Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 55
Filtrar
Mais filtros

Medicinas Complementares
Métodos Terapêuticos e Terapias MTCI
Base de dados
Tipo de documento
Intervalo de ano de publicação
1.
Methods Mol Biol ; 2225: 241-255, 2021.
Artigo em Inglês | MEDLINE | ID: mdl-33108667

RESUMO

Systemic lupus erythematosus (SLE) is a multifactorial and heterogeneous autoimmune disease involving multiple organ systems and tissues. Lupus nephritis occurs in approximately 60% of patients with SLE and is the leading cause of morbidity. Diffuse alveolar hemorrhage (DAH) is a rare but very serious complication of SLE with a greater than 50% associated mortality. The etiology of SLE is unclear but has proposed genetic, hormonal, and environmental aspects. Pristane is a saturated terpenoid alkane and has become the most popular laboratory model for inducing lupus in mice. The pristane model of SLE has the capacity to reproduce many components of the human presentation of the disease. Previous studies have demonstrated that virus-derived immune-modulating proteins have the potential to control inflammatory and autoimmune disorders. Serp-1, a 55 kDa secreted and highly glycosylated immune modulator derived from myxoma virus (MYXV), has potent immunomodulatory activity in models of vasculitis, viral sepsis, collagen-induced arthritis, and transplant rejection. This chapter describes the mouse preclinical pristane lupus model as a method to examine virus-derived protein efficacy for treating autoimmune diseases and specifically lupus nephritis and DAH.


Assuntos
Avaliação Pré-Clínica de Medicamentos/métodos , Hemorragia/prevenção & controle , Fatores Imunológicos/farmacologia , Nefrite Lúpica/tratamento farmacológico , Myxoma virus/química , Proteinúria/tratamento farmacológico , Proteínas Virais/farmacologia , Animais , Autoanticorpos/biossíntese , Citocinas/biossíntese , Modelos Animais de Doenças , Feminino , Hemorragia/imunologia , Hemorragia/patologia , Humanos , Fatores Imunológicos/imunologia , Injeções Intraperitoneais , Pulmão/irrigação sanguínea , Pulmão/efeitos dos fármacos , Pulmão/patologia , Nefrite Lúpica/induzido quimicamente , Nefrite Lúpica/imunologia , Nefrite Lúpica/patologia , Camundongos , Camundongos Endogâmicos BALB C , Proteinúria/induzido quimicamente , Proteinúria/imunologia , Proteinúria/patologia , Terpenos/administração & dosagem , Resultado do Tratamento , Proteínas Virais/imunologia
2.
Nature ; 581(7808): 252-255, 2020 05.
Artigo em Inglês | MEDLINE | ID: mdl-32415276

Assuntos
Antivirais/farmacologia , Betacoronavirus/química , Betacoronavirus/imunologia , Desenho de Fármacos , Proteínas Virais/antagonistas & inibidores , Proteínas Virais/química , Vacinas Virais , Monofosfato de Adenosina/análogos & derivados , Monofosfato de Adenosina/farmacologia , Monofosfato de Adenosina/uso terapêutico , Alanina/análogos & derivados , Alanina/farmacologia , Alanina/uso terapêutico , Enzima de Conversão de Angiotensina 2 , Animais , Antivirais/química , Azóis/farmacologia , Betacoronavirus/efeitos dos fármacos , Betacoronavirus/enzimologia , Vacinas contra COVID-19 , China , Proteases 3C de Coronavírus , Infecções por Coronavirus/imunologia , Infecções por Coronavirus/prevenção & controle , Proteases Semelhantes à Papaína de Coronavírus , RNA-Polimerase RNA-Dependente de Coronavírus , Microscopia Crioeletrônica , Cristalização , Cristalografia por Raios X , Cisteína Endopeptidases/química , Cisteína Endopeptidases/metabolismo , Avaliação Pré-Clínica de Medicamentos , Alemanha , Ensaios de Triagem em Larga Escala , Humanos , Isoindóis , Camundongos , National Institutes of Health (U.S.)/economia , National Institutes of Health (U.S.)/organização & administração , Compostos Organosselênicos/farmacologia , Peptidil Dipeptidase A/química , Peptidil Dipeptidase A/metabolismo , Inibidores de Proteases/farmacologia , RNA Polimerase Dependente de RNA/antagonistas & inibidores , RNA Polimerase Dependente de RNA/química , RNA Polimerase Dependente de RNA/metabolismo , SARS-CoV-2 , Glicoproteína da Espícula de Coronavírus/química , Glicoproteína da Espícula de Coronavírus/metabolismo , Síncrotrons , Fatores de Tempo , Reino Unido , Estados Unidos , Proteínas não Estruturais Virais/antagonistas & inibidores , Proteínas não Estruturais Virais/química , Proteínas não Estruturais Virais/metabolismo , Proteínas Virais/imunologia
3.
Vet Microbiol ; 242: 108604, 2020 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-32122610

RESUMO

Here, we examined the efficacy of are combinant subunit antigen-based oral vaccine for preventing porcine epidemic diarrhea virus (PEDV). First, we generated a soluble recombinant partial spike S1 protein (aP2) from PEDV in E. coli and then evaluated the utility of aP2 subunit vaccine-loaded hydroxypropyl methylcellulose phthalate microspheres (HPMCP) and RANKL-secreting L. lactis (LLRANKL) as a candidate oral vaccine in pregnant sows. Pregnant sows were vaccinated twice (with a 2 week interval between doses) at 4 weeks before farrowing. Titers of virus-specific IgA antibodies in colostrum, and neutralizing antibodies in serum, of sows vaccinated with HPMCP (aP2) plus LL RANKL increased significantly at 4 weeks post-first vaccination. Furthermore, the survival rate of newborn suckling piglets delivered by sows vaccinated with HPMCP (aP2) plus LL RANKL was similar to that of piglets delivered by sows vaccinated with a commercial killed porcine epidemic diarrhea virus (PED) vaccine. The South Korean government promotes a PED vaccine program (live-killed-killed) to increase the titers of IgA and IgG antibodies in pregnant sows and prevent PEDV. The oral vaccine strategy described herein, which is based on a safe and efficient recombinant subunit antigen, is an alternative PED vaccination strategy that could replace the traditional strategy, which relies on attenuated live oral vaccines or artificial infection with virulent PEDV.


Assuntos
Infecções por Coronavirus/veterinária , Lactobacillus/imunologia , Metilcelulose/análogos & derivados , Ligante RANK/imunologia , Doenças dos Suínos/prevenção & controle , Vacinas Virais/imunologia , Administração Oral , Animais , Anticorpos Neutralizantes/sangue , Anticorpos Antivirais/sangue , Colostro/imunologia , Infecções por Coronavirus/prevenção & controle , Feminino , Metilcelulose/administração & dosagem , Microesferas , Vírus da Diarreia Epidêmica Suína , Gravidez , Ligante RANK/administração & dosagem , Suínos , Doenças dos Suínos/virologia , Vacinas de Subunidades Antigênicas/administração & dosagem , Vacinas de Subunidades Antigênicas/imunologia , Proteínas Virais/genética , Proteínas Virais/imunologia , Vacinas Virais/administração & dosagem
4.
Viruses ; 10(4)2018 04 01.
Artigo em Inglês | MEDLINE | ID: mdl-29614779

RESUMO

An efficacious HIV-1 vaccine is regarded as the best way to halt the ongoing HIV-1 epidemic. However, despite significant efforts to develop a safe and effective vaccine, the modestly protective RV144 trial remains the only efficacy trial to provide some level of protection against HIV-1 acquisition. This review will outline the history of HIV vaccine development, novel technologies being applied to HIV vaccinology and immunogen design, as well as the studies that are ongoing to advance our understanding of vaccine-induced immune correlates of protection.


Assuntos
Vacinas contra a AIDS/imunologia , Infecções por HIV/prevenção & controle , Infecções por HIV/terapia , HIV-1/imunologia , Vacinas contra a AIDS/administração & dosagem , Animais , Anticorpos Neutralizantes/sangue , Anticorpos Neutralizantes/imunologia , Ensaios Clínicos como Assunto , Avaliação Pré-Clínica de Medicamentos , Anticorpos Anti-HIV/sangue , Anticorpos Anti-HIV/imunologia , Infecções por HIV/transmissão , HIV-1/genética , Interações Hospedeiro-Patógeno/imunologia , Humanos , Imunogenicidade da Vacina , Avaliação de Resultados em Cuidados de Saúde , Relação Estrutura-Atividade , Vacinação , Proteínas Virais/química , Proteínas Virais/genética , Proteínas Virais/imunologia
5.
Plant Biotechnol J ; 16(2): 572-580, 2018 02.
Artigo em Inglês | MEDLINE | ID: mdl-28710796

RESUMO

The global Zika virus (ZIKV) outbreak and its link to foetal and newborn microcephaly and severe neurological complications in adults call for the urgent development of ZIKV vaccines. In response, we developed a subunit vaccine based on the ZIKV envelope (E) protein and investigated its immunogenicity in mice. Transient expression of ZIKV E (zE) resulted in its rapid accumulation in leaves of Nicotiana benthamiana plants. Biochemical analysis revealed that plant-produced ZIKV E (PzE) exhibited specific binding to a panel of monoclonal antibodies that recognize various zE conformational epitopes. Furthermore, PzE can be purified to >90% homogeneity with a one-step Ni2+ affinity chromatography process. PzE are found to be highly immunogenic, as two doses of PzE elicited both potent zE-specific antibody and cellular immune responses in mice. The delivery of PzE with alum induced a mixed Th1/Th2 immune response, as the antigen-specific IgG isotypes were a mixture of high levels of IgG1/IgG2c and splenocyte cultures from immunized mice secreted significant levels of IFN-gamma, IL-4 and IL-6. Most importantly, the titres of zE-specific and neutralizing antibodies exceeded the threshold that correlates with protective immunity against multiple strains of ZIKV. Thus, our results demonstrated the feasibility of plant-produced ZIKV protein antigen as effective, safe and affordable vaccines against ZIKV.


Assuntos
Infecção por Zika virus/imunologia , Zika virus/imunologia , Zika virus/patogenicidade , Animais , Anticorpos Neutralizantes/imunologia , Camundongos , Proteínas do Envelope Viral/imunologia , Proteínas Virais/imunologia
6.
Mol Biotechnol ; 59(11-12): 469-481, 2017 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-28921459

RESUMO

Large quantities of potato leafroll virus (PLRV) antigen are difficult to obtain because this virus accumulates in plants at a low titer. To overcome this problem, we constructed a binary vector containing chimeric cDNA, in which the coat protein (CP) gene of the crucifer infecting tobacco mosaic virus (crTMV) was substituted for the coat protein gene of PLRV. The PLRV movement protein (MP) gene, which overlaps completely with the CP gene, was doubly mutated to eliminate priming of the PLRV MP translation from ATG codons with no changes to the amino acid sequence of the CP. The untranslated long intergenic region located upstream of the CP gene was removed from the construct. Transcribed powerful tobamovirus polymerase of the produced vector synthesized PLRV CP gene that was, in turn, translated into the protein. CP PLRV packed RNAs from the helical crTMV in spherical virions. Morphology, size and antigenic specificities of the wild-type and chimeric virus were similar. The yield of isolated chimera was about three orders higher than the yield of native PLRV. The genetic manipulations facilitated the generation of antibodies against the chimeric virus, which recognize the wild-type PLRV.


Assuntos
Antígenos Virais/imunologia , Luteoviridae/imunologia , Nicotiana/imunologia , Plantas Geneticamente Modificadas/imunologia , Solanum tuberosum/imunologia , Vírus do Mosaico do Tabaco/imunologia , Proteínas Virais/imunologia , Antígenos Virais/genética , Genoma Viral , Luteoviridae/genética , Plantas Geneticamente Modificadas/genética , Plantas Geneticamente Modificadas/virologia , Biossíntese de Proteínas , RNA Viral , Solanum tuberosum/genética , Solanum tuberosum/virologia , Nicotiana/genética , Nicotiana/virologia , Vírus do Mosaico do Tabaco/genética , Proteínas Virais/genética , Vírion/genética , Vírion/imunologia
7.
J Virol Methods ; 242: 35-45, 2017 04.
Artigo em Inglês | MEDLINE | ID: mdl-28025125

RESUMO

There is an urgent need for Ebola virus (EBOV) proteins, EBOV-specific antibodies and recombinant antigens to be used in diagnostics and as potential vaccine candidates. Our objective was to produce and purify recombinant proteins for immunological assays and for the production of polyclonal EBOV specific antibodies. In addition, a limited comparison of the adjuvant effects of Freund's complete adjuvant (FCA) and adjuvant system 03 (AS03) was carried out. Recombinant EBOV GST-VP24, -VP30, -VP35, -VP40 and -NP were produced in E. coli and purified with affinity chromatography followed by preparative gel electrophoresis. Recombinant EBOV GP-His was produced in Sf9 insect cells and purified by preparative gel electrophoresis. To compare the adjuvant effect of FCA and AS03, 12 rabbits were immunized four times with one of the six recombinant EBOV proteins using FCA or AS03. In addition, three guinea pigs were immunized with EBOV VP24 using FCA. With the exception of sera from two rabbits immunized with GST-VP24, the antisera against all other EBOV proteins showed very high and specific antibody responses after three to four immunizations. The adjuvant effect of AS03 was comparable to that of FCA. The produced antibodies recognized the corresponding EBOV proteins in wild type EBOV-infected cells.


Assuntos
Adjuvantes Imunológicos , Ebolavirus/imunologia , Adjuvante de Freund , Polissorbatos , Esqualeno , Proteínas Virais/imunologia , Proteínas Virais/isolamento & purificação , alfa-Tocoferol , Animais , Anticorpos Antivirais/biossíntese , Anticorpos Antivirais/sangue , Anticorpos Antivirais/imunologia , Baculoviridae/genética , Combinação de Medicamentos , Ebolavirus/química , Ebolavirus/isolamento & purificação , Adjuvante de Freund/provisão & distribuição , Cobaias , Coelhos , Proteínas Recombinantes/imunologia , Proteínas Recombinantes/isolamento & purificação , Proteínas Virais/genética
8.
mBio ; 7(4)2016 08 02.
Artigo em Inglês | MEDLINE | ID: mdl-27486196

RESUMO

UNLABELLED: Obesity is a risk factor for developing severe influenza virus infection, making vaccination of utmost importance for this high-risk population. However, vaccinated obese animals and adults have decreased neutralizing antibody responses. In these studies, we tested the hypothesis that the addition of either alum or a squalene-based adjuvant (AS03) to an influenza vaccine would improve neutralizing antibody responses and protect obese mice from challenge. Our studies demonstrate that adjuvanted vaccine does increase both neutralizing and nonneutralizing antibody levels compared to vaccine alone. Although obese mice mount significantly decreased virus-specific antibody responses, both the breadth and the magnitude of the responses against hemagglutinin (HA) and neuraminidase (NA) are decreased compared to the responses in lean mice. Importantly, even with a greater than fourfold increase in neutralizing antibody levels, obese mice are not protected against influenza virus challenge and viral loads remain elevated in the respiratory tract. Increasing the antigen dose affords no added protection, and a decreasing viral dose did not fully mitigate the increased mortality seen in obese mice. Overall, these studies highlight that, while the use of an adjuvant does improve seroconversion, vaccination does not fully protect obese mice from influenza virus challenge, possibly due to the increased sensitivity of obese animals to infection. Given the continued increase in the global obesity epidemic, our findings have important implications for public health. IMPORTANCE: Vaccination is the most effective strategy for preventing influenza virus infection and is a key component for pandemic preparedness. However, vaccines may fail to provide optimal protection in high-risk groups, including overweight and obese individuals. Given the worldwide obesity epidemic, it is imperative that we understand and improve vaccine efficacy. No work to date has investigated whether adjuvants increase the protective capacity of influenza vaccines in the obese host. In these studies, we show that adjuvants increased the neutralizing and nonneutralizing antibody responses during vaccination of lean and obese mice to levels considered "protective," and yet, obese mice still succumbed to infection. This vulnerability is likely due to a combination of factors, including the increased susceptibility of obese animals to develop severe and even lethal disease when infected with very low viral titers. Our studies highlight the critical public health need to translate these findings and better understand vaccination in this increasing population.


Assuntos
Adjuvantes Imunológicos/administração & dosagem , Anticorpos Neutralizantes/sangue , Anticorpos Antivirais/sangue , Vacinas contra Influenza/administração & dosagem , Vacinas contra Influenza/imunologia , Influenza Humana/prevenção & controle , Obesidade/imunologia , Compostos de Alúmen/administração & dosagem , Animais , Modelos Animais de Doenças , Combinação de Medicamentos , Glicoproteínas de Hemaglutininação de Vírus da Influenza/imunologia , Humanos , Subtipo H7N9 do Vírus da Influenza A/isolamento & purificação , Camundongos Endogâmicos C57BL , Camundongos Obesos , Neuraminidase/imunologia , Polissorbatos/administração & dosagem , Sistema Respiratório/virologia , Esqualeno/administração & dosagem , Análise de Sobrevida , Resultado do Tratamento , Carga Viral , Proteínas Virais/imunologia , alfa-Tocoferol/administração & dosagem
9.
Science ; 349(6249): 739-42, 2015 Aug 14.
Artigo em Inglês | MEDLINE | ID: mdl-26249231

RESUMO

The latest Ebola virus (EBOV) epidemic spread rapidly through Guinea, Sierra Leone, and Liberia, creating a global public health crisis and accelerating the assessment of experimental therapeutics and vaccines in clinical trials. One of those vaccines is based on recombinant vesicular stomatitis virus expressing the EBOV glycoprotein (VSV-EBOV), a live-attenuated vector with marked preclinical efficacy. Here, we provide the preclinical proof that VSV-EBOV completely protects macaques against lethal challenge with the West African EBOV-Makona strain. Complete and partial protection was achieved with a single dose given as late as 7 and 3 days before challenge, respectively. This indicates that VSV-EBOV may protect humans against EBOV infections in West Africa with relatively short time to immunity, promoting its use for immediate public health responses.


Assuntos
Vacinas contra Ebola/administração & dosagem , Ebolavirus/imunologia , Glicoproteínas/imunologia , Doença pelo Vírus Ebola/prevenção & controle , Proteínas Virais/imunologia , Animais , Avaliação Pré-Clínica de Medicamentos , Vacinas contra Ebola/imunologia , Ebolavirus/genética , Vetores Genéticos , Glicoproteínas/genética , Macaca , Vesiculovirus , Proteínas Virais/genética
10.
BMC Bioinformatics ; 15 Suppl 4: I1, 2014.
Artigo em Inglês | MEDLINE | ID: mdl-25104130

RESUMO

Following last year's computational vaccinology workshop in Shanghai, China, the third ISV Pre-conference Computational Vaccinology Workshop (ICoVax 2013) was held in Barcelona, Spain. ICoVax 2013 provided an international platform for the attendees to showcase their research and discuss problems and solutions in the development and application of computational vaccinology and vaccine informatics tools. The first of the three full-length papers presented at ICoVax discussed the discovery of viral "camouflage" through cross-conservation of T-cell epitopes using a tool called JanusMatrix. This important paper reports that viruses may camouflage their presence in the human body by incorporating sequences in their proteins that are highly cross-conserved at the T-cell receptor surface with human genome proteins, a discovery that has wide ranging implications for the development of vaccines against viruses that use the camouflage method. The other papers described a database for storing experimentally verified data on DNA vaccines and compared therapeutic targets of western drugs to Chinese herbal medicines for cardiovascular diseases. The short poster presentations covered various uses of informatics tools for processing the DNA and microRNA of pathogens to improve vaccine coverage, efficacy and development. A live (on-line) demonstration of the vaccine design toolkit, iVax, presented by Frances Terry of EpiVax, illustrated how computational vaccinology could be used in the design of next generation vaccines.


Assuntos
Biologia Computacional/educação , Biologia Computacional/métodos , Vacinas Virais/imunologia , Doenças Cardiovasculares/tratamento farmacológico , China , Bases de Dados de Ácidos Nucleicos , Medicamentos de Ervas Chinesas/uso terapêutico , Epitopos de Linfócito T/imunologia , Genoma Humano , Humanos , Receptores de Antígenos de Linfócitos T/imunologia , Vacinas de DNA/imunologia , Proteínas Virais/imunologia , Vacinas Virais/química , Viroses/imunologia , Viroses/prevenção & controle , Vírus/imunologia
11.
PLoS One ; 9(7): e102235, 2014.
Artigo em Inglês | MEDLINE | ID: mdl-25014219

RESUMO

Hepatitis C virus (HCV) infection is still a serious global health burden. Despite improved therapeutic options, a preventative vaccine would be desirable especially in undeveloped countries. Traditionally, highly conserved epitopes are targets for antibody-based prophylactic vaccines. In HCV-infected patients, however, neutralizing antibodies are primarily directed against hypervariable region I (HVRI) in the envelope protein E2. HVRI is the most variable region of HCV, and this heterogeneity contributes to viral persistence and has thus far prevented the development of an effective HVRI-based vaccine. The primary goal of an antibody-based HCV vaccine should therefore be the induction of cross-reactive HVRI antibodies. In this study we approached this problem by presenting selected cross-reactive HVRI variants in a highly symmetric repeated array on capsid-like particles (CLPs). SplitCore CLPs, a novel particulate antigen presentation system derived from the HBV core protein, were used to deliberately manipulate the orientation of HVRI and therefore enable the presentation of conserved parts of HVRI. These HVRI-CLPs induced high titers of cross-reactive antibodies, including neutralizing antibodies. The combination of only four HVRI CLPs was sufficient to induce antibodies cross-reactive with 81 of 326 (24.8%) naturally occurring HVRI peptides. Most importantly, HVRI CLPs with AS03 as an adjuvant induced antibodies with a 10-fold increase in neutralizing capability. These antibodies were able to neutralize infectious HCVcc isolates and 4 of 19 (21%) patient-derived HCVpp isolates. Taken together, these results demonstrate that the induction of at least partially cross-neutralizing antibodies is possible. This approach might be useful for the development of a prophylactic HCV vaccine and should also be adaptable to other highly variable viruses.


Assuntos
Anticorpos Neutralizantes/biossíntese , Anticorpos Antivirais/biossíntese , Hepacivirus/imunologia , Vírus da Hepatite B/imunologia , Hepatite C/prevenção & controle , Vacinas contra Hepatite Viral/imunologia , Proteínas Virais/imunologia , Animais , Apresentação de Antígeno , Antígenos Virais/química , Antígenos Virais/genética , Antígenos Virais/imunologia , Proteínas do Capsídeo/química , Proteínas do Capsídeo/genética , Proteínas do Capsídeo/imunologia , Proteção Cruzada , Combinação de Medicamentos , Escherichia coli/genética , Escherichia coli/metabolismo , Feminino , Expressão Gênica , Vírus da Hepatite B/genética , Hepatite C/imunologia , Hepatite C/virologia , Humanos , Imunidade Humoral , Injeções Intramusculares , Injeções Subcutâneas , Camundongos , Camundongos Endogâmicos C57BL , Polissorbatos/administração & dosagem , Proteínas Recombinantes/química , Proteínas Recombinantes/genética , Proteínas Recombinantes/imunologia , Esqualeno/administração & dosagem , Vacinas contra Hepatite Viral/administração & dosagem , Vacinas contra Hepatite Viral/genética , Proteínas Virais/química , Proteínas Virais/genética , alfa-Tocoferol/administração & dosagem
12.
J Virol ; 88(14): 8180-8, 2014 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-24829334

RESUMO

The interferon antiviral system is a primary barrier to virus replication triggered upon recognition of nonself RNAs by the cytoplasmic sensors encoded by retinoic acid-inducible gene I (RIG-I), melanoma differentiation-associated gene 5 (MDA5), and laboratory of genetics and physiology gene 2 (LGP2). Paramyxovirus V proteins are interferon antagonists that can selectively interact with MDA5 and LGP2 through contact with a discrete helicase domain region. Interaction with MDA5, an activator of antiviral signaling, disrupts interferon gene expression and antiviral responses. LGP2 has more diverse reported roles as both a coactivator of MDA5 and a negative regulator of both RIG-I and MDA5. This functional dichotomy, along with the concurrent interference with both cellular targets, has made it difficult to assess the unique consequences of V protein interaction with LGP2. To directly evaluate the impact of LGP2 interference, MDA5 and LGP2 variants unable to be recognized by measles virus and parainfluenza virus 5 (PIV5) V proteins were tested in signaling assays. Results indicate that interaction with LGP2 specifically prevents coactivation of MDA5 signaling and that LGP2's negative regulatory capacity was not affected. V proteins only partially antagonize RIG-I at high concentrations, and their expression had no additive effects on LGP2-mediated negative regulation. However, conversion of RIG-I to a direct V protein target was accomplished by only two amino acid substitutions that allowed both V protein interaction and efficient interference. These results clarify the unique consequences of MDA5 and LGP2 interference by paramyxovirus V proteins and help resolve the distinct roles of LGP2 in both activation and inhibition of antiviral signal transduction. Importance: Paramyxovirus V proteins interact with two innate immune receptors, MDA5 and LGP2, but not RIG-I. V proteins prevent MDA5 from signaling to the beta interferon promoter, but the consequences of LGP2 targeting are poorly understood. As the V protein targets MDA5 and LGP2 simultaneously, and LGP2 is both a positive and negative regulator of both MDA5 and RIG-I, it has been difficult to evaluate the specific advantages conferred by LGP2 targeting. Experiments with V-insensitive proteins revealed that the primary outcome of LGP2 interference is suppression of its ability to synergize with MDA5. LGP2's negative regulation of MDA5 and RIG-I remains intact irrespective of V protein interaction. Complementary experiments demonstrate that RIG-I can be converted to V protein sensitivity by two amino acid substitutions. These findings clarify the functions of LGP2 as a positive regulator of MDA5 signaling, demonstrate the basis for V-mediated LGP2 targeting, and broaden our understanding of paramyxovirus-host interactions.


Assuntos
RNA Helicases DEAD-box/metabolismo , Interações Hospedeiro-Patógeno , Evasão da Resposta Imune , Vírus do Sarampo/imunologia , Vírus da Parainfluenza 5/imunologia , RNA Helicases/metabolismo , Transdução de Sinais , Proteínas Virais/imunologia , Linhagem Celular , Humanos , Helicase IFIH1 Induzida por Interferon , Ligação Proteica
13.
J Econ Entomol ; 107(1): 75-82, 2014 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-24665687

RESUMO

Thrips-transmitted Iris yellow spot virus (IYSV) is an important economic constraint to the production of bulb and seed onion crops in the United States and many other parts of the world. Because the virus is exclusively spread by thrips, the ability to rapidly detect the virus in thrips vectors would facilitate studies on the role of thrips in virus epidemiology, and thus formulation of better vector management strategies. Using a polyclonal antiserum produced against the recombinant, Escherichia coli-expressed nonstructural protein coded by the small (S) RNA of IYSV, an enzyme linked immunosorbent assay was developed for detecting IYSV in individual as well as groups of adult thrips. The approach enabled estimating the proportion of potential thrips transmitters in a large number of field-collected thrips collected from field-grown onion plants. Availability of a practical and inexpensive test to identify viruliferous thrips would be useful in epidemiological studies to better understand the role of thrips vectors in outbreaks of this economically important virus of onion.


Assuntos
Bunyaviridae/isolamento & purificação , Insetos Vetores/virologia , Tisanópteros/virologia , Animais , Bunyaviridae/imunologia , Ensaio de Imunoadsorção Enzimática , Cebolas/virologia , Proteínas Virais/imunologia
14.
Clin Exp Immunol ; 176(2): 199-206, 2014 May.
Artigo em Inglês | MEDLINE | ID: mdl-24387268

RESUMO

Autoimmune diabetes is characterized by autoantigen-specific T cell-mediated destruction of pancreatic islet beta cells, and CD8(+) T cells are key players during this process. We assessed whether the bitransgenic RIP-CD80 x RIP-LCMV-GP (RIP-CD80GP) mice may be a versatile antigen-specific model of inducible CD8(+) T cell-mediated autoimmune diabetes. Antigen-encoding DNA, peptide-loaded dendritic cells and antigen plus incomplete Freund's adjuvant were used for vaccination. Of 14 pancreatic proteins tested by DNA vaccination, murine pre-proinsulin 2 (100% of mice; median time after vaccination, 60 days) and islet-specific glucose-6-phosphatase catalytic subunit-related protein (IGRP) (77%, 58 days) could induce diabetes. Vaccination with DNA encoding for zinc transporter 8, Ia-2, Ia-2ß, glutamic acid decarboxylase 67 (Gad67), chromogranin A, insulinoma amyloid polypeptide and homeobox protein Nkx-2.2 induced diabetes development in 25-33% of mice. Vaccination with DNA encoding for Gad65, secretogranin 5, pancreas/duodenum homeobox protein 1 (Pdx1), carboxyl ester lipase, glucagon and control hepatitis B surface antigen (HBsAg) induced diabetes in <20% of mice. Diabetes induction efficiency could be increased by DNA vaccination with a vector encoding a ubiquitin-antigen fusion construct. Diabetic mice had florid T cell islet infiltration. CD8(+) T cell targets of IGRP were identified with a peptide library-based enzyme-linked immunospot assay, and diabetes could also be induced by vaccination with major histocompatibility complex (MHC) class I-restricted IGRP peptides loaded on mature dendritic cells. Vaccination with antigen plus incomplete Freund's adjuvant, which can prevent diabetes in other models, led to rapid diabetes development in the RIP-CD80GP mouse. We conclude that RIP-CD80GP mice are a versatile model of antigen specific autoimmune diabetes and may complement existing mouse models of autoimmune diabetes for evaluating CD8(+) T cell-targeted prevention strategies.


Assuntos
Linfócitos T CD8-Positivos/imunologia , Diabetes Mellitus Tipo 1/imunologia , Glucose-6-Fosfatase/imunologia , Insulina/imunologia , Precursores de Proteínas/imunologia , Vacinação/métodos , Animais , Antígeno B7-1/genética , Antígeno B7-1/imunologia , Linfócitos T CD8-Positivos/metabolismo , DNA/genética , DNA/imunologia , Diabetes Mellitus Tipo 1/etiologia , Diabetes Mellitus Tipo 1/genética , Adjuvante de Freund/imunologia , Glucose-6-Fosfatase/genética , Glicoproteínas/genética , Glicoproteínas/imunologia , Insulina/genética , Ilhotas Pancreáticas/imunologia , Estimativa de Kaplan-Meier , Lipídeos/imunologia , Vírus da Coriomeningite Linfocítica/genética , Vírus da Coriomeningite Linfocítica/imunologia , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Regiões Promotoras Genéticas/genética , Regiões Promotoras Genéticas/imunologia , Precursores de Proteínas/genética , Ratos , Fatores de Tempo , Vacinação/efeitos adversos , Vacinas de DNA/imunologia , Proteínas Virais/genética , Proteínas Virais/imunologia
16.
Mol Med ; 19: 276-85, 2013.
Artigo em Inglês | MEDLINE | ID: mdl-23979709

RESUMO

Chemokines facilitate the recruitment of inflammatory cells into tissues, contributing to target organ injury in a wide range of inflammatory and autoimmune diseases. Targeting either single chemokines or chemokine receptors alters the progression of disease in animal models of rheumatoid arthritis and lupus with varying degrees of efficacy but clinical trials in humans have been less successful. Given the redundancy of chemokine-chemokine receptor interactions, targeting of more than one chemokine may be required to inhibit active inflammatory disease. To test the effects of multiple-chemokine blockade in inflammation, we generated an adenovirus expressing bovine herpesvirus 1 glycoprotein G (BHV1gG), a viral chemokine antagonist that binds to a wide spectrum of murine and human chemokines, fused to the Fc portion of murine IgG2a. Administration of the adenovirus significantly inhibited thioglycollate-induced migration of polymorphonuclear leukocytes into the peritoneal cavity of BALB/c mice and reduced both clinical severity and articular damage in K/BxN serum transfer-induced arthritis. However, treatment with BHV1gG-Ig fusion protein did not prevent monocyte infiltration into the peritoneum in the thioglycollate model and did not prevent renal monocyte infiltration or nephritis in lupus-prone NZB/W mice. These observations suggest that the simultaneous inhibition of multiple chemokines by BHV1gG has the potential to interfere with acute inflammatory responses mediated by polymorphonuclear leukocytes, but is less effective in chronic inflammatory disease mediated by macrophages.


Assuntos
Movimento Celular/imunologia , Inflamação/imunologia , Monócitos/imunologia , Neutrófilos/imunologia , Proteínas Virais/imunologia , Animais , Artrite Experimental/imunologia , Artrite Experimental/prevenção & controle , Cálcio/imunologia , Cálcio/metabolismo , Bovinos , Movimento Celular/efeitos dos fármacos , Quimiocinas/metabolismo , Herpesvirus Bovino 1/genética , Soros Imunes/imunologia , Fragmentos Fc das Imunoglobulinas/genética , Fragmentos Fc das Imunoglobulinas/imunologia , Inflamação/metabolismo , Camundongos Endogâmicos BALB C , Camundongos Endogâmicos , Camundongos SCID , Monócitos/efeitos dos fármacos , Neutrófilos/efeitos dos fármacos , Ligação Proteica , Proteínas Recombinantes de Fusão/imunologia , Proteínas Recombinantes de Fusão/farmacologia , Tioglicolatos/imunologia , Tioglicolatos/farmacologia , Proteínas Virais/metabolismo , Proteínas Virais/farmacologia
17.
Fish Shellfish Immunol ; 30(1): 135-42, 2011 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-20887791

RESUMO

Killed viral vaccines and bacterial toxoids are weakly immunogenic. Numerous compounds are under evaluation as immunological adjuvants and peptide-carriers to improve the immune response. The hemocyanins, giant extracellular copper proteins in the blood of many mollusks, are widely used as immune stimulants. In the present study we investigated the adjuvant properties of hemocyanins isolated from marine gastropods Rapana thomasiana and Megathura crenulata. An immunization with Influenza vaccine or tetanus toxoid combined with Rapana thomasiana hemocyanin (RtH) and Keyhole limpet hemocyanin (KLH) in mice induced an anti-influenza cytotoxic response lasting at least 5 months and an antibody response to viral proteins. The IgG antibody response to the tetanus toxoid (TT) combined with RtH or KLH was comparable to the response of the toxoid in complete Freund's adjuvant. The results obtained demonstrate that the both hemocyanins are acceptable as potential bio-adjuvants for subunit vaccines.


Assuntos
Adjuvantes Imunológicos/farmacologia , Gastrópodes/metabolismo , Hemocianinas/análogos & derivados , Hemocianinas/farmacologia , Animais , Proteínas de Bactérias/imunologia , Linhagem Celular , Cães , Feminino , Hemocianinas/química , Imunoglobulina G/sangue , Imunoglobulina M/sangue , Vacinas contra Influenza/imunologia , Camundongos , Camundongos Endogâmicos BALB C , Orthomyxoviridae , Toxoide Tetânico/imunologia , Proteínas Virais/imunologia
18.
Immunol Lett ; 120(1-2): 108-16, 2008 Oct 30.
Artigo em Inglês | MEDLINE | ID: mdl-18706443

RESUMO

Transgenic (Tg) mice expressing HLA class I alleles and lacking murine MHC class I represent a useful model for the pre-clinical evaluation of human vaccines, which focus on induction of CD8(+) T-cell responses. We have developed a platform to be used in Tg mice for exploring the immunogenicity of T-cell targets, whose immunologic epitopes have yet to be defined. To test the attributes of the evaluation system in the context of an important human pathogen, we have explored multiple antigens from cytomegalovirus (CMV). A panel of recombinant modified vaccinia Ankara (MVA) vectors, expressing various CMV proteins (CMV-MVA) was used to immunize HLA-A*0201, B*0702 and A*1101 Tg mice. Immune splenocytes were in vitro stimulated (IVS) either using syngeneic lipo-polysaccharide activated lymphoblasts or Tg HLA-I matched human EBV-transformed B-lymphoblastoid cells (LCL), both loaded with peptide libraries, encompassing the CMV protein under investigation. IVS performed with peptide library loaded lymphoblasts failed to provide a reliable stimulation. In contrast, the usage of LCL as antigen presenting cells (APC) of CMV peptide libraries resulted in a consistent and specific amplification of the Tg T-cell response in animals immunized with CMV-MVAs. The LCL IVS method reliably allowed defining the immunogenicity and immunodominant CD8(+) T-cell regions of uncharacterized CMV antigens. The combination of CMV-MVA vectors, unbiased pools of CMV-specific peptide libraries presented by Tg HLA-I matched LCL constitutes a valid tool for the pre-clinical evaluation of model candidate vaccines. This convenient method could find application to investigate the immunogenicity profile of cancer antigens or proteins from infectious human pathogens.


Assuntos
Antígenos Virais/imunologia , Avaliação Pré-Clínica de Medicamentos/métodos , Antígenos HLA/genética , Antígenos HLA/imunologia , Animais , Células Apresentadoras de Antígenos/imunologia , Linfócitos T CD8-Positivos/imunologia , Citomegalovirus/imunologia , Epitopos de Linfócito T/imunologia , Humanos , Proteínas Imediatamente Precoces/imunologia , Camundongos , Camundongos Knockout , Fosfoproteínas/imunologia , Transativadores/imunologia , Proteínas Virais/imunologia , Vacinas Virais/imunologia
19.
Vet Immunol Immunopathol ; 121(1-2): 83-90, 2008 Jan 15.
Artigo em Inglês | MEDLINE | ID: mdl-18006078

RESUMO

The expression of recombinant antigens in transgenic plants is increasingly used as an alternative method of producing experimental immunogens. In this report, we describe the production of transgenic tomato plants that express the structural polyprotein, P1-2A, and protease, 3C, from foot-and-mouth disease (FMDV). P1-2A3C was inserted into the plant binary vector, pBin438, and transformed into tomato plants using Agrobacterium tumefaciens strain, GV3101. The presence of P1-2A3C was confirmed by PCR, transcription was verified by RT-PCR, and recombinant protein expression was confirmed by sandwich-ELISA and Western blot analyses. Guinea pigs immunized intramuscularly with foliar extracts from P1-2A3C-transgenic tomato plants were found to develop a virus-specific antibody response against FMDV. Vaccinated guinea pigs were fully protected against a challenge infection, while guinea pigs injected with untransformed plant extracts failed to elicit an antibody response and were not protected against challenge. These results demonstrate that transgenic tomato plants expressing the FMDV structural polyprotein, P1-2A, and the protease, 3C, can be used as a source of recombinant antigen for vaccine production.


Assuntos
Cisteína Endopeptidases/imunologia , Vírus da Febre Aftosa/imunologia , Febre Aftosa/imunologia , Imunização/métodos , Plantas Geneticamente Modificadas/química , Poliproteínas/imunologia , Proteínas Virais/imunologia , Proteases Virais 3C , Animais , Anticorpos Antivirais/sangue , Cisteína Endopeptidases/genética , DNA Viral/genética , Ensaio de Imunoadsorção Enzimática , Febre Aftosa/prevenção & controle , Vírus da Febre Aftosa/genética , Cobaias , Solanum lycopersicum/química , Solanum lycopersicum/genética , Extratos Vegetais/imunologia , Extratos Vegetais/farmacologia , Plantas Geneticamente Modificadas/genética , Plantas Geneticamente Modificadas/imunologia , Reação em Cadeia da Polimerase , Poliproteínas/genética , Distribuição Aleatória , Transcrição Gênica , Transformação Genética , Proteínas Virais/genética
20.
J Virol Methods ; 137(1): 1-6, 2006 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-16884787

RESUMO

Three single-chain variable fragment (scFv) antibodies recognizing the nuclear inclusion a (NIa) and capsid proteins of potato virus Y were obtained from two mouse derived hybridoma clones secreting, respectively, an anti-NIa (22-1) and an anti-coat protein (136-13) monoclonal antibodies. The first monoclonal antibody was able to inhibit in vitro the PVY polyprotein cleavage by blocking the NIa protease activity. The amplified scFv cDNAs were first inserted into the TOPO vector and then sequenced. Several recombinant E. coli clones carrying the accurate scFv sequences were selected and the corresponding cDNAs were subcloned in pHEN phagemid and transferred in E. coli strain. The expressed scFv fragments showed an antibody activity that recognized the viral target proteins in infected tissues. Their activity was comparable to the parental monoclonal antibodies.


Assuntos
Anticorpos Antivirais/genética , Anticorpos Antivirais/imunologia , Região Variável de Imunoglobulina/genética , Região Variável de Imunoglobulina/imunologia , Potyvirus/imunologia , Proteínas Virais/imunologia , Sequência de Aminoácidos , Anticorpos Monoclonais , Anticorpos Antivirais/biossíntese , Proteínas do Capsídeo/imunologia , Clonagem Molecular , DNA Complementar , Endopeptidases , Escherichia coli/genética , Técnica Indireta de Fluorescência para Anticorpo , Hibridomas , Região Variável de Imunoglobulina/biossíntese , Dados de Sequência Molecular , Proteínas Recombinantes/biossíntese , Proteínas Recombinantes/genética , Proteínas Recombinantes/imunologia , Análise de Sequência de DNA , Solanum tuberosum/virologia
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA