1
|
Yeom JH, Shin E, Jin H, Liu H, Luo Y, Nam Y, Ryu M, Song W, Chi H, Kim J, Lee K, Bae J. Aptamer-conjugated gold nanoparticles platform as the intracellular delivery of antibodies for cancer therapy. J IND ENG CHEM 2023; 126:480-491. [DOI: 10.1016/j.jiec.2023.06.036] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2024]
|
2
|
McKellar J, Arnaud-Arnould M, Chaloin L, Tauziet M, Arpin-André C, Pourcelot O, Blaise M, Moncorgé O, Goujon C. An evolutionarily conserved N-terminal leucine is essential for MX1 GTPase antiviral activity against different families of RNA viruses. J Biol Chem 2023; 299:102747. [PMID: 36436557 PMCID: PMC9808005 DOI: 10.1016/j.jbc.2022.102747] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2022] [Revised: 11/14/2022] [Accepted: 11/16/2022] [Indexed: 11/27/2022] Open
Abstract
Myxovirus resistance protein 1 (MX1) and MX2 are homologous, dynamin-like large GTPases, induced upon interferon exposure. Human MX1 (HsMX1) is known to inhibit many viruses, including influenza A virus, by likely acting at various steps of their life cycles. Despite decades of studies, the mechanism(s) of action with which MX1 proteins manage to inhibit target viruses is not fully understood. MX1 proteins are mechano-enzymes and share a similar organization to dynamin, with a GTPase domain and a carboxy-terminal stalk domain, connected by a bundle signaling element. These three elements are known to be essential for antiviral activity. HsMX1 has two unstructured regions, the L4 loop, also essential for antiviral activity, and a short amino (N)-terminal region, which greatly varies between MX1 proteins of different species. The role of this N-terminal domain in antiviral activity is not known. Herein, using mutagenesis, imaging, and biochemical approaches, we demonstrate that the N-terminal domain of HsMX1 is essential for antiviral activity against influenza A virus, Vesicular Stomatitis Virus, and La Crosse virus. Furthermore, we pinpoint a highly conserved leucine within this region, which is absolutely crucial for human, mouse, and bat MX1 protein antiviral activity. Importantly, mutation of this leucine does not compromise GTPase activity or oligomerization capabilities but does modify MX1 protein subcellular localization. The discovery of this essential and highly conserved residue defines this region as key for antiviral activity and may reveal insights as to the mechanism(s) of action of MX1 proteins.
Collapse
Affiliation(s)
- Joe McKellar
- IRIM, CNRS, Montpellier University, Montpellier, France
| | | | | | | | | | | | | | | | | |
Collapse
|
3
|
Anti-Myxovirus Resistance Protein-1 Immunoglobulin A Autoantibody in Idiopathic Pulmonary Fibrosis. Can Respir J 2022; 2022:1107673. [PMID: 35391716 PMCID: PMC8983265 DOI: 10.1155/2022/1107673] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2021] [Revised: 01/22/2022] [Accepted: 02/07/2022] [Indexed: 12/02/2022] Open
Abstract
Background We have previously analysed serum autoantibody levels in patients with idiopathic pulmonary fibrosis (IPF), idiopathic nonspecific interstitial pneumonia (iNSIP), and healthy controls and identified the autoantibody against anti-myxovirus resistance protein-1 (MX1) to be a specific autoantibody in iNSIP. We found that a higher anti-MX1 autoantibody level was a significant predictor of a good prognosis in patients with non-IPF idiopathic interstitial pneumonias. In this retrospective study, we sought to clarify the prognostic significance of the anti-MX1 autoantibody in IPF. Methods We measured anti-MX1 immunoglobulin (Ig) G, IgA, and IgM autoantibody levels by enzyme-linked immunosorbent assay in serum collected at the time of diagnosis from 71 patients with IPF diagnosed according to the 2018 IPF guideline. The gender-age-physiology (GAP) index was calculated in each case. Results The study population (59 men and 12 women) had a median age of 67 years. Serum anti-MX1 IgG and IgA autoantibody levels correlated positively with GAP stage (p < 0.05). Univariate Cox proportional hazards regression analysis did not identify an elevated anti-MX1 IgG, IgA, or IgM autoantibody level as a significant prognostic factor; however, a higher anti-MX1 IgA autoantibody level heralded significantly poorer survival after adjustment for GAP stage (p=0.030) and for percent forced vital capacity and modified Medical Research Council score (p=0.018). Neither the anti-MX1 IgG autoantibody nor the IgM autoantibody could predict survival after these adjustments. Conclusions The serum anti-MX1 IgA autoantibody level is a significant prognostic factor in IPF. Further studies are needed to clarify the pathophysiological role of this autoantibody in IPF.
Collapse
|
4
|
Soetens E, Ballegeer M, Saelens X. An Inside Job: Applications of Intracellular Single Domain Antibodies. Biomolecules 2020; 10:biom10121663. [PMID: 33322697 PMCID: PMC7764588 DOI: 10.3390/biom10121663] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2020] [Revised: 12/09/2020] [Accepted: 12/10/2020] [Indexed: 02/06/2023] Open
Abstract
Sera of camelid species contain a special kind of antibody that consists only of heavy chains. The variable antigen binding domain of these heavy chain antibodies can be expressed as a separate entity, called a single domain antibody that is characterized by its small size, high solubility and oftentimes exceptional stability. Because of this, most single domain antibodies fold correctly when expressed in the reducing environment of the cytoplasm, and thereby retain their antigen binding specificity. Single domain antibodies can thus be used to target a broad range of intracellular proteins. Such intracellular single domain antibodies are also known as intrabodies, and have proven to be highly useful tools for basic research by allowing visualization, disruption and even targeted degradation of intracellular proteins. Furthermore, intrabodies can be used to uncover prospective new therapeutic targets and have the potential to be applied in therapeutic settings in the future. In this review we provide a brief overview of recent advances in the field of intracellular single domain antibodies, focusing on their use as research tools and potential therapeutic applications. Special attention is given to the available methods that allow delivery of single domain antibodies into cells.
Collapse
Affiliation(s)
- Eline Soetens
- VIB-UGent Center for Medical Biotechnology, VIB, B-9052 Ghent, Belgium; (E.S.); (M.B.)
- Department of Biochemistry and Microbiology, Ghent University, B-9000 Ghent, Belgium
| | - Marlies Ballegeer
- VIB-UGent Center for Medical Biotechnology, VIB, B-9052 Ghent, Belgium; (E.S.); (M.B.)
- Department of Biochemistry and Microbiology, Ghent University, B-9000 Ghent, Belgium
| | - Xavier Saelens
- VIB-UGent Center for Medical Biotechnology, VIB, B-9052 Ghent, Belgium; (E.S.); (M.B.)
- Department of Biochemistry and Microbiology, Ghent University, B-9000 Ghent, Belgium
- Correspondence:
| |
Collapse
|
5
|
Singh K, Ejaz W, Dutta K, Thayumanavan S. Antibody Delivery for Intracellular Targets: Emergent Therapeutic Potential. Bioconjug Chem 2019; 30:1028-1041. [PMID: 30830750 PMCID: PMC6470022 DOI: 10.1021/acs.bioconjchem.9b00025] [Citation(s) in RCA: 65] [Impact Index Per Article: 10.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
Abstract
Proteins have sparked fast growing interest as biological therapeutic agents for several diseases. Antibodies, in particular, carry an enormous potential as drugs owing to their remarkable target specificity and low immunogenicity. Although the market has numerous antibodies directed toward extracellular targets, their use in targeting therapeutically important intracellular targets is limited by their inability to cross cellular membrane. Realizing the potential for antibody therapy in disease treatment, progress has been made in the development of methods to deliver antibodies intracellularly. In this review, we address various platforms for delivery of antibodies and their merits and drawbacks.
Collapse
|
6
|
Haller O, Arnheiter H, Pavlovic J, Staeheli P. The Discovery of the Antiviral Resistance Gene Mx: A Story of Great Ideas, Great Failures, and Some Success. Annu Rev Virol 2018; 5:33-51. [PMID: 29958082 DOI: 10.1146/annurev-virology-092917-043525] [Citation(s) in RCA: 31] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
The discovery of the Mx gene-dependent, innate resistance of mice against influenza virus was a matter of pure chance. Although the subsequent analysis of this antiviral resistance was guided by straightforward logic, it nevertheless led us into many blind alleys and was full of surprising turns and twists. Unexpectedly, this research resulted in the identification of one of the first interferon-stimulated genes and provided a new view of interferon action. It also showed that in many species, MX proteins have activities against a broad range of viruses. To this day, Mx research continues to flourish and to provide insights into the never-ending battle between viruses and their hosts.
Collapse
Affiliation(s)
- Otto Haller
- Institute of Virology, Medical Center University of Freiburg, D-79104 Freiburg, Germany; .,Faculty of Medicine, University of Freiburg, D-79104 Freiburg, Germany
| | - Heinz Arnheiter
- National Institute of Neurological Disorders and Stroke, National Institutes of Health, Bethesda, Maryland 20892, USA
| | - Jovan Pavlovic
- Institute of Medical Virology, University of Zürich, 8057 Zürich, Switzerland
| | - Peter Staeheli
- Institute of Virology, Medical Center University of Freiburg, D-79104 Freiburg, Germany; .,Faculty of Medicine, University of Freiburg, D-79104 Freiburg, Germany
| |
Collapse
|
7
|
Van der Poorten O, Legrand B, Vezenkov LL, García-Pindado J, Bettache N, Knuhtsen A, Pedersen DS, Sánchez-Navarro M, Martinez J, Teixidó M, Garcia M, Tourwé D, Amblard M, Ballet S. Indoloazepinone-Constrained Oligomers as Cell-Penetrating and Blood-Brain-Barrier-Permeating Compounds. Chembiochem 2018; 19:696-705. [PMID: 29377388 DOI: 10.1002/cbic.201700678] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2017] [Indexed: 12/29/2022]
Abstract
Non-cationic and amphipathic indoloazepinone-constrained (Aia) oligomers have been synthesized as new vectors for intracellular delivery. The conformational preferences of the [l-Aia-Xxx]n oligomers were investigated by circular dichroism (CD) and NMR spectroscopy. Whereas Boc-[l-Aia-Gly]2,4 -OBn oligomers 12 and 13 and Boc-[l-Aia-β3 -h-l-Ala]2,4 -OBn oligomers 16 and 17 were totally or partially disordered, Boc-[l-Aia-l-Ala]2 -OBn (14) induced a typical turn stabilized by C5 - and C7 -membered H-bond pseudo-cycles and aromatic interactions. Boc-[l-Aia-l-Ala]4 -OBn (15) exhibited a unique structure with remarkable T-shaped π-stacking interactions involving the indole rings of the four l-Aia residues forming a dense hydrophobic cluster. All of the proposed FITC-6-Ahx-[l-Aia-Xxx]4 -NH2 oligomers 19-23, with the exception of FITC-6-Ahx-[l-Aia-Gly]4 -NH2 (18), were internalized by MDA-MB-231 cells with higher efficiency than the positive references penetratin and Arg8 . In parallel, the compounds of this series were successfully explored in an in vitro blood-brain barrier (BBB) permeation assay. Although no passive diffusion permeability was observed for any of the tested Ac-[l-Aia-Xxx]4 -NH2 oligomers in the PAMPA model, Ac-[l-Aia-l-Arg]4 -NH2 (26) showed significant permeation in the in vitro cell-based human model of the BBB, suggesting an active mechanism of cell penetration.
Collapse
Affiliation(s)
- Olivier Van der Poorten
- Research Group of Organic Chemistry, Departments of Chemistry and Bioengineering Sciences, Vrije Universiteit Brussel, Pleinlaan 2, 1050, Brussels, Belgium
| | - Baptiste Legrand
- Institut des Biomolécules Max Mousseron (IBMM), UMR 5247, Université de Montpellier, CNRS, ENSCM, 15 Avenue Charles Flahault, 34093, Montpellier, Cedex 5, France
| | - Lubomir L Vezenkov
- Institut des Biomolécules Max Mousseron (IBMM), UMR 5247, Université de Montpellier, CNRS, ENSCM, 15 Avenue Charles Flahault, 34093, Montpellier, Cedex 5, France
| | - Júlia García-Pindado
- Institute for Research in Biomedicine (IRB Barcelona), The Barcelona Institute of Science and Technology (BIST), Baldiri Reixac 10, 08028, Barcelona, Spain
| | - Nadir Bettache
- Institut des Biomolécules Max Mousseron (IBMM), UMR 5247, Université de Montpellier, CNRS, ENSCM, 15 Avenue Charles Flahault, 34093, Montpellier, Cedex 5, France
| | - Astrid Knuhtsen
- Department of Drug Design and Pharmacology, University of Copenhagen, Jagtvej 162, 2100, Copenhagen, Denmark
| | - Daniel Sejer Pedersen
- Department of Drug Design and Pharmacology, University of Copenhagen, Jagtvej 162, 2100, Copenhagen, Denmark
| | - Macarena Sánchez-Navarro
- Institute for Research in Biomedicine (IRB Barcelona), The Barcelona Institute of Science and Technology (BIST), Baldiri Reixac 10, 08028, Barcelona, Spain
| | - Jean Martinez
- Institut des Biomolécules Max Mousseron (IBMM), UMR 5247, Université de Montpellier, CNRS, ENSCM, 15 Avenue Charles Flahault, 34093, Montpellier, Cedex 5, France
| | - Meritxell Teixidó
- Institute for Research in Biomedicine (IRB Barcelona), The Barcelona Institute of Science and Technology (BIST), Baldiri Reixac 10, 08028, Barcelona, Spain
| | - Marcel Garcia
- Institut des Biomolécules Max Mousseron (IBMM), UMR 5247, Université de Montpellier, CNRS, ENSCM, 15 Avenue Charles Flahault, 34093, Montpellier, Cedex 5, France
| | - Dirk Tourwé
- Research Group of Organic Chemistry, Departments of Chemistry and Bioengineering Sciences, Vrije Universiteit Brussel, Pleinlaan 2, 1050, Brussels, Belgium
| | - Muriel Amblard
- Institut des Biomolécules Max Mousseron (IBMM), UMR 5247, Université de Montpellier, CNRS, ENSCM, 15 Avenue Charles Flahault, 34093, Montpellier, Cedex 5, France
| | - Steven Ballet
- Research Group of Organic Chemistry, Departments of Chemistry and Bioengineering Sciences, Vrije Universiteit Brussel, Pleinlaan 2, 1050, Brussels, Belgium
| |
Collapse
|
8
|
|
9
|
Yamada Y, Perez SMV, Tabata M, Abe J, Yasuzaki Y, Harashima H. Efficient and High-Speed Transduction of an Antibody into Living Cells Using a Multifunctional Nanocarrier System to Control Intracellular Trafficking. J Pharm Sci 2015; 104:2845-54. [DOI: 10.1002/jps.24310] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2014] [Revised: 11/13/2014] [Accepted: 11/21/2014] [Indexed: 12/24/2022]
|
10
|
|
11
|
Funamoto D, Asai D, Sato K, Yamaguchi Y, Kim CW, Sato H, Nakhaei E, Matsumoto S, Yoshikawa T, Sasaki K, Yamamoto T, Kishimura A, Mori T, Katayama Y. Antibody Internalization into Living Cells via Crosslinker-mediated Endocytosis. CHEM LETT 2015. [DOI: 10.1246/cl.141157] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022]
Affiliation(s)
- Daiki Funamoto
- Graduate School of System Life Sciences, Kyushu University
| | - Daisuke Asai
- Department of Microbiology, St. Marianna University School of Medicine
| | - Kazuki Sato
- Department of Environmental Science, Fukuoka Women’s University
| | - Yoko Yamaguchi
- Department of Environmental Science, Fukuoka Women’s University
| | - Chan Woo Kim
- Graduate School of System Life Sciences, Kyushu University
| | - Hikari Sato
- Graduate School of System Life Sciences, Kyushu University
| | - Elnaz Nakhaei
- Graduate School of System Life Sciences, Kyushu University
| | | | | | - Koichi Sasaki
- Graduate School of System Life Sciences, Kyushu University
| | | | | | - Takeshi Mori
- Graduate School of System Life Sciences, Kyushu University
| | | |
Collapse
|
12
|
Haller O, Staeheli P, Schwemmle M, Kochs G. Mx GTPases: dynamin-like antiviral machines of innate immunity. Trends Microbiol 2015; 23:154-63. [PMID: 25572883 DOI: 10.1016/j.tim.2014.12.003] [Citation(s) in RCA: 346] [Impact Index Per Article: 34.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2014] [Revised: 11/28/2014] [Accepted: 12/03/2014] [Indexed: 01/09/2023]
Abstract
The Mx dynamin-like GTPases are key antiviral effector proteins of the type I and type III interferon (IFN) systems. They inhibit several different viruses by blocking early steps of the viral replication cycle. We focus on new structural and functional insights and discuss recent data revealing that human MxA (MX1) provides a safeguard against introduction of avian influenza A viruses (FLUAV) into the human population. The related human MxB (MX2) serves as restriction factor for HIV-1 and other primate lentiviruses.
Collapse
Affiliation(s)
- Otto Haller
- Institute of Virology, University Medical Center Freiburg, Freiburg, Germany.
| | - Peter Staeheli
- Institute of Virology, University Medical Center Freiburg, Freiburg, Germany.
| | - Martin Schwemmle
- Institute of Virology, University Medical Center Freiburg, Freiburg, Germany
| | - Georg Kochs
- Institute of Virology, University Medical Center Freiburg, Freiburg, Germany
| |
Collapse
|
13
|
Mx proteins: antiviral gatekeepers that restrain the uninvited. Microbiol Mol Biol Rev 2014; 77:551-66. [PMID: 24296571 DOI: 10.1128/mmbr.00024-13] [Citation(s) in RCA: 229] [Impact Index Per Article: 20.8] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022] Open
Abstract
Fifty years after the discovery of the mouse Mx1 gene, researchers are still trying to understand the molecular details of the antiviral mechanisms mediated by Mx proteins. Mx proteins are evolutionarily conserved dynamin-like large GTPases, and GTPase activity is required for their antiviral activity. The expression of Mx genes is controlled by type I and type III interferons. A phylogenetic analysis revealed that Mx genes are present in almost all vertebrates, usually in one to three copies. Mx proteins are best known for inhibiting negative-stranded RNA viruses, but they also inhibit other virus families. Recent structural analyses provide hints about the antiviral mechanisms of Mx proteins, but it is not known how they can suppress such a wide variety of viruses lacking an obvious common molecular pattern. Perhaps they interact with a (partially) symmetrical invading oligomeric structure, such as a viral ribonucleoprotein complex. Such an interaction may be of a fairly low affinity, in line with the broad target specificity of Mx proteins, yet it would be strong enough to instigate Mx oligomerization and ring assembly. Such a model is compatible with the broad "substrate" specificity of Mx proteins: depending on the size of the invading viral ribonucleoprotein complexes that need to be wrapped, the assembly process would consume the necessary amount of Mx precursor molecules. These Mx ring structures might then act as energy-consuming wrenches to disassemble the viral target structure.
Collapse
|
14
|
microRNA control of interferons and interferon induced anti-viral activity. Mol Immunol 2013; 56:781-93. [PMID: 23962477 DOI: 10.1016/j.molimm.2013.07.009] [Citation(s) in RCA: 48] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2013] [Revised: 07/11/2013] [Accepted: 07/14/2013] [Indexed: 12/22/2022]
Abstract
Interferons (IFNs) are cytokines that are spontaneously produced in response to virus infection. They act by binding to IFN-receptors (IFN-R), which trigger JAK/STAT cell signalling and the subsequent induction of hundreds of IFN-inducible genes, including both protein-coding and microRNA genes. IFN-induced genes then act synergistically to prevent virus replication and create an anti-viral state. miRNA are therefore integral to the innate response to virus infection and are important components of IFN-mediated biology. On the other hand viruses also encode miRNAs that in some cases interfere directly with the IFN response to infection. This review summarizes the important roles of miRNAs in virus infection acting both as IFN-stimulated anti-viral molecules and as critical regulators of IFNs and IFN-stimulated genes. It also highlights how recent knowledge in RNA editing influence miRNA control of virus infection.
Collapse
|
15
|
Abstract
Peptide based drug design efforts have gained renewed interest with the discovery of cargo-carrying or cell-penetrating peptides. Understanding the translocation mechanism of these peptides and identifying the residues or elements that contribute to uptake can provide valuable clues toward the design of novel peptides. To this end, we have performed steered molecular dynamics (SMD) simulations on the pVEC peptide from murine vascular endothelial-cadherin protein and its two variants. Translocation was found to occur in three stages, adsorption via the cationic residues, inclusion of the whole peptide inside the membrane accompanied by formation of a water defect, and exit of both peptide and water molecules from the bilayer. Our simulation results suggest that the precise order in which the hydrophobic, cationic, and the polar regions are located in the amphipathic pVEC peptide contributes to its uptake mechanism. These results present new opportunities for the design of novel cell-penetrating and antimicrobial peptides.
Collapse
|
16
|
Dermine M, Desmecht D. In Vivo modulation of the innate response to pneumovirus by type-I and -III interferon-induced Bos taurus Mx1. J Interferon Cytokine Res 2012; 32:332-7. [PMID: 22385204 DOI: 10.1089/jir.2011.0123] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022] Open
Abstract
The respiratory syncytial virus (RSV) is a major pathogen of the human species. This pneumovirus is a prominent cause of airway morbidity in children and maintains an excessive hospitalization rate despite decades of research. As involvement of a genetic vulnerability is a possibility supported by recent data, we addressed the question of whether the Mx gene products, the typical target of which consists in single-stranded negative-polarity RNA viruses, could alter the course of pneumovirus-associated disease in vivo. Wild-type and Bos taurus Mx1-expressing transgenic FVB/J mice were inoculated with the mouse counterpart and closest phylogenetic relative of RSV, pneumonia virus of mice. Survival data and follow-up of body weight, histological scores, lung virus spread, and lung viral load unequivocally showed that the viral infection was severely repressed in Mx-transgenic mice, thus suggesting that pneumoviruses belong to the antiviral spectrum of mammalian Mx GTPases. Elucidating the underlying mechanisms at the molecular level could reveal critical information for the development of new anti-RSV molecules.
Collapse
Affiliation(s)
- Martin Dermine
- Department of Pathology, Faculty of Veterinary Medicine, University of Liège, Liège, Belgium
| | | |
Collapse
|
17
|
Gao S, von der Malsburg A, Dick A, Faelber K, Schröder G, Haller O, Kochs G, Daumke O. Structure of Myxovirus Resistance Protein A Reveals Intra- and Intermolecular Domain Interactions Required for the Antiviral Function. Immunity 2011; 35:514-25. [DOI: 10.1016/j.immuni.2011.07.012] [Citation(s) in RCA: 166] [Impact Index Per Article: 11.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2011] [Revised: 07/20/2011] [Accepted: 07/21/2011] [Indexed: 01/26/2023]
|
18
|
A peptide derived from herpes simplex virus type 1 glycoprotein H: membrane translocation and applications to the delivery of quantum dots. NANOMEDICINE-NANOTECHNOLOGY BIOLOGY AND MEDICINE 2011; 7:925-34. [PMID: 21664490 DOI: 10.1016/j.nano.2011.04.009] [Citation(s) in RCA: 62] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/26/2010] [Revised: 03/31/2011] [Accepted: 04/15/2011] [Indexed: 12/28/2022]
Abstract
UNLABELLED Cell membranes are impermeable to most molecules that are not actively imported by living cells, including all macromolecules and even small molecules whose physiochemical properties prevent passive membrane diffusion. However, recently, we have seen the development of increasingly sophisticated methodology for intracellular drug delivery. Cell-penetrating peptides (CPPs), short peptides believed to enter cells by penetrating cell membranes, have attracted great interest in the hope of enhancing gene therapy, vaccine development and drug delivery. Nevertheless, to achieve an efficient intracellular delivery, further strategies to bypass the endocytotic pathway must be investigated. We report on a novel peptide molecule derived from glycoprotein gH of herpes simplex type I virus that is able to traverse the membrane bilayer and to transport a cargo into the cytoplasm with novel properties in comparison with existing CPPs. We use as cargo molecule quantum dots that do not significantly traverse the membrane bilayer on their own. FROM THE CLINICAL EDITOR Cell-penetrating peptides have recently attracted great interest in optimizing gene therapy, vaccine development and drug delivery. In this study, a peptide derived from glycoprotein gH of herpes simplex I is investigated from this standpoint.
Collapse
|
19
|
Varkouhi AK, Scholte M, Storm G, Haisma HJ. Endosomal escape pathways for delivery of biologicals. J Control Release 2011; 151:220-8. [DOI: 10.1016/j.jconrel.2010.11.004] [Citation(s) in RCA: 1102] [Impact Index Per Article: 78.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2010] [Accepted: 10/19/2010] [Indexed: 11/29/2022]
|
20
|
Haller O, Gao S, von der Malsburg A, Daumke O, Kochs G. Dynamin-like MxA GTPase: structural insights into oligomerization and implications for antiviral activity. J Biol Chem 2010; 285:28419-24. [PMID: 20538602 DOI: 10.1074/jbc.r110.145839] [Citation(s) in RCA: 84] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
The interferon-inducible MxA GTPase is a key mediator of cell-autonomous innate immunity against a broad range of viruses such as influenza and bunyaviruses. MxA shares a similar domain structure with the dynamin superfamily of mechanochemical enzymes, including an N-terminal GTPase domain, a central middle domain, and a C-terminal GTPase effector domain. Recently, crystal structures of a GTPase domain dimer of dynamin 1 and of the oligomerized stalk of MxA (built by the middle and GTPase effector domains) were determined. These data provide exciting insights into the architecture and antiviral function of the MxA oligomer. Moreover, the structural knowledge paves the way for the development of novel antiviral drugs against influenza and other highly pathogenic viruses.
Collapse
Affiliation(s)
- Otto Haller
- Department of Virology, Institute for Medical Microbiology and Hygiene, University of Freiburg, Hermann-Herder-Strasse 11, D-79104 Freiburg, Germany.
| | | | | | | | | |
Collapse
|
21
|
Structural basis of oligomerization in the stalk region of dynamin-like MxA. Nature 2010; 465:502-6. [PMID: 20428112 DOI: 10.1038/nature08972] [Citation(s) in RCA: 196] [Impact Index Per Article: 13.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2009] [Accepted: 02/25/2010] [Indexed: 12/11/2022]
Abstract
The interferon-inducible dynamin-like myxovirus resistance protein 1 (MxA; also called MX1) GTPase is a key mediator of cell-autonomous innate immunity against pathogens such as influenza viruses. MxA partially localizes to COPI-positive membranes of the smooth endoplasmic reticulum-Golgi intermediate compartment. At the point of infection, it redistributes to sites of viral replication and promotes missorting of essential viral constituents. It has been proposed that the middle domain and the GTPase effector domain of dynamin-like GTPases constitute a stalk that mediates oligomerization and transmits conformational changes from the G domain to the target structure; however, the molecular architecture of this stalk has remained elusive. Here we report the crystal structure of the stalk of human MxA, which folds into a four-helical bundle. This structure tightly oligomerizes in the crystal in a criss-cross pattern involving three distinct interfaces and one loop. Mutations in each of these interaction sites interfere with native assembly, oligomerization, membrane binding and antiviral activity of MxA. On the basis of these results, we propose a structural model for dynamin oligomerization and stimulated GTP hydrolysis that is consistent with previous structural predictions and has functional implications for all members of the dynamin family.
Collapse
|
22
|
Wu K, Liu P, Meng XX, Liu L, Li YH, Ge ZZ, Yang JC. Preparation of polyclonal antibody against human MxA protein and its specificity to diversified myxovirus resistant protein A. BIOMEDICAL AND ENVIRONMENTAL SCIENCES : BES 2010; 23:76-82. [PMID: 20486440 DOI: 10.1016/s0895-3988(10)60035-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/29/2023]
Abstract
OBJECTIVE To study the human myxovirus resistant protein A (MxA), a specifically induced peptide by interferon I, and to use its level as a diagnostic criterion for viral infections. METHODS Anti-MxA antisera from immunized mice were prepared with the expressed MxA protein of pET32a-MxA in E. coli BL-21(DE3). To confirm the antiserum activity and specificity, the expression product of BL21, wild type MxA pEGFP-C1-wMxA and site-directed mutant MxA pEGFP-C1-mMxA(N589S) stably transfected 3T3 cells and induced A549 cells were detected by Western blot with the antisera using non-MxA transfected or non-IFN-beta induced cells, intact A549, NIH 3T3 cells transfected with pEGFP-C1 and pET32a (+)-transformed BL-21 as controls. RESULTS The antisera had specific positive immunoreactivity to the NIH3T3 cells transformed with pEGFP-C1-wMxA and pEGFP-C1-mMxA, INF-beta induced A549 cells and BL21 proteins expressed with pET32a (+)-MxA. The hybridization signals from IFN-beta induced A549 cells depended on the IFN-beta inducing concentrations. Meanwhile, immunohistochemical assay showed that NIH 3T3 cells with pEGFP-C1-wMxA and pEGFP-C1-mMxA had > 98% of positive cells at 1:50 dilution of the serum and A549 cells induced by 20 ng/mL IFN-beta for 48 h showed 95% positive cells. pEGFP-C1-transfected NIH 3T3 cells were all negative. CONCLUSION Anti-sera are highly specific to diversified MxAs. The antibody is detectable by Western blot, immunocytochemistry and immunofluorescence assay.
Collapse
Affiliation(s)
- Kang Wu
- School of Basic Medicine and Biology, Suzhou University, Suzhou 215123, Jiangsu, China
| | | | | | | | | | | | | |
Collapse
|
23
|
Palm M, Garigliany MM, Cornet F, Desmecht D. Interferon-induced Sus scrofa Mx1 blocks endocytic traffic of incoming influenza A virus particles. Vet Res 2010; 41:29. [PMID: 20167191 PMCID: PMC2826089 DOI: 10.1051/vetres/2010001] [Citation(s) in RCA: 29] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2009] [Accepted: 01/07/2010] [Indexed: 01/13/2023] Open
Abstract
The interferon-induced Mx proteins of vertebrates are dynamin-like GTPases, some isoforms of which can additionally inhibit the life cycle of certain RNA viruses. Here we show that the porcine Mx1 protein (poMx1) inhibits replication of influenza A virus and we attempt to identify the step at which the viral life cycle is blocked. In infected cells expressing poMx1, the level of transcripts encoding the viral nucleoprotein is significantly lower than normal, even when secondary transcription is prevented by exposure to cycloheximide. This reveals that a pretranscriptional block participates to the anti-influenza activity. Binding and internalization of incoming virus particles are normal in the presence of poMx1 but centripetal traffic to the late endosomes is interrupted. Surprisingly but decisively, poMx1 significantly alters binding of early endosome autoantigen 1 to early endosomes and/or early endosome size and spatial distribution. This is compatible with impairment of traffic of the endocytic vesicles to the late endosomes.
Collapse
Affiliation(s)
- Mélanie Palm
- Department of Pathology, University of Liège, FMV Sart Tilman B43, 4000 Liège, Belgium
| | | | | | | |
Collapse
|
24
|
Goparaju GN, Satishchandran C, Gupta PK. The effect of the structure of small cationic peptides on the characteristics of peptide-DNA complexes. Int J Pharm 2009; 369:162-9. [DOI: 10.1016/j.ijpharm.2008.10.028] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2008] [Accepted: 10/21/2008] [Indexed: 11/24/2022]
|
25
|
Vidal SM, Malo D, Marquis JF, Gros P. Forward genetic dissection of immunity to infection in the mouse. Annu Rev Immunol 2008; 26:81-132. [PMID: 17953509 DOI: 10.1146/annurev.immunol.26.021607.090304] [Citation(s) in RCA: 50] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
Forward genetics is an experimental approach in which gene mapping and positional cloning are used to elucidate the molecular mechanisms underlying phenotypic differences between two individuals for a given trait. This strategy has been highly successful for the study of inbred mouse strains that show differences in innate susceptibility to bacterial, parasitic, fungal, and viral infections. Over the past 20 years, these studies have led to the identification of a number of cell populations and critical biochemical pathways and proteins that are essential for the early detection of and response to invading pathogens. Strikingly, the macrophage is the point of convergence for many of these genetic studies. This has led to the identification of diverse pathways involved in extracellular and intracellular pathogen recognition, modification of the properties and content of phagosomes, transcriptional response, and signal transduction for activation of adaptive immune mechanisms. In models of viral infections, elegant genetic studies highlighted the pivotal role of natural killer cells in the detection and destruction of infected cells.
Collapse
Affiliation(s)
- S M Vidal
- Department of Microbiology and Immunology, McGill University, Montreal, Quebec, Canada H3G 1Y6
| | | | | | | |
Collapse
|
26
|
Torchilin VP. Tat peptide-mediated intracellular delivery of pharmaceutical nanocarriers. Adv Drug Deliv Rev 2008; 60:548-58. [PMID: 18053612 DOI: 10.1016/j.addr.2007.10.008] [Citation(s) in RCA: 333] [Impact Index Per Article: 19.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2007] [Accepted: 10/01/2007] [Indexed: 12/16/2022]
Abstract
Cell-penetrating peptides (CPPs) including TAT peptide (TATp) have been successfully used for intracellular delivery of a broad variety of cargoes including various nanoparticulate pharmaceutical carriers (liposomes, micelles, nanoparticles). Here, we will consider the main results in this area, with a special emphasis on TATp-mediated delivery of liposomes and DNA. We will also address the development of "smart" stimuli-sensitive nanocarriers, where cell-penetrating function can be activated by the decreased pH only inside the biological target minimizing thus the interaction of drug-loaded nanocarriers with non-target cells.
Collapse
|
27
|
Costantini DL, Hu M, Reilly RM. Update:Peptide Motifs for Insertion of Radiolabeled Biomolecules into Cells and Routing to the Nucleus for Cancer Imaging or Radiotherapeutic Applications. Cancer Biother Radiopharm 2008; 23:3-24. [DOI: 10.1089/cbr.2007.0430] [Citation(s) in RCA: 50] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/16/2023] Open
Affiliation(s)
- Danny L. Costantini
- Leslie Dan Faculty of Pharmacy, Department of Pharmaceutical Sciences, University of Toronto, Toronto, Ontario, Canada
| | - Meiduo Hu
- Leslie Dan Faculty of Pharmacy, Department of Pharmaceutical Sciences, University of Toronto, Toronto, Ontario, Canada
| | - Raymond M. Reilly
- Leslie Dan Faculty of Pharmacy, Department of Pharmaceutical Sciences, University of Toronto, Toronto, Ontario, Canada
| |
Collapse
|
28
|
Torchilin V. Intracellular delivery of protein and peptide therapeutics. DRUG DISCOVERY TODAY. TECHNOLOGIES 2008; 5:e95-e103. [PMID: 24981097 DOI: 10.1016/j.ddtec.2009.01.002] [Citation(s) in RCA: 120] [Impact Index Per Article: 7.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/03/2023]
Abstract
Many proteins and peptides are used as highly specific and effective therapeutic agents. Their use is, however, complicated by their instability and side effects. Because many protein and peptide drugs have their therapeutic targets inside cells, there is also an important task to bring these drugs into target cells without subjecting them to the lysosomal degradation. This review describes current approaches to the intracellular delivery of protein and peptide drugs. Various drug delivery systems and methods are considered allowing for safe and effective transport of protein and peptide drugs into the cell cytoplasm.:
Collapse
Affiliation(s)
- Vladimir Torchilin
- Department of Pharmaceutical Sciences and Center for Pharmaceutical Biotechnology and Nanomedicine, Northeastern University, 360 Huntington Avenue, Boston, MA 02115, USA.
| |
Collapse
|
29
|
Toyokawa K, Leite F, Ott TL. Cellular Localization and Function of the Antiviral Protein, Ovine Mx1 (oMx1): II. The oMx1 Protein Is a Regulator of Secretion in an Ovine Glandular Epithelial Cell Line. Am J Reprod Immunol 2007; 57:23-33. [PMID: 17156188 DOI: 10.1111/j.1600-0897.2006.00439.x] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022] Open
Abstract
PROBLEM Embryonic loss is a major contributor to infertility. Understanding factors affecting embryonic loss will help increase fertility. METHOD OF STUDY We investigated if ovine Mx1 (oMx1) mediated secretion by ovine glandular epithelial (oGE) cells using small interfering RNA (siRNA). Effects on secretion were examined through the conventional endoplasmic reticulum-Golgi pathway using beta2- microglobulin (beta2MG) as a marker, and interferon-stimulated gene 15 (ISG15) as a marker for unconventional secretion. RESULTS Mx1 siRNA reduced oMx1 mRNA levels at 12 and 24 hr after IFN-tau treatment (P < 0.05), without affecting levels of oMx2, ISG15, 2',5'-oligoadenylate synthetas or beta2MG. Mx1 siRNA reduced Mx1 protein levels at 48 and 120 hr after treatment (P < 0.05) and protein levels remained low at 120 hr. Transient oMx1 knock-down reduced secretion of oMx1 (P < 0.01). ISG15 protein in secretions was reduced without affecting intracellular levels (P < 0.05). Levels of beta2MG in secretions were not affected by Mx1 siRNA. CONCLUSION We showed that oMx1 protein is secreted by oGE cells and that reduction in oMx1 protein levels by siRNA reduced secretion of ISG15, but not beta2MG. Results support the hypothesis that oMx1 is a regulator of secretion through unconventional secretory pathway(s).
Collapse
Affiliation(s)
- Koji Toyokawa
- Department of Animal and Veterinary Science, Center for Reproductive Biology, University of Idaho, Moscow, ID, USA
| | | | | |
Collapse
|
30
|
Abstract
Intracellular delivery of various drugs, including DNA, and drug carriers can sharply increase the efficiency of various treatment protocols. However, the receptor-mediated endocytosis of drugs, drug carriers, and DNA results in their lysosomal delivery and significant degradation. The problem can be solved and therapy efficacy still further increased if the approaches for direct intracytoplasmic delivery that bypass the endocytic pathway are developed. This is especially important for many anticancer drugs (proapoptotic drugs whose primary action site is the mitochondrial membrane) and gene therapy (nuclear or mitochondrial genomes should be targeted). This review considers several current approaches for intracellular drug delivery: the use of pH-sensitive liposomes, the use of cell-penetrating proteins and peptides, and the use of immunoliposomes targeting intracellular antigens. Among intracellular targets, nuclei (gene therapy), mitochondria (proapoptotic cancer therapy and targeting of the mitochondrial genome), and lysosomes (lysosomal targeting of enzymes for the therapy of the lysosomal storage diseases) are considered. Examples of successful intracellular and organelle-specific delivery of biologically active molecules, including DNA, are presented; unanswered questions, challenges, and future trends are also discussed.
Collapse
Affiliation(s)
- Vladimir P Torchilin
- Department of Pharmaceutical Sciences, Northeastern University, Boston, Massachusetts 02115, USA.
| |
Collapse
|
31
|
Gupta B, Torchilin VP. Transactivating transcriptional activator-mediated drug delivery. Expert Opin Drug Deliv 2006; 3:177-90. [PMID: 16506946 DOI: 10.1517/17425247.3.2.177] [Citation(s) in RCA: 33] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Abstract
Cell-penetrating peptides (CPPs) are peptide vectors that can traverse through the plasma membrane barrier without breaching the integrity of the cell, and deliver various cargoes inside cell. The range of cargoes that can be delivered intracellularly by CPPs encompasses a broad variety of hydrophilic molecules, such as peptides, proteins, antibodies, imaging agents, DNA and even nanosized entities, including polymer-based systems, solid nanoparticles and liposomes. Multiple studies have focused on CPPs such as transactivating transcriptional activator peptide (TATp), penetratin, VP22, transportan and synthetic oligoarginines because of their high inherent potential as intracellular delivery vectors. However, the TATp remains the most popular CPP used for a variety of purposes. This review article attempts to bring together the available data on TAT-mediated intracellular uptake of a broad range of molecules and nanoparticles. It also considers potential practical applications of this approach.
Collapse
Affiliation(s)
- Bhawna Gupta
- Department of Pharmaceutical Sciences, Northeastern University, Boston, MA 02115, USA
| | | |
Collapse
|
32
|
Hu M, Chen P, Wang J, Chan C, Scollard DA, Reilly RM. Site-specific conjugation of HIV-1 tat peptides to IgG: a potential route to construct radioimmunoconjugates for targeting intracellular and nuclear epitopes in cancer. Eur J Nucl Med Mol Imaging 2005; 33:301-10. [PMID: 16258763 DOI: 10.1007/s00259-005-1908-7] [Citation(s) in RCA: 34] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2004] [Accepted: 07/04/2005] [Indexed: 11/29/2022]
Abstract
PURPOSE Our objective was to study the cellular and nuclear uptake of (123)I-mouse IgG ((123)I-mIgG) linked to peptides [GRKKRRQRRRPPQGYGC] harbouring the membrane-translocating and nuclear import sequences of HIV-1 tat protein. METHODS Carbohydrates on mIgG were oxidized by NaIO(4), then reacted with a 40-fold excess of peptides. Displacement of binding of anti-mouse IgG (Fab specific; alpha-mFab) to (123)I-mIgG by tat-mIgG or mIgG was compared. Internalization and nuclear translocation of (123)I-tat-mIgG in MDA-MB-468, MDA-MB-231 or MCF-7 breast cancer cells were measured. The immunoreactivity of imported tat-mIgG was evaluated by measuring binding of (123)I-alpha-mFab to cell lysate and by displacement of binding of (123)I-mIgG to alpha-mFab by cell lysate. Biodistribution and nuclear uptake of (123)I-tat-mIgG, (123)I-mIgG and (123)I-tat were compared in mice bearing s.c. MDA-MB-468 tumours. RESULTS There was a 15-fold decrease in affinity of alpha-mFab for tat-mIgG compared with mIgG. Internalized radioactivity imported into the nucleus for (123)I-tat-mIgG in MDA-MB-468, MDA-MB-231 and MCF-7 cells was 61.5+/-0.6%, 60.3+/-3.6% and 64.7+/-1.0%, respectively. The binding of (123)I-alpha-mFab to lysate from MDA-MB-468 cells importing tat-mIgG was 17-fold higher than that for cells not exposed to tat-mIgG. Imported tat-mIgG competed with tat-mIgG for displacement of binding of (123)I-mIgG to alpha-mFab. Conjugation of mIgG to tat peptides did not change tissue distribution. Nuclear localization for (123)I-tat-mIgG in MDA-MB-468 tumours was 28.1+/-5.6%, and for liver, spleen and kidneys it was 41.7+/-2.7%, 13.8+/-0.8% and 36.9+/-3.3%, respectively. CONCLUSION (123)I-tat-mIgG radioimunoconjugates suggest a route to the design of radiopharmaceuticals exploiting intracellular and nuclear epitopes.
Collapse
Affiliation(s)
- Meiduo Hu
- Division of Nuclear Medicine, University Health Network, Toronto, Ontario, Canada
| | | | | | | | | | | |
Collapse
|
33
|
Leroy M, Baise E, Pire G, Gérardin J, Desmecht D. Resistance of Paramyxoviridae to Type I Interferon-InducedBos taurusMx1 Dynamin. J Interferon Cytokine Res 2005; 25:192-201. [PMID: 15812245 DOI: 10.1089/jir.2005.25.192] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022] Open
Abstract
Typical targets of type I interferon (IFN)-induced antiviral Mx proteins known to date have been shown to share a common profile: single-stranded negative-sense RNA viruses. Among them, human MxA is known to interfere with the replication of measles, human, and bovine parainfluenza-3 viruses (BoPi3V), that is, three members of the Paramyxoviridae family. Recently, bovine Mx1 protein (BoMx1) was included in the group of Mx proteins with authenticated antiviral potential, as it dramatically represses the replication of vesicular stomatitis virus (VSV). As replication in bovine cells of Pi3, respiratory syncytial (RS), and Sendai (Se) viruses, all members of the same family, is known to be reduced on IFN-alpha incorporation into the culture medium, it was hypothesized that the BoMx1 pathway possibly was involved, its antiviral spectrum thus probably extending to Paramyxoviridae. In this study, probing of BoMx1-inhibiting effects was carried out by infecting a transgenic Vero cell line that allows tightly regulated conditional expression of BoMx1 after doxycycline treatment with a wide array of Paramyxoviridae. Expressing and nonexpressing cells displayed similar viability, cytopathic effects (CPEs), and amounts of infectious virus yields, whatever the infecting virus or the multiplicity of infection (moi) imposed. It is, therefore, concluded that BoMx1 does not interfere with Paramyxoviridae.
Collapse
Affiliation(s)
- Michael Leroy
- Department of Pathology, Faculty of Veterinary Medicine, University of Liège, B-4000 Liège, Belgium
| | | | | | | | | |
Collapse
|
34
|
Lin OE, Ohira T, Hirono I, Saito-Taki T, Aoki T. Immunoanalysis of antiviral Mx protein expression in Japanese flounder (Paralichthys olivaceus) cells. DEVELOPMENTAL AND COMPARATIVE IMMUNOLOGY 2004; 29:443-455. [PMID: 15707665 DOI: 10.1016/j.dci.2004.10.003] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/05/2004] [Revised: 09/30/2004] [Accepted: 10/03/2004] [Indexed: 05/24/2023]
Abstract
Mx proteins are interferon-inducible GTPases that possess antiviral properties in vertebrates. Japanese flounder (Paralichthys olivaceus) Mx protein has previously been shown to possess some antiviral activity against rhabdoviruses. A polyclonal antibody was generated against a purified peptide fragment of Japanese flounder Mx protein that had been produced in an Escherichia coli expression system. The PAb detected the approximately 71 kDa Mx protein from Japanese flounder (hirame) natural embryo (HINAE) cells that had been cultured with poly I:C, an interferon inducer, but not in unstimulated cells. The polyclonal antibody did not cross react with Mx protein from carp epithelial, grouper fin and zebrafish embryo cell lines that had been similarly induced or transfected with poly I:C. By immunofluorescence cytochemistry, Japanese flounder Mx protein was localized to the cell cytoplasm. Hirame rhabdovirus stimulated expression of Mx protein in the infected and surrounding HINAE cells. Within virus-infected cells, there was some indication of Mx protein colocalizing with viral proteins. Poly I:C stimulation of HINAE cells induced an early increase in Mx protein mRNA transcripts, but maximum Mx mRNA transcript and protein expression was reached after 48 h. Both Mx mRNA transcripts and protein levels were maintained till at least 72 h.
Collapse
Affiliation(s)
- Ooi Ei Lin
- Laboratory of Genome Science, Graduate School of Marine Science and Technology, Tokyo University of Marine Science and Technology, Konan 4-5-7, Minato-ku, Tokyo 108-8477, Japan
| | | | | | | | | |
Collapse
|
35
|
Mie M, Takahashi F, Funabashi H, Yanagida Y, Aizawa M, Kobatake E. Intracellular delivery of antibodies using TAT fusion protein A. Biochem Biophys Res Commun 2003; 310:730-4. [PMID: 14550263 DOI: 10.1016/j.bbrc.2003.09.071] [Citation(s) in RCA: 47] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
Abstract
Internalization of antibodies into mammalian cells is a useful method for analyzing and regulating cellular function. In this study, we developed a novel method for the delivery of antibodies into cells using the TAT-fused protein. This fusion protein consists of two functional domains, the protein transduction domain of HIV-1 TAT and the B domain of staphylococcal protein A (SpA), which has an ability to bind to the IgG. The TAT-SpA fusion protein was mixed with fluorescence-labeled rabbit IgG and added to cells. The internalization of antibody was analyzed using confocal microscopy and flow cytometry in living cells. As a result, fluorescence-labeled IgG with the TAT-SpA fusion protein was observed intracellularly. Flow cytometry results demonstrated time course and dose dependence relationships of antibody internalization. These results suggest that the TAT-SpA fusion protein can be a useful reagent for the delivery of antibody into cells.
Collapse
Affiliation(s)
- Masayasu Mie
- Department of Biological Information, Graduate School of Bioscience and Biotechnology, Tokyo Institute of Technology, Yokohama, Japan
| | | | | | | | | | | |
Collapse
|
36
|
Jin HK, Takada A, Kon Y, Haller O, Watanabe T. Identification of the murine Mx2 gene: interferon-induced expression of the Mx2 protein from the feral mouse gene confers resistance to vesicular stomatitis virus. J Virol 1999; 73:4925-30. [PMID: 10233954 PMCID: PMC112536 DOI: 10.1128/jvi.73.6.4925-4930.1999] [Citation(s) in RCA: 67] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
The mouse genome contains two related interferon-regulated genes, Mx1 and Mx2. Whereas Mx1 codes for the nuclear 72-kDa protein that interferes with influenza virus replication after interferon treatment, the Mx2 gene is nonfunctional in all laboratory mouse strains examined, since its open reading frame (ORF) is interrupted by an insertional mutation and a subsequent frameshift mutation. In the present study, we demonstrate that Mx2 mRNA of cells from feral mouse strains NJL (Mus musculus musculus) and SPR (Mus spretus) differs from that of the laboratory mouse strains tested. The Mx2 mRNA of the feral strains contains a single long ORF consisting of 656 amino acids. We further show that Mx2 protein in the feral strains is expressed upon interferon treatment and localizes to the cytoplasm much like the rat Mx2 protein, which inhibits vesicular stomatitis virus replication. Furthermore, transfected 3T3 cell lines of laboratory mouse origin expressing Mx2 from feral strains acquire slight resistance to vesicular stomatitis virus.
Collapse
Affiliation(s)
- H K Jin
- Laboratory of Experimental Animal Science, Graduate School of Veterinary Medicine, Hokkaido University, Sapporo 060-0818, Japan
| | | | | | | | | |
Collapse
|
37
|
Kochs G, Haller O. GTP-bound human MxA protein interacts with the nucleocapsids of Thogoto virus (Orthomyxoviridae). J Biol Chem 1999; 274:4370-6. [PMID: 9933640 DOI: 10.1074/jbc.274.7.4370] [Citation(s) in RCA: 86] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/04/2023] Open
Abstract
Human MxA protein is an interferon-induced member of the dynamin superfamily of large GTPases. MxA inhibits the multiplication of several RNA viruses, including Thogoto virus, an influenza virus-like orthomyxovirus transmitted by ticks. Previous studies have indicated that GTP binding is required for antiviral activity, but the mechanism of action is still unknown. Here, we have used an in vitro cosedimentation assay to demonstrate, for the first time, a GTP-dependent interaction between MxA GTPase and a viral target structure. The assay is based on highly active MxA GTPase as effector molecules, Thogoto virus nucleocapsids as viral targets, and guanosine 5'-O-(3-thiotriphosphate) (GTPgammaS) as a stabilizing factor. We show that MxA tightly interacts with viral nucleocapsids by binding to the nucleoprotein component. This interaction requires the presence of GTPgammaS and is mediated by domains in the carboxyl-terminal moiety of MxA. We propose that GTP-bound MxA adopts an antivirally active conformation that allows interaction with viral nucleocapsids, thereby impairing their normal function.
Collapse
Affiliation(s)
- G Kochs
- Abteilung Virologie, Institut für Medizinische Mikrobiologie und Hygiene, Universität Freiburg, D-79008 Freiburg, Germany.
| | | |
Collapse
|
38
|
Abstract
Mx proteins are members of a family of interferon-inducible genes expressed when cells are treated with double-stranded RNA or virus infection. These proteins are important components of the antiviral response and form the first line of the body's defense against virus infections. The exact mechanism of action for these proteins has not been discovered, but mice missing the Mx genes are extremely sensitive to influenza virus infection. Mammals have between two and three Mx genes whose functions may vary with regard to the inhibition of a specific virus, cellular localization, and activity. The cDNA of three rainbow trout Mx proteins has been cloned and a comparison of their sequences with that of avian and mammalian species reveals striking conservation of domains. They all maintain the tripartite ATP/GTP binding domain and the dynamin family signature in the amino terminal half of the protein. In the carboxyl terminal half of the Mx proteins are the localization signals and the leucine zipper motifs which account for the trimerization of Mx in the cell. Like the rat and human Mx proteins, the different trout Mx proteins exhibit distinctly different immunohistochemical staining patterns in cells transfected with plasmids expressing RBTMx1, RBTMx2, or RBTMx3. To date, the antiviral function of the trout Mx proteins has not been satisfactorily established.
Collapse
Affiliation(s)
- J C Leong
- Department of Microbiology, Oregon State University, Corvallis, USA.
| | | | | | | | | |
Collapse
|
39
|
Johannes L, Kambadur R, Lee-Hellmich H, Hodgkinson CA, Arnheiter H, Meier E. Antiviral determinants of rat Mx GTPases map to the carboxy-terminal half. J Virol 1997; 71:9792-5. [PMID: 9371647 PMCID: PMC230291 DOI: 10.1128/jvi.71.12.9792-9795.1997] [Citation(s) in RCA: 31] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/05/2023] Open
Abstract
Rat Mx2 and rat Mx3 are two alpha/beta interferon-inducible cytoplasmic GTPases that differ in three residues in the amino-terminal third, which also contains the tripartite GTP-binding domain, and that differ in five residues in the carboxy-terminal quarter, which also contains a dimerization domain. While Mx2 is active against vesicular stomatitis virus (VSV), Mx3 lacks antiviral activity. We mapped the functional difference between Mx2 and Mx3 protein to two critical residues in the carboxy-terminal parts of the molecules. An exchange of either residue 588 or 630 of Mx2 with the corresponding residues of Mx3 abolished anti-VSV activity, and the introduction of the two Mx2 residues on an Mx3 background partially restored anti-VSV activity. These results are consistent with the facts that Mx2 and Mx3 have similar intrinsic GTPase activities and that the GTPase domain of Mx3 can fully substitute for the GTPase domain of Mx2. Nevertheless, the amino-terminal third containing the GTP-binding domain is necessary for antiviral activity, since an amino-terminally truncated Mx2 protein is devoid of anti-VSV activity. Furthermore, Fab fragments of a monoclonal antibody known to neutralize antiviral activity block GTPase activity by binding an epitope in the carboxy-terminal half of Mx2 or Mx3 protein. The results are consistent with a two-domain model in which both the conserved amino-terminal half and the less-well-conserved carboxy-terminal half of Mx proteins carry functionally important domains.
Collapse
Affiliation(s)
- L Johannes
- Laboratory of Developmental Neurogenetics, National Institute of Neurological Disorders and Stroke, Bethesda, Maryland 20892, USA
| | | | | | | | | | | |
Collapse
|
40
|
Novoa I, Benavente J, Cotten M, Carrasco L. Permeabilization of mammalian cells to proteins: poliovirus 2A(pro) as a probe to analyze entry of proteins into cells. Exp Cell Res 1997; 232:186-90. [PMID: 9141636 DOI: 10.1006/excr.1997.3533] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/04/2023]
Abstract
Two hybrid protein molecules containing the poliovirus protease 2A (MBP-2A(pro)) (maltose-binding protein-2A(pro) and MBP-Pseudomonas exotoxin A-2A(pro)) have been constructed and purified. Both hybrid proteins efficiently cleave the translation initiation factor eIF-4G when they are co-internalized into cells with adenovirus particles. Almost no intact eIF-4G can be detected in cells incubated with these proteins following this method. Reovirus infectious subviral particles also promote the delivery of MBP-2A(pro) into cells, although less efficiently than adenovirus particles. None of the other methods employed to permeabilize cells to MBP-2A(pro) achieves the degree of eIF-4G cleavage observed with adenovirus particles. By comparison about 30% of cells electroporated with MBP-2A(pro) still contain intact eIF-4G. More drastic electroporation conditions lead to a significant decrease of cell survival. Osmotic lysis of pinocytic vesicles resulted in 30% of the eIF-4G being cleaved in cells treated in suspension. Delivery of MBP-2A(pro) by pH-sensitive liposomes leads to poor hydrolysis of eIF-4G. Taken together our results indicate that permeabilization of cells with adenovirus particles is the most efficient method for introducing MBP-2A(pro) into cells.
Collapse
Affiliation(s)
- I Novoa
- Centro de Biología Molecular (CSIC-UAM), Universidad Autónoma de Madrid, Canto Blanco, Spain
| | | | | | | |
Collapse
|
41
|
Kalvakolanu DV, Borden EC. An overview of the interferon system: signal transduction and mechanisms of action. Cancer Invest 1996; 14:25-53. [PMID: 8597888 DOI: 10.3109/07357909609018435] [Citation(s) in RCA: 108] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/31/2023]
Affiliation(s)
- D V Kalvakolanu
- Department of Microbology, University of Maryland School of Medicine, Baltimore, Maryland, USA
| | | |
Collapse
|
42
|
Arnheiter H, Frese M, Kambadur R, Meier E, Haller O. Mx transgenic mice--animal models of health. Curr Top Microbiol Immunol 1996; 206:119-47. [PMID: 8608714 DOI: 10.1007/978-3-642-85208-4_8] [Citation(s) in RCA: 30] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/31/2023]
Affiliation(s)
- H Arnheiter
- Laboratory of Developmental Neurogenetics, National Institute of Neurological Disorders and Stroke, National Institutes of Health, Bethesda, MD 20892-4160, USA
| | | | | | | | | |
Collapse
|
43
|
Frese M, Kochs G, Meier-Dieter U, Siebler J, Haller O. Human MxA protein inhibits tick-borne Thogoto virus but not Dhori virus. J Virol 1995; 69:3904-9. [PMID: 7745744 PMCID: PMC189115 DOI: 10.1128/jvi.69.6.3904-3909.1995] [Citation(s) in RCA: 115] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/26/2023] Open
Abstract
Thogoto and Dhori viruses are tick-borne orthomyxoviruses infecting humans and livestock in Africa, Asia, and Europe. Here, we show that human MxA protein is an efficient inhibitor of Thogoto virus but is inactive against Dhori virus. When expressed in the cytoplasm of stably transfected cell lines, MxA protein interfered with the accumulation of Thogoto viral RNA and proteins. Likewise, MxA(R645), a mutant MxA protein known to be active against influenza virus but inactive against vesicular stomatitis virus, was equally efficient in blocking Thogoto virus growth. Hence, a common antiviral mechanism that is distinct from the antiviral action against vesicular stomatitis virus may operate against both influenza virus and Thogoto virus. When moved to the nucleus with the help of a foreign nuclear transport signal, MxA(R645) remained active against Thogoto virus, indicating that a nuclear step of virus replication was inhibited. In contrast, Dhori virus was not affected by wild-type or mutant MxA protein, indicating substantial differences between these two tick-transmitted orthomyxoviruses. Human MxB protein had no antiviral activity against either virus.
Collapse
Affiliation(s)
- M Frese
- Abteilung Virologie, Institut für Medizinische Mikrobiologie und Hygiene, Universität Freiburg, Germany
| | | | | | | | | |
Collapse
|
44
|
Haller O, Frese M, Rost D, Nuttall PA, Kochs G. Tick-borne thogoto virus infection in mice is inhibited by the orthomyxovirus resistance gene product Mx1. J Virol 1995; 69:2596-601. [PMID: 7884909 PMCID: PMC188937 DOI: 10.1128/jvi.69.4.2596-2601.1995] [Citation(s) in RCA: 92] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/27/2023] Open
Abstract
We show that tick-transmitted Thogoto virus is sensitive to interferon-induced nuclear Mx1 protein, which is known for its specific antiviral action against orthomyxoviruses. Influenza virus-susceptible BALB/c mice (lacking a functional Mx1 gene) developed severe disease symptoms and died within days after intracerebral or intraperitoneal infection with a lethal challenge dose of Thogoto virus. In contrast, Mx1-positive congenic, influenza virus-resistant BALB.A2G-Mx1 mice remained healthy and survived. Likewise, A2G, congenic B6.A2G-Mx1 and CBA.T9-Mx1 mice (derived from influenza virus-resistant wild mice) as well as Mx1-transgenic 979 mice proved to be resistant. Peritoneal macrophages and interferon-treated embryo cells from resistant mice exhibited the same resistance phenotype in vitro. Moreover, stable lines of transfected mouse 3T3 cells that constitutively express Mx1 protein showed increased resistance to Thogoto virus infection. We conclude that an Mx1-sensitive step has been conserved during evolution of orthomyxoviruses and suggest that the Mx1 gene in rodents may serve to combat infections by influenza virus-like arboviruses.
Collapse
Affiliation(s)
- O Haller
- Abteilung Virologie, Universität Freiburg, Germany
| | | | | | | | | |
Collapse
|
45
|
Abstract
Human resistance to infectious diseases is often regulated by multiple genes that control different aspects of host-parasite interaction. Genetically distinct inbred strains of mice that differ in their susceptibility to specific pathogens are invaluable for dissecting such complex patterns and have allowed the identification of several host-resistance loci that regulate natural and acquired immunity in response to infection. Cloning these genes is the first step in elucidating their roles in host defense.
Collapse
Affiliation(s)
- D Malo
- McGill Centre, Montreal General Hospital, Canada
| | | |
Collapse
|
46
|
Heinz H, Marquardt J, Schuberth HJ, Adolf GR, Leibold W. Proteins induced by recombinant equine interferon-beta 1 within equine peripheral blood mononuclear cells and polymorphonuclear neutrophilic granulocytes. Vet Immunol Immunopathol 1994; 42:221-35. [PMID: 7810057 DOI: 10.1016/0165-2427(94)90069-8] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/27/2023]
Abstract
Peripheral blood mononuclear cells (PBMC) and polymorphonuclear neutrophilic granulocytes (PMN) as well as embryonic equine dermal fibroblasts and the equine fibroblast line E. Derm which were used as controls, were treated with recombinant equine interferon-beta 1 (rEqIFN-beta 1) in vitro which induced the expression of different proteins in these cells. A 74 kDa protein was induced in PBMC and an 82 kDa protein was additionally found in the equine fibroblast E. Derm cell line following treatment with rEqFN-beta 1. Both proteins reacted with anti-mouse and anti-human Mx protein antisera in immunoblot tests. The 74 kDa and perhaps the 82 kDa components may thus represent equine 'Mxanalogous proteins'. The 74 kDa protein was only detected in PBMC of ten out of 20 horses examined. The induction of Mx protein in the horse by Type 1 interferon may therefore resemble that in the mouse, where Mx protein is involved in selective resistance to influenza virus. The influence of rEqIFN-beta 1 on protein expression in equine PBMC and PMN was monitored by metabolic labeling and 2-D gel electrophoresis. Proteins of 82, 74, 58 and 40 kDa were induced in PBMC following exposure to rEqIFN-beta 1. A constitutively expressed 35 kDa protein, however, was no longer demonstrable upon treatment with interferon. None of the proteins induced within PBMC was found in highly purified PMN treated with interferon. PMN exposed to rEqIFN-beta 1 synthesized four proteins in the range of 25 to 27 kDa. These proteins have not been described in interferon-treated PMN of any other species.
Collapse
Affiliation(s)
- H Heinz
- Immunology Unit, Veterinary School, Hannover, Germany
| | | | | | | | | |
Collapse
|
47
|
IFP 35 is an interferon-induced leucine zipper protein that undergoes interferon-regulated cellular redistribution. J Biol Chem 1994. [DOI: 10.1016/s0021-9258(17)42225-3] [Citation(s) in RCA: 51] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022] Open
|
48
|
Nakayama M, Yazaki K, Kusano A, Nagata K, Hanai N, Ishihama A. Structure of mouse Mx1 protein. Molecular assembly and GTP-dependent conformational change. J Biol Chem 1993. [DOI: 10.1016/s0021-9258(18)82434-6] [Citation(s) in RCA: 58] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/27/2022] Open
|
49
|
Affiliation(s)
- G C Sen
- Department of Molecular Biology, Cleveland Clinic Foundation, Ohio 44195-5285
| | | |
Collapse
|
50
|
Romantsev FE, Khodarev NN, Votrin II. Intermolecular homologies of human interferon-alpha. Biochem Biophys Res Commun 1992; 186:211-8. [PMID: 1632767 DOI: 10.1016/s0006-291x(05)80795-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2022]
Abstract
Human interferon-alpha 2 (IFN) was analyzed by homology search computer program with the use of protein primary structures data bases. Results indicate that four domains with heightened ability to form homology pairs with different proteins exist in the IFN molecule. These domains occupy regions 35-56, 72-85, 97-110 and 124-136, mainly between the alpha-helical cylinders on the tertiary structure models. Additionally, results show in IFN structure the presence of amino-acid motifs that create the opportunity for this cytokine to influence directly the processes of DNA functioning in cell nuclei.
Collapse
|