21 results on '"Hauke M"'
Search Results
2. The influence of firing parameters on the formation of nitride phases in nitride bonded silicon carbides
- Author
-
Kehren, J.T., Steffen, T., Hauke, M., Linden, C., Dannert, C., and Krause, O.
- Published
- 2023
- Full Text
- View/download PDF
3. Early REperfusion therapy with intravenous alteplase for recovery of VISION in acute central retinal artery occlusion (REVISION): Study protocol of a phase III trial.
- Author
-
Poli, Sven, Grohmann, Carsten, Wenzel, Daniel A, Poli, Khouloud, Tünnerhoff, Johannes, Nedelmann, Max, Fiehler, Jens, Burghaus, Ina, Lehmann, Monika, Glauch, Monika, Schadwinkel, Hauke M, Kalmbach, Pia, Zeller, Julia, Peters, Tobias, Eschenfelder, Christoph, Agostini, Hansjürgen, Campbell, Bruce CV, Fischer, M Dominik, Sykora, Marek, and Mac Grory, Brian
- Abstract
Rationale: Meta-analyses of case series of non-arteritic central retinal artery occlusion (CRAO) indicate beneficial effects of intravenous thrombolysis when initiated early after symptom onset. Randomized data are lacking to address this question. Aims: The REperfusion therapy with intravenous alteplase for recovery of VISION in acute central retinal artery occlusion (REVISION) investigates intravenous alteplase within 4.5 h of monocular vision loss due to acute CRAO. Methods: This study is the randomized (1:1), double-blind, placebo-controlled, multicenter adaptive phase III trial. Study outcomes: Primary outcome is functional recovery to normal or mildly impaired vision in the affected eye defined as best-corrected visual acuity of the Logarithm of the Minimum Angle of Resolution of 0.5 or less at 30 days (intention-to-treat analysis). Secondary efficacy outcomes include modified Rankin Score at 90 days and quality of life. Safety outcomes include symptomatic intracranial hemorrhage, major bleeding (International Society on Thrombosis and Haemostasis definition) and mortality. Exploratory analyses of optical coherence tomography/angiography, ultrasound and magnetic resonance imaging (MRI) biomarkers will be conducted. Sample size: Using an adaptive design with interim analysis at 120 patients, up to 422 participants (211 per arm) would be needed for 80% power (one-sided alpha = 0.025) to detect a difference of 15%, assuming functional recovery rates of 10% in the placebo arm and 25% in the alteplase arm. Discussion: By enrolling patients within 4.5 h of CRAO onset, REVISION uses insights from meta-analyses of CRAO case series and randomized thrombolysis trials in acute ischemic stroke. Increased rates of early reperfusion and good neurological outcomes in stroke may translate to CRAO with its similar pathophysiology. Trial registration: ClinicalTrials.gov: NCT04965038; EU Trial Number: 2023-507388-21-00. [ABSTRACT FROM AUTHOR]
- Published
- 2024
- Full Text
- View/download PDF
4. A MHz X-ray diffraction set-up for dynamic compression experiments in the diamond anvil cell
- Author
-
Rachel J. Husband, Cornelius Strohm, Karen Appel, Orianna B. Ball, Richard Briggs, Johannes Buchen, Valerio Cerantola, Stella Chariton, Amy L. Coleman, Hyunchae Cynn, Dana Dattelbaum, Anand Dwivedi, Jon H. Eggert, Lars Ehm, William J. Evans, Konstantin Glazyrin, Alexander F. Goncharov, Heinz Graafsma, Alex Howard, Larissa Huston, Trevor M. Hutchinson, Huijeong Hwang, Sony Jacob, Johannes Kaa, Jaeyong Kim, Minseob Kim, Egor Koemets, Zuzana Konôpková, Falko Langenhorst, Torsten Laurus, Xinyang Li, Jona Mainberger, Hauke Marquardt, Emma E. McBride, Christopher McGuire, James D. McHardy, Malcolm I. McMahon, R. Stewart McWilliams, Alba S. J. Méndez, Anshuman Mondal, Guillaume Morard, Earl F. O'Bannon, Christoph Otzen, Charles M. Pépin, Vitali B. Prakapenka, Clemens Prescher, Thomas R. Preston, Ronald Redmer, Michael Roeper, Carmen Sanchez-Valle, Dean Smith, Raymond F. Smith, Daniel Sneed, Sergio Speziale, Tobias Spitzbart, Stephan Stern, Blake T. Sturtevant, Jolanta Sztuk-Dambietz, Peter Talkovski, Nenad Velisavljevic, Cara Vennari, Zhongyan Wu, Choong-Shik Yoo, Ulf Zastrau, Zsolt Jenei, and Hanns-Peter Liermann
- Subjects
extreme conditions science ,x-ray free-electron lasers ,diamond anvil cells ,dynamic compression ,Nuclear and particle physics. Atomic energy. Radioactivity ,QC770-798 ,Crystallography ,QD901-999 - Abstract
An experimental platform for dynamic diamond anvil cell (dDAC) research has been developed at the High Energy Density (HED) Instrument at the European X-ray Free Electron Laser (European XFEL). Advantage was taken of the high repetition rate of the European XFEL (up to 4.5 MHz) to collect pulse-resolved MHz X-ray diffraction data from samples as they are dynamically compressed at intermediate strain rates (≤103 s−1), where up to 352 diffraction images can be collected from a single pulse train. The set-up employs piezo-driven dDACs capable of compressing samples in ≥340 µs, compatible with the maximum length of the pulse train (550 µs). Results from rapid compression experiments on a wide range of sample systems with different X-ray scattering powers are presented. A maximum compression rate of 87 TPa s−1 was observed during the fast compression of Au, while a strain rate of ∼1100 s−1 was achieved during the rapid compression of N2 at 23 TPa s−1.
- Published
- 2023
- Full Text
- View/download PDF
5. Strong Effect of Stress on the Seismic Signature of the Post‐Stishovite Phase Transition in the Earth's Lower Mantle
- Author
-
Biao Wang, Johannes Buchen, Alba San José Méndez, Alexander Kurnosov, Giacomo Criniti, Hanns‐Peter Liermann, and Hauke Marquardt
- Subjects
Earth's lower mantle ,stishovite to post‐stishovite phase transition ,stress ,bulk modulus ,high pressure experiments ,seismic scatterers ,Geophysics. Cosmic physics ,QC801-809 - Abstract
Abstract The stishovite to post‐stishovite phase transition may modify the scattering of seismic waves by stishovite‐bearing rocks in the Earth's lower mantle. A series of continuous compression experiments on sintered polycrystalline stishovite was performed to study the effect of stress on the phase transition. The experimental results show that the phase transition shifts to lower pressures as the magnitude of deviatoric stress increases. Our results further show that the bulk modulus of sintered polycrystalline stishovite differs from that derived from single crystal measurements and decreases at the phase transition. In cold regions, such as subducted slabs, stresses may accumulate and shift the phase transition to a shallower depth. In hot regions with less stress, such as rising plumes, the phase transition is shifted to a greater depth. In addition, the phase transition may have varying seismic signatures depending on the behavior of the grain boundaries in mantle rocks and the micro‐stresses present in neighboring grains.
- Published
- 2023
- Full Text
- View/download PDF
6. GLUT1-mediated glucose import in B cells is critical for anaplerotic balance and humoral immunity.
- Author
-
Bierling TEH, Gumann A, Ottmann SR, Schulz SR, Weckwerth L, Thomas J, Gessner A, Wichert M, Kuwert F, Rost F, Hauke M, Freudenreich T, Mielenz D, Jäck HM, and Pracht K
- Subjects
- Animals, Mice, Glucose, Glucose Transporter Type 1, Plasma Cells, B-Lymphocytes, Immunity, Humoral
- Abstract
Glucose uptake increases during B cell activation and antibody-secreting cell (ASC) differentiation, but conflicting findings prevent a clear metabolic profile at different stages of B cell activation. Deletion of the glucose transporter type 1 (GLUT1) gene in mature B cells (GLUT1-cKO) results in normal B cell development, but it reduces germinal center B cells and ASCs. GLUT1-cKO mice show decreased antigen-specific antibody titers after vaccination. In vitro, GLUT1-deficient B cells show impaired activation, whereas established plasmablasts abolish glycolysis, relying on mitochondrial activity and fatty acids. Transcriptomics and metabolomics reveal an altered anaplerotic balance in GLUT1-deficient ASCs. Despite attempts to compensate for glucose deprivation by increasing mitochondrial mass and gene expression associated with glycolysis, the tricarboxylic acid cycle, and hexosamine synthesis, GLUT1-deficient ASCs lack the metabolites for energy production and mitochondrial respiration, limiting protein synthesis. We identify GLUT1 as a critical metabolic player defining the germinal center response and humoral immunity., Competing Interests: Declaration of interests The authors declare no competing interests., (Copyright © 2024 The Author(s). Published by Elsevier Inc. All rights reserved.)
- Published
- 2024
- Full Text
- View/download PDF
7. Deletion of vascular thromboxane A 2 receptors and its impact on angiotensin II-induced hypertension and atherosclerotic lesion formation in the aorta of Ldlr-deficient mice.
- Author
-
Braun H, Hauke M, Petermann M, Eckenstaler R, Ripperger A, Schwedhelm E, Ludwig-Kraus B, Bernhard Kraus F, Jalal Ahmed Shawon M, Dubourg V, Zernecke A, Schreier B, Gekle M, and Benndorf RA
- Subjects
- Animals, Female, Male, Mice, Angiotensin II toxicity, Aorta, Mice, Inbred C57BL, Mice, Knockout, Atherosclerosis chemically induced, Atherosclerosis genetics, Atherosclerosis pathology, Hypertension chemically induced, Hypertension genetics, Hypertension pathology, Receptors, Thromboxane genetics
- Abstract
The thromboxane A
2 receptor (TP) has been shown to play a role in angiotensin II (Ang II)-mediated hypertension and pathological vascular remodeling. To assess the impact of vascular TP on Ang II-induced hypertension, atherogenesis, and pathological aortic alterations, i.e. aneurysms, we analysed Western-type diet-fed and Ang II-infused TPVSMC KO /Ldlr KO, TPEC KO /Ldlr KO mice and their respective wild-type littermates (TPWT /Ldlr KO). These analyses showed that neither EC- nor VSMC-specific deletion of the TP significantly affected basal or Ang II-induced blood pressure or aortic atherosclerotic lesion area. In contrast, VSMC-specific TP deletion abolished and EC-specific TP deletion surprisingly reduced the ex vivo reactivity of aortic rings to the TP agonist U-46619, whereas VSMC-specific TP knockout also diminished the ex vivo response of aortic rings to Ang II. Furthermore, despite similar systemic blood pressure, there was a trend towards less atherogenesis in the aortic arch and a trend towards fewer pathological aortic alterations in Ang II-treated female TPVSMC KO /Ldlr KO mice. Survival was impaired in male mice after Ang II infusion and tended to be higher in TPVSMC KO /Ldlr KO mice than in TPWT /Ldlr KO littermates. Thus, our data may suggest a deleterious role of the TP expressed in VSMC in the pathogenesis of Ang II-induced aortic atherosclerosis in female mice, and a surprising role of the endothelial TP in TP-mediated aortic contraction. However, future studies are needed to substantiate and further elucidate the role of the vascular TP in the pathogenesis of Ang II-induced hypertension, aortic atherosclerosis and aneurysm formation., Competing Interests: Declaration of Competing Interest The authors declare that they have no known competing financial interests or personal relationships that could have appeared to influence the work reported in this paper., (Copyright © 2023 The Author(s). Published by Elsevier Inc. All rights reserved.)- Published
- 2024
- Full Text
- View/download PDF
8. Identification of miR-128 Target mRNAs That Are Expressed in B Cells Using a Modified Dual Luciferase Vector.
- Author
-
Schreiber S, Daum P, Danzer H, Hauke M, Jäck HM, and Wittmann J
- Subjects
- RNA, Messenger genetics, RNA, Messenger metabolism, Cell Line, B-Lymphocytes metabolism, Luciferases genetics, MicroRNAs metabolism
- Abstract
MicroRNAs (miRNAs) are 21-25 nucleotide long non-coding ribonucleic acids that modulate gene expression by degrading transcripts or inhibiting translation. The miRNA miR-128, originally thought to be brain-specific, was later also found in immune cells. To identify a valuable immune cell model system to modulate endogenous miR-128 amounts and to validate predicted miR-128 target mRNAs in B cells, we first investigated miR-128 expression using Northern blot analysis in several cell lines representing different stages of B cell development. The results showed that only primary brain cells showed significant levels of mature miR-128. To study the function of miR-128 in immune cells, we modified dual luciferase vectors to allow easy transfer of 3' UTR fragments with predicted miR-128 binding sites from widely used single to dual luciferase vectors. Comparison of in silico predicted miR-128-regulated mRNAs in single and dual luciferase constructs yielded similar results, validating the dual luciferase vector for miRNA target analysis. Furthermore, we confirmed miR-128-regulated mRNAs identified in silico and in vivo using the Ago HITS-CLIP technique and known to be expressed in B cells using the dual luciferase assay. In conclusion, this study provides new insights into the expression and function of miR-128 by validating novel target mRNAs expressed in B cells and identifying additional pathways likely controlled by this miRNA in the immune system.
- Published
- 2023
- Full Text
- View/download PDF
9. Innate activation of human neutrophils and neutrophil-like cells by the pro-inflammatory bacterial metabolite ADP-heptose and Helicobacter pylori.
- Author
-
Faass L, Hauke M, Stein SC, and Josenhans C
- Subjects
- Humans, Lipopolysaccharides metabolism, Helicobacter Infections microbiology, Helicobacter pylori, Heptoses metabolism, Neutrophils metabolism
- Abstract
Lipopolysaccharide inner core heptose metabolites, including ADP-heptose, play a substantial role in the activation of cell-autonomous innate immune responses in eukaryotic cells, via the ALPK1-TIFA signaling pathway, as demonstrated for various pathogenic bacteria. The important role of LPS heptose metabolites during Helicobacter pylori infection of the human gastric niche has been demonstrated for gastric epithelial cells and macrophages, while the role of heptose metabolites on human neutrophils has not been investigated. In this study, we aimed to gain a better understanding of the activation potential of bacterial heptose metabolites for human neutrophil cells. To do so, we used pure ADP-heptose and, as a bacterial model, H. pylori, which can transport heptose metabolites into the human host cell via the Cag Type 4 Secretion System (CagT4SS). Main questions were how bacterial heptose metabolites impact on the pro-inflammatory activation, alone and in the bacterial context, and how they influence maturation of human neutrophils. Results of the present study demonstrated that neutrophils respond with high sensitivity to pure heptose metabolites, and that global regulation networks and neutrophil maturation are influenced by heptose exposure. Furthermore, activation of human neutrophils by live H. pylori is strongly impacted by the presence of LPS heptose metabolites and the functionality of its CagT4SS. Similar activities were determined in cell culture neutrophils of different maturation states and in human primary neutrophils. In conclusion, we demonstrated that specific heptose metabolites or bacteria producing heptoses exhibit a strong activity on cell-autonomous innate responses of human neutrophils., (Copyright © 2023 The Authors. Published by Elsevier GmbH.. All rights reserved.)
- Published
- 2023
- Full Text
- View/download PDF
10. Helicobacter pylori Modulates Heptose Metabolite Biosynthesis and Heptose-Dependent Innate Immune Host Cell Activation by Multiple Mechanisms.
- Author
-
Hauke M, Metz F, Rapp J, Faass L, Bats SH, Radziej S, Link H, Eisenreich W, and Josenhans C
- Subjects
- Humans, Lipopolysaccharides metabolism, Heptoses chemistry, Heptoses metabolism, Inflammation, Immunity, Innate, Bacterial Proteins metabolism, Helicobacter pylori genetics, Helicobacter pylori metabolism
- Abstract
Heptose metabolites including ADP-d-glycero-β-d-manno-heptose (ADP-heptose) are involved in bacterial lipopolysaccharide and cell envelope biosynthesis. Recently, heptoses were also identified to have potent proinflammatory activity on human cells as novel microbe-associated molecular patterns. The gastric pathogenic bacterium Helicobacter pylori produces heptose metabolites, which it transports into human cells through its Cag type 4 secretion system. Using H. pylori as a model, we have addressed the question of how proinflammatory ADP-heptose biosynthesis can be regulated by bacteria. We have characterized the interstrain variability and regulation of heptose biosynthesis genes and the modulation of heptose metabolite production by H. pylori, which impact cell-autonomous proinflammatory human cell activation. HldE, a central enzyme of heptose metabolite biosynthesis, showed strong sequence variability between strains and was also variably expressed between strains. Amounts of gene transcripts in the hldE gene cluster displayed intrastrain and interstrain differences, were modulated by host cell contact and the presence of the cag pathogenicity island, and were affected by carbon starvation regulator A (CsrA). We reconstituted four steps of the H. pylori lipopolysaccharide (LPS) heptose biosynthetic pathway in vitro using recombinant purified GmhA, HldE, and GmhB proteins. On the basis of one- and two-dimensional nuclear magnetic resonance (NMR) spectroscopy and mass spectrometry, the structures of major reaction products were identified as β-d-ADP-heptose and β-heptose-1-monophosphate. A proinflammatory heptose-monophosphate variant was also identified for the first time as a novel cell-active product in H. pylori bacteria. Separate purified HldE subdomains and variant HldE allowed us to uncover additional strain variation in generating heptose metabolites. IMPORTANCE Bacterial heptose metabolites, intermediates of lipopolysaccharide (LPS) biosynthesis, are novel microbe-associated molecular patterns (MAMPs) that activate proinflammatory signaling. In the gastric pathogen Helicobacter pylori, heptoses are transferred into host cells by the Cag type IV secretion system, which is also involved in carcinogenesis. Little is known about how H. pylori, which is highly strain variable, regulates heptose biosynthesis and downstream host cell activation. We report here that the regulation of proinflammatory heptose production by H. pylori is strain specific. Heptose gene cluster activity is modulated by the presence of an active cag pathogenicity island ( cag PAI), contact with human cells, and the carbon starvation regulator A. Reconstitution with purified biosynthesis enzymes and purified bacterial lysates allowed us to biochemically characterize heptose pathway products, identifying a heptose-monophosphate variant as a novel proinflammatory metabolite. These findings emphasize that the bacteria use heptose biosynthesis to fine-tune inflammation and also highlight opportunities to mine the heptose biosynthesis pathway as a potential therapeutic target against infection, inflammation, and cancer., Competing Interests: The authors declare no conflict of interest.
- Published
- 2023
- Full Text
- View/download PDF
11. Innate immune activation and modulatory factors of Helicobacter pylori towards phagocytic and nonphagocytic cells.
- Author
-
Faass L, Hauke M, Stein SC, and Josenhans C
- Subjects
- Humans, Neutrophils metabolism, Immunity, Innate, Epithelial Cells, Bacterial Proteins metabolism, Helicobacter pylori
- Abstract
Helicobacter pylori is an intriguing obligate host-associated human pathogen with a specific host interaction biology, which has been shaped by thousands of years of host-pathogen coevolution. Molecular mechanisms of interaction of H. pylori with the local immune cells in the human system are less well defined than epithelial cell interactions, although various myeloid cells, including neutrophils and other phagocytes, are locally present or attracted to the sites of infection and interact with H. pylori. We have recently addressed the question of novel bacterial innate immune stimuli, including bacterial cell envelope metabolites, that can activate and modulate cell responses via the H. pylori Cag type IV secretion system. This review article gives an overview of what is currently known about the interaction modes and mechanisms of H. pylori with diverse human cell types, with a focus on bacterial metabolites and cells of the myeloid lineage including phagocytic and antigen-presenting cells., (Copyright © 2023 Elsevier Ltd. All rights reserved.)
- Published
- 2023
- Full Text
- View/download PDF
12. In vitro kinase assay reveals ADP-heptose-dependent ALPK1 autophosphorylation and altered kinase activity of disease-associated ALPK1 mutants.
- Author
-
García-Weber D, Dangeard AS, Teixeira V, Hauke M, Carreaux A, Josenhans C, and Arrieumerlou C
- Subjects
- Humans, Phosphorylation, Immunity, Innate, NF-kappa B genetics, NF-kappa B metabolism, Heptoses chemistry, Heptoses metabolism, Helicobacter Infections microbiology, Helicobacter pylori metabolism
- Abstract
Alpha-protein kinase 1 (ALPK1) is a pathogen recognition receptor that detects ADP-heptose (ADPH), a lipopolysaccharide biosynthesis intermediate, recently described as a pathogen-associated molecular pattern in Gram-negative bacteria. ADPH binding to ALPK1 activates its kinase domain and triggers TIFA phosphorylation on threonine 9. This leads to the assembly of large TIFA oligomers called TIFAsomes, activation of NF-κB and pro-inflammatory gene expression. Furthermore, mutations in ALPK1 are associated with inflammatory syndromes and cancers. While this kinase is of increasing medical interest, its activity in infectious or non-infectious diseases remains poorly characterized. Here, we use a non-radioactive ALPK1 in vitro kinase assay based on the use of ATPγS and protein thiophosphorylation. We confirm that ALPK1 phosphorylates TIFA T9 and show that T2, T12 and T19 are also weakly phosphorylated by ALPK1. Interestingly, we find that ALPK1 itself is phosphorylated in response to ADPH recognition during Shigella flexneri and Helicobacter pylori infection and that disease-associated ALPK1 mutants exhibit altered kinase activity. In particular, T237M and V1092A mutations associated with ROSAH syndrome and spiradenoma/spiradenocarcinoma respectively, exhibit enhanced ADPH-induced kinase activity and constitutive assembly of TIFAsomes. Altogether, this study provides new insights into the ADPH sensing pathway and disease-associated ALPK1 mutants., (© 2023. The Author(s).)
- Published
- 2023
- Full Text
- View/download PDF
13. Chitinase A, a tightly regulated virulence factor of Salmonella enterica serovar Typhimurium, is actively secreted by a Type 10 Secretion System.
- Author
-
Krone L, Faass L, Hauke M, Josenhans C, and Geiger T
- Subjects
- Humans, Salmonella typhimurium, N-Acetylmuramoyl-L-alanine Amidase genetics, N-Acetylmuramoyl-L-alanine Amidase metabolism, Serogroup, Intestinal Mucosa microbiology, Bacterial Secretion Systems, Bacterial Proteins genetics, Bacterial Proteins metabolism, Gene Expression Regulation, Bacterial, Virulence Factors genetics, Virulence Factors metabolism, Chitinases genetics, Chitinases metabolism
- Abstract
As a facultative intracellular pathogen, Salmonella enterica serovar Typhimurium is one of the leading causes of food-borne diseases in humans. With the ingestion of fecal contaminated food or water, S. Typhimurium reaches the intestine. Here, the pathogen efficiently invades intestinal epithelial cells of the mucosal epithelium by the use of multiple virulence factors. Recently, chitinases have been described as emerging virulence factors of S. Typhimurium that contribute to the attachment and invasion of the intestinal epithelium, prevent immune activation, and modulate the host glycome. Here we find that the deletion of chiA leads to diminished adhesion and invasion of polarized intestinal epithelial cells (IEC) compared to wild-type S. Typhimurium. Interestingly, no apparent impact on interaction was detected when using non-polarized IEC or HeLa epithelial cells. In concordance, we demonstrate that chiA gene and ChiA protein expression was solely induced when bacteria gain contact with polarized IEC. The induction of chiA transcripts needs the specific activity of transcriptional regulator ChiR, which is co-localized with chiA in the chitinase operon. Moreover, we established that after chiA is induced, a major portion of the bacterial population expresses chiA, analyzed by flow cytometry. Once expressed, we found ChiA in the bacterial supernatants using Western blot analyses. ChiA secretion was completely abolished when accessory genes within the chitinase operon encoding for a holin and a peptidoglycan hydrolase were deleted. Holins, peptidoglycan hydrolases, and large extracellular enzymes in close proximity have been described as components of the bacterial holin/peptidoglycan hydrolase-dependent protein secretion system or Type 10 Secretion System. Overall, our results confirm that chitinase A is an important virulence factor, tightly regulated by ChiR, that promotes adhesion and invasion upon contact with polarized IEC and is likely secreted by a Type 10 Secretion System (T10SS)., Competing Interests: The authors have declared that no competing interests exist., (Copyright: © 2023 Krone et al. This is an open access article distributed under the terms of the Creative Commons Attribution License, which permits unrestricted use, distribution, and reproduction in any medium, provided the original author and source are credited.)
- Published
- 2023
- Full Text
- View/download PDF
14. The microRNA processing subunit DGCR8 is required for a T cell-dependent germinal center response.
- Author
-
Daum P, Ottmann SR, Meinzinger J, Schulz SR, Côrte-Real J, Hauke M, Roth E, Schuh W, Mielenz D, Jäck HM, and Pracht K
- Subjects
- Mice, Animals, RNA-Binding Proteins genetics, RNA-Binding Proteins metabolism, T-Lymphocytes metabolism, Germinal Center metabolism, Immunoglobulin G metabolism, MicroRNAs genetics, MicroRNAs metabolism
- Abstract
We have previously shown that the microRNA (miRNA) processor complex consisting of the RNAse Drosha and the DiGeorge Critical Region (DGCR) 8 protein is essential for B cell maturation. To determine whether miRNA processing is required to initiate T cell-mediated antibody responses, we deleted DGCR8 in maturing B2 cells by crossing a mouse with loxP-flanked DGCR8 alleles with a CD23-Cre mouse. As expected, non-immunized mice showed reduced numbers of mature B2 cells and IgG-secreting cells and diminished serum IgG titers. In accordance, germinal centers and antigen-specific IgG-secreting cells were absent in mice immunized with T-dependent antigens. Therefore, DGCR8 is required to mount an efficient T-dependent antibody response. However, DGCR8 deletion in B1 cells was incomplete, resulting in unaltered B1 cell numbers and normal IgM and IgA titers in DGCR8-knock-out mice. Therefore, this mouse model could be used to analyze B1 responses in the absence of functional B2 cells., Competing Interests: The authors declare that the research was conducted in the absence of any commercial or financial relationships that could be construed as a potential conflict of interest., (Copyright © 2022 Daum, Ottmann, Meinzinger, Schulz, Côrte-Real, Hauke, Roth, Schuh, Mielenz, Jäck and Pracht.)
- Published
- 2022
- Full Text
- View/download PDF
15. A current overview of RhoA, RhoB, and RhoC functions in vascular biology and pathology.
- Author
-
Eckenstaler R, Hauke M, and Benndorf RA
- Subjects
- rhoC GTP-Binding Protein metabolism, rho GTP-Binding Proteins genetics, Cell Movement, Biology, rhoB GTP-Binding Protein genetics, rhoB GTP-Binding Protein metabolism, rhoA GTP-Binding Protein genetics
- Abstract
The Rho subfamily members of Rho GTPases, RhoA, RhoB, and RhoC, are key regulators of signal transduction in a variety of cellular processes, including regulation of actomyosin and microtubule dynamics, cell shape, cell adhesion, cell division, cell migration, vesicle/membrane trafficking, and cell proliferation. Traditionally, the focus of research on RhoA/B/C has been on tumor biology, as dysregulation of expression or function of these proteins plays an important role in the pathogenesis of various cancer entities. However, RhoA, RhoB, and RhoC are also important in the context of vascular biology and pathology because they influence endothelial barrier function, vascular smooth muscle contractility and proliferation, vascular function and remodelling as well as angiogenesis. In this context, RhoA/B/C exploit numerous effector molecules to transmit their signals, and their activity is regulated by a variety of guanine nucleotide exchange factors (RhoGEFs) and GTPase-activating proteins (RhoGAPs) that enable precise spatiotemporal activation often in concert with other Rho GTPases. Although their protein structure is very similar, different mechanisms of regulation of gene expression, different localization, and to some extent different interaction with RhoGAPs and RhoGEFs have been observed for RhoA/B/C. In this review, we aim to provide a current overview of the Rho subfamily as regulators of vascular biology and pathology, analyzing database information and existing literature on expression, protein structure, and interaction with effectors and regulatory proteins. In this setting, we will also discuss recent findings on Rho effectors, RhoGEFs, RhoGAPs, as well as guanine nucleotide dissociation inhibitors (RhoGDIs)., Competing Interests: Declaration of Competing Interest The authors declare that they have no known competing financial interests or personal relationships that could have appeared to influence the work reported in this paper., (Copyright © 2022 The Author(s). Published by Elsevier Inc. All rights reserved.)
- Published
- 2022
- Full Text
- View/download PDF
16. Thromboxane A 2 receptor activation via G α13 -RhoA/C-ROCK-LIMK2-dependent signal transduction inhibits angiogenic sprouting of human endothelial cells.
- Author
-
Eckenstaler R, Ripperger A, Hauke M, Braun H, Ergün S, Schwedhelm E, and Benndorf RA
- Subjects
- 15-Hydroxy-11 alpha,9 alpha-(epoxymethano)prosta-5,13-dienoic Acid pharmacology, Humans, Neovascularization, Physiologic, Signal Transduction, Vascular Endothelial Growth Factor A metabolism, rho-Associated Kinases, rhoC GTP-Binding Protein, GTP-Binding Protein alpha Subunits, G12-G13 metabolism, Human Umbilical Vein Endothelial Cells cytology, Human Umbilical Vein Endothelial Cells metabolism, Lim Kinases metabolism, Receptors, Thromboxane A2, Prostaglandin H2 metabolism, rhoA GTP-Binding Protein genetics, rhoA GTP-Binding Protein metabolism
- Abstract
We could previously show that thromboxane A
2 receptor (TP) activation inhibits the angiogenic capacity of human endothelial cells, but the underlying mechanisms remained unclear. Therefore, the aim of this study was to elucidate TP signal transduction pathways relevant to angiogenic sprouting of human endothelial cells. To clarify this matter, we used RNAi-mediated gene silencing as well as pharmacological inhibition of potential TP downstream targets in human umbilical vein endothelial cells (HUVEC) and VEGF-induced angiogenic sprouting of HUVEC spheroids in vitro as a functional read-out. In this experimental set-up, the TP agonist U-46619 completely blocked VEGF-induced angiogenic sprouting of HUVEC spheroids. Moreover, in live-cell analyses TP activation induced endothelial cell contraction, sprout retraction as well as endothelial cell tension and focal adhesion dysregulation of HUVEC. These effects were reversed by pharmacological TP inhibition or TP knockdown. Moreover, we identified a TP-Gα13 -RhoA/C-ROCK-LIMK2-dependent signal transduction pathway to be relevant for U-46619-induced inhibition of VEGF-mediated HUVEC sprouting. In line with these results, U-46619-mediated TP activation potently induced RhoA and RhoC activity in live HUVEC as measured by FRET biosensors. Interestingly, pharmacological inhibition of ROCK and LIMK2 also normalized U-46619-induced endothelial cell tension and focal adhesion dysregulation of HUVEC. In summary, our work reveals mechanisms by which the TP may disturb angiogenic endothelial function in disease states associated with sustained endothelial TP activation., (Copyright © 2022 Elsevier Inc. All rights reserved.)- Published
- 2022
- Full Text
- View/download PDF
17. Active RhoA Exerts an Inhibitory Effect on the Homeostasis and Angiogenic Capacity of Human Endothelial Cells.
- Author
-
Hauke M, Eckenstaler R, Ripperger A, Ender A, Braun H, and Benndorf RA
- Subjects
- Animals, Cell Movement, Homeostasis, Human Umbilical Vein Endothelial Cells metabolism, Humans, Lim Kinases metabolism, Mice, Neovascularization, Pathologic metabolism, rho-Associated Kinases metabolism, rhoA GTP-Binding Protein, Signal Transduction, Vascular Endothelial Growth Factor A metabolism
- Abstract
Background The small GTPase RhoA (Ras homolog gene family, member A) regulates a variety of cellular processes, including cell motility, proliferation, survival, and permeability. In addition, there are reports indicating that RhoA-ROCK (rho associated coiled-coil containing protein kinase) activation is essential for VEGF (vascular endothelial growth factor)-mediated angiogenesis, whereas other work suggests VEGF-antagonistic effects of the RhoA-ROCK axis. Methods and Results To elucidate this issue, we examined human umbilical vein endothelial cells and human coronary artery endothelial cells after stable overexpression (lentiviral transduction) of constitutively active (G14V/Q63L), dominant-negative (T19N), or wild-type RhoA using a series of in vitro angiogenesis assays (proliferation, migration, tube formation, angiogenic sprouting, endothelial cell viability) and a human umbilical vein endothelial cells xenograft assay in immune-incompetent NOD scid gamma mice in vivo. Here, we report that expression of active and wild-type RhoA but not dominant-negative RhoA significantly inhibited endothelial cell proliferation, migration, tube formation, and angiogenic sprouting in vitro. Moreover, active RhoA increased endothelial cell death in vitro and decreased human umbilical vein endothelial cell-related angiogenesis in vivo. Inhibition of RhoA by C3 transferase antagonized the inhibitory effects of RhoA and strongly enhanced VEGF-induced angiogenic sprouting in control-treated cells. In contrast, inhibition of RhoA effectors ROCK1/2 and LIMK1/2 (LIM domain kinase 1/2) did not significantly affect RhoA-related effects, but increased angiogenic sprouting and migration of control-treated cells. In agreement with these data, VEGF did not activate RhoA in human umbilical vein endothelial cells as measured by a Förster resonance energy transfer-based biosensor. Furthermore, global transcriptome and subsequent bioinformatic gene ontology enrichment analyses revealed that constitutively active RhoA induced a differentially expressed gene pattern that was enriched for gene ontology biological process terms associated with mitotic nuclear division, cell proliferation, cell motility, and cell adhesion, which included a significant decrease in VEGFR-2 (vascular endothelial growth factor receptor 2) and NOS3 (nitric oxide synthase 3) expression. Conclusions Our data demonstrate that increased RhoA activity has the potential to trigger endothelial dysfunction and antiangiogenic effects independently of its well-characterized downstream effectors ROCK and LIMK.
- Published
- 2022
- Full Text
- View/download PDF
18. The F2-isoprostane 8-iso-PGF 2α attenuates atherosclerotic lesion formation in Ldlr-deficient mice - Potential role of vascular thromboxane A 2 receptors.
- Author
-
Braun H, Hauke M, Eckenstaler R, Petermann M, Ripperger A, Kühn N, Schwedhelm E, Ludwig-Kraus B, Kraus FB, Dubourg V, Zernecke A, Schreier B, Gekle M, and Benndorf RA
- Subjects
- Animals, Dinoprost analogs & derivatives, F2-Isoprostanes, Mice, Mice, Knockout, Placenta Growth Factor, Receptors, Thromboxane genetics, Thromboxane A2, Thromboxanes, Atherosclerosis genetics, Cardiovascular Diseases
- Abstract
The F2-isoprostane 8-iso-PGF
2α (also known as 15-F2t -isoprostane, iPF2α -III, 8-epi PGF2α , 15(S)-8-iso-PGF2α , or 8-Isoprostane), a thromboxane A2 receptor (TP) agonist, stable biomarker of oxidative stress, and risk marker of cardiovascular disease, has been proposed to aggravate atherogenesis in genetic mouse models of atherosclerotic vascular disease. Moreover, the TP plays an eminent role in the pathophysiology of endothelial dysfunction, atherogenesis, and cardiovascular disease. Yet it is unknown, how the TP expressed by vascular cells affects atherogenesis or 8-iso-PGF2α -related effects in mouse models of atherosclerosis. We studied Ldlr-deficient vascular endothelial-specific (EC) and vascular smooth muscle cell (VSMC)-specific TP knockout mice (TPEC KO /Ldlr KO; TPVSMC KO /Ldlr KO) and corresponding wild-type littermates (TPWT /Ldlr KO). The mice were fed a Western-type diet for eight weeks and received either 8-iso-PGF2α or vehicle infusions via osmotic pumps. Subsequently, arterial blood pressure, atherosclerotic lesion formation, and lipid profiles were analyzed. We found that VSMC-, but not EC-specific TP deletion, attenuated atherogenesis without affecting blood pressure or plasma lipid profiles of the mice. In contrast to a previous report, 8-iso-PGF2α tended to reduce atherogenesis in TPWT /Ldlr KO and TPEC KO /Ldlr KO mice, again without significantly affecting blood pressure or lipid profiles of these mice. However, no further reduction in atherogenesis was observed in 8-iso-PGF2α -treated TPVSMC KO /Ldlr KO mice. Our work suggests that the TP expressed in VSMC but not the TP expressed in EC is involved in atherosclerotic lesion formation in Ldlr-deficient mice. Furthermore, we report an inhibitory effect of 8-iso-PGF2α on atherogenesis in this experimental atherosclerosis model, which paradoxically appears to be related to the presence of the TP in VSMC., (Copyright © 2022 The Authors. Published by Elsevier Inc. All rights reserved.)- Published
- 2022
- Full Text
- View/download PDF
19. A pair of noncompeting neutralizing human monoclonal antibodies protecting from disease in a SARS-CoV-2 infection model.
- Author
-
Peter AS, Roth E, Schulz SR, Fraedrich K, Steinmetz T, Damm D, Hauke M, Richel E, Mueller-Schmucker S, Habenicht K, Eberlein V, Issmail L, Uhlig N, Dolles S, Grüner E, Peterhoff D, Ciesek S, Hoffmann M, Pöhlmann S, McKay PF, Shattock RJ, Wölfel R, Socher E, Wagner R, Eichler J, Sticht H, Schuh W, Neipel F, Ensser A, Mielenz D, Tenbusch M, Winkler TH, Grunwald T, Überla K, and Jäck HM
- Subjects
- Animals, Antibodies, Monoclonal, Antibodies, Neutralizing, Antibodies, Viral, Humans, Mice, SARS-CoV-2, COVID-19, Spike Glycoprotein, Coronavirus
- Abstract
TRIANNI mice carry an entire set of human immunoglobulin V region gene segments and are a powerful tool to rapidly isolate human monoclonal antibodies. After immunizing these mice with DNA encoding the spike protein of SARS-CoV-2 and boosting with spike protein, we identified 29 hybridoma antibodies that reacted with the SARS-CoV-2 spike protein. Nine antibodies neutralize SARS-CoV-2 infection at IC50 values in the subnanomolar range. ELISA-binding studies and DNA sequence analyses revealed one cluster of three clonally related neutralizing antibodies that target the receptor-binding domain and compete with the cellular receptor hACE2. A second cluster of six clonally related neutralizing antibodies bind to the N-terminal domain of the spike protein without competing with the binding of hACE2 or cluster 1 antibodies. SARS-CoV-2 mutants selected for resistance to an antibody from one cluster are still neutralized by an antibody from the other cluster. Antibodies from both clusters markedly reduced viral spread in mice transgenic for human ACE2 and protected the animals from SARS-CoV-2-induced weight loss. The two clusters of potent noncompeting SARS-CoV-2 neutralizing antibodies provide potential candidates for therapy and prophylaxis of COVID-19. The study further supports transgenic animals with a human immunoglobulin gene repertoire as a powerful platform in pandemic preparedness initiatives., (© 2021 The Authors. European Journal of Immunology published by Wiley-VCH GmbH.)
- Published
- 2022
- Full Text
- View/download PDF
20. A Thromboxane A 2 Receptor-Driven COX-2-Dependent Feedback Loop That Affects Endothelial Homeostasis and Angiogenesis.
- Author
-
Eckenstaler R, Ripperger A, Hauke M, Petermann M, Hemkemeyer SA, Schwedhelm E, Ergün S, Frye M, Werz O, Koeberle A, Braun H, and Benndorf RA
- Subjects
- Cyclooxygenase 2 genetics, Cyclooxygenase 2 metabolism, Cyclooxygenase 2 pharmacology, Feedback, Homeostasis, Humans, Receptors, Thromboxane A2, Prostaglandin H2 genetics, Thromboxane A2 metabolism, Thromboxanes metabolism, Thromboxanes pharmacology, Endothelial Cells metabolism, Receptors, Thromboxane metabolism
- Abstract
Background: TP (thromboxane A
2 receptor) plays an eminent role in the pathophysiology of endothelial dysfunction and cardiovascular disease. Moreover, its expression is reported to increase in the intimal layer of blood vessels of cardiovascular high-risk individuals. Yet it is unknown, whether TP upregulation per se has the potential to affect the homeostasis of the vascular endothelium., Methods: We combined global transcriptome analysis, lipid mediator profiling, functional cell analyses, and in vivo angiogenesis assays to study the effects of endothelial TP overexpression or knockdown/knockout on the angiogenic capacity of endothelial cells in vitro and in vivo., Results: Here we report that endothelial TP expression induces COX-2 (cyclooxygenase-2) in a Gi/o - and Gq/11 -dependent manner, thereby promoting its own activation via the auto/paracrine release of TP agonists, such as PGH2 (prostaglandin H2 ) or prostaglandin F2 but not TxA2 (thromboxane A2 ). TP overexpression induces endothelial cell tension and aberrant cell morphology, affects focal adhesion dynamics, and inhibits the angiogenic capacity of human endothelial cells in vitro and in vivo, whereas TP knockdown or endothelial-specific TP knockout exerts opposing effects. Consequently, this TP-dependent feedback loop is disrupted by pharmacological TP or COX-2 inhibition and by genetic reconstitution of PGH2 -metabolizing prostacyclin synthase even in the absence of functional prostacyclin receptor expression., Conclusions: Our work uncovers a TP-driven COX-2-dependent feedback loop and important effector mechanisms that directly link TP upregulation to angiostatic TP signaling in endothelial cells. By these previously unrecognized mechanisms, pathological endothelial upregulation of the TP could directly foster endothelial dysfunction, microvascular rarefaction, and systemic hypertension even in the absence of exogenous sources of TP agonists.- Published
- 2022
- Full Text
- View/download PDF
21. Krüppel-like factor 2 controls IgA plasma cell compartmentalization and IgA responses.
- Author
-
Wittner J, Schulz SR, Steinmetz TD, Berges J, Hauke M, Channell WM, Cunningham AF, Hauser AE, Hutloff A, Mielenz D, Jäck HM, and Schuh W
- Subjects
- Animals, Flagellin, Intestinal Mucosa, Mice, Immunoglobulin A metabolism, Kruppel-Like Transcription Factors genetics, Peyer's Patches, Plasma Cells
- Abstract
Krüppel-like factor 2 (KLF2) is a potent regulator of lymphocyte differentiation, activation and migration. However, its functional role in adaptive and humoral immunity remains elusive. Therefore, by using mice with a B cell-specific deletion of KLF2, we investigated plasma cell differentiation and antibody responses. We revealed that the deletion of KLF2 resulted in perturbed IgA plasma cell compartmentalization, characterized by the absence of IgA plasma cells in the bone marrow, their reductions in the spleen, the blood and the lamina propria of the colon and the small intestine, concomitant with their accumulation and retention in mesenteric lymph nodes and Peyer's patches. Most intriguingly, secretory IgA in the intestinal lumen was almost absent, dimeric serum IgA was drastically reduced and antigen-specific IgA responses to soluble Salmonella flagellin were blunted in KLF2-deficient mice. Perturbance of IgA plasma cell localization was caused by deregulation of CCR9, Integrin chains αM, α4, β7, and sphingosine-1-phosphate receptors. Hence, KLF2 not only orchestrates the localization of IgA plasma cells by fine-tuning chemokine receptors and adhesion molecules but also controls IgA responses to Salmonella flagellin., (© 2022. The Author(s).)
- Published
- 2022
- Full Text
- View/download PDF
Catalog
Discovery Service for Jio Institute Digital Library
For full access to our library's resources, please sign in.