ArticlePDF Available

Identification of ubiquitin-specific protease 32 as an oncogene in glioblastoma and the underlying mechanisms

Authors:

Abstract and Figures

Glioblastoma (GBM) patients present poor prognosis. Deubiquitination by deubiquitinating enzymes (DUBs) is a critical process in cancer progression. Ubiquitin-specific proteases (USPs) constitute the largest sub-family of DUBs. Evaluate the role of USP32 in GBM progression and provide a potential target for GBM treatment. Clinical significance of USP32 was investigated using Gene Expression Omnibus databases. Effects of USP32 on cell growth and metastasis were studied in vitro and in vivo. Differentially expressive genes between USP32-knockdown U-87 MG cells and negative control cells were detected using RNA sequencing and used for Gene Ontology and Kyoto Encyclopedia of Genes and Genomic pathway enrichment analyses. Finally, RT-qPCR was used to validate the divergent expression of genes involved in the enriched pathways. USP32 was upregulated in GBM patients, being correlated to poor prognosis. USP32 downregulation inhibited cell growth and metastasis in vitro. Furthermore, USP32 knockdown inhibited tumorigenesis in vivo. In addition, UPS32 was identified as a crucial regulator in different pathways including cell cycle, cellular senescence, DNA replication, base excision repair, and mismatch repair pathways. USP32 acts as an oncogene in GBM through regulating several biological processes/pathways. It could be a potential target for GBM treatment.
This content is subject to copyright. Terms and conditions apply.
1
Vol.:(0123456789)
Scientic Reports | (2022) 12:6445 | https://doi.org/10.1038/s41598-022-09497-y
www.nature.com/scientificreports
Identication of ubiquitin‑specic
protease 32 as an oncogene
in glioblastoma and the underlying
mechanisms
Sifang Chen1, Xi Chen1, Zhangyu Li1, Jianyao Mao1, Weichao Jiang1, Zhi Zhu2, Yukui Li1,
Zhengye Jiang1, Wenpeng Zhao1, Guowei Tan1 & Zhanxiang Wang1,3,4*
Glioblastoma (GBM) patients present poor prognosis. Deubiquitination by deubiquitinating enzymes
(DUBs) is a critical process in cancer progression. Ubiquitin‑specic proteases (USPs) constitute the
largest sub‑family of DUBs. Evaluate the role of USP32 in GBM progression and provide a potential
target for GBM treatment. Clinical signicance of USP32 was investigated using Gene Expression
Omnibus databases. Eects of USP32 on cell growth and metastasis were studied in vitro and in vivo.
Dierentially expressive genes between USP32‑knockdown U‑87 MG cells and negative control cells
were detected using RNA sequencing and used for Gene Ontology and Kyoto Encyclopedia of Genes
and Genomic pathway enrichment analyses. Finally, RT‑qPCR was used to validate the divergent
expression of genes involved in the enriched pathways. USP32 was upregulated in GBM patients,
being correlated to poor prognosis. USP32 downregulation inhibited cell growth and metastasis
in vitro. Furthermore, USP32 knockdown inhibited tumorigenesis in vivo. In addition, UPS32 was
identied as a crucial regulator in dierent pathways including cell cycle, cellular senescence, DNA
replication, base excision repair, and mismatch repair pathways. USP32 acts as an oncogene in GBM
through regulating several biological processes/pathways. It could be a potential target for GBM
treatment.
Glioblastoma (GBM), the most aggressive type of glioma, are characterized by cellular heterogeneity, a diusive
inltration of tumor cells, and the presence of glioma stem-like cells capable of signicantly expanding and
generating new tumors. GBM patients have an average overall survival time of barely more than 15 months14.
Understanding the molecular mechanisms driving malignancy helps in the development of agents specically
targeting the tumor cells or tumor microenvironment5. Despite improvements in therapeutic strategies, GBM
remains a clinical challenge. ere is an urgent need to nd novel regulators or targets for exploring highly
bioactive and brain-penetrating targeted therapies.
Ubiquitination is an ATP-dependent cascade process that ligates ubiquitin to a substrate protein, catalyzed
by the E1, E2, and E3 three-enzyme cascade6. e ubiquitinated proteins can be specially recognized by the 26S
proteasome, and this leads to the proteasomal degradation of these proteins, regulating dierent cell processes.
Many studies have revealed that the ubiquitin–proteasome system is involved in the progression of GBM710. A
siRNA screening analysis showed that 22% of GBM-survival-relevant genes were components of the 20S and 26S
proteasome subunits11. Epidermal growth factor receptor (EGFR) amplication and mutations are commonly
observed in GBM; EGFR stability and downstream signaling are subject to the ubiquitin regulatory network10.
Homologous to E6-AP Carboxyl Terminus (HECT) E3 ligase Smurf2 suppresses TGF-β signaling by targeting
TbR-I for proteasomal degradation, promoting the progression of GBM12. HECT E3 ligase HERC3 play an essen-
tial role in autophagy-induced EMT, resulting in the chemoresistance of GBM13. In GBM, the p53 and c-Myc
levels are regulated by E3 ligases MDM2 and TRIM3, respectively14,15. e ubiquitination is oen antagonized by
deubiquitinating enzymes (DUBs) which remove the ubiquitin chains from ubiquitinated proteins16. More than
100 DUBs have been identied and categorized into 8 subfamilies, of which ubiquitin-specic proteases (USPs)
OPEN
1Department of Neurosurgery, The First Aliated Hospital of Xiamen University, No. 55 Zhenghai Road, Siming
District, Xiamen 361000, Fujian, China. 2Department of Neurosurgery, Heze Municipal Hospital, Heze 274000,
Shandong, China. 3Xiamen Key Laboratory of Brain Center, The First Aliated Hospital of Xiamen University,
Xiamen 361000, Fujian, China. 4Department of Neuroscience, Institute of Neurosurgery, School of Medicine,
Xiamen University, Xiamen 361000, Fujian, China. *email: csfsong@163.com
Content courtesy of Springer Nature, terms of use apply. Rights reserved
2
Vol:.(1234567890)
Scientic Reports | (2022) 12:6445 | https://doi.org/10.1038/s41598-022-09497-y
www.nature.com/scientificreports/
constitute the largest one. DUBs not only function to reverse ubiquitination but are also involved in protein
tracking, chromatin remodeling, cell cycle regulation, base excision repair, mismatch repair, and signaling
pathway modulations, which are tightly associated with the development of cancer17,18. DUBs have emerged
as attractive targets for targeted therapy in cancer19,20 aer the clinical success of proteasomal inhibitors, such
as bortezomib, ixazomib, and carlzomib. NSC 144303 (G5) and NSC 632839 (F6), two DUBs inhibitors, are
under pre-clinical trial6. Several DUBs were reported to regulate multiple cellular processes such as apoptosis,
proliferation, and stemness in GBM7,21.
USP32 is a member of USPs discovered in recent years. Previous studies had shown that USP32 is highly
expressed in breast cancer and promotes the growth of breast tumor cells22. Hu etal. revealed that USP32
knockdown suppresses cellular proliferation and cell metastasis in small cell lung cancer23. e study of Dou
etal. reported that high expression of USP32 is signicantly associated with high T stage and poor prognosis in
gastric cancer patients24. USP32 was also identied as an oncogene in epithelial ovarian25. However, USP32 is still
rarely reported in malignant disease processes, especially in GBM. In this study, USP32 was knocked down in
GBM cells, evaluating the eect of this enzyme on cell growth and metastasis. Tumor xenogra experiments in
nude mice were also performed to validate the role of USP32 in GBM development. Transcriptional sequencing
identied the dierential expressed genes between stably USP32-knockdown U-87 MG cells and negative control
cells. Functional annotations, including Gene Ontology (GO) and Kyoto Encyclopedia of Genes and Genomes
(KEGG) pathway enrichment analyses, were used to uncover the underlying mechanism of USP32 on promoting
the progression of GBM. Our study may provide a potential target for GBM treatment.
Methods
Cell lines and cell culture. 293T, SVG p12, U-87 MG, U-118 MG, U-251 MG, T98G, and A172 cells were
purchased from Xiamen Immocell Biotechnology Co., Ltd (Xiamen, Fujian, China). All cells were maintained
using Dulbecco’s modied Eagle’s medium (DMEM, IMMOCELL) containing 15% fetal bovine serum (FBS,
Gibco) at 37°C in an incubator with an atmosphere containing 5% CO2 and 21% O2.
High‑content screening. Small interfering RNAs (siRNAs) targeting USPs were obtained from GeneP-
harma (Shanghai, China). Table1 describes these siRNAs. U-87 MG and U-251 MG cells were seeded into
96-well plates at a density of 1 × 104 per well. Lipofectamine RNAiMAX (Life Technology, Carlsbad, CA, USA)
was used for siRNA transfection at 37°C. Aer a 36h of transfection, the cells were incubated with 10μL/well
CCK-8 solution (Yeasen, Shanghai, China). e cell viability was evaluated 2h later by detecting the optical
density at 450nm (OD450) using a microplate reader (Molecular Devices, San Francisco, CA, USA).
Data mining from Gene Expression Omnibus (GEO) databases. Dataset GSE59612 (normal 17,
GBM 39) was downloaded from the GEO website (https:// www. ncbi. nlm. nih. gov/ geo/) to evaluate the dier-
ential expression of USP32 between normal tissues and tumor tissues. e mRNA expression data and survival
information of GBM patients in datasets GSE74187 (GBM 60) and GSE83300 (GBM 50) were also downloaded.
e association between USP32 expression level and prognosis was analyzed. GBM patients were divided into
two groups (high and low) using the optimal cut-o value of USP32 expression level, which was determined
using the surv_cutpoint function of the R package survminer via RStudio soware (version 2021.09.0 + 351,
https:// www. rstud io. com/).
RT‑qPCR. Total RNA from U-87 MG was obtained using an RNA Extraction Kit (Vazyme, Nanjing, Jiangsu,
China) and reverse transcribed into cDNA using HiScript II Reverse Transcriptase (Vazyme). One hundred
nanogram of cDNA was used for qPCR per well. qPCR was performed using a Bio-Rad CFX96 system (Bio-Rad
Laboratories, Hercules, CA, USA) with an AceQ qPCR SYBR Green Master Mix Kit (Vazyme). e thermocy-
cling condition was 96°C for 5min, followed by 96°C for 15s, 60°C for 25s, and 72°C for 20s, for 45 cycles.
e 2−ΔΔCt method was used to calculate the relative mRNA levels, which was calibrated to 18S RNA. e primers
for RT-qPCR are shown in Table2.
Western blotting. U-251 MG and U-87 MG cells were lysed using RIPA buer (Vazyme). e protein
concentrations of samples were measured using a bicinchoninic acid (BCA) protein quantication kit (Abcam,
Shanghai, China). Samples (12μg/lane) were loaded into a 12% SDS-PAGE gel for electrophoresis and then
transferred onto a PVDF membrane (Roche, Basel, Switzerland). e membrane was incubated with primary
antibody Anti-USP32 (1:1000, CAT#ab251903, Abcam) or Anti-GAPDH (1:2000, CAT#ab9485, Abcam) at
4°C overnight, followed by incubation with secondary antibody HRP-conjugated Goat Anti-Rabbit IgG H&L
(1:2000, CAT#ab6721, Abcam) at 28°C for 30min. e signals were visualized using the ECL detection system
(ermo Fisher Scientic, Waltham, MA, USA) and quantied by densitometry using Image J v1.48u.
USP32 knockdown by transfection. U-251 MG and U-87 MG cells were seeded into 6-well plates at
a density of 5 × 105 per well and transfected with 200pmol/well siRNA NC, siUSP32-2261, or siUSP32-386
using LipofectamineRNAiMAX. e cells were harvested and used for further experiments aer incubation for
6–48h at 37°C.
CCK‑8 assay for cell viability. U-251 MG and U-87 MG cells were tripsinized aer a 24h of transfection
and then seeded into 96-well plates at a density of 5 × 103 per well. At dierent time points (0, 24, 48, 72h), 10μL/
well CCK-8 solution was added into cells and the cell viability was evaluated by detecting OD450.
Content courtesy of Springer Nature, terms of use apply. Rights reserved
3
Vol.:(0123456789)
Scientic Reports | (2022) 12:6445 | https://doi.org/10.1038/s41598-022-09497-y
www.nature.com/scientificreports/
siRNAs Sense (5–3) Antisense (5–3)
siUSP1-1780 GGU UAA AGU CUG CAA CUA AUU UUA GUU GCA GAC UUU AAC CUU
siUSP1-1501 GCA UAG AGA UGG ACA GUA UUU AUA CUG UCC AUC UCU AUG CUU
siUSP2-1339 GCG CUU UGU UGG CUA UAA UUU AUU AUA GCC AAC AAA GCG CUU
siUSP2-1921 CCU GUA CGC UGU GUC CAA UUU AUU GGA CAC AGC GUA CAG GUU
siUSP3-1109 GGG ACA GAA UCU AGA AAG UUU ACU UUC UAG AUU CUG UCC CUU
siUSP3-1569 GCU GGU UCC ACU UCA AUG AUU UCA UUG AAG UGG AAC CAG CUU
siUSP4-377 GCG UGG AAU AAA CUA CUA AUU UUA GUA GUU UAU UCC ACG CUU
siUSP4-860 GCA AAU GGU GAU AGC ACU AUU UAG UGC UAU CAC CAU UUG CUU
siUSP5-822 GGA GCU GAC GUG UAC UCA UUU AUG AGU ACA CGU CAG CUC CUU
siUSP5-1265 GCC AGA ACA GAA GGA AGU UUU AAC UUC CUU CUG UUC UGG CUU
siUSP6-2274 GGA AGG ACA UAC UUA UGA AUU UUC AUA AGU AUG UCC UUC CUU
siUSP6-2494 GCA CAG UAG CAA ACU CAU AUU UAU GAG UUU GCU ACU GUG CUU
siUSP7-2625 GUG GUU ACG UUA UCA AAU AUU UAU UUG AUA ACG UAA CCA CUU
siUSP7-603 GCA GUG CUG AAG AUA AUA AUU UUA UUA UCU UCA GCA CUG CUU
siUSP8-873 CCA AAG AGA AAG GAG CAA UUU AUU GCU CCU UUC UCU UUG GUU
siUSP8-3569 GCA AGA CAA CGG UGG UUU AUU UAA ACC ACC GUU GUC UUG CUU
siUSP9X-7849 GGG CAA UGG AGA UCU UAA AUU UUU AAG AUC UCC AUU GCC CUU
siUSP9X-2333 CCC GCA CUG AAA CAA AUU AUU UAA UUU GUU UCA GUG CGG GUU
siUSP9Y-2805 CCU UGC AAC CUA CAU GAA UUU AUU CAU GUA GGU UGC AAG GUU
siUSP9Y-8135 GCA GUU GUC CUG UUG CUU AUU UAA GCA ACA GGA CAA CUG CUU
siUSP10-630 GCU UUG GAU GGA AGU UCU AUU UAG AAC UUC CAU CCA AAG CUU
siUSP10-1175 GCA CAC CAC GGA AAG CAU AUU UAU GCU UUC CGU GGU GUG CUU
siUSP11-1415 GCA AUG UAU CUG UGA CCU UUU AAG GUC ACA GAU ACA UUG CUU
siUSP11-2088 CCU CCU GGA CAA UUG CCU UUU AAG GCA AUU GUC CAG GAG GUU
siUSP12-314 CUU CGG CAU UAG AGA AAG AUU UCU UUC UCU AAU GCC GAA GUU
siUSP12-648 CCU ACU AAA UAC AAU UGC UUU AGC AAU UGU AUU UAG UAG GUU
siUSP13-1685 CGU GCC AAG AUA CCA UUU AUU UAA AUG GUA UCU UGG CAC GUU
siUSP13-2004 GCC UGA UGA ACC AAU UGA UUU AUC AAU UGG UUC AUC AGG CUU
siUSP14-655 GCU UCA GCG CAG UAU AUU AUU UAA UAU ACU GCG CUG AAG CUU
siUSP14-1632 GCA UAU CGC UUA CGU UCU AUU UAG AAC GUA AGC GAU AUG CUU
siUSP15-249 GGA ACA CCU UAU UGA UGA AUU UUC AUC AAU AAG GUG UUC CUU
siUSP15-1150 GCA GAU GGA AGG CCA GAU AUU UAU CUG GCC UUC CAU CUG CUU
siUSP16-331 GGA AUG GAA UAU CUG CCA AUU UUG GCA GAU AUU CCA UUC CUU
siUSP16-469 GCA UGC CUU GAA GCA CUA UUU AUA GUG CUU CAA GGC AUG CUU
siUSP17-1431 CCA UCA UCC UGA ACA GCA AUU UUG CUG UUC AGG AUG AUG GUU
siUSP17-1538 GGA GAU CCA AAG GGA AGA AUU UUC UUC CCU UUG GAU CUC CUU
siUSP18-527 GCU GCC UUA ACU CCU UGA UUU AUC AAG GAG UUA AGG CAG CUU
siUSP18-1418 CUG CAU AUC UUC UGG UUU AUU UAA ACC AGA AGA UAU GCA GUU
siUSP19-2439 GCA UUC AGA ACA AGC CCU AUU UAG GGC UUG UUC UGA AUG CUU
siUSP19-2518 GCG GCA CAA GAU GAG GAA UUU AUU CCU CAU CUU GUG CCG CUU
siUSP20-249 CCA UAG GAG AGG UGA CCA AUU UUG GUC ACC UCU CCU AUG GUU
siUSP20-1041 GCC CAU CAG AAG AUG AGU UUU AAC UCA UCU UCU GAU GGG CUU
siUSP21-641 CCA ACU UAG CCC GUU CCA AUU UUG GAA CGG GCU AAG UUG GUU
siUSP21-1353 GCU AGA AGA ACC UGA GUU AUU UAA CUC AGG UUC UUC UAG CUU
siUSP22-1367 GCU ACC AGG AGU CCA CAA AUU UUU GUG GAC UCC UGG UAG CUU
siUSP22-695 GGA GAA AGA UCA CCU CGA AUU UUC GAG GUG AUC UUU CUC CUU
siUSP24-611 GGA AUU GAA UUC CCU ACA AUU UUG UAG GGA AUU CAA UUC CUU
siUSP24-719 GCA UCU ACC UAC CUA GCA AUU UUG CUA GGU AGG UAG AUG CUU
siUSP25-897 GCC AAA GAA CCC UAU GGU AUU UAC CAU AGG GUU CUU UGG CUU
siUSP25-1128 GCC GGU AUU AAC AUU UGA AUU UUC AAA UGU UAA UAC CGG CUU
siUSP26-1600 CCU UAU UGU UCA CCU CAA AUU UUU GAG GUG AAC AAU AAG GUU
siUSP26-2426 GGU UCC AAU AAG AAU CCA AUU UUG GAU UCU UAU UGG AAC CUU
siUSP27-1398 GGC GCA AGA UCA CUA CAU AUU UAU GUA GUG AUC UUG CGC CUU
siUSP27-855 CUC CUC AUG UGC CCU AUA AUU UUA UAG GGC ACA UGA GGA GUU
siUSP28-1836 GGG CCU AUA UCU AUA AUC AUU UGA UUA UAG AUA UAG GCC CUU
siUSP28-841 GCA UUC CAG CUA GCU GUU AUU UAA CAG CUA GCU GGA AUG CUU
Continued
Content courtesy of Springer Nature, terms of use apply. Rights reserved
4
Vol:.(1234567890)
Scientic Reports | (2022) 12:6445 | https://doi.org/10.1038/s41598-022-09497-y
www.nature.com/scientificreports/
Table 1. Sequences of siRNAs targeting USPs.
siRNAs Sense (5–3) Antisense (5–3)
siUSP29-983 CCC AUC AAG UUU AGA GGA UUU AUC CUC UAA ACU UGA UGG GUU
siUSP29-1922 GGU GAA GAA UAA CGA GCA AUU UUG CUC GUU AUU CUU CAC CUU
siUSP30-176 CCG UCA GAU AUA AAG UCA UUU AUG ACU UUA UAU CUG ACG GUU
siUSP30-521 GCU GCU UGU UGG AUG UCU UUU AAG ACA UCC AAC AAG CAG CUU
siUSP31-954 GCC UCU CUA UGU CAC UGU AUU UAC AGU GAC AUA GAG AGG CUU
siUSP31-3922 GCU CGC AAA UCC AAG UCU UUU AAG ACU UGG AUU UGC GAG CUU
siUSP32-2261 GCG CAU UAA AGA GGA AGA UUU AUC UUC CUC UUU AAU GCG CUU
siUSP32-386 GAC CUG UGG ACU CUC AUA UUU AUA UGA GAG UCC ACA GGU CUU
siUSP33-829 CCC AGU AAU ACA ACA UUA AUU UUA AUG UUG UAU UAC UGG GUU
siUSP33-597 GGA GAA UAG AUG UUC AUA UUU AUA UGA ACA UCU AUU CUC CUU
siUSP34-1228 GCG ACU GAG UAC UCA ACA UUU AUG UUG AGU ACU CAG UCG CUU
siUSP34-3023 CCU GAU CAU UUC AGG UUA AUU UUA ACC UGA AAU GAU CAG GUU
siUSP35-3188 CCC UGC ACA AGG ACU UGA UUU AUC AAG UCC UUG UGC AGG GUU
siUSP35-1916 GCU CGG AGU AUC UGA AGU AUU UAC UUC AGA UAC UCC GAG CUU
siUSP36-741 CCA ACU ACC UGC UCU CCA AUU UUG GAG AGC AGG UAG UUG GUU
siUSP36-474 GCA AAU AUG UGU UGC UCA AUU UUG AGC AAC ACA UAU UUG CUU
siUSP37-555 CCA AGG AUA UUU CAG CUA AUU UUA GCU GAA AUA UCC UUG GUU
siUSP37-2235 GCA CAU AUG GCA AUU UCU AUU UAG AAA UUG CCA UAU GUG CUU
siUSP38-3501 GGU AAG UUG GAA AUA CAA GUU CUU GUA UUU CCA ACU UAC CUU
siUSP38-1047 GGU UCG AAC GAU AGG CCA UUU AUG GCC UAU CGU UCG AAC CUU
siUSP39-958 GGA ACC CUC GAA AUU UCA AUU UUG AAA UUU CGA GGG UUC CUU
siUSP39-1375 GCA UCA CUG AGA AGG AAU AUU UAU UCC UUC UCA GUG AUG CUU
siUSP40 -721 GCA GCA AAG UCG GCC AAA UUU AUU UGG CCG ACU UUG CUG CUU
siUSP40 -1212 GCU CCA UUC UCA GAU AUU UUU AAA UAU CUG AGA AUG GAG CUU
siUSP42-459 GCU CCA GAA UUU GGG CAA UUU AUU GCC CAA AUU CUG GAG CUU
siUSP42-750 GCA GAA AGC AUG CUU GAA UUU AUU CAA GCA UGC UUU CUG CUU
siUSP43-813 GCC ACU UUC AAG CAC AAU AUU UAU UGU GCU UGA AAG UGG CUU
siUSP43-2196 GGG CUU AUA UCC UGU UCU AUU UAG AAC AGG AUA UAA GCC CUU
siUSP44-653 GGG UAC AGG UGA UGA UUC UUU AGA AUC AUC ACC UGU ACC CUU
siUSP44-1553 CGC UCA GGA AUU UCU UUG UUU ACA AAG AAA UUC CUG AGC GUU
siUSP45-1377 GGC ACC UCG AUU UAA AGA UUU AUC UUU AAA UCG AGG UGC CUU
siUSP45-753 GCA GCU AGU ACU UAC UUC UUU AGA AGU AAG UAC UAG CUG CUU
siUSP46-204 GGU CCA GAG CAG UUU CCA AUU UUG GAA ACU GCU CUG GAC CUU
siUSP46-426 CCA CCA AAG AAG UUC AUU UUU AAA UGA ACU UCU UUG GUG GUU
siUSP47-2463 GCU GUC GCC UUG UUA AAU AUU UAU UUA ACA AGG CGA CAG CUU
siUSP47-3757 CCA GCA AUC AAG AGU UUG AUU UCA AAC UCU UGA UUG CUG GUU
siUSP48-676 GCA UCU CCA GUA CUU GUU UUU AAA CAA GUA CUG GAG AUG CUU
siUSP48-871 GCA GUU CUG UGG AGA AUA UUU AUA UUC UCC ACA GAA CUG CUU
siUSP49-1925 GGG UCC AUG UCG UCU UUG AUU UCA AAG ACG ACA UGG ACC CUU
siUSP49-1825 GAA GCU AGA AAG CAG UUA AUU UUA ACU GCU UUC UAG CUU CUU
siUSP50-571 GCU CAG GAA UUC UUG AUU UUU AAA UCA AGA AUU CCU GAG CUU
siUSP50-692 CCA CUG AGA CAU CCA UCA UUU AUG AUG GAU GUC UCA GUG GUU
siUSP51-895 CCA UUU AGC UGU AGA CCU UUU AAG GUC UAC AGC UAA AUG GUU
siUSP51-1225 CCA UAU UCC UCU ACU GAA AUU UUU CAG UAG AGG AAU AUG GUU
siUSP52-896 GCU GCA GAA UCA CAU ACU AUU UAG UAU GUG AUU CUG CAG CUU
siUSP52-1247 GCG CUU CAU UCC UAC AUA UUU AUA UGU AGG AAU GAA GCG CUU
siUSP53-3296 GAG CCA ACA UCA CUU AGA AUU UUC UAA GUG AUG UUG GCU CUU
siUSP53-1605 GUG CGG UAC AUU UCU ACA AUU UUG UAG AAA UGU ACC GCA CUU
siUSP54-3654 GCU GCC UAA UGG UGA AAC UUU AGU UUC ACC AUU AGG CAG CUU
siUSP54-2546 GAG CCC UAG UCG AUA AGA AUU UUC UUA UCG ACU AGG GCU CUU
siRNA NC CUC CGA ACG UGU CAC GUU CGU GAC ACG UUC GGA GUU
Content courtesy of Springer Nature, terms of use apply. Rights reserved
5
Vol.:(0123456789)
Scientic Reports | (2022) 12:6445 | https://doi.org/10.1038/s41598-022-09497-y
www.nature.com/scientificreports/
Colony formation assay for cell growth. U-251 MG and U-87 MG cells were tripsinized and then
seeded into 6-well plates at a density of 1 × 103 per well 6h aer transfection. e cells were continuously cul-
tured at 37°C for 7–10days. e colonies were xed with 4% methanal (Sangon Biotech, Shanghai, China) in
phosphate buer saline (PBS) at 28°C for 15min. Aer washing with PBS three times, the colonies were pho-
tographed using HUAWEI Mate 40 (Huawei, Shenzhen, China) and the number of colonies was counted using
Image J v1.48u (NIH, Bethesda, MD, USA).
Microscopic analysis of EdU incorporation for cell proliferation. U-251 MG and U-87 MG cells
were tripsinized aer a 24h of transfection and then seeded into 96-well plates at a density of 2 × 104 per well.
Twelve hours later, the cells were labeled with 10μM EdU solution (Beyotime, Shanghai, China) at 37°C for 3h.
Aer xation with 4% methanal in PBS at 28°C for 20min and subsequent permeation with 0.5% Triton X-100
(Sangon Biotech) in PBS at 28°C for 10min, the cells were incubated with 50µL of 1 × Click Additive Solution
(Beyotime) at 28°C for 30min. e nucleus was stained with 5μg/mL DAPI (Yeasen). Finally, the uorescent
dots were observed and photographed using a uorescence microscope (MOTIC, Hongkong, China) and then
photographed. Image J v1.48u was used to count the number of cells.
Flow cytometry‑based cell cycle. U-251 MG and U-87 MG cells were collected aer a 24h of transfec-
tion and then xed using 70% ethanol in PBS at − 20°C for 6h. e cells were then treated with 0.5% Triton
X-100 and 10μg/mL RNase (Sangon Biotech) at 28°C for 25min. Finally, the cells were stained with 20μg/
mL propidium iodide (PI, Vazyme) in the dark at 28°C for 25min and then placed into to the ow cytometer
NovoCyte 1300 (ACEA, San Diego, CA, USA) for uorescent detection within the PE-channel (Ex: 488nm/Em:
578nm).
Transwell assay for cell migration and invasion. U-251 MG and U-87 MG cells were tripsinized aer
a 24h of transfection and then suspended in FBS-free DMEM at a density of 3 × 105/mL. In the transwell migra-
tion assay, 100μL of suspended cells were placed into the upper chambers of transwell plates (NEST, Wuxi,
Jiangsu, China). e lower chamber was supplied with 550μL 10% FBS DMEM. In the Matrigel invasion assay,
the membrane of the upper chamber was pre-coated with sixfold diluted Matrigel (Corning, Corning, NY, USA)
before seeding cells. e migrated and invasive cells were xed with 4% methanal in PBS and then stained with
1% crystal violet (Sangon Biotech) in PBS at 28°C for 20min, respectively. e stained cells were observed and
photographed using a light microscope (MOTIC). e number of migrated/invasive cells were counted using
Image J v1.48u in three random elds.
Construction of stable U‑87 MG cell lines with USP32 knockdown (shUSP32). e lentiviral
vector PLKO.1-TRC-Puro (Antihela, Xiamen, Fujian, China) was used to construct plasmid overexpressing
short hairpin RNA targeting USP32 and shctrl plasmid. e primers for shUSP32 plasmid construction were
designed based on the sequence of siUSP32-386 (Table2). For lentiviral packaging, 293T cells (4 × 106/well)
were seeded into 6-well plates and transfected with 3µg shUSP32 or shctrl plasmid, 2µg psPAX2 (Antihela),
Table 2. Primers for RT-qPCR and plasmid construction.
Name Forward primer (5–3) Reverse primer (5–3)
CCNB1 GAC CTG TGT CAG GCT TTC TCTG GGT ATT TTG GTC TGA CTG CTTGC
CDC25A TCT GGA CAG CTC CTC TCG TCAT ACT TCC AGG TGG AGA CTC CTCT
CDC45C TGG ATG CTG TCC AAG GAC CTGA CAG GAC ACC AAC ATC AGT CACG
CDK1 GGA AAC CAG GAA GCC TAG CATC GGA TGA TTC AGT GCC ATT TTGCC
MCM3 CGA GAC CTA GAA AAT GGC AGCC GCA GTG CAA AGC ACA TAC CGCA
MCM4 CTT GCT TCA GCC TTG GCT CCAA GTC GCC ACA CAG CAA GAT GTTG
MCM6 GAC AAC AGG AGA AGG GAC CTCT GGA CGC TTT ACC ACT GGT GTAG
MCM7 GCC AAG TCT CAG CTC CTG TCAT CCT CTA AGG TCA GTT CTC CACTC
FEN1 ACT AAG CGG CTG GTG AAG GTCA GCA GCA TAG ACT TTG CCA GCCT
NEIL3 AGT GGT CTC CAC CCA GCT GTTA AGA GCA AGT CCT GCT TTA CGGC
POLE ACG CTG GAA GAG GTG TAT GGCT GGA ACG GTT CTC AGA GAT GAGC
POLE2 TGC GTC CGT TTT CCT AGC AGCA GGG CAG ACA TAA AGA GGT AGGG
EXO1 TCG GAT CTC CTA GCT TTT GGCTG AGC TGT CTG CAC ATT CCT AGCC
RFC2 GTC GGG AAT GAA GAC ACC GTGA CAG AAT GCT TGT GGT CTT GCCG
RFC3 CCT GAG ACA GAT TGG GAG GTGT AGC TCA TAC AGC CTT CCA CGAAC
RFC4 GGC AGC TTT AAG ACG TAC CATGG TCT GAC AGA GGC TTG AAG CGGA
Shctrl
CCG GCT CCG AAC GTG TCA CGCTC AAT TAA AAA CTC CGA ACG TGT CAC GCT CGA GCG TGA CAC GTT CGGAG
GAG CGT GAC ACG TTC GGA GTT TTT AAT TAA AAA GAC CTG TGG ACT CTC ATA TCT CGA GAT ATG AGA GTC CAC
AGGTC
shUSP32 CCG GGA CCT GTG GAC TCT CAT ATC TCG AGA TAT GAG AGT CCA CAG GTC
TTTTT
Content courtesy of Springer Nature, terms of use apply. Rights reserved
6
Vol:.(1234567890)
Scientic Reports | (2022) 12:6445 | https://doi.org/10.1038/s41598-022-09497-y
www.nature.com/scientificreports/
and 1µg pMD2.G (Antihela) using LipofectamineRNAiMAX at 37°C. Aer incubation for 48h, the lentivirus
was harvested and used to infect U-87 MG cells at a multiplicity of infection of 30 with the addition of 10µg/mL
polybrene (ermo Fisher Scientic). Forty-eight hours aer infection, U-87 MG cells were treated with 1.0μg/
mL puromycin (Yeasen) for 3days, constructing shUSP32 and shctrl U-87 MG cells.
Animal experiments. Six-week-old female BALB/c nude mice were obtained from Vitalriver (Beijing,
China). e subcutaneous injection of shctrl or shUSP32 cells (4 × 106) was performed into the right ank of six
mice. e long diameter (a) and short diameter (b) of tumors were measured every 4days. e tumor volume
(V) was calculated using the formula V = ab2/226. e mice were euthanized using isourane (RWD life science,
Shenzhen, China) at day 48. Tumors were dissected o and photographed using HUAWEI Mate 40. e tumor
was also weighed.
RNA sequencing (RNA‑Seq) and data analysis. Total RNA from shctrl or shUSP32 U-87 MG cells was
used as input material for the RNA sample preparations. mRNA was puried from total RNA using poly-T oligo-
attached magnetic beads. e RNA-Seq library was built by Novogene (Beijing, China). Aer cluster generation
using TruSeq PE Cluster Kit v3-cBot-HS (Illumia, San Diego, CA, USA) on a cBot Cluster Generation System
(Illumia), the library preparations were sequenced on a Novaseq platform (Illumina). RNA-Seq data analysis
was performed according to the protocol of Novogene. In brief, reads were aligned to the human transcriptome
and genome hg19 using T Hisat2 v2.0.5. Transcripts and genes were quantied using featureCounts v1.5.0-
p3. Dierential expression analysis was performed using DESeq2 R package v1.20.0. Genes with an adjusted
p value < 0.05 were considered as dierentially expressed. GO and KEGG pathway enrichment analyses2729 of
dierentially expressed genes were performed using the clusterProler R package.
Figure1. High content screening based on cell viability to screen USPs may function in glioblastoma. Data are
represented as mean ± standard deviation (SD) of three biological replicates.
Content courtesy of Springer Nature, terms of use apply. Rights reserved
7
Vol.:(0123456789)
Scientic Reports | (2022) 12:6445 | https://doi.org/10.1038/s41598-022-09497-y
www.nature.com/scientificreports/
Statistical analysis. All statistical analyses were performed using GraphPad Prism v8.2.1 (GraphPad So-
ware, San Diego, CA, USA). e data are presented as mean ± standard deviation (SD) unless otherwise shown.
ANOVA followed by Tukey’s post-hoc test was used for multiple comparisons among three groups. Unpaired
Student’s t test was performed to compare the dierence between two groups. Survival curves were calculated
using the Kaplan–Meier method, and the signicance was determined by the log-rank test. Statistical signi-
cance was accepted at p < 0.05.
Ethics approval. Animal experiments were conducted in accordance with the national guidelines for the
humane treatment of animals and were approved by the Institutional Animal Care and Use Committee (IACUC)
at Xiamen University. e study is reported in accordance with ARRIVE guidelines.
Results
High content screening. High content screening was performed to identify the USPs regulating GBM cell
survival. As shown in Fig.1, knockdown of USP1, USP8, or USP32 in U-87 MG cells inhibited cell viability by
at least half. Knockdown of USP32, USP9X, or USP1 suppressed the viability of U-251 MG. erefore, USP32
was chosen for further study.
USP32 expression level linked to poor prognosis. First, we investigated the clinical signicance of
USP32 using GEO dataset. As can be seen in Fig.2A, the USP32 expression level was higher in GBM tissues
compared to normal tissues. Moreover, higher USP32 expression level indicated poorer prognosis (Fig.2B).
Next, we evaluated the USP32 expression level in normal brain cells and GBM cells. e results showed that the
USP32 mRNA and protein levels in GBM cells (U-118 MG, U-87 MG, A172, T98G, and U-251 MG) were higher
than those in the normal brain cell SVG p12 (Fig.2C,D). Furthermore, U-87 MG and U-251 MG have the high-
est USP32 expression levels. Based on these ndings, we chose to knock down USP32 in U-87 MG and U-251
MG cells to study the function of USP32 in GBM.
Figure2. USP32 expression level in glioblastoma (GBM) tissues and cell lines and the association of USP32
expression level with prognosis. (A,B) USP32 is upregulated in GBM patients (A), which indicates poor
prognosis (B). e USP32 mRNA expression data and survival information of GBM patients were downloaded
from the Gene Expression Omnibus databases. (C,D) e USP32 mRNA and protein levels in GBM cells (U-118
MG, U-87 MG, A172, T198G, and U-251 MG) and normal brain cell SVG p12 were determined using RT-qPCR
(C) and western blotting (D), respectively. Data are represented as mean ± SD of three technical replicates.
Unpaired Student’s t test for (A). Kaplan–Meier method and the signicance was determined using the log-rank
test for (B). One way ANOVA followed by Tukey’s post-hoc test for (C,D): vs SVG p12, *p < 0.05, ***p < 0.001,
****p < 0.0001.
Content courtesy of Springer Nature, terms of use apply. Rights reserved
8
Vol:.(1234567890)
Scientic Reports | (2022) 12:6445 | https://doi.org/10.1038/s41598-022-09497-y
www.nature.com/scientificreports/
Knockdown of USP32 inhibits cell growth. USP32 in U-87 MG and U-251 MG cells was knocked down
using siRNAs. As shown in Fig.3A,B, the mRNA and protein levels of USP32 were signicantly reduced aer
transfection with siUSP32-2261 and siUSP32-386. Moreover, siUSP32-386 had higher knockdown eciency
Figure3. Knockdown of USP32 inhibits cell growth. (A) e mRNA level of USP32 was reduced by siUSP32-
2261 and siUSP32-386. Data are represented as mean ± SD of three biological replicates. (B) Le panel:
Representative images of three independent western bolting analyses showing the knockdown eciency of
siUSP32-2261 and siUSP32-386. Right panel: statistical quantication of le panel. (C) MTT assay determining
the cell viability of U-87 MG and U-251 MG. Data are represented as mean ± SD of six biological replicates. (D)
Representative images of three independent colony formation assays showing that USP32 knockdown inhibited
cell proliferation. (E) Statistical quantication of (D). (F) USP32 knockdown reduced the number of EdU+ cells.
Bar: 10μm. (G) Statistical quantication of (F). Data are represented as mean ± SD of three technical replicates.
(H) Representative images of three independent cell cycle assays by ow cytometry. (I) Histogram showing the
percentage of each cell-cycle phase in (H). One way ANOVA followed by Tukey’s post-hoc test: vs siRNA NC,
*p < 0.05, **p < 0.01, ***p < 0.001, ****p < 0.0001.
Content courtesy of Springer Nature, terms of use apply. Rights reserved
9
Vol.:(0123456789)
Scientic Reports | (2022) 12:6445 | https://doi.org/10.1038/s41598-022-09497-y
www.nature.com/scientificreports/
than siUSP32-2261. Similarly, the CCK-8 assay indicated that USP32 knockdown signicantly suppressed the
viability of U-87 MG and U-251 MG cells (Fig.3C). Cellular proliferation was analyzed using colony formation
and EdU assays in U-87 MG and U-251 MG cells; Fig.3D,E show that USP32 knockdown signicantly reduced
the number of colonies. In U-87 MG cells, the number of colonies decreased from 125 ± 11 to 80 ± 14 (p = 0.0058)
for siUSP32-2261 and 46 ± 5 (p = 0.0003) for siUSP32-386. In U-251 MG cells, the number of colonies decreased
from 48 ± 8 to 24 ± 3 (p = 0.0051) for siUSP32-2261 and 8 ± 3 (p = 0.0003) for siUSP32-386. e percentage of
EdU+ cells was also signicantly reduced by silencing USP32 (Fig.3F). In U-87 MG cells, the percentage of
EdU+ cells decreased from 25.5 ± 2.5% to 16.9 ± 2.0% (p = 0.0045) for siUSP32-2261 and 8.6 ± 1.1% (p = 0.0001)
for siUSP32-386. In U-251 MG cells, the percentage of EdU+ cells decreased from 39.3 ± 1.4% to 28.3 ± 2.3%
(p = 0.0016) for siUSP32-2261 and 24.2 ± 2.3% (p = 0.0003) for siUSP32-386 (Fig.3G). Next, we investigated the
eect of USP32 on cell cycle progression. e results show that USP32 knockdown promotes the arrest of cells
in the G0/G1 phase (Fig.3H,I). ese ndings suggested that silencing USP32 may inhibit cell growth due to
cell-cycle arrest.
Knockdown of USP32 inhibits cell metastasis. Transwell migration and Matrigel invasion assays were
performed to study the eect of USP32 on cell metastasis. Figure4A shows that USP32 downregulation inhib-
ited cell migration. In U-87 MG cells, the number of migrated cells was 330 ± 26, 186 ± 17, and 123 ± 13 for
siRNA NC, siUSP32-2261 (p = 0.0003, vs siRNA NC), and siUSP-386 (p < 0.0001, vs siRNA NC), respectively. In
U-251 MG cells, the number of migrated cells was 431 ± 20, 305 ± 21, and 246 ± 13 for siRNA NC, siUSP32-2261
(p = 0.0004, vs siRNA NC), and siUSP-386 (p < 0.0001, vs siRNA NC), respectively (Fig.4B). is downregula-
tion also inhibited cell invasion (Fig.4C). In U-87 MG cells, the number of invasive cells decreased from 136 ± 22
to 53 ± 10 (p = 0.0010) for siUSP32-2261 and 24 ± 3 (p = 0.0002) for siUSP32-386. In U-251 MG cells, the number
of invasive cells decreased from 136 ± 22 to 53 ± 10 (p = 0.0010) for siUSP32-2261 and 24 ± 3 (p = 0.0002) for
siUSP32-386. In U-251 MG cells, the number of invasive cells were reduced from 156 ± 19 to 77 ± 10 (p = 0.0008)
for siUSP32-2261 and 34 ± 6 (p < 0.0001) for siUSP32-386 (Fig.4D). Taken together, these data indicate that
USP32 knockdown inhibited cell metastasis.
Figure4. Knockdown of USP32 inhibits cell metastasis. (A) Transwell migration assay showing that silencing
USP32 suppressed cell migration. Bar: 10μm. Images were representatives of three independent experiments.
(B) Statistical quantication of (A). (C) Matrigel invasion assay showing that USP32 deciency suppressed
cell invasion. Bar: 10μm. Images were representatives of three independent experiments. (D) Statistical
quantication of (C). One way ANOVA followed by Tukey’s post-hoc test: vs siRNA NC, **p < 0.01, ***p < 0.001,
****p < 0.0001.
Content courtesy of Springer Nature, terms of use apply. Rights reserved
10
Vol:.(1234567890)
Scientic Reports | (2022) 12:6445 | https://doi.org/10.1038/s41598-022-09497-y
www.nature.com/scientificreports/
Knockdown of USP32 inhibits tumor growth in vivo. Next, stably USP32-knockdown U-87 MG cells
were constructed to validate the function of USP32 invivo. Figure5A–C show that the stable cell lines were suc-
cessfully constructed. e constructed cells were then subcutaneously injected into nude mice. As can be seen
in Fig.5D, the tumor in group shUSP32 grew more slowly compared to group shctrl. e dissected tumors are
shown in Fig.5E, being the weight lighter when group shUSP32 was compared to shctrl (Fig.5F). Moreover, the
mRNA and protein levels of USP32 were indeed lower in group shUSP32 than those in group shctrl (Fig.5G–I).
ese data demonstrate that USP32 knockdown inhibits tumor growth invivo.
Analysis of dierentially expressive genes (DEGs). Transcriptional sequencing was performed to
nd the dierentially expressive genes between shctrl and shUSP32 U87-MG cells. e volcano plot indicates
that 2017 genes were signicantly upregulated and 2333 genes were signicantly downregulated aer USP32
knockdown (Fig.6A). e heat map was used to show the distinguishable mRNA expression patterns between
shUSP32 and shctrl samples (Fig.6B).
GO and KEGG pathway enrichment analyses. All upregulated and downregulated genes were used for
GO and KEGG enrichment analyses. In the GO enrichment analysis, 10 molecular functions such as cadherin
binding and catalytic activity acting on DNA, 11 cellular components such as condensed chromosome and cell
cycle checkpoint, and 9 biological processes such as mitotic cell cycle phase initiation and DNA replication
were signicantly modulated (Fig.6C). KEGG pathway enrichment analysis discovers 20 pathways signicantly
linked to USP32 expression, including cell cycle and DNA replication pathways (Fig.6D). e enriched DEGs in
GO and KEGG pathway enrichment analyses are listed in Supplementary TableS1 and Supplementary TableS2,
respectively. e expression prole of several DEGs associated with cell cycle, DNA replication, base excision
repair, and mismatch repair are shown in Fig.7A. RT-qPCR analysis conrmed that USP32 knockdown reduced
the expression of these genes (Fig.7B).
Figure5. Knockdown of USP32 suppresses tumor growth invivo. (A,C) Stably USP32-knockdown U-87
MG cell lines were successfully constructed, determined using RT-qPCR (A) and western blotting (B,C). Data
are represented as mean ± SD of three technical replicates. (D) Tumor volume was tracked every 4days by
calculating using the formula: volume = long diameter × short diameter2/2. Data are represented as mean ± SD
of six mice. (E) e image of tumors. e mice were euthanized using isourane at day 48 and tumors were
dissected o. (F) USP32 knockdown reduced the weight of tumors. Data are represented as mean ± SD of six
mice. (G) RT-qPCR analysis degerming the mRNA level of USP32 in tumor tissues. Data are represented as
mean ± SD of six mice. (H) Western blotting. (I) Statistical quantication of (H). Unpaired Student’s t test: vs
shctrl, *p < 0.05, ***p < 0.001, ****p < 0.0001.
Content courtesy of Springer Nature, terms of use apply. Rights reserved
11
Vol.:(0123456789)
Scientic Reports | (2022) 12:6445 | https://doi.org/10.1038/s41598-022-09497-y
www.nature.com/scientificreports/
Discussion
GBM accounts for 16% of primary brain tumors with an incidence rate of thirty-two per million population1,2.
GBM oen causes poor prognosis and the median-survival time of patients is less than 2 years30,31. It is urgent
to nd novel targets for brain-penetrating targeted therapies. Recently, more and more USPs were reported to
play important roles in GBM progression7,21. USP1, USP8, USP9x, and USP28 were identied as oncogenes in
GBM3235. USPs with antitumor activity such as USP11 and USP286 promote cell viability aer being silenced36,37,
which is consistent with the results in Fig.1. In this study, USP32 knockdown inhibited cell growth and metastasis
invitro, and suppressed tumor growth invivo, which suggests that USP32 acts as an oncogene in GBM and may
serve as a potential target for GBM treatment.
EdU+ cells indicate the cells in DNA replication. DNA replication, occurring in the S phase of interphase
during cell cycle, is an important step for cell proliferation and division38. Results showed that the percentages of
EdU+ cells and G0/G1-phase cells were reduced and increased aer USP32 knockdown, respectively. Moreover,
GO and KEGG pathway analyses revealed that this enzyme is involved in DNA replication and cell cycle pro-
cesses or pathways. is suggests that USP32 may promote cells passing through the G0/G1 phase and initiate
the DNA replication, promoting the proliferation of cancer cells, which is consistent with the study of Hu etal.23.
Base excision repair is an essential genome-maintenance pathway by which cells repair damaged DNA bases
that arise at a high level during DNA replication. Failure to remove the damaged DNA bases causes increasing
Figure6. Gene Ontology (GO) and Kyoto Encyclopedia of Genes and Genomic (KEGG) pathway enrichment
analyses using dierentially expressive genes (DEGs) screened out by transcriptional sequencing. (A) Volcano
plot based on the results of RNA sequencing for transcriptomes from shctrl and shUSP32 U87-MG cells. (B)
e heat map showing the distinguishable mRNA expression patterns between the shUSP32 and shctrl samples.
(C) GO enrichment analysis showing the considerable molecular functions, cellular components, and biological
processes. (D) KEGG pathway enrichment analysis showing the considerable pathways.
Content courtesy of Springer Nature, terms of use apply. Rights reserved
12
Vol:.(1234567890)
Scientic Reports | (2022) 12:6445 | https://doi.org/10.1038/s41598-022-09497-y
www.nature.com/scientificreports/
levels of mutation and chromosomal instability, nally resulting in carcinogenesis39. DNA mismatch repair is a
rescue system that conserves the DNA sequences by removing the erroneously mismatched, inserted, and deleted
bases during DNA duplication and recombination. Defects in DNA mismatch repair are also associated with
carcinogenesis40. Moreover, DUBs are oen involved in base excision repair and mismatch repair processes17,18.
erefore, USP32 eect on the expression of molecules functioning in these processes was validated using RT-
qPCR, although the adjusted p-values for base excision repair and mismatch repair pathways in KEGG pathway
analysis were greater than 0.05. e results indicate that USP32 regulates the expressions of MCM3, MCM4,
MCM6, MCM7, FEN1, NEIL3, and POLE, suggesting that USP32 plays an important role in base excision repair
and mismatch repair.
ere are few reports about elements of the ubiquitin–proteasome system involved in GBM metastasis. USP18
was reported to promote epithelial-mesenchymal transition in GBM cells by deubiquitinating and stabilizing
Twist141. UBE2T, a ubiquitin-conjugating enzyme, stabilizes GRP78 to promote the metastasis of GBM cells42.
Our study demonstrates that USP32 facilitates the migration and invasion of GBM cells, which supports that
the ubiquitin–proteasome system plays an important role in the GBM metastasis.
Further experiments are needed to conrm the mechanism of action by which USP32 upregulates the expres-
sion of several genes (Fig.7B). Further, the eect of USP32 modulation on cell function in normal glial cells
and a broader panel of GBM cells will be evaluated. In addition, a collection of clinical samples to analyze the
expression of this enzyme in GBM and normal tissue will also be included.
In conclusion, our study demonstrates that USP32 acts as an oncogene in GBM through regulating cell
cycle, DNA replication, base excision repair, and mismatch repair. USP32 could be a potential target for GBM
treatment.
Data availability
e data in this study will be made available from the corresponding author on reasonable request.
Figure7. USP32 is involved in cell cycle, DNA replication, base excision repair, and mismatch repair processes/
pathways. (A) e expression prole of several DEGs. (B) RT-qPCR analysis conrming the regulatory eect of
USP32 on cell cycle, DNA replication, base excision repair, and mismatch repair processes/pathways. Data are
represented as mean ± SD of three technical replicates.
Content courtesy of Springer Nature, terms of use apply. Rights reserved
13
Vol.:(0123456789)
Scientic Reports | (2022) 12:6445 | https://doi.org/10.1038/s41598-022-09497-y
www.nature.com/scientificreports/
Received: 26 October 2021; Accepted: 7 March 2022
References
1. Gusyatiner, O. & Hegi, M. E. Glioma epigenetics: From subclassication to novel treatment options. Semin. Cancer Biol. 51, 50–58.
https:// doi. org/ 10. 1016/j. semca ncer. 2017. 11. 010 (2018).
2. Davis, M. E. Glioblastoma: Overview of disease and treatment. Clin. J. Oncol. Nurs. 20, S2–S8. https:// doi. org/ 10. 1188/ 16. CJON.
S1.2-8 (2016).
3. Johnson, D. R., Ma, D. J., Buckner, J. C. & Hammack, J. E. Conditional probability of long-term survival in glioblastoma: A
population-based analysis. Cancer 118, 5608–5613. https:// doi. org/ 10. 1002/ cncr. 27590 (2012).
4. Holland, E. C. Progenitor cells and glioma formation. Curr. Opin. Neurol. 14, 683–688. https:// doi. org/ 10. 1097/ 00019 052- 20011
2000- 00002 (2001).
5. Touat, M., Idbaih, A., Sanson, M. & Ligon, K. L. Glioblastoma targeted therapy: Updated approaches from recent biological insights.
Ann. Oncol. 28, 1457–1472. https:// doi. org/ 10. 1093/ annonc/ mdx106 (2017).
6. Sun, T., Liu, Z. & Yang, Q. e role of ubiquitination and deubiquitination in cancer metabolism. Mol. Cancer 19, 146–146. https://
doi. org/ 10. 1186/ s12943- 020- 01262-x (2020).
7. Maksoud, S. e role of the ubiquitin proteasome system in glioma: Analysis emphasizing the main molecular players and thera-
peutic strategies identied in glioblastoma multiforme. Mol. Neurobiol. 58, 3252–3269. https:// doi. org/ 10. 1007/ s12035- 021- 02339-4
(2021).
8. Zaky, W. et al. e ubiquitin-proteasome pathway in adult and pediatric brain tumors: Biological insights and therapeutic oppor-
tunities. Cancer Metastasis Rev. 36, 617–633. https:// doi. org/ 10. 1007/ s10555- 017- 9700-2 (2017).
9. Vlachostergios, P. J., Voutsadakis, I. A. & Papandreou, C. N. e role of ubiquitin-proteasome system in glioma survival and
growth. Growth Factors 31, 106–113. https:// doi. org/ 10. 3109/ 08977 194. 2013. 799156 (2013).
10. Scholz, N., Kurian, K. M., Siebzehnrubl, F. A. & Licchesi, J. D. F. Targeting the ubiquitin system in glioblastoma. Front. Oncol. 10,
574011. https:// doi. org/ 10. 3389/ fonc. 2020. 574011 (2020).
11. aker, N. G. et al. Identication of survival genes in human glioblastoma cells by small interfering RNA screening. Mol. Pharmacol.
76, 1246–1255. https:// doi. org/ 10. 1124/ mol. 109. 058024 (2009).
12. Kavsak, P. et al. Smad7 binds to Smurf2 to form an E3 ubiquitin ligase that targets the TGF beta receptor for degradation. Mol.
Cell 6, 1365–1375. https:// doi. org/ 10. 1016/ s1097- 2765(00) 00134-9 (2000).
13. Li, H. et al. HERC3-mediated SMAD7 ubiquitination degradation promotes autophagy-induced EMT and chemoresistance in
glioblastoma. Clin. Cancer Res. 25, 3602–3616. https:// doi. org/ 10. 1158/ 1078- 0432. Ccr- 18- 3791 (2019).
14. Zhang, Y. et al. e p53 pathway in glioblastoma. Cancers (Basel) 10, 297. https:// doi. org/ 10. 3390/ cance rs100 90297 (2018).
15. Chen, G. et al. Human Brat ortholog TRIM3 is a tumor suppressor that regulates asymmetric cell division in glioblastoma. Cancer
Res. 74, 4536–4548. https:// doi. org/ 10. 1158/ 0008- 5472. Can- 13- 3703 (2014).
16. Antao, A. M., Tyagi, A., Kim, K.-S. & Ramakrishna, S. Advances in deubiquitinating enzyme inhibition and applications in cancer
therapeutics. Cancers (Basel) 12, 1579. https:// doi. org/ 10. 3390/ cance rs120 61579 (2020).
17. Fraile, J. M., Quesada, V., Rodríguez, D., Freije, J. M. & López-Otín, C. Deubiquitinases in cancer: New functions and therapeutic
options. Oncogene 31, 2373–2388. https:// doi. org/ 10. 1038/ onc. 2011. 443 (2012).
18. Komander, D., Clague, M. J. & Urbé, S. Breaking the chains: Structure and function of the deubiquitinases. Nat. Rev. Mol. Cell Biol.
10, 550–563. https:// doi. org/ 10. 1038/ nrm27 31 (2009).
19. Chen, D., Frezza, M., Schmitt, S., Kanwar, J. & Dou, Q. P. Bortezomib as the rst proteasome inhibitor anticancer drug: Current
status and future perspectives. Curr Cancer Drug Targets 11, 239–253. https:// doi. org/ 10. 2174/ 15680 09117 94519 752 (2011).
20. Richardson, P. G., Hideshima, T. & Anderson, K. C. Bortezomib (PS-341): A novel, rst-in-class proteasome inhibitor for the treat-
ment of multiple myeloma and other cancers. Cancer Control 10, 361–369. https:// doi. org/ 10. 1177/ 10732 74803 01000 502 (2003).
21. Jin, W. L., Mao, X. Y. & Qiu, G. Z. Targeting deubiquitinating enzymes in glioblastoma multiforme: Expectations and challenges.
Med. Res. Rev. 37, 627–661. https:// doi. org/ 10. 1002/ med. 21421 (2017).
22. Akhavantabasi, S. et al. USP32 is an active, membrane-bound ubiquitin protease overexpressed in breast cancers. Mamm. Genome
21, 388–397. https:// doi. org/ 10. 1007/ s00335- 010- 9268-4 (2010).
23. Hu, W. et al. Downregulation of USP32 inhibits cell proliferation, migration and invasion in human small cell lung cancer. Cell
Prolif. 50, e12343. https:// doi. org/ 10. 1111/ cpr. 12343 (2017).
24. Dou, N. et al. USP32 promotes tumorigenesis and chemoresistance in gastric carcinoma via upregulation of SMAD2. Int. J. Biol.
Sci. 16, 1648–1657. https:// doi. org/ 10. 7150/ ijbs. 43117 (2020).
25. Nakae, A. et al. Ubiquitin specic peptidase 32 acts as an oncogene in epithelial ovarian cancer by deubiquitylating farnesyl-
diphosphate farnesyltransferase 1. Biochem. Biophys. Res. Commun. 552, 120–127. https:// doi. org/ 10. 1016/j. bbrc. 2021. 03. 049
(2021).
26. Lai, M. C., Zhu, Q. Q., Xu, J. & Zhang, W. J. Experimental and clinical evidence suggests that GRPEL2 plays an oncogenic role in
HCC development. Am. J. Cancer Res. 11, 4175–4198 (2021).
27. Kanehisa, M., Furumichi, M., Sato, Y., Ishiguro-Watanabe, M. & Tanabe, M. KEGG: Integrating viruses and cellular organisms.
Nucleic Acids Res. 49, D545–D551. https:// doi. org/ 10. 1093/ nar/ gkaa9 70 (2021).
28. Kanehisa, M. Toward understanding the origin and evolution of cellular organisms. Protein Sci. 28, 1947–1951. https:// doi. org/
10. 1002/ pro. 3715 (2019).
29. Kanehisa, M. & Goto, S. KEGG: Kyoto encyclopedia of genes and genomes. Nucleic Acids Res. 28, 27–30. https:// doi. org/ 10. 1093/
nar/ 28.1. 27 (2000).
30. Lin, Z. et al. Establishment of age group classication for risk stratication in glioma patients. BMC Neurol. 20, 310–310. https://
doi. org/ 10. 1186/ s12883- 020- 01888-w (2020).
31. Stupp, R. et al. Eects of radiotherapy with concomitant and adjuvant temozolomide versus radiotherapy alone on survival in
glioblastoma in a randomised phase III study: 5-year analysis of the EORTC-NCIC trial. Lancet Oncol. 10, 459–466. https:// doi.
org/ 10. 1016/ s1470- 2045(09) 70025-7 (2009).
32. Lee, J.-K. et al. USP1 targeting impedes GBM growth by inhibiting stem cell maintenance and radioresistance. Neuro Oncol. 18,
37–47. https:// doi. org/ 10. 1093/ neuonc/ nov091 (2016).
33. Panner, A. et al. Ubiquitin-specic protease 8 links the PTEN-Akt-AIP4 pathway to the control of FLIPS stability and TRAIL
sensitivity in glioblastoma multiforme. Cancer Res. 70, 5046–5053. https:// doi. org/ 10. 1158/ 0008- 5472. CAN- 09- 3979 (2010).
34. Chen, Z. et al. USP9X deubiquitinates ALDH1A3 and maintains mesenchymal identity in glioblastoma stem cells. J. Clin. Investig.
129, 2043–2055. https:// doi. org/ 10. 1172/ JCI12 6414 (2019).
35. Wang, Z., Song, Q., Xue, J., Zhao, Y. & Qin, S. Ubiquitin-specic protease 28 is overexpressed in human glioblastomas and contrib-
utes to glioma tumorigenicity by regulating MYC expression. Exp. Biol. Med. 241, 255–264. https:// doi. org/ 10. 1177/ 15353 70215
595468 (2016).
36. Wu, H. C. et al. USP11 regulates PML stability to control Notch-induced malignancy in brain tumours. Nat. Commun. 5, 3214.
https:// doi. org/ 10. 1038/ ncomm s4214 (2014).
Content courtesy of Springer Nature, terms of use apply. Rights reserved
14
Vol:.(1234567890)
Scientic Reports | (2022) 12:6445 | https://doi.org/10.1038/s41598-022-09497-y
www.nature.com/scientificreports/
37. Kit Leng Lui, S. et al. USP26 regulates TGF-β signaling by deubiquitinating and stabilizing SMAD7. EMBO Rep. 21, e49618. https://
doi. org/ 10. 15252/ embr. 20194 9618 (2020).
38. Nabais Sá, M. J. et al. Biallelic GINS2 variant p.(Arg114Leu) causes Meier–Gorlin syndrome with craniosynostosis. J. Med. Genet.
https:// doi. org/ 10. 1136/ jmedg enet- 2020- 107572 (2021).
39. Arczewska, K. D., Michalickova, K., Donaldson, I. M. & Nilsen, H. e contribution of DNA base damage to human cancer is
modulated by the base excision repair interaction network. Crit. Rev. Oncog. 14, 217–273. https:// doi. org/ 10. 1615/ critr evonc og.
v14. i4. 20 (2008).
40. Li, G. M. Mechanisms and functions of DNA mismatch repair. Cell Res. 18, 85–98. https:// doi. org/ 10. 1038/ cr. 2007. 115 (2008).
41. Cai, X. et al. USP18 deubiquitinates and stabilizes Twist1 to promote epithelial-mesenchymal transition in glioblastoma cells. Am.
J. Cancer Res. 10, 1156–1169 (2020).
42. Huang, P. et al. UBE2T promotes glioblastoma invasion and migration via stabilizing GRP78 and regulating EMT. Aging (Albany
NY) 12, 10275–10289. https:// doi. org/ 10. 18632/ aging. 103239 (2020).
Acknowledgements
e study is supported by National Natural Science Foundation of China (No. 82072777).
Author contributions
W.Z.X. conceived and designed the study. C.S.F. collected the data. W.Z.X. and C.S.F. veried and analyzed the
data. C.X., L.Z., M.J.Y., J.W., Z.Z., L.Y.K., J.Z.Y., Z.W.P., and T.G.W. performed the experiments and draed the
manuscript. All authors read and approved the nal manuscript.
Competing interests
e authors declare no competing interests.
Additional information
Supplementary Information e online version contains supplementary material available at https:// doi. org/
10. 1038/ s41598- 022- 09497-y.
Correspondence and requests for materials should be addressed to Z.W.
Reprints and permissions information is available at www.nature.com/reprints.
Publisher’s note Springer Nature remains neutral with regard to jurisdictional claims in published maps and
institutional aliations.
Open Access is article is licensed under a Creative Commons Attribution 4.0 International
License, which permits use, sharing, adaptation, distribution and reproduction in any medium or
format, as long as you give appropriate credit to the original author(s) and the source, provide a link to the
Creative Commons licence, and indicate if changes were made. e images or other third party material in this
article are included in the article’s Creative Commons licence, unless indicated otherwise in a credit line to the
material. If material is not included in the article’s Creative Commons licence and your intended use is not
permitted by statutory regulation or exceeds the permitted use, you will need to obtain permission directly from
the copyright holder. To view a copy of this licence, visit http:// creat iveco mmons. org/ licen ses/ by/4. 0/.
© e Author(s) 2022
Content courtesy of Springer Nature, terms of use apply. Rights reserved
1.
2.
3.
4.
5.
6.
Terms and Conditions
Springer Nature journal content, brought to you courtesy of Springer Nature Customer Service Center GmbH (“Springer Nature”).
Springer Nature supports a reasonable amount of sharing of research papers by authors, subscribers and authorised users (“Users”), for small-
scale personal, non-commercial use provided that all copyright, trade and service marks and other proprietary notices are maintained. By
accessing, sharing, receiving or otherwise using the Springer Nature journal content you agree to these terms of use (“Terms”). For these
purposes, Springer Nature considers academic use (by researchers and students) to be non-commercial.
These Terms are supplementary and will apply in addition to any applicable website terms and conditions, a relevant site licence or a personal
subscription. These Terms will prevail over any conflict or ambiguity with regards to the relevant terms, a site licence or a personal subscription
(to the extent of the conflict or ambiguity only). For Creative Commons-licensed articles, the terms of the Creative Commons license used will
apply.
We collect and use personal data to provide access to the Springer Nature journal content. We may also use these personal data internally within
ResearchGate and Springer Nature and as agreed share it, in an anonymised way, for purposes of tracking, analysis and reporting. We will not
otherwise disclose your personal data outside the ResearchGate or the Springer Nature group of companies unless we have your permission as
detailed in the Privacy Policy.
While Users may use the Springer Nature journal content for small scale, personal non-commercial use, it is important to note that Users may
not:
use such content for the purpose of providing other users with access on a regular or large scale basis or as a means to circumvent access
control;
use such content where to do so would be considered a criminal or statutory offence in any jurisdiction, or gives rise to civil liability, or is
otherwise unlawful;
falsely or misleadingly imply or suggest endorsement, approval , sponsorship, or association unless explicitly agreed to by Springer Nature in
writing;
use bots or other automated methods to access the content or redirect messages
override any security feature or exclusionary protocol; or
share the content in order to create substitute for Springer Nature products or services or a systematic database of Springer Nature journal
content.
In line with the restriction against commercial use, Springer Nature does not permit the creation of a product or service that creates revenue,
royalties, rent or income from our content or its inclusion as part of a paid for service or for other commercial gain. Springer Nature journal
content cannot be used for inter-library loans and librarians may not upload Springer Nature journal content on a large scale into their, or any
other, institutional repository.
These terms of use are reviewed regularly and may be amended at any time. Springer Nature is not obligated to publish any information or
content on this website and may remove it or features or functionality at our sole discretion, at any time with or without notice. Springer Nature
may revoke this licence to you at any time and remove access to any copies of the Springer Nature journal content which have been saved.
To the fullest extent permitted by law, Springer Nature makes no warranties, representations or guarantees to Users, either express or implied
with respect to the Springer nature journal content and all parties disclaim and waive any implied warranties or warranties imposed by law,
including merchantability or fitness for any particular purpose.
Please note that these rights do not automatically extend to content, data or other material published by Springer Nature that may be licensed
from third parties.
If you would like to use or distribute our Springer Nature journal content to a wider audience or on a regular basis or in any other manner not
expressly permitted by these Terms, please contact Springer Nature at
onlineservice@springernature.com
... We queried USP32 in human tissues through the Human Protein Atlas website, the testis showed the greatest RNA expression of USP32 (Fig. 3). USP32 is upregulated in a variety of cancers, including small cell lung cancer [24], gastric cancer [25,26], breast cancer [23,27,28], epithelial ovarian cancer [29], glioblastoma [30], gastrointestinal stromal tumor [31], pancreatic duct adenocarcinoma [32] and acute myeloid leukemia [33]. Endogenous USP32 is found in the cytoplasm and membrane, according to the findings of a subcellular separation experiment [34] and a fluorescence protection experiment [23]. ...
... The elevated expression of USP32 protein in GBM tissues was validated by a study. Glioblastoma cell lines (Umur118 MG, Umur87 MG, A172, T98G, and Umur251 MG) had higher mRNA and protein levels of USP32 than normal brain cells SVG p12 [30]. In addition, they found that the higher the expression of USP32 in GBM, the worse the prognosis. ...
... Cancer cell proliferation and migration can be stopped in vitro by inhibiting USP32, and tumor growth can be stopped in vivo by down-regulating the USP32 gene. In their research, they discovered that USP32 can speed up the transition of the cell cycle from the G0 phase to the G1 phase [30], start DNA replication, and so enhance the growth of cancer cells. Then, RT-qPCR tests were used to confirm the effects of USP32 on a few functional molecules. ...
Article
Full-text available
An essential protein regulatory system in cells is the ubiquitin-proteasome pathway. The substrate is modified by the ubiquitin ligase system (E1-E2-E3) in this pathway, which is a dynamic protein bidirectional modification regulation system. Deubiquitinating enzymes (DUBs) are tasked with specifically hydrolyzing ubiquitin molecules from ubiquitin-linked proteins or precursor proteins and inversely regulating protein degradation, which in turn affects protein function. The ubiquitin-specific peptidase 32 (USP32) protein level is associated with cell cycle progression, proliferation, migration, invasion, and other cellular biological processes. It is an important member of the ubiquitin-specific protease family. It is thought that USP32, a unique enzyme that controls the ubiquitin process, is closely linked to the onset and progression of many cancers, including small cell lung cancer, gastric cancer, breast cancer, epithelial ovarian cancer, glioblastoma, gastrointestinal stromal tumor, acute myeloid leukemia, and pancreatic adenocarcinoma. In this review, we focus on the multiple mechanisms of USP32 in various tumor types and show that USP32 controls the stability of many distinct proteins. Therefore, USP32 is a key and promising therapeutic target for tumor therapy, which could provide important new insights and avenues for antitumor drug development. The therapeutic importance of USP32 in cancer treatment remains to be further proven. In conclusion, there are many options for the future direction of USP32 research.
... Aberrant expression of USP32 triggers certain diseases such as Parkinson's, Fragile X Syndrome, Chronic Kidney Disease and Cancer [11,[17][18][19]. Recent research has demonstrated the high expression of USP32 in a range of cancers [20][21][22][23][24][25][26] and its role in the initiation and progression of cancer. ...
... For example, USP32 in uences the invasion, migration, and proliferation of small cell lung cancer [21]; USP32 affects the development of epithelial ovarian cancer [20], glioblastoma [22], and gastric cancer [23] through the regulation of related proteins. USP32 interacts with Rab35 due to the deubiquitinating enzyme activity of USP32, therefore promoting the mesenchymal malignancies in the gastrointestinal tract to acquire imatinib resistance [27]. ...
Preprint
Full-text available
The regulatory significance of ubiquitin-specific peptidase 32 (USP32) in tumor is significant, nevertheless, the biological roles and regulatory mechanisms of USP32 in non-small cell lung cancer (NSCLC) remain unclear. According to our research, USP32 was strongly expressed in NSCLC cell lines and tissues and was linked to a bad prognosis for NSCLC patients. Interference with USP32 resulted in a significant inhibition of NSCLC cell proliferation, migration potential, and EMT development; on the other hand, USP32 overexpression had the opposite effect. To further elucidate the mechanism of action of USP32 in NSCLC, we screened H1299 cells for interacting proteins and found that USP32 interacts with BAG3 (Bcl2-associated athanogene 3) and deubiquitinates and stabilizes BAG3 in a deubiquitinating activity-dependent manner. Functionally, restoration of BAG3 expression abrogated the antitumor effects of USP32 silencing. Furthermore, USP32 increased the phosphorylation level of the RAF/MEK/ERK signaling pathway in NSCLC cells by stabilizing BAG3. In summary, these findings imply that USP32 is critical to the development of NSCLC and could offer a theoretical framework for the clinical diagnosis and management of NSCLC patients in the future.
... USP32 belongs to the ubiquitin-specific protease family, deubiquitinating enzymes which have been reported to be involved in several cancer initiation and progression (ovarian cancer [34], gastric cancer [35], glioblastoma [36], breast cancer [37], small cell lung cancer [38]). A recent work suggests a pivotal role of USP32 in pancreatic ductal adenocarcinoma given its higher expression levels compared to normal pancreatic tissues and a significant association with tumor grade and stage [39]. ...
... However, these data must be considered preliminary and need to be confirmed on a larger sample of patients. The two other patients who were followed-up after medical treatment without statistically significant USP32 Ovarian cancer Overexpressed [34] Gastric cancer Overexpressed [35] Glioblastoma Overexpressed [36] Breast cancer Overexpressed [37] Small cell lung cancer Overexpressed [38] Pancreatic ductal adenocarcinoma ...
Article
Full-text available
Background Neuroendocrine tumors (NETs) early diagnosis is a clinical challenge that require a deep understanding of molecular and genetic features of this heterogeneous group of neoplasms. However, few biomarkers exist to aid diagnosis and to predict prognosis and treatment response. In the oncological field, tumor-educated platelets (TEPs) have been implicated as central players in the systemic and local responses to tumor growth, thereby altering tumor specific RNA profile. Although TEPs have been found to be enriched in RNAs, few studies have investigated the potential of a type of RNA, circular RNAs (circRNA), as platelet-derived biomarkers for cancer. In this proof-of-concept study, we aim to demonstrate whether the circRNAs signature of tumor educated platelets can be used as a liquid biopsy biomarker for the detection of gastroenteropancreatic (GEP)-NETs and the prediction of the early response to treatment. Methods We performed a 24-months, prospective proof-of-concept study in men and women with histologically proven well-differentiated G1-G2 GEP-NET, aged 18–80 years, naïve to treatment. We performed a RNAseq analysis of circRNAs obtained from TEPs samples of 10 GEP-NETs patients at baseline and after 3 months from therapy (somatostatin analogs or surgery) and from 5 patients affected by non-malignant endocrinological diseases enrolled as a control group. Results Statistical analysis based on p < 0.05 resulted in the identification of 252 circRNAs differentially expressed between GEP-NET and controls of which 109 were up-regulated and 143 were down-regulated in NET patients. Further analysis based on an FDR value ≤ 0.05 resulted in the selection of 5 circRNAs all highly significant downregulated. The same analysis on GEP-NETs at baseline and after therapy in 5 patients revealed an average of 4983 remarkably differentially expressed circRNAs between follow-up and baseline samples of which 2648 up-regulated and 2334 down-regulated, respectively. Applying p ≤ 0.05 and FDR ≤ 0.05 filters, only 3/5 comparisons gave statistically significant results. Conclusions Our findings identified for the first time a circRNAs signature from TEPs as potential diagnostic and predictive biomarkers for GEP-NETs.
... In 2021, USP32 was found to be overexpressed in epithelial ovarian cancer (EOC), particularly in metastatic peritoneal tumors, and it positively regulates the proliferation and epithelial-mesenchymal transition (EMT) capabilities of cancer cells [9]. In 2022 and 2023, researchers confirmed that USP32 act as an oncogene in glioblastoma, gastrointestinal stromal tumors (GISTs) and acute myeloid leukemia through performing in vivo and in vitro experiments [10][11][12]. Through a series of preliminary pan-cancer analyses, we discovered that USP32 is significantly overexpressed in several cancer types such as BC, cholangiocarcinoma, esophageal carcinoma, head and neck squamous cell carcinoma, HCC and GC (Fig. 1A). ...
Article
Full-text available
Background Ubiquitin-specific protease 32 (USP32) is a highly conserved gene that promotes cancer progression. However, its role in hepatocellular carcinoma (HCC) is not well understood. The aim of this project is to explore the clinical significance and functions of USP32 in HCC. Methods The expression of USP32 in HCC was evaluated using data from TCGA, GEO, TISCH, tissue microarray, and human HCC samples from our hospital. Survival analysis, PPI analysis and GSEA analysis were performed to evaluate USP32-related clinical significance, key molecules and enrichment pathways. Using the ssGSEA algorithm and TIMER, we investigated the relationships between USP32 and immune infiltrates in the TME. Univariate and multivariate Cox regression analyses were then used to identify key USP32-related immunomodulators and constructed a USP32-related immune prognostic model. Finally, CCK8, transwell and colony formation assays of HCC cells were performed and an HCC nude mouse model was established to verify the oncogenic role of USP32. Results USP32 is overexpressed in HCC and its expression is an independent predictive factor for outcomes of HCC patients. USP32 is associated with pathways related to cell behaviors and cancer signaling, and its expression is significantly correlated with the infiltration of immune cells in the TME. We also successfully constructed a USP32-related immune prognostic model using 5 genes. Wet experiments confirmed that knockdown of USP32 could repress the proliferation, colony formation and migration of HCC cells in vitro and inhibit tumor growth in vivo. Conclusion USP32 is highly expressed in HCC and closely correlates with the TME of HCC. It is a potential target for improving the efficacy of chemotherapy and developing new strategies for targeted therapy and immunotherapy in HCC.
... Ubiquitin-specific protease 32 (USP32) is recognized as a new member of the ubiquitin-specific proteases subfamily. It alters protein stability and localization, thereby regulating their activity in different pathological stages of many human diseases, like cancer (6)(7)(8). So far, it has been reported that USP32 was over-expressed in lung and breast cancers, enhancing cellular proliferation and tissue metastasis (9). ...
Article
Full-text available
Objective: Gastric cancer is the fifth most common neoplasm and the fourth reason for mortality globally. Incidence rates are highly variable and dependent on risk factors, epidemiologic and carcinogenesis patterns. Previous studies reported that Helicobacter pylori (H. pylori) infection is one the strongest known risk factor for gastric cancer. USP32 is a deubiquitinating enzyme identified as a potential factor associated with tumor progression and a key player in cancer development. On the other hand, SHMT2 is involved in serine-glycine metabolism to support cancer cell proliferation. Both USP32 and SHMT2 are reported to be upregulated in many cancer types, including gastric cancer, but its complete mechanism is not fully explored yet. The present study explored possible mechanism of action of USP32 and SHMT2 in the progression of gastric cancer. Materials and methods: In this experimental study, Capsaicin (0.3 g/kg/day) and H. pylori infection combination was used to successfully initiate gastric cancer conditions in mice. It was followed by 40 and 70 days of treatment to establish initial and advanced conditions of gastric cancer. Results: Histopathology confirmed formation of signet ring cell and initiation of cellular proliferation in the initial gastric cancer. More proliferative cells were also observed. In addition, tissue hardening was confirmed in the advanced stage of gastric cancer. USP32 and SHMT2 showed progressive upregulated expression, as gastric cancer progress. Immunohistologically, it showed signals in abnormal cells and high-intensity signals in the advanced stage of cancer. In USP32 silenced tissue, expression of SHMT2 was completely blocked and reverted cancer development as evident with less abnormal cell in initial gastric cancer. Reduction of SHMT2 level to one-fourth was observed in the advanced gastric cancer stages of USP32 silenced tissue. Conclusion: USP32 had a direct role in regulating SHMT2 expression, which attracted therapeutic target for future treatment.
... However, an open question is how RAB7 monoubiquitination contributes to USP32 depletion-dependent defects in EGFR degradation and tumorigenesis. Notably, our and others' work has already linked USP32 to breast and other cancers, highlighting the implications of deregulated USP32 function [19][20][21]. ...
Article
Full-text available
Behind the scenes of signaling cascades initiated by activated receptors, endocytosis determines the fate of internalized proteins through degradation in lysosomes or recycling. Over the years, significant progress has been made in understanding the mechanisms of endocytosis and deregulation in disease states. Here we review the role of the EGF-SNX3-EGFR axis in breast cancers with an extended discussion on deregulated EGFR endocytosis in cancer.
... USP32 was reported to be one protein that contributed to the chimeric Tre2 (USP6) oncogene [154]. The pro-cancer effects of USP32 have been observed in breast cancer and glioblastoma [155,156]. Additionally, USP32 promotes GC cell growth, metastasis, and chemoresistance by upregulating SMAD2, an important protein in the TGF-β signaling pathway [55]. However, it is unclear whether USP32 regulates the ubiquitination level of SMAD2. ...
Article
Full-text available
Gastric cancers (GCs) are malignant tumors with a high incidence that threaten global public health. Despite advances in GC diagnosis and treatment, the prognosis remains poor. Therefore, the mechanisms underlying GC progression need to be identified to develop prognostic biomarkers and therapeutic targets. Ubiquitination, a post-translational modification that regulates the stability, activity, localization, and interactions of target proteins, can be reversed by deubiquitinases (DUBs), which can remove ubiquitin monomers or polymers from modified proteins. The dysfunction of DUBs has been closely linked to tumorigenesis in various cancer types, and targeting certain DUBs may provide a potential option for cancer therapy. Multiple DUBs have been demonstrated to function as oncogenes or tumor suppressors in GC. In this review, we summarize the DUBs involved in GC and their associated upstream regulation and downstream mechanisms and present the benefits of targeting DUBs for GC treatment, which could provide new insights for GC diagnosis and therapy.
Article
Full-text available
Introduction Replication of the nuclear genome is an essential step for cell division. Pathogenic variants in genes coding for highly conserved components of the DNA replication machinery cause Meier-Gorlin syndrome (MGORS). Objective Identification of novel genes associated with MGORS. Methods Exome sequencing was performed to investigate the genotype of an individual presenting with prenatal and postnatal growth restriction, a craniofacial gestalt of MGORS and coronal craniosynostosis. The analysis of the candidate variants employed bioinformatic tools, in silico structural protein analysis and modelling in budding yeast. Results A novel homozygous missense variant NM_016095.2:c.341G>T, p.(Arg114Leu), in GINS2 was identified. Both non-consanguineous healthy parents carried this variant. Bioinformatic analysis supports its classification as pathogenic. Functional analyses using yeast showed that this variant increases sensitivity to nicotinamide, a compound that interferes with DNA replication processes. The phylogenetically highly conserved residue p.Arg114 localises at the docking site of CDC45 and MCM5 at GINS2. Moreover, the missense change possibly disrupts the effective interaction between the GINS complex and CDC45, which is necessary for the CMG helicase complex (Cdc45/MCM2–7/GINS) to accurately operate. Interestingly, our patient’s phenotype is strikingly similar to the phenotype of patients with CDC45 -related MGORS, particularly those with craniosynostosis, mild short stature and patellar hypoplasia. Conclusion GINS2 is a new disease-associated gene, expanding the genetic aetiology of MGORS.
Article
Full-text available
Gliomas constitute the most frequent tumors of the brain. High-grade gliomas are characterized by a poor prognosis caused by a set of attributes making treatment difficult, such as heterogeneity and cell infiltration. Additionally, there is a subgroup of glioma cells with properties similar to those of stem cells responsible for tumor recurrence after treatment. Since proteasomal degradation regulates multiple cellular processes, any mutation causing disturbances in the function or expression of its elements can lead to various disorders such as cancer. Several studies have focused on protein degradation modulation as a mechanism of glioma control. The ubiquitin proteasome system is the main mechanism of cellular proteolysis that regulates different events, intervening in pathological processes with exacerbating or suppressive effects on diseases. This review analyzes the role of proteasomal degradation in gliomas, emphasizing the elements of this system that modulate different cellular mechanisms in tumors and discussing the potential of distinct compounds controlling brain tumorigenesis through the proteasomal pathway.
Article
Full-text available
Glioblastoma is the most common primary brain tumor in adults with poor overall outcome and 5-year survival of less than 5%. Treatment has not changed much in the last decade or so, with surgical resection and radio/chemotherapy being the main options. Glioblastoma is highly heterogeneous and frequently becomes treatment-resistant due to the ability of glioblastoma cells to adopt stem cell states facilitating tumor recurrence. Therefore, there is an urgent need for novel therapeutic strategies. The ubiquitin system, in particular E3 ubiquitin ligases and deubiquitinating enzymes, have emerged as a promising source of novel drug targets. In addition to conventional small molecule drug discovery approaches aimed at modulating enzyme activity, several new and exciting strategies are also being explored. Among these, PROteolysis TArgeting Chimeras (PROTACs) aim to harness the endogenous protein turnover machinery to direct therapeutically relevant targets, including previously considered “undruggable” ones, for proteasomal degradation. PROTAC and other strategies targeting the ubiquitin proteasome system offer new therapeutic avenues which will expand the drug development toolboxes for glioblastoma. This review will provide a comprehensive overview of E3 ubiquitin ligases and deubiquitinating enzymes in the context of glioblastoma and their involvement in core signaling pathways including EGFR, TGF-β, p53 and stemness-related pathways. Finally, we offer new insights into how these ubiquitin-dependent mechanisms could be exploited therapeutically for glioblastoma.
Article
Full-text available
KEGG (https://www.kegg.jp/) is a manually curated resource integrating eighteen databases categorized into systems, genomic, chemical and health information. It also provides KEGG mapping tools, which enable understanding of cellular and organism-level functions from genome sequences and other molecular datasets. KEGG mapping is a predictive method of reconstructing molecular network systems from molecular building blocks based on the concept of functional orthologs. Since the introduction of the KEGG NETWORK database, various diseases have been associated with network variants, which are perturbed molecular networks caused by human gene variants, viruses, other pathogens and environmental factors. The network variation maps are created as aligned sets of related networks showing, for example, how different viruses inhibit or activate specific cellular signaling pathways. The KEGG pathway maps are now integrated with network variation maps in the NETWORK database, as well as with conserved functional units of KEGG modules and reaction modules in the MODULE database. The KO database for functional orthologs continues to be improved and virus KOs are being expanded for better understanding of virus-cell interactions and for enabling prediction of viral perturbations.
Article
Full-text available
Metabolic reprogramming, including enhanced biosynthesis of macromolecules, altered energy metabolism, and maintenance of redox homeostasis, is considered a hallmark of cancer, sustaining cancer cell growth. Multiple signaling pathways, transcription factors and metabolic enzymes participate in the modulation of cancer metabolism and thus, metabolic reprogramming is a highly complex process. Recent studies have observed that ubiquitination and deubiquitination are involved in the regulation of metabolic reprogramming in cancer cells. As one of the most important type of post-translational modifications, ubiquitination is a multistep enzymatic process, involved in diverse cellular biological activities. Dysregulation of ubiquitination and deubiquitination contributes to various disease, including cancer. Here, we discuss the role of ubiquitination and deubiquitination in the regulation of cancer metabolism, which is aimed at highlighting the importance of this post-translational modification in metabolic reprogramming and supporting the development of new therapeutic approaches for cancer treatment.
Article
Full-text available
Background: Age is associated with the prognosis of glioma patients, but there is no uniform standard of age-group classification to evaluate the prognosis of glioma patients. In this study, we aimed to establish an age group classification for risk stratification in glioma patients. Methods: 1502 patients diagnosed with gliomas at Nanfang Hospital between 2000 and 2018 were enrolled. The WHO grade of glioma was used as a dependent variable to evaluate the effect of age on risk stratification. The evaluation model was established by logistic regression, and the Akaike information criterion (AIC) value of the model was used to determine the optimal cutoff points for age-classification. The differences in gender, WHO grade, pathological subtype, tumor cell differentiation, tumor size, tumor location, and molecular markers between different age groups were analyzed. The molecular markers included GFAP, EMA, MGMT, P53, NeuN, Oligo2, EGFR, VEGF, IDH1, Ki-67, PR, CD3, H3K27M, TS, and 1p/19q status. Results: The proportion of men with glioma was higher than that of women with glioma (58.3% vs 41.7%). Analysis of age showed that appropriate classifications of age group were 0-14 years old (pediatric group), 15-47 years old (youth group), 48-63 years old (middle-aged group), and ≥ 64 years old (elderly group).The proportions of glioblastoma and large tumor size (4-6 cm) increased with age (p = 0.000, p = 0.018, respectively). Analysis of the pathological molecular markers across the four age groups showed that the proportion of patients with larger than 10% area of Ki-67 expression or positive PR expression increased with age (p = 0.000, p = 0.017, respectively). Conclusions: Appropriate classifications of the age group for risk stratification are 0-14 years old (pediatric group), 15-47 years old (young group), 48-63 years old (middle age group) and ≥ 64 years old (elderly group). This age group classification is effective in evaluating the risk of glioblastoma in glioma patients.
Article
Full-text available
Since the discovery of the ubiquitin proteasome system (UPS), the roles of ubiquitinating and deubiquitinating enzymes (DUBs) have been widely elucidated. The ubiquitination of proteins regulates many aspects of cellular functions such as protein degradation and localization, and also modifies protein-protein interactions. DUBs cleave the attached ubiquitin moieties from substrates and thereby reverse the process of ubiquitination. The dysregulation of these two paramount pathways has been implicated in numerous diseases, including cancer. Attempts are being made to identify inhibitors of ubiquitin E3 ligases and DUBs that potentially have clinical implications in cancer, making them an important target in the pharmaceutical industry. Therefore, studies in medicine are currently focused on the pharmacological disruption of DUB activity as a rationale to specifically target cancer-causing protein aberrations. Here, we briefly discuss the pathophysiological and physiological roles of DUBs in key cancer-related pathways. We also discuss the clinical applications of promising DUB inhibitors that may contribute to the development of DUBs as key therapeutic targets in the future.
Article
Full-text available
Glioblastoma (GBM) generally has a dismal prognosis, and it is associated with a poor quality of life as the disease progresses. However, the development of effective therapies for GBM has been deficient. Ubiquitin-conjugating enzyme E2T (UBE2T) is a member of the E2 family in the ubiquitin-proteasome pathway and a vital regulator of tumour progression, but its role in GBM is unclear. In this study, we aimed to clarify the role of UBE2T in GBM. Bioinformatics analysis identified UBE2T as an independent risk factor for gliomas. Immunohistochemistry was used to measure UBE2T expression in GBM and normal tissue samples obtained from patients with GBM. The effects of UBE2T on GBM cell invasion and migration were analysed using the Transwell assay. BALB/c nude mice were used for the in vivo assays. Immunoblotting and immunoprecipitation were performed to determine the molecular mechanisms. UBE2T was highly expressed in GBM tissues, and its expression was linked to a poor prognosis. In vitro, depletion of UBE2T significantly suppressed cell invasion and migration. Moreover, UBE2T depletion suppressed the growth of GBM subcutaneous tumours in vivo. Further experiments revealed that UBE2T suppressed invasion and migration by regulating epithelial- mesenchymal transition (EMT) via stabilising GRP78 in GBM cells. We uncovered a novel UBE2T/GRP78/EMT regulatory axis that modulates the malignant progression and recurrence of GBM, indicating that the axis might be a valuable therapeutic target.
Article
Hepatocellular carcinoma (HCC) continues to cause severe burden worldwide. The limited options especially toward HCC with metastasis prompts us to identify novel molecules for either diagnostic/prognostic or therapeutic purposes. GRPEL2 is well defined in maintaining mitochondrial homeostasis, which is critical to multiple biological processes for cancer survival. However, its role in HCC progression was not investigated before. In our analysis using data from The Cancer Genome Atlas Liver Hepatocellular Carcinoma (TCGA LIHC) dataset and tissue microarray, higher expression levels of GRPEL2 were obseved in HCC tissues compared to in normal liver tissues, and indicated higher tumor grade, higher tumor stage, and shorter overall survival (OS). Consistent with the results of above analyses, the functional experiments validated that GRPEL2 acted as a tumor-promoting factor in HCC progression. GRPEL2 knockdown suppressed cell growth, migration, and invasion in vitro, as well as inhibited tumor growth in vivo. Moreover, GRPEL2 deficiency also accelerated reactive oxygen species (ROS) production and increased mitochondrial membrane potential (MMP), leading to cell apoptosis. In addition, we found that the cell cycle and NF-κB signaling pathways were responsible for GRPEL2-induced HCC progression, based on the results of Gene Set Enrichment Analysis (GSEA) and subsequent experimental validation. Our study, for the first time, identified the role of GRPEL2 in HCC development and provided a compelling biomarker for targted therapy in HCC treatment.
Article
Epithelial ovarian cancer (EOC) is the seventh most common cancer worldwide and the deadliest gynecological malignancy because of its aggressiveness and high recurrence rate. To discover new therapeutic targets for EOC, we combined public EOC microarray datasets with our previous in vivo shRNA screening dataset. The top-ranked gene ubiquitin specific peptidase 32 (USP32), coding a deubiquitinating enzyme, is a component of the ubiquitin proteasome system. Clinically, USP32 is expressed in primary ovarian cancer, especially in metastatic peritoneal tumors, and negatively impacts the survival outcome. USP32 regulates proliferative and epithelial mesenchymal transition capacities that are associated with EOC progression. Proteomic analysis identified farnesyl-diphosphate farnesyltransferase 1 (FDFT1) as a novel substrate of USP32 that is an enzyme in the mevalonate pathway, essentially associated with cell proliferation and stemness. USP32 and FDFT1 expression was higher in tumor spheres than in adherent cells. Inhibition of USP32, FDFT1, or mevalonate pathway considerably suppressed tumor sphere formation, which was restored by adding squalene, a downstream product of FDFT1. These findings suggested that USP32-FDFT1 axis contributes to EOC progression, and could be novel therapeutic targets for EOC treatment.