Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 10 de 10
Filter
Add more filters










Publication year range
1.
J Enzyme Inhib Med Chem ; 39(1): 2301772, 2024 Dec.
Article in English | MEDLINE | ID: mdl-38221792

ABSTRACT

The viral genome of the SARS-CoV-2 coronavirus, the aetiologic agent of COVID-19, encodes structural, non-structural, and accessory proteins. Most of these components undergo rapid genetic variations, though to a lesser extent the essential viral proteases. Consequently, the protease and/or deubiquitinase activities of the cysteine proteases Mpro and PLpro became attractive targets for the design of antiviral agents. Here, we develop and evaluate new bis(benzylidene)cyclohexanones (BBC) and identify potential antiviral compounds. Three compounds were found to be effective in reducing the SARS-CoV-2 load, with EC50 values in the low micromolar concentration range. However, these compounds also exhibited inhibitory activity IC50 against PLpro at approximately 10-fold higher micromolar concentrations. Although originally developed as PLpro inhibitors, the comparison between IC50 and EC50 of BBC indicates that the mechanism of their in vitro antiviral activity is probably not directly related to inhibition of viral cysteine proteases. In conclusion, our study has identified new potential noncytotoxic antiviral compounds suitable for in vivo testing and further improvement.


Subject(s)
COVID-19 , Cysteine Proteases , Humans , SARS-CoV-2 , Cysteine Endopeptidases/metabolism , Viral Nonstructural Proteins/chemistry , Antiviral Agents/pharmacology , Antiviral Agents/chemistry , Protease Inhibitors/pharmacology , Protease Inhibitors/chemistry , Molecular Docking Simulation
2.
Article in English | MEDLINE | ID: mdl-31293981

ABSTRACT

The M3 protein (M3) encoded by murine gammaherpesvirus 68 (MHV-68) is a unique viral immunomodulator with a high-affinity for a broad spectrum of chemokines, key mediators responsible for the migration of immune cells to sites of inflammation. M3 is currently being studied as a very attractive and desirable tool for blocking the chemokine signaling involved in some inflammatory diseases and cancers. In this study, we elucidated the role of M3 residues E70 and T272 in binding to chemokines by examining the effects of the E70A and T272G mutations on the ability of recombinant M3, prepared in Escherichia coli cells, to bind the human chemokines CCL5 and CXCL8. We found that the E70A mutation enhanced binding of M3 to CCL5 two-fold but had little effect on its binding to CXCL8. In contrast, the T272G mutation was found to be important for the thermal stability of M3 and significantly decreased M3's binding to both CCL5 (by about 4×) and CXCL8 (by about 5×). We also constructed in silico models of the wild-type M3-CCL5 and M3-CCL8 complexes and found substantial differences in their physical and chemical properties. M3 models with single mutation E70A and T272G suggested the role of E70 and T272 in binding M3 protein to chemokines. In sum, we have confirmed that site-directed mutagenesis could be an effective tool for modulating the blockade of particular chemokines by M3, as desired in therapeutic treatments for severe inflammatory illnesses arising from chemokine network dysregulation.


Subject(s)
Chemokines/metabolism , Mutation , Protein Binding , Rhadinovirus/genetics , Viral Proteins/genetics , Viral Proteins/immunology , Amino Acid Sequence , Animals , Cell Line , Chemokine CCL5/metabolism , Cloning, Molecular , Escherichia coli/genetics , Escherichia coli/metabolism , Gene Expression Regulation, Bacterial , Humans , Immunologic Factors/genetics , Immunologic Factors/immunology , Interleukin-8 , Models, Molecular , Mutagenesis, Site-Directed , Protein Conformation , Protein Structure, Tertiary , Recombinant Proteins/chemistry , Recombinant Proteins/genetics , Recombinant Proteins/immunology , Recombinant Proteins/metabolism , Viral Proteins/chemistry
3.
J Biol Chem ; 294(27): 10519-10529, 2019 07 05.
Article in English | MEDLINE | ID: mdl-31126984

ABSTRACT

Human cytomegalovirus (HCMV) is a ß-herpesvirus that has co-evolved with the host immune system to establish lifelong persistence. HCMV encodes many immunomodulatory molecules, including the glycoprotein UL144. UL144 is a structural mimic of the tumor necrosis factor receptor superfamily member HVEM (herpesvirus entry mediator), which binds to the various ligands LIGHT, LTα, BTLA, CD160, and gD. However, in contrast to HVEM, UL144 only binds BTLA, inhibiting T-cell activation. Here, we report the crystal structure of the UL144-BTLA complex, revealing that UL144 utilizes residues from its N-terminal cysteine-rich domain 1 (CRD1) to interact uniquely with BTLA. The shorter CRD2 loop of UL144 also alters the relative orientation of BTLA binding with both N-terminal CRDs. By employing structure-guided mutagenesis, we have identified a mutant of BTLA (L123A) that interferes with HVEM binding but preserves UL144 interactions. Furthermore, our results illuminate structural differences between UL144 and HVEM that explain its binding selectivity and highlight it as a suitable scaffold for designing superior, immune inhibitory BTLA agonists.


Subject(s)
Cytomegalovirus/metabolism , Membrane Glycoproteins/chemistry , Receptors, Immunologic/metabolism , Receptors, Tumor Necrosis Factor, Member 14/chemistry , Viral Proteins/chemistry , Amino Acid Sequence , Binding Sites , Crystallography, X-Ray , Humans , Membrane Glycoproteins/metabolism , Mutagenesis, Site-Directed , Protein Binding , Receptors, Immunologic/chemistry , Receptors, Immunologic/genetics , Receptors, Tumor Necrosis Factor, Member 14/metabolism , Recombinant Proteins/biosynthesis , Recombinant Proteins/chemistry , Recombinant Proteins/genetics , Sequence Alignment , Viral Proteins/metabolism
4.
J Biol Chem ; 292(51): 21060-21070, 2017 12 22.
Article in English | MEDLINE | ID: mdl-29061848

ABSTRACT

The human cytomegalovirus opening reading frame UL144 is an ortholog of the TNF receptor superfamily member, herpesvirus entry mediator (HVEM; TNFRSF14). HVEM binds the TNF ligands, LIGHT and LTa; the immunoglobulin inhibitory receptor, B and T lymphocyte attenuator (BTLA); and the natural killer cell-activating receptor CD160. However, UL144 selectively binds BTLA, avoiding activation of inflammatory signaling initiated by CD160 in natural killer cells. BTLA and CD160 cross-compete for binding HVEM, but the structural basis for the ligand selectivity by UL144 and how it acts as an anti-inflammatory agonist remains unclear. Here, we modeled the UL144 structure and characterized its binding with BTLA. The UL144 structure was predicted to closely mimic the surface of HVEM, and we also found that both HVEM and UL144 bind a common epitope of BTLA, whether engaged in trans or in cis, that is shared with a BTLA antibody agonist. On the basis of the UL144 selectivity, we engineered a BTLA-selective HVEM protein to understand the basis for ligand selectivity and BTLA agonism to develop novel anti-inflammatory agonists. This HVEM mutein did not bind CD160 or TNF ligands but did bind BTLA with 10-fold stronger affinity than wild-type HVEM and retained potent inhibitory activity that reduced T-cell receptor, B-cell receptor, and interferon signaling in B cells. In conclusion, using a viral immune evasion strategy that shows broad immune-ablating activity, we have identified a novel anti-inflammatory BTLA-selective agonist.


Subject(s)
B-Lymphocytes/metabolism , Killer Cells, Natural/metabolism , Membrane Glycoproteins/metabolism , Models, Molecular , Receptors, Immunologic/agonists , Receptors, Tumor Necrosis Factor, Member 14/metabolism , T-Lymphocytes/metabolism , Viral Proteins/metabolism , Amino Acid Substitution , Anti-Inflammatory Agents, Non-Steroidal/chemistry , Anti-Inflammatory Agents, Non-Steroidal/metabolism , Anti-Inflammatory Agents, Non-Steroidal/pharmacology , Antigens, CD/chemistry , Antigens, CD/genetics , Antigens, CD/metabolism , B-Lymphocytes/cytology , B-Lymphocytes/drug effects , B-Lymphocytes/immunology , Binding Sites , Cell Line, Tumor , Drug Design , GPI-Linked Proteins/chemistry , GPI-Linked Proteins/genetics , GPI-Linked Proteins/metabolism , HEK293 Cells , Humans , Killer Cells, Natural/cytology , Killer Cells, Natural/drug effects , Killer Cells, Natural/immunology , Kinetics , Ligands , Membrane Glycoproteins/chemistry , Membrane Glycoproteins/genetics , Mutation , Protein Conformation , Protein Engineering , Protein Interaction Domains and Motifs , Receptors, Immunologic/chemistry , Receptors, Immunologic/genetics , Receptors, Immunologic/metabolism , Receptors, Tumor Necrosis Factor, Member 14/chemistry , Receptors, Tumor Necrosis Factor, Member 14/genetics , Recombinant Fusion Proteins/chemistry , Recombinant Fusion Proteins/metabolism , Recombinant Fusion Proteins/pharmacology , T-Lymphocytes/cytology , T-Lymphocytes/drug effects , T-Lymphocytes/immunology , Viral Proteins/chemistry , Viral Proteins/genetics
5.
Biochemistry ; 55(19): 2735-47, 2016 05 17.
Article in English | MEDLINE | ID: mdl-27149221

ABSTRACT

Mammalian ß-hexosaminidases have been shown to play essential roles in cellular physiology and health. These enzymes are responsible for the cleavage of the monosaccharides N-acetylglucosamine (GlcNAc) and N-acetylgalactosamine (GalNAc) from cellular substrates. One of these ß-hexosaminidases, hexosaminidase D (HexD), encoded by the HEXDC gene, has received little attention. No mechanistic studies have focused on the role of this unusual nucleocytoplasmically localized ß-hexosaminidase, and its cellular function remains unknown. Using a series of kinetic and mechanistic investigations into HexD, we define the precise catalytic mechanism of this enzyme and establish the identities of key enzymic residues. The preparation of synthetic aryl N-acetylgalactosaminide substrates for HexD in combination with measurements of kinetic parameters for wild-type and mutant enzymes, linear free energy analyses of the enzyme-catalyzed hydrolysis of these substrates, evaluation of the reaction by nuclear magnetic resonance, and inhibition studies collectively reveal the detailed mechanism of action employed by HexD. HexD is a retaining glycosidase that operates using a substrate-assisted catalytic mechanism, has a preference for galactosaminide over glucosaminide substrates, and shows a pH optimum in its second-order rate constant at pH 6.5-7.0. The catalytically important residues are Asp148 and Glu149, with Glu149 serving as the general acid/base residue and Asp148 as the polarizing residue. HexD is inhibited by Gal-NAG-thiazoline (Ki = 420 nM). The fundamental insights gained from this study will aid in the development of potent and selective probes for HexD, which will serve as useful tools to improve our understanding of the physiological role played by this unusual enzyme.


Subject(s)
Enzyme Inhibitors/chemistry , Hexosaminidases/antagonists & inhibitors , Hexosaminidases/chemistry , Thiazolidines/chemistry , Catalysis , Hexosaminidases/genetics , Hexosaminidases/metabolism , Humans , Hydrogen-Ion Concentration , Hydrolysis , Kinetics
6.
J Exp Med ; 211(7): 1433-48, 2014 Jun 30.
Article in English | MEDLINE | ID: mdl-24958847

ABSTRACT

Biologics to TNF family receptors are prime candidates for therapy of immune disease. Whereas recent studies have highlighted a requirement for Fcγ receptors in enabling the activity of CD40, TRAILR, and GITR when engaged by antibodies, other TNFR molecules may be controlled by additional mechanisms. Antibodies to 4-1BB (CD137) are currently in clinical trials and can both augment immunity in cancer and promote regulatory T cells that inhibit autoimmune disease. We found that the action of agonist anti-4-1BB in suppressing autoimmune and allergic inflammation was completely dependent on Galectin-9 (Gal-9). Gal-9 directly bound to 4-1BB, in a site distinct from the binding site of antibodies and the natural ligand of 4-1BB, and Gal-9 facilitated 4-1BB aggregation, signaling, and functional activity in T cells, dendritic cells, and natural killer cells. Conservation of the Gal-9 interaction in humans has important implications for effective clinical targeting of 4-1BB and possibly other TNFR superfamily molecules.


Subject(s)
Antibodies/pharmacology , Autoimmune Diseases/drug therapy , Galectins/immunology , Hypersensitivity/drug therapy , Neoplasms/drug therapy , Tumor Necrosis Factor Receptor Superfamily, Member 9/antagonists & inhibitors , Animals , Antibodies/immunology , Autoimmune Diseases/genetics , Autoimmune Diseases/immunology , Galectins/genetics , Humans , Hypersensitivity/genetics , Hypersensitivity/immunology , Mice , Mice, Knockout , Neoplasms/genetics , Neoplasms/immunology , T-Lymphocytes, Regulatory/immunology , Tumor Necrosis Factor Receptor Superfamily, Member 9/genetics , Tumor Necrosis Factor Receptor Superfamily, Member 9/immunology
7.
Acta Crystallogr D Biol Crystallogr ; 70(Pt 3): 851-62, 2014 Mar.
Article in English | MEDLINE | ID: mdl-24598754

ABSTRACT

Natural killer (NK) cells are critical components of the innate immune system as they rapidly detect and destroy infected cells. To avoid immune recognition and to allow long-term persistence in the host, Human cytomegalovirus (HCMV) has evolved a number of genes to evade or inhibit immune effector pathways. In particular, UL141 can inhibit cell-surface expression of both the NK cell-activating ligand CD155 as well as the TRAIL death receptors (TRAIL-R1 and TRAIL-R2). The crystal structure of unliganded HCMV UL141 refined to 3.25 Šresolution allowed analysis of its head-to-tail dimerization interface. A `dimerization-deficient' mutant of UL141 (ddUL141) was further designed, which retained the ability to bind to TRAIL-R2 or CD155 while losing the ability to cross-link two receptor monomers. Structural comparison of unliganded UL141 with UL141 bound to TRAIL-R2 further identified a mobile loop that makes intimate contacts with TRAIL-R2 upon receptor engagement. Superposition of the Ig-like domain of UL141 on the CD155 ligand T-cell immunoreceptor with Ig and ITIM domains (TIGIT) revealed that UL141 can potentially engage CD155 similar to TIGIT by using the C'C'' and GF loops. Further mutations in the TIGIT binding site of CD155 (Q63R and F128R) abrogated UL141 binding, suggesting that the Ig-like domain of UL141 is a viral mimic of TIGIT, as it targets the same binding site on CD155 using similar `lock-and-key' interactions. Sequence alignment of the UL141 gene and its orthologues also showed conservation in this highly hydrophobic (L/A)X6G `lock' motif for CD155 binding as well as conservation of the TRAIL-R2 binding patches, suggesting that these host-receptor interactions are evolutionary conserved.


Subject(s)
Cytomegalovirus/chemistry , Immunoglobulins/chemistry , Membrane Glycoproteins/chemistry , Receptors, TNF-Related Apoptosis-Inducing Ligand/chemistry , Receptors, Virus/chemistry , Viral Proteins/chemistry , Animals , Crystallography, X-Ray , Cytomegalovirus/genetics , Cytomegalovirus/immunology , Evolution, Molecular , Humans , Immunoglobulins/genetics , Immunologic Factors/chemistry , Immunologic Factors/genetics , Killer Cells, Natural/chemistry , Killer Cells, Natural/immunology , Killer Cells, Natural/metabolism , Membrane Glycoproteins/genetics , Membrane Glycoproteins/immunology , Mutation , Protein Binding/genetics , Protein Binding/immunology , Protein Folding , Receptors, TNF-Related Apoptosis-Inducing Ligand/genetics , Receptors, Virus/genetics , Sf9 Cells , Signal Transduction/genetics , Signal Transduction/immunology , Spodoptera/genetics , Viral Proteins/genetics , Viral Proteins/immunology
8.
Glycoconj J ; 30(9): 899-909, 2013 Dec.
Article in English | MEDLINE | ID: mdl-23979800

ABSTRACT

Homology searches indicated that up to five class I α-mannosidases (glycohydrolase family 47) and eight class II α-mannosidases (glycohydrolase family 38) are encoded by the fruitfly (Drosophila melanogaster) genome. Selected example mannosidases were expressed in secreted form using the yeast Pichia pastoris. A number of characteristics of these enzymes were determined with p-nitrophenyl-α-mannoside as substrate; particularly striking were the low optima (pH 5) of three class II mannosidases most closely related to known lysosomal mannosidases and the distinct Co(II)-requirement of a mannosidase previously named ManIIb. Some of the recombinant mannosidases were demonstrably active towards oligomannosidic glycans, specifically, the Co(II)-requiring ManIIb, two 'acidic' mannosidases and the class I mas-1 mannosidase. Other than previous characterisations of the well-known Golgi mannosidase II, this is the first study summarising various properties of recombinant mannosidases from the fruitfly.


Subject(s)
Drosophila Proteins/chemistry , Drosophila melanogaster/enzymology , alpha-Mannosidase/chemistry , Amino Acid Sequence , Animals , Drosophila Proteins/genetics , Drosophila Proteins/metabolism , Kinetics , Molecular Sequence Data , Polysaccharides/metabolism , Substrate Specificity , alpha-Mannosidase/genetics , alpha-Mannosidase/metabolism
9.
PLoS Pathog ; 9(3): e1003224, 2013 Mar.
Article in English | MEDLINE | ID: mdl-23555243

ABSTRACT

The TRAIL (TNF-related apoptosis inducing ligand) death receptors (DRs) of the tumor necrosis factor receptor superfamily (TNFRSF) can promote apoptosis and regulate antiviral immunity by maintaining immune homeostasis during infection. In turn, human cytomegalovirus (HCMV) expresses immunomodulatory proteins that down-regulate cell surface expression of TNFRSF members as well as poliovirus receptor-related proteins in an effort to inhibit host immune effector pathways that would lead to viral clearance. The UL141 glycoprotein of human cytomegalovirus inhibits host defenses by blocking cell surface expression of TRAIL DRs (by retention in ER) and poliovirus receptor CD155, a nectin-like Ig-fold molecule. Here we show that the immunomodulatory function of HCMV UL141 is associated with its ability to bind diverse proteins, while utilizing at least two distinct binding sites to selectively engage TRAIL DRs or CD155. Binding studies revealed high affinity interaction of UL141 with both TRAIL-R2 and CD155 and low affinity binding to TRAIL-R1. We determined the crystal structure of UL141 bound to TRAIL-R2 at 2.1 Å resolution, which revealed that UL141 forms a homodimer that engages two TRAIL-R2 monomers 90° apart to form a heterotetrameric complex. Our structural and biochemical data reveal that UL141 utilizes its Ig-domain to facilitate non-canonical death receptor interactions while UL141 partially mimics the binding site of TRAIL on TRAIL-R2, which we found to be distinct from that of CD155. Moreover, UL141 also binds to an additional surface patch on TRAIL-R2 that is distinct from the TRAIL binding site. Therefore, the breadth of UL141-mediated effects indicates that HCMV has evolved sophisticated strategies to evade the immune system by modulating multiple effector pathways.


Subject(s)
Cytomegalovirus/metabolism , Death Domain Receptor Signaling Adaptor Proteins/metabolism , Receptors, Death Domain/metabolism , Receptors, TNF-Related Apoptosis-Inducing Ligand/metabolism , Binding Sites , Cytomegalovirus/chemistry , Death Domain Receptor Signaling Adaptor Proteins/chemistry , Host-Pathogen Interactions , Humans , Immune Evasion/genetics , Immunomodulation , Mutation , Protein Binding , Protein Folding , Protein Structure, Tertiary , Receptors, Death Domain/chemistry , Receptors, TNF-Related Apoptosis-Inducing Ligand/chemistry , Receptors, TNF-Related Apoptosis-Inducing Ligand/genetics
10.
Cell Host Microbe ; 13(3): 324-35, 2013 Mar 13.
Article in English | MEDLINE | ID: mdl-23498957

ABSTRACT

Death receptors (DRs) of the TNFR superfamily contribute to antiviral immunity by promoting apoptosis and regulating immune homeostasis during infection, and viral inhibition of DR signaling can alter immune defenses. Here we identify the human cytomegalovirus (HCMV) UL141 glycoprotein as necessary and sufficient to restrict TRAIL DR function. Despite showing no primary sequence homology to TNF family cytokines, UL141 binds the ectodomains of both human TRAIL DRs with affinities comparable to the natural ligand TRAIL. UL141 binding promotes intracellular retention of the DRs, thus protecting virus infected cells from TRAIL and TRAIL-dependent NK cell-mediated killing. The identification of UL141 as a herpesvirus modulator of the TRAIL DRs strongly implicates this pathway as a regulator of host defense to HCMV and highlights UL141 as a pleiotropic inhibitor of NK cell effector function.


Subject(s)
Cytomegalovirus Infections/immunology , Cytomegalovirus/immunology , Immunity, Innate , Membrane Glycoproteins/immunology , Membrane Glycoproteins/metabolism , Receptors, TNF-Related Apoptosis-Inducing Ligand/immunology , Viral Proteins/immunology , Viral Proteins/metabolism , Cytomegalovirus/genetics , Cytomegalovirus/metabolism , Cytomegalovirus Infections/genetics , Cytomegalovirus Infections/metabolism , Cytomegalovirus Infections/virology , Host-Pathogen Interactions , Humans , Killer Cells, Natural/immunology , Membrane Glycoproteins/genetics , Protein Binding , Receptors, TNF-Related Apoptosis-Inducing Ligand/genetics , Receptors, TNF-Related Apoptosis-Inducing Ligand/metabolism , Viral Proteins/genetics
SELECTION OF CITATIONS
SEARCH DETAIL
...