1
|
Togni A, Piermartiri T, Tasca CI, Nedel CB. The intricate relationship between SUMOylation and gliomas: a review with a perspective on natural compounds. Nat Prod Res 2025:1-12. [PMID: 39849680 DOI: 10.1080/14786419.2025.2456093] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2023] [Revised: 12/31/2024] [Accepted: 01/16/2025] [Indexed: 01/25/2025]
Abstract
Gliomas are tumours that affect the nervous system, with glioblastoma, also known as grade IV astrocytoma, being the most aggressive type, associated with poor prognosis. Glioblastoma is characterised by its highly invasive nature, rapid growth, and resistance to conventional chemotherapy and radiation treatments, resulting in a median survival of about 14 months. To improve patient outcomes, novel therapeutic approaches are needed. Targeting SUMOylation, a post-translational modification involving the attachment of Small Ubiquitin-like Modifier (SUMO) proteins to lysine residues in target proteins, is emerging as a promising strategy. SUMOylation regulates various biological processes, including the cell cycle, apoptosis, and senescence. Dysregulation of this pathway has been linked to glioblastoma tumorigenesis, as well as the invasion and proliferation of glioblastoma cells. Therefore, focusing on the SUMOylation pathway offers the potential for developing innovative therapeutic strategies, including the use of natural compounds as adjuvant therapies, to address glioblastoma more effectively.
Collapse
Affiliation(s)
- Anderson Togni
- Programa de Pós-Graduação em Biologia Celular e do Desenvolvimento, Universidade Federal de Santa Catarina, Florianópolis, Brazil
| | - Tetsade Piermartiri
- Programa de Pós-Graduação em Bioquímica, Universidade Federal de Santa Catarina, Florianópolis, Brazil
| | - Carla Inês Tasca
- Programa de Pós-Graduação em Bioquímica, Universidade Federal de Santa Catarina, Florianópolis, Brazil
| | - Cláudia Beatriz Nedel
- Programa de Pós-Graduação em Biologia Celular e do Desenvolvimento, Universidade Federal de Santa Catarina, Florianópolis, Brazil
| |
Collapse
|
2
|
Wang J, Xie Y, Zeng C, Nie L, Yang Y. Predictive value of abnormal expression of MPHOSPH9 in reintervention after high intensity focused ultrasound treatment of uterine fibroids. J Obstet Gynaecol Res 2025; 51:e16205. [PMID: 39780408 DOI: 10.1111/jog.16205] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2024] [Accepted: 12/24/2024] [Indexed: 01/11/2025]
Abstract
AIMS Aberrantly expressed MPHOSPH9 has been reported to be associated with poor prognosis in many diseases. Previous study indicates that MPHOSPH9 is abnormally expressed in patients with uterine fibroids (UFs). This study focused on the possible prognostic value of MPHOSPH9 in UFs patients after high intensity focused ultrasound (HIFU) treatment. METHODS A total of 455 UFs patients participated in the study, including 95 patients who needed reintervention (Reintervention group) and 360 patients who did not need reintervention (Non-reintervention group) after HIFU treatment. They volunteered blood samples before HIFU treatment. The relative expression of MPHOSPH9 was assessed by qRT-PCR. Cox regression analysis was performed to assess the prognostic value of MPHOSPH9 and risk factors of postoperative reintervention. RESULTS The expression of MPHOSPH9 was markedly elevated in Reintervention, compared to that of the Non-reintervention group (p < 0.001). Five-year follow-up results showed that among these postoperative patients, 95 patients required reintervention and the reintervention time mainly occurred during 10-40 months after surgery. The Cox regression analysis demonstrated that UFs size (hazard ratio = 2.769, 95% confidence interval: 1.366-5.610) and the high MPHOSPH9 expression level (hazard ratio = 3.160, 95% confidence interval: 1.506-6.630) were independent risk factors for postoperative reintervention. CONCLUSIONS The enhanced MPHOSPH9 was a potential candidate for predicting postoperative reintervention in UFs patients.
Collapse
Affiliation(s)
- Jing Wang
- Department of Imaging, Beijing Anzhen Nanchong Hospital, Capital Medical University & Nanchong Central Hospital, Nanchong, China
| | - Yingjie Xie
- One Departments of Cadre Ward, General Hospital of Southern Theater Command of Chinese PLA, Guangzhou, China
| | - Chaoqiang Zeng
- Department of Imaging, Beijing Anzhen Nanchong Hospital, Capital Medical University & Nanchong Central Hospital, Nanchong, China
| | - Lin Nie
- Department of Imaging, Beijing Anzhen Nanchong Hospital, Capital Medical University & Nanchong Central Hospital, Nanchong, China
| | - Yang Yang
- Department of Radiology, Ya'an People's Hospital, Ya'an, China
| |
Collapse
|
3
|
He J, Guo J, Sun P. Prognostic value of CTF1 in glioma and its role in the tumor microenvironment. Transl Cancer Res 2024; 13:6862-6879. [PMID: 39816535 PMCID: PMC11730202 DOI: 10.21037/tcr-24-1258] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2024] [Accepted: 11/10/2024] [Indexed: 01/18/2025]
Abstract
Background Within the realm of primary brain tumors, specifically glioblastoma (GBM), presents a notable obstacle due to their unfavorable prognosis and differing median survival rates contingent upon tumor grade and subtype. Despite a plethora of research connecting cardiotrophin-1 (CTF1) modifications to a range of illnesses, its correlation with glioma remains uncertain. This study investigated the clinical value of CTF1 in glioma and its potential as a biomarker of the disease. Methods Glioma project in The Cancer Genome Atlas (TCGA) database served as the training cohort, and CGGA 325 series in the Chinese Glioma Genome Atlas (CGGA) database served as the external independent validation cohort. First, the difference in the expression level of CTF1 between glioma tissue and normal tissue was analyzed, and the results were verified with the CGGA database. The relationship between CTF1 expression and the prognosis of glioma patients was evaluated using Univariate and Multivariate Cox analysis and the Kaplan-Meier (KM) curve. We used CIBERSOFT to explore the association between CTF1 and immune cell infiltration in GBM, as well as performing gene set enrichment analysis (GSEA) and gene set variation analysis (GSVA) analyses. Furthermore, we analyzed the relationship between CTF1 and gene mutations and drug sensitivity. Using Weighted gene co-expression network analysis (WGCNA) analysis, we pinpointed the gene set most correlated with CTF1 and conducted Gene Ontology (GO) and Kyoto Encyclopedia of Genes and Genomes (KEGG) gene enrichment analyses to anticipate the pathways that could be influenced by CTF1. Finally, we constructed a nomogram using a multifactorial regression model to further predict patient prognosis. Results CTF1 expression was significantly elevated in glioma tissues compared to normal tissues in the TCGA dataset (P<0.001) and was associated with poorer survival in both TCGA and CGGA datasets (P<0.001). Receiver operating characteristic (ROC) analysis demonstrated the diagnostic potential of CTF1, with an area under the curve (AUC) of 0.889 [95% confidence interval (CI): 0.803-0.974] in TCGA and 0.664 (95% CI: 0.599-0.729) in CGGA. High CTF1 levels were correlated with advanced glioma grades, and Cox regression analysis identified CTF1 as an independent risk factor. A nomogram incorporating CTF1 levels, isocitrate dehydrogenase 1 (IDH1) mutation status, O6-methylguanine-DNA methyltransferase (MGMT) methylation status, age, and gender were developed and validated to predict 1- and 2-year survival probabilities. In GBM, drug sensitivity analysis revealed significant associations between CTF1 expression and responsiveness to gemcitabine, dasatinib, and other agents. CTF1 expression was also linked to immune infiltration (monocytes, neutrophils, M0 macrophages) and pathways involved in tumor progression, including IL2_STAT5, P53, and IL6_JAK_STAT3 signaling pathways. Conclusions CTF1 could serve as a prognostic marker for glioma. It acts as a predictive indicator and is associated with immune cell infiltration in GBM. These findings provide a foundation for further research into the molecular function of CTF1 and offer new insights for exploring the underlying mechanisms and developing treatments for glioma.
Collapse
Affiliation(s)
- Jiakai He
- The Second Clinical Medical College, Lanzhou University, Lanzhou, China
- Department of Radiation Oncology, The Second Hospital of Lanzhou University, Lanzhou, China
| | - Jiayin Guo
- The Second Clinical Medical College, Lanzhou University, Lanzhou, China
| | - Pengfei Sun
- Department of Radiation Oncology, The Second Hospital of Lanzhou University, Lanzhou, China
| |
Collapse
|
4
|
Liu X, Zhang S, Dong Y, Xie Y, Li Q. SENP1-mediated SUMOylation of SIRT1 affects glioma development through the NF-κB pathway. Exp Cell Res 2023; 433:113822. [PMID: 37866458 DOI: 10.1016/j.yexcr.2023.113822] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2023] [Revised: 09/15/2023] [Accepted: 10/15/2023] [Indexed: 10/24/2023]
Abstract
Gliomas are the most common primary brain tumors in adults. Although they exist in different malignant stages, most gliomas are clinically challenging because of their infiltrative growth patterns and inherent relapse tendency with increased malignancy. Epigenetic alterations have been suggested to be an important factor for glioma genesis. Using mRNA probe hybridization, we identified SUMO-specific protease 1 (SENP1) as the most significantly upregulated SUMOylation regulator in glioma. Moreover, SENP1 was overexpressed in gliomas and predicted poor prognoses. Depletion of SENP1 reduced glioma cell activity, cycle arrest, and increased apoptotic activity. Mechanistically, SENP1 inhibited the protein expression of sirtuin 1 (SIRT1) through de-SUMOylation, and SIRT1 inhibited the activity of nuclear factor kappaB (NF-κB) by deacetylation. Rescue experiments revealed that downregulation of SIRT1 reversed the inhibitory effect of sh-SENP1 on glioma cell malignant phenotype, while downregulation of NF-κB reversed the activating effect of sh-SIRT1 on glioma cell malignant phenotype. Thus, SENP1-mediated de-SUMOylation of SIRT1 might be therapeutically important in gliomas.
Collapse
Affiliation(s)
- Xin Liu
- Department of Oncology, Affiliated Hospital of Chengde Medical University, Chengde, 067000, Hebei, PR China
| | - Shenglin Zhang
- Department of Oncology, Affiliated Hospital of Chengde Medical University, Chengde, 067000, Hebei, PR China
| | - Yi Dong
- Department of Oncology, Affiliated Hospital of Chengde Medical University, Chengde, 067000, Hebei, PR China
| | - Yunpeng Xie
- Department of Neurosurgery, Affiliated Hospital of Chengde Medical University, Chengde, 067000, Hebei, PR China.
| | - Qingshan Li
- Department of Oncology, Affiliated Hospital of Chengde Medical University, Chengde, 067000, Hebei, PR China.
| |
Collapse
|
5
|
Liu H, Qi L, Tang X, Tan S, Gou Z, Qi J, Lu X, Li D, Chen C. Astragalus Polysaccharides Affect Glioblastoma Cells Through Targeting miR-34a-5p. J BIOMATER TISS ENG 2022. [DOI: 10.1166/jbt.2022.3048] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/23/2022]
Abstract
This study discussed Astragalus Polysaccharides (APS)’s effect on the cytobiology of glioma. U87 glioma cells were assigned into control group (U87 cells), miR-34a-5p mimic group (transfected with miR-34a-5p mimic), and APS group (treated with 10 μM APS) followed by
analysis of miR-34a-5p level, cell proliferation and invasion, Caspase3 and SOD activity as well as E-cadherin, Vimentin and survivn expression. APS treatment significantly upregulated miR-34a-5p expression, inhibits cell proliferation and invasion, and promoted cell apoptosis. In addition,
APS also significantly upregulated E-cadherin, downregulated Vimentin and survivn level in glioma cells as well as inhibited ROS generation and increased SOD activity. In conclusion, the level of miR-34a-5a in glioma cells is up-regulated by APS so as to restrain the biological behaviors of
glioma cells, indicating that it might be used as novel agent for the treatment of glioma.
Collapse
Affiliation(s)
- Hongjun Liu
- Department of Neurosurgery, Affiliated Hospital of North Sichuan Medical College, Nanchong, Sichuan, 637000, China
| | - Lingjun Qi
- Department of Neurosurgery, Daping Hospital, Army Medical University, Chongqing, 400042, China
| | - Xiaoping Tang
- Department of Neurosurgery, Affiliated Hospital of North Sichuan Medical College, Nanchong, Sichuan, 637000, China
| | - Shasha Tan
- Department of Neurosurgery, Affiliated Hospital of North Sichuan Medical College, Nanchong, Sichuan, 637000, China
| | - Zhangyang Gou
- Department of Neurosurgery, Affiliated Hospital of North Sichuan Medical College, Nanchong, Sichuan, 637000, China
| | - Jian Qi
- Department of Neurosurgery, Affiliated Hospital of North Sichuan Medical College, Nanchong, Sichuan, 637000, China
| | - Xingyu Lu
- Department of Neurosurgery, Affiliated Hospital of North Sichuan Medical College, Nanchong, Sichuan, 637000, China
| | - Dong Li
- Department of Neurosurgery, Affiliated Hospital of North Sichuan Medical College, Nanchong, Sichuan, 637000, China
| | - Chunbao Chen
- Department of Neurosurgery, Affiliated Hospital of North Sichuan Medical College, Nanchong, Sichuan, 637000, China
| |
Collapse
|
6
|
Liu B, Liu Z, Wang Y, Lian X, Han Z, Cheng X, Zhu Y, Liu R, Zhao Y, Gao Y. Overexpression of GINS4 is associated with poor prognosis and survival in glioma patients. Mol Med 2021; 27:117. [PMID: 34556022 PMCID: PMC8461916 DOI: 10.1186/s10020-021-00378-0] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2021] [Accepted: 09/11/2021] [Indexed: 12/21/2022] Open
Abstract
Background GINS4, an indispensable component of the GINS complex, is vital for a variety of cancer. However, no known empirical research has focused on exploring relationships between GINS4 and glioma. Thus, this study aims to understand and explain the role of GINS4 in glioma. Method First, we used the data in the CGGA, TCGA, GEO, GEPIA, and HPA databases to explore the expression level of GINS4 in glioma, the correlation between GINS4 expression and the clinical features of glioma, its impact on the survival of glioma patients, and verified the analysis results through RT-qPCR, IHC, and meta-analysis. Subsequently, GSEA enrichment analysis is used to find the potential molecular mechanism of GINS4 to promote the malignant process of glioma and the anti-glioma drugs that may target GINS4 screened by CMap analysis. Moreover, we further explored the influence of the GINS4 expression on the immune microenvironment of glioma patients through the TIMER database. Results Our results suggested that GINS4 was elevated in glioma, and the overexpression of GINS4 was connected with a vast number of clinical features. The next, GINS4 as an independent prognostic factor, which can result in an unfavorable prognosis of glioma. Once more, GINS4 may be participating in the oncogenesis of glioma through JAK-STAT signaling pathways, etc. 6-thioguanine, Doxazosin, and Emetine had potential value in the clinical application of drugs targeting GINS4. Finally, the expression exhibited a close relationship with some immune cells, especially Dendritic cells. Conclusion GINS4 is an independent prognostic factor that led to a poor prognosis of glioma. The present study revealed the probable underlying molecular mechanisms of GINS4 in glioma and provided a potential target for improving the prognosis of glioma. Supplementary Information The online version contains supplementary material available at 10.1186/s10020-021-00378-0.
Collapse
Affiliation(s)
- Binfeng Liu
- Zhengzhou University People's Hospital, Henan Provincial People's Hospital, Henan, 450003, Zhengzhou, China
| | - Zhendong Liu
- Department of Surgery of Spine and Spinal Cord, Henan Provincial People's Hospital, Henan International Joint Laboratory of Intelligentized Orthopedics Innovation and Transformation, Henan Key Laboratory for Intelligent Precision Orthopedics, People's Hospital of Zhengzhou University, People's Hospital of Henan University, Henan, 450003, Zhengzhou, China
| | - Yanbiao Wang
- Department of Orthopedics, First Affiliated Hospital of Xinxiang Medical College, Xinjiang, China
| | - Xiaoyu Lian
- Zhengzhou University People's Hospital, Henan Provincial People's Hospital, Henan, 450003, Zhengzhou, China
| | - Zhibin Han
- Department of Neurosurgery of the First Affiliate Hospital of Harbin Medical University, Harbin, China
| | - Xingbo Cheng
- Department of Neurosurgery of the First Affiliate Hospital of Harbin Medical University, Harbin, China
| | - Yongjie Zhu
- Department of Surgery of Spine and Spinal Cord, Henan University People's Hospital, Henan Provincial People's Hospital, Henan, 450003, Zhengzhou, China
| | - Runze Liu
- Department of Surgery of Spine and Spinal Cord, Henan University People's Hospital, Henan Provincial People's Hospital, Henan, 450003, Zhengzhou, China
| | - Yaoye Zhao
- Zhengzhou University People's Hospital, Henan Provincial People's Hospital, Henan, 450003, Zhengzhou, China
| | - Yanzheng Gao
- Department of Surgery of Spine and Spinal Cord, Henan Provincial People's Hospital, Henan International Joint Laboratory of Intelligentized Orthopedics Innovation and Transformation, Henan Key Laboratory for Intelligent Precision Orthopedics, People's Hospital of Zhengzhou University, People's Hospital of Henan University, Henan, 450003, Zhengzhou, China.
| |
Collapse
|
7
|
Establishment of an Immune-Related Gene Signature for Risk Stratification for Patients with Glioma. COMPUTATIONAL AND MATHEMATICAL METHODS IN MEDICINE 2021; 2021:2191709. [PMID: 34497663 PMCID: PMC8420975 DOI: 10.1155/2021/2191709] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/30/2021] [Revised: 07/14/2021] [Accepted: 08/11/2021] [Indexed: 12/14/2022]
Abstract
Glioma is a frequently seen primary malignant intracranial tumor, characterized by poor prognosis. The study is aimed at constructing a prognostic model for risk stratification in patients suffering from glioma. Weighted gene coexpression network analysis (WGCNA), integrated transcriptome analysis, and combining immune-related genes (IRGs) were used to identify core differentially expressed IRGs (DE IRGs). Subsequently, univariate and multivariate Cox regression analyses were utilized to establish an immune-related risk score (IRRS) model for risk stratification for glioma patients. Furthermore, a nomogram was developed for predicting glioma patients' overall survival (OS). The turquoise module (cor = 0.67; P < 0.001) and its genes (n = 1092) were significantly pertinent to glioma progression. Ultimately, multivariate Cox regression analysis constructed an IRRS model based on VEGFA, SOCS3, SPP1, and TGFB2 core DE IRGs, with a C-index of 0.811 (95% CI: 0.786-0.836). Then, Kaplan-Meier (KM) survival curves revealed that patients presenting high risk had a dismal outcome (P < 0.0001). Also, this IRRS model was found to be an independent prognostic indicator of gliomas' survival prediction, with HR of 1.89 (95% CI: 1.252-2.85) and 2.17 (95% CI: 1.493-3.14) in the Cancer Genome Atlas (TCGA) and Chinese Glioma Genome Atlas (CGGA) datasets, respectively. We established the IRRS prognostic model, capable of effectively stratifying glioma population, convenient for decision-making in clinical practice.
Collapse
|
8
|
Li X, Meng Y. Construction of a SUMOylation regulator-based prognostic model in low-grade glioma. J Cell Mol Med 2021; 25:5434-5442. [PMID: 33951297 PMCID: PMC8184686 DOI: 10.1111/jcmm.16553] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/28/2020] [Revised: 03/30/2021] [Accepted: 04/01/2021] [Indexed: 12/23/2022] Open
Abstract
Low‐grade glioma (LGG) is an intracranial malignant tumour that mainly originates from astrocytes and oligodendrocytes. SUMOylation is one of the post‐translational modifications but studies of SUMOylation in LGG is quite limited. Transcriptome data, single nucleotide variant (SNV) data and clinical data of LGG were derived from public databases. The differences between the expression of SUMOylation regulators in LGG and normal brain tissue were analysed. Cox regression was used to construct a prognostic model in the training cohort. Kaplan‐Meier survival curves and ROC curves were plotted in the training and the validation cohort to evaluate the effectiveness of the prognostic model. GO and KEGG analyses were applied to preliminarily analyse the biological functions. Compared with normal brain tissue, SENP1 and SENP7 were up‐regulated and SENP5 was down‐regulated in LGG. SUMOylation regulators may be involved in functions such as mRNA splicing, DNA replication, ATPase activity and spliceosome. One prognostic model was established based on the 4 SUMOylation regulator‐related signatures (RFWD3, MPHOSPH9, WRN and NUP155), which had a good predictive ability for overall survival. This study is expected to provide targets for the diagnosis and treatment of low‐grade glioma.
Collapse
Affiliation(s)
- Xiaozhi Li
- Department of Neurosurgery, Shengjing Hospital of China Medical University, Shenyang, China
| | - Yutong Meng
- Department of Stomatology, Shengjing Hospital of China Medical University, Shenyang, China
| |
Collapse
|