525 results on '"Junin virus"'
Search Results
2. Restoration of virulence in the attenuated Candid#1 vaccine virus requires reversion at both positions 168 and 427 in the envelope glycoprotein GPC.
- Author
-
Nunberg, Jack H., Westover, Jonna B., Joanne York, Kie Hoon Jung, Bailey, Kevin W., Boardman, Kirsten M., Minghao Li, Furnell, Rachel S., Wasson, Samantha R., Murray, Justin S., Kaundal, Rakesh, Thomas, Aaron J., and Gowen, Brian B.
- Subjects
- *
VIRAL vaccines , *TRANSMEMBRANE domains , *GUINEA pigs , *HEMORRHAGIC fever , *VIRUS diseases - Abstract
Live-attenuated virus vaccines provide long-lived protection against viral disease but carry inherent risks of residual pathogenicity and genetic reversion. The live-attenuated Candid#1 vaccine was developed to protect Argentines against lethal infection by the Argentine hemorrhagic fever arenavirus, Junín virus. Despite its safety and efficacy in Phase III clinical study, the vaccine is not licensed in the US, in part due to concerns regarding the genetic stability of attenuation. Previous studies had ident ified a single F427I mutation in the transmembrane domain of the Candid#1 envelope glycoprotein GPC as the key determinant of attenuation, as well as the propensity of this mutation to revert upon passage in cell culture and neonatal mice. To ascertain the consequences of this reversion event, we introduced the I427F mutation into recombinant Candid#1 (I427F rCan) and investigated the effects in two validated small-animal models: in mice expressing the essential virus receptor (human transferrin receptor 1; huTfR1) and in the conventional guinea pig model. We report that I427F rCan displays only modest virulence in huTfR1 mice and appears attenuated in guinea pigs. Reversion at another attenuating locus in Candid#1 GPC (T168A) was also examined, and a similar pattern was observed. By contrast, virus bearing both revertant mutations (A168T+I427F rCan) approached the lethal virulence of the pathogenic Romero strain in huTfR1 mice. Virulence was less extreme in guinea pigs. Our findings suggest that genetic stabilization at both positions is required to minimize the likelihood of reversion to virulence in a second-generation Candid#1 vaccine. IMPORTANCE Live-attenuated virus vaccines, such as measles/mumps/rubella and oral poliovirus, provide robust protection against disease but carry with them the risk of genetic reversion to the virulent form. Here, we analyze the genetics of reversion in the live-attenuated Candid#1 vaccine that is used to protect against Argentine hemorrhagic fever, an often-lethal disease caused by the Junín arenavirus. In two validated smallanimal models, we find that restoration of virulence in recombinant Candid#1 viruses requires back-mutation at two positions specific to the Candid#1 envelope glycoprotein GPC, at positions 168 and 427. Viruses bearing only a single change showed only modest virulence. We discuss strategies to genetically harden Candid#1 GPC against these two reversion events in order to develop a safer second-generation Candid#1 vaccine virus. [ABSTRACT FROM AUTHOR]
- Published
- 2024
- Full Text
- View/download PDF
3. Multi-epitope-based vaccine designing against Junín virus glycoprotein: immunoinformatics approach
- Author
-
Mallari Praveen
- Subjects
Junín virus ,Glycoprotein ,Vaccine designing ,Immunoinformatics ,Molecular docking ,Multi-epitope vaccine ,Therapeutics. Pharmacology ,RM1-950 ,Pharmacy and materia medica ,RS1-441 - Abstract
Abstract Background The Junín virus (JUNV) is well known for causing argentine haemorrhagic fever (AHF), a severe endemic disease in farming premises. The glycoprotein of JUNV is an important therapeutic target in vaccine design. Despite using drugs and neutralizing weakened antibodies being used in the medication, neither the severity reduced nor eradicated the infection. However, this constraint can be resolved by immunoinformatic approaches. Results The glycoprotein fasta sequence was retrieved from NCBI to anticipate the B cell and T cell epitopes through the Immune Epitope Database. Furthermore, each epitope underwent validation in Vaxijen 2.0, Aller Top, and Toxin Pred to find antigenic, nonallergic, and non-toxic peptides. Moreover, the vaccine is designed with appropriate adjuvants and linkers. Subsequently, physicochemical properties were determined in ProtParam including solubility and disulphide bonds in the SCRATCH server. The vaccine 3D structure was built using I-TASSER and refined in ModRefine. Docking between JUNV glycoprotein (PDB ID:5NUZ) with a built vaccine revealed a balanced docked complex visualized in the Drug Discovery studio, identified 280 hydrogen bonds between them. The docking score of − 15.5 kcal/mol was determined in the MM/GBSA analysis in HawkDock. MD simulations employed using the GROMACS at 20 ns resulted in minimal deviation and fewer fluctuations, particularly with high hydrogen bond-forming residues. Conclusion However, these findings present a potential vaccine for developing against JUNV glycoprotein after validating the epitopes and 3D vaccine construct through in silico methods. Therefore, further investigation in the wet laboratory is necessary to confirm the potentiality of the predicted vaccine.
- Published
- 2024
- Full Text
- View/download PDF
4. Multi-epitope-based vaccine designing against Junín virus glycoprotein: immunoinformatics approach
- Author
-
Praveen, Mallari
- Published
- 2024
- Full Text
- View/download PDF
5. Alternative translation contributes to the generation of a cytoplasmic subpopulation of the Junín virus nucleoprotein that inhibits caspase activation and innate immunity.
- Author
-
Bostedt, Linus, Fénéant, Lucie, Leske, Anne, Holzerland, Julia, Günther, Karla, Waßmann, Irke, Bohn, Patrick, and Groseth, Allison
- Subjects
- *
CASPASES , *NATURAL immunity , *RNA synthesis , *DOUBLE-stranded RNA , *CELLULAR inclusions , *CELL anatomy , *NUCLEOPROTEINS , *EXONUCLEASES - Abstract
The highly pathogenic arenavirus, Junín virus (JUNV), expresses three truncated alternative isoforms of its nucleoprotein (NP), i.e., NP53kD, NP47kD, and NP40kD. While both NP47kD and NP40kD have been previously shown to be products of caspase cleavage, here, we show that expression of the third isoform NP53kD is due to alternative in-frame translation from M80. Based on this information, we were able to generate recombinant JUNVs lacking each of these isoforms. Infection with these mutants revealed that, while all three isoforms contribute to the efficient control of caspase activation, NP40kD plays the predominant role. In contrast to full-length NP (i.e., NP65kD), which is localized to inclusion bodies, where viral RNA synthesis takes place, the loss of portions of the N-terminal coiled-coil region in these isoforms leads to a diffuse cytoplasmic distribution and a loss of function in viral RNA synthesis. Nonetheless, NP53kD, NP47kD, and NP40kD all retain robust interferon antagonistic and 3′−5′ exonuclease activities. We suggest that the altered localization of these NP isoforms allows them to be more efficiently targeted by activated caspases for cleavage as decoy substrates, and to be better positioned to degrade viral double-stranded (ds)RNA species that accumulate in the cytoplasm during virus infection and/or interact with cytosolic RNA sensors, thereby limiting dsRNA-mediated innate immune responses. Taken together, this work provides insight into the mechanism by which JUNV leverages apoptosis during infection to generate biologically distinct pools of NP and contributes to our understanding of the expression and biological relevance of alternative protein isoforms during virus infection. IMPORTANCE A limited coding capacity means that RNA viruses need strategies to diversify their proteome. The nucleoprotein (NP) of the highly pathogenic arenavirus Junín virus (JUNV) produces three N-terminally truncated isoforms: two (NP47kD and NP40kD) are known to be produced by caspase cleavage, while, here, we show that NP53kD is produced by alternative translation initiation. Recombinant JUNVs lacking individual NP isoforms revealed that all three isoforms contribute to inhibiting caspase activation during infection, but cleavage to generate NP40kD makes the biggest contribution. Importantly, all three isoforms retain their ability to digest double-stranded (ds)RNA and inhibit interferon promoter activation but have a diffuse cytoplasmic distribution. Given the cytoplasmic localization of both aberrant viral dsRNAs, as well as dsRNA sensors and many other cellular components of innate immune activation pathways, we suggest that the generation of NP isoforms not only contributes to evasion of apoptosis but also robust control of the antiviral response. [ABSTRACT FROM AUTHOR]
- Published
- 2024
- Full Text
- View/download PDF
6. The Estimation of Efficacy of Nonspecific Medications Against Hemorrhagic Fevers Caused by Arenaviruses
- Author
-
Т. Е. Sizikova, V. N. Lebedev, and S. V. Borisevich
- Subjects
hemorrhagic fevers caused by arenaviruses ,junin virus ,lassa virus ,therapeutic ,favipiravir ,Neoplasms. Tumors. Oncology. Including cancer and carcinogens ,RC254-282 - Abstract
The article presents an analysis of studies assessing the effectiveness of new nonspecific medications against hemorrhagic fevers caused by arenaviruses. The possible targets for nonspecific medications, classes of researched antiviral preparations, methods of preclinical investigation of antiviral preparations in vitro and on laboratory animals, as well as prospects for their use in healthcare at present are considered. It has been shown that the level of development of nonspecific medications against hemorrhagic fevers caused by arenaviruses is significantly inferior to those against filovirus infections. Favipiravir should currently be considered as the most effective nonspecific medication against hemorrhagic fevers caused by arenaviruses.
- Published
- 2023
- Full Text
- View/download PDF
7. Host receptor-targeted therapeutic approach to counter pathogenic New World mammarenavirus infections
- Author
-
Hickerson, Brady T, Daniels-Wells, Tracy R, Payes, Cristian, Clark, Lars E, Candelaria, Pierre V, Bailey, Kevin W, Sefing, Eric J, Zink, Samantha, Ziegenbein, James, Abraham, Jonathan, Helguera, Gustavo, Penichet, Manuel L, and Gowen, Brian B
- Subjects
Medical Microbiology ,Biomedical and Clinical Sciences ,Prevention ,Infectious Diseases ,Biodefense ,Emerging Infectious Diseases ,Vaccine Related ,5.1 Pharmaceuticals ,Development of treatments and therapeutic interventions ,Aetiology ,2.1 Biological and endogenous factors ,Infection ,A549 Cells ,Animals ,Antibodies ,Monoclonal ,Antigens ,CD ,Arenaviridae ,Chlorocebus aethiops ,Hemorrhagic Fever ,American ,Host-Pathogen Interactions ,Humans ,Junin virus ,Mice ,Inbred C57BL ,Mice ,Transgenic ,Molecular Docking Simulation ,Protein Binding ,Receptors ,Transferrin ,Vero Cells ,Viral Envelope Proteins - Abstract
Five New World mammarenaviruses (NWMs) cause life-threatening hemorrhagic fever (HF). Cellular entry by these viruses is mediated by human transferrin receptor 1 (hTfR1). Here, we demonstrate that an antibody (ch128.1/IgG1) which binds the apical domain of hTfR1, potently inhibits infection of attenuated and pathogenic NWMs in vitro. Computational docking of the antibody Fab crystal structure onto the known structure of hTfR1 shows an overlapping receptor-binding region shared by the Fab and the viral envelope glycoprotein GP1 subunit that binds hTfR1, and we demonstrate competitive inhibition of NWM GP1 binding by ch128.1/IgG1 as the principal mechanism of action. Importantly, ch128.1/IgG1 protects hTfR1-expressing transgenic mice against lethal NWM challenge. Additionally, the antibody is well-tolerated and only partially reduces ferritin uptake. Our findings provide the basis for the development of a novel, host receptor-targeted antibody therapeutic broadly applicable to the treatment of HF of NWM etiology.
- Published
- 2022
8. Progress toward the development of Lassa vaccines.
- Author
-
Ly, Hinh
- Published
- 2024
- Full Text
- View/download PDF
9. Recent Discoveries of Novel Mammarenaviruses Infecting Humans and Other Mammals in Asia and Southeast Asia
- Author
-
Brigitte Flannery, Michaela Cain, and Hinh Ly
- Subjects
Mammarenavirus ,arenavirus ,Wenzhou virus ,Plateau pika virus ,Lassa virus ,Junin virus ,Infectious and parasitic diseases ,RC109-216 - Abstract
ABSTRACTMammarenaviruses, a genus of the family Arenaviridae, are capable of infecting mammals and are primarily found in rodent reservoirs worldwide. Mammarenaviruses can be transmitted to humans through contact with infected rodents, and though infection is often asymptomatic, some members of this genus can cause viral haemorrhagic fever which has mortality rates ranging from 1% to 50%. These viruses are typically restricted geographically, based on the geographical range of their host reservoirs. Lymphocytic choriomeningitis virus (LCMV) was previously thought to be the only mammarenavirus found across the globe. However, recent discoveries of two novel human mammarenaviruses, Wenzhou Virus (WENV) and Plateau Pika Virus (PPV), in Asia and Southeast Asia show that mammarenaviruses are more widespread than previously thought. This editorial article aims to raise awareness about these emerging viruses, their genetic and ecological diversities, and clinical significance, and to encourage further study of these emerging viruses.
- Published
- 2023
- Full Text
- View/download PDF
10. N6-methyladenosine is required for efficient RNA synthesis of Ebola virus and other haemorrhagic fever viruses
- Author
-
Lisa Wendt, Matthew J. Pickin, Bianca S. Bodmer, Sven Reiche, Lucie Fénéant, Julia E. Hölper, Walter Fuchs, Allison Groseth, and Thomas Hoenen
- Subjects
Ebola virus ,Junín virus ,Crimean-Congo haemorrhagic fever virus ,filovirus ,arenavirus ,orthonairovirus ,Infectious and parasitic diseases ,RC109-216 ,Microbiology ,QR1-502 - Abstract
N6-methyladenosine (m6A) is one of the most abundant modifications of cellular RNA, where it serves various functions. m6A methylation of many viral RNA species has also been described; however, little is known about the m6A epitranscriptome of haemorrhagic fever-causing viruses like Ebola virus (EBOV). Here, we analysed the importance of the methyltransferase METTL3 for the life cycle of this virus. We found that METTL3 interacts with the EBOV nucleoprotein and the transcriptional activator VP30 to support viral RNA synthesis, and that METTL3 is recruited into EBOV inclusions bodies, where viral RNA synthesis occurs. Analysis of the m6A methylation pattern of EBOV mRNAs showed that they are methylated by METTL3. Further studies revealed that METTL3 interaction with the viral nucleoprotein, as well as its importance for RNA synthesis and protein expression, is also observed for other haemorrhagic fever viruses such as Junín virus (JUNV) and Crimean-Congo haemorrhagic fever virus (CCHFV). The negative effects on viral RNA synthesis due to loss of m6A methylation are independent of innate immune sensing, as METTL3 knockout did not affect type I interferon induction in response to viral RNA synthesis or infection. Our results suggest a novel function for m6A that is conserved among diverse haemorrhagic fever-causing viruses (i.e. EBOV, JUNV and CCHFV), making METTL3 a promising target for broadly-acting antivirals.
- Published
- 2023
- Full Text
- View/download PDF
11. Nuclease Activity of the Junín Virus Nucleoprotein C-Terminal Domain.
- Author
-
Sierra, Alicia Armella, Loureiro, María Eugenia, Esperante, Sebastián, Borkosky, Silvia Susana, Gallo, Giovanna L., de Prat Gay, Gonzalo, and Lopez, Nora
- Subjects
- *
NUCLEOPROTEINS , *EXTRACELLULAR matrix proteins , *VIRAL proteins , *RNA synthesis , *DOUBLE-stranded RNA , *HEMORRHAGIC fever - Abstract
The mammarenavirus Junín (JUNV) is the causative agent of Argentine hemorrhagic fever, a severe disease of public health concern. The most abundant viral protein is the nucleoprotein (NP), a multifunctional, two-domain protein with the primary role as structural component of the viral nucleocapsids, used as template for viral polymerase RNA synthesis activities. Here, we report that the C-terminal domain (CTD) of the attenuated Candid#1 strain of the JUNV NP can be purified as a stable soluble form with a secondary structure in line with known NP structures from other mammarenaviruses. We show that the JUNV NP CTD interacts with the viral matrix protein Z in vitro, and that the full-length NP and Z interact with each other in cellulo, suggesting that the NP CTD is responsible for this interaction. This domain comprises an arrangement of four acidic residues and a histidine residue conserved in the active site of exoribonucleases belonging to the DEDDh family. We show that the JUNV NP CTD displays metal-ion-dependent nuclease activity against DNA and single- and double-stranded RNA, and that this activity is impaired by the mutation of a catalytic residue within the DEDDh motif. These results further support this activity, not previously observed in the JUNV NP, which could impact the mechanism of the cellular immune response modulation of this important pathogen. [ABSTRACT FROM AUTHOR]
- Published
- 2023
- Full Text
- View/download PDF
12. Effects of the Natural Flavonoid Quercetin on Arenavirus Junín Infection.
- Author
-
Alvarez De Lauro, Aaron Ezequiel, Pelaez, Miguel Angel, Marquez, Agostina Belén, Wagner, Mariel Selene, Scolaro, Luis Alberto, García, Cybele Carina, Damonte, Elsa Beatriz, and Sepúlveda, Claudia Soledad
- Subjects
- *
ARENAVIRUS diseases , *FLAVONOIDS , *PI3K/AKT pathway , *HEMORRHAGIC fever , *CELL receptors , *QUERCETIN - Abstract
There is no specific chemotherapy approved for the treatment of pathogenic arenaviruses that cause severe hemorrhagic fever (HF) in the population of endemic regions in America and Africa. The present study reports the effects of the natural flavonoid quercetin (QUER) on the infection of A549 and Vero cells with Junín virus (JUNV), agent of the Argentine HF. By infectivity assays, a very effective dose-dependent reduction of JUNV multiplication was shown by cell pretreatment at 2–6 h prior to the infection at non-cytotoxic concentrations, with 50% effective concentration values in the range of 6.1–7.5 µg/mL. QUER was also active by post-infection treatment but with minor efficacy. Mechanistic studies indicated that QUER mainly affected the early steps of virus adsorption and internalization in the multiplication cycle of JUNV. Treatment with QUER blocked the phosphorylation of Akt without changes in the total protein expression, detected by Western blot, and the consequent perturbation of the PI3K/Akt pathway was also associated with the fluorescence redistribution from membrane to cytoplasm of TfR1, the cell receptor recognized by JUNV. Then, it appears that the cellular antiviral state, induced by QUER treatment, leads to the prevention of JUNV entry into the cell. [ABSTRACT FROM AUTHOR]
- Published
- 2023
- Full Text
- View/download PDF
13. The roles of XJ13 and XJ44-specific mutations within the Candid #1 GPC in Junin virus attenuation.
- Author
-
Manning, John Tyler, Junki Maruyama, Wanninger, Timothy, Reyna, Rachel A., Stevenson, Heather L., Bi-Hung Peng, Mantlo, Emily K., Cheng Huang, and Paessler, Slobodan
- Subjects
GUINEA pigs ,VACCINE immunogenicity ,HEMORRHAGIC fever ,RHESUS monkeys ,GENETIC mutation - Abstract
Junin virus (JUNV) is a member of the Arenaviridae family of viruses and is the pathogen responsible for causing Argentine hemorrhagic fever, a potentially lethal disease endemic to Argentina. A live attenuated vaccine for human use, called Candid#1, is approved only in Argentina. Candid#1 vaccine strain of Junin virus was obtained through serial passage in mouse brain tissues followed by passage in Fetal Rhesus macaque lung fibroblast (FRhL) cells. Previously, the mutations responsible for attenuation of this virus in Guinea pigs were mapped in the gene encoding for glycoprotein precursor (GPC) protein. The resulting Candid#1 glycoprotein complex has been shown to cause endoplasmic reticulum (ER) stress in vitro resulting in the degradation of the GPC. To evaluate the attenuating properties of specific mutations within GPC, we created recombinant viruses expressing GPC mutations specific to key Candid#1 passages and evaluated their pathogenicity in our outbred Hartley guinea pig model of Argentine hemorrhagic fever. Here, we provide evidence that early mutations in GPC obtained through serial passaging attenuate the visceral disease and increase immunogenicity in guinea pigs. Specific mutations acquired prior to the 13th mouse brain passage (XJ13) are responsible for attenuation of the visceral disease while having no impact on the neurovirulence of Junin virus. Additionally, our findings demonstrate that the mutation within an N-linked glycosylation motif, acquired prior to the 44th mouse brain passage (XJ44), is unstable but necessary for complete attenuation and enhanced immunogenicity of Candid#1 vaccine strain. The highly conserved N-linked glycosylation profiles of arenavirus glycoproteins could therefore be viable targets for designing attenuating viruses for vaccine development against other arenavirus-associated illnesses. [ABSTRACT FROM AUTHOR]
- Published
- 2023
- Full Text
- View/download PDF
14. The roles of XJ13 and XJ44-specific mutations within the Candid #1 GPC in Junin virus attenuation
- Author
-
John Tyler Manning, Junki Maruyama, Timothy Wanninger, Rachel A. Reyna, Heather L. Stevenson, Bi-Hung Peng, Emily K. Mantlo, Cheng Huang, and Slobodan Paessler
- Subjects
Candid #1 vaccine ,Junin virus ,neurovirulence ,vaccine safety ,arenavirus diseases ,Immunologic diseases. Allergy ,RC581-607 - Abstract
Junin virus (JUNV) is a member of the Arenaviridae family of viruses and is the pathogen responsible for causing Argentine hemorrhagic fever, a potentially lethal disease endemic to Argentina. A live attenuated vaccine for human use, called Candid#1, is approved only in Argentina. Candid#1 vaccine strain of Junin virus was obtained through serial passage in mouse brain tissues followed by passage in Fetal Rhesus macaque lung fibroblast (FRhL) cells. Previously, the mutations responsible for attenuation of this virus in Guinea pigs were mapped in the gene encoding for glycoprotein precursor (GPC) protein. The resulting Candid#1 glycoprotein complex has been shown to cause endoplasmic reticulum (ER) stress in vitro resulting in the degradation of the GPC. To evaluate the attenuating properties of specific mutations within GPC, we created recombinant viruses expressing GPC mutations specific to key Candid#1 passages and evaluated their pathogenicity in our outbred Hartley guinea pig model of Argentine hemorrhagic fever. Here, we provide evidence that early mutations in GPC obtained through serial passaging attenuate the visceral disease and increase immunogenicity in guinea pigs. Specific mutations acquired prior to the 13th mouse brain passage (XJ13) are responsible for attenuation of the visceral disease while having no impact on the neurovirulence of Junin virus. Additionally, our findings demonstrate that the mutation within an N-linked glycosylation motif, acquired prior to the 44th mouse brain passage (XJ44), is unstable but necessary for complete attenuation and enhanced immunogenicity of Candid#1 vaccine strain. The highly conserved N-linked glycosylation profiles of arenavirus glycoproteins could therefore be viable targets for designing attenuating viruses for vaccine development against other arenavirus-associated illnesses.
- Published
- 2023
- Full Text
- View/download PDF
15. Editorial: Insights in virus and host: 2021
- Author
-
John Hiscott and Curtis R. Brandt
- Subjects
KHSV ,RIO kinase ,Junin virus ,Dengue virus ,Chikungunya virus ,Porcine epidemic diarrhea virus ,Microbiology ,QR1-502 - Published
- 2023
- Full Text
- View/download PDF
16. Vaccine Candidates against Arenavirus Infections.
- Author
-
Saito, Takeshi, Reyna, Rachel A., Taniguchi, Satoshi, Littlefield, Kirsten, Paessler, Slobodan, and Maruyama, Junki
- Subjects
ARENAVIRUS diseases ,VIRAL vaccines ,VACCINE approval ,VACCINES ,RECOMBINANT proteins - Abstract
The viral family Arenaviridae contains several members that cause severe, and often lethal, diseases in humans. Several highly pathogenic arenaviruses are classified as Risk Group 4 agents and must be handled in the highest biological containment facility, biosafety level-4 (BSL-4). Vaccines and treatments are very limited for these pathogens. The development of vaccines is crucial for the establishment of countermeasures against highly pathogenic arenavirus infections. While several vaccine candidates have been investigated, there are currently no approved vaccines for arenavirus infection except for Candid#1, a live-attenuated Junin virus vaccine only licensed in Argentina. Current platforms under investigation for use include live-attenuated vaccines, recombinant virus-based vaccines, and recombinant proteins. We summarize here the recent updates of vaccine candidates against arenavirus infections. [ABSTRACT FROM AUTHOR]
- Published
- 2023
- Full Text
- View/download PDF
17. Modulation of the Aryl Hydrocarbon Receptor Signaling Pathway Impacts on Junín Virus Replication.
- Author
-
Pelaez, Miguel Angel, Torti, María Florencia, Alvarez De Lauro, Aaron Ezequiel, Marquez, Agostina Belén, Giovannoni, Federico, Damonte, Elsa Beatriz, and García, Cybele Carina
- Subjects
- *
ARYL hydrocarbon receptors , *IMMUNOMODULATORS , *CELLULAR signal transduction , *VIRAL replication , *VIRAL proteins - Abstract
Junín virus (JUNV), a member of the family Arenaviridae, is the etiological agent of the Argentine hemorrhagic fever, an endemic disease in the rural region of Argentina lacking a specific chemotherapy. Aryl hydrocarbon receptor (AHR) is expressed in several mammalian tissues and has been indicated as a sensor of ligands from variable sources and a modulator of the cell immune response. Interestingly, recent studies have suggested that the activation or depression of the AHR signaling pathway may play a role in the outcome of diverse human viral infections. In the present report, the effect of the pharmacological modulation of AHR on JUNV in vitro infection was analyzed. An initial microarray screening showed that the AHR pathway was overexpressed in JUNV-infected hepatic cells. Concomitantly, the infection of Vero and Huh-7 cells with the JUNV strains IV4454 and Candid#1 was significantly inhibited in a dose-dependent manner by treatment with CH223191, a specific AHR antagonist, as detected by infectivity assays, real-time RT-PCR and immunofluorescence detection of viral proteins. Furthermore, the pro-viral role of AHR in JUNV infection appears to be independent of the IFN-I pathway. Our findings support the promising perspectives of the pharmacological modulation of AHR as a potential target for the control of AHF. [ABSTRACT FROM AUTHOR]
- Published
- 2023
- Full Text
- View/download PDF
18. VelcroVax: a 'Bolt-On' Vaccine Platform for Glycoprotein Display
- Author
-
Natalie J. Kingston, Keith Grehan, Joseph S. Snowden, Mark Hassall, Jehad Alzahrani, Guido C. Paesen, Lee Sherry, Connor Hayward, Amy Roe, Sam Stephen, Darren Tomlinson, Antra Zeltina, Katie J. Doores, Neil A. Ranson, Martin Stacey, Mark Page, Nicola J. Rose, Thomas A. Bowden, David J. Rowlands, and Nicola J. Stonehouse
- Subjects
HBcAg ,VLP ,Junín virus ,platform ,vaccine ,Microbiology ,QR1-502 - Abstract
ABSTRACT Having varied approaches to the design and manufacture of vaccines is critical in being able to respond to worldwide needs and newly emerging pathogens. Virus-like particles (VLPs) form the basis of two of the most successful licensed vaccines (against hepatitis B virus [HBV] and human papillomavirus). They are produced by recombinant expression of viral structural proteins, which assemble into immunogenic nanoparticles. VLPs can be modified to present unrelated antigens, and here we describe a universal “bolt-on” platform (termed VelcroVax) where the capturing VLP and the target antigen are produced separately. We utilize a modified HBV core (HBcAg) VLP with surface expression of a high-affinity binding sequence (Affimer) directed against a SUMO tag and use this to capture SUMO-tagged gp1 glycoprotein from the arenavirus Junín virus (JUNV). Using this model system, we have solved the first high-resolution structures of VelcroVax VLPs and shown that the VelcroVax-JUNV gp1 complex induces superior humoral immune responses compared to the noncomplexed viral protein. We propose that this system could be modified to present a range of antigens and therefore form the foundation of future rapid-response vaccination strategies. IMPORTANCE The hepatitis B core protein (HBc) forms noninfectious virus-like particles, which can be modified to present a capturing molecule, allowing suitably tagged antigens to be bound on their surface. This system can be adapted and provides the foundation for a universal “bolt-on” vaccine platform (termed VelcroVax) that can be easily and rapidly modified to generate nanoparticle vaccine candidates.
- Published
- 2023
- Full Text
- View/download PDF
19. The Pan-ErbB tyrosine kinase inhibitor afatinib inhibits multiple steps of the mammarenavirus life cycle.
- Author
-
Mizuma, Keita, Takashima, Ayako, Cubitt, Beatrice, de la Torre, Juan C., and Iwasaki, Masaharu
- Subjects
- *
PROTEIN-tyrosine kinase inhibitors , *LIFE cycles (Biology) , *AFATINIB , *LYMPHOCYTIC choriomeningitis virus , *LASSA fever , *PROTEIN-tyrosine kinases - Abstract
The mammarenavirus Lassa virus (LASV) causes a life-threatening acute febrile disease, Lassa fever (LF). To date, no US Food and Drug Administration (FDA)-licensed medical countermeasures against LASV are available. This underscores the need for the development of novel anti-LASV drugs. Here, we screen an FDA-approved drug library to identify novel anti-LASV drug candidates using an infectious-free cell line expressing a functional LASV ribonucleoprotein (vRNP), where levels of vRNP-directed reporter gene expression serve as a surrogate for vRNP activity. Our screen identified the pan-ErbB tyrosine kinase inhibitor afatinib as a potent inhibitor of LASV vRNP activity. Afatinib inhibited multiplication of lymphocytic choriomeningitis virus (LCMV) a mammarenavirus closely related to LASV. Cell-based assays revealed that afatinib inhibited multiple steps of the LASV and LCMV life cycles. Afatinib also inhibited multiplication of Junín virus vaccine strain Candid#1, indicating that afatinib can have antiviral activity against a broad range of human pathogenic mammarenaviruses. • LASV RNP-expressing cell-based screening against a US FDA-approved drug library. • Pan-ErbB tyrosine kinase inhibitor afatinib is a potent inhibitor of LASV RNP. • Afatinib inhibits multiplication of LCMV and Junín virus (JUNV). • Afatinib inhibits multiple steps of the life cycles of LASV, LCMV, and JUNV. [ABSTRACT FROM AUTHOR]
- Published
- 2022
- Full Text
- View/download PDF
20. Nuclease Activity of the Junín Virus Nucleoprotein C-Terminal Domain
- Author
-
Alicia Armella Sierra, María Eugenia Loureiro, Sebastián Esperante, Silvia Susana Borkosky, Giovanna L. Gallo, Gonzalo de Prat Gay, and Nora Lopez
- Subjects
Junín virus ,nucleoprotein ,exoribonuclease activity ,biophysical characterization ,Microbiology ,QR1-502 - Abstract
The mammarenavirus Junín (JUNV) is the causative agent of Argentine hemorrhagic fever, a severe disease of public health concern. The most abundant viral protein is the nucleoprotein (NP), a multifunctional, two-domain protein with the primary role as structural component of the viral nucleocapsids, used as template for viral polymerase RNA synthesis activities. Here, we report that the C-terminal domain (CTD) of the attenuated Candid#1 strain of the JUNV NP can be purified as a stable soluble form with a secondary structure in line with known NP structures from other mammarenaviruses. We show that the JUNV NP CTD interacts with the viral matrix protein Z in vitro, and that the full-length NP and Z interact with each other in cellulo, suggesting that the NP CTD is responsible for this interaction. This domain comprises an arrangement of four acidic residues and a histidine residue conserved in the active site of exoribonucleases belonging to the DEDDh family. We show that the JUNV NP CTD displays metal-ion-dependent nuclease activity against DNA and single- and double-stranded RNA, and that this activity is impaired by the mutation of a catalytic residue within the DEDDh motif. These results further support this activity, not previously observed in the JUNV NP, which could impact the mechanism of the cellular immune response modulation of this important pathogen.
- Published
- 2023
- Full Text
- View/download PDF
21. Effects of the Natural Flavonoid Quercetin on Arenavirus Junín Infection
- Author
-
Aaron Ezequiel Alvarez De Lauro, Miguel Angel Pelaez, Agostina Belén Marquez, Mariel Selene Wagner, Luis Alberto Scolaro, Cybele Carina García, Elsa Beatriz Damonte, and Claudia Soledad Sepúlveda
- Subjects
arenavirus ,Junín virus ,Argentine hemorrhagic fever ,quercetin ,natural flavonoid ,pretreatment ,Microbiology ,QR1-502 - Abstract
There is no specific chemotherapy approved for the treatment of pathogenic arenaviruses that cause severe hemorrhagic fever (HF) in the population of endemic regions in America and Africa. The present study reports the effects of the natural flavonoid quercetin (QUER) on the infection of A549 and Vero cells with Junín virus (JUNV), agent of the Argentine HF. By infectivity assays, a very effective dose-dependent reduction of JUNV multiplication was shown by cell pretreatment at 2–6 h prior to the infection at non-cytotoxic concentrations, with 50% effective concentration values in the range of 6.1–7.5 µg/mL. QUER was also active by post-infection treatment but with minor efficacy. Mechanistic studies indicated that QUER mainly affected the early steps of virus adsorption and internalization in the multiplication cycle of JUNV. Treatment with QUER blocked the phosphorylation of Akt without changes in the total protein expression, detected by Western blot, and the consequent perturbation of the PI3K/Akt pathway was also associated with the fluorescence redistribution from membrane to cytoplasm of TfR1, the cell receptor recognized by JUNV. Then, it appears that the cellular antiviral state, induced by QUER treatment, leads to the prevention of JUNV entry into the cell.
- Published
- 2023
- Full Text
- View/download PDF
22. Evidence for Viral mRNA Export from Ebola Virus Inclusion Bodies by the Nuclear RNA Export Factor NXF1.
- Author
-
Wendt, Lisa, Brandt, Janine, Ushakov, Dmitry S., Bodmer, Bianca S., Pickin, Matthew J., Groseth, Allison, and Hoenen, Thomas
- Subjects
- *
EBOLA virus , *CELLULAR inclusions , *RNA , *LIFE cycles (Biology) , *RNA synthesis , *MESSENGER RNA - Abstract
Many negative-sense RNA viruses, including the highly pathogenic Ebola virus (EBOV), use cytoplasmic inclusion bodies (IBs) for viral RNA synthesis. However, it remains unclear how viral mRNAs are exported from these IBs for subsequent translation. We recently demonstrated that the nuclear RNA export factor 1 (NXF1) is involved in a late step in viral protein expression, i.e., downstream of viral mRNA transcription, and proposed it to be involved in this mRNA export process. We now provide further evidence for this function by showing that NXF1 is not required for translation of viral mRNAs, thus pinpointing its function to a step between mRNA transcription and translation. We further show that RNA binding of both NXF1 and EBOV NP is necessary for export of NXF1 from IBs, supporting a model in which NP hands viral mRNA over to NXF1 for export. Mapping of NP-NXF1 interactions allowed refinement of this model, revealing two separate interaction sites, one of them directly involving the RNA binding cleft of NP, even though these interactions are RNA-independent. Immuno-fluorescence analyses demonstrated that individual NXF1 domains are sufficient for its recruitment into IBs, and complementation assays helped to define NXF1 domains important for its function in the EBOV life cycle. Finally, we show that NXF1 is also required for protein expression of other viruses that replicate in cytoplasmic IBs, including Lloviu and Junín virus. These data suggest a role for NXF1 in viral mRNA export from IBs for various viruses, making it a potential target for broadly active antivirals. [ABSTRACT FROM AUTHOR]
- Published
- 2022
- Full Text
- View/download PDF
23. South American Hemorrhagic Fevers: A summary for clinicians
- Author
-
Maria G. Frank, Adam Beitscher, Camille M. Webb, Vanessa Raabe, Nahid Bhadelia, Theodore J. Cieslak, Richard T. Davey, Kerry Dierberg, Jared D. Evans, Jonathan Grein, Mark G. Kortepeter, Colleen S. Kraft, Chris J. Kratochvil, Karen Martins, Susan McLellan, Aneesh K. Mehta, George Risi, Lauren Sauer, Erica S. Shenoy, and Tim Uyeki
- Subjects
South American hemorrhagic fevers ,New World Arenavirus ,Junín virus ,Argentine hemorrhagic fever ,Venezuelan hemorrhagic fever ,Machupo virus ,Infectious and parasitic diseases ,RC109-216 - Abstract
Objectives: This article is one of a series on acute, severe diseases of humans caused by emerging viruses for which there are no or limited licensed medical countermeasures. We approached this summary on South American Hemorrhagic Fevers (SAHF) from a clinical perspective that focuses on pathogenesis, clinical features, and diagnostics with an emphasis on therapies and vaccines that have demonstrated potential for use in an emergency situation through their evaluation in nonhuman primates (NHPs) and/or in humans. Methods: A standardized literature review was conducted on the clinical, pathological, vaccine, and treatment factors for SAHF as a group and for each individual virus/disease. Results: We identified 2 treatments and 1 vaccine platform that have demonstrated potential benefit for treating or preventing infection in humans and 4 other potential treatments currently under investigation. Conclusion: We provide succinct summaries of these countermeasures to give the busy clinician a head start in reviewing the literature if faced with a patient with South American Hemorrhagic Fever. We also provide links to other authoritative sources of information.
- Published
- 2021
- Full Text
- View/download PDF
24. The Virus–Host Interplay in Junín Mammarenavirus Infection.
- Author
-
Gallo, Giovanna Lucrecia, López, Nora, and Loureiro, María Eugenia
- Subjects
- *
ARENAVIRUSES , *HEMORRHAGIC fever , *ENDEMIC diseases , *REVERSE genetics , *VIRUS diseases , *INFECTION - Abstract
Junín virus (JUNV) belongs to the Arenaviridae family and is the causative agent of Argentine hemorrhagic fever (AHF), a severe human disease endemic to agricultural areas in Argentina. At this moment, there are no effective antiviral therapeutics to battle pathogenic arenaviruses. Cumulative reports from recent years have widely provided information on cellular factors playing key roles during JUNV infection. In this review, we summarize research on host molecular determinants that intervene in the different stages of the viral life cycle: viral entry, replication, assembly and budding. Alongside, we describe JUNV tight interplay with the innate immune system. We also review the development of different reverse genetics systems and their use as tools to study JUNV biology and its close teamwork with the host. Elucidating relevant interactions of the virus with the host cell machinery is highly necessary to better understand the mechanistic basis beyond virus multiplication, disease pathogenesis and viral subversion of the immune response. Altogether, this knowledge becomes essential for identifying potential targets for the rational design of novel antiviral treatments to combat JUNV as well as other pathogenic arenaviruses. [ABSTRACT FROM AUTHOR]
- Published
- 2022
- Full Text
- View/download PDF
25. Junin Virus Activates p38 MAPK and HSP27 Upon Entry.
- Author
-
Fitzpatrick, Collin J., Mudhasani, Rajini R., Altamura, Louis A., Campbell, Catherine E., Tran, Julie P., Beitzel, Brett F., Narayanan, Aarthi, de la Fuente, Cynthia L., Kehn-Hall, Kylene, Smith, Jeffrey M., Schmaljohn, Connie S., and Garrison, Aura R.
- Subjects
HEAT shock proteins ,NF-kappa B ,MITOGEN-activated protein kinases ,HUMAN cell culture ,PROTEIN microarrays - Abstract
Junín virus (JUNV), a New World arenavirus, is a rodent-borne virus and the causative agent of Argentine hemorrhagic fever. Humans become infected through exposure to rodent host secreta and excreta and the resulting infection can lead to an acute inflammatory disease with significant morbidity and mortality. Little is understood about the molecular pathogenesis of arenavirus hemorrhagic fever infections. We utilized Reverse Phase Protein Microarrays (RPPA) to compare global alterations in the host proteome following infection with an attenuated vaccine strain, Candid#1 (CD1), and the most parental virulent strain, XJ13, of JUNV in a human cell culture line. Human small airway epithelial cells were infected with CD1 or XJ13 at an MOI of 10, or mock infected. To determine proteomic changes at early timepoints (T = 1, 3, 8 and 24 h), the JUNV infected or mock infected cells were lysed in compatible buffers for RPPA. Out of 113 proteins that were examined by RPPA, 14 proteins were significantly altered following JUNV infection. Several proteins were commonly phosphorylated between the two strains and these correspond to entry and early replication events, to include p38 mitogen-activated protein kinase (MAPK), heat shock protein 27 (HSP27), and nuclear factor kappa B (NFκB). We qualitatively confirmed the alterations of these three proteins following infection by western blot analysis. We also determined that the inhibition of either p38 MAPK, with the small molecule inhibitor SB 203580 or siRNA knockdown, or HSP27, by siRNA knockdown, significantly decreases JUNV replication. Our data suggests that HSP27 phosphorylation at S82 upon virus infection is dependent on p38 MAPK activity. This work sheds light on the nuances of arenavirus replication. [ABSTRACT FROM AUTHOR]
- Published
- 2022
- Full Text
- View/download PDF
26. Junin Virus Activates p38 MAPK and HSP27 Upon Entry
- Author
-
Collin J. Fitzpatrick, Rajini R. Mudhasani, Louis A. Altamura, Catherine E. Campbell, Julie P. Tran, Brett F. Beitzel, Aarthi Narayanan, Cynthia L. de la Fuente, Kylene Kehn-Hall, Jeffrey M. Smith, Connie S. Schmaljohn, and Aura R. Garrison
- Subjects
Junin virus ,cellular pathways ,HSP27 ,p38 MAPK ,antiviral ,Microbiology ,QR1-502 - Abstract
Junín virus (JUNV), a New World arenavirus, is a rodent-borne virus and the causative agent of Argentine hemorrhagic fever. Humans become infected through exposure to rodent host secreta and excreta and the resulting infection can lead to an acute inflammatory disease with significant morbidity and mortality. Little is understood about the molecular pathogenesis of arenavirus hemorrhagic fever infections. We utilized Reverse Phase Protein Microarrays (RPPA) to compare global alterations in the host proteome following infection with an attenuated vaccine strain, Candid#1 (CD1), and the most parental virulent strain, XJ13, of JUNV in a human cell culture line. Human small airway epithelial cells were infected with CD1 or XJ13 at an MOI of 10, or mock infected. To determine proteomic changes at early timepoints (T = 1, 3, 8 and 24 h), the JUNV infected or mock infected cells were lysed in compatible buffers for RPPA. Out of 113 proteins that were examined by RPPA, 14 proteins were significantly altered following JUNV infection. Several proteins were commonly phosphorylated between the two strains and these correspond to entry and early replication events, to include p38 mitogen-activated protein kinase (MAPK), heat shock protein 27 (HSP27), and nuclear factor kappa B (NFκB). We qualitatively confirmed the alterations of these three proteins following infection by western blot analysis. We also determined that the inhibition of either p38 MAPK, with the small molecule inhibitor SB 203580 or siRNA knockdown, or HSP27, by siRNA knockdown, significantly decreases JUNV replication. Our data suggests that HSP27 phosphorylation at S82 upon virus infection is dependent on p38 MAPK activity. This work sheds light on the nuances of arenavirus replication.
- Published
- 2022
- Full Text
- View/download PDF
27. Clinical Management of Argentine Hemorrhagic Fever using Ribavirin and Favipiravir, Belgium, 2020
- Author
-
Ioannis Veliziotis, Alain Roman, Delphine Martiny, Gerlind Schuldt, Marc Claus, Nicolas Dauby, Sigi Van den Wijngaert, Charlotte Martin, Rakan Nasreddine, Claudia Perandones, Romain Mahieu, Corien Swaan, Serge Van Praet, Deborah Konopnicki, Maria A. Morales, Denis Malvy, Etienne Stevens, Philippe Dechamps, Erika Vlieghe, Olivier Vandenberg, Stephan Günther, and Michèle Gérard
- Subjects
Junin virus ,New World arenaviruses ,viruses ,Argentine hemorrhagic fever ,travel ,ribavirin ,Medicine ,Infectious and parasitic diseases ,RC109-216 - Abstract
We report a case of Argentine hemorrhagic fever diagnosed in a woman in Belgium who traveled from a disease-endemic area. Patient management included supportive care and combination therapy with ribavirin and favipiravir. Of 137 potential contacts, including friends, relatives, and healthcare and laboratory workers, none showed development of clinical symptoms of this disease.
- Published
- 2020
- Full Text
- View/download PDF
28. Vaccine Candidates against Arenavirus Infections
- Author
-
Takeshi Saito, Rachel A. Reyna, Satoshi Taniguchi, Kirsten Littlefield, Slobodan Paessler, and Junki Maruyama
- Subjects
arenavirus ,vaccine ,Lassa virus ,Junin virus ,virus attenuation ,Medicine - Abstract
The viral family Arenaviridae contains several members that cause severe, and often lethal, diseases in humans. Several highly pathogenic arenaviruses are classified as Risk Group 4 agents and must be handled in the highest biological containment facility, biosafety level-4 (BSL-4). Vaccines and treatments are very limited for these pathogens. The development of vaccines is crucial for the establishment of countermeasures against highly pathogenic arenavirus infections. While several vaccine candidates have been investigated, there are currently no approved vaccines for arenavirus infection except for Candid#1, a live-attenuated Junin virus vaccine only licensed in Argentina. Current platforms under investigation for use include live-attenuated vaccines, recombinant virus-based vaccines, and recombinant proteins. We summarize here the recent updates of vaccine candidates against arenavirus infections.
- Published
- 2023
- Full Text
- View/download PDF
29. Modulation of the Aryl Hydrocarbon Receptor Signaling Pathway Impacts on Junín Virus Replication
- Author
-
Miguel Angel Pelaez, María Florencia Torti, Aaron Ezequiel Alvarez De Lauro, Agostina Belén Marquez, Federico Giovannoni, Elsa Beatriz Damonte, and Cybele Carina García
- Subjects
aryl hydrocarbon receptor ,Junín virus ,arenavirus ,Argentine hemorrhagic fever ,host therapeutic target ,Microbiology ,QR1-502 - Abstract
Junín virus (JUNV), a member of the family Arenaviridae, is the etiological agent of the Argentine hemorrhagic fever, an endemic disease in the rural region of Argentina lacking a specific chemotherapy. Aryl hydrocarbon receptor (AHR) is expressed in several mammalian tissues and has been indicated as a sensor of ligands from variable sources and a modulator of the cell immune response. Interestingly, recent studies have suggested that the activation or depression of the AHR signaling pathway may play a role in the outcome of diverse human viral infections. In the present report, the effect of the pharmacological modulation of AHR on JUNV in vitro infection was analyzed. An initial microarray screening showed that the AHR pathway was overexpressed in JUNV-infected hepatic cells. Concomitantly, the infection of Vero and Huh-7 cells with the JUNV strains IV4454 and Candid#1 was significantly inhibited in a dose-dependent manner by treatment with CH223191, a specific AHR antagonist, as detected by infectivity assays, real-time RT-PCR and immunofluorescence detection of viral proteins. Furthermore, the pro-viral role of AHR in JUNV infection appears to be independent of the IFN-I pathway. Our findings support the promising perspectives of the pharmacological modulation of AHR as a potential target for the control of AHF.
- Published
- 2023
- Full Text
- View/download PDF
30. Novel Arenavirus Entry Inhibitors Discovered by Using a Minigenome Rescue System for High-Throughput Drug Screening
- Author
-
Rathbun, Jessica Y, Droniou, Magali E, Damoiseaux, Robert, Haworth, Kevin G, Henley, Jill E, Exline, Colin M, Choe, Hyeryun, and Cannon, Paula M
- Subjects
Medical Microbiology ,Biomedical and Clinical Sciences ,Biological Sciences ,Rare Diseases ,Vaccine Related ,Emerging Infectious Diseases ,Infectious Diseases ,Vector-Borne Diseases ,Biotechnology ,Orphan Drug ,Prevention ,Biodefense ,2.2 Factors relating to the physical environment ,Aetiology ,Infection ,Good Health and Well Being ,Antiviral Agents ,Arenaviridae Infections ,Arenavirus ,Drug Evaluation ,Preclinical ,High-Throughput Screening Assays ,Humans ,Junin virus ,Reverse Genetics ,Virus Internalization ,Agricultural and Veterinary Sciences ,Medical and Health Sciences ,Virology ,Agricultural ,veterinary and food sciences ,Biological sciences ,Biomedical and clinical sciences - Abstract
UnlabelledCertain members of the Arenaviridae family are category A agents capable of causing severe hemorrhagic fevers in humans. Specific antiviral treatments do not exist, and the only commonly used drug, ribavirin, has limited efficacy and can cause severe side effects. The discovery and development of new antivirals are inhibited by the biohazardous nature of the viruses, making them a relatively poorly understood group of human pathogens. We therefore adapted a reverse-genetics minigenome (MG) rescue system based on Junin virus, the causative agent of Argentine hemorrhagic fever, for high-throughput screening (HTS). The MG rescue system recapitulates all stages of the virus life cycle and enables screening of small-molecule libraries under biosafety containment level 2 (BSL2) conditions. The HTS resulted in the identification of four candidate compounds with potent activity against a broad panel of arenaviruses, three of which were completely novel. The target for all 4 compounds was the stage of viral entry, which positions the compounds as potentially important leads for future development.ImportanceThe arenavirus family includes several members that are highly pathogenic, causing acute viral hemorrhagic fevers with high mortality rates. No specific effective treatments exist, and although a vaccine is available for Junin virus, the causative agent of Argentine hemorrhagic fever, it is licensed for use only in areas where Argentine hemorrhagic fever is endemic. For these reasons, it is important to identify specific compounds that could be developed as antivirals against these deadly viruses.
- Published
- 2015
31. Second-Generation Live-Attenuated Candid#1 Vaccine Virus Resists Reversion and Protects against Lethal Junín Virus Infection in Guinea Pigs.
- Author
-
Gowen, Brian B., Hickerson, Brady T., York, Joanne, Westover, Jonna B., Sefing, Eric J., Bailey, Kevin W., Wandersee, Luci, and Nunberg, Jack H.
- Subjects
- *
VIRUS diseases , *GUINEA pigs , *VIRAL vaccines , *RNA sequencing , *VACCINE effectiveness , *VIRAL envelope proteins , *NEUTRALIZATION tests - Abstract
Live-attenuated virus vaccines are highly effective in preventing viral disease but carry intrinsic risks of residual virulence and reversion to pathogenicity. The classically derived Candid#1 virus protects seasonal field workers in Argentina against zoonotic infection by Junín virus (JUNV) but is not approved in the United States, in part due to the potential for reversion at the attenuating locus, a phenylalanine-to-isoleucine substitution at position 427 in the GP2 subunit of the GPC envelope glycoprotein. Previously, we demonstrated facile reversion of recombinant Candid#1 (rCan) in cell culture and identified an epistatic interaction between the attenuating I427 and a secondary K33S mutation in the stable signal peptide (SSP) subunit of GPC that imposes an evolutionary barrier to reversion. The magnitude of this genetic barrier is manifest in our repeated failures to rescue the hypothetical revertant virus. In this study, we show that K33S rCan is safe and attenuated in guinea pigs and capable of eliciting potent virus-neutralizing antibodies. Immunized animals are fully protected against lethal challenge with virulent JUNV. In addition, we employed a more permissive model of infection in neonatal mice to investigate genetic reversion. RNA sequence analysis of the recovered virus identified revertant viruses in pups inoculated with the parental rCan virus and none in mice receiving K33S rCan (P,0.0001). Taken together, our findings support the further development of K33S rCan as a safe second-generation JUNV vaccine. IMPORTANCE Our most successful vaccines comprise weakened strains of virus that initiate a limited and benign infection in immunized persons. The live-attenuated Candid#1 strain of Junín virus (JUNV) was developed to protect field workers in Argentina from rodent-borne hemorrhagic fever but is not licensed in the United States, in part due to the likelihood of genetic reversion to virulence. A single-amino-acid change in the GPC envelope glycoprotein of the virus is responsible for attenuation, and a single nucleotide change may regenerate the pathogenic virus. Here, we take advantage of a unique genetic interaction between GPC subunits to design a mutant Candid#1 virus that establishes an evolutionary barrier to reversion. The mutant virus (K33S rCan) is fully attenuated and protects immunized guinea pigs against lethal JUNV infection. We find no instances of reversion in mice inoculated with K33S rCan. This work supports the further development of K33S rCan as a second-generation JUNV vaccine. [ABSTRACT FROM AUTHOR]
- Published
- 2021
- Full Text
- View/download PDF
32. Chapter 33 Diseases of the central nervous system caused by lymphocytic choriomeningitis virus and other arenaviruses
- Author
-
Wilson, Michael R and Peters, Clarence J
- Subjects
Medical Microbiology ,Biomedical and Clinical Sciences ,Emerging Infectious Diseases ,Rare Diseases ,Vaccine Related ,Prevention ,Infectious Diseases ,Immunization ,Biodefense ,Infection ,Arenaviridae Infections ,Central Nervous System Diseases ,Humans ,Lymphocytic choriomeningitis virus ,Argentine hemorrhagic fever ,Bolivian hemorrhagic fever ,Guanarito virus ,Junin virus ,Lassa fever ,Machupo virus ,Venezuelan hemorrhagic fever ,arenavirus ,lymphocytic choriomeningitis virus ,viral encephalitis ,viral hemorrhagic fever ,viral meningitis ,zoonosis ,Neurology & Neurosurgery ,Neurosciences - Abstract
This chapter highlights the neurologic sequelae of viruses from two major groups of arenaviruses, the Lassa-lymphocytic choriomeningitis serocomplex and the Tacaribe serocomplex. Fundamental features of these viruses are reviewed, including the rich history of their discovery and the large influence that the study of arenaviruses has had on the disciplines of virology and immunology more generally. Virus morphology, viral genome organization, individual viral protein functions, and small-animal models of disease are also discussed. The epidemiology, natural history, and laboratory evaluation of the arenaviruses that cause human illness are presented. In particular, the neurologic complications of lymphocytic choriomeningitis virus in immunocompetent, pregnant, and solid-organ transplant patients are highlighted. The neurologic sequelae of the arenaviruses that cause hemorrhagic fever (i.e., Lassa fever, Argentine hemorrhagic fever, Bolivian hemorrhagic fever, Venezuelan hemorrhagic fever) are also presented. Lastly, potential treatment and vaccine strategies of these diseases are reviewed. © 2014 Elsevier B.V.
- Published
- 2014
33. Diseases of the central nervous system caused by lymphocytic choriomeningitis virus and other arenaviruses.
- Author
-
Wilson, Michael R and Peters, Clarence J
- Subjects
Humans ,Lymphocytic choriomeningitis virus ,Arenaviridae Infections ,Central Nervous System Diseases ,Argentine hemorrhagic fever ,Bolivian hemorrhagic fever ,Guanarito virus ,Junin virus ,Lassa fever ,Machupo virus ,Venezuelan hemorrhagic fever ,arenavirus ,lymphocytic choriomeningitis virus ,viral encephalitis ,viral hemorrhagic fever ,viral meningitis ,zoonosis ,Neurology & Neurosurgery - Abstract
This chapter highlights the neurologic sequelae of viruses from two major groups of arenaviruses, the Lassa-lymphocytic choriomeningitis serocomplex and the Tacaribe serocomplex. Fundamental features of these viruses are reviewed, including the rich history of their discovery and the large influence that the study of arenaviruses has had on the disciplines of virology and immunology more generally. Virus morphology, viral genome organization, individual viral protein functions, and small-animal models of disease are also discussed. The epidemiology, natural history, and laboratory evaluation of the arenaviruses that cause human illness are presented. In particular, the neurologic complications of lymphocytic choriomeningitis virus in immunocompetent, pregnant, and solid-organ transplant patients are highlighted. The neurologic sequelae of the arenaviruses that cause hemorrhagic fever (i.e., Lassa fever, Argentine hemorrhagic fever, Bolivian hemorrhagic fever, Venezuelan hemorrhagic fever) are also presented. Lastly, potential treatment and vaccine strategies of these diseases are reviewed. © 2014 Elsevier B.V.
- Published
- 2014
34. South American Hemorrhagic Fevers: A summary for clinicians.
- Author
-
Frank, Maria G., Beitscher, Adam, Webb, Camille M., and Raabe, Vanessa
- Subjects
- *
HEMORRHAGIC fever , *MEDICAL personnel , *INFECTION , *INFLUENZA - Abstract
• South American Hemorrhagic Fevers are caused by 5 Mammarenaviruses. • Infection occurs from contact with infected persons, rodents, or their environment. • The nonspecific initial symptoms resemble influenza without respiratory symptoms. • Diagnosis of viral hemorrhagic fevers requires a skillfully obtained travel history. • Two treatments and one vaccine are potential SAHF medical countermeasures. This article is one of a series on acute, severe diseases of humans caused by emerging viruses for which there are no or limited licensed medical countermeasures. We approached this summary on South American Hemorrhagic Fevers (SAHF) from a clinical perspective that focuses on pathogenesis, clinical features, and diagnostics with an emphasis on therapies and vaccines that have demonstrated potential for use in an emergency situation through their evaluation in nonhuman primates (NHPs) and/or in humans. A standardized literature review was conducted on the clinical, pathological, vaccine, and treatment factors for SAHF as a group and for each individual virus/disease. We identified 2 treatments and 1 vaccine platform that have demonstrated potential benefit for treating or preventing infection in humans and 4 other potential treatments currently under investigation. We provide succinct summaries of these countermeasures to give the busy clinician a head start in reviewing the literature if faced with a patient with South American Hemorrhagic Fever. We also provide links to other authoritative sources of information. [ABSTRACT FROM AUTHOR]
- Published
- 2021
- Full Text
- View/download PDF
35. The Virus–Host Interplay in Junín Mammarenavirus Infection
- Author
-
Giovanna Lucrecia Gallo, Nora López, and María Eugenia Loureiro
- Subjects
Junín virus ,Argentine hemorrhagic fever ,arenavirus ,host–virus interactions ,entry ,replication ,Microbiology ,QR1-502 - Abstract
Junín virus (JUNV) belongs to the Arenaviridae family and is the causative agent of Argentine hemorrhagic fever (AHF), a severe human disease endemic to agricultural areas in Argentina. At this moment, there are no effective antiviral therapeutics to battle pathogenic arenaviruses. Cumulative reports from recent years have widely provided information on cellular factors playing key roles during JUNV infection. In this review, we summarize research on host molecular determinants that intervene in the different stages of the viral life cycle: viral entry, replication, assembly and budding. Alongside, we describe JUNV tight interplay with the innate immune system. We also review the development of different reverse genetics systems and their use as tools to study JUNV biology and its close teamwork with the host. Elucidating relevant interactions of the virus with the host cell machinery is highly necessary to better understand the mechanistic basis beyond virus multiplication, disease pathogenesis and viral subversion of the immune response. Altogether, this knowledge becomes essential for identifying potential targets for the rational design of novel antiviral treatments to combat JUNV as well as other pathogenic arenaviruses.
- Published
- 2022
- Full Text
- View/download PDF
36. Type I interferon underlies severe disease associated with Junín virus infection in mice
- Author
-
Brady T Hickerson, Eric J Sefing, Kevin W Bailey, Arnaud J Van Wettere, Manuel L Penichet, and Brian B Gowen
- Subjects
mammarenavirus ,arenavirus ,junin virus ,transferrin receptor ,interferon ,viral hemorrhagic fever ,Medicine ,Science ,Biology (General) ,QH301-705.5 - Abstract
Junín virus (JUNV) is one of five New World mammarenaviruses (NWMs) that causes fatal hemorrhagic disease in humans and is the etiological agent of Argentine hemorrhagic fever (AHF). The pathogenesis underlying AHF is poorly understood; however, a prolonged, elevated interferon-α (IFN-α) response is associated with a negative disease outcome. A feature of all NWMs that cause viral hemorrhagic fever is the use of human transferrin receptor 1 (hTfR1) for cellular entry. Here, we show that mice expressing hTfR1 develop a lethal disease course marked by an increase in serum IFN-α concentration when challenged with JUNV. Further, we provide evidence that the type I IFN response is central to the development of severe JUNV disease in hTfR1 mice. Our findings identify hTfR1-mediated entry and the type I IFN response as key factors in the pathogenesis of JUNV infection in mice.
- Published
- 2020
- Full Text
- View/download PDF
37. Immunization with GP1 but Not Core-like Particles Displaying Isolated Receptor-Binding Epitopes Elicits Virus-Neutralizing Antibodies against Junín Virus
- Author
-
Gleyder Roman-Sosa, Anne Leske, Xenia Ficht, Tung Huy Dau, Julia Holzerland, Thomas Hoenen, Martin Beer, Robert Kammerer, Reinhold Schirmbeck, Felix A. Rey, Sandra M. Cordo, and Allison Groseth
- Subjects
arenavirus ,Junín virus ,immune response ,neutralizing antibodies ,glycoprotein ,GP1 ,Medicine - Abstract
New World arenaviruses are rodent-transmitted viruses and include a number of pathogens that are responsible for causing severe human disease. This includes Junín virus (JUNV), which is the causative agent of Argentine hemorrhagic fever. The wild nature and mobility of the rodent reservoir host makes it difficult to control the disease, and currently passive immunization with high-titer neutralizing antibody-containing plasma from convalescent patients is the only specific therapy. However, dwindling supplies of naturally available convalescent plasma, and challenges in developing similar resources for other closely related viruses, have made the development of alternative antibody-based therapeutic approaches of critical importance. In this study, we sought to induce a neutralizing antibody response in rabbits against the receptor-binding subunit of the viral glycoprotein, GP1, and the specific peptide sequences in GP1 involved in cellular receptor contacts. While these specific receptor-interacting peptides did not efficiently induce the production of neutralizing antibodies when delivered as a particulate antigen (as part of hepatitis B virus core-like particles), we showed that recombinant JUNV GP1 purified from transfected mammalian cells induced virus-neutralizing antibodies at high titers in rabbits. Further, neutralization was observed across a range of unrelated JUNV strains, a feature that is critical for effectiveness in the field. These results underscore the potential of GP1 alone to induce a potent neutralizing antibody response and highlight the importance of epitope presentation. In addition, effective virus neutralization by rabbit antibodies supports the potential applicability of this species for the future development of immunotherapeutics (e.g., based on humanized monoclonal antibodies). Such information can be applied in the design of vaccines and immunogens for both prevention and specific therapies against this and likely also other closely related pathogenic New World arenaviruses.
- Published
- 2022
- Full Text
- View/download PDF
38. The Current State of Vaccine Development for Specific Prophylactics of Arenaviral Hemorrhagic Fevers
- Author
-
T. E. Sizikova, V. N. Lebedev, S. I. Syromyatnikova, and S. V. Borisevich
- Subjects
hemorrhagic fevers ,arenaviruses ,lassa virus ,machupo virus ,junin virus ,specific prophylactics ,live vaccines ,vector recombinant vaccines ,rna-replicons ,dna vaccines ,Infectious and parasitic diseases ,RC109-216 - Abstract
Presently, the Arenavirus genus (Arenaviridae family) includes 26 individual species of viruses. It is divided into two main groups – Old World arenaviruses and New World arenaviruses. The New World arenaviruses comprise four clades: А, B, C, D; pathogenic for humans New World arenaviruses are attributed to clade B. Lassa, Lujo, Machupo, Junin, Guanarito and Sabia viruses are the agents of extremely hazardous hemorrhagic fevers (Lassa hemorrhagic fever, Lujo hemorrhagic fever, Bolivian hemorrhagic fever, Argentinean hemorrhagic fever, Venezuelan hemorrhagic fever, Brazilian hemorrhagic fever, accordingly). These arenaviruses pose a potential threat to national public health due to the possibility of their accidental importation into the territory of the Russian Federation. The vaccination of risk group is the most effective and money-saving means of protection against epidemic spread. Objective of this review is to analyze the specific preparations for arenaviral hemorrhagic fever prevention that are currently under development. Production of live vaccines based on attenuated strains of the agents, the DNA vaccines, vector recombinant vaccines and vaccines on the basis of RNA-replicons is viewed as the main trends in the area. Тhe most important results in the development of effective prophylactic means against arenaviral hemorrhagic fevers are discussed in this paper.
- Published
- 2018
- Full Text
- View/download PDF
39. Description and characterization of a novel live-attenuated tri-segmented Machupo virus in Guinea pigs
- Author
-
Amélie D. Zaza, Cécile H. Herbreteau, and Christophe N. Peyrefitte
- Subjects
Vaccine candidate development ,Therapeutic ,Mammarenaviruses ,Machupo virus ,Junín virus ,Candid #1 ,Infectious and parasitic diseases ,RC109-216 - Abstract
Abstract Background Machupo virus (MACV) is a member of the Mammarenavirus genus, Arenaviridae family and is the etiologic agent of Bolivian hemorrhagic fever, which causes small outbreaks or sporadic cases. Several other arenaviruses in South America Junín virus (JUNV) in Argentina, Guanarito in Venezuela, Sabiá in Brazil and Chapare in Bolivia, also are responsible for human hemorrhagic fevers. Among these arenaviruses, JUNV caused thousands of human cases until 1991, when the live attenuated Candid #1 vaccine, was used. Other than Candid #1 vaccine, few other therapeutic or prophylactic treatments exist. Therefore, new strategies for production of safe countermeasures with broad spectrum activity are needed. Findings We tested a tri-segmented MACV, a potential vaccine candidate with several mutations, (r3MACV). In cell culture, r3MACV showed a 2-log reduction in infectious virus particle production and the MACV inhibition of INF-1β was removed from the construct and produced by infected cells. Furthermore, in an animal experiment, r3MACV was able to protect 50% of guinea pigs from a simultaneous lethal JUNV challenge. Protected animals didn’t display clinical symptoms nor were virus particles found in peripheral blood (day 14) or in organs (day 28 post-inoculation). The r3MACV provided a higher protection than the Candid #1 vaccine. Conclusions The r3MACV provides a potential countermeasure against two South America arenaviruses responsible of human hemorrhagic fever.
- Published
- 2018
- Full Text
- View/download PDF
40. Clinical Management of Argentine Hemorrhagic Fever using Ribavirin and Favipiravir, Belgium, 2020.
- Author
-
Veliziotis, Ioannis, Roman, Alain, Martiny, Delphine, Schuldt, Gerlind, Claus, Marc, Dauby, Nicolas, Van den Wijngaert, Sigi, Martin, Charlotte, Nasreddine, Rakan, Perandones, Claudia, Mahieu, Romain, Swaan, Corien, Van Praet, Serge, Konopnicki, Deborah, Morales, Maria A., Malvy, Denis, Stevens, Etienne, Dechamps, Philippe, Vlieghe, Erika, and Vandenberg, Olivier
- Abstract
We report a case of Argentine hemorrhagic fever diagnosed in a woman in Belgium who traveled from a disease-endemic area. Patient management included supportive care and combination therapy with ribavirin and favipiravir. Of 137 potential contacts, including friends, relatives, and healthcare and laboratory workers, none showed development of clinical symptoms of this disease. [ABSTRACT FROM AUTHOR]
- Published
- 2020
- Full Text
- View/download PDF
41. The Glycoprotein of the Live-Attenuated Junin Virus Vaccine Strain Induces Endoplasmic Reticulum Stress and Forms Aggregates prior to Degradation in the Lysosome.
- Author
-
Manning, John T., Yun, Nadya E., Seregin, Alexey V., Takaaki Koma, Sattler, Rachel A., Ezeomah, Chiomah, Cheng Huang, de la Torre, Juan C., and Paessler, Slobodan
- Subjects
- *
VIRAL vaccines , *ENDOPLASMIC reticulum , *HEMORRHAGIC fever , *GLYCOSYLATION - Abstract
Argentine hemorrhagic fever is a potentially lethal disease that is caused by Junin virus (JUNV). There are currently around 5 million individuals at risk of infection within regions of endemicity in Argentina. The live attenuated vaccine strain Candid #1 (Can) is approved for use in regions of endemicity and has substantially decreased the number of annual Argentine hemorrhagic fever (AHF) cases. The glycoprotein (GPC) gene is primarily responsible for attenuation of the Can strain, and we have shown that the absence of an N-linked glycosylation motif in the subunit G1 of the glycoprotein complex of Can, which is otherwise present in the wildtype pathogenic JUNV, causes GPC retention in the endoplasmic reticulum (ER). Here, we show that Can GPC aggregates in the ER of infected cells, forming incorrect cross-chain disulfide bonds, which results in impaired GPC processing into G1 and G2. The GPC fails to cleave into its G1 and G2 subunits and is targeted for degradation within lysosomes. Cells infected with the wild-type Romero (Rom) strain do not produce aggregates that are observed in Can infection, and the stress on the ER remains minimal. While the mutation of the N-linked glycosylation motif (T168A) is primarily responsible for the formation of aggregates, other mutations within G1 that occurred earlier in the passage history of the Can strain also contribute to aggregation of the GPC within the ER. [ABSTRACT FROM AUTHOR]
- Published
- 2020
- Full Text
- View/download PDF
42. Guinea pig transferrin receptor 1 mediates cellular entry of Junín virus and other pathogenic New World arenaviruses.
- Author
-
Hickerson, Brady T., Westover, Jonna B., Zhongde Wang, Young-Min Lee, and Gowen, Brian B.
- Subjects
- *
TRANSFERRIN , *GUINEA pigs , *TRANSFERRIN receptors , *PATHOGENIC viruses , *MOUSE leukemia viruses , *ARENAVIRUSES - Abstract
Several clade B New World arenaviruses (NWAs) can cause severe and often fatal hemorrhagic fever, for which preventive and therapeutic measures are severely limited. These NWAs use human transferrin receptor 1 (hTfR1) as a host cell receptor for virus entry. The most prevalent of the pathogenic NWAs is Junín virus (JUNV), the etiological agent of Argentine hemorrhagic fever. Small animal models of JUNV infection are limited because most laboratory rodent species are refractory to disease. Only guinea pigs are known to develop disease following JUNV infection, but the underlying mechanisms are not well characterized. In the present study, we demonstrate marked susceptibility of Hartley guinea pigs to uniformly lethal disease when challenged with as few as 4 plaque-forming units of the Romero strain of JUNV. In vitro, we show that infection of primary guinea pig macrophages results in greater JUNV replication compared to infection of hamster or mouse macrophages. We provide evidence that the guinea pig TfR1 (gpTfR1) is the principal receptor for JUNV, while hamster and mouse orthologs fail to support viral entry/infection of pseudotyped murine leukemia viruses expressing pathogenic NWA glycoproteins or JUNV. Together, our results indicate that gpTfR1 serves as the primary receptor for pathogenic NWAs, enhancing viral infection in guinea pigs. [ABSTRACT FROM AUTHOR]
- Published
- 2020
- Full Text
- View/download PDF
43. Junin Virus Triggers Macrophage Activation and Modulates Polarization According to Viral Strain Pathogenicity
- Author
-
María F. Ferrer, Pablo Thomas, Aída O. López Ortiz, Andrea E. Errasti, Nancy Charo, Victor Romanowski, Juan Gorgojo, María E. Rodriguez, Eugenio A. Carrera Silva, and Ricardo M. Gómez
- Subjects
junin virus ,human macrophages ,TAM receptors ,macrophage activation ,macrophage polarization ,IFN-I ,Immunologic diseases. Allergy ,RC581-607 - Abstract
The New World arenavirus Junin (JUNV) is the etiological agent of Argentine hemorrhagic fever (AHF). Previous studies of human macrophage infection by the Old-World arenaviruses Mopeia and Lassa showed that while the non-pathogenic Mopeia virus replicates and activates human macrophages, the pathogenic Lassa virus replicates but fails to activate human macrophages. Less is known in regard to the impact of New World arenavirus infection on the human macrophage immune response. Macrophage activation is critical for controlling infections but could also be usurped favoring immune evasion. Therefore, it is crucial to understand how the JUNV infection modulates macrophage plasticity to clarify its role in AHF pathogenesis. With this aim in mind, we compared infection with the attenuated Candid 1 (C#1) or the pathogenic P strains of the JUNV virus in human macrophage cultures. The results showed that both JUNV strains similarly replicated and induced morphological changes as early as 1 day post-infection. However, both strains differentially induced the expression of CD71, the receptor for cell entry, the activation and maturation molecules CD80, CD86, and HLA-DR and selectively modulated cytokine production. Higher levels of TNF-α, IL-10, and IL-12 were detected with C#1 strain, while the P strain induced only higher levels of IL-6. We also found that C#1 strain infection skewed macrophage polarization to M1, whereas the P strain shifted the response to an M2 phenotype. Interestingly, the MERTK receptor, that negatively regulates the immune response, was down-regulated by C#1 strain and up-regulated by P strain infection. Similarly, the target genes of MERTK activation, the cytokine suppressors SOCS1 and SOCS3, were also increased after P strain infection, in addition to IRF-1, that regulates type I IFN levels, which were higher with C#1 compared with P strain infection. Together, this differential activation/polarization pattern of macrophages elicited by P strain suggests a more evasive immune response and may have important implications in the pathogenesis of AHF and underpinning the development of new potential therapeutic strategies.
- Published
- 2019
- Full Text
- View/download PDF
44. Editorial: Insights in virus and host: 2021.
- Author
-
Hiscott, John and Brandt, Curtis R.
- Subjects
CHIKUNGUNYA virus ,PORCINE epidemic diarrhea virus ,PLANT viruses ,MEDICAL microbiology ,ALTERNATIVE RNA splicing ,HOST-virus relationships - Published
- 2023
- Full Text
- View/download PDF
45. Inhibitors of the p38 cell signaling pathway as antiviral compounds against Junín virus
- Author
-
Jesús Emanuel Brunetti, Verónica Mara Quintana, Luis Alberto Scolaro, and Viviana Castilla
- Subjects
HEK293 Cells ,Junin virus ,Virology ,Chlorocebus aethiops ,Animals ,Humans ,General Medicine ,Virus Replication ,Antiviral Agents ,Vero Cells ,HeLa Cells ,Signal Transduction - Abstract
In the present study, we analyzed the modulation of p38 cell signaling by Junín virus (JUNV) and evaluated the antiviral activity of p38 inhibitors against JUNV. While JUNV induced a progressive activation of p38 throughout the infection in Vero cells, a partial downregulation of p38 phosphorylation was observed in HEK293 and HeLa cells. The compounds SB203580 and SB202190, which are selective inhibitors of p38, significantly reduced viral protein expression and viral yield in the cell lines examined, indicating that the p38 signaling pathway might be a promising antiviral target against JUNV infection.
- Published
- 2022
- Full Text
- View/download PDF
46. Family Arenaviridae
- Author
-
Susan Payne
- Subjects
chemistry.chemical_classification ,Arenavirus ,biology ,viruses ,Family Arenaviridae ,virus diseases ,RNA ,biology.organism_classification ,medicine.disease ,Genome ,Virology ,Ribosome ,chemistry ,Junin virus ,medicine ,Glycoprotein ,Lassa fever - Abstract
The name arenavirus comes from the Latin arenosus (sandy). Ribosomes are packaged within arenavirus virions and these are responsible for the “sandy” appearance of the particles. Arenavirus virions are enveloped with helical nucleocapsids and are pleomorphic, ranging in size from 100 to 130 nm in diameter. Structural proteins include a nucleocapsid protein (N), a matrix-like protein (Z), and two glycoproteins derived from a single precursor. Arenaviruses are segmented RNA viruses that replicate using an overall strategy common to negative-strand RNA viruses. However, both genome segments (large (L) and small (S)) are ambisense in their coding strategy. Rodents are natural hosts of many arenaviruses though some can infect humans through contact with infected rodents or inhalation of infectious urine or feces.
- Published
- 2023
- Full Text
- View/download PDF
47. Development of a Reverse Genetic System to Generate Recombinant Chimeric Tacaribe Virus that Expresses Junín Virus Glycoproteins
- Author
-
Sabrina Foscaldi, María Eugenia Loureiro, Claudia Sepúlveda, Carlos Palacios, María Belén Forlenza, and Nora López
- Subjects
Tacaribe virus ,infectious clone ,chimeric virus ,Junín virus ,non-coding region ,viral attenuation ,Medicine - Abstract
Mammarenaviruses are enveloped and segmented negative-stranded RNA viruses that comprise several pathogenic members associated with severe human hemorrhagic fevers. Tacaribe virus (TCRV) is the prototype for the New World group of mammarenaviruses and is not only naturally attenuated but also phylogenetically and antigenically related to all South American pathogenic mammarenaviruses, particularly the Junín virus (JUNV), which is the etiological agent of Argentinian hemorrhagic fever (AHF). Moreover, since TCRV protects guinea pigs and non-human primates from lethal challenges with pathogenic strains of JUNV, it has already been considered as a potential live-attenuated virus vaccine candidate against AHF. Here, we report the development of a reverse genetic system that relies on T7 polymerase-driven intracellular expression of the complementary copy (antigenome) of both viral S and L RNA segments. Using this approach, we successfully recovered recombinant TCRV (rTCRV) that displayed growth properties resembling those of authentic TCRV. We also generated a chimeric recombinant TCRV expressing the JUNV glycoproteins, which propagated similarly to wild-type rTCRV. Moreover, a controlled modification within the S RNA 5′ non-coding terminal sequence diminished rTCRV propagation in a cell-type dependent manner, giving rise to new perspectives where the incorporation of additional attenuation markers could contribute to develop safe rTCRV-based vaccines against pathogenic mammarenaviruses.
- Published
- 2020
- Full Text
- View/download PDF
48. Review of Mammarenavirus Biology and Replication
- Author
-
Steven J. Hallam, Takaaki Koma, Junki Maruyama, and Slobodan Paessler
- Subjects
arenavirus ,virus replication ,lassa virus ,junin virus ,viral entry ,virus immune evasion ,Microbiology ,QR1-502 - Abstract
The family Arenaviridae is divided into three genera: Mammarenavirus, Reptarenavirus, and Hartmanivirus. The Mammarenaviruses contain viruses responsible for causing human hemorrhagic fever diseases including New World viruses Junin, Machupo, Guanarito, Sabia, and Chapare virus and Old World viruses Lassa, and Lujo virus. These two groups of arenaviruses share the same genome organization composed of two ambisense RNA segments. These segments contain four open reading frames that encode for four proteins: the nucleoprotein, glycoprotein precursor, L protein, and Z. Despite their genome similarities, these groups exhibit marked differences in their replication life cycles. This includes differences in attachment, entry, and immune evasion. By understanding the intricacy of replication in each of these viral species we can work to develop counter measures against human diseases. This includes the development of vaccines and antivirals for these emerging viral threats. Currently only the vaccine against Junin virus, Candid#1, is in use as well as Ribavirin for treatment of Lassa Fever. In addition, small molecule inhibitors can be developed to target various aspects of the virus life cycle. In these ways an understanding of the arenavirus replication cycle can be used to alleviate the mortality and morbidity of these infections worldwide.
- Published
- 2018
- Full Text
- View/download PDF
49. Visualization of Double-Stranded RNA Colocalizing With Pattern Recognition Receptors in Arenavirus Infected Cells
- Author
-
Elizabeth J. Mateer, Slobodan Paessler, and Cheng Huang
- Subjects
dsRNA ,negative-sense RNA virus ,arenaviruses ,Junín virus ,pattern recognition receptor ,RIG-I ,Microbiology ,QR1-502 - Abstract
An important step in the initiation of the innate immune response to virus infection is the recognition of non-self, viral RNA, including double-stranded RNA (dsRNA), by cytoplasmic pattern recognition receptors (PRRs). For many positive-sense RNA viruses and DNA viruses, the production of viral dsRNA, and the interaction of viral dsRNA and PRRs are well characterized. However, for negative-sense RNA viruses, viral dsRNA was thought to be produced at low to undetectable levels and PRR recognition of viral dsRNA is still largely unclear. In the case of arenaviruses, the nucleocaspid protein (NP) has been identified to contain an exoribonuclease activity that preferentially degrades dsRNA in biochemical studies. Nevertheless, pathogenic New World (NW) arenavirus infections readily induce an interferon (IFN) response in a RIG-I dependent manner, and also activate the dsRNA-dependent Protein Kinase R (PKR). To better understand the innate immune response to pathogenic arenavirus infection, we used a newly identified dsRNA-specific antibody that efficiently detects viral dsRNA in negative-sense RNA virus infected cells. dsRNA was detected in NW arenavirus infected cells colocalizing with virus NP in immunofluorescence assay. Importantly, the dsRNA signals also colocalized with cytoplasmic PRRs, namely, PKR, RIG-I and MDA-5, as well as with the phosphorylated, activated form of PKR in infected cells. Our data clearly demonstrate the PRR recognition of dsRNA and their activation in NW arenavirus infected cells. These findings provide new insights into the interaction between NW arenaviruses and the host innate immune response.
- Published
- 2018
- Full Text
- View/download PDF
50. EXPERIENCE OF STUDY AND POSSIBLE WAYS OF ELIMINATION OF FALSE POSITIVE AND FALSE NEGATIVE RESULTS DURING EXECUTION OF POLYMERASE CHAIN REACTION ON AN EXAMPLE OF JUNIN VIRUS RNA DETECTION
- Author
-
T. E Sizikova, V. N Lebedev, V. B Pantyukhov, S. V Borisevich, and V. A Merkulov
- Subjects
polymerase chain reaction ,argentine hemorrhagic fever ,junin virus ,false positive results ,false negative results ,Microbiology ,QR1-502 - Abstract
Aim. Experience of study and possible ways of elimination of false positive and false negative results during execution of polymerase chain reaction on an example of Junin virus RNA detection. Materials and methods. Junin virus - causative agent ofArgentine hemorrhagic fever (AHF) strain XJpR37/5787 was obtained from the State collection of pathogenicity group I causative agents of the 48th Central Research Institute. Reagent kit for detection of Junin virus RNA by RT-PCR was developed in the Institute and consists of 4 sets: for isolation of RNA, execution of reverse-transcription reaction, execution of PCR and electrophoretic detection of PCR products. RT-PCR was carried out by a standard technique. Continuous cell cultures of African green monkey Yhro B, GMK-AH-1(D) were obtained from the museum of cell culture department of the Centre. Results. An experimental study of the effect of various factors of impact on the sample under investigation («thawing-freezing», presence of formaldehyde, heparin) on the obtaining of false negative results during Junin virus RNA detection by using RT-PCR was studied. Addition of 0.01% heparin to the samples was shown to completely inhibit PCR. Addition of 0.05% formaldehyde significantly reduces sensitivity of the method. A possibility of reduction of analysis timeframe from 15 to 5 days was shown during detection of the causative agent in samples with low concentration of the latter by growing the samples and subsequent analysis of the material obtained by using RT-PCR. Conclusion. During detection of causative agent by using RT-PCR false negative results could appear in the presence of formaldehyde and heparin in the sample. A possibility of elimination of false negative PCR results due to concentration of the causative agent in the sample under investigation at a level below sensitivity threshold was shown on the example of Junin virus RNA detection by using growing of the pathogen in appropriate accumulation system with subsequent analysis of the material obtained using PCR.
- Published
- 2015
Catalog
Discovery Service for Jio Institute Digital Library
For full access to our library's resources, please sign in.