Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 594
Filter
1.
J Med Virol ; 96(7): e29788, 2024 Jul.
Article in English | MEDLINE | ID: mdl-38982767

ABSTRACT

Molecular surveillance is vital for monitoring arboviruses, often employing genus-specific quantitative reverse-transcription polymerase chain reaction (RT-qPCR). Despite this, an overlooked chikungunya fever outbreak occurred in Yunnan province, China, in 2019 and false negatives are commonly encountered during alphaviruses screening practice, highlighting the need for improved detection methods. In this study, we developed an improved alphaviruses-specific RT-qPCR capable of detecting chikungunya virus, eastern equine encephalitis virus, western equine encephalitis virus, Venezuelan equine encephalitis virus, Sindbis virus, Mayaro virus, and Ross River virus with high sensitivity and specificity. The assay identified three chikungunya virus-positive cases out of 188 sera retrospectively. Later genetic characterization suggested that imported cases from neighboring countries may be responsible for the neglected chikungunya fever outbreak of 2019 in Yunnan. Our findings underscore the value of improved alphaviruses-specific RT-qPCR in bolstering alphaviruses surveillance and informing preventive strategies.


Subject(s)
Alphavirus Infections , Alphavirus , Chikungunya virus , Real-Time Polymerase Chain Reaction , Sensitivity and Specificity , Humans , Alphavirus/genetics , Alphavirus/isolation & purification , Alphavirus Infections/diagnosis , Alphavirus Infections/virology , Alphavirus Infections/prevention & control , Alphavirus Infections/epidemiology , China/epidemiology , Real-Time Polymerase Chain Reaction/methods , Chikungunya virus/genetics , Chikungunya virus/isolation & purification , Retrospective Studies , Chikungunya Fever/diagnosis , Chikungunya Fever/prevention & control , Chikungunya Fever/virology , Chikungunya Fever/epidemiology , Encephalitis Virus, Eastern Equine/genetics , Disease Outbreaks/prevention & control , Sindbis Virus/genetics , Encephalitis Virus, Western Equine/genetics , Ross River virus/genetics , Ross River virus/isolation & purification , Encephalitis Virus, Venezuelan Equine/genetics , Reverse Transcriptase Polymerase Chain Reaction/methods , RNA, Viral/genetics
2.
J Gen Virol ; 105(7)2024 Jul.
Article in English | MEDLINE | ID: mdl-38995674

ABSTRACT

Mayaro virus (MAYV), a mosquito-borne alphavirus, is considered an emerging threat to public health with epidemic potential. Phylogenetic studies show the existence of three MAYV genotypes. In this study, we provide a preliminary analysis of the pathogenesis of all three MAYV genotypes in cynomolgus macaques (Macaca facicularis, Mauritian origin). Significant MAYV-specific RNAemia and viremia were detected during acute infection in animals challenged intravenously with the three MAYV genotypes, and strong neutralizing antibody responses were observed. MAYV RNA was detected at high levels in lymphoid tissues, joint muscle and synovia over 1 month after infection, suggesting that this model could serve as a promising tool in studying MAYV-induced chronic arthralgia, which can persist for years. Significant leucopenia was observed across all MAYV genotypes, peaking with RNAemia. Notable differences in the severity of acute RNAemia and composition of cytokine responses were observed among the three MAYV genotypes. Our model showed no outward signs of clinical disease, but several major endpoints for future MAYV pathology and intervention studies are described. Disruptions to normal blood cell counts and cytokine responses were markedly distinct from those observed in macaque models of CHIKV infection, underlining the importance of developing non-human primate models specific to MAYV infection.


Subject(s)
Alphavirus Infections , Alphavirus , Genotype , Macaca fascicularis , RNA, Viral , Viremia , Animals , Macaca fascicularis/virology , Alphavirus/genetics , Alphavirus/pathogenicity , Alphavirus/classification , Alphavirus/isolation & purification , Alphavirus Infections/virology , Alphavirus Infections/veterinary , Viremia/virology , RNA, Viral/genetics , Antibodies, Viral/blood , Antibodies, Neutralizing/blood , Disease Models, Animal , Phylogeny , Cytokines/genetics , Cytokines/blood
3.
Nat Commun ; 15(1): 4906, 2024 Jun 08.
Article in English | MEDLINE | ID: mdl-38851803

ABSTRACT

Various low-density lipoprotein receptors (LPRs) have been identified as entry factors for alphaviruses, and structures of the corresponding virion-receptor complexes have been determined. Here, we analyze the similarities and differences in the receptor binding modes of multiple alphaviruses to understand their ability to infect a wide range of hosts. We further discuss the challenges associated with the development of broad-spectrum treatment strategies against a diverse range of alphaviruses.


Subject(s)
Alphavirus , Antiviral Agents , Receptors, LDL , Virus Internalization , Animals , Humans , Alphavirus/drug effects , Alphavirus/physiology , Alphavirus/genetics , Alphavirus Infections/drug therapy , Alphavirus Infections/virology , Antiviral Agents/therapeutic use , Antiviral Agents/pharmacology , Protein Binding , Receptors, LDL/metabolism , Receptors, LDL/genetics , Receptors, Virus/metabolism , Receptors, Virus/chemistry , Virion/metabolism , Virus Internalization/drug effects
4.
Viruses ; 16(6)2024 May 30.
Article in English | MEDLINE | ID: mdl-38932175

ABSTRACT

Newly emerging viruses, primarily zoonotic or vector-borne, pose a persistent threat to public health and have led to outbreaks of global concern [...].


Subject(s)
Alphavirus Infections , Alphavirus , Flavivirus Infections , Flavivirus , Alphavirus/physiology , Alphavirus/genetics , Humans , Animals , Flavivirus/genetics , Flavivirus/physiology , Alphavirus Infections/virology , Alphavirus Infections/epidemiology , Flavivirus Infections/virology , Flavivirus Infections/epidemiology
5.
Viruses ; 16(5)2024 05 20.
Article in English | MEDLINE | ID: mdl-38793690

ABSTRACT

The Mayaro virus (MAYV) is an arbovirus with emerging potential, though with a limited understanding of its epidemiology and evolution due to the lack of studies and surveillance. Here, we investigated 71 MAYV genome sequences from the Americas available at GenBank and characterized the phylogenetic relationship among virus strains. A phylogenetic analysis showed that sequences were grouped according to the genotypes L, D, and N. Genotype D sequences were closely related to sequences collected in adjacent years and from their respective countries, suggesting that isolates may have originated from circulating lineages. The coalescent analysis demonstrated similar results, indicating the continuous circulation of the virus between countries as well. An unidentified sequence from the USA was grouped with genotype D, suggesting the insertion of this genotype in the country. Furthermore, the recombination analysis detected homologous and three heterologous hybrids which presented an insertion into the nsP3 protein. Amino acid substitutions among sequences indicated selective pressure sites, suggesting viral adaptability. This also impacted the binding affinity between the E1-E2 protein complex and the Mxra8 receptor, associated with MAYV entry into human cells. These results provide information for a better understanding of genotypes circulating in the Americas.


Subject(s)
Evolution, Molecular , Genetic Variation , Genome, Viral , Genotype , Phylogeny , Americas/epidemiology , Humans , Alphavirus/genetics , Alphavirus/classification , Alphavirus/isolation & purification , Animals , Recombination, Genetic , Alphavirus Infections/virology , Alphavirus Infections/epidemiology
6.
Viruses ; 16(4)2024 03 25.
Article in English | MEDLINE | ID: mdl-38675846

ABSTRACT

Replicating RNA, including self-amplifying RNA (saRNA) and trans-amplifying RNA (taRNA), holds great potential for advancing the next generation of RNA-based vaccines. Unlike in vitro transcribed mRNA found in most current RNA vaccines, saRNA or taRNA can be massively replicated within cells in the presence of RNA-amplifying enzymes known as replicases. We recently demonstrated that this property could enhance immune responses with minimal injected RNA amounts. In saRNA-based vaccines, replicase and antigens are encoded on the same mRNA molecule, resulting in very long RNA sequences, which poses significant challenges in production, delivery, and stability. In taRNA-based vaccines, these challenges can be overcome by splitting the replication system into two parts: one that encodes replicase and the other that encodes a short antigen-encoding RNA called transreplicon. Here, we review the identification and use of transreplicon RNA in alphavirus research, with a focus on the development of novel taRNA technology as a state-of-the art vaccine platform. Additionally, we discuss remaining challenges essential to the clinical application and highlight the potential benefits related to the unique properties of this future vaccine platform.


Subject(s)
Alphavirus , RNA, Viral , Vaccine Development , Animals , Humans , Alphavirus/genetics , Alphavirus/immunology , Alphavirus Infections/immunology , Alphavirus Infections/virology , RNA, Viral/genetics , Viral Vaccines/immunology , Viral Vaccines/genetics , Virus Replication
8.
Int J Biol Macromol ; 265(Pt 1): 130847, 2024 Apr.
Article in English | MEDLINE | ID: mdl-38490381

ABSTRACT

Getah virus (GETV) belongs to the Alphavirus genus in the Togaviridae family and is a zoonotic arbovirus causing disease in both humans and animals. The capsid protein (CP) of GETV regulates the viral core assembly, but the mechanism underlying this process is poorly understood. In this study, we demonstrate that CP undergoes liquid-liquid phase separation (LLPS) with the GETV genome RNA (gRNA) in vitro and forms cytoplasmic puncta in cells. Two regions of GETV gRNA (nucleotides 1-4000 and 5000-8000) enhance CP droplet formation in vitro and the lysine-rich Link region of CP is essential for its phase separation. CP(K/R) mutant with all lysines in the Link region replaced by arginines exhibits improved LLPS versus wild type (WT) CP, but CP(K/E) mutant with lysines substituted by glutamic acids virtually loses condensation ability. Consistently, recombinant virus mutant with CP(K/R) possesses significantly higher gRNA binding affinity, virion assembly efficiency and infectivity than the virus with WT-CP. Overall, our findings provide new insights into the understanding of GETV assembly and development of new antiviral drugs against alphaviruses.


Subject(s)
Alphavirus , Animals , Humans , Alphavirus/genetics , Alphavirus/metabolism , Capsid Proteins/genetics , Capsid Proteins/metabolism , RNA, Viral/genetics , RNA, Guide, CRISPR-Cas Systems , Genomics , Virion/genetics
9.
Parasit Vectors ; 17(1): 73, 2024 Feb 19.
Article in English | MEDLINE | ID: mdl-38374048

ABSTRACT

BACKGROUND: Increasing global temperatures and unpredictable climatic extremes have contributed to the spread of vector-borne diseases. The mosquito Aedes aegypti is the main vector of multiple arboviruses that negatively impact human health, mostly in low socioeconomic areas of the world. Co-circulation and co-infection of these viruses in humans have been increasingly reported; however, how vectors contribute to this alarming trend remains unclear. METHODS: Here, we examine single and co-infection of Mayaro virus (D strain, Alphavirus) and dengue virus (serotype 2, Flavivirus) in Ae. aegypti adults and cell lines at two constant temperatures, moderate (27 °C) and hot (32 °C), to quantify vector competence and the effect of temperature on infection, dissemination and transmission, including on the degree of interaction between the two viruses. RESULTS: Both viruses were primarily affected by temperature but there was a partial interaction with co-infection. Dengue virus quickly replicates in adult mosquitoes with a tendency for higher titers in co-infected mosquitoes at both temperatures, and mosquito mortality was more severe at higher temperatures in all conditions. For dengue, and to a lesser extent Mayaro, vector competence and vectorial capacity were higher at hotter temperature in co- vs. single infections and was more evident at earlier time points (7 vs. 14 days post infection) for Mayaro. The temperature-dependent phenotype was confirmed in vitro by faster cellular infection and initial replication at higher temperatures for dengue but not for Mayaro virus. CONCLUSIONS: Our study suggests that contrasting kinetics of the two viruses could be related to their intrinsic thermal requirements, where alphaviruses thrive better at lower temperatures compared to flaviviruses. However, more studies are necessary to clarify the role of co-infection at different temperature regimes, including under more natural temperature settings.


Subject(s)
Aedes , Alphavirus , Coinfection , Dengue Virus , Dengue , Flavivirus , Animals , Humans , Temperature , Mosquito Vectors , Alphavirus/genetics , Flavivirus/genetics
10.
Nat Commun ; 15(1): 246, 2024 Jan 04.
Article in English | MEDLINE | ID: mdl-38172096

ABSTRACT

Members of the low-density lipoprotein receptor (LDLR) family, including LDLRAD3, VLDLR, and ApoER2, were recently described as entry factors for different alphaviruses. However, based on studies with gene edited cells and knockout mice, blockade or abrogation of these receptors does not fully inhibit alphavirus infection, indicating the existence of additional uncharacterized entry factors. Here, we perform a CRISPR-Cas9 genome-wide loss-of-function screen in mouse neuronal cells with a chimeric alphavirus expressing the Eastern equine encephalitis virus (EEEV) structural proteins and identify LDLR as a candidate receptor. Expression of LDLR on the surface of neuronal or non-neuronal cells facilitates binding and infection of EEEV, Western equine encephalitis virus, and Semliki Forest virus. Domain mapping and binding studies reveal a low-affinity interaction with LA domain 3 (LA3) that can be enhanced by concatenation of LA3 repeats. Soluble decoy proteins with multiple LA3 repeats inhibit EEEV infection in cell culture and in mice. Our results establish LDLR as a low-affinity receptor for multiple alphaviruses and highlight a possible path for developing inhibitors that could mitigate infection and disease.


Subject(s)
Alphavirus Infections , Alphavirus , Encephalitis Virus, Eastern Equine , Horses , Animals , Mice , Alphavirus/genetics , Encephalitis Virus, Eastern Equine/genetics , Semliki forest virus/genetics , Lipoproteins, LDL
11.
Virology ; 591: 109990, 2024 03.
Article in English | MEDLINE | ID: mdl-38224661

ABSTRACT

Getah virus (GETV) is an emerging mosquito-borne alphavirus that can infect horses, pigs and other animals. Given the public health threat posed by GETV, research on its pathogenesis, diagnosis and prevention is urgently needed. In the current study, prokaryotic expression systems were used to express the capsid protein of GETV. This protein was then used to immunize BALB/c mice in order to generate monoclonal antibodies (mAbs). Subsequently, hybridoma cells secreting a mAb (2B11-4) against the capsid protein were obtained using the hybridoma technique. A B cell linear epitope, 18-PAYRPWR-24, located at the capsid protein's N-terminal region was identified using western blotting analysis with the produced mAb, 2B11-4. Sequence alignment indicated that this epitope was highly conserved in group III (GIII) strains of GETV, but varied among the other genotypes. Western blotting showed that mAb 2B11-4 could discriminate Group III GETVs from other genotypes. This study describes the preparation of a mAb against the GETV capsid protein and the identification of the specific localization of B-cell epitopes, and will contribute towards a better understanding of the biological importance of the GETV capsid protein. It will also pave the way for developing immunological detection methods and genotype diagnosis for GETVs.


Subject(s)
Alphavirus , Culicidae , Mice , Animals , Swine , Horses , Alphavirus/genetics , Capsid Proteins/genetics , Antibodies, Monoclonal , Epitopes, B-Lymphocyte/genetics
12.
Nat Commun ; 15(1): 622, 2024 Jan 20.
Article in English | MEDLINE | ID: mdl-38245515

ABSTRACT

Alphaviruses are arboviruses transmitted by mosquitoes and are pathogenic to humans and livestock, causing a substantial public health burden. So far, several receptors have been identified for alphavirus entry; however, they cannot explain the broad host range and tissue tropism of certain alphaviruses, such as Getah virus (GETV), indicating the existence of additional receptors. Here we identify the evolutionarily conserved low-density lipoprotein receptor (LDLR) as a new cell entry factor for GETV, Semliki Forest virus (SFV), Ross River virus (RRV) and Bebaru virus (BEBV). Ectopic expression of LDLR facilitates cellular binding and internalization of GETV, which is mediated by the interaction between the E2-E1 spike of GETV and the ligand-binding domain (LBD) of LDLR. Antibodies against LBD block GETV infection in cultured cells. In addition, the GST-LBD fusion protein inhibits GETV infection both in vitro and in vivo. Notably, we identify the key amino acids in LDLR-LBD that played a crucial role in viral entry; specific mutations in the CR4 and CR5 domain of LDLR-LBD reduce viral entry to cells by more than 20-fold. These findings suggest that targeting the LDLR-LBD could be a potential strategy for the development of antivirals against multiple alphaviruses.


Subject(s)
Alphavirus Infections , Alphavirus , Culicidae , Animals , Humans , Alphavirus/genetics , Virus Internalization , Semliki forest virus/genetics , Semliki forest virus/metabolism , Alphavirus Infections/genetics
13.
Viruses ; 15(12)2023 12 17.
Article in English | MEDLINE | ID: mdl-38140691

ABSTRACT

Salmonid alphavirus strain 3 is responsible for outbreaks of pancreas disease in salmon and rainbow trout in Norway. Although the extensive amount of research on SAV3 focused mainly on the heart and pancreas (of clinical importance), tropism and pathogenesis studies of the virus in other salmon tissues are limited. Here, we used a combination of RT-qPCR (Q_nsp1 gene) and in situ hybridization (RNAscope®) to demonstrate the tropism of SAV3 in situ in tissues of Atlantic salmon, employing a challenge model (by cohabitation). In addition, as previous results suggested that the pseudobranch may harbor the virus, the change in the expression of different immune genes upon SAV3 infection (RT-qPCR) was focused on the pseudobranch in this study. In situ hybridization detected SAV3 in different tissues of Atlantic salmon during the acute phase of the infection, with the heart ventricle showing the most extensive infection. Furthermore, the detection of the virus in different adipose tissues associated with the internal organs of the salmon suggests a specific affinity of SAV3 to adipocyte components. The inconsistent immune response to SAV3 in the pseudobranch after infection did not mitigate the infection in that tissue and is probably responsible for the persistent low infection at 4 weeks post-challenge. The early detection of SAV3 in the pseudobranch after infection, along with the persistent low infection over the experimental infection course, suggests a pivotal role of the pseudobranch in SAV3 pathogenesis in Atlantic salmon.


Subject(s)
Alphavirus Infections , Alphavirus , Fish Diseases , Salmo salar , Animals , Alphavirus/genetics , Heart
14.
Int J Mol Sci ; 24(21)2023 Nov 03.
Article in English | MEDLINE | ID: mdl-37958918

ABSTRACT

Infection by arthritogenic alphaviruses (aavs) can lead to reactive arthritis, which is characterized by inflammation and persistence of the virus; however, its mechanisms remain ill-characterized. Intriguingly, it has been shown that viral persistence still takes place in spite of robust innate and adaptive immune responses, characterized notably by the infiltration of macrophages (sources of TNF-alpha) as well as T/NK cells (sources of IFN-gamma) in the infected joint. Aavs are known to target mesenchymal stem cells (MSCs) in the synovium, and we herein tested the hypothesis that the infection of MSCs may promote the expression of immunoregulators to skew the anti-viral cellular immune responses. We compared the regulated expression via human synovial MSCs of pro-inflammatory mediators (e.g., IL-1ß, IL6, CCL2, miR-221-3p) to that of immunoregulators (e.g., IDO, TSG6, GAS6, miR146a-5p). We used human synovial tissue-derived MSCs which were infected with O'Nyong-Nyong alphavirus (ONNV, class II aav) alone, or combined with recombinant human TNF-α or IFN-γ, to mimic the clinical settings. We confirmed via qPCR and immunofluorescence that ONNV infected human synovial tissue-derived MSCs. Interestingly, ONNV alone did not regulate the expression of pro-inflammatory mediators. In contrast, IDO, TSG6, and GAS6 mRNA expression were increased in response to ONNV infection alone, but particularly when combined with both recombinant cytokines. ONNV infection equally decreased miR-146a-5p and miR-221-3p in the untreated cells and abrogated the stimulatory activity of the recombinant TNF-α but not the IFN-gamma. Our study argues for a major immunoregulatory phenotype of MSCs infected with ONNV which may favor virus persistence in the inflamed joint.


Subject(s)
Alphavirus , Arthritis, Infectious , Mesenchymal Stem Cells , MicroRNAs , Humans , Alphavirus/genetics , Alphavirus/metabolism , Immunity , Inflammation Mediators , Mesenchymal Stem Cells/metabolism , MicroRNAs/genetics , MicroRNAs/metabolism , Tumor Necrosis Factor-alpha
15.
J Clin Microbiol ; 61(12): e0015223, 2023 12 19.
Article in English | MEDLINE | ID: mdl-37982611

ABSTRACT

Eastern equine encephalitis virus (EEEV), Madariaga virus (MADV), and Venezuelan equine encephalitis virus complex (VEEV) are New World alphaviruses transmitted by mosquitoes. They cause febrile and sometimes severe neurological diseases in human and equine hosts. Detecting them during the acute phase is hindered by non-specific symptoms and limited diagnostic tools. We designed and clinically assessed real-time reverse transcription polymerase chain reaction assays (rRT-PCRs) for VEEV complex, MADV, and EEEV using whole-genome sequences. Validation involved 15 retrospective serum samples from 2015 to 2017 outbreaks, 150 mosquito pools from 2015, and 118 prospective samples from 2021 to 2022 surveillance in Panama. The rRT-PCRs detected VEEV complex RNA in 10 samples (66.7%) from outbreaks, with one having both VEEV complex and MADV RNAs. VEEV complex RNA was found in five suspected dengue cases from disease surveillance. The rRT-PCR assays identified VEEV complex RNA in three Culex (Melanoconion) vomerifer pools, leading to VEEV isolates in two. Phylogenetic analysis revealed the VEEV ID subtype in positive samples. Notably, 11.9% of dengue-like disease patients showed VEEV infections. Together, our rRT-PCR validation in human and mosquito samples suggests that this method can be incorporated into mosquito and human encephalitic alphavirus surveillance programs in endemic regions.


Subject(s)
Alphavirus , Culicidae , Dengue , Encephalitis Virus, Eastern Equine , Encephalomyelitis, Eastern Equine , Encephalomyelitis, Venezuelan Equine , Humans , Animals , Horses/genetics , Encephalitis Virus, Eastern Equine/genetics , Encephalomyelitis, Venezuelan Equine/diagnosis , Encephalomyelitis, Venezuelan Equine/epidemiology , Culicidae/genetics , Reverse Transcriptase Polymerase Chain Reaction , Phylogeny , Prospective Studies , Public Health Surveillance , Retrospective Studies , Alphavirus/genetics , RNA
16.
Viruses ; 15(10)2023 10 14.
Article in English | MEDLINE | ID: mdl-37896867

ABSTRACT

As an epizootic causative agent, the Getah virus (GETV) can cause moderate illness in horses, lethal disease in foxes, and reproductive disorders and fetal death in pigs. Due to the wide range of hosts and multiple routes of transmission, GETV has become a growing potential threat to the global livestock industry, and even to public health. More attention and research on GETV are urgently needed. In this study, we successfully isolated a novel GETV strain, named BJ0304, from a commercial live vaccine against porcine reproductive and respiratory syndrome virus (PRRSV) and determined its growth kinetics. Then, genetic and phylogenetic analyses were performed. The results revealed that BJ0304 was clustered into Group III, and it was most related to the GETV-V1 strain based on the complete genome sequence. Furthermore, the pathogenicity of the isolate was assessed and found to be a low virulent strain in mice relative to its closest homolog GETV-V1. Finally, mutation and glycosylation analysis showed that a unique mutation (171 T > I) at one amino acid of E2, which affected the glycosylation of E2, may be associated with viral pathogenicity. In summary, the general characteristic of a novel Group III-classified GETV-BJ0304 isolated from commercial live PRRSV vaccine was defined and then mutation/glycosylation-related potential virulence factor was discussed. This study highlights the complexity of GETV transmission routes in swine and the need for more surveillance on commercial animal vaccines, contributes to the understanding of genetic characterization of clinical isolates, provides possible virulence factors in favor of unveiling the viral pathogenesis, and eventually lays the foundation for the prevention and control of GETV.


Subject(s)
Alphavirus , Porcine Reproductive and Respiratory Syndrome , Porcine respiratory and reproductive syndrome virus , Vaccines , Animals , Swine , Horses , Mice , Porcine respiratory and reproductive syndrome virus/genetics , Alphavirus/genetics , Phylogeny , Porcine Reproductive and Respiratory Syndrome/prevention & control
17.
Int J Mol Sci ; 24(19)2023 Oct 06.
Article in English | MEDLINE | ID: mdl-37834397

ABSTRACT

Sindbis alphavirus vectors offer a promising platform for cancer therapy, serving as valuable models for alphavirus-based treatment. This review emphasizes key studies that support the targeted delivery of Sindbis vectors to tumor cells, highlighting their effectiveness in expressing tumor-associated antigens and immunomodulating proteins. Among the various alphavirus vectors developed for cancer therapy, Sindbis-vector-based imaging studies have been particularly extensive. Imaging modalities that enable the in vivo localization of Sindbis vectors within lymph nodes and tumors are discussed. The correlation between laminin receptor expression, tumorigenesis, and Sindbis virus infection is examined. Additionally, we present alternative entry receptors for Sindbis and related alphaviruses, such as Semliki Forest virus and Venezuelan equine encephalitis virus. The review also discusses cancer treatments that are based on the alphavirus vector expression of anti-tumor agents, including tumor-associated antigens, cytokines, checkpoint inhibitors, and costimulatory immune molecules.


Subject(s)
Alphavirus , Encephalitis Virus, Venezuelan Equine , Neoplasms , Humans , Alphavirus/genetics , Genetic Vectors/genetics , Neoplasms/therapy , Genetic Therapy/methods
18.
J Virol ; 97(11): e0122523, 2023 Nov 30.
Article in English | MEDLINE | ID: mdl-37877718

ABSTRACT

IMPORTANCE: Alphavirus replicons are being developed as self-amplifying RNAs aimed at improving the efficacy of mRNA vaccines. These replicons are convenient for genetic manipulations and can express heterologous genetic information more efficiently and for a longer time than standard mRNAs. However, replicons mimic many aspects of viral replication in terms of induction of innate immune response, modification of cellular transcription and translation, and expression of nonstructural viral genes. Moreover, all replicons used in this study demonstrated expression of heterologous genes in cell- and replicon's origin-specific modes. Thus, many aspects of the interactions between replicons and the host remain insufficiently investigated, and further studies are needed to understand the biology of the replicons and their applicability for designing a new generation of mRNA vaccines. On the other hand, our data show that replicons are very flexible expression systems, and additional modifications may have strong positive impacts on protein expression.


Subject(s)
Alphavirus , Gene Expression Regulation, Viral , Host Microbial Interactions , Replicon , Viral Proteins , Alphavirus/genetics , Alphavirus/metabolism , mRNA Vaccines/genetics , Replicon/genetics , Virus Replication/genetics , RNA, Viral/biosynthesis , RNA, Viral/genetics , Host Microbial Interactions/genetics , Viral Proteins/biosynthesis , Viral Proteins/genetics
19.
Int Rev Cell Mol Biol ; 379: 143-168, 2023.
Article in English | MEDLINE | ID: mdl-37541722

ABSTRACT

Alphaviruses have frequently been engineered for cancer therapy, cancer immunotherapy, and cancer vaccine development. As members of self-replicating RNA viruses, alphaviruses provide high levels of transgene expression through efficient self-amplifying of their RNA genome in host cells. Alphavirus vectors can be used as recombinant viral particles or oncolytic viruses. Alternatively, either naked or nanoparticle-encapsulated RNA and DNA replicons can be utilized. In the context of cancer prevention and treatment, antitumor, cytotoxic and suicide genes have been expressed from alphavirus vectors to provide tumor regression and tumor eradication. Moreover, immunostimulatory genes such as cytokines and chemokines have been used for cancer immunotherapy approaches. Expression of tumor antigens has been applied for cancer vaccine development. Alphavirus vectors has demonstrated tumor regression and even cure in various preclinical animal models. Immunization has elicited strong immune responses and showed protection against challenges with tumor cells in animal models. Several clinical trials have confirmed good safety and tolerability of alphaviruses in cancer patients although therapeutic efficacy will still require optimization.


Subject(s)
Alphavirus , Cancer Vaccines , Neoplasms , Animals , Alphavirus/genetics , Genetic Vectors , Immunotherapy , RNA , Neoplasms/therapy
SELECTION OF CITATIONS
SEARCH DETAIL