Open Access

Role of ubiquitin regulatory X domain‑containing protein 3B in the development of hepatocellular carcinoma (Review)

  • Authors:
    • Ziwei Guo
    • Jun Liang
  • View Affiliations

  • Published online on: February 9, 2023     https://doi.org/10.3892/or.2023.8494
  • Article Number: 57
  • Copyright: © Guo et al. This is an open access article distributed under the terms of Creative Commons Attribution License.

Metrics: Total Views: 0 (Spandidos Publications: | PMC Statistics: )
Total PDF Downloads: 0 (Spandidos Publications: | PMC Statistics: )


Abstract

The majority of new cases and fatalities from hepatocellular carcinoma (HCC) occur in China; however, the overall morbidity and mortality rates are decreasing. A major risk factor due to the evolving epidemiology is improper lipid metabolism. Although investigations on aberrant lipid metabolism are numerous, there are only a limited number of studies available on proteasomal degradation processes. The degradation process is mainly involved in endoplasmic reticulum stabilization, the balance of lipid metabolism, and physiological functions of Golgi apparatus, endoplasmic reticulum, lysosomes and other organelles, however, this process has been little studied in the development of tumorigenesis. In order to provide some theoretical support for future research on ubiquitin regulatory X domain‑containing protein 3B (UBXN3B), the present review focuses on the role of UBXN3B, which is involved in the stabilization of the endoplasmic reticulum and the maintenance of lipid homeostasis, as well as in the promotion and development of non‑alcoholic fatty liver disease and HCC.

Introduction

Hepatocellular carcinoma (HCC) is the fourth most prevalent type of cancer worldwide and the sixth most frequent malignancy (1). In 2020, 45% of the year's new cases (910,000) and 47% of the year's fatalities (830,000) occurred in China (2). The data for GCC in China are still not encouraging, despite the recent worldwide reduction in the incidence of this type of cancer. The explanation for this may be related to a shift in the etiology; an increasing number of HCC cases are being linked to faulty lipid metabolism, which also causes non-alcoholic fatty liver disease (NAFLD), non-alcoholic steatohepatitis and even HCC. As a result, a new risk factor is replacing the classic theory of the evolution of viral hepatitis, namely abnormal lipid metabolism.

The dysregulation of lipid droplets (LDs) may interact with the endoplasmic reticulum (ER), mitochondria, peroxisomes, vesicles and lysosomes under normal oxygen conditions (3). In addition, the tumor microenvironment may lead to dysregulation via factors, such hyperinsulinemia caused by insulin resistance and increased pro-inflammatory cytokine levels (4). Previous research has emphasized the role of modifications in signaling pathways and enzyme metabolism during the production of fatty acid synthesis (FAS) (5). However, available data on the association between the ubiquitin regulatory X domain-containing protein (UBX) family and endoplasmic reticulum-associated degradation (ERAD) are limited. The UBX family, in contrast to earlier research (6), increases cellular stability, primarily through ERAD or increased intranuclear translocation mechanisms, a process that even inhibits FAS. Despite this apparent contradiction with the main line of inquiry, there is still evidence to support the increased expression of UBX family genes or proteins in HCC and their roles as poor prognostic factors (7).

The UBX family, which consists of the 13 members UBXN1, UBXN2A-2C, UBXN3A-3B, UBXN4 and UBXN6-11, has a structural domain that is comparable to the N terminus of ubiquitin. The N terminus of the receptor protein is identified and transported to the proteasome, where it undergoes the standard protein degradation process and finally degrades into smaller polypeptides and amino acids (8). An essential organelle for the synthesis, folding, secretion and recycling of proteins across organelles is the ER. When elements such as Ca2+ levels, energy and nutrition are aberrant, abnormal protein folding occurs in the ER, a process known as ER stress, which is required to maintain ER homeostasis (9). As a result, ERAD is a crucial process that eliminates unfolded or misfolded proteins when ER stress arises in order to return the ER to its normal state and function (10).

Of the UBX family, nine members, namely UBXN1, UBXN2A-2C, UBXN3B, UBXN4, UBXN6, UBXN8 and UBXN10, are involved in ERAD during lipid metabolism (Fig. 1). UBXN1 interacts with apoptosis protein inhibitors, blocks and interferes with retinoic acid-inducible gene (RIG-I)-like receptors and the nuclear factor (NF)-κB pathway, and is a key player in cell signaling, endocytosis and DNA damage repair (11). UBXN1 is a component of the reverse translocation degradation complex (11) and is involved in ER stress-mediated de-glycosylation (11). Trimers of UBXN2A, 2B and 2C bind to cytoplasmic p97 and are involved in the stability of the ER, the Golgi apparatus and the rearrangement of the mitotic terminal (12). Based on remodeling mechanisms, p97, an enzyme belonging to the adenosine triphosphate (ATP) enzyme family, is connected to a number of cellular processes and activities (13). p97 may alternatively be considered as an ‘enzymatic dissociative activity’ protein (13). p97 has two different mechanisms for protein degradation: One involves the p97-Ufd1-Npl4 complex, which binds to various ubiquitinated ERAD substrates on the cytoplasmic side of the ER membrane before being reverse transcribed and transported to the proteasome for degradation (14); the other involves a protein degradation pathway that connects the ER to p97 through a number of cofactors, primarily members of the UBX family. In addition to regulating the ERAD pathway to preserve ER function under conditions of ER stress (15), UBXN3B plays a crucial role in ERAD by limiting the activity of phospholipases in LDs and preventing the degradation of LDs (15). In order to attract valosin-containing protein (VCP) to the ER and encourage ERAD, UBXN4, an essential membrane protein of the ER, serves as a platform (16). UBXN6 participates in lysosomal degradation, may play a role in misfolded proteins in ERAD, and may adversely affect ATP-driven VCP (17). One of the cofactors of p97, VCP, is a component of the ATPase complex known as UBXN8, which binds to p97 and promotes ERAD. VCP/p97 factor UBXN10, a VCP/p97 binding protein necessary for cilia promotion, has a VCP/p97 substrate specificity (18).

In the UBX family, UBXN3B has a specific and crucial function in cancer cells when LD production and enhanced ERAD occur simultaneously. Other members, on the other hand, perform unique tasks relating to cellular activity, apoptosis, innate immunity, Golgi apparatus and lysosomes. As a result, the main aim of the present review was to describe the mechanisms through which UBXN3B contributes to the development of HCC.

Transcriptional regulation of UBXN3B

UBXN3B maintains ER stability

UBXN3B is a hairpin-like structural protein that resembles a hairpin and is comprised of hydrophobic amino acid residues that are introduced into the ER cytoplasm. UBXD8 migrates from the ER to the surface of fatty acid-rich LDs in fatty acid-deficient cells (19), and it has a UBX structural domain that interacts with p97 (13), which is necessary for ERAD (8). By repressing the transcriptional activity and target of NAFLD, UBXD8 inhibits the transcriptional activity of the liver X receptor (LXR), a key regulator of the enterohepatic cycle. It also inhibits the transcriptional activity of the sterol regulatory element-binding protein 1 (SREBP-1), and it stimulates the transcription of genes encoding proteins necessary for FAS (20). FAS, acetyl coenzyme A carboxylase, and stearoyl coenzyme A desaturase-1 are all directly activated by the LXR (20). The first and rate-limiting steps of FAS, which are carried out by these three enzymes, control how rapidly monounsaturated fatty acids are produced. The transcription factor termed SREBP-1, which is found in the ER, stimulates the expression of all the genes necessary for fatty acid metabolism (21). Its cytoplasmic N-terminal structural domain is connected to the insulin-inducible gene 1 (INSIG-1) and is located there (22). Without fatty acids, UBXD8 binds to INSIG-1 and induces the rapid proteasomal degradation of the receptor protein by attracting p97 to the protein (23). SREBP-1 is moved from the ER to the Golgi without INSIG-1, where it is broken by two Golgi-localized proteases (24). This cleavage allows SREBP-1 to enter the nucleus and activate all FAS-required genes, as it frees the protein's N-terminal structural domain from the membrane (21).

By identifying and destroying the ubiquitin-like structural domains of lipid-binding proteins in the ER, ERAD guarantees the structural normalcy of the ER. Hepatocytes release apolipoprotein (Apo) B-100, a glycoprotein with a molecular weight >500 kDa. Several phases in the lipoprotein transport mechanism control the production of this protein. When there are more lipids in the ER lumen, Apo B-100 may develop. Lipid-carrying Apo B-100 is ubiquitinated by ERAD, which causes the proteasome to degrade lipid-poor Apo B-100. On the other hand, ubiquitinated Apo B-100 builds up in the LDs when the proteasome is blocked, and it has been proposed that this may act as a platform for Apo B-100 breakdown (25). It should be noted that this is only conjecture. According to another study, Apo B-100 predominantly lipidates and builds up in LDs (25). As lipidation only occurs in the ER lumen, lipidated Apo B-100 is moved to the cytoplasmic side near the LDs. Thus, the buildup of lipidated Apo B-100 aids in the development of specific Apo B-100 structures linked to ER-LD. It should be noted that UBX family members are ubiquitinated proteins that the ubiquitination-proteasome system degrades under ‘non-essential’ conditions (26). In other words, ubiquitinated proteins may be deubiquitinated and retrieved in a hypothesized non-degradation mechanism rather than necessarily being subject to destruction (26). According to this hypothesis, damaged proteins either accumulate or continue to degrade in response to environmental changes and ERAD only functions under specified circumstances. An essential protein involved in ER stability is the p97-UBXN3B complex.

UBXN3B maintains fatty acid and triglyceride homeostasis

The p97-UBXN3B complex regulates the production of triglycerides and breaks down ubiquitinated proteins in the ER. p97-UBXN3B controls triglyceride metabolism, in addition to serving as a sensor of long-chain unsaturated fatty acids (15,23,24). Unsaturated fatty acids enhance the purification and polymerization of UBXN3B (15) when it is cultivated in vitro; in cells without lipids, UBXN3B prevents triglyceride production, since this process requires the attachment and the conversion of fatty acids (27). Fatty acids in cells are linked to phospholipids and do not participate in triglyceride production when triglyceride synthesis is terminated (15). Excess long-chain unsaturated fatty acids have the ability to polymerize UBXN3B and interfere with its ability to perform its functions, resulting in unaltered triglyceride production (15). By blocking the rate-limiting enzyme of triglyceride production, the recruitment of p97 from the ER to the LD surface by UBXN3B increases the size of LDs and prevents triglyceride hydrolysis to fatty acids (15). While saturated fatty acids are unable to interact with UBXN3B, they promote the conversion of extra unsaturated fatty acids into triglycerides for storage in the LDs and prevent breakdown by attaching to phospholipases and blocking their activity (15). Since UBXN3B promotes triglyceride accumulation in the LDs, while inhibiting triglyceride production and binding to phospholipids in the ER, this protein constantly cycles between the two tissues to carry out its various roles (19).

Other mechanisms

Coenzyme 3-hydroxy-3-methylglutaryl coenzyme A reductase (HMGCR) is a necessary protein for the ER membrane, and p97 and ERAD are required for its destruction (26). The degradation of HMGCR induced by sterols is prevented by the silencing of UBXN3B (28). In hepatocytes, UBXN3B is a poor predictor of HMGCR degradation. It can thus be hypothesized that UBXN3B knockdown promotes the production of cholesterol. However, in hepatocytes with higher HMGCR levels induced by sterol depletion, there were no changes in messenger ribonucleic acid (mRNA) or protein expression levels, indicating that sterol-dependent UBXN3B expression did not promote HMGCR breakdown (28). Type I interferon and immunological inflammatory reactions are promoted by the stimulatory interferon genes (STING) (29). The UBX protein family activates and controls the biological activities of the interferon-stimulated response element (ISRE) (30). High amounts of UBXN3B do not, however, substantially promote ISRE, indicating that UBXN3B and STING-dependent signaling pathways only have a positive connection. Additionally, STING is ubiquitinated, dimerized and is maintained in a phosphorylated state by UBXN3B, which causes SRING to be destroyed by binding to p97 and the other E3 ubiquitin ligases (8). Although the aforementioned HMGCR degradation, ISRE activation and STING stimulation do not appear to directly interact with UBXN3B, numerous specific mechanisms of these pathways still need to be addressed by more extensive research.

Mechanisms of progression from NAFLD to HCC

LD metabolic disorders are associated with a number of metabolic illnesses, including obesity, fatty liver, diabetes and cardiovascular disease (31). In clinical practice, blood concentrations of total cholesterol, high-density lipoprotein (HDL), low-density lipoprotein (LDL), triglycerides and free fatty acids (FFA) are used to determine the lipid status of a patient. Diseases with high levels of LDs enhance the risk of tumorigenesis in viral infections (32).

The liver serves as the hub for lipid metabolism (33). Issues regarding systemic glucose and NAFLD are tightly related (34). Although excess hepatic lipid levels have been established to be an independent risk factor and are strongly associated with the development of HCC, the mechanisms through which this contributes to fatty acid metabolism remain unclear (35).

By interfering with signaling pathways and cytokines via a variety of methods, lipid dysregulation may either directly or indirectly contribute to the development of cancers (e.g., lipid regulation, spontaneous synthesis of lipids, ER stress, increased inflammatory cytokines and immune cells).

Abnormal lipid regulation

Enzymes expressed by a variety of key transcriptional proteins control normal lipid metabolism (36). Triglycerides do not specifically harm or destroy cells (8). When the accumulation of triglycerides sensitizes cells to damage, it disrupts signaling pathways and gene function, leading to the dysfunction of lipid-related factors (37).

De novo biosynthesis

The proliferation, invasion and metastasis of cancer cells, including those in hepatocellular, breast, renal, colorectal and prostate malignancies, are mediated by high levels of LDs (38). The unique function of the liver is to both use and resynthesize FFAs, which are released by other organs. External lipid absorption is the primary mechanism through which the enhanced metabolism required for lipolysis as a means of cell survival is accomplished. Instead, during the process of cellular energy acquisition, the citric acid cycle, ATP citrate lyase, and acetyl coenzyme A carboxylase may transform the pyruvate generated by glycolysis into oxaloacetate and acetyl coenzyme in the mitochondria (39). Saturated fatty acids may be created from acetyl coenzyme A via binding and conversion mechanisms (40). Stearoyl coenzyme A desaturase converts them to monounsaturated fatty acids (39,40). In this particular de novo biosynthesis procedure, the key building blocks of prostaglandin and membrane production are unsaturated fatty acids. By triggering autophagy, promoting cell membrane renewal, affecting intracellular signaling and gene transcription, and boosting energy generation, the unsaturated fatty acids are crucial for cell survival. Energy is produced in this process by lipid ‘starvation’, which combines extracellular resources with lipid de novo production (41). ATP citrate lyase and acetyl coenzyme A carboxylase are abundantly expressed in NAFLD after FAS has developed, increasing FAS, an early sign of NAFLD and fibrosis (42).

ER stress

ER stress may be observed in patients with NAFLD (43). The reduction of ER stress improves NAFLD (43). Even in the absence of carcinogenic therapy, spontaneous fatty liver disease may result in HCC, indicating that ER stress is sufficient to convert NAFLD to HCC. Mechanistically, increased levels of ER stress cause macrophages to produce increased levels of tumor necrosis factor (TNF), which promotes cell growth, anti-apoptotic activity and eventually, tumor development (13). These findings suggest that when NAFLD progresses to HCC, ER stress may initiate malignant transformation. The section that follows provides a more in-depth discussion of what is connected to ER stress.

Inflammatory factors associated with NAFLD

TNF-α and interleukin (IL)-6 are two inflammatory factors that are linked to NAFLD (44). Additionally, NAFLD induces the production of anti-inflammatory cytokines, such as IL-10 or IL-1 receptor antagonists, which prevent NF-κB from being activated and prevent the release of chemokines, TNF-α and IL-6 (44,45). TNF-α expression is elevated in individuals with NAFLD, cirrhosis and HCC, which results in the release of additional cytokines and chemokines (45,46). TNF-α gene polymorphisms also have a greater propensity to cirrhosis and NAFLD (45,46).

IL-6 is a significant inducer of C-reactive protein and hepatocyte production (47) and may play a role in NAFLD. The direct pathogenic factors are IL-6 and TNF-α. In addition to IL-6 and TNF-α, hepatocyte injury, the disruption of signaling cascades and functional protein loss are also induced by IL-10, IL-1 inhibitors and growth factors, which in turn result in aberrant lipid metabolism. In response to stress-induced intracellular changes, a surge in inflammatory substances alters LD proteins and signaling pathways, which in turn causes cancer cell conversion.

Macrophage function

Numerous immune cells are found in the liver, where they are affected by portal blood and endure a special tolerant environment where they may react to foreign pathogens, but avoid innocuous antigens caused by dietary antigens and microbial products (48,49). An increased inflammatory activity is associated with the lipid burden in macrophages (50). M1- and M2-macrophages may arise as a result of macrophage involvement, depending on the changed microenvironment. While the latter has an anti-inflammatory and immunomodulatory function, the former causes the generation of inflammatory cytokines (51).

M1-macrophages are known to play a role in NAFLD, with the bacterial endotoxin, lipopolysaccharide, and interferon-γ helping to activate M1-macrophages and increase the production of pro-inflammatory cytokines and chemokines, reactive oxygen species and nitric oxide. According to research, individuals with NAFLD have higher hepatic levels of certain M1-specific cytokines and chemokines, such as IL-1β, IL-18, and chemokines 2–5 and 9–11 (52). The increased activation of cytokines and chemokines, in addition to lipids, oxidation products, or chemicals produced following hepatocyte damage that may directly contribute to liver injury, can aggravate M1-transformation (53). Changes in the M1/M2 phenotype of macrophages may be influenced by the environment. Lipid-rich dendritic cells (DCs) struggle to digest antigens in the presence of tumors (54). Similar to this, under conditions of steatosis, DCs are recruited into hepatocytes and are maintained at high levels, despite the fact that their function is activated. Conversely, the depletion of DCs leads to an increase in liver inflammation, a decrease in the numbers of Treg, an increase in CD8+ T-cell function, an increase in immune effector cell activity and the production of pro-inflammatory cytokines, an increase in hepatocyte apoptosis, and ultimately, in the acceleration of liver fibrosis (55). However, another study found higher levels of natural killer (NK)p46+ cells in NAFLD, which trigger local and invading macrophages to differentiate into M1 and stop fibrosis from inducing M2-cells (56). M2-macrophages, however, have been linked to HCC brought on by NAFLD (57). The influence of the macroenvironment on macrophage activity and its involvement in the development of NAFLD-promoted HCC warrant further investigation.

Role of UBXN3B in HCC

ER stress

Increasing attention has been paid to the role that ER stress plays in the growth, metastasis, angiogenesis and even treatment resistance of HCC cells (58). Proteotoxic ER stress refers to disruptions in protein folding in the ER, which triggers unfolded protein responses (UPRs). UPR activation by ER stress is often observed as an adaptive mechanism to preserve in vivo protein homeostasis. In terms of stability to mRNA and ERAD substrates and modified signaling pathways, the present review also discusses the mechanisms through which the UBXN3B protein reacts to ER stress.

Stability of mRNA

The biogenesis and metabolic functions of mRNAs are connected to a number of different proteins. Pre-mRNA processing, nuclear export, translation, localization and mRNA decay processes are the key factors influencing the remodeling events that the resultant mRNA-protein complexes (mRNPs) go through (59). The most extensively studied effect on mRNPs is the influence of ATP-dependent RNA helicases, through which mRNAs are modified to facilitate the ‘metabolism’ of mRNPs (58). The UBX family plays a significant role in preserving their stability, since they rely on ubiquitination signals; HuR, a dominant binding protein that binds multiple AU-rich regions, is one of these mRNA stabilizers and performs a crucial stabilizing function as a cytokine and transcription factor under conditions of cellular stress (58). HuR is often overexpressed and serves as a representation of the very dynamic alterations that occur during the recombination and dissociation of mRNPs (60). Researchers have investigated how phosphorylated HuR regulates protein abundance by influencing the location and stability of organelles (61). That is, phosphorylated HuR maintains mRNA stability, while transporting proteins to the nucleus or degrading proteins in the ER to regulate protein abundance in the cytoplasm. Through a non-degradative ubiquitination signaling mechanism that disrupts the metabolism of mRNPs, the p97-UBXD8 complex affects HuR. In fact, HuR only interacts with the p97-UBXD8 complex and not p97 or UBXD8 alone (7). The primary characteristic of the p97-UBXD8 complex, among the ubiquitination signals of the non-degradation route, is that the ubiquitinated HuR is not degraded by the ubiquitin-proteasome system (62). The protein may instead be deubiquitinated and regenerated via a process known as a non-degradation route. According to a previous study (63), the p97-UBXD8 complex is involved in HuR-mRNA modification during the stress response. When cancer develops, the stabilizing factors, the p97-UBXD8 complex and the phosphorylated HuR transition, are activated to serve their stabilizing roles after organelle function and structure have been compromised, mostly by transitory proliferation and metabolic abnormalities. The UBX family, particularly the p97-UBXD8 complex, is one of these modifications that not only improves cellular stability, but also controls it through autoregulation.

Stabilization of ERAD substrates

Based on the heterogeneity of the N-terminal structural domains of the UBX family, wherein only five of the 13 proteins in this family possess N-terminal AAA-enriched structures in mammals (UBXN1, UBXN2C, UBXN3A, UBXN3B and UBXN7), the UBX proteins have been split into two groups (8,11,64,). Park et al (64) reported that these five proteins fold abnormally to the ER, as ERAD substrates. The expression of all five proteins was shown to be upregulated in cells treated with cyclooxygenase, according to RT-PCR data (64). Among these proteins, UBXN2C, UBXN1 and UBXN3B are ‘immediate’ responders to endogenous stress, whereas UBXN3A and UBXN7 are ‘late’ responders. This indicates that whereas UBXN3A and UBXN7 are affected by other variables and do not reflect the ER stress in a timely and efficient manner, UBXN2C, UBXN1 and UBXN3B directly reflect increased ER stress.

Of note, UBXN2C and UBXN3B have higher expression levels than UBXN1 across all ERAD substrates, but UBXN1 has lower expression levels (11). Additional research revealed that UBXN1 has no affinity for certain ERAD substrates. Stable α-T-cell antigen receptor (α-TCR)-expressing cells under conditions of stress exhibit higher levels of UBXN2C and UBXN3B and lower levels of UBXN1. When UBXN2C or UBXN3B are overexpressed, the degradation of α-TCR occurs more rapidly; UBXN1 has the reverse effect. The degradation of α-TCR is caused by the overexpression of UBXN1. ERAD is one of the key elements in overcoming ER stress by activating the UPR (11). These findings imply that the expression levels of these five genes are altered by both ER stress and the overexpression of ERAD substrates (64). Although these five proteins function as the primary proteins for proteasomal degradation, the expression of several genes affects the outcomes when a certain role is played. This paradox can be explained by the fact that, on the one hand, UPRs build up in the ER and that, under conditions of ER stress, the overexpression of ERAD substrates can partially reduce this buildup; on the other hand, an increase in misfolded proteins in the cell and an increase in their demand can decrease the rate of proteasomal degradation of ERAD substrates. When UBXN2C and UBXN3B levels are high in a significant number of misfolded proteins, particularly in cells expressing α-TCR, and when UBXN1 is downregulated to further promote degradation, this may explain the enhanced participation of UBXN2C and UBXN3B in the feedback loop during ER stress.

Altered signaling pathways

Under typical circumstances, the three primary transmembrane sensors inositolase-1α, protein kinase RNA-like ER kinase (PERK) and activated transcription factor-6α (ATF-6α) are all active in the lumen. Apoptosis may be caused if ER homeostasis is not recovered.

The inositolase-1α pathway is a transmembrane protein that is present in the UPRs and has kinase and ribonucleic acid endonuclease as its cytoplasmic structural domains (65). In a healthy state, inositolase-1 binds to immunoglobulins and is inactive. In response to stress, inositolase-1 is released, dimerized and activated, resulting in conformational alterations. As an alternative, unfolded proteins may bind directly to the structural domain of inositolase-1, causing conformational alterations and activation (66). The transcription factor known as the X-box binding protein, which is produced as a result of activated inositolase-1α, improves the capacity of the protein to fold and the function of ERAD in the ER. When ER stress is unrecoverable, the function of inositolase-1 is interrupted, which causes mRNA that is linked to it to degrade or to be recruited by TNF, which then stimulates cellular and mitochondrial death via apoptotic signaling (67). As a result, the inositolase-1 pathway reacts to ER stress by either boosting apoptotic pathways or upregulating ERAD, both of which include the UBX protein family. By inhibiting and interfering with the RIG-I-like receptors and the NF-κB pathway, UBXN1 is a member of the complex necessary to engage in the de-glycosylation and proteasome-mediated destruction of misfolded proteins under ER stress via reverse translocation (11). Similar to inositolase-1, PERK is a transmembrane protein that is inactive in a physiological setting (68). Dimerization and tetrameric pattern-mediated autophosphorylation are required for its activation (69). When PERK is activated, it may also phosphorylate translation initiation factors (70), which causes ERAD to reduce the amount of protein (71). This pathway also causes cell death by upregulating apoptosis-related genes (71). ATF-6α is translocated to the Golgi apparatus when the ER is stressed, despite the fact that it is primarily engaged in the control of ER plasmalogens (72).

Abnormal lipid metabolism

As a type of proteotoxic ER stress, the changed protein folding burden in the ER (73) interferes with aberrant protein folding, misfolded, unfolded or altered folding density. UPRs are also known as lipotoxic ER stress, as they may be directly triggered by toxic lipids in addition to being reliant on the buildup of misfolded proteins. Both forms of stress are induced by the involvement of the ER in the folding and transport of proteins, which is connected to lipid production and transport and activates UPRs to bring about homeostasis.

Genes associated with FAS, such as ATP acetyl coenzyme A carboxylase, which causes the conversion of citric acid into acetyl coenzyme A, malonyl coenzyme A, and fatty acids, are often overexpressed or upregulated in HCC (74). Through the examination of gene expression, a number of studies have investigated the causes and purposes of HCC development (6–8,11,14–18). As has already been established, as UBXN3B levels increase, so do the amounts of lipidated Apo B-100 in LDs and ubiquitinated Apo B-100 in ER. Both ubiquitinated and lipidated Apo B-100 have the ability to speed up proteasomal breakdown, while promoting lipid transport. The associated metabolism between LD and ER is regulated by UBXN3B.

Changes in the microenvironment

NK T-cells (NKT cells) are innate and adaptive immune cells with a variety of immunomodulatory functions (75). Changes in the metabolic profile of tumor lipids may also modify the type of lipid antigens, which may affect the immunomodulatory activity of NKT cells, as they predominantly detect lipid antigens (76). In a mouse model of NAFLD, increased lipids were shown to cause NKT cell death, which reduced the amount of hepatic NKT cells (77). Type I NKT cells are activated by a lipid surplus, and this leads to a more potent pro-inflammatory cytokine environment. However, despite the higher hepatic lipid content in HCC, another study found no significant difference in the number of NKT cells (78). These contradictory results suggest that further more in-depth experiments are required to examine the effects of lipid changes on NKT cells in NAFLD and HCC.

Additionally, processes including aberrant lipid metabolism and Golgi expansion control the proliferation of cells, such as myeloid-derived suppressor cells, CD8+ T-cells, DCs and tumor-associated macrophages, which are all implicated in the growth of tumors (79). The expression of the UBX protein and the development of HCC may be affected by mitochondrial defects, changes in lipid signaling molecules and pathways, fatty acid biosynthetic pathways, lipidomics, other genetic mutations, chronic viral infections, cholesterol efflux factors, ER autophagy, and post-translational modifications of proteins. The mechanisms through which the UBX protein family affects ER stress, lipid metabolism and microenvironmental changes remain unknown.

Conclusions and future perspectives

As a member of the UBX family, UBXN3B is primarily involved in the ER stress mechanism known as ERAD. It performs a variety of roles in LDs and ERs under various clinical conditions and develops into a new HCC biomarker. In order to maintain fatty acid and triglyceride homeostasis, maintain intracellular signaling pathways, and normalize cytokines, UBXN3B participates in ER stability, maintains fatty acid and triglyceride homeostasis, and has an impact on cholesterol production. UBXN3B is also involved in the progression of ER stress, the dysregulation of lipid metabolism, the driving of inflammatory factors and the immune microenvironment. However, a number of physiological processes of UBXN3B remain unknown and require support and clarification by further fundamental experimental and clinical studies.

Acknowledgements

Not applicable.

Funding

The present study was supported by a Peking University International Hospital Research Grant (no. YN2022QN12).

Availability of data and materials

Not applicable.

Authors' contributions

ZG and JL drafted, read and approved the final manuscript, and conceived and designed the study. Data authentication is not applicable.

Ethics approval and consent to participate

Not applicable.

Patient consent for publication

Not applicable.

Competing interests

The authors declare that they have no competing interests.

Glossary

Abbreviations

Abbreviations:

HCC

hepatocellular carcinoma

UBXN3B

ubiquitin regulatory X domain-containing proteins 3B

ER

endoplasmic reticulum

NAFLD

non-alcoholic fatty liver disease

ERAD

endoplasmic reticulum-associated degradation

NASH

non-alcoholic steatohepatitis

LDs

lipid droplets

FAS

fatty acid synthase

RIG-I

retinoic acid-inducible

NF-κB

nuclear factor-κB

ATP

adenosine triphosphate

VCP

valosin-containing protein

LXRα

liver X receptor α

SREBP-1

sterol regulatory element-binding protein 1

INSIG-1

insulin-inducible gene 1

HMGCR

3-hydroxy-3-methylglutaryl coenzyme A reductase

mRNA

messenger ribonucleic acid

STING

stimulator of interferon genes

IFN-I

type I interferon

ISRE

interferon-stimulated response element

HDL

high-density lipoprotein

LDL

low-density lipoprotein

FFAs

free fatty acids

TNF

tumor necrosis factor

IL

interleukin

DCs

dendritic cells

NKT cells

natural killer T-cells

UPR

unfolded protein response

mRNPs

mRNA-protein complexes

AREs

AU-rich elements

α-TCR

α-T-cell antigen receptor

PERK

protein kinase RNA like ER kinase

ATF-6α

activated transcription factor-6α

References

1 

Rawla P, Sunkara T, Muralidharan P and Raj JP: Update in global trends and aetiology of hepatocellular carcinoma. Contemp Oncol (Pozn). 22:141–150. 2018.PubMed/NCBI

2 

World Health Organization, . GLOBOCAN 2020 Graph production. http://gco.iarc.fr/todaySeptember. 2021

3 

Blanchette-Mackie EJ, Dwyer NK, Barber T, Coxey RA, Takeda T, Rondinone CM, Theodorakis JL, Greenberg AS and Londos C: Perilipin is located on the surface layer of intracellular lipid droplets in adipocytes. J Lipid Res. 36:1211–1226. 1995. View Article : Google Scholar : PubMed/NCBI

4 

Shalapour S, Lin XJ, Bastian IN, Brain J, Burt AD, Aksenov AA, Vrbanac AF, Li W, Perkins A, Matsutani T, et al: Inflammation-induced IgA+ cells dismantle anti-liver cancer immunity. Nature. 551:340–345. 2017. View Article : Google Scholar : PubMed/NCBI

5 

Muir K, Hazim A, He Y, Peyressatre M, Kim DY, Song X and Beretta L: Proteomic and lipidomic signatures of lipid metabolism in NASH-associated hepatocellular carcinoma. Cancer Res. 73:4722–4731. 2013. View Article : Google Scholar : PubMed/NCBI

6 

Zhou HL, Geng C, Luo G and Lou H: The p97-UBXD8 complex destabilizes mRNA by promoting release of ubiquitinated HuR from mRNP. Genes Dev. 27:1046–1058. 2013. View Article : Google Scholar : PubMed/NCBI

7 

Kloppsteck P, Ewens CA, Förster A, Zhang X and Freemont PS: Regulation of p97 in the ubiquitin-proteasome system by the UBX protein-family. Biochim Biophys Acta. 1823:125–129. 2012. View Article : Google Scholar : PubMed/NCBI

8 

Rezvani K: UBXD proteins: A family of proteins with diverse functions in cancer. Int J Mol Sci. 17:17242016. View Article : Google Scholar : PubMed/NCBI

9 

Liao Z, Luo R, Li G, Song Y, Zhan S, Zhao K, Hua W, Zhang Y, Wu X and Yang C: Exosomes from mesenchymal stem cells modulate endoplasmic reticulum stress to protect against nucleus pulposus cell death and ameliorate intervertebral disc degeneration in vivo. Theranostics. 9:4084–4100. 2019. View Article : Google Scholar : PubMed/NCBI

10 

Nakatsukasa K, Huyer G, Michaelis S and Brodsky JL: Dissecting the ER-associated degradation of a misfolded polytopic membrane protein. Cell. 132:101–112. 2008. View Article : Google Scholar : PubMed/NCBI

11 

Mukkavalli S, Klickstein JA, Ortiz B, Juo P and Raman M: The p97-UBXN1 complex regulates aggresome formation. J Cell Sci. 134:jcs2542012021. View Article : Google Scholar : PubMed/NCBI

12 

Uchiyama K, Totsukawa G, Puhka M, Kaneko Y, Jokitalo E, Dreveny I, Beuron F, Zhang X, Freemont P and Kondo H: p37 is a p97 adaptssswor required for Golgi and ER biogenesis in interphase and at the end of mitosis. Dev Cell. 11:803–816. 2006. View Article : Google Scholar : PubMed/NCBI

13 

Her NG, Toth JI, Ma CT, Wei Y, Motamedchaboki K, Sergienko E and Petroski MD: p97 composition changes caused by allosteric inhibition are suppressed by an on-target mechanism that increases the enzyme's ATPase activity. Cell Chem Biol. 23:517–528. 2016. View Article : Google Scholar : PubMed/NCBI

14 

Ye Y, Meyer HH and Rapoport TA: The AAA ATPase Cdc48/p97 and its partners transport proteins from the ER into the cytosol. Nature. 414:652–656. 2001. View Article : Google Scholar : PubMed/NCBI

15 

Olzmann JA, Richter CM and Kopito RR: Spatial regulation of UBXD8 and p97/VCP controls ATGL-mediated lipid droplet turnover. Proc Natl Acad Sci USA. 110:1345–1350. 2013. View Article : Google Scholar : PubMed/NCBI

16 

Liang J, Yin C, Doong H, Fang S, Peterhoff C, Nixon RA and Monteiro MJ: Characterization of erasin (UBXD2): A new ER protein that promotes ER-associated protein degradation. J Cell Sci. 119:4011–4024. 2006. View Article : Google Scholar : PubMed/NCBI

17 

Nagahama M, Ohnishi M, Kawate Y, Matsui T, Miyake H, Yuasa K, Tani K, Tagaya M and Tsuji A: UBXD1 is a VCP-interacting protein that is involved in ER-associated degradation. Biochem Biophys Res Commun. 382:303–308. 2009. View Article : Google Scholar : PubMed/NCBI

18 

Raman M, Sergeev M, Garnaas M, Lydeard JR, Huttlin EL, Goessling W, Shah JV and Harper JW: Systematic proteomics of the VCP-UBXD adaptor network identifies a role for UBXN10 in regulating ciliogenesis. Nat Cell Biol. 17:1356–1369. 2015. View Article : Google Scholar : PubMed/NCBI

19 

Schrul B and Kopito RR: Peroxin-dependent targeting of a lipid-droplet-destined membrane protein to ER subdomains. Nat Cell Biol. 18:740–751. 2016. View Article : Google Scholar : PubMed/NCBI

20 

Pawar A, Botolin D, Mangelsdorf DJ and Jump DB: The role of liver X receptor-alpha in the fatty acid regulation of hepatic gene expression. J Biol Chem. 278:40736–40743. 2003. View Article : Google Scholar : PubMed/NCBI

21 

Horton JD, Shah NA, Warrington JA, Anderson NN, Park SW, Brown MS and Goldstein JL: Combined analysis of oligonucleotide microarray data from transgenic and knockout mice identifies direct SREBP target genes. Proc Natl Acad Sci USA. 100:12027–12032. 2003. View Article : Google Scholar : PubMed/NCBI

22 

Gong Yi, Lee JN, Lee PC, Goldstein JL, Brown MS and Ye J: Sterol-regulated ubiquitination and degradation of Insig-1 creates a convergent mechanism for feedback control of cholesterol synthesis and uptake. Cell Metab. 3:15–24. 2006. View Article : Google Scholar : PubMed/NCBI

23 

Lee JN, Zhang X, Feramisco JD, Gong Y and Ye J: Unsaturated fatty acids inhibit proteasomal degradation of Insig-1 at a postubiquitination step. J Biol Chem. 283:33772–33783. 2008. View Article : Google Scholar : PubMed/NCBI

24 

Nohturfft A, Yabe D, Goldstein JL, Brown MS and Espenshade PJ: Regulated step in cholesterol feedback localized to budding of SCAP from ER membranes. Cell. 102:315–323. 2000. View Article : Google Scholar : PubMed/NCBI

25 

Ohsaki Y, Cheng J, Suzuki M, Fujita A and Fujimoto T: Lipid droplets are arrested in the ER membrane by tight binding of lipidated apolipoprotein B-100. J Cell Sci. 121:2415–2422. 2008. View Article : Google Scholar : PubMed/NCBI

26 

Sasako T, Ohsugi M, Kubota N, Itoh S, Okazaki Y, Terai A, Kubota T, Yamashita S, Nakatsukasa K, Kamura T, et al: Hepatic Sdf2l1 controls feeding-induced ER stress and regulates metabolism. Nat Commun. 10:9472019. View Article : Google Scholar : PubMed/NCBI

27 

Yen CL, Monetti M, Burri BJ and Farese RV Jr: The triacylglycerol synthesis enzyme DGAT1 also catalyzes the synthesis of diacylglycerols, waxes, and retinyl esters. J Lipid Res. 46:1502–1511. 2005. View Article : Google Scholar : PubMed/NCBI

28 

Loregger A, Raaben M, Tan J, Scheij S, Moeton M, van den Berg M, Gelberg-Etel H, Stickel E, Roitelman J, Brummelkamp T and Zelcer N: Haploid mammalian genetic screen identifies UBXD8 as a key determinant of HMGCR degradation and cholesterol biosynthesis. Arterioscler Thromb Vasc Biol. 37:2064–2074. 2017. View Article : Google Scholar : PubMed/NCBI

29 

Ishikawa H, Ma Z and Barber GN: STING regulates intracellular DNA-mediated, type I interferon-dependent innate immunity. Nature. 461:788–792. 2009. View Article : Google Scholar : PubMed/NCBI

30 

Yang L, Wang L, Ketkar H, Ma J, Yang G, Cui S, Geng T, Mordue DG, Fujimoto T, Cheng G, et al: UBXN3B positively regulates STING-mediated antiviral immune responses. Nat Commun. 9:23292018. View Article : Google Scholar : PubMed/NCBI

31 

Preuss C, Jelenik T, Bódis K, Müssig K, Burkart V, Szendroedi J, Roden M and Markgraf DF: A new targeted lipidomics approach reveals lipid droplets in liver, muscle and heart as a repository for diacylglycerol and ceramide species in non-alcoholic fatty liver. Cells. 8:2772019. View Article : Google Scholar : PubMed/NCBI

32 

Tian Y, Yang B, Qiu W, Hao Y, Zhang Z, Yang B, Li N, Cheng S, Lin Z, Rui YC, et al: ER-residential Nogo-B accelerates NAFLD-associated HCC mediated by metabolic reprogramming of oxLDL lipophagy. Nat Commun. 10:33912019. View Article : Google Scholar : PubMed/NCBI

33 

Trefts E, Gannon M and Wasserman DH: The liver. Curr Biol. 27:R1147–R1151. 2017. View Article : Google Scholar : PubMed/NCBI

34 

Lonardo A, Ballestri S, Marchesini G, Angulo P and Loria P: Nonalcoholic fatty liver disease: A precursor of the metabolic syndrome. Dig Liver Dis. 47:181–190. 2015. View Article : Google Scholar : PubMed/NCBI

35 

Guri Y, Colombi M, Dazert E, Hindupur SK, Roszik J, Moes S, Jenoe P, Heim MH, Riezman I, Riezman H and Hall MN: mTORC2 promotes tumorigenesis via lipid synthesis. Cancer Cell. 32:807–823.e12. 2017. View Article : Google Scholar : PubMed/NCBI

36 

Huang X, Fan M and Huang W: Pleiotropic roles of FXR in liver and colorectal cancers. Mol Cell Endocrinol. 543:1115432022. View Article : Google Scholar : PubMed/NCBI

37 

Yang S, Koteish A, Lin H, Huang J, Roskams T, Dawson V and Diehl AM: Oval cells compensate for damage and replicative senescence of mature hepatocytes in mice with fatty liver disease. Hepatology. 39:403–411. 2004. View Article : Google Scholar : PubMed/NCBI

38 

Zhang T, Zhang Y, Liu J, Ma Y, Ye Q, Yan X and Ding L: MicroRNA-377-3p inhibits hepatocellular carcinoma growth and metastasis through negative regulation of CPT1C-mediated fatty acid oxidation. Cancer Metab. 10:22022. View Article : Google Scholar : PubMed/NCBI

39 

Bauer DE, Hatzivassiliou G, Zhao F, Andreadis C and Thompson CB: ATP citrate lyase is an important component of cell growth and transformation. Oncogene. 24:6314–6322. 2005. View Article : Google Scholar : PubMed/NCBI

40 

Litwack G: Chapter 9-lipids. Litwack G: Human biochemistry Boston: Academic Press; pp. 199–255. 2018

41 

Medes G, Thomas A and Wernhouse S: Metabolism of neoplastic tissue. IV. A study of lipid synthesis in neoplastic tissue slices in vitro. Cancer Res. 13:27–29. 1953.PubMed/NCBI

42 

Fullerton MD, Galic S, Marcinko K, Sikkema S, Pulinilkunnil T, Chen ZP, O'Neill HM, Ford RJ, Palanivel R, O'Brien M, et al: Single phosphorylation sites in Acc1 and Acc2 regulate lipid homeostasis and the insulin-sensitizing effects of metformin. Nat Med. 19:1649–1654. 2013. View Article : Google Scholar : PubMed/NCBI

43 

Lake AD, Novak P, Hardwick RN, Flores-Keown B, Zhao F, Klimecki WT and Cherrington NJ: The adaptive endoplasmic reticulum stress response to lipotoxicity in progressive human nonalcoholic fatty liver disease. Toxicol Sci. 137:26–35. 2014.Oyadomari S, Harding HP, Zhang Y, Oyadomari M and Ron D: Dephosphorylation of translation initiation factor 2alpha enhances glucose tolerance and attenuates hepatosteatosis in mice. Cell Metab. 7:520–532. 2008. View Article : Google Scholar : PubMed/NCBI

44 

Pinto Lde F, Compri CM, Fornari JV, Bartchewsky W, Cintra DE, Trevisan M, Carvalho Pde O, Ribeiro ML, Velloso LA, Saad MJ, et al: The immunosuppressant drug, thalidomide, improves hepatic alterations induced by a high-fat diet in mice. Liver Int. 30:603–610. 2010. View Article : Google Scholar : PubMed/NCBI

45 

Crespo J, Cayón A, Fernández-Gil P, Hernández-Guerra M, Mayorga M, Domínguez-Díez A, Fernández-Escalante JC and Pons-Romero F: Gene expression of tumor necrosis factor alpha and TNF-receptors, p55 and p75, in nonalcoholic steatohepatitis patients. Hepatology. 34:1158–1163. 2001. View Article : Google Scholar : PubMed/NCBI

46 

Zhou YJ, Li YY, Nie YQ, Yang H, Zhan Q, Huang J, Shi SL, Lai XB and Huang HL: Influence of polygenetic polymorphisms on the susceptibility to non-alcoholic fatty liver disease of Chinese people. J Gastroenterol Hepatol. 25:772–777. 2010. View Article : Google Scholar : PubMed/NCBI

47 

Kramer F, Torzewski J, Kamenz J, Veit K, Hombach V, Dedio J and Ivashchenko Y: Interleukin-1beta stimulates acute phase response and C-reactive protein synthesis by inducing an NFkappaB- and C/EBPbeta-dependent autocrine interleukin-6 loop. Mol Immunol. 45:2678–2689. 2008. View Article : Google Scholar : PubMed/NCBI

48 

Banroques J, Doère M, Dreyfus M, Linder P and Tanner NK: Motif III in superfamily 2 ‘helicases’ helps convert the binding energy of ATP into a high-affinity RNA binding site in the yeast DEAD-box protein Ded1. J Mol Biol. 396:949–966. 2010. View Article : Google Scholar : PubMed/NCBI

49 

Catalá M, Antón A and Portolés MT: Characterization of the simultaneous binding of Escherichia coli endotoxin to Kupffer and endothelial liver cells by flow cytometry. Cytometry. 36:123–130. 1999. View Article : Google Scholar : PubMed/NCBI

50 

Luo W, Xu Q, Wang Q, Wu H and Hua J: Effect of modulation of PPAR-γ activity on Kupffer cells M1/M2 polarization in the development of non-alcoholic fatty liver disease. Sci Rep. 7:446122017. View Article : Google Scholar : PubMed/NCBI

51 

Liu PS, Wang H, Li X, Chao T, Teav T, Christen S, Di Conza G, Cheng WC, Chou CH, Vavakova M, et al: α-ketoglutarate orchestrates macrophage activation through metabolic and epigenetic reprogramming. Nat Immunol. 18:985–994. 2017. View Article : Google Scholar : PubMed/NCBI

52 

Bertola A, Bonnafous S, Anty R, Patouraux S, Saint-Paul MC, Iannelli A, Gugenheim J, Barr J, Mato JM, Le Marchand-Brustel Y, et al: Hepatic expression patterns of inflammatory and immune response genes associated with obesity and NASH in morbidly obese patients. PLoS One. 5:e135772010. View Article : Google Scholar : PubMed/NCBI

53 

Maina V, Sutti S, Locatelli I, Vidali M, Mombello C, Bozzola C and Albano E: Bias in macrophage activation pattern influences non-alcoholic steatohepatitis (NASH) in mice. Clin Sci (Lond). 122:545–553. 2012. View Article : Google Scholar : PubMed/NCBI

54 

Herber DL, Cao W, Nefedova Y, Novitskiy SV, Nagaraj S, Tyurin VA, Corzo A, Cho HI, Celis E, Lennox B, et al: Lipid accumulation and dendritic cell dysfunction in cancer. Nat Med. 16:880–886. 2010. View Article : Google Scholar : PubMed/NCBI

55 

Henning JR, Graffeo CS, Rehman A, Fallon NC, Zambirinis CP, Ochi A, Barilla R, Jamal M, Deutsch M, Greco S, et al: Dendritic cells limit fibroinflammatory injury in nonalcoholic steatohepatitis in mice. Hepatology. 58:589–602. 2013. View Article : Google Scholar : PubMed/NCBI

56 

Tosello-Trampont AC, Krueger P, Narayanan S, Landes SG, Leitinger N and Hahn YS: NKp46(+) natural killer cells attenuate metabolism-induced hepatic fibrosis by regulating macrophage activation in mice. Hepatology. 63:799–812. 2016. View Article : Google Scholar : PubMed/NCBI

57 

Ambade A, Satishchandran A, Saha B, Gyongyosi B, Lowe P, Kodys K, Catalano D and Szabo G: Hepatocellular carcinoma is accelerated by NASH involving M2 macrophage polarization mediated by hif-1αinduced IL-10. Oncoimmunology. 5:e12215572016. View Article : Google Scholar : PubMed/NCBI

58 

Wang W, Furneaux H, Cheng H, Caldwell MC, Hutter D, Liu Y, Holbrook N and Gorospe M: HuR regulates p21 mRNA stabilization by UV light. Mol Cell Biol. 20:760–769. 2000. View Article : Google Scholar : PubMed/NCBI

59 

Gerber AP, Herschlag D and Brown PO: Extensive association of functionally and cytotopically related mRNAs with Puf family RNA-binding proteins in yeast. PLoS Biol. 2:E792004. View Article : Google Scholar : PubMed/NCBI

60 

Clement SL, Scheckel C, Stoecklin G and Lykke-Andersen J: Phosphorylation of tristetraprolin by MK2 impairs AU-rich element mRNA decay by preventing deadenylase recruitment. Mol Cell Biol. 31:256–266. 2011. View Article : Google Scholar : PubMed/NCBI

61 

Lafarga V, Cuadrado A, Lopez de Silanes I, Bengoechea R, Fernandez-Capetillo O and Nebreda AR: p38 Mitogen-activated protein kinase- and HuR-dependent stabilization of p21(Cip1) mRNA mediates the G(1)/S checkpoint. Mol Cell Biol. 9:4341–4351. 2009. View Article : Google Scholar : PubMed/NCBI

62 

Meerang M, Ritz D, Paliwal S, Garajova Z, Bosshard M, Mailand N, Janscak P, Hübscher U, Meyer H and Ramadan K: The ubiquitin-selective segregase VCP/p97 orchestrates the response to DNA double-strand breaks. Nat Cell Biol. 13:1376–1382. 2011. View Article : Google Scholar : PubMed/NCBI

63 

Kim HH, Abdelmohsen K, Lal A, Pullmann R Jr, Yang X, Galban S, Srikantan S, Martindale JL, Blethrow J, Shokat KM and Gorospe M: Nuclear HuR accumulation through phosphorylation by Cdk1. Genes Dev. 22:1804–1815. 2008. View Article : Google Scholar : PubMed/NCBI

64 

Park ES, Yoo YJ and Elangovan M: The opposite role of two UBA-UBX containing proteins, p47 and SAKS1 in the degradation of a single ERAD substrate, α-TCR. Mol Cell Biochem. 425:37–45. 2017. View Article : Google Scholar : PubMed/NCBI

65 

Sepulveda D, Rojas-Rivera D, Rodríguez DA, Groenendyk J, Köhler A, Lebeaupin C, Ito S, Urra H, Carreras-Sureda A, Hazari Y, et al: Interactome screening identifies the ER luminal chaperone Hsp47 as a regulator of the unfolded protein response transducer IRE1α. Mol Cell. 69:238–252.e7. 2018. View Article : Google Scholar : PubMed/NCBI

66 

Karagöz GE, Acosta-Alvear D, Nguyen HT, Lee CP, Chu F and Walter P: An unfolded protein-induced conformational switch activates mammalian IRE1. Elife. 6:e307002017. View Article : Google Scholar : PubMed/NCBI

67 

Urano F, Wang X, Bertolotti A, Zhang Y, Chung P, Harding HP and Ron D: Coupling of stress in the ER to activation of JNK protein kinases by transmembrane protein kinase IRE1. Science. 287:664–666. 2000. View Article : Google Scholar : PubMed/NCBI

68 

Bertolotti A, Zhang Y, Hendershot LM, Harding HP and Ron D: Dynamic interaction of BiP and ER stress transducers in the unfolded-protein response. Nat Cell Biol. 2:326–332. 2000. View Article : Google Scholar : PubMed/NCBI

69 

Carrara M, Prischi F, Nowak PR and Ali MM: Crystal structures reveal transient PERK luminal domain tetramerization in endoplasmic reticulum stress signaling. EMBO J. 34:1589–1600. 2015. View Article : Google Scholar : PubMed/NCBI

70 

Harding HP, Zhang Y, Zeng H, Novoa I, Lu PD, Calfon M, Sadri N, Yun C, Popko B, Paules R, et al: An integrated stress response regulates amino acid metabolism and resistance to oxidative stress. Mol Cell. 11:619–633. 2003. View Article : Google Scholar : PubMed/NCBI

71 

Blais JD, Filipenko V, Bi M, Harding HP, Ron D, Koumenis C, Wouters BG and Bell JC: Activating transcription factor 4 is translationally regulated by hypoxic stress. Mol Cell Biol. 24:7469–7482. 2004. View Article : Google Scholar : PubMed/NCBI

72 

Adachi Y, Yamamoto K, Okada T, Yoshida H, Harada A and Mori K: ATF6 is a transcription factor specializing in the regulation of quality control proteins in the endoplasmic reticulum. Cell Struct Funct. 33:75–89. 2008. View Article : Google Scholar : PubMed/NCBI

73 

Schindler AJ and Schekman R: In vitro reconstitution of ER-stress induced ATF6 transport in COPII vesicles. Proc Natl Acad Sci USA. 106:17775–17780. 2009. View Article : Google Scholar : PubMed/NCBI

74 

Lally JSV, Ghoshal S, DePeralta DK, Moaven O, Wei L, Masia R, Erstad DJ, Fujiwara N, Leong V, Houde VP, et al: Inhibition of acetyl-CoA carboxylase by phosphorylation or the inhibitor ND-654 suppresses lipogenesis and hepatocellular carcinoma. Cell Metab. 29:174–182.e5. 2019. View Article : Google Scholar : PubMed/NCBI

75 

Zhu H, Zhang Q and Chen G: CXCR6 deficiency ameliorates ischemia-reperfusion injury by reducing the recruitment and cytokine production of hepatic NKT cells in a mouse model of non-alcoholic fatty liver disease. Int Immunopharmacol. 72:224–234. 2019. View Article : Google Scholar : PubMed/NCBI

76 

Zhou ZJ, Xin HY, Li J, Hu ZQ, Luo CB and Zhou SL: Intratumoral plasmacytoid dendritic cells as a poor prognostic factor for hepatocellular carcinoma following curative resection. Cancer Immunol Immunother. 68:1223–1233. 2019. View Article : Google Scholar : PubMed/NCBI

77 

Tang T, Sui Y, Lian M, Li Z and Hua J: Pro-inflammatory activated Kupffer cells by lipids induce hepatic NKT cells deficiency through activation-induced cell death. PLoS One. 8:e819492013. View Article : Google Scholar : PubMed/NCBI

78 

Wu L, Parekh VV, Gabriel CL, Bracy DP, Marks-Shulman PA, Tamboli RA, Kim S, Mendez-Fernandez YV, Besra GS, Lomenick JP, et al: Activation of invariant natural killer T cells by lipid excess promotes tissue inflammation, insulin resistance, and hepatic steatosis in obese mice. Proc Natl Acad Sci USA. 109:E1143–E1152. 2012. View Article : Google Scholar : PubMed/NCBI

79 

Ben Haij N, Planès R, Leghmari K, Serrero M, Delobel P, Izopet J, BenMohamed L and Bahraoui E: HIV-1 Tat protein induces production of proinflammatory cytokines by human dendritic cells and monocytes/macrophages through engagement of TLR4-MD2-CD14 complex and activation of NF-κB pathway. PLoS One. 10:e01294252015. View Article : Google Scholar : PubMed/NCBI

Related Articles

Journal Cover

March-2023
Volume 49 Issue 3

Print ISSN: 1021-335X
Online ISSN:1791-2431

Sign up for eToc alerts

Recommend to Library

Copy and paste a formatted citation
x
Spandidos Publications style
Guo Z and Guo Z: Role of ubiquitin regulatory X domain‑containing protein 3B in the development of hepatocellular carcinoma (Review). Oncol Rep 49: 57, 2023
APA
Guo, Z., & Guo, Z. (2023). Role of ubiquitin regulatory X domain‑containing protein 3B in the development of hepatocellular carcinoma (Review). Oncology Reports, 49, 57. https://doi.org/10.3892/or.2023.8494
MLA
Guo, Z., Liang, J."Role of ubiquitin regulatory X domain‑containing protein 3B in the development of hepatocellular carcinoma (Review)". Oncology Reports 49.3 (2023): 57.
Chicago
Guo, Z., Liang, J."Role of ubiquitin regulatory X domain‑containing protein 3B in the development of hepatocellular carcinoma (Review)". Oncology Reports 49, no. 3 (2023): 57. https://doi.org/10.3892/or.2023.8494