Daniel Sauter’s research while affiliated with University of Tübingen and other places

What is this page?


This page lists works of an author who doesn't have a ResearchGate profile or hasn't added the works to their profile yet. It is automatically generated from public (personal) data to further our legitimate goal of comprehensive and accurate scientific recordkeeping. If you are this author and want this page removed, please let us know.

Publications (222)


High‐throughput screening for endogenous inhibitors of SARS‐CoV‐2 entry. A) Caco‐2 cells were pre‐treated with peptide/protein containing fractions of a hemofiltrate library before transduction with luciferase‐encoding lentiviral pseudoparticles harboring the SARS‐CoV‐2 Hu‐1 spike. Transduction rates were assessed 48 h later by measuring luciferase activity in cell lysates. Columns represent pseudoparticle entry (expressed as % versus H2O‐treated condition) and black line absorbance at 280 nm of the corresponding fraction. The serine protease inhibitor camostat mesylate (CM) was included as a reference inhibitor. Hit fraction 19 was subpurified and screened again (Figure S1, Supporting Information). Shown are the mean values of one experiment performed in triplicates ± SEM. CM = Camostat mesylate. # indicates cellular toxicity as observed by light microscopy B) Schematic representation of serine protease inhibitor Bikunin with protease inhibitor domains from the kunitz‐type. C,D) Caco‐2 cells were pre‐treated with serial dilution of compounds before transduction with luciferase‐encoding lentiviral pseudoparticles harboring the SARS‐CoV‐2 Hu‐1 spike or VSV‐G glycoprotein. Transduction rates were assessed 48 h later by measuring luciferase activity in cell lysates. Shown are the mean values of three independent experiments performed in triplicates ± SEM.
Trypstatin inhibits TMPRSS2 and related proteases. A,C,D,G–J) Serial dilutions of Trypstatin, serine protease inhibitor camostat mesylate, kunitz inhibitor aprotinin, and cysteine protease inhibitor E64‐d were mixed with the respective enzyme before the addition of a fluorogenic reporter substrate. Fluorescence intensity was measured at an excitation wavelength of 380 nm and an emission wavelength of 460 nm. B,E,F) HEK293T‐cells were transfected with TMPRSS2‐, TMPRSS11D‐ and TMPRSS13‐encoding plasmid or mock control, respectively, before treatment with serial dilutions of inhibitors and the addition of a fluorogenic reporter substrate. Values were corrected for the signal of mock‐transfected HEK293T‐cells. Shown are the mean values of three independent experiments performed in triplicates ± SEM. K) Heat‐map summarizing the IC50 values of A‐J. L) Binding affinity prediction between protease inhibitors and TMPRSS2 with PPI‐Affinity. The complexes are representative structures obtained by molecular dynamics simulations. Zoom: S1 pocket of TMPRSS2. Hydrogen bonds are shown in light blue, salt bridges in orange. The catalytic residues (H296, D345, and S441) are depicted in Corey–Pauling–Koltun representation.
Trypstatin inhibits SARS‐CoV‐2 in vitro and in vivo. A) Caco‐2 cells were pre‐treated with serial dilution of compounds before transduction with luciferase‐encoding lentiviral pseudoparticles harboring the SARS‐CoV‐2 Hu‐1 spike. Transduction rates were assessed 48 h later by measuring luciferase activity in cell lysates. Shown is the mean of three independent experiments performed in triplicates ± SEM. B) Caco‐2 cells were treated with indicated compounds for 30 min before inoculation with SARS‐CoV‐2 B.1 D614G (MOI 0.0005) or the Omicron BA.1 variant (MOI 0.2). Nucleocapsid protein‐positive cells were quantified one day post‐infection by flow cytometry. Shown are the mean values of two (BA.1) or three (B.1) experiments performed in triplicates ± SEM. C) Calu‐3 cells were treated with Trypstatin (10 µm), camostat (80 µm), or PBS right before (pre) or 3 hours post (post) infection with SARS‐CoV‐2 BA.5 at a MOI of 0.001. Two hours later, the inoculum was removed, cells were washed and fresh medium supplemented with respective compounds was added. Complete supernatants were sampled at 2, 24, 48, and 72 h postinfection (hpi) and fresh medium with the compound was added after sampling. RT‐qPCR targeting SARS‐CoV‐2 ORF1b nsp14 was performed on RNA isolated from harvested supernatants. D) Cumulative SARS‐CoV‐2 RNA copies until 3 dpi assessed by area under the curve analysis from (C). E) The apical site of human airway epithelial cells (HAEC) grown at the air–liquid interface was exposed to PBS, Trypstatin (10 µm), or camostat mesylate (10 µm) before inoculation with SARS‐CoV‐2 Omicron BA. 5 (MOI 0.5) for 2 h before apical washing and further culturing at the air–liquid interface. Two days‐post infection, mucus was washed off and PBS was added to the apical side for 30 min before the sample was subjected to TCID50 titration. Shown are TCID50/ml values of pooled apical washes from two cultures per donor. F) Syrian gold hamsters were infected with SARS‐CoV‐2 (Delta) and treated with Trypstatin or PBS. Hamsters were sacrificed 72 h post infection and bronchoalveolar lavage was collected for each animal. BAL samples were subjected to RT‐qPCR and TCID50 analysis. Mann–Whitney‐U‐Test was applied to test for significance. G) Hamsters were monitored daily for clinical symptoms.
Trypstatin is a broad‐spectrum inhibitor of corona‐ and influenza viruses. A,B) Caco‐2 cells were treated with serial dilutions of compound right before transduction with lentiviral pseudoparticles harboring the SARS‐CoV‐1‐ or MERS‐CoV spike. Transduction rates were assessed 48 h later by measuring luciferase activity in cell lysates. C) Caco‐2 cells were treated with serial dilutions of Trypstatin or control inhibitors before infection with the hCoV‐NL63 (MOI 0.018) virus. Nucleocapsid protein‐positive cells were quantified three days post‐infection by flow cytometry. D–F) Calu‐3 cells were exposed to serial dilution of compounds for 30 min before inoculation with 100xTCID50 per well of respective influenza virus. Three days post‐infection, nucleocapsid‐positive cells were quantified by immunostaining and high‐content imaging. Shown are the mean values of three independent experiments performed in triplicates ± SEM for all graphs. Tryp = Trypstatin, CM = Camostat mesylate, BA = Baloxavir acid.
Trypstatin as a Novel TMPRSS2 Inhibitor with Broad‐Spectrum Efficacy against Corona and Influenza Viruses
  • Article
  • Full-text available

May 2025

·

33 Reads

·

Lukas Wettstein

·

·

[...]

·

Respiratory viruses, such as SARS‐CoV‐2 and influenza, exploit host proteases like TMPRSS2 for entry, making TMPRSS2 a prime antiviral target. Here, the identification and characterization of Trypstatin, a 61‐amino acid Kunitz‐type protease inhibitor derived from human hemofiltrate are reported. Trypstatin inhibits TMPRSS2 and related proteases with high potency, exhibiting half‐maximal inhibitory concentration values in the nanomolar range, comparable to the small molecule inhibitor camostat mesylate. In vitro assays demonstrate that Trypstatin effectively blocks spike‐driven entry of SARS‐CoV‐2, SARS‐CoV‐1, MERS‐CoV, and hCoV‐NL63, as well as hemagglutinin‐mediated entry of influenza A and B viruses. In primary human airway epithelial cultures, Trypstatin significantly reduces SARS‐CoV‐2 replication and retained activity in the presence of airway mucus. In vivo, intranasal administration of Trypstatin to SARS‐CoV‐2‐infected Syrian hamsters reduces viral titers and alleviates clinical symptoms. These findings highlight Trypstatin's potential as a broad‐spectrum antiviral agent against TMPRSS2‐dependent respiratory viruses.

Download

Fig. 1. Antiviral proteins inhibit Syncytin-mediated cell fusion. (A) heK293t split-GFP assay. two subclones of heK293t cells express one part of GFP each. Upon Syncytin overexpression, the cells fuse and GFP is complemented. (B) heK239t split-GFP cells were cotransfected with Syncytin and antiviral protein. An empty vector was used to adjust total dnA amounts. Fusion was monitored by quantifying GFP fluorescence for 48 hours. Means ± Sd, n = 4 to 8. (C and D) Average n fold changes of the 10 most strongly up-regulated genes in placentas from patients with FGR or severe Pe compared to healthy controls (n = 8 per group), and the genes analyzed in Fig. 1B were derived from (24). (E) GBP5 protein levels in BeWo cells 72 hours after valproate stimulation. (F) BeWo split-GFP reporter assay. Upon forskolin stimulation, the cells fuse and GFP is complemented. (G) BeWo cell fusion upon valproate stimulation. Means ± Sd, n = 4. (H) GBP5 expression upon iFn-γ stimulation (100 ng/ml) in BeWo cells, quantified by quantitative PcR. Means ± SeM, n = 3. (I) BeWo cell fusion upon iFn-γ stimulation (100 ng/ml). Means ± Sd, n = 4. (J) BeWo cell fusion after GBP2/5 overexpression. BeWo split-GFP cells were transduced with GBP2/5 and treated with forskolin, and GFP fluorescence was monitored for 72 hours. Means ± Sd, n = 3 to 4. (K) cell fusion after GBP5 depletion. BeWo split-GFP cells were transduced with cRiSPR/cas lentiviruses expressing GBP5-targeting single guide RnA (sgRnA) or a negative control. three days later, cells were stimulated with iFn-γ, before fusion was induced by forskolin 2 days later. GFP fluorescence was monitored for 44 hours. Means ± SeM are shown on top, n = 3. One representative Western blot is shown at the bottom. A one-way analysis of variance (AnOvA) with multiple comparisons (dunnett's test) was performed for (G), (J), and (K). A paired t test was performed for (i) (*P < 0.05, **P < 0.01, ***P < 0.001, and ****P < 0.0001). AUc, area under the curve; dMSO, dimethyl sulfoxide; h, hours.
Inhibition of placental trophoblast fusion by guanylate-binding protein 5

May 2025

·

33 Reads

Science Advances

Syncytin-1 and Syncytin-2 are envelope glycoproteins encoded by human endogenous retroviruses that have been exapted for the fusion of cytotrophoblast cells into syncytiotrophoblasts during placental development. Pregnancy complications like preeclampsia are associated with altered expression of interferon-stimulated genes, including guanylate-binding protein 5 (GBP5). Here, we show that misdirected antiviral activity of GBP5 impairs processing and activation of Syncytin-1. In contrast, the proteolytic activation of Syncytin-2 is not affected by GBP5, and its fusogenic activity is only modestly reduced. Mechanistic analyses revealed that Syncytin-1 is mainly cleaved by the GBP5 target furin, whereas Syncytin-2 is also efficiently processed by the proprotein convertase subtilisin/kexin type 7 (PCSK7) and thus resistant to GBP5-mediated restriction. Mutational analyses mapped PCSK7 processing of Syncytin-2 to a leucine residue upstream of the polybasic cleavage site. In summary, we identified an innate immune mechanism that impairs the activity of a co-opted endogenous retroviral envelope protein during pregnancy and may potentially contribute to the pathogenesis of pregnancy disorders.


Fig. 2. Susceptibility of SIVmus to ZAP restriction. (A) Image of a mustached guenon (Cercopithecus cephus); pictures taken from Zoo Beauval in France, courtesy of Dr. Baptiste Mulot. (B) Genomic organization of the molecular clone of SIVmus-11Gab-Pts02(34). (C) HEK293T cells were transfected with SIVmus IMC or empty vector control. Supernatants obtained 2 d later were used to infect TZM-bl cells and β-galactosidase activities determined 3 d later. Data present the mean of three independent experiments each tested in triplicates (±SEM). RLU/s, relative light unit per second. Significant differences compared to control using the t test are indicated as ***P < 0.001. (D) Stimulated PBMCs and CD4+T cells from four different healthy human donors were infected with the indicated viruses including VSV-G pseudotyped SIVmus. Virus stocks were treated with EF-C, a fibrillar attachment factor, to enhance initial infection (37). Supernatants were harvested at 3 d intervals and examined for infectious virus production by TZM-bl reporter assay. Shown are mean values from the four donors (±SEM) each measured in duplicate. (E) HEK293T ZAP KO cells were cotransfected with increasing amounts of humZAP-L expression vector and indicated proviral constructs of HIV-1 NL4-3, HIV-1 NHG, and HIV-1 NHG L CpG high mutant. Infectious virus yields determined by TZM-bl assay are provided relative to those obtained in the absence of ZAP set as (100%). Shown are mean values from three to four independent experiments, each tested in technical triplicates (±SEM).
Fig. 3. Species-dependent differences in the expression of ZAP splice variants. (A) Domain organization of different ZAP isoforms: four N-terminal CCCH zinc finger RNA binding domains (orange boxes) and two central WWE domains (purple boxes) are shared by all four isoforms. The catalytically inactive C-terminal PARP domain (green box) is shared by ZAP-L and -XL. The ZAP-XL and M isoforms harbor an extended exon 4 (blue box). Domains are not drawn to scale. Primers used for ZAP amplification are indicated by blue and red arrows. (B) Amplification of ZAP genes from blood cells derived from the indicated primate species. (C) Schematic of the ZAP gene encompassing a total of 13 exons and generation of ZAP-L and ZAP-XL via usage of splice donor 4 (SD4) and 5 (SD5), respectively. (D) Splice donor usage for ZAP orthologs of the indicated primate species was predicted using SpliceRover (44). The respective scores are shown on the Right. Scores range from 0.0 to 1.0, with high scores indicating high probability of splice donor usage. The phylogenetic tree on the Left is based on timetree.org and Shao et al. 2023 (45). Cercopithecus spec. are highlighted in blue.
Fig. 4. Antiviral activity of primate ZAPs. (A) HEK293T ZAP KO cells were cotransfected with increasing amounts of vectors expressing the indicated N-terminally HA-tagged ZAP isoforms or empty vector control (CTRL) and analyzed by western blot. (B). Inhibition of HIV-1 and SIVmus by increasing amounts of ZAP orthologs, amplified from HUM, MUS, or MON blood or chemically synthesized ZAP genes. Stars indicate ZAP isoforms amplified from primate PBMCs, while their absence indicates synthesized isoforms. Shown are means of three independent experiments, each tested in duplicates (SIVmus) or triplicates (HIV-1), ± SEM. (C) Impact of ZAP on HIV-1 protein expression. HEK293T ZAP KO cells were cotransfected with 0.05 µg of indicated ZAP constructs and HIV-1 CH058 provirus. Whole-cell lysates and supernatants were analyzed for ZAP and viral proteins expression levels by western blot. Values shown under the blot represent normalized Env and p24 levels, adjusted to the internal control GAPDH and normalized to the no-ZAP condition (100%) (D) HEK293T ZAP KO cells were cotransfected with CH058 (Left panel) or SIVmus (Right panel) proviral constructs and increasing amounts of vectors expressing the ZAP-XL or ZAP-L isoforms. Infectious virus yield values were normalized to no ZAP set as (100%). ZAP orthologs amplified from primate blood are indicated with a star symbol. Shown are mean values (± SEM) obtained from three to eight independent experiments, each tested in triplicate.
Fig. 5. A combination of five mutations restores musZAP antiviral activity. (A) Schematic presentation of mutations in HUM and MUS ZAPs analyzed. Amino acid residues selected for functional analyses are highlighted in pink. Black lines indicate amino acid differences between HUM and MUS ZAPs. (B) Representative western blot of whole cell lysates showing expression levels of parental and mutant ZAPs. (C-E) HEK293T ZAP KO cells were cotransfected with proviral HIV-1 CH058 or SIVmus constructs and increasing amounts of vectors expressing the N-terminally HA-tagged isoforms of wt or mutant humZAP-L (C), musZAP-L (D), or mus ZAP-XL (E). Infectious virus yield was determined using TZM-bl assay. For each proviral construct, values were normalized to the infectious virus yield obtained in the absence of ZAP (100%). Shown are the mean (± SEM) of three to four independent experiments each performed in triplicates.
Fig. 6. CpG suppression and ZAP function in lemurs. (A) Suppression of CpGs dinucleotides in the genomes of the indicated lentiviruses (SI Appendix). (B) HEK293T ZAP KO cells were cotransfected with increasing amounts of indicated vectors expressing N-terminally HA-tagged ZAP-L proteins or empty vector control (CTRL) and analyzed by western blot for expression levels. (C) HEK293T ZAP KO cells were cotransfected with HIV-1 CH058 proviral construct and increasing amounts of vectors expressing HUM, MUS, or lemur (GML and CSL) ZAP-L isoforms. Infectious virus yields were normalized to the values obtained in the absence of ZAP set as (100%). Data are shown as mean of three to four independent experiments, each tested in triplicates, ± SEM. (D and E) Comparison of the frequency (D) and suppression (E) of CpG dinucleotides in 27 innate immune genes from the indicated primate species. Asterisks indicate statistical significance using Dunnett's multiple comparisons test ***P < 0.001; ****P < 0.0001.
Host ZAP activity correlates with the levels of CpG suppression in primate lentiviruses

April 2025

·

22 Reads

Proceedings of the National Academy of Sciences

Zinc-finger antiviral protein (ZAP) is thought to drive the suppression of CpG dinucleotides in many viruses to mimic the composition of their host genomes. However, in vivo evidence is sparse. Here, we investigated the reasons for unusually high CpG levels in SIVmus and SIVmon from mustached and mona monkeys, descendants of one of the precursors of HIV-1. We show that SIVmus is not resistant to ZAP inhibition. Instead, these Cercopithecus monkey hosts differ from other primate species by a splice site mutation and express the poorly active extralarge XL rather than the highly active L isoform of ZAP. Similarly, higher CpG levels in endogenous prosimian lentiviruses were associated with low activity of the corresponding host lemur ZAPs. In addition, lemur genes also show lower CpG suppression than other primates. Thus, the antiviral activity of ZAP not only affects suppression of CpG dinucleotides in viral transcripts but possibly also host genomes.


Antagonistic Regulation of LINE-1/Alu Elements and Their Repressor APOBEC3B in Cellular Senescence

March 2025

·

5 Reads

Long Interspersed Nuclear Elements-1 (LINE-1 or L1) make up approximately 21% of the human genome, with some L1 loci containing intact open reading frames (ORFs) that facilitate retrotransposition. Because retrotransposition can have deleterious effects leading to mutations and genomic instability, L1 activity is typically suppressed in somatic cells through transcriptional and post-transcriptional mechanisms. However, L1 elements are derepressed in senescent cells causing age-associated inflammation. Despite the recognition of L1 activity as a hallmark of aging, the underlying molecular mechanisms governing L1 derepression in these cells are not fully understood. In this study, we employed high throughput sequencing datasets and validated our findings through independent experiments to investigate the regulation of L1 elements in senescent cells. Our results reveal that both replicative and oncogene-induced senescence are associated with reduced expression of the cytidine deaminase APOBEC3B, a known suppressor of L1 retrotransposition. Consequently, senescent cells exhibited diminished levels of C-to-U editing of full-length L1 elements. Moreover, Ribo-seq profiling indicated that progression to senescence is not only associated with increased L1 transcription, but also translation of L1 ORFs. In summary, our results suggest that the depletion of APOBEC3B contributes to enhanced activity of L1 in senescent cells and promotion of L1-induced DNA damage and aging.


Evolution and sequence characteristics of LTR12 repeats. (A) Endogenous retroviruses are the result of retroviral integration events in the host genome that were ultimately fixed in the population. During evolution, the retroviral gag, pol, and env genes are frequently lost due to recombination events of the retroviral long terminal repeats (LTRs). In this case, only so-called solo-LTRs remain. Some solo-LTRs, including several LTR12 repeats, have retained their regulatory activity and may regulate the expression of host genes. (B) Arrangement of a prototypical LTR12 repeat in U3, R, and U5 regions, where the U3-R border marks the transcription start site (TSS). The U3 enhancer region is characterized by 5–17 tandem repeats. Each repeat has a length of about 41 nt and can be grouped into one of four closely related subtypes (1, 2, 3, or 4). These repeats comprise recurrent binding sites for transcription factors (TFBS), such as NF-Y, GATA-2, and MZF1. Like U3, the U5 region also varies in length due to varying numbers of 72 bp repeats.
Mechanisms of LTR12-mediated regulation of cellular gene expression. LTR12 repeats can act as (A) primary promoters, (B) alternative promoters, or (C) enhancers for cellular genes. Furthermore, some LTR12 loci code for regulatory RNAs, including (D) lncRNAs, (E) miRNA sponges, and (F) transcription factor decoys. Finally, splice donor or acceptor sites within LTR12 repeats could enable (G) alternative splicing of cellular genes.
Mechanisms of LTR12 regulation. At the pre-transcriptional level (top), LTR12 repeats are repressed by DNA marks (e.g., CpG methylation) and histone marks (e.g., H3K9 trimethylation). DNA methylation is associated with binding of the KRAB zinc finger protein ZNF676 and potentially piRNA/piwi complexes. The HUSH complex, including MPP8, TASOR, and periphilin contributes to LTR12 restriction by recruiting the histone methyltransferase SETDB1 for H3K9 trimethylation and stabilizing repressive chromatin. The adaptor protein TNRC18 is also involved in LTR12 restriction, recruiting HDACs to H3K9 marks. Finally, TE-derived antisense transcripts may suppress LTR12 transcription via a sequestration of transcription factors, such as NF-Y. At the post-transcriptional level (bottom), piRNA/piwi complexes may potentially induce the targeted degradation of LTR12-derived transcripts.
LTR12-regulated genes protect us from infections and cancer. Several LTR12 target genes are involved in the immune response to infection (left). These include genes encoding cytokines and their receptors (CXCL11 and CCR4), immune cell activators (SEMA4D), and components of the cell-intrinsic immune response against intracellular pathogens (IRGM, GBP2, and GBP5). Moreover, LTR12 repeats may exert anti-tumor activities by regulating proteins and RNAs involved in DNA repair (PRLH1), apoptosis (TNFRSF10B, p63, and DHRS2), and/or senescence (DHRS2).
Viral influencers: deciphering the role of endogenous retroviral LTR12 repeats in cellular gene expression

January 2025

·

49 Reads

·

2 Citations

The human genome is like a museum of ancient retroviral infections. It contains a large number of endogenous retroviruses (ERVs) that bear witness to past integration events. About 5,000 of them are so-called long terminal repeat 12 (LTR12) elements. Compared with 20,000 human genes, this is a remarkable number. Although LTR12 elements can act as promoters or enhancers of cellular genes, the function of most of these retroviral elements has remained unclear. In our mini-review, we show that different LTR12 elements share many similarities, including common transcription factor binding sites. Furthermore, we summarize novel insights into the epigenetic mechanisms governing their silencing and activation. Specific examples of genes and pathways that are regulated by LTR12 loci are used to illustrate the regulatory network built by these repetitive elements. A particular focus is on their role in the regulation of antiviral immune responses, tumor cell proliferation, and senescence. Finally, we describe how a targeted activation of this fascinating ERV family could be used for diagnostic or therapeutic purposes.


Reactivation of latent HIV-1 by the glucocorticoid receptor modulator AZD9567

January 2025

·

39 Reads

One key determinant of HIV-1 latency reversal is the activation of the viral long terminal repeat (LTR) by cellular transcription factors such as NF-κB and AP-1. Interestingly, the activity of these two transcription factors can be modulated by glucocorticoid receptors (GRs). Furthermore, the HIV-1 genome contains multiple binding sites for GRs. We therefore hypothesized that glucocorticoids and other GR modulators may influence HIV-1 latency and reactivation. To investigate how GR signaling affects latent HIV-1 reservoirs, we assembled a representative panel of GR modulators including natural steroidal agonists, selective and non-selective GR modulators, and clinically approved GR-modulating drugs. The effects of these compounds on HIV-1 reactivation were assessed using latently HIV-1-infected cell lines and primary cells, as well as reporter assays that monitored GR and LTR activities. We found that AZD9567 (Mizacorat), a non-steroidal partial GR agonist, reactivates latent HIV-1 in both lymphoid and myeloid cell lines and primary CD4+ T cells. Conversely, the GR antagonist mifepristone suppresses HIV-1 LTR-driven gene expression. Mechanistic analyses revealed that AZD9567-mediated reactivation partially depends on both GR and AP-1 binding sites in the LTR. In summary, we, here, identify the GR modulator AZD9567 as novel latency-reversing agent that activates LTR-driven gene expression, which may aid in advancing current shock-and-kill approaches in the treatment of HIV-1 infection. IMPORTANCE Latently infected cells of people living with HIV are constantly exposed to fluctuating levels of glucocorticoid hormones such as cortisol. In addition, many HIV-infected individuals regularly take corticosteroids as anti-inflammatory drugs. Although corticosteroids are known to affect the activity of the viral long terminal repeat (LTR) promoter and influence ongoing HIV-1 replication, relatively little is known about the effect of corticosteroid hormones and other glucocorticoid receptor (GR) modulators on latent HIV-1. By systematically comparing natural and synthetic GR modulators, we, here, identify a first first-in-class, oral, partial GR agonist that reactivates latent HIV-1 from different cell types. This drug, AZD9567, was previously tested in clinical trials for rheumatoid arthritis. Mutational analyses shed light on the underlying mode of action and revealed transcription factor binding sites in the HIV-1 LTR that determine responsiveness to AZD9567.


Polymorphism T746K enhances S protein cleavage. (A) Schematic illustration of the MERS-CoV S protein and mutations studied. SP, signal peptide; RBD, receptor-binding domain; FP, fusion peptide; HR, heptad repeat; TD, transmembrane domain; S1/S2 and S2′, cleavage sites. (B) T746K enhances MERS-S cleavage. Whole-cell lysates (WCL) of cells expressing the indicated S proteins (or no S protein, control; top left panel) or VSVpp bearing the indicated S proteins (or no S protein, control; top right panel) were subjected to SDS-PAGE, and S protein cleavage was analyzed using immunoblot (via C-terminal V5-epitope tag). Detection of beta-actin (ACTB) or VSV-M protein served as loading control. The immunoblot data are representative of five (WCL) or six (VSVpp) independent experiments. For quantification of S protein expression (bottom left panel), total S protein signals (S0 +S2) were normalized against the corresponding ACTB (WCL) or VSV-M (VSVpp) signals and subsequently compared to EMC (WT) S (set as 1). For quantification of S protein cleavage, total S protein signals (S0 +S2) were set as 100% and the proportion of S2 signals calculated (bottom right panel). Graphs show mean ± SEM of five (WCL) or six (VSVpp) independent experiments. Statistical significance was assessed by two-tailed, unpaired Student’s t-test with Welch correction (P > 0.05, not significant [ns]; *P ≤ 0.05; **P ≤ 0.01; ***P ≤ 0.001). (C) Peptides covering the MERS-S S1/S2 cleavage site with or without mutation T746K (Suc-TL[T/K]PRSVR-AMC), or a furin consensus sequence (Pyr-RTKR-AMC) were incubated with recombinant furin and the mean fluorescence intensity (MFI; excitation wave length: 355 nm; emission wave length: 460 nm) was recorded every 60 s for a total of 1 h, and background signals (addition of water instead of peptide) were subtracted. Graphs show average (mean) data ± SEM of four independent experiments, conducted with technical triplicates.
Mutation T746K does not augment cell–cell fusion. (A) A MERS-S mutant, in which the S1/S2 site was changed from RSVR to SSVR was included as control in the cell–cell fusion assay, since this mutant is known to exhibit reduced cell–cell fusion. (B) VSV particles bearing no spike, EMC WT, or mutant SSVR were concentrated, and expression of S protein and VSV-M in cell lysates was analyzed using immunoblot. Similar results were obtained in two separate experiments. (C) For analysis of S protein-driven cell-to-cell fusion, effector 293T cells transfected with plasmids encoding the indicated S proteins and β-galactosidase α-fragment were mixed with target 293T cells transfected to express DPP4 and the Ω-fragment of β-galactosidase. β-galactosidase activity in cell lysates was measured at the indicated time points post cell mixing and normalized against the assay background (effector cells expressing no S protein). The mean of three independent experiments with technical quadruplicates is shown; error bars indicate SEM. (D) Cell–cell fusion activity at 24 h post cell mixing is shown. Results obtained for MERS-S WT were set as 1. Statistical significance in panels (C) and (D) was assessed by two-way analysis of variance with Tukey’s multiple comparison test and two-tailed, unpaired Student’s t-test with Welch correction, respectively (P > 0.05, not significant [ns]; *P ≤ 0.05; **P ≤ 0.01; ***P ≤ 0.001).
Mutation T746K does not augment S protein-driven entry into Calu-3 lung cells. The indicated cell lines were inoculated with VSVpp bearing the indicated S proteins. S protein-driven cell entry was analyzed by quantification of virus-encoded luciferase activity in cell lysates. For normalization, cell entry of particles bearing EMC (WT) S was set as 1. Mean ± SEM of three independent experiments conducted with four technical replicates is shown. Statistical significance was assessed by two-tailed, unpaired Student’s t-test with Welch correction, respectively (P > 0.05, not significant [ns]; *P ≤ 0.05; **P ≤ 0.01; ***P ≤ 0.001).
Mutation T746K reduces DPP4 binding. (A) Spike scheme and location of mutation I529T. A spike protein harboring mutation I529T, which is known to reduce DPP4 binding, was included as control in the DPP4 binding assay. (B) To measure S protein binding to DPP4, 293T cells were transfected with expression plasmids for the indicated S proteins or empty vector and incubated with DPP4-Fc fusion protein and labeled antibody and subjected to flow cytometric analysis. Left panel: Mean fluorescence intensities were normalized to cells expressing no S protein. The average of three independent experiments conducted with unicates is shown; error bars indicate SEM. Right panel: Data were normalized against binding of MERS-S WT to DPP4, which was set as 1. Error bars indicate the SEM. Statistical significance was assessed by two-tailed, unpaired Student’s t-test with Welch correction (P > 0.05, not significant [ns]; *P ≤ 0.05; **P ≤ 0.01; ***P ≤ 0.001).
Optimization of the S1/S2 site increases S protein cleavage but not lung cell entry. (A) Spike scheme and location of mutations. The minimal furin motif at the S1/S2 site in MERS-S was either optimized (mutant S1/S2OPT), or the complete S1/S2 loop was exchanged against the corresponding sequences found in the hemagglutinin of H5N1 influenza A virus (mutant S1/S2H5) or the S protein of SARS-CoV-2 (mutant S1/S2SARS2). (B) Optimization of the furin motif at the S1/S2 site of MERS-S increased S protein cleavage. Whole-cell lysates (WCL) of cells expressing the indicated S proteins (or no S protein, control; left panel), or vesicular stomatitis virus pseudovirus particles (VSVpp) bearing the indicated S proteins (or no S protein, control; right panel) were subjected to SDS-PAGE. and S protein cleavage was analyzed using immunoblot (via C-terminal V5-epitope tag). Detection of beta-actin (ACTB) or VSV-M protein served as loading control. Representative immunoblot data of three independent experiments are shown. Bands corresponding to uncleaved (S0) and cleaved (S2) MERS-S are indicated. (C) For quantification of S protein expression (left panel), total S protein signals (S0 +S2) were normalized against the corresponding ACTB (WCL) or VSV-M (VSVpp) signals and subsequently compared against EMC (WT) S (set as 1). For quantification of S protein cleavage, total S protein signals (S0 +S2) were set as 100% and the proportion of S2 signals calculated (right panel). Graphs show the mean of three independent experiments. Error bars indicate SEM. (D) S protein binding to DPP4 was analyzed as described for panel B of Fig. 4. Left panel: The average of four independent experiments conducted with unicates is shown, error bars indicate SEM. Right panel: Data were normalized against binding of MERS-S WT to DPP4, which was set as 1. Error bars indicate the SEM. (E) The indicated cell lines were inoculated with VSVpp bearing the indicated S proteins. S protein-driven cell entry was analyzed by quantification of virus-encoded luciferase activity in cell lysates. For normalization, cell entry of particles bearing EMC (WT) S was set as 1. Mean ± SEM of three independent experiments conducted with four technical replicates are shown. Statistical significance in panels (C)–(E) was assessed by two-tailed, unpaired Student’s t-test with Welch correction (P > 0.05, not significant [ns]; *P ≤ 0.05; **P ≤ 0.01; ***P ≤ 0.001).
Acquisition of a multibasic cleavage site does not increase MERS-CoV entry into Calu-3 human lung cells

October 2024

·

53 Reads

Human-to-human transmission of the highly pathogenic Middle East respiratory syndrome coronavirus (MERS-CoV) is currently inefficient. However, there is concern that the virus might mutate and thereby increase its transmissibility and thus pandemic potential. The pandemic SARS-CoV-2 depends on a highly cleavable furin motif at the S1/S2 site of the viral spike (S) protein for efficient lung cell entry, transmission, and pathogenicity. Here, by employing pseudotyped particles, we investigated whether augmented cleavage at the S1/S2 site also increases MERS-CoV entry into Calu-3 human lung cells. We report that polymorphism T746K at the S1/S2 cleavage site or optimization of the furin motif increases S protein cleavage but not lung cell entry. These findings suggest that, unlike what has been reported for SARS-CoV-2, a highly cleavable S1/S2 site might not augment MERS-CoV infectivity for human lung cells. IMPORTANCE The highly cleavable furin motif in the spike protein is required for robust lung cell entry, transmission, and pathogenicity of SARS-CoV-2. In contrast, it is unknown whether optimization of the furin motif in the spike protein of the pre-pandemic MERS-CoV increases lung cell entry and allows for robust human–human transmission. The present study indicates that this might not be the case. Thus, neither a naturally occurring polymorphism that increased MERS-CoV spike protein cleavage nor artificial optimization of the cleavage site allowed for increased spike-protein-driven entry into Calu-3 human lung cells.


GBP5-mediated restriction of viral glycoprotein function. GBP5 reduces virion infectivity of various viral pathogens by inhibiting the (i) anterograde transport, (ii) N-linked glycosylation, and/or (iii) proteolytic processing of viral glycoproteins (GP). The host protease furin (green) and the oligosaccharyltransferase complex (OST, pink) have been described as targets of GBP5 (red), resulting in impaired proteolytic processing and glycosylation of viral glycoproteins (blue), respectively. The figure was generated using BioRender.
Antiviral mechanisms of guanylate-binding protein 5: versatile inhibition of multiple viral glycoproteins

October 2024

·

9 Reads

·

1 Citation

Guanylate-binding proteins (GBPs) are interferon-inducible cellular factors known to inhibit a wide variety of pathogens. Humans encode seven GBPs that have functionally diversified to provide broad protection against a variety of bacteria, protozoa, and viruses. Here, we discuss recent data on the mechanisms underlying the broad antiviral activity of GBP5 (H. Veler, C. M. Lun, A. A. Waheed, and E. O. Freed, mBio e02086-24, 2024, https://doi.org/10.1128/mbio.02086-24) and place them in the context of previous studies on the ability of this antiviral factor to impair the function of numerous viral envelope (Env) glycoproteins. We focus on the effects of GBP5 on the glycosylation, proteolytic processing, and anterograde transport of Env and discuss mechanistic interdependencies of these maturation steps. Understanding the induction and action of broadly acting immune factors, such as GBP5, may help develop effective immune-based strategies against numerous pathogens.


Attenuated replication and damaging effects of SARS-CoV-2 Omicron variants in an intestinal epithelial barrier model

July 2024

·

19 Reads

·

4 Citations

Journal of Medical Virology

Many COVID‐19 patients suffer from gastrointestinal symptoms and impaired intestinal barrier function is thought to play a key role in Long COVID. Despite its importance, the impact of severe acute respiratory syndrome coronavirus 2 (SARS‐CoV‐2) on intestinal epithelia is poorly understood. To address this, we established an intestinal barrier model integrating epithelial Caco‐2 cells, mucus‐secreting HT29 cells and Raji cells. This gut epithelial model allows efficient differentiation of Caco‐2 cells into microfold‐like cells, faithfully mimics intestinal barrier function, and is highly permissive to SARS‐CoV‐2 infection. Early strains of SARS‐CoV‐2 and the Delta variant replicated with high efficiency, severely disrupted barrier function, and depleted tight junction proteins, such as claudin‐1, occludin, and ZO‐1. In comparison, Omicron subvariants also depleted ZO‐1 from tight junctions but had fewer damaging effects on mucosal integrity and barrier function. Remdesivir, the fusion inhibitor EK1 and the transmembrane serine protease 2 inhibitor Camostat inhibited SARS‐CoV‐2 replication and thus epithelial barrier damage, while the Cathepsin inhibitor E64d was ineffective. Our results support that SARS‐CoV‐2 disrupts intestinal barrier function but further suggest that circulating Omicron variants are less damaging than earlier viral strains.


A Variety of Mouse PYHIN Proteins Restrict Murine and Human Retroviruses

March 2024

·

49 Reads

·

1 Citation

PYHIN proteins are only found in mammals and play key roles in the defense against bacterial and viral pathogens. The corresponding gene locus shows variable deletion and expansion ranging from 0 genes in bats, over 1 in cows, and 4 in humans to a maximum of 13 in mice. While initially thought to act as cytosolic immune sensors that recognize foreign DNA, increasing evidence suggests that PYHIN proteins also inhibit viral pathogens by more direct mechanisms. Here, we examined the ability of all 13 murine PYHIN proteins to inhibit HIV-1 and murine leukemia virus (MLV). We show that overexpression of p203, p204, p205, p208, p209, p210, p211, and p212 strongly inhibits production of infectious HIV-1; p202, p207, and p213 had no significant effects, while p206 and p214 showed intermediate phenotypes. The inhibitory effects on infectious HIV-1 production correlated significantly with the suppression of reporter gene expression by a proviral Moloney MLV-eGFP construct and HIV-1 and Friend MLV LTR luciferase reporter constructs. Altogether, our data show that the antiretroviral activity of PYHIN proteins is conserved between men and mice and further support the key role of nuclear PYHIN proteins in innate antiviral immunity.


Citations (61)


... This histone deacetylase (HDAC) inhibitor is contra-indicated during pregnancy as it can cause a range of developmental delays and congenital malformation that are referred to as fetal valproate spectrum disorder (FVSD) or fetal valproate syndrome (40). Since GBP5 expression is driven by an endogenous retroviral promoter that is activated upon HDAC inhibition (41)(42)(43), we hypothesized that valproate may induce its expression in placental cells. Valproate dosedependently increased GBP5 protein levels in BeWo cells, a wellcharacterized trophoblast cell line (Fig. 1E) (44). ...

Reference:

Inhibition of placental trophoblast fusion by guanylate-binding protein 5
Viral influencers: deciphering the role of endogenous retroviral LTR12 repeats in cellular gene expression

... GBP5 inhibits a diverse array of viral infections by binding to the oligosaccharyltransferase (OST) complex, thereby obstructing its interaction with viral glycoproteins, including the spike protein of severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2). In addition, it impedes the glycosylation of glycoproteins from other pathogens, such as Middle East Respiratory Syndrome Coronavirus (MERS-CoV), HIV-1, and Influenza Virus (IAV) (Hotter et al. 2017;Sauter and Kirchhoff 2024;Veler et al. 2024;Wang et al. 2024). This mechanism of action offers novel insights into the potential development of broad-spectrum antiviral therapies. ...

Antiviral mechanisms of guanylate-binding protein 5: versatile inhibition of multiple viral glycoproteins

... Significant changes to the intestinal microbiota were observed, disrupting local immune responses [7]. Additionally, it compromises the barrier's structural integrity by altering the expression of tight junction proteins, which has also been demonstrated in cell culture models [14,15]. Persistent SARS-CoV-2 reservoirs in host organs are plausible, as other respiratory RNA viruses, such as latent respiratory syncytial virus or influenza A virus, have been observed to persist for extended periods in murine models [16,17]. ...

Attenuated replication and damaging effects of SARS-CoV-2 Omicron variants in an intestinal epithelial barrier model
  • Citing Article
  • July 2024

Journal of Medical Virology

... The unique PYD and HIN domains of AIM2 compared to the domains found in other ALRs supports its different function [18,65]. Indeed, the HIN domains of the other ALRs, including IFI16 in humans and IFI203 and IFI204 in mice, bind DNA and induce IFN-I signaling in response to pathogens rather than inflammation [68][69][70][71][72]. ...

A Variety of Mouse PYHIN Proteins Restrict Murine and Human Retroviruses

... Mutations in the SARS-CoV-2 3CLpro/Mpro gene are pivotal for modulating hostvirus interactions and significantly influence viral replication efficiency and immune evasion [111] 3CLpro is essential for the processing of viral polyproteins into functional components required for viral replication. Mutations in this protease can modify its substrate specificity and catalytic efficiency, leading to altered viral protein maturation [112]. Additionally, these mutations might influence protease interactions with host proteins, such as those involved in antiviral signalling pathways, further dampening the host immune response. ...

Restriction of Viral Glycoprotein Maturation by Cellular Protease Inhibitors

... • Tetherin Inhibition: ORF3a increases viral particle release by rerouting tetherin to late endosomes/lysosomes [125]. ORF7a inhibits BST-2 (tetherin) activity, aiding in the release of mature virions [162]. • Lysosomal Exocytosis and Viral Egress: ORF3a inhibits autophagy and promotes lysosomal exocytosis, as well as forming ion channels mediating viral egress [126,127,129]. ...

Tetherin Restricts SARS-CoV-2 despite the Presence of Multiple Viral Antagonists

... ORF3a has been shown to inhibit JAK-STAT signalling [36,37], whilst ORF3b [11] and ORF3c suppress IFN-β production. For ORF3c, this effect is mediated by the induction of MAVS degradation [53,59]. ...

ORF3c is expressed in SARS‐CoV‐2‐infected cells and inhibits innate sensing by targeting MAVS
  • Citing Article
  • October 2023

EMBO Reports

... For this purpose, we used a luciferase reporter construct preceded by the promoter region of duck tetherin gene. Tetherin (also called bone marrow stromal antigen 2; Bst2) is a highly IFN-induced avian gene, which we previously characterized in chicken and other avian species (Krchlíková et al. 2020(Krchlíková et al. , 2023. We generated two variants of the tetherin-luciferase construct: i) with native ISRE sequence (complying with the ISRE consensus; (Santhakumar et al. 2018)) and ii) a mutant with a key guanine changed to adenine (Fig. 8). ...

Independent loss events of a functional tetherin gene in galliform birds

... Similarly, LTR69, an ERV3 element, is elevated in an adenocarcinoma cell line, Calu-3, following SARS-CoV-2-induced H3K27 acetylation. LTR69 expression is also prone to increase following stimulation by polyinosinic:polycytidylic acid (poly:IC) and overexpression of interferon regulatory factor 3 (IRF3) and NF-κB subunit, p65/RELA [127], which are also triggered upon SARS-CoV-2 in- fection. In contrast, one study showed downregulation of HERV-H, HERV-W and HERV-K in individuals reinfected with SARS-CoV-2 [128]. ...

SARS-CoV-2 infection induces epigenetic changes in the LTR69 subfamily of endogenous retroviruses

Mobile DNA

... While there are no direct orthologs of human IFI16, IFIX, or MNDA in mice, mouse p204, p205, p207, and p211 cluster with human IFI16 and IFIX in phylogenetic analyses [7,18], suggesting that they may exert similar activities. Gene duplications are commonly observed in immune genes and may allow diversification of expression patterns, targeting of newly emerging pathogens, or evasion of viral antagonists [32][33][34]. Mice are infected with a large variety of viral families, and it is tempting to speculate that the multiplication of Pyhin genes was driven by the need to defend and control a wide range of pathogens. ...

The more the merrier? Gene duplications in the coevolution of primate lentiviruses with their hosts
  • Citing Article
  • August 2023