149 results on '"Johannes Beckers"'
Search Results
2. Author Correction: Maternal whole blood cell miRNA-340 is elevated in gestational diabetes and inversely regulated by glucose and insulin
- Author
-
Laura Stirm, Peter Huypens, Steffen Sass, Richa Batra, Louise Fritsche, Sara Brucker, Harald Abele, Anita M. Hennige, Fabian Theis, Johannes Beckers, Martin Hrabě de Angelis, Andreas Fritsche, Hans-Ulrich Häring, and Harald Staiger
- Subjects
Medicine ,Science - Published
- 2022
- Full Text
- View/download PDF
3. The Notch ligand DNER regulates macrophage IFNγ release in chronic obstructive pulmonary diseaseResearch in context
- Author
-
Carolina Ballester-López, Thomas M. Conlon, Zeynep Ertüz, Flavia R. Greiffo, Martin Irmler, Stijn E. Verleden, Johannes Beckers, Isis E. Fernandez, Oliver Eickelberg, and Ali Önder Yildirim
- Subjects
Medicine ,Medicine (General) ,R5-920 - Abstract
Background: Chronic Obstructive Pulmonary Disease (COPD) is the third leading cause of death worldwide with no curative therapy. A non-canonical Notch ligand, DNER, has been recently identified in GWAS to associate with COPD severity, but its function and contribution to COPD is unknown. Methods: DNER localisation was assessed in lung tissue from healthy and COPD patients, and cigarette smoke (CS) exposed mice. Microarray analysis was performed on WT and DNER deficient M1 and M2 bone marrow-derived macrophages (BMDM), and gene set enrichment undertaken. WT and DNER deficient mice were exposed to CS or filtered air for 3 day and 2 months to assess IFNγ-expressing macrophages and emphysema development. Notch and NFKB active subunits were quantified in WT and DNER deficient LPS-treated and untreated BMDM. Findings: Immunofluorescence staining revealed DNER localised to macrophages in lung tissue from COPD patients and mice. Human and murine macrophages showed enhanced DNER expression in response to inflammation. Interestingly, pro-inflammatory DNER deficient BMDMs exhibited impaired NICD1/NFKB dependent IFNγ signalling and reduced nuclear NICD1/NFKB translocation. Furthermore, decreased IFNγ production and Notch1 activation in recruited macrophages from CS exposed DNER deficient mice were observed, protecting against emphysema and lung dysfunction. Interpretation: DNER is a novel protein induced in COPD patients and 6 months CS-exposed mice that regulates IFNγ secretion via non-canonical Notch in pro-inflammatory recruited macrophages. These results provide a new pathway involved in COPD immunity that could contribute to the discovery of innovative therapeutic targets. Funding: This work was supported from the Helmholtz Alliance ‘Aging and Metabolic Programming, AMPro’. Keywords: COPD, Macrophages, DNER, Notch signalling, IFNγ, Cigarette smoke, Lung, NFkB
- Published
- 2019
- Full Text
- View/download PDF
4. Standardized, systemic phenotypic analysis reveals kidney dysfunction as main alteration of Kctd1 I27N mutant mice
- Author
-
Sudhir Kumar, Birgit Rathkolb, Sibylle Sabrautzki, Stefan Krebs, Elisabeth Kemter, Lore Becker, Johannes Beckers, Raffi Bekeredjian, Robert Brommage, Julia Calzada-Wack, Lillian Garrett, Sabine M. Hölter, Marion Horsch, Martin Klingenspor, Thomas Klopstock, Kristin Moreth, Frauke Neff, Jan Rozman, Helmut Fuchs, Valérie Gailus-Durner, Martin Hrabe de Angelis, Eckhard Wolf, and Bernhard Aigner
- Subjects
Animal model ,Kctd1 ,SEN syndrome ,Systematic phenotype analysis ,Medicine - Abstract
Abstract Background Increased levels of blood plasma urea were used as phenotypic parameter for establishing novel mouse models for kidney diseases on the genetic background of C3H inbred mice in the phenotype-driven Munich ENU mouse mutagenesis project. The phenotypically dominant mutant line HST014 was established and further analyzed. Methods Analysis of the causative mutation as well as the standardized, systemic phenotypic analysis of the mutant line was carried out. Results The causative mutation was detected in the potassium channel tetramerization domain containing 1 (Kctd1) gene which leads to the amino acid exchange Kctd1 I27N thereby affecting the functional BTB domain of the protein. This line is the first mouse model harboring a Kctd1 mutation. Kctd1 I27N homozygous mutant mice die perinatally. Standardized, systemic phenotypic analysis of Kctd1 I27N heterozygous mutants was carried out in the German Mouse Clinic (GMC). Systematic morphological investigation of the external physical appearance did not detect the specific alterations that are described in KCTD1 mutant human patients affected by the scalp-ear-nipple (SEN) syndrome. The main pathological phenotype of the Kctd1 I27N heterozygous mutant mice consists of kidney dysfunction and secondary effects thereof, without gross additional primary alterations in the other phenotypic parameters analyzed. Genome-wide transcriptome profiling analysis at the age of 4 months revealed about 100 differentially expressed genes (DEGs) in kidneys of Kctd1 I27N heterozygous mutants as compared to wild-type controls. Conclusions In summary, the main alteration of the Kctd1 I27N heterozygous mutants consists in kidney dysfunction. Additional analyses in 9–21 week-old heterozygous mutants revealed only few minor effects.
- Published
- 2017
- Full Text
- View/download PDF
5. Extensive phenotypic characterization of a new transgenic mouse reveals pleiotropic perturbations in physiology due to mesenchymal hGH minigene expression
- Author
-
Aimilios Kaklamanos, Jan Rozman, Manolis Roulis, Niki Karagianni, Maria Armaka, Moya Wu, Laura Brachthäuser, Julia Calzada-Wack, Marion Horsch, Johannes Beckers, Birgit Rathkolb, Thure Adler, Frauke Neff, Eckhard Wolf, Valerie Gailus-Durner, Helmut Fuchs, Martin Hrabe de Angelis, and George Kollias
- Subjects
Medicine ,Science - Abstract
Abstract The human growth hormone (hGH) minigene used for transgene stabilization in mice has been recently identified to be locally expressed in the tissues where transgenes are active and associated with phenotypic alterations. Here we extend these findings by analyzing the effect of the hGH minigene in TgC6hp55 transgenic mice which express the human TNFR1 under the control of the mesenchymal cell-specific CollagenVI promoter. These mice displayed a fully penetrant phenotype characterized by growth enhancement accompanied by perturbations in metabolic, skeletal, histological and other physiological parameters. Notably, this phenotype was independent of TNF-TNFR1 signaling since the genetic ablation of either Tnf or Tradd did not rescue the phenotype. Further analyses showed that the hGH minigene was expressed in several tissues, also leading to increased hGH protein levels in the serum. Pharmacological blockade of GH signaling prevented the development of the phenotype. Our results indicate that the unplanned expression of the hGH minigene in CollagenVI expressing mesenchymal cells can lead through local and/or systemic mechanisms to enhanced somatic growth followed by a plethora of primary and/or secondary effects such as hyperphagia, hypermetabolism, disturbed glucose homeostasis, altered hematological parameters, increased bone formation and lipid accumulation in metabolically critical tissues.
- Published
- 2017
- Full Text
- View/download PDF
6. Maternal Pre-Pregnancy Obesity Is Associated with Altered Placental Transcriptome.
- Author
-
Signe Altmäe, Maria Teresa Segura, Francisco J Esteban, Sabine Bartel, Pilar Brandi, Martin Irmler, Johannes Beckers, Hans Demmelmair, Carmen López-Sabater, Berthold Koletzko, Susanne Krauss-Etschmann, and Cristina Campoy
- Subjects
Medicine ,Science - Abstract
Maternal obesity has a major impact on pregnancy outcomes. There is growing evidence that maternal obesity has a negative influence on placental development and function, thereby adversely influencing offspring programming and health outcomes. However, the molecular mechanisms underlying these processes are poorly understood. We analysed ten term placenta's whole transcriptomes in obese (n = 5) and normal weight women (n = 5), using the Affymetrix microarray platform. Analyses of expression data were carried out using non-parametric methods. Hierarchical clustering and principal component analysis showed a clear distinction in placental transcriptome between obese and normal weight women. We identified 72 differentially regulated genes, with most being down-regulated in obesity (n = 61). Functional analyses of the targets using DAVID and IPA confirm the dysregulation of previously identified processes and pathways in the placenta from obese women, including inflammation and immune responses, lipid metabolism, cancer pathways, and angiogenesis. In addition, we detected new molecular aspects of obesity-derived effects on the placenta, involving the glucocorticoid receptor signalling pathway and dysregulation of several genes including CCL2, FSTL3, IGFBP1, MMP12, PRG2, PRL, QSOX1, SERPINE2 and TAC3. Our global gene expression profiling approach demonstrates that maternal obesity creates a unique in utero environment that impairs the placental transcriptome.
- Published
- 2017
- Full Text
- View/download PDF
7. Generation and Standardized, Systemic Phenotypic Analysis of Pou3f3L423P Mutant Mice.
- Author
-
Sudhir Kumar, Birgit Rathkolb, Elisabeth Kemter, Sibylle Sabrautzki, Dian Michel, Thure Adler, Lore Becker, Johannes Beckers, Dirk H Busch, Lillian Garrett, Wolfgang Hans, Sabine M Hölter, Marion Horsch, Martin Klingenspor, Thomas Klopstock, Ildikó Rácz, Jan Rozman, Ingrid Liliana Vargas Panesso, Alexandra Vernaleken, Andreas Zimmer, Helmut Fuchs, Valérie Gailus-Durner, Martin Hrabě de Angelis, Eckhard Wolf, and Bernhard Aigner
- Subjects
Medicine ,Science - Abstract
Increased levels of blood plasma urea were used as phenotypic parameter for establishing novel mouse models for kidney diseases on the genetic background of C3H inbred mice in the phenotype-driven Munich ENU mouse mutagenesis project. The phenotypically recessive mutant line HST011 was established and further analyzed. The causative mutation was detected in the POU domain, class 3 transcription factor 3 (Pou3f3) gene, which leads to the amino acid exchange Pou3f3L423P thereby affecting the conserved homeobox domain of the protein. Pou3f3 homozygous knockout mice are published and show perinatal death. Line Pou3f3L423P is a viable mouse model harboring a homozygous Pou3f3 mutation. Standardized, systemic phenotypic analysis of homozygous mutants was carried out in the German Mouse Clinic. Main phenotypic changes were low body weight and a state of low energy stores, kidney dysfunction and secondary effects thereof including low bone mineralization, multiple behavioral and neurological defects including locomotor, vestibular, auditory and nociceptive impairments, as well as multiple subtle changes in immunological parameters. Genome-wide transcriptome profiling analysis of kidney and brain of Pou3f3L423P homozygous mutants identified significantly regulated genes as compared to wild-type controls.
- Published
- 2016
- Full Text
- View/download PDF
8. Sphingomyelin Synthase 1 Is Essential for Male Fertility in Mice.
- Author
-
Anke Wittmann, Marcus O W Grimm, Harry Scherthan, Marion Horsch, Johannes Beckers, Helmut Fuchs, Valerie Gailus-Durner, Martin Hrabě de Angelis, Steven J Ford, Neal C Burton, Daniel Razansky, Dietrich Trümbach, Michaela Aichler, Axel Karl Walch, Julia Calzada-Wack, Frauke Neff, Wolfgang Wurst, Tobias Hartmann, and Thomas Floss
- Subjects
Medicine ,Science - Abstract
Sphingolipids and the derived gangliosides have critical functions in spermatogenesis, thus mutations in genes involved in sphingolipid biogenesis are often associated with male infertility. We have generated a transgenic mouse line carrying an insertion in the sphingomyelin synthase gene Sms1, the enzyme which generates sphingomyelin species in the Golgi apparatus. We describe the spermatogenesis defect of Sms1-/- mice, which is characterized by sloughing of spermatocytes and spermatids, causing progressive infertility of male homozygotes. Lipid profiling revealed a reduction in several long chain unsaturated phosphatidylcholins, lysophosphatidylcholins and sphingolipids in the testes of mutants. Multi-Spectral Optoacoustic Tomography indicated blood-testis barrier dysfunction. A supplementary diet of the essential omega-3 docosahexaenoic acid and eicosapentaenoic acid diminished germ cell sloughing from the seminiferous epithelium and restored spermatogenesis and fertility in 50% of previously infertile mutants. Our findings indicate that SMS1 has a wider than anticipated role in testis polyunsaturated fatty acid homeostasis and for male fertility.
- Published
- 2016
- Full Text
- View/download PDF
9. CIP2A Promotes T-Cell Activation and Immune Response to Listeria monocytogenes Infection.
- Author
-
Christophe Côme, Anna Cvrljevic, Mohd Moin Khan, Irina Treise, Thure Adler, Juan Antonio Aguilar-Pimentel, Byron Au-Yeung, Eleonora Sittig, Teemu Daniel Laajala, Yiling Chen, Sebastian Oeder, Julia Calzada-Wack, Marion Horsch, Tero Aittokallio, Dirk H Busch, Markus W Ollert, Frauke Neff, Johannes Beckers, Valerie Gailus-Durner, Helmut Fuchs, Martin Hrabě de Angelis, Zhi Chen, Riitta Lahesmaa, and Jukka Westermarck
- Subjects
Medicine ,Science - Abstract
The oncoprotein Cancerous Inhibitor of Protein Phosphatase 2A (CIP2A) is overexpressed in most malignancies and is an obvious candidate target protein for future cancer therapies. However, the physiological importance of CIP2A-mediated PP2A inhibition is largely unknown. As PP2A regulates immune responses, we investigated the role of CIP2A in normal immune system development and during immune response in vivo. We show that CIP2A-deficient mice (CIP2AHOZ) present a normal immune system development and function in unchallenged conditions. However when challenged with Listeria monocytogenes, CIP2AHOZ mice display an impaired adaptive immune response that is combined with decreased frequency of both CD4+ T-cells and CD8+ effector T-cells. Importantly, the cell autonomous effect of CIP2A deficiency for T-cell activation was confirmed. Induction of CIP2A expression during T-cell activation was dependent on Zap70 activity. Thus, we reveal CIP2A as a hitherto unrecognized mediator of T-cell activation during adaptive immune response. These results also reveal CIP2AHOZ as a possible novel mouse model for studying the role of PP2A activity in immune regulation. On the other hand, the results also indicate that CIP2A targeting cancer therapies would not cause serious immunological side-effects.
- Published
- 2016
- Full Text
- View/download PDF
10. Cox4i2, Ifit2, and Prdm11 Mutant Mice: Effective Selection of Genes Predisposing to an Altered Airway Inflammatory Response from a Large Compendium of Mutant Mouse Lines.
- Author
-
Marion Horsch, Juan Antonio Aguilar-Pimentel, Clemens Bönisch, Christophe Côme, Cathrine Kolster-Fog, Klaus T Jensen, Anders H Lund, Icksoo Lee, Lawrence I Grossman, Christopher Sinkler, Maik Hüttemann, Erwin Bohn, Helmut Fuchs, Markus Ollert, Valérie Gailus-Durner, Martin Hrabĕ de Angelis, and Johannes Beckers
- Subjects
Medicine ,Science - Abstract
We established a selection strategy to identify new models for an altered airway inflammatory response from a large compendium of mutant mouse lines that were systemically phenotyped in the German Mouse Clinic (GMC). As selection criteria we included published gene functional data, as well as immunological and transcriptome data from GMC phenotyping screens under standard conditions. Applying these criteria we identified a few from several hundred mutant mouse lines and further characterized the Cox4i2tm1Hutt, Ifit2tm1.1Ebsb, and Prdm11tm1.1ahl lines following ovalbumin (OVA) sensitization and repeated OVA airway challenge. Challenged Prdm11tm1.1ahl mice exhibited changes in B cell counts, CD4+ T cell counts, and in the number of neutrophils in bronchoalveolar lavages, whereas challenged Ifit2tm1.1Ebsb mice displayed alterations in plasma IgE, IgG1, IgG3, and IgM levels compared to the challenged wild type littermates. In contrast, challenged Cox4i2tm1Hutt mutant mice did not show alterations in the humoral or cellular immune response compared to challenged wild type mice. Transcriptome analyses from lungs of the challenged mutant mouse lines showed extensive changes in gene expression in Prdm11tm1.1ahl mice. Functional annotations of regulated genes of all three mutant mouse lines were primarily related to inflammation and airway smooth muscle (ASM) remodeling. We were thus able to define an effective selection strategy to identify new candidate genes for the predisposition to an altered airway inflammatory response under OVA challenge conditions. Similar selection strategies may be used for the analysis of additional genotype-envirotype interactions for other diseases.
- Published
- 2015
- Full Text
- View/download PDF
11. Uromodulin retention in thick ascending limb of Henle's loop affects SCD1 in neighboring proximal tubule: renal transcriptome studies in mouse models of uromodulin-associated kidney disease.
- Author
-
Marion Horsch, Johannes Beckers, Helmut Fuchs, Valérie Gailus-Durner, Martin Hrabě de Angelis, Birgit Rathkolb, Eckhard Wolf, Bernhard Aigner, and Elisabeth Kemter
- Subjects
Medicine ,Science - Abstract
Uromodulin-associated kidney disease (UAKD) is a hereditary progressive renal disease which can lead to renal failure and requires renal replacement therapy. UAKD belongs to the endoplasmic reticulum storage diseases due to maturation defect of mutant uromodulin and its retention in the enlarged endoplasmic reticulum in the cells of the thick ascending limb of Henle's loop (TALH). Dysfunction of TALH represents the key pathogenic mechanism of UAKD causing the clinical symptoms of this disease. However, the molecular alterations underlying UAKD are not well understood. In this study, transcriptome profiling of whole kidneys of two mouse models of UAKD, UmodA227T and UmodC93F, was performed. Genes differentially abundant in UAKD affected kidneys of both Umod mutant lines at different disease stages were identified and verified by RT-qPCR. Additionally, differential protein abundances of SCD1 and ANGPTL7 were validated by immunohistochemistry and Western blot analysis. ANGPTL7 expression was down-regulated in TALH cells of Umod mutant mice which is the site of the mutant uromodulin maturation defect. SCD1 was expressed selectively in the S3 segment of proximal tubule cells, and SCD1 abundance was increased in UAKD affected kidneys. This finding demonstrates that a cross talk between two functionally distinct tubular segments of the kidney, the TALH segment and the S3 segment of proximal tubule, exists.
- Published
- 2014
- Full Text
- View/download PDF
12. Prdm6 is essential for cardiovascular development in vivo.
- Author
-
Andreas Gewies, Mercedes Castineiras-Vilarino, Uta Ferch, Nina Jährling, Katja Heinrich, Ulrike Hoeckendorf, Gerhard K H Przemeck, Matthias Munding, Olaf Groß, Timm Schroeder, Marion Horsch, E Loraine Karran, Aneela Majid, Stefan Antonowicz, Johannes Beckers, Martin Hrabé de Angelis, Hans-Ulrich Dodt, Christian Peschel, Irmgard Förster, Martin J S Dyer, and Jürgen Ruland
- Subjects
Medicine ,Science - Abstract
Members of the PRDM protein family have been shown to play important roles during embryonic development. Previous in vitro and in situ analyses indicated a function of Prdm6 in cells of the vascular system. To reveal physiological functions of Prdm6, we generated conditional Prdm6-deficient mice. Complete deletion of Prdm6 results in embryonic lethality due to cardiovascular defects associated with aberrations in vascular patterning. However, smooth muscle cells could be regularly differentiated from Prdm6-deficient embryonic stem cells and vascular smooth muscle cells were present and proliferated normally in Prdm6-deficient embryos. Conditional deletion of Prdm6 in the smooth muscle cell lineage using a SM22-Cre driver line resulted in perinatal lethality due to hemorrhage in the lungs. We thus identified Prdm6 as a factor that is essential for the physiological control of cardiovascular development.
- Published
- 2013
- Full Text
- View/download PDF
13. Standardized, systemic phenotypic analysis of Umod(C93F) and Umod(A227T) mutant mice.
- Author
-
Elisabeth Kemter, Petra Prückl, Birgit Rathkolb, Kateryna Micklich, Thure Adler, Lore Becker, Johannes Beckers, Dirk H Busch, Alexander A Götz, Wolfgang Hans, Marion Horsch, Boris Ivandic, Martin Klingenspor, Thomas Klopstock, Jan Rozman, Anja Schrewe, Holger Schulz, Helmut Fuchs, Valérie Gailus-Durner, Martin Hrabé de Angelis, Eckhard Wolf, and Bernhard Aigner
- Subjects
Medicine ,Science - Abstract
Uromodulin-associated kidney disease (UAKD) summarizes different clinical features of an autosomal dominant heritable disease syndrome in humans with a proven uromodulin (UMOD) mutation involved. It is often characterized by hyperuricemia, gout, alteration of urine concentrating ability, as well as a variable rate of disease progression inconstantly leading to renal failure and histological alterations of the kidneys. We recently established the two Umod mutant mouse lines Umod(C93F) and Umod(A227T) on the C3H inbred genetic background both showing kidney defects analogous to those found in human UAKD patients. In addition, disease symptoms were revealed that were not yet described in other published mouse models of UAKD. To examine if further organ systems and/or metabolic pathways are affected by Umod mutations as primary or secondary effects, we describe a standardized, systemic phenotypic analysis of the two mutant mouse lines Umod(A227T) and Umod(C93F) in the German Mouse Clinic. Different genotypes as well as different ages were tested. Beside the already published changes in body weight, body composition and bone metabolism, the influence of the Umod mutation on energy metabolism was confirmed. Hematological analysis revealed a moderate microcytic and erythropenic anemia in older Umod mutant mice. Data of the other analyses in 7-10 month-old mutant mice showed single small additional effects.
- Published
- 2013
- Full Text
- View/download PDF
14. Identification of C/EBPβ target genes in ALK+ anaplastic large cell lymphoma (ALCL) by gene expression profiling and chromatin immunoprecipitation.
- Author
-
Irina Bonzheim, Martin Irmler, Margit Klier-Richter, Julia Steinhilber, Nataša Anastasov, Sabine Schäfer, Patrick Adam, Johannes Beckers, Mark Raffeld, Falko Fend, and Leticia Quintanilla-Martinez
- Subjects
Medicine ,Science - Abstract
C/EBPβ (CCAAT enhancer binding protein) is a transcription factor that plays a crucial role in survival and transformation of ALK+ anaplastic large cell lymphoma (ALCL). The aim of this study was to identify the downstream targets of C/EBPβ responsible for ALK-mediated oncogenesis. C/EBPβ was knocked down in ALK+ ALCL cell lines with a C/EBPβ-shRNA, followed by gene expression profiling (GEP). GEP analysis revealed a reproducible signature of genes that were significantly regulated by C/EBPβ. Classification into biological categories revealed overrepresentation of genes involved in the immune response, apoptosis and cell proliferation. Transcriptional regulation by C/EBPβ was found in 6 of 11 (BCL2A1, G0S2, TRIB1, S100A9, DDX21 and DDIT4) genes investigated by chromatin immunoprecipitation. We demonstrated that BCL2A1, G0S2 and DDX21 play a crucial role in survival and proliferation of ALK+ ALCL cells. DDX21, a gene involved in rRNA biogenesis, was found differentially overexpressed in primary ALK+ ALCL cases. All three candidate genes were validated in primary ALCL cases by either immunohistochemistry or RT-qPCR. In conclusion, we identified and validated several key C/EBPβ-regulated genes with major impact on survival and cell growth in ALK+ ALCL, supporting the central role of C/EBPβ in ALK-mediated oncogenesis.
- Published
- 2013
- Full Text
- View/download PDF
15. In vivo functional requirement of the mouse Ifitm1 gene for germ cell development, interferon mediated immune response and somitogenesis.
- Author
-
Ingeborg Klymiuk, Lukas Kenner, Thure Adler, Dirk H Busch, Auke Boersma, Martin Irmler, Barbara Fridrich, Valérie Gailus-Durner, Helmut Fuchs, Nicole Leitner, Mathias Müller, Ralf Kühn, Michaela Schlederer, Irina Treise, Martin Hrabě de Angelis, and Johannes Beckers
- Subjects
Medicine ,Science - Abstract
The mammalian Interferon induced transmembrane protein 1 (Ifitm1) gene was originally identified as a member of a gene family highly inducible by type I and type II interferons. Based on expression analyses, it was suggested to be required for normal primordial germ cell migration. The knockdown of Ifitm1 in mouse embryos provided evidence for a role in somitogenesis. We generated the first targeted knockin allele of the Ifitm1 gene to systematically reassess all inferred functions. Sperm motility and the fertility of male and female mutant mice are as in wild type littermates. Embryonic somites and the adult vertebral column appear normal in homozygous Ifitm1 knockout mice, demonstrating that Ifitm1 is not essential for normal segmentation of the paraxial mesoderm. Proportions of leucocyte subsets, including granulocytes, monocytes, B-cells, T-cells, NK-cells, and NKT-cells, are unchanged in mutant mice. Based on a normal immune response to Listeria monocytogenes infection, there is no evidence for a dysfunction in downstream IFNγ signaling in Ifitm1 mutant mice. Expression from the Ifitm1 locus from E8.5 to E14.5 is highly dynamic. In contrast, in adult mice, Ifitm1 expression is highly restricted and strong in the bronchial epithelium. Intriguingly, IFITM1 is highly overexpressed in tumor epithelia cells of human squamous cell carcinomas and in adenocarcinomas of NSCLC patients. These analyses underline the general importance of targeted in vivo studies for the functional annotation of the mammalian genome. The first comprehensive description of the Ifitm1 expression pattern provides a rational basis for the further examination of Ifitm1 gene functions. Based on our data, the fact that IFITM1 can function as a negative regulator of cell proliferation, and because the gene maps to chromosome band 11p15.5, previously associated with NSCLC, it is likely that IFITM1 in man has a key role in tumor formation.
- Published
- 2012
- Full Text
- View/download PDF
16. Correction: Functional Requirement of the Mouse Gene for Germ Cell Development, Interferon Mediated Immune Response and Somitogenesis.
- Author
-
Ingeborg Klymiuk, Lukas Kenner, Thure Adler, Dirk H. Busch, Auke Boersma, Martin Irmler, Barbara Fridrich, Valérie Gailus-Durner, Helmut Fuchs, Nicole Leitner, Mathias Müller, Ralf Kühn, Michaela Schlederer, Irina Treise, Martin Hrabě de Angelis, and Johannes Beckers
- Subjects
Medicine ,Science - Published
- 2012
- Full Text
- View/download PDF
17. Integration of transcriptomics, proteomics, and microRNA analyses reveals novel microRNA regulation of targets in the mammalian inner ear.
- Author
-
Tal Elkan-Miller, Igor Ulitsky, Ronna Hertzano, Anya Rudnicki, Amiel A Dror, Danielle R Lenz, Ran Elkon, Martin Irmler, Johannes Beckers, Ron Shamir, and Karen B Avraham
- Subjects
Medicine ,Science - Abstract
We have employed a novel approach for the identification of functionally important microRNA (miRNA)-target interactions, integrating miRNA, transcriptome and proteome profiles and advanced in silico analysis using the FAME algorithm. Since miRNAs play a crucial role in the inner ear, demonstrated by the discovery of mutations in a miRNA leading to human and mouse deafness, we applied this approach to microdissected auditory and vestibular sensory epithelia. We detected the expression of 157 miRNAs in the inner ear sensory epithelia, with 53 miRNAs differentially expressed between the cochlea and vestibule. Functionally important miRNAs were determined by searching for enriched or depleted targets in the transcript and protein datasets with an expression consistent with the dogma of miRNA regulation. Importantly, quite a few of the targets were detected only in the protein datasets, attributable to regulation by translational suppression. We identified and experimentally validated the regulation of PSIP1-P75, a transcriptional co-activator previously unknown in the inner ear, by miR-135b, in vestibular hair cells. Our findings suggest that miR-135b serves as a cellular effector, involved in regulating some of the differences between the cochlear and vestibular hair cells.
- Published
- 2011
- Full Text
- View/download PDF
18. Loss of the actin remodeler Eps8 causes intestinal defects and improved metabolic status in mice.
- Author
-
Arianna Tocchetti, Charlotte Blanche Ekalle Soppo, Fabio Zani, Fabrizio Bianchi, Maria Cristina Gagliani, Benedetta Pozzi, Jan Rozman, Ralf Elvert, Nicole Ehrhardt, Birgit Rathkolb, Corinna Moerth, Marion Horsch, Helmut Fuchs, Valérie Gailus-Durner, Johannes Beckers, Martin Klingenspor, Eckhard Wolf, Martin Hrabé de Angelis, Eugenio Scanziani, Carlo Tacchetti, Giorgio Scita, Pier Paolo Di Fiore, and Nina Offenhäuser
- Subjects
Medicine ,Science - Abstract
BACKGROUND: In a variety of organisms, including mammals, caloric restriction improves metabolic status and lowers the incidence of chronic-degenerative diseases, ultimately leading to increased lifespan. METHODOLOGY/PRINCIPAL FINDINGS: Here we show that knockout mice for Eps8, a regulator of actin dynamics, display reduced body weight, partial resistance to age- or diet-induced obesity, and overall improved metabolic status. Alteration in the liver gene expression profile, in behavior and metabolism point to a calorie restriction-like phenotype in Eps8 knockout mice. Additionally, and consistent with a calorie restricted metabolism, Eps8 knockout mice show increased lifespan. The metabolic alterations in Eps8 knockout mice correlated with a significant reduction in intestinal fat absorption presumably caused by a 25% reduction in intestinal microvilli length. CONCLUSIONS/SIGNIFICANCE: Our findings implicate actin dynamics as a novel variable in the determination of longevity. Additionally, our observations suggest that subtle differences in energy balance can, over time, significantly affect bodyweight and metabolic status in mice.
- Published
- 2010
- Full Text
- View/download PDF
19. Dll1 haploinsufficiency in adult mice leads to a complex phenotype affecting metabolic and immunological processes.
- Author
-
Isabel Rubio-Aliaga, Gerhard K H Przemeck, Helmut Fuchs, Valérie Gailus-Durner, Thure Adler, Wolfgang Hans, Marion Horsch, Birgit Rathkolb, Jan Rozman, Anja Schrewe, Sibylle Wagner, Sabine M Hoelter, Lore Becker, Thomas Klopstock, Wolfgang Wurst, Eckhard Wolf, Martin Klingenspor, Boris T Ivandic, Dirk H Busch, Johannes Beckers, and Martin Hrabé de Angelis
- Subjects
Medicine ,Science - Abstract
BACKGROUND: The Notch signaling pathway is an evolutionary conserved signal transduction pathway involved in embryonic patterning and regulation of cell fates during development and self-renewal. Recent studies have demonstrated that this pathway is integral to a complex system of interactions, involving as well other signal transduction pathways, and implicated in distinct human diseases. Delta-like 1 (Dll1) is one of the known ligands of the Notch receptors. The role of the Notch ligands is less well understood. Loss-of-function of Dll1 leads to embryonic lethality, but reduction of Delta-like 1 protein levels has not been studied in adult stage. METHODOLOGY/PRINCIPAL FINDINGS: Here we present the haploinsufficient phenotype of Dll1 and a missense mutant Dll1 allele (Dll1(C413Y)). Haploinsufficiency leads to a complex phenotype with several biological processes altered. These alterations reveal the importance of Dll1 mainly in metabolism, energy balance and in immunology. The animals are smaller, lighter, with altered fat to lean ratio and have increased blood pressure and a slight bradycardia. The animals have reduced cholesterol and triglyceride levels in blood. At the immunological level a subtle phenotype is observed due to the effect and fine-tuning of the signaling network at the different levels of differentiation, proliferation and function of lymphocytes. Moreover, the importance of the proteolytic regulation of the Notch signaling network emphasized. CONCLUSIONS/SIGNIFICANCE: In conclusion, slight alterations in one player of Notch signaling alter the entire organism, emphasizing the fine-tuning character of this pathway in a high number of processes.
- Published
- 2009
- Full Text
- View/download PDF
20. Inceptor counteracts insulin signalling in β-cells to control glycaemia
- Author
-
Michal Grzybek, Matthias H. Tschöp, Oliver Plettenburg, Sara Bilekova, Lena Oppenländer, Johanna Siehler, Michael Sterr, Amir Morshedi, Sarah Homberg, Timo D. Müller, Aurelia Raducanu, Katharina Wißmiller, Felizitas Gräfin von Hahn, Ansarullah, Chirag Jain, Julius Wiener, Ünal Coskun, Johannes Beckers, Heiko Lickert, Gustav Collden, Fataneh Fathi Far, Silvia Schirge, Aimée Bastidas-Ponce, Christin Ahlbrecht, Annette Feuchtinger, Matthias Meier, Martin Irmler, and Regina Feederle
- Subjects
Blood Glucose ,Male ,0301 basic medicine ,medicine.medical_treatment ,Golgi Apparatus ,Endoplasmic Reticulum ,Insulin Antagonists ,Mice ,0302 clinical medicine ,Insulin-Secreting Cells ,Insulin ,Insulin-Like Growth Factor I ,Receptor ,Glucose tolerance test ,Multidisciplinary ,medicine.diagnostic_test ,biology ,Chemistry ,Endocytosis ,Neoplasm Proteins ,Knockout mouse ,hormones, hormone substitutes, and hormone antagonists ,Signal Transduction ,endocrine system ,medicine.medical_specialty ,030209 endocrinology & metabolism ,Cell Line ,03 medical and health sciences ,Commentaries ,Internal medicine ,medicine ,Animals ,Humans ,Cell Proliferation ,Cell Size ,Insulin-like growth factor 1 receptor ,Growth factor ,Insulin-like growth factor 2 receptor ,Membrane Proteins ,nutritional and metabolic diseases ,Glucose Tolerance Test ,Clathrin ,Receptor, Insulin ,Tamoxifen ,Insulin receptor ,030104 developmental biology ,Endocrinology ,Commentary ,biology.protein ,Endocrine Cells ,Lysosomes - Abstract
Resistance to insulin and insulin-like growth factor 1 (IGF1) in pancreatic β-cells causes overt diabetes in mice; thus, therapies that sensitize β-cells to insulin may protect patients with diabetes against β-cell failure1–3. Here we identify an inhibitor of insulin receptor (INSR) and IGF1 receptor (IGF1R) signalling in mouse β-cells, which we name the insulin inhibitory receptor (inceptor; encoded by the gene Iir). Inceptor contains an extracellular cysteine-rich domain with similarities to INSR and IGF1R4, and a mannose 6-phosphate receptor domain that is also found in the IGF2 receptor (IGF2R)5. Knockout mice that lack inceptor (Iir−/−) exhibit signs of hyperinsulinaemia and hypoglycaemia, and die within a few hours of birth. Molecular and cellular analyses of embryonic and postnatal pancreases from Iir−/− mice showed an increase in the activation of INSR–IGF1R in Iir−/− pancreatic tissue, resulting in an increase in the proliferation and mass of β-cells. Similarly, inducible β-cell-specific Iir−/− knockout in adult mice and in ex vivo islets led to an increase in the activation of INSR–IGF1R and increased proliferation of β-cells, resulting in improved glucose tolerance in vivo. Mechanistically, inceptor interacts with INSR–IGF1R to facilitate clathrin-mediated endocytosis for receptor desensitization. Blocking this physical interaction using monoclonal antibodies against the extracellular domain of inceptor resulted in the retention of inceptor and INSR at the plasma membrane to sustain the activation of INSR–IGF1R in β-cells. Together, our findings show that inceptor shields insulin-producing β-cells from constitutive pathway activation, and identify inceptor as a potential molecular target for INSR–IGF1R sensitization and diabetes therapy. The insulin inhibitory receptor (inceptor) is identified as a negative regulator of insulin and IGF1 signalling that could be targeted for β-cell regeneration in treatments for diabetes.
- Published
- 2021
21. Non-canonical Wnt/PCP signalling regulates intestinal stem cell lineage priming towards enteroendocrine and Paneth cell fates
- Author
-
Johannes Beckers, Fabian J. Theis, Wolfgang Enard, Maren Büttner, Christoph Ziegenhain, Heiko Lickert, Fien M. Verhamme, Lena Oppenländer, Anika Böttcher, Sophie Tritschler, Oliver Eickelberg, Michael Sterr, Andrea C. Schamberger, Alexandra Aliluev, Martin Irmler, Steffen Sass, and Ingo Burtscher
- Subjects
inorganic chemicals ,Male ,Paneth Cells ,Lineage (genetic) ,Enteroendocrine Cells ,Enteroendocrine cell ,Biology ,digestive system ,Receptors, G-Protein-Coupled ,Mice ,03 medical and health sciences ,0302 clinical medicine ,Single-cell analysis ,Cell polarity ,medicine ,Animals ,Cell Lineage ,Cell Self Renewal ,Intestinal Mucosa ,beta Catenin ,030304 developmental biology ,Mice, Knockout ,0303 health sciences ,Gene Expression Profiling ,Stem Cells ,Wnt signaling pathway ,LGR5 ,Cell Polarity ,Cell Biology ,Cell biology ,Mice, Inbred C57BL ,Wnt Proteins ,medicine.anatomical_structure ,030220 oncology & carcinogenesis ,Paneth cell ,Female ,Single-Cell Analysis ,Stem cell - Abstract
A detailed understanding of intestinal stem cell (ISC) self-renewal and differentiation is required to treat chronic intestinal diseases. However, the different models of ISC lineage hierarchy1–6 and segregation7–12 are subject to debate. Here, we have discovered non-canonical Wnt/planar cell polarity (PCP)-activated ISCs that are primed towards the enteroendocrine or Paneth cell lineage. Strikingly, integration of time-resolved lineage labelling with single-cell gene expression analysis revealed that both lineages are directly recruited from ISCs via unipotent transition states, challenging the existence of formerly predicted bi- or multipotent secretory progenitors7–12. Transitory cells that mature into Paneth cells are quiescent and express both stem cell and secretory lineage genes, indicating that these cells are the previously described Lgr5+ label-retaining cells7. Finally, Wnt/PCP-activated Lgr5+ ISCs are molecularly indistinguishable from Wnt/β-catenin-activated Lgr5+ ISCs, suggesting that lineage priming and cell-cycle exit is triggered at the post-transcriptional level by polarity cues and a switch from canonical to non-canonical Wnt/PCP signalling. Taken together, we redefine the mechanisms underlying ISC lineage hierarchy and identify the Wnt/PCP pathway as a new niche signal preceding lateral inhibition in ISC lineage priming and segregation. Polarity cues regulate intestinal stem cell fate. Bottcher et al. demonstrate that mouse intestinal stem cells, which express the Wnt/planar cell polarity reporter Flattop, are primed either towards the enteroendocrine or Paneth cell lineage.
- Published
- 2021
22. Nutrition and its role in epigenetic inheritance of obesity and diabetes across generations
- Author
-
Johannes Beckers, Sieglinde Hastreiter, Daniela Kaspar, Martin Hrabé de Angelis, and Martin Irmler
- Subjects
Male ,Inheritance Patterns ,Embryonic Development ,Endocrine Disruptors ,Article ,Histones ,03 medical and health sciences ,Overnutrition ,0302 clinical medicine ,Insulin resistance ,Glucose Intolerance ,Genetics ,medicine ,Animals ,Humans ,Obesity ,RNA, Messenger ,Epigenetics ,Gene ,030304 developmental biology ,2. Zero hunger ,0303 health sciences ,biology ,Malnutrition ,Inheritance (genetic algorithm) ,DNA Methylation ,medicine.disease ,Human genetics ,ddc ,medicine.anatomical_structure ,Histone ,Diabetes Mellitus, Type 2 ,biology.protein ,Female ,Gene-Environment Interaction ,Insulin Resistance ,Reprogramming ,030217 neurology & neurosurgery ,Germ cell - Abstract
Nutritional constraints including not only caloric restriction or protein deficiency, but also energy-dense diets affect metabolic health and frequently lead to obesity and insulin resistance, as well as glucose intolerance and type 2 diabetes. The effects of these environmental factors are often mediated via epigenetic modifiers that target the expression of metabolic genes. More recently, it was discovered that such parentally acquired metabolic changes can alter the metabolic health of the filial and grand-filial generations. In mammals, this epigenetic inheritance can either follow an intergenerational or transgenerational mode of inheritance. In the case of intergenerational inheritance, epimutations established in gametes persist through the first round of epigenetic reprogramming occurring during preimplantation development. For transgenerational inheritance, epimutations persist additionally throughout the reprogramming that occurs during germ cell development later in embryogenesis. Differentially expressed transcripts, genomic cytosine methylations, and several chemical modifications of histones are prime candidates for tangible marks which may serve as epimutations in inter- and transgenerational inheritance and which are currently being investigated experimentally. We review, here, the current literature in support of epigenetic inheritance of metabolic traits caused by nutritional constraints and potential mechanisms in man and in rodent model systems.
- Published
- 2020
23. A novel stem cell type at the basal side of the subventricular zone maintains adult neurogenesis
- Author
-
Johannes Beckers, Priti Khatri, Claudia Mandl, Yan Shi, Gabriele Hoelzl-Wenig, Yomn Abdullah, Katja Baur, Udo Schmidt-Edelkraut, Martin Irmler, Anna M. Hagenston, and Francesca Ciccolini
- Subjects
Neurogenesis ,Subventricular zone ,Biology ,Neural Stem Cells ,Notch Signalling ,Olfactory Bulb ,Subventricular Zone ,Biochemistry ,Basal (phylogenetics) ,Mice ,Lateral Ventricles ,Genetics ,medicine ,Animals ,Progenitor cell ,Molecular Biology ,reproductive and urinary physiology ,EMBO27 ,EMBO34 ,Article ,Articles ,neural stem cells ,neurogenesis ,notch signalling ,olfactory bulb ,subventricular zone ,Cell Differentiation ,Nestin ,Neural stem cell ,nervous system diseases ,Cell biology ,Olfactory bulb ,ddc ,medicine.anatomical_structure ,nervous system ,Female ,biological phenomena, cell phenomena, and immunity ,Stem cell - Abstract
According to the current consensus, neural stem cells (NSCs) apically contacting the lateral ventricle generate differentiated progenitors by rare asymmetric divisions or by relocating to the basal side of the ventricular-subventricular zone V-SVZ. Both processes will then ultimately lead to the generation of adult-born olfactory bulb (OB) interneurons. In contrast to this view, we here found that adult-born OB interneurons largely derive from an additional NSC type resident in the basal V-SVZ. Despite being both capable of self-renewal and long-term quiescence, apical and basal NSCs differ in Nestin expression, primary cilia extension and frequency of cell division. The expression of Notch-related genes also differed between the two NSC groups and Notch-activation was greatest in apical NSCs. Apical downregulation of Notch-effector Hes1 decreased Notch activation while increasing proliferation across the niche and neurogenesis from apical NSCs. Underscoring their different roles in neurogenesis, lactation-dependent increase in neurogenesis was paralleled by extra activation of basal but not apical NSCs. Thus, basal NSCs support OB neurogenesis whereas apical NSCs impart Notch-mediated lateral inhibition across the V-SVZ.
- Published
- 2021
24. Point mutations in the PDX1 transactivation domain impair human β-cell development and function
- Author
-
Filippo M. Cernilogar, Gunnar Schotta, Julia Beckenbauer, Ingo Burtscher, Anika Böttcher, Ansarullah, Martin Irmler, Thomas Meitinger, Xianming Wang, Johanna Siehler, Harald Staiger, Michael Sterr, Johannes Beckers, Mostafa Bakhti, Heiko Lickert, Hans-Ulrich Häring, and Christopher V.E. Wright
- Subjects
Adult ,Male ,0301 basic medicine ,lcsh:Internal medicine ,endocrine system ,Lineage (genetic) ,endocrine system diseases ,β-Cell differentiation ,030209 endocrinology & metabolism ,Biology ,medicine.disease_cause ,digestive system ,Cell Line ,03 medical and health sciences ,Transactivation ,0302 clinical medicine ,Protein Domains ,Loss of Function Mutation ,Insulin-Secreting Cells ,Pdx1 ,Transactivation Domain ,Beta-cell Differentiation ,Insulin Secretion ,Pdx1-bound Genes ,Diabetes Mellitus ,medicine ,Humans ,Point Mutation ,Missense mutation ,lcsh:RC31-1245 ,Molecular Biology ,Gene ,Homeodomain Proteins ,MEG3 ,PDX1 ,Mutation ,Transactivation domain ,Insulin secretion ,Point mutation ,Cell Differentiation ,Cell Biology ,Molecular biology ,ddc ,3. Good health ,030104 developmental biology ,Trans-Activators ,Original Article ,Female ,RNA, Long Noncoding ,Carboxylic Ester Hydrolases ,PDX1-Bound genes ,Transcription Factors - Abstract
Objective Hundreds of missense mutations in the coding region of PDX1 exist; however, if these mutations predispose to diabetes mellitus is unknown. Methods In this study, we screened a large cohort of subjects with increased risk for diabetes and identified two subjects with impaired glucose tolerance carrying common, heterozygous, missense mutations in the PDX1 coding region leading to single amino acid exchanges (P33T, C18R) in its transactivation domain. We generated iPSCs from patients with heterozygous PDX1P33T/+, PDX1C18R/+ mutations and engineered isogenic cell lines carrying homozygous PDX1P33T/P33T, PDX1C18R/C18R mutations and a heterozygous PDX1 loss-of-function mutation (PDX1+/−). Results Using an in vitro β-cell differentiation protocol, we demonstrated that both, heterozygous PDX1P33T/+, PDX1C18R/+ and homozygous PDX1P33T/P33T, PDX1C18R/C18R mutations impair β-cell differentiation and function. Furthermore, PDX1+/− and PDX1P33T/P33T mutations reduced differentiation efficiency of pancreatic progenitors (PPs), due to downregulation of PDX1-bound genes, including transcription factors MNX1 and PDX1 as well as insulin resistance gene CES1. Additionally, both PDX1P33T/+ and PDX1P33T/P33T mutations in PPs reduced the expression of PDX1-bound genes including the long-noncoding RNA, MEG3 and the imprinted gene NNAT, both involved in insulin synthesis and secretion. Conclusions Our results reveal mechanistic details of how common coding mutations in PDX1 impair human pancreatic endocrine lineage formation and β-cell function and contribute to the predisposition for diabetes., Highlights • Missense mutations in the transactivation domain reduce PDX1 target gene expression. • Lack of PDX1 target gene activation impairs both β-cell development and function. • Common PDX1 coding mutations likely predispose for diabetes.
- Published
- 2019
25. Combination therapies induce cancer cell death through the integrated stress response and disturbed pyrimidine metabolism
- Author
-
Johannes Beckers, Oliver Plettenburg, Sean Lin, Foivos-Filippos Tsokanos, Anna Artati, Susanne Seitz, Arlett Schäfer, Goetz Hartleben, Martin Irmler, Yun Kwon, Lisa Mehr, Mauricio Berriel Diaz, Kenji Schorpp, Anja Zeigerer, Barbara Betz, Zahra Dantes, Jerzy Adamsky, Pauline Morigny, José Manuel Monroy Kuhn, Stephan Herzig, Janina Tokarz, Kamyar Hadian, Ina Rothenaigner, Dominik Lutter, and Maximilian Reichert
- Subjects
0301 basic medicine ,pyrimidine ,Medicine (General) ,pyrimidine derivative ,cancer metabolism ,CHOP ,QH426-470 ,chemistry.chemical_compound ,0302 clinical medicine ,cell stress ,metabolic vulnerabilities ,Neoplasms ,Medicine ,antineoplastic agent ,Cancer ,cancer cell ,Cell Death ,catabolism ,Articles ,integrated stress response ,Drug repositioning ,Paclitaxel ,Pyrimidine metabolism ,tricyclic antidepressants ,Molecular Medicine ,metabolome ,Signal Transduction ,organoid ,animal experiment ,Antineoplastic Agents ,Article ,03 medical and health sciences ,R5-920 ,male ,pyrimidine synthesis ,Chemical Biology ,Metabolome ,Genetics ,Integrated stress response ,Humans ,controlled study ,ddc:610 ,human ,Cancer Metabolism ,Integrated Stress Response ,Metabolic Vulnerabilities ,Pyrimidine Metabolism ,Tricyclic Antidepressants ,mouse ,nonhuman ,business.industry ,animal model ,human cell ,niclosamide ,growth arrest and DNA damage inducible protein 153 ,medicine.disease ,pyrimidine metabolism ,030104 developmental biology ,Pyrimidines ,chemistry ,Cancer cell ,Cancer research ,Dewey Decimal Classification::600 | Technik::610 | Medizin, Gesundheit ,business ,transcriptome ,030217 neurology & neurosurgery ,neoplasm - Abstract
By accentuating drug efficacy and impeding resistance mechanisms, combinatorial, multi‐agent therapies have emerged as key approaches in the treatment of complex diseases, most notably cancer. Using high‐throughput drug screens, we uncovered distinct metabolic vulnerabilities and thereby identified drug combinations synergistically causing a starvation‐like lethal catabolic response in tumor cells from different cancer entities. Domperidone, a dopamine receptor antagonist, as well as several tricyclic antidepressants (TCAs), including imipramine, induced cancer cell death in combination with the mitochondrial uncoupler niclosamide ethanolamine (NEN) through activation of the integrated stress response pathway and the catabolic CLEAR network. Using transcriptome and metabolome analyses, we characterized a combinatorial response, mainly driven by the transcription factors CHOP and TFE3, which resulted in cell death through enhanced pyrimidine catabolism as well as reduced pyrimidine synthesis. Remarkably, the drug combinations sensitized human organoid cultures to the standard‐of‐care chemotherapy paclitaxel. Thus, our combinatorial approach could be clinically implemented into established treatment regimen, which would be further facilitated by the advantages of drug repurposing., This study identifies novel combinatorial drug treatments to induce death of different tumor cells, and defines the mechanisms of synergism between a mitochondrial uncoupler and antidepressants or dopamine receptor antagonists.
- Published
- 2021
26. Epithelial cell plasticity drives endoderm formation during gastrulation
- Author
-
Ansarullah, Dapeng Yang, Heiko Lickert, Johannes Beckers, Michael Sterr, Filippo M. Cernilogar, Fabian J. Theis, Ingo Burtscher, Katharina Scheibner, Martin Irmler, Maren Büttner, Silvia Schirge, Gunnar Schotta, and Anika Böttcher
- Subjects
Mesoderm ,Embryology ,Embryonic stem cells ,animal structures ,Epithelial-Mesenchymal Transition ,Time Factors ,Cellular differentiation ,Cell Plasticity ,Gestational Age ,Mice, Transgenic ,Germ layer ,Biology ,Article ,Cell Line ,Mice ,Endoderm formation ,medicine ,Animals ,Cell lineage ,Primitive streak ,Endoderm ,Gastrulation ,Gene Expression Regulation, Developmental ,Cell Differentiation ,Epithelial Cells ,Mouse Embryonic Stem Cells ,Cell Biology ,Publisher Correction ,Cell biology ,medicine.anatomical_structure ,Blastocyst ,Phenotype ,Epiblast ,embryonic structures ,Hepatocyte Nuclear Factor 3-beta ,Snail Family Transcription Factors - Abstract
It is generally accepted that epiblast cells ingress into the primitive streak by epithelial-to-mesenchymal transition (EMT) to give rise to the mesoderm; however, it is less clear how the endoderm acquires an epithelial fate. Here, we used embryonic stem cell and mouse embryo knock‐in reporter systems to combine time-resolved lineage labelling with high-resolution single-cell transcriptomics. This allowed us to resolve the morphogenetic programs that segregate the mesoderm from the endoderm germ layer. Strikingly, while the mesoderm is formed by classical EMT, the endoderm is formed independent of the key EMT transcription factor Snail1 by mechanisms of epithelial cell plasticity. Importantly, forkhead box transcription factor A2 (Foxa2) acts as an epithelial gatekeeper and EMT suppressor to shield the endoderm from undergoing a mesenchymal transition. Altogether, these results not only establish the morphogenetic details of germ layer formation, but also have broader implications for stem cell differentiation and cancer metastasis., Scheibner et al. demonstrate that, during gastrulation in the mouse, epithelial epiblast progenitors upregulate Foxa2 and form the definitive endoderm independently of a full EMT–MET cycle.
- Published
- 2021
27. Insights into energy balance dysregulation from a mouse model of methylmalonic aciduria
- Author
-
Julia Calzada-Wack, D. S. Froese, Johannes Beckers, Céline Bürer, G. T. Bommer, Jan Rozman, Andres Kaech, M. Forny, Daniel Hoces, J. P. Dewulf, Samuel E. Wuest, Stefan Kölker, M. Lucienne, Patrick Forny, Martin Irmler, Sven W. Sauer, Helmut Fuchs, Juan Antonio Aguilar-Pimentel, Matthias R. Baumgartner, F. Traversi, Birgit Rathkolb, Raffaele Gerlini, V. Gailus-Durner, M Hrabe de Angelis, and University of Zurich
- Subjects
medicine.medical_specialty ,Chemistry ,610 Medicine & health ,Metabolism ,Peroxisome ,Endocrinology ,medicine.anatomical_structure ,Mutase ,Methylmalonic aciduria ,10036 Medical Clinic ,Internal medicine ,Brown adipose tissue ,medicine ,Lean body mass ,Energy source ,Homeostasis - Abstract
Inherited disorders of mitochondrial metabolism, including isolated methylmalonic aciduria (MMAuria), present unique challenges to energetic homeostasis by disrupting energy producing pathways. To better understand global responses to energy shortage, we investigated a hemizygous mouse model of methylmalonyl-CoA mutase (Mmut) type MMAuria. We found Mmut mutant mice to have reduced appetite, energy expenditure and body mass compared to littermate controls, along with a relative reduction in lean mass but increase in fat mass. Brown adipose tissue showed a process of whitening, in line with lower body surface temperature and lesser ability to cope with cold challenge. Mutant mice had dysregulated plasma glucose, delayed glucose clearance and a lesser ability to regulate energy sources when switching from the fed to fasted state, while liver investigations indicated metabolite accumulation and altered expression of peroxisome proliferator-activated receptor and Fgf21-controlled pathways. Together, these indicate hypometabolism, energetic inflexibility and increased stores at the expense of active tissue as energy shortage consequences.
- Published
- 2021
28. Diet-induced alteration of intestinal stem cell function underlies obesity and prediabetes in mice
- Author
-
Johannes Beckers, Alexandra Aliluev, Sophie Tritschler, Michael Sterr, Alida S.D. Kindt, Malte D Luecken, Anika Böttcher, Julia Hinterdobler, Fabian J. Theis, Lena Oppenländer, Axel Walch, Kerstin Stemmer, Heiko Lickert, Na Sun, Tobias Greisle, Matthias H. Tschöp, Jan Krumsiek, and Martin Irmler
- Subjects
medicine.medical_specialty ,Endocrinology, Diabetes and Metabolism ,Peroxisome Proliferator-Activated Receptors ,Peroxisome proliferator-activated receptor ,Enteroendocrine cell ,Biology ,Diet, High-Fat ,digestive system ,Article ,Prediabetic State ,Mice ,Physiology (medical) ,Internal medicine ,Internal Medicine ,medicine ,Animals ,Cell Lineage ,Secretion ,Obesity ,ddc:610 ,Progenitor cell ,Cancer ,Cell Proliferation ,Progenitor ,chemistry.chemical_classification ,Stem Cells ,Fatty Acids ,Cell Biology ,medicine.disease ,ddc ,Intestines ,Metabolism ,Endocrinology ,chemistry ,Stem cell ,Metabolic syndrome ,Signal Transduction ,Hormone - Abstract
Excess nutrient uptake and altered hormone secretion in the gut contribute to a systemic energy imbalance, which causes obesity and an increased risk of type 2 diabetes and colorectal cancer. This functional maladaptation is thought to emerge at the level of the intestinal stem cells (ISCs). However, it is not clear how an obesogenic diet affects ISC identity and fate. Here we show that an obesogenic diet induces ISC and progenitor hyperproliferation, enhances ISC differentiation and cell turnover and changes the regional identities of ISCs and enterocytes in mice. Single-cell resolution of the enteroendocrine lineage reveals an increase in progenitors and peptidergic enteroendocrine cell types and a decrease in serotonergic enteroendocrine cell types. Mechanistically, we link increased fatty acid synthesis, Ppar signaling and the Insr–Igf1r–Akt pathway to mucosal changes. This study describes molecular mechanisms of diet-induced intestinal maladaptation that promote obesity and therefore underlie the pathogenesis of the metabolic syndrome and associated complications., A combination of single-cell approaches, lineage tracing and metabolomics is used to characterize the changes to intestinal stem cell function in the small intestine that underlie intestinal maladaptation in mice fed an obesogenic diet.
- Published
- 2020
29. Type 2 diabetes risk gene Dusp8 regulates hypothalamic Jnk signaling and insulin sensitivity
- Author
-
Florian Giesert, Dhiraj G. Kabra, Timo D. Müller, Kerstin Stemmer, Meri De Angelis, Martin Hrabé de Angelis, Fabian Seebacher, Martin Heni, Sonja C. Schriever, Ruchi Jain, Luke Harrison, Moya Wu, Martin Irmler, Rubén Nogueiras, Johannes Beckers, Hans-Ulrich Häring, Serge Luquet, Stephanie Kullmann, Chun-Xia Yi, Natalie Krahmer, Felipe Correa-da-Silva, Emily Violette Baumgart, Julien Castel, Paul T. Pfluger, Sarah Martinez, Peter Baumann, Mathias V. Schmidt, Jeffery D. Molkentin, Katrin Pfuhlmann, Hannah Schug, Jan Rozman, Wolfgang Wurst, Joachim Nagler, Matthias H. Tschöp, Helmholtz-Zentrum München (HZM), German Center for Diabetes Research - Deutsches Zentrum für Diabetesforschung [Neuherberg] (DZD), Technische Universität Munchen - Université Technique de Munich [Munich, Allemagne] (TUM), Institute for Diabetes Research and Metabolic Diseases [Tübingen, Germany], VU University Medical Center [Amsterdam], Netherlands Institute for Neuroscience (NIN), Royal Netherlands Academy of Arts and Sciences (KNAW), TUM School of Life Sciences Weihenstephan, Skane University Hospital [Malmo], Lund University [Lund], Synlab [Lausanne], Unité de Biologie Fonctionnelle et Adaptative (BFA (UMR_8251 / U1133)), Institut National de la Santé et de la Recherche Médicale (INSERM)-Centre National de la Recherche Scientifique (CNRS)-Université de Paris (UP), School Life Science Weihenstephan (TUM), Eberhard Karls Universität Tübingen = Eberhard Karls University of Tuebingen, German Research Center for Neurodegenerative Diseases - Deutsches Zentrum für Neurodegenerative Erkrankungen (DZNE), Munich Cluster for systems neurology [Munich] (SyNergy), Technische Universität Munchen - Université Technique de Munich [Munich, Allemagne] (TUM)-Ludwig-Maximilians-Universität München (LMU), Institute of Molecular Genetics of the Czech Academy of Sciences (IMG / CAS), Czech Academy of Sciences [Prague] (CAS), Howard Hughes Medical Institute (HHMI), Cincinnati Children's Hospital Medical Center, Institute of the Royal Netherlands Academy of Arts and Sciences, Max Planck Institute of Psychiatry, Max-Planck-Gesellschaft, Endocrinology, Laboratory for Endocrinology, Amsterdam Neuroscience - Cellular & Molecular Mechanisms, and Amsterdam Gastroenterology Endocrinology Metabolism
- Subjects
0301 basic medicine ,MAP Kinase Kinase 4 ,[SDV]Life Sciences [q-bio] ,Type 2 diabetes ,Mice ,chemistry.chemical_compound ,0302 clinical medicine ,Corticosterone ,enzymology [Hypothalamus] ,Glucose homeostasis ,genetics [Diabetes Mellitus, Experimental] ,Mice, Knockout ,Kinase ,Diabetes ,enzymology [Diabetes Mellitus, Experimental] ,General Medicine ,metabolism [Dual-Specificity Phosphatases] ,DUSP8 protein, mouse ,030220 oncology & carcinogenesis ,Dual-Specificity Phosphatases ,medicine.symptom ,Signal Transduction ,medicine.medical_specialty ,Hypothalamus ,enzymology [Diabetes Mellitus, Type 2] ,Inflammation ,genetics [Diabetes Mellitus, Type 2] ,Diabetes Mellitus, Experimental ,03 medical and health sciences ,Insulin resistance ,Commentaries ,Internal medicine ,Diabetes mellitus ,medicine ,Animals ,genetics [MAP Kinase Kinase 4] ,ddc:610 ,Obesity ,genetics [Dual-Specificity Phosphatases] ,business.industry ,metabolism [MAP Kinase Kinase 4] ,medicine.disease ,030104 developmental biology ,Endocrinology ,Metabolism ,Diabetes Mellitus, Type 2 ,chemistry ,Commentary ,Insulin Resistance ,business ,Hormone - Abstract
International audience; Recent genome-wide association studies (GWAS) identified DUSP8, encoding a dual-specificity phosphatase targeting mitogen-activated protein kinases, as a type 2 diabetes (T2D) risk gene. Here, we reveal that Dusp8 is a gatekeeper in the hypothalamic control of glucose homeostasis in mice and humans. Male, but not female, Dusp8 loss-of-function mice, either with global or corticotropin-releasing hormone neuron-specific deletion, had impaired systemic glucose tolerance and insulin sensitivity when exposed to high-fat diet (HFD). Mechanistically, we found impaired hypothalamic-pituitary-adrenal axis feedback, blunted sympathetic responsiveness, and chronically elevated corticosterone levels driven by hypothalamic hyperactivation of Jnk signaling. Accordingly, global Jnk1 ablation, AAV-mediated Dusp8 overexpression in the mediobasal hypothalamus, or metyrapone-induced chemical adrenalectomy rescued the impaired glucose homeostasis of obese male Dusp8-KO mice, respectively. The sex-specific role of murine Dusp8 in governing hypothalamic Jnk signaling, insulin sensitivity, and systemic glucose tolerance was consistent with functional MRI data in human volunteers that revealed an association of the DUSP8 rs2334499 risk variant with hypothalamic insulin resistance in men. Further, expression of DUSP8 was increased in the infundibular nucleus of T2D humans. In summary, our findings suggest the GWAS-identified gene Dusp8 as a novel hypothalamic factor that plays a functional role in the etiology of T2D.
- Published
- 2020
30. PAX6 mutation alters circadian rhythm and β cell function in mice without affecting glucose tolerance
- Author
-
Daniel Gradinger, Oana V. Amarie, Marina Rubey, Johannes Beckers, Jan Rozman, Anna-Lena Amend, Gerhard K. H. Przemeck, Moya Wu, Birgit Rathkolb, Raffaele Teperino, Annette Feuchtinger, Peter Huypens, Martin Hrabě de Angelis, Martin Irmler, Nirav Florian Chhabra, and Eckhard Wolf
- Subjects
Blood Glucose ,Male ,Retinal Ganglion Cells ,0301 basic medicine ,medicine.medical_specialty ,PAX6 Transcription Factor ,Physiology ,Mutant ,Medicine (miscellaneous) ,Context (language use) ,Biology ,medicine.disease_cause ,Article ,Retina ,General Biochemistry, Genetics and Molecular Biology ,03 medical and health sciences ,0302 clinical medicine ,Insulin-Secreting Cells ,Internal medicine ,Insulin Secretion ,medicine ,Animals ,Circadian rhythm ,lcsh:QH301-705.5 ,Transcription factor ,Mice, Inbred C3H ,Mutation ,Pancreatic islets ,Point mutation ,Endocrine system and metabolic diseases ,Optic Nerve ,Mice, Mutant Strains ,eye diseases ,Circadian Rhythm ,Glucose ,030104 developmental biology ,Endocrinology ,medicine.anatomical_structure ,Gene Expression Regulation ,Liver ,lcsh:Biology (General) ,sense organs ,PAX6 ,General Agricultural and Biological Sciences ,030217 neurology & neurosurgery - Abstract
The transcription factor PAX6 is involved in the development of the eye and pancreatic islets, besides being associated with sleep–wake cycles. Here, we investigated a point mutation in the RED subdomain of PAX6, previously described in a human patient, to present a comprehensive study of a homozygous Pax6 mutation in the context of adult mammalian metabolism and circadian rhythm. Pax6Leca2 mice lack appropriate retinal structures for light perception and do not display normal daily rhythmic changes in energy metabolism. Despite β cell dysfunction and decreased insulin secretion, mutant mice have normal glucose tolerance. This is associated with reduced hepatic glucose production possibly due to altered circadian variation in expression of clock and metabolic genes, thereby evading hyperglycemia. Hence, our findings show that while the RED subdomain is important for β cell functional maturity, the Leca2 mutation impacts peripheral metabolism via loss of circadian rhythm, thus revealing pleiotropic effects of PAX6., Nirav Chhabra et al. characterize adult mice carrying a homozygous mutation in Pax6 that was identified in a patient with foveal hypoplasia. They find that the Pax6 point mutation has pleiotropic effects, including defects in the mouse retinal structures, loss of the optic nerve, changes in energy metabolism and circadian rhythms, and dysregulation of genes expressed in the pancreas.
- Published
- 2020
31. Dose-Dependent and Subset-Specific Regulation of Midbrain Dopaminergic Neuron Differentiation by LEF1-Mediated WNT1/b-Catenin Signaling
- Author
-
Parivash Nouri, Sebastian Götz, Benedict Rauser, Martin Irmler, Changgeng Peng, Dietrich Trümbach, Christian Kempny, Carina G. Lechermeier, Agnes Bryniok, Andrea Dlugos, Ellen Euchner, Johannes Beckers, Claude Brodski, Claudia Klümper, Wolfgang Wurst, and Nilima Prakash
- Subjects
0301 basic medicine ,Biology ,Midbrain dopaminergic neuron differentiation ,Cell and Developmental Biology ,03 medical and health sciences ,0302 clinical medicine ,ddc:570 ,medicine ,WNT1 ,RSPO2 ,lcsh:QH301-705.5 ,mouse ,Original Research ,Wnt signaling pathway ,Cell Biology ,Cell biology ,030104 developmental biology ,medicine.anatomical_structure ,regenerative therapy ,lcsh:Biology (General) ,030220 oncology & carcinogenesis ,Catenin ,Parkinson’s disease ,Neuron ,Signal transduction ,dopamine ,nerve cell ,Developmental Biology ,Lymphoid enhancer-binding factor 1 - Abstract
The mesodiencephalic dopaminergic (mdDA) neurons, including the nigrostriatal subset that preferentially degenerates in Parkinson's Disease (PD), strongly depend on an accurately balanced Wingless-type MMTV integration site family member 1 (WNT1)/beta-catenin signaling pathway during their development. Loss of this pathway abolishes the generation of these neurons, whereas excessive WNT1/b-catenin signaling prevents their correct differentiation. The identity of the cells responding to this pathway in the developing mammalian ventral midbrain (VM) as well as the precise progression of WNT/b-catenin action in these cells are still unknown. We show that strong WNT/b-catenin signaling inhibits the differentiation of WNT/b-catenin-responding mdDA progenitors into PITX3+ and TH+ mdDA neurons by repressing the Pitx3 gene in mice. This effect is mediated by RSPO2, a WNT/b-catenin agonist, and lymphoid enhancer binding factor 1 (LEF1), an essential nuclear effector of the WNT/b-catenin pathway, via conserved LEF1/T-cell factor binding sites in the Pitx3 promoter. LEF1 expression is restricted to a caudolateral mdDA progenitor subset that preferentially responds to WNT/b-catenin signaling and gives rise to a fraction of all mdDA neurons. Our data indicate that an attenuation of WNT/b-catenin signaling in mdDA progenitors is essential for their correct differentiation into specific mdDA neuron subsets. This is an important consideration for stem cell-based regenerative therapies and in vitro models of neuropsychiatric diseases.
- Published
- 2020
32. Specific induction of double negative B cells during protective and pathogenic immune responses
- Author
-
Gisela Gabernet, Ulf Ziemann, Bernhard Hemmer, Sven Nahnsen, Simon Heumos, Greg Owens, Martin Irmler, Gildas Lepennetier, Miriam Kaminski, Jeffrey Bennett, Markus C. Kowarik, Christoph Ruschil, Zsuzsanna Hrasko, and Johannes Beckers
- Subjects
Male ,double negative B cells ,Antibodies, Viral ,Lymphocyte Activation ,autoimmune disorders ,Transcriptome ,Immunogenicity, Vaccine ,Autoimmune Disorders ,B Cells ,Double Negative B Cells ,Influenza Vaccination ,Neuromyelitis Optica Spectrum Disorder ,Tbe Vaccination ,Vaccination ,Original Research ,Aged, 80 and over ,B-Lymphocytes ,education.field_of_study ,biology ,neuromyelitis optica spectrum disorder ,Middle Aged ,influenza vaccination ,Phenotype ,medicine.anatomical_structure ,Influenza Vaccines ,Female ,Antibody ,Encephalitis ,Adult ,Adolescent ,Immunology ,Antigens, CD19 ,Population ,Naive B cell ,Communicable Diseases ,CD19 ,TBE vaccination ,Encephalitis Viruses, Tick-Borne ,Young Adult ,Immune system ,medicine ,Humans ,ddc:610 ,education ,B cell ,Aged ,Cell Proliferation ,Inflammation ,B cells ,Viral Vaccines ,Antigens, CD20 ,medicine.disease ,Immunity, Humoral ,Tumor Necrosis Factor Receptor Superfamily, Member 7 ,Case-Control Studies ,biology.protein - Abstract
1AbstractDouble negative (DN) (CD19+CD20lowCD27−IgD−) B cells are expanded in patients with autoimmune and infectious diseases; however their role in the humoral immune response remains unclear. Using systematic flow cytometric analyses of peripheral blood B cell subsets, we observed an inflated DN B cell population in patients with variety of active inflammatory conditions: myasthenia gravis, Guillain-Barré syndrome, neuromyelitis optica spectrum disorder, meningitis/encephalitis, and rheumatic disorders. Furthermore, we were able to induce DN B cells in healthy subjects following vaccination against influenza and tick borne encephalitis virus. Transcriptome analysis revealed a gene expression profile in DN B cells that clustered with naïve B cells, memory B cells, and plasmablasts. Immunoglobulin VH transcriptome sequencing and analysis of recombinant antibodies revealed clonal expansion of DN B cells, that were targeted against the vaccine antigen. Our study suggests that DN B cells are expanded in multiple inflammatory neurologic diseases and represent an inducible B cell population that responds to antigenic stimulation, possibly through an extra-follicular maturation pathway.
- Published
- 2020
33. Wnt/PCP-primed intestinal stem cells directly differentiate into enteroendocrine or Paneth cells
- Author
-
Alexandra Aliluev, Fien M. Verhamme, Lena Oppenländer, Heiko Lickert, Maren Büttner, Sophie Tritschler, Johannes Beckers, Christoph Ziegenhain, Wolfgang Enard, Andrea C. Schamberger, Michael Sterr, Ingo Burtscher, Anika Böttcher, Oliver Eickelberg, Fabian J. Theis, Martin Irmler, and Steffen Sass
- Subjects
medicine.anatomical_structure ,Effector ,Paneth cell ,Gene expression ,Wnt signaling pathway ,medicine ,LGR5 ,Priming (immunology) ,Enteroendocrine cell ,Stem cell ,Biology ,digestive system ,Cell biology - Abstract
SUMMARYA detailed understanding of intestinal stem cell (ISC) self-renewal and differentiation is required to better treat chronic intestinal diseases. However, different models of ISC lineage hierarchy1–6 and segregation7–12 are debated. Here we report the identification of Lgr5+ ISCs that express Flattop (Fltp), a Wnt/planar cell polarity (PCP) reporter and effector gene. Lineage labelling revealed that Wnt/PCP-activated Fltp+ ISCs are primed either towards the enteroendocrine or the Paneth cell lineage in vivo. Integration of time-resolved lineage labelling with genome-wide and targeted single-cell gene expression analysis allowed us to delineate the ISC differentiation path into enteroendocrine and Paneth cells at the molecular level. Strikingly, we found that both lineages are directly recruited from ISCs via unipotent transition states, challenging the existence of formerly predicted bi- or multipotent secretory progenitors7–12. Transitory cells that mature into Paneth cells are quiescent and express both stem cell and secretory lineage genes, indicating that these cells are the previously described Lgr5+ labelretaining cells7. Wnt/PCP-activated Lgr5+ ISCs are indistinguishable from Wnt/β-catenin-activated Lgr5+ ISCs based on the expression of stem-cell signature or secretory lineagespecifying genes but possess less self-renewal activity. This suggests that lineage priming and cell-cycle exit is triggered at the post-transcriptional level by polarity cues and a switch from canonical to non-canonical Wnt/PCP signalling. Taken together, we identified the Wnt/PCP pathway as a new niche signal and polarity cue regulating stem cell fate. Active Wnt/PCP signalling represents one of the earliest events in ISC lineage priming towards the Paneth and enteroendocrine cell fate, preceding lateral inhibition and expression of secretory lineagespecifying genes. Thus, our findings provide a better understanding of the niche signals and redefine the mechanisms underlying ISC lineage hierarchy and segregation.
- Published
- 2020
34. A mosaic renal myeloid subtype with T-cell inhibitory and protumoral features is linked to immune escape and survival in clear cell renal cell cancer
- Author
-
Tobias Straub, Elfriede Noessner, Evangelos Kokolakis, Stefan Winter, Elke Schaeffeler, Johannes Beckers, Dorothee Brech, Peter J. Nelson, Florian Buettner, Matthias Schwab, and Martin Irmler
- Subjects
Cell type ,Myeloid ,Angiogenesis ,business.industry ,medicine.medical_treatment ,T cell ,Immunotherapy ,medicine.disease ,Clear cell renal cell carcinoma ,medicine.anatomical_structure ,Immune system ,medicine ,Cancer research ,business ,Clear cell - Abstract
SummaryMononuclear phagocytes moderate tissue repair, immune activation and tolerance. In the renal tubulo-interstitium specialized dendritic cells help maintain homeostasis and protect tubuli from immune injury. Human renal cell carcinoma (RCC) is immunogenic; yet immunotherapies that target T-cell dysfunction show limited clinical efficacy suggesting additional mechanisms of immunoinhibiton. We previously described “enriched-in-renal cell carcinoma” (erc)DCs that are often found in tight contact with T cells which are dysfunctional. Here we describe that ercDCs exhibit a distinct polarization state imparted by tissue-specific signals characteristic for RCC and renal tissue homeostasis. The resulting mosaic transcript signature includes features associated with host defense activity, angiogenesis/invasion and T-cell inhibition. An ercDC-specific profile was predictive for patient survival and suggests potential therapeutic targets for improved immunotherapy.SignificanceImmunotherapies, which re-invigorate T-cell activity, achieve clinical responses in subsets of patients only revealing additional layers of T-cell inhibition. Mononuclear phagocytes can be immunoinhibitory. But, they are highly plastic and repolarization may be possible if key programming molecules can be identified, potentially enabling antitumor responses in tumors refractory to checkpoint blockade. We describe a myeloid cell type with mosaic feature including tumor-promotion and immunoinhibition in human clear cell renal cell carcinoma. Observed tight contacts with T cells may translate into T-cell dysfunction. A high ercDC score in tumor tissue correlates with poor patient survival suggesting ercDCs as targets for therapeutic intervention. Targeting molecules that are identified in the ercDC profile may expand the range of patients effectively treated by immunotherapy.HighlightsBullet points:Renal cell carcinoma (ccRCC) harbors polarized mosaic myeloid cells (ercDCs)ercDCs are found in contact with dysfunctional T cells in ccRCCercDCs express novel immunoinhibitory proteinsHigh ercDC z-score in ccRCC tissue correlates with poor patient survival
- Published
- 2020
- Full Text
- View/download PDF
35. From the roundabout of molecular events to nanomaterial-induced chronic inflammation prediction
- Author
-
Olivier Joubert, Mojca Pušnik, Nick Quirke, Ulla Vogel, Iztok Urbančič, Sabina Halappanavar, Boštjan Kokot, Polona Umek, Pernille Høgh Danielsen, Stane Pajk, Lobaskin, Petra Čotar, López Cb, Patrycja Zawilska, Johannes Beckers, Otmar Schmid, Qiaoxia Zhou, T. Stoeger, Claudia Contini, Rok Podlipec, Ana Krišelj, Mikhail Ivanov, Trine Berthing, Hana Majaron, Jessica Ponti, Martin Irmler, Zahra Doumandji, Janez Štrancar, M. Schneemilch, Alexander P. Lyubartsev, Carola Voss, Zhernovkov, Aleksandar Sebastijanović, and Tilen Koklic
- Subjects
0303 health sciences ,Single exposure ,Mechanism (biology) ,Chemistry ,In silico ,Inflammation ,Lipid metabolism ,02 engineering and technology ,021001 nanoscience & nanotechnology ,In vitro ,3. Good health ,Cell biology ,Proinflammatory cytokine ,03 medical and health sciences ,In vivo ,medicine ,medicine.symptom ,0210 nano-technology ,030304 developmental biology - Abstract
Many chronic diseases manifest in prolonged inflammation and often ignored dysregulated lipid metabolism. When associated with inhalation of nanomaterials, limited information is available on the relevant molecular events and their causal connections. This prevents reliable prediction of outcomes by efficient testing strategies. To unravel how acute nanomaterial exposure leads to chronic conditions, we employed advanced microscopy and omics in vitro and in vivo together with in silico modelling. For selected metal-oxide nanomaterials, we show that lung epithelial cells survive the exposure by excreting internalized nanomaterials and passivating them on the surface, employing elevated lipid synthesis. Macrophages, on the contrary, lose their integrity whilst degrading the passivized bio-nano agglomerates, releasing the nanomaterials, which are taken up again by the epithelial cells. Constant proinflammatory signalling recruits new phagocytes that feed the vicious cycle of events, resulting in a long-lasting response to a single exposure. The proposed mechanism explains the nanomaterial-associated in vivo chronic outcomes and allows its prediction based on in vitro measurements. Similar mechanisms may trigger other chronic diseases affecting millions of lives worldwide.
- Published
- 2020
- Full Text
- View/download PDF
36. Intrauterine smoke exposure deregulates lung function, pulmonary transcriptomes, and in particular insulin-like growth factor (IGF)-1 in a sex-specific manner
- Author
-
Petra Nathan, A. O. Yildirim, Johannes Beckers, Martin Irmler, Katrin Milger, Hagen Scherb, Natalia El-Merhie, Susanne Krauss-Etschmann, Machteld N. Hylkema, Gerrit John-Schuster, Oliver Eickelberg, Sabine Bartel, Stefan Dehmel, Bianca Schaub, Reproductive Origins of Adult Health and Disease (ROAHD), and Groningen Research Institute for Asthma and COPD (GRIAC)
- Subjects
0301 basic medicine ,Male ,Adolescent ,Offspring ,medicine.medical_treatment ,Physiology ,lcsh:Medicine ,Tobacco smoke ,Article ,Transcriptome ,03 medical and health sciences ,Insulin-like growth factor ,Mice ,Pregnancy ,Medicine ,Animals ,Humans ,Insulin-Like Growth Factor I ,Child ,lcsh:Science ,Lung ,Fetus ,Sex Characteristics ,Multidisciplinary ,business.industry ,Gene Expression Profiling ,lcsh:R ,medicine.disease ,Disease Models, Animal ,030104 developmental biology ,medicine.anatomical_structure ,Gene Expression Regulation ,In utero ,Maternal Exposure ,Prenatal Exposure Delayed Effects ,Female ,Tobacco Smoke Pollution ,lcsh:Q ,business - Abstract
Prenatal exposure to tobacco smoke is a significant risk-factor for airway disease development. Furthermore, the high prevalence of pregnant smoking women requires the establishment of strategies for offspring lung protection. Therefore, we here aimed to understand the molecular mechanism of how prenatal smoke exposure affects fetal lung development. We used a mouse model recapitulating clinical findings of prenatally exposed children, where pregnant mice were exposed to smoke until c-section or spontaneous delivery, and offspring weight development and lung function was monitored. Additionally, we investigated pulmonary transcriptome changes in fetal lungs (GD18.5) by mRNA/miRNA arrays, network analyses and qPCR. The results demonstrated that prenatally exposed mice showed intrauterine and postnatal growth retardation, and impaired lung function. 1340 genes and 133 miRNAs were found to be significantly dysregulated by in utero smoke exposure, and we identified Insulin-like growth factor 1 (Igf1) as a top hierarchical node in a network analysis. Moreover, Igf1 mRNA was increased in female murine offspring and in prenatally exposed children. These findings suggest that prenatal smoking is associated with a dysregulation of several genes, including Igf1 in a sex-specific manner. Thus, our results could represent a novel link between smoke exposure, abberant lung development and impaired lung function.
- Published
- 2018
37. Characterization of neuroendocrine tumors in heterozygous mutant MENX rats: a novel model of invasive medullary thyroid carcinoma
- Author
-
Andrea Kügler, Frauke Neff, Natalia S. Pellegata, Sara Molatore, Mercedes Robledo, Federico Roncaroli, Johannes Beckers, Annette Feuchtinger, Martin Irmler, and Tobias Wiedemann
- Subjects
Male ,0301 basic medicine ,congenital, hereditary, and neonatal diseases and abnormalities ,Cancer Research ,medicine.medical_specialty ,Endocrinology, Diabetes and Metabolism ,Adrenal Gland Neoplasms ,Pheochromocytoma ,Biology ,Neuroendocrine tumors ,Rats, Mutant Strains ,Thyroid carcinoma ,03 medical and health sciences ,0302 clinical medicine ,Endocrinology ,Internal medicine ,medicine ,Animals ,Humans ,Pituitary Neoplasms ,Thyroid Neoplasms ,pre-clinical studies ,Thyroid ,Pituitary tumors ,Medullary thyroid cancer ,p27 ,Menx ,Medullary Thyroid Cancer ,P27 Haploinsufficiency ,medicine.disease ,Pituitary adenoma ,Carcinoma, Neuroendocrine ,Rats ,Gene Expression Regulation, Neoplastic ,Disease Models, Animal ,030104 developmental biology ,medicine.anatomical_structure ,Oncology ,Calcitonin ,030220 oncology & carcinogenesis ,Female ,Transcriptome ,Haploinsufficiency ,Cyclin-Dependent Kinase Inhibitor p27 - Abstract
Rats affected by the MENX syndrome spontaneously develop multiple neuroendocrine tumors (NETs) including adrenal, pituitary and thyroid gland neoplasms. MENX was initially reported to be inherited as a recessive trait and affected rats were found to be homozygous for the predisposingCdkn1bmutation encoding p27. We here report that heterozygous MENX-mutant rats (p27+/mut) develop the same spectrum of NETs seen in the homozygous (p27mut/mut) animals but with slower progression. Consequently, p27+/mut rats have a significantly shorter lifespan compared with their wild-type (p27+/+) littermates. In the tumors of p27+/mut rats, the wild-typeCdkn1ballele is neither lost nor silenced, implying that p27 is haploinsufficient for tumor suppression in this model. Transcriptome profiling of rat adrenal (pheochromocytoma) and pituitary tumors having different p27 dosages revealed a tissue-specific, dose-dependent effect of p27 on gene expression. In p27+/mut rats, thyroid neoplasms progress to invasive and metastatic medullary thyroid carcinomas (MTCs) accompanied by increased calcitonin levels, as in humans. Comparison of expression signatures of late-stage vs early-stage MTCs from p27+/mut rats identified genes potentially involved in tumor aggressiveness. The expression of a subset of these genes was evaluated in human MTCs and found to be associated with aggressive RET-M918T-positive tumors. Altogether, p27 haploinsufficiency in MENX rats uncovered a novel, representative model of invasive and metastatic MTC exploitable for translational studies of this often aggressive and incurable cancer.
- Published
- 2018
38. Animal models of obesity and diabetes mellitus
- Author
-
Michael Ristow, Mostafa Bakhti, Peter Huypens, Timo D. Müller, Mark L. Heiman, Susanna M. Hofmann, Alan D. Cherrington, Heiko Lickert, Martin Hrabé de Angelis, Stephen C. Woods, Annette Schürmann, Johannes Beckers, Mary Courtney Moore, Peter J. Havel, Christoffer Clemmensen, Simone Renner, Eckhard Wolf, Martin Klingenspor, Matthias H. Tschöp, and Maximilian Kleinert
- Subjects
0301 basic medicine ,medicine.medical_specialty ,Swine ,Endocrinology, Diabetes and Metabolism ,Clinical Sciences ,Population ,Type 2 diabetes ,Overweight ,Risk Assessment ,Sensitivity and Specificity ,Body Mass Index ,Endocrinology & Metabolism ,Mice ,03 medical and health sciences ,Dogs ,Endocrinology ,SAFER ,Diabetes mellitus ,Diabetes Mellitus ,medicine ,Animals ,Humans ,Obesity ,Intensive care medicine ,education ,education.field_of_study ,Type 1 diabetes ,Animal ,business.industry ,Fishes ,Type 2 Diabetes Mellitus ,Haplorhini ,medicine.disease ,Rats ,Disease Models, Animal ,030104 developmental biology ,Diabetes Mellitus, Type 2 ,Disease Models ,medicine.symptom ,business ,Risk assessment ,Type 2 - Abstract
More than one-third of the worldwide population is overweight or obese and therefore at risk of developing type 2 diabetes mellitus. In order to mitigate this pandemic, safer and more potent therapeutics are urgently required. This necessitates the continued use of animal models to discover, validate and optimize novel therapeutics for their safe use in humans. In order to improve the transition from bench to bedside, researchers must not only carefully select the appropriate model but also draw the right conclusions. In this Review, we consolidate the key information on the currently available animal models of obesity and diabetes and highlight the advantages, limitations and important caveats of each of these models.
- Published
- 2018
39. Exercise prevents fatty liver by modifying the compensatory response of mitochondrial metabolism to excess substrate availability
- Author
-
Rainer Lehmann, Laxmikanth Kollipara, Andreas Peter, Hans-Ulrich Häring, Miriam Hoene, Andreas L. Birkenfeld, Martin Irmler, Albert Sickmann, Christoph Hoffmann, Chunxiu Hu, Johannes Beckers, Guowang Xu, Martin Hrabě de Angelis, Lisa Kappler, Daniel Bleher, and Cora Weigert
- Subjects
Male ,Proteomics ,medicine.medical_specialty ,MAFLD ,Mitochondrion ,Mice ,Insulin resistance ,Endurance training ,Physical Conditioning, Animal ,Internal medicine ,medicine ,Animals ,Mitochondrial supercomplexes ,Exercise ,Molecular Biology ,Chemistry ,Fatty liver ,Skeletal muscle ,Lipid metabolism ,Cell Biology ,Metabolism ,medicine.disease ,RC31-1245 ,Mafld ,Acetyl-coa ,Lipidomics ,Mitochondrial Supercomplexes ,ddc ,Mitochondria ,Fatty Liver ,Mice, Inbred C57BL ,Endocrinology ,medicine.anatomical_structure ,Acetyl-CoA ,Lipogenesis ,Original Article - Abstract
Objective Liver mitochondria adapt to high-calorie intake. We investigated how exercise alters the early compensatory response of mitochondria, thus preventing fatty liver disease as a long-term consequence of overnutrition. Methods We compared the effects of a steatogenic high-energy diet (HED) for six weeks on mitochondrial metabolism of sedentary and treadmill-trained C57BL/6N mice. We applied multi-OMICs analyses to study the alterations in the proteome, transcriptome, and lipids in isolated mitochondria of liver and skeletal muscle as well as in whole tissue and examined the functional consequences by high-resolution respirometry. Results HED increased the respiratory capacity of isolated liver mitochondria, both in sedentary and in trained mice. However, proteomics analysis of the mitochondria and transcriptomics indicated that training modified the adaptation of the hepatic metabolism to HED on the level of respiratory complex I, glucose oxidation, pyruvate and acetyl-CoA metabolism, and lipogenesis. Training also counteracted the HED-induced glucose intolerance, the increase in fasting insulin, and in liver fat by lowering diacylglycerol species and c-Jun N-terminal kinase (JNK) phosphorylation in the livers of trained HED-fed mice, two mechanisms that can reverse hepatic insulin resistance. In skeletal muscle, the combination of HED and training improved the oxidative capacity to a greater extent than training alone by increasing respiration of isolated mitochondria and total mitochondrial protein content. Conclusion We provide a comprehensive insight into the early adaptations of mitochondria in the liver and skeletal muscle to HED and endurance training. Our results suggest that exercise disconnects the HED-induced increase in mitochondrial substrate oxidation from pyruvate and acetyl-CoA-driven lipid synthesis. This could contribute to the prevention of deleterious long-term effects of high fat and sugar intake on hepatic mitochondrial function and insulin sensitivity., Highlights • High-energy diet promotes mitochondrial respiration in liver independent of training. • High-energy diet combined with training disconnects substrate oxidation from lipid synthesis. • High-energy diet combined with training reduces complex I formation in the liver. • Trained skeletal muscle unburdens the liver from substrate overload. • Comprehensive resource of mitochondrial adaptations to high-energy diet and training.
- Published
- 2021
40. Publisher Correction: Epithelial cell plasticity drives endoderm formation during gastrulation
- Author
-
Maren Büttner, Ingo Burtscher, Katharina Scheibner, Heiko Lickert, Gunnar Schotta, Martin Irmler, Michael Sterr, Anika Böttcher, Ansarullah, Fabian J. Theis, Silvia Schirge, Filippo M. Cernilogar, Dapeng Yang, and Johannes Beckers
- Subjects
Gastrulation ,medicine.anatomical_structure ,Endoderm formation ,Embryology ,medicine ,Cell Biology ,Cell lineage ,Epithelial–mesenchymal transition ,Plasticity ,Biology ,Embryonic stem cell ,Epithelium ,Cell biology - Published
- 2021
41. Modeling coexistence of oscillation and Delta/Notch-mediated lateral inhibition in pancreas development and neurogenesis
- Author
-
Elida Schneltzer, Johannes Beckers, Hendrik B. Tiedemann, Gerhard K. H. Przemeck, and Martin Hrabě de Angelis
- Subjects
0301 basic medicine ,Statistics and Probability ,endocrine system ,medicine.medical_specialty ,Neurogenesis ,Gene regulatory network ,Endocrine System ,Nerve Tissue Proteins ,Biology ,Models, Biological ,General Biochemistry, Genetics and Molecular Biology ,Mice ,03 medical and health sciences ,Lateral inhibition ,Oscillometry ,Internal medicine ,Gene expression ,Basic Helix-Loop-Helix Transcription Factors ,medicine ,Animals ,Cell Lineage ,HES1 ,Pancreas ,Body Patterning ,Progenitor ,Feedback, Physiological ,Pancreatic duct ,Receptors, Notch ,General Immunology and Microbiology ,Applied Mathematics ,Gene Expression Regulation, Developmental ,General Medicine ,Cell biology ,030104 developmental biology ,medicine.anatomical_structure ,Endocrinology ,Modeling and Simulation ,Transcription Factor HES-1 ,General Agricultural and Biological Sciences ,Neurog3 ,Pancreatogenesis ,Cycling Gene Expression ,Endocrine Progenitor ,Simulation - Abstract
During pancreas development, Neurog3 positive endocrine progenitors are specified by Delta/Notch (D/N) mediated lateral inhibition in the growing ducts. During neurogenesis, genes that determine the transition from the proneural state to neuronal or glial lineages are oscillating before their expression is sustained. Although the basic gene regulatory network is very similar, cycling gene expression in pancreatic development was not investigated yet, and previous simulations of lateral inhibition in pancreas development excluded by design the possibility of oscillations. To explore this possibility, we developed a dynamic model of a growing duct that results in an oscillatory phase before the determination of endocrine progenitors by lateral inhibition. The basic network (D/N + Hes1 + Neurog3) shows scattered, stable Neurog3 expression after displaying transient expression. Furthermore, we included the Hes1 negative feedback as previously discussed in neurogenesis and show the consequences for Neurog3 expression in pancreatic duct development. Interestingly, a weakened HES1 action on the Hes1 promoter allows the coexistence of stable patterning and oscillations. In conclusion, cycling gene expression and lateral inhibition are not mutually exclusive. In this way, we argue for a unified mode of D/N mediated lateral inhibition in neurogenic and pancreatic progenitor specification.
- Published
- 2017
42. A history of obesity leaves an inflammatory fingerprint in liver and adipose tissue
- Author
-
Martin Irmler, Carola W. Meyer, I P Fischer, M. H. Tschöp, M. Hrabě de Angelis, Frauke Neff, Johannes Beckers, Siegfried Ussar, Stephan Sachs, and Susanna M. Hofmann
- Subjects
Male ,0301 basic medicine ,medicine.medical_specialty ,Endocrinology, Diabetes and Metabolism ,medicine.medical_treatment ,Medicine (miscellaneous) ,Adipose tissue ,Inflammation ,Proinflammatory cytokine ,Mice ,03 medical and health sciences ,Weight loss ,Internal medicine ,Weight Loss ,medicine ,Animals ,Obesity ,Caloric Restriction ,Nutrition and Dietetics ,business.industry ,Insulin ,medicine.disease ,Fatty Liver ,Mice, Inbred C57BL ,030104 developmental biology ,Endocrinology ,Adipose Tissue ,Liver ,Original Article ,medicine.symptom ,Steatosis ,Metabolic syndrome ,business ,Weight gain ,Biomarkers - Abstract
Background/Objectives: Dieting is a popular yet often ineffective way to lower body weight, as the majority of people regain most of their pre-dieting weights in a relatively short time. The underlying molecular mechanisms driving weight regain and the increased risk for metabolic disease are still incompletely understood. Here we investigate the molecular alterations inherited from a history of obesity. Methods: In our model, male high-fat diet (HFD)-fed obese C57BL/6J mice were switched to a low caloric chow diet, resulting in a decline of body weight to that of lean mice. We measured body composition, as well as metrics of glucose, insulin and lipid homeostasis. This was accompanied by histological and gene expression analysis of adipose tissue and liver to assess adipose tissue inflammation and hepatosteatosis. Moreover, acute hypothalamic response to (re-) exposure to HFD was assessed by qPCR. Results & Conclusions: Within 7 weeks after diet switch, most obesity-associated phenotypes, such as body mass, glucose intolerance and blood metabolite levels were reversed. However, hepatic inflammation, hepatic steatosis as well as hypertrophy and inflammation of perigonadal, but not subcutaneous, adipocytes persisted in formerly obese mice. Transcriptional profiling of liver and perigonadal fat revealed an upregulation of pathways associated with immune function and cellularity. Thus, we show that weight reduction leaves signs of inflammation in liver and perigonadal fat, indicating that persisting proinflammatory signals in liver and adipose tissue could contribute to an increased risk of formerly obese subjects to develop the metabolic syndrome upon recurring weight gain.
- Published
- 2017
43. Pulmonary CCR2+CD4+T cells are immune regulatory and attenuate lung fibrosis development
- Author
-
Johannes Beckers, Oliver Eickelberg, Susanne Krauss-Etschmann, Jürgen Behr, Frank Reichenberger, Michael Mayinger, Mareike Lehmann, Alla Skapenko, Katrin Milger, Eva Brudy, Yingyan Yu, Melanie Königshoff, and Martin Irmler
- Subjects
0301 basic medicine ,Pulmonary and Respiratory Medicine ,Adoptive cell transfer ,business.industry ,animal diseases ,FOXP3 ,hemic and immune systems ,medicine.disease ,03 medical and health sciences ,Idiopathic pulmonary fibrosis ,Interleukin 21 ,030104 developmental biology ,0302 clinical medicine ,Immune system ,Fibrosis ,parasitic diseases ,Pulmonary fibrosis ,Immunology ,medicine ,IL-2 receptor ,business ,030215 immunology - Abstract
Background Animal models have suggested that CCR2-dependent signalling contributes to the pathogenesis of pulmonary fibrosis, but global blockade of CCL2 failed to improve the clinical course of patients with lung fibrosis. However, as levels of CCR2 + CD4 + T cells in paediatric lung fibrosis had previously been found to be increased, correlating with clinical symptoms, we hypothesised that distinct CCR2 + cell populations might either increase or decrease disease pathogenesis depending on their subtype. Objective To investigate the role of CCR2 + CD4 + T cells in experimental lung fibrosis and in patients with idiopathic pulmonary fibrosis and other fibrosis. Methods Pulmonary CCR2 + CD4 + T cells were analysed using flow cytometry and mRNA profiling, followed by in silico pathway analysis, in vitro assays and adoptive transfer experiments. Results Frequencies of CCR2 + CD4 + T cells were increased in experimental fibrosis—specifically the CD62L - CD44 + effector memory T cell phenotype, displaying a distinct chemokine receptor profile. mRNA profiling of isolated CCR2 + CD4 + T cells from fibrotic lungs suggested immune regulatory functions, a finding that was confirmed in vitro using suppressor assays. Importantly, adoptive transfer of CCR2 + CD4 + T cells attenuated fibrosis development. The results were partly corroborated in patients with lung fibrosis, by showing higher percentages of Foxp3 + CD25 + cells within bronchoalveolar lavage fluid CCR2 + CD4 + T cells as compared with CCR2 - CD4 + T cells. Conclusion Pulmonary CCR2 + CD4 + T cells are immunosuppressive, and could attenuate lung inflammation and fibrosis. Therapeutic strategies completely abrogating CCR2-dependent signalling will therefore also eliminate cell populations with protective roles in fibrotic lung disease. This emphasises the need for a detailed understanding of the functions of immune cell subsets in fibrotic lung disease.
- Published
- 2017
44. Extensive phenotypic characterization of a new transgenic mouse reveals pleiotropic perturbations in physiology due to mesenchymal hGH minigene expression
- Author
-
Martin Hrabé de Angelis, Eckhard Wolf, Niki Karagianni, Julia Calzada-Wack, Birgit Rathkolb, Maria Armaka, Manolis Roulis, Helmut Fuchs, Aimilios Kaklamanos, George Kollias, Marion Horsch, Jan Rozman, Thure Adler, Moya Wu, Laura Brachthäuser, Johannes Beckers, Valerie Gailus-Durner, and Frauke Neff
- Subjects
Male ,0301 basic medicine ,Genetically modified mouse ,medicine.medical_specialty ,Transgene ,Science ,Gene Expression ,Mice, Transgenic ,Collagen Type VI ,Biology ,Article ,Mice ,03 medical and health sciences ,Internal medicine ,medicine ,Animals ,Humans ,Glucose homeostasis ,Transgenes ,Promoter Regions, Genetic ,Regulation of gene expression ,Multidisciplinary ,Human Growth Hormone ,Tumor Necrosis Factor-alpha ,Mesenchymal stem cell ,TRADD ,Phenotype ,Glucose ,030104 developmental biology ,Endocrinology ,Gene Expression Regulation ,Receptors, Tumor Necrosis Factor, Type I ,Medicine ,Female ,Signal Transduction ,Minigene - Abstract
The human growth hormone (hGH) minigene used for transgene stabilization in mice has been recently identified to be locally expressed in the tissues where transgenes are active and associated with phenotypic alterations. Here we extend these findings by analyzing the effect of the hGH minigene in TgC6hp55 transgenic mice which express the human TNFR1 under the control of the mesenchymal cell-specific CollagenVI promoter. These mice displayed a fully penetrant phenotype characterized by growth enhancement accompanied by perturbations in metabolic, skeletal, histological and other physiological parameters. Notably, this phenotype was independent of TNF-TNFR1 signaling since the genetic ablation of either Tnf or Tradd did not rescue the phenotype. Further analyses showed that the hGH minigene was expressed in several tissues, also leading to increased hGH protein levels in the serum. Pharmacological blockade of GH signaling prevented the development of the phenotype. Our results indicate that the unplanned expression of the hGH minigene in CollagenVI expressing mesenchymal cells can lead through local and/or systemic mechanisms to enhanced somatic growth followed by a plethora of primary and/or secondary effects such as hyperphagia, hypermetabolism, disturbed glucose homeostasis, altered hematological parameters, increased bone formation and lipid accumulation in metabolically critical tissues.
- Published
- 2017
45. Inhibition of LTβR signalling activates WNT-induced regeneration in lung
- Author
-
Zeynep Ertüz, Giorgi Beroshvili, Dominik Pfister, Adrien Guillot, Oliver Eickelberg, Mathias Heikenwalder, Eric Goffin, Reinoud Gosens, Gerald Burgstaller, Danijela Heide, Maximilian Strunz, Fabian J. Theis, Mareike Lehmann, Martin A. Lopez, Michael Boutros, Aicha Jeridi, Darcy E. Wagner, Yan Hu, Marlene Kohlhepp, Tracy O'Connor, Lore Becker, Frank Tacke, Thomas M. Conlon, Meshal Ansari, Indrabahadur Singh, Christoph Mayr, Martin Hrabé de Angelis, Tobias Stoeger, Emmanuel Dejardin, Hani N. Alsafadi, Melanie Königshoff, Sandra Prokosch, Chiara Ciminieri, Michael Dudek, Bernard Pirotte, Herbert B. Schiller, Johannes Beckers, Maja C. Funk, Percy A. Knolle, Martin Irmler, Stijn E. Verleden, Michael Lindner, Gizem Gunes, Rita Costa, Jenny Hetzer, Gerrit John-Schuster, Jakob Janzen, Ali Önder Yildirim, Groningen Research Institute for Asthma and COPD (GRIAC), Molecular Pharmacology, and Nanomedicine & Drug Targeting
- Subjects
0301 basic medicine ,Aging ,Apoptosis ,Adaptive Immunity ,Article ,03 medical and health sciences ,Mice ,Pulmonary Disease, Chronic Obstructive ,0302 clinical medicine ,Fibrosis ,Lymphotoxin beta Receptor ,Smoke ,medicine ,Animals ,Humans ,Regeneration ,Progenitor cell ,Lung ,beta Catenin ,Emphysema ,Multidisciplinary ,Innate immune system ,business.industry ,Regeneration (biology) ,Stem Cells ,Wnt signaling pathway ,NF-kappa B ,Acquired immune system ,medicine.disease ,Epithelium ,Immunity, Innate ,3. Good health ,Mice, Inbred C57BL ,Wnt Proteins ,030104 developmental biology ,Lymphotoxin ,medicine.anatomical_structure ,030228 respiratory system ,Alveolar Epithelial Cells ,Cancer research ,Female ,business ,Engineering sciences. Technology ,Signal Transduction - Abstract
Blockade of lymphotoxin beta-receptor (LT beta R) signalling restores WNT signalling and epithelial repair in a model of chronic obstructive pulmonary disease. Lymphotoxin beta-receptor (LT beta R) signalling promotes lymphoid neogenesis and the development of tertiary lymphoid structures(1,2), which are associated with severe chronic inflammatory diseases that span several organ systems(3-6). How LT beta R signalling drives chronic tissue damage particularly in the lung, the mechanism(s) that regulate this process, and whether LT beta R blockade might be of therapeutic value have remained unclear. Here we demonstrate increased expression of LT beta R ligands in adaptive and innate immune cells, enhanced non-canonical NF-kappa B signalling, and enriched LT beta R target gene expression in lung epithelial cells from patients with smoking-associated chronic obstructive pulmonary disease (COPD) and from mice chronically exposed to cigarette smoke. Therapeutic inhibition of LT beta R signalling in young and aged mice disrupted smoking-related inducible bronchus-associated lymphoid tissue, induced regeneration of lung tissue, and reverted airway fibrosis and systemic muscle wasting. Mechanistically, blockade of LT beta R signalling dampened epithelial non-canonical activation of NF-kappa B, reduced TGF beta signalling in airways, and induced regeneration by preventing epithelial cell death and activating WNT/beta-catenin signalling in alveolar epithelial progenitor cells. These findings suggest that inhibition of LT beta R signalling represents a viable therapeutic option that combines prevention of tertiary lymphoid structures(1) and inhibition of apoptosis with tissue-regenerative strategies.
- Published
- 2019
46. Murine tissue factor disulfide mutation causes a bleeding phenotype with sex specific organ pathology and lethality
- Author
-
Wolfram Ruf, Johannes Beckers, Marion Horsch, Felix C. Tanner, Tanja Klein-Rodewald, Thomas F. Lüscher, Andrea S. Rothmeier, Frauke Neff, Birgit Rathkolb, Martin Hrabě de Angelis, Simon F. Stämpfli, Anja Schrewe, Helmut Fuchs, Susanna H. M. Sluka, Alexander Akhmedov, Paula Grest, Valerie Gailus-Durner, Adrián Sanz-Moreno, Giovanni G. Camici, Eckhard Wolf, and University of Zurich
- Subjects
Male ,medicine.medical_specialty ,Offspring ,610 Medicine & health ,Hemorrhage ,030204 cardiovascular system & hematology ,Biology ,medicine.disease_cause ,Article ,Thromboplastin ,11459 Center for Molecular Cardiology ,03 medical and health sciences ,Tissue factor ,Arterial Thrombosis ,Blood Coagulation and Fibrinolysis ,Disorders of Coagulation and Fibrinolysis ,Mice ,0302 clinical medicine ,In vivo ,Pregnancy ,Internal medicine ,medicine ,Extracellular ,Animals ,Disulfides ,Mutation ,Hematology ,Phenotype ,In vitro ,Endocrinology ,10036 Medical Clinic ,10076 Center for Integrative Human Physiology ,Hemostasis ,10209 Clinic for Cardiology ,Female ,030215 immunology - Abstract
Tissue factor is highly expressed in sub-endothelial tissue. The extracellular allosteric disulfide bond Cys186-Cys209 of human tissue factor shows high evolutionary conservation and in vitro evidence suggests that it significantly contributes to tissue factor procoagulant activity. To investigate the role of this allosteric disulfide bond in vivo, we generated a C213G mutant tissue factor mouse by replacing Cys213 of the corresponding disulfide Cys190-Cys213 in murine tissue factor. A bleeding phenotype was prominent in homozygous C213G tissue factor mice. Pre-natal lethality of 1/3rd of homozygous offspring was observed between E9.5 and E14.5 associated with placental hemorrhages. After birth, homozygous mice suffered from bleedings in different organs and reduced survival. Homozygous C213G tissue factor male mice showed higher incidence of lung bleedings and lower survival rates than females. In both sexes, C213G mutation evoked a reduced protein expression (about 10-fold) and severely reduced pro-coagulant activity (about 1000-fold). Protein glycosylation was impaired and cell membrane exposure decreased in macrophages in vivo. Single housing of homozygous C213G tissue factor males reduced the occurrence of severe bleeding and significantly improved survival, suggesting that inter-male aggressiveness might significantly account for the sex differences. These experiments show that the tissue factor allosteric disulfide bond is of crucial importance for normal in vivo expression, post-translational processing and activity of murine tissue factor. Although C213G tissue factor mice do not display the severe embryonic lethality of tissue factor knock-out mice, their postnatal bleeding phenotype emphasizes the importance of fully functional tissue factor for hemostasis.
- Published
- 2019
47. The Notch ligand DNER regulates macrophage IFNγ release in chronic obstructive pulmonary disease
- Author
-
Oliver Eickelberg, Carolina Ballester‐López, Isis E. Fernandez, Zeynep Ertüz, Martin Irmler, Ali Önder Yildirim, Thomas M. Conlon, Stijn E. Verleden, Johannes Beckers, and Flavia R. Greiffo
- Subjects
0301 basic medicine ,Research paper ,NF-KAPPA-B ,Fluorescent Antibody Technique ,Gene Expression ,EMPHYSEMA ,Research & Experimental Medicine ,Monocytes ,ACTIVATION ,Mice ,Pulmonary Disease, Chronic Obstructive ,0302 clinical medicine ,Macrophage ,ALVEOLAR MACROPHAGES ,Lung ,Mice, Knockout ,COPD ,Receptors, Notch ,Cigarette smoke ,General Medicine ,medicine.anatomical_structure ,DNER ,Medicine, Research & Experimental ,030220 oncology & carcinogenesis ,Notch signalling ,SIGNALING PATHWAY ,medicine.symptom ,Life Sciences & Biomedicine ,Signal Transduction ,EXPRESSION ,DEFENSE ,Notch signaling pathway ,Inflammation ,Nerve Tissue Proteins ,Receptors, Cell Surface ,IFN gamma ,Models, Biological ,General Biochemistry, Genetics and Molecular Biology ,NFkB ,MECHANISMS ,Immunophenotyping ,03 medical and health sciences ,Interferon-gamma ,Medicine, General & Internal ,Immunity ,General & Internal Medicine ,medicine ,Animals ,Humans ,Secretion ,Science & Technology ,INTERFERON-GAMMA ,RECEPTOR ,business.industry ,Microarray analysis techniques ,Macrophages ,Macrophage Activation ,medicine.disease ,respiratory tract diseases ,Disease Models, Animal ,030104 developmental biology ,Copd ,Dner ,Notch Signalling ,Ifn Gamma ,Cigarette Smoke ,Nfkb ,Cancer research ,Human medicine ,business ,Biomarkers - Abstract
BACKGROUND: Chronic Obstructive Pulmonary Disease (COPD) is the third leading cause of death worldwide with no curative therapy. A non-canonical Notch ligand, DNER, has been recently identified in GWAS to associate with COPD severity, but its function and contribution to COPD is unknown. METHODS: DNER localisation was assessed in lung tissue from healthy and COPD patients, and cigarette smoke (CS) exposed mice. Microarray analysis was performed on WT and DNER deficient M1 and M2 bone marrow-derived macrophages (BMDM), and gene set enrichment undertaken. WT and DNER deficient mice were exposed to CS or filtered air for 3 day and 2 months to assess IFNγ-expressing macrophages and emphysema development. Notch and NFKB active subunits were quantified in WT and DNER deficient LPS-treated and untreated BMDM. FINDINGS: Immunofluorescence staining revealed DNER localised to macrophages in lung tissue from COPD patients and mice. Human and murine macrophages showed enhanced DNER expression in response to inflammation. Interestingly, pro-inflammatory DNER deficient BMDMs exhibited impaired NICD1/NFKB dependent IFNγ signalling and reduced nuclear NICD1/NFKB translocation. Furthermore, decreased IFNγ production and Notch1 activation in recruited macrophages from CS exposed DNER deficient mice were observed, protecting against emphysema and lung dysfunction. INTERPRETATION: DNER is a novel protein induced in COPD patients and 6 months CS-exposed mice that regulates IFNγ secretion via non-canonical Notch in pro-inflammatory recruited macrophages. These results provide a new pathway involved in COPD immunity that could contribute to the discovery of innovative therapeutic targets. FUNDING: This work was supported from the Helmholtz Alliance 'Aging and Metabolic Programming, AMPro'. ispartof: EBIOMEDICINE vol:43 pages:562-575 ispartof: location:Netherlands status: published
- Published
- 2019
48. Publisher Correction: Inhibition of LTβR signalling activates WNT-induced regeneration in lung
- Author
-
Dominik Pfister, Adrien Guillot, Frank Tacke, Zeynep Ertüz, Thomas M. Conlon, Meshal Ansari, Gizem Gunes, Indrabahadur Singh, Ilias Angelidis, Maja C. Funk, Darcy E. Wagner, Gerrit John-Schuster, Emmanuel Dejardin, Michael Lindner, Mareike Lehmann, Jenny Hetzer, Bernard Pirotte, Jakob Janzen, Martin Irmler, Tracy O'Connor, Lore Becker, Mathias Heikenwalder, Giorgi Beroshvili, Yan Hu, Michael Boutros, Johannes Beckers, Ali Önder Yildirim, Reinoud Gosens, Fabian J. Theis, Danijela Heide, Maximilian Strunz, Stijn E. Verleden, Melanie Königshoff, Martin Hrabé de Angelis, Gerald Burgstaller, Eric Goffin, Tobias Stoeger, Martin A. Lopez, Hani N. Alsafadi, Aicha Jeridi, Marlene Kohlhepp, Chiara Ciminieri, Percy A. Knolle, Christoph Mayr, Oliver Eickelberg, Michael Dudek, Sandra Prokosch, Herbert B. Schiller, and Rita Costa
- Subjects
Cellular immunity ,Multidisciplinary ,Signalling ,Lung ,medicine.anatomical_structure ,business.industry ,Regeneration (biology) ,medicine ,Wnt signaling pathway ,business ,Cell biology - Abstract
A Correction to this paper has been published: https://doi.org/10.1038/s41586-020-03087-6
- Published
- 2021
49. Dynamic landscape of pancreatic carcinogenesis reveals early molecular networks of malignancy
- Author
-
Susanne Raulefs, Jörg Kleeff, Helmut Friess, Irene Esposito, Mert Erkan, Christoph W. Michalski, Ziying Jian, Martin Irmler, Nataliya Valkovskaya, Susanne Roth, Jing Cao, Thilo Hackert, Bo Kong, Fabian J. Theis, Anna Melissa Schlitter, Philipp Bruns, Andreas Gewies, Ligong Chang, Johannes Beckers, Nora Behler, Sina Fritzsche, Ivonne Regel, Nikola S. Mueller, and Jürgen Ruland
- Subjects
0301 basic medicine ,Pathology ,medicine.medical_specialty ,Carcinogenesis ,Pancreas regeneration ,Mice, Transgenic ,Acinar Cells ,Biology ,medicine.disease_cause ,Proto-Oncogene Proteins p21(ras) ,03 medical and health sciences ,Pancreatic cancer ,medicine ,Acinar cell ,Animals ,Humans ,Regeneration ,Gene Regulatory Networks ,Pancreas ,Cell Proliferation ,Gene Expression Profiling ,Regeneration (biology) ,Gastroenterology ,Cancer ,Mesenchymal Stem Cells ,medicine.disease ,Gene Expression Regulation, Neoplastic ,Pancreatic Neoplasms ,Disease Models, Animal ,030104 developmental biology ,Pancreatitis ,Acute Disease ,Disease Progression ,Cancer research ,KRAS ,Precancerous Conditions ,Carcinoma, Pancreatic Ductal - Abstract
ObjectiveThe initial steps of pancreatic regeneration versus carcinogenesis are insufficiently understood. Although a combination of oncogenic Kras and inflammation has been shown to induce malignancy, molecular networks of early carcinogenesis remain poorly defined.DesignWe compared early events during inflammation, regeneration and carcinogenesis on histological and transcriptional levels with a high temporal resolution using a well-established mouse model of pancreatitis and of inflammation-accelerated KrasG12D-driven pancreatic ductal adenocarcinoma. Quantitative expression data were analysed and extensively modelled in silico.ResultsWe defined three distinctive phases—termed inflammation, regeneration and refinement—following induction of moderate acute pancreatitis in wild-type mice. These corresponded to different waves of proliferation of mesenchymal, progenitor-like and acinar cells. Pancreas regeneration required a coordinated transition of proliferation between progenitor-like and acinar cells. In mice harbouring an oncogenic Kras mutation and challenged with pancreatitis, there was an extended inflammatory phase and a parallel, continuous proliferation of mesenchymal, progenitor-like and acinar cells. Analysis of high-resolution transcriptional data from wild-type animals revealed that organ regeneration relied on a complex interaction of a gene network that normally governs acinar cell homeostasis, exocrine specification and intercellular signalling. In mice with oncogenic Kras, a specific carcinogenic signature was found, which was preserved in full-blown mouse pancreas cancer.ConclusionsThese data define a transcriptional signature of early pancreatic carcinogenesis and a molecular network driving formation of preneoplastic lesions, which allows for more targeted biomarker development in order to detect cancer earlier in patients with pancreatitis.
- Published
- 2016
50. Impact of fibroblast growth factor 21 on the secretome of human perivascular preadipocytes and adipocytes: a targeted proteomics approach
- Author
-
Lucia Berti, Stefan Lehr, Sonja Hartwig, Martin Irmler, Bernhard Rädle, Harald Staiger, Martin Hrabě de Angelis, Dorothea Siegel-Axel, Hans-Ulrich Häring, Johannes Beckers, and Hadi Al-Hasani
- Subjects
Proteomics ,medicine.medical_specialty ,FGF21 ,Physiology ,Adipokine ,Adipose tissue ,030209 endocrinology & metabolism ,030204 cardiovascular system & hematology ,Real-Time Polymerase Chain Reaction ,Fibroblast growth factor ,Proinflammatory cytokine ,03 medical and health sciences ,0302 clinical medicine ,Physiology (medical) ,Internal medicine ,Adipocytes ,medicine ,Humans ,Chemerin ,Cells, Cultured ,Inflammation ,biology ,Genome, Human ,Gene Expression Profiling ,Leptin ,General Medicine ,Fibroblast Growth Factors ,Endocrinology ,Adipogenesis ,Radial Artery ,biology.protein ,Biomarkers - Abstract
Perivascular adipose tissue (PVAT) is suggested to impact on vascular cells via humoral factors, possibly contributing to endothelial dysfunction and atherosclerosis.To address whether the hepatokine fibroblast growth factor (FGF) 21 affects the PVAT secretome.Human perivascular (pre)adipocytes were subjected to targeted proteomics and whole-genome gene expression analysis.Preadipocytes, as compared to adipocytes, secreted higher amounts of inflammatory cytokines and chemokines. Adipocytes released higher amounts of adipokines [e.g. adipisin, visfatin, dipeptidyl peptidase 4 (DPP4), leptin; p 0.05, all]. In preadipocytes, omentin 1 release was 1.28-fold increased by FGF-21 (p 0.05). In adipocytes, FGF-21 reduced chemerin release by 5% and enhanced DPP4 release by 1.15-fold (p 0.05, both). FGF-21 altered the expression of four secretory genes in preadipocytes and of 18 in adipocytes (p 0.01, all).The hepatokine FGF-21 exerts secretome-modulating effects in human perivascular (pre)adipocytes establishing a new liver-PVAT-blood vessel axis that possibly contributes to vascular inflammation and atherosclerosis.
- Published
- 2016
Catalog
Discovery Service for Jio Institute Digital Library
For full access to our library's resources, please sign in.