1
|
Dewangan N, Jana ID, Yadav S, Sardar A, Mallick AI, Mondal A, Tarafdar PK. Design of Flavonoid-Based Lipid Domains as Fusion Inhibitors to Efficiently Block Coronavirus and Other Enveloped Virus Infection. SMALL (WEINHEIM AN DER BERGSTRASSE, GERMANY) 2025; 21:e2410727. [PMID: 39828665 DOI: 10.1002/smll.202410727] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/11/2024] [Revised: 12/27/2024] [Indexed: 01/22/2025]
Abstract
Developing a broad-spectrum antiviral is imperative in light of the recent emergence of recurring viral infections. The critical role of host-virus attachment and membrane fusion during enveloped virus entry is a suitable target for developing broad-spectrum antivirals. A new class of flavonoid-based fusion inhibitors are designed to alter the membrane's physical properties. These flavonoid-based molecules (MFDA; myristoyl flavonoid di-aspartic acid) are self-assembled in the membrane, creating distinct nanodomains and effectively inhibiting membrane fusion by modulating the membrane's interfacial properties. The broad-spectrum antiviral efficacy of these compounds are established in effectively blocking the entry of Severe Acute Respiratory Syndrome Coronavirus 2 (SARS-CoV-2), Type A Influenza, Human coronavirus OC43 (HCoV-OC43), and Vesicular stomatitis virus (VSV). A slightly more effectivity of MFDA in coronavirus infection than other enveloped viruses may be attributed to its secondary interaction with the receptor binding domain (RBD) of the SARS-CoV-2 spike protein. A membrane nanodomain formation strategy is highlighted with natural-product-based fusion inhibitors, effectively thwarting the infection of several enveloped viruses, entailing their broad-spectrum antiviral functionality.
Collapse
Affiliation(s)
- Nikesh Dewangan
- Department of Chemical Sciences, Indian Institute of Science Education and Research Kolkata, Mohanpur, 741246, India
| | - Indrani Das Jana
- Department of Bioscience & Biotechnology, Indian Institute of Technology Kharagpur, Kharagpur, 721302, India
| | - Sandeep Yadav
- Department of Biological Sciences, Indian Institute of Science Education and Research Kolkata, Mohanpur, 741246, India
| | - Avijit Sardar
- Department of Chemical Sciences, Indian Institute of Science Education and Research Kolkata, Mohanpur, 741246, India
| | - Amirul I Mallick
- Department of Biological Sciences, Indian Institute of Science Education and Research Kolkata, Mohanpur, 741246, India
| | - Arindam Mondal
- Department of Bioscience & Biotechnology, Indian Institute of Technology Kharagpur, Kharagpur, 721302, India
| | - Pradip K Tarafdar
- Department of Chemical Sciences, Indian Institute of Science Education and Research Kolkata, Mohanpur, 741246, India
| |
Collapse
|
2
|
Sardar A, Bhowmick S, Kamble M, Dewangan N, Hazra B, Mallick AI, Tarafdar PK. De Novo Design of Lipopeptide-Based β-Sheet-Like Self-Assemblies: A Strategy to Develop Fusion Inhibitors as Broad-Spectrum Antivirals. Chemistry 2025; 31:e202403039. [PMID: 39716967 DOI: 10.1002/chem.202403039] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2024] [Revised: 12/15/2024] [Accepted: 12/16/2024] [Indexed: 12/25/2024]
Abstract
The recent surge in emerging viral infections warrants the design of broad-spectrum antivirals. We aim to develop a lead molecule that targets a common biochemical feature of many enveloped viruses, membrane fusion. To achieve the broad-spectrum ability, instead of targeting the fusion machinery, we plan to modulate the physicochemical properties of the host and viral membranes to block fusion. The approach is based on the Coronin-1 protein of Mycobacterium, which presumably inhibits the phagosome-lysosome fusion, and a unique Trp-Asp (WD) sequence is placed at the distorted β-meander motif. We designed a WD-based branched lipopeptide (Myr-D(WD)2) that supported the intermolecular interactions to create a β-sheet-like supramolecular assembly at the membrane surface. TEM and confocal fluorescence experiments also suggest that the lipopeptide self-assembled at the bilayer interface and modulated the interfacial order and the water penetration. We demonstrated that the supramolecular organization of Myr-D(WD)2 could block artificial membrane fusion completely and restrict pH-dependent influenza virus (H1N1, H9N2), and pH-independent mouse hepatitis virus, human coronavirus (HCoV-OC43) infections. The present study provided an evidence-based broad-spectrum antiviral potential of a designed self-assembled lipopeptide.
Collapse
Affiliation(s)
- Avijit Sardar
- Department of Chemical Sciences, Indian Institute of Science Education and Research Kolkata, Mohanpur, 741 246, India
| | - Sucharita Bhowmick
- Department of Biological Sciences, Indian Institute of Science Education and Research Kolkata, Mohanpur, 741 246, India
| | - Mithila Kamble
- Department of Biological Sciences, Indian Institute of Science Education and Research Kolkata, Mohanpur, 741 246, India
| | - Nikesh Dewangan
- Department of Chemical Sciences, Indian Institute of Science Education and Research Kolkata, Mohanpur, 741 246, India
| | - Bibhas Hazra
- Department of Chemical Sciences, Indian Institute of Science Education and Research Kolkata, Mohanpur, 741 246, India
| | - Amirul I Mallick
- Department of Biological Sciences, Indian Institute of Science Education and Research Kolkata, Mohanpur, 741 246, India
| | - Pradip K Tarafdar
- Department of Chemical Sciences, Indian Institute of Science Education and Research Kolkata, Mohanpur, 741 246, India
| |
Collapse
|
3
|
Lü Z, Dai X, Xu J, Liu Z, Guo Y, Gao Z, Meng F. Medicinal chemistry strategies toward broad-spectrum antiviral agents to prevent next pandemics. Eur J Med Chem 2024; 271:116442. [PMID: 38685143 DOI: 10.1016/j.ejmech.2024.116442] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2023] [Revised: 04/02/2024] [Accepted: 04/19/2024] [Indexed: 05/02/2024]
Abstract
The pandemic and tremendous impact of severe acute respiratory syndrome coronavirus 2 alert us, despite great achievements in prevention and control of infectious diseases, we still lack universal and powerful antiviral strategies to rapidly respond to the potential threat of serious infectious disease. Various highly contagious and pathogenic viruses, as well as other unknown viruses may appear or reappear in human society at any time, causing a catastrophic epidemic. Developing broad-spectrum antiviral drugs with high security and efficiency is of great significance for timely meeting public health emergency and protecting the lives and health of the people. Hence, in this review, we summarized diverse broad-spectrum antiviral targets and corresponding agents from a medicinal chemistry prospective, compared the pharmacological advantages and disadvantages of different targets, listed representative agents, showed their structures, pharmacodynamics and pharmacokinetics characteristics, and conducted a critical discussion on their development potential, in the hope of providing up-to-date guidance for the development of broad-spectrum antivirals and perspectives for applications of antiviral therapy.
Collapse
Affiliation(s)
- Zirui Lü
- State Key Laboratory of NBC Protection for Civilian, Beijing, 102205, China
| | - Xiandong Dai
- State Key Laboratory of NBC Protection for Civilian, Beijing, 102205, China
| | - Jianjie Xu
- State Key Laboratory of NBC Protection for Civilian, Beijing, 102205, China
| | - Zhenming Liu
- State Key Laboratory of Natural and Biomimetic Drugs, Peking University, Beijing, China
| | - Yongbiao Guo
- State Key Laboratory of NBC Protection for Civilian, Beijing, 102205, China
| | - Zhenhua Gao
- State Key Laboratory of NBC Protection for Civilian, Beijing, 102205, China
| | - Fanhua Meng
- State Key Laboratory of NBC Protection for Civilian, Beijing, 102205, China.
| |
Collapse
|
4
|
Groß R, Reßin H, von Maltitz P, Albers D, Schneider L, Bley H, Hoffmann M, Cortese M, Gupta D, Deniz M, Choi JY, Jansen J, Preußer C, Seehafer K, Pöhlmann S, Voelker DR, Goffinet C, Pogge-von Strandmann E, Bunz U, Bartenschlager R, El Andaloussi S, Sparrer KMJ, Herker E, Becker S, Kirchhoff F, Münch J, Müller JA. Phosphatidylserine-exposing extracellular vesicles in body fluids are an innate defence against apoptotic mimicry viral pathogens. Nat Microbiol 2024; 9:905-921. [PMID: 38528146 PMCID: PMC10994849 DOI: 10.1038/s41564-024-01637-6] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2023] [Accepted: 02/14/2024] [Indexed: 03/27/2024]
Abstract
Some viruses are rarely transmitted orally or sexually despite their presence in saliva, breast milk, or semen. We previously identified that extracellular vesicles (EVs) in semen and saliva inhibit Zika virus infection. However, the antiviral spectrum and underlying mechanism remained unclear. Here we applied lipidomics and flow cytometry to show that these EVs expose phosphatidylserine (PS). By blocking PS receptors, targeted by Zika virus in the process of apoptotic mimicry, they interfere with viral attachment and entry. Consequently, physiological concentrations of EVs applied in vitro efficiently inhibited infection by apoptotic mimicry dengue, West Nile, Chikungunya, Ebola and vesicular stomatitis viruses, but not severe acute respiratory syndrome coronavirus 2, human immunodeficiency virus 1, hepatitis C virus and herpesviruses that use other entry receptors. Our results identify the role of PS-rich EVs in body fluids in innate defence against infection via viral apoptotic mimicries, explaining why these viruses are primarily transmitted via PS-EV-deficient blood or blood-ingesting arthropods rather than direct human-to-human contact.
Collapse
Affiliation(s)
- Rüdiger Groß
- Institute of Molecular Virology, Ulm University Medical Center, Ulm, Germany
| | - Hanna Reßin
- Institute of Molecular Virology, Ulm University Medical Center, Ulm, Germany
| | - Pascal von Maltitz
- Institute of Molecular Virology, Ulm University Medical Center, Ulm, Germany
| | - Dan Albers
- Institute of Molecular Virology, Ulm University Medical Center, Ulm, Germany
| | - Laura Schneider
- Institute of Virology, Philipps University Marburg, Marburg, Germany
| | - Hanna Bley
- Institute of Virology, Philipps University Marburg, Marburg, Germany
| | - Markus Hoffmann
- Infection Biology Unit, German Primate Center, Göttingen, Germany
- Georg-August University Göttingen, Göttingen, Germany
| | - Mirko Cortese
- Department of Infectious Diseases, Molecular Virology, University of Heidelberg, Heidelberg, Germany
| | - Dhanu Gupta
- Biomolecular Medicine, Clinical Research Center, Department of Laboratory Medicine, Karolinska Institutet, Stockholm, Sweden
- Department of Paediatrics, University of Oxford, Oxford, UK
| | - Miriam Deniz
- Clinic for Gynecology and Obstetrics, Ulm University Medical Center, Ulm, Germany
| | - Jae-Yeon Choi
- Department of Medicine, National Jewish Health, Denver, CO, USA
| | - Jenny Jansen
- Institute of Virology, Charité-Universitätsmedizin Berlin, Berlin, Germany
| | - Christian Preußer
- Core Facility Extracellular Vesicles, Institute for Tumor Immunology, Center for Tumor Biology and Immunology, Philipps University Marburg, Marburg, Germany
| | - Kai Seehafer
- Organisch-Chemisches Institut, Ruprecht-Karls-Universität, Heidelberg, Germany
| | - Stefan Pöhlmann
- Infection Biology Unit, German Primate Center, Göttingen, Germany
- Georg-August University Göttingen, Göttingen, Germany
| | | | - Christine Goffinet
- Institute of Virology, Charité-Universitätsmedizin Berlin, Berlin, Germany
- Department of Tropical Disease Biology, Liverpool School of Tropical Medicine, Liverpool, UK
| | - Elke Pogge-von Strandmann
- Core Facility Extracellular Vesicles, Institute for Tumor Immunology, Center for Tumor Biology and Immunology, Philipps University Marburg, Marburg, Germany
| | - Uwe Bunz
- Organisch-Chemisches Institut, Ruprecht-Karls-Universität, Heidelberg, Germany
| | - Ralf Bartenschlager
- Department of Infectious Diseases, Molecular Virology, University of Heidelberg, Heidelberg, Germany
| | - Samir El Andaloussi
- Biomolecular Medicine, Clinical Research Center, Department of Laboratory Medicine, Karolinska Institutet, Stockholm, Sweden
| | | | - Eva Herker
- Institute of Virology, Philipps University Marburg, Marburg, Germany
| | - Stephan Becker
- Institute of Virology, Philipps University Marburg, Marburg, Germany
| | - Frank Kirchhoff
- Institute of Molecular Virology, Ulm University Medical Center, Ulm, Germany
| | - Jan Münch
- Institute of Molecular Virology, Ulm University Medical Center, Ulm, Germany
| | - Janis A Müller
- Institute of Molecular Virology, Ulm University Medical Center, Ulm, Germany.
- Institute of Virology, Philipps University Marburg, Marburg, Germany.
| |
Collapse
|
5
|
Adeosun WB, Loots DT. Medicinal Plants against Viral Infections: A Review of Metabolomics Evidence for the Antiviral Properties and Potentials in Plant Sources. Viruses 2024; 16:218. [PMID: 38399995 PMCID: PMC10892737 DOI: 10.3390/v16020218] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2023] [Revised: 01/22/2024] [Accepted: 01/29/2024] [Indexed: 02/25/2024] Open
Abstract
Most plants have developed unique mechanisms to cope with harsh environmental conditions to compensate for their lack of mobility. A key part of their coping mechanisms is the synthesis of secondary metabolites. In addition to their role in plants' defense against pathogens, they also possess therapeutic properties against diseases, and their use by humans predates written history. Viruses are a unique class of submicroscopic agents, incapable of independent existence outside a living host. Pathogenic viruses continue to pose a significant threat to global health, leading to innumerable fatalities on a yearly basis. The use of medicinal plants as a natural source of antiviral agents has been widely reported in literature in the past decades. Metabolomics is a powerful research tool for the identification of plant metabolites with antiviral potentials. It can be used to isolate compounds with antiviral capacities in plants and study the biosynthetic pathways involved in viral disease progression. This review discusses the use of medicinal plants as antiviral agents, with a special focus on the metabolomics evidence supporting their efficacy. Suggestions are made for the optimization of various metabolomics methods of characterizing the bioactive compounds in plants and subsequently understanding the mechanisms of their operation.
Collapse
Affiliation(s)
- Wilson Bamise Adeosun
- Human Metabolomics, North-West University, Private Bag X6001, Box 269, Potchefstroom 2531, South Africa;
| | | |
Collapse
|
6
|
Negi G, Sharma A, Chaudhary M, Parveen N. Disruption Mechanisms of Enveloped Viruses by Ionic and Nonionic Surfactants. J Phys Chem B 2024; 128:768-780. [PMID: 38228291 DOI: 10.1021/acs.jpcb.3c05531] [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: 01/18/2024]
Abstract
The world has witnessed multiple pandemics and endemics caused by enveloped viruses in the past century. To name a few, the ongoing COVID-19 pandemic and other pandemics/endemics caused by coronaviruses, influenza viruses, HIV-1, etc. The external and topical applications of surfactants have been effective in limiting the spread of viruses. While it is well-known that surfactants inactivate virus particles (virions), the mechanism of action of surfactants against enveloped virions has not yet been established. In this work, we have evaluated the surfactant-induced disruption mechanism of a cocktail of enveloped viruses containing particles of mumps, measles, and rubella viruses. We applied the total internal reflection fluorescence microscopy technique to trace the temporal changes in the fluorescence signal from single virions upon the addition of a surfactant solution. We report that surfactants solubilize either the viral lipid membrane, proteins, or both. Ionic surfactants, depending on their charge and interaction type with the viral lipids and proteins, can cause bursting or perforation of the viral envelope, whereas a nonionic surfactant can cause either symmetric expansion or perforation of the viral envelope depending on the surfactant concentration.
Collapse
Affiliation(s)
- Geetanjali Negi
- Department of Chemistry, Indian Institute of Technology Kanpur, 208016 Kanpur, India
| | - Anurag Sharma
- Department of Chemistry, Indian Institute of Technology Kanpur, 208016 Kanpur, India
| | - Monika Chaudhary
- Department of Chemistry, Indian Institute of Technology Kanpur, 208016 Kanpur, India
| | - Nagma Parveen
- Department of Chemistry, Indian Institute of Technology Kanpur, 208016 Kanpur, India
| |
Collapse
|
7
|
Johnson TG, Langton MJ. Molecular Machines For The Control Of Transmembrane Transport. J Am Chem Soc 2023; 145:27167-27184. [PMID: 38062763 PMCID: PMC10740008 DOI: 10.1021/jacs.3c08877] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2023] [Revised: 11/21/2023] [Accepted: 11/22/2023] [Indexed: 12/21/2023]
Abstract
Nature embeds some of its molecular machinery, including ion pumps, within lipid bilayer membranes. This has inspired chemists to attempt to develop synthetic analogues to exploit membrane confinement and transmembrane potential gradients, much like their biological cousins. In this perspective, we outline the various strategies by which molecular machines─molecular systems in which a nanomechanical motion is exploited for function─have been designed to be incorporated within lipid membranes and utilized to mediate transmembrane ion transport. We survey molecular machines spanning both switches and motors, those that act as mobile carriers or that are anchored within the membrane, mechanically interlocked molecules, and examples that are activated in response to external stimuli.
Collapse
Affiliation(s)
- Toby G. Johnson
- Department of Chemistry, Chemistry
Research Laboratory, University of Oxford Mansfield Road, Oxford OX1 3TA United Kingdom
| | - Matthew J. Langton
- Department of Chemistry, Chemistry
Research Laboratory, University of Oxford Mansfield Road, Oxford OX1 3TA United Kingdom
| |
Collapse
|
8
|
Ostroumova OS, Efimova SS. Lipid-Centric Approaches in Combating Infectious Diseases: Antibacterials, Antifungals and Antivirals with Lipid-Associated Mechanisms of Action. Antibiotics (Basel) 2023; 12:1716. [PMID: 38136750 PMCID: PMC10741038 DOI: 10.3390/antibiotics12121716] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2023] [Revised: 12/05/2023] [Accepted: 12/08/2023] [Indexed: 12/24/2023] Open
Abstract
One of the global challenges of the 21st century is the increase in mortality from infectious diseases against the backdrop of the spread of antibiotic-resistant pathogenic microorganisms. In this regard, it is worth targeting antibacterials towards the membranes of pathogens that are quite conservative and not amenable to elimination. This review is an attempt to critically analyze the possibilities of targeting antimicrobial agents towards enzymes involved in pathogen lipid biosynthesis or towards bacterial, fungal, and viral lipid membranes, to increase the permeability via pore formation and to modulate the membranes' properties in a manner that makes them incompatible with the pathogen's life cycle. This review discusses the advantages and disadvantages of each approach in the search for highly effective but nontoxic antimicrobial agents. Examples of compounds with a proven molecular mechanism of action are presented, and the types of the most promising pharmacophores for further research and the improvement of the characteristics of antibiotics are discussed. The strategies that pathogens use for survival in terms of modulating the lipid composition and physical properties of the membrane, achieving a balance between resistance to antibiotics and the ability to facilitate all necessary transport and signaling processes, are also considered.
Collapse
Affiliation(s)
- Olga S. Ostroumova
- Laboratory of Membrane and Ion Channel Modeling, Institute of Cytology, Russian Academy of Sciences, Tikhoretsky Ave. 4, St. Petersburg 194064, Russia;
| | | |
Collapse
|
9
|
Shahpasand-Kroner H, Siddique I, Malik R, Linares GR, Ivanova MI, Ichida J, Weil T, Münch J, Sanchez-Garcia E, Klärner FG, Schrader T, Bitan G. Molecular Tweezers: Supramolecular Hosts with Broad-Spectrum Biological Applications. Pharmacol Rev 2023; 75:263-308. [PMID: 36549866 PMCID: PMC9976797 DOI: 10.1124/pharmrev.122.000654] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2022] [Revised: 10/14/2022] [Accepted: 10/19/2022] [Indexed: 12/24/2022] Open
Abstract
Lysine-selective molecular tweezers (MTs) are supramolecular host molecules displaying a remarkably broad spectrum of biologic activities. MTs act as inhibitors of the self-assembly and toxicity of amyloidogenic proteins using a unique mechanism. They destroy viral membranes and inhibit infection by enveloped viruses, such as HIV-1 and SARS-CoV-2, by mechanisms unrelated to their action on protein self-assembly. They also disrupt biofilm of Gram-positive bacteria. The efficacy and safety of MTs have been demonstrated in vitro, in cell culture, and in vivo, suggesting that these versatile compounds are attractive therapeutic candidates for various diseases, infections, and injuries. A lead compound called CLR01 has been shown to inhibit the aggregation of various amyloidogenic proteins, facilitate their clearance in vivo, prevent infection by multiple viruses, display potent anti-biofilm activity, and have a high safety margin in animal models. The inhibitory effect of CLR01 against amyloidogenic proteins is highly specific to abnormal self-assembly of amyloidogenic proteins with no disruption of normal mammalian biologic processes at the doses needed for inhibition. Therapeutic effects of CLR01 have been demonstrated in animal models of proteinopathies, lysosomal-storage diseases, and spinal-cord injury. Here we review the activity and mechanisms of action of these intriguing compounds and discuss future research directions. SIGNIFICANCE STATEMENT: Molecular tweezers are supramolecular host molecules with broad biological applications, including inhibition of abnormal protein aggregation, facilitation of lysosomal clearance of toxic aggregates, disruption of viral membranes, and interference of biofilm formation by Gram-positive bacteria. This review discusses the molecular and cellular mechanisms of action of the molecular tweezers, including the discovery of distinct mechanisms acting in vitro and in vivo, and the application of these compounds in multiple preclinical disease models.
Collapse
Affiliation(s)
- Hedieh Shahpasand-Kroner
- Department of Neurology, David Geffen School of Medicine (H.S.-K., I.S., R.M., G.B.), Brain Research Institute (G.B.), and Molecular Biology Institute (G.B.), University of California, Los Angeles, California; Department of Stem Cell Biology & Regenerative Medicine, Keck School of Medicine, University of Southern California, Los Angeles, California (G.R.L., J.I.); Department of Neurology, University of Michigan, Ann Arbor, Michigan (M.I.I.); Institute of Molecular Virology, Ulm University Medical Center, Ulm, Germany (T.W., J.M.); and Department of Computational Biochemistry (E.S.-G.) and Faculty of Chemistry (F-G.K., T.S.), University of Duisburg-Essen, Essen, Germany
| | - Ibrar Siddique
- Department of Neurology, David Geffen School of Medicine (H.S.-K., I.S., R.M., G.B.), Brain Research Institute (G.B.), and Molecular Biology Institute (G.B.), University of California, Los Angeles, California; Department of Stem Cell Biology & Regenerative Medicine, Keck School of Medicine, University of Southern California, Los Angeles, California (G.R.L., J.I.); Department of Neurology, University of Michigan, Ann Arbor, Michigan (M.I.I.); Institute of Molecular Virology, Ulm University Medical Center, Ulm, Germany (T.W., J.M.); and Department of Computational Biochemistry (E.S.-G.) and Faculty of Chemistry (F-G.K., T.S.), University of Duisburg-Essen, Essen, Germany
| | - Ravinder Malik
- Department of Neurology, David Geffen School of Medicine (H.S.-K., I.S., R.M., G.B.), Brain Research Institute (G.B.), and Molecular Biology Institute (G.B.), University of California, Los Angeles, California; Department of Stem Cell Biology & Regenerative Medicine, Keck School of Medicine, University of Southern California, Los Angeles, California (G.R.L., J.I.); Department of Neurology, University of Michigan, Ann Arbor, Michigan (M.I.I.); Institute of Molecular Virology, Ulm University Medical Center, Ulm, Germany (T.W., J.M.); and Department of Computational Biochemistry (E.S.-G.) and Faculty of Chemistry (F-G.K., T.S.), University of Duisburg-Essen, Essen, Germany
| | - Gabriel R Linares
- Department of Neurology, David Geffen School of Medicine (H.S.-K., I.S., R.M., G.B.), Brain Research Institute (G.B.), and Molecular Biology Institute (G.B.), University of California, Los Angeles, California; Department of Stem Cell Biology & Regenerative Medicine, Keck School of Medicine, University of Southern California, Los Angeles, California (G.R.L., J.I.); Department of Neurology, University of Michigan, Ann Arbor, Michigan (M.I.I.); Institute of Molecular Virology, Ulm University Medical Center, Ulm, Germany (T.W., J.M.); and Department of Computational Biochemistry (E.S.-G.) and Faculty of Chemistry (F-G.K., T.S.), University of Duisburg-Essen, Essen, Germany
| | - Magdalena I Ivanova
- Department of Neurology, David Geffen School of Medicine (H.S.-K., I.S., R.M., G.B.), Brain Research Institute (G.B.), and Molecular Biology Institute (G.B.), University of California, Los Angeles, California; Department of Stem Cell Biology & Regenerative Medicine, Keck School of Medicine, University of Southern California, Los Angeles, California (G.R.L., J.I.); Department of Neurology, University of Michigan, Ann Arbor, Michigan (M.I.I.); Institute of Molecular Virology, Ulm University Medical Center, Ulm, Germany (T.W., J.M.); and Department of Computational Biochemistry (E.S.-G.) and Faculty of Chemistry (F-G.K., T.S.), University of Duisburg-Essen, Essen, Germany
| | - Justin Ichida
- Department of Neurology, David Geffen School of Medicine (H.S.-K., I.S., R.M., G.B.), Brain Research Institute (G.B.), and Molecular Biology Institute (G.B.), University of California, Los Angeles, California; Department of Stem Cell Biology & Regenerative Medicine, Keck School of Medicine, University of Southern California, Los Angeles, California (G.R.L., J.I.); Department of Neurology, University of Michigan, Ann Arbor, Michigan (M.I.I.); Institute of Molecular Virology, Ulm University Medical Center, Ulm, Germany (T.W., J.M.); and Department of Computational Biochemistry (E.S.-G.) and Faculty of Chemistry (F-G.K., T.S.), University of Duisburg-Essen, Essen, Germany
| | - Tatjana Weil
- Department of Neurology, David Geffen School of Medicine (H.S.-K., I.S., R.M., G.B.), Brain Research Institute (G.B.), and Molecular Biology Institute (G.B.), University of California, Los Angeles, California; Department of Stem Cell Biology & Regenerative Medicine, Keck School of Medicine, University of Southern California, Los Angeles, California (G.R.L., J.I.); Department of Neurology, University of Michigan, Ann Arbor, Michigan (M.I.I.); Institute of Molecular Virology, Ulm University Medical Center, Ulm, Germany (T.W., J.M.); and Department of Computational Biochemistry (E.S.-G.) and Faculty of Chemistry (F-G.K., T.S.), University of Duisburg-Essen, Essen, Germany
| | - Jan Münch
- Department of Neurology, David Geffen School of Medicine (H.S.-K., I.S., R.M., G.B.), Brain Research Institute (G.B.), and Molecular Biology Institute (G.B.), University of California, Los Angeles, California; Department of Stem Cell Biology & Regenerative Medicine, Keck School of Medicine, University of Southern California, Los Angeles, California (G.R.L., J.I.); Department of Neurology, University of Michigan, Ann Arbor, Michigan (M.I.I.); Institute of Molecular Virology, Ulm University Medical Center, Ulm, Germany (T.W., J.M.); and Department of Computational Biochemistry (E.S.-G.) and Faculty of Chemistry (F-G.K., T.S.), University of Duisburg-Essen, Essen, Germany
| | - Elsa Sanchez-Garcia
- Department of Neurology, David Geffen School of Medicine (H.S.-K., I.S., R.M., G.B.), Brain Research Institute (G.B.), and Molecular Biology Institute (G.B.), University of California, Los Angeles, California; Department of Stem Cell Biology & Regenerative Medicine, Keck School of Medicine, University of Southern California, Los Angeles, California (G.R.L., J.I.); Department of Neurology, University of Michigan, Ann Arbor, Michigan (M.I.I.); Institute of Molecular Virology, Ulm University Medical Center, Ulm, Germany (T.W., J.M.); and Department of Computational Biochemistry (E.S.-G.) and Faculty of Chemistry (F-G.K., T.S.), University of Duisburg-Essen, Essen, Germany
| | - Frank-Gerrit Klärner
- Department of Neurology, David Geffen School of Medicine (H.S.-K., I.S., R.M., G.B.), Brain Research Institute (G.B.), and Molecular Biology Institute (G.B.), University of California, Los Angeles, California; Department of Stem Cell Biology & Regenerative Medicine, Keck School of Medicine, University of Southern California, Los Angeles, California (G.R.L., J.I.); Department of Neurology, University of Michigan, Ann Arbor, Michigan (M.I.I.); Institute of Molecular Virology, Ulm University Medical Center, Ulm, Germany (T.W., J.M.); and Department of Computational Biochemistry (E.S.-G.) and Faculty of Chemistry (F-G.K., T.S.), University of Duisburg-Essen, Essen, Germany
| | - Thomas Schrader
- Department of Neurology, David Geffen School of Medicine (H.S.-K., I.S., R.M., G.B.), Brain Research Institute (G.B.), and Molecular Biology Institute (G.B.), University of California, Los Angeles, California; Department of Stem Cell Biology & Regenerative Medicine, Keck School of Medicine, University of Southern California, Los Angeles, California (G.R.L., J.I.); Department of Neurology, University of Michigan, Ann Arbor, Michigan (M.I.I.); Institute of Molecular Virology, Ulm University Medical Center, Ulm, Germany (T.W., J.M.); and Department of Computational Biochemistry (E.S.-G.) and Faculty of Chemistry (F-G.K., T.S.), University of Duisburg-Essen, Essen, Germany
| | - Gal Bitan
- Department of Neurology, David Geffen School of Medicine (H.S.-K., I.S., R.M., G.B.), Brain Research Institute (G.B.), and Molecular Biology Institute (G.B.), University of California, Los Angeles, California; Department of Stem Cell Biology & Regenerative Medicine, Keck School of Medicine, University of Southern California, Los Angeles, California (G.R.L., J.I.); Department of Neurology, University of Michigan, Ann Arbor, Michigan (M.I.I.); Institute of Molecular Virology, Ulm University Medical Center, Ulm, Germany (T.W., J.M.); and Department of Computational Biochemistry (E.S.-G.) and Faculty of Chemistry (F-G.K., T.S.), University of Duisburg-Essen, Essen, Germany
| |
Collapse
|
10
|
Park S, Cho NJ. Lipid Membrane Interface Viewpoint: From Viral Entry to Antiviral and Vaccine Development. LANGMUIR : THE ACS JOURNAL OF SURFACES AND COLLOIDS 2023; 39:1-11. [PMID: 36576966 DOI: 10.1021/acs.langmuir.2c02501] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/17/2023]
Abstract
Membrane-enveloped viruses are responsible for most viral pandemics in history, and more effort is needed to advance broadly applicable countermeasures to mitigate the impact of future outbreaks. In this Perspective, we discuss how biosensing techniques associated with lipid model membrane platforms are contributing to improving our mechanistic knowledge of membrane fusion and destabilization that is closely linked to viral entry as well as vaccine and antiviral drug development. A key benefit of these platforms is the simplicity of interpreting the results which can be complemented by other techniques to decipher more complicated biological observations and evaluate the biophysical functionalities that can be correlated to biological activities. Then, we introduce exciting application examples of membrane-targeting antivirals that have been refined over time and will continue to improve based on biophysical insights. Two ways to abrogate the function of viral membranes are introduced here: (1) selective disruption of the viral membrane structure and (2) alteration of the membrane component. While both methods are suitable for broadly useful antivirals, the latter also has the potential to produce an inactivated vaccine. Collectively, we emphasize how biosensing tools based on membrane interfacial science can provide valuable information that could be translated into biomedicines and improve their selectivity and performance.
Collapse
Affiliation(s)
- Soohyun Park
- School of Materials Science and Engineering, Nanyang Technological University, 50 Nanyang Avenue, Singapore 639798, Singapore
| | - Nam-Joon Cho
- School of Materials Science and Engineering, Nanyang Technological University, 50 Nanyang Avenue, Singapore 639798, Singapore
| |
Collapse
|
11
|
Weil T, Münch J. Molekulare Pinzetten als Breitbandinhibitor viraler Infektionen. BIOSPEKTRUM : ZEITSCHRIFT DER GESELLSCHAFT FUR BIOLOGISHE CHEMIE (GBCH) UND DER VEREINIGUNG FUR ALLGEMEINE UND ANGEWANDTE MIKROBIOLOGIE (VAAM) 2023; 29:150-152. [PMID: 37073322 PMCID: PMC10101532 DOI: 10.1007/s12268-023-1906-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 04/20/2023]
Abstract
The SARS-CoV-2 pandemic once again highlighted the constant threat posed by viruses. Specific therapeutics are highly warranted, but their development is time consuming and cost intensive. Broad-spectrum antivirals provide a promising option for fast application to treat circulating or newly emerged viruses. Here, we introduce molecular tweezers as broad-spectrum antivirals, which abrogate viral infection by directly targeting the viral membrane. Furthermore, we discuss the current stage of tweezer development to fight SARS-CoV-2 and other respiratory viruses.
Collapse
Affiliation(s)
- Tatjana Weil
- Institut für Molekulare Virologie, Universitätsklinikum Ulm, Meyerhofstraße 1 (ZQB), D-89081 Ulm, Deutschland
| | - Jan Münch
- Institut für Molekulare Virologie, Universitätsklinikum Ulm, Meyerhofstraße 1 (ZQB), D-89081 Ulm, Deutschland
| |
Collapse
|
12
|
Biria D. Tailored lipopeptide surfactants as potentially effective drugs to treat SARS-CoV-2 infection. Med Hypotheses 2022; 167:110948. [PMID: 36157252 PMCID: PMC9482169 DOI: 10.1016/j.mehy.2022.110948] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2022] [Revised: 07/18/2022] [Accepted: 07/23/2022] [Indexed: 11/22/2022]
Abstract
Finding effective drugs to treat SARS-CoV-2 infection as a complementary step to the extensive vaccination is of the great importance to overcome the current pandemic situation. It has been shown that some bio-active unsaturated fatty acids such as Arachidonic Acid (AA) can reduce the infection severity and even destroy the virus by disintegration of the virus lipid envelope. On the other hand, it has been reported that several designed peptides with an activity similar to the angiotensin converting enzyme 2 (ACE-2), which has a high affinity towards the novel corona virus spike protein, can inhibit the viral infection through concealing the spike proteins from the cell surfaces ACE-2. Binding the mentioned peptides to the bio-active lipids like AA will result in a lipopeptide surfactant molecule with the synergistic effect of both the active moieties in its structure to treat the novel corona infection. In addition, the peptide segment increases the aqueous solubility of the lipid segment and enables the targeted delivery of the surfactant molecule to the virus. The resultant lipopeptide would be a potentially effective drug for SARS-CoV-2 infection treatment with the minimum side effects.
Collapse
Affiliation(s)
- Davoud Biria
- Department of Biotechnology, Faculty of Biological Sciences and Technologies, University of Isfahan, Isfahan, Iran
| |
Collapse
|
13
|
Weil T, Kirupakaran A, Le MH, Rebmann P, Mieres-Perez J, Issmail L, Conzelmann C, Müller JA, Rauch L, Gilg A, Wettstein L, Groß R, Read C, Bergner T, Pålsson SA, Uhlig N, Eberlein V, Wöll H, Klärner FG, Stenger S, Kümmerer BM, Streeck H, Fois G, Frick M, Braubach P, Spetz AL, Grunwald T, Shorter J, Sanchez-Garcia E, Schrader T, Münch J. Advanced Molecular Tweezers with Lipid Anchors against SARS-CoV-2 and Other Respiratory Viruses. JACS AU 2022; 2:2187-2202. [PMID: 36186568 PMCID: PMC9516563 DOI: 10.1021/jacsau.2c00220] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/07/2022] [Revised: 07/06/2022] [Accepted: 07/07/2022] [Indexed: 06/16/2023]
Abstract
The COVID-19 pandemic caused by SARS-CoV-2 presents a global health emergency. Therapeutic options against SARS-CoV-2 are still very limited but urgently required. Molecular tweezers are supramolecular agents that destabilize the envelope of viruses resulting in a loss of viral infectivity. Here, we show that first-generation tweezers, CLR01 and CLR05, disrupt the SARS-CoV-2 envelope and abrogate viral infectivity. To increase the antiviral activity, a series of 34 advanced molecular tweezers were synthesized by insertion of aliphatic or aromatic ester groups on the phosphate moieties of the parent molecule CLR01. A structure-activity relationship study enabled the identification of tweezers with a markedly enhanced ability to destroy lipid bilayers and to suppress SARS-CoV-2 infection. Selected tweezer derivatives retain activity in airway mucus and inactivate the SARS-CoV-2 wildtype and variants of concern as well as respiratory syncytial, influenza, and measles viruses. Moreover, inhibitory activity of advanced tweezers against respiratory syncytial virus and SARS-CoV-2 was confirmed in mice. Thus, potentiated tweezers are broad-spectrum antiviral agents with great prospects for clinical development to combat highly pathogenic viruses.
Collapse
Affiliation(s)
- Tatjana Weil
- Institute
of Molecular Virology, Ulm University Medical
Center, Ulm89081, Germany
| | - Abbna Kirupakaran
- Faculty
of Chemistry, University of Duisburg-Essen, Essen45117, Germany
| | - My-Hue Le
- Faculty
of Chemistry, University of Duisburg-Essen, Essen45117, Germany
| | - Philipp Rebmann
- Faculty
of Chemistry, University of Duisburg-Essen, Essen45117, Germany
| | - Joel Mieres-Perez
- Computational
Biochemistry, University of Duisburg-Essen, Essen45117, Germany
| | - Leila Issmail
- Fraunhofer
Institute for Cell Therapy and Immunology IZI, Leipzig04103, Germany
| | - Carina Conzelmann
- Institute
of Molecular Virology, Ulm University Medical
Center, Ulm89081, Germany
| | - Janis A. Müller
- Institute
of Virology, Philipps University of Marburg, Marburg35043, Germany
| | - Lena Rauch
- Institute
of Molecular Virology, Ulm University Medical
Center, Ulm89081, Germany
| | - Andrea Gilg
- Institute
of Molecular Virology, Ulm University Medical
Center, Ulm89081, Germany
| | - Lukas Wettstein
- Institute
of Molecular Virology, Ulm University Medical
Center, Ulm89081, Germany
| | - Rüdiger Groß
- Institute
of Molecular Virology, Ulm University Medical
Center, Ulm89081, Germany
| | - Clarissa Read
- Central
Facility for Electron Microscopy, Ulm University, Ulm89081, Germany
- Institute
of Virology, Ulm University Medical Center, Ulm89081, Germany
| | - Tim Bergner
- Central
Facility for Electron Microscopy, Ulm University, Ulm89081, Germany
| | - Sandra Axberg Pålsson
- Department
of Molecular Biosciences, The Wenner-Gren Institute, Stockholm University, Stockholm10691, Sweden
| | - Nadja Uhlig
- Fraunhofer
Institute for Cell Therapy and Immunology IZI, Leipzig04103, Germany
| | - Valentina Eberlein
- Fraunhofer
Institute for Cell Therapy and Immunology IZI, Leipzig04103, Germany
| | - Heike Wöll
- Faculty
of Chemistry, University of Duisburg-Essen, Essen45117, Germany
| | | | - Steffen Stenger
- Institute
for Microbiology and Hygiene, Ulm University
Medical Center, Ulm89081, Germany
| | - Beate M. Kümmerer
- Institute
of Virology, Medical Faculty, University
of Bonn, Bonn53127, Germany
- German
Centre for Infection Research (DZIF),
partner site Bonn-Cologne, Bonn53127, Germany
| | - Hendrik Streeck
- Institute
of Virology, Medical Faculty, University
of Bonn, Bonn53127, Germany
- German
Centre for Infection Research (DZIF),
partner site Bonn-Cologne, Bonn53127, Germany
| | - Giorgio Fois
- Institute
of General Physiology, Ulm University, Ulm89081, Germany
| | - Manfred Frick
- Institute
of General Physiology, Ulm University, Ulm89081, Germany
| | - Peter Braubach
- Institute
of Pathology, Hannover Medical School (MHH), Hannover30625, Germany
| | - Anna-Lena Spetz
- Department
of Molecular Biosciences, The Wenner-Gren Institute, Stockholm University, Stockholm10691, Sweden
| | - Thomas Grunwald
- Fraunhofer
Institute for Cell Therapy and Immunology IZI, Leipzig04103, Germany
| | - James Shorter
- Department
of Biochemistry and Biophysics, Perelman
School of Medicine at the University of Pennsylvania, Philadelphia19104, United States
| | - Elsa Sanchez-Garcia
- Computational
Biochemistry, University of Duisburg-Essen, Essen45117, Germany
| | - Thomas Schrader
- Faculty
of Chemistry, University of Duisburg-Essen, Essen45117, Germany
| | - Jan Münch
- Institute
of Molecular Virology, Ulm University Medical
Center, Ulm89081, Germany
| |
Collapse
|
14
|
Vogel V, Olari LR, Jachmann M, Reich SJ, Häring M, Kissmann AK, Rosenau F, Riedel CU, Münch J, Spellerberg B. The bacteriocin Angicin interferes with bacterial membrane integrity through interaction with the mannose phosphotransferase system. Front Microbiol 2022; 13:991145. [PMID: 36147850 PMCID: PMC9486217 DOI: 10.3389/fmicb.2022.991145] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2022] [Accepted: 08/17/2022] [Indexed: 12/29/2022] Open
Abstract
In a natural environment, bacteria are members of multispecies communities. To compete with rival species, bacteria produce antimicrobial peptides (AMPs), called bacteriocins. Bacteriocins are small, cationic, ribosomally synthesized peptides, which normally inhibit closely related species of the producing organism. Bacteriocin production is best studied in lactic bacteria (LAB). Streptococcus anginosus, belonging to LAB, produces the potent bacteriocin Angicin, which shows inhibitory activity against other streptococci, Listeria monocytogenes and vancomycin resistant Enterococcus faecium (VRE). Furthermore, Angicin shows a high resistance toward pH changes and heat, rendering it an interesting candidate for food preservation or clinical applications. The inhibitory activity of Angicin depends on the presence of a mannose phosphotransferase system (Man-PTS) in target cells, since L. monocytogenes harboring a deletion in an extracellular loop of this system is no longer sensitive to Angicin. Furthermore, we demonstrated by liposome leakage and pHluorin assays that Angicin destroys membrane integrity but shows only low cytotoxicity against human cell lines. In conclusion, we show that Angicin has a detrimental effect on the membrane of target organisms by using the Man-PTS as a receptor.
Collapse
Affiliation(s)
- Verena Vogel
- Institute of Medical Microbiology and Hygiene, Ulm University Medical Center, Ulm, Germany
| | - Lia-Raluca Olari
- Institute of Molecular Virology, Ulm University Medical Center, Ulm, Germany
| | - Marie Jachmann
- Institute of Medical Microbiology and Hygiene, Ulm University Medical Center, Ulm, Germany
| | - Sebastian J. Reich
- Institute of Microbiology and Biotechnology, University of Ulm, Ulm, Germany
| | - Michelle Häring
- Institute of Pharmaceutical Biotechnology, University of Ulm, Ulm, Germany
| | | | - Frank Rosenau
- Institute of Pharmaceutical Biotechnology, University of Ulm, Ulm, Germany
| | - Christian U. Riedel
- Institute of Microbiology and Biotechnology, University of Ulm, Ulm, Germany
| | - Jan Münch
- Institute of Molecular Virology, Ulm University Medical Center, Ulm, Germany
| | - Barbara Spellerberg
- Institute of Medical Microbiology and Hygiene, Ulm University Medical Center, Ulm, Germany
- *Correspondence: Barbara Spellerberg,
| |
Collapse
|
15
|
Groß R, Dias Loiola LM, Issmail L, Uhlig N, Eberlein V, Conzelmann C, Olari L, Rauch L, Lawrenz J, Weil T, Müller JA, Cardoso MB, Gilg A, Larsson O, Höglund U, Pålsson SA, Tvilum AS, Løvschall KB, Kristensen MM, Spetz A, Hontonnou F, Galloux M, Grunwald T, Zelikin AN, Münch J. Macromolecular Viral Entry Inhibitors as Broad-Spectrum First-Line Antivirals with Activity against SARS-CoV-2. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2022; 9:e2201378. [PMID: 35543527 PMCID: PMC9284172 DOI: 10.1002/advs.202201378] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/09/2022] [Revised: 04/11/2022] [Indexed: 05/03/2023]
Abstract
Inhibitors of viral cell entry based on poly(styrene sulfonate) and its core-shell nanoformulations based on gold nanoparticles are investigated against a panel of viruses, including clinical isolates of SARS-CoV-2. Macromolecular inhibitors are shown to exhibit the highly sought-after broad-spectrum antiviral activity, which covers most analyzed enveloped viruses and all of the variants of concern for SARS-CoV-2 tested. The inhibitory activity is quantified in vitro in appropriate cell culture models and for respiratory viral pathogens (respiratory syncytial virus and SARS-CoV-2) in mice. Results of this study comprise a significant step along the translational path of macromolecular inhibitors of virus cell entry, specifically against enveloped respiratory viruses.
Collapse
Affiliation(s)
- Rüdiger Groß
- Institute of Molecular VirologyUlm University Medical CenterUlm89081Germany
| | - Lívia Mesquita Dias Loiola
- Department of Chemistry and iNano Interdisciplinary Nanoscience CentreAarhus UniversityAarhus8000Denmark
- Brazilian Synchrotron Light LaboratoryBrazilian Center for Research in Energy and MaterialsCampinasSão Paulo13083‐970Brazil
| | - Leila Issmail
- Fraunhofer Institute for Cell Therapy and Immunology IZILeipzig04103Germany
| | - Nadja Uhlig
- Fraunhofer Institute for Cell Therapy and Immunology IZILeipzig04103Germany
| | - Valentina Eberlein
- Fraunhofer Institute for Cell Therapy and Immunology IZILeipzig04103Germany
| | - Carina Conzelmann
- Institute of Molecular VirologyUlm University Medical CenterUlm89081Germany
| | - Lia‐Raluca Olari
- Institute of Molecular VirologyUlm University Medical CenterUlm89081Germany
| | - Lena Rauch
- Institute of Molecular VirologyUlm University Medical CenterUlm89081Germany
| | - Jan Lawrenz
- Institute of Molecular VirologyUlm University Medical CenterUlm89081Germany
| | - Tatjana Weil
- Institute of Molecular VirologyUlm University Medical CenterUlm89081Germany
| | - Janis A. Müller
- Institute of Molecular VirologyUlm University Medical CenterUlm89081Germany
| | - Mateus Borba Cardoso
- Brazilian Synchrotron Light LaboratoryBrazilian Center for Research in Energy and MaterialsCampinasSão Paulo13083‐970Brazil
| | - Andrea Gilg
- Institute of Molecular VirologyUlm University Medical CenterUlm89081Germany
| | | | | | - Sandra Axberg Pålsson
- Department of Molecular BiosciencesThe Wenner‐Gren Institute Stockholm UniversityStockholm10691Sweden
| | - Anna Selch Tvilum
- Department of Chemistry and iNano Interdisciplinary Nanoscience CentreAarhus UniversityAarhus8000Denmark
| | - Kaja Borup Løvschall
- Department of Chemistry and iNano Interdisciplinary Nanoscience CentreAarhus UniversityAarhus8000Denmark
| | - Maria M. Kristensen
- Department of Chemistry and iNano Interdisciplinary Nanoscience CentreAarhus UniversityAarhus8000Denmark
| | - Anna‐Lena Spetz
- Department of Molecular BiosciencesThe Wenner‐Gren Institute Stockholm UniversityStockholm10691Sweden
| | | | - Marie Galloux
- Université Paris‐SaclayINRAE, UVSQ, VIMJouy‐en‐Josas78352France
| | - Thomas Grunwald
- Fraunhofer Institute for Cell Therapy and Immunology IZILeipzig04103Germany
| | - Alexander N. Zelikin
- Department of Chemistry and iNano Interdisciplinary Nanoscience CentreAarhus UniversityAarhus8000Denmark
| | - Jan Münch
- Institute of Molecular VirologyUlm University Medical CenterUlm89081Germany
| |
Collapse
|
16
|
Yao L, Zhou Z, Wang S, Zou Q, Wang HX, Ma LX, Wang S, Zhang X. Phosphorylation of covalent organic framework nanospheres for inhibition of amyloid-β peptide fibrillation. Chem Sci 2022; 13:5902-5912. [PMID: 35685783 PMCID: PMC9132083 DOI: 10.1039/d2sc00253a] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2022] [Accepted: 04/21/2022] [Indexed: 12/27/2022] Open
Abstract
The development and exploration of new nanostructural inhibitors against Alzheimer's disease (AD)-associated amyloid-β (Aβ) fibrillation have attracted extensive attention and become a new frontier in nanomedicine. However, focusing on finding an effective nanostructure is one of the most challenging parts of the therapeutics task. Herein, nanoscale spherical covalent organic frameworks (COFs) via post-synthetic functionalization with sodium phosphate (SP) groups on the channel networks were found to efficiently inhibit Aβ fibrillation. The as-prepared uniform SP-COF nanospheres with high surface area, good crystallinity, and chemical stability were characterized by multifarious microscopic and spectroscopic techniques. Moreover, molecular dynamics simulation together with fibrillation kinetics and cytotoxicity assay experiments shows that there were restricted-access adsorption channels in the SP-COFs which were formed by the cavities with size and functional groups accommodated to the Aβ peptide sequence and significantly affected the fibrillation and cytotoxicity of Aβ. Transmission electron microscopy (TEM), dynamic light scattering (DLS) monitoring, isothermal titration calorimetry (ITC), Fourier transform infrared (FT-IR) and circular dichroism (CD) spectra measurements, and confocal imaging observation were performed to understand the inhibition mechanism and influencing factors of the SP-COFs. To our knowledge, our strategy is the first exploration of COF-based anti-amyloidogenic nanomaterials with high affinity and specific targeting, which are crucial for the inhibition of Aβ fibrillation for AD prevention and treatment. Nanoscale spherical COFs via phosphorylation functionalization were found to efficiently inhibit fibrillation of the Alzheimer's disease-associated Aβ peptide.![]()
Collapse
Affiliation(s)
- Linli Yao
- Collaborative Innovation Center for Advanced Organic Chemical Materials Co-constructed by the Province and Ministry, College of Chemistry and Chemical Engineering, Hubei University Wuhan 430062 China
| | - Zhe Zhou
- Department of Neurology, The First Hospital of Lanzhou University Lanzhou 730000 China
| | - Suxiao Wang
- Collaborative Innovation Center for Advanced Organic Chemical Materials Co-constructed by the Province and Ministry, College of Chemistry and Chemical Engineering, Hubei University Wuhan 430062 China
| | - Qichao Zou
- Collaborative Innovation Center for Advanced Organic Chemical Materials Co-constructed by the Province and Ministry, College of Chemistry and Chemical Engineering, Hubei University Wuhan 430062 China
| | - Hang-Xing Wang
- Collaborative Innovation Center for Advanced Organic Chemical Materials Co-constructed by the Province and Ministry, College of Chemistry and Chemical Engineering, Hubei University Wuhan 430062 China
| | - Li-Xin Ma
- State Key Laboratory of Biocatalysis and Enzyme Engineering, School of Life Science, Hubei University Wuhan 430062 China
| | - Shengfu Wang
- Collaborative Innovation Center for Advanced Organic Chemical Materials Co-constructed by the Province and Ministry, College of Chemistry and Chemical Engineering, Hubei University Wuhan 430062 China
| | - Xiuhua Zhang
- Collaborative Innovation Center for Advanced Organic Chemical Materials Co-constructed by the Province and Ministry, College of Chemistry and Chemical Engineering, Hubei University Wuhan 430062 China
| |
Collapse
|
17
|
Zhou X, Shi M, Wang X, Xu D. Exploring the Binding Mechanism of a Supramolecular Tweezer CLR01 to 14-3-3σ Protein via Well-Tempered Metadynamics. Front Chem 2022; 10:921695. [PMID: 35646830 PMCID: PMC9133541 DOI: 10.3389/fchem.2022.921695] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2022] [Accepted: 04/25/2022] [Indexed: 11/17/2022] Open
Abstract
Using supramolecules for protein function regulation is an effective strategy in chemical biology and drug discovery. However, due to the presence of multiple binding sites on protein surfaces, protein function regulation via selective binding of supramolecules is challenging. Recently, the functions of 14-3-3 proteins, which play an important role in regulating intracellular signaling pathways via protein–protein interactions, have been modulated using a supramolecular tweezer, CLR01. However, the binding mechanisms of the tweezer molecule to 14-3-3 proteins are still unclear, which has hindered the development of novel supramolecules targeting the 14-3-3 proteins. Herein, the binding mechanisms of the tweezer to the lysine residues on 14-3-3σ (an isoform in 14-3-3 protein family) were explored by well-tempered metadynamics. The results indicated that the inclusion complex formed between the protein and supramolecule is affected by both kinetic and thermodynamic factors. In particular, simulations confirmed that K214 could form a strong binding complex with the tweezer; the binding free energy was calculated to be −10.5 kcal·mol−1 with an association barrier height of 3.7 kcal·mol−1. In addition, several other lysine residues on 14-3-3σ were identified as being well-recognized by the tweezer, which agrees with experimental results, although only K214/tweezer was co-crystallized. Additionally, the binding mechanisms of the tweezer to all lysine residues were analyzed by exploring the representative conformations during the formation of the inclusion complex. This could be helpful for the development of new inhibitors based on tweezers with more functions against 14-3-3 proteins via modifications of CLR01. We also believe that the proposed computational strategies can be extended to understand the binding mechanism of multi-binding sites proteins with supramolecules and will, thus, be useful toward drug design.
Collapse
Affiliation(s)
- Xin Zhou
- College of Chemistry, MOE Key Laboratory of Green Chemistry and Technology, Sichuan University, Chengdu, China
| | - Mingsong Shi
- State Key Laboratory of Biotherapy/Collaborative Innovation Center of Biotherapy and Cancer Center, West China Hospital, Sichuan University, Chengdu, China
| | - Xin Wang
- College of Chemistry, MOE Key Laboratory of Green Chemistry and Technology, Sichuan University, Chengdu, China
- *Correspondence: Xin Wang, ; Dingguo Xu,
| | - Dingguo Xu
- College of Chemistry, MOE Key Laboratory of Green Chemistry and Technology, Sichuan University, Chengdu, China
- Research Center for Material Genome Engineering, Sichuan University, Chengdu, China
- *Correspondence: Xin Wang, ; Dingguo Xu,
| |
Collapse
|
18
|
Extracellular vesicle-derived miR-1249-5p regulates influenza A virus-induced acute lung injury in RAW246.7 cells through targeting SLC4A1. Microbes Infect 2022; 24:104998. [DOI: 10.1016/j.micinf.2022.104998] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2021] [Revised: 04/27/2022] [Accepted: 04/29/2022] [Indexed: 11/19/2022]
|
19
|
Dergham M, Lin S, Geng J. Supramolecular Self-Assembly in Living Cells. Angew Chem Int Ed Engl 2022; 61:e202114267. [PMID: 35037350 DOI: 10.1002/anie.202114267] [Citation(s) in RCA: 51] [Impact Index Per Article: 17.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2021] [Indexed: 12/17/2022]
Abstract
Supramolecular interactions rely on non-covalent forces, such as hydrophobic effects, hydrogen-bonding, and electrostatic interactions, which govern many intracellular biological pathways. In cellulo supramolecular self-assembly is mainly based on host-guest interactions, changes in pH, enzymes, and polymerization-induced self-assembly to accurately induce various unnatural reactions without disturbing natural biological processes. This process can produce synthetic biocompatible macromolecules to control cell properties and regulate biological functions, such as cell proliferation and differentiation. This Minireview focuses on the latest reports in the field of in cellulo supramolecular self-assembly and anticipates future advances regarding its activation in response to internal and external stimuli, such as pH changes, reactive oxygen species, and enzymes, as well as external light illumination.
Collapse
Affiliation(s)
- Mohamed Dergham
- Centre for Polymers in Medicine, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen, Nanshan, 518055, China.,University of Chinese Academy of Science, Beijing, 100049, China
| | - Shanmeng Lin
- Centre for Polymers in Medicine, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen, Nanshan, 518055, China
| | - Jin Geng
- Centre for Polymers in Medicine, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen, Nanshan, 518055, China
| |
Collapse
|
20
|
Gupta A, Dey S, Bhowmik D, Maiti S. Coexisting Ordered and Disordered Membrane Phases Have Distinct Modes of Interaction with Disease-Associated Oligomers. J Phys Chem B 2022; 126:1016-1023. [PMID: 35104126 DOI: 10.1021/acs.jpcb.1c09421] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Abstract
Ordered membrane domains are thought to influence the attachment and insertion of toxic amyloid oligomers, and consequently, their toxicity. However, if and how the molecular aspects of this interaction depend on the membrane order is poorly understood. Here we measure the affinity, location, and degree of insertion of the small oligomers of hIAPP (human Islet Amyloid Polypeptide, associated with Type II diabetes) at near-physiological concentrations to adjacent domains of a biphasic lipid bilayer. Using simultaneous atomic force, confocal and fluorescence lifetime microscopy (AFM-FLIM), we find that hIAPP oligomers have a nearly 8-fold higher affinity to the disordered domains over the ordered domains. To probe whether this difference indicates different modes of interaction, we measure the change of lifetime of peptide-attached fluorescent labels induced by soluble fluorescence quenchers and also measure the kinetics of localized photobleaching. We find that in the raft-like ordered domains, the oligomers primarily lie on the aqueous interface with limited membrane penetration. However, in the neighboring disordered domains, their C-termini penetrate deeper into the lipid bilayer. We conclude that local membrane order determines not only the affinity but also the mode of interaction of amyloid oligomers, which may have significant implications for disease mechanisms.
Collapse
Affiliation(s)
- Ankur Gupta
- Tata Institute of Fundamental Research, Homi Bhabha Road, Colaba, Mumbai 400005, India
| | - Simli Dey
- Tata Institute of Fundamental Research, Homi Bhabha Road, Colaba, Mumbai 400005, India
| | - Debanjan Bhowmik
- Tata Institute of Fundamental Research, Homi Bhabha Road, Colaba, Mumbai 400005, India
| | - Sudipta Maiti
- Tata Institute of Fundamental Research, Homi Bhabha Road, Colaba, Mumbai 400005, India
| |
Collapse
|
21
|
Dergham M, Lin S, Geng J. Supramolecular Self‐assembly in Living Cells. Angew Chem Int Ed Engl 2022. [DOI: 10.1002/ange.202114267] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
Affiliation(s)
- Mohamed Dergham
- Shenzhen Institutes of Advanced Technology Chinese Academy of Sciences Biomedicine and Biotechnology CHINA
| | - Shanmeng Lin
- Shenzhen Institutes of Advanced Technology Chinese Academy of Sciences Biomedicine and Biotechnology CHINA
| | - Jin Geng
- Shenzhen Institutes of Advanced Technology Chinese Academy of Sciences Institute of Biomedicine and Biotechnology Xuyuan Road 518055 Shenzhen CHINA
| |
Collapse
|
22
|
Le MH, Taghuo-Kaptouom E, Schrader T. Molecular Tweezers – a new class of potent broad-spectrum antivirals against enveloped viruses. Chem Commun (Camb) 2022; 58:2954-2966. [DOI: 10.1039/d1cc06737k] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
Abstract
A new supramolecular approach to broad spectrum antivirals utilizes host guest chemistry between molecular tweezers and lysine/arginine as well as choline. Basic amino acids in amyloid-forming SEVI peptides (semen-derived enhancers...
Collapse
|
23
|
Li Z, Siddique I, Hadrović I, Kirupakaran A, Li J, Zhang Y, Klärner FG, Schrader T, Bitan G. Lysine-selective molecular tweezers are cell penetrant and concentrate in lysosomes. Commun Biol 2021; 4:1076. [PMID: 34521989 PMCID: PMC8440717 DOI: 10.1038/s42003-021-02603-2] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2021] [Accepted: 08/24/2021] [Indexed: 11/09/2022] Open
Abstract
Lysine-selective molecular tweezers are promising drug candidates against proteinopathies, viral infection, and bacterial biofilm. Despite demonstration of their efficacy in multiple cellular and animal models, important questions regarding their mechanism of action, including cell penetrance and intracellular distribution, have not been answered to date. The main impediment to answering these questions has been the low intrinsic fluorescence of the main compound tested to date, called CLR01. Here, we address these questions using new fluorescently labeled molecular tweezers derivatives. We show that these compounds are internalized in neurons and astrocytes, at least partially through dynamin-dependent endocytosis. In addition, we demonstrate that the molecular tweezers concentrate rapidly in acidic compartments, primarily lysosomes. Accumulation of molecular tweezers in lysosomes may occur both through the endosomal-lysosomal pathway and via the autophagy-lysosome pathway. Moreover, by visualizing colocalization of molecular tweezers, lysosomes, and tau aggregates we show that lysosomes likely are the main site for the intracellular anti-amyloid activity of molecular tweezers. These findings have important implications for the mechanism of action of molecular tweezers in vivo, explaining how administration of low doses of the compounds achieves high effective concentrations where they are needed, and supporting the development of these compounds as drugs for currently cureless proteinopathies.
Collapse
Affiliation(s)
- Zizheng Li
- Department of Neurology, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA, USA
| | - Ibrar Siddique
- Department of Neurology, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA, USA
| | - Inesa Hadrović
- Institute of Chemistry, University of Duisburg-Essen, Essen, Germany
| | - Abbna Kirupakaran
- Institute of Chemistry, University of Duisburg-Essen, Essen, Germany
| | - Jiwen Li
- Department of Psychiatry and Biobehavioral Sciences, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA, USA
| | - Ye Zhang
- Department of Psychiatry and Biobehavioral Sciences, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA, USA.,Brain Research Institute, University of California, Los Angeles, Los Angeles, CA, USA.,Molecular Biology Institute, University of California, Los Angeles, Los Angeles, CA, USA
| | | | - Thomas Schrader
- Institute of Chemistry, University of Duisburg-Essen, Essen, Germany
| | - Gal Bitan
- Department of Neurology, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA, USA. .,Brain Research Institute, University of California, Los Angeles, Los Angeles, CA, USA. .,Molecular Biology Institute, University of California, Los Angeles, Los Angeles, CA, USA.
| |
Collapse
|
24
|
Brenner S, Braun B, Read C, Weil T, Walther P, Schrader T, Münch J, von Einem J. The Molecular Tweezer CLR01 Inhibits Antibody-Resistant Cell-to-Cell Spread of Human Cytomegalovirus. Viruses 2021; 13:v13091685. [PMID: 34578265 PMCID: PMC8472163 DOI: 10.3390/v13091685] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2021] [Revised: 08/20/2021] [Accepted: 08/23/2021] [Indexed: 02/06/2023] Open
Abstract
Human cytomegalovirus (HCMV) uses two major ways for virus dissemination: infection by cell-free virus and direct cell-to-cell spread. Neutralizing antibodies can efficiently inhibit infection by cell-free virus but mostly fail to prevent cell-to-cell transmission. Here, we show that the ‘molecular tweezer’ CLR01, a broad-spectrum antiviral agent, is not only highly active against infection with cell-free virus but most remarkably inhibits antibody-resistant direct cell-to-cell spread of HCMV. The inhibition of cell-to-cell spread by CLR01 was not limited to HCMV but was also shown for the alphaherpesviruses herpes simplex viruses 1 and 2 (HSV-1, -2). CLR01 is a rapid acting small molecule that inhibits HCMV entry at the attachment and penetration steps. Electron microscopy of extracellular virus particles indicated damage of the viral envelope by CLR01, which likely impairs the infectivity of virus particles. The rapid inactivation of viral particles by CLR01, the viral envelope as the main target, and the inhibition of virus entry at different stages are presumably the key to inhibition of cell-free virus infection and cell-to-cell spread by CLR01. Importance: While cell-free spread enables the human cytomegalovirus (HCMV) and other herpesviruses to transmit between hosts, direct cell-to-cell spread is thought to be more relevant for in vivo dissemination within infected tissues. Cell-to-cell spread is resistant to neutralizing antibodies, thus contributing to the maintenance of virus infection and virus dissemination in the presence of an intact immune system. Therefore, it would be therapeutically interesting to target this mode of spread in order to treat severe HCMV infections and to prevent dissemination of virus within the infected host. The molecular tweezer CLR01 exhibits broad-spectrum antiviral activity against a number of enveloped viruses and efficiently blocks antibody-resistant cell-to-cell spread of HCMV, thus representing a novel class of small molecules with promising antiviral activity.
Collapse
Affiliation(s)
- Sina Brenner
- Institute of Virology, Ulm University Medical Center, 89081 Ulm, Germany; (S.B.); (B.B.); (C.R.)
| | - Berenike Braun
- Institute of Virology, Ulm University Medical Center, 89081 Ulm, Germany; (S.B.); (B.B.); (C.R.)
| | - Clarissa Read
- Institute of Virology, Ulm University Medical Center, 89081 Ulm, Germany; (S.B.); (B.B.); (C.R.)
- Central Facility for Electron Microscopy, Ulm University, 89081 Ulm, Germany;
| | - Tatjana Weil
- Institute of Molecular Virology, Ulm University Medical Center, 89081 Ulm, Germany; (T.W.); (J.M.)
| | - Paul Walther
- Central Facility for Electron Microscopy, Ulm University, 89081 Ulm, Germany;
| | - Thomas Schrader
- Faculty of Chemistry, University of Duisburg-Essen, 45117 Essen, Germany;
| | - Jan Münch
- Institute of Molecular Virology, Ulm University Medical Center, 89081 Ulm, Germany; (T.W.); (J.M.)
- Core Facility Functional Peptidomics, Ulm University Medical Center, 89081 Ulm, Germany
| | - Jens von Einem
- Institute of Virology, Ulm University Medical Center, 89081 Ulm, Germany; (S.B.); (B.B.); (C.R.)
- Correspondence: ; Tel.: +49-(0)731-500-65104; Fax: +49-(0)731-500-65102
| |
Collapse
|
25
|
Schütz D, Conzelmann C, Fois G, Groß R, Weil T, Wettstein L, Stenger S, Zelikin A, Hoffmann TK, Frick M, Müller JA, Münch J. Carrageenan-containing over-the-counter nasal and oral sprays inhibit SARS-CoV-2 infection of airway epithelial cultures. Am J Physiol Lung Cell Mol Physiol 2021; 320:L750-L756. [PMID: 33561380 PMCID: PMC8384564 DOI: 10.1152/ajplung.00552.2020] [Citation(s) in RCA: 35] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2020] [Revised: 02/02/2021] [Accepted: 02/04/2021] [Indexed: 02/06/2023] Open
Abstract
Pharmaceutical interventions are urgently needed to prevent severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) infection and transmission. As SARS-CoV-2 infects and spreads via the nasopharyngeal airways, we analyzed the antiviral effect of selected nasal and oral sprays on virus infection in vitro. Two nose sprays showed virucidal activity but were cytotoxic precluding further analysis in cell culture. One nasal and one mouth spray suppressed SARS-CoV-2 infection of TMPRSS2-expressing Vero E6 cells and primary differentiated human airway epithelial cultures. The antiviral activity in both sprays could be attributed to polyanionic ι- and κ-carrageenans. Thus, application of carrageenan-containing nasal and mouth sprays may reduce the risk of acquiring SARS-CoV-2 infection and may limit viral spread, warranting further clinical evaluation.
Collapse
Affiliation(s)
- Desiree Schütz
- Institute of Molecular Virology, Ulm University Medical Center, Ulm, Germany
| | - Carina Conzelmann
- Institute of Molecular Virology, Ulm University Medical Center, Ulm, Germany
| | - Giorgio Fois
- Institute of General Physiology, Ulm University, Ulm, Germany
| | - Rüdiger Groß
- Institute of Molecular Virology, Ulm University Medical Center, Ulm, Germany
| | - Tatjana Weil
- Institute of Molecular Virology, Ulm University Medical Center, Ulm, Germany
| | - Lukas Wettstein
- Institute of Molecular Virology, Ulm University Medical Center, Ulm, Germany
| | - Steffen Stenger
- Institute for Microbiology and Hygiene, Ulm University Medical Center, Ulm, Germany
| | - Alexander Zelikin
- Department of Chemistry and iNano Interdisciplinary Nanoscience Centre, Aarhus University, Aarhus, Denmark
| | - Thomas K Hoffmann
- Department of Otorhinolaryngology, Head and Neck Surgery, Ulm University, Ulm, Germany
| | - Manfred Frick
- Institute of General Physiology, Ulm University, Ulm, Germany
| | - Janis A Müller
- Institute of Molecular Virology, Ulm University Medical Center, Ulm, Germany
| | - Jan Münch
- Institute of Molecular Virology, Ulm University Medical Center, Ulm, Germany
| |
Collapse
|
26
|
Malishev R, Salinas N, Gibson J, Eden AB, Mieres-Perez J, Ruiz-Blanco YB, Malka O, Kolusheva S, Klärner FG, Schrader T, Sanchez-Garcia E, Wang C, Landau M, Bitan G, Jelinek R. Inhibition of Staphylococcus aureus biofilm-forming functional amyloid by molecular tweezers. Cell Chem Biol 2021; 28:1310-1320.e5. [PMID: 33852903 DOI: 10.1016/j.chembiol.2021.03.013] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2020] [Revised: 01/19/2021] [Accepted: 03/19/2021] [Indexed: 02/07/2023]
Abstract
Biofilms are rigid and largely impenetrable three-dimensional matrices constituting virulence determinants of various pathogenic bacteria. Here, we demonstrate that molecular tweezers, unique supramolecular artificial receptors, modulate biofilm formation of Staphylococcus aureus. In particular, the tweezers affect the structural and assembly properties of phenol-soluble modulin α1 (PSMα1), a biofilm-scaffolding functional amyloid peptide secreted by S. aureus. The data reveal that CLR01, a diphosphate tweezer, exhibits significant S. aureus biofilm inhibition and disrupts PSMα1 self-assembly and fibrillation, likely through inclusion of lysine side chains of the peptide. In comparison, different peptide binding occurs in the case of CLR05, a tweezer containing methylenecarboxylate units, which exhibits lower affinity for the lysine residues yet disrupts S. aureus biofilm more strongly than CLR01. Our study points to a possible role for molecular tweezers as potent biofilm inhibitors and antibacterial agents, particularly against untreatable biofilm-forming and PSM-producing bacteria, such as methicillin-resistant S. aureus.
Collapse
Affiliation(s)
- Ravit Malishev
- Department of Chemistry, Ben Gurion University of the Negev, Beer Sheva 84105, Israel
| | - Nir Salinas
- Department of Biology, Technion-Israel Institute of Technology, Haifa 3200003, Israel
| | - James Gibson
- Center for Biotechnology and Interdisciplinary Studies, Department of Biological Sciences, Rensselaer Polytechnic Institute, Troy, NY 12180, USA
| | - Angela Bailey Eden
- Center for Biotechnology and Interdisciplinary Studies, Department of Biological Sciences, Rensselaer Polytechnic Institute, Troy, NY 12180, USA
| | - Joel Mieres-Perez
- Department of Computational Biochemistry, University of Duisburg-Essen, Universitätsstrasse 2, 45141 Essen, Germany
| | - Yasser B Ruiz-Blanco
- Department of Computational Biochemistry, University of Duisburg-Essen, Universitätsstrasse 2, 45141 Essen, Germany
| | - Orit Malka
- Department of Chemistry, Ben Gurion University of the Negev, Beer Sheva 84105, Israel
| | - Sofiya Kolusheva
- Ilse Katz Institute for Nanotechnology, Ben Gurion University of the Negev, Beer Sheva 84105, Israel
| | | | - Thomas Schrader
- Faculty of Chemistry, University of Duisburg-Essen, Essen, Germany
| | - Elsa Sanchez-Garcia
- Department of Computational Biochemistry, University of Duisburg-Essen, Universitätsstrasse 2, 45141 Essen, Germany
| | - Chunyu Wang
- Center for Biotechnology and Interdisciplinary Studies, Department of Biological Sciences, Rensselaer Polytechnic Institute, Troy, NY 12180, USA
| | - Meytal Landau
- Department of Biology, Technion-Israel Institute of Technology, Haifa 3200003, Israel; European Molecular Biology Laboratory (EMBL), 22607 Hamburg, Germany
| | - Gal Bitan
- Department of Neurology, David Geffen School of Medicine, Brain Research Institute, and Molecular Biology Institute, University of California, Los Angeles, Los Angeles, CA, USA
| | - Raz Jelinek
- Department of Chemistry, Ben Gurion University of the Negev, Beer Sheva 84105, Israel; Ilse Katz Institute for Nanotechnology, Ben Gurion University of the Negev, Beer Sheva 84105, Israel.
| |
Collapse
|
27
|
Schütz D, Read C, Groß R, Röcker A, Rode S, Annamalai K, Fändrich M, Münch J. Negatively Charged Peptide Nanofibrils from Immunoglobulin Light Chain Sequester Viral Particles but Lack Cell-Binding and Viral Transduction-Enhancing Properties. ACS OMEGA 2021; 6:7731-7738. [PMID: 33778283 PMCID: PMC7992169 DOI: 10.1021/acsomega.1c00068] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/05/2021] [Accepted: 02/09/2021] [Indexed: 05/08/2023]
Abstract
Positively charged naturally occurring or engineered peptide nanofibrils (PNF) are effective enhancers of lentiviral and retroviral transduction, an often rate-limiting step in gene transfer and gene therapy approaches. These polycationic PNF are thought to bridge the electrostatic repulsions between negatively charged membranes of virions and cells, thereby enhancing virion attachment to and infection of target cells. Here, we analyzed PNF, which are formed by the peptide AL1, that represents a fragment of an immunoglobulin light chain that causes systemic AL amyloidosis. We found that negatively charged AL1 PNF interact with viral particles to a comparable extent as positively charged PNF. However, AL1 PNF lacked cell-binding activity, and consequently, did not enhance retroviral infection. These findings show that virion capture and cell binding of PNF are mediated by different mechanisms, offering avenues for the design of advanced PNF with selective functions.
Collapse
Affiliation(s)
- Desiree Schütz
- Institute
of Molecular Virology, Ulm University Medical
Center, 89081 Ulm, Germany
| | - Clarissa Read
- Central
Facility for Electron Microscopy, Ulm University, 89081 Ulm, Germany
| | - Rüdiger Groß
- Institute
of Molecular Virology, Ulm University Medical
Center, 89081 Ulm, Germany
| | - Annika Röcker
- Institute
of Molecular Virology, Ulm University Medical
Center, 89081 Ulm, Germany
| | - Sascha Rode
- Institute
of Molecular Virology, Ulm University Medical
Center, 89081 Ulm, Germany
| | | | - Marcus Fändrich
- Institute
of Protein Biochemistry, Ulm University, 89081 Ulm, Germany
| | - Jan Münch
- Institute
of Molecular Virology, Ulm University Medical
Center, 89081 Ulm, Germany
- Core
Facility Functional Peptidomics, Ulm University
Medical Center, 89081 Ulm, Germany
- . Phone: +49 731 500 65154
| |
Collapse
|
28
|
Bolaamphiphile-based supramolecular gels with drugs eliciting membrane effects. J Colloid Interface Sci 2021; 594:857-863. [PMID: 33794407 DOI: 10.1016/j.jcis.2021.03.026] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2021] [Revised: 03/02/2021] [Accepted: 03/06/2021] [Indexed: 12/11/2022]
Abstract
Supramolecular chemistry has garnered important interest in recent years toward improving therapeutic efficacy via drug delivery approaches. Although self-assemblies have been deeply investigated, the design of novel drugs leveraging supramolecular chemistry is less known. In this contribution, we show that a Low Molecular Weight Gel (LMWG) can elicit cancer cell apoptosis. This biological effect results from the unique supramolecular properties of a bolaamphiphile-based gelator, which allow for strong interaction with the lipid membrane. This novel supramolecular-drug paradigm opens up new possibilities for therapeutic applications targeting membrane lipids.
Collapse
|
29
|
Linet A, Joseph MM, Haritha M, Shamna K, Varughese S, Devi PS, Suresh CH, Maiti KK, Neogi I. De novo design and synthesis of boomerang-shaped molecules and their in silico and SERS-based interactions with SARS-CoV-2 spike protein and ACE2. NEW J CHEM 2021. [DOI: 10.1039/d1nj02955j] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022]
Abstract
Boomerang shaped molecule and its interaction study with SARS-CoV-2 S-protein–ACE2 using molecular docking, SERS and UV-Vis spectroscopy.
Collapse
Affiliation(s)
- Amrutham Linet
- CSIR-National Institute for Interdisciplinary Sciences and Technology (CSIR-NIIST), Thiruvananthapuram, 695019, India
- Academy of Scientific and Innovative Research (AcSIR), Ghaziabad, 201002, India
| | - Manu M. Joseph
- CSIR-National Institute for Interdisciplinary Sciences and Technology (CSIR-NIIST), Thiruvananthapuram, 695019, India
| | - Mambatta Haritha
- CSIR-National Institute for Interdisciplinary Sciences and Technology (CSIR-NIIST), Thiruvananthapuram, 695019, India
- Academy of Scientific and Innovative Research (AcSIR), Ghaziabad, 201002, India
| | - K. Shamna
- CSIR-National Institute for Interdisciplinary Sciences and Technology (CSIR-NIIST), Thiruvananthapuram, 695019, India
| | - Sunil Varughese
- CSIR-National Institute for Interdisciplinary Sciences and Technology (CSIR-NIIST), Thiruvananthapuram, 695019, India
- Academy of Scientific and Innovative Research (AcSIR), Ghaziabad, 201002, India
| | - P. Sujatha Devi
- CSIR-National Institute for Interdisciplinary Sciences and Technology (CSIR-NIIST), Thiruvananthapuram, 695019, India
- Academy of Scientific and Innovative Research (AcSIR), Ghaziabad, 201002, India
| | - C. H. Suresh
- CSIR-National Institute for Interdisciplinary Sciences and Technology (CSIR-NIIST), Thiruvananthapuram, 695019, India
- Academy of Scientific and Innovative Research (AcSIR), Ghaziabad, 201002, India
| | - Kaustabh Kumar Maiti
- CSIR-National Institute for Interdisciplinary Sciences and Technology (CSIR-NIIST), Thiruvananthapuram, 695019, India
- Academy of Scientific and Innovative Research (AcSIR), Ghaziabad, 201002, India
| | - Ishita Neogi
- CSIR-National Institute for Interdisciplinary Sciences and Technology (CSIR-NIIST), Thiruvananthapuram, 695019, India
- Academy of Scientific and Innovative Research (AcSIR), Ghaziabad, 201002, India
| |
Collapse
|
30
|
Selective Recognition of Amino Acids and Peptides by Small Supramolecular Receptors. Molecules 2020; 26:molecules26010106. [PMID: 33379401 PMCID: PMC7796322 DOI: 10.3390/molecules26010106] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2020] [Revised: 12/14/2020] [Accepted: 12/22/2020] [Indexed: 12/30/2022] Open
Abstract
To this day, the recognition and high affinity binding of biomolecules in water by synthetic receptors remains challenging, while the necessity for systems for their sensing, transport and modulation persists. This problematic is prevalent for the recognition of peptides, which not only have key roles in many biochemical pathways, as well as having pharmacological and biotechnological applications, but also frequently serve as models for the study of proteins. Taking inspiration in nature and on the interactions that occur between several receptors and peptide sequences, many researchers have developed and applied a variety of different synthetic receptors, as is the case of macrocyclic compounds, molecular imprinted polymers, organometallic cages, among others, to bind amino acids, small peptides and proteins. In this critical review, we present and discuss selected examples of synthetic receptors for amino acids and peptides, with a greater focus on supramolecular receptors, which show great promise for the selective recognition of these biomolecules in physiological conditions. We decided to focus preferentially on small synthetic receptors (leaving out of this review high molecular weight polymeric systems) for which more detailed and accurate molecular level information regarding the main structural and thermodynamic features of the receptor biomolecule assemblies is available.
Collapse
|