3,627 results on '"Vesicular Stomatitis"'
Search Results
2. Intravenous Vesicular Stomatitis Virus in Patients With Peripheral T-cell Lymphoma
- Author
-
National Cancer Institute (NCI)
- Published
- 2024
3. Novel Endogenous Engineering Platform for Robust Loading and Delivery of Functional mRNA by Extracellular Vesicles.
- Author
-
Zickler, Antje M., Liang, Xiuming, Gupta, Dhanu, Mamand, Doste R., De Luca, Mariacristina, Corso, Giulia, Errichelli, Lorenzo, Hean, Justin, Sen, Titash, Elsharkasy, Omnia M., Kamei, Noriyasu, Niu, Zheyu, Zhou, Guannan, Zhou, Houze, Roudi, Samantha, Wiklander, Oscar P. B., Görgens, André, Nordin, Joel Z., Castilla‐Llorente, Virginia, and EL Andaloussi, Samir
- Subjects
- *
VESICULAR stomatitis , *EXTRACELLULAR vesicles , *NUCLEIC acids , *BIOENGINEERING , *IMMUNE response - Abstract
Messenger RNA (mRNA) has emerged as an attractive therapeutic molecule for a plethora of clinical applications. For in vivo functionality, mRNA therapeutics require encapsulation into effective, stable, and safe delivery systems to protect the cargo from degradation and reduce immunogenicity. Here, a bioengineering platform for efficient mRNA loading and functional delivery using bionormal nanoparticles, extracellular vesicles (EVs), is established by expressing a highly specific RNA‐binding domain fused to CD63 in EV producer cells stably expressing the target mRNA. The additional combination with a fusogenic endosomal escape moiety, Vesicular Stomatitis Virus Glycoprotein, enables functional mRNA delivery in vivo at doses substantially lower than currently used clinically with synthetic lipid‐based nanoparticles. Importantly, the application of EVs loaded with effective cancer immunotherapy proves highly effective in an aggressive melanoma mouse model. This technology addresses substantial drawbacks currently associated with EV‐based nucleic acid delivery systems and is a leap forward to clinical EV applications. [ABSTRACT FROM AUTHOR]
- Published
- 2024
- Full Text
- View/download PDF
4. Molecular characterization of Ebola virus, immune response, and therapeutic challenges: a narrative review.
- Author
-
Ndayambaje, Martin, Yadufashije, Callixte, Habyarimana, Thierry, Niyonsaba, Theogene, Wahnou, Hicham, Iradukunda, Patrick Gad, Izere, Cedrick, Uwishema, Olivier, Ndishimye, Pacifique, and Oudghiri, Mounia
- Subjects
- *
EBOLA virus , *VACCINE effectiveness , *VESICULAR stomatitis , *VIRAL proteins , *CYTOSKELETAL proteins - Abstract
The Ebola virus (EBOV) remains a major public health challenge due to its complex structure and the lack of appropriate and effective vaccines and therapies. This review characterizes the Ebola virus, its immune response, and its therapeutic challenges. Structural EBOV proteins include the envelope glycoprotein, nucleoprotein, RNA polymerase L, and viral proteins VP30, VP24, VP35, and VP40. The proteins play a role in the virus's pathogenesis by evading the host's immune response. The immune system evasion mechanisms of EBOV are critical in its pathogenesis. Some vaccines, such as the recombinant vesicular stomatitis virus-Zaire Ebola virus (RVSV-ZEBOV), have proven to be very effective and have been approved by the Food and Drug Administration (FDA) additionally, four other vaccines have been approved including Gam Evac-Combi (licensed in Russia), ad5-EBOV (approved in China), Zabdeno and Mvabea (approved in Europe). However, some challenges remain in developing effective vaccines, such as the selection of immunogens, cross-protecting immunity, long-term protection, mechanism of protection, and rapid response vaccination. Despite the progress made, there is still a need for an effective vaccine that offers durable and broad protection against multiple strains of the Ebola virus. This will be achieved through the collaboration of various organizations and government and Non-Governmental Organization (NGO) agencies. [ABSTRACT FROM AUTHOR]
- Published
- 2024
- Full Text
- View/download PDF
5. Characterisation and Sensitivity of a Canine Mast Cell Tumour Line to Oncolytic Viruses.
- Author
-
Mehrani, Yeganeh, Kakish, Julia E., Napoleoni, Christina, Thompson, Jennifer Jane, Knapp, Jason P., Minott, Jessica A., Yates, Jacob G. E., Stuart, Deirdre, Coomber, Brenda L., Foster, Robert A., Bridle, Byram W., and Karimi, Khalil
- Subjects
- *
ONCOLYTIC virotherapy , *MAST cells , *VESICULAR stomatitis , *CANCER relapse , *CELL lines - Abstract
ABSTRACT Canine mast cell tumours (MCTs) are one of the most common skin cancers of dogs. Surgical removal is the primary treatment, but recurrence and metastasis can occur even with low‐grade tumours. As a result, new treatment strategies are being sought. We tested the potential of several oncolytic viruses (OVs) to infect and kill a cell line isolated from a canine MCT. Employing a resazurin‐based metabolic assay and flow cytometry technology, we used recombinant vesicular stomatitis virus (rVSV‐Δm51), avian orthoavulavirus‐1 (AOaV‐1), and Orf viruses in our assessment. Our study aimed to evaluate the potential of oncolytic virotherapy in treating canine cancers. We found that MCT‐1 cells showed different sensitivities to the OVs, with rVSV‐Δm51 showing the most promising results in vitro. These findings suggest that further investigation into using OVs for treating canine MCTs is needed, although clinical efficacy is yet to be determined. [ABSTRACT FROM AUTHOR]
- Published
- 2024
- Full Text
- View/download PDF
6. Oncolytic viruses: a potential breakthrough immunotherapy for multiple myeloma patients.
- Author
-
Raimondi, Vincenzo, Vescovini, Rosanna, Dessena, Mattia, Donofrio, Gaetano, Storti, Paola, and Giuliani, Nicola
- Subjects
ONCOLYTIC virotherapy ,PLASMA cells ,VESICULAR stomatitis ,MULTIPLE myeloma ,MEASLES virus - Abstract
Oncolytic virotherapy represents an innovative and promising approach for the treatment of cancer, including multiple myeloma (MM), a currently incurable plasma cell (PC) neoplasm. Despite the advances that new therapies, particularly immunotherapy, have been made, relapses still occur in MM patients, highlighting the medical need for new treatment options. Oncolytic viruses (OVs) preferentially infect and destroy cancer cells, exerting a direct and/or indirect cytopathic effect, combined with a modulation of the tumor microenvironment leading to an activation of the immune system. Both naturally occurring and genetically modified viruses have demonstrated significant preclinical effects against MM cells. Currently, the OVs genetically modified measles virus strains, reovirus, and vesicular stomatitis virus are employed in clinical trials for MM. Nevertheless, significant challenges remain, including the efficiency of the virus delivery to the tumor, overcoming antiviral immune responses, and the specificity of the virus for MM cells. Different strategies are being explored to optimize OV therapy, including combining it with standard treatments and targeted therapies to enhance efficacy. This review will provide a comprehensive analysis of the mechanism of action of the different OVs, and preclinical and clinical evidence, focusing on the role of oncolytic virotherapy as a new possible immunotherapeutic approach also in combination with the current therapeutic armamentarium and underlying the future directions in the context of MM treatments. [ABSTRACT FROM AUTHOR]
- Published
- 2024
- Full Text
- View/download PDF
7. In memory of an exquisite medicinal chemist, Prof. Morris Robins.
- Author
-
De Clercq, Erik
- Subjects
- *
VACCINIA , *VESICULAR stomatitis , *VARICELLA-zoster virus , *ACYCLOVIR , *HERPES simplex virus , *CHEMISTS - Abstract
AbstractAmong the most prominent realizations of Morris J. Robins in the antiviral nucleoside chemistry are
(i) the synthesis of 8-substituted (methyl-, amino-, bromo-, iodo) derivatives of acyclovir,(ii) xylotubercidin as an inhibitor of herpes simplex virus (HSV) infections,(iii) the anti-HIV activity of the 2’,3’-dideoxyriboside of 2,6-diaminopurine (ddDAPR) and the 3’-azido- and 3’-fluoro derivatives thereof (AzddDAPR and FddDAPR, respectively),(iv) the potentiating effect of ribavirin on the anti-HIV activity of 2’,3’-dideoxyinosine (ddI) and ddDAPR,(v) S-adenosylhomocysteine hydrolase (SAH) inhibitors principally active against vaccinia virus (VV) and vesicular stomatitis virus (VSV), and(vi) furo[2,3-d]pyrimidinone derivatives active against varicella-zoster virus (VZV). [ABSTRACT FROM AUTHOR]- Published
- 2024
- Full Text
- View/download PDF
8. Cytoskeletal β-tubulin and cysteine cathepsin L deregulation by SARS-CoV-2 spike protein interaction with the neuronal model cell line SH-SY5Y.
- Author
-
Oliveira, Bernardo R., Nehlmeier, Inga, Kempf, Amy Madeleine, Venugopalan, Vaishnavi, Rehders, Maren, Ceniza, Marianne E.P., Cavalcanti, Pedro A.de T.P.V., Hoffmann, Markus, Pöhlmann, Stefan, and Brix, Klaudia
- Subjects
- *
SARS-CoV-2 , *SARS-CoV-2 Omicron variant , *VESICULAR stomatitis , *CENTRAL nervous system , *CELL membranes , *TUBULINS , *CYTOSKELETAL proteins - Abstract
SARS-CoV-2 mainly infects the respiratory tract but can also target other organs, including the central nervous system. While it was recently shown that cells of the blood-brain-barrier are permissive to SARS-CoV-2 infection in vitro , it remains debated whether neurons can be infected. In this study, we demonstrate that vesicular stomatitis virus particles pseudotyped with the spike protein of SARS-CoV-2 variants WT, Alpha, Delta and Omicron enter the neuronal model cell line SH-SY5Y. Cell biological analyses of the pseudo-virus treated cultures showed marked alterations in microtubules of SH-SY5Y cells. Because the changes in β-tubulin occurred in most cells, but only few were infected, we further asked whether interaction of the cells with spike protein might be sufficient to cause molecular and structural changes. For this, SH-SY5Y cells were incubated with trimeric spike proteins for time intervals of up to 24 h. CellProfiler™-based image analyses revealed changes in the intensities of microtubule staining in spike protein-incubated cells. Furthermore, expression of the spike protein-processing protease cathepsin L was found to be up-regulated by wild type, Alpha and Delta spike protein pseudotypes and cathepsin L was found to be secreted from spike protein-treated cells. We conclude that the mere interaction of the SARS-CoV-2 with neuronal cells can affect cellular architecture and proteolytic capacities. The molecular mechanisms underlying SARS-CoV-2 spike protein induced cytoskeletal changes in neuronal cells remain elusive and require future studies. [Display omitted] • ACE2, Basigin and Neuropilin-1 are detected at the plasma membrane of SH-SY5Y cells. • SARS-CoV-2 spike protein mediates entry of pseudotyped particles into SH-SY5Y cells. • SARS-CoV-2 spike variants rearrange the cytoskeleton of SH-SY5Y cells. • SARS-CoV-2 spike variants have differential effects on cathepsin L expression. [ABSTRACT FROM AUTHOR]
- Published
- 2024
- Full Text
- View/download PDF
9. Antiviral effects of Sarcodia suae water extracts against vesicular stomatitis virus infection.
- Author
-
Chang, Tsai-Fei, Lin, Chiou-Feng, Wu, Shan-Ying, Lin, Lin, Lai, Pei-Chun, Liao, Chen-Ting, Woodson, Caitlin, Chen, Shiow-Yi, Lee, Po-Tsang, Lee, Meng-Chou, and Lin, Shih-Chao
- Subjects
- *
VESICULAR stomatitis , *MEDICAL sciences , *VETERINARY medicine , *VIRUS diseases , *MARINE algae - Abstract
Marine algae, a rich source of bioactive substances, have long been utilized in biomedical and veterinary sciences. This study breaks new ground by assessing the antiviral therapeutic potential of water extracts from four algal species – Colaconema formosanum, Caulerpa microphysa, Gelidium amansii and Sarcodia suae – against vesicular stomatitis virus (VSV) infection. The standout among these was Sarcodia suae water extract (SSWE), which not only significantly repressed VSV replication and enhanced cell survival without cytotoxic effects, but also demonstrated its antiviral activity through interference with viral attachment, entry, RNA replication, and egress, and reduced VSV-induced autophagy in Mv1Lu cells during the late stages of infection. The EC50 of SSWE was 0.422 ± 0.14 mg ml‒1. The cytotoxicity assay confirmed the high biocompatibility of SSWE. This study identifies SSWE as a promising natural antiviral agent and elucidates its mechanisms of action, highlighting the need for further research to optimize its use in controlling VSV-related diseases. Highlights: The bioactive water extract of Sarcodia suae was identified as a sustainable antiviral agent for the algal cultivation industry. The antiviral activity of S. suae against vesicular stomatitis virus (VSV) infection was confirmed. The mechanisms of action of the extract from Sarcodia suae on the autophagic pathway were demonstrated. [ABSTRACT FROM AUTHOR]
- Published
- 2024
- Full Text
- View/download PDF
10. A single amino acid substitution in the Borna disease virus glycoprotein enhances the infectivity titer of vesicular stomatitis virus pseudotyped virus by altering membrane fusion activity.
- Author
-
Akiba, Yusa, Matsugo, Hiromichi, Kanda, Takehiro, Sakai, Modoka, Makino, Akiko, and Tomonaga, Keizo
- Subjects
BORNA disease virus ,MEMBRANE fusion ,VESICULAR stomatitis ,AMINO acids ,TITERS - Abstract
Borna disease virus 1 (BoDV‐1) causes acute fatal encephalitis in mammals, including humans. Despite its importance, research on BoDV‐1 cell entry has been hindered by low infectious viral particle production in cells and the lack of cytopathic effects, which are typically useful for screening. To address these issues, we developed a method to efficiently produce vesicular stomatitis virus (VSV) pseudotyped with glycoprotein (G) of members of the genus Orthobornavirus, including BoDV‐1. We discovered that optimal G expression is required to obtain a high infectivity titer of the VSV pseudotyped virus. Remarkably, the infectivity of the VSV pseudotyped virus with G from the BoDV‐1 strain huP2br was significantly higher than that of the VSV pseudotyped virus with G from the He/80 strain. Mutational analysis demonstrated that the methionine at BoDV‐1–G residue 307 increases the infectivity titer of VSV pseudotyped with BoDV‐1–G (VSV–BoDV‐1–G). A cell‒cell fusion assay indicated that this residue plays a pivotal role in membrane fusion, thus suggesting that high membrane fusion activity and a broad pH range for membrane fusion are crucial for achieving a high infectivity titer of VSV–BoDV‐1–G. This finding may be extended to increase the infectivity titer of VSV pseudotyped virus with other orthobornavirus G. Our study also contributes to identifying functional domains of BoDV‐1–G and provides insight into G‐mediated cell entry. [ABSTRACT FROM AUTHOR]
- Published
- 2024
- Full Text
- View/download PDF
11. Extensively harvested hay contaminated with common windgrass induced an ulcerative stomatitis–gingivitis outbreak in horses.
- Author
-
Vervuert, I., Müller, S., and Karl, H.
- Subjects
- *
BLOOD cell count , *PLANT identification , *SHOW horses , *VESICULAR stomatitis , *HORSE health , *HORSE breeding , *HAY , *ORAL mucosa - Abstract
Summary After starting feeding on a new batch of hay, 29 out of 33 horses showed clinical signs of oral ulcerative and bleeding papules and mucous membrane erythema of the gingiva of the mandible within 2–3 days. The results of the complete blood cell count and biochemistry analysis did not yield significant findings in relation to horses affected by ulcerative stomatitis. Swab analyses of the oral mucosa for vesicular stomatitis virus (VS, Type Indiana and Type New Jersey) were negative in all horses (n = 33). Common windgrass with long awns in the deeper stomal‐gingival ulcerations was confirmed through botanical analysis by botanical identification. The hay batch fed to the affected horses was assessed via botanical analysis, finding 15% common windgrass (Apera spica‐venti). After the removal of the contaminated hay batch, full recovery was seen in all horses within 2–6 weeks depending on the severity of the lesions. The grassland from which the hay was harvested belonged to a semi‐natural landscape restored to increase biodiversity and as a protection zone in case of overflow of the river Rhine. In conclusion, feeding forages harvested from these semi‐natural landscapes may be harmful to the health of horses. [ABSTRACT FROM AUTHOR]
- Published
- 2024
- Full Text
- View/download PDF
12. MAVS signaling shapes microglia responses to neurotropic virus infection.
- Author
-
Gern, Olivia Luise, Pavlou, Andreas, Mulenge, Felix, Busker, Lena Mareike, Ghita, Luca, Aringo, Angela, Costa, Bibiana, Spanier, Julia, Waltl, Inken, Stangel, Martin, and Kalinke, Ulrich
- Subjects
- *
MYELOID cells , *OLFACTORY bulb , *VESICULAR stomatitis , *VIRAL transmission , *VIRUS diseases , *VIRAL encephalitis - Abstract
Viral encephalitis is characterized by a series of immunological reactions that can control virus infection in the brain, but dysregulated responses may cause excessive inflammation and brain damage. Microglia are brain-resident myeloid cells that are specialized in surveilling the local CNS environment and in case of viral brain infection they contribute to the control of the infection and to restriction of viral dissemination. Here, we report that after exposure to neurotropic vesicular stomatitis virus (VSV), murine in vitro microglia cultures showed rapid upregulation of a broad range of pro-inflammatory and antiviral genes, which were stably expressed over the entire 8 h infection period. Additionally, a set of immunomodulatory genes was upregulated between 6 and 8 h post infection. In microglia cultures, the induction of several immune response pathways including cytokine responses was dependent on mitochondrial antiviral-signaling protein (MAVS). Consequently, in Mavs-deficient microglia the control of virus propagation failed as indicated by augmented virus titers and the accumulation of viral transcripts. Thus, in the analyzed in vitro system, MAVS signaling is critically required to achieve full microglia activation and to mediate profound antiviral effects. In Mavs-deficient mice, intranasal VSV instillation caused higher disease severity than in WT mice and virus dissemination was noticed beyond the olfactory bulb. Virus spread to inner regions of the olfactory bulb, i.e., the granular cell layer, correlated with the recruitment of highly inflammatory non-microglia myeloid cells into the olfactory bulb in Mavs−/− mice. Furthermore, increased cytokine levels were detected in the nasal cavity, the olfactory bulb and in other brain regions. Thus, microglial MAVS signaling is critically needed for virus sensing, full microglia activation, and for orchestration of protective immunity in the virus-infected CNS. [ABSTRACT FROM AUTHOR]
- Published
- 2024
- Full Text
- View/download PDF
13. Molecular Engineering of Virus Tropism.
- Author
-
He, Bo, Wilson, Belinda, Chen, Shih-Heng, Sharma, Kedar, Scappini, Erica, Cook, Molly, Petrovich, Robert, and Martin, Negin P.
- Subjects
- *
GENETIC vectors , *VIRAL tropism , *RECOMBINANT viruses , *CHIMERIC proteins , *VESICULAR stomatitis - Abstract
Engineered viral vectors designed to deliver genetic material to specific targets offer significant potential for disease treatment, safer vaccine development, and the creation of novel biochemical research tools. Viral tropism, the specificity of a virus for infecting a particular host, is often modified in recombinant viruses to achieve precise delivery, minimize off-target effects, enhance transduction efficiency, and improve safety. Key factors influencing tropism include surface protein interactions between the virus and host-cell, the availability of host-cell machinery for viral replication, and the host immune response. This review explores current strategies for modifying the tropism of recombinant viruses by altering their surface proteins. We provide an overview of recent advancements in targeting non-enveloped viruses (adenovirus and adeno-associated virus) and enveloped viruses (retro/lentivirus, Rabies, Vesicular Stomatitis Virus, and Herpesvirus) to specific cell types. Additionally, we discuss approaches, such as rational design, directed evolution, and in silico and machine learning-based methods, for generating novel AAV variants with the desired tropism and the use of chimeric envelope proteins for pseudotyping enveloped viruses. Finally, we highlight the applications of these advancements and discuss the challenges and future directions in engineering viral tropism. [ABSTRACT FROM AUTHOR]
- Published
- 2024
- Full Text
- View/download PDF
14. Leveraging Synthetic Virology for the Rapid Engineering of Vesicular Stomatitis Virus (VSV).
- Author
-
Moles, Chad M., Basu, Rupsa, Weijmarshausen, Peter, Ho, Brenda, Farhat, Manal, Flaat, Taylor, and Smith, Bruce F.
- Subjects
- *
MOLECULAR biology , *VESICULAR stomatitis , *VIROLOGY , *SYNTHETIC biology , *CONTRACTING out - Abstract
Vesicular stomatitis virus (VSV) is a prototype RNA virus that has been instrumental in advancing our understanding of viral molecular biology and has applications in vaccine development, cancer therapy, antiviral screening, and more. Current VSV genome plasmids for purchase or contract virus services provide limited options for modification, restricted to predefined cloning sites and insert locations. Improved methods and tools to engineer VSV will unlock further insights into long-standing virology questions and new opportunities for innovative therapies. Here, we report the design and construction of a full-length VSV genome. The 11,161 base pair synthetic VSV (synVSV) was assembled from four modularized DNA fragments. Following rescue and titration, phenotypic analysis showed no significant differences between natural and synthetic viruses. To demonstrate the utility of a synthetic virology platform, we then engineered VSV with a foreign glycoprotein, a common use case for studying viral entry and developing anti-virals. To show the freedom of design afforded by this platform, we then modified the genome of VSV by rearranging the gene order, switching the positions of VSV-P and VSV-M genes. This work represents a significant technical advance, providing a flexible, cost-efficient platform for the rapid construction of VSV genomes, facilitating the development of innovative therapies. [ABSTRACT FROM AUTHOR]
- Published
- 2024
- Full Text
- View/download PDF
15. Stearoyl coenzyme A desaturase 1 (SCD1) regulates foot-and-mouth disease virus replication by modulating host cell lipid metabolism and viral protein 2C-mediated replication complex formation.
- Author
-
Bonan Lv, Yuncong Yuan, Zhuang Yang, Xingran Wang, Jianjun Hu, Yidan Sun, Hang Du, Xuemei Liu, Huimin Duan, Ruyi Ding, Zishu Pan, Xiao-Feng Tang, and Chao Shen
- Subjects
- *
REOVIRUSES , *RNA virus infections , *VESICULAR stomatitis , *VIRUS diseases , *CELL metabolism - Abstract
The life cycle of foot-and-mouth disease virus (FMDV) is tightly regulated by host cell lipid metabolism. In previous studies, we reported downregulated expression of stearoyl coenzyme A desaturase-1 (SCD1), a key enzyme of fatty acid metabolism, in BHK-VEC cells (a virus-negative cell line derived from BKH-21 cells with persistent FMDV infection) on comparing transcriptomic data for BHK-VEC and BHK-21 cells (Y. Yuan et al., Front Cell Infect Microbiol 12:940906, 2022, https://doi.org/10.3389/fcimb.2022.940906; L. Han et al., Vet Microbiol 263:109247, 2021, https://doi.org/10.1016/j.vetmic.2021.109247). In the present study, we identify that SCD1 regulates FMDV replication. SCD1 overexpression or exogenous addition of oleic acid (OA), a product of the enzymatic activity of SCD1, increased FMDV replication in both BHK-21 cells and SCD1-knockdown cells. Overexpression of SCD1 or exogenous addition of OA restored FMDV infection and replication in BHK-VEC cells, and OA also promoted FMDV replication in BHK-21 cells with persistent FMDV infection. SCD1 recruited the nonstructural FMDV protein 2C to a detergent-resistant membrane located in the perinuclear region of cells to form replication complexes. Inhibiting SCD1 enzyme activity resulted in a significantly decreased number of FMDV replication complexes with abnormal morphology. Inhibition of SCD1 activity also effectively decreased the replication of other RNA viruses such as respiratory enteric orphan virus-3-176, poliovirus-1, enterovirus 71, and vesicular stomatitis virus. Our results demonstrate that SCD1, as a key host regulator of RNA virus replication, is a potential target for developing novel drugs against infections by RNA viruses. [ABSTRACT FROM AUTHOR]
- Published
- 2024
- Full Text
- View/download PDF
16. Matrix Protein of Vesicular Stomatitis Virus Targets the Mitochondria, Reprograms Glucose Metabolism, and Sensitizes to 2-Deoxyglucose in Glioblastoma.
- Author
-
Zhou, Yi, Li, Yongzhong, Chenm, Jing, Mei, Kai, Kang, Mingxiang, Chen, Ping, and Li, Qiu
- Subjects
- *
CELL metabolism , *METABOLIC reprogramming , *VESICULAR stomatitis , *EXTRACELLULAR matrix proteins , *GLUCOSE metabolism , *GLYCOLYSIS - Abstract
A potential therapeutic approach for cancer treatment is target oxidative phosphorylation and glycolysis simultaneously. The matrix protein of vesicular stomatitis virus (VSV MP) can target the surface of mitochondria, causing morphological changes that may be associated with mitochondrial dysfunction and oxidative phosphorylation inhibition. Previous research has shown that mitochondrial abnormalities can direct glucose metabolism toward glycolysis. Thus, after treatment with VSV MP, glycolysis inhibition is necessary to completely block glucose metabolism and eradicate cancer. Here, to inhibit glycolysis, the 2-deoxy-D-glucose (2-DG), a synthetic glucose analog was used to combine with VSV MP to treat cancer. This study aims to determine how VSV MP affects the glucose bioenergetic metabolism of cancer cells and to evaluate the synergistic effect of 2-DG when combined with VSV. Our results indicated that in U87 and C6 glioblastoma cell lines, VSV MP caused mitochondrial membrane potential loss, cytochrome c release, and glucose bioenergetics metabolism reprogramming. When combined with 2-DG, VSV MP synergistically aggravated cell viability, apoptosis, and G2/M phase arrest. Meanwhile, the combination therapy exacerbated ATP depletion, activated AMPK, and inhibited mammalian target of rapamycin signaling pathways. In addition, 2-DG treatment alone induced autophagy in glioblastoma cells; however, VSV MP inhibited the autophagy induced by 2-DG in combined treatment and finally contributed to the enhanced cytotoxic effect of the combination strategy in U87 and C6 cancer cells. In the orthotopic U87 glioblastoma model and subcutaneous C6 glioblastoma model, the combined treatment led to significant tumor regression and prolonged survival. A potent therapeutic approach for treating glioblastoma may be found in the combination of VSV MP and glycolytic inhibitors. [ABSTRACT FROM AUTHOR]
- Published
- 2024
- Full Text
- View/download PDF
17. A novel vesicular stomatitis virus armed with IL-2 mimic for oncolytic therapy.
- Author
-
Manman Wu, Yiwei Wang, Chuanjian Wu, Huang Huang, Xinyuan Zhou, Jun Wang, Sidong Xiong, and Chunsheng Dong
- Subjects
VESICULAR stomatitis ,IMMUNE checkpoint inhibitors ,IMMUNITY ,REGULATORY T cells ,IMMUNE response - Abstract
Oncolytic virus (OV) is increasingly being recognized as a novel vector in cancer immunotherapy. Increasing evidence suggests that OV has the ability to change the immune status of tumor microenvironment, so called transformation of 'cold' tumors into 'hot' tumors. The improved anti-tumor immunity can be induced by OV and further enhanced through the combination of various immunomodulators. The Neo-2/15 is a newly de novo synthesized cytokine that functions as both IL-2 and IL-15. However, it specifically lacks the binding site of IL-2 receptor α subunit (CD25), therefore unable to induce the Treg proliferation. In present study, a recombinant vesicular stomatitis virus expressing the Neo-2/15 (VSV
M51R -Neo-2/15) was generated. Intratumoral delivery of VSVM51R -Neo-2/15 efficiently inhibited tumor growth in mice without causing the IL-2-related toxicity previously observed in clinic. Moreover, treatment with VSVM51R -Neo-2/15 increased the number of activated CD8+ T cells but not Treg cells in tumors. More tumor-bearing mice were survival with VSVM51R -Neo-2/15 treatment, and the surviving mice displayed enhanced protection against tumor cell rechallenge due to the induced anti-tumor immunity. In addition, combination therapy of OV and anti-PD-L1 immune checkpoint inhibitors further enhanced the anti-tumor immune response. These findings suggest that our novel VSVM51R -Neo-2/15 can effectively inhibit the tumor growth and enhance the sensitivity to immune checkpoint inhibitors, providing promising attempts for further clinical trials. [ABSTRACT FROM AUTHOR]- Published
- 2024
- Full Text
- View/download PDF
18. Intranasal Prime–Boost with Spike Vectors Generates Antibody and T-Cell Responses at the Site of SARS-CoV-2 Infection.
- Author
-
Metko, Muriel, Tonne, Jason, Veliz Rios, Alexa, Thompson, Jill, Mudrick, Haley, Masopust, David, Diaz, Rosa Maria, Barry, Michael A., and Vile, Richard G.
- Subjects
COVID-19 vaccines ,VIRAL proteins ,GENETIC vectors ,VESICULAR stomatitis ,VIRUS diseases - Abstract
Background: Long-lived, re-activatable immunity to SARS-CoV-2 and its emerging variants will rely on T cells recognizing conserved regions of viral proteins across strains. Heterologous prime–boost regimens can elicit elevated levels of circulating CD8+ T cells that provide a reservoir of first responders upon viral infection. Although most vaccines are currently delivered intramuscularly (IM), the initial site of infection is the nasal cavity. Methods: Here, we tested the hypothesis that a heterologous prime and boost vaccine regimen delivered intranasally (IN) will generate improved immune responses locally at the site of virus infection compared to intramuscular vaccine/booster regimens. Results: In a transgenic human ACE2 murine model, both a Spike-expressing single-cycle adenovirus (SC-Ad) and an IFNß safety-enhanced replication-competent Vesicular Stomatitis Virus (VSV) platform generated anti-Spike antibody and T-cell responses that diminished with age. Although SC-Ad-Spike boosted a prime with VSV-Spike-mIFNß, SC-Ad-Spike alone induced maximal levels of IgG, IgA, and CD8+ T-cell responses. Conclusions: There were significant differences in T-cell responses in spleens compared to lungs, and the intranasal boost was significantly superior to the intramuscular boost in generating sentinel immune effectors at the site of the virus encounter in the lungs. These data show that serious consideration should be given to intranasal boosting with anti-SARS-CoV-2 vaccines. [ABSTRACT FROM AUTHOR]
- Published
- 2024
- Full Text
- View/download PDF
19. A comprehensive study of SARS-CoV-2 mfigain protease (Mpro) inhibitor-resistant mutants selected in a VSV-based system.
- Author
-
Costacurta, Francesco, Dodaro, Andrea, Bante, David, Schöppe, Helge, Peng, Ju-Yi, Sprenger, Bernhard, He, Xi, Moghadasi, Seyed Arad, Egger, Lisa Maria, Fleischmann, Jakob, Pavan, Matteo, Bassani, Davide, Menin, Silvia, Rauch, Stefanie, Krismer, Laura, Sauerwein, Anna, Heberle, Anne, Rabensteiner, Toni, Ho, Joses, and Harris, Reuben S.
- Subjects
- *
SARS-CoV-2 Omicron variant , *VESICULAR stomatitis , *SARS-CoV-2 , *PHENOTYPES , *BIOSAFETY - Abstract
Nirmatrelvir was the first protease inhibitor specifically developed against the SARS-CoV-2 main protease (3CLpro/Mpro) and licensed for clinical use. As SARS-CoV-2 continues to spread, variants resistant to nirmatrelvir and other currently available treatments are likely to arise. This study aimed to identify and characterize mutations that confer resistance to nirmatrelvir. To safely generate Mpro resistance mutations, we passaged a previously developed, chimeric vesicular stomatitis virus (VSV-Mpro) with increasing, yet suboptimal concentrations of nirmatrelvir. Using Wuhan-1 and Omicron Mpro variants, we selected a large set of mutants. Some mutations are frequently present in GISAID, suggesting their relevance in SARS-CoV-2. The resistance phenotype of a subset of mutations was characterized against clinically available protease inhibitors (nirmatrelvir and ensitrelvir) with cell-based, biochemical and SARS-CoV-2 replicon assays. Moreover, we showed the putative molecular mechanism of resistance based on in silico molecular modelling. These findings have implications on the development of future generation Mpro inhibitors, will help to understand SARS-CoV-2 protease inhibitor resistance mechanisms and show the relevance of specific mutations, thereby informing treatment decisions. Author summary: Studying dangerous viruses comes with risks and strict safety requirements. This is also true when new medications against viruses are developed and their effectiveness over time has to be tested. Unfortunately, viruses are quick to mutate and become resistant against almost any new medication. Ideally, this information is available before the medication is widely available. Then, when resistant viruses arise, patient caretakers can switch to other available treatments. However, to study the development of resistance with dangerous viruses is considered as 'gain-of-function' research, which is highly controversial and experiments have to be performed with high biological containment to prevent biosafety breaches. To facilitate studying the development of resistance and circumvent gain-of-function research of dangerous viruses, we describe a new, safe method. With this method, we can generate resistance data without using the actual virus, namely SARS-CoV-2 (alias 'Corona'). We provide resistance data against two clinically used antiviral medications that are relevant for the treatment of SARS-CoV-2. [ABSTRACT FROM AUTHOR]
- Published
- 2024
- Full Text
- View/download PDF
20. 水泡性口炎病毒对体外血管内皮屏障功能的损伤作用及其机制.
- Author
-
曹宇璇, 陈 为, 孙成彪, 赵 娜, 王 燕, 董明鑫, 许 娜, 刘文森, and 李咏梅
- Subjects
- *
LOW density lipoprotein receptors , *WNT proteins , *VESICULAR stomatitis , *ADHERENS junctions , *GENE expression - Abstract
Objective: To discuss the damage effect of vesicular stomatitis virus (VSV) on the vascular endothelial (VE) barrier, and to clarify its mechanism. Methods: The canine kidney cells were used to amplify VSV. The half tissue culture infective dose (TCID50) of VSV was determined using mouse brain endothelial tumor bEnd. 3 cells, and subsequent experiment was conducted using 300 times the TCID50. The bEnd. 3 cells were divided into infection 0 h group, infection 4 h group, infection 8 h group, and infection 12 h group for VE barrier damage experiments due to VSV infection. The bEnd. 3 cells were also divided into control group, infection group, and correction group for experiments to inhibit the VSV replication and restore the VE barrier. The bEnd. 3 cells were inoculated into Transwell chambers to construct an in vitro VE barrier model. Cell voltage resistance meter was used to detect the transepithelial resistance (TER) in various groups after the bEnd. 3 cells were infected with VSV at different time points; fluorescein isothiocyanate-dextran leakage assay was used to detect the permeability coefficients of the cells in various groups; immunofluorescence staining was used to observe the localization changes of VE-cadherin, β-catenin, and phosphorylated β-catenin (p-β-catenin) in cytoskeleton and adherens junctions (AJs) of the bEnd. 3 cells after VSV infection; real-time fluorescence quantitative PCR (RT-qPCR) method was used to detect the expression levels of Wnt and β-catenin mRNA in the cells in various groups; Western blotting method was used to detect the expression levels of Wnt, β -catenin, and p- β -catenin proteins in the cells in various groups. Results: The TCID50 of VSV was 10-4. 5 ·100 μL-1. The Transwell chamber experiment results showed that compared with infection 0 h group, the TERs in the cells in the other groups were significantly decreased (P<0. 05), and the permeability coefficients were significantly increased (P<0. 05). The immunofluorescence staining results showed that compared with control group, the cytoskeleton of the bEnd. 3 cells in infection group was disordered, the cell gaps was increased, the linear index of AJs was significantly decreased (P<0. 05), and β-catenin and p-β-catenin translocated from the cell membrane to the perinuclear area. The RT-qPCR results showed that compared with infection 0 h group, the expression levels of Wnt mRNA in the cells in the other groups were significantly decreased (P<0. 05), while the expression levels of β -catenin mRNA showed no statistically significant difference (P>0. 05). The Western blotting results showed that compared with infection 0 h group, the expression levels of Wnt protein in the cells in the other groups were significantly decreased (P<0. 05), the expression levels of β -catenin showed no statistically significant differences (P>0. 05), and the expression levels of p-β-catenin were significantly increased (P<0. 05). After inhibiting the VSV replication and correcting the low density lipoprotein receptor (LDLR) abnormalities, the Transwell chamber experiment results showed that compared with infection group, the TER in the cells in correction group was significantly increased (P<0. 05), and the permeability coefficient was significantly decreased (P<0. 05). The immunofluorescence staining results showed that compared with infection group, the gaps in the cells in correction group were reduced, and the perinuclear aggregation of β -catenin and p- β-catenin in the cells was restrained. The RT-qPCR results showed that compared with infection group, the expression level of Wnt mRNA in the cells in correction group was significantly increased (P<0. 05). The Western blotting results showed that compared with infection group, the expression level of Wnt protein in the cells in correction group was significantly increased (P<0. 05), the expression level of β -catenin showed no statistically significant difference (P>0. 05), and the expression level of p- β -catenin was significantly decreased (P<0. 05). Conclusion: VSV infection can cause the LDLR inactivation, reduce the expression level of Wnt protein, increase the phosphorylation level of β-catenin and cause its internalization, disrupt the stability of AJs, and ultimately lead to VE barrier damage. [ABSTRACT FROM AUTHOR]
- Published
- 2024
- Full Text
- View/download PDF
21. The Autonomous Fusion Activity of Human Cytomegalovirus Glycoprotein B Is Regulated by Its Carboxy-Terminal Domain.
- Author
-
Reuter, Nina, Kropff, Barbara, Chen, Xiaohan, Britt, William J., Sticht, Heinrich, Mach, Michael, and Thomas, Marco
- Subjects
- *
CELL fusion , *CHIMERIC proteins , *HUMAN cytomegalovirus , *VESICULAR stomatitis , *SITE-specific mutagenesis - Abstract
The human cytomegalovirus (HCMV) glycoprotein B (gB) is the viral fusogen required for entry into cells and for direct cell-to-cell spread of the virus. We have previously demonstrated that the exchange of the carboxy-terminal domain (CTD) of gB for the CTD of the structurally related fusion protein G of the vesicular stomatitis virus (VSV-G) resulted in an intrinsically fusion-active gB variant (gB/VSV-G). In this present study, we employed a dual split protein (DSP)-based cell fusion assay to further characterize the determinants of fusion activity in the CTD of gB. We generated a comprehensive library of gB CTD truncation mutants and identified two mutants, gB-787 and gB-807, which were fusion-competent and induced the formation of multinucleated cell syncytia in the absence of other HCMV proteins. Structural modeling coupled with site-directed mutagenesis revealed that gB fusion activity is primarily mediated by the CTD helix 2, and secondarily by the recruitment of cellular SH2/WW-domain-containing proteins. The fusion activity of gB-807 was inhibited by gB-specific monoclonal antibodies (MAbs) targeting the antigenic domains AD-1 to AD-5 within the ectodomain and not restricted to MAbs directed against AD-4 and AD-5 as observed for gB/VSV-G. This finding suggested a differential regulation of the fusion-active conformational state of both gB variants. Collectively, our findings underscore a pivotal role of the CTD in regulating the fusogenicity of HCMV gB, with important implications for understanding the conformations of gB that facilitate membrane fusion, including antigenic structures that could be targeted by antibodies to block this essential step in HCMV infection. [ABSTRACT FROM AUTHOR]
- Published
- 2024
- Full Text
- View/download PDF
22. Tuning VSV-G Expression Improves Baculovirus Integrity, Stability and Mammalian Cell Transduction Efficiency.
- Author
-
Mattioli, Martina, Raele, Renata A., Gautam, Gunjan, Borucu, Ufuk, Schaffitzel, Christiane, Aulicino, Francesco, and Berger, Imre
- Subjects
- *
ALFALFA looper , *GENETIC vectors , *VESICULAR stomatitis , *GENOME editing , *CRISPRS - Abstract
Baculoviral vectors (BVs) derived from Autographa californica multiple nucleopolyhedrovirus (AcMNPV) are an attractive tool for multigene delivery in mammalian cells, which is particularly relevant for CRISPR technologies. Most applications in mammalian cells rely on BVs that are pseudotyped with vesicular stomatitis virus G-protein (VSV-G) to promote efficient endosomal release. VSV-G expression typically occurs under the control of the hyperactive polH promoter. In this study, we demonstrate that polH-driven VSV-G expression results in BVs characterised by reduced stability, impaired morphology, and VSV-G induced toxicity at high multiplicities of transduction (MOTs) in target mammalian cells. To overcome these drawbacks, we explored five alternative viral promoters with the aim of optimising VSV-G levels displayed on the pseudotyped BVs. We report that Orf-13 and Orf-81 promoters reduce VSV-G expression to less than 5% of polH, rescuing BV morphology and stability. In a panel of human cell lines, we elucidate that BVs with reduced VSV-G support efficient gene delivery and CRISPR-mediated gene editing, at levels comparable to those obtained previously with polH VSV-G-pseudotyped BVs (polH VSV-G BV). These results demonstrate that VSV-G hyperexpression is not required for efficient transduction of mammalian cells. By contrast, reduced VSV-G expression confers similar transduction dynamics while substantially improving BV integrity, structure, and stability. [ABSTRACT FROM AUTHOR]
- Published
- 2024
- Full Text
- View/download PDF
23. Structural Heterogeneity of the Rabies Virus Virion.
- Author
-
Cai, Xiaoying, Zhou, Kang, Alvarez-Cabrera, Ana Lucia, Si, Zhu, Wang, Hui, He, Yao, Li, Cally, and Zhou, Z. Hong
- Subjects
- *
VESICULAR stomatitis , *RABIES virus , *EXTRACELLULAR matrix proteins , *VIRAL vaccines , *ELECTRON microscopy - Abstract
Rabies virus (RABV) is among the first recognized viruses of public health concern and has historically contributed to the development of viral vaccines. Despite these significances, the three-dimensional structure of the RABV virion remains unknown due to the challenges in isolating structurally homogenous virion samples in sufficient quantities needed for structural investigation. Here, by combining the capabilities of cryogenic electron tomography (cryoET) and microscopy (cryoEM), we determined the three-dimensional structure of the wild-type RABV virion. Tomograms of RABV virions reveal a high level of structural heterogeneity among the bullet-shaped virion particles encompassing the glycoprotein (G) trimer-decorated envelope and the nucleocapsid composed of RNA, nucleoprotein (N), and matrix protein (M). The structure of the trunk region of the virion was determined by cryoEM helical reconstruction, revealing a one-start N-RNA helix bound by a single layer of M proteins at an N:M ratio of 1. The N-M interaction differs from that in fellow rhabdovirus vesicular stomatitis virus (VSV), which features two layers of M stabilizing the N-RNA helix at an M:N ratio of 2. These differences in both M-N stoichiometry and binding allow RABV to flex its N-RNA helix more freely and point to different mechanisms of viral assembly between these two bullet-shaped rhabdoviruses. [ABSTRACT FROM AUTHOR]
- Published
- 2024
- Full Text
- View/download PDF
24. Vesicular Stomatitis Virus Detected in Biting Midges and Black Flies during the 2023 Outbreak in Southern California.
- Author
-
Scroggs, Stacey L. P., Swanson, Dustin A., Steele, Taylor D., Hudson, Amy R., Reister-Hendricks, Lindsey M., Gutierrez, Jessica, Shults, Phillip, McGregor, Bethany L., Taylor, Caitlin E., Davis, Travis M., Lamberski, Nadine, Phair, Kristen A., Howard, Lauren L., McConnell, Nathan E., Gurfield, Nikos, Drolet, Barbara S., Pelzel-McCluskey, Angela M., and Cohnstaedt, Lee W.
- Subjects
- *
BLOODSUCKING insects , *VESICULAR stomatitis , *SIMULIIDAE , *VIRUS diseases , *WATCHFUL waiting - Abstract
Vesicular stomatitis (VS) is a viral disease that affects horses, cattle, and swine that is transmitted by direct contact and hematophagous insects. In 2023, a multi-state outbreak of vesicular stomatitis New Jersey virus (VSNJV) occurred in California, Nevada, and Texas, infecting horses, cattle, and rhinoceros. To identify possible insect vectors, we conducted insect surveillance at various locations in San Diego County, CA, including at a wildlife park. CO2 baited traps set from mid-May to mid-August 2023 collected 2357 Culicoides biting midges and 1215 Simulium black flies, which are insect genera implicated in VSNJV transmission. Insects were pooled by species, location, and date, then tested for viral RNA. Nine RNA-positive pools of Culicoides spp. and sixteen RNA-positive pools of Simulium spp were detected. Infectious virus was detected by cytopathic effect in 96% of the RNA-positive pools. This is the first report of VSNJV in wild-caught C. bergi, C. freeborni, C. occidentalis, S. argus, S. hippovorum, and S. tescorum. The vector competency of these species for VSNJV has yet to be determined but warrants examination. Active vector surveillance and testing during disease outbreaks increases our understanding of the ecology and epidemiology of VS and informs vector control efforts. [ABSTRACT FROM AUTHOR]
- Published
- 2024
- Full Text
- View/download PDF
25. Viral and cellular determinants of polarized trafficking of viral envelope proteins from insect-specific and insect-vectored viruses in insect midgut and salivary gland cells.
- Author
-
Hodgson, Jeffrey J., Chen, Robin Y., Blissard, Gary W., and Buchon, Nicolas
- Subjects
- *
VIRAL envelope proteins , *LIFE cycles (Biology) , *SALIVARY glands , *INSECT viruses , *VESICULAR stomatitis - Abstract
Systemic viral infection of insects typically begins with the primary infection of midgut epithelial cells (enterocytes) and subsequent transit of the progeny virus in an apical-to-basal orientation into the hemocoel. For insect-vectored viruses, an oppositely oriented process (basal-to-apical transit) occurs upon secondary infection of salivary glands and is necessary for virus transmission to non-insect hosts. To examine this inversely oriented virus transit in these polarized tissues, we assessed the intracellular trafficking of two model viral envelope proteins (baculovirus GP64 and vesicular stomatitis virus G) in the midgut and salivary gland cells of the model insect, Drosophila melanogaster. Using fly lines that inducibly express either GP64 or VSV G, we found that each protein, expressed alone, was trafficked basally in midgut enterocytes. In salivary gland cells, VSV G was trafficked apically in most but not all cells, whereas GP64 was consistently trafficked basally. We demonstrated that a YxxØ motif present in both proteins was critical for basal trafficking in midgut enterocytes but dispensable for trafficking in salivary gland cells. Using RNAi, we found that clathrin adaptor protein complexes AP-1 and AP-3, as well as seven Rab GTPases, were involved in polarized VSV G trafficking in midgut enterocytes. Our results indicate that these viral envelope proteins encode the requisite information and require no other viral factors for appropriately polarized trafficking. In addition, they exploit tissue-specific differences in protein trafficking pathways to facilitate virus egress in the appropriate orientation for establishing systemic infections and vectoring infection to other hosts. IMPORTANCE Viruses that use insects as hosts must navigate specific routes through different insect tissues to complete their life cycles. The routes may differ substantially depending on the life cycle of the virus. Both insect pathogenic viruses and insect-vectored viruses must navigate through the polarized cells of the midgut epithelium to establish a systemic infection. In addition, insect-vectored viruses must also navigate through the polarized salivary gland epithelium for transmission. Thus, insect-vectored viruses appear to traffic in opposite directions in these two tissues. In this study, we asked whether two viral envelope proteins (VSV G and baculovirus GP64) alone encode the signals necessary for the polarized trafficking associated with their respective life cycles. Using Drosophila as a model to examine tissue-specific polarized trafficking of these viral envelope proteins, we identified one of the virus-encoded signals and several host proteins associated with regulating the polarized trafficking in the midgut epithelium. [ABSTRACT FROM AUTHOR]
- Published
- 2024
- Full Text
- View/download PDF
26. Optimizing lentiviral vector formulation conditions for efficient ex vivo transduction of primary human T cells in chimeric antigen receptor T-cell manufacturing.
- Author
-
Luostarinen, Annu, Kailaanmäki, Anssi, Turkki, Vesa, Köylijärvi, Marjut, Käyhty, Piia, Leinonen, Hanna, Albers-Skirdenko, Vita, Lipponen, Eevi, Ylä-Herttuala, Seppo, Kaartinen, Tanja, Lesch, Hanna P., and Kekarainen, Tuija
- Subjects
- *
CHIMERIC antigen receptors , *VESICULAR stomatitis , *REPORTER genes , *T cells , *CELLULAR therapy , *CRYOPROTECTIVE agents - Abstract
Chimeric antigen receptor (CAR) T-cell products are commonly generated using lentiviral vector (LV) transduction. Optimal final formulation buffer (FFB) supporting LV stability during cryostorage is crucial for cost-effective manufacturing. To identify the ideal LV FFB composition for ex vivo CAR-T production, primary human T cells were transduced with vesicular stomatitis virus G-protein (VSV-G) -pseudotyped LVs (encoding a reporter gene or an anti-CD19-CAR). The formulations included phosphate-buffered saline (PBS), HEPES, or X-VIVOTM 15, and stabilizing excipients. The functional and viral particle titers and vector copy number were measured after LV cryopreservation and up to 24 h post-thaw incubation. CAR-Ts were produced with LVs in selected FFBs, and the resulting cells were characterized. Post-cryopreservation, HEPES-based FFBs provided higher LV functional titers than PBS and X-VIVOTM 15, and 10% trehalose-20 mM MgCl 2 improved LV transduction efficiency in PBS and 50 mM HEPES. Thawed vectors remained stable at +4°C, while a ≤ 25% median decrease in the functional titer occurred during 24 h at room temperature. Tested excipients did not enhance LV post-thaw stability. CAR-Ts produced using LVs cryopreserved in 10% trehalose- or sucrose-20 mM MgCl 2 in 50 mM HEPES showed comparable transduction rates, cell yield, viability, phenotype, and in vitro functionality. A buffer consisting of 10% trehalose-20 mM MgCl 2 in 50 mM HEPES provided a feasible FFB to cryopreserve a VSV-G -pseudotyped LV for CAR-T-cell production. The LVs remained relatively stable for at least 24 h post-thaw, even at ambient temperatures. This study provides insights into process development, showing LV formulation data generated using the relevant target cell type for CAR-T-cell manufacturing. [ABSTRACT FROM AUTHOR]
- Published
- 2024
- Full Text
- View/download PDF
27. An Unconventional Case Study of Neoadjuvant Oncolytic Virotherapy for Recurrent Breast Cancer.
- Author
-
Forčić, Dubravko, Mršić, Karmen, Perić-Balja, Melita, Kurtović, Tihana, Ramić, Snježana, Silovski, Tajana, Pedišić, Ivo, Milas, Ivan, and Halassy, Beata
- Subjects
ONCOLYTIC virotherapy ,VESICULAR stomatitis ,MEASLES virus ,CANCER invasiveness ,MEASLES vaccines - Abstract
Intratumoural oncolytic virotherapy may have promise as a means to debulk and downstage inoperable tumours in preparation for successful surgery. Here, we describe the unique case of a 50-year-old self-experimenting female virologist with locally recurrent muscle-invasive breast cancer who was able to proceed to simple, non-invasive tumour resection after receiving multiple intratumoural injections of research-grade virus preparations, which first included an Edmonston-Zagreb measles vaccine strain (MeV) and then a vesicular stomatitis virus Indiana strain (VSV), both prepared in her own laboratory. The intratumoural virus therapy was well tolerated. Frequent imaging studies and regular clinical observations documenting size, consistency and mobility of the injected tumour demonstrate that both the MeV- and VSV-containing parts of the protocol contributed to the overall favourable response. Two months after the start of the virus injections, the shrunken tumour was no longer invading the skin or underlying muscle and was surgically excised. The excised tumour showed strong lymphocytic infiltration, with an increase in CD20-positive B cells, CD8-positive T cells and macrophages. PD-L1 expression was detected in contrast to the baseline PD-L1-negative phenotype. The patient completed one-year trastuzumab adjuvant therapy and remains well and recurrence-free 45 months post-surgery. Although an isolated case, it encourages consideration of oncolytic virotherapy as a neoadjuvant treatment modality. [ABSTRACT FROM AUTHOR]
- Published
- 2024
- Full Text
- View/download PDF
28. Study to Evaluate the Recombinant Vesicular Stomatitis Virus (rVSV)-Marburg Virus Vaccine Candidate (PHV01) in Healthy Adult Subjects (PHV01)
- Author
-
Biomedical Advanced Research and Development Authority
- Published
- 2024
29. Abortive and productive infection of CNS cell types following in vivo delivery of VSV.
- Author
-
Krause, Tyler B. and Cepko, Constance L.
- Subjects
- *
VIRAL genes , *VESICULAR stomatitis , *CENTRAL nervous system , *GENE expression ,CENTRAL nervous system infections - Abstract
Viral infection is frequently assayed by ongoing expression of viral genes. These assays fail to identify cells that have been exposed to the virus but limit or inhibit viral replication. To address this limitation, we used a dual-labeling vesicular stomatitis virus (DL-VSV), which has a deletion of the viral glycoprotein gene, to allow evaluation of primary infection outcomes. This virus encodes Cre, which can stably mark any cell with even a minimal level of viral gene expression. Additionally, the virus encodes GFP, which distinguishes cells with higher levels of viral gene expression, typically due to genome replication. Stereotactic injections of DL-VSV into the murine brain showed that different cell types had very different responses to the virus. Almost all neurons hosted high levels of viral gene expression, while glial cells varied in their responses. Astrocytes (Sox9+) were predominantly productively infected, while oligodendrocytes (Sox10+) were largely abortively infected. Microglial cells (Iba1+) were primarily uninfected. Furthermore, we monitored the early innate immune response to viral infection and identified unique patterns of interferon (IFN) induction. Shortly after infection, microglia were the main producers of IFNb, whereas later, oligodendrocytes were the main producers. IFNb+ cells were primarily abortively infected regardless of cell type. Last, we investigated whether IFN signaling had any impact on the outcome of primary infection and did not observe significant changes, suggesting that intrinsic factors are likely responsible for determining the outcome of primary infection. [ABSTRACT FROM AUTHOR]
- Published
- 2024
- Full Text
- View/download PDF
30. Phosphorylation of the selective autophagy receptor TAX1BP1 by TBK1 and IKBKE/IKKi promotes ATG8-family protein-dependent clearance of MAVS aggregates.
- Author
-
White, Jesse, Choi, Young Bong, Zhang, Jiawen, Vo, Mai Tram, He, Chaoxia, Shaikh, Kashif, and Harhaj, Edward W.
- Subjects
- *
RNA virus infections , *VESICULAR stomatitis , *VIRUS diseases , *LYSOSOMES , *AUTOPHAGY - Abstract
TAX1BP1 is a selective macroautophagy/autophagy receptor that inhibits NFKB and RIGI-like receptor (RLR) signaling to prevent excessive inflammation and maintain homeostasis. Selective autophagy receptors such as SQSTM1/p62 and OPTN are phosphorylated by the kinase TBK1 to stimulate their selective autophagy function. However, it is unknown if TAX1BP1 is regulated by TBK1 or other kinases under basal conditions or during RNA virus infection. Here, we found that TBK1 and IKBKE/IKKi function redundantly to phosphorylate TAX1BP1 and regulate its autophagic turnover through canonical macroautophagy. TAX1BP1 phosphorylation promotes its localization to lysosomes, resulting in its degradation. Additionally, we found that during vesicular stomatitis virus infection, TAX1BP1 is targeted to lysosomes in an ATG8-family protein-independent manner. Furthermore, TAX1BP1 plays a critical role in the clearance of MAVS aggregates, and phosphorylation of TAX1BP1 controls its MAVS aggrephagy function. Together, our data support a model whereby TBK1 and IKBKE license TAX1BP1-selective autophagy function to inhibit MAVS and RLR signaling.
Abbreviations: ATG: autophagy related; BafA1: bafilomycin A1; CALCOCO2: calcium binding and coiled-coil domain 2; GFP: green fluorescent protein; IFA: indirect immunofluorescence assay; IFN: interferon; IκB: inhibitor of nuclear factor kappa B; IKK: IκB kinase; IRF: interferon regulatory factor; KO: knockout; LAMP1: lysosomal associated membrane protein 1; LIR: LC3-interacting region; MAP1LC3/LC3: microtubule associated protein 1 light chain 3; MAVS: mitochondrial antiviral signaling protein; MEF: mouse embryonic fibroblast; MOI: multiplicity of infection; IKBKG/NEMO: inhibitor of nuclear factor kappa B kinase regulatory subunit gamma; NFKB: nuclear factor kappa B; OPTN: optineurin; Poly(I:C): polyinosinic-polycytidylic acid; RB1CC1/FIP200: RB1 inducible coiled-coil 1; RIGI: RNA sensor RIG-I; RLR: RIGI-like receptor; SDD-AGE: semi-denaturing detergent-agarose gel electrophoresis; SeV: Sendai virus; SLR: SQSTM1-like receptor; SQSTM1: sequestosome 1; TAX1BP1: Tax1 binding protein 1; TBK1: TANK binding kinase 1; TNF: tumor necrosis factor; TRAF: TNF receptor associated factor; VSV: vesicular stomatitis virus; ZnF: zinc finger. [ABSTRACT FROM AUTHOR]- Published
- 2024
- Full Text
- View/download PDF
31. Improved thermal stabilization of VSV-vector with enhanced vacuum drying in pullulan and trehalose-based films.
- Author
-
Iwashkiw, Jeremy A., Mohamud, Abdulhamid O., Kazhdan, Natallia, Ameen, Aaisha, Beecher, Jody E., Filipe, Carlos D. M., and Lichty, Brian D.
- Subjects
- *
VACCINE effectiveness , *VESICULAR stomatitis , *COMBINED vaccines , *VIRAL vaccines , *DISEASE vectors - Abstract
One major limitation of effective vaccine delivery is its dependency on a robust cold chain infrastructure. While Vesicular stomatitis virus (VSV) has been demonstrated to be an effective viral vaccine vector for diseases including Ebola, its −70 °C storage requirement is a significant limitation for accessing disadvantaged locations and populations. Previous work has shown thermal stabilization of viral vaccines with a combination of pullulan and trehalose (PT) dried films. To improve the thermal stability of VSV, we optimized PT formulation concentrations and components, as well as drying methodology with enhanced vacuum drying. When formulated in PT films, VSV can be stored for 32 weeks at 4 °C with less than 2 log PFU loss, at 25 °C with 2.5 log PFU loss, and at 37 °C with 3.1 log PFU loss. These results demonstrate a significant advancement in VSV thermal stabilization, decreasing the cold chain requirements for VSV vectored vaccines. [ABSTRACT FROM AUTHOR]
- Published
- 2024
- Full Text
- View/download PDF
32. Evaluation of a novel intramuscular prime/intranasal boost vaccination strategy against influenza in the pig model.
- Author
-
Avanthay, Robin, Garcia-Nicolas, Obdulio, Ruggli, Nicolas, Grau-Roma, Llorenç, Párraga-Ros, Ester, Summerfield, Artur, and Zimmer, Gert
- Subjects
- *
BOOSTER vaccines , *VACCINE effectiveness , *VIRUS diseases , *INFLUENZA A virus, H1N1 subtype , *VESICULAR stomatitis - Abstract
Live-attenuated influenza vaccines (LAIV) offer advantages over the commonly used inactivated split influenza vaccines. However, finding the optimal balance between sufficient attenuation and immunogenicity has remained a challenge. We recently developed an alternative LAIV based on the 2009 pandemic H1N1 virus with a truncated NS1 protein and lacking PA-X protein expression (NS1(1–126)-ΔPAX). This virus showed a blunted replication and elicited a strong innate immune response. In the present study, we evaluated the efficacy of this vaccine candidate in the porcine animal model as a pertinent in vivo system. Immunization of pigs via the nasal route with the novel NS1(1–126)-ΔPAX LAIV did not cause disease and elicited a strong mucosal immune response that completely blocked replication of the homologous challenge virus in the respiratory tract. However, we observed prolonged shedding of our vaccine candidate from the upper respiratory tract. To improve LAIV safety, we developed a novel prime/boost vaccination strategy combining primary intramuscular immunization with a haemagglutinin-encoding propagation-defective vesicular stomatitis virus (VSV) replicon, followed by a secondary immunization with the NS1(1–126)-ΔPAX LAIV via the nasal route. This two-step immunization procedure significantly reduced LAIV shedding, increased the production of specific serum IgG, neutralizing antibodies, and Th1 memory cells, and resulted in sterilizing immunity against homologous virus challenge. In conclusion, our novel intramuscular prime/intranasal boost regimen interferes with virus shedding and transmission, a feature that will help combat influenza epidemics and pandemics. Author summary: Nasally administered live-attenuated influenza vaccines induce a mucosal immune response that can effectively prevent primary infection and virus shedding. However, the development of alternative live vaccine strategies must consider the balance between eliciting a robust immune response and reducing the virulence of the vaccine candidate. To address this problem, we have developed a novel prime/boost vaccination strategy consisting of an initial intramuscular immunization with a propagation-defective RNA virus vector and subsequent nasal immunization with a modified attenuated influenza virus that has lost its ability to counteract the host innate immune response. Using the porcine model, we demonstrate that this novel strategy is capable of eliciting a robust immune response both systemically and at mucosal surfaces. Importantly, intranasal infection with homologous challenge virus remained undetectable in vaccinated animals. In conclusion, our innovative vaccination regimen represents a promising strategy to control influenza disease and virus spread in both humans and livestock. [ABSTRACT FROM AUTHOR]
- Published
- 2024
- Full Text
- View/download PDF
33. Modeling the 2014–2015 Vesicular Stomatitis Outbreak in the United States Using an SEIR-SEI Approach.
- Author
-
Humphreys, John M., Pelzel-McCluskey, Angela M., Shults, Phillip T., Velazquez-Salinas, Lauro, Bertram, Miranda R., McGregor, Bethany L., Cohnstaedt, Lee W., Swanson, Dustin A., Scroggs, Stacey L. P., Fautt, Chad, Mooney, Amber, Peters, Debra P. C., and Rodriguez, Luis L.
- Subjects
- *
VESICULAR stomatitis , *MARKOV chain Monte Carlo , *VECTOR-borne diseases , *INFECTIOUS disease transmission , *INSECT hosts - Abstract
Vesicular stomatitis (VS) is a vector-borne livestock disease caused by the vesicular stomatitis New Jersey virus (VSNJV). This study presents the first application of an SEIR-SEI compartmental model to analyze VSNJV transmission dynamics. Focusing on the 2014–2015 outbreak in the United States, the model integrates vertebrate hosts and insect vector demographics while accounting for heterogeneous competency within the populations and observation bias in documented disease cases. Key epidemiological parameters were estimated using Bayesian inference and Markov chain Monte Carlo (MCMC) methods, including the force of infection, effective reproduction number ( R t ), and incubation periods. The model revealed significant underreporting, with only 10–24% of infections documented, 23% of which presented with clinical symptoms. These findings underscore the importance of including competence and imperfect detection in disease models to depict outbreak dynamics and inform effective control strategies accurately. As a baseline model, this SEIR-SEI implementation is intended to serve as a foundation for future refinements and expansions to improve our understanding of VS dynamics. Enhanced surveillance and targeted interventions are recommended to manage future VS outbreaks. [ABSTRACT FROM AUTHOR]
- Published
- 2024
- Full Text
- View/download PDF
34. Efficacy and Immunogenicity of a Recombinant Vesicular Stomatitis Virus-Vectored Marburg Vaccine in Cynomolgus Macaques.
- Author
-
Camargos, Vidyleison N., Rossi, Shannan L., Juelich, Terry L., Smith, Jennifer K., Vasilakis, Nikos, Freiberg, Alexander N., Nichols, Rick, and Fusco, Joan
- Subjects
- *
VESICULAR stomatitis , *MARBURG virus , *HEMORRHAGIC fever , *VACCINE effectiveness , *DEATH rate - Abstract
Filoviruses, like the Marburg (MARV) and Ebola (EBOV) viruses, have caused outbreaks associated with significant hemorrhagic morbidity and high fatality rates. Vaccines offer one of the best countermeasures for fatal infection, but to date only the EBOV vaccine has received FDA licensure. Given the limited cross protection between the EBOV vaccine and Marburg hemorrhagic fever (MHF), we analyzed the protective efficacy of a similar vaccine, rVSV-MARV, in the lethal cynomolgus macaque model. NHPs vaccinated with a single dose (as little as 1.6 × 107 pfu) of rVSV-MARV seroconverted to MARV G-protein prior to challenge on day 42. Vaccinemia was measured in all vaccinated primates, self-resolved by day 14 post vaccination. Importantly, all vaccinated NHPs survived lethal MARV challenge, and showed no significant alterations in key markers of morbid disease, including clinical signs, and certain hematological and clinical chemistry parameters. Further, apart from one primate (from which tissues were not collected and no causal link was established), no pathology associated with Marburg disease was observed in vaccinated animals. Taken together, rVSV-MARV is a safe and efficacious vaccine against MHF in cynomolgus macaques. [ABSTRACT FROM AUTHOR]
- Published
- 2024
- Full Text
- View/download PDF
35. Design of novel broad‐spectrum antiviral nucleoside analogues using natural bases ring‐opening strategy.
- Author
-
Du, Xingyi, Yang, Xingxing, Zhao, Jianyuan, Zhang, Jinyan, Yu, Jiahui, Ma, Ling, Zhang, Weina, Cen, Shan, Ren, Xuhong, and He, Xinhua
- Subjects
- *
RNA virus infections , *VESICULAR stomatitis , *RNA polymerases , *INFLUENZA A virus , *BASE pairs - Abstract
The global prevalence of RNA virus infections has presented significant challenges to public health in recent years, necessitating the expansion of its alternative therapeutic library. Due to its evolutional conservation, RNA‐dependent RNA polymerase (RdRp) has emerged as a potential target for broad‐spectrum antiviral nucleoside analogues. However, after over half a century of structural modification, exploring unclaimed chemical space using frequently‐used structural substitution methods to design new nucleoside analogues is challenging. In this study, we explore the use of the "ring‐opening" strategy to design new base mimics, thereby using these base mimics to design new nucleoside analogues with broad‐spectrum antiviral activities. A total of 29 compounds were synthesized. Their activity against viral RdRp was initially screened using an influenza A virus RdRp high‐throughput screening model. Then, the antiviral activity of 38a was verified against influenza virus strain A/PR/8/34 (H1N1), demonstrating a 50% inhibitory concentration (IC50) value of 9.95 μM, which was superior to that of ribavirin (the positive control, IC50 = 11.43 μM). Moreover, 38a also has inhibitory activity against coronavirus 229E with an IC50 of 30.82 μM. In addition, compounds 42 and 46f exhibit an 82% inhibition rate against vesicular stomatitis virus at a concentration of 20 μM and hardly induce cytotoxicity in host cells. This work demonstrates the feasibility of designing nucleoside analogues with "ring‐opening" bases and suggests the "ring‐opening" nucleosides may have greater polarity, and designing prodrugs is an important aspect of optimizing their antiviral activity. Future research should focus on enhancing the conformational restriction of open‐loop bases to mimic Watson‐Crick base pairing better and improve antiviral activity. [ABSTRACT FROM AUTHOR]
- Published
- 2024
- Full Text
- View/download PDF
36. Vaccine Platform Comparison: Protective Efficacy against Lethal Marburg Virus Challenge in the Hamster Model.
- Author
-
O'Donnell, Kyle L., Henderson, Corey W., Anhalt, Hanna, Fusco, Joan, Erasmus, Jesse H., Lambe, Teresa, and Marzi, Andrea
- Subjects
- *
VESICULAR stomatitis , *MARBURG virus , *VACCINE approval , *HUMORAL immunity , *IMMUNE response - Abstract
Marburg virus (MARV), a filovirus, was first identified in 1967 in Marburg, Germany, and Belgrade, former Yugoslavia. Since then, MARV has caused sporadic outbreaks of human disease with high case fatality rates in parts of Africa, with the largest outbreak occurring in 2004/05 in Angola. From 2021 to 2023, MARV outbreaks occurred in Guinea, Ghana, New Guinea, and Tanzania, emphasizing the expansion of its endemic area into new geographical regions. There are currently no approved vaccines or therapeutics targeting MARV, but several vaccine candidates have shown promise in preclinical studies. We compared three vaccine platforms simultaneously by vaccinating hamsters with either a single dose of an adenovirus-based (ChAdOx-1 MARV) vaccine, an alphavirus replicon-based RNA (LION-MARV) vaccine, or a recombinant vesicular stomatitis virus-based (VSV-MARV) vaccine, all expressing the MARV glycoprotein as the antigen. Lethal challenge with hamster-adapted MARV 4 weeks after vaccination resulted in uniform protection of the VSV-MARV and LION-MARV groups and 83% of the ChAdOx-1 MARV group. Assessment of the antigen-specific humoral response and its functionality revealed vaccine-platform-dependent differences, particularly in the Fc effector functions. [ABSTRACT FROM AUTHOR]
- Published
- 2024
- Full Text
- View/download PDF
37. Preclinical Safety Assessment of the EBS-LASV Vaccine Candidate against Lassa Fever Virus.
- Author
-
Matassov, Demetrius, DeWald, Lisa Evans, Hamm, Stefan, Nowak, Rebecca M., Gerardi, Cheryl S., Latham, Theresa E., Xu, Rong, Luckay, Amara, Chen, Tracy, Tremblay, Marc, Shearer, Jeffry, Wynn, Melissa, Eldridge, John H., Warfield, Kelly, and Spurgers, Kevin
- Subjects
LASSA fever ,VESICULAR stomatitis ,DELETION mutation ,LYMPH nodes ,IMMUNE response - Abstract
There are currently no prophylactic vaccines licensed to protect against Lassa fever caused by Lassa virus (LASV) infection. The Emergent BioSolutions (EBS) vaccine candidate, EBS-LASV, is being developed for the prevention of Lassa fever. EBS-LASV is a live-attenuated recombinant Vesicular Stomatitis Virus (rVSV)-vectored vaccine encoding the surface glycoprotein complex (GPC) from LASV and has two attenuating vector modifications: a gene shuffle of the VSV N gene and a deletion of the VSV G gene. Preclinical studies were performed to evaluate EBS-LASV's neurovirulence potential following intracranial (IC) injection and to determine the biodistribution and vector replication following intramuscular (IM) inoculation in mice. In addition, the potential EBS-LASV toxicity was assessed using repeated-dose IM EBS-LASV administration to rabbits. All mice receiving the IC injection of EBS-LASV survived, while mice administered the unattenuated control vector did not. The vaccine was only detected in the muscle at the injection site, draining lymph nodes, and the spleen over the first week following IM EBS-LASV injection in mice, with no detectable plasma viremia. No toxicity was observed in rabbits receiving a three-dose regimen of EBS-LASV. These studies demonstrate that EBS-LASV is safe when administered to animals and supported a first-in-human dose-escalation, safety, and immunogenicity clinical study. [ABSTRACT FROM AUTHOR]
- Published
- 2024
- Full Text
- View/download PDF
38. Eucalyptus Essential Oil Inhibits Cell Infection by SARS-CoV-2 Spike Pseudotyped Lentivirus.
- Author
-
Fernandez, Sara Alonso, Pelaez-Prestel, Hector F., Ras-Carmona, Alvaro, Mozas-Gutierrez, Juan, Reyes-Manzanas, Raquel, and Reche, Pedro A.
- Subjects
SARS-CoV-2 ,VESICULAR stomatitis ,ANGIOTENSIN converting enzyme ,ESSENTIAL oils ,CYTOTOXINS - Abstract
Severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) remains a public health concern due to infections with new SARS-CoV-2 variants. Therefore, finding effective preventive and therapeutic treatments against all SARS-CoV-2 variants is of great interest. In this study, we examined the capacity of eucalyptus essential oil (EEO) and eucalyptol (EOL) to prevent SARS-CoV-2 infection, using as a model SARS-CoV-2 Spike pseudotyped lentivirus (SARS-CoV-2 pseudovirus) and 293T cells transfected with human angiotensin-converting enzyme 2 (hACE2-293T cells). First, we determined the cytotoxicity of EEO and EOL using the MTT colorimetric assay, selecting non-cytotoxic concentrations ≤ 0.1% (v/v) for further analysis. Subsequently, we evaluated the capacity of EEO and EOL in cell cultures to preclude infection of hACE2-293T cells by SARS-CoV-2 pseudovirus, using a luciferase-based assay. We found that EEO and EOL significantly reduced SARS-CoV-2 pseudovirus infection, obtaining IC
50 values of 0.00895% and 0.0042% (v/v), respectively. Likewise, EEO and EOL also reduced infection by vesicular stomatitis virus (VSV) pseudovirus, although higher concentrations were required. Hence, EEO and EOL may be able to inhibit SARS-CoV-2 infection, at least partially, through a Spike-independent pathway, supporting the implementation of aromatherapy with these agents as a cost-effective antiviral measure. [ABSTRACT FROM AUTHOR]- Published
- 2024
- Full Text
- View/download PDF
39. The RNA helicase DHX35 functions as a co-sensor for RIG-I-mediated innate immunity.
- Author
-
Qiao, Yuan, Zhu, Shan, Yang, Ning, Zou, Shan-Shan, Gao, Bao, Wu, Jing, Liu, Chunyan, Li, Xiaoping, Liu, Yong-Jun, and Chen, Jingtao
- Subjects
- *
RNA helicase , *NATURAL immunity , *INTERFERON regulatory factors , *NUCLEIC acids , *VESICULAR stomatitis , *DNA helicases , *ADAPTOR proteins - Abstract
RNA helicases are involved in the innate immune response against pathogens, including bacteria and viruses; however, their mechanism in the human airway epithelial cells is still not fully understood. Here, we demonstrated that DEAH (Asp-Glu-Ala-His) box polypeptide 35 (DHX35), a member of the DExD/H (Asp-Glu-x-Asp/His)-box helicase family, boosts antiviral innate immunity in human airway epithelial cells. DHX35 knockdown attenuated the production of interferon-β (IFN-β), IL6, and CXCL10, whereas DHX35 overexpression increased their production. Upon stimulation, DHX35 was constitutively expressed, but it translocated from the nucleus into the cytosol, where it recognized cytosolic poly(I:C) and poly(dA:dT) via its HELICc domain. Mitochondrial antiviral signaling protein (MAVS) acted as an adaptor for DHX35 and interacted with the HELICc domain of DHX35 using amino acids 360–510. Interestingly, DHX35 interacted with retinoic acid-inducible gene 1 (RIG-I), enhanced the binding affinity of RIG-I with poly(I:C) and poly(dA:dT), and formed a signalsome with MAVS to activate interferon regulatory factor 3 (IRF3), NF-κB-p65, and MAPK signaling pathways. These results indicate that DHX35 not only acted as a cytosolic nucleic acid sensor but also synergized with RIG-I to enhance antiviral immunity in human airway epithelial cells. Our results demonstrate a novel molecular mechanism for DHX35 in RIG-I-mediated innate immunity and provide a novel candidate for drug and vaccine design to control viral infections in the human airway. Author summary: RIG-I plays an important role in defending against RNA viral infections, such as SARS-CoV-2, influenza virus, and vesicular stomatitis virus (VSV). DHX35 is a member of the Asp-Glu-x-Asp/His (DExD/H)-box helicase family. This study revealed a novel function of DHX35 in regulating activation of the RIG-I signaling pathway. DHX35 translocates from the nucleus into cytosol where it recognizes cytosolic nucleic acids upon stimulation, and interacts with MAVS to activate downstream signaling pathways. More importantly, DHX35 interacts with RIG-I to enhance its binding affinity to nucleic acids. Taken together, our findings demonstrate a novel molecular mechanism of RIG-I-mediated innate immunity in human airway epithelial cells. [ABSTRACT FROM AUTHOR]
- Published
- 2024
- Full Text
- View/download PDF
40. Recombinant OC43 SARS-CoV-2 spike replacement virus: An improved BSL-2 proxy virus for SARS-CoV-2 neutralization assays.
- Author
-
Zhe Hu, Domingo López-Muñoz, Alberto, Kosik, Ivan, Tiansheng Li, Callahan, Victoria, Brooks, Kelsie, Yee, Debra S., Holly, Jaroslav, Santos, Jefferson J. S., Brant, Ayslan Castro, Johnson, Reed F., Kazuyo Takeda, Zhi-Ming Zheng, Brenchley, Jason M., Yewdell, Jonathan W., and Fox, Julie M.
- Subjects
- *
SARS-CoV-2 , *IMMUNOGLOBULIN A , *COVID-19 vaccines , *VESICULAR stomatitis , *IMMUNOGLOBULIN G - Abstract
We generated a replication-competent OC43 human seasonal coronavirus (CoV) expressing the severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) spike in place of the native spike (rOC43-CoV2 S). This virus is highly attenuated relative to OC43 and SARS-CoV-2 in cultured cells and animals and is classified as a biosafety level 2 (BSL-2) agent by the NIH biosafety committee. Neutralization of rOC43-CoV2 S and SARS-CoV-2 by S-specific monoclonal antibodies and human sera is highly correlated, unlike recombinant vesicular stomatitis virus-CoV2 S. Single-dose immunization with rOC43-CoV2 S generates high levels of neutralizing antibodies against SARS-CoV-2 and fully protects human ACE2 transgenic mice from SARS-CoV-2 lethal challenge, despite nondetectable replication in respiratory and nonrespiratory organs. rOC43-CoV2 S induces S-specific serum and airway mucosal immunoglobulin A and IgG responses in rhesus macaques. rOC43-CoV2 S has enormous value as a BSL-2 agent to measure S-specific antibodies in the context of a bona fide CoV and is a candidate live attenuated SARS-CoV-2 mucosal vaccine that preferentially replicates in the upper airway. [ABSTRACT FROM AUTHOR]
- Published
- 2024
- Full Text
- View/download PDF
41. Histone deacetylase 8 promotes innate antiviral immunity through deacetylation of RIG-I.
- Author
-
Huijun Zhang, Tingli Liu, Xinhua Liu, Fenfen You, Jiaheng Yang, Nan Zhang, Ying Huang, and Gaofeng Liang
- Subjects
NATURAL immunity ,VESICULAR stomatitis ,DEACETYLATION ,VIRUS diseases ,CELLULAR signal transduction ,SIRTUINS ,INTERFERON receptors - Abstract
Histone deacetylates family proteins have been studied for their function in regulating viral replication by deacetylating non-histone proteins. RIG-I (Retinoic acid-inducible gene I) is a critical protein in RNA virus-induced innate antiviral signaling pathways. Our previous research showed that HDAC8 (histone deacetylase 8) involved in innate antiviral immune response, but the underlying mechanism during virus infection is still unclear. In this study, we showed that HDAC8 was involved in the regulation of vesicular stomatitis virus (VSV) replication. Over-expression of HDAC8 inhibited while knockdown promoted VSV replication. Further exploration demonstrated that HDAC8 interacted with and deacetylated RIG-I, which eventually lead to enhance innate antiviral immune response. Collectively, our data clearly demonstrated that HDAC8 inhibited VSV replication by promoting RIG-I mediated interferon production and downstream signaling pathway. [ABSTRACT FROM AUTHOR]
- Published
- 2024
- Full Text
- View/download PDF
42. Reassortment incompetent live attenuated and replicon influenza vaccines provide improved protection against influenza in piglets.
- Author
-
Graaf-Rau, Annika, Schmies, Kathrin, Breithaupt, Angele, Ciminski, Kevin, Zimmer, Gert, Summerfield, Artur, Sehl-Ewert, Julia, Lillie-Jaschniski, Kathrin, Helmer, Carina, Bielenberg, Wiebke, grosse Beilage, Elisabeth, Schwemmle, Martin, Beer, Martin, and Harder, Timm
- Subjects
INFLUENZA vaccines ,PIGLETS ,ANIMAL herds ,INFLUENZA ,VESICULAR stomatitis ,SWINE influenza - Abstract
Swine influenza A viruses (swIAV) cause an economically important respiratory disease in modern pig production. Continuous virus transmission and antigenic drift are difficult to control in enzootically infected pig herds. Here, antibody-positive piglets from a herd enzootically infected with swIAV H1N2 (clade 1 A.3.3.2) were immunized using a homologous prime-boost vaccination strategy with novel live attenuated influenza virus (LAIV) based on a reassortment-incompetent bat influenza-swIAV chimera or a vesicular stomatitis virus-based replicon vaccine. Challenge infection of vaccinated piglets by exposure to H1N2 swIAV-infected unvaccinated seeder pigs showed that both LAIV and replicon vaccine markedly reduced virus replication in the upper and lower respiratory tract, respectively, compared to piglets immunized with commercial heterologous or autologous adjuvanted whole-inactivated virus vaccines. Our novel vaccines may aid in interrupting continuous IAV transmission chains in large enzootically infected pig herds, improve the health status of the animals, and reduce the risk of zoonotic swIAV transmission. [ABSTRACT FROM AUTHOR]
- Published
- 2024
- Full Text
- View/download PDF
43. Interrogating Genomes and Geography to Unravel Multiyear Vesicular Stomatitis Epizootics.
- Author
-
Humphreys, John M., Shults, Phillip T., Velazquez-Salinas, Lauro, Bertram, Miranda R., Pelzel-McCluskey, Angela M., Pauszek, Steven J., Peters, Debra P. C., and Rodriguez, Luis L.
- Subjects
- *
VESICULAR stomatitis , *VIRAL variation , *VIRUS diversity , *VECTOR-borne diseases , *SUMMER - Abstract
We conducted an integrative analysis to elucidate the spatial epidemiological patterns of the Vesicular Stomatitis New Jersey virus (VSNJV) during the 2014–15 epizootic cycle in the United States (US). Using georeferenced VSNJV genomics data, confirmed vesicular stomatitis (VS) disease cases from surveillance, and a suite of environmental factors, our study assessed environmental and phylogenetic similarity to compare VS cases reported in 2014 and 2015. Despite uncertainties from incomplete virus sampling and cross-scale spatial processes, patterns suggested multiple independent re-invasion events concurrent with potential viral overwintering between sequential seasons. Our findings pointed to a geographically defined southern virus pool at the US–Mexico interface as the source of VSNJV invasions and overwintering sites. Phylodynamic analysis demonstrated an increase in virus diversity before a rise in case numbers and a pronounced reduction in virus diversity during the winter season, indicative of a genetic bottleneck and a significant narrowing of virus variation between the summer outbreak seasons. Environment–vector interactions underscored the central role of meta-population dynamics in driving disease spread. These insights emphasize the necessity for location- and time-specific management practices, including rapid response, movement restrictions, vector control, and other targeted interventions. [ABSTRACT FROM AUTHOR]
- Published
- 2024
- Full Text
- View/download PDF
44. Hexestrol, an estrogen receptor agonist, inhibits Lassa virus entry.
- Author
-
ZihanZhang, Toru Takenaga, Fehling, Sarah Katharina, Manabu Igarashi, Takatsugu Hirokawa, Yukiko Muramoto, Koji Yamauchi, Chiho Onishi, Masahiro Nakano, Shuzo Urata, Groseth, Allison, Strecker, Thomas, and Takeshi Noda
- Subjects
- *
ESTROGEN receptors , *MEMBRANE fusion , *LASSA fever , *VESICULAR stomatitis , *HEMORRHAGIC fever , *VACCINE approval - Abstract
Lassa virus (LASV) is the causative agent of human Lassa fever which in severe cases manifests as hemorrhagic fever leading to thousands of deaths annually. However, no approved vaccines or antiviral drugs are currently available. Recently, we screened approximately 2,500 compounds using a recombinant vesicular stomatitis virus (VSV) expressing LASV glycoprotein GP (VSV-LASVGP) and identified a P-glycoprotein inhibitor as a potential LASV entry inhibitor. Here, we show that another identified candidate, hexestrol (HES), an estrogen receptor agonist, is also a LASV entry inhibitor. HES inhibited VSV-LASVGP replication with a 50% inhibitory concentration (IC50) of 0.63 µM. Importantly, HES also inhibited authentic LASV replication with IC50 values of 0.31 µM–0.61 µM. Time-of-addition and cell-based membrane fusion assays suggested that HES inhibits the membrane fusion step during virus entry. Alternative estrogen receptor agonists did not inhibit VSV-LASVGP replication, suggesting that the estrogen receptor itself is unlikely to be involved in the antiviral activity of HES. Generation of a HES-resistant mutant revealed that the phenylalanine at amino acid position 446 (F446) of LASVGP, which is located in the transmembrane region, conferred resistance to HES. Although mutation of F446 enhanced the membrane fusion activity of LASVGP, it exhibited reduced VSV-LASVGP replication, most likely due to the instability of the pre-fusion state of LASVGP. Collectively, our results demonstrated that HES is a promising anti-LASV drug that acts by inhibiting the membrane fusion step of LASV entry. This study also highlights the importance of the LASVGP transmembrane region as a target for anti-LASV drugs. IMPORTANCE Lassa virus (LASV), the causative agent of Lassa fever, is the most devastating mammarenavirus with respect to its impact on public health in West Africa. However, no approved antiviral drugs or vaccines are currently available. Here, we identified hexestrol (HES), an estrogen receptor agonist, as the potential antiviral candidate drug. We showed that the estrogen receptor itself is not involved in the antiviral activity. HES directly bound to LASVGP and blocked membrane fusion, thereby inhibiting LASV infection. Through the generation of a HES-resistant virus, we found that phenylalanine at position 446 (F446) within the LASVGP transmembrane region plays a crucial role in the antiviral activity of HES. The mutation at F446 caused reduced virus replication, likely due to the instability of the pre-fusion state of LASVGP. These findings highlight the potential of HES as a promising candidate for the development of antiviral compounds targeting LASV. [ABSTRACT FROM AUTHOR]
- Published
- 2024
- Full Text
- View/download PDF
45. Characterization of human tibrovirus envelope glycoproteins.
- Author
-
Yannick Munyeku-Bazitama, Takeshi Saito, Takanari Hattori, Hiroko Miyamoto, Boniface Pongombo Lombe, Akina Mori-Kajihara, Masahiro Kajihara, Jean-Jacques Muyembe-Tamfum, Manabu Igarashi, Eun-sil Park, Shigeru Morikawa, Makiala-Mandanda, Sheila, and Ayato Takada
- Subjects
- *
VIRAL tropism , *GLYCOPROTEINS , *VESICULAR stomatitis , *VIRUS-like particles , *MEMBRANE fusion , *DEATH rate , *VIRAL envelope proteins - Abstract
Tibroviruses are novel rhabdoviruses detected in humans, cattle, and arthropods. Four tibroviruses are known to infect humans: Bas-Congo virus (BASV), Ekpoma virus 1 (EKV-1), Ekpoma virus 2, and Mundri virus. However, since none of them has been isolated, their biological properties are largely unknown. We aimed to characterize the human tibrovirus glycoprotein (G), which likely plays a pivotal role in viral tropism and pathogenicity. Human tibrovirus Gs were found to share some primary structures and display 14 conserved cysteine residues, although their overall amino acid homology was low (29%–48%). Multiple potential glycosylation sites were found on the G molecules, and endoglycosidase H- and peptide-N-glycosidase F-sensitive glycosylation was confirmed. AlphaFold-predicted three-dimensional (3D) structures of human tibrovirus Gs were overall similar. Membrane fusion mediated by these tibrovirus Gs was induced by acidic pH. The low pH-induced conformational change that triggers fusion was reversible. Virus-like particles (VLPs) were produced by transient expression of Gs in cultured cells and used to produce mouse antisera. Using vesicular stomatitis Indiana virus pseudotyped with Gs, we found that the antisera to the respective tibrovirus VLPs showed limited cross-neutralizing activity. It was also found that human C-type lectins and T-cell immunoglobulin mucin 1 acted as attachment factors for G-mediated entry into cells. Interestingly, BASV-G showed the highest ability to utilize these molecules. The viruses infected a wide range of cell lines with preferential tropism for humanderived cells whereas the preference of EKV-1 was unique compared with the other human tibroviruses. These findings provide fundamental information to understand the biological properties of the human tibroviruses. IMPORTANCE Human tibroviruses are poorly characterized emerging rhabdoviruses associated with either asymptomatic infection or severe disease with a case fatality rate as high as 60% in humans. However, the extent and burden of human infection as well as factors behind differences in infection outcomes are largely unknown. In this study, we characterized human tibrovirus glycoproteins, which play a key role in virus-host interactions, mainly focusing on their structural and antigenic differences and cellular tropism. Our results provide critical information for understanding the biological properties of these novel viruses and for developing appropriate preparedness interventions such as diagnostic tools, vaccines, and effective therapies. [ABSTRACT FROM AUTHOR]
- Published
- 2024
- Full Text
- View/download PDF
46. Development of a Cell-Based Reporter Potency Assay for Live Virus Vaccines.
- Author
-
Sun, Dengyun, Meyer, Brian K., Deevi, Dhanvanthri S., Mirza, Asra, He, Li, Gruber, Ashley, Abbondanzo, Susan J., Benton, Noah A., Whiteman, Melissa C., Capen, Robert C., and Gurney, Kevin B.
- Subjects
VIRAL vaccines ,VESICULAR stomatitis ,MEASLES virus ,TISSUE culture ,VIRUS diseases - Abstract
The rapid development of potency assays is critical in the development of life-saving vaccines. The traditional plaque assay or fifty percent tissue culture infectious dose (TCID
50 ) assay used to measure the potency of live virus vaccines is time consuming, labor intensive, low throughput and with high variability. Described here is the development and qualification of a cell-based reporter potency assay for two vaccines for respiratory viral infection, one based on the recombinant vesicular stomatitis virus (rVSV) backbone, termed Vaccine 1 in this paper, and the other based on the measles virus vector, termed Vaccine 2. The reporter potency assay used a Vero E6 cell line engineered to constitutively express NanuLuc® luciferase, termed the VeroE6-NLuc or JM-1 cell line. Infection of JM-1 cells by a live virus, such as rVSV or measles virus, causes a cytopathic effect (CPE) and release of NanuLuc® from the cytoplasm into the supernatant, the amount of which reflects the intensity of the viral infection. The relative potency was calculated by comparison to a reference standard using parallel line analysis (PLA) in a log–log linear model. The reporter assay demonstrated good linearity, accuracy, and precision, and is therefore suitable for a vaccine potency assay. Further evaluation of the Vaccine 1 reporter assay demonstrated the robustness to a range of deliberate variation of the selected assay parameters and correlation with the plaque assay. In conclusion, we have demonstrated that the reporter assay using the JM-1 cell line could be used as a potency assay to support the manufacturing and release of multiple live virus vaccines. [ABSTRACT FROM AUTHOR]- Published
- 2024
- Full Text
- View/download PDF
47. The lipopeptide Pam3CSK4 inhibits Rift Valley fever virus infection and protects from encephalitis.
- Author
-
Griesman, Trevor, McMillen, Cynthia M., Negatu, Seble Getenet, Hulahan, Jesse J., Whig, Kanupriya, Dohnalová, Lenka, Dittmar, Mark, Thaiss, Christoph A., Jurado, Kellie A., Schultz, David C., Hartman, Amy L., and Cherry, Sara
- Subjects
- *
RIFT Valley fever , *VIRUS diseases , *MOSQUITO control , *ENCEPHALITIS , *VESICULAR stomatitis , *MEMBRANE fusion - Abstract
Rift Valley fever virus (RVFV) is an encephalitic bunyavirus that can infect neurons in the brain. There are no approved therapeutics that can protect from RVFV encephalitis. Innate immunity, the first line of defense against infection, canonically antagonizes viruses through interferon signaling. We found that interferons did not efficiently protect primary cortical neurons from RVFV, unlike other cell types. To identify alternative neuronal antiviral pathways, we screened innate immune ligands and discovered that the TLR2 ligand Pam3CSK4 inhibited RVFV infection, and other bunyaviruses. Mechanistically, we found that Pam3CSK4 blocks viral fusion, independent of TLR2. In a mouse model of RVFV encephalitis, Pam3CSK4 treatment protected animals from infection and mortality. Overall, Pam3CSK4 is a bunyavirus fusion inhibitor active in primary neurons and the brain, representing a new approach toward the development of treatments for encephalitic bunyavirus infections. Author summary: Rift Valley fever virus (RVFV) is an emerging mosquito-borne pathogen, which has shown the potential to spread geographically, and which can cause severe disease in humans. One of the most serious complications of RVFV infection is invasion into the brain. There are no approved treatments for RVFV infection, or any other bunyavirus that infects the brain. We found that the classically antiviral interferon pathway was not protective in neurons. This led us to screen for new antivirals, and we identified Pam3CSK4, which inhibited RVFV and related bunyaviruses, but not vesicular stomatitis virus. We found that the antiviral activity of Pam3CSK4 was separate from its previously characterized activity as a TLR2 agonist. Instead, Pam3CSK4 blocked RVFV entry at the step of viral membrane fusion. Importantly, we found that Pam3CSK4 prevented RVFV infection in the mouse brain. This work reveals that Pam3CSK4 prevents encephalitic bunyavirus infection in neurons, a vulnerable cell type, and future research should evaluate the therapeutic potential of Pam3CSK4. [ABSTRACT FROM AUTHOR]
- Published
- 2024
- Full Text
- View/download PDF
48. Expression of tumor antigens within an oncolytic virus enhances the anti-tumor T cell response.
- Author
-
Webb, Mason J., Sangsuwannukul, Thanich, van Vloten, Jacob, Evgin, Laura, Kendall, Benjamin, Tonne, Jason, Thompson, Jill, Metko, Muriel, Moore, Madelyn, Chiriboga Yerovi, Maria P., Olin, Michael, Borgatti, Antonella, McNiven, Mark, Monga, Satdarshan P. S., Borad, Mitesh J., Melcher, Alan, Roberts, Lewis R., and Vile, Richard
- Subjects
T cells ,TUMOR antigens ,IMMUNE checkpoint inhibitors ,ONCOLYTIC virotherapy ,VESICULAR stomatitis ,CELL populations ,T cell receptors - Abstract
Although patients benefit from immune checkpoint inhibition (ICI) therapy in a broad variety of tumors, resistance may arise from immune suppressive tumor microenvironments (TME), which is particularly true of hepatocellular carcinoma (HCC). Since oncolytic viruses (OV) can generate a highly immune-infiltrated, inflammatory TME, OVs could potentially restore ICI responsiveness via recruitment, priming, and activation of anti-tumor T cells. Here we find that on the contrary, an oncolytic vesicular stomatitis virus, expressing interferon-ß (VSV-IFNß), antagonizes the effect of anti-PD-L1 therapy in a partially anti-PD-L1-responsive model of HCC. Cytometry by Time of Flight shows that VSV-IFNß expands dominant anti-viral effector CD8 T cells with concomitant relative disappearance of anti-tumor T cell populations, which are the target of anti-PD-L1. However, by expressing a range of HCC tumor antigens within VSV, combination OV and anti-PD-L1 therapeutic benefit could be restored. Our data provide a cautionary message for the use of highly immunogenic viruses as tumor-specific immune-therapeutics by showing that dominant anti-viral T cell responses can inhibit sub-dominant anti-tumor T cell responses. However, through encoding tumor antigens within the virus, oncolytic virotherapy can generate anti-tumor T cell populations upon which immune checkpoint blockade can effectively work. Oncolytic viruses create an inflamed tumour microenvironment allowing T cells to respond to immune checkpoint blockade therapy more efficiently. Authors here show that in a hepatocellular carcinoma model, a dominant anti-viral rather than anti-tumour T cell response is elicited by an oncolytic vesicular stomatitis virus, unless the virus is designed to express tumour antigens, which restores therapeutic benefit. [ABSTRACT FROM AUTHOR]
- Published
- 2024
- Full Text
- View/download PDF
49. Development of a novel PCV2 and PCV3 vaccine using virus-like vesicles incorporating Venezuelan equine encephalomyelitis virus-containing vesicular stomatitis virus glycoprotein.
- Author
-
Ying Wang, Min Su, Yongshuang Huang, Jianle Ren, Sheng Niu, Yujun Zhao, Fang Yan, Yi Yan, and Wen-xia Tian
- Subjects
VENEZUELAN equine encephalomyelitis ,VESICULAR stomatitis ,SEMLIKI Forest virus ,CIRCOVIRUS diseases ,CYTOSKELETAL proteins - Abstract
Porcine circovirus disease (PCV) causes substantial economic losses in the pig industry, primarily from porcine circovirus type 2 (PCV2) and porcine circovirus type 3 (PCV3). Novel vaccines are necessary to prevent and control PCV infections. PCV coat proteins are crucial for eliciting immunogenic proteins that induce the production of antibodies and immune responses. A vaccine platform utilizing Semliki Forest virus RNA replicons expressing vesicular stomatitis virus glycoprotein (VSV-G), was recently developed. This platform generates virus-like vesicles (VLVs) containing VSV-G exclusively, excluding other viral structural proteins. In our study, we developed a novel virus-like vesicle vaccine by constructing recombinant virus-like vesicles (rVLVs) that also express EGFP. These rVLVs were created using the RNA replicon of Venezuelan equine encephalomyelitis (VEEV) and New Jersey serotype VSV-G. The rVLVs underwent characterization and safety evaluation in vitro. Subsequently, rVLVs expressing PCV2d-Cap and PCV3-Cap proteins were constructed. Immunization of C57 mice with these rVLVs led to a significant increase in anti-porcine circovirus type 2 and type 3 capsid protein antibodies in mouse serum. Additionally, a cellular immune response was induced, as evidenced by high production of IFN-γ and IL-4 cytokines. Overall, this study demonstrates the feasibility of developing a novel porcine circovirus disease vaccine based on rVLVs. [ABSTRACT FROM AUTHOR]
- Published
- 2024
- Full Text
- View/download PDF
50. Development of Robust Freeze-Drying Process for Long-Term Stability of rVSV-SARS-CoV-2 Vaccine.
- Author
-
Khan, MD Faizul Hussain, Youssef, Maryam, Nesdoly, Sean, and Kamen, Amine A.
- Subjects
- *
FREEZE-drying , *VESICULAR stomatitis , *VIRAL vaccines , *VIRUS inactivation , *VIRUS isolation , *GELATIN , *TREHALOSE - Abstract
The thermostability of vaccines, particularly enveloped viral vectored vaccines, remains a challenge to their delivery wherever needed. The freeze-drying of viral vectored vaccines is a promising approach but remains challenging due to the water removal process from the outer and inner parts of the virus. In the case of enveloped viruses, freeze-drying induces increased stress on the envelope, which often leads to the inactivation of the virus. In this study, we designed a method to freeze-dry a recombinant vesicular stomatitis virus (VSV) expressing the SARS-CoV-2 spike glycoprotein. Since the envelope of VSV is composed of 50% lipids and 50% protein, the formulation study focused on both the protein and lipid portions of the vector. Formulations were prepared primarily using sucrose, trehalose, and sorbitol as cryoprotectants; mannitol as a lyoprotectant; and histidine as a buffer. Initially, the infectivity of rVSV-SARS-CoV-2 and the cake stability were investigated at different final moisture content levels. High recovery of the infectious viral titer (~0.5 to 1 log loss) was found at 3–6% moisture content, with no deterioration in the freeze-dried cakes. To further minimize infectious viral titer loss, the composition and concentration of the excipients were studied. An increase from 5 to 10% in both the cryoprotectants and lyoprotectant, together with the addition of 0.5% gelatin, resulted in the improved recovery of the infectious virus titer and stable cake formation. Moreover, the secondary drying temperature of the freeze-drying process showed a significant impact on the infectivity of rVSV-SARS-CoV-2. The infectivity of the vector declined drastically when the temperature was raised above 20 °C. Throughout a long-term stability study, formulations containing 10% sugar (sucrose/trehalose), 10% mannitol, 0.5% gelatin, and 10 mM histidine showed satisfactory stability for six months at 2–8 °C. The development of this freeze-drying process and the optimized formulation minimize the need for a costly cold chain distribution system. [ABSTRACT FROM AUTHOR]
- Published
- 2024
- Full Text
- View/download PDF
Catalog
Discovery Service for Jio Institute Digital Library
For full access to our library's resources, please sign in.