Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 36
Filtrar
2.
Front Immunol ; 12: 694959, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-34421904

RESUMEN

Avian Tembusu virus (TMUV) is a novel flavivirus causing severe egg drop and fatal encephalitis in avian in Asia. In the present study, we screened the structural and functional requirements of TMUV capsid protein (CP) for viral morphogenesis using reverse genetics methods in combination with replicon packaging assays. TMUV-CP showed dramatic functional and structural flexibility, and even though 44 residues were removed from the N-terminus, it was still capable of packaging replicon RNA; in addition, 33 residues were deleted from the C-terminus (containing nearly the entire α4-helix), and infectious particles were still produced, although α4-α4' is supposedly vital for CP dimerization and nucleocapsid formation. We further analyzed two mutants (ΔC20-43 and ΔC64-96 viruses) with relatively large deletions that still replicated well in BHK-21 cells. Our data indicate that internal deletions within CP impaired viral replication or assembly, resulting in attenuated virus proliferation in cells and attenuated virulence in duck embryos, and these deletion mutations are quite stable in cell culture. An in vivo assay indicated that both ΔC20-43 virus and ΔC64-96 virus were highly attenuated in ducklings but still immunogenic. Single-dose immunization with ΔC20-43 virus or ΔC64-96 virus could protect ducklings from a lethal challenge with good antigen clearance. Together, our data shed light on replication/assembly defective TMUV with internal deletions in CP and provide an effective approach to attenuate viral virulence in live vaccines without changing the antigen composition.


Asunto(s)
Proteínas de la Cápside/genética , Infecciones por Flavivirus/prevención & control , Flavivirus/genética , Enfermedades de las Aves de Corral/prevención & control , Eliminación de Secuencia , Vacunas Virales/genética , Ensamble de Virus/genética , Replicación Viral/genética , Animales , Proteínas de la Cápside/inmunología , Línea Celular , Cricetinae , Patos , Flavivirus/crecimiento & desarrollo , Flavivirus/inmunología , Flavivirus/patogenicidad , Infecciones por Flavivirus/inmunología , Infecciones por Flavivirus/virología , Inmunogenicidad Vacunal , Enfermedades de las Aves de Corral/inmunología , Enfermedades de las Aves de Corral/virología , Vacunación , Vacunas Vivas no Atenuadas/administración & dosificación , Vacunas Vivas no Atenuadas/genética , Vacunas Vivas no Atenuadas/inmunología , Vacunas Virales/administración & dosificación , Vacunas Virales/inmunología , Virulencia
3.
BMC Infect Dis ; 21(1): 117, 2021 Jan 26.
Artículo en Inglés | MEDLINE | ID: mdl-33499826

RESUMEN

BACKGROUND: Herpes zoster (HZ) infection of hematopoietic stem cell transplant (HSCT) patients is of clinical concern. Vaccination could help restore immunity to varicella zoster virus (VZV); however, temporal changes in immunogenicity and safety of live HZ vaccines after HSCT is still unclear. The aim of this study was to elucidate the temporal immunogenicity and safety of the HZ vaccine according to time since HSCT and to determine optimal timing of vaccination. METHODS: Live HZ vaccine was administered to patients 2-5 years or > 5 years post-HSCT. Control groups comprised patients with a hematologic malignancy who received cytotoxic chemotherapy and healthy volunteers. Humoral and cellular immunogenicity were measured using a glycoprotein enzyme-linked immunosorbent assay (gpELISA) and an interferon-γ (IFN-γ) enzyme-linked immunospot (ELISPOT) assay. Vaccine-related adverse events were also monitored. RESULTS: Fifty-six patients with hematologic malignancy (41 in the HSCT group and 15 in the chemotherapy group) along with 30 healthy volunteers were enrolled. The geometric mean fold rises (GMFRs) in humoral immune responses of the 2-5 year and > 5 year HSCT groups, and the healthy volunteer group, were comparable and significantly higher than that of the chemotherapy group (3.15, 95% CI [1.96-5.07] vs 5.05, 95% CI [2.50-10.20] vs 2.97, 95% CI [2.30-3.83] vs 1.42, 95% CI [1.08-1.86]). The GMFR of cellular immune responses was highest in the HSCT 2-5 year group and lowest in the chemotherapy group. No subject suffered clinically significant adverse events or reactivation of VZV within the follow-up period. CONCLUSION: Our findings demonstrate that a live HZ vaccine is immunogenic and safe when administered 2 years post-HSCT.


Asunto(s)
Trasplante de Células Madre Hematopoyéticas , Vacuna contra el Herpes Zóster , Herpes Zóster/prevención & control , Herpesvirus Humano 3/inmunología , Receptores de Trasplantes , Vacunas Vivas no Atenuadas , Anciano , Anticuerpos Antivirales/inmunología , Estudios de Casos y Controles , Femenino , Estudios de Seguimiento , Neoplasias Hematológicas/epidemiología , Neoplasias Hematológicas/inmunología , Neoplasias Hematológicas/terapia , Trasplante de Células Madre Hematopoyéticas/efectos adversos , Trasplante de Células Madre Hematopoyéticas/estadística & datos numéricos , Vacuna contra el Herpes Zóster/efectos adversos , Vacuna contra el Herpes Zóster/inmunología , Humanos , Inmunogenicidad Vacunal/fisiología , Masculino , Persona de Mediana Edad , Receptores de Trasplantes/estadística & datos numéricos , Resultado del Tratamiento , Vacunación/efectos adversos , Vacunación/métodos , Vacunación/estadística & datos numéricos , Vacunas Vivas no Atenuadas/efectos adversos , Vacunas Vivas no Atenuadas/inmunología
4.
Front Immunol ; 11: 1725, 2020.
Artículo en Inglés | MEDLINE | ID: mdl-33193290

RESUMEN

Background and Objectives: The live non-pathogenic Leishmania tarantolae has recently provided a promising approach as an effective vaccine candidate against experimental leishmaniasis (ILL). Here, we evaluated the immunoprotective potential of the live Iranian Lizard Leishmania mixed with CpG adjuvant against L. major infection in BALB/c mice. Methods: Four groups of female BALB/c mice were included in the study. The first and second groups received PBS and CpG, respectively. The immunized groups received 2 × 105 ILL promastigotes and the CpG-mixed ILL (ILL+CpG). Injections were performed subcutaneously in the right footpad. Three weeks later, all mice were challenged with 2 × 105 metacyclic promastigotes of Leishmania majorEGFP ; inoculation was done in the left footpad. The measurement of footpad swelling and in vivo fluorescent imaging were used to evaluate disease progress during infection course. Eight weeks after challenge, all mice were sacrificed and the cytokines levels (IFN-γ, IL-4, and IL-10) and sera antibodies concentrations (IgG2a and IgG1) using ELISA assay, nitric oxide production using Griess assay, and arginase activity in cultured splenocytes, were measured. In addition, direct fluorescent microscopy analysis and qPCR assay were used to quantify the splenic parasite burden. Result: The results showed that mice immunized with ILL+CpG were protected against the development of the dermal lesion. Moreover, they showed a significant reduction in the parasite load, in comparison to the control groups. The observed protection was associated with higher production of IFN-γ, as well as a reduction in IL-4 level. Additionally, the results demonstrated that arginase activity was decreased in ILL+CpG group compared to other groups. Conclusion: Immunization using ILL+CpG induces a protective immunity; indicating that ILL with an appropriate adjuvant would be a suitable choice for vaccination against leishmaniasis.


Asunto(s)
Adyuvantes Inmunológicos/farmacología , Leishmania major/inmunología , Vacunas contra la Leishmaniasis/farmacología , Leishmaniasis Cutánea/prevención & control , Lagartos/parasitología , Oligodesoxirribonucleótidos/farmacología , Piel/efectos de los fármacos , Vacunas Vivas no Atenuadas/farmacología , Animales , Anticuerpos Antiprotozoarios/sangre , Arginasa/metabolismo , Células Cultivadas , Citocinas/sangre , Modelos Animales de Enfermedad , Femenino , Inmunización , Inmunogenicidad Vacunal , Leishmaniasis Cutánea/sangre , Leishmaniasis Cutánea/inmunología , Leishmaniasis Cutánea/parasitología , Macrófagos/efectos de los fármacos , Macrófagos/inmunología , Macrófagos/metabolismo , Macrófagos/parasitología , Ratones Endogámicos BALB C , Carga de Parásitos , Piel/inmunología , Piel/parasitología , Bazo/efectos de los fármacos , Bazo/inmunología , Bazo/metabolismo , Bazo/parasitología , Vacunas Vivas no Atenuadas/inmunología
5.
EBioMedicine ; 62: 103132, 2020 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-33232870

RESUMEN

BACKGROUND: Due to the lack of protective immunity of humans towards the newly emerged SARS-CoV-2, this virus has caused a massive pandemic across the world resulting in hundreds of thousands of deaths. Thus, a vaccine is urgently needed to contain the spread of the virus. METHODS: Here, we describe Newcastle disease virus (NDV) vector vaccines expressing the spike protein of SARS-CoV-2 in its wild type format or a membrane-anchored format lacking the polybasic cleavage site. All described NDV vector vaccines grow to high titers in embryonated chicken eggs. In a proof of principle mouse study, the immunogenicity and protective efficacy of these NDV-based vaccines were investigated. FINDINGS: We report that the NDV vector vaccines elicit high levels of antibodies that are neutralizing when the vaccine is given intramuscularly in mice. Importantly, these COVID-19 vaccine candidates protect mice from a mouse-adapted SARS-CoV-2 challenge with no detectable viral titer and viral antigen in the lungs. INTERPRETATION: The results suggested that the NDV vector expressing either the wild type S or membrane-anchored S without the polybasic cleavage site could be used as live vector vaccine against SARS-CoV-2. FUNDING: This work is supported by an NIAID funded Center of Excellence for Influenza Research and Surveillance (CEIRS) contract, the Collaborative Influenza Vaccine Innovation Centers (CIVIC) contract, philanthropic donations and NIH grants.


Asunto(s)
Vacunas contra la COVID-19 , COVID-19 , Regulación Viral de la Expresión Génica/inmunología , Virus de la Enfermedad de Newcastle , SARS-CoV-2 , Glicoproteína de la Espiga del Coronavirus , Animales , COVID-19/genética , COVID-19/inmunología , COVID-19/prevención & control , Vacunas contra la COVID-19/genética , Vacunas contra la COVID-19/inmunología , Chlorocebus aethiops , Femenino , Ratones , Ratones Endogámicos BALB C , Virus de la Enfermedad de Newcastle/genética , Virus de la Enfermedad de Newcastle/inmunología , SARS-CoV-2/genética , SARS-CoV-2/inmunología , Glicoproteína de la Espiga del Coronavirus/genética , Glicoproteína de la Espiga del Coronavirus/inmunología , Vacunas Vivas no Atenuadas/genética , Vacunas Vivas no Atenuadas/inmunología , Células Vero
6.
Parasit Vectors ; 13(1): 415, 2020 Aug 12.
Artículo en Inglés | MEDLINE | ID: mdl-32787908

RESUMEN

BACKGROUND: Leishmaniasis is a serious health problem in some parts of the world. In spite of the many known leishmaniasis control measures, the disease has continued to increase in endemic areas, and no effective vaccine has been discovered. METHODS: In this study, Leishmania tarentulae was used as a living factory for the production of two LACK and KMP11 immunogenic antigens in the mice body, and safety profiles were investigated. The sequences of the KMP11 and LACK L. major antigens were synthesized in the pLEXSY-neo 2.1 plasmid and cloned into E. coli strain Top10, and after being linearized with the SwaI enzyme, they were transfected into the genome of L. tarentolae. The L. tarentolae-LACK/KMP11/EGFP in the stationary phase with CpG ODN as an adjuvant was used for vaccination in BALB/c mice. Vaccination was performed into the left footpad. Three weeks later, the booster was injected in the same manner. To examine the effectiveness of the injected vaccine, pathogenic L. major (MRHO/IR/75/ER) was injected into the right footpad of all mice three weeks following the booster vaccination. In order to assess humoral immunity, the levels of IgG1, and IgG2a antibodies before and 6 weeks after the challenge were studied in the groups. In addition, in order to investigate cellular immunity in the groups, the study measured IFN-γ, IL-5, TNF-α, IL-6 and IL-17 cytokines before, 3 weeks and 8 weeks after the challenge, and also the parasite load in the lymph node with real-time PCR. RESULTS: The lowest level of the parasitic load was observed in the G1 group (mice vaccinated with L. tarentolae-LACK/KMP11/EGFP with CpG) in comparison with other groups (L. tarentolae-LACK/KMP11/EGFP +non-CpG (G2); L. tarentolae-EGFP + CpG (G3, control); L. tarentolae-EGFP + non-CpG (G4, control); and mice injected with PBS (G5, control). Moreover, the evaluation of immune response showed a delayed-type hypersensitivity towards Th1. CONCLUSIONS: According to the results of this study, the live recombinant vaccine of L. tarentolae-LACK/KMP11/EGFP with the CpG adjuvant reduced the parasitic load and footpad induration in infected mice. The long-term effects of this vaccine can be evaluated in volunteers as a clinical trial in future planning.


Asunto(s)
Leishmania/inmunología , Vacunas contra la Leishmaniasis , Leishmaniasis Cutánea , Vacunas Vivas no Atenuadas , Animales , Anticuerpos Antiprotozoarios , Antígenos de Protozoos/biosíntesis , Antígenos de Protozoos/genética , Antígenos de Protozoos/inmunología , Clonación Molecular , Citocinas/metabolismo , Escherichia coli/genética , Genes Protozoarios , Inmunidad Humoral , Inmunoglobulina G/metabolismo , Leishmania/efectos de los fármacos , Leishmania/patogenicidad , Leishmania major/efectos de los fármacos , Leishmania major/inmunología , Leishmania major/patogenicidad , Vacunas contra la Leishmaniasis/biosíntesis , Vacunas contra la Leishmaniasis/inmunología , Vacunas contra la Leishmaniasis/farmacología , Leishmaniasis Cutánea/tratamiento farmacológico , Leishmaniasis Cutánea/inmunología , Leishmaniasis Cutánea/prevención & control , Glicoproteínas de Membrana/genética , Glicoproteínas de Membrana/inmunología , Ratones , Ratones Endogámicos BALB C/inmunología , Ratones Endogámicos BALB C/parasitología , Carga de Parásitos , Proteínas Protozoarias/biosíntesis , Proteínas Protozoarias/genética , Proteínas Protozoarias/inmunología , Proteínas Recombinantes/biosíntesis , Proteínas Recombinantes/inmunología , Vacunas Vivas no Atenuadas/biosíntesis , Vacunas Vivas no Atenuadas/inmunología , Vacunas Vivas no Atenuadas/farmacología , Vacunas Sintéticas/biosíntesis , Vacunas Sintéticas/inmunología , Vacunas Sintéticas/farmacología
8.
Front Immunol ; 11: 204, 2020.
Artículo en Inglés | MEDLINE | ID: mdl-32210953

RESUMEN

Although viruses and bacteria have been known as agents of diseases since 1546, 250 years went by until the first vaccines against these pathogens were developed (1796 and 1800s). In contrast, Malaria, which is a protozoan-neglected disease, has been known since the 5th century BCE and, despite 2,500 years having passed since then, no human vaccine has yet been licensed for Malaria. Additionally, no modern human vaccine is currently licensed against Visceral or Cutaneous leishmaniasis. Vaccination against Malaria evolved from the inoculation of irradiated sporozoites through the bite of Anopheles mosquitoes in 1930's, which failed to give protection, to the use of controlled human Malaria infection (CHMI) provoked by live sporozoites of Plasmodium falciparum and curtailed with specific chemotherapy since 1940's. Although the use of CHMI for vaccination was relatively efficacious, it has some ethical limitations and was substituted by the use of injected recombinant vaccines expressing the main antigens of the parasite cycle, starting in 1980. Pre-erythrocytic (PEV), Blood stage (BSV), transmission-blocking (TBV), antitoxic (AT), and pregnancy-associated Malaria vaccines are under development. Currently, the RTS,S-PEV vaccine, based on the circumsporozoite protein, is the only one that has arrived at the Phase III trial stage. The "R" stands for the central repeat region of Plasmodium (P.) falciparum circumsporozoite protein (CSP); the "T" for the T-cell epitopes of the CSP; and the "S" for hepatitis B surface antigen (HBsAg). In Africa, this latter vaccine achieved only 36.7% vaccine efficacy (VE) in 5-7 years old children and was associated with an increase in clinical cases in one assay. Therefore, in spite of 35 years of research, there is no currently licensed vaccine against Malaria. In contrast, more progress has been achieved regarding prevention of leishmaniasis by vaccine, which also started with the use of live vaccines. For ethical reasons, these were substituted by second-generation subunit or recombinant DNA and protein vaccines. Currently, there is one live vaccine for humans licensed in Uzbekistan, and four licensed veterinary vaccines against visceral leishmaniasis: Leishmune® (76-80% VE) and CaniLeish® (68.4% VE), which give protection against strong endpoints (severe disease and deaths under natural conditions), and, under less severe endpoints (parasitologically and PCR-positive cases), Leishtec® developed 71.4% VE in a low infective pressure area but only 35.7% VE and transient protection in a high infective pressure area, while Letifend® promoted 72% VE. A human recombinant vaccine based on the Nucleoside hydrolase NH36 of Leishmania (L.) donovani, the main antigen of the Leishmune® vaccine, and the sterol 24-c-methyltransferase (SMT) from L. (L.) infantum has reached the Phase I clinical trial phase but has not yet been licensed against the disease. This review describes the history of vaccine development and is focused on licensed formulations that have been used in preventive medicine. Special attention has been given to the delay in the development and licensing of human vaccines against Protozoan infections, which show high incidence worldwide and still remain severe threats to Public Health.


Asunto(s)
Leishmania donovani/inmunología , Vacunas contra la Leishmaniasis/historia , Leishmaniasis Visceral/prevención & control , Concesión de Licencias/historia , Vacunas contra la Malaria/historia , Malaria Falciparum/prevención & control , Vacunación Masiva/historia , Plasmodium falciparum/inmunología , Adulto , Animales , Niño , Preescolar , Femenino , Historia del Siglo XVII , Historia del Siglo XVIII , Historia del Siglo XIX , Historia del Siglo XX , Historia del Siglo XXI , Humanos , Vacunas contra la Leishmaniasis/inmunología , Leishmaniasis Visceral/parasitología , Leishmaniasis Visceral/veterinaria , Vacunas contra la Malaria/inmunología , Malaria Falciparum/parasitología , Vacunación Masiva/métodos , Embarazo , Vacunas Atenuadas/historia , Vacunas Atenuadas/inmunología , Vacunas Vivas no Atenuadas/historia , Vacunas Vivas no Atenuadas/inmunología , Vacunas Sintéticas/historia , Vacunas Sintéticas/inmunología
9.
J Virol Methods ; 273: 113722, 2019 11.
Artículo en Inglés | MEDLINE | ID: mdl-31422118

RESUMEN

The drawbacks of conventional inactivated Foot and Mouth Disease (FMD) vaccine, such as escaping of the virus during manufacture processes prompted researchers to explore novel types of vaccine to overcome these disadvantages. Listeria ivanovii (LI) is an intracellular microorganism that possesses immune-stimulatory properties, making it appropriate for use as a live bacterial vaccine vector. The Foot and mouth disease virus (FMDV) VP1 protein is the most immunogenic part of FMDV capsid, it has most of the antigenic sites for viral neutralization. The expression of antigen gene cassette in vitro was confirmed by Western blot analysis. Mice were able to eliminate LI△actAplcB-vp1 from the liver and spleen within few days revealed a safety of the candidate vaccine. Two doses of LI△actAplcB-vp1 with 14 days of interval were injected into mice. High levels of specific IgG antibodies and CD8+ and CD4+ T cells secreted cytokines including IFN-γ, TNF-α and IL-2 against FMDV-VP1 were achieved. Based on the obtained results, LI△actAplcB-vp1 candidate vaccine utilizing Listeria ivanovii as a live vector-based vaccine could enhance a specific cellular and humoral immune responses against the inserted FMDV-vp1 heterologous genes. LI△actAplcB-vp1 candidate vaccine could be a modern tool to overcome the disadvantages of the traditional inactivated FMD vaccine.


Asunto(s)
Proteínas de la Cápside/inmunología , Fiebre Aftosa/prevención & control , Listeria/genética , Vacunas Virales/inmunología , Animales , Anticuerpos Antivirales/sangre , Linfocitos T CD4-Positivos/inmunología , Linfocitos T CD8-positivos/inmunología , Proteínas de la Cápside/genética , Citocinas/inmunología , Femenino , Virus de la Fiebre Aftosa , Inmunoglobulina G/sangre , Ratones , Ratones Endogámicos C57BL , Vacunas de ADN/inmunología , Vacunas Vivas no Atenuadas/genética , Vacunas Vivas no Atenuadas/inmunología , Vacunas Virales/genética
10.
Front Immunol ; 10: 1689, 2019.
Artículo en Inglés | MEDLINE | ID: mdl-31447829

RESUMEN

The goals of our study were to compare the immune response to different killed and modified live vaccines against PRRS virus and to monitor the antibody production and the cell mediated immunity both at the systemic and local level. In the experiment, we immunized four groups of piglets with two commercial inactivated (A1-Progressis, A2-Suivac) and two modified live vaccines (B3-Amervac, B4-Porcilis). Twenty-one days after the final vaccination, all piglets, including the control non-immunized group (C5), were i.n., infected with the Lelystad strain of PRRS virus. The serum antibody response (IgM and IgG) was the strongest in group A1 followed by two MLV (B3 and B4) groups. Locally, we demonstrated the highest level of IgG antibodies in bronchoalveolar lavages (BALF), and saliva in group A1, whereas low IgA antibody responses in BALF and feces were detected in all groups. We have found virus neutralization antibody at DPV 21 (days post vaccination) and higher levels in all groups including the control at DPI 21 (days post infection). Positive antigen specific cell-mediated response in lymphocyte transformation test (LTT) was observed in groups B3 and B4 at DPV 7 and in group B4 at DPV 21 and in all intervals after infection. The IFN-γ producing lymphocytes after antigen stimulation were found in CD4-CD8+ and CD4+CD8+ subsets of all immunized groups 7 days after infection. After infection, there were obvious differences in virus excretion. The virus was detected in all groups of piglets in serum, saliva, and occasionally in feces at DPI 3. Significantly lower virus load was found in groups A1 and B3 at DPI 21. Negative samples appeared at DPI 21 in B3 group in saliva. It can be concluded that antibodies after immunization and infection, and the virus after infection can be detected in all the compartments monitored. Immunization with inactivated vaccine A1-Progressis induces high levels of antibodies produced both systemically and locally. Immunization with MLV-vaccines (Amervac and Porcilis) produces sufficient antibody levels and also cell-mediated immunity. After infection virus secretion gradually decreases in group B3, indicating tendency to induce sterile immunity.


Asunto(s)
Anticuerpos Antivirales/biosíntesis , Activación de Linfocitos , Virus del Síndrome Respiratorio y Reproductivo Porcino/inmunología , Vacunación , Vacunas Virales/inmunología , Animales , Porcinos , Vacunas de Productos Inactivados/inmunología , Vacunas Vivas no Atenuadas/inmunología , Carga Viral
11.
mSphere ; 4(4)2019 08 21.
Artículo en Inglés | MEDLINE | ID: mdl-31434744

RESUMEN

Vibrio cholerae infection provides long-lasting protective immunity, while oral, inactivated cholera vaccines (OCV) result in more-limited protection. To identify characteristics of the innate immune response that may distinguish natural V. cholerae infection from OCV, we stimulated differentiated, macrophage-like THP-1 cells with live versus heat-inactivated V. cholerae with and without endogenous or exogenous cholera holotoxin (CT). Interleukin 23A gene (IL23A) expression was higher in cells exposed to live V. cholerae than in cells exposed to inactivated organisms (mean change, 38-fold; 95% confidence interval [95% CI], 4.0 to 42; P < 0.01). IL-23 secretion was also higher in cells exposed to live V. cholerae than in cells exposed to inactivated V. cholerae (mean change, 5.6-fold; 95% CI, 4.4 to 11; P < 0.001). This increase in IL-23 secretion was more marked than for other key innate immune cytokines (e.g., IL-1ß and IL-6) and dependent on exposure to the combination of both live V. cholerae and CT. While IL-23 secretion was reduced following stimulation with either heat-inactivated wild-type V. cholerae or a live isogenic ctxAB mutant of V. cholerae, the addition of exogenous CT restored IL-23 secretion in combination with the live isogenic ctxAB mutant V. cholerae, but not when it was paired with stimulation by heat-inactivated V. cholerae The posttranslational regulation of IL-23 under these conditions was dependent on the activity of the cysteine protease cathepsin B. In humans, IL-23 promotes the differentiation of Th17 cells to T follicular helper cells, which maintain and support long-term memory B cell generation after infection. Based on these findings, the stimulation of IL-23 production may be a determinant of protective immunity following V. cholerae infection.IMPORTANCE An episode of cholera provides better protection against reinfection than oral cholera vaccines, and the reasons for this are still under study. To better understand this, we compared the immune responses of human cells exposed to live Vibrio cholerae with those of cells exposed to heat-killed V. cholerae (similar to the contents of oral cholera vaccines). We also compared the effects of active cholera toxin and the inactive cholera toxin B subunit (which is included in some cholera vaccines). One key immune signaling molecule, IL-23, was uniquely produced in response to the combination of live bacteria and active cholera holotoxin. Stimulation with V. cholerae that did not produce the active toxin or was killed did not produce an IL-23 response. The stimulation of IL-23 production by cholera toxin-producing V. cholerae may be important in conferring long-term immunity after cholera.


Asunto(s)
Antígenos Bacterianos/inmunología , Inmunidad Innata , Subunidad p19 de la Interleucina-23/genética , Monocitos/inmunología , Procesamiento Postranscripcional del ARN/inmunología , Vibrio cholerae/inmunología , Anticuerpos Antibacterianos/inmunología , Antígenos Bacterianos/química , Toxina del Cólera/inmunología , Vacunas contra el Cólera/inmunología , Citocinas/inmunología , Regulación de la Expresión Génica/inmunología , Calor , Humanos , Subunidad p19 de la Interleucina-23/inmunología , Monocitos/microbiología , Células THP-1 , Vacunas de Productos Inactivados/inmunología , Vacunas Vivas no Atenuadas/inmunología , Vibrio cholerae/patogenicidad
12.
Front Immunol ; 10: 923, 2019.
Artículo en Inglés | MEDLINE | ID: mdl-31156614

RESUMEN

BCG is currently the only licensed vaccine against tuberculosis (TB) and confers protection against meningitis and miliary tuberculosis in infants, although pulmonary disease protection in adults is inconsistent. Recently, promising HIV-1 immunogens were developed, such as the T-cell immunogens "tHIVconsvX," designed using functionally conserved protein regions across group M strains, with mosaic immunogens to improve HIV-1 variant match and response breadth. In this study, we constructed an integrative E. coli-mycobacterial shuttle plasmid, p2auxo.HIVconsvXint, expressing the immunogens HIVconsv1&2. This expression vector used an antibiotic resistance-free mechanism for plasmid selection and maintenance. It was first transformed into a glycine auxotrophic E. coli strain and subsequently transformed into a lysine auxotrophic Mycobacterium bovis BCG strain to generate vaccines BCG.HIVconsv12auxo.int and BCG.HIVconsv22auxo.int. The DNA sequence coding for the HIVconsv1&2 immunogens and protein expression were confirmed and working vaccine stocks were genetically and phenotypically characterized. We demonstrated that BCG.HIVconsv1&22auxo.int in combination with ChAdOx1.tHIVconsv5&6 were well tolerated and induced HIV-1-specific T-cell responses in adult BALB/c mice. In addition, we showed that the BCG.HIVconsv1&22auxo.int vaccine strains were stable in vitro after 35 bacterial generations and in vivo 7 weeks after inoculation. The use of integrative expression vectors and novel HIV-1 immunogens are likely to have improved the mycobacterial vaccine stability and specific immunogenicity and may enable the development of a useful vaccine platform for priming protective responses against HIV-1/TB and other prevalent pediatric pathogens shortly following birth.


Asunto(s)
Vacunas contra el SIDA/inmunología , Vacuna BCG/inmunología , VIH-1/inmunología , Linfocitos T/inmunología , Vacunas Sintéticas/inmunología , Animales , Antígenos Virales/inmunología , Femenino , Ratones , Ratones Endogámicos BALB C , Vacunas Vivas no Atenuadas/inmunología
13.
J Infect Dis ; 220(7): 1147-1151, 2019 08 30.
Artículo en Inglés | MEDLINE | ID: mdl-31095689

RESUMEN

Pneumonic plague, caused by Yersinia pestis, is a rapidly progressing contagious disease. In the plague mouse model, a single immunization with the EV76 live attenuated Y. pestis strain rapidly induced the expression of hemopexin and haptoglobin in the lung and serum, both of which are important in iron sequestration. Immunization against a concomitant lethal Y. pestis respiratory challenge was correlated with temporary inhibition of disease progression. Combining EV76-immunization and second-line antibiotic treatment, which are individually insufficient, led to a synergistic protective effect that represents a proof of concept for efficient combinational therapy in cases of infection with antibiotic-resistant strains.


Asunto(s)
Antibacterianos/uso terapéutico , Vacunas Bacterianas/uso terapéutico , Ceftriaxona/uso terapéutico , Peste/tratamiento farmacológico , Peste/prevención & control , Profilaxis Posexposición/métodos , Yersinia pestis/inmunología , Animales , Modelos Animales de Enfermedad , Sinergismo Farmacológico , Quimioterapia Combinada/métodos , Femenino , Haptoglobinas/análisis , Hemopexina/análisis , Hierro/metabolismo , Ratones , Ratones Endogámicos C57BL , Peste/microbiología , Resultado del Tratamiento , Vacunas Vivas no Atenuadas/inmunología
14.
J Vet Med Sci ; 81(4): 612-619, 2019 Apr 27.
Artículo en Inglés | MEDLINE | ID: mdl-30867350

RESUMEN

In this study, we evaluated antibody and cell-mediated immune (CMI) responses in the mucosal and systemic compartments and protection against challenge with a nephropathogenic Brazilian (BR-I) strain of infectious bronchitis virus (IBV) in chickens submitted to a vaccination regime comprising a priming dose of heterologous live attenuated Massachusetts vaccine followed by a booster dose of an experimental homologous inactivated vaccine two weeks later. This immunization protocol elicited significant increases in serum and lachrymal levels of anti-IBV IgG antibodies and upregulated the expression of CMI response genes, such as those encoding CD8ß chain and Granzyme homolog A in tracheal and kidney tissues at 3, 7, and 11 days post-infection in the vaccinated chickens. Additionally, vaccinated and challenged chickens showed reduced viral loads and microscopic lesion counts in tracheal and kidney tissues, and their antibody and CMI responses were negatively correlated with viral loads in the trachea and kidney. In conclusion, the combination of live attenuated vaccine containing the Massachusetts strain with a booster dose of an inactivated vaccine, containing a BR-I IBV strain, confers effective protection against infection with nephropathogenic homologous IBV strain because of the induction of consistent memory immune responses mediated by IgG antibodies and TCD8 cells in the mucosal and systemic compartments of chickens submitted to this vaccination regime.


Asunto(s)
Pollos , Infecciones por Coronavirus/inmunología , Inmunogenicidad Vacunal , Memoria Inmunológica , Virus de la Bronquitis Infecciosa , Enfermedades de las Aves de Corral/virología , Vacunas Virales/inmunología , Animales , Anticuerpos Antivirales/inmunología , Inmunidad Celular , Enfermedades de las Aves de Corral/inmunología , Vacunas Atenuadas/administración & dosificación , Vacunas Atenuadas/inmunología , Vacunas Vivas no Atenuadas/administración & dosificación , Vacunas Vivas no Atenuadas/inmunología , Vacunas Virales/administración & dosificación
15.
J Cell Physiol ; 234(4): 5196-5202, 2019 04.
Artículo en Inglés | MEDLINE | ID: mdl-30203415

RESUMEN

Vaccinations are the cornerstone of influenza prevention strategies. We carried out a meta-analysis of the messenger RNA expression profiles from recipients of trivalent inactivated vaccines (TIV) or live attenuated vaccines (LAIV) to determine the different recipients' responses to these two types of vaccines, which may provide information to improve the design of future improved vaccines. We executed meta-analysis on these datasets using a random-effects model and identified 191 and 195 differentially expressed genes in TIV and LAIV, respectively, with an false discovery rate <0.05. The genes significantly upregulated by TIV were associated with both the innate immune response and the humoral immune response, whereas LAIV mainly activated the innate immune system. The identified genes that responsible for the immune difference between LAIV and TIV might provide new information to improve current vaccines to have better efficacy in children, adults, and the elderly.


Asunto(s)
Anticuerpos Antivirales/inmunología , Inmunidad Humoral/genética , Inmunidad Innata/genética , Inmunogenicidad Vacunal/genética , Vacunas contra la Influenza/inmunología , Gripe Humana/prevención & control , Orthomyxoviridae/inmunología , Anticuerpos Antivirales/sangre , Redes Reguladoras de Genes , Interacciones Huésped-Patógeno , Humanos , Vacunas contra la Influenza/administración & dosificación , Gripe Humana/genética , Gripe Humana/inmunología , Gripe Humana/virología , Mapas de Interacción de Proteínas , Factores de Tiempo , Resultado del Tratamiento , Vacunas de Productos Inactivados/inmunología , Vacunas Vivas no Atenuadas/inmunología
16.
J Virol ; 92(17)2018 09 01.
Artículo en Inglés | MEDLINE | ID: mdl-29925653

RESUMEN

The poliovirus eradication initiative has spawned global immunization infrastructure and dramatically decreased the prevalence of the disease, yet the original virus eradication goal has not been met. The suboptimal properties of the existing vaccines are among the major reasons why the program has repeatedly missed eradication deadlines. Oral live poliovirus vaccine (OPV), while affordable and effective, occasionally causes the disease in the primary recipients, and the attenuated viruses rapidly regain virulence and can cause poliomyelitis outbreaks. Inactivated poliovirus vaccine (IPV) is safe but expensive and does not induce the mucosal immunity necessary to interrupt virus transmission. While the need for a better vaccine is widely recognized, current efforts are focused largely on improvements to the OPV or IPV, which are still beset by the fundamental drawbacks of the original products. Here we demonstrate a different design of an antipoliovirus vaccine based on in situ production of virus-like particles (VLPs). The poliovirus capsid protein precursor, together with a protease required for its processing, are expressed from a Newcastle disease virus (NDV) vector, a negative-strand RNA virus with mucosal tropism. In this system, poliovirus VLPs are produced in the cells of vaccine recipients and are presented to their immune systems in the context of active replication of NDV, which serves as a natural adjuvant. Intranasal administration of the vectored vaccine to guinea pigs induced strong neutralizing systemic and mucosal antibody responses. Thus, the vectored poliovirus vaccine combines the affordability and efficiency of a live vaccine with absolute safety, since no full-length poliovirus genome is present at any stage of the vaccine life cycle.IMPORTANCE A new, safe, and effective vaccine against poliovirus is urgently needed not only to complete the eradication of the virus but also to be used in the future to prevent possible virus reemergence in a postpolio world. Currently, new formulations of the oral vaccine, as well as improvements to the inactivated vaccine, are being explored. In this study, we designed a viral vector with mucosal tropism that expresses poliovirus capsid proteins. Thus, poliovirus VLPs are produced in vivo, in the cells of a vaccine recipient, and are presented to the immune system in the context of vector virus replication, stimulating the development of systemic and mucosal immune responses. Such an approach allows the development of an affordable and safe vaccine that does not rely on the full-length poliovirus genome at any stage.


Asunto(s)
Vectores Genéticos , Virus de la Enfermedad de Newcastle/genética , Poliomielitis/prevención & control , Vacunas contra Poliovirus/inmunología , Poliovirus/genética , Vacunas de Partículas Similares a Virus/inmunología , Animales , Anticuerpos Antivirales/sangre , Proteínas de la Cápside/genética , Proteínas de la Cápside/inmunología , Cobayas , Inmunidad Mucosa , Inmunoglobulina A/inmunología , Inmunoglobulina G/inmunología , Virus de la Enfermedad de Newcastle/inmunología , Virus de la Enfermedad de Newcastle/fisiología , Poliomielitis/inmunología , Poliomielitis/virología , Poliovirus/enzimología , Poliovirus/inmunología , Vacuna Antipolio de Virus Inactivados/administración & dosificación , Vacuna Antipolio de Virus Inactivados/efectos adversos , Vacuna Antipolio de Virus Inactivados/genética , Vacuna Antipolio de Virus Inactivados/inmunología , Vacunas contra Poliovirus/efectos adversos , Vacunas contra Poliovirus/normas , Vacunación , Vacunas Vivas no Atenuadas/administración & dosificación , Vacunas Vivas no Atenuadas/efectos adversos , Vacunas Vivas no Atenuadas/genética , Vacunas Vivas no Atenuadas/inmunología , Vacunas de Partículas Similares a Virus/administración & dosificación , Vacunas de Partículas Similares a Virus/efectos adversos , Vacunas de Partículas Similares a Virus/genética
17.
PLoS Negl Trop Dis ; 12(6): e0006511, 2018 06.
Artículo en Inglés | MEDLINE | ID: mdl-29889829

RESUMEN

BACKGROUND: To establish correlates of human immunity to the live plague vaccine (LPV), we analyzed parameters of cellular and antibody response to the plasminogen activator Pla of Y. pestis. This outer membrane protease is an essential virulence factor that is steadily expressed by Y. pestis. METHODOLOGY/PRINCIPAL FINDINGS: PBMCs and sera were obtained from a cohort of naïve (n = 17) and LPV-vaccinated (n = 34) donors. Anti-Pla antibodies of different classes and IgG subclasses were determined by ELISA and immunoblotting. The analysis of antibody response was complicated with a strong reactivity of Pla with normal human sera. The linear Pla B-cell epitopes were mapped using a library of 15-mer overlapping peptides. Twelve peptides that reacted specifically with sera of vaccinated donors were found together with a major cross-reacting peptide IPNISPDSFTVAAST located at the N-terminus. PBMCs were stimulated with recombinant Pla followed by proliferative analysis and cytokine profiling. The T-cell recall response was pronounced in vaccinees less than a year post-immunization, and became Th17-polarized over time after many rounds of vaccination. CONCLUSIONS/SIGNIFICANCE: The Pla protein can serve as a biomarker of successful vaccination with LPV. The diagnostic use of Pla will require elimination of cross-reactive parts of the antigen.


Asunto(s)
Antígenos Bacterianos/inmunología , Proteínas Bacterianas/inmunología , Inmunidad Celular , Inmunidad Humoral , Vacuna contra la Peste/inmunología , Activadores Plasminogénicos/inmunología , Yersinia pestis/inmunología , Adulto , Anciano , Biomarcadores/sangre , Citocinas/biosíntesis , Citocinas/inmunología , Epítopos de Linfocito B/inmunología , Femenino , Humanos , Inmunoglobulina G/sangre , Masculino , Persona de Mediana Edad , Peste/sangre , Peste/inmunología , Peste/microbiología , Peste/prevención & control , Células Th17/inmunología , Vacunación , Vacunas Vivas no Atenuadas/administración & dosificación , Vacunas Vivas no Atenuadas/inmunología , Factores de Virulencia
18.
Viruses ; 10(4)2018 03 31.
Artículo en Inglés | MEDLINE | ID: mdl-29614716

RESUMEN

Wild ducks are known to be able to carry avian influenza viruses over long distances and infect domestic ducks, which in their turn infect domestic chickens. Therefore, prevention of virus transmission between ducks and chickens is important to control the spread of avian influenza. Here we used a low pathogenic wild aquatic bird virus A/duck/Moscow/4182/2010 (H5N3) for prevention of highly pathogenic avian influenza virus (HPAIV) transmission between ducks and chickens. We first confirmed that the ducks orally infected with H5N1 HPAIV A/chicken/Kurgan/3/2005 excreted the virus in feces. All chickens that were in contact with the infected ducks became sick, excreted the virus, and died. However, the ducks orally inoculated with 104 50% tissue culture infective doses of A/duck/Moscow/4182/2010 and challenged 14 to 90 days later with H5N1 HPAIV did not excrete the challenge virus. All contact chickens survived and did not excrete the virus. Our results suggest that low pathogenic virus of wild aquatic birds can be used for prevention of transmission of H5N1 viruses between ducks and chickens.


Asunto(s)
Virus de la Influenza A/inmunología , Vacunas contra la Influenza/inmunología , Enfermedades de las Aves de Corral/prevención & control , Enfermedades de las Aves de Corral/transmisión , Vacunas Vivas no Atenuadas/inmunología , Esparcimiento de Virus/inmunología , Administración Oral , Animales , Animales Domésticos , Pollos , Patos , Heces/virología , Inmunización , Subtipo H5N1 del Virus de la Influenza A/inmunología , Virus de la Influenza A/clasificación , Virus de la Influenza A/patogenicidad , Vacunas contra la Influenza/administración & dosificación , Enfermedades de las Aves de Corral/mortalidad , Vacunas Vivas no Atenuadas/administración & dosificación
19.
Transbound Emerg Dis ; 65(5): 1227-1234, 2018 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-29536637

RESUMEN

The objective of this study was to determine the efficacy of a commercially available porcine reproductive and respiratory syndrome virus (PRRSV)-1 modified-live virus (MLV) vaccine against PRRSV-1 and PRRSV-2 challenge in late-term pregnancy gilts. Gilts were vaccinated with the PRRSV-1 MLV vaccine at 4 weeks prior to breeding and then challenged intranasally with PRRSV-1 or PRRSV-2 at 93 days of gestation. After PRRSV-1 challenge, vaccinated pregnant gilts had a significantly longer gestation period, significantly higher numbers of live-born and weaned piglets and a significantly lower number of stillborn piglets at birth compared to unvaccinated pregnant gilts. No significant improvement in reproductive performance was observed between vaccinated and unvaccinated pregnant gilts following PRRSV-2 challenge. Vaccinated pregnant gilts also exhibited a significantly improved reproductive performance after challenge with PRRSV-1 compared to vaccinated pregnant gilts following PRRSV-2 challenge. The PRRSV-1 MLV vaccine was able to reduce PRRSV-1 but not PRRSV-2 viremia in pregnant gilts. Vaccinated gilts also showed a significantly higher number of PRRSV-1-specific IFN-γ-secreting cells (IFN-γ-SC) compared to PRRSV-2-specific IFN-γ-SC. The data presented here suggest that the vaccination of pregnant gilts with a PRRSV-1 MLV vaccine provides good protection against PRRSV-1 but only limited protection against PRRSV-2 challenge in late-term pregnancy gilts based on improvement of reproductive performance, reduction in viremia and induction of IFN-γ-SC.


Asunto(s)
Síndrome Respiratorio y de la Reproducción Porcina/prevención & control , Virus del Síndrome Respiratorio y Reproductivo Porcino/inmunología , Complicaciones Infecciosas del Embarazo/veterinaria , Vacunación/veterinaria , Vacunas Virales/administración & dosificación , Administración Intranasal , Animales , Anticuerpos Antivirales/sangre , Femenino , Embarazo , Complicaciones Infecciosas del Embarazo/prevención & control , Reproducción , Mortinato , Porcinos , Vacunas Vivas no Atenuadas/inmunología , Vacunas Virales/inmunología , Viremia/inmunología
20.
Internist (Berl) ; 59(2): 205-212, 2018 Feb.
Artículo en Alemán | MEDLINE | ID: mdl-29230484

RESUMEN

Infectious diseases are responsible for up to 5% of fatalities even in developed countries. In addition, there is an increasing susceptibility for infections in elderly people due to physiological aging of the immune system. The principles of vaccination are based on a targeted activation of the human immune system. Principally, a distinction is made between passive immunization, i.e. the application of specific antibodies against a pathogen and active immunization. In active immunization, i.e. vaccination, weakened (attenuated) or dead pathogens or components of pathogens (antigens) are administered. After a latency period that depends on the vaccine, complete immune protection is achieved and immunity is maintained for a certain period of time. In contrast to dead vaccines, by the use of live vaccines there is always a risk for infection with the administered vaccine. In passive immunization antibodies are administered. As a rule passive immunization is carried out in persons who have had contact with an infected person and in whom no or uncertain immunity against the corresponding disease is present. Based on the recommendations of the Standing Committee on Vaccination (STIKO), influenza, pneumococcal, herpes zoster, early summer meningoencephalitis (FSME) and travel vaccines are described.


Asunto(s)
Enfermedades Transmisibles/inmunología , Vacunación/métodos , Vacuna contra el Herpes Zóster/efectos adversos , Vacuna contra el Herpes Zóster/inmunología , Humanos , Inmunidad Activa/inmunología , Inmunización Pasiva , Inmunocompetencia/inmunología , Vacunas contra la Influenza/efectos adversos , Vacunas contra la Influenza/inmunología , Infecciones Neumocócicas/inmunología , Infecciones Neumocócicas/prevención & control , Factores de Riesgo , Streptococcus pneumoniae/inmunología , Vacunas Atenuadas/administración & dosificación , Vacunas Atenuadas/inmunología , Vacunas Vivas no Atenuadas/efectos adversos , Vacunas Vivas no Atenuadas/inmunología
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...