Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 24
Filter
Add more filters










Publication year range
1.
Neuropharmacology ; 226: 109397, 2023 03 15.
Article in English | MEDLINE | ID: mdl-36623805

ABSTRACT

We recently reported that the "Dopamine Neuron Challenge Test" (DNC Test), a diagnostic method that measures the levels of dopamine metabolites in cerebrospinal fluid (CSF) and plasma samples after pharmacologically inducing a transient dopamine release, can detect early-stage Parkinson's disease (PD) with high sensitivity and selectivity in mouse models. The use of haloperidol in the original DNC test to challenge dopamine neurons was less than ideal, as it may cause extrapyramidal motor symptoms. Here we report an improved DNC Test, in which the original challenging agents, haloperidol and methylphenidate, are replaced by a single challenging agent, a dopamine autoreceptor preferring antagonist AJ76 or UH232. We show that the improved DNC Test can achieve the same level of sensitivity and selectivity in detecting early PD in a mouse model without causing motor side effects. These findings significantly improve the practicality of using the DNC Test as a screening or diagnostic test for detecting early-stage PD in the high-risk population in humans.


Subject(s)
Dopamine , Parkinson Disease , Animals , Mice , Humans , Dopamine/metabolism , Parkinson Disease/diagnosis , Parkinson Disease/drug therapy , Haloperidol/pharmacology , Haloperidol/therapeutic use , Homovanillic Acid/metabolism , Dopamine Antagonists/pharmacology , 3,4-Dihydroxyphenylacetic Acid/metabolism
2.
Curr Protoc ; 2(11): e587, 2022 Nov.
Article in English | MEDLINE | ID: mdl-36373979

ABSTRACT

This article describes how to assemble and operate a spectrometer-based fiber photometry system for in vivo simultaneous measurements of multiple fluorescent biosensors in freely moving mice. The first section of the article describes the step-by-step procedure to assemble a basic single-spectrometer fiber photometry system and how to expand it into a dual-spectrometer system that allows for simultaneous recordings from two sites. The second part describes the steps for a typical fiber probe implantation surgery. The last section describes how to acquire and analyze the time-lapsed spectral data. This article is intended for teaching labs how to build their own fiber photometry systems (with a video tutorial) from commercially available parts and perform in vivo recordings in behaving mice. © Published 2022. This article is a U.S. Government work and is in the public domain in the USA. Basic Protocol 1: Assembling a dual-laser, single-spectrometer fiber photometry system Support Protocol: Dual-spectrometer fiber photometry assembly Basic Protocol 2: Optical fiber probe implantation Basic Protocol 3: Data acquisition and analysis.


Subject(s)
Optical Fibers , Photometry , Animals , Mice , Photometry/methods
3.
NPJ Parkinsons Dis ; 7(1): 116, 2021 Dec 16.
Article in English | MEDLINE | ID: mdl-34916526

ABSTRACT

Diagnosing Parkinson's disease (PD) before the clinical onset proves difficult because the hallmark PD symptoms do not manifest until more than 60% of dopamine neurons in the substantia nigra pars compacta have been lost. Here we show that, by evoking a transient dopamine release and subsequently measuring the levels of dopamine metabolites in the cerebrospinal fluid and plasma, a hypodopaminergic state can be revealed when less than 30% of dopamine neurons are lost in mouse PD models. These findings may lead to sensitive and practical screening and diagnostic tests for detecting early PD in the high-risk population.

4.
J Virol Methods ; 281: 113863, 2020 07.
Article in English | MEDLINE | ID: mdl-32371233

ABSTRACT

Recombinant adeno-associated viruses (rAAVs) are robust and versatile tools for in vivo gene delivery. Natural and designer capsid variations in rAAVs allow for targeted gene delivery to specific cell types. Low immunogenicity and lack of pathogenesis also add to the popularity of this virus as an innocuous gene delivery vector for gene therapy. rAAVs are routinely used to express recombinases, sensors, detectors, CRISPR-Cas9 components, or to simply overexpress a gene of interest for functional studies. High production demand has given rise to multiple platforms for the production and purification of rAAVs. However, most platforms rely heavily on large amounts of starting material and multiple purification steps to produce highly purified viral particles. Often, researchers require several small-scale purified rAAVs. Here, we describe a simple and efficient technique for purification of recombinant rAAVs from small amounts of starting material in a two-step purification method. In this method, rAAVs are released into the packaging cell medium using high salt concentration, pelleted by ultracentrifugation to remove soluble impurities. Then, the resuspended pellet is purified using a protein spin-concentrator. In this protocol, we modify the conventional rAAV purification methods to eliminate the need for fraction collection and the labor-intensive steps for evaluating the titer and purity of individual fractions. The resulting rAAV preparations are comparable in titer and purity to commercially available samples. This simplified process can be used to generate highly purified rAAV particles on a small scale, thereby saving resources, generating less waste, and reducing a laboratory's environmental footprint.


Subject(s)
Dependovirus/isolation & purification , Virology/methods , Animals , Genetic Vectors , HEK293 Cells , Humans , Mice , Mice, Inbred C57BL , Ultracentrifugation
5.
J Virol ; 93(6)2019 03 15.
Article in English | MEDLINE | ID: mdl-30567978

ABSTRACT

Marburg virus (MARV) is a filovirus related to Ebola virus (EBOV) associated with human hemorrhagic disease. Outbreaks are sporadic and severe, with a reported case mortality rate of upward of 88%. There is currently no antiviral or vaccine available. Given the sporadic nature of outbreaks, vaccines provide the best approach for long-term control of MARV in regions of endemicity. We have developed an inactivated rabies virus-vectored MARV vaccine (FILORAB3) to protect against Marburg virus disease. Immunogenicity studies in our labs have shown that a Th1-biased seroconversion to both rabies virus and MARV glycoproteins (GPs) is beneficial for protection in a preclinical murine model. As such, we adjuvanted FILORAB3 with glucopyranosyl lipid adjuvant (GLA), a Toll-like receptor 4 agonist, in a squalene-in-water emulsion. Across two different BALB/c mouse challenge models, we achieved 92% protection against murine-adapted Marburg virus (ma-MARV). Although our vaccine elicited strong MARV GP antibodies, it did not strongly induce neutralizing antibodies. Through both in vitro and in vivo approaches, we elucidated a critical role for NK cell-dependent antibody-mediated cellular cytotoxicity (ADCC) in vaccine-induced protection. Overall, these findings demonstrate that FILORAB3 is a promising vaccine candidate for Marburg virus disease.IMPORTANCE Marburg virus (MARV) is a virus similar to Ebola virus and also causes a hemorrhagic disease which is highly lethal. In contrast to EBOV, only a few vaccines have been developed against MARV, and researchers do not understand what kind of immune responses are required to protect from MARV. Here we show that antibodies directed against MARV after application of our vaccine protect in an animal system but fail to neutralize the virus in a widely used virus neutralization assay against MARV. This newly discovered activity needs to be considered more when analyzing MARV vaccines or infections.


Subject(s)
Antibodies, Neutralizing/immunology , Antibodies, Viral/immunology , Glycoproteins/immunology , Marburg Virus Disease/immunology , Marburgvirus/immunology , Rabies virus/immunology , Rabies/immunology , Animals , Cell Line , Chlorocebus aethiops , Disease Models, Animal , Female , HEK293 Cells , Humans , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Rabies Vaccines/immunology , Vaccination/methods , Vero Cells , Viral Vaccines/immunology
6.
Neuron ; 98(4): 707-717.e4, 2018 05 16.
Article in English | MEDLINE | ID: mdl-29731250

ABSTRACT

To achieve simultaneous measurement of multiple cellular events in molecularly defined groups of neurons in vivo, we designed a spectrometer-based fiber photometry system that allows for spectral unmixing of multiple fluorescence signals recorded from deep brain structures in behaving animals. Using green and red Ca2+ indicators differentially expressed in striatal direct- and indirect-pathway neurons, we were able to simultaneously monitor the neural activity in these two pathways in freely moving animals. We found that the activities were highly synchronized between the direct and indirect pathways within one hemisphere and were desynchronized between the two hemispheres. We further analyzed the relationship between the movement patterns and the magnitude of activation in direct- and indirect-pathway neurons and found that the striatal direct and indirect pathways coordinately control the dynamics and fate of movement. VIDEO ABSTRACT.


Subject(s)
Corpus Striatum/physiology , Neural Pathways/physiology , Neurons/physiology , Spectrometry, Fluorescence/methods , Animals , Brain/diagnostic imaging , Corpus Striatum/diagnostic imaging , Green Fluorescent Proteins , Intravital Microscopy , Luminescent Proteins , Mice , Neural Pathways/diagnostic imaging , Optical Imaging , Photometry , Red Fluorescent Protein
7.
J Infect Dis ; 214(suppl 3): S342-S354, 2016 10 15.
Article in English | MEDLINE | ID: mdl-27456709

ABSTRACT

The 2013-2016 West African Ebola virus (EBOV) disease outbreak was the largest filovirus outbreak to date. Over 28 000 suspected, probable, or confirmed cases have been reported, with a 53% case-fatality rate. The magnitude and international impact of this EBOV outbreak has highlighted the urgent need for a safe and efficient EBOV vaccine. To this end, we demonstrate the immunogenicity and protective efficacy of FILORAB1, a recombinant, bivalent, inactivated rabies virus-based EBOV vaccine, in rhesus and cynomolgus monkeys. Our results demonstrate that the use of the synthetic Toll-like receptor 4 agonist glucopyranosyl lipid A in stable emulsion (GLA-SE) as an adjuvant increased the efficacy of FILORAB1 to 100% protection against lethal EBOV challenge, with no to mild clinical signs of disease. Furthermore, all vaccinated subjects developed protective anti-rabies virus antibody titers. Taken together, these results support further development of FILORAB1/GLA-SE as an effective preexposure EBOV vaccine.


Subject(s)
Ebola Vaccines/immunology , Ebolavirus/immunology , Glucosides/immunology , Hemorrhagic Fever, Ebola/prevention & control , Lipid A/immunology , Rabies virus/immunology , Rabies/prevention & control , Adjuvants, Immunologic , Animals , Antibodies, Viral/immunology , Emulsions , Female , Hemorrhagic Fever, Ebola/immunology , Hemorrhagic Fever, Ebola/virology , Macaca fascicularis , Macaca mulatta , Male , Rabies/immunology , Rabies/virology , Rabies Vaccines/immunology , Toll-Like Receptor 4/immunology , Vaccines, Inactivated/immunology , Vaccines, Synthetic/immunology
8.
J Gen Virol ; 97(8): 1942-1954, 2016 08.
Article in English | MEDLINE | ID: mdl-27166137

ABSTRACT

We previously demonstrated that small-particle (0.5-3.0 µm) aerosol infection of rhesus monkeys (Macaca mulatta) with cowpox virus (CPXV)-Brighton Red (BR) results in fulminant respiratory tract disease characterized by severe lung parenchymal pathology but only limited systemic virus dissemination and limited classic epidermal pox-like lesion development (Johnson et al., 2015). Based on these results, and to further develop CPXV as an improved model of human smallpox, we evaluated a novel large-particle aerosol (7.0-9.0 µm) exposure of rhesus monkeys to CPXV-BR and monitored for respiratory tract disease by serial computed tomography (CT). As expected, the upper respiratory tract and large airways were the major sites of virus-induced pathology following large-particle aerosol exposure. Large-particle aerosol CPXV exposure of rhesus macaques resulted in severe upper airway and large airway pathology with limited systemic dissemination.


Subject(s)
Aerosols , Cowpox virus/pathogenicity , Cowpox/pathology , Cowpox/virology , Disease Models, Animal , Respiratory Tract Infections/pathology , Respiratory Tract Infections/virology , Animals , Macaca mulatta , Respiratory Tract Infections/diagnostic imaging , Tomography, X-Ray Computed
9.
J Infect Dis ; 212 Suppl 2: S414-24, 2015 Oct 01.
Article in English | MEDLINE | ID: mdl-26063224

ABSTRACT

We previously described the generation of a novel Ebola virus (EBOV) vaccine based on inactivated rabies virus (RABV) containing EBOV glycoprotein (GP) incorporated in the RABV virion. Our results demonstrated safety, immunogenicity, and protective efficacy in mice and nonhuman primates (NHPs). Protection against viral challenge depended largely on the quality of the humoral immune response against EBOV GP.Here we present the extension and improvement of this vaccine by increasing the amount of GP incorporation into virions via GP codon-optimization as well as the addition of Sudan virus (SUDV) and Marburg virus (MARV) GP containing virions. Immunogenicity studies in mice indicate similar immune responses for both SUDV GP and MARV GP compared to EBOV GP. Immunizing mice with multiple antigens resulted in immune responses similar to immunization with a single antigen. Moreover, immunization of NHP with the new inactivated RABV EBOV vaccine resulted in high titer neutralizing antibody levels and 100% protection against lethal EBOV challenge when applied with adjuvant.Our results indicate that an inactivated polyvalent vaccine against RABV filoviruses is achievable. Finally, the novel vaccines are produced on approved VERO cells and a clinical grade RABV/EBOV vaccine for human trials has been produced.


Subject(s)
Filoviridae/immunology , Rabies Vaccines/immunology , Rabies virus/immunology , Rabies/immunology , Vaccines, Inactivated/immunology , Animals , Antibodies, Viral/immunology , Antibody Formation/immunology , Chlorocebus aethiops , Drug Evaluation, Preclinical/methods , Ebola Vaccines/immunology , Ebolavirus/immunology , Glycoproteins/immunology , Hemorrhagic Fever, Ebola/immunology , Hemorrhagic Fever, Ebola/virology , Macaca fascicularis , Marburgvirus/immunology , Mice , Mice, Inbred C57BL , Rabies/virology , Sudan , Vaccination/methods , Vero Cells
10.
Expert Rev Vaccines ; 14(7): 949-62, 2015 Jul.
Article in English | MEDLINE | ID: mdl-25864502

ABSTRACT

The recent emergence of Middle East respiratory syndrome (MERS) highlights the need to engineer new methods for expediting vaccine development against emerging diseases. However, several obstacles prevent pursuit of a licensable MERS vaccine. First, the lack of a suitable animal model for MERS complicates the in vivo testing of candidate vaccines. Second, due to the low number of MERS cases, pharmaceutical companies have little incentive to pursue MERS vaccine production as the costs of clinical trials are high. In addition, the timeline from bench research to approved vaccine use is 10 years or longer. Using novel methods and cost-saving strategies, genetically engineered vaccines can be produced quickly and cost-effectively. Along with progress in MERS animal model development, these obstacles can be circumvented or at least mitigated.


Subject(s)
Coronavirus Infections/prevention & control , Viral Vaccines/immunology , Viral Vaccines/isolation & purification , Animals , Clinical Trials as Topic , Disease Models, Animal , Drug Discovery/economics , Drug Discovery/trends , Drug Evaluation, Preclinical/economics , Drug Evaluation, Preclinical/methods , Humans
11.
Virus Res ; 197: 54-8, 2015 Feb 02.
Article in English | MEDLINE | ID: mdl-25481284

ABSTRACT

Using a recombinant rabies (RABV) vaccine platform, we have developed several safe and effective vaccines. Most recently, we have developed a RABV-based ebolavirus (EBOV) vaccine that is efficacious in nonhuman primates. One safety feature of this vaccine is the utilization of a live but replication-deficient RABV construct. In this construct, the RABV glycoprotein (G) has been deleted from the genome, requiring G trans complementation in order for new infectious viruses to be released from the initial infected cell. Here we analyze this safety feature of the bivalent RABV-based EBOV vaccine comprised of the G-deleted RABV backbone expressing EBOV glycoprotein (GP). We found that, while the level of RABV genome in infected cells is equivalent regardless of G supplementation, the production of infectious virus is indeed restricted by the lack of G, and most importantly, that the presence of EBOV GP does not substitute for G. These findings further support the safety profile of this replication-deficient RABV-EBOV bivalent vaccine.


Subject(s)
Antigens, Viral/biosynthesis , Ebola Vaccines/immunology , Gene Expression , Glycoproteins/biosynthesis , Rabies Vaccines/immunology , Viral Envelope Proteins/biosynthesis , Viral Envelope Proteins/immunology , Animals , Antigens, Viral/genetics , Ebola Vaccines/genetics , Gene Deletion , Genetic Complementation Test , Glycoproteins/genetics , Hemorrhagic Fever, Ebola/prevention & control , Primates , Rabies/prevention & control , Rabies Vaccines/genetics , Recombinant Proteins/genetics , Recombinant Proteins/immunology , Vaccines, Attenuated/genetics , Vaccines, Attenuated/immunology , Vaccines, Synthetic/genetics , Vaccines, Synthetic/immunology , Viral Envelope Proteins/genetics
12.
Vaccine ; 31(49): 5897-902, 2013 Dec 02.
Article in English | MEDLINE | ID: mdl-24120673

ABSTRACT

The safety and availability of the human polyclonal sera that is currently utilized for post-exposure treatment (PET) of rabies virus (RABV) infection remain a concern. Recombinant monoclonal antibodies have been postulated as suitable alternatives by WHO. To this extent, CL184, the RABV human antibody combination comprising monoclonal antibodies (mAbs) CR57 and CR4098, has been developed and has delivered promising clinical data to support its use for RABV PET. For this fully human IgG1 cocktail, mAbs CR57 and CR4098 are produced in the PER.C6 human cell line and combined in equal amounts in the final product. During preclinical evaluation, CR57 was shown to bind to antigenic site I whereas CR4098 neutralization was influenced by a mutation of position 336 (N336) located within antigenic site III. Here, alanine scanning was used to analyze the influence of mutations within the potential binding site for CR4098, antigenic site III, in order to evaluate the possibility of mutated rabies viruses escaping neutralization. For this approach, twenty flanking amino acids (10 upstream and 10 downstream) of the RABV glycoprotein (G) asparagine (N336) were exchanged to alanine (or serine, if already alanine) by site-directed mutagenesis. Analysis of G expression revealed four of the twenty mutant Gs to be non-functional, as shown by their lack of cell surface expression, which is a requirement for the production of infectious RABV. Therefore, these mutants were excluded from further study. The remaining sixteen mutants were introduced in an infectious clone of RABV, and recombinant RABVs (rRABVs) were recovered and utilized for in vitro neutralization assays. All of the viruses were effectively neutralized by CR4098 as well as by CR57, indicating that single amino acid exchanges in this region does not affect the broad neutralizing capability of the CL184 mAb combination.


Subject(s)
Antibodies, Monoclonal/immunology , Antigens, Viral/genetics , Glycoproteins/genetics , Post-Exposure Prophylaxis , Rabies/therapy , Alanine/genetics , Animals , Antibodies, Monoclonal/therapeutic use , Antibodies, Neutralizing/immunology , Antibodies, Neutralizing/therapeutic use , Antibodies, Viral/immunology , Antigens, Viral/immunology , Binding Sites, Antibody , Cell Line , Cricetinae , Glycoproteins/immunology , Humans , Mutagenesis, Site-Directed , Neutralization Tests , Rabies Vaccines/therapeutic use , Viral Envelope Proteins/genetics , Viral Envelope Proteins/immunology
13.
PLoS Pathog ; 9(5): e1003389, 2013.
Article in English | MEDLINE | ID: mdl-23737747

ABSTRACT

We have previously described the generation of a novel Ebola virus (EBOV) vaccine platform based on (a) replication-competent rabies virus (RABV), (b) replication-deficient RABV, or (c) chemically inactivated RABV expressing EBOV glycoprotein (GP). Mouse studies demonstrated safety, immunogenicity, and protective efficacy of these live or inactivated RABV/EBOV vaccines. Here, we evaluated these vaccines in nonhuman primates. Our results indicate that all three vaccines do induce potent immune responses against both RABV and EBOV, while the protection of immunized animals against EBOV was largely dependent on the quality of humoral immune response against EBOV GP. We also determined if the induced antibodies against EBOV GP differ in their target, affinity, or the isotype. Our results show that IgG1-biased humoral responses as well as high levels of GP-specific antibodies were beneficial for the control of EBOV infection after immunization. These results further support the concept that a successful EBOV vaccine needs to induce strong antibodies against EBOV. We also showed that a dual vaccine against RABV and filoviruses is achievable; therefore addressing concerns for the marketability of this urgently needed vaccine.


Subject(s)
Antibodies, Viral/immunology , Ebola Vaccines , Ebolavirus , Hemorrhagic Fever, Ebola , Immunoglobulin G/immunology , Rabies Vaccines , Rabies virus , Viral Matrix Proteins , Animals , Ebola Vaccines/genetics , Ebola Vaccines/immunology , Ebola Vaccines/pharmacology , Ebolavirus/genetics , Ebolavirus/immunology , Female , Hemorrhagic Fever, Ebola/immunology , Hemorrhagic Fever, Ebola/prevention & control , Macaca mulatta , Male , Mice , Rabies Vaccines/genetics , Rabies Vaccines/immunology , Rabies Vaccines/pharmacology , Rabies virus/genetics , Rabies virus/immunology , Viral Matrix Proteins/genetics , Viral Matrix Proteins/immunology , Viral Matrix Proteins/pharmacology
14.
Virology ; 434(1): 18-26, 2012 Dec 05.
Article in English | MEDLINE | ID: mdl-22889613

ABSTRACT

We are developing inactivated and live-attenuated rabies virus (RABV) vaccines expressing Ebola virus (EBOV) glycoprotein for use in humans and endangered wildlife, respectively. Here, we further characterize the pathogenesis of the live-attenuated RABV/EBOV vaccine candidates in mice in an effort to define their growth properties and potential for safety. RABV vaccines expressing GP (RV-GP) or a replication-deficient derivative with a deletion of the RABV G gene (RVΔG-GP) are both avirulent after intracerebral inoculation of adult mice. Furthermore, RVΔG-GP is completely avirulent upon intracerebral inoculation of suckling mice unlike parental RABV vaccine or RV-GP. Analysis of RVΔG-GP in the brain by quantitative PCR, determination of virus titer, and immunohistochemistry indicated greatly restricted virus replication. In summary, our findings indicate that RV-GP retains the attenuation phenotype of the live-attenuated RABV vaccine, and RVΔG-GP would appear to be an even safer alternative for use in wildlife or consideration for human use.


Subject(s)
Ebola Vaccines/adverse effects , Ebola Vaccines/immunology , Rabies Vaccines/adverse effects , Rabies Vaccines/immunology , Animals , Animals, Newborn , Brain/pathology , Brain/virology , Disease Models, Animal , Ebola Vaccines/genetics , Female , Gene Deletion , Genes, Viral , Immunohistochemistry , Mice , Mice, Inbred BALB C , Mice, SCID , Rabies Vaccines/genetics , Real-Time Polymerase Chain Reaction , Vaccines, Attenuated/adverse effects , Vaccines, Attenuated/genetics , Vaccines, Attenuated/immunology , Viral Load , Virulence
15.
Vaccine ; 30(43): 6136-41, 2012 Sep 21.
Article in English | MEDLINE | ID: mdl-22884661

ABSTRACT

We have previously developed (a) replication-competent, (b) replication-deficient, and (c) chemically inactivated rabies virus (RABV) vaccines expressing Ebola virus (EBOV) glycoprotein (GP) that induce humoral immunity against each virus and confer protection from both lethal RABV and mouse-adapted EBOV challenge in mice. Here, we expand our investigation of the immunogenic properties of these bivalent vaccines in mice. Both live and killed vaccines induced primary EBOV GP-specific T-cells and a robust recall response as measured by interferon-γ ELISPOT assay. In addition to cellular immunity, an effective filovirus vaccine will likely require a multivalent humoral immune response against multiple virus species. As a proof-of-principle experiment, we demonstrated that inactivated RV-GP could be formulated with another inactivated RABV vaccine expressing the nontoxic fragment of botulinum neurotoxin A heavy chain (HC50) without a reduction in immunity to each component. Finally, we demonstrated that humoral immunity to GP could be induced by immunization of mice with inactivated RV-GP in the presence of pre-existing immunity to RABV. The ability of these novel vaccines to induce strong humoral and cellular immunity indicates that they should be further evaluated in additional animal models of infection.


Subject(s)
Antibody Specificity , Ebola Vaccines/immunology , Hemorrhagic Fever, Ebola/prevention & control , Membrane Glycoproteins/immunology , Rabies Vaccines/immunology , Viral Matrix Proteins/immunology , Animals , Antibodies, Viral/blood , Ebolavirus/immunology , Immunity, Cellular , Immunity, Humoral , Interferon-gamma/immunology , Mice , Mice, Inbred BALB C , T-Lymphocytes/immunology , Vaccines, Inactivated/immunology
16.
J Virol ; 85(20): 10605-16, 2011 Oct.
Article in English | MEDLINE | ID: mdl-21849459

ABSTRACT

The search for a safe and efficacious vaccine for Ebola virus continues, as no current vaccine candidate is nearing licensure. We have developed (i) replication-competent, (ii) replication-deficient, and (iii) chemically inactivated rabies virus (RABV) vaccines expressing Zaire Ebola virus (ZEBOV) glycoprotein (GP) by a reverse genetics system based on the SAD B19 RABV wildlife vaccine. ZEBOV GP is efficiently expressed by these vaccine candidates and is incorporated into virions. The vaccine candidates were avirulent after inoculation of adult mice, and viruses with a deletion in the RABV glycoprotein had greatly reduced neurovirulence after intracerebral inoculation in suckling mice. Immunization with live or inactivated RABV vaccines expressing ZEBOV GP induced humoral immunity against each virus and conferred protection from both lethal RABV and EBOV challenge in mice. The bivalent RABV/ZEBOV vaccines described here have several distinct advantages that may speed the development of inactivated vaccines for use in humans and potentially live or inactivated vaccines for use in nonhuman primates at risk of EBOV infection in endemic areas.


Subject(s)
Ebola Vaccines/immunology , Rabies Vaccines/immunology , Animals , Antibodies, Viral/blood , Brain/virology , Disease Models, Animal , Ebola Vaccines/administration & dosage , Ebola Vaccines/adverse effects , Ebola Vaccines/genetics , Ebolavirus/genetics , Ebolavirus/immunology , Hemorrhagic Fever, Ebola/prevention & control , Mice , Mice, Inbred BALB C , Rabies/prevention & control , Rabies Vaccines/administration & dosage , Rabies Vaccines/adverse effects , Rabies Vaccines/genetics , Rabies virus/genetics , Rabies virus/immunology , Rodent Diseases/prevention & control , Vaccines, Attenuated/administration & dosage , Vaccines, Attenuated/adverse effects , Vaccines, Attenuated/genetics , Vaccines, Attenuated/immunology , Vaccines, Inactivated/administration & dosage , Vaccines, Inactivated/adverse effects , Vaccines, Inactivated/genetics , Vaccines, Inactivated/immunology , Vaccines, Synthetic/administration & dosage , Vaccines, Synthetic/adverse effects , Vaccines, Synthetic/genetics , Vaccines, Synthetic/immunology , Virulence
17.
Nat Rev Microbiol ; 8(1): 51-61, 2010 Jan.
Article in English | MEDLINE | ID: mdl-19946287

ABSTRACT

Rabies virus, the prototypical neurotropic virus, causes one of the most lethal zoonotic diseases. According to official estimates, over 55,000 people die of the disease annually, but this is probably a severe underestimation. A combination of virulence factors enables the virus to enter neurons at peripheral sites and travel through the spinal cord to the brain of the infected host, where it often induces aggression that facilitates the transfer of the virus to a new host. This Review summarizes the current knowledge of the replication cycle of rabies virus and virus- host cell interactions, both of which are fundamental elements in our quest to understand the life cycle of rabies virus and the pathogenesis of rabies.


Subject(s)
Brain/virology , Peripheral Nervous System/virology , Rabies virus/physiology , Spinal Cord/virology , Animals , Humans , Models, Biological , Rabies virus/pathogenicity , Viral Proteins/physiology , Virulence Factors/physiology
18.
Vaccine ; 28(2): 299-308, 2009 Dec 11.
Article in English | MEDLINE | ID: mdl-19879223

ABSTRACT

Highly attenuated rabies virus (RV) vaccine vectors were evaluated for their ability to protect against highly pathogenic SIV(mac251) challenge. Mamu-A*01 negative rhesus macaques were immunized in groups of four with either: RV expressing SIV(mac239)-GagPol, a combination of RV expressing SIV(mac239)-Env and RV expressing SIV(mac239)-GagPol, or with empty RV vectors. Eight weeks later animals received a booster immunization with a heterologous RV expressing the same antigens. At 12 weeks post-boost, all animals were challenged intravenously with 100 TCID(50) of pathogenic SIV(mac251-CX). Immunized macaques in both vaccine groups had 1.3-1.6-log-fold decrease in viral set point compared to control animals. The GagPol/Env immunized animals also had a significantly lower peak viral load. When compared to control animals following challenge, vaccinated macaques had a more rapid induction of SIV(mac251) neutralizing antibodies and of CD8(+) T cell responses to various SIV epitopes. Moreover, vaccinated macaques better maintained peripheral memory CD4(+) T cells and were able to mount a poly-functional CD8(+) T cell response in the mucosa. These findings indicate promise for RV-based vectors and have important implications for the development of an efficacious HIV vaccine.


Subject(s)
Antibodies, Neutralizing/immunology , CD8-Positive T-Lymphocytes/immunology , Macaca mulatta/immunology , Macaca mulatta/virology , Rabies virus/immunology , SAIDS Vaccines/immunology , Simian Acquired Immunodeficiency Syndrome/immunology , Animals , Immunity, Cellular/immunology , Immunity, Humoral/immunology , Simian Immunodeficiency Virus/immunology
19.
J Infect Dis ; 200(8): 1251-60, 2009 Oct 15.
Article in English | MEDLINE | ID: mdl-19764884

ABSTRACT

Although current postexposure prophylaxis rabies virus (RV) vaccines are effective, approximately 40,000-70,000 rabies-related deaths are reported annually worldwide. The development of effective formulations requiring only 1-2 applications would significantly reduce mortality. We assessed in mice and nonhuman primates the efficacy of replication-deficient RV vaccine vectors that lack either the matrix (M) or phosphoprotein (P) gene. A single dose of M gene-deficient RV induced a more rapid and efficient anti-RV response than did P gene-deficient RV immunization. Furthermore, the M gene-deleted RV vaccine induced 4-fold higher virus-neutralizing antibody (VNA) levels in rhesus macaques than did a commercial vaccine within 10 days after inoculation, and at 180 days after immunization rhesus macaques remained healthy and had higher-avidity antibodies, higher VNA titers, and a more potent antibody response typical of a type 1 T helper response than did animals immunized with a commercial vaccine. The data presented in this article suggest that the M gene-deleted RV vaccine is safe and effective and holds the potential of replacing current pre- and postexposure RV vaccines.


Subject(s)
Antibodies, Viral/blood , Rabies Vaccines/immunology , Rabies virus/physiology , Rabies/prevention & control , Vaccines, Attenuated/immunology , Animals , Antibody Affinity , DNA-Binding Proteins/genetics , DNA-Binding Proteins/metabolism , Female , Gene Deletion , Macaca mulatta , Male , Mice , Mice, Inbred BALB C , Mice, Knockout , Rabies/immunology , Rabies Vaccines/adverse effects , Vaccines, Attenuated/adverse effects , Virus Replication
20.
Vaccine ; 26(50): 6405-14, 2008 Nov 25.
Article in English | MEDLINE | ID: mdl-18804506

ABSTRACT

The type of immune response induced by a vaccine is a critical factor that determines its effectiveness in preventing infection or disease. Inactivated and live rabies virus (RV) vaccine strains elicit an IgG1-biased and IgG1/IgG2a-balanced antibody response, respectively. However, IgG2a antibodies are potent inducers of anti-viral effector functions, and therefore, a viral vaccine vector that can elicit an IgG2a-biased antibody response may be more effective against RV infection. Here we describe the humoral immune response of a live replication-deficient phosphoprotein (P)-deleted RV vector (SPBN-DeltaP), or a recombinant P-deleted virus that expresses two copies of the RV glycoprotein (G) gene (SPBN-DeltaP-RVG), and compare it to a UV-inactivated RV. Mice inoculated with UV-inactivated RV induced predominantly an IgG1-specific antibody response, while live recombinant SPBN-DeltaP exhibited a mixed IgG1/IgG2a antibody response, which is consistent with the isotype profiles from the replication-competent parental viruses. Survivorship in mice after pathogenic RV challenge indicates a 10-fold higher efficiency of live SPBN-DeltaP compared to UV-inactivated SPBN-DeltaP. In addition, SPBN-DeltaP-RVG induced a more rapid and robust IgG2a response that protected mice more effectively than SPBN-DeltaP. Of note, 10(3)ffu of SPBN-DeltaP-RVG-induced anti-RV antibodies that were 100% protective in mice against pathogenic RV challenge. The increased immune response was directed not only against RV G but also against the ribonucleoprotein (RNP), indicating that the expression of two RV G genes from SPBN-DeltaP-RVG enhances the immune response to other RV antigens as well. In addition, Rag2 mice inoculated intramuscularly with 10(5)ffu/mouse of SPBN-DeltaP showed no clinical signs of rabies, and no viral RNA was detected in the spinal cord or brain of inoculated mice. Therefore, the safety of the P-deleted vectors along with the onset and magnitude of the IgG2a-induced immune response by SPBN-DeltaP-RVG indicate that this vector holds great promise as either a therapeutic or preventative vaccine against RV or other infectious diseases.


Subject(s)
Antibodies, Viral/blood , Gene Dosage , Genetic Vectors , Rabies Vaccines/immunology , Rabies/prevention & control , Viral Envelope Proteins/immunology , Virus Diseases/prevention & control , Animals , Cricetinae , Female , Gene Deletion , Mice , Mice, Inbred BALB C , Phosphoproteins/genetics , Rabies/immunology , Rabies/virology , Rabies Vaccines/administration & dosage , Rabies Vaccines/genetics , Rabies virus/immunology , Viral Envelope Proteins/genetics , Viral Envelope Proteins/metabolism , Virus Diseases/immunology , Virus Diseases/virology
SELECTION OF CITATIONS
SEARCH DETAIL
...