Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 11 de 11
Filter
Add more filters










Publication year range
1.
Viruses ; 15(12)2023 11 28.
Article in English | MEDLINE | ID: mdl-38140570

ABSTRACT

Interferon-induced transmembrane proteins (IFITM1, 2 and 3) are important host antiviral defense factors. They are active against viruses like the influenza A virus (IAV), dengue virus (DENV), Ebola virus (EBOV), Zika virus (ZIKV) and severe acute respiratory syndrome coronavirus (SARS-CoV). In this review, we focus on IFITM3 S-palmitoylation, a reversible lipid modification, and describe its role in modulating IFITM3 antiviral activity. Our laboratory discovered S-palmitoylation of IFITMs using chemical proteomics and demonstrated the importance of highly conserved fatty acid-modified Cys residues in IFITM3 antiviral activity. Further studies showed that site-specific S-palmitoylation at Cys72 is important for IFITM3 trafficking to restricted viruses (IAV and EBOV) and membrane-sterol interactions. Thus, site-specific lipid modification of IFITM3 directly regulates its antiviral activity, cellular trafficking, and membrane-lipid interactions.


Subject(s)
Influenza A virus , Zika Virus Infection , Zika Virus , Humans , Lipoylation , RNA-Binding Proteins/metabolism , Zika Virus/metabolism , Influenza A virus/metabolism , Antiviral Agents/metabolism , Lipids , Membrane Proteins/metabolism
2.
Protein Sci ; 32(4): e4618, 2023 04.
Article in English | MEDLINE | ID: mdl-36883310

ABSTRACT

Post-translational modifications (PTMs) of proteins not only exponentially increase the diversity of proteoforms, but also contribute to dynamically modulating the localization, stability, activity, and interaction of proteins. Understanding the biological consequences and functions of specific PTMs has been challenging for many reasons, including the dynamic nature of many PTMs and the technical limitations to access homogenously modified proteins. The genetic code expansion technology has emerged to provide unique approaches for studying PTMs. Through site-specific incorporation of unnatural amino acids (UAAs) bearing PTMs or their mimics into proteins, genetic code expansion allows the generation of homogenous proteins with site-specific modifications and atomic resolution both in vitro and in vivo. With this technology, various PTMs and mimics have been precisely introduced into proteins. In this review, we summarize the UAAs and approaches that have been recently developed to site-specifically install PTMs and their mimics into proteins for functional studies of PTMs.


Subject(s)
Protein Processing, Post-Translational , Proteins , Proteins/chemistry , Amino Acids/chemistry , Genetic Code , Codon
3.
ACS Chem Biol ; 17(8): 2109-2120, 2022 08 19.
Article in English | MEDLINE | ID: mdl-35861660

ABSTRACT

Interferon-induced transmembrane proteins (IFITM1, 2, and 3) are important antiviral proteins that are active against many viruses, including influenza A virus (IAV), dengue virus (DENV), Ebola virus (EBOV), Zika virus (ZIKV), and severe acute respiratory syndrome coronavirus (SARS-CoV). IFITM proteins exhibit specificity in activity, but their distinct mechanisms of action and regulation are unclear. Since S-palmitoylation and cholesterol homeostasis are crucial for viral infections, we investigated IFITM interactions with cholesterol by photoaffinity cross-linking in mammalian cells along with molecular dynamic simulations and nuclear magnetic resonance analysis in vitro. These studies suggest that cholesterol can directly interact with S-palmitoylated IFITMs in cells and alter the conformation of IFITMs in membrane bilayers. Notably, we discovered that the S-palmitoylation levels regulate differential IFITM protein interactions with cholesterol in mammalian cells and specificity of antiviral activity toward IAV, SARS-CoV-2, and EBOV. Our studies suggest that modulation of IFITM S-palmitoylation levels and cholesterol interaction influence host susceptibility to different viruses.


Subject(s)
Antiviral Agents , Lipoylation , Membrane Proteins , Sterols , Animals , Antiviral Agents/pharmacology , Cholesterol/metabolism , Influenza A virus , Membrane Proteins/metabolism , Membrane Proteins/pharmacology , SARS-CoV-2 , Sterols/metabolism , Zika Virus
4.
Curr Opin Chem Biol ; 65: 109-117, 2021 12.
Article in English | MEDLINE | ID: mdl-34333222

ABSTRACT

Protein S-fatty acylation or S-palmitoylation is a reversible and regulated lipid post-translational modification (PTM) in eukaryotes. Loss-of-function mutagenesis studies have suggested important roles for protein S-fatty acylation in many fundamental biological pathways in development, neurobiology, and immunity that are also associated with human diseases. However, the hydrophobicity and reversibility of this PTM have made site-specific gain-of-function studies more challenging to investigate. In this review, we summarize recent chemical biology approaches and methods that have enabled site-specific gain-of-function studies of protein S-fatty acylation and the investigation of the mechanisms and significance of this PTM in eukaryotic biology.


Subject(s)
Lipoylation , Protein S , Acylation , Humans , Protein Processing, Post-Translational , Protein S/metabolism
5.
ACS Chem Biol ; 16(5): 844-856, 2021 05 21.
Article in English | MEDLINE | ID: mdl-33887136

ABSTRACT

Interferon-induced transmembrane proteins (IFITMs) are S-palmitoylated proteins in vertebrates that restrict a diverse range of viruses. S-palmitoylated IFITM3 in particular engages incoming virus particles, prevents their cytoplasmic entry, and accelerates their lysosomal clearance by host cells. However, how S-palmitoylation modulates the structure and biophysical characteristics of IFITM3 to promote its antiviral activity remains unclear. To investigate how site-specific S-palmitoylation controls IFITM3 antiviral activity, we employed computational, chemical, and biophysical approaches to demonstrate that site-specific lipidation of cysteine 72 enhances the antiviral activity of IFITM3 by modulating its conformation and interaction with lipid membranes. Collectively, our results demonstrate that site-specific S-palmitoylation of IFITM3 directly alters its biophysical properties and activity in cells to prevent virus infection.


Subject(s)
Antiviral Agents/chemistry , Cell Membrane/metabolism , Interferons/chemistry , Lipids/chemistry , Membrane Proteins/metabolism , RNA-Binding Proteins/metabolism , Amino Acid Sequence , Antiviral Agents/pharmacology , Binding Sites , Cell Membrane/ultrastructure , Computational Biology , Drug Design , Humans , Interferons/pharmacology , Lipoylation , Lysosomes/metabolism , Molecular Dynamics Simulation , Protein Binding , Protein Conformation , Signal Transduction
6.
Open Biol ; 11(3): 200411, 2021 03.
Article in English | MEDLINE | ID: mdl-33653086

ABSTRACT

S-palmitoylation is a reversible posttranslational lipid modification of proteins. It controls protein activity, stability, trafficking and protein-protein interactions. Recent global profiling of immune cells and targeted analysis have identified many S-palmitoylated immunity-associated proteins. Here, we review S-palmitoylated immune receptors and effectors, and their dynamic regulation at cellular membranes to generate specific and balanced immune responses. We also highlight how this understanding can drive therapeutic advances to pharmacologically modulate immune responses.


Subject(s)
Adaptive Immunity , Immunity, Innate , Protein Processing, Post-Translational , Animals , Humans , Lipoylation , Receptors, Immunologic/metabolism
7.
Res Sq ; 2021 Dec 29.
Article in English | MEDLINE | ID: mdl-34981045

ABSTRACT

Interferon-induced transmembrane proteins (IFITM1, 2 and 3) are important antiviral proteins that are active against many viruses, including influenza A virus (IAV), dengue virus (DENV), Ebola virus (EBOV), Zika virus (ZIKV) and severe acute respiratory syndrome coronavirus (SARS-CoV). IFITMs exhibit isoform-specific activity, but their distinct mechanisms of action and regulation are unclear. Since S -palmitoylation and cholesterol homeostasis are crucial for viral infections, we investigated IFITM interactions with cholesterol by molecular dynamic stimulations, nuclear magnetic resonance analysis in vitro and photoaffinity crosslinking in mammalian cells. These studies suggest that cholesterol can alter the conformation of IFITMs in membrane bilayers and directly interact with S -palmitoylated IFITMs in cells. Notably, we discovered that the S -palmitoylation levels regulate differential IFITM isoform interactions with cholesterol in mammalian cells and specificity of antiviral activity towards IAV, SARS-CoV-2 and EBOV. Our studies suggest that modulation of IFITM S -palmitoylation levels and cholesterol interaction may influence host susceptibility to different viruses.

8.
Cell Chem Biol ; 27(5): 571-585.e6, 2020 05 21.
Article in English | MEDLINE | ID: mdl-32243810

ABSTRACT

Interferon-induced transmembrane protein 3 (IFITM3) is a key interferon effector that broadly prevents infection by diverse viruses. However, the cellular factors that control IFITM3 homeostasis and antiviral activity have not been fully elucidated. Using site-specific photo-crosslinking and quantitative proteomic analysis, here we present the identification and functional characterization of VCP/p97 AAA-ATPase as a primary interaction partner of IFITM3. We show that IFITM3 ubiquitination at lysine 24 is crucial for VCP binding, trafficking, turnover, and engagement with incoming virus particles. Consistently, pharmacological inhibition of VCP/p97 ATPase activity leads to defective IFITM3 lysosomal sorting, turnover, and co-trafficking with virus particles. Our results showcase the utility of site-specific protein photo-crosslinking in mammalian cells and reveal VCP/p97 as a key cellular factor involved in IFITM3 trafficking and homeostasis.


Subject(s)
Membrane Proteins/metabolism , RNA-Binding Proteins/metabolism , Valosin Containing Protein/metabolism , HEK293 Cells , Humans , Protein Interaction Maps , Protein Transport , Proteomics , Ubiquitination
9.
Nat Chem Biol ; 15(3): 259-268, 2019 03.
Article in English | MEDLINE | ID: mdl-30643282

ABSTRACT

Interferon-induced transmembrane proteins (IFITMs 1, 2 and 3) have emerged as important innate immune effectors that prevent diverse virus infections in vertebrates. However, the cellular mechanisms and live-cell imaging of these small membrane proteins have been challenging to evaluate during viral entry of mammalian cells. Using CRISPR-Cas9-mediated IFITM-mutant cell lines, we demonstrate that human IFITM1, IFITM2 and IFITM3 act cooperatively and function in a dose-dependent fashion in interferon-stimulated cells. Through site-specific fluorophore tagging and live-cell imaging studies, we show that IFITM3 is on endocytic vesicles that fuse with incoming virus particles and enhances the trafficking of this pathogenic cargo to lysosomes. IFITM3 trafficking is specific to restricted viruses, requires S-palmitoylation and is abrogated with loss-of-function mutants. The site-specific protein labeling and live-cell imaging approaches described here should facilitate the functional analysis of host factors involved in pathogen restriction as well as their mechanisms of regulation.


Subject(s)
Membrane Proteins/metabolism , Membrane Proteins/physiology , RNA-Binding Proteins/metabolism , RNA-Binding Proteins/physiology , Transport Vesicles/physiology , A549 Cells , Animals , Antigens, Differentiation/metabolism , Antiviral Agents , Endosomes/physiology , HeLa Cells , Humans , Lysosomes/physiology , Optical Imaging/methods , Protein Transport , Virion/pathogenicity , Virus Internalization
10.
Angew Chem Int Ed Engl ; 55(4): 1412-6, 2016 Jan 22.
Article in English | MEDLINE | ID: mdl-26663633

ABSTRACT

Organophosphorus-based nerve agents, such as paraoxon, parathion, and malathion, inhibit acetylcholinesterase, which results in paralysis, respiratory failure, and death. Bacteria are known to use the enzyme phosphotriesterase (PTE) to break down these compounds. In this work, we designed vacancy-engineered nanoceria (VE CeO2 NPs) as PTE mimetic hotspots for the rapid degradation of nerve agents. We observed that the hydrolytic effect of the nanomaterial is due to the synergistic activity between both Ce(3+) and Ce(4+) ions located in the active site-like hotspots. Furthermore, the catalysis by nanoceria overcomes the product inhibition generally observed for PTE and small molecule-based PTE mimetics.


Subject(s)
Cerium/chemistry , Enzymes/metabolism , Molecular Mimicry , Nerve Agents/metabolism , Organophosphorus Compounds/metabolism , Biodegradation, Environmental , Microscopy, Electron, Transmission , X-Ray Diffraction
11.
Chem Asian J ; 11(1): 72-6, 2016 Jan.
Article in English | MEDLINE | ID: mdl-26377634

ABSTRACT

Nanomaterials-based enzyme mimetics (nanozymes) have attracted considerable interest due to their applications in imaging, diagnostics, and therapeutic treatments. Particularly, metal-oxide nanozymes have been shown to mimic the interesting redox properties and biological activities of metalloenzymes. Here we describe an efficient synthesis of MnFe2 O4 nanomaterials and show how the morphology can be controlled by using a simple co-precipitation method. The nanomaterials prepared by this method exhibit a remarkable oxidase-like activity. Interestingly, the activity is morphology-dependent, with nanooctahedra (NOh) exhibiting a catalytic efficiency of 2.21×10(9) m(-1) s(-1) , the highest activity ever reported for a nanozyme.


Subject(s)
Iron/metabolism , Manganese/metabolism , Nanostructures/chemistry , Oxidoreductases/metabolism , Oxygen/metabolism , Iron/chemistry , Manganese/chemistry , Oxidoreductases/chemistry , Oxygen/chemistry , Particle Size , Surface Properties
SELECTION OF CITATIONS
SEARCH DETAIL
...