Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 13 de 13
Filter
Add more filters










Publication year range
1.
Nat Commun ; 15(1): 3747, 2024 May 03.
Article in English | MEDLINE | ID: mdl-38702310

ABSTRACT

In malaria parasites, the regulation of mRNA translation, storage and degradation during development and life-stage transitions remains largely unknown. Here, we functionally characterized the DEAD-box RNA helicase PfDOZI in P. falciparum. Disruption of pfdozi enhanced asexual proliferation but reduced sexual commitment and impaired gametocyte development. By quantitative transcriptomics, we show that PfDOZI is involved in the regulation of invasion-related genes and sexual stage-specific genes during different developmental stages. PfDOZI predominantly participates in processing body-like mRNPs in schizonts but germ cell granule-like mRNPs in gametocytes to impose opposing actions of degradation and protection on different mRNA targets. We further show the formation of stress granule-like mRNPs during nutritional deprivation, highlighting an essential role of PfDOZI-associated mRNPs in stress response. We demonstrate that PfDOZI participates in distinct mRNPs to maintain mRNA homeostasis in response to life-stage transition and environmental changes by differentially executing post-transcriptional regulation on the target mRNAs.


Subject(s)
DEAD-box RNA Helicases , Plasmodium falciparum , Protozoan Proteins , RNA, Messenger , DEAD-box RNA Helicases/metabolism , DEAD-box RNA Helicases/genetics , Plasmodium falciparum/genetics , Plasmodium falciparum/metabolism , Plasmodium falciparum/growth & development , RNA, Messenger/metabolism , RNA, Messenger/genetics , Protozoan Proteins/metabolism , Protozoan Proteins/genetics , Ribonucleoproteins/metabolism , Ribonucleoproteins/genetics , Life Cycle Stages/genetics , RNA, Protozoan/metabolism , RNA, Protozoan/genetics , RNA Stability , Humans , Malaria, Falciparum/parasitology
2.
Nat Commun ; 14(1): 8302, 2023 Dec 14.
Article in English | MEDLINE | ID: mdl-38097652

ABSTRACT

The proteasome of the malaria parasite Plasmodium falciparum (Pf20S) is an advantageous drug target because its inhibition kills P. falciparum in multiple stages of its life cycle and synergizes with artemisinins. We recently developed a macrocyclic peptide, TDI-8304, that is highly selective for Pf20S over human proteasomes and is potent in vitro and in vivo against P. falciparum. A mutation in the Pf20S ß6 subunit, A117D, confers resistance to TDI-8304, yet enhances both enzyme inhibition and anti-parasite activity of a tripeptide vinyl sulfone ß2 inhibitor, WLW-vs. Here we present the high-resolution cryo-EM structures of Pf20S with TDI-8304, of human constitutive proteasome with TDI-8304, and of Pf20Sß6A117D with WLW-vs that give insights into the species selectivity of TDI-8304, resistance to it, and the collateral sensitivity associated with resistance, including that TDI-8304 binds ß2 and ß5 in wild type Pf20S as well as WLW-vs binds ß2 and ß5 in Pf20Sß6A117D. We further show that TDI-8304 kills P. falciparum as quickly as chloroquine and artemisinin and is active against P. cynomolgi at the liver stage. This increases interest in using these structures to facilitate the development of Pf20S inhibitors that target multiple proteasome subunits and limit the emergence of resistance.


Subject(s)
Antimalarials , Malaria, Falciparum , Humans , Plasmodium falciparum/genetics , Proteasome Inhibitors/pharmacology , Proteasome Inhibitors/chemistry , Proteasome Endopeptidase Complex/metabolism , Drug Collateral Sensitivity , Malaria, Falciparum/drug therapy , Malaria, Falciparum/parasitology , Antimalarials/pharmacology , Antimalarials/chemistry , Drug Resistance/genetics , Protozoan Proteins/genetics
3.
Cell Chem Biol ; 30(5): 457-469.e11, 2023 05 18.
Article in English | MEDLINE | ID: mdl-37148884

ABSTRACT

Artemisinins (ART) are critical anti-malarials and despite their use in combination therapy, ART-resistant Plasmodium falciparum is spreading globally. To counter ART resistance, we designed artezomibs (ATZs), molecules that link an ART with a proteasome inhibitor (PI) via a non-labile amide bond and hijack parasite's own ubiquitin-proteasome system to create novel anti-malarials in situ. Upon activation of the ART moiety, ATZs covalently attach to and damage multiple parasite proteins, marking them for proteasomal degradation. When damaged proteins enter the proteasome, their attached PIs inhibit protease function, potentiating the parasiticidal action of ART and overcoming ART resistance. Binding of the PI moiety to the proteasome active site is enhanced by distal interactions of the extended attached peptides, providing a mechanism to overcome PI resistance. ATZs have an extra mode of action beyond that of each component, thereby overcoming resistance to both components, while avoiding transient monotherapy seen when individual agents have disparate pharmacokinetic profiles.


Subject(s)
Antimalarials , Artemisinins , Parasites , Plasmodium , Animals , Antimalarials/chemistry , Proteasome Endopeptidase Complex/metabolism , Parasites/metabolism , Pharmacophore , Ubiquitin , Plasmodium/metabolism , Artemisinins/pharmacology , Drug Resistance
4.
Small ; 18(47): e2206468, 2022 Nov.
Article in English | MEDLINE | ID: mdl-36424186

ABSTRACT

Small 2021, 17, 2008165 DOI: 10.1002/smll.202008165 The above article in Small, published online on 26 March 2021 in Wiley Online Library (https://onlinelibrary.wiley.com/doi/abs/10.1002/smll.202008165),[1] has been retracted by agreement between the authors, the Editor-in-Chief, José Oliveira, and Wiley-VCH GmbH. The retraction has been agreed following an investigation by the corresponding author. The electrochemical measurements on the anode were performed in a wrong manner and cannot reliably be reproduced. The conclusions of this article are considered to be invalid. The authors agree with the retraction but were not available to confirm the final wording of the retraction. [1] Z. Cao, Y. Yang, J. Qin, J. He, Z. Su, Small 2021, 17, 2008165.

5.
mBio ; 13(4): e0189722, 2022 08 30.
Article in English | MEDLINE | ID: mdl-35938722

ABSTRACT

Artemisinin resistance in Plasmodium falciparum has been associated with a mutation in the NLI-interacting factor-like phosphatase PfNIF4, in addition to the mutations in the Kelch13 protein as the major determinant. We found that PfNIF4 was predominantly expressed at the schizont stage and localized in the nuclei of the parasite. To elucidate the functions of PfNIF4 in P. falciparum, we performed PfNIF4 knockdown (KD) using the inducible ribozyme system. PfNIF4 KD attenuated merozoite invasion and affected gametocytogenesis. PfNIF4 KD parasites also showed significantly increased in vitro susceptibility to artemisinins. Transcriptomic and proteomic analysis revealed that PfNIF4 KD led to the downregulation of gene categories involved in invasion and artemisinin resistance (e.g., mitochondrial function, membrane, and Kelch13 interactome) at the trophozoite and/or schizont stage. Consistent with PfNIF4 being a protein phosphatase, PfNIF4 KD resulted in an overall upregulation of the phosphoproteome of infected erythrocytes. Quantitative phosphoproteomic profiling identified a set of PfNIF4-regulated phosphoproteins with functional similarity to FCP1 substrates, particularly proteins involved in chromatin organization and transcriptional regulation. Specifically, we observed increased phosphorylation of Ser2/5 of the tandem repeats in the C-terminal domain (CTD) of RNA polymerase II (RNAPII) upon PfNIF4 KD. Furthermore, using the TurboID-based proteomic approach, we identified that PfNIF4 interacted with the RNAPII components, AP2-domain transcription factors, and chromatin-modifiers and binders. These findings suggest that PfNIF4 may act as the RNAPII CTD phosphatase, regulating the expression of general and parasite-specific cellular pathways during the blood-stage development. IMPORTANCE Protein phosphorylation regulates a multitude of cellular processes. The eukaryotic FCP1 phosphatase acts as a CTD-phosphatase to critically balance the phosphorylation status of the CTD of the RNAPII, controlling the accurate execution of the transcription process. Here, we identified PfNIF4 as the FCP1-like phosphatase in P. falciparum. PfNIF4 KD specifically downregulated genes involved in merozoite invasion, resulting in the attenuation of this process. Consistent with the earlier finding of the association of PfNIF4 mutations with artemisinin resistance in Southeast Asian parasite populations, PfNIF4 KD significantly increased in vitro susceptibility to artemisinins. The regulation of these cellular processes in P. falciparum by PfNIF4 is likely realized through RNAPII-mediated transcription, because PfNIF4 was found to interact with RNAPII subunits and KD of PfNIF4 caused CTD hyperphosphorylation. Our results reveal the functions of the PfNIF4 phosphatase in controlling the transcription of invasion- and resistance-related genes in the malaria parasite.


Subject(s)
Antimalarials , Artemisinins , Malaria, Falciparum , Animals , Antimalarials/pharmacology , Artemisinins/metabolism , Artemisinins/pharmacology , Malaria, Falciparum/parasitology , Merozoites , Phosphoprotein Phosphatases/genetics , Phosphoprotein Phosphatases/metabolism , Plasmodium falciparum/metabolism , Proteomics , Protozoan Proteins/genetics , Protozoan Proteins/metabolism , RNA Polymerase II/metabolism , Schizonts/genetics
6.
Microbiol Spectr ; 10(3): e0278221, 2022 06 29.
Article in English | MEDLINE | ID: mdl-35510853

ABSTRACT

By fusing catalytically dead Cas9 (dCas9) to active domains of histone deacetylase (Sir2a) or acetyltransferase (GCN5), this CRISPR interference/activation (CRISPRi/a) system allows gene regulation at the transcriptional level without causing permanent changes in the parasite genome. However, the constitutive expression of dCas9 poses a challenge for studying essential genes, which may lead to adaptive changes in the parasite, masking the true phenotypes. Here, we developed a leak-free inducible CRISPRi/a system by integrating the DiCre/loxP regulon to allow the expression of dCas9-GCN5/-Sir2a upon transient induction with rapamycin, which allows convenient transcriptional regulation of a gene of interest by introducing a guide RNA targeting its transcription start region. Using eight genes that are either silent or expressed from low to high levels during asexual erythrocytic development, we evaluated the robustness and versatility of this system in the asexual parasites. For most genes analyzed, this inducible CRISPRi/a system led to 1.5- to 3-fold up-or downregulation of the target genes at the mRNA level. Alteration in the expression of PfK13 and PfMYST resulted in altered sensitivities to artemisinin. For autophagy-related protein 18, an essential gene related to artemisinin resistance, a >2-fold up- or downregulation was obtained by inducible CRISPRi/a, leading to growth retardation. For the master regulator of gametocytogenesis, PfAP2-G, a >10-fold increase of the PfAP2-G transcripts was obtained by CRISPRa, resulting in >4-fold higher gametocytemia in the induced parasites. Additionally, inducible CRISPRi/a could also regulate gene expression in gametocytes. This inducible epigenetic regulation system offers a fast way of studying gene functions in Plasmodium falciparum. IMPORTANCE Understanding the fundamental biology of malaria parasites through functional genetic/genomic studies is critical for identifying novel targets for antimalarial development. Conditional knockout/knockdown systems are required to study essential genes in the haploid blood stages of the parasite. In this study, we developed an inducible CRISPRi/a system via the integration of DiCre/loxP. We evaluated the robustness and versatility of this system by activating or repressing eight selected genes and achieved up- and downregulation of the targeted genes located in both the euchromatin and heterochromatin regions. This system offers the malaria research community another tool for functional genetic studies.


Subject(s)
Antimalarials , Artemisinins , CRISPR-Cas Systems , Epigenesis, Genetic , Gene Expression Regulation , Plasmodium falciparum/genetics
7.
Small ; 17(19): e2008165, 2021 May.
Article in English | MEDLINE | ID: mdl-33768724

ABSTRACT

In this work, a novel lollipop nanostructure of Co3 O4 @MnO2 composite is prepared as anode material in lithium-ion batteries (LIBs). Cobalt metal-organic framework (ZIF-67) is grown on the open end of MnO2 nanotubes via a self-assembly process. The obtained ZIF-67@MnO2 is then converted to Co3 O4 @MnO2 by a simple annealing treatment in air. Scanning electron microscopy, transmission electron microscopy, and X-ray diffraction characterizations indicate that the prepared Co3 O4 @MnO2 takes a lollipop nanostructure with a stick of ≈100 nm in diameter, consisting of MnO2 nanotube, and a head part of ≈1 µm, consisting of Co3 O4 nanoparticles. The charge-discharge tests illustrate that this unique novel configuration endows the resulting Co3 O4 @MnO2 with excellent electrochemical performances, delivering a capacity of 1080 mAh g-1 at 300 mA g-1 after 160 cycles, and 696 mAh g-1 at 1 A g-1 after 210 cycles, compared with 404 mAh g-1 and 590 for pure Co3 O4 polyhedrons and pure MnO2 nanotubes at 300 mA g-1 after 160 cycles, respectively. The lollipop configuration consisting of porous Co3 O4 polyhedron and MnO2 nanotube shows excellent structural stability and facilitates lithium insertion/extraction, leading to excellent cyclic stability and rate capacity of Co3 O4 @MnO2 -based LIBs.

8.
J Infect Dis ; 222(9): 1561-1569, 2020 10 01.
Article in English | MEDLINE | ID: mdl-32386321

ABSTRACT

Deletion of the pfhrp2 gene in Plasmodium falciparum can lead to false-negative rapid diagnostic test (RDT) results, constituting a major challenge for evidence-based malaria treatment. Here we analyzed the whole genome sequences of 138 P. falciparum clinical samples collected from the China-Myanmar boarder for pfhrp2 and pfhrp3 gene deletions. We found pfhrp2 and pfhrp3 deletions in 9.4% and 3.6% of samples, respectively, with no samples harboring deletions of both genes. The pfhrp2 deletions showed 2 distinct breakpoints, representing 2 different chromosomal deletion events. A phylogenetic analysis performed using genome-wide single-nucleotide polymorphisms revealed that the 2 pfhrp2 breakpoint groups as well as all the pfhrp3-negative parasites formed separate clades, suggesting they might have resulted from clonal expansion of pfhrp2- and pfhrp3-negative parasites. These findings highlight the need for urgent surveys to determine the prevalence of pfhrp2-negative parasites causing false-negative RDT results and a plan for switching of RDTs pending the survey results.


Subject(s)
Antigens, Protozoan/genetics , Malaria, Falciparum/parasitology , Plasmodium falciparum/genetics , Protozoan Proteins/genetics , China/epidemiology , False Negative Reactions , Gene Deletion , Genome, Protozoan/genetics , Humans , Malaria, Falciparum/diagnosis , Malaria, Falciparum/epidemiology , Myanmar/epidemiology , Phylogeny , Polymerase Chain Reaction , Polymorphism, Single Nucleotide/genetics , Prevalence , Sequence Alignment
9.
mBio ; 11(1)2020 02 25.
Article in English | MEDLINE | ID: mdl-32098812

ABSTRACT

Mutations in the Plasmodium falciparum Kelch 13 (PfK13) protein are associated with artemisinin resistance. PfK13 is essential for asexual erythrocytic development, but its function is not known. We tagged the PfK13 protein with green fluorescent protein in P. falciparum to study its expression and localization in asexual and sexual stages. We used a new antibody against PfK13 to show that the PfK13 protein is expressed ubiquitously in both asexual erythrocytic stages and gametocytes and is localized in punctate structures, partially overlapping an endoplasmic reticulum marker. We introduced into the 3D7 strain four PfK13 mutations (F446I, N458Y, C469Y, and F495L) identified in parasites from the China-Myanmar border area and characterized the in vitro artemisinin response phenotypes of the mutants. We found that all the parasites with the introduced PfK13 mutations showed higher survival rates in the ring-stage survival assay (RSA) than the wild-type (WT) control, but only parasites with N458Y displayed a significantly higher RSA value (26.3%) than the WT control. After these PfK13 mutations were reverted back to the WT in field parasite isolates, all revertant parasites except those with the C469Y mutation showed significantly lower RSA values than their respective parental isolates. Although the 3D7 parasites with introduced F446I, the predominant PfK13 mutation in northern Myanmar, did not show significantly higher RSA values than the WT, they had prolonged ring-stage development and showed very little fitness cost in in vitro culture competition assays. In comparison, parasites with the N458Y mutations also had a prolonged ring stage and showed upregulated resistance pathways in response to artemisinin, but this mutation produced a significant fitness cost, potentially leading to their lower prevalence in the Greater Mekong subregion.IMPORTANCE Artemisinin resistance has emerged in Southeast Asia, endangering the substantial progress in malaria elimination worldwide. It is associated with mutations in the PfK13 protein, but how PfK13 mediates artemisinin resistance is not completely understood. Here we used a new antibody against PfK13 to show that the PfK13 protein is expressed in all stages of the asexual intraerythrocytic cycle as well as in gametocytes and is partially localized in the endoplasmic reticulum. By introducing four PfK13 mutations into the 3D7 strain and reverting these mutations in field parasite isolates, we determined the impacts of these mutations identified in the parasite populations from northern Myanmar on the ring stage using the in vitro ring survival assay. The introduction of the N458Y mutation into the 3D7 background significantly increased the survival rates of the ring-stage parasites but at the cost of the reduced fitness of the parasites. Introduction of the F446I mutation, the most prevalent PfK13 mutation in northern Myanmar, did not result in a significant increase in ring-stage survival after exposure to dihydroartemisinin (DHA), but these parasites showed extended ring-stage development. Further, parasites with the F446I mutation showed only a marginal loss of fitness, partially explaining its high frequency in northern Myanmar. Conversely, reverting all these mutations, except for the C469Y mutation, back to their respective wild types reduced the ring-stage survival of these isolates in response to in vitro DHA treatment.


Subject(s)
Antimalarials/pharmacology , Artemisinins/pharmacology , Drug Resistance/genetics , Plasmodium falciparum/drug effects , Plasmodium falciparum/genetics , Protozoan Proteins/genetics , Asia, Southeastern , Drug Resistance/drug effects , Humans , Malaria, Falciparum/parasitology , Mutation , Organisms, Genetically Modified
10.
PLoS One ; 10(9): e0138184, 2015.
Article in English | MEDLINE | ID: mdl-26372641

ABSTRACT

OBJECTIVE: Adipose tissue derived stem cells (ADSCs) transplantation has recently gained widespread enthusiasm, particularly in the perspective to use them as potential alternative cell sources for hepatocytes in cell based therapy, mainly because of their capability of hepatogenic differentiation in vitro and in vivo. But some challenges remain to be addressed, including whether ADSCs can be provided effectively to the target organ and whether subsequent proliferation of transplanted cells can be achieved. To date, intrasplenic injection is the conventional method to deliver ADSCs into the liver; however, a number of donor cells retained in the spleen has been reported. In this study, our objective is to evaluate a novel route to transplant ADSCs specifically to the liver. We aimed to test the feasibility of in situ transplantation of ADSCs by injecting bioencapsulated ADSCs into the liver in mouse model. METHODS: The ADSCs isolated from human alpha 1 antitrypsin (M-hAAT) transgenic mice were used to allow delivered ADSCs be readily identified in the liver of recipient mice, and alginate was selected as a cell carrier. We first evaluated whether alginate microspheres are implantable into the liver tissue by injection and whether ADSCs could migrate from alginate microspheres (study one). Once proven, we then examined the in vivo fate of ADSCs loaded microspheres in the liver. Specifically, we evaluated whether transplanted, undifferentiated ASDCs could be induced by the local microenvironment toward hepatogenic differentiation and the distribution of surviving ADSCs in major tissue organs (study two). RESULTS: Our results indicated ADSCs loaded alginate microspheres were implantable into the liver. Both degraded and residual alginate microspheres were observed in the liver up to three weeks. The viable ADSCs were detectable surrounding degraded and residual alginate microspheres in the liver and other major organs such as bone marrow and the lungs. Importantly, transplanted ADSCs underwent hepatogenic differentiation to become cells expressing albumin in the liver. These findings improve our understanding of the interplay between ADSCs (donor cells), alginate (biomaterial), and local microenvironment in a hepatectomized mouse model, and might improve the strategy of in situ transplantation of ADSCs in treating liver diseases.


Subject(s)
Adipose Tissue/cytology , Alginates/chemistry , Liver/cytology , Stem Cell Transplantation/methods , Stem Cells/chemistry , Stem Cells/cytology , Animals , Capsules , Cell Differentiation , Feasibility Studies , Glucuronic Acid/chemistry , Hepatocytes/cytology , Hexuronic Acids/chemistry , Humans , Injections , Male , Mice , Mice, Inbred C57BL , Mice, Transgenic , Microspheres , alpha 1-Antitrypsin/genetics
11.
Infect Genet Evol ; 20: 257-69, 2013 Dec.
Article in English | MEDLINE | ID: mdl-24056016

ABSTRACT

Microglia plays a crucial role during virus pathogenesis in the central nervous system (CNS). Infection by rabies virus (RABV) causes a fatal infection in the CNS of all warm-blooded animals. However, the microglial responses to RABV infection have been scarcely reported. To better understand microglia-RABV interactions at the transcriptional level, a genome wide gene expression profile in mouse microglial cells line BV2 was performed using microarray analysis. The global messenger RNA changes in murine microglial cell line BV2 after 12, 24 and 48 h of infection with rabies virus CVS-11 strain were investigated using DNA Microarray and quantitative real-time PCR. Infection of CVS-11 at different time points induced different gene expression signatures in BV2 cells. The expression patterns of differentially expressed genes are shown by K-means clustering in four clusters in RABV- or mock-infected microglia at 12, 24 and 48h post infection (hpi). Gene ontology and network analysis of the differentially expressed genes in responses to RABV were performed by the Ingenuity Pathway Analysis system (IPA, Ingenuity® Systems, http://www.ingenuity.com). The results revealed that 28 genes were significantly up-regulated (P<0.01) and 1 gene was significantly down-regulated (P<0.01) in microglial cells at 12hpi, 72 genes were significantly up-regulated (P<0.01) and 24 genes were significantly down-regulated (P<0.01) at 24hpi, and 671 genes were significantly up-regulated (P<0.01) and 190 genes were significantly down-regulated (P<0.01) at 48hpi. Genes in BV2 were significantly regulated (P<0.01) in response to RABV infection and they were found to be interferon stimulated genes (Isg15, Isg20, Oasl1, Oasl2, Ifit2, Irf7 and Ifi203), chemokine genes (Ccl5, Cxcl10 and Ccrl2) and the proinflammatory factor gene (Interleukin 6). The results indicated that the differentially expressed genes from microglial cells after RABV infection were mainly involved in innate immune responses, inflammatory responses and host antiviral responses.


Subject(s)
Microglia/immunology , Microglia/virology , Rabies virus , Rabies/genetics , Animals , Brain/immunology , Brain/virology , Cell Line , Chemokines/biosynthesis , Chemokines/genetics , Gene Expression , Gene Expression Profiling , Immunity, Innate/genetics , Immunity, Innate/immunology , Inflammation/genetics , Inflammation/immunology , Interferons/immunology , Mice , Microarray Analysis , RNA, Messenger/biosynthesis , RNA, Messenger/genetics , Transcription Factors/biosynthesis , Transcription Factors/genetics , Transcription, Genetic
12.
PLoS One ; 7(5): e37170, 2012.
Article in English | MEDLINE | ID: mdl-22623990

ABSTRACT

BACKGROUND: Peste des petits ruminants (PPR) is a highly contagious infectious disease of goats, sheep and small wild ruminant species with high morbidity and mortality rates. The Peste des petits ruminants virus (PPRV) expresses a hemagglutinin (H) glycoprotein on its outer envelope that is crucial for viral attachment to host cells and represents a key antigen for inducing the host immune response. METHODOLOGY/PRINCIPAL FINDINGS: To determine whether H can be exploited to generate an effective PPRV vaccine, a replication-competent recombinant canine adenovirus type-2 (CAV-2) expressing the H gene of PPRV (China/Tibet strain) was constructed by the in vitro ligation method. The H expression cassette, including the human cytomegalovirus (hCMV) promoter/enhancer and the BGH early mRNA polyadenylation signal, was inserted into the SspI site of the E3 region, which is not essential for proliferation of CAV-2. Infectious recombinant rCAV-2-PPRV-H virus was generated in transfected MDCK cells and used to immunize goats. All vaccinated animals produced antibodies upon primary injection that were effective in neutralizing PPRV in vitro. Higher antibody titer was obtained following booster inoculation, and the antibody was detectable in goats for at least seven months. No serious recombinant virus-related adverse effect was observed in immunized animals and no adenovirus could be isolated from the urine or feces of vaccinated animals. Results showed that the recombinant virus was safe and could stimulate a long-lasting immune response in goats. CONCLUSIONS/SIGNIFICANCE: This strategy not only provides an effective PPR vaccine candidate for goats but may be a valuable mean by which to differentiate infected from vaccinated animals (the so-called DIVA approach).


Subject(s)
Adenoviruses, Canine/immunology , Goat Diseases/prevention & control , Goat Diseases/virology , Peste-des-Petits-Ruminants/veterinary , Peste-des-petits-ruminants virus/immunology , Viral Vaccines/therapeutic use , Adenoviruses, Canine/genetics , Analysis of Variance , Animals , Antibodies, Viral/blood , Blotting, Western , Cell Line , DNA Primers/genetics , Dogs , Fluorescent Antibody Technique , Goats , Hemagglutinins, Viral/genetics , Hemagglutinins, Viral/metabolism , In Vitro Techniques , Leukocytes, Mononuclear/immunology , Neutralization Tests , Peste-des-Petits-Ruminants/prevention & control , Peste-des-petits-ruminants virus/genetics , Vaccines, Synthetic/genetics , Vaccines, Synthetic/therapeutic use , Viral Vaccines/genetics
13.
Arch Virol ; 156(5): 753-8, 2011 May.
Article in English | MEDLINE | ID: mdl-21221673

ABSTRACT

A pseudotype baculovirus with the glycoprotein of vesicular stomatitis virus (VSV-G) on the envelope was used as a vector for the construction of recombinant baculovirus expressing the G protein of rabies virus (RABV) under the cytomegalovirus (CMV) promoter. The generated recombinant baculovirus (BV-G) efficiently expressed the RABV G proteins in mammalian cells. Intramuscular vaccination with BV-G (10(9) PFU/mouse) induced the production of RABV G-specific neutralizing antibodies and strong T cell responses in mice. Our data clearly indicate that pseudotype baculovirus-mediated gene delivery can be utilized as an alternative strategy to develop a new generation of vaccine against RABV infection.


Subject(s)
Antigens, Viral/immunology , Baculoviridae/genetics , Drug Carriers , Genetic Vectors , Glycoproteins/immunology , Rabies Vaccines/immunology , Rabies virus/immunology , Vesiculovirus/genetics , Viral Envelope Proteins/immunology , Animals , Antibodies, Neutralizing/blood , Antibodies, Viral/blood , Antigens, Viral/genetics , Female , Glycoproteins/genetics , Injections, Intramuscular , Mice , Mice, Inbred ICR , Rabies Vaccines/administration & dosage , Rabies Vaccines/genetics , Rabies virus/genetics , Vaccination/methods , Vaccines, Synthetic/administration & dosage , Vaccines, Synthetic/genetics , Vaccines, Synthetic/immunology , Viral Envelope Proteins/genetics
SELECTION OF CITATIONS
SEARCH DETAIL
...