Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 11 de 11
Filtrar
Mais filtros











Base de dados
Intervalo de ano de publicação
1.
J Immunother Cancer ; 11(8)2023 08.
Artigo em Inglês | MEDLINE | ID: mdl-37607769

RESUMO

BACKGROUND: Multiple myeloma (MM) cancers originate from plasma cells that have passed through the germinal center reaction where somatic hypermutation of Ig V regions takes place. Myeloma protein V regions often express many mutations and are thus a rich source of neoantigens (traditionally called idiotopes (Id)). Therefore, these are highly tumor-specific and excellent targets for immunotherapy. METHODS: We have developed a DNA Id vaccine which as translated protein targets conventional dendritic cells (cDC) for CCL3-mediated delivery of myeloma protein V regions in a single-chain fragment variable (scFv) format. Vaccine efficacy was studied in the mouse MM model, mineral oil-induced plasmacytoma 315.BM. RESULTS: The Id vaccine protected mice against a challenge with MM cells. Moreover, the vaccine had a therapeutic effect. However, in some of the vaccinated mice, MM cells not producing H chains escaped rejection, resulting in free light chain (FLC) MM. Depletion of CD8+ T cells abrogated vaccine efficacy, and protection was observed to be dependent on cDC1s, using Batf3-/- mice. Modifications of scFv in the vaccine demonstrated that CD8+ T cells were specific for two mutated VH sequences. CONCLUSIONS: VH neoantigen-specific CD8+ T cells elicited by CCL3-containing Id vaccines had a therapeutic effect against MM in a mouse model. MM cells could escape rejection by losing expression of the H chain, thus giving rise to FLC MM.


Assuntos
Mieloma Múltiplo , Vacinas de DNA , Animais , Camundongos , Mieloma Múltiplo/terapia , Linfócitos T CD8-Positivos , Imunoterapia , Células Dendríticas
2.
Cell Rep ; 39(9): 110901, 2022 05 31.
Artigo em Inglês | MEDLINE | ID: mdl-35649357

RESUMO

Antibodies are important for vaccine efficacy. Targeting antigens to antigen-presenting cells (APCs) increases antibody levels. Here, we explore the role of antigen valency in MHC class II (MHCII)-targeted vaccines delivered as DNA. We design heterodimeric proteins that carry either two identical (bivalent vaccines), or two different antigens (monovalent vaccines). Bivalent vaccines with two identical influenza hemagglutinins (HA) elicit higher amounts of anti-HA antibodies in mice than monovalent versions with two different HAs. Bivalent vaccines increase the levels of germinal center (GC) B cells and long-lived plasma cells. Only HA-bivalent vaccines completely protect mice against challenge with homologous influenza virus. Similar results are obtained with other antigens by targeting CD11c and Xcr1 on dendritic cells (DCs) or when administering the vaccine as protein with adjuvant. Bivalency probably increases B cell responses by cross-linking BCRs in readily observable DC-B cell synapses. These results are important for generating potent APC-targeted vaccines.


Assuntos
Vacinas Anticâncer , Vacinas contra Influenza , Vacinas de DNA , Animais , Anticorpos Antivirais , Células Apresentadoras de Antígenos , Hemaglutininas , Camundongos , Vacinas Combinadas , Vacinas de DNA/genética
3.
EMBO Rep ; 22(4): e52196, 2021 04 07.
Artigo em Inglês | MEDLINE | ID: mdl-33719206

RESUMO

T and B cells continually recirculate between blood and secondary lymphoid organs. To promote their trans-endothelial migration (TEM), chemokine receptors control the activity of RHO family small GTPases in part via GTPase-activating proteins (GAPs). T and B cells express several RHO-GAPs, the function of most of which remains unknown. The ARHGAP45 GAP is predominantly expressed in hematopoietic cells. To define its in vivo function, we describe two mouse models where ARHGAP45 is ablated systemically or selectively in T cells. We combine their analysis with affinity purification coupled to mass spectrometry to determine the ARHGAP45 interactome in T cells and with time-lapse and reflection interference contrast microscopy to assess the role of ARGHAP45 in T-cell polarization and motility. We demonstrate that ARHGAP45 regulates naïve T-cell deformability and motility. Under physiological conditions, ARHGAP45 controls the entry of naïve T and B cells into lymph nodes whereas under competitive repopulation it further regulates hematopoietic progenitor cell engraftment in the bone marrow, and T-cell progenitor thymus seeding. Therefore, the ARGHAP45 GAP controls multiple key steps in the life of T and B cells.


Assuntos
Linfócitos T , Internalização do Vírus , Animais , Linfócitos B , Movimento Celular , Proteínas Ativadoras de GTPase/genética , Linfonodos , Camundongos , Timo
4.
Mol Ther Methods Clin Dev ; 17: 378-392, 2020 Jun 12.
Artigo em Inglês | MEDLINE | ID: mdl-32128342

RESUMO

Immunogenicity of DNA vaccines can be increased by constructing the DNA in such a way that it encodes secreted homodimeric fusion proteins that target antigen-presenting cells (APCs). In this study, we have developed novel APC-targeting vaccine molecules with an increased flexibility due to introduction of a heterodimerization motif. The heterodimeric proteins permit four different fusions within a single molecule, thus allowing expression of two different APC-targeting moieties and two different antigens. Two types of heterodimeric fusion proteins were developed that employed either the ACID/BASE or the Barnase/Barstar motifs, respectively. The ACID/BASE heterodimeric vaccines conferred protection against challenges with either influenza virus or tumor cells in separate preclinical models. The ACID/BASE motif was flexible since a large number of different targeting moieties and antigens could be introduced with maintenance of specificity, antigenicity, and secretion. APC-targeting ACID/BASE vaccines expressing two different antigens induced antibody and T cell responses against either of the two antigens. Heterodimeric ACID/BASE DNA vaccines were of approximately the same potency as previously reported homodimeric DNA vaccines. The flexibility and potency of the ACID/BASE format suggest that it could be a useful platform for DNA vaccines that encode APC-targeting fusion proteins.

5.
Immunohorizons ; 2(1): 38-53, 2018 01 18.
Artigo em Inglês | MEDLINE | ID: mdl-31022690

RESUMO

Upon APC-targeted DNA vaccination, transfected cells secrete fusion proteins with targeting units specific for surface molecules on APC. In this study, we have tested several different targeting units for their ability to influence the magnitude and subclass of Ab responses to hemagglutinin from influenza A virus. The experiments employed bivalent homodimeric Ig-based molecules (vaccibodies). The overall efficiency in BALB/c mice depended on the targeting units in the following order: αMHC class II > αCD11c > αCD40 > Xcl-1 = MIP-1α > FliC > GM-CSF > Flt-3L > αDEC205. GM-CSF induced mainly IgG1, whereas Xcl1, MIP-1α, αCD40, and αDEC205 induced predominantly IgG2a. A more balanced mixture of IgG1 and IgG2a was observed with αCD11c, αMHC class II, Flt-3L, and FliC. Similar results of IgG subclass-skewing were obtained in Th1-prone C57BL/6 mice with a more limited panel of vaccines. IgG1 responses in BALB/c occurred early after immunization but declined relatively rapidly over time. IgG2a responses appeared later but lasted longer (>252 d) than IgG1 responses. The most efficient targeting units elicited short- and long-term protection against PR8 influenza (H1N1) virus in BALB/c mice. The results suggest that targeting of Xcr1+ conventional type 1 dendritic cells preferentially induces IgG2a responses, whereas simultaneous targeting of several dendritic cell subtypes also induces IgG1 responses. The induction of distinct subclass profiles by different surface molecules supports the APC-B cell synapse hypothesis. The results may contribute to generation of more potent DNA vaccines that elicit high levels of Abs with desired biologic effector functions.


Assuntos
Células Apresentadoras de Antígenos/imunologia , Vírus da Influenza A/imunologia , Vacinas contra Influenza/imunologia , Vacinas de DNA/imunologia , Animais , Anticorpos Antivirais/biossíntese , Formação de Anticorpos , Linhagem Celular , Células Dendríticas/imunologia , Células HEK293 , Hemaglutininas/imunologia , Humanos , Vacinas contra Influenza/genética , Macrófagos , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Endogâmicos C57BL , Transfecção
6.
Sci Rep ; 6: 38597, 2016 12 09.
Artigo em Inglês | MEDLINE | ID: mdl-27934903

RESUMO

Bats carry and shed many emerging infectious disease agents including Ebola virus and SARS-like Coronaviruses, yet they rarely display clinical symptoms of infection. Bat epithelial or fibroblast cell lines were previously established to study the bat immune response against viral infection. However, the lack of professional immune cells such as dendritic cells (DC) and macrophages has greatly limited the significance of current investigations. Using Pteropus alecto (P. alecto) GM-CSF plus IL4, FLT3L and CSF-1, we successfully generated bat bone marrow-derived DC and macrophages. Cells with the phenotype, morphology and functional features of monocyte-derived DC, bona fide DC or macrophages were obtained in GM-CSF/IL4, FLT3L or CSF-1 cultures, respectively. The successful generation of the first bat bone marrow-derived immune cells paves the way to unlocking the immune mechanisms that confer host resilience to pathogens in bats.


Assuntos
Quirópteros/imunologia , Imunidade , Animais , Biomarcadores , Quirópteros/metabolismo , Citocinas/metabolismo , Células Dendríticas/imunologia , Células Dendríticas/metabolismo , Imunidade Inata , Imunofenotipagem , Macrófagos/imunologia , Macrófagos/metabolismo , Fagocitose , Fenótipo , Linfócitos T/imunologia , Linfócitos T/metabolismo
7.
J Immunol ; 194(12): 5895-902, 2015 Jun 15.
Artigo em Inglês | MEDLINE | ID: mdl-25941327

RESUMO

The development of vaccines inducing efficient CD8(+) T cell responses is the focus of intense research. Dendritic cells (DCs) expressing the XCR1 chemokine receptor, also known as CD103(+) or CD8α(+) DCs, excel in the presentation of extracellular Ags to CD8(+) T cells. Because of its high numbers of DCs, including XCR1(+) DCs, the skin dermis is an attractive site for vaccine administration. By creating laser-generated micropores through the epidermis, we targeted a model protein Ag fused to XCL1, the ligand of XCR1, to dermal XCR1(+) DCs and induced Ag-specific CD8(+) and CD4(+) T cell responses. Efficient immunization required the emigration of XCR1(+) dermal DCs to draining lymph nodes and occurred irrespective of TLR signaling. Moreover, a single intradermal immunization protected mice against melanoma tumor growth in prophylactic and therapeutic settings, in the absence of exogenous adjuvant. The mild inflammatory milieu created in the dermis by skin laser microporation itself most likely favored the development of potent T cell responses in the absence of exogenous adjuvants. The existence of functionally equivalent XCR1(+) dermal DCs in humans should permit the translation of laser-assisted intradermal delivery of a tumor-specific vaccine targeting XCR1(+) DCs to human cancer immunotherapy. Moreover, considering that the use of adjuvants in vaccines is often associated with safety issues, the possibility of inducing protective responses against melanoma tumor growth independently of the administration of exogenous adjuvants should facilitate the development of safer vaccines.


Assuntos
Vacinas Anticâncer/imunologia , Células Dendríticas/imunologia , Células Dendríticas/metabolismo , Neoplasias/imunologia , Receptores de Quimiocinas/metabolismo , Proteínas Adaptadoras de Transporte Vesicular/metabolismo , Animais , Vacinas Anticâncer/administração & dosagem , Quimiocinas C/genética , Quimiocinas C/metabolismo , Modelos Animais de Doenças , Injeções Intradérmicas , Melanoma Experimental , Camundongos , Camundongos Knockout , Fator 88 de Diferenciação Mieloide/metabolismo , Neoplasias/patologia , Neoplasias/terapia , Ovalbumina/genética , Ovalbumina/imunologia , Ligação Proteica , Receptores de Quimiocinas/genética , Subpopulações de Linfócitos T/imunologia , Carga Tumoral/imunologia
8.
J Virol ; 84(23): 12255-64, 2010 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-20861242

RESUMO

In cells infected with the Kaposi's sarcoma-associated herpesvirus (KSHV), CSL/CBF1 signaling is essential for viral replication and promotes the survival of KSHV-infected cells. CSL/CBF1 is a DNA adaptor molecule which recruits coactivator and corepressor complexes to regulate viral and cellular gene transcription and which is a major downstream effector molecule of activated Notch. The interaction of KSHV RTA and LANA with CSL/CBF1 has been shown to balance the lytic and latent viral life cycle. Here we report that a third KSHV protein, viral interferon regulatory factor 4 (vIRF4/K10), but none of the three other KSHV-encoded vIRFs, interacts with CSL/CBF1. Two regions of vIRF4 with dissimilar affinities contribute to CSL/CBF1 binding. Similar to Notch, vIRF4 targets the hydrophobic pocket in the beta trefoil domain of CSL/CBF1 through a short peptide motif which closely resembles a motif found in Notch but does not strictly follow the ΦWΦP consensus conserved in human and mouse Notch proteins. Our results suggest that vIRF4 might compete with Notch for CSL/CBF1 binding and signaling.


Assuntos
Herpesvirus Humano 8/metabolismo , Proteína de Ligação a Sequências Sinal de Recombinação J de Imunoglobina/metabolismo , Fatores Reguladores de Interferon/metabolismo , Transdução de Sinais/fisiologia , Proteínas Virais/metabolismo , Ligação Competitiva , Linhagem Celular Tumoral , Cromatografia de Afinidade , Primers do DNA/genética , Eletroforese em Gel de Poliacrilamida , Ensaio de Desvio de Mobilidade Eletroforética , Humanos , Immunoblotting , Imunoprecipitação , Plasmídeos/genética , Ligação Proteica , Receptores Notch/metabolismo , Técnicas do Sistema de Duplo-Híbrido
9.
PLoS Pathog ; 5(9): e1000570, 2009 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-19730696

RESUMO

Herpesviruses constitute a family of large DNA viruses widely spread in vertebrates and causing a variety of different diseases. They possess dsDNA genomes ranging from 120 to 240 kbp encoding between 70 to 170 open reading frames. We previously reported the protein interaction networks of two herpesviruses, varicella-zoster virus (VZV) and Kaposi's sarcoma-associated herpesvirus (KSHV). In this study, we systematically tested three additional herpesvirus species, herpes simplex virus 1 (HSV-1), murine cytomegalovirus and Epstein-Barr virus, for protein interactions in order to be able to perform a comparative analysis of all three herpesvirus subfamilies. We identified 735 interactions by genome-wide yeast-two-hybrid screens (Y2H), and, together with the interactomes of VZV and KSHV, included a total of 1,007 intraviral protein interactions in the analysis. Whereas a large number of interactions have not been reported previously, we were able to identify a core set of highly conserved protein interactions, like the interaction between HSV-1 UL33 with the nuclear egress proteins UL31/UL34. Interactions were conserved between orthologous proteins despite generally low sequence similarity, suggesting that function may be more conserved than sequence. By combining interactomes of different species we were able to systematically address the low coverage of the Y2H system and to extract biologically relevant interactions which were not evident from single species.


Assuntos
Herpesviridae/genética , Mapeamento de Interação de Proteínas/métodos , Proteínas Virais/genética , Proteínas Virais/metabolismo , Análise por Conglomerados , Evolução Molecular , Células HeLa , Herpesviridae/metabolismo , Herpesvirus Humano 1/genética , Herpesvirus Humano 3/genética , Herpesvirus Humano 4/genética , Herpesvirus Humano 8/genética , Humanos , Imuno-Histoquímica , Muromegalovirus/genética , Filogenia , Transdução de Sinais , Proteínas do Core Viral/genética , Proteínas do Core Viral/metabolismo , Vírion/metabolismo
10.
J Virol ; 83(2): 1140-6, 2009 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-18987133

RESUMO

Upon viral infection, the major defense mounted by the host immune system is activation of the interferon (IFN)-mediated antiviral pathway, which is mediated by IFN regulatory factors (IRFs). In order to complete their life cycle, viruses must modulate host IFN-mediated immune responses. Despite its association with significant human health problems, activities of Epstein-Barr virus (EBV), a human tumor-inducing herpesvirus, to evade host IFN-mediated innate immunity have not been well characterized. To search for EBV genes that block IFN signal transduction, we carried out a screening of EBV open reading frames for their abilities to block IFN-alpha/beta-mediated luciferase expression upon Sendai virus infection. This screening demonstrates that EBV LF2 tegument protein specifically interacts with the central inhibitory association domain of IRF7, and this interaction leads to inhibition of the dimerization of IRF7, which suppresses IFN-alpha production and IFN-mediated immunity. This demonstrates a novel immune evasion mechanism of EBV LF2 in blocking cellular IRF7-mediated innate immunity.


Assuntos
Glicoproteínas/metabolismo , Herpesvirus Humano 4/fisiologia , Interferon Tipo I/antagonistas & inibidores , Proteínas Virais/metabolismo , Linhagem Celular , Genes Reporter , Glicoproteínas/imunologia , Herpesvirus Humano 4/imunologia , Humanos , Fator Regulador 7 de Interferon/antagonistas & inibidores , Fator Regulador 7 de Interferon/metabolismo , Luciferases/genética , Luciferases/metabolismo , Ligação Proteica , Proteínas Virais/imunologia
11.
Science ; 311(5758): 239-42, 2006 Jan 13.
Artigo em Inglês | MEDLINE | ID: mdl-16339411

RESUMO

The comprehensive yeast two-hybrid analysis of intraviral protein interactions in two members of the herpesvirus family, Kaposi sarcoma-associated herpesvirus (KSHV) and varicella-zoster virus (VZV), revealed 123 and 173 interactions, respectively. Viral protein interaction networks resemble single, highly coupled modules, whereas cellular networks are organized in separate functional submodules. Predicted and experimentally verified interactions between KSHV and human proteins were used to connect the viral interactome into a prototypical human interactome and to simulate infection. The analysis of the combined system showed that the viral network adopts cellular network features and that protein networks of herpesviruses and possibly other intracellular pathogens have distinguishing topologies.


Assuntos
Herpesvirus Humano 3/metabolismo , Herpesvirus Humano 8/metabolismo , Proteoma/metabolismo , Proteínas Virais/metabolismo , Linhagem Celular , Clonagem Molecular , Humanos , Fases de Leitura Aberta , Ligação Proteica , Biologia de Sistemas , Técnicas do Sistema de Duplo-Híbrido
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA