Next Article in Journal
Hypothalamic Astrocytes as a Specialized and Responsive Cell Population in Obesity
Next Article in Special Issue
Small-Molecule Inhibitors Targeting Proteasome-Associated Deubiquitinases
Previous Article in Journal
Proteomic Profiling of BRAFV600E Mutant Colon Cancer Cells Reveals the Involvement of Nucleophosmin/c-Myc Axis in Modulating the Response and Resistance to BRAF Inhibition by Vemurafenib
Previous Article in Special Issue
Ubiquitination and Deubiquitination in Oral Disease
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Regulation of Cancer Metabolism by Deubiquitinating Enzymes: The Warburg Effect

Department of Biomedical Science, CHA University, Seongnam-si 13488, Gyeonggi-do, Korea
*
Author to whom correspondence should be addressed.
Int. J. Mol. Sci. 2021, 22(12), 6173; https://doi.org/10.3390/ijms22126173
Submission received: 10 April 2021 / Revised: 31 May 2021 / Accepted: 5 June 2021 / Published: 8 June 2021

Abstract

:
Cancer is a disorder of cell growth and proliferation, characterized by different metabolic pathways within normal cells. The Warburg effect is a major metabolic process in cancer cells that affects the cellular responses, such as proliferation and apoptosis. Various signaling factors down/upregulate factors of the glycolysis pathway in cancer cells, and these signaling factors are ubiquitinated/deubiquitinated via the ubiquitin–proteasome system (UPS). Depending on the target protein, DUBs act as both an oncoprotein and a tumor suppressor. Since the degradation of tumor suppressors and stabilization of oncoproteins by either negative regulation by E3 ligases or positive regulation of DUBs, respectively, promote tumorigenesis, it is necessary to suppress these DUBs by applying appropriate inhibitors or small molecules. Therefore, we propose that the DUBs and their inhibitors related to the Warburg effect are potential anticancer targets.

1. Introduction

Cellular respiration is the process by which living organisms decompose organic matter to inorganic matter for producing energy required for survival. Sequentially, this encompasses the glycolysis process, the tricarboxylic acid cycle (TCA cycle, also called Krebs cycle), and the electron transport system [1]. In normal cells, glucose is converted to pyruvate through glycolysis in the cytoplasm, and the pyruvate enters the mitochondria where it is completely degraded [2]. This process requires oxygen, and 38 ATPs are finally produced from one molecule of glucose: 2 ATPs during glycolysis, 2 ATPs in the TCA cycle, and 34 ATPs in the electron transport system [3]. Under hypoxic conditions, the pyruvate is converted to lactic acid, which accumulates instead of entering the TCA cycle. In cancer cells, there is an abnormal progression of metabolism that only utilizes glycolysis. In normal cells, 38 ATPs are generated with one glucose molecule, whereas only 2 ATPs are generated in cancer cells (Figure 1) [4]. Consequently, the cancer cells require more glucose molecules than normal cells to obtain enough energy to survive. This is a remarkable characteristic of cancer cells and has recently been applied as a method of detecting cancer by exploiting the characteristic of excessive glucose utilization by cancer cells [5]. It is essential to consider that cancer cells complete the glycolysis process regardless of absence or presence of oxygen [6]. When cancer cells only obtain glycolysis-dependent energy even in the presence of oxygen, the effect is called the “Warburg effect” [7].
Homeostasis, which is a property of maintaining a constant state in response to various stimuli in an individual or a cell, is an essential factor. However, the imbalance of homeostasis leads to various diseases, including cancer. Therefore, it is important to maintain an optimized state by restoring an equilibrium state broken by changes in the surrounding environment. The degradation and synthesis of proteins in cells are an example of maintaining homeostasis. Proteins that need to be discarded after their half-life, and unstable proteins due to damage, are degraded. Here, ubiquitin serves a marker for labeling proteins that need to be degraded. Ubiquitin is finally covalently bound to the target protein through a series of processes using the E1, E2, and E3 enzymes [8]. Ubiquitin molecules attached to the substrate form a polyubiquitin chain, regulate the activity and function of the substrate protein, and induce degradation through the 26S proteasome [9]. The E3 ligase plays a role in attaching ubiquitin to the target protein, whereas the deubiquitinating enzyme (DUB) induces a reversible reaction that breaks the bond between the target protein and ubiquitin or between the ubiquitin [10]. The ubiquitinated proteins mediated by an E3 ligase are degraded by the 26S proteasome, thereby reducing the cellular functions of the proteins. Alternatively, DUBs stabilize the substrate proteins and improve their cellular functions by modulating the degradation [11]. Thus, degradation and expression of proteins by the ubiquitin–proteasome system (UPS) play an important role in cell homeostasis [12] and can be used as an anticancer drug to remove oncoproteins or stabilize tumor suppressor proteins through UPS [13].

2. Cancer Metabolism Involved in the Warburg Effect

2.1. Changes in the Tumor Microenvironment

Unlike normal cells, cancer cells require considerable energy to replicate due to their inherent characteristic of abnormally rapid proliferation [14]. The tumor microenvironment (TME), such as hypoxia, results in metabolic changes, including the Warburg effect, in cancer cells. Alterations of the tumor microenvironment due to HIF initiate transcription programs under hypoxic stress conditions [15]. HIF is a transcription factor that regulates angiogenesis, and heterodimers include HIF-1, HIF-2, and HIF-3. HIF-1α is expressed in all cells, whereas HIF-2α is expressed in endothelial cells or hepatocytes and controls angiogenesis and red blood cell production [16]. The level of hypoxia-induced neovascularization varies and is dependent on the expression levels of ANGPT-2 and VEGF-A [17]. In non-small-cell lung cancer cells, inhibition of the HIF-1α/VEGF signaling pathway by GLA inhibits the hypoxia-induced cell invasion and proliferation [18]. The representative target of pVHL is HIF. pVHL binds to elongin C and CUL2, thus forming the VCB–CUL2 complex with elongin B. Under normal oxygen conditions, acetylation and hydroxylation of proline residues in the oxygen-dependent degradation domain (ODD) of HIF-α induces binding to the VCB–CUL2 complex and functions as an anti-tumor when HIF-1α is degraded via the ubiquitin–proteasome pathway [19]. Under hypoxia, the HIF-1α is not degraded, accumulates in the cells, and subsequently interacts with HIF-β. Blood supply to all cancer cells is restricted with tumor progression, and the tumor microenvironment stabilizes HIF-1α and HIF-2α [20,21], which are therefore found to be upregulated in tumors; however, this does not apply to all cancer cell types. In lung adenocarcinoma, HIF-1α is upregulated, but HIF-2α is downregulated [22].

2.2. Changes in the Signaling Pathways

AKT is an important signaling molecule that induces the Warburg effect in cancer cells. PI3K phosphorylates and activates AKT located on the cell membrane [23]. The phosphorylated AKT separates from the cell membrane, causing intracellular reactions with the mTORC-containing substrates. AKT promotes lipid biosynthesis and glucose transport to cancer cells with improved glycolysis through activation of mTOR [24]. The PI3K/AKT signaling pathway promotes the breakdown and uptake of glucose and lactic acid production, thus playing an important role in the metabolic reprogramming of cancer cells [25]. It also regulates the growth, proliferation, and apoptosis of cancer cells by stabilizing, inhibiting, and regulating downstream factors [26]. Growth factors regulating HIF-1α activate the PI3K/AKT signaling pathway [27]. Conversely, hindrance of the AKT signaling pathway inhibits glycolysis and prevents cell growth. CTMP inhibits AKT phosphorylation with subsequent inhibition of the PI3K/AKT pathway [28], and FDFT1 negatively regulates the AKT/mTOR signal to inhibit glycolysis and proliferation in colon cancer [29]. Moreover, PTEN also acts as a negative regulator of the PI3K/AKT pathway [30].

2.3. Change from Oxidative Metabolism to Reduced Metabolism

NADH is an important factor involved in altering the redox metabolism of cells and is used by the mitochondria for electron transport. NAD+ is regenerated from NADH by a redox-linked mitochondrial shuttle, of which malate-aspartate is the most commonly recognized shuttle [31]. During glycolysis, a malate-aspartate shuttle through the mitochondria restores the NADH imbalance. However, when transport through the intracellular membrane attains the maximum rate, the shuttle exceeds the range that accommodates glycolysis and subsequently converts pyruvate to lactic acid via LDH to produce NAD+ [32]. The conversion of NADH to NAD+ in the cytoplasm satisfies the redox imbalance occurring in the glycolysis process, and the mitochondrial NADH produced enters the electron transport system and eventually produces more ATPs [33]. Thus, the balance of NADH redox contributes to a direct signaling role for the Warburg effect [34]. Inhibition of lactate dehydrogenase A (LDH-A) suppresses the Warburg effect and returns to oxidative phosphorylation to re-oxidize NADH and produce ATP [35].

2.4. Consumption of Glutamine

The Warburg effect is a major metabolic characteristic of cancer. Proliferating cells require not only ATP but also other cellular components. Along with glucose, glutamine is one of the most abundant nutrients in plasma. Similar to glucose, glutamine is degraded to lactic acid and does not undergo complete oxidative phosphorylation in mammalian cells. Glutamine is also utilized in biosynthetic pathways that provide a secondary carbon source for fatty acid synthesis. Glutamine-derived α-ketoglutarate is supplied for the production of citrate through forward flux via the TCA cycle and malic enzyme-dependent production of pyruvate [36].

2.5. Lipid Biosynthesis

Lipids are partially derived from acetyl CoA and many contain fatty acids (FAs), which are essential for cancer cell proliferation. Adipocytes provide FAs for rapid tumor growth. FAs are considered a major source of electrons for ATP production and are used to produce energy in cancer cells [37]. In addition, mitochondrial fatty acid oxidation (β-oxidation) produces flavin adenine dinucleotide (FADH2) and nicotinamide adenine dinucleotide (NADH) via oxidative phosphorylation (OxPhos). A recent study found that a lack of glucose in cancer cells does not reduce ATP levels; however, blocking fatty acid oxidation (FAO) showed no effect in normal cells but reduced ATP production in cancer cells by 40% [38]. Further studies are required to determine which pathways among glucose, glutamine, and fatty acid metabolism are primarily used by cancer cells to produce ATP. In addition, cancer cells are known to improve biosynthetic ability by expressing pyruvate kinase isozymes M2 (PKM2) [39].

2.6. Drug Resistance

The inflow of glucose into cells is promoted by 14 GLUT transporter groups. GLUT1 and GLUT3 have been extensively investigated for cancer metabolism [40,41]. GLUT1 is upregulated in malignant tumors, such as prostate cancer [40] and breast cancer [42]. Increased GLUT1 expression activates mTOR [43], and activated mTOR increases the expression of GLUT1 [44], thereby forming a positive feedback loop. LoVo cells resistant to doxorubicin (an anticancer drug) showed a higher dependence on glucose metabolism and expression of GLUT1 and MCT4 for survival as compared to doxorubicin-sensitive LoVo cells. Silybin, an inhibitor of GLUT, is reported to decrease the expression of the GLUT protein. Exposure to 50 μM silybin resulted in decreased expression of GLUT1 only in the doxorubicin-resistant cells; however, exposure to 10 μM silybin affected both doxorubicin-sensitive and -resistant cells [45]. This result indicates that upregulation of GLUT is a target of resistance to drugs, such as doxorubicin. GLUT3 is highly expressed in brain tumor cells [46], and long-term treatment with temozolomide in human astrocytes resulted in increased expression of GLUT3, indicating that GLUT3 has acquired resistance to the temozolomide drug [47].

3. Ubiquitin–Proteasome System and DUBs

Ubiquitin is a small protein expressed in all eukaryotes, is composed of 76 amino acids, and is covalently bound to substrates. Ubiquitination is a post-translational modification that has various roles in cellular processes, including cell cycle, DNA repair, and signal transduction [48]. Ubiquitination is a multistep enzyme cascade that functions via the E1 (ubiquitin-activating enzyme), E2 (ubiquitin-conjugating enzyme), and E3 ligase (ubiquitin ligase) [49]. The glycine at the ubiquitin end first links to the cysteine residue of the E1 enzyme. ATP is required for this process, and the activated E1 enzyme transfers the linked ubiquitin to the E2 enzyme, which then binds to the E3 enzyme and transmits ubiquitin to the target protein [50]. The polyubiquitin chain formed by repeating this process leads the target protein to the 26S proteasome and eventual degradation of substrates [51]. This process is defined as the ubiquitin–proteasome system (UPS). The E3 enzyme is also called the E3 ligase, which links the ubiquitin to a target protein in UPS; other enzymes that function opposite to E3 ligase are called the deubiquitinating enzymes (DUBs) (Figure 2) [12]. Ubiquitin has the capability to form a polyubiquitin chain at 7 Lys sites and 1 Met site: K6, K11, K27, K29, K33, K48, K63, and M1. It is well known that the K6-linked polyubiquitin responds to mitophagy and DNA repair [52]. K11-linked polyubiquitin is known to control the cell cycle, proteasomal degradation, protein stability, mitophagy, trafficking, and endoplasmic reticulum-associated protein degradation [52,53,54]. K27-linked polyubiquitin activates kinases and regulates DNA repair [54]; K29-linked polyubiquitin plays a role in kinase modification and proteasomal/lysosomal degradation [55]; and K33-linked polyubiquitin induces kinases modification, innate immunity, and autophagy [54,56]. It is also known that K48-linked polyubiquitin induces proteasomal degradation [57], and the K63-linked polyubiquitin plays a role in protein kinase activation and DNA damage [58]. Lastly, M1-linked polyubiquitin activates gene expression and innate immunity [52].
DUBs are involved in two major roles. The first is to break the bond between ubiquitin and a protein, so that the protein from which ubiquitin has been removed can continue to function in the cells. The second involves breaking the bond between ubiquitin and ubiquitin [59]. After the target protein enters the proteasome, the remaining ubiquitins are broken down one by one in the form of a chain, and free ubiquitin is recycled back to the UPS. DUBs are classified into nine subfamilies [60]: ubiquitin-specific protease (USP), ubiquitin C-terminal hydrolases protease (UCH), Machado–Joseph disease protein domain protease (MJD), ovarian tumor protease (OTU), Jab1/Pab1/MPN metallo-enzyme motif protease (JAMM), monocyte chemotactic protein-induced protease (MCPIP), permuted papain fold peptidase of dsDNA viruses and eukaryotes (PPPDE), motif interacting with Ub-containing novel DUB family (MINDY), and zinc finger with UFM1-specific peptidase domain protein (ZUFSP, also called ZUP1 or C60rf113) [61,62,63]. The USP, UCH, OTU, MJD, MCPIP, PPPDE, MINDY, and ZUFSP subfamilies have cysteine peptidase activity, whereas the JAMM subfamily contains zinc metalloisopeptidase activity [64,65].

4. DUBs of the Warburg Effect Factors

4.1. USP7

Ubiquitin-specific protease 7 (USP7), also called the herpesvirus-associated ubiquitin-specific protease (HAUSP), is a protein belonging to USP, the largest subfamily of DUB. It targets proteins associated with numerous tumors, such as p53 [66], MDM2 [67], PTEN [68], FOXO4 [69], and histone H2B [70], and is responsible for various biological functions, such as tumor suppression, DNA repair, immune response, and apoptosis [71].
SIRT7, a new target of USP7, plays a role in controlling a variety of cellular processes, ranging from cell homeostasis [72], senescence [73], and DNA repair to cancer progression [74]. It also acts to resist various types of stress, such as hypoxia [75], endoplasmic reticulum stress [76], and low glucose levels [77]. USP7 is a DUB of SIRT7 and decreases the K-63 linked ubiquitination level of SIRT7 in HCT116 cells. However, the binding affinity of USP7 and SIRT7 is found to decrease in glucose starvation, and the ubiquitin level of SIRT7 decreases with increasing glucose concentration. SIRT7 deubiquitination by USP7 inhibits the expression of G6PC, a key regulator of SIRT7-mediated glucose production [78].
FoxO1, a member of the FoxO subfamily of forkhead/winged helix transcription factors, is important for inhibition of the insulin-mediated glucose production [79]. FoxO1 interacts with the insulin responsive enzyme (IRE) in the promoter regions of G6PC and Pck1 [80]. Insulin signaling inhibits the transcriptional activity of FoxO1 through AKT-dependent phosphorylation of FoxO1 at certain conserved residues (T24, S256, and S319 of human FoxO1) [81]. USP7 deubiquitinates the FoxO1 and inhibits transcriptional activity in HEK293A cells. In addition, USP7 regulates FoxO1 occupancy in the promoter of the glucose-generating gene [82]. Thus, the activity of USP7 alleviates excessive glucose production in a hypoxic environment.

4.2. USP19

The ubiquitin-specific protease 19 (USP19) protein belongs to the USP subfamily. USP19 promotes tumor formation in gastric cancer by upregulating MMP2 and MMP9 involved in cancer migration and invasion [83] and regulates the growth of Ewing sarcoma by acting as a DUB of the chimeric transcription factor EWS-FLI1 [84]. USP19 not only regulates cancer cells but also performs various functions within cells, such as differentiation of muscle cells [85], viral immune response [86], autophagy [87], macrophage polarization [88], cell cycle regulation [89], chromosome stabilization, and repair of DNA damage [90].
HIF is a transcription activator having subunits HIF-α (HIF-1α, HIF-2α, and HIF-3α) and HIF-β, and mediates the response of cell proliferation/survival, angiogenesis, and glucose metabolism [91]. USP19 interacts with HIF-1α and stabilizes HIF-1α, regardless of the ER localization and catalytic activity of USP19 in HeLa cells. Knockdown of USP19 decreases the protein levels of HIF-1α and the mRNA expression levels of VEGF and GLUT1, which are target genes of HIF-1α in hypoxia [92].

4.3. USP28

The ubiquitin-specific protease 28 (USP28) protein belongs to the USP subfamily and USP28 responds to DNA damage and stabilizes proteins such as Myc and cyclin E [93]. In addition, USP28 is overexpressed in tumors of human colon cancer patients and acts as an oncogene that promotes the formation of intestinal tumors [94].
Fbw7, a substrate of USP28, is an E3 ligase that plays a role in Myc- or cyclin E-mediated cancer development [95]. Loss of Fbw7 in mice decelerates the cardiovascular development and increases embryonic mortality via an increase in the notch1 mRNA levels [96]. USP28 does not directly bind to c-Myc but deubiquitinates the c-Myc through interaction with Fbw7 in vivo and in vitro, the E3 ligase of c-Myc [97].

4.4. USP37

The ubiquitin-specific protease 37 (USP37) protein belongs to the USP subfamily. USP37 has a role in regulating the cell cycle and accelerates conversion of the G1/S phase. USP37 is also reported to be a transcriptional target of the oncogenic transcription factor E2F1, which is upregulated in several cancers [98]. Recent studies report that USP37 is a potential factor related to breast cancer progression [99].
The Myc oncogene, which contributes to the development of numerous human cancers, encodes the transcription factor c-Myc, thereby linking altered cellular metabolism with oncogenesis [100]. c-Myc plays an important role in promoting glycolysis in the Warburg effect [101] and cooperates with E2F1 in regulating the expression of genes involved in nucleotide metabolism. Along with HIF-1, it is reported to regulate the expression of genes involved in glucose metabolism [102,103]. The overexpression of c-Myc can be controlled by the UPS. c-Myc is deubiquitinated and stabilized by direct binding to USP37 in HEK293T cells. Moreover, depletion of USP37 results in decreased expressions of GLUT1 and LDHA mRNAs required for glucose uptake and lactic acid production in H1299 cells. This suggests that USP37 regulates the Warburg effect via stabilizing c-Myc in lung cancer [104]. Interestingly, unlike USP28 [97], USP37 directly binds to c-Myc and acts independently of Fbw37.

4.5. USP44

The ubiquitin-specific protease 44 (USP44) belongs to the USP family and is a DUB containing a zinc-finger domain and a USP domain. USP44 is involved in numerous cellular functions, including stem cell differentiation [105] and central body position regulation [106], and responses to DNA damage [107]. In addition, USP44 plays an important role in human tumors acting as both a tumor suppressor and an oncogene. It inhibits cell growth by inhibiting AKT signaling in non-small-cell lung cancer [108] and promotes the growth of prostate cancer by stabilizing the EZH2 protein [109].
Fructose-1, 6-bisphosphatase 1 (FBP1) is an enzyme that regulates glucose production and catalyzes the decomposition of fructose 1, 6-bisphosphate into fructose 6-phosphate and inorganic phosphate. FBP1 acts as a tumor suppressor in diverse malignancies, such as kidney cancer [110], breast cancer [111], lung cancer [112], and liver cancer [113]. Knockdown of USP44 improves glucose utilization and lactic acid production capacity by reducing only FBP1, and not HK2 or PKM2, in PANC-1 cells. Conversely, overexpression of USP44 increases the protein expression level of FBP1. This suggests that FBP1 mediates USP44 and plays an important role in glucose metabolism [114].

4.6. OTUB2

Ubiquitin aldehyde binding 2 (OTUB2) belongs to the OTU subfamily, which relies on DNA damage to fine-tune the ubiquitination level and supports the DNA repair pathway selection [115]. In addition, OTUB2 promotes cancer metastasis by activating YAP and TAZ, independent of the Hippo signaling pathway [116].
AKT, also known as protein kinase B (PKB), is a serine/threonine kinase that induces cell growth and differentiation. AKT is capable of phosphorylating various downstream factors related to apoptosis, transcription, and oncogene [117]. Phosphorylation-induced AKT activation, overexpression, and mutation are frequently observed in human cancers [118]. Knockdown of OTUB2 reduces glycolysis and its ability to absorb glucose or produce extracellular lactic acid in A549 and H1299 cells. In NSCLC cells, the expressions of HIF-1α and c-Myc are decreased. In addition, OTUB2 increases the phosphorylation levels of mTOR and AKT and protein expression levels of GLUT1, U2AF2, HK2, PGAM1, PGK1, HIF-1α, and c-Myc in XL-2 cells. This indicates that the mTOR/AKT signaling pathway can be activated by OTUB2 and suggests that OTUB2 is a regulator of the Warburg effect through interaction with U2AF2 [119].

4.7. OTUD6B

A double allele mutation in the ovarian tumor domain-containing 6B (OTUD6B) gene is associated with development of the intellectual disability syndrome [120]. Additionally, the long noncoding RNA OTUD6B-AS1 (lncRNA OTUD6B-AS1) of OTUD6B exacerbates oxidative damage in bladder cancer [121] and inhibits the growth, invasion, and migration of thyroid cancer [122]. The lncRNA OTUD6B-AS1 also regulates the Wnt/β-catenin signaling pathway to induce the proliferation and invasion of hepatocellular carcinoma [123]. However, it contrarily inhibits the proliferation of renal cell carcinoma [124].
The protein von Hippel–Lindau (pVHL) is an E3 ligase that plays a role as a tumor suppressor. pVHL is present on chromosome 3(3p25–26) and mutations cause von Hippel–Lindau disease, a genetic disease that is transmitted through generations [125]. In HCC cells with OTUD6B knockdown, the protein levels of HIF-1α and HIF-2α were increased in both the normal oxygen state as well as the hypoxic state. In addition, overexpression of OTUD6B resulted in increased ubiquitination of HIF-1α in MHCC-LM3 cells. However, OTUD6B did not bind to HIF-1α but instead directly bound to pVHL. OTUD6B binds to elongin B and enhances the interaction with pVHL-elongin C, thereby inhibiting pVHL from proteasome degradation in HEK293T cells. The stabilized pVHL recognizes and degrades hydroxylated or SUMOylated HIF-1α in hypoxia. As a result, OTUD6B increases the stability of pVHL to prevent metastasis of hepatocellular carcinoma [126,127].

4.8. OTUD7B

The OTU domain-containing protein 7B (OTUD7B), also called Cezanne, is a member of the OTU subfamily that contains the OTU domain. OTUD7B positively modulates and activates T cells and induces an inflammatory response [128]. Moreover, by inhibiting the NF-κB signaling pathway, OTUD7B negatively regulates and inhibits B cell activity [129]. OTUD7B not only responds to the immune system but also functions as an oncogene; OTUD7B induces lung squamous carcinoma via the AKT/VEGF signaling pathway [130] and activates the NF-κB signaling pathway to increase resistance to apoptosis in hepatocellular carcinoma [131].
Under hypoxia, HIF-1 upregulates the expression of Pdk1, an enzyme regulating glycolysis in cancer cells and VHL-deficient osteoblasts, and promotes the conversion of cytoplasmic pyruvate to lactic acid [132]. Knockdown of OTUD7B decreases the protein levels of HIF-1α in HeLa cells and mouse embryonic fibroblasts (MEFs) isolated from OTUD7B knockout mice (OTUD7B−/−) and increases the K11-linked polyubiquitin chain formed. However, it is confirmed that HIF-1α, which is degraded by OTUD7B, is regulated through a proteasome-independent process in OTUD7B knockdown cells (Table 1 and Figure 3) [133].

4.9. Other DUBs

There are DUBs excluded from this paper because they are not identified to be related to the Warburg effect, glycolysis, or cancer, yet. For example, USP1 limits PI3K–AKT–FoxO signaling by removing the K63-linked polyubiquitin chain from AKT in prolonged starvation mouse muscle and depletion of USP1 increases ubiquitination level of AKT, glucose uptake, and PI3K–AKT–FoxO signaling [134]. However, different results may occur when applied to humans or other diseases such as cancer, because DUBs function differently depending on the cell types. Another DUB that regulates AKT, UCH-L1, induces lymphoma by deregulating AKT signaling [135]. However, the effect of UCH-L1 on the Warburg effect or glycolysis is not known yet.
There are other DUBs excluded from this paper for the same reason as described. DUBs which affect c-Myc are CYLD [136], USP2a [137,138,139], USP7 [140,141], USP13 [142], USP16 [143], USP18 [144], USP22 [145,146], USP28 [147], USP36 [148,149], and OTUD6B isoform 2 [150]. DUBs that regulate FBP1 include USP22 [151] and USP44 [114]. The DUBs of HIF-1α are MCPIP1 [152], UCH-L1 [153,154,155], USP7 [156], USP8 [157], USP20 [158], and USP28 [158]. USP4 [159], USP5 [160], USP9X [161], USP10 [162], and OTUD5 [163] regulate mTOR signaling, and USP22 [164] and OTUB1 [165] regulate activity of mTOR.
These DUBs affect signaling pathways, but it is not known how they affect survival, apoptosis, the Warburg effect, or glycolysis in cancer cells. Therefore, follow-up studies related to metabolism in cancer cells and clinical samples are required.

5. Small Molecules of DUBs Associated with the Warburg Effect

Depending on the target protein, DUBs can play a role in tumor formation as well as suppression. For example, when a DUB acts on the tumor suppressor protein p53, the degradation of p53 is inhibited, and the tumor suppressor role is restored by stabilizing p53 [166,167]. However, if the DUB targets an oncoprotein such as c-Myc, it plays a role in tumorigenesis [97,104,119]. Therefore, inhibiting a DUB, which acts as an oncoprotein, may be another way to inhibit the Warburg effect and treat cancer. Table 2 reveals the inhibitors and small molecules of DUBs featured in this paper (Table 2). However, since inhibitors or small molecules of DUBs are yet to be developed, such DUBs are other potential targets for anticancer therapeutics.

5.1. USP7

FT671 and FT827 are small molecule inhibitors that specifically target USP7 and no other DUBs. FT671 is a non-covalent inhibitor, whereas FT827 is a covalent inhibitor, which binds and inhibits the catalytic domain of USP7. By inhibiting USP7, FT671 induces increased protein levels of p53 and p53 target proteins in HCT116 and U2OS cells and stabilization of the p53 protein and degradation of N-Myc in the IMR-32 cells [168].
GNE-6640 and GNE-6776 non-covalently target 12Å remote from the catalytic cysteine and not the catalytic domain of USP7. GNE-6640 and GNE-6776 interact with the side chain and acidic residues of ubiquitin K48. This means that USP7 binds and decomposes ubiquitin moieties having the side chain of K48 and, consequently, inhibits the activity of USP7 by weakening the binding with ubiquitin [169].
HBX 19,818, HBX 28,258, and HBX 41,108 are small molecule substances that inhibit the activity of USP7, where HBX 19,818 and HBX 28,258 inhibit HAUbVS binding to USP7. In particular, HBX 19,818 covalently binds to the catalytic cysteine C223 of USP7 and increases the deubiquitination level of the USP7-mediated MDM2 and functional activation of p53 [170]. In addition to the p53-dependent apoptosis, HBX 41,108 induces USP7-mediated p53 stabilization and activation. Unlike HBX 41,108, which non-specifically inhibits DUB, HBX 19,818 and HBX 28,258 target USP7 as specific targets [170,171].
P22077 is an inhibitor that jointly targets USP7, USP47, and USP10, and P50429 inhibits USP7 and USP47. P22077 and P50429 covalently modify the catalytic cysteine (C233) of USP7 and promote structural changes of enzymes related to rearrangement of the active site. In addition, P22077 causes a partial loss, but P50429 completely abolishes the link between Ub-vinyl methyl ester (Ub-VME) and USP7, which is an ubiquitin variant that binds to the catalytic cysteine of DUB [173]. The inhibition of USP7 by P22077 results in accumulation of the 26S proteasome complex and polyubiquitinated substances but does not directly block the proteasome proteolytic activity. In the early stage of P22077 treatment, there is a decrease in the USP7-mediated HDM2, with increased protein expressions of p53 and p21; however, in the later stages, there is a decrease in the protein expression level of HDM2 due to the p53/HDM2 feedback loop. In addition, P22077 decreases the expression levels of claspin and Chk1, resulting in a checkpoint arrest [172].
P5091 is a trisubstituted thiophene with dichlorophenylthio, nitro, and acetyl substituents and reacts specifically with USP7, but not with other DUBs. P5091 inhibits USP7 by binding to USP7 competitively with Ub-VME, and inhibiting the deubiquitinating activity of USP7, but not the proteasome [174]. In multiple myeloma (MM) cells treated with P5091, the level of ubiquitination of HDM2 and protein expression levels of HDM2 and HDMX are increased, and the protein levels of p53 and p21 are downregulated. Moreover, there is also a reduction in the angiogenic activity [175].
vif1 and vif2 are peptides derived from Kaposi’s sarcoma-associated herpesvirus (KSHV) vIRF4; vif1 binds competitively with the TRAF domain of USP7, which is known to bind to a substrate having P/A/E-x-x-S, whereas vif2 inhibits the deubiquitination activity of USP7 by binding both the TRAF domain and the catalytic domain of USP7. Treatment with vif1 and vif2 promotes anti-tumor effects via p53-mediated apoptosis and p21-mediated cell cycle arrest [176].
XL177A is an irreversible inhibitor that acts on the catalytic cysteine C223 of USP7. When treated with XL177A, the MCF7 cells expressing USP7 show reduced expression of HDM2 and upregulation in the expression levels of p53 and p21, CDKN1A and GADD45A (proteins that arrest the cell cycle), and Bax and DDB2 (apoptotic proteins) [177]. XL188 is a small molecule inhibitor, which was developed before XL177A, and is reported to inhibit noncovalent active sites. XL188 treatment induces downregulation of HDM2 and upregulation of the p53 and p21 proteins [178].

5.2. USP28

AZ1 is a double inhibitor targeting both USP25 and USP28. The fluorophenyl ring of AZ1 forms an arene–H interaction with Phe370, while the bromophenyl ring forms a π–π interaction with Phe370. The fluoro and OH groups form hydrogen bond interactions with the Glu366 and Lys381 residues. Treatment of AZ1 induces apoptosis by downregulating the expression level of the tumor protein c-Myc in cancer cell lines [179]. Binding of the [1,2,3]triazolo[4,5-d]pyrimidine to the catalytic domain of USP28 and attachment of the benzyl group to the triazole ring of [1,2,3]triazolo[4,5-d]pyrimidine are important for activity. [1,2,3]triazolo[4,5-d]pyrimidine inhibits the survival, proliferation, cell cycle, and migration of cancer cells and induces apoptosis through the degradation of c-Myc by the proteasome [180].

5.3. Broad Spectrum of DUB Inhibitors

Betulinic acid is a natural anticancer drug derived from birch, which induces apoptosis by inhibiting the proteasome reaction in various cancer cells. Betulinic acid induces apoptosis by activating NF-κB, a major mediator of the cellular stress response [181]. It also upregulates the Bax protein [182], downregulates the expression of VEGF [183], and induces cell cycle arrest [184]. Interestingly, normal tissues and cells are not affected by betulinic acid [185].
PR-619 targets a wide range of DUBs but not all cysteine proteases. PR-619 induces G2/M cell cycle arrest in esophageal squamous cell carcinomas cells (ESCCs) and inhibits cell growth by inhibiting cyclin B1 and stabilizing p21. It also induces ER stress and promotes apoptosis [186]. In fibroblasts, there is increased sensitivity to the TNF-related apoptosis ligand (TRAIL) that selectively targets cancer cells but not non-malignant normal cells [187].

6. Conclusions

From the discovery of Warburg in the 1920s till today of 2021, the Warburg effect has received considerable attention, and the number of publications related to cancer cell metabolism is rapidly increasing. Therefore, to treat malignant tumors, it is important to comprehend the metabolic changes in cancer cells.
Among the substances that cells metabolize during glycolysis (glutamine, FA, and glucose), the Warburg effect is associated with the process that catalyzes glucose. Normal cells go through three steps (glycolysis process, TCA cycle, and electron transport system) in a normoxic environment and generate 38 ATPs per glucose molecule. Under hypoxia, normal cells produce two ATPs per glucose molecule as well as lactic acid solely through the glycolysis process. However, since cancer cells prefer anaerobic glycolysis under both normoxia and hypoxia, they require more glucose to produce the same energy as normal cells. One method being applied to detect tumors is by using the difference in metabolism between normal cells and cancer cells, and metabolic anticancer drugs are also being studied along with cytotoxic chemotherapy, targeted anticancer drugs, and cancer immunotherapy.
In cancer cells, proteins related to glycolysis are either upregulated or downregulated, and these proteins can be stabilized by DUBs or prevented from degradation by E3 ligases. Therefore, if UPS can be applied to control the expression levels of proteins related to glycolysis, it can prevent cancer cells from obtaining energy and, thus, be used as a new anticancer therapy. However, the expression levels and cellular roles of DUBs vary depending on the cell type or their substrates. On the other hand, clinical results are insufficient compared to in vivo or in vitro experiments. Therefore, follow-up clinical studies are required.

Author Contributions

Manuscript writing, S.-H.K. and K.-H.B.; final approval of manuscript, K.-H.B. All authors have read and agreed to the published version of the manuscript.

Funding

The study was supported by Basic Science Research Program through the National Research Foundation of Korea (NRF) funded by the Ministry of Education (No. RF-2020R1I1A207500312).

Institutional Review Board Statement

Not applicable.

Informed Consent Statement

Not applicable.

Data Availability Statement

Not applicable.

Acknowledgments

We would like to thank the members of Baek’s laboratory for their critical comments on the manuscript.

Conflicts of Interest

The authors declare no conflict of interest.

References

  1. Jiang, B. Aerobic glycolysis and high level of lactate in cancer metabolism and microenvironment. Genes Dis. 2017, 4, 25–27. [Google Scholar] [CrossRef] [PubMed]
  2. Anderson, N.M.; Mucka, P.; Kern, J.G.; Feng, H. The emerging role and targetability of the TCA cycle in cancer metabolism. Protein Cell 2018, 9, 216–237. [Google Scholar] [CrossRef]
  3. Devic, S. Warburg effect-A consequence or the cause of carcinogenesis? J. Cancer 2016, 7, 817–822. [Google Scholar] [CrossRef] [Green Version]
  4. Vander Heiden, M.G.; Cantley, L.C.; Thompson, C.B. Understanding the Warburg effect: The metabolic requirements of cell proliferation. Science 2009, 324, 1029–1033. [Google Scholar] [CrossRef] [Green Version]
  5. Jadvar, H. PET of glucose metabolism and cellular proliferation in prostate cancer. J. Nucl. Med. 2016, 57, 25S–29S. [Google Scholar] [CrossRef] [Green Version]
  6. Vaupel, P.; Multhoff, G. Revisiting the Warburg effect: Historical dogma versus current understanding. J. Physiol. 2020, 599, 1745–1757. [Google Scholar] [CrossRef]
  7. Warburg, O. On the origin of cancer cells. Science 1956, 123, 309–314. [Google Scholar] [CrossRef]
  8. Pickart, C.M. Mechanisms underlying ubiquitination. Annu. Rev. Biochem. 2001, 70, 503–533. [Google Scholar] [CrossRef]
  9. Hershko, A.; Ciechanover, A. The ubiquitin system. Annu. Rev. Biochem. 1998, 67, 425–479. [Google Scholar] [CrossRef]
  10. Smit, J.J.; Sixma, T.K. RBR E3-ligases at work. EMBO Rep. 2014, 15, 142–154. [Google Scholar] [CrossRef] [Green Version]
  11. Reyes-Turcu, F.E.; Ventii, K.H.; Wilkinson, K.D. Regulation and cellular roles of ubiquitin-specific deubiquitinating enzymes. Annu. Rev. Biochem. 2009, 78, 363–397. [Google Scholar] [CrossRef] [Green Version]
  12. Komander, D.; Clague, M.J.; Urbe, S. Breaking the chains: Structure and function of the deubiquitinases. Nat. Rev. Mol. Cell. Biol. 2009, 10, 550–563. [Google Scholar] [CrossRef]
  13. Park, J.; Cho, J.; Song, E.J. Ubiquitin-proteasome system (UPS) as a target for anticancer treatment. Arch. Pharm. Res. 2020, 43, 1144–1161. [Google Scholar] [CrossRef] [PubMed]
  14. Hanahan, D.; Weinberg, R.A. Hallmarks of cancer: The next generation. Cell 2011, 144, 646–674. [Google Scholar] [CrossRef] [Green Version]
  15. Kaelin, W.G., Jr.; Ratcliffe, P.J. Oxygen sensing by metazoans: The central role of the HIF hydroxylase pathway. Mol. Cell 2008, 30, 393–402. [Google Scholar] [CrossRef] [PubMed]
  16. Gruber, M.; Hu, C.J.; Johnson, R.S.; Brown, E.J.; Keith, B.; Simon, M.C. Acute postnatal ablation of Hif-2alpha results in anemia. Proc. Natl. Acad. Sci. USA 2007, 104, 2301–2306. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  17. Wiesener, M.S.; Jurgensen, J.S.; Rosenberger, C.; Scholze, C.K.; Horstrup, J.H.; Warnecke, C.; Mandriota, S.; Bechmann, I.; Frei, U.A.; Pugh, C.W.; et al. Widespread hypoxia-inducible expression of HIF-2alpha in distinct cell populations of different organs. FASEB J. 2003, 17, 271–273. [Google Scholar] [CrossRef] [Green Version]
  18. Wang, Y.; Shi, J.; Gong, L. Gamma linolenic acid suppresses hypoxia-induced proliferation and invasion of non-small cell lung cancer cells by inhibition of HIF1alpha. Genes Genom. 2020, 42, 927–935. [Google Scholar] [CrossRef]
  19. Zhang, J.; Zhang, Q. VHL and hypoxia signaling: Beyond HIF in cancer. Biomedicines 2018, 6, 35. [Google Scholar] [CrossRef] [Green Version]
  20. Foster, J.G.; Wong, S.C.; Sharp, T.V. The hypoxic tumor microenvironment: Driving the tumorigenesis of non-small-cell lung cancer. Future Oncol. 2014, 10, 2659–2674. [Google Scholar] [CrossRef] [PubMed]
  21. Franovic, A.; Holterman, C.E.; Payette, J.; Lee, S. Human cancers converge at the HIF-2alpha oncogenic axis. Proc. Natl. Acad. Sci. USA 2009, 106, 21306–21311. [Google Scholar] [CrossRef] [Green Version]
  22. Gao, Z.J.; Wang, Y.; Yuan, W.D.; Yuan, J.Q.; Yuan, K. HIF-2alpha not HIF-1alpha overexpression confers poor prognosis in non-small cell lung cancer. Tumour Biol. 2017, 39, 1010428317709637. [Google Scholar] [CrossRef] [Green Version]
  23. King, D.; Yeomanson, D.; Bryant, H.E. PI3King the lock: Targeting the PI3K/Akt/mTOR pathway as a novel therapeutic strategy in neuroblastoma. J. Pediatr. Hematol. Oncol. 2015, 37, 245–251. [Google Scholar] [CrossRef]
  24. Reiter, R.J.; Sharma, R.; Rosales-Corral, S. Anti-warburg effect of melatonin: A proposed mechanism to explain its inhibition of multiple diseases. Int. J. Mol. Sci. 2021, 22, 764. [Google Scholar] [CrossRef]
  25. Polivka, J., Jr.; Janku, F. Molecular targets for cancer therapy in the PI3K/AKT/mTOR pathway. Pharmacol. Ther. 2014, 142, 164–175. [Google Scholar] [CrossRef]
  26. Sarbassov, D.D.; Guertin, D.A.; Ali, S.M.; Sabatini, D.M. Phosphorylation and regulation of Akt/PKB by the rictor-mTOR complex. Science 2005, 307, 1098–1101. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  27. Niu, G.; Briggs, J.; Deng, J.; Ma, Y.; Lee, H.; Kortylewski, M.; Kujawski, M.; Kay, H.; Cress, W.D.; Jove, R.; et al. Signal transducer and activator of transcription 3 is required for hypoxia-inducible factor-1alpha RNA expression in both tumor cells and tumor-associated myeloid cells. Mol. Cancer Res. 2008, 6, 1099–1105. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  28. Zhao, S.; Fu, J.; Liu, F.; Rastogi, R.; Zhang, J.; Zhao, Y. Small interfering RNA directed against CTMP reduces acute traumatic brain injury in a mouse model by activating Akt. Neurol. Res. 2014, 36, 483–490. [Google Scholar] [CrossRef]
  29. Weng, M.L.; Chen, W.K.; Chen, X.Y.; Lu, H.; Sun, Z.R.; Yu, Q.; Sun, P.F.; Xu, Y.J.; Zhu, M.M.; Jiang, N.; et al. Fasting inhibits aerobic glycolysis and proliferation in colorectal cancer via the Fdft1-mediated AKT/mTOR/HIF1alpha pathway suppression. Nat. Commun. 2020, 11, 1869. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  30. Li, A.; Qiu, M.; Zhou, H.; Wang, T.; Guo, W. PTEN, insulin resistance and cancer. Curr. Pharm. Des. 2017, 23, 3667–3676. [Google Scholar] [CrossRef] [PubMed]
  31. LaNoue, K.F.; Williamson, J.R. Interrelationships between malate-aspartate shuttle and citric acid cycle in rat heart mitochondria. Metabolism 1971, 20, 119–140. [Google Scholar] [CrossRef]
  32. Kaplon, J.; Zheng, L.; Meissl, K.; Chaneton, B.; Selivanov, V.A.; Mackay, G.; van der Burg, S.H.; Verdegaal, E.M.; Cascante, M.; Shlomi, T.; et al. A key role for mitochondrial gatekeeper pyruvate dehydrogenase in oncogene-induced senescence. Nature 2013, 498, 109–112. [Google Scholar] [CrossRef] [PubMed]
  33. Locasale, J.W.; Cantley, L.C. Metabolic flux and the regulation of mammalian cell growth. Cell Metab. 2011, 14, 443–451. [Google Scholar] [CrossRef] [Green Version]
  34. Liberti, M.V.; Locasale, J.W. The warburg effect: How does it benefit cancer cells? Trends Biochem. Sci. 2016, 41, 211–218. [Google Scholar] [CrossRef] [Green Version]
  35. Fantin, V.R.; St-Pierre, J.; Leder, P. Attenuation of LDH-A expression uncovers a link between glycolysis, mitochondrial physiology, and tumor maintenance. Cancer Cell 2006, 9, 425–434. [Google Scholar] [CrossRef] [Green Version]
  36. DeBerardinis, R.J.; Mancuso, A.; Daikhin, E.; Nissim, I.; Yudkoff, M.; Wehrli, S.; Thompson, C.B. Beyond aerobic glycolysis: Transformed cells can engage in glutamine metabolism that exceeds the requirement for protein and nucleotide synthesis. Proc. Natl. Acad. Sci. USA 2007, 104, 19345–19350. [Google Scholar] [CrossRef] [Green Version]
  37. Nieman, K.M.; Kenny, H.A.; Penicka, C.V.; Ladanyi, A.; Buell-Gutbrod, R.; Zillhardt, M.R.; Romero, I.L.; Carey, M.S.; Mills, G.B.; Hotamisligil, G.S.; et al. Adipocytes promote ovarian cancer metastasis and provide energy for rapid tumor growth. Nat. Med. 2011, 17, 1498–1503. [Google Scholar] [CrossRef] [Green Version]
  38. Lee, J.S.; Oh, S.J.; Choi, H.J.; Kang, J.H.; Lee, S.H.; Ha, J.S.; Woo, S.M.; Jang, H.; Lee, H.; Kim, S.Y. ATP production relies on fatty acid oxidation rather than glycolysis in pancreatic ductal adenocarcinoma. Cancers 2020, 12, 2477. [Google Scholar] [CrossRef] [PubMed]
  39. Christofk, H.R.; Vander Heiden, M.G.; Harris, M.H.; Ramanathan, A.; Gerszten, R.E.; Wei, R.; Fleming, M.D.; Schreiber, S.L.; Cantley, L.C. The M2 splice isoform of pyruvate kinase is important for cancer metabolism and tumour growth. Nature 2008, 452, 230–233. [Google Scholar] [CrossRef]
  40. Xiao, H.; Wang, J.; Yan, W.; Cui, Y.; Chen, Z.; Gao, X.; Wen, X.; Chen, J. GLUT1 regulates cell glycolysis and proliferation in prostate cancer. Prostate 2018, 78, 86–94. [Google Scholar] [CrossRef]
  41. Dai, W.; Xu, Y.; Mo, S.; Li, Q.; Yu, J.; Wang, R.; Ma, Y.; Ni, Y.; Xiang, W.; Han, L.; et al. GLUT3 induced by AMPK/CREB1 axis is key for withstanding energy stress and augments the efficacy of current colorectal cancer therapies. Signal Transduct. Target Ther. 2020, 5, 177. [Google Scholar] [CrossRef] [PubMed]
  42. Deng, Y.; Zou, J.; Deng, T.; Liu, J. Clinicopathological and prognostic significance of GLUT1 in breast cancer: A meta-analysis. Medicine 2018, 97, e12961. [Google Scholar] [CrossRef] [PubMed]
  43. Buller, C.L.; Heilig, C.W.; Brosius, F.C., III. GLUT1 enhances mTOR activity independently of TSC2 and AMPK. Am. J. Physiol. Renal. Physiol. 2011, 301, F588–F596. [Google Scholar] [CrossRef] [PubMed]
  44. Buller, C.L.; Loberg, R.D.; Fan, M.H.; Zhu, Q.; Park, J.L.; Vesely, E.; Inoki, K.; Guan, K.L.; Brosius, F.C., III. A GSK-3/TSC2/mTOR pathway regulates glucose uptake and GLUT1 glucose transporter expression. Am. J. Physiol. Cell Physiol. 2008, 295, C836–C843. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  45. Catanzaro, D.; Gabbia, D.; Cocetta, V.; Biagi, M.; Ragazzi, E.; Montopoli, M.; Carrara, M. Silybin counteracts doxorubicin resistance by inhibiting GLUT1 expression. Fitoterapia 2018, 124, 42–48. [Google Scholar] [CrossRef]
  46. Flavahan, W.A.; Wu, Q.; Hitomi, M.; Rahim, N.; Kim, Y.; Sloan, A.E.; Weil, R.J.; Nakano, I.; Sarkaria, J.N.; Stringer, B.W.; et al. Brain tumor initiating cells adapt to restricted nutrition through preferential glucose uptake. Nat. Neurosci. 2013, 16, 1373–1382. [Google Scholar] [CrossRef]
  47. Le Calve, B.; Rynkowski, M.; Le Mercier, M.; Bruyere, C.; Lonez, C.; Gras, T.; Haibe-Kains, B.; Bontempi, G.; Decaestecker, C.; Ruysschaert, J.M.; et al. Long-term in vitro treatment of human glioblastoma cells with temozolomide increases resistance in vivo through up-regulation of GLUT transporter and aldo-keto reductase enzyme AKR1C expression. Neoplasia 2010, 12, 727–739. [Google Scholar] [CrossRef]
  48. Song, L.; Luo, Z.Q. Post-translational regulation of ubiquitin signaling. J. Cell Biol. 2019, 218, 1776–1786. [Google Scholar] [CrossRef]
  49. Zheng, N.; Shabek, N. Ubiquitin ligases: Structure, function, and regulation. Annu. Rev. Biochem. 2017, 86, 129–157. [Google Scholar] [CrossRef]
  50. Finley, D. Recognition and processing of ubiquitin-protein conjugates by the proteasome. Annu. Rev. Biochem. 2009, 78, 477–513. [Google Scholar] [CrossRef] [Green Version]
  51. Coux, O.; Tanaka, K.; Goldberg, A.L. Structure and functions of the 20S and 26S proteasomes. Annu. Rev. Biochem. 1996, 65, 801–847. [Google Scholar] [CrossRef]
  52. Lafont, E.; Hartwig, T.; Walczak, H. Paving TRAIL’s path with ubiquitin. Trends Biochem. Sci. 2018, 43, 44–60. [Google Scholar] [CrossRef]
  53. Suresh, B.; Lee, J.; Kim, K.S.; Ramakrishna, S. The importance of ubiquitination and deubiquitination in cellular reprogramming. Stem. Cells Int. 2016, 2016, 6705927. [Google Scholar] [CrossRef] [Green Version]
  54. Suresh, B.; Lee, J.; Kim, H.; Ramakrishna, S. Regulation of pluripotency and differentiation by deubiquitinating enzymes. Cell Death Differ. 2016, 23, 1257–1264. [Google Scholar] [CrossRef]
  55. Zhou, B.; Zeng, L. Conventional and unconventional ubiquitination in plant immunity. Mol. Plant. Pathol. 2017, 18, 1313–1330. [Google Scholar] [CrossRef]
  56. Huang, Q.; Zhang, X. Emerging roles and research tools of atypical ubiquitination. Proteomics 2020, 20, e1900100. [Google Scholar] [CrossRef]
  57. Grice, G.L.; Nathan, J.A. The recognition of ubiquitinated proteins by the proteasome. Cell Mol. Life Sci. 2016, 73, 3497–3506. [Google Scholar] [CrossRef] [Green Version]
  58. Li, W.; Ye, Y. Polyubiquitin chains: Functions, structures, and mechanisms. Cell Mol. Life Sci. 2008, 65, 2397–2406. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  59. Clague, M.J.; Barsukov, I.; Coulson, J.M.; Liu, H.; Rigden, D.J.; Urbe, S. Deubiquitylases from genes to organism. Physiol. Rev. 2013, 93, 1289–1315. [Google Scholar] [CrossRef] [PubMed]
  60. Das, T.; Shin, S.C.; Song, E.J.; Kim, E.E. Regulation of deubiquitinating enzymes by post-translational modifications. Int. J. Mol. Sci. 2020, 21, 4028–4045. [Google Scholar] [CrossRef] [PubMed]
  61. Abdul Rehman, S.A.; Kristariyanto, Y.A.; Choi, S.Y.; Nkosi, P.J.; Weidlich, S.; Labib, K.; Hofmann, K.; Kulathu, Y. MINDY-1 is a member of an evolutionarily conserved and structurally distinct new family of deubiquitinating enzymes. Mol. Cell 2016, 63, 146–155. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  62. Haahr, P.; Borgermann, N.; Guo, X.; Typas, D.; Achuthankutty, D.; Hoffmann, S.; Shearer, R.; Sixma, T.K.; Mailand, N. ZUFSP deubiquitylates K63-linked polyubiquitin chains to promote genome stability. Mol. Cell 2018, 70, 165–174.e6. [Google Scholar] [CrossRef] [Green Version]
  63. Hermanns, T.; Pichlo, C.; Woiwode, I.; Klopffleisch, K.; Witting, K.F.; Ovaa, H.; Baumann, U.; Hofmann, K. A family of unconventional deubiquitinases with modular chain specificity determinants. Nat. Commun. 2018, 9, 799. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  64. Shrestha, R.K.; Ronau, J.A.; Davies, C.W.; Guenette, R.G.; Strieter, E.R.; Paul, L.N.; Das, C. Insights into the mechanism of deubiquitination by JAMM deubiquitinases from cocrystal structures of the enzyme with the substrate and product. Biochemistry 2014, 53, 3199–3217. [Google Scholar] [CrossRef]
  65. Kwasna, D.; Abdul Rehman, S.A.; Natarajan, J.; Matthews, S.; Madden, R.; De Cesare, V.; Weidlich, S.; Virdee, S.; Ahel, I.; Gibbs-Seymour, I.; et al. Discovery and characterization of ZUFSP/ZUP1, a distinct deubiquitinase class important for genome stability. Mol. Cell 2018, 70, 150–164.e6. [Google Scholar] [CrossRef] [Green Version]
  66. Kwon, S.K.; Saindane, M.; Baek, K.H. p53 stability is regulated by diverse deubiquitinating enzymes. Biochim. Biophys. Acta Rev. Cancer 2017, 1868, 404–411. [Google Scholar] [CrossRef] [PubMed]
  67. Sun, Y.; Cao, L.; Sheng, X.; Chen, J.; Zhou, Y.; Yang, C.; Deng, T.; Ma, H.; Feng, P.; Liu, J.; et al. WDR79 promotes the proliferation of non-small cell lung cancer cells via USP7-mediated regulation of the Mdm2-p53 pathway. Cell Death Dis. 2017, 8, e2743. [Google Scholar] [CrossRef] [Green Version]
  68. Noguera, N.I.; Song, M.S.; Divona, M.; Catalano, G.; Calvo, K.L.; Garcia, F.; Ottone, T.; Florenzano, F.; Faraoni, I.; Battistini, L.; et al. Nucleophosmin/B26 regulates PTEN through interaction with HAUSP in acute myeloid leukemia. Leukemia 2013, 27, 1037–1043. [Google Scholar] [CrossRef]
  69. Van der Horst, A.; de Vries-Smits, A.M.; Brenkman, A.B.; van Triest, M.H.; van den Broek, N.; Colland, F.; Maurice, M.M.; Burgering, B.M. FOXO4 transcriptional activity is regulated by monoubiquitination and USP7/HAUSP. Nat. Cell Biol. 2006, 8, 1064–1073. [Google Scholar] [CrossRef]
  70. Cole, A.J.; Clifton-Bligh, R.; Marsh, D.J. Histone H2B monoubiquitination: Roles to play in human malignancy. Endocr. Relat. Cancer 2015, 22, T19–T33. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  71. Park, J.J.; Lim, K.H.; Baek, K.H. Annexin-1 regulated by HAUSP is essential for UV-induced damage response. Cell Death Dis. 2015, 6, e1654. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  72. Korogi, W.; Yoshizawa, T.; Karim, M.F.; Tanoue, H.; Yugami, M.; Sobuz, S.U.; Hinoi, E.; Sato, Y.; Oike, Y.; Mizuta, H.; et al. SIRT7 is an important regulator of cartilage homeostasis and osteoarthritis development. Biochem. Biophys. Res. Commun. 2018, 496, 891–897. [Google Scholar] [CrossRef] [PubMed]
  73. Bi, S.; Liu, Z.; Wu, Z.; Wang, Z.; Liu, X.; Wang, S.; Ren, J.; Yao, Y.; Zhang, W.; Song, M.; et al. SIRT7 antagonizes human stem cell aging as a heterochromatin stabilizer. Protein Cell 2020, 11, 483–504. [Google Scholar] [CrossRef]
  74. Tang, M.; Li, Z.; Zhang, C.; Lu, X.; Tu, B.; Cao, Z.; Li, Y.; Chen, Y.; Jiang, L.; Wang, H.; et al. SIRT7-mediated ATM deacetylation is essential for its deactivation and DNA damage repair. Sci. Adv. 2019, 5, eaav1118. [Google Scholar] [CrossRef] [Green Version]
  75. Zhan, L.; Lei, S.; Li, W.; Zhang, Y.; Wang, H.; Shi, Y.; Tian, Y. Suppression of microRNA-142-5p attenuates hypoxia-induced apoptosis through targeting SIRT7. Biomed. Pharmacother. 2017, 94, 394–401. [Google Scholar] [CrossRef] [PubMed]
  76. Shin, J.; He, M.; Liu, Y.; Paredes, S.; Villanova, L.; Brown, K.; Qiu, X.; Nabavi, N.; Mohrin, M.; Wojnoonski, K.; et al. SIRT7 represses Myc activity to suppress ER stress and prevent fatty liver disease. Cell Rep. 2013, 5, 654–665. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  77. Yan, W.W.; Liang, Y.L.; Zhang, Q.X.; Wang, D.; Lei, M.Z.; Qu, J.; He, X.H.; Lei, Q.Y.; Wang, Y.P. Arginine methylation of SIRT7 couples glucose sensing with mitochondria biogenesis. EMBO Rep. 2018, 19, e46377. [Google Scholar] [CrossRef]
  78. Jiang, L.; Xiong, J.; Zhan, J.; Yuan, F.; Tang, M.; Zhang, C.; Cao, Z.; Chen, Y.; Lu, X.; Li, Y.; et al. Ubiquitin-specific peptidase 7 (USP7)-mediated deubiquitination of the histone deacetylase SIRT7 regulates gluconeogenesis. J. Biol. Chem. 2017, 292, 13296–13311. [Google Scholar] [CrossRef] [Green Version]
  79. Chiefari, E.; Arcidiacono, B.; Palmieri, C.; Corigliano, D.M.; Morittu, V.M.; Britti, D.; Armoni, M.; Foti, D.P.; Brunetti, A. Cross-talk among HMGA1 and FoxO1 in control of nuclear insulin signaling. Sci. Rep. 2018, 8, 8540. [Google Scholar] [CrossRef] [Green Version]
  80. Singh, B.K.; Sinha, R.A.; Zhou, J.; Xie, S.Y.; You, S.H.; Gauthier, K.; Yen, P.M. FoxO1 deacetylation regulates thyroid hormone-induced transcription of key hepatic gluconeogenic genes. J. Biol. Chem. 2013, 288, 30365–30372. [Google Scholar] [CrossRef] [Green Version]
  81. Saline, M.; Badertscher, L.; Wolter, M.; Lau, R.; Gunnarsson, A.; Jacso, T.; Norris, T.; Ottmann, C.; Snijder, A. AMPK and AKT protein kinases hierarchically phosphorylate the N-terminus of the FOXO1 transcription factor, modulating interactions with 14-3-3 proteins. J. Biol. Chem. 2019, 294, 13106–13116. [Google Scholar] [CrossRef]
  82. Hall, J.A.; Tabata, M.; Rodgers, J.T.; Puigserver, P. USP7 attenuates hepatic gluconeogenesis through modulation of FoxO1 gene promoter occupancy. Mol. Endocrinol. 2014, 28, 912–924. [Google Scholar] [CrossRef]
  83. Dong, Z.; Guo, S.; Wang, Y.; Zhang, J.; Luo, H.; Zheng, G.; Yang, D.; Zhang, T.; Yan, L.; Song, L.; et al. USP19 enhances MMP2/MMP9-mediated tumorigenesis in gastric cancer. OncoTargets Ther. 2020, 13, 8495–8510. [Google Scholar] [CrossRef] [PubMed]
  84. Gierisch, M.E.; Pedot, G.; Walser, F.; Lopez-Garcia, L.A.; Jaaks, P.; Niggli, F.K.; Schafer, B.W. USP19 deubiquitinates EWS-FLI1 to regulate Ewing sarcoma growth. Sci. Rep. 2019, 9, 951. [Google Scholar] [CrossRef] [PubMed]
  85. Wiles, B.; Miao, M.; Coyne, E.; Larose, L.; Cybulsky, A.V.; Wing, S.S. USP19 deubiquitinating enzyme inhibits muscle cell differentiation by suppressing unfolded-protein response signaling. Mol. Biol. Cell 2015, 26, 913–923. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  86. Gu, Z.; Shi, W.; Zhang, L.; Hu, Z.; Xu, C. USP19 suppresses cellular type I interferon signaling by targeting TRAF3 for deubiquitination. Future Microbiol. 2017, 12, 767–779. [Google Scholar] [CrossRef] [PubMed]
  87. Jin, S.; Tian, S.; Chen, Y.; Zhang, C.; Xie, W.; Xia, X.; Cui, J.; Wang, R.F. USP19 modulates autophagy and antiviral immune responses by deubiquitinating Beclin-1. EMBO J. 2016, 35, 866–880. [Google Scholar] [CrossRef] [Green Version]
  88. Liu, T.; Wang, L.; Liang, P.; Wang, X.; Liu, Y.; Cai, J.; She, Y.; Wang, D.; Wang, Z.; Guo, Z.; et al. USP19 suppresses inflammation and promotes M2-like macrophage polarization by manipulating NLRP3 function via autophagy. Cell Mol. Immunol. 2020, 4, 1–12. [Google Scholar] [CrossRef]
  89. Lu, Y.; Adegoke, O.A.; Nepveu, A.; Nakayama, K.I.; Bedard, N.; Cheng, D.; Peng, J.; Wing, S.S. USP19 deubiquitinating enzyme supports cell proliferation by stabilizing KPC1, a ubiquitin ligase for p27Kip1. Mol. Cell Biol. 2009, 29, 547–558. [Google Scholar] [CrossRef] [Green Version]
  90. Wu, M.; Tu, H.Q.; Chang, Y.; Tan, B.; Wang, G.; Zhou, J.; Wang, L.; Mu, R.; Zhang, W.N. USP19 deubiquitinates HDAC1/2 to regulate DNA damage repair and control chromosomal stability. Oncotarget 2017, 8, 2197–2208. [Google Scholar] [CrossRef] [Green Version]
  91. Ke, Q.; Costa, M. Hypoxia-inducible factor-1 (HIF-1). Mol. Pharmacol. 2006, 70, 1469–1480. [Google Scholar] [CrossRef]
  92. Altun, M.; Zhao, B.; Velasco, K.; Liu, H.; Hassink, G.; Paschke, J.; Pereira, T.; Lindsten, K. Ubiquitin-specific protease 19 (USP19) regulates hypoxia-inducible factor 1alpha (HIF-1alpha) during hypoxia. J. Biol. Chem. 2012, 287, 1962–1969. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  93. Zhang, D.; Zaugg, K.; Mak, T.W.; Elledge, S.J. A role for the deubiquitinating enzyme USP28 in control of the DNA-damage response. Cell 2006, 126, 529–542. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  94. Diefenbacher, M.E.; Popov, N.; Blake, S.M.; Schulein-Volk, C.; Nye, E.; Spencer-Dene, B.; Jaenicke, L.A.; Eilers, M.; Behrens, A. The deubiquitinase USP28 controls intestinal homeostasis and promotes colorectal cancer. J. Clin. Invest. 2014, 124, 3407–3418. [Google Scholar] [CrossRef] [Green Version]
  95. Diefenbacher, M.E.; Chakraborty, A.; Blake, S.M.; Mitter, R.; Popov, N.; Eilers, M.; Behrens, A. Usp28 counteracts Fbw7 in intestinal homeostasis and cancer. Cancer Res. 2015, 75, 1181–1186. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  96. Tetzlaff, M.T.; Yu, W.; Li, M.; Zhang, P.; Finegold, M.; Mahon, K.; Harper, J.W.; Schwartz, R.J.; Elledge, S.J. Defective cardiovascular development and elevated cyclin E and Notch proteins in mice lacking the Fbw7 F-box protein. Proc. Natl. Acad. Sci. USA 2004, 101, 3338–3345. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  97. Popov, N.; Wanzel, M.; Madiredjo, M.; Zhang, D.; Beijersbergen, R.; Bernards, R.; Moll, R.; Elledge, S.J.; Eilers, M. The ubiquitin-specific protease USP28 is required for Myc stability. Nat. Cell Biol. 2007, 9, 765–774. [Google Scholar] [CrossRef]
  98. Huang, X.; Summers, M.K.; Pham, V.; Lill, J.R.; Liu, J.; Lee, G.; Kirkpatrick, D.S.; Jackson, P.K.; Fang, G.; Dixit, V.M. Deubiquitinase USP37 is activated by CDK2 to antagonize APC(CDH1) and promote S phase entry. Mol. Cell 2011, 42, 511–523. [Google Scholar] [CrossRef]
  99. Qin, T.; Li, B.; Feng, X.; Fan, S.; Liu, L.; Liu, D.; Mao, J.; Lu, Y.; Yang, J.; Yu, X.; et al. Abnormally elevated USP37 expression in breast cancer stem cells regulates stemness, epithelial-mesenchymal transition and cisplatin sensitivity. J. Exp. Clin. Cancer Res. 2018, 37, 287. [Google Scholar] [CrossRef] [Green Version]
  100. Dang, C.V. Myc on the path to cancer. Cell 2012, 149, 22–35. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  101. Xiang, S.; Gu, H.; Jin, L.; Thorne, R.F.; Zhang, X.D.; Wu, M. LncRNA IDH1-AS1 links the functions of c-Myc and HIF1alpha via IDH1 to regulate the Warburg effect. Proc. Natl. Acad. Sci. USA 2018, 115, E1465–E1474. [Google Scholar] [CrossRef] [Green Version]
  102. Cairns, R.A.; Harris, I.S.; Mak, T.W. Regulation of cancer cell metabolism. Nat. Rev. Cancer 2011, 11, 85–95. [Google Scholar] [CrossRef] [Green Version]
  103. Dang, C.V.; Kim, J.W.; Gao, P.; Yustein, J. The interplay between Myc and HIF in cancer. Nat. Rev. Cancer 2008, 8, 51–56. [Google Scholar] [CrossRef]
  104. Pan, J.; Deng, Q.; Jiang, C.; Wang, X.; Niu, T.; Li, H.; Chen, T.; Jin, J.; Pan, W.; Cai, X.; et al. USP37 directly deubiquitinates and stabilizes c-Myc in lung cancer. Oncogene 2015, 34, 3957–3967. [Google Scholar] [CrossRef]
  105. Fuchs, G.; Shema, E.; Vesterman, R.; Kotler, E.; Wolchinsky, Z.; Wilder, S.; Golomb, L.; Pribluda, A.; Zhang, F.; Haj-Yahya, M.; et al. RNF20 and USP44 regulate stem cell differentiation by modulating H2B monoubiquitylation. Mol. Cell 2012, 46, 662–673. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  106. Zhang, Y.; Foreman, O.; Wigle, D.A.; Kosari, F.; Vasmatzis, G.; Salisbury, J.L.; van Deursen, J.; Galardy, P.J. USP44 regulates centrosome positioning to prevent aneuploidy and suppress tumorigenesis. J. Clin. Invest. 2012, 122, 4362–4374. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  107. Mosbech, A.; Lukas, C.; Bekker-Jensen, S.; Mailand, N. The deubiquitylating enzyme USP44 counteracts the DNA double-strand break response mediated by the RNF8 and RNF168 ubiquitin ligases. J. Biol. Chem. 2013, 288, 16579–16587. [Google Scholar] [CrossRef] [Green Version]
  108. Zhang, Y.K.; Tian, W.Z.; Zhang, R.S.; Zhang, Y.J.; Ma, H.T. Ubiquitin-specific protease 44 inhibits cell growth by suppressing AKT signaling in non-small cell lung cancer. Kaohsiung J. Med. Sci. 2019, 35, 535–541. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  109. Park, J.M.; Lee, J.E.; Park, C.M.; Kim, J.H. USP44 promotes the tumorigenesis of prostate cancer cells through EZH2 protein stabilization. Mol. Cells 2019, 42, 17–27. [Google Scholar] [PubMed]
  110. Li, B.; Qiu, B.; Lee, D.S.; Walton, Z.E.; Ochocki, J.D.; Mathew, L.K.; Mancuso, A.; Gade, T.P.; Keith, B.; Nissim, I.; et al. Fructose-1,6-bisphosphatase opposes renal carcinoma progression. Nature 2014, 513, 251–255. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  111. Dong, C.; Yuan, T.; Wu, Y.; Wang, Y.; Fan, T.W.; Miriyala, S.; Lin, Y.; Yao, J.; Shi, J.; Kang, T.; et al. Loss of FBP1 by Snail-mediated repression provides metabolic advantages in basal-like breast cancer. Cancer Cell 2013, 23, 316–331. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  112. Zhang, J.; Wang, J.; Xing, H.; Li, Q.; Zhao, Q.; Li, J. Down-regulation of FBP1 by ZEB1-mediated repression confers to growth and invasion in lung cancer cells. Mol. Cell Biochem. 2016, 411, 331–340. [Google Scholar] [CrossRef] [PubMed]
  113. Yang, J.; Wang, C.; Zhao, F.; Luo, X.; Qin, M.; Arunachalam, E.; Ge, Z.; Wang, N.; Deng, X.; Jin, G.; et al. Loss of FBP1 facilitates aggressive features of hepatocellular carcinoma cells through the Warburg effect. Carcinogenesis 2017, 38, 134–143. [Google Scholar] [CrossRef] [Green Version]
  114. Yang, C.; Zhu, S.; Yang, H.; Deng, S.; Fan, P.; Li, M.; Jin, X. USP44 suppresses pancreatic cancer progression and overcomes gemcitabine resistance by deubiquitinating FBP1. Am. J. Cancer Res. 2019, 9, 1722–1733. [Google Scholar]
  115. Kato, K.; Nakajima, K.; Ui, A.; Muto-Terao, Y.; Ogiwara, H.; Nakada, S. Fine-tuning of DNA damage-dependent ubiquitination by OTUB2 supports the DNA repair pathway choice. Mol. Cell 2014, 53, 617–630. [Google Scholar] [CrossRef] [Green Version]
  116. Zhang, Z.; Du, J.; Wang, S.; Shao, L.; Jin, K.; Li, F.; Wei, B.; Ding, W.; Fu, P.; van Dam, H.; et al. OTUB2 promotes cancer metastasis via Hippo-independent activation of YAP and TAZ. Mol. Cell 2019, 73, 7–21.e7. [Google Scholar] [CrossRef] [Green Version]
  117. Revathidevi, S.; Munirajan, A.K. Akt in cancer: Mediator and more. Semin. Cancer Biol. 2019, 59, 80–91. [Google Scholar] [CrossRef]
  118. Kim, D.H.; Suh, J.; Surh, Y.J.; Na, H.K. Regulation of the tumor suppressor PTEN by natural anticancer compounds. Ann. N. Y. Acad. Sci. 2017, 1401, 136–149. [Google Scholar] [CrossRef]
  119. Li, J.; Cheng, D.; Zhu, M.; Yu, H.; Pan, Z.; Liu, L.; Geng, Q.; Pan, H.; Yan, M.; Yao, M. OTUB2 stabilizes U2AF2 to promote the Warburg effect and tumorigenesis via the AKT/mTOR signaling pathway in non-small cell lung cancer. Theranostics 2019, 9, 179–195. [Google Scholar] [CrossRef] [PubMed]
  120. Santiago-Sim, T.; Burrage, L.C.; Ebstein, F.; Tokita, M.J.; Miller, M.; Bi, W.; Braxton, A.A.; Rosenfeld, J.A.; Shahrour, M.; Lehmann, A.; et al. Biallelic variants in OTUD6B cause an intellectual disability syndrome associated with seizures and dysmorphic features. Am. J. Hum. Genet. 2017, 100, 676–688. [Google Scholar] [CrossRef] [Green Version]
  121. Wang, Y.; Yang, T.; Han, Y.; Ren, Z.; Zou, J.; Liu, J.; Xi, S. lncRNA OTUD6B-AS1 exacerbates As2O3-induced oxidative damage in bladder cancer via miR-6734-5p-mediated functional inhibition of IDH2. Oxid. Med. Cell Longev. 2020, 2020, 3035624. [Google Scholar] [CrossRef] [PubMed]
  122. Wang, Z.; Xia, F.; Feng, T.; Jiang, B.; Wang, W.; Li, X. OTUD6B-AS1 inhibits viability, migration, and invasion of thyroid carcinoma by targeting miR-183-5p and miR-21. Front. Endocrinol. 2020, 11, 136. [Google Scholar] [CrossRef] [Green Version]
  123. Kong, S.; Xue, H.; Li, Y.; Li, P.; Ma, F.; Liu, M.; Li, W. The long noncoding RNA OTUD6B-AS1 enhances cell proliferation and the invasion of hepatocellular carcinoma cells through modulating GSKIP/Wnt/beta-catenin signalling via the sequestration of miR-664b-3p. Exp. Cell Res. 2020, 395, 112180. [Google Scholar] [CrossRef] [PubMed]
  124. Wang, G.; Zhang, Z.J.; Jian, W.G.; Liu, P.H.; Xue, W.; Wang, T.D.; Meng, Y.Y.; Yuan, C.; Li, H.M.; Yu, Y.P.; et al. Novel long noncoding RNA OTUD6B-AS1 indicates poor prognosis and inhibits clear cell renal cell carcinoma proliferation via the Wnt/beta-catenin signaling pathway. Mol. Cancer 2019, 18, 15. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  125. Maher, E.R.; Iselius, L.; Yates, J.R.; Littler, M.; Benjamin, C.; Harris, R.; Sampson, J.; Williams, A.; Ferguson-Smith, M.A.; Morton, N. Von Hippel-Lindau disease: A genetic study. J. Med. Genet. 1991, 28, 443–447. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  126. Liu, X.; Zhang, X.; Peng, Z.; Li, C.; Wang, Z.; Wang, C.; Deng, Z.; Wu, B.; Cui, Y.; Wang, Z.; et al. Deubiquitylase OTUD6B governs pVHL stability in an enzyme-independent manner and suppresses hepatocellular carcinoma metastasis. Adv. Sci. 2020, 7, 1902040. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  127. Dai, X.; Liu, J.; Wei, W. DUB-independent regulation of pVHL by OTUD6B suppresses hepatocellular carcinoma. Protein Cell 2020, 11, 546–548. [Google Scholar] [CrossRef] [Green Version]
  128. Hu, H.; Wang, H.; Xiao, Y.; Jin, J.; Chang, J.H.; Zou, Q.; Xie, X.; Cheng, X.; Sun, S.C. Otud7b facilitates T cell activation and inflammatory responses by regulating Zap70 ubiquitination. J. Exp. Med. 2016, 213, 399–414. [Google Scholar] [CrossRef] [Green Version]
  129. Hu, H.; Brittain, G.C.; Chang, J.H.; Puebla-Osorio, N.; Jin, J.; Zal, A.; Xiao, Y.; Cheng, X.; Chang, M.; Fu, Y.X.; et al. OTUD7B controls non-canonical NF-kappaB activation through deubiquitination of TRAF3. Nature 2013, 494, 371–374. [Google Scholar] [CrossRef] [Green Version]
  130. Lin, D.D.; Shen, Y.; Qiao, S.; Liu, W.W.; Zheng, L.; Wang, Y.N.; Cui, N.; Wang, Y.F.; Zhao, S.; Shi, J.H. Upregulation of OTUD7B (Cezanne) promotes tumor progression via AKT/VEGF pathway in lung squamous carcinoma and adenocarcinoma. Front. Oncol. 2019, 9, 862. [Google Scholar] [CrossRef] [Green Version]
  131. Tan, G.; Wu, L.; Tan, J.; Zhang, B.; Tai, W.C.; Xiong, S.; Chen, W.; Yang, J.; Li, H. MiR-1180 promotes apoptotic resistance to human hepatocellular carcinoma via activation of NF-kappaB signaling pathway. Sci. Rep. 2016, 6, 22328. [Google Scholar] [CrossRef] [Green Version]
  132. Dirckx, N.; Tower, R.J.; Mercken, E.M.; Vangoitsenhoven, R.; Moreau-Triby, C.; Breugelmans, T.; Nefyodova, E.; Cardoen, R.; Mathieu, C.; Van der Schueren, B.; et al. Vhl deletion in osteoblasts boosts cellular glycolysis and improves global glucose metabolism. J. Clin. Invest. 2018, 128, 1087–1105. [Google Scholar] [CrossRef]
  133. Bremm, A.; Moniz, S.; Mader, J.; Rocha, S.; Komander, D. Cezanne (OTUD7B) regulates HIF-1alpha homeostasis in a proteasome-independent manner. EMBO Rep. 2014, 15, 1268–1277. [Google Scholar] [CrossRef]
  134. Goldbraikh, D.; Neufeld, D.; Eid-Mutlak, Y.; Lasry, I.; Gilda, J.E.; Parnis, A.; Cohen, S. USP1 deubiquitinates Akt to inhibit PI3K-Akt-FoxO signaling in muscle during prolonged starvation. EMBO Rep. 2020, 21, e48791. [Google Scholar] [CrossRef] [PubMed]
  135. Hussain, S.; Foreman, O.; Perkins, S.L.; Witzig, T.E.; Miles, R.R.; van Deursen, J.; Galardy, P.J. The de-ubiquitinase UCH-L1 is an oncogene that drives the development of lymphoma in vivo by deregulating PHLPP1 and Akt signaling. Leukemia 2010, 24, 1641–1655. [Google Scholar] [CrossRef] [PubMed]
  136. Pannem, R.R.; Dorn, C.; Ahlqvist, K.; Bosserhoff, A.K.; Hellerbrand, C.; Massoumi, R. CYLD controls c-MYC expression through the JNK-dependent signaling pathway in hepatocellular carcinoma. Carcinogenesis 2014, 35, 461–468. [Google Scholar] [CrossRef] [Green Version]
  137. Benassi, B.; Marani, M.; Loda, M.; Blandino, G. USP2a alters chemotherapeutic response by modulating redox. Cell Death Dis. 2013, 4, e812. [Google Scholar] [CrossRef]
  138. Benassi, B.; Flavin, R.; Marchionni, L.; Zanata, S.; Pan, Y.; Chowdhury, D.; Marani, M.; Strano, S.; Muti, P.; Blandino, G.; et al. MYC is activated by USP2a-mediated modulation of microRNAs in prostate cancer. Cancer Discov. 2012, 2, 236–247. [Google Scholar] [CrossRef] [Green Version]
  139. Nelson, W.G.; De Marzo, A.M.; Yegnasubramanian, S. USP2a activation of MYC in prostate cancer. Cancer Discov. 2012, 2, 206–207. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  140. Bhattacharya, S.; Ghosh, M.K. HAUSP regulates c-MYC expression via de-ubiquitination of TRRAP. Cell Oncol. 2015, 38, 265–277. [Google Scholar] [CrossRef]
  141. Nicklas, S.; Hillje, A.L.; Okawa, S.; Rudolph, I.M.; Collmann, F.M.; van Wuellen, T.; Del Sol, A.; Schwamborn, J.C. A complex of the ubiquitin ligase TRIM32 and the deubiquitinase USP7 balances the level of c-Myc ubiquitination and thereby determines neural stem cell fate specification. Cell Death Differ. 2019, 26, 728–740. [Google Scholar] [CrossRef] [Green Version]
  142. Fang, X.; Zhou, W.; Wu, Q.; Huang, Z.; Shi, Y.; Yang, K.; Chen, C.; Xie, Q.; Mack, S.C.; Wang, X.; et al. Deubiquitinase USP13 maintains glioblastoma stem cells by antagonizing FBXL14-mediated Myc ubiquitination. J. Exp. Med. 2017, 214, 245–267. [Google Scholar] [CrossRef] [Green Version]
  143. Ge, J.; Yu, W.; Li, J.; Ma, H.; Wang, P.; Zhou, Y.; Wang, Y.; Zhang, J.; Shi, G. USP16 regulates castration-resistant prostate cancer cell proliferation by deubiquitinating and stablizing c-Myc. J. Exp. Clin. Cancer Res. 2021, 40, 59. [Google Scholar] [CrossRef]
  144. Feng, L.; Wang, K.; Tang, P.; Chen, S.; Liu, T.; Lei, J.; Yuan, R.; Hu, Z.; Li, W.; Yu, X. Deubiquitinase USP18 promotes the progression of pancreatic cancer via enhancing the Notch1-c-Myc axis. Aging 2020, 12, 19273–19292. [Google Scholar] [CrossRef] [PubMed]
  145. Kim, D.; Hong, A.; Park, H.I.; Shin, W.H.; Yoo, L.; Jeon, S.J.; Chung, K.C. Deubiquitinating enzyme USP22 positively regulates c-Myc stability and tumorigenic activity in mammalian and breast cancer cells. J. Cell Physiol. 2017, 232, 3664–3676. [Google Scholar] [CrossRef] [PubMed]
  146. Liu, Y.L.; Zheng, J.; Tang, L.J.; Han, W.; Wang, J.M.; Liu, D.W.; Tian, Q.B. The deubiquitinating enzyme activity of USP22 is necessary for regulating HeLa cell growth. Gene 2015, 572, 49–56. [Google Scholar] [CrossRef] [PubMed]
  147. Gersch, M.; Wagstaff, J.L.; Toms, A.V.; Graves, B.; Freund, S.M.V.; Komander, D. Distinct USP25 and USP28 oligomerization states regulate deubiquitinating activity. Mol. Cell 2019, 74, 436–451.e7. [Google Scholar] [CrossRef] [PubMed]
  148. Sun, X.X.; He, X.; Yin, L.; Komada, M.; Sears, R.C.; Dai, M.S. The nucleolar ubiquitin-specific protease USP36 deubiquitinates and stabilizes c-Myc. Proc. Natl. Acad. Sci. USA 2015, 112, 3734–3739. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  149. Liu, Q.; Sheng, W.; Ma, Y.; Zhen, J.; Roy, S.; Alvira Jafar, C.; Xin, W.; Wan, Q. USP36 protects proximal tubule cells from ischemic injury by stabilizing c-Myc and SOD2. Biochem. Biophys. Res. Commun. 2019, 513, 502–508. [Google Scholar] [CrossRef]
  150. Sobol, A.; Askonas, C.; Alani, S.; Weber, M.J.; Ananthanarayanan, V.; Osipo, C.; Bocchetta, M. Deubiquitinase OTUD6B isoforms are important regulators of growth and proliferation. Mol. Cancer Res. 2017, 15, 117–127. [Google Scholar] [CrossRef] [Green Version]
  151. Atanassov, B.S.; Dent, S.Y. USP22 regulates cell proliferation by deubiquitinating the transcriptional regulator FBP1. EMBO Rep. 2011, 12, 924–930. [Google Scholar] [CrossRef]
  152. Sun, P.; Lu, Y.X.; Cheng, D.; Zhang, K.; Zheng, J.; Liu, Y.; Wang, X.; Yuan, Y.F.; Tang, Y.D. Monocyte chemoattractant protein-induced protein 1 targets hypoxia-inducible factor 1alpha to protect against hepatic ischemia/reperfusion injury. Hepatology 2018, 68, 2359–2375. [Google Scholar] [CrossRef]
  153. Goto, Y.; Zeng, L.; Yeom, C.J.; Zhu, Y.; Morinibu, A.; Shinomiya, K.; Kobayashi, M.; Hirota, K.; Itasaka, S.; Yoshimura, M.; et al. UCHL1 provides diagnostic and antimetastatic strategies due to its deubiquitinating effect on HIF-1alpha. Nat. Commun. 2015, 6, 6153. [Google Scholar] [CrossRef] [Green Version]
  154. Li, X.; Hattori, A.; Takahashi, S.; Goto, Y.; Harada, H.; Kakeya, H. Ubiquitin carboxyl-terminal hydrolase L1 promotes hypoxia-inducible factor 1-dependent tumor cell malignancy in spheroid models. Cancer Sci. 2020, 111, 239–252. [Google Scholar] [CrossRef] [PubMed]
  155. Nakashima, R.; Goto, Y.; Koyasu, S.; Kobayashi, M.; Morinibu, A.; Yoshimura, M.; Hiraoka, M.; Hammond, E.M.; Harada, H. UCHL1-HIF-1 axis-mediated antioxidant property of cancer cells as a therapeutic target for radiosensitization. Sci. Rep. 2017, 7, 6879. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  156. Wu, H.T.; Kuo, Y.C.; Hung, J.J.; Huang, C.H.; Chen, W.Y.; Chou, T.Y.; Chen, Y.; Chen, Y.J.; Chen, Y.J.; Cheng, W.C.; et al. K63-polyubiquitinated HAUSP deubiquitinates HIF-1alpha and dictates H3K56 acetylation promoting hypoxia-induced tumour progression. Nat. Commun. 2016, 7, 13644. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  157. Troilo, A.; Alexander, I.; Muehl, S.; Jaramillo, D.; Knobeloch, K.P.; Krek, W. HIF1alpha deubiquitination by USP8 is essential for ciliogenesis in normoxia. EMBO Rep. 2014, 15, 77–85. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  158. Park, J.J.; Yun, J.H.; Baek, K.H. Polyclonal and monoclonal antibodies specific for ubiquitin-specific protease 20. Monoclon. Antib. Immunodiagn. Immunother. 2013, 32, 193–199. [Google Scholar] [CrossRef] [PubMed]
  159. Xu, J.; Chen, D.; Jin, L.; Chen, Z.; Tu, Y.; Huang, X.; Xue, F.; Xu, J.; Chen, M.; Wang, X.; et al. Ubiquitously specific protease 4 inhibitor-Vialinin A attenuates inflammation and fibrosis in S100-induced hepatitis mice through Rheb/mTOR signalling. J. Cell Mol. Med. 2021, 25, 1140–1150. [Google Scholar] [CrossRef] [PubMed]
  160. Li, Y.; Zhou, J. USP5 promotes uterine corpus endometrial carcinoma cell growth and migration via mTOR/4EBP1 activation. Cancer Manag. Res. 2021, 13, 3913–3924. [Google Scholar] [CrossRef]
  161. Wrobel, L.; Siddiqi, F.H.; Hill, S.M.; Son, S.M.; Karabiyik, C.; Kim, H.; Rubinsztein, D.C. mTORC2 assembly is regulated by USP9X-mediated deubiquitination of RICTOR. Cell Rep. 2020, 33, 108564. [Google Scholar] [CrossRef] [PubMed]
  162. Lu, C.; Ning, Z.; Wang, A.; Chen, D.; Liu, X.; Xia, T.; Tekcham, D.S.; Wang, W.; Li, T.; Liu, X.; et al. USP10 suppresses tumor progression by inhibiting mTOR activation in hepatocellular carcinoma. Cancer Lett. 2018, 436, 139–148. [Google Scholar] [CrossRef] [PubMed]
  163. Cho, J.H.; Kim, K.; Kim, S.A.; Park, S.; Park, B.O.; Kim, J.H.; Kim, S.Y.; Kwon, M.J.; Han, M.H.; Lee, S.B.; et al. Deubiquitinase OTUD5 is a positive regulator of mTORC1 and mTORC2 signaling pathways. Cell Death Differ. 2021, 28, 900–914. [Google Scholar] [CrossRef]
  164. Kosinsky, R.L.; Zerche, M.; Saul, D.; Wang, X.; Wohn, L.; Wegwitz, F.; Begus-Nahrmann, Y.; Johnsen, S.A. USP22 exerts tumor-suppressive functions in colorectal cancer by decreasing mTOR activity. Cell Death Differ. 2020, 27, 1328–1340. [Google Scholar] [CrossRef] [PubMed]
  165. Zhao, L.; Wang, X.; Yu, Y.; Deng, L.; Chen, L.; Peng, X.; Jiao, C.; Gao, G.; Tan, X.; Pan, W.; et al. OTUB1 protein suppresses mTOR complex 1 (mTORC1) activity by deubiquitinating the mTORC1 inhibitor DEPTOR. J. Biol. Chem. 2018, 293, 4883–4892. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  166. Zhou, J.; Wang, J.; Chen, C.; Yuan, H.; Wen, X.; Sun, H. USP7: Target validation and drug discovery for cancer therapy. Med. Chem. 2018, 14, 3–18. [Google Scholar] [CrossRef] [PubMed]
  167. Liu, H.; Li, X.; Ning, G.; Zhu, S.; Ma, X.; Liu, X.; Liu, C.; Huang, M.; Schmitt, I.; Wullner, U.; et al. The Machado-Joseph disease deubiquitinase ataxin-3 regulates the stability and apoptotic function of p53. PLoS Biol. 2016, 14, e2000733. [Google Scholar] [CrossRef] [Green Version]
  168. Turnbull, A.P.; Ioannidis, S.; Krajewski, W.W.; Pinto-Fernandez, A.; Heride, C.; Martin, A.C.L.; Tonkin, L.M.; Townsend, E.C.; Buker, S.M.; Lancia, D.R.; et al. Molecular basis of USP7 inhibition by selective small-molecule inhibitors. Nature 2017, 550, 481–486. [Google Scholar] [CrossRef]
  169. Kategaya, L.; Di Lello, P.; Rouge, L.; Pastor, R.; Clark, K.R.; Drummond, J.; Kleinheinz, T.; Lin, E.; Upton, J.P.; Prakash, S.; et al. USP7 small-molecule inhibitors interfere with ubiquitin binding. Nature 2017, 550, 534–538. [Google Scholar] [CrossRef] [Green Version]
  170. Reverdy, C.; Conrath, S.; Lopez, R.; Planquette, C.; Atmanene, C.; Collura, V.; Harpon, J.; Battaglia, V.; Vivat, V.; Sippl, W.; et al. Discovery of specific inhibitors of human USP7/HAUSP deubiquitinating enzyme. Chem. Biol. 2012, 19, 467–477. [Google Scholar] [CrossRef] [Green Version]
  171. Colland, F.; Formstecher, E.; Jacq, X.; Reverdy, C.; Planquette, C.; Conrath, S.; Trouplin, V.; Bianchi, J.; Aushev, V.N.; Camonis, J.; et al. Small-molecule inhibitor of USP7/HAUSP ubiquitin protease stabilizes and activates p53 in cells. Mol. Cancer Ther. 2009, 8, 2286–2295. [Google Scholar] [CrossRef] [Green Version]
  172. Altun, M.; Kramer, H.B.; Willems, L.I.; McDermott, J.L.; Leach, C.A.; Goldenberg, S.J.; Kumar, K.G.; Konietzny, R.; Fischer, R.; Kogan, E.; et al. Activity-based chemical proteomics accelerates inhibitor development for deubiquitylating enzymes. Chem. Biol. 2011, 18, 1401–1412. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  173. Pozhidaeva, A.; Valles, G.; Wang, F.; Wu, J.; Sterner, D.E.; Nguyen, P.; Weinstock, J.; Kumar, K.G.S.; Kanyo, J.; Wright, D.; et al. USP7-specific inhibitors target and modify the enzyme’s active ite via distinct chemical mechanisms. Cell Chem. Biol. 2017, 24, 1501–1512. [Google Scholar] [CrossRef] [Green Version]
  174. Wang, F.; Wang, L.; Wu, J.; Sokirniy, I.; Nguyen, P.; Bregnard, T.; Weinstock, J.; Mattern, M.; Bezsonova, I.; Hancock, W.W.; et al. Active site-targeted covalent irreversible inhibitors of USP7 impair the functions of Foxp3+ T-regulatory cells by promoting ubiquitination of Tip60. PLoS ONE 2017, 12, e0189744. [Google Scholar] [CrossRef] [Green Version]
  175. Chauhan, D.; Tian, Z.; Nicholson, B.; Kumar, K.G.; Zhou, B.; Carrasco, R.; McDermott, J.L.; Leach, C.A.; Fulcinniti, M.; Kodrasov, M.P.; et al. A small molecule inhibitor of ubiquitin-specific protease-7 induces apoptosis in multiple myeloma cells and overcomes bortezomib resistance. Cancer Cell 2012, 22, 345–358. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  176. Lee, H.R.; Choi, W.C.; Lee, S.; Hwang, J.; Hwang, E.; Guchhait, K.; Haas, J.; Toth, Z.; Jeon, Y.H.; Oh, T.K.; et al. Bilateral inhibition of HAUSP deubiquitinase by a viral interferon regulatory factor protein. Nat. Struct. Mol. Biol. 2011, 18, 1336–1344. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  177. Schauer, N.J.; Liu, X.; Magin, R.S.; Doherty, L.M.; Chan, W.C.; Ficarro, S.B.; Hu, W.; Roberts, R.M.; Iacob, R.E.; Stolte, B.; et al. Selective USP7 inhibition elicits cancer cell killing through a p53-dependent mechanism. Sci. Rep. 2020, 10, 5324–5338. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  178. Lamberto, I.; Liu, X.; Seo, H.S.; Schauer, N.J.; Iacob, R.E.; Hu, W.; Das, D.; Mikhailova, T.; Weisberg, E.L.; Engen, J.R.; et al. Structure-guided development of a potent and selective non-covalent active-site inhibitor of USP7. Cell Chem. Biol. 2017, 24, 1490–1500. [Google Scholar] [CrossRef] [Green Version]
  179. Wrigley, J.D.; Gavory, G.; Simpson, I.; Preston, M.; Plant, H.; Bradley, J.; Goeppert, A.U.; Rozycka, E.; Davies, G.; Walsh, J.; et al. Identification and characterization of dual inhibitors of the USP25/28 deubiquitinating enzyme subfamily. ACS Chem. Biol. 2017, 12, 3113–3125. [Google Scholar] [CrossRef] [Green Version]
  180. Liu, Z.; Zhao, T.; Li, Z.; Sun, K.; Fu, Y.; Cheng, T.; Guo, J.; Yu, B.; Shi, X.; Liu, H. Discovery of [1,2,3]triazolo[4,5-d]pyrimidine derivatives as highly potent, selective, and cellularly active USP28 inhibitors. Acta Pharm. Sin. B 2020, 10, 1476–1491. [Google Scholar] [CrossRef]
  181. Kasperczyk, H.; La Ferla-Bruhl, K.; Westhoff, M.A.; Behrend, L.; Zwacka, R.M.; Debatin, K.M.; Fulda, S. Betulinic acid as new activator of NF-kappaB: Molecular mechanisms and implications for cancer therapy. Oncogene 2005, 24, 6945–6956. [Google Scholar] [CrossRef] [Green Version]
  182. Fulda, S.; Kroemer, G. Targeting mitochondrial apoptosis by betulinic acid in human cancers. Drug. Discov. Today 2009, 14, 885–890. [Google Scholar] [CrossRef] [PubMed]
  183. Chintharlapalli, S.; Papineni, S.; Lei, P.; Pathi, S.; Safe, S. Betulinic acid inhibits colon cancer cell and tumor growth and induces proteasome-dependent and -independent downregulation of specificity proteins (Sp) transcription factors. BMC Cancer 2011, 11, 371–382. [Google Scholar] [CrossRef] [Green Version]
  184. Reiner, T.; Parrondo, R.; de Las Pozas, A.; Palenzuela, D.; Perez-Stable, C. Betulinic acid selectively increases protein degradation and enhances prostate cancer-specific apoptosis: Possible role for inhibition of deubiquitinase activity. PLoS ONE 2013, 8, e56234. [Google Scholar] [CrossRef] [Green Version]
  185. Kumar, P.; Bhadauria, A.S.; Singh, A.K.; Saha, S. Betulinic acid as apoptosis activator: Molecular mechanisms, mathematical modeling and chemical modifications. Life Sci. 2018, 209, 24–33. [Google Scholar] [CrossRef]
  186. Wang, L.; Li, M.; Sha, B.; Hu, X.; Sun, Y.; Zhu, M.; Xu, Y.; Li, P.; Wang, Y.; Guo, Y.; et al. Inhibition of deubiquitination by PR-619 induces apoptosis and autophagy via ubi-protein aggregation-activated ER stress in oesophageal squamous cell carcinoma. Cell Prolif. 2021, 54, e12919. [Google Scholar] [CrossRef] [PubMed]
  187. Crowder, R.N.; Dicker, D.T.; El-Deiry, W.S. The deubiquitinase inhibitor PR-619 sensitizes normal human fibroblasts to tumor necrosis factor-related apoptosis-inducing ligand (TRAIL)-mediated cell death. J. Biol. Chem. 2016, 291, 5960–5970. [Google Scholar] [CrossRef] [PubMed] [Green Version]
Figure 1. Differences in glycolysis pathways between normal cells and cancer cells. (A) In the presence of oxygen, normal cells produce carbon dioxide up to 38 ATPs per glucose molecule through glycolysis, TCA cycle, and electron transport system. In a hypoxic environment, pyruvates are accumulated without going through the TCA cycle. These accumulated pyruvates in the muscle tissue are converted to lactic acid and only produce 2 ATPs. (B) Cancer cells only use the glycolysis process, regardless of the presence or absence of oxygen; 2 ATPs are produced per glucose molecule and, therefore, compared to normal cells, more glucose is required to obtain energy.
Figure 1. Differences in glycolysis pathways between normal cells and cancer cells. (A) In the presence of oxygen, normal cells produce carbon dioxide up to 38 ATPs per glucose molecule through glycolysis, TCA cycle, and electron transport system. In a hypoxic environment, pyruvates are accumulated without going through the TCA cycle. These accumulated pyruvates in the muscle tissue are converted to lactic acid and only produce 2 ATPs. (B) Cancer cells only use the glycolysis process, regardless of the presence or absence of oxygen; 2 ATPs are produced per glucose molecule and, therefore, compared to normal cells, more glucose is required to obtain energy.
Ijms 22 06173 g001
Figure 2. The schematic diagram of the ubiquitin–proteasome system (UPS). The ATP-activated E1 enzyme binds to glycine at the end of ubiquitin. The E1 enzyme delivers the ubiquitin to the E2 enzyme. The E2 enzyme binds to the E3 enzyme (E3 ligase) bound to the substrate protein. Ubiquitin linked to the E2 enzyme moves to the substrate protein. By repeating this process, several ubiquitins form a polyubiquitin chain, and the substrate is degraded through the 26S proteasome. Deubiquitinating enzyme (DUB) acts in opposition to the E3 ligase, which links ubiquitin to the substrate protein.
Figure 2. The schematic diagram of the ubiquitin–proteasome system (UPS). The ATP-activated E1 enzyme binds to glycine at the end of ubiquitin. The E1 enzyme delivers the ubiquitin to the E2 enzyme. The E2 enzyme binds to the E3 enzyme (E3 ligase) bound to the substrate protein. Ubiquitin linked to the E2 enzyme moves to the substrate protein. By repeating this process, several ubiquitins form a polyubiquitin chain, and the substrate is degraded through the 26S proteasome. Deubiquitinating enzyme (DUB) acts in opposition to the E3 ligase, which links ubiquitin to the substrate protein.
Ijms 22 06173 g002
Figure 3. The schematic model for the Warburg effect modulated by DUBs and their substrates. The black arrow indicates direct stimulatory/inhibitory modification, and the blue arrow indicates transcriptional stimulatory modification. The blue circles represent the factors of the Warburg effect, and the gray square boxes represent their DUBs.
Figure 3. The schematic model for the Warburg effect modulated by DUBs and their substrates. The black arrow indicates direct stimulatory/inhibitory modification, and the blue arrow indicates transcriptional stimulatory modification. The blue circles represent the factors of the Warburg effect, and the gray square boxes represent their DUBs.
Ijms 22 06173 g003
Table 1. The list of DUBs and cellular substrates involved in the Warburg effect.
Table 1. The list of DUBs and cellular substrates involved in the Warburg effect.
DUBsSubstratesFunctionsReferences
USP7SIRT7USP7 decreases the K-63 linked ubiquitination level of SIRT7.[78]
USP7 decreases the ubiquitin level of SIRT7 as the glucose concentration increased.
G6PCDeubiquitination of SIRT7 by USP7 inhibits the expression of G6PC, a key regulator of SIRT7-mediated glucose production.
FoxO1USP7 deubiquitinates and regulates FoxO1 occupancy in the promoter of the glucose-generating gene.[82]
USP19HIF-1αSilence of USP19 decreased the protein level of HIF-1α.[92]
VEGFSilence of USP19 decreased the mRNA level of VEGF.
GLUT1Silence of USP19 decreased the mRNA level of GLUT1.
USP28c-MycUSP28 deubiquitinates c-Myc through interaction with Fbw7.[97]
USP37c-MycUSP37 directly binds to c-Myc and deubiquitinates the c-Myc.[104]
GLUT1Depletion of USP37 leads to decreased expression of GLUT1 mRNAs required for glucose uptake.
LDHADepletion of USP37 leads to decreased expression of LDHA mRNAs required for lactic acid production.
USP44FBP1Knockdown of USP44 improves glucose utilization and lactic acid production capacity by reducing FBP1.[114]
OTUB2c-MycOTUB2 increases the expression level of c-Myc.[119]
HIF-1αOTUB2 increases the expression level of HIF-1α.
GLUT1OTUB2 increases the expression level of GLUT1.
U2AF2OTUB2 increases the expression level of U2AF2.
OTUB2 regulates the Warburg effect via interaction with U2AF2.
HK2OTUB2 increases the expression level of HK2.
PGAM1OTUB2 increases the expression level of PGAM1.
PGK1OTUB2 increases the expression level of PGK1.
mTOROTUB2 increases the level of phosphorylation of mTOR.
AKTOTUB2 increases the level of phosphorylation of AKT.
OTUD6BHIF-1αOverexpression of OTUD6B increased the ubiquitination level of HIF-1α and decreased protein level of HIF-1α.[126,127]
pVHLOTUD6B inhibits pVHL from proteasome degradation through binding with elongin B and enhancing the interaction with pVHL-elongin C.
OTUD7BHIF-1αKnockdown of OTUD7B decreases the protein levels of HIF-1α and increases the K11-linked polyubiquitin chain. [133]
Table 2. The list of DUBs and inhibitors involved in the Warburg effect.
Table 2. The list of DUBs and inhibitors involved in the Warburg effect.
DUBsInhibitorsReferences
USP7FT671[168]
FT827[168]
GNE-6640[169]
GNE-6776[169]
HBX 19,818[170]
HBX 28,258[170]
HBX 41,108[171]
P22077[172]
P50429[173]
P5091[174,175]
vif1[176]
vif2[176]
XL177A[177]
XL188[178]
USP19unknown
USP28AZ1[179]
[1,2,3]triazolo[4,5-d]pyrimidine[180]
USP37unknown
USP44unknown
OTUB2unknown
OTUD6Bunknown
OTUD7Bunknown
Broad spectrum DUB inhibitorBetulinic acid[181,182,183,184,185]
PR-619[186,187]
Publisher’s Note: MDPI stays neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Share and Cite

MDPI and ACS Style

Kim, S.-H.; Baek, K.-H. Regulation of Cancer Metabolism by Deubiquitinating Enzymes: The Warburg Effect. Int. J. Mol. Sci. 2021, 22, 6173. https://doi.org/10.3390/ijms22126173

AMA Style

Kim S-H, Baek K-H. Regulation of Cancer Metabolism by Deubiquitinating Enzymes: The Warburg Effect. International Journal of Molecular Sciences. 2021; 22(12):6173. https://doi.org/10.3390/ijms22126173

Chicago/Turabian Style

Kim, So-Hee, and Kwang-Hyun Baek. 2021. "Regulation of Cancer Metabolism by Deubiquitinating Enzymes: The Warburg Effect" International Journal of Molecular Sciences 22, no. 12: 6173. https://doi.org/10.3390/ijms22126173

Note that from the first issue of 2016, this journal uses article numbers instead of page numbers. See further details here.

Article Metrics

Back to TopTop