Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 20
Filtrar
1.
Cancer Immunol Immunother ; 67(9): 1425-1436, 2018 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-30019146

RESUMO

There has recently been a paradigm shift in the field of dendritic cell (DC)-based immunotherapy, where several clinical studies have confirmed the feasibility and advantageousness of using directly isolated human blood-derived DCs over in vitro differentiated subsets. There are two major DC subsets found in blood; plasmacytoid DCs (pDCs) and myeloid DCs (mDCs), and both have been tested clinically. CD1c+ mDCs are highly efficient antigen-presenting cells that have the ability to secrete IL-12p70, while pDCs are professional IFN-α-secreting cells that are shown to induce innate immune responses in melanoma patients. Hence, combining mDCs and pDCs poses as an attractive, multi-functional vaccine approach. However, type I IFNs have been reported to inhibit IL-12p70 production and mDC-induced T-cell activation. In this study, we investigate the effect of IFN-α on mDC maturation and function. We demonstrate that both recombinant IFN-α and activated pDCs strongly enhance mDC maturation and increase IL-12p70 production. Co-cultured mDCs and pDCs additionally have beneficial effect on NK and NKT-cell activation and also enhances IFN-γ production by allogeneic T cells. In contrast, the presence of type I IFNs reduces the proliferative T-cell response. The mere presence of a small fraction of activated pDCs is sufficient for these effects and the required ratio between the subsets is non-stringent. Taken together, these results support the usage of mDCs and pDCs combined into one immunotherapeutic vaccine with broad immunostimulatory features.


Assuntos
Células Dendríticas/imunologia , Interferon Tipo I/farmacologia , Interleucina-12/biossíntese , Células Mieloides/imunologia , Antígenos CD1/imunologia , Antígenos CD1/farmacologia , Técnicas de Cocultura , Células Dendríticas/citologia , Células Dendríticas/efeitos dos fármacos , Glicoproteínas/imunologia , Glicoproteínas/farmacologia , Humanos , Imunidade Inata , Interferon Tipo I/imunologia , Interferon alfa-2 , Interferon-alfa/imunologia , Interferon-alfa/farmacologia , Interferon gama/biossíntese , Interferon gama/imunologia , Interleucina-12/imunologia , Interleucina-12/farmacologia , Ativação Linfocitária , Células Mieloides/citologia , Células Mieloides/efeitos dos fármacos , Quinolinas/farmacologia , Proteínas Recombinantes/imunologia , Proteínas Recombinantes/farmacologia , Linfócitos T/citologia , Linfócitos T/efeitos dos fármacos , Linfócitos T/imunologia
2.
Eur J Immunol ; 45(10): 2798-809, 2015 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-26255864

RESUMO

Novel approaches of dendritic cell (DC) based cancer immunotherapy aim at harnessing the unique attributes of different DC subsets. Classical monocyte-derived DC vaccines are currently being replaced by either applying primary DCs or specifically targeting antigens and adjuvants to these subsets in vivo. Appropriate DC activation in both strategies is essential for optimal effect. For this purpose TLR agonists are favorable adjuvant choices, with TLR7 triggering being essential for inducing strong Th1 responses. However, mouse CD8α(+) DCs, considered to be the major cross-presenting subset, lack TLR7 expression. Interestingly, this DC subset can respond to TLR7 ligand upon concurrent TLR3 triggering. Nevertheless, the mechanism underlying this synergy remains obscure. We now show that TLR3 ligation results in the production of IFN-α, which rapidly induces the expression of TLR7, resulting in synergistic activation. Moreover, we demonstrate that this mechanism conversely holds for plasmacytoid DCs that respond to TLR3 ligation when TLR7 pathway is mobilized. We further demonstrate that this mechanism of sharpening DC senses is also conserved in human BDCA1(+) DCs and plasmacytoid DCs. These findings have important implications for future clinical trials as it suggests that combinations of TLR ligands should be applied irrespective of initial TLR expression profiles on natural DC subsets for optimal stimulation.


Assuntos
Células Dendríticas/imunologia , Interferon Tipo I/imunologia , Glicoproteínas de Membrana/agonistas , Receptor 3 Toll-Like/imunologia , Receptor 7 Toll-Like/agonistas , Animais , Células Dendríticas/citologia , Humanos , Glicoproteínas de Membrana/imunologia , Camundongos , Células Th1/citologia , Células Th1/imunologia , Receptor 7 Toll-Like/imunologia
3.
Mediators Inflamm ; 2016: 3605643, 2016.
Artigo em Inglês | MEDLINE | ID: mdl-27057096

RESUMO

Dendritic cells (DCs) are central players of immune responses; they become activated upon infection or inflammation and migrate to lymph nodes, where they can initiate an antigen-specific immune response by activating naive T cells. Two major types of naturally occurring DCs circulate in peripheral blood, namely, myeloid and plasmacytoid DCs (pDCs). Myeloid DCs (mDCs) can be subdivided based on the expression of either CD1c or CD141. These human DC subsets differ in surface marker expression, Toll-like receptor (TLR) repertoire, and transcriptional profile, suggesting functional differences between them. Here, we directly compared the capacity of human blood mDCs and pDCs to activate and polarize CD4(+) T cells. CD141(+) mDCs show an overall more mature phenotype over CD1c(+) mDC and pDCs; they produce less IL-10 and more IL-12 than CD1c(+) mDCs. Despite these differences, all subsets can induce the production of IFN-γ in naive CD4(+) T cells. CD1c(+) and CD141(+) mDCs especially induce a strong T helper 1 profile. Importantly, naive CD4(+) T cells are not polarized towards regulatory T cells by any subset. These findings further establish all three human blood DCs-despite their differences-as promising candidates for immunostimulatory effectors in cancer immunotherapy.


Assuntos
Células Dendríticas/metabolismo , Linfócitos T/metabolismo , Antígenos CD1/metabolismo , Antígenos de Superfície/metabolismo , Linfócitos T CD4-Positivos/metabolismo , Células Cultivadas , Células Dendríticas/efeitos dos fármacos , Glicoproteínas/metabolismo , Humanos , Interleucina-10/metabolismo , Interleucina-12/metabolismo , Células Mieloides/imunologia , Células Mieloides/metabolismo , Linfócitos T/efeitos dos fármacos , Trombomodulina , Receptores Toll-Like/metabolismo
4.
Cancer Immunol Immunother ; 64(11): 1461-73, 2015 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-26275446

RESUMO

Dendritic cells (DCs) are key in connecting innate and adaptive immunity. Their potential in inducing specific immune responses has made them interesting targets for immunotherapeutic approaches. Our research group was the first to exploit the naturally occurring myeloid DCs (mDCs) and plasmacytoid DCs (pDCs) in therapeutic vaccination trials against melanoma. To develop primary DC subsets as an optimal vaccine, the identification of a clinically applicable adjuvant activating both subsets is required. Although the expression of pathogen recognition receptors differs distinctly between the DC subsets, both pDCs and mDCs can respond to single-stranded RNA (ssRNA) via Toll-like receptors 7 and 8, respectively. Since ssRNA is easily degraded by RNases, we stabilized anionic RNA by complexing it with the positively charged protein protamine. This leads to the formation of protamine-RNA complexes with varying features depending on ionic content. We subsequently investigated the immunostimulatory effect of complexes that formed various salt concentrations on purified DC subsets. Both mDCs and pDCs upregulated maturation markers and produced pro-inflammatory cytokines in a dose-dependent way to the protamine-RNA complexes. This was dependent on endosomal acidification and correlated partly with the uptake of protamine-RNA complexes. Furthermore, both DC subsets induced T cell proliferation and IFN gamma secretion in a beneficial ratio to IL-10. These results indicate that protamine-RNA complexes can be used to stimulate human mDC and pDC ex vivo for use in immunotherapeutic settings.


Assuntos
Adjuvantes Imunológicos/farmacologia , Células Dendríticas/imunologia , Protaminas/farmacologia , RNA/farmacologia , Células Cultivadas , Citocinas/metabolismo , Células Dendríticas/classificação , Relação Dose-Resposta a Droga , Endossomos/fisiologia , Humanos , Interferon gama/biossíntese , Ativação Linfocitária , Estabilidade de RNA , Cloreto de Sódio/farmacologia , Receptores Toll-Like/fisiologia
5.
Crit Rev Immunol ; 34(6): 517-36, 2014.
Artigo em Inglês | MEDLINE | ID: mdl-25597314

RESUMO

Dendritic cells (DCs) are key players in the induction of immune responses. Adoptive transfer of autologous mature DCs loaded with tumor-associated antigens is a promising therapy for the treatment of immunogenic tumors. For a long time, its therapeutic activity was thought to depend solely on the induction of tumor-specific CD8+ and CD4+ T cell responses. More recently, DCs were shown to bidirectionally interact with innate and innate-like immune cells, including natural killer (NK), invariant natural killer T (iNKT), and γδ T cells. These effector cells can amplify responses induced by DCs via several mechanisms, including induction of DC maturation and conventional T cell priming. In addition, NK, iNKT, and γδ T cells possess cytolytic activity and can act directly on tumor cells. Therapeutic strategies targeting these innate and innate-like immune cells hence hold potential to improve current DC vaccination protocols.


Assuntos
Antígenos de Neoplasias/imunologia , Linfócitos T CD4-Positivos/imunologia , Células Dendríticas/imunologia , Células Matadoras Naturais/imunologia , Neoplasias/prevenção & controle , Linfócitos T Citotóxicos/imunologia , Transferência Adotiva , Animais , Antígenos de Neoplasias/genética , Linfócitos T CD4-Positivos/citologia , Vacinas Anticâncer/administração & dosagem , Vacinas Anticâncer/genética , Vacinas Anticâncer/imunologia , Comunicação Celular/imunologia , Células Dendríticas/citologia , Células Dendríticas/transplante , Regulação Neoplásica da Expressão Gênica , Humanos , Imunidade Inata , Células Matadoras Naturais/citologia , Camundongos , Neoplasias/imunologia , Neoplasias/patologia , Receptores de Antígenos de Linfócitos T gama-delta/genética , Receptores de Antígenos de Linfócitos T gama-delta/imunologia , Transdução de Sinais , Linfócitos T Citotóxicos/citologia , Vacinação
6.
J Immunol ; 190(7): 3346-53, 2013 Apr 01.
Artigo em Inglês | MEDLINE | ID: mdl-23427247

RESUMO

Cytokines and IFNs, such as TNF-α and IFN-α, upregulate costimulatory molecules in monocyte-derived dendritic cells (MDDCs), enabling effective Ag presentation to T cells. This activation of MDDCs is often accompanied by upregulation of apolipoprotein B mRNA-editing, enzyme-catalytic, polypeptide-like 3 (APOBEC3) (A3) family proteins that are able to restrict HIV-1 replication in MDDCs by inducing hypermutations in the viral genome. In this study, we show that TNF-α upregulates costimulatory molecules and are able to restrict HIV-1BaL replication in MDDCs without significant induction of A3G, A3A, or A3F. Conversely, low quantities of IFN-α failed to upregulate costimulatory molecules, did not induce IL-12p40 or migration, but significantly induced A3G, A3A, and A3F mRNA expression and restricted viral replication in MDDCs. We also showed that transmission of HIV-1 from MDDCs to autologous T cells was significantly reduced in the presence of IFN-α. Sequence analyses detected the induction of high frequency of G-to-A hypermutations in the env genes from HIV-1BaL-infected MDDCs treated with low quantities of IFN-α2b. These findings show that low quantities of IFN-α can induce functional A3 family proteins and restrict HIV-1 replication in MDDCs while keeping an immature nonmigratory phenotype, supporting further investigations of modalities that enhance retroviral restriction factors. In addition, the findings highlight the role of IFN-α as a double-edged sword in HIV-1 infection, and we show that IFN-α can be powerful in reducing HIV-1 infection both in MDDCs and T cells.


Assuntos
Linfócitos T CD4-Positivos/efeitos dos fármacos , Linfócitos T CD4-Positivos/virologia , Citidina Desaminase/metabolismo , Células Dendríticas/efeitos dos fármacos , Células Dendríticas/metabolismo , HIV-1/fisiologia , Interferon-alfa/farmacologia , Desaminase APOBEC-3G , Antígeno B7-1/metabolismo , Antígeno B7-2/metabolismo , Células da Medula Óssea/efeitos dos fármacos , Células da Medula Óssea/imunologia , Células da Medula Óssea/metabolismo , Linfócitos T CD4-Positivos/imunologia , Movimento Celular/imunologia , Células Cultivadas , Citidina Desaminase/genética , Citosina Desaminase/genética , Citosina Desaminase/metabolismo , Células Dendríticas/imunologia , Regulação da Expressão Gênica/efeitos dos fármacos , Humanos , Interferon alfa-2 , Mutação/efeitos dos fármacos , Polietilenoglicóis/farmacologia , Proteínas/genética , Proteínas/metabolismo , Proteínas Recombinantes/farmacologia , Fator de Necrose Tumoral alfa/farmacologia , Replicação Viral/efeitos dos fármacos , Produtos do Gene env do Vírus da Imunodeficiência Humana/genética
7.
Blood ; 120(4): 768-77, 2012 Jul 26.
Artigo em Inglês | MEDLINE | ID: mdl-22700721

RESUMO

TLR3 is a key receptor for recognition of double-stranded RNA and initiation of immune responses against viral infections. However, hyperactive responses can have adverse effects, such as virus-induced asthma. Strategies to prevent TLR3-mediated pathology are therefore desired. We investigated the effect of single-stranded DNA oligonucleotides (ssDNA-ODNs) on TLR3 activation. Human monocyte-derived dendritic cells up-regulate maturation markers and secrete proinflammatory cytokines on treatment with the synthetic TLR3 ligand polyinosine-polycytidylic acid (poly I:C). These events were inhibited in cultures with ssDNA-ODNs. Poly I:C activation of nonhematopoietic cells was also inhibited by ssDNA-ODNs. The uptake of poly I:C into cells was reduced in the presence of ssDNA-ODNs, preventing TLR3 engagement from occurring. To confirm this inhibition in vivo, we administered ssDNA-ODNs and poly I:C, alone or in combination, via the intranasal route in cynomolgus macaques. Proinflammatory cytokines were detected in nasal secretions in the poly I:C group, while the levels were reduced in the groups receiving ssDNA-ODNs or both substances. Our results demonstrate that TLR3-triggered immune activation can be modulated by ssDNA-ODNs and provide evidence of dampening proinflammatory cytokine release in the airways of cynomolgus macaques. These findings may open novel perspectives for clinical strategies to prevent or treat inflammatory conditions exacerbated by TLR3 signaling.


Assuntos
Proliferação de Células , DNA de Cadeia Simples/farmacologia , Células Dendríticas/imunologia , Monócitos/imunologia , Oligonucleotídeos/farmacologia , Receptor 3 Toll-Like/metabolismo , Animais , Western Blotting , Diferenciação Celular , Células Cultivadas , Citocinas/metabolismo , Células Dendríticas/efeitos dos fármacos , Células Dendríticas/metabolismo , Humanos , Macaca fascicularis , Monócitos/efeitos dos fármacos , Monócitos/metabolismo , Mucosa Nasal/citologia , Mucosa Nasal/efeitos dos fármacos , Mucosa Nasal/metabolismo , Poli I-C/farmacologia , RNA de Cadeia Dupla/farmacologia , RNA Mensageiro/genética , Reação em Cadeia da Polimerase em Tempo Real , Sistema Respiratório/efeitos dos fármacos , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Transdução de Sinais/efeitos dos fármacos , Células Estromais/citologia , Células Estromais/efeitos dos fármacos , Células Estromais/metabolismo , Receptor 3 Toll-Like/antagonistas & inibidores
8.
J Biol Chem ; 287(17): 13731-42, 2012 Apr 20.
Artigo em Inglês | MEDLINE | ID: mdl-22396536

RESUMO

Dendritic cells (DCs) are professional antigen-presenting cells playing a central role in connecting innate and adaptive immunity. Maturation signals are, however, required for DCs to undergo phenotypic and functional changes to acquire a fully competent antigen-presenting capacity. We previously reported that activated apoptotic peripheral lymphocytes (ActApo) provide activation/maturation signals to human monocyte-derived DCs. In this paper, we have characterized the signaling pathways and molecules involved in ActApo-mediated DC maturation. We found that both cellular and supernatant fractions from ActApo are required for DC maturation signaling. ActApoSup-induced CD80 and CD86 expression was significantly blocked in the presence of neutralizing antibodies against tumor necrosis factor-α (TNF-α). Cell-cell contact-dependent signaling involved ß2 integrins, dendritic cell-specific ICAM-3-grabbing nonintegrin (DC-SIGN), and TLR4 because ActApo-induced up-regulation of the maturation markers CD80 and CD86 was significantly inhibited in the presence of neutralizing antibodies against CD18, CD11a, CD11b, and DC-SIGN as well as TLR4. The role of TLR4 was further confirmed by silencing of TLR4 in DCs. In addition, the endogenous adjuvant effect exerted by activated apoptotic splenocytes (ActApoSp) was reduced after immunization with human serum albumin in TLR4(-/-) mice. We detected activation of multiple signaling pathways and transcription factors in DCs upon co-culture with ActApo, including p38, JNK, PI3K-Akt, Src family kinases, NFκB p65, and AP1 transcription factor family members c-Jun and c-Fos, demonstrating the complex interactions occurring between ActApo and DCs. These studies provide important mechanistic insight into the responses of DCs during encounter with cells undergoing immunogenic cell death.


Assuntos
Antígenos CD/metabolismo , Apoptose , Antígenos CD18/metabolismo , Moléculas de Adesão Celular/metabolismo , Células Dendríticas/citologia , Lectinas Tipo C/metabolismo , Monócitos/citologia , Receptores de Superfície Celular/metabolismo , Receptor 4 Toll-Like/metabolismo , Animais , Diferenciação Celular , Técnicas de Cocultura , Humanos , Leucócitos Mononucleares/citologia , Receptores de Lipopolissacarídeos/biossíntese , Camundongos , Camundongos Endogâmicos C57BL , Fenótipo , Transdução de Sinais , Fatores de Transcrição/metabolismo
9.
MAbs ; 15(1): 2223750, 2023.
Artigo em Inglês | MEDLINE | ID: mdl-37332119

RESUMO

CD40 agonism by systemic administration of CD40 monoclonal antibodies has been explored in clinical trials for immunotherapy of cancer, uncovering enormous potential, but also dosing challenges in terms of systemic toxicity. CD40-dependent activation of antigen presenting cells is dependent on crosslinking of the CD40 receptor. Here we exploited this requisite by coupling crosslinking to cancer-receptor density by dual-targeting of CD40 and platelet-derived growth factor receptor beta (PDGFRB), which is highly expressed in the stroma of various types of tumors. A novel PDGFRBxCD40 Fc-silenced bispecific AffiMab was developed to this end to test whether it is possible to activate CD40 in a PDGFRB-targeted manner. A PDGFRB-binding Affibody molecule was fused to each heavy chain of an Fc-silenced CD40 agonistic monoclonal antibody to obtain a bispecific "AffiMab". Binding of the AffiMab to both PDGFRB and CD40 was confirmed by surface plasmon resonance, bio-layer interferometry and flow cytometry, through analysis of cells expressing respective target. In a reporter assay, the AffiMab displayed increased CD40 potency in the presence of PDGFRB-conjugated beads, in a manner dependent on PDGFRB amount/bead. To test the concept in immunologically relevant systems with physiological levels of CD40 expression, the AffiMab was tested in human monocyte-derived dendritic cells (moDCs) and B cells. Expression of activation markers was increased in moDCs specifically in the presence of PDGFRB-conjugated beads upon AffiMab treatment, while the Fc-silenced CD40 mAb did not stimulate CD40 activation. As expected, the AffiMab did not activate moDCs in the presence of unconjugated beads. Finally, in a co-culture experiment, the AffiMab activated moDCs and B cells in the presence of PDGFRB-expressing cells, but not in co-cultures with PDGFRB-negative cells. Collectively, these results suggest the possibility to activate CD40 in a PDGFRB-targeted manner in vitro. This encourages further investigation and the development of such an approach for the treatment of solid cancers.


Assuntos
Neoplasias , Receptor beta de Fator de Crescimento Derivado de Plaquetas , Humanos , Antígenos CD40 , Anticorpos Monoclonais , Monócitos/metabolismo
10.
J Transl Med ; 10: 144, 2012 Jul 11.
Artigo em Inglês | MEDLINE | ID: mdl-22784600

RESUMO

Europrise is a Network of Excellence supported by the European Commission within the 6th Framework programme from 2007 to 2012. The Network has involved over 50 institutions from 13 European countries together with 3 industrial partners and 6 African countries. The Network encompasses an integrated program of research, training, dissemination and advocacy within the field of HIV vaccines and microbicides. A central and timely theme of the Network is the development of the unique concept of co-usage of vaccines and microbicides. Training of PhD students has been a major task, and some of these post-graduate students have here summarized novel ideas emanating from presentations at the last annual Europrise meeting in Prague. The latest data and ideas concerning HIV vaccine and microbicide studies are included in this review; these studies are so recent that the majority have yet to be published. Data were presented and discussed concerning novel immunisation strategies; microbicides and PrEP (alone and in combination with vaccines); mucosal transmission of HIV/SIV; mucosal vaccination; novel adjuvants; neutralizing antibodies; innate immune responses; HIV/SIV pathogenesis and disease progression; new methods and reagents. These - necessarily overlapping topics - are comprehensively summarised by the Europrise students in the context of other recent exciting data.


Assuntos
Vacinas contra a AIDS , Fármacos Anti-HIV/uso terapêutico , Desenho de Fármacos , Infecções por HIV/imunologia , Animais , Infecções por HIV/prevenção & controle , Humanos
11.
J Transl Med ; 9: 40, 2011 Apr 12.
Artigo em Inglês | MEDLINE | ID: mdl-21486446

RESUMO

Novel, exciting intervention strategies to prevent infection with HIV have been tested in the past year, and the field is rapidly evolving. EUROPRISE is a network of excellence sponsored by the European Commission and concerned with a wide range of activities including integrated developmental research on HIV vaccines and microbicides from discovery to early clinical trials. A central and timely theme of the network is the development of the unique concept of co-usage of vaccines and microbicides. This review, prepared by the PhD students of the network captures much of the research ongoing between the partners. The network is in its 5th year and involves over 50 institutions from 13 European countries together with 3 industrial partners; GSK, Novartis and Sanofi-Pasteur. EUROPRISE is involved in 31 separate world-wide trials of Vaccines and Microbicides including 6 in African countries (Tanzania, Mozambique, South Africa, Kenya, Malawi, Rwanda), and is directly supporting clinical trials including MABGEL, a gp140-hsp70 conjugate trial and HIVIS, vaccine trials in Europe and Africa.


Assuntos
Vacinas contra a AIDS/imunologia , Anti-Infecciosos/imunologia , Desenho de Fármacos , Animais , Formação de Anticorpos/imunologia , Ensaios Clínicos como Assunto , Humanos
12.
J Transl Med ; 8: 72, 2010 Jul 26.
Artigo em Inglês | MEDLINE | ID: mdl-20659333

RESUMO

EUROPRISE is a Network of Excellence sponsored from 2007 to 2011 by the European Commission within the 6th Framework Program. The Network encompasses a wide portfolio of activities ranging from an integrated research program in the field of HIV vaccines and microbicides to training, dissemination and advocacy. The research program covers the whole pipeline of vaccine and microbicide development from discovery to early clinical trials. The Network is composed of 58 partners representing more than 65 institutions from 13 European countries; it also includes three major pharmaceutical companies (GlaxoSmithKline, Novartis and Sanofi-Pasteur) involved in HIV microbicide and vaccine research. The Network displays a dedicated and informative web page: http://www.europrise.org. Finally, a distinguishing trait of EUROPRISE is its PhD School of students from across Europe, a unique example in the world of science aimed at spreading excellence through training. EUROPRISE held its second annual conference in Budapest in November, 2009. The conference had 143 participants and their presentations covered aspects of vaccine and microbicide research, development and discovery. Since training is a major task of the Network, the students of the EUROPRISE PhD program summarized certain presentations and their view of the conference in this paper.


Assuntos
Vacinas contra a AIDS/imunologia , Anti-Infecciosos/síntese química , Desenho de Fármacos , Imunidade Adaptativa/imunologia , Animais , Ensaios Clínicos como Assunto , Suscetibilidade a Doenças , Europa (Continente) , Infecções por HIV/imunologia , HIV-1/imunologia , Humanos , Imunidade nas Mucosas/imunologia , Camundongos , Testes de Neutralização
13.
Sci Rep ; 8(1): 15841, 2018 10 26.
Artigo em Inglês | MEDLINE | ID: mdl-30367171

RESUMO

Recognition of nucleic acids by endosomal Toll-like receptors (TLR) is essential to combat pathogens, but requires strict control to limit inflammatory responses. The mechanisms governing this tight regulation are unclear. We found that single-stranded oligonucleotides (ssON) inhibit endocytic pathways used by cargo destined for TLR3/4/7 signaling endosomes. Both ssDNA and ssRNA conferred the endocytic inhibition, it was concentration dependent, and required a certain ssON length. The ssON-mediated inhibition modulated signaling downstream of TLRs that localized within the affected endosomal pathway. We further show that injection of ssON dampens dsRNA-mediated inflammatory responses in the skin of non-human primates. These studies reveal a regulatory role for extracellular ssON in the endocytic uptake of TLR ligands and provide a mechanistic explanation of their immunomodulation. The identified ssON-mediated interference of endocytosis (SOMIE) is a regulatory process that temporarily dampens TLR3/4/7 signaling, thereby averting excessive immune responses.


Assuntos
Clatrina/metabolismo , Endocitose/efeitos dos fármacos , Oligonucleotídeos/farmacologia , Receptor 3 Toll-Like/metabolismo , Receptor 4 Toll-Like/metabolismo , Receptor 7 Toll-Like/metabolismo , Animais , Células Cultivadas , Quimiocinas/metabolismo , Citocinas/metabolismo , DNA de Cadeia Simples/farmacologia , Endossomos/metabolismo , Humanos , Leucócitos Mononucleares/citologia , Leucócitos Mononucleares/efeitos dos fármacos , Leucócitos Mononucleares/metabolismo , Macaca fascicularis , Poli I-C/farmacologia , Transdução de Sinais/efeitos dos fármacos , Pele/metabolismo , Pele/patologia , Receptor 3 Toll-Like/antagonistas & inibidores , Receptor 4 Toll-Like/antagonistas & inibidores , Receptor 7 Toll-Like/antagonistas & inibidores
14.
Oncotarget ; 8(12): 19879-19893, 2017 Mar 21.
Artigo em Inglês | MEDLINE | ID: mdl-28186996

RESUMO

Effective stimulation of immune cells is crucial for the success of cancer immunotherapies. Current approaches to evaluate the efficiency of stimuli are mainly defined by known flow cytometry-based cell activation or cell maturation markers. This method however does not give a complete overview of the achieved activation state and may leave important side effects unnoticed. Here, we used an unbiased RNA sequencing (RNA-seq)-based approach to compare the capacity of four clinical-grade dendritic cell (DC) activation stimuli used to prepare DC-vaccines composed of various types of DC subsets; the already clinically applied GM-CSF and Frühsommer meningoencephalitis (FSME) prophylactic vaccine and the novel clinical grade adjuvants protamine-RNA complexes (pRNA) and CpG-P. We found that GM-CSF and pRNA had similar effects on their target cells, whereas pRNA and CpG-P induced stronger type I interferon (IFN) expression than FSME. In general, the pathways most affected by all stimuli were related to immune activity and cell migration. GM-CSF stimulation, however, also induced a significant increase of genes related to nonsense-mediated decay, indicating a possible deleterious effect of this stimulus. Taken together, the two novel stimuli appear to be promising alternatives. Our study demonstrates how RNA-seq based investigation of changes in a large number of genes and gene groups can be exploited for fast and unbiased, global evaluation of clinical-grade stimuli, as opposed to the general limited evaluation of a pre-specified set of genes, by which one might miss important biological effects that are detrimental for vaccine efficacy.


Assuntos
Células Dendríticas/imunologia , Imunoterapia/métodos , Análise de Sequência de RNA/métodos , Vacinas/imunologia , Adjuvantes Imunológicos/farmacologia , Antígenos CD1/imunologia , Antígenos CD1/metabolismo , Células Cultivadas , Análise por Conglomerados , Células Dendríticas/efeitos dos fármacos , Células Dendríticas/metabolismo , Citometria de Fluxo , Perfilação da Expressão Gênica/métodos , Ontologia Genética , Glicoproteínas/imunologia , Glicoproteínas/metabolismo , Fator Estimulador de Colônias de Granulócitos e Macrófagos/imunologia , Fator Estimulador de Colônias de Granulócitos e Macrófagos/farmacologia , Humanos , Interferon Tipo I/biossíntese , Interferon Tipo I/genética , Interferon Tipo I/imunologia , Oligodesoxirribonucleotídeos/imunologia , Oligodesoxirribonucleotídeos/farmacologia , Protaminas/imunologia , Protaminas/farmacologia , RNA/imunologia , RNA/farmacologia , Transcriptoma/efeitos dos fármacos , Transcriptoma/imunologia , Vacinas/uso terapêutico , Vacinas Virais/imunologia , Vacinas Virais/farmacologia
15.
Oncoimmunology ; 5(10): e1227902, 2016.
Artigo em Inglês | MEDLINE | ID: mdl-27853652

RESUMO

Human blood dendritic cells (DCs) hold great potential for use in anticancer immunotherapies. CD1c+ myeloid DCs and plasmacytoid DCs (pDCs) have been successfully utilized in clinical vaccination trials against melanoma. We hypothesize that combining both DC subsets in a single vaccine can further improve vaccine efficacy. Here, we have determined the potential synergy between the two subsets in vitro on the level of maturation, cytokine expression, and effector cell induction. Toll-like receptor (TLR) stimulation of CD1c+ DCs induced cross-activation of immature pDCs and vice versa. When both subsets were stimulated together using TLR agonists, CD86 expression on pDCs was increased and higher levels of interferon (IFN)-α were produced by DC co-cultures. Although the two subsets did not display any synergistic effect on naive CD4+ and CD8+ T cell polarization, CD1c+ DCs and pDCs were able to complement each other's induction of other immune effector cells. The mere presence of pDCs in DC co-cultures promoted plasma cell differentiation from activated autologous B cells. Similarly, CD1c+ DCs, alone or in co-cultures, induced high levels of IFN-γ from allogeneic peripheral blood lymphocytes or activated autologous natural killer (NK) cells. Both CD1c+ DCs and pDCs could enhance NK cell cytotoxicity, and interestingly DC co-cultures further enhanced NK cell-mediated killing of an NK-resistant tumor cell line. These results indicate that co-application of human blood DC subsets could render DC-based anticancer vaccines more efficacious.

16.
Cancer Res ; 76(15): 4332-46, 2016 08 01.
Artigo em Inglês | MEDLINE | ID: mdl-27325645

RESUMO

The tumor microenvironment is characterized by regulatory T cells, type II macrophages, myeloid-derived suppressor cells, and other immunosuppressive cells that promote malignant progression. Here we report the identification of a novel BDCA1(+)CD14(+) population of immunosuppressive myeloid cells that are expanded in melanoma patients and are present in dendritic cell-based vaccines, where they suppress CD4(+) T cells in an antigen-specific manner. Mechanistic investigations showed that BDCA1(+)CD14(+) cells expressed high levels of the immune checkpoint molecule PD-L1 to hinder T-cell proliferation. While this BDCA1(+)CD14(+) cell population expressed markers of both BDCA1(+) dendritic cells and monocytes, analyses of function, transcriptome, and proteome established their unique nature as exploited by tumors for immune escape. We propose that targeting these cells may improve the efficacy of cancer immunotherapy. Cancer Res; 76(15); 4332-46. ©2016 AACR.


Assuntos
Vacinas Anticâncer/genética , Células Dendríticas/imunologia , Receptores de Lipopolissacarídeos/metabolismo , Melanoma/genética , Células Progenitoras Mieloides/metabolismo , Proliferação de Células , Humanos , Melanoma/patologia , Microambiente Tumoral
17.
Cell Rep ; 16(11): 2953-2966, 2016 09 13.
Artigo em Inglês | MEDLINE | ID: mdl-27626665

RESUMO

Dendritic cells (DCs) play a key role in orchestrating adaptive immune responses. In human blood, three distinct subsets exist: plasmacytoid DCs (pDCs) and BDCA3+ and CD1c+ myeloid DCs. In addition, a DC-like CD16+ monocyte has been reported. Although RNA-expression profiles have been previously compared, protein expression data may provide a different picture. Here, we exploited label-free quantitative mass spectrometry to compare and identify differences in primary human DC subset proteins. Moreover, we integrated these proteomic data with existing mRNA data to derive robust cell-specific expression signatures with more than 400 differentially expressed proteins between subsets, forming a solid basis for investigation of subset-specific functions. We illustrated this by extracting subset identification markers and by demonstrating that pDCs lack caspase-1 and only express low levels of other inflammasome-related proteins. In accordance, pDCs were incapable of interleukin (IL)-1ß secretion in response to ATP.


Assuntos
Biomarcadores/metabolismo , Células Dendríticas/metabolismo , Inflamassomos/metabolismo , Proteômica/métodos , Caspase 1/metabolismo , Perfilação da Expressão Gênica , Regulação da Expressão Gênica , Humanos , Mapas de Interação de Proteínas , Reprodutibilidade dos Testes , Transcriptoma/genética
18.
Front Immunol ; 5: 165, 2014.
Artigo em Inglês | MEDLINE | ID: mdl-24782868

RESUMO

Dendritic cell (DC)-based immunotherapy employs the patients' immune system to fight neoplastic lesions spread over the entire body. This makes it an important therapy option for patients suffering from metastatic melanoma, which is often resistant to chemotherapy. However, conventional cellular vaccination approaches, based on monocyte-derived DCs (moDCs), only achieved modest response rates despite continued optimization of various vaccination parameters. In addition, the generation of moDCs requires extensive ex vivo culturing conceivably hampering the immunogenicity of the vaccine. Recent studies, thus, focused on vaccines that make use of primary DCs. Though rare in the blood, these naturally circulating DCs can be readily isolated and activated thereby circumventing lengthy ex vivo culture periods. The first clinical trials not only showed increased survival rates but also the induction of diversified anti-cancer immune responses. Upcoming treatment paradigms aim to include several primary DC subsets in a single vaccine as pre-clinical studies identified synergistic effects between various antigen-presenting cells.

19.
Front Immunol ; 5: 191, 2014.
Artigo em Inglês | MEDLINE | ID: mdl-24834066

RESUMO

Prostate cancer (PCa) is the most common cancer in men and the second most common cause of cancer-related death in men. In recent years, novel therapeutic options for PCa have been developed and studied extensively in clinical trials. Sipuleucel-T is the first cell-based immunotherapeutic vaccine for treatment of cancer. This vaccine consists of autologous mononuclear cells stimulated and loaded with an immunostimulatory fusion protein containing the prostate tumor antigen prostate acid posphatase. The choice of antigen might be key for the efficiency of cell-based immunotherapy. Depending on the treatment strategy, target antigens should be immunogenic, abundantly expressed by tumor cells, and preferably functionally important for the tumor to prevent loss of antigen expression. Autoimmune responses have been reported against several antigens expressed in the prostate, indicating that PCa is a suitable target for immunotherapy. In this review, we will discuss PCa antigens that exhibit immunogenic features and/or have been targeted in immunotherapeutic settings with promising results, and we highlight the hurdles and opportunities for cancer immunotherapy.

20.
PLoS One ; 6(6): e21171, 2011.
Artigo em Inglês | MEDLINE | ID: mdl-21698207

RESUMO

Dendritic cells (DCs) are activated by signaling via pathogen-specific receptors or exposure to inflammatory mediators. Here we show that co-culturing DCs with apoptotic HIV-infected activated CD4(+) T cells (ApoInf) or apoptotic uninfected activated CD4(+) T cells (ApoAct) induced expression of co-stimulatory molecules and cytokine release. In addition, we measured a reduced HIV infection rate in DCs after co-culture with ApoAct. A prerequisite for reduced HIV infection in DCs was activation of CD4(+) T cells before apoptosis induction. DCs exposed to ApoAct or ApoInf secreted MIP-1α, MIP-1ß, MCP-1, and TNF-α; this effect was retained in the presence of exogenous HIV. The ApoAct-mediated induction of co-stimulatory CD86 molecules and reduction of HIV infection in DCs were partially abrogated after blocking TNF-α using monoclonal antibodies. APOBEC3G expression in DCs was increased in co-cultures of DCs and ApoAct but not by apoptotic resting CD4(+) T cells (ApoRest). Silencing of APOBEC3G in DC abrogated the HIV inhibitory effect mediated by ApoAct. Sequence analyses of an env region revealed significant induction of G-to-A hypermutations in the context of GG or GA dinucleotides in DNA isolated from DCs exposed to HIV and ApoAct. Thus, ApoAct-mediated DC maturation resulted in induction of APOBEC3G that was important for inhibition of HIV-infection in DCs. These findings underscore the complexity of differential DC responses evoked upon interaction with resting as compared with activated dying cells during HIV infection.


Assuntos
Apoptose , Linfócitos T CD4-Positivos/citologia , Citidina Desaminase/fisiologia , Células Dendríticas/virologia , Desaminase APOBEC-3G , Sequência de Bases , Diferenciação Celular , Quimiocinas/metabolismo , Técnicas de Cocultura , Meios de Cultivo Condicionados , Citocinas/metabolismo , Primers do DNA , Células Dendríticas/citologia , Humanos , Reação em Cadeia da Polimerase
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA