1
|
Yadav K, Saurav GK, Rana VS, Rawat N, Anjali, Jamwal R, Singh OP, Bandyopadhyay A, Rajagopal R. Polyubiquitin protein of Aedes aegypti as an interacting partner of dengue virus envelope protein. MEDICAL AND VETERINARY ENTOMOLOGY 2024; 38:48-58. [PMID: 37807654 DOI: 10.1111/mve.12696] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/06/2023] [Accepted: 09/07/2023] [Indexed: 10/10/2023]
Abstract
Dengue virus (DENV) is an arbovirus that comprises four antigenically different serotypes. Aedes aegypti (Diptera: Culicidae) acts as the principal vector for DENV transmission, and vector control is crucial for dengue fever epidemic management. To design effective vector control strategies, a comprehensive understanding of the insect vector and virus interaction is required. Female Ae. aegypti ingests DENV during the acquisition of a blood meal from an infected human. DENV enters the insect midgut, replicates inside it and reaches the salivary gland for transmitting DENV to healthy humans during the subsequent feeding cycles. DENV must interact with the proteins present in the midgut and salivary glands to gain entry and accomplish successful replication and transmission. Ae. aegypti midgut cDNA library was prepared, and yeast two-hybrid screening was performed against the envelope protein domain III (EDIII) protein of DENV-2. The polyubiquitin protein was selected from the various candidate proteins for subsequent analysis. Polyubiquitin gene was amplified, and the protein was purified in a heterologous expression system for in vitro interaction studies. In vitro pull-down assay presented a clear interaction between polyubiquitin protein and EDIII. To further confirm this interaction, a dot blot assay was employed, and polyubiquitin protein was found to interact with DENV particles. Our results enable us to suggest that polyubiquitin plays an important role in DENV infection within mosquitoes.
Collapse
Affiliation(s)
- Karuna Yadav
- Gut Biology Laboratory, Department of Zoology, University of Delhi, Delhi, India
| | - Gunjan Kumar Saurav
- Gut Biology Laboratory, Department of Zoology, University of Delhi, Delhi, India
- Department of Zoology, Rajiv Gandhi University, Doimukh, Arunachal Pradesh, India
| | - Vipin Singh Rana
- Gut Biology Laboratory, Department of Zoology, University of Delhi, Delhi, India
- Department of Veterinary Medicine, University of Maryland, College Park, Maryland, USA
| | - Nitish Rawat
- Gut Biology Laboratory, Department of Zoology, University of Delhi, Delhi, India
| | - Anjali
- Gut Biology Laboratory, Department of Zoology, University of Delhi, Delhi, India
| | - Rohit Jamwal
- Gut Biology Laboratory, Department of Zoology, University of Delhi, Delhi, India
| | | | - Anannya Bandyopadhyay
- Protein Homeostasis Laboratory, Department of Zoology, University of Delhi, Delhi, India
| | - Raman Rajagopal
- Gut Biology Laboratory, Department of Zoology, University of Delhi, Delhi, India
| |
Collapse
|
2
|
Munt JE, Henein S, Adams C, Young E, Hou YJ, Conrad H, Zhu D, Dong S, Kose N, Yount B, Meganck RM, Tse LPV, Kuan G, Balmaseda A, Ricciardi MJ, Watkins DI, Crowe JE, Harris E, DeSilva AM, Baric RS. Homotypic antibodies target novel E glycoprotein domains after natural DENV 3 infection/vaccination. Cell Host Microbe 2023; 31:1850-1865.e5. [PMID: 37909048 PMCID: PMC11221912 DOI: 10.1016/j.chom.2023.10.004] [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: 03/01/2023] [Revised: 07/31/2023] [Accepted: 10/04/2023] [Indexed: 11/02/2023]
Abstract
The envelope (E) glycoprotein is the primary target of type-specific (TS) neutralizing antibodies (nAbs) after infection with any of the four distinct dengue virus serotypes (DENV1-4). nAbs can be elicited to distinct structural E domains (EDs) I, II, or III. However, the relative contribution of these domain-specific antibodies is unclear. To identify the primary DENV3 nAb targets in sera after natural infection or vaccination, chimeric DENV1 recombinant encoding DENV3 EDI, EDII, or EDIII were generated. DENV3 EDII is the principal target of TS polyclonal nAb responses and encodes two or more neutralizing epitopes. In contrast, some were individuals vaccinated with a DENV3 monovalent vaccine-elicited serum TS nAbs targeting each ED in a subject-dependent fashion, with an emphasis on EDI and EDIII. Vaccine responses were also sensitive to DENV3 genotypic variation. This DENV1/3 panel allows the measurement of serum ED TS nAbs, revealing differences in TS nAb immunity after natural infection or vaccination.
Collapse
Affiliation(s)
- Jennifer E Munt
- Department of Epidemiology, University of North Carolina, Chapel Hill, NC, USA
| | - Sandra Henein
- Department of Microbiology and Immunology, University of North Carolina, Chapel Hill, NC, USA
| | - Cameron Adams
- Department of Microbiology and Immunology, University of North Carolina, Chapel Hill, NC, USA
| | - Ellen Young
- Department of Epidemiology, University of North Carolina, Chapel Hill, NC, USA
| | - Yixuan J Hou
- Department of Epidemiology, University of North Carolina, Chapel Hill, NC, USA
| | - Helen Conrad
- Department of Epidemiology, University of North Carolina, Chapel Hill, NC, USA
| | - Deanna Zhu
- Department of Epidemiology, University of North Carolina, Chapel Hill, NC, USA
| | - Stephanie Dong
- Department of Epidemiology, University of North Carolina, Chapel Hill, NC, USA
| | - Nurgun Kose
- Vanderbilt Vaccine Center, Nashville, TN, USA
| | - Boyd Yount
- Department of Epidemiology, University of North Carolina, Chapel Hill, NC, USA
| | - Rita M Meganck
- Department of Epidemiology, University of North Carolina, Chapel Hill, NC, USA
| | - Long Ping V Tse
- Department of Epidemiology, University of North Carolina, Chapel Hill, NC, USA
| | - Guillermina Kuan
- Health Center Socrates Flores Vivas, Ministry of Health, Managua, Nicaragua; Sustainable Sciences Institute, Managua, Nicaragua
| | - Angel Balmaseda
- Sustainable Sciences Institute, Managua, Nicaragua; National Virology Laboratory, National Center for Diagnosis and Reference, Ministry of Health, Managua, Nicaragua
| | | | - David I Watkins
- University of Massachusetts Medical School, Worcester, MA, USA
| | | | - Eva Harris
- Division of Infectious Diseases and Vaccinology, School of Public Health, University of California Berkeley, Berkeley, CA, USA
| | - Aravinda M DeSilva
- Department of Microbiology and Immunology, University of North Carolina, Chapel Hill, NC, USA
| | - Ralph S Baric
- Department of Epidemiology, University of North Carolina, Chapel Hill, NC, USA; Department of Microbiology and Immunology, University of North Carolina, Chapel Hill, NC, USA.
| |
Collapse
|
3
|
Modak A, Mishra SR, Awasthi M, Sreedevi S, Sobha A, Aravind A, Kuppusamy K, Sreekumar E. Higher-temperature-adapted dengue virus serotype 2 strain exhibits enhanced virulence in AG129 mouse model. FASEB J 2023; 37:e23062. [PMID: 37389962 DOI: 10.1096/fj.202300098r] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2023] [Revised: 05/13/2023] [Accepted: 06/14/2023] [Indexed: 07/02/2023]
Abstract
The factors that drive dengue virus (DENV) evolution, and selection of virulent variants are yet not clear. Higher environmental temperature shortens DENV extrinsic incubation period in mosquitoes, increases human transmission, and plays a critical role in outbreak dynamics. In the present study, we looked at the effect of temperature in altering the virus virulence. We found that DENV cultured at a higher temperature in C6/36 mosquito cells was significantly more virulent than the virus grown at a lower temperature. In a mouse model, the virulent strain induced enhanced viremia and aggressive disease with a short course, hemorrhage, severe vascular permeability, and death. Higher inflammatory cytokine response, thrombocytopenia, and severe histopathological changes in vital organs such as heart, liver, and kidney were hallmarks of the disease. Importantly, it required only a few passages for the virus to acquire a quasi-species population harboring virulence-imparting mutations. Whole genome comparison with a lower temperature passaged strain identified key genomic changes in the structural protein-coding regions as well as in the 3'UTR of the viral genome. Our results point out that virulence-enhancing genetic changes could occur in the dengue virus genome under enhanced growth temperature conditions in mosquito cells.
Collapse
Affiliation(s)
- Ayan Modak
- Molecular Virology Laboratory, Rajiv Gandhi Centre for Biotechnology (RGCB), Thiruvananthapuram, India
| | - Srishti Rajkumar Mishra
- Molecular Virology Laboratory, Rajiv Gandhi Centre for Biotechnology (RGCB), Thiruvananthapuram, India
| | - Mansi Awasthi
- Molecular Virology Laboratory, Rajiv Gandhi Centre for Biotechnology (RGCB), Thiruvananthapuram, India
| | - Sreeja Sreedevi
- Molecular Virology Laboratory, Rajiv Gandhi Centre for Biotechnology (RGCB), Thiruvananthapuram, India
| | - Archana Sobha
- Animal Research Facility, Rajiv Gandhi Centre for Biotechnology (RGCB), Thiruvananthapuram, India
| | - Arya Aravind
- Animal Research Facility, Rajiv Gandhi Centre for Biotechnology (RGCB), Thiruvananthapuram, India
| | - Krithiga Kuppusamy
- Bioscience Research & Training Centre (BRTC), Kerala Veterinary and Animal Sciences University, Bio360 Life Sciences Park, Thiruvananthapuram, India
| | - Easwaran Sreekumar
- Molecular Virology Laboratory, Rajiv Gandhi Centre for Biotechnology (RGCB), Thiruvananthapuram, India
- Molecular Bioassay Laboratory, Institute of Advanced Virology (IAV), Bio360 Life Sciences Park, Thiruvananthapuram, India
| |
Collapse
|
4
|
Yadav K, Rana VS, Anjali, Saurav GK, Rawat N, Kumar A, Sunil S, Singh OP, Rajagopal R. Mucin Protein of Aedes aegypti Interacts with Dengue Virus 2 and Influences Viral Infection. Microbiol Spectr 2023; 11:e0250322. [PMID: 36847498 PMCID: PMC10101019 DOI: 10.1128/spectrum.02503-22] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2022] [Accepted: 01/26/2023] [Indexed: 03/01/2023] Open
Abstract
Dengue, caused by dengue virus (DENV), is the most prevalent vector-borne viral disease, posing a serious health concern to 2.5 billion people worldwide. DENV is primarily transmitted among humans by its mosquito vector Aedes aegypti; hence, the identification of a novel dengue virus receptor in mosquitoes is critical for the development of new anti-mosquito measures. In the current study, we have identified peptides which potentially interact with the surface of the virion particles and facilitate virus infection and movement during their life cycle in the mosquito vector. To identify these candidate proteins, we performed phage-display library screening against domain III of the envelope protein (EDIII), which plays an essential role during host cell receptor binding for viral entry. The mucin protein, which shared sequence similarity with the peptide identified in the screening, was cloned, expressed, and purified for in vitro interaction studies. Using in vitro pulldown and virus overlay protein-binding assay (VOPBA), we confirmed the positive interaction of mucin with purified EDIII and whole virion particles. Finally, blocking of mucin protein with anti-mucin antibodies partially reduced DENV titers in infected mosquitos. Moreover, mucin protein was found to be localized in the midgut of Ae. aegypti. IMPORTANCE Identification of interacting protein partners of DENV in the insect vector Aedes aegypti is crucial for designing vector control-based strategies and for understanding the molecular mechanism DENV uses to modulate the host, gain entry, and survive successfully. Similar proteins can be used in generating transmission-blocking vaccines.
Collapse
Affiliation(s)
- Karuna Yadav
- Gut Biology Laboratory, Department of Zoology, University of Delhi, New Delhi, India
| | - Vipin Singh Rana
- Gut Biology Laboratory, Department of Zoology, University of Delhi, New Delhi, India
- Department of Veterinary Medicine, University of Maryland, College Park, Maryland, USA
| | - Anjali
- Gut Biology Laboratory, Department of Zoology, University of Delhi, New Delhi, India
| | - Gunjan Kumar Saurav
- Gut Biology Laboratory, Department of Zoology, University of Delhi, New Delhi, India
- Department of Zoology, Rajiv Gandhi University, Doimukh, Arunachal Pradesh, India
| | - Nitish Rawat
- Gut Biology Laboratory, Department of Zoology, University of Delhi, New Delhi, India
| | - Ankit Kumar
- Vector Borne Disease Group, International Centre for Genetic Engineering and Biotechnology, New Delhi, India
| | - Sujatha Sunil
- Vector Borne Disease Group, International Centre for Genetic Engineering and Biotechnology, New Delhi, India
| | - Om P. Singh
- National Institute of Malaria Research, New Delhi, India
| | - Raman Rajagopal
- Gut Biology Laboratory, Department of Zoology, University of Delhi, New Delhi, India
| |
Collapse
|
5
|
Reactivity of DENV-positive sera against recombinant envelope proteins produced in bacteria and eukaryotic cells. Immunol Res 2023; 71:39-50. [PMID: 36192522 DOI: 10.1007/s12026-022-09326-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2022] [Accepted: 09/25/2022] [Indexed: 01/20/2023]
Abstract
Dengue is a mosquito-borne disease endemic in many tropical and subtropical countries. It is caused by the dengue virus (DENV) that can be classified into 4 different serotypes (DENV-1-4). Early diagnosis and management can reduce morbidity and mortality rates of severe forms of the disease, as well as decrease the risk of larger outbreaks. Hiperendemicity in some regions of the world and the possibility that some people develop a more severe form of disease after a secondary infection caused by antibody-dependent enhancement justify the need to understand more thoroughly the antibody response induced against the virus. Here, we successfully produced a recombinant DENV-2 envelope (E) protein and its domains (EDI/II and EDIII) in two distinct expression systems: the Drosophila S2 insect cell system and the BL21 (DE3) pLySs bacterial system. We then evaluated the reactivity of sera from patients previously infected with DENV to each recombinant protein and to each domain separately. Our results show that the E protein produced in Drosophila S2 cells is recognized more frequently than the protein produced in bacteria. However, the recognition of E protein produced in bacteria correlates better with the DENV-2 sera neutralization capacity. The results described here emphasize the differences observed when antigens produced in bacteria or eukaryotic cells are used and may be useful to gain more insight into the humoral immune responses induced by dengue infection.
Collapse
|
6
|
Dos Santos Nascimento IJ, da Silva Rodrigues ÉE, da Silva MF, de Araújo-Júnior JX, de Moura RO. Advances in Computational Methods to Discover New NS2B-NS3 Inhibitors Useful Against Dengue and Zika Viruses. Curr Top Med Chem 2022; 22:2435-2462. [PMID: 36415099 DOI: 10.2174/1568026623666221122121330] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2022] [Revised: 09/20/2022] [Accepted: 09/27/2022] [Indexed: 11/24/2022]
Abstract
The Flaviviridae virus family consists of the genera Hepacivirus, Pestivirus, and Flavivirus, with approximately 70 viral types that use arthropods as vectors. Among these diseases, dengue (DENV) and zika virus (ZIKV) serotypes stand out, responsible for thousands of deaths worldwide. Due to the significant increase in cases, the World Health Organization (WHO) declared DENV a potential threat for 2019 due to being transmitted by infected travelers. Furthermore, ZIKV also has a high rate of transmissibility, highlighted in the outbreak in 2015, generating consequences such as Guillain-Barré syndrome and microcephaly. According to clinical outcomes, those infected with DENV can be asymptomatic, and in other cases, it can be lethal. On the other hand, ZIKV has severe neurological symptoms in newborn babies and adults. More serious symptoms include microcephaly, brain calcifications, intrauterine growth restriction, and fetal death. Despite these worrying data, no drug or vaccine is approved to treat these diseases. In the drug discovery process, one of the targets explored against these diseases is the NS2B-NS3 complex, which presents the catalytic triad His51, Asp75, and Ser135, with the function of cleaving polyproteins, with specificity for basic amino acid residues, Lys- Arg, Arg-Arg, Arg-Lys or Gln-Arg. Since NS3 is highly conserved in all DENV serotypes and plays a vital role in viral replication, this complex is an excellent drug target. In recent years, computer-aided drug discovery (CADD) is increasingly essential in drug discovery campaigns, making the process faster and more cost-effective, mainly explained by discovering new drugs against DENV and ZIKV. Finally, the main advances in computational methods applied to discover new compounds against these diseases will be presented here. In fact, molecular dynamics simulations and virtual screening is the most explored approach, providing several hit and lead compounds that can be used in further optimizations. In addition, fragment-based drug design and quantum chemistry/molecular mechanics (QM/MM) provides new insights for developing anti-DENV/ZIKV drugs. We hope that this review offers further helpful information for researchers worldwide and stimulates the use of computational methods to find a promising drug for treating DENV and ZIKV.
Collapse
Affiliation(s)
- Igor José Dos Santos Nascimento
- Department of Pharmacy, Estácio of Alagoas College, Maceió, Brazil.,Department of Pharmacy, Cesmac University Center, Maceió, Brazil.,Department of Pharmacy, Drug Development and Synthesis Laboratory, State University of Paraíba, Campina Grande, Brazil
| | | | - Manuele Figueiredo da Silva
- Laboratory of Medicinal Chemistry, Pharmaceutical Sciences Institute, Federal University of Alagoas, Maceió, Brazil
| | - João Xavier de Araújo-Júnior
- Laboratory of Medicinal Chemistry, Pharmaceutical Sciences Institute, Federal University of Alagoas, Maceió, Brazil
| | - Ricardo Olimpio de Moura
- Department of Pharmacy, Drug Development and Synthesis Laboratory, State University of Paraíba, Campina Grande, Brazil
| |
Collapse
|
7
|
Mittal A, Chauhan A. Aspects of Biological Replication and Evolution Independent of the Central Dogma: Insights from Protein-Free Vesicular Transformations and Protein-Mediated Membrane Remodeling. J Membr Biol 2022; 255:185-209. [PMID: 35333977 PMCID: PMC8951669 DOI: 10.1007/s00232-022-00230-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2022] [Accepted: 03/06/2022] [Indexed: 11/21/2022]
Abstract
Biological membrane remodeling is central to living systems. In spite of serving as “containers” of whole-living systems and functioning as dynamic compartments within living systems, biological membranes still find a “blue collar” treatment compared to the “white collar” nucleic acids and proteins in biology. This may be attributable to the fact that scientific literature on biological membrane remodeling is only 50 years old compared to ~ 150 years of literature on proteins and a little less than 100 years on nucleic acids. However, recently, evidence for symbiotic origins of eukaryotic cells from data only on biological membranes was reported. This, coupled with appreciation of reproducible amphiphilic self-assemblies in aqueous environments (mimicking replication), has already initiated discussions on origins of life beyond nucleic acids and proteins. This work presents a comprehensive compilation and meta-analyses of data on self-assembly and vesicular transformations in biological membranes—starting from model membranes to establishment of Influenza Hemagglutinin-mediated membrane fusion as a prototypical remodeling system to a thorough comparison between enveloped mammalian viruses and cellular vesicles. We show that viral membrane fusion proteins, in addition to obeying “stoichiometry-driven protein folding”, have tighter compositional constraints on their amino acid occurrences than general-structured proteins, regardless of type/class. From the perspective of vesicular assemblies and biological membrane remodeling (with and without proteins) we find that cellular vesicles are quite different from viruses. Finally, we propose that in addition to pre-existing thermodynamic frameworks, kinetic considerations in de novo formation of metastable membrane structures with available “third-party” constituents (including proteins) were not only crucial for origins of life but also continue to offer morphological replication and/or functional mechanisms in modern life forms, independent of the central dogma.
Collapse
Affiliation(s)
- Aditya Mittal
- Kusuma School of Biological Sciences, Indian Institute of Technology Delhi (IIT Delhi), Hauz Khas, New Delhi, 110016, India. .,Supercomputing Facility for Bioinformatics and Computational Biology (SCFBio), IIT Delhi, Hauz Khas, New Delhi, 110016, India.
| | - Akanksha Chauhan
- Kusuma School of Biological Sciences, Indian Institute of Technology Delhi (IIT Delhi), Hauz Khas, New Delhi, 110016, India
| |
Collapse
|
8
|
Ling J, Li J, Khan A, Lundkvist Å, Li JP. Is heparan sulfate a target for inhibition of RNA virus infection? Am J Physiol Cell Physiol 2022; 322:C605-C613. [PMID: 35196165 PMCID: PMC8977144 DOI: 10.1152/ajpcell.00028.2022] [Citation(s) in RCA: 13] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/23/2023]
Abstract
Heparan sulfate (HS) is a linear polysaccharide attached to a core protein, forming heparan sulfate proteoglycans (HSPGs) that are ubiquitously expressed on the surface of almost all mammalian cells and the extracellular matrix. HS orchestrates the binding of various signal molecules to their receptors, thus, regulating many biological processes, including homeostasis, metabolism, and various pathological processes. Due to its wide distribution and negatively charged properties, HS is exploited by many viruses as a co-factor to attach to host cells. Therefore, inhibition of the interaction between virus and HS is proposed as a promising approach to mitigate viral infection, including SARS-CoV-2. In this review, we summarize the interaction manners of HS with viruses with focus on significant pathogenic RNA viruses, including alphaviruses, flaviviruses, and coronaviruses. We also provide an overview of the challenges we may face when using HS-mimetics as antivirals for clinical treatment. More studies are needed to provide a further understanding of the interplay between HS and viruses both in vitro and in vivo, which will favor the development of specific antiviral inhibitors.
Collapse
Affiliation(s)
- Jiaxin Ling
- Department of Medical Biochemistry and Microbiology & The Biomedical Center; Zoonosis Science Center, University of Uppsala, Uppsala, Sweden.,Zoonosis Science Center, University of Uppsala, Uppsala, Sweden
| | - Jinlin Li
- Department of Medical Biochemistry and Microbiology & The Biomedical Center; Zoonosis Science Center, University of Uppsala, Uppsala, Sweden
| | - Asifa Khan
- Department of Medical Biochemistry and Microbiology & The Biomedical Center; Zoonosis Science Center, University of Uppsala, Uppsala, Sweden
| | - Åke Lundkvist
- Department of Medical Biochemistry and Microbiology & The Biomedical Center; Zoonosis Science Center, University of Uppsala, Uppsala, Sweden.,Zoonosis Science Center, University of Uppsala, Uppsala, Sweden
| | - Jin-Ping Li
- Department of Medical Biochemistry and Microbiology & The Biomedical Center; Zoonosis Science Center, University of Uppsala, Uppsala, Sweden.,SciLifeLab Uppsala, University of Uppsala, Uppsala, Sweden
| |
Collapse
|
9
|
Anwar MN, Akhtar R, Abid M, Khan SA, Rehman ZU, Tayyub M, Malik MI, Shahzad MK, Mubeen H, Qadir MS, Hameed M, Wahaab A, Li Z, Liu K, Li B, Qiu Y, Ma Z, Wei J. The interactions of flaviviruses with cellular receptors: Implications for virus entry. Virology 2022; 568:77-85. [DOI: 10.1016/j.virol.2022.02.001] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2021] [Revised: 01/10/2022] [Accepted: 02/02/2022] [Indexed: 12/17/2022]
|
10
|
Mertinková P, Mochnáčová E, Bhide K, Kulkarni A, Tkáčová Z, Hruškovicová J, Bhide M. Development of peptides targeting receptor binding site of the envelope glycoprotein to contain the West Nile virus infection. Sci Rep 2021; 11:20131. [PMID: 34635758 PMCID: PMC8505397 DOI: 10.1038/s41598-021-99696-w] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2021] [Accepted: 09/24/2021] [Indexed: 11/09/2022] Open
Abstract
West Nile virus (WNV), re-emerging neurotropic flavivirus, can cross the blood-brain barrier (BBB) and cause fatal encephalitis and meningitis. Infection of the human brain microvascular endothelial cells (hBMECs), building blocks of the BBB, represents the pivotal step in neuroinvasion. Domain III (DIII) of the envelope (E) glycoprotein is a key receptor-binding domain, thus, it is an attractive target for anti-flavivirus strategies. Here, two combinatorial phage display peptide libraries, Ph.D.-C7C and Ph.D.-12, were panned against receptor-binding site (RBS) on DIII to isolate peptides that could block DIII. From series of pannings, nine peptides (seven 7-mer cyclic and two 12-mer linear) were selected and overexpressed in E. coli SHuffle T5. Presence of disulfide bond in 7-mer peptides was confirmed with thiol-reactive maleimide labeling. Except for linear peptide 19 (HYSWSWIAYSPG), all peptides proved to be DIII binders. Among all peptides, 4 cyclic peptides (CTKTDVHFC, CIHSSTRAC, CTYENHRTC, and CLAQSHPLC) showed significant blocking of the interaction between DIII and hBMECs, and ability to neutralize infection in cultured cells. None of these peptides showed toxic or hemolytic activity. Peptides identified in this study may serve as potential candidates for the development of novel antiviral therapeutics against WNV.
Collapse
Affiliation(s)
- Patrícia Mertinková
- grid.412971.80000 0001 2234 6772Laboratory of Biomedical Microbiology and Immunology, The University of Veterinary Medicine and Pharmacy in Košice, Komenského 73, 04181 Košice, Slovakia
| | - Evelína Mochnáčová
- grid.412971.80000 0001 2234 6772Laboratory of Biomedical Microbiology and Immunology, The University of Veterinary Medicine and Pharmacy in Košice, Komenského 73, 04181 Košice, Slovakia
| | - Katarína Bhide
- grid.412971.80000 0001 2234 6772Laboratory of Biomedical Microbiology and Immunology, The University of Veterinary Medicine and Pharmacy in Košice, Komenského 73, 04181 Košice, Slovakia
| | - Amod Kulkarni
- grid.412971.80000 0001 2234 6772Laboratory of Biomedical Microbiology and Immunology, The University of Veterinary Medicine and Pharmacy in Košice, Komenského 73, 04181 Košice, Slovakia ,grid.419303.c0000 0001 2180 9405Institute of Neuroimmunology of Slovak Academy of Sciences, Dubravska cesta 9, 84510 Bratislava, Slovakia
| | - Zuzana Tkáčová
- grid.412971.80000 0001 2234 6772Laboratory of Biomedical Microbiology and Immunology, The University of Veterinary Medicine and Pharmacy in Košice, Komenského 73, 04181 Košice, Slovakia
| | - Jana Hruškovicová
- grid.412971.80000 0001 2234 6772Laboratory of Biomedical Microbiology and Immunology, The University of Veterinary Medicine and Pharmacy in Košice, Komenského 73, 04181 Košice, Slovakia
| | - Mangesh Bhide
- grid.412971.80000 0001 2234 6772Laboratory of Biomedical Microbiology and Immunology, The University of Veterinary Medicine and Pharmacy in Košice, Komenského 73, 04181 Košice, Slovakia ,grid.419303.c0000 0001 2180 9405Institute of Neuroimmunology of Slovak Academy of Sciences, Dubravska cesta 9, 84510 Bratislava, Slovakia
| |
Collapse
|
11
|
Insights on Dengue and Zika NS5 RNA-dependent RNA polymerase (RdRp) inhibitors. Eur J Med Chem 2021; 224:113698. [PMID: 34274831 DOI: 10.1016/j.ejmech.2021.113698] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2021] [Revised: 07/09/2021] [Accepted: 07/10/2021] [Indexed: 11/20/2022]
Abstract
Over recent years, many outbreaks caused by (re)emerging RNA viruses have been reported worldwide, including life-threatening Flaviviruses, such as Dengue (DENV) and Zika (ZIKV). Currently, there is only one licensed vaccine against Dengue, Dengvaxia®. However, its administration is not recommended for children under nine years. Still, there are no specific inhibitors available to treat these infectious diseases. Among the flaviviral proteins, NS5 RNA-dependent RNA polymerase (RdRp) is a metalloenzyme essential for viral replication, suggesting that it is a promising macromolecular target since it has no human homolog. Nowadays, several NS5 RdRp inhibitors have been reported, while none inhibitors are currently in clinical development. In this context, this review constitutes a comprehensive work focused on RdRp inhibitors from natural, synthetic, and even repurposing sources. Furthermore, their main aspects associated with the structure-activity relationship (SAR), proposed mechanisms of action, computational studies, and other topics will be discussed in detail.
Collapse
|
12
|
Tangudu CS, Charles J, Nunez-Avellaneda D, Hargett AM, Brault AC, Blitvich BJ. Chimeric Zika viruses containing structural protein genes of insect-specific flaviviruses cannot replicate in vertebrate cells due to entry and post-translational restrictions. Virology 2021; 559:30-39. [PMID: 33812340 DOI: 10.1016/j.virol.2021.03.014] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2021] [Revised: 03/04/2021] [Accepted: 03/21/2021] [Indexed: 02/06/2023]
Abstract
Long Pine Key virus (LPKV) and Lammi virus are insect-specific flaviviruses that phylogenetically affiliate with dual-host flaviviruses. The goal of this study was to provide insight into the genetic determinants that condition this host range restriction. Chimeras were initially created by replacing select regions of the Zika virus genome, including the premembrane and envelope protein (prM-E) genes, with the corresponding regions of the LPKV genome. Of the four chimeras produced, one (the prM-E swap) yielded virus that replicated in mosquito cells. Another chimeric virus with a mosquito replication-competent phenotype was created by inserting the prM-E genes of Lammi virus into a Zika virus genetic background. Vertebrate cells did not support the replication of either chimeric virus although trace to modest amounts of viral antigen were produced, consistent with suboptimal viral entry. These data suggest that dual-host affiliated insect-specific flaviviruses cannot replicate in vertebrate cells due to entry and post-translational restrictions.
Collapse
Affiliation(s)
- Chandra S Tangudu
- Department of Veterinary Microbiology and Preventive Medicine, College of Veterinary Medicine, Iowa State University, Ames, IA, USA
| | - Jermilia Charles
- Department of Veterinary Microbiology and Preventive Medicine, College of Veterinary Medicine, Iowa State University, Ames, IA, USA
| | - Daniel Nunez-Avellaneda
- Department of Veterinary Microbiology and Preventive Medicine, College of Veterinary Medicine, Iowa State University, Ames, IA, USA
| | - Alissa M Hargett
- Department of Veterinary Microbiology and Preventive Medicine, College of Veterinary Medicine, Iowa State University, Ames, IA, USA
| | - Aaron C Brault
- Division of Vector-Borne Diseases, Centers for Disease Control and Prevention, Fort Collins, CO, USA
| | - Bradley J Blitvich
- Department of Veterinary Microbiology and Preventive Medicine, College of Veterinary Medicine, Iowa State University, Ames, IA, USA.
| |
Collapse
|
13
|
Westlake D, Bielefeldt-Ohmann H, Prow NA, Hall RA. Novel Flavivirus Attenuation Markers Identified in the Envelope Protein of Alfuy Virus. Viruses 2021; 13:v13020147. [PMID: 33498300 PMCID: PMC7909262 DOI: 10.3390/v13020147] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2020] [Revised: 01/12/2021] [Accepted: 01/14/2021] [Indexed: 11/16/2022] Open
Abstract
Alfuy (ALFV) is an attenuated flavivirus related to the Murray Valley encephalitis virus (MVEV). We previously identified markers of attenuation in the envelope (E) protein of the prototype strain (ALFV3929), including the hinge region (E273-277) and lack of glycosylation at E154-156. To further determine the mechanisms of attenuation we assessed ALFV3929 binding to glycosaminoglycans (GAG), a known mechanism of flaviviruses attenuation. Indeed, ALFV3929 exhibited reduced binding to GAG-rich cells in the presence of heparin; however, low-passage ALFV isolates were relatively unaffected. Sequence comparisons between ALFV strains and structural modelling incriminated a positively-charged residue (K327) in ALFV3929 as a GAG-binding motif. Substitution of this residue to the corresponding uncharged residue in MVEV (L), using a previously described chimeric virus containing the prM & E genes of ALFV3929 in the backbone of MVEV (MVEV/ALFV-prME), confirmed a role for K327 in enhanced GAG binding. When the wild type residues at E327, E273-277 and E154-156 of ALFV3929 were replaced with the corresponding residues from virulent MVEV, it revealed each motif contributed to attenuation of ALFV3929, with the E327/E273-277 combination most dominant. These data demonstrate that attenuation of ALFV3929 is multifactorial and provide new insights for the rational design of attenuated flavivirus vaccines.
Collapse
Affiliation(s)
- Daniel Westlake
- School of Chemistry and Molecular Biosciences, The University of Queensland, St. Lucia, QLD 4072, Australia; (D.W.); (H.B.-O.)
| | - Helle Bielefeldt-Ohmann
- School of Chemistry and Molecular Biosciences, The University of Queensland, St. Lucia, QLD 4072, Australia; (D.W.); (H.B.-O.)
- Australian Infectious Diseases Research Centre, The University of Queensland, St. Lucia, QLD 4072, Australia
- School of Veterinary Sciences, The University of Queensland, St. Lucia, QLD 4072, Australia
| | - Natalie A. Prow
- School of Chemistry and Molecular Biosciences, The University of Queensland, St. Lucia, QLD 4072, Australia; (D.W.); (H.B.-O.)
- Australian Infectious Diseases Research Centre, The University of Queensland, St. Lucia, QLD 4072, Australia
- Experimental Therapeutics Laboratory, School of Clinical and Health Sciences, University of South Australia Cancer Research Institute, Adelaide, SA 5000, Australia
- Correspondence: (N.A.P.); (R.A.H.)
| | - Roy A. Hall
- School of Chemistry and Molecular Biosciences, The University of Queensland, St. Lucia, QLD 4072, Australia; (D.W.); (H.B.-O.)
- Australian Infectious Diseases Research Centre, The University of Queensland, St. Lucia, QLD 4072, Australia
- Correspondence: (N.A.P.); (R.A.H.)
| |
Collapse
|
14
|
Pawar R, Patravale V. A Step towards Treating Dengue Viral Infection: An In Silico Approach to Identify Potential Antidengue Phytoconstituents. ChemistrySelect 2020. [DOI: 10.1002/slct.202004137] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022]
Affiliation(s)
- Rohit Pawar
- Department of Pharmaceutical Sciences and Technology Institute of Chemical Technology Nathalal Parekh Marg, Matunga Mumbai 400019 Maharashtra India
| | - Vandana Patravale
- Department of Pharmaceutical Sciences and Technology Institute of Chemical Technology Nathalal Parekh Marg, Matunga Mumbai 400019 Maharashtra India
| |
Collapse
|
15
|
Abstract
At the beginning of 2020, the national health system and medical communities are faced with unprecedented public health challenges. A novel strain of coronavirus, later identified as severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2), has spread globally, marking another pandemic of coronaviruses. This viral disease is responsible for devastating pneumonia, named coronavirus disease of 2019 (COVID-19), and projected to persist until the end of the year. In tropical countries, however, concerns arise regarding the similarities of COVID-19 with other infectious diseases due to the same chief complaint, which is fever. One of the infectious disease of a primary concern is dengue infection, which its peak season is approaching. Others report that there are cases of serological cross-reaction of COVID-19 and dengue infection. In this comprehensive review, we underscore the importance of knowing similar clinical presentations of both diseases and emphasize why excluding COVID-19 in the differentials in the setting of a pandemic is imprudent.
Collapse
|
16
|
Pong LY, Yew PN, Lee WL, Lim YY, Sharifah SH. Anti-dengue virus serotype 2 activity of tannins from porcupine dates. Chin Med 2020; 15:49. [PMID: 32467721 PMCID: PMC7238553 DOI: 10.1186/s13020-020-00329-7] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/02/2020] [Accepted: 05/12/2020] [Indexed: 11/10/2022] Open
Abstract
Background Dengue fever is currently endemic in tropical and subtropical countries worldwide and effective drug against DENV infection is still unavailable. Porcupine dates, which are traditionally used to treat dengue fever, might contain potential anti-dengue compounds. Two porcupine dates, black date (BD) and powdery date (PD) from Himalayan porcupine (Hystrix brachyura), were investigated for their antiviral activities against DENV-2 in vitro. Methods The methanol crude extracts (MBD and MPD) were prepared from the raw material of porcupine dates. The tannin-rich fractions (BDTF and PDTF) were isolated from their methanol crude extracts using column chromatography. The presence of tannins in BDTF and PDTF extracts was determined by fourier-transform infrared spectroscopy (FTIR) and nuclear magnetic resonance (NMR) analyses. The cytotoxicity and anti-DENV-2 activities including virus yield inhibition, virucidal, virus attachment and pre-treatment assays of the extracts were examined in Vero cells. Results Our findings revealed that all the extracts of porcupine dates exhibited antiviral activity against DENV-2 in Vero cells. The IC50 of BDTF and PDTF were 25 µg/mL and 11 µg/mL respectively, while their methanol crude extracts demonstrated lower antiviral efficacy (IC50 ≈ 101–107 µg/mL). BDTF and PDTF also exerted a similar higher virucidal effect (IC50 of 11 µg/mL) than methanol crude extracts (IC50 ≈ 52–66 µg/mL). Furthermore, all the extracts inhibited the attachment of DENV-2 by at least 80%. Pre-treatments of cells with BDTF and PDTF markedly prevented DENV-2 infection when compared to methanol crude extracts. Conclusion This study suggests that porcupine dates possess antiviral properties against DENV-2, which is attributed to its tannin compounds.
Collapse
Affiliation(s)
- Lian Yih Pong
- 1Jeffrey Cheah School of Medicine and Health Sciences, Monash University Malaysia, Jalan Lagoon Selatan, 47500 Bandar Sunway, Selangor Darul Ehsan Malaysia.,2Infectious Diseases and Health Cluster, Tropical Medicine and Biology Platform, Monash University Malaysia, Jalan Lagoon Selatan, 47500 Bandar Sunway, Selangor Darul Ehsan Malaysia
| | - Peng Nian Yew
- 3School of Science, Monash University Malaysia, Jalan Lagoon Selatan, 47500 Bandar Sunway, Selangor Darul Ehsan Malaysia.,4Present Address: Department of Bioscience, Faculty of Applied Science, Tunku Abdul Rahman University College, Jalan Genting Kelang, 53300 Kuala Lumpur, Malaysia
| | - Wai Leng Lee
- 3School of Science, Monash University Malaysia, Jalan Lagoon Selatan, 47500 Bandar Sunway, Selangor Darul Ehsan Malaysia
| | - Yau Yan Lim
- 3School of Science, Monash University Malaysia, Jalan Lagoon Selatan, 47500 Bandar Sunway, Selangor Darul Ehsan Malaysia
| | - Syed Hassan Sharifah
- 1Jeffrey Cheah School of Medicine and Health Sciences, Monash University Malaysia, Jalan Lagoon Selatan, 47500 Bandar Sunway, Selangor Darul Ehsan Malaysia.,2Infectious Diseases and Health Cluster, Tropical Medicine and Biology Platform, Monash University Malaysia, Jalan Lagoon Selatan, 47500 Bandar Sunway, Selangor Darul Ehsan Malaysia
| |
Collapse
|
17
|
Rana VS, Popli S, Saurav GK, Yadav K, Kumar A, Sunil S, Kumar N, Singh OP, Natarajan K, Rajagopal R. Aedes aegypti lachesin protein binds to the domain III of envelop protein of Dengue virus-2 and inhibits viral replication. Cell Microbiol 2020; 22:e13200. [PMID: 32141690 DOI: 10.1111/cmi.13200] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2019] [Revised: 01/31/2020] [Accepted: 02/16/2020] [Indexed: 12/21/2022]
Abstract
Dengue virus (DENV) comprises of four serotypes (DENV-1 to -4) and is medically one of the most important arboviruses (arthropod-borne virus). DENV infection is a major human health burden and is transmitted between humans by the insect vector, Aedes aegypti. Ae. aegypti ingests DENV while feeding on infected humans, which traverses through its gut, haemolymph and salivary glands of the mosquito before being injected into a healthy human. During this process of transmission, DENV must interact with many proteins of the insect vector, which are important for its successful transmission. Our study focused on the identification and characterisation of interacting protein partners in Ae. aegypti to DENV. Since domain III (DIII) of envelope protein (E) is exposed on the virion surface and is involved in virus entry into various cells, we performed phage display library screening against domain III of the envelope protein (EDIII) of DENV-2. A peptide sequence showing similarity to lachesin protein was found interacting with EDIII. The lachesin protein was cloned, heterologously expressed, purified and used for in vitro interaction studies. Lachesin protein interacted with EDIII and also with DENV. Further, lachesin protein was localised in neuronal cells of different organs of Ae. aegypti by confocal microscopy. Blocking of lachesin protein in Ae. aegypti with anti-lachesin antibody resulted in a significant reduction in DENV replication.
Collapse
Affiliation(s)
- Vipin S Rana
- Gut Biology Laboratory, Department of Zoology, University of Delhi, Delhi, India.,Department of Veterinary Medicine, University of Maryland, College Park, Maryland, USA
| | - Sonam Popli
- Gut Biology Laboratory, Department of Zoology, University of Delhi, Delhi, India.,Department of Medical Microbiology and Immunology, College of Medicine, University of Toledo, Toledo, Ohio, USA
| | - Gunjan K Saurav
- Gut Biology Laboratory, Department of Zoology, University of Delhi, Delhi, India.,Department of Zoology, Munshi Lal Arya College, Purnea University, Purnia, Bihar, India
| | - Karuna Yadav
- Gut Biology Laboratory, Department of Zoology, University of Delhi, Delhi, India
| | - Ankit Kumar
- Vector Borne Diseases Group, International Centre for Genetic Engineering and Biotechnology, Delhi, India
| | - Sujatha Sunil
- Vector Borne Diseases Group, International Centre for Genetic Engineering and Biotechnology, Delhi, India
| | - Narendra Kumar
- Department of Zoology, Shaheed Mangal Pandey Government Girls Post Graduate College, Meerut, Uttar Pradesh, India
| | - Om P Singh
- National Institute of Malaria Research, Delhi, India
| | | | - Raman Rajagopal
- Gut Biology Laboratory, Department of Zoology, University of Delhi, Delhi, India
| |
Collapse
|
18
|
Chia PY, Thein TL, Ong SWX, Lye DC, Leo YS. Severe dengue and liver involvement: an overview and review of the literature. Expert Rev Anti Infect Ther 2020; 18:181-189. [PMID: 31971031 DOI: 10.1080/14787210.2020.1720652] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Introduction: Two billion population are at risk of dengue fever and by 2080, over six billion population will be at risk. Hepatitis is common in dengue and the liver is invariably involved in severe cases. We conducted a literature review using the PubMed database on articles covering a broad range of issues related to dengue and hepatitis.Areas covered: This article overviews available literature on changes in the definition of severe dengue, pathogenesis of liver involvement in dengue, clinical manifestations, and predictors of mortality in severe dengue with liver involvement, impact of viral hepatitis co-infections and hepatotoxic drugs, and hemophagocytic lymphohistiocytosis.Expert commentary: Hepatitis is commonly seen in dengue however the degree of elevation of transaminases did not correlate well with severity of illness in observational studies, except in the elderly. The underlying pathogenesis of liver injury is still being elucidated and further studies are required to fully understand the cellular pathways. Acute or chronic viral hepatitis does not appear to affect dengue outcomes. Commonly used medications such as paracetamol and statins may influence dengue outcomes.
Collapse
Affiliation(s)
- Po Ying Chia
- National Centre for Infectious Diseases, Singapore, Singapore.,Department of Infectious Diseases, Tan Tock Seng Hospital, Singapore, Singapore.,Lee Kong Chian School of Medicine, Singapore, Singapore
| | - Tun-Linn Thein
- National Centre for Infectious Diseases, Singapore, Singapore
| | - Sean Wei Xiang Ong
- National Centre for Infectious Diseases, Singapore, Singapore.,Department of Infectious Diseases, Tan Tock Seng Hospital, Singapore, Singapore
| | - David Chien Lye
- National Centre for Infectious Diseases, Singapore, Singapore.,Department of Infectious Diseases, Tan Tock Seng Hospital, Singapore, Singapore.,Lee Kong Chian School of Medicine, Singapore, Singapore.,Department of Medicine, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
| | - Yee Sin Leo
- National Centre for Infectious Diseases, Singapore, Singapore.,Department of Infectious Diseases, Tan Tock Seng Hospital, Singapore, Singapore.,Lee Kong Chian School of Medicine, Singapore, Singapore.,Department of Medicine, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore.,Saw Swee Hock School of Public Health, National University of Singapore, Singapore, Singapore
| |
Collapse
|
19
|
Wong RR, Abd-Aziz N, Affendi S, Poh CL. Role of microRNAs in antiviral responses to dengue infection. J Biomed Sci 2020; 27:4. [PMID: 31898495 PMCID: PMC6941309 DOI: 10.1186/s12929-019-0614-x] [Citation(s) in RCA: 58] [Impact Index Per Article: 14.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2019] [Accepted: 12/29/2019] [Indexed: 12/13/2022] Open
Abstract
Dengue virus (DENV) is the etiological agent of dengue fever. Severe dengue could be fatal and there is currently no effective antiviral agent or vaccine. The only licensed vaccine, Dengvaxia, has low efficacy against serotypes 1 and 2. Cellular miRNAs are post-transcriptional regulators that could play a role in direct regulation of viral genes. Host miRNA expressions could either promote or repress viral replications. Induction of some cellular miRNAs could help the virus to evade the host immune response by suppressing the IFN-α/β signaling pathway while others could upregulate IFN-α/β production and inhibit the viral infection. Understanding miRNA expressions and functions during dengue infections would provide insights into the development of miRNA-based therapeutics which could be strategized to act either as miRNA antagonists or miRNA mimics. The known mechanisms of how miRNAs impact DENV replication are diverse. They could suppress DENV multiplication by directly binding to the viral genome, resulting in translational repression. Other miRNA actions include modulation of host factors. In addition, miRNAs that could modulate immunopathogenesis are discussed. Major hurdles lie in the development of chemical modifications and delivery systems for in vivo delivery. Nevertheless, advancement in miRNA formulations and delivery systems hold great promise for the therapeutic potential of miRNA-based therapy, as supported by Miravirsen for treatment of Hepatitis C infection which has successfully completed phase II clinical trial.
Collapse
Affiliation(s)
- Rui Rui Wong
- Centre for Virus and Vaccine Research (CVVR), Sunway University, 47500, Subang Jaya, Selangor, Malaysia
| | - Noraini Abd-Aziz
- Centre for Virus and Vaccine Research (CVVR), Sunway University, 47500, Subang Jaya, Selangor, Malaysia
| | - Sarah Affendi
- Centre for Virus and Vaccine Research (CVVR), Sunway University, 47500, Subang Jaya, Selangor, Malaysia
| | - Chit Laa Poh
- Centre for Virus and Vaccine Research (CVVR), Sunway University, 47500, Subang Jaya, Selangor, Malaysia.
| |
Collapse
|
20
|
Montes-Grajales D, Puerta-Guardo H, Espinosa DA, Harris E, Caicedo-Torres W, Olivero-Verbel J, Martínez-Romero E. In silico drug repurposing for the identification of potential candidate molecules against arboviruses infection. Antiviral Res 2019; 173:104668. [PMID: 31786251 DOI: 10.1016/j.antiviral.2019.104668] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2019] [Accepted: 11/27/2019] [Indexed: 01/09/2023]
Abstract
Arboviral diseases caused by dengue (DENV), Zika (ZIKV) and chikungunya (CHIKV) viruses represent a major public health problem worldwide, especially in tropical areas where millions of infections occur every year. The aim of this research was to identify candidate molecules for the treatment of these diseases among the drugs currently available in the market, through in silico screening and subsequent in vitro evaluation with cell culture models of DENV and ZIKV infections. Numerous pharmaceutical compounds from antibiotics to chemotherapeutic agents presented high in silico binding affinity for the viral proteins, including ergotamine, antrafenine, natamycin, pranlukast, nilotinib, itraconazole, conivaptan and novobiocin. These five last compounds were tested in vitro, being pranlukast the one that exhibited the best antiviral activity. Further in vitro assays for this compound showed a significant inhibitory effect on DENV and ZIKV infection of human monocytic cells and human hepatocytes (Huh-7 cells) with potential abrogation of virus entry. Finally, intrinsic fluorescence analyses suggest that pranlukast may have some level of interaction with three viral proteins of DENV: envelope, capsid, and NS1. Due to its promising results, suitable accessibility in the market and reduced restrictions compared to other pharmaceuticals; the anti-asthmatic pranlukast is proposed as a drug candidate against DENV, ZIKV, and CHIKV, supporting further in vitro and in vivo assessment of the potential of this and other lead compounds that exhibited good affinity scores in silico as therapeutic agents or scaffolds for the development of new drugs against arboviral diseases.
Collapse
Affiliation(s)
- Diana Montes-Grajales
- Environmental and Computational Chemistry Group, School of Pharmaceutical Sciences, Zaragocilla Campus, University of Cartagena, Cartagena, 130015, Colombia.
| | - Henry Puerta-Guardo
- Division of Infectious Diseases and Vaccinology, School of Public Health, University of California, Berkeley, Berkeley, CA, 94720-3370, USA
| | - Diego A Espinosa
- Division of Infectious Diseases and Vaccinology, School of Public Health, University of California, Berkeley, Berkeley, CA, 94720-3370, USA
| | - Eva Harris
- Division of Infectious Diseases and Vaccinology, School of Public Health, University of California, Berkeley, Berkeley, CA, 94720-3370, USA
| | - William Caicedo-Torres
- Grupo de Investigación de Tecnologías Aplicadas y Sistemas de Información, School of Engineering, Universidad Tecnológica de Bolívar, Cartagena, 130010, Colombia
| | - Jesus Olivero-Verbel
- Environmental and Computational Chemistry Group, School of Pharmaceutical Sciences, Zaragocilla Campus, University of Cartagena, Cartagena, 130015, Colombia
| | - Esperanza Martínez-Romero
- Centro de Ciencias Genómicas, Universidad Nacional Autónoma de México, Cuernavaca-Morelos 565-A, Mexico
| |
Collapse
|
21
|
Chang YJ, Pong LY, Hassan SS, Choo WS. Antiviral activity of betacyanins from red pitahaya ( Hylocereus polyrhizus) and red spinach ( Amaranthus dubius) against dengue virus type 2 (GenBank accession no. MH488959). Access Microbiol 2019; 2:acmi000073. [PMID: 33062932 PMCID: PMC7525058 DOI: 10.1099/acmi.0.000073] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2019] [Accepted: 10/03/2019] [Indexed: 02/04/2023] Open
Abstract
This study investigated the antiviral activity of betacyanins from red pitahaya (Hylocereus polyrhizus) and red spinach (Amaranthus dubius) against dengue virus type 2 (DENV-2). The pulp of red pitahaya and the leaves of red spinach were extracted using methanol followed by sub-fractionation and Amberlite XAD16N column chromatography to obtain betacyanin fractions. The half maximum cytotoxicity concentration for betacyanin fractions from red pitahaya and red spinach on Vero cells were 4.346 and 2.287 mg ml-1, respectively. The half-maximal inhibitory concentration (IC50) of betacyanin fraction from red pitahaya was 125.8 μg ml-1 with selectivity index (SI) of 5.8. For betacyanin fraction from red spinach, the IC50 value was 14.62 µg ml-1 with SI of 28.51. Using the maximum non-toxic betacyanin concentration, direct virucidal effect against DENV-2 was obtained from betacyanin fraction from red pitahaya (IC50 of 126.70 μg ml-1; 95.0 % virus inhibition) and red spinach (IC50 value of 106.80 μg ml-1; 65.9 % of virus inhibition). Betacyanin fractions from red pitahaya and red spinach inhibited DENV-2 in vitro.
Collapse
Affiliation(s)
- Ying Jun Chang
- School of Science, Monash University Malaysia, Jalan Lagoon Selatan, 47500 Bandar Sunway, Selangor, Malaysia
| | - Lian Yih Pong
- Jeffrey Cheah School of Medicine and Health Sciences, Monash University Malaysia, Jalan Lagoon Selatan, 47500 Bandar Sunway, Selangor, Malaysia
- Infectious Diseases and Health Cluster, Tropical Medicine and Biology Platform, Monash University Malaysia, Jalan Lagoon Selatan, 47500 Bandar Sunway, Selangor, Malaysia
| | - Sharifah S. Hassan
- Jeffrey Cheah School of Medicine and Health Sciences, Monash University Malaysia, Jalan Lagoon Selatan, 47500 Bandar Sunway, Selangor, Malaysia
- Infectious Diseases and Health Cluster, Tropical Medicine and Biology Platform, Monash University Malaysia, Jalan Lagoon Selatan, 47500 Bandar Sunway, Selangor, Malaysia
| | - Wee Sim Choo
- School of Science, Monash University Malaysia, Jalan Lagoon Selatan, 47500 Bandar Sunway, Selangor, Malaysia
| |
Collapse
|
22
|
Faheem M, Barbosa Lima JC, Jamal SB, Silva PA, Barbosa JARG. An insight into dengue virus proteins as potential drug/vaccine targets. Future Virol 2019. [DOI: 10.2217/fvl-2019-0107] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
Abstract
Dengue virus (DENV) is an arbovirus that belongs to family flaviviridae. Its genome is composed of a single stranded RNA molecule that encodes a single polyprotein. The polyprotein is processed by viral and cellular proteases to generate ten viral proteins. There are four antigenically distinct serotypes of DENV (DENV1, DENV2, DENV3 and DENV4), which are genetically related. Although protein variability is a major problem in dengue treatment, the functional and structural studies of individual proteins are equally important in treatment development. The data accumulated on dengue proteins are significant to provide detailed understanding of viral infection, replication, host-immune evasion and pathogenesis. In this review, we summarized the detailed current knowledge about DENV proteins.
Collapse
Affiliation(s)
- Muhammad Faheem
- Laboratory of Biophysics, Department of Cellular Biology, University of Brasilia, Brasilia-DF 70910-900, Brazil
- Post-graduate program of Genomics Sciences & Biotechnology, Catholic University of Brasilia, Brasília-DF 70790-160, Brazil
| | - Jônatas Cunha Barbosa Lima
- Laboratory of Biophysics, Department of Cellular Biology, University of Brasilia, Brasilia-DF 70910-900, Brazil
| | - Syed Babar Jamal
- Department of Biological Sciences, National University of Medical Sciences, The Mall road, Rawalpindi, Punjab 46000, Pakistan
| | - Paula Andreia Silva
- Post-graduate program of Genomics Sciences & Biotechnology, Catholic University of Brasilia, Brasília-DF 70790-160, Brazil
| | - João Alexandre Ribeiro Gonçalves Barbosa
- Laboratory of Biophysics, Department of Cellular Biology, University of Brasilia, Brasilia-DF 70910-900, Brazil
- Post-graduate program of Genomics Sciences & Biotechnology, Catholic University of Brasilia, Brasília-DF 70790-160, Brazil
| |
Collapse
|
23
|
Kerrigan SW, Devine T, Fitzpatrick G, Thachil J, Cox D. Early Host Interactions That Drive the Dysregulated Response in Sepsis. Front Immunol 2019; 10:1748. [PMID: 31447831 PMCID: PMC6691039 DOI: 10.3389/fimmu.2019.01748] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2019] [Accepted: 07/10/2019] [Indexed: 01/18/2023] Open
Abstract
Sepsis is defined as life-threatening organ dysfunction caused by a dysregulated host response to infection. While many individual cells and systems in the body are involved in driving the excessive and sometimes sustained host response, pathogen engagement with endothelial cells and platelets early in sepsis progression, are believed to be key. Significant progress has been made in establishing key molecular interactions between platelets and pathogens and endothelial cells and pathogens. This review will explore the growing number of compensatory connections between bacteria and viruses with platelets and endothelial cells and how a better understanding of these interactions are informing the field of potential novel ways to treat the dysregulated host response during sepsis.
Collapse
Affiliation(s)
- Steven W Kerrigan
- Cardiovascular Infection Research Group, Royal College of Surgeons in Ireland, Dublin, Ireland.,School of Pharmacy, Royal College of Surgeons in Ireland, Dublin, Ireland.,Molecular and Cellular Therapeutics, Royal College of Surgeons in Ireland, Dublin, Ireland.,Irish Centre for Vascular Biology, Royal College of Surgeons in Ireland, Dublin, Ireland
| | - Tatyana Devine
- Cardiovascular Infection Research Group, Royal College of Surgeons in Ireland, Dublin, Ireland.,Molecular and Cellular Therapeutics, Royal College of Surgeons in Ireland, Dublin, Ireland.,Irish Centre for Vascular Biology, Royal College of Surgeons in Ireland, Dublin, Ireland
| | - Glenn Fitzpatrick
- Cardiovascular Infection Research Group, Royal College of Surgeons in Ireland, Dublin, Ireland.,School of Pharmacy, Royal College of Surgeons in Ireland, Dublin, Ireland.,Irish Centre for Vascular Biology, Royal College of Surgeons in Ireland, Dublin, Ireland
| | - Jecko Thachil
- Department of Haematology, Central Manchester University Hospitals NHS Foundation Trust, Manchester, United Kingdom
| | - Dermot Cox
- Cardiovascular Infection Research Group, Royal College of Surgeons in Ireland, Dublin, Ireland.,Molecular and Cellular Therapeutics, Royal College of Surgeons in Ireland, Dublin, Ireland.,Irish Centre for Vascular Biology, Royal College of Surgeons in Ireland, Dublin, Ireland
| |
Collapse
|
24
|
Niu J, Jiang Y, Xu H, Zhao C, Zhou G, Chen P, Cao R. TIM-1 Promotes Japanese Encephalitis Virus Entry and Infection. Viruses 2018; 10:E630. [PMID: 30441759 PMCID: PMC6265761 DOI: 10.3390/v10110630] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2018] [Revised: 11/09/2018] [Accepted: 11/10/2018] [Indexed: 01/23/2023] Open
Abstract
Japanese encephalitis virus (JEV) is a mosquito-borne Flavivirus, the leading cause of viral-induced encephalitis. Several host molecules have been identified as the JEV attachment factor; however, the molecules involved in JEV entry remain poorly understood. In the present study, we demonstrate that TIM-1 is important for efficient infection by JEV. Firstly, three TIM-1 variants (V1, V2, and V3) were cloned from A549 cells, and we revealed that only ectopically TIM-1 V2 expression in 293T cells significantly promotes JEV attachment, entry and infection. Point mutation of phosphatidylserine (Ptdser) binding pocket in the TIM-1 IgV domain dampened JEV entry, indicating that TIM-1-mediated JEV infection is Ptdser-dependent. Furthermore, we found the cytoplasmic domain of TIM-1 is also required for enhancing JEV entry. Additionally, knock down of TIM-1 expression in A549 cells impaired JEV entry and infection, but not attachment, suggesting that additional factors exist in A549 cells that allow the virus to bind. In conclusion, our findings demonstrate that TIM-1 promotes JEV infection as an entry cofactor, and the polymorphism of TIM-1 is associated with JEV susceptibility to host cells.
Collapse
Affiliation(s)
- Jichen Niu
- MOE Joint International Research Laboratory of Animal Health and Food Safety, College of Veterinary Medicine, Nanjing Agricultural University, Nanjing 210095, China.
| | - Ya Jiang
- MOE Joint International Research Laboratory of Animal Health and Food Safety, College of Veterinary Medicine, Nanjing Agricultural University, Nanjing 210095, China.
| | - Hao Xu
- MOE Joint International Research Laboratory of Animal Health and Food Safety, College of Veterinary Medicine, Nanjing Agricultural University, Nanjing 210095, China.
| | - Changjing Zhao
- MOE Joint International Research Laboratory of Animal Health and Food Safety, College of Veterinary Medicine, Nanjing Agricultural University, Nanjing 210095, China.
| | - Guodong Zhou
- MOE Joint International Research Laboratory of Animal Health and Food Safety, College of Veterinary Medicine, Nanjing Agricultural University, Nanjing 210095, China.
| | - Puyan Chen
- MOE Joint International Research Laboratory of Animal Health and Food Safety, College of Veterinary Medicine, Nanjing Agricultural University, Nanjing 210095, China.
| | - Ruibing Cao
- MOE Joint International Research Laboratory of Animal Health and Food Safety, College of Veterinary Medicine, Nanjing Agricultural University, Nanjing 210095, China.
| |
Collapse
|
25
|
Azami NAM, Moi ML, Ami Y, Suzaki Y, Lim CK, Taniguchi S, Saijo M, Takasaki T, Kurane I. Genotype-specific and cross-reactive neutralizing antibodies induced by dengue virus infection: detection of antibodies with different levels of neutralizing activities against homologous and heterologous genotypes of dengue virus type 2 in common marmosets (Callithrix jacchus). Virol J 2018; 15:51. [PMID: 29587780 PMCID: PMC5870686 DOI: 10.1186/s12985-018-0967-x] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2017] [Accepted: 03/19/2018] [Indexed: 12/02/2022] Open
Abstract
BACKGROUND A vaccine against all four dengue virus (DENV) serotypes includes the formulation of one genotype of each serotype. Although genetic similarities among genotypes within a serotype are higher as compared to those among serotypes, differences in the immunogenicity of the included genotypes would be a critical issue in maximizing successful dengue vaccine development. Thus, we determined the neutralizing antibody responses against three genotypes of dengue virus serotype 2 (DENV-2), namely Cosmopolitan, Asian I, and Asian/American, after primary and secondary inoculation with DENV-2 in a dengue animal model, the common marmoset (Callithrix jacchus). METHODS A total of fifty-four plasma samples were obtained from thirty-four marmosets that were inoculated with clinically-isolated DENV strains or DENV candidate vaccines, were used in this study. Plasma samples were obtained from marmosets after primary inoculation with DENV-2 infection, secondary inoculation with homologous or heterologous genotypes, and tertiary inoculation with heterologous DENV. Neutralizing antibody titers against DENV-2 (Cosmopolitan, Asian I, and Asian/American genotypes) and DENV-1 were determined using a conventional plaque reduction neutralization assay. RESULTS In marmosets that were inoculated with the Cosmopolitan genotype in primary infection, neutralizing antibody neutralized 3 genotypes, and the titers to Asian I genotype were significantly higher than those to homologous Cosmopolitan genotype. After secondary DENV-2 infection with heterologous genotype (Asian I in primary and Asian/American in secondary), neutralizing antibody titers to Asian/American genotype was significantly higher than those against Cosmopolitan and Asian I genotypes. Following tertiary infection with DENV-1 following DENV-2 Asian I and Cosmopolitan genotypes, neutralizing antibody titers to Asian/American were also significantly higher than those against Cosmopolitan and Asian I genotypes. CONCLUSION The present study demonstrated that different levels of neutralizing antibodies were induced against variable DENV-2 genotypes after primary, secondary and tertiary infections, and that neutralizing antibody titers to some heterologous genotypes were higher than those to homologous genotypes within a serotype. The results indicate that heterogeneity and homogeneity of infecting genotypes influence the levels and cross-reactivity of neutralizing antibodies induced in following infections. The results also suggest that certain genotypes may possess advantage in terms of breakthrough infections against vaccination.
Collapse
Affiliation(s)
- Nor Azila Muhammad Azami
- Department of Virology 1, National Institute of Infectious Diseases, Tokyo, Japan
- Graduate School of Comprehensive Human Sciences, University of Tsukuba, Ibaraki, Japan
| | - Meng Ling Moi
- Department of Virology 1, National Institute of Infectious Diseases, Tokyo, Japan
- Institute of Tropical Medicine, Nagasaki University, Nagasaki, Japan
| | - Yasushi Ami
- Division of Experimental Animal Research, National Institute of Infectious Diseases, Tokyo, Japan
| | - Yuriko Suzaki
- Division of Experimental Animal Research, National Institute of Infectious Diseases, Tokyo, Japan
| | - Chang-Kweng Lim
- Department of Virology 1, National Institute of Infectious Diseases, Tokyo, Japan
| | - Satoshi Taniguchi
- Department of Virology 1, National Institute of Infectious Diseases, Tokyo, Japan
| | - Masayuki Saijo
- Department of Virology 1, National Institute of Infectious Diseases, Tokyo, Japan
| | | | - Ichiro Kurane
- National Institute of Infectious Diseases, 1-23-1 Toyama, Shinjuku, Tokyo, 162-8640 Japan
| |
Collapse
|
26
|
Abdul Ahmad SA, Palanisamy UD, Tejo BA, Chew MF, Tham HW, Syed Hassan S. Geraniin extracted from the rind of Nephelium lappaceum binds to dengue virus type-2 envelope protein and inhibits early stage of virus replication. Virol J 2017; 14:229. [PMID: 29162124 PMCID: PMC5698958 DOI: 10.1186/s12985-017-0895-1] [Citation(s) in RCA: 36] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2017] [Accepted: 11/13/2017] [Indexed: 01/17/2023] Open
Abstract
Background The rapid rise and spread in dengue cases, together with the unavailability of safe vaccines and effective antiviral drugs, warrant the need to discover and develop novel anti-dengue treatments. In this study the antiviral activity of geraniin, extracted from the rind of Nephelium lappaceum, against dengue virus type-2 (DENV-2) was investigated. Methods Geraniin was prepared from Nephelium lappaceum rind by reverse phase C-18 column chromatography. Cytotoxicity of geraniin towards Vero cells was evaluated using MTT assay while IC50 value was determined by plaque reduction assay. The mode-of-action of geraniin was characterized using the virucidal, attachment, penetration and the time-of-addition assays’. Docking experiments with geraniin molecule and the DENV envelope (E) protein was also performed. Finally, recombinant E Domain III (rE-DIII) protein was produced to physiologically test the binding of geraniin to DENV-2 E-DIII protein, through ELISA competitive binding assay. Results Cytotoxicity assay confirmed that geraniin was not toxic to Vero cells, even at the highest concentration tested. The compound exhibited DENV-2 plaque formation inhibition, with an IC50 of 1.75 μM. We further revealed that geraniin reduced viral infectivity and inhibited DENV-2 from attaching to the cells but had little effect on its penetration. Geraniin was observed to be most effective when added at the early stage of DENV-2 infection. Docking experiments showed that geraniin binds to DENV E protein, specifically at the DIII region, while the ELISA competitive binding assay confirmed geraniin’s interaction with rE-DIII with high affinity. Conclusions Geraniin from the rind of Nephelium lappaceum has antiviral activity against DENV-2. It is postulated that the compound inhibits viral attachment by binding to the E-DIII protein and interferes with the initial cell-virus interaction. Our results demonstrate that geraniin has the potential to be developed into an effective antiviral treatment, particularly for early phase dengue viral infection.
Collapse
Affiliation(s)
- Siti Aisyah Abdul Ahmad
- Virus-Host Interaction Research Group, Jeffrey Cheah School of Medicine and Health Sciences, Monash University Malaysia, Jalan Lagoon Selatan, 47500, Bandar Sunway, Selangor, Malaysia
| | - Uma D Palanisamy
- Virus-Host Interaction Research Group, Jeffrey Cheah School of Medicine and Health Sciences, Monash University Malaysia, Jalan Lagoon Selatan, 47500, Bandar Sunway, Selangor, Malaysia
| | - Bimo A Tejo
- Department of Chemistry, Universiti Putra Malaysia, 43400, Serdang, Malaysia.,Faculty of Applied Sciences, UCSI University, No. 1, Jalan Menara Gading, UCSI Heights, 56000, Kuala Lumpur, Cheras, Malaysia
| | - Miaw Fang Chew
- Virus-Host Interaction Research Group, Jeffrey Cheah School of Medicine and Health Sciences, Monash University Malaysia, Jalan Lagoon Selatan, 47500, Bandar Sunway, Selangor, Malaysia.,Research Centre for Biomedical Sciences, Sunway University, 47500, Bandar Sunway, Selangor, Malaysia
| | - Hong Wai Tham
- Virus-Host Interaction Research Group, Jeffrey Cheah School of Medicine and Health Sciences, Monash University Malaysia, Jalan Lagoon Selatan, 47500, Bandar Sunway, Selangor, Malaysia.,Faculty of Pharmacy, SEGI University, 9 Jalan Teknologi, Taman Sains Selangor, PJU 5, Kota Damansara, 47810, Petaling Jaya, Selangor, Malaysia
| | - Sharifah Syed Hassan
- Virus-Host Interaction Research Group, Jeffrey Cheah School of Medicine and Health Sciences, Monash University Malaysia, Jalan Lagoon Selatan, 47500, Bandar Sunway, Selangor, Malaysia. .,Infectious Diseases and Health Cluster, Tropical Medicine and Biology Platform, Monash University Malaysia, Bandar Sunway, 47500, Selangor, Malaysia.
| |
Collapse
|
27
|
Raj M, Langley M, McArthur SJ, Jean F. Moonlighting glycolytic enzyme glyceraldehyde-3-phosphate dehydrogenase (GAPDH) is required for efficient hepatitis C virus and dengue virus infections in human Huh-7.5.1 cells. J Gen Virol 2017; 98:977-991. [PMID: 28548037 DOI: 10.1099/jgv.0.000754] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022] Open
Abstract
Hijacking of cellular biosynthetic pathways by human enveloped viruses is a shared molecular event essential for the viral lifecycle. In this study, the accumulating evidence of the importance of human glyceraldehyde-3-phosphate dehydrogenase (GAPDH) in the host secretory pathway led us to hypothesize that this moonlighting enzyme could play a key role in the lifecycle steps of two important Flaviviridae members, hepatitis C virus (HCV) and dengue virus (DENV). We used short interfering RNA (siRNA)-mediated knockdown of human GAPDH in Huh-7.5.1 cells- both pre- and post-HCV infection- to demonstrate that GAPDH is a host factor for HCV infection. siRNA-induced GAPDH knockdown performed pre-HCV infection inhibits HCV core production in infected cells and leads to a decrease in infectivity of the HCV-infected cell supernatants. siRNA-induced GAPDH knockdown performed post-HCV infection does not have an effect on HCV core abundance in infected cells, but does lead to a decrease in infectivity of the HCV-infected cell supernatants. Exogenous expression of V5-tagged human GAPDH, pre- and post-infection, increases the infectivity of HCV-infected cell supernatants, suggesting a role for GAPDH during HCV post-replication steps. Interestingly, siRNA-induced GAPDH knockdown in HCV replicon-harbouring cells had no effect on viral RNA replication. Importantly, we confirmed the important role of GAPDH in the HCV lifecycle using Huh-7-derived stable GAPDH-knockdown clones. Finally, siRNA-induced GAPDH knockdown inhibits intracellular DENV-2 E glycoprotein production in infected cells. Collectively, our findings suggest that the moonlighting enzyme, GAPDH, is an important host factor for HCV infection, and they support its potential role in the DENV lifecycle.
Collapse
Affiliation(s)
- Meera Raj
- Department of Microbiology and Immunology, University of British Columbia, Vancouver, BC V6T 1Z4, Canada.,Present address: Canadian Blood Services and the Centre for Blood Research, University of British Columbia, Vancouver, BC V6T 1Z3, Canada
| | - Mary Langley
- Department of Microbiology and Immunology, University of British Columbia, Vancouver, BC V6T 1Z4, Canada.,Present address: School of Medicine, Flinders University, Adelaide, South Australia
| | - Steven J McArthur
- Department of Microbiology and Immunology, University of British Columbia, Vancouver, BC V6T 1Z4, Canada
| | - François Jean
- Department of Microbiology and Immunology, University of British Columbia, Vancouver, BC V6T 1Z4, Canada
| |
Collapse
|
28
|
Amir-Hassan A, Lee VS, Baharuddin A, Othman S, Xu Y, Huang M, Yusof R, Rahman NA, Othman R. Conformational and energy evaluations of novel peptides binding to dengue virus envelope protein. J Mol Graph Model 2017; 74:273-287. [PMID: 28458006 DOI: 10.1016/j.jmgm.2017.03.010] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2017] [Revised: 03/15/2017] [Accepted: 03/16/2017] [Indexed: 12/13/2022]
Abstract
Effective novel peptide inhibitors which targeted the domain III of the dengue envelope (E) protein by blocking dengue virus (DENV) entry into target cells, were identified. The binding affinities of these peptides towards E-protein were evaluated by using a combination of docking and explicit solvent molecular dynamics (MD) simulation methods. The interactions of these complexes were further investigated by using the Molecular Mechanics-Poisson Boltzmann Surface Area (MMPBSA) and Molecular Mechanics Generalized Born Surface Area (MMGBSA) methods. Free energy calculations of the peptides interacting with the E-protein demonstrated that van der Waals (vdW) and electrostatic interactions were the main driving forces stabilizing the complexes. Interestingly, calculated binding free energies showed good agreement with the experimental dissociation constant (Kd) values. Our results also demonstrated that specific residues might play a crucial role in the effective binding interactions. Thus, this study has demonstrated that a combination of docking and molecular dynamics simulations can accelerate the identification process of peptides as potential inhibitors of dengue virus entry into host cells.
Collapse
Affiliation(s)
- Asfarina Amir-Hassan
- Drug Design & Development Research Group, University of Malaya, 50603 Kuala Lumpur, Malaysia; Department of Pharmacy, Faculty of Medicine, University of Malaya, 50603 Kuala Lumpur, Malaysia
| | - Vannajan Sanghiran Lee
- Drug Design & Development Research Group, University of Malaya, 50603 Kuala Lumpur, Malaysia; Department of Chemistry, Faculty of Science, University of Malaya, 50603 Kuala Lumpur, Malaysia
| | - Aida Baharuddin
- Drug Design & Development Research Group, University of Malaya, 50603 Kuala Lumpur, Malaysia; Department of Molecular Medicine, Faculty of Medicine, University of Malaya, 50603 Kuala Lumpur, Malaysia
| | - Shatrah Othman
- Drug Design & Development Research Group, University of Malaya, 50603 Kuala Lumpur, Malaysia; Department of Molecular Medicine, Faculty of Medicine, University of Malaya, 50603 Kuala Lumpur, Malaysia
| | - Yongtao Xu
- School of Chemistry and Chemical Engineering, Queen's University Belfast, Northern Ireland, United Kingdom; School of Biomedical Engineering, Xinxiang Medical University, Xinxiang, Henan 453003, China
| | - Meilan Huang
- School of Chemistry and Chemical Engineering, Queen's University Belfast, Northern Ireland, United Kingdom
| | - Rohana Yusof
- Drug Design & Development Research Group, University of Malaya, 50603 Kuala Lumpur, Malaysia; Department of Molecular Medicine, Faculty of Medicine, University of Malaya, 50603 Kuala Lumpur, Malaysia
| | - Noorsaadah Abd Rahman
- Drug Design & Development Research Group, University of Malaya, 50603 Kuala Lumpur, Malaysia; Department of Chemistry, Faculty of Science, University of Malaya, 50603 Kuala Lumpur, Malaysia
| | - Rozana Othman
- Drug Design & Development Research Group, University of Malaya, 50603 Kuala Lumpur, Malaysia; Department of Pharmacy, Faculty of Medicine, University of Malaya, 50603 Kuala Lumpur, Malaysia; Centre for Natural Product Research and Drug Discovery, University of Malaya, 50603 Kuala Lumpur, Malaysia.
| |
Collapse
|
29
|
Recovery of West Nile Virus Envelope Protein Domain III Chimeras with Altered Antigenicity and Mouse Virulence. J Virol 2016; 90:4757-4770. [PMID: 26912625 DOI: 10.1128/jvi.02861-15] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2015] [Accepted: 02/20/2016] [Indexed: 11/20/2022] Open
Abstract
UNLABELLED Flaviviruses are positive-sense, single-stranded RNA viruses responsible for millions of human infections annually. The envelope (E) protein of flaviviruses comprises three structural domains, of which domain III (EIII) represents a discrete subunit. The EIII gene sequence typically encodes epitopes recognized by virus-specific, potently neutralizing antibodies, and EIII is believed to play a major role in receptor binding. In order to assess potential interactions between EIII and the remainder of the E protein and to assess the effects of EIII sequence substitutions on the antigenicity, growth, and virulence of a representative flavivirus, chimeric viruses were generated using the West Nile virus (WNV) infectious clone, into which EIIIs from nine flaviviruses with various levels of genetic diversity from WNV were substituted. Of the constructs tested, chimeras containing EIIIs from Koutango virus (KOUV), Japanese encephalitis virus (JEV), St. Louis encephalitis virus (SLEV), and Bagaza virus (BAGV) were successfully recovered. Characterization of the chimeras in vitro and in vivo revealed differences in growth and virulence between the viruses, within vivo pathogenesis often not being correlated within vitro growth. Taken together, the data demonstrate that substitutions of EIII can allow the generation of viable chimeric viruses with significantly altered antigenicity and virulence. IMPORTANCE The envelope (E) glycoprotein is the major protein present on the surface of flavivirus virions and is responsible for mediating virus binding and entry into target cells. Several viable West Nile virus (WNV) variants with chimeric E proteins in which the putative receptor-binding domain (EIII) sequences of other mosquito-borne flaviviruses were substituted in place of the WNV EIII were recovered, although the substitution of several more divergent EIII sequences was not tolerated. The differences in virulence and tissue tropism observed with the chimeric viruses indicate a significant role for this sequence in determining the pathogenesis of the virus within the mammalian host. Our studies demonstrate that these chimeras are viable and suggest that such recombinant viruses may be useful for investigation of domain-specific antibody responses and the more extensive definition of the contributions of EIII to the tropism and pathogenesis of WNV or other flaviviruses.
Collapse
|
30
|
Drumond BP, Fagundes LGDS, Rocha RP, Fumagalli MJ, Araki CS, Colombo TE, Nogueira ML, Castilho TE, da Silveira NJF, Malaquias LCC, Coelho LFL. Phylogenetic analysis of Dengue virus 1 isolated from South Minas Gerais, Brazil. Braz J Microbiol 2016; 47:251-8. [PMID: 26887252 PMCID: PMC4827697 DOI: 10.1016/j.bjm.2015.11.016] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2014] [Accepted: 07/24/2015] [Indexed: 11/24/2022] Open
Abstract
Dengue is a major worldwide public health problem, especially in the tropical and subtropical regions of the world. Primary infection with a single Dengue virus serotype causes a mild, self-limiting febrile illness called dengue fever. However, a subset of patients who experience secondary infection with a different serotype can progress to a more severe form of the disease, called dengue hemorrhagic fever. The four Dengue virus serotypes (1–4) are antigenically and genetically distinct and each serotype is composed of multiple genotypes. In this study we isolated one Dengue virus 1 serotype, named BR/Alfenas/2012, from a patient with dengue hemorrhagic fever in Alfenas, South Minas Gerais, Brazil and molecular identification was performed based on the analysis of NS5 gene. Swiss mice were infected with this isolate to verify its potential to induce histopathological alterations characteristic of dengue. Liver histopathological analysis of infected animals showed the presence of inflammatory infiltrates, hepatic steatosis, as well as edema, hemorrhage and necrosis focal points. Phylogenetic and evolutionary analyses based on the envelope gene provided evidence that the isolate BR/Alfenas/2012 belongs to genotype V, lineage I and it is probably derived from isolates of Rio de Janeiro, Brazil. The isolate BR/Alfenas/2012 showed two unique amino acids substitutions (SER222THRE and PHE306SER) when compared to other Brazilian isolates from the same genotype/lineage. Molecular models were generated for the envelope protein indicating that the amino acid alteration PHE 306 SER could contribute to a different folding in this region located within the domain III. Further genetic and animal model studies using BR/Alfenas/2012 and other isolates belonging to the same lineage/genotype could help determine the relation of these genetic alterations and dengue hemorrhagic fever in a susceptible population.
Collapse
Affiliation(s)
- Betania Paiva Drumond
- Laboratório de Virologia, Instituto de Ciências Biológicas, Universidade Federal de Juiz de Fora, Juiz de Fora, Minas Gerais, Brazil
| | - Luiz Gustavo da Silva Fagundes
- Laboratório de Vacinas, Instituto de Ciências Biomédicas, Universidade Federal de Alfenas, Alfenas, Minas Gerais, Brazil
| | - Raissa Prado Rocha
- Laboratório de Vacinas, Instituto de Ciências Biomédicas, Universidade Federal de Alfenas, Alfenas, Minas Gerais, Brazil
| | - Marcilio Jorge Fumagalli
- Laboratório de Vacinas, Instituto de Ciências Biomédicas, Universidade Federal de Alfenas, Alfenas, Minas Gerais, Brazil
| | - Carlos Shigueru Araki
- Laboratório de Pesquisas Em Virologia, Departamento de Doenças Infecciosas e Parasitárias, Faculdade de Medicina de São José do Rio Preto, São Paulo, Brazil
| | - Tatiana Elisa Colombo
- Laboratório de Pesquisas Em Virologia, Departamento de Doenças Infecciosas e Parasitárias, Faculdade de Medicina de São José do Rio Preto, São Paulo, Brazil
| | - Mauricio Lacerda Nogueira
- Laboratório de Pesquisas Em Virologia, Departamento de Doenças Infecciosas e Parasitárias, Faculdade de Medicina de São José do Rio Preto, São Paulo, Brazil
| | - Thiago Elias Castilho
- Laboratório de Bioinformática, Universidade Federal de Alfenas, Alfenas, Minas Gerais, Brazil
| | | | - Luiz Cosme Cotta Malaquias
- Laboratório de Virologia, Instituto de Ciências Biológicas, Universidade Federal de Juiz de Fora, Juiz de Fora, Minas Gerais, Brazil
| | - Luiz Felipe Leomil Coelho
- Laboratório de Virologia, Instituto de Ciências Biológicas, Universidade Federal de Juiz de Fora, Juiz de Fora, Minas Gerais, Brazil.
| |
Collapse
|
31
|
The Potential Use of Natural and Structural Analogues of Antimicrobial Peptides in the Fight against Neglected Tropical Diseases. Molecules 2015; 20:15392-433. [PMID: 26305243 PMCID: PMC6332049 DOI: 10.3390/molecules200815392] [Citation(s) in RCA: 38] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2015] [Revised: 08/02/2015] [Accepted: 08/10/2015] [Indexed: 12/13/2022] Open
Abstract
Recently, research into the development of new antimicrobial agents has been driven by the increase in resistance to traditional antibiotics and Emerging Infectious Diseases. Antimicrobial peptides (AMPs) are promising candidates as alternatives to current antibiotics in the treatment and prevention of microbial infections. AMPs are produced by all known living species, displaying direct antimicrobial killing activity and playing an important role in innate immunity. To date, more than 2000 AMPs have been discovered and many of these exhibit broad-spectrum antibacterial, antiviral and anti-parasitic activity. Neglected tropical diseases (NTDs) are caused by a variety of pathogens and are particularly wide-spread in low-income and developing regions of the world. Alternative, cost effective treatments are desperately needed to effectively battle these medically diverse diseases. AMPs have been shown to be effective against a variety of NTDs, including African trypanosomes, leishmaniosis and Chagas disease, trachoma and leprosy. In this review, the potential of selected AMPs to successfully treat a variety of NTD infections will be critically evaluated.
Collapse
|
32
|
de Bruycker-Nogueira F, Nogueira RMR, Faria NRDC, Simões JBS, Nunes PCG, de Filippis AMB, dos Santos FB. Insights of the genetic diversity of DENV-1 detected in Brazil in 25 years: Analysis of the envelope domain III allows lineages characterization. INFECTION GENETICS AND EVOLUTION 2015; 34:126-36. [PMID: 26160541 DOI: 10.1016/j.meegid.2015.07.007] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/17/2015] [Revised: 07/03/2015] [Accepted: 07/04/2015] [Indexed: 01/03/2023]
Abstract
Dengue virus type 1 (DENV-1) was first isolated in Brazil in 1986 in the state of Rio de Janeiro (RJ) and during 25years, this serotype emerged and re-emerged causing explosive epidemics in the country. Here, we aimed to present the phylogeny and molecular characterization based on the envelope gene (E) of DENV-1 (n=48) isolated during epidemics occurred from 1986 to 2011. Six full coding region genomes of DENV-1 were fully sequenced and possible genomic recombination events were analyzed. The results showed that the Brazilian DENV-1 isolates analyzed belong to genotype V (Americas/Africa), but grouping into distinct clades. Three groups were identified, one dating from 1986 to 2002 (lineage 1a), a second group isolated from 2009 to 2011 and a representative strain isolated in 2002 (lineage 2), and a group of strains isolated from 2010 to 2011 (lineage 1b). The lineages 1a and 1b were more closely related to the American strains, while lineage 2 to the Asian strains. Amino acids (aa) substitutions were observed in the domains I and III of the E protein and were associated to the lineages segregation. A substitution on E297 differentiated the lineage 1a from the lineages 1b and 2. Substitutions on E338, E394 (domain III), E428 and E436 (stem region) differentiated lineages 1a, 1b and 2. With the exception of the C gene, all the others genes analyzed allowed the DENV-1 classification into the distinct genotypes. Interestingly, the E gene's domain III and stem regions alone were able to characterize the distinct lineages, as observed by the analysis of the entire E gene and the complete coding region. No recombinant events were detected, but a strain belonging to lineage 1a was closely related to a known recombinant strain (AF513110/BR/2001).
Collapse
Affiliation(s)
| | - Rita Maria Ribeiro Nogueira
- Laboratory of Flavivirus, Oswaldo Cruz Institute-FIOCRUZ, Av. Brasil 4365, 21045-900 Rio de Janeiro, RJ, Brazil
| | | | | | | | - Ana Maria Bispo de Filippis
- Laboratory of Flavivirus, Oswaldo Cruz Institute-FIOCRUZ, Av. Brasil 4365, 21045-900 Rio de Janeiro, RJ, Brazil
| | - Flávia Barreto dos Santos
- Laboratory of Flavivirus, Oswaldo Cruz Institute-FIOCRUZ, Av. Brasil 4365, 21045-900 Rio de Janeiro, RJ, Brazil.
| |
Collapse
|
33
|
Analogs of LDL Receptor Ligand Motifs in Dengue Envelope and Capsid Proteins as Potential Codes for Cell Entry. ACTA ACUST UNITED AC 2015; 2015. [PMID: 27123468 DOI: 10.1155/2015/646303] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022]
Abstract
It is established that cell entry of low density lipoprotein particles (LLPs) containing Apo B100 and Apo E is mediated by receptors and GAGs. Receptor ligand motifs, XBBBXXBX, XBBXBX, and ΨBΨXB, and mono- and bipartite NLS sequences are abundant in Apo E and Apo B100 as well as in envelope and capsid proteins of Dengue viruses 1-4 (DENV1-4). Synthetic, fluorescence-labeled peptides of sequences in DENV2 envelope protein, and DENV3 capsid that include these motifs were used to conduct a qualitative assessment of cell binding and entry capacity using HeLa cells. DENV2 envelope peptide, Dsp2EP, 0564Gly-Gly0595, was shown to bind and remain at the cell surface. In contrast, DENV3 capsid protein peptide, Dsp3CP, 0002Asn-Gln0028, readily enters HeLa cells and accumulates at discrete loci in the nucleus. FITC-labeled dengue synthetic peptides colocalize with Low Density Lipoprotein-CM-DiI and Apo E-CM-DiI to a degree that suggests that Dengue viruses may utilize cell entry pathways used by LLPs.
Collapse
|
34
|
Suzarte E, Gil L, Valdés I, Marcos E, Lazo L, Izquierdo A, García A, López L, Álvarez M, Pérez Y, Castro J, Romero Y, Guzmán MG, Guillén G, Hermida L. A novel tetravalent formulation combining the four aggregated domain III-capsid proteins from dengue viruses induces a functional immune response in mice and monkeys. Int Immunol 2015; 27:367-79. [PMID: 25795768 DOI: 10.1093/intimm/dxv011] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2014] [Accepted: 03/16/2015] [Indexed: 11/14/2022] Open
Abstract
Our group developed a subunit vaccine candidate against dengue virus based on two different viral regions: the domain III of the envelope protein and the capsid protein. The novel chimeric protein from dengue-2 virus [domain III-capsid (DIIIC-2)], when presented as aggregated incorporating oligodeoxynucleotides, induced anti-viral and neutralizing antibodies, a cellular immune response and conferred significant protection to mice and monkeys. The remaining constructs were already obtained and properly characterized. Based on this evidence, this work was aimed at assessing the immune response in mice of the chimeric proteins DIIIC of each serotype, as monovalent and tetravalent formulations. Here, we demonstrated the immunogenicity of each protein in terms of humoral and cell-mediated immunity, without antigen competition on the mixture forming the formulation tetra DIIIC. Accordingly, significant protection was afforded as measured by the limited viral load in the mouse encephalitis model. The assessment of the tetravalent formulation in non-human primates was also conducted. In this animal model, it was demonstrated that the formulation induced neutralizing antibodies and memory cell-mediated immune response with IFN-γ-secreting and cytotoxic capacity, regardless the route of immunization used. Taken together, we can assert that the tetravalent formulation of DIIIC proteins constitutes a promising vaccine candidate against dengue virus, and propose it for further efficacy experiments in monkeys or in the dengue human infection model, as it has been recently proposed.
Collapse
Affiliation(s)
- Edith Suzarte
- Vaccine Division, Center for Genetic Engineering and Biotechnology, Havana 6 10 600, Cuba
| | - Lázaro Gil
- Vaccine Division, Center for Genetic Engineering and Biotechnology, Havana 6 10 600, Cuba
| | - Iris Valdés
- Vaccine Division, Center for Genetic Engineering and Biotechnology, Havana 6 10 600, Cuba
| | - Ernesto Marcos
- Vaccine Division, Center for Genetic Engineering and Biotechnology, Havana 6 10 600, Cuba
| | - Laura Lazo
- Vaccine Division, Center for Genetic Engineering and Biotechnology, Havana 6 10 600, Cuba
| | - Alienys Izquierdo
- Pan American Health Organization/Word Health Organization Collaborating Center for the Study of Dengue and its Vector, Department of Virology, Pedro Kourí Tropical Medicine Institute, Havana 13 11 400, Cuba
| | - Angélica García
- Pan American Health Organization/Word Health Organization Collaborating Center for the Study of Dengue and its Vector, Department of Virology, Pedro Kourí Tropical Medicine Institute, Havana 13 11 400, Cuba
| | - Lázaro López
- Vaccine Division, Center for Genetic Engineering and Biotechnology, Havana 6 10 600, Cuba
| | - Maylin Álvarez
- Pan American Health Organization/Word Health Organization Collaborating Center for the Study of Dengue and its Vector, Department of Virology, Pedro Kourí Tropical Medicine Institute, Havana 13 11 400, Cuba
| | - Yusleydis Pérez
- Vaccine Division, Center for Genetic Engineering and Biotechnology, Havana 6 10 600, Cuba
| | - Jorge Castro
- Vaccine Division, Center for Genetic Engineering and Biotechnology, Havana 6 10 600, Cuba
| | - Yaremis Romero
- Vaccine Division, Center for Genetic Engineering and Biotechnology, Havana 6 10 600, Cuba
| | - María G Guzmán
- Pan American Health Organization/Word Health Organization Collaborating Center for the Study of Dengue and its Vector, Department of Virology, Pedro Kourí Tropical Medicine Institute, Havana 13 11 400, Cuba
| | - Gerardo Guillén
- Vaccine Division, Center for Genetic Engineering and Biotechnology, Havana 6 10 600, Cuba
| | - Lisset Hermida
- Vaccine Division, Center for Genetic Engineering and Biotechnology, Havana 6 10 600, Cuba
| |
Collapse
|
35
|
Colombelli C, Palmisano M, Eshed-Eisenbach Y, Zambroni D, Pavoni E, Ferri C, Saccucci S, Nicole S, Soininen R, McKee KK, Yurchenco PD, Peles E, Wrabetz L, Feltri ML. Perlecan is recruited by dystroglycan to nodes of Ranvier and binds the clustering molecule gliomedin. J Cell Biol 2015; 208:313-29. [PMID: 25646087 PMCID: PMC4315246 DOI: 10.1083/jcb.201403111] [Citation(s) in RCA: 34] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2014] [Accepted: 12/18/2014] [Indexed: 01/09/2023] Open
Abstract
Fast neural conduction requires accumulation of Na(+) channels at nodes of Ranvier. Dedicated adhesion molecules on myelinating cells and axons govern node organization. Among those, specific laminins and dystroglycan complexes contribute to Na(+) channel clustering at peripheral nodes by unknown mechanisms. We show that in addition to facing the basal lamina, dystroglycan is found near the nodal matrix around axons, binds matrix components, and participates in initial events of nodogenesis. We identify the dystroglycan-ligand perlecan as a novel nodal component and show that dystroglycan is required for the selective accumulation of perlecan at nodes. Perlecan binds the clustering molecule gliomedin and enhances clustering of node of Ranvier components. These data show that proteoglycans have specific roles in peripheral nodes and indicate that peripheral and central axons use similar strategies but different molecules to form nodes of Ranvier. Further, our data indicate that dystroglycan binds free matrix that is not organized in a basal lamina.
Collapse
Affiliation(s)
- Cristina Colombelli
- Division of Genetics and Cell Biology, San Raffaele Hospital, 20132 Milan, Italy
| | - Marilena Palmisano
- Division of Genetics and Cell Biology, San Raffaele Hospital, 20132 Milan, Italy Department of Biochemistry and Department of Neurology, Hunter James Kelly Research Institute, School of Medicine and Biomedical Sciences, University at Buffalo, Buffalo, NY 14203 Department of Biochemistry and Department of Neurology, Hunter James Kelly Research Institute, School of Medicine and Biomedical Sciences, University at Buffalo, Buffalo, NY 14203
| | - Yael Eshed-Eisenbach
- Department of Molecular Cell Biology, The Weizmann Institute of Science, Rehovot 76100, Israel
| | - Desirée Zambroni
- Division of Genetics and Cell Biology, San Raffaele Hospital, 20132 Milan, Italy
| | - Ernesto Pavoni
- Division of Genetics and Cell Biology, San Raffaele Hospital, 20132 Milan, Italy
| | - Cinzia Ferri
- Division of Genetics and Cell Biology, San Raffaele Hospital, 20132 Milan, Italy
| | - Stefania Saccucci
- Division of Genetics and Cell Biology, San Raffaele Hospital, 20132 Milan, Italy
| | - Sophie Nicole
- Institut du Cerveau et de la Moelle Épinière, 75013 Paris, France Institut National de la Santé et de la Recherche Médicale, U1127, 75019 Paris, France Sorbonne Universités, Université Pierre et Marie Currie, UMRS1127, 75252 Paris, France Centre National de la Recherche Scientifique, UMR 7225, 75013 Paris, France
| | - Raija Soininen
- Oulu Center for Cell-Extracellular Matrix Research, University of Oulu, 90014 Oulu, Finland
| | | | | | - Elior Peles
- Department of Molecular Cell Biology, The Weizmann Institute of Science, Rehovot 76100, Israel
| | - Lawrence Wrabetz
- Division of Genetics and Cell Biology, San Raffaele Hospital, 20132 Milan, Italy Department of Biochemistry and Department of Neurology, Hunter James Kelly Research Institute, School of Medicine and Biomedical Sciences, University at Buffalo, Buffalo, NY 14203 Department of Biochemistry and Department of Neurology, Hunter James Kelly Research Institute, School of Medicine and Biomedical Sciences, University at Buffalo, Buffalo, NY 14203
| | - M Laura Feltri
- Division of Genetics and Cell Biology, San Raffaele Hospital, 20132 Milan, Italy Department of Biochemistry and Department of Neurology, Hunter James Kelly Research Institute, School of Medicine and Biomedical Sciences, University at Buffalo, Buffalo, NY 14203 Department of Biochemistry and Department of Neurology, Hunter James Kelly Research Institute, School of Medicine and Biomedical Sciences, University at Buffalo, Buffalo, NY 14203
| |
Collapse
|
36
|
Ramakrishnan L, Pillai MR, Nair RR. Dengue vaccine development: strategies and challenges. Viral Immunol 2014; 28:76-84. [PMID: 25494228 DOI: 10.1089/vim.2014.0093] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022] Open
Abstract
Infection with dengue virus may result in dengue fever or a more severe outcome, such as dengue hemorrhagic syndrome/shock. Dengue virus infection poses a threat to endemic regions for four reasons: the presence of four serotypes, each with the ability to cause a similar disease outcome, including fatality; difficulties related to vector control; the lack of specific treatment; and the nonavailability of a suitable vaccine. Vaccine development is considered challenging due to the severity of the disease observed in individuals who have acquired dengue-specific immunity, either passively or actively. Therefore, the presence of vaccine-induced immunity against a particular serotype may prime an individual to severe disease on exposure to dengue virus. Vaccine development strategies include live attenuated vaccines, chimeric, DNA-based, subunit, and inactivated vaccines. Each of the candidates is in various stages of preclinical and clinical development. Issues pertaining to selection pressures, viral interaction, and safety still need to be evaluated in order to induce a complete protective immune response against all four serotypes. This review highlights the various strategies that have been employed in vaccine development, and identifies the obstacles to producing a safe and effective vaccine.
Collapse
Affiliation(s)
- Lakshmy Ramakrishnan
- 1 Laboratory Medicine and Molecular Diagnostics, Rajiv Gandhi Centre for Biotechnology , Trivandrum, India
| | | | | |
Collapse
|
37
|
Vervaeke P, Vermeire K, Liekens S. Endothelial dysfunction in dengue virus pathology. Rev Med Virol 2014; 25:50-67. [PMID: 25430853 DOI: 10.1002/rmv.1818] [Citation(s) in RCA: 42] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2014] [Revised: 10/07/2014] [Accepted: 10/14/2014] [Indexed: 02/04/2023]
Abstract
Dengue virus (DENV) is a leading cause of illness and death, mainly in the (sub)tropics, where it causes dengue fever and/or the more serious diseases dengue hemorrhagic fever and dengue shock syndrome that are associated with changes in vascular permeability. Despite extensive research, the pathogenesis of DENV is still poorly understood and, although endothelial cells represent the primary fluid barrier of the blood vessels, the extent to which these cells contribute to DENV pathology is still under debate. The primary target cells for DENV are dendritic cells and monocytes/macrophages that release various chemokines and cytokines upon infection, which can activate the endothelium and are thought to play a major role in DENV-induced vascular permeability. However, recent studies indicate that DENV also replicates in endothelial cells and that DENV-infected endothelial cells may directly contribute to viremia, immune activation, vascular permeability and immune targeting of the endothelium. Also, the viral non-structural protein-1 and antibodies directed against this secreted protein have been reported to be involved in endothelial cell dysfunction. This review provides an extensive overview of the effects of DENV infection on endothelial cell physiology and barrier function.
Collapse
Affiliation(s)
- Peter Vervaeke
- KU Leuven, Rega Institute for Medical Research, Leuven, Belgium
| | | | | |
Collapse
|
38
|
Zhang ZS, Weng YW, Huang HL, Zhang JM, Yan YS. Neutralizing antibodies respond to a bivalent dengue DNA vaccine or/and a recombinant bivalent antigen. Mol Med Rep 2014; 11:1009-16. [PMID: 25371092 DOI: 10.3892/mmr.2014.2850] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2013] [Accepted: 06/10/2014] [Indexed: 11/06/2022] Open
Abstract
There is currently no effective vaccine to prevent dengue infection, despite the existence of multiple studies on potential methods of immunization. The aim of the present study was to explore the effect of DNA and/or recombinant protein on levels of neutralizing antibodies. For this purpose, envelope domain IIIs of dengue serotypes 1 and 2 (DEN-1/2)were spliced by a linker (Gly‑Gly‑Ser‑Gly‑Ser)3 and cloned into the prokaryotic expression plasmid pET30a (+) and eukaryotic vector pcDNA3.1 (+). The chimeric bivalent protein was expressed in Escherichia coli, and one‑step purification by high‑performance liquid chromatography was conducted. Protein expression levels of the DNA plasmid were tested in BHK‑21 cells by indirect immunofluorescent assay. In order to explore a more effective immunization strategy and to develop neutralizing antibodies against the two serotypes, mice were inoculated with recombinant bivalent protein, the DNA vaccine, or the two given simultaneously. Presence of the specific antibodies was tested by ELISA and the presence of the neutralizing antibodies was determined by plaque reduction neutralization test. Results of the analysis indicated that the use of a combination of DNA and protein induced significantly higher titers of neutralizing antibodies against either DEN‑1 or DEN‑2 (1:64.0 and 1:76.1, respectively) compared with the DNA (1:24.7 and 1:26.9, DEN‑1 and DEN‑2, respectively) or the recombinant protein (1:34.9 and 1:45.3 in DEN‑1 and DEN‑2, respectively). The present study demonstrated that the combination of recombinant protein and DNA as an immunization strategy may be an effective method for the development of a vaccine to prevent dengue virus infection.
Collapse
Affiliation(s)
- Zhi-Shan Zhang
- Clinical Laboratory, Affiliated Quanzhou First Hospital of Fujian Medical University, Fuzhou, Fujian 350001, P.R. China
| | - Yu-Wei Weng
- Fujian Center for Disease Control and Prevention, Fuzhou, Fujian 350001, P.R. China
| | - Hai-Long Huang
- Fujian Center for Disease Control and Prevention, Fuzhou, Fujian 350001, P.R. China
| | - Jian-Ming Zhang
- Fujian Center for Disease Control and Prevention, Fuzhou, Fujian 350001, P.R. China
| | - Yan-Sheng Yan
- Fujian Center for Disease Control and Prevention, Fuzhou, Fujian 350001, P.R. China
| |
Collapse
|
39
|
Lisova O, Belkadi L, Bedouelle H. Direct and indirect interactions in the recognition between a cross-neutralizing antibody and the four serotypes of dengue virus. J Mol Recognit 2014; 27:205-14. [PMID: 24591178 DOI: 10.1002/jmr.2352] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2013] [Revised: 12/16/2013] [Accepted: 12/16/2013] [Indexed: 11/11/2022]
Abstract
Dengue fever is the most important vector-borne viral disease. Four serotypes of dengue virus, DENV1 to DENV4, coexist. Secondary infection by a different serotype is a risk factor for severe dengue. Monoclonal antibody mAb4E11 neutralizes the four serotypes of DENV with varying efficacies by recognizing an epitope located within domain-III (ED3) of the viral envelope (E) protein. To better understand the cross-reactivities between mAb4E11 and the four serotypes of DENV, we constructed mutations in both Fab4E11 fragment and ED3, and we searched for indirect interactions in the crystal structures of the four complexes. According to the serotype, 7 to 12 interactions are mediated by one water molecule, 1 to 10 by two water molecules, and several of these interactions are conserved between serotypes. Most interfacial water molecules make hydrogen bonds with both antibody and antigen. Some residues or atomic groups are engaged in both direct and water-mediated interactions. The doubly-indirect interactions are more numerous in the complex of lowest affinity. The third complementarity determining region of the light chain (L-CDR3) of mAb4E11 does not contact ED3. The structures and double-mutant thermodynamic cycles showed that the effects of (hyper)-mutations in L-CDR3 on affinity were caused by conformational changes and indirect interactions with ED3 through other CDRs. Exchanges of residues between ED3 serotypes showed that their effects on affinity were context dependent. Thus, conformational changes, structural context, and indirect interactions should be included when studying cross-reactivity between antibodies and different serotypes of viral antigens for a better design of diagnostics, vaccine, and therapeutic tools against DENV and other Flaviviruses.
Collapse
Affiliation(s)
- Olesia Lisova
- Institut Pasteur, Unit of Molecular Prevention and Therapy of Human Diseases, Department of Infection and Epidemiology, rue du Dr. Roux, F-75015, Paris, France; CNRS, URA3012, rue du Dr. Roux, F-75015, Paris, France
| | | | | |
Collapse
|
40
|
Saiyasombat R, Carrillo-Tripp J, Miller WA, Bredenbeek PJ, Blitvich BJ. Substitution of the premembrane and envelope protein genes of Modoc virus with the homologous sequences of West Nile virus generates a chimeric virus that replicates in vertebrate but not mosquito cells. Virol J 2014; 11:150. [PMID: 25151534 PMCID: PMC4148964 DOI: 10.1186/1743-422x-11-150] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2014] [Accepted: 08/19/2014] [Indexed: 11/23/2022] Open
Abstract
Background Most known flaviviruses, including West Nile virus (WNV), are maintained in natural transmission cycles between hematophagous arthropods and vertebrate hosts. Other flaviviruses such as Modoc virus (MODV) and Culex flavivirus (CxFV) have host ranges restricted to vertebrates and insects, respectively. The genetic elements that modulate the differential host ranges and transmission cycles of these viruses have not been identified. Methods Fusion polymerase chain reaction (PCR) was used to replace the capsid (C), premembrane (prM) and envelope (E) genes and the prM-E genes of a full-length MODV infectious cDNA clone with the corresponding regions of WNV and CxFV. Fusion products were directly transfected into baby hamster kidney-derived cells that stably express T7 RNA polymerase. At 4 days post-transfection, aliquots of each supernatant were inoculated onto vertebrate (BHK-21 and Vero) and mosquito (C6/36) cells which were then assayed for evidence of viral infection by reverse transcription-PCR, Western blot and plaque assay. Results Chimeric virus was recovered in cells transfected with the fusion product containing the prM-E genes of WNV. The virus could infect vertebrate but not mosquito cells. The in vitro replication kinetics and yields of the chimeric virus were similar to MODV but the chimeric virus produced larger plaques. Chimeric virus was not recovered in cells transfected with any of the other fusion products. Conclusions Our data indicate that genetic elements outside of the prM-E gene region of MODV condition its vertebrate-specific phenotype.
Collapse
Affiliation(s)
| | | | | | | | - Bradley J Blitvich
- Department of Veterinary Microbiology and Preventive Medicine, College of Veterinary Medicine, Iowa State University, Ames, Iowa, USA.
| |
Collapse
|
41
|
Huerta V, Toledo P, Fleitas N, Martín A, Pupo D, Yero A, Sarría M, Sánchez A, Besada V, Ramos Y, Márquez G, Guirola O, Chinea G. Receptor-activated human α2-macroglobulin interacts with the envelope protein of dengue virus and protects virions from temperature-induced inactivation through multivalent binding. J Gen Virol 2014; 95:2668-2676. [PMID: 25100798 DOI: 10.1099/vir.0.068544-0] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022] Open
Abstract
Based on the hypothesis that interactions between virions and serum components may influence the outcome of dengue virus (DENV) infections, we decided to use affinity chromatography with domain III from the envelope (E) protein of DENV2 (DIIIE2) as a ligand to isolate virus-binding proteins from human plasma. This approach yielded serum amyloid P (SAP) and α2-macroglobulin (α2M) as novel viral interactors. After confirming the specific binding of both SAP and α2M to DIIIE2 by ELISA, the latter interaction was examined in greater detail. We obtain evidence suggesting that the binding species was actually the receptor-activated form of α2M (α2M*), that α2M* could bind monovalently to recombinant domain III from all four DENV serotypes with affinities in the micromolar range ranking as DENV4>DENV1~DENV2>DENV3 and that this interaction exhibited a strong avidity effect when multivalent binding was favoured (KD 8 × 10(-8) M for DIIIE2). We also showed that α2M* bound to DENV virions of the four serotypes, protecting the virus from temperature-induced inactivation in the absence of serum and enhancing infectivity. The latter effect exhibited an ED50 of 2.9 × 10(-8) M, also suggesting an avidity effect due to multivalent binding. These results will further contribute to the characterization of the virus-host factor interaction network during human DENV infection.
Collapse
Affiliation(s)
- Vivian Huerta
- Center for Genetic Engineering and Biotechnology (CIGB), PO Box 6162, Havana 10600, Cuba
| | - Patricia Toledo
- Center for Genetic Engineering and Biotechnology (CIGB), PO Box 6162, Havana 10600, Cuba
| | - Noralvis Fleitas
- Center for Genetic Engineering and Biotechnology (CIGB), PO Box 6162, Havana 10600, Cuba
| | - Alejandro Martín
- Center for Genetic Engineering and Biotechnology (CIGB), PO Box 6162, Havana 10600, Cuba
| | - Dianne Pupo
- Center for Genetic Engineering and Biotechnology (CIGB), PO Box 6162, Havana 10600, Cuba
| | - Alexis Yero
- Center for Genetic Engineering and Biotechnology (CIGB), PO Box 6162, Havana 10600, Cuba
| | - Mónica Sarría
- Center for Genetic Engineering and Biotechnology (CIGB), PO Box 6162, Havana 10600, Cuba
| | - Aniel Sánchez
- Center for Genetic Engineering and Biotechnology (CIGB), PO Box 6162, Havana 10600, Cuba
| | - Vladimir Besada
- Center for Genetic Engineering and Biotechnology (CIGB), PO Box 6162, Havana 10600, Cuba
| | - Yassel Ramos
- Center for Genetic Engineering and Biotechnology (CIGB), PO Box 6162, Havana 10600, Cuba
| | - Gabriel Márquez
- Center for Genetic Engineering and Biotechnology (CIGB), PO Box 6162, Havana 10600, Cuba
| | - Osmany Guirola
- Center for Genetic Engineering and Biotechnology (CIGB), PO Box 6162, Havana 10600, Cuba
| | - Glay Chinea
- Center for Genetic Engineering and Biotechnology (CIGB), PO Box 6162, Havana 10600, Cuba
| |
Collapse
|
42
|
Dalrymple NA, Mackow ER. Virus interactions with endothelial cell receptors: implications for viral pathogenesis. Curr Opin Virol 2014; 7:134-40. [PMID: 25063986 PMCID: PMC4206553 DOI: 10.1016/j.coviro.2014.06.006] [Citation(s) in RCA: 48] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2014] [Revised: 06/10/2014] [Accepted: 06/27/2014] [Indexed: 01/14/2023]
Abstract
The endothelial lining of the vasculature performs a vital role in maintaining fluid barrier functions despite balancing nutrient and fluid content of tissues, repairing localized damage, coordinating responses of a plethora of factors, immune cells and platelets through a multitude of endothelial cell surface receptors. Viruses that nonlytically cause lethal hemorrhagic or edematous diseases engage receptors on vascular and lymphatic endothelial cells, altering normal cellular responses that control capillary leakage and fluid clearance functions with lethal consequences. Recent studies indicate that receptors directing dengue virus and hantavirus infection of the endothelium contribute to the dysregulation of normal endothelial cell signaling responses that control capillary permeability and immune responses that contribute to pathogenesis. Here we present recent studies of virally altered endothelial functions that provide new insight into targeting barrier functions of the endothelium as a potential therapeutic approach.
Collapse
|
43
|
Nguyen NL, So KK, Kim JM, Kim SH, Jang YS, Yang MS, Kim DH. Expression and characterization of an M cell-specific ligand-fused dengue virus tetravalent epitope using Saccharomyces cerevisiae. J Biosci Bioeng 2014; 119:19-27. [PMID: 25027708 DOI: 10.1016/j.jbiosc.2014.06.005] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2014] [Revised: 05/19/2014] [Accepted: 06/11/2014] [Indexed: 11/29/2022]
Abstract
A fusion construct (Tet-EDIII-Co1) consisting of an M cell-specific peptide ligand (Co1) at the C-terminus of a recombinant tetravalent gene encoding the amino acid sequences of dengue envelope domain III (Tet-EDIII) from four serotypes was expressed and tested for binding activity to the mucosal immune inductive site M cells for the development of an oral vaccine. The yeast episomal expression vector, pYEGPD-TER, which was designed to direct gene expression using the glyceraldehyde-3-phosphate dehydrogenase (GPD) promoter, a functional signal peptide of the amylase 1A protein from rice, and the GAL7 terminator, was used to clone the Tet-EDIII-Co1 gene and resultant plasmids were then used to transform Saccharomyces cerevisiae. PCR and back-transformation into Escherichia coli confirmed the presence of the Tet-EDIII-Co1 gene-containing plasmid in transformants. Northern blot analysis of transformed S. cerevisiae identified the presence of the Tet-EDIII-Co1-specific transcript. Western blot analysis indicated that the produced Tet-EDIII-Co1 protein with the expected molecular weight was successfully secreted into the culture medium. Quantitative Western blot analysis and ELISA revealed that the recombinant Tet-EDIII-Co1 protein comprised approximately 0.1-0.2% of cell-free extracts (CFEs). In addition, 0.1-0.2 mg of Tet-EDIII-Co1 protein per liter of culture filtrate was detected on day 1, and this quantity peaked on day 3 after cultivation. In vivo binding assays showed that the Tet-EDIII-Co1 protein was delivered specifically to M cells in Peyer's patches (PPs) while the Tet-EDIII protein lacking the Co1 ligand did not, which demonstrated the efficient targeting of this antigenic protein through the mucosal-specific ligand.
Collapse
Affiliation(s)
- Ngoc-Luong Nguyen
- Research Center of Bioactive Materials, Center for Fungal Pathogenesis, Chonbuk National University, Jeonju, Chonbuk 561-756, Republic of Korea
| | - Kum-Kang So
- Research Center of Bioactive Materials, Center for Fungal Pathogenesis, Chonbuk National University, Jeonju, Chonbuk 561-756, Republic of Korea
| | - Jung-Mi Kim
- Department of Bio-Environmental Chemistry, Wonkwang University, Iksan, Chonbuk 570-749, Republic of Korea
| | - Sae-Hae Kim
- Research Center of Bioactive Materials, Center for Fungal Pathogenesis, Chonbuk National University, Jeonju, Chonbuk 561-756, Republic of Korea
| | - Yong-Suk Jang
- Research Center of Bioactive Materials, Center for Fungal Pathogenesis, Chonbuk National University, Jeonju, Chonbuk 561-756, Republic of Korea
| | - Moon-Sik Yang
- Research Center of Bioactive Materials, Center for Fungal Pathogenesis, Chonbuk National University, Jeonju, Chonbuk 561-756, Republic of Korea
| | - Dae-Hyuk Kim
- Research Center of Bioactive Materials, Center for Fungal Pathogenesis, Chonbuk National University, Jeonju, Chonbuk 561-756, Republic of Korea.
| |
Collapse
|
44
|
Lazo L, Izquierdo A, Suzarte E, Gil L, Valdés I, Marcos E, Álvarez M, Romero Y, Guzmán MG, Guillén G, Hermida Cruz L. Evaluation in mice of the immunogenicity and protective efficacy of a tetravalent subunit vaccine candidate against dengue virus. Microbiol Immunol 2014; 58:219-26. [DOI: 10.1111/1348-0421.12140] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2013] [Revised: 01/16/2014] [Accepted: 01/25/2014] [Indexed: 01/12/2023]
Affiliation(s)
- Laura Lazo
- Vaccines Division; Center for Genetic Engineering and Biotechnology; P.O. Box 6162 Playa Havana 11 600 Cuba
| | - Alienys Izquierdo
- Virology Department; Tropical Medicine Institute “Pedro Kourí,” Pan American Health Organization/World Health Organization Collaborating Center for the Study of Dengue and its Vector; P.O. Box Marianao 13 Havana 11 600 Cuba
| | - Edith Suzarte
- Vaccines Division; Center for Genetic Engineering and Biotechnology; P.O. Box 6162 Playa Havana 11 600 Cuba
| | - Lázaro Gil
- Vaccines Division; Center for Genetic Engineering and Biotechnology; P.O. Box 6162 Playa Havana 11 600 Cuba
| | - Iris Valdés
- Vaccines Division; Center for Genetic Engineering and Biotechnology; P.O. Box 6162 Playa Havana 11 600 Cuba
| | - Ernesto Marcos
- Vaccines Division; Center for Genetic Engineering and Biotechnology; P.O. Box 6162 Playa Havana 11 600 Cuba
| | - Mayling Álvarez
- Virology Department; Tropical Medicine Institute “Pedro Kourí,” Pan American Health Organization/World Health Organization Collaborating Center for the Study of Dengue and its Vector; P.O. Box Marianao 13 Havana 11 600 Cuba
| | - Yaremis Romero
- Vaccines Division; Center for Genetic Engineering and Biotechnology; P.O. Box 6162 Playa Havana 11 600 Cuba
| | - María Guadalupe Guzmán
- Virology Department; Tropical Medicine Institute “Pedro Kourí,” Pan American Health Organization/World Health Organization Collaborating Center for the Study of Dengue and its Vector; P.O. Box Marianao 13 Havana 11 600 Cuba
| | - Gerardo Guillén
- Vaccines Division; Center for Genetic Engineering and Biotechnology; P.O. Box 6162 Playa Havana 11 600 Cuba
| | - Lisset Hermida Cruz
- Vaccines Division; Center for Genetic Engineering and Biotechnology; P.O. Box 6162 Playa Havana 11 600 Cuba
| |
Collapse
|
45
|
Mutagenesis analysis of T380R mutation in the envelope protein of yellow fever virus. Virol J 2014; 11:60. [PMID: 24678844 PMCID: PMC3974419 DOI: 10.1186/1743-422x-11-60] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2014] [Accepted: 03/27/2014] [Indexed: 11/23/2022] Open
Abstract
Background The RGD motif in the mosquito-borne flaviviruses envelope protein domain III (EDIII) FG loop was shown to bind negatively charged cellular molecules and mediate virus entry in mammals. However, its importance in virus entry in the mosquito has not yet been defined. The sequences of RGD motifs are conserved in JEV-serocomplex members primarily transmitted by Culex mosquitoes but absent from members of the DENV serocomplex, which utilize Aedes mosquitoes as vectors. Interestingly, the RGD sequence is present in the attenuated 17D strain of yellow fever virus as a result of the T380R mutation in the EDIII of Asibi strain following extensive in vitro passage in mice and chicken embryos and was found to contribute to the more rapid clearance in mice challenged with 17D. However, viral infectivity and dissemination in mosquitoes had not been evaluated for this mutant. Findings The study utilized the reverse genetics system of YFV and Ae. aegypti RexD WE mosquitoes to assess the impact of a T380R mutation in YFV Asibi and 17D/Asibi M-E chimera. The T380R mutation led to higher infection rates but similar dissemination rates when introduced into the YFV Asibi strain and 17D/Asibi M-E chimera. Conclusions While the increase of the positive charge in EDIII may reduce the virulence of YFV in mice, this mutation favored the establishment of the viral infection in Ae. aegypti. However, such gain in viral infectivity did not increase dissemination in infected mosquitoes.
Collapse
|
46
|
Proteomic identification of dengue virus binding proteins in Aedes aegypti mosquitoes and Aedes albopictus cells. BIOMED RESEARCH INTERNATIONAL 2013; 2013:875958. [PMID: 24324976 PMCID: PMC3842078 DOI: 10.1155/2013/875958] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/08/2013] [Revised: 09/19/2013] [Accepted: 09/25/2013] [Indexed: 12/22/2022]
Abstract
The main vector of dengue in America is the mosquito Aedes aegypti, which is infected by dengue virus (DENV) through receptors of midgut epithelial cells. The envelope protein (E) of dengue virus binds to receptors present on the host cells through its domain III that has been primarily recognized to bind cell receptors. In order to identify potential receptors, proteins from mosquito midgut tissue and C6/36 cells were purified by affinity using columns with the recombinant E protein domain III (rE-DIII) or DENV particles bound covalently to Sepharose 4B to compare and evaluate their performance to bind proteins including putative receptors from female mosquitoes of Ae. aegypti. To determine their identity mass spectrometric analysis of purified proteins separated by polyacrylamide gel electrophoresis was performed. Our results indicate that both viral particles and rE-DIII bound proteins with the same apparent molecular weights of 57 and 67 kDa. In addition, viral particles bound high molecular weight proteins. Purified proteins identified were enolase, beta-adrenergic receptor kinase (beta-ARK), translation elongation factor EF-1 alpha/Tu, and cadherin.
Collapse
|
47
|
Yun SI, Lee YM. Japanese encephalitis: the virus and vaccines. Hum Vaccin Immunother 2013; 10:263-79. [PMID: 24161909 PMCID: PMC4185882 DOI: 10.4161/hv.26902] [Citation(s) in RCA: 176] [Impact Index Per Article: 16.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2013] [Revised: 10/14/2013] [Accepted: 10/22/2013] [Indexed: 12/11/2022] Open
Abstract
Japanese encephalitis (JE) is an infectious disease of the central nervous system caused by Japanese encephalitis virus (JEV), a zoonotic mosquito-borne flavivirus. JEV is prevalent in much of Asia and the Western Pacific, with over 4 billion people living at risk of infection. In the absence of antiviral intervention, vaccination is the only strategy to develop long-term sustainable protection against JEV infection. Over the past half-century, a mouse brain-derived inactivated vaccine has been used internationally for active immunization. To date, however, JEV is still a clinically important, emerging, and re-emerging human pathogen of global significance. In recent years, production of the mouse brain-derived vaccine has been discontinued, but 3 new cell culture-derived vaccines are available in various parts of the world. Here we review current aspects of JEV biology, summarize the 4 types of JEV vaccine, and discuss the potential of an infectious JEV cDNA technology for future vaccine development.
Collapse
Affiliation(s)
- Sang-Im Yun
- Department of Animal, Dairy, and Veterinary Sciences; Utah Science Technology and Research; College of Agriculture and Applied Sciences; Utah State University; Logan, UT USA
| | - Young-Min Lee
- Department of Animal, Dairy, and Veterinary Sciences; Utah Science Technology and Research; College of Agriculture and Applied Sciences; Utah State University; Logan, UT USA
| |
Collapse
|
48
|
García-Machorro J, López-González M, Barrios-Rojas O, Fernández-Pomares C, Sandoval-Montes C, Santos-Argumedo L, Villegas-Sepúlveda N, Gutiérrez-Castañeda B, Cedillo-Barrón L. DENV-2 subunit proteins fused to CR2 receptor-binding domain (P28)-induces specific and neutralizing antibodies to the Dengue virus in mice. Hum Vaccin Immunother 2013; 9:2326-35. [PMID: 23880886 DOI: 10.4161/hv.25673] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022] Open
Abstract
Domain III (DIII) of the dengue virus (DENV) envelope (E) protein induces strong neutralizing type-specific antibodies. In addition, a region near the fusion loop in domain II (DII) induces the production of cross-reactive antibodies with neutralizing potential. Thus, this study aimed to generate DENV-2 recombinant fusion proteins (i.e., rEII*EIII and rEII*EIII/NS1*) either alone or fused to 3 copies of P28, the minimum CR2-binding domain of the complement protein C3d. The 4 recombinant proteins were generated in a Drosophila melanogaster Schneider 2 (S2) cell system. The expression and secretion of the recombinant proteins were confirmed in vitro using immunofluorescence (IF) and western blot (WB) analyses. Human dengue immune serum samples recognized recombinant proteins. The immunogenicity of the 4 proteins in BALB/c mice was analyzed using ELISA, and the results revealed that the induced specific antibody response was higher in the groups of mice immunized with the P28 fusion proteins. Interestingly, although the 4 recombinant proteins were able to elicit high levels of neutralizing antibodies in BALB/c mice; no adjuvant effect was observed in terms of neutralizing antibodies in the groups immunized with proteins containing P28. Thus, ELISA and PRNT50 assays may evaluate different epitopes and responses, where ELISA showed a wider response that did not always correlate with neutralization. Furthermore, the elicited antibodies were able to recognize the immobilized E glycoprotein of DENV. All mice vaccinated with the DENV-2 recombinant proteins showed induction of higher levels of IgG1 antibodies than of IgG2a antibodies.
Collapse
Affiliation(s)
- Jazmín García-Machorro
- Department of Molecular Biomedicine Centre for Research and Advanced Studies (CINVESTAV-IPN) Av. IPN # 2508 Col.; San Pedro Zacatenco, D.F. Mexico, Mexico
| | | | | | | | | | | | | | | | | |
Collapse
|
49
|
Zidane N, Dussart P, Bremand L, Bedouelle H. Cross-reactivities between human IgMs and the four serotypes of dengue virus as probed with artificial homodimers of domain-III from the envelope proteins. BMC Infect Dis 2013; 13:302. [PMID: 23815496 PMCID: PMC3701519 DOI: 10.1186/1471-2334-13-302] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2013] [Accepted: 06/26/2013] [Indexed: 12/23/2022] Open
Abstract
Background Dengue fever is the most important vector-borne viral disease. Four serotypes of dengue virus, DENV1 to DENV4, coexist. Infection by one serotype elicits long-lasting immunity to that serotype but not the other three. Subsequent infection by a different serotype is a risk factor for severe dengue. Domain III (ED3) of the viral envelope protein interacts with cell receptors and contains epitopes recognized by neutralizing antibodies. We determined the serotype specificity and cross-reactivity of human IgMs directed against ED3 by using a well-characterized collection of 90 DENV-infected and 89 DENV-uninfected human serums. Methods The recognitions between the four serotypes of ED3 and the serums were assayed with an IgM antibody-capture ELISA (MAC-ELISA) and artificial homodimeric antigens. The results were analyzed with Receiving Operator Characteristic (ROC) curves. Results The DENV-infected serums contained IgMs that reacted with one or several ED3 serotypes. The discrimination by ED3 between serums infected by the homotypic DENV and uninfected serums varied with the serotype in the decreasing order DENV1 > DENV2 > DENV3 > DENV4. The ED3 domain of DENV1 gave the highest discrimination between DENV-infected and DENV-uninfected serums, whatever the infecting serotype, and thus behaved like a universal ED3 domain for the detection of IgMs against DENV. Some ED3 serotypes discriminated between IgMs directed against the homotypic and heterotypic DENVs. The patterns of cross-reactivities and discriminations varied with the serotype. Conclusions The results should help better understand the IgM immune response and protection against DENV since ED3 is widely used as an antigen in diagnostic assays and an immunogen in vaccine candidates.
Collapse
Affiliation(s)
- Nora Zidane
- Unit of Molecular Prevention and Therapy of Human Diseases, Department of Infection and Epidemiology, Institut Pasteur, Rue du Docteur Roux, F-75015 Paris, France
| | | | | | | |
Collapse
|
50
|
Shah M, Wadood A, Rahman Z, Husnain T. Interaction and inhibition of dengue envelope glycoprotein with mammalian receptor DC-sign, an in-silico approach. PLoS One 2013; 8:e59211. [PMID: 23527139 PMCID: PMC3601059 DOI: 10.1371/journal.pone.0059211] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2012] [Accepted: 02/12/2013] [Indexed: 12/23/2022] Open
Abstract
Membrane fusion is the central molecular event during the entry of enveloped viruses into cells. The critical agents of this process are viral surface proteins, primed to facilitate cell bilayer fusion. The important role of Dendritic-cell-specific ICAM3-grabbing non-integrin (DC-SIGN) in Dengue virus transmission makes it an attractive target to interfere with Dengue virus Propagation. Receptor mediated endocytosis allows the entry of virions due to the presence of endosomal membranes and low pH-induced fusion of the virus. DC-SIGN is the best characterized molecule among the candidate protein receptors and is able to mediate infection with the four serotypes of dengue virus (DENV). Unrestrained pair wise docking was used for the interaction of dengue envelope protein with DC-SIGN and monoclonal antibody 2G12. Pre-processed the PDB coordinates of dengue envelope glycoprotein and other candidate proteins were prepared and energy minimized through AMBER99 force field distributed in MOE software. Protein-protein interaction server, ZDOCK was used to find molecular interaction among the candidate proteins. Based on these interactions it was found that antibody successfully blocks the glycosylation site ASN 67 and other conserved residues present at DC-SIGN-Den-E complex interface. In order to know for certain, the exact location of the antibody in the envelope protein, co-crystallize of the envelope protein with these compounds is needed so that their exact docking locations can be identified with respect to our results.
Collapse
Affiliation(s)
- Masaud Shah
- Bioinformatics Research Laboratory, National Center of Excellence in Molecular Biology (CEMB), University of the Punjab, Lahore, Pakistan
- Computational Medicinal Chemistry Laboratory, Department of Biochemistry Abdul Wali Khan University, Mardan, Pakistan
| | - Abdul Wadood
- Computational Medicinal Chemistry Laboratory, Department of Biochemistry Abdul Wali Khan University, Mardan, Pakistan
- * E-mail: (AW); (ZR)
| | - Ziaur Rahman
- Bioinformatics Research Laboratory, National Center of Excellence in Molecular Biology (CEMB), University of the Punjab, Lahore, Pakistan
- * E-mail: (AW); (ZR)
| | - Tayyab Husnain
- Bioinformatics Research Laboratory, National Center of Excellence in Molecular Biology (CEMB), University of the Punjab, Lahore, Pakistan
| |
Collapse
|