Open Access

MG132 induces cell type‑specific anticancer effects in uterine leiomyosarcoma cell lines

  • Authors:
    • Hosouk Joung
    • Suho Seo
    • Hyunju Liu
  • View Affiliations

  • Published online on: April 10, 2025     https://doi.org/10.3892/mmr.2025.13524
  • Article Number: 159
  • Copyright: © Joung 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

Uterine leiomyosarcoma (Ut‑LMS) is a rare and aggressive malignant tumor with limited therapeutic options. Therefore, exploration of novel treatment strategies is necessary. MG132 is a potent proteasome inhibitor that has shown promising potential in cancer therapy by inducing apoptosis through disruption of protein homeostasis. Despite its promising applications in various cancers, its effects on Ut‑LMS remains largely unexplored. Therefore, the present study investigated the effects of MG132 on Ut‑LMS cell lines (SK‑LMS‑1, SK‑UT‑1 and SK‑UT‑1B) in terms of cytotoxicity, apoptosis induction, cell cycle progression, autophagy and reactive oxygen species (ROS) production. Treatment with MG132 (0‑2 µM for 24 h) induced a dose‑dependent reduction in cell viability across all three cell lines, and the lactate dehydrogenase release assays confirmed membrane damage. Moreover, apoptosis induction was assessed using annexin V and 7‑AAD staining, which revealed dose‑dependent apoptosis in all three cell lines. Western blot analysis revealed increased cleaved poly‑adenosine diphosphate ribose polymerase and caspase‑3 levels, thereby indicating activation of the apoptotic pathway in response to MG132 treatment. MG132 also induced G2/M phase arrest in SK‑LMS‑1 and SK‑UT‑1 cells and altered the expression of cell cycle regulatory proteins, such as p21, p27 and p53. Furthermore, MG132 promoted autophagy in all three cell lines by increasing light chain 3 II levels. ROS levels remained unchanged in SK‑LMS‑1 cells but increased in SK‑UT‑1B and SK‑UT‑1 cells. Furthermore, the ROS scavenger N‑acetylcysteine effectively reduced MG132‑induced apoptosis in SK‑UT‑1 cells. These findings highlight the cytotoxicity of MG132 in Ut‑LMS cells, emphasize its potential as a therapeutic agent for Ut‑LMS, provide insights into its mechanisms of action, and suggest possible strategies for improving treatment efficacy.

Introduction

Uterine leiomyosarcoma (Ut-LMS) is a rare and aggressive malignant tumor that originates from the smooth muscle cells of the uterus (1). Ut-LMS constitutes approximately 1–2% of all uterine malignancies and is characterized by poor prognosis, high recurrence rates, and a tendency to metastasize (2,3). Pathologically, Ut-LMS is characterized by atypical spindle cells with considerable nuclear pleomorphisms, high mitotic activity, and areas of necrosis. These characteristics distinguish it from benign leiomyomas (fibroids) that lack malignant potential (4,5). Ut-LMS diagnosis typically requires histopathological examinations, which are often supplemented by immunohistochemical staining, to confirm the smooth muscle origin of the tumor cells (6). Clinically, patients with Ut-LMS may present with nonspecific symptoms, such as abnormal uterine bleeding, pelvic pain, and a palpable mass (1). Owing to its aggressive nature, Ut-LMS often presents at an advanced stage, and early diagnosis is challenging. Imaging techniques, including ultrasound, magnetic resonance imaging, and computed tomography, aid in the evaluation of tumor size, extent, and metastatic spread; however, definitive diagnosis relies on tissue biopsy (7).

Depending on the extent of the disease, Ut-LMS treatment primarily involves surgical resection, often in the form of total hysterectomy with or without bilateral salpingo-oophorectomy (1). Conservative treatment for patients who wish to preserve fertility may be an option; however, it requires close and intensive follow-up (8). Although laparoscopic approaches provide minimally invasive options, the abdominal incision approach remains commonly used owing to its lower cost and ease of application (9). Despite surgical intervention, the high recurrence rate of Ut-LMS necessitates additional therapeutic strategies. Owing to the limited data on this rare tumor, preoperative diagnosis is challenging, and uncertainty regarding optimal postoperative management render treatment decisions complex (8). Adjuvant therapies, including radiation and chemotherapy, have been explored; however, their efficacy remains limited, and the optimal treatment regimen remains a subject of ongoing research (10). Therefore, factors, such as safety, fertility preservation, long-term prognosis, and cost should be carefully considered when determining the best surgical approach (8).

Proteasome inhibitors are a class of compounds that target the ubiquitin-proteasome system, which is a critical pathway for degrading misfolded, damaged, or unnecessary proteins within cells (11). By inhibiting proteasome activity, these inhibitors disrupt cellular protein homeostasis and trigger stress responses, which results in programmed cell death (apoptosis) in some cases (12). MG132 is a potent peptide aldehyde-based inhibitor that primarily blocks the chymotrypsin-like activity of proteasomes, thereby resulting in accumulation of ubiquitinated proteins (13,14). Hence, MG132 is widely used in experimental settings to study proteasome function, apoptosis induction, and stress response pathways (15,16). Furthermore, MG132 is utilized to investigate signaling mechanisms, such as nuclear factor kappa B (NF-κB) pathway inhibition, which is crucial in inflammation and immune responses (17,18).

Clinically, proteasome inhibitors, including MG132, have shown notable therapeutic potential, particularly in cancer treatment (19,20). Moreover, MG132 serves as a foundational compound for developing Food and Drug Administration (FDA)-approved drugs, such as bortezomib and carfilzomib, which are used to treat multiple myeloma and mantle cell lymphoma (21). However, in patients with metastatic sarcomas, including leiomyosarcoma, single-agent bortezomib showed minimal efficacy and led to early study termination after the first stage of patient accrual (22). In addition to oncology, MG132 has been used in preclinical studies to explore its role in neurodegenerative diseases, such as Alzheimer's and Parkinson's disease, where protein aggregation is a hallmark (23,24). Furthermore, its anti-inflammatory properties and ability to modulate oxidative stress suggest its potential application in the treatment of inflammatory, cardiovascular, and metabolic disorders (2527). Accordingly, continued research on MG132 and its related inhibitors may uncover new therapeutic avenues and deepen our understanding of proteasome biology.

However, the effect of MG132 on the growth of Ut-LMS remains poorly understood. Therefore, this study aimed to investigate the effects of MG132 on Ut-LMS cells by focusing on the relevant molecular mechanisms and cellular responses. By elucidating the underlying processes, this study will provide valuable insights that will contribute to the development of more effective therapeutic strategies for Ut-LMS. This will ultimately enhance patient outcomes and offer renewed hope to individuals affected by this challenging malignancy.

Materials and methods

Cell lines and reagents

Human Ut-LMS cancer cell lines (SK-LMS-1, SK-UT-1, and SK-UT-1B) were obtained from the American Type Culture Collection (ATCC, Manassas, VA, USA). These cells were cultured in Minimal Essential Medium (LM007-07; Gyeongsan, South Korea, Welgene) supplemented with 10% fetal bovine serum (SH30919.03; Hyclone, Logan, UT, USA) and 1% streptomycin-penicillin (15140122; Gibco, Waltham, MA, USA) and were maintained in a humidified incubator at 37°C and 5% CO2. MG132 was obtained from Selleckchem (S2619; Houston, TX, USA), and dimethyl sulfoxide (DMSO; LPS Solution) was used as the control. N-acetyl-L-cysteine (NAC; A7250) and catalase (CAT; C1345) were purchased from Sigma-Aldrich (St. Louis, MI, USA).

The 2,5-diphenyl-2H-tetrazolium bromide (MTT) assay

Cells were seeded in 96-well plates at a density of 5,000 cells/well and allowed to adhere overnight. After treatment with various concentrations of MG132 for 24 h, 20 µl of MTT solution (5 mg/ml, M2128; Sigma-Aldrich) was added to each well and incubated for 2 h at 37°C. The resulting formazan crystals were dissolved by adding 150 µl of DMSO to each well, and the absorbance was measured at 570 nm using an INNO microplate spectrophotometer (LTek, Seongnam, South Korea). Cell viability was calculated as a percentage of the untreated control group.

Lactate dehydrogenase (LDH) release assay

The cells were cultured in 96-well plates and treated with MG132 for 24 h. After treatment, LDH release was measured using a Dyne LDH Plus Cytotoxicity Assay Kit (GBL-P500; Dyne Bio, Seongnam, South Korea) following the manufacturer's instructions. LDH PLUS Reaction Mixture (100 µl) was added to each well, and the plate was gently mixed. Thereafter, the reaction was allowed to proceed in the dark for 30 min at room temperature. The absorbance of the samples was measured at 490 nm using a microplate reader. Absorbance values were normalized to those of the control group, thus allowing for comparison between treated and untreated samples.

Protein preparation and western blotting analysis

Ut-LMS cells were treated with varying concentrations of MG132 (0–2 µM) for 24 h. Following treatment, the cells were lysed using radioimmunoprecipitation assay buffer (R2002; Biosesang, Yongin, South Korea) supplemented with a protease inhibitor cocktail (04693132001; Roche, Basel, Switzerland). Subsequently, equal amounts of protein were denatured by heating and were resolved using 12–15% sodium dodecyl-sulfate polyacrylamide gel electrophoresis. The separated proteins were transferred onto polyvinylidene fluoride membranes (IPVH00010; Merck Millipore, Burlington, MA, USA) and blocked with 5% skim milk (262100; BD Difco, Franklin Lakes, NJ, USA) for 1 h at room temperature. The membranes were then incubated overnight at 4°C with gentle shaking using primary antibodies. The following primary antibodies were used: caspase-3 (9665S), cleaved caspase-3 (9664S), caspase-9 (9502S), poly-adenosine diphosphate ribose polymerase (PARP; 9542S), p21 (2947S), p27 (3686T), p53 (2527S), light chain 3 (LC3) beta (2775S), β-actin (4967S; Cell Signaling Technology, Danvers, MA, USA), and ubiquitin (BML-PW0930; Enzo Biochem Inc., Farmingdale, NY, USA). After washing with Tris-buffered saline with 0.1% Tween® 20 detergent, membranes were incubated with horseradish peroxidase-conjugated secondary antibodies (7074S or 7076S; Cell Signaling Technology) for 1 h at room temperature. Protein signals were detected using the Western Pico ECL Kit (PICO-250; LPS Solution) and visualized using an Azure c280 chemiluminescent imaging system. The intensity of the protein bands was quantified using ImageJ version 1.53a software to analyze relative protein expression levels.

Apoptotic profile analysis

The apoptotic profile of Ut-LMS cells was assessed using the Muse Annexin V & Dead Cell Kit (MCH100105; Luminex, Austin, TX, USA) according to the manufacturer's instructions. Ut-LMS cells were seeded in six-well plates at a density of 5×10 cells/well and treated with various concentrations of MG132 for 24 h. After treatment, the cells were detached using trypsin, harvested by centrifugation, and resuspended in 100 µl of fresh medium. Subsequently, 100 µl of Muse Annexin V & Dead Cell reagent was added, and the mixture was incubated for 20 min at room temperature in the dark. The stained cells were then analyzed using a Guava Muse Cell Analyzer to evaluate apoptotic and dead cell populations.

Flow cytometric evaluation of cell cycle distribution

Cell cycle distribution was analyzed using the Muse Cell Cycle Kit (MCH100106; Luminex) following the manufacturer's protocol. Cells were harvested, centrifuged to form a pellet, and fixed in 70% ethanol at −20°C for 3 h. After fixation, the cells were stained with the Muse Cell Cycle Reagent and incubated in the dark for 30 min. Thereafter, the stained samples were analyzed using a Guava Muse Cell Analyzer to evaluate the distribution of cells across different cell cycle phases.

Assessment of autophagy using flow cytometry

Autophagy induction by MG132 was assessed using the Muse Autophagy LC3-Antibody Based Kit (MCH200109; Luminex) and Muse Cell Analyzer following the manufacturer's instructions. Both treated and untreated cells were cultured, detached, and incubated with anti-LC3 Alexa Fluor 555 and 1X Autophagy Reagent for 30 min in the dark. After incubation, the cells were resuspended in 1X assay buffer and analyzed using a Muse Cell Analyzer. The level of autophagy was then quantified by calculating the ratio of fluorescence intensity in the treated samples relative to the controls.

ROS quantification using flow cytometry

ROS production was quantified using flow cytometry with the Muse Oxidative Stress Kit (cat. no. MCH100111; Luminex) according to manufacturer's instructions. Cells were seeded at 5×104 cells/well in six-well plates and cultured at 37°C for 24 h. The cells were then treated with 0, 0.25, 0.5, or 1 µM MG132, diluted in fresh medium, and incubated for 24 h. After treatment, the cells were detached, resuspended in 1X Assay Buffer, and incubated with the Muse Oxidative Stress Reagent working solution for 30 min at 37°C. Samples were then analyzed using a Guava Muse Cell Analyzer (cat. no. 0500-3115; Luminex), and the data were processed using Muse analysis software (version 1.5; Luminex).

Statistical analyses

Statistical analyses were performed using GraphPad Prism software (version 8.0). Data are expressed as the mean ± standard deviation. One-way analysis of variance was used to compare multiple groups, followed by Tukey's post hoc test or Dunnett's multiple comparison test, as appropriate. Statistical significance was set at *P<0.05 and **P<0.01.

Results

Assessment of MG132 cytotoxicity in Ut-LMS cell lines

To evaluate the cytotoxic effects of MG132 (Fig. 1A), Ut-LMS cell lines (SK-LMS-1, SK-UT-1B, and SK-UT-1) were treated with varying concentrations (0–1 or 2 µM) of MG132 for 24 h. Cytotoxicity was then evaluated using the MTT and LDH release assays. As shown in Fig. 1B, all three Ut-LMS cell lines exhibited a dose-dependent decrease in viability as MG132 concentrations increased. This indicated that MG132 effectively reduced cell viability in a concentration-dependent manner. Additionally, LDH release assays were conducted on Ut-LMS cells treated with MG132 for 24 h, which confirmed enhanced membrane damage. As the concentration of MG132 increased, all three cell lines showed a considerable increase in LDH release (Fig. 1C). Moreover, a relative increase in ubiquitinated proteins was observed in Ut-LMS cells treated with 0.5 and 1 µM MG132 for 24 h compared with those in control cells (Fig. S1). This indicated that Ut-LMS cell lines experienced reduced cell viability and membrane damage. These findings demonstrate that MG132 induces cytotoxicity in Ut-LMS cells, rendering it a promising therapeutic agent for uterine smooth muscle tumors.

Assessment of apoptosis induction by MG132 in Ut-LMS cell lines

To investigate the pro-apoptotic effects of MG132 in Ut-LMS cells, apoptotic markers were examined using Annexin V and 7-AAD staining via flow cytometry after treating SK-LMS-1, SK-UT-1B, and SK-UT-1 cell lines with MG132 (0, 0.25, 0.5, or 1 µM) for 24 h. Apoptosis markedly increased in a dose-dependent manner upon MG132 treatment in all three cell lines (Fig. 2A). To further confirm apoptosis induction, western blot analysis was performed to evaluate the expression of apoptosis-related proteins including PARP, caspase-3, and caspase-9. The concentrations of MG132 (0.5 and 1 µM) were carefully selected based on the cell viability data. Following MG132 treatment, increased cleaved PARP and cleaved caspase-3 levels were observed in all three cell lines, whereas no changes in cleaved caspase-9 levels were detected in any of the cell lines (Fig. 2B). These results confirmed that MG132 induced apoptosis via activation of apoptosis-related markers in all three Ut-LMS cell lines.

Effects of MG132 on cell cycle progression and regulatory proteins in Ut-LMS cells

To investigate the impact of MG132 on cell cycle progression in Ut-LMS cells, cell cycle distribution was analyzed using propidium iodide staining, followed by flow cytometry after treating the cells with varying concentrations of MG132 for 24 h. As shown in Fig. 3A, MG132 treatment induced a G2/M phase arrest at concentrations of 0.5 and 1 µM in SK-LMS-1 and SK-UT-1 cells, whereas no significant changes in cell cycle distribution were observed in SK-UT-1B cells. Western blot analysis was performed to evaluate the expression of cell cycle regulatory proteins, including p21, p27, and p53, in Ut-LMS cells (Fig. 3B). The p21 expression was markedly upregulated by MG132 in all three cell lines. In contrast, p27 levels pronouncedly decreased in all three cell lines following MG132 treatment. However, a differential response was observed in p53 expression; its expression decreased in SK-LMS-1 cells, increased in SK-UT-1B cells, and showed no significant changes in SK-UT-1 cells. These findings suggest that MG132 differentially regulates cell cycle progression and expression of cell cycle-related proteins in Ut-LMS cell lines, thereby highlighting the distinct cellular responses to treatment.

Effects of MG132 on autophagy in Ut-LMS cell lines

Autophagy is an essential cellular mechanism that maintains homeostasis under normal and stressful conditions (28). To assess the induction of autophagy, LC3 protein levels, a marker of autophagosome formation, were evaluated using flow cytometry and western blotting. Following MG132 treatment at the indicated concentrations (0.5 and 1 µM) for 24 h, flow cytometry analysis revealed an increase in autophagy induction in SK-LMS-1, SK-UT-1B, and SK-UT-1 cells (Fig. 4A). Consistent with the flow cytometry results, western blot analysis showed elevated LC3 II levels in all three cell lines, thereby indicating the conversion of LC3 I to LC3 II, which is a hallmark of autophagy induction (Fig. 4B). These findings suggest that MG132 promotes autophagy in Ut-LMS cells, thus potentially contributing to its cellular effects.

Effects of MG132 on intracellular ROS levels in Ut-LMS cells

Excessive ROS accumulation in cells leads to oxidative stress, which damages nucleic acids, lipids, proteins, membranes, and mitochondria (29). To assess whether MG132 increases ROS levels in Ut-LMS cells, cells were treated with varying concentrations of MG132 (0.5 and 1 µM) for 24 h, followed by flow cytometry analysis. In SK-LMS-1 cells, no significant changes in ROS levels were observed after MG132 treatment (Fig. 5A, upper panels). Based on the results shown in Fig. 2A and these findings, apoptosis induced by MG132 in SK-LMS-1 cells appeared to occur independently of ROS production. However, ROS levels were increased in SK-UT-1B and SK-UT-1 cells treated with MG132 at the indicated concentrations for 24 h (Fig. 5A, middle and bottom panels). Therefore, to investigate whether ROS production contributes to MG132-induced apoptosis in SK-UT-1B and SK-UT-1 cells, cells were treated with MG132 (1 µM) with or without the ROS scavengers NAC and CAT (30,31). In SK-UT-1B cells, NAC and CAT had no significant effect on MG132-induced apoptosis (Fig. S2A). However, NAC effectively reduced MG132-induced apoptosis (Fig. 5B), whereas CAT had no significant effect (Fig. S2B) in SK-UT-1 cells. These results suggest that MG132 induced ROS-mediated apoptosis in SK-UT-1 cells, whereas MG132-induced apoptosis in SK-LMS-1 and SK-UT-1B cells occurs in an ROS-independent manner.

Discussion

The findings of this study (Fig. 6) provide valuable insights into the molecular mechanisms underlying the effects of MG132 on Ut-LMS cell lines. The study emphasizes the therapeutic potential of MG132 and the distinct responses observed in different cell lines. Ut-LMS is a rare and aggressive malignancy of the uterine smooth muscle cells, characterized by poor prognosis and considerable challenges in treatment and management. Thus, the study findings contribute to the growing understanding of this disease and offer new perspectives on potential therapeutic strategies for difficult-to-treat malignancies.

This study demonstrated that MG132 induces various forms of cell death in Ut-LMS cell lines. MG132 induced cytotoxicity in SK-LMS-1, SK-UT-1B, and SK-UT-1 cells, with a dose-dependent decrease in cell viability and increased LDH release observed in all three cell lines. Furthermore, MG132 treatment affected cell cycle progression, resulting in G2/M arrest in SK-LMS-1 and SK-UT-1 cells. However, no significant changes were observed in SK-UT-1B cells. In addition, MG132 induced apoptosis, with ROS-independent apoptosis observed in SK-LMS-1 and SK-UT-1B cells, and ROS-dependent apoptosis in SK-UT-1 cells. Additionally, MG132 differentially regulated the expression of cell cycle proteins (p21, p27, and p53) in each cell line and induced autophagy, as evidenced by increased LC3 II levels. This suggests that MG132 simultaneously activates multiple pathways, thereby disrupting key survival mechanisms.

This study also revealed crucial differences in protein expression and cellular behavior across these cell lines. MG132 treatment decreased p53 expression in SK-LMS-1 cells, whereas it increased p53 expression in SK-UT-1B cells. Furthermore, ROS analysis demonstrated variability; SK-LMS-1 cells exhibited higher basal ROS levels, whereas MG132 treatment elevated ROS levels in SK-UT-1B and SK-UT-1 cells solely. These findings underscore the molecular heterogeneity of Ut-LMS cell lines and align with previous reports of variability in cell doubling times and tumor formation rates among these models (32).

The differences between MG132-treated SK-LMS-1, SK-UT-1, and SK-UT-1 cell lines in this study could be attributed to several key factors. A detailed comparison of the cell line characteristics based on ATCC product sheets supports these observations. SK-UT-1B and SK-UT-1, which are derived from the uterus of a 75-year-old female, differ in pathology. SK-UT-1B is associated with Grade III endometrial leiomyosarcoma (LMS), whereas SK-UT-1 originates from a mesodermal mixed tumor. In contrast, SK-LMS-1 cells, which are derived from the vulva of a 43-year-old female patient with LMS, exhibited fibroblast morphology indicative of its stromal tissue origin. However, SK-UT-1B and SK-UT-1 display epithelial morphology consistent with uterine and endometrial tissues. Therefore, these differences in tissue origin are likely to contribute to variations in molecular pathways that subsequently influence drug responses. For example, the expression levels of key proteins, such as p21 and p27, and ROS levels, play key roles in the response of each cell line to MG132-induced proteasome inhibition and stress. Additionally, according to the ATCC product sheet, SK-UT-1B is a subclone derived from SK-UT-1. Although SK-UT-1, similar to SK-LMS-1, harbors two independent p53 point mutations, some reports suggest that SK-UT-1B retains wild-type p53 (33,34). Owing to these conflicting findings, the p53 status of SK-UT-1B remains unclear, and further sequencing or validation may be necessary for confirmation. These factors collectively contributed to the variability in MG132-induced signaling observed in this study. Therefore, future studies should investigate these differences in detail to gain a comprehensive understanding of the diverse mechanisms underlying Ut-LMS progression and therapeutic responses.

In addition to MG132, research on proteasome inhibitors has markedly expanded, thereby contributing to advancements in cancer treatment by targeting the ubiquitin-proteasome system (19,20). Proteasome inhibitors, such as bortezomib, carfilzomib, ixazomib, and marizomib, have demonstrated efficacy in inducing apoptosis and disrupting critical cellular processes in various malignancies (20,35). Bortezomib is the first FDA-approved proteasome inhibitor and is notably effective in treating multiple myeloma (MM) by inhibiting the NF-κB pathway and stabilizing pro-apoptotic factors, thereby improving survival rates (36). Despite the associated cardiovascular risks, carfilzomib, which is a second-generation inhibitor, provides sustained proteasome inhibition and has shown enhanced efficacy in combination therapies for relapsed/refractory MM (37,38). Ixazomib is the first oral proteasome inhibitor that is convenient for outpatient treatment and is effective in extending progression-free survival in patients with MM (39). Furthermore, marizomib has emerged as a promising candidate because of its ability to cross the blood-brain barrier, thus showing potential for treating glioblastoma by inducing oxidative stress and apoptosis in cancer cells (40). Research on the mechanisms of action of these proteasome inhibitors is ongoing to optimize treatment protocols and expand their use to other cancer types, including Ut-LMS. These efforts highlight the critical role of proteasome inhibition in contemporary cancer therapy, with ongoing studies focusing on improving efficacy, reducing toxicity, and enhancing patient outcomes.

This study had some limitations. Although the cell death mechanisms and therapeutic potential of MG132 were investigated, the full extent of its effects across different Ut-LMS cell lines is not yet clear. In addition, the long-term toxicity of MG132 and its potential synergistic effects in combination with other treatments have not yet been explored. Therefore, further research is needed to address these aspects and enhance current understanding of the mechanism by which MG132 can be effectively and safely incorporated into treatment strategies for Ut-LMS.

In conclusion, this study demonstrates the anticancer potential of MG132 in Ut-LMS cell lines, thereby revealing diverse and cell-specific responses. The variability in the effects of MG132 on these cell lines underscores the molecular heterogeneity of this aggressive malignancy and emphasizes the importance of personalized therapeutic approaches. Therefore, the study findings provide a strong foundation for further research to elucidate the precise mechanisms underlying MG132-induced cell death and to explore its synergistic potential in combination therapies.

Supplementary Material

Supporting Data

Acknowledgements

Not applicable.

Funding

This study was supported by the Basic Science Research Program through the National Research Foundation of Korea, funded by the Ministry of Education (grant no. NRF-2022R1l1A1A01053069) and the Korean Government (Ministry of Science and Information and Communication Technology) (grant no. RS-2022-00166501).

Availability of data and materials

The data generated in the present study may be requested from the corresponding author.

Authors' contributions

HL and SS designed the experiments and revised the manuscript accordingly. HL and HJ conducted the experiments and wrote the manuscript. HJ and SS performed the data analysis. All authors have read and approved the final manuscript. HL and HJ confirm the authenticity of all the raw data.

Ethics approval and consent to participate

Not applicable.

Patient consent for publication

Not applicable.

Competing interests

The authors declare that they have no competing interests.

Glossary

Abbreviations

Abbreviations:

Ut-LMS

uterine leiomyosarcoma

LDH

lactate dehydrogenase

ROS

reactive oxygen species

NAC

N-acetylcysteine

CAT

catalase

PARP

poly-adenosine diphosphate ribose polymerase

NF-κB

nuclear factor κB

FDA

Food and Drug Administration

DMSO

dimethyl sulfoxide

MTT

2,5-diphenyl-2H-tetrazolium bromide

LC3

light chain 3

LMS

leiomyosarcoma

MM

multiple myeloma

References

1 

Byar KL and Fredericks T: Uterine leiomyosarcoma. J Adv Pract Oncol. 13:70–76. 2022. View Article : Google Scholar : PubMed/NCBI

2 

Ahuja A, Agarwal P, Sardana R and Bhaskar S: Extensively metastasizing leiomyosarcoma: A diagnostic challenge. J Midlife Health. 8:148–150. 2017.PubMed/NCBI

3 

Chapel DB, Sharma A, Lastra RR, Maccio L, Bragantini E, Zannoni GF, George S, Quade BJ, Parra-Herran C and Nucci MR: A novel morphology-based risk stratification model for stage I uterine leiomyosarcoma: An analysis of 203 cases. Mod Pathol. 35:794–807. 2022. View Article : Google Scholar : PubMed/NCBI

4 

Chapel DB, Nucci MR, Quade BJ and Parra-Herran C: Epithelioid leiomyosarcoma of the uterus: Modern outcome-based appraisal of diagnostic criteria in a large institutional series. Am J Surg Pathol. 46:464–475. 2022. View Article : Google Scholar : PubMed/NCBI

5 

Menon G, Mangla A and Yadav U: Leiomyosarcoma. StatPearls [Internet]. StatPearls Publishing; Treasure Island, FL: 2024

6 

Soares Queirós C, Filipe P and Soares de Almeida L: Cutaneous leiomyosarcoma: A 20-year retrospective study and review of the literature. An Bras Dermatol. 96:278–283. 2021. View Article : Google Scholar : PubMed/NCBI

7 

Bathan AJ, Constantinidou A, Pollack SM and Jones RL: Diagnosis, prognosis, and management of leiomyosarcoma: Recognition of anatomic variants. Curr Opin Oncol. 25:384–389. 2013. View Article : Google Scholar : PubMed/NCBI

8 

Giannini A, Golia D'Augè T, Bogani G, Laganà AS, Chiantera V, Vizza E, Muzii L and Di Donato V: Uterine sarcomas: A critical review of the literature. Eur J Obstet Gynecol Reprod Biol. 287:166–170. 2023. View Article : Google Scholar : PubMed/NCBI

9 

Giannini A, Cuccu I, D'Auge TG, De Angelis E, Laganà AS, Chiantera V, Caserta D, Vitale SG, Muzii L, D'Oria O, et al: The great debate: Surgical outcomes of laparoscopic versus laparotomic myomectomy. A meta-analysis to critically evaluate current evidence and look over the horizon. Eur J Obstet Gynecol Reprod Biol. 297:50–58. 2024. View Article : Google Scholar : PubMed/NCBI

10 

Kyriazoglou A, Liontos M, Ntanasis-Stathopoulos I and Gavriatopoulou M: The systemic treatment of uterine leiomyosarcomas: A systematic review. No news is good news? Medicine (Baltimore). 100:e253092021.PubMed/NCBI

11 

Bhat SA, Vasi Z, Adhikari R, Gudur A, Ali A, Jiang L, Ferguson R, Liang D and Kuchay S: Ubiquitin proteasome system in immune regulation and therapeutics. Curr Opin Pharmacol. 67:1023102022. View Article : Google Scholar : PubMed/NCBI

12 

Nunes AT and Annunziata CM: Proteasome inhibitors: Structure and function. Semin Oncol. 44:377–380. 2017. View Article : Google Scholar : PubMed/NCBI

13 

Ji MM, Lee JM, Mon H, Xu J, Tatsuke T and Kusakabe T: Proteasome inhibitor MG132 impairs autophagic flux through compromising formation of autophagosomes in Bombyx cells. Biochem Biophys Res Commun. 479:690–696. 2016. View Article : Google Scholar : PubMed/NCBI

14 

Momose I and Kawada M: The therapeutic potential of microbial proteasome inhibitors. Int Immunopharmacol. 37:23–30. 2016. View Article : Google Scholar : PubMed/NCBI

15 

Guo N and Peng Z: MG132, a proteasome inhibitor, induces apoptosis in tumor cells. Asia Pac J Clin Oncol. 9:6–11. 2013. View Article : Google Scholar : PubMed/NCBI

16 

Bulangalire N, Claeyssen C, Agbulut O and Cieniewski-Bernard C: Impact of MG132 induced-proteotoxic stress on αB-crystallin and desmin phosphorylation and O-GlcNAcylation and their partition towards cytoskeleton. Biochimie. 226:121–135. 2024. View Article : Google Scholar : PubMed/NCBI

17 

La Frazia S, Amici C and Santoro MG: Antiviral activity of proteasome inhibitors in herpes simplex virus-1 infection: Role of nuclear factor-kappaB. Antivir Ther. 11:995–1004. 2006. View Article : Google Scholar : PubMed/NCBI

18 

Ortiz-Lazareno PC, Hernandez-Flores G, Dominguez-Rodriguez JR, Lerma-Diaz JM, Jave-Suarez LF, Aguilar-Lemarroy A, Gomez-Contreras PC, Scott-Algara D and Bravo-Cuellar A: MG132 proteasome inhibitor modulates proinflammatory cytokines production and expression of their receptors in U937 cells: Involvement of nuclear factor-kappaB and activator protein-1. Immunology. 124:534–541. 2008. View Article : Google Scholar : PubMed/NCBI

19 

Kaplan GS, Torcun CC, Grune T, Ozer NK and Karademir B: Proteasome inhibitors in cancer therapy: Treatment regimen and peripheral neuropathy as a side effect. Free Radic Biol Med. 103:1–13. 2017. View Article : Google Scholar : PubMed/NCBI

20 

Manasanch EE and Orlowski RZ: Proteasome inhibitors in cancer therapy. Nat Rev Clin Oncol. 14:417–433. 2017. View Article : Google Scholar : PubMed/NCBI

21 

Goldberg AL: Development of proteasome inhibitors as research tools and cancer drugs. J Cell Biol. 199:583–588. 2012. View Article : Google Scholar : PubMed/NCBI

22 

Maki RG, Kraft AS, Scheu K, Yamada J, Wadler S, Antonescu CR, Wright JJ and Schwartz GK: A multicenter phase II study of bortezomib in recurrent or metastatic sarcomas. Cancer. 103:1431–1438. 2005. View Article : Google Scholar : PubMed/NCBI

23 

Steinhilb ML, Turner RS and Gaut JR: The protease inhibitor, MG132, blocks maturation of the amyloid precursor protein Swedish mutant preventing cleavage by beta-secretase. J Biol Chem. 276:4476–4484. 2001. View Article : Google Scholar : PubMed/NCBI

24 

Sun F, Anantharam V, Zhang D, Latchoumycandane C, Kanthasamy A and Kanthasamy AG: Proteasome inhibitor MG-132 induces dopaminergic degeneration in cell culture and animal models. Neurotoxicology. 27:807–815. 2006. View Article : Google Scholar : PubMed/NCBI

25 

Ma Y, Chen B, Liu D, Yang Y, Xiong Z, Zeng J and Dong Y: MG132 treatment attenuates cardiac remodeling and dysfunction following aortic banding in rats via the NF-κB/TGFβ1 pathway. Biochem Pharmacol. 81:1228–1236. 2011. View Article : Google Scholar : PubMed/NCBI

26 

Zeng W, Qi W, Mu J, Wei Y, Yang LL, Zhang Q, Wu Q, Tang JY and Feng B: MG132 protects against renal dysfunction by regulating Akt-mediated inflammation in diabetic nephropathy. Sci Rep. 9:20492019. View Article : Google Scholar : PubMed/NCBI

27 

Shu Z, Li X, Zhang W, Huyan Z, Cheng D, Xie S, Cheng H, Wang J and Du B: MG-132 activates sodium palmitate-induced autophagy in human vascular smooth muscle cells and inhibits senescence via the PI3K/AKT/mTOR axis. Lipids Health Dis. 23:2822024. View Article : Google Scholar : PubMed/NCBI

28 

Gómez-Virgilio L, Silva-Lucero MD, Flores-Morelos DS, Gallardo-Nieto J, Lopez-Toledo G, Abarca-Fernandez AM, Zacapala-Gómez AE, Luna-Muñoz J, Montiel-Sosa F, Soto-Rojas LO, et al: Autophagy: A key regulator of homeostasis and disease: An overview of molecular mechanisms and modulators. Cells. 11:22622022. View Article : Google Scholar : PubMed/NCBI

29 

Guo C, Sun L, Chen X and Zhang D: Oxidative stress, mitochondrial damage and neurodegenerative diseases. Neural Regen Res. 8:2003–2014. 2013.PubMed/NCBI

30 

Zhitkovich A: N-acetylcysteine: Antioxidant, aldehyde scavenger, and more. Chem Res Toxicol. 32:1318–1319. 2019. View Article : Google Scholar : PubMed/NCBI

31 

Anwar S, Alrumaihi F, Sarwar T, Babiker AY, Khan AA, Prabhu SV and Rahmani AH: Exploring therapeutic potential of catalase: Strategies in disease prevention and management. Biomolecules. 14:6972024. View Article : Google Scholar : PubMed/NCBI

32 

Mills J, Matos T, Charytonowicz E, Hricik T, Castillo-Martin M, Remotti F, Lee FY and Matushansky I: Characterization and comparison of the properties of sarcoma cell lines in vitro and in vivo. Hum Cell. 22:85–93. 2009. View Article : Google Scholar : PubMed/NCBI

33 

Smardová J, Pavlová S, Svitáková M, Grochová D and Ravcuková B: Analysis of p53 status in human cell lines using a functional assay in yeast: Detection of new non-sense p53 mutation in codon 124. Oncol Rep. 14:901–907. 2005.PubMed/NCBI

34 

Coley HM, Shotton CF, Kokkinos MI and Thomas H: The effects of the CDK inhibitor seliciclib alone or in combination with cisplatin in human uterine sarcoma cell lines. Gynecol Oncol. 105:462–469. 2007. View Article : Google Scholar : PubMed/NCBI

35 

Potts BC, Albitar MX, Anderson KC, Baritaki S, Berkers C, Bonavida B, Chandra J, Chauhan D, Cusack JC Jr, Fenical W, et al: Marizomib, a proteasome inhibitor for all seasons: Preclinical profile and a framework for clinical trials. Curr Cancer Drug Targets. 11:254–284. 2011. View Article : Google Scholar : PubMed/NCBI

36 

Alwahsh M, Farhat J, Talhouni S, Hamadneh L and Hergenröder R: Bortezomib advanced mechanisms of action in multiple myeloma, solid and liquid tumors along with its novel therapeutic applications. EXCLI J. 22:146–168. 2023.PubMed/NCBI

37 

Latif A, Kapoor V, Lateef N, Ahsan MJ, Usman RM, Malik SU, Ahmad N, Rosko N, Rudoni J, William P, et al: Incidence and management of carfilzomib-induced cardiovascular toxicity; A systematic review and meta-analysis. Cardiovasc Hematol Disord Drug Targets. 21:30–45. 2021. View Article : Google Scholar : PubMed/NCBI

38 

Zhai Y, Ye X, Hu F, Xu J, Guo X, Cao Y, Lin Z, Zhou X, Guo Z and He J: Cardiovascular toxicity of carfilzomib: The real-world evidence based on the adverse event reporting system database of the FDA, the United States. Front Cardiovasc Med. 8:7354662021. View Article : Google Scholar : PubMed/NCBI

39 

Richardson PG, Zweegman S, O'Donnell EK, Laubach JP, Raje N, Voorhees P, Ferrari RH, Skacel T, Kumar SK and Lonial S: Ixazomib for the treatment of multiple myeloma. Expert Opin Pharmacother. 19:1949–1968. 2018. View Article : Google Scholar : PubMed/NCBI

40 

Di K, Lloyd GK, Abraham V, MacLaren A, Burrows FJ, Desjardins A, Trikha M and Bota DA: Marizomib activity as a single agent in malignant gliomas: Ability to cross the blood-brain barrier. Neuro Oncol. 18:840–848. 2016. View Article : Google Scholar : PubMed/NCBI

Related Articles

Journal Cover

June-2025
Volume 31 Issue 6

Print ISSN: 1791-2997
Online ISSN:1791-3004

Sign up for eToc alerts

Recommend to Library

Copy and paste a formatted citation
x
Spandidos Publications style
Joung H, Seo S and Liu H: MG132 induces cell type‑specific anticancer effects in uterine leiomyosarcoma cell lines. Mol Med Rep 31: 159, 2025.
APA
Joung, H., Seo, S., & Liu, H. (2025). MG132 induces cell type‑specific anticancer effects in uterine leiomyosarcoma cell lines. Molecular Medicine Reports, 31, 159. https://doi.org/10.3892/mmr.2025.13524
MLA
Joung, H., Seo, S., Liu, H."MG132 induces cell type‑specific anticancer effects in uterine leiomyosarcoma cell lines". Molecular Medicine Reports 31.6 (2025): 159.
Chicago
Joung, H., Seo, S., Liu, H."MG132 induces cell type‑specific anticancer effects in uterine leiomyosarcoma cell lines". Molecular Medicine Reports 31, no. 6 (2025): 159. https://doi.org/10.3892/mmr.2025.13524