Author
-
Samo Turk, Zala Jevnikar, Urban Švajger, Bojan Doljak, Janko Kos, Stanislav Gobec, Stephan Hailfinger, Nataša Obermajer, and Margot Thome
- Subjects
Models, Molecular ,Talin ,T-Lymphocytes ,Immunology ,Integrin ,Blotting, Western ,Cell Separation ,Cleavage (embryo) ,Transfection ,Polymerase Chain Reaction ,Jurkat Cells ,Immunology and Allergy ,Humans ,Immunoprecipitation ,chemistry.chemical_classification ,biology ,Signal transducing adaptor protein ,Cell Biology ,Flow Cytometry ,Carboxypeptidase ,Talin binding ,Cathepsins ,Lymphocyte Function-Associated Antigen-1 ,Cell biology ,Amino acid ,Chemotaxis, Leukocyte ,chemistry ,Microscopy, Fluorescence ,Cytoplasm ,CD18 Antigens ,biology.protein ,Electrophoresis, Polyacrylamide Gel ,Protein Processing, Post-Translational ,Cysteine ,Protein Binding - Abstract
T cell migration, essential for immune surveillance and response, is mediated by the integrin LFA-1. CatX, a cysteine carboxypeptidase, is involved in the regulation of T cell migration by interaction with LFA-1. We show that sequential cleavage of C-terminal amino acids from the β2 cytoplasmic tail of LFA-1, by CatX, enhances binding of the adaptor protein talin to LFA-1 and triggers formation of the latter's high-affinity form. As shown by SPR analysis of peptides constituting the truncated β2 tail, the cleavage of three C-terminal amino acids by CatX resulted in a 1.6-fold increase of talin binding. Removal of one more amino acid resulted in a 2.5-fold increase over the intact tail. CatX cleavage increased talin-binding affinity to the MD but not the MP talin-binding site on the β2 tail. This was shown by molecular modeling of the β2 tail/talin F3 complex to be a result of conformational changes affecting primarily the distal-binding site. Analysis of LFA-1 by conformation-specific mAb showed that CatX modulates LFA-1 affinity, promoting formation of high-affinity from intermediate-affinity LFA-1 but not the initial activation of LFA-1 from a bent to extended form. CatX post-translational modifications may thus represent a mechanism of LFA-1 fine-tuning that enables the trafficking of T cells.
- Published
- 2011
50. Essential role of MALT1 protease activity in activated B cell-like diffuse large B-cell lymphoma
- Author
-
Georg Lenz, Vu L. Ngo, Fabien Rebeaud, Judith Dierlamm, Anita Posvitz-Fejfar, Eva-Maria Murga Penas, Wing C. Chan, Montserrat Guzzardi, Margot Thome, Stephan Hailfinger, and Louis M. Staudt
- Subjects
Lung Neoplasms ,Antineoplastic Agents ,medicine.disease_cause ,Proto-Oncogene Mas ,Mice ,immune system diseases ,hemic and lymphatic diseases ,Cell Line, Tumor ,medicine ,Animals ,Humans ,Caspase ,B cell ,Adaptor Proteins, Signal Transducing ,Cell Proliferation ,Oligonucleotide Array Sequence Analysis ,Multidisciplinary ,biology ,Cell growth ,Gene Expression Profiling ,NF-kappa B ,Biological Sciences ,medicine.disease ,B-Cell CLL-Lymphoma 10 Protein ,Molecular biology ,Caspase Inhibitors ,Lymphoma ,Neoplasm Proteins ,CARD Signaling Adaptor Proteins ,Gene Expression Regulation, Neoplastic ,MALT1 ,medicine.anatomical_structure ,Guanylate Cyclase ,Mucosa-Associated Lymphoid Tissue Lymphoma Translocation 1 Protein ,Caspases ,biology.protein ,Cancer research ,Lymphoma, Large B-Cell, Diffuse ,Signal transduction ,Carcinogenesis ,Diffuse large B-cell lymphoma ,Signal Transduction - Abstract
A key element for the development of suitable anti-cancer drugs is the identification of cancer-specific enzymatic activities that can be therapeutically targeted. Mucosa-associated lymphoid tissue transformation protein 1 (MALT1) is a proto-oncogene that contributes to tumorigenesis in diffuse large B-cell lymphoma (DLBCL) of the activated B-cell (ABC) subtype, the least curable subtype of DLBCL. Recent data suggest that MALT1 has proteolytic activity, but it is unknown whether this activity is relevant for tumor growth. Here we report that MALT1 is constitutively active in DLBCL lines of the ABC but not the GCB subtype. Inhibition of the MALT1 proteolytic activity led to reduced expression of growth factors and apoptosis inhibitors, and specifically affected the growth and survival of ABC DLBCL lines. These results demonstrate a key role for the proteolytic activity of MALT1 in DLBCL of the ABC subtype, and provide a rationale for the development of pharmacological inhibitors of MALT1 in DLBCL therapy.
- Published
- 2009
Catalog
Discovery Service for Jio Institute Digital Library
For full access to our library's resources, please sign in.