1
|
Marquardt AE, Basu M, VanRyzin JW, Ament SA, McCarthy MM. The transcriptome of playfulness is sex biased in the juvenile rat medial amygdala: A role for inhibitory neurons. Cell Rep 2025; 44:115782. [PMID: 40478737 DOI: 10.1016/j.celrep.2025.115782] [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: 09/13/2024] [Revised: 02/28/2025] [Accepted: 05/13/2025] [Indexed: 06/11/2025] Open
Abstract
Social play is a well-conserved, dynamic behavior known to be sexually differentiated. In most species, males play more than females, a sex difference largely driven by the medial amygdala (MeA), yet the full mechanism establishing this bias is unknown. Here, we explore "the transcriptome of playfulness" in both sexes, demonstrating that the transcriptomic profile in the juvenile rat MeA associated with playfulness is markedly distinct in males and females. Parallel single-cell RNA sequencing experiments from newborn rats suggest that inhibitory neurons drive this sex difference. Furthermore, we show that inhibitory neurons comprise the majority of play-active cells in the juvenile MeA, with males having more play-active cells than females, of which a greater proportion are GABAergic. Through integrative bioinformatic analyses, we further explore the expression, function, and cell-type specificity of key play-associated gene modules, providing valuable insight into the sex-biased mechanisms underlying this fundamental social behavior.
Collapse
Affiliation(s)
- Ashley E Marquardt
- Program in Neuroscience, University of Maryland School of Medicine, Baltimore, MD 21201, USA; University of Maryland - Medicine Institute for Neuroscience Discovery, University of Maryland School of Medicine, Baltimore, MD 21201, USA
| | - Mahashweta Basu
- Institute for Genome Sciences, University of Maryland School of Medicine, Baltimore, MD 21201, USA
| | - Jonathan W VanRyzin
- University of Maryland - Medicine Institute for Neuroscience Discovery, University of Maryland School of Medicine, Baltimore, MD 21201, USA; Department of Pharmacology and Physiology, University of Maryland School of Medicine, Baltimore, MD 21201, USA
| | - Seth A Ament
- University of Maryland - Medicine Institute for Neuroscience Discovery, University of Maryland School of Medicine, Baltimore, MD 21201, USA; Institute for Genome Sciences, University of Maryland School of Medicine, Baltimore, MD 21201, USA; Department of Psychiatry, University of Maryland School of Medicine, Baltimore, MD 21201, USA; Maryland Psychiatric Research Center, University of Maryland School of Medicine, Baltimore, MD 21201, USA
| | - Margaret M McCarthy
- University of Maryland - Medicine Institute for Neuroscience Discovery, University of Maryland School of Medicine, Baltimore, MD 21201, USA; Department of Pharmacology and Physiology, University of Maryland School of Medicine, Baltimore, MD 21201, USA.
| |
Collapse
|
2
|
Pilcher C, Buco PAV, Truong JQ, Ramsland PA, Smeets MF, Walkley CR, Holien JK. Characteristics of the Kelch domain containing (KLHDC) subfamily and relationships with diseases. FEBS Lett 2025; 599:1094-1112. [PMID: 39887712 PMCID: PMC12035522 DOI: 10.1002/1873-3468.15108] [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: 10/28/2024] [Revised: 01/08/2025] [Accepted: 01/13/2025] [Indexed: 02/01/2025]
Abstract
The Kelch protein superfamily is an evolutionary conserved family containing 63 alternate protein coding members. The superfamily is split into three subfamilies: Kelch like (KLHL), Kelch-repeat and bric-a-bracs (BTB) domain containing (KBTBD) and Kelch domain containing protein (KLHDC). The KLHDC subfamily is one of the smallest within the Kelch superfamily, containing 10 primary members. There is little known about the structures and functions of the subfamily; however, they are thought to be involved in several cellular and molecular processes. Recently, there have been significant structural and biochemical advances for KLHDC2, which has aided our understanding of other KLHDC family members. Furthermore, small molecules directly targeting KLHDC2 have been identified, which act as tools for targeted protein degradation. This review utilises this information, in conjunction with a thorough exploration of the structural aspects and potential biological functions to summarise the relationship between KLHDCs and human disease.
Collapse
Affiliation(s)
- Courtney Pilcher
- School of Science, STEM CollegeRMIT UniversityMelbourneAustralia
- St Vincent's Institute of Medical ResearchFitzroyAustralia
| | - Paula Armina V. Buco
- St Vincent's Institute of Medical ResearchFitzroyAustralia
- Department of Medicine, Eastern Hill Academic Centre, Melbourne Medical SchoolThe University of MelbourneCarltonAustralia
| | - Jia Q. Truong
- School of Science, STEM CollegeRMIT UniversityMelbourneAustralia
| | - Paul A. Ramsland
- School of Science, STEM CollegeRMIT UniversityMelbourneAustralia
- Department of ImmunologyMonash UniversityMelbourneAustralia
- Department of Surgery, Austin HealthThe University of MelbourneMelbourneAustralia
| | | | - Carl R. Walkley
- St Vincent's Institute of Medical ResearchFitzroyAustralia
- Department of Medicine, Eastern Hill Academic Centre, Melbourne Medical SchoolThe University of MelbourneCarltonAustralia
- Centre for Innate Immunity and Infectious DiseasesHudson Institute of Medical ResearchMelbourneAustralia
- Department of Molecular and Translational ScienceMonash UniversityMelbourneAustralia
| | - Jessica K. Holien
- School of Science, STEM CollegeRMIT UniversityMelbourneAustralia
- St Vincent's Institute of Medical ResearchFitzroyAustralia
- Department of Medicine, Eastern Hill Academic Centre, Melbourne Medical SchoolThe University of MelbourneCarltonAustralia
| |
Collapse
|
3
|
Fang J, Wang J, Zhao X, Yang Y, Xiao Y. KLHDC8A knockdown in normal ovarian epithelial cells promoted the polarization of pro-tumoral macrophages via the C5a/C5aR/p65 NFκB signaling pathway. Cell Immunol 2025; 409-410:104913. [PMID: 39805213 DOI: 10.1016/j.cellimm.2024.104913] [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: 09/10/2024] [Revised: 12/13/2024] [Accepted: 12/22/2024] [Indexed: 01/16/2025]
Abstract
AIMS Tumor-associated macrophages (TAM) is related to Ovarian cancer (OC) pathogenesis, but the exact mechanism remains unclear. This study investigated the expression of Kelch Domain Containing 8 A (KLHDC8A) in OC and the mechanism associated with TAM. MAIN METHODS Bioinformatics analysis of differential expression genes between normal and OC tissues were analyzed based on the Tumor Genome Atlas (TCGA) databases. KLHDC8A mRNA expression was knocked down in normal epithelial cells (IOSE80), and then the effects of siKLHDC8A on the proliferation, invasion, migration and C5a secretion of IOSE80 cells were explored. THP1-derived macrophages were cultured with medium of NC-IOSE80 cells, siKLHDC8A-IOSE80 cells with or without C5aR antagonists. KEY FINDINGS KLHDC8A was lowly expressed in OC and negatively correlated with the infiltration of tumor-promoting macrophages, contributing to the survival of OC patients. Furthermore, siKLHDC8A promotes the proliferation, invasion and migration of IOSE80 cells and leads to polarization of pro-tumoral macrophages, which can be rescued by C5aR antagonists. SIGNIFICANCE Our results indicated that KLHDC8A knockdown could modulate the development of OC by affecting macrophage polarization to pro-tumoral type via the C5a/C5aR/p65 NFκB signaling pathway. It may play an essential role as the tumor suppressor genes in diagnosis and treatment of OC.
Collapse
Affiliation(s)
- Jie Fang
- Department of Gynecology, the Affiliated Hospital of Jiangsu University, Jiangsu University, Zhenjiang, Jiangsu 212001, China.
| | - Jin Wang
- Department of Gynecology, the Affiliated Hospital of Jiangsu University, Jiangsu University, Zhenjiang, Jiangsu 212001, China
| | - Xinyue Zhao
- Department of Gynecology, the Affiliated Hospital of Jiangsu University, Jiangsu University, Zhenjiang, Jiangsu 212001, China
| | - Yaping Yang
- Department of Gynecology, the Affiliated Hospital of Jiangsu University, Jiangsu University, Zhenjiang, Jiangsu 212001, China
| | - Yujia Xiao
- Department of Gynecology, the Affiliated Hospital of Jiangsu University, Jiangsu University, Zhenjiang, Jiangsu 212001, China
| |
Collapse
|
4
|
Marquardt AE, Basu M, VanRyzin JW, Ament SA, McCarthy MM. The transcriptome of playfulness is sex-biased in the juvenile rat medial amygdala: a role for inhibitory neurons. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.09.11.612456. [PMID: 39314276 PMCID: PMC11419002 DOI: 10.1101/2024.09.11.612456] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 09/25/2024]
Abstract
Social play is a dynamic behavior known to be sexually differentiated; in most species, males play more than females, a sex difference driven in large part by the medial amygdala (MeA). Despite the well-conserved nature of this sex difference and the importance of social play for appropriate maturation of brain and behavior, the full mechanism establishing the sex bias in play is unknown. Here, we explore "the transcriptome of playfulness" in the juvenile rat MeA, assessing differences in gene expression between high- and low-playing animals of both sexes via bulk RNA-sequencing. Using weighted gene co-expression network analysis (WGCNA) to identify gene modules combined with analysis of differentially expressed genes (DEGs), we demonstrate that the transcriptomic profile in the juvenile rat MeA associated with playfulness is largely distinct in males compared to females. Of the 13 play-associated WGCNA networks identified, only two were associated with play in both sexes, and very few DEGs associated with playfulness were shared between males and females. Data from our parallel single-cell RNA-sequencing experiments using amygdala samples from newborn male and female rats suggests that inhibitory neurons drive this sex difference, as the majority of sex-biased DEGs in the neonatal amygdala are enriched within this population. Supporting this notion, we demonstrate that inhibitory neurons comprise the majority of play-active cells in the juvenile MeA, with males having a greater number of play-active cells than females, of which a larger proportion are GABAergic. Through integrative bioinformatic analyses, we further explore the expression, function, and cell-type specificity of key play-associated modules and the regulator "hub genes" predicted to drive them, providing valuable insight into the sex-biased mechanisms underlying this fundamental social behavior.
Collapse
|
5
|
Wu X, Fu M, Ge C, Zhou H, Huang H, Zhong M, Zhang M, Xu H, Zhu G, Hua W, Lv K, Yang H. m 6A-Mediated Upregulation of lncRNA CHASERR Promotes the Progression of Glioma by Modulating the miR-6893-3p/TRIM14 Axis. Mol Neurobiol 2024; 61:5418-5440. [PMID: 38193984 DOI: 10.1007/s12035-023-03911-w] [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: 08/03/2023] [Accepted: 12/28/2023] [Indexed: 01/10/2024]
Abstract
Long noncoding RNAs (lncRNAs) play crucial roles in tumor progression and are dysregulated in glioma. However, the functional roles of lncRNAs in glioma remain largely unknown. In this study, we utilized the TCGA (the Cancer Genome Atlas database) and GEPIA2 (Gene Expression Profiling Interactive Analysis 2) databases and observed the overexpression of lncRNA CHASERR in glioma tissues. We subsequently investigated this phenomenon in glioma cell lines. The effects of lncRNA CHASERR on glioma proliferation, migration, and invasion were analyzed using in vitro and in vivo experiments. Additionally, the regulatory mechanisms among PTEN/p-Akt/mTOR and Wnt/β-catenin, lncRNA CHASERR, Micro-RNA-6893-3p(miR-6893-3p), and tripartite motif containing14 (TRIM14) were investigated via bioinformatics analyses, quantitative real-time PCR (qRT-PCR), western blot (WB), RNA immunoprecipitation (RIP), dual luciferase reporter assay, fluorescence in situ hybridization (FISH), and RNA sequencing assays. RIP and RT-qRCR were used to analyze the regulatory effect of N6-methyladenosine(m6A) on the aberrantly expressed lncRNA CHASERR. High lncRNA CHASERR expression was observed in glioma tissues and was associated with unfavorable prognosis in glioma patients. Further functional assays showed that lncRNA CHASERR regulates glioma growth and metastasis in vitro and in vivo. Mechanistically, lncRNA CHASERR sponged miR-6893-3p to upregulate TRIM14 expression, thereby facilitating glioma progression. Additionally, the activation of PTEN/p-Akt/mTOR and Wnt/β-catenin pathways by lncRNA CHASERR, miR-6893-3p, and TRIM14 was found to regulate glioma progression. Moreover, the upregulation of lncRNA CHASERR was observed in response to N6-methyladenosine modification, which was facilitated by METTL3/YTHDF1-mediated RNA transcripts. This study elucidates the m6A/lncRNACHASERR/miR-6893-3p/TRIM14 pathway that contributes to glioma progression and underscores the potential of lncRNA CHASERR as a novel prognostic indicator and therapeutic target for glioma.
Collapse
Affiliation(s)
- Xingwei Wu
- Anhui Province Key Laboratory of Non-Coding RNA Basic Research and Clinical Transformation, Wannan Medical College, Wuhu, 241001, China
- Key Laboratory of Non-Coding RNA Transformation Research of Anhui Higher Education Institution (Yijishan Hospital of Wannan Medical College), Wuhu, 241001, Anhui, China
- Central Laboratory, Yijishan Hospital, The First Affiliated Hospital of Wannan Medical College, Wuhu, 241001, Anhui, China
| | - Minjie Fu
- Department of Neurosurgery, Huashan Hospital, Fudan University, Shanghai, 200040, China
- Shanghai Key Laboratory of Brain Function Restoration and Neural Regeneration, Neurosurgical Institute of Fudan University, Shanghai, China
| | - Chang Ge
- Department of Psychology, Zhejiang Sci-Tech University, Hangzhou, 310000, Zhejiang, China
| | - Hanyu Zhou
- Anhui Province Key Laboratory of Non-Coding RNA Basic Research and Clinical Transformation, Wannan Medical College, Wuhu, 241001, China
- Key Laboratory of Non-Coding RNA Transformation Research of Anhui Higher Education Institution (Yijishan Hospital of Wannan Medical College), Wuhu, 241001, Anhui, China
- Central Laboratory, Yijishan Hospital, The First Affiliated Hospital of Wannan Medical College, Wuhu, 241001, Anhui, China
- Department of Psychology, Zhejiang Sci-Tech University, Hangzhou, 310000, Zhejiang, China
| | - Haoyu Huang
- Anhui Province Key Laboratory of Non-Coding RNA Basic Research and Clinical Transformation, Wannan Medical College, Wuhu, 241001, China
- Key Laboratory of Non-Coding RNA Transformation Research of Anhui Higher Education Institution (Yijishan Hospital of Wannan Medical College), Wuhu, 241001, Anhui, China
- Central Laboratory, Yijishan Hospital, The First Affiliated Hospital of Wannan Medical College, Wuhu, 241001, Anhui, China
| | - Min Zhong
- Anhui Province Key Laboratory of Non-Coding RNA Basic Research and Clinical Transformation, Wannan Medical College, Wuhu, 241001, China
- Key Laboratory of Non-Coding RNA Transformation Research of Anhui Higher Education Institution (Yijishan Hospital of Wannan Medical College), Wuhu, 241001, Anhui, China
- Central Laboratory, Yijishan Hospital, The First Affiliated Hospital of Wannan Medical College, Wuhu, 241001, Anhui, China
- Department of Psychology, Zhejiang Sci-Tech University, Hangzhou, 310000, Zhejiang, China
| | - Mengying Zhang
- Anhui Province Key Laboratory of Non-Coding RNA Basic Research and Clinical Transformation, Wannan Medical College, Wuhu, 241001, China
- Key Laboratory of Non-Coding RNA Transformation Research of Anhui Higher Education Institution (Yijishan Hospital of Wannan Medical College), Wuhu, 241001, Anhui, China
- Central Laboratory, Yijishan Hospital, The First Affiliated Hospital of Wannan Medical College, Wuhu, 241001, Anhui, China
- Department of Psychology, Zhejiang Sci-Tech University, Hangzhou, 310000, Zhejiang, China
| | - Hao Xu
- Department of Neurosurgery, Huashan Hospital, Fudan University, Shanghai, 200040, China
| | - Guoping Zhu
- Anhui Provincial Key Laboratory of Molecular Enzymology and Mechanism of Major Diseases, College of Life Sciences, Anhui Normal University, Wuhu, 241001, Anhui, China.
- College of Life Sciences, Key Laboratory of Biomedicine in Gene Diseases and Health of Anhui Higher Education Institutes, Anhui Normal University, Wuhu, 241001, Anhui, China.
- Auhui Provincial Engineering Research Centre for Molecular Detection and Diagnostics, College of Life Sciences, Anhui Normal University, Wuhu, 241001, Anhui, China.
| | - Wei Hua
- Department of Neurosurgery, Huashan Hospital, Fudan University, Shanghai, 200040, China.
- Shanghai Key Laboratory of Brain Function Restoration and Neural Regeneration, Neurosurgical Institute of Fudan University, Shanghai, China.
| | - Kun Lv
- Anhui Province Key Laboratory of Non-Coding RNA Basic Research and Clinical Transformation, Wannan Medical College, Wuhu, 241001, China.
- Key Laboratory of Non-Coding RNA Transformation Research of Anhui Higher Education Institution (Yijishan Hospital of Wannan Medical College), Wuhu, 241001, Anhui, China.
- Central Laboratory, Yijishan Hospital, The First Affiliated Hospital of Wannan Medical College, Wuhu, 241001, Anhui, China.
- Anhui Provincial Key Laboratory of Molecular Enzymology and Mechanism of Major Diseases, College of Life Sciences, Anhui Normal University, Wuhu, 241001, Anhui, China.
- College of Life Sciences, Key Laboratory of Biomedicine in Gene Diseases and Health of Anhui Higher Education Institutes, Anhui Normal University, Wuhu, 241001, Anhui, China.
- Auhui Provincial Engineering Research Centre for Molecular Detection and Diagnostics, College of Life Sciences, Anhui Normal University, Wuhu, 241001, Anhui, China.
- Clinical Research Center for Critical Respiratory Medicine of Anhui Province, Yijishan Hospital, The First Affiliated Hospital of Wannan Medical College, Wuhu, 241001, Anhui, China.
| | - Hui Yang
- Anhui Province Key Laboratory of Non-Coding RNA Basic Research and Clinical Transformation, Wannan Medical College, Wuhu, 241001, China.
- Key Laboratory of Non-Coding RNA Transformation Research of Anhui Higher Education Institution (Yijishan Hospital of Wannan Medical College), Wuhu, 241001, Anhui, China.
- Central Laboratory, Yijishan Hospital, The First Affiliated Hospital of Wannan Medical College, Wuhu, 241001, Anhui, China.
- Anhui Provincial Key Laboratory of Molecular Enzymology and Mechanism of Major Diseases, College of Life Sciences, Anhui Normal University, Wuhu, 241001, Anhui, China.
- College of Life Sciences, Key Laboratory of Biomedicine in Gene Diseases and Health of Anhui Higher Education Institutes, Anhui Normal University, Wuhu, 241001, Anhui, China.
- Auhui Provincial Engineering Research Centre for Molecular Detection and Diagnostics, College of Life Sciences, Anhui Normal University, Wuhu, 241001, Anhui, China.
- Clinical Research Center for Critical Respiratory Medicine of Anhui Province, Yijishan Hospital, The First Affiliated Hospital of Wannan Medical College, Wuhu, 241001, Anhui, China.
| |
Collapse
|
6
|
Ying T, Lai Y, Lu S, E S. Identification and validation of a glycolysis-related taxonomy for improving outcomes in glioma. CNS Neurosci Ther 2024; 30:e14601. [PMID: 38332637 PMCID: PMC10853657 DOI: 10.1111/cns.14601] [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: 08/26/2023] [Revised: 12/11/2023] [Accepted: 12/29/2023] [Indexed: 02/10/2024] Open
Abstract
BACKGROUND Reprogramming of glucose metabolism is a prominent abnormal energy metabolism in glioma. However, the efficacy of treatments targeting glycolysis varies among patients. The present study aimed to classify distinct glycolysis subtypes (GS) of glioma, which may help to improve the therapy response. METHODS The expression profiles of glioma were downloaded from public datasets to perform an enhanced clustering analysis to determine the GS. A total of 101 combinations based on 10 machine learning algorithms were performed to screen out the most valuable glycolysis-related glioma signature (GGS). Through RSF and plsRcox algorithms, adrenomedullin (ADM) was eventually obtained as the most significant glycolysis-related gene for prognostic prediction in glioma. Furthermore, drug sensitivity analysis, molecular docking, and in vitro experiments were utilized to verify the efficacy of ADM and ingenol mebutate (IM). RESULTS Glioma patients were classified into five distinct GS (GS1-GS5), characterized by varying glycolytic metabolism levels, molecular expression, immune cell infiltration, immunogenic modulators, and clinical features. Anti-CTLA4 and anti-PD-L1 antibodies significantly improved the prognosis for GS2 and GS5, respectively. ADM has been identified as a potential biomarker for targeted glycolytic therapy in glioma patients. In vitro experiments demonstrated that IM inhibited glioma cell progression by inhibiting ADM. CONCLUSION This study elucidates that evaluating GS is essential for comprehending the heterogeneity of glioma, which is pivotal for predicting immune cell infiltration (ICI) characterization, prognosis, and personalized immunotherapy regimens. We also explored the glycolysis-related genes ADM and IM to develop a theoretical framework for anti-tumor strategies targeting glycolysis.
Collapse
Affiliation(s)
- Tianshu Ying
- Department of OncologyShengjing Hospital of China Medical UniversityShenyangChina
| | - Yaming Lai
- Department of UrologyGuangyuan Central HospitalGuangyuanChina
| | - Shiyang Lu
- Department of UrologyShengjing Hospital of China Medical UniversityShenyangChina
| | - Shaolong E
- Department of UrologyShengjing Hospital of China Medical UniversityShenyangChina
| |
Collapse
|
7
|
YU Z, LI J, JIANG J, LI Y, LIN L, XIA Y, WANG L. [miRNA-128-3p inhibits malignant behavior of glioma cells by downregulating KLHDC8A expression]. NAN FANG YI KE DA XUE XUE BAO = JOURNAL OF SOUTHERN MEDICAL UNIVERSITY 2023; 43:1447-1459. [PMID: 37814858 PMCID: PMC10563106 DOI: 10.12122/j.issn.1673-4254.2023.09.01] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Subscribe] [Scholar Register] [Received: 05/23/2023] [Accepted: 09/18/2023] [Indexed: 10/11/2023]
Abstract
OBJECTIVE To determine whether miRNA-128-3p regulates malignant biological behavior of glioma cells by targeting KLHDC8A. METHODS Dual-luciferase reporter assays, qRT-PCR and Western blotting were used to verify the targeting of miRNA-128-3p to KLHDC8A. Edu assay, flow cytometry, Transwell assay and would healing assay were used to determine the effects of changes in miRNA-128-3p and KLHDC8A expression levels on malignant behavior of glioma cells. Rescue experiment was carried out to verify that miRNA-128-3p regulated glioma cell proliferation, apoptosis, invasion and migration by targeting KLHDC8A. RESULTS The expression level of KLHDC8A was significantly increased in high-grade glioma tissue and was closely related to a poor survival outcome of the patients. Overexpression of KLHDC8A promoted glioma cell proliferation, migration and invasion, and miRNA-128-3p overexpression inhibited proliferative and metastatic capacities of glioma cells. Mechanistically, KLHDC8A expression was directly modulated by miRNA-128-3p, which, by targeting KLHDC8A, inhibited malignant behavior of glioma cells. CONCLUSION Upregulation of miRNA-128-3p inhibits uncontrolled growth of glioma cells by negatively regulating KLHDC8A expression and its downstream effectors, suggesting that the miRNA-128-3p-KLHDC8A axis may serve as a potential prognostic indicator and a therapeutic target for developing new strategies for glioma treatment.
Collapse
Affiliation(s)
- Zhengtao YU
- Department of Neurosurgery, Affiliated Haikou Hospital of Xiangya School of Central South University, Haikou 570208, China中南大学湘雅医学院附属海口医院神经外科,海南 海口 570208
| | - Jiameng LI
- Department of Neurosurgery, Affiliated Haikou Hospital of Xiangya School of Central South University, Haikou 570208, China中南大学湘雅医学院附属海口医院神经外科,海南 海口 570208
| | - Junwen JIANG
- Department of Neurosurgery, Affiliated Haikou Hospital of Xiangya School of Central South University, Haikou 570208, China中南大学湘雅医学院附属海口医院神经外科,海南 海口 570208
| | - You LI
- Department of Neurosurgery, Affiliated Haikou Hospital of Xiangya School of Central South University, Haikou 570208, China中南大学湘雅医学院附属海口医院神经外科,海南 海口 570208
| | - Long LIN
- Department of Neurosurgery, Affiliated Haikou Hospital of Xiangya School of Central South University, Haikou 570208, China中南大学湘雅医学院附属海口医院神经外科,海南 海口 570208
| | - Ying XIA
- Department of Neurosurgery, Affiliated Haikou Hospital of Xiangya School of Central South University, Haikou 570208, China中南大学湘雅医学院附属海口医院神经外科,海南 海口 570208
| | - Lei WANG
- Department of Neurosurgery, Hunan Cancer Hospital and Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University, Changsha 410006, China中南大学湘雅医学院附属肿瘤医院神经外科,湖南 长沙 410006
| |
Collapse
|
8
|
Zhu X, Wu X, Yang H, Xu Q, Zhang M, Liu X, Lv K. m 6A-mediated upregulation of LINC01003 regulates cell migration by targeting the CAV1/FAK signaling pathway in glioma. Biol Direct 2023; 18:27. [PMID: 37270527 DOI: 10.1186/s13062-023-00386-6] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2022] [Accepted: 05/30/2023] [Indexed: 06/05/2023] Open
Abstract
BACKGROUND Long non-coding RNAs (lncRNAs) play important roles in the progression of glioma. Here, we examined the potential functions of a lncRNA, LINC01003, in glioma and characterized the underlying molecular mechanisms. METHODS The GEIPA2 and Chinese Glioma Genome Atlas (CCGA) databases were employed to analyze gene expression and the overall survival curve in patients with glioma. The functions of LINC01003 in glioma growth and migration were assessed by loss-of-function experiments in vitro and in vivo. RNA sequencing was used to determine the signaling pathways effected by LINC01003. Bioinformatics analysis and RNA immunoprecipitation (RIP) assays were used to explore the mechanism underlying the N6-methyladenine (m6A) modification-dependent upregulation of LINC01003 in glioma. RESULTS LINC01003 expression was upregulated in glioma cell lines and tissues. Higher LINC01003 expression predicted shorter overall survival time in glioma patients. Functionally, LINC01003 knockdown inhibited the cell cycle and cell proliferation and migration in glioma cells. Mechanistically, RNA sequencing revealed that LINC01003 mediated the focal adhesion signaling pathway. Furthermore, LINC01003 upregulation is induced by m6A modification regulated by METTL3. CONCLUSION This study characterized LINC01003 as a lncRNA that contributes to tumorigenesis in glioma and demonstrated that the LINC01003-CAV1-FAK axis serves as a potential therapeutic target for glioma.
Collapse
Affiliation(s)
- Xiaolong Zhu
- Central Laboratory, The First Affiliated Hospital of Wannan Medical College (Yijishan Hospital of Wannan Medical College), Wuhu, 241001, People's Republic of China
- Key Laboratory of Non-Coding RNA Transformation Research of Anhui Higher Education Institutes (Wannan Medical College), Wuhu, 241001, People's Republic of China
- Non-Coding RNA Research Center of Wannan Medical College, Wuhu, 241001, People's Republic of China
- Anhui Provincial Clinical Research Center for Critical Respiratory Disease, Wuhu, 241001, People's Republic of China
| | - Xingwei Wu
- Central Laboratory, The First Affiliated Hospital of Wannan Medical College (Yijishan Hospital of Wannan Medical College), Wuhu, 241001, People's Republic of China
- Key Laboratory of Non-Coding RNA Transformation Research of Anhui Higher Education Institutes (Wannan Medical College), Wuhu, 241001, People's Republic of China
- Non-Coding RNA Research Center of Wannan Medical College, Wuhu, 241001, People's Republic of China
| | - Hui Yang
- Central Laboratory, The First Affiliated Hospital of Wannan Medical College (Yijishan Hospital of Wannan Medical College), Wuhu, 241001, People's Republic of China
- Key Laboratory of Non-Coding RNA Transformation Research of Anhui Higher Education Institutes (Wannan Medical College), Wuhu, 241001, People's Republic of China
- Non-Coding RNA Research Center of Wannan Medical College, Wuhu, 241001, People's Republic of China
- Anhui Provincial Clinical Research Center for Critical Respiratory Disease, Wuhu, 241001, People's Republic of China
| | - Qiancheng Xu
- Anhui Provincial Clinical Research Center for Critical Respiratory Disease, Wuhu, 241001, People's Republic of China
- Department of Critical Care Medicine, The First Affiliated Hospital of Wannan Medical College (Yijishan Hospital), Wuhu, 241001, People's Republic of China
| | - Mengying Zhang
- Central Laboratory, The First Affiliated Hospital of Wannan Medical College (Yijishan Hospital of Wannan Medical College), Wuhu, 241001, People's Republic of China
- Key Laboratory of Non-Coding RNA Transformation Research of Anhui Higher Education Institutes (Wannan Medical College), Wuhu, 241001, People's Republic of China
- Non-Coding RNA Research Center of Wannan Medical College, Wuhu, 241001, People's Republic of China
- Anhui Provincial Clinical Research Center for Critical Respiratory Disease, Wuhu, 241001, People's Republic of China
| | - Xiaocen Liu
- Department of Nuclear Medicine, The First Affiliated Hospital of Wannan Medical College (Yijishan Hospital of Wannan Medical College), Wuhu, 241001, Anhui, People's Republic of China
| | - Kun Lv
- Central Laboratory, The First Affiliated Hospital of Wannan Medical College (Yijishan Hospital of Wannan Medical College), Wuhu, 241001, People's Republic of China.
- Key Laboratory of Non-Coding RNA Transformation Research of Anhui Higher Education Institutes (Wannan Medical College), Wuhu, 241001, People's Republic of China.
- Non-Coding RNA Research Center of Wannan Medical College, Wuhu, 241001, People's Republic of China.
- Anhui Provincial Clinical Research Center for Critical Respiratory Disease, Wuhu, 241001, People's Republic of China.
| |
Collapse
|
9
|
Allegra A, Murdaca G, Mirabile G, Gangemi S. Redox Signaling Modulates Activity of Immune Checkpoint Inhibitors in Cancer Patients. Biomedicines 2023; 11:1325. [PMID: 37238995 PMCID: PMC10215686 DOI: 10.3390/biomedicines11051325] [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: 04/06/2023] [Revised: 04/23/2023] [Accepted: 04/28/2023] [Indexed: 05/28/2023] Open
Abstract
Although immunotherapy is already a staple of cancer care, many patients may not benefit from these cutting-edge treatments. A crucial field of research now focuses on figuring out how to improve treatment efficacy and assess the resistance mechanisms underlying this uneven response. For a good response, immune-based treatments, in particular immune checkpoint inhibitors, rely on a strong infiltration of T cells into the tumour microenvironment. The severe metabolic environment that immune cells must endure can drastically reduce effector activity. These immune dysregulation-related tumour-mediated perturbations include oxidative stress, which can encourage lipid peroxidation, ER stress, and T regulatory cells dysfunction. In this review, we have made an effort to characterize the status of immunological checkpoints, the degree of oxidative stress, and the part that latter plays in determining the therapeutic impact of immunological check point inhibitors in different neoplastic diseases. In the second section of the review, we will make an effort to assess new therapeutic possibilities that, by affecting redox signalling, may modify the effectiveness of immunological treatment.
Collapse
Affiliation(s)
- Alessandro Allegra
- Division of Hematology, Department of Human Pathology in Adulthood and Childhood “Gaetano Barresi”, University of Messina, 98125 Messina, Italy;
| | - Giuseppe Murdaca
- Department of Internal Medicine, Ospedale Policlinico San Martino IRCCS, University of Genova, Viale Benedetto XV, n. 6, 16132 Genova, Italy
| | - Giuseppe Mirabile
- Division of Hematology, Department of Human Pathology in Adulthood and Childhood “Gaetano Barresi”, University of Messina, 98125 Messina, Italy;
| | - Sebastiano Gangemi
- Allergy and Clinical Immunology Unit, Department of Clinical and Experimental Medicine, University of Messina, 98125 Messina, Italy;
| |
Collapse
|
10
|
Lee D, Gimple RC, Wu X, Prager BC, Qiu Z, Wu Q, Daggubati V, Mariappan A, Gopalakrishnan J, Sarkisian MR, Raleigh DR, Rich JN. Superenhancer activation of KLHDC8A drives glioma ciliation and hedgehog signaling. J Clin Invest 2023; 133:e163592. [PMID: 36394953 PMCID: PMC9843063 DOI: 10.1172/jci163592] [Citation(s) in RCA: 15] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2022] [Accepted: 11/15/2022] [Indexed: 11/18/2022] Open
Abstract
Glioblastoma ranks among the most aggressive and lethal of all human cancers. Self-renewing, highly tumorigenic glioblastoma stem cells (GSCs) contribute to therapeutic resistance and maintain cellular heterogeneity. Here, we interrogated superenhancer landscapes of primary glioblastoma specimens and patient-derived GSCs, revealing a kelch domain-containing gene, specifically Kelch domain containing 8A (KLHDC8A) with a previously unknown function as an epigenetically driven oncogene. Targeting KLHDC8A decreased GSC proliferation and self-renewal, induced apoptosis, and impaired in vivo tumor growth. Transcription factor control circuitry analyses revealed that the master transcriptional regulator SOX2 stimulated KLHDC8A expression. Mechanistically, KLHDC8A bound chaperonin-containing TCP1 (CCT) to promote the assembly of primary cilia to activate hedgehog signaling. KLHDC8A expression correlated with Aurora B/C Kinase inhibitor activity, which induced primary cilia and hedgehog signaling. Combinatorial targeting of Aurora B/C kinase and hedgehog displayed augmented benefit against GSC proliferation. Collectively, superenhancer-based discovery revealed KLHDC8A as what we believe to be a novel molecular target of cancer stem cells that promotes ciliogenesis to activate the hedgehog pathway, offering insights into therapeutic vulnerabilities for glioblastoma treatment.
Collapse
Affiliation(s)
- Derrick Lee
- UPMC Hillman Cancer Center, Pittsburgh, Pennsylvania, USA
- Department of Medicine, University of Pittsburgh, Pittsburgh, Pennsylvania, USA
- Division of Regenerative Medicine, Department of Medicine, UCSD, La Jolla, California, USA
| | - Ryan C. Gimple
- Division of Regenerative Medicine, Department of Medicine, UCSD, La Jolla, California, USA
- Department of Pathology, Case Western Reserve University, Cleveland, Ohio, USA
| | - Xujia Wu
- UPMC Hillman Cancer Center, Pittsburgh, Pennsylvania, USA
- Department of Medicine, University of Pittsburgh, Pittsburgh, Pennsylvania, USA
| | - Briana C. Prager
- Division of Regenerative Medicine, Department of Medicine, UCSD, La Jolla, California, USA
- Cleveland Clinic Lerner College of Medicine, Cleveland, Ohio, USA
| | - Zhixin Qiu
- UPMC Hillman Cancer Center, Pittsburgh, Pennsylvania, USA
- Department of Medicine, University of Pittsburgh, Pittsburgh, Pennsylvania, USA
- Division of Regenerative Medicine, Department of Medicine, UCSD, La Jolla, California, USA
| | - Qiulian Wu
- UPMC Hillman Cancer Center, Pittsburgh, Pennsylvania, USA
- Department of Medicine, University of Pittsburgh, Pittsburgh, Pennsylvania, USA
- Division of Regenerative Medicine, Department of Medicine, UCSD, La Jolla, California, USA
| | - Vikas Daggubati
- Department of Radiation Oncology and
- Department of Neurological Surgery, UCSF, San Francisco, California, USA
| | - Aruljothi Mariappan
- Institute of Human Genetics, University Hospital Düsseldorf, Heinrich-Heine-Universität, Düsseldorf, Germany
| | - Jay Gopalakrishnan
- Institute of Human Genetics, University Hospital Düsseldorf, Heinrich-Heine-Universität, Düsseldorf, Germany
| | - Matthew R. Sarkisian
- Department of Neuroscience, McKnight Brain Institute and
- Preston A. Wells, Jr. Center for Brain Tumor Therapy, University of Florida, Gainesville, Florida, USA
| | - David R. Raleigh
- Department of Radiation Oncology and
- Department of Neurological Surgery, UCSF, San Francisco, California, USA
| | - Jeremy N. Rich
- UPMC Hillman Cancer Center, Pittsburgh, Pennsylvania, USA
- Division of Regenerative Medicine, Department of Medicine, UCSD, La Jolla, California, USA
- Department of Neurology, University of Pittsburgh, Pittsburgh, Pennsylvania, USA
| |
Collapse
|
11
|
Abstract
Histone lactylation, an indicator of lactate level and glycolysis, has intrinsic connections with cell metabolism that represents a novel epigenetic code affecting the fate of cells including carcinogenesis. Through delineating the relationship between histone lactylation and cancer hallmarks, we propose histone lactylation as a novel epigenetic code priming cells toward the malignant state, and advocate the importance of identifying novel therapeutic strategies or dual-targeting modalities against lactylation toward effective cancer control. This review underpins important yet less-studied area in histone lactylation, and sheds insights on its clinical impact as well as possible therapeutic tools targeting lactylation.
Collapse
|
12
|
Zhang L, Wu X, Fan X, Ai H. MUM1L1 as a Tumor Suppressor and Potential Biomarker in Ovarian Cancer: Evidence from Bioinformatics Analysis and Basic Experiments. Comb Chem High Throughput Screen 2023; 26:2487-2501. [PMID: 36856181 DOI: 10.2174/1386207326666230301141912] [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: 07/22/2022] [Revised: 12/01/2022] [Accepted: 12/15/2022] [Indexed: 03/02/2023]
Abstract
BACKGROUND Ovarian cancer (OC) is the most prevalent gynecologic malignancy, with high mortality rates. However, its pathogenesis remains unclear. The current study aimed to explore potential biomarkers and suppressor genes for diagnosing and treating OC. METHODS Biochemical and bioinformatics approaches were used to detect differentially expressed genes (DEGs) in ovarian tissues via integration analysis. Kaplan-Meier plot analysis was performed to assess progression-free survival and overall survival according to DEGs. Then, we constructed a protein-protein interaction (PPI) network based on data from the STRING database to identify the related target genes of DEGs. Finally, DEGs regulating the proliferation, migration, and invasion of SKOV3 cell lines were validated via in vitro experiments. RESULTS Four DEGs (MUM1L1, KLHDC8A, CRYGD, and GREB1) with enriched expression in ovarian tissues were explicitly expressed in the ovary based on an analysis of all human proteins. MUM1L1 had high specificity, and its expression was higher in normal ovarian tissues than in OC tissues. Kaplan-Meier plot analysis showed that a high MUM1L1 expression was associated with longer progression-free survival and overall survival in OC. Based on the PPI analysis results, CBLN4, CBLN1, PTH2R, TMEM255B, and COL23A1 were associated with MUM1L1. In vitro studies revealed that MUM1L1 overexpression decreased the proliferation, migration, and invasion ability of SKOV3 cell lines. Meanwhile, MUM1L1 knockdown had contrasting results. CONCLUSION MUM1L1 is a tumor suppressor gene and is a potential biomarker for diagnosing and treating OC.
Collapse
Affiliation(s)
- Lu Zhang
- Graduate School, Jinzhou Medical University, Jinzhou, Liaoning 121000, China
| | - Xue Wu
- Graduate School, Jinzhou Medical University, Jinzhou, Liaoning 121000, China
| | - Xue Fan
- Department of Obstetrics and Gynecology, 3rd Affiliated Hospital of Jinzhou Medical University, Jinzhou, Liaoning 121000, China
| | - Hao Ai
- Graduate School, Jinzhou Medical University, Jinzhou, Liaoning 121000, China
- Department of Obstetrics and Gynecology, 3rd Affiliated Hospital of Jinzhou Medical University, Jinzhou, Liaoning 121000, China
| |
Collapse
|
13
|
Cheng T, Xu M, Zhang H, Lu B, Zhang X, Wang Z, Huang J. KLHDC8A Expression in Association with Macrophage Infiltration and Oxidative Stress Predicts Unfavorable Prognosis for Glioma. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2022; 2022:2694377. [PMID: 36199422 PMCID: PMC9527113 DOI: 10.1155/2022/2694377] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/29/2022] [Revised: 08/22/2022] [Accepted: 09/01/2022] [Indexed: 12/21/2022]
Abstract
Background The tumor immune microenvironment (TME) is associated with cancer progression and immune escape. Although KLHDC8A has been reported in glioma in vitro, the expression and clinical significance of this gene in clinical samples are unknown. Methods The Cancer Genome Atlas and Chinese Glioma Genome Atlas databases were used to evaluate the mRNA expression level of KLHDC8A and its significance in the glioma TME. Tissue microarray-based multiple immunohistochemical staining was conducted to determine KLHDC8A protein levels and characterize the immune signature of tumor-infiltrating immune cells in gliomas. Results Tumor cells and tumor-associated macrophages expressed KLHDC8A. The expression of KLHDC8A was higher in glioma tissues than in normal brain tissues and was associated with patient clinical characteristics. Gliomas exhibited a high abundance of macrophages, neutrophils, regulatory T cells, and the immune checkpoint PD-L1, as well as high KLHDC8A expression. Cox regression analysis showed that KLHDC8A+CD68+ macrophages and KLHDC8A predicted unfavorable survival in patients with glioma. Finally, protein-protein interaction network analysis showed that the KLHDC8A expression was associated with hypoxia and oxidative stress. Conclusions KLHDC8A is a potential marker for the clinical diagnosis of glioma. The immune characteristics of macrophages play a crucial role in predicting patients with glioma, providing a new avenue for targeted glioma therapy.
Collapse
Affiliation(s)
- Tong Cheng
- Department of Clinical Biobank, Affiliated Hospital of Nantong University & Medical School of Nantong University, Jiangsu 226001, China
| | - Manyu Xu
- Department of Clinical Biobank, Affiliated Hospital of Nantong University & Medical School of Nantong University, Jiangsu 226001, China
| | - Hui Zhang
- Department of Clinical Biobank, Affiliated Hospital of Nantong University & Medical School of Nantong University, Jiangsu 226001, China
| | - Bing Lu
- Department of Clinical Biobank, Affiliated Hospital of Nantong University & Medical School of Nantong University, Jiangsu 226001, China
| | - Xiaojing Zhang
- Department of Clinical Biobank, Affiliated Hospital of Nantong University & Medical School of Nantong University, Jiangsu 226001, China
| | - Ziheng Wang
- Department of Clinical Biobank, Affiliated Hospital of Nantong University & Medical School of Nantong University, Jiangsu 226001, China
| | - Jianfei Huang
- Department of Clinical Biobank, Affiliated Hospital of Nantong University & Medical School of Nantong University, Jiangsu 226001, China
| |
Collapse
|
14
|
Maeda M, Ko M, Mane MM, Cohen IJ, Shindo M, Vemuri K, Serganova I, Blasberg R. Genetic and Drug Inhibition of LDH-A: Effects on Murine Gliomas. Cancers (Basel) 2022; 14:2306. [PMID: 35565435 PMCID: PMC9105502 DOI: 10.3390/cancers14092306] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2021] [Revised: 04/11/2022] [Accepted: 04/26/2022] [Indexed: 02/04/2023] Open
Abstract
The effects of the LDH-A depletion via shRNA knockdown on three murine glioma cell lines and corresponding intracranial (i.c.) tumors were studied and compared to pharmacologic (GNE-R-140) inhibition of the LDH enzyme complex, and to shRNA scrambled control (NC) cell lines. The effects of genetic-shRNA LDH-A knockdown and LDH drug-targeted inhibition (GNE-R-140) on tumor-cell metabolism, tumor growth, and animal survival were similar. LDH-A KD and GNE-R-140 unexpectedly increased the aggressiveness of GL261 intracranial gliomas, but not CT2A and ALTS1C1 i.c. gliomas. Furthermore, the bioenergetic profiles (ECAR and OCR) of GL261 NC and LDH-A KD cells under different nutrient limitations showed that (a) exogenous pyruvate is not a major carbon source for metabolism through the TCA cycle of native GL261 cells; and (b) the unique upregulation of LDH-B that occurs in GL261 LDH-A KD cells results in these cells being better able to: (i) metabolize lactate as a primary carbon source through the TCA cycle, (ii) be a net consumer of lactate, and (iii) showed a significant increase in the proliferation rate following the addition of 10 mM lactate to the glucose-free media (only seen in GL261 KD cells). Our study suggests that inhibition of LDH-A/glycolysis may not be a general strategy to inhibit the i.c. growth of all gliomas, since the level of LDH-A expression and its interplay with LDH-B can lead to complex metabolic interactions between tumor cells and their environment. Metabolic-inhibition treatment strategies need to be carefully assessed, since the inhibition of glycolysis (e.g., inhibition of LDH-A) may lead to the unexpected development and activation of alternative metabolic pathways (e.g., upregulation of lipid metabolism and fatty-acid oxidation pathways), resulting in enhanced tumor-cell survival in a nutrient-limited environment and leading to increased tumor aggressiveness.
Collapse
Affiliation(s)
- Masatomo Maeda
- Department of Neurology, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA; (M.M.); (M.K.); (M.M.M.); (M.S.); (K.V.); (I.S.)
- Molecular Pharmacology and Chemistry Program, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA;
- Department of Neurosurgery, Nozaki Tokushukai Hospital, Osaka 5740074, Japan
| | - Myat Ko
- Department of Neurology, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA; (M.M.); (M.K.); (M.M.M.); (M.S.); (K.V.); (I.S.)
- Molecular Pharmacology and Chemistry Program, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA;
- Human Oncology and Pathogenesis Program, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA
| | - Mayuresh M. Mane
- Department of Neurology, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA; (M.M.); (M.K.); (M.M.M.); (M.S.); (K.V.); (I.S.)
- Molecular Pharmacology and Chemistry Program, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA;
- Department of Radiology, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA
| | - Ivan J. Cohen
- Molecular Pharmacology and Chemistry Program, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA;
- Gerstner Sloan Kettering Graduate School of Biomedical Sciences, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA
- Center for Cellular Immunotherapies, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Masahiro Shindo
- Department of Neurology, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA; (M.M.); (M.K.); (M.M.M.); (M.S.); (K.V.); (I.S.)
- Molecular Pharmacology and Chemistry Program, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA;
- Department of Neurosurgery, Nozaki Tokushukai Hospital, Osaka 5740074, Japan
- Department of Neurosurgery, Yukioka Hospital, Osaka 5740074, Japan
| | - Kiranmayi Vemuri
- Department of Neurology, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA; (M.M.); (M.K.); (M.M.M.); (M.S.); (K.V.); (I.S.)
- Molecular Pharmacology and Chemistry Program, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA;
- Department of Genetics, Rutgers University, New Brunswick, NJ 08901, USA
| | - Inna Serganova
- Department of Neurology, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA; (M.M.); (M.K.); (M.M.M.); (M.S.); (K.V.); (I.S.)
- Molecular Pharmacology and Chemistry Program, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA;
- Weill Cornell Medicine, New York, NY 10021, USA
| | - Ronald Blasberg
- Department of Neurology, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA; (M.M.); (M.K.); (M.M.M.); (M.S.); (K.V.); (I.S.)
- Molecular Pharmacology and Chemistry Program, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA;
- Human Oncology and Pathogenesis Program, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA
| |
Collapse
|
15
|
Carbajo-García MC, Corachán A, Juárez-Barber E, Monleón J, Payá V, Trelis A, Quiñonero A, Pellicer A, Ferrero H. Integrative analysis of the DNA methylome and transcriptome in uterine leiomyoma shows altered regulation of genes involved in metabolism, proliferation, extracellular matrix and vesicles. J Pathol 2022; 257:663-673. [PMID: 35472162 DOI: 10.1002/path.5920] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2022] [Revised: 04/07/2022] [Accepted: 04/22/2022] [Indexed: 11/08/2022]
Abstract
Uterine leiomyomas are the most common benign tumors in women of reproductive age. Despite the high prevalence, tumor pathology remains unclear, which hampers development of safe and effective treatments. Epigenetic mechanisms appear to be involved in uterine leiomyoma development, particularly via DNA methylation that regulates gene expression. We aimed to determine the relationship between DNA methylation and gene expression in uterine leiomyoma compared to adjacent myometrium to identify molecular mechanisms involved in uterine leiomyoma formation that are under epigenetic control. Our results showed a different DNA methylation profile between uterine leiomyoma and myometrium, leading to hypermethylation of uterine leiomyoma, and a different global transcriptome profile. Integration of DNA methylation and whole-transcriptome RNA-sequencing data identified 93 genes regulated by methylation, with 22 hypomethylated/upregulated and 71 hypermethylated/downregulated. Functional enrichment analysis showed dysregulated biological processes and molecular functions involved in metabolism and cell physiology, response to extracellular signals, invasion, and proliferation, as well as pathways related to uterine biology and cancer. Cellular components such as cell membranes, vesicles, extracellular matrix, and cell junctions were dysregulated in uterine leiomyoma. In addition, we found hypomethylation/upregulation of oncogenes (PRL, ATP8B4, CEMIP, ZPMS2-AS1, RIMS2, TFAP2C) and hypermethylation/downregulation of tumor suppressor genes (EFEMP1, FBLN2, ARHGAP10, HTATIP2), which are related to proliferation, invasion, altered metabolism, deposition of extracellular matrix, and Wnt/β-catenin pathway dysregulation. This confirms that key processes of uterine leiomyoma development are under DNA methylation control. Finally, inhibition of DNA methyltransferases by 5-aza-2'-deoxycitidine increased expression of hypermethylated/downregulated genes in uterine leiomyoma cells in vitro. In conclusion, gene regulation by DNA methylation is implicated in uterine leiomyoma pathogenesis, and reversion of this methylation could offer a therapeutic option for uterine leiomyoma. This article is protected by copyright. All rights reserved.
Collapse
Affiliation(s)
- María Cristina Carbajo-García
- Fundación IVI, Instituto de Investigación Sanitaria La Fe, Valencia, Spain.,Departamento de Pediatría, Obstetricia y Ginecología, Universidad de Valencia, Spain
| | - Ana Corachán
- Fundación IVI, Instituto de Investigación Sanitaria La Fe, Valencia, Spain.,Departamento de Pediatría, Obstetricia y Ginecología, Universidad de Valencia, Spain
| | | | - Javier Monleón
- Hospital Universitario y Politécnico La Fe, Valencia, Spain
| | - Vicente Payá
- Hospital Universitario y Politécnico La Fe, Valencia, Spain
| | | | - Alicia Quiñonero
- Fundación IVI, Instituto de Investigación Sanitaria La Fe, Valencia, Spain
| | - Antonio Pellicer
- Fundación IVI, Instituto de Investigación Sanitaria La Fe, Valencia, Spain.,IVIRMA, Rome, Rome, Italy
| | - Hortensia Ferrero
- Fundación IVI, Instituto de Investigación Sanitaria La Fe, Valencia, Spain
| |
Collapse
|
16
|
Zhu X, Chen T, Yang H, Lv K. Lactate induced up-regulation of KLHDC8A (Kelch domain-containing 8A) contributes to the proliferation, migration and apoptosis of human glioma cells. J Cell Mol Med 2020; 24:11691-11702. [PMID: 32851798 PMCID: PMC7579713 DOI: 10.1111/jcmm.15780] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2020] [Revised: 06/19/2020] [Accepted: 07/30/2020] [Indexed: 12/11/2022] Open
Abstract
Glioma is a common type of malignant brain tumour with high mortality and relapse rate. However, the molecular mechanisms of glioma development have not been clarified. Differentially expressed genes in normal brain tissues and glioma tissues, low-grade and high-grade gliomas were screened out with GEO database analysis. We found that KLHDC8A (Kelch domain-containing 8A) expression level was significantly increased in high-grade glioma tissues and that high KLHDC8A expression was closely related with poor prognosis. Function assays indicated that KLHDC8A knockdown inhibited proliferation, migration and invasion, blocked the cell cycle and promoted apoptosis in glioma cells. Mechanistically, KLHDC8A regulated various functions in glioma by directly mediating Bcl2, BAX, p21, CDK2, MMP2 transcription and ERK and P38 MAPK activation. KLHDC8A overexpression enhances glioma tumorgenesis such as cell proliferation, migration and invasion. The ERK and P38 MAPK which activated by KLHDC8A overexpression could be reversed by U0126 and SB203580, respectively. Meanwhile, stimulation of lactate which produced by glycolysis is responsible for induction of KLHDC8A expression. Collectively, we demonstrated that KLHDC8A plays an important role in tumorgenesis of glioma, suggesting that it is a promising prognostic marker and a potential therapy target for the treatment of glioma.
Collapse
Affiliation(s)
- Xiaolong Zhu
- Key Laboratory of Non-coding RNA Transformation Research of Anhui Higher Education Institutes (Wannan Medical College), Wuhu, China.,Non-coding RNA Research Center of Wannan Medical College, Wuhu, China.,Central Laboratory, The first affiliated hospital of Wannan Medical College, Wuhu, China
| | - Tianbing Chen
- Key Laboratory of Non-coding RNA Transformation Research of Anhui Higher Education Institutes (Wannan Medical College), Wuhu, China.,Non-coding RNA Research Center of Wannan Medical College, Wuhu, China.,Central Laboratory, The first affiliated hospital of Wannan Medical College, Wuhu, China
| | - Hui Yang
- Key Laboratory of Non-coding RNA Transformation Research of Anhui Higher Education Institutes (Wannan Medical College), Wuhu, China.,Non-coding RNA Research Center of Wannan Medical College, Wuhu, China.,Central Laboratory, The first affiliated hospital of Wannan Medical College, Wuhu, China
| | - Kun Lv
- Key Laboratory of Non-coding RNA Transformation Research of Anhui Higher Education Institutes (Wannan Medical College), Wuhu, China.,Non-coding RNA Research Center of Wannan Medical College, Wuhu, China.,Central Laboratory, The first affiliated hospital of Wannan Medical College, Wuhu, China
| |
Collapse
|