Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 30
Filter
1.
Cell ; 186(16): 3427-3442.e22, 2023 08 03.
Article in English | MEDLINE | ID: mdl-37421949

ABSTRACT

SARS-CoV-2 is associated with broad tissue tropism, a characteristic often determined by the availability of entry receptors on host cells. Here, we show that TMEM106B, a lysosomal transmembrane protein, can serve as an alternative receptor for SARS-CoV-2 entry into angiotensin-converting enzyme 2 (ACE2)-negative cells. Spike substitution E484D increased TMEM106B binding, thereby enhancing TMEM106B-mediated entry. TMEM106B-specific monoclonal antibodies blocked SARS-CoV-2 infection, demonstrating a role of TMEM106B in viral entry. Using X-ray crystallography, cryogenic electron microscopy (cryo-EM), and hydrogen-deuterium exchange mass spectrometry (HDX-MS), we show that the luminal domain (LD) of TMEM106B engages the receptor-binding motif of SARS-CoV-2 spike. Finally, we show that TMEM106B promotes spike-mediated syncytium formation, suggesting a role of TMEM106B in viral fusion. Together, our findings identify an ACE2-independent SARS-CoV-2 infection mechanism that involves cooperative interactions with the receptors heparan sulfate and TMEM106B.


Subject(s)
COVID-19 , SARS-CoV-2 , Humans , SARS-CoV-2/metabolism , Angiotensin-Converting Enzyme 2/metabolism , Receptors, Virus/metabolism , Virus Internalization , Protein Binding , Membrane Proteins/metabolism , Nerve Tissue Proteins/metabolism
2.
J Gen Virol ; 100(4): 583-601, 2019 04.
Article in English | MEDLINE | ID: mdl-30762518

ABSTRACT

The possible resistance of influenza virus against existing antiviral drugs calls for new therapeutic concepts. One appealing strategy is to inhibit virus entry, in particular at the stage of internalization. This requires a better understanding of virus-host interactions during the entry process, including the role of receptor tyrosine kinases (RTKs). To search for cellular targets, we evaluated a panel of 276 protein kinase inhibitors in a multicycle antiviral assay in Madin-Darby canine kidney cells. The RTK inhibitor Ki8751 displayed robust anti-influenza A and B virus activity and was selected for mechanistic investigations. Ki8751 efficiently disrupted the endocytic process of influenza virus in different cell lines carrying platelet-derived growth factor receptor ß (PDGFRß), an RTK that is known to act at GM3 ganglioside-positive lipid rafts. The more efficient virus entry in CHO-K1 cells compared to the wild-type ancestor (CHO-wt) cells indicated a positive effect of GM3, which is abundant in CHO-K1 but not in CHO-wt cells. Entering virus localized to GM3-positive lipid rafts and the PDGFRß-containing endosomal compartment. PDGFRß/GM3-dependent virus internalization involved PDGFRß phosphorylation, which was potently inhibited by Ki8751, and desialylation of activated PDGFRß by the viral neuraminidase. Virus uptake coincided with strong activation of the Raf/MEK/Erk cascade, but not of PI3K/Akt or phospholipase C-γ. We conclude that influenza virus efficiently hijacks the GM3-enhanced PDGFRß signalling pathway for cell penetration, providing an opportunity for host cell-targeting antiviral intervention.


Subject(s)
G(M3) Ganglioside/metabolism , Influenza, Human/metabolism , Influenza, Human/virology , Orthomyxoviridae Infections/metabolism , Orthomyxoviridae/pathogenicity , Receptor, Platelet-Derived Growth Factor beta/metabolism , Signal Transduction/physiology , Animals , CHO Cells , Cell Line , Cricetulus , Dogs , HEK293 Cells , Humans , Influenza, Human/drug therapy , Madin Darby Canine Kidney Cells , Orthomyxoviridae/drug effects , Orthomyxoviridae Infections/drug therapy , Phenylurea Compounds/pharmacology , Phosphatidylinositol 3-Kinases/metabolism , Phosphorylation/physiology , Protein Kinase Inhibitors/pharmacology , Quinolines/pharmacology , Receptor Protein-Tyrosine Kinases/metabolism , Signal Transduction/drug effects , Virus Internalization/drug effects
3.
J Cell Mol Med ; 21(6): 1206-1216, 2017 06.
Article in English | MEDLINE | ID: mdl-27997763

ABSTRACT

The mast/stem cell growth factor receptor KIT has long been assumed to be a specific marker for interstitial cells of Cajal (ICC) in the bladder, with possible druggable perspectives. However, several authors have challenged the presence of KIT+ ICC in recent years. The aim of this study was therefore to attempt to clarify the conflicting reports on KIT expression in the bladder of human beings, rat, mouse and guinea pig and to elucidate the possible role of antibody-related issues and interspecies differences in this matter. Fresh samples were obtained from human, rat, mouse and guinea pig cystectomies and processed for single/double immunohistochemistry/immunofluorescence. Specific antibodies against KIT, mast cell tryptase (MCT), anoctamin-1 (ANO1) and vimentin were used to characterize the cell types expressing KIT. Gut (jejunum) tissue was used as an external antibody control. Our results revealed KIT expression on mast cells but not on ICC in human, rat, mouse and guinea pig bladder. Parallel immunohistochemistry showed KIT expression on ICC in human, rat, mouse and guinea pig gut, which confirmed the selectivity of the KIT antibody clones. In conclusion, we have shown that KIT+ cells in human, rat, mouse and guinea pig bladder are mast cells and not ICC. The present report is important as it opposes the idea that KIT+ ICC are present in bladder. In this perspective, functional concepts of KIT+ ICC being involved in sensory and/or motor aspects of bladder physiology should be revised.


Subject(s)
Proto-Oncogene Proteins c-kit/genetics , Stem Cells/metabolism , Urinary Bladder/metabolism , Animals , Gene Expression Regulation/genetics , Guinea Pigs , Humans , Interstitial Cells of Cajal/cytology , Interstitial Cells of Cajal/metabolism , Mast Cells/metabolism , Mice , Muscle, Smooth/growth & development , Muscle, Smooth/metabolism , Proto-Oncogene Proteins c-kit/metabolism , Rats , Urinary Bladder/cytology
4.
Cell Tissue Res ; 370(3): 403-416, 2017 12.
Article in English | MEDLINE | ID: mdl-28963588

ABSTRACT

With most research on interstitial cells (IC) in the bladder being conducted on animal models, it remains unclear whether all structural and functional data on IC from animal models can be translated to the human context. This prompted us to compare the structural and immunohistochemical properties of IC in bladders from mouse, rat and human. Tissue samples were obtained from the bladder dome and subsequently processed for immunohistochemistry and electron microscopy. The ultrastructural properties of IC were compared by means of electron microscopy and IC were additionally characterized with single/double immunohistochemistry/immunofluorescence. Our results reveal a similar organization of the IC network in the upper lamina propria (ULP), the deep lamina propria (DLP) and the detrusor muscle in human, rat and mouse bladders. Furthermore, despite several similarities in IC phenotypes, we also found several obvious inter-species differences in IC, especially in the ULP. Most remarkably in this respect, ULP IC in human bladder predominantly displayed a myoid phenotype with abundant presence of contractile micro-filaments, while those in rat and mouse bladders showed a fibroblast phenotype. In conclusion, the organization of ULP IC, DLP IC and detrusor IC is comparable in human, rat and mouse bladders, although several obvious inter-species differences in IC phenotypes were found. The present data show that translating research data on IC in laboratory animals to the human setting should be carried out with caution.


Subject(s)
Interstitial Cells of Cajal/ultrastructure , Mucous Membrane/ultrastructure , Myocytes, Smooth Muscle/ultrastructure , Urinary Bladder/anatomy & histology , Animals , Female , Humans , Immunohistochemistry , Male , Mice , Microscopy, Electron , Rats , Rats, Sprague-Dawley
5.
J Biol Chem ; 289(18): 12494-506, 2014 May 02.
Article in English | MEDLINE | ID: mdl-24634210

ABSTRACT

Lens epithelium-derived growth factor (LEDGF/p75) is a transcriptional co-activator involved in targeting human immunodeficiency virus (HIV) integration and the development of MLL fusion-mediated acute leukemia. A previous study revealed that LEDGF/p75 dynamically scans the chromatin, and upon interaction with HIV-1 integrase, their complex is locked on chromatin. At present, it is not known whether LEDGF/p75-mediated chromatin locking is typical for interacting proteins. Here, we employed continuous photobleaching and fluorescence correlation and cross-correlation spectroscopy to investigate in vivo chromatin binding of JPO2, a LEDGF/p75- and c-Myc-interacting protein involved in transcriptional regulation. In the absence of LEDGF/p75, JPO2 performs chromatin scanning inherent to transcription factors. However, whereas the dynamics of JPO2 chromatin binding are decelerated upon interaction with LEDGF/p75, very strong locking of their complex onto chromatin is absent. Similar results were obtained with the domesticated transposase PogZ, another cellular interaction partner of LEDGF/p75. We furthermore show that diffusive JPO2 can oligomerize; that JPO2 and LEDGF/p75 interact directly and specifically in vivo through the specific interaction domain of JPO2 and the C-terminal domain of LEDGF/p75, comprising the integrase-binding domain; and that modulation of JPO2 dynamics requires a functional PWWP domain in LEDGF/p75. Our results suggest that the dynamics of the LEDGF/p75-chromatin interaction depend on the specific partner and that strong chromatin locking is not a property of all LEDGF/p75-binding proteins.


Subject(s)
Chromatin/metabolism , Intercellular Signaling Peptides and Proteins/metabolism , Multiprotein Complexes/metabolism , Repressor Proteins/metabolism , Amino Acid Motifs , Amino Acid Sequence , Binding Sites , Chromatin/genetics , Fluorescence Resonance Energy Transfer , Gene Knockdown Techniques , Green Fluorescent Proteins/genetics , Green Fluorescent Proteins/metabolism , HeLa Cells , Humans , Intercellular Signaling Peptides and Proteins/chemistry , Intercellular Signaling Peptides and Proteins/genetics , Kinetics , Microscopy, Confocal , Multiprotein Complexes/chemistry , Multiprotein Complexes/genetics , Protein Binding , Protein Multimerization , Repressor Proteins/chemistry , Repressor Proteins/genetics , Transcriptional Activation , Transposases/genetics , Transposases/metabolism
6.
J Urol ; 192(5): 1555-63, 2014 Nov.
Article in English | MEDLINE | ID: mdl-24893312

ABSTRACT

PURPOSE: There is increasing evidence for an important role of the lamina propria in bladder physiology and interstitial cells seem to have a key role in this area. Interstitial cells in the upper lamina propria have been studied most frequently. We characterized interstitial cells in the deeper lamina propria and hypothesized that the 2 interstitial cell populations have different phenotypes based on their ultrastructural and immunohistochemical properties. MATERIALS AND METHODS: Tissue samples were obtained from macroscopically and microscopically normal areas of radical cystectomy specimens. A panel of immunohistochemical markers was used to characterize lamina propria interstitial cells. Single/double immunohistochemistry/immunofluorescence was performed. At a second phase electron microscopy was used to compare upper and deeper lamina propria interstitial cells. RESULTS: Overall the phenotype of upper lamina propria interstitial cells was vimentin, α-smooth muscle actin, caveolin-1 and 2, PDGFRα, and nonphosphorylated and phosphorylated connexin 43 positive, and CD34 and c-kit negative. The overall phenotype of deeper lamina propria interstitial cells was vimentin, CD34 and nonphosphorylated connexin 43 positive, and α-smooth actin, caveolin-1 and 2, PDGFRα, phosphorylated connexin 43 and c-kit negative. Based on ultrastructural findings upper lamina propria interstitial cells were fibroblasts with myoid features and sparse myofibroblasts while deeper lamina propria interstitial cells were interstitial cell of Cajal-like cells. CONCLUSIONS: To our knowledge this is the first study of 2 main interstitial cell populations in the upper and deeper lamina propria of the human bladder with distinct ultrastructural and immunohistochemical phenotypes. Future research is needed to elucidate whether these morphological findings reflect different roles for upper and deeper lamina propria interstitial cells in bladder physiology.


Subject(s)
Interstitial Cells of Cajal/metabolism , Mucous Membrane/metabolism , Urinary Bladder Neoplasms/pathology , Urinary Bladder/ultrastructure , Aged , Biomarkers, Tumor/metabolism , Caveolin 1/metabolism , Connexin 43/metabolism , Female , Fibroblasts/metabolism , Fibroblasts/ultrastructure , Humans , Immunohistochemistry , Interstitial Cells of Cajal/ultrastructure , Male , Microscopy, Confocal , Microscopy, Electron , Mucous Membrane/ultrastructure , Phenotype , Proto-Oncogene Proteins c-kit/metabolism , Receptor, Platelet-Derived Growth Factor alpha/metabolism , Urinary Bladder/metabolism , Urinary Bladder Neoplasms/metabolism , Vimentin/metabolism
7.
Am J Respir Cell Mol Biol ; 48(5): 589-600, 2013 May.
Article in English | MEDLINE | ID: mdl-23328641

ABSTRACT

Malaria-associated acute respiratory distress syndrome (MA-ARDS) is a deadly complication of malaria, and its pathophysiology is insufficiently understood. Both in humans and in murine models, MA-ARDS is characterized by marked pulmonary inflammation. We investigated the role of hemozoin in MA-ARDS in C57Bl/6 mice infected with Plasmodium berghei NK65, P. berghei ANKA, and P. chabaudi AS. By quantifying hemozoin in the lungs and measuring the disease parameters of MA-ARDS, we demonstrated a highly significant correlation between pulmonary hemozoin concentrations, lung weights, and alveolar edema. Histological analysis of the lungs demonstrated that hemozoin is localized in phagocytes and infected erythrocytes, and only occasionally in granulocytes. Species-specific differences in hemozoin production, as measured among individual schizonts, were associated with variations in pulmonary pathogenicity. Furthermore, both pulmonary hemozoin and lung pathology were correlated with the number of infiltrating inflammatory cells, an increased pulmonary expression of cytokines, chemokines, and enzymes, and concentrations of alveolar vascular endothelial growth factor. The causal relationship between hemozoin and inflammation was investigated by injecting P. falciparum-derived hemozoin intravenously into malaria-free mice. Hemozoin potently induced the pulmonary expression of proinflammatory chemokines (interferon-γ inducible protein-10/CXC-chemokine ligand (CXCL)10, monocyte chemotactic protein-1/CC-chemokine ligand 2, and keratinocyte-derived chemokine/CXCL1), cytokines (IL-1ß, IL-6, IL-10, TNF, and transforming growth factor-ß), and other inflammatory mediators (inducible nitric oxide synthase, heme oxygenase-1, nicotinamide adenine dinucleotide phosphate- oxidase-2, and intercellular adhesion molecule-1). Thus, hemozoin correlates with MA-ARDS and induces pulmonary inflammation.


Subject(s)
Hemeproteins/metabolism , Malaria/metabolism , Plasmodium berghei/metabolism , Plasmodium chabaudi/metabolism , Pneumonia/parasitology , Respiratory Distress Syndrome/metabolism , Animals , Bronchoalveolar Lavage Fluid/chemistry , CD4 Lymphocyte Count , Chemokine CCL2/genetics , Chemokine CCL2/metabolism , Erythrocytes/metabolism , Erythrocytes/parasitology , Gene Expression , Hemeproteins/physiology , Host-Parasite Interactions , Humans , Interleukin-10/genetics , Interleukin-10/metabolism , Lung/immunology , Lung/metabolism , Lung/parasitology , Macrophages/metabolism , Macrophages/parasitology , Malaria/complications , Malaria/parasitology , Mice , Mice, Inbred C57BL , Organ Size , Plasmodium berghei/immunology , Plasmodium berghei/physiology , Plasmodium chabaudi/immunology , Plasmodium chabaudi/physiology , Pneumonia/immunology , Pneumonia/metabolism , Respiratory Distress Syndrome/etiology , Schizonts/immunology , Schizonts/metabolism , Schizonts/physiology , Species Specificity , Tumor Necrosis Factor-alpha/genetics , Tumor Necrosis Factor-alpha/metabolism , Vascular Endothelial Growth Factor A/genetics , Vascular Endothelial Growth Factor A/metabolism
8.
J Virol ; 86(17): 9416-31, 2012 Sep.
Article in English | MEDLINE | ID: mdl-22740402

ABSTRACT

We report on a new anti-influenza virus agent, SA-19, a lipophilic glycopeptide derivative consisting of aglycoristocetin coupled to a phenylbenzyl-substituted cyclobutenedione. In Madin-Darby canine kidney cells infected with influenza A/H1N1, A/H3N2, or B virus, SA-19 displayed a 50% antivirally effective concentration of 0.60 µM and a selectivity index (ratio of cytotoxic versus antiviral concentration) of 112. SA-19 was 11-fold more potent than unsubstituted aglycoristocetin and was active in human and nonhuman cell lines. Virus yield at 72 h p.i. was reduced by 3.6 logs at 0.8 µM SA-19. In contrast to amantadine and oseltamivir, SA-19 did not select for resistance upon prolonged virus exposure. SA-19 was shown to inhibit an early postbinding step in virus replication. The compound had no effect on hemagglutinin (HA)-mediated membrane fusion in an HA-polykaryon assay and did not inhibit the low-pH-induced refolding of the HA in a tryptic digestion assay. However, a marked inhibitory effect on the transduction exerted by retroviral pseudoparticles carrying an HA or vesicular stomatitis virus glycoprotein (VSV-G) fusion protein was noted, suggesting that SA-19 targets a cellular factor with a role in influenza virus and VSV entry. Using confocal microscopy with antinucleoprotein staining, SA-19 was proven to completely prevent the influenza virus nuclear entry. This virus arrest was characterized by the formation of cytoplasmic aggregates. SA-19 appeared to disturb the endocytic uptake and trap the influenza virus in vesicles distinct from early, late, or recycling endosomes. The aglycoristocetin derivative SA-19 represents a new class of potent and broad-acting influenza virus inhibitors with potential clinical relevance.


Subject(s)
Antiviral Agents/pharmacology , Cytoplasm/virology , Glycopeptides/pharmacology , Influenza A virus/drug effects , Influenza B virus/drug effects , Animals , Antiviral Agents/chemistry , Cell Line , Cytoplasm/drug effects , Dogs , Glycopeptides/chemistry , Humans , Influenza A Virus, H1N1 Subtype/drug effects , Influenza A Virus, H1N1 Subtype/physiology , Influenza A Virus, H3N2 Subtype/drug effects , Influenza A Virus, H3N2 Subtype/physiology , Influenza A virus/physiology , Influenza B virus/physiology , Influenza, Human/drug therapy , Influenza, Human/virology , Molecular Structure , Virus Internalization/drug effects , Virus Replication/drug effects
9.
mBio ; 13(4): e0137622, 2022 08 30.
Article in English | MEDLINE | ID: mdl-35913162

ABSTRACT

The continuous emergence of new variants of severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) urges better understanding of the functional motifs in the spike (S) protein and their tolerance to mutations. Here, we focused on the S2' motif, which, during virus entry, requires cleavage by a host cell protease to release the fusion peptide. Though belonging to an immunogenic region, the SARS-CoV-2 S2' motif (811-KPSKR-815) has shown hardly any variation, with its three basic (K/R) residues being >99.99% conserved thus far. By creating a series of mutant pseudoviruses bearing the spikes of Wuhan-Hu-1, its G614 mutant or the Delta and Omicron variants, we show that residue K814 (preceding the scissile R815) is dispensable for TMPRSS2 yet favored by the alternative TMPRSS13 protease. Activation by TMPRSS13 was drastically reduced when the SARS-CoV-2 S2' motif was swapped with that of the low pathogenic 229E coronavirus (685-RVAGR-689), and also, the reverse effect was seen. This swap had no impact on recognition by TMPRSS2. In the Middle East respiratory syndrome coronavirus (MERS-CoV) spike, introducing a dibasic scissile motif was easily accepted by TMPRSS13 but less so by TMPRSS2, confirming that TMPRSS13 favors a sequence rich in K/R residues. Pseudovirus entry experiments in Calu-3 cells confirmed that the S2' mutations have minor impact on TMPRSS2. Our findings are the first to demonstrate which S2' residues are important for SARS-CoV-2 spike activation by these two airway proteases, with TMPRSS2 being more tolerant to variation than TMPRSS13. This preemptive insight will help to estimate the impact of S2' motif changes as they appear in new SARS-CoV-2 variants. IMPORTANCE Since its introduction in humans, SARS-CoV-2 is evolving with frequent appearance of new variants. The surveillance would benefit from proactive characterization of the functional motifs in the spike (S) protein, the most variable viral factor. This is linked to immune evasion but also influences spike functioning. Remarkably, though located in a strongly immunogenic region, the S2' cleavage motif has, thus far, remained highly conserved. This suggests that its sequence is critical for spike activation by airway proteases. To investigate this, we assessed how pseudovirus entry is affected by changes in the S2' motif. We demonstrate that TMPRSS2 readily accepts variations in this motif, whereas the alternative TMPRSS13 protease is more fastidious. The Wuhan-Hu-1, G614, Delta and Omicron spikes showed no difference in this regard. Being the first in its kind, our study will help to assess the impact of S2' variations as soon as they are detected during variant surveillance.


Subject(s)
COVID-19 , SARS-CoV-2 , Humans , Membrane Proteins/genetics , Mutation , Peptide Hydrolases/genetics , SARS-CoV-2/genetics , Serine Endopeptidases/genetics , Spike Glycoprotein, Coronavirus/metabolism , Virus Internalization
10.
J Biol Chem ; 285(28): 21768-80, 2010 Jul 09.
Article in English | MEDLINE | ID: mdl-20406803

ABSTRACT

The human immunodeficiency virus, type 1 (HIV-1)-encoded Rev protein is essential for the expression of late viral mRNAs. Rev forms a large organized multimeric protein-protein complex on the Rev response element of these viral mRNA species and transports them from the nucleus to the cytoplasm, exploiting the CRM1-mediated cellular machinery. Here we report the selection of a nanobody, derived from a llama heavy-chain only antibody, that efficiently blocks the assembly of Rev multimers. The nanobody inhibits HIV-1 replication in cells and specifically suppresses the Rev-dependent expression of partially spliced and unspliced HIV-1 RNA. In HIV-susceptible cells, this nanobody thus has potential as an effective anti-HIV agent using genetic immunization strategies. Its binding site was mapped to Rev residues Lys-20 and Tyr-23 located in the N-terminal alpha-helical multimerization domain. In the presence of this nanobody, we observed an accumulation of dimeric Rev species, supporting a head-to-head/tail-to-tail molecular model for Rev assembly. The results indicate that the oligomeric assembly of Rev follows an ordered stepwise process and identify a new epitope within Rev that could guide strategies for the development of novel HIV inhibitors.


Subject(s)
Gene Products, rev/chemistry , HIV Infections/drug therapy , HIV-1/metabolism , Active Transport, Cell Nucleus , Amino Acid Sequence , Animals , Anti-HIV Agents/pharmacology , Antibodies/chemistry , Camelids, New World , Dimerization , Fluorescence Resonance Energy Transfer , Gene Products, rev/metabolism , HeLa Cells , Humans , Lysine/chemistry , Molecular Sequence Data , Protein Interaction Mapping , Protein Structure, Secondary , Protein Structure, Tertiary , Tyrosine/chemistry
11.
Nat Microbiol ; 6(10): 1219-1232, 2021 10.
Article in English | MEDLINE | ID: mdl-34471255

ABSTRACT

The coronavirus disease 2019 (COVID-19) pandemic has claimed millions of lives and caused a global economic crisis. No effective antiviral drugs are currently available to treat infections of severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2). The medical need imposed by the pandemic has spurred unprecedented research efforts to study coronavirus biology. Every virus depends on cellular host factors and pathways for successful replication. These proviral host factors represent attractive targets for antiviral therapy as they are genetically more stable than viral targets and may be shared among related viruses. The application of various 'omics' technologies has led to the rapid discovery of proviral host factors that are required for the completion of the SARS-CoV-2 life cycle. In this Review, we summarize insights into the proviral host factors that are required for SARS-CoV-2 infection that were mainly obtained using functional genetic and interactome screens. We discuss cellular processes that are important for the SARS-CoV-2 life cycle, as well as parallels with non-coronaviruses. Finally, we highlight host factors that could be targeted by clinically approved molecules and molecules in clinical trials as potential antiviral therapies for COVID-19.


Subject(s)
COVID-19/metabolism , SARS-CoV-2/physiology , Antiviral Agents/pharmacology , Antiviral Agents/therapeutic use , COVID-19/virology , Host-Pathogen Interactions/drug effects , Humans , Peptide Hydrolases/metabolism , RNA, Viral/metabolism , Receptors, Virus/metabolism , SARS-CoV-2/drug effects , Viral Proteins/metabolism , Virus Internalization/drug effects , Virus Replication/drug effects , COVID-19 Drug Treatment
12.
Pharmaceuticals (Basel) ; 14(10)2021 Oct 17.
Article in English | MEDLINE | ID: mdl-34681276

ABSTRACT

We used classical linear and microwave-assisted synthesis methods to prepare novel N-substituted, benzimidazole-derived acrylonitriles with antiproliferative activity against several cancer cells in vitro. The most potent systems showed pronounced activity against all tested hematological cancer cell lines, with favorable selectivity towards normal cells. The selection of lead compounds was also tested in vitro for tubulin polymerization inhibition as a possible mechanism of biological action. A combination of docking and molecular dynamics simulations confirmed the suitability of the employed organic skeleton for the design of antitumor drugs and demonstrated that their biological activity relies on binding to the colchicine binding site in tubulin. In addition, it also underlined that higher tubulin affinities are linked with (i) bulkier alkyl and aryl moieties on the benzimidazole nitrogen and (ii) electron-donating substituents on the phenyl group that allow deeper entrance into the hydrophobic pocket within the tubulin's ß-subunit, consisting of Leu255, Leu248, Met259, Ala354, and Ile378 residues.

13.
Nat Genet ; 53(4): 435-444, 2021 04.
Article in English | MEDLINE | ID: mdl-33686287

ABSTRACT

The ongoing COVID-19 pandemic has caused a global economic and health crisis. To identify host factors essential for coronavirus infection, we performed genome-wide functional genetic screens with severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) and human coronavirus 229E. These screens uncovered virus-specific as well as shared host factors, including TMEM41B and PI3K type 3. We discovered that SARS-CoV-2 requires the lysosomal protein TMEM106B to infect human cell lines and primary lung cells. TMEM106B overexpression enhanced SARS-CoV-2 infection as well as pseudovirus infection, suggesting a role in viral entry. Furthermore, single-cell RNA-sequencing of airway cells from patients with COVID-19 demonstrated that TMEM106B expression correlates with SARS-CoV-2 infection. The present study uncovered a collection of coronavirus host factors that may be exploited to develop drugs against SARS-CoV-2 infection or future zoonotic coronavirus outbreaks.


Subject(s)
COVID-19/genetics , CRISPR-Cas Systems , Genome, Human/genetics , Genome-Wide Association Study/methods , Membrane Proteins/genetics , Nerve Tissue Proteins/genetics , Bronchoalveolar Lavage Fluid/cytology , COVID-19/epidemiology , COVID-19/virology , Cell Line, Tumor , Cells, Cultured , Coronavirus 229E, Human/genetics , Epidemics , Epithelial Cells/virology , Gene Expression , Host-Pathogen Interactions , Humans , Proviruses/physiology , SARS-CoV-2/physiology , Virus Internalization
14.
Antivir Ther ; 13(8): 977-90, 2008.
Article in English | MEDLINE | ID: mdl-19195323

ABSTRACT

BACKGROUND: ST-246 is a potent anti-orthopoxviral molecule targeting the F13L protein of vaccinia virus, which is involved in the wrapping of viruses. The discrepancy in sensitivities of several orthopoxviruses to ST-246 has raised questions about potential differences in their replicative cycles and/or the presence of another drug target. METHODS: Density gradients were used to evaluate the differences between the viral cycles of vaccinia, cowpox and camelpox viruses. Also, to investigate if ST-246 inhibits a single target, we compared its activity to that of small interfering RNAs designed to silence the F13L gene (siF13Ls). RESULTS: We showed that the spread of vaccinia virus involved both intracellular and extracellular enveloped viruses, whereas both cowpox and camelpox viruses seemed to propagate via non-enveloped intracellular forms and cell-associated viral particles. Although ST-246 exerted a clear antiviral activity by interfering with the egress of the virus from infected cells, we observed that cowpox and camelpox viruses, in contrast to vaccinia virus, could be directed towards a lytic cycle under ST-246 treatment. We specifically knocked down the F13L transcripts of vaccinia and camelpox viruses by > 85%, reduced virus progeny by 90% and showed that siF13Ls affect camelpox and vaccinia virus propagation differently. Flow cytometry data validated that ST-246 interfered with the activity of the F13L protein, whereas siF13Ls silenced the F13L gene. CONCLUSIONS: Our observations support that vaccinia, cowpox and camelpox viruses exhibit different levels of sensitivity to ST-246 because of dissimilarities between their ways of propagation, and provide a better understanding of the mode of action of ST-246.


Subject(s)
Benzamides/pharmacology , Cowpox virus/drug effects , Isoindoles/pharmacology , Membrane Proteins/metabolism , Orthopoxvirus/drug effects , Vaccinia virus/drug effects , Viral Envelope Proteins/metabolism , Antiviral Agents/pharmacology , Cell Line , Gene Silencing , Humans , RNA, Small Interfering , Species Specificity , Virus Replication/drug effects
15.
Bioorg Med Chem ; 16(21): 9487-97, 2008 Nov 01.
Article in English | MEDLINE | ID: mdl-18835718

ABSTRACT

CRM1-mediated nucleocytoplasmic transport plays an important role in many cellular processes and diseases. To investigate the structural basis required for the inhibition of the CRM1-mediated nuclear export we have synthesized analogs of a previously identified small molecule lead compound and monitored their activity against the Rev function of the human immunodeficiency virus. Microscopy studies show that the active congeners of this series inhibit the nucleocytoplasmic transport of Rev and the co-localization between Rev and CRM1 in living cells. Mechanism of action studies show their interaction with the Cys528 residue of CRM1 involving a Michael-addition type of reaction. However, structure-activity relationship demonstrates strict constraints to the structure of the inhibitors, and shows that activity is not solely correlated to Michael-addition suggesting a more complex mechanism of action. Our results are suggestive for the existence of a well-defined interaction at the CRM1-NES binding site. In addition, the most selective congener inhibited the HIV-1 production in latently infected cells. These specific CRM1 inhibitors are of interest as tool for analyzing the mechanisms of post-transcriptional control of gene expression and provide insight in the design of new agents.


Subject(s)
Acrylates/pharmacology , Active Transport, Cell Nucleus/drug effects , HIV-1/metabolism , Karyopherins/pharmacology , Virus Replication/drug effects , rev Gene Products, Human Immunodeficiency Virus/antagonists & inhibitors , Binding Sites , HIV Infections , HeLa Cells , Humans , Kidney/drug effects , Kidney/metabolism , Nuclear Export Signals , Receptors, Cytoplasmic and Nuclear , Structure-Activity Relationship , rev Gene Products, Human Immunodeficiency Virus/genetics , rev Gene Products, Human Immunodeficiency Virus/metabolism , Exportin 1 Protein
16.
Int J Food Microbiol ; 266: 190-199, 2018 Feb 02.
Article in English | MEDLINE | ID: mdl-29227906

ABSTRACT

Postharvest losses of fruit and vegetables can reach up to 30%, the main cause being microbial decay. For apple fruit, mostly fungal pathogens, such as Penicillium expansum, Colletotrichum spp., Neofabraea spp. and Botrytis cinerea, are important. As such losses are unsustainable in many ways, it is necessary that research is conducted to prevent them. Generally, for plants and fruit grown under non-sterile field conditions, disinfection is carried out prior to the start of a phytopathological experiment. The motivation for this practice is the removal of background contamination so that it will not affect the experimental outcome and its interpretation. In literature, a plethora of disinfection methods exists, differing in disinfectant, strength and duration. The following two disinfectants are commonly used: sodium hypochlorite (NaOCl) and ethanol. This article presents a targeted investigation into the effects of these two disinfectants on apple fruit surface and physiology. The results clearly demonstrate that both were affected by both disinfectants. NaOCl caused oxidative damage to the apple's wax layer, causing it to crack. Ethanol affected a redistribution of the wax on the fruit surface and altered the wax composition and/or metabolism. Both NaOCl and ethanol treatment resulted in an increased respiration rate. Therefore, apple and possibly other fruit should not be disinfected in phytopathological studies. A negative control, as is typically used, is not solving this issue, as we clearly demonstrate that the living tissue shows metabolic effects following disinfection, and hence the study objects are changed, hampering a clear interpretation of the experimental outcomes. Moreover, fungal inoculation during experiments is typically taking place at rather large levels in wounded tissue (as infection success is the exception), outnumbering the variable levels of background population, if present.


Subject(s)
Ethanol/pharmacology , Food Microbiology , Food Preservation/methods , Fruit/drug effects , Malus/drug effects , Sodium Hypochlorite/pharmacology , Disinfectants/pharmacology , Food Microbiology/trends , Fruit/microbiology , Malus/microbiology
17.
J Infect ; 77(2): 119-130, 2018 08.
Article in English | MEDLINE | ID: mdl-29752963

ABSTRACT

OBJECTIVES: The opportunistic pathogen Staphylococcus epidermidis is progressively involved in device-related infections. Since these infections involve biofilm formation, antibiotics are not effective. Conversely, a vaccine can be advantageous to prevent these infections. In view of vaccine development, predicted surface proteins were evaluated on their potential as a vaccine target. METHODS: Immunoglobulins directed against S. epidermidis surface proteins SesB, M, O, Q and R were used to firstly affirm their surface location. Further, inhibitory effects of these IgGs on biofilm formation were determined in vitro on polystyrene and polyurethane surfaces and in vivo using a subcutaneous catheter mouse model. We also examined the opsonophagocytotic capacity of these IgGs. RESULTS: Surface localization of the five Ses proteins was demonstrated both for planktonic and sessile cells, though to a variable extent. Ses-specific IgGs added to planktonic cells had a variable inhibitory effect on cell adhesion to polystyrene, while only anti-SesO IgGs decreased cell attachment to polyurethane catheters. Although phagocytic killing was only obtained after opsonization with SesB-specific IgGs, a significant reduction of in vivo formed biofilms was observed after administration of SesB-, SesM- and SesO-specific IgGs. CONCLUSIONS: Regardless of their characterization or function, S. epidermidis surface proteins can be adequate targets for vaccine development aiming the prevention of device-related infections caused by invasive S. epidermidis strains.


Subject(s)
Bacterial Proteins/immunology , Bacterial Vaccines/immunology , Membrane Proteins/immunology , Staphylococcal Infections/prevention & control , Staphylococcus epidermidis/metabolism , Animals , Antibody Specificity , Bacterial Proteins/genetics , Bacterial Proteins/metabolism , Biofilms , Gene Expression Regulation, Bacterial/physiology , HL-60 Cells , Humans , Immunoglobulin G/immunology , Rabbits , Staphylococcal Infections/microbiology , Staphylococcus epidermidis/immunology
18.
Eur J Med Chem ; 132: 219-235, 2017 May 26.
Article in English | MEDLINE | ID: mdl-28365318

ABSTRACT

5-(2-(4-Methoxyphenyl)ethyl)-2-amino-3-methylcarboxylate thiophene (TR560) is the prototype drug of a recently discovered novel class of tumor-selective compounds that preferentially inhibit the proliferation of specific tumor cell types (e.g. leukemia/lymphoma). Here, we further increased tumor selectivity by simplification of the molecule through replacing the 4-methoxyphenyl moiety by an alkyl chain. Several 2-amino-3-methylcarboxylate thiophene derivatives containing at C-5 an alkyl group consisting of at least 6 (hexyl) to 9 (nonyl) carbon units showed pronounced anti-proliferative activity in the mid-nanomolar range with 500- to 1000-fold tumor cell selectivity. The compounds preferentially inhibited the proliferation of T-lymphoma CEM and Molt/4, prostate PC-3, kidney Caki-1 and hepatoma Huh-7 tumor cells, but were virtually inactive against other tumor cell lines including B-lymphoma Raji and cervix carcinoma HeLa cells. The novel prototype drug 3j (containing a 5-heptyl chain) elicited a cytotoxic, rather than cytostatic activity, already after 4 h of exposure. The unusual tumor selectivity could not be explained by a differential uptake (or efflux) of the drug by sensitive versus resistant tumor cells. Exposure of a fluorescent derivative of 3j revealed pronounced uptake of the drug in the cytoplasm, no visible appearance in the nucleus, and a predominant localization in the endoplasmic reticulum. These observations may be helpful to narrow down the intracellular localization and identification of the molecular target of the 5-substituted thiophene derivatives.


Subject(s)
Antineoplastic Agents/pharmacology , Thiophenes/chemical synthesis , Alkylation , Antineoplastic Agents/chemical synthesis , Antineoplastic Agents/chemistry , Cell Line, Tumor , Cell Nucleus/metabolism , Cell Proliferation/drug effects , Endoplasmic Reticulum/metabolism , Humans , Structure-Activity Relationship , Thiophenes/pharmacokinetics , Thiophenes/pharmacology , Tissue Distribution
19.
Clin Cancer Res ; 23(10): 2528-2541, 2017 May 15.
Article in English | MEDLINE | ID: mdl-27780859

ABSTRACT

Purpose: Human exportin-1 (XPO1) is the key nuclear-cytoplasmic transport protein that exports different cargo proteins out of the nucleus. Inducing nuclear accumulation of these proteins by inhibiting XPO1 causes cancer cell death. First clinical validation of pharmacological inhibition of XPO1 was obtained with the Selective Inhibitor of Nuclear Export (SINE) compound selinexor (KPT-330) demonstrating activity in phase-II/IIb clinical trials when dosed 1 to 3 times weekly. The second-generation SINE compound KPT-8602 shows improved tolerability and can be dosed daily. Here, we investigate and validate the drug-target interaction of KPT-8602 and explore its activity against acute lymphoblastic leukemia (ALL).Experimental Design: We examined the effect of KPT-8602 on XPO1 function and XPO1-cargo as well as on a panel of leukemia cell lines. Mutant XPO1 leukemia cells were designed to validate KPT-8602's drug-target interaction. In vivo, anti-ALL activity was measured in a mouse ALL model and patient-derived ALL xenograft models.Results: KPT-8602 induced caspase-dependent apoptosis in a panel of leukemic cell lines in vitro Using CRISPR/Cas9 genome editing, we demonstrated the specificity of KPT-8602 for cysteine 528 in the cargo-binding groove of XPO1 and validated the drug target interaction. In vivo, KPT-8602 showed potent anti-leukemia activity in a mouse ALL model as well as in patient-derived T- and B-ALL xenograft models without affecting normal hematopoiesis.Conclusions: KPT-8602 is highly specific for XPO1 inhibition and demonstrates potent anti-leukemic activity supporting clinical application of the second-generation SINE compound for the treatment of ALL. Clin Cancer Res; 23(10); 2528-41. ©2016 AACR.


Subject(s)
Antineoplastic Agents/administration & dosage , Karyopherins/antagonists & inhibitors , Precursor Cell Lymphoblastic Leukemia-Lymphoma/drug therapy , Receptors, Cytoplasmic and Nuclear/antagonists & inhibitors , Thiazoles/administration & dosage , Active Transport, Cell Nucleus/drug effects , Animals , Antineoplastic Agents/chemistry , Apoptosis/drug effects , CRISPR-Cas Systems , Cell Line, Tumor , Gene Editing , Humans , Karyopherins/genetics , Mice , Precursor Cell Lymphoblastic Leukemia-Lymphoma/genetics , Precursor Cell Lymphoblastic Leukemia-Lymphoma/pathology , Receptors, Cytoplasmic and Nuclear/genetics , Xenograft Model Antitumor Assays , Exportin 1 Protein
20.
Oncotarget ; 7(42): 68842-68850, 2016 Oct 18.
Article in English | MEDLINE | ID: mdl-27634897

ABSTRACT

Exportin-1 (CRM1/XPO1) is a crucial nuclear export protein that transports a wide variety of proteins from the nucleus to the cytoplasm. These cargo proteins include tumor suppressors and growth-regulatory factors and as such XPO1 is considered a potential anti-cancer target. From this perspective, inhibition of the XPO1-mediated nuclear export by selective inhibitor of nuclear export (SINE) compounds has shown broad-spectrum anti-cancer activity. Furthermore, the clinical candidate SINE, selinexor, is currently in multiple phase I/II/IIb trials for treatment of cancer. Resistance against selinexor has not yet been observed in the clinic, but in vitro selection of resistance did not reveal any mutations in the target protein, XPO1. However, introduction of a homozygous mutation at the drug's target site, the cysteine 528 residue inside the XPO1 cargo-binding pocket, by genetic engineering, confers resistance to selinexor. Here we investigated whether this resistance to selinexor is recessive or dominant. For this purpose we have engineered multiple leukemia cell lines containing heterozygous or homozygous C528S substitutions using CRISPR/Cas9-mediated genome editing. Our findings show that heterozygous mutation confers similar resistance against selinexor as homozygous substitution, demonstrating that SINE resistance can be obtained by a single and dominant mutation of the cysteine528 residue in XPO1.


Subject(s)
Active Transport, Cell Nucleus , Antineoplastic Agents/pharmacology , Drug Resistance, Neoplasm , Karyopherins/genetics , Receptors, Cytoplasmic and Nuclear/genetics , CRISPR-Cas Systems , Cell Survival , Cysteine/genetics , Gene Editing , Genetic Engineering , HL-60 Cells , Heterozygote , Homozygote , Humans , Jurkat Cells , K562 Cells , Mutation , Protein Binding , Exportin 1 Protein
SELECTION OF CITATIONS
SEARCH DETAIL