1
|
Hu WM, Jiang WJ. A prognostic model for laryngeal squamous cell carcinoma based on the mitochondrial metabolism-related genes. Transl Cancer Res 2025; 14:966-979. [PMID: 40104737 PMCID: PMC11912054 DOI: 10.21037/tcr-24-1436] [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: 08/16/2024] [Accepted: 12/17/2024] [Indexed: 03/20/2025]
Abstract
Background Mitochondrial metabolism-related genes (MMRGs) have emerged as potential therapeutic targets in cancer. This study aimed to construct a prognosis model based on MMRGs for patients with laryngeal squamous cell carcinoma (LSCC). Methods Differentially expressed MMRGs in LSCC were identified from The Cancer Genome Atlas (TCGA) and Molecular Signatures Database (MSigDB). Their functions were characterized by Gene Ontology (GO) and Kyoto Encyclopedia of Genes and Genomes (KEGG). A prognostic model was established using univariate, least absolute shrinkage and selection operator (LASSO), and multivariate Cox regression analyses, and its performance was evaluated using Kaplan-Meier and receiver operating characteristic (ROC) curves. Gene set enrichment analysis (GSEA) was performed to elucidate the biological pathways associated with the hub prognostic MMRGs. Genetic perturbation similarity analysis (GPSA) was used to determine the regulatory network of hub genes. Additionally, the correlation of the hub MMRGs with the immune microenvironment and drug sensitivity was investigated. Results We identified 308 differentially expressed MMRGs, enriched in various metabolic processes and pathways. The prognostic model comprising four hub MMRGs (POLD1, PON2, SMS, and THEM5) accurately predicted patient outcomes, with the high-risk group exhibiting poorer survival. Additionally, high expression of POLD1 and THEM5 while low expression of PON2 and SMS indicated better prognosis for LSCC patients. GSEA revealed pathways correlated with each prognostic MMRG, such as PI3K-AKT-mTOR signaling pathways, while GPSA identified key regulatory genes interacting with four hub MMRGs. Furthermore, differences in the tumor immune microenvironment and somatic mutation profiles were observed between high- and low-risk groups. Finally, the correlation of four hub MMRGs with 30 drug sensitivity was revealed. Conclusions This study highlights the prognostic significance of MMRGs in LSCC and underscores their potential as biomarkers for LSCC therapy.
Collapse
Affiliation(s)
- Wei-Ming Hu
- Head and Neck & Otolaryngology Center, Plastic Surgery Center, Cancer Center, Department of Otolaryngology, Zhejiang Provincial People's Hospital, Hangzhou, China
| | - Wen-Jing Jiang
- Head and Neck & Otolaryngology Center, Plastic Surgery Center, Cancer Center, Department of Otolaryngology, Zhejiang Provincial People's Hospital, Hangzhou, China
| |
Collapse
|
2
|
Kang Z, Zhang C, Huangfu H. Exosomal lncRNA LINC02191 Promotes Laryngeal Squamous cell Carcinoma Progression by Targeting miR-204-5p/RAB22A Axis and Regulating PI3K/Akt/mTOR Pathway. Biochem Genet 2024; 62:2117-2133. [PMID: 37863866 DOI: 10.1007/s10528-023-10541-3] [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: 06/26/2023] [Accepted: 10/01/2023] [Indexed: 10/22/2023]
Abstract
Recent research has explored the potential use of serum-derived biomarkers in cancer screening, and mounting evidence has illustrated the pivotal roles of long noncoding RNAs (lncRNAs) in regulating laryngeal squamous cell carcinoma (LSCC) progression. LINC02191 is a newly identified lncRNA and no studies have investigated its role in malignant tumors. This study aims to explore the functions and mechanisms of lncRNA LINC02191 in LSCC. LINC02191 was knocked down in LSCC cells using shRNAs for loss-of-function experiments. RT-qPCR revealed that LINC02191 was upregulated in LSCC patients' serum exosomes, tissues and cells. Western blotting and RT-qPCR were implemented for detecting molecular protein and RNA levels. Colony formation, CCK-8, wound healing and Transwell assays were employed for examining LSCC cell malignant behaviors in vitro. A tumor-bearing mouse model (n = 4/group) was established for examining LINC02191 role in vivo. The results showed that LINC02191 silencing hindered LSCC cell proliferation, invasiveness, migration as well as EMT in vitro and impeded tumorigenesis in xenograft mouse model. Luciferase reporter assay was utilized for verifying the interaction between LINC02191, miR-204-5p and RAB22A. Pearson correlation analysis was employed to evaluate their expression correlation in LSCC tissue specimens (N = 30). Mechanistically, LINC02191 upregulated RAB22A by binding to miR-204-5p, and knocking down LINC02191 inhibited PI3K/Akt/mTOR signaling transduction in LSCC cells and tumor-bearing mice. Moreover, RAB22A overexpression reversed LINC02191 depletion-triggered suppression of LSCC cell aggressiveness and inactivation of PI3K/Akt/mTOR signaling. In conclusion, LINC02191 aggravates LSCC by targeting miR-204-5p/RAB22A/PI3K/Akt/mTOR signaling pathway, which indicates that LINC02191 may serve as a promising target for LSCC treatment.
Collapse
Affiliation(s)
- Zhiwei Kang
- Department of Otolaryngology Head & Neck Surgery, Shanxi Key Laboratory of Otorhinolaryngology Head and Neck Cancer, Shanxi Clinical Medical Research Center for Precision Medicine of Head and Neck Cancer, First Hospital of Shanxi Medical University, Taiyuan, 030001, China
| | - Chunming Zhang
- Department of Otolaryngology Head & Neck Surgery, Shanxi Key Laboratory of Otorhinolaryngology Head and Neck Cancer, Shanxi Clinical Medical Research Center for Precision Medicine of Head and Neck Cancer, First Hospital of Shanxi Medical University, Taiyuan, 030001, China
| | - Hui Huangfu
- Department of Otolaryngology Head & Neck Surgery, Shanxi Key Laboratory of Otorhinolaryngology Head and Neck Cancer, Shanxi Clinical Medical Research Center for Precision Medicine of Head and Neck Cancer, First Hospital of Shanxi Medical University, Taiyuan, 030001, China.
| |
Collapse
|
3
|
Wang J, Liu L, Li Z, Wang H, Ren Y, Wang K, Liu Y, Tao X, Zheng L. JMJD3 regulate H3K27me3 modification via interacting directly with TET1 to affect spermatogonia self-renewal and proliferation. BMC Genomics 2024; 25:225. [PMID: 38424516 PMCID: PMC10905883 DOI: 10.1186/s12864-024-10120-9] [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: 10/20/2023] [Accepted: 02/13/2024] [Indexed: 03/02/2024] Open
Abstract
BACKGROUND In epigenetic modification, histone modification and DNA methylation coordinate the regulation of spermatogonium. Not only can methylcytosine dioxygenase 1 (TET1) function as a DNA demethylase, converting 5-methylcytosine to 5-hydroxymethylcytosine, it can also form complexes with other proteins to regulate gene expression. H3K27me3, one of the common histone modifications, is involved in the regulation of stem cell maintenance and tumorigenesis by inhibiting gene transcription. METHODS we examined JMJD3 at both mRNA and protein levels and performed Chip-seq sequencing of H3K27me3 in TET1 overexpressing cells to search for target genes and signaling pathways of its action. RESULTS This study has found that JMJD3 plays a leading role in spermatogonia self-renewal and proliferation: at one extreme, the expression of the self-renewal gene GFRA1 and the proliferation-promoting gene PCNA was upregulated following the overexpression of JMJD3 in spermatogonia; at the other end of the spectrum, the expression of differentiation-promoting gene DAZL was down-regulated. Furthermore, the fact that TET1 and JMJD3 can form a protein complex to interact with H3K27me3 has also been fully proven. Then, through analyzing the sequencing results of CHIP-Seq, we found that TET1 targeted Pramel3 when it interacted with H3K27me3. Besides, TET1 overexpression not only reduced H3K27me3 deposition at Pramel3, but promoted its transcriptional activation as well, and the up-regulation of Pramel3 expression was verified in JMJD3-overexpressing spermatogonia. CONCLUSION In summary, our study identified a novel link between TET1 and H3K27me3 and established a Tet1-JMJD3-H3K27me3-Pramel3 axis to regulate spermatogonia self-renewal and proliferation. Judging from the evidence offered above, we can safely conclude that this study provides new ideas for further research regarding the mechanism of spermatogenesis and spermatogenesis disorders on an apparent spectrum.
Collapse
Affiliation(s)
- Jin Wang
- School of Basic Medical Sciences, Anhui Medical University, Hefei, China
| | - Lingling Liu
- School of Basic Medical Sciences, Anhui Medical University, Hefei, China
| | - Zebin Li
- School of Basic Medical Sciences, Anhui Medical University, Hefei, China
| | - Haoyu Wang
- School of Basic Medical Sciences, Anhui Medical University, Hefei, China
| | - Yuanyuan Ren
- School of Basic Medical Sciences, Anhui Medical University, Hefei, China
| | - Kaisheng Wang
- School of Basic Medical Sciences, Anhui Medical University, Hefei, China
| | - Yang Liu
- School of Basic Medical Sciences, Anhui Medical University, Hefei, China
| | - Xinjie Tao
- School of Basic Medical Sciences, Anhui Medical University, Hefei, China
| | - Liming Zheng
- School of Basic Medical Sciences, Anhui Medical University, Hefei, China.
| |
Collapse
|
4
|
Wang Q, Fan X, Sheng Q, Yang M, Zhou P, Lu S, Gao Y, Kong Z, Shen N, Lv Z, Wang R. N6-methyladenosine methylation in kidney injury. Clin Epigenetics 2023; 15:170. [PMID: 37865763 PMCID: PMC10590532 DOI: 10.1186/s13148-023-01586-7] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2023] [Accepted: 10/11/2023] [Indexed: 10/23/2023] Open
Abstract
Multiple mechanisms are involved in kidney damage, among which the role of epigenetic modifications in the occurrence and development of kidney diseases is constantly being revealed. However, N6-methyladenosine (M6A), a well-known post-transcriptional modification, has been regarded as the most prevalent epigenetic modifications in higher eukaryotic, which is involved in various biological processes of cells such as maintaining the stability of mRNA. The role of M6A modification in the mechanism of kidney damage has attracted widespread attention. In this review, we mainly summarize the role of M6A modification in the progression of kidney diseases from the following aspects: the regulatory pattern of N6-methyladenosine, the critical roles of N6-methyladenosine in chronic kidney disease, acute kidney injury and renal cell carcinoma, and then reveal its potential significance in the diagnosis and treatment of various kidney diseases. A better understanding of this field will be helpful for future research and clinical treatment of kidney diseases.
Collapse
Affiliation(s)
- Qimeng Wang
- Department of Nephrology, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, 250021, Shandong, China
- Department of Nephrology, Shandong Provincial Hospital, Shandong University, Jinan, 250021, Shandong, China
| | - Xiaoting Fan
- Department of Nephrology, Shandong Provincial Hospital, Shandong University, Jinan, 250021, Shandong, China
| | - Qinghao Sheng
- Department of Nephrology, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, 250021, Shandong, China
| | - Meilin Yang
- Department of Nephrology, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, 250021, Shandong, China
| | - Ping Zhou
- Department of Nephrology, Shandong Provincial Hospital, Shandong University, Jinan, 250021, Shandong, China
| | - Shangwei Lu
- Department of Nephrology, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, 250021, Shandong, China
| | - Ying Gao
- Department of Nephrology, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, 250021, Shandong, China
| | - Zhijuan Kong
- Department of Nephrology, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, 250021, Shandong, China
| | - Ning Shen
- Department of Nephrology, Shandong Provincial Hospital, Shandong University, Jinan, 250021, Shandong, China
| | - Zhimei Lv
- Department of Nephrology, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, 250021, Shandong, China.
- Department of Nephrology, Shandong Provincial Hospital, Shandong University, Jinan, 250021, Shandong, China.
| | - Rong Wang
- Department of Nephrology, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, 250021, Shandong, China.
- Department of Nephrology, Shandong Provincial Hospital, Shandong University, Jinan, 250021, Shandong, China.
| |
Collapse
|
5
|
Hedrich V, Breitenecker K, Ortmayr G, Pupp F, Huber H, Chen D, Sahoo S, Jolly MK, Mikulits W. PRAME Is a Novel Target of Tumor-Intrinsic Gas6/Axl Activation and Promotes Cancer Cell Invasion in Hepatocellular Carcinoma. Cancers (Basel) 2023; 15:2415. [PMID: 37173882 PMCID: PMC10177160 DOI: 10.3390/cancers15092415] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2023] [Revised: 04/14/2023] [Accepted: 04/19/2023] [Indexed: 05/15/2023] Open
Abstract
(1) Background: Activation of the receptor tyrosine kinase Axl by Gas6 fosters oncogenic effects in hepatocellular carcinoma (HCC), associating with increased mortality of patients. The impact of Gas6/Axl signaling on the induction of individual target genes in HCC and its consequences is an open issue. (2) Methods: RNA-seq analysis of Gas6-stimulated Axl-proficient or Axl-deficient HCC cells was used to identify Gas6/Axl targets. Gain- and loss-of-function studies as well as proteomics were employed to characterize the role of PRAME (preferentially expressed antigen in melanoma). Expression of Axl/PRAME was assessed in publicly available HCC patient datasets and in 133 HCC cases. (3) Results: Exploitation of well-characterized HCC models expressing Axl or devoid of Axl allowed the identification of target genes including PRAME. Intervention with Axl signaling or MAPK/ERK1/2 resulted in reduced PRAME expression. PRAME levels were associated with a mesenchymal-like phenotype augmenting 2D cell migration and 3D cell invasion. Interactions with pro-oncogenic proteins such as CCAR1 suggested further tumor-promoting functions of PRAME in HCC. Moreover, PRAME showed elevated expression in Axl-stratified HCC patients, which correlates with vascular invasion and lowered patient survival. (4) Conclusions: PRAME is a bona fide target of Gas6/Axl/ERK signaling linked to EMT and cancer cell invasion in HCC.
Collapse
Affiliation(s)
- Viola Hedrich
- Center for Cancer Research, Comprehensive Cancer Center, Medical University of Vienna, 1090 Vienna, Austria; (V.H.)
| | - Kristina Breitenecker
- Center for Cancer Research, Comprehensive Cancer Center, Medical University of Vienna, 1090 Vienna, Austria; (V.H.)
| | - Gregor Ortmayr
- Center for Cancer Research, Comprehensive Cancer Center, Medical University of Vienna, 1090 Vienna, Austria; (V.H.)
| | - Franziska Pupp
- Center for Cancer Research, Comprehensive Cancer Center, Medical University of Vienna, 1090 Vienna, Austria; (V.H.)
| | - Heidemarie Huber
- Center for Cancer Research, Comprehensive Cancer Center, Medical University of Vienna, 1090 Vienna, Austria; (V.H.)
| | - Doris Chen
- Department of Chromosome Biology, Max Perutz Labs Vienna, University of Vienna, 1030 Vienna, Austria
| | - Sarthak Sahoo
- Centre for BioSystems Science and Engineering, Indian Institute of Science, Bangalore 560012, India
| | - Mohit Kumar Jolly
- Centre for BioSystems Science and Engineering, Indian Institute of Science, Bangalore 560012, India
| | - Wolfgang Mikulits
- Center for Cancer Research, Comprehensive Cancer Center, Medical University of Vienna, 1090 Vienna, Austria; (V.H.)
| |
Collapse
|