1
|
Sun Z, Zhang Z, Zhang J, Yang Z, Pan S, Li X, Wang X, Zhu X. F-box only protein 25-mediated α-actinin 1 upregulation drives ovarian cancer progression via ERK1/2 signaling in tumor cells and macrophage M2 polarization. Int Immunopharmacol 2025; 153:114479. [PMID: 40117808 DOI: 10.1016/j.intimp.2025.114479] [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: 10/13/2024] [Revised: 03/07/2025] [Accepted: 03/10/2025] [Indexed: 03/23/2025]
Abstract
BACKGROUND ACTN1 belongs to the α-actinin family and is considered a tumor-promoting gene in various tumor types; however, the biological function and fundamental molecular mechanisms of ACTN1 in ovarian cancer remain unclear. METHODS The HPA and The Cancer Genome Atlas (TCGA) databases were used to compare the expression of ACTN1 in normal ovarian and OC tissues. The Kaplan-Meier Plotter database was used to analyze the relationship between the expression of ACTN1 and the prognosis of ovarian cancer. The TIMER2.0 database was used to analyze the correlation between the expression of ACTN1 and macrophages. CCK-8, colony formation, and Transwell assays as well as flow cytometry were used to determine the biological properties of the cells. Protein expression was assessed by immunohistochemistry, immunofluorescence, and western blot analysis. A co-culture experiment was used to analyze the effect of ovarian cancer cells on the polarization of macrophages. Co-immunoprecipitation was performed to validate the interaction between FBXO25 and ACTN1. RESULTS ACTN1 was highly expressed in OC tissues and cell lines. Downregulation of ACTN1 attenuated the proliferation, migration, and invasion of OC cells, promoted apoptosis and reduced the aggregation of M2 macrophages and the expression of CD163. The opposite effect was observed following the upregulation of ACTN1. Mechanistically, ACTN1 knockdown reduced ERK1/2 phosphorylation and inhibited epithelial-mesenchymal transition (EMT), whereas its overexpression resulted in the opposite effect. The ERK1/2 inhibitor LY3214996 partially reversed cell proliferation, migration, and M2 polarization of macrophages promoted by ACTN1 overexpression. Moreover, FBXO25, which is upstream of ACTN1 and interacts with it. FBXO25 upregulation partially reversed cell proliferation and migration inhibited by ACTN1 knockdown. CONCLUSION Upregulation of ACTN1 by FBXO25 promotes the progression of ovarian cancer by activating the ERK1/2 signaling pathway and M2 polarization of macrophages. The FBXO25/ACTN1/ERK1/2 axis and M2 macrophages may represent promising targets for developing ovarian cancer treatments.
Collapse
Affiliation(s)
- Zhengwei Sun
- Zhejiang Provincial Clinical Research Center for Gynecological Diseases, Department of Obstetrics and Gynecology, The Second Affiliated Hospital of Wenzhou Medical University, Wenzhou 325027, China
| | - Zihan Zhang
- Zhejiang Provincial Clinical Research Center for Gynecological Diseases, Department of Obstetrics and Gynecology, The Second Affiliated Hospital of Wenzhou Medical University, Wenzhou 325027, China
| | - Jiamin Zhang
- Zhejiang Provincial Clinical Research Center for Gynecological Diseases, Department of Obstetrics and Gynecology, The Second Affiliated Hospital of Wenzhou Medical University, Wenzhou 325027, China
| | - Ziyi Yang
- Zhejiang Provincial Clinical Research Center for Gynecological Diseases, Department of Obstetrics and Gynecology, The Second Affiliated Hospital of Wenzhou Medical University, Wenzhou 325027, China
| | - Shuya Pan
- Zhejiang Provincial Clinical Research Center for Gynecological Diseases, Department of Obstetrics and Gynecology, The Second Affiliated Hospital of Wenzhou Medical University, Wenzhou 325027, China
| | - Xiaosheng Li
- Zhejiang Provincial Clinical Research Center for Gynecological Diseases, Department of Obstetrics and Gynecology, The Second Affiliated Hospital of Wenzhou Medical University, Wenzhou 325027, China
| | - Xujing Wang
- Zhejiang Provincial Clinical Research Center for Gynecological Diseases, Department of Obstetrics and Gynecology, The Second Affiliated Hospital of Wenzhou Medical University, Wenzhou 325027, China
| | - Xueqiong Zhu
- Zhejiang Provincial Clinical Research Center for Gynecological Diseases, Department of Obstetrics and Gynecology, The Second Affiliated Hospital of Wenzhou Medical University, Wenzhou 325027, China.
| |
Collapse
|
2
|
Karaman E, Yay F, Ayan D, Bayram E, Erturk S. The Clinopathological and Prognostic Significance of SPOCK1 in Gynecological Cancers: A Bioinformatics Based Analysis. BIOLOGY 2025; 14:209. [PMID: 40001977 PMCID: PMC11852031 DOI: 10.3390/biology14020209] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/18/2025] [Revised: 02/09/2025] [Accepted: 02/13/2025] [Indexed: 02/27/2025]
Abstract
Background: Sparc/osteonectin, cwcv, and kazal-like domains proteoglycan 1 (SPOCK1) is an oncogene that promotes tumor formation and progression in certain types of cancer and is associated with poor survival rates. However, there is limited information on the importance of SPOCK1 in gynecological cancers in the literature. The aim of this study was to explore the role of SPOCK1 in ovarian serous cystadenocarcinoma (OV), cervical squamous cell carcinoma and endocervical adenocarcinoma (CESC), and uterine corpus endometrial carcinomas (UCEC). Methods: The data used in this study were obtained from the GEPIA2, TCGA, Kaplan-Meier Plotter, GeneMANIA, UALCAN, cBioPortal, and TIMER databases. Overall survival (OS) and relapse-free survival (RFS) rates were evaluated by Kaplan-Meier survival analysis. Spearman's rho and statistical significance values were obtained for the correlation between SPOCK1 expression and tumor infiltration by different immune cells. Results: Lower SPOCK1 gene expression was observed in CESC and UCEC compared to normal tissue (p < 0.05), but the OV did not differ significantly (p > 0.05). In OV, SPOCK1 gene expression was solely linked to age; in CESC, it was linked to age, stage, weight, and histology; and in UCEC, it was linked to age, stage, weight, and menopausal status. Conclusions:SPOCK1 gene expression in UCEC showed weak positive correlations with CD8+ T cells and weak negative correlations with CD4+ T cells. SPOCK1 may be a potential prognostic and therapeutic target for gynecological cancers.
Collapse
Affiliation(s)
- Enes Karaman
- Department of Obstetrics and Gynecology, Faculty of Medicine, Nigde Omer Halisdemir University, 51240 Nigde, Turkey
| | - Fatih Yay
- Medical Biochemistry, Nigde Omer Halisdemir University Research and Training Hospital, 51100 Nigde, Turkey
| | - Durmus Ayan
- Department of Medical Biochemistry, Faculty of Medicine, Nigde Omer Halisdemir University, 51240 Nigde, Turkey
| | - Ergul Bayram
- Medical Biochemistry, Nigde Omer Halisdemir University Research and Training Hospital, 51100 Nigde, Turkey
| | - Sefa Erturk
- Department of Biophysics, Faculty of Medicine, Nigde Omer Halisdemir University, 51240 Nigde, Turkey
| |
Collapse
|
3
|
Wesley T, Escalona RM, Kannourakis G, Ahmed N. Plakin Expression in Serous Epithelial Ovarian Cancer Has the Potential to Impede Metastatic Spread and Epithelial-Mesenchymal Transition: A Comparative Expression Analysis of Immunohistochemical and In Silico Datasets. Cancers (Basel) 2024; 16:4087. [PMID: 39682273 DOI: 10.3390/cancers16234087] [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: 10/23/2024] [Revised: 11/27/2024] [Accepted: 12/02/2024] [Indexed: 12/18/2024] Open
Abstract
Epithelial ovarian cancer is aggressive and causes high mortality among women worldwide. Members of the plakin family are essential to maintain cytoskeletal integrity and key cellular processes. In this study we characterised the expression of plakins, particularly plectin (PLEC), periplakin (PPL), envoplakin (EVPL), and EMT-related proteins by immunohistochemistry in n = 48 patients' samples to evaluate a potential correlation of plakin expression with EMT as EOC progresses. These tissue plakin and EMT expression analyses were further evaluated by in vitro cell line expression and correlated with the expression of these molecules using publicly available datasets such as Cancer Genome Atlas (TCGA) and Clinical Proteome Tumour Analysis Consortium (CPTAC) datasets. We demonstrate that the expression of PPL and PLEC plakins is decreased in high-grade compared to low-grade EOCs with mixed EMT marker protein expression. This is supported by the correlation of high PPL and PLEC expression with an epithelial rather than mesenchymal phenotype. Our data suggest a partial loss of plakin expression as EOC tumours progress. This may impact the connections of plakins with membrane-bound receptors, which impede the downstream signalling required for the initiation of EMT as the tumours progress.
Collapse
Affiliation(s)
- Tamsin Wesley
- Fiona Elsey Cancer Research Institute, Ballarat, VIC 3353, Australia
- Health Innovation and Transformation Centre, Mt Helen Campus, Federation University Australia, Ballarat, VIC 3353, Australia
| | - Ruth M Escalona
- Fiona Elsey Cancer Research Institute, Ballarat, VIC 3353, Australia
- Centre for Reproductive Health, Hudson Institute of Medical Research, Clayton, VIC 3168, Australia
| | - George Kannourakis
- Fiona Elsey Cancer Research Institute, Ballarat, VIC 3353, Australia
- Health Innovation and Transformation Centre, Mt Helen Campus, Federation University Australia, Ballarat, VIC 3353, Australia
| | - Nuzhat Ahmed
- Fiona Elsey Cancer Research Institute, Ballarat, VIC 3353, Australia
- Health Innovation and Transformation Centre, Mt Helen Campus, Federation University Australia, Ballarat, VIC 3353, Australia
- Centre for Reproductive Health, Hudson Institute of Medical Research, Clayton, VIC 3168, Australia
- Department of Molecular & Translational Science, Monash University, Clayton, VIC 3168, Australia
- Department of Obstetrics and Gynaecology, University of Melbourne, Parkville, VIC 3050, Australia
- Department of Surgery, St Vincent Hospital, University of Melbourne, Fitzroy, VIC 3065, Australia
| |
Collapse
|
4
|
Ren S, Yang K, Fan Q, Wang Q, Zhu M, Yin S, Gu Y, Xu L. Bioinformatics analysis of key candidate genes and pathways in Chinese patients with keratoconus. Exp Eye Res 2023; 231:109488. [PMID: 37116607 DOI: 10.1016/j.exer.2023.109488] [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: 03/21/2023] [Revised: 04/10/2023] [Accepted: 04/19/2023] [Indexed: 04/30/2023]
Abstract
Keratoconus (KC) is a multifactorial disease in which genetic factors played important roles in its pathogenesis. The purpose of the current study was to identify the key candidate genes and pathways in Chinese patients with KC through bioinformatics analysis. Totally, we identified 71 candidate genes by analyzing the results of whole exome sequencing on 51 Chinese patients with KC, combining with previous reports on differential expression at transcription and protein levels in KC. Gene enrichment analysis with GeneCodis demonstrated that two significantly enriched terms including 21 genes in biological process (BP) were detected, and six significantly enriched terms containing 14 genes in Kyoto Encyclopedia of Genes and Genomes (KEGG) pathway were discovered. The STRING was utilized to construct the protein-protein interaction (PPI) network of identified genes. The result showed that a PPI network consisted of 14 nodes with 14 edges was constructed, and two gene modules were obtained. Eight hub genes (LAMB3, LAMA3, LAMA1, ITGA6, ITGA3, COL6A3, COL6A2, and COL6A1) were identified as key candidate genes for KC by cytoHubba in Cytoscape. Functional enrichment analysis with ClueGO and CluePedia indicated that the ECM-receptor interaction was the key pathway accounted for KC. The findings might provide novel insights on the genetic basis of KC.
Collapse
Affiliation(s)
- Shengwei Ren
- Henan Provincial People's Hospital, Henan Eye Hospital, Henan Eye Institute, People's Hospital of Zhengzhou University, Zhengzhou, 450003, China; Zhengzhou University People's Hospital, Henan Provincial People's Hospital, Henan Eye Hospital, Henan Eye Institution, Zhengzhou, 450003, China
| | - Kaili Yang
- Henan Provincial People's Hospital, Henan Eye Hospital, Henan Eye Institute, People's Hospital of Zhengzhou University, Zhengzhou, 450003, China
| | - Qi Fan
- Henan Provincial People's Hospital, Henan Eye Hospital, Henan Eye Institute, People's Hospital of Zhengzhou University, Zhengzhou, 450003, China
| | - Qing Wang
- Henan University People's Hospital, Henan Provincial People's Hospital, Henan Eye Hospital, Henan Eye Institute, Zhengzhou, 450003, China
| | - Meng Zhu
- Zhengzhou University People's Hospital, Henan Provincial People's Hospital, Henan Eye Hospital, Henan Eye Institution, Zhengzhou, 450003, China
| | - Shanshan Yin
- Zhengzhou University People's Hospital, Henan Provincial People's Hospital, Henan Eye Hospital, Henan Eye Institution, Zhengzhou, 450003, China
| | - Yuwei Gu
- Henan Provincial People's Hospital, Henan Eye Hospital, Henan Eye Institute, People's Hospital of Zhengzhou University, Zhengzhou, 450003, China
| | - Liyan Xu
- Henan Provincial People's Hospital, Henan Eye Hospital, Henan Eye Institute, People's Hospital of Zhengzhou University, Zhengzhou, 450003, China.
| |
Collapse
|
5
|
Uncovering Novel Features of the Pc Locus in Horn Development from Gene-Edited Holstein Cattle by RNA-Sequencing Analysis. Int J Mol Sci 2022; 23:ijms232012060. [PMID: 36292916 PMCID: PMC9603690 DOI: 10.3390/ijms232012060] [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: 09/08/2022] [Revised: 10/01/2022] [Accepted: 10/04/2022] [Indexed: 11/19/2022] Open
Abstract
The Polled Celtic (Pc) mutation locus is a genetically simple single mutation that is the best choice for breeding polled cattle using gene editing. However, the mechanism of the Pc locus for regulating horn development is unclear, so we used gene editing, somatic cell nuclear transfer and embryo transfer to obtain polled Holstein fetal bovine (gestation time 90 days) with a homozygous Pc insertion (gene-edited Holstein fetal bovine, EH) and the wild-type 90 days Holstein fetal bovine (WH) as controls. The hematoxylin-eosin (HE) staining results showed that, compared to the WH, the EH horn buds had no white keratinized projections or vacuolated keratinocytes and no thick nerve bundles under the dermal tissue. Furthermore, DNA sequencing results showed that the Pc locus was homozygously inserted into the fetal bovine genome. A total of 791 differentially expressed genes were identified by transcriptome sequencing analysis. Enrichment analysis and protein interaction analysis results of differentially expressed genes showed that abundant gene changes after Pc insertion were associated with the adhesion molecule regulation, actin expression, cytoskeletal deformation and keratin expression and keratinization. It was also noted that the results contained several genes that had been reported to be associated with the development of horn traits, such as RXFP2 and TWIST1. This study identified these changes for the first time and summarized them. The results suggested that the Pc mutant locus may inhibit neural crest cell EMT generation and keratin expression, leading to failures in neural crest cell migration and keratinization of the horn bud tissue, regulating the production of the polled phenotype.
Collapse
|