1
|
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, Jäck H. A pair of noncompeting neutralizing human monoclonal antibodies protecting from disease in a SARS-CoV-2 infection model. Eur J Immunol 2022; 52:770-783. [PMID: 34355795 PMCID: PMC8420377 DOI: 10.1002/eji.202149374] [Citation(s) in RCA: 20] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2021] [Revised: 07/05/2021] [Accepted: 08/03/2021] [Indexed: 11/18/2022]
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.
Collapse
|
2
|
IMU-838, a Developmental DHODH Inhibitor in Phase II for Autoimmune Disease, Shows Anti-SARS-CoV-2 and Broad-Spectrum Antiviral Efficacy In Vitro. Viruses 2020; 12:v12121394. [PMID: 33291455 PMCID: PMC7762174 DOI: 10.3390/v12121394] [Citation(s) in RCA: 31] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2020] [Revised: 12/01/2020] [Accepted: 12/02/2020] [Indexed: 12/17/2022] Open
Abstract
The ongoing pandemic spread of the severe acute respiratory syndrome coronavirus type 2 (SARS-CoV-2) demands skillful strategies for novel drug development, drug repurposing and cotreatments, in particular focusing on existing candidates of host-directed antivirals (HDAs). The developmental drug IMU-838, currently being investigated in a phase 2b trial in patients suffering from autoimmune diseases, represents an inhibitor of human dihydroorotate dehydrogenase (DHODH) with a recently proven antiviral activity in vitro and in vivo. Here, we established an analysis system for assessing the antiviral potency of IMU-838 and DHODH-directed back-up drugs in cultured cell-based infection models. By the use of SARS-CoV-2-specific immunofluorescence, Western blot, in-cell ELISA, viral yield reduction and RT-qPCR methods, we demonstrated the following: (i) IMU-838 and back-ups show anti-SARS-CoV-2 activity at several levels of viral replication, i.e., protein production, double-strand RNA synthesis, and release of infectious virus; (ii) antiviral efficacy in Vero cells was demonstrated in a micromolar range (IMU-838 half-maximal effective concentration, EC50, of 7.6 ± 5.8 µM); (iii) anti-SARS-CoV-2 activity was distinct from cytotoxic effects (half-cytotoxic concentration, CC50, >100 µM); (iv) the drug in vitro potency was confirmed using several Vero lineages and human cells; (v) combination with remdesivir showed enhanced anti-SARS-CoV-2 activity; (vi) vidofludimus, the active determinant of IMU-838, exerted a broad-spectrum activity against a selection of major human pathogenic viruses. These findings strongly suggest that developmental DHODH inhibitors represent promising candidates for use as anti-SARS-CoV-2 therapeutics.
Collapse
|
3
|
Brachs S, Polack J, Brachs M, Jahn-Hofmann K, Elvert R, Pfenninger A, Bärenz F, Margerie D, Mai K, Spranger J, Kannt A. Genetic Nicotinamide N-Methyltransferase ( Nnmt) Deficiency in Male Mice Improves Insulin Sensitivity in Diet-Induced Obesity but Does Not Affect Glucose Tolerance. Diabetes 2019; 68:527-542. [PMID: 30552109 DOI: 10.2337/db18-0780] [Citation(s) in RCA: 42] [Impact Index Per Article: 8.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/29/2018] [Accepted: 12/03/2018] [Indexed: 11/13/2022]
Abstract
Antisense oligonucleotide knockdown (ASO-KD) of nicotinamide N-methyltransferase (NNMT) in high-fat diet (HFD)-fed mice has been reported to reduce weight gain and plasma insulin levels and to improve glucose tolerance. Using NNMT-ASO-KD or NNMT knockout mice (NNMT-/-), we tested the hypothesis that Nnmt deletion protects against diet-induced obesity and its metabolic consequences in males and females on obesity-inducing diets. We also examined samples from a human weight reduction (WR) study for adipose NNMT (aNNMT) expression and plasma 1-methylnicotinamide (MNAM) levels. In Western diet (WD)-fed female mice, NNMT-ASO-KD reduced body weight, fat mass, and insulin level and improved glucose tolerance. Although NNMT-/- mice fed a standard diet had no obvious phenotype, NNMT-/- males fed an HFD showed strongly improved insulin sensitivity (IS). Furthermore, NNMT-/- females fed a WD showed reduced weight gain, less fat, and lower insulin levels. However, no improved glucose tolerance was observed in NNMT-/- mice. Although NNMT expression in human fat biopsy samples increased during WR, corresponding plasma MNAM levels significantly declined, suggesting that other mechanisms besides aNNMT expression modulate circulating MNAM levels during WR. In summary, upon NNMT deletion or knockdown in males and females fed different obesity-inducing diets, we observed sex- and diet-specific differences in body composition, weight, and glucose tolerance and estimates of IS.
Collapse
Affiliation(s)
- Sebastian Brachs
- Department of Endocrinology and Metabolism, Charité-Universitätsmedizin Berlin, Berlin, Germany
- DZHK (German Centre for Cardiovascular Research), partner site Berlin, Germany
- Center for Cardiovascular Research (CCR), Charité-Universitätsmedizin Berlin, Berlin, Germany
| | - James Polack
- Department of Endocrinology and Metabolism, Charité-Universitätsmedizin Berlin, Berlin, Germany
- DZHK (German Centre for Cardiovascular Research), partner site Berlin, Germany
- Center for Cardiovascular Research (CCR), Charité-Universitätsmedizin Berlin, Berlin, Germany
| | - Maria Brachs
- Department of Endocrinology and Metabolism, Charité-Universitätsmedizin Berlin, Berlin, Germany
- Center for Cardiovascular Research (CCR), Charité-Universitätsmedizin Berlin, Berlin, Germany
| | | | - Ralf Elvert
- Sanofi Research and Development, Frankfurt am Main, Germany
| | | | - Felix Bärenz
- Sanofi Research and Development, Frankfurt am Main, Germany
| | | | - Knut Mai
- Department of Endocrinology and Metabolism, Charité-Universitätsmedizin Berlin, Berlin, Germany
- DZHK (German Centre for Cardiovascular Research), partner site Berlin, Germany
- Center for Cardiovascular Research (CCR), Charité-Universitätsmedizin Berlin, Berlin, Germany
- Clinical Research Unit, Berlin Institute of Health (BIH), Berlin, Germany
| | - Joachim Spranger
- Department of Endocrinology and Metabolism, Charité-Universitätsmedizin Berlin, Berlin, Germany
- DZHK (German Centre for Cardiovascular Research), partner site Berlin, Germany
- Center for Cardiovascular Research (CCR), Charité-Universitätsmedizin Berlin, Berlin, Germany
- Clinical Research Unit, Berlin Institute of Health (BIH), Berlin, Germany
| | - Aimo Kannt
- Sanofi Research and Development, Frankfurt am Main, Germany
- Institute of Experimental and Clinical Pharmacology and Toxicology, Medical Faculty Mannheim, Heidelberg University, Mannheim, Germany
| |
Collapse
|
4
|
Fürnrohr BG, Mielenz D. Quantification of Human Swiprosin-1/EFhd2 Expression on Protein and RNA Level. Methods Mol Biol 2019; 1929:595-605. [PMID: 30710299 DOI: 10.1007/978-1-4939-9030-6_37] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022]
Abstract
Many Ca2+-binding proteins are differentially regulated under pro-inflammatory conditions in different organs. Specific quantification of RNA and protein expression of those proteins demands validated protocols. Peripheral blood mononuclear cells (PBMC) can mirror an inflammatory status originating from several organs and can therefore be an important diagnostic tool. Swiprosin-1/EFhd2 (EFhd2) is a ~30 kDa Ca2+ and F-actin binding, cytoskeletal protein with two central EF hands and a C-terminal coiled-coil domain. Unbiased gene expression analyses and proteomics revealed that EFhd2 is regulated under pro-inflammatory conditions in several cell types and tissues. Here we describe validated protocols to quantify the expression of the human orthologue of Swiprosin-1/EFhd2 on RNA and protein level in PBMC. Both methods reveal that EFhd2 is stronger expressed in monocytes than in B cells of healthy donors. Thus, initial experiments relying on qPCR are likely to provide results with functional relevance. The higher expression of EFhd2 in monocytes could be related to monocyte migration under inflammatory conditions.
Collapse
Affiliation(s)
- Barbara G Fürnrohr
- Division of Molecular immunology, Department of Internal Medicine III, Nikolaus-Fiebiger-Center, University of Erlangen-Nürnberg, Erlangen, Germany
| | - Dirk Mielenz
- Division of Molecular immunology, Department of Internal Medicine III, Nikolaus-Fiebiger-Center, University of Erlangen-Nürnberg, Erlangen, Germany.
| |
Collapse
|
5
|
Physiological and pathophysiological functions of Swiprosin-1/EFhd2 in the nervous system. Biochem J 2017; 473:2429-37. [PMID: 27515255 DOI: 10.1042/bcj20160168] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2016] [Accepted: 05/03/2016] [Indexed: 12/15/2022]
Abstract
Synaptic dysfunction and dysregulation of Ca(2+) are linked to neurodegenerative processes and behavioural disorders. Our understanding of the causes and factors involved in behavioural disorders and neurodegeneration, especially Alzheimer's disease (AD), a tau-related disease, is on the one hand limited and on the other hand controversial. Here, we review recent data about the links between the Ca(2+)-binding EF-hand-containing cytoskeletal protein Swiprosin-1/EFhd2 and neurodegeneration. Specifically, we summarize the functional biochemical data obtained in vitro with the use of recombinant EFhd2 protein, and integrated them with in vivo data in order to interpret the emerging role of EFhd2 in synaptic plasticity and in the pathophysiology of neurodegenerative disorders, particularly involving the tauopathies. We also discuss its functions in actin remodelling through cofilin and small GTPases, thereby linking EFhd2, synapses and the actin cytoskeleton. Expression data and functional experiments in mice and in humans have led to the hypothesis that down-regulation of EFhd2, especially in the cortex, is involved in dementia.
Collapse
|
6
|
Stein M, Dütting S, Mougiakakos D, Bösl M, Fritsch K, Reimer D, Urbanczyk S, Steinmetz T, Schuh W, Bozec A, Winkler TH, Jäck HM, Mielenz D. A defined metabolic state in pre B cells governs B-cell development and is counterbalanced by Swiprosin-2/EFhd1. Cell Death Differ 2017; 24:1239-1252. [PMID: 28524857 DOI: 10.1038/cdd.2017.52] [Citation(s) in RCA: 43] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2016] [Revised: 02/10/2017] [Accepted: 03/08/2017] [Indexed: 12/14/2022] Open
Abstract
B-cell development in the bone marrow comprises proliferative and resting phases in different niches. We asked whether B-cell metabolism relates to these changes. Compared to pro B and small pre B cells, large pre B cells revealed the highest glucose uptake and ROS but not mitochondrial mass, whereas small pre B cells exhibited the lowest mitochondrial membrane potential. Small pre B cells from Rag1-/-;33.C9 μ heavy chain knock-in mice revealed decreased glycolysis (ECAR) and mitochondrial spare capacity compared to pro B cells from Rag1-/- mice. We were interested in the step regulating this metabolic switch from pro to pre B cells and uncovered that Swiprosin-2/EFhd1, a Ca2+-binding protein of the inner mitochondrial membrane involved in Ca2+-induced mitoflashes, is expressed in pro B cells, but downregulated by surface pre B-cell receptor expression. Knockdown and knockout of EFhd1 in 38B9 pro B cells decreased the oxidative phosphorylation/glycolysis (OCR/ECAR) ratio by increasing glycolysis, glycolytic capacity and reserve. Prolonged expression of EFhd1 in EFhd1 transgenic mice beyond the pro B cell stage increased expression of the mitochondrial co-activator PGC-1α in primary pre B cells, but reduced mitochondrial ATP production at the pro to pre B cell transition in IL-7 cultures. Transgenic EFhd1 expression caused a B-cell intrinsic developmental disadvantage for pro and pre B cells. Hence, coordinated expression of EFhd1 in pro B cells and by the pre BCR regulates metabolic changes and pro/pre B-cell development.
Collapse
Affiliation(s)
- Merle Stein
- Department of Internal Medicine 3, Division of Molecular Immunology, Nikolaus-Fiebiger-Center, Friedrich-Alexander-University of Erlangen-Nuremberg, Erlangen, Germany
| | - Sebastian Dütting
- Department of Internal Medicine 3, Division of Molecular Immunology, Nikolaus-Fiebiger-Center, Friedrich-Alexander-University of Erlangen-Nuremberg, Erlangen, Germany
| | - Dimitrios Mougiakakos
- Department of Internal Medicine V, University Clinic, Friedrich-Alexander-University of Erlangen-Nuremberg, Erlangen, Germany
| | - Michael Bösl
- Department of Experimental Biomedicine, University Hospital and Rudolf Virchow Center, University of Würzburg, Würzburg D-97080, Germany
| | - Kristin Fritsch
- Department of Internal Medicine 3, Division of Molecular Immunology, Nikolaus-Fiebiger-Center, Friedrich-Alexander-University of Erlangen-Nuremberg, Erlangen, Germany
| | - Dorothea Reimer
- Department of Internal Medicine 3, Division of Molecular Immunology, Nikolaus-Fiebiger-Center, Friedrich-Alexander-University of Erlangen-Nuremberg, Erlangen, Germany
| | - Sophia Urbanczyk
- Department of Internal Medicine 3, Division of Molecular Immunology, Nikolaus-Fiebiger-Center, Friedrich-Alexander-University of Erlangen-Nuremberg, Erlangen, Germany
| | - Tobit Steinmetz
- Department of Internal Medicine 3, Division of Molecular Immunology, Nikolaus-Fiebiger-Center, Friedrich-Alexander-University of Erlangen-Nuremberg, Erlangen, Germany
| | - Wolfgang Schuh
- Department of Internal Medicine 3, Division of Molecular Immunology, Nikolaus-Fiebiger-Center, Friedrich-Alexander-University of Erlangen-Nuremberg, Erlangen, Germany
| | - Aline Bozec
- Department of Internal Medicine III, University Clinic, Friedrich-Alexander-University of Erlangen-Nuremberg, Erlangen, Germany
| | - Thomas H Winkler
- Department of Biology, Nikolaus-Fiebiger-Center, Friedrich-Alexander-University of Erlangen-Nuremberg, Erlangen, Germany
| | - Hans-Martin Jäck
- Department of Internal Medicine 3, Division of Molecular Immunology, Nikolaus-Fiebiger-Center, Friedrich-Alexander-University of Erlangen-Nuremberg, Erlangen, Germany
| | - Dirk Mielenz
- Department of Internal Medicine 3, Division of Molecular Immunology, Nikolaus-Fiebiger-Center, Friedrich-Alexander-University of Erlangen-Nuremberg, Erlangen, Germany
| |
Collapse
|
7
|
Wang ZB, Han P, Tong LC, Luo Y, Su WH, Wei X, Yu XH, Liu WY, Zhang XH, Lei H, Li ZZ, Wang F, Chen JG, Ma TH, Su DF, Li L. Low level of swiprosin-1/EFhd2 in vestibular nuclei of spontaneously hypersensitive motion sickness mice. Sci Rep 2017; 7:40986. [PMID: 28128226 PMCID: PMC5269593 DOI: 10.1038/srep40986] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2016] [Accepted: 12/14/2016] [Indexed: 11/13/2022] Open
Abstract
Susceptibility to motion sickness (MS) varies considerably among humans. However, the cause of such variation is unclear. Here, we used a classical genetic approach to obtain mouse strains highly sensitive and resistant to MS (SMS and RMS). Proteomics analysis revealed substantially lower swiprosin-1 expression in SMS mouse brains. Inducing MS via rotary stimulation decreased swiprosin-1 in the mouse brains. Swiprosin-1 knockout mice were much more sensitive to motion disturbance. Immunohistochemistry revealed strong swiprosin-1 expression in the vestibular nuclei (VN). Over-expressing swiprosin-1 in the VN of SMS mice decreased MS susceptibility. Down-regulating swiprosin-1 in the VN of RMS mice by RNAi increased MS susceptibility. Additional in vivo experiments revealed decreased swiprosin-1 expression by glutamate via the NMDA receptor. Glutamate increased neuronal excitability in SMS or swiprosin-1 knockout mice more prominently than in RMS or wild-type mice. These results indicate that swiprosin-1 in the VN is a critical determinant of the susceptibility to MS.
Collapse
Affiliation(s)
- Zhi-Bin Wang
- Department of Pharmacology, College of Pharmacy, Second Military Medical University, Shanghai 200433, China
| | - Ping Han
- Department of Pharmacology, College of Pharmacy, Second Military Medical University, Shanghai 200433, China
| | - Ling-Chang Tong
- Department of Pharmacology, College of Pharmacy, Second Military Medical University, Shanghai 200433, China
| | - Yi Luo
- Department of Pharmacology, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430030, China
| | - Wei-Heng Su
- Basal medical College, Dalian Medical University, Dalian, Liaoning 130041, China
| | - Xin Wei
- Department of Pharmacology, College of Pharmacy, Second Military Medical University, Shanghai 200433, China
| | - Xu-Hong Yu
- Department of Pharmacology, College of Pharmacy, Second Military Medical University, Shanghai 200433, China
| | - Wei-Ye Liu
- Department of Pharmacology, College of Pharmacy, Second Military Medical University, Shanghai 200433, China
| | - Xiu-Hua Zhang
- Department of Pharmacology, College of Pharmacy, Second Military Medical University, Shanghai 200433, China
| | - Hong Lei
- Department of Pharmacology, College of Pharmacy, Second Military Medical University, Shanghai 200433, China
| | - Zhen-Zhen Li
- Department of Pharmacology, College of Pharmacy, Second Military Medical University, Shanghai 200433, China
| | - Fang Wang
- Department of Pharmacology, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430030, China
| | - Jian-Guo Chen
- Department of Pharmacology, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430030, China
| | - Tong-Hui Ma
- Basal medical College, Dalian Medical University, Dalian, Liaoning 130041, China
| | - Ding-Feng Su
- Department of Pharmacology, College of Pharmacy, Second Military Medical University, Shanghai 200433, China
| | - Ling Li
- Department of Pharmacology, College of Pharmacy, Second Military Medical University, Shanghai 200433, China
| |
Collapse
|
8
|
The Ca2+ sensor protein swiprosin-1/EFhd2 is present in neurites and involved in kinesin-mediated transport in neurons. PLoS One 2014; 9:e103976. [PMID: 25133820 PMCID: PMC4136728 DOI: 10.1371/journal.pone.0103976] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2014] [Accepted: 07/08/2014] [Indexed: 01/10/2023] Open
Abstract
Swiprosin-1/EFhd2 (EFhd2) is a cytoskeletal Ca2+ sensor protein strongly expressed in the brain. It has been shown to interact with mutant tau, which can promote neurodegeneration, but nothing is known about the physiological function of EFhd2 in the nervous system. To elucidate this question, we analyzed EFhd2−/−/lacZ reporter mice and showed that lacZ was strongly expressed in the cortex, the dentate gyrus, the CA1 and CA2 regions of the hippocampus, the thalamus, and the olfactory bulb. Immunohistochemistry and western blotting confirmed this pattern and revealed expression of EFhd2 during neuronal maturation. In cortical neurons, EFhd2 was detected in neurites marked by MAP2 and co-localized with pre- and post-synaptic markers. Approximately one third of EFhd2 associated with a biochemically isolated synaptosome preparation. There, EFhd2 was mostly confined to the cytosolic and plasma membrane fractions. Both synaptic endocytosis and exocytosis in primary hippocampal EFhd2−/− neurons were unaltered but transport of synaptophysin-GFP containing vesicles was enhanced in EFhd2−/− primary hippocampal neurons, and notably, EFhd2 inhibited kinesin mediated microtubule gliding. Therefore, we found that EFhd2 is a neuronal protein that interferes with kinesin-mediated transport.
Collapse
|