Next Article in Journal
The Expression and Functionality of CB1R-NMDAR Complexes Are Decreased in A Parkinson’s Disease Model
Next Article in Special Issue
Bone Morphogenic Proteins in Pediatric Diffuse Midline Gliomas: How to Make New Out of Old?
Previous Article in Journal
RETRACTED: Wang et al. Panoramic Manifold Projection (Panoramap) for Single-Cell Data Dimensionality Reduction and Visualization. Int. J. Mol. Sci. 2022, 23, 7775
Previous Article in Special Issue
Identification of Metabolomic Markers in Frozen or Formalin-Fixed and Paraffin-Embedded Samples of Diffuse Glioma from Adults
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Communication

Gene Expression of CSF3R/CD114 Is Associated with Poorer Patient Survival in Glioma

by
Samir Ale Bark
1,2,
Matheus Dalmolin
3,4,
Osvaldo Malafaia
1,
Rafael Roesler
5,6,7,*,
Marcelo A. C. Fernandes
3,4,8 and
Gustavo R. Isolan
1,2,7,*
1
Graduate Program in Principles of Surgery, Mackenzie Evangelical University, Curitiba 80730-000, PR, Brazil
2
The Center for Advanced Neurology and Neurosurgery (CEANNE), Porto Alegre 90560-010, RS, Brazil
3
InovAI Lab, nPITI/IMD, Federal University of Rio Grande do Norte, Natal 59078-970, RN, Brazil
4
Bioinformatics Multidisciplinary Environment (BioME), Federal University of Rio Grande do Norte, Natal 59078-970, RN, Brazil
5
Department of Pharmacology, Institute for Basic Health Sciences, Federal University of Rio Grande do Sul, Porto Alegre 90035-003, RS, Brazil
6
Cancer and Neurobiology Laboratory, Experimental Research Center, Clinical Hospital (CPE-HCPA), Federal University of Rio Grande do Sul, Porto Alegre 90035-003, RS, Brazil
7
National Science and Technology Institute for Children’s Cancer Biology and Pediatric Oncology—INCT BioOncoPed, Porto Alegre 90035-003, RS, Brazil
8
Department of Computer Engineering and Automation, Federal University of Rio Grande do Norte, Natal 59078-970, RN, Brazil
*
Authors to whom correspondence should be addressed.
Int. J. Mol. Sci. 2024, 25(5), 3020; https://doi.org/10.3390/ijms25053020
Submission received: 5 January 2024 / Revised: 27 February 2024 / Accepted: 29 February 2024 / Published: 5 March 2024
(This article belongs to the Special Issue Recent Advances in Brain Cancers)

Abstract

:
Gliomas comprise most cases of central nervous system (CNS) tumors. Gliomas afflict both adults and children, and glioblastoma (GBM) in adults represents the clinically most important type of malignant brain cancer, with a very poor prognosis. The cell surface glycoprotein CD114, which is encoded by the CSF3R gene, acts as the receptor for the granulocyte colony stimulating factor (GCSF), and is thus also called GCSFR or CSFR. CD114 is a marker of cancer stem cells (CSCs), and its expression has been reported in several cancer types. In addition, CD114 may represent one among various cases where brain tumors hijack molecular mechanisms involved in neuronal survival and synaptic plasticity. Here, we describe CSF3R mRNA expression in human gliomas and their association with patient prognosis as assessed by overall survival (OS). We found that the levels of CSF3R/CD114 transcripts are higher in a few different types of gliomas, namely astrocytoma, pilocytic astrocytoma, and GBM, in comparison to non-tumoral neural tissue. We also observed that higher expression of CSF3R/CD114 in gliomas is associated with poorer outcome as measured by a shorter OS. Our findings provide early evidence suggesting that CSF3R/CD114 shows a potential role as a prognosis marker of OS in patients with GBM.

1. Introduction

Gliomas comprise about 80 percent of central nervous system (CNS) cancers in adults. In children, CNS cancers including pediatric gliomas represent the majority of solid tumors. Gliomas are classified into different types, namely astrocytoma, oligodendroglioma and glioblastoma (GBM). The most prevalent and lethal glioma type is GBM. Patients with this tumor type have a poor prognosis even after undergoing multimodal therapy combining surgical resection, radiotherapy, and treatment with temozolomide. Status of the isocitrate dehydrogenase (IDH) gene allows the classification of GBM into three groups, namely IDH wild-type GBM, which represents about 90% of cases, mutated IDH, or not specified GBM (NOS, unevaluated status) [1,2,3,4,5].
The cell surface protein CD114, encoded by the CSF3R gene, is a receptor for the granulocyte colony stimulating factor (GCSF), being thus also called GCSFR or CSFR [6,7]. Upon stimulation by GCSF, CD114 activates the transcription factor signal transducer and activator of transcription 3 (STAT3), which promotes a cancer stem cell (CSC) phenotype [8]. CD114 has been proposed as a marker for CSCs in neural crest-derived tumors such as neuroblastoma (NB) and melanoma [7,9,10]. Expression of CD114 is found in solid tumors including brain, ovarian, cervical, bladder, and skin cancers [7,10,11,12,13,14,15,16,17,18]. In addition, CSF3R mutations have been found in rare types of leukemia [19,20].
Brain cancer hijacks molecular and cellular mechanisms of neuronal plasticity [21,22,23,24,25,26]. GCSF acts as a growth factor stimulating the survival and plasticity of neurons and neural stem cells [27]. GCSF stimulates neuronal survival and neurogenesis [6] and acts synergistically with stem cell factor (SCF) to stimulate neurite outgrowth in cortical neurons [28]. The combination of GCSF and SCF also protects from neurodegeneration and promotes neurostructure network reorganization in a mouse model of traumatic brain injury [29]. Systemic administration of GCSF ameliorates learning and memory impairments and improves disruptions in dendritic morphology spine density, and mature spines in hippocampal CA1 neurons induced by brain ischemia in rats [30]. In vivo treatment with of GCSF restores long-term depression in hippocampal slices from transgenic the APP/PS1 mouse model of Alzheimer’s disease (AD) [31]. In the most common type of pediatric brain tumor, medulloblastoma (MB), the expression of the GCSF receptor CD144 is found in MB cell lines, patient-derived xenograft (PDX) tumors, and primary patient tumors. CD114+ cells show resistance against cytotoxic chemotherapy and are responsive to stimulation by GCSF [17]. In addition, transcript levels of the CSF3R gene have been identified across molecular subgroups of MB [18]. A previous study focusing on human gliomas described widespread RNA and protein expression of GCSF. In addition, proliferation and migration were stimulated by exposure to GCSF in CD114+ glioma cells, whereas GCSF inhibition by a neutralizing antibody impaired cell growth and migration [11]. However, the potential role of CSF3R/CD114 as a biomarker in gliomas remains poorly understood and warrants further investigation. In the present study, we describe transcript levels of CSF3R in gliomas and their association with patient prognosis as assessed by overall survival (OS).

2. Results

2.1. CSF3R/CD114 Transcript Levels Are Higher in Different Glioma Types Compared to Non-Tumoral Neural Tissue

In comparison to non-tumoral neural tissue (n = 8), significantly higher levels of CSF3R transcripts were observed in astrocytoma, pilocytic astrocytoma, and glioblastoma (all ps < 0.001; Figure 1; Table 1), whereas oligoastrocytoma and oligodendroglioma did not show significant differences in the French cohort.

2.2. Higher Gene Expression of CSF3R/CD114 Is Associated with Poorer Patient Outcome in GBM

OS was analyzed using 266 glioma samples from the French dataset. Patients were divided into two groups (low or high expression) based on the expression level of the CSF3R/CD114 gene. Analysis of glioma patient OS in relation to CSF3R/CD114 transcript levels in tumors showed that, when all glioma types were pooled together, higher CSF3R/CD114 expression was significantly associated with a poorer prognosis as assessed by shorter OS (Figure 2A). We then assessed each glioma type, grouping grade II and III samples together due to the limited number of samples in each subgroup. A significant association between high CSF3R mRNA levels and shorter OS was observed in patients with GBM (n = 150; p < 0.05; Figure 2B).

2.3. CSF3R/CD114 Transcript Levels and OS in Patients Bearing IDH-Mutated versus IDH Wild-Type GBM Tumors

Patients were divided into IDH-mutated and IDH wild-type tumor groups, and then patients within each group were also classified according to high or low CSF3R expression levels. There was no significant difference in CSF3R mRNA expression between patients with IDH-mutated versus wild-type GBM tumors (Figure 3A). There were no significant differences in OS between patients with low- or high-expressing tumors within the IDH-mutated or IDH wild-type groups. Patients with wild-type IDH showed an apparent reduction in OS regardless of CSF3R levels (Figure 3B).

2.4. CSF3R/CD114 Transcript Levels in the Brain Lower Grade Glioma (TCGA-LGG) Cohort

We went on to analyze CSF3R/CD114 expression in The Cancer Genome Atlas (TCGA) Brain Lower Grade Glioma (TCGA-LGG) cohort, which contains 513 glioma samples distributed across tumor types astrocytoma, oligoastrocytoma, and oligodendroglioma. Consistently with the data obtained with the French cohort, astrocytoma tumors displayed the highest gene expression of CSF3R (Figure 4).

2.5. Higher CSF3R/CD114 Gene Expression Is Associated with Shorter Patient OS in Glioma Tumors from the TCGA-LGG Cohort

In glioma tumors from the TCGA-LGG dataset, a poorer patient prognosis as assessed by shorter OS was significantly associated with high CSF3R/CD114 expression when all tumor types were polled together (Figure 5A), as well as when astrocytoma (n = 194), oligoastrocytoma (n = 130), and oligodendroglioma (n = 189) tumors were analyzed separately (Figure 5B–D).

3. Discussion

GCSF is a cytokine encoded by the CSF3 gene that acts as a hematopoietic growth factor regulating the function of granulocytic precursors and neutrophils. GCSF actions are mediated by activation of its receptor, named GCSFR or CD114. Recombinant human GCSF is clinically used to prevent neutropenia, due to its effects on neutrophil mobilization and maturation [32]. GCSF/CD114 signaling has also been investigated as a modulator of neuronal survival, synaptic plasticity [6,28,29,30,31], and cancer [7,10,11,12,13,14,15,16,17,18,19,20]. Specifically, CD114 has been put forward as a marker to identify CSC subpopulations associated with tumorigenicity, metastasis, and resistance to treatment [7,9,10].
In epithelial skin tumors, the presence of CD114 is significantly higher compared to normal skin, Bowen’s disease, or actinic keratosis, and was associated with carcinogenesis. However, no association between the protein expression of CD114 and patient mortality was found [13]. Similarly, different levels of CSF3R transcripts occur among different tumor subgroups and subtypes of MB, but no significant association with patient survival was established [18]. A previous study in glioma analyzed the RNA and protein expression of GCSF and CD114 in a set of 22 human gliomas (WHO grade II, III, and IV) and cell cultures derived from these tumors. Although the expression of GCSF and CD114, as well as that of granulocyte macrophage colony-stimulating factor (GMCSF) and its receptor, was found in all glioma tumors and cell cultures, the coexpression of both factors and their receptors was selectively observed in grade IV tumors (GBMs), and thus, the expression correlates with advanced tumor stage [11]. The present transcript analyses indicate that a significant association between high CSF3R mRNA levels and poorer prognosis measured by shorter OS was found in patients with gliomas. These early in silico findings suggest that further experimental studies should characterize the effects of GCSF/CD114 inhibition in experimental GBM models.
We did not find a significant impact of CSF3R mRNA expression on OS when GBM patients were divided according to IDH status (mutated versus wild-type), possibly because of the limited number of samples available in each subgroup. Also, there was no significant difference in CSF3R levels between mutated or IDH wild-type GBM tumors. Some IDH1 mutations are considered prognostic markers, with patients bearing mutated tumors showing improved OS [33] Consistently with these data, we found an apparently poorer OS in patients with wild-type IDH GBM. One study reported that in mice bearing mutated IDH GBM, GCSF is secreted by GBM CSCs, and blocking GCSF accelerates tumor progression by acting on tumor-infiltrating myeloid cells [34].
STAT3 is involved in mediating the cellular effects of CD114 activation. Several studies have indicated that STAT3 is an oncogene in GBM. The activation of STAT3 is associated with shorter OS and progression-free survival in patients with GBM [35], and STAT3 is required for the maintenance of a CSC phenotype in GBM cells [36]. STAT3 may have a dual role in GBM, either promoting or suppressing GBM tumor progression [37,38]. Future experiments should investigate the role of STAT3 downstream of CD114 in different glioma types.

4. Materials and Methods

4.1. Gene Expression, and Tumor and Patient Data

CSF3R mRNA expression levels were normalized using the R2 Genomics Analysis and Visualization Platform (http://r2.amc.nl). Data were obtained from the French cohort (total n = 284; Gene Expression Omnibus—GEO ID: GSE16011; https://www.ncbi.nlm.nih.gov/geo/query/acc.cgi?acc=gse16011, website accessed on 31 January 2024), which included samples from different primary glioma types as well as non-tumoral neural tissue as a control [26,39,40]. Patient characteristics are summarized in Table 1. Tumors were classified into different types according to data available in the dataset.
Normalization of raw microarray data was performed using the Robust Multichip Average (RMA) method, and quality control was conducted through Affy Bioconductor/R. We also used data from the TCGA-LGG cohort (total n = 513). The already processed and normalized expression data were obtained from the cBioPortal. Characteristics of patients in the TCGA-LGG dataset are shown in Table 2.

4.2. Statistical Analysis

Clinical information of patients in the French cohort was obtained through the ‘geoquery’ package and data described by Gravendeel et al. [39]. Clinical information of patients in the TCGA-LGG cohort was acquired through the cBioPortal. To investigate differences between glioma tumor types and control neural tissue in the French cohort, we used the Wilcoxon test and the Dunn test to perform specific comparisons among tissue types. Assessment of statistical significance was conducted through the Holm-adjusted p-value test. Analyses were carried out using the ‘ggstatsplot’ package.
To examine associations between gene expression and patient OS in the French cohort, 8 control samples and additional 12 glioma samples that lacked information about patient status (‘alive’ or ‘dead’) were excluded from the analysis, resulting in a total of 266 samples. To classify patients into high and low CSF3R gene expression groups, we used the ‘Survminer’ package with ‘minprop = 0.2’. Patients within specific IDH-mutated and IDH wild-type groups were also stratified based on high and low CSF3R expression levels. Survival analysis was conducted using the ‘Survival’ package. Patient overall survival (OS) was measured from the day of diagnosis until death or date of last follow-up. OS was calculated using the Kaplan–Meier estimate.
The R2 Genomics Analysis and Visualization Platform (http://r2.amc.nl) was used to compare patients with IDH-mutated versus IDH wild-type GBM tumors from the French cohort. Welch’s ANOVA was carried out for these comparisons, with p values < 0.01 considered to indicate statistical significance. Kaplan–Meier survival curves were also estimated for IDH-mutated and IDH wild-type groups to assess patient OS.

5. Conclusions

The main novel finding of the present study, obtained by analyzing public glioma and neural tissue data, is the association of poorer patient outcome assessed by a reduction in OS in patients with high CSF3R/CD114 mRNA expression in different types of glioma tumors. Further studies should explore the role of CD114 in glioma tumor cell lines, primary tumors, and tumor microenvironment to increase our understanding of the role of GCSF and similar growth factors in brain cancer progression.

Author Contributions

Conception and design, S.A.B., M.D., O.M., R.R., M.A.C.F. and G.R.I.; Acquisition of data: S.A.B., M.D., M.A.C.F. and G.R.I.; Analysis and interpretation of data: S.A.B., M.D., O.M., R.R., M.A.C.F. and G.R.I.; Drafting the article: S.A.B., R.R. and G.R.I.; Critically revising the article: S.A.B., M.D., O.M., R.R., M.A.C.F. and G.R.I. All authors have read and agreed to the published version of the manuscript.

Funding

This research was supported by the National Council for Scientific and Technological Development (CNPq, MCTI, Brazil) grants 305647/2019-9, 405608/2021-7, and 406484/2022-8 (INCT BioOncoPed); The Center for Advanced Neurology and Neurosurgery (CEANNE); the Mackenzie Evangelical University; and the Children’s Cancer Institute (ICI).

Institutional Review Board Statement

Not applicable.

Informed Consent Statement

Not applicable.

Data Availability Statement

The data presented in this study are available upon request from the corresponding author.

Acknowledgments

The authors thank Barbara Kunzler Souza and Epigenica Biosciences for providing assistance in gene expression analysis and data interpretation.

Conflicts of Interest

The authors declare no conflicts of interest.

References

  1. Wen, P.Y.; Kesari, S. Malignant gliomas in adults. N. Engl. J. Med. 2008, 359, 492–507. [Google Scholar] [CrossRef]
  2. Alexander, B.M.; Cloughesy, T.F. Adult glioblastoma. J. Clin. Oncol. 2017, 35, 2402–2409. [Google Scholar] [CrossRef]
  3. Funakoshi, Y.; Hata, N.; Kuga, D.; Hatae, R.; Sangatsuda, Y.; Fujioka, Y.; Takigawa, K.; Mizoguchi, M. Pediatric glioma: An update of diagnosis, biology, and treatment. Cancers 2021, 13, 758. [Google Scholar] [CrossRef]
  4. Louis, D.N.; Perry, A.; Wesseling, P.; Brat, D.J.; Cree, I.A.; Figarella-Branger, D.; Hawkins, C.; Ng, H.K.; Pfister, S.M.; Reifenberger, G.; et al. The 2021 WHO Classification of Tumors of the Central Nervous System: A summary. Neuro Oncol. 2021, 23, 1231–1251. [Google Scholar] [CrossRef]
  5. Hardigan, A.A.; Jackson, J.D.; Patel, A.P. Surgical management and advances in the treatment of glioma. Semin. Neurol. 2023, 43, 810–824. [Google Scholar] [CrossRef] [PubMed]
  6. Schneider, A.; Krüger, C.; Steigleder, T.; Weber, D.; Pitzer, C.; Laage, R.; Aronowski, J.; Maurer, M.H.; Gassler, N.; Mier, W.; et al. The hematopoietic factor G-CSF is a neuronal ligand that counteracts programmed cell death and drives neurogenesis. J. Clin. Investig. 2005, 115, 2083–2098. [Google Scholar] [CrossRef] [PubMed]
  7. Zage, P.E.; Whittle, S.B.; Shohet, J.M. CD114: A new member of the neural crest-derived cancer stem cell marker family. J. Cell Biochem. 2017, 118, 221–231. [Google Scholar] [CrossRef] [PubMed]
  8. Agarwal, S.; Lakoma, A.; Chen, Z.; Hicks, J.; Metelitsa, L.S.; Kim, E.S.; Shohet, J.M. G-CSF promotes neuroblastoma tumorigenicity and metastasis via STAT3-dependent cancer stem cell activation. Cancer Res. 2015, 75, 2566–2579. [Google Scholar] [CrossRef] [PubMed]
  9. Hsu, D.M.; Agarwal, S.; Benham, A.; Coarfa, C.; Trahan, D.N.; Chen, Z.; Stowers, P.N.; Courtney, A.N.; Lakoma, A.; Barbieri, E.; et al. G-CSF receptor positive neuroblastoma subpopulations are enriched in chemotherapy-resistant or relapsed tumors and are highly tumorigenic. Cancer Res. 2013, 73, 4134–4146. [Google Scholar] [CrossRef] [PubMed]
  10. Zhang, L.; Agarwal, S.; Shohet, J.M.; Zage, P.E. CD114 expression mediates melanoma tumor cell growth and treatment resistance. Anticancer Res. 2015, 35, 3787–3792. [Google Scholar] [PubMed]
  11. Mueller, M.M.; Herold-Mende, C.C.; Riede, D.; Lange, M.; Steiner, H.H.; Fusenig, N.E. Autocrine growth regulation by granulocyte colony-stimulating factor and granulocyte macrophage colony-stimulating factor in human gliomas with tumor progression. Am. J. Pathol. 1999, 155, 1557–1567. [Google Scholar] [CrossRef]
  12. Kyo, S.; Kanaya, T.; Takakura, M.; Inoue, M. A case of cervical cancer with aggressive tumor growth: Possible autocrine growth stimulation by G-CSF and Il-6. Gynecol. Oncol. 2000, 78 Pt 1, 383–387. [Google Scholar] [CrossRef]
  13. Hirai, K.; Kumakiri, M.; Fujieda, S.; Sunaga, H.; Lao, L.M.; Imamura, Y.; Ueda, K.; Fukuda, M. Expression of granulocyte colony-stimulating factor and its receptor in epithelial skin tumors. J. Dermatol. Sci. 2001, 25, 179–188. [Google Scholar] [CrossRef] [PubMed]
  14. Savarese, T.M.; Mitchell, K.; McQuain, C.; Campbell, C.L.; Guardiani, R.; Wuu, J.; Ollari, C.; Reale, F.; Nelson, B.E.; Chen, A.; et al. Coexpression of granulocyte colony stimulating factor and its receptor in primary ovarian carcinomas. Cancer Lett. 2001, 162, 105–115. [Google Scholar] [CrossRef] [PubMed]
  15. Chakraborty, A.; Guha, S. Granulocyte colony-stimulating factor/granulocyte colony-stimulating factor receptor biological axis promotes survival and growth of bladder cancer cells. Urology 2007, 69, 1210–1215. [Google Scholar] [CrossRef]
  16. Hanumanthu, V.S.; Pirruccello, S.J. GCSF-R expression in myelodysplastic and myeloproliferative disorders and blast dysmaturation in CML. Am. J. Clin. Pathol. 2013, 140, 155–164. [Google Scholar] [CrossRef]
  17. Paul, M.R.; Huo, Y.; Liu, A.; Lesperance, J.; Garancher, A.; Wechsler-Reya, R.J.; Zage, P.E. Characterization of G-CSF receptor expression in medulloblastoma. Neurooncol. Adv. 2020, 2, vdaa062. [Google Scholar] [CrossRef]
  18. Monteiro, J.M.; Reis Ramos, J.I.; Teixeira e Sousa, I.; Bighetti-Trevisan, R.L.; Ribas Filho, J.M.; Isolan, G.R. Identification of CD114 membrane receptors as a molecular target in medulloblastomas. Int. J. Mol Sci. 2023, 24, 5331. [Google Scholar] [CrossRef]
  19. Maxson, J.E.; Gotlib, J.; Pollyea, D.A.; Fleischman, A.G.; Agarwal, A.; Eide, C.A.; Bottomly, D.; Wilmot, B.; McWeeney, S.K.; Tognon, C.E.; et al. Oncogenic CSF3R mutations in chronic neutrophilic leukemia and atypical CML. N. Engl. J. Med. 2013, 368, 1781–1790. [Google Scholar] [CrossRef]
  20. Maxson, J.E.; Ries, R.E.; Wang, Y.C.; Gerbing, R.B.; Kolb, E.A.; Thompson, S.L.; Guidry Auvil, J.M.; Marra, M.A.; Ma, Y.; Zong, Z.; et al. CSF3R mutations have a high degree of overlap with CEBPA mutations in pediatric AML. Blood 2016, 127, 3094–3098. [Google Scholar] [CrossRef] [PubMed]
  21. Venkatesh, H.S.; Johung, T.B.; Caretti, V.; Noll, A.; Tang, Y.; Nagaraja, S.; Gibson, E.M.; Mount, C.W.; Polepalli, J.; Mitra, S.S.; et al. Neuronal activity promotes glioma growth through neuroligin-3 secretion. Cell 2015, 161, 803–816. [Google Scholar] [CrossRef] [PubMed]
  22. Venkatesh, H.S.; Morishita, W.; Geraghty, A.C.; Silverbush, D.; Gillespie, S.M.; Arzt, M.; Tam, L.T.; Espenel, C.; Ponnuswami, A.; Ni, L.; et al. Electrical and synaptic integration of glioma into neural circuits. Nature 2019, 573, 539–545. [Google Scholar] [CrossRef] [PubMed]
  23. Pinheiro, K.V.; Thomaz, A.; Souza, B.K.; Metcalfe, V.A.; Freire, N.H.; Brunetto, A.T.; de Farias, C.B.; Jaeger, M.; Bambini, V.; Smith, C.G.S.; et al. Expression and pharmacological inhibition of TrkB and EGFR in glioblastoma. Mol. Biol. Rep. 2020, 47, 6817–6828. [Google Scholar] [CrossRef] [PubMed]
  24. Thomaz, A.; Jaeger, M.; Brunetto, A.L.; Brunetto, A.T.; Gregianin, L.; de Farias, C.B.; Ramaswamy, V.; Nör, C.; Taylor, M.D.; Roesler, R. Neurotrophin signaling in medulloblastoma. Cancers 2020, 12, 2542. [Google Scholar] [CrossRef] [PubMed]
  25. Krishna, S.; Choudhury, A.; Keough, M.B.; Seo, K.; Ni, L.; Kakaizada, S.; Lee, A.; Aabedi, A.; Popova, G.; Lipkin, B.; et al. Glioblastoma remodelling of human neural circuits decreases survival. Nature 2023, 617, 599–607. [Google Scholar] [CrossRef] [PubMed]
  26. Rodrigues, E.M.; Giovanini, A.F.; Ribas, C.A.P.M.; Malafaia, O.; Roesler, R.; Isolan, G.R. The nervous system development regulator neuropilin-1 as a potential prognostic marker and therapeutic target in brain cancer. Cancers 2023, 15, 4922. [Google Scholar] [CrossRef]
  27. Wallner, S.; Peters, S.; Pitzer, C.; Resch, H.; Bogdahn, U.; Schneider, A. The granulocyte-colony stimulating factor has a dual role in neuronal and vascular plasticity. Front. Cell Dev. Biol. 2015, 3, 48. [Google Scholar] [CrossRef]
  28. Su, Y.; Cui, L.; Piao, C.; Li, B.; Zhao, L.R. The effects of hematopoietic growth factors on neurite outgrowth. PLoS ONE 2013, 8, e75562. [Google Scholar] [CrossRef]
  29. He, J.; Russell, T.; Qiu, X.; Hao, F.; Kyle, M.; Chin, L.; Zhao, L.R. The contribution of stem cell factor and granulocyte colony-stimulating factor in reducing neurodegeneration and promoting neurostructure network reorganization after traumatic brain injury. Brain Res. 2020, 1746, 147000. [Google Scholar] [CrossRef]
  30. Sarkala, H.B.; Jahanshahi, M.; Dolatabadi, L.K.; Namavar, M.R. G-CSF improved the memory and dendritic morphology impairments in the hippocampal CA1 pyramidal neurons after brain ischemia in the male rats. Metab. Brain Dis. 2023, 38, 2573–2581. [Google Scholar] [CrossRef]
  31. Song, S.; Wang, X.; Sava, V.; Weeber, E.J.; Sanchez-Ramos, J. In vivo administration of granulocyte colony-stimulating factor restores long-term depression in hippocampal slices prepared from transgenic APP/PS1 mice. J. Neurosci. Res. 2014, 92, 975–980. [Google Scholar] [CrossRef] [PubMed]
  32. Park, S.D.; Saunders, A.S.; Reidy, M.A.; Bender, D.E.; Clifton, S.; Morris, K.T. A review of granulocyte colony-stimulating factor receptor signaling and regulation with implications for cancer. Front. Oncol. 2022, 12, 932608. [Google Scholar] [CrossRef]
  33. Sareen, H.; Ma, Y.; Becker, T.M.; Roberts, T.L.; de Souza, P.; Powter, B. Molecular biomarkers in glioblastoma: A systematic review and meta-analysis. Int. J. Mol. Sci. 2022, 23, 8835. [Google Scholar] [CrossRef]
  34. Alghamri, M.S.; McClellan, B.L.; Avvari, R.P.; Thalla, R.; Carney, S.; Hartlage, C.S.; Haase, S.; Ventosa, M.; Taher, A.; Kamran, N.; et al. G-CSF secreted by mutant IDH1 glioma stem cells abolishes myeloid cell immunosuppression and enhances the efficacy of immunotherapy. Sci. Adv. 2021, 7, eabh3243. [Google Scholar] [CrossRef]
  35. Masliantsev, K.; Pinel, B.; Balbous, A.; Guichet, P.O.; Tachon, G.; Milin, S.; Godet, J.; Duchesne, M.; Berger, A.; Petropoulos, C.; et al. Impact of STAT3 phosphorylation in glioblastoma stem cells radiosensitization and patient outcome. Oncotarget 2017, 9, 3968–3979. [Google Scholar] [CrossRef]
  36. Im, C.N.; Yun, H.H.; Song, B.; Youn, D.Y.; Cui, M.N.; Kim, H.S.; Park, G.S.; Lee, J.H. BIS-mediated STAT3 stabilization regulates glioblastoma stem cell-like phenotypes. Oncotarget 2016, 7, 35056–35070. [Google Scholar] [CrossRef]
  37. de la Iglesia, N.; Konopka, G.; Lim, K.L.; Nutt, C.L.; Bromberg, J.F.; Frank, D.A.; Mischel, P.S.; Louis, D.N.; Bonni, A. Deregulation of a STAT3-interleukin 8 signaling pathway promotes human glioblastoma cell proliferation and invasiveness. J. Neurosci. 2008, 28, 5870–5878. [Google Scholar] [CrossRef]
  38. Luwor, R.B.; Stylli, S.S.; Kaye, A.H. The role of Stat3 in glioblastoma multiforme. J. Clin. Neurosci. 2013, 20, 907–911. [Google Scholar] [CrossRef]
  39. Gravendeel, L.A.; Kouwenhoven, M.C.; Gevaert, O.; de Rooi, J.J.; Stubbs, A.P.; Duijm, J.E.; Daemen, A.; Bleeker, F.E.; Bralten, L.B.; Kloosterhof, N.K.; et al. Intrinsic gene expression profiles of gliomas are a better predictor of survival than histology. Cancer Res. 2009, 69, 9065–9072. [Google Scholar] [CrossRef]
  40. Gritsenko, P.G.; Atlasy, N.; Dieteren, C.E.J.; Navis, A.C.; Venhuizen, J.H.; Veelken, C.; Schubert, D.; Acker-Palmer, A.; Westerman, B.A.; Wurdinger, T.; et al. p120-catenin-dependent collective brain infiltration by glioma cell networks. Nat. Cell Biol. 2020, 22, 97–107. [Google Scholar] [CrossRef]
Figure 1. CSF3R/CD114 gene expression in different types of glioma and non-tumoral neural tissue. Data were obtained from the French cohort (total dataset n = 284) and analyzed with the R2 Genomics Analysis and Visualization Platform (http://r2.amc.nl). Results are presented in boxplot format as log2-transformed signal intensity. Glioma tumor samples were classified as astrocytoma, pilocytic astrocytoma, oligoastrocytoma, oligodendroglioma, and glioblastoma.; p values for comparisons are indicated in the figure.
Figure 1. CSF3R/CD114 gene expression in different types of glioma and non-tumoral neural tissue. Data were obtained from the French cohort (total dataset n = 284) and analyzed with the R2 Genomics Analysis and Visualization Platform (http://r2.amc.nl). Results are presented in boxplot format as log2-transformed signal intensity. Glioma tumor samples were classified as astrocytoma, pilocytic astrocytoma, oligoastrocytoma, oligodendroglioma, and glioblastoma.; p values for comparisons are indicated in the figure.
Ijms 25 03020 g001
Figure 2. CSF3R/CD114 gene expression and OS in patients with glioma. Results are shown for (A) all gliomas pooled together (n = 266) or (B) GBM patients only (n = 150). Data were obtained from the French cohort. Patient OS was measured from the day of diagnosis until death or date of last follow-up, and calculated using the Kaplan–Meier estimate, with median values and long-rank statistics; p values are indicated in the figure.
Figure 2. CSF3R/CD114 gene expression and OS in patients with glioma. Results are shown for (A) all gliomas pooled together (n = 266) or (B) GBM patients only (n = 150). Data were obtained from the French cohort. Patient OS was measured from the day of diagnosis until death or date of last follow-up, and calculated using the Kaplan–Meier estimate, with median values and long-rank statistics; p values are indicated in the figure.
Ijms 25 03020 g002
Figure 3. CSF3R/CD114 expression and OS in patients bearing IDH-mutated versus IDH wild-type GBM tumors. Data were obtained from the French cohort. (A) Results for gene expression are presented in boxplot format as log2-transformed signal intensity. (B) Patient OS was measured from the day of diagnosis until death or date of last follow-up and calculated using the Kaplan–Meier estimate.
Figure 3. CSF3R/CD114 expression and OS in patients bearing IDH-mutated versus IDH wild-type GBM tumors. Data were obtained from the French cohort. (A) Results for gene expression are presented in boxplot format as log2-transformed signal intensity. (B) Patient OS was measured from the day of diagnosis until death or date of last follow-up and calculated using the Kaplan–Meier estimate.
Ijms 25 03020 g003
Figure 4. CSF3R/CD114 expression in astrocytoma (n = 194), oligoastrocytoma (n = 130), and oligodendroglioma (n = 189) tumors from the TCGA-LGG cohort; p values are indicated in the figure.
Figure 4. CSF3R/CD114 expression in astrocytoma (n = 194), oligoastrocytoma (n = 130), and oligodendroglioma (n = 189) tumors from the TCGA-LGG cohort; p values are indicated in the figure.
Ijms 25 03020 g004
Figure 5. CSF3R/CD114 gene expression and OS of patients in the TCGA-LGG cohort. (A) glioma types polled together, (B) astrocytoma (n = 194), (C) oligoastrocytoma (n = 130), and (D) oligodendroglioma (n = 189).
Figure 5. CSF3R/CD114 gene expression and OS of patients in the TCGA-LGG cohort. (A) glioma types polled together, (B) astrocytoma (n = 194), (C) oligoastrocytoma (n = 130), and (D) oligodendroglioma (n = 189).
Ijms 25 03020 g005
Table 1. Summary of characteristics of patients from the French cohort selected for survival analysis.
Table 1. Summary of characteristics of patients from the French cohort selected for survival analysis.
Glioma Type
CharacteristicsAstrocytomaGlioblastomaOligoastrocytomaOligodendrogliomaPilocytic
Astrocytoma
Total number of samples2815326518
Mean age (years)42.5453.8448.0249.1625.49
GenderMale2010318314
Female8508204
Mean overall
survival (OS, months)
32.5716.2545.6070.9974.72
StatusAlive43276
Dead2314324430
GradeGrade_II120370
Grade_III16023440
Table 2. Summary of characteristics of patients in the TCGA-LGG cohort.
Table 2. Summary of characteristics of patients in the TCGA-LGG cohort.
Glioma Type
CharacteristicsAstrocytomaOligoastrocytomaOligodendroglioma
Total number of samples194130189
Mean age (years)41.840.9645.39
GenderMale10872105
Female865884
Mean overall
survival (OS, months)
28.9330.535.28
StatusAlive136105147
Dead582542
GradeGrade_II6374111
Grade_III1315578
Disclaimer/Publisher’s Note: The statements, opinions and data contained in all publications are solely those of the individual author(s) and contributor(s) and not of MDPI and/or the editor(s). MDPI and/or the editor(s) disclaim responsibility for any injury to people or property resulting from any ideas, methods, instructions or products referred to in the content.

Share and Cite

MDPI and ACS Style

Bark, S.A.; Dalmolin, M.; Malafaia, O.; Roesler, R.; Fernandes, M.A.C.; Isolan, G.R. Gene Expression of CSF3R/CD114 Is Associated with Poorer Patient Survival in Glioma. Int. J. Mol. Sci. 2024, 25, 3020. https://doi.org/10.3390/ijms25053020

AMA Style

Bark SA, Dalmolin M, Malafaia O, Roesler R, Fernandes MAC, Isolan GR. Gene Expression of CSF3R/CD114 Is Associated with Poorer Patient Survival in Glioma. International Journal of Molecular Sciences. 2024; 25(5):3020. https://doi.org/10.3390/ijms25053020

Chicago/Turabian Style

Bark, Samir Ale, Matheus Dalmolin, Osvaldo Malafaia, Rafael Roesler, Marcelo A. C. Fernandes, and Gustavo R. Isolan. 2024. "Gene Expression of CSF3R/CD114 Is Associated with Poorer Patient Survival in Glioma" International Journal of Molecular Sciences 25, no. 5: 3020. https://doi.org/10.3390/ijms25053020

Note that from the first issue of 2016, this journal uses article numbers instead of page numbers. See further details here.

Article Metrics

Back to TopTop