Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 37
Filtrar
Mais filtros

Intervalo de ano de publicação
1.
Proc Natl Acad Sci U S A ; 119(12): e2200065119, 2022 03 22.
Artigo em Inglês | MEDLINE | ID: mdl-35286211

RESUMO

SignificanceConcern has increased about the pandemic potential of Nipah virus (NiV). Similar to SARS-CoV-2, NiV is an RNA virus that is transmitted by respiratory droplets. There are currently no NiV vaccines licensed for human use. While several preventive vaccines have shown promise in protecting animals against lethal NiV disease, most studies have assessed protection 1 mo after vaccination. However, in order to contain and control outbreaks, vaccines that can rapidly confer protection in days rather than months are needed. Here, we show that a recombinant vesicular stomatitis virus vector expressing the NiV glycoprotein can completely protect monkeys vaccinated 7 d prior to NiV exposure and 67% of animals vaccinated 3 d before NiV challenge.


Assuntos
Infecções por Henipavirus/veterinária , Vírus Nipah/imunologia , Doenças dos Primatas/prevenção & controle , Vacinas Sintéticas/imunologia , Vacinas Virais/imunologia , Animais , Anticorpos Neutralizantes , Anticorpos Antivirais/imunologia , Biomarcadores , Vetores Genéticos , Estimativa de Kaplan-Meier , Testes de Neutralização , Avaliação de Resultados em Cuidados de Saúde , Doenças dos Primatas/diagnóstico , Doenças dos Primatas/mortalidade , Doenças dos Primatas/virologia , Vacinação , Carga Viral
2.
Am J Primatol ; 82(8): e23176, 2020 08.
Artigo em Inglês | MEDLINE | ID: mdl-32686188

RESUMO

The emergence of SARS-CoV-2 in late 2019 and human responses to the resulting COVID-19 pandemic in early 2020 have rapidly changed many aspects of human behavior, including our interactions with wildlife. In this commentary, we identify challenges and opportunities at human-primate interfaces in light of COVID-19, focusing on examples from Asia, and make recommendations for researchers working with wild primates to reduce zoonosis risk and leverage research opportunities. First, we briefly review the evidence for zoonotic origins of SARS-CoV-2 and discuss risks of zoonosis at the human-primate interface. We then identify challenges that the pandemic has caused for primates, including reduced nutrition, increased intraspecific competition, and increased poaching risk, as well as challenges facing primatologists, including lost research opportunities. Subsequently, we highlight opportunities arising from pandemic-related lockdowns and public health messaging, including opportunities to reduce the intensity of problematic human-primate interfaces, opportunities to reduce the risk of zoonosis between humans and primates, opportunities to reduce legal and illegal trade in primates, new opportunities for research on human-primate interfaces, and opportunities for community education. Finally, we recommend specific actions that primatologists should take to reduce contact and aggression between humans and primates, to reduce demand for primates as pets, to reduce risks of zoonosis in the context of field research, and to improve understanding of human-primate interfaces. Reducing the risk of zoonosis and promoting the well-being of humans and primates at our interfaces will require substantial changes from "business as usual." We encourage primatologists to help lead the way.


Assuntos
Infecções por Coronavirus/prevenção & controle , Pandemias/prevenção & controle , Pneumonia Viral/prevenção & controle , Doenças dos Primatas/prevenção & controle , Zoonoses/prevenção & controle , Animais , COVID-19 , Conservação dos Recursos Naturais/tendências , Infecções por Coronavirus/transmissão , Comportamento Alimentar/fisiologia , Humanos , Pneumonia Viral/transmissão , Doenças dos Primatas/transmissão , Doenças dos Primatas/virologia , Primatas , Fatores de Risco , Zoonoses/transmissão
3.
Am J Primatol ; 82(8): e23161, 2020 08.
Artigo em Inglês | MEDLINE | ID: mdl-32583538

RESUMO

Year 2020 has brought the greatest global pandemic to hit the world since the end of the First World War. The severe acute respiratory syndrome coronavirus 2 and the resulting disease named coronavirus disease 2019 has brought the world to its knees both financially and medically. The American Society of Primatologists has postponed their annual meetings from the end of May 2020 until the end of September 2020, while the International Primatological Society have postponed their biennial congress from August 2020 to August 2021, which has also resulted in their 2022 meetings in Malaysia being pushed back until 2023. Here, I explore the potential dangers of pursuing any primate fieldwork during this pandemic on our study species, their ecosystems, and local peoples. I believe that the risk of bringing this virus into our study ecosystems is too great and that primatologists should cancel all field research until the pandemic ends or a vaccine/reliable treatment is widely available. This is the year we all must become One Health practitioners!


Assuntos
Infecções por Coronavirus/prevenção & controle , Educação em Veterinária , Espécies em Perigo de Extinção , Pandemias/prevenção & controle , Pneumonia Viral/prevenção & controle , Doenças dos Primatas/prevenção & controle , Primatas , Zoologia/métodos , Animais , COVID-19 , Congressos como Assunto/tendências , Infecções por Coronavirus/transmissão , Educação em Veterinária/métodos , Educação em Veterinária/tendências , Humanos , Pneumonia Viral/transmissão , Doenças dos Primatas/virologia , Zoologia/tendências
4.
J Infect Dis ; 218(2): 218-227, 2018 06 20.
Artigo em Inglês | MEDLINE | ID: mdl-29566184

RESUMO

Background: The emerging zoonotic paramyxovirus Nipah virus (NiV) causes severe respiratory and neurological disease in humans, with high fatality rates. Nipah virus can be transmitted via person-to-person contact, posing a high risk for epidemic outbreaks. However, a broadly applicable approach for human NiV outbreaks in field settings is lacking. Methods: We engineered new antiviral lipopeptides and analyzed in vitro fusion inhibition to identify an optimal candidate for prophylaxis of NiV infection in the lower respiratory tract, and we assessed antiviral efficiency in 2 different animal models. Results: We show that lethal NiV infection can be prevented with lipopeptides delivered via the respiratory route in both hamsters and nonhuman primates. By targeting retention of peptides for NiV prophylaxis in the respiratory tract, we avoid its systemic delivery in individuals who need only prevention, and thus we increase the safety of treatment and enhance utility of the intervention. Conclusions: The experiments provide a proof of concept for the use of antifusion lipopeptides for prophylaxis of lethal NiV. These results advance the goal of rational development of potent lipopeptide inhibitors with desirable pharmacokinetic and biodistribution properties and a safe effective delivery method to target NiV and other pathogenic viruses.


Assuntos
Quimioprevenção/métodos , Infecções por Henipavirus/prevenção & controle , Lipopeptídeos/administração & dosagem , Vírus Nipah/fisiologia , Doenças dos Primatas/prevenção & controle , Proteínas do Envelope Viral/antagonistas & inibidores , Inibidores de Proteínas Virais de Fusão/administração & dosagem , Animais , Broncopneumonia/prevenção & controle , Broncopneumonia/veterinária , Chlorocebus aethiops , Modelos Animais de Doenças , Feminino , Humanos , Masculino , Mesocricetus
5.
J Virol ; 89(3): 1781-93, 2015 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-25410871

RESUMO

UNLABELLED: Varicella-zoster virus (VZV) is a human neurotropic alphaherpesvirus and the etiological agent of varicella (chickenpox) and herpes zoster (HZ, shingles). Previously, inoculation of monkeys via the subcutaneous, intratracheal, intravenous, or oral-nasal-conjunctival routes did not recapitulate all the hallmarks of VZV infection, including varicella, immunity, latency, and reactivation. Intrabronchial inoculation of rhesus macaques (RMs) with simian varicella virus (SVV), a homolog of VZV, recapitulates virologic and immunologic hallmarks of VZV infection in humans. Given that VZV is acquired primarily via the respiratory route, we investigated whether intrabronchial inoculation of RMs with VZV would result in a robust model. Despite the lack of varicella and viral replication in either the lungs or whole blood, all four RMs generated an immune response characterized by the generation of VZV-specific antibodies and T cells. Two of 4 VZV-inoculated RMs were challenged with SVV to determine cross-protection. VZV-immune RMs displayed no varicella rash and had lower SVV viral loads and earlier and stronger humoral and cellular immune responses than controls. In contrast to the results for SVV DNA, no VZV DNA was detected in sensory ganglia at necropsy. In summary, following an abortive VZV infection, RMs developed an adaptive immune response that conferred partial protection against SVV challenge. These data suggest that a replication-incompetent VZV vaccine that does not establish latency may provide sufficient protection against VZV disease and that VZV vaccination of RMs followed by SVV challenge provides a model to evaluate new vaccines and therapeutics against VZV. IMPORTANCE: Although VZV vaccine strain Oka is attenuated, it can cause mild varicella, establish latency, and in rare cases, reactivate to cause herpes zoster (HZ). Moreover, studies suggest that the HZ vaccine (Zostavax) only confers short-lived immunity. The development of more efficacious vaccines would be facilitated by a robust animal model of VZV infection. The data presented in this report show that intrabronchial inoculation of rhesus macaques (RMs) with VZV resulted in an abortive VZV infection. Nevertheless, all animals generated a humoral and cellular immune response that conferred partial cross-protection against simian varicella virus (SVV) challenge. Additionally, VZV DNA was not detected in the sensory ganglia, suggesting that viremia might be required for the establishment of latency. Therefore, VZV vaccination of RMs followed by SVV challenge is a model that will support the development of vaccines that boost protective T cell responses against VZV.


Assuntos
Varicela/veterinária , Proteção Cruzada , Herpesvirus Humano 3/imunologia , Doenças dos Primatas/prevenção & controle , Animais , Anticorpos Antivirais/sangue , Varicela/imunologia , Varicela/patologia , Varicela/prevenção & controle , DNA Viral/genética , DNA Viral/isolamento & purificação , Gânglios/virologia , Macaca mulatta , Masculino , Doenças dos Primatas/imunologia , Linfócitos T/imunologia
6.
Arch Virol ; 161(2): 465-70, 2016 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-26590068

RESUMO

There are several dengue vaccine candidates at advanced stages of development, but none of them are licensed. Despite the reactogenicity and immunogenicity profile in humans of the tetravalent ChimeriVax™ dengue vaccine candidate, in efficacy trials, it has failed to confer complete protection against dengue virus (DENV)-1 and DENV-2. However, full protection against the four serotypes had been observed previously in monkeys immunized with this vaccine candidate. Some authors have tried to explain this contradiction by hypothesizing that protection rates in non-human primates (NHPs) are associated with a lack of post-challenge anamnestic immune responses. Here, we studied the protection and anamnestic response patterns after homologous challenge in NHPs previously infected with DENV-2. Two immunization schemes were used, varying the viral doses and the intervals between them. Animals developed immunity against DENV-2 that provided full protection against reinfection with a homologous virus. However, all monkeys showed a significant increase in antiviral and neutralizing antibody titers after challenge. Our results suggest that sterilizing immunity could not be induced by infection with the virus despite the lack of detectable viremia in some animals in which an increase in antibody titer was observed. For this reason, we propose that the lack of an anamnestic neutralizing antibody response after challenge, as suggested by some authors, should be carefully reviewed as a criterion for evaluating the functionality of vaccine candidates.


Assuntos
Vírus da Dengue/imunologia , Dengue/veterinária , Doenças dos Primatas/prevenção & controle , Animais , Anticorpos Neutralizantes/sangue , Anticorpos Antivirais/sangue , Chlorocebus aethiops , Dengue/imunologia , Dengue/prevenção & controle , Memória Imunológica
7.
J Virol ; 88(14): 8139-52, 2014 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-24807726

RESUMO

Live attenuated cold-adapted (ca) H5N1, H7N3, H6N1, and H9N2 influenza vaccine viruses replicated in the respiratory tract of mice and ferrets, and 2 doses of vaccines were immunogenic and protected these animals from challenge infection with homologous and heterologous wild-type (wt) viruses of the corresponding subtypes. However, when these vaccine candidates were evaluated in phase I clinical trials, there were inconsistencies between the observations in animal models and in humans. The vaccine viruses did not replicate well and immune responses were variable in humans, even though the study subjects were seronegative with respect to the vaccine viruses before vaccination. Therefore, we sought a model that would better reflect the findings in humans and evaluated African green monkeys (AGMs) as a nonhuman primate model. The distribution of sialic acid (SA) receptors in the respiratory tract of AGMs was similar to that in humans. We evaluated the replication of wt and ca viruses of avian influenza (AI) virus subtypes H5N1, H6N1, H7N3, and H9N2 in the respiratory tract of AGMs. All of the wt viruses replicated efficiently, while replication of the ca vaccine viruses was restricted to the upper respiratory tract. Interestingly, the patterns and sites of virus replication differed among the different subtypes. We also evaluated the immunogenicity and protective efficacy of H5N1, H6N1, H7N3, and H9N2 ca vaccines. Protection from wt virus challenge correlated well with the level of serum neutralizing antibodies. Immune responses were slightly better when vaccine was delivered by both intranasal and intratracheal delivery than when it was delivered intranasally by sprayer. We conclude that live attenuated pandemic influenza virus vaccines replicate similarly in AGMs and human subjects and that AGMs may be a useful model to evaluate the replication of ca vaccine candidates. Importance: Ferrets and mice are commonly used for preclinical evaluation of influenza vaccines. However, we observed significant inconsistencies between observations in humans and in these animal models. We used African green monkeys (AGMs) as a nonhuman primate (NHP) model for a comprehensive and comparative evaluation of pairs of wild-type and pandemic live attenuated influenza virus vaccines (pLAIV) representing four subtypes of avian influenza viruses and found that pLAIVs replicate similarly in AGMs and humans and that AGMs can be useful for evaluation of the protective efficacy of pLAIV.


Assuntos
Vírus da Influenza A/crescimento & desenvolvimento , Vírus da Influenza A/imunologia , Vacinas contra Influenza/imunologia , Infecções por Orthomyxoviridae/prevenção & controle , Doenças dos Primatas/prevenção & controle , Animais , Anticorpos Neutralizantes/sangue , Anticorpos Antivirais/sangue , Chlorocebus aethiops , Modelos Animais de Doenças , Feminino , Humanos , Vacinas contra Influenza/administração & dosagem , Influenza Humana , Masculino , Camundongos , Infecções por Orthomyxoviridae/imunologia , Pandemias , Doenças dos Primatas/imunologia , Sistema Respiratório/virologia , Vacinas Atenuadas/administração & dosagem , Vacinas Atenuadas/imunologia
8.
J Virol ; 88(22): 13418-28, 2014 11.
Artigo em Inglês | MEDLINE | ID: mdl-25210172

RESUMO

UNLABELLED: Current influenza virus vaccines primarily aim to induce neutralizing antibodies (NAbs). Modified vaccinia virus Ankara (MVA) is a safe and well-characterized vector for inducing both antibody and cellular immunity. We evaluated the immunogenicity and protective efficacy of MVA encoding influenza virus hemagglutinin (HA) and/or nucleoprotein (NP) in cynomolgus macaques. Animals were given 2 doses of MVA-based vaccines 4 weeks apart and were challenged with a 2009 pandemic H1N1 isolate (H1N1pdm) 8 weeks after the last vaccination. MVA-based vaccines encoding HA induced potent serum antibody responses against homologous H1 or H5 HAs but did not stimulate strong T cell responses prior to challenge. However, animals that received MVA encoding influenza virus HA and/or NP had high frequencies of virus-specific CD4(+) and CD8(+) T cell responses within the first 7 days of H1N1pdm infection, while animals vaccinated with MVA encoding irrelevant antigens did not. We detected little or no H1N1pdm replication in animals that received vaccines encoding H1 (homologous) HA, while a vaccine encoding NP from an H5N1 isolate afforded no protection. Surprisingly, H1N1pdm viral shedding was reduced in animals vaccinated with MVA encoding HA and NP from an H5N1 isolate. This reduced shedding was associated with cross-reactive antibodies capable of mediating antibody-dependent cellular cytotoxicity (ADCC) effector functions. Our results suggest that ADCC plays a role in cross-protective immunity against influenza. Vaccines optimized to stimulate cross-reactive antibodies with ADCC function may provide an important measure of protection against emerging influenza viruses when NAbs are ineffective. IMPORTANCE: Current influenza vaccines are designed to elicit neutralizing antibodies (NAbs). Vaccine-induced NAbs typically are effective but highly specific for particular virus strains. Consequently, current vaccines are poorly suited for preventing the spread of newly emerging pandemic viruses. Therefore, we evaluated a vaccine strategy designed to induce both antibody and T cell responses, which may provide more broadly cross-protective immunity against influenza. Here, we show in a translational primate model that vaccination with a modified vaccinia virus Ankara encoding hemagglutinin from a heterosubtypic H5N1 virus was associated with reduced shedding of a pandemic H1N1 virus challenge, while vaccination with MVA encoding nucleoprotein, an internal viral protein, was not. Unexpectedly, this reduced shedding was associated with nonneutralizing antibodies that bound H1 hemagglutinin and activated natural killer cells. Therefore, antibody-dependent cellular cytotoxicity (ADCC) may play a role in cross-protective immunity to influenza virus. Vaccines that stimulate ADCC antibodies may enhance protection against pandemic influenza virus.


Assuntos
Citotoxicidade Celular Dependente de Anticorpos , Reações Cruzadas , Portadores de Fármacos/administração & dosagem , Glicoproteínas de Hemaglutininação de Vírus da Influenza/imunologia , Vírus da Influenza A Subtipo H1N1/imunologia , Vacinas contra Influenza/imunologia , Infecções por Orthomyxoviridae/prevenção & controle , Animais , Anticorpos Antivirais/sangue , Linfócitos T CD4-Positivos/imunologia , Linfócitos T CD8-Positivos/imunologia , Modelos Animais de Doenças , Vetores Genéticos , Glicoproteínas de Hemaglutininação de Vírus da Influenza/genética , Humanos , Vírus da Influenza A Subtipo H1N1/genética , Virus da Influenza A Subtipo H5N1/imunologia , Vacinas contra Influenza/administração & dosagem , Vacinas contra Influenza/genética , Macaca fascicularis , Masculino , Doenças dos Primatas/prevenção & controle , Vacinação/métodos , Vacinas Sintéticas/administração & dosagem , Vacinas Sintéticas/genética , Vacinas Sintéticas/imunologia , Vaccinia virus/genética
9.
J Virol ; 85(6): 2878-90, 2011 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-21191005

RESUMO

The use of animal models of human cytomegalovirus (HCMV) infection is critical to refine HCMV vaccine candidates. Previous reports have demonstrated that immunization of rhesus monkeys against rhesus cytomegalovirus (RhCMV) can reduce both local and systemic replication of RhCMV following experimental RhCMV challenge. These studies used prime/boost combinations of DNA expression plasmids alone or DNA priming and boosting with either inactivated virion particles or modified vaccinia virus Ankara (MVA) expressing the same antigens. Viral outcomes included reduced RhCMV replication at the site of subcutaneous inoculation and RhCMV viremia following intravenous inoculation. Since shedding of cytomegalovirus from mucosal surfaces is critical for horizontal transmission of the virus, DNA priming/MVA boosting was evaluated for the ability to reduce oral shedding of RhCMV following subcutaneous challenge. Of six rhesus monkeys vaccinated exclusively against RhCMV glycoprotein B (gB), phosphoprotein 65 (pp65), and immediate-early 1 (IE1), half showed viral loads in saliva that were lower than those of control monkeys by 1 to 3 orders of magnitude. Further, there was a strong association of memory pp65 T cell responses postchallenge in animals exhibiting the greatest reduction in oral shedding. These results highlight the fact that a DNA/MVA vaccination regimen can achieve a notable reduction in a critical parameter of viral replication postchallenge. The recently completed clinical trial of a gB subunit vaccine in which the rate of HCMV infection was reduced by 50% in the individuals receiving the vaccine is consistent with the results of this study suggesting that additional immunogens are likely essential for maximum protection in an outbred human population.


Assuntos
Infecções por Citomegalovirus/prevenção & controle , Vacinas contra Citomegalovirus/imunologia , Doenças dos Primatas/prevenção & controle , Vacinas de DNA/imunologia , Eliminação de Partículas Virais , Animais , Infecções por Citomegalovirus/imunologia , Infecções por Citomegalovirus/virologia , Vacinas contra Citomegalovirus/administração & dosagem , Modelos Animais de Doenças , Feminino , Imunização Secundária/métodos , Macaca mulatta , Masculino , Mucosa Bucal/virologia , Doenças dos Primatas/imunologia , Doenças dos Primatas/virologia , Saliva/virologia , Vacinação/métodos , Vacinas de DNA/administração & dosagem , Carga Viral
10.
Am J Primatol ; 74(6): 518-27, 2012 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-21905063

RESUMO

In captive research environments for nonhuman primates (NHP), social housing strategies are often in conflict with protocols designed to minimize disease transmission. This is particularly true in breeding colonies, and is especially relevant when attempting to eliminate specific pathogens from a population of primates. Numerous strategies have been used to establish such specific pathogen free (SPF) breeding colonies (primarily of macaques), ranging from nursery rearing of neonates to single housing of socially reared yearlings to the rearing of infants in large social groups. All these strategies attempt to balance the effects of the chosen socialization strategy on parameters related to disease transmission, including the ultimate elimination of the target pathogens. Such strategies may affect the overall disease states of NHP breeding colonies through selective breeding processes. This can occur either by creating subpopulations of animals that do not have target diseases (SPF colonies), but may have other issues; or by creating situations in which the "best" animals are sold and the breeding colony is stocked with animals that may be more disease susceptible than those that were sold. The disease states of NHP research colonies also may be affected by selective utilization programs, in which animals removed from the breeding colony for health/behavior reasons, are preferentially chosen for use in scientific investigations. Such utilization criteria raise the question of whether ideal subjects are being chosen for use in research. Finally, captive primate colonies, where both socialization and disease states are intensely managed, may provide opportunities for those testing predictions from models of the interactions of socialization and disease transmission in the evolution of wild populations of NHP. This would be especially true for some extreme conditions of these disease ecology models, given the exceedingly high social densities and levels of pathogen control that exist in many captive nonhuman primate colonies.


Assuntos
Animais de Laboratório/virologia , Doenças dos Primatas/transmissão , Comportamento Social , Viroses/veterinária , Animais , Animais de Laboratório/psicologia , Feminino , Abrigo para Animais , Masculino , Doenças dos Primatas/prevenção & controle , Primatas , Viroses/prevenção & controle , Viroses/transmissão
11.
J Infect Dis ; 204(9): 1437-49, 2011 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-21921206

RESUMO

The prophylactic efficacy of a schistosome antigen (Sm-p80) was tested in a nonhuman primate model, the baboon. Using a total of 28 baboons, different vaccination strategies were used including recombinant Sm-p80 protein formulated in Toll-like receptor 7 and Toll-like receptor 9 agonists, and DNA priming followed by boosting with protein plus adjuvants. Recombinant protein approaches provided levels of prophylactic efficacy of 52%-58%, whereas prime-boost approaches conferred 38%-47% protection in baboons. An appropriately balanced pro-inflammatory (T-helper 17 [Th17] and Th1) and anti-inflammatory (Th2) type of response was generated; the Th1 and Th17 types of immune responses appear to be indicative of increased prophylactic efficacy. Production and expression of several cytokines (interleukin 2 [IL-2], interferon γ, IL-12α, IL-1ß, IL-6, and IL-22) were up-regulated in vaccinated animals. Human correlate studies revealed Sm-p80 reactivity with immunoglobulin G in human serum samples from schistosome-infected individuals. In addition, a complete lack of prevailing Sm-p80-specific immunoglobulin E in a high-risk or infected population was observed, thus minimizing the risk of hypersensitivity reaction following vaccination with Sm-p80 in humans. This study provided the proof of concept to move Sm-p80 forward into further preclinical development leading to human clinical trials.


Assuntos
Anticorpos Anti-Helmínticos/sangue , Antígenos de Helmintos/imunologia , Doenças Endêmicas , Schistosoma mansoni/imunologia , Esquistossomose mansoni/imunologia , Esquistossomose mansoni/prevenção & controle , Vacinas de DNA/imunologia , Animais , Antígenos de Helmintos/administração & dosagem , Citocinas/metabolismo , Modelos Animais de Doenças , Feminino , Humanos , Imunoglobulina E/sangue , Imunoglobulina G/sangue , Masculino , Papio , Doenças dos Primatas/imunologia , Doenças dos Primatas/prevenção & controle , Soro/imunologia , Vacinação/métodos , Vacinas de DNA/administração & dosagem , Vacinas de Subunidades Antigênicas/administração & dosagem , Vacinas de Subunidades Antigênicas/imunologia
12.
Hum Vaccin ; 7 Suppl: 108-14, 2011.
Artigo em Inglês | MEDLINE | ID: mdl-21263229

RESUMO

PURPOSE: BCG is not efficacious against M. tuberculosis (TB) in adult. Therefore, novel TB vaccines were established by using three kinds of animal models (cynomolgus monkey model which is the best animal model of human TB, IL-2R knock out SCID mice as a human immune model, and granulysin transgenic mouse). METHODS AND RESULTS: DNA vaccine expressing TB Hsp65 and IL-12 was delivered by the hemagglutinating virus of Japan (HVJ)-envelope. The BCG prime followed by Hsp65+IL-12/HVJ vaccine boost showed a synergistic effect in the TB-infected cynomolgus monkey (100% survival). In contrast, 33% of monkeys were alive in BCG alone group. Furthermore, the prolongation of survival period of the monkey was observed by the combination of BCG and DNA vaccine even when the boost was performed after long-term period (4month) from prime. This combination also improved the erythrocyte sedimentation rate (ESR), increased the body weight, and augmented the proliferation of PBL and IL-12 production at higher levels than BCG alone or saline. Furthermore, this vaccine exerted therapeutic efficacy in IL-2R knock out SCID-PBL/hu mice, which were transplanted with human T cells. Granulysin is an important defensive molecule expressed by human T cells and NK cells and has a cytolytic activity against microbes including Mycobacterium tuberculosis (TB) and tumors. Expression of 15kD (15K) granulysin protein and mRNA in CD8 positive T cells in the patients infected with drug sensitive (TB) or multi-drug resistant (MDR-TB) M. tuberculosis were lower than that in the healthy volunteers, suggesting that granulysin treatment might improve the tuberculous disease in human. Therefore, we established two kinds of granulysin transgenic mice (15K granulysin transgenic mice and 9K granulysin transgenic mice). It was demonstrated that 15K granulysin transgenic mice as well as 9K granulysin transgenic mice exerted in vivo anti-TB effect, including the decrease of the number of TB and augmentation of the CTL activity. These are the first findings which demonstrate in vivo effects of 15K granulysin and 9K granulysin against TB infection. Moreover, DNA vaccine expressing 15K granulysin showed a therapeutic activity against TB in mice. CONCLUSION: These data indicate that monkey, IL-2R gene-knock out SCID-PBL/hu and granulysin transgenic mice models provide useful tools for the development of novel vaccines (HVJ-Envelope/Hsp65 DNA + IL-12 DNA vaccine and granulysin vaccine) against TB.


Assuntos
Proteínas de Bactérias/imunologia , Chaperonina 60/imunologia , Mycobacterium tuberculosis/imunologia , Vacinas contra a Tuberculose/administração & dosagem , Vacinas contra a Tuberculose/imunologia , Animais , Proteínas de Bactérias/genética , Proliferação de Células , Chaperonina 60/genética , Modelos Animais de Doenças , Imunização Secundária/métodos , Interleucina-12/genética , Interleucina-12/imunologia , Leucócitos Mononucleares/imunologia , Macaca fascicularis , Camundongos , Camundongos SCID , Camundongos Transgênicos , Mycobacterium tuberculosis/genética , Doenças dos Primatas/imunologia , Doenças dos Primatas/prevenção & controle , Proteínas Recombinantes/genética , Proteínas Recombinantes/imunologia , Doenças dos Roedores/imunologia , Doenças dos Roedores/prevenção & controle , Vacinação/métodos
13.
Xenotransplantation ; 16(1): 34-44, 2009.
Artigo em Inglês | MEDLINE | ID: mdl-19243559

RESUMO

UNLABELLED: Porcine endogenous retroviruses (PERVs) released from pig tissue can infect selected human cells in vitro and therefore represent a safety risk for xenotransplantation using pig cells, tissues, or organs. Although PERVs infect cells of numerous species in vitro, attempts to establish reliable animal models failed until now. Absence of PERV transmission has been shown in first experimental and clinical xenotransplantations; however, these trials suffered from the absence of long-term exposure (transplant survival) and profound immunosuppression. METHODS: We conducted infectivity studies in rhesus monkeys, pig-tailed monkeys, and baboons under chronic immunosuppression with cyclosporine A, methylprednisolone, and the rapamycin derivative. These species were selected because they are close to the human species and PERVs can be transmitted in vitro to cells of these species. In addition, the animals received twice, a C1 esterase inhibitor to block complement activation before inoculation of PERV. In order to overcome the complications of microchimerism, animals were inoculated with high titers of cell-free PERV. In addition, to enable transmission via cell-cell contact, some animals also received virus-producing cells. For inoculation the primate cell-adapted strain PERV/5 degrees was used which is characterized by a high infectious titer. Produced on human cells, this virus does not express alpha 1,3 Gal epitopes, does not contain porcine antigens on the viral surface and is therefore less immunogenic in non-human primates compared with pig cell-derived virus. Finally, we present evidence that PERV/5 degrees productively infects cells from baboons and rhesus monkeys. RESULTS: In a follow-up period of 11 months, no antibody production against PERV and no integration of proviral DNA in blood cells was observed. Furthermore, no PERV sequences were detected in the DNA of different organs taken after necropsy. CONCLUSION: These results indicate that in a primate model, in the presence of chronic immunosuppression, neither the inoculation of cell-free nor cell-associated PERV using a virus already adapted to primate cells results in an infection; this is despite the fact that peripheral blood mononuclear cells of the same animals are infectible in vitro.


Assuntos
Retrovirus Endógenos/patogenicidade , Imunossupressores/uso terapêutico , Macaca mulatta/imunologia , Macaca nemestrina/imunologia , Papio/imunologia , Doenças dos Primatas/prevenção & controle , Infecções por Retroviridae/veterinária , Suínos/virologia , Animais , Ciclosporina/uso terapêutico , DNA Viral/sangue , Quimioterapia Combinada , Retrovirus Endógenos/genética , Feminino , Masculino , Metilprednisolona/uso terapêutico , Doenças dos Primatas/imunologia , Doenças dos Primatas/virologia , Infecções por Retroviridae/imunologia , Infecções por Retroviridae/prevenção & controle , Sirolimo/uso terapêutico , Transplante Heterólogo
14.
Rev Sci Tech ; 15(1): 267-87, 1996 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-8924710

RESUMO

The author summarises the occurrence of major diseases in wild animals maintained in captivity in South America. The epidemiology, impact and significance of the diseases are discussed, together with appropriate husbandry practices to control and prevent transmissible diseases. The following animal groups and pathologies are considered in this review: poxvirus dermatitis, gastroenteritis, pneumonia, amoebosis and coccidiosis in reptiles, management practices and diseases (including botulism, bacterial enteritis, psittacosis, aspergillosis and parasitic diseases in birds), enterocolitis, pneumonias and internal parasites in non-human primates, canine distemper, parvoviruses, babesiosis, internal and external parasites in carnivores, tuberculosis and enteritis in tapirs, haemorrhagic disease in cervids.


Assuntos
Criação de Animais Domésticos , Animais de Zoológico , Doenças Transmissíveis/veterinária , Animais , Doenças das Aves/epidemiologia , Doenças das Aves/prevenção & controle , Aves , Carnívoros , Controle de Doenças Transmissíveis , Doenças Transmissíveis/epidemiologia , Cervos , Perissodáctilos , Doenças dos Primatas/epidemiologia , Doenças dos Primatas/prevenção & controle , Primatas , Répteis , América do Sul/epidemiologia
15.
Rev Sci Tech ; 20(1): 291-303, 2001 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-11288517

RESUMO

Tuberculosis caused by Mycobacterium tuberculosis and M. tuberculosis-like organisms has been identified in a wide range of species, including non-human primates, elephants and other exotic ungulates, carnivores, marine mammals and psittacine birds. Disease associated with M. tuberculosis has occurred mostly within captive settings and does not appear to occur naturally in free-living mammals. Mycobacterium tuberculosis probably originated as an infection of humans, but from the zoonotic standpoint, non-human primates, Asian elephants and psittacine birds have the potential to transmit this disease to humans. However, the overall prevalence of disease in these susceptible species is low and documented transmissions of M. tuberculosis between animals and humans are uncommon. Mycobacterium tuberculosis causes progressive pulmonary disease in mammals and a muco-cutaneous disease in parrots. In all cases, the disease can disseminate and be shed into the environment. Diagnosis in living animals is based on intradermal tuberculin testing in non-human primates, culturing trunk secretions in elephants, and biopsy and culture of external lesions in parrots. Ancillary testing with deoxyribonucleic acid probes and nucleic acid amplification, and enzyme-linked immunosorbent assays have been adapted to some of these species with promising results. Additionally, new guidelines for controlling tuberculosis in elephants in the United States of America, and programmes for tuberculosis prevention in animal handlers have been established.


Assuntos
Animais Selvagens , Animais de Zoológico , Mycobacterium tuberculosis/isolamento & purificação , Tuberculose/veterinária , Animais , Doenças das Aves/prevenção & controle , Doenças das Aves/transmissão , Elefantes , Humanos , Mycobacterium tuberculosis/genética , Mycobacterium tuberculosis/imunologia , Prevalência , Doenças dos Primatas/prevenção & controle , Doenças dos Primatas/transmissão , Primatas , Psittaciformes , Tuberculose/diagnóstico , Tuberculose/epidemiologia , Tuberculose/transmissão , Zoonoses
16.
J Zoo Wildl Med ; 31(3): 361-9, 2000 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-11237144

RESUMO

Vitamin D deficiency rickets was diagnosed in three juvenile chimpanzees (Pan troglodytes) raised indoors under skylights and consuming only breast milk. Two cases detected early had mild but characteristic radiographic changes. More advanced disease presented with florid x-ray features of rickets and pathologic fractures, as well as hypocalcemia, hypophosphatemia, and low serum 25-hydroxyvitamin D levels. Treatment by a single injection of vitamin D2 in sesame oil (slow release) followed by daily oral supplementation with vitamin D2 corrected the condition. On the basis of experience with these cases and comparison with rickets in humans, a prevention protocol for mother-reared, inside-housed, chimpanzee juveniles was developed. Injection with slow release vitamin D2 (5,000 IU i.m. once) at 4 mo of age, followed by oral supplementation of 400 IU vitamin D2 daily until weaning, prevents rickets in juvenile chimpanzees raised indoors.


Assuntos
Doenças dos Primatas/prevenção & controle , Raquitismo/veterinária , Administração Oral , Animais , Animais de Zoológico , Osso e Ossos/diagnóstico por imagem , Cálcio/sangue , Dieta/veterinária , Suplementos Nutricionais , Ergocalciferóis/administração & dosagem , Ergocalciferóis/uso terapêutico , Feminino , Masculino , Pan troglodytes , Doenças dos Primatas/dietoterapia , Radiografia , Raquitismo/dietoterapia , Raquitismo/prevenção & controle , Óleo de Gergelim
17.
Vaccine ; 30(2): 336-41, 2012 Jan 05.
Artigo em Inglês | MEDLINE | ID: mdl-22085548

RESUMO

A prototype dengue-1 DNA vaccine was shown to be safe and immunogenic in a previous Phase 1 clinical trial. Anti-dengue-1 neutralizing antibody responses were detectable only in the group of volunteers receiving the high dose of nonadjuvanted vaccine and the antibody titers were low. Vaxfectin(®), a lipid-based adjuvant, enhances the immunogenicity of DNA vaccines. We conducted a nonhuman primate study to evaluate the effect of Vaxfectin(®) on the immunogenicity of a tetravalent dengue DNA vaccine. Animals were immunized on days 0, 28 and 84, with each immunization consisting of 3mg of Vaxfectin(®)-adjuvanted tetravalent dengue DNA vaccine. The use of Vaxfectin(®) resulted in a significant increase in anti-dengue neutralizing antibody responses against dengue-1, -3 and -4. There was little to no effect on T cell responses as measured by interferon gamma ELISPOT assay. Animals immunized with the Vaxfectin(®)-formulated tetravalent DNA vaccine showed significant protection against live dengue-2 virus challenge compared to control animals (0.75 mean days of viremia vs 3.3 days). Animals vaccinated with nonadjuvanted DNA had a mean 2.0 days of viremia. These results support further evaluation of the Vaxfectin(®)-adjuvanted tetravalent dengue DNA vaccine in a Phase 1 clinical trial.


Assuntos
Adjuvantes Imunológicos/administração & dosagem , Vacinas contra Dengue/imunologia , Dengue/veterinária , Fosfatidiletanolaminas/administração & dosagem , Doenças dos Primatas/prevenção & controle , Vacinas de DNA/imunologia , Animais , Anticorpos Neutralizantes/sangue , Anticorpos Antivirais/sangue , Dengue/prevenção & controle , Vacinas contra Dengue/administração & dosagem , Modelos Animais de Doenças , ELISPOT , Macaca mulatta , Primatas , Linfócitos T/imunologia , Vacinas de DNA/administração & dosagem , Viremia/prevenção & controle
18.
Vaccine ; 30(38): 5603-10, 2012 Aug 17.
Artigo em Inglês | MEDLINE | ID: mdl-22789506

RESUMO

We studied the replication of influenza A/California/07/09 (H1N1) wild type (CA09wt) virus in two non-human primate species and used one of these models to evaluate the immunogenicity and protective efficacy of a live attenuated cold-adapted vaccine, which contains the hemagglutinin and neuraminidase from the H1N1 wild type (wt) virus and six internal protein gene segments of the A/Ann Arbor/6/60 cold-adapted (ca) master donor virus. We infected African green monkeys (AGMs) and rhesus macaques with 2×10(6) TCID(50) of CA09wt and CA09ca influenza viruses. The virus CA09wt replicated in the upper respiratory tract of all animals but the titers in upper respiratory tract tissues of rhesus macaques were significant higher than in AGMs (mean peak titers 10(4.5) TCID(50)/g and 10(2.0) TCID(50)/g on days 4 and 2 post-infection, respectively; p<0.01). Virus replication was observed in the lungs of all rhesus macaques (10(2.0)-10(5.4) TCID(50)/g) whereas only 2 out of 4 AGMs had virus recovered from the lungs (10(2.5)-10(3.5) TCID(50)/g). The CA09ca vaccine virus was attenuated and highly restricted in replication in both AGMs and rhesus macaques. We evaluated the immunogenicity and protective efficacy of the CA09ca vaccine in rhesus macaques because CA09wt virus replicated more efficiently in this species. One or two doses of vaccine were administered intranasally and intratracheally to rhesus macaques. For the two-dose group, the vaccine was administered 4-weeks apart. Immunogenicity was assessed by measuring hemagglutination-inhibiting (HAI) antibodies in the serum and specific IgA antibodies to CA09wt virus in the nasal wash. One or two doses of the vaccine elicited a significant rise in HAI titers (range 40-320). Two doses of CA09ca elicited higher pH1N1-specific IgA titers than in the mock-immunized group (p<0.01). Vaccine efficacy was assessed by comparing titers of CA09wt challenge virus in the respiratory tract of mock-immunized and CA09ca vaccinated monkeys. Significantly lower virus titers were observed in the lungs of vaccinated animals than mock-immunized animals (p≤0.01). Our results demonstrate that AGMs and rhesus macaques support the replication of pandemic H1N1 influenza virus to different degrees and a cold-adapted pH1N1 vaccine elicits protective immunity against pH1N1 virus infection in rhesus macaques.


Assuntos
Adaptação Biológica , Vírus da Influenza A Subtipo H1N1/imunologia , Vírus da Influenza A Subtipo H1N1/fisiologia , Vacinas contra Influenza/imunologia , Replicação Viral , Administração Intranasal , Animais , Anticorpos Antivirais/análise , Sangue/imunologia , Chlorocebus aethiops , Temperatura Baixa , Feminino , Testes de Inibição da Hemaglutinação , Imunoglobulina A/análise , Vírus da Influenza A Subtipo H1N1/genética , Vírus da Influenza A Subtipo H1N1/patogenicidade , Vacinas contra Influenza/administração & dosagem , Vacinas contra Influenza/genética , Pulmão/virologia , Macaca mulatta , Masculino , Mucosa Nasal/imunologia , Infecções por Orthomyxoviridae/prevenção & controle , Doenças dos Primatas/prevenção & controle , Vírus Reordenados/genética , Vírus Reordenados/imunologia , Vírus Reordenados/fisiologia , Sistema Respiratório/virologia , Vacinas Atenuadas/administração & dosagem , Vacinas Atenuadas/genética , Vacinas Atenuadas/imunologia , Carga Viral
19.
Artigo em Inglês | MEDLINE | ID: mdl-23226683

RESUMO

Antibodies that neutralize infectivity of malaria sporozoites target the central repeat region of the circumsporozoite (CS) protein, which in Plasmodium falciparum is comprised primarily of 30-40 tandem NANP tetramer repeats. We evaluated immunogenicity of an alum-adsorbed (NANP)(6) peptide conjugated to an outer membrane protein complex (OMPC) derived from Neisseria meningitidis, a carrier protein used in a licensed Haemophilus influenzae pediatric vaccine. Mice immunized with (NANP)(6)-OMPC adsorbed to Merck's alum adjuvant (MAA), with or without Iscomatrix® as co-adjuvant, developed high levels of anti-repeat peptide antibody that inhibited in vitro invasion of human hepatoma cells by transgenic P. berghei sporozoites that express P. falciparum CS repeats (PfPb). Inhibition of sporozoite invasion in vitro correlated with in vivo resistance to challenge by the bites of PfPb-infected mosquitoes. Challenged mice had >90% reduction of hepatic stage parasites as measured by real-time PCR, and either sterile immunity, i.e., no detectable blood stage parasites, or delayed prepatent periods which indicate neutralization of a majority, but not all, sporozoites. Rhesus macaques immunized with two doses of (NANP)(6)-OMPC/MAA formulated with Iscomatrix® developed anti-repeat antibodies that persisted for ~2 years. A third dose of (NANP)(6)-OMPC/MAA+ Iscomatrix® at that time elicited strong anamnestic antibody responses. Rhesus macaque immune sera obtained post second and third dose of vaccine displayed high levels of sporozoite neutralizing activity in vitro that correlated with presence of high anti-repeat antibody titers. These preclinical studies in mice of different MHC haplotypes and a non-human primate support use of CS peptide-OMPC conjugates as a highly immunogenic platform to evaluate CS protective epitopes. Potential pre-erythrocytic vaccines can be combined with sexual blood stage vaccines as a multi-antigen malaria vaccine to block invasion and transmission of Plasmodium parasites.


Assuntos
Anticorpos Neutralizantes/sangue , Vacinas Antimaláricas/imunologia , Malária/prevenção & controle , Plasmodium falciparum/imunologia , Proteínas de Protozoários/imunologia , Vacinação/métodos , Adjuvantes Imunológicos/administração & dosagem , Compostos de Alúmen/administração & dosagem , Animais , Anticorpos Antiprotozoários/sangue , Proteínas da Membrana Bacteriana Externa/administração & dosagem , Proteínas da Membrana Bacteriana Externa/isolamento & purificação , Modelos Animais de Doenças , Feminino , Macaca mulatta , Vacinas Antimaláricas/administração & dosagem , Vacinas Antimaláricas/genética , Camundongos , Camundongos Endogâmicos BALB C , Neisseria meningitidis/química , Doenças dos Primatas/prevenção & controle , Proteínas de Protozoários/genética , Vacinas Conjugadas/administração & dosagem , Vacinas Conjugadas/genética , Vacinas Conjugadas/imunologia , Vacinas Sintéticas/administração & dosagem , Vacinas Sintéticas/genética , Vacinas Sintéticas/imunologia
20.
Vaccine ; 29(5): 873-7, 2011 Jan 29.
Artigo em Inglês | MEDLINE | ID: mdl-21115053

RESUMO

Safe and effective adjuvants are needed for many vaccines with limited commercial appeal, such as vaccines to infrequent (orphan) diseases or to neglected and poverty-related diseases. Here we found that three nonproprietary liposome formulations containing monophosphoryl lipid A each induced 3-fold to 5-fold increased titers of binding and neutralizing antibodies to anthrax protective antigen compared to aluminum hydroxide-adsorbed antigen in monkeys. All vaccinated monkeys were protected against lethal challenge with aerosolized Ames strain spores.


Assuntos
Adjuvantes Imunológicos/administração & dosagem , Vacinas contra Antraz/imunologia , Lipossomos/administração & dosagem , Adjuvantes Imunológicos/efeitos adversos , Hidróxido de Alumínio/administração & dosagem , Hidróxido de Alumínio/efeitos adversos , Animais , Antraz/prevenção & controle , Vacinas contra Antraz/efeitos adversos , Anticorpos Antibacterianos/sangue , Anticorpos Neutralizantes/sangue , Lipossomos/efeitos adversos , Macaca mulatta , Doenças dos Primatas/prevenção & controle , Análise de Sobrevida
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA