Your browser doesn't support javascript.
loading
: 20 | 50 | 100
1 - 20 de 1.325
1.
J Phys Chem B ; 128(20): 4986-4995, 2024 May 23.
Article En | MEDLINE | ID: mdl-38739415

Membrane fusion is considered the first step in the entry of enveloped viruses into the host cell. Several targeted strategies have been implemented to block viral entry by limiting the fusion protein to form a six-helix bundle, which is a prerequisite for fusion. Nonetheless, the development of broad-spectrum fusion inhibitors is essential to combat emerging and re-emerging viral infections. TG-23, a coronin 1, a tryptophan-aspartate-rich phagosomal protein-derived peptide, demonstrated inhibition of fusion between small unilamellar vesicles (SUVs) by modulating the membrane's physical properties. However, its inhibitory efficacy reduces with an increasing concentration of membrane cholesterol. The present work aims to develop a fusion inhibitor whose efficacy would be unaltered in the presence of membrane cholesterol. A stretch of the tryptophan-aspartic acid-containing peptide with a similar secondary structure and hydrophobicity profile of TG-23 from coronin 1 was synthesized, and its ability to inhibit SUV-SUV fusion with varying concentrations of membrane cholesterol was evaluated. Our results demonstrate that the GG-21 peptide inhibits fusion irrespective of the cholesterol content of the membrane. We have further evaluated the peptide-induced change in the membrane organization and dynamics utilizing arrays of steady-state and time-resolved fluorescence measurements and correlated these results with their effect on fusion. Interestingly, GG-21 displays inhibitory efficacy in a wide variety of lipid compositions despite having a secondary structure and physical properties similar to those of TG-23. Overall, our results advocate that the secondary structure and physical properties of the peptide may not be sufficient to predict its inhibitory efficacy.


Cholesterol , Membrane Fusion , Membrane Fusion/drug effects , Cholesterol/chemistry , Peptides/chemistry , Peptides/pharmacology , Unilamellar Liposomes/chemistry , Unilamellar Liposomes/metabolism , Microfilament Proteins/metabolism , Microfilament Proteins/chemistry
2.
Mol Biol Rep ; 50(3): 2033-2039, 2023 Mar.
Article En | MEDLINE | ID: mdl-36538173

BACKGROUND: Based on our previous research conducted on cinnamaldehyde (CA) exhibiting its ability to improve the growth performance of fattening pigs and the adipogenesis induction model of C2C12 cells constructed in our laboratory, we explored the effects of CA on the generation and development of lipid droplets (LDs) in adipogenic differentiated C2C12 cells. METHODS AND RESULTS: C2C12 cells were treated with either 0.4 mM or 0.8 mM CA. BODIPY staining and triglyceride measurements were conducted to observe the morphology of LDs, and Western blotting was used to measure the expression of their metabolism-related proteins. The results showed that the average number of LDs in the CA treatment groups was more than the control group (P < 0.05), whereas the average LD size and triglyceride content decreased (P < 0.05). Compared with the control group, the expression levels of fusion-related genes in the LDs of the CA treatment group significantly decreased, while decomposition-related genes and autophagy-related genes in the LDs in C2C12 cells significantly increased (P < 0.01). CONCLUSION: Cinnamaldehyde promoted the decomposition and autophagy of lipid droplets in C2C12 cells and inhibited the fusion of lipid droplets.


Acrolein , Adipocytes , Cell Differentiation , Lipid Droplets , Lipid Metabolism , Lipid Droplets/drug effects , Lipid Droplets/metabolism , Adipocytes/cytology , Adipocytes/drug effects , Adipocytes/metabolism , Autophagy/drug effects , Autophagy/genetics , Membrane Fusion/drug effects , Lipid Metabolism/drug effects , Meat/standards , Food Quality , Animals , Mice , Cell Line , Acrolein/analogs & derivatives , Triglycerides
3.
J Mater Chem B ; 9(47): 9658-9669, 2021 12 08.
Article En | MEDLINE | ID: mdl-34647566

Specific interactions between viruses and host cells provide essential insights into material science-based strategies to combat emerging viral diseases. pH-triggered viral fusion is ubiquitous to multiple viral families and is important for understanding the viral infection cycle. Inspired by this process, virus detection has been achieved using nanomaterials with host-mimetic membranes, enabling interactions with amphiphilic hemagglutinin fusion peptides of viruses. Most research has been on designing functional nanoparticles with fusogenic capability for virus detection, and there has been little exploitation of the kinetic stability to alter the ability of nanoparticles to interact with viral membranes and improve their sensing performance. In this study, a homogeneous fluorescent assay using self-assembled polymeric nanoparticles (PNPs) with tunable responsiveness to external stimuli is developed for rapid and straightforward detection of an activated influenza A virus. Dissociation of PNPs induced by virus insertion can be readily controlled by varying the fraction of hydrophilic segments in copolymers constituting PNPs, giving rise to fluorescence signals within 30 min and detection of various influenza viruses, including H9N2, CA04(H1N1), H4N6, and H6N8. Therefore, the designs demonstrated in this study propose underlying approaches for utilizing engineered PNPs through modulation of their kinetic stability for direct and sensitive identification of infectious viruses.


Influenza A virus/isolation & purification , Nanoparticles/chemistry , Peptides/chemistry , Polyethylene Glycols/chemistry , Viral Fusion Proteins/metabolism , Animals , Carbocyanines/chemistry , Chickens , Eggs/virology , Fluorescence Resonance Energy Transfer/methods , Fluorescent Dyes/chemistry , Influenza A virus/metabolism , Limit of Detection , Membrane Fusion/drug effects , Membranes, Artificial , Peptides/chemical synthesis , Peptides/metabolism , Polyethylene Glycols/chemical synthesis , Polyethylene Glycols/metabolism
4.
Cell Death Dis ; 12(10): 939, 2021 10 13.
Article En | MEDLINE | ID: mdl-34645799

Lysosome-autophagosome fusion is critical to autophagosome maturation. Although several proteins that regulate this fusion process have been identified, the prefusion architecture and its regulation remain unclear. Herein, we show that upon stimulation, multiple lysosomes form clusters around individual autophagosomes, setting the stage for membrane fusion. The soluble N-ethylmaleimide-sensitive factor attachment protein receptor (SNARE) protein on lysosomes-vesicle-associated membrane protein 8 (VAMP8)-plays an important role in forming this prefusion state of lysosomal clusters. To study the potential role of phosphorylation on spontaneous fusion, we investigated the effect of phosphorylation of C-terminal residues of VAMP8. Using a phosphorylation mimic, we observed a decrease of fusion in an ensemble lipid mixing assay and an increase of unfused lysosomes associated with autophagosomes. These results suggest that phosphorylation not only reduces spontaneous fusion for minimizing autophagic flux under normal conditions, but also preassembles multiple lysosomes to increase the fusion probability for resuming autophagy upon stimulation. VAMP8 phosphorylation may thus play an important role in chemotherapy drug resistance by influencing autophagosome maturation.


Autophagosomes/metabolism , Lysosomes/metabolism , Membrane Fusion , R-SNARE Proteins/metabolism , Autophagosomes/drug effects , Autophagosomes/ultrastructure , Carbonyl Cyanide m-Chlorophenyl Hydrazone/pharmacology , Drug Resistance, Neoplasm/drug effects , HeLa Cells , Humans , Lysosomes/drug effects , Lysosomes/ultrastructure , Membrane Fusion/drug effects , Phosphorylation/drug effects , R-SNARE Proteins/chemistry , SNARE Proteins/metabolism , Temozolomide/pharmacology
5.
Nutrients ; 13(10)2021 Sep 25.
Article En | MEDLINE | ID: mdl-34684361

Lactoferrin (LF) was used at first as a vehicle to deliver non-soluble active compounds to the body, including the central nervous system (CNS). Nonetheless, it soon became evident that, apart from acting as a vehicle, LF itself owns active effects in the CNS. In the present study, the effects of LF are assessed both in baseline conditions, as well as to counteract methamphetamine (METH)-induced neurodegeneration by assessing cell viability, cell phenotype, mitochondrial status, and specific autophagy steps. In detail, cell integrity in baseline conditions and following METH administration was carried out by using H&E staining, Trypan blue, Fluoro Jade B, and WST-1. Western blot and immuno-fluorescence were used to assess the expression of the neurofilament marker ßIII-tubulin. Mitochondria were stained using Mito Tracker Red and Green and were further detailed and quantified by using transmission electron microscopy. Autophagy markers were analyzed through immuno-fluorescence and electron microscopy. LF counteracts METH-induced degeneration. In detail, LF significantly attenuates the amount of cell loss and mitochondrial alterations produced by METH; and mitigates the dissipation of autophagy-related proteins from the autophagy compartment, which is massively induced by METH. These findings indicate a protective role of LF in the molecular mechanisms of neurodegeneration.


Autophagy , Lactoferrin/pharmacology , Methamphetamine/toxicity , Mitochondria/metabolism , Protective Agents/pharmacology , Animals , Autophagosomes/drug effects , Autophagosomes/metabolism , Autophagosomes/ultrastructure , Autophagy/drug effects , Cathepsin D/metabolism , Cell Line, Tumor , Cell Survival/drug effects , Humans , Lactoferrin/administration & dosage , Lysosomal-Associated Membrane Protein 1/metabolism , Lysosomes/drug effects , Lysosomes/metabolism , Membrane Fusion/drug effects , Methamphetamine/administration & dosage , Microtubule-Associated Proteins/metabolism , Mitochondria/drug effects , Mitochondria/ultrastructure , PC12 Cells , Phenotype , Rats , Time Factors , Tubulin/metabolism , Vacuoles/drug effects , Vacuoles/metabolism , Vacuoles/ultrastructure
6.
Cell Death Dis ; 12(10): 917, 2021 10 07.
Article En | MEDLINE | ID: mdl-34620841

We previously demonstrated that sulforaphane (SFN) inhibited autophagy leading to apoptosis in human non-small cell lung cancer (NSCLC) cells, but the underlying subcellular mechanisms were unknown. Hereby, high-performance liquid chromatography-tandem mass spectrometry uncovered that SFN regulated the production of lipoproteins, and microtubule- and autophagy-associated proteins. Further, highly expressed fatty acid synthase (FASN) contributed to cancer malignancy and poor prognosis. Results showed that SFN depolymerized microtubules, downregulated FASN, and decreased its binding to α-tubulin; SFN downregulated FASN, acetyl CoA carboxylase (ACACA), and ATP citrate lyase (ACLY) via activating proteasomes and downregulating transcriptional factor SREBP1; SFN inhibited the interactions among α-tubulin and FASN, ACACA, and ACLY; SFN decreased the amount of intracellular fatty acid (FA) and mitochondrial phospholipids; and knockdown of FASN decreased mitochondrial membrane potential (ΔΨm) and increased reactive oxygen species, mitochondrial abnormality, and apoptosis. Further, SFN downregulated mitophagy-associated proteins Bnip3 and NIX, and upregulated mitochondrial LC3 II/I. Transmission electron microscopy showed mitochondrial abnormality and accumulation of mitophagosomes in response to SFN. Combined with mitophagy inducer CCCP or autophagosome-lysosome fusion inhibitor Bafilomycin A1, we found that SFN inhibited mitophagosome-lysosome fusion leading to mitophagosome accumulation. SFN reduced the interaction between NIX and LC3 II/I, and reversed CCCP-caused FA increase. Furthermore, knockdown of α-tubulin downregulated NIX and BNIP3 production, and upregulated LC3 II/I. Besides, SFN reduced the interaction and colocalization between α-tubulin and NIX. Thus, SFN might cause apoptosis via inhibiting microtubule-mediated mitophagy. These results might give us a new insight into the mechanisms of SFN-caused apoptosis in the subcellular level.


Apoptosis , Down-Regulation , Fatty Acid Synthases/metabolism , Isothiocyanates/pharmacology , Microtubules/metabolism , Mitophagy , Sulfoxides/pharmacology , Aged , Apoptosis/drug effects , Autophagosomes/drug effects , Autophagosomes/metabolism , Autophagosomes/ultrastructure , Cell Line, Tumor , Down-Regulation/drug effects , Fatty Acids/biosynthesis , Female , Humans , Lysosomes/drug effects , Lysosomes/metabolism , Lysosomes/ultrastructure , Male , Membrane Fusion/drug effects , Microtubule-Associated Proteins/metabolism , Microtubules/drug effects , Microtubules/ultrastructure , Middle Aged , Mitophagy/drug effects , Models, Biological , Polymerization , Proteasome Endopeptidase Complex/metabolism , Protein Multimerization/drug effects , Tubulin/metabolism
7.
Curr Opin Virol ; 51: 34-47, 2021 12.
Article En | MEDLINE | ID: mdl-34592709

The Paramyxoviridae family includes enveloped single-stranded negative-sense RNA viruses such as measles, mumps, human parainfluenza, canine distemper, Hendra, and Nipah viruses, which cause a tremendous global health burden. The ability of paramyxoviral glycoproteins to merge viral and host membranes allows entry of the viral genome into host cells, as well as cell-cell fusion, an important contributor to disease progression. Recent molecular and structural advances in our understanding of the paramyxovirus membrane fusion machinery gave rise to various therapeutic approaches aiming at inhibiting viral infection, spread, and cytopathic effects. These therapeutic approaches include peptide mimics, antibodies, and small molecule inhibitors with various levels of success at inhibiting viral entry, increasing the potential of effective antiviral therapeutic development.


Antiviral Agents/pharmacology , Cell Membrane/metabolism , Membrane Fusion/drug effects , Paramyxoviridae Infections/drug therapy , Paramyxoviridae Infections/virology , Paramyxoviridae/drug effects , Paramyxoviridae/metabolism , Animals , Antiviral Agents/therapeutic use , Cell Fusion , Cell Membrane/drug effects , Humans
8.
Antiviral Res ; 193: 105125, 2021 09.
Article En | MEDLINE | ID: mdl-34197863

Several arenaviruses, including Lassa and Lujo viruses in Africa and five New World arenavirus (NWA) species in the Americas, cause life-threatening viral hemorrhagic fevers. In the absence of licensed antiviral therapies, these viruses pose a significant public health risk. The envelope glycoprotein complex (GPC) mediates arenavirus entry through a pH-dependent fusion of the viral and host endosomal membranes. It thus is recognized as a viable target for small-molecule fusion inhibitors. Here, we report on the antiviral activity and pre-clinical development of the novel broad-spectrum arenavirus fusion inhibitors, ARN-75039 and ARN-75041. In Tacaribe virus (TCRV) pseudotyped and native virus assays, the ARN compounds were active in the low to sub-nanomolar range with selectivity indices exceeding 1000. Pharmacokinetic analysis of the orally administered compounds revealed an extended half-life in mice supporting once-daily dosing, and the compounds were well tolerated at the highest tested dose of 100 mg/kg. In a proof-of-concept prophylactic efficacy study, doses of 10 and 35 mg/kg of either compound dramatically improved survival outcome and potently inhibited TCRV replication in serum and various tissues. Additionally, in contrast to surviving mice that received ribavirin or placebo, animals treated with ARN-75039 or ARN-75041 were cured of TCRV infection. In a follow-up study with ARN-75039, impressive therapeutic efficacy was demonstrated under conditions where treatment was withheld until after the onset of disease. Taken together, the data strongly support the continued development of ARN-75039 as a candidate therapeutic for the treatment of severe arenaviral diseases.


Antiviral Agents/pharmacology , Arenaviridae Infections/drug therapy , Arenaviruses, New World/drug effects , Membrane Fusion/drug effects , Small Molecule Libraries/pharmacology , Administration, Oral , Animals , Antiviral Agents/pharmacokinetics , Chlorocebus aethiops , Male , Mice , Ribavirin/pharmacology , Small Molecule Libraries/pharmacokinetics , Vero Cells , Viral Envelope Proteins/chemistry , Viral Envelope Proteins/metabolism , Virus Internalization/drug effects
9.
Sci Rep ; 11(1): 10955, 2021 05 26.
Article En | MEDLINE | ID: mdl-34040104

The primary hallmark of Parkinson's disease (PD) is the generation of Lewy bodies of which major component is α-synuclein (α-Syn). Because of increasing evidence of the fundamental roles of α-Syn oligomers in disease progression, α-Syn oligomers have become potential targets for therapeutic interventions for PD. One of the potential toxicities of α-Syn oligomers is their inhibition of SNARE-mediated vesicle fusion by specifically interacting with vesicle-SNARE protein synaptobrevin-2 (Syb2), which hampers dopamine release. Here, we show that α-Syn monomers and oligomers cooperatively inhibit neuronal SNARE-mediated vesicle fusion. α-Syn monomers at submicromolar concentrations increase the fusion inhibition by α-Syn oligomers. This cooperative pathological effect stems from the synergically enhanced vesicle clustering. Based on this cooperative inhibition mechanism, we reverse the fusion inhibitory effect of α-Syn oligomers using small peptide fragments. The small peptide fragments, derivatives of α-Syn, block the binding of α-Syn oligomers to Syb2 and dramatically reverse the toxicity of α-Syn oligomers in vesicle fusion. Our findings demonstrate a new strategy for therapeutic intervention in PD and related diseases based on this specific interaction of α-Syn.


Membrane Fusion/drug effects , SNARE Proteins/antagonists & inhibitors , alpha-Synuclein/pharmacology , Amino Acid Sequence , Amino Acid Substitution , Dopamine/metabolism , Dopamine/pharmacology , Drug Evaluation, Preclinical , Liposomes , Membrane Lipids/metabolism , Models, Molecular , Mutation, Missense , Peptide Fragments/pharmacology , Point Mutation , Protein Binding , Protein Multimerization , Proteolipids/chemistry , Recombinant Fusion Proteins/pharmacology , SNARE Proteins/physiology , Vesicle-Associated Membrane Protein 2/antagonists & inhibitors , Vesicle-Associated Membrane Protein 2/physiology , alpha-Synuclein/chemistry , alpha-Synuclein/genetics , alpha-Synuclein/toxicity
10.
Viruses ; 13(5)2021 04 23.
Article En | MEDLINE | ID: mdl-33922579

HIV-1 (human immunodeficiency virus type 1) infection begins with the attachment of the virion to a host cell by its envelope glycoprotein (Env), which subsequently induces fusion of viral and cell membranes to allow viral entry. Upon binding to primary receptor CD4 and coreceptor (e.g., chemokine receptor CCR5 or CXCR4), Env undergoes large conformational changes and unleashes its fusogenic potential to drive the membrane fusion. The structural biology of HIV-1 Env and its complexes with the cellular receptors not only has advanced our knowledge of the molecular mechanism of how HIV-1 enters the host cells but also provided a structural basis for the rational design of fusion inhibitors as potential antiviral therapeutics. In this review, we summarize our latest understanding of the HIV-1 membrane fusion process and discuss related therapeutic strategies to block viral entry.


HIV Fusion Inhibitors/pharmacology , HIV-1/drug effects , HIV-1/physiology , Membrane Fusion/drug effects , Virus Internalization/drug effects , Anti-Retroviral Agents/pharmacology , HIV Fusion Inhibitors/classification , HIV Infections/virology , Humans
11.
Cells ; 10(4)2021 04 17.
Article En | MEDLINE | ID: mdl-33920542

Autophagy is a specific macromolecule and organelle degradation process. The target macromolecule or organelle is first enclosed in an autophagosome, and then delivered along acetylated microtubules to the lysosome. Autophagy is triggered by stress and largely contributes to cell survival. We have previously shown that S6K1 kinase is essential for autophagic flux under stress conditions. Here, we aimed to elucidate the underlying mechanism of S6K1 involvement in autophagy. We stimulated autophagy in S6K1/2 double-knockout mouse embryonic fibroblasts by exposing them to different stress conditions. Transient gene overexpression or silencing, immunoblotting, immunofluorescence, flow cytometry, and ratiometric fluorescence analyses revealed that the perturbation of autophagic flux in S6K1-deficient cells did not stem from impaired lysosomal function. Instead, the absence of S6K1 abolished stress-induced tubulin acetylation and disrupted the acetylated microtubule network, in turn impairing the autophagosome-lysosome fusion. S6K1 overexpression restored tubulin acetylation and autophagic flux in stressed S6K1/2-deficient cells. Similar effect of S6K1 status was observed in prostate cancer cells. Furthermore, overexpression of an acetylation-mimicking, but not acetylation-resistant, tubulin variant effectively restored autophagic flux in stressed S6K1/2-deficient cells. Collectively, S6K1 controls tubulin acetylation, hence contributing to the autophagic flux induced by different stress conditions and in different cells.


Autophagy , Microtubules/metabolism , Ribosomal Protein S6 Kinases, 70-kDa/metabolism , Stress, Physiological , Acetylation/drug effects , Animals , Autophagosomes/drug effects , Autophagosomes/metabolism , Autophagy/drug effects , Cell Line, Tumor , Embryo, Mammalian/cytology , Fibroblasts/drug effects , Fibroblasts/metabolism , Glucose/deficiency , Humans , Isothiocyanates/pharmacology , Lysosomes/drug effects , Lysosomes/metabolism , Membrane Fusion/drug effects , Mice, Knockout , Microtubule-Associated Proteins/metabolism , Microtubules/drug effects , Models, Biological , Phenotype , Phosphorylation/drug effects , Proteolysis/drug effects , Stress, Physiological/drug effects , Sulfoxides/pharmacology , Tubulin/metabolism
12.
Photochem Photobiol Sci ; 20(2): 321-326, 2021 Feb.
Article En | MEDLINE | ID: mdl-33721250

Charge recombination kinetics of bacterial photosynthetic protein Reaction Center displays an exquisite sensitivity to the actual occupancy of ubiquinone-10 in its QB-binding site. Here, we have exploited such phenomenon for assessing the growth and the aggregation/fusion of phosphocholine vesicles embedding RC in their membrane, when treated with sodium oleate.


Bacterial Proteins/chemistry , Liposomes/chemistry , Photosynthetic Reaction Center Complex Proteins/chemistry , Rhodobacter sphaeroides/metabolism , Bacterial Proteins/metabolism , Dynamic Light Scattering , Membrane Fusion/drug effects , Oleic Acid/chemistry , Oleic Acid/pharmacology , Photosynthetic Reaction Center Complex Proteins/metabolism , Ubiquinone/analogs & derivatives , Ubiquinone/chemistry , Ubiquinone/metabolism
13.
Sci Rep ; 11(1): 5558, 2021 03 10.
Article En | MEDLINE | ID: mdl-33692386

The recent COVID-19 pandemic poses a serious threat to global public health, thus there is an urgent need to define the molecular mechanisms involved in SARS-CoV-2 spike (S) protein-mediated virus entry that is essential for preventing and/or treating this emerging infectious disease. In this study, we examined the blocking activity of human COVID-19 convalescent plasma by cell-cell fusion assays using SARS-CoV-2-S-transfected 293 T as effector cells and ACE2-expressing 293 T as target cells. We demonstrate that the SARS-CoV-2 S protein exhibits a very high capacity for membrane fusion and is efficient in mediating virus fusion and entry into target cells. Importantly, we find that COVID-19 convalescent plasma with high titers of IgG neutralizing antibodies can block cell-cell fusion and virus entry by interfering with the SARS-CoV-2-S/ACE2 or SARS-CoV-S/ACE2 interactions. These findings suggest that COVID-19 convalescent plasma may not only inhibit SARS-CoV-2-S but also cross-neutralize SARS-CoV-S-mediated membrane fusion and virus entry, supporting its potential as a preventive and/or therapeutic agent against SARS-CoV-2 as well as other SARS-CoV infections.


COVID-19/immunology , COVID-19/therapy , Spike Glycoprotein, Coronavirus/immunology , Adult , Antibodies, Neutralizing/immunology , Antibodies, Viral/immunology , COVID-19/prevention & control , Cell Fusion/methods , Female , Humans , Immunization, Passive/methods , Male , Membrane Fusion/drug effects , Middle Aged , Pandemics/prevention & control , Plasma/chemistry , Receptors, Virus/metabolism , SARS-CoV-2/immunology , SARS-CoV-2/pathogenicity , Spike Glycoprotein, Coronavirus/metabolism , Virus Internalization/drug effects , COVID-19 Serotherapy
14.
Science ; 371(6536): 1379-1382, 2021 03 26.
Article En | MEDLINE | ID: mdl-33597220

Containment of the COVID-19 pandemic requires reducing viral transmission. Severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) infection is initiated by membrane fusion between the viral and host cell membranes, which is mediated by the viral spike protein. We have designed lipopeptide fusion inhibitors that block this critical first step of infection and, on the basis of in vitro efficacy and in vivo biodistribution, selected a dimeric form for evaluation in an animal model. Daily intranasal administration to ferrets completely prevented SARS-CoV-2 direct-contact transmission during 24-hour cohousing with infected animals, under stringent conditions that resulted in infection of 100% of untreated animals. These lipopeptides are highly stable and thus may readily translate into safe and effective intranasal prophylaxis to reduce transmission of SARS-CoV-2.


COVID-19/transmission , Lipopeptides/administration & dosage , Membrane Fusion/drug effects , SARS-CoV-2/drug effects , Viral Fusion Protein Inhibitors/administration & dosage , Virus Internalization/drug effects , Administration, Intranasal , Animals , COVID-19/prevention & control , COVID-19/virology , Chlorocebus aethiops , Disease Models, Animal , Drug Design , Ferrets , Lipopeptides/chemistry , Lipopeptides/pharmacokinetics , Lipopeptides/pharmacology , Mice , Pre-Exposure Prophylaxis , SARS-CoV-2/isolation & purification , SARS-CoV-2/physiology , Spike Glycoprotein, Coronavirus/metabolism , Tissue Distribution , Vero Cells , Viral Fusion Protein Inhibitors/chemistry , Viral Fusion Protein Inhibitors/pharmacokinetics , Viral Fusion Protein Inhibitors/pharmacology
15.
Cell Death Dis ; 12(1): 80, 2021 01 13.
Article En | MEDLINE | ID: mdl-33441536

It is widely accepted that lysosomes are essential for cell homeostasis, and autophagy plays an important role in tumor development. Here, we found FV-429, a synthetic flavonoid compound, inhibited autophagy flux, promoted autophagosomes accumulation, and inhibited lysosomal degradation in T-cell malignancies. These effects were likely to be achieved by lysosomal dysregulation. The destructive effects of FV-429 on lysosomes resulted in blockage of lysosome-associated membrane fusion, lysosomal membrane permeabilization (LMP), and cathepsin-mediated caspase-independent cell death (CICD). Moreover, we initially investigated the effects of autophagy inhibition by FV-429 on the therapeutic efficacy of chemotherapy and found that FV-429 sensitized cancer cells to chemotherapy agents. Our findings suggest that FV-429 could be a potential novel autophagy inhibitor with notable antitumor efficacy as a single agent.


Flavonoids/pharmacology , Hematologic Neoplasms/drug therapy , Lysosomes/drug effects , Antineoplastic Agents/pharmacology , Autophagy/drug effects , Cell Death/drug effects , Cell Line, Tumor , Cell Membrane Permeability , HEK293 Cells , Hematologic Neoplasms/pathology , Humans , Jurkat Cells , Leukemia-Lymphoma, Adult T-Cell/drug therapy , Leukemia-Lymphoma, Adult T-Cell/pathology , Lymphoma, Large-Cell, Anaplastic/drug therapy , Lymphoma, Large-Cell, Anaplastic/pathology , Lysosomes/metabolism , Membrane Fusion/drug effects
16.
Eur J Pharmacol ; 894: 173836, 2021 Mar 05.
Article En | MEDLINE | ID: mdl-33387467

The COVID-19 pandemic has spread rapidly and poses an unprecedented threat to the global economy and human health. Broad-spectrum antivirals are currently being administered to treat severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2). China's prevention and treatment guidelines suggest the use of an anti-influenza drug, arbidol, for the clinical treatment of COVID-19. Reports indicate that arbidol could neutralize SARS-CoV-2. Monotherapy with arbidol is superior to lopinavir-ritonavir or favipiravir for treating COVID-19. In SARS-CoV-2 infection, arbidol acts by interfering with viral binding to host cells. However, the detailed mechanism by which arbidol induces the inhibition of SARS-CoV-2 is not known. Here, we present atomistic insights into the mechanism underlying membrane fusion inhibition of SARS-CoV-2 by arbidol. Molecular dynamics (MD) simulation-based analyses demonstrate that arbidol binds and stabilizes at the receptor-binding domain (RBD)/ACE2 interface with a high affinity. It forms stronger intermolecular interactions with the RBD than ACE2. Analyses of the detailed decomposition of energy components and binding affinities revealed a substantial increase in the affinity between the RBD and ACE2 in the arbidol-bound RBD/ACE2 complex, suggesting that arbidol generates favorable interactions between them. Based on our MD simulation results, we propose that the binding of arbidol induces structural rigidity in the viral glycoprotein, thus restricting the conformational rearrangements associated with membrane fusion and virus entry. Furthermore, key residues of the RBD and ACE2 that interact with arbidol were identified, opening the door for developing therapeutic strategies and higher-efficacy arbidol derivatives or lead drug candidates.


Antiviral Agents/metabolism , Antiviral Agents/pharmacology , Indoles/metabolism , Indoles/pharmacology , SARS-CoV-2/drug effects , Angiotensin-Converting Enzyme 2/metabolism , Computer Simulation , Glycoproteins/drug effects , Glycoproteins/metabolism , Humans , Membrane Fusion/drug effects , Models, Molecular , Molecular Conformation , Molecular Docking Simulation , Molecular Dynamics Simulation , Protein Domains , COVID-19 Drug Treatment
17.
Int J Mol Sci ; 22(1)2021 Jan 05.
Article En | MEDLINE | ID: mdl-33466417

Liposomes are highly biocompatible and versatile drug carriers with an increasing number of applications in the field of nuclear medicine and diagnostics. So far, only negatively charged liposomes with intercalated radiometals, e.g., 64Cu, 99mTc, have been reported. However, the process of cellular uptake of liposomes by endocytosis is rather slow. Cellular uptake can be accelerated by recently developed cationic liposomes, which exhibit extraordinarily high membrane fusion ability. The aim of the present study was the development of the formulation and the characterization of such cationic fusogenic liposomes with intercalated radioactive [131I]I- for potential use in therapeutic applications. The epithelial human breast cancer cell line MDA-MB-231 was used as a model for invasive cancer cells and cellular uptake of [131I]I- was monitored in vitro. Delivery efficiencies of cationic and neutral liposomes were compared with uptake of free iodide. The best cargo delivery efficiency (~10%) was achieved using cationic fusogenic liposomes due to their special delivery pathway of membrane fusion. Additionally, human blood cells were also incubated with cationic control liposomes and free [131I]I-. In these cases, iodide delivery efficiencies remained below 3%.


Cations/chemistry , Drug Carriers/chemistry , Iodine Radioisotopes/administration & dosage , Iodine Radioisotopes/chemistry , Liposomes/chemistry , Nanoparticles/chemistry , Animals , CHO Cells , Cell Line , Cell Line, Tumor , Cricetulus , Endocytosis/drug effects , Humans , Membrane Fusion/drug effects
18.
Angew Chem Int Ed Engl ; 60(11): 6101-6106, 2021 03 08.
Article En | MEDLINE | ID: mdl-33241871

The entry of enveloped virus requires the fusion of viral and host cell membranes. An effective fusion inhibitor aiming at impeding such membrane fusion may emerge as a broad-spectrum antiviral agent against a wide range of viral infections. Mycobacterium survives inside the phagosome by inhibiting phagosome-lysosome fusion with the help of a coat protein coronin 1. Structural analysis of coronin 1 and other WD40-repeat protein suggest that the trp-asp (WD) sequence is placed at distorted ß-meander motif (more exposed) in coronin 1. The unique structural feature of coronin 1 was explored to identify a simple lipo-peptide sequence (myr-WD), which effectively inhibits membrane fusion by modulating the interfacial order, water penetration, and surface potential. The mycobacterium inspired lipo-dipeptide was successfully tested to combat type 1 influenza virus (H1N1) and murine coronavirus infections as a potential broad-spectrum antiviral agent.


Antiviral Agents/pharmacology , Dipeptides/pharmacology , Lipopeptides/pharmacology , Membrane Fusion/drug effects , Virus Internalization/drug effects , Animals , Antiviral Agents/chemistry , Antiviral Agents/toxicity , Dipeptides/chemistry , Dipeptides/toxicity , Dogs , Humans , Influenza A Virus, H1N1 Subtype/drug effects , Lipopeptides/chemistry , Lipopeptides/toxicity , Liposomes/chemistry , Madin Darby Canine Kidney Cells , Murine hepatitis virus/drug effects , Phosphatidylcholines/chemistry , Phosphatidylethanolamines/chemistry , Rats
19.
Viruses ; 12(12)2020 12 21.
Article En | MEDLINE | ID: mdl-33371476

Flaviviruses bear class II fusion proteins as their envelope (E) proteins. Here, we describe the development of an in vitro quantitative mosquito-cell-based membrane-fusion assay for the E protein using dual split proteins (DSPs). The assay does not involve the use of live viruses and allows the analysis of a membrane-fusion step independent of other events in the viral lifecycle, such as endocytosis. The progress of membrane fusion can be monitored continuously by measuring the activities of Renilla luciferase derived from the reassociation of DSPs during cell fusion. We optimized the assay to screen an FDA-approved drug library for a potential membrane fusion inhibitor using the E protein of Zika virus. Screening results identified atovaquone, which was previously described as an antimalarial agent. Atovaquone potently blocked the in vitro Zika virus infection of mammalian cells with an IC90 of 2.1 µM. Furthermore, four distinct serotypes of dengue virus were also inhibited by atovaquone with IC90 values of 1.6-2.5 µM, which is a range below the average blood concentration of atovaquone after its oral administration in humans. These findings make atovaquone a likely candidate drug to treat illnesses caused by Zika as well as dengue viruses. Additionally, the DSP assay is useful to study the mechanism of membrane fusion in Flaviviruses.


Antiviral Agents/pharmacology , Dengue Virus/drug effects , Dengue/virology , Membrane Fusion/drug effects , Viral Envelope Proteins/metabolism , Zika Virus Infection/virology , Zika Virus/drug effects , Animals , Cell Line , Culicidae , Dengue/drug therapy , Dengue Virus/physiology , Flow Cytometry , High-Throughput Nucleotide Sequencing , Humans , Microbial Sensitivity Tests , Virus Internalization/drug effects , Zika Virus/physiology , Zika Virus Infection/drug therapy
20.
Int J Mol Sci ; 21(23)2020 Nov 30.
Article En | MEDLINE | ID: mdl-33265989

Discovering antibiotic molecules able to hold the growing spread of antimicrobial resistance is one of the most urgent endeavors that public health must tackle. The case of Gram-negative bacterial pathogens is of special concern, as they are intrinsically resistant to many antibiotics, due to an outer membrane that constitutes an effective permeability barrier. Antimicrobial peptides (AMPs) have been pointed out as potential alternatives to conventional antibiotics, as their main mechanism of action is membrane disruption, arguably less prone to elicit resistance in pathogens. Here, we investigate the in vitro activity and selectivity of EcDBS1R4, a bioinspired AMP. To this purpose, we have used bacterial cells and model membrane systems mimicking both the inner and the outer membranes of Escherichia coli, and a variety of optical spectroscopic methodologies. EcDBS1R4 is effective against the Gram-negative E. coli, ineffective against the Gram-positive Staphylococcus aureus and noncytotoxic for human cells. EcDBS1R4 does not form stable pores in E. coli, as the peptide does not dissipate its membrane potential, suggesting an unusual mechanism of action. Interestingly, EcDBS1R4 promotes a hemi-fusion of vesicles mimicking the inner membrane of E. coli. This fusogenic ability of EcDBS1R4 requires the presence of phospholipids with a negative curvature and a negative charge. This finding suggests that EcDBS1R4 promotes a large lipid spatial reorganization able to reshape membrane curvature, with interesting biological implications herein discussed.


Escherichia coli/drug effects , Pore Forming Cytotoxic Proteins/pharmacology , Animals , Anions , Cell Death/drug effects , Cell Membrane/drug effects , Cell Membrane/metabolism , Humans , Kinetics , Membrane Fusion/drug effects , Membrane Potentials/drug effects , Mice, Inbred C57BL , Microbial Sensitivity Tests , Microbial Viability/drug effects , Pore Forming Cytotoxic Proteins/chemistry , Protein Conformation
...