Abstract
During malignant transformation and cancer progression, tumor cells face both intrinsic and extrinsic stress, endoplasmic reticulum (ER) stress in particular. To survive and proliferate, tumor cells use multiple stress response pathways to mitigate ER stress, promoting disease aggression and treatment resistance. Among the stress response pathways is ER-associated degradation (ERAD), which consists of multiple components and steps working together to ensure protein quality and quantity. In addition to its established role in stress responses and tumor cell survival, ERAD has recently been shown to regulate tumor immunity. Here we summarize current knowledge on how ERAD promotes protein degradation, regulates immune cell development and function, participates in antigen presentation, exerts paradoxical roles on tumorigenesis and immunity, and thus impacts current cancer therapy. Collectively, ERAD is a critical protein homeostasis pathway intertwined with cancer development and tumor immunity. Of particular importance is the need to further unveil ERAD’s enigmatic roles in tumor immunity to develop effective targeted and combination therapy for successful treatment of cancer.
Keywords: Cancer, ERAD, tumor immunity, ER stress, immune cells
Introduction
In eukaryotes, approximately one-third of all proteins, including those important for immunity, are synthesized and folded in the endoplasmic reticulum (ER) (Jahn and Radford 2005; Oakes and Papa 2015). Physiological and pathological variations in the cellular environment often lead to the accumulation of misfolded and unfolded proteins in the ER, referred to as ER stress (Ma and Hendershot 2004). To restore ER homeostasis, cells have evolved two major protein quality control systems: the unfolded protein response (UPR) and ER-associated degradation (ERAD) (Chen et al. 2011). The UPR is triggered upstream of ERAD to restore protein homeostasis by transiently reducing protein translation, while selectively upregulating chaperone proteins to enhance protein folding and degradation (Janssens et al. 2014). The primary responsibility of ERAD is the elimination of misfolded and unfolded proteins to ensure protein quality. In addition, ERAD also regulates the quantity of properly folded proteins, including certain enzymes and lipid carriers (Fisher and Ginsberg 2002; Hampton 2002). Due to its crucial role in controlling both protein quality and quantity, altered ERAD function has been linked to numerous diseases, including neurodegenerative disorders, metabolic diseases, autoimmune diseases, and cancer (Guerriero and Brodsky 2012; Chen et al. 2018).
A critical component for cancer initiation and progression is the tumor microenvironment (TME), which is composed of tumor cells, immune cells, blood, and lymphatic vessels, as well as the many other types of cells (Junttila and de Sauvage 2013; Quail and Joyce 2013). The TME is a battlefield where immune cells struggle to gain control over the growth of tumor mass through antitumor immune responses. As the cycle of tumor regression and regrowth progresses, the TME becomes increasingly hypoxic and acidotic (Vaupel and Multhoff 2017). These adverse conditions thus heavily burden tumor and immune cells with ER stress (Cubillos-Ruiz et al. 2017). The increased ER stress impacts the proliferation and survival of tumor cells, as well as the activation and function of immune cells, thereby modulating tumor immunity, which is the process in which immune cells react to tumors (Janssens et al. 2014; Bettigole and Glimcher 2015; Grootjans et al. 2016; Cubillos-Ruiz et al. 2017).
To survive and proliferate in the harsh microenvironment, tumor cells activate the UPR to enhance ERAD for accelerated degradation of misfolded/unfolded proteins (Yadav et al. 2014). Due to the dependence of tumor cells on ERAD, a growing number of small-molecule inhibitors targeting ERAD have been developed as potential anticancer therapeutics (Tax et al. 2019). Interestingly, emerging evidence has established the role of ER stress and ERAD in regulating immunity, including ER stress induced by cancer treatment (Cubillos-Ruiz and Glimcher 2016; Cha et al. 2018). Hence, it is crucial to gain a comprehensive understanding of how ERAD impacts tumor immunity to guide both therapeutic development and cancer treatment. Here we present an overview of each individual step of the ERAD pathway and summarize the role of ERAD in immune cell development and function, as well as antigen presentation and antitumor immune responses. Finally, we discuss how small-molecule inhibitors, including standard cytotoxic chemotherapy and targeted therapy, may impact ERAD and tumor immunity.
ERAD, stepping down to degradation
ERAD research began in the 1980s when it demonstrated that unassembled membrane proteins in the ER were degraded (Lippincott-Schwartz et al. 1988; Bonifacino et al. 1990). Subsequent biochemical and genetic approaches led to the identification of specific ERAD components, spanning from yeast to metazoan (Table 1). In sequential steps, these components work together in this highly conserved pathway to recognize, retrotranslocate, and ubiquitinate proteins for proteasome-mediated degradation (Figure 1 and Table 1).
Table 1.
Components and substrates in the ERAD pathway.
Components/complexes Human (yeast) | Substrates | Function(s) | References |
---|---|---|---|
Substrate recognition ERMan1 (Mns1p) |
ASGPR H2a | Glycan trimming | Avezov et al. (2008) |
EDEM1, EDEM2, EDEM3 (Htm1p) | NHK AAT variant | Glycan trimming | Hosokawa et al. (2001), Hirao et al (2006), Hosokawa et al. (2010), Hotamisligil (2010), and Ninagawa et al. (2014) |
GI/GII | Glycan trimming | D’Alessio and Dahms (2015) | |
OS-9 (Yos9p) | NHK AAT variant | Glycan binding | Bhamidipati et al. (2005), Szathmary et al. (2005), and Christianson et al. (2008, 2011) |
XTP3-B | Glycan binding | Christianson et al. (2011) and Tyler et al. (2012) | |
GRP170 | misfolded glycosylated client NHK | Nucleotide exchange factor | Inoue and Tsai (2016) |
BiP (Kar2p) | nonsecreted Igκ light chain | Substrate recognition and recruitment | Okuda-Shimizu and Hendershot (2007) |
PDI (Pdi1) | glyco-pro-α-factor; BACE457 | Disulfide bonds rearrangement | Gillece et al. (1999), Molinari et al. (2002) |
ERdj5-EDEM1-BiP | Substrate recognition | Ushioda et al. (2008) | |
UGGT | Glycoprotein glucosyltransferase | D’Alessio et al. (2010) | |
GRP94 | NHK AAT variant | Chaperone | Christianson et al. (2008) |
ERdj4 | surfactant protein C | Co-chaperone | Dong et al. (2008) |
ERdj5 (Scj1p) | NHK AAT variant | Co-chaperone | Dong et al. (2008) and Ushioda et al. (2008) |
SEC22b (Sec22) | ER-phagosome traffic mediator | Alloatti et al. (2017) | |
ERp57 | MHC-I heavy chain | Glycoprotein folding | Zhang, Baig et al. (2006) |
Retrotranslocation and Ubiquitination Derlin-1,2,3 (Der1p) | US11-associated MHC-I heavy chain | Retrotranslocation channel | Lilley and Ploegh (2004) and Ye et al. (2004) |
SEL1L-OS9-UBC6E-AUP1 | US11-associated MHC-I heavy chain | Substrate recognition and dislocation | Mueller et al. (2008) |
SEL1L (Hrd3p) | Igμ chain; US11-associated MHCI heavy chain | Substrate recognition and recruitment | Cattaneo et al. (2008) and Mueller et al. (2008) |
SEC61 (Sec61p) | Retrotranslocation channel | Koopmann et al. (2000) | |
RMA1 | mutant CFTR | E3 ubiquitin ligase | Morito et al. (2008) |
TMEM129 | US11-associated MHC-I heavy chain | E3 ubiquitin ligase | van de Weijer et al. (2014) and van den Boomen et al. (2014) |
TMEM129-Derlin1 | US11-associated MHC-I heavy chain | Substrate ubiquitination and translocation | van de Weijer et al. (2014) and van den Boomen et al. (2014) |
HRD1-UBE2J1 | non-β2m bound MHC I heavy chain | Substrate ubiquitination | Burr et al. (2011) |
UBE2G2 (Ubc7p) | HMGCR | E2-conjugating enzyme | Miao et al. (2010) |
SKP1-Cullin-F-box | Intergrin-β1 | E3 ligase complex | Yoshida et al. (2005) |
UBE2J1 (Ubc6p) | TCRα; mutant CFTR | E2-conjugating enzyme | Lenk et al. (2002) and Miao et al. (2010) |
CHIP | mutant CFTR | U-box ubiquitin ligase | Younger et al. (2006) |
TRC8 | US2-associated MHC-I heavy chain | E3 ubiquitin ligase | Stagg et al. (2009) |
HRD1 (Hrd1p) | unassembled Igμ; FAS; BLIMP-1; Nrf1 | E3 ubiquitin ligase | Cattaneo et al. (2008), Tsuchiya et al. (2011), Yang et al. (2014), and Kong et al. (2016) |
SEL1L-HRD1 | IRE1α; P53; Pre-BCR; pro-AVP | Substrate ubiquitination and retrotranslocation | Yamasaki et al. (2007), Sun et al. (2015), Ji et al. (2016), and Shi et al. (2017) |
HERP (Usa1p) | TCRα | Substrate ubiquitination | Schulze et al. (2005) |
GP78 | TCRα | E3 ubiquitin ligase | Zhang, Xu, et al. (2015) |
TEB4 (Doa10) | E3 ubiquitin ligase | Hassink et al. (2005) | |
WDR20 | TCRα | Adaptor Protein | Ju et al. (2018) |
Dislocation and Degradation UBXD8 (Ubx2p) | ApoB100 | P97 recruitment | Suzuki et al. (2012) |
NGly (Png1) | glycosylated RTA | Deglycosylating enzyme | Kim et al. (2006) |
RAD23 (Rad23) | glycosylated RTA | Ubiquitin binding protein | Kim et al. (2006) |
AUP1 (Cue1p) | HMGCR | UBE2G2 recruitment | Jo et al. (2013) |
Ataxin-3 | TCRα; CD3δ | Deubiquitinating enzyme | Zhong and Pittman (2006) |
P97-UFD1-NPL4 (Cdc48p-Ufd1p-Npl4p) | Substrates dislocation | Ye et al. (2001) | |
HERP (Usa1p) | CD3δ | Shuttle factors | Kim et al. (2008) |
YOD1 | TCRα; AAT | Deubiquitinating enzyme | Ernst et al. (2009) |
BAG6-UBL4A-TRC35 | Shuttle factors | Xu et al. (2012) | |
USP13 | TCRα | Deubiquitinating enzyme | Sowa et al. (2009) |
Figure 1.
Schematic illustration of the ERAD pathway. The first step of ERAD is substrate recognition. BiP recognizes and recruits nonglycosylated misfolded or unfolded proteins while ER resident lectins recognize glycosylated substrates. Substrate recognition is followed by retrotranslocation of substrates from the ER to the cytosol through translocon channels. The P97-UFD1-NPL4 complex facilitates the retrotranslocation of substrates that bind to P97. This complex serves as a platform for ubiquitination, which is carried out by E1 ubiquitin-activating enzymes, E2 ubiquitin-conjugating enzymes, and E3s such as HRD1. The polyubiquitinated protein is then degraded by the proteasome in the cytosol. A colored version of the figure is available online.
i. Substrate recognition
Substrate recognition, the first step in ERAD, must be precisely controlled to ensure accurate detection of target proteins (Figure 2) (Ruggiano et al. 2014; Moon et al. 2018). Initially, polysomes on the ER membrane synthesize proteins that enter the ER lumen in an unfolded state. Once inside the ER lumen, these polypeptides encounter an oxidizing and calcium-rich microenvironment, wherein N-linked glycosylation modification occurs and disulfide bonds form as proteins mature (Ma and Hendershot 2004). Properly folded polypeptides are subsequently shuttled to their respective destinations (Denic et al. 2006; Liu and Ye 2011; Walter and Ron 2011). If the protein fails to fold properly, it will bind to mannose-specific lectins that trim the terminal mannose residues from the core glycans, thereby marking this misfolded/unfolded protein for ERAD (Figure 2) (Xu and Ng 2015). Examples of these lectins include ER degradation-enhancing α-mannosidase-like lectin 2 (EDEM2) and ER mannosidase-I (ERmanI), which trim the first mannose, followed by further trimming of the glycan by EDEM1/EDEM3 (Hirao et al. 2006; Hosokawa et al. 2010; Ninagawa et al. 2014). The ER-resident lectin, Yeast OS-9 homolog (Yos9), or human osteosarcoma amplified 9 (OS-9) and XTP3-transactivated gene B (XTP3-B)/Erlectin, will then target misfolded/unfolded proteins for retrotranslocation (Szathmary et al. 2005; Christianson et al. 2011). Chaperone proteins, including binding immunoglobulin protein (BiP) and other members of the heat shock protein 70 (HSP70) family, can bind to hydrophobic patches of an unfolded substrate (Hammond and Helenius 1994). This prevents the aggregation of both glycosylated and nonglycosylated substrates (Hammond and Helenius 1994).
Figure 2.
Glycosylation-dependent recognition and targeting of ERAD substrates. Upon entry into the ER, the misfolded/unfolded proteins are often modified by core glycans, GlcNAc2Man9Glc3, which is composed of three glucose (Glc3), nine mannose (Man9), and two N-Acetylglucosamine (GlcNAc2). The addition of the glycans occurs at the Asn residue within the Asn-X-Ser/Thr motif. GI and GII remove two glucoses from the glycans. The resulting monoglucosylated proteins are then recognized by the ER chaperones, calnexin or CRT, which facilitate the folding of the immature proteins. Meanwhile, GI will remove the final glucose from the glycans. If proper folding occurs, the protein will then be modified by ERmanI that will remove the outer mannose. The mature protein achieving its native conformation will exit the ER. However, if the protein is improperly folded, UGGT will recognize and glucosylate this protein, allowing its reentry into the folding cycle again. If this protein fails to achieve its native conformation, it will undergo mannose trimming by EDEM and ERmanI, and subsequently targeted by OS-9 or XTP3-B for ERAD. GI: Glucosidase-I; GII: Glucosidase-I; CRT: calrecticulin; ERmanI: ER mannosidase-I; OST: oligosaccharyl transferase; UGGT: UDP-glucose: glycoprotein glucosyltransferase; and EDEM: ER degradation-enhancing α-mannosidase-like lectins.
In contrast to glycosylated proteins, the process of nonglycosylated substrate recognition and targeting is less clear. Nonglycosylated substrates are primarily recognized and recruited by BiP (Plemper et al. 1997; Skowronek et al. 1998). Additionally, mammalian non-glycosylated proteins can be recognized by EDEM2/OS-9 or EDEM1 (Cormier et al. 2009; Tang HY et al. 2014). Homocysteine-induced ER protein (HERP), a transmembrane protein, directs nonglycosylated substrates to the proteasome for degradation (Okuda-Shimizu and Hendershot 2007). In yeast, nonglycosylated substrates are often recognized by U1Snp1-asscociating 1 (Horn et al. 2009; Kanehara et al. 2010); however, some of them can also be directly recognized by Yos9 for degradation (Jaenicke et al. 2011).
ERAD utilizes the same general machinery to degrade misfolded/unfolded and properly folded proteins; however, the process of substrate recognition and targeting differs (Ruggiano et al. 2014). Each properly folded substrate tends to have its own specific adaptor although most of these adaptors are yet to be identified (Ruggiano et al. 2014). One interesting example is the identification of Derlin-related iRhom proteins as adaptors for the degradation of epidermal growth factor receptor (EGFR) ligands in regulation of sleep (Zettl et al. 2011). Additionally, cellular signaling also controls the recognition of certain enzymes, such as those in lipid metabolism and sterol biosynthesis (Ruggiano et al. 2014). One example is the degradation of 3-hydroxy-3-methyl-glutaryl-CoA reductase (HMGR), a rate-limiting enzyme in cholesterol synthesis. Under low levels of sterol, HMGR does not bind to its adaptor, ER membrane protein insulin-induced gene 1(INSIG-1) or 2, and thus remains stable, serving as a precursor for sterol synthesis. Upon active sterol synthesis, the concentration of intermediate 24,25-dihydrolanosterol increases, enabling the binding of HMGR to INSIG-1 or -2 and the delivery of HMGR to its E3 ligase complex for ubiquitination and degradation (Song et al. 2005).
ii. Substrate retrotranslocation and ubiquitination
After recognition, substrates will be retrotranslocated (or dislocated) to the cytosol depending on their specific location in the ER, a step tightly coupled with substrate ubiquitination. If the substrate is in the lumen, the complete polypeptide will be retrotranslocated. For substrates located in the membrane, only certain domains will be retrotranslocated. Therefore, ubiquitination of the luminal substrates often occurs at later stages of retrotranslocation, whereas ubiquitination of most membrane substrates takes place simultaneously with retrotranslocation. Specific retrotranslocation channel proteins or certain E3 ligases can mediate substrate retrotranslocation. In yeast, Sec61 is a candidate channel protein for retrotranslocation, working within a multiprotein ensemble (Lilley and Ploegh 2004; Ravid et al. 2006; Kohlmann et al. 2008). The Derlin family of proteins serve as alternative candidate components for retrotranslocation (Jarosch et al. 2002; Richly et al. 2005), and there are three in mammalians, Derlin-1, 2, and 3 (Wahlman et al. 2007; Mehnert et al. 2014). Suppressor enhancer Lin12 1 like (SEL1L), an ortholog of yeast HMG-CoA reductase degradation 3 (Hrd3p), is essential for retrotranslocation of many ERAD substrates in human cells (Mueller et al. 2006). In chickens, the SEL1L-dependent substrate, ERAD-L, requires Derlin 2/3 and Herp 1/2 for its degradation (Sugimoto et al. 2017).
The P97 valosin-containing protein (VCP) is a type II AAA+protein family of ATPases, critical for substrate retrotranslocation from yeast to mammals (DeLaBarre and Brunger 2003; Huyton et al. 2003; Meyer 2012; Meyer et al. 2012). The N-terminal domain of P97 binds to substrates, while the C-terminal domain interacts with its adaptor proteins, mediated through the P97/VCP-interacting motif, P97/VCP-binding region, or SHP box (Meyer et al. 2012). Among P97 cofactors, the ubiquitin fusion degradation protein 1 (UFD1) and nuclear protein localization protein 4 (NPL4) form one of the major retrotranslocation complexes with P97 (i.e., the P97-UFD1-NPL4 complex) (Ye Y et al. 2001; Rabinovich et al. 2002). Because P97 cofactors also possess ubiquitin-binding domains, they can directly interact with ubiquitinated substrates. The P97 complexes not only retrotranslocate substrates through the ER membrane, but also provide a platform for various ERAD components, such as the E3 ligases, to interact with and subsequently ubiquitinate substrates (Ye et al. 2004; Liang et al. 2006; Greenblatt et al. 2011; Suzuki et al. 2012; Christianson and Ye 2014).
Ubiquitination is carried out through a series of sequential enzymatic reactions, facilitated by the ubiquitin-activating enzyme (E1), then the ubiquitin-conjugating enzyme (E2), and finally the ubiquitin ligase (E3). Ubiquitin is first covalently conjugated to a reactive cysteine residue of E1 and then transferred to a catalytic cysteine of E2. Finally, E3s facilitate the transfer of ubiquitin from E2 to a lysine residue or another ubiquitin moiety on ERAD substrates (Sommer and Jentsch 1993; Kikkert et al. 2004; Hassink et al. 2005). This reaction can be repeated several times to form polyubiquitin chains. Occasionally, an E4 ligase, a specialized ubiquitin ligase such as UFD2, works after E3s to elongate ubiquitin chains of oligoubiquitinated proteins to generate polyubiquitinated conjugates (Hoppe 2005). This suggests that the polyubiquitin appendage must reach a crucial length before a substrate can be retrotranslocated (Mayer et al. 1998).
A number of E3s involved in ERAD have been identified. Transmembrane E3s include HRD1, glycoprotein 78 (GP78), membrane-associated really interesting new gene (RING) finger protein 6, and RING finger protein 5 (Fang et al. 2001; Nadav et al. 2003; Kikkert et al. 2004; Hassink et al. 2005; Younger et al. 2006; Christianson and Ye 2014; Jeon et al. 2015; Tomati et al. 2015). Their functions are described in later sections. Cytosolic E3s include PARKIN, the C-terminus of HSP70-interacting protein (CHIP), ubiquitin protein ligase E3 component N-recognin 1/2 (UBR1/2), SMAD ubiquitination regulatory factor 1, E6-associated protein (E6-AP), and neuregulin receptor degradation pathway protein 1/fetal lever ring finger (Cummings et al. 1999; Meacham et al. 2001; Murata et al. 2001; Qiu and Goldberg 2002; Tsai et al. 2003; Nillegoda et al. 2010; Guo et al. 2011). Among them, CHIP, UBR1/2, PARKIN, and E6-AP can specifically recognize chaperone-bound misfolded proteins for proteasome-mediated degradation (Cummings et al. 1999; Murata et al. 2001; Tsai et al. 2003; Nillegoda et al. 2010). The SCF complexes, another major family of cytosolic E3s, are composed of Skp1, Cullin1, and specific F-box protein (Fbx, e.g., Fbx2, Fbx6, and β-transducin repeat containing proteins 1 and 2). This class of E3s can recognize and target certain glycoprotein substrates for degradation (Yoshida et al. 2003; 2005; Gong B et al. 2010; Magadan et al. 2010).
In yeast, E3s can function in cytosolic (ERAD-C), luminal (ERAD-L), and membrane (ERAD-M) pathways based on the location of their substrates (Deshaies et al. 1991; Wiertz et al. 1996; Vashist and Ng 2004). For instance, the degradation of alpha 10 (Doa10) is an E3 that targets substrates with lesions in the cytosolic domain, whereas the E3 Hrd1 can target the substrates with lesions in the transmembrane or luminal domain for degradation (Vashist and Ng 2004; Carvalho et al. 2006). Thus far, the three ERAD pathways mentioned above have only been identified in yeast. Mammals possess a more elaborate repertoire of ERAD components. Another unique property of yeast ubiquitin ligases is that they can form multispanning membrane protein complexes that perform a variety of functions including substrate recognition, targeting, and retrotranslocation. For instance, Hrd1 possesses retrotranslocation function (Schoebel et al. 2017). Autoubiquitination of Hrd1 on lysine residues of its RING-finger domain allows misfolded substrates to move across the membrane without the requirement of P97 ATPase (Baldridge and Rapoport 2016). Allowing a single E3 enzyme to harbor both ubiquitination and retrotranslocation activities may provide the most efficient way to target ERAD substrates for degradation in lower species (Bays et al. 2001). HRD1 and GP78 in mammals are homologous to the E3 Hrd1 in yeast. HRD1 ubiquitinates glycosylated substrates in a SEL1L-dependent manner, which can in turn regulate the stabilization of HRD1 (Gardner RG et al. 2000; Carvalho et al. 2006; Gauss et al. 2006; Christianson et al. 2008; Sun S et al. 2014). GP78 functions downstream of HRD1 through association with the human BCL2-associated athanogene 6 (BAG6) chaperone complex to prevent substrate aggregation, thus ensuring the efficient retrotranslocation of substrates for degradation (Zhang, Xu, et al. 2015).
iii. Substrate degradation
Once polyubiquitinated, substrates are extracted from the membrane prior to or during proteasome-mediated degradation. Interestingly, a substantial percentage of proteasome complexes reside at the surface of the ER membrane, ideally positioned to retrotranslocate, receive, and degrade substrates (Rivett 1993; Verma et al. 2000; Hollien and Weissman 2006). This begs the question: how do proteasome-ready substrates exist in ERAD? Quite often, P97 serves as a bridge to link the retrotranslocated substrates with cofactors in cytoplasm. For instance, the cytosolic N-glycanase 1 directly binds to P97 and cleaves N-linked glycans from retrotranslocated substrates, presenting these substrates to the proteasome for degradation (Kim et al. 2006; Bettigole and Glimcher 2015). P97 alone can recognize nonubiquitinated substrates, whereas polyubiquitinated substrates require both P97 and its cofactor UFD1-NPL4 for cytosolic dislocation, a process to export substrates from the ER to the cytosol (Ye et al. 2003). Deubiquitylation occurs during retrotranslocation or immediately prior to degradation, and is mediated by deubiquinating enzymes (DUBs) (Martinon 2012). Deubiquitination may be essential for substrates to enter the proteasome or escape from degradation. Example of DUBs, which associate with P97 directly or indirectly, include the ovarian tumor family of deubiquitinating enzyme 2, valosin-containing protein P97/P47 complex-interacting protein P135, ubiquitin-specific protease 13, and Ataxin-3 (Uchiyama et al. 2002; Wang et al. 2006; Ernst et al. 2009; Sowa et al. 2009).
In order to be degraded by the proteasome, misfolded substrates with hydrophobic motifs or transmembrane domains must remain soluble. The BAG6/ubiquitin-like protein 4 A (UBL4A)/transmembrane domain recognition complex 35 (TRC35) complex aids in this process. BAG6 maintains polypeptide solubility through its chaperone-like activity and subsequently transfers the polypeptides to the proteasome (Wang et al. 2011). The interaction of the BAG6-UBL4A-TRC35 complex with the co-chaperone small glutamine-rich TPR-containing protein further prevents the aggregation of ERAD substrates (Xu et al. 2012). Only properly folded proteins are transported to their destination through the secretory pathway, raising the question: how are they transported from the ER? It is likely that competition exists between protein transportation and ERAD selection machinery, suggesting that the decision to transport or degrade substrates may depend on cell type, stress, secretory-protein load, and/or signaling pathways.
ERAD impinges on immune cells
The adaptive and innate immune responses of our body must be tightly regulated, which is primarily carried out by different immune cells: T cells, B cells, natural killer (NK) cells, dendritic cells (DCs), macrophages, and neutrophils. ERAD plays a crucial role in regulating immune responses to avoid self-reactive (i.e., autoimmunity) or insufficient immune responses (i.e., immunodeficiency or immunosuppression) (Kim et al. 2011; Gong et al. 2014; Yang et al. 2014). ERAD regulates immune cell development by controlling the quantity of both T-cell receptor (TCR) and pre-B-cell receptor (pre-BCR) complexes (Ho et al. 2000; Ji et al. 2016). Additionally, ERAD is important for proper antigen assembly and cytokine secretion (Xu et al. 2016).
T cells
T cells are a type of lymphocytes that function in adaptive immunity and play a key role in the defense against pathogens and tumors. In mammals, the bone marrow is the primary site of hematopoiesis where both T and B lymphoid precursors originate, whereas the thymus is where early thymic progenitor cells seed and develop. A stepwise selection proceeds inside the thymus, starting as double negative (DN, CD4−CD8−) cells, then double positive (DP, CD4+CD8+) cells, and finally single positive (SP, CD4+ or CD8+) cells. During this long journey, the unique composition of the TCR complex identifies each T-cell clone and marks the self-responsive population for elimination. The TCR complex consists of heterodimer of TCRα/β together with homodimers or heterodimers of CD3γ, δ, ε, and ζ chains (Chan et al. 1994). The expression of the TCRβ chain on the DN thymocytes dictates their development into αβ T cells (Starr et al. 2003; Takahama 2006). The DN thymocytes first express the pre-TCR complex, which contains pre-α rather than mature α chains (Germain 2002). This complex is subsequently replaced by the mature TCR complex to facilitate positive and negative selection of thymocytes. Specifically, thymocytes expressing TCR complexes with the ability to bind to the major histocompatibility complexes (MHC) will survive (i.e., positive selection); conversely, thymocytes expressing TCR complexes that have strong affinity to self-antigens will be eliminated (i.e., negative selection). Mature SP T cells that survive the selection will exit the thymus and participate in the adaptive immune response (Germain 2002; von Boehmer and Melchers 2010).
Since the proper expression and assembly of the TCR complex on T cells is a prerequisite for their development in the thymus, the quality and quantity of TCR subunits must be properly controlled. TCR subunits that fail to assemble into the heteromeric complex will be degraded through the ERAD pathway (Bonifacino et al. 1989; Yang et al. 1998). Among these subunits, TCRα and CD3δ are particularly susceptible to ERAD. Recent studies have identified the specific components required for ERAD of TCR subunits. For instance, the mannose-trimming of TCRα is mediated by the ER lectin EDEM3 together with an ER-resident oxidoreductase ER-resident protein 46 (Yu S et al. 2018). Moreover, the ubiquitination of TCRα is essential for its retrotranslocation, which is a multi-step process regulated by P97 (Huppa and Ploegh 1997; Yu H and Kopito 1999; Moore et al. 2013). Interestingly, impaired UFD1 or NPL4 function does not impact TCRα levels, suggesting the P97-UFD1-NPL4 complex does not regulate its retrotranslocation (Nowis et al. 2006). The ER-localized E3 HRD1 mediates the ubiquitination of TCRα (Ishikura et al. 2010), whereas ubiquitin specific peptidase 19, a human DUB, can reverse this process (Hassink et al. 2009). Recently, WD repeat-containing protein 20 (WDR20), a novel regulator of ERAD, was found to facilitate TCRα ubiquitination by mediating the interaction of P97 and TCRα (Ju et al. 2018). Although TCRα and CD3δ are both TCR subunits, their ERAD components differ. The E3 GP78 facilitates the degradation of CD3δ through its physical interaction with P97 (Zhong et al. 2004); however, deletion of P97 only impacts the degradation of TCRα but not CD3δ (Wojcik et al. 2006; Lass et al. 2007). Instead, the degradation of CD3δ was mediated by the E3 Ring finger protein 2 (Lerner et al. 2007).
Despite the above knowledge, how ERAD specifically impacts T-cell development remains elusive. Using T-cell-specific Hrd1 knockout mice, a recent study showed that HRD1 depletion markedly reduced all T-cell populations, including CD4+ and CD8+ SP T cells (Xu et al. 2016). This observation is intriguing and suggests the contribution of the E3 HRD1 to the development of CD4+ and CD8+ T cells; however, the specific mechanism remains unclear. Additionally, the absolute number of T regulatory cells (Tregs) also decreased, likely due to fewer T cells in the spleen of Hrd1 knockout mice (Xu et al. 2016). Interestingly, HRD1 was shown to maintain the stability of Tregs through suppressing ER stress responses (Xu et al. 2019). Moreover, loss of HRD1 leads to a blockade in T-cell proliferation and differentiation, as well as interleukin-2 (IL-2) production (Xu et al. 2016). Despite these advances, the contribution of other ERAD components to T-cell immunity has yet to be elucidated. The use of animal models in which specific ERAD components are depleted could be particularly useful in defining their contribution to T-cell development and function.
B cells
In contrast to T cells, B cells develop in the bone marrow and primarily function in humoral adaptive immunity (LeBien and Tedder 2008). B cell precursors in the bone marrow undergo a multi-step developmental process, starting as B cell progenitors, then precursors, and finally immature B cells (Cambier et al. 2007). During early B-cell development, the SEL1L-HRD1 complex mediates ERAD of the pre-B-cell receptor (pre-BCR) complex, which is critical for the transition of large to small pre-B cells (Ji et al. 2016). Upon stimulation of thymus-independent antigens, such as lipopolysaccharides (LPS), B cells proliferate and differentiate into plasma cells that primarily secrete immunoglobulin M (IgM) antibodies (Shih et al. 2002; Nutt et al. 2015). To cope with this output, B cells must drastically improve their protein quality control system and secretory machinery (Gass et al. 2002); therefore, they enhance ERAD functions to reduce the accumulation of misfolded/unfolded Ig in the ER that results from escalating Ig synthesis. One such example is the upregulation of SEL1L and HRD1 in LPS-stimulated B cells, consistent with the abundant expression and secretion of IgM (Cattaneo et al. 2008).
IgM consists of two forms: a membrane-bound monomer and a secreted pentamer. The pentameric IgM consists of five monomers held together by J chains and disulfide bonds (Kubagawa et al. 2019). Similar to TCRα and CD3δ, unassembled IgM heavy chain (Igμ) is also degraded by ERAD (Ho et al. 2000). Igμ and J chains trimmed by ERManI are targeted to the cytosol for ERAD, as the use of Kifunensine, an inhibitor of ERManI, prevents degradation of Igμ and dislocation and degradation of the J chain (Fagioli and Sitia 2001). Other ERAD components, such as SEL1 and HRD1, are also involved in degradation of unassembled Igμ (Cattaneo et al. 2008). Additionally, unfolded/unassembled IgG is degraded by a Herp-dependent ERAD pathway (Lee J et al. 2012). In addition to impacting B-cell immunity through controlling the quality of Ig, HRD1 can also regulate other critical factors to affect B-cell activation. This is supported by a study that shows the death receptor FAS being degraded by HRD1 to sustain B-cell survival (Kong et al. 2016). The above evidence demonstrates the critical contribution of ERAD to B-cell development and activation. Further understanding of how ERAD controls plasma cell survival could guide the treatment of multiple myeloma, a disease with abnormal expansion of plasma cells.
Other immune cells
Recent studies also reveal the regulatory role of ERAD on other immune cells, such as DCs, macrophages, and NK cells. DCs are professional antigen-presenting cells (APCs), which play a critical role in initiating adaptive immune responses, as well as maintaining the tolerance of self-antigens (Heath and Carbone 2001; Mellman 2013). DCs can engulf pathogen-infected or malignantly transformed cells, a process that initiates exogenous antigen presentation through MHC class I molecules (MHC-I) to CD8+ T cells or MHC class II molecules (MHC-II) to CD4+ T cells (Joffre et al. 2012). B lymphocyte-induced maturation protein 1 (BLIMP-1), a transcriptional repressor, can suppress systemic lupus erythematosus in mice by maintaining tolerogenic function of DCs (Kim et al. 2011). Using mice with a DC-specific deletion of Blimp-1, a study showed that BLIMP-1 can transcriptionally repress the expression of MHC-II on the surface of DCs (Kim et al. 2013). Moreover, enhanced expression of HRD1 through Toll-like receptor stimulation promotes MHC-II expression by ubiquitinating and degrading BLIMP-1 (Yang et al. 2014). This regulatory role of HRD1 only impinges on MHC-II but not on MHC-I, impairing the priming of CD4+ T cells while sparing CD8+ T cells (Yang et al. 2014). Loss of HRD1 function in DCs protects mice from experimental autoimmune encephalomyelitis, a direct evidence demonstrating a physiological role of HRD1 in immunity (Yang et al. 2014). Additionally, ERAD has a vital role in antigen cross-presentation, which will be discussed below in the antigen presentation section.
Macrophages are derived from monocytes and exert the first line immune responses against infections (Labonte et al. 2014). Macrophages can differentiate into two subtypes: M1 that possesses antitumor activity and M2 that is detrimental to the host, contributing to tumor growth, angiogenesis, and metastasis (Dumont et al. 2008; Niino et al. 2010; Alfano et al. 2013; Labonte et al. 2014). A recent in vitro study showed that overexpression of HERP, a shuttling factor that delivers ubiquitinated substrates to the proteasome for degradation, promotes IL-4-induced polarization and migration of M2 macrophages (Kim et al. 2008; Li Y et al. 2018). Lipoprotein lipase (LPL) is a protein guarding the entry of fatty acids into tissues (Williams 2008). SEL1L mediates LPL secretion and systemic lipid metabolism in adipocytes, myocytes, and macrophages by preventing aggregation of LPL and facilitating its ER exit (Sha et al. 2014). Despite these studies, further studies are needed to understand how the ERAD pathway regulates macrophage differentiation and functions.
NK cells play a vital role in immunosurveillance by directly inducing cytotoxic responses to tumor cells (Degli-Esposti and Smyth 2005). The recognition of human tumor cells by NK cells is mediated by the binding of NK-cell activated receptor CD226 to its ligand CD155 (Carlsten et al. 2009). Although there is no direct evidence showing the regulation of ERAD in NK cells, activation of UPR and ERAD in hepatocellular carcinoma cell (HCC) can suppress NK-mediated cytotoxicity. Activation of two branches of the UPR pathways, transcription factor 6 (ATF6) and inositol-requiring enzyme 1α (IRE1α), can enhance HRD1 expression in HCC. Increased levels of HRD1 promote degradation of CD155 in HCC, which further perturbs the cytotoxic activity of NK cells (Gong et al. 2014). Since these experiments are only performed in vitro, additional in vivo work is required to establish the role of HRD1 in the development and activation of NK cells.
ERAD in antigen presentation
Antigen presentation is a prerequisite for initiating T-cell mediated antitumor responses. Dysfunctions in antigen processing and presentation are common causes of immune evasion in cancer, creating barriers to T-cell based immunotherapies (de Charette et al. 2016). To initiate antitumor immune responses, peptides derived from tumor antigens must be presented on the surface of APCs by MHCs to effector T cells (Burgdorf et al. 2008). MHC-I are expressed by all nucleated cells, including tumor cells and APCs, primarily presenting antigens to CD8+ T cells (Vyas et al. 2008). MHC-II are predominantly expressed by professional APCs, such as DCs, B cells, and macrophages, mainly presenting antigens to CD4+ T cells (Jensen 2007). Endogenous antigen presentation, which occurs in all nucleated cells, is the presentation of self-antigenic peptides to the cell surface through MHC-I (Neefjes et al. 2011). Endogenous antigen presentation can also occur through MHC-II in certain cell types, such as DCs, tumor cells, and thymic epithelial cells (Roche and Furuta 2015). In a similar manner, APCs present extracellular antigens through MHC-II to CD4+ T cells, a process referred to as exogenous antigen presentation (Watts 2004). Cross-presentation is the process in which APCs present exogenous antigens to naïve CD8+ T cells through MHC-I, thus activating them to cytotoxic CD8+ T cells (Kurts et al. 2010). Despite little known about how ERAD regulates antigen presentation, accumulating evidence demonstrates that ERAD regulates degradation of MHC-I and translocation of exogenous antigens from phagosomes to the cytosol, a critical step for cross-presentation (Grotzke and Cresswell 2015).
i. ERAD in endogenous antigen presentation
Cancer cells present tumor antigens through endogenous antigen presentation. One of the key players in this process is MHC-I, which is composed of a heavy chain and an invariant light chain known as β2-microglobulin (β2 m) (Vyas et al. 2008). The MHC-I is assembled in the ER and loaded with peptides derived from tumor antigens through the following sequential steps (Figure 3(A)). Tumor antigens, including proteins, peptides, and polysaccharides, are degraded by the proteasome into small peptides, which are subsequently translocated into the ER by the transporter associated with antigen presentation (TAP) (Howard 1995). These peptides are typically composed of eight to sixteen amino acids, which exceeds the peptide length required for MHC-I association. Hence, aminopeptidase, another ER luminal component, will trim these peptides into an approximate length of eight amino acids for MHC-I loading (York et al. 2002; Cruz et al. 2017). In mice, there is only one such enzyme called ER aminopeptidase that is associated with antigen processing, whereas in humans, two such enzymes have been identified: ER aminopeptidase-1 (ERAP1) and 2 (ERAP2) (Saric et al. 2002; Serwold et al. 2002; Saveanu et al. 2005). Before MHC-I can load the peptides, they must be stabilized by ER chaperone proteins, such as calreticulin (CRT), ER-resident protein 57 (ERp57), protein disulfide isomerase, and tapasin. Once stabilized, TAP and tapasin help translocate the peptides into the ER and deliver them to MHC-I (Sadasivan et al. 1996). The fully assembled peptide-MHC-I complexes will then be transported via the Golgi from the ER to the cell surface (Vyas et al. 2008).
Figure 3.
Endogenous and exogenous antigen presentation by MHC-I. (A) MHC-I presentation of endogenous antigenic peptides in cancer cells begins with degradation of the antigen by the proteasome. The degraded peptides are then translocated into the ER lumen through TAP. In the lumen, the peptides are trimmed by ERAP1/2 and loaded onto MHC-I. The fully assembled peptide-MHC-I complex is transported via the Golgi to the cell surface for presentation to CD8+ T cells. When β2m is absent or downregulated, the MHC-I heavy chain will be removed from the ER through ERAD. (B) Exogenous antigens can be presented to T cells by antigen presenting cells through two major pathways. The first pathway, the vacuolar pathway, begins with the exogenous antigen internalized into a phagosome, where it is degraded by cathepsin S and then loaded onto MHC-I for transportation to the cell surface. The second pathway, the endosome pathway, begins with the antigen exiting the phagosome through SEC61 and P97 to the cytosol for proteasome-mediated degradation. From this point, the peptides will be transported to the cell surface following a path similar to (A). Alternatively, the peptides will be transported back to the phagosome by TAP and loaded onto MHC-I for transportation to the cell surface. TAP: transporter associated with antigenic presentation; ERAP: ER aminopeptidase associated with antigen processing.
Cancer cells often escape immunosurveillance due to defective antigen presentation. Newly synthesized MHC-I heavy chain must bind to β2 m to incorporate into the peptide-loading complex (PLC), comprised of TAP, tapasin, CRT, and ERp57 (Chapman and Williams 2010). Upon increased ER stress, cancer cells often downregulate MHC-I, TAP, tapasin, CRT, and/or ERp57, impairing the formation of PLC and endogenous antigen presentation (Leone et al. 2013). Reduced presentation of the peptide-MHC-I complexes is observed in approximately 20–60% of solid tumors, such as melanoma, lung, and breast cancer (Campoli and Ferrone 2008). When β2 m is absent or downregulated, the MHC-I heavy chain cannot fold properly and will be removed from the ER through ERAD (Hughes et al. 1997). Misfolded MHC-I heavy chain is recognized in the ER lumen by EDEM1, OS-9, and XTP3-B in a glycan-dependent manner (Burr et al. 2013), and then targeted by HRD1/SEL11L/UBE2J1(ubiquitin conjugating enzyme E2 J1) for ERAD (Burr et al. 2011). The loss of UBE2J1 impairs the ERAD of misfolded MHC-I heavy chain (Burr et al. 2011). The extraction of the misfolded heavy chain from the ER membrane requires the UFD1-NPL4-P97 complex (Burr et al. 2013). Disruption of the complex through UFD1 deletion restores MHC-I levels in the β2 m-depleted cells (Burr et al. 2013); however, it is unknown whether the dysfunction of the UFD1-NPL4-P97 complex can increase the presentation of tumor antigens. Antigenic peptides that fail to bind to MHC-I will be transported from the ER lumen into the cytosol for degradation. One way to degrade these peptides is through cytosolic peptidase; however, this enzyme can only partially degrade the peptides (Roelse et al. 1994). TAP can then translocate these peptides into the ER, allowing them to re-associate with MHC-I (Roelse et al. 1994). ERAD serves as an additional way to eliminate antigen peptides that fail to associate with MHC-I (Koopmann et al. 2000). Hence, enhanced ERAD function in tumor cells may promote the degradation of MHC-I and antigen peptides, thereby preventing the reassociation of peptides with MHC-I, impairing antigen presentation and promoting immune evasion.
In addition to MHC-I, endogenous antigens can also be presented by MHC-II through autophagy-mediated antigen processing (Schmid and Munz 2007). This allows the recognition of intracellular antigens by CD4+ T cells, a process particularly important for positive and negative selection of thymocytes (Kasai et al. 2009). MHC-II is expressed by APCs, as well as by a variety of cancer cells, including melanoma, glioma, breast, colorectal, ovarian, prostate, and nonsmall cell lung cancer (NSCLC) cells (Axelrod et al. 2019). Interestingly, tumor-associated MHC-II expression predicts a favorable outcome in patients (Axelrod et al. 2019). Unlike MHC-I, MHC-II is composed of an α and a β chain, which are then associated with an invariant chain (Ii) in the ER. Binding of Ii to MHC-II prevents the loading of antigen peptides onto MHC-II through the blockade of the antigen-binding groove (Busch et al. 2000; Bryant and Ploegh 2004). The Ii-MHC-II complex is then transported to the late endosome, where part of Ii is cleaved, leaving the MHC-II-associated Ii peptide (CLIP) in the peptide binding groove. The late endosome is fused with an autophagosome that contains cytosolic antigens (van den Boorn et al. 2011). The nonclassical MHC-II molecule, human leukocyte antigen-DM, interacts with MHC-II and catalyzes the exchange of CLIP with the antigenic peptides. MHC-II loaded with antigenic peptide is then transported to the cell surface through endolysosomal tubules to present the antigen to CD4+ T cells (Vyas et al. 2008).
ii. Cross-presentation
In contrast to MHC II-restricted exogenous presentation, the process of cross-presentation is more complicated, and is primarily referred to as the presentation of exogenous antigens by APCs through MHC-I to CD8+ T cells (Figure 3(B)) (Vyas et al. 2008). Although other APCs, such as macrophages and B cells, can cross-present exogenous antigens (Ramirez and Sigal 2002; Marino et al. 2012), DCs are considered to be the most prominent cross-presenting APCs. DCs uptake exogenous antigens, including tumor cell debris, through endocytic pathways such as phagocytosis, macro-pinocytosis, and endocytosis (Guermonprez et al. 2002; Kamphorst et al. 2010). These extracellular antigens are then presented on the surface of APCs through two major pathways: TAP-dependent (endosome-to-cytosol) and independent (vacuolar) (Figure 3(B)). The endosome-to-cytosol pathway is unique to DCs, enabling the antigens to be internalized in the endosome and transported into the cytosol for proteasome-mediated degradation (Kovacsovics-Bankowski and Rock 1995; Rodriguez et al. 1999; Ackerman et al. 2003). The vacuolar pathway is utilized by APCs to cross-present virus and tumor antigens (Liu et al. 1997; Campbell et al. 2000; Ma et al. 2016). Exosomes secreted by tumor cells contain tumor antigens, which can be presented by DCs through their uptake of exosomes (Wolfers et al. 2001). Phagocytosis also results in efficient cross-presentation of tumor antigen (Berard et al. 2000; Nouri-Shirazi et al. 2000; Russo et al. 2000).
A key step in the endosome to cytosol pathway is the transfer of antigens from phagosomes to the cytosol (Ackerman et al. 2003; Embgenbroich and Burgdorf 2018). It is well accepted that ERAD components participate in the retrotranslocation of exogenous antigens, as demonstrated by the detection of ERAD components on the phagosomal membrane (Houde et al. 2003). One ERAD component involved in antigen presentation is the E3 CHIP. Downregulation of CHIP results in decreased cross-presentation of ovalbumin (OVA) and the accumulation of OVA in DCs (Imai et al. 2005). P97 is also involved in cross-presentation (Imai et al. 2005; Ackerman et al. 2006; Zehner et al. 2011; Menager et al. 2014). For instance, P97 can interact with exogenous OVA, and the knockdown of P97 reduces the degradation and cross-presentation of OVA (Imai et al. 2005). Mutant P97 that lacks the ATPase can inhibit cross-presentation, indicating that the ATPase activity of P97 is required for retrotranslocation of exogenous antigens (Ackerman et al. 2006). The recruitment of P97 toward the endosomal membrane is regulated by the polyubiquitination of the mannose receptor, C-type lectin (Zehner et al. 2011). Interestingly, P97, but not SEC61 or Derlin-1, can translocate the synthetic tumor antigen Melan-A peptides from the endosome into the cytosol (Menager et al. 2014). Although SEC61’s role in antigen presentation is controversial, several studies indicate that SEC61 participates in cross-presentation. Genetic inactivation of Sec61 decreases the degradation of exogenous OVA and its cross-presentation (Imai et al. 2005). Pharmacological inhibition of SEC61 by exotoxin A (ExoA) also impairs antigen cross-presentation through impairment of SEC61-dependent protein transport across the ER and phagosome membranes (Ackerman et al. 2006). An intracellular antibody, which can trap SEC61 inside the ER preventing its recruitment to the endosome, inhibits antigen translocation and cross-presentation (Zehner et al. 2015). Interestingly, a recent study shows that SEC61 blockade inhibits cross-presentation independently of the endosome to cytosol pathway (Grotzke et al. 2017). Together, these studies demonstrate an important role of ERAD components in antigen presentation, supporting its involvement in tumor immunity.
Paradoxical role of ERAD in tumor immunity
Tumor cells experience increased ER stress often resulting from oncogene activation, uncontrolled proliferation, hypoxia, nutrient deprivation, as well as anticancer therapy (Nakagawa et al. 2014; Cruz et al. 2017). Diverse strategies, such as the UPR and ERAD, are exploited by tumor cells to mitigate ER stress. However, the intrinsic stress response pathways in malignant cells exert paradoxical roles during tumor development. For instance, certain ERAD components can promote tumorigenesis, while others inhibit tumor cell survival and growth (Tax et al. 2019). In the TME, both the UPR and ERAD can impart extrinsic effects to promote immunosurveillance or immune evasion (Cubillos-Ruiz et al. 2017; Cha et al. 2018; Gong et al. 2014). When treated with chemotherapy or radiotherapy, ER stress can induce immunogenic cell death (ICD) of cancer cells, which activates antitumor immune responses (Garg et al. 2017). In DCs, ERAD facilitates the cross-presentation of tumor antigens (Li et al. 2011; Menager et al. 2014; Alloatti et al. 2017). However, increased ER stress in the TME can also dampen antitumor immune responses, leading to immune evasion of cancers.
i. The effect of ER stress on tumor immunity
In the TME, ER stress impacts tumor and immune cells simultaneously, exerting a paradoxical role in tumor immunity. When cells experience ER stress, BiP is dissociated from three stress sensors on the ER membrane: RNA-dependent protein kinase-like kinase (PERK), IRE1α, and ATF6 (Moenner et al. 2007; Yadav et al. 2014). BiP, as well as IRE1α in certain conditions, binds to misfolded/unfolded proteins to activate the UPR, the signaling pathways upstream of ERAD (Dorner et al. 1992; Bertolotti et al. 2000; Gardner BM and Walter 2011). When the UPR is triggered by ER stress, PERK phosphorylates eukaryotic initiation factor 2α (eIF2α), which in turn induces the expression of transcription factor C/EBP-homologous protein (CHOP), a proapoptotic protein (Harding et al. 1999; Ma et al. 2002; Marciniak et al. 2004). In various types of cancer, ER stress resulting from high hydrostatic pressure induces tumor cell death by activating the PERK-eIF2α pathway (McCullough et al. 2001; Moserova et al. 2017), allowing DCs to engulf tumor cell debris to activate antitumor immune responses. Oligomerized IRE1α, which possesses RNase activity, splices XBP1. As a highly active transcription factor, spliced XBP1 can enhance the expression of ER chaperones and ERAD components (Lee AH et al. 2003; Shaffer et al. 2004; Mohamed et al. 2017). In addition, the IRE1α-XBP1 axis has been shown to suppress the development of colorectal cancer (Niederreiter et al. 2013). However, at later stages of tumor development, the UPR transitions from being cytotoxic to cytoprotective, enabling tumor cells to thrive and proliferate (Vanacker et al. 2017; Sundaram et al. 2018).
Besides cell death induced by unresolved ER stress, cancer cells can also release “danger signals” known as damage-associated molecular patterns (DAMPs) to initiate ICD and protective immune responses (Ma et al. 2011; van Vliet et al. 2015). There are primarily three types of DAMPs: surface exposure of CRT, the secretion of ATP, and the release of high mobility group box 1 (HMGB1) (Krysko et al. 2012). ICD induced by DAMPs under ER stress promotes antitumor immunity (Casares et al. 2005; Garg et al. 2012). Exposure of CRT on the cell surface signals DCs to engulf dying tumor cells (Gardai et al. 2005). Secretion of ATP attracts DCs to promote activation of the pyrin domain containing-3 inflammasome and increased secretion of pro-inflammatory cytokines such as IL-1β and IL-18 (Ghiringhelli et al. 2009; Tschopp and Schroder 2010). HMGB1 can bind to Toll-like receptor 4 on DCs and subsequently enhance cross-presentation of tumor antigens to CD8+ T cells (Apetoh et al. 2007; Kepp et al. 2013). Since ICD induction elicits long-term antitumor immunity, it has been proposed as a potential novel strategy in the treatment of cancer.
ER stress is the ultimate driving force of ICD induction (Sagar et al. 2019). In refractory endometrial cancer cells, ER stress can increase membrane expression of CRT, the key signal in ICD induction (Xu et al. 2017). Blockade or knockdown of CRT can eliminate antitumor immune responses in mice (Obeid et al. 2007). Furthermore, in NSCLC, CRT exposure is correlated with the recruitment of antitumor immune cells to the TME (Zitvogel et al. 2010; Fucikova et al. 2016). ERp75, an ER chaperone, can promote phagocytosis by DCs through the enhancement of CRT exposure (Obeid 2008). CRT exposure on the cell surface is usually mediated through activation of the PERK-eIF2α pathway. Phosphorylation of eIF2α by PERK correlates with CRT exposure, allowing for the use of phospho-eIF2α as a biomarker of ICD (Bezu et al. 2018). In melanoma cells, however, eIF2α is phosphorylated by protein kinase R and general control nonderepressible 2 to facilitate the translocation of CRT to the surface of tumor cells (Giglio et al. 2018). Although it is clear that the UPR contributes to ICD induction, whether ERAD can stimulate ICD remains unknown.
While ER stress resulting from cancer therapy primarily enhances antitumor immunity through ICD induction, sustained ER stress in cancer cells can also contribute to the establishment of the immunosuppressive network within the TME. Cancer cells experiencing ER stress secrete unknown factors into the TME to activate and promote the UPR in tumor-infiltrating immune cells such as macrophages (Mahadevan et al. 2011). This process is referred to as “transmissible ER stress”, which induces a robust proinflammatory response in tumor cells and pro-tumor microenvironment (Mahadevan et al. 2011). Transmissible ER stress can suppress the antigen-presenting capacity of DCs, impairing the activation of CD8+ T cells (Mahadevan et al. 2012).
ER stress activation in immune cells can also inhibit antitumor immunity. Activation of the IRE1α-XBP1 axis in macrophage promotes the secretion of cathepsin to enhance tumor cell invasion (Yan et al. 2016). ER stress in myeloid-derived suppressor cells (MDSCs) increases their immunosuppressive capacity, accelerating cancer progression (Condamine et al. 2014; Lee BR et al. 2014; Condamine et al. 2016; Nan et al. 2018). Induction of ER stress in neutrophils from healthy donors can transform the cells into immunosuppressive MDSCs (Condamine et al. 2016). CHOP deficiency in mouse MDSCs leads to decreased immunosuppressive functions (Thevenot et al. 2014). Additionally, CHOP is upregulated in tumor-infiltrating CD8+ T cells to suppress their cytotoxic response (Cao et al. 2019). ER stress can also promote tumor immune escape through disruption of antigen presentation. In advanced ovarian cancer, activation of the IRE1α-XBP1 axis in tumor-associated DCs disrupts ER homeostasis, inhibiting their antigen-presentation capacity and ultimately suppressing T-cell antitumor immunity (Cubillos-Ruiz et al. 2015; Cubillos-Ruiz and Glimcher 2016). Increased ER stress in thymoma cells activates the PERK-eIF2α pathway, leading to the blockade of protein synthesis that impairs tumor antigen presentation mediated by MHC-I (Granados et al. 2009). ER stress also suppresses the expression of components of the MHC-I machinery, including tapasin and TAP, to decrease tumor antigen presentation (Pellicciotta et al. 2008; Bartoszewski et al. 2011).
While it is apparent that ER stress can impede antitumor immunity, it has also been conversely implicated in immunosurveillance. A low protein diet can increase IRE1α signaling and subsequently activate the regulated IRE1α-dependent decay of ER-localized RNAs to enhance the cytotoxic response of CD8+ T cells thus suppressing tumor growth (Rubio-Patino et al. 2018). Activation of eIF2α enhances antitumor immunity by producing novel tumor peptides associated with MHC-I (Schwab et al. 2004). In murine melanoma cell lines, accumulation of misfolded/unfolded mutant tyrosinase in the ER promotes the presentation of certain tyrosinase epitopes by MHC-I (Ostankovitch et al. 2005). Furthermore, autophagy induced by ER stress is another means to activate protective antitumor immunity. As demonstrated in an in-vivo murine model, autophagy activity is required to sustain CD8α-type DCs for efficient cross-presentation of tumor antigens (Parekh et al. 2017).
ii. The effect of enhanced ERAD on tumor cells
While ERAD alleviates ER stress through facilitating the degradation of misfolded/unfolded proteins, components of ERAD exert paradoxical roles in tumorigenesis (Table 2). It has been shown that certain components of ERAD, such as ERmanI, Derlin-1, Derlin-3, HRD1, P97, NPL4, and UFD1, exert pro-tumor effects (Table 2) (Tax et al. 2019). Reduced expression of ERmanI can inhibit N-glycosylation and impair survival of breast cancer cells (Legler et al. 2018). Derlin-1 and 3, retrotranslocation channel proteins, promote overall cancer growth of breast origin (Guiliano et al. 2014; Shibata et al. 2017). Additionally, overexpression of Derlin-1 has been found in several human cancers including NSCLC, bladder, and colorectal cancer, predicting poor prognosis (Tan et al. 2015; Dong et al. 2017; Mao et al. 2018). Moreover, upregulation of the E3 ligase HRD1 promotes invasion of colorectal cancer (Tan et al. 2019). Recently, UFD1, a component facilitating retrotranslocation of ERAD substrates, has been found to mediate MYC-driven leukemia aggression (Huiting et al. 2018). Inhibitors of P97, a UFD1 functional partner, can interfere with autophagy and induce cancer cell death (Magnaghi et al. 2013). Disulfiram, a drug that can target NPL4, another UFD1 functional partner, can inhibit cancer cell growth (Skrott et al. 2017). Collectively, the P97-NPL4-UFD1 complex in the ERAD pathway plays an essential role in the survival of cancer cells.
Table 2.
Paradoxical role of ERAD components in tumor development.
ERAD components | Effects on tumor | Tumor types | Experimental approaches | Mechanisms of action | References |
---|---|---|---|---|---|
Derlin-1 | Tumor promoting | Breast, bladder, colorectal cancer, and NSCLC | Knockdown studies by shRNA in human cell culture | A retrotranslocation channel protein upregulated in colorectal cancer cells | Tan et al. (2015), Dong et al. (2017), and Mao et al. (2018) |
Derlin-3 | Tumor promoting | Breast cancer | SiRNA inhibition in breast cancer cell lines | A retrotranslocation channel protein, whose inhibition results in decreased invasion and proliferation of cancer cells | Guiliano et al. (2014) and Shibata et al. (2017) |
ERmanI | Tumor promoting | Breast cancer | Gene expression profiling of patient samples and gene inactivation by shRNA in human cancer cell lines | A mannosidase, its reduced expression inhibits N-glycosylation of substrates, impairing cancer cell survival | Legler et al. (2018) |
HRD1 | Tumor promoting | Colorectal cancer | Knockdown studies by shRNA in human cancer cell line | An E3, promoting degradation of misfolded proteins overexpressed in colorectal cancer | Tan et al. (2019) |
P97 | Tumor promoting | Colorectal, cervical cancer, and osteosarcoma | Small molecule and RNAi inhibition in human cancer cell lines | In complex with UFD1-NPL4 to facilitate substrate retrotranslocation in ERAD | Magnaghi et al. (2013) |
NPL4 | Tumor promoting | Breast cancer, myeloma, and osteosarcoma | Human cancer cell line and a murine model treated with disulfiram | In complex with P97 and UFD1 to facilitate substrate retrotranslocation in ERAD | Skrott et al. (2017) |
UFD1 | Tumor promoting | T-cell acute lymphoblastic leukemia | Gene inactivation studies in zebrafish models of leukemia and human leukemic cell lines. | In complex with P97 and NPL4 to facilitate substrate retrotranslocation in ERAD and its inactivation resulting in cytotoxic UPR responses | Huiting et al. (2018) |
SEC22b | Tumor suppressing | Lymphoma and fibrosarcoma | Conditional mouse knockout specific in DCs | An ER-phagosome traffic mediator, facilitating cross-presentation in DCs | Alloatti et al. (2017) |
CHIP | Tumor suppressing | Breast cancer | Gene inactivation by shRNA in mouse xenograft models and human cancer cell lines | An E3 facilitating degradation of misfolded substrates | Kajiro et al. (2009) |
OS-9 | Tumor suppressing | Pancreatic ductal adenocarcinoma | Measuring mRNA expression levels in patient samples of pancreatic ductal adenocarcinoma | A substrate recognition factor facilitating ERAD | Sun YW et al. (2014) |
SEL1L | Tumor promoting or suppressing | Pancreatic and breast cancer | Overexpression and downregulation studies in murine models | A substrate recognition and recruitment factor, facilitating substrate retrotranslocation; overexpression of SEL1L results in reduced pancreatic cancer invasion, while its downregulation predicts poor prognosis of breast cancer | Orlandi et al. (2002), Cattaneo et al. (2005), and Jeon et al. (2015) |
GP78 | Tumor promoting or suppressing | HCC, colorectal, esophageal, bladder, and breast cancer | Correlation studies in patients, knockout studies in mice, and gene inactivation studies by shRNA in murine models | An E3 participating in substrate degradation; high GP78 expression predicts poor prognosis of patients with esophageal squamous cell carcinoma, colorectal, and bladder cancer; Gp78−/− knockout increases HCC development; Gene inactivation shows tumor suppressive effect of GP78 in breast cancer | Nakamori et al. (1994), Otto et al. (1994), Maruyama et al. (1995), Silletti and Raz (1996), Zhang, Kho, et al. (2015), and Chang et al. (2016) |
Conversely, some ERAD components engage in tumor suppression (Table 2). Expression of CHIP, a ubiquitin ligase, is inversely correlated with breast cancer progression (Kajiro et al. 2009). Depletion of CHIP correlates with an increase in growth of subcutaneous tumors in mice, indicating its role as a tumor suppressor (Kajiro et al. 2009). Similarly, enforced expression of OS-9, a substrate recognition factor, can suppress invasion of pancreatic ductal adenocarcinoma (Sun YW et al. 2014). The roles of SEL1L and GP78, two other ERAD components, differ based on the context of cancer (Table 2). Due to the complex roles of ERAD components, it is important to understand their respective function in ERAD and effect on tumor cell survival, which can impact tumor immunity.
iii. The effect of ERAD on tumor immunity
While the role of ERAD in regulating immune cell development and function is evident, its involvement in tumor-infiltrating immune cells is not well-defined. Nevertheless, ERAD participates in cross-presentation, indicating its importance in tumor immunity. In murine models, deficiency in SEC22b, an ERAD component required for effective cross-presentation, leads to impaired antitumor immune responses (Alloatti et al. 2017). Additionally, treatment of DCs with ExoA, an inhibitor of the SEC61 translocon protein, partially suppresses activation of CD8+ T cells (Li et al. 2011). Similarly, studies in DCs demonstrate a critical role of ERAD in processing tumor antigens (Xing et al. 2016). An alternative ERAD pathway was found in the cross-presentation of synthetic long peptides (SLP), which have been used as successful antitumor vaccines against multiple types of cancer (Menager et al. 2014). This study demonstrates that cross-presentation is dependent on P97, a critical ERAD component, to translocate the SLP16–40 antigen (Menager et al. 2014). Moreover, the ubiquitin proteasome system depends on P97 to translocate antigens to MHC-I for cross-presentation (Palmer and Dolan 2013). Glucose-regulated protein 170 (GRP170), an ER chaperone, can deliver tumor antigens to APCs for cross-presentation, resulting in antitumor immune responses by cytotoxic T cells (Wang et al. 2010; Wang et al. 2014). For instance, GRP170 in DCs facilitates the processing and presentation of melanoma antigen glycoprotein 100 by enhancing its interaction with ERAD components, such as SEC61, P97, CHIP and GRP78 (Wang et al. 2013; Wang et al., 2014). These findings suggest that upregulation of ERAD in DCs may potentially enhance cross-presentation of tumor antigens to elicit a stronger cytotoxic antitumor response.
To evade immunosurveillance, cancer cells upregulate programed death-ligand 1 (PD-L1), which binds to its receptor PD-1, a critical immune checkpoint protein on activated T cells (Pardoll 2012). Since ERAD can promote degradation of PD-L1, enhanced ERAD in tumor cells may be beneficial for antitumor immune responses (Cha et al. 2018). However, ERAD can be detrimental to antitumor responses in different contexts. In human HCC, IRE1α upregulates HRD1 to degrade the CD155 ligand for CD226, a major NK-cell activating receptor (Gong et al. 2014). Thus, ERAD upregulation in HCC has an adverse effect on immunosurveillance and results in a poor prognosis, demonstrating the paradoxical function of ERAD in tumor immunity (Gong et al. 2014). Hence, more studies on ERAD’s function in the TME and tumor immunity are needed to guide the design and efficacy of current immunotherapies.
Therapeutic influences on ER stress and tumor immunity
The key for successful immunosurveillance and effective cancer therapy is their ability to induce immune responses against tumor cells (Blankenstein et al. 2012). Most chemotherapy are immunosuppressive, as they are cytotoxic to immune cells (Table 3). Additionally, some of them can induce ER stress and treatment resistance (Table 3). However, targeted therapy and subsets of cytotoxic chemotherapy are able to promote antitumor immune responses (Table 3). This effect can occur through ER stress induced ICD, modulation of immune cells in the TME, and/or reduction of immunosuppressive cytokines (Garg et al. 2010). The initiation of antitumor immune responses by cancer therapy aids the immune system in eradicating cancer cells, allowing for sustained cancer remission (Hughes and Yong 2017). Given the diverse effects of anticancer therapy on tumor immunity, understanding their respective roles on cancer cells and immunity is critical to guide clinical application of immunotherapy and combination treatment.
Table 3.
Therapeutic effects on ER stress and tumor immunity.
Therapy | Tumor types | Mechanisms of action | Effects on ER stress/ERAD | References |
---|---|---|---|---|
Camptothecin | Leukemia | Cytotoxic therapy, targeting topoisomerase and inducing apoptosis but not ICD | Sauter et al. (2000), Obeid et al. (2007), and Tesniere et al. (2010) | |
Etoposide | Testicular cancer, lung, lymphoma, leukemia, neuroblastoma, and ovarian cancer | Cytotoxic therapy inducing apoptosis but not ICD | Promoting apoptosis in human hepatic stellate cells via ER stress induction | Sauter et al. (2000), Obeid et al. 2007), and Wang et al. (2016) |
Mitomycin C | Adenocarcinoma of stomach and pancreas | Cytotoxic therapy inducing apoptosis but not ICD | Inducing ER stress | Sauter et al. (2000), Obeid et al. 2007), and Shi et al. (2013) |
Cyclophosphamide | Lymphoma, leukemia, NSCLC, breast, and ovarian cancer | Cytotoxic therapy, high dose with immunosuppressive effects by inhibiting immune cell proliferation, while counteracting immunosuppression by Tregs in single low dose | Inducing ER stress to expose CRT and release HMGB1 | Emadi et al. (2009), Schiavoni et al. (2011), Inoue and Tani (2014), and Ahlmann and Hempel (2016) |
Cisplatin | Various types of cancers, such as breast, ovarian, testicular, brain, and lung cancer | Cytotoxic therapy, inducing apoptosis but not ICD unless combined with ER stress inducer | Martins et al. (2011) | |
Oxaliplatin | Colorectal cancer | Cytotoxic therapy, inducing ICD | Activation of the PERK-eIF2α pathway, resulting in CRT exposure | Tesniere et al. (2010) and Garg et al. (2012) |
Mitoxantrone, idarubicin, epirubicin, and doxorubicin | Acute lymphoblastic leukemia, ovarian, and prostate cancer | Cytotoxic therapy, inducing ICD | Activation of the PERK-eIF2α pathway, resulting in CRT exposure | Fucikova et al. (2011) and Bezu et al. (2018) |
Bleomycin | Testicular cancer and Hodgkin’s lymphoma | Cytotoxic therapy, inducing ICD and may also promoting Treg proliferation | Inducing ER stress through the generation of reactive oxygen species and exposing CRT though activation of the PERK-eIF2α pathway | Bugaut et al. (2013) and Garg et al. (2017) |
Imatinib | Chronic myeloid leukemia, and gastric cancer | BCR-ABL tyrosine kinase inhibitor, promoting antitumor immune responses by inhibiting MDSCs and Treg function, while suppressing tumor immunity by inhibiting tumor antigens, DCs, and effector T cells | Inducing ER stress and resulting in apoptosis | Brauer et al. (2007), Larmonier et al. 2008), Heine et al. (2011), Hughes et al. (2017), and Kim et al. (2019) |
Nilotinib and dasatinib | Chronic myeloid leukemia | BCR-ABL tyrosine kinase inhibitor, reducing MDSCs but not inducing ICD | Hughes et al. (2017) | |
Abemaciclib | Breast cancer | CDK4/6 inhibitor, enhancing tumor antigen presentation | Goel et al. (2017) and Schaer et al. (2018) | |
Palbociclib | Breast cancer | CDK4/6 inhibitor, activating effector T cells | Deng et al. (2018) | |
Gefitinib, erlontinib, afatinib, osimertinib, and olmutinib | NSCLC | EGFR inhibitors, decreasing PD-L1 expression and reducing Tregs | Yamaoka et al. (2017) and Li et al. (2018) | |
Cetuximab | Colorectal cancer | mAb competitively inhibiting EGFR, inducing ADCC and enhancing cross-presentation in DCs | Kimura et al. (2007), Srivastava et al. (2013), and Yamaoka et al. (2017) | |
Anti-GD2 | Neuroblastoma | mAb targeting cell surface disialoganglioside GD2 and resulting in ADCC | Horta et al. (2016) | |
Ipilimumab | NSCLC and melanoma | mAb inhibiting CTLA-4 | Pennock and Chow (2015) and Buchbinder and Desai (2016) | |
Nivolumab | NSCLC and melanoma | mAb inhibiting PD-1 | Pennock and Chow (2015) and Buchbinder and Desai (2016) | |
Durvalumab | NSCLC | mAb targeting PD-L1 | Antonia et al. (2017) | |
Metformin | Activating AMPK to phosphorylate PD-L1 and promote its degradation via ERAD | PD-L1 is degraded by ERAD but metformin does not target ERAD | Cha et al. (2018) and Dreher and Hoppe (2018) | |
Disulfram | Off-target effect on NPL4 | Impairing ERAD by disrupting P97-NPL4-UFD1 complex and inhibiting proinflammatory macrophage responses | Skrott et al. (2017) |
i. Effects of cytotoxic chemotherapy on ER stress and tumor immunity
Cytotoxic chemotherapy has historically been considered immunogenically silent or immunosuppressive (Harris et al. 1976). When killing cancer cells, most cytotoxic chemotherapy can induce ER stress to dampen antitumor immune responses (Bugaut et al. 2013). Additionally, this type of chemotherapy can interfere with hematopoiesis by eliminating hematopoietic stem cells, blood cells, and immune cells (Emadi et al. 2009). Cytotoxic drugs, for example, camptothecin, etoposide, and mitomycin C, induce apoptosis but not ICD, and thus are nonimmunogenic (Sauter et al. 2000; Obeid et al. 2007). High dose cyclophosphamide, used for treating hematological malignancies, has been shown to actively suppress the immune system by decreasing proliferation of immune cells (Emadi et al. 2009). Moreover, it can induce ER stress and activation of the antiapoptotic UPR, resulting in chemoresistance (Yarapureddy et al. 2019). In osteosarcoma, activation of ATF6 increases resistance to chemotherapy, leading to decreased overall and progression-free survival (Yarapureddy et al. 2019). A similar effect is seen upon BiP induction due to increased ER stress, which is correlated with increased tumor cell survival and chemoresistance (Lee AS 2007).
Although ER stress exerts a pro-tumor role, chemotherapy that results in ICD often utilizes the ER stress response pathways. For example, cisplatin alone is insufficient to induce ICD unless it is combined with ER stress inducers, such as thapsigargin or tunicamycin (Martins et al. 2011). Certain cytotoxic chemotherapy that do induce ER stress, such as oxaliplatin, mitoxantrone, idarubicin, epirubicin, doxorubicin, and cyclophosphamide, can cause ICD (Wu and Waxman 2018). Those chemotherapy can directly stimulate immunogenic responses primarily through the emission of one or multiple DAMPs (Krysko et al. 2012). Among them, the exposure of CRT on the cell surface is the primary determining factor for ICD induction by chemotherapy (Obeid et al. 2007; Krysko et al. 2012). Anthracycline chemotherapies, which include mitoaxantrone, idarubicin, epirubicin, and doxorubicin, have been shown to cause an antitumor immune response by inducing ICD in human prostate cancer, ovarian cancer, and acute lymphoblastic leukemia (Fucikova et al. 2011). To accomplish this function, this type of chemotherapy relies on the activation of the PERK-eIF2α pathway, which causes the translocation of CRT to the cell surface (Bezu et al. 2018). However, anthracyclines and oxaliplatin do not trigger the ATF6 or IRE1α branch of the UPR, suggesting that these UPR pathways do not contribute to ICD induction (Fucikova et al. 2011). Similarly, oxaliplatin, but not cisplatin, causes the translocation of CRT from the ER to the cell surface through the PERK-eIF2α pathway, and can induce ICD in colorectal cancer (Garg et al. 2012). Consistently, oxaliplatin treatment in colorectal cancer cells with CRT depletion cannot induce ICD (Tesniere et al. 2010).
Other ICD-inducing chemotherapy, such as cyclophosphamide and bleomycin, have a paradoxical role on antitumor immunity. Cyclophosphamide, one of the first anticancer agents, is immunosuppressive with high dose treatments depleting hematopoietic lineages, thus patients commonly receive bone marrow transplantation (Emadi et al. 2009). However, cyclophosphamide has also been shown to induce ICD in lymphoma cells by exposing CRT and releasing HMGB1 (Schiavoni et al. 2011; Inoue H and Tani 2014). When applied as a single and low dose, cyclophosphamide can mitigate the immunosuppressive effects of Tregs, aiding in cancer treatment (Ahlmann and Hempel 2016). Bleomycin, a glycopeptide antibiotic used in the treatment of testicular cancer and Hodgkin’s lymphoma, has also been shown to result in the induction of ICD (Bugaut et al. 2013; Garg et al. 2017). However, this effect is counteracted by bleomycin’s ability to stimulate Treg proliferation, making this drug both immunomodulatory and immunosuppressive, similar to cyclophosphamide (Bugaut et al. 2013).
ii. Effects of targeted therapy on ER stress and tumor immunity
Targeted cancer therapy can exert similar antitumor immune responses as some cytotoxic chemotherapy and are particularly important as they often minimize treatment side-effects. One of the first targeted cancer therapy, imatinib (Gleevec) and the more potent second-generation inhibitors nilotinib and dasatinib, can bolster the immune responses in chronic myeloid leukemia patients through reduction of MDSCs (Hughes et al. 2017). While imatinib, nilotinib, and dasatinib have not been shown to induce ICD, their ability to reduce immunosuppressive cells has lasting effects of promoting disease remission without the need for continuous treatment (Hughes and Yong 2017). Moreover, imatinib can impair the immunosuppressive functions of Tregs and enhance immunotherapy against lymphoma (Larmonier et al. 2008). However, imatinib downregulates tumor antigens in leukemic cells and inhibits DCs and effector T cells, suppressing antitumor immunity (Brauer et al. 2007; Heine et al. 2011). Inhibitors of cyclin-dependent kinase 4 and 6 (CDK4/6), such as ribociclib, abemaciclib, and palbociclib, recently gained traction in treating advanced breast cancer. In addition to inhibiting tumor cell proliferation, these inhibitors can also evoke antitumor immune responses. Abemaciclib enhances antigen presentation in tumor cells and suppresses Treg functions, while palbociclib activates effector T cells (Goel et al. 2017; Deng et al. 2018; Schaer et al. 2018).
Other targeted therapy inhibitors, such as EGFR inhibitors, have since been developed for the treatment of multiple cancers - such as NSCLC, colorectal, and breast cancer - and have been shown to promote antitumor immune responses (Li X et al. 2018). Gefitinib, erlotinib, afatinib, osimertinib, and olmutinib are approved treatments for metastatic NSCLC, while lapatinib is approved for HER2+ breast cancer (Yamaoka et al. 2017). Although EGFR inhibition has not been shown to induce ICD, it can increase antitumor immune responses. In NSCLC, EGFR inhibition with gefitinib results in decreased expression of PD-L1 (Li X et al. 2018). Additionally, gefitinib treatment reduces Tregs in mice engrafted with squamous cell carcinoma (Mascia et al. 2016). EGFR inhibitors, therefore, can affect both tumor cells and the TME. However, treatment resistance to EGFR inhibitors is common, demanding the use of other therapeutic strategies to overcome this clinical challenge (Stewart et al. 2015; Tang Z-H and Lu 2018).
Similar to cytotoxic chemotherapy, targeted therapy can also induce ER stress and alter ERAD, leading to treatment resistance. Melanomas with the BRAFV600E mutation often become resistant to BRAF inhibitors, such as vemurafenib, due to the induction of ER stress and cytoprotective autophagy (Ma et al. 2014). This resistance can be overcome when combined with autophagy inhibition (Ma et al. 2014). Vemurafenib can also induce miR-211 to downregulate EDEM1, an ER chaperone critical for ERAD of tyrosinase, subsequently leading to tyrosinase accumulation and treatment resistance (Vitiello et al. 2017). Another targeted therapy is monoclonal antibodies (mAbs). Cetuximab, a mAb against EGFR and an effective competitive inhibitor, can result in antibody-dependent cell-mediated cytotoxicity (ADCC) in colorectal cancer (Kimura et al. 2007; Yamaoka et al. 2017). Additionally, cetuximab induces crosstalk between NK cells and DCs to promote the maturation of DCs, enhancing cross-presentation (Srivastava et al. 2013). A similar result is seen in the use of HER2 mAbs, which has led to better clinical outcomes in HER2+ patients (Costa and Czerniecki 2020). In neuroblastoma treatment, anti-GD2, the first mAb approved for treating childhood malignancies, has also been shown to result in ADCC (Horta et al. 2016). Due to their ability to specifically target cancer cells while eliciting immune responses, mAbs represent the most commonly used and approved immunotherapy in the treatment of cancer (Kimiz-Gebologlu et al. 2018).
Given the known role of ERAD in promoting tumor cell survival, it is reasonable to suspect that enhanced ERAD in tumor cells might result in resistance to ICD and decreased availability of tumor cell debris as antigens for DCs. As such, the benefits of ERAD inhibitors to cancer treatment could be two-fold: inducing cancer cell death and increasing antitumor immunity. The efficacy of such treatments is seen in proteasome inhibitors, such as bortezomib, which result in cancer cell death due to irresolvable ER stress as well as ICD induction (Obeng et al. 2006; Spisek et al. 2007; Strasser and Puthalakath 2008). The mechanism of ICD induction by bortezomib is through exposure of HSP90 (another DAMP) on the tumor cell surface (Spisek et al. 2007). Other components of the ERAD pathway may be similarly targetable. P97 is the AAA ATPase mainly associated with retrotranslocation of ERAD substrates (Carvalho et al. 2006). CB-5083, a selective and potent inhibitor for P97, results in irresolvable ER stress and subsequent cell death in multiple myeloma (Anderson et al. 2015). However, CB-5083 recently failed in phase I clinical trials due to high toxicity (NCT02243917). Interestingly, disulfram, an alcohol-aversion drug, can disrupt ERAD by inhibiting NPL4, a P97 partner. Disulfram’s use in cancer patients has led to lower mortality, due to its double-edged effects: promoting cancer cell death through ERAD impairment and inhibiting macrophage proinflammatory responses (Skrott et al. 2017; Terashima et al. 2020).
iii. ERAD’s impact on immunotherapy
Immunotherapy functions by promoting cytotoxic responses to tumor cells through cytokines and effector immune cells, leading to enduring antitumor immune responses. Advances in immunotherapy have improved cancer treatment, especially through the application of chimeric antigen receptor T-cells and immune checkpoint inhibitors, such as ipilimumab and nivolumab (Buchbinder and Desai 2016). However, current immunotherapy possesses significant side effects. A major adverse event, for example, is abnormal reactivity to normal tissues by hyperactivated T cells (Weber et al. 2015). Tumors in most patients respond to the treatment and are controllable for a long time, yet one-third of them relapse by some unknown mechanism (Ribas and Wolchok 2018).
Two immune checkpoints, cytotoxic T-lymphocyte-associated antigen 4 (CTLA-4) and PD-1, put the brakes on T-cell functions (Buchbinder and Desai 2016). CTLA-4 is expressed on Tregs, which ultimately inhibit T-cell activation and proliferation (Wing et al. 2008). PD-1 is expressed on activated T cells, suppressing their functions by binding to its ligand PD-L1 (Okazaki and Honjo 2007; Yokosuka et al. 2012). Ipilimumab and nivolumab, mAbs against CTLA-4 and PD-1 respectively, have achieved some success in treating melanoma and NSCLC (Pennock and Chow 2015; Buchbinder and Desai 2016). Since PD-L1 is often upregulated in malignant cells, the mAb durvalumab has been developed to target PD-L1, leading to increased progression-free survival in NSCLC (Antonia et al. 2017). Interestingly, metformin, a commonly used drug for type 2 diabetes, induces the degradation of PD-L1 through ERAD (Dreher and Hoppe 2018). Metformin can interact with the AMP-activated protein kinase (AMPK) and enhance AMPK-mediated phosphorylation of PD-L1. Although it has been observed that the phosphorylated form of PD-L1 accumulates in the ER and is subsequently degraded through ERAD, how ERAD targets PD-L1 remains unknown (Cha et al. 2018). In contrast, disruption of ERAD by disulfram upregulates PD-L1, leading to immunosuppression and treatment resistance in cancer patients (Zhou et al. 2019). A better understanding of ERAD in tumor immunity is needed for successful application of immunotherapy.
Future perspectives
Over the past four decades, significant insights have been acquired through in-vitro co-culture and in-vivo humanized animal model studies, establishing ERAD as a principle protein quality control mechanism, which often enables the survival and growth of tumor cells (Tax et al. 2019). Due to its ability to degrade key regulators of immune cells, such as TCR, BCR, and BLIMP-1, ERAD plays an indispensable role in immune cell development and function (Bonifacino et al. 1989; Yang et al. 1998; Yang et al. 2014; Ji et al. 2016). Through degrading MHC-I, processing tumor antigens, and translocating exogenous antigens for cross-presentation, components of ERAD participate in antitumor immune responses (Gong et al. 2014; Grotzke and Cresswell 2015). Despite some known effects of current anticancer therapy in evoking ER stress and their paradoxical roles in tumor immunity, our limited understanding of ERAD presents a significant barrier to applying current therapy for successful eradication of cancer.
While emerging evidence demonstrates ERAD’s regulatory role in immune cell development and function, little is known about how it impacts immune cells in the TME. In particular, several questions remain unanswered. How does altered ERAD in tumor cells affect functions of tumor-infiltrating immune cells and the induction of ICD by cancer therapy? How does altered ERAD in tumor-infiltrating immune cells impact their antitumor capacities? How can other forms of cancer therapy, particularly immunotherapy, be combined with ERAD inhibitors? Due to these unanswered questions, caution must be taken in combination treatment utilizing current immunotherapy with other therapy that are known to evoke ER stress and ERAD.
Therapy targeting ERAD is still in its infancy due to the complex and multifaceted roles of ERAD in tumorigenesis and immunity (Guerriero and Brodsky 2012). Although a number of small-molecule inhibitors targeting ERAD have been developed, none except proteasome inhibitors have progressed to clinical applications due to their toxicities. Therefore, a better understanding of ERAD is necessary for selection of therapeutic targets for development of minimally toxic treatments. Animal models with lineage-specific depletion of ERAD components will facilitate the dissection of ERAD’s effects on specific types of immune cells, as well as on tumor cells, in both physiological and pathological settings. With improved understanding of how ERAD regulates proteins relevant to immune checkpoints, such as PD-L1, a more effective combination therapy could be designed. Of course, the successful combination of ERAD inhibitors or other therapy with immunotherapy is likely to be context dependent as the ability for each therapy to evoke ER stress and impact tumor immunity differs in each cancer type. It is our hope that an improved understanding of ERAD’s role in tumor immunity will not only lead to development of novel and specific inhibitors but also guide rational application of currently available therapy in our battle against cancer.
Acknowledgement
The authors thank members of the Feng laboratory for proofreading of the manuscript.
Funding
This work was supported by the St. Baldrick’s Foundation (a career development scholar grant), the National Institute of Health [grant number 1UL1TR001430], [grant number CA215059] to HF; the American Cancer Society [grant number RSG-17-204-01-TBG HF]; the National Science Foundation [grant number 1911253]; and the Undergraduate Research of Opportunity Program Award from Boston University [to KS].
Abbreviations:
- AAT
alpha 1 antitrypsin
- ADCC
antibody-dependent cell-mediated cytotoxicity
- AMPK
AMP-activated protein kinase
- APCs
antigen-presenting cells
- ApoB100
apolipoprotein B
- ASGPR
asialoglycoprotein receptor
- ATF6
activating transcription factor 6
- AUP1
ancient ubiquitous protein 1
- BACE457
pancreatic isoform of human β-secretase
- β2m
β2-microglobulin
- BAG6
BCL2-associated athanogene 6
- Pre-BCR
pre-B-cell receptor
- BiP
binding immunoglobulin protein
- BLIMP-1
B lymphocyte-induced maturation protein-1
- CDK4/6
cyclin-dependent kinase 4 and 6
- CFTR
mutant cystic fibrosis transmembrane regulator
- CHIP
C-terminus of HSP70-interacting protein
- CHOP
transcription factor C/EBP-homologous protein
- CRT
calreticulin
- CTLA-4
cytotoxic T-lymphocyte-associated antigen 4
- DAMP
damage-associated molecular pattern
- Doa10
degradation of alpha 10
- DCs
dendritic cells
- DUBs
deubiquinating enzymes
- E1
ubiquitin-activating enzyme
- E2
ubiquitin-conjugating enzyme
- E3
ubiquitin ligase
- E4
specialized ubiquitin ligase
- E6-AP
E6-associated protein
- EDEM 1/2/3
ER degradation-enhancing α-mannosidase-like lectin 1/2/3
- EGFR
epidermal growth factor receptor
- ER
endoplasmic reticulum
- ERAD
endoplasmic reticulum-associated protein degradation
- ERAP1/2
ER aminopeptidase-1/2
- ERdj5
endoplasmic reticulum (ER)-resident protein-containing DnaJ and thioredoxin domains
- ERManI
ER mannosidase-I
- ERp57
ER-resident protein 57
- eIF2α
eukaryotic initiation factor 2α
- ExoA
exotoxin A
- Fbx
F-box protein
- Glc
glucose
- GlcNAc2
N-Acetylglucosamine
- GP78
glycoprotein 78
- GRP170
Glucose-regulated protein 170
- HCC
hepatocellular carcinoma cell
- HERP
Homocysteine-induced ER protein
- HMGB1
high mobility group box 1
- HMGR
hydroxy-3-methyl-glutaryl-CoA reductase
- Hrd3
hmg CoA reductase degradation 3
- HSP70
heat shock protein 70
- IL-2
interleukin-2
- Igμ
IgM heavy chain
- IgG/M
immunoglobulin G/M
- Ii
invariant chain
- ICD
immunogenic cell death
- INSIG-1
insulin-induced gene 1
- IRE1α
inositol-requiring enzyme 1α
- LPL
lipoprotein lipase
- LPS
lipopolysaccharides
- mAb
monoclonal antibody
- MDSCs
myeloid-derived suppressor cells
- MHC
major histocompatibility complexes
- MHC-I
MHC class I molecules
- MHC-II
MHC class II molecules
- Man
mannose
- NGly
N-glycanase
- NK cells
natural killer cells
- NPL4
nuclear protein localization protein 4
- NSCLC
nonsmall cell lung cancer
- OS-9
osteosarcoma 9
- OST
oligosaccharyl transferase
- OVA
ovalbumin
- PERK
protein kinase RNA-like ER kinase
- PDIs
protein disulfide isomerases
- PD-L1
programed death ligand 1
- PLC
peptide loading complex
- RAD23
radiation-sensitive 23
- RMA1
RING finger protein 5
- RING
really interesting new gene
- RTA
glycosylated protein-rich A chain
- SEL1L
suppressor enhancer Lin12 1 like
- SKP1
S-phase kinase-associated protein 1
- SLP
synthetic long peptide
- TAP
transporter associated with antigen processing
- TCRα
T-cell receptor alpha chain
- TCR
T-cell receptor
- TME
tumor microenvironment
- TMEM129
transmembrane protein 129
- TRC8
translocation in renal carcinoma, chromosome 8 gene
- TRC35
transmembrane domain recognition complex 35
- Tregs
T regulatory cells
- Ubc6p
ubiquitin-conjugating enzyme 6p
- Ubx2p
UBX domain-containing protein 2
- UBC6E
ubiquitin-conjugating enzyme 6e
- UBE2G2
ubiquitin-conjugating enzyme E2 G2
- UBE2J1
ubiquitin-conjugating enzyme E2 J1
- UBL4A
ubiquitin-like protein 4A
- UBR1/2
E3 component N-recognin 1/2
- UBXD8
ubiquitinlike-domain-containing protein
- UFD1
ubiquitin fusion degradation protein
- UGGT
glycoprotein glycosyl-transferase
- UPR
unfolded protein response
- USP13
ubiquitin-specific protease 13
- VCP
valosin-containing-protein
- WDR20
WD repeat-containing protein 20
- XTP3-B
XTP3-transactivated gene B
- YOD1
ubiquitin thioesterase OTU1
- Yos9
Yeast OS-9 homolog
Footnotes
Disclosure statement
No potential conflict of interest was reported by the author(s).
References
- Ackerman AL, Giodini A, Cresswell P. 2006. A role for the endoplasmic reticulum protein retrotranslocation machinery during crosspresentation by dendritic cells. Immunity. 25(4):607–617. [DOI] [PubMed] [Google Scholar]
- Ackerman AL, Kyritsis C, Tampe R, Cresswell P. 2003. Early phagosomes in dendritic cells form a cellular compartment sufficient for cross presentation of exogenous antigens. Proc Natl Acad Sci U S A. 100(22):12889–12894. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Ahlmann M, Hempel G. 2016. The effect of cyclophosphamide on the immune system: implications for clinical cancer therapy. Cancer Chemother Pharmacol. 78(4):661–671. [DOI] [PubMed] [Google Scholar]
- Alfano M, Graziano F, Genovese L, Poli G. 2013. Macrophage polarization at the crossroad between HIV-1 infection and cancer development. Arterioscler Thromb Vasc Biol. 33(6): 1145–1152. [DOI] [PubMed] [Google Scholar]
- Alloatti A, Rookhuizen DC, Joannas L, Carpier JM, Iborra S, Magalhaes JG, Yatim N, Kozik P, Sancho D, Albert ML, et al. 2017. Critical role for Sec22b-dependent antigen cross-presentation in antitumor immunity. J Exp Med. 214(8):2231–2241. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Anderson DJ, Le Moigne R, Djakovic S, Kumar B, Rice J, Wong S, Wang J, Yao B, Valle E, Kiss von Soly S, et al. 2015. Targeting the AAA ATPase p97 as an Approach to Treat Cancer through Disruption of Protein Homeostasis. Cancer Cell. 28(5):653–665. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Antonia SJ, Villegas A, Daniel D, Vicente D, Murakami S, Hui R, Yokoi T, Chiappori A, Lee KHD, Wit M, et al. 2017. Durvalumab after chemoradiotherapy in Stage III non--small-cell lung cancer. N Engl J Med. 377(20):1919–1929. [DOI] [PubMed] [Google Scholar]
- Apetoh L, Ghiringhelli F, Tesniere A, Obeid M, Ortiz C, Criollo A, Mignot G, Maiuri MC, Ullrich E, Saulnier P, et al. 2007. Toll-like receptor 4-dependent contribution of the immune system to anticancer chemotherapy and radiotherapy. Nat Med. 13(9):1050–1059. [DOI] [PubMed] [Google Scholar]
- Avezov E, Frenkel Z, Ehrlich M, Herscovics A, Lederkremer GZ. 2008. Endoplasmic reticulum (ER) mannosidase I is compartmentalized and required for N-glycan trimming to Man5–6GlcNAc2 in glycoprotein ER-associated degradation. Mol Biol Cell. 19(1):216–225. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Axelrod ML, Cook RS, Johnson DB, Balko JM. 2019. Biological Consequences of MHC-II Expression by Tumor Cells in Cancer. Clin Cancer Res. 25(8):2392–2402. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Baldridge RD, Rapoport TA. 2016. Autoubiquitination of the Hrd1 Ligase Triggers Protein Retrotranslocation in ERAD. Cell. 166(2):394–407. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Bartoszewski R, Brewer JW, Rab A, Crossman DK, Bartoszewska S, Kapoor N, Fuller C, Collawn JF, Bebok Z. 2011. The unfolded protein response (UPR)-activated transcription factor X-box-binding protein 1 (XBP1) induces microRNA-346 expression that targets the human antigen peptide transporter 1 (TAP1) mRNA and governs immune regulatory genes. J Biol Chem. 286(48):41862–41870. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Bays NW, Gardner RG, Seelig LP, Joazeiro CA, Hampton RY. 2001. Hrd1p/Der3p is a membrane-anchored ubiquitin ligase required for ER-associated degradation. Nat Cell Biol. 3(1):24–29. [DOI] [PubMed] [Google Scholar]
- Berard F, Blanco P, Davoust J, Neidhart-Berard EM, Nouri-Shirazi M, Taquet N, Rimoldi D, Cerottini JC, Banchereau J, Palucka AK. 2000. Cross-priming of naive CD8 T cells against melanoma antigens using dendritic cells loaded with killed allogeneic melanoma cells. J Exp Med. 192(11): 1535–1544. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Bertolotti A, Zhang Y, Hendershot LM, Harding HP, Ron D. 2000. Dynamic interaction of BiP and ER stress transducers in the unfolded-protein response. Nat Cell Biol. 2(6): 326–332. [DOI] [PubMed] [Google Scholar]
- Bettigole SE, Glimcher LH. 2015. Endoplasmic reticulum stress in immunity. Annu Rev Immunol. 33:107–138. [DOI] [PubMed] [Google Scholar]
- Bezu L, Sauvat A, Humeau J, Gomes-da-Silva LC, Iribarren K, Forveille S, Garcia P, Zhao L, Liu P, Zitvogel L, et al. 2018. eIF2α phosphorylation is pathognomonic for immunogenic cell death. Cell Death Differ. 25(8):1375–1393. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Bhamidipati A, Denic V, Quan EM, Weissman JS. 2005. Exploration of the topological requirements of ERAD identifies Yos9p as a lectin sensor of misfolded glycoproteins in the ER lumen. Mol Cell. 19(6):741–751. [DOI] [PubMed] [Google Scholar]
- Blankenstein T, Coulie PG, Gilboa E, Jaffee EM. 2012. The determinants of tumour immunogenicity. Nat Rev Cancer. 12(4):307–313. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Bonifacino JS, Suzuki CK, Lippincott-Schwartz J, Weissman AM, Klausner RD. 1989. Pre-Golgi degradation of newly synthesized T-cell antigen receptor chains: intrinsic sensitivity and the role of subunit assembly. J Cell Biol. 109(1): 73–83. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Bonifacino JS, Cosson P, Klausner RD. 1990. Colocalized transmembrane determinants for ER degradation and subunit assembly explain the intracellular fate of TCR chains. Cell. 63(3):503–513. [DOI] [PubMed] [Google Scholar]
- Brauer KM, Werth D, von Schwarzenberg K, Bringmann A, Kanz L, Grunebach F, Brossart P. 2007. BCR-ABL activity is critical for the immunogenicity of chronic myelogenous leukemia cells. Cancer Res. 67(11):5489–5497. [DOI] [PubMed] [Google Scholar]
- Bryant P, Ploegh H. 2004. Class II MHC peptide loading by the professionals. Curr Opin Immunol. 16(1):96–102. [DOI] [PubMed] [Google Scholar]
- Buchbinder EI, Desai A. 2016. CTLA-4 and PD-1 pathways: similarities, differences, and implications of their inhibition. Am J Clin Oncol. 39(1):98–106. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Bugaut H, Bruchard M, Berger H, Derangère V, Odoul L, Euvrard R, Ladoire S, Chalmin F, Végran F, Rébé C, et al. 2013. Bleomycin exerts ambivalent antitumor immune effect by triggering both immunogenic cell death and proliferation of regulatory T cells. PLoS One. 8(6):e65181. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Burgdorf S, Schölz C, Kautz A, Tampé R, Kurts C. 2008. Spatial and mechanistic separation of cross-presentation and endogenous antigen presentation. Nat Immunol. 9(5): 558–566. [DOI] [PubMed] [Google Scholar]
- Burr ML, Cano F, Svobodova S, Boyle LH, Boname JM, Lehner PJ. 2011. HRD1 and UBE2J1 target misfolded MHC class I heavy chains for endoplasmic reticulum-associated degradation. Proc Natl Acad Sci USA. 108(5):2034–2039. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Burr ML, van den Boomen DJ, Bye H, Antrobus R, Wiertz EJ, Lehner PJ. 2013. MHC class I molecules are preferentially ubiquitinated on endoplasmic reticulum luminal residues during HRD1 ubiquitin E3 ligase-mediated dislocation. Proc Natl Acad Sci USA. 110(35):14290–14295. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Busch R, Doebele RC, Patil NS, Pashine A, Mellins ED. 2000. Accessory molecules for MHC class II peptide loading. Curr Opin Immunol. 12(1):99–106. [DOI] [PubMed] [Google Scholar]
- Cambier JC, Gauld SB, Merrell KT, Vilen BJ. 2007. B-cell anergy: from transgenic models to naturally occurring anergic B cells?. Nat Rev Immunol. 7(8):633–643. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Campbell DJ, Serwold T, Shastri N. 2000. Bacterial proteins can be processed by macrophages in a transporter associated with antigen processing-independent, cysteine protease-dependent manner for presentation by MHC class I molecules. J Immunol. 164(1):168–175. [DOI] [PubMed] [Google Scholar]
- Campoli M, Ferrone S. 2008. HLA antigen changes in malignant cells: epigenetic mechanisms and biologic significance. Oncogene. 27(45):5869–5885. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Cao Y, Trillo-Tinoco J, Sierra RA, Anadon C, Dai W, Mohamed E, Cen L, Costich TL, Magliocco A, Marchion D, et al. 2019. ER stress-induced mediator C/EBP homologous protein thwarts effector T cell activity in tumors through T-bet repression. Nat Commun. 10(1):1280. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Carlsten M, Norell H, Bryceson YT, Poschke I, Schedvins K, Ljunggren HG, Kiessling R, Malmberg KJ. 2009. Primary human tumor cells expressing CD155 impair tumor targeting by down-regulating DNAM-1 on NK cells. J Immunol. 183(8):4921–4930. [DOI] [PubMed] [Google Scholar]
- Carvalho P, Goder V, Rapoport TA. 2006. Distinct ubiquitinligase complexes define convergent pathways for the degradation of ER proteins. Cell. 126(2):361–373. [DOI] [PubMed] [Google Scholar]
- Casares N, Pequignot MO, Tesniere A, Ghiringhelli F, Roux S, Chaput N, Schmitt E, Hamai A, Hervas-Stubbs S, Obeid M, et al. 2005. Caspase-dependent immunogenicity of doxorubicin-induced tumor cell death. J Exp Med. 202(12): 1691–1701. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Cattaneo M, Fontanella E, Canton C, Delia D, Biunno I. 2005. SEL1L affects human pancreatic cancer cell cycle and invasiveness through modulation of PTEN and genes related to cell-matrix interactions. Neoplasia. 7(11):1030–1038. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Cattaneo M, Otsu M, Fagioli C, Martino S, Lotti LV, Sitia R, Biunno I. 2008. SEL1L and HRD1 are involved in the degradation of unassembled secretory Ig-mu chains. J Cell Physiol. 215(3):794–802. [DOI] [PubMed] [Google Scholar]
- Cha JH, Yang WH, Xia W, Wei Y, Chan LC, Lim SO, Li CW, Kim T, Chang SS, Lee HH, et al. 2018. Metformin Promotes Antitumor Immunity via Endoplasmic-Reticulum-Associated Degradation of PD-L1. Mol Cell. 71(4):606–620 e607. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Chan AC, Desai DM, Weiss A. 1994. The role of protein tyrosine kinases and protein tyrosine phosphatases in T cell antigen receptor signal transduction. Annu Rev Immunol. 12:555–592. [DOI] [PubMed] [Google Scholar]
- Chang YW, Tseng CF, Wang MY, Chang WC, Lee CC, Chen LT, Hung MC, Su JL. 2016. Deacetylation of HSPA5 by HDAC6 leads to GP78-mediated HSPA5 ubiquitination at K447 and suppresses metastasis of breast cancer. Oncogene. 35(12): 1517–1528. [DOI] [PubMed] [Google Scholar]
- Chapman DC, Williams DB. 2010. ER quality control in the biogenesis of MHC class I molecules. Semin Cell Dev Biol. 21(5):512–519. [DOI] [PubMed] [Google Scholar]
- Chen B, Retzlaff M, Roos T, Frydman J. 2011. Cellular strategies of protein quality control. Cold Spring Harb Perspect Biol. 3(8):a004374. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Chen K, Han M, Tang M, Xie Y, Lai Y, Hu X, Zhang J, Yang J, Li H. 2018. Differential Hrd1 expression and B-cell accumulation in eosinophilic and non-eosinophilic chronic rhinosinusitis with nasal polyps. Allergy Asthma Immunol Res. 10(6):698–715. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Christianson JC, Olzmann JA, Shaler TA, Sowa ME, Bennett EJ, Richter CM, Tyler RE, Greenblatt EJ, Harper JW, Kopito RR. 2011. Defining human ERAD networks through an integrative mapping strategy. Nat Cell Biol. 14(1):93–105. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Christianson JC, Shaler TA, Tyler RE, Kopito RR. 2008. OS-9 and GRP94 deliver mutant alpha1-antitrypsin to the Hrd1-SEL1L ubiquitin ligase complex for ERAD. Nat Cell Biol. 10(3):272–282. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Christianson JC, Ye Y. 2014. Cleaning up in the endoplasmic reticulum: ubiquitin in charge. Nat Struct Mol Biol. 21(4): 325–335. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Condamine T, Dominguez GA, Youn JI, Kossenkov AV, Mony S, Alicea-Torres K, Tcyganov E, Hashimoto A, Nefedova Y, Lin C, et al. 2016. Lectin-type oxidized LDL receptor-1 distinguishes population of human polymorphonuclear myeloid-derived suppressor cells in cancer patients. Sci Immunol. 1(2):aaf8943. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Condamine T, Kumar V, Ramachandran IR, Youn JI, Celis E, Finnberg N, El-Deiry WS, Winograd R, Vonderheide RH, English NR, et al. 2014. ER stress regulates myeloid-derived suppressor cell fate through TRAIL-R-mediated apoptosis. J Clin Invest. 124(6):2626–2639. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Cormier JH, Tamura T, Sunryd JC, Hebert DN. 2009. EDEM1 recognition and delivery of misfolded proteins to the SEL1L-containing ERAD complex. Mol Cell. 34(5):627–633. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Costa RLB, Czerniecki BJ. 2020. Clinical development of immunotherapies for HER2+ breast cancer: a review of HER2-directed monoclonal antibodies and beyond. Npj Breast Cancer. 6(1):10. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Cruz FM, Colbert JD, Merino E, Kriegsman BA, Rock KL. 2017. The Biology and Underlying Mechanisms of Cross-Presentation of Exogenous Antigens on MHC-I Molecules. Annu Rev Immunol. 35:149–176. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Cubillos-Ruiz JR, Bettigole SE, Glimcher LH. 2017. Tumorigenic and Immunosuppressive Effects of Endoplasmic Reticulum Stress in Cancer. Cell. 168(4): 692–706. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Cubillos-Ruiz JR, Glimcher LH. 2016. Targeting abnormal ER stress responses in tumors: A new approach to cancer immunotherapy. Oncoimmunology. 5(3):e1098802. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Cubillos-Ruiz JR, Silberman PC, Rutkowski MR, Chopra S, Perales-Puchalt A, Song M, Zhang S, Bettigole SE, Gupta D, Holcomb K, et al. 2015. ER stress sensor XBP1 controls anti-tumor immunity by disrupting dendritic cell homeostasis. Cell. 161(7):1527–1538. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Cummings CJ, Reinstein E, Sun Y, Antalffy B, Jiang Y, Ciechanover A, Orr HT, Beaudet AL, Zoghbi HY. 1999. Mutation of the E6-AP ubiquitin ligase reduces nuclear inclusion frequency while accelerating polyglutamineinduced pathology in SCA1 mice. Neuron. 24(4):879–892. [DOI] [PubMed] [Google Scholar]
- D’Alessio C, Caramelo JJ, Parodi AJ. 2010. UDP-GlC:glycoprotein glucosyltransferase-glucosidase II, the ying-yang of the ER quality control. Semin Cell Dev Biol. 21(5):491–499. [DOI] [PMC free article] [PubMed] [Google Scholar]
- D’Alessio C, Dahms NM. 2015. Glucosidase II and MRH-domain containing proteins in the secretory pathway. Curr Protein Pept Sci. 16(1):31–48. [DOI] [PMC free article] [PubMed] [Google Scholar]
- de Charette M, Marabelle A, Houot R. 2016. Turning tumour cells into antigen presenting cells: The next step to improve cancer immunotherapy? Eur J Cancer. 68: 134–147. [DOI] [PubMed] [Google Scholar]
- Degli-Esposti MA, Smyth MJ. 2005. Close encounters of different kinds: dendritic cells and NK cells take centre stage. Nat Rev Immunol. 5(2):112–124. [DOI] [PubMed] [Google Scholar]
- DeLaBarre B, Brunger AT. 2003. Complete structure of p97/valosin-containing protein reveals communication between nucleotide domains. Nat Struct Biol. 10(10): 856–863. [DOI] [PubMed] [Google Scholar]
- Deng J, Wang ES, Jenkins RW, Li S, Dries R, Yates K, Chhabra S, Huang W, Liu H, Aref AR, et al. 2018. CDK4/6 inhibition augments antitumor immunity by enhancing T-cell activation. Cancer Discov. 8(2):216–233. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Denic V, Quan EM, Weissman JS. 2006. A luminal surveillance complex that selects misfolded glycoproteins for ER-associated degradation. Cell. 126(2):349–359. [DOI] [PubMed] [Google Scholar]
- Deshaies RJ, Sanders SL, Feldheim DA, Schekman R. 1991. Assembly of yeast Sec proteins involved in translocation into the endoplasmic reticulum into a membrane-bound multisubunit complex. Nature. 349(6312):806–808. [DOI] [PubMed] [Google Scholar]
- Dong M, Bridges JP, Apsley K, Xu Y, Weaver TE. 2008. ERdj4 and ERdj5 are required for endoplasmic reticulum-associated protein degradation of misfolded surfactant protein C. Mol Biol Cell. 19(6):2620–2630. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Dong Q, Fu L, Zhao Y, Tan S, Wang E. 2017. Derlin-1 overexpression confers poor prognosis in muscle invasive bladder cancer and contributes to chemoresistance and invasion through PI3K/AKT and ERK/MMP signaling. Oncotarget. 8(10):17059–17069. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Dorner AJ, Wasley LC, Kaufman RJ. 1992. Overexpression of GRP78 mitigates stress induction of glucose regulated proteins and blocks secretion of selective proteins in Chinese hamster ovary cells. Embo J. 11(4):1563–1571. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Dreher LS, Hoppe T. 2018. ERADicate tumor progression with metformin. Mol Cell. 71(4):481–482. [DOI] [PubMed] [Google Scholar]
- Dumont P, Berton A, Nagy N, Sandras F, Tinton S, Demetter P, Mascart F, Allaoui A, Decaestecker, Salmon. 2008. Expression of galectin-3 in the tumor immune response in colon cancer. Lab Invest. 88(8):896–906. [DOI] [PubMed] [Google Scholar]
- Emadi A, Jones RJ, Brodsky RA. 2009. Cyclophosphamide and cancer: golden anniversary. Nat Rev Clin Oncol. 6(11): 638–647. [DOI] [PubMed] [Google Scholar]
- Embgenbroich M, Burgdorf S. 2018. Current concepts of antigen cross-presentation. Front Immunol. 9:1643. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Ernst R, Mueller B, Ploegh HL, Schlieker C. 2009. The otubain YOD1 is a deubiquitinating enzyme that associates with p97 to facilitate protein dislocation from the ER. Mol Cell. 36(1):28–38. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Fagioli C, Sitia R. 2001. Glycoprotein quality control in the endoplasmic reticulum. Mannose trimming by endoplasmic reticulum mannosidase I times the proteasomal degradation of unassembled immunoglobulin subunits. J Biol Chem. 276(16):12885–12892. [DOI] [PubMed] [Google Scholar]
- Fang S, Ferrone M, Yang C, Jensen JP, Tiwari S, Weissman AM. 2001. The tumor autocrine motility factor receptor, gp78, is a ubiquitin protein ligase implicated in degradation from the endoplasmic reticulum. Proc Natl Acad Sci USA. 98(25):14422–14427. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Fisher EA, Ginsberg HN. 2002. Complexity in the secretory pathway: the assembly and secretion of apolipoprotein B-containing lipoproteins. J Biol Chem. 277(20):17377–17380. [DOI] [PubMed] [Google Scholar]
- Fucikova J, Becht E, Iribarren K, Goc J, Remark R, Damotte D, Alifano M, Devi P, Biton J, Germain C, et al. 2016. Calreticulin expression in human non-small cell lung cancers correlates with increased accumulation of antitumor immune cells and favorable prognosis. Cancer Res. 76(7): 1746–1756. [DOI] [PubMed] [Google Scholar]
- Fucikova J, Kralikova P, Fialova A, Brtnicky T, Rob L, Bartunkova J, Spisek R. 2011. Human tumor cells killed by anthracyclines induce a tumor-specific immune response. Cancer Res. 71(14):4821–4833. [DOI] [PubMed] [Google Scholar]
- Gardai SJ, McPhillips KA, Frasch SC, Janssen WJ, Starefeldt A, Murphy-Ullrich JE, Bratton DL, Oldenborg PA, Michalak M, Henson PM. 2005. Cell-surface calreticulin initiates clearance of viable or apoptotic cells through trans-activation of LRP on the phagocyte. Cell. 123(2):321–334. [DOI] [PubMed] [Google Scholar]
- Gardner BM, Walter P. 2011. Unfolded proteins are Ire1-activating ligands that directly induce the unfolded protein response. Science. 333(6051):1891–1894. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Gardner RG, Swarbrick GM, Bays NW, Cronin SR, Wilhovsky S, Seelig L, Kim C, Hampton RY. 2000. Endoplasmic reticulum degradation requires lumen to cytosol signaling. Transmembrane control of Hrd1p by Hrd3p. J Cell Biol. 151(1):69–82. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Garg AD, Krysko DV, Verfaillie T, Kaczmarek A, Ferreira GB, Marysael T, Rubio N, Firczuk M, Mathieu C, Roebroek AJ, et al. 2012. A novel pathway combining calreticulin exposure and ATP secretion in immunogenic cancer cell death. Embo J. 31(5):1062–1079. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Garg AD, More S, Rufo N, Mece O, Sassano ML, Agostinis P, Zitvogel L, Kroemer G, Galluzzi L. 2017. Trial watch: Immunogenic cell death induction by anticancer chemotherapeutics. Oncoimmunology. 6(12):e1386829. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Garg AD, Nowis D, Golab J, Vandenabeele P, Krysko DV, Agostinis P. 2010. Immunogenic cell death, DAMPs and anticancer therapeutics: an emerging amalgamation. Biochim Biophys Acta. 1805(1):53–71. [DOI] [PubMed] [Google Scholar]
- Gass JN, Gifford NM, Brewer JW. 2002. Activation of an unfolded protein response during differentiation of antibody-secreting B cells. J Biol Chem. 277(50):49047–49054. [DOI] [PubMed] [Google Scholar]
- Gauss R, Jarosch E, Sommer T, Hirsch C. 2006. A complex of Yos9p and the HRD ligase integrates endoplasmic reticulum quality control into the degradation machinery. Nat Cell Biol. 8(8):849–854. [DOI] [PubMed] [Google Scholar]
- Germain RN. 2002. T-cell development and the CD4-CD8 lineage decision. Nat Rev Immunol. 2(5):309–322. [DOI] [PubMed] [Google Scholar]
- Ghiringhelli F, Apetoh L, Tesniere A, Aymeric L, Ma Y, Ortiz C, Vermaelen K, Panaretakis T, Mignot G, Ullrich E, et al. 2009. Activation of the NLRP3 inflammasome in dendritic cells induces IL-1beta-dependent adaptive immunity against tumors. Nat Med. 15(10):1170–1178. [DOI] [PubMed] [Google Scholar]
- Giglio P, Gagliardi M, Tumino N, Antunes F, Smaili S, Cotella D, Santoro C, Bernardini R, Mattei M, Piacentini M, et al. 2018. PKR and GCN2 stress kinases promote an ER stressindependent eIF2α phosphorylation responsible for calreticulin exposure in melanoma cells. Oncoimmunology. 7(8): e1466765. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Gillece P, Luz JM, Lennarz WJ, de La Cruz FJ, Romisch K. 1999. Export of a cysteine-free misfolded secretory protein from the endoplasmic reticulum for degradation requires interaction with protein disulfide isomerase. J Cell Biol. 147(7):1443–1456. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Goel S, DeCristo MJ, Watt AC, BrinJones H, Sceneay J, Li BB, Khan N, Ubellacker JM, Xie S, Metzger-Filho O, et al. 2017. CDK4/6 inhibition triggers anti-tumour immunity. Nature. 548(7668):471–475. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Gong B, Chen F, Pan Y, Arrieta-Cruz I, Yoshida Y, Haroutunian V, Pasinetti GM. 2010. SCFFbx2-E3-ligase-mediated degradation of BACE1 attenuates Alzheimer’s disease amyloidosis and improves synaptic function. Aging Cell. 9(6):1018–1031. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Gong J, Fang L, Liu R, Wang Y, Xing J, Chen Y, Zhuang R, Zhang Y, Zhang C, Yang A, et al. 2014. UPR decreases CD226 ligand CD155 expression and sensitivity to NK cell-mediated cytotoxicity in hepatoma cells. Eur J Immunol. 44(12):3758–3767. [DOI] [PubMed] [Google Scholar]
- Granados DP, Tanguay PL, Hardy MP, Caron E, de Verteuil D, Meloche S, Perreault C. 2009. ER stress affects processing of MHC class I-associated peptides. BMC Immunol. 10:10. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Greenblatt EJ, Olzmann JA, Kopito RR. 2011. Derlin-1 is a rhomboid pseudoprotease required for the dislocation of mutant α-1 antitrypsin from the endoplasmic reticulum. Nat Struct Mol Biol. 18(10):1147–1152. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Grootjans J, Kaser A, Kaufman RJ, Blumberg RS. 2016. The unfolded protein response in immunity and inflammation. Nat Rev Immunol. 16(8):469–484. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Grotzke JE, Cresswell P. 2015. Are ERAD components involved in cross-presentation? Mol Immunol. 68(2 Pt A): 112–115. [DOI] [PubMed] [Google Scholar]
- Grotzke JE, Kozik P, Morel JD, Impens F, Pietrosemoli N, Cresswell P, Amigorena S, Demangel C. 2017. Sec61 blockade by mycolactone inhibits antigen cross-presentation independently of endosome-to-cytosol export. Proc Natl Acad Sci USA. 114(29):E5910–E5919. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Guermonprez P, Valladeau J, Zitvogel L, Thery C, Amigorena S. 2002. Antigen presentation and T cell stimulation by dendritic cells. Annu Rev Immunol. 20:621–667. [DOI] [PubMed] [Google Scholar]
- Guerriero CJ, Brodsky JL. 2012. The delicate balance between secreted protein folding and endoplasmic reticulum-associated degradation in human physiology. Physiol Rev. 92(2):537–576. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Guiliano DB, Fussell H, Lenart I, Tsao E, Nesbeth D, Fletcher AJ, Campbell EC, Yousaf N, Williams S, Santos S, et al. 2014. Endoplasmic reticulum degradation-enhancing α-mannosidase-like protein 1 targets misfolded HLA-B27 dimers for endoplasmic reticulum-associated degradation. Arthritis Rheumatol. 66(11):2976–2988. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Guo X, Shen S, Song S, He S, Cui Y, Xing G, Wang J, Yin Y, Fan L, He F, et al. 2011. The E3 ligase Smurf1 regulates Wolfram syndrome protein stability at the endoplasmic reticulum. J Biol Chem. 286(20):18037–18047. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Hammond C, Helenius A. 1994. Folding of VSV G protein: sequential interaction with BiP and calnexin. Science. 266(5184):456–458. [DOI] [PubMed] [Google Scholar]
- Hampton RY. 2002. ER-associated degradation in protein quality control and cellular regulation. Curr Opin Cell Biol. 14(4):476–482. [DOI] [PubMed] [Google Scholar]
- Harding HP, Zhang Y, Ron D. 1999. Protein translation and folding are coupled by an endoplasmic-reticulum-resident kinase. Nature. 397(6716):271–274. [DOI] [PubMed] [Google Scholar]
- Harris J, Sengar D, Stewart T, Hyslop D. 1976. The effect of immunosuppressive chemotherapy on immune function in patients with malignant disease. Cancer. 37(S2):1058–1069. [DOI] [PubMed] [Google Scholar]
- Hassink G, Kikkert M, van Voorden S, Lee SJ, Spaapen R, van Laar T, Coleman CS, Bartee E, Fruh K, Chau V, et al. 2005. TEB4 is a C4HC3 RING finger-containing ubiquitin ligase of the endoplasmic reticulum. Biochem J. 388(Pt 2):647–655. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Hassink GC, Zhao B, Sompallae R, Altun M, Gastaldello S, Zinin NV, Masucci MG, Lindsten K. 2009. The ER-resident ubiquitin-specific protease 19 participates in the UPR and rescues ERAD substrates. EMBO Rep. 10(7):755–761. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Heath WR, Carbone FR. 2001. Cross-presentation in viral immunity and self-tolerance. Nat Rev Immunol. 1(2): 126–134. [DOI] [PubMed] [Google Scholar]
- Heine A, Held SA, Bringmann A, Holderried TA, Brossart P. 2011. Immunomodulatory effects of anti-angiogenic drugs. Leukemia. 25(6):899–905. [DOI] [PubMed] [Google Scholar]
- Hirao K, Natsuka Y, Tamura T, Wada I, Morito D, Natsuka S, Romero P, Sleno B, Tremblay LO, Herscovics A, et al. 2006. EDEM3, a soluble EDEM homolog, enhances glycoprotein endoplasmic reticulum-associated degradation and mannose trimming. J Biol Chem. 281(14):9650–9658. [DOI] [PubMed] [Google Scholar]
- Ho SC, Chaudhuri S, Bachhawat A, McDonald K, Pillai S. 2000. Accelerated proteasomal degradation of membrane ig heavy chains. J Immunol. 164(9):4713–4719. [DOI] [PubMed] [Google Scholar]
- Hollien J, Weissman JS. 2006. Decay of endoplasmic reticulum-localized mRNAs during the unfolded protein response. Science. 313(5783):104–107. [DOI] [PubMed] [Google Scholar]
- Hoppe T 2005. Multiubiquitylation by E4 enzymes: ‘one size’ doesn’t fit all. Trends Biochem Sci. 30(4):183–187. [DOI] [PubMed] [Google Scholar]
- Horn SC, Hanna J, Hirsch C, Volkwein C, Schutz A, Heinemann U, Sommer T, Jarosch E. 2009. Usa1 functions as a scaffold of the HRD-ubiquitin ligase. Mol Cell. 36(5): 782–793. [DOI] [PubMed] [Google Scholar]
- Horta ZP, Goldberg JL, Sondel PM. 2016. Anti-GD2 mAbs and next-generation mAb-based agents for cancer therapy. Immunotherapy. 8(9):1097–1117. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Hosokawa N, Tremblay LO, Sleno B, Kamiya Y, Wada I, Nagata K, Kato K, Herscovics A. 2010. EDEM1 accelerates the trimming of alpha1,2-linked mannose on the C branch of N-glycans. Glycobiology. 20(5):567–575. [DOI] [PubMed] [Google Scholar]
- Hosokawa N, Wada I, Hasegawa K, Yorihuzi T, Tremblay LO, Herscovics A, Nagata K. 2001. A novel ER alpha-mannosidase-like protein accelerates ER-associated degradation. EMBO Rep. 2(5):415–422. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Hotamisligil GS. 2010. Endoplasmic reticulum stress and the inflammatory basis of metabolic disease. Cell. 140(6): 900–917. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Houde M, Bertholet S, Gagnon E, Brunet S, Goyette G, Laplante A, Princiotta MF, Thibault P, Sacks D, Desjardins M. 2003. Phagosomes are competent organelles for antigen cross-presentation. Nature. 425(6956):402–406. [DOI] [PubMed] [Google Scholar]
- Howard JC. 1995. Supply and transport of peptides presented by class I MHC molecules. Curr Opin Immunol. 7(1): 69–76. [DOI] [PubMed] [Google Scholar]
- Hughes A, Clarson J, Tang C, Vidovic L, White DL, Hughes TP, Yong A. 2017. CML patients with deep molecular responses to TKI have restored immune effectors and decreased PD-1 and immune suppressors. Blood. 129(9): 1166–1176. [DOI] [PubMed] [Google Scholar]
- Hughes A, Yong A. 2017. Immune effector recovery in chronic myeloid leukemia and treatment-free remission. Front Immunol. 8:469. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Hughes EA, Hammond C, Cresswell P. 1997. Misfolded major histocompatibility complex class I heavy chains are translocated into the cytoplasm and degraded by the proteasome. Proc Natl Acad Sci USA. 94(5):1896–1901. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Huiting LN, Samaha Y, Zhang GL, Roderick JE, Li B, Anderson NM, Wang YW, Wang L, Laroche F, Choi JW, et al. 2018. UFD1 contributes to MYC-mediated leukemia aggressiveness through suppression of the proapoptotic unfolded protein response. Leukemia. 32(11):2339–2351. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Huppa JB, Ploegh HL. 1997. The alpha chain of the T cell antigen receptor is degraded in the cytosol. Immunity. 7(1):113–122. [DOI] [PubMed] [Google Scholar]
- Huyton T, Pye VE, Briggs LC, Flynn TC, Beuron F, Kondo H, Ma J, Zhang X, Freemont PS. 2003. The crystal structure of murine p97/VCP at 3.6A. J Struct Biol. 144(3):337–348. [DOI] [PubMed] [Google Scholar]
- Imai J, Hasegawa H, Maruya M, Koyasu S, Yahara I. 2005. Exogenous antigens are processed through the endoplasmic reticulum-associated degradation (ERAD) in cross-presentation by dendritic cells. Int Immunol. 17(1):45–53. [DOI] [PubMed] [Google Scholar]
- Inoue H, Tani K. 2014. Multimodal immunogenic cancer cell death as a consequence of anticancer cytotoxic treatments. Cell Death Differ. 21(1):39–49. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Inoue T, Tsai B. 2016. The Grp170 nucleotide exchange factor executes a key role during ERAD of cellular misfolded clients. Mol Biol Cell. 27(10):1650–1662. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Ishikura S, Weissman AM, Bonifacino JS. 2010. Serine residues in the cytosolic tail of the T-cell antigen receptor alpha-chain mediate ubiquitination and endoplasmic reticulum-associated degradation of the unassembled protein. J Biol Chem. 285(31):23916–23924. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Jaenicke LA, Brendebach H, Selbach M, Hirsch C. 2011. Yos9p assists in the degradation of certain nonglycosylated proteins from the endoplasmic reticulum. Mol Biol Cell. 22(16):2937–2945. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Jahn TR, Radford SE. 2005. The Yin and Yang of protein folding. Febs J. 272(23):5962–5970. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Janssens S, Pulendran B, Lambrecht BN. 2014. Emerging functions of the unfolded protein response in immunity. Nat Immunol. 15(10):910–919. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Jarosch E, Taxis C, Volkwein C, Bordallo J, Finley D, Wolf DH, Sommer T. 2002. Protein dislocation from the ER requires polyubiquitination and the AAA-ATPase Cdc48. Nat Cell Biol. 4(2):134–139. [DOI] [PubMed] [Google Scholar]
- Jensen PE. 2007. Recent advances in antigen processing and presentation. Nat Immunol. 8(10):1041–1048. [DOI] [PubMed] [Google Scholar]
- Jeon YJ, Khelifa S, Ratnikov B, Scott DA, Feng Y, Parisi F, Ruller C, Lau E, Kim H, Brill LM, et al. 2015. Regulation of glutamine carrier proteins by RNF5 determines breast cancer response to ER stress-inducing chemotherapies. Cancer Cell. 27(3):354–369. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Ji Y, Kim H, Yang L, Sha H, Roman CA, Long Q, Qi L. 2016. The Sel1L-Hrd1 endoplasmic reticulum-associated degradation complex manages a key checkpoint in B cell development. Cell Rep. 16(10):2630–2640. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Jo Y, Hartman IZ, DeBose-Boyd RA. 2013. Ancient ubiquitous protein-1 mediates sterol-induced ubiquitination of 3-hydroxy-3-methylglutaryl CoA reductase in lipid droplet-associated endoplasmic reticulum membranes. Mol Biol Cell. 24(3):169–183. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Joffre OP, Segura E, Savina A, Amigorena S. 2012. Cross-presentation by dendritic cells. Nat Rev Immunol. 12(8): 557–569. [DOI] [PubMed] [Google Scholar]
- Ju LG, Lin X, Yan D, Li QL, Wu M, Li LY. 2018. Characterization of WDR20: A new regulator of the ERAD machinery. Biochim Biophys Acta Mol Cell Res. 1865(7): 970–980. [DOI] [PubMed] [Google Scholar]
- Junttila MR, de Sauvage FJ. 2013. Influence of tumour microenvironment heterogeneity on therapeutic response. Nature. 501(7467):346–354. [DOI] [PubMed] [Google Scholar]
- Kajiro M, Hirota R, Nakajima Y, Kawanowa K, So-Ma K, Ito I, Yamaguchi Y, Ohie SH, Kobayashi Y, Seino Y, et al. 2009. The ubiquitin ligase CHIP acts as an upstream regulator of oncogenic pathways. Nat Cell Biol. 11(3):312–319. [DOI] [PubMed] [Google Scholar]
- Kamphorst AO, Guermonprez P, Dudziak D, Nussenzweig MC. 2010. Route of antigen uptake differentially impacts presentation by dendritic cells and activated monocytes. J Immunol. 185(6):3426–3435. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Kanehara K, Xie W, Ng DT. 2010. Modularity of the Hrd1 ERAD complex underlies its diverse client range. J Cell Biol. 188(5):707–716. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Kasai M, Tanida I, Ueno T, Kominami E, Seki S, Ikeda T, Mizuochi T. 2009. Autophagic compartments gain access to the MHC class II compartments in thymic epithelium. J Immunol. 183(11):7278–7285. [DOI] [PubMed] [Google Scholar]
- Kepp O, Menger L, Vacchelli E, Locher C, Adjemian S, Yamazaki T, Martins I, Sukkurwala AQ, Michaud M, Senovilla L, et al. 2013. Crosstalk between ER stress and immunogenic cell death. Cytokine Growth Factor Rev. 24(4):311–318. [DOI] [PubMed] [Google Scholar]
- Kikkert M, Doolman R, Dai M, Avner R, Hassink G, van Voorden S, Thanedar S, Roitelman J, Chau V, Wiertz E. 2004. Human HRD1 is an E3 ubiquitin ligase involved in degradation of proteins from the endoplasmic reticulum. J Biol Chem. 279(5):3525–3534. [DOI] [PubMed] [Google Scholar]
- Kim I, Ahn J, Liu C, Tanabe K, Apodaca J, Suzuki T, Rao H. 2006. The Png1-Rad23 complex regulates glycoprotein turnover. J Cell Biol. 172(2):211–219. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Kim JL, Lee DH, Jeong S, Kim BR, Na YJ, Park SH, Jo MJ, Jeong YA, Oh SC. 2019. Imatinib-induced apoptosis of gastric cancer cells is mediated by endoplasmic reticulum stress. Oncol Rep. 41(3):1616–1626. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Kim SJ, Gregersen PK, Diamond B. 2013. Regulation of dendritic cell activation by microRNA let-7c and BLIMP1. J Clin Invest. 123(2):823–833. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Kim SJ, Zou YR, Goldstein J, Reizis B, Diamond B. 2011. Tolerogenic function of Blimp-1 in dendritic cells. J Exp Med. 208(11):2193–2199. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Kim TY, Kim E, Yoon SK, Yoon JB. 2008. Herp enhances ER-associated protein degradation by recruiting ubiquilins. Biochem Biophys Res Commun. 369(2):741–746. [DOI] [PubMed] [Google Scholar]
- Kimiz-Gebologlu I, Gulce-Iz S, Biray-Avci C. 2018. Monoclonal antibodies in cancer immunotherapy. Mol Biol Rep. 45(6): 2935–2940. [DOI] [PubMed] [Google Scholar]
- Kimura H, Sakai K, Arao T, Shimoyama T, Tamura T, Nishio K. 2007. Antibody-dependent cellular cytotoxicity of cetuximab against tumor cells with wild-type or mutant epidermal growth factor receptor. Cancer Science. 98(8): 1275–1280. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Kohlmann S, Schafer A, Wolf DH. 2008. Ubiquitin ligase Hul5 is required for fragment-specific substrate degradation in endoplasmic reticulum-associated degradation. J Biol Chem. 283(24):16374–16383. [DOI] [PubMed] [Google Scholar]
- Kong S, Yang Y, Xu Y, Wang Y, Zhang Y, Melo-Cardenas J, Xu X, Gao B, Thorp EB, Zhang DD, et al. 2016. Endoplasmic reticulum-resident E3 ubiquitin ligase Hrd1 controls B-cell immunity through degradation of the death receptor CD95/Fas. Proc Natl Acad Sci USA. 113(37):10394–10399. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Koopmann JO, Albring J, Huter E, Bulbuc N, Spee P, Neefjes J, Hammerling GJ, Momburg F. 2000. Export of antigenic peptides from the endoplasmic reticulum intersects with retrograde protein translocation through the Sec61p channel. Immunity. 13(1):117–127. [DOI] [PubMed] [Google Scholar]
- Kovacsovics-Bankowski M, Rock KL. 1995. A phagosome-to-cytosol pathway for exogenous antigens presented on MHC class I molecules. Science. 267(5195):243–246. [DOI] [PubMed] [Google Scholar]
- Krysko DV, Garg AD, Kaczmarek A, Krysko O, Agostinis P, Vandenabeele P. 2012. Immunogenic cell death and DAMPs in cancer therapy. Nat Rev Cancer. 12(12):860–875. [DOI] [PubMed] [Google Scholar]
- Kubagawa H, Honjo K, Ohkura N, Sakaguchi S, Radbruch A, Melchers F, Jani PK. 2019. Functional Roles of the IgM Fc Receptor in the Immune System. Front Immunol. 10:945. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Kurts C, Robinson BW, Knolle PA. 2010. Cross-priming in health and disease. Nat Rev Immunol. 10(6):403–414. [DOI] [PubMed] [Google Scholar]
- Labonte AC, Tosello-Trampont AC, Hahn YS. 2014. The role of macrophage polarization in infectious and inflammatory diseases. Mol Cells. 37(4):275–285. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Larmonier N, Janikashvili N, LaCasse CJ, Larmonier CB, Cantrell J, Situ E, Lundeen T, Bonnotte B, Katsanis E. 2008. Imatinib mesylate inhibits CD4+ CD25+ regulatory T cell activity and enhances active immunotherapy against BCR-ABL- tumors. J Immunol. 181(10):6955–6963. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Lass A, McConnell E, Nowis D, Mechref Y, Kang P, Novotny MV, Wójcik C. 2007. A novel function of VCP (valosin-con-taining protein; p97) in the control of N-glycosylation of proteins in the endoplasmic reticulum. Arch Biochem Biophys. 462(1):62–73. [DOI] [PMC free article] [PubMed] [Google Scholar]
- LeBien TW, Tedder TF. 2008. B lymphocytes: how they develop and function. Blood. 112(5):1570–1580. 5):1570–1580. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Lee AH, Iwakoshi NN, Glimcher LH. 2003. XBP-1 regulates a subset of endoplasmic reticulum resident chaperone genes in the unfolded protein response. Mol Cell Biol. 23(21):7448–7459. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Lee AS. 2007. GRP78 Induction in Cancer: therapeutic and Prognostic Implications: Figure 1. Cancer Res. 67(8): 3496–3499. [DOI] [PubMed] [Google Scholar]
- Lee BR, Chang SY, Hong EH, Kwon BE, Kim HM, Kim YJ, Lee J, Cho HJ, Cheon JH, Ko HJ. 2014. Elevated endoplasmic reticulum stress reinforced immunosuppression in the tumor microenvironment via myeloid-derived suppressor cells. Oncotarget. 5(23):12331–12345. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Lee J, Choi TG, Ha J, Kim SS. 2012. Cyclosporine A suppresses immunoglobulin G biosynthesis via inhibition of cyclophilin B in murine hybridomas and B cells. Int Immunopharmacol. 12(1):42–49. [DOI] [PubMed] [Google Scholar]
- Legler K, Rosprim R, Karius T, Eylmann K, Rossberg M, Wirtz RM, Muller V, Witzel I, Schmalfeldt B, Milde-Langosch K, et al. 2018. Reduced mannosidase MAN1A1 expression leads to aberrant N-glycosylation and impaired survival in breast cancer. Br J Cancer. 118(6):847–856. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Lenk U, Yu H, Walter J, Gelman MS, Hartmann E, Kopito RR, Sommer T. 2002. A role for mammalian Ubc6 homologues in ER- associated protein degradation. J Cell Sci. 115(Pt 14):3007–3014. [DOI] [PubMed] [Google Scholar]
- Leone P, Shin EC, Perosa F, Vacca A, Dammacco F, Racanelli V. 2013. MHC class I antigen processing and presenting machinery: organization, function, and defects in tumor cells. J Natl Cancer Inst. 105(16):1172–1187. [DOI] [PubMed] [Google Scholar]
- Lerner M, Corcoran M, Cepeda D, Nielsen ML, Zubarev R, Pontén F, Uhlén M, Hober S, Grandér D, Sangfelt O. 2007. The RBCC gene RFP2 (Leu5) encodes a novel transmembrane E3 ubiquitin ligase involved in ERAD. Mol Biol Cell. 18(5):1670–1682. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Li X, Lian Z, Wang S, Xing L, Yu J. 2018. Interactions between EGFR and PD-1/PD-L1 pathway: Implications for treatment of NSCLC. Cancer Lett. 418:1–9. [DOI] [PubMed] [Google Scholar]
- Li Y, Wang LX, Pang P, Cui Z, Aung S, Haley D, Fox BA, Urba WJ, Hu HM. 2011. Tumor-derived autophagosome vaccine: mechanism of cross-presentation and therapeutic efficacy. Clin Cancer Res. 17(22):7047–7057. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Li Y, Xie Y, Hao J, Liu J, Ning Y, Tang Q, Ma M, Zhou H, Guan S, Zhou Q, et al. 2018. ER-localized protein-Herpud1 is a new mediator of IL-4-induced macrophage polarization and migration. Exp Cell Res. 368(2):167–173. [DOI] [PubMed] [Google Scholar]
- Liang J, Yin C, Doong H, Fang S, Peterhoff C, Nixon RA, Monteiro MJ. 2006. Characterization of erasin (UBXD2): a new ER protein that promotes ER-associated protein degradation. J Cell Sci. 119(Pt 19):4011–4024. [DOI] [PubMed] [Google Scholar]
- Lilley BN, Ploegh HL. 2004. A membrane protein required for dislocation of misfolded proteins from the ER. NATURE. 429(6994):834–840. [DOI] [PubMed] [Google Scholar]
- Lippincott-Schwartz J, Bonifacino JS, Yuan LC, Klausner RD. 1988. Degradation from the endoplasmic reticulum: disposing of newly synthesized proteins. Cell. 54(2):209–220. [DOI] [PubMed] [Google Scholar]
- Liu T, Chambers B, Diehl AD, Van Kaer L, Jondal M, Ljunggren HG. 1997. TAP peptide transporter-independent presentation of heat-killed Sendai virus antigen on MHC class I molecules by splenic antigen-presenting cells. J Immunol. 159(11):5364–5371. [PubMed] [Google Scholar]
- Liu Y, Ye Y. 2011. Proteostasis regulation at the endoplasmic reticulum: a new perturbation site for targeted cancer therapy. Cell Res. 21(6):867–883. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Ma W, Zhang Y, Vigneron N, Stroobant V, Thielemans K, van der Bruggen P, Van den Eynde BJ. 2016. Long-peptide cross-presentation by human dendritic cells occurs in vacuoles by peptide exchange on nascent MHC Class I molecules. J Immunol. 196(4):1711–1720. [DOI] [PubMed] [Google Scholar]
- Ma XH, Piao SF, Dey S, McAfee Q, Karakousis G, Villanueva J, Hart LS, Levi S, Hu J, Zhang G, et al. 2014. Targeting ER stress-induced autophagy overcomes BRAF inhibitor resistance in melanoma. J Clin Invest. 124(3):1406–1417. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Ma Y, Brewer JW, Diehl JA, Hendershot LM. 2002. Two distinct stress signaling pathways converge upon the CHOP promoter during the mammalian unfolded protein response. J Mol Biol. 318(5):1351–1365. [DOI] [PubMed] [Google Scholar]
- Ma Y, Conforti R, Aymeric L, Locher C, Kepp O, Kroemer G, Zitvogel L. 2011. How to improve the immunogenicity of chemotherapy and radiotherapy. Cancer Metastasis Rev. 30(1):71–82. [DOI] [PubMed] [Google Scholar]
- Ma Y, Hendershot LM. 2004. The role of the unfolded protein response in tumour development: friend or foe?. Nat Rev Cancer. 4(12):966–977. [DOI] [PubMed] [Google Scholar]
- Magadan JG, Perez-Victoria FJ, Sougrat R, Ye Y, Strebel K, Bonifacino JS. 2010. Multilayered mechanism of CD4 downregulation by HIV-1 Vpu involving distinct ER retention and ERAD targeting steps. PLoS Pathog. 6(4): e1000869. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Magnaghi P, D’Alessio R, Valsasina B, Avanzi N, Rizzi S, Asa D, Gasparri F, Cozzi L, Cucchi U, Orrenius C, et al. 2013. Covalent and allosteric inhibitors of the ATPase VCP/p97 induce cancer cell death. Nat Chem Biol. 9(9):548–556. [DOI] [PubMed] [Google Scholar]
- Mahadevan NR, Anufreichik V, Rodvold JJ, Chiu KT, Sepulveda H, Zanetti M. 2012. Cell-extrinsic effects of tumor ER stress imprint myeloid dendritic cells and impair CD8(+) T cell priming. PLoS One. 7(12):e51845. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Mahadevan NR, Rodvold J, Sepulveda H, Rossi S, Drew AF, Zanetti M. 2011. Transmission of endoplasmic reticulum stress and pro-inflammation from tumor cells to myeloid cells. Proc Natl Acad Sci USA. 108(16):6561–6566. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Mao M, Zhang J, Jiang J. 2018. Overexpression of Derlin-1 is associated with poor prognosis in patients with non-small cell lung cancer. Ann Clin Lab Sci. 48(1):29–34. [PubMed] [Google Scholar]
- Marciniak SJ, Yun CY, Oyadomari S, Novoa I, Zhang Y, Jungreis R, Nagata K, Harding HP, Ron D. 2004. CHOP induces death by promoting protein synthesis and oxidation in the stressed endoplasmic reticulum. Genes Dev. 18(24):3066–3077. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Marino E, Tan B, Binge L, Mackay CR, Grey ST. 2012. B-cell cross-presentation of autologous antigen precipitates diabetes. Diabetes. 61(11):2893–2905. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Martinon F 2012. Targeting endoplasmic reticulum signaling pathways in cancer. Acta Oncol. 51(7):822–830. [DOI] [PubMed] [Google Scholar]
- Martins I, Kepp O, Schlemmer F, Adjemian S, Tailler M, Shen S, Michaud M, Menger L, Gdoura A, Tajeddine N, et al. 2011. Restoration of the immunogenicity of cisplatin-induced cancer cell death by endoplasmic reticulum stress. Oncogene. 30(10):1147–1158. [DOI] [PubMed] [Google Scholar]
- Maruyama K, Watanabe H, Shiozaki H, Takayama T, Gofuku J, Yano H, Inoue M, Tamura S, Raz A, Monden M. 1995. Expression of autocrine motility factor receptor in human esophageal squamous cell carcinoma. Int J Cancer. 64(5): 316–321. [DOI] [PubMed] [Google Scholar]
- Mascia F, Schloemann DT, Cataisson C, McKinnon KM, Krymskaya L, Wolcott KM, Yuspa SH. 2016. Cell autonomous or systemic EGFR blockade alters the immune-environment in squamous cell carcinomas. Int J Cancer. 139(11):2593–2597. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Mayer TU, Braun T, Jentsch S. 1998. Role of the proteasome in membrane extraction of a short-lived ER-transmembrane protein. Embo J. 17(12):3251–3257. [DOI] [PMC free article] [PubMed] [Google Scholar]
- McCullough KD, Martindale JL, Klotz LO, Aw TY, Holbrook NJ. 2001. Gadd153 sensitizes cells to endoplasmic reticulum stress by down-regulating Bcl2 and perturbing the cellular redox state. Mol Cell Biol. 21(4):1249–1259. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Meacham GC, Patterson C, Zhang W, Younger JM, Cyr DM. 2001. The Hsc70 co-chaperone CHIP targets immature CFTR for proteasomal degradation. Nat Cell Biol. 3(1): 100–105. [DOI] [PubMed] [Google Scholar]
- Mehnert M, Sommer T, Jarosch E. 2014. Der1 promotes movement of misfolded proteins through the endoplasmic reticulum membrane. Nat Cell Biol. 16(1):77–86. [DOI] [PubMed] [Google Scholar]
- Mellman I 2013. Dendritic cells: master regulators of the immune response. Cancer Immunol Res. 1(3):145–149. [DOI] [PubMed] [Google Scholar]
- Menager J, Ebstein F, Oger R, Hulin P, Nedellec S, Duverger E, Lehmann A, Kloetzel PM, Jotereau F, Guilloux Y. 2014. Cross-presentation of synthetic long peptides by human dendritic cells: a process dependent on ERAD component p97/VCP but Not sec61 and/or Derlin-1. PLoS One. 9(2): e89897. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Meyer H 2012. p97 complexes as signal integration hubs. BMC Biol. 10:48. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Meyer H, Bug M, Bremer S. 2012. Emerging functions of the VCP/p97 AAA-ATPase in the ubiquitin system. Nat Cell Biol. 14(2):117–123. [DOI] [PubMed] [Google Scholar]
- Miao H, Jiang W, Ge L, Li B, Song B. 2010. Tetra-glutamic acid residues adjacent to Lys248 in HMG-CoA reductase are critical for the ubiquitination mediated by gp78 and UBE2G2. Acta Biochim Biophys Sin (Shanghai)). 42(5): 303–310. [DOI] [PubMed] [Google Scholar]
- Moenner M, Pluquet O, Bouchecareilh M, Chevet E. 2007. Integrated endoplasmic reticulum stress responses in cancer. Cancer Res. 67(22):10631–10634. [DOI] [PubMed] [Google Scholar]
- Mohamed E, Cao Y, Rodriguez PC. 2017. Endoplasmic reticulum stress regulates tumor growth and anti-tumor immunity: a promising opportunity for cancer immunotherapy. Cancer Immunol Immunother. 66(8):1069–1078. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Molinari M, Galli C, Piccaluga V, Pieren M, Paganetti P. 2002. Sequential assistance of molecular chaperones and transient formation of covalent complexes during protein degradation from the ER. J Cell Biol. 158(2):247–257. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Moon HW, Han HG, Jeon YJ. 2018. Protein quality control in the endoplasmic reticulum and cancer. Int J Mol Sci. 19(10) [DOI] [PMC free article] [PubMed] [Google Scholar]
- Moore P, He K, Tsai B. 2013. Establishment of an in vitro transport assay that reveals mechanistic differences in cytosolic events controlling cholera toxin and T-cell receptor α retro-translocation. PLoS One. 8(10):e75801. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Morito D, Hirao K, Oda Y, Hosokawa N, Tokunaga F, Cyr DM, Tanaka K, Iwai K, Nagata K. 2008. Gp78 cooperates with RMA1 in endoplasmic reticulum-associated degradation of CFTRDeltaF508. Mol Biol Cell. 19(4):1328–1336. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Moserova I, Truxova I, Garg AD, Tomala J, Agostinis P, Cartron PF, Vosahlikova S, Kovar M, Spisek R, Fucikova J. 2017. Caspase-2 and oxidative stress underlie the immunogenic potential of high hydrostatic pressure-induced cancer cell death. Oncoimmunology. 6(1): e1258505. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Mueller B, Klemm EJ, Spooner E, Claessen JH, Ploegh HL. 2008. SEL1L nucleates a protein complex required for dislocation of misfolded glycoproteins. Proc Natl Acad Sci USA. 105(34):12325–12330. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Mueller B, Lilley BN, Ploegh HL. 2006. SEL1L, the homologue of yeast Hrd3p, is involved in protein dislocation from the mammalian ER. J Cell Biol. 175(2):261–270. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Murata S, Minami Y, Minami M, Chiba T, Tanaka K. 2001. CHIP is a chaperone-dependent E3 ligase that ubiquitylates unfolded protein. EMBO Rep. 2(12):1133–1138. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Nadav E, Shmueli A, Barr H, Gonen H, Ciechanover A, Reiss Y. 2003. A novel mammalian endoplasmic reticulum ubiquitin ligase homologous to the yeast Hrd1. Biochem Biophys Res Commun. 303(1):91–97. [DOI] [PubMed] [Google Scholar]
- Nakagawa H, Umemura A, Taniguchi K, Font-Burgada J, Dhar D, Ogata H, Zhong Z, Valasek MA, Seki E, Hidalgo J, et al. 2014. ER stress cooperates with hypernutrition to trigger TNF-dependent spontaneous HCC development. Cancer Cell. 26(3):331–343. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Nakamori S, Watanabe H, Kameyama M, Imaoka S, Furukawa H, Ishikawa O, Sasaki Y, Kabuto T, Raz A. 1994. Expression of autocrine motility factor receptor in colorectal cancer as a predictor for disease recurrence. Cancer. 74(7): 1855–1862. [DOI] [PubMed] [Google Scholar]
- Nan J, Xing YF, Hu B, Tang JX, Dong HM, He YM, Ruan DY, Ye QJ, Cai JR, Ma XK, et al. 2018. Endoplasmic reticulum stress induced LOX-1+ CD15+ polymorphonuclear myeloid-derived suppressor cells in hepatocellular carcinoma. Immunology. 154(1):144–155. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Neefjes J, Jongsma ML, Paul P, Bakke O. 2011. Towards a systems understanding of MHC class I and MHC class II antigen presentation. Nat Rev Immunol. 11(12):823–836. [DOI] [PubMed] [Google Scholar]
- Niederreiter L, Fritz TM, Adolph TE, Krismer AM, Offner FA, Tschurtschenthaler M, Flak MB, Hosomi S, Tomczak MF, Kaneider NC, et al. 2013. ER stress transcription factor Xbp1 suppresses intestinal tumorigenesis and directs intestinal stem cells. J Exp Med. 210(10):2041–2056. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Niino D, Komohara Y, Murayama T, Aoki R, Kimura Y, Hashikawa K, Kiyasu J, Takeuchi M, Suefuji N, Sugita Y, et al. 2010. Ratio of M2 macrophage expression is closely associated with poor prognosis for Angioimmunoblastic T-cell lymphoma (AITL). Pathol Int. 60(4):278–283. [DOI] [PubMed] [Google Scholar]
- Nillegoda NB, Theodoraki MA, Mandal AK, Mayo KJ, Ren HY, Sultana R, Wu K, Johnson J, Cyr DM, Caplan AJ. 2010. Ubr1 and Ubr2 function in a quality control pathway for degradation of unfolded cytosolic proteins. Mol Biol Cell. 21(13):2102–2116. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Ninagawa S, Okada T, Sumitomo Y, Kamiya Y, Kato K, Horimoto S, Ishikawa T, Takeda S, Sakuma T, Yamamoto T, et al. 2014. EDEM2 initiates mammalian glycoprotein ERAD by catalyzing the first mannose trimming step. J Cell Biol. 206(3):347–356. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Nouri-Shirazi M, Banchereau J, Bell D, Burkeholder S, Kraus ET, Davoust J, Palucka KA. 2000. Dendritic cells capture killed tumor cells and present their antigens to elicit tumor-specific immune responses. J Immunol. 165(7): 3797–3803. [DOI] [PubMed] [Google Scholar]
- Nowis D, McConnell E, Wojcik C. 2006. Destabilization of the VCP-Ufd1-Npl4 complex is associated with decreased levels of ERAD substrates. Exp Cell Res. 312(15):2921–2932. [DOI] [PubMed] [Google Scholar]
- Nutt SL, Hodgkin PD, Tarlinton DM, Corcoran LM. 2015. The generation of antibody-secreting plasma cells. Nat Rev Immunol. 15(3):160–171. [DOI] [PubMed] [Google Scholar]
- Oakes SA, Papa FR. 2015. The role of endoplasmic reticulum stress in human pathology. Annu Rev Pathol. 10:173–194. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Obeid M 2008. ERP57 membrane translocation dictates the immunogenicity of tumor cell death by controlling the membrane translocation of calreticulin. J Immunol. 181(4): 2533–2543. [DOI] [PubMed] [Google Scholar]
- Obeid M, Tesniere A, Ghiringhelli F, Fimia GM, Apetoh L, Perfettini JL, Castedo M, Mignot G, Panaretakis T, Casares N, et al. 2007. Calreticulin exposure dictates the immunogenicity of cancer cell death. Nat Med. 13(1):54–61. [DOI] [PubMed] [Google Scholar]
- Obeng EA, Carlson LM, Gutman DM, Harrington WJ Jr., Lee KP, Boise LH. 2006. Proteasome inhibitors induce a terminal unfolded protein response in multiple myeloma cells. Blood. 107(12):4907–4916. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Okazaki T, Honjo T. 2007. PD-1 and PD-1 ligands: from discovery to clinical application. Int Immunol. 19(7):813–824. [DOI] [PubMed] [Google Scholar]
- Okuda-Shimizu Y, Hendershot LM. 2007. Characterization of an ERAD pathway for nonglycosylated BiP substrates, which require Herp. Mol Cell. 28(4):544–554. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Orlandi R, Cattaneo M, Troglio F, Casalini P, Ronchini C, Menard S, Biunno I. 2002. SEL1L expression decreases breast tumor cell aggressiveness in vivo and in vitro. Cancer Res. 62(2):567–574. [PubMed] [Google Scholar]
- Ostankovitch M, Robila V, Engelhard VH. 2005. Regulated folding of tyrosinase in the endoplasmic reticulum demonstrates that misfolded full-length proteins are efficient substrates for class I processing and presentation. J Immunol. 174(5):2544–2551. [DOI] [PubMed] [Google Scholar]
- Otto T, Birchmeier W, Schmidt U, Hinke A, Schipper J, Rubben H, Raz A. 1994. Inverse relation of E-cadherin and autocrine motility factor receptor expression as a prognostic factor in patients with bladder carcinomas. Cancer Res. 54(12):3120–3123. [PubMed] [Google Scholar]
- Palmer AL, Dolan BP. 2013. MHC class I antigen presentation of DRiP-derived peptides from a model antigen is not dependent on the AAA ATPase p97. PLoS One. 8(7): e67796. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Pardoll DM. 2012. The blockade of immune checkpoints in cancer immunotherapy. Nat Rev Cancer. 12(4):252–264. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Parekh VV, Pabbisetty SK, Wu L, Sebzda E, Martinez J, Zhang J, Van Kaer L. 2017. Autophagy-related protein Vps34 controls the homeostasis and function of antigen cross-presenting CD8α+ dendritic cells. Proc Natl Acad Sci USA. 114(31):E6371–E6380. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Pellicciotta I, Cortez-Gonzalez X, Sasik R, Reiter Y, Hardiman G, Langlade-Demoyen P, Zanetti M. 2008. Presentation of telomerase reverse transcriptase, a self-tumor antigen, is down-regulated by histone deacetylase inhibition. Cancer Res. 68(19):8085–8093. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Pennock GK, Chow LQ. 2015. The evolving role of immune checkpoint inhibitors in cancer treatment. Oncologist. 20(7):812–822. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Plemper RK, Bohmler S, Bordallo J, Sommer T, Wolf DH. 1997. Mutant analysis links the translocon and BiP to retrograde protein transport for ER degradation. Nature. 388(6645):891–895. [DOI] [PubMed] [Google Scholar]
- Qiu XB, Goldberg AL. 2002. Nrdp1/FLRF is a ubiquitin ligase promoting ubiquitination and degradation of the epidermal growth factor receptor family member, ErbB3. Proc Natl Acad Sci USA. 99(23):14843–14848. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Quail DF, Joyce JA. 2013. Microenvironmental regulation of tumor progression and metastasis. Nat Med. 19(11): 1423–1437. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Rabinovich E, Kerem A, Frohlich KU, Diamant N, Bar-Nun S. 2002. AAA-ATPase p97/Cdc48p, a cytosolic chaperone required for endoplasmic reticulum-associated protein degradation. Mol Cell Biol. 22(2):626–634. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Ramirez MC, Sigal LJ. 2002. Macrophages and dendritic cells use the cytosolic pathway to rapidly cross-present antigen from live, vaccinia-infected cells. J Immunol. 169(12): 6733–6742. [DOI] [PubMed] [Google Scholar]
- Ravid T, Kreft SG, Hochstrasser M. 2006. Membrane and soluble substrates of the Doa10 ubiquitin ligase are degraded by distinct pathways. Embo J. 25(3):533–543. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Ribas A, Wolchok JD. 2018. Cancer immunotherapy using checkpoint blockade. Cancer Immunology, Immunotherapy. 359(6382):1350–1355. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Richly H, Rape M, Braun S, Rumpf S, Hoege C, Jentsch S. 2005. A series of ubiquitin binding factors connects CDC48/p97 to substrate multiubiquitylation and proteasomal targeting. Cell. 120(1):73–84. [DOI] [PubMed] [Google Scholar]
- Rivett AJ. 1993. Proteasomes: multicatalytic proteinase complexes. Biochem J. 291 (1):1–10. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Roche PA, Furuta K. 2015. The ins and outs of MHC class II-mediated antigen processing and presentation. Nat Rev Immunol. 15(4):203–216. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Rodriguez A, Regnault A, Kleijmeer M, Ricciardi-Castagnoli P, Amigorena S. 1999. Selective transport of internalized antigens to the cytosol for MHC class I presentation in dendritic cells. Nat Cell Biol. 1(6):362–368. [DOI] [PubMed] [Google Scholar]
- Roelse J, Gromme M, Momburg F, Hammerling G, Neefjes J. 1994. Trimming of TAP-translocated peptides in the endoplasmic reticulum and in the cytosol during recycling. J Exp Med. 180(5):1591–1597. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Rubio-Patino C, Bossowski JP, De Donatis GM, Mondragon L, Villa E, Aira LE, Chiche J, Mhaidly R, Lebeaupin C, Marchetti S, et al. 2018. Low-Protein Diet Induces IRE1α-Dependent Anticancer Immunosurveillance. Cell Metab. 27(4):828–842 e827. [DOI] [PubMed] [Google Scholar]
- Ruggiano A, Foresti O, Carvalho P. 2014. Quality control: ER-associated degradation: protein quality control and beyond. J Cell Biol. 204(6):869–879. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Russo V, Tanzarella S, Dalerba P, Rigatti D, Rovere P, Villa A, Bordignon C, Traversari C. 2000. Dendritic cells acquire the MAGE-3 human tumor antigen from apoptotic cells and induce a class I-restricted T cell response. Proc Natl Acad Sci USA. 97(5):2185–2190. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Sadasivan B, Lehner PJ, Ortmann B, Spies T, Cresswell P. 1996. Roles for calreticulin and a novel glycoprotein, tapasin, in the interaction of MHC class I molecules with TAP. Immunity. 5(2):103–114. [DOI] [PubMed] [Google Scholar]
- Sagar V, Vatapalli R, Lysy B, Pamarthy S, Anker JF, Rodriguez Y, Han H, Unno K, Stadler WM, Catalona WJ, et al. 2019. EPHB4 inhibition activates ER stress to promote immunogenic cell death of prostate cancer cells. Cell Death Dis. 10(11):801. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Saric T, Chang SC, Hattori A, York IA, Markant S, Rock KL, Tsujimoto M, Goldberg AL. 2002. An IFN-gamma-induced aminopeptidase in the ER, ERAP1, trims precursors to MHC class I-presented peptides. Nat Immunol. 3(12):1169–1176. [DOI] [PubMed] [Google Scholar]
- Sauter B, Albert ML, Francisco L, Larsson M, Somersan S, Bhardwaj N. 2000. Consequences of Cell death: exposure to necrotic tumor cells, but not primary tissue cells or apoptotic cells, induces the maturation of immunostimulatory dendritic cells. J Exp Med. 191(3):423–434. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Saveanu L, Carroll O, Lindo V, Del Val M, Lopez D, Lepelletier Y, Greer F, Schomburg L, Fruci D, Niedermann G, et al. 2005. Concerted peptide trimming by human ERAP1 and ERAP2 aminopeptidase complexes in the endoplasmic reticulum. Nat Immunol. 6(7):689–697. [DOI] [PubMed] [Google Scholar]
- Schaer DA, Beckmann RP, Dempsey JA, Huber L, Forest A, Amaladas N, Li Y, Wang YC, Rasmussen ER, Chin D, et al. 2018. The CDK4/6 inhibitor abemaciclib induces a T cell inflamed tumor microenvironment and enhances the efficacy of PD-L1 checkpoint blockade. Cell Rep. 22(11): 2978–2994. [DOI] [PubMed] [Google Scholar]
- Schiavoni G, Sistigu A, Valentini M, Mattei F, Sestili P, Spadaro F, Sanchez M, Lorenzi S, D’Urso MT, Belardelli F, et al. 2011. Cyclophosphamide synergizes with type I interferons through systemic dendritic cell reactivation and induction of immunogenic tumor apoptosis. Cancer Res. 71(3):768–778. [DOI] [PubMed] [Google Scholar]
- Schmid D, Munz C. 2007. Innate and adaptive immunity through autophagy. Immunity. 27(1):11–21. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Schoebel S, Mi W, Stein A, Ovchinnikov S, Pavlovicz R, DiMaio F, Baker D, Chambers MG, Su H, Li D, et al. 2017. Cryo-EM structure of the protein-conducting ERAD channel Hrd1 in complex with Hrd3. Nature. 548(7667): 352–355. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Schulze A, Standera S, Buerger E, Kikkert M, van Voorden S, Wiertz E, Koning F, Kloetzel PM, Seeger M. 2005. The ubiquitin-domain protein HERP forms a complex with components of the endoplasmic reticulum associated degradation pathway. J Mol Biol. 354(5):1021–1027. [DOI] [PubMed] [Google Scholar]
- Schwab SR, Shugart JA, Horng T, Malarkannan S, Shastri N. 2004. Unanticipated antigens: translation initiation at CUG with leucine. PLoS Biol. 2(11):e366. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Serwold T, Gonzalez F, Kim J, Jacob R, Shastri N. 2002. ERAAP customizes peptides for MHC class I molecules in the endoplasmic reticulum. Nature. 419(6906):480–483. [DOI] [PubMed] [Google Scholar]
- Sha H, Sun S, Francisco AB, Ehrhardt N, Xue Z, Liu L, Lawrence P, Mattijssen F, Guber RD, Panhwar MS, et al. 2014. The ER-associated degradation adaptor protein Sel1L regulates LPL secretion and lipid metabolism. Cell Metab. 20(3):458–470. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Shaffer AL, Shapiro-Shelef M, Iwakoshi NN, Lee AH, Qian SB, Zhao H, Yu X, Yang L, Tan BK, Rosenwald A, et al. 2004. XBP1, downstream of Blimp-1, expands the secretory apparatus and other organelles, and increases protein synthesis in plasma cell differentiation. Immunity. 21(1):81–93. [DOI] [PubMed] [Google Scholar]
- Shi G, Somlo DRM, Kim GH, Prescianotto-Baschong, Sun S, Beuret N, Long Q, Rutishauser J, Arvan P, Spiess M, et al. 2017. ER-associated degradation is required for vasopressin prohormone processing and systemic water homeostasis. J Clin Invest. 127(10):3897–3912. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Shi K, Wang D, Cao X, Ge Y. 2013. Endoplasmic reticulum stress signaling is involved in mitomycin C (MMC)-induced apoptosis in human fibroblasts via PERK pathway. PLoS One. 8(3):e59330. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Shibata M, Kanda M, Tanaka H, Umeda S, Miwa T, Shimizu D, Hayashi M, Inaishi T, Miyajima N, Adachi Y, et al. 2017. Overexpression of Derlin 3 is associated with malignant phenotype of breast cancer cells. Oncol Rep. 38(3): 1760–1766. [DOI] [PubMed] [Google Scholar]
- Shih TA, Roederer M, Nussenzweig MC. 2002. Role of antigen receptor affinity in T cell-independent antibody responses in vivo. Nat Immunol. 3(4):399–406. [DOI] [PubMed] [Google Scholar]
- Silletti S, Raz A. 1996. Regulation of autocrine motility factor receptor expression in tumor cell locomotion and metastasis. Curr Top Microbiol Immunol. 213 (Pt 2):137–169. [DOI] [PubMed] [Google Scholar]
- Skowronek MH, Hendershot LM, Haas IG. 1998. The variable domain of nonassembled Ig light chains determines both their half-life and binding to the chaperone BiP. Proc Natl Acad Sci USA. 95(4):1574–1578. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Skrott Z, Mistrik M, Andersen KK, Friis S, Majera D, Gursky J, Ozdian T, Bartkova J, Turi Z, Moudry P, et al. 2017. Alcohol-abuse drug disulfiram targets cancer via p97 segregase adaptor NPL4. Nature. 552(7684):194–199. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Sommer T, Jentsch S. 1993. A protein translocation defect linked to ubiquitin conjugation at the endoplasmic reticulum. Nature. 365(6442):176–179. [DOI] [PubMed] [Google Scholar]
- Song BL, Javitt NB, DeBose-Boyd RA. 2005. Insig-mediated degradation of HMG CoA reductase stimulated by lanosterol, an intermediate in the synthesis of cholesterol. Cell Metab. 1(3):179–189. [DOI] [PubMed] [Google Scholar]
- Sowa ME, Bennett EJ, Gygi SP, Harper JW. 2009. Defining the human deubiquitinating enzyme interaction landscape. Cell. 138(2):389–403. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Spisek R, Charalambous A, Mazumder A, Vesole DH, Jagannath S, Dhodapkar MV. 2007. Bortezomib enhances dendritic cell (DC)-mediated induction of immunity to human myeloma via exposure of cell surface heat shock protein 90 on dying tumor cells: therapeutic implications. Blood. 109(11):4839–4845. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Srivastava RM, Lee SC, Andrade Filho PA, Lord CA, Jie HB, Davidson HC, Lopez-Albaitero A, Gibson SP, Gooding WE, Ferrone S, et al. 2013. Cetuximab-activated natural killer and dendritic cells collaborate to trigger tumor antigen-specific T-cell immunity in head and neck cancer patients. Clin Cancer Res. 19(7):1858–1872. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Stagg HR, Thomas M, van den Boomen D, Wiertz EJ, Drabkin HA, Gemmill RM, Lehner PJ. 2009. The TRC8 E3 ligase ubiquitinates MHC class I molecules before dislocation from the ER. J Cell Biol. 186(5):685–692. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Starr TK, Jameson SC, Hogquist KA. 2003. Positive and negative selection of T cells. Annu Rev Immunol. 21:139–176. [DOI] [PubMed] [Google Scholar]
- Stewart EL, Tan SZ, Liu G, Tsao M-S. 2015. Known and putative mechanisms of resistance to EGFR targeted therapies in NSCLC patients with EGFR mutations-a review. Transl Lung Cancer Res. 4(1):67–81. eng. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Strasser A, Puthalakath H. 2008. Fold up or perish: unfolded protein response and chemotherapy. Cell Death Differ. 15(2):223–225. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Sugimoto T, Ninagawa S, Yamano S, Ishikawa T, Okada T, Takeda S, Mori K. 2017. SEL1L-dependent substrates require Derlin2/3 and Herp1/2 for endoplasmic reticulum-associated degradation. Cell Struct Funct. 42(2):81–94. [DOI] [PubMed] [Google Scholar]
- Sun S, Shi G, Han X, Francisco AB, Ji Y, Mendonca N, Liu X, Locasale JW, Simpson KW, Duhamel GE, et al. 2014. Sel1L is indispensable for mammalian endoplasmic reticulum-associated degradation, endoplasmic reticulum homeostasis, and survival. Proc Natl Acad Sci USA. 111(5):E582–591. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Sun S, Shi G, Sha H, Ji Y, Han X, Shu X, Ma H, Inoue T, Gao B, Kim H, et al. 2015. IRE1α is an endogenous substrate of endoplasmic-reticulum-associated degradation. Nat Cell Biol. 17(12):1546–1555. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Sun YW, Chen YF, Li J, Huo YM, Liu DJ, Hua R, Zhang JF, Liu W, Yang JY, Fu XL, et al. 2014. A novel long non-coding RNA ENST00000480739 suppresses tumour cell invasion by regulating OS-9 and HIF-1α in pancreatic ductal adenocarcinoma. Br J Cancer. 111(11):2131–2141. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Sundaram A, Appathurai S, Plumb R, Mariappan M. 2018. Dynamic changes in complexes of IRE1α, PERK, and ATF6α during endoplasmic reticulum stress. Mol Biol Cell. 29(11): 1376–1388. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Suzuki M, Otsuka T, Ohsaki Y, Cheng J, Taniguchi T, Hashimoto H, Taniguchi H, Fujimoto T. 2012. Derlin-1 and UBXD8 are engaged in dislocation and degradation of lipidated ApoB-100 at lipid droplets. Mol Biol Cell. 23(5): 800–810. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Szathmary R, Bielmann R, Nita-Lazar M, Burda P, Jakob CA. 2005. Yos9 protein is essential for degradation of misfolded glycoproteins and may function as lectin in ERAD. Mol Cell. 19(6):765–775. [DOI] [PubMed] [Google Scholar]
- Takahama Y 2006. Journey through the thymus: stromal guides for T-cell development and selection. Nat Rev Immunol. 6(2):127–135. [DOI] [PubMed] [Google Scholar]
- Tan X, He X, Fan Z. 2019. Upregulation of HRD1 promotes cell migration and invasion in colon cancer. Mol Cell Biochem. 454(1–2):1–9. [DOI] [PubMed] [Google Scholar]
- Tan X, He X, Jiang Z, Wang X, Ma L, Liu L, Wang X, Fan Z, Su D. 2015. Derlin-1 is overexpressed in human colon cancer and promotes cancer cell proliferation. Mol Cell Biochem. 408(1–2):205–213. [DOI] [PubMed] [Google Scholar]
- Tang HY, Huang CH, Zhuang YH, Christianson JC, Chen X. 2014. EDEM2 and OS-9 are required for ER-associated degradation of non-glycosylated sonic hedgehog. PLoS One. 9(6):e92164. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Tang Z-H, Lu J-J. 2018. Osimertinib resistance in non-small cell lung cancer: mechanisms and therapeutic strategies. Cancer Lett. 420:242–246. [DOI] [PubMed] [Google Scholar]
- Tax G, Lia A, Santino A, Roversi P. 2019. Modulation of ERQC and ERAD: a broad-spectrum spanner in the works of cancer cells? J Oncol. 2019:8384913. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Terashima Y, Toda E, Itakura M, Otsuji M, Yoshinaga S, Okumura K, Shand FHW, Komohara Y, Takeda M, Kokubo K, et al. 2020. Targeting FROUNT with disulfiram suppresses macrophage accumulation and its tumor-promoting properties. Nat Commun. 11(1):609. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Tesniere A, Schlemmer F, Boige V, Kepp O, Martins I, Ghiringhelli F, Aymeric L, Michaud M, Apetoh L, Barault L, et al. 2010. Immunogenic death of colon cancer cells treated with oxaliplatin. Oncogene. 29(4):482–491. [DOI] [PubMed] [Google Scholar]
- Thevenot PT, Sierra RA, Raber PL, Al-Khami AA, Trillo-Tinoco J, Zarreii P, Ochoa AC, Cui Y, Del Valle L, Rodriguez PC. 2014. The stress-response sensor chop regulates the function and accumulation of myeloid-derived suppressor cells in tumors. Immunity. 41(3):389–401. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Tomati V, Sondo E, Armirotti A, Caci E, Pesce E, Marini M, Gianotti A, Jeon YJ, Cilli M, Pistorio A, et al. 2015. Genetic inhibition of the ubiquitin ligase Rnf5 attenuates phenotypes associated to F508del cystic fibrosis mutation. Sci Rep. 5:12138. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Tsai YC, Fishman PS, Thakor NV, Oyler GA. 2003. Parkin facilitates the elimination of expanded polyglutamine proteins and leads to preservation of proteasome function. J Biol Chem. 278(24):22044–22055. [DOI] [PubMed] [Google Scholar]
- Tschopp J, Schroder K. 2010. NLRP3 inflammasome activation: The convergence of multiple signalling pathways on ROS production?. Nat Rev Immunol. 10(3):210–215. [DOI] [PubMed] [Google Scholar]
- Tsuchiya Y, Morita T, Kim M, Iemura S, Natsume T, Yamamoto M, Kobayashi A. 2011. Dual regulation of the transcriptional activity of Nrf1 by β-TrCP- and Hrd1-dependent degradation mechanisms. Mol Cell Biol. 31(22):4500–4512. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Tyler RE, Pearce MM, Shaler TA, Olzmann JA, Greenblatt EJ, Kopito RR. 2012. Unassembled CD147 is an endogenous endoplasmic reticulum-associated degradation substrate. Mol Biol Cell. 23(24):4668–4678. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Uchiyama K, Jokitalo E, Kano F, Murata M, Zhang X, Canas B, Newman R, Rabouille C, Pappin D, Freemont P, et al. 2002. VCIP135, a novel essential factor for p97/p47-mediated membrane fusion, is required for Golgi and ER assembly in vivo. J Cell Biol. 159(5):855–866. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Ushioda R, Hoseki J, Araki K, Jansen G, Thomas DY, Nagata K. 2008. ERdj5 is required as a disulfide reductase for degradation of misfolded proteins in the ER. Science. 321(5888): 569–572. [DOI] [PubMed] [Google Scholar]
- van de Weijer ML, Bassik MC, Luteijn RD, Voorburg CM, Lohuis MA, Kremmer E, Hoeben RC, LeProust EM, Chen S, Hoelen H, et al. 2014. A high-coverage shRNA screen identifies TMEM129 as an E3 ligase involved in ER-associated protein degradation. Nat Commun. 5:3832. [DOI] [PMC free article] [PubMed] [Google Scholar]
- van den Boomen DJ, Timms RT, Grice GL, Stagg HR, Skodt K, Dougan G, Nathan JA, Lehner PJ. 2014. TMEM129 is a Derlin-1 associated ERAD E3 ligase essential for virus-induced degradation of MHC-I. Proc Natl Acad Sci USA. 111(31):11425–11430. [DOI] [PMC free article] [PubMed] [Google Scholar]
- van den Boorn JG, Picavet DI, van Swieten PF, van Veen HA, Konijnenberg D, van Veelen PA, van Capel T, Jong EC, Reits EA, Drijfhout JW, et al. 2011. Skin-depigmenting agent monobenzone induces potent T-cell autoimmunity toward pigmented cells by tyrosinase haptenation and melanosome autophagy. J Invest Dermatol. 131(6): 1240–1251. [DOI] [PubMed] [Google Scholar]
- van Vliet AR, Martin S, Garg AD, Agostinis P. 2015. The PERKs of damage-associated molecular patterns mediating cancer immunogenicity: from sensor to the plasma membrane and beyond. Semin Cancer Biol. 33:74–85. [DOI] [PubMed] [Google Scholar]
- Vanacker H, Vetters J, Moudombi L, Caux C, Janssens S, Michallet MC. 2017. Emerging role of the unfolded protein response in tumor immunosurveillance. Trends Cancer. 3(7):491–505. [DOI] [PubMed] [Google Scholar]
- Vashist S, Ng DT. 2004. Misfolded proteins are sorted by a sequential checkpoint mechanism of ER quality control. J Cell Biol. 165(1):41–52. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Vaupel P, Multhoff G. 2017. Accomplices of the hypoxic tumor microenvironment compromising antitumor immunity: adenosine, lactate, acidosis, vascular endothelial growth factor, potassium ions, and phosphatidylserine. Front Immunol. 8:1887. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Verma R, Chen S, Feldman R, Schieltz D, Yates J, Dohmen J, Deshaies RJ. 2000. Proteasomal proteomics: identification of nucleotide-sensitive proteasome-interacting proteins by mass spectrometric analysis of affinity-purified proteasomes. Mol Biol Cell. 11(10):3425–3439. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Vitiello M, Tuccoli A, D’Aurizio R, Sarti S, Giannecchini L, Lubrano S, Marranci A, Evangelista M, Peppicelli S, Ippolito C, et al. 2017. Context-dependent miR-204 and miR-211 affect the biological properties of amelanotic and melanotic melanoma cells. Oncotarget. 8(15):25395–25417. [DOI] [PMC free article] [PubMed] [Google Scholar]
- von Boehmer H, Melchers F. 2010. Checkpoints in lymphocyte development and autoimmune disease. Nat Immunol. 11(1):14–20. [DOI] [PubMed] [Google Scholar]
- Vyas JM, Van der Veen AG, Ploegh HL. 2008. The known unknowns of antigen processing and presentation. Nat Rev Immunol. 8(8):607–618. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Wahlman J, DeMartino GN, Skach WR, Bulleid NJ, Brodsky JL, Johnson AE. 2007. Real-time fluorescence detection of ERAD substrate retrotranslocation in a mammalian in vitro system. Cell. 129(5):943–955. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Walter P, Ron D. 2011. The unfolded protein response: from stress pathway to homeostatic regulation. Science. 334(6059):1081–1086. [DOI] [PubMed] [Google Scholar]
- Wang C, Zhang F, Cao Y, Zhang M, Wang A, Xu M, Su M, Zhang M, Zhuge Y. 2016. Etoposide Induces Apoptosis in Activated Human Hepatic Stellate Cells via ER Stress. Sci Rep. 6:34330. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Wang H, Pezeshki AM, Yu X, Guo C, Subjeck JR, Wang XY. 2014. The endoplasmic reticulum chaperone GRP170: from immunobiology to cancer therapeutics. Front Oncol. 4:377. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Wang H, Yu X, Guo C, Zuo D, Fisher PB, Subjeck JR, Wang XY. 2013. Enhanced endoplasmic reticulum entry of tumor antigen is crucial for cross-presentation induced by dendritic cell-targeted vaccination. J Immunol. 191(12): 6010–6021. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Wang Q, Li L, Ye Y. 2006. Regulation of retrotranslocation by p97-associated deubiquitinating enzyme ataxin-3. J Cell Biol. 174(7):963–971. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Wang Q, Liu Y, Soetandyo N, Baek K, Hegde R, Ye Y. 2011. A ubiquitin ligase-associated chaperone holdase maintains polypeptides in soluble states for proteasome degradation. Mol Cell. 42(6):758–770. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Wang XY, Sun X, Chen X, Facciponte J, Repasky EA, Kane J, Subjeck JR. 2010. Superior antitumor response induced by large stress protein chaperoned protein antigen compared with peptide antigen. J Immunol. 184(11):6309–6319. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Watts C 2004. The exogenous pathway for antigen presentation on major histocompatibility complex class II and CD1 molecules. Nat Immunol. 5(7):685–692. [DOI] [PubMed] [Google Scholar]
- Weber JS, Yang JC, Atkins MB, Disis ML. 2015. Toxicities of immunotherapy for the practitioner. J Clin Oncol. 33(18): 2092–2099. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Wiertz EJ, Jones TR, Sun L, Bogyo M, Geuze HJ, Ploegh HL. 1996. The human cytomegalovirus US11 gene product dislocates MHC class I heavy chains from the endoplasmic reticulum to the cytosol. Cell. 84(5):769–779. [DOI] [PubMed] [Google Scholar]
- Williams KJ. 2008. Molecular processes that handle––and mishandle––dietary lipids. J Clin Invest. 118(10):3247–3259. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Wing K, Onishi Y, Prieto-Martin P, Yamaguchi T, Miyara M, Fehervari Z, Nomura T, Sakaguchi S. 2008. CTLA-4 control over Foxp3+ regulatory T cell function. Science. 322(5899):271–275. eng. [DOI] [PubMed] [Google Scholar]
- Wojcik C, Rowicka M, Kudlicki A, Nowis D, McConnell E, Kujawa M, DeMartino GN. 2006. Valosin-containing protein (p97) is a regulator of endoplasmic reticulum stress and of the degradation of N-end rule and ubiquitin-fusion degradation pathway substrates in mammalian cells. Mol Biol Cell. 17(11):4606–4618. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Wolfers J, Lozier A, Raposo G, Regnault A, Thery C, Masurier C, Flament C, Pouzieux S, Faure F, Tursz T, et al. 2001. Tumor-derived exosomes are a source of shared tumor rejection antigens for CTL cross-priming. Nat Med. 7(3): 297–303. [DOI] [PubMed] [Google Scholar]
- Wu J, Waxman DJ. 2018. Immunogenic chemotherapy: dose and schedule dependence and combination with immunotherapy. Cancer Lett. 419:210–221. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Xing Y, Cao R, Hu HM. 2016. TLR and NLRP3 inflammasome-dependent innate immune responses to tumor-derived autophagosomes (DRibbles). Cell Death Dis. 7(8):e2322. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Xu C, Ng DT. 2015. Glycosylation-directed quality control of protein folding. Nat Rev Mol Cell Biol. 16(12):742–752. [DOI] [PubMed] [Google Scholar]
- Xu Q, Chen C, Lin A, Xie Y. 2017. Endoplasmic reticulum stress-mediated membrane expression of CRT/ERp57 induces immunogenic apoptosis in drug-resistant endometrial cancer cells. Oncotarget. 8(35):58754–58764. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Xu Y, Cai M, Yang Y, Huang L, Ye Y. 2012. SGTA recognizes a noncanonical ubiquitin-like domain in the Bag6-Ubl4A-Trc35 complex to promote endoplasmic reticulum-associated degradation. Cell Rep. 2(6):1633–1644. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Xu Y, Melo-Cardenas J, Zhang Y, Gau I, Wei J, Montauti E, Zhang Y, Gao B, Jin H, Sun Z, et al. 2019. The E3 ligase Hrd1 stabilizes Tregs by antagonizing inflammatory cytokine-induced ER stress response. JCI Insight. 4(5):e121887. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Xu Y, Zhao F, Qiu Q, Chen K, Wei J, Kong Q, Gao B, Melo-Cardenas J, Zhang B, Zhang J, et al. 2016. The ER membrane-anchored ubiquitin ligase Hrd1 is a positive regulator of T-cell immunity. Nat Commun. 7:12073. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Yadav RK, Chae SW, Kim HR, Chae HJ. 2014. Endoplasmic reticulum stress and cancer. J Cancer Prev. 19(2):75–88. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Yamaoka T, Ohba M, Ohmori T. 2017. Molecular-targeted therapies for epidermal growth factor receptor and its resistance mechanisms. IJMS. 18(11):2420. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Yamasaki S, Yagishita N, Sasaki T, Nakazawa M, Kato Y, Yamadera T, Bae E, Toriyama S, Ikeda R, Zhang L, et al. 2007. Cytoplasmic destruction of p53 by the endoplasmic reticulum-resident ubiquitin ligase ‘Synoviolin’. Embo J. 26(1):113–122. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Yan D, Wang HW, Bowman RL, Joyce JA. 2016. STAT3 and STAT6 Signaling Pathways Synergize to Promote Cathepsin Secretion from Macrophages via IRE1α Activation. Cell Rep. 16(11):2914–2927. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Yang H, Qiu Q, Gao B, Kong S, Lin Z, Fang D. 2014. Hrd1-mediated BLIMP-1 ubiquitination promotes dendritic cell MHCII expression for CD4 T cell priming during inflammation. J Exp Med. 211(12):2467–2479. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Yang M, Omura S, Bonifacino JS, Weissman AM. 1998. Novel aspects of degradation of T cell receptor subunits from the endoplasmic reticulum (ER) in T cells: importance of oligosaccharide processing, ubiquitination, and proteasome-dependent removal from ER membranes. J Exp Med. 187(6):835–846. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Yarapureddy S, Abril J, Foote J, Kumar S, Asad O, Sharath V, Faraj J, Daniel D, Dickman P, White-Collins A, et al. 2019. ATF6α activation enhances survival against chemotherapy and serves as a prognostic indicator in osteosarcoma. Neoplasia. 21(6):516–532. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Ye Y, Meyer HH, Rapoport TA. 2001. The AAA ATPase Cdc48: p97 and its partners transport proteins from the ER into the cytosol. Nature. 414(6864):652–656. [DOI] [PubMed] [Google Scholar]
- Ye Y, Meyer HH, Rapoport TA. 2003. Function of the p97-Ufd1-Npl4 complex in retrotranslocation from the ER to the cytosol: dual recognition of nonubiquitinated polypeptide segments and polyubiquitin chains. J Cell Biol. 162(1): 71–84. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Ye Y, Shibata Y, Yun C, Ron D, Rapoport TA. 2004. A membrane protein complex mediates retro-translocation from the ER lumen into the cytosol. Nature. 429(6994):841–847. [DOI] [PubMed] [Google Scholar]
- Yokosuka T, Takamatsu M, Kobayashi-Imanishi W, Hashimoto-Tane A, Azuma M, Saito T. 2012. Programmed cell death 1 forms negative costimulatory microclusters that directly inhibit T cell receptor signaling by recruiting phosphatase SHP2. J Exp Med. 209(6):1201–1217. [DOI] [PMC free article] [PubMed] [Google Scholar]
- York IA, Chang SC, Saric T, Keys JA, Favreau JM, Goldberg AL, Rock KL. 2002. The ER aminopeptidase ERAP1 enhances or limits antigen presentation by trimming epitopes to 8–9 residues. Nat Immunol. 3(12):1177–1184. [DOI] [PubMed] [Google Scholar]
- Yoshida Y, Adachi E, Fukiya K, Iwai K, Tanaka K. 2005. Glycoprotein-specific ubiquitin ligases recognize N-glycans in unfolded substrates. EMBO Rep. 6(3):239–244. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Yoshida Y, Tokunaga F, Chiba T, Iwai K, Tanaka K, Tai T. 2003. Fbs2 is a new member of the E3 ubiquitin ligase family that recognizes sugar chains. J Biol Chem. 278(44): 43877–43884. [DOI] [PubMed] [Google Scholar]
- Younger JM, Chen L, Ren HY, Rosser MF, Turnbull EL, Fan CY, Patterson C, Cyr DM. 2006. Sequential quality-control checkpoints triage misfolded cystic fibrosis transmembrane conductance regulator. Cell. 126(3):571–582. [DOI] [PubMed] [Google Scholar]
- Yu H, Kopito RR. 1999. The role of multiubiquitination in dislocation and degradation of the alpha subunit of the T cell antigen receptor. J Biol Chem. 274(52):36852–36858. [DOI] [PubMed] [Google Scholar]
- Yu S, Ito S, Wada I, Hosokawa N. 2018. ER-resident protein 46 (ERp46) triggers the mannose-trimming activity of ER degradation-enhancing α-mannosidase-like protein 3 (EDEM3)). J Biol Chem. 293(27):10663–10674. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Zehner M, Chasan AI, Schuette V, Embgenbroich M, Quast T, Kolanus W, Burgdorf S. 2011. Mannose receptor polyubiquitination regulates endosomal recruitment of p97 and cytosolic antigen translocation for cross-presentation. Proc Natl Acad Sci USA. 108(24):9933–9938. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Zehner M, Marschall AL, Bos E, Schloetel JG, Kreer C, Fehrenschild D, Limmer A, Ossendorp F, Lang T, Koster AJ, et al. 2015. The translocon protein Sec61 mediates antigen transport from endosomes in the cytosol for cross-presentation to CD8(+) T cells. Immunity. 42(5):850–863. [DOI] [PubMed] [Google Scholar]
- Zettl M, Adrain C, Strisovsky K, Lastun V, Freeman M. 2011. Rhomboid family pseudoproteases use the ER quality control machinery to regulate intercellular signaling. Cell. 145(1):79–91. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Zhang T, Kho DH, Wang Y, Harazono Y, Nakajima K, Xie Y, Raz A. 2015. Gp78, an E3 ubiquitin ligase acts as a gate-keeper suppressing nonalcoholic steatohepatitis (NASH) and liver cancer. PLoS One. 10(3):e0118448. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Zhang T, Xu Y, Liu Y, Ye Y. 2015. gp78 functions downstream of Hrd1 to promote degradation of misfolded proteins of the endoplasmic reticulum. Mol Biol Cell. 26(24): 4438–4450. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Zhang Y, Baig E, Williams DB. 2006. Functions of ERp57 in the folding and assembly of major histocompatibility complex class I molecules. J Biol Chem. 281(21):14622–14631. [DOI] [PubMed] [Google Scholar]
- Zhong X, Pittman RN. 2006. Ataxin-3 binds VCP/p97 and regulates retrotranslocation of ERAD substrates. Hum Mol Genet. 15(16):2409–2420. [DOI] [PubMed] [Google Scholar]
- Zhong X, Shen Y, Ballar P, Apostolou A, Agami R, Fang S. 2004. AAA ATPase p97/valosin-containing protein interacts with gp78, a ubiquitin ligase for endoplasmic reticulum-associated degradation. J Biol Chem. 279(44):45676–45684. [DOI] [PubMed] [Google Scholar]
- Zhou B, Guo L, Zhang B, Liu S, Zhang K, Yan J, Zhang W, Yu M, Chen Z, Xu Y, et al. 2019. Disulfiram combined with copper induces immunosuppression via PD-L1 stabilization in hepatocellular carcinoma. Am J Cancer Res. 9(11): 2442–2455. [PMC free article] [PubMed] [Google Scholar]
- Zitvogel L, Kepp O, Senovilla L, Menger L, Chaput N, Kroemer G. 2010. Immunogenic tumor cell death for optimal anticancer therapy: the calreticulin exposure pathway. Clin Cancer Res. 16(12):3100–3104. [DOI] [PubMed] [Google Scholar]