Elsevier

Cellular Signalling

Volume 79, March 2021, 109882
Cellular Signalling

CAMTA1, a novel antitumor gene, regulates proliferation and the cell cycle in glioma by inhibiting AKT phosphorylation

https://doi.org/10.1016/j.cellsig.2020.109882Get rights and content

Highlights

  • CAMTA1 expression is downregulated within glioma.

  • CAMTA1 expression is negatively correlated with ITGA5/ITGB1.

  • CAMTA1 regulates glioma cell proliferation, migration, invasion and cell cycle via ITGA5/ITGB1 and AKT signaling.

Abstract

Identifying biomarkers for the early diagnosis of glioma and elucidating the molecular mechanisms underlying the development of this cancer are of considerable clinical importance. Recently, studies performing microarray profiling of genes to identify distinct gene signatures reported specific subtypes with predictive and prognostic relevance. Thus, we performed deep sequencing on a total of 26 glioma tissue samples to identify the frequently mutated of oncogenes and tumor suppressors in gliomas. A total of 2306 single-nucleotide polymorphisms (SNPs) and 2010 insertion and deletion sites (indels) were found by aligning sequencing information from 26 glioma samples with sequences from the normal human gene database (GRCh37/hg19). GSEA results suggest that an underexpressed gene, calmodulin binding transcription activator 1 (CAMTA1), participates in the cell proliferation and cell cycle regulation of glioma cells. Moreover, overexpression of CAMTA1 in glioma cells notably inhibited cell growth, migration, invasion and cell cycle and enhanced temozolomide (TMZ)-induced cell apoptosis in glioma cells, while CAMTA1 overexpression decreased the ITGA5, ITGB1, p-AKT, p-FAK, and Myc protein levels, suggesting that the signaling pathways of these proteins might be involved in the cellular functions of CAMTA1 in glioma. Moreover, overexpression of CAMTA1 attenuated the growth and tumorigenesis of glioma in vivo. In summary, we identified high-frequency mutant genes in glioma and provided an experimental basis for a novel mechanism by which CAMTA1 may serve as a tumor suppressor in glioma.

Introduction

Gliomas are brain tumors generated by glial cells and are the most commonly diagnosed primary tumors of the central nervous system (accounting for approximately 30% of these tumors). Gliomas have a high mortality rate, which is close to 80% within one year after diagnosis. Although the metastatic potential of gliomas is limited, one of their features is a higher death rate, which is primarily attributable to high rates of local infiltration and angiogenesis with immunosuppression. In addition, delayed diagnosis of gliomas leads to advanced/graded tumors and poor prognosis, while high-grade gliomas are often associated with extensive blood vessel invasion, massive loss of oxygen and neck inflammation [1]. Histologically, gliomas can be divided into glioblastomas, astrocytomas, and oligodendrogliomas. Moreover, the World Health Organization (WHO) introduced a four-tiered grading system in 2002 that identified four classes of malignancies: low-grade (WHO grade I–II), anaplastic (grade III) and glioblastoma multiform (grade IV) [2]. The most recent WHO Classification of Tumors of the Central Nervous System was proposed in 2016; this classification not only integrated the morphological characteristics, growing patterns and molecular features of tumor cells [3] but also defined the degree of malignancy with sufficient prognosis to play a critical role in determining the clinical settings and treatment programs [4]. Recently, studies performing on gene expression profiling to identify distinct gene signatures reported specific subtypes with predictive and prognostic relevance [[5], [6], [7], [8]]. However, due to the risks of radiation therapy and the limited effectiveness of surgery, there is an urgent need for new options for the treatment of glioma. Identifying early diagnostic biomarkers of glioma and elucidating the molecular mechanisms underlying the development of this cancer are of considerable clinical importance.

To explore the mechanisms underlying tumor initiation, progression and metastasis and to identify novel diagnostic biomarkers and new targeted therapies for glioma, in the past several years, studies have focused on gene alterations related to glioma and the signaling pathway deregulation. Recently, The Cancer Genome Atlas (TCGA) (http://cancergenome.nih.gov) analyses of gliomas have utilized a variety of technology platforms, such as mutation arrays, copy number arrays, expression arrays and methylation arrays [5,6,9]. Expression array date have revealed that molecular subtypes are related to grade and outcome, suggesting that expression profiles are more predictive of outcome, than are histological subtypes [5,6,9]. For example, CD44 is overexpressed in glioma and is involved in many important cell functions that contribute to tumorigenesis [10,11]. Cyclin dependent kinase 1 (CDK1) and cadherin 1 (CDH1) play important roles in the protein-protein interaction (PPI) network according to Gene Ontology functional and KEGG pathway enrichment analyses based on the GSE31262 dataset [12]. Many other glioma-related genes such as FN1 [13], TGF-β [14], SPP1 and EGFR [15] have also been studied and their potential applications as targets for the diagnosis and therapy of glioma have also been evaluated.

There is increasing evidence that the risk and prognosis of gliomas may be influenced by the genetic backgrounds of patients. As single nucleotide polymorphisms (SNPs) are the most commonly seen genetic variation, they may be useful alternative biomarkers of patients' genetic backgrounds and may be employed to predict treatment response and prognosis. Barbashina et al. reported that the loss of combined 1p/19q was strongly correlated with the histology of typical oligodendroglioma and determined that a small segment of 1p36 in calmodulin binding transcription activator 1 (CAMTA1) was deleted within all oligodendroglial tumors with loss of heterozygosity of 1p, which could overlap with the minimal deleted region of the neuroblastoma 1p36 [16]. More importantly, CAMTA1 is a novel tumor suppressor in glioblastoma, and colony formation is significantly decreased by the overexpression of CAMTA1. Consistent with the finding, CAMTA1 expression correlates with glioblastoma patient survival [17]. Nevertheless, the biological function CAMTA1 in glioma still unclear and need further clarification.

In the present study, we performed deep sequencing on a total of 26 glioma tissue samples to identify the frequently occurring mutation of oncogenes and tumor suppressors in gliomas. The SNPs and indels that were detected were analyzed and compared to the normal human genetic database (hg19) to identify the high-frequency mutant genes. Based on the essential role played by CAMTA1 in glioma, we subsequently analyzed the correlation between the gene polymorphism and the expression of CAMTA1. The expression of CAMTA1 in glioma was analyzed based on the Chinese Glioma Genome Atlas (CGGA) and TCGA and examined in tissue samples using real-time PCR and immunoblotting. Next, gene set enrichment analysis (GSEA) was performed to identify signaling pathways related to CAMTA1. The correlation between the expression of CAMTA1 and related factors and the cellular function of CAMTA1 in glioma cells and relevant pathways were examined. In summary, we identified frequently occurring mutant genes in glioma and provided an experimental basis for a novel mechanism by which CAMTA1 may serve as a tumor suppressor in glioma.

Section snippets

Clinical tissue samples

The study was approved by the appropriate ethics review committees. Appropriate informed consent forms were completed by all subjects. A total of 26 glioma and peritumoral brain tissues were obtained from the glioma patients who underwent surgery in the Department of Neurosurgery of the People's Hospital of Sichuan Province between October 2016 and July 2018. The inclusion criteria for the case group are as follows: 1. Patients with pathological diagnosis of glioma during the initial diagnosis

High-frequency mutation of oncogenes and tumor suppressors in gliomas including CAMTA1

A total of 26 tissue samples were collected, and the sequencing information of 26 samples was obtained. According to the sequencing analysis, we observed a large number of mutations in the glioma cells of the Han population. A total of 2306 single nucleotide polymorphisms (SNPs) were identified by aligning the sequencing information from 26 cases with sequences from the normal human gene database (GRCh37/hg19). Among these SNPs, the high-frequency mutant genes with mutation sites ≥ 30 include

Discussion

To identify differentially expressed genes, high-frequency mutant genes, and related signaling pathways in gliomas, we performed deep sequencing on a total of 26 glioma samples. A total of 2306 single nucleotide polymorphisms (SNPs) and 2010 insertion and deletion sites (indels) were identified by aligning sequencing information from 26 glioma cases with sequences from the normal human gene database (GRCh37/hg19). High-frequency mutated genes (mutations sites≥ 30) include previously reported

Conclusion

The results of this study demonstrate that CAMTA1 is a novel antitumor gene in gliomas and that CAMTA1 regulates proliferation, cell apoptosis, invasion and cell cycle in gliomas by inhibiting AKT phosphorylation via the ITGA1/ITGB5 pathway.

Disclosure statement

None.

Funding

This study was supported by the Department of Science and Technology of Sichuan Province, China (No. 2017SZ0203), Foundation for Technology and Science Bureau of Chengdu, China (No. 2018-YF05-00336-SN), Department of Science and Technology of Sichuan Province, China (No. 2019YFS0399) and Health and Family Planning Commission of Sichuan Province, China (No. 18PJ426).

Ethical approval and consent to participate

All procedures performed in studies involving human participants were in accordance with the ethical standards of People's Hospital of Sichuan Province and with the 1964 Helsinki declaration. Informed consent to participate in the study has been obtained from participants.

Consent for publication

Consent for publication was obtained from the participants.

Data availability statements

All the available data were presented in the study.

Declaration of Competing Interest

None.

Acknowledgment

Not applicable.

References (61)

  • P. Fiorilli et al.

    Integrins mediate adhesion of medulloblastoma cells to tenascin and activate pathways associated with survival and proliferation

    Lab. Investig.

    (2008)
  • S. Kesari

    Understanding glioblastoma tumor biology: the potential to improve current diagnosis and treatments

    Semin. Oncol.

    (2011)
  • P. Kleihues et al.

    The WHO classification of tumors of the nervous system

    J. Neuropathol. Exp. Neurol.

    (2002)
  • B.S. Kim et al.

    Clinical outcomes of intracranial solitary fibrous tumor and hemangiopericytoma: analysis according to the 2016 WHO classification of central nervous system tumors

    J. Neurosurg.

    (2018)
  • D.N. Louis et al.

    The 2007 WHO classification of tumours of the central nervous system

    Acta Neuropathol.

    (2007)
  • L.A. Gravendeel et al.

    Intrinsic gene expression profiles of gliomas are a better predictor of survival than histology

    Cancer Res.

    (2009)
  • A. Li et al.

    Unsupervised analysis of transcriptomic profiles reveals six glioma subtypes

    Cancer Res.

    (2009)
  • Cancer Genome Atlas Research N

    Comprehensive genomic characterization defines human glioblastoma genes and core pathways

    Nature.

    (2008)
  • C. Xie et al.

    Candidate genes and microRNAs for glioma pathogenesis and prognosis based on gene expression profiles

    Mol. Med. Rep.

    (2018)
  • T.C. Ramos et al.

    Treatment of high-grade glioma patients with the humanized anti-epidermal growth factor receptor (EGFR) antibody h-R3: report from a phase I/II trial

    Cancer Biol Ther.

    (2006)
  • T. Nickl-Jockschat et al.

    An imbalance between Smad and MAPK pathways is responsible for TGF-beta tumor promoting effects in high-grade gliomas

    Int. J. Oncol.

    (2007)
  • V. Barbashina et al.

    Allelic losses at 1p36 and 19q13 in gliomas: correlation with histologic classification, definition of a 150-kb minimal deleted region on 1p36, and evaluation of CAMTA1 as a candidate tumor suppressor gene

    Clin. Cancer Res.

    (2005)
  • D. Schraivogel et al.

    CAMTA1 is a novel tumour suppressor regulated by miR-9/9* in glioblastoma stem cells

    EMBO J.

    (2011)
  • X. Jin et al.

    CASC2/miR-24/miR-221 modulates the TRAIL resistance of hepatocellular carcinoma cell through caspase-8/caspase-3

    Cell Death Dis.

    (2018)
  • H. He et al.

    Silencing of MED27 inhibits adrenal cortical carcinogenesis by targeting the Wnt/beta-catenin signaling pathway and the epithelial-mesenchymal transition process

    Biol. Chem.

    (2018)
  • L.A. Edwards et al.

    Effect of brain- and tumor-derived connective tissue growth factor on glioma invasion

    J. Natl. Cancer Inst.

    (2011)
  • V. Milinkovic et al.

    Identification of novel genetic alterations in samples of malignant glioma patients

    PLoS One

    (2013)
  • L. Feng et al.

    miR-330-5p suppresses glioblastoma cell proliferation and invasiveness through targeting ITGA5

    Biosci. Rep.

    (2017)
  • S. Choi et al.

    Temozolomide-associated hypermutation in gliomas

    Neuro-Oncology

    (2018)
  • A. Karachi et al.

    Temozolomide for immunomodulation in the treatment of glioblastoma

    Neuro-Oncology

    (2018)
  • Cited by (12)

    • Narciclasine suppresses esophageal cancer cell proliferation and migration by inhibiting the FAK signaling pathway

      2022, European Journal of Pharmacology
      Citation Excerpt :

      Narciclasine dose-dependently downregulated the expression of alpha-tubulin (Fig. 5D–F). Various studies have indicated that the downregulation of FAK and cell cycle arrest can promote apoptosis (Fan et al., 2020; He et al., 2021; Kassab and Hassan, 2018). Therefore, flow cytometric analysis was performed to examine the effect of narciclasine on apoptosis.

    • LncRNA RBPMS-AS1 promotes NRGN transcription to enhance the radiosensitivity of glioblastoma through the microRNA-301a-3p/CAMTA1 axis

      2022, Translational Oncology
      Citation Excerpt :

      The Genecards website shows that CAMTA1 contains a transcription factor immunoglobulin domain, ankyrin repeats, as well as calmodulin-binding IQ motifs. CAMTA1 acts as a tumor inhititory gene in gliomas [21], and there is a report revealing that CAMTA1 is acted as a transcription factor to activate the transcription of cardiac hormone sodium A (NPPA), and induces anti-proliferation effects in glioma cells [24]. In our research, we performed RT-qPCR to detect the CAMTA1 expression level in GBM tissues and the corresponding normal tissues, which revealed that decreased CAMTA1 expression was found in GBM tissues versus normal tissues (Fig. 2A).

    View all citing articles on Scopus
    1

    The authors contribute equally to this study.

    View full text