Next Article in Journal / Special Issue
Functions of the Proteasome on Chromatin
Previous Article in Journal
Long Chain Fatty Acid Acylated Derivatives of Quercetin-3-O-Glucoside as Antioxidants to Prevent Lipid Oxidation
Previous Article in Special Issue
Nuclear Transport of Yeast Proteasomes
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Proteasome Subtypes and Regulators in the Processing of Antigenic Peptides Presented by Class I Molecules of the Major Histocompatibility Complex

by
Nathalie Vigneron
1,2,3,* and
Benoît J. Van den Eynde
1,2,3
1
Ludwig Institute for Cancer Research, Brussels 1200, Belgium
2
WELBIO (Walloon Excellence in Life Sciences and Biotechnology), Brussels 1200, Belgium
3
De Duve Institute, Université catholique de Louvain, Brussels 1200, Belgium
*
Author to whom correspondence should be addressed.
Biomolecules 2014, 4(4), 994-1025; https://doi.org/10.3390/biom4040994
Submission received: 30 July 2014 / Revised: 2 October 2014 / Accepted: 29 October 2014 / Published: 18 November 2014
(This article belongs to the Special Issue Proteasomes and Its Regulators)

Abstract

:
The proteasome is responsible for the breakdown of cellular proteins. Proteins targeted for degradation are allowed inside the proteasome particle, where they are cleaved into small peptides and released in the cytosol to be degraded into amino acids. In vertebrates, some of these peptides escape degradation in the cytosol, are loaded onto class I molecules of the major histocompatibility complex (MHC) and displayed at the cell surface for scrutiny by the immune system. The proteasome therefore plays a key role for the immune system: it provides a continued sampling of intracellular proteins, so that CD8-positive T-lymphocytes can kill cells expressing viral or tumoral proteins. Consequently, the repertoire of peptides displayed by MHC class I molecules at the cell surface depends on proteasome activity, which may vary according to the presence of proteasome subtypes and regulators. Besides standard proteasomes, cells may contain immunoproteasomes, intermediate proteasomes and thymoproteasomes. Cells may also contain regulators of proteasome activity, such as the 19S, PA28 and PA200 regulators. Here, we review the effects of these proteasome subtypes and regulators on the production of antigenic peptides. We also discuss an unexpected function of the proteasome discovered through the study of antigenic peptides: its ability to splice peptides.

1. Introduction to the Ubiquitin Proteasome System

The ubiquitin-proteasome system (UPS) is the major ATP-dependent protein degradation system in cells. It is essential to maintain cellular protein homeostasis and ensure the elimination of misfolded proteins (reviewed in [1,2]). Proteasome-dependent proteolysis is also essential to regulate a number of other cellular processes, such as cell differentiation, cell-cycle progression or apoptosis. Proteins targeted for degradation by the proteasome are tagged with polyubiquitin chains that are attached to lysine residues within the protein sequence. These ubiquitin-tagged proteins are recognized by the regulatory particle 19S (PA700), which is associated with the 20S proteasome catalytic core to form the 26S proteasome. Upon degradation, these ubiquitin-tagged proteins are degraded into small peptides of about three to 22 amino acids [3], which can be further degraded by cytosolic peptidases to recycle the pool of amino acids [4]. The immune system takes advantage of this system to monitor cellular integrity. Thus, a fraction of the peptides released by proteasomal degradation is transferred into the lumen of the endoplasmic reticulum (ER) by a dedicated transporter, named TAP (transporter associated with antigen processing) [5]. Inside the ER, those peptides will be further trimmed by ER-resident proteases, and peptides of an appropriate size (8–10 amino acids) and sequence will then associate with class I molecules of the major histocompatibility complex (MHC). Peptide-MHC complexes are finally displayed at the cell surface for potential recognition by cytolytic T-lymphocytes (CTLs). CTLs are major sentinels poised to rapidly recognize and destroy cells expressing mutant, infectious or tumoral proteins. It is the display of peptides (named antigenic peptides) derived from such altered proteins that marks cells for CTL recognition. The sensitivity of CTL, which can recognize target cells expressing as little as 10 molecules of antigenic peptide [6], enables the detection of only subtle changes in the cellular protein content. The detailed characterization of the antigenic peptides recognized by anti-tumor CTL that were isolated from the blood or tumor of cancer patients has provided valuable insights into the processing of antigenic peptides [7,8]. Such peptides usually derive from mutated proteins [9,10], from differentiation proteins [11,12] and from tumor proteins encoded by cancer-germline genes, such as the MAGE genes [13].

2. Role of the Proteasome in Antigen Processing

The involvement of the proteasome in the production of MHC class I peptides was originally stressed by the observation that the presentation of the model antigen, ovalbumin, was dependent on ubiquitination and, therefore, on the 26S proteasome [14,15]. In line with this, MHC class I presentation was accelerated when the N-terminus of protein antigens was genetically modified with a bulky or a charged residue to increase their rate of ubiquitination by the ubiquitin ligase E3 α (N-end rule) [16,17,18]. Another piece of evidence for the involvement of the proteasome in antigen processing came from the observation that proteasome inhibitors blocked the presentation of a number of antigenic peptides while limiting the overall peptide supply to MHC class I [19,20,21,22,23,24,25,26,27]. Although inhibitors reduced the cell ability to express specific antigenic peptides from full-length proteins, two studies showed that production of the antigenic peptides from minigenes of the correct size was not affected, suggesting that the inhibitors indeed specifically act on the peptide release from the full-length protein [19,28]. However, the role of proteasome inhibitors is often hard to interpret, first because inhibitors generally induce ER stress, which can affect the rate of synthesis of some proteins [29,30], and second, because they inhibit some catalytic activities of the proteasome, but not others. Their effect may therefore vary according to the main catalytic activity involved in the production of the peptide studied. In addition, the production of a number of peptides depends on a balance between productive cleavages, occurring mostly at the C-terminus, and destructive cleavages occurring within the sequence of the antigenic peptide [31,32]. The net effect of proteasome inhibitors on the production of such peptides therefore depends on the relative inhibition of the catalytic activities responsible for the productive versus destructive cleavages [33]. Studies on the presentation of constructs involving antigenic peptides extended at the C-terminus or the N-terminus indicated that the proteasome was usually required to produce the C-terminal cleavage, but was dispensable for the N-terminal cleavage [28,34]. Subsequent studies showed that N-terminally extended peptide precursors produced by the proteasome can be further trimmed by other peptidases, either in the cytosol by tripeptidyl peptidase II (TPPII) [35] or, more prominently, in the ER by the endoplasmic reticulum aminopeptidase ERAP1 [36,37]. Another strong argument for the role of the proteasome in the production of class I peptides lies in the observation that many antigenic peptides are differentially processed by the various proteasomes subtypes, as we will discuss below (Table 1) [31,32,38].
Although the proteasome is considered as the main source of antigenic peptides, it is now clear that it is not the only protease able to generate antigenic peptides [27,39]. Several alternative cytosolic proteases were shown to process specific antigenic peptides, including tripeptidyl peptidase II (TPPII) and the metallopeptidases thimet oligopeptidase, nardilysin and insulin degrading enzyme (IDE) [40,41,42,43,44]. However, the function of these proteases in general MHC class I presentation appears quite limited [45,46,47]. Additionally, antigenic peptides can also be produced by proteases present in the ER or in the secretory compartment. For example, signal peptidase and/or signal peptide peptidase are both involved in the production of TAP-independent peptides derived from signal sequences [48,49]. Moreover, in cells with a defect in peptide loading complex quality control, trans-Golgi proteases, such as protease pro-protein convertase 7, can rescue a significant proportion of the unstable MHC class I peptides, potentially by making peptides available from full-length proteins [50]. Coupling a murine cytomegalovirus (CMV) epitope to the C-terminus of the secretory hepatitis B protein, HBe, Gil-Torregrosa et al. showed that the trans-Golgi resident protease, furin, which is involved in HBe maturation along the secretory route, helped with releasing the CMV peptide for loading onto H-2Ld in TAP-deficient cells [51]. Finally, another alternative pathway involved in the presentation of MHC class I-restricted peptides is autophagy, a process that enables non-proteasomal degradation of proteins at steady-state and in conditions of stress, such as starvation or oxidative damage. In the MHC class I pathway, autophagy appears to contribute to the processing of a growing number of antigenic peptides presented by MHC class I and derived from endogenously expressed proteins [52,53,54]. In the autophagy-based MHC class I processing, cellular proteins are brought to the autophagosome/lysosomal compartment for degradation, and the peptides released are then loaded onto local MHC class I. This process is TAP-independent and gives rise to peptides that can be similar to those produced through the classical proteasome processing pathway [52,53,54]. Although the relative importance of autophagy in antigen processing still needs to be evaluated further, this process may be important to circumvent viral or tumoral evasion strategies affecting TAP transport. In this review, we will recapitulate the role played by the proteasome and its regulators in the processing of antigenic peptides.
Table 1. Processing efficiency of tumor antigens by the different proteasome types.
Table 1. Processing efficiency of tumor antigens by the different proteasome types.
Peptide sourceMHC restrictionPeptide SequenceStandard ProteasomeImmuno-proteasomeIntermediate Proteasome β5iIntermediate Proteasome β1i–β5iReferences
Self
RU134–42HLA-B51VPYGSFKHV+ + §+/−n.d.n.d.[20,31]
FGF-5172–176 and 217–220 *HLA-A3NTYAS_PRFK+ + n.d.n.d.[55,56]
Differentiation
gp10040–42 and 47–52 *HLA-A32RTK_QLYPEW+ + +/−n.d.n.d.[22,56]
gp100209–217HLA-A2ITDQVPFSV+ + +/−+/−+/−[12,31,38]
gp100195–202 and 191 or 192 *HLA-A3RSYVPLAH_R+ +n.d.n.d.n.d.[57]
Tyrosinase369−377HLA-A2YMDGTMSQV+ + +/−+/−[31,38,58]
Tyrosinase368–373 and 336–340 *HLA-A24IYMDGT_ADFSF+ + n.d.n.d.[59]
Melan-A26–35HLA-A2EAAGIGILTV+ ++/−+/−[38,60]
Cancer germline
MAGE-A3114–122HLA-B40AELVHFLLL+ ++ ++ +[31,38,61]
MAGE-A3271–279HLA-A2FLWGPRALV+ +[32,62]
MAGE-A10254–262HLA-A2GLYDGMEHL+ +[32,63]
MAGE-C2336–344HLA-A2ALKDVEERV+ ++ +[31,38,64]
MAGE-C242–50HLA-B57ASSTLYLVF+ ++ ++ +[38,65]
MAGE-C2191–200HLA-A2LLFGLALIEV+ +[38,64]
Mutation
CLPP240–248HLA-A2ILDKVLVHL+/−+ + n.d.n.d.[66]
HAUS3154–162HLA-A2ILNAMIAKIn.d.n.d.+ +[67]
Polymorphism (mHC ||)
SP110296–301 and 286−289 *HLA-A3SLPRGT_STPK+/−+ +n.d.n.d.[56,68]
* Spliced peptide. § + +, efficiently produced; +/−, slightly produced; −, not produced; n.d., not determined. || mHC, minor histocompatibility antigen.

3. The 20S Proteasome

The 20S proteasome is a cylindric particle of about 700 kDa, which is composed of four stacked heptameric rings, each comprising seven subunits. The two outer rings are identical and made of structural α subunits, and the two inner rings each contain seven β subunits, three of which mediate the catalytic activity of the proteasome: β1, β2 and β5 (Figure 1A). Mutagenesis studies and the analysis of the crystal structure of proteasomes complexed with inhibitors showed that peptide bond hydrolysis is mediated by the N-terminal threonine of the catalytically-active β subunits, which is exposed to the luminal side of the proteasome particle, in the catalytic chamber [69,70,71,72]. The hydroxyl group of the side chain of this threonine produces a nucleophilic attack on the carbonyl of the peptide bond, leading to the formation of an acyl-enzyme intermediate in which a peptide fragment remains attached to the proteasome by an ester link. Water molecules present in the chamber rapidly hydrolyze this acyl-enzyme intermediate, releasing a peptide fragment that is then transferred back into the cytosol [73]. A few years ago, we found that the proteasome was also able to produce antigenic peptides by creating a peptide bond between peptide fragments originally distant in the parental protein [22]. So far, five antigenic peptides were shown to be produced by this new function of the proteasome, called peptide splicing (Table 1) [22,55,57,59,68,74]. We showed that peptide splicing takes place in the proteasome chamber, through a process of transpeptidation that involves the nucleophilic attack of the acyl-enzyme intermediate by the N-terminus of a peptide fragment present in the catalytic chamber (Figure 2) [22]. Interestingly, among the five spliced peptides identified to date, three were composed of two non-contiguous peptide fragments spliced together in the reverse order to that in which they occur in the parental protein [57,59,68]. Given their nature, spliced antigenic peptides therefore appear as byproducts of the normal proteasome degradation activity. This process leads to the production of a large variety of peptides, as peptide splicing could theoretically involve any peptide fragment released by proteolysis, provided that the nucleophile peptide contains at least three amino acids [57]. Studying the peptide RSYVPLAH_R (splicing of fragment RSYVPLAH with a single arginine) derived from gp100 and presented by HLA-A3, we showed that this minimal size requirement for the nucleophile peptide resulted in the production of a spliced peptide bearing an extended C-terminus, which is then further trimmed by the proteasome in order to produce the final antigenic peptide (Figure 2) [57]. Recently, Mishto et al. suggested the existence of an additional pocket that would accommodate the nucleophile peptide and would be distinct from the primed substrate binding site, which accommodates the part of the peptide substrate located C-terminally from the cleavage site [75]. They suggested that this additional pocket might already be occupied by the splice reactant prior to the formation of the acyl-enzyme intermediate, thereby facilitating and speeding up the splicing reaction. This suggestion arose from their observation that the hierarchy of splicing at various sites within a given peptide did not strictly follow the hierarchy of cleavage at those sites, suggesting additional constraints determining splicing efficiency, such as the affinity of the nucleophile peptide for this postulated pocket.
Figure 1. Proteasome subtypes and regulators. (A) Mammalian proteasomes are composed of a 20S core particle, made of four stacked rings of seven subunits each. The two outer rings are made of α subunits, and the two inner rings are made of β subunits, three of which (β1, β2, β5) are catalytically active. Upon induction with IFNγ or in immune cells, these catalytic subunits are replaced with their inducible counterparts, β1i, β2i, β5i, to form immunoproteasomes. Besides standard and immunoproteasomes, two additional forms of proteasome exist, which contain a mixture of standard and immune catalytic subunits, as indicated. The thymic cortex expresses a specific catalytic subunit, called β5t, which combines with β1i and β2i to form the thymoproteasome. The percentage of each proteasome type as measured by a sandwich ELISA approach [32] is indicated for different tissue and tumor types. Thymoproteasome is specifically expressed in cortical epithelial cells (cortical thymic epithelial cells (cTEC)). (B) The two α-rings of the 20S proteasome interact with regulatory particles of four different types: PA700 (19S), PA28αβ, PA28γ and PA200. These regulatory particles can bind to one or both sides of the 20S particle or form hybrid proteasomes where the 20S core binds two different regulators. Representative examples are shown. NT, not tested.
Figure 1. Proteasome subtypes and regulators. (A) Mammalian proteasomes are composed of a 20S core particle, made of four stacked rings of seven subunits each. The two outer rings are made of α subunits, and the two inner rings are made of β subunits, three of which (β1, β2, β5) are catalytically active. Upon induction with IFNγ or in immune cells, these catalytic subunits are replaced with their inducible counterparts, β1i, β2i, β5i, to form immunoproteasomes. Besides standard and immunoproteasomes, two additional forms of proteasome exist, which contain a mixture of standard and immune catalytic subunits, as indicated. The thymic cortex expresses a specific catalytic subunit, called β5t, which combines with β1i and β2i to form the thymoproteasome. The percentage of each proteasome type as measured by a sandwich ELISA approach [32] is indicated for different tissue and tumor types. Thymoproteasome is specifically expressed in cortical epithelial cells (cortical thymic epithelial cells (cTEC)). (B) The two α-rings of the 20S proteasome interact with regulatory particles of four different types: PA700 (19S), PA28αβ, PA28γ and PA200. These regulatory particles can bind to one or both sides of the 20S particle or form hybrid proteasomes where the 20S core binds two different regulators. Representative examples are shown. NT, not tested.
Biomolecules 04 00994 g001
Figure 2. Peptide splicing and trimming of the antigenic peptide RSYVPLAH_R derived from gp100 by the proteasome. In the course of peptide-bond hydrolysis, the hydroxyl group of the N-terminal threonine produces a nucleophile attack on the carbonyl of the peptide bond. This leads to the formation of an acyl-enzyme intermediate in which the peptide fragment RSYVPLAH remains attached to the proteasome through an ester link. Normally, this acyl-enzyme intermediate is rapidly hydrolyzed. However, peptide fragments, such as RGS, present in the chamber can compete with water molecules, their free N-terminal amino-group performing a nucleophilic attack on the acyl-enzyme intermediate. This transpeptidation leads to the creation of a new peptide bond, which assembles both fragments, leading to the formation of a spliced peptide bearing an extended C-terminus. The C-terminal extension is then further trimmed by the proteasome to release the antigenic peptide, RSYVPLAHR. Balls represent the catalytic β subunits of the proteasome. The hydroxyl group of the N-terminal threonine is indicated.
Figure 2. Peptide splicing and trimming of the antigenic peptide RSYVPLAH_R derived from gp100 by the proteasome. In the course of peptide-bond hydrolysis, the hydroxyl group of the N-terminal threonine produces a nucleophile attack on the carbonyl of the peptide bond. This leads to the formation of an acyl-enzyme intermediate in which the peptide fragment RSYVPLAH remains attached to the proteasome through an ester link. Normally, this acyl-enzyme intermediate is rapidly hydrolyzed. However, peptide fragments, such as RGS, present in the chamber can compete with water molecules, their free N-terminal amino-group performing a nucleophilic attack on the acyl-enzyme intermediate. This transpeptidation leads to the creation of a new peptide bond, which assembles both fragments, leading to the formation of a spliced peptide bearing an extended C-terminus. The C-terminal extension is then further trimmed by the proteasome to release the antigenic peptide, RSYVPLAHR. Balls represent the catalytic β subunits of the proteasome. The hydroxyl group of the N-terminal threonine is indicated.
Biomolecules 04 00994 g002
So far, the identification of spliced antigenic peptides has been limited by the availability of specific CTL directed against these peptides. Using a specific computer-based algorithm applied to the mass spectrometry analysis of peptide digests, Liepe at al. developed a novel approach (SpliceMet) to predict spliced antigenic peptides [76]. One could verify the relevance of these peptides by isolating CTL in vitro using the reverse immunology approach [77]. The presence of the predicted peptide at the surface of target cells would then be confirmed by analyzing the ability of these cells to activate the corresponding CTL.
In essence, the proteolytic activity of the proteasome is not specific: any protein or peptide entering the proteasome can be degraded. It is therefore essential for cellular homeostasis to tightly control the activity of such an abundant intracellular protease, so as to prevent undesired degradation of intracellular proteins. The proteasome therefore evolved as a compartmentalized particle delineating a chamber that is not accessible to surrounding proteins and that contains the catalytically active sites [78]. This enables the control of proteasome activity at the level of the entry of substrates in the catalytic chamber. Indeed, regulators of proteasome activity, such as the 19S regulator, function as gate-keepers, allowing entry into the proteasome particle of only those substrates that need to be degraded and preventing entry of others. Entrance to the chamber is first limited by an aperture consisting of a 13-Å pore delimited by the ring formed by the α subunits, as observed in the Thermoplasma proteasome [69]. The small size of this α-annulus ensures that only unfolded proteins can enter the catalytic chamber. Additionally, the entrance of the chamber is further obstructed in yeast and bovine proteasome by the N-terminal tails of the α subunits that form a tight polypeptide net, which is projected inside the pore and prevents access of smaller substrates to the catalytic chamber [70,79]. Free 20S proteasome therefore has a low activity, at least toward folded protein substrates [2]. Opening of the α-gate is tightly regulated through the binding of specific activator complexes, called regulatory particles, which are believed to interact with the N-terminal tails of the α-subunits, inducing their displacement and the opening of the gate [2]. Four types of regulators exist: the 19S regulator (PA700), the 11S regulators, PA28αβ and PA28γ, and the PA200 regulator, whose functions will be discussed below.
As an exception to this rule of access-control by regulators, it has been shown that unfolded protein substrates and oxidant-damaged proteins can be degraded by free 20S proteasomes [80,81]. A large excess of enzymatically-active, free 20S proteasome over 26S proteasome is indeed found within cells [82,83]. A recent study from Baugh et al. suggested that about 20% of total cellular proteins can be degraded by the 20S proteasome [84]. However, how unstructured proteins or oxidant-damaged proteins enter the catalytic core of the 20S chamber is still not well understood. Various studies suggested that access to the catalytic chamber could be controlled by the substrate itself or facilitated by accessory proteins, such as Hsp90 [85] or arginine-rich histones [86]. It has been suggested that hydrophobic patches that are often exposed after protein oxidation might facilitate the entry of oxidized proteins into the catalytic chamber [80], in line with the fact that hydrophobic peptides appear to promote the opening of the α-ring of the 20S proteasome [87]. Conditions of stress, such as oxidative damage or prolonged starvation, induce 26S proteasomes to dissociate into 20S proteasomes and 19S sub-complexes [88,89,90]. This leads to a rapid decrease in proteolysis and an accumulation of ubiquitinated proteins, but could favor the degradation of poorly-structured or oxidized proteins by 20S proteasomes [88,91]. Reports have suggested that PA28αβ could also assist the 20S proteasome in the degradation of oxidized proteins [81,92].

4. Regulation of the Proteasome Activity

The first level of regulation is achieved by changes in the structure of the 20S proteasome, which exists in several isoforms or subtypes, having different catalytic activities.

4.1. Proteasome Subtypes

4.1.1. Standard Proteasome and Immunoproteasome

In immune cells or after exposure to the inflammatory cytokines, IFN-γ or TNF-α, alternative catalytic subunits, named LMP2 (β1i), MECL-1 (β2i) and LMP7 (β5i), are expressed and incorporated into the proteasome in place of their constitutive counterparts, β1, β2 and β5, to form another particle called the immunoproteasome (Figure 1A) [93]. Based on the degradation of small fluorogenic peptides, three major catalytic activities of the proteasome were described, the caspase-like, trypsin-like and chymotrypsin-like activities, which cleave after acidic, basic and hydrophobic residues, respectively. The study of yeast proteasome mutants suggested that each of these catalytic activities was associated with one specific catalytic subunit. The caspase-like activity is linked to β1, while the trypsin-like and chymotrypsin-like activities are associated with β2 and β5, respectively [72,94,95]. However, it is now clear that the catalytic specificity is more complex than originally expected, since some proteasome subunits have overlapping specificities [95], and the position of the cleavage site can also be influenced by the surrounding residues [96,97]. Nevertheless, the immunoproteasome activity, as measured with fluorogenic peptides, is characterized by a lower caspase-like activity and higher trypsin-like and chymotrypsin-like activities [98,99]. Due to these differences in their catalytic sites, the standard proteasome and the immunoproteasome produce distinct peptide sets, which only partly overlap [97,98,100,101]. Because the immunoproteasome shows an increased propensity to cleave after basic or hydrophobic residues, it was predicted to be more efficient at producing antigenic peptides with high affinity for MHC class I molecules, which often call for peptides bearing basic or hydrophobic residues at their C-termini [98,100]. This idea was corroborated by the study of knockout mice for either β1i or β5i, in which the presentation of several epitopes was decreased [102,103]. This was recently confirmed using knockout mice for all three immunosubunits, whose dendritic cells fail to present a number of MHC class I epitopes and whose splenocytes display a peptide repertoire that is about 50% different from that of wild-type cells [104]. In the same line, the peptide repertoire of dendritic cells from β2i−/−β5i−/− mice is less abundant and diverse than that of wild-type mice [105]. Overall, these results show that the immunoproteasome plays an important role in MHC class I antigen presentation. Several human tumor antigens are also better produced by the immunoproteasome than by the standard proteasome (Table 1) [31,32,38,61,65]. However, it is also clear that a number of antigenic peptides are better produced by the standard proteasome. The first example of such a peptide was described by Morel et al. who studied a human CTL clone recognizing a peptide derived from ubiquitous protein RU1. This peptide was presented by kidney cancer cells, but not by the autologous EBV-B cells [20]. This was explained by the fact that the peptide was processed efficiently by the standard proteasome, which was present in kidney cancer cells, but not by the immunoproteasome, which was abundant in the EBV-transformed B-cells. Other peptides that are efficiently processed by the standard proteasome, but not by the immunoproteasome, were subsequently identified (Table 1) [31,106]. In vitro digestions of peptide precursors with purified proteasomes indicated that the lack of processing by one proteasome type generally resulted from a prominent cleavage within the antigenic peptide, resulting in its destruction [31,32,38]. Similar observations were made when Basler et al. studied the immunoproteasome-dependent generation of the murine minor histocompatibility antigen UTY246–254 and the human influenza matrix peptide58–66 [107]. The authors showed that inhibition of the β1 or the β5 subunit, respectively, rescued the presentation of these peptides, suggesting that the standard subunits were responsible for their degradation.
In the past few years, we and others identified several peptides produced by peptide splicing in the proteasome. As described above, the peptide splicing reaction takes place in the proteasome trough a reaction of transpeptidation involving the nucleophilic attack of an acyl-enzyme intermediate by another peptide fragment present in the chamber [22]. We found that three spliced peptides were better produced by the standard proteasome than by the immunoproteasome, while another was more efficiently produced by the immunoproteasome [56]. This demonstrated that both proteasome types can perform the splicing reaction. However, we observed that the production of a given spliced peptide depended on the ability of the proteasome to perform the cleavages required to liberate the fragments that composed the spliced peptide. Additionally, this ability differed among the two proteasome subtypes. Of note, even though it was initially identified in tumor cells, peptide splicing is not restricted to tumor cells, but can also occur in non-tumor cells, such as melanocytes [59]. In vitro, it was produced using proteasomes originating from human erythrocytes, but also from yeast cells [22,75]. It is therefore expected that peptide splicing also occurs in the thymus, where spliced peptides should be expressed like any other self-peptides to establish natural immune tolerance, on the condition that the type of proteasome (standard or immunoproteasome) present in medullary thymic epithelial cells is adequate for the production of these peptides.
The induction of T-cell immune responses is critically dependent on dendritic cells, which are professional antigen-presenting cells located mostly in lymph nodes. These cells uniquely express the set of cytokines and costimulatory molecules required to activate naive T-cells in an antigen-specific manner. This means that CTL responses will be induced only against antigenic peptides presented by dendritic cells. Dendritic cells contain immunoproteasomes and intermediate proteasomes (see below), but very little standard proteasome (Figure 1A) [32]. Therefore, they will mostly induce CTL responses against peptides produced by the immunoproteasome (or intermediate proteasomes), but not against peptides produced by the standard proteasome. Regarding anti-viral responses, this is not an issue, because viral infections usually trigger inflammation, which induces immunoproteasomes in infected cells, which therefore will present the adequate antigenic peptides. Indeed, immunoproteasome-dependent peptides were shown to be the dominant targets of anti-viral CTL responses [108]. However, in the case of anti-tumor immune responses, the situation is different, because tumor cells usually do not contain immunoproteasomes, unless they are exposed to inflammation, and therefore, they may not present the peptides against which the immune response was triggered by dendritic cells [106]. Moreover, as indicated above, a number of clinically relevant tumor antigens are produced by the standard proteasome, but not by the immunoproteasome, and therefore are not presented efficiently by dendritic cells [20,31]. Vaccination strategies aimed at inducing CTL against such antigens should therefore be adapted in order to by-pass processing by the proteasome of dendritic cells. This can be done by immunizing with short “pre-processed” synthetic peptides or with recombinant minigene constructs encoding only the short peptide. This notion was first confirmed by a study comparing the CTL responses induced against the HLA-A2-restricted peptide, melan-A26-35, after vaccination of wild-type and β1i-knock-out HLA-A2 transgenic mice with either the peptide-encoding minigene or the full-length melan-A [109]. Stronger responses were induced in wild-type mice when the immunization was performed using the minigene retroviral construct when compared to the full-length construct, and this difference was reduced in the β1i-knockout mice, which developed stronger responses to the full-length construct than the wild-type mice. This confirmed that the immunoproteasome was responsible for the poor responses in wild-type mice. Another approach to induce CTL responses against standard proteasome-dependent peptides is to engineer dendritic cells so that they no longer contain immunoproteasomes, but only standard proteasomes. Dannull et al. successfully used this approach in humans, using siRNA for the catalytic immunosubunits. They showed that dendritic cells transfected with those siRNA and with RNA encoding full-length tumor antigens induced better anti-tumor CTL responses, not only in vitro, but also in vivo in melanoma patients [110,111].
As mentioned above, the immunodominance hierarchy of viral antigens is linked to their dependency on processing by the immunoproteasome. Indeed, studying CTL responses against various influenza, MCMV (mouse cytomegalovirus) and LCMV (lymphocytic choriomeningitis virus) peptides, several groups observed changes in the responses to these peptides in knockout mice for some of the immunoproteasome subunits [108,112,113,114,115]. Responses to antigenic peptides were generally decreased, but in some cases, responses were improved or remained stable. Interestingly, this ended up modifying the immunodominance hierarchy, with dominant epitopes becoming subdominant, and vice versa [108]. The modification of the responses observed in these different models were generally attributed to a difference in the processing of the antigenic peptide by the proteasome types and/or to an alteration of the T-cell repertoire in immunoproteasome-deficient animals [108,112,114]. This latter point implies that, in immunoproteasome-deficient animals, the negative selection operated in the thymus has led to the deletion of T-cell precursors recognizing those viral epitopes. Negative selection occurs in the thymic medulla, where medullary thymic epithelial cells (mTEC) present MHC class I-restricted peptides derived from a variety of self-proteins. This induces the selective deletion of T-cell precursors reactive to these peptides. The proteasome content of mTEC has not yet been clearly evaluated, but it is thought to mostly correspond to immunoproteasomes. Alteration of the anti-viral repertoire in immunoproteasome subunit-deficient animals therefore suggests that the absence of some immunoproteasome subunits in mTECs enables the expression of novel self-peptides that mimic the viral epitopes and lead to the deletion of the corresponding anti-viral T-cell precursors.
Studying the role of the immunoproteasome in the establishment of a T-cell response to MCMV in wild-type and β5i−/− mice, Hutchinson et al. observed that although acute responses to MCMV mostly targeted immunoproteasome-dependent peptides, memory responses targeted those two epitopes that showed the least dependency on the immunoproteasome [115]. Acute MCMV T-cell responses would therefore be elicited by cells containing some immunoproteasome. However, this response would not be totally efficient at destroying target cells infected by MCMV, as this virus impedes immunoproteasome formation through the action of M27, a protein that induces the proteasomal degradation of STAT2 and thereby inhibits the IFNγ response pathway [116]. Chronic responses to MCMV would then involve antigen-presenting cells containing only standard proteasomes and stimulating CTL against those epitopes that are the least dependent on the immunoproteasome.

4.1.2. Intermediate Proteasomes

Using a unique set of antibodies directed against proteasome catalytic subunits, our group additionally described two alternative proteasome forms, which are intermediate between the standard and the immunoproteasome, and contain only one (β5i) or two (β1i, β5i) of the three catalytic subunits of the immunoproteasome (immunosubunits) (Figure 1A) [32]. The existence of intermediate proteasome forms had previously been inferred from the observation that a number of tissues contained only some of the three immunosubunits [117]. It is only the development of immunosubunit-specific antibodies able to capture proteasomes in their native form that allowed us to define the exact stoichiometry of the intermediate proteasomes that are found in human tissues [32]. The existence of only two forms of intermediate proteasomes, namely β5i and β1i–β5i, is in agreement with the previous works dissecting the general rules for proteasome cooperative assembly [93,118], which is dependent on the nature of the subunit propeptides [119,120]. This cooperative assembly ensures a preferential incorporation of immunosubunits over standard subunits, where, e.g., incorporation of β2i is dependent on that of β1i [93]. This may explain why we did not detect the presence of intermediate proteasomes containing β2i. In addition, catalytic subunits are initially incorporated in an immature form, containing an N-terminal peptide that needs to be cleaved to liberate the N-terminal threonine of the mature active site [121]. This maturation step occurs at the last stage of proteasome assembly, when two hemiproteasomes join together to form the final particle. Again, this ensures protection of the cellular proteins, as catalytic sites only become active when the final particle is assembled and confines the proteolytic activity to the inner chamber. Maturation of the immunosubunits is interdependent: β5i was shown to be required for the maturation of β1i and β2i [93]. This may explain why we did not detect intermediate proteasomes lacking β5i. Because the proteasome particle contains two β rings, the existence of proteasomes containing an asymmetric assortment of immunosubunits and standard subunits is conceivable. Such asymmetric proteasomes were observed in cells transfected with a tagged β1 and exposed to IFNγ [122], but it remains unclear whether they exist in unmanipulated cells or tissues. The intermediate proteasomes we described were symmetric. We searched for asymmetric proteasomes containing β5 on one β ring and β5i on the other, and we did not find any in human liver and melanoma [32].
Interestingly, intermediate proteasomes β5i and β1i–β5i represent 10% to 20% of the total proteasomes found in tumors and 30% to 50% of those found in liver, kidney, small bowel, colon and dendritic cells [32] (Figure 1A). Studying the specific cleavage properties of each proteasome type, we showed that intermediate proteasomes β5i and β1i–β5i both display trypsin and chymotrypsin-like activities that are intermediate between those of the standard and the immunoproteasome. The caspase-like activity of intermediate proteasome β5i is similar to that of the standard proteasome, while this activity is low in intermediate proteasome β1i–β5i. This is easily explained by the fact that the caspase-like activity is generally associated with the β1 subunit, which is present in the β5i intermediate proteasome and not in the β1i–β5i intermediate proteasome [32]. Owing to their specific cleavage properties, intermediate proteasomes produce a unique set of antigenic peptides. The HLA-A2-restricted peptides, MAGE-A10254–262 and MAGE-C2191–200, are exclusively produced by the intermediate proteasome, β1i–β5i, while the HLA-A2-restricted peptide, MAGE-A3271–279, is processed only by intermediate proteasome β5i (Table 1) [32,38]. Hence, intermediate proteasomes enlarge the MHC class I repertoire to peptides that can be processed by both dendritic cells (containing immunoproteasomes and intermediate proteasomes) and tumor cells (containing standard proteasomes and intermediate proteasomes) and might therefore represent valuable targets for cancer immunotherapy. Of note, the presence of intermediate proteasomes should also be taken into account when analyzing the role of the immunoproteasome in single-immunosubunit KO mice.

4.1.3. Thymoproteasome

A few years ago, Murata et al. discovered the existence of an additional proteasome catalytic subunit, called β5t, which is specifically expressed in the cortical thymic epithelial cells (cTEC) and is homologous to the β5 and β5i subunits [123,124]. The cTEC are responsible for positive selection, a process by which immature T-cell precursors able to bind to self-MHC class I/peptide complexes on cTEC are triggered to survive. The β5t subunit has a dramatic effect on the establishment of the CTL repertoire, as β5t-deficient mice display an 80% reduction of splenic CTL and do not survive influenza virus infection [123,125]. The β5t subunit is incorporated in the 20S proteasome instead of β5i along with β1i and β2i (Figure 1A) [123]. Incorporation of β5t into HEK-293T proteasomes induced a dramatic decrease of the chymotrypsin-like activity when compared to the β5-containing proteasome, while the trypsin-like and caspase-like activities were not affected [123]. This is in line with the fact that the S1 pocket of β5t is lined by hydrophilic amino acids, while that of β5 or β5i contains hydrophobic residues [123]. Using a unique set of antibodies recognizing peptide-bound or empty MHC class I molecules, Nitta et al. showed that cTECs from both wild-type or β5t−/− mice express MHC class I molecules associated with peptides. Some of these peptides appeared unique to cTECs, as two specific peptide populations bound to H-2Kb were found to be differentially expressed in cTECs when compared to other APCs [125], suggesting that cTECs display a qualitatively distinct repertoire of MHC class I/peptide complexes. It was originally proposed that the low chymotrypsin-like activity of the thymoproteasome might produce low-affinity MHC class I ligands that may limit the duration or the avidity of the interaction of the peptide/MHC complexes with the TCR and facilitate positive selection [123,126]. However, recent work by Xing et al. showed that the half-life of MHC/peptide complexes on the surface cTEC and mTEC was similar, disagreeing with the idea that cTEC would carry unstable MHC complexes responsible for positive selection [127]. Moreover, similar surface MHC class I levels are found in β5t+/− and β5t−/− cTECs [123]. To reconcile these observations, it was proposed that cTECs carried peptides binding MHC class I using alternative anchor positions or developed a mechanism enabling them to stabilize MHC class I molecules bearing low affinity peptides [128]. In the future, a more detailed analysis of the peptide repertoire found on cTECs might help to understand the role played by the thymoproteasome in the formation of an immunocompetent T-cell repertoire.

4.1.4. Proteasome Subtypes and Negative Selection of T-Cell Precursors in the Thymus

It is interesting to note that, so far, most tumor antigenic peptides found to be poorly processed by the immunoproteasome derive from self-proteins expressed in normal adult tissues (Table 1). Because the thymus contains abundant amounts of immunoproteasome, this precludes the processing of these peptides by the medullary thymic epithelial cells (mTEC), resulting in a lack of negative selection of T-cells recognizing these antigens and explaining why these T-cells persist in the periphery. In contrast, tumor antigens, which are better produced by the immunoproteasome, often correspond to MAGE-type antigens encoded by cancer-germline genes or to mutated antigens [31,32,66]. The limited immune tolerance against MAGE-type antigens has been attributed to the very low expression of cancer-germline genes in mTEC, resulting in the negative selection of only those T-cells recognizing these antigens with a high avidity [129]. The anti-tumor T-lymphocytes present in the blood of cancer patients would then derive from low-avidity T-cells that escape negative selection in the thymus. To confirm this concept, our group recently produced knockout mice for a mouse cancer-germline gene, namely P1A [130]. The expectation was that these mice, which are devoid of P1A expression in the thymus, should contain an unselected anti-P1A T-cell repertoire still containing high-affinity T-cells. Surprisingly, although the frequency of anti-P1A T-cells was slightly higher in P1A-KO mice, the anti-P1A T-cell repertoire of P1A-KO mice was barely different from that of wild-type mice, indicating that there is only minimal tolerance against this antigen in wild-type mice, despite detectable expression of P1A in mTEC [130]. Further analysis of the processing of the P1A antigen and of the type of proteasome contained in mTEC might help understanding the reason for the lack of tolerance against antigens encoded by cancer-germline genes.

4.1.5. Structure-Related Properties

The crystal structures of the mouse standard proteasome and the immunoproteasome were solved recently and revealed that the cleavage preferences of the immunoproteasome relied on subtle conformational changes in the substrate-binding pockets of catalytic subunits β1i and β5i [131]. Indeed, the substrate-binding channel of β1i is lined with hydrophobic amino acids, resulting in an increased hydrophobicity and a decreased size of the binding pocket, thereby accounting for the higher cleavage activity of β1i after small hydrophobic and branched residues [131]. Compared to β5, β5i harbors a more spacious S1 pocket, favoring access to large non-polar residues and explaining the higher chymotrypsin-like activity of β5i [131]. Additionally, the vicinity of the active site of β5i is more hydrophilic: this is predicted to favor proteolysis by attracting water molecules, but also by stabilizing the tetrahedral intermediate through additional hydrogen bonds with the oxyanion hole [131].
Studying the processing of the HLA-A2-restricted peptide, MAGE-A10254–262, we observed that this peptide could only be processed by the intermediate proteasome β1i–β5i and that the inability of the other proteasomes to produce the peptide was related to the occurrence of a destructive cleavage within the peptide after aspartate257 (D257) (Table 1) [32]. The caspase-like activity, which is defined by cleavage after acidic residues, is generally associated with the presence of the β1 subunit, and digestion of fluorogenic peptides showed that the intermediate proteasome β1i–β5i and the immunoproteasome display a much lower caspase-like activity than the standard proteasome. However, in the case of the MAGE-A10254–262 peptide, the immunoproteasome still performed the destructive cleavage after D257, despite the presence of the β1i subunit. Therefore, the ability of the intermediate proteasome β1i–β5i, as opposed to the immunoproteasome, to produce the antigenic peptide suggests that β2i negatively influences the production of this peptide, either because β2i itself displays sufficient caspase-like activity to destroy the peptide or because it affects the structure of the immunoproteasome and increases its ability to cleave after D257. Such an allosteric effect of one subunit on the activity of the others was previously suggested by Sijts et al. who showed that subunit β5i, by impacting the structure of the 20S proteasome, increases the activity of subunits β2i and β1i [132].

4.1.6. Other Functions of the Immunoproteasome

It is quite likely that the function of the different proteasome types is not restricted to antigen processing. The work of Seifert et al., for example, suggested a role for the immunoproteasome in the degradation of polyubiquitinated, oxidant-damaged proteins accumulating after IFNγ treatment [133]. However, the prominent role of the immunoproteasome in the degradation of polyubiquitinated proteins was not confirmed by others [134]. Another role for the immunoproteasome was suggested by the observation that the absence of β1i in NOD mice impaired the processing of the NF-κB p105–p50 [135]. However, this study was refuted early after publication, and the function of the immunoproteasome in the NF-κB signaling pathway has remained largely debated since then [136,137,138,139,140]. Interestingly, the use of the β5i-specific inhibitor, PR-957, affected the production of inflammatory cytokines IL-23, TNF-α and IL-6 by endotoxin-stimulated peripheral blood mononuclear cells in an NF-κB-independent manner [141]. The inhibitor also affected the production of IFNγ and IL-2 in stimulated T-cells and helped with controlling the pathogenic response in a mouse model of rheumatoid arthritis [141].

4.2. Proteasome Regulators

As mentioned above, a key step in the control of proteasome activity is exerted at the level of substrate entry into the catalytic chamber of the particle and depends on the presence of regulators.

4.2.1. The 19S Regulator

The 19S regulator (PA700) is a large particle of about 700 kDa that associates with one or both sides of the 20S proteasome core to enable the proteasome to bind, deubiquitylate, unfold and translocate the protein substrate into the catalytic chamber. The 19S regulator specifically recognizes proteins targeted for degradation by the covalent attachment of ubiquitin molecules (74 amino acids in length) to lysine residues within protein substrates. The ubiquitination requires the sequential action of an ubiquitin-activating enzyme, E1, an ubiquitin-conjugating enzyme, E2, and an ubiquitin-ligase enzyme, E3 (reviewed in [142]). Additional ubiquitin molecules can then be ligated to a previously bound ubiquitin to form polyubiquitin chains. Chains of four or more ubiquitins are sufficient to target proteins for degradation.
The 19S regulator is composed of at least 19 subunits that are assembled in two different parts: the lid and the base (reviewed in [2]). In yeast, the base comprises six ATPases (Rpt1-6), the two large non-ATPase subunits, Rpn1 and Rpn2, and two ubiquitin receptor subunits, Rpn10 and Rpn13. The six AAA-ATPase subunits, Rpt1-6, form a heterohexameric ring that directly contacts the α ring of the core 20S particle and is responsible for the ATP-dependent unfolding and translocation of the substrate into the chamber. The Rpn10 and Rpn13 subunits directly bind the polyubiquitin chains of protein substrates. The lid comprises nine Rpn subunits (Rpn3, 5–9, 11, 12 and 15). Subunits Rpn3, 5, 6, 7, 9, 12 have very similar domain structures and comprise an N-terminal helix repeat segment, a proteasome-COP9/signalosome-eIF3 (PCI) domain and a long C-terminal helix [143]. The Rpn8 and 11 subunits contain an Mpr1-Pad1-N-terminal (MPN) domain, and Rpn11 bears a deubiquitylation activity (DUB) [144,145]. The role of the other PCI and MPN subunits has not yet been characterized, except for Rpn6, whose crystal structure suggested a critical role in stabilizing the interaction between the 20S core particle and the regulatory complex [146]. The 19S regulator can associate with one or both ends of the 20S proteasome particle, but can also be part of a hybrid proteasome composed of one 19S regulator and one PA28 or PA200 regulator bound to either end of the 20S proteasome complex (Figure 1B) [147,148].

4.2.2. The PA28αβ Regulator

The PA28αβ regulator (11S regulator) was originally isolated owing to its ability to stimulate the hydrolysis by the 20S proteasome of small peptide substrates, but not intact, folded proteins [149,150,151]. It is composed of two subunits, α and β, whose transcription is induced following exposure to IFNγ [152,153]. Electron-microscopic studies showed that the PA28αβ subunits assemble to form a ring-shaped heptameric complex with a 3α4β conformation that interacts with the α-ring of the 20S proteasome complex [154,155,156]. The PA28αβ complex binds either to both ends of the 20S proteasome or to only one side of the particle, which binds a 19S regulator on the other side to form the so-called hybrid proteasome (Figure 1B) [147,157,158]. A number of mechanisms have been proposed to explain how PA28αβ increases proteasome activity, including the allosteric activation of the active sites of the proteasome [149] and the stimulation of peptide entry [159] or exit [160,161] from the 20S chamber. Other explanations include the coupling of the proteasome to chaperones that facilitate substrate delivery inside the catalytic chamber or the interaction of PA28-containing proteasomes with ER membranes to facilitate the transfer of the produced peptides into the ER lumen [151,162]. Studying antigen presentation in mice deficient for PA28β, Preckel et al. showed that these mice, which lack both PA28α and PA28β, were impaired in their ability to process antigens [163]. In addition, these PA28-deficient cells displayed a dramatic decrease in immunoproteasome content, which, together with the fact that PA28 was found to be associated with immunosubunit-containing hemiproteasomes, led the authors to conclude that PA28αβ was necessary for immunoproteasome assembly [163]. Yet, in subsequent studies using PA28α and PA28β double-knockout mice, Murata et al. observed normal immunoproteasome assembly and normal immunity to influenza infection [164]. In this study, however, the authors observed that PA28αβ−/− cells were unable to process a peptide derived from the murine melanoma differentiation antigen, TRP2 (TRP2181–188). Overall, the authors concluded that PA28 was not required for proteasome assembly nor overall MHC class I presentation, but was, rather, involved in the processing of a subset of antigenic peptides [164]. Later on, several in vitro and in vivo studies highlighted a role for PA28αβ in the processing of other MHC class I peptides (Table 2). In vitro digestions of precursor peptides by 20S proteasomes in the presence or absence of recombinant PA28αβ showed that PA28αβ increases cleavage efficiency and the production of MHC ligands by the proteasome [165,166]. Association of the proteasome with PA28 also favors double cleavages, which was assumed to originate from the retention by PA28 of the peptide substrate inside the proteasome chamber, thereby increasing the probability for a second cleavage to occur [165]. Transfecting PA28αβ into mouse fibroblasts expressing the murine cytomegalovirus antigen pp89, Groettrup et al. observed a marked increase in recognition by the virus-specific cytotoxic T-cells [167]. In this system, presentation of the influenza nucleoprotein was also significantly improved. Later on, the same authors showed that the increase in antigen presentation of the pp89 peptide was independent of the proteasome-subunit composition and therefore, rather, resulted from the general ability of PA28 to activate proteasome complexes without affecting proteasome-subunit composition [168]. Overexpression of PA28αβ also increased the presentation of a number of viral epitopes, while it did not affect the production of others [169,170]. Studying the role of PA28αβ on the processing of the peptide TRP2360–368 derived from the human melanoma differentiation antigen, TRP2 [171], Sun et al. observed that this peptide was not processed in a melanoma cell line lacking PA28, but this processing was rescued after transient transfection of PA28 cDNAs. Interestingly, the replacement of the flanking sequences of a PA28-independent antigenic peptide by those of the PA28-dependent peptide TRP2360–368, revealed that the terminal flanking sequence of the peptide TRP2360–368 was sufficient to confer PA28αβ dependency [172]. The origin of this sequence-driven dependency has not yet been clarified. Lastly, an allele-specific involvement of PA28αβ was observed, as it reduced the processing of Kd and Dd ligands, while favoring the presentation of Kb and Ld peptides [173]. Overall, PA28αβ appears not to be essential for the MHC class I processing pathway, but rather to be involved in the presentation of a selective set of antigenic peptides.
Table 2. The effect on the proteasome regulator PA28αβ on the processing of viral and tumor antigens. LCMV, lymphocytic choriomeningitis virus.
Table 2. The effect on the proteasome regulator PA28αβ on the processing of viral and tumor antigens. LCMV, lymphocytic choriomeningitis virus.
Peptide SourceMHC RestrictionPeptide SequenceDependence on PA28αβReferences
Viral antigens
Murine cytomegalovirus pp89168–176H-2LdYPHFMPTNL+[167,168]
Murine Moloney MuLV Gag-leader75–83H-2DbCCLCLTVFL+[169]
Murine Moloney MuLV gp75-env189–196H-2KbSSWDFITV[169]
LCMV nuceoprotein118–126H-2LdRPQASGVYM[174]
JAK1 tyrosine kinase355–363H-2KdSYFPEITHI+[165]
Influenza A/PR/8 nucleoprotein146–154H-2KdTYQRTRALV+[167]
Murine tumor Antigens
TRP2181–188H-2KbVYDFFVWL+[164]
Human Tumor Antigen
TRP2360–368HLA-A2TLDSQVMSL+[171,172]
TRP2288–296HLA-A2SLDDYNHLV[172]

4.2.3. The PA28γ

PA28γ was first identified as the Ki antigen, target of autoantibodies in the sera of patients with systemic lupus erythematosus [175]. In contrast to PA28αβ, PA28γ is not induced by IFNγ and has a predominant localization in the nucleus [151], where it binds and activates the 20S proteasome as an active homoheptamer [176]. PA28γ-deficient mice display reduced body size and defects in cell proliferation [177,178]. PA28γ-deficient embryonic fibroblasts show defects in mitotic progression and increased apoptosis, suggesting that the regulator is involved in cell cycle progression and apoptosis [177,178]. Accordingly, several binding partners of PA28γ were identified, whose functions are related to apoptosis, such as caspase-7 and MEKK3 [179,180]. Moreover, PA28γ was shown to be involved in the ubiquitin- and ATP-independent degradation of a number of cell-cycle regulatory proteins, such as the cyclin-dependent inhibitor p21, or the steroid receptor coactivator 3 (SRC-3), which is an oncogene often amplified in breast cancer and involved in cell growth [181,182]. Recent studies have also shown that PA28γ is involved in DNA repair and chromosome stability and takes part in the organization of the nuclear speckle and the trafficking of SR proteins (proteins with long serine and arginine repeats, involved in RNA splicing) to the transcription sites [183,184,185]. Interestingly, PA28γ was shown to inhibit the activity of p53, a central transcription factor involved in cell cycle arrest and apoptosis, by inducing p53 nuclear export and promoting its MDM2-dependent ubiquitination and proteasome degradation [186,187]. The role of PA28γ in the stability of p53 was recently shown to originate from the PA28γ-dependent degradation of casein kinase 1 (CK1), which negatively regulates MDM-2 [188]. Additionally, a negative feedback loop between p53 and PA28γ helps with maintaining the balance of p53 and PA28γ in cells [189].
Contrary to PA28αβ, which improves all three catalytic activities of the proteasome; PA28γ was shown to selectively activate the trypsin-like activity without affecting the chymotrypsin-like and the caspase-like activities [190]. However, the role of PA28γ in antigen processing appears relatively insignificant, as PA28γ-deficient mice display normal levels of MHC class I molecules and are not affected in their responses to influenza virus [178]. However, a slower clearance of the intracellular fungal pathogen Histoplasma capsulatum was observed, in parallel with a slight decrease in the number of total CD8+ T-cells in infected animals [178]. The origin of this defect was not identified.

4.2.4. The PA200 Regulator

PA200 is a 200 kDa protein originally identified in rabbit reticulocyte lysate and bovine testis, where it was bound to 20S proteasomes [148,191]. Similarly to the proteasome regulator, PA28αβ, PA200 stimulates the proteasome hydrolysis of small peptides, but not folded proteins, and it does so in an ATP-independent manner [148]. Contrary to PA28αβ, which activates all three proteasome catalytic activities, or to PA28γ, which activates only the trypsin-like activity, PA200 was shown to increase the hydrolysis of peptides bearing an acidic residue in P1 (caspase-like activity) [148]. PA200 is particularly abundant in testis, where it might play an important role in the maturation of spermatozoa and in the progression of spermatogenesis. Interestingly, after gamma irradiation, cellular levels of PA200/19S hybrid proteasomes are increased [192], and the complex relocalizes to intranuclear foci [148], where PA200 accumulates on chromatin. This leads to an increase in proteasome-related caspase-like activity at the chromatin site [192]. Interestingly, PA200 knockdown was found to impair cell survival after gamma-irradiation [148], a phenomenon that could be counteracted by the addition of exogenous glutamine. This suggested that PA200-deficient cells are unable to cope with the increased glutamine demand that arises following irradiation. The reason for this is not yet clear, but it was suggested that the caspase-like activity of the PA200-19S hybrid proteasome might help with restoring the intracellular levels of glutamine or glutamate necessary for the long-term survival of tumor cells following radiation exposure [192]. So far, no involvement of PA200 in the production of antigenic peptides has been reported.

5. Conclusions

The proteasome is a major regulator of protein homeostasis in cells. Through its function in protein degradation, the proteasome enables the cell to adapt to the various assaults and changes occurring in its environment. For each situation, a specific proteasome regulator is involved, granting access to protein substrates into the chamber. Proteasome activity is also regulated by its composition in catalytic subunits, which modulates the cleavage specificities of the particle. As a result of its protease activity, the proteasome releases small peptides that can be loaded onto MHC class I molecules and transported to the cell surface for recognition by the immune system. Proteasome regulation is an important parameter determining the type of peptides presented at the cell surface and thereby affecting the ability of the immune system to respond to the presence of viruses and tumor cells. This occurs at various levels including the establishment of an immunocompetent T-cell repertoire devoid of autoimmune reactivity, the activation of naive T-cells into active effectors cells and the recognition and destruction of tumor or virus-infected cells. Altogether, understanding the mechanisms involved in the production of antigenic peptides by the proteasome is of crucial importance in order to adapt the mode of delivery of immunotherapeutic vaccines and to define which peptides are the most likely to improve the clinical responses to viruses and cancer.

Acknowledgments

We are grateful to Julie Klein for her valuable help in the preparation of this manuscript. Nathalie Vigneron is supported by a grant from the Belgium Programme on Interuniversity Poles of Attraction initiated by the Belgian State, prime Minister’s Office, Science Policy Programming.

Conflicts of Interest

The authors declare no conflict of interest.

References

  1. Rock, K.L.; Goldberg, A.L. Degradation of cell proteins and the generation of MHC class I-presented peptides. Annu. Rev. Immunol. 1999, 17, 739–779. [Google Scholar] [CrossRef] [PubMed]
  2. Tomko, R.J., Jr.; Hochstrasser, M. Molecular architecture and assembly of the eukaryotic proteasome. Annu. Rev. Biochem. 2013, 82, 415–445. [Google Scholar] [CrossRef] [PubMed]
  3. Kisselev, A.F.; Akopian, T.N.; Woo, K.M.; Goldberg, A.L. The sizes of peptides generated from protein by mammalian 26 and 20S proteasomes. J. Biol. Chem. 1999, 274, 3363–3371. [Google Scholar] [CrossRef] [PubMed]
  4. Reits, E.; Griekspoor, A.; Neijssen, J.; Groothuis, T.; Jalink, K.; van Veelen, P.; Janssen, H.; Calafat, J.; Drijfhout, J.W.; Neefjes, J. Peptide diffusion, protection, and degradation in nuclear and cytoplasmic compartments before antigen presentation by MHC class I. Immunity 2003, 18, 97–108. [Google Scholar] [CrossRef] [PubMed]
  5. Androlewicz, M.J.; Anderson, K.S.; Cresswell, P. Evidence that transporters associated with antigen processing translocate a major histocompatibility complex class I-binding peptide into the endoplasmic reticulum in an ATP-dependent manner. Proc. Natl. Acad. Sci. USA 1993, 90, 9130–9134. [Google Scholar] [CrossRef] [PubMed]
  6. Kageyama, S.; Tsomides, T.J.; Sykulev, Y.; Eisen, H.N. Variations in the number of peptide-MHC class I complexes required to activate cytotoxic T-cell responses. J. Immunol. 1995, 154, 567–576. [Google Scholar] [PubMed]
  7. Coulie, P.G.; Van den Eynde, B.J.; van der Bruggen, P.; Boon, T. Tumour antigens recognized by T-lymphocytes: At the core of cancer immunotherapy. Nat. Rev. Cancer 2014, 14, 135–146. [Google Scholar] [CrossRef] [PubMed]
  8. Vigneron, N.; Van den Eynde, B.J. Insights into the processing of MHC class I ligands gained from the study of human tumor epitopes. Cell. Mol. Life Sci. 2011, 68, 1503–1520. [Google Scholar] [CrossRef] [PubMed]
  9. Wölfel, T.; Hauer, M.; Schneider, J.; Serrano, M.; Wölfel, C.; Klehmann-Hieb, E.; de Plaen, E.; Hankeln, T.; Meyer zum Büschenfelde, K.-H.; Beach, D. A p16INK4a-insensitive CDK4 mutant targeted by cytolytic T-lymphocytes in a human melanoma. Science 1995, 269, 1281–1284. [Google Scholar] [CrossRef] [PubMed]
  10. Vigneron, N.; Ooms, A.; Morel, S.; Degiovanni, G.; Van den Eynde, B. Identification of a new peptide recognized by autologous cytolytic T-lymphocytes on a human melanoma. Cancer Immun. 2002, 2, 9–18. [Google Scholar] [PubMed]
  11. Coulie, P.G.; Brichard, V.; van Pel, A.; Wölfel, T.; Schneider, J.; Traversari, C.; Mattei, S.; de Plaen, E.; Lurquin, C.; Szikora, J.-P.; et al. A new gene coding for a differentiation antigen recognized by autologous cytolytic T-lymphocytes on HLA-A2 melanomas. J. Exp. Med. 1994, 180, 35–42. [Google Scholar] [CrossRef] [PubMed]
  12. Kawakami, Y.; Eliyahu, S.; Jennings, C.; Sakaguchi, K.; Kang, X.; Southwood, S.; Robbins, P.F.; Sette, A.; Appella, E.; Rosenberg, S.A. Recognition of multiple epitopes in the human melanoma antigen gp100 by tumor-infiltrating T-lymphocytes associated with in vivo tumor regression. J. Immunol. 1995, 154, 3961–3968. [Google Scholar] [PubMed]
  13. Van der Bruggen, P.; Traversari, C.; Chomez, P.; Lurquin, C.; de Plaen, E.; Van den Eynde, B.; Knuth, A.; Boon, T. A gene encoding an antigen recognized by cytolytic T-lymphocytes on a human melanoma. Science 1991, 254, 1643–1647. [Google Scholar]
  14. Michalek, M.T.; Grant, E.P.; Gramm, C.; Goldberg, A.L.; Rock, K.L. A role for the ubiquitin-dependent proteolytic pathway in MHC class I-restricted antigen presentation. Nature 1993, 363, 552–554. [Google Scholar] [CrossRef] [PubMed]
  15. Ben-Shahar, S.; Cassouto, B.; Novak, L.; Porgador, A.; Reiss, Y. Production of a specific major histocompatibility complex class I-restricted epitope by ubiquitin-dependent degradation of modified ovalbumin in lymphocyte lysate. J. Biol. Chem. 1997, 272, 21060–21066. [Google Scholar] [CrossRef] [PubMed]
  16. Grant, E.P.; Michalek, M.T.; Goldberg, A.L.; Rock, K.L. Rate of antigen degradation by the ubiquitin-proteasome pathway influences MHC class I presentation. J. Immunol. 1995, 155, 3750–3758. [Google Scholar] [PubMed]
  17. Townsend, A.; Bastin, J.; Gould, K.; Brownlee, G.; Andrew, M.; Coupar, B.; Boyle, D.; Chan, S.; Smith, G. Defective presentation to class I-restricted cytotoxic T-lymphocytes in vaccinia-infected cells is overcome by enhanced degradation of antigen. J. Exp. Med. 1988, 168, 1211–1224. [Google Scholar] [CrossRef] [PubMed]
  18. Kwon, Y.T.; Reiss, Y.; Fried, V.A.; Hershko, A.; Yoon, J.K.; Gonda, D.K.; Sangan, P.; Copeland, N.G.; Jenkins, N.A.; Varshavsky, A. The mouse and human genes encoding the recognition component of the N-end rule pathway. Proc. Natl. Acad. Sci. USA 1998, 95, 7898–7903. [Google Scholar] [CrossRef] [PubMed]
  19. Rock, K.L.; Gramm, C.; Rothstein, L.; Clark, K.; Stein, R.; Dick, L.; Hwang, D.; Goldberg, A.L. Inhibitors of the proteasome block the degradation of most cell proteins and the generation of peptides presented on MHC class I molecules. Cell 1994, 78, 761–771. [Google Scholar] [CrossRef] [PubMed]
  20. Morel, S.; Lévy, F.; Burlet-Schiltz, O.; Brasseur, F.; Probst-Kepper, M.; Peitrequin, A.-L.; Monsarrat, B.; van Velthoven, R.; Cerottini, J.-C.; Boon, T.; et al. Processing of some antigens by the standard proteasome but not by the immunoproteasome results in poor presentation by dendritic cells. Immunity 2000, 12, 107–117. [Google Scholar] [CrossRef] [PubMed]
  21. Ottaviani, S.; Zhang, Y.; Boon, T.; van der Bruggen, P. A MAGE-1 antigenic peptide recognized by human cytolytic T-lymphocytes on HLA-A2 tumor cells. Cancer Immunol. Immunother. 2005, 54, 1214–1220. [Google Scholar] [CrossRef] [PubMed]
  22. Vigneron, N.; Stroobant, V.; Chapiro, J.; Ooms, A.; Degiovanni, G.; Morel, S.; van der Bruggen, P.; Boon, T.; Van den Eynde, B. An antigenic peptide produced by peptide splicing in the proteasome. Science 2004, 304, 587–590. [Google Scholar] [CrossRef] [PubMed]
  23. Cerundolo, V.; Benham, A.; Braud, V.; Mukherjee, S.; Gould, K.; Macino, B.; Neefjes, J.; Townsend, A. The proteasome-specific inhibitor lactacystin blocks presentation of cytotoxic T-lymphocyte epitopes in human and murine cells. Eur. J. Immunol. 1997, 27, 336–341. [Google Scholar] [CrossRef] [PubMed]
  24. Harding, C.V.; France, J.; Song, R.; Farah, J.M.; Chatterjee, S.; Iqbal, M.; Siman, R. Novel dipeptide aldehydes are proteasome inhibitors and block the MHC-I antigen-processing pathway. J. Immunol. 1995, 155, 1767–1775. [Google Scholar] [PubMed]
  25. Yang, B.; Hahn, Y.S.; Hahn, C.S.; Braciale, T.J. The requirement for proteasome activity class I major histocompatibility complex antigen presentation is dictated by the length of preprocessed antigen. J. Exp. Med. 1996, 183, 1545–1552. [Google Scholar] [CrossRef] [PubMed]
  26. Niedermann, G.; Butz, S.; Ihlenfeldt, H.G.; Grimm, R.; Lucchiari, M.; Hoschutzky, H.; Jung, G.; Maier, B.; Eichmann, K. Contribution of proteasome-mediated proteolysis to the hierarchy of epitopes presented by major histocompatibility complex class I molecules. Immunity 1995, 2, 289–299. [Google Scholar] [CrossRef]
  27. Vinitsky, A.; Anton, L.C.; Snyder, H.L.; Orlowski, M.; Bennink, J.R.; Yewdell, J.W. The generation of MHC class I-associated peptides is only partially inhibited by proteasome inhibitors-involvement of nonproteasomal cytosolic proteases in antigen processing? J. Immunol. 1997, 159, 554–564. [Google Scholar]
  28. Craiu, A.; Akopian, T.; Goldberg, A.; Rock, K.L. Two distinct proteolytic processes in the generation of a major histocompatibility complex class I-presented peptide. Proc. Natl. Acad. Sci. USA 1997, 94, 10850–10855. [Google Scholar] [CrossRef] [PubMed]
  29. Milner, E.; Gutter-Kapon, L.; Bassani-Strenberg, M.; Barnea, E.; Beer, I.; Admon, A. The effect of proteasome inhibition on the generation of the human leukocyte antigen (HLA) peptidome. Mol. Cell. Proteomics 2013, 12, 1853–1864. [Google Scholar] [CrossRef] [PubMed]
  30. Nawrocki, S.T.; Carew, J.S.; Dunner, K., Jr.; Boise, L.H.; Chiao, P.J.; Huang, P.; Abbruzzese, J.L.; McConkey, D.J. Bortezomib inhibits PKR-like endoplasmic reticulum (ER) kinase and induces apoptosis via ER stress in human pancreatic cancer cells. Cancer. Res. 2005, 65, 11510–11519. [Google Scholar] [CrossRef] [PubMed]
  31. Chapiro, J.; Claverol, S.; Piette, F.; Ma, W.; Stroobant, V.; Guillaume, B.; Gairin, J.-E.; Morel, S.; Burlet-Schiltz, O.; Monsarrat, B.; et al. Destructive cleavage of antigenic peptides either by the immunoproteasome or by the standard proteasome results in differential antigen presentation. J. Immunol. 2006, 176, 1053–1061. [Google Scholar] [CrossRef] [PubMed]
  32. Guillaume, B.; Chapiro, J.; Stroobant, V.; Colau, D.; van Holle, B.; Parvizi, G.; Bousquet-Dubouch, M.P.; Theate, I.; Parmentier, N.; Van den Eynde, B.J. Two abundant proteasome subtypes that uniquely process some antigens presented by HLA class I molecules. Proc. Natl. Acad. Sci. USA 2010, 107, 18599–18604. [Google Scholar] [CrossRef] [PubMed]
  33. Gelman, J.S.; Sironi, J.; Berezniuk, I.; Dasgupta, S.; Castro, L.M.; Gozzo, F.C.; Ferro, E.S.; Fricker, L.D. Alterations of the intracellular peptidome in response to the proteasome inhibitor bortezomib. PLoS One 2013, 8, e53263. [Google Scholar] [CrossRef] [PubMed]
  34. Mo, X.Y.; Cascio, P.; Lemerise, K.; Goldberg, A.L.; Rock, K. Distinct proteolytic processes generate the C and N termini of MHC class I-binding peptides. J. Immunol. 1999, 163, 5851–5859. [Google Scholar] [PubMed]
  35. Reits, E.; Neijssen, J.; Herberts, C.; Benckhuijsen, W.; Janssen, L.; Drijfhout, J.W.; Neefjes, J. A major role for TPPII in trimming proteasomal degradation products for MHC class I antigen presentation. Immunity 2004, 20, 495–506. [Google Scholar] [CrossRef] [PubMed]
  36. Saric, T.; Chang, S.C.; Hattori, A.; York, I.A.; Markant, S.; Rock, K.L.; Tsujimoto, M.; Goldberg, A.L. An IFN-γ-induced aminopeptidase in the ER, ERAP1, trims precursors to MHC class I-presented peptides. Nat. Immunol. 2002, 3, 1169–1176. [Google Scholar] [CrossRef] [PubMed]
  37. Kanaseki, T.; Blanchard, N.; Hammer, G.E.; Gonzalez, F.; Shastri, N. ERAAP synergizes with MHC class I molecules to make the final cut in the antigenic peptide precursors in the endoplasmic reticulum. Immunity 2006, 25, 795–806. [Google Scholar] [CrossRef] [PubMed]
  38. Guillaume, B.; Stroobant, V.; Bousquet-Dubouch, M.P.; Colau, D.; Chapiro, J.; Parmentier, N.; Dalet, A.; Van den Eynde, B.J. Analysis of the processing of seven human tumor antigens by intermediate proteasomes. J. Immunol. 2012, 189, 3538–3547. [Google Scholar] [CrossRef] [PubMed]
  39. Luckey, C.J.; King, G.M.; Marto, J.A.; Venketeswaran, S.; Maier, B.F.; Crotzer, V.L.; Colella, T.A.; Shabanowitz, J.; Hunt, D.F.; Engelhard, V.H. Proteasomes can either generate or destroy MHC class I epitopes: Evidence for nonproteasomal epitope generation in the cytosol. J. Immunol. 1998, 161, 112–121. [Google Scholar] [PubMed]
  40. Seifert, U.; Maranon, C.; Shmueli, A.; Desoutter, J.F.; Wesoloski, L.; Janek, K.; Henklein, P.; Diescher, S.; Andrieu, M.; de la Salle, H.; et al. An essential role for tripeptidyl peptidase in the generation of an MHC class I epitope. Nat. Immunol. 2003, 4, 375–379. [Google Scholar] [CrossRef] [PubMed]
  41. Guil, S.; Rodriguez-Castro, M.; Aguilar, F.; Villasevil, E.M.; Anton, L.C.; del Val, M. Need for tripeptidyl-peptidase II in major histocompatibility complex class I viral antigen processing when proteasomes are detrimental. J. Biol. Chem. 2006, 281, 39925–39934. [Google Scholar] [CrossRef] [PubMed]
  42. Diekmann, J.; Adamopoulou, E.; Beck, O.; Rauser, G.; Lurati, S.; Tenzer, S.; Einsele, H.; Rammensee, H.G.; Schild, H.; Topp, M.S. Processing of two latent membrane protein 1 MHC class I epitopes requires tripeptidyl peptidase II involvement. J. Immunol. 2009, 183, 1587–1597. [Google Scholar] [CrossRef] [PubMed]
  43. Parmentier, N.; Stroobant, V.; Colau, D.; de Diesbach, P.; Morel, S.; Chapiro, J.; Van Endert, P.; Van den Eynde, B.J. Production of an antigenic peptide by insulin-degrading enzyme. Nat. Immunol. 2010, 11, 449–454. [Google Scholar] [CrossRef] [PubMed]
  44. Kessler, J.H.; Khan, S.; Seifert, U.; le Gall, S.; Chow, K.M.; Paschen, A.; Bres-Vloemans, S.A.; de Ru, A.; van Montfoort, N.; Franken, K.L.; et al. Antigen processing by nardilysin and thimet oligopeptidase generates cytotoxic T-cell epitopes. Nat. Immunol. 2011, 12, 45–53. [Google Scholar] [CrossRef] [PubMed]
  45. Kessler, B.; Hong, X.; Petrovic, J.; Borodovsky, A.; Dantuma, N.P.; Bogyo, M.; Overkleeft, H.S.; Ploegh, H.; Glas, R. Pathways accessory to proteasomal proteolysis are less efficient in major histocompatibility complex class I antigen production. J. Biol. Chem. 2003, 278, 10013–10021. [Google Scholar] [CrossRef] [PubMed]
  46. Van Endert, P. Role of tripeptidyl peptidase II in MHC class I antigen processing—The end of controversies? Eur. J. Immunol. 2008; 38, 609–613. [Google Scholar]
  47. Culina, S.; Mauvais, F.X.; Hsu, H.T.; Burgevin, A.; Guenette, S.; Moser, A.; van Endert, P. No major role for insulin-degrading enzyme in antigen presentation by MHC molecules. PLoS One 2014, 9, e88365. [Google Scholar] [CrossRef] [PubMed]
  48. El Hage, F.; Stroobant, V.; Vergnon, I.; Baurain, J.F.; Echchakir, H.; Lazar, V.; Chouaib, S.; Coulie, P.G.; Mami-Chouaib, F. Preprocalcitonin signal peptide generates a cytotoxic T-lymphocyte-defined tumor epitope processed by a proteasome-independent pathway. Proc. Natl. Acad. Sci. USA 2008, 105, 10119–10124. [Google Scholar]
  49. Wölfel, C.; Drexler, I.; van Pel, A.; Thres, T.; Leister, N.; Herr, W.; Sutter, G.; Huber, C.; Wölfel, T. Transporter (TAP)- and proteasome-independent presentation of a melanoma-associated tyrosinase epitope. Int. J. Cancer 2000, 88, 432–438. [Google Scholar] [CrossRef] [PubMed]
  50. Leonhardt, R.M.; Fiegl, D.; Rufer, E.; Karger, A.; Bettin, B.; Knittler, M.R. Post-endoplasmic reticulum rescue of unstable MHC class I requires proprotein convertase PC7. J. Immunol. 2010, 184, 2985–2998. [Google Scholar] [CrossRef] [PubMed]
  51. Gil-Torregrosa, B.C.; Raul Castano, A.; del Val, M. Major histocompatibility complex class I viral antigen processing in the secretory pathway defined by the trans-Golgi network protease furin. J. Exp. Med. 1998, 188, 1105–1116. [Google Scholar] [PubMed]
  52. Tey, S.K.; Khanna, R. Autophagy mediates transporter associated with antigen processing-independent presentation of viral epitopes through MHC class I pathway. Blood 2012, 120, 994–1004. [Google Scholar] [CrossRef] [PubMed]
  53. English, L.; Chemali, M.; Duron, J.; Rondeau, C.; Laplante, A.; Gingras, D.; Alexander, D.; Leib, D.; Norbury, C.; Lippe, R.; et al. Autophagy enhances the presentation of endogenous viral antigens on MHC class I molecules during HSV-1 infection. Nat. Immunol. 2009, 10, 480–487. [Google Scholar] [CrossRef] [PubMed]
  54. Demachi-Okamura, A.; Torikai, H.; Akatsuka, Y.; Miyoshi, H.; Yoshimori, T.; Kuzushima, K. Autophagy creates a CTL epitope that mimics tumor-associated antigens. PLoS One 2012, 7, e47126. [Google Scholar] [CrossRef] [PubMed]
  55. Hanada, K.; Yewdell, J.W.; Yang, J.C. Immune recognition of a human renal cancer antigen through post-translational protein splicing. Nature 2004, 427, 252–256. [Google Scholar] [CrossRef] [PubMed]
  56. Dalet, A.; Stroobant, V.; Vigneron, N.; Van den Eynde, B.J. Differences in the production of spliced antigenic peptides by the standard proteasome and the immunoproteasome. Eur. J. Immunol. 2011, 41, 39–46. [Google Scholar] [CrossRef] [PubMed]
  57. Michaux, A.; Larrieu, P.; Stroobant, V.; Fonteneau, J.F.; Jotereau, F.; Van den Eynde, B.J.; Moreau-Aubry, A.; Vigneron, N. A spliced antigenic peptide comprising a single spliced amino acid is produced in the proteasome by reverse splicing of a longer peptide fragment followed by trimming. J. Immunol. 2014, 192, 1962–1971. [Google Scholar] [CrossRef] [PubMed]
  58. Wölfel, T.; van Pel, A.; Brichard, V.; Schneider, J.; Seliger, B.; Meyer zum Büschenfelde, K.-H.; Boon, T. Two tyrosinase nonapeptides recognized on HLA-A2 melanomas by autologous cytolytic T-lymphocytes. Eur. J. Immunol. 1994, 24, 759–764. [Google Scholar] [CrossRef] [PubMed]
  59. Dalet, A.; Robbins, P.F.; Stroobant, V.; Vigneron, N.; Li, Y.F.; El-Gamil, M.; Hanada, K.; Yang, J.C.; Rosenberg, S.A.; Van den Eynde, B.J. An antigenic peptide produced by reverse splicing and double asparagine deamidation. Proc. Natl. Acad. Sci. USA 2011, 108, E323–E331. [Google Scholar] [CrossRef] [PubMed]
  60. Kawakami, Y.; Eliyahu, S.; Sakaguchi, K.; Robbins, P.F.; Rivoltini, L.; Yannelli, J.R.; Appella, E.; Rosenberg, S.A. Identification of the immunodominant peptides of the MART-1 human melanoma antigen recognized by the majority of HLA-A2-restricted tumor infiltrating lymphocytes. J. Exp. Med. 1994, 180, 347–352. [Google Scholar] [CrossRef] [PubMed]
  61. Schultz, E.S.; Chapiro, J.; Lurquin, C.; Claverol, S.; Burlet-Schiltz, O.; Warnier, G.; Russo, V.; Morel, S.; Levy, F.; Boon, T.; et al. The production of a new MAGE-3 peptide presented to cytolytic T-lymphocytes by HLA-B40 requires the immunoproteasome. J. Exp. Med. 2002, 195, 391–399. [Google Scholar] [CrossRef] [PubMed]
  62. Van der Bruggen, P.; Bastin, J.; Gajewski, T.; Coulie, P.G.; Boël, P.; de Smet, C.; Traversari, C.; Townsend, A.; Boon, T. A peptide encoded by human gene MAGE-3 and presented by HLA-A2 induces cytolytic T-lymphocytes that recognize tumor cells expressing MAGE-3. Eur. J. Immunol. 1994, 24, 3038–3043. [Google Scholar]
  63. Huang, L.-Q.; Brasseur, F.; Serrano, A.; de Plaen, E.; van der Bruggen, P.; Boon, T.; van Pel, A. Cytolytic T-lymphocytes recognize an antigen encoded by MAGE-A10 on a human melanoma. J. Immunol. 1999, 162, 6849–6854. [Google Scholar] [PubMed]
  64. Ma, W.; Germeau, C.; Vigneron, N.; Maernoudt, A.-S.; Morel, S.; Boon, T.; Coulie, P.G.; Van den Eynde, B. Two new tumor-specific antigenic peptides encoded by gene MAGE-C2 and presented to cytolytic T-lymphocytes by HLA-A2. Int. J. Cancer 2004, 109, 698–702. [Google Scholar] [CrossRef] [PubMed]
  65. Ma, W.; Vigneron, N.; Chapiro, J.; Stroobant, V.; Germeau, C.; Boon, T.; Coulie, P.G.; van den Eynde, B.J. A MAGE-C2 antigenic peptide processed by the immunoproteasome is recognized by cytolytic T-cells isolated from a melanoma patient after successful immunotherapy. Int. J. Cancer 2011, 129, 2427–2434. [Google Scholar] [CrossRef] [PubMed]
  66. Corbière, V.; Chapiro, J.; Stroobant, V.; Ma, W.; Lurquin, C.; Lethe, B.; Van Baren, N.; van den Eynde, B.J.; Boon, T.; Coulie, P.G. Antigen spreading contributes to MAGE vaccination-induced regression of melanoma metastases. Cancer. Res. 2011, 71, 1253–1262. [Google Scholar] [CrossRef] [PubMed]
  67. Robbins, P.F.; Lu, Y.C.; El-Gamil, M.; Li, Y.F.; Gross, C.; Gartner, J.; Lin, J.C.; Teer, J.K.; Cliften, P.; Tycksen, E.; et al. Mining exomic sequencing data to identify mutated antigens recognized by adoptively transferred tumor-reactive T-cells. Nat. Med. 2013, 19, 747–752. [Google Scholar] [CrossRef] [PubMed]
  68. Warren, E.H.; Vigneron, N.J.; Gavin, M.A.; Coulie, P.G.; Stroobant, V.; Dalet, A.; Tykodi, S.S.; Xuereb, S.M.; Mito, J.K.; Riddell, S.R.; et al. An antigen produced by splicing of noncontiguous peptides in the reverse order. Science 2006, 313, 1444–1447. [Google Scholar] [CrossRef] [PubMed]
  69. Löwe, J.; Stock, D.; Jap, B.; Zwickl, P.; Baumeister, W.; Huber, R. Crystal structure of the 20S proteasome from the archaeon T. acidophilum at 3.4 Å resolution. Science 1995, 268, 533–539. [Google Scholar] [CrossRef] [PubMed]
  70. Groll, M.; Ditzel, L.; Löwe, J.; Stock, D.; Bochtler, M.; Bartunik, H.D.; Huber, R. Structure of 20S proteasome from yeast at 2.4 Å resolution. Nature 1997, 386, 463–471. [Google Scholar] [CrossRef] [PubMed]
  71. Seemuller, E.; Lupas, A.; Stock, D.; Lowe, J.; Huber, R.; Baumeister, W. Proteasome from Thermoplasma acidophilum: A threonine protease. Science 1995, 268, 579–582. [Google Scholar] [CrossRef] [PubMed]
  72. Heinemeyer, W.; Fischer, M.; Krimmer, T.; Stachon, U.; Wolf, D.H. The active sites of the eukaryotic 20 S proteasome and their involvement in subunit precursor processing. J. Biol. Chem. 1997, 272, 25200–25209. [Google Scholar] [CrossRef] [PubMed]
  73. Groll, M.; Huber, R. Substrate access and processing by the 20S proteasome core particle. Int. J. Biochem. Cell Biol. 2003, 35, 606–616. [Google Scholar] [CrossRef] [PubMed]
  74. Dalet, A.; Vigneron, N.; Stroobant, V.; Hanada, K.; Van den Eynde, B.J. Splicing of distant peptide fragments occurs in the proteasome by transpeptidation and produces the spliced antigenic peptide derived from fibroblast growth factor-5. J. Immunol. 2010, 184, 3016–3024. [Google Scholar] [CrossRef] [PubMed]
  75. Mishto, M.; Goede, A.; Taube, K.T.; Keller, C.; Janek, K.; Henklein, P.; Niewienda, A.; Kloss, A.; Gohlke, S.; Dahlmann, B.; et al. Driving forces of proteasome-catalyzed peptide splicing in yeast and humans. Mol. Cell. Proteomics 2012, 11, 1008–1023. [Google Scholar] [CrossRef] [PubMed]
  76. Liepe, J.; Mishto, M.; Textoris-Taube, K.; Janek, K.; Keller, C.; Henklein, P.; Kloetzel, P.M.; Zaikin, A. The 20S proteasome splicing activity discovered by SpliceMet. PLoS Comput. Biol. 2010, 6, e1000830. [Google Scholar] [CrossRef] [PubMed]
  77. Van der Bruggen, P.; Zhang, Y.; Chaux, P.; Stroobant, V.; Panichelli, C.; Schultz, E.S.; Chapiro, J.; Van den Eynde, B.J.; Brasseur, F.; Boon, T. Tumor-specific shared antigenic peptides recognized by human T-cells. Immunol. Rev. 2002, 188, 51–64. [Google Scholar] [CrossRef] [PubMed]
  78. Baumeister, W.; Walz, J.; Zühl, F.; Seemüller, E. The proteasome: Paradigm of a self-compartmentalizing protease. Cell 1998, 92, 367–380. [Google Scholar] [CrossRef] [PubMed]
  79. Unno, M.; Mizushima, T.; Morimoto, Y.; Tomisugi, Y.; Tanaka, K.; Yasuoka, N.; Tsukihara, T. The structure of the mammalian 20S proteasome at 2.75 Å resolution. Structure 2002, 10, 609–618. [Google Scholar] [CrossRef] [PubMed]
  80. Davies, K.J. Degradation of oxidized proteins by the 20S proteasome. Biochimie 2001, 83, 301–310. [Google Scholar] [CrossRef] [PubMed]
  81. Pickering, A.M.; Koop, A.L.; Teoh, C.Y.; Ermak, G.; Grune, T.; Davies, K.J. The immunoproteasome, the 20S proteasome and the PA28αβ proteasome regulator are oxidative-stress-adaptive proteolytic complexes. Biochem. J. 2010, 432, 585–594. [Google Scholar] [CrossRef] [PubMed]
  82. Brooks, P.; Fuertes, G.; Murray, R.Z.; Bose, S.; Knecht, E.; Rechsteiner, M.C.; Hendil, K.B.; Tanaka, K.; Dyson, J.; Rivett, J. Subcellular localization of proteasomes and their regulatory complexes in mammalian cells. Biochem. J. 2000, 346, 155–161. [Google Scholar] [CrossRef] [PubMed]
  83. Peters, J.M.; Franke, W.W.; Kleinschmidt, J.A. Distinct 19S and 20S subcomplexes of the 26S proteasome and their distribution in the nucleus and the cytoplasm. J. Biol. Chem. 1994, 269, 7709–7718. [Google Scholar] [PubMed]
  84. Baugh, J.M.; Viktorova, E.G.; Pilipenko, E.V. Proteasomes can degrade a significant proportion of cellular proteins independent of ubiquitination. J. Mol. Biol. 2009, 386, 814–827. [Google Scholar] [CrossRef] [PubMed]
  85. Whittier, J.E.; Xiong, Y.; Rechsteiner, M.C.; Squier, T.C. Hsp90 enhances degradation of oxidized calmodulin by the 20S proteasome. J. Biol. Chem. 2004, 279, 46135–46142. [Google Scholar] [CrossRef] [PubMed]
  86. Orlowski, M. Selective activation of the 20S proteasome (multicatalytic proteinase complex) by histone H3. Biochemistry 2001, 40, 15318–15326. [Google Scholar] [CrossRef] [PubMed]
  87. Kisselev, A.F.; Kaganovich, D.; Goldberg, A.L. Binding of hydrophobic peptides to several non-catalytic sites promotes peptide hydrolysis by all active sites of 20S proteasomes. Evidence for peptide-induced channel opening in the α-rings. J. Biol. Chem. 2002, 277, 22260–22270. [Google Scholar] [CrossRef] [PubMed]
  88. Wang, X.; Yen, J.; Kaiser, P.; Huang, L. Regulation of the 26S proteasome complex during oxidative stress. Sci. Signal. 2010. [Google Scholar] [CrossRef]
  89. Grune, T.; Catalgol, B.; Licht, A.; Ermak, G.; Pickering, A.M.; Ngo, J.K.; Davies, K.J. Hsp70 mediates dissociation and reassociation of the 26S proteasome during adaptation to oxidative stress. Free Radic. Biol. Med. 2011, 51, 1355–1364. [Google Scholar] [CrossRef] [PubMed]
  90. Bajorek, M.; Finley, D.; Glickman, M.H. Proteasome disassembly and downregulation is correlated with viability during stationary phase. Curr. Biol. 2003, 13, 1140–1144. [Google Scholar] [CrossRef] [PubMed]
  91. Aiken, C.T.; Kaake, R.M.; Wang, X.; Huang, L. Oxidative stress-mediated regulation of proteasome complexes. Mol. Cell. Proteomics 2011. [Google Scholar] [CrossRef]
  92. Li, J.; Powell, S.R.; Wang, X. Enhancement of proteasome function by PA28α; overexpression protects against oxidative stress. FASEB J. 2011, 25, 883–893. [Google Scholar] [CrossRef] [PubMed]
  93. Griffin, T.A.; Nandi, D.; Cruz, M.; Fehling, H.J.; van Kaer, L.; Monaco, J.J.; Colbert, R.A. Immunoproteasome assembly: Cooperative incorporation of interferon γ (IFN-γ)-inducible subunits. J. Exp. Med. 1998, 187, 97–104. [Google Scholar] [CrossRef] [PubMed]
  94. Enenkel, C.; Lehmann, H.; Kipper, J.; Guckel, R.; Hilt, W.; Wolf, D.H. PRE3, highly homologous to the human major histocompatibility complex-linked LMP2 (RING12) gene, codes for a yeast proteasome subunit necessary for the peptidylglutamyl-peptide hydrolyzing activity. FEBS Lett. 1994, 341, 193–196. [Google Scholar] [CrossRef] [PubMed]
  95. Dick, T.P.; Nussbaum, A.K.; Deeg, M.; Heinemeyer, W.; Groll, M.; Schirle, M.; Keilholz, W.; Stevanovic, S.; Wolf, D.H.; Huber, R.; et al. Contribution of proteasomal β-subunits to the cleavage of peptide substrates analyzed with yeast mutants. J. Biol. Chem. 1998, 273, 25637–25646. [Google Scholar] [CrossRef] [PubMed]
  96. Nussbaum, A.K.; Dick, T.P.; Keilholz, W.; Schirle, M.; Stevanovic, S.; Dietz, K.; Heinemeyer, W.; Groll, M.; Wolf, D.H.; Huber, R.; et al. Cleavage motifs of the yeast 20S proteasome β subunits deduced from digests of enolase 1. Proc. Natl. Acad. Sci. USA 1998, 95, 12504–12509. [Google Scholar] [CrossRef] [PubMed]
  97. Toes, R.E.M.; Nussbaum, A.K.; Degermann, S.; Schirle, M.; Emmerich, N.P.N.; Kraft, M.; Laplace, C.; Zwinderman, A.; Dick, T.P.; Müller, J.; et al. Discrete cleavage motifs of constitutive and immunoproteasomes revealed by quantitative analysis of cleavage products. J. Exp. Med. 2001, 194, 1–12. [Google Scholar] [CrossRef] [PubMed]
  98. Gaczynska, M.; Rock, K.L.; Goldberg, A.L. γ-interferon and expression of MHC genes regulate peptide hydrolysis by proteasomes. Nature 1993, 365, 264–267. [Google Scholar] [CrossRef] [PubMed]
  99. Gaczynska, M.; Rock, K.L.; Spies, T.; Goldberg, A.L. Peptidase activities of proteasomes are differentially regulated by the major histocompatibility complex-encoded genes for LMP2 and LMP7. Proc. Natl. Acad. Sci. USA 1994, 91, 9213–9217. [Google Scholar] [CrossRef] [PubMed]
  100. Driscoll, J.; Brown, M.; Finley, D.; Monaco, J. MHC-linked LMP gene products specifically alter peptidase activities of the proteasome. Nature 1993, 365, 262–264. [Google Scholar] [CrossRef] [PubMed]
  101. Boes, B.; Hengel, H.; Ruppert, T.; Multhaup, G.; Koszinowski, U.H.; Kloetzel, P.M. Interferon γ stimulation modulates the proteolytic activity and cleavage site preference of 20S mouse proteasomes. J. Exp. Med. 1994, 179, 901–909. [Google Scholar] [CrossRef] [PubMed]
  102. Fehling, H.J.; Swat, W.; Laplace, C.; Kühn, R.; Rajewsky, K.; Müller, U.; von Boehmer, H. MHC class I expression in mice lacking the proteasome subunit LMP-7. Science 1994, 265, 1234–1237. [Google Scholar] [CrossRef] [PubMed]
  103. Van Kaer, L.; Ashton-Rickardt, P.G.; Eichelberger, M.; Gaczynska, M.; Nagashima, K.; Rock, K.L.; Goldberg, A.L.; Doherty, P.C.; Tonegawa, S. Altered peptidase and viral-specific T-cell response in LMP2 mutant mice. Immunity 1994, 1, 533–541. [Google Scholar] [CrossRef] [PubMed]
  104. Kincaid, E.Z.; Che, J.W.; York, I.; Escobar, H.; Reyes-Vargas, E.; Delgado, J.C.; Welsh, R.M.; Karow, M.L.; Murphy, A.J.; Valenzuela, D.M.; et al. Mice completely lacking immunoproteasomes show major changes in antigen presentation. Nat. Immunol. 2012, 13, 129–135. [Google Scholar] [CrossRef]
  105. De Verteuil, D.; Muratore-Schroeder, T.L.; Granados, D.P.; Fortier, M.H.; Hardy, M.P.; Bramoulle, A.; Caron, E.; Vincent, K.; Mader, S.; Lemieux, S.; et al. Deletion of immunoproteasome subunits imprints on the transcriptome and has a broad impact on peptides presented by major histocompatibility complex I molecules. Mol. Cell. Proteomics 2010, 9, 2034–2047. [Google Scholar] [CrossRef] [PubMed]
  106. Vigneron, N.; Van den Eynde, B.J. Proteasome subtypes and the processing of tumor antigens: Increasing antigenic diversity. Curr. Opin. Immunol. 2012, 24, 84–91. [Google Scholar] [CrossRef] [PubMed]
  107. Basler, M.; Lauer, C.; Moebius, J.; Weber, R.; Przybylski, M.; Kisselev, A.F.; Tsu, C.; Groettrup, M. Why the structure but not the activity of the immunoproteasome subunit low molecular mass polypeptide 2 rescues antigen presentation. J. Immunol. 2012, 189, 1868–1877. [Google Scholar] [CrossRef] [PubMed]
  108. Chen, W.; Norbury, C.C.; Cho, Y.; Yewdell, J.W.; Bennink, J.R. Immunoproteasomes shape immunodominance hierarchies of antiviral CD8+ T-cells at the levels of T-cell repertoire and presentation of viral antigens. J. Exp. Med. 2001, 193, 1319–1326. [Google Scholar] [CrossRef] [PubMed]
  109. Chapatte, L.; Ayyoub, M.; Morel, S.; Peitrequin, A.L.; Levy, N.; Servis, C.; Van den Eynde, B.J.; Valmori, D.; Levy, F. Processing of tumor-associated antigen by the proteasomes of dendritic cells controls in vivo T-cell responses. Cancer Res. 2006, 66, 5461–5468. [Google Scholar] [CrossRef]
  110. Dannull, J.; Haley, N.R.; Archer, G.; Nair, S.; Boczkowski, D.; Harper, M.; de Rosa, N.; Pickett, N.; Mosca, P.J.; Burchette, J.; et al. Melanoma immunotherapy using mature DCs expressing the constitutive proteasome. J. Clin. Invest. 2013, 123, 3135–3145. [Google Scholar] [CrossRef] [PubMed]
  111. Dannull, J.; Lesher, D.T.; Holzknecht, R.; Qi, W.; Hanna, G.; Seigler, H.; Tyler, D.S.; Pruitt, S.K. Immunoproteasome down-modulation enhances the ability of dendritic cells to stimulate antitumor immunity. Blood 2007, 110, 4341–4350. [Google Scholar] [CrossRef] [PubMed]
  112. Basler, M.; Moebius, J.; Elenich, L.; Groettrup, M.; Monaco, J.J. An altered T-cell repertoire in MECL-1-deficient mice. J. Immunol. 2006, 176, 6665–6672. [Google Scholar] [CrossRef] [PubMed]
  113. Basler, M.; Youhnovski, N.; van den Broek, M.; Przybylski, M.; Groettrup, M. Immunoproteasomes down-regulate presentation of a subdominant T-cell epitope from lymphocytic choriomeningitis virus. J. Immunol. 2004, 173, 3925–3934. [Google Scholar] [CrossRef] [PubMed]
  114. Pang, K.C.; Sanders, M.T.; Monaco, J.J.; Doherty, P.C.; Turner, S.J.; Chen, W. Immunoproteasome subunit deficiencies impact differentially on two immunodominant influenza virus-specific CD8+ T-cell responses. J. Immunol. 2006, 177, 7680–7688. [Google Scholar] [CrossRef] [PubMed]
  115. Hutchinson, S.; Sims, S.; O’Hara, G.; Silk, J.; Gileadi, U.; Cerundolo, V.; Klenerman, P. A dominant role for the immunoproteasome in CD8+ T-cell responses to murine cytomegalovirus. PLoS One 2011, 6, e14646. [Google Scholar] [CrossRef] [PubMed]
  116. Zimmermann, A.; Trilling, M.; Wagner, M.; Wilborn, M.; Bubic, I.; Jonjic, S.; Koszinowski, U.; Hengel, H. A cytomegaloviral protein reveals a dual role for STAT2 in IFN-γ signaling and antiviral responses. J. Exp. Med. 2005, 201, 1543–1553. [Google Scholar] [CrossRef] [PubMed]
  117. Dahlmann, B.; Ruppert, T.; Kuehn, L.; Merforth, S.; Kloetzel, P.-M. Different proteasome subtypes in a single tissue exhibit different enzymatic properties. J. Mol. Biol. 2000, 303, 643–653. [Google Scholar] [CrossRef] [PubMed]
  118. Groettrup, M.; Standera, S.; Stohwasser, R.; Kloetzel, P.M. The subunits MECL-1 and LMP2 are mutually required for incorporation into the 20S proteasome. Proc. Natl. Acad. Sci. USA 1997, 94, 8970–8975. [Google Scholar] [CrossRef] [PubMed]
  119. De, M.; Jayarapu, K.; Elenich, L.; Monaco, J.J.; Colbert, R.A.; Griffin, T.A. β2 subunit propeptides influence cooperative proteasome assembly. J. Biol. Chem. 2003, 278, 6153–6159. [Google Scholar] [CrossRef] [PubMed]
  120. Kingsbury, D.J.; Griffin, T.A.; Colbert, R.A. Novel propeptide function in 20S proteasome assembly influences β subunit composition. J. Biol. Chem. 2000, 275, 24156–24162. [Google Scholar] [CrossRef] [PubMed]
  121. Groll, M.; Heinemeyer, W.; Jäger, S.; Ullrich, T.; Bochtler, M.; Wolf, D.H.; Huber, R. The catalytic sites of 20S proteasomes and their role in subunit maturation: A mutational and crystallographic study. Proc. Natl. Acad. Sci. USA 1999, 96, 10976–10983. [Google Scholar] [CrossRef] [PubMed]
  122. Klare, N.; Seeger, M.; Janek, K.; Jungblut, P.R.; Dahlmann, B. Intermediate-type 20S proteasomes in HeLa cells: “Asymmetric” subunit composition, diversity and adaptation. J. Mol. Biol. 2007, 373, 1–10. [Google Scholar] [CrossRef] [PubMed]
  123. Murata, S.; Sasaki, K.; Kishimoto, T.; Niwa, S.; Hayashi, H.; Takahama, Y.; Tanaka, K. Regulation of CD8+ T-cell development by thymus-specific proteasomes. Science 2007, 316, 1349–1353. [Google Scholar] [CrossRef] [PubMed]
  124. Tomaru, U.; Ishizu, A.; Murata, S.; Miyatake, Y.; Suzuki, S.; Takahashi, S.; Kazamaki, T.; Ohara, J.; Baba, T.; Iwasaki, S.; et al. Exclusive expression of proteasome subunit β5t in the human thymic cortex. Blood 2009, 113, 5186–5191. [Google Scholar] [CrossRef] [PubMed]
  125. Nitta, T.; Murata, S.; Sasaki, K.; Fujii, H.; Ripen, A.M.; Ishimaru, N.; Koyasu, S.; Tanaka, K.; Takahama, Y. Thymoproteasome shapes immunocompetent repertoire of CD8+ T-cells. Immunity 2010, 32, 29–40. [Google Scholar] [CrossRef] [PubMed]
  126. Ziegler, A.; Muller, C.A.; Bockmann, R.A.; Uchanska-Ziegler, B. Low-affinity peptides and T-cell selection. Trends Immunol. 2009, 30, 53–60. [Google Scholar] [CrossRef] [PubMed]
  127. Xing, Y.; Jameson, S.C.; Hogquist, K.A. Thymoproteasome subunit-β5t generates peptide-MHC complexes specialized for positive selection. Proc. Natl. Acad. Sci. USA 2013, 110, 6979–6984. [Google Scholar] [CrossRef] [PubMed]
  128. Groettrup, M.; Kirk, C.J.; Basler, M. Proteasomes in immune cells: more than peptide producers? Nat. Rev. Immunol. 2010, 10, 73–78. [Google Scholar] [CrossRef]
  129. Gotter, J.; Brors, B.; Hergenhahn, M.; Kyewski, B. Medullary epithelial cells of the human thymus express a highly diverse selection of tissue-specific genes colocalized in chromosomal clusters. J. Exp. Med. 2004, 199, 155–166. [Google Scholar] [CrossRef] [PubMed]
  130. Huijbers, I.J.; Soudja, S.M.; Uyttenhove, C.; Buferne, M.; Inderberg-Suso, E.M.; Colau, D.; Pilotte, L.; Chomez, P.; Brasseur, F.; Schmitt-Verhulst, A.M.; et al. Minimal tolerance to a tumor antigen encoded by a cancer-germline gene. J. Immunol. 2012, 188, 111–121. [Google Scholar] [CrossRef] [PubMed]
  131. Huber, E.M.; Basler, M.; Schwab, R.; Heinemeyer, W.; Kirk, C.J.; Groettrup, M.; Groll, M. Immuno- and constitutive proteasome crystal structures reveal differences in substrate and inhibitor specificity. Cell 2012, 148, 727–738. [Google Scholar] [CrossRef] [PubMed]
  132. Sijts, A.J.A.M.; Ruppert, T.; Rehermann, B.; Schmidt, M.; Koszinowski, U.; Kloetzel, P.-M. Efficient generation of a hepatitis B virus cytotoxic T-lymphocyte epitope requires the structural features of immunoproteasomes. J. Exp. Med. 2000, 191, 503–513. [Google Scholar] [CrossRef] [PubMed]
  133. Seifert, U.; Bialy, L.P.; Ebstein, F.; Bech-Otschir, D.; Voigt, A.; Schroter, F.; Prozorovski, T.; Lange, N.; Steffen, J.; Rieger, M.; et al. Immunoproteasomes preserve protein homeostasis upon interferon-induced oxidative stress. Cell 2010, 142, 613–624. [Google Scholar] [CrossRef] [PubMed]
  134. Nathan, J.A.; Spinnenhirn, V.; Schmidtke, G.; Basler, M.; Groettrup, M.; Goldberg, A.L. Immuno- and constitutive proteasomes do not differ in their abilities to degrade ubiquitinated proteins. Cell 2013, 152, 1184–1194. [Google Scholar] [CrossRef] [PubMed]
  135. Hayashi, T.; Faustman, D. NOD mice are defective in proteasome production and activation of NF-κB. Mol. Cell. Biol. 1999, 19, 8646–8659. [Google Scholar] [PubMed]
  136. Kessler, B.M.; Lennon-Dumenil, A.M.; Shinohara, M.L.; Lipes, M.A.; Ploegh, H.L. LMP2 expression and proteasome activity in NOD mice. Nat. Med. 2000, 6, 1064. [Google Scholar] [PubMed]
  137. Runnels, H.A.; Watkins, W.A.; Monaco, J.J. LMP2 expression and proteasome activity in NOD mice. Nat. Med. 2000, 6, 1064–1065. [Google Scholar] [PubMed]
  138. Hensley, S.E.; Zanker, D.; Dolan, B.P.; David, A.; Hickman, H.D.; Embry, A.C.; Skon, C.N.; Grebe, K.M.; Griffin, T.A.; Chen, W.; et al. Unexpected role for the immunoproteasome subunit LMP2 in antiviral humoral and innate immune responses. J. Immunol. 2010, 184, 4115–4122. [Google Scholar] [CrossRef] [PubMed]
  139. Moebius, J.; van den Broek, M.; Groettrup, M.; Basler, M. Immunoproteasomes are essential for survival and expansion of T-cells in virus-infected mice. Eur. J. Immunol. 2010, 40, 3439–3449. [Google Scholar] [CrossRef] [PubMed]
  140. Maldonado, M.; Kapphahn, R.J.; Terluk, M.R.; Heuss, N.D.; Yuan, C.; Gregerson, D.S.; Ferrington, D.A. Immunoproteasome deficiency modifies the alternative pathway of NF-κB signaling. PLoS One 2013, 8, e56187. [Google Scholar] [CrossRef] [PubMed]
  141. Muchamuel, T.; Basler, M.; Aujay, M.A.; Suzuki, E.; Kalim, K.W.; Lauer, C.; Sylvain, C.; Ring, E.R.; Shields, J.; Jiang, J.; et al. A selective inhibitor of the immunoproteasome subunit LMP7 blocks cytokine production and attenuates progression of experimental arthritis. Nat. Med. 2009, 15, 781–787. [Google Scholar] [CrossRef] [PubMed]
  142. Pickart, C.M. Mechanisms underlying ubiquitination. Annu. Rev. Biochem. 2001, 70, 503–533. [Google Scholar] [CrossRef] [PubMed]
  143. Forster, F.; Unverdorben, P.; Sledz, P.; Baumeister, W. Unveiling the long-held secrets of the 26S proteasome. Structure 2013, 21, 1551–1562. [Google Scholar] [CrossRef] [PubMed]
  144. Yao, T.; Cohen, R.E. A cryptic protease couples deubiquitination and degradation by the proteasome. Nature 2002, 419, 403–407. [Google Scholar] [CrossRef] [PubMed]
  145. Verma, R.; Aravind, L.; Oania, R.; McDonald, W.H.; Yates, J.R., 3rd; Koonin, E.V.; Deshaies, R.J. Role of Rpn11 metalloprotease in deubiquitination and degradation by the 26S proteasome. Science 2002, 298, 611–615. [Google Scholar] [CrossRef] [PubMed]
  146. Pathare, G.R.; Nagy, I.; Bohn, S.; Unverdorben, P.; Hubert, A.; Korner, R.; Nickell, S.; Lasker, K.; Sali, A.; Tamura, T.; et al. The proteasomal subunit Rpn6 is a molecular clamp holding the core and regulatory subcomplexes together. Proc. Natl. Acad. Sci. USA 2012, 109, 149–154. [Google Scholar] [CrossRef] [PubMed]
  147. Tanahashi, N.; Murakami, Y.; Minami, Y.; Shimbara, N.; Hendil, K.B.; Tanaka, K. Hybrid proteasomes. Induction by interferon-γ and contribution to ATP-dependent proteolysis. J. Biol. Chem. 2000, 275, 14336–14345. [Google Scholar] [CrossRef] [PubMed]
  148. Ustrell, V.; Hoffman, L.; Pratt, G.; Rechsteiner, M. PA200, a nuclear proteasome activator involved in DNA repair. EMBO J. 2002, 21, 3516–3525. [Google Scholar] [CrossRef] [PubMed]
  149. Ma, C.P.; Slaughter, C.A.; DeMartino, G.N. Identification, purification, and characterization of a protein activator (PA28) of the 20 S proteasome (macropain). J. Biol. Chem. 1992, 267, 10515–10523. [Google Scholar] [PubMed]
  150. Dubiel, W.; Pratt, G.; Ferrell, K.; Rechsteiner, M. Purification of an 11S regulator of the multicatalytic protease. J. Biol. Chem. 1992, 267, 22369–22377. [Google Scholar] [PubMed]
  151. Rechsteiner, M.; Realini, C.; Ustrell, V. The proteasome activator 11S REG (PA28) and class I antigen presentation. Biochem. J. 2000, 345, 1–15. [Google Scholar] [CrossRef] [PubMed]
  152. Realini, C.; Dubiel, W.; Pratt, G.; Ferrell, K.; Rechsteiner, M. Molecular cloning and expression of a γ-interferon-inducible activator of the multicatalytic protease. J. Biol. Chem. 1994, 269, 20727–20732. [Google Scholar] [PubMed]
  153. Ahn, J.Y.; Tanahashi, N.; Akiyama, K.; Hisamatsu, H.; Noda, C.; Tanaka, K.; Chung, C.H.; Shibmara, N.; Willy, P.J.; Mott, J.D.; et al. Primary structures of two homologous subunits of PA28, a γ-interferon-inducible protein activator of the 20S proteasome. FEBS Lett. 1995, 366, 37–42. [Google Scholar] [CrossRef] [PubMed]
  154. Gray, C.W.; Slaughter, C.A.; DeMartino, G.N. PA28 activator protein forms regulatory caps on proteasome stacked rings. J. Mol. Biol. 1994, 236, 7–15. [Google Scholar] [CrossRef] [PubMed]
  155. Knowlton, J.R.; Johnston, S.C.; Whitby, F.G.; Realini, C.; Zhang, Z.; Rechsteiner, M.; Hill, C.P. Structure of the proteasome activator REGα (PA28α). Nature 1997, 390, 639–643. [Google Scholar] [CrossRef] [PubMed]
  156. Zhang, Z.; Krutchinsky, A.; Endicott, S.; Realini, C.; Rechsteiner, M.; Standing, K.G. Proteasome activator 11S REG or PA28: Recombinant REG α/REG β hetero-oligomers are heptamers. Biochemistry 1999, 38, 5651–5658. [Google Scholar] [CrossRef] [PubMed]
  157. Hendil, K.B.; Khan, S.; Tanaka, K. Simultaneous binding of PA28 and PA700 activators to 20S proteasomes. Biochem. J. 1998, 332, 749–754. [Google Scholar] [PubMed]
  158. Cascio, P.; Call, M.; Petre, B.M.; Walz, T.; Goldberg, A.L. Properties of the hybrid form of the 26S proteasome containing both 19S and PA28 complexes. EMBO J. 2002, 21, 2636–2645. [Google Scholar] [CrossRef] [PubMed]
  159. DeMartino, G.N.; Slaughter, C.A. The proteasome, a novel protease regulated by multiple mechanisms. J. Biol. Chem. 1999, 274, 22123–22126. [Google Scholar] [CrossRef] [PubMed]
  160. Whitby, F.G.; Masters, E.I.; Kramer, L.; Knowlton, J.R.; Yao, Y.; Wang, C.C.; Hill, C.P. Structural basis for the activation of 20S proteasomes by 11S regulators. Nature 2000, 408, 115–120. [Google Scholar] [CrossRef] [PubMed]
  161. Kohler, A.; Cascio, P.; Leggett, D.S.; Woo, K.M.; Goldberg, A.L.; Finley, D. The axial channel of the proteasome core particle is gated by the Rpt2 ATPase and controls both substrate entry and product release. Mol. Cell 2001, 7, 1143–1152. [Google Scholar] [CrossRef] [PubMed]
  162. Yamano, T.; Murata, S.; Shimbara, N.; Tanaka, N.; Chiba, T.; Tanaka, K.; Yui, K.; Udono, H. Two distinct pathways mediated by PA28 and hsp90 in major histocompatibility complex class I antigen processing. J. Exp. Med. 2002, 196, 185–196. [Google Scholar] [CrossRef] [PubMed]
  163. Preckel, T.; Fung-Leung, W.-P.; Cai, Z.; Vitiello, A.; Salter-Cid, L.; Winqvist, O.; Wolfe, T.G.; von Herrath, M.; Angulo, A.; Ghazal, P.; et al. Impaired immunoproteasome assembly and immune responses in PA28−/− mice. Science 1999, 286, 2162–2165. [Google Scholar] [CrossRef] [PubMed]
  164. Murata, S.; Udono, H.; Tanahashi, N.; Hamada, H.; Watanabe, K.; Adachi, K.; Yamano, T.; Yui, K.; Kobayashi, H.; Kasahara, M.; et al. Immunoproteasome assembly and antigen presentation in mice lacking both PA28α and PA28β. EMBO J. 2001, 20, 5898–5907. [Google Scholar] [CrossRef] [PubMed]
  165. Dick, T.P.; Ruppert, T.; Groettrup, M.; Kloetzel, P.M.; Kuehn, L.; Koszinowski, U.H.; Stevanovic, S.; Schild, H.; Rammensee, H.-G. Coordinated dual cleavages induced by the proteasome regulator PA28 lead to dominant MHC ligands. Cell 1996, 86, 253–262. [Google Scholar] [CrossRef] [PubMed]
  166. De Graaf, N.; van Helden, M.J.; Textoris-Taube, K.; Chiba, T.; Topham, D.J.; Kloetzel, P.M.; Zaiss, D.M.; Sijts, A.J. PA28 and the proteasome immunosubunits play a central and independent role in the production of MHC class I-binding peptides in vivo. Eur. J. Immunol. 2011, 41, 926–935. [Google Scholar] [CrossRef] [PubMed]
  167. Groettrup, M.; Soza, A.; Eggers, M.; Kuehn, L.; Dick, T.P.; Schild, H.; Rammensee, H.-G.; Koszinowski, U.H.; Kloetzel, P.M. A role for the proteasome regulator PA28α in antigen presentation. Nature 1996, 381, 166–168. [Google Scholar] [CrossRef] [PubMed]
  168. Schwarz, K.; Eggers, M.; Soza, A.; Koszinowski, U.H.; Kloetzel, P.M.; Groettrup, M. The proteasome regulator PA28α/β can enhance antigen presentation without affecting 20S proteasome subunit composition. Eur. J. Immunol. 2000, 30, 3672–3679. [Google Scholar] [CrossRef] [PubMed]
  169. Van Hall, T.; Sijts, A.; Camps, M.; Offringa, R.; Melief, C.; Kloetzel, P.-M.; Ossendorp, F. Differential influence on cytotoxic T-lymphocyte epitope presentation by controlled expression of either proteasome immunosubunits or PA28. J. Exp. Med. 2000, 192, 483–494. [Google Scholar] [CrossRef] [PubMed]
  170. Sijts, A.; Sun, Y.; Janek, K.; Kral, S.; Paschen, A.; Schadendorf, D.; Kloetzel, P.M. The role of the proteasome activator PA28 in MHC class I antigen processing. Mol. Immunol. 2002, 39, 165–169. [Google Scholar] [CrossRef] [PubMed]
  171. Sun, Y.; Sijts, A.J.A.M.; Song, M.; Janek, K.; Nussbaum, A.K.; Kral, S.; Schirle, M.; Stevanovic, S.; Paschen, A.; Schild, H.; et al. Expression of the proteasome activator PA28 rescues the presentation of a cytotoxic T-lymphocyte epitope on melanoma cells. Cancer Res. 2002, 62, 2875–2882. [Google Scholar] [PubMed]
  172. Textoris-Taube, K.; Henklein, P.; Pollmann, S.; Bergann, T.; Weisshoff, H.; Seifert, U.; Drung, I.; Mugge, C.; Sijts, A.; Kloetzel, P.M.; et al. The N-terminal flanking region of the TRP2360–368 melanoma antigen determines proteasome activator PA28 requirement for epitope liberation. J. Biol. Chem. 2007, 282, 12749–12754. [Google Scholar] [CrossRef] [PubMed]
  173. Yamano, T.; Sugahara, H.; Mizukami, S.; Murata, S.; Chiba, T.; Tanaka, K.; Yui, K.; Udono, H. Allele-selective effect of PA28 in MHC class I antigen processing. J. Immunol. 2008, 181, 1655–1664. [Google Scholar] [CrossRef] [PubMed]
  174. Schwarz, K.; van den Broek, M.; Kostka, S.; Kraft, R.; Soza, A.; Schmidtke, G.; Kloetzel, P.-M.; Groettrup, M. Overexpression of the proteasome subunits LMP2, LMP7, and MECL-1, but not PA28α/β, enhances the presentation of an immunodominant lymphocytic choriomeningitis virus T-cell epitope. J. Immunol. 2000, 165, 768–778. [Google Scholar] [CrossRef] [PubMed]
  175. Nikaido, T.; Shimada, K.; Shibata, M.; Hata, M.; Sakamoto, M.; Takasaki, Y.; Sato, C.; Takahashi, T.; Nishida, Y. Cloning and nucleotide sequence of cDNA for Ki antigen, a highly conserved nuclear protein detected with sera from patients with systemic lupus erythematosus. Clin. Exp. Immunol. 1990, 79, 209–214. [Google Scholar] [CrossRef] [PubMed]
  176. Wilk, S.; Chen, W.E.; Magnusson, R.P. Properties of the nuclear proteasome activator PA28γ (REGγ). Arch. Biochem. Biophys. 2000, 383, 265–271. [Google Scholar] [CrossRef] [PubMed]
  177. Murata, S.; Kawahara, H.; Tohma, S.; Yamamoto, K.; Kasahara, M.; Nabeshima, Y.; Tanaka, K.; Chiba, T. Growth retardation in mice lacking the proteasome activator PA28γ. J. Biol. Chem. 1999, 274, 38211–38215. [Google Scholar] [CrossRef] [PubMed]
  178. Barton, L.F.; Runnels, H.A.; Schell, T.D.; Cho, Y.; Gibbons, R.; Tevethia, S.S.; Deepe, G.S.; Monaco, J.J. Immune defects in 28-kDa proteasome activator γ-deficient mice. J. Immunol. 2004, 172, 3948–3954. [Google Scholar] [CrossRef] [PubMed]
  179. Araya, R.; Takahashi, R.; Nomura, Y. Yeast two-hybrid screening using constitutive-active caspase-7 as bait in the identification of PA28γ as an effector caspase substrate. Cell Death Differ. 2002, 9, 322–328. [Google Scholar] [CrossRef] [PubMed]
  180. Hagemann, C.; Patel, R.; Blank, J.L. MEKK3 interacts with the PA28γ regulatory subunit of the proteasome. Biochem. J. 2003, 373, 71–79. [Google Scholar] [CrossRef]
  181. Li, X.; Lonard, D.M.; Jung, S.Y.; Malovannaya, A.; Feng, Q.; Qin, J.; Tsai, S.Y.; Tsai, M.J.; O’Malley, B.W. The SRC-3/AIB1 coactivator is degraded in a ubiquitin- and ATP-independent manner by the REGγ proteasome. Cell 2006, 124, 381–392. [Google Scholar] [CrossRef] [PubMed]
  182. Li, X.; Amazit, L.; Long, W.; Lonard, D.M.; Monaco, J.J.; O’Malley, B.W. Ubiquitin- and ATP-independent proteolytic turnover of p21 by the REGγ-proteasome pathway. Mol. Cell 2007, 26, 831–842. [Google Scholar] [CrossRef] [PubMed]
  183. Levy-Barda, A.; Lerenthal, Y.; Davis, A.J.; Chung, Y.M.; Essers, J.; Shao, Z.; van Vliet, N.; Chen, D.J.; Hu, M.C.; Kanaar, R.; et al. Involvement of the nuclear proteasome activator PA28γ in the cellular response to DNA double-strand breaks. Cell Cycle 2011, 10, 4300–4310. [Google Scholar] [CrossRef]
  184. Zannini, L.; Lecis, D.; Buscemi, G.; Carlessi, L.; Gasparini, P.; Fontanella, E.; Lisanti, S.; Barton, L.; Delia, D. REGγ proteasome activator is involved in the maintenance of chromosomal stability. Cell Cycle 2008, 7, 504–512. [Google Scholar] [CrossRef] [PubMed]
  185. Baldin, V.; Militello, M.; Thomas, Y.; Doucet, C.; Fic, W.; Boireau, S.; Jariel-Encontre, I.; Piechaczyk, M.; Bertrand, E.; Tazi, J.; et al. A novel role for PA28γ-proteasome in nuclear speckle organization and SR protein trafficking. Mol. Biol. Cell 2008, 19, 1706–1716. [Google Scholar] [CrossRef] [PubMed]
  186. Zhang, Z.; Zhang, R. Proteasome activator PA28γ regulates p53 by enhancing its MDM2-mediated degradation. EMBO J. 2008, 27, 852–864. [Google Scholar] [CrossRef] [PubMed]
  187. Liu, J.; Yu, G.; Zhao, Y.; Zhao, D.; Wang, Y.; Wang, L.; Li, L.; Zeng, Y.; Dang, Y.; Wang, C.; et al. REGγ modulates p53 activity by regulating its cellular localization. J. Cell. Sci. 2010, 123, 4076–4084. [Google Scholar] [CrossRef] [PubMed]
  188. Li, L.; Zhao, D.; Wei, H.; Yao, L.; Dang, Y.; Amjad, A.; Xu, J.; Liu, J.; Guo, L.; Li, D.; et al. REGγ deficiency promotes premature aging via the casein kinase 1 pathway. Proc. Natl. Acad. Sci. USA 2013, 110, 11005–11010. [Google Scholar] [CrossRef] [PubMed]
  189. Wan, Z.X.; Yuan, D.M.; Zhuo, Y.M.; Yi, X.; Zhou, J.; Xu, Z.X.; Zhou, J.L. The proteasome activator PA28γ, a negative regulator of p53, is transcriptionally up-regulated by p53. Int. J. Mol. Sci. 2014, 15, 2573–2584. [Google Scholar] [CrossRef] [PubMed]
  190. Realini, C.; Jensen, C.C.; Zhang, Z.; Johnston, S.C.; Knowlton, J.R.; Hill, C.P.; Rechsteiner, M. Characterization of recombinant REGα, REGβ, and REGγ proteasome activators. J. Biol. Chem. 1997, 272, 25483–25492. [Google Scholar] [CrossRef] [PubMed]
  191. Hoffman, L.; Pratt, G.; Rechsteiner, M. Multiple forms of the 20S multicatalytic and the 26S ubiquitin/ATP-dependent proteases from rabbit reticulocyte lysate. J. Biol. Chem. 1992, 267, 22362–22368. [Google Scholar] [PubMed]
  192. Blickwedehl, J.; Agarwal, M.; Seong, C.; Pandita, R.K.; Melendy, T.; Sung, P.; Pandita, T.K.; Bangia, N. Role for proteasome activator PA200 and postglutamyl proteasome activity in genomic stability. Proc. Natl. Acad. Sci. USA 2008, 105, 16165–16170. [Google Scholar] [CrossRef] [PubMed]

Share and Cite

MDPI and ACS Style

Vigneron, N.; Van den Eynde, B.J. Proteasome Subtypes and Regulators in the Processing of Antigenic Peptides Presented by Class I Molecules of the Major Histocompatibility Complex. Biomolecules 2014, 4, 994-1025. https://doi.org/10.3390/biom4040994

AMA Style

Vigneron N, Van den Eynde BJ. Proteasome Subtypes and Regulators in the Processing of Antigenic Peptides Presented by Class I Molecules of the Major Histocompatibility Complex. Biomolecules. 2014; 4(4):994-1025. https://doi.org/10.3390/biom4040994

Chicago/Turabian Style

Vigneron, Nathalie, and Benoît J. Van den Eynde. 2014. "Proteasome Subtypes and Regulators in the Processing of Antigenic Peptides Presented by Class I Molecules of the Major Histocompatibility Complex" Biomolecules 4, no. 4: 994-1025. https://doi.org/10.3390/biom4040994

Article Metrics

Back to TopTop