Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 54
Filtrar
1.
J Gen Virol ; 99(8): 1078-1085, 2018 08.
Artigo em Inglês | MEDLINE | ID: mdl-29923817

RESUMO

Dengue virus (DENV) is one the most important viral pathogens worldwide. Currently there is an imperative need for a reliable vaccine capable of inducing durable protection against all four serotypes. We have previously reported strongly neutralizing and highly specific antibody responses from all four serotypes to a DNA vaccine based on an engineered version of DENV E protein's domain III (DIII). Here, we show that monovalent and tetravalent immunizations with the DIII-based DNA vaccines are also capable of inducing highly stable antibody responses that remain strongly neutralizing over long periods of time. Our results demonstrate that DNA-vaccinated mice maintain a strong antibody response in terms of titre, avidity and virus-neutralizing capability 1 year after immunization.


Assuntos
Vacinas contra Dengue/imunologia , Vírus da Dengue/classificação , Dengue/prevenção & controle , Animais , Anticorpos Neutralizantes/sangue , Anticorpos Neutralizantes/imunologia , Anticorpos Antivirais/sangue , Anticorpos Antivirais/imunologia , Formação de Anticorpos , Dengue/virologia , Vírus da Dengue/imunologia , Feminino , Humanos , Camundongos , Camundongos Endogâmicos BALB C , Vacinas de DNA/imunologia , Proteínas do Envelope Viral
2.
Sci Rep ; 7(1): 966, 2017 04 19.
Artigo em Inglês | MEDLINE | ID: mdl-28424472

RESUMO

Dengue and Zika are two of the most important human viral pathogens worldwide. In both cases, the envelope glycoprotein E is the main target of the antibody response. Recently, new complex quaternary epitopes were identified which are the consequence of the arrangement of the antiparallel E dimers on the viral surface. Such epitopes can be exploited to develop more efficient cross-neutralizing vaccines. Here we describe a successful covalent stabilization of E dimers from Dengue and Zika viruses in mammalian cells. Folding and dimerization of secretory E was found to be strongly dependent on temperature but independent of PrM co-expression. In addition, we found that, due to the close relationship between flaviviruses, Dengue and Zika viruses E proteins can form heterodimers and assemble into mosaic viral particles. Finally, we present new virus-free analytical platforms to study and screen antibody responses against Dengue and Zika, which allow for differentiation of epitopes restricted to specific domains, dimers and higher order arrangements of E.


Assuntos
Vírus da Dengue/metabolismo , Dobramento de Proteína , Multimerização Proteica , Proteínas do Envelope Viral/metabolismo , Zika virus/metabolismo , Animais , Chlorocebus aethiops , Vírus da Dengue/fisiologia , Células HEK293 , Células HeLa , Humanos , Camundongos , Estabilidade Proteica , Temperatura , Células Vero , Proteínas do Envelope Viral/química , Zika virus/fisiologia
3.
Biochem Biophys Res Commun ; 492(4): 579-586, 2017 10 28.
Artigo em Inglês | MEDLINE | ID: mdl-28069378

RESUMO

Zika virus has rapidly spread reaching a global distribution pattern similar to that of dengue virus, and has been associated with serious neurological and developmental pathologies, like congenital malformation during pregnancy and Guillain-Barré syndrome. Sequence analysis of different clinical and laboratory isolates has shown the existence of mutants with loss of the conserved N-glycosylation motif on domain I of protein E that is common to all flaviviruses. We found that loss of E N-linked glycosylation leads to compromised expression and secretion of E ectodomain from mammalian cells. For both, wild type and glycosylation-negative mutant, secretion was independent of co-expression of the PrM viral protein, but highly dependent on temperature. Low temperature (28 °C) favoured secretion, although the glycosylation mutant E ectodomain showed impaired secretion and membrane display compared to the wild type. Production of pseudoviral particles with a West Nile virus replicon packaged with the Zika virus structural proteins C-PrM-E was significantly reduced with the non-glycosylated E. Similarly, glycosylation-negative pseudoviral particles showed impaired infectivity of Vero cells and reduced ability to infect K562 cells upon particles opsonisation with anti-E antibodies.


Assuntos
Proteínas do Envelope Viral/metabolismo , Ativação Viral/fisiologia , Montagem de Vírus/fisiologia , Infecção por Zika virus/virologia , Zika virus/fisiologia , Animais , Glicosilação , Humanos , Células K562 , Domínios Proteicos , Células Vero , Zika virus/patogenicidade
4.
Clin Exp Immunol ; 186(3): 356-363, 2016 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-27548641

RESUMO

New data suggest the involvement of rotavirus (RV) in triggering autoimmunity in coeliac disease (CD) by molecular mimicry between the human-transglutaminase protein and the dodecapeptide (260-271 aa) of the RV protein VP7 (pVP7). To assess the role of RV in the onset of CD, we measured anti-pVP7 antibodies in the sera of children with CD and of control groups. We analysed serum samples of 118 biopsy-proven CD patients and 46 patients with potential CD; 32 children with other gastrointestinal diseases; 107 no-CD children and 107 blood donors. Using enzyme-linked immunosorbent assay (ELISA) assay, we measured immunoglobulin (Ig)A-IgG antibodies against the synthetic peptides pVP7, the human transglutaminase-derived peptide (476-487 aa) which shows a homology with VP7 protein and a control peptide. The triple-layered RV particles (TLPs) containing the VP7 protein and the double-layered RV-particles (DLPs) lacking the VP7 protein were also used as antigens in ELISA assay. Antibody reactivity to the RV-TLPs was positive in 22 of 118 (18%) CD patients and in both paediatric (17 of 107, 16%) and adult (29 of 107, 27%) control groups, without showing a statistically significant difference among them (P = 0·6, P = 0·1). Biopsy-proven CD patients as well as the adult control group demonstrated a high positive antibody reactivity against both pVP7 (34 of 118, 29% CD patients; 66 of 107, 62% adult controls) and control synthetic peptides (35 of 118, 30% CD patients; 56 of 107, 52% adult controls), suggesting a non-specific response against RV pVP7. We show that children with CD do not have higher immune reactivity to RV, thus questioning the molecular mimicry mechanism as a triggering factor of CD.


Assuntos
Doença Celíaca/etiologia , Mimetismo Molecular , Infecções por Rotavirus/complicações , Infecções por Rotavirus/imunologia , Rotavirus/imunologia , Adolescente , Adulto , Anticorpos Antivirais/sangue , Anticorpos Antivirais/imunologia , Antígenos Virais/imunologia , Criança , Pré-Escolar , Feminino , Humanos , Imunoglobulina A/sangue , Imunoglobulina A/imunologia , Imunoglobulina G/sangue , Imunoglobulina G/imunologia , Lactente , Masculino , Infecções por Rotavirus/virologia , Adulto Jovem
5.
J Gen Virol ; 96(11): 3265-3279, 2015 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-26358704

RESUMO

Dengue virus (DENV) is currently among the most important human pathogens and affects millions of people throughout the tropical and subtropical regions of the world. Although it has been a World Health Organization priority for several years, there is still no efficient vaccine available to prevent infection. The envelope glycoprotein (E), exposed on the surface on infective viral particles, is the main target of neutralizing antibodies. For this reason it has been used as the antigen of choice for vaccine development efforts. Here we show a detailed analysis of factors involved in the expression, secretion and folding of E ectodomain from all four DENV serotypes in mammalian cells, and how this affects their ability to induce neutralizing antibody responses in DNA-vaccinated mice. Proper folding of E domain II (DII) is essential for efficient E ectodomain secretion, with DIII playing a significant role in stabilizing soluble dimers. We also show that the level of protein secreted from transfected cells determines the strength and efficiency of antibody responses in the context of DNA vaccination and should be considered a pivotal feature for the development of E-based DNA vaccines against DENV.


Assuntos
Vacinas contra Dengue/imunologia , Vírus da Dengue/metabolismo , Dengue/imunologia , Proteínas do Envelope Viral/química , Proteínas do Envelope Viral/metabolismo , Animais , Dengue/genética , Dengue/virologia , Vacinas contra Dengue/administração & dosagem , Vacinas contra Dengue/genética , Vírus da Dengue/química , Vírus da Dengue/classificação , Vírus da Dengue/imunologia , Feminino , Humanos , Camundongos , Camundongos Endogâmicos BALB C , Dobramento de Proteína , Estrutura Terciária de Proteína , Sistemas de Translocação de Proteínas/genética , Sistemas de Translocação de Proteínas/metabolismo , Transporte Proteico , Sorogrupo , Vacinas de DNA/administração & dosagem , Vacinas de DNA/genética , Vacinas de DNA/imunologia , Proteínas do Envelope Viral/genética , Proteínas do Envelope Viral/imunologia
6.
Gene Ther ; 22(12): 960-7, 2015 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-26181624

RESUMO

The ganglioside GM3(Neu5Gc) has gained increasing attention as therapeutic target because of its selective expression in various human tumours, such as melanoma, breast and lung cancer. 14F7 is a mouse IgG1 with specific reactivity to GM3(Neu5Gc)-positive tumours. The therapeutic activity of 14F7 has also been demonstrated in vivo, through its repetitive passive administration in tumour-bearing animals. In this work we used an alternative strategy to deliver recombinant 14F7 in vivo and analysed the therapeutic efficacy of this approach. We engineered a recombinant adeno-associated vector to direct the expression of secretable recombinant 14F7 in BALB/c animals. A single administration of the rAAV induced efficient production and secretion of the antibody in the bloodstream, with an expression level reaching plateau at ∼3 weeks after injection and persisting for almost a year. Strikingly, upon challenge with GM3(Neu5Gc)-positive X63-AG8.653 myeloma cells, tumour development was significantly delayed in animals treated with rAAV-14F7 with respect to animals treated with a control rAAV codifying for an irrelevant antibody. Finally, no significant differences in survival proportion were detected in animals injected with rAAV-14F7 or treated by standard administration of repetitive doses of purified monoclonal antibody 14F7.


Assuntos
Anticorpos Monoclonais/biossíntese , Anticorpos Monoclonais/imunologia , Gangliosídeo G(M3)/imunologia , Sequência de Aminoácidos , Animais , Dependovirus/genética , Dependovirus/metabolismo , Células HEK293 , Humanos , Imunoglobulina G/biossíntese , Imunoglobulina G/imunologia , Camundongos , Camundongos Endogâmicos BALB C , Dados de Sequência Molecular , Neoplasias Experimentais/imunologia , Neoplasias Experimentais/terapia , Proteínas Recombinantes/genética , Proteínas Recombinantes/metabolismo
7.
J Gen Virol ; 93(Pt 7): 1474-1482, 2012 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-22442113

RESUMO

Although inserting exogenous viral genome segments into rotavirus particles remains a hard challenge, this study describes the in vivo incorporation of a recombinant viral capsid protein (VP6) into newly assembled rotavirus particles. In vivo biotinylation technology was exploited to biotinylate a recombinant VP6 protein fused to a 15 aa biotin-acceptor peptide (BAP) by the bacterial biotin ligase BirA contextually co-expressed in mammalian cells. To avoid toxicity of VP6 overexpression, a stable HEK293 cell line was constructed with tetracycline-inducible expression of VP6-BAP and constitutive expression of BirA. Following tetracycline induction and rotavirus infection, VP6-BAP was biotinylated, recruited into viroplasms and incorporated into newly assembled virions. The biotin molecules in the capsid allowed the use of streptavidin-coated magnetic beads as a purification technique instead of CsCl gradient ultracentrifugation. Following transfection, double-layered particles attached to beads were able to induce viroplasm formation and to generate infective viral progeny.


Assuntos
Biotinilação/métodos , Rotavirus/crescimento & desenvolvimento , Coloração e Rotulagem/métodos , Virologia/métodos , Montagem de Vírus , Proteínas do Capsídeo/genética , Proteínas do Capsídeo/metabolismo , Linhagem Celular , Humanos , Proteínas Recombinantes/genética , Proteínas Recombinantes/metabolismo , Recombinação Genética , Rotavirus/genética , Rotavirus/fisiologia
8.
J Virol ; 85(6): 2781-92, 2011 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-21228236

RESUMO

The ubiquitin-proteasome system has been shown to play an important role in the replication cycle of different viruses. In this study, we describe a strong impairment of rotavirus replication upon inhibition of proteasomal activity. The effect was evidenced at the level of accumulation of viral proteins, viral RNA, and yield of infective particles. Kinetic studies revealed that the early steps of the replicative cycle following attachment, entry, and uncoating were clearly more sensitive to proteasome inhibition. We ruled out a direct inhibition of the viral polymerase activities and stability of viral proteins and found that the crucial step that was impaired by blocking proteasome activity was the assembly of new viroplasms. This was demonstrated by using chemical inhibitors of proteasome and by gene silencing using small interfering RNAs (siRNAs) specific for different proteasomal subunits and for the ubiquitin precursor RPS27A. In addition, we show that the effect of proteasome inhibition on virus infection is not due to increased levels of beta interferon (IFN-ß).


Assuntos
Complexo de Endopeptidases do Proteassoma/metabolismo , Rotavirus/fisiologia , Internalização do Vírus , Replicação Viral , Animais , Linhagem Celular , Inativação Gênica , Inibidores de Proteassoma , RNA Viral/metabolismo , Proteínas Virais/metabolismo
9.
J Gen Virol ; 91(Pt 7): 1782-93, 2010 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-20200190

RESUMO

Rotavirus genome replication and the first steps of virus morphogenesis take place in cytoplasmic viral factories, called viroplasms, containing four structural (VP1, VP2, VP3 and VP6) and two non-structural (NSP2 and NSP5) proteins. NSP2 and NSP5 have been shown to be essential for viroplasm formation and, when co-expressed in uninfected cells, to form viroplasm-like structures (VLS). In the present work, VLS formation was shown upon co-expression of NSP5 with the core protein VP2 despite the absence of NSP2, indicating a central role for NSP5 in VLS assembly. Since VP2 and NSP2 also induce NSP5 hyperphosphorylation, the possible correlation between VLS formation and the NSP5 phosphorylation status was investigated without evidence of a direct link. In VLS induced by NSP2, the polymerase VP1 was recruited, while the middle layer protein VP6 was not, forming instead tubular structures. On the other hand, VLS induced by VP2 were able to recruit both VP1 and VP6. More importantly, in VLS formed when NSP5 was expressed with both inducers, all viroplasmic proteins were found co-localized, resembling their distribution in viroplasms. Our results suggest a key role for NSP5 in architectural assembly of viroplasms and in recruitment of viroplasmic proteins. A new role for VP2 as an inducer of viroplasms and of NSP5 hyperphosphorylation is also described. These data may contribute to the understanding of rotavirus morphogenesis.


Assuntos
Rotavirus/fisiologia , Proteínas não Estruturais Virais/metabolismo , Montagem de Vírus/fisiologia , Animais , Proteínas do Capsídeo/genética , Proteínas do Capsídeo/metabolismo , Linhagem Celular , Regulação Viral da Expressão Gênica/fisiologia , Rim/citologia , Proteínas não Estruturais Virais/genética
10.
Scand J Immunol ; 68(1): 58-66, 2008 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-18482206

RESUMO

Thanks to the safety of administration, efficiency of in vivo transduction and persistence of transgene expression, vectors based on the adeno-associated virus (AAV) are extensively utilized in both preclinical and clinical experimentation. Here we thoroughly explore the potential of AAV-mediated antigen delivery for tumour vaccination. A recombinant AAV vector (rAAV) encoding a lymphoma idiotype (Id) in a single-chain variable fragment format was found to induce an efficient anti-Id immune response upon injection in immunocompetent animals. The intensity of the immune response and the protective effect of rAAV administration in vivo were systematically compared with those elicited by simple injection of naked DNA or biolistic immunization. The results indicate that Id delivery via rAAV enhances the intensity of immune response compared with injection of naked DNA, while anti-idiotypic antibodies titres are not considerably increased compared with biolistic vaccination. On the contrary, a prime-boost vaccination strategy combining biolistic and AAV DNA delivery results in a major increase in anti-Id antibody response compared with the repetitive biolistic immunization. This increased anti-Id humoral response strictly correlated with a significant improvement on tumour protection in vivo.


Assuntos
Anticorpos Anti-Idiotípicos/imunologia , Biolística/métodos , Vacinas Anticâncer/imunologia , Dependovirus/genética , Idiótipos de Imunoglobulinas/genética , Linfoma de Células B/terapia , Animais , Antígenos de Neoplasias/imunologia , Vetores Genéticos , Humanos , Idiótipos de Imunoglobulinas/administração & dosagem , Idiótipos de Imunoglobulinas/imunologia , Injeções Intramusculares , Injeções Intraperitoneais , Injeções Subcutâneas , Linfoma de Células B/imunologia , Camundongos , Transdução Genética , Vacinas de DNA
11.
J Virol ; 81(5): 2128-37, 2007 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-17182692

RESUMO

Rotavirus morphogenesis starts in intracellular inclusion bodies called viroplasms. RNA replication and packaging are mediated by several viral proteins, of which VP1, the RNA-dependent RNA polymerase, and VP2, the core scaffolding protein, were shown to be sufficient to provide replicase activity in vitro. In vivo, however, viral replication complexes also contain the nonstructural proteins NSP2 and NSP5, which were shown to be essential for replication, to interact with each other, and to form viroplasm-like structures (VLS) when coexpressed in uninfected cells. In order to gain a better understanding of the intermediates formed during viral replication, this work focused on the interactions of NSP5 with VP1, VP2, and NSP2. We demonstrated a strong interaction of VP1 with NSP5 but only a weak one with NSP2 in cotransfected cells in the absence of other viral proteins or viral RNA. By contrast, we failed to coimmunoprecipitate VP2 with anti-NSP5 antibodies or NSP5 with anti-VP2 antibodies. We constructed a tagged form of VP1, which was found to colocalize in viroplasms and in VLS formed by NSP5 and NSP2. The tagged VP1 was able to replace VP1 structurally by being incorporated into progeny viral particles. When applying anti-tag-VP1 or anti-NSP5 antibodies, coimmunoprecipitation of tagged VP1 with NSP5 was found. Using deletion mutants of NSP5 or different fragments of NSP5 fused to enhanced green fluorescent protein, we identified the 48 C-terminal amino acids as the region essential for interaction with VP1.


Assuntos
RNA Polimerases Dirigidas por DNA/fisiologia , Proteínas de Ligação a RNA/fisiologia , Rotavirus/fisiologia , Proteínas do Core Viral/fisiologia , Proteínas não Estruturais Virais/fisiologia , Animais , Sequência de Bases , Linhagem Celular , DNA Viral/genética , RNA Polimerases Dirigidas por DNA/genética , Microscopia de Fluorescência , Ligação Proteica , Proteínas de Ligação a RNA/genética , Proteínas Recombinantes/genética , Proteínas Recombinantes/metabolismo , Rotavirus/genética , Deleção de Sequência , Transfecção , Proteínas do Core Viral/genética , Proteínas não Estruturais Virais/química , Proteínas não Estruturais Virais/genética
12.
Curr Gene Ther ; 2(2): 235-42, 2002 May.
Artigo em Inglês | MEDLINE | ID: mdl-12109219

RESUMO

Vaccination protocols based on targeting of the idiotype expressed on malignant B cells have so far provided encouraging results in clinical trials. The essential requirement to induce an immune response is the inclusion of carriers to overcome T-cell tolerance. Chemical cross-linking of idiotypic protein is so far the method of choice to induce protective responses in human studies. Meanwhile, a flurry of alternative strategies to simplify vaccine production is being tested in murine model. Thanks to the advance in antibody engineering the two relevant antigenic domains of the lymphoma immunoglobulin can be assembled into an appropriate format, genetically linked to molecules that act as immunological adjuvants and directly delivered as plasmid DNA. Upon immunization, rejection of tumor cells may depend on cellular or humoral mechanisms, whose relative importance has not been entirely estimated. We have recently analyzed the specificity of anti-idiotypic antibodies induced by DNA vaccination and characterised the elements contributing to optimal anti-idiotypic responses.


Assuntos
Vacinas Anticâncer/farmacologia , Neoplasias Hematológicas/terapia , Imunoterapia/métodos , Animais , Membrana Celular/metabolismo , Ensaios Clínicos como Assunto , DNA/metabolismo , Humanos , Camundongos , Camundongos Endogâmicos BALB C , Modelos Biológicos , Plasmídeos/metabolismo
13.
Gene Ther ; 8(20): 1555-61, 2001 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-11704816

RESUMO

DNA vaccines encoding the idiotype of immunoglobulins of tumour B cells were shown to induce protection in several mouse lymphoma models. The mechanism of rejection of tumour cells has not been fully understood, but there is strong evidence suggesting that engagement of the idiotype by anti-idiotypic antibodies may directly result in inhibition of tumour growth. In this study, we have investigated the structural basis of the idiotypic/anti-idiotypic interaction following immunisation with DNA vaccines. scFvs containing only one of the two tumour-derived V regions recombined to an irrelevant V region partner were generated. These constructs encoding a secretory form of the scFv were used as immunogens to induce anti-Id antibodies. The same scFvs were expressed as membrane-bound molecules on the surface of mammalian cells. Analysis of immune sera on the membrane-displayed idiotypes revealed that DNA immunisation induced a polyclonal antibody response restricted to conformational combined epitopes formed by the parental V(L)/V(H) association. Immune sera raised by scFv DNA vaccination did not show any detectable reactivity towards chimeric scFvs containing only one of the two immunising V regions, indicating that the response against combined V(L)/V(H)determinants is highly dominant. Remarkably, the same immunogen, delivered as scFv protein, induced antibodies also directed against chain-specific determinants. These findings indicate that presentation of properly folded idiotypes results in a highly specific antibody response directed exclusively to private idiotypic determinants of the V(L)/V(H) combination of the immunogen.


Assuntos
Anticorpos Anti-Idiotípicos/análise , Idiótipos de Imunoglobulinas/genética , Região Variável de Imunoglobulina/imunologia , Imunoterapia Ativa/métodos , Linfoma/terapia , Vacinas de DNA/administração & dosagem , Animais , Linfócitos B/imunologia , Sequência de Bases , Western Blotting/métodos , Clonagem Molecular , Ensaio de Imunoadsorção Enzimática/métodos , Citometria de Fluxo/métodos , Engenharia Genética , Linfoma/imunologia , Camundongos , Camundongos Endogâmicos BALB C , Modelos Animais , Dados de Sequência Molecular , Transfecção , Células Tumorais Cultivadas
14.
Biochemistry ; 40(35): 10686-92, 2001 Sep 04.
Artigo em Inglês | MEDLINE | ID: mdl-11524014

RESUMO

Membrane-bound immunoglobulins have, in addition to the transmembrane and cytoplasmic portions, an extracellular membrane-proximal domain (EMPD), absent in the secretory forms. EMPDs of immunoglobulin isotypes alpha, gamma, and epsilon contain cysteines whose role has so far not been elucidated. Using a genetic strategy, we investigated the ability of these cysteines to form disulfide bridges. Shortened versions of human membrane immunoglobulins, depleted of cysteines known to form intermolecular disulfide bonds, were constructed and expressed on the surface of a B-cell line. The resulting membrane proteins contain a single chain fragment of variable regions (scFv) linked to the dimerizing domain from the immunoglobulin heavy chains (CH3 for alpha and gamma or CH4 for epsilon isotypes), followed by the corresponding EMPD and the transmembrane and cytoplasmic domains. The two functional membrane versions of the epsilon chain, containing the short and long EMPD, were analyzed. Our results show that the single cysteine within alpha1L and gamma1 EMPD and the short version of epsilon EMPD form an interchain disulfide bond. Conversely, the cysteine resident in the epsilon transmembrane domain remains unreacted. epsilon-long EMPD contains four cysteines; two are involved in interchain bonds while the remaining two are likely forming an intrachain bridge. Expression of a full-length membrane epsilon heavy chain mutant, in which Cys(121) and Cys(209) within domain CH2 (involved in interchain bridges) were mutated to alanines, confirmed that, within the complete IgE, EMPD cysteines form interchain disulfide bonds. In conclusion, we unveil evidence for additional covalent stabilization of membrane-bound immunoglobulins.


Assuntos
Dissulfetos/química , Receptores de Antígenos de Linfócitos B/química , Sequência de Aminoácidos , Biotina/metabolismo , Cisteína/química , Humanos , Dados de Sequência Molecular , Mutação , Receptores de Antígenos de Linfócitos B/genética , Alinhamento de Sequência
15.
Gene Ther ; 7(7): 605-11, 2000 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-10819576

RESUMO

Vaccination with immunogenic formulations of lymphoma-derived immunoglobulin can elicit strong anti-idiotypic immune responses which have proved effective in murine B cell tumor challenge experiments and suggested possible benefits in recent human clinical trials. Naked plasmid DNA vaccines encoding the Id determinants as scFv fragments provide the most promising alternative to protein immunization. With this approach the addition of an immunogenic domain linked to the scFv has proved essential for the induction of a protective immune response. In this study we have produced a scFv gene construct linked to the CH3 exon of the human IgG1 constant region and tested its efficacy in inducing protective immunity against the mouse BCL1 lymphoma. We have also generated a second construct in which the BCL1 VL gene was deleted to investigate whether the VH region domain contains sufficient antigenic determinants for a protective immune response. Both constructs induced anti-idiotypic antibodies that specifically reacted with the BCL1 IgM protein in ELISA and with BCL1 tumor cells in flow cytometry assays. Protection against tumor challenge was fully achieved with the complete scFv construct whereas immunization with the construct lacking the VL gene resulted in only a slight prolongation of the survival. We therefore conclude that a plasmid DNA vaccine containing the VH and VL genes of the lymphoma Ig linked to the human IgG1 CH3 exon is highly effective in inducing a protective immune response in the BCL1 model. We also demonstrated that VH gene immunization can induce strong anti-idiotypic antibody responses.


Assuntos
Anticorpos Anti-Idiotípicos/genética , Região Variável de Imunoglobulina/genética , Linfoma/terapia , Transfecção/métodos , Vacinas de DNA/administração & dosagem , Animais , Linhagem Celular , Impressões Digitais de DNA/métodos , Ensaio de Imunoadsorção Enzimática/métodos , Engenharia Genética/métodos , Linfoma/imunologia , Camundongos , Camundongos Endogâmicos BALB C
16.
Immunotechnology ; 4(3-4): 267-72, 1999 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-10231095

RESUMO

BACKGROUND: Unlike other immunoglobulin isotypes, the human C epsilon gene generates by alternative splicing two types of secretory and two types of membrane epsilon chains. The two secreted epsilon heavy chains, epsilon(S1) and epsilon(S2), differ only in the sequence of the last eight C-terminal amino acids, being epsilon(S2) six amino acids longer. The two types of membrane isoforms differ in the extracellular membrane proximal domain, with the longer variant, epsilon(mL), containing 52 extra amino acids which are absent in the shorter epsilon(mS) isoform. OBJECTIVES: We wished to produce quality antibody reagents that specifically detect epitopes that are epsilon isoform-specific. STUDY DESIGN: Short sequences of seven or ten amino acids were chosen as target epitopes and expressed as part of the highly immunogenic loops of deletion variants of engineered Flock House Virus capsid protein RNA2. Chimeric proteins were expressed in E. coli, and used to immunize rabbits. Antisera were screened by immunoblotting of purified IgE isoforms expressed by murine transfectomas. RESULTS: Chimeric proteins expressing epsilon isoform-specific epitopes proved to be strong immunogens in vivo and induced highly specific rabbit antisera. Two antisera so obtained recognize specifically the IgE-S2 isoform. A third one recognizes the long membrane variant m(L)IgE and a fourth one detects an epitope specific to m(S)IgE. CONCLUSION: Here we describe a simplified and efficient protocol of immunization which does not require peptide synthesis and conjugation to carrier protein. Our results show that short peptides of unknown immunogenicity, when genetically introduced into the modified Flock House Virus epitope display system, successfully induced IgE isoform-specific polyclonal antisera in rabbits. These are valuable tools to specifically identify secretory and membrane isoforms of human IgE, and the method is potentially applicable to other variant isoforms or mutants of a given protein.


Assuntos
Anticorpos Anti-Idiotípicos/imunologia , Epitopos/imunologia , Imunoglobulina E/imunologia , Cadeias épsilon de Imunoglobulina/imunologia , Sequência de Aminoácidos , Animais , Anticorpos Anti-Idiotípicos/sangue , Western Blotting , Capsídeo/genética , Capsídeo/imunologia , Citometria de Fluxo , Genes de Imunoglobulinas , Humanos , Imunização , Imunoglobulina E/química , Imunoglobulina E/genética , Cadeias épsilon de Imunoglobulina/química , Cadeias épsilon de Imunoglobulina/genética , Vírus de Insetos/genética , Dados de Sequência Molecular , Coelhos , Proteínas Recombinantes de Fusão/genética , Proteínas Recombinantes de Fusão/imunologia
17.
Eur J Immunol ; 29(3): 936-45, 1999 03.
Artigo em Inglês | MEDLINE | ID: mdl-10092098

RESUMO

We have recently reported that besides the most abundant form epsilonS1, there exists another human secretory epsilon H chain isoform, epsilonS2, resulting from alternative splicing in the epsilonCH4 exon. Using a specific antibody targeted to the epsilonS2-specific C-terminal tailpiece, we now show that this second secretory IgE isoform (IgE-S2) is constitutively co-expressed with the classical secretory IgE-S1 by human myeloma cells. The epsilonS2 variant was also detected in tonsils and in the serum of three non-atopic donors, but was absent in the vast majority of sera of both atopic and non-atopic individuals tested, indicating rare serum expression. IgE-S2 is capable of binding to cells expressing Fc epsilonRI, the high-affinity receptor for IgE. Analysis of intracellular tyrosine phosphorylation signal, degranulation, and rate of receptor internalization suggest a quantitatively lower response by IgE-S2 compared to IgE-S1. The modest differences observed do not appear to overall affect the degranulation competency of IgE-S2, but suggest that the unique structure of the epsilonS2 tailpiece can exert an effect on the interaction with the alpha chain of Fc epsilonRI.


Assuntos
Imunoglobulina E/imunologia , Receptores de IgE/imunologia , Humanos , Immunoblotting , Imunoglobulina E/genética , Leucócitos Mononucleares/imunologia , Mastócitos/imunologia , Tonsila Palatina/imunologia , Tonsila Palatina/patologia , Isoformas de Proteínas/genética , Isoformas de Proteínas/imunologia , RNA Mensageiro , Células Tumorais Cultivadas
18.
J Gen Virol ; 80 ( Pt 2): 333-339, 1999 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-10073692

RESUMO

In rotavirus-infected cells, the non-structural proteins NSP5 and NSP2 localize in complexes called viroplasms, where replication and assembly occur. Recently, we have demonstrated direct interaction of NSP5 with NSP2, and as a consequence of that, up-regulation of NSP5 hyperphosphorylation. To investigate a possible structural role for the NSP2-NSP5 interaction, we analysed the cytoplasmic distribution of the two proteins in transfected cells by immunofluorescence using specific antibodies. Here we report that NSP2 and NSP5 can drive the formation of viroplasm-like structures (VLS) in the absence of other rotaviral proteins and rotavirus replication. Several NSP5 deletion mutants were constructed and expressed in combination with NSP2. Both the N- and C-terminal domains of NSP5 were found to be essential for VLS formation. Only one mutant, with an internal deletion of residues 81-130, was able to interact with NSP2 to form VLS. Analysis of the phosphorylation capacity of the different mutants in vivo indicated that hyperphosphorylation of NSP5 is necessary, but not sufficient, for VLS formation. Our results suggest a role for the non-structural protein NSP5 in the structure of viroplasms mediated by its interaction with NSP2.


Assuntos
Proteínas de Ligação a RNA/química , Proteínas de Ligação a RNA/metabolismo , Rotavirus/metabolismo , Proteínas não Estruturais Virais/química , Proteínas não Estruturais Virais/metabolismo , Proteínas Virais/química , Proteínas Virais/metabolismo , Animais , Sequência de Bases , Linhagem Celular , Primers do DNA/genética , Substâncias Macromoleculares , Microscopia de Fluorescência , Fosforilação , Proteínas de Ligação a RNA/genética , Rotavirus/genética , Deleção de Sequência , Proteínas não Estruturais Virais/genética , Proteínas Virais/genética , Replicação Viral
19.
Exp Cell Res ; 246(2): 368-75, 1999 Feb 01.
Artigo em Inglês | MEDLINE | ID: mdl-9925752

RESUMO

Endothelial cells form the inner lining of blood and lymphatic vessels. In mice, only tumors of the blood vessel endothelium (haemangiomas) have been thus far reported. Here we describe a highly reproducible method for the induction of benign tumors of the lymphatic endothelial cells (lymphangiomas) in mice by intraperitoneal injection of incomplete Freund's adjuvant. Morphological and histopathological studies of the lesions revealed the presence of cells at various levels of vascular development. The lymphangiomas developed in the peritoneal cavity and expressed the endothelial markers CD31/PECAM (platelet endothelial cell adhesion molecule), CD54/ICAM-1 (InterCellular Adhesion Molecule-1), and CD102/ICAM-2, as well as the vascular endothelial growth factor (VEGF) receptor Flk-1, the endothelial cell specific receptors Tie-1 and Tie-2 and the lymphatic endothelial cell specific Flt4 receptor as shown by in situ hybridization. The Flk-1 and Flt4 receptors were also identified in immunoblots of the tumors and in cells cultured from them. When induced in beta-galactosidase knock-in Flt4(+/-) mice, the tumor endothelia could be stained blue in a number of tumor cells although the staining was of lower intensity than in normal lymphatic vessels. The tumor-derived cells could be propagated in vitro and they spontaneously differentiated, forming vessel-like structures. Murine lymphangiomas thus represent a highly reproducible and convenient source of lymphatic endothelial cells.


Assuntos
Carcinógenos/toxicidade , Adjuvante de Freund/toxicidade , Linfangioma/induzido quimicamente , Neoplasias Peritoneais/induzido quimicamente , Animais , Biomarcadores Tumorais/biossíntese , Divisão Celular , Endotélio Linfático , Expressão Gênica , Injeções Intraperitoneais , Linfangioma/metabolismo , Linfangioma/patologia , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Endogâmicos CBA , Neoplasias Peritoneais/metabolismo , Neoplasias Peritoneais/patologia , Proteínas Proto-Oncogênicas/genética , Coelhos , Receptores Proteína Tirosina Quinases/genética , Receptor 1 de Fatores de Crescimento do Endotélio Vascular
20.
J Gen Virol ; 79 ( Pt 11): 2679-86, 1998 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-9820143

RESUMO

We have previously shown that a number of isoforms of the non-structural rotavirus protein NSP5 are found in virus-infected cells. These isoforms differ in their level of phosphorylation which, at least in part, appears to occur through autophosphorylation. NSP5 co-localizes with another non-structural protein, NSP2, in the viroplasms of infected cells where virus replication takes place. We now show that NSP5 can be chemically cross-linked in living cells with the viral polymerase VP1 and NSP2. Interaction of NSP5 with NSP2 was also demonstrated by co-immunoprecipitation of NSP2 and NSP5 from extracts of UV-treated rotavirus-infected cells. In addition, in transient transfection assays, NSP5 phosphorylation in vivo was enhanced by co-expression of NSP2. An NSP5 C-terminal domain deletion mutant, was completely unable to be phosphorylated either in the presence or absence of NSP2. However, a 33 aa N-terminal deletion mutant of NSP5 was shown to become hyperphosphorylated in vivo and to be insensitive to NSP2 activation, suggesting a regulatory role for this domain in NSP5 phosphorylation and making it a candidate for the interaction with NSP2. These mutants also allow a preliminary mapping of NSP5 autophosphorylation activity.


Assuntos
Proteínas de Ligação a RNA/metabolismo , Rotavirus/metabolismo , Proteínas não Estruturais Virais/metabolismo , Proteínas Virais/metabolismo , Animais , Regulação Viral da Expressão Gênica , Mutação , Fosforilação , Proteínas de Ligação a RNA/genética , Regulação para Cima , Proteínas não Estruturais Virais/genética , Proteínas Virais/genética
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...