1
|
Dong X, Qiu Y, Jia N, Wu Y, Nie Q, Wen J, Zhao C, Zhai Y. Recent advances of edible marine algae-derived sulfated polysaccharides in antiviral treatments: challenges vs. opportunities. Front Nutr 2025; 12:1561119. [PMID: 40206958 PMCID: PMC11978671 DOI: 10.3389/fnut.2025.1561119] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2025] [Accepted: 03/12/2025] [Indexed: 04/11/2025] Open
Abstract
Marine polysaccharides, particularly those derived from red, brown, and green algae, have shown promising antiviral activity. Among them, sulfated polysaccharides are particularly notable due to their broad-spectrum antiviral properties. These include direct viral destruction, inhibition of virus adsorption, disruption of viral transcription and replication, and the stimulation of the host's antiviral immunity. With low toxicity, minimal drug resistance, and excellent biocompatibility, these polysaccharides represent promising candidates for the development of antiviral medications. For instance, carrageenan, a polysaccharide from red algae, and fucoidan, a polymer from brown algae, have both been proven to effectively inhibit viral infections. Sulfated polysaccharides from green algae, such as those found in Ulva species, also exhibit antiviral properties, including activity against the Japanese encephalitis virus. These polysaccharides function by blocking the attachment of viruses to host cells or interfering with various stages of the viral life cycle. Moreover, marine polysaccharides have been shown to enhance host immune responses, thereby aiding in viral clearance. Although these findings highlight the antiviral potential of marine polysaccharides, most studies have been conducted in vitro or in animal models. Further clinical trials are necessary to validate their effectiveness and safety for therapeutic use.
Collapse
Affiliation(s)
- Xiaoying Dong
- Department of Infectious Disease, Shengjing Hospital of China Medical University, Shenyang, China
| | - Yusong Qiu
- State Key Laboratory of Mariculture Breeding, Key Laboratory of Marine Biotechnology of Fujian Province, Fujian Agriculture and Forestry University, Fuzhou, China
| | - Nan Jia
- State Key Laboratory of Mariculture Breeding, Key Laboratory of Marine Biotechnology of Fujian Province, Fujian Agriculture and Forestry University, Fuzhou, China
- College of Marine Sciences, Fujian Agriculture and Forestry University, Fuzhou, China
| | - Yinfeng Wu
- State Key Laboratory of Mariculture Breeding, Key Laboratory of Marine Biotechnology of Fujian Province, Fujian Agriculture and Forestry University, Fuzhou, China
| | - Qing Nie
- College of Marine and Biology Engineering, Yancheng Institute of Technology, Yancheng, China
| | - Jiahui Wen
- State Key Laboratory of Mariculture Breeding, Key Laboratory of Marine Biotechnology of Fujian Province, Fujian Agriculture and Forestry University, Fuzhou, China
- College of Marine Sciences, Fujian Agriculture and Forestry University, Fuzhou, China
| | - Chao Zhao
- State Key Laboratory of Mariculture Breeding, Key Laboratory of Marine Biotechnology of Fujian Province, Fujian Agriculture and Forestry University, Fuzhou, China
- College of Marine Sciences, Fujian Agriculture and Forestry University, Fuzhou, China
| | - Yongzhen Zhai
- Department of Infectious Disease, Shengjing Hospital of China Medical University, Shenyang, China
| |
Collapse
|
2
|
Wang TT, Hirons A, Doerflinger M, Morris KV, Ledger S, Purcell DFJ, Kelleher AD, Ahlenstiel CL. Current State of Therapeutics for HTLV-1. Viruses 2024; 16:1616. [PMID: 39459949 PMCID: PMC11512412 DOI: 10.3390/v16101616] [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: 09/02/2024] [Revised: 10/07/2024] [Accepted: 10/11/2024] [Indexed: 10/28/2024] Open
Abstract
Human T cell leukaemia virus type-1 (HTLV-1) is an oncogenic retrovirus that causes lifelong infection in ~5-10 million individuals globally. It is endemic to certain First Nations populations of Northern and Central Australia, Japan, South and Central America, Africa, and the Caribbean region. HTLV-1 preferentially infects CD4+ T cells and remains in a state of reduced transcription, often being asymptomatic in the beginning of infection, with symptoms developing later in life. HTLV-1 infection is implicated in the development of adult T cell leukaemia/lymphoma (ATL) and HTLV-1-associated myelopathies (HAM), amongst other immune-related disorders. With no preventive or curative interventions, infected individuals have limited treatment options, most of which manage symptoms. The clinical burden and lack of treatment options directs the need for alternative treatment strategies for HTLV-1 infection. Recent advances have been made in the development of RNA-based antiviral therapeutics for Human Immunodeficiency Virus Type-1 (HIV-1), an analogous retrovirus that shares modes of transmission with HTLV-1. This review highlights past and ongoing efforts in the development of HTLV-1 therapeutics and vaccines, with a focus on the potential for gene therapy as a new treatment modality in light of its successes in HIV-1, as well as animal models that may help the advancement of novel antiviral and anticancer interventions.
Collapse
Affiliation(s)
- Tiana T. Wang
- Kirby Institute, University of New South Wales, Sydney, NSW 2052, Australia; (T.T.W.); (S.L.); (A.D.K.)
| | - Ashley Hirons
- The Peter Doherty Institute for Infection and Immunity, University of Melbourne, Melbourne, VIC 3052, Australia; (A.H.); (D.F.J.P.)
| | - Marcel Doerflinger
- Walter and Eliza Hall Institute of Medical Research, Parkville, VIC 3052, Australia;
- Department of Medical Biology, University of Melbourne, Melbourne, VIC 3050, Australia
| | - Kevin V. Morris
- Centre for Genomics and Personalised Health, School of Biomedical Sciences, Queensland University of Technology, Kelvin Grove, QLD 4059, Australia;
| | - Scott Ledger
- Kirby Institute, University of New South Wales, Sydney, NSW 2052, Australia; (T.T.W.); (S.L.); (A.D.K.)
| | - Damian F. J. Purcell
- The Peter Doherty Institute for Infection and Immunity, University of Melbourne, Melbourne, VIC 3052, Australia; (A.H.); (D.F.J.P.)
| | - Anthony D. Kelleher
- Kirby Institute, University of New South Wales, Sydney, NSW 2052, Australia; (T.T.W.); (S.L.); (A.D.K.)
- UNSW RNA Institute, University of New South Wales, Sydney, NSW 2052, Australia
| | - Chantelle L. Ahlenstiel
- Kirby Institute, University of New South Wales, Sydney, NSW 2052, Australia; (T.T.W.); (S.L.); (A.D.K.)
- UNSW RNA Institute, University of New South Wales, Sydney, NSW 2052, Australia
| |
Collapse
|
3
|
Fantini J, Azzaz F, Chahinian H, Yahi N. Electrostatic Surface Potential as a Key Parameter in Virus Transmission and Evolution: How to Manage Future Virus Pandemics in the Post-COVID-19 Era. Viruses 2023; 15:284. [PMID: 36851498 PMCID: PMC9964723 DOI: 10.3390/v15020284] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/26/2022] [Revised: 01/14/2023] [Accepted: 01/18/2023] [Indexed: 01/20/2023] Open
Abstract
Virus-cell interactions involve fundamental parameters that need to be considered in strategies implemented to control viral outbreaks. Among these, the surface electrostatic potential can give valuable information to deal with new epidemics. In this article, we describe the role of this key parameter in the hemagglutination of red blood cells and in the co-evolution of synaptic receptors and neurotransmitters. We then establish the functional link between lipid rafts and the electrostatic potential of viruses, with special emphasis on gangliosides, which are sialic-acid-containing, electronegatively charged plasma membrane components. We describe the common features of ganglioside binding domains, which include a wide variety of structures with little sequence homology but that possess key amino acids controlling ganglioside recognition. We analyze the role of the electrostatic potential in the transmission and intra-individual evolution of HIV-1 infections, including gatekeeper and co-receptor switch mechanisms. We show how to organize the epidemic surveillance of influenza viruses by focusing on mutations affecting the hemagglutinin surface potential. We demonstrate that the electrostatic surface potential, by modulating spike-ganglioside interactions, controls the hemagglutination properties of coronaviruses (SARS-CoV-1, MERS-CoV, and SARS-CoV-2) as well as the structural dynamics of SARS-CoV-2 evolution. We relate the broad-spectrum antiviral activity of repositioned molecules to their ability to disrupt virus-raft interactions, challenging the old concept that an antibiotic or anti-parasitic cannot also be an antiviral. We propose a new concept based on the analysis of the electrostatic surface potential to develop, in real time, therapeutic and vaccine strategies adapted to each new viral epidemic.
Collapse
Affiliation(s)
- Jacques Fantini
- Department of Biology, Faculty of Medicine, University of Aix-Marseille, INSERM UMR_S 1072, 13015 Marseille, France
| | | | | | | |
Collapse
|
4
|
Fröba M, Große M, Setz C, Rauch P, Auth J, Spanaus L, Münch J, Ruetalo N, Schindler M, Morokutti-Kurz M, Graf P, Prieschl-Grassauer E, Grassauer A, Schubert U. Iota-Carrageenan Inhibits Replication of SARS-CoV-2 and the Respective Variants of Concern Alpha, Beta, Gamma and Delta. Int J Mol Sci 2021; 22:ijms222413202. [PMID: 34947999 PMCID: PMC8709357 DOI: 10.3390/ijms222413202] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2021] [Revised: 12/01/2021] [Accepted: 12/03/2021] [Indexed: 12/15/2022] Open
Abstract
The COVID-19 pandemic continues to spread around the world and remains a major public health threat. Vaccine inefficiency, vaccination breakthroughs and lack of supply, especially in developing countries, as well as the fact that a non-negligible part of the population either refuse vaccination or cannot be vaccinated due to age, pre-existing illness or non-response to existing vaccines intensify this issue. This might also contribute to the emergence of new variants, being more efficiently transmitted, more virulent and more capable of escaping naturally acquired and vaccine-induced immunity. Hence, the need of effective and viable prevention options to reduce viral transmission is of outmost importance. In this study, we investigated the antiviral effect of iota-, lambda- and kappa-carrageenan, sulfated polysaccharides extracted from red seaweed, on SARS-CoV-2 Wuhan type and the spreading variants of concern (VOCs) Alpha, Beta, Gamma and Delta. Carrageenans as part of broadly used nasal and mouth sprays as well as lozenges have the potential of first line defense to inhibit the infection and transmission of SARS-CoV-2. Here, we demonstrate by using a SARS-CoV-2 spike pseudotyped lentivirus particles (SSPL) system and patient-isolated SARS-CoV-2 VOCs to infect transgenic A549ACE2/TMPRSS2 and Calu-3 human lung cells that all three carrageenan types exert antiviral activity. Iota-carrageenan exhibits antiviral activity with comparable IC50 values against the SARS-CoV-2 Wuhan type and the VOCs. Altogether, these results indicate that iota-carrageenan might be effective for prophylaxis and treatment of SARS-CoV-2 infections independent of the present and potentially future variants.
Collapse
Affiliation(s)
- Maria Fröba
- Institute of Virology, Friedrich-Alexander University Erlangen-Nürnberg (FAU), 91054 Erlangen, Germany; (M.F.); (M.G.); (C.S.); (P.R.); (J.A.); (L.S.)
| | - Maximilian Große
- Institute of Virology, Friedrich-Alexander University Erlangen-Nürnberg (FAU), 91054 Erlangen, Germany; (M.F.); (M.G.); (C.S.); (P.R.); (J.A.); (L.S.)
| | - Christian Setz
- Institute of Virology, Friedrich-Alexander University Erlangen-Nürnberg (FAU), 91054 Erlangen, Germany; (M.F.); (M.G.); (C.S.); (P.R.); (J.A.); (L.S.)
| | - Pia Rauch
- Institute of Virology, Friedrich-Alexander University Erlangen-Nürnberg (FAU), 91054 Erlangen, Germany; (M.F.); (M.G.); (C.S.); (P.R.); (J.A.); (L.S.)
| | - Janina Auth
- Institute of Virology, Friedrich-Alexander University Erlangen-Nürnberg (FAU), 91054 Erlangen, Germany; (M.F.); (M.G.); (C.S.); (P.R.); (J.A.); (L.S.)
| | - Lucas Spanaus
- Institute of Virology, Friedrich-Alexander University Erlangen-Nürnberg (FAU), 91054 Erlangen, Germany; (M.F.); (M.G.); (C.S.); (P.R.); (J.A.); (L.S.)
| | - Jan Münch
- Institute of Molecular Virology, Ulm University Medical Center, 89081 Ulm, Germany;
| | - Natalia Ruetalo
- Institute for Medical Virology and Epidemiology of Viral Diseases, University Hospital Tübingen, 72076 Tübingen, Germany; (N.R.); (M.S.)
| | - Michael Schindler
- Institute for Medical Virology and Epidemiology of Viral Diseases, University Hospital Tübingen, 72076 Tübingen, Germany; (N.R.); (M.S.)
| | | | - Philipp Graf
- Marinomed Biotech AG, A-2100 Korneuburg, Austria; (M.M.-K.); (P.G.); (E.P.-G.); (A.G.)
| | | | - Andreas Grassauer
- Marinomed Biotech AG, A-2100 Korneuburg, Austria; (M.M.-K.); (P.G.); (E.P.-G.); (A.G.)
| | - Ulrich Schubert
- Institute of Virology, Friedrich-Alexander University Erlangen-Nürnberg (FAU), 91054 Erlangen, Germany; (M.F.); (M.G.); (C.S.); (P.R.); (J.A.); (L.S.)
- Correspondence: ; Tel.: +49-9131-85-26478
| |
Collapse
|
5
|
The challenge of structural heterogeneity in the native mass spectrometry studies of the SARS-CoV-2 spike protein interactions with its host cell-surface receptor. Anal Bioanal Chem 2021; 413:7205-7214. [PMID: 34389878 PMCID: PMC8362873 DOI: 10.1007/s00216-021-03601-3] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2021] [Revised: 07/28/2021] [Accepted: 08/04/2021] [Indexed: 11/13/2022]
Abstract
Native mass spectrometry (MS) enjoyed tremendous success in the past two decades in a wide range of studies aiming at understanding the molecular mechanisms of physiological processes underlying a variety of pathologies and accelerating the drug discovery process. However, the success record of native MS has been surprisingly modest with respect to the most recent challenge facing the biomedical community—the novel coronavirus infection (COVID-19). The major reason for the paucity of successful studies that use native MS to target various aspects of SARS-CoV-2 interaction with its host is the extreme degree of heterogeneity of the viral protein playing a key role in the host cell invasion. Indeed, the SARS-CoV-2 spike protein (S-protein) is extensively glycosylated, presenting a formidable challenge for native MS as a means of characterizing its interactions with both the host cell–surface receptor ACE2 and the drug candidates capable of disrupting this interaction. In this work, we evaluate the utility of native MS complemented with the experimental methods using gas-phase chemistry (limited charge reduction) to obtain meaningful information on the association of the S1 domain of the S-protein with the ACE2 ectodomain, and the influence of a small synthetic heparinoid on this interaction. Native MS reveals the presence of several different S1 oligomers in solution and allows the stoichiometry of the most prominent S1/ACE2 complexes to be determined. This enables meaningful interpretation of the changes in native MS that are observed upon addition of a small synthetic heparinoid (the pentasaccharide fondaparinux) to the S1/ACE2 solution, confirming that the small polyanion destabilizes the protein/receptor binding.
Collapse
|
6
|
Yang Y, Ivanov DG, Kaltashov IA. The challenge of structural heterogeneity in the native mass spectrometry studies of the SARS-CoV-2 spike protein interactions with its host cell-surface receptor. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2021. [PMID: 34189525 DOI: 10.1101/2021.06.20.449191] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/24/2022]
Abstract
Native mass spectrometry (MS) enjoyed tremendous success in the past two decades in a wide range of studies aiming at understanding the molecular mechanisms of physiological processes underlying a variety of pathologies and accelerating the drug discovery process. However, the success record of native MS has been surprisingly modest with respect to the most recent challenge facing the biomedical community â€" the novel coronavirus infection (COVID-19). The major reason for the paucity of successful studies that use native MS to target various aspects of SARS-CoV-2 interaction with its host is the extreme degree of structural heterogeneity of the viral protein playing a key role in the host cell invasion. Indeed, the SARS-CoV-2 spike protein (S-protein) is extensively glycosylated, presenting a formidable challenge for native mass spectrometry (MS) as a means of characterizing its interactions with both the host cell-surface receptor ACE2 and the drug candidates capable of disrupting this interaction. In this work we evaluate the utility of native MS complemented with the experimental methods using gas-phase chemistry (limited charge reduction) to obtain meaningful information on the association of the S1 domain of the S-protein with the ACE2 ectodomain, and the influence of a small synthetic heparinoid on this interaction. Native MS reveals the presence of several different S1 oligomers in solution and allows the stoichiometry of the most prominent S1/ACE2 complexes to be determined. This enables meaningful interpretation of the changes in native MS that are observed upon addition of a small synthetic heparinoid (the pentasaccharide fondaparinux) to the S1/ACE2 solution, confirming that the small polyanion destabilizes the protein/receptor binding.
Collapse
|
7
|
Mathez G, Cagno V. Viruses Like Sugars: How to Assess Glycan Involvement in Viral Attachment. Microorganisms 2021; 9:1238. [PMID: 34200288 PMCID: PMC8230229 DOI: 10.3390/microorganisms9061238] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2021] [Revised: 06/01/2021] [Accepted: 06/04/2021] [Indexed: 12/12/2022] Open
Abstract
The first step of viral infection requires interaction with the host cell. Before finding the specific receptor that triggers entry, the majority of viruses interact with the glycocalyx. Identifying the carbohydrates that are specifically recognized by different viruses is important both for assessing the cellular tropism and for identifying new antiviral targets. Advances in the tools available for studying glycan-protein interactions have made it possible to identify them more rapidly; however, it is important to recognize the limitations of these methods in order to draw relevant conclusions. Here, we review different techniques: genetic screening, glycan arrays, enzymatic and pharmacological approaches, and surface plasmon resonance. We then detail the glycan interactions of enterovirus D68 and severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2), highlighting the aspects that need further clarification.
Collapse
Affiliation(s)
| | - Valeria Cagno
- Institute of Microbiology, Lausanne University Hospital, University of Lausanne, 1011 Lausanne, Switzerland;
| |
Collapse
|
8
|
Wells L, Vierra C, Hardman J, Han Y, Dimas D, Gwarada‐Phillips LN, Blackeye R, Eggers DK, LaBranche CC, Král P, McReynolds KD. Sulfoglycodendrimer Therapeutics for HIV-1 and SARS-CoV-2. ADVANCED THERAPEUTICS 2021; 4:2000210. [PMID: 33786368 PMCID: PMC7995185 DOI: 10.1002/adtp.202000210] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2020] [Revised: 01/04/2021] [Indexed: 12/05/2022]
Abstract
Hexavalent sulfoglycodendrimers (SGDs) are synthesized as mimics of host cell heparan sulfate proteoglycans (HSPGs) to inhibit the early stages in viral binding/entry of HIV-1 and SARS-CoV-2. Using an HIV neutralization assay, the most promising of the seven candidates are found to have sub-micromolar anti-HIV activities. Molecular dynamics simulations are separately implemented to investigate how/where the SGDs interacted with both pathogens. The simulations revealed that the SGDs: 1) develop multivalent binding with polybasic regions within and outside of the V3 loop on glycoprotein 120 (gp120) for HIV-1, and consecutively bind with multiple gp120 subunits, and 2) interact with basic amino acids in both the angiotensin-converting enzyme 2 (ACE2) and HSPG binding regions of the Receptor Binding Domain (RBD) from SARS-CoV-2. These results illustrate the considerable potential of SGDs as inhibitors in viral binding/entry of both HIV-1 and SARS-CoV-2 pathogens, leading the way for further development of this class of molecules as broad-spectrum antiviral agents.
Collapse
Affiliation(s)
- Lauren Wells
- Department of ChemistryCalifornia State UniversitySacramento, 6000 J StreetSacramentoCA95819–6057USA
| | - Cory Vierra
- Department of ChemistryCalifornia State UniversitySacramento, 6000 J StreetSacramentoCA95819–6057USA
| | - Janee’ Hardman
- Department of ChemistryCalifornia State UniversitySacramento, 6000 J StreetSacramentoCA95819–6057USA
| | - Yanxiao Han
- Department of ChemistryUniversity of IllinoisChicago 845 W. Taylor St.ChicagoIL60607USA
| | - Dustin Dimas
- Department of ChemistryCalifornia State UniversitySacramento, 6000 J StreetSacramentoCA95819–6057USA
| | - Lucia N. Gwarada‐Phillips
- Department of ChemistryCalifornia State UniversitySacramento, 6000 J StreetSacramentoCA95819–6057USA
| | - Rachel Blackeye
- Department of ChemistryCalifornia State UniversitySacramento, 6000 J StreetSacramentoCA95819–6057USA
| | - Daryl K. Eggers
- Department of ChemistrySan José State UniversityOne Washington SquareSan JoséCA95192USA
| | | | - Petr Král
- Department of ChemistryUniversity of IllinoisChicago 845 W. Taylor St.ChicagoIL60607USA
- Departments of Physics, Pharmaceutical Sciences, and Chemical EngineeringUniversity of IllinoisChicago 845 W. Taylor St.ChicagoIL60607USA
| | - Katherine D. McReynolds
- Department of ChemistryCalifornia State UniversitySacramento, 6000 J StreetSacramentoCA95819–6057USA
| |
Collapse
|
9
|
Antiviral activity of lambda-carrageenan against influenza viruses and severe acute respiratory syndrome coronavirus 2. Sci Rep 2021; 11:821. [PMID: 33436985 PMCID: PMC7804421 DOI: 10.1038/s41598-020-80896-9] [Citation(s) in RCA: 56] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2020] [Accepted: 12/30/2020] [Indexed: 01/04/2023] Open
Abstract
Influenza virus and coronavirus, belonging to enveloped RNA viruses, are major causes of human respiratory diseases. The aim of this study was to investigate the broad spectrum antiviral activity of a naturally existing sulfated polysaccharide, lambda-carrageenan (λ-CGN), purified from marine red algae. Cell culture-based assays revealed that the macromolecule efficiently inhibited both influenza A and B viruses with EC50 values ranging from 0.3 to 1.4 μg/ml, as well as currently circulating severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) with an EC50 value of 0.9 ± 1.1 μg/ml. No toxicity to the host cells was observed at concentrations up to 300 μg/ml. Plaque titration and western blot analysis verified that λ-CGN reduced expression of viral proteins in cell lysates and suppressed progeny virus production in culture supernatants in a dose-dependent manner. This polyanionic compound exerts antiviral activity by targeting viral attachment to cell surface receptors and preventing virus entry. Moreover, its intranasal administration to mice during influenza A viral challenge not only alleviated infection-mediated reductions in body weight but also protected 60% of mice from virus-induced mortality. Thus, λ-CGN could be a promising antiviral agent for preventing infection with several respiratory viruses.
Collapse
|
10
|
Chitray M, Kotecha A, Nsamba P, Ren J, Maree S, Ramulongo T, Paul G, Theron J, Fry EE, Stuart DI, Maree FF. Symmetrical arrangement of positively charged residues around the 5-fold axes of SAT type foot-and-mouth disease virus enhances cell culture of field viruses. PLoS Pathog 2020; 16:e1008828. [PMID: 32991636 PMCID: PMC7577442 DOI: 10.1371/journal.ppat.1008828] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2020] [Revised: 10/21/2020] [Accepted: 07/22/2020] [Indexed: 11/18/2022] Open
Abstract
Field isolates of foot-and-mouth disease viruses (FMDVs) utilize integrin-mediated cell entry but many, including Southern African Territories (SAT) viruses, are difficult to adapt to BHK-21 cells, thus hampering large-scale propagation of vaccine antigen. However, FMDVs acquire the ability to bind to cell surface heparan sulphate proteoglycans, following serial cytolytic infections in cell culture, likely by the selection of rapidly replicating FMDV variants. In this study, fourteen SAT1 and SAT2 viruses, serially passaged in BHK-21 cells, were virulent in CHO-K1 cells and displayed enhanced affinity for heparan, as opposed to their low-passage counterparts. Comparative sequence analysis revealed the fixation of positively charged residues clustered close to the icosahedral 5-fold axes of the virus, at amino acid positions 83-85 in the βD-βE loop and 110-112 in the βF-βG loop of VP1 upon adaptation to cultured cells. Molecular docking simulations confirmed enhanced binding of heparan sulphate to a model of the adapted SAT1 virus, with the region around VP1 arginine 112 contributing the most to binding. Using this information, eight chimeric field strain mutant viruses were constructed with additional positive charges in repeated clusters on the virion surface. Five of these bound heparan sulphate with expanded cell tropism, which should facilitate large-scale propagation. However, only positively charged residues at position 110-112 of VP1 enhanced infectivity of BHK-21 cells. The symmetrical arrangement of even a single amino acid residue in the FMD virion is a powerful strategy enabling the virus to generate novel receptor binding and alternative host-cell interactions.
Collapse
Affiliation(s)
- Melanie Chitray
- Vaccine and Diagnostic Development Programme, Onderstepoort Veterinary Institute, Agricultural Research Council, Onderstepoort, South Africa
- Department of Biochemistry, Genetics and Microbiology, Faculty of Natural and Agricultural Sciences, University of Pretoria, Pretoria, South Africa
| | - Abhay Kotecha
- Division of Structural Biology, The Henry Wellcome Building for Genomic Medicine, Roosevelt Drive, Headington, Oxford, United Kingdom
| | - Peninah Nsamba
- Vaccine and Diagnostic Development Programme, Onderstepoort Veterinary Institute, Agricultural Research Council, Onderstepoort, South Africa
- Department of Veterinary Tropical Diseases, Faculty of Veterinary Sciences, University of Pretoria, Onderstepoort, South Africa
- Makerere University, College of Veterinary Medicine, Animal Resources and Biosecurity, Kampala, Uganda
| | - Jingshan Ren
- Division of Structural Biology, The Henry Wellcome Building for Genomic Medicine, Roosevelt Drive, Headington, Oxford, United Kingdom
| | - Sonja Maree
- Vaccine and Diagnostic Development Programme, Onderstepoort Veterinary Institute, Agricultural Research Council, Onderstepoort, South Africa
| | - Tovhowani Ramulongo
- Vaccine and Diagnostic Development Programme, Onderstepoort Veterinary Institute, Agricultural Research Council, Onderstepoort, South Africa
- Department of Biochemistry, Genetics and Microbiology, Faculty of Natural and Agricultural Sciences, University of Pretoria, Pretoria, South Africa
| | | | - Jacques Theron
- Department of Biochemistry, Genetics and Microbiology, Faculty of Natural and Agricultural Sciences, University of Pretoria, Pretoria, South Africa
| | - Elizabeth E. Fry
- Division of Structural Biology, The Henry Wellcome Building for Genomic Medicine, Roosevelt Drive, Headington, Oxford, United Kingdom
| | - David I. Stuart
- Division of Structural Biology, The Henry Wellcome Building for Genomic Medicine, Roosevelt Drive, Headington, Oxford, United Kingdom
| | - Francois F. Maree
- Vaccine and Diagnostic Development Programme, Onderstepoort Veterinary Institute, Agricultural Research Council, Onderstepoort, South Africa
- Department of Biochemistry, Genetics and Microbiology, Faculty of Natural and Agricultural Sciences, University of Pretoria, Pretoria, South Africa
| |
Collapse
|
11
|
van Haren FMP, Page C, Laffey JG, Artigas A, Camprubi-Rimblas M, Nunes Q, Smith R, Shute J, Carroll M, Tree J, Carroll M, Singh D, Wilkinson T, Dixon B. Nebulised heparin as a treatment for COVID-19: scientific rationale and a call for randomised evidence. Crit Care 2020; 24:454. [PMID: 32698853 PMCID: PMC7374660 DOI: 10.1186/s13054-020-03148-2] [Citation(s) in RCA: 74] [Impact Index Per Article: 14.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2020] [Accepted: 07/03/2020] [Indexed: 12/16/2022] Open
Abstract
Nebulised unfractionated heparin (UFH) has a strong scientific and biological rationale and warrants urgent investigation of its therapeutic potential, for COVID-19-induced acute respiratory distress syndrome (ARDS). COVID-19 ARDS displays the typical features of diffuse alveolar damage with extensive pulmonary coagulation activation resulting in fibrin deposition in the microvasculature and formation of hyaline membranes in the air sacs. Patients infected with SARS-CoV-2 who manifest severe disease have high levels of inflammatory cytokines in plasma and bronchoalveolar lavage fluid and significant coagulopathy. There is a strong association between the extent of the coagulopathy and poor clinical outcomes.The anti-coagulant actions of nebulised UFH limit fibrin deposition and microvascular thrombosis. Trials in patients with acute lung injury and related conditions found inhaled UFH reduced pulmonary dead space, coagulation activation, microvascular thrombosis and clinical deterioration, resulting in increased time free of ventilatory support. In addition, UFH has anti-inflammatory, mucolytic and anti-viral properties and, specifically, has been shown to inactivate the SARS-CoV-2 virus and prevent its entry into mammalian cells, thereby inhibiting pulmonary infection by SARS-CoV-2. Furthermore, clinical studies have shown that inhaled UFH safely improves outcomes in other inflammatory respiratory diseases and also acts as an effective mucolytic in sputum-producing respiratory patients. UFH is widely available and inexpensive, which may make this treatment also accessible for low- and middle-income countries.These potentially important therapeutic properties of nebulised UFH underline the need for expedited large-scale clinical trials to test its potential to reduce mortality in COVID-19 patients.
Collapse
Affiliation(s)
- Frank M P van Haren
- Australian National University, Medical School, Canberra, Australia.
- Intensive Care Unit, the Canberra Hospital, Canberra, Australia.
| | - Clive Page
- Sackler Institute of Pulmonary Pharmacology, King's College London, London, UK
| | - John G Laffey
- Anaesthesia and Intensive Care Medicine, School of Medicine, and Regenerative Medicine Institute (REMEDI) at CÚRAM Centre for Research in Medical Devices, Biomedical Sciences Building, National University of Ireland Galway, Galway, Ireland
- Department of Anaesthesia, University Hospital Galway, Saolta Hospital Group, Galway, Ireland
| | - Antonio Artigas
- Critical Center, Corporació Sanitaria Parc Tauli , CIBER Enfermedades Respiratorias, Autonomous University of Barcelona, Sabadell, Spain
| | - Marta Camprubi-Rimblas
- Institut d'Investigació I Innovació Parc Tauli (I3PT), CIBER de Enfermedades Respiratorias, Sabadell, Spain
| | - Quentin Nunes
- Institute of Systems, Molecular and Integrative Biology, University of Liverpool, Liverpool, UK
| | - Roger Smith
- Department of Critical Care Medicine, St Vincent's Hospital, Melbourne, Australia
| | - Janis Shute
- School of Pharmacy and Biomedical Science, University of Portsmouth, Portsmouth, UK
| | - Mary Carroll
- Department of Respiratory Medicine, University of Southampton, Southampton, UK
| | - Julia Tree
- National Infection Service, Public Health England, Porton Down, UK
| | - Miles Carroll
- National Infection Service, Public Health England, Porton Down, UK
| | - Dave Singh
- Medicines Evaluation Unit, University of Manchester, Manchester, UK
| | - Tom Wilkinson
- Department of Respiratory Medicine, University of Southampton, Southampton, UK
| | - Barry Dixon
- Department of Critical Care Medicine, St Vincent's Hospital, Melbourne, Australia
| |
Collapse
|
12
|
Anti-Influenza A Virus Activity of Rhamnan Sulfate from Green Algae Monostroma nitidum in Mice with Normal and Compromised Immunity. Mar Drugs 2020; 18:md18050254. [PMID: 32414158 PMCID: PMC7281209 DOI: 10.3390/md18050254] [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: 03/06/2020] [Revised: 05/03/2020] [Accepted: 05/09/2020] [Indexed: 11/16/2022] Open
Abstract
Influenza viruses cause a significant public health burden each year despite the availability of anti-influenza drugs and vaccines. Therefore, new anti-influenza virus agents are needed. Rhamnan sulfate (RS) is a sulfated polysaccharide derived from the green alga Monostroma nitidum. Here, we aimed to demonstrate the antiviral activity of RS, especially against influenza A virus (IFV) infection, in vitro and in vivo. RS showed inhibitory effects on viral proliferation of enveloped viruses in vitro. Evaluation of the anti-IFV activity of RS in vitro showed that it inhibited both virus adsorption and entry steps. The oral administration of RS in IFV-infected immunocompetent and immunocompromised mice suppressed viral proliferation in both mouse types. The oral administration of RS also had stimulatory effects on neutralizing antibody production. Fluorescent analysis showed that RS colocalized with M cells in Peyer’s patches, suggesting that RS bound to the M cells and may be incorporated into the Peyer’s patches, which are essential to intestinal immunity. In summary, RS inhibits influenza virus infection and promotes antibody production, suggesting that RS is a potential candidate for the treatment of influenza virus infections.
Collapse
|
13
|
Afosah DK, Al-Horani RA. Sulfated Non-Saccharide Glycosaminoglycan Mimetics as Novel Drug Discovery Platform for Various Pathologies. Curr Med Chem 2020; 27:3412-3447. [PMID: 30457046 PMCID: PMC6551317 DOI: 10.2174/0929867325666181120101147] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2018] [Revised: 11/06/2018] [Accepted: 11/13/2018] [Indexed: 01/14/2023]
Abstract
Glycosaminoglycans (GAGs) are very complex, natural anionic polysaccharides. They are polymers of repeating disaccharide units of uronic acid and hexosamine residues. Owing to their template-free, spatiotemporally-controlled, and enzyme-mediated biosyntheses, GAGs possess enormous polydispersity, heterogeneity, and structural diversity which often translate into multiple biological roles. It is well documented that GAGs contribute to physiological and pathological processes by binding to proteins including serine proteases, serpins, chemokines, growth factors, and microbial proteins. Despite advances in the GAG field, the GAG-protein interface remains largely unexploited by drug discovery programs. Thus, Non-Saccharide Glycosaminoglycan Mimetics (NSGMs) have been rationally developed as a novel class of sulfated molecules that modulate GAG-protein interface to promote various biological outcomes of substantial benefit to human health. In this review, we describe the chemical, biochemical, and pharmacological aspects of recently reported NSGMs and highlight their therapeutic potentials as structurally and mechanistically novel anti-coagulants, anti-cancer agents, anti-emphysema agents, and anti-viral agents. We also describe the challenges that complicate their advancement and describe ongoing efforts to overcome these challenges with the aim of advancing the novel platform of NSGMs to clinical use.
Collapse
Affiliation(s)
- Daniel K. Afosah
- Department of Medicinal Chemistry and Institute for Structural Biology, Drug Discovery and Development, Virginia Commonwealth University, Richmond, Virginia 23219
| | - Rami A. Al-Horani
- Division of Basic Pharmaceutical Sciences, College of Pharmacy, Xavier University of Louisiana, New Orleans, Louisiana 70125
| |
Collapse
|
14
|
Barrios-Gumiel A, Sepúlveda-Crespo D, Jiménez JL, Gómez R, Muñoz-Fernández MÁ, de la Mata FJ. Dendronized magnetic nanoparticles for HIV-1 capture and rapid diagnostic. Colloids Surf B Biointerfaces 2019; 181:360-368. [PMID: 31158698 DOI: 10.1016/j.colsurfb.2019.05.050] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2019] [Revised: 05/18/2019] [Accepted: 05/21/2019] [Indexed: 01/29/2023]
Abstract
Human immunodeficiency virus type 1 (HIV-1) remains a global public health problem. Detection and reduction of the rates of late diagnosis of HIV-1 infection are one of the main challenges in combating the HIV-1 epidemic. Magnetic nanoparticles (MNPs) have several characteristics that make them susceptible to capture HIV-1 of a wide range of biological samples reducing waiting times between the acquisition of HIV-1 infection and its detection by current techniques. Carbosilane dendrons decorated with peripheral carboxyl groups and alcoxysilane function at the focal point have been used to stabilize MNPs by co-precipitation method in one step. The characterization of these systems and of their carboxylate analogues was performed by Fourier transform infrared spectroscopy (FTIR), transmission electron microscopy (TEM), dynamic light scattering (DLS), ζ potential and thermal gravimetric analysis (TGA). The ability of carboxyl and carboxylate MNPs to capture R5-HIV-1 and X4-HIV-1 strains was evaluated to achieve a rapid and easy diagnostic method in order to reduce or eliminate the risk of HIV-1 transmission.
Collapse
Affiliation(s)
- Andrea Barrios-Gumiel
- Dpto. de Química Orgánica y Química Inorgánica, Universidad de Alcalá (UAH), Campus Universitario, E-28871 Alcalá de Henares (Madrid), Spain; Instituto de Investigación Química "Andrés M. del Río" (IQAR), Universidad de Alcalá (UAH), Spain; Networking Research Center for Bioengineering, Biomaterials and Nanomedicine, (CIBER-BBN), Spain; Instituto Ramón y Cajal de Investigación Sanitaria, IRYCIS, Spain
| | - Daniel Sepúlveda-Crespo
- Networking Research Center for Bioengineering, Biomaterials and Nanomedicine, (CIBER-BBN), Spain; Sección Inmunología. Laboratorio InmunoBiología Molecular, Hospital General Universitario Gregorio Marañón, Madrid, Spain; Instituto de Investigación Sanitaria Gregorio Marañón (IiSGM), Madrid, Spain; Spanish HIV HGM BioBank, Madrid, Spain
| | - José Luis Jiménez
- Networking Research Center for Bioengineering, Biomaterials and Nanomedicine, (CIBER-BBN), Spain; Sección Inmunología. Laboratorio InmunoBiología Molecular, Hospital General Universitario Gregorio Marañón, Madrid, Spain; Spanish HIV HGM BioBank, Madrid, Spain; Plataforma de Laboratorio, Hospital General Universitario Gregorio Marañón, Madrid, Spain; IiSGM, Madrid, Spain
| | - Rafael Gómez
- Dpto. de Química Orgánica y Química Inorgánica, Universidad de Alcalá (UAH), Campus Universitario, E-28871 Alcalá de Henares (Madrid), Spain; Instituto de Investigación Química "Andrés M. del Río" (IQAR), Universidad de Alcalá (UAH), Spain; Networking Research Center for Bioengineering, Biomaterials and Nanomedicine, (CIBER-BBN), Spain; Instituto Ramón y Cajal de Investigación Sanitaria, IRYCIS, Spain
| | - María Ángeles Muñoz-Fernández
- Networking Research Center for Bioengineering, Biomaterials and Nanomedicine, (CIBER-BBN), Spain; Sección Inmunología. Laboratorio InmunoBiología Molecular, Hospital General Universitario Gregorio Marañón, Madrid, Spain; Instituto de Investigación Sanitaria Gregorio Marañón (IiSGM), Madrid, Spain; Spanish HIV HGM BioBank, Madrid, Spain; Plataforma de Laboratorio, Hospital General Universitario Gregorio Marañón, Madrid, Spain; IiSGM, Madrid, Spain.
| | - F Javier de la Mata
- Dpto. de Química Orgánica y Química Inorgánica, Universidad de Alcalá (UAH), Campus Universitario, E-28871 Alcalá de Henares (Madrid), Spain; Instituto de Investigación Química "Andrés M. del Río" (IQAR), Universidad de Alcalá (UAH), Spain; Networking Research Center for Bioengineering, Biomaterials and Nanomedicine, (CIBER-BBN), Spain; Instituto Ramón y Cajal de Investigación Sanitaria, IRYCIS, Spain.
| |
Collapse
|
15
|
Besednova NN, Zvyagintseva TN, Kuznetsova TA, Makarenkova ID, Smolina TP, Fedyanina LN, Kryzhanovsky SP, Zaporozhets TS. Marine Algae Metabolites as Promising Therapeutics for the Prevention and Treatment of HIV/AIDS. Metabolites 2019; 9:E87. [PMID: 31052506 PMCID: PMC6572556 DOI: 10.3390/metabo9050087] [Citation(s) in RCA: 36] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2019] [Revised: 04/18/2019] [Accepted: 04/26/2019] [Indexed: 11/17/2022] Open
Abstract
This review presents an analysis of works devoted to the anti-human immunodeficiency virus (HIV) activity of algae metabolites-sulfated polysaccharides (fucoidans, carrageenans), lectins, laminarans, and polyphenols. Despite the presence of a significant number of antiretroviral drugs, the development of new therapeutic and prophylactic agents against this infection remains very urgent problem. This is due to the variability of HIV, the absence of an animal model (except monkeys) and natural immunity to this virus and the toxicity of therapeutic agents and their high cost. In this regard, the need for new therapeutic approaches and broad-spectrum drugs, which in addition to antiviral effects can have anti-inflammatory, antioxidant, and immunomodulatory effects, and to which the minimum resistance of HIV strains would be formed. These requirements meet the biologically active substances of marine algae. The results of experimental and clinical studies conducted in vitro and in vivo are presented, and the issues of the anti-HIV activity of these compounds are considered depending on their structural features. On the whole, the presented data prove the high efficiency of seaweed metabolites and justify the possibility of their use as a potential basis for the development of new drugs with a wide spectrum of activity.
Collapse
Affiliation(s)
- Natalya N Besednova
- Federal State Budgetary Scientific Institution, Somov Research Institute of Epidemiology and Microbiology, Sel'skaya street, 1, 690087 Vladivostok, Russia.
| | - Tatyana N Zvyagintseva
- G.B. Elyakov Pacific Institute of Bioorganic Chemistry, Far Eastern Branch of the Russian Academy of Sciences, Pr. 100-letiya Vladivostoka, 159, 690022 Vladivostok, Russia.
| | - Tatyana A Kuznetsova
- Federal State Budgetary Scientific Institution, Somov Research Institute of Epidemiology and Microbiology, Sel'skaya street, 1, 690087 Vladivostok, Russia.
| | - Ilona D Makarenkova
- Federal State Budgetary Scientific Institution, Somov Research Institute of Epidemiology and Microbiology, Sel'skaya street, 1, 690087 Vladivostok, Russia.
| | - Tatyana P Smolina
- Federal State Budgetary Scientific Institution, Somov Research Institute of Epidemiology and Microbiology, Sel'skaya street, 1, 690087 Vladivostok, Russia.
| | - Ludmila N Fedyanina
- Far Eastern Federal University, School of Biomedicine, bldg. M25 FEFU Campus, Ajax Bay, Russky Isl., 690922 Vladivostok, Russia.
| | | | - Tatyana S Zaporozhets
- Federal State Budgetary Scientific Institution, Somov Research Institute of Epidemiology and Microbiology, Sel'skaya street, 1, 690087 Vladivostok, Russia.
| |
Collapse
|
16
|
Kokotidou C, Jonnalagadda SVR, Orr AA, Seoane-Blanco M, Apostolidou CP, van Raaij MJ, Kotzabasaki M, Chatzoudis A, Jakubowski JM, Mossou E, Forsyth VT, Mitchell EP, Bowler MW, Llamas-Saiz AL, Tamamis P, Mitraki A. A novel amyloid designable scaffold and potential inhibitor inspired by GAIIG of amyloid beta and the HIV-1 V3 loop. FEBS Lett 2018; 592:1777-1788. [PMID: 29772603 DOI: 10.1002/1873-3468.13096] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2018] [Accepted: 05/02/2018] [Indexed: 12/11/2022]
Abstract
The GAIIG sequence, common to the amyloid beta peptide (residues 29-33) and to the HIV-1 gp120 (residues 24-28 in a typical V3 loop), self-assembles into amyloid fibrils, as suggested by theory and the experiments presented here. The longer YATGAIIGNII sequence from the V3 loop also self-assembles into amyloid fibrils, of which the first three and the last two residues are outside the amyloid GAIIG core. We postulate that this sequence, with suitably selected modifications at the flexible positions, can serve as a designable scaffold for novel amyloid-based materials. Moreover, we report the single crystal X-ray structure of the beta-breaker peptide GAIPIG at 1.05 Å resolution. The structural information provided in this study could serve as the basis for structure-based design of potential inhibitors of amyloid formation.
Collapse
Affiliation(s)
- Chrysoula Kokotidou
- Department of Materials Science and Technology, University of Crete, Heraklion, Greece.,Institute of Electronic Structure and Laser (IESL), FORTH, Heraklion, Greece
| | | | - Asuka A Orr
- Artie McFerrin Department of Chemical Engineering, Texas A&M University, College Station, TX, USA
| | - Mateo Seoane-Blanco
- Departamento de Estructura de Macromoleculas, Centro Nacional de Biotecnologia (CSIC), Madrid, Spain
| | - Chrysanthi Pinelopi Apostolidou
- Department of Materials Science and Technology, University of Crete, Heraklion, Greece.,Institute of Electronic Structure and Laser (IESL), FORTH, Heraklion, Greece
| | - Mark J van Raaij
- Departamento de Estructura de Macromoleculas, Centro Nacional de Biotecnologia (CSIC), Madrid, Spain
| | - Marianna Kotzabasaki
- Department of Materials Science and Technology, University of Crete, Heraklion, Greece
| | - Apostolos Chatzoudis
- Department of Materials Science and Technology, University of Crete, Heraklion, Greece
| | - Joseph M Jakubowski
- Artie McFerrin Department of Chemical Engineering, Texas A&M University, College Station, TX, USA
| | - Estelle Mossou
- Institut Laue Langevin, Grenoble Cedex 9, France.,Faculty of Natural Sciences/Institute for Science and Technology in Medicine, Keele University, Staffordshire, UK
| | - V Trevor Forsyth
- Institut Laue Langevin, Grenoble Cedex 9, France.,Faculty of Natural Sciences/Institute for Science and Technology in Medicine, Keele University, Staffordshire, UK
| | - Edward P Mitchell
- Faculty of Natural Sciences/Institute for Science and Technology in Medicine, Keele University, Staffordshire, UK.,European Synchrotron Radiation Facility, Grenoble Cedex 9, France
| | - Matthew W Bowler
- European Molecular Biology Laboratory, Grenoble, France.,Unit for Virus Host Cell Interactions, University Grenoble Alpes-EMBL-CNRS, Grenoble, France
| | - Antonio L Llamas-Saiz
- X-Ray Unit, RIAIDT, University of Santiago de Compostela, Santiago de Compostela, Spain
| | - Phanourios Tamamis
- Artie McFerrin Department of Chemical Engineering, Texas A&M University, College Station, TX, USA
| | - Anna Mitraki
- Department of Materials Science and Technology, University of Crete, Heraklion, Greece.,Institute of Electronic Structure and Laser (IESL), FORTH, Heraklion, Greece
| |
Collapse
|
17
|
Shah HR, Savjani JK. Recent updates for designing CCR5 antagonists as anti-retroviral agents. Eur J Med Chem 2018; 147:115-129. [PMID: 29425816 DOI: 10.1016/j.ejmech.2018.01.085] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2017] [Revised: 01/11/2018] [Accepted: 01/26/2018] [Indexed: 01/23/2023]
Abstract
The healthcare system faces various challenges in human immunodeficiency virus (HIV) therapy due to resistance to Anti-Retroviral Therapy (ART) as a consequence of the evolutionary process. Despite the success of antiretroviral drugs like Zidovudine, Zalcitabine, Raltegravir WHO ranks HIV as one of the deadliest diseases with a mortality of one million lives in 2016. Thus, there emerges an urgency of developing a novel anti-retroviral agent that combat resistant HIV strains. The clinical development of ART from a single drug regimen to current triple drug combination is very slow. The progression in the structural biology of the viral envelope prompted the discovery of novel targets, which can be demonstrated a proficient approach for drug design of anti-retroviral agents. The current review enlightens the recent updates in the structural biology of the viral envelope and focuses on CCR5 as a validated target as well as ways to overcome CCR5 resistance. The article also throws light on the SAR studies and most prevalent mutations in the receptor for designing CCR5 antagonists that can combat HIV-1 infection. To conclude, the paper lists diversified scaffolds that are in pipeline by various pharmaceutical companies that could provide an aid for developing novel CCR5 antagonists.
Collapse
Affiliation(s)
- Harshil R Shah
- Department of Pharmaceutical Chemistry, Institute of Pharmacy, Nirma University, S.G. Highway, Ahmedabad 382481, India
| | - Jignasa Ketan Savjani
- Department of Pharmaceutical Chemistry, Institute of Pharmacy, Nirma University, S.G. Highway, Ahmedabad 382481, India.
| |
Collapse
|
18
|
Hollingsworth LR, Brown AM, Gandour RD, Bevan DR. Computational study of HIV gp120 as a target for polyanionic entry inhibitors: Exploiting the V3 loop region. PLoS One 2018; 13:e0190658. [PMID: 29346393 PMCID: PMC5773097 DOI: 10.1371/journal.pone.0190658] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2017] [Accepted: 12/18/2017] [Indexed: 01/09/2023] Open
Abstract
Multiple approaches are being utilized to develop therapeutics to treat HIV infection. One approach is designed to inhibit entry of HIV into host cells, with a target being the viral envelope glycoprotein, gp120. Polyanionic compounds have been shown to be effective in inhibiting HIV entry, with a mechanism involving electrostatic interactions with the V3 loop of gp120 being proposed. In this study, we applied computational methods to elucidate molecular interactions between the repeat unit of the precisely alternating polyanion, Poly(4,4′-stilbenedicarboxylate-alt–maleic acid) (DCSti-alt-MA) and the V3 loop of gp120 from strains of HIV against which these polyanions were previously tested (IIIb, BaL, 92UG037, JR-CSF) as well as two strains for which gp120 crystal structures are available (YU2, 2B4C). Homology modeling was used to create models of the gp120 proteins. Using monomers of the gp120 protein, we applied extensive molecular dynamics simulations to obtain dominant morphologies that represent a variety of open-closed states of the V3 loop to examine the interaction of 112 ligands of the repeating units of DCSti-alt-MA docked to the V3 loop and surrounding residues. Using the distance between the V1/V2 and V3 loops of gp120 as a metric, we revealed through MD simulations that gp120 from the lab-adapted strains (BaL and IIIb), which are more susceptible to inhibition by DCSti-alt-MA, clearly transitioned to the closed state in one replicate of each simulation set, whereas none of the replicates from the Tier II strains (92UG037 and JR-CSF) did so. Docking repeat unit microspecies to the gp120 protein before and after MD simulation enabled identification of residues that were key for binding. Notably, only a few residues were found to be important for docking both before and after MD simulation as a result of the conformational heterogeneity provided by the simulations. Consideration of the residues that were consistently involved in interactions with the ligand revealed the importance of both hydrophilic and hydrophobic moieties of the ligand for effective binding. The results also suggest that polymers of DCSti-alt-MA with repeating units of different configurations may have advantages for therapeutic efficacy.
Collapse
Affiliation(s)
- Louis R. Hollingsworth
- Department of Chemical Engineering, Virginia Tech, Blacksburg, Virginia, United States of America
- Department of Biochemistry, Virginia Tech, Blacksburg, Virginia, United States of America
- Department of Chemistry, Virginia Tech, Blacksburg, Virginia, United States of America
| | - Anne M. Brown
- Department of Biochemistry, Virginia Tech, Blacksburg, Virginia, United States of America
- Research and Informatics, University Libraries, Virginia Tech, Blacksburg, Virginia, United States of America
| | - Richard D. Gandour
- Department of Chemistry, Virginia Tech, Blacksburg, Virginia, United States of America
- Virginia Tech Center for Drug Discovery, Virginia Tech, Blacksburg, Virginia, United States of America
| | - David R. Bevan
- Department of Biochemistry, Virginia Tech, Blacksburg, Virginia, United States of America
- Virginia Tech Center for Drug Discovery, Virginia Tech, Blacksburg, Virginia, United States of America
- * E-mail:
| |
Collapse
|
19
|
Ayerst BI, Smith RAA, Nurcombe V, Day AJ, Merry CLR, Cool SM. Growth Differentiation Factor 5-Mediated Enhancement of Chondrocyte Phenotype Is Inhibited by Heparin: Implications for the Use of Heparin in the Clinic and in Tissue Engineering Applications. Tissue Eng Part A 2017; 23:275-292. [PMID: 27899064 PMCID: PMC5397242 DOI: 10.1089/ten.tea.2016.0364] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/30/2023] Open
Abstract
The highly sulfated glycosaminoglycan (GAG) heparin is widely used in the clinic as an anticoagulant, and researchers are now using it to enhance stem cell expansion/differentiation protocols, as well as to improve the delivery of growth factors for tissue engineering (TE) strategies. Growth differentiation factor 5 (GDF5) belongs to the bone morphogenetic protein family of proteins and is vital for skeletal formation; however, its interaction with heparin and heparan sulfate (HS) has not been studied. We identify GDF5 as a novel heparin/HS binding protein and show that HS proteoglycans are vital in localizing GDF5 to the cell surface. Clinically relevant doses of heparin (≥10 nM), but not equivalent concentrations of HS, were found to inhibit GDF5's biological activity in both human mesenchymal stem/stromal cell-derived chondrocyte pellet cultures and the skeletal cell line ATDC5. We also found that heparin inhibited both GDF5 binding to cell surface HS and GDF5-induced induction of Smad 1/5/8 signaling. Furthermore, GDF5 significantly increased aggrecan gene expression in chondrocyte pellet cultures, without affecting collagen type X expression, making it a promising target for the TE of articular cartilage. Importantly, this study may explain the variable (and disappointing) results seen with heparin-loaded biomaterials for skeletal TE and the adverse skeletal effects reported in the clinic following long-term heparin treatment. Our results caution the use of heparin in the clinic and in TE applications, and prompt the transition to using more specific GAGs (e.g., HS derivatives), with better-defined structures and fewer off-target effects.
Collapse
Affiliation(s)
- Bethanie I Ayerst
- 1 Institute of Medical Biology , Agency for Science, Technology and Research (A*STAR), Singapore, Singapore .,2 Wellcome Trust Centre for Cell-Matrix Research, Division of Cell Matrix Biology and Regenerative Medicine, School of Biology, Faculty of Biology Medicine and Health, Manchester Academic Health Science Centre, University of Manchester , Manchester, United Kingdom
| | - Raymond A A Smith
- 1 Institute of Medical Biology , Agency for Science, Technology and Research (A*STAR), Singapore, Singapore
| | - Victor Nurcombe
- 1 Institute of Medical Biology , Agency for Science, Technology and Research (A*STAR), Singapore, Singapore
| | - Anthony J Day
- 2 Wellcome Trust Centre for Cell-Matrix Research, Division of Cell Matrix Biology and Regenerative Medicine, School of Biology, Faculty of Biology Medicine and Health, Manchester Academic Health Science Centre, University of Manchester , Manchester, United Kingdom
| | - Catherine L R Merry
- 3 School of Materials, University of Manchester , Manchester, United Kingdom .,4 Wolfson Centre for Stem Cells, Tissue Engineering and Modelling, Centre for Biomolecular Sciences, University of Nottingham , Nottingham, United Kingdom
| | - Simon M Cool
- 1 Institute of Medical Biology , Agency for Science, Technology and Research (A*STAR), Singapore, Singapore .,5 Department of Orthopaedic Surgery, Yong Loo Lin School of Medicine, National University of Singapore , Singapore, Singapore
| |
Collapse
|
20
|
Zhernov YV, Kremb S, Helfer M, Schindler M, Harir M, Mueller C, Hertkorn N, Avvakumova NP, Konstantinov AI, Brack-Werner R, Schmitt-Kopplin P, Perminova IV. Supramolecular combinations of humic polyanions as potent microbicides with polymodal anti-HIV-activities. NEW J CHEM 2017. [DOI: 10.1039/c6nj00960c] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022]
Abstract
Anti-HIV potency of humic PAs is governed by scaffolds diversity.
Collapse
Affiliation(s)
- Yury V. Zhernov
- State Research Center “Institute of Immunology” of the Federal Medical-Biological Agency of Russia
- Moscow
- Russia
| | - Stephan Kremb
- Institute of Virology, Helmholtz Zentrum München – German Research Center for Environmental Health
- Neuherberg
- Germany
| | - Markus Helfer
- Institute of Virology, Helmholtz Zentrum München – German Research Center for Environmental Health
- Neuherberg
- Germany
| | - Michael Schindler
- University Hospital Tübingen
- Institute for Medical Virology and Epidemiology of Viral Diseases
- Tübingen
- Germany
| | - Mourad Harir
- Research Unit Analytical BioGeoChemistry
- Helmholtz Zentrum München – German Research Center for Environmental Health
- Neuherberg
- Germany
| | - Constanze Mueller
- Research Unit Analytical BioGeoChemistry
- Helmholtz Zentrum München – German Research Center for Environmental Health
- Neuherberg
- Germany
| | - Norbert Hertkorn
- Research Unit Analytical BioGeoChemistry
- Helmholtz Zentrum München – German Research Center for Environmental Health
- Neuherberg
- Germany
| | - Nadezhda P. Avvakumova
- Samara State Medical University
- Department of General, Bioinorganic and Bioorganic Chemistry
- Samara
- Russia
| | | | - Ruth Brack-Werner
- Institute of Virology, Helmholtz Zentrum München – German Research Center for Environmental Health
- Neuherberg
- Germany
| | - Philippe Schmitt-Kopplin
- Research Unit Analytical BioGeoChemistry
- Helmholtz Zentrum München – German Research Center for Environmental Health
- Neuherberg
- Germany
- Technical University of Munich
| | | |
Collapse
|
21
|
Connell BJ, Chang SY, Prakash E, Yousfi R, Mohan V, Posch W, Wilflingseder D, Moog C, Kodama EN, Clayette P, Lortat-Jacob H. A Cinnamon-Derived Procyanidin Compound Displays Anti-HIV-1 Activity by Blocking Heparan Sulfate- and Co-Receptor- Binding Sites on gp120 and Reverses T Cell Exhaustion via Impeding Tim-3 and PD-1 Upregulation. PLoS One 2016; 11:e0165386. [PMID: 27788205 PMCID: PMC5082894 DOI: 10.1371/journal.pone.0165386] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2016] [Accepted: 10/11/2016] [Indexed: 11/18/2022] Open
Abstract
Amongst the many strategies aiming at inhibiting HIV-1 infection, blocking viral entry has been recently recognized as a very promising approach. Using diverse in vitro models and a broad range of HIV-1 primary patient isolates, we report here that IND02, a type A procyanidin polyphenol extracted from cinnamon, that features trimeric and pentameric forms displays an anti-HIV-1 activity against CXCR4 and CCR5 viruses with 1–7 μM ED50 for the trimer. Competition experiments, using a surface plasmon resonance-based binding assay, revealed that IND02 inhibited envelope binding to CD4 and heparan sulphate (HS) as well as to an antibody (mAb 17b) directed against the gp120 co-receptor binding site with an IC50 in the low μM range. IND02 has thus the remarkable property of simultaneously blocking gp120 binding to its major host cell surface counterparts. Additionally, the IND02-trimer impeded up-regulation of the inhibitory receptors Tim-3 and PD-1 on CD4+ and CD8+ cells, thereby demonstrating its beneficial effect by limiting T cell exhaustion. Among naturally derived products significantly inhibiting HIV-1, the IND02-trimer is the first component demonstrating an entry inhibition property through binding to the viral envelope glycoprotein. These data suggest that cinnamon, a widely consumed spice, could represent a novel and promising candidate for a cost-effective, natural entry inhibitor for HIV-1 which can also down-modulate T cell exhaustion markers Tim-3 and PD-1.
Collapse
Affiliation(s)
- Bridgette Janine Connell
- Institut de Biologie Structurale, UMR 5075, Univ. Grenoble Alpes, CNRS, CEA, F-38027 Grenoble, France
| | - Sui-Yuan Chang
- School of Medical Technology, College of Medicine, National Taiwan University Hospital, Taipei, Taiwan
| | | | - Rahima Yousfi
- Laboratoire de Neurovirologie, Bertin Pharma, CEA, 92265 Fontenay aux Roses, France
| | | | - Wilfried Posch
- Division of Hygiene and Medical Microbiology, Innsbruck Medical University, Innsbruck, Austria
| | - Doris Wilflingseder
- Division of Hygiene and Medical Microbiology, Innsbruck Medical University, Innsbruck, Austria
| | - Christiane Moog
- INSERM U1110, Fédération de médecine translationnelle de Strasbourg (FMTS), Institut de Virologie, 3 rue Koeberlé, 67000 Strasbourg, France
| | - Eiichi N. Kodama
- Division of Emerging Infectious Diseases, Miyagi Communitiy Health Promotion, Tohoku University School of Medicine, Bldg. 1, Rm. 515, 2–1 Seiryocho, Aoba-ku, Sendai 980–8575, Japan
| | - Pascal Clayette
- Laboratoire de Neurovirologie, Bertin Pharma, CEA, 92265 Fontenay aux Roses, France
| | - Hugues Lortat-Jacob
- Institut de Biologie Structurale, UMR 5075, Univ. Grenoble Alpes, CNRS, CEA, F-38027 Grenoble, France
- * E-mail: (HLJ); (EP)
| |
Collapse
|
22
|
Sepúlveda-Crespo D, Ceña-Díez R, Jiménez JL, Ángeles Muñoz-Fernández M. Mechanistic Studies of Viral Entry: An Overview of Dendrimer-Based Microbicides As Entry Inhibitors Against Both HIV and HSV-2 Overlapped Infections. Med Res Rev 2016; 37:149-179. [PMID: 27518199 DOI: 10.1002/med.21405] [Citation(s) in RCA: 37] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2016] [Revised: 06/16/2016] [Accepted: 07/06/2016] [Indexed: 12/12/2022]
Abstract
This review provides an overview of the development of different dendrimers, mainly polyanionic, against human immunodeficiency virus (HIV) and genital herpes (HSV-2) as topical microbicides targeting the viral entry process. Vaginal topical microbicides to prevent sexually transmitted infections such as HIV and HSV-2 are urgently needed. To inhibit HIV/HSV-2 entry processes, new preventive targets have been established to maximize the current therapies against wild-type and drug-resistant viruses. The entry of HIV/HSV-2 into target cells is a multistep process that triggers a cascade of molecular interactions between viral envelope proteins and cell surface receptors. Polyanionic dendrimers are highly branched nanocompounds with potent activity against HIV/HSV-2. Inhibitors of each entry step have been identified with regard to generations and surface groups, and possible roles for these agents in anti-HIV/HSV-2 therapies have also been discussed. Four potential binding sites for impeding HIV infection (HSPG, DC-SIGN, GSL, and CD4/gp120 inhibitors) and HSV-2 infection (HS, gB, gD, and gH/gL inhibitors) exist according to their mechanisms of action and structures. This review clarifies that inhibition of HIV/HSV-2 entry continues to be a promising target for drug development because nanotechnology can transform the field of HIV/HSV-2 prevention by improving the efficacy of the currently available antiviral treatments.
Collapse
Affiliation(s)
- Daniel Sepúlveda-Crespo
- Laboratorio InmunoBiología Molecular, Hospital General Universitario Gregorio Marañón, Madrid, Spain.,Instituto de Investigación Sanitaria Gregorio Marañón (IiSGM), Spanish HIV-HGM BioBank, Madrid, Spain.,Networking Research Center on Bioengineering, Biomaterials and Nanomedicine (CIBER-BBN), Madrid, Spain
| | - Rafael Ceña-Díez
- Laboratorio InmunoBiología Molecular, Hospital General Universitario Gregorio Marañón, Madrid, Spain.,Instituto de Investigación Sanitaria Gregorio Marañón (IiSGM), Spanish HIV-HGM BioBank, Madrid, Spain.,Networking Research Center on Bioengineering, Biomaterials and Nanomedicine (CIBER-BBN), Madrid, Spain
| | - José Luis Jiménez
- Instituto de Investigación Sanitaria Gregorio Marañón (IiSGM), Spanish HIV-HGM BioBank, Madrid, Spain.,Networking Research Center on Bioengineering, Biomaterials and Nanomedicine (CIBER-BBN), Madrid, Spain.,Plataforma de Laboratorio, Hospital General Universitario Gregorio Marañón, Madrid, Spain
| | - Ma Ángeles Muñoz-Fernández
- Laboratorio InmunoBiología Molecular, Hospital General Universitario Gregorio Marañón, Madrid, Spain.,Instituto de Investigación Sanitaria Gregorio Marañón (IiSGM), Spanish HIV-HGM BioBank, Madrid, Spain.,Networking Research Center on Bioengineering, Biomaterials and Nanomedicine (CIBER-BBN), Madrid, Spain
| |
Collapse
|
23
|
Nakamura M, Terada M, Kamada M, Yokono A, Nakamori S, Ohno T. Mechanistic Effect of NMSO3 on Replication of Human Immunodeficiency Virus. ACTA ACUST UNITED AC 2016; 14:171-6. [PMID: 14582845 DOI: 10.1177/095632020301400401] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
NMSO3, a sulphated sialyl lipid, was evaluated for its efficacy against human immunodeficiency virus type 1 (HIV-1). The compound exhibited concentration-dependent inhibition of HIV-1 replication in primary infection cell culture systems. Substantial inhibition was observed at concentrations of NMSO3 that showed little cytotoxicity. NMSO3 also exhibited anti HIV-1 activity in chronically HIV-1 infected cultures. The production of progeny viruses was completely abolished without cytotoxicity by continuous addition of NMSO3 to chronically infected U937 cells. Furthermore, in attempting to define the inhibitory mechanism of NMSO3, we investigated its effect on several steps of the HIV-1 replication cycle. NMSO3 competes with gp120 for binding to CD4 receptors on cells and inhibits the entry of HIV-1. By epitope analysis of the human CD4 molecule, NMSO3 inhibits the binding of antibodies, which recognize the D1 domain of CD4. Moreover, semi-quantitative reverse transcribed polymerase chain reaction (RT-PCR) showed that the integrated provirus is transcriptionally inactive in NMSO3-treated cells, supporting the lack of progeny in the culture supernatant of chronically HIV-1-infected cells treated with NMSO3. These findings indicate that NMSO3 has a unique mechanism of action against HIV-1 in both primary and chronic infection, and may be a valuable compound for the treatment of HIV-1 infection.
Collapse
Affiliation(s)
- Mariko Nakamura
- Department of Microbiology, Jikei University School of Medicine, 3-25-8 Nishi-Shinbashi, Minato-ku, Tokyo 105-8461 Japan.
| | | | | | | | | | | |
Collapse
|
24
|
Yamada H, Nagase S, Takahashi K, Sakoda Y, Kida H, Okamoto S. Toll-like receptor 9 ligand D-type oligodeoxynucleotide D35 as a broad inhibitor for influenza A virus replication that is associated with suppression of neuraminidase activity. Antiviral Res 2016; 129:81-92. [PMID: 26923882 PMCID: PMC7113795 DOI: 10.1016/j.antiviral.2016.02.012] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2015] [Revised: 02/09/2016] [Accepted: 02/17/2016] [Indexed: 12/18/2022]
Abstract
The most effective drugs available to treat influenza are neuraminidase (NA) inhibitors, which provide important additional measures for the control of influenza virus infections. However, since the emergence of NA inhibitor-resistant viruses may compromise the clinical utility of this class of anti-influenza agents, it is very important to develop new anti-influenza agents which target a different region in NA responsible for its sensitivity from that for NA inhibitors and could be used to treat NA inhibitors-resistant isolates. The oligodeoxynucleotide D35, multimerized and aggregated, suppressed replication of influenza A viruses except A/WSN/33 (WSN). The suppressive viral replication by D35 depended on G-terad and multimer formation. The range of the suppressive viral replication at the late stage, including virus assembly and release from infected cells, was much larger than that at the initial stage, viral attachment and entry. D35 suppressed NA activity of influenza A viruses. Furthermore, replacing the NA gene of A/Puerto Rico/8/34 (PR8), in which viral replication was inhibited by D35 at the late stage, with the NA gene from WSN, in which viral replication was not inhibited, eliminated the D35-dependent suppression. D35 showed an additive anti-influenza effect with oseltamivir. It was also effective in vivo. These results suggest that the influenza virus NA mainly contributse to the D35-suppressible virus release from infected cells at the late stage. In addition, because administration of D35 into the virus-infected mice suppressed viral replication and weight loss, clinical application of D35 could be considered. The oligodeoxynucleotide D35 suppressed replication of some influenza A viruses. D35 inhibits viral replication at the late step which is dependent on NA activity. Antiviral mechanism by D35 is different from that by oseltamivir.
Collapse
Affiliation(s)
- Hiroshi Yamada
- Laboratory of Virology and Vaccinology, National Institute of Biomedical Innovation, Ibaraki, Osaka, Japan
| | - Satoshi Nagase
- Department of Laboratory Sciences, Division of Health Sciences, Kanazawa University Graduate School of Medical Science, Kanazawa, Japan
| | - Kazuo Takahashi
- Department of Infectious Diseases, Osaka Prefectural Institute of Public Health, Osaka, Japan
| | - Yoshihiro Sakoda
- Laboratory of Microbiology, Graduate School of Veterinary Medicine, Hokkaido University, Sapporo, Japan
| | - Hiroshi Kida
- Laboratory of Microbiology, Graduate School of Veterinary Medicine, Hokkaido University, Sapporo, Japan; Research Center for Zoonosis Control, Hokkaido University, Sapporo, Japan
| | - Shigefumi Okamoto
- Department of Laboratory Sciences, Division of Health Sciences, Kanazawa University Graduate School of Medical Science, Kanazawa, Japan.
| |
Collapse
|
25
|
Peña-González CE, García-Broncano P, Ottaviani MF, Cangiotti M, Fattori A, Hierro-Oliva M, González-Martín ML, Pérez-Serrano J, Gómez R, Muñoz-Fernández MÁ, Sánchez-Nieves J, de la Mata FJ. Dendronized Anionic Gold Nanoparticles: Synthesis, Characterization, and Antiviral Activity. Chemistry 2016; 22:2987-99. [PMID: 26875938 DOI: 10.1002/chem.201504262] [Citation(s) in RCA: 29] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2015] [Indexed: 12/23/2022]
Abstract
Anionic carbosilane dendrons decorated with sulfonate functions and one thiol moiety at the focal point have been used to synthesize water-soluble gold nanoparticles (AuNPs) through the direct reaction of dendrons, gold precursor, and reducing agent in water, and also through a place-exchange reaction. These nanoparticles have been characterized by NMR spectroscopy, TEM, thermogravimetric analysis, X-ray photoelectron spectroscopy (XPS), UV/Vis spectroscopy, elemental analysis, and zeta-potential measurements. The interacting ability of the anionic sulfonate functions was investigated by EPR spectroscopy with copper(II) as a probe. Different structures and conformations of the AuNPs modulate the availability of sulfonate and thiol groups for complexation by copper(II). Toxicity assays of AuNPs showed that those produced through direct reaction were less toxic than those obtained by ligand exchange. Inhibition of HIV-1 infection was higher in the case of dendronized AuNPs than in dendrons.
Collapse
Affiliation(s)
- Cornelia E Peña-González
- Dpto. de Química Orgánica y Química Inorgánica, Edificio de Farmacia, Universidad de Alcalá, Campus Universitario, Alcalá de Henares (Madrid), Spain
| | - Pilar García-Broncano
- Unidad de Infección Viral e Inmunidad, Centro Nacional de Microbiología, Instituto de Salud Carlos III, Campus Majadahonda, Madrid, Spain.,Laboratorio de InmunoBiología Molecular, Hospital General Universitario Gregorio Marañón, Madrid, Spain.,Instituto de Investigación Sanitaria Gregorio Marañón, Madrid, Spain.,Spanish HIV HGM BioBank, Madrid, Spain
| | - M Francesca Ottaviani
- Department of Earth, Life and Environment Sciences, University of Urbino, Urbino, 61029, Italy
| | - Michela Cangiotti
- Department of Earth, Life and Environment Sciences, University of Urbino, Urbino, 61029, Italy
| | - Alberto Fattori
- Department of Earth, Life and Environment Sciences, University of Urbino, Urbino, 61029, Italy
| | - Margarita Hierro-Oliva
- Departamento de Física Aplicada, Facultad de Ciencias, Campus Universitario, Universidad de Extremadura, Badajoz, Spain.,Networking Research Center for Bioengineering Biomaterials and Nanomedicine (CIBER-BBN), Instituto de Salud Carlos III, Madrid, Spain
| | - M Luisa González-Martín
- Departamento de Física Aplicada, Facultad de Ciencias, Campus Universitario, Universidad de Extremadura, Badajoz, Spain.,Networking Research Center for Bioengineering Biomaterials and Nanomedicine (CIBER-BBN), Instituto de Salud Carlos III, Madrid, Spain
| | - Jorge Pérez-Serrano
- Departamento de Biomedicina y Biotecnología, Edificio de Farmacia, Campus Universitario, Universidad de Alcalá, 28871, Alcalá de Henares, Spain
| | - Rafael Gómez
- Dpto. de Química Orgánica y Química Inorgánica, Edificio de Farmacia, Universidad de Alcalá, Campus Universitario, Alcalá de Henares (Madrid), Spain.,Networking Research Center for Bioengineering Biomaterials and Nanomedicine (CIBER-BBN), Instituto de Salud Carlos III, Madrid, Spain
| | - M Ángeles Muñoz-Fernández
- Laboratorio de InmunoBiología Molecular, Hospital General Universitario Gregorio Marañón, Madrid, Spain.,Instituto de Investigación Sanitaria Gregorio Marañón, Madrid, Spain.,Spanish HIV HGM BioBank, Madrid, Spain.,Networking Research Center for Bioengineering Biomaterials and Nanomedicine (CIBER-BBN), Instituto de Salud Carlos III, Madrid, Spain
| | - Javier Sánchez-Nieves
- Dpto. de Química Orgánica y Química Inorgánica, Edificio de Farmacia, Universidad de Alcalá, Campus Universitario, Alcalá de Henares (Madrid), Spain. .,Networking Research Center for Bioengineering Biomaterials and Nanomedicine (CIBER-BBN), Instituto de Salud Carlos III, Madrid, Spain.
| | - F Javier de la Mata
- Dpto. de Química Orgánica y Química Inorgánica, Edificio de Farmacia, Universidad de Alcalá, Campus Universitario, Alcalá de Henares (Madrid), Spain. .,Networking Research Center for Bioengineering Biomaterials and Nanomedicine (CIBER-BBN), Instituto de Salud Carlos III, Madrid, Spain.
| |
Collapse
|
26
|
Zhang X, Ao Z, Bello A, Ran X, Liu S, Wigle J, Kobinger G, Yao X. Characterization of the inhibitory effect of an extract of Prunella vulgaris on Ebola virus glycoprotein (GP)-mediated virus entry and infection. Antiviral Res 2016; 127:20-31. [PMID: 26778707 PMCID: PMC7113790 DOI: 10.1016/j.antiviral.2016.01.001] [Citation(s) in RCA: 35] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2015] [Revised: 01/05/2016] [Accepted: 01/06/2016] [Indexed: 11/25/2022]
Abstract
Currently, no approved antiviral therapeutic is available for treatment or prevention of Ebola virus (EBOV) infection. In this study, we characterized an EBOV-glycoprotein (GP) pseudotyped HIV-1-based vector system in different cell cultures, including human umbilical vein endothelial cells (HUVECs) and human macrophages, for the screening of anti-EBOV-GP agent(s). Based on this system, we demonstrated that an aqueous extract (CHPV) from the Chinese herb Prunella vulgaris displayed a potent inhibitory effect on EBOV-GP pseudotyped virus (EBOV-GP-V)-mediated infection in various cell lines, including HUVEC and macrophage. In addition, our results indicated that CHPV was able to block an eGFP-expressing Zaire ebola virus (eGFP-ZEBOV) infection in VeroE6 cells. The anti-EBOV activity of CHPV was exhibited in a dose-dependent manner. At a 12.5 μg/ml concentration, the CHPV showed a greater than 80% inhibition of EBOV-GP-V and eGFP-EBOV infections. Likewise, our studies suggested that the inhibitory effect of CHPV occurred by binding directly to EBOV-GP-Vs and blocking the early viral events. Interestingly, our results have shown that CHPV was able to enhance the anti-EBOV activity of the monoclonal antibody MAb 2G4 against EBOV-GP. Overall, this study provides evidence that CHPV has anti-EBOV activity and may be developed as a novel antiviral approach against EBOV infection.
Collapse
Affiliation(s)
- Xu Zhang
- Laboratory of Molecular Human Retrovirology, Department of Medical Microbiology, Faculty of Medicine, University of Manitoba, Canada
| | - Zhujun Ao
- Laboratory of Molecular Human Retrovirology, Department of Medical Microbiology, Faculty of Medicine, University of Manitoba, Canada; Department of Microbiology, School of Basic Medical Sciences, Central South University, Changsha, Hunan, 410078, PR China
| | - Alexander Bello
- Special Pathogens Program, National Microbiology Laboratory, Public Health Agency of Canada, Canada
| | - Xiaozhuo Ran
- Laboratory of Molecular Human Retrovirology, Department of Medical Microbiology, Faculty of Medicine, University of Manitoba, Canada
| | - Shuiping Liu
- Department of Microbiology, School of Basic Medical Sciences, Central South University, Changsha, Hunan, 410078, PR China
| | - Jeffrey Wigle
- Department of Biochemistry and Medical Genetics, University of Manitoba, Canada
| | - Gary Kobinger
- Special Pathogens Program, National Microbiology Laboratory, Public Health Agency of Canada, Canada
| | - Xiaojian Yao
- Laboratory of Molecular Human Retrovirology, Department of Medical Microbiology, Faculty of Medicine, University of Manitoba, Canada; Department of Microbiology, School of Basic Medical Sciences, Central South University, Changsha, Hunan, 410078, PR China.
| |
Collapse
|
27
|
Ceña-Díez R, Sepúlveda-Crespo D, Maly M, Muñoz-Fernández MA. Dendrimeric based microbicides against sexual transmitted infections associated to heparan sulfate. RSC Adv 2016. [DOI: 10.1039/c6ra06969j] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022] Open
Abstract
Cell surface heparan sulfate (HS) represents a common link that many sexually transmitted infections (STIs) require for infection.
Collapse
Affiliation(s)
- Rafael Ceña-Díez
- Laboratorio InmunoBiología Molecular
- Hospital General Universitario Gregorio Marañón
- 28007 Madrid
- Spain
- Instituto de Investigación Sanitaria Gregorio Marañón (IiSGM)
| | - Daniel Sepúlveda-Crespo
- Laboratorio InmunoBiología Molecular
- Hospital General Universitario Gregorio Marañón
- 28007 Madrid
- Spain
- Instituto de Investigación Sanitaria Gregorio Marañón (IiSGM)
| | - Marek Maly
- Department of Innovative Technologies
- University of Applied Science of Southern Switzerland
- Switzerland
- Faculty of Science
- J. E. Purkinje University
| | - Mª Angeles Muñoz-Fernández
- Laboratorio InmunoBiología Molecular
- Hospital General Universitario Gregorio Marañón
- 28007 Madrid
- Spain
- Instituto de Investigación Sanitaria Gregorio Marañón (IiSGM)
| |
Collapse
|
28
|
Heredia A, Latinovic OS, Barbault F, de Leeuw EPH. A novel small-molecule inhibitor of HIV-1 entry. DRUG DESIGN DEVELOPMENT AND THERAPY 2015; 9:5469-78. [PMID: 26491257 PMCID: PMC4598220 DOI: 10.2147/dddt.s89338] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
Background Antiretroviral therapy has transformed HIV-1 infection into a managed condition with near-normal life expectancy. However, a significant number of patients remain with limited therapeutic options due to HIV-1 resistance, side effects, or drug costs. Further, it is likely that current drugs will not retain efficacy, due to risks of side effects and transmitted resistance. Results We describe compound 5660386 (3-ethyl-2-[3-(1,3,3-trimethyl-1,3-dihydro-2H-indol-2-ylidene)-1-propen-1-yl]-1,3-benzothiazol-3-ium) as a novel inhibitor of HIV-1 entry. Compound 5660386 inhibits HIV-1 entry in cell lines and primary cells, binds to HIV-1 envelope protein, and inhibits the interaction of GP120 to CD4. Further, compound 5660386 showed a unique and broad-range activity against primary HIV-1 isolates from different subtypes and geographical areas. Conclusion Development of small-molecule entry inhibitors of HIV-1 such as 5660386 may lead to novel classes of anti-HIV-1 therapeutics. These inhibitors may be particularly effective against viruses resistant to current antiretroviral drugs and could have potential applications in both treatment and prevention.
Collapse
Affiliation(s)
- Alonso Heredia
- Department of Medicine, University of Maryland Baltimore School of Medicine, Baltimore, MD, USA ; Institute of Human Virology, University of Maryland Baltimore School of Medicine, Baltimore, MD, USA
| | - Olga S Latinovic
- Department of Microbiology and Immunology, University of Maryland Baltimore School of Medicine, Baltimore, MD, USA ; Institute of Human Virology, University of Maryland Baltimore School of Medicine, Baltimore, MD, USA
| | - Florent Barbault
- Univ Paris Diderot, Sorbonne Paris Cité, ITODYS, UMRCNRS7086, Paris, France
| | - Erik P H de Leeuw
- Institute of Human Virology, University of Maryland Baltimore School of Medicine, Baltimore, MD, USA ; Department of Biochemistry and Molecular Biology, University of Maryland Baltimore School of Medicine, Baltimore, MD, USA
| |
Collapse
|
29
|
Shcherbakov DN, Bakulina AY, Karpenko LI, Ilyichev AA. Broadly Neutralizing Antibodies against HIV-1 As a Novel Aspect of the Immune Response. Acta Naturae 2015; 7:11-21. [PMID: 26798488 PMCID: PMC4717246] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022] Open
Abstract
The human immunodeficiency virus-1 (HIV-1) has the ability to evade the adaptive immune response due to high mutation rates. Soon after the discovery of HIV-1, it was originally proposed that neutralizing of antibodies to the virus occurs rarely or cannot be elicited at all. In the 1990s, there appeared reports that sera of select HIV-1-infected individuals contained antibodies capable of neutralizing different virus subtypes. Such antibodies were named broadly neutralizing antibodies (bNAbs). Since 2009, the development of new cell technologies has intensified research efforts directed at identifying new bNAbs with a neutralization potency of over 90% of primary HIV-1 isolates. These antibodies have unique characteristics which include high levels of somatic mutations and unusually long variable loops that penetrate through the glycan shield of HIV-1 Env to contact the protein surface. In this review, we will attempt to summarize the latest data on bNAbs against HIV-1 in terms of their interactions with the sites of vulnerability on HIV-1 glycoproteins.
Collapse
Affiliation(s)
- D. N. Shcherbakov
- State research center of virology and biotechnology “Vector”, Koltsovo, 630559, Novosibirsk region, Russia
- Altai State University, 61 Lenin St., 656049, Barnaul, Russia
| | - A. Y. Bakulina
- State research center of virology and biotechnology “Vector”, Koltsovo, 630559, Novosibirsk region, Russia
- Novosibirsk State University, 2 Pirogova St., 630090, Novosibirsk, Russia
| | - L. I. Karpenko
- State research center of virology and biotechnology “Vector”, Koltsovo, 630559, Novosibirsk region, Russia
| | - A. A. Ilyichev
- State research center of virology and biotechnology “Vector”, Koltsovo, 630559, Novosibirsk region, Russia
| |
Collapse
|
30
|
Characterization of the Determinants of NS2-3-Independent Virion Morphogenesis of Pestiviruses. J Virol 2015; 89:11668-80. [PMID: 26355097 DOI: 10.1128/jvi.01646-15] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2015] [Accepted: 09/04/2015] [Indexed: 01/09/2023] Open
Abstract
UNLABELLED A peculiarity of the Flaviviridae is the critical function of nonstructural (NS) proteins for virus particle formation. For pestiviruses, like bovine viral diarrhea virus (BVDV), uncleaved NS2-3 represents an essential factor for virion morphogenesis, while NS3 is an essential component of the viral replicase. Accordingly, in natural pestivirus isolates, processing at the NS2-3 cleavage site is not complete, to allow for virion morphogenesis. Virion morphogenesis of the related hepatitis C virus (HCV) shows a major deviation from that of pestiviruses: while RNA replication also requires free NS3, virion formation does not depend on uncleaved NS2-NS3. Recently, we described a BVDV-1 chimera based on strain NCP7 encompassing the NS2-4B*-coding region of strain Osloss (E. Lattwein, O. Klemens, S. Schwindt, P. Becher, and N. Tautz, J Virol 86:427-437, 2012, doi:10.1128/JVI.06133-11). This chimera allowed for the production of infectious virus particles in the absence of uncleaved NS2-3. The Osloss sequence deviates in the NS2-4B* part from NCP7 in 48 amino acids and also has a ubiquitin insertion between NS2 and NS3. The present study demonstrates that in the NCP7 backbone, only two amino acid exchanges in NS2 (E1576V) and NS3 (V1721A) are sufficient and necessary to allow for efficient NS2-3-independent virion morphogenesis. The adaptation of a bicistronic virus encompassing an internal ribosomal entry site element between the NS2 and NS3 coding sequences to efficient virion morphogenesis led to the identification of additional amino acids in E2, NS2, and NS5B that are critically involved in this process. The surprisingly small requirements for approximating the packaging schemes of pestiviruses and HCV with respect to the NS2-3 region is in favor of a common mechanism in an ancestral virus. IMPORTANCE For positive-strand RNA viruses, the processing products of the viral polyprotein serve in RNA replication as well as virion morphogenesis. For bovine viral diarrhea virus, nonstructural protein NS2-3 is of critical importance to switch between these processes. While free NS3 is essential for RNA replication, uncleaved NS2-3, which accumulates over time in the infected cell, is required for virion morphogenesis. In contrast, the virion morphogenesis of the related hepatitis C virus is independent from uncleaved NS2-NS3. Here, we demonstrate that pestiviruses can adapt to virion morphogenesis in the absence of uncleaved NS2-3 by just two amino acid exchanges. While the mechanism behind this gain of function remains elusive, the fact that it can be achieved by such minor changes is in line with the assumption that an ancestral virus already used this mechanism but lost it in the course of adapting to a new host/infection strategy.
Collapse
|
31
|
Sepúlveda-Crespo D, Sánchez-Rodríguez J, Serramía MJ, Gómez R, De La Mata FJ, Jiménez JL, Muñoz-Fernández MÁ. Triple combination of carbosilane dendrimers, tenofovir and maraviroc as potential microbicide to prevent HIV-1 sexual transmission. Nanomedicine (Lond) 2015; 10:899-914. [PMID: 25867856 DOI: 10.2217/nnm.14.79] [Citation(s) in RCA: 40] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/17/2023] Open
Abstract
AIM To research the synergistic activity by triple combinations of carbosilane dendrimers with tenofovir and maraviroc as topical microbicide. METHODS Cytotoxicity, anti-HIV-1 activity, vaginal irritation and histological analysis of triple combinations were determined. Analysis of combined effects and the median effective concentration were performed using CalcuSyn software. RESULTS Combinations showed a greater broad-spectrum anti-HIV-1 activity than the single-drug, and preserved this activity in acid environment or seminal fluid. The strongest combinations were G2-STE16/G2-S24P/tenofovir, G2-STE16/G2-S16/maraviroc and G2-STE16/tenofovir/maraviroc at 2:2:1, 10:10:1 10:5:1 ratios, respectively. They demonstrated strong synergistic activity profile due to the weighted average combination indices varied between 0.06 and 0.38. No irritation was detected in female BALB/c mice. CONCLUSION The three-drug combination increases their antiviral potency and act synergistically as potential microbicide.
Collapse
Affiliation(s)
- Daniel Sepúlveda-Crespo
- Laboratorio InmunoBiología Molecular, Hospital General Universitario Gregorio Marañón, Instituto de Investigación Sanitaria Gregorio Marañón, Spanish HIV-HGM Biobank, Networking Research Center on Bioengineering, Biomaterials & Nanomedicine (CIBER-BBN), Madrid, Spain
| | | | | | | | | | | | | |
Collapse
|
32
|
Konstantoulas CJ, Lamp B, Rumenapf TH, Indik S. Single amino acid substitution (G42E) in the receptor binding domain of mouse mammary tumour virus envelope protein facilitates infection of non-murine cells in a transferrin receptor 1-independent manner. Retrovirology 2015; 12:43. [PMID: 25980759 PMCID: PMC4445801 DOI: 10.1186/s12977-015-0168-2] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2015] [Accepted: 04/20/2015] [Indexed: 12/11/2022] Open
Abstract
Background Mouse mammary tumour virus (MMTV) is a betaretrovirus that infects rodent cells and uses mouse tranferrin receptor 1 (TfR1) for cell entry. Several MMTV strains have been shown to productively infect, in addition to murine cells, various heterologous cell lines including those of human origin, albeit less efficiently than murine cells. Furthermore, there have been reports that the continued passage of MMTV in heterologous cell lines gives rise to novel variants that are able to infect naive non-murine cells with higher efficiency than the parental virus. Results We show that MMTV(C3H), like other MMTV strains, that had undergone a number of replication cycles in non-murine cells displayed an increased replication kinetic, as compared to parental virus, when applied on naive human cells. Sequence analysis of several replication kinetic variants and the parental virus, together with calculation of the ratio of non-synonymous to synonymous mutations at individual codons, revealed that several regions within the viral genome were under strong positive selection pressure during viral replication in human cells. The mutation responsible, at least in part, for the phenotypic change was subsequently mapped to the segment of env encoding the receptor binding site (F40HGFR44). Introduction of the identified mutation, leading to single amino acid substitution (G42E), into egfp-containing recombinant MMTV virions enhanced their ability to bind to and infect human cells. Interestingly, neither the replication kinetic mutant nor the parental virus required human TfR1 for infection. Knock-out of TFR1 gene from the human genome did not decrease the susceptibility of Hs578T cells to virus infection. Furthermore, the expression of human TfR1, in contrast to mouse TfR1, did not enhance the susceptibility of MMTV-resistant Chinese hamster ovary cells. Thus, human TfR1 is dispensable for infection and another cell surface molecule mediates the MMTV entry into human cells. Conclusion Taken together, our data explain the mechanism enabling MMTV to form ‘host-range variants’ in non-murine cells that has been known for a long time, the basis of which remained obscure. Our findings may expand our understanding of how viruses gain capability to cross species-specific barriers to infect new hosts. Electronic supplementary material The online version of this article (doi:10.1186/s12977-015-0168-2) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
| | - Benjamin Lamp
- Institute of Virology, University of Veterinary Medicine, Veterinaerplatz 1, 1210, Vienna, Austria.
| | - Tillman Hans Rumenapf
- Institute of Virology, University of Veterinary Medicine, Veterinaerplatz 1, 1210, Vienna, Austria.
| | - Stanislav Indik
- Institute of Virology, University of Veterinary Medicine, Veterinaerplatz 1, 1210, Vienna, Austria.
| |
Collapse
|
33
|
Thuy TTT, Ly BM, Van TTT, Quang NV, Tu HC, Zheng Y, Seguin-Devaux C, Mi B, Ai U. Anti-HIV activity of fucoidans from three brown seaweed species. Carbohydr Polym 2015; 115:122-8. [PMID: 25439876 DOI: 10.1016/j.carbpol.2014.08.068] [Citation(s) in RCA: 82] [Impact Index Per Article: 8.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2014] [Revised: 08/13/2014] [Accepted: 08/17/2014] [Indexed: 12/20/2022]
Abstract
Fucoidans are sulfated polysaccharides derived from marine brown algae. In the current work the anti-HIV activity of three fucoidans, extracted from three brown seaweeds Sargassum mcclurei, Sargassum polycystum and Turbinara ornata and collected from Nha Trang bay, Vietnam was investigated. Fucoidans extracted from the three species displayed similar antiviral activities with a mean IC50 ranging from 0.33 to 0.7 μg/ml while displaying no cell toxicity. Our results showed that the anti-HIV activity of fucoidans is not primarily linked to the sulfate content and the appropriate position of sulfate groups in the fucoidan backbones was also not associated with the antiviral activity. Fucoidans inhibited HIV-1 infection when they were pre-incubated with the virus but not with the cells, and not after infection, blocking the early steps of HIV entry into target cells. These data contribute to a better understanding of the influence of fucoidans structural characteristics on their biological activity.
Collapse
Affiliation(s)
- Thanh Thi Thu Thuy
- Institute of Chemistry, Vietnam Academy of Science and Technology, Viet Nam.
| | - Bui Minh Ly
- Nha Trang Institute of Technology Research and Application, Vietnam Academy of Science and Technology, Viet Nam
| | - Tran Thi Thanh Van
- Nha Trang Institute of Technology Research and Application, Vietnam Academy of Science and Technology, Viet Nam
| | - Ngo Van Quang
- Institute of Chemistry, Vietnam Academy of Science and Technology, Viet Nam
| | - Ho Cam Tu
- Laboratory of Retrovirology, CRP-Santé, Luxembourg
| | - Yue Zheng
- Laboratory of Retrovirology, CRP-Santé, Luxembourg
| | | | - Bilan Mi
- N.D.Zelinsky Institute of Organic Chemistry, Russian Academy of Sciences, Leninskii Prospect 47, Moscow 119991, Russian Federation
| | - Usov Ai
- N.D.Zelinsky Institute of Organic Chemistry, Russian Academy of Sciences, Leninskii Prospect 47, Moscow 119991, Russian Federation
| |
Collapse
|
34
|
Pemmaraju BP, Malekar S, Agarwal HK, Tiwari RK, Oh D, Doncel GF, Worthen DR, Parang K. Design, synthesis, antiviral activity, and pre-formulation development of poly-L-arginine-fatty acyl derivatives of nucleoside reverse transcriptase inhibitors. NUCLEOSIDES, NUCLEOTIDES & NUCLEIC ACIDS 2015; 34:1-15. [PMID: 25513860 PMCID: PMC4269296 DOI: 10.1080/15257770.2014.945649] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/24/2022]
Abstract
The objective of this work was to design conjugates of anti-HIV nucleosides conjugated with fatty acids and cell-penetrating poly-L-arginine (polyArg) peptides. Three conjugates of polyArg cell-penetrating peptides with fatty acyl derivatives of alovudine (FLT), lamivudine (3TC), and emtricitabine (FTC) were synthesized. In general, the compounds exhibited anti-HIV activity against X4 and R5 cell-free virus with EC50 values of 1.5-16.6 μM. FLT-CO-(CH2)12-CO-(Arg)7 exhibited EC50 values of 2.9 μM and 3.1 μM against X4 and R5 cell-free virus, respectively. The FLT conjugate was selected for further preformulation studies by determination of solution state degradation and lipid solubility. The compound was found to be stable in neutral and oxidative conditions and moderately stable in heated conditions.
Collapse
Affiliation(s)
- Bhanu P. Pemmaraju
- Department of Biomedical and Pharmaceutical Sciences, College of Pharmacy, University of Rhode Island, Kingston, RI 02881, United States
| | - Swapnil Malekar
- Department of Biomedical and Pharmaceutical Sciences, College of Pharmacy, University of Rhode Island, Kingston, RI 02881, United States
| | - Hitesh K. Agarwal
- Department of Biomedical and Pharmaceutical Sciences, College of Pharmacy, University of Rhode Island, Kingston, RI 02881, United States
| | - Rakesh K. Tiwari
- Department of Biomedical and Pharmaceutical Sciences, College of Pharmacy, University of Rhode Island, Kingston, RI 02881, United States
- School of Pharmacy, Chapman University, Irvine, CA 92618, United States
| | - Donghoon Oh
- Department of Biomedical and Pharmaceutical Sciences, College of Pharmacy, University of Rhode Island, Kingston, RI 02881, United States
| | - Gustavo F. Doncel
- CONRAD, Department of Obstetrics and Gynecology, Eastern Virginia Medical School, Norfolk, VA, USA 23507
| | - David R. Worthen
- Department of Biomedical and Pharmaceutical Sciences, College of Pharmacy, University of Rhode Island, Kingston, RI 02881, United States
| | - Keykavous Parang
- Department of Biomedical and Pharmaceutical Sciences, College of Pharmacy, University of Rhode Island, Kingston, RI 02881, United States
- School of Pharmacy, Chapman University, Irvine, CA 92618, United States
- Chao Family Comprehensice Cancer Center, School of Medicine, University of California, Irvine, Shanbrom Hall, 101 The City Drive, Orange, CA 92868 (USA)
| |
Collapse
|
35
|
Costantini LM, Irvin SC, Kennedy SC, Guo F, Goldstein H, Herold BC, Snapp EL. Engineering and exploitation of a fluorescent HIV-1 gp120 for live cell CD4 binding assays. Virology 2014; 476:240-248. [PMID: 25555152 DOI: 10.1016/j.virol.2014.12.019] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2014] [Revised: 10/08/2014] [Accepted: 12/09/2014] [Indexed: 10/24/2022]
Abstract
The HIV-1 envelope glycoprotein, gp120, binds the host cell receptor, CD4, in the initial step of HIV viral entry and infection. This process is an appealing target for the development of inhibitory drugs and neutralizing antibodies. To study gp120 binding and intracellular trafficking, we engineered a fluorescent fusion of the humanized gp120 JRFL HIV-1 variant and GFP. Gp120-sfGFP is glycosylated with human sugars, robustly expressed, and secreted from cultured human cells. Protein dynamics, quality control, and trafficking can be visualized in live cells. The fusion protein can be readily modified with different gp120 variants or fluorescent proteins. Finally, secreted gp120-sfGFP enables a sensitive and easy binding assay that can quantitatively screen potential inhibitors of gp120-CD4 binding on live cells via fluorescence imaging or laser scanning cytometry. This adaptable research tool should aid in studies of gp120 cell biology and the development of novel anti-HIV drugs.
Collapse
Affiliation(s)
- Lindsey M Costantini
- Department of Anatomy and Structural Biology, Albert Einstein College of Medicine, 1300 Morris Park Avenue, Bronx, NY 10461, USA
| | - Susan C Irvin
- Department of Pediatrics, Albert Einstein College of Medicine, 1300 Morris Park Avenue, Bronx, NY 10461, USA; Department of Microbiology and Immunology, Albert Einstein College of Medicine, 1300 Morris Park Avenue, Bronx, NY 10461, USA
| | - Steven C Kennedy
- Department of Pediatrics, Albert Einstein College of Medicine, 1300 Morris Park Avenue, Bronx, NY 10461, USA
| | - Feng Guo
- Department of Anatomy and Structural Biology, Albert Einstein College of Medicine, 1300 Morris Park Avenue, Bronx, NY 10461, USA
| | - Harris Goldstein
- Department of Pediatrics, Albert Einstein College of Medicine, 1300 Morris Park Avenue, Bronx, NY 10461, USA; Department of Microbiology and Immunology, Albert Einstein College of Medicine, 1300 Morris Park Avenue, Bronx, NY 10461, USA
| | - Betsy C Herold
- Department of Pediatrics, Albert Einstein College of Medicine, 1300 Morris Park Avenue, Bronx, NY 10461, USA; Department of Microbiology and Immunology, Albert Einstein College of Medicine, 1300 Morris Park Avenue, Bronx, NY 10461, USA
| | - Erik L Snapp
- Department of Anatomy and Structural Biology, Albert Einstein College of Medicine, 1300 Morris Park Avenue, Bronx, NY 10461, USA.
| |
Collapse
|
36
|
Hu QY, Fink E, Grant CK, Elder JH. Selective interaction of heparin with the variable region 3 within surface glycoprotein of laboratory-adapted feline immunodeficiency virus. PLoS One 2014; 9:e115252. [PMID: 25521480 PMCID: PMC4270745 DOI: 10.1371/journal.pone.0115252] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2014] [Accepted: 11/20/2014] [Indexed: 12/22/2022] Open
Abstract
Heparan sulfate proteoglycans (HSPG) can act as binding receptors for certain laboratory-adapted (TCA) strains of feline immunodeficiency virus (FIV) and human immunodeficiency virus (HIV). Heparin, a soluble heparin sulfate (HS), can inhibit TCA HIV and FIV entry mediated by HSPG interaction in vitro. In the present study, we further determined the selective interaction of heparin with the V3 loop of TCA of FIV. Our current results indicate that heparin selectively inhibits infection by TCA strains, but not for field isolates (FS). Heparin also specifically interferes with TCA surface glycoprotein (SU) binding to CXCR4, by interactions with HSPG binding sites on the V3 loop of the FIV envelope protein. Peptides representing either the N- or C-terminal side of the V3 loop and containing HSPG binding sites were able to compete away the heparin block of TCA SU binding to CXCR4. Heparin does not interfere with the interaction of SU with anti-V3 antibodies that target the CXCR4 binding region or with the interaction between FS FIV and anti-V3 antibodies since FS SU has no HSPG binding sites within the HSPG binding region. Our data show that heparin blocks TCA FIV infection or entry not only through its competition of HSPG on the cell surface interaction with SU, but also by its interference with CXCR4 binding to SU. These studies aid in the design and development of heparin derivatives or analogues that can inhibit steps in virus infection and are informative regarding the HSPG/SU interaction.
Collapse
Affiliation(s)
- Qiong-Ying Hu
- School of Medicine, Taizhou University, Taizhou, Zhejiang, China
- Department of Immunology and Microbial Science, The Scripps Research Institute, La Jolla, California, United States
| | - Elizabeth Fink
- Department of Immunology and Microbial Science, The Scripps Research Institute, La Jolla, California, United States
| | - Chris K. Grant
- Custom Monoclonals International, Inc., W. Sacramento, California, United States
| | - John H. Elder
- Department of Immunology and Microbial Science, The Scripps Research Institute, La Jolla, California, United States
- * E-mail:
| |
Collapse
|
37
|
Yamada H, Nagao C, Haredy AM, Mori Y, Mizuguchi K, Yamanishi K, Okamoto S. Dextran sulfate-resistant A/Puerto Rico/8/34 influenza virus is associated with the emergence of specific mutations in the neuraminidase glycoprotein. Antiviral Res 2014; 111:69-77. [PMID: 25234090 DOI: 10.1016/j.antiviral.2014.09.002] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2014] [Revised: 09/02/2014] [Accepted: 09/07/2014] [Indexed: 11/28/2022]
Abstract
Dextran sulfate (DS) is a negatively charged sulfated polysaccharide that suppresses the replication of influenza A viruses. The suppression was thought to be associated with inhibition of the hemagglutinin-dependent fusion activity. However, we previously showed that suppression by DS was observed not only at the initial stage of viral infection, but also later when virus is released from infected cells due to inhibition of neuraminidase (NA) activity. In the present study, we isolated DS-resistant A/Puerto Rico/8/34 (PR8) influenza viruses and analyzed the inhibition by DS. We found six mutations in NA genes of five independent resistant PR8 viruses and each resistant NA gene had two mutations. All mutations were from basic to acidic or neutral amino acids. In addition, R430L, K432E or K435E in the 430-435 region was a common mutation in all resistant NA genes. To determine which amino acid(s) are responsible for this resistance, a panel of recombinant viruses containing a PR8 and A/WSN/33(WSN) chimeric NA gene or an NA gene with different mutation(s) was generated using reverse genetics. Using recombinant viruses containing a PR8/WSN chimeric NA, we showed that one third of the C-terminal region of PR8 NA was responsible for DS-sensitivity. Recombinant viruses with a single mutation in NA replicated better than wild-type PR8 in the presence of DS, but were still DS-sensitive. However, replication of recombinant viruses with double mutations from the resistant viruses was not affected by the presence or absence of DS. In addition, resistant recombinant viruses were found to be sensitive to the NA inhibitor, oseltamivir and the oseltamivir-resistant recombinant virus was sensitive to DS. These results suggested that DS is an NA inhibitor with a different mechanism of action from the currently used NA inhibitors and that DS could be used in combination with these inhibitors to treat influenza virus infections.
Collapse
Affiliation(s)
- Hiroshi Yamada
- Laboratory of Virology and Vaccinology, Division of Biomedical Research, National Institute of Biomedical Innovation, Ibaraki, Osaka, Japan.
| | - Chioko Nagao
- National Institute of Biomedical Innovation, Ibaraki, Osaka, Japan
| | - Ahmad M Haredy
- Laboratory of Virology and Vaccinology, Division of Biomedical Research, National Institute of Biomedical Innovation, Ibaraki, Osaka, Japan
| | - Yasuko Mori
- Division of Clinical Virology, Kobe University Graduate School of Medicine, Kobe, Japan
| | - Kenji Mizuguchi
- National Institute of Biomedical Innovation, Ibaraki, Osaka, Japan
| | - Koichi Yamanishi
- Laboratory of Virology and Vaccinology, Division of Biomedical Research, National Institute of Biomedical Innovation, Ibaraki, Osaka, Japan
| | - Shigefumi Okamoto
- Laboratory of Virology and Vaccinology, Division of Biomedical Research, National Institute of Biomedical Innovation, Ibaraki, Osaka, Japan
| |
Collapse
|
38
|
Savage AM, Li Y, Matolyak LE, Doncel GF, Turner SR, Gandour RD. Anti-HIV Activities of Precisely Defined, Semirigid, Carboxylated Alternating Copolymers. J Med Chem 2014; 57:6354-63. [DOI: 10.1021/jm401913w] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Affiliation(s)
- Alice M. Savage
- Department
of Chemistry MC0212 and Macromolecules and Interfaces Institute MC0344, Virginia Tech, Blacksburg, Virginia 24061, United States
| | - Yi Li
- Department
of Chemistry MC0212 and Macromolecules and Interfaces Institute MC0344, Virginia Tech, Blacksburg, Virginia 24061, United States
| | - Lindsay E. Matolyak
- Department
of Chemistry MC0212 and Macromolecules and Interfaces Institute MC0344, Virginia Tech, Blacksburg, Virginia 24061, United States
| | - Gustavo F. Doncel
- CONRAD, Eastern Virginia Medical School, 601 Colley Avenue, Norfolk, Virginia 23507, United States
| | - S. Richard Turner
- Department
of Chemistry MC0212 and Macromolecules and Interfaces Institute MC0344, Virginia Tech, Blacksburg, Virginia 24061, United States
| | - Richard D. Gandour
- Department
of Chemistry MC0212 and Macromolecules and Interfaces Institute MC0344, Virginia Tech, Blacksburg, Virginia 24061, United States
- Virginia
Tech Center for Drug Discovery, Virginia Tech, Blacksburg, Virginia 24061, United States
| |
Collapse
|
39
|
Entry Inhibitors of Human Immunodeficiency Virus. Antiviral Res 2014. [DOI: 10.1128/9781555815493.ch2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022]
|
40
|
Helfer M, Koppensteiner H, Schneider M, Rebensburg S, Forcisi S, Müller C, Schmitt-Kopplin P, Schindler M, Brack-Werner R. The root extract of the medicinal plant Pelargonium sidoides is a potent HIV-1 attachment inhibitor. PLoS One 2014; 9:e87487. [PMID: 24489923 PMCID: PMC3906173 DOI: 10.1371/journal.pone.0087487] [Citation(s) in RCA: 60] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2013] [Accepted: 12/28/2013] [Indexed: 12/21/2022] Open
Abstract
Global HIV-1 treatment would benefit greatly from safe herbal medicines with scientifically validated novel anti-HIV-1 activities. The root extract from the medicinal plant Pelargonium sidoides (PS) is licensed in Germany as the herbal medicine EPs®7630, with numerous clinical trials supporting its safety in humans. Here we provide evidence from multiple cell culture experiments that PS extract displays potent anti-HIV-1 activity. We show that PS extract protects peripheral blood mononuclear cells and macrophages from infection with various X4 and R5 tropic HIV-1 strains, including clinical isolates. Functional studies revealed that the extract from PS has a novel mode-of-action. It interferes directly with viral infectivity and blocks the attachment of HIV-1 particles to target cells, protecting them from virus entry. Analysis of the chemical footprint of anti-HIV activity indicates that HIV-1 inhibition is mediated by multiple polyphenolic compounds with low cytotoxicity and can be separated from other extract components with higher cytotoxicity. Based on our data and its excellent safety profile, we propose that PS extract represents a lead candidate for the development of a scientifically validated herbal medicine for anti-HIV-1 therapy with a mode-of-action different from and complementary to current single-molecule drugs.
Collapse
Affiliation(s)
- Markus Helfer
- Institute of Virology, Helmholtz Zentrum München - German Research Center for Environmental Health, Neuherberg, Germany
| | - Herwig Koppensteiner
- Institute of Virology, Helmholtz Zentrum München - German Research Center for Environmental Health, Neuherberg, Germany
| | - Martha Schneider
- Institute of Virology, Helmholtz Zentrum München - German Research Center for Environmental Health, Neuherberg, Germany
| | - Stephanie Rebensburg
- Institute of Virology, Helmholtz Zentrum München - German Research Center for Environmental Health, Neuherberg, Germany
| | - Sara Forcisi
- Research Unit Analytical BioGeoChemistry, Helmholtz Zentrum München - German Research Center for Environmental Health, Neuherberg, Germany
| | - Constanze Müller
- Research Unit Analytical BioGeoChemistry, Helmholtz Zentrum München - German Research Center for Environmental Health, Neuherberg, Germany
| | - Philippe Schmitt-Kopplin
- Research Unit Analytical BioGeoChemistry, Helmholtz Zentrum München - German Research Center for Environmental Health, Neuherberg, Germany
| | - Michael Schindler
- Institute of Virology, Helmholtz Zentrum München - German Research Center for Environmental Health, Neuherberg, Germany
| | - Ruth Brack-Werner
- Institute of Virology, Helmholtz Zentrum München - German Research Center for Environmental Health, Neuherberg, Germany
- * E-mail:
| |
Collapse
|
41
|
Arnáiz E, Vacas-Córdoba E, Galán M, Pion M, Gómez R, Muñoz-Fernández MAÁ, de la Mata FJ. Synthesis of anionic carbosilane dendrimers via “click chemistry” and their antiviral properties against HIV. ACTA ACUST UNITED AC 2014. [DOI: 10.1002/pola.27090] [Citation(s) in RCA: 31] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/06/2023]
Affiliation(s)
- Eduardo Arnáiz
- Departamento de Química Inorgánica; Universidad de Alcalá, Campus Universitario; E-28871 Alcalá de Henares Spain
- Networking Research Center on Bioengineering; Biomaterials and Nanomedicine (CIBER-BBN); Spain
| | - Enrique Vacas-Córdoba
- Networking Research Center on Bioengineering; Biomaterials and Nanomedicine (CIBER-BBN); Spain
- Laboratorio de Inmunobiología Molecular; Hospital General Universitario Gregorio Marañón and Instituto de Investigación Sanitaria Gregorio Marañón; E-28007 Madrid Spain
| | - Marta Galán
- Departamento de Química Inorgánica; Universidad de Alcalá, Campus Universitario; E-28871 Alcalá de Henares Spain
- Networking Research Center on Bioengineering; Biomaterials and Nanomedicine (CIBER-BBN); Spain
| | - Marjorie Pion
- Networking Research Center on Bioengineering; Biomaterials and Nanomedicine (CIBER-BBN); Spain
- Laboratorio de Inmunobiología Molecular; Hospital General Universitario Gregorio Marañón and Instituto de Investigación Sanitaria Gregorio Marañón; E-28007 Madrid Spain
| | - Rafael Gómez
- Departamento de Química Inorgánica; Universidad de Alcalá, Campus Universitario; E-28871 Alcalá de Henares Spain
- Networking Research Center on Bioengineering; Biomaterials and Nanomedicine (CIBER-BBN); Spain
| | - MA Ángeles Muñoz-Fernández
- Networking Research Center on Bioengineering; Biomaterials and Nanomedicine (CIBER-BBN); Spain
- Laboratorio de Inmunobiología Molecular; Hospital General Universitario Gregorio Marañón and Instituto de Investigación Sanitaria Gregorio Marañón; E-28007 Madrid Spain
| | - F. Javier de la Mata
- Departamento de Química Inorgánica; Universidad de Alcalá, Campus Universitario; E-28871 Alcalá de Henares Spain
- Networking Research Center on Bioengineering; Biomaterials and Nanomedicine (CIBER-BBN); Spain
| |
Collapse
|
42
|
Dürr R, Keppler O, Christ F, Crespan E, Garbelli A, Maga G, Dietrich U. Targeting Cellular Cofactors in HIV Therapy. TOPICS IN MEDICINAL CHEMISTRY 2014. [DOI: 10.1007/7355_2014_45] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/12/2023]
|
43
|
Connell BJ, Lortat-Jacob H. Human immunodeficiency virus and heparan sulfate: from attachment to entry inhibition. Front Immunol 2013; 4:385. [PMID: 24312095 PMCID: PMC3834540 DOI: 10.3389/fimmu.2013.00385] [Citation(s) in RCA: 89] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2013] [Accepted: 11/05/2013] [Indexed: 11/13/2022] Open
Abstract
By targeting cells that provide protection against infection, HIV-1 causes acquired immunodeficiency syndrome. Infection starts when gp120, the viral envelope glycoprotein, binds to CD4 and to a chemokine receptor usually CCR5 or CXCR4. As many microorganisms, HIV-1 also interacts with heparan sulfate (HS), a complex group of cell surface associated anionic polysaccharides. It has been thought that this binding, occurring at a step prior to CD4 recognition, increases infectivity by pre-concentrating the virion particles at the cell surface. Early work, dating from before the identification of CCR5 and CXCR4, showed that a variety of HS mimetics bind to the gp120 V3 loop through electrostatic interactions, compete with cell surface associated HS to bind the virus and consequently, neutralize the infectivity of a number of T-cell line-adapted HIV-1 strains. However, progress made to better understand HIV-1 attachment and entry, coupled with the recent identification of additional gp120 regions mediating HS recognition, have considerably modified this view. Firstly, the V3 loop from CXCR4-using viruses is much more positively charged compared to those using CCR5. HS inhibition of cell attachment is thus restricted to CXCR4-using viruses (such as T-cell line-adapted HIV-1). Secondly, studies aiming at characterizing the gp120/HS complex revealed that HS binding was far more complex than previously thought: in addition to the V3 loop of CXCR4 tropic gp120, HS interacts with several other cryptic areas of the protein, which can be induced upon CD4 binding, and are conserved amongst CCR5 and CXCR4 viruses. In view of these data, this review will detail the present knowledge on HS binding to HIV-1, with regards to attachment and entry processes. It will discuss the perspective of targeting the gp120 co-receptor binding site with HS mimetic compounds, a strategy that recently gave rise to entry inhibitors that work in the low nanomolar range, independently of co-receptor usage.
Collapse
Affiliation(s)
- Bridgette J Connell
- University of Grenoble Alpes, Institut de Biologie Structurale , Grenoble , France ; Centre National de la Recherche Scientifique, Institut de Biologie Structurale , Grenoble , France ; Commissariat à l'Énergie Atomique, Direction des Sciences du Vivant, Institut de Biologie Structurale , Grenoble , France
| | | |
Collapse
|
44
|
Bon I, Lembo D, Rusnati M, Clò A, Morini S, Miserocchi A, Bugatti A, Grigolon S, Musumeci G, Landolfo S, Re MC, Gibellini D. Peptide-derivatized SB105-A10 dendrimer inhibits the infectivity of R5 and X4 HIV-1 strains in primary PBMCs and cervicovaginal histocultures. PLoS One 2013; 8:e76482. [PMID: 24116111 PMCID: PMC3792046 DOI: 10.1371/journal.pone.0076482] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2013] [Accepted: 08/30/2013] [Indexed: 11/19/2022] Open
Abstract
Peptide dendrimers are a class of molecules that exhibit a large array of biological effects including antiviral activity. In this report, we analyzed the antiviral activity of the peptide-derivatized SB105-A10 dendrimer, which is a tetra-branched dendrimer synthetized on a lysine core, in activated peripheral blood mononuclear cells (PBMCs) that were challenged with reference and wild-type human immunodeficiency virus type 1 (HIV-1) strains. SB105-A10 inhibited infections by HIV-1 X4 and R5 strains, interfering with the early phases of the viral replication cycle. SB105-A10 targets heparan sulfate proteoglycans (HSPGs) and, importantly, the surface plasmon resonance (SPR) assay revealed that SB105-A10 strongly binds gp41 and gp120, most likely preventing HIV-1 attachment/entry through multiple mechanisms. Interestingly, the antiviral activity of SB105-A10 was also detectable in an organ-like structure of human cervicovaginal tissue, in which SB105-A10 inhibited the HIV-1ada R5 strain infection without altering the tissue viability. These results demonstrated the strong antiviral activity of SB105-A10 and suggest a potential microbicide use of this dendrimer to prevent the heterosexual transmission of HIV-1.
Collapse
Affiliation(s)
- Isabella Bon
- Department of Experimental, Diagnostic and Specialty Medicine (DIMES), Microbiology Section, University of Bologna, Bologna, Italy
| | - David Lembo
- Department of Clinical and Biological Sciences, University of Torino, Orbassano, Torino, Italy
| | - Marco Rusnati
- Department of Biomedical Sciences and Biotechnology, University of Brescia, Brescia, Italy
| | - Alberto Clò
- Department of Experimental, Diagnostic and Specialty Medicine (DIMES), Microbiology Section, University of Bologna, Bologna, Italy
| | - Silvia Morini
- Department of Experimental, Diagnostic and Specialty Medicine (DIMES), Microbiology Section, University of Bologna, Bologna, Italy
| | - Anna Miserocchi
- Department of Experimental, Diagnostic and Specialty Medicine (DIMES), Microbiology Section, University of Bologna, Bologna, Italy
| | - Antonella Bugatti
- Department of Biomedical Sciences and Biotechnology, University of Brescia, Brescia, Italy
| | | | - Giuseppina Musumeci
- Department of Experimental, Diagnostic and Specialty Medicine (DIMES), Microbiology Section, University of Bologna, Bologna, Italy
| | - Santo Landolfo
- Department of Public Health and Microbiology, University of Torino, Torino, Italy
| | - Maria Carla Re
- Department of Experimental, Diagnostic and Specialty Medicine (DIMES), Microbiology Section, University of Bologna, Bologna, Italy
- Interuniversity Consortium, National Institute Biostructure and Biosystems (INBB) Roma, Italy
| | - Davide Gibellini
- Department of Experimental, Diagnostic and Specialty Medicine (DIMES), Microbiology Section, University of Bologna, Bologna, Italy
| |
Collapse
|
45
|
Lian W, Wu M, Huang N, Gao N, Xiao C, Li Z, Zhang Z, Zheng Y, Peng W, Zhao J. Anti-HIV-1 activity and structure–activity-relationship study of a fucosylated glycosaminoglycan from an echinoderm by targeting the conserved CD4 induced epitope. Biochim Biophys Acta Gen Subj 2013; 1830:4681-91. [DOI: 10.1016/j.bbagen.2013.06.003] [Citation(s) in RCA: 42] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2013] [Revised: 05/31/2013] [Accepted: 06/04/2013] [Indexed: 11/25/2022]
|
46
|
Rusnati M, Chiodelli P, Bugatti A, Urbinati C. Bridging the past and the future of virology: surface plasmon resonance as a powerful tool to investigate virus/host interactions. Crit Rev Microbiol 2013; 41:238-60. [PMID: 24059853 DOI: 10.3109/1040841x.2013.826177] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/20/2023]
Abstract
Despite decades of antiviral drug research and development, viruses still remain a top global healthcare problem. Compared to eukaryotic cells, viruses are composed by a limited numbers of proteins that, nevertheless, set up multiple interactions with cellular components, allowing the virus to take control of the infected cell. Each virus/host interaction can be considered as a therapeutical target for new antiviral drugs but, unfortunately, the systematic study of a so huge number of interactions is time-consuming and expensive, calling for models overcoming these drawbacks. Surface plasmon resonance (SPR) is a label-free optical technique to study biomolecular interactions in real time by detecting reflected light from a prism-gold film interface. Launched 20 years ago, SPR has become a nearly irreplaceable technology for the study of biomolecular interactions. Accordingly, SPR is increasingly used in the field of virology, spanning from the study of biological interactions to the identification of putative antiviral drugs. From the literature available, SPR emerges as an ideal link between conventional biological experimentation and system biology studies functional to the identification of highly connected viral or host proteins that act as nodal points in virus life cycle and thus considerable as therapeutical targets for the development of innovative antiviral strategies.
Collapse
Affiliation(s)
- Marco Rusnati
- Department of Molecular and Translational Medicine, University of Brescia , Brescia , Italy
| | | | | | | |
Collapse
|
47
|
Prokofjeva MM, Imbs TI, Shevchenko NM, Spirin PV, Horn S, Fehse B, Zvyagintseva TN, Prassolov VS. Fucoidans as potential inhibitors of HIV-1. Mar Drugs 2013; 11:3000-14. [PMID: 23966033 PMCID: PMC3766878 DOI: 10.3390/md11083000] [Citation(s) in RCA: 49] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2013] [Revised: 06/26/2013] [Accepted: 07/31/2013] [Indexed: 11/17/2022] Open
Abstract
The antiviral activity of different structure fucoidans (α-l-fucans and galactofucans) was studied using two model viral systems based on a lentiviral vectors and a replication competent Moloney murine leukemia virus (Mo-MuLV). It was found that investigated fucoidans have no cytotoxic effects on Jurkat and SC-1cell at the concentration range of 0.001-100 µg/mL. Fucoidans with different efficiency suppressed transduction of Jurkat cell line by pseudo-HIV-1 particles carrying the envelope protein of HIV-1 and infection of SC-1 cells by Mo-MuLV. According to our data, all natural fucoidans can be considered as potential anti-HIV agents regardless of their carbohydrate backbone and degree of sulfating, since their activity is shown at low concentrations (0.001-0.05 µg/mL). High molecular weight fucoidans isolated from Saccharina cichorioides (1.3-α-l-fucan), and S. japonica (galactofucan) were the most effective inhibitors.
Collapse
Affiliation(s)
- Maria M. Prokofjeva
- Laboratory of Cell Biology, Engelhardt-Institute of Molecular Biology, Moscow 119991, Russia; E-Mails: (M.M.P.); (P.V.S.); (V.S.P.)
| | - Tatyana I. Imbs
- Laboratory of Enzyme Chemistry, G.B. Elyakov Pacific Institute of Bioorganic Chemistry, Far Eastern Branch, Russian Academy of Sciences, 159 100-Let Vladivostoku Ave., Vladivostok 690022, Russia; E-Mails: (T.I.I.); (N.M.S.)
| | - Natalya M. Shevchenko
- Laboratory of Enzyme Chemistry, G.B. Elyakov Pacific Institute of Bioorganic Chemistry, Far Eastern Branch, Russian Academy of Sciences, 159 100-Let Vladivostoku Ave., Vladivostok 690022, Russia; E-Mails: (T.I.I.); (N.M.S.)
| | - Pavel V. Spirin
- Laboratory of Cell Biology, Engelhardt-Institute of Molecular Biology, Moscow 119991, Russia; E-Mails: (M.M.P.); (P.V.S.); (V.S.P.)
| | - Stefan Horn
- Research Department of Cell and Gene Therapy, Clinic for Stem Cell Transplantation, UCCH, University Medical Center Hamburg-Eppendorf (UKE), Hamburg D-20246, Germany; E-Mails: (S.H.); (B.F.)
| | - Boris Fehse
- Research Department of Cell and Gene Therapy, Clinic for Stem Cell Transplantation, UCCH, University Medical Center Hamburg-Eppendorf (UKE), Hamburg D-20246, Germany; E-Mails: (S.H.); (B.F.)
| | - Tatyana N. Zvyagintseva
- Laboratory of Enzyme Chemistry, G.B. Elyakov Pacific Institute of Bioorganic Chemistry, Far Eastern Branch, Russian Academy of Sciences, 159 100-Let Vladivostoku Ave., Vladivostok 690022, Russia; E-Mails: (T.I.I.); (N.M.S.)
| | - Vladimir S. Prassolov
- Laboratory of Cell Biology, Engelhardt-Institute of Molecular Biology, Moscow 119991, Russia; E-Mails: (M.M.P.); (P.V.S.); (V.S.P.)
| |
Collapse
|
48
|
Abstract
The human immunodeficiency virus (HIV) enters cells through a series of molecular interactions between the HIV envelope protein and cellular receptors, thus providing many opportunities to block infection. Entry inhibitors are currently being used in the clinic, and many more are under development. Unfortunately, as is the case for other classes of antiretroviral drugs that target later steps in the viral life cycle, HIV can become resistant to entry inhibitors. In contrast to inhibitors that block viral enzymes in intracellular compartments, entry inhibitors interfere with the function of the highly variable envelope glycoprotein as it continuously adapts to changing immune pressure and available target cells in the extracellular environment. Consequently, pathways and mechanisms of resistance for entry inhibitors are varied and often involve mutations across the envelope gene. This review provides a broad overview of entry inhibitor resistance mechanisms that inform our understanding of HIV entry and the design of new inhibitors and vaccines.
Collapse
Affiliation(s)
- Christopher J De Feo
- Office of Vaccine Research and Review, Center for Biologics Evaluation and Research, US Food and Drug Administration, 8800 Rockville Pike, Bethesda, MD 20892, USA.
| | | |
Collapse
|
49
|
Haqqani AA, Tilton JC. Entry inhibitors and their use in the treatment of HIV-1 infection. Antiviral Res 2013; 98:158-70. [PMID: 23541872 DOI: 10.1016/j.antiviral.2013.03.017] [Citation(s) in RCA: 95] [Impact Index Per Article: 7.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2013] [Revised: 03/18/2013] [Accepted: 03/19/2013] [Indexed: 12/20/2022]
Abstract
Entry of HIV into target cells is a complex, multi-stage process involving sequential attachment and CD4 binding, coreceptor binding, and membrane fusion. HIV entry inhibitors are a complex group of drugs with multiple mechanisms of action depending on the stage of the viral entry process they target. Two entry inhibitors are currently approved for the treatment of HIV-infected patients. Maraviroc, a CCR5 antagonist, blocks interactions between the viral envelope proteins and the CCR5 coreceptor. Enfuvirtide, a fusion inhibitor, disrupts conformational changes in gp41 that drive membrane fusion. A wide array of additional agents are in various stages of development. This review covers the entry inhibitors and their use in the treatment of HIV-infected patients.
Collapse
Affiliation(s)
- Aiman A Haqqani
- Case Center for Proteomics and Bioinformatics, Case Western Reserve University, Cleveland, OH 44106, USA
| | | |
Collapse
|
50
|
Tan S, Lu L, Li L, Liu J, Oksov Y, Lu H, Jiang S, Liu S. Polyanionic candidate microbicides accelerate the formation of semen-derived amyloid fibrils to enhance HIV-1 infection. PLoS One 2013; 8:e59777. [PMID: 23544097 PMCID: PMC3609764 DOI: 10.1371/journal.pone.0059777] [Citation(s) in RCA: 33] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2012] [Accepted: 02/18/2013] [Indexed: 12/21/2022] Open
Abstract
Polyanionic candidate microbicides, including cellulose sulfate, carrageenan, PRO 2000, were proven ineffective in preventing HIV-1 transmission and even cellulose sulfate showed increased risk of HIV acquisition in the Phase III efficacy trials. Semen plays critical roles in HIV-1 sexual transmission. Specifically, amyloid fibrils formed by fragments of prostatic acidic phosphatase (PAP) in semen termed semen-derived enhancer of virus infection (SEVI) could drastically enhance HIV-1 infection. Here we investigated the interaction between polyanions and PAP248-286, a prototype peptide of SEVI, to understand the possible cause of polyanionic candidate microbicides to fail in clinical trials. We found anionic polymers could efficiently promote SEVI fibril formation, most likely mediated by the natural electrostatic interaction between polyanions and PAP248-286, as revealed by acid native PAGE and Western blot. The overall anti-HIV-1 activity of polyanions in the presence or absence of PAP248-286 or semen was evaluated. In the viral infection assay, the supernatants of polyanions/PAP248-286 or polyanions/semen mixtures containing the free, unbound polyanionic molecules showed a general reduction in antiviral efficacy, while the pellets containing amyloid fibrils formed by the polyanion-bound PAP248-286 showed aggravated enhancement of viral infection. Collectively, from the point of drug-host protein interaction, our study revealed that polyanions facilitate SEVI fibril formation to promote HIV-1 infection, thus highlighting a molecular mechanism underlying the failure of polyanions in clinical trials and the importance of drug-semen interaction in evaluating the anti-HIV-1 efficacy of candidate microbicides.
Collapse
Affiliation(s)
- Suiyi Tan
- School of Pharmaceutical Sciences, Southern Medical University, Guangzhou, China
- Lindsley F. Kimball Research Institute, New York Blood Center, New York, New York, United States of America
| | - Lu Lu
- Key Laboratory of Medical Molecular Virology of Ministries of Education & Health, Shanghai Medical College and Institute of Medical Microbiology, Fudan University, Shanghai, China
- Lindsley F. Kimball Research Institute, New York Blood Center, New York, New York, United States of America
| | - Lin Li
- School of Pharmaceutical Sciences, Southern Medical University, Guangzhou, China
| | - Jixiang Liu
- Lindsley F. Kimball Research Institute, New York Blood Center, New York, New York, United States of America
| | - Yelena Oksov
- Lindsley F. Kimball Research Institute, New York Blood Center, New York, New York, United States of America
| | - Hong Lu
- Lindsley F. Kimball Research Institute, New York Blood Center, New York, New York, United States of America
| | - Shibo Jiang
- School of Pharmaceutical Sciences, Southern Medical University, Guangzhou, China
- Key Laboratory of Medical Molecular Virology of Ministries of Education & Health, Shanghai Medical College and Institute of Medical Microbiology, Fudan University, Shanghai, China
- Lindsley F. Kimball Research Institute, New York Blood Center, New York, New York, United States of America
- * E-mail: (SJ); (SL)
| | - Shuwen Liu
- School of Pharmaceutical Sciences, Southern Medical University, Guangzhou, China
- * E-mail: (SJ); (SL)
| |
Collapse
|