terça-feira, 15 de novembro de 2011

Células dendriticas e vacinas


Dendritic Cells In Vivo: A Key Target for a New Vaccine Science

Ralph M. Steinman1Go To Corresponding Author 
1 Laboratory of Cellular Physiology and Immunology, the Rockefeller University and Chris Browne Center for Immunology and Immune Diseases, New York, NY 10065, USA
Corresponding author

  • Summary

  • Dendritic cells (DCs) are the antigen presenting cells that initiate and regulate immunity. By studying these cells in vivo, we will be able to move beyond standard approaches and design vaccines that directly harness the elaborate properties of DCs to control immunity.
 

MAIN TEXT

“The challenge for us as immunologists is to understand how the various elements work and fit together, and then to develop innovative solutions that do better than nature…. Immunity ranks with the most complex of complex systems, along with neurobiology and climate change.” P.C. Doherty and S.J. Turner, Immunity 27: 363–365
The traditional vaccines that we know induce immunity against specific microbial antigens and prevent infectious diseases. These vaccines are a major success story and emanate in large part from the discoveries of Louis Pasteur. His first vaccine against chicken cholera was created in 1879, and his most famous vaccine against human rabies was created in 1885 (Dubos, 1988). Pasteur's research was based on the science of microbiology, i.e., his discovery that distinct microbes are the causes of disease and that an attenuated microbe can induce long-lived protection against infection by the nonattenuated form of that organism. These breakthroughs occurred before there was any clear understanding of vaccine immunity, which began with the discovery of antibodies by von Behring and Kitasato in the 1890s. After the great advances in immunology during the twentieth century, a new vaccine era has finally arrived on the basis of key immune principles.
Vaccines can be defined as formulations that induce specific, nontoxic, and long-lasting immune responses to prevent or treat disease. The new vaccines that immunological research can now develop will deliver the relevant antigens and adjuvants (substances that work with antigens to either enhance or silence immunity) to redirect the immune system for the individual's benefit (Pulendran and Ahmed, 2006) (Figure 1). The dendritic cell (DC) biology that is described in this issue of Immunity provides a foundation on which to create vaccines that not only induce protection against microbes but also deal with cancer (see Melief, 2008), autoimmunity, and allergy (see Belkaid and Oldenhove, 2008).
Display high quality version of this figure
View a PowerPoint of this figure
Figure 1
Some Key Challenges for Vaccines in Immunological Science and Medicine
Here, I will discuss four features of DCs that establish their central role in developing new vaccine strategies: location, antigen handling, maturation, and subsets. As Doherty and Turner write in the introductory quotation, vaccine biology compels us to pull these features of DCs together and “develop innovative solutions that do better than nature.”

Location of Dendritic Cells

In the past, emphasis has been placed on the capacity of DCs to pick up antigens in the periphery, including vaccines at an injection site, and then migrate from peripheral tissues to the T cell areas of lymphoid organs to initiate immunity. The underlying biology is elegant (see Alvarez et al., 2008, this issue), yet more information is needed on the types of DCs that pick up vaccine antigens at an injection site, such as skin or muscle, and initiate immunity. Now additional origins of DCs are apparent (Shortman and Naik, 2007) (see López-Bravo and Ardavín, 2008, this issue). In the steady state, most DCs in lymphoid organs actually arise from a blood precursor (Fogg et al., 2006,Liu et al., 2007). These precursors can proliferate in the lymphoid organ, a process driven by flt-3 ligand (Waskow et al., 2008). Another potentially major source of DCs is monocytes. Monocyte-derived DCs accumulate in lymphoid tissues during some infections, e.g., Leishmania major (Leon et al., 2007). Learning how to mobilize and access these other reservoirs of DCs in vivo could enhance the quality and efficacy of vaccine-induced immunity.
Effective mucosal immunity or resistance at body surfaces is a major hurdle for vaccine development, probably because mucosal surfaces are specialized to sustain nonreactivity to all the innocuous antigens within commensal microorganisms and the proteins in the air we breathe and the foods we eat. HIV-1 vaccines for example will probably need to induce mucosal immunity because this virus is most often transmitted via a mucosal route and quickly leads to a rapid loss of T cells in the intestine (Brenchley et al., 2004,Mehandru et al., 2004). DCs are uniquely located beneath the epithelium at several mucosal surfaces, such as the airway, intestine, and stomach. Intravital microscopy has helped reveal that these cells insinuate their dendritic processes between epithelial cells to enter the mucosal lumen (Chieppa et al., 2006,Niess et al., 2005). A critical goal is to determine whether vaccines can be designed to access these mucosal DCs to bring about better mucosal immunity. Depending upon the medical condition under investigation, desirable mucosal vaccines need to induce both antibodies and T cells that block infection at the site of pathogen entry, or alternatively activate suppressor T cells (“regulatory T cells”), which have the potential to block the inflammatory and allergic diseases at mucosal surfaces (see Belkaid, 2008). One gap in current knowledge relates to antigen and adjuvant delivery to DCs associated with organized mucosal lymphoid tissues such as Peyer's patches. Because the latter are covered by a distinctive epithelium rich in antigen-transporting M cells, receptors on these M cells, if ligated, could improve vaccine delivery to the DCs that lie underneath (Nochi et al., 2007).
A hallmark of DC location is their abundance in lymphoid tissues, particularly the T cell areas (Alvarez et al., 2008). Numerically, DCs represent a tiny fraction of total cells, but their processes constantly probe the environment, forming a vast and labyrinthine network through which lymphocytes must pass (Lindquist et al., 2004). This sets the stage for the selection of rare but specific clones of lymphocytes during the initial steps of vaccination. It is now feasible to target vaccine antigens directly to these numerous DCs in the T cell areas and modulate their function with adjuvants (Bonifaz et al., 2004,Boscardin et al., 2006,Trumpfheller et al., 2008,Trumpfheller et al., 2006).

Antigen Handling by Dendritic Cells

Because of the molecular identification of the DEC-205 (CD205) antigen uptake receptor on DCs (Jiang et al., 1995), it has become apparent that these cells express a plethora of such receptors, often lectins. These molecules deliver antigens to processing compartments, leading to the presentation of antigen fragments on MHC and CD1 molecules (see Villadangos and Young, 2008). Endocytic receptors in some cases also signal DC activation or deactivation (Robinson et al., 2006). Although DCs are able to capture antigens as solutes in endocytic vacuoles (“pinocytosis”), the identification of uptake receptors changes the study of DC biology in vivo and opens new possibilities for efficient vaccine delivery to DCs and vaccine design.
By identifying specific ligands for antigen uptake receptors on DCs, or by using monoclonal anti-receptor antibodies as surrogate ligands, one can efficiently target vaccine antigens to DCs or their subsets (see below) in vivo (Hawiger et al., 2001). Retroviral vectors gain improved immunogenicity if the envelope is engineered to target the DC-SIGN (CD209) receptor on DCs (Yang et al., 2008). The efficacy of HIV DNA vaccines can be improved by targeting to DEC-205 (Nchinda et al., 2008). Low doses of protein-based vaccines have also been targeted to DEC-205 along with poly IC as adjuvant. This leads to a large T helper 1 (Th1) cell type of T cell response, whereas targeting to the DCIR2 receptor on another subset of DCs allows both IFN-γ and IL-4 to be induced (Soares et al., 2007,Trumpfheller et al., 2008). Receptor targeting not only enhances antigen uptake and processing a hundred fold but also facilitates research on DCs and receptor function in vivo without the need to isolate the cells (Hawiger et al., 2001)(Bonifaz et al., 2004).
Another major hurdle for vaccine design is “crosspresentation.” Nonreplicating vaccines, e.g., the Salk inactivated viral vaccine, as well as protein vaccines, e.g., the diphtheria-pertussis-tetanus (DPT) vaccine, are capable of inducing antibody and CD4+ T cell immunity, sufficient in quantity and quality to provide protection against the polio virus and DPT toxins, respectively. However, in order for protein-based vaccines to elicit resistance to HIV and cancer, a nonreplicating form of an antigen needs to elicit MHC-class-I-restricted, CD8+ or cytotoxic T cells. Typically, MHC I presentation requires microbial replication and antigen synthesis in an infected cell; e.g., attenuated viral vectors can elicit CD8+ cytotoxic T cells, but they also elicit antivector immunity that can compromise the efficacy of booster doses of vaccine. Crosspresentation could overcome these hurdles because it provides a route for “exogenous” vaccine proteins to be processed and to gain access to MHC I. Mechanisms are under study (Kasturi and Pulendran, 2008); the latest concept is that there are special endosomal compartments where internalized antigens are crosspresented (Burgdorf et al., 2008,Di Pucchio et al., 2008). Certain receptors may be specialized to traverse the crosspresentation pathway, such as Fc receptors for antibody complexes (Dhodapkar et al., 2002a,Regnault et al., 1999), receptors for dying cells (den Haan et al., 2000,Dhodapkar et al., 2002b,Iyoda et al., 2002), and certain C-type lectin receptors such as DEC-205 (Bonifaz et al., 2004,Bozzacco et al., 2007). Targeting these receptors with vaccines may help to overcome the crosspresentation hurdle during vaccination. Nonetheless, I urge the field to move beyond the dominant use of ovalbumin in C57Bl/6 mice as the model antigen for these studies because it is orders of magnitude more sensitive than the antigens that need to be crosspresented to create effective vaccines. Ovalbumin may be distorting the standards for discovering safe, defined, protein-based vaccines.

Maturation of Dendritic Cells

The most intricate feature of DCs is their capacity to differentiate or mature along many different lines. This is driven by many different types of stimuli including (1) microbial ligands for pattern recognition receptors, (2) innate lymphocytes, (3) immune complexes acting on Fc receptors, and (4) additional environmental and endogenous stimuli termed “alarmins.” Maturing DCs, depending on the stimulus as well as environmental factors affecting lymphocytes, then determine the type of response, which can be either immunogenic, providing resistance, or tolerogenic, silencing an immune response. One sphere of immunity that is particularly sensitive to the type of stimulus encountered by DCs is CD4+ helper T cell differentiation. The specific pathway followed by CD4+ T cells, whether it involves Th1, Th2, Th17, Tf, Tr1, or Treg cell differentiation, is significantly governed by DCs.
A major challenge is to understand how vaccine adjuvants influence DC maturation in vivo, so that the resulting immunity will appropriately resist a particular pathogen or disease. Two important areas of science are pattern recognition receptors and DC subsets (next) (Agrawal et al., 2003,Shah et al., 2003). Synthetic double-stranded RNA, poly IC, is recognized by TLR3 and MDA-5 receptors, and it can polarize CD4+ T cells along a Th1 cell pathway when delivered with antigens to the CD205+ DC subset (Soares et al., 2007,Trumpfheller et al., 2008). TLR7-TLR8 and TLR9 ligands also can serve as adjuvants for responses by IFN-γ-producing T cells (Wille-Reece et al., 2006). In contrast, ligands for the dectin receptor, when applied to bone-marrow-derived DCs, lead to IL-2 and IL-10 production and seem to favor Th17 cell differentiation (LeibundGut-Landmann et al., 2007). Ligation of c-kit and TSLP receptors allows DCs to induce Th2 cell responses, a process that takes place in the presence of certain allergens such as the house dust-mite allergen (Krishnamoorthy et al., 2008,Soumelis et al., 2002). These observations set the stage to design vaccines that direct the antigen-presenting DCs and/or the responding T cells to develop selected types of immunity; e.g., Th1 T cells help resist many viruses and tumors, Th2 cells mediate parasitic infections, and Th17 cells respond to certain extracellular bacteria and fungi.
An exciting new area will be to design vaccines to specifically silence unwanted immune reactions by taking advantage of DC programs that lead to tolerance. DCs can induce tolerance by deleting or silencing T cells (Brimnes et al., 2003,Hawiger et al., 2004), but they can also induce differentiation of suppressive T cells, e.g., IL-10 producers (Levings et al., 2005,Macdonald et al., 2005), and foxp3+ Treg cells (Kretschmer et al., 2005,Luo et al., 2007). Environmental cytokines such as IL-10 and TGF-β, as well as vitamin A, can be critical. The CD8+ or CD205+ subset of DCs in mice produces more TGF-β (Wang et al., 2008; our unpublished data) and also metabolizes vitamin A to retinoic acid; TGF-β and retinoic acid are cofactors for Treg cell development (Coombes et al., 2007,Sun et al., 2007). A different pathway involves the E-cadherin that DCs use to bind other cells or other DCs; when the DCs detach, the E-cadherin signals upregulation of the lymph-node-homing receptor CCR7 and production of tolerizing amounts of IL-10 (Jiang et al., 2007). In yet another route (and there will be many others!), ligation of select ILT molecules on DCs makes them tolerogenic (Liang et al., 2008,Manavalan et al., 2003). For these reasons, we can start to think about designing vaccines that harness antigen-presenting DCs to regulate and suppress specific, undesirable immune responses in allergy, autoimmunity, and transplantation.

Subsets of Dendritic Cells

Certain markers divide DCs into distinct forms termed subsets (Villadangos and Young, 2008). Many of these markers are receptors involved in pattern recognition and antigen presentation, the key steps in innate and adaptive immunity. All subsets, however, can share characteristic features of the DC lineage such as an unusual, probing dendritic morphology, high amounts of MHC class II, and potent T cell-stimulating activity.
DC subsets are likely to be selected to recognize distinct pathogens or forms of antigen and then follow through with distinct innate and adaptive responses (Liu, 2005,Shortman and Naik, 2007). To illustrate, plasmacytoid DCs express Toll-like receptors (TLR7, TLR8, and TLR9) and respond to viral and self-nucleic acids with vigorous type I interferon production (Kadowaki et al., 2000) (Boonstra et al., 2003) (Boule et al., 2004) (Lande et al., 2007); the plasmacytoid subset also has special endosomal compartments for crosspresentation on MHC I (Di Pucchio et al., 2008). Another subset of CD205+ “myeloid” DCs seems specialized to take up and crosspresent antigens from dying cells (den Haan et al., 2000,Iyoda et al., 2002). Within the skin, there are DC subsets that are specialized to induce either cytotoxic T cells or antibody-forming B cells (Klechevsky et al., 2008, this issue). In all these instances, it is likely that the immunogenic DC subset, and conceivably a tolerogenic DC subset as well, must present the relevant vaccine antigen and directly respond to the vaccine adjuvant (Sporri and Reis e Sousa, 2005,Sporri and Reis e Sousa, 2003). DC subsets therefore greatly expand the number of targets that vaccines can exploit to control immunity.
Monocyte-derived DCs comprise another type of DC subset(s). The term is used in two contexts and their relationship is still not established. In vivo, monocyte-derived DCs were recently described in lymph nodes during infection of mice with the parasite L. major (Leon et al., 2007; see López-Bravo and Ardavín, 2008), but their functional properties relative to other DC subsets in lymphoid tissues remain undefined. In addition, for many years scientists have been stimulating monocytes in vitro to develop many features of DCs (Romani et al., 1994,Sallusto and Lanzavecchia, 1994). Human monocyte-derived DCs are notably plastic depending upon the type of cytokines in the culture, e.g., IL-4, IL-15, or type I interferon (Dubsky et al., 2007). It still needs to be determined whether a vaccine approach based on monocyte-derived DCs can influence the immune outcome.
DC subsets can vary from one another in all the other features described above: location, receptors, and maturation programs. Nonetheless, given the potential importance of DC-subset diversity in determining the outcome of immunization, particularly the need to generate CD8+ cytotoxic T cell immunity in cancer and infections like AIDS, it is unsettling that the information base for DC subsets, including pattern-recognition receptors, is scant for DCs in intact human lymphoid and mucosal tissues.

An Exciting but Challenging Time for Vaccine Science

It is widely acknowledged that fundamental discoveries are needed to develop new vaccines (Klausner et al., 2003). Vaccine science will identify and understand the antigens, adjuvants, and protective cells that provide specific, durable, and nontoxic therapies. In the case of AIDS vaccines, discovery-dedicated academic scientists are finally beginning to receive the support and coordination they require, particularly through the new initiatives of the Bill and Melinda Gates Foundation and the National Institute of Allergy and Infectious Diseases. Unfortunately, the same cannot be said for other disease targets, particularly cancer. Immunologic approaches to cancer have proven to be effective especially with monoclonal antibodies. However, cellular- and vaccine-based therapies remain underappreciated and poorly supported areas of research, either as stand-alone approaches or in combination with other therapies (see Melief, 2008). Cancer patients also illustrate most poignantly the difficulties with the currently prevailing therapies against diseases that interact with the immune system. These drugs can have toxic and “offtarget” side effects and typically require frequent use. Vaccines, in contrast, offer the ultimate “targeted” therapy because they have the capacity to reach many specific targets in tandem, such as the many alterations in cancer cells, and because they remain directed and durable in their effects.
Vaccine design is often considered an empirical activity. But let us not be derailed by the “empirical” or “descriptive” label. The fundamental challenges are scientific, in that we must discover disease-relevant antigens, adjuvants, and protective mechanisms. Immunology is ready to address these challenges to understand how the various elements work and fit together. This is because the immune system deals directly with the myriad of disease targets, not only hundreds of different infections but also many cancers as well as autoimmune, inflammatory, and allergic conditions.
Yet there are at least three ways in which vaccine science differs from the mainstream of modern immunology. The first is that teams of scientists often have to interact on a regular basis, either within laboratories, between laboratories, or among institutions and countries, to solve the challenges of vaccine biology. There are just too many unknowns when it comes to antigens, adjuvants, and protective immune responses (Figure 1). Nevertheless, on a more hopeful note as emphasized here, many medical conditions can now be addressed from the focused perspective of vaccination.
The second difference from what immunologists are accustomed to is that vaccine science involves humans, not only mice. I am not referring to large-scale clinical trials testing whether existing practices and concepts are effective. Rather, we need to work out principles that govern the regulation of the human immune system in vivo and integrate this research with other medical sciences such as microbiology, genetics, and cancer biology. Basic research in humans is much more demanding in terms of costs, multiple regulations, and time constraints. Along with our current successful research enterprise, we now need to expand our research to study directly in patients the distinct pathogens and medical conditions that threaten us.
The third difference is that vaccine science involves discoveries that differ from what most scientists are so good at, which is to dissect disease-relevant mechanisms. The other less followed research path, which is imperative for vaccine science, is to identify basic principles that direct immune responses and apply these to “develop innovative solutions that do better than nature.” It is important to dissect immune function in model systems. But it is equally challenging to discover how to direct immunity to create vaccines that guide the human immune system. Dendritic cells, as key orchestrators of these responses in vivo, should help in the quest to bring new vaccines into medicine.


ACKNOWLEDGMENTS

I thank C. Moberg, M. Nussenzweig, and R. Seder for help with the manuscript.

REFERENCES

Agrawal et al., 2003 Agrawal, S., Agrawal, A., Doughty, B., Gerwitz, A., Blenis, J., Van Dyke, T., and Pulendran, B. (2003). Different toll-like receptor agonists instruct dendritic cells to induce distinct Th responses via differential modulation of extracellular signal-regulated kinase-mitogen-activated protein kinase and c-FosJ. Immunol. 171, 4984–4989PubMed
Alvarez et al., 2008 Alvarez, D., Vollmann, E.H., and von Andrian, U.H. (2008). Mechanisms and consequences of dendritic cell migrationImmunity 29, 325–342this issueAbstract | Full Text | PDF (2022 kb) |CrossRef | PubMed
Belkaid and Oldenhove, 2008 Belkaid, Y., and Oldenhove, G. (2008). Tuning microenvironments: Induction of regulatory T cells by dendritic cellsImmunity 29, 362–371this issueAbstract | Full Text | PDF(879 kb) | CrossRef | PubMed
Bonifaz et al., 2004 Bonifaz, L.C., Bonnyay, D.P., Charalambous, A., Darguste, D.I., Fujii, S., Soares, H., Brimnes, M.K., Moltedo, B., Moran, T.M., and Steinman, R.M. (2004). In vivo targeting of antigens to maturing dendritic cells via the DEC-205 receptor improves T cell vaccinationJ. Exp. Med. 199, 815–824CrossRef | PubMed
Boonstra et al., 2003 Boonstra, A., Asselin-Paturel, C., Gilliet, M., Crain, C., Trinchieri, G., Liu, Y.-J., and O'Garra, A. (2003). Flexibility of mouse classical and plasmacytoid-derived dendritic cells in directing T helper type 1 and 2 cell development: Dependency on antigen dose and differential Toll-like receptor ligationJ. Exp. Med. 197, 101–109CrossRef | PubMed
Boscardin et al., 2006 Boscardin, S.B., Hafalla, J.C.R., Kamphorst, A.O., Malilamani, R.F., Zebroski, H.A., Rai, U., Morrot, A., Zavala, F., Steinman, R.M., Nussenzweig, R.S., et al. (2006). Antigen targeting to dendritic cells elicits long-lived T cell help for antibody responsesJ. Exp. Med. 203, 599–606CrossRef | PubMed
Boule et al., 2004 Boule, M.W., Broughton, C., Mackay, F., Akira, S., Marshak-Rothstein, A., and Rifkin, I.R. (2004). Toll-like receptor 9-dependent and -independent dendritic cell activation by chromatin-immunoglobulin G complexesJ. Exp. Med. 199, 1631–1640CrossRef | PubMed
Bozzacco et al., 2007 Bozzacco, L., Trumpfheller, C., Siegal, F.P., Mehandru, S., Markowitz, M., Carrington, M., Nussenzweig, M.C., Piperno, A.G., and Steinman, R.M. (2007). DEC-205 receptor on dendritic cells mediates presentation of HIV gag protein to CD8+ T cells in a spectrum of human MHC I haplotypesProc. Natl. Acad. Sci. USA 104, 1289–1294CrossRef | PubMed
Brenchley et al., 2004 Brenchley, J.M., Schacker, T.W., Ruff, L.E., Price, D.A., Taylor, J.H., Beilman, G.J., Nguyen, P.L., Khoruts, A., Larson, M., Haase, A.T., et al. (2004). CD4+ T cell depletion during all stages of HIV disease occurs predominantly in the gastrointestinal tractJ. Exp. Med. 200, 749–759CrossRef | PubMed
Brimnes et al., 2003 Brimnes, M.K., Bonifaz, L., Steinman, R.M., and Moran, T.M. (2003). Influenza virus-induced dendritic cell maturation is associated with the induction of strong T cell immunity to a coadministered, normally nonimmunogenic proteinJ. Exp. Med. 198, 133–144CrossRef | PubMed
Burgdorf et al., 2008 Burgdorf, S., Scholz, C., Kautz, A., Tampe, R., and Kurts, C. (2008). Spatial and mechanistic separation of cross-presentation and endogenous antigen presentationNat. Immunol. 9, 558–566CrossRef | PubMed
Chieppa et al., 2006 Chieppa, M., Rescigno, M., Huang, A.Y.C., and Germain, R.N. (2006). Dynamic imaging of dendritic cell extension into the small bowel lumen in response to epithelial cell TLR engagementJ. Exp. Med. 203, 2841–2852CrossRef | PubMed
Coombes et al., 2007 Coombes, J.L., Siddiqui, K.R., Arancibia-Carcamo, C.V., Hall, J., Sun, C.M., Belkaid, Y., and Powrie, F. (2007). A functionally specialized population of mucosal CD103+ DCs induces Foxp3+ regulatory T cells via a TGF-β- and retinoic acid-dependent mechanismJ. Exp. Med. 204, 1757–1764CrossRef | PubMed
den Haan et al., 2000 den Haan, J.M., Lehar, S.M., and Bevan, M.J. (2000). CD8+ but not CD8- dendritic cells cross-prime cytotoxic T cells in vivoJ. Exp. Med. 192, 1685–1696CrossRef | PubMed
Dhodapkar et al., 2002a Dhodapkar, K.M., Krasovsky, J., Williamson, B., and Dhodapkar, M.V. (2002). Anti-tumor monoclonal antibodies enhance cross-presentation of cellular antigens and the generation of myeloma-specific killer T cells by dendritic cellsJ. Exp. Med. 195, 125–133CrossRef | PubMed
Dhodapkar et al., 2002b Dhodapkar, M.V., Krasovsky, J., and Olson, K. (2002). T cells from the tumor microenvironment of patients with progressive myeloma can generate strong, tumor-specific cytolytic responses to autologous, tumor-loaded dendritic cellsProc. Natl. Acad. Sci. USA 99, 13009–13013CrossRef | PubMed
Di Pucchio et al., 2008 Di Pucchio, T., Chatterjee, B., Smed-Sorensen, A., Clayton, S., Palazzo, A., Montes, M., Xue, Y., Mellman, I., Banchereau, J., and Connolly, J.E. (2008). Direct proteasome-independent cross-presentation of viral antigen by plasmacytoid dendritic cells on major histocompatibility complex class INat. Immunol. 9, 551–557CrossRef | PubMed
Dubos, 1988 Dubos, R. (1988). Pasteur and Modern ScienceSecond Edition, (Madison, WI: Science Tech Publishers)PubMed
Dubsky et al., 2007 Dubsky, P., Saito, H., Leogier, M., Dantin, C., Connolly, J.E., Banchereau, J., and Palucka, A.K. (2007). IL-15-induced human DC efficiently prime melanoma-specific naive CD8+ T cells to differentiate into CTLEur. J. Immunol. 37, 1678–1690CrossRef | PubMed
Fogg et al., 2006 Fogg, D.K., Sibon, C., Miled, C., Jung, S., Aucouturier, P., Littman, D.R., Cumano, A., and Geissmann, F. (2006). A clonogenic bone marrow progenitor specific for macrophages and dendritic cellsScience 311, 83–87CrossRef | PubMed
Hawiger et al., 2001 Hawiger, D., Inaba, K., Dorsett, Y., Guo, M., Mahnke, K., Rivera, M., Ravetch, J.V., Steinman, R.M., and Nussenzweig, M.C. (2001). Dendritic cells induce peripheral T cell unresponsiveness under steady state conditions in vivoJ. Exp. Med. 194, 769–780CrossRef | PubMed
Hawiger et al., 2004 Hawiger, D., Masilamani, R.F., Bettelli, E., Kuchroo, V.K., and Nussenzweig, M.C. (2004). Immunological unresponsiveness characterized by increased expression of CD5 on peripheral T cells induced by dendritic cells in vivoImmunity 20, 695–705Abstract | Full Text | PDF (195 kb) | CrossRef | PubMed
Iyoda et al., 2002 Iyoda, T., Shimoyama, S., Liu, K., Omatsu, Y., Akiyama, Y., Maeda, Y., Takahara, K., Steinman, R.M., and Inaba, K. (2002). The CD8+ dendritic cell subset selectively endocytoses dying cells in culture and in vivoJ. Exp. Med. 195, 1289–1302CrossRef | PubMed
Jiang et al., 2007 Jiang, A., Bloom, O., Ono, S., Cui, W., Unternaehrer, J., Jiang, S., Whitney, J.A., Connolly, J., Banchereau, J., and Mellman, I. (2007). Disruption of E-cadherin-mediated adhesion induces a functionally distinct pathway of dendritic cell maturationImmunity 27, 610–624Abstract | Full Text | PDF (1963 kb) | CrossRef | PubMed
Jiang et al., 1995 Jiang, W., Swiggard, W.J., Heufler, C., Peng, M., Mirza, A., Steinman, R.M., and Nussenzweig, M.C. (1995). The receptor DEC-205 expressed by dendritic cells and thymic epithelial cells is involved in antigen processingNature 375, 151–155CrossRef | PubMed
Kadowaki et al., 2000 Kadowaki, N., Antoneko, S., Lau, J.Y.-N., and Liu, Y.-J. (2000). Natural interferon-α/β-producing cells link innate and adaptive immunityJ. Exp. Med. 192, 219–226CrossRef | PubMed
Kasturi and Pulendran, 2008 Kasturi, S.P., and Pulendran, B. (2008). Cross-presentation: Avoiding trafficking chaos?Nat. Immunol. 9, 461–463CrossRef | PubMed
Klausner et al., 2003 Klausner, R.D., Fauci, A.S., Corey, L., Nabel, G.J., Gayle, H., Berkley, S., Haynes, B.F., Baltimore, D., Collins, C., Douglas, R.G., et al. (2003). Medicine. The need for a global HIV vaccine enterpriseScience 300, 2036–2039CrossRef | PubMed
Klechevsky et al., 2008 Klechevsky, E., Morita, R., Liu, M., Cao, Y., Coquery, S., Thompson-Snipes, L., Briere, F., Chaussabel, D., Zurawski, G., Palucka, A.K., et al. (2008). Functional specializations of human epidermal Langerhans cells and CD14+ dermal dendritic cellsImmunity 29, 497–510this issueAbstract | Full Text | PDF (1710 kb) | CrossRef | PubMed
Kretschmer et al., 2005 Kretschmer, K., Apostolou, I., Hawiger, D., Khazaie, K., Nussenzweig, M.C., and von Boehmer, H. (2005). Inducing and expanding regulatory T cell populations by foreign antigenNat. Immunol. 6, 1219–1227CrossRef | PubMed
Krishnamoorthy et al., 2008 Krishnamoorthy, N., Oriss, T.B., Paglia, M., Fei, M., Yarlagadda, M., Vanhaesebroeck, B., Ray, A., and Ray, P. (2008). Activation of c-Kit in dendritic cells regulates T helper cell differentiation and allergic asthmaNat. Med. 14, 565–573CrossRef | PubMed
Lande et al., 2007 Lande, R., Gregorio, J., Facchinetti, V., Chatterjee, B., Wang, Y.H., Homey, B., Cao, W., Wang, Y.H., Su, B., Nestle, F.O., et al. (2007). Plasmacytoid dendritic cells sense self-DNA coupled with antimicrobial peptideNature 449, 564–569CrossRef | PubMed
LeibundGut-Landmann et al., 2007 LeibundGut-Landmann, S., Gross, O., Robinson, M.J., Osorio, F., Slack, E.C., Tsoni, S.V., Schweighoffer, E., Tybulewicz, V., Brown, G.D., Ruland, J., et al. (2007). Syk- and CARD9-dependent coupling of innate immunity to the induction of T helper cells that produce interleukin 17Nat. Immunol. 8, 630–638CrossRef | PubMed
Leon et al., 2007 Leon, B., Lopez-Bravo, M., and Ardavin, C. (2007). Monocyte-derived dendritic cells formed at the infection site control the induction of protective T helper 1 responses against Leishmania.Immunity 26, 519–531Abstract | Full Text | PDF (1853 kb) | CrossRef | PubMed
Levings et al., 2005 Levings, M.K., Gregori, S., Tresoldi, E., Cazzaniga, S., Bonini, C., and Roncarolo, M.G. (2005). Differentiation of Tr1 cells by immature dendritic cells requires IL-10 but not CD25+CD4+ Tr cellsBlood 105, 1162–1169CrossRef | PubMed
Liang et al., 2008 Liang, S., Ristich, V., Arase, H., Dausset, J., Carosella, E.D., and Horuzsko, A. (2008). Modulation of dendritic cell differentiation by HLA-G and ILT4 requires the IL-6–STAT3 signaling pathwayProc. Natl. Acad. Sci. USA 105, 8357–8362CrossRef | PubMed
Lindquist et al., 2004 Lindquist, R.L., Shakhar, G., Dudziak, D., Wardemann, H., Eisenreich, T., Dustin, M.L., and Nussenzweig, M.C. (2004). Visualizing dendritic cell networks in vivoNat. Immunol. 5, 1243–1250CrossRef | PubMed
Liu et al., 2007 Liu, K., Waskow, C., Liu, X., Yao, K., Hoh, J., and Nussenzweig, M. (2007). Origin of dendritic cells in peripheral lymphoid organs of miceNat. Immunol. 8, 578–583CrossRef | PubMed
Liu, 2005 Liu, Y.J. (2005). IPC: Professional type 1 interferon-producing cells and plasmacytoid dendritic cell precursorsAnnu. Rev. Immunol. 23, 275–306CrossRef | PubMed
López-Bravo and Ardavín, 2008 López-Bravo, M., and Ardavín, C. (2008). In vivo induction of immune responses to pathogens by conventional dendritic cellsImmunity 29, 343–351this issueAbstract | Full Text | PDF (1053 kb) | CrossRef | PubMed
Luo et al., 2007 Luo, X., Tarbell, K.V., Yang, H., Pothoven, K., Bailey, S.L., Ding, R., Steinman, R.M., and Suthanthiran, M. (2007). Dendritic cells with TGF-β1 differentiate naive CD4+CD25 T cells into islet-protective Foxp3+ regulatory T cellsProc. Natl. Acad. Sci. USA 104, 2821–2826CrossRef | PubMed
Macdonald et al., 2005 Macdonald, K.P., Rowe, V., Clouston, A.D., Welply, J.K., Kuns, R.D., Ferrara, J.L., Thomas, R., and Hill, G.R. (2005). Cytokine expanded myeloid precursors function as regulatory antigen-presenting cells and promote tolerance through IL-10-producing regulatory T cellsJ. Immunol. 174, 1841–1850PubMed
Manavalan et al., 2003 Manavalan, J.S., Rossi, P.C., Vlad, G., Piazza, F., Yarilina, A., Cortesini, R., Mancini, D., and Suciu-Foca, N. (2003). High expression of ILT3 and ILT4 is a general feature of tolerogenic dendritic cellsTranspl. Immunol. 11, 245–258CrossRef | PubMed
Mehandru et al., 2004 Mehandru, S., Poles, M.A., Tenner-Racz, K., Horowitz, A., Hurley, A., Hogan, C., Boden, D., Racz, P., and Markowitz, M. (2004). Primary HIV-1 infection is associated with preferential depletion of CD4+ T lymphocytes from effector sites in the gastrointestinal tractJ. Exp. Med. 200, 761–770CrossRef | PubMed
Melief, 2008 Melief, C.J.M. (2008). Cancer immunotherapy by dendritic cellsImmunity 29, 372–383this issueAbstract | Full Text | PDF (796 kb) | CrossRef | PubMed
Nchinda et al., 2008 Nchinda, G., Kuroiwa, J., Oks, M., Trumpfheller, C., Park, C.G., Huang, Y., Hannaman, D., Schlesinger, S.J., Mizenina, O., Nussenzweig, M.C., et al. (2008). The efficacy of DNA vaccination is enhanced by targeting the encoded protein to dendritic cellsJ. Clin. Invest. 118, 1427–1436CrossRef | PubMed
Niess et al., 2005 Niess, J.H., Brand, S., Gu, X., Landsman, L., Jung, S., McCormick, B.A., Vyas, J.M., Boes, M., Ploegh, H.L., Fox, J.G., et al. (2005). CX3CR1-mediated dendritic cell access to the intestinal lumen and bacterial clearanceScience 307, 254–258CrossRef | PubMed
Nochi et al., 2007 Nochi, T., Yuki, Y., Matsumura, A., Mejima, M., Terahara, K., Kim, D.Y., Fukuyama, S., Iwatsuki-Horimoto, K., Kawaoka, Y., Kohda, T., et al. (2007). A novel M cell-specific carbohydrate-targeted mucosal vaccine effectively induces antigen-specific immune responsesJ. Exp. Med. 204, 2789–2796CrossRef | PubMed
Pulendran and Ahmed, 2006 Pulendran, B., and Ahmed, R. (2006). Translating innate immunity into immunological memory: Implications for vaccine developmentCell 124, 849–863Abstract | Full Text | PDF(778 kb) | CrossRef | PubMed
Regnault et al., 1999 Regnault, A., Lankar, D., Lacabanne, V., Rodriguez, A., Thery, C., Rescigno, M., Saito, T., Verbeek, S., Bonnerot, C., Ricciardi-Castagnoli, P., et al. (1999). Fcγ receptor-mediated induction of dendritic cell maturation and major histocompatibility complex class I-restricted antigen presentation after immune complex internalizationJ. Exp. Med. 189, 371–380CrossRef | PubMed
Robinson et al., 2006 Robinson, M.J., Sancho, D., Slack, E.C., Leibundgut-Landmann, S., and Sousa, C.R. (2006). Myeloid C-type lectins in innate immunityNat. Immunol. 7, 1258–1265CrossRef | PubMed
Romani et al., 1994 Romani, N., Gruner, S., Brang, D., Kampgen, E., Lenz, A., Trockenbacher, B., Konwalinka, G., Fritsch, P.O., Steinman, R.M., and Schuler, G. (1994). Proliferating dendritic cell progenitors in human bloodJ. Exp. Med. 180, 83–93CrossRef | PubMed
Sallusto and Lanzavecchia, 1994 Sallusto, F., and Lanzavecchia, A. (1994). Efficient presentation of soluble antigen by cultured human dendritic cells is maintained by granulocyte/macrophage colony-stimulating factor plus interleukin 4 and downregulated by tumor necrosis factor αJ. Exp. Med. 179, 1109–1118CrossRef | PubMed
Shah et al., 2003 Shah, J.A., Darrah, P.A., Ambrozak, D.R., Turon, T.N., Mendez, S., Kirman, J., Wu, C.Y., Glaichenhaus, N., and Seder, R.A. (2003). Dendritic cells are responsible for the capacity of CpG oligodeoxynucleotides to act as an adjuvant for protective vaccine immunity against Leishmania major in miceJ. Exp. Med. 198, 281–291CrossRef | PubMed
Shortman and Naik, 2007 Shortman, K., and Naik, S.H. (2007). Steady-state and inflammatory dendritic-cell developmentNat. Rev. Immunol. 7, 19–30PubMed
Soares et al., 2007 Soares, H., Waechter, H., Glaichenhaus, N., Mougneau, E., Yagita, H., Mizenina, O., Dudziak, D., Nussenzweig, M.C., and Steinman, R.M. (2007). A subset of dendritic cells induces CD4+T cells to produce IFN-γ by an IL-12-independent but CD70-dependent mechanism in vivoJ. Exp. Med. 204, 1095–1106CrossRef | PubMed
Soumelis et al., 2002 Soumelis, V., Reche, P.A., Kanzler, H., Yuan, W., Edward, G., Homey, B., Gilliet, M., Ho, S., Antonenko, S., Lauerma, A., et al. (2002). Human epithelial cells trigger dendritic cell-mediated allergic inflammation by producing TSLPNat. Immunol. 3, 673–680PubMed
Sporri and Reis e Sousa, 2005 Sporri, R., and Reis e Sousa, C. (2005). Inflammatory mediators are insufficient for full dendritic cell activation and promote expansion of CD4+ T cell populations lacking helper functionNat. Immunol. 6, 163–170CrossRef | PubMed
Sporri and Reis e Sousa, 2003 Sporri, R., and Reis e Sousa, C. (2003). Newly activated T cells promote maturation of bystander dendritic cells but not IL-12 productionJ. Immunol. 171, 6406–6413PubMed
Sun et al., 2007 Sun, C.M., Hall, J.A., Blank, R.B., Bouladoux, N., Oukka, M., Mora, J.R., and Belkaid, Y. (2007). Small intestine lamina propria dendritic cells promote de novo generation of Foxp3 T reg cells via retinoic acidJ. Exp. Med. 204, 1775–1785CrossRef | PubMed
Trumpfheller et al., 2006 Trumpfheller, C., Finke, J.S., Lopez, C.B., Moran, T.M., Moltedo, B., Soares, H., Huang, Y., Schlesinger, S.J., Park, C.G., Nussenzweig, M.C., et al. (2006). Intensified and protective CD4+ T cell immunity in mice with anti-dendritic cell HIV gag fusion antibody vaccineJ. Exp. Med. 203, 607–617CrossRef | PubMed
Trumpfheller et al., 2008 Trumpfheller, C., Caskey, M., Nchinda, G., Longhi, M.P., Mizenina, O., Huang, Y., Schlesinger, S.J., Colonna, M., and Steinman, R.M. (2008). The microbial mimic polyIC induces durable and protective CD4+ T cell immunity together with a dendritic cell targeted vaccineProc. Natl. Acad. Sci. USA 105, 2574–2579CrossRef | PubMed
Villadangos and Young, 2008 Villadangos, J.A., and Young, L. (2008). Plasmacytoid dendritic cell antigen presentation in vivoImmunity 29, 352–361this issueAbstract | Full Text | PDF (521 kb) | CrossRef |PubMed
Wang et al., 2008 Wang, L., Pino-Lagos, K., de Vries, V.C., Guleria, I., Sayegh, M.H., and Noelle, R.J. (2008). Programmed death 1 ligand signaling regulates the generation of adaptive Foxp3+CD4+ regulatory T cellsProc. Natl. Acad. Sci. USA 105, 9331–9336CrossRef | PubMed
Waskow et al., 2008 Waskow, C., Liu, K., Darrasse-Jeze, G., Guermonprez, P., Ginhoux, F., Merad, M., Shengelia, T., Yao, K., and Nussenzweig, M. (2008). The receptor tyrosine kinase Flt3 is required for dendritic cell development in peripheral lymphoid tissuesNat. Immunol. 9, 676–683CrossRef | PubMed
Wille-Reece et al., 2006 Wille-Reece, U., Flynn, B.J., Lore, K., Koup, R.A., Miles, A.P., Saul, A., Kedl, R.M., Mattapallil, J.J., Weiss, W.R., Roederer, M., et al. (2006). Toll-like receptor agonists influence the magnitude and quality of memory T cell responses after prime-boost immunization in nonhuman primatesJ. Exp. Med. 203, 1249–1258CrossRef | PubMed
Yang et al., 2008 Yang, L., Yang, H., Rideout, K., Cho, T., Joo, K.I., Ziegler, L., Elliot, A., Walls, A., Yu, D., Baltimore, D., et al. (2008). Engineered lentivector targeting of dendritic cells for in vivo immunization.Nat. Biotechnol. 26, 326–334CrossRef | PubMed

Nenhum comentário:

Postar um comentário