Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 21
Filtrar
1.
Front Immunol ; 14: 1256094, 2023.
Artículo en Inglés | MEDLINE | ID: mdl-37691927

RESUMEN

The first exposure to influenza is presumed to shape the B-cell antibody repertoire, leading to preferential enhancement of the initially formed responses during subsequent exposure to viral variants. Here, we investigated whether this principle remains applicable when there are large genetic and antigenic differences between primary and secondary influenza virus antigens. Because humans usually have a complex history of influenza virus exposure, we conducted this investigation in influenza-naive cynomolgus macaques. Two groups of six macaques were immunized four times with influenza virus-like particles (VLPs) displaying either one (monovalent) or five (pentavalent) different hemagglutinin (HA) antigens derived from seasonal H1N1 (H1N1) strains. Four weeks after the final immunization, animals were challenged with pandemic H1N1 (H1N1pdm09). Although immunization resulted in robust virus-neutralizing responses to all VLP-based vaccine strains, there were no cross-neutralization responses to H1N1pdm09, and all animals became infected. No reductions in viral load in the nose or throat were detected in either vaccine group. After infection, strong virus-neutralizing responses to H1N1pdm09 were induced. However, there were no increases in virus-neutralizing titers against four of the five H1N1 vaccine strains; and only a mild increase was observed in virus-neutralizing titer against the influenza A/Texas/36/91 vaccine strain. After H1N1pdm09 infection, both vaccine groups showed higher virus-neutralizing titers against two H1N1 strains of intermediate antigenic distance between the H1N1 vaccine strains and H1N1pdm09, compared with the naive control group. Furthermore, both vaccine groups had higher HA-stem antibodies early after infection than the control group. In conclusion, immunization with VLPs displaying HA from antigenically distinct H1N1 variants increased the breadth of the immune response during subsequent H1N1pdm09 challenge, although this phenomenon was limited to intermediate antigenic variants.


Asunto(s)
Subtipo H1N1 del Virus de la Influenza A , Vacunas contra la Influenza , Gripe Humana , Animales , Humanos , Estaciones del Año , Anticuerpos Neutralizantes , Macaca fascicularis
2.
Sci Rep ; 13(1): 5074, 2023 03 28.
Artículo en Inglés | MEDLINE | ID: mdl-36977691

RESUMEN

Influenza virosomes serve as antigen delivery vehicles and pre-existing immunity toward influenza improves the immune responses toward antigens. Here, vaccine efficacy was evaluated in non-human primates with a COVID-19 virosome-based vaccine containing a low dose of RBD protein (15 µg) and the adjuvant 3M-052 (1 µg), displayed together on virosomes. Vaccinated animals (n = 6) received two intramuscular administrations at week 0 and 4 and challenged with SARS-CoV-2 at week 8, together with unvaccinated control animals (n = 4). The vaccine was safe and well tolerated and serum RBD IgG antibodies were induced in all animals and in the nasal washes and bronchoalveolar lavages in the three youngest animals. All control animals became strongly sgRNA positive in BAL, while all vaccinated animals were protected, although the oldest vaccinated animal (V1) was transiently weakly positive. The three youngest animals had also no detectable sgRNA in nasal wash and throat. Cross-strain serum neutralizing antibodies toward Wuhan-like, Alpha, Beta, and Delta viruses were observed in animals with the highest serum titers. Pro-inflammatory cytokines IL-8, CXCL-10 and IL-6 were increased in BALs of infected control animals but not in vaccinated animals. Virosomes-RBD/3M-052 prevented severe SARS-CoV-2, as shown by a lower total lung inflammatory pathology score than control animals.


Asunto(s)
COVID-19 , Vacunas contra la Influenza , Gripe Humana , Animales , Humanos , Macaca mulatta , Virosomas , SARS-CoV-2 , Receptor Toll-Like 7 , COVID-19/prevención & control , Adyuvantes Inmunológicos , Anticuerpos ampliamente neutralizantes , Vacunas contra la COVID-19 , Anticuerpos Antivirales , Anticuerpos Neutralizantes
3.
Front Immunol ; 13: 1026951, 2022.
Artículo en Inglés | MEDLINE | ID: mdl-36405682

RESUMEN

Several studies have shown that the first encounter with influenza virus shapes the immune response to future infections or vaccinations. However, a detailed analysis of the primary antibody response is lacking as this is difficult to study in humans. It is therefore not known what the frequency and dynamics of the strain-specific hemagglutinin (HA) head- and stem-directed antibody responses are directly after primary influenza virus infection. Here, sera of twelve H1N1pdm2009 influenza virus-infected cynomolgus macaques were evaluated for HA-head and HA-stem domain antibody responses. We observed an early induction of HA-stem antibody responses, which was already decreased by day 56. In contrast, responses against the HA-head domain were low early after infection and increased at later timepoint. The HA-specific B cell repertoires in each animal showed diverse VH-gene usage with preferred VH-gene and JH-gene family usage for HA-head or HA-stem B cells but a highly diverse allelic variation within the VH-usage. HA-head B cells had shorter CDRH3s and higher VH-gene somatic hyper mutation levels relative to HA-stem B cells. In conclusion, our data suggest that HA-stem antibodies are the first to react to the infection while HA-head antibodies show a delayed response, but a greater propensity to enter the germinal center and undergo affinity maturation.


Asunto(s)
Subtipo H1N1 del Virus de la Influenza A , Gripe Humana , Infecciones por Orthomyxoviridae , Humanos , Animales , Formación de Anticuerpos , Hemaglutininas , Glicoproteínas Hemaglutininas del Virus de la Influenza/genética , Anticuerpos Antivirales
4.
NPJ Vaccines ; 7(1): 54, 2022 May 18.
Artículo en Inglés | MEDLINE | ID: mdl-35585071

RESUMEN

Rift Valley fever virus (RVFV) is an emerging mosquito-borne bunyavirus that is highly pathogenic to wild and domesticated ruminants, camelids, and humans. While animals are exclusively infected via mosquito bites, humans can also be infected via contact with contaminated tissues or blood. No human vaccine is available and commercialized veterinary vaccines do not optimally combine efficacy with safety. We previously reported the development of two novel live-attenuated RVF vaccines, created by splitting the M genome segment and deleting the major virulence determinant NSs. The vaccine candidates, referred to as the veterinary vaccine vRVFV-4s and the human vaccine hRVFV-4s, were shown to induce protective immunity in multiple species after a single vaccination. Anticipating accidental exposure of humans to the veterinary vaccine and the application of hRVFV-4s to humans, the safety of each vaccine was evaluated in the most susceptible nonhuman primate model, the common marmoset (Callithrix jacchus). Marmosets were inoculated with high doses of each vaccine and were monitored for clinical signs as well as for vaccine virus dissemination, shedding, and spreading to the environment. To accurately assess the attenuation of both vaccine viruses, separate groups of marmosets were inoculated with the parent wild-type RVFV strains. Both wild-type strains induced high viremia and disseminated to primary target organs, associated with mild-to-severe morbidity. In contrast, both vaccines were well tolerated with no evidence of dissemination and shedding while inducing potent neutralizing antibody responses. The results of the studies support the unprecedented safety profile of both vaccines for animals and humans.

5.
Front Immunol ; 13: 845887, 2022.
Artículo en Inglés | MEDLINE | ID: mdl-35371043

RESUMEN

Novel safe, immunogenic, and effective vaccines are needed to control the COVID-19 pandemic, caused by SARS-CoV-2. Here, we describe the safety, robust immunogenicity, and potent efficacy elicited in rhesus macaques by a modified vaccinia virus Ankara (MVA) vector expressing a full-length SARS-CoV-2 spike (S) protein (MVA-S). MVA-S vaccination was well tolerated and induced S and receptor-binding domain (RBD)-binding IgG antibodies and neutralizing antibodies against SARS-CoV-2 and several variants of concern. S-specific IFNγ, but not IL-4, -producing cells were also elicited. After SARS-CoV-2 challenge, vaccinated animals showed a significant strong reduction of virus loads in bronchoalveolar lavages (BAL) and decreased levels in throat and nasal mucosa. Remarkably, MVA-S also protected macaques from fever and infection-induced cytokine storm. Computed tomography and histological examination of the lungs showed reduced lung pathology in MVA-S-vaccinated animals. These findings favor the use of MVA-S as a potential vaccine for SARS-CoV-2 in clinical trials.


Asunto(s)
COVID-19 , Virus Vaccinia , Animales , Anticuerpos Antivirales , COVID-19/prevención & control , Vacunas contra la COVID-19 , Humanos , Macaca mulatta , Pandemias , SARS-CoV-2/genética , Glicoproteína de la Espiga del Coronavirus , Virus Vaccinia/genética
6.
Viruses ; 13(8)2021 08 23.
Artículo en Inglés | MEDLINE | ID: mdl-34452537

RESUMEN

The post-acute phase of SARS-CoV-2 infection was investigated in rhesus (Macaca mulatta) and cynomolgus macaques (Macaca fascicularis). During the acute phase of infection, SARS-CoV-2 was shed via the nose and throat, and viral RNA was occasionally detected in feces. This phase coincided with a transient change in systemic immune activation. Even after the alleged resolution of the infection, computed tomography (CT) and positron emission tomography (PET)-CT revealed pulmonary lesions and activated tracheobronchial lymph nodes in all animals. Post-mortem histological examination of the lung tissue revealed mostly marginal or resolving minimal lesions that were indicative of SARS-CoV-2 infection. Evidence for SARS-CoV-2-induced histopathology was also found in extrapulmonary tissue samples, such as conjunctiva, cervical, and mesenteric lymph nodes. However, 5-6 weeks after SARS-CoV-2 exposure, upon necropsy, viral RNA was still detectable in a wide range of tissue samples in 50% of the macaques and included amongst others the heart, the respiratory tract and surrounding lymph nodes, salivary gland, and conjunctiva. Subgenomic messenger RNA was detected in the lungs and tracheobronchial lymph nodes, indicative of ongoing virus replication during the post-acute phase. These results could be relevant for understanding the long-term consequences of COVID-19 in humans.


Asunto(s)
COVID-19/patología , COVID-19/virología , Pulmón/patología , SARS-CoV-2/fisiología , Animales , Anticuerpos Antivirales/sangre , COVID-19/inmunología , Citocinas/sangre , Modelos Animales de Enfermedad , Humanos , Pulmón/virología , Ganglios Linfáticos/patología , Ganglios Linfáticos/fisiopatología , Macaca fascicularis , Macaca mulatta , ARN Mensajero/análisis , ARN Viral/análisis , Sistema Respiratorio/patología , Sistema Respiratorio/virología , SARS-CoV-2/inmunología , Replicación Viral
7.
Viruses ; 13(2)2021 02 22.
Artículo en Inglés | MEDLINE | ID: mdl-33671829

RESUMEN

Infection with highly pathogenic avian H5N1 influenza virus in humans often leads to severe respiratory disease with high mortality. Experimental infection in non-human primates can provide additional insight into disease pathogenesis. However, such a model should recapitulate the disease symptoms observed in humans, such as pneumonia and inflammatory cytokine response. While previous studies in macaques have demonstrated the occurrence of typical lesions in the lungs early after infection and a high level of immune activation, progression to severe disease and lethality were rarely observed. Here, we evaluated a routinely used combined route of infection via intra-bronchial, oral, and intra-nasal virus inoculation with aerosolized H5N1 exposure, with or without the regular collection of bronchoalveolar lavages early after infection. Both combined route and aerosol exposure resulted in similar levels of virus replication in nose and throat and similar levels of immune activation, cytokine, and chemokine release in the blood. However, while animals exposed to H5N1 by combined-route inoculation developed severe disease with high lethality, aerosolized exposure resulted in less lesions, as measured by consecutive computed tomography and less fever and lethal disease. In conclusion, not virus levels or immune activation, but route of infection determines fatal outcome for highly pathogenic avian H5N1 influenza infection.


Asunto(s)
Aerosoles/análisis , Bronquios/virología , Subtipo H5N1 del Virus de la Influenza A/fisiología , Gripe Humana/virología , Macaca fascicularis/virología , Boca/virología , Nariz/virología , Microbiología del Aire , Animales , Citocinas/genética , Citocinas/inmunología , Modelos Animales de Enfermedad , Exposición a Riesgos Ambientales , Humanos , Subtipo H5N1 del Virus de la Influenza A/genética , Gripe Humana/genética , Gripe Humana/inmunología , Masculino
8.
J Gen Virol ; 101(12): 1229-1241, 2020 12.
Artículo en Inglés | MEDLINE | ID: mdl-32975505

RESUMEN

Non-human primates form an important animal model for the evaluation of immunogenicity and efficacy of novel 'universal' vaccine candidates against influenza virus. However, in most studies a combination of intra-tracheal or intra-bronchial, oral and nasal virus inoculation is used with a standard virus dose of between 1 and 10 million tissue culture infective doses, which differs from typical modes of virus exposure in humans. This paper studies the systemic and local inflammatory and immune effects of aerosolized versus combined-route exposure to pandemic H1N1 influenza virus. In agreement with a previous study, both combined-route and aerosol exposure resulted in similar levels of virus replication in nose, throat and lung lavages. However, the acute release of pro-inflammatory cytokines and chemokines, acute monocyte activation in peripheral blood as well as increased cytokine production and T-cell proliferation in the lungs were only observed after combined-route infection and not after aerosol exposure. Longitudinal evaluation by computed tomography demonstrated persistence of lung lesions after resolution of the infection and a tendency for more lesions in the lower lung lobes after combined-route exposure versus upper and middle lung lobes after aerosol exposure. Computed tomography scores were observed to correlate with fever. In conclusion, influenza virus infection by aerosol exposure is accompanied by less immune-activation and inflammation in comparison with direct virus installation, despite similar levels of virus replication and development of lesions in the lungs.


Asunto(s)
Modelos Animales de Enfermedad , Subtipo H1N1 del Virus de la Influenza A , Pulmón/inmunología , Macaca fascicularis , Infecciones por Orthomyxoviridae/inmunología , Infecciones por Orthomyxoviridae/virología , Animales , Bronquios/virología , Líquido del Lavado Bronquioalveolar/inmunología , Líquido del Lavado Bronquioalveolar/virología , Citocinas/sangre , Citocinas/metabolismo , Humanos , Inmunidad Celular , Inmunidad Humoral , Subtipo H1N1 del Virus de la Influenza A/inmunología , Subtipo H1N1 del Virus de la Influenza A/fisiología , Gripe Humana/inmunología , Gripe Humana/virología , Pulmón/virología , Linfopenia , Masculino , Boca/virología , Nariz/virología , Infecciones por Orthomyxoviridae/patología , Replicación Viral , Esparcimiento de Virus
9.
J Gen Virol ; 100(5): 738-751, 2019 05.
Artículo en Inglés | MEDLINE | ID: mdl-30920368

RESUMEN

Antibodies directed against the conserved regions within the influenza A haemagglutinin (HA) protein are detected at low frequency in humans. These antibodies display a broad reactivity against divergent influenza virus strains and could potentially offer broad protection. The in vivo protective effect of these antibodies is mainly mediated through Fc receptor effector functions. While antibody-dependent phagocytosis (ADP) of anti-HA antibodies has been demonstrated in human sera and sera from influenza virus-infected macaques, it is not known whether ADP can also be induced by vaccination and what the relative strength of ADP responses is in comparison to other antibody functions. Using a cohort of influenza virus-infected and immunized macaques, we demonstrate that infection as well as immunization with DNA-encoding HA induces high-titre ADP responses against HA, which are of potency 100-1000 times higher than virus inhibitory functions including antibody-dependent cell-mediated cytotoxicity (ADCC), virus neutralization (VN) and haemagglutinin inhibition (HAI). ADP activity was equally high against HA of heterologous influenza strains of the same subtype, in contrast to virus inhibitory functions, which were all greatly diminished. ADP titres against H5, representing a hetero-subtypic virus, were much lower. ADP was measured in THP-1 cells but was also observed in primary peripheral blood monocytes and neutrophils. Furthermore, at high serum dilution enhanced infection of both monocytes and myeloid dendritic cells (mDC) was observed. Hence, influenza virus infection as well as DNA-immunization against HA can induce high-titre ADP responses that can potentially enhance influenza virus infection of primary phagocytic and dendritic cells.


Asunto(s)
Anticuerpos Antivirales/inmunología , Glicoproteínas Hemaglutininas del Virus de la Influenza/inmunología , Vacunas contra la Influenza/inmunología , Monocitos/inmunología , Orthomyxoviridae/inmunología , Fagocitosis , Vacunas de ADN/inmunología , Animales , Células Cultivadas , Células Dendríticas/inmunología , Modelos Animales de Enfermedad , Humanos , Vacunas contra la Influenza/administración & dosificación , Macaca , Neutrófilos/inmunología , Infecciones por Orthomyxoviridae/inmunología , Infecciones por Orthomyxoviridae/prevención & control , Vacunas de ADN/administración & dosificación
10.
Vaccine ; 37(6): 817-826, 2019 02 04.
Artículo en Inglés | MEDLINE | ID: mdl-30638800

RESUMEN

Conventional influenza vaccines are hampered by slow and limited production capabilities, whereas DNA vaccines can be rapidly produced for global coverage in the event of an emerging pandemic. However, a drawback of DNA vaccines is their generally low immunogenicity in non-human primates and humans. We have previously demonstrated that targeting of influenza hemagglutinin to human HLA class II molecules can increase antibody responses in larger animals such as ferrets and pigs. Here, we extend these observations by immunizing non-human primates (rhesus macaques) with a DNA vaccine encoding a bivalent fusion protein that targets influenza virus hemagglutinin (HA) to Mamu class II molecules. Such immunization induced neutralizing antibodies and antigen-specific T cells. The DNA was delivered by pain- and needle-free jet injections intradermally. No adverse effects were observed. Most importantly, the immunized rhesus macaques were protected against a challenge with influenza virus.


Asunto(s)
Anticuerpos Antivirales/sangre , Glicoproteínas Hemaglutininas del Virus de la Influenza/inmunología , Antígenos de Histocompatibilidad Clase II/inmunología , Infecciones por Orthomyxoviridae/prevención & control , Vacunas de ADN/administración & dosificación , Proteínas Virales de Fusión/inmunología , Animales , Anticuerpos Neutralizantes/sangre , Citocinas/inmunología , Subtipo H1N1 del Virus de la Influenza A , Inyecciones Intradérmicas/métodos , Inyecciones a Chorro , Macaca mulatta , Masculino , Vacunación/métodos , Vacunas de ADN/inmunología , Proteínas Virales de Fusión/administración & dosificación
11.
NPJ Vaccines ; 3: 25, 2018.
Artículo en Inglés | MEDLINE | ID: mdl-29977611

RESUMEN

Seasonal vaccines are currently the most effective countermeasure against influenza. However, seasonal vaccines are only effective against strains closely related to the influenza strains contained in the vaccine. Recently a new hemagglutinin (HA) stem-based antigen, the so-called "mini-HA", has been shown to induce a cross-protective immune response in influenza-naive mice and non-human primates (NHP). However, prior exposure to influenza can have a profound effect on the immune response to subsequent influenza infection and the protective efficacy of vaccination. Here we show that mini-HA, compared to a trivalent influenza vaccine (TIV), elicits a broadened influenza-specific humoral immune response in NHP previously exposed to influenza. Serum transfer experiments showed that antibodies induced by both mini-HA and seasonal vaccine protected mice against lethal challenge with a H1N1 influenza strain heterologous to the H1 HA included in the TIV. However, antibodies elicited by mini-HA showed an additional benefit of protecting mice against lethal heterosubtypic H5N1 influenza challenge, associated with H5 HA-specific functional antibodies.

12.
Influenza Other Respir Viruses ; 12(6): 675-686, 2018 11.
Artículo en Inglés | MEDLINE | ID: mdl-30019448

RESUMEN

BACKGROUND: The immunological response to influenza vaccine and/or natural infection is evaluated by serological techniques, the most common being hemagglutination inhibition (HI), single radial hemolysis (SRH), and virus neutralization assays, which is commonly used in a micro-neutralization (MN) format. ELISA is not officially required; however, this assay is able to measure different class-specific antibodies. The four assays identify different sets or subsets of antibodies. OBJECTIVES: The aim of this study was to establish the correlation among four serological assays using four seasonal influenza strains. METHODS: The HI, SRH, MN assays, and ELISA were performed on four seasonal influenza strains. RESULTS: A strong positive correlation was found between HI and MN and between SRH and MN assays for influenza A strains. The B strains also showed good correlations among the three assays. A positive correlation was also found between ELISA and the "classical" assays for all strains. Concerning the correlates of protection, as defined by HI ≥ 40 and SRH ≥ 25 mm2 , good agreement was observed for the influenza A strains. By contrast, the agreement for the B strains was very low. CONCLUSIONS: There is a positive strong correlation among the four serological assays for both A and B strains, especially for the HI and MN assays. There is good agreement on correlates of protection between HI and SRH assays for the A strains, but very low agreement for the B strains, suggesting higher sensitivity of SRH than HI assay in detecting antibodies against the influenza B viruses.


Asunto(s)
Anticuerpos Neutralizantes/sangre , Anticuerpos Antivirales/sangre , Inmunoensayo/métodos , Gripe Humana/inmunología , Orthomyxoviridae/inmunología , Adulto , Humanos , Sensibilidad y Especificidad
13.
J Virol ; 91(19)2017 10 01.
Artículo en Inglés | MEDLINE | ID: mdl-28701402

RESUMEN

Strategies are needed to improve the immunogenicity of HIV-1 envelope (Env) antigens (Ag) for more long-lived, efficacious HIV-1 vaccine-induced B-cell responses. HIV-1 Env gp140 (native or uncleaved molecules) or gp120 monomeric proteins elicit relatively poor B-cell responses which are short-lived. We hypothesized that Env engagement of the CD4 receptor on T-helper cells results in anergic effects on T-cell recruitment and consequently a lack of strong, robust, and durable B-memory responses. To test this hypothesis, we occluded the CD4 binding site (CD4bs) of gp140 by stable cross-linking with a 3-kDa CD4 miniprotein mimetic, serving to block ligation of gp140 on CD4+ T cells while preserving CD4-inducible (CDi) neutralizing epitopes targeted by antibody-dependent cellular cytotoxicity (ADCC) effector responses. Importantly, immunization of rhesus macaques consistently gave superior B-cell (P < 0.001) response kinetics and superior ADCC (P < 0.014) in a group receiving the CD4bs-occluded vaccine compared to those of animals immunized with gp140. Of the cytokines examined, Ag-specific interleukin-4 (IL-4) T-helper enzyme-linked immunosorbent spot (ELISpot) assays of the CD4bs-occluded group increased earlier (P = 0.025) during the inductive phase. Importantly, CD4bs-occluded gp140 antigen induced superior B-cell and ADCC responses, and the elevated B-cell responses proved to be remarkably durable, lasting more than 60 weeks postimmunization.IMPORTANCE Attempts to develop HIV vaccines capable of inducing potent and durable B-cell responses have been unsuccessful until now. Antigen-specific B-cell development and affinity maturation occurs in germinal centers in lymphoid follicles through a critical interaction between B cells and T follicular helper cells. The HIV envelope binds the CD4 receptor on T cells as soluble shed antigen or as antigen-antibody complexes, causing impairment in the activation of these specialized CD4-positive T cells. We proposed that CD4-binding impairment is partly responsible for the relatively poor B-cell responses to HIV envelope-based vaccines. To test this hypothesis, we blocked the CD4 binding site of the envelope antigen and compared it to currently used unblocked envelope protein. We found superior and durable B-cell responses in macaques vaccinated with an occluded CD4 binding site on the HIV envelope antigen, demonstrating a potentially important new direction in future design of new HIV vaccines.


Asunto(s)
Anticuerpos Neutralizantes/inmunología , Linfocitos B/inmunología , Antígenos CD4/inmunología , Anticuerpos Anti-VIH/inmunología , Macaca mulatta/inmunología , Linfocitos T Colaboradores-Inductores/inmunología , Productos del Gen env del Virus de la Inmunodeficiencia Humana/inmunología , Vacunas contra el SIDA/inmunología , Animales , Citotoxicidad Celular Dependiente de Anticuerpos/inmunología , Sitios de Unión de Anticuerpos/inmunología , VIH-1/inmunología , Macaca mulatta/virología , Vacunación
14.
J Virol ; 90(2): 1023-33, 2016 01 15.
Artículo en Inglés | MEDLINE | ID: mdl-26537681

RESUMEN

UNLABELLED: Influenza virus infection of nonhuman primates is a well-established animal model for studying pathogenesis and for evaluating prophylactic and therapeutic intervention strategies. However, usually a standard dose is used for the infection, and there is no information on the relation between challenge dose and virus replication or the induction of immune responses. Such information is also very scarce for humans and largely confined to evaluation of attenuated virus strains. Here, we have compared the effect of a commonly used dose (4 × 10(6) 50% tissue culture infective doses) versus a 100-fold-higher dose, administered by intrabronchial installation, to two groups of 6 cynomolgus macaques. Animals infected with the high virus dose showed more fever and had higher peak levels of gamma interferon in the blood. However, virus replication in the trachea was not significantly different between the groups, although in 2 out of 6 animals from the high-dose group it was present at higher levels and for a longer duration. The virus-specific antibody response was not significantly different between the groups. However, antibody enzyme-linked immunosorbent assay, virus neutralization, and hemagglutination inhibition antibody titers correlated with cumulative virus production in the trachea. In conclusion, using influenza virus infection in cynomolgus macaques as a model, we demonstrated a relationship between the level of virus production upon infection and induction of functional antibody responses against the virus. IMPORTANCE: There is only very limited information on the effect of virus inoculation dose on the level of virus production and the induction of adaptive immune responses in humans or nonhuman primates. We found only a marginal and variable effect of virus dose on virus production in the trachea but a significant effect on body temperature. The induction of functional antibody responses, including virus neutralization titer, hemagglutination inhibition titer, and antibody-dependent cell-mediated cytotoxicity, correlated with the level of virus replication measured in the trachea. The study reveals a relationship between virus production and functional antibody formation, which could be relevant in defining appropriate criteria for new influenza virus vaccine candidates.


Asunto(s)
Anticuerpos Antivirales/sangre , Formación de Anticuerpos , Subtipo H1N1 del Virus de la Influenza A/inmunología , Subtipo H1N1 del Virus de la Influenza A/fisiología , Replicación Viral , Animales , Anticuerpos Neutralizantes/sangre , Relación Dosis-Respuesta Inmunológica , Ensayo de Inmunoadsorción Enzimática , Pruebas de Inhibición de Hemaglutinación , Macaca fascicularis , Masculino , Pruebas de Neutralización , Tráquea/virología , Carga Viral
15.
PLoS One ; 10(5): e0126132, 2015.
Artículo en Inglés | MEDLINE | ID: mdl-25946071

RESUMEN

The close immunological and physiological resemblance with humans makes non-human primates a valuable model for studying influenza virus pathogenesis and immunity and vaccine efficacy against infection. Although both cynomolgus and rhesus macaques are frequently used in influenza virus research, a direct comparison of susceptibility to infection and disease has not yet been performed. In the current study a head-to-head comparison was made between these species, by using a recently described swine-origin pandemic H1N1 strain, A/Mexico/InDRE4487/2009. In comparison to rhesus macaques, cynomolgus macaques developed significantly higher levels of virus replication in the upper airways and in the lungs, involving both peak level and duration of virus production, as well as higher increases in body temperature. In contrast, clinical symptoms, including respiratory distress, were more easily observed in rhesus macaques. Expression of sialyl-α-2,6-Gal saccharides, the main receptor for human influenza A viruses, was 50 to 73 times more abundant in trachea and bronchus of cynomolgus macaques relative to rhesus macaques. The study also shows that common marmosets, a New World non-human primate species, are susceptible to infection with pandemic H1N1. The study results favor the cynomolgus macaque as model for pandemic H1N1 influenza virus research because of the more uniform and high levels of virus replication, as well as temperature increases, which may be due to a more abundant expression of the main human influenza virus receptor in the trachea and bronchi.


Asunto(s)
Callithrix/virología , Subtipo H1N1 del Virus de la Influenza A/inmunología , Subtipo H1N1 del Virus de la Influenza A/fisiología , Macaca fascicularis/inmunología , Macaca fascicularis/virología , Macaca mulatta/inmunología , Macaca mulatta/virología , Animales , Quimiocinas/sangre , Citocinas/sangre , Modelos Animales de Enfermedad , Fiebre/etiología , Especificidad del Huésped , Interacciones Huésped-Patógeno/inmunología , Humanos , Mediadores de Inflamación/sangre , Subtipo H1N1 del Virus de la Influenza A/patogenicidad , Gripe Humana/epidemiología , Gripe Humana/inmunología , Gripe Humana/virología , Pulmón/patología , Masculino , Infecciones por Orthomyxoviridae/inmunología , Infecciones por Orthomyxoviridae/patología , Infecciones por Orthomyxoviridae/virología , Pandemias , Receptores Virales/metabolismo , Porcinos/virología , Carga Viral , Virulencia/inmunología , Replicación Viral
16.
J Exp Med ; 209(6): 1091-103, 2012 Jun 04.
Artículo en Inglés | MEDLINE | ID: mdl-22641382

RESUMEN

Llamas (Lama glama) naturally produce heavy chain-only antibodies (Abs) in addition to conventional Abs. The variable regions (VHH) in these heavy chain-only Abs demonstrate comparable affinity and specificity for antigens to conventional immunoglobulins despite their much smaller size. To date, immunizations in humans and animal models have yielded only Abs with limited ability to neutralize HIV-1. In this study, a VHH phagemid library generated from a llama that was multiply immunized with recombinant trimeric HIV-1 envelope proteins (Envs) was screened directly for HIV-1 neutralization. One VHH, L8CJ3 (J3), neutralized 96 of 100 tested HIV-1 strains, encompassing subtypes A, B, C, D, BC, AE, AG, AC, ACD, CD, and G. J3 also potently neutralized chimeric simian-HIV strains with HIV subtypes B and C Env. The sequence of J3 is highly divergent from previous anti-HIV-1 VHH and its own germline sequence. J3 achieves broad and potent neutralization of HIV-1 via interaction with the CD4-binding site of HIV-1 Env. This study may represent a new benchmark for immunogens to be included in B cell-based vaccines and supports the development of VHH as anti-HIV-1 microbicides.


Asunto(s)
Anticuerpos Neutralizantes/inmunología , Camélidos del Nuevo Mundo/inmunología , VIH-1/inmunología , Animales , Afinidad de Anticuerpos , Antígenos CD4/metabolismo , Epítopos/metabolismo , Región Variable de Inmunoglobulina/genética , Región Variable de Inmunoglobulina/inmunología , Pruebas de Neutralización , Biblioteca de Péptidos , Proteínas Recombinantes/genética , Proteínas Recombinantes/inmunología , Vacunas de ADN/inmunología , Productos del Gen env del Virus de la Inmunodeficiencia Humana/genética , Productos del Gen env del Virus de la Inmunodeficiencia Humana/inmunología
17.
PLoS One ; 6(12): e28974, 2011.
Artículo en Inglés | MEDLINE | ID: mdl-22216149

RESUMEN

BACKGROUND: A vaccine is needed to control the spread of human immunodeficiency virus type 1 (HIV-1). An in vitro assay that can predict the protection induced by a vaccine would facilitate the development of such a vaccine. A potential candidate would be an assay to quantify neutralization of HIV-1. METHODS AND FINDINGS: We have used sera from rhesus macaques that have been immunized with HIV candidate vaccines and subsequently challenged with simian human immunodeficiency virus (SHIV). We compared neutralization assays with different formats. In experiments with the standardized and validated TZMbl assay, neutralizing antibody titers against homologous SHIV(SF162P4) pseudovirus gave a variable correlation with reductions in plasma viremia levels. The target cells used in the assays are not just passive indicators of virus infection but are actively involved in the neutralization process. When replicating virus was used with GHOST cell assays, events during the absorption phase, as well as the incubation phase, determine the level of neutralization. Sera that are associated with protection have properties that are closest to the traditional concept of neutralization: the concentration of antibody present during the absorption phase has no effect on the inactivation rate. In GHOST assays, events during the absorption phase may inactivate a fixed number, rather than a proportion, of virus so that while complete neutralization can be obtained, it can only be found at low doses particularly with isolates that are relatively resistant to neutralization. CONCLUSIONS: Two scenarios have the potential to predict protection by neutralizing antibodies at concentrations that can be induced by vaccination: antibodies that have properties close to the traditional concept of neutralization may protect against a range of challenge doses of neutralization sensitive HIV isolates; a window of opportunity also exists for protection against isolates that are more resistant to neutralization but only at low challenge doses.


Asunto(s)
Vacunas contra el SIDA/inmunología , Anticuerpos Neutralizantes/inmunología , VIH-1/inmunología , Animales , Cinética , Macaca mulatta
18.
J Gen Virol ; 90(Pt 4): 915-926, 2009 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-19223489

RESUMEN

Human immunodeficiency virus (HIV) infection in humans and simian immunodeficiency virus (SIV) infection in macaques are accompanied by a combined early loss of CCR5 (CD195)-expressing CD4(+) memory T cells, loss of T-helper function and T-cell hyperactivation, which have all been associated with development of high virus load and disease progression. Here, a cohort of vaccinated simian-human immunodeficiency virus strain 89.6p (SHIV(89.6p))-infected rhesus macaques, where preferential depletion of these memory T-cell subsets does not take place and CD4(+) T cells are relatively well maintained, was used to study the role of hyperactivation as an independent factor in the establishment of set-point virus load. In the acute phase of the infection, a transient loss of CD4(+) T cells, as well as strong increases in expression of proliferation and activation markers on CD4(+) and CD8(+) T cells, together with CD152 expression on CD4(+) T cells, were observed. Peak expression levels of these markers on CD4(+) T cells, but not on CD8(+) T cells, were correlated with high virus replication in the chronic phase of the infection. In addition, the peak expression level of these markers was correlated inversely with acute-phase, but not chronic-phase, HIV/SIV-specific gamma interferon responses. These data highlight a central role for an acute but transient CD4 decrease, as well as CD4(+) T-cell activation, as independent factors for prediction of set-point levels of virus replication.


Asunto(s)
Vacunas contra el SIDA/administración & dosificación , Antígenos CD/metabolismo , Linfocitos T CD4-Positivos/inmunología , Infecciones por VIH , Vacunas contra el SIDAS/administración & dosificación , Síndrome de Inmunodeficiencia Adquirida del Simio , Regulación hacia Arriba , Replicación Viral , Enfermedad Aguda , Animales , Antígeno CTLA-4 , VIH/inmunología , VIH/patogenicidad , Infecciones por VIH/inmunología , Infecciones por VIH/prevención & control , Infecciones por VIH/virología , Humanos , Interferón gamma/metabolismo , Activación de Linfocitos/inmunología , Macaca , Síndrome de Inmunodeficiencia Adquirida del Simio/inmunología , Síndrome de Inmunodeficiencia Adquirida del Simio/prevención & control , Síndrome de Inmunodeficiencia Adquirida del Simio/virología , Virus de la Inmunodeficiencia de los Simios/inmunología , Virus de la Inmunodeficiencia de los Simios/patogenicidad
19.
Virology ; 382(2): 217-25, 2008 Dec 20.
Artículo en Inglés | MEDLINE | ID: mdl-18947849

RESUMEN

Immune correlates of vaccine protection from HIV-1 infection would provide important milestones to guide HIV-1 vaccine development. In a proof of concept study using mucosal priming and systemic boosting, the titer of neutralizing antibodies in sera was found to correlate with protection of mucosally exposed rhesus macaques from SHIV infection. Mucosal priming consisted of two sequential immunizations at 12-week intervals with replicating host range mutants of adenovirus type 5 (Ad5hr) expressing the HIV-1(89.6p) env gene. Following boosting with either heterologous recombinant protein or alphavirus replicons at 12-week intervals animals were intrarectally exposed to infectious doses of the CCR5 tropic SHIV(SF162p4). Heterologous mucosal prime systemic boost immunization elicited neutralizing antibodies (Nabs), antibody-dependent cytotoxicity (ADCC), and specific patterns of antibody binding to envelope peptides. Vaccine induced protection did not correlate with the type of boost nor T-cell responses, but rather with the Nab titer prior to exposure.


Asunto(s)
Vacunas contra el SIDA/administración & dosificación , Anticuerpos Antivirales/biosíntesis , Anticuerpos Anti-VIH/biosíntesis , VIH-1/inmunología , Vacunas contra el SIDAS/administración & dosificación , Virus de la Inmunodeficiencia de los Simios/inmunología , Animales , Secuencia de Bases , ADN Viral/análisis , ADN Viral/genética , Femenino , Genes env , Infecciones por VIH/inmunología , Infecciones por VIH/prevención & control , Infecciones por VIH/virología , VIH-1/genética , Inmunidad Celular , Inmunidad Mucosa , Esquemas de Inmunización , Inmunización Secundaria , Macaca mulatta , Masculino , Pruebas de Neutralización , Síndrome de Inmunodeficiencia Adquirida del Simio/inmunología , Síndrome de Inmunodeficiencia Adquirida del Simio/prevención & control , Síndrome de Inmunodeficiencia Adquirida del Simio/virología , Virus de la Inmunodeficiencia de los Simios/genética , Viremia/inmunología , Viremia/prevención & control
20.
J Gen Virol ; 89(Pt 2): 540-5533, 2008 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-18198386

RESUMEN

Current data suggest that prophylactic human immunodeficiency virus type 1 (HIV) vaccines will be most efficacious if they elicit a combination of adaptive humoral and T-cell responses. Here, we explored the use of different vaccine strategies in heterologous prime-boost regimes and evaluated the breadth and nature of immune responses in rhesus monkeys induced by epidermally delivered plasmid DNA or recombinant HIV proteins formulated in the AS02A adjuvant system. These immunogens were administered alone or as either prime or boost in mixed-modality regimes. DNA immunization alone induced cell-mediated immune (CMI) responses, with a strong bias towards Th1-type cytokines, and no detectable antibodies to the vaccine antigens. Whenever adjuvanted protein was used as a vaccine, either alone or in a regime combined with DNA, high-titre antibody responses to all vaccine antigens were detected in addition to strong Th1- and Th2-type CMI responses. As the vaccine antigens included HIV-1 Env, Nef and Tat, as well as simian immunodeficiency virus (SIV)mac239 Nef, the animals were subsequently exposed to a heterologous, pathogenic simian-human immunodeficiency virus (SHIV)89.6p challenge. Protection against sustained high virus load was observed to some degree in all vaccinated groups. Suppression of virus replication to levels below detection was observed most frequently in the group immunized with protein followed by DNA immunization, and similarly in the group immunized with DNA alone. Interestingly, control of virus replication was associated with increased SIV Nef- and Gag-specific gamma interferon responses observed immediately following challenge.


Asunto(s)
Vacunas contra el SIDA/inmunología , Productos del Gen env/metabolismo , Productos del Gen tat/metabolismo , VIH/inmunología , VIH/metabolismo , Proteínas del Virus de la Inmunodeficiencia Humana/administración & dosificación , Virus de la Inmunodeficiencia de los Simios/fisiología , Animales , Anticuerpos Antivirales/inmunología , Anticuerpos Antivirales/farmacología , Productos del Gen env/genética , Productos del Gen env/inmunología , Productos del Gen tat/genética , Productos del Gen tat/inmunología , VIH/genética , Proteínas del Virus de la Inmunodeficiencia Humana/genética , Humanos , Inmunización , Macaca mulatta , Vacunas de ADN/inmunología , Vacunas de Subunidad/inmunología , Viremia , Replicación Viral
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA