Skip to main content
Immunology logoLink to Immunology
. 2004 Jul;112(3):345–351. doi: 10.1111/j.1365-2567.2004.01920.x

Cross-presentation: dendritic cells and macrophages bite off more than they can chew!

Sven Brode *, Paul A Macary
PMCID: PMC1782510  PMID: 15196201

Abstract

As immunologists, our knowledge of the molecular mechanisms which underlie the presentation of antigens derived from extracellular or ‘exogenous’ sources to CD8 cytotoxic lymphocytes (CTL) has been limited. This process, termed ‘cross-presentation’, has been linked to the elicitation of protective CTL responses against tumours and may be extremely important in generating immune responses against clinically relevant pathogens that do not infect tissues of haemopoietic origin. It is now known that cross-presentation of exogenous antigens on major histocompatibility complex (MHC) class I occurs through several distinct cellular pathways. In this review we outline and discuss some recent advances in our understanding of these pathways.

Keywords: antigen processing, cross-presentation, dendritic cell, exogenous antigen, MHC class I, phagosome

Introduction

The major histocompatibility complex (MHC) class-I restricted cytotoxic T-cell (CTL) response is the principal arm of the immune response that destroys intracellular pathogens such as viruses and some bacteria. CTL responses have also been implicated in the elimination of cells that have undergone malignant transformation. To initiate a protective CTL response, the antigens derived from pathogens and transformed cells must be processed and presented on professional antigen-presenting cells (APC) in the context of MHC class I molecules. Only professional APC such as dendritic cells (DC) and possibly some macrophages express a combination of co-receptors and MHC-class I molecules at levels high enough to stimulate naive CD8 T cells.1

Most peptides found on MHC class I molecules are derived from endogenous polypeptides synthesized on the cells' own ribosomes. Such endogenous antigens comprise peptides of host cell origin along with those from viruses and intracellular bacteria. The majority of antigenic peptides are generated by the proteolytic cleavage of malfolded proteins. This protein fraction has been collectively termed defective ribosomal products (DRiPs) and accounts for almost 30% of all synthesized proteins in cells.2

Upon DC activation, DRiPs accumulate as large cytosolic aggregates, called dendritic cell aggresome-like inducible structures (DALIS).3 Ubiquitinating enzymes present within these complexes generate substrates for the cytosolic multi-catalytic proteasome complex which cleaves large ubiquitinated polypeptides into smaller peptides.4 Peptides derived from proteasomal degradation are transported into the lumen of the endoplasmic reticulum (ER) by the transporter associated with antigen presentation (TAP).

In the lumen of the ER, newly imported peptides require further trimming by the ER-aminopeptidase I (ERAP).5 The resulting peptides, 8–9 amino acids long, are loaded onto nascent MHC class I chains by the MHC class I loading complex. This involves the ER chaperones calnexin, calreticulin and tapasin. This is part of a process that forms the fully folded, mature MHC class I molecules. MHC class I–peptide complexes then dissociate from the MHC class I loading complex and are transported through the secretory pathway to the plasma membrane for display to the immune system.

This presentation system evolved to exploit the dependence of replicating viruses on utilizing the host's protein synthesis machinery. However, it raises two important questions. How can we elicit CTL responses against tumour cells of a non-haematopoietic origin? How can we resolve viral infections where the virus does not infect professional APC? This paradox was solved when Bevan and colleagues characterized MHC class I-restricted CTL responses against peptides derived from ‘exogenous’ or extracellular protein sources.6,7 This postulated pathway for the transfer of exogenous antigen into the MHC class I antigen processing machinery was subsequently termed ‘cross-presentation’. With very few exceptions8,9 cross-presentation is restricted to DC and some macrophages but is not normally seen in other nucleated cells.10–14 In addition to ‘cross-priming’ naïve CD8 T cells, cross-presentation appears to be central to the maintenance of peripheral tolerance to self-antigens, that is ‘cross-tolerance’.15

Cross-presentation has been shown for viruses, allografts, tumours, particulate antigens and purified proteins, both, in vitro and in vivo.13,16,17

The role of dc in cross-presentation

DC are the most potent professional APC capable of priming naïve T cells in vivo.1 While resident in peripheral tissues DC exhibit high levels of receptor-mediated endocytosis, macropinocytosis and low levels of phagocytic activity and thus are efficient cells for capturing invading pathogens.1820 After capturing invading pathogens in the periphery, DC migrate to the local draining lymph nodes where they present antigens derived from the pathogens to circulating lymphocytes. Effective stimulation of naïve T cells occurs in the draining lymph nodes and depends on two main criteria: the efficient uptake and processing of antigens and a regulated maturation process in response to contact with inflammatory mediators. The exposure of DC to inflammatory stimuli induces their differentiation into professional APC. These unique aspects of DC function ensure that they are the ideal cell type for mediating cross-presentation in vitro and in vivo.12,13,2130 Deletion of DC abrogates cross-presentation of self-antigen.31 Both TAP-dependent and independent routes for the presentation of exogenous antigen have been identified in DC.

Routes of entry for exogenous antigens

There are four major cellular processes used by DC and macrophages to internalize exogenous antigens (Fig. 1). These are endocytosis, pinocytosis, phagocytosis and marcopinocytosis. The form of the antigen, its solubility, and whether it is part of an immune complex or still associated with a pathogen all determine the route of entry.

Figure 1.

Figure 1

Common pathways for the internalization of exogenous antigens by professional antigen presenting cells (pAPC). DC and macrophages acquire exogenous antigens through four major pathways. The nature of the antigen determines which internalization route is used. (a) Large particulate antigens (such as opsonized/complement fixed bacteria, apoptotic cells and biologically inert particles) are internalized by phagocytosis. (b) Small particulate antigens enter the cell by receptor-mediated endocytosis. (c) Pinocytosis describes the uptake of soluble antigens as part of the extracellular fluid present in the vicinity of the budding endosome. Uptake of endocytic vesicles can be both clathrin dependent and independent. (d) Large fluid volumes are internalized by marcopinocytosis. Antigen from endocytic vesicles is either translocated into the cytosol for presentation via the classical pathway, or loaded on MHC class I molecules within the endocytic compartment.

Endocytosis is the term used to describe the formation of vesicles of between ∼150–200 nm that form at sites of membrane invaginations, termed coated pits. Antigens induce the formation of coated pits by clustering specific cell surface receptors.32 In pinocytosis, soluble antigen is taken up at the same time as extracellular fluid present in the vicinity of the budding endosome. Uptake of these vesicles can be both clathrin dependent and independent.

Phagocytosis is the clathrin-independent process by which cells internalize large particulate material such as apoptotic cells, cellular debris, or bacteria, which are usually destined to be degraded by lysosomal enzymes. The phagosome is a membrane-bound organelle formed when a phagocytic cell engulfs particulate material.33 The phagocytic process can be divided into three stages: attachment of the particle to the cell surface, mediated by surface receptors; engulfment, characterized by the flow redistribution of the plasma membrane to surround the particle; and formation of the phagosome (phagolysosome). Macropinocytosis is a process whereby large vacuoles, termed macropinosomes, form at the plasma membrane; these nonspecifically trap large volumes of the extracellular media. Macropinosomes are usually 200–500 nm in diameter and are thought to form at sites of membrane ruffling.

Receptors involved in the recognition and uptake of exogenous antigens

Phagocytosis and receptor-mediated endocytosis are mediated by an increasing number of characterized cellular receptors; many of which have been implicated in cross-presentation. Targeting antigens to Fc receptors for immunoglobulin G (FcγR)1 or administered in the form of immune complexes,34 opsonized liposomes,35 or dead cells36 strongly augments cross-presentation by DC and macrophages. Mice lacking these receptors fail to cross-present efficiently.34

The uptake of cell-associated antigens from apoptotic and necrotic bodies is largely accomplished by complement receptors such as the calreticulin/CD91 receptor complex, αv integrins,24,37 class A-scavenger receptors and CD36.24,29,38 Bacterial antigens are recognized by a large panel of surface receptors, including the mannose receptor,39,40 dectin-141 and scavenger receptors.42

Bacterial and cellular antigens are also efficiently cross-presented when transferred as peptides associated to heat-shock proteins (hsp). Cross-priming activities have been reported for cytosolic hsp (hsp 70, hsc 70, hsp 90) and ER chaperones grp94/gp96,43–47 as well as their bacterial homologues.44,48,49 The uptake of hsp–peptide complexes appears to be mediated by specific cellular receptors such as the α2-macroglobulin receptor CD91,50,51 and the scavenger receptors LOX-152 and SR-A.53

The cell biology of cross-presentation

Nearly all cells can bind and represent peptides by β2-microglobulin-stabilized MHC class I dimers.54,55 However, this form of antigen presentation is restricted to a very limited number of antigens that are derived either by extracellular processing in the surrounding plasma, or by regurgitation of endosomally processed polypeptides. Efficient loading of MHC class I molecules has been considered to be restricted to the ER because cells lacking TAP or tapasin are severely impaired in their ability to present MHC class I-associated antigens.56,57 How can exogenous antigens access the MHC class I pathway? So far evidence has supported two principal pathways. Rodriguez and colleagues showed that exogenous antigen internalized by phagocytosis can escape lysosomal degradation by translocation into the cytosol in a murine DC cell line.22 A similar observation was made with antigens from internalized cytomegalovirus-infected fibroblasts in human DC58 and vaccinia virus-infected fibroblasts in murine macrophages and DC.59

Once in the cytosol, antigenic peptides derived from the translocated material by proteasomal degradation access the ER lumen via TAP. Three observations support this model: Cross-presentation is abrogated by the inhibition of proteasomal degradation (using lactacystin);22 by inhibition of TAP or in TAP-deficient professional APC;16 and by inhibition of the secretory pathway and the trans-Golgi network using brefeldin A.60

The second model proposes that MHC class I molecules encounter and bind exogenously derived peptides in post-Golgi or endolysosomal compartments, in the same way as MHC class II molecules, before being transported to the cell surface. Indeed, MHC class I–β2-microglobulin dimers are present in the endosomes of immature DC and can traffic rapidly to the surface upon encounter with a maturation signal.6163 Specific sorting to endolysosomal compartments is mediated by a highly conserved tyrosine motif within the cytoplasmic tail region of MHC class I. Intriguingly, deletion or mutations of this motif abrogate acquisition and cross-presentation of exogenously derived peptides in vitro and attenuate T lymphocyte responses to immunodominant viral epitopes in vivo.64 Cross-presentation within a endolysosomal compartment is independent of TAP.

Phagocytosis and cross-presentation

Recent revelations regarding the phagocytic process support an alternative molecular model for cross-presentation. This may occur autonomously through an involvement of the ER in the generation of phagosome compartments but in a TAP-dependent manner. It has been a paradigm for more than two decades that the plasma membrane, through invagination, provides all of the membrane required to form complete phagosomes.65 However, active phagocytes are capable of engulfing large numbers of particles without any apparent loss from their plasma membrane. This observation is difficult to explain by membrane regeneration processes alone. Desjardins and colleagues characterized phagosomal membranes by mass spectrometry and two-dimensional gel electrophoresis.66 Surprisingly, these studies revealed that several ER-resident proteins are present in phagosomes. Further analysis showed that phagosomes fuse with the ER during particle engulfment, and that ER membranes constitute a large part of phagosomal membranes.67 Other studies have shown that phagosomes from murine macrophages68 and DC,69 as well as human DC,63 acquire the entire MHC class I loading complex and other elements involved in peptide processing and translocation. They also obtain the sec61 translocon complex, which is capable of transporting entire proteins through membranes. Phagosome compartments retain these ER elements until maturation to phagolysosomes at which point they display increasing numbers of lysosomal markers in parallel with a decrease in luminal pH.

Exogenous antigens from within phagosomes are rapidly translocated to the cytosol within 1–2 hr post-internalisation.68,69 This translocation was also observed with the cholera toxin subunit 1 (CTA1), a known substrate for Sec61,70 suggesting a role for this complex in the translocation events observed. Following export from the phagosome to the cytosol, antigens would become available for degradation by proteasomes into shorter peptides which are suitable for TAP-mediated import into the ER, and thus access the classical pathway for MHC class I presentation. Desjardins and colleagues found that the proteasomal α- and β-subunits are transiently recruited to the cytoplasmic surface of phagosomal membranes. This recruitment peaks approximately 1 hr after phagocytosis and is time independent from the acquisition of ER-luminal proteins. This raises the alternative possibility that peptides derived from phagocytic antigens can be retro-translocated back into the lumen of the phagosome compartment for loading onto MHC class I molecules after proteasomal degradation. Data demonstrating that the TAP molecules and the MHC class I loading complex in phagosomes are fully functional support the idea that the phagosome could function as an autonomous cross-presentation organelle. Indeed, the import of peptides is dependent on TAP, as demonstrated by both antibodies against TAP and the inhibitory peptide ICP47, derived from herpes simplex virus.63 When antigenic peptides were added to isolated phagosomes, or fed to macrophages in the form of whole proteins, MHC class I peptide complexes were detectable 1–2 hr after phagocytosis. Membranes extracted from these vesicles could stimulate interferon-γ production from T-cell receptor transgenic CD8 T cells and responses were inhibited by lactacystin.

Amigorena and colleagues have presented strong evidence that MHC class I loading occurs in the same phagosome compartment utilized for internalization of the antigen.69 Thus, in the current model (Fig. 2), phagocytosed antigens for cross-presentation on MHC class I are internalized in a process identical to that used for degradation of apoptotic bodies and for presentation on MHC class II molecules. After ER-phagosome formation and internalization of antigens, maturation of ER-phagosomes down the endocytic pathway results in acquisition of hydrolases that aid the breakdown of internalized antigens and export into the cytosol.71 In the cytosol, these polypeptides are polyubiquitinated and further degraded by recruited proteasomes. Peptides that are suitable for transport by TAP are then re-imported and loaded onto MHC class I.

Figure 2.

Figure 2

Schematic representation of ER-mediated phagocytosis and antigen cross-presentation in a mixed ER-phagosome compartment. (a) Phagocytosis is initiated by binding and cross-linking of cell surface receptors by particulate antigens. (b) ER membranes fuse with the plasma membrane to form the complete phagosome with a large proportion of ER-derived membranes and proteins including all the major elements of the MHC class I loading complex such as TAP, tapasin, calnexin, MHC class I heavy chain, ER chaperones and disulphide isomerase (Grp78, ERp57), ER-aminopeptidase associated with peptide trimming (ERAP) and the peptide translocation channel (Sec61). (c) Phagosomes gradually progress through the endocytic pathway and acquire hydrolases such as cathepsin D. (d) Partial proteolysis by cathepsins generates polypeptides which are suitable for export into the cytosol by Sec61. (e) Cytosolic peptides are poly-ubiquitinated by the ubiquitinating enzyme complex (UBC) and are substrates for proteasomal degradation. Processed peptides are re-imported by the TAP and trimmed by ERAP. MHC class I molecules are loaded by the MHC class I loading complex. (f) Phagosomes containing loaded MHC class I may recycle back to the plasma membrane by exocytosis. Specific inhibitors (depicted in red) that interfere at various points in the cross-presentation pathway have allowed the development of the current model.

Macropinocytosis and cross-presentation

The extension of the phagosome loading compartment model to DC remains controversial as the phagocytic activity of these cells is thought to be significantly less than that of macrophages or neutrophils. However, DC are highly efficient in macropinocytosis. Watts and colleagues demonstrated that exogenous antigenic material delivered to the interior of the DC via macropinocytosis could be processed and presented on MHC class I.72 Whether the formation of macropinosomes involves ER membrane fusion remains to be formally demonstrated, but there are emerging data suggesting that soluble antigens can be cross-presented in the same way as phagocytosed antigens. DC form large numbers of membrane-bound macropinocytic vesicles without a detectable loss from the plasma membrane.

Peter Cresswell and colleagues first demonstrated that soluble proteins internalized by marcopinocytosis can access the MHC class I peptide-loading complex. Internalization of soluble ovalbumin and an anti-tapasin antibody appeared in similar peripheral vesicles, some of them positive for the ER marker calnexin.63 Also, the group utilized a soluble form of the TAP inhibitor US6, a human cytomegalovirus membrane protein, to show the TAP dependency of this process. Cross-presentation of soluble ovalbumin in the presence of US6 was inhibited in a dose-dependent manner.

Concluding remarks

Data linking aspects of ER membrane trafficking with the immunological phenomena of cross-presentation provides compelling evidence for the existence of a specific cross-presentation organelle. From a mechanistic point of view, this model is advantageous as internalized antigens could potentially avoid direct competition with DRiPs for access to TAP and the class I loading complex in the ER. Moreover, it has been demonstrated that phagosome loaded MHC class I peptide complexes are rapidly displayed to the immune system within just 1 hr ‘post’ internalization.69

However, a number of questions about this model remain. It has been suggested (but not formally demonstrated) that the export of polypeptides from phagosome compartments is mediated by the ER translocon Sec61. Localization of Sec61 is usually restricted to the rough ER where it associates with ribosomal subunits.73 The presence of ribosomes on phagosomes has never been demonstrated.

Moreover, the relevance of this model for in vivo cross-presentation has not been addressed and the efficiency of MHC class-I loading in phagosomes remains unclear. How many molecules can be loaded within this compartment, are they all derived from the ER, or to what extend from recycling MHC class I. Using an approach designed for looking at intracellular protein trafficking, Cresswell and colleagues demonstrated that most loaded phagosomal MHC class I molecules are sensitive to endoglycosidase H (endoH) digestion, which results from a lack of post-translational modification within the medial-Golgi.63 This suggests that MHC class I molecules in phagosomes must be derived primarily as newly synthesized protein from the ER rather than by recycling from the plasma membrane.63 In addition, such nascent MHC class I/peptide complexes are also present on the plasma membrane. This is at least suggestive of direct transport back to the cell surface.

Hence, by utilizing similar approaches based on a greater understanding of the cell biology that underlies cross-presentation, immunologists will be able to provide definitive answers to the remaining questions.

References

  • 1.Guermonprez P, Valladeau J, Zitvogel L, Thery C, Amigorena S. Antigen presentation and T cell stimulation by dendritic cells. Annu Rev Immunol. 2002;20:621–67. doi: 10.1146/annurev.immunol.20.100301.064828. [DOI] [PubMed] [Google Scholar]
  • 2.Princiotta MF, Finzi D, Qian SB, Gibbs J, Schuchmann S, Buttgereit F, Bennink JR, Yewdell JW. Quantitating protein synthesis, degradation, and endogenous antigen processing. Immunity. 2003;18:343–54. doi: 10.1016/s1074-7613(03)00051-7. [DOI] [PubMed] [Google Scholar]
  • 3.Lelouard H, Gatti E, Cappello F, Gresser O, Camosseto V, Pierre P. Transient aggregation of ubiquitinated proteins during dendritic cell maturation. Nature. 2002;417:177–82. doi: 10.1038/417177a. [DOI] [PubMed] [Google Scholar]
  • 4.Lelouard H, Ferrand V, Marguet D, Bania J, Camosseto V, David A, Gatti E, Pierre P. Dendritic cell aggresome-like induced structures are dedicated areas for ubiquitination and storage of newly synthesized defective proteins. J Cell Biol. 2004;164:667–75. doi: 10.1083/jcb.200312073. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 5.York IA, Chang SC, Saric T, Keys JA, Favreau JM, Goldberg AL, Rock KL. The ER aminopeptidase ERAP1 enhances or limits antigen presentation by trimming epitopes to 8–9 residues. Nat Immunol. 2002;3:1177–84. doi: 10.1038/ni860. [DOI] [PubMed] [Google Scholar]
  • 6.Bevan MJ. Cross-priming for a secondary cytotoxic response to minor H antigens with H-2 congenic cells which do not cross-react in the cytotoxic assay. J Exp Med. 1976;143:1283–8. doi: 10.1084/jem.143.5.1283. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 7.Bevan MJ. Minor H antigens introduced on H-2 different stimulating cells cross-react at the cytotoxic T cell level during in vivo priming. J Immunol. 1976;117:2233–8. [PubMed] [Google Scholar]
  • 8.Limmer A, Ohl J, Kurts C, et al. Efficient presentation of exogenous antigen by liver endothelial cells to CD8 T cells results in antigen-specific T-cell tolerance. Nat Med. 2000;6:1348–54. doi: 10.1038/82161. [DOI] [PubMed] [Google Scholar]
  • 9.Heit A, Huster KM, Schmitz F, Schiemann M, Busch DH, Wagner H. CpG-DNA aided cross-priming by cross-presenting B cells. J Immunol. 2004;172:1501–7. doi: 10.4049/jimmunol.172.3.1501. [DOI] [PubMed] [Google Scholar]
  • 10.Huang AY, Golumbek P, Ahmadzadeh M, Jaffee E, Pardoll D, Levitsky H. Role of bone marrow-derived cells in presenting MHC class I-restricted tumor antigens. Science. 1994;264:961–5. doi: 10.1126/science.7513904. [DOI] [PubMed] [Google Scholar]
  • 11.Bennett SR, Carbone FR, Karamalis F, Miller JF, Heath WR. Induction of a CD8+ cytotoxic T lymphocyte response by cross-priming requires cognate CD4+ T cell help. J Exp Med. 1997;186:65–70. doi: 10.1084/jem.186.1.65. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 12.Kurts C, Cannarile M, Klebba I, Brocker T. Dendritic cells are sufficient to cross-present self-antigens to CD8 T cells in vivo. J Immunol. 2001;166:1439–42. doi: 10.4049/jimmunol.166.3.1439. [DOI] [PubMed] [Google Scholar]
  • 13.Sigal LJ, Crotty S, Andino R, Rock KL. Cytotoxic T-cell immunity to virus-infected non-haematopoietic cells requires presentation of exogenous antigen. Nature. 1999;398:77–80. doi: 10.1038/18038. [DOI] [PubMed] [Google Scholar]
  • 14.Freigang S, Egger D, Bienz K, Hengartner H, Zinkernagel RM. Endogenous neosynthesis vs. cross-presentation of viral antigens for cytotoxic T cell priming. Proc Natl Acad Sci USA. 2003;100:13477–82. doi: 10.1073/pnas.1835685100. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 15.Carbone FR, Kurts C, Bennett SR, Miller JF, Heath WR. Cross-presentation: a general mechanism for CTL immunity and tolerance. Immunol Today. 1998;19:368–73. doi: 10.1016/s0167-5699(98)01301-2. [DOI] [PubMed] [Google Scholar]
  • 16.den Haan JM, Lehar SM, Bevan MJ. CD8(+) but not CD8(–) dendritic cells cross-prime cytotoxic T cells in vivo. J Exp Med. 2000;192:1685–96. doi: 10.1084/jem.192.12.1685. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 17.Pooley JL, Heath WR, Shortman K. Cutting edge: intravenous soluble antigen is presented to CD4 T cells by CD8– dendritic cells, but cross-presented to CD8 T cells by CD8+ dendritic cells. J Immunol. 2001;166:5327–30. doi: 10.4049/jimmunol.166.9.5327. [DOI] [PubMed] [Google Scholar]
  • 18.Cella M, Engering A, Pinet V, Pieters J, Lanzavecchia A. Inflammatory stimuli induce accumulation of MHC class II complexes on dendritic cells. Nature. 1997;388:782–7. doi: 10.1038/42030. [DOI] [PubMed] [Google Scholar]
  • 19.Iyoda T, Shimoyama S, Liu K, Omatsu Y, Akiyama Y, Maeda Y, Takahara K, Steinman RM, Inaba K. The CD8+ dendritic cell subset selectively endocytoses dying cells in culture and in vivo. J Exp Med. 2002;195:1289–302. doi: 10.1084/jem.20020161. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 20.Steinman RM, Inaba K, Turley S, Pierre P, Mellman I. Antigen capture, processing, and presentation by dendritic cells: recent cell biological studies. Hum Immunol. 1999;60:562–7. doi: 10.1016/s0198-8859(99)00030-0. [DOI] [PubMed] [Google Scholar]
  • 21.Shen Z, Reznikoff G, Dranoff G, Rock KL. Cloned dendritic cells can present exogenous antigens on both MHC class I and class II molecules. J Immunol. 1997;158:2723–30. [PubMed] [Google Scholar]
  • 22.Rodriguez A, Regnault A, Kleijmeer M, Ricciardi-Castagnoli P, Amigorena S. Selective transport of internalized antigens to the cytosol for MHC class I presentation in dendritic cells. Nat Cell Biol. 1999;1:362–68. doi: 10.1038/14058. [DOI] [PubMed] [Google Scholar]
  • 23.Rock KL, Rothstein L, Gamble S, Fleischacker C. Characterization of antigen-presenting cells that present exogenous antigens in association with class I MHC molecules. J Immunol. 1993;150:438–46. [PubMed] [Google Scholar]
  • 24.Albert ML, Pearce SF, Francisco LM, Sauter B, Roy P, Silverstein RL, Bhardwaj N. Immature dendritic cells phagocytose apoptotic cells via alphavbeta5 and CD36, and cross-present antigens to cytotoxic T lymphocytes. J Exp Med. 1998;188:1359–68. doi: 10.1084/jem.188.7.1359. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 25.Albert ML, Sauter B, Bhardwaj N. Dendritic cells acquire antigen from apoptotic cells and induce class I-restricted CTLs. Nature. 1998;392:86–9. doi: 10.1038/32183. [DOI] [PubMed] [Google Scholar]
  • 26.Sauter B, Albert ML, Francisco L, Larsson M, Somersan S, Bhardwaj N. Consequences of cell death: exposure to necrotic tumor cells, but not primary tissue cells or apoptotic cells, induces the maturation of immunostimulatory dendritic cells. J Exp Med. 2000;191:423–34. doi: 10.1084/jem.191.3.423. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 27.Larsson M, Fonteneau JF, Somersan S, Sanders C, Bickham K, Thomas EK, Mahnke K, Bhardwaj N. Efficiency of cross presentation of vaccinia virus-derived antigens by human dendritic cells. Eur J Immunol. 2001;31:3432–42. doi: 10.1002/1521-4141(200112)31:12<3432::aid-immu3432>3.0.co;2-r. [DOI] [PubMed] [Google Scholar]
  • 28.Motta I, Andre F, Lim A, Tartaglia J, Cox WI, Zitvogel L, Angevin E, Kourilsky P. Cross-presentation by dendritic cells of tumor antigen expressed in apoptotic recombinant canarypox virus-infected dendritic cells. J Immunol. 2001;167:1795–802. doi: 10.4049/jimmunol.167.3.1795. [DOI] [PubMed] [Google Scholar]
  • 29.Schulz O, Reis e Sousa C. Cross-presentation of cell-associated antigens by CD8alpha+ dendritic cells is attributable to their ability to internalize dead cells. Immunology. 2002;107:183–9. doi: 10.1046/j.1365-2567.2002.01513.x. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 30.Schulz O, Pennington DJ, Hodivala-Dilke K, Febbraio M, Reis e Sousa C. CD36 or alphavbeta3 and alphavbeta5 integrins are not essential for MHC class I cross-presentation of cell-associated antigen by CD8 alpha+ murine dendritic cells. J Immunol. 2002;168:6057–65. doi: 10.4049/jimmunol.168.12.6057. [DOI] [PubMed] [Google Scholar]
  • 31.Jung S, Unutmaz D, Wong P, et al. In vivo depletion of CD11c(+) dendritic cells abrogates priming of CD8(+) T cells by exogenous cell-associated antigens. Immunity. 2002;17:211–20. doi: 10.1016/s1074-7613(02)00365-5. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 32.Mellman I. Endocytosis and molecular sorting. Annu Rev Cell Dev Biol. 1996;12:575–625. doi: 10.1146/annurev.cellbio.12.1.575. [DOI] [PubMed] [Google Scholar]
  • 33.Brown EJ. The role of extracellular matrix proteins in the control of phagocytosis. J Leukoc Biol. 1986;39:579–91. doi: 10.1002/jlb.39.5.579. [DOI] [PubMed] [Google Scholar]
  • 34.Regnault A, Lankar D, Lacabanne V, et al. Fcgamma receptor-mediated induction of dendritic cell maturation and major histocompatibility complex class I-restricted antigen presentation after immune complex internalization. J Exp Med. 1999;189:371–80. doi: 10.1084/jem.189.2.371. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 35.Machy P, Serre K, Leserman L. Class I-restricted presentation of exogenous antigen acquired by Fcgamma receptor-mediated endocytosis is regulated in dendritic cells. Eur J Immunol. 2000;30:848–57. doi: 10.1002/1521-4141(200003)30:3<848::AID-IMMU848>3.0.CO;2-Q. [DOI] [PubMed] [Google Scholar]
  • 36.Rovere P, Sabbadini MG, Vallinoto C, et al. Dendritic cell presentation of antigens from apoptotic cells in a proinflammatory context: role of opsonizing anti-beta2-glycoprotein I antibodies. Arthritis Rheum. 1999;42:1412–20. doi: 10.1002/1529-0131(199907)42:7<1412::AID-ANR15>3.0.CO;2-T. [DOI] [PubMed] [Google Scholar]
  • 37.Fadok VA, Bratton DL, Rose DM, Pearson A, Ezekewitz RA, Henson PM. A receptor for phosphatidylserine-specific clearance of apoptotic cells. Nature. 2000;405:85–90. doi: 10.1038/35011084. [DOI] [PubMed] [Google Scholar]
  • 38.Belz GT, Vremec D, Febbraio M, Corcoran L, Shortman K, Carbone FR, Heath WR. CD36 is differentially expressed by CD8+ splenic dendritic cells but is not required for cross-presentation in vivo. J Immunol. 2002;168:6066–70. doi: 10.4049/jimmunol.168.12.6066. [DOI] [PubMed] [Google Scholar]
  • 39.Ezekowitz RA, Sastry K, Bailly P, Warner A. Molecular characterization of the human macrophage mannose receptor: demonstration of multiple carbohydrate recognition-like domains and phagocytosis of yeasts in Cos-1 cells. J Exp Med. 1990. pp. 1785–94. [DOI] [PMC free article] [PubMed]
  • 40.Engering AJ, Cella M, Fluitsma DM, Hoefsmit EC, Lanzavecchia A, Pieters J. Mannose receptor mediated antigen uptake and presentation in human dendritic cells. Adv Exp Med Biol. 1997;417:183–7. doi: 10.1007/978-1-4757-9966-8_31. [DOI] [PubMed] [Google Scholar]
  • 41.Brown GD, Gordon S. Immune recognition. A new receptor for beta-glucans. Nature. 2001;413:36–7. doi: 10.1038/35092620. [DOI] [PubMed] [Google Scholar]
  • 42.Hampton RY, Golenbock DT, Penman M, Krieger M, Raetz CR. Recognition and plasma clearance of endotoxin by scavenger receptors. Nature. 1991;352:342–4. doi: 10.1038/352342a0. [DOI] [PubMed] [Google Scholar]
  • 43.Moroi Y, Mayhew M, Trcka J, Hoe MH, Takechi Y, Hartl FU, Rothman JE, Houghton AN. Induction of cellular immunity by immunization with novel hybrid peptides complexed to heat shock protein 70. Proc Natl Acad Sci USA. 2000;97:3485–90. doi: 10.1073/pnas.070550797. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 44.Suzue K, Young RA. Adjuvant-free hsp70 fusion protein system elicits humoral and cellular immune responses to HIV-1 p24. J Immunol. 1996;156:873–9. [PubMed] [Google Scholar]
  • 45.Tamura Y, Peng P, Liu K, Daou M, Srivastava PK. Immunotherapy of tumors with autologous tumor-derived heat shock protein. Science. 1997;278:117–20. doi: 10.1126/science.278.5335.117. [DOI] [PubMed] [Google Scholar]
  • 46.Blachere NE, Li Z, Chandawarkar RY, Suto R, Jaikaria NS, Basu S, Udono H, Srivastava PK. Heat shock protein-peptide complexes, reconstituted in vitro, elicit peptide-specific cytotoxic T lymphocyte response and tumor immunity. J Exp Med. 1997;186:1315–22. doi: 10.1084/jem.186.8.1315. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 47.Binder RJ, Blachere NE, Srivastava PK. Heat shock protein-chaperoned peptides but not free peptides introduced into the cytosol are presented efficiently by major histocompatibility complex I molecules. J Biol Chem. 2001;276:17163–71. doi: 10.1074/jbc.M011547200. [DOI] [PubMed] [Google Scholar]
  • 48.Huang Q, Richmond JF, Suzue K, Eisen HN, Young RA. In vivo cytotoxic T lymphocyte elicitation by mycobacterial heat shock protein 70 fusion proteins maps to a discrete domain and is CD4 (+) T cell independent. J Exp Med. 2000;191:403–8. doi: 10.1084/jem.191.2.403. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 49.MacAry PA, Javid B, Floto RA, Smith KG, Oehlmann W, Singh M, Lehner PJ. HSP70 peptide binding mutants separate antigen delivery from dendritic cell stimulation. Immunity. 2004;20:95–106. doi: 10.1016/s1074-7613(03)00357-1. [DOI] [PubMed] [Google Scholar]
  • 50.Basu S, Binder RJ, Ramalingam T, Srivastava PK. CD91 is a common receptor for heat shock proteins gp96, hsp90, hsp70, and calreticulin. Immunity. 2001;14:303–13. doi: 10.1016/s1074-7613(01)00111-x. [DOI] [PubMed] [Google Scholar]
  • 51.Binder RJ, Han DK, Srivastava PK. CD91: a receptor for heat shock protein gp96. Nat Immunol. 2000;1:151–55. doi: 10.1038/77835. [DOI] [PubMed] [Google Scholar]
  • 52.Delneste Y, Magistrelli G, Gauchat J, et al. Involvement of LOX-1 in dendritic cell-mediated antigen cross-presentation. Immunity. 2002;17:353–62. doi: 10.1016/s1074-7613(02)00388-6. [DOI] [PubMed] [Google Scholar]
  • 53.Berwin B, Hart JP, Rice S, Gass C, Pizzo SV, Post SR, Nicchitta CV. Scavenger receptor-A mediates gp96/GRP94 and calreticulin internalization by antigen-presenting cells. Embo J. 2003;22:6127–36. doi: 10.1093/emboj/cdg572. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 54.Rock KL, Gamble S, Rothstein L, Benacerraf B. Reassociation with beta 2-microglobulin is necessary for Db class I major histocompatibility complex binding of an exogenous influenza peptide. Proc Natl Acad Sci USA. 1991;88:301–4. doi: 10.1073/pnas.88.1.301. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 55.Rock KL, Gramm C, Benacerraf B. Low temperature and peptides favor the formation of class I heterodimers on RMA-S cells at the cell surface. Proc Natl Acad Sci USA. 1991;88:4200–4. doi: 10.1073/pnas.88.10.4200. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 56.Spies T, DeMars R. Restored expression of major histocompatibility class I molecules by gene transfer of a putative peptide transporter. Nature. 1991;351:323–4. doi: 10.1038/351323a0. [DOI] [PubMed] [Google Scholar]
  • 57.Lehner PJ, Surman MJ, Cresswell P. Soluble tapasin restores MHC class I expression and function in the tapasin-negative cell line.220. Immunity. 1998;8:221–31. doi: 10.1016/s1074-7613(00)80474-4. [DOI] [PubMed] [Google Scholar]
  • 58.Arrode G, Boccaccio C, Lule J, Allart S, Moinard N, Abastado JP, Alam A, Davrinche C. Incoming human cytomegalovirus pp65 (UL83) contained in apoptotic infected fibroblasts is cross-presented to CD8(+) T cells by dendritic cells. J Virol. 2000;74:10018–24. doi: 10.1128/jvi.74.21.10018-10024.2000. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 59.Ramirez MC, Sigal LJ. Macrophages and dendritic cells use the cytosolic pathway to rapidly cross-present antigen from live, vaccinia-infected cells. J Immunol. 2002;169:6733–42. doi: 10.4049/jimmunol.169.12.6733. [DOI] [PubMed] [Google Scholar]
  • 60.Kovacsovics-Bankowski M, Rock KL. A phagosome-to-cytosol pathway for exogenous antigens presented on MHC class I molecules. Science. 1995;267:243–6. doi: 10.1126/science.7809629. [DOI] [PubMed] [Google Scholar]
  • 61.MacAry PA, Lindsay M, Scott MA, Craig JI, Luzio JP, Lehner PJ. Mobilization of MHC class I molecules from late endosomes to the cell surface following activation of CD34-derived human Langerhans cells. Proc Natl Acad Sci USA. 2001;98:3982–7. doi: 10.1073/pnas.071477498. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 62.Kleijmeer MJ, Escola JM, UytdeHaag FG, et al. Antigen loading of MHC class I molecules in the endocytic tract. Traffic. 2001;2:124–37. doi: 10.1034/j.1600-0854.2001.020207.x. [DOI] [PubMed] [Google Scholar]
  • 63.Ackerman AL, Cresswell P. Regulation of MHC class I transport in human dendritic cells and the dendritic-like cell line KG-1. J Immunol. 2003;170:4178–88. doi: 10.4049/jimmunol.170.8.4178. [DOI] [PubMed] [Google Scholar]
  • 64.Lizee G, Basha G, Tiong J, Julien JP, Tian M, Biron KE, Jefferies WA. Control of dendritic cell cross-presentation by the major histocompatibility complex class I cytoplasmic domain. Nat Immunol. 2003;4:1065–73. doi: 10.1038/ni989. [DOI] [PubMed] [Google Scholar]
  • 65.Werb Z, Cohn ZA. Plasma membrane synthesis in the macrophage following phagocytosis of polystyrene latex particles. J Biol Chem. 1972;247:2439–46. [PubMed] [Google Scholar]
  • 66.Garin J, Diez R, Kieffer S, et al. The phagosome proteome: insight into phagosome functions. J Cell Biol. 2001;152:165–80. doi: 10.1083/jcb.152.1.165. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 67.Gagnon E, Duclos S, Rondeau C, et al. Endoplasmic reticulum-mediated phagocytosis is a mechanism of entry into macrophages. Cell. 2002;110:119–37. doi: 10.1016/s0092-8674(02)00797-3. [DOI] [PubMed] [Google Scholar]
  • 68.Houde M, Bertholet S, Gagnon E, et al. Phagosomes are competent organelles for antigen cross-presentation. Nature. 2003;425:402–6. doi: 10.1038/nature01912. [DOI] [PubMed] [Google Scholar]
  • 69.Guermonprez P, Saveanu L, Kleijmeer M, Davoust J, Van Endert P, Amigorena S. ER-phagosome fusion defines an MHC class I cross-presentation compartment in dendritic cells. Nature. 2003;425:397–402. doi: 10.1038/nature01911. [DOI] [PubMed] [Google Scholar]
  • 70.Schmitz A, Herrgen H, Winkeler A, Herzog V. Cholera toxin is exported from microsomes by the Sec61p complex. J Cell Biol. 2000;148:1203–12. doi: 10.1083/jcb.148.6.1203. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 71.Fonteneau JF, Kavanagh DG, Lirvall M, Sanders C, Cover TL, Bhardwaj N, Larsson M. Characterization of the MHC class I cross-presentation pathway for cell-associated antigens by human dendritic cells. Blood. 2003;102:4448–55. doi: 10.1182/blood-2003-06-1801. [DOI] [PubMed] [Google Scholar]
  • 72.Norbury CC, Hewlett LJ, Prescott AR, Shastri N, Watts C. Class I MHC presentation of exogenous soluble antigen via macropinocytosis in bone marrow macrophages. Immunity. 1995;3:783–91. doi: 10.1016/1074-7613(95)90067-5. [DOI] [PubMed] [Google Scholar]
  • 73.Levy R, Wiedmann M, Kreibich G. In vitro binding of ribosomes to the beta subunit of the Sec61p protein translocation complex. J Biol Chem. 2001;276:2340–6. doi: 10.1074/jbc.M004867200. [DOI] [PubMed] [Google Scholar]

Articles from Immunology are provided here courtesy of British Society for Immunology

RESOURCES