167 results on '"Jäck HM"'
Search Results
2. Tfg (Trk fused gene) is a Carma-1/IKKγ interacting protein involved in CD40-induced canonical NF-κB signaling
- Author
-
Mielenz D, Jäck HM, Kalbacher H, Karas M, Hampel M, Hermann I, and Grohmann M
- Subjects
Medicine ,Cytology ,QH573-671 - Published
- 2009
- Full Text
- View/download PDF
3. Stages of germinal center transit are defined by B cell transcription factor coexpression and relative abundance
- Author
-
Cattoretti, G, Shaknovich, R, Smith, P, Jäck, H, Murty, V, Alobeid, B, CATTORETTI, GIORGIO, Smith, PM, Jäck, HM, Murty, VV, Alobeid, B., Cattoretti, G, Shaknovich, R, Smith, P, Jäck, H, Murty, V, Alobeid, B, CATTORETTI, GIORGIO, Smith, PM, Jäck, HM, Murty, VV, and Alobeid, B.
- Abstract
The transit of T cell-activated B cells through the germinal center (GC) is controlled by sequential activation and repression of key transcription factors, executing the pre- and post-GC B cell program. B cell lymphoma (BCL) 6 and IFN regulatory factor (IRF) 8 are necessary for GC formation and for its molecular activity in Pax5+PU.1+ B cells. IRF4, which is highly expressed in BCL6- GC B cells, is necessary for class switch recombination and the plasma cell differentiation at exit from the GC. In this study, we show at the single-cell level broad coexpression of IRF4 with BCL6, Pax5, IRF8, and PU.1 in pre- and post-GC B cells in human and mouse. IRF4 is down-regulated in BCL6+ human GC founder cells (IgD+CD38+), is absent in GC centroblasts, and is re-expressed in positive regulatory domain 1-positive centrocytes, which are negative for all the B cell transcription factors. Activated (CD30+) and activation-induced cytidine deaminase-positive extrafollicular blasts coexpress Pax5 and IRF4. PU.1-negative plasma cells and CD30+ blasts uniquely display the conformational epitope of IRF4 recognized by the MUM1 Ab, an epitope that is absent from any other IRF4+PU.1+ lymphoid and hemopoietic subsets. Low grade B cell lymphomas, representing the malignant counterpart of pre- and post-GC B cells, accordingly express IRF4. However, a fraction of BCL6+ diffuse large B cell lymphomas express IRF4 bearing the MUM1 epitope, indicative of a posttranscriptional modification of IRF4 not seen in the normal counterpart.
- Published
- 2006
4. Swiprosin 1 – regulator of proximal BCR signaling
- Author
-
Kroczek, C, primary, Lang, C, additional, Nitschke, L, additional, Elter, A, additional, Brachs, S, additional, Jäck, HM, additional, and Mielenz, D, additional
- Published
- 2009
- Full Text
- View/download PDF
5. Tfg (Trk fused gene) is a Carma-1/IKKγ interacting protein involved in CD40-induced canonical NF-κB signaling
- Author
-
Grohmann, M, primary, Hermann, I, additional, Hampel, M, additional, Karas, M, additional, Kalbacher, H, additional, Jäck, HM, additional, and Mielenz, D, additional
- Published
- 2009
- Full Text
- View/download PDF
6. Pre-B cell receptor-mediated cell cycle arrest in Philadelphia chromosome-positive acute lymphoblastic leukemia requires IKAROS function
- Author
-
Hans-Martin Jäck, Giovanni Martinelli, Yong-Mi Kim, Ilaria Iacobucci, Hassan Jumaa, Daniel Trageser, Wolfgang Schuh, Clelia Tiziana Storlazzi, Lars Klemm, Rahul Nahar, Eugene Park, Cihangir Duy, John Groffen, Tanja A. Gruber, Markus Müschen, Wolf-Karsten Hofmann, Sebastian Herzog, Aihong Li, Nora Heisterkamp, Gregor von Levetzow, Trageser D, Iacobucci I, Nahar R, Duy C, von Levetzow G, Klemm L, Park E, Schuh W, Gruber T, Herzog S, Kim YM, Hofmann WK, Li A, Storlazzi CT, Jäck HM, Groffen J, Martinelli G, Heisterkamp N, Jumaa H, and Müschen M.
- Subjects
Adult ,Cell cycle checkpoint ,Immunology ,B-cell receptor ,Down-Regulation ,Mice, Transgenic ,Biology ,Genes, abl ,Philadelphia chromosome ,Article ,03 medical and health sciences ,Ikaros Transcription Factor ,Mice ,0302 clinical medicine ,medicine ,Immunology and Allergy ,Animals ,Humans ,Philadelphia Chromosome ,PRE-B CELL RECEPTOR ,B cell ,030304 developmental biology ,Adaptor Proteins, Signal Transducing ,Mice, Knockout ,0303 health sciences ,ABL ,Leukemia, Prolymphocytic, B-Cell ,Cell Cycle ,ACUTE LYMPHOBLASTIC LEUKEMIA ,IKAROS ,Cell cycle ,medicine.disease ,medicine.anatomical_structure ,Cell Transformation, Neoplastic ,030220 oncology & carcinogenesis ,Pre-B Cell Receptors ,Cancer research ,Tyrosine kinase ,Gene Deletion ,Signal Transduction - Abstract
B cell lineage acute lymphoblastic leukemia (ALL) arises in virtually all cases from B cell precursors that are arrested at pre–B cell receptor–dependent stages. The Philadelphia chromosome–positive (Ph+) subtype of ALL accounts for 25–30% of cases of adult ALL, has the most unfavorable clinical outcome among all ALL subtypes and is defined by the oncogenic BCR-ABL1 kinase and deletions of the IKAROS gene in >80% of cases. Here, we demonstrate that the pre–B cell receptor functions as a tumor suppressor upstream of IKAROS through induction of cell cycle arrest in Ph+ ALL cells. Pre–B cell receptor–mediated cell cycle arrest in Ph+ ALL cells critically depends on IKAROS function, and is reversed by coexpression of the dominant-negative IKAROS splice variant IK6. IKAROS also promotes tumor suppression through cooperation with downstream molecules of the pre–B cell receptor signaling pathway, even if expression of the pre–B cell receptor itself is compromised. In this case, IKAROS redirects oncogenic BCR-ABL1 tyrosine kinase signaling from SRC kinase-activation to SLP65, which functions as a critical tumor suppressor downstream of the pre–B cell receptor. These findings provide a rationale for the surprisingly high frequency of IKAROS deletions in Ph+ ALL and identify IKAROS-mediated cell cycle exit as the endpoint of an emerging pathway of pre–B cell receptor–mediated tumor suppression.
- Published
- 2009
7. An Enhanced Retroviral Vector for Efficient Genetic Manipulation and Selection in Mammalian Cells.
- Author
-
Triller J, Prots I, Jäck HM, and Wittmann J
- Subjects
- Humans, Animals, Puromycin pharmacology, HEK293 Cells, Transgenes, Mice, Genetic Vectors genetics, Green Fluorescent Proteins genetics, Green Fluorescent Proteins metabolism, Moloney murine leukemia virus genetics, Retroviridae genetics
- Abstract
Introducing genetic material into hard-to-transfect mammalian cell lines and primary cells is often best achieved through retroviral infection. An ideal retroviral vector should offer a compact, selectable, and screenable marker while maximizing transgene delivery capacity. However, a previously published retroviral vector featuring an EGFP/Puromycin fusion protein failed to meet these criteria in our experiments. We encountered issues such as low infection efficiency, weak EGFP fluorescence, and selection against infected cells. To address these shortcomings, we developed a novel retroviral vector based on the Moloney murine leukemia virus. This vector includes a compact bifunctional EGFP and Puromycin resistance cassette connected by a 2A peptide. Our extensively tested vector demonstrated superior EGFP expression, efficient Puromycin selection, and no growth penalty in infected cells compared with the earlier design. These benefits were consistent across multiple mammalian cell types, underscoring the versatility of our vector. In summary, our enhanced retroviral vector offers a robust solution for efficient infection, reliable detection, and effective selection in mammalian cells. Its improved performance and compact design make it an ideal choice for a wide range of applications involving precise genetic manipulation and characterization in cell-based studies.
- Published
- 2024
- Full Text
- View/download PDF
8. Metabolic profiling of single cells by exploiting NADH and FAD fluorescence via flow cytometry.
- Author
-
Abir AH, Weckwerth L, Wilhelm A, Thomas J, Reichardt CM, Munoz L, Völkl S, Appelt U, Mroz M, Niesner R, Hauser A, Sophie Fischer R, Pracht K, Jäck HM, Schett G, Krönke G, and Mielenz D
- Subjects
- Humans, Mitochondria metabolism, T-Lymphocytes metabolism, Oxidation-Reduction, Fluorescence, Arthritis, Rheumatoid metabolism, Glycolysis, Oxidative Phosphorylation, Female, Male, Glucose metabolism, Flow Cytometry methods, NAD metabolism, Flavin-Adenine Dinucleotide metabolism, Single-Cell Analysis methods, B-Lymphocytes metabolism
- Abstract
Objective: The metabolism of different cells within the same microenvironment can differ and dictate physiological or pathological adaptions. Current single-cell analysis methods of metabolism are not label-free., Methods: The study introduces a label-free, live-cell analysis method assessing endogenous fluorescence of NAD(P)H and FAD in surface-stained cells by flow cytometry., Results: OxPhos inhibition, mitochondrial uncoupling, glucose exposure, genetic inactivation of glucose uptake and mitochondrial respiration alter the optical redox ratios of FAD and NAD(P)H as measured by flow cytometry. Those alterations correlate strongly with measurements obtained by extracellular flux analysis. Consequently, metabolically distinct live B-cell populations can be resolved, showing that human memory B-cells from peripheral blood exhibit a higher glycolytic flexibility than naïve B cells. Moreover, the comparison of blood-derived B- and T-lymphocytes from healthy donors and rheumatoid arthritis patients unleashes rheumatoid arthritis-associated metabolic traits in human naïve and memory B-lymphocytes., Conclusions: Taken together, these data show that the optical redox ratio can depict metabolic differences in distinct cell populations by flow cytometry., 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 © 2024 The Authors. Published by Elsevier GmbH.. All rights reserved.)
- Published
- 2024
- Full Text
- View/download PDF
9. Identification of TFG- and Autophagy-Regulated Proteins and Glycerophospholipids in B Cells.
- Author
-
Steinmetz TD, Thomas J, Reimann L, Himmelreich AK, Schulz SR, Golombek F, Castiglione K, Jäck HM, Brodesser S, Warscheid B, and Mielenz D
- Subjects
- Animals, Mice, Lipid Metabolism, Lipidomics methods, Mitochondria metabolism, Proteomics methods, Autophagy, B-Lymphocytes metabolism, Glycerophospholipids metabolism, Lysosomes metabolism
- Abstract
Autophagy supervises the proteostasis and survival of B lymphocytic cells. Trk-fused gene (TFG) promotes autophagosome-lysosome flux in murine CH12 B cells, as well as their survival. Hence, quantitative proteomics of CH12 tfg KO and WT B cells in combination with lysosomal inhibition should identify proteins that are prone to lysosomal degradation and contribute to autophagy and B cell survival. Lysosome inhibition via NH
4 Cl unexpectedly reduced a number of proteins but increased a large cluster of translational, ribosomal, and mitochondrial proteins, independent of TFG. Hence, we propose a role for lysosomes in ribophagy in B cells. TFG-regulated proteins include CD74, BCL10, or the immunoglobulin JCHAIN. Gene ontology (GO) analysis reveals that proteins regulated by TFG alone, or in concert with lysosomes, localize to mitochondria and membrane-bound organelles. Likewise, TFG regulates the abundance of metabolic enzymes, such as ALDOC and the fatty acid-activating enzyme ACOT9. To test consequently for a function of TFG in lipid metabolism, we performed shotgun lipidomics of glycerophospholipids. Total phosphatidylglycerol is more abundant in CH12 tfg KO B cells. Several glycerophospholipid species with similar acyl side chains, such as 36:2 phosphatidylethanolamine and 36:2 phosphatidylinositol, show a dysequilibrium. We suggest a role for TFG in lipid homeostasis, mitochondrial functions, translation, and metabolism in B cells.- Published
- 2024
- Full Text
- View/download PDF
10. Virological Traits of the SARS-CoV-2 BA.2.87.1 Lineage.
- Author
-
Zhang L, Dopfer-Jablonka A, Nehlmeier I, Kempf A, Graichen L, Calderón Hampel N, Cossmann A, Stankov MV, Morillas Ramos G, Schulz SR, Jäck HM, Behrens GMN, Pöhlmann S, and Hoffmann M
- Abstract
Transmissibility and immune evasion of the recently emerged, highly mutated SARS-CoV-2 BA.2.87.1 are unknown. Here, we report that BA.2.87.1 efficiently enters human cells but is more sensitive to antibody-mediated neutralization than the currently dominating JN.1 variant. Acquisition of adaptive mutations might thus be needed for efficient spread in the population.
- Published
- 2024
- Full Text
- View/download PDF
11. 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
12. SARS-CoV-2 BA.2.86 enters lung cells and evades neutralizing antibodies with high efficiency.
- Author
-
Zhang L, Kempf A, Nehlmeier I, Cossmann A, Richter A, Bdeir N, Graichen L, Moldenhauer AS, Dopfer-Jablonka A, Stankov MV, Simon-Loriere E, Schulz SR, Jäck HM, Čičin-Šain L, Behrens GMN, Drosten C, Hoffmann M, and Pöhlmann S
- Subjects
- Humans, Caspases metabolism, Lung virology, Virus Internalization, Spike Glycoprotein, Coronavirus genetics, Antibodies, Neutralizing metabolism, Antibodies, Viral metabolism, COVID-19 immunology, COVID-19 virology, SARS-CoV-2 classification, SARS-CoV-2 genetics, SARS-CoV-2 pathogenicity, SARS-CoV-2 physiology
- Abstract
BA.2.86, a recently identified descendant of the severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) Omicron BA.2 sublineage, contains ∼35 mutations in the spike (S) protein and spreads in multiple countries. Here, we investigated whether the virus exhibits altered biological traits, focusing on S protein-driven viral entry. Employing pseudotyped particles, we show that BA.2.86, unlike other Omicron sublineages, enters Calu-3 lung cells with high efficiency and in a serine- but not cysteine-protease-dependent manner. Robust lung cell infection was confirmed with authentic BA.2.86, but the virus exhibited low specific infectivity. Further, BA.2.86 was highly resistant against all therapeutic antibodies tested, efficiently evading neutralization by antibodies induced by non-adapted vaccines. In contrast, BA.2.86 and the currently circulating EG.5.1 sublineage were appreciably neutralized by antibodies induced by the XBB.1.5-adapted vaccine. Collectively, BA.2.86 has regained a trait characteristic of early SARS-CoV-2 lineages, robust lung cell entry, and evades neutralizing antibodies. However, BA.2.86 exhibits low specific infectivity, which might limit transmissibility., Competing Interests: Declaration of interests A.K., I.N., S.P. and M.H. conducted contract research (testing of vaccinee sera for neutralizing activity against SARS-CoV-2) for Valneva unrelated to this work. G.M.N.B. served as advisor for Moderna, and S.P. served as advisor for BioNTech, unrelated to this work. A.D-J. served as advisor for Pfizer, unrelated to this work. The authors declare no competing interests., (Copyright © 2023 Elsevier Inc. All rights reserved.)
- Published
- 2024
- Full Text
- View/download PDF
13. 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
14. Neutralisation sensitivity of SARS-CoV-2 lineages EG.5.1 and XBB.2.3.
- Author
-
Zhang L, Kempf A, Nehlmeier I, Cossmann A, Dopfer-Jablonka A, Stankov MV, Schulz SR, Jäck HM, Behrens GMN, Pöhlmann S, and Hoffmann M
- Subjects
- Humans, SARS-CoV-2 genetics, COVID-19
- Abstract
Competing Interests: AK, IN, SP, and MH did the contract research (testing of vaccinee sera for neutralising activity against SARS-CoV-2) for Valneva, unrelated to this work. GMNB served as an adviser for Moderna, unrelated to this work. SP served as an adviser for BioNTech, unrelated to this work. AD-J served as an adviser for Pfizer, unrelated to this work. All other authors declare no competing interests. SP acknowledges funding by the EU project UNDINE (grant agreement number 101057100), the Ministry for Science and Culture of Lower Saxony (Niedersächsisches Ministerium für Wissenschaft und Kultur; 14-76103-184, COFONI Network, projects 7FF22, 6FF22, 10FF22) and the German Research Foundation (Deutsche Forschungsgemeinschaft; PO 716/11-1). H-MJ received funding from BMBF (01KI2043, NaFoUniMedCovid19-COVIM: 01KX2021), Bavarian State Ministry for Science and the Arts, and Deutsche Forschungsgemeinschaft through the research training groups RTG1660 and TRR130, the Bayerische Forschungsstiftung (Project CORAd) and the Kastner Foundation. GMNB acknowledges funding by German Center for Infection Research (grant 80018019238). GMNB and AD-J acknowledge funding by a European Regional Development Fund (Defeat Corona, ZW7-8515131). The funding sources had no role in the design and execution of the study, the writing of the manuscript, and the decision to submit the manuscript for publication. The authors did not receive payment by a pharmaceutical company or other agency to write the publication. The authors were not precluded from accessing data in the study, and they accept responsibility to submit for publication.
- Published
- 2023
- Full Text
- View/download PDF
15. Host cell entry and neutralisation sensitivity of the SARS-CoV-2 XBB.1.16 lineage.
- Author
-
Nehlmeier I, Kempf A, Arora P, Cossmann A, Dopfer-Jablonka A, Stankov MV, Schulz SR, Jäck HM, Behrens GMN, Pöhlmann S, and Hoffmann M
- Subjects
- Humans, SARS-CoV-2, Virus Internalization, COVID-19
- Published
- 2023
- Full Text
- View/download PDF
16. Profound neutralization evasion and augmented host cell entry are hallmarks of the fast-spreading SARS-CoV-2 lineage XBB.1.5.
- Author
-
Hoffmann M, Arora P, Nehlmeier I, Kempf A, Cossmann A, Schulz SR, Morillas Ramos G, Manthey LA, Jäck HM, Behrens GMN, and Pöhlmann S
- Subjects
- Humans, SARS-CoV-2, Virus Internalization, Antibodies, Viral, Spike Glycoprotein, Coronavirus, Immune Evasion, Antibodies, Neutralizing, COVID-19
- Published
- 2023
- Full Text
- View/download PDF
17. The intestine: A highly dynamic microenvironment for IgA plasma cells.
- Author
-
Pracht K, Wittner J, Kagerer F, Jäck HM, and Schuh W
- Subjects
- Intestinal Mucosa, Cytokines, Immunoglobulin A, Plasma Cells, Intestines
- Abstract
To achieve longevity, IgA plasma cells require a sophisticated anatomical microenvironment that provides cytokines, cell-cell contacts, and nutrients as well as metabolites. The intestinal epithelium harbors cells with distinct functions and represents an important defense line. Anti-microbial peptide-producing paneth cells, mucus-secreting goblet cells and antigen-transporting microfold (M) cells cooperate to build a protective barrier against pathogens. In addition, intestinal epithelial cells are instrumental in the transcytosis of IgA to the gut lumen, and support plasma cell survival by producing the cytokines APRIL and BAFF. Moreover, nutrients are sensed through specialized receptors such as the aryl hydrocarbon receptor (AhR) by both, intestinal epithelial cells and immune cells. However, the intestinal epithelium is highly dynamic with a high cellular turn-over rate and exposure to changing microbiota and nutritional factors. In this review, we discuss the spatial interplay of the intestinal epithelium with plasma cells and its potential contribution to IgA plasma cell generation, homing, and longevity. Moreover, we describe the impact of nutritional AhR ligands on intestinal epithelial cell-IgA plasma cell interaction. Finally, we introduce spatial transcriptomics as a new technology to address open questions in intestinal IgA plasma cell biology., 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 © 2023 Pracht, Wittner, Kagerer, Jäck and Schuh.)
- Published
- 2023
- Full Text
- View/download PDF
18. Broadly neutralizing SARS-CoV-2 antibodies through epitope-based selection from convalescent patients.
- Author
-
Rouet R, Henry JY, Johansen MD, Sobti M, Balachandran H, Langley DB, Walker GJ, Lenthall H, Jackson J, Ubiparipovic S, Mazigi O, Schofield P, Burnett DL, Brown SHJ, Martinello M, Hudson B, Gilroy N, Post JJ, Kelleher A, Jäck HM, Goodnow CC, Turville SG, Rawlinson WD, Bull RA, Stewart AG, Hansbro PM, and Christ D
- Subjects
- Humans, Female, Animals, Mice, Broadly Neutralizing Antibodies, Leukocytes, Mononuclear, Antibodies, Viral, Antibodies, Monoclonal, Antibodies, Neutralizing, Epitopes, Spike Glycoprotein, Coronavirus genetics, Neutralization Tests, SARS-CoV-2, COVID-19
- Abstract
Emerging variants of concern (VOCs) are threatening to limit the effectiveness of SARS-CoV-2 monoclonal antibodies and vaccines currently used in clinical practice; broadly neutralizing antibodies and strategies for their identification are therefore urgently required. Here we demonstrate that broadly neutralizing antibodies can be isolated from peripheral blood mononuclear cells of convalescent patients using SARS-CoV-2 receptor binding domains carrying epitope-specific mutations. This is exemplified by two human antibodies, GAR05, binding to epitope class 1, and GAR12, binding to a new epitope class 6 (located between class 3 and 5). Both antibodies broadly neutralize VOCs, exceeding the potency of the clinical monoclonal sotrovimab (S309) by orders of magnitude. They also provide prophylactic and therapeutic in vivo protection of female hACE2 mice against viral challenge. Our results indicate that exposure to SARS-CoV-2 induces antibodies that maintain broad neutralization against emerging VOCs using two unique strategies: either by targeting the divergent class 1 epitope in a manner resistant to VOCs (ACE2 mimicry, as illustrated by GAR05 and mAbs P2C-1F11/S2K14); or alternatively, by targeting rare and highly conserved epitopes, such as the new class 6 epitope identified here (as illustrated by GAR12). Our results provide guidance for next generation monoclonal antibody development and vaccine design., (© 2023. The Author(s).)
- Published
- 2023
- Full Text
- View/download PDF
19. Neutralisation sensitivity of the SARS-CoV-2 XBB.1 lineage.
- Author
-
Arora P, Cossmann A, Schulz SR, Ramos GM, Stankov MV, Jäck HM, Behrens GMN, Pöhlmann S, and Hoffmann M
- Subjects
- Humans, SARS-CoV-2, Antibodies, Viral, COVID-19
- Abstract
Competing Interests: SP and MH do contract research on the testing of vaccinee serum samples for neutralising activity against SARS-CoV-2 for Valneva, unrelated to this work. GMNB served as an advisor for Moderna and SP served as an advisor for BioNTech, unrelated to this work. All other authors declare no competing interests. SP acknowledges funding for this project by the German Federal Ministry of Education and Research (01KI2006D), the EU project UNDINE (grant agreement number 101057100), the Ministry for Science and Culture of Lower Saxony (14-76103-184, MWK HZI COVID-19), and the German Research Foundation (PO 716/11-1 and PO 716/14-1). H-MJ received funding from the German Federal Ministry of Education and Research (01KI2043, NaFoUniMedCovid19-COVIM 01KX2021), Bavarian State Ministry for Science and the Arts; and DFG through the research training groups RTG1660 and TRR130, the Bayerische Forschungsstiftung (Project CORAd), and the Kastner Foundation. GMNB acknowledges funding by the German Center for Infection Research (grant number 80018019238) and a European Regional Development Fund (Defeat Corona, ZW7-8515131). The funding sources had no role in study design, data collection, data analysis, data interpretation, writing of the Correspondence, or the decision to submit the manuscript for publication. We did not receive payment by a pharmaceutical company or other agency to write this Correspondence. We were not precluded from accessing data in the study and we accept responsibility to submit for publication.
- Published
- 2023
- Full Text
- View/download PDF
20. Omicron sublineage BQ.1.1 resistance to monoclonal antibodies.
- Author
-
Arora P, Kempf A, Nehlmeier I, Schulz SR, Jäck HM, Pöhlmann S, and Hoffmann M
- Subjects
- Humans, Antibodies, Neutralizing, Antibodies, Monoclonal therapeutic use, Antibodies, Viral
- Abstract
Competing Interests: AK, IN, and MH do contract research (testing of vaccinee sera for neutralising activity against SARS-CoV-2) for Valneva, unrelated to this Correspondence. SP does contract research (testing of vaccinee sera for neutralising activity against SARS-CoV-2) for Valneva and served as advisor for BioNTech, unrelated to Correspondence, and acknowledges funding by the Federal Ministry of Education and Research (Bundesministerium für Bildung und Forschung [BMBF]; 01KI2006D, 01KI20328A, and 01KX2021), the EU project UNDINE (grant agreement number 101057100), the Ministry for Science and Culture of Lower Saxony (14–76103–184, MWK HZI COVID-19), and the German Research Foundation (DFG; PO 716/11–1 and PO 716/14–1). H-MJ received funding from BMBF (01KI2043 and NaFoUniMedCovid19-COVIM: 01KX2021), the Bavarian State Ministry for Science and the Arts and DFG through the research training groups RTG1660 and TRR130, the Bayerische Forschungsstiftung (Project CORAd), and the Kastner Foundation. All other authors declare no competing interests.
- Published
- 2023
- Full Text
- View/download PDF
21. 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
22. The effect of cilgavimab and neutralisation by vaccine-induced antibodies in emerging SARS-CoV-2 BA.4 and BA.5 sublineages.
- Author
-
Arora P, Zhang L, Nehlmeier I, Kempf A, Cossmann A, Dopfer-Jablonka A, Schulz SR, Jäck HM, Behrens GMN, Pöhlmann S, and Hoffmann M
- Subjects
- Humans, SARS-CoV-2, Antibodies, Viral, Antibodies, Neutralizing, COVID-19 prevention & control, Vaccines
- Abstract
Competing Interests: AK, IN, SP, and MH conduct contract research (testing of vaccinee sera for neutralising activity against SARS-CoV-2) for Valneva, unrelated to this work. GMNB served as an advisor for Moderna and acknowledges funding by German Center for Infection Research (grant no 80018019238) and a European Regional Development Fund (Defeat Corona, ZW78–515131). SP served as an advisor for BioNTech, unrelated to this work. SP acknowledges funding by Bundesministerium fur Bildung und Forshung (Bundesministerium fur Bildung und Forshung; 01KI2006D, 01KI20328A, 01KX2021), the EU hunam genetic and immunological determinants of the clinical manifestations of SARS-Cov-2 infection: Towards personalised medicine project (grant agreement number 101057100), the Ministry for Science and Culture of Lower Saxony (147–61031–84, MWK HZI COVID-19), and the German Research Foundation (DFG; PO716/111–, PO716/141–). H-MJ received funding from BMBF (01KI2043, NaFoUniMedCovid19-COVIM: 01KX2021), Bavarian State Ministry for Science and the Arts and DFG through the research training groups RTG1660 and TRR130, the Bayerische Forschungsstiftung (Project CORAd), and the Kastner Foundation. All other authors declare no competing interests.
- Published
- 2022
- Full Text
- View/download PDF
23. Host Cell Entry and Neutralization Sensitivity of SARS-CoV-2 Lineages B.1.620 and R.1.
- Author
-
Sidarovich A, Krüger N, Rocha C, Graichen L, Kempf A, Nehlmeier I, Lier M, Cossmann A, Stankov MV, Schulz SR, Behrens GMN, Jäck HM, Pöhlmann S, and Hoffmann M
- Subjects
- Humans, Spike Glycoprotein, Coronavirus, Angiotensin-Converting Enzyme 2, Virus Internalization, Peptidyl-Dipeptidase A metabolism, Antibodies, Neutralizing, Antibodies, Viral, Mutation, SARS-CoV-2 genetics, COVID-19
- Abstract
The spike (S) protein of severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) facilitates viral entry into host cells and is the key target for neutralizing antibodies. The SARS-CoV-2 lineage B.1.620 carries fifteen mutations in the S protein and is spread in Africa, the US and Europe, while lineage R.1 harbors four mutations in S and infections were observed in several countries, particularly Japan and the US. However, the impact of the mutations in B.1.620 and R.1 S proteins on antibody-mediated neutralization and host cell entry are largely unknown. Here, we report that these mutations are compatible with robust ACE2 binding and entry into cell lines, and they markedly reduce neutralization by vaccine-induced antibodies. Our results reveal evasion of neutralizing antibodies by B.1.620 and R.1, which might have contributed to the spread of these lineages.
- Published
- 2022
- Full Text
- View/download PDF
24. Lung cell entry, cell-cell fusion capacity, and neutralisation sensitivity of omicron sublineage BA.2.75.
- Author
-
Arora P, Nehlmeier I, Kempf A, Cossmann A, Schulz SR, Dopfer-Jablonka A, Baier E, Tampe B, Moerer O, Dickel S, Winkler MS, Jäck HM, Behrens GMN, Pöhlmann S, and Hoffmann M
- Subjects
- Humans, Cell Fusion, Thorax, Lung, Antibodies, Viral, Virus Internalization, HIV-1
- Abstract
Competing Interests: AK, IN, SP, and MH conduct contract research (testing of vaccine sera for neutralising activity against SARS-CoV-2) for Valneva unrelated to this work. GMNB was an advisor for Moderna and SP was an advisor for BioNTech, unrelated to this work. SP acknowledges funding by the Federal Ministry of Education and Research (BMBF; grant numbers 01KI2006D, 01KI20328A, and 01KX2021), the EU UNDINE project (grant agreement number 101057100), the Ministry for Science and Culture of Lower Saxony (grant numbers 14-76103-184 and MWK HZI COVID-19), and the German Research Foundation (Deutsche Forschungsgemeinschaft; PO 716/11-1 and PO 716/14-1). MSW received unrestricted funding from Sartorius AG, Lung research. H-MJ received funding from BMBF (grant numbers 01KI2043 and NaFoUniMedCovid19-COVIM: 01KX2021), Bavarian State Ministry for Science and the Arts and Deutsche Forschungsgemeinschaft through the research training groups RTG1660 and TRR130, the Bayerische Forschungsstiftung (Project CORAd), and the Kastner Foundation. GMNB acknowledges funding by German Center for Infection Research (grant number 80018019238) and a European Regional Development Fund (Defeat Corona, grant number ZW7-8515131). All other authors declare no competing interests.
- Published
- 2022
- Full Text
- View/download PDF
25. IRF4 deficiency vulnerates B-cell progeny for leukemogenesis via somatically acquired Jak3 mutations conferring IL-7 hypersensitivity.
- Author
-
Das Gupta D, Paul C, Samel N, Bieringer M, Staudenraus D, Marini F, Raifer H, Menke L, Hansal L, Camara B, Roth E, Daum P, Wanzel M, Mernberger M, Nist A, Bauer UM, Helmprobst F, Buchholz M, Roth K, Bastian L, Hartmann AM, Baldus C, Ikuta K, Neubauer A, Burchert A, Jäck HM, Klein M, Bopp T, Stiewe T, Pagenstecher A, and Lohoff M
- Subjects
- Adult, Animals, Humans, Mice, B-Lymphocytes, Interleukin-7 genetics, Janus Kinase 3 genetics, Mutation genetics, Signal Transduction, Burkitt Lymphoma pathology, Precursor Cell Lymphoblastic Leukemia-Lymphoma genetics
- Abstract
The processes leading from disturbed B-cell development to adult B-cell progenitor acute lymphoblastic leukemia (BCP-ALL) remain poorly understood. Here, we describe Irf4
-/- mice as prone to developing BCP-ALL with age. Irf4-/- preB-I cells exhibited impaired differentiation but enhanced proliferation in response to IL-7, along with reduced retention in the IL-7 providing bone marrow niche due to decreased CXCL12 responsiveness. Thus selected, preB-I cells acquired Jak3 mutations, probably following irregular AID activity, resulting in malignant transformation. We demonstrate heightened IL-7 sensitivity due to Jak3 mutants, devise a model to explain it, and describe structural and functional similarities to Jak2 mutations often occurring in human Ph-like ALL. Finally, targeting JAK signaling with Ruxolitinib in vivo prolonged survival of mice bearing established Irf4-/- leukemia. Intriguingly, organ infiltration including leukemic meningeosis was selectively reduced without affecting blood blast counts. In this work, we present spontaneous leukemogenesis following IRF4 deficiency with potential implications for high-risk BCP-ALL in adult humans., (© 2022. The Author(s).)- Published
- 2022
- Full Text
- View/download PDF
26. Efficient antibody evasion but reduced ACE2 binding by the emerging SARS-CoV-2 variant B.1.640.2.
- Author
-
Arora P, Kempf A, Nehlmeier I, Graichen L, Schulz S, Cossmann A, Dopfer-Jablonka A, Winkler MS, Jäck HM, Behrens GMN, Pöhlmann S, and Hoffmann M
- Subjects
- Angiotensin-Converting Enzyme 2, Binding Sites, Humans, Mutation, Protein Binding, Spike Glycoprotein, Coronavirus genetics, Spike Glycoprotein, Coronavirus metabolism, COVID-19, SARS-CoV-2
- Published
- 2022
- Full Text
- View/download PDF
27. SARS-CoV-2 Omicron sublineages show comparable cell entry but differential neutralization by therapeutic antibodies.
- Author
-
Arora P, Zhang L, Krüger N, Rocha C, Sidarovich A, Schulz S, Kempf A, Graichen L, Moldenhauer AS, Cossmann A, Dopfer-Jablonka A, Behrens GMN, Jäck HM, Pöhlmann S, and Hoffmann M
- Subjects
- Antibodies, Monoclonal, Antibodies, Monoclonal, Humanized, Antibodies, Neutralizing therapeutic use, Antibodies, Viral therapeutic use, BNT162 Vaccine, Humans, Virus Internalization, SARS-CoV-2, COVID-19 Drug Treatment
- Abstract
The Omicron variant of SARS-CoV-2 evades antibody-mediated neutralization with unprecedented efficiency. At least three Omicron sublineages have been identified-BA.1, BA.2, and BA.3-and BA.2 exhibits increased transmissibility. However, it is currently unknown whether BA.2 differs from the other sublineages regarding cell entry and antibody-mediated inhibition. Here, we show that BA.1, BA.2, and BA.3 enter and fuse target cells with similar efficiency and in an ACE2-dependent manner. However, BA.2 was not efficiently neutralized by seven of eight antibodies used for COVID-19 therapy, including Sotrovimab, which robustly neutralized BA.1. In contrast, BA.2 and BA.3 (but not BA.1) were appreciably neutralized by Cilgavimab, which could constitute a treatment option. Finally, all sublineages were comparably and efficiently neutralized by antibodies induced by BNT162b2 booster vaccination after previous two-dose homologous or heterologous vaccination. Collectively, the Omicron sublineages show comparable cell entry and neutralization by vaccine-induced antibodies but differ in susceptibility to therapeutic antibodies., Competing Interests: Declaration of interests The authors declare no competing interests., (Copyright © 2022 Elsevier Inc. All rights reserved.)
- Published
- 2022
- Full Text
- View/download PDF
28. Augmented neutralisation resistance of emerging omicron subvariants BA.2.12.1, BA.4, and BA.5.
- Author
-
Arora P, Kempf A, Nehlmeier I, Schulz SR, Cossmann A, Stankov MV, Jäck HM, Behrens GMN, Pöhlmann S, and Hoffmann M
- Abstract
Competing Interests: AK, IN, SP, and MH conduct contract research (ie, testing of vaccinee sera for neutralising activity against SARS-CoV-2) for an industrial entity, unrelated to this Correspondence. GMNB served as an adviser for Moderna, unrelated to this Correspondence. All other authors declare no competing interests. SP acknowledges funding by Bundesministerium für Bildung und Forschung (01KI2006D, 01KI20328A, 01KX2021), the Ministry for Science and Culture of Lower Saxony (14–76103–184, MWK HZI COVID-19), and the German Research Foundation (PO 716/11–1, PO 716/14–1). H-MJ received funding from BMBF (01KI2043, NaFoUniMedCovid19-COVIM: 01KX2021), Bavarian State Ministry for Science and the Arts, and Deutsche Forschungsgemeinschaft through the research training groups RTG1660 and TRR130, the Bayerische Forschungsstiftung (Project CORAd), and the Kastner Foundation. GMNB acknowledges funding by German Center for Infection Research (grant no 80018019238) and a European Regional Development Fund (Defeat Corona, ZW7–8515131).
- Published
- 2022
- Full Text
- View/download PDF
29. Evidence for an ACE2-Independent Entry Pathway That Can Protect from Neutralization by an Antibody Used for COVID-19 Therapy.
- Author
-
Hoffmann M, Sidarovich A, Arora P, Krüger N, Nehlmeier I, Kempf A, Graichen L, Winkler MS, Niemeyer D, Goffinet C, Drosten C, Schulz S, Jäck HM, and Pöhlmann S
- Subjects
- Angiotensin-Converting Enzyme 2, Animals, Antibodies, Viral, Cell Line, Chlorocebus aethiops, Humans, Mutation, Protein Binding, Receptors, Virus metabolism, Spike Glycoprotein, Coronavirus metabolism, Vero Cells, Antibodies, Monoclonal, Humanized pharmacology, Antibodies, Neutralizing pharmacology, COVID-19 therapy, SARS-CoV-2 physiology, Virus Internalization
- Abstract
SARS-CoV-2 variants of concern (VOC) acquired mutations in the spike (S) protein, including E484K, that confer resistance to neutralizing antibodies. However, it is incompletely understood how these mutations impact viral entry into host cells. Here, we analyzed how mutations at position 484 that have been detected in COVID-19 patients impact cell entry and antibody-mediated neutralization. We report that mutation E484D markedly increased SARS-CoV-2 S-driven entry into the hepatoma cell line Huh-7 and the lung cell NCI-H1299 without augmenting ACE2 binding. Notably, mutation E484D largely rescued Huh-7 but not Vero cell entry from blockade by the neutralizing antibody Imdevimab and rendered Huh-7 cell entry ACE2-independent. These results suggest that the naturally occurring mutation E484D allows SARS-CoV-2 to employ an ACE2-independent mechanism for entry that is largely insensitive against Imdevimab, an antibody employed for COVID-19 therapy. IMPORTANCE The interaction of the SARS-CoV-2 spike protein (S) with the cellular receptor ACE2 is considered essential for infection and constitutes the key target for antibodies induced upon infection and vaccination. Here, using a surrogate system for viral entry, we provide evidence that a naturally occurring mutation can liberate SARS-CoV-2 from ACE2-dependence and that ACE2-independent entry may protect the virus from neutralization by an antibody used for COVID-19 therapy.
- Published
- 2022
- Full Text
- View/download PDF
30. BAFFR activates PI3K/AKT signaling in human naive but not in switched memory B cells through direct interactions with B cell antigen receptors.
- Author
-
Sevdali E, Block V, Lataretu M, Li H, Smulski CR, Briem JS, Heitz Y, Fischer B, Ramirez NJ, Grimbacher B, Jäck HM, Voll RE, Hölzer M, Schneider P, and Eibel H
- Subjects
- B-Cell Activating Factor immunology, B-Cell Activating Factor metabolism, Humans, B-Cell Activation Factor Receptor immunology, B-Cell Activation Factor Receptor metabolism, Memory B Cells immunology, Memory B Cells metabolism, Phosphatidylinositol 3-Kinases immunology, Phosphatidylinositol 3-Kinases metabolism, Proto-Oncogene Proteins c-akt immunology, Proto-Oncogene Proteins c-akt metabolism, Receptors, Antigen, B-Cell immunology, Receptors, Antigen, B-Cell metabolism
- Abstract
Binding of BAFF to BAFFR activates in mature B cells PI3K/AKT signaling regulating protein synthesis, metabolic fitness, and survival. In humans, naive and memory B cells express the same levels of BAFFR, but only memory B cells seem to survive without BAFF. Here, we show that BAFF activates PI3K/AKT only in naive B cells and changes the expression of genes regulating migration, proliferation, growth, and survival. BAFF-induced PI3K/AKT activation requires direct interactions between BAFFR and the B cell antigen receptor (BCR) components CD79A and CD79B and is enhanced by the AKT coactivator TCL1A. Compared to memory B cells, naive B cells express more surface BCRs, which interact better with BAFFR than IgG or IgA, thus allowing stronger responses to BAFF. As ablation of BAFFR in naive and memory B cells causes cell death independent of BAFF-induced signaling, BAFFR seems to act also as an intrinsic factor for B cell survival., Competing Interests: Declaration of interests The authors declare no competing interests., (Copyright © 2022 The Author(s). Published by Elsevier Inc. All rights reserved.)
- Published
- 2022
- Full Text
- View/download PDF
31. Comparable neutralisation evasion of SARS-CoV-2 omicron subvariants BA.1, BA.2, and BA.3.
- Author
-
Arora P, Zhang L, Rocha C, Sidarovich A, Kempf A, Schulz S, Cossmann A, Manger B, Baier E, Tampe B, Moerer O, Dickel S, Dopfer-Jablonka A, Jäck HM, Behrens GMN, Winkler MS, Pöhlmann S, and Hoffmann M
- Subjects
- Humans, Mutation, Spike Glycoprotein, Coronavirus genetics, COVID-19, SARS-CoV-2
- Abstract
Competing Interests: SP acknowledges funding from Bundesministerium für Bildung und Forschung (BMBF; grant numbers 01KI2006D, 01KI20328A, 01KX2021), the Ministry for Science and Culture of Lower Saxony (grant numbers 14-76103-184, MWK HZI COVID-19), and the German Research Foundation (DFG; grant numbers PO 716/11-1, PO 716/14-1). MSW received unrestricted funding from Sartorius, Lung research. H-MJ received funding from BMBF (grant numbers 01KI2043, NaFoUniMedCovid19-COVIM 01KX2021), Bavarian State Ministry for Science and the Arts, and DFG through the research training groups RTG1660 and TRR130, the Bayerische Forschungsstiftung (Project CORAd), and the Kastner Foundation. GMNB acknowledges funding from the German Center for Infection Research (grant number 80018019238) and a European Regional Development Fund (Defeat Corona, grant number ZW7-8515131, together with AD-J). All other authors declare no competing interests.
- Published
- 2022
- Full Text
- View/download PDF
32. Augmented neutralization of SARS-CoV-2 Omicron variant by boost vaccination and monoclonal antibodies.
- Author
-
Schulz SR, Hoffmann M, Roth E, Pracht K, Burnett DL, Mazigi O, Schuh W, Manger B, Mielenz D, Goodnow CC, Christ D, Pöhlmann S, and Jäck HM
- Subjects
- Antibodies, Monoclonal, Antibodies, Neutralizing, Antibodies, Viral, BNT162 Vaccine, COVID-19 Vaccines, Humans, SARS-CoV-2, Vaccination, Antineoplastic Agents, Immunological, COVID-19
- Abstract
Effective vaccines and monoclonal antibodies have been developed against coronavirus disease 2019 (COVID-19) caused by severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2). However, the appearance of virus variants with higher transmissibility and pathogenicity is a major concern because of their potential to escape vaccines and clinically approved SARS-CoV-2- antibodies. Here, we use flow cytometry-based binding and pseudotyped SARS-CoV-2 neutralization assays to determine the efficacy of boost immunization and therapeutic antibodies to neutralize the dominant Omicron variant. We provide compelling evidence that the third vaccination with BNT162b2 increases the amount of neutralizing serum antibodies against Delta and Omicron variants, albeit to a lower degree when compared to the parental Wuhan strain. Therefore, a third vaccination is warranted to increase titers of protective serum antibodies, especially in the case of the Omicron variant. We also found that most clinically approved and otherwise potent therapeutic antibodies against the Delta variant failed to recognize and neutralize the Omicron variant. In contrast, some antibodies under preclinical development potentially neutralized the Omicron variant. Our studies also support using a flow cytometry-based antibody binding assay to rapidly monitor therapeutic candidates and serum titers against emerging SARS-CoV-2 variants., (© 2022 The Authors. European Journal of Immunology published by Wiley-VCH GmbH.)
- Published
- 2022
- Full Text
- View/download PDF
33. Platform for isolation and characterization of SARS-CoV-2 variants enables rapid characterization of Omicron in Australia.
- Author
-
Aggarwal A, Stella AO, Walker G, Akerman A, Esneau C, Milogiannakis V, Burnett DL, McAllery S, Silva MR, Lu Y, Foster CSP, Brilot F, Pillay A, Van Hal S, Mathivanan V, Fichter C, Kindinger A, Hoppe AC, Munier ML, Amatayakul-Chantler S, Roth N, Coppola G, Symonds GP, Schofield P, Jackson J, Lenthall H, Henry JY, Mazigi O, Jäck HM, Davenport MP, Darley DR, Matthews GV, Khoury DS, Cromer D, Goodnow CC, Christ D, Robosa R, Starck DJ, Bartlett NW, Rawlinson WD, Kelleher AD, and Turville SG
- Subjects
- Australia, Humans, Pandemics, COVID-19 diagnosis, SARS-CoV-2 genetics
- Abstract
Genetically distinct variants of severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) have emerged since the start of the COVID-19 pandemic. Over this period, we developed a rapid platform (R-20) for viral isolation and characterization using primary remnant diagnostic swabs. This, combined with quarantine testing and genomics surveillance, enabled the rapid isolation and characterization of all major SARS-CoV-2 variants circulating in Australia in 2021. Our platform facilitated viral variant isolation, rapid resolution of variant fitness using nasopharyngeal swabs and ranking of evasion of neutralizing antibodies. In late 2021, variant of concern Omicron (B1.1.529) emerged. Using our platform, we detected and characterized SARS-CoV-2 VOC Omicron. We show that Omicron effectively evades neutralization antibodies and has a different entry route that is TMPRSS2-independent. Our low-cost platform is available to all and can detect all variants of SARS-CoV-2 studied so far, with the main limitation being that our platform still requires appropriate biocontainment., (© 2022. The Author(s).)
- Published
- 2022
- Full Text
- View/download PDF
34. Inter-domain communication in SARS-CoV-2 spike proteins controls protease-triggered cell entry.
- Author
-
Qing E, Li P, Cooper L, Schulz S, Jäck HM, Rong L, Perlman S, and Gallagher T
- Subjects
- Communication, Humans, Peptide Hydrolases, SARS-CoV-2, Virus Internalization, COVID-19, Spike Glycoprotein, Coronavirus
- Abstract
SARS-CoV-2 continues to evolve into variants of concern (VOC), with greatest variability in the multidomain, entry-facilitating spike proteins. To recognize the significance of adaptive spike protein changes, we compare variant SARS-CoV-2 virus particles in several assays reflecting authentic virus-cell entry. Virus particles with adaptive changes in spike amino-terminal domains (NTDs) are hypersensitive to proteolytic activation of membrane fusion, an essential step in virus-cell entry. Proteolysis is within fusion domains (FDs), at sites over 10 nm from the VOC-specific NTD changes, indicating allosteric inter-domain control of fusion activation. In addition, NTD-specific antibodies block FD cleavage, membrane fusion, and virus-cell entry, suggesting restriction of inter-domain communication as a neutralization mechanism. Finally, using structure-guided mutagenesis, we identify an inter-monomer β sheet structure that facilitates NTD-to-FD transmissions and subsequent fusion activation. This NTD-to-FD axis that sensitizes viruses to infection and to NTD-specific antibody neutralization provides new context for understanding selective forces driving SARS-CoV-2 evolution., Competing Interests: Declaration of interests The authors declare no competing interests., (Copyright © 2022 The Authors. Published by Elsevier Inc. All rights reserved.)
- Published
- 2022
- Full Text
- View/download PDF
35. SARS-CoV-2 variants C.1.2 and B.1.621 (Mu) partially evade neutralization by antibodies elicited upon infection or vaccination.
- Author
-
Arora P, Kempf A, Nehlmeier I, Graichen L, Winkler MS, Lier M, Schulz S, Jäck HM, Cossmann A, Stankov MV, Behrens GMN, Pöhlmann S, and Hoffmann M
- Subjects
- Antibodies, Monoclonal, Humanized, Antibodies, Neutralizing, Antibodies, Viral, COVID-19 Vaccines, Humans, Spike Glycoprotein, Coronavirus, Vaccination, COVID-19, SARS-CoV-2
- Abstract
Rapid spread of SARS-CoV-2 variants C.1.2 and B.1.621 (Mu variant) in Africa and the Americas, respectively, as well as a high number of mutations in the viral spike proteins raised concerns that these variants might pose an elevated threat to human health. Here, we show that C.1.2 and B.1.621 spike proteins mediate increased entry into certain cell lines but do not exhibit increased ACE2 binding. Further, we demonstrate that C.1.2 and B.1.621 are resistant to neutralization by bamlanivimab but remain sensitive to inhibition by antibody cocktails used for COVID-19 therapy. Finally, we show that C.1.2 and B.1.621 partially escape neutralization by antibodies induced upon infection and vaccination, with escape of vaccine-induced antibodies being as potent as that measured for B.1.351 (Beta variant), which is known to be highly neutralization resistant. Collectively, C.1.2 and B.1.621 partially evade control by vaccine-induced antibodies, suggesting that close monitoring of these variants is warranted., Competing Interests: Declaration of interests M.S.W. received unrestricted funding for independent research projects from Sartorius., (Copyright © 2022 The Author(s). Published by Elsevier Inc. All rights reserved.)
- Published
- 2022
- Full Text
- View/download PDF
36. 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
37. 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
38. No evidence for increased cell entry or antibody evasion by Delta sublineage AY.4.2.
- Author
-
Arora P, Kempf A, Nehlmeier I, Graichen L, Winkler MS, Lier M, Schulz S, Jäck HM, Pöhlmann S, and Hoffmann M
- Subjects
- Humans, Mutation immunology, Neutralization Tests methods, Spike Glycoprotein, Coronavirus immunology, Virus Internalization, Antibodies, Neutralizing immunology, Antibodies, Viral immunology, COVID-19 immunology, SARS-CoV-2 immunology
- Abstract
Since the beginning of the COVID-19 pandemic, multiple SARS-CoV-2 variants have emerged. While some variants spread only locally, others, referred to as variants of concern, disseminated globally and became drivers of the pandemic. All SARS-CoV-2 variants harbor mutations relative to the virus circulating early in the pandemic, and mutations in the viral spike (S) protein are considered of particular relevance since the S protein mediates host cell entry and constitutes the key target of the neutralizing antibody response. As a consequence, mutations in the S protein may increase SARS-CoV-2 infectivity and enable its evasion of neutralizing antibodies. Furthermore, mutations in the S protein can modulate viral transmissibility and pathogenicity., (© 2021. The Author(s).)
- Published
- 2022
- Full Text
- View/download PDF
39. The Omicron variant is highly resistant against antibody-mediated neutralization: Implications for control of the COVID-19 pandemic.
- Author
-
Hoffmann M, Krüger N, Schulz S, Cossmann A, Rocha C, Kempf A, Nehlmeier I, Graichen L, Moldenhauer AS, Winkler MS, Lier M, Dopfer-Jablonka A, Jäck HM, Behrens GMN, and Pöhlmann S
- Subjects
- Adaptive Immunity, Angiotensin-Converting Enzyme 2 metabolism, Animals, Antibodies, Neutralizing pharmacology, Antibodies, Viral immunology, BNT162 Vaccine immunology, COVID-19 prevention & control, COVID-19 Vaccines immunology, Cell Line, Chlorocebus aethiops, Female, Humans, Male, Protein Binding, SARS-CoV-2 chemistry, Spike Glycoprotein, Coronavirus chemistry, Spike Glycoprotein, Coronavirus metabolism, Vaccination, Vero Cells, Antibodies, Monoclonal, Humanized pharmacology, Antibodies, Neutralizing immunology, COVID-19 immunology, COVID-19 virology, SARS-CoV-2 drug effects, SARS-CoV-2 immunology, Spike Glycoprotein, Coronavirus immunology
- Abstract
The rapid spread of the SARS-CoV-2 Omicron variant suggests that the virus might become globally dominant. Further, the high number of mutations in the viral spike protein raised concerns that the virus might evade antibodies induced by infection or vaccination. Here, we report that the Omicron spike was resistant against most therapeutic antibodies but remained susceptible to inhibition by sotrovimab. Similarly, the Omicron spike evaded neutralization by antibodies from convalescent patients or individuals vaccinated with the BioNTech-Pfizer vaccine (BNT162b2) with 12- to 44-fold higher efficiency than the spike of the Delta variant. Neutralization of the Omicron spike by antibodies induced upon heterologous ChAdOx1 (Astra Zeneca-Oxford)/BNT162b2 vaccination or vaccination with three doses of BNT162b2 was more efficient, but the Omicron spike still evaded neutralization more efficiently than the Delta spike. These findings indicate that most therapeutic antibodies will be ineffective against the Omicron variant and that double immunization with BNT162b2 might not adequately protect against severe disease induced by this variant., Competing Interests: Declaration of interests The authors declare no competing interests., (Copyright © 2021 Elsevier Inc. All rights reserved.)
- Published
- 2022
- Full Text
- View/download PDF
40. Single-cell resolution of plasma cell fate programming in health and disease.
- Author
-
Delaloy C, Schuh W, Jäck HM, Bonaud A, and Espéli M
- Subjects
- Animals, Humans, Cell Differentiation genetics, Cell Differentiation immunology, Chromatin Immunoprecipitation Sequencing, Immune System Diseases genetics, Immune System Diseases immunology, Plasma Cells immunology, RNA-Seq, Single-Cell Analysis
- Abstract
Long considered a homogeneous population dedicated to antibody secretion, plasma cell phenotypic and functional heterogeneity is increasingly recognized. Plasma cells were first segregated based on their maturation level, but the complexity of this subset might well be underestimated by this simple dichotomy. Indeed, in the last decade new functions have been attributed to plasma cells including but not limited to cytokine secretion. However, a proper characterization of plasma cell heterogeneity has remained elusive partly due to technical issues and cellular features that are specific to this cell type. Cell intrinsic and cell extrinsic signals could be at the origin of this heterogeneity. Recent advances in technologies such as single cell RNA-seq, ATAC-seq, or ChIP-seq on low cell numbers helped to elucidate the fate decision in other cell lineages and similar approaches could be implemented to evaluate the heterogeneous fate of activated B cells in health and disease. Here, we summarized published work shedding some lights on the stimuli and genetic program shaping B-cell terminal differentiation at the single cell level in mice and men. We also discuss the fate and heterogeneity of plasma cells during immune responses, vaccination, and in the frame of human plasma cell disorders., (© 2021 Wiley-VCH GmbH.)
- Published
- 2022
- Full Text
- View/download PDF
41. Immunizations with diverse sarbecovirus receptor-binding domains elicit SARS-CoV-2 neutralizing antibodies against a conserved site of vulnerability.
- Author
-
Burnett DL, Jackson KJL, Langley DB, Aggrawal A, Stella AO, Johansen MD, Balachandran H, Lenthall H, Rouet R, Walker G, Saunders BM, Singh M, Li H, Henry JY, Jackson J, Stewart AG, Witthauer F, Spence MA, Hansbro NG, Jackson C, Schofield P, Milthorpe C, Martinello M, Schulz SR, Roth E, Kelleher A, Emery S, Britton WJ, Rawlinson WD, Karl R, Schäfer S, Winkler TH, Brink R, Bull RA, Hansbro PM, Jäck HM, Turville S, Christ D, and Goodnow CC
- Subjects
- Animals, Antibodies, Neutralizing metabolism, Antibodies, Viral metabolism, Conserved Sequence genetics, Evolution, Molecular, Humans, Immunization, Mice, Mice, Inbred BALB C, Mice, Inbred C57BL, Protein Binding, Protein Domains genetics, Spike Glycoprotein, Coronavirus genetics, Spike Glycoprotein, Coronavirus immunology, Vaccine Development, Betacoronavirus physiology, COVID-19 Vaccines immunology, Coronavirus Infections immunology, Severe acute respiratory syndrome-related coronavirus physiology, Spike Glycoprotein, Coronavirus metabolism
- Abstract
Viral mutations are an emerging concern in reducing SARS-CoV-2 vaccination efficacy. Second-generation vaccines will need to elicit neutralizing antibodies against sites that are evolutionarily conserved across the sarbecovirus subgenus. Here, we immunized mice containing a human antibody repertoire with diverse sarbecovirus receptor-binding domains (RBDs) to identify antibodies targeting conserved sites of vulnerability. Antibodies with broad reactivity against diverse clade B RBDs targeting the conserved class 4 epitope, with recurring IGHV/IGKV pairs, were readily elicited but were non-neutralizing. However, rare class 4 antibodies binding this conserved RBD supersite showed potent neutralization of SARS-CoV-2 and all variants of concern. Structural analysis revealed that the neutralizing ability of cross-reactive antibodies was reserved only for those with an elongated CDRH3 that extends the antiparallel beta-sheet RBD core and orients the antibody light chain to obstruct ACE2-RBD interactions. These results identify a structurally defined pathway for vaccine strategies eliciting escape-resistant SARS-CoV-2 neutralizing antibodies., Competing Interests: Declaration of interests All affiliations are listed on the title page of the manuscript. The authors and their immediate family members have no financial interests to declare. The authors have lodged a patent (Australian provisional patent application no. 2021901716) based on this work., (Copyright © 2021 The Author(s). Published by Elsevier Inc. All rights reserved.)
- Published
- 2021
- Full Text
- View/download PDF
42. Endothelial dysfunction contributes to severe COVID-19 in combination with dysregulated lymphocyte responses and cytokine networks.
- Author
-
Ruhl L, Pink I, Kühne JF, Beushausen K, Keil J, Christoph S, Sauer A, Boblitz L, Schmidt J, David S, Jäck HM, Roth E, Cornberg M, Schulz TF, Welte T, Höper MM, and Falk CS
- Subjects
- Biomarkers blood, C-Reactive Protein metabolism, COVID-19 immunology, COVID-19 mortality, COVID-19 virology, Chemokine CXCL10 blood, Chemokine CXCL9 blood, Cluster Analysis, Convalescence, Cytokine Release Syndrome immunology, Cytokine Release Syndrome mortality, Cytokine Release Syndrome virology, Disease Progression, Endothelium, Vascular immunology, Granulocytes immunology, Granulocytes virology, Hematopoietic Cell Growth Factors blood, Hepatocyte Growth Factor blood, Humans, Intensive Care Units, Interleukin-12 Subunit p40 blood, Interleukin-6 blood, Interleukin-8 blood, Killer Cells, Natural immunology, Killer Cells, Natural virology, Lectins, C-Type blood, Lymphopenia immunology, Lymphopenia mortality, Lymphopenia virology, Plasma Cells immunology, Plasma Cells virology, Survival Analysis, T-Lymphocytes immunology, T-Lymphocytes virology, Antibodies, Viral blood, Blood Proteins metabolism, COVID-19 diagnosis, Cytokine Release Syndrome diagnosis, Endothelium, Vascular virology, Lymphopenia diagnosis, SARS-CoV-2 pathogenicity
- Abstract
The systemic processes involved in the manifestation of life-threatening COVID-19 and in disease recovery are still incompletely understood, despite investigations focusing on the dysregulation of immune responses after SARS-CoV-2 infection. To define hallmarks of severe COVID-19 in acute disease (n = 58) and in disease recovery in convalescent patients (n = 28) from Hannover Medical School, we used flow cytometry and proteomics data with unsupervised clustering analyses. In our observational study, we combined analyses of immune cells and cytokine/chemokine networks with endothelial activation and injury. ICU patients displayed an altered immune signature with prolonged lymphopenia but the expansion of granulocytes and plasmablasts along with activated and terminally differentiated T and NK cells and high levels of SARS-CoV-2-specific antibodies. The core signature of seven plasma proteins revealed a highly inflammatory microenvironment in addition to endothelial injury in severe COVID-19. Changes within this signature were associated with either disease progression or recovery. In summary, our data suggest that besides a strong inflammatory response, severe COVID-19 is driven by endothelial activation and barrier disruption, whereby recovery depends on the regeneration of the endothelial integrity., (© 2021. The Author(s).)
- Published
- 2021
- Full Text
- View/download PDF
43. The spike protein of SARS-CoV-2 variant A.30 is heavily mutated and evades vaccine-induced antibodies with high efficiency.
- Author
-
Arora P, Rocha C, Kempf A, Nehlmeier I, Graichen L, Winkler MS, Lier M, Schulz S, Jäck HM, Cossmann A, Stankov MV, Behrens GMN, Pöhlmann S, and Hoffmann M
- Subjects
- Antibodies, Neutralizing immunology, Cell Line, Humans, SARS-CoV-2 immunology, Spike Glycoprotein, Coronavirus immunology, Virus Internalization, Antibodies, Viral immunology, COVID-19 Vaccines immunology, Mutation, SARS-CoV-2 genetics, Spike Glycoprotein, Coronavirus genetics
- Published
- 2021
- Full Text
- View/download PDF
44. Guidelines for the use of flow cytometry and cell sorting in immunological studies (third edition).
- Author
-
Cossarizza A, Chang HD, Radbruch A, Abrignani S, Addo R, Akdis M, Andrä I, Andreata F, Annunziato F, Arranz E, Bacher P, Bari S, Barnaba V, Barros-Martins J, Baumjohann D, Beccaria CG, Bernardo D, Boardman DA, Borger J, Böttcher C, Brockmann L, Burns M, Busch DH, Cameron G, Cammarata I, Cassotta A, Chang Y, Chirdo FG, Christakou E, Čičin-Šain L, Cook L, Corbett AJ, Cornelis R, Cosmi L, Davey MS, De Biasi S, De Simone G, Del Zotto G, Delacher M, Di Rosa F, Di Santo J, Diefenbach A, Dong J, Dörner T, Dress RJ, Dutertre CA, Eckle SBG, Eede P, Evrard M, Falk CS, Feuerer M, Fillatreau S, Fiz-Lopez A, Follo M, Foulds GA, Fröbel J, Gagliani N, Galletti G, Gangaev A, Garbi N, Garrote JA, Geginat J, Gherardin NA, Gibellini L, Ginhoux F, Godfrey DI, Gruarin P, Haftmann C, Hansmann L, Harpur CM, Hayday AC, Heine G, Hernández DC, Herrmann M, Hoelsken O, Huang Q, Huber S, Huber JE, Huehn J, Hundemer M, Hwang WYK, Iannacone M, Ivison SM, Jäck HM, Jani PK, Keller B, Kessler N, Ketelaars S, Knop L, Knopf J, Koay HF, Kobow K, Kriegsmann K, Kristyanto H, Krueger A, Kuehne JF, Kunze-Schumacher H, Kvistborg P, Kwok I, Latorre D, Lenz D, Levings MK, Lino AC, Liotta F, Long HM, Lugli E, MacDonald KN, Maggi L, Maini MK, Mair F, Manta C, Manz RA, Mashreghi MF, Mazzoni A, McCluskey J, Mei HE, Melchers F, Melzer S, Mielenz D, Monin L, Moretta L, Multhoff G, Muñoz LE, Muñoz-Ruiz M, Muscate F, Natalini A, Neumann K, Ng LG, Niedobitek A, Niemz J, Almeida LN, Notarbartolo S, Ostendorf L, Pallett LJ, Patel AA, Percin GI, Peruzzi G, Pinti M, Pockley AG, Pracht K, Prinz I, Pujol-Autonell I, Pulvirenti N, Quatrini L, Quinn KM, Radbruch H, Rhys H, Rodrigo MB, Romagnani C, Saggau C, Sakaguchi S, Sallusto F, Sanderink L, Sandrock I, Schauer C, Scheffold A, Scherer HU, Schiemann M, Schildberg FA, Schober K, Schoen J, Schuh W, Schüler T, Schulz AR, Schulz S, Schulze J, Simonetti S, Singh J, Sitnik KM, Stark R, Starossom S, Stehle C, Szelinski F, Tan L, Tarnok A, Tornack J, Tree TIM, van Beek JJP, van de Veen W, van Gisbergen K, Vasco C, Verheyden NA, von Borstel A, Ward-Hartstonge KA, Warnatz K, Waskow C, Wiedemann A, Wilharm A, Wing J, Wirz O, Wittner J, Yang JHM, and Yang J
- Subjects
- Animals, Chronic Disease, Humans, Mice, Practice Guidelines as Topic, Autoimmune Diseases immunology, Flow Cytometry, Infections immunology, Neoplasms immunology
- Abstract
The third edition of Flow Cytometry Guidelines provides the key aspects to consider when performing flow cytometry experiments and includes comprehensive sections describing phenotypes and functional assays of all major human and murine immune cell subsets. Notably, the Guidelines contain helpful tables highlighting phenotypes and key differences between human and murine cells. Another useful feature of this edition is the flow cytometry analysis of clinical samples with examples of flow cytometry applications in the context of autoimmune diseases, cancers as well as acute and chronic infectious diseases. Furthermore, there are sections detailing tips, tricks and pitfalls to avoid. All sections are written and peer-reviewed by leading flow cytometry experts and immunologists, making this edition an essential and state-of-the-art handbook for basic and clinical researchers., (© 2021 Wiley-VCH GmbH.)
- Published
- 2021
- Full Text
- View/download PDF
45. Delta variant (B.1.617.2) sublineages do not show increased neutralization resistance.
- Author
-
Arora P, Kempf A, Nehlmeier I, Graichen L, Sidarovich A, Winkler MS, Schulz S, Jäck HM, Stankov MV, Behrens GMN, Pöhlmann S, and Hoffmann M
- Subjects
- Animals, COVID-19 Vaccines, Chlorocebus aethiops, Databases, Genetic, Humans, Mutation, Spike Glycoprotein, Coronavirus immunology, T-Lymphocytes immunology, T-Lymphocytes virology, Vero Cells, Vietnam, Antibodies, Neutralizing chemistry, COVID-19 virology, Neutralization Tests, SARS-CoV-2 genetics, SARS-CoV-2 physiology
- Published
- 2021
- Full Text
- View/download PDF
46. A surrogate cell-based SARS-CoV-2 spike blocking assay.
- Author
-
Schuh W, Baus L, Steinmetz T, Schulz SR, Weckwerth L, Roth E, Hauke M, Krause S, Morhart P, Rauh M, Hoffmann M, Vesper N, Reth M, Schneider H, Jäck HM, and Mielenz D
- Subjects
- Antibodies, Blocking immunology, Antibodies, Neutralizing immunology, COVID-19 immunology, Humans, SARS-CoV-2, Spike Glycoprotein, Coronavirus immunology, Antibodies, Blocking blood, Antibodies, Neutralizing blood, Antibodies, Viral analysis, COVID-19 diagnosis, COVID-19 Serological Testing methods, Flow Cytometry methods
- Abstract
To monitor infection by the severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) and successful vaccination against coronavirus disease 2019 (COVID-19), the kinetics of neutralizing or blocking anti-SARS-CoV-2 antibody titers need to be assessed. Here, we report the development of a quick and inexpensive surrogate SARS-CoV-2 blocking assay (SUBA) using immobilized recombinant human angiotensin-converting enzyme 2 (hACE2) and human cells expressing the native form of surface SARS-CoV-2 spike protein. Spike protein-expressing cells bound to hACE2 in the absence or presence of blocking antibodies were quantified by measuring the optical density of cell-associated crystal violet in a spectrophotometer. The advantages are that SUBA is a fast and inexpensive assay, which does not require biosafety level 2- or 3-approved laboratories. Most importantly, SUBA detects blocking antibodies against the native trimeric cell-bound SARS-CoV-2 spike protein and can be rapidly adjusted to quickly pre-screen already approved therapeutic antibodies or sera from vaccinated individuals for their ACE2 blocking activities against any emerging SARS-CoV-2 variants., (© 2021 The Authors. European Journal of Immunology published by Wiley-VCH GmbH.)
- Published
- 2021
- Full Text
- View/download PDF
47. B.1.617.2 enters and fuses lung cells with increased efficiency and evades antibodies induced by infection and vaccination.
- Author
-
Arora P, Sidarovich A, Krüger N, Kempf A, Nehlmeier I, Graichen L, Moldenhauer AS, Winkler MS, Schulz S, Jäck HM, Stankov MV, Behrens GMN, Pöhlmann S, and Hoffmann M
- Subjects
- Adult, Antibodies, Monoclonal immunology, Antibodies, Neutralizing immunology, Antibodies, Viral immunology, BNT162 Vaccine, COVID-19 metabolism, COVID-19 therapy, COVID-19 Vaccines immunology, Cell Fusion, Cell Line, Female, HEK293 Cells, Humans, Immune Evasion physiology, Immunization, Passive methods, Lung pathology, Lung virology, Male, Middle Aged, Neutralization Tests, SARS-CoV-2 metabolism, SARS-CoV-2 pathogenicity, Spike Glycoprotein, Coronavirus immunology, Vaccination methods, COVID-19 Serotherapy, COVID-19 immunology, Immune Evasion immunology, SARS-CoV-2 immunology
- Abstract
The Delta variant of severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2), B.1.617.2, emerged in India and has spread to over 80 countries. B.1.617.2 replaced B.1.1.7 as the dominant virus in the United Kingdom, resulting in a steep increase in new infections, and a similar development is expected for other countries. Effective countermeasures require information on susceptibility of B.1.617.2 to control by antibodies elicited by vaccines and used for coronavirus disease 2019 (COVID-19) therapy. We show, using pseudotyping, that B.1.617.2 evades control by antibodies induced upon infection and BNT162b2 vaccination, although to a lesser extent as compared to B.1.351. We find that B.1.617.2 is resistant against bamlanivimab, a monoclonal antibody with emergency use authorization for COVID-19 therapy. Finally, we show increased Calu-3 lung cell entry and enhanced cell-to-cell fusion of B.1.617.2, which may contribute to augmented transmissibility and pathogenicity of this variant. These results identify B.1.617.2 as an immune evasion variant with increased capacity to enter and fuse lung cells., Competing Interests: Declaration of interests The authors declare no competing interests., (Copyright © 2021 The Author(s). Published by Elsevier Inc. All rights reserved.)
- Published
- 2021
- Full Text
- View/download PDF
48. A Barcoded Flow Cytometric Assay to Explore the Antibody Responses Against SARS-CoV-2 Spike and Its Variants.
- Author
-
Vesper N, Ortiz Y, Bartels-Burgahn F, Yang J, de la Rosa K, Tenbusch M, Schulz S, Finzel S, Jäck HM, Eibel H, Voll RE, and Reth M
- Subjects
- Angiotensin-Converting Enzyme 2 metabolism, Antibodies, Viral metabolism, Antibody Formation, Female, Humans, Immunization, Secondary, Immunoglobulin A metabolism, Immunoglobulin G metabolism, Male, Middle Aged, Mutation genetics, Spike Glycoprotein, Coronavirus genetics, Vaccination, COVID-19 immunology, Cell Separation methods, Flow Cytometry methods, SARS-CoV-2 physiology, Spike Glycoprotein, Coronavirus metabolism
- Abstract
The SARS-CoV-2 pandemic has spread to all parts of the world and can cause life-threatening pneumonia and other severe disease manifestations known as COVID-19. This health crisis has resulted in a significant effort to stop the spread of this new coronavirus. However, while propagating itself in the human population, the virus accumulates mutations and generates new variants with increased fitness and the ability to escape the human immune response. Here we describe a color-based barcoded spike flow cytometric assay (BSFA) that is particularly useful to evaluate and directly compare the humoral immune response directed against either wild type (WT) or mutant spike (S) proteins or the receptor-binding domains (RBD) of SARS-CoV-2. This assay employs the human B lymphoma cell line Ramos, transfected for stable expression of WT or mutant S proteins or a chimeric RBD-CD8 fusion protein. We find that the alpha and beta mutants are more stably expressed than the WT S protein on the Ramos B cell surface and/or bind with higher affinity to the viral entry receptor ACE2. However, we find a reduce expression of the chimeric RBD-CD8 carrying the point mutation N501Y and E484K characteristic for the alpha and beta variant, respectively. The comparison of the humoral immune response of 12 vaccinated probands with 12 COVID-19 patients shows that after the boost, the S-specific IgG class immune response in the vaccinated group is similar to that of the patient group. However, in comparison to WT the specific IgG serum antibodies bind less well to the alpha variant and only poorly to the beta variant S protein. This is in line with the notion that the beta variant is an immune escape variant of SARS-CoV-2. The IgA class immune response was more variable than the IgG response and higher in the COVID-19 patients than in the vaccinated group. In summary, we think that our BSFA represents a useful tool to evaluate the humoral immunity against emerging variants of SARS-CoV-2 and to analyze new vaccination protocols against these variants., 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 © 2021 Vesper, Ortiz, Bartels-Burgahn, Yang, de la Rosa, Tenbusch, Schulz, Finzel, Jäck, Eibel, Voll and Reth.)
- Published
- 2021
- Full Text
- View/download PDF
49. Increased risk of chronic fatigue and hair loss following COVID-19 in individuals with hypohidrotic ectodermal dysplasia.
- Author
-
Hennig V, Schuh W, Neubert A, Mielenz D, Jäck HM, and Schneider H
- Subjects
- Adolescent, Adult, Alopecia, Child, Humans, Longitudinal Studies, Middle Aged, SARS-CoV-2, Spike Glycoprotein, Coronavirus, Young Adult, COVID-19, Ectodermal Dysplasia 1, Anhidrotic, Fatigue Syndrome, Chronic
- Abstract
Background: Hypohidrotic ectodermal dysplasia (HED) is a group of genodermatoses in which deficient ectodysplasin A signalling leads to maldevelopment of skin appendages, various eccrine glands, and teeth. Individuals with HED often have disrupted epithelial barriers and, therefore, were suspected to be more susceptible to coronavirus infection., Methods: 56 households with at least one member who had coronavirus disease of 2019 (COVID-19) were enrolled in a longitudinal study to compare the course of illness, immune responses, and long-term consequences of severe acute respiratory syndrome-coronavirus 2 (SARS-CoV-2) infection in HED patients (n = 15, age 9-52 years) and control subjects of the same age group (n = 149)., Results: In 14 HED patients, mild or moderate typical COVID-19 symptoms were observed that lasted for 4-45 days. Fever during the first days sometimes required external cooling measures. The course of COVID-19 was similar to that in control subjects if patients developed antibodies blocking the SARS-CoV-2 spike protein. Five out of six HED patients with completely abrogated ectodysplasin A signalling (83%) suffered from chronic, in two cases very severe fatigue following COVID-19, while only 25% of HED patients with residual activity of this pathway and 21% of control subjects recovering from COVID-19 experienced postinfectious fatigue. Hair loss after COVID-19 was also more frequent among HED patients (64%) than in the control group (13%)., Conclusions: HED appears to be associated with an increased risk of long-term consequences of a SARS-CoV-2 infection. Preventive vaccination against COVID-19 should be recommended for individuals affected by this rare genetic disorder., (© 2021. The Author(s).)
- Published
- 2021
- Full Text
- View/download PDF
50. TFG is required for autophagy flux and to prevent endoplasmic reticulum stress in CH12 B lymphoma cells.
- Author
-
Steinmetz TD, Schlötzer-Schrehardt U, Hearne A, Schuh W, Wittner J, Schulz SR, Winkler TH, Jäck HM, and Mielenz D
- Subjects
- Adaptor Proteins, Signal Transducing metabolism, Animals, Autophagosomes metabolism, Cell Line, Tumor, Endoplasmic Reticulum metabolism, Mice, Autophagy, Endoplasmic Reticulum Stress, Lymphoma, B-Cell, Proteins metabolism
- Abstract
Plasma cells depend on quality control of newly synthesized antibodies in the endoplasmic reticulum (ER) via macroautophagy/autophagy and proteasomal degradation. The cytosolic adaptor protein TFG (Trk-fused gene) regulates ER-Golgi transport, the secretory pathway and proteasome activity in non-immune cells. We show here that TFG is upregulated during lipopolysaccharide- and CpG-induced differentiation of B1 and B2 B cells into plasmablasts, with the highest expression of TFG in mature plasma cells. CRISPR-CAS9-mediated gene disruption of tfg in the B lymphoma cell line CH12 revealed increased apoptosis, which was reverted by BCL2 but even more by ectopic TFG expression. Loss of TFG disrupted ER structure, leading to an expanded ER and increased expression of ER stress genes. When compared to wild-type CH12 cells, tfg KO CH12 cells were more sensitive toward ER stress induced by tunicamycin, monensin and proteasome inhibition or by expression of an ER-bound immunoglobulin (Ig) μ heavy (µH) chain. CH12 tfg KO B cells displayed more total LC3, lower LC3-II turnover and increased numbers and size of autophagosomes. Tandem-fluorescent-LC3 revealed less accumulation of GFP-LC3 in starved and chloroquine-treated CH12 tfg KO B cells. The GFP:RFP ratio of tandem-fluorescent-LC3 was higher in tunicamycin-treated CH12 tfg KO B cells, suggesting less autophagy flux during induced ER stress. Based on these data, we suggest that TFG controls autophagy flux in CH12 B cells and propose that TFG is a survival factor that alleviates ER stress through the support of autophagy flux in activated B cells and mature plasma cells. Abbreviations : Ab, antibody; Ag, antigen; ASC, antibody-secreting cells; ATG, autophagy-related; BCR, B cell receptor; COPII, coat protein complex II; CpG, non-methylated CpG oligonucleotide; ER, endoplasmic reticulum; ERAD, ER-associated degradation; FO, follicular; GFP, green fluorescent protein; HC, heavy chain; Ig, immunoglobulin; IRES, internal ribosomal entry site; LC, light chain; MZ, marginal zone; NFKB, nuclear factor of kappa light polypeptide gene enhancer in B cells; TLR, toll-like receptor; UPR, unfolded protein response.
- Published
- 2021
- Full Text
- View/download PDF
Catalog
Discovery Service for Jio Institute Digital Library
For full access to our library's resources, please sign in.