1
|
Zhou Y, Ma W, Hu H, He Q, Yu C, Chen W, Yu G. Angiogenesis related gene signatures predict prognosis and guide therapeutic strategies in renal clear cell carcinoma. Sci Rep 2025; 15:17030. [PMID: 40379825 PMCID: PMC12084375 DOI: 10.1038/s41598-025-02134-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2024] [Accepted: 05/12/2025] [Indexed: 05/19/2025] Open
Abstract
Kidney tumors are hypervascular tumors with crucial antiangiogenic effects in tumor therapy. This study aimed to develop a predictive model for kidney renal clear cell carcinoma (KIRC) by utilizing angiogenesis-related genes to formulate targeted therapy and immunotherapy strategies. Angiogenesis-related genes were screened via the GeneCard and Molecular Signatures Database (MSigDB). The KIRC data downloaded from The Cancer Genome Atlas (TCGA) were randomly divided into an experimental cohort and a validation cohort. In the experimental cohort, a risk score prediction model was constructed through successive analyses via univariate Cox regression, LASSO regression, and multivariate Cox regression. Receiver operating characteristic (ROC) curves were employed to assess the sensitivity of the model's predictions. The model's stability and generalizability were subsequently validated in both the validation cohort and the E-MTAB-1980 cohort. Subsequently, the TCGA-KIRC dataset was stratified into two distinct groups: a localized tumor cohort and a progression/metastasis cohort, based on tumor staging criteria. The efficacy of the prognostic prediction model was evaluated within each subgroup. A nomogram model was developed in conjunction with each independent prognostic factor to accurately predict patient outcomes. Additionally, single-cell and intercellular communication analyses were conducted via KIRC single-cell data obtained from the Gene Expression Omnibus (GEO) database. The effects of immunotherapy and targeted therapy on patients were predicted via prognostic modeling. A total of 260 angiogenesis-related genes were identified through screening in the GeneCards and Molecular Signatures Database(MSigDB). We subsequently developed a risk model comprising five genes: MEOX2, PLG, PROX1, TEK, and TIMP1. Survival analysis indicated that the prognosis for high-risk patients was significantly poorer than that for low-risk patients (P < 0.001), and the model demonstrated satisfactory accuracy in predicting 1-, 3-, and 5-year survival rates. This finding was further validated in both internal and external validation cohorts. The model demonstrated applicability for prognostic predictions in both the localized tumor cohort and the progression/metastasis cohort, with proficiency in forecasting the prognosis of patients diagnosed with metastatic renal cancer. The AUC values for 1, 3, and 5 years were recorded at 0.691, 0.709, and 0.773, respectively. We successfully constructed a nomogram model to facilitate accurate prognostic predictions for patients. Analysis of single-cell data revealed that PLG was expressed predominantly in tumor cell clusters, whereas TEK was highly expressed primarily in pericytes. TIMP1 was found to be highly expressed in vascular smooth muscle cells. In contrast, MEOX2 and PROX1 were highly expressed in specific cell clusters but presented low expression levels across the overall cell population. Cell communication analysis indicated that the modeling gene TEK was involved in the angiogenic pathway, with the interaction between the ligand ANGPT2 and the receptor ITGA5-ITGB1 being particularly prominent in this study. Furthermore, the immune dysfunction and rejection scores for high-risk patients within the non-localized renal cancer cohort were markedly elevated compared to those observed in the low-risk group. In terms of targeted pharmacological intervention, individuals classified in the low-risk group exhibited a heightened sensitivity to sorafenib. The KIRC prognostic prediction model, which is based on five angiogenesis-related genes, demonstrated reliable performance, indicating that high-risk patients have a significantly poorer prognosis than low-risk patients do. The developed nomogram model effectively visualizes and accurately predicts patient prognosis. It is essential to highlight that individuals diagnosed with low-risk metastatic KIRC may experience greater advantages from the administration of immunotherapy and sorafenib.
Collapse
Affiliation(s)
- Yuhe Zhou
- Department of Urology, Jinshan Branch of the Sixth People's Hospital of Shanghai, 147 Jiankang Road, Jinshan District, Shanghai, China
| | - Weixiong Ma
- Department of Urology, Jinshan Branch of the Sixth People's Hospital of Shanghai, 147 Jiankang Road, Jinshan District, Shanghai, China.
| | - Hengda Hu
- Department of Urology, Jinshan Branch of the Sixth People's Hospital of Shanghai, 147 Jiankang Road, Jinshan District, Shanghai, China
| | - Qirui He
- Department of Urology, Jinshan Branch of the Sixth People's Hospital of Shanghai, 147 Jiankang Road, Jinshan District, Shanghai, China
| | - Chengshuai Yu
- Department of Urology, Jinshan Branch of the Sixth People's Hospital of Shanghai, 147 Jiankang Road, Jinshan District, Shanghai, China
| | - Wenpu Chen
- Department of Urology, Jinshan Branch of the Sixth People's Hospital of Shanghai, 147 Jiankang Road, Jinshan District, Shanghai, China
| | - Guofeng Yu
- Department of Urology, Jinshan Branch of the Sixth People's Hospital of Shanghai, 147 Jiankang Road, Jinshan District, Shanghai, China
| |
Collapse
|
2
|
Wang H, Tang Y, Wang M, Zhao J, Ding C, Yang X, Han P, Liu P. Low expression of MEOX2 is associated with poor survival in patients with breast cancer. Biomark Med 2022; 16:1161-1170. [PMID: 36625258 DOI: 10.2217/bmm-2022-0468] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/11/2023] Open
Abstract
Aim: To investigate associations of MEOX2 expression with clinicopathological features and survival of breast cancer patients. Materials & methods: We used a breast cancer tissue microarray for immunohistochemistry. Associations between MEOX2 expression and clinicopathological features were analyzed using the χ-square test. Survival analysis was determined using a Kaplan-Meier curve. Multivariate Cox regression was used to determine associations of MEOX2 expression with overall survival. Results: We found that 74.1% of patients (100/135) had expression of MEOX2 at varying levels. MEOX2 was associated with histological grade and negatively correlated with Ki67 expression. Lower MEOX2 expression was significantly associated with decreased overall survival (p = 0.0011). Conclusion: MEOX2 expression could be a novel diagnostic and prognostic biomarker of breast cancer.
Collapse
Affiliation(s)
- Huxia Wang
- Center for Translational Medicine, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, 710061, China.,Department of Mammary, Shaanxi Provincial Cancer Hospital, Xi'an, 710061, China
| | - Yanan Tang
- Department of Vascular Surgery, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, 710061, China
| | - Meixia Wang
- Department of Health Examination, Shenmu Hospital, Yulin, 719300, China
| | - Jing Zhao
- Department of Mammary, Shaanxi Provincial Cancer Hospital, Xi'an, 710061, China
| | - Caixia Ding
- Department of Pathology, Shaanxi Provincial Cancer Hospital, Xi'an, 710061, China
| | - Xiaomin Yang
- Department of Mammary, Shaanxi Provincial Cancer Hospital, Xi'an, 710061, China
| | - Pihua Han
- Department of Mammary, Shaanxi Provincial Cancer Hospital, Xi'an, 710061, China
| | - Peijun Liu
- Center for Translational Medicine, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, 710061, China
| |
Collapse
|
3
|
Wang H, Tang Y, Yang X, Wang W, Han P, Zhao J, He S, Liu P. A Crucial Angiogenesis-Associated Gene MEOX2 Could Be a Promising Biomarker Candidate for Breast Cancer. Front Oncol 2022; 12:759300. [PMID: 35615155 PMCID: PMC9124839 DOI: 10.3389/fonc.2022.759300] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2021] [Accepted: 04/14/2022] [Indexed: 11/13/2022] Open
Abstract
BackgroundAngiogenesis plays a critical role in the growth and metastasis of breast cancer and angiogenesis inhibition has become an effective strategy for cancer therapy. Our study aimed to clarify the key candidate genes and pathways related to breast cancer angiogenesis.MethodsDifferentially expressed genes (DEGs) in the raw breast cancer (BRCA) gene dataset from the Cancer Genome Atlas (TCGA) database were identified and gene ontology analysis of the DEGs was performed. Hub genes were subsequently determined using the Gene Expression Omnibus database. The expression of the mesenchyme homeobox 2 (MEOX2) in breast cancer cells and tissues was assessed by quantification real-time polymerase chain reaction (qRT-PCR) and immunohistochemistry (IHC), respectively. The prognostic value of the MEOX2 gene in breast cancer tissue was evaluated with the Kaplan-Meier plotter.ResultsA total of 61 angiogenesis-related DEGs were identified in the TCGA dataset, among which the gene MEOX2 was significantly down-regulated. GO functional annotation and pathway enrichment analyses showed that MEOX2 was significantly enriched in the regulation of vasculature development. The IHC results confirmed that MEOX2 expression was repressed in breast cancer tissues and the relatively low level indicated the tissue was densely vascularized. Moreover, MEOX2 expression was significantly elevated in breast cancer cells after treatment with cisplatin (DDP) and epirubicin (EPI). Finally, the Kaplan-Meier plotter confirmed that higher expression levels of MEOX2 were related to better overall survival.ConclusionOur study revealed that the angiogenesis-associated gene MEOX2 can be used as a novel biomarker for breast cancer diagnosis and clinical therapy.
Collapse
Affiliation(s)
- Huxia Wang
- Center for Translational Medicine, The First Affiliated Hospital of Xi’an Jiaotong University, Xi’an, China
- Mammary Department, Shaanxi Provincial Cancer Hospital, Xi’an, China
| | - Yanan Tang
- Vascular Surgery Department, The First Affiliated Hospital of Xi’an Jiaotong University, Xi’an, China
| | - Xiaomin Yang
- Mammary Department, Shaanxi Provincial Cancer Hospital, Xi’an, China
| | - Weiyi Wang
- Vascular Surgery Department, The First Affiliated Hospital of Xi’an Jiaotong University, Xi’an, China
| | - Pihua Han
- Mammary Department, Shaanxi Provincial Cancer Hospital, Xi’an, China
| | - Jing Zhao
- Mammary Department, Shaanxi Provincial Cancer Hospital, Xi’an, China
| | - Sai He
- Mammary Department, Shaanxi Provincial Cancer Hospital, Xi’an, China
| | - Peijun Liu
- Center for Translational Medicine, The First Affiliated Hospital of Xi’an Jiaotong University, Xi’an, China
- *Correspondence: Peijun Liu,
| |
Collapse
|
4
|
Wang J, Chen Y, Wang Q, Xu H, Wu C, Jiang Q, Wu G, Zhou H, Xiao Z, Chen Y, Zhang T, Lan Q. MEOX2-mediated regulation of Cathepsin S promotes cell proliferation and motility in glioma. Cell Death Dis 2022; 13:360. [PMID: 35436995 PMCID: PMC9016080 DOI: 10.1038/s41419-022-04845-2] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2021] [Revised: 04/01/2022] [Accepted: 04/06/2022] [Indexed: 11/30/2022]
Abstract
Nuclear transcription factor Mesenchyme Homeobox 2 (MEOX2) is a homeobox gene that is originally discovered to suppress the growth of vascular smooth muscle and endothelial cells. However, whether or not it is connected to cancer is yet unknown. Here, we report that MEOX2 functions as a tumor-initiating element in glioma. Bioinformatic analyses of public databases and investigation of MEOX2 expression in patients with glioma demonstrated that MEOX2 was abundant at both mRNA and protein levels in glioma. MEOX2 expression was shown to be inversely linked with the prognosis of glioma patients. MEOX2 inhibition changed the morphology of glioma cells, inhibited cell proliferation and motility, whereas had no effect on cell apoptosis. Besides, silencing MEOX2 also hampered the epithelial-mesenchymal transition (EMT), focal adhesion formation, and F-actin assembly. Overexpression of MEOX2 exhibited opposite effects. Importantly, RNA-sequencing, ChIP-qPCR assay, and luciferase reporter assay revealed Cathepsin S (CTSS) as a novel transcriptional target of MEOX2 in glioma cells. Consistently, MEOX2 causes glioma tumor development in mice and greatly lowers the survival period of tumor-bearing mice. Our findings indicate that MEOX2 promotes tumorigenesis and progression of glioma partially through the regulation of CTSS. Targeting MEOX2-CTSS axis might be a promising alternative for the treatment of glioma.
Collapse
|
5
|
Cardinale A, Cantalupo S, Lasorsa VA, Montella A, Cimmino F, Succoio M, Vermeulen M, Baltissen MP, Esposito M, Avitabile M, Formicola D, Testori A, Bonfiglio F, Ghiorzo P, Scalvenzi M, Ayala F, Zambrano N, Iles MM, Xu M, Law MH, Brown KM, Iolascon A, Capasso M. Functional annotation and investigation of the 10q24.33 melanoma risk locus identifies a common variant that influences transcriptional regulation of OBFC1. Hum Mol Genet 2022; 31:863-874. [PMID: 34605909 PMCID: PMC9077268 DOI: 10.1093/hmg/ddab293] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2021] [Revised: 09/07/2021] [Accepted: 09/29/2021] [Indexed: 12/15/2022] Open
Abstract
The 10q24.33 locus is known to be associated with susceptibility to cutaneous malignant melanoma (CMM), but the mechanisms underlying this association have been not extensively investigated. We carried out an integrative genomic analysis of 10q24.33 using epigenomic annotations and in vitro reporter gene assays to identify regulatory variants. We found two putative functional single nucleotide polymorphisms (SNPs) in an enhancer and in the promoter of OBFC1, respectively, in neural crest and CMM cells, one, rs2995264, altering enhancer activity. The minor allele G of rs2995264 correlated with lower OBFC1 expression in 470 CMM tumors and was confirmed to increase the CMM risk in a cohort of 484 CMM cases and 1801 controls of Italian origin. Hi-C and chromosome conformation capture (3C) experiments showed the interaction between the enhancer-SNP region and the promoter of OBFC1 and an isogenic model characterized by CRISPR-Cas9 deletion of the enhancer-SNP region confirmed the potential regulatory effect of rs2995264 on OBFC1 transcription. Moreover, the presence of G-rs2995264 risk allele reduced the binding affinity of the transcription factor MEOX2. Biologic investigations showed significant cell viability upon depletion of OBFC1, specifically in CMM cells that were homozygous for the protective allele. Clinically, high levels of OBFC1 expression associated with histologically favorable CMM tumors. Finally, preliminary results suggested the potential effect of decreased OBFC1 expression on telomerase activity in tumorigenic conditions. Our results support the hypothesis that reduced expression of OBFC1 gene through functional heritable DNA variation can contribute to malignant transformation of normal melanocytes.
Collapse
Affiliation(s)
- Antonella Cardinale
- Dipartimento di Medicina Molecolare e Biotecnologie Mediche, Università degli Studi di Napoli Federico II, Naples 80136, Italy
- CEINGE Biotecnologie Avanzate, Naples 80145, Italy
- Department of Pediatric Hematology and Oncology, Bambino Gesù Children's Hospital, IRCCS, Rome, Italy
| | - Sueva Cantalupo
- Dipartimento di Medicina Molecolare e Biotecnologie Mediche, Università degli Studi di Napoli Federico II, Naples 80136, Italy
- CEINGE Biotecnologie Avanzate, Naples 80145, Italy
| | - Vito Alessandro Lasorsa
- Dipartimento di Medicina Molecolare e Biotecnologie Mediche, Università degli Studi di Napoli Federico II, Naples 80136, Italy
- CEINGE Biotecnologie Avanzate, Naples 80145, Italy
| | - Annalaura Montella
- Dipartimento di Medicina Molecolare e Biotecnologie Mediche, Università degli Studi di Napoli Federico II, Naples 80136, Italy
- CEINGE Biotecnologie Avanzate, Naples 80145, Italy
| | | | | | - Michiel Vermeulen
- Department of Molecular Biology, Radboud Institute for Molecular Life Sciences, Oncode Institute, Radboud University, Nijmegen, the Netherlands
| | - Marijke P Baltissen
- Department of Molecular Biology, Radboud Institute for Molecular Life Sciences, Oncode Institute, Radboud University, Nijmegen, the Netherlands
| | - Matteo Esposito
- Dipartimento di Medicina Molecolare e Biotecnologie Mediche, Università degli Studi di Napoli Federico II, Naples 80136, Italy
| | - Marianna Avitabile
- Dipartimento di Medicina Molecolare e Biotecnologie Mediche, Università degli Studi di Napoli Federico II, Naples 80136, Italy
- CEINGE Biotecnologie Avanzate, Naples 80145, Italy
| | - Daniela Formicola
- Dipartimento di Medicina Molecolare e Biotecnologie Mediche, Università degli Studi di Napoli Federico II, Naples 80136, Italy
- CEINGE Biotecnologie Avanzate, Naples 80145, Italy
- SOC Genetica Medica, Azienda Ospedaliera Universitaria Meyer, Firenze 50139, Italy
| | - Alessandro Testori
- Dipartimento di Medicina Molecolare e Biotecnologie Mediche, Università degli Studi di Napoli Federico II, Naples 80136, Italy
| | - Ferdinando Bonfiglio
- CEINGE Biotecnologie Avanzate, Naples 80145, Italy
- Dipartimento di Ingegneria chimica, dei Materiali e della Produzione industriale, Università degli Studi di Napoli Federico II, Napoli, Italy
| | - Paola Ghiorzo
- Genetica dei Rumori Rari, IRCCS Ospedale Policlinico San Martino, Genoa, Italy
- Dipartimento di Medicina Interna e Specialità Mediche, Università degli Studi di Genova, Genova, Italy
| | - Massimiliano Scalvenzi
- Dipartimento di Medicina clinica e Chirurgia, Università degli Studi di Napoli Federico II, Naples 80136, Italy
| | - Fabrizio Ayala
- Department of Melanoma and Cancer Immunotherapy, Istituto Nazionale Tumori IRCCS Fondazione Pascale, Napoli, Italy
| | - Nicola Zambrano
- Dipartimento di Medicina Molecolare e Biotecnologie Mediche, Università degli Studi di Napoli Federico II, Naples 80136, Italy
- CEINGE Biotecnologie Avanzate, Naples 80145, Italy
| | - Mark M Iles
- Section of Epidemiology and Biostatistics, Leeds Institute of Cancer and Pathology, University of Leeds, Leeds, UK
| | - Mai Xu
- Laboratory of Translational Genomics, Division of Cancer Epidemiology and Genetics, National Cancer Institute, National Institutes of Health, Bethesda, MD 20892, USA
| | - Matthew H Law
- Statistical Genetics, QIMR Berghofer Medical Research Institute Brisbane, Queensland 4006, Australia
- School of Biomedical Sciences, Faculty of Health, Queensland University of Technology, Brisbane, Queensland, Australia
| | - Kevin M Brown
- Laboratory of Translational Genomics, Division of Cancer Epidemiology and Genetics, National Cancer Institute, National Institutes of Health, Bethesda, MD 20892, USA
| | - Achille Iolascon
- Dipartimento di Medicina Molecolare e Biotecnologie Mediche, Università degli Studi di Napoli Federico II, Naples 80136, Italy
- CEINGE Biotecnologie Avanzate, Naples 80145, Italy
| | - Mario Capasso
- Dipartimento di Medicina Molecolare e Biotecnologie Mediche, Università degli Studi di Napoli Federico II, Naples 80136, Italy
- CEINGE Biotecnologie Avanzate, Naples 80145, Italy
| |
Collapse
|
6
|
Cai Y, Liu Y, Sun Y, Ren Y. Mesenchyme homeobox 2 has a cancer-inhibiting function in breast carcinoma via affection of the PI3K/AKT/mTOR and ERK1/2 pathways. Biochem Biophys Res Commun 2022; 593:20-27. [DOI: 10.1016/j.bbrc.2022.01.011] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2022] [Accepted: 01/05/2022] [Indexed: 12/22/2022]
|
7
|
Peralta-Arrieta I, Trejo-Villegas OA, Armas-López L, Ceja-Rangel HA, Ordóñez-Luna MDC, Pineda-Villegas P, González-López MA, Ortiz-Quintero B, Mendoza-Milla C, Zatarain-Barrón ZL, Arrieta O, Zúñiga J, Ávila-Moreno F. Failure to EGFR-TKI-based therapy and tumoural progression are promoted by MEOX2/GLI1-mediated epigenetic regulation of EGFR in the human lung cancer. Eur J Cancer 2021; 160:189-205. [PMID: 34844838 DOI: 10.1016/j.ejca.2021.10.032] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2021] [Accepted: 10/26/2021] [Indexed: 01/22/2023]
Abstract
BACKGROUND Mesenchyme homeobox-2 (MEOX2)-mediated regulation of glioma-associated oncogene-1 (GLI1) has been associated with poor overall survival, conferring chemoresistance in lung cancer. However, the role of MEOX2/GLI1 in resistance to epidermal growth factor receptor-tyrosine kinase inhibitors (EGFR-TKIs)-based therapy remains unexplored in human lung cancer. METHODS Functional assays using genetic silencing strategy by short hairpin RNAs, as well as cytotoxic (tetrazolium dye MTT) and clonogenic assays, were performed to evaluate MEOX2/GLI1-induced malignancy capacity in lung cancer cells. Further analysis performed includes western blot, qPCR and ChIP-qPCR assays to identify whether MEOX2/GLI1 promote EGFR/AKT/ERK activation, as well as EGFR overexpression through epigenetic mechanisms. Finally, preclinical tumour progression in vivo and progression-free disease interval analyses in patients treated with EGFR-TKI were included. RESULTS Overexpressed MEOX2/GLI1 in both EGFR wild-type and EGFR/KRAS-mutated lung cancer cells were detected and involved in the activation/expression of EGFR/AKT/ERK biomarkers. In addition, MEOX2/GLI1 was shown to be involved in the increased proliferation of tumour cells and resistance capacity to cisplatin, EGFR-TKIs (erlotinib and AZD9291 'osimertinib'), AZD8542-SMO, and AZD6244-MEKK1/2. In addition, we identified that MEOX2/GLI1 promote lung tumour cells progression in vivo and are clinically associated with poorer progression-free disease intervals. Finally, both MEOX2 and GLI1 were detected to be epigenetically involved in EGFR expression by reducing both repressive markers polycomb-EZH2 and histone H3K27me3, but, particularly, increasing an activated histone profile H3K27Ac/H3K4me3 at EGFR-gene enhancer-promoter sequences that probably representing a novel EGFR-TKI-based therapy resistance mechanism. CONCLUSION MEOX2/GLI1 promote resistance to cisplatin and EGFR-TKI-based therapy in lung cancer cells, modulating EGFR/AKT/ERK signalling pathway activation, as well as inducing an aberrant epigenetic modulation of the EGFR-gene expression in human lung cancer.
Collapse
Affiliation(s)
- Irlanda Peralta-Arrieta
- Universidad Nacional Autónoma de México (UNAM), Unidad de Investigación en Biomedicina (UBIMED), Facultad de Estudios Superiores (FES) Iztacala, Tlalnepantla de Baz, 54090, Estado de México, Mexico.
| | - Octavio A Trejo-Villegas
- Universidad Nacional Autónoma de México (UNAM), Unidad de Investigación en Biomedicina (UBIMED), Facultad de Estudios Superiores (FES) Iztacala, Tlalnepantla de Baz, 54090, Estado de México, Mexico.
| | - Leonel Armas-López
- Universidad Nacional Autónoma de México (UNAM), Unidad de Investigación en Biomedicina (UBIMED), Facultad de Estudios Superiores (FES) Iztacala, Tlalnepantla de Baz, 54090, Estado de México, Mexico.
| | - Hugo A Ceja-Rangel
- Universidad Nacional Autónoma de México (UNAM), Unidad de Investigación en Biomedicina (UBIMED), Facultad de Estudios Superiores (FES) Iztacala, Tlalnepantla de Baz, 54090, Estado de México, Mexico.
| | - María Del Carmen Ordóñez-Luna
- Universidad Nacional Autónoma de México (UNAM), Unidad de Investigación en Biomedicina (UBIMED), Facultad de Estudios Superiores (FES) Iztacala, Tlalnepantla de Baz, 54090, Estado de México, Mexico.
| | - Priscila Pineda-Villegas
- Universidad Nacional Autónoma de México (UNAM), Unidad de Investigación en Biomedicina (UBIMED), Facultad de Estudios Superiores (FES) Iztacala, Tlalnepantla de Baz, 54090, Estado de México, Mexico.
| | - Marco A González-López
- Unidad Funcional de Oncología Torácica, Instituto Nacional de Cancerología (INCan), Av. San Fernando 22, Belisario Domínguez Secc 16, Tlalpan, 14080, Ciudad de México, Mexico.
| | - Blanca Ortiz-Quintero
- Instituto Nacional de Enfermedades Respiratorias Ismael Cosío Villegas, Calz de Tlalpan, 14080, Ciudad de México, Mexico.
| | - Criselda Mendoza-Milla
- Instituto Nacional de Enfermedades Respiratorias Ismael Cosío Villegas, Calz de Tlalpan, 14080, Ciudad de México, Mexico.
| | - Zyanya L Zatarain-Barrón
- Tecnologico de Monterrey, Escuela de Medicina y Ciencias de la Salud, Campus Ciudad de México, Mexico.
| | - Oscar Arrieta
- Unidad Funcional de Oncología Torácica, Instituto Nacional de Cancerología (INCan), Av. San Fernando 22, Belisario Domínguez Secc 16, Tlalpan, 14080, Ciudad de México, Mexico.
| | - Joaquín Zúñiga
- Instituto Nacional de Enfermedades Respiratorias Ismael Cosío Villegas, Calz de Tlalpan, 14080, Ciudad de México, Mexico; Tecnologico de Monterrey, Escuela de Medicina y Ciencias de la Salud, Campus Ciudad de México, Mexico.
| | - Federico Ávila-Moreno
- Universidad Nacional Autónoma de México (UNAM), Unidad de Investigación en Biomedicina (UBIMED), Facultad de Estudios Superiores (FES) Iztacala, Tlalnepantla de Baz, 54090, Estado de México, Mexico; Instituto Nacional de Enfermedades Respiratorias Ismael Cosío Villegas, Calz de Tlalpan, 14080, Ciudad de México, Mexico.
| |
Collapse
|
8
|
Tachon G, Masliantsev K, Rivet P, Petropoulos C, Godet J, Milin S, Wager M, Guichet PO, Karayan-Tapon L. Prognostic significance of MEOX2 in gliomas. Mod Pathol 2019; 32:774-786. [PMID: 30659268 DOI: 10.1038/s41379-018-0192-6] [Citation(s) in RCA: 30] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2018] [Revised: 11/20/2018] [Accepted: 11/21/2018] [Indexed: 01/08/2023]
Abstract
Gliomas are the most common malignant primary tumors in the central nervous system and have variable predictive clinical courses. Glioblastoma, the most aggressive form of glioma, is a complex disease with unsatisfactory therapeutic solutions and a very poor prognosis. Some processes at stake in gliomagenesis have been discovered but little is known about the role of homeobox genes, even though they are highly expressed in gliomas, particularly in glioblastoma. Among them, the transcription factor Mesenchyme Homeobox 2 (MEOX2) had previously been associated with malignant progression and clinical prognosis in lung cancer and hepatocarcinoma but never studied in glioma. The aim of our study was to investigate the clinical significance of MEOX2 in gliomas. We assessed the expression of MEOX2 according to IDH1/2 molecular profile and patient survival among three different public datasets: The Cancer Genome Atlas (TCGA), The Chinese Glioma Genome Atlas (CGGA) and the US National Cancer Institute Repository for Molecular Brain Neoplasia Data (Rembrandt). We then evaluated the prognostic significance of MEOX2 protein expression on 112 glioma clinical samples including; 56 IDH1 wildtype glioblastomas, 7 IDH1 wild-type lower grade gliomas, 49 IDH1 mutated lower grade gliomas. Survival rates were estimated by the Kaplan-Meier method followed by uni/multivariate analyses. We demonstrated that MEOX2 was one of the transcription factors most closely associated with overall survival in glioma. Moreover, MEOX2 expression was associated with IDH1/2 wildtype molecular subtype and was significantly correlated with overall survival of all gliomas and, more interestingly, in lower grade glioma. To conclude, our results may be the first to provide insight into the clinical significance of MEOX2 in gliomas, which is a factor closely related to patient outcome. MEOX2 could constitute an interesting prognostic biomarker, especially for lower grade glioma.
Collapse
Affiliation(s)
- Gaelle Tachon
- Inserm U1084, Laboratoire de Neurosciences Expérimentales et Cliniques, Poitiers, F-86073, France.,Université de Poitiers, F-86073, Poitiers, France.,CHU de Poitiers, Laboratoire de Cancérologie Biologique, Poitiers, F-86022, France
| | - Konstantin Masliantsev
- Inserm U1084, Laboratoire de Neurosciences Expérimentales et Cliniques, Poitiers, F-86073, France.,Université de Poitiers, F-86073, Poitiers, France.,CHU de Poitiers, Laboratoire de Cancérologie Biologique, Poitiers, F-86022, France
| | - Pierre Rivet
- CHU de Poitiers, Laboratoire de Cancérologie Biologique, Poitiers, F-86022, France
| | - Christos Petropoulos
- Inserm U1084, Laboratoire de Neurosciences Expérimentales et Cliniques, Poitiers, F-86073, France.,Université de Poitiers, F-86073, Poitiers, France.,CHU de Poitiers, Laboratoire de Cancérologie Biologique, Poitiers, F-86022, France
| | - Julie Godet
- CHU de Poitiers, Service d'Anatomo-Cytopathologie, Poitiers, F-86021, France
| | - Serge Milin
- CHU de Poitiers, Service d'Anatomo-Cytopathologie, Poitiers, F-86021, France
| | - Michel Wager
- Inserm U1084, Laboratoire de Neurosciences Expérimentales et Cliniques, Poitiers, F-86073, France.,Université de Poitiers, F-86073, Poitiers, France.,CHU de Poitiers, Service de Neurochirurgie, Poitiers, F-86021, France
| | - Pierre-Olivier Guichet
- Inserm U1084, Laboratoire de Neurosciences Expérimentales et Cliniques, Poitiers, F-86073, France. .,Université de Poitiers, F-86073, Poitiers, France. .,CHU de Poitiers, Laboratoire de Cancérologie Biologique, Poitiers, F-86022, France.
| | - Lucie Karayan-Tapon
- Inserm U1084, Laboratoire de Neurosciences Expérimentales et Cliniques, Poitiers, F-86073, France. .,Université de Poitiers, F-86073, Poitiers, France. .,CHU de Poitiers, Laboratoire de Cancérologie Biologique, Poitiers, F-86022, France.
| |
Collapse
|
9
|
An Z, Wang D, Yang G, Zhang WQ, Ren J, Fu JL. Role of microRNA-130a in the pathogeneses of obstructive sleep apnea hypopnea syndrome-associated pulmonary hypertension by targeting the GAX gene. Medicine (Baltimore) 2017; 96:e6746. [PMID: 28514291 PMCID: PMC5440128 DOI: 10.1097/md.0000000000006746] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/25/2022] Open
Abstract
OBJECTIVE The purpose of this study was to elucidate the role of microRNA-130a (miR-130a) in obstructive sleep apnea hypopnea syndrome (OSAHS)-associated pulmonary hypertension (PHT) by targeting the growth arrest-specific homeobox (GAX) gene. METHODS A total of 108 patients with OSAHS-associated PHT were recruited as the OSAHS-associated PHT group and 110 healthy individuals were randomly selected as the normal control group. Human umbilical vein endothelial cells (HUVECs) were selected and divided into the control, miR-130a mimic, mimic negative control (NC), miR-130a inhibitor, and inhibitor-NC groups. The dual luciferase reporter gene assay was used to identify the relationship between miR-130a and the GAX gene. The quantitative real-time polymerase chain reaction (qRT-PCR) and Western blotting were applied for the relative expressions of miR-130a and the mRNA and protein expressions of GAX. Serum levels of endothelin-1 (ET-1), vascular endothelial growth factor (VEGF), nitric oxide (NO), and super oxide dismutase (SOD) were detected. Cell apoptosis and angiogenic activity were analyzed by flow cytometry and cell tube formation assay. RESULTS GAX was a target gene of miR-130a. Compared with the normal control group, the relative expression of miR-130a and the serum levels of ET-1 and VEGF were increased, whereas the mRNA expression of GAX and the serum levels of NO and SOD were decreased in the OSAHS-associated PHT group. Compared with the control, mimic-NC, and inhibitor-NC groups, the relative expressions of miR-130a in the miR-130a mimic group were enhanced, whereas the expression of miR-130a in the miR-130a inhibitor group was reduced. However, the mRNA and protein expressions of GAX showed an opposite trend in the miR-130a mimic and miR-130a inhibitor groups. In comparison to the control, mimic-NC, and inhibitor-NC groups, the miR-130a mimic group had an increase of ET-1 and VEGF expressions, whereas the expressions of NO and SOD were reduced. However, the miR-130a inhibitor group exhibited an opposite trend. The apoptosis rate and tube formation number in the miR-130a mimic group were obviously increased, whereas the miR-130a inhibitor group showed an obvious decrease. CONCLUSION These data provided strong evidence that miR-130a may be involved in the progression of OSAHS-associated PHT by down-regulating GAX gene.
Collapse
Affiliation(s)
- Zhe An
- Department of Cardiology, China-Japan Union Hospital of Jilin University
| | - Dan Wang
- Department of Breast Surgery, The Second Hospital of Jilin University
| | - Guang Yang
- Department of Molecular Biology, College of Basic Medical Sciences, Jilin University
| | - Wen-Qi Zhang
- Department of Cardiology, China-Japan Union Hospital of Jilin University
| | - Jin Ren
- Department of Respiratory Medicine, the Second Hospital of Jilin University
| | - Jin-Ling Fu
- Department of Ophthalmology, The Second Hospital of Jilin University, Changchun, Jilin, P.R. China
| |
Collapse
|