Open Access

Roles of USP9X in cellular functions and tumorigenesis (Review)

  • Authors:
    • Yimei Meng
    • Chaojin Hong
    • Sifu Yang
    • Zhiquan Qin
    • Liu Yang
    • Yumei Huang
  • View Affiliations

  • Published online on: October 10, 2023     https://doi.org/10.3892/ol.2023.14093
  • Article Number: 506
  • Copyright: © Meng 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

Ubiquitin‑specific peptidase 9X (USP9X) is involved in certain human diseases, including malignancies, atherosclerosis and certain diseases of the nervous system. USP9X promotes the deubiquitination and stabilization of diverse substrates, thereby exerting a versatile range of effects on pathological and physiological processes. USP9X serves vital roles in the processes of cell survival, invasion and migration in various types of cancer. The present review aims to highlight the current knowledge of USP9X in terms of its structure and the possible mediatory mechanisms involved in certain types of cancer, providing a thorough introduction to its biological functions in carcinogenesis and further outlining its oncogenic or suppressive properties in a diverse range of cancer types. Finally, several perspectives regarding USP9X‑targeted pharmacological therapeutics in cancer development are discussed.

Introduction

The precise control of cell activity in response to systemic or local signals is largely governed by the regulation of homoeostasis (1). Proteins are known to execute a diverse range of intracellular processes (2). In order to accurately perform various physiological functions, the intracellular protein quality must be precisely and strictly controlled, and this level of control depends on the balance between protein degradation and synthesis (3). The lysosomal systems and the ubiquitin-proteasome system (UPS) form an interconnected protein quality control network for lysosomal and proteasomal protein degradation, respectively (4,5). In the UPS, ubiquitin is conjugated to targeted proteins, which are subsequently recognized by the proteasome for degradation (6,7). Ubiquitinases and deubiquitinases (DUBs) in the ubiquitin proteasome pathway are involved in various pathological and physiological processes, such as DNA repair (8), apoptosis (9) and cancer metabolism (10). DUBs are classified into seven families on the basis of their sequences and structural homology, namely ubiquitin-specific proteases (USPs), ovarian tumor proteases, the JAB1/MPN/MOV34 family of metalloenzymes, ubiquitin carboxy-terminal hydrolases (UCHs), the motif interacting with ubiquitin-containing novel DUB family, Machado-Josephin domain-containing proteases and the zinc finger-containing ubiquitin peptidase 1. The structural or functional abnormalities of these enzymes can lead to numerous diseases, including cancer (11,12). The USP family accounts for the largest proportion of DUBs that have been identified, and share a catalytic domain that comprises 300–800 amino acids (13).

Ubiquitin-specific peptidase 9X (USP9X), a member of the USP family, regulates numerous signaling pathways by deubiquitinating essential proteins, such as myeloid leukemia cell differentiation 1 (MCL-1) (14,15). USP9X is closely associated with neurological disorders (1618), atherosclerosis (19) and cancers (20). However, to date, USP9X and its precise roles in different types of cancer have not been widely investigated or specifically and systematically reviewed. In addition, the value of USP9X in terms of its clinical and potential application as a cancer target may be underestimated. Therefore, in the present review, the current evidence in support of the cellular functions and underlying regulatory mechanisms of USP9X in tumorigenesis is summarized, as well as the biological mechanisms associated with USP9X in different types of cancer.

Structure of USP9X

The highly conserved DUB USP9X is located on chromosome Xp11.4 and was first identified as a human homologue of the Drosophila fat facets (faf) gene, subsequently named DFFRX, which escapes X-chromosome inactivation and serves vital roles in embryo development (21). The protein sequence of USP9X and its molecular functions are evolutionarily conserved across species (2225). The two recognizable domains of USP9X are the ubiquitin-like module (UBL) domain (amino acids 886–970) and the catalytic domain with USP-definitive cysteine (amino acid 1,566) and histidine (amino acid 1,879) box catalytic motifs (Fig. 1A) (15). The long non-conserved N- and C-terminal extensions flank the catalytic domains of USP9X (26,27). The homology model of USP9X and its functional domains were constructed from the Alphafold Database (https://alphafold.com/) with PyMOL (The PyMOL Molecular Graphics System; version 2.3; Schrödinger, LLC; Fig. 1B-D). The crystal structure of USP9X has been solved and reported as a conserved and canonical USP-fold consisting of palm, finger and thumb subdomains, and a β-hairpin insertion (28). The catalytic domain of USP9X harbors a zinc finger motif and three ubiquitin binding sites in the fingers subdomain, and the β-hairpin insertion, contributing to polyubiquitin chain processing and cleavage of lysine (Lys)11-, Lys63-, Lys48- and Lys6-linkages, enabling the protein to perform a variety of cellular functions (2934).

USP9X is predominantly present in the cytoplasm (35,36) and at the cell membrane (37), whereas small amounts of the protein are internalized into the mitochondria (38), the nucleus (39) and the centrosome (40). USP9X regulates apoptosis, mitotic fidelity, inflammation, ribosomal stalling, proliferation, epithelial-mesenchymal transition (EMT), oxidative stress, the stemness of cancer cells, chromatin reprogramming and drug resistance through the precise recognition, recruitment and binding of diverse substrates for targeted deubiquitination and stabilization (4157) (Table I; Fig. 2). USP9X is also involved in several vital and classical signaling pathways, including the transforming growth factor-β (TGF-β), Hippo, Wnt/β-catenin and Janus kinase (JAK)-STAT signaling pathways (Fig. 3). Moreover, USP9X has been found to be enriched in the majority of cancer samples, and the deleterious genetic variants of USP9X are associated with neurodevelopmental disorders and neurodegeneration (1618), indicating its essential and potential function in clinical treatment.

Table I.

Substrates and mechanisms of ubiquitin-specific peptidase 9X in cancers described in this review.

Table I.

Substrates and mechanisms of ubiquitin-specific peptidase 9X in cancers described in this review.

First author, yearType of cancerIn vivo or in vitroExpression regulationTargetEffectTumor biological behaviors(Refs.)
Engel et al, 2016Aggressive B-cell lymphomaIn vivo and in vitroUpXIAPStabilizeInhibits apoptosis and increases chemoresistance(41)
Grou et al, 2012Chronic myelogenous leukemiaIn vitroUpMCL-1StabilizeInhibits apoptosis(42)
Akiyama et al, 2019AMLIn vivo and in vitro-MCL-1StabilizeInhibits apoptosis(43)
Wang et al, 2023AMLIn vivo and in vitroUpALKBH5StabilizePromotes cell survival(44)
Akiyama et al, 2020Multiple myelomaIn vitroUpMCL-1StabilizePromotes cell survival(45)
Wu et al, 2017Breast cancerIn vivo and in vitroUpSMAD4InhibitPromotes cancer progression and metastasis(46)
Li et al, 2018Breast cancerIn vivo and in vitroUpYAP1StabilizePromotes cell survival and regulates chemotherapy resistance(47)
Guan et al, 2022Breast cancerIn vivo and in vitroUpSnailStabilizePromotes metastasis and chemoresistance(48)
Li et al, 2017Breast cancerIn vivo and in vitroUpCEP131StabilizePromotes centrosome amplification and chromosome instability(40)
Narayanan et al, 2017Breast cancerIn vitroUpTDRD3StabilizeInhibits apoptosis(35)
Lu et al, 2021Breast cancerIn vitroUpHIF-1αStabilizeImproves cancer stem cell self-renewal(49)
Jie et al, 2021Lung cancerIn vivo and in vitroUpKDM4CStabilizePromotes radioresistance(50)
Chen et al, 2018NSCLCIn vivo and in vitroUpTTKStabilizePromotes tumorigenesis(51)
Kushwaha, et al, 2015NSCLCIn vitroUpMCL-1StabilizePromotes apoptosis(52)
Sulkshane et al, 2021OSCCIn vivo and in vitroUpMCL-1StabilizePromotes tumorigenesis and inhibits apoptosis(53)
Cheng et al, 2016GlioblastomaIn vivo and in vitroUpALDH1A3StabilizeMaintains self-renewal and mesenchymal features(54)
Khan et al, 2018Colorectal cancerIn vivo and in vitroDownFBW7StabilizeSuppresses tumor formation(55)
Potu et al, 2017MelanomaIn vivo and in vitroUpEts-1StabilizeEnhances tumorigenicity(56)
Chen et al, 2021 CholangiocarcinomaIn vivo and in vitroDownEGLN3StabilizePromotes apoptosis(57)

[i] XIAP, X-linked inhibitor of apoptosis protein; MCL-1, myeloid cell leukemia 1; AML, acute myeloid leukemia; ALKBH5, AlkB homolog 5; SMAD4, SMAD family member 4; YAP1, yes-associated protein1; CEP131, Centrosomal protein 131; TDRD3, tudor domain-containing protein 3; HIF-1α, hypoxia-inducible factor-1α; KDM4C, histone lysine demethylase 4C; NSCLC, non-small-cell lung cancer; TTK, threonine tyrosine kinase; OSCC, oral squamous cell carcinoma; ALDH1A3, aldehyde dehydrogenase 1A3; FBW7, F-box and WD repeat domain-containing 7; Ets-1, E26 transformation-specific-1; EGLN3, prolyl hydroxylase 3.

Cellular and biological functions of USP9X

Regulation of both the expression level of USP9X and its functions could lead to diverse and dynamic biological behavioral changes under a variety of cellular conditions. USP9X has been identified to have a series of substrates that enable the DUB to regulate cell apoptosis and survival, mitotic fidelity and the cell cycle, cell migration and invasion, and DNA damage repair. USP9X exhibits considerable control over these cellular functions and also in the development of a number of diseases.

Regulation of cell apoptosis and survival

The execution of apoptosis is countered by the action of anti-apoptotic proteins (58). Since certain substrates of USP9X, such as MCL-1 and XIAP, are key factors in cellular apoptosis signaling pathways that drive cell apoptosis, targeting these substrates via USP9X might lead to the regulation of cell apoptosis (4145). USP9X displays both pro- and anti-apoptotic functions, mediated by the deubiquitination of critical components of the apoptotic signaling networks (53,59). Previous studies have reported that the expression of stress-sensing, pro-apoptotic kinases is regulated by USP9X to initiate the apoptotic JNK signaling cascade (59,60). The pro-apoptotic kinase, apoptosis signal-regulating kinase 1 (ASK-1), is activated under conditions of oxidative stress, leading to the selective activation of the JNK signaling pathways (59). USP9X also interacts with ASK-1, protecting it from proteasomal degradation to mediate oxidative stress-induced cell death (59). USP9X also activates and stabilizes the dual leucine zipper kinase in response to extracellular and intracellular stress in neurons, thereby enabling the activation of pro-apoptotic JNK signaling (60).

By contrast, USP9X enhances the activities of a large spectrum of anti-apoptotic factors for cell survival. In the classical mitochondrial apoptosis pathway, the pro-survival B-cell lymphoma-2 family proteins, including MCL-1, preserve mitochondrial integrity and indirectly inhibit the activation of caspase-3 and −7, ultimately limiting the rate of cell apoptosis (17). USP9X stabilizes MCL-1 through its interaction with the protein and the removal of the Lys48-linked polyubiquitin chains that mark a protein for proteasomal degradation (53). A recent study reported that the RNA helicase Asp-Glu-Ala-Asp-box polypeptide 3 interacted with the N-terminus of USP9X and participated in deubiquitination of MCL-1 (61). Therefore, human tumors with a high level of MCL-1 expression may be accompanied by the overexpression of USP9X (53,62). Upregulated expression of USP9X promotes tumorigenicity and cell survival through stabilizing cell death regulators, including X-linked inhibitor of apoptosis protein (XIAP), and inhibiting the induction of apoptosis by specifically stabilizing MCL-1 (53). WP1130 is a small molecule that directly decreases the DUB activity of USP9X, which leads to the downregulation of MCL-1, ultimately facilitating apoptosis (63,64). A previous study reported that apoptosis was induced by the inhibition of USP9X at least partly through oxidative stress, which activated DNA damage responses and stress-associated mitogen-activated protein kinase (MAPK) signaling pathways (43). Therefore, USP9X, when co-expressed with multiple apoptosis-associated proteins, exerts an anti-apoptotic role in cancers, such as oral cancer, prostate cancer, chronic myeloid leukemia (CML) and acute myeloid leukemia (AML) (6567). Overexpression or depletion of USP9X is therefore an important factor in cell apoptosis and survival.

Regulation of mitotic fidelity and the cell cycle

Regulation of mitosis safeguards cellular integrity and its failure contributes to the progression, maintenance and drug resistance of cancer (6870). In the cell cycle, the mitotic checkpoint complex (MCC) senses the orientation of sister chromatids on the mitotic spindle and restricts the activity of anaphase-promoting complex/cyclosome (APC/C) ubiquitin ligase to initiate mitotic exit, consequently ensuring that chromosome segregation and anaphase are able to occur (71). In this way, the spindle assembly checkpoint is strengthened and chromosomal stability is made more secure through restricting APC/C-mediated MCC turnover (71). By contrast, downregulation of USP9X contributes to a reduction in the efficacy of the spindle assembly checkpoint, an increase in chromosome segregation defects and chromosomal instability, which leads to the subsequent promotion of cancer (71). Activating the mitotic phosphorylation of USP9X promotes cell survival through counteracting mitotic ubiquitination and the ensuing proteasomal degradation of Wilms' tumor protein 1, the latter of which modulates the transcription and secretion of CXC motif chemokine ligand 8/interleukin-8 in mitosis (72). In addition, the dephosphorylation of USP9X mediated by cell division cycle 14B (CDC14B) has previously been shown to promote mitotic apoptosis (72). USP9X is an integral component of the centrosome, where it functions to stabilize certain centrosome proteins, centrosomal protein 55 and pericentriolar material 1, thereby promoting centrosome biogenesis (73). Activation of USP9X enhances centrosome amplification and chromosome instability, whereas inactivation of USP9X leads to an impairment of centrosome duplication (40). It has been previously reported that USP9X may stabilize cell cycle-associated proteins to control ribosomal stalling (74), regulate centrosome duplication (40), and antagonize mitotic cell death and chemoresistance (41). Proteins, such as USP9X, that coordinate the normal functioning of the cell cycle may also trigger inappropriate cell divisions, thereby taking on dysfunctional roles in certain pathological disorders, including X-linked intellectual disability (16,18) and malignancies (40).

Regulation of cell migration and invasion

USP9X also serves pivotal roles in cell migration and invasion. Doublecortin (DCX) is a microtubule-associated protein involved in vesicle transport and microtubule dynamics (75). The C-terminus of USP9X binds DCX, which acts as a regulator of neuronal cell migration (76). Loss of USP9X has been shown not only to reduce axon growth, but also to cause a reduction in neuronal cell migration both in vivo and in vitro via USP9X-mediated disruption of the neuronal cytoskeleton (18). Accordingly, USP9X is required for normal neuronal cell migration. In addition, USP9X-mediated deubiquitination of integrin a5b1 serves a role in a5b1-dependent cell migration (77). A previous study reported that USP9X promoted TGF-β-dependent cancer progression and metastasis through interaction with SMAD4 in the TGF-β signaling pathway, thereby inhibiting the E3 ubiquitin-protein ligase TIF1γ-mediated ubiquitination of SMAD4 (46). SMAD-specific E3 ubiquitin protein ligase 1 (SMURF1) was originally identified to block the TGF/bone morphogenetic protein (BMP) signaling pathway by specifically degrading SMAD1 and SMAD5, as well as TGF/BMP receptors (78,79). The negative regulation of USP9X destabilizes SMURF1 and inhibits SMURF1-dependent cell migration in breast cancer cells (80). USP9X has also been shown to activate the prostaglandin E synthase (PTGES)/prostaglandin E2 (PGE2) pathway, thereby promoting metastatic features of non-small cell lung cancer (NSCLC) cells through the deubiquitination and stabilization of PTGES, which functions as a key enzyme for the process of PGE2 synthesis in the arachidonic acid pathway (81). It has previously been demonstrated that USP9X is able to disrupt neuronal cell migration and growth, which is associated with X-linked intellectual disability; therefore, USP9X may serve roles in both neurodevelopment and the modulation of neural apoptosis (18). Taken together, these findings demonstrate the importance of UPS9X in regulating cell migration and invasion.

Regulation of DNA damage repair

USP9X serves important roles in DNA damage repair (82). Genomic instability is a cancer hallmark, often discovered at early stages of the tumorigenesis process (83). The repair of DNA double-strand breaks may trigger the modification of proteins via polyubiquitin or monoubiquitin around the damage sites (84). During DNA replication, the ataxia telangiectasia-mutated and Rad3-related kinase/checkpoint kinase 1 pathway is activated to coordinate both fork-repair processes and checkpoint responses. In a previous study, USP9X was found to enhance the maintenance of DNA damage checkpoint responses and improve DNA replication fork stability by modulating the checkpoint adaptor and DNA replication factor claspin during S phase, thereby preventing DNA damage accumulation or the DNA replication blockade (85). In addition, USP9X depletion was shown to block the progression of DNA replication forks, which increased sensitivity to ionizing radiation, indirectly impairing genome integrity and leading to excessive endogenous DNA damage (85). Loss of USP9X also affects the radiosensitivity of, and cell survival in, glioblastoma through a number of MCL-independent and -dependent mechanisms (86). The potential involvement of USP9X during radiotherapy remains unknown; however, a previous study reported that it may promote lung cancer radioresistance via epigenetically inducing TGF-β2 transcription, which serves to increase the survival of lung cancer cells (26). Therefore, USP9X serves an important role in the stability of the genome during DNA replication. The positive regulation of the DNA damage repair process may also indicate that USP9X is a potential tumor promoter in cancer, also suggesting its candidacy as a potential clinical target in tumor therapy.

Roles of USP9X in human cancer

USP9X expression in different cells and cancer types

USP9X expression level has been shown to differ in tissues and cell lines (Fig. 4A). Furthermore, increased expression levels of USP9X are found in certain cancer types, including breast and lung cancer, and glioblastoma (Fig. 4B; Table I), according to the data from the ProteomicsDB database (87). The therapeutic potential of USP9X in different types of cancer has been demonstrated through considering its deubiquitination capabilities on multiple regulated substrates and critical signaling pathways (88,89). As for the survival and prognosis value of USP9X, high expression of USP9X in adrenocortical, and bladder urothelial carcinoma tissues is associated with lower disease-free survival rates of patients, although it is associated with higher overall survival (OS) rates in cholangiocarcinoma (CHOL; Fig. 5), according to the data from GEPIA (http://gepia.cancer pku.cn/) (90).

USP9X-associated non-coding RNAs in cancer

The available evidence demonstrates that the USP9X-induced aggressive and metastatic phenotypes of cancer are regulated by the abnormal expression of non-coding RNAs (9197) (Table II). The long non-coding RNA LINC01433 has been shown to enhance the interaction between USP9X and Yes-associated protein (YAP), thereby stabilizing YAP and leading to the promotion of tumor progression and chemotherapy resistance in gastric cancer (GC) cells (91). In addition, the circular RNA hsa_circ_0008434 is highly expressed in GC and serves the role of upregulating the expression of USP9X, thereby promoting the malignant phenotypes of GC cells by serving as a microRNA (miR) sponge for miR-6838-5p (93). Moreover, downregulation of the cellular FADD-like interleukin-1β-converting enzyme-inhibitory protein, which is induced by WP1130, a type of selective USP9X inhibitor that decreases the activity of USP9X, was shown to be regulated by miR-708 through the inhibition of USP9X, which induces the apoptosis of CaSki cells (98). In addition, miR-132 was previously shown to target USP9X-induced EMT, which led to inhibition of the migration and invasive capabilities of NSCLC cells (97).

Table II.

USP9X-related non-coding RNAs in various types of cancer.

Table II.

USP9X-related non-coding RNAs in various types of cancer.

A, LncRNA

Non-coding RNAType of cancerExpression regulationMechanismsMain functions(Refs.)
lnc01433Gastric cancerUp lnc01433-USP9X-YAPPromotes proliferation, migration, invasion and chemotherapy resistance(91)
lnc473HCCUp lnc473-USP9X-survivinPromotes cell proliferation and invasion(92)

B, circRNA

Non-coding RNAType of cancerExpression regulation MechanismsMain functions(Refs.)

hsa_circ_0008434-miR-6838-5pGastric cancerUp Hsa_circ_0008434-miR-6838-5p-USP9XPromotes cell proliferation, migration and invasion(93)

C, miRNA

Non-coding RNAType of cancerExpression regulation MechanismsMain functions(Refs.)

miR-26bHCCDownmiR-26b-USP9X-SMAD4 and TGF-β-EMTInhibits EMT(94)
miR-708Renal carcinomaDown WP1130-USP9X-miR-708-c-FLIPPromotes apoptosis(66)
miR-212Non-small-cell lung cancerDown miR-212-USP9X-EMTInhibits cell invasion and migration(95)
miR-212Pancreatic ductal adenocarcinomaDownmiR-212-USP9XEnhances sensitivity to doxorubicin and promotes EMT, apoptosis and autophagy(96)
miR-132Lung cancerDown miR-132-USP9X-EMTInhibits cell migration and invasion(97)

[i] miR/miRNA, microRNA; lncRNA, long non-coding RNA; circRNA, circular RNA; USP9X, ubiquitin-specific peptidase 9X; YAP, yes-associated protein; HCC, hepatocellular carcinoma; TGF-β, transforming growth factor-β; EMT, epithelial-mesenchymal transition; c-FLIP, cellular FADD-like interleukin-1β-converting enzyme-inhibitory protein.

USP9X and hematological malignancies

USP9X has been shown to exert opposing effects in hematological malignancies. It has been reported that USP9X can promote tumor cell survival in human diffuse large B-cell lymphoma, multiple myeloma and follicular lymphoma through deubiquitinating MCL-1, which protects it from degradation, resulting in poor clinical outcomes (38,45,99,100). In addition, a high level of USP9X in patients with aggressive B-cell lymphoma has been shown to be associated with excessive B-cell proliferation, resulting in an adverse prognosis and resistance to treatment therapies through the deubiquitination and stabilization of XIAP, independent of MCL-1 (41). A BCR-ABL-positive diagnosis is a typical characteristic of patients with CML. The WP1130-mediated inhibition of USP9X is associated with a reduction in MCL-1 levels, followed by blockade of BCR-ABL kinase signaling, which leads to the rapid onset of CML cell apoptosis (21). However, USP9X is not involved in BCR-ABL ubiquitination or cellular localization (101). Moreover, USP9X silencing was shown to lead to lymphoma growth suppression, leading to decreased chemotherapy resistance in B-cell lymphoma (63). Internal tandem duplications of FMS-like tyrosine kinase 3 (FLT3-ITD) occur frequently in AML, and is associated with poor outcomes in patients with AML. USP9X interacts with FLT3-ITD to inhibit its Lys63-linked polyubiquitination (43,102). Furthermore, FLT3-ITD reversely induces the ubiquitination and tyrosine-phosphorylation of USP9X, thereby promoting its proteasomal degradation (102). USP9X also stabilizes RNA m6A demethylase ALKBH5 by removing the K48-linked polyubiquitin chain at K57 and promotes AML cell survival (44). Taken together, these findings identify USP9X as an oncogenic gene that promotes the development of hematological malignancies, and the therapeutic targeting of USP9X may lead to preferential inhibition of certain types of leukemia.

USP9X also exerts negative effects in hematological malignancies by acting as a tumor suppressor gene. USP9X has been identified as a novel leukemia susceptibility gene associated with B-cell acute lymphoblastic leukemia (B-ALL) and multiple neurodevelopmental and congenital abnormalities (103). Low USP9X expression is associated with decreased survival in patients with high-risk B-ALL (103). Genetic or pharmacological inhibition of USP9X can restrict JAK signaling to enhance the survival of cytokine receptor-like factor 2-positive B-ALL in patients with Down syndrome (104), which suggests that USP9X may exert differential effects in different types of leukemia.

USP9X and breast cancer

The underlying mechanisms of USP9X in breast cancer are complex. YAP is an important oncogene that drives cancer progression, and dysregulation of the Hippo/YAP1 signaling pathway is involved in breast cancer development (105). YAP1 is deubiquitinated and stabilized by USP9X, thereby promoting breast cancer cell survival and resulting in chemotherapy resistance (47). USP9X stabilizes Snail1, a key factor regulating the EMT process, contributing to metastasis and chemoresistance in triple-negative breast cancer (48). Furthermore, downregulation of USP9X renders estrogen receptor α-positive breast cancer resistant to tamoxifen, leading to a poor outcome for patients following adjuvant tamoxifen treatment (89). USP9X can modulate centrosome biogenesis and enhance breast carcinogenesis by deubiquitinating and stabilizing the centriolar satellite protein, CEP131 (40). In a previous study, the arginine methylation of USP9X was shown to improve its interaction with Tudor domain-containing protein 3 (TDRD3), which led to a subsequent enhancement of its anti-apoptotic activities in breast cancer cells (35). The aforementioned study also reported that downregulation of TDRD3 improved the sensitivity of chemotherapeutic drug-induced apoptosis in breast cancer cells, which is likely due to its regulation of USP9X DUB activity on the anti-apoptotic protein MCL-1 and stress granule localization (35).

Obesity may be a risk factor for the development of breast cancer, as it is associated with reduced survival and increases the risk of distant metastasis for female patients diagnosed with breast cancer (106). Plasma free fatty acids further facilitate this biological progression of breast cancer in obese patients (46). USP9X can also be recruited by the transcription factor Nanog to stabilize the hypoxia-inducible factor-1α protein, which leads to an enhancement of self-renewal in breast cancer stem cells (49). Moreover, USP9X selectively promotes activation of the Notch developmental signaling pathway in triple-negative breast cancer, and small molecule EOAI3402143 (G9)-mediated USP9X inhibition specifically inhibits the Notch pathway, which leads to a remodeling of the tumor immune landscape and suppresses tumor growth (107). Taken together, these studies suggest that a diverse range of USP9X-associated mechanisms are involved in breast cancer.

USP9X and lung cancer

Histone lysine demethylase 4C (KDM4C), a substrate of USP9X, is associated with poor clinical outcomes in patients with lung cancer (50). A recent study reported that USP9X activated the TGF-β/SMAD signaling pathway, thereby inducing radioresistance by deubiquitinating and stabilizing KDM4C in lung cancer (50). USP9X is also positively correlated with the dual-specificity protein kinase known as threonine tyrosine kinase, which modulates cell proliferation, migration and tumorigenesis in NSCLC (51). Furthermore, the inhibition of USP9X has been shown to contribute towards MCL-1-mediated proteasomal degradation, radiosensitivity and apoptosis in NSCLC cells (52). Moreover, the chemotherapeutic agent pemetrexed has been shown to induce apoptosis by increasing the expression of the pro-apoptotic protein Noxa, thereby activating the Noxa-USP9X-MCL-1 axis, which demonstrates that USP9X serves a critical role in human lung cancer cells (108). Collectively, these studies have shown that USP9X exerts a range of different functions in lung cancer.

USP9X and pancreatic cancer

Pancreatic cancer is a lethal malignancy, and effective targeted therapies are urgently required to combat its high morbidity rates (2.6%) and mortality (4.7%) rates worldwide in 2020 (109). To date, USP9X has been reported to show tumor-suppressing potential in pancreatic cancer (110112). USP9X suppressed tumorigenesis in a V-Ki-ras2 Kirsten rat sarcoma viral oncogene homolog mouse model of pancreatic ductal adenocarcinoma (PDAC) and its downregulation enhanced malignant transformation, protecting PDAC cells from anoikis (110). However, these properties of USP9X in pancreatic cancer predominantly depend on its intrinsic deubiquitinase activity. USP9X mediates acute MCL-1 stabilization and protection from apoptosis in response to MAPK suppression caused by MEK inhibitors (113), which may provide a promising therapeutic strategy for pancreatic cancer.

USP9X and other types of cancer

The differential expression or genetic alterations of USP9X have been identified in multiple types of human cancer, which suggests that it potentially exerts different roles in tumor progression. Higher expression levels of USP9X are observed in oral squamous cell carcinoma (OSCC) cells, where it drives oral tumorigenesis by deubiquitinating and stabilizing the anti-apoptotic protein MCL-1, which was shown to correlate with poor outcomes in patients with OSCC (53). In addition, aldehyde dehydrogenase 1 family member A3 (ALDH1A3) functions as a key enzyme for maintaining the self-renewal and mesenchymal (MES) features of glioblastoma stem cells (54). Depletion of USP9X leads to a marked downregulation of ALDH1A3, which leads to a loss of the tumorigenic and self-renewal capabilities of MES glioblastoma stem cells. By contrast, a high expression level of USP9X is indicative of potent tumorigenic capability in MES glioblastoma stem cells with enrichment of ALDH1A3 (114). A previous study reported that USP9X-mediated deubiquitination and stabilization of Ets-1 promoted the expression of the N-RAS oncogene and carcinogenesis in melanoma (56).

Although USP9X has been reported to exert oncogenic functions in several different types of cancer, a number of previous studies have also reported the tumor-suppressive properties of USP9X in carcinogenesis. In the murine intestine, USP9X is necessary for tissue homeostasis and regeneration following acute colitis (55); it regulates the function and protein expression levels of the tumor suppressor FBW7, which consequently protects it from proteasomal degradation (55). Therefore, the restricted level of c-Myc that is regulated by USP9X via stabilization of FBW7 reduces the risk of colitis-mediated colorectal cancer (CRC) in mice, whereas the silencing of USP9X is associated with a poor prognosis in human CRC (55). USP9X expression is downregulated in CRC and CHOL (89). Moreover, USP9X is involved in the activation of apoptosis in CHOL, which also suppresses tumor cell proliferation (57). USP9X exerts its tumor-suppressive functions through deubiquitinating Egl-9 family hypoxia inducible factor 3, thereby activating the apoptosis signaling pathway components, kinesin KIF1Bβ and cleaved caspase-3 (57). USP9X also targets and regulates the stability of angiomotin to indirectly inhibit YAP/transcriptional coactivator with PDZ-binding motif activity, and a low level of USP9X is correlated with poor clinical outcome in renal clear cell carcinoma (115). Taken together, these findings suggest that there may be potential for USP9X to be used as a therapeutic target for the treatment of certain types of cancer.

USP9X protein as a therapeutic target

Small molecule inhibitors that target DUBs are emerging as a novel form of anticancer therapeutic strategy (82). A previous study reported that USP9X overexpression inhibited apoptosis and promoted tumor cell survival (41,57). Moreover, previous studies have identified USP9X as an oncogene in various types of cancer (47,53), suggesting that it may be a future potential target for therapeutic development. However, no small molecule inhibitors specific to USP9X are currently available for use in a clinical setting. WP1130, previously known as Degrasyn (116), is the best-characterized USP9X inhibitor capable of inhibiting USP9X activity, in addition to the activity of several other DUBs (USP5, USP14, USP24, UCH37 and UCH-L1) (117), thereby highlighting its role as a type of partially selective DUB inhibitor. The proliferation, inhibition and anti-apoptotic effects of WP1130 against diverse tumors have been reported in B-cell malignancies (118), AML (43), NSCLC (51) and glioblastoma (114), mainly through the inhibition of USP9X with the subsequent accumulation of polyubiquitinated proteins and downregulation of antiapoptotic proteins, including MCL-1 and p53. WP1130 has also been shown to increase tumor cell sensitivity to chemotherapy (43). BIX-01294 is an inhibitor developed to inhibit the activity of euchromatic histone-lysine N-methyltransferase 2 and has been shown to promote USP9X downregulation by stimulating both endoplasmic reticulum stress and the expression of phorbol-12-myristate-13-acetate-induced protein 1, resulting in a downregulation of the level of MCL-1, which promotes apoptosis in bladder cancer cells (119). Peterson et al (118) reported on a small molecule inhibitor of USP9X/USP24, EOAI3402143(G9), which inhibited the DUB activity of USP9X and USP24 in a dose-dependent manner, increased apoptosis in myeloma and fully regressed or blocked myeloma tumors in mice. G9 was shown to induce apoptosis in lymphoma and myeloma cell lines in vitro, inhibiting tumor progression with little overt toxicity (120). BRD0476 selectively inhibits USP9X activity to suppress the JAK-STAT pathway, which protects human pancreatic β-cells and cancer cells from cytokine-induced apoptosis (121). It was also demonstrated that the disruption of USP9X by the CRISPR/Cas9 system and small interfering RNA intervention brought about similar protective effects resulting from BRD0476 treatment, which suggests that BRD0476 may function as a modulator of USP9X (121). Isothiocyanates, such as phenethyl isothiocyanate and benzyl isothiocyanate, have also been shown to exert anticancer activity by inhibiting USP9X and other DUBs in physiologically relevant time scales and concentrations, particularly in hematological malignancies (122). Taken together, these study results highlight the potential of pharmacological inhibitors targeting USP9X, which may be an effective therapeutic target courtesy of its DUB activity. Although inhibitors of USP9X are not at present available for clinical use, novel therapeutic approaches targeting USP9X in human cancers may be developed further in the future.

Conclusions and perspectives

USP9X interacts with an extensive range of substrates and has the potential to regulate multiple signaling and survival pathways with cellular responses. USP9X controls a wide variety of cellular pathological and physiological processes through its DUB activity. Previous studies have reported on the significance of USP9X in the cell cycle, apoptosis and survival, and its crucial functions in cell migration and invasion, and DNA damage repair are mediated predominantly via deubiquitination-mediated regulation of the turnover of certain substrates. However, the roles of USP9X in tumor development are complex, and contradictory functions have been identified in different developmental contexts, wherein USP9X displays both oncogenic activity and tumor suppressor functions. In addition to the substrates and downstream signaling pathways regulated by USP9X in tumor progression, the upstream regulatory factors involved in modulating USP9X protein activation in carcinogenesis also merit further exploration. In the future, the development of specific novel inhibitors against USP9X may be a successful strategy for tumor therapy.

Acknowledgements

Not applicable.

Funding

This study was supported by grants from the National Natural Science Foundation of China (grant no. 82102816), the Zhejiang Provincial Natural Science Foundation of China (grant no. LQ22H160014) and Zhejiang Provincial People's Hospital Scientific Research Foundation for The Excellent Youth (grant no. ZRY2020B003).

Availability of data and materials

Not applicable.

Authors' contributions

YH conceived and designed the study. YM, YH, ZQ, CH and SY prepared the figures and tables. YH, YM and LY wrote and edited the manuscript. ZQ, CH and SY revised the manuscript. All authors read and approved the final version of the manuscript. 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.

References

1 

Ermolaeva M, Neri F, Ori A and Rudolph KL: Cellular and epigenetic drivers of stem cell ageing. Nat Rev Mol Cell Biol. 19:594–610. 2018. View Article : Google Scholar : PubMed/NCBI

2 

Holguin-Cruz JA, Foster LJ and Gsponer J: Where protein structure and cell diversity meet. Trends Cell Biol. 32:996–1007. 2022. View Article : Google Scholar : PubMed/NCBI

3 

Toyama BH and Hetzer MW: Protein homeostasis: Live long, won't prosper. Nat Rev Mol Cell Biol. 14:55–61. 2013. View Article : Google Scholar : PubMed/NCBI

4 

Beynon RJ and Bond JS: Catabolism of intracellular protein: Molecular aspects. Am J Physiol. 251:141–152. 1986. View Article : Google Scholar : PubMed/NCBI

5 

Komander D and Rape M: The ubiquitin code. Annu Rev Biochem. 81:203–229. 2012. View Article : Google Scholar : PubMed/NCBI

6 

Li X, Elmira E, Rohondia S, Wang J, Liu J and Dou QP: A patent review of the ubiquitin ligase system: 2015–2018. Expert Opin Ther Pat. 28:919–937. 2018. View Article : Google Scholar : PubMed/NCBI

7 

Jang HH: Regulation of protein degradation by proteasomes in cancer. J Cancer Prev. 23:153–161. 2018. View Article : Google Scholar : PubMed/NCBI

8 

Becker JR, Clifford G, Bonnet C, Groth A, Wilson MD and Chapman JR: BARD1 reads H2A lysine 15 ubiquitination to direct homologous recombination. Nature. 596:433–437. 2021. View Article : Google Scholar : PubMed/NCBI

9 

Roberts JZ, Crawford N and Longley DB: The role of ubiquitination in apoptosis and necroptosis. Cell Death Differ. 29:272–284. 2022. View Article : Google Scholar : PubMed/NCBI

10 

Sun T, Liu Z and Yang Q: The role of ubiquitination and deubiquitination in cancer metabolism. Mol Cancer. 19:1462020. View Article : Google Scholar : PubMed/NCBI

11 

Schmidt MF, Gan ZY, Komander D and Dewson G: Ubiquitin signalling in neurodegeneration: Mechanisms and therapeutic opportunities. Cell Death Differ. 28:570–590. 2021. View Article : Google Scholar : PubMed/NCBI

12 

Fhu CW and Ali A: Dysregulation of the ubiquitin proteasome system in human malignancies: A window for therapeutic intervention. Cancers (Basel). 13:15132021. View Article : Google Scholar : PubMed/NCBI

13 

Grabbe C, Husnjak K and Dikic I: The spatial and temporal organization of ubiquitin networks. Nat Rev Mol Cell Biol. 12:295–307. 2011. View Article : Google Scholar : PubMed/NCBI

14 

Shimizu K, Gi M, Suzuki S, North BJ, Watahiki A, Fukumoto S, Asara JM, Tokunaga F, Wei W and Inuzuka H: Interplay between protein acetylation and ubiquitination controls MCL1 protein stability. Cell Rep. 37:1099882021. View Article : Google Scholar : PubMed/NCBI

15 

Murtaza M, Jolly LA, Gecz J and Wood SA: La FAM fatale: USP9X in development and disease. Cell Mol Life Sci. 72:2075–2089. 2015. View Article : Google Scholar : PubMed/NCBI

16 

Johnson BV, Kumar R, Oishi S, Alexander S, Kasherman M, Vega MS, Ivancevic A, Gardner A, Domingo D, Corbett M, et al: Partial loss of USP9X function leads to a male neurodevelopmental and behavioral disorder converging on transforming growth factor beta signaling. Biol Psychiatry. 87:100–112. 2020. View Article : Google Scholar : PubMed/NCBI

17 

Delbridge ARD, Kueh AJ, Ke F, Zamudio NM, El-Saafin F, Jansz N, Wang GY, Iminitoff M, Beck T, Haupt S, et al: Loss of p53 causes stochastic aberrant x-chromosome inactivation and female-specific neural tube defects. Cell Rep. 27:442–454. 2019. View Article : Google Scholar : PubMed/NCBI

18 

Homan CC, Kumar R, Nguyen LS, Haan E, Raymond FL, Abidi F, Raynaud M, Schwartz CE, Wood SA, Gecz J, et al: Mutations in USP9X are associated with X-linked intellectual disability and disrupt neuronal cell migration and growth. Am J Hum Genet. 94:470–478. 2014. View Article : Google Scholar : PubMed/NCBI

19 

Wang B, Tang X, Yao L, Wang Y, Chen Z, Li M, Wu N, Wu D, Dai X, Jiang H and Ai D: Disruption of USP9X in macrophages promotes foam cell formation and atherosclerosis. J Clin Invest. 132:e1542172022. View Article : Google Scholar : PubMed/NCBI

20 

Greenhill C: Pancreatic cancer: USP9X can be used to predict pancreatic cancer outcomes. Nat Rev Gastroenterol Hepatol. 9:3022012. View Article : Google Scholar : PubMed/NCBI

21 

Jones MH, Furlong RA, Burkin H, Chalmers IJ, Brown GM, Khwaja O and Affara NA: The Drosophila developmental gene fat facets has a human homologue in Xp11.4 which escapes X-inactivation and has related sequences on Yq11.2. Hum Mol Gene. 5:1695–1701. 1996. View Article : Google Scholar : PubMed/NCBI

22 

Wood SA, Pascoe WS, Ru K, Yamada T, Hirchenhain J, Kemler R and Mattick JS: Cloning and expression analysis of a novel mouse gene with sequence similarity to the Drosophila fat facets gene. Mech Dev. 63:29–38. 1997. View Article : Google Scholar : PubMed/NCBI

23 

Chen X, Overstreet E, Wood SA and Fischer JA: On the conservation of function of the Drosophila fat facets deubiquitinating enzyme and Fam, its mouse homolog. Dev Genes Evol. 210:603–610. 2000. View Article : Google Scholar : PubMed/NCBI

24 

Fischer-Vize JA, Rubin GM and Lehmann R: The fat facets gene is required for Drosophila eye and embryo development. Development. 116:985–1000. 1992. View Article : Google Scholar : PubMed/NCBI

25 

Khut PY, Tucker B, Lardelli M and Wood SA: Evolutionary and expression analysis of the zebrafish deubiquitylating enzyme, usp9. Zebrafish. 4:95–101. 2007. View Article : Google Scholar : PubMed/NCBI

26 

Nijman SM, Luna-Vargas MP, Velds A, Brummelkamp TR, Dirac AM, Sixma TK and Bernards R: A genomic and functional inventory of deubiquitinating enzymes. Cell. 123:773–786. 2005. View Article : Google Scholar : PubMed/NCBI

27 

Amerik AY and Hochstrasser M: Mechanism and function of deubiquitinating enzymes. Biochim Biophys Acta. 1695:189–207. 2004. View Article : Google Scholar : PubMed/NCBI

28 

Paudel P, Zhang Q, Leung C, Greenberg HC, Guo Y, Chern YH, Dong A, Li Y, Vedadi M, Zhuang Z and Tong Y: Crystal structure and activity-based labeling reveal the mechanisms for linkage-specific substrate recognition by deubiquitinase USP9X. Proc Natl Acad Sci USA. 116:7288–7297. 2019. View Article : Google Scholar : PubMed/NCBI

29 

Marx C, Held JM, Gibson BW and Benz CC: ErbB2 trafficking and degradation associated with K48 and K63 polyubiquitination. Cancer Res. 70:3709–3717. 2010. View Article : Google Scholar : PubMed/NCBI

30 

Al-Hakim AK, Zagorska A, Chapman L, Deak M, Peggie M and Alessi DR: Control of AMPK-related kinases by USP9X and atypical Lys(29)/Lys(33)-linked polyubiquitin chains. Biochem J. 411:249–260. 2008. View Article : Google Scholar : PubMed/NCBI

31 

Dupont S, Mamidi A, Cordenonsi M, Montagner M, Zacchigna L, Adorno M, Martello G, Stinchfield MJ, Soligo S, Morsut L, et al: FAM/USP9×, a deubiquitinating enzyme essential for TGFbeta signaling, controls Smad4 monoubiquitination. Cell. 136:123–135. 2009. View Article : Google Scholar : PubMed/NCBI

32 

Mouchantaf R, Azakir BA, McPherson PS, Millard SM, Wood SA and Angers A: The ubiquitin ligase itch is auto-ubiquitylated in vivo and in vitro but is protected from degradation by interacting with the deubiquitylating enzyme FAM/USP9X. J Biol Chem. 281:38738–38747. 2006. View Article : Google Scholar : PubMed/NCBI

33 

Taya S, Yamamoto T, Kano K, Kawano Y, Iwamatsu A, Tsuchiya T, Tanaka K, Kanai-Azuma M, Wood SA, Mattick JS and Kaibuchi K: The Ras target AF-6 is a substrate of the fam deubiquitinating enzyme. J Cell Biol. 142:1053–1062. 1998. View Article : Google Scholar : PubMed/NCBI

34 

Vong QP, Cao K, Li HY, Iglesias PA and Zheng Y: Chromosome alignment and segregation regulated by ubiquitination of survivin. Science. 310:1499–1504. 2005. View Article : Google Scholar : PubMed/NCBI

35 

Narayanan N, Wang Z, Li L and Yang Y: Arginine methylation of USP9X promotes its interaction with TDRD3 and its anti-apoptotic activities in breast cancer cells. Cell Discov. 3:160482017. View Article : Google Scholar : PubMed/NCBI

36 

Murray RZ, Jolly LA and Wood SA: The FAM deubiquitylating enzyme localizes to multiple points of protein trafficking in epithelia, where it associates with E-cadherin and beta-catenin. Mol Biol Cell. 15:1591–1599. 2004. View Article : Google Scholar : PubMed/NCBI

37 

Theard D, Labarrade F, Partisani M, Milanini J, Sakagami H, Fon EA, Wood SA, Franco M and Luton F: USP9×-mediated deubiquitination of EFA6 regulates de novo tight junction assembly. EMBO J. 29:1499–1509. 2010. View Article : Google Scholar : PubMed/NCBI

38 

Schwickart M, Huang X, Lill JR, Liu J, Ferrando R, French DM, Maecker H, O'Rourke K, Bazan F, Eastham-Anderson J, et al: Deubiquitinase USP9X stabilizes MCL1 and promotes tumour cell survival. Nature. 463:103–107. 2010. View Article : Google Scholar : PubMed/NCBI

39 

Trinkle-Mulcahy L, Boulon S, Lam YW, Urcia R, Boisvert FM, Vandermoere F, Morrice NA, Swift S, Rothbauer U, Leonhardt H and Lamond A: Identifying specific protein interaction partners using quantitative mass spectrometry and bead proteomes. J Cell Biol. 183:223–239. 2008. View Article : Google Scholar : PubMed/NCBI

40 

Li X, Song N, Liu L, Liu X, Ding X, Song X, Yang S, Shan L, Zhou X, Su D, et al: USP9X regulates centrosome duplication and promotes breast carcinogenesis. Nat Commun. 8:148662017. View Article : Google Scholar : PubMed/NCBI

41 

Engel K, Rudelius M, Slawska J, Jacobs L, Abhari BA, Altmann B, Kurutz J, Rathakrishnan A, Fernandez-Saiz V, Brunner A, et al: USP9X stabilizes XIAP to regulate mitotic cell death and chemoresistance in aggressive B-cell lymphoma. EMBO Mol Med. 8:851–862. 2016. View Article : Google Scholar : PubMed/NCBI

42 

Grou CP, Francisco T, Rodrigues TA, Freitas MO, Pinto MP, Carvalho AF, Domingues P, Wood SA, Rodriguez-Borges JE, Sa-Miranda C, et al: Identification of ubiquitin-specific protease 9X (USP9X) as a deubiquitinase acting on ubiquitin-peroxin 5 (PEX5) thioester conjugate. J Biol Chem. 287:12815–12827. 2012. View Article : Google Scholar : PubMed/NCBI

43 

Akiyama H, Umezawa Y, Ishida S, Okada K, Nogami A and Miura O: Inhibition of USP9X induces apoptosis in FLT3-ITD-positive AML cells cooperatively by inhibiting the mutant kinase through aggresomal translocation and inducing oxidative stress. Cancer Lett. 453:84–94. 2019. View Article : Google Scholar : PubMed/NCBI

44 

Wang P, Wang J, Yao S, Cui M, Cheng Y, Liu W, Gao Z, Hu J, Zhang J and Zhang H: Deubiquitinase USP9X stabilizes RNA m(6)A demethylase ALKBH5 and promotes acute myeloid leukemia cell survival. J Biol Chem. 299:1050552023. View Article : Google Scholar : PubMed/NCBI

45 

Akiyama H, Umezawa Y, Watanabe D, Okada K, Ishida S, Nogami A and Miura O: Inhibition of USP9X downregulates JAK2-V617F and induces apoptosis synergistically with BH3 mimetics preferentially in ruxolitinib-persistent JAK2-V617F-positive leukemic cells. Cancers (Basel). 12:4062020. View Article : Google Scholar : PubMed/NCBI

46 

Wu Y, Yu X, Yi X, Wu K, Dwabe S, Atefi M, Elshimali Y, Kemp KT II, Bhat K, Haro J, et al: Aberrant phosphorylation of SMAD4 Thr277-mediated USP9×-SMAD4 interaction by free fatty acids promotes breast cancer metastasis. Cancer Res. 77:1383–1394. 2017. View Article : Google Scholar : PubMed/NCBI

47 

Li L, Liu T, Li Y, Wu C, Luo K, Yin Y, Chen Y, Nowsheen S, Wu J, Lou Z and Yuan J: The deubiquitinase USP9X promotes tumor cell survival and confers chemoresistance through YAP1 stabilization. Oncogene. 37:2422–2431. 2018. View Article : Google Scholar : PubMed/NCBI

48 

Guan T, Yang X, Liang H, Chen J, Chen Y, Zhu Y and Liu T: Deubiquitinating enzyme USP9X regulates metastasis and chemoresistance in triple-negative breast cancer by stabilizing Snail1. J Cell Physiol. 237:2992–3000. 2022. View Article : Google Scholar : PubMed/NCBI

49 

Lu H, Lyu Y, Tran L, Lan J, Xie Y, Yang Y, Murugan NL, Wang YJ and Semenza GL: HIF-1 recruits NANOG as a coactivator for TERT gene transcription in hypoxic breast cancer stem cells. Cell Rep. 36:1097572021. View Article : Google Scholar : PubMed/NCBI

50 

Jie X, Fong WP, Zhou R, Zhao Y, Zhao Y, Meng R, Zhang S, Dong X, Zhang T, Yang K, et al: USP9X-mediated KDM4C deubiquitination promotes lung cancer radioresistance by epigenetically inducing TGF-β2 transcription. Cell Death Differ. 28:2095–2111. 2021. View Article : Google Scholar : PubMed/NCBI

51 

Chen X, Yu C, Gao J, Zhu H, Cui B, Zhang T, Zhou Y, Liu Q, He H, Xiao R, et al: A novel USP9X substrate TTK contributes to tumorigenesis in non-small-cell lung cancer. Theranostics. 8:2348–2360. 2018. View Article : Google Scholar : PubMed/NCBI

52 

Kushwaha D, O'Leary C, Cron KR, Deraska P, Zhu K, D'Andrea AD and Kozono D: USP9X inhibition promotes radiation-induced apoptosis in non-small cell lung cancer cells expressing mid-to-high MCL1. Cancer Biol Ther. 16:392–401. 2015. View Article : Google Scholar : PubMed/NCBI

53 

Sulkshane P, Pawar SN, Waghole R, Pawar SS, Rajput P, Uthale A, Oak S, Kalkar P, Wani H, Patil R, et al: Elevated USP9X drives early-to-late-stage oral tumorigenesis via stabilisation of anti-apoptotic MCL-1 protein and impacts outcome in oral cancers. Br J Cancer. 125:547–560. 2021. View Article : Google Scholar : PubMed/NCBI

54 

Cheng P, Wang J, Waghmare I, Sartini S, Coviello V, Zhang Z, Kim SH, Mohyeldin A, Pavlyukov MS, Minata M, et al: FOXD1-ALDH1A3 signaling is a determinant for the self-renewal and tumorigenicity of mesenchymal glioma stem cells. Cancer Res. 76:7219–7230. 2016. View Article : Google Scholar : PubMed/NCBI

55 

Khan OM, Carvalho J, Spencer-Dene B, Mitter R, Frith D, Snijders AP, Wood SA and Behrens A: The deubiquitinase USP9X regulates FBW7 stability and suppresses colorectal cancer. J Clin Invest. 128:1326–1337. 2018. View Article : Google Scholar : PubMed/NCBI

56 

Potu H, Peterson LF, Kandarpa M, Pal A, Sun H, Durham A, Harms PW, Hollenhorst PC, Eskiocak U, Talpaz M and Donato NJ: Usp9× regulates Ets-1 ubiquitination and stability to control NRAS expression and tumorigenicity in melanoma. Nat Commun. 8:144492017. View Article : Google Scholar : PubMed/NCBI

57 

Chen W, Song J, Liu S, Tang B, Shen L, Zhu J, Fang S, Wu F, Zheng L, Qiu R, et al: USP9X promotes apoptosis in cholangiocarcinoma by modulation expression of KIF1Bbeta via deubiquitinating EGLN3. J Biomed Sci. 28:442021. View Article : Google Scholar : PubMed/NCBI

58 

Czabotar PE, Lessene G, Strasser A and Adams JM: Control of apoptosis by the BCL-2 protein family: Implications for physiology and therapy. Nat Rev Mol Cell Biol. 15:49–63. 2014. View Article : Google Scholar : PubMed/NCBI

59 

Nagai H, Noguchi T, Homma K, Katagiri K, Takeda K, Matsuzawa A and Ichijo H: Ubiquitin-like sequence in ASK1 plays critical roles in the recognition and stabilization by USP9X and oxidative stress-induced cell death. Mol Cell. 36:805–818. 2009. View Article : Google Scholar : PubMed/NCBI

60 

Huntwork-Rodriguez S, Wang B, Watkins T, Ghosh AS, Pozniak CD, Bustos D, Newton K, Kirkpatrick DS and Lewcock JW: JNK-mediated phosphorylation of DLK suppresses its ubiquitination to promote neuronal apoptosis. J Cell Biol. 202:747–763. 2013. View Article : Google Scholar : PubMed/NCBI

61 

Lai MC, Chen YP, Li DA, Yu JS, Hung HY and Tarn WY: DDX3 interacts with USP9X and participates in deubiquitination of the anti-apoptotic protein MCL1. FEBS J. 289:1043–1061. 2022. View Article : Google Scholar : PubMed/NCBI

62 

Hogh-Binder SA, Klein D, Wolfsperger F, Huber SM, Hennenlotter J, Stenzl A and Rudner J: Protein levels of anti-apoptotic Mcl-1 and the deubiquitinase USP9× are cooperatively upregulated during prostate cancer progression and limit response of prostate cancer cells to radiotherapy. Cancers (Basel). 15:24962023. View Article : Google Scholar : PubMed/NCBI

63 

Sun H, Kapuria V, Peterson LF, Fang D, Bornmann WG, Bartholomeusz G, Talpaz M and Donato NJ: Bcr-Abl ubiquitination and Usp9× inhibition block kinase signaling and promote CML cell apoptosis. Blood. 117:3151–3162. 2011. View Article : Google Scholar : PubMed/NCBI

64 

Kapuria V, Peterson LF, Fang D, Bornmann WG, Talpaz M and Donato NJ: Deubiquitinase inhibition by small-molecule WP1130 triggers aggresome formation and tumor cell apoptosis. Cancer Res. 70:9265–9276. 2010. View Article : Google Scholar : PubMed/NCBI

65 

Karpel-Massler G, Ishida CT, Bianchetti E, Shu C, Perez-Lorenzo R, Horst B, Banu M, Roth KA, Bruce JN, Canoll P, et al: Inhibition of mitochondrial matrix chaperones and antiapoptotic Bcl-2 family proteins empower antitumor therapeutic responses. Cancer Res. 77:3513–3526. 2017. View Article : Google Scholar : PubMed/NCBI

66 

Kim S, Woo SM, Min KJ, Seo SU, Lee TJ, Kubatka P, Kim DE and Kwon TK: WP1130 enhances TRAIL-induced apoptosis through USP9X-dependent miR-708-mediated downregulation of c-FLIP. Cancers (Basel). 11:3442019. View Article : Google Scholar : PubMed/NCBI

67 

Liu Y, Xu X, Lin P, He Y, Zhang Y, Cao B, Zhang Z, Sethi G, Liu J, Zhou X and Mao X: Inhibition of the deubiquitinase USP9× induces pre-B cell homeobox 1 (PBX1) degradation and thereby stimulates prostate cancer cell apoptosis. J Biol Chem. 294:4572–4582. 2019. View Article : Google Scholar : PubMed/NCBI

68 

Dominguez-Brauer C, Thu KL, Mason JM, Blaser H, Bray MR and Mak TW: Targeting mitosis in cancer: Emerging strategies. Mol Cell. 60:524–536. 2015. View Article : Google Scholar : PubMed/NCBI

69 

Motofei IG: Biology of cancer: Understanding the supracellular control of mitosis in physiological processes and malignancy. Semin Cancer Biol. 92:42–44. 2023. View Article : Google Scholar : PubMed/NCBI

70 

Schoonen PM, Talens F, Stok C, Gogola E, Heijink AM, Bouwman P, Foijer F, Tarsounas M, Blatter S, Jonkers J, et al: Progression through mitosis promotes PARP inhibitor-induced cytotoxicity in homologous recombination-deficient cancer cells. Nat Commun. 8:159812017. View Article : Google Scholar : PubMed/NCBI

71 

Skowyra A, Allan LA, Saurin AT and Clarke PR: USP9X limits mitotic checkpoint complex turnover to strengthen the spindle assembly checkpoint and guard against chromosomal instability. Cell Rep. 23:852–865. 2018. View Article : Google Scholar : PubMed/NCBI

72 

Dietachmayr M, Rathakrishnan A, Karpiuk O, von Zweydorf F, Engleitner T, Fernandez-Saiz V, Schenk P, Ueffing M, Rad R, Eilers M, et al: Antagonistic activities of CDC14B and CDK1 on USP9X regulate WT1-dependent mitotic transcription and survival. Nat Commun. 11:12682020. View Article : Google Scholar : PubMed/NCBI

73 

Wang Q, Tang Y, Xu Y, Xu S, Jiang Y, Dong Q, Zhou Y and Ge W: The X-linked deubiquitinase USP9X is an integral component of centrosome. J Biol Chem. 292:12874–12884. 2017. View Article : Google Scholar : PubMed/NCBI

74 

Clancy A, Heride C, Pinto-Fernandez A, Elcocks H, Kallinos A, Kayser-Bricker KJ, Wang W, Smith V, Davis S, Fessler S, et al: The deubiquitylase USP9X controls ribosomal stalling. J Cell Biol. 220:2020042112021. View Article : Google Scholar

75 

Weimer JM and Anton ES: Doubling up on microtubule stabilizers: Synergistic functions of doublecortin-like kinase and doublecortin in the developing cerebral cortex. Neuron. 49:3–4. 2006. View Article : Google Scholar : PubMed/NCBI

76 

Gleeson JG, Lin PT, Flanagan LA and Walsh CA: Doublecortin is a microtubule-associated protein and is expressed widely by migrating neurons. Neuron. 23:257–271. 1999. View Article : Google Scholar : PubMed/NCBI

77 

Kharitidi D, Apaja PM, Manteghi S, Suzuki K, Malitskaya E, Roldan A, Gingras MC, Takagi J, Lukacs GL and Pause A: Interplay of endosomal pH and ligand occupancy in integrin alpha5beta1 ubiquitination, endocytic sorting, and cell migration. Cell Rep. 13:599–609. 2015. View Article : Google Scholar : PubMed/NCBI

78 

Zhu H, Kavsak P, Abdollah S, Wrana JL and Thomsen GH: A SMAD ubiquitin ligase targets the BMP pathway and affects embryonic pattern formation. Nature. 400:687–693. 1999. View Article : Google Scholar : PubMed/NCBI

79 

Ebisawa T, Fukuchi M, Murakami G, Chiba T, Tanaka K, Imamura T and Miyazono K: Smurf1 interacts with transforming growth factor-beta type I receptor through Smad7 and induces receptor degradation. J Biol Chem. 276:12477–12480. 2001. View Article : Google Scholar : PubMed/NCBI

80 

Xie Y, Avello M, Schirle M, McWhinnie E, Feng Y, Bric-Furlong E, Wilson C, Nathans R, Zhang J, Kirschner MW, et al: Deubiquitinase FAM/USP9X interacts with the E3 ubiquitin ligase SMURF1 protein and protects it from ligase activity-dependent self-degradation. J Biol Chem. 288:2976–2985. 2013. View Article : Google Scholar : PubMed/NCBI

81 

Wang T, Jing B, Sun B, Liao Y, Song H, Xu D, Guo W, Li K, Hu M, Liu S, et al: Stabilization of PTGES by deubiquitinase USP9X promotes metastatic features of lung cancer via PGE(2) signaling. Am J Cancer Res. 9:1145–1160. 2019.PubMed/NCBI

82 

Harrigan JA, Jacq X, Martin NM and Jackson SP: Deubiquitylating enzymes and drug discovery: Emerging opportunities. Nat Rev Drug Discov. 17:57–78. 2018. View Article : Google Scholar : PubMed/NCBI

83 

Negrini S, Gorgoulis VG and Halazonetis TD: Genomic instability-an evolving hallmark of cancer. Nat Rev Mol Cell Biol. 11:220–228. 2010. View Article : Google Scholar : PubMed/NCBI

84 

Kang JW, Zhan Z, Ji G, Sang Y, Zhou D, Li Y, Feng H and Cheng T: PUMA facilitates EMI1-promoted cytoplasmic Rad51 ubiquitination and inhibits DNA repair in stem and progenitor cells. Signal Transduct Target Ther. 6:1292021. View Article : Google Scholar : PubMed/NCBI

85 

McGarry E, Gaboriau D, Rainey MD, Restuccia U, Bachi A and Santocanale C: The deubiquitinase USP9X maintains DNA replication fork stability and DNA damage checkpoint responses by regulating CLASPIN during S-Phase. Cancer Res. 76:2384–2393. 2016. View Article : Google Scholar : PubMed/NCBI

86 

Wolfsperger F, Hogh-Binder SA, Schittenhelm J, Psaras T, Ritter V, Bornes L, Huber SM, Jendrossek V and Rudner J: Deubiquitylating enzyme USP9× regulates radiosensitivity in glioblastoma cells by Mcl-1-dependent and -independent mechanisms. Cell Death Dis. 7:20392016. View Article : Google Scholar : PubMed/NCBI

87 

Lautenbacher L, Samaras P, Muller J, Grafberger A, Shraideh M, Rank J, Fuchs ST, Schmidt TK, The M, Dallago C, et al: ProteomicsDB: Toward a FAIR open-source resource for life-science research. Nucleic Acids Res. 50:D1541–D1552. 2022. View Article : Google Scholar : PubMed/NCBI

88 

Zhang C, Cai TY, Zhu H, Yang LQ, Jiang H, Dong XW, Hu YZ, Lin NM, He QJ and Yang B: Synergistic antitumor activity of gemcitabine and ABT-737 in vitro and in vivo through disrupting the interaction of USP9X and Mcl-1. Mol Cancer Ther. 10:1264–1275. 2011. View Article : Google Scholar : PubMed/NCBI

89 

Oosterkamp HM, Hijmans EM, Brummelkamp TR, Canisius S, Wessels LF, Zwart W and Bernards R: USP9X downregulation renders breast cancer cells resistant to tamoxifen. Cancer Res. 74:3810–3820. 2014. View Article : Google Scholar : PubMed/NCBI

90 

Tang Z, Li C, Kang B, Gao G, Li C and Zhang Z: GEPIA: A web server for cancer and normal gene expression profiling and interactive analyses. Nucleic Acids Res. 45:W98–W102. 2017. View Article : Google Scholar : PubMed/NCBI

91 

Zhang C, Qian H, Liu K, Zhao W and Wang L: A feedback loop regulation of LINC01433 and YAP promotes malignant behavior in gastric cancer cells. Onco Targets Ther. 12:7949–7962. 2019. View Article : Google Scholar : PubMed/NCBI

92 

Chen H, Yang F, Li X, Gong ZJ and Wang LW: Long noncoding RNA LNC473 inhibits the ubiquitination of survivin via association with USP9X and enhances cell proliferation and invasion in hepatocellular carcinoma cells. Biochem Biophys Res Commun. 499:702–710. 2018. View Article : Google Scholar : PubMed/NCBI

93 

Xu X, Wang S, Wang H, Pan C, Yang W and Yu J: Hsa_circ_0008434 regulates USP9X expression by sponging miR-6838-5p to promote gastric cancer growth, migration and invasion. BMC Cancer. 21:12892021. View Article : Google Scholar : PubMed/NCBI

94 

Shen G, Lin Y, Yang X, Zhang J, Xu Z and Jia H: MicroRNA-26b inhibits epithelial-mesenchymal transition in hepatocellular carcinoma by targeting USP9X. BMC Cancer. 14:3932014. View Article : Google Scholar : PubMed/NCBI

95 

Chen W, Huang Y, Zhang S, Zheng X, Xie S, Mao J, Cai Y, Lu X, Hu L, Shen J, et al: MicroRNA-212 suppresses nonsmall lung cancer invasion and migration by regulating ubiquitin-specific protease-9. J Cell Biochem. 120:6482–6489. 2019. View Article : Google Scholar : PubMed/NCBI

96 

Chen W, Zhou Y, Zhi X, Ma T, Liu H, Chen BW, Zheng X, Xie S, Zhao B, Feng X, et al: Delivery of miR-212 by chimeric peptide-condensed supramolecular nanoparticles enhances the sensitivity of pancreatic ductal adenocarcinoma to doxorubicin. Biomaterials. 192:590–600. 2019. View Article : Google Scholar : PubMed/NCBI

97 

Guo H, Zhang X, Chen Q, Bao Y, Dong C and Wang X: miR-132 suppresses the migration and invasion of lung cancer cells by blocking USP9X-induced epithelial-mesenchymal transition. Am J Transl Res. 10:224–234. 2018.PubMed/NCBI

98 

Chen E, Li E, Liu H, Zhou Y, Wen L, Wang J, Wang Y, Ye L and Liang T: miR-26b enhances the sensitivity of hepatocellular carcinoma to Doxorubicin via USP9X-dependent degradation of p53 and regulation of autophagy. Int J Biol Sci. 17:781–795. 2021. View Article : Google Scholar : PubMed/NCBI

99 

Madan V, Li J, Zhou S, Teoh WW, Han L, Meggendorfer M, Malcovati L, Cazzola M, Ogawa S, Haferlach T, et al: Distinct and convergent consequences of splice factor mutations in myelodysplastic syndromes. Am J Hematol. 95:133–143. 2020. View Article : Google Scholar : PubMed/NCBI

100 

Aqaqe N, Yassin M, Yassin AA, Ershaid N, Katz-Even C, Zipin-Roitman A, Kugler E, Lechman ER, Gan OI, Mitchell A, et al: An ERG enhancer-based reporter identifies leukemia cells with elevated leukemogenic potential driven by ERG-USP9X feed-forward regulation. Cancer Res. 79:3862–3876. 2019. View Article : Google Scholar : PubMed/NCBI

101 

Shen SM, Zhang C, Ge MK, Dong SS, Xia L, He P, Zhang N, Ji Y, Yang S, Yu Y, et al: PTENalpha and PTENbeta promote carcinogenesis through WDR5 and H3K4 trimethylation. Nat Cell Biol. 21:1436–1448. 2019. View Article : Google Scholar : PubMed/NCBI

102 

Kapoor S, Natarajan K, Baldwin PR, Doshi KA, Lapidus RG, Mathias TJ, Scarpa M, Trotta R, Davila E, Kraus M, et al: Concurrent inhibition of pim and FLT3 kinases enhances apoptosis of FLT3-ITD acute myeloid leukemia cells through increased Mcl-1 proteasomal degradation. Clin Cancer Res. 24:234–247. 2018. View Article : Google Scholar : PubMed/NCBI

103 

Sisoudiya SD, Mishra P, Li H, Schraw JM, Scheurer ME, Salvi S, Doddapaneni H, Muzny D, Mitchell D, Taylor O, et al: Identification of USP9X as a leukemia susceptibility gene. Blood Adv. 7:4563–4575. 2023. View Article : Google Scholar : PubMed/NCBI

104 

Schwartzman O, Savino AM, Gombert M, Palmi C, Cario G, Schrappe M, Eckert C, von Stackelberg A, Huang JY, Hameiri-Grossman M, et al: Suppressors and activators of JAK-STAT signaling at diagnosis and relapse of acute lymphoblastic leukemia in Down syndrome. Proc Natl Acad Sci USA. 114:4030–4039. 2017. View Article : Google Scholar

105 

Moroishi T, Hansen CG and Guan KL: The emerging roles of YAP and TAZ in cancer. Nat Rev Cancer. 15:73–79. 2015. View Article : Google Scholar : PubMed/NCBI

106 

Demark-Wahnefried W, Platz EA, Ligibel JA, Blair CK, Courneya KS, Meyerhardt JA, Ganz PA, Rock CL, Schmitz KH, Wadden T, et al: The role of obesity in cancer survival and recurrence. Cancer Epidemiol Biomarkers Prev. 21:1244–1259. 2012. View Article : Google Scholar : PubMed/NCBI

107 

Jaiswal A, Murakami K, Elia A, Shibahara Y, Done SJ, Wood SA, Donato NJ, Ohashi PS and Reedijk M: Therapeutic inhibition of USP9×-mediated Notch signaling in triple-negative breast cancer. Proc Natl Acad Sci USA. 118:e21015921182021. View Article : Google Scholar : PubMed/NCBI

108 

Yan J, Zhong N, Liu G, Chen K, Liu X, Su L and Singhal S: Usp9×- and Noxa-mediated Mcl-1 downregulation contributes to pemetrexed-induced apoptosis in human non-small-cell lung cancer cells. Cell Death Dis. 5:e13162014. View Article : Google Scholar : PubMed/NCBI

109 

Sung H, Ferlay J, Siegel RL, Laversanne M, Soerjomataram I, Jemal A and Bray F: Global cancer statistics 2020: GLOBOCAN estimates of incidence and mortality worldwide for 36 cancers in 185 countries. CA Cancer J Clin. 71:209–249. 2021. View Article : Google Scholar : PubMed/NCBI

110 

Perez-Mancera PA, Rust AG, van der Weyden L, Kristiansen G, Li A, Sarver AL, Silverstein KA, Grutzmann R, Aust D, Rummele P, et al: The deubiquitinase USP9X suppresses pancreatic ductal adenocarcinoma. Nature. 486:266–270. 2012. View Article : Google Scholar : PubMed/NCBI

111 

Pal A, Dziubinski M, Di Magliano MP, Simeone DM, Owens S, Thomas D, Peterson L, Potu H, Talpaz M and Donato NJ: Usp9× promotes survival in human pancreatic cancer and its inhibition suppresses pancreatic ductal adenocarcinoma in vivo tumor growth. Neoplasia. 20:152–164. 2018. View Article : Google Scholar : PubMed/NCBI

112 

Guo M, Luo G, Jin K, Long J, Cheng H, Lu Y, Wang Z, Yang C, Xu J, Ni Q, et al: Somatic genetic variation in solid pseudopapillary tumor of the pancreas by whole exome sequencing. Int J Mol Sci. 18:812017. View Article : Google Scholar : PubMed/NCBI

113 

Perurena N, Lock R, Davis RA, Raghavan S, Pilla NF, Ng R, Loi P, Guild CJ, Miller AL, Sicinska E, et al: USP9X mediates an acute adaptive response to MAPK suppression in pancreatic cancer but creates multiple actionable therapeutic vulnerabilities. Cell Rep Med. 4:1010072023. View Article : Google Scholar : PubMed/NCBI

114 

Chen Z, Wang HW, Wang S, Fan L, Feng S, Cai X, Peng C, Wu X, Lu J, Chen D, et al: USP9X deubiquitinates ALDH1A3 and maintains mesenchymal identity in glioblastoma stem cells. J Clin Invest. 129:2043–2055. 2019. View Article : Google Scholar : PubMed/NCBI

115 

Nguyen HT, Andrejeva D, Gupta R, Choudhary C, Hong X, Eichhorn PJ, Loya AC and Cohen SM: Deubiquitylating enzyme USP9× regulates hippo pathway activity by controlling angiomotin protein turnover. Cell Discov. 2:160012016. View Article : Google Scholar : PubMed/NCBI

116 

Bartholomeusz G, Talpaz M, Bornmann W, Kong LY and Donato NJ: Degrasyn activates proteasomal-dependent degradation of c-Myc. Cancer Res. 67:3912–3918. 2007. View Article : Google Scholar : PubMed/NCBI

117 

Luo H, Jing B, Xia Y, Zhang Y, Hu M, Cai H, Tong Y, Zhou L, Yang L, Yang J, et al: WP1130 reveals USP24 as a novel target in T-cell acute lymphoblastic leukemia. Cancer Cell Int. 19:562019. View Article : Google Scholar : PubMed/NCBI

118 

Peterson LF, Sun H, Liu Y, Potu H, Kandarpa M, Ermann M, Courtney SM, Young M, Showalter HD, Sun D, et al: Targeting deubiquitinase activity with a novel small-molecule inhibitor as therapy for B-cell malignancies. Blood. 125:3588–3597. 2015. View Article : Google Scholar : PubMed/NCBI

119 

Cui J, Sun W, Hao X, Wei M, Su X, Zhang Y, Su L and Liu X: EHMT2 inhibitor BIX-01294 induces apoptosis through PMAIP1-USP9X-MCL1 axis in human bladder cancer cells. Cancer Cell Int. 15:42015. View Article : Google Scholar : PubMed/NCBI

120 

Boise LH: DUB-ling down on B-cell malignancies. Blood. 125:3522–3523. 2015. View Article : Google Scholar : PubMed/NCBI

121 

Chou DH, Vetere A, Choudhary A, Scully SS, Schenone M, Tang A, Gomez R, Burns SM, Lundh M, Vital T, et al: Kinase-independent small-molecule inhibition of JAK-STAT signaling. J Am Chem Soc. 137:7929–7934. 2015. View Article : Google Scholar : PubMed/NCBI

122 

Lawson AP, Long MJC, Coffey RT, Qian Y, Weerapana E, El Oualid F and Hedstrom L: Naturally occurring isothiocyanates exert anticancer effects by inhibiting deubiquitinating enzymes. Cancer Res. 75:5130–5142. 2015. View Article : Google Scholar : PubMed/NCBI

Related Articles

Journal Cover

December-2023
Volume 26 Issue 6

Print ISSN: 1792-1074
Online ISSN:1792-1082

Sign up for eToc alerts

Recommend to Library

Copy and paste a formatted citation
x
Spandidos Publications style
Meng Y, Hong C, Yang S, Qin Z, Yang L and Huang Y: Roles of USP9X in cellular functions and tumorigenesis (Review). Oncol Lett 26: 506, 2023
APA
Meng, Y., Hong, C., Yang, S., Qin, Z., Yang, L., & Huang, Y. (2023). Roles of USP9X in cellular functions and tumorigenesis (Review). Oncology Letters, 26, 506. https://doi.org/10.3892/ol.2023.14093
MLA
Meng, Y., Hong, C., Yang, S., Qin, Z., Yang, L., Huang, Y."Roles of USP9X in cellular functions and tumorigenesis (Review)". Oncology Letters 26.6 (2023): 506.
Chicago
Meng, Y., Hong, C., Yang, S., Qin, Z., Yang, L., Huang, Y."Roles of USP9X in cellular functions and tumorigenesis (Review)". Oncology Letters 26, no. 6 (2023): 506. https://doi.org/10.3892/ol.2023.14093