1
|
Bal Albayrak MG, Simsek T, Akpinar G, Kasap M, Canturk NZ. Proteomic insights into lymph node metastasis in breast cancer subtypes: Key biomarkers and pathways. Pathol Res Pract 2025; 269:155938. [PMID: 40179440 DOI: 10.1016/j.prp.2025.155938] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/28/2024] [Revised: 02/27/2025] [Accepted: 03/26/2025] [Indexed: 04/05/2025]
Abstract
BACKGROUND Breast cancer (BC) is a significant global cause of death in women, primarily due to its diversity and metastatic potential. METHODS BC, healthy lymph node (HL), and metastatic lymph node (ML) tissues were collected from 19 patients diagnosed with infiltrating ductal carcinoma. Protein isolation was performed, followed by two-dimensional gel electrophoresis (2DE) and mass spectrometry (MALDI-TOF/TOF) to identify differentially expressed proteins. Bioinformatic analyses, including protein-protein interaction networks and molecular pathways, were conducted using STRING. Kaplan-Meier analysis was performed with KM plotter to evaluate the prognostic significance of identified proteins. Receiver operating characteristic (ROC) curves were generated using TCGA and GTEx data from UCSC Xena and easyROC to assess diagnostic relevance. RESULTS Distinct pathways related to cytoskeletal regulation, immune modulation, and oxidative stress response were enriched in each subtype. Key proteins such as TUBA1C, CCT6A, and Vimentin (LNA), CAPZB and ENO1 (LNB), GSTO1 (HER2 OE), and CORO1A and LAP3 (TNBC) were identified as significant in driving metastatic behavior. KM survival analysis showed that CAPZB (LNB) and CORO1A (TNBC) were associated with patient outcomes, while GSTO1 was linked to improved distant metastasis-free survival in HER2 OE. ROC analysis highlighted CAPZB as a strong diagnostic marker. CONCLUSIONS These findings form a basis for comprehending the molecular mechanisms underlying metastasis in different subtypes of breast cancer. They may lead to the identification of new therapeutic targets for customized interventions against invasion and metastasis. Further validation is required to confirm their clinical utility in larger cohorts.
Collapse
Affiliation(s)
| | - Turgay Simsek
- Department of General Surgery, Medical School, Kocaeli University, Kocaeli 41001, Turkiye
| | - Gurler Akpinar
- Department of Medical Biology, Medical School, Kocaeli University, Kocaeli 41001, Turkiye.
| | - Murat Kasap
- Department of Medical Biology, Medical School, Kocaeli University, Kocaeli 41001, Turkiye
| | - Nuh Zafer Canturk
- Department of General Surgery, Medical School, Kocaeli University, Kocaeli 41001, Turkiye
| |
Collapse
|
2
|
Yu P, Zhao Z, Sun Q, Diao B, Sun C, Wang Y, Qiao H, Li H, Yang P. N-glycosylation of GSTO1 promotes cervical cancer migration and invasion through JAK/STAT3 pathway activation. Funct Integr Genomics 2025; 25:51. [PMID: 40032681 DOI: 10.1007/s10142-025-01565-6] [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: 09/03/2024] [Revised: 02/13/2025] [Accepted: 02/23/2025] [Indexed: 03/05/2025]
Abstract
Protein glycosylation is strongly associated with tumor progression. Glutathione S-transferase omega 1 (GSTO1) is a member of the glutathione S-transferase family. The significance of GSTO1 N-glycosylation in the progression of cervical cancer (CC) has remained elusive. In this study, we investigated the functional significance of GSTO1 N-glycosylation in CC progression. We employed immunohistochemistry to detect the relative expression of evaluating the link between GSTO1 in CC and benign tissues and the overall survival (OS) and progression-free survival (PFS) in CC patients.In vitro and in vivo experiments to detect CC cell proliferation or metastatic ability after GSTO1 downregulation. NetNGly1.0 Server database predicts potential N-glycosylation modification sites of GSTO1 (Asn55, Asn135, Asn190). Investigating GSTO1 N-glycosylation's function in cellular migration, invasion and epithelial-mesenchymal transition (EMT), we mutated the N-glycosylation sites of GSTO1 through lentivirus-based insertional mutagenesis. Detection of signalling pathways associated with N-glycosylation-modified GSTO1 by enrichment analysis and Western blot. Compared to normal cervical tissue, CC tissue showed significantly higher GSTO1 expression. Further, high GSTO1 levels were a poor predictor of OS and PFS. Both cell and animal experiments suggested that down-regulation of GSTO1 inhibited cell proliferation and metastasis. Glycosylation modification of targeted mutant GSTO1 at positions 55, 135 and 190 significantly inhibits migration and invasion of CC cells. GSTO1 N-glycosylation fixed point mutation inhibits EMT process in CC cells. Mechanistically, N-glycosylated GSTO1 promoted the expression of JAK/STAT3 pathway related markers. GSTO1 N-glycosylation is associated with CC progression and may promote EMT via JAK/STAT3 signaling.
Collapse
Affiliation(s)
- Panpan Yu
- First Affiliated Hospital, School of Medicine, Shihezi University, Shihezi, China
- School of Medicine, Shihezi University, Shihezi, China
| | - Zouyu Zhao
- First Affiliated Hospital, School of Medicine, Shihezi University, Shihezi, China
| | - Qianyu Sun
- First Affiliated Hospital, School of Medicine, Shihezi University, Shihezi, China
| | - Bowen Diao
- First Affiliated Hospital, School of Medicine, Shihezi University, Shihezi, China
| | - Chongfeng Sun
- First Affiliated Hospital, School of Medicine, Shihezi University, Shihezi, China
| | - Yan Wang
- First Affiliated Hospital, School of Medicine, Shihezi University, Shihezi, China
| | - Hui Qiao
- First Affiliated Hospital, School of Medicine, Shihezi University, Shihezi, China
| | - Hong Li
- First Affiliated Hospital, School of Medicine, Shihezi University, Shihezi, China
| | - Ping Yang
- First Affiliated Hospital, School of Medicine, Shihezi University, Shihezi, China.
| |
Collapse
|
3
|
Dong J, Mao J, Wu W, Qian X, Yu Z. FTO Suppresses Proliferation and Induces Apoptosis of T98G Glioblastoma Cells via N6-methyladenosine Modification of GSTO1. Neurochem Res 2025; 50:83. [PMID: 39843621 DOI: 10.1007/s11064-025-04334-w] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2024] [Revised: 12/27/2024] [Accepted: 01/06/2025] [Indexed: 01/24/2025]
Abstract
Glioblastoma (GBM) is the most malignant type of glioma with a very poor prognosis. N6-methyladenosine (m6A) is well-documented to be involved in GBM progression, and FTO is a demethylase. GSTO1 is also associated with tumor progression. This study aimed to investigate the impact of FTO and GSTO1 on GBM progression and the regulation of FTO on m6A modification of GSTO1. T98G cell phenotypes including proliferation and apoptosis were analyzed by cell counting kit 8, colony formation assay, and flow cytometry. The regulation of m6A methylation mediated by FTO was evaluated by methylated RNA immunoprecipitation, RNA immunoprecipitation, and dual-luciferase reporter assay. The results showed that FTO expression was downregulated in GBM. Overexpression of FTO inhibited cell proliferation and facilitated apoptosis in vitro. Additionally, GSTO1 expression was elevated in GBM, and knockdown of GSTO1 suppressed cell proliferation and promoted apoptosis and oxidative stress. Moreover, FTO inhibited m6A methylation of GSTO1 and reduced the stability of GSTO1. Overexpression of GSTO1 abrogated T98G cellular processes mediated by FTO. The in vivo experiments showed that FTO inhibited tumor growth by downregulating GSTO1 expression. In conclusion, FTO decelerates GBM progression by inducing apoptosis through suppressing m6A methylation of GSTO1.
Collapse
Affiliation(s)
- Jinjiang Dong
- Neurosurgery Department, Chun'an First People's Hospital, Hangzhou Medical College Affiliated Chun'an Hospital, No. 1869, Huanhu North Road, Chunan, Hangzhou, Zhejiang, 311700, China
| | - Jianhao Mao
- Neurosurgery Department, Chun'an First People's Hospital, Hangzhou Medical College Affiliated Chun'an Hospital, No. 1869, Huanhu North Road, Chunan, Hangzhou, Zhejiang, 311700, China
| | - Weihua Wu
- Dept Intens Care Unit, Hangzhou TCM Hospital Affiliated to Zhejiang Chinese Medical University, No.453, Stadium Road, Hangzhou, Zhejiang, 310007, China
| | - Xiaoling Qian
- Dept Intens Care Unit, Hangzhou TCM Hospital Affiliated to Zhejiang Chinese Medical University, No.453, Stadium Road, Hangzhou, Zhejiang, 310007, China
| | - Zhenfei Yu
- Dept Intens Care Unit, Hangzhou TCM Hospital Affiliated to Zhejiang Chinese Medical University, No.453, Stadium Road, Hangzhou, Zhejiang, 310007, China.
| |
Collapse
|
4
|
Zhu J, Zhang T, Jiang J, Yang M, Xia N, Chen Y. Genetic variation perspective reveals potential drug targets for subtypes of endometrial cancer. Sci Rep 2024; 14:28180. [PMID: 39548148 PMCID: PMC11568156 DOI: 10.1038/s41598-024-78689-5] [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: 06/15/2024] [Accepted: 11/04/2024] [Indexed: 11/17/2024] Open
Abstract
The study aims to identify potential drug targets for endometrial cancer (EC) subtypes through a Mendelian randomization (MR) approach, assessing their clinical relevance. We utilized genetic instruments for 4,907 plasma proteins from the deCODE Genetics study dataset, and data with EC (n = 12,906) from a genome-wide study (GWAS) meta-analysis in European populations for MR analyses. Complementary analyses included protein-protein interactions (PPI) network analysis, therapeutic efficacy evaluation, differential gene expression assessment, and prognosis evaluation. The expression levels of key drug targets were quantitatively measured at both the transcriptional and translational stages utilizing reverse transcription quantitative PCR (RT-qPCR) and immunohistochemistry (IHC). Additionally, we analyzed various clinicopathological features. Five drug targets for EC (CBR3, GSTO1, HHIP, IGF2R, and MMP10), seven for endometrioid subtypes (ACAP2, CBR3, GSTO1, HHIP, IGF2R, MMP10, and TLR2), and seven for non-endometrioid subtypes (CST3, DNAJB14, FSTL5, GMPR2, IFI16, MAPK9, and NEO1) were identified. Among these, IGF2R (OR = 1.165; 95% CI 1.067-1.272; p = 1.046 × 10- 2) and CST3 (OR = 0.523; 95% CI 0.339-0.804; p = 7.010 × 10- 3) were highlighted as key drug targets with causal evidence both at transcriptional and translational levels. This study preliminarily confirms that IGF2R and CST3 may serve as novel targets for the treatment of EC, providing a foundational reference for innovative clinical approaches to this disease.
Collapse
Affiliation(s)
- Jiamei Zhu
- Department of Obstetrics and Gynecology, The First Affiliated Hospital of Soochow University, Suzhou, China
- Department of Obstetrics and Gynecology, Jingjiang People's Hospital Affiliated to Yangzhou University, Taizhou, China
- Advanced Molecular Pathology Institute of Soochow University and SANO, Suzhou, China
| | - Ting Zhang
- Department of Obstetrics and Gynecology, The First Affiliated Hospital of Soochow University, Suzhou, China
| | - Juan Jiang
- Department of Obstetrics and Gynecology, Jingjiang People's Hospital Affiliated to Yangzhou University, Taizhou, China
| | - Mei Yang
- Advanced Molecular Pathology Institute of Soochow University and SANO, Suzhou, China
| | - Nan Xia
- Department of Obstetrics and Gynecology, Jingjiang People's Hospital Affiliated to Yangzhou University, Taizhou, China
| | - Youguo Chen
- Department of Obstetrics and Gynecology, The First Affiliated Hospital of Soochow University, Suzhou, China.
| |
Collapse
|
5
|
Pohjolainen L, Ruskoaho H, Talman V. Transcriptomics reveal stretched human pluripotent stem cell-derived cardiomyocytes as an advantageous hypertrophy model. JOURNAL OF MOLECULAR AND CELLULAR CARDIOLOGY PLUS 2022; 2:100020. [PMID: 39802492 PMCID: PMC11708431 DOI: 10.1016/j.jmccpl.2022.100020] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 08/04/2022] [Revised: 11/10/2022] [Accepted: 11/10/2022] [Indexed: 01/16/2025]
Abstract
Left ventricular hypertrophy, characterized by hypertrophy of individual cardiomyocytes, is an adaptive response to an increased cardiac workload that eventually leads to heart failure. Previous studies using neonatal rat ventricular myocytes (NRVMs) and animal models have revealed several genes and signaling pathways associated with hypertrophy and mechanical load. However, these models are not directly applicable to humans. Here, we studied the effect of cyclic mechanical stretch on gene expression of human induced pluripotent stem cell-derived cardiomyocytes (hiPSC-CMs) using RNA sequencing. hiPSC-CMs showed distinct hypertrophic changes in gene expression at the level of individual genes and in biological processes. We also identified several differentially expressed genes that have not been previously associated with cardiomyocyte hypertrophy and thus serve as attractive targets for future studies. When compared to previously published data attained from stretched NRVMs and human embryonic stem cell-derived cardiomyocytes, hiPSC-CMs displayed a smaller number of changes in gene expression, but the differentially expressed genes revealed more pronounced enrichment of hypertrophy-related biological processes and pathways. Overall, these results establish hiPSC-CMs as a valuable in vitro model for studying human cardiomyocyte hypertrophy.
Collapse
Affiliation(s)
- Lotta Pohjolainen
- Drug Research Program and Division of Pharmacology and Pharmacotherapy, Faculty of Pharmacy, University of Helsinki, FI-00014 Helsinki, Finland
| | - Heikki Ruskoaho
- Drug Research Program and Division of Pharmacology and Pharmacotherapy, Faculty of Pharmacy, University of Helsinki, FI-00014 Helsinki, Finland
| | - Virpi Talman
- Drug Research Program and Division of Pharmacology and Pharmacotherapy, Faculty of Pharmacy, University of Helsinki, FI-00014 Helsinki, Finland
| |
Collapse
|
6
|
Wang C, Li Y, Li S, Chen M, Hu Y. Proteomics Combined with RNA Sequencing to Screen Biomarkers of Sepsis. Infect Drug Resist 2022; 15:5575-5587. [PMID: 36172619 PMCID: PMC9512028 DOI: 10.2147/idr.s380137] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2022] [Accepted: 09/10/2022] [Indexed: 12/23/2022] Open
Abstract
Purpose To screen biomarkers in the serum of patients with sepsis by proteomics combined with RNA sequencing technology, and to find new diagnostic and therapeutic targets for sepsis. Patients and Methods Blood samples of 22 sepsis patients (sepsis group) and 10 healthy volunteers (normal group) were collected from January 2019 to December 2020. Data-independent acquisition (DIA) method was employed for protein profiling, RNA sequencing was employed for gene sequencing. Subsequently, quality control and differential analysis (FC≥2; FDR<0.05) of DIA data and RNA sequencing data were performed. Then we identified expression trend-consistent divergence factors by nine-quadrant analysis; subsequent protein-protein interaction (PPI) and gene ontology (GO) functional enrichment analysis of intersection factors was performed, and meta-analysis of targets at transcriptome level was implemented using public datasets. Finally, five Peripheral blood mononuclear cell (PBMC) samples (NC=2; SIRS=1; SEPSIS =2) were collected, and cell localization analysis of core genes was performed by 10× single-cell RNA sequencing (scRNA-seq). Results Compared with the normal group, there were 4681 differentially expressed genes and 202 differentially expressed proteins in the sepsis group. Among them, 25 factors were expressed in both proteome and transcriptome, and the analysis of PPI and GO found that they were mainly involved in biological processes such as white blood cell and neutrophil response, inflammatory and immune response. Four core genes GSTO1, C1QA, RETN, and GRN were screened by meta-analysis, all of which were highly expressed in the sepsis group compared with the normal group (P<0.05); scRNA-seq showed the core genes were mainly localized in macrophage cell lines. Conclusion The core genes GSTO1, C1QA, RETN and GRN are mainly expressed in macrophages, widely involved in inflammation and immune responses, and are highly expressed in plasma in the sepsis, suggesting that they may become potential research targets for sepsis.
Collapse
Affiliation(s)
- Chenglin Wang
- Department of Emergency Medicine, The Affiliated Hospital of Southwest Medical University, Luzhou, People's Republic of China
| | - Yang Li
- Department of Emergency Medicine, The Affiliated Hospital of Southwest Medical University, Luzhou, People's Republic of China
| | - Shilin Li
- Department of Emergency Medicine, The Affiliated Hospital of Southwest Medical University, Luzhou, People's Republic of China
| | - Muhu Chen
- Department of Emergency Medicine, The Affiliated Hospital of Southwest Medical University, Luzhou, People's Republic of China
| | - Yingchun Hu
- Department of Emergency Medicine, The Affiliated Hospital of Southwest Medical University, Luzhou, People's Republic of China
| |
Collapse
|
7
|
Wang S, Ma Y, Li Y, Ge X, Lu C, Cai C, Yang Y, Zhao Y, Liang G, Guo X, Cao G, Li B, Gao P. Long non-coding RNAs in <i>Sus scrofa</i> ileum under starvation stress. Anim Biosci 2022; 35:975-988. [PMID: 35240026 PMCID: PMC9271390 DOI: 10.5713/ab.21.0483] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2021] [Accepted: 02/04/2022] [Indexed: 11/27/2022] Open
Abstract
Objective In this study, we aimed to identify long non-coding RNAs (lncRNAs) that play important roles in starvation stress, analyze their functions, and discover potential molecular targets to alleviate starvation stress to provide a theoretical reference for subsequent in-depth research. Methods We generated a piglet starvation stress animal model. Nine Yorkshire weaned piglets were randomly divided into a long-term starvation stress group (starved for 72 h), short-term starvation stress group (starved for 48 h), and the control group. LncRNA libraries were constructed using high-throughput sequencing of piglet ileums. Results We obtained 11,792 lncRNAs, among which, 2,500 lncRNAs were novel. In total, 509 differentially expressed (DE)lncRNAs were identified in this study. Target genes of DElncRNAs were predicted via cis and trans interactions, and functional and pathway analyses were performed. Gene ontology functions and Kyoto encyclopedia of genes and genomes analysis revealed that lncRNA-targeted genes mainly participated in metabolic pathways, cellular processes, immune system processes, digestive systems, and transport activities. To reveal the mechanism underlying starvation stress, the interaction network between lncRNAs and their targets was constructed based on 26 DElncRNAs and 72 DEmRNAs. We performed an interaction network analysis of 121 DElncRNA–DEmRNA pairs with a Pearson correlation coefficient greater than 0.99. Conclusion We found that MSTRG.19894.13, MSTRG.16726.3, and MSTRG.12176.1 might play important roles in starvation stress. This study not only generated a library of enriched lncRNAs in piglets, but its outcomes also provide a strong foundation to screen key lncRNAs involved in starvation stress and a reference for subsequent in-depth research.
Collapse
|
8
|
Wang K, Zhang FL, Jia W. Glutathione S‑transferase ω 1 promotes the proliferation, migration and invasion, and inhibits the apoptosis of non‑small cell lung cancer cells, via the JAK/STAT3 signaling pathway. Mol Med Rep 2021; 23:71. [PMID: 33236161 PMCID: PMC7716429 DOI: 10.3892/mmr.2020.11709] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2020] [Accepted: 10/21/2020] [Indexed: 12/25/2022] Open
Abstract
Glutathione S‑transferase ω 1 (GSTO1) expression levels have been discovered to be upregulated in various types of cancer. However, to the best of our knowledge, the role of GSTO1 in non‑small cell lung cancer (NSCLC) has not been investigated. The present study aimed to investigate the role of GSTO1 in NSCLC and to determine the potential molecular mechanism. GSTO1 expression levels in A549 cells were knocked down using short hairpin RNA and GSTO1 overexpression in H2122 cells was achieved using cDNA constructs. Reverse transcription‑quantitative PCR was used to analyze the mRNA expression levels of GSTO1. Cell proliferation was determined using a Cell Counting Kit‑8 assay, whereas cell migration and invasion were analyzed using Transwell assays. Flow cytometric analysis was performed to determine the levels of cell apoptosis. The expression levels of GSTO1, Bax, caspase 3, JAK and STAT3 were analyzed using western blotting. The results revealed that GSTO1 overexpression significantly promoted the proliferation, migration and invasion, and inhibited the apoptosis of H2122 cells, whereas the opposite trend was achieved in A549 cells with GSTO1 knockdown. GSTO1 overexpression also significantly increased the phosphorylation levels of JAK and STAT3, whereas the knockdown of GSTO1 promoted the opposite effects. In conclusion, the findings of the present study indicated that GSTO1 may serve as an oncogene in NSCLC. The results suggested that GSTO1 may have an important role in NSCLC by regulating the JAK/STAT3 signaling pathway. Therefore, inhibiting the expression levels of GSTO1 may represent a potential novel therapeutic strategy for NSCLC.
Collapse
Affiliation(s)
- Kai Wang
- Department of Respiratory and Critical Care Medicine, Tianjin Chest Hospital, Tianjin 300222, P.R. China
| | - Fu-Lian Zhang
- Integrated TCM and Western Medicine Department, Tianjin Medical University Chu Hsien-I Memorial Hospital, Tianjin 300134, P.R. China
| | - Wei Jia
- Department of Respiratory and Critical Care Medicine, Tianjin Chest Hospital, Tianjin 300222, P.R. China
| |
Collapse
|