Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 17 de 17
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
2.
J Transl Med ; 21(1): 495, 2023 07 23.
Artigo em Inglês | MEDLINE | ID: mdl-37482614

RESUMO

BACKGROUND: Recombinant MVAs (rMVAs) are widely used both in basic and clinical research. Our previously developed Red-to-Green Gene Swapping Method (RGGSM), a cytometry-based Cell-Sorting protocol, revolves around the transient expression of a green fluorescent cytoplasmic marker, to subsequently obtain purified untagged rMVA upon loss of that marker by site-specific recombination. The standard RGSSM is quite costly in terms of bench work, reagents, and Sorting Facility fees. Although faster than other methods to obtain recombinant MVAs, the standard RGSSM still is time-consuming, taking at least 25 days to yield the final product. METHODS: The direct sorting of fluorescent virions is made amenable by the marker HAG, a flu hemagglutinin/EGFP fusion protein, integrated into the external envelope of extracellular enveloped virions (EEVs). Fluorescent EEVs-containing supernatants of infected cultures are used instead of purified virus. Direct Virus-Sorting was performed on BD FACSAria Fusion cell sorter equipped with 4 lasers and a 100-mm nozzle, with 20 psi pressure and a minimal flow rate, validated using Megamix beads. RESULTS: Upon infection of cells with recombinant EEVs, at the first sorting step virions that contain HAG are harvested and cloned, while the second sorting step yields EEVs that have lost HAG, allowing to clone untagged rMVA. Because only virion-containing supernatants are used, no virus purification steps and fewer sortings are necessary. Therefore, the final untagged rMVA product can be obtained in a mere 8 days. CONCLUSIONS: Altogether, we report that the original RGSSM has been markedly improved in terms of time- and cost efficiency by substituting Cell-Sorting with direct Virus-Sorting from the supernatants of infected cells. The improved virometry-based RGGSM may find wide applicability, considering that rMVAs hold great promise to serve as personalized vaccines for therapeutic intervention against cancer and various types of infectious diseases.


Assuntos
Vaccinia virus , Vírion , Análise Custo-Benefício , Vírion/metabolismo
3.
Environ Microbiol ; 22(6): 1997-2000, 2020 06.
Artigo em Inglês | MEDLINE | ID: mdl-32342578

RESUMO

The current SARS-CoV-2 pandemic is wreaking havoc throughout the world and has rapidly become a global health emergency. A central question concerning COVID-19 is why some individuals become sick and others not. Many have pointed already at variation in risk factors between individuals. However, the variable outcome of SARS-CoV-2 infections may, at least in part, be due also to differences between the viral subspecies with which individuals are infected. A more pertinent question is how we are to overcome the current pandemic. A vaccine against SARS-CoV-2 would offer significant relief, although vaccine developers have warned that design, testing and production of vaccines may take a year if not longer. Vaccines are based on a handful of different designs (i), but the earliest vaccines were based on the live, attenuated virus. As has been the case for other viruses during earlier pandemics, SARS-CoV-2 will mutate and may naturally attenuate over time (ii). What makes the current pandemic unique is that, thanks to state-of-the-art nucleic acid sequencing technologies, we can follow in detail how SARS-CoV-2 evolves while it spreads. We argue that knowledge of naturally emerging attenuated SARS-CoV-2 variants across the globe should be of key interest in our fight against the pandemic.


Assuntos
Betacoronavirus , Coronavírus Relacionado à Síndrome Respiratória Aguda Grave , COVID-19 , Infecções por Coronavirus , Surtos de Doenças , Humanos , Pandemias , Pneumonia Viral , SARS-CoV-2
4.
J Virol Methods ; 251: 7-14, 2018 01.
Artigo em Inglês | MEDLINE | ID: mdl-28987424

RESUMO

As a vaccination vector, MVA has been widely investigated both in animal models and humans. The construction of recombinant MVA (rMVA) relies on homologous recombination between an acceptor virus and a donor plasmid in infected/transfected permissive cells. Our construction strategy "Red-to-Green gene swapping" - based on the exchange of two fluorescent markers within the flanking regions of MVA deletion ΔIII, coupled to fluorescence activated cell sorting - is here extended to a second insertion site, within the flanking regions of MVA deletion ΔVI. Exploiting this strategy, both double and triple rMVA were constructed, expressing as transgenes the influenza A proteins HA, NP, M1, and PB1. Upon validation of the harbored transgenes co-expression, double and triple recombinants rMVA(ΔIII)-NP-P2A-M1 and rMVA(ΔIII)-NP-P2A-M1-(ΔVI)-PB1 were assayed for in vivo immunogenicity and protection against lethal challenge. In vivo responses were identical to those obtained with the reported combinations of single recombinants, supporting the feasibility and reliability of the present improvement and the extension of Red-to-Green gene swapping to insertion sites other than ΔIII.


Assuntos
Portadores de Fármacos , Vetores Genéticos , Vacinas contra Influenza/imunologia , Vaccinia virus/genética , Animais , Anticorpos Antivirais/sangue , Antígenos Virais/genética , Antígenos Virais/imunologia , Linfócitos T CD8-Positivos/imunologia , Expressão Gênica , Vacinas contra Influenza/administração & dosagem , Vacinas contra Influenza/genética , Camundongos Endogâmicos C57BL , Proteínas Recombinantes/genética , Proteínas Recombinantes/imunologia , Análise de Sobrevida , Vacinas Sintéticas/administração & dosagem , Vacinas Sintéticas/genética , Vacinas Sintéticas/imunologia , Proteínas Virais/genética , Proteínas Virais/imunologia
5.
J Immunol ; 197(7): 2583-8, 2016 10 01.
Artigo em Inglês | MEDLINE | ID: mdl-27566822

RESUMO

Exogenous IgE acts as an adjuvant in tumor vaccination in mice, and therefore a direct role of endogenous IgE in tumor immunosurveillance was investigated. By using genetically engineered mice, we found that IgE ablation rendered mice more susceptible to the growth of transplantable tumors. Conversely, a strengthened IgE response provided mice with partial or complete resistance to tumor growth, depending on the tumor type. By genetic crosses, we showed that IgE-mediated tumor protection was mostly lost in mice lacking FcεRI. Tumor protection was also lost after depletion of CD8(+) T cells, highlighting a cross-talk between IgE and T cell-mediated tumor immunosurveillance. Our findings provide the rationale for clinical observations that relate atopy with a lower risk for developing cancer and open new avenues for the design of immunotherapeutics relevant for clinical oncology.


Assuntos
Imunoglobulina E/imunologia , Vigilância Imunológica/imunologia , Neoplasias/imunologia , Receptores de IgE/imunologia , Adjuvantes Imunológicos , Animais , Engenharia Genética , Imunoterapia , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Knockout , Neoplasias/genética , Neoplasias/patologia , Neoplasias/terapia , Receptores de IgE/deficiência
6.
J Immunol ; 188(1): 103-10, 2012 Jan 01.
Artigo em Inglês | MEDLINE | ID: mdl-22124126

RESUMO

The IgE-mediated immune system activation can be redirected to combat tumors. Mouse and human IgE have been shown to provide a potent adjuvant effect in antitumor vaccination, with a crucial role played by FcεRI. This effect results from T cell-mediated adaptive immune response. Modified vaccinia virus Ankara (MVA) has been used to infect IgE-loaded tumor cells. These results led to a shift toward a highly safe protocol employing membrane IgE (mIgE), thus eliminating any possible anaphylactogenicity caused by circulating IgE. Evidence that human mIgE and a truncated version lacking IgE Fabs (tmIgE) bind and activate FcεRI has been fundamental and forms the core of this report. Human tmIgE has been engineered into a recombinant MVA (rMVA-tmIgE), and the expression of tmIgE and its transport to the surface of rMVA-tmIgE-infected cells has been detected by Western blot and cytofluorimetry, respectively. FcεRI activation by tmIgE has been confirmed by the release of ß-hexosaminidase in a cell-to-cell contact assay using human FcεRI-transfected RBL-SX38 cells. The rMVA-tmIgE antitumor vaccination strategy has been investigated in FcεRIα(-/-) human FcεRIα(+) mice, with results indicating a level of protection comparable to that obtained using soluble human IgE tumor cell loading. The rMVA-tmIgE vector represents a device that suits safe IgE-based antitumor vaccines, harboring the possibility to couple tmIgE with other gene insertions that might enhance the antitumor effect, thus bringing the field closer to the clinics.


Assuntos
Vacinas Anticâncer/imunologia , Membrana Celular/imunologia , Imunoglobulina E/imunologia , Neoplasias/imunologia , Vaccinia virus , Animais , Vacinas Anticâncer/biossíntese , Vacinas Anticâncer/genética , Linhagem Celular Tumoral , Membrana Celular/genética , Membrana Celular/metabolismo , Feminino , Humanos , Imunoglobulina E/biossíntese , Imunoglobulina E/genética , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Knockout , Neoplasias/genética , Neoplasias/metabolismo , Proteínas Recombinantes/biossíntese , Proteínas Recombinantes/genética , Proteínas Recombinantes/imunologia , Vacinação
7.
PLoS One ; 6(12): e28001, 2011.
Artigo em Inglês | MEDLINE | ID: mdl-22162996

RESUMO

The interest in broad-range anti-influenza A monoclonal antibodies (mAbs) has recently been strengthened by the identification of anti-hemagglutinin (HA) mAbs endowed with heterosubtypic neutralizing activity to be used in the design of "universal" prophylactic or therapeutic tools. However, the majority of the single mAbs described to date do not bind and neutralize viral isolates belonging to highly divergent subtypes clustering into the two different HA-based influenza phylogenetic groups: the group 1 including, among others, subtypes H1, H2, H5 and H9 and the group 2 including, among others, H3 subtype. Here, we describe a human mAb, named PN-SIA28, capable of binding and neutralizing all tested isolates belonging to phylogenetic group 1, including H1N1, H2N2, H5N1 and H9N2 subtypes and several isolates belonging to group 2, including H3N2 isolates from the first period of the 1968 pandemic. Therefore, PN-SIA28 is capable of neutralizing isolates belonging to subtypes responsible of all the reported pandemics, as well as other subtypes with pandemic potential. The region recognized by PN-SIA28 has been identified on the stem region of HA and includes residues highly conserved among the different influenza subtypes. A deep characterization of PN-SIA28 features may represent a useful help in the improvement of available anti-influenza therapeutic strategies and can provide new tools for the development of universal vaccinal strategies.


Assuntos
Anticorpos Monoclonais/química , Vacinas contra Influenza/uso terapêutico , Influenza Humana/metabolismo , Orthomyxoviridae/genética , Orthomyxoviridae/imunologia , Alanina/genética , Animais , Anticorpos Neutralizantes/química , Linhagem Celular , Análise por Conglomerados , Cães , Glicoproteínas/química , Hemaglutininas Virais/química , Humanos , Influenza Humana/prevenção & controle , Modelos Moleculares , Conformação Molecular , Mutagênese Sítio-Dirigida , Testes de Neutralização , Filogenia , Ligação Proteica
8.
J Virol Methods ; 174(1-2): 22-8, 2011 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-21419167

RESUMO

Pairs of recombinant MVA (Modified Vaccinia Ankara) and FPV (Fowlpox Virus) expressing the same transgene are reasonable candidates for prime/boost regimens, because cross-reacting immune responses between the two vectors, both non-replicative in mammalian hosts, are very limited. The acceptor virus FPD-Red, a derivative of FPV, carrying a red fluorescent protein gene flanked by the homology regions of MVA deletion III, was constructed. The same MVA Transfer Plasmid Green, designed to insert transgenes into the MVA deletion III locus, can therefore be used to transfer transgenes into both acceptor viruses MVA-Red and FPD-Red with the described recently Red-to-Green gene swapping method. Cells infected by either recombinant virus can be sorted differentially by a simple and reliable FACS-based purification protocol. The procedure is carried out in primary chick embryo fibroblasts grown in serum-free media and was applied to the production of three rMVA/rFPV pairs expressing the H5N1 avian influenza antigens M1, M2 and NP. The viral genes were human codon-optimized and expressed at high levels in both chick and mammalian cells. Both single-step and multiple-step growth analyses showed no significant differences in growth due to the transgenes in either rMVA or rFPV derivatives.


Assuntos
Portadores de Fármacos , Vírus da Varíola das Aves Domésticas/genética , Vetores Genéticos , Vaccinia virus/genética , Vacinas Virais/genética , Animais , Linhagem Celular , Galinhas , Terapia Genética/métodos , Humanos , Virus da Influenza A Subtipo H5N1/genética , Transdução Genética , Transgenes , Vacinas Sintéticas/genética , Proteínas Virais/genética
9.
J Gen Virol ; 91(Pt 1): 235-41, 2010 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-19776241

RESUMO

The hypothesis that open conformers of HLA-C on target cells might directly exert an effect on their infectability by human immunodeficiency virus (HIV) has been suggested previously. This was tested by exploiting the peculiar specificity of monoclonal antibody (mAb) L31 for HLA-C open conformers to show that normal levels of Env-driven fusion were restored in HLA-C transfectants of a major histocompatibility complex-deleted (fusion-incompetent) cell line. The physiological relevance of this finding is now confirmed in this report, where small interfering RNA (siRNA) technology was used to silence HLA-C expression in peripheral blood lymphocytes (PBLs) from 11 healthy donors. Infectability by HIV (strains IIIB and Bal and primary isolates) was significantly reduced (P=0.016) in silenced cells compared with cells that maintained HLA-C expression in 10 of the 11 PBL donors. Normal infectability was resumed, together with HLA-C expression, when the effect of siRNA interference waned after several days in culture. Additional confirmation of the HLA-C effect was obtained in several assays employing HLA-C-positive and -negative cell lines, a number of HIV strains and also pseudoviruses. In particular, viruses pseudotyped with env genes from HIV strains AC10 and QH0692.42 were assayed on siRNA-silenced lymphocytes from three healthy donors: the differences in infection with pseudoviruses were even higher than those observed in infections with normal viruses.


Assuntos
Linfócitos T CD4-Positivos/virologia , HIV-1/fisiologia , Antígenos HLA-C/fisiologia , Internalização do Vírus , Anticorpos Monoclonais/imunologia , Linhagem Celular , Células Cultivadas , Inativação Gênica , Antígenos HLA-C/genética , Antígenos HLA-C/imunologia , Humanos , Leucócitos Mononucleares/virologia , RNA Interferente Pequeno/genética , RNA Interferente Pequeno/metabolismo
10.
J Virol Methods ; 163(2): 195-204, 2010 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-19778556

RESUMO

Modified vaccinia virus Ankara (MVA) is employed as a human vaccine vector for the high expression of heterologous genes and the lack of replication in mammalian cells. This study demonstrates that cells infected by recombinant viruses can be obtained by fluorescence-activated cell sorting. Recombinant viruses are generated by a swapping event between a red fluorescent protein gene in the acceptor virus and a plasmid cassette coding for both a green fluorescent marker and a transgene. To prevent the carry-over of parental virus, due to superinfection of the cells harbouring recombinant viruses, the sorting is performed on cells infected at low m.o.i. in the presence of a reversible inhibitor of viral particle release. Terminal dilution cloning is then used to isolate both green and marker-free recombinant viruses, which can be identified by whole-plate fluoroimaging. The differential visualization of all the viral types involved allows a stepwise monitoring of all recombinations and leads to a straightforward and efficient flow cytometry-based cell sorting purification protocol. As an example of the efficacy of this sorting procedure, the construction of rMVA's coding for the rat nuclear protein HMGB1 and H5N1 influenza A virus hemagglutinin is reported. The entire recombinant MVA production process is carried out in serum-free media employing primary chicken embryo fibroblasts (CEF), which are certified for the preparation of human vaccines. This rMVA production method is faster, simpler and more reliable than any other available procedure for obtaining safe vaccine stocks for human use.


Assuntos
Genes Virais , Recombinação Genética , Vacina Antivariólica , Vaccinia virus/crescimento & desenvolvimento , Vaccinia virus/isolamento & purificação , Animais , Técnicas de Cultura de Células/métodos , Células Cultivadas , Embrião de Galinha , Galinhas , Meios de Cultura Livres de Soro , Fibroblastos/virologia , Citometria de Fluxo/métodos , Proteínas de Fluorescência Verde/genética , Proteína HMGB1/genética , Virus da Influenza A Subtipo H5N1/genética , Proteínas Luminescentes/genética , Ratos , Coloração e Rotulagem/métodos , Vaccinia virus/genética , Proteína Vermelha Fluorescente
11.
J Immunol ; 183(7): 4530-6, 2009 Oct 01.
Artigo em Inglês | MEDLINE | ID: mdl-19748979

RESUMO

Working with C57BL/6 mouse tumor models, we had previously demonstrated that vaccination with IgE-coated tumor cells can protect against tumor challenge, an observation that supports the involvement of IgE in antitumor immunity. The adjuvant effect of IgE was shown to result from eosinophil-dependent priming of the T cell-mediated adaptive immune response. The protective effect is likely to be mediated by the interaction of tumor cell-bound IgE with receptors, which then trigger the release of mediators, recruitment of effector cells, cell killing and tumor Ag cross-priming. It was therefore of utmost importance to demonstrate the strict dependence of the protective effect on IgE receptor activation. First, the protective effect of IgE was confirmed in a BALB/c tumor model, in which IgE-loaded modified VV Ankara-infected tumor cells proved to be an effective cellular vaccine. However, the protective effect was lost in Fc(epsilon)RIalpha(-/-) (but not in CD23(-/-)) knockout mice, showing the IgE-Fc(epsilo)nRI interaction to be essential. Moreover, human IgE (not effective in BALB/c mice) had a protective effect in the humanized knockin mouse (Fc(epsilon)RIalpha(-/-) hFc(epsilon)RIalpha(+)). This finding suggests that the adjuvant effect of IgE could be exploited for human therapeutics.


Assuntos
Adjuvantes Imunológicos/fisiologia , Adjuvantes Imunológicos/uso terapêutico , Antineoplásicos/uso terapêutico , Imunoglobulina E/fisiologia , Imunoglobulina E/uso terapêutico , Receptores de IgE/fisiologia , Adenocarcinoma/imunologia , Adenocarcinoma/patologia , Adenocarcinoma/terapia , Adjuvantes Imunológicos/metabolismo , Animais , Antígenos de Neoplasias/imunologia , Antineoplásicos/administração & dosagem , Vacinas Anticâncer/administração & dosagem , Vacinas Anticâncer/genética , Vacinas Anticâncer/imunologia , Linhagem Celular Tumoral , Reagentes de Ligações Cruzadas/administração & dosagem , Reagentes de Ligações Cruzadas/metabolismo , Reagentes de Ligações Cruzadas/uso terapêutico , Feminino , Técnicas de Introdução de Genes , Humanos , Imunoglobulina E/metabolismo , Leucemia Basofílica Aguda/imunologia , Leucemia Basofílica Aguda/patologia , Leucemia Basofílica Aguda/terapia , Linfoma de Células T/imunologia , Linfoma de Células T/patologia , Linfoma de Células T/terapia , Neoplasias Mamárias Experimentais/imunologia , Neoplasias Mamárias Experimentais/patologia , Neoplasias Mamárias Experimentais/terapia , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Endogâmicos C57BL , Camundongos Knockout , Camundongos Transgênicos , Ratos , Receptores de IgE/deficiência , Receptores de IgE/genética
12.
J Virol Methods ; 156(1-2): 37-43, 2009 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-19038289

RESUMO

Modified Vaccinia Virus Ankara (MVA) is employed widely as an experimental and human vaccine vector for its lack of replication in mammalian cells and high expression of heterologous genes. Recombinant MVA technology can be improved greatly by combining transient host-range selection (based on the restoration in MVA of the deleted vaccinia gene K1L) with the differential expression of fluorescent proteins. Recombinant virus results from swapping a red protein gene (in the acceptor virus) with a cassette of the transfer plasmid comprising the transgene and the green marker K1Lgfp (a chimeric gene comprising K1L and EGFP). Recombinant selection is performed in the selective host RK13. Finally, in the non-selective host BHK-21, a single crossover between identical flanking regions excises the marker gene. The three types of viruses involved (red parental, green intermediate and colourless final recombinant) are visualized differentially by fluorescence microscopy or fluoro-imaging of terminal dilution microcultures, leading to a straightforward and efficient purification protocol. This method (Red-to-Green gene swapping) reduces greatly the time needed to obtain marker-free recombinant MVA and increases the reliability of the construction process.


Assuntos
Vírus Defeituosos/genética , Engenharia Genética/métodos , Vaccinia virus/genética , Animais , Linhagem Celular , Cricetinae , DNA Recombinante/genética , DNA Viral/genética , Corantes Fluorescentes/metabolismo , Genes Reporter , Genes Virais , Vetores Genéticos , Microscopia de Fluorescência , Plasmídeos , Coelhos , Especificidade da Espécie , Transfecção , Transgenes
13.
Retrovirology ; 5: 68, 2008 Aug 01.
Artigo em Inglês | MEDLINE | ID: mdl-18673537

RESUMO

BACKGROUND: A recently identified genetic polymorphism located in the 5' region of the HLA-C gene is associated with individual variations in HIV-1 viral load and with differences in HLA-C expression levels. HLA-C has the potential to restrict HIV-1 by presenting epitopes to cytotoxic T cells but it is also a potent inhibitor of NK cells. In addition, HLA-C molecules incorporated within the HIV-1 envelope have been shown to bind to the envelope glycoprotein gp120 and enhance viral infectivity. We investigated this last property in cell fusion assays where the expression of HLA-C was silenced by small interfering RNA sequences. Syncytia formation was analyzed by co-cultivating cell lines expressing HIV-1 gp120/gp41 from different laboratory and primary isolates with target cells expressing different HIV-1 co-receptors. Virus infectivity was analyzed using pseudoviruses. Molecular complexes generated during cell fusion (fusion complexes) were purified and analyzed for their HLA-C content. RESULTS: HLA-C positive cells co-expressing HIV-1 gp120/gp41 fused more rapidly and produced larger syncytia than HLA-C negative cells. Transient transfection of gp120/gp41 from different primary isolates in HLA-C positive cells resulted in a significant cell fusion increase. Fusion efficiency was reduced in HLA-C silenced cells compared to non-silenced cells when co-cultivated with different target cell lines expressing HIV-1 co-receptors. Similarly, pseudoviruses produced from HLA-C silenced cells were significantly less infectious. HLA-C was co-purified with gp120 from cells before and after fusion and was associated with the fusion complex. CONCLUSION: Virionic HLA-C molecules associate to Env and increase the infectivity of both R5 and X4 viruses. Genetic polymorphisms associated to variations in HLA-C expression levels may therefore influence the individual viral set point not only by means of a regulation of the virus-specific immune response but also via a direct effect on the virus replicative capacity. These findings have implications for the understanding of the HIV-1 entry mechanism and of the role of Env conformational modifications induced by virion-associated host proteins.


Assuntos
Proteína gp120 do Envelope de HIV/metabolismo , Infecções por HIV/metabolismo , HIV-1/fisiologia , Antígenos HLA-C/metabolismo , Animais , Células CHO , Fusão Celular , Linhagem Celular , Cricetinae , Cricetulus , Expressão Gênica , Células Gigantes/metabolismo , Células Gigantes/virologia , Proteína gp120 do Envelope de HIV/genética , Infecções por HIV/virologia , HIV-1/patogenicidade , Antígenos HLA-C/genética , Células HeLa , Humanos , Camundongos , Células NIH 3T3 , Ligação Proteica
14.
Proc Natl Acad Sci U S A ; 102(33): 11805-10, 2005 Aug 16.
Artigo em Inglês | MEDLINE | ID: mdl-16091471

RESUMO

The activating receptor NKG2D recognizes a wide range of different ligands, some of which are primarily expressed in "stressed" tissues or on tumor cells. Until now, similar stimulatory effects on natural killer and CD8+ T cells have been described for all NKG2D ligands, and the NKG2D receptor/ligand system has therefore been interpreted as a sensor system involved in tumor immune surveillance and activation of immune responses. We show here that the NKG2D ligands H60 and MIC class 1 chain-related protein A (MICA) can also mediate strong suppressive effects on T cell proliferation. Responsiveness to H60- and MICA-mediated suppression requires IL-10 and involves a receptor other than NKG2D. These findings might provide explanations for the observation that strong in vivo NKG2D ligand expression, such as that on tumor cells, sometimes fails to support effective immune responses and links this observation to a distinct subgroup of NKG2D ligands.


Assuntos
Antígenos de Histocompatibilidade Classe I/metabolismo , Antígenos de Histocompatibilidade Menor/metabolismo , Receptores Imunológicos/metabolismo , Linfócitos T/citologia , Linfócitos T/metabolismo , Animais , Linhagem Celular , Proliferação de Células , Interleucina-10/metabolismo , Ligantes , Camundongos , Camundongos Knockout , Subfamília K de Receptores Semelhantes a Lectina de Células NK , Receptores de Células Matadoras Naturais
15.
J Immunol ; 174(9): 5602-11, 2005 May 01.
Artigo em Inglês | MEDLINE | ID: mdl-15843559

RESUMO

Interaction of secretory IgE with FcepsilonRI is the prerequisite for allergen-driven cellular responses, fundamental events in immediate and chronic allergic manifestations. Previous studies reported the binding of soluble FcepsilonRIalpha to membrane IgE exposed on B cells. In this study, the functional interaction between human membrane IgE and human FcepsilonRI is presented. Four different IgE versions were expressed in mouse B cell lines, namely: a truncation at the Cepsilon2-Cepsilon3 junction of membrane IgE isoform long, membrane IgE isoform long (without Igalpha/Igbeta BCR accessory proteins), and both epsilonBCRs (containing membrane IgE isoforms short and long). All membrane IgE versions activated a rat basophilic leukemia cell line transfected with human FcepsilonRI, as detected by measuring the release of both preformed and newly synthesized mediators. The interaction led also to Ca(2+) responses in the basophil cell line, while membrane IgE-FcepsilonRI complexes were detected by immunoprecipitation. FcepsilonRI activation by membrane IgE occurs in an Ag-independent manner. Noteworthily, human peripheral blood basophils and monocytes also were activated upon contact with cells bearing membrane IgE. In humans, the presence of FcepsilonRI in several cellular entities suggests a possible membrane IgE-FcepsilonRI-driven cell-cell dialogue, with likely implications for IgE homeostasis in physiology and pathology.


Assuntos
Antígenos/fisiologia , Imunoglobulina E/metabolismo , Receptores de Antígenos de Linfócitos B/metabolismo , Receptores de IgE/metabolismo , Animais , Subpopulações de Linfócitos B/imunologia , Subpopulações de Linfócitos B/metabolismo , Basófilos/imunologia , Basófilos/metabolismo , Sítios de Ligação de Anticorpos , Ligação Competitiva/imunologia , Células CHO , Cálcio/metabolismo , Comunicação Celular/imunologia , Contagem de Células , Linhagem Celular Tumoral , Cricetinae , Humanos , Imunoglobulina E/fisiologia , Camundongos , Monócitos/imunologia , Monócitos/metabolismo , Complexos Multiproteicos/metabolismo , Ligação Proteica/imunologia , Isoformas de Proteínas/biossíntese , Isoformas de Proteínas/metabolismo , Ratos , Receptores de Antígenos de Linfócitos B/fisiologia , Receptores de IgE/antagonistas & inibidores , Receptores de IgE/biossíntese , SRS-A/análogos & derivados , SRS-A/metabolismo , Solubilidade , Fatores de Tempo
16.
AIDS ; 17(11): 1621-30, 2003 Jul 25.
Artigo em Inglês | MEDLINE | ID: mdl-12853744

RESUMO

OBJECTIVE: In HIV-positive individuals administration of intermittent interleukin (IL)-2 in addition to highly active antiretroviral therapy (HAART) induces expansion of the peripheral T cell pool with dilution of signal joint T cell receptor excision circles (sjTREC) that cannot be used to measure thymic output. We analysed whether in vitro thymopoiesis could be used to predict in vivo thymic output in IL-2 treated subjects. DESIGN AND METHODS: We correlated the relative variation of peripheral CD4 T cells over 12 months in HIV-positive subjects on HAART or HAART + IL-2 with the mean levels of both sjTREC and T cells developed in chimeric murine foetal thymic organ cultures (FTOC) reconstituted with circulating progenitors. RESULTS: In contrast with HAART treated individuals in which these values were directly correlated, in subjects receiving HAART + IL-2 the increase of CD4 T cells in vivo was correlated to neither sjTREC number nor to reconstitution of FTOC, probably reflecting a main effect of IL-2 in the expansion of the peripheral T cell pool. Nevertheless, addition of IL-2 to HAART determined a significant increase of in vitro thymopoietic potential in individuals with undetectable viraemia. CONCLUSIONS: The increased T cell development in vitro after addition of IL-2 to HAART suggests that intermittent IL-2 administration may exert a positive influence on lymphopoiesis. In two subjects with positive viraemia treated with IL-2 we observed reduced in vitro development of T cell precursors suggesting that the positive influence of IL-2 on thymopoiesis could be secondary to the control of viral replication by HAART. These observations provide novel evidence in support of the potential beneficial use of IL-2 in HAART treated individuals.


Assuntos
Fármacos Anti-HIV/uso terapêutico , Linfócitos T CD4-Positivos/imunologia , Infecções por HIV/tratamento farmacológico , Infecções por HIV/imunologia , HIV-1/fisiologia , Interleucina-2/uso terapêutico , Adulto , Animais , Terapia Antirretroviral de Alta Atividade , Contagem de Linfócito CD4 , Células Cultivadas , Feminino , Citometria de Fluxo , Humanos , Linfopoese/efeitos dos fármacos , Camundongos , Camundongos Endogâmicos C57BL , Pessoa de Meia-Idade , Replicação Viral
17.
J Immunol ; 168(12): 6388-95, 2002 Jun 15.
Artigo em Inglês | MEDLINE | ID: mdl-12055257

RESUMO

Bacterial LPS protects primary human macrophages from infection by CCR5-tropic HIV-1 isolates through the release of the CC chemokines RANTES and macrophage inflammatory protein-1 alpha and -1 beta. Here, we show that LPS also suppresses infection of macrophages by CXCR4-tropic HIV-1 isolates. A marked down-regulation of both CD4 and CXCR4 expression was associated with this effect. Furthermore, a soluble factor(s) released by macrophages upon LPS treatment inhibited infection with CXCR4-tropic HIV-1 isolate viruses in both macrophages and T lymphocytes. Infection of both cell types appeared to be blocked at the level of viral entry and was independent of stromal cell-derived factor-1, the only known natural ligand of CXCR4. Moreover, the suppressive effect of LPS was unrelated to the release of IFN-alpha and -beta, macrophage-derived chemokine, leukemia inhibitory factor, or TNF-alpha. These results suggest the existence of potent HIV-1 inhibitory factor(s), uncharacterized to date, released by activated cells of the mononuclear phagocytic system.


Assuntos
Fármacos Anti-HIV/farmacologia , HIV-1/imunologia , Lipopolissacarídeos/farmacologia , Macrófagos/imunologia , Macrófagos/virologia , Proteínas , Receptores CXCR4/fisiologia , Linfócitos T/imunologia , Linfócitos T/virologia , Antagonistas dos Receptores CCR5 , Antígenos CD4/biossíntese , Antígenos CD4/imunologia , Sistema Livre de Células/imunologia , Células Cultivadas , Quimiocina CCL22 , Quimiocinas CC/fisiologia , Meios de Cultivo Condicionados/farmacologia , Regulação para Baixo/imunologia , Infecções por HIV/imunologia , Infecções por HIV/prevenção & controle , Infecções por HIV/virologia , HIV-1/isolamento & purificação , Humanos , Soros Imunes/farmacologia , Interferon-alfa/antagonistas & inibidores , Interferon-alfa/metabolismo , Interferon-alfa/fisiologia , Interleucina-6 , Fator Inibidor de Leucemia , Lipopolissacarídeos/antagonistas & inibidores , Macrófagos/metabolismo , Chaperonas Moleculares/fisiologia , Monócitos/imunologia , Monócitos/metabolismo , Monócitos/virologia , Receptores CCR5/biossíntese , Receptores CXCR4/antagonistas & inibidores , Receptores CXCR4/biossíntese , Solubilidade , Fatores Supressores Imunológicos/metabolismo , Fatores Supressores Imunológicos/fisiologia , Linfócitos T/metabolismo , Células Tumorais Cultivadas , Fator de Necrose Tumoral alfa/fisiologia
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...