Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 64
Filter
Add more filters

Country/Region as subject
Publication year range
1.
J Med Virol ; 95(1): e28260, 2023 01.
Article in English | MEDLINE | ID: mdl-36305515

ABSTRACT

Several viruses are known to be associated with the development of certain cancers, including human papilloma virus (HPV), an established causative agent for a range of anogenital and head and neck cancers. However, the causality has been based on the presence of the virus, or its genetic material, in the sampled tumors. We have long wondered if viruses cause cancer via a "hit and run" mechanism such that they are no longer present in the resulting tumors. Here, we hypothesize that the absence of viral genes from the tumor does not necessarily exclude the viral aetiology. To test this, we used an HPV-driven oropharyngeal cancer (OPC) tumor model and CRISPR to delete the viral oncogene, E7. Indeed, the genetic removal of HPV E7 oncogene eliminates tumors in vivo. Remarkably, E7 deleted tumors recurred over time and develop new mutations not previously seen in HPV+ OPC tumors. Importantly, a number of these new mutations are found to be already present in HPV- OPC tumors.


Subject(s)
Head and Neck Neoplasms , Oncogene Proteins, Viral , Oropharyngeal Neoplasms , Papillomavirus Infections , Humans , Human Papillomavirus Viruses , Oncogene Proteins, Viral/genetics , Papillomavirus Infections/pathology , Repressor Proteins/genetics , Neoplasm Recurrence, Local , Oropharyngeal Neoplasms/complications , Oropharyngeal Neoplasms/pathology , Papillomavirus E7 Proteins/genetics
2.
Virus Genes ; 59(2): 312-316, 2023 Apr.
Article in English | MEDLINE | ID: mdl-36474086

ABSTRACT

Clinical and pre-clinical work for a number of cancer types has demonstrated relatively positive outcomes and effective tumour regression when the level and function of p53, a well-established tumour suppressor, is restored. Human papillomavirus (HPV)-driven cancers encode the E6 oncoprotein, which leads to p53 degradation, to allow the carcinogenic process to proceed. Indeed, there have been several attempts to revive p53 function in HPV-driven cancers by both pharmacological and genetic means to increase p53 bioavailability. Here, we employed a CRISPR activation (CRISPRa) approach to overcome HPV-mediated silencing of p53 by hyperexpressing the p53 gene promoter. Our data show that CRISPRa-mediated hyperexpression of p53 leads to HPV+ cervical cancer cell killing and the reduction of cell proliferation. This proof-of-concept data suggest that increasing p53 bioavailability may potentially be a promising therapeutic approach for the treatment of HPV-driven cancers.


Subject(s)
Oncogene Proteins, Viral , Papillomavirus Infections , Uterine Cervical Neoplasms , Female , Humans , Uterine Cervical Neoplasms/genetics , Tumor Suppressor Protein p53/genetics , Tumor Suppressor Protein p53/metabolism , Human Papillomavirus Viruses , Papillomavirus Infections/complications , Papillomavirus Infections/genetics , Clustered Regularly Interspaced Short Palindromic Repeats , Oncogene Proteins, Viral/genetics , Oncogene Proteins, Viral/metabolism , Repressor Proteins/genetics , Repressor Proteins/metabolism , Human papillomavirus 16/genetics , Human papillomavirus 16/metabolism
3.
J Virol ; 95(6)2021 02 24.
Article in English | MEDLINE | ID: mdl-33328314

ABSTRACT

Type I interferon (IFN)-mediated antiviral responses are critical for modulating host-virus responses, and indeed, viruses have evolved strategies to antagonize this pathway. Encephalomyocarditis virus (EMCV) is an important zoonotic pathogen, which causes myocarditis, encephalitis, neurological disease, reproductive disorders, and diabetes in pigs. This study aims to understand how EMCV interacts with the IFN pathway. EMCV circumvents the type I IFN response by expressing proteins that antagonize cellular innate immunity. Here, we show that EMCV VP2 is a negative regulator of the IFN-ß pathway. This occurs via the degradation of the MDA5-mediated cytoplasmic double-stranded RNA (dsRNA) antiviral sensing RIG-I-like receptor (RLR) pathway. We show that structural protein VP2 of EMCV interacts with MDA5, MAVS, and TBK1 through its C terminus. In addition, we found that EMCV VP2 could significantly degrade RLRs by the proteasomal and lysosomal pathways. For the first time, EMCV VP2 was shown to play an important role in EMCV evasion of the type I IFN signaling pathway. This study expands our understanding that EMCV utilizes its capsid protein VP2 to evade the host antiviral response.IMPORTANCE Encephalomyocarditis virus is an important pathogen that can cause encephalitis, myocarditis, neurological diseases, and reproductive disorders. It also causes huge economic losses for the swine industry worldwide. Innate immunity plays an important role in defending the host from pathogen infection. Understanding pathogen microorganisms evading the host immune system is of great importance. Currently, whether EMCV evades cytosolic RNA sensing and signaling is still poorly understood. In the present study, we found that viral protein VP2 antagonized the RLR signaling pathway by degrading MDA5, MAVS, and TBK1 protein expression to facilitate viral replication in HEK293 cells. The findings in this study identify a new mechanism for EMCV evading the host's innate immune response, which provide new insights into the virus-host interaction and help develop new antiviral approaches against EMCV.


Subject(s)
Capsid Proteins/metabolism , Encephalomyocarditis virus/physiology , Interferon-beta/metabolism , Signal Transduction , Adaptor Proteins, Signal Transducing/metabolism , Capsid Proteins/chemistry , Capsid Proteins/genetics , Cardiovirus Infections/immunology , Cardiovirus Infections/virology , DEAD Box Protein 58/antagonists & inhibitors , DEAD Box Protein 58/metabolism , Encephalomyocarditis virus/genetics , Encephalomyocarditis virus/metabolism , HEK293 Cells , Humans , Immune Evasion , Immunity, Innate , Interferon Type I/metabolism , Interferon-Induced Helicase, IFIH1/metabolism , Mutation , Protein Interaction Domains and Motifs , Protein Serine-Threonine Kinases/metabolism , Receptors, Immunologic/antagonists & inhibitors , Receptors, Immunologic/metabolism , Virus Replication
4.
Mol Ther ; 29(7): 2219-2226, 2021 07 07.
Article in English | MEDLINE | ID: mdl-33992805

ABSTRACT

Coronavirus disease 2019 (COVID-19) is caused by severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) infection in humans. Despite several emerging vaccines, there remains no verifiable therapeutic targeted specifically to the virus. Here we present a highly effective small interfering RNA (siRNA) therapeutic against SARS-CoV-2 infection using a novel lipid nanoparticle (LNP) delivery system. Multiple siRNAs targeting highly conserved regions of the SARS-CoV-2 virus were screened, and three candidate siRNAs emerged that effectively inhibit the virus by greater than 90% either alone or in combination with one another. We simultaneously developed and screened two novel LNP formulations for the delivery of these candidate siRNA therapeutics to the lungs, an organ that incurs immense damage during SARS-CoV-2 infection. Encapsulation of siRNAs in these LNPs followed by in vivo injection demonstrated robust repression of virus in the lungs and a pronounced survival advantage to the treated mice. Our LNP-siRNA approaches are scalable and can be administered upon the first sign of SARS-CoV-2 infection in humans. We suggest that an siRNA-LNP therapeutic approach could prove highly useful in treating COVID-19 disease as an adjunctive therapy to current vaccine strategies.


Subject(s)
COVID-19 Drug Treatment , Drug Delivery Systems/methods , Lipids/chemistry , Nanoparticles/chemistry , RNA, Double-Stranded/administration & dosage , RNA, Small Interfering/administration & dosage , RNA, Small Interfering/genetics , SARS-CoV-2/genetics , Administration, Intravenous , Angiotensin-Converting Enzyme 2/genetics , Animals , COVID-19/metabolism , COVID-19/virology , Female , Gene Silencing , HEK293 Cells , Humans , Lung/metabolism , Male , Mice , Mice, Transgenic , RNA, Double-Stranded/genetics , RNA, Viral/genetics , Transcriptome/drug effects , Treatment Outcome
5.
Microb Pathog ; 132: 162-165, 2019 Jul.
Article in English | MEDLINE | ID: mdl-31054871

ABSTRACT

Head and neck cancers (HNCs) are a major health problem and a leading cause of morbidity and mortality worldwide. More than 90% of these tumours are head and neck squamous cell carcinomas (HNSCCs). Amongst the common risk factors for HNCs (tobacco and alcohol use), there is a strong association of human papillomavirus (HPV) with HNSCCs. HPV type 16 (HPV 16), the major high-risk HPV type, is most commonly associated with HPV-driven HNSCCs. The promiscuous nature of the major HPV oncogene, E7, allows its interaction with a myriad of host proteins including STING, a component of the viral DNA-sensing cyclic GMP-AMP synthase (cGAS) - stimulator of interferon genes (STING) machinery. Sensing of viral DNA by the cGAS-STING machinery results in a type I interferon (IFN)-mediated anti-viral response. Amelioration of IFN responses resulting from the direct blockade of STING by E7 was first demonstrated in high-risk HPV type 18 (HPV 18) positive (+) cervical squamous cell carcinoma (CESC) cells. However, the role of E7 from HPV 16 (HPV 16E7) in antagonising cGAS-STING responses have not been investigated, let alone in the context of HNSCCs. Here, we show that HPV 16E7+, but not HPV 16E7 negative (-), HNSCC cells respond poorly to cGAS-STING activation stimulus. We further confirm that this inhibition occurred via the highly conserved LXCXE motif in 16E7. This finding contributes to the better understanding of role of high-risk HPV E7 in blocking cGAS-STING pathway, especially in the context of HNSCCs.


Subject(s)
DNA, Viral/isolation & purification , Head and Neck Neoplasms/virology , Human papillomavirus 16/genetics , Nucleotidyltransferases/genetics , Papillomavirus Infections/virology , Squamous Cell Carcinoma of Head and Neck/virology , Cell Line, Tumor , DNA, Viral/genetics , Gene Expression Regulation, Viral , Head and Neck Neoplasms/complications , Human papillomavirus 16/metabolism , Humans , Interferon Type I/genetics , Interferon Type I/metabolism , Nucleotidyltransferases/antagonists & inhibitors , Nucleotidyltransferases/metabolism , Papillomavirus E7 Proteins/genetics , Papillomavirus E7 Proteins/metabolism , Papillomavirus Infections/complications , Squamous Cell Carcinoma of Head and Neck/complications
6.
Cent Eur J Public Health ; 27(4): 340-344, 2019 12.
Article in English | MEDLINE | ID: mdl-31951696

ABSTRACT

OBJECTIVE: Drug-resistant staphylococci have been a growing threat to the community and hospitals due to the misuse of antibiotics by humans, industrialisation and lack of novel antimicrobials currently available. Little is known about the prevalence of drug-resistant staphylococci in non-healthcare environments outside hospitals in the London area. Staphylococci can spread via contact with contaminated objects. Traffic light buttons present a fast and easy transmission route for staphylococci. METHODS: Traffic light buttons outside a major hospital in London were swabbed and cultured onto selective media to isolate staphylococci bacteria before performing antimicrobial susceptibility testing on the isolates. The identity of the isolates were determined using MALDI-TOF mass spectrometry (MS). Staphylococci isolates resistant to oxacillin were further tested for minimum inhibitory concentration (MIC). PCR analysis of the mecA gene, a gene that confers resistance to oxacillin, is used to determine the level of resistance to oxacillin. RESULTS: Eight different staphylococcal species were identified by MALDI-TOF-MS analysis. Out of the 66 staphylococci isolates, 16 were resistant to multiple antibiotics including six isolates which were oxacillin resistant. CONCLUSION: This work provides evidence of the presence of multidrug-resistant staphylococci in the vicinity of the hospital environment in London.


Subject(s)
Anti-Bacterial Agents/pharmacology , Drug Resistance, Multiple , Staphylococcus/drug effects , Staphylococcus/isolation & purification , Hospitals , Humans , London , Microbial Sensitivity Tests , Oxacillin/pharmacology
7.
Intervirology ; 61(3): 149-154, 2018.
Article in English | MEDLINE | ID: mdl-30145592

ABSTRACT

BACKGROUND/AIMS: Human metapneumovirus (hMPV) is an important human respiratory pathogen and is implicated in an array of respiratory illnesses, ranging from asymptomatic infection to severe bronchiolitis. Currently, there is no reliable vaccine or specific antiviral therapy for hMPV infection and treatment is supportive. The use of ribonucleic acid interference has the potential to change that with the targeting of essential viral genes via small interfering RNAs (siRNAs) offering the ability to directly and rapidly treat viral infections. METHOD: The human lung carcinoma epithelial cell line, A549, was transfected with siRNAs targeting the N and P genes before infecting with hMPV A2 CAN97-83. Viral growth inhibition was then measured by the viral plaque assay and nucleoprotein (N) and phosphoprotein (P) gene knockdown was determined by real-time PCR. RESULTS: In vitro prophylactic use of siRNAs targeting the 3'-abundantly expressed N and P genes of hMPV resulted in potent, sequence-specific viral inhibition. The viral inhibition was specific and not mediated by an anti-viral interferon-ß response or cell death. CONCLUSION: The findings presented here confirmed the highly potent, sequence-specific antiviral effect of siRNAs targeting the N and P gene of hMPV. These results may facilitate the development of a novel therapeutic agent for hMPV control.


Subject(s)
Metapneumovirus/growth & development , Metapneumovirus/genetics , Nucleoproteins/genetics , Phosphoproteins/genetics , RNA Interference , A549 Cells , Gene Knockdown Techniques , Genes, Viral , Humans , RNA, Small Interfering/genetics , RNA, Viral/genetics , Transfection , Viral Proteins/genetics
8.
Eur J Immunol ; 46(2): 329-39, 2016 Feb.
Article in English | MEDLINE | ID: mdl-26542182

ABSTRACT

Cross-presentation is the mechanism by which exogenous Ag is processed for recognition by CD8(+) T cells. Murine CD8α(+) DCs are specialized at cross-presenting soluble and cellular Ag, but in humans this process is poorly characterized. In this study, we examined uptake and cross-presentation of soluble and cellular Ag by human blood CD141(+) DCs, the human equivalent of mouse CD8α(+) DCs, and compared them with human monocyte-derived DCs (MoDCs) and blood CD1c(+) DC subsets. MoDCs were superior in their capacity to internalize and cross-present soluble protein whereas CD141(+) DCs were more efficient at ingesting and cross-presenting cellular Ag. Whilst cross-presentation by CD1c(+) DCs and CD141(+) DCs was dependent on the proteasome, and hence cytosolic translocation, cross-presentation by MoDCs was not. Inhibition of endosomal acidification enhanced cross-presentation by CD1c(+) DCs and MoDCs but not by CD141(+) DCs. These data demonstrate that CD1c(+) DCs, CD141(+) DCs, and MoDCs are capable of cross-presentation; however, they do so via different mechanisms. Moreover, they demonstrate that human CD141(+) DCs, like their murine CD8α(+) DC counterparts, are specialized at cross-presenting cellular Ag, most likely mediated by an enhanced capacity to ingest cellular Ag combined with subtle changes in lysosomal pH during Ag processing and use of the cytosolic pathway.


Subject(s)
CD8-Positive T-Lymphocytes/immunology , Dendritic Cells/immunology , Endocytosis , Phosphoproteins/metabolism , Proteasome Endopeptidase Complex/metabolism , Viral Matrix Proteins/metabolism , Antigens, CD1/metabolism , Antigens, Surface/metabolism , Blood Circulation , Cell Line , Cross-Priming , Humans , Monocytes/immunology , Necrosis , Protein Transport , Solubility , Thrombomodulin
9.
Pharm Biol ; 55(1): 1093-1113, 2017 Dec.
Article in English | MEDLINE | ID: mdl-28198202

ABSTRACT

CONTEXT: Medicinal plants have attracted global attention for their hidden therapeutic potential. Clinacanthus nutans (Burm.f) Lindau (Acanthaceae) (CN) is endemic in Southeast Asia. CN contains phytochemicals common to medicinal plants, such as flavonoids. Traditionally, CN has been used for a broad range of human ailments including snake bites and cancer. OBJECTIVES: This article compiles the ethnomedicinal uses of CN and its phytochemistry, and thus provides a phytochemical library of CN. It also discusses the known pharmacological and biological effects of CN to enable better investigation of CN. METHODS: This literature review was limited to articles and websites published in the English language. MEDLINE and Google Scholar databases were searched from December 2014 to September 2016 using the following keywords: "Clinacanthus nutans" and "Belalai gajah". The results were reviewed to identify relevant articles. Information from relevant selected studies was systematically analyzed from contemporary ethnopharmacological sources, evaluated against scientific literature, and extracted into tables. RESULTS: The literature search yielded 124 articles which were then further scrutinized revealing the promising biological activities of CN, including antimicrobial, antiproliferative, antitumorigenic and anti-inflammatory effects. Few articles discussed the mechanisms for these pharmacological activities. Furthermore, CN was beneficial in small-scale clinical trials for genital Herpes and aphthous stomatitis. CONCLUSION: Despite the rich ethnomedicinal knowledge behind the traditional uses of CN, the current scientific evidence to support these claims remains scant. More research is still needed to validate these medicinal claims, beginning by increasing the understanding of the biological actions of this plant.


Subject(s)
Acanthaceae , Medicine, Traditional , Plant Extracts/pharmacology , Acanthaceae/anatomy & histology , Acanthaceae/chemistry , Anti-Infective Agents/pharmacology , Anti-Inflammatory Agents/pharmacology , Antineoplastic Agents, Phytogenic/pharmacology , Antioxidants/pharmacology , Asia, Southeastern , Humans , Phytochemicals/analysis , Phytotherapy , Plant Extracts/toxicity
10.
Cent Eur J Immunol ; 42(3): 324-330, 2017.
Article in English | MEDLINE | ID: mdl-29204100

ABSTRACT

Carbon dot (Cdot) nanoparticles are an emerging class of carbon nanomaterials with a promising potential for drug delivery and bio imaging applications. Although the interaction between Cdots and non-immune cell types has been well studied, Cdot interactions with macrophages have not been investigated. Exposure of Cdot nanoparticles to J774.1 cells, a murine macrophage cell line, resulted in minimal toxicity, where notable toxicity was only seen with Cdot concentrations higher than 0.5 mg/ml. Flow cytometric analysis revealed that Cdots prepared from citric acid were internalized at significantly higher levels by macrophages compared with those prepared from bamboo leaves. Interestingly, macrophages preferentially took up phenylboronic acid (PB)-modified nanoparticles. By fluorescence microscopy, strong blue light-specific punctate Cdot fluorescence resembling Cdot structures in the cytosolic space was mostly observed in J774.1 macrophages exposed to PB-modified nanoparticles and not unmodified Cdot nanoparticles. PB binds to sialic acid residues that are overexpressed on diseased cell surfaces. Our findings demonstrate that PB-conjugated Cdots can be taken up by macrophages with low toxicity and high efficiency. These modified Cdots can be used to deliver drugs to suppress or eliminate aberrant immune cells such as macrophages associated with tumors such as tumor-associated macrophages.

11.
J Immunol ; 192(4): 1982-9, 2014 Feb 15.
Article in English | MEDLINE | ID: mdl-24453245

ABSTRACT

We established a humanized mouse model incorporating FLT3-ligand (FLT3-L) administration after hematopoietic cell reconstitution to investigate expansion, phenotype, and function of human dendritic cells (DC). FLT3-L increased numbers of human CD141(+) DC, CD1c(+) DC, and, to a lesser extent, plasmacytoid DC (pDC) in the blood, spleen, and bone marrow of humanized mice. CD1c(+) DC and CD141(+) DC subsets were expanded to a similar degree in blood and spleen, with a bias toward expansion of the CD1c(+) DC subset in the bone marrow. Importantly, the human DC subsets generated after FLT3-L treatment of humanized mice are phenotypically and functionally similar to their human blood counterparts. CD141(+) DC in humanized mice express C-type lectin-like receptor 9A, XCR1, CADM1, and TLR3 but lack TLR4 and TLR9. They are major producers of IFN-λ in response to polyinosinic-polycytidylic acid but are similar to CD1c(+) DC in their capacity to produce IL-12p70. Although all DC subsets in humanized mice are efficient at presenting peptide to CD8(+) T cells, CD141(+) DC are superior in their capacity to cross-present protein Ag to CD8(+) T cells following activation with polyinosinic-polycytidylic acid. CD141(+) DC can be targeted in vivo following injection of Abs against human DEC-205 or C-type lectin-like receptor 9A. This model provides a feasible and practical approach to dissect the function of human CD141(+) and CD1c(+) DC and evaluate adjuvants and DC-targeting strategies in vivo.


Subject(s)
Adjuvants, Immunologic/pharmacology , Antigens, CD1/metabolism , Antigens, Surface/metabolism , Dendritic Cells/drug effects , Dendritic Cells/immunology , Glycoproteins/metabolism , Membrane Proteins/pharmacology , Adjuvants, Immunologic/administration & dosage , Animals , Antigen Presentation/immunology , Antigens, CD/immunology , CD8-Positive T-Lymphocytes/immunology , Cell Adhesion Molecule-1 , Cell Adhesion Molecules/metabolism , Female , Humans , Immunoglobulins/metabolism , Interferon-gamma/metabolism , Interleukin-12/metabolism , Lectins, C-Type/immunology , Lectins, C-Type/metabolism , Lymphocyte Activation/immunology , Membrane Proteins/administration & dosage , Mice , Mice, Inbred NOD , Mice, SCID , Minor Histocompatibility Antigens , Poly I-C/immunology , Receptors, Cell Surface/immunology , Receptors, Chemokine/metabolism , Thrombomodulin , Toll-Like Receptor 3/metabolism , Toll-Like Receptor 4/metabolism , Toll-Like Receptor 9/metabolism
12.
Molecules ; 21(8)2016 Aug 17.
Article in English | MEDLINE | ID: mdl-27548121

ABSTRACT

The mistletoes, stem hemiparasites of Asia and Europe, have been used as medicinal herbs for many years and possess sophisticated systems to obtain nutrients from their host plants. Although knowledge about ethnomedicinal uses of mistletoes is prevalent in Asia, systematic scientific study of these plants is still lacking, unlike its European counterparts. This review aims to evaluate the literature on Scurrula and Viscum mistletoes. Both mistletoes were found to have anticancer, antimicrobial, antioxidant and antihypertensive properties. Plants from the genus Scurrula were found to inhibit cancer growth due to presence of phytoconstituents such as quercetin and fatty acid chains. Similar to plants from the genus Viscum, Scurrula also possesses TNFα activity to strengthen the immune system to combat cancer. In line with its anticancer activity, both mistletoes are rich in antioxidants that confer protection against cancer as well as neurodegeneration. Extracts from plants of both genera showed evidence of vasodilation and thus, antihypertensive effects. Other therapeutic effects such as weight loss, postpartum and gastrointestinal healing from different plants of the genus Scurrula are documented. As the therapeutic effects of plants from Scurrula are still in exploration stage, there is currently no known clinical trial on these plants. However, there are few on-going clinical trials for Viscum album that demonstrate the functionalities of these mistletoes. Future work required for exploring the benefits of these plants and ways to develop both parasitic plants as a source of pharmacological drug are explained in this article.


Subject(s)
Mistletoe/physiology , Plant Extracts/pharmacology , Plants, Medicinal/physiology , Animals , Anti-Infective Agents/chemistry , Anti-Infective Agents/pharmacology , Antihypertensive Agents/chemistry , Antihypertensive Agents/pharmacology , Antineoplastic Agents, Phytogenic/chemistry , Antineoplastic Agents, Phytogenic/pharmacology , Antioxidants/chemistry , Antioxidants/pharmacology , China , Ethnopharmacology , Europe , Humans , Mistletoe/chemistry , Plant Extracts/chemistry , Plants, Medicinal/chemistry
13.
Virulence ; 15(1): 2383559, 2024 Dec.
Article in English | MEDLINE | ID: mdl-39066684

ABSTRACT

A variety of animals can be infected by encephalomyocarditis virus (EMCV). EMCV is the established causative agent of myocarditis and encephalitis in some animals. EMCV causes high fatality in suckling and weaning piglets, making pigs the most susceptible domestic animal species. Importantly, EMCV has zoonotic potential to infect the human population. The ability of the pathogen to avoid and undermine the initial defence mechanism of the host contributes to its virulence and pathogenicity. A large body of literature highlights the intricate strategies employed by EMCV to escape the innate immune machinery to suit its "pathogenic needs." Here, we also provide examples on how EMCV interacts with certain host proteins to dampen the infection process. Hence, this concise review aims to summarize these findings in a compendium of decades of research on this exciting yet underappreciated topic.


Subject(s)
Cardiovirus Infections , Encephalomyocarditis virus , Host-Pathogen Interactions , Immunity, Innate , Encephalomyocarditis virus/pathogenicity , Encephalomyocarditis virus/immunology , Encephalomyocarditis virus/physiology , Animals , Cardiovirus Infections/virology , Cardiovirus Infections/immunology , Cardiovirus Infections/veterinary , Swine , Humans , Host-Pathogen Interactions/immunology , Myocarditis/virology , Myocarditis/immunology , Virulence , Swine Diseases/virology , Swine Diseases/immunology
14.
Expert Rev Anti Infect Ther ; : 1-11, 2024 Jul 09.
Article in English | MEDLINE | ID: mdl-38973346

ABSTRACT

INTRODUCTION: Respiratory syncytial virus (RSV) causes bronchiolitis and other respiratory issues in immunocompromised individuals, the elderly, and children. After six decades of research, we have only recently seen the approval of two RSV vaccines, Arexvy and Abrysvo. Direct-acting antivirals against RSV have been more difficult to develop with ribavirin and palivizumab giving very modest reductions in hospitalizations and no differences in mortality. Recently, nirsevimab was licensed and has proven to be much more effective when given prophylactically. These are delivered intravenously (IV) and intramuscularly (IM), but an intranasal (IN) antiviral has several advantages in terms of ease of use, lower resource need, and acting at the site of infection. AREAS COVERED: In this paper, we review the available literature on the current pre-clinical research landscape of anti-RSV therapeutics tested for IN delivery. EXPERT OPINION: As RSV is a respiratory virus that infects both the upper and lower respiratory tracts, efforts are focused on developing a therapeutic that can be delivered via the nasal route. The rationale is to directly target the replicating virus with an obvious respiratory tract tropism. This approach will not only pave the way for a nasal delivery approach aimed at reducing respiratory viral illness but also controlling aerosol virus transmission.

15.
Antiviral Res ; 222: 105815, 2024 02.
Article in English | MEDLINE | ID: mdl-38246206

ABSTRACT

There remains a striking overall mortality burden of COVID-19 worldwide. Given the waning effectiveness of current SARS-CoV-2 antivirals due to the rapid emergence of new variants of concern (VOC), we employed a direct-acting molecular therapy approach using gene silencing RNA interference (RNAi) technology. In this study, we developed and screened several ultra-conserved small-interfering RNAs (siRNAs) before selecting one potent siRNA candidate for pre-clinical in vivo testing. This non-immunostimulatory, anti-SARS-CoV-2 siRNA candidate maintains its antiviral activity against all tested SARS-CoV-2 VOC and works effectively as a single agent. For the first time, significant antiviral effects in both the lungs and nasal cavities of SARS-CoV-2 infected mice were observed when this siRNA candidate was delivered intranasally (IN) as a prophylactic agent with the aid of lipid nanoparticles (LNPs). Importantly, a pre-exposure prophylactic IN-delivered anti-SARS-CoV-2 siRNA antiviral that can ameliorate viral replication in the nasal cavity could potentially prevent aerosol spread of respiratory viruses. An IN delivery approach would allow for the development of a direct-acting nasal spray approach that could be self-administered prophylactically.


Subject(s)
COVID-19 , Animals , Mice , RNA, Small Interfering/genetics , COVID-19/prevention & control , Nasal Cavity , SARS-CoV-2/genetics , Antiviral Agents/therapeutic use , Lung
16.
Cancer Manag Res ; 16: 1053-1061, 2024.
Article in English | MEDLINE | ID: mdl-39183757

ABSTRACT

Purpose: This study aimed to investigate the clinicopathological characteristics of oral squamous cell carcinoma (OSCC) patients and analyze the relationship between the degree of differentiation and factors including age, sex, stage, and tumor location in West Java, Indonesia. Patients and Methods: A retrospective cross-sectional study was conducted at the Central Referral and Teaching Hospital in West Java, Indonesia. The data were collected by reviewing medical records with International Classification of Diseases (ICD) codes C00-C06 from 2016 to 2023. Descriptive statistics were employed to summarize the clinicopathological characteristics of OSCC patients. Chi-square, rank Spearman tests, and contingency correlation coefficients were used to analyze the relationship between the degree of differentiation and various factors, such as age, sex, stage, and tumor location of OSCC. Results: Out of the 627 oral cancer patients, 70.49% were diagnosed with OSCC with a gender distribution of 45.7% males and 54.3% females, predominantly within the age range of 30-49 (37.2%). Most OSCC cases were stage IV (37.7%), with the tongue identified as the most common site (68.8%). A consistent trend of higher well-differentiated and moderately differentiated OSCC by age and gender was observed. Statistical analysis revealed no significant correlation between age, gender, tumor location, and the degree of OSCC differentiation (p>0.05). However, a statistically significant correlation was identified between the degree of OSCC differentiation and stage (p<0.001, r=0.460). Conclusion: There is a correlation between the degree of differentiation of OSCC and stage, suggesting significant prognostic implications that can aid in treatment planning and outcome prediction. However, further studies are needed due to the lack of comprehensive data on risk factors and survival rates of oral cancer patients, which is essential for enhancing prevention and treatment strategies for OSCC.

17.
Int J Nanomedicine ; 19: 6931-6943, 2024.
Article in English | MEDLINE | ID: mdl-39005960

ABSTRACT

Purpose: Over the past three years, extensive research has been dedicated to understanding and combating COVID-19. Targeting the interaction between the SARS-CoV-2 Spike protein and the ACE2 receptor has emerged as a promising therapeutic strategy against SARS-CoV-2. This study aimed to develop ACE2-coated virus-like particles (ACE2-VLPs), which can be utilized to prevent viral entry into host cells and efficiently neutralize the virus. Methods: Virus-like particles were generated through the utilization of a packaging plasmid in conjunction with a plasmid containing the ACE2 envelope sequence. Subsequently, ACE2-VLPs and ACE2-EVs were purified via ultracentrifugation. The quantification of VLPs was validated through multiple methods, including Nanosight 3000, TEM imaging, and Western blot analysis. Various packaging systems were explored to optimize the ACE2-VLP configuration for enhanced neutralization capabilities. The evaluation of neutralization effectiveness was conducted using pseudoviruses bearing different spike protein variants. Furthermore, the study assessed the neutralization potential against the Omicron BA.1 variant in Vero E6 cells. Results: ACE2-VLPs showed a high neutralization capacity even at low doses and demonstrated superior efficacy in in vitro pseudoviral assays compared to extracellular vesicles carrying ACE2. ACE2-VLPs remained stable under various environmental temperatures and effectively blocked all tested variants of concern in vitro. Notably, they exhibited significant neutralization against Omicron BA.1 variant in Vero E6 cells. Given their superior efficacy compared to extracellular vesicles and proven success against live virus, ACE2-VLPs stand out as crucial candidates for treating SARS-CoV-2 infections. Conclusion: This novel therapeutic approach of coating VLPs with receptor particles provides a proof-of-concept for designing effective neutralization strategies for other viral diseases in the future.


Subject(s)
Angiotensin-Converting Enzyme 2 , COVID-19 , SARS-CoV-2 , Spike Glycoprotein, Coronavirus , Angiotensin-Converting Enzyme 2/metabolism , Animals , Vero Cells , Chlorocebus aethiops , Humans , COVID-19/virology , Spike Glycoprotein, Coronavirus/metabolism , Spike Glycoprotein, Coronavirus/genetics , Spike Glycoprotein, Coronavirus/chemistry , Antibodies, Neutralizing/pharmacology , HEK293 Cells , Virus Internalization/drug effects
18.
Mol Immunol ; 170: 131-143, 2024 Jun.
Article in English | MEDLINE | ID: mdl-38663254

ABSTRACT

Mammalian reovirus (MRV) is a non-enveloped, gene segmented double-stranded RNA (dsRNA) virus. It is an important zoonotic pathogen that infects many mammals and vertebrates that act as natural hosts and causes respiratory and digestive tract diseases. Studies have reported that RIG-I and MDA5 in the innate immune cytoplasmic RNA-sensing RIG-like receptor (RLR) signaling pathway can recognize dsRNA from MRV and promote antiviral type I interferon (IFN) responses. However, the mechanism by which many MRV-encoded proteins evade the host innate immune response remains unclear. Here, we show that exogenous µ1 protein promoted the proliferation of MRV in vitro, while knockdown of MRV µ1 protein expression by shRNA could impair MRV proliferation. Specifically, µ1 protein inhibited MRV or poly(I:C)-induced IFN-ß expression, and attenuated RIG-I/MDA5-mediated signaling axis transduction during MRV infection. Importantly, we found that µ1 protein significantly decreased IFN-ß mRNA expression induced by MDA5, RIG-I, MAVS, TBK1, IRF3(5D), and degraded the protein expression of exogenous MDA5, RIG-I, MAVS, TBK1 and IRF3 via the proteasomal and lysosomal pathways. Additionally, we show that µ1 protein can physically interact with MDA5, RIG-I, MAVS, TBK1, and IRF3 and attenuate the RIG-I/MDA5-mediated signaling cascades by blocking the phosphorylation and nuclear translocation of IRF3. In conclusion, our findings reveal that MRV outer capsid protein µ1 is a key factor in antagonizing RLRs signaling cascades and provide new strategies for effective prevention and treatment of MRV infection.


Subject(s)
Capsid Proteins , DEAD Box Protein 58 , Interferon Regulatory Factor-3 , Interferon-Induced Helicase, IFIH1 , Orthoreovirus, Mammalian , Receptors, Immunologic , Signal Transduction , Animals , Humans , Active Transport, Cell Nucleus , Cell Nucleus/metabolism , DEAD Box Protein 58/metabolism , HEK293 Cells , Immunity, Innate/immunology , Interferon Regulatory Factor-3/metabolism , Interferon-beta/metabolism , Interferon-beta/immunology , Interferon-Induced Helicase, IFIH1/metabolism , Interferon-Induced Helicase, IFIH1/genetics , Orthoreovirus, Mammalian/immunology , Orthoreovirus, Mammalian/physiology , Phosphorylation , Protein Serine-Threonine Kinases , Reoviridae Infections/immunology , Signal Transduction/immunology , Viral Proteins/metabolism , Capsid Proteins/metabolism
19.
Virol Sin ; 39(4): 587-599, 2024 Aug.
Article in English | MEDLINE | ID: mdl-38823782

ABSTRACT

Herpesviruses antagonize host antiviral responses through a myriad of molecular strategies culminating in the death of the host cells. Pseudorabies virus (PRV) is a significant veterinary pathogen in pigs, causing neurological sequalae that ultimately lead to the animal's demise. PRV is known to trigger apoptotic cell death during the late stages of infection. The virion host shutdown protein (VHS) encoded by UL41 plays a crucial role in the PRV infection process. In this study, we demonstrate that UL41 inhibits PRV-induced activation of inflammatory cytokine and negatively regulates the cGAS-STING-mediated antiviral activity by targeting IRF3, thereby inhibiting the translocation and phosphorylation of IRF3. Notably, mutating the conserved amino acid sites (E192, D194, and D195) in the RNase domain of UL41 or knocking down UL41 inhibits the immune evasion of PRV, suggesting that UL41 may play a crucial role in PRV's evasion of the host immune response during infection. These results enhance our understanding of how PRV structural proteins assist the virus in evading the host immune response.


Subject(s)
Herpesvirus 1, Suid , Immune Evasion , Interferon Regulatory Factor-3 , NF-kappa B , Herpesvirus 1, Suid/immunology , Herpesvirus 1, Suid/genetics , Interferon Regulatory Factor-3/metabolism , Interferon Regulatory Factor-3/genetics , Animals , Swine , NF-kappa B/metabolism , NF-kappa B/genetics , NF-kappa B/immunology , Humans , Interferons/immunology , Interferons/metabolism , Interferons/genetics , Pseudorabies/virology , Pseudorabies/immunology , Cell Line , Host-Pathogen Interactions/immunology , Viral Proteins/genetics , Viral Proteins/metabolism , Viral Proteins/immunology , HEK293 Cells , Phosphorylation , Protein Transport
20.
Nucleic Acid Ther ; 34(3): 101-108, 2024.
Article in English | MEDLINE | ID: mdl-38530082

ABSTRACT

Long antisense RNAs (asRNAs) have been observed to repress HIV and other virus expression in a manner that is refractory to viral evolution. Severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2), the causative agent of the coronavirus disease 2019 (COVID-19) disease, has a distinct ability to evolve resistance around antibody targeting, as was evident from the emergence of various SARS-CoV-2 spike antibody variants. Importantly, the effectiveness of current antivirals is waning due to the rapid emergence of new variants of concern, more recently the omicron variant. One means of avoiding the emergence of viral resistance is by using long asRNA to target SARS-CoV-2. Similar work has proven successful with HIV targeting by long asRNA. In this study, we describe a long asRNA targeting SARS-CoV-2 RNA-dependent RNA polymerase gene and the ability to deliver this RNA in extracellular vesicles (EVs) to repress virus expression. The observations presented in this study suggest that EV-delivered asRNAs are one means to targeting SARS-CoV-2 infection, which is both effective and broadly applicable as a means to control viral expression in the absence of mutation. This is the first demonstration of the use of engineered EVs to deliver long asRNA payloads for antiviral therapy.


Subject(s)
COVID-19 , Extracellular Vesicles , RNA, Antisense , SARS-CoV-2 , Extracellular Vesicles/genetics , Extracellular Vesicles/virology , Extracellular Vesicles/metabolism , SARS-CoV-2/genetics , SARS-CoV-2/drug effects , Humans , RNA, Antisense/genetics , RNA, Antisense/therapeutic use , COVID-19/virology , COVID-19/genetics , COVID-19/therapy , Animals , RNA-Dependent RNA Polymerase/genetics , RNA-Dependent RNA Polymerase/antagonists & inhibitors , Vero Cells , Chlorocebus aethiops , Antiviral Agents/therapeutic use , Antiviral Agents/pharmacology , COVID-19 Drug Treatment
SELECTION OF CITATIONS
SEARCH DETAIL