Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 12 de 12
Filtrar
1.
Eur J Immunol ; 51(1): 64-75, 2021 01.
Artigo em Inglês | MEDLINE | ID: mdl-32949466

RESUMO

Immune responses to Epstein-Barr virus (EBV) infection synergize with the main genetic risk factor HLA-DRB1*15:01 (HLA-DR15) to increase the likelihood to develop the autoimmune disease multiple sclerosis (MS) at least sevenfold. In order to gain insights into this synergy, we investigated HLA-DR15 positive human immune compartments after reconstitution in immune-compromised mice (humanized mice) with and without EBV infection. We detected elevated activation of both CD4+ and CD8+ T cells in HLA-DR15 donor-reconstituted humanized mice at steady state, even when compared to immune compartments carrying HLA-DRB1*04:01 (HLA-DR4), which is associated with other autoimmune diseases. Increased CD8+ T cell expansion and activation was also observed in HLA-DR15 donor-reconstituted humanized mice after EBV infection. Despite this higher immune activation, EBV viral loads were less well controlled in the context of HLA-DR15. Indeed, HLA-DR15-restricted CD4+ T cell clones recognized EBV-transformed B cell lines less efficiently and demonstrated cross-reactivity toward allogeneic target cells and one MS autoantigen. These findings suggest that EBV as one of the main environmental risk factors and HLA-DR15 as the main genetic risk factor for MS synergize by priming hyperreactive T-cell compartments, which then control the viral infection less efficiently and contain cross-reactive CD4+ T cell clones.


Assuntos
Infecções por Vírus Epstein-Barr/imunologia , Subtipos Sorológicos de HLA-DR/imunologia , Esclerose Múltipla/imunologia , Imunidade Adaptativa , Animais , Linfócitos T CD4-Positivos/imunologia , Linfócitos T CD8-Positivos/imunologia , Reações Cruzadas , Modelos Animais de Doenças , Infecções por Vírus Epstein-Barr/complicações , Infecções por Vírus Epstein-Barr/virologia , Predisposição Genética para Doença , Antígeno HLA-A2/genética , Antígeno HLA-A2/imunologia , Subtipos Sorológicos de HLA-DR/genética , Herpesvirus Humano 4/imunologia , Humanos , Isoantígenos , Ativação Linfocitária , Camundongos , Camundongos Endogâmicos NOD , Camundongos SCID , Camundongos Transgênicos , Esclerose Múltipla/etiologia , Esclerose Múltipla/genética , Bainha de Mielina/imunologia , Fatores de Risco
2.
Cancer Immunol Immunother ; 68(4): 673-685, 2019 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-30569204

RESUMO

Many pathogens, ranging from viruses to multicellular parasites, promote expansion of MDSCs, which are myeloid cells that exhibit immunosuppressive features. The roles of MDSCs in infection depend on the class and virulence mechanisms of the pathogen, the stage of the disease, and the pathology associated with the infection. This work compiles evidence supported by functional assays on the roles of different subsets of MDSCs in acute and chronic infections, including pathogen-associated malignancies, and discusses strategies to modulate MDSC dynamics to benefit the host.


Assuntos
Doenças Transmissíveis/etiologia , Doenças Transmissíveis/metabolismo , Células Supressoras Mieloides/imunologia , Células Supressoras Mieloides/metabolismo , Doença Aguda , Animais , Biomarcadores , Doença Crônica , Doenças Transmissíveis/tratamento farmacológico , Suscetibilidade a Doenças , Interações Hospedeiro-Patógeno/imunologia , Humanos , Imunomodulação , Terapia de Alvo Molecular , Células Supressoras Mieloides/efeitos dos fármacos
3.
J Virol ; 89(22): 11711-4, 2015 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-26339045

RESUMO

Functions of Epstein-Barr virus (EBV)-encoded RNAs (EBERs) were tested in lymphoblastoid cell lines containing EBER mutants of EBV. Binding of EBER1 to ribosomal protein L22 (RPL22) was confirmed. Deletion of EBER1 or EBER2 correlated with increased levels of cytoplasmic EBV LMP2 RNA and with small effects on specific cellular microRNA (miRNA) levels, but protein levels of LMP1 and LMP2A were not affected. Wild-type EBV and EBER deletion EBV had approximately equal abilities to infect immunodeficient mice reconstituted with a human hematopoietic system.


Assuntos
Herpesvirus Humano 4/genética , RNA Viral/genética , Proteínas da Matriz Viral/metabolismo , Animais , Linhagem Celular , Cisplatino/farmacologia , Humanos , Camundongos , Camundongos Knockout , Camundongos SCID , MicroRNAs/genética , RNA Viral/biossíntese , Proteínas de Ligação a RNA/metabolismo , Proteínas Ribossômicas/metabolismo , Proteína Supressora de Tumor p53/metabolismo , Proteínas da Matriz Viral/genética
4.
Eur J Immunol ; 43(9): 2246-54, 2013 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-23913412

RESUMO

Despite many theoretical incompatibilities between mouse and human cells, mice with reconstituted human immune system components contain nearly all human leukocyte populations. Accordingly, several human-tropic pathogens have been investigated in these in vivo models of the human immune system, including viruses such as human immunodeficiency virus (HIV) and Epstein-Barr virus (EBV), as well as bacteria such as Mycobacterium tuberculosis and Salmonella enterica Typhi. While these studies initially aimed to establish similarities in the pathogenesis of infections between these models and the pathobiology in patients, recent investigations have provided new and interesting functional insights into the protective value of certain immune compartments and altered pathology upon mutant pathogen infections. As more tools and methodologies are developed to make these models more versatile to study human immune responses in vivo, such improvements build toward small animal models with human immune components, which could predict immune responses to therapies and vaccination in human patients.


Assuntos
Infecções por Vírus Epstein-Barr/imunologia , Infecções por HIV/imunologia , Leucócitos/imunologia , Tuberculose/imunologia , Febre Tifoide/imunologia , Animais , Modelos Animais de Doenças , HIV-1/imunologia , Herpesvirus Humano 4/imunologia , Humanos , Camundongos , Mycobacterium tuberculosis/imunologia , Salmonella typhi/imunologia
5.
Proc Natl Acad Sci U S A ; 108(18): 7499-504, 2011 May 03.
Artigo em Inglês | MEDLINE | ID: mdl-21502499

RESUMO

Recombinant adenoviruses (rAds) based on types 5 (rAd5) and 35 (rAd35) have emerged as important vaccine delivery vectors in clinical testing for a variety of pathogens. A major difference between these vectors is their binding to cellular receptors used for infection. Whereas rAd5 binds coxsackie-adenovirus receptor (CAR), rAd35 binds the complement regulatory protein CD46. Although rAd35 infected and phenotypically matured human blood dendritic cells (DCs) more efficiently than rAd5, we show here that rAd35 markedly suppressed DC-induced activation of naive CD4(+) T cells. rAd35 specifically blocked both DCs and anti-CD3/CD28 mAb-induced naive T-cell proliferation and IL-2 production. This effect was also observed in CD4(+) memory T cells but to a lesser extent. The suppression occurred by rAd35 binding to CD46 on T cells and was independent of infection. CD46 engagement with mAb mimicked the effects of rAd35 and also led to deficient NF-κB nuclear translocation. In contrast, rAd5 and rAd35 vectors with ablated CD46 binding did not inhibit T-cell activation. Our findings provide insights into the basic biology of adenoviruses and indicate that CD46 binding may have an impact on the generation of primary CD4(+) T-cell responses by Ad35.


Assuntos
Adenoviridae/metabolismo , Linfócitos T CD4-Positivos/metabolismo , Vetores Genéticos/metabolismo , Ativação Linfocitária/fisiologia , Proteína Cofatora de Membrana/metabolismo , Receptores Virais/metabolismo , Adenoviridae/genética , Western Blotting , Linfócitos T CD4-Positivos/imunologia , Proteína de Membrana Semelhante a Receptor de Coxsackie e Adenovirus , Células Dendríticas/metabolismo , Citometria de Fluxo , Humanos
6.
Immunology ; 134(3): 257-69, 2011 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-21977996

RESUMO

Interferon-α (IFN-α) produced at high levels by human plasmacytoid dendritic cells (pDCs) can specifically regulate B-cell activation to Toll-like receptor (TLR) 7/8 stimulation. To explore the influence of IFN-α and pDCs on B-cell functions in vivo, studies in non-human primates that closely resemble humans in terms of TLR expression on different subsets of immune cells are valuable. Here, we performed a side-by side comparison of the response pattern between human and rhesus macaque B cells and pDCs in vitro to well-defined TLR ligands and tested whether IFN-α enhanced B-cell function comparably. We found that both human and rhesus B cells proliferated while pDCs from both species produced high levels of IFN-α in response to ligands targeting TLR7/8 and TLR9. Both human and rhesus B-cell proliferation to TLR7/8 ligand and CpG class C was significantly increased in the presence of IFN-α. Although both human and rhesus B cells produced IgM upon stimulation, only human B cells acquired high expression of CD27 associated with plasmablast formation. Instead, rhesus B-cell differentiation and IgM levels correlated to down-regulation of CD20. These data suggest that the response pattern of human and rhesus B cells and pDCs to TLR7/8 and TLR9 is similar, although some differences in the cell surface phenotype of the differentiating cells exist. A more thorough understanding of potential similarities and differences between human and rhesus cells and their response to potential vaccine components will provide important information for translating non-human primate studies into human trials.


Assuntos
Linfócitos B/imunologia , Células Dendríticas/imunologia , Interferon-alfa/imunologia , Macaca mulatta/imunologia , Receptores Toll-Like/imunologia , Animais , Antígenos CD20/imunologia , Antígenos CD20/metabolismo , Linfócitos B/efeitos dos fármacos , Linfócitos B/metabolismo , Diferenciação Celular/efeitos dos fármacos , Diferenciação Celular/imunologia , Proliferação de Células/efeitos dos fármacos , Células Cultivadas , Células Dendríticas/efeitos dos fármacos , Células Dendríticas/metabolismo , Ensaio de Imunoadsorção Enzimática , Citometria de Fluxo , Humanos , Imidazóis/farmacologia , Imunoglobulina M/imunologia , Imunoglobulina M/metabolismo , Interferon-alfa/metabolismo , Interferon-alfa/farmacologia , Macaca mulatta/metabolismo , Oligodesoxirribonucleotídeos/farmacologia , Quinolinas/farmacologia , Receptor 7 Toll-Like/agonistas , Receptor 7 Toll-Like/imunologia , Receptor 7 Toll-Like/metabolismo , Receptor 8 Toll-Like/agonistas , Receptor 8 Toll-Like/imunologia , Receptor 8 Toll-Like/metabolismo , Receptor Toll-Like 9/agonistas , Receptor Toll-Like 9/imunologia , Receptor Toll-Like 9/metabolismo , Receptores Toll-Like/agonistas , Receptores Toll-Like/metabolismo , Membro 7 da Superfamília de Receptores de Fatores de Necrose Tumoral/imunologia , Membro 7 da Superfamília de Receptores de Fatores de Necrose Tumoral/metabolismo
7.
J Immunol ; 182(4): 1991-2001, 2009 Feb 15.
Artigo em Inglês | MEDLINE | ID: mdl-19201852

RESUMO

Selected TLR ligands are under evaluation as vaccine adjuvants and are known to activate dendritic cells (DCs) and B cells to affect vaccine-induced Ab responses. However, the relative contribution of the two main human DC subsets, myeloid (MDCs) and plasmacytoid (PDCs), in supporting B cell responses to TLR ligands remains poorly understood. We found that PDCs but not MDCs markedly enhanced B cell proliferation in response to TLR7/8-L, an imidazoquinoline derivative, and to a lesser extent to TLR9 ligands (CpG ODN classes A, B, and C). PDCs strongly enhanced TLR7/8-L-induced proliferation of both memory and naive B cells but were only able to support memory cells to differentiate to CD27(high) plasmablasts. In response to TLR7/8 stimulation, PDCs mediated the up-regulation of transcription factors B lymphocyte-induced maturation protein 1 and X-box binding protein 1 and enhanced differentiation of B cells into IgM-, IgG-, and IgA-producing cells. Type I IFN produced to high levels by PDCs was the principal mediator of the effects on TLR7/8 stimulation. Although MDCs expressed higher levels of the known B cell growth factors IL-6, IL-10, and B cell-activating factor in response to TLR7/8 stimulation, they were unable to enhance B cell responses in this system. These data help decipher the different roles of PDCs and MDCs for modulating human B cell responses and can contribute to selection of specific TLR ligands as vaccine adjuvants.


Assuntos
Linfócitos B/imunologia , Células Dendríticas/imunologia , Ativação Linfocitária/imunologia , Células Mieloides/imunologia , Receptores Toll-Like/imunologia , Diferenciação Celular/imunologia , Proliferação de Células , Células Dendríticas/citologia , Humanos , Interferon gama/biossíntese , Ligantes , Células Mieloides/citologia , Reação em Cadeia da Polimerase Via Transcriptase Reversa
8.
Blood Adv ; 3(7): 1129-1144, 2019 04 09.
Artigo em Inglês | MEDLINE | ID: mdl-30952679

RESUMO

Infectious mononucleosis, caused by infection with the human gamma-herpesvirus Epstein-Barr virus (EBV), manifests with one of the strongest CD8+ T-cell responses described in humans. The resulting T-cell memory response controls EBV infection asymptomatically in the vast majority of persistently infected individuals. Whether and how dendritic cells (DCs) contribute to the priming of this near-perfect immune control remains unclear. Here we show that of all the human DC subsets, plasmacytoid DCs (pDCs) play a central role in the detection of EBV infection in vitro and in mice with reconstituted human immune system components. pDCs respond to EBV by producing the interferon (IFN) subtypes α1, α2, α5, α7, α14, and α17. However, the virus curtails this type I IFN production with its latent EBV gene products EBNA3A and EBNA3C. The induced type I IFNs inhibit EBV entry and the proliferation of latently EBV-transformed B cells but do not influence lytic reactivation of the virus in vitro. In vivo, exogenous IFN-α14 and IFN-α17, as well as pDC expansion, delay EBV infection and the resulting CD8+ T-cell expansion, but pDC depletion does not significantly influence EBV infection. Thus, consistent with the observation that primary immunodeficiencies compromising type I IFN responses affect only alpha- and beta-herpesvirus infections, we found that EBV elicits pDC responses that transiently suppress viral replication and attenuate CD8+ T-cell expansion but are not required to control primary infection.


Assuntos
Células Dendríticas/imunologia , Infecções por Vírus Epstein-Barr/imunologia , Interferon Tipo I/biossíntese , Animais , Linfócitos T CD8-Positivos/patologia , Proliferação de Células , Humanos , Interferon Tipo I/farmacologia , Camundongos , Internalização do Vírus/efeitos dos fármacos , Replicação Viral
9.
Nat Commun ; 9(1): 1980, 2018 05 17.
Artigo em Inglês | MEDLINE | ID: mdl-29773792

RESUMO

The type I interferon (IFN) system plays an important role in controlling herpesvirus infections, but it is unclear which IFN-mediated effectors interfere with herpesvirus replication. Here we report that human myxovirus resistance protein B (MxB, also designated Mx2) is a potent human herpesvirus restriction factor in the context of IFN. We demonstrate that ectopic MxB expression restricts a range of herpesviruses from the Alphaherpesvirinae and Gammaherpesvirinae, including herpes simplex virus 1 and 2 (HSV-1 and HSV-2), and Kaposi's sarcoma-associated herpesvirus (KSHV). MxB restriction of HSV-1 and HSV-2 requires GTPase function, in contrast to restriction of lentiviruses. MxB inhibits the delivery of incoming HSV-1 DNA to the nucleus and the appearance of empty capsids, but not the capsid delivery to the cytoplasm or tegument dissociation from the capsid. Our study identifies MxB as a potent pan-herpesvirus restriction factor which blocks the uncoating of viral DNA from the incoming viral capsid.


Assuntos
Infecções por Herpesviridae/imunologia , Herpesviridae/fisiologia , Interferon Tipo I/imunologia , Proteínas de Resistência a Myxovirus/imunologia , Replicação Viral/imunologia , Capsídeo/imunologia , Proteínas do Capsídeo/imunologia , Linhagem Celular Tumoral , Núcleo Celular/imunologia , Núcleo Celular/virologia , Citoplasma , DNA Viral/imunologia , Células HEK293 , Herpesviridae/patogenicidade , Infecções por Herpesviridae/virologia , Humanos , Proteínas de Resistência a Myxovirus/genética , RNA Interferente Pequeno/metabolismo , Desenvelopamento do Vírus/imunologia
10.
Curr Opin Virol ; 13: 6-10, 2015 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-25846986

RESUMO

Epstein Barr virus (EBV) was the first human tumor virus to be described. Despite its discovery now more than fifty years ago, immune control of this virus is still not very well understood and no vaccine is available. This knowledge gap is due in part to the lack of a preclinical small animal model which can faithfully recapitulate EBV infection and immune control, and would allow testing of EBV specific vaccine candidates. With the advent of mice with reconstituted human immune system compartments (HIS mice) during the past decade this is changing. We will discuss which aspects of EBV infection and its immune control can already be modeled in HIS mice, and which shortcomings still need to be overcome in order to recapitulate the immunobiology of oncogenic EBV infection.


Assuntos
Modelos Animais de Doenças , Infecções por Vírus Epstein-Barr/virologia , Herpesvirus Humano 4/fisiologia , Animais , Infecções por Vírus Epstein-Barr/imunologia , Herpesvirus Humano 4/genética , Humanos , Camundongos , Linfócitos T/imunologia
11.
J Leukoc Biol ; 89(6): 811-21, 2011 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-21233412

RESUMO

The development and quality of a humoral immune response are largely influenced by the environment that supports the activation of naïve B cells. Human PDCs, through their unique capacity to produce high levels of IFN-α, have been shown earlier to enhance B cell responses stimulated by selected TLR ligands. In this study, we investigated whether PDCs also promote B cell activation induced by Th cell interactions and BCR ligation. Sorted human naive CD19(+) CD27(-) B cells were activated in vitro with anti-Ig and irradiated CD4(+) T cells. Under these conditions, the presence of supernatants from TLR-stimulated PDCs increased B cell proliferation, the frequency of B cells that differentiated to CD27(high) CD38(high) cells, and secretion of IgM. Similar results were observed when the B cells were activated in the presence of purified IFN-α. In contrast, supernatants from stimulated MDCs did not augment these functions. Also, IFN-α treatment of B cells up-regulated the expression of costimulatory molecule CD86 but not CD40, CD80, MHC class II, or CD25. Although direct IFN-α exposure of T cells suppressed their proliferative capacity, IFN-α treatment of B cells led to a small increase in their capacity to induce superantigen-driven activation of autologous CD4(+) T cells. In summary, PDCs, via their production of IFN-α, may render B cells more responsive to T cell contact, which in turn, facilitates B cell proliferation and differentiation to antibody-producing cells.


Assuntos
Células Produtoras de Anticorpos/imunologia , Linfócitos B/imunologia , Diferenciação Celular/imunologia , Células Dendríticas/metabolismo , Interferon-alfa/metabolismo , Linfócitos T/imunologia , Células Cultivadas , Citocinas/metabolismo , Humanos , Imunoglobulina M/metabolismo , Memória Imunológica , Ativação Linfocitária
12.
J Virol ; 79(13): 8454-69, 2005 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-15956589

RESUMO

We studied the effect of entry inhibitors on 58 virus isolates derived during acute and chronic infection to validate these inhibitors in vitro and to probe whether viruses at early and chronic disease stages exhibit general differences in the interaction with entry receptors. We included members of all types of inhibitors currently identified: (i) agents that block gp120 binding to CD4 (CD4-IgG2 and monoclonal antibody [MAb] IgG1b12), (ii) compounds that block the interaction with CCR5 (the chemokine RANTES/CCL5, the small-molecule inhibitor AD101, and the anti-CCR5 antibody PRO 140), (iii) the fusion inhibitor enfuvirtide (T-20), and (iv) neutralizing antibodies directed against gp120 (MAb 2G12) and gp41 (MAbs 2F5 and 4E10). No differences between viruses from acute and chronic infections in the susceptibility to inhibitors targeting the CD4 binding site, CCR5, or fusion or to MAb 2G12 were apparent, rendering treatment with entry inhibitors feasible across disease stages. The notable exceptions were antibodies 2F5 and 4E10, which were more potent in inhibiting viruses from acute infection (P = 0.0088 and 0.0005, respectively), although epitopes of these MAbs were equally well preserved in both groups. Activities of these MAbs correlated significantly with each other, suggesting that common features of the viral envelope modulate their potencies.


Assuntos
Síndrome da Imunodeficiência Adquirida/virologia , Fármacos Anti-HIV/farmacologia , Inibidores da Fusão de HIV/farmacologia , Infecções por HIV/virologia , HIV-1/isolamento & purificação , Doença Aguda , Adulto , Antagonistas dos Receptores CCR5 , Doença Crônica , Enfuvirtida , Feminino , Anticorpos Anti-HIV/sangue , Proteína gp41 do Envelope de HIV/farmacologia , HIV-1/efeitos dos fármacos , HIV-1/fisiologia , Humanos , Masculino , Pessoa de Meia-Idade , Fragmentos de Peptídeos/farmacologia , Análise de Regressão
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA