Your browser doesn't support javascript.
loading
Montrer: 20 | 50 | 100
Résultats 1 - 9 de 9
Filtrer
Plus de filtres










Base de données
Gamme d'année
1.
mBio ; 9(2)2018 03 06.
Article de Anglais | MEDLINE | ID: mdl-29511072

RÉSUMÉ

Chikungunya virus (CHIKV) is a reemerging arbovirus capable of causing explosive outbreaks of febrile illness, polyarthritis, and polyarthralgia, inflicting severe morbidity on affected populations. CHIKV can be genetically classified into 3 major lineages: West African (WA); East, Central, and South African (ECSA); Indian Ocean (IOL); and Asian. Additionally, the Indian Ocean (IOL) sublineage emerged within the ECSA clade and the Asian/American sublineage emerged within the Asian clade. While differences in epidemiological and pathological characteristics among outbreaks involving different CHIKV lineages and sublineages have been suggested, few targeted investigations comparing lineage virulence levels have been reported. We compared the virulence levels of CHIKV isolates representing all major lineages and sublineages in the type I interferon receptor-knockout A129 mouse model and found lineage-specific differences in virulence. We also evaluated the cross-protective efficacy of the IOL-derived, live-attenuated vaccine strain CHIKV/IRESv1 against the Asian/American CHIKV isolate YO123223 in both murine and nonhuman primate models, as well as the WA strain SH2830 in a murine model. The CHIKV/IRES vaccine provided protection both in mice and in nonhuman primate cohorts against Caribbean strain challenge and protected mice against WA challenge. Taken together, our data suggest that Asian/American CHIKV strains are less virulent than those in the Asian, ECSA, and WA lineages and that despite differences in virulence, IOL-based vaccine strains offer robust cross-protection against strains from other lineages. Further research is needed to elucidate the genetic basis for variation in CHIKV virulence in the A129 mouse model and to corroborate this variation with human pathogenicity.IMPORTANCE Chikungunya virus (CHIKV) is a reemerging human pathogen capable of causing debilitating and disfiguring polyarthritis, which can last for months to years after initial fever has resolved. There are four major genetic lineages of CHIKV, as well as two recently emerged sublineages, none of which have been evaluated for differences in virulence. Moreover, the ability of chikungunya vaccines to cross-protect against heterologous CHIKV lineages has not been explored. Therefore, we sought to compare the virulence levels among CHIKV lineages, as well as to evaluate the cross-protective efficacy of the CHIKV/IRESv1 vaccine candidate, in two different models of CHIKV infection. Our results suggest that, although significant differences in virulence were observed among CHIKV lineages, the CHIKV/IRESv1 vaccine elicits cross-lineage protective immunity. These findings provide valuable information for predicting the severity of CHIKV-associated morbidity in future outbreaks, as well as vaccine development considerations.


Sujet(s)
Virus du chikungunya/pathogénicité , Alphavirus/génétique , Alphavirus/immunologie , Alphavirus/pathogénicité , Animaux , Fièvre chikungunya/immunologie , Fièvre chikungunya/virologie , Virus du chikungunya/génétique , Virus du chikungunya/immunologie , Souris , Souches mutantes de souris , Primates , Vaccins antiviraux/usage thérapeutique , Virulence/génétique
2.
PLoS Negl Trop Dis ; 9(5): e0003797, 2015 May.
Article de Anglais | MEDLINE | ID: mdl-26020513

RÉSUMÉ

Venezuelan equine encephalitis virus (VEEV) is an arbovirus endemic to the Americas that is responsible for severe, sometimes fatal, disease in humans and horses. We previously described an IRES-based VEE vaccine candidate based up the IE serotype that offers complete protection against a lethal subtype IE VEEV challenge in mice. Here we demonstrate the IRES-based vaccine's ability to protect against febrile disease in cynomolgus macaques. Vaccination was well tolerated and elicited robust neutralizing antibody titers noticed as early as day 14. Moreover, complete protection from disease characterized by absence of viremia and characteristic fever following aerosolized IE VEEV challenge was observed in all vaccinees compared to control animals, which developed clinical disease. Together, these results highlight the safety and efficacy of IRES-based VEEV vaccine to protect against an endemic, pathogenic VEEV IE serotype.


Sujet(s)
Anticorps antiviraux/sang , Virus de l'encéphalite équine du Venezuela/immunologie , Encéphalomyélite équine du Vénézuéla/prévention et contrôle , Maladies des chevaux/prévention et contrôle , Vaccination , Vaccins antiviraux/immunologie , Aérosols , Animaux , Anticorps neutralisants/sang , Chlorocebus aethiops , Modèles animaux de maladie humaine , Encéphalomyélite équine du Vénézuéla/immunologie , Femelle , Maladies des chevaux/immunologie , Equus caballus , Humains , Sites internes d'entrée des ribosomes/immunologie , Macaca fascicularis , Mâle , Agents protecteurs , Répartition aléatoire , Vaccins atténués/immunologie , Cellules Vero , Virémie
3.
Proc Natl Acad Sci U S A ; 112(12): 3782-7, 2015 Mar 24.
Article de Anglais | MEDLINE | ID: mdl-25775591

RÉSUMÉ

Ricin toxin (RT) is the second most lethal toxin known; it has been designated by the CDC as a select agent. RT is made by the castor bean plant; an estimated 50,000 tons of RT are produced annually as a by-product of castor oil. RT has two subunits, a ribotoxic A chain (RTA) and galactose-binding B chain (RTB). RT binds to all mammalian cells and once internalized, a single RTA catalytically inactivates all of the ribosomes in a cell. Administered as an aerosol, RT causes rapid lung damage and fibrosis followed by death. There are no Food and Drug Administration-approved vaccines and treatments are only effective in the first few hours after exposure. We have developed a recombinant RTA vaccine that has two mutations V76M/Y80A (RiVax). The protein is expressed in Escherichia coli and is nontoxic and immunogenic in mice, rabbits, and humans. When vaccinated mice are challenged with injected, aerosolized, or orally administered (gavaged) RT, they are completely protected. We have now developed a thermostable, aluminum-adjuvant-containing formulation of RiVax and tested it in rhesus macaques. After three injections, the animals developed antibodies that completely protected them from a lethal dose of aerosolized RT. These antibodies neutralized RT and competed to varying degrees with a panel of neutralizing and nonneutralizing mouse monoclonal antibodies known to recognize specific epitopes on native RTA. The resulting antibody competition profile could represent an immunologic signature of protection. Importantly, the same signature was observed using sera from RiVax-immunized humans.


Sujet(s)
Anticorps neutralisants/composition chimique , Épitopes/composition chimique , Ricine/composition chimique , Vaccins/composition chimique , Aérosols , Animaux , Anticorps monoclonaux/composition chimique , Test ELISA , Cellules épithéliales/composition chimique , Humains , Immunoglobuline G/composition chimique , Poumon/anatomopathologie , Macaca mulatta , Souris , Conformation moléculaire , Température
4.
Vaccine ; 33(5): 686-92, 2015 Jan 29.
Article de Anglais | MEDLINE | ID: mdl-25533326

RÉSUMÉ

The Gram-negative Burkholderia mallei is a zoonotic pathogen and the causative agent of glanders disease. Because the bacteria maintain the potential to be used as a biothreat agent, vaccine strategies are required for human glanders prophylaxis. A rhesus macaque (Macaca mulatta) model of pneumonic (inhalational) glanders was established and the protective properties of a nanoparticle glycoconjugate vaccine composed of Burkholderia thailandensis LPS conjugated to FliC was evaluated. An aerosol challenge dose of ∼1×10(4) CFU B. mallei produced mortality in 50% of naïve animals (n=2/4), 2-3 days post-exposure. Although survival benefit was not observed by vaccination with a glycoconjugate glanders vaccine (p=0.42), serum LPS-specific IgG titers were significantly higher on day 80 in 3 vaccinated animals who survived compared with 3 vaccinated animals who died. Furthermore, B. mallei was isolated from multiple organs of both non-vaccinated survivors, but not from any organs of 3 vaccinated survivors at 30 days post-challenge. Taken together, this is the first time a candidate vaccine has been evaluated in a non-human primate aerosol model of glanders and represents the initial step for consideration in pre-clinical studies.


Sujet(s)
Vaccins antibactériens/immunologie , Burkholderia mallei/immunologie , Morve/prévention et contrôle , Or/administration et posologie , Nanoparticules/administration et posologie , Animaux , Anticorps antibactériens/sang , Antigènes bactériens/administration et posologie , Antigènes bactériens/immunologie , Vaccins antibactériens/administration et posologie , Modèles animaux de maladie humaine , Glycoconjugués/administration et posologie , Glycoconjugués/immunologie , Lipopolysaccharides/administration et posologie , Lipopolysaccharides/immunologie , Macaca mulatta , Analyse de survie , Vaccins conjugués/administration et posologie , Vaccins conjugués/immunologie
5.
Clin Vaccine Immunol ; 21(12): 1668-80, 2014 Dec.
Article de Anglais | MEDLINE | ID: mdl-25298110

RÉSUMÉ

Despite the availability of annually formulated vaccines, influenza virus infection remains a worldwide public health burden. Therefore, it is important to develop preclinical challenge models that enable the evaluation of vaccine candidates while elucidating mechanisms of protection. Here, we report that naive rhesus macaques challenged with 2009 pandemic H1N1 (pH1N1) influenza virus do not develop observable clinical symptoms of disease but develop a subclinical biphasic fever on days 1 and 5 to 6 postchallenge. Whole blood microarray analysis further revealed that interferon activity was associated with fever. We then tested whether type I interferon activity in the blood is a correlate of vaccine efficacy. The animals immunized with candidate vaccines carrying hemagglutinin (HA) or nucleoprotein (NP) exhibited significantly reduced interferon activity on days 5 to 6 postchallenge. Supported by cellular and serological data, we conclude that blood interferon activity is a prominent marker that provides a convenient metric of influenza virus vaccine efficacy in the subclinical rhesus macaque model.


Sujet(s)
Anticorps antiviraux/sang , Sous-type H1N1 du virus de la grippe A/immunologie , Vaccins antigrippaux/immunologie , Grippe humaine/immunologie , Infections à Orthomyxoviridae/prévention et contrôle , Animaux , Humains , Immunisation , Macaca mulatta , Vaccination
6.
J Infect Dis ; 210(10): 1639-48, 2014 Nov 15.
Article de Anglais | MEDLINE | ID: mdl-24879799

RÉSUMÉ

The hallmark of disease caused by tick- and louse-borne relapsing fever due to Borrelia infection is cyclic febrile episodes, which in humans results in severe malaise and may lead to death. To evaluate the pathogenesis of relapsing fever due to spirochetes in an animal model closely related to humans, disease caused by Borrelia turicatae after tick bite was compared in 2 rhesus macaques in which radiotelemetry devices that recorded body temperatures in 24-hour increments were implanted. The radiotelemetry devices enabled real-time acquisition of core body temperatures and changes in heart rates and electrocardiogram intervals for 28 consecutive days without the need to constantly manipulate the animals. Blood specimens were also collected from all animals for 14 days after tick bite, and spirochete densities were assessed by quantitative polymerase chain reaction. The complexity of disease caused by relapsing-fever spirochetes was demonstrated in the nonhuman primates monitored in real time. The animals experienced prolonged episodes of hyperthermia and hypothermia; disruptions in their diurnal patterns and repolarization of the heart were also observed. This is the first report of the characterizing disease progression with continuous monitoring in an animal model of relapsing fever due to Borrelia infection.


Sujet(s)
Borrelia/isolement et purification , Fièvre récurrente/microbiologie , Fièvre récurrente/anatomopathologie , Morsures de tiques/complications , Animaux , Température du corps , Modèles animaux de maladie humaine , Évolution de la maladie , Rythme cardiaque , Macaca mulatta , Télémétrie , Facteurs temps
7.
J Infect Dis ; 209(12): 1891-9, 2014 Jun 15.
Article de Anglais | MEDLINE | ID: mdl-24403555

RÉSUMÉ

Chikungunya virus (CHIKV) is a mosquito-borne alphavirus that causes major epidemics of rash, fever, and debilitating arthritis. Currently, there are no vaccines or antivirals available for prevention or treatment. We therefore generated 2 live-attenuated vaccine candidates based on the insertion of a picornavirus internal ribosome entry site (IRES) sequence into the genome of CHIKV. Vaccination of cynomolgus macaques with a single dose of either vaccine produced no signs of disease but was highly immunogenic. After challenge with a subcutaneous inoculation of wild-type CHIKV, both vaccine candidates prevented the development of detectable viremia. Protected animals also exhibited no significant changes in core body temperature or cardiovascular rhythm, whereas sham-vaccinated animals showed hyperthermia, followed by sustained hypothermia, as well as significant changes in heart rate. These CHIKV/IRES vaccine candidates appear to be safe and efficacious, supporting their strong potential as human vaccines to protect against CHIKV infection and reduce transmission and further spread.


Sujet(s)
Infections à alphavirus/prévention et contrôle , Virus du chikungunya/isolement et purification , Macaca fascicularis/immunologie , Vaccins antiviraux/administration et posologie , Animaux , Anticorps neutralisants/sang , Anticorps neutralisants/immunologie , Anticorps antiviraux/sang , Anticorps antiviraux/immunologie , Fièvre chikungunya , Virus du chikungunya/génétique , Virus du chikungunya/immunologie , Modèles animaux de maladie humaine , Relation dose-effet des médicaments , Macaca fascicularis/virologie , Télémétrie , Vaccination , Vaccins atténués/administration et posologie , Vaccins atténués/immunologie , Vaccins antiviraux/immunologie
8.
Am J Pathol ; 177(3): 1286-97, 2010 Sep.
Article de Anglais | MEDLINE | ID: mdl-20671267

RÉSUMÉ

Recent studies suggest a link between neuropsychiatric disorders and HIV/SIV infection. Most evidence indicates that monocytes/macrophages are the primary cell type infected within the CNS and that they contribute to CNS inflammation and neurological disease. Substance P (SP), a pleotropic neuropeptide implicated in inflammation, depression, and immune modulation via interaction with its cognate receptor, the neurokinin 1 receptor (NK1-R), is produced by monocyte/macrophages. While the presence of NK1-R on neurons is well known, its role on cells of the immune system such as monocyte/macrophages is just beginning to emerge. Therefore, we have examined the expression of SP and NK1-R and their relationship to SIV/HIV encephalitis (SIVE/HIVE) lesions and SIV-infected cells. These studies demonstrated intense expression of SP and NK1-R in SIVE lesions, with macrophages being the principal cell expressing NK1-R. Interestingly, all of the SIV-infected macrophages expressed NK1-R. Additionally, we examined the functional role of SP as a proinflammatory mediator of monocyte activation and chemotaxis. These studies demonstrated that treatment of monocytes with SP elicited changes in cell-surface expression for CCR5 and NK1-R in a dose-dependent manner. Moreover, pretreatment with SP enhanced both SP- and CCL5-mediated chemotaxis. All of these findings suggest that SP and NK1-R are important in SIV infection of macrophages and the development of SIVE lesions.


Sujet(s)
Encéphale/métabolisme , Récepteur de la neurokinine 1/métabolisme , Syndrome d'immunodéficience acquise du singe/métabolisme , Virus de l'immunodéficience simienne/immunologie , Substance P/métabolisme , Animaux , Astrocytes/immunologie , Astrocytes/métabolisme , Astrocytes/virologie , Encéphale/immunologie , Encéphale/virologie , Mouvement cellulaire , Cellules cultivées , Cytométrie en flux , Immunohistochimie , Macaca mulatta , Macrophages/immunologie , Macrophages/métabolisme , Macrophages/virologie , Microscopie confocale , Neurones/immunologie , Neurones/métabolisme , Neurones/virologie , Récepteur de la neurokinine 1/immunologie , Syndrome d'immunodéficience acquise du singe/immunologie , Syndrome d'immunodéficience acquise du singe/virologie , Substance P/immunologie
9.
Blood ; 114(14): 2917-25, 2009 Oct 01.
Article de Anglais | MEDLINE | ID: mdl-19383966

RÉSUMÉ

It is widely accepted that destruction of CD4(+) T cells and viral load are the primary markers for immunodeficiency in HIV-1-infected humans and in simian immunodeficiency virus (SIV)-infected macaques. However, monocyte/macrophages are also important targets of HIV/SIV infection and a critical link between innate and adaptive immunity. We therefore examined whether changes in cells of the monocyte/macrophage lineage could be linked to the pathogenesis of AIDS in the rhesus macaque model. Here, we show that massive turnover of peripheral monocytes associated with death of tissue macrophages correlates with AIDS progression in macaques. More importantly, the level of monocyte turnover was not linked to the CD4(+) T-cell count and was a better predictive marker for AIDS progression than was viral load or lymphocyte activation. Our results show the importance of monocyte/macrophages in the pathogenesis of AIDS and suggest the dynamic changes of the monocyte/macrophages as a new marker for AIDS progression.


Sujet(s)
Modèles animaux de maladie humaine , Monocytes/métabolisme , Syndrome d'immunodéficience acquise du singe/physiopathologie , Virus de l'immunodéficience simienne/physiologie , Animaux , Cellules de la moelle osseuse/physiologie , Évolution de la maladie , Cytométrie en flux , Numération des leucocytes , Macaca mulatta , Macrophages/métabolisme , Macrophages/virologie , Modèles théoriques , Monocytes/virologie , ARN viral/sang , Syndrome d'immunodéficience acquise du singe/virologie , Lymphocytes T/immunologie , Lymphocytes T/anatomopathologie , Lymphocytes T/virologie , Charge virale
SÉLECTION CITATIONS
DÉTAIL DE RECHERCHE
...