Buscar en
Inmunología
Toda la web
Inicio Inmunología Dendritic cells: Nearly 40 years later…
Información de la revista
Vol. 31. Núm. 2.
Páginas 49-57 (Abril - Junio 2012)
Compartir
Compartir
Descargar PDF
Más opciones de artículo
Visitas
4342
Vol. 31. Núm. 2.
Páginas 49-57 (Abril - Junio 2012)
Review article
Acceso a texto completo
Dendritic cells: Nearly 40 years later…
Células dendríticas: casi 40 años después…
Visitas
4342
Daniel Benitez-Ribasa, Francesc E. Borràsb, Margarita del Valc, Juan José Lasarted, Concepción Marañóne, Enrique Martín-Gayof, Pablo Sarobed, Maria L. Toribiof, María Montoyag,h,
Autor para correspondencia
maria.montoya@cresa.uab.es

Corresponding author.
a CIBERehd, Department of Experimental Gastroenterology, Barcelona, Spain
b Institut Investigació Germans Trias i Pujol (IGTP), Badalona (Barcelona), Spain
c Unidad de Inmunología Viral, Centro de Biología Molecular Severo Ochoa (CSIC-UAM), Universidad Autónoma de Madrid, Campus Cantoblanco, Madrid, Spain
d Área de Hepatología y Terapia Génica, Centro de Investigación Médica Aplicada CIMA, Pamplona, Spain
e GENYO, Centro de Genómica e Investigación Oncológica: Pfizer-Universidad de Granada-Junta de Andalucía, Granada, Spain
f Centro de Biología Molecular “Severo Ochoa” CSIC-UAM, Campus de la Universidad Autónoma de Madrid, Madrid, Spain
g Centre de Recerca en Sanitat Animal (CReSA), UAB-IRTA, Campus de la Universitat Autònoma de Barcelona, Barcelona, Spain
h Institut de Recerca i Tecnologia Agroalimentarias (IRTA), Barcelona, Spain
Este artículo ha recibido
Información del artículo
Resumen
Texto completo
Bibliografía
Descargar PDF
Estadísticas
Figuras (1)
Abstract

The immune system is probably one of the most complex cellular organizations in the body. Its complexity is not superfluous, but rather it is required to fulfill the complicated purpose of the immune system, namely: the recognition of the diverse repertoire of microorganisms and pathogens; the detection of neoplastic lesions originating from a range of tissues; and, while executing these tasks, the maintenance of peripheral tolerance by suppressing detrimental responses against healthy tissues. Since they were discovered by R. Steinman et al. nearly 40 years ago, dendritic cells (DCs) have emerged to be critical players in conducting the immune response to fulfill these roles. Here, we provide a general view on some aspects of DC immunology, highlighting the crucial role that R. Steinman's research in the DC field has played during all those years. This review will also give an outline on DC research in the particular aspects that represent the focus of research groups in Spain (recently organized as the DC.esp working group within SEI). Firstly, some of the subtypes of DC will be described, particularly thymic DC and their role on tolerance; then the DC role in tolerance will be examined, followed by their implications in viral infections. Finally, antigen targeting DCs will be reviewed taking into account the crucial contributions made by R. Steinman et al. This chapter will end by reviewing some DCs based therapies in viral infections.

Keywords:
Dendritic cells (DCs)
Neoplastic lesions
Thymic DC
Resumen

El sistema inmune es probablemente una de las estructuras más complejas del cuerpo. Esta complejidad no es superflua, sino que es necesaria para realizar todas las complicadas tareas a las que se enfrenta, tales como el reconocimiento de un amplísimo espectro de microorganismos y patógenos, la detección de lesiones oncogénicas en un amplio rango de tejidos y, mientras ejecuta estas tareas, el mantenimiento de la tolerancia periférica mediante la supresión de respuestas perjudiciales dirigidas a tejidos sanos. Desde que fueron descubiertas por R. Steinman y colaboradores hace casi cuarenta años, las células dendríticas (CD) se han posicionado como elementos clave que dirigen el sistema inmune para acometer las tareas propias del mismo. En esta revisión, mostraremos una visión general sobre algunos aspectos de la inmunología de las CD así como subrayaremos el papel primordial que la investigación de R. Steinman ejerció en el campo de las CD durante todos estos años. Además, esta revisión da unas pinceladas sobre la investigación en CD que abordan la mayoría de los grupos de investigación en este campo en España (recientemente organizados en un grupo de trabajo llamado DC.esp, dentro de la SEI). En primer lugar, se describen la mayoría de subtipos de CD, y en concreto las CD tímicas y su papel en tolerancia, para después hablar de la tolerancia y las CD, seguido de los descubrimientos sobre las implicaciones de las CD en infecciones virales. Finalmente, se revisará el direccionamiento de antígenos a CD, teniendo en cuenta las importantes contribuciones de R. Steinman y colaboradores en el campo. Este capítulo finalizará con una revisión sobre las terapias basadas en CD en infecciones virales.

Palabras clave:
Células dendríticas (CD)
Lesiones oncogénicas
CD tímicas
Texto completo

The understanding of the role of dendritic cells (DCs) in immune responses has come a long way since Steinman and colleagues described these cells in 1972. Extensive research during nearly 40 years has provided a good understanding of the complexity of the DC system and its pivotal role in immunity. The following review will attempt to provide a general view on some aspects of DC immunology and to underline the crucial role of Steinman's research on the DC field during all those years. Several groups are working on DC in Spain, some because they were fascinated by DC research and some others because they stumble upon them and they were intrigued about these cells. This review will give an overview on DC research in the aspects that represent the focus of the majority of research groups working in DC immunology in Spain (recently organized as the DC.esp working group within SEI). Firstly, some of the subtypes of DC will be described, particularly thymic DC whose role on tolerance has been recently unraveled and which are in a separated section; then the DC role on tolerance will be examined, followed by a description of DC implications in viral infections. Finally, antigen targeting to DCs will be reviewed taking into account the crucial contributions made by R. Steinman and colleagues. This chapter will end by reviewing some DC-based therapies in viral infections. The role of DCs in cancer and its applications in DC-based cancer immunotherapy are topics not discussed in this review, as they are dealt with by Sureda et al. in other paper in this issue. All authors have contributed similarly in this review and the authorship was decided on alphabetical order.

DCs are integral to the initiation and regulation of immune responses. DCs are professional antigen presenting cells that play essential roles in mediating immunity in front of invading pathogens and also in tolerance to self and innocuous antigens. These apparently opposed functions are performed by DCs due to their plasticity and adaptability to respond to different physiological and pathological stimuli. DCs are located throughout the body and form a refined and complex network that allows them to communicate with different populations of lymphocytes, thereby forming an interface between the external environment and the adaptive immune system. In order to provide this protection, different subsets of DCs have evolved, and these DCs subsets are specialized to exist in distinct locations, where they acquire antigens and transport them to draining lymph nodes for T cell priming. This classification was initially based on their distinct patterns of cell-surface molecule expression. The four major categories of DCs are conventional DCs (cDCs), which predominate in the steady state; plasmacytoid DCs (pDCs); monocyte-derived DCs and Langerhans cells. More information on DCs can be found in Vàquez et al.1

Conventional DCs. Conventional DCs are specialized for antigen processing and presentation. They can be grouped into two main classes based on their localization in tissues and their migratory pathways as they circulate in the body. The first category of conventional DCs is generally referred to as the migratory DCs. Conventional DCs migrate from the blood to peripheral tissues where they sample the environment, and traffic from tissues via apherent lymphatic vessels to local lymph nodes for a highly efficient antigen presentation. Migratory DCs are not found in the spleen and are restricted to the lymph nodes.2 On the contrary, lymphoid tissue-resident DCs do not traffic from other tissues. In the absence of infection, they exist in an immature state (with high endocytic capacity and lower MHC class II expression compared to activated DCs), and their residency in lymphoid tissues makes them ideally placed to sense antigens or pathogens that are transported in the blood.3

Plasmacytoid DCs. pDCs correspond to natural interferon producing cells and are specialized in producing high levels of IFNα in response to virus infection.4 They are quiescent cells that are broadly distributed in the body and they are less competent than cDCs in antigen uptake. They are present in the thymus and secondary lymph nodes but rarely found in non-inflamed tissues and apherent lymphatics. However, under inflammatory conditions, pDCs migrate to and accumulate in tissues and in local lymph nodes, preferentially through high endothelial venules.5

Monocyte-derived DCs. They are induced in response to inflammation when circulating blood monocytes can be rapidly mobilized and can differentiate into cells that possess many prototypical features of DCs (Fig. 1).6 In response to growth factors such as granulocyte–macrophage colony-stimulating factor (GM-CSF) in vitro or to Toll-like receptor 4 (TLR4) ligands or bacteria in vivo, fully differentiated monocyte-derived DCs emerge. In addition, these cells acquire potent antigen-presenting capacity, including the ability to cross-present antigens.7 Thus, it is emerging that monocyte-derived DCs are a crucial reservoir of professional antigen-presenting cells (APCs) that are recruited into immune responses to certain microorganisms and potentially have an emergency back-up role in cases of acute inflammation.

Fig. 1.

Human cytokine-matured monocyte-derived DC floating in culture media exhibiting prominent dendrites (Line: 20μm).

(0,1MB).

Langerhans cells. Langerhans cells are resident in the skin and, like migratory DCs, migrate to the lymph nodes to present antigens. However, unlike conventional DCs, which arise from a bone marrow precursor cell, Langerhans cells are derived from a local LY6C+ myelomonocytic precursor cell population in the skin. This precursor population originates from macrophages that are present early in embryonic development and that undergo a proliferative burst in the epidermis in the first few days after birth.8

Thymic DCs

Dendritic cells represent a minor proportion of the hematopoietic compartment present in the postnatal thymus under steady-state conditions. Thymic DCs are able to present antigen in a tolerogenic fashion9 to those CD4+CD8+ double positive (DP) thymocytes that have survived positive selection. Depending on the overall TCR signal quantity and/or quality delivered during this interaction, potentially self-reactive thymocytes are either deleted by apoptosis (negative selection), or undergo commitment towards CD4+ CD25+ Foxp3+ natural regulatory T cells (nTregs), both mechanisms critically contributing to the establishment of central tolerance.10–12 While the role that thymic DCs play in negative selection has been firmly established, their contribution to the generation of nTreg cells remains controversial, especially because thymic epithelial cells (TECs) can function as inducers of nTregs.13–16 Adding complexity to this scheme, several groups have shown that thymus-resident DCs include both pDCs and cDCs subtypes.17–20 In the last years, nTregs have emerged as key players in the induction of immunological tolerance and, therefore, in the protection against fatal autoimmunity throughout life.21 As a consequence, a great interest has emerged in understanding how each intrathymic DC subtype could contribute to the generation of nTregs, especially in humans.

Seminal studies by Liu and coworkers provided direct evidence that human thymic cDCs display tolerogenic potential and generate nTregs, at least under allogeneic conditions in vitro.22 A latter work revealed that only one of the two subsets of cDCs found in mouse thymus supports nTregs development in vivo, and suggested that thymic pDCs display poor tolerogenic function.15 In contrast, we have recently shown that pDCs resident in the human thymus support the differentiation of autologous nTregs from DP thymocytes as efficiently as cDCs, through a mechanism that may involve the CD40-CD40L pathway.23 Moreover, pDCs and cDCs were shown to play a non-redundant functional role, as they selectively induced one of the two nTreg subsets, i.e. IL-10- or TGF-β-producing nTregs, previously identified in the human thymus.24 Therefore, current information points to thymic DCs as key regulators of central tolerance. Whether functional specialization of thymic pDCs and cDCs does exist owing to their distinct antigen presenting abilities25 is an interesting and still open question.

The important tolerogenic role of thymic DCs and their expected therapeutic potential has led to a great interest in defining the developmental origin of these cells. Pioneering work by Ardavin and collaborators showed that early thymic progenitors (ETPs) from the murine thymus displayed cDCs potential in vivo.26 Thereafter, our group extended this finding to human ETPs, which were shown to display non-T cell potentials, including cDCs, macrophage and NK cell potentials in vitro.27–29 As a whole, these data support the notion that thymic DCs are generated in situ from ETPs that display multipotent potential and commit to the T-cell lineage upon thymus entry. More recently, independent studies have shown that human ETPs can also generate pDCs both in vitro30 and in vivo,31 supporting their intrathymic origin. How non-T cells, and specifically DC, developmental pathways branch off from the main T-cell stream has been largely discussed. However, the recent identification in the human postnatal thymus of myeloid-restricted progenitors with pDC/cDC/macrophage potentials (unpublished results) supports a myeloid origin for thymic DCs.

In conclusion, current views support the notion that at least part of thymic pDCs and cDCs are derived in situ from myeloid-primed progenitors. How specific developmental fates are imposed within the thymic microenvironments at specific niches is still an open question that requires further study.

Tolerogenic DCs

The involvement of DCs in central and peripheral tolerance was soon considered as one of the functions played by DCs. It was earlier shown that, under steady-state conditions, iDCs may capture apoptotic bodies derived from natural cell turnover and, after migration to the draining LNs, DCs present self-antigens and silenciate autoreactive T cells.32 It is also well documented that the role of thymic DCs in tolerance induction against self-antigens and numerous studies have revealed mechanisms by which subsets of DCs induce or maintain self-tolerance. Tolerogenic DCs are immature, maturation-resistant or alternatively activated DCs that express low levels surface MHC molecules, have a low ratio of costimulatory to inhibitory signals, and an impaired ability to synthesize Th1- or Th17-cell-driving cytokines (such as IL-12p70, IL-23 or TNF-α) compared with fully mature cells. Furthermore, the expression of inhibitory receptors as well as the secretion of anti-inflammatory cytokines (IL-10 and TGF-β) contributes to tolerance induction. In general, the molecular mechanisms by which DCs exhibit a tolerogenic function include: antigen presentation with inappropriated costimulation (induction of anergy), presentation of very low levels of antigen in the absence of other stimuli, production of cytokines such as IL-10, transforming growth factor (TGF-α), tumor necrosis factor (TNF)-α, granulocyte colony-stimulating factor (G-CSF) or retinoic acid (RA), expression of inhibitory receptors such as ILT3, expression of some molecules that induce T cell death (deletion) such as indoleamine 2,3-dioxygenase (IDO), and also express membrane receptors that may instruct T-cell deletion, such as the interaction through the Fas-ligand, or the programmed death ligands (PDL)-1 and -2. Several evidences support that DCs, through the above mentioned mechanisms, contribute to peripheral tolerance by limiting the expansion of autoreactive T cells and by activating Tregs.33,34

Remarkably the role of DCs in maintaining tolerance is fully independent of their maturation state, since immature DCs, semi-mature DCs and also mature DCs have shown to expand antigen-specific Treg cells both in vitro and in vivo.35–38 Hence, authors recommend defining DCs as ‘immunogenic DCs’ or ‘tolerogenic DCs’ according to their functionality.

Some of the mechanisms indicated above have been exploited for the generation of tolerogenic DCs for therapeutic purposes. Of course, the clinical potential of TolDCs in autoimmunity and transplantation relates in part in the capacity of DCs to induce and expand regulatory T lymphocytes (Tregs) that suppress unwanted immunity. A number of murine studies suggest the possibility that potent antigen-specific regulatory T cells can be readily induced in vivo by targeting antigens to DC specific receptors under non-activating conditions. In certain conditions, mature DCs expand CD4+CD25+ Tregs with retained suppressive activity and are capable of inhibiting diabetes development in NOD mice. Tolerogenic properties of immature DCs have already been documented in healthy human volunteers, providing proof of principle that antigen-specific T cell tolerance can be achieved using this approach in humans.39In vitro, several agents, including dexamethasone, vitamin D3, retinoic acid, or IL-10, can be used to render DCs resistant to maturation when DCs are derived from monocytes (Fig. 1). However, so far, only one recent clinical study has taken advantage of their specific tolerogenic properties by utilizing CD40, CD80 and CD86 antisense transfected DCs to treat diabetic patients.40 The application of these tolerogenic DCs was safe and well tolerated. It is not difficult to speculate that tolerogenic DCs will be evaluated in other autoimmune or chronic inflammatory diseases like multiple sclerosis, arthritis or prevent organ transplant rejection. In vitro experiments have already shown promising results in some of these situations. Moreover, GMP grade TolDCs may be already obtained in GMP facilities, thus further supporting their potential use in a therapeutic scenario.

DCs in viral infections

A number of viral pathogens can infect cells of the immune system – particularly DCs and macrophages – reducing or even impairing the host's capacity for immune defense. DCs are highly responsive to stimuli such as microbial infection, inflammation, and tissue damage, and their maturation is further driven by interaction with T cells.41 The manner in which different viruses interact with DC function depends both on the virus and on the DC subset involved.42,43 In contrast, the outcome is less dependent on the capacity of the virus to replicate in the DC. A crucial issue is how antigens are presented to T cells by DCs.

Antigen presentation to CD8+ T lymphocytes by virus-infected dendritic cells

A mechanism for initiating CD8+ T-cell immunity to pathogens is the cross-presentation by MHC class I molecules of pathogen antigens after uptake of infected cells by DCs. The most efficient cross-presenting DCs also express the CD8 surface molecule.44 This mechanism is thought to exist to cope with some pathogens that cannot infect immature DCs, such as measles virus,45 or for which infection does not lead to full-blown viral antigen expression in DCs, such as vaccinia virus,46 as well as to deal with some pathogens that compromise the DCs antigen presentation function, such as respiratory syncytial virus,47,48 or that interfere with the MHC class I presentation pathway.49 An extreme interesting case is exemplified by cytomegalovirus, which is cleared by CD8 T-cell immunity in most organs; however, absence of cross-presentation together with interference with direct presentation by MHC class I molecules in the salivary gland result in local virus control exclusively by CD4+ T-cell-produced IFNγ.50

However, for many pathogens, notably for several viruses, direct presentation of viral proteins synthesized in these professional APC upon infection represents a straightforward pathway to prime naïve virus-specific CD8+ T lymphocytes. Interestingly, a need for maturation of influenza-virus-infected DCs results in a delayed direct viral antigen presentation as compared with other cell types,51 while in vivo direct presentation by vaccinia-virus-infected DCs already peaks at 6h post infection.52 The relative in vivo contribution of each mechanism differs for each infection, and there are examples of efficient CD8+ T-lymphocyte priming solely by cross-presentation, such as for herpes simplex virus 1 of for the Ankara strain of vaccinia virus,53,54 or solely by direct presentation, such as for vaccinia virus.55 Actually, it is important to recall that viral antigen presentation by DCs is only relevant if the elicited CD8+ T lymphocytes recognize pathogenically relevant infected tissue cells in the infected organism; for two herpesviruses, cytomegalovirus56 and Epstein-Barr virus,57 DCs have been shown to prime CD8+ T cells specific for viral antigens that are not presented in relevant tissues and that thus do not confer antiviral protection. It is thus very likely that both mechanisms contribute to CD8+ T-lymphocyte-mediated virus control, as the sets of epitopes generated by cross-presentation and direct presentation are not completely overlapping.49

Antigen cross-presentation by dendritic cells

Endocytosed or phagocytosed antigens are generally presented to T lymphocytes as peptide-class II MHC complexes, while endogenous cytosolic antigens are predominantly presented as peptide-class I MHC complexes. Dendritic cells have developed exclusive cross-presentation pathways58 allowing class I MHC-restricted presentation of exogenous antigens, taken up by endocytosis or phagocytosis.59 This pathway allows the induction of immunity or tolerance towards vaccines, pathogens that do not infect DCs, tissue-specific autoantigens, or tumoral antigens. The loading of class I MHC molecules with endocytosed or phagocytosed antigens implies their exit to the cytosol using specific channels to reach the classical class I pathway, or the fusion of the endo/phagolysosomal compartment with the endoplasmic reticulum, where mature class I molecules reside. Although several reports have provided evidences supporting both mechanisms, the routes by which exogenous antigens access newly formed class I MHC molecules remain unclear (revised in60).

The antigen to be cross-presented can be soluble (proteins, lysates) or particulated (pathogens, beads, cell-associated antigens).61 Regarding cell-associated antigen cross-presentation, studies have focused in dead cells (necrotic or apoptotic). In patients with different types of cancer, monocyte-derived DCs (Fig. 1) loaded with autologous or allogenic killed tumor cells have been extensively used for immunotherapy.62 However, only some types of death induce immunogenic stress signals.63 Quite recently it has been demonstrated that human monocyte-derived DCs (Fig. 1) can also acquire antigens from live cells for cross-presentation64 through nibbling, a mechanism similar to trogocytosis.65 It has also been shown that the in vivo cross-presentation of tumoral antigens from live melanoma cells by bone marrow-derived DCs is protective in a therapeutic model.66

In the mouse, CD8α+ DCs were thought to be unique in performing cross-presentation, and their human counterparts (BDCA-3+ cDCs) also present a high cross-presentation activity ex vivo.67,68 However, this cross-presentation activity is not exclusive of the former population and can also be detected in CD1c+ cDCs and pDCs ex vivo.69 Indeed it has also been shown that murine splenic pDCs cells efficiently cross-prime naive T cells in vivo after TLR activation.70 In summary, cross-presentation by DCs is crucial to reach immunoprotection and immunotolerance, but the mechanisms implicated in this exclusive pathway and the DC subsets involved in vivo remain elusive.

Targeting antigens to DCs

The identification of DCs as key regulators of T and B cell adaptive immunity prompted their introduction as adjuvants in vaccination strategies aimed to induce anti-tumor/virus antigen-specific T cell responses. In general, these strategies consist in the isolation of DCs from patients to be then loaded with tumor/viral antigens followed by their in vitro maturation using different stimuli previous to their transfer into the patient. A more direct strategy involves the “in vivo loading” or “targeting” of antigens to DCs through their surface receptors. Ralph Steinman et al. identified and exploited the use of CD205, a C-type lectin receptor (CLR) which is highly expressed in DCs, for the in vivo antigen targeting. CD205 recycles through late endosomal and lysosomal compartments and mediates antigen presentation very efficiently.71 They demonstrated that proteins could be targeted to DEC to greatly enhance antigen presentation to both CD4+ and CD8+ T cells in vivo. They assessed the potential of antigen targeting to dendritic cells to improve immunity, by the incorporation of ovalbumin protein (OVA) into a monoclonal antibody to the DEC-205 receptor. Targeting of anti-DEC-205:OVA to DCs in the steady state induced 4–7 cycles of T cell division, but the T cells were then deleted and the mice became specifically unresponsive to rechallenge with OVA. In contrast, simultaneous delivery of a DC maturation stimulus via CD40, together with anti-DEC-205:OVA, induced strong immunity with the activation of cytolytic CD8+ T cells producing large amounts of interleukin 2 and interferon gamma. They found that DEC-205 provides an efficient receptor-based mechanism for DCs to process proteins for MHC class I presentation in vivo, leading to tolerance in the steady state and immunity after DC maturation.72 Steinman et al. demonstrated that a DEC-targeted vaccine is able to induce a strong and protective CD4+ T cell response, which will improve vaccine efficacy as a stand-alone approach or with other modalities.73 To extend their finding to humans, they coupled the HIV p24 gag protein to anti-DEC antibodies and assessed cross-presentation for recognition by human CD8+ T cells. They found that DCs and DEC-205 can cross-present several different peptides from a single protein, supporting the testing of anti-DEC-205 fusion mAb as a protein-based vaccine for HIV.74,75 When administered together with synthetic double-stranded RNA, polyriboinosinic:polyribocytidylic acid (poly IC) or its analogue poly ICLC as adjuvant, HIV gag-p24 within anti-DEC-205 mAb was found to be highly immunogenic in mice, rhesus macaques, and in ongoing research, healthy human volunteers (reviewed in76). Steinman et al. made a great effort to develop a safe T-cell-based protein vaccine to target antigens to DCs and exploit their pivotal role in initiating adaptive immunity. Many laboratories have extended this approach using a variety of DC receptors. Thus, mannose receptors, DC-SIGN, LOX1, Dectin-1, Fc receptors, CD11c, CD209, CD40 (reviewed in Tacken et al.,77 CD36,78 CD91,79 or different ligands for toll-like receptors such as CpG,80 flagellin81 or the extra domain A from fibronectin82 among others are being exploited to target Ag to DCs and induce humoral and cellular immune responses. Thus, DC-targeted protein vaccines are a potential new vaccine platform, either alone or in combination with other adjuvants, to induce integrated immune responses against microbial or cancer antigens, with improved ease of manufacturing and clinical use.

DC-based therapies in viral infections

The properties of DCs as professional APC have led to their use as vaccines in different therapeutic settings. Although most of these applications have been related to the induction of anti-tumor immunity,83 chronic viral infections may also benefit of the advantages of DCs as immunogens. It has been described that among other mechanisms, some viruses may subvert anti-viral immunity by targeting functional properties of DCs,84–87 a feature common to some viruses causing chronic infections. This can be mediated either by a direct effect upon viral infection of DCs or by indirect mechanisms, such as interaction of viral products with DC receptors. Therefore, these individuals are often characterized by their poor T cell responses, mainly those of the Th1 subtype, which display the most potent anti-viral effects.88 The lack of a correct DC function during the natural course of viral infection has suggested that those strategies based on the administration of fully competent DCs may be able to activate T cells with proper anti-viral activity. Thus, initial preclinical studies carried out in animal models showed that DC vaccination could induce potent anti-viral immunity.89 Indeed, it has been shown that in some cases DC vaccination is the most appropriate strategy to break tolerance against viral antigens.90 Regarding human studies, although DCs obtained directly from blood of patients with chronic infections caused by viruses like HIV, HBV or HCV may present some phenotypic and/or functional impairment, protocols have been developed to differentiate functional monocyte-derived from these individuals. In vitro studies using these cells have shown that they can activate T cell responses against viral antigens, suggesting that this approach may be useful in these individuals.91,92 According to it, several clinical trials using DCs have been carried out. HIV infection is the field with the highest number of antiviral therapeutic vaccination clinical trials based on DCs (reviewed in93). They are characterized by their heterogeneity, both on trial design as well as on their DC manufacturing process. Although most of them use monocyte-derived DCs differentiated with GM-CSF and IL-4 (Fig. 1), they include several types of antigens (virus, recombinant proteins, peptides), as well as different loading strategies and maturation stimuli. These clinical trials have shown that DC vaccination is safe in these individuals, showing only minor local side-effects in some patients, in agreement with results obtained in cancer patients. Concerning immunogenicity, DC vaccination induced T cells of the Th1 profile able to recognize viral antigens. However, only in some cases a virological effect was reported, with some one-log decreases of viral load. Other fields using DC vaccination are chronic infections caused by HBV, HCV, HPV and EBV, although few examples exist in these cases.94–97 As for HIV, these protocols have shown to be safe and no significant toxicities were reported. In a similar manner, anti-viral T cell responses were induced or boosted after vaccination. Finally, poor clinical results have been reported, detectable only in a minority of vaccinated patients. The low number of clinical trials as well as their different design and experimental procedures precludes from obtaining solid conclusions regarding their antiviral effect. Nevertheless, current results are promising and further trials are warranted to ascertain the efficacy of DC vaccines as anti-viral therapies.

Future directions

It is now clear that an extraordinarily complex network of DC subsets contributes in different ways to the induction of immune responses and the maintenance of tolerance. A major challenge now will be to determine the overall contributions of different DC subsets to these processes, to understand the changes in the transcriptional network that are induced by inflammatory states to ensure the mobilization and activation of each DCs precursor and also to decipher DC interactions with other cells in the immune system.

DCs have long been considered ideal candidates for targeted vaccine approaches, but this promise remains largely unfulfilled. However, DC research has reached a point where clinical application has become a real possibility. So far, the rapid pace of immunology research and the fact that a vast number of human diseases have been proven to exhibit an immune or inflammatory component, suggests that the applications envisaged at the moment will likely be expanded in the future in a way that we cannot yet anticipate.

Conflict of interest

The authors declare no conflict of interest.

References
[1]
B. Vázquez, M. Sureda, J. Rebollo.
Células dendríticas I: aspectos básicos de su biología y funciones.
Inmunología, 31 (2012), pp. 21-30
[2]
K. Liu, M.C. Nussenzweig.
Origin and development of dendritic cells.
Immunol Rev, 234 (2010), pp. 45-54
[3]
R.J. Lundie, T.F. de Koning-Ward, G.M. Davey, C.Q. Nie, D.S. Hansen, L.S. Lau, et al.
Blood-stage Plasmodium infection induces CD8+ T lymphocytes to parasite-expressed antigens, largely regulated by CD8alpha+ dendritic cells.
Proc Natl Acad Sci U S A, 105 (2008), pp. 14509-14514
[4]
F.P. Siegal, N. Kadowaki, M. Shodell, P.A. Fitzgerald-Bocarsly, K. Shah, S. Ho, et al.
The nature of the principal type 1 interferon-producing cells in human blood.
Science, 284 (1999), pp. 1835-1837
[5]
H. Yoneyama, K. Matsuno, Y. Zhang, T. Nishiwaki, M. Kitabatake, S. Ueha, et al.
Evidence for recruitment of plasmacytoid dendritic cell precursors to inflamed lymph nodes through high endothelial venules.
Int Immunol, 16 (2004), pp. 915-928
[6]
S.H. Naik, D. Metcalf, A. van Nieuwenhuijze, I. Wicks, L. Wu, M. O’Keeffe, et al.
Intrasplenic steady-state dendritic cell precursors that are distinct from monocytes.
Nat Immunol, 7 (2006), pp. 663-671
[7]
C. Cheong, I. Matos, J.H. Choi, D.B. Dandamudi, E. Shrestha, M.P. Longhi, et al.
Microbial stimulation fully differentiates monocytes to DC-SIGN/CD209(+) dendritic cells for immune T cell areas.
[8]
L. Chorro, A. Sarde, M. Li, K.J. Woollard, P. Chambon, B. Malissen, et al.
Langerhans cell (LC) proliferation mediates neonatal development, homeostasis, and inflammation-associated expansion of the epidermal LC network.
J Exp Med, 206 (2009), pp. 3089-3100
[9]
R.M. Steinman, D. Hawiger, M.C. Nussenzweig.
Tolerogenic dendritic cells.
Annu Rev Immunol, 21 (2003), pp. 685-711
[10]
T. Brocker, M. Riedinger, K. Karjalainen.
Targeted expression of major histocompatibility complex (MHC) class II molecules demonstrates that dendritic cells can induce negative but not positive selection of thymocytes in vivo.
J Exp Med, 185 (1997), pp. 541-550
[11]
G.J. Nossal.
Negative selection of lymphocytes.
Cell, 76 (1994), pp. 229-239
[12]
J.P. van Meerwijk, S. Marguerat, R.K. Lees, R.N. Germain, B.J. Fowlkes, H.R. MacDonald.
Quantitative impact of thymic clonal deletion on the T cell repertoire.
J Exp Med, 185 (1997), pp. 377-383
[13]
S.J. Bensinger, A. Bandeira, M.S. Jordan, A.J. Caton, T.M. Laufer.
Major histocompatibility complex class II-positive cortical epithelium mediates the selection of CD4(+)25(+) immunoregulatory T cells.
J Exp Med, 194 (2001), pp. 427-438
[14]
C. Ohnmacht, A. Pullner, S.B. King, I. Drexler, S. Meier, T. Brocker, et al.
Constitutive ablation of dendritic cells breaks self-tolerance of CD4 T cells and results in spontaneous fatal autoimmunity.
J Exp Med, 206 (2009), pp. 549-559
[15]
A.I. Proietto, S. van Dommelen, P. Zhou, A. Rizzitelli, A. D’Amico, R.J. Steptoe, et al.
Dendritic cells in the thymus contribute to T-regulatory cell induction.
Proc Natl Acad Sci U S A, 105 (2008), pp. 19869-19874
[16]
G. Wirnsberger, F. Mair, L. Klein.
Regulatory T cell differentiation of thymocytes does not require a dedicated antigen-presenting cell but is under T cell-intrinsic developmental control.
Proc Natl Acad Sci U S A, 106 (2009), pp. 10278-10283
[17]
N. Bendriss-Vermare, C. Barthelemy, I. Durand, C. Bruand, C. Dezutter-Dambuyant, N. Moulian, et al.
Human thymus contains IFN-alpha-producing CD11c(−), myeloid CD11c(+), and mature interdigitating dendritic cells.
J Clin Invest, 107 (2001), pp. 835-844
[18]
T. Okada, Z.X. Lian, M. Naiki, A.A. Ansari, S. Ikehara, M.E. Gershwin.
Murine thymic plasmacytoid dendritic cells.
Eur J Immunol, 33 (2003), pp. 1012-1019
[19]
K. Shortman, Y.J. Liu.
Mouse and human dendritic cell subtypes.
Nat Rev Immunol, 2 (2002), pp. 151-161
[20]
S. Vandenabeele, H. Hochrein, N. Mavaddat, K. Winkel, K. Shortman.
Human thymus contains 2 distinct dendritic cell populations.
Blood, 97 (2001), pp. 1733-1741
[21]
S. Sakaguchi.
Naturally arising CD4+ regulatory t cells for immunologic self-tolerance and negative control of immune responses.
Annu Rev Immunol, 22 (2004), pp. 531-562
[22]
N. Watanabe, Y.H. Wang, H.K. Lee, T. Ito, W. Cao, Y.J. Liu.
Hassall's corpuscles instruct dendritic cells to induce CD4+CD25+ regulatory T cells in human thymus.
Nature, 436 (2005), pp. 1181-1185
[23]
E. Martin-Gayo, E. Sierra-Filardi, A.L. Corbi, M.L. Toribio.
Plasmacytoid dendritic cells resident in human thymus drive natural Treg cell development.
Blood, 115 (2010), pp. 5366-5375
[24]
T. Ito, S. Hanabuchi, Y.H. Wang, W.R. Park, K. Arima, L. Bover, et al.
Two functional subsets of FOXP3+ regulatory T cells in human thymus and periphery.
Immunity, 28 (2008), pp. 870-880
[25]
L.J. Young, N.S. Wilson, P. Schnorrer, A. Proietto, T. ten Broeke, Y. Matsuki, et al.
Differential MHC class II synthesis and ubiquitination confers distinct antigen-presenting properties on conventional and plasmacytoid dendritic cells.
Nat Immunol, 9 (2008), pp. 1244-1252
[26]
C. Ardavin, L. Wu, C.L. Li, K. Shortman.
Thymic dendritic cells and T cells develop simultaneously in the thymus from a common precursor population.
Nature, 362 (1993), pp. 761-763
[27]
V.G. de Yebenes, Y.R. Carrasco, A.R. Ramiro, M.L. Toribio.
Identification of a myeloid intrathymic pathway of dendritic cell development marked by expression of the granulocyte macrophage-colony-stimulating factor receptor.
Blood, 99 (2002), pp. 2948-2956
[28]
C. Marquez, C. Trigueros, E. Fernandez, M.L. Toribio.
The development of T and non-T cell lineages from CD34+ human thymic precursors can be traced by the differential expression of CD44.
J Exp Med, 181 (1995), pp. 475-483
[29]
C. Marquez, C. Trigueros, J.M. Franco, A.R. Ramiro, Y.R. Carrasco, M. Lopez-Botet, et al.
Identification of a common developmental pathway for thymic natural killer cells and dendritic cells.
Blood, 91 (1998), pp. 2760-2771
[30]
H. Fohrer, I.M. Audit, A. Sainz, C. Schmitt, C. Dezutter-Dambuyant, A.H. Dalloul.
Analysis of transcription factors in thymic and CD34+ progenitor-derived plasmacytoid and myeloid dendritic cells: evidence for distinct expression profiles.
Exp Hematol, 32 (2004), pp. 104-112
[31]
K. Weijer, C.H. Uittenbogaart, A. Voordouw, F. Couwenberg, J. Seppen, B. Blom, et al.
Intrathymic and extrathymic development of human plasmacytoid dendritic cell precursors in vivo.
Blood, 99 (2002), pp. 2752-2759
[32]
R.M. Steinman, S. Turley, I. Mellman, K. Inaba.
The induction of tolerance by dendritic cells that have captured apoptotic cells.
J Exp Med, 191 (2000), pp. 411-416
[33]
A.L. Mellor, D.H. Munn.
IDO expression by dendritic cells: tolerance and tryptophan catabolism.
Nat Rev Immunol, 4 (2004), pp. 762-774
[34]
D.H. Munn, A.L. Mellor, M. Rossi, J.W. Young.
Dendritic cells have the option to express IDO-mediated suppression or not.
[35]
M.L. Albert, M. Jegathesan, R.B. Darnell.
Dendritic cell maturation is required for the cross-tolerization of CD8+ T cells.
Nat Immunol, 2 (2001), pp. 1010-1017
[36]
M.B. Lutz, N.A. Kukutsch, M. Menges, S. Rossner, G. Schuler.
Culture of bone marrow cells in GM-CSF plus high doses of lipopolysaccharide generates exclusively immature dendritic cells which induce alloantigen-specific CD4 T cell anergy in vitro.
[37]
M. Menges, S. Rossner, C. Voigtlander, H. Schindler, N.A. Kukutsch, C. Bogdan, et al.
Repetitive injections of dendritic cells matured with tumor necrosis factor alpha induce antigen-specific protection of mice from autoimmunity.
J Exp Med, 195 (2002), pp. 15-21
[38]
S. Yamazaki, T. Iyoda, K. Tarbell, K. Olson, K. Velinzon, K. Inaba, et al.
Direct expansion of functional CD25+ CD4+ regulatory T cells by antigen-processing dendritic cells.
J Exp Med, 198 (2003), pp. 235-247
[39]
M.V. Dhodapkar, R.M. Steinman, J. Krasovsky, C. Munz, N. Bhardwaj.
Antigen-specific inhibition of effector T cell function in humans after injection of immature dendritic cells.
J Exp Med, 193 (2001), pp. 233-238
[40]
N. Giannoukakis, B. Phillips, D. Finegold, J. Harnaha, M. Trucco.
Phase I (safety) study of autologous tolerogenic dendritic cells in type 1 diabetic patients.
Diabetes Care, 34 (2011), pp. 2026-2032
[41]
C. Reis e Sousa.
Dendritic cells as sensors of infection.
Immunity, 14 (2001), pp. 495-498
[42]
S.S. Diebold, M. Montoya, H. Unger, L. Alexopoulou, P. Roy, L.E. Haswell, et al.
Viral infection switches non-plasmacytoid dendritic cells into high interferon producers.
Nature, 424 (2003), pp. 324-328
[43]
M. Montoya, M.J. Edwards, D.M. Reid, P. Borrow.
Rapid activation of spleen dendritic cell subsets following lymphocytic choriomeningitis virus infection of mice: analysis of the involvement of type 1 IFN.
J Immunol, 174 (2005), pp. 1851-1861
[44]
J.M. den Haan, S.M. Lehar, M.J. Bevan.
CD8+ but not CD8− dendritic cells cross-prime cytotoxic. T cells in vivo.
J Exp Med, 192 (2000), pp. 1685-1696
[45]
M. van der Vlist, L. de Witte, R.D. de Vries, M. Litjens, M.A. de Jong, D. Fluitsma, et al.
Human Langerhans cells capture measles virus through Langerin and present viral antigens to CD4 T cells but are incapable of cross-presentation.
Eur J Immunol, 41 (2011), pp. 2619-2631
[46]
E.F. Tewalt, J.M. Grant, E.L. Granger, D.C. Palmer, N.D. Heuss, D.S. Gregerson, et al.
Viral sequestration of antigen subverts cross presentation to CD8+ T cells.
PLoS Pathog, 5 (2009), pp. e1000457
[47]
P.M. De Graaff, E.C. de Jong, T.M. van Capel, M.E. van Dijk, P.J. Roholl, J. Boes, et al.
Respiratory syncytial virus infection of monocyte-derived dendritic cells decreases their capacity to activate CD4 T cells.
J Immunol, 175 (2005), pp. 5904-5911
[48]
P.A. Gonzalez, C.E. Prado, E.D. Leiva, L.J. Carreno, S.M. Bueno, C.A. Riedel, et al.
Respiratory syncytial virus impairs T cell activation by preventing synapse assembly with dendritic cells.
Proc Natl Acad Sci U S A, 105 (2008), pp. 14999-15004
[49]
S. Basta, W. Chen, J.R. Bennink, J.W. Yewdell.
Inhibitory effects of cytomegalovirus proteins US2 and US11 point to contributions from direct priming and cross-priming in induction of vaccinia virus-specific CD8+ T cells.
J Immunol, 168 (2002), pp. 5403-5408
[50]
S.M. Walton, S. Mandaric, N. Torti, A. Zimmermann, H. Hengel, A. Oxenius.
Absence of cross-presenting cells in the salivary gland and viral immune evasion confine cytomegalovirus immune control to effector CD4 T cells.
PLoS Pathog, 7 (2011), pp. e1002214
[51]
S. Herter, P. Osterloh, N. Hilf, G. Rechtsteiner, J. Hohfeld, H.G. Rammensee, et al.
Dendritic cell aggresome-like-induced structure formation and delayed antigen presentation coincide in influenza virus-infected dendritic cells.
J Immunol, 175 (2005), pp. 891-898
[52]
C.C. Norbury, D. Malide, J.S. Gibbs, J.R. Bennink, J.W. Yewdell.
Visualizing priming of virus-specific CD8+ T cells by infected dendritic cells in vivo.
Nat Immunol, 3 (2002), pp. 265-271
[53]
G. Gasteiger, W. Kastenmuller, R. Ljapoci, G. Sutter, I. Drexler.
Cross-priming of cytotoxic T cells dictates antigen requisites for modified vaccinia virus Ankara vector vaccines.
J Virol, 81 (2007), pp. 11925-11936
[54]
A.C. Jirmo, C.H. Nagel, C. Bohnen, B. Sodeik, G.M. Behrens.
Contribution of direct and cross-presentation to CTL immunity against herpes simplex virus 1.
J Immunol, 182 (2009), pp. 283-292
[55]
R.H. Xu, S. Remakus, X. Ma, F. Roscoe, L.J. Sigal.
Direct presentation is sufficient for an efficient anti-viral CD8+ T cell response.
PLoS Pathog, 6 (2010), pp. e1000768
[56]
R. Holtappels, J. Podlech, M.F. Pahl-Seibert, M. Julch, D. Thomas, C.O. Simon, et al.
Cytomegalovirus misleads its host by priming of CD8 T cells specific for an epitope not presented in infected tissues.
J Exp Med, 199 (2004), pp. 131-136
[57]
A.B. Hill, S.P. Lee, J.S. Haurum, N. Murray, Q.Y. Yao, M. Rowe, et al.
Class I major histocompatibility complex-restricted cytotoxic T lymphocytes specific for Epstein-Barr virus (EBV) nuclear antigens fail to lyse the EBV-transformed B lymphoblastoid cell lines against which they were raised.
J Exp Med, 181 (1995), pp. 2221-2228
[58]
S. Jung, D. Unutmaz, P. Wong, G. Sano, K. De los Santos, T. Sparwasser, et al.
In vivo depletion of CD11c+ dendritic cells abrogates priming of CD8+ T cells by exogenous cell-associated antigens.
Immunity, 17 (2002), pp. 211-220
[59]
E. Segura, J.A. Villadangos.
Antigen presentation by dendritic cells in vivo.
Curr Opin Immunol, 21 (2009), pp. 105-110
[60]
J.M. Vyas, A.G. Van der Veen, H.L. Ploegh.
The known unknowns of antigen processing and presentation.
Nat Rev Immunol, 8 (2008), pp. 607-618
[61]
T.W. Flinsenberg, E.B. Compeer, J.J. Boelens, M. Boes.
Antigen cross-presentation: extending recent laboratory findings to therapeutic intervention.
Clin Exp Immunol, 165 (2011), pp. 8-18
[62]
A.K. Palucka, H. Ueno, J. Connolly, F. Kerneis-Norvell, J.P. Blanck, D.A. Johnston, et al.
Dendritic cells loaded with killed allogeneic melanoma cells can induce objective clinical responses and MART-1 specific CD8+ T-cell immunity.
J Immunother, 29 (2006), pp. 545-557
[63]
D.R. Green, T. Ferguson, L. Zitvogel, G. Kroemer.
Immunogenic and tolerogenic cell death.
Nat Rev Immunol, 9 (2009), pp. 353-363
[64]
C. Maranon, J.F. Desoutter, G. Hoeffel, W. Cohen, D. Hanau, A. Hosmalin.
Dendritic cells cross-present HIV antigens from live as well as apoptotic infected CD4+ T lymphocytes.
Proc Natl Acad Sci U S A, 101 (2004), pp. 6092-6097
[65]
L.A. Harshyne, S.C. Watkins, A. Gambotto, S.M. Barratt-Boyes.
Dendritic cells acquire antigens from live cells for cross-presentation to CTL.
J Immunol, 166 (2001), pp. 3717-3723
[66]
D. Matheoud, C. Baey, L. Vimeux, A. Tempez, M. Valente, P. Louche, et al.
Dendritic cells crosspresent antigens from live B16 cells more efficiently than from apoptotic cells and protect from melanoma in a therapeutic model.
[67]
A. Bachem, S. Guttler, E. Hartung, F. Ebstein, M. Schaefer, A. Tannert, et al.
Superior antigen cross-presentation and XCR1 expression define human CD11c+CD141+ cells as homologues of mouse CD8+ dendritic cells.
J Exp Med, 207 (2010), pp. 1273-1281
[68]
K. Crozat, R. Guiton, V. Contreras, V. Feuillet, C.A. Dutertre, E. Ventre, et al.
The XC chemokine receptor 1 is a conserved selective marker of mammalian cells homologous to mouse CD8alpha+ dendritic cells.
J Exp Med, 207 (2010), pp. 1283-1292
[69]
G. Hoeffel, A.C. Ripoche, D. Matheoud, M. Nascimbeni, N. Escriou, P. Lebon, et al.
Antigen crosspresentation by human plasmacytoid dendritic cells.
Immunity, 27 (2007), pp. 481-492
[70]
J. Mouries, G. Moron, G. Schlecht, N. Escriou, G. Dadaglio, C. Leclerc.
Plasmacytoid dendritic cells efficiently cross-prime naive T cells in vivo after TLR activation.
Blood, 112 (2008), pp. 3713-3722
[71]
W. Jiang, W.J. Swiggard, C. Heufler, M. Peng, A. Mirza, R.M. Steinman, et al.
The receptor DEC-205 expressed by dendritic cells and thymic epithelial cells is involved in antigen processing.
Nature, 375 (1995), pp. 151-155
[72]
L. Bonifaz, D. Bonnyay, K. Mahnke, M. Rivera, M.C. Nussenzweig, R.M. Steinman.
Efficient targeting of protein antigen to the dendritic cell receptor DEC-205 in the steady state leads to antigen presentation on major histocompatibility complex class I products and peripheral CD8+ T cell tolerance.
J Exp Med, 196 (2002), pp. 1627-1638
[73]
C. Trumpfheller, J.S. Finke, C.B. Lopez, T.M. Moran, B. Moltedo, H. Soares, et al.
Intensified and protective CD4+ T cell immunity in mice with anti-dendritic cell HIV gag fusion antibody vaccine.
J Exp Med, 203 (2006), pp. 607-617
[74]
L. Bozzacco, C. Trumpfheller, F.P. Siegal, S. Mehandru, M. Markowitz, M. Carrington, et al.
DEC-205 receptor on dendritic cells mediates presentation of HIV gag protein to CD8+ T cells in a spectrum of human MHC I haplotypes.
Proc Natl Acad Sci U S A, 104 (2007), pp. 1289-1294
[75]
C. Trumpfheller, M. Caskey, G. Nchinda, M.P. Longhi, O. Mizenina, Y. Huang, et al.
The microbial mimic poly IC induces durable and protective CD4+ T cell immunity together with a dendritic cell targeted vaccine.
Proc Natl Acad Sci U S A, 105 (2008), pp. 2574-2579
[76]
C. Trumpfheller, M.P. Longhi, M. Caskey, J. Idoyaga, L. Bozzacco, T. Keler, et al.
Dendritic cell-targeted protein vaccines: a novel approach to induce T-cell immunity.
J Intern Med, 271 (2012), pp. 183-192
[77]
P.J. Tacken, I.J. de Vries, R. Torensma, C.G. Figdor.
Dendritic-cell immunotherapy: from ex vivo loading to in vivo targeting.
Nat Rev Immunol, 7 (2007), pp. 790-802
[78]
E. Tagliani, P. Guermonprez, J. Sepulveda, M. Lopez-Bravo, C. Ardavin, S. Amigorena, et al.
Selection of an antibody library identifies a pathway to induce immunity by targeting CD36 on steady-state CD8 alpha+ dendritic cells.
J Immunol, 180 (2008), pp. 3201-3209
[79]
J.P. Hart, M.D. Gunn, S.V. Pizzo.
A CD91-positive subset of CD11c+ blood dendritic cells: characterization of the APC that functions to enhance adaptive immune responses against CD91-targeted antigens.
J Immunol, 172 (2004), pp. 70-78
[80]
H. Wagner.
The immunogenicity of CpG-antigen conjugates.
Adv Drug Deliv Rev, 61 (2009), pp. 243-247
[81]
C. Cuadros, F.J. Lopez-Hernandez, A.L. Dominguez, M. McClelland, J. Lustgarten.
Flagellin fusion proteins as adjuvants or vaccines induce specific immune responses.
Infect Immun, 72 (2004), pp. 2810-2816
[82]
J.J. Lasarte, N. Casares, M. Gorraiz, S. Hervas-Stubbs, L. Arribillaga, C. Mansilla, et al.
The extra domain A from fibronectin targets antigens to TLR4-expressing cells and induces cytotoxic T cell responses in vivo.
J Immunol, 178 (2007), pp. 748-756
[83]
C.J. Melief.
Cancer immunotherapy by dendritic cells.
Immunity, 29 (2008), pp. 372-383
[84]
S. Beckebaum, V.R. Cicinnati, G. Dworacki, J. Muller-Berghaus, D. Stolz, J. Harnaha, et al.
Reduction in the circulating pDC1/pDC2 ratio and impaired function of ex vivo-generated DC1 in chronic hepatitis B infection.
Clin Immunol, 104 (2002), pp. 138-150
[85]
J. Engelmayer, M. Larsson, M. Subklewe, A. Chahroudi, W.I. Cox, R.M. Steinman, et al.
Vaccinia virus inhibits the maturation of human dendritic cells: a novel mechanism of immune evasion.
J Immunol, 163 (1999), pp. 6762-6768
[86]
B. Majumder, M.L. Janket, E.A. Schafer, K. Schaubert, X.L. Huang, J. Kan-Mitchell, et al.
Human immunodeficiency virus type 1 Vpr impairs dendritic cell maturation and T-cell activation: implications for viral immune escape.
J Virol, 79 (2005), pp. 7990-8003
[87]
P. Sarobe, J.J. Lasarte, A. Zabaleta, L. Arribillaga, A. Arina, I. Melero, et al.
Hepatitis C virus structural proteins impair dendritic cell maturation and inhibit in vivo induction of cellular immune responses.
J Virol, 77 (2003), pp. 10862-10871
[88]
B. Rehermann, M. Nascimbeni.
Immunology of hepatitis B virus and hepatitis C virus infection.
Nat Rev Immunol, 5 (2005), pp. 215-229
[89]
B. Ludewig, S. Ehl, U. Karrer, B. Odermatt, H. Hengartner, R.M. Zinkernagel.
Dendritic cells efficiently induce protective antiviral immunity.
J Virol, 72 (1998), pp. 3812-3818
[90]
Y. Shimizu, L.G. Guidotti, P. Fowler, F.V. Chisari.
Dendritic cell immunization breaks cytotoxic T lymphocyte tolerance in hepatitis B virus transgenic mice.
J Immunol, 161 (1998), pp. 4520-4529
[91]
I. Echeverria, A. Pereboev, L. Silva, A. Zabaleta, J.I. Riezu-Boj, M. Bes, et al.
T cell responses against hepatitis C virus by ex vivo targeting of adenoviral particles to dendritic cells.
Hepatology, 54 (2011), pp. 28-37
[92]
X.L. Huang, Z. Fan, L. Borowski, C.R. Rinaldo.
Multiple T-cell responses to human immunodeficiency virus type 1 are enhanced by dendritic cells.
Clin Vaccine Immunol, 16 (2009), pp. 1504-1516
[93]
F. Garcia, J.P. Routy.
Challenges in dendritic cells-based therapeutic vaccination in HIV-1 infection Workshop in dendritic cell-based vaccine clinical trials in HIV-1.
Vaccine, 29 (2011), pp. 6454-6463
[94]
S.M. Akbar, S. Furukawa, N. Horiike, M. Abe, Y. Hiasa, M. Onji.
Safety and immunogenicity of hepatitis B surface antigen-pulsed dendritic cells in patients with chronic hepatitis B.
J Viral Hepat, 18 (2011), pp. 408-414
[95]
E.J. Gowans, S. Roberts, K. Jones, I. Dinatale, P.A. Latour, B. Chua, et al.
A phase I clinical trial of dendritic cell immunotherapy in HCV-infected individuals.
J Hepatol, 53 (2010), pp. 599-607
[96]
C.L. Lin, W.F. Lo, T.H. Lee, Y. Ren, S.L. Hwang, Y.F. Cheng, et al.
Immunization with Epstein-Barr Virus (EBV) peptide-pulsed dendritic cells induces functional CD8+ T-cell immunity and may lead to tumor regression in patients with EBV-positive nasopharyngeal carcinoma.
Cancer Res, 62 (2002), pp. 6952-6958
[97]
A.D. Santin, S. Bellone, M. Palmieri, A. Zanolini, A. Ravaggi, E.R. Siegel, et al.
Human papillomavirus type 16 and 18 E7-pulsed dendritic cell vaccination of stage IB or IIA cervical cancer patients: a phase I escalating-dose trial.
J Virol, 82 (2008), pp. 1968-1979
Copyright © 2012. Sociedad Española de Inmunología
Opciones de artículo
Herramientas