Skip to main content

Prognostic values and immune infiltration of KLF15, AQP7, AGPAT9 in glioma and glioblastoma

Abstract

Backgrounds

The overall survival of patients with lower-grade gliomas and glioblastoma varies greatly. No reliable or existing procedures can accurately forecast survival and prognostic biomarkers for early diagnosis in glioma and glioblastoma. However, investigations are progressing in immunotherapy, tumor purity, and tumor microenvironment which may be therapeutic targets for glioma and glioblastoma.

Results

This study indicated the possible prognostic signatures that can be used to identify immune-related prognostic biomarkers in the prediction of the survival of low-grade glioma (LGG) patients which may be a possible therapeutic target. In addition, the Kaplan–Meier plot, ESTIMATE algorithm, and TIMER 2.0 analysis indicated that Krüppel-like factor 15 (KLF15) p = 0.030, Aquaporin 7 (AQP7) p = 0.001, and Human 1-acylglycerol-3-phosphate O-acyltransferase 9 (AGPAT9) p = 0.005 are significantly associated in glioma. Hence, they may be possible prognostic biomarkers in glioma. Meanwhile, in the glioblastoma, only KLF15 has a significant association with glioblastoma (p = 0.025). Stromal and immune scores of gliomas were determined from transcriptomic profiles of LGG cohort from TCGA (The Cancer Genome Atlas) using the ESTIMATE (Estimation of Stromal and Immune cells in Malignant Tumours using Expression data algorithm). The immune infiltration of the KLF15, AQP7, and AGPAT9 for low-grade glioma and glioblastoma was determined using TIMER immune 2.0 which indicates correlation with tumor purity for KLF15, AQP7, and AGPAT9, but only KLF15 and AGPAT9 are significantly associated in both glioma and glioblastoma, respectively.

Conclusions

These results highlight the significance of microenvironment monitoring, analysis of glioma and glioblastoma prognosis, and targeted immunotherapy. To our knowledge, this is the first time to investigate an analysis that revealed that KLF15, AQP7, and AGPAT9 may be important prognostic biomarkers for patients with glioma and KLF15 for patients with glioblastoma. Meanwhile, KLF15 and AGPAT9 are significantly associated in both glioma and glioblastoma, respectively, for tumor purity.

Backgrounds

Glioma is the most prevalent primary malignant brain tumor and can be divided into distinct categories. According to the WHO grading system, it can be categorized into astrocytomas, diffuse low-grade, intermediate-grade, oligodendrogliomas, and mixed oligoastrocytomas [1,2,3]. The most frequent treatment for glioma is surgical resection in combination with chemoradiotherapy. Due to its highly invasive nature, surgical resection may be difficult to treat, and residual tumor could lead to malignant progressions and even reoccurrence in the long run [4]. Although the classification of low-grade glioma (LGG) is recognized worldwide, it may not adequately predict its survival rate; however, clinicians tend to depend on genetic classifications to guide its treatment [5,6,7]. The survival outcomes of LGG vary widely among different patients [8]. However, some LGGs stay stable for a long period while some progress into glioblastoma [9,10,11]. Notwithstanding more investigations are required to elucidate whether gliomas progress to glioblastoma. Gliomas account for approximately 75% of primary cancerous brain tumors [12]. In the USA, about 13,000 people die and 18,000 new cases of CNS tumors and malignant brain tumors arise each year due to glioma prognosis and occurrence [13, 14], hence a need for therapeutics and early diagnosis of the diseases [15].

Glioma is a cancerous tumor of the central nervous system that begins in the glial cells that surround and nourish the brain's neurons [16]. In the treatment of gliomas, great progress has been made in genomic, transcriptomic, and epigenetic profiling [17,18,19,20,21]. Astrocytoma, ependymoma, glioblastoma, and oligodendroglioma are some of the different kinds of glioma [22]. Glioblastoma (GBM), the most common and aggressive primary kind of malignant brain tumor, is assumed to have started in glial cells [23,24,25]. Scientific evidence, on the other hand, reveals that GBM could have developed from a variety of cells with neural stem cell characteristics [26, 27]. GBM is slightly more common in men than in women, as well as in Caucasians and other white races and ethnicities [28, 29]. GBM is usually found in the supratentorial region of the brain such as hypothalamus, pituitary gland, pineal, and the four lobes: temporal, parietal, frontal, and occipital lobes, with cerebellum being a rare exception [30, 31]. Sixty-one percent of all primary gliomas are found in the brain's four lobes: 20% in the temporal lobe, 25% in the frontal lobe, 3% in the occipital lobe, and 13% in the parietal lobe [32]. Glioblastomas are divided into primary and secondary subtypes that originate along different genetic routes and affect individuals of various ages [33, 34]. Quite recently, glioblastoma with oligodendroglioma component is an uncommon subtype of glioblastoma that features certain parts that resemble anaplastic oligodendroglioma, according to the WHO [35, 36].

In clinical practices, mutated genes such as isocitrate dehydrogenase 1 (IDHI), IDH2, tumor protein 53 (TP53), epidermal growth factor receptor (EGFR), and alpha-thalassemia/mental retardation, X-linked (ATRX) are factors for the prognosis of patients with LGG [37,38,39]. Some other biomarkers, including 1p/19q codeletion and methylguanine methyltransferase (MGMT) promoter methylation, are also well-recognized and essential prognostic factors for LGGs [40,41,42]. Sometimes, these genetic factors fail to indicate accurate survival outcomes [43, 44]. Hence, further investigations are required to elucidate the functions and the mechanisms of the prognostic signatures.

Several studies have shown that cancer recurrence and progression are caused not only by the tumor's underlying genetic changes but also by tumor microenvironment (TME) [45,46,47]. The TME is basically composed of numerous cytokines, extracellular matrix molecules, immune cells, chemokines, fluids, and stromal cells [23, 48, 49]. The cells found in the TME reflect the evolutionary nature of cancer and together, promotes the tumor immune escape, tumor growth, and metastasis [50, 51]. Cancer researchers are not vividly aware of the impact of the TME on immune response or tumor progressions although multiple genetic mutations increase the prevalence of cancer [52]. TME can induce metabolic stress on immune cell infiltration thereby causing local immunosuppression and limited reinvigoration of antitumor immunity [53, 54]. However, having an in-depth understanding of the epigenetic, molecular composition, and function of the TME is essential to manage and treat cancer progressions, recurrence, and immune response [55,56,57]. Integrating multiple gene biomarkers instead of a single model would improve the accuracy of the prediction significantly [58,59,60,61].

The survival of glioma patients has received so much research and discovery in the aspect of neurosurgery, radiotherapy, and chemotherapy. However, a lot of challenges of glioma are yet to be solved. Currently, immunotherapy has unveiled possible therapy for cancer [54, 62, 63]. Investigations are currently going on in the area of immunotherapy, but there is still need for efficient molecular biomarkers to differentiate patients with possible sensitivity to immunotherapy [64, 65]. Therefore, it is very crucial to identify immune-related prognostic biomarkers which may be a possible therapeutic target and may be utilized for immunotherapy in patients.

Taken together, differential expressed genes (DEGs) using an immune stromal score in glioma and glioblastoma, transcriptional microarray of glioma cases from multiple TCGA cohorts were investigated to predict the survival of LGG and GBM patients. The following prognostic signatures, such as KLF15, AQP7, and AGPAT9, were used in this investigation to determine whether they have a significant association with glioma and glioblastoma using TCGA and the immune infiltration was unveiled for precise immunotherapy. To our knowledge, this is the first time to use these signatures for glioma and glioblastoma, hence unveiling prognostic biomarker and immune infiltration.

Methods

In this investigation, utilization of the Kaplan–Meier plots using Xena bower (http://xena.ucsc.edu/), ESTIMATE algorithm (Estimation of Stromal and Immune cells in Malignant Tumours using Expression), Timer 2.0 (http://timer.comp-genomics.org/timer/), and The Cancer Genome Atlas (TCGA) database were used to unveil the prognostic signature and immune infiltration of glioma and glioblastoma analysis.

Bioinformatics approaches were applied to integrate copy number variations and differential expressed genes of low-grade glioma. The immune cell proportion of the prognostic signatures, such as KLF15, AQP7, and AGPAT9, were determined using TIMER immune 2.0. In TIMER, the Gene module was used to identify the relationship between tumor gene expression and immune infiltration in low-grade glioma and glioblastoma. Stromal and immune scores of gliomas were estimated in transcriptomic profiles of LGG cohort from TCGA using the ESTIMATE. One hundred entries of TCGA cohort were entered and used to plot the graph showing the presence of stromal scores in tumor tissues, immune scores for the infiltration of immune cells in tumor tissues, and the ESTIMATE scores that infers tumor purity. Herein, we analyzed the immune infiltration landscape in LGGs, by applying single-sample gene set enrichment analysis (ssGSEA) to evaluate the relative abundance of each immune cell subpopulation using RNA sequencing (RNA-Seq V2) data of 100 LGGs from TCGA. The survival analysis of significant DEGs in glioma using TCGA database was determined. Kaplan–Meier curves were used to produce graphs showing the survival probability of prognostic signature genes of glioma and glioblastoma and their statistical significance. For example, p values of less than 0.05 in all tests were significantly linked to low-grade glioma and glioblastoma. The gene expression profiles of the prognostic signatures (KLF15, AQP7, and AGPAT9) were determined using TIMER immune 2.0.

Results

Kaplan–Meier curve showing the expression of KLF 15, AQP7, and AGPAT9 gene in glioma

Herein, we unveiled the survival analysis for glioma patients using the TCGA database and Kaplan–Meier plots and discovered that the KLF 15 is significantly associated (p = 0.03) with the overall survival of the patient which indicates that it may be a very possible prognostic biomarker useful for glioma patients Fig. 1a. In our investigations, the Kaplan–Meier plot showed that AQP7 is significantly associated (p = 0.001) with overall survival of the glioma patients using the TCGA database Fig. 1b. Hence, it showed that it may be a prognostic biomarker which may be useful for the glioma patient. The Kaplan–Meier plot showed that AGPAT9 is significantly associated (p = 0.005) with overall survival of the glioma patients using the TCGA database (Fig. 1c).

Fig. 1
figure 1

Kaplan–Meier curve showing the expression of KLF 15, AQP7, and AGPAT9 gene in glioma

Kaplan–Meier curve showing the expression of KLF 15, AQP7, and AGPAT9 gene in glioblastoma

This is a visual representation of expression level of prognostic signature KLF15, which indicates that the p value is 0.025 that means it has a significant association with glioblastoma and thus can be used as a prognostic signature in the early detection of glioblastoma (Fig. 2a). Meanwhile, the expression level of prognostic signature AQP7 has p value of 0.59 that means it has no significant association with glioblastoma and thus cannot be used as a prognostic signature in the early detection for glioblastoma patients (Fig. 2b). Also, AGPAT9 has a p value of 0.10 that means it has no significant association with glioblastoma and thus cannot be used as a prognostic signature in the early detection of glioblastoma. Thus, AQP7 and AGPAT9 have no significant association and so they are predictive biomarkers and may not be potential prognostic signatures for glioblastoma patients. However, KLF15 showed a significant association with glioblastoma and so can be a prognostic biomarker in glioblastoma patients.

Fig. 2
figure 2

Kaplan–Meier curve showing the expression of KLF 15, AQP7, and AGPAT9 gene in glioblastoma

The stromal, immune, and estimate scores of low-grade glioma

To analyze the immune infiltration landscape in LGGs, single-sample gene set enrichment analysis (ssGSEA) was applied to evaluate the relative abundance of each immune cell subpopulation using RNA sequencing (RNA-Seq V2) data of 100 LGGs patients from The Cancer Genome Atlas (TCGA). By performing single-sample gene set enrichment analysis (ssGSEA), we calculated stromal and immune scores to predict the level of infiltrating stromal and immune cells and these form the basis for the ESTIMATE score to infer tumor purity in tumor tissue Fig. 3.

Fig. 3
figure 3

The stromal, immune, and estimate scores of low-grade glioma

The expression levels of KLF15 in LGG using different immune infiltrate variables

KLF15 has a correlation with low-grade glioma. Correlation value of 0.124 and the genes are highly expressed in the tumor cells which show a positive correlation with tumor purity and significantly association (p = 0.000005) Fig. 4. B cell, CD8+ T cell, CD4+ T cell, macrophages, neutrophil, and dendritic cells are the immune infiltrates which show that the expression level of KLF15 has a partial correlation, and it is significantly associated with immune infiltration in all the cells except that of the macrophages where the P value is 0.07.

Fig. 4
figure 4

The expression levels of KLF15 in LGG using different immune infiltrate variables

The expression levels of AQP7 in LGG using different immune infiltrate variables

Based on investigations, it shows that the tumor purity of AQP7 has a negative correlation with low-grade glioma; correlation value = − 0.007 (Fig. 5). Also, purity and B cells do not show a significant association with glioma; p values = 0.8, 0.4, respectively. CD8+ T cell, CD4+ T cell, macrophages, neutrophil, and dendritic cells show that the expression level of AQP7 has a partial correlation with the infiltration level and significantly associated. CD8+ T cell, CD4+ T cell, macrophages, neutrophil, and dendritic cells also show a significant association with glioma with p values of 0.006, 0.05, 0.003, 0.003, and 0.01, respectively.

Fig. 5
figure 5

The expression levels of AQP7 in LGG using different immune infiltrate variables

The expression levels of AGPAT9 IN LGG using different immune infiltrate variables

Based on the investigation, it also shows that tumor purity of AGPAT9 has a negative correlation with low-grade glioma; correlation value = − 0.238, and it shows a significant association with the tumor purity with p value (p = 0.000000014) (Fig. 6). All the immune cells have positive correlation and show significant association with glioma except macrophages.

Fig. 6
figure 6

The expression levels of AGPAT9 IN LGG using different immune infiltrate variables

The expression levels of KLF15 in GBM using different immune infiltrate variables

The dendritic cells, CD4 T cell, neutrophil, and CD8+ T cell immune infiltrate are significantly associated with glioblastoma. Meanwhile, B cell, CD8+ T cell, CD4+ T cell, macrophages, neutrophil, and dendritic cells immune infiltrates indicated that KLF15 expression level is partially correlated with immune infiltration level in GBM, and purity immune infiltrate is correlated. Herein, during the analysis of the KLF15, we realized that the immune infiltrates are significantly association except that of the B cells (p = 0.1) and the macrophages (p = 0.3). The correlation value was 0.254 and the genes are highly expressed in the tumor cells. In the tumor purity, it shows positive correlation and significantly associated (p = 0.00000013) (Fig. 7) in glioblastoma.

Fig. 7
figure 7

The expression levels of KLF15 in GBM using different immune infiltrate variables

The expression levels of AQP7 in GBM using different immune infiltrate variables

Here, no significant association (p = 0.26). It was also indicated that the tumor purity of AQP7 has a negative correlation with its immune infiltration level; Correlation value = − 0.055 (Fig. 8). However, neutrophil and CD4 T cells have partial correlation and significantly associated in glioblastoma patients with p value of 0.0000195 and 0.0000177, respectively.

Fig. 8
figure 8

The expression levels of AQP7 in GBM using different immune infiltrate variables

The expression levels of AGPAT9 in GBM using different immune infiltrate variables

B cells and macrophages show no significant association with glioblastoma multiforme. The purity, CD8+ T cell, CD4+ T cell, neutrophil, and dendritic cells show a significant association with glioblastoma multiforme. Based on the analysis, the tumor purity of AGPAT9 has a negative correlation with low-grade glioma; correlation value = − 0.363. It also shows a significant association p = 0.000012 (Fig. 9).

Fig. 9
figure 9

The expression levels of AGPAT9 in GBM using different immune infiltrate variables

Discussions

Krüppel-like factor 15 (KLF15) is a signature that is yet to be fully elucidated in the glioma patient, but previous investigations have been done in the area of clear cell renal cell carcinoma [66] adenocarcinoma lung cancer [67]. Krüppel-like factor 15 (KLF15) is useful in a lot of biological processes which include cell proliferation, cell cycle, adipogenesis, etc. [68,69,70]. KLF15 has an important role in RNA polymerase II-specific DNA-binding transcription factor activity [71, 72]. Hence, it is known to have significant functions in different types of cancer. KLF15 is responsible for the suppression and activation of genes in carcinogenesis. Previous investigation has shown that KLF15 is a positive regulator of carcinogenesis [73,74,75,76]. Therefore, KLF15 may be useful immune-related prognostic signature in glioma and glioblastoma patients.

Investigations have been conducted on the Aquaporin 7 (AQP7) association with lymphatic metastasis, breast cancer, liver cancer, and clear renal cancer [77,78,79,80,81]. It is otherwise known as water channels which have been known to be related to the invasion, proliferation, and migration of human breast tumors [77, 82,83,84]. However, investigations are yet to discover the potential roles of AQP7 in glioma and glioblastoma patients as a possible therapeutic target and prognostic biomarker. Aquaporin (AQP) family members were first investigated in 1992 [85,86,87]. Various investigations have shown that it can be expressed in epithelial and non-epithelial cells [88]. AQP7 is also important in fatty acid metabolism and enhances the migration of water and glycerol [78]. Human 1-acylglycerol-3-phosphate O-acyltransferase 9 (AGPAT9, also known as GPAT3 or LPCAT1) is correlated with tumor progression and tumor microenvironment [89]. It is related to fatty acid metabolisms and involved in a lot of biological processes. It catalyzes de novo synthesis of triacylglycerol [89]. Hence, AQP7 and AGPAT9 indicate usefulness as prognostic biomarker which may be advantageous for the glioma patient.

Stromal and immune scores were estimated from transcriptomic profiles of LGG cohort from TCGA using the ESTIMATE. One hundred entries of TCGA cohort were entered and used for the investigation. Hence, the presence of stromal scores in tumor tissues, immune scores for the infiltration of immune cells in tumor tissues and the ESTIMATE scores that infers tumor purity is observed [90,91,92].

Immune infiltration of malignancies correlates strongly with clinical outcomes. In terms of chemotherapy and immunotherapy, the makeup of tumor-infiltrating immune cells (TIICs) can serve as biomarkers for predicting treatment response and survival in distinct patient subgroups [93]. Hence, the immune cell proportion of the three-signature for LGG were determined using Timer immune 2.0. The Gene module allows a user to identify the relationship between tumor gene expression and immune infiltration in a fast, comprehensive, and unbiased way [94]. Therefore, the signatures may be a potential prognostic signature for glioma and useful for screening immunotherapy for glioma patients. Hence, this is in consistent with previous investigations [95, 96]. Therefore, B cell, CD8+ T cell, CD4+ T cell, macrophages, Neutrophil, and dendritic cells immune infiltrates indicated that AQP7 expression level is partially correlated with immune infiltration level in LGG [97], while purity infiltrate is correlated. Hence, it may be a potential prognostic signature for glioma and useful for screening immunotherapy for glioma patients [98].

B cell, CD8+ T cell, CD4+ T cell, macrophages, neutrophil, and dendritic cells show that the expression level of AGPAT9 has a partial correlation with the infiltration level. Purity, B cell, CD8+ T cell, CD4+ T cell, neutrophil, and dendritic cells show a significant association with glioma [99], while macrophages do not have any significant association with glioma. Therefore, B cell, CD8+ T cell, CD4+ T cell, macrophages, neutrophil, and dendritic cells immune infiltrates indicated that AGPAT9 expression level is partially correlated with immune infiltration level in low-grade glioma, while purity immune infiltrate is correlated [100].

Determination of immune cell proportion of the KLF15, AQP7, and AGPAT9 signatures on the glioblastoma multiforme prognostic using Timer immune 2.0 indicated that the tumor purity of KLF15 has a positive correlation with glioma and glioblastoma [101]. Hence, KLF 15 may be a potential prognostic biomarker and useful for screening immunotherapy for glioma and glioblastoma patients. B cell, CD8+ T cell, CD4+ T cell, macrophages, neutrophil, and dendritic cells show that the expression level of KLF15 has a partial correlation with the infiltration level. Therefore, CD8+ T cell, CD4+ T cell, neutrophil, and dendritic cells immune infiltrates indicated that KLF15 expression level is significantly associated with the GBM patients. Thus, KLF15 may be a useful signature for monitoring immunotherapy in GBM [102].

B cell, CD8+ T cell, CD4+ T cell, macrophages, neutrophil, and dendritic cells show that the expression level of AGPAT9 has a partial correlation with the infiltration level. CD4+ T cell, macrophages, and neutrophil cells do not show a significant association with glioblastoma multiforme. The tumor purity, dendritic cells, and CD8+ T cell immune infiltrate have a significant association with glioblastoma using the AGPAT9 gene [66]. Therefore, B cell, CD8+ T cell, CD4+ T cell, macrophages, neutrophil, and dendritic cells immune infiltrates indicated that AGPAT9 expression level is partially correlated with immune infiltration level in GBM [89], while purity immune infiltrate is correlated. Dendritic cells are known for their ability of promoting tumor immunosuppression [103]. Dendritic cells are divided into two forms, myeloid DC and plasmacytoid DC, which can produce large amount of Interferon gamma [104]. It can also induce T cell immunity or tolerance [105, 106]. Hence, AGPT9 may be useful for monitoring immunotherapy in glioblastoma patients. Concerning the association of CD8 T cells, it shows that CD8+ T lymphocytes are crucial components of the tumor-specific adaptive immunity that attacks tumor cells [107]. Clinical outcomes are highly connected to the immune infiltration of malignancies [108]. The composition of tumor-infiltrating immune cells (TIICs) may serve as biomarkers for predicting treatment response and survival in various patients subgroups in terms of chemotherapy and immunotherapy [109, 110].

Conclusions

The analysis revealed that KLF15, AQP7, and AGPAT9 may be prognostic biomarker genes that may be useful for prognosis of patients with glioma. Utilization of bioinformatics tools such as TIMER, ESTIMATE, Kaplan–Meier plot, TCGA database; the immune proportion, stromal and immune scores, various expression levels of the prognostic signatures, infiltrating levels, and tumor purity of glioma and glioblastoma multiforme were determined. Further investigations will be required using X-tile software, Database for Annotation, Visualization, and Integrated Discovery (DAVID), string, cytoscape, Kyoto Encyclopedia of Genes and Genomes (KEGGs) databases to unveil the molecular mechanisms of glioma and glioblastoma. Use of single cell sequencing will be of great usefulness in the treatment of glioma and glioblastoma. Investigations into hormone-based therapy will be fascinating. It would be enormously fascinating to validate maybe the biomarker predicts both precision immunotherapy and prognosis. The determination of real-time quantitative PCR analysis is also important.

Availability of data and materials

The data that support the findings of this study are available from the corresponding author, upon reasonable request http://xena.ucsc.edu/, TCGA database.

Abbreviations

TCGA:

The Cancer Genome Atlas

KLF15:

Krüppel-like factor 15

AQP7:

Aquaporin 7

AGPAT9:

Human 1-acylglycerol-3-phosphate O-acyltransferase 9

LGG:

Low-grade glioma

GBM:

Glioblastoma

ESTIMATE:

Estimation of Stromal and Immune cells in Malignant Tumors using Expression data algorithm

TIICs:

Tumor-infiltrating immune cells

ssGSEA:

Single-sample gene set enrichment analysis.

DEG:

Differentially Expressed Genes

DAVID:

Database for Annotation, Visualization, and Integrated Discovery

KEGGs:

Encyclopedia of Genes and Genomes

TME:

Tumor microenvironment

TIMER:

Tumor IMmune Estimation Resource

References

  1. Aoki K, Nakamura H, Suzuki H, Matsuo K, Kataoka K, Shimamura T et al (2018) Prognostic relevance of genetic alterations in diffuse lower-grade gliomas. Neuro-Oncol 20(1):66–77. https://doi.org/10.1093/neuonc/nox132

    Article  CAS  PubMed  Google Scholar 

  2. Salari N, Ghasemi H, Fatahian R, Mansouri K, Dokaneheifard S, Shiri MH et al (2023) The global prevalence of primary central nervous system tumors: a systematic review and meta-analysis. Eur J Med Res 28(1):39. https://doi.org/10.1186/s40001-023-01011-y

    Article  PubMed  PubMed Central  Google Scholar 

  3. Kessler T, Ito J, Wick W, Wick A (2023) Conventional and emerging treatments of astrocytomas and oligodendrogliomas. J Neurooncol 162(3):471–478. https://doi.org/10.1007/s11060-022-04216-z

    Article  PubMed  Google Scholar 

  4. Song L-R, Weng J-C, Li C-B, Huo X-L, Li H, Hao S-Y et al (2020) Prognostic and predictive value of an immune infiltration signature in diffuse lower-grade gliomas. JCI insight 5(8):e133811. https://doi.org/10.1172/jci.insight.133811

    Article  PubMed  PubMed Central  Google Scholar 

  5. Liang X, Wang Z, Dai Z, Zhang H, Cheng Q, Liu Z (2021) Promoting prognostic model application: a review based on gliomas. J Oncol 2021:1–14. https://doi.org/10.1155/2021/7840007

    Article  CAS  Google Scholar 

  6. Luo J, Pan M, Mo K, Mao Y, Zou D (2023) Emerging role of artificial intelligence in diagnosis, classification, and clinical management of glioma. Sem Cancer Bio 91:110–123. https://doi.org/10.1016/j.semcancer.2023.03.006

    Article  CAS  Google Scholar 

  7. Aiman W, Gasalberti DP, Rayi A. Low-Grade Gliomas (2023). StatPearls [Internet] 1–10

  8. Hu Y, Yang Q, Cai S, Wang W, Fu S (2023) The integrative analysis based on super-enhancer related genes for predicting different subtypes and prognosis of patient with lower-grade glioma. Front Gent 14:1085584. https://doi.org/10.3389/fgene.2023.1085584

    Article  CAS  Google Scholar 

  9. Jiang H, Zhu Q, Wang X, Li M, Shen S, Yang C et al (2023) Characterization and clinical implications of different malignant transformation patterns in diffuse low-grade gliomas. Cancer Sci 114(9):3708–3718. https://doi.org/10.1111/cas.15889

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  10. Nakasu S, Nakasu Y, Tsuji A, Fukami T, Nitta N, Kawano H et al (2023) Incidental diffuse low-grade gliomas: a systematic review and meta-analysis of treatment results with correction of lead-time and lengthy-time biases. Neurooncol Pract 10(2):113–125. https://doi.org/10.1093/nop/npac073

    Article  PubMed  Google Scholar 

  11. van den Bent M (2023) Thirty years of progress in the management of low-grade gliomas. Revue Neurologique 5:425–429. https://doi.org/10.1016/j.neurol.2023.03.001

    Article  Google Scholar 

  12. Melnyk T, Masiá E, Zagorodko O, Conejos-Sánchez I, Vicent MJ (2023) Rational design of poly-L-glutamic acid-palbociclib conjugates for pediatric glioma treatment. J Control Release 355:385–394. https://doi.org/10.1016/j.jconrel.2023.01.079

    Article  CAS  PubMed  Google Scholar 

  13. Kupfer SS, Ellis NA (2017) Hereditary colorectal cancer. In: The molecular basis of human cancer, pp 381–400. https://doi.org/10.1053/j.scrs.2010.12.002

  14. Chen H-L, Hsu F-T, Kao Y-CJ, Liu H-S, Huang W-Z, Lu C-F et al (2017) Identification of epidermal growth factor receptor-positive glioblastoma using lipid-encapsulated targeted superparamagnetic iron oxide nanoparticles in vitro. J Nanobiotechnol 15(1):1–13. https://doi.org/10.1186/s12951-017-0313-2

    Article  CAS  Google Scholar 

  15. Pickering L, Main KM, Feldt-Rasmussen U, Sehested A, Mathiasen R, Klose M et al (2023) Survival and long-term socioeconomic consequences of childhood and adolescent onset of brain tumors. Dev Med Child Neurol 65(7):942–952. https://doi.org/10.1111/dmcn.15467

    Article  PubMed  Google Scholar 

  16. Ji J, Huh Y, Ji R-R (2023) Immune and glial cells in pain and their interactions with nociceptive neurons. In: Neuroimmune interact pain: mech and therap. Springer, Berlin, pp 121–51. https://doi.org/10.1007/978-3-031-29231-6-5

  17. Shi DD, Anand S, Abdullah KG, McBrayer SK (2023) DNA damage in IDH-mutant gliomas: mechanisms and clinical implications. J Neuro-Oncol 162(3):515–523. https://doi.org/10.1007/s11060-022-04172-8

    Article  CAS  Google Scholar 

  18. Navickas SM, Giles KA, Brettingham-Moore KH, Taberlay PC (2023) The role of chromatin remodeler SMARCA4/BRG1 in brain cancers: a potential therapeutic target. Oncogene. https://doi.org/10.1038/s41388-023-02773-9

    Article  PubMed  PubMed Central  Google Scholar 

  19. Ajuyah P, Mayoh C, Lau LM, Barahona P, Wong M, Chambers H et al (2023) Histone H3-wild type diffuse midline gliomas with H3K27me3 loss are a distinct entity with exclusive EGFR or ACVR1 mutation and differential methylation of homeobox genes. Sci Rep 3(1):3775. https://doi.org/10.1038/s41598-023-30395-4

    Article  ADS  CAS  Google Scholar 

  20. O’Donohue T, Farouk Sait S, Glade Bender J (2023) Progress in precision therapy in pediatric oncology. Curr Opin Ped 35(1):41–47. https://doi.org/10.1097/MOP.0000000000001198

    Article  Google Scholar 

  21. Sun Y, Liu Z, Fu Y, Yang Y, Lu J, Pan M et al (2023) Single-cell multi-omics sequencing and its application in tumor heterogeneity. Brief Func Gen 22(4):313–328. https://doi.org/10.1093/bfgp/elad009

    Article  CAS  Google Scholar 

  22. Broggi G, Salvatorelli L, Barbagallo D, Certo F, Altieri R, Tirrò E et al (2021) Diagnostic utility of the immunohistochemical expression of serine and arginine rich splicing factor 1 (SRSF1) in the differential diagnosis of adult gliomas. Cancers 3(9):2086. https://doi.org/10.3390/cancers13092086

    Article  CAS  Google Scholar 

  23. Sisakht AK, Malekan M, Ghobadinezhad F, Firouzabadi SNM, Jafari A, Mirazimi SMA et al (2023) Cellular conversations in glioblastoma progression, diagnosis, and treatment. Cell Mol Neurobio 43(2):585–603. https://doi.org/10.1007/s10571-022-01212-9

    Article  Google Scholar 

  24. Elshaer SS, Abulsoud AI, Fathi D, Abdelmaksoud NM, Zaki MB, El-Mahdy HA et al (2023) miRNAs role in glioblastoma pathogenesis and targeted therapy: Signaling pathways interplay. Pathol Res Pract. https://doi.org/10.1016/j.prp.2023.154511

    Article  PubMed  Google Scholar 

  25. Sharma A, Guerrero-Cázares H, Maciaczyk J (2023) Editorial to special issue “glioblastoma: recapitulating the key breakthroughs and future perspective”. MDPI, p 2548. https://doi.org/10.1186/s40478-023-01605-x

  26. Loras A, Gonzalez-Bonet LG, Gutierrez-Arroyo JL, Martinez-Cadenas C, Marques-Torrejon MA (2023) Neural stem cells as potential glioblastoma cells of origin. Life 13(4):905. https://doi.org/10.3390/life13040905

    Article  ADS  CAS  PubMed  PubMed Central  Google Scholar 

  27. Ah-Pine F, Khettab M, Bedoui Y, Slama Y, Daniel M, Doray B, Gasque P (2023) On the origin and development of glioblastoma: multifaceted role of perivascular mesenchymal stromal cells. Acta Neuropathol Commun 11(1):1–15. https://doi.org/10.1186/s40478-023-01605-x

    Article  CAS  Google Scholar 

  28. Colopi A, Fuda S, Santi S, Onorato A, Cesarini V, Salvati M et al (2023) Impact of age and gender on glioblastoma onset, progression, and management. Mech Ageing Dev 211:111801. https://doi.org/10.1016/j.mad.2023.111801

    Article  PubMed  Google Scholar 

  29. Shobeiri P, Seyedmirzaei H, Kalantari A, Mohammadi E, Rezaei N, Hanaei S (2023) The epidemiology of brain and spinal cord tumors. In: Human brain and spinal cord tumors: from bench to bedside volume 1: neuroimmunology and neurogenetics. Springer, Berlin, pp 19–39. https://doi.org/10.1007/978-3-031-14732-6-2

  30. Sahrizan NSA, Manan HA, Abdul Hamid H, Abdullah JM, Yahya N (2023) Functional alteration in the brain due to tumour invasion in paediatric patients: a systematic review. Cancers 15(7):2168. https://doi.org/10.3390/cancers15072168

    Article  PubMed  PubMed Central  Google Scholar 

  31. Arnold LM, DeWitt JC, Thomas AA (2023) Tumors of the nervous system. In: Neurobiology of brain disorders. Elsevier, New York, pp 203–228

  32. Datta S, Jones LD, Pingle S (2023) Brain tumors: focus on glioblastoma. Dis Rev 1–22

  33. Bao H, Ren P, Yi L, Lv Z, Ding W, Li C et al (2023) New insights into glioma frequency maps: from genetic and transcriptomic correlate to survival prediction. Int J Cancer 152(5):998. https://doi.org/10.1002/ijc.34336

    Article  CAS  PubMed  Google Scholar 

  34. Goodenberger ML, Jenkins RB (2012) Genetics of adult glioma. Cancer Genet 205(12):613–621. https://doi.org/10.1016/j.cancergen.2012.10.009

    Article  CAS  PubMed  Google Scholar 

  35. Louis DN, Perry A, Reifenberger G, Von Deimling A, Figarella-Branger D, Cavenee WK et al (2016) The 2016 World Health Organization classification of tumors of the central nervous system: a summary. Acta Neuropathol 131:803–820. https://doi.org/10.1007/s00401-016-1545-1

    Article  PubMed  Google Scholar 

  36. Esparragosa Vazquez I, Ndiaye M, Di Stefano AL, Younan N, Larrieu-Ciron D, Seyve A et al (2023) FLAIR pseudoprogression in patients with anaplastic oligodendrogliomas treated with PCV chemotherapy alone. Euro J Neurol. https://doi.org/10.1111/ene.15873

    Article  Google Scholar 

  37. Bernabéu-Sanz Á, Fuentes-Baile M, Alenda C (2021) Main genetic differences in high-grade gliomas may present different MR imaging and MR spectroscopy correlates. Eur Radiol 31:749–763. https://doi.org/10.1007/s00330-020-07138-4

    Article  PubMed  Google Scholar 

  38. Balana C, Castañer S, Carrato C, Moran T, Lopez-Paradís A, Domenech M et al (2022) Preoperative diagnosis and molecular characterization of gliomas with liquid biopsy and radiogenomics. Front Neurol 13:865171. https://doi.org/10.3389/fneur.2022.865171

    Article  PubMed  PubMed Central  Google Scholar 

  39. Zhang J, Peng H, Wang Y-L, Xiao H-F, Cui Y-Y, Bian X-B et al (2021) Predictive role of the apparent diffusion coefficient and MRI morphologic features on IDH status in patients with diffuse glioma: a retrospective cross-sectional study. Front Oncol 11:640738. https://doi.org/10.3389/fonc.2021.640738

    Article  PubMed  PubMed Central  Google Scholar 

  40. Li M, Wang J, Chen X, Dong G, Zhang W, Shen S et al (2023) The sinuous, wave-like intratumoral-wall sign is a sensitive and specific radiological biomarker for oligodendrogliomas. Euro Radiol 33(6):4440–4452. https://doi.org/10.1007/s00330-022-09314-0

    Article  CAS  Google Scholar 

  41. Fisher BJ, Hu C, Macdonald DR, Lesser GJ, Coons SW, Brachman DG et al (2015) Phase 2 study of temozolomide-based chemoradiation therapy for high-risk low-grade gliomas: preliminary results of Radiation Therapy Oncology Group 0424. Int J Radiat Oncol Biol Phys 91(3):497–504. https://doi.org/10.1016/j.ijrobp.2014.11.012

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  42. Wang Y, Wahafu A, Wu W, Xiang J, Huo L, Ma X et al (2021) FABP5 enhances malignancies of lower-grade gliomas via canonical activation of NF-κB signaling. J Cell Mol Med 9:4487–4500. https://doi.org/10.1111/jcmm.16536

    Article  CAS  Google Scholar 

  43. Kumari S, Gupta R, Ambasta RK, Kumar P (2023) Multiple therapeutic approaches of glioblastoma multiforme: from terminal to therapy. Biochim Biophys Acta Rev Cancer. https://doi.org/10.1016/j.bbcan.2023.188913

    Article  PubMed  Google Scholar 

  44. Steyaert S, Qiu YL, Zheng Y, Mukherjee P, Vogel H, Gevaert O (2023) Multimodal deep learning to predict prognosis in adult and pediatric brain tumors. Commun Med Lond 3(1):44. https://doi.org/10.1038/s43856-023-00276-y

    Article  PubMed  PubMed Central  Google Scholar 

  45. Cui X, Wang Y, Zhou J, Wang Q, Kang C (2023) Expert opinion on translational research for advanced glioblastoma treatment. Cancer Biol Med 20(5):344. https://doi.org/10.20892/j.issn.2095-3941.2023.0012

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  46. Dhungel L, Harris C, Romine L, Sarkaria J, Raucher D (2023) Targeted c-Myc inhibition and systemic temozolomide therapy extend survival in glioblastoma xenografts. Bioenginering 10(6):718. https://doi.org/10.3390/bioengineering10060718

    Article  CAS  Google Scholar 

  47. Hu Y, Li Z, Zhang Y, Wu Y, Liu Z, Zeng J et al (2023) The evolution of tumor microenvironment evolution in gliomas and its implication for target therapy. Int J Biol Sci 19(13):4311–4326. https://doi.org/10.7150/ijbs.83531

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  48. Siminzar P, Tohidkia MR, Eppard E, Vahidfar N, Tarighatnia A, Aghanejad A (2023) Recent trends in diagnostic biomarkers of tumor microenvironment. Mol Imaging Biol 25(3):464–482. https://doi.org/10.1007/s11307-022-01795-11

    Article  PubMed  Google Scholar 

  49. Neophytou CM, Panagi M, Stylianopoulos T, Papageorgis P (2021) The role of tumor microenvironment in cancer metastasis: Molecular mechanisms and therapeutic opportunities. Cancers 13(9):2053. https://doi.org/10.3390/cancers13092053

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  50. Francescangeli F, De Angelis ML, Rossi R, Cuccu A, Giuliani A, De Maria R, Zeuner A (2023) Dormancy, stemness, and therapy resistance: interconnected players in cancer evolution. Cancer Metastasis Rev 42(1):197–215. https://doi.org/10.1007/s10555-023-10092-4

    Article  PubMed  PubMed Central  Google Scholar 

  51. van Weverwijk A, de Visser KE (2023) Mechanisms driving the immunoregulatory function of cancer cells. Nat Rev Cancer 23(4):193–215. https://doi.org/10.1038/s41568-022-00544-4

    Article  CAS  PubMed  Google Scholar 

  52. Margetis AT (2023) Metabolic targeting of malignant tumors: a need for systemic approach. J Cancer Res Clin Oncol 149(5):2115–2138. https://doi.org/10.1007/s00432-022-04212-w

    Article  PubMed  Google Scholar 

  53. Wang S, Yan W, Kong L, Zuo S, Wu J, Zhu C et al (2023) Oncolytic viruses engineered to enforce cholesterol efflux restore tumor-associated macrophage phagocytosis and anti-tumor immunity in glioblastoma. Nat Commun 14(1):4367. https://doi.org/10.1038/s41467-023-39683-z

    Article  ADS  CAS  PubMed  PubMed Central  Google Scholar 

  54. Wu B, Shi X, Jiang M, Liu H (2023) Crosstalk between cancer stem cells and immune cells: potential therapeutic targets in the tumor immune microenvironment. Mol Cancer 22(1):1–22. https://doi.org/10.1186/s12943-023-01748-4

    Article  Google Scholar 

  55. Kumari S, Kumar S, Muthuswamy S (2023) RNA N6-methyladenosine modification in regulating cancer stem cells and tumor immune microenvironment and its implication for cancer therapy. J Cancer Res Clin Oncol 149(4):1621–1633. https://doi.org/10.1007/s00432-022-04158-z

    Article  CAS  PubMed  Google Scholar 

  56. Luo H, Zhang H, Mao J, Cao H, Tao Y, Zhao G et al (2023) Exosome-based nanoimmunotherapy targeting TAMs, a promising strategy for glioma. Cell Death Dis 14(4):235. https://doi.org/10.1038/s41419-023-05753-9

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  57. Yang J, Xu J, Wang W, Zhang B, Yu X, Shi S (2023) Epigenetic regulation in the tumor microenvironment: molecular mechanisms and therapeutic targets. Signal Transduct Target Ther 8(1):210. https://doi.org/10.1038/s41392-023-01480-x

    Article  PubMed  PubMed Central  Google Scholar 

  58. Li Z-S, Wang B, Liang H, Li Y, Zhang Z-Y, Han L (2023) A three-stage eccDNA-based molecular profiling significantly improves the identification, prognosis assessment and recurrence prediction accuracy in patients with glioma. Cancer Lett. https://doi.org/10.1016/j.canlet.2023.216369

    Article  PubMed  Google Scholar 

  59. Karabacak M, Ozkara BB, Senparlak K, Bisdas S (2023) Deep learning-based radiomics for prognostic stratification of low-grade gliomas using a multiple-gene signature. Appl Sci 13(6):3873. https://doi.org/10.3390/app13063873

    Article  CAS  Google Scholar 

  60. Lee M (2023) Deep learning techniques with genomic data in cancer prognosis: a comprehensive review of the 2021–2023 literature. Biology 12(7):893. https://doi.org/10.3390/biology12070893

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  61. Choi SR, Lee M (2023) Transformer architecture and attention mechanisms in genome data analysis: a comprehensive review. Biology 12(7):1033. https://doi.org/10.3390/biology12071033

    Article  PubMed  PubMed Central  Google Scholar 

  62. Li S, Wang C, Chen J, Lan Y, Zhang W, Kang Z et al (2023) Signaling pathways in brain tumors and therapeutic interventions. Signal Transduct Target Ther 8(1):8. https://doi.org/10.1038/s41392-022-01260-z

    Article  PubMed  PubMed Central  Google Scholar 

  63. Zhou X, Jin G, Zhang J, Liu F (2023) Recruitment mechanisms and therapeutic implications of tumor-associated macrophages in the glioma microenvironment. Front Immunol 14:1067641. https://doi.org/10.3389/fimmu.2023.1067641

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  64. Arrieta VA, Dmello C, McGrail DJ, Brat DJ, Lee-Chang C, Heimberger AB et al (2023) Immune checkpoint blockade in glioblastoma: from tumor heterogeneity to personalized treatment. J Clin Invest 133(2):e163447. https://doi.org/10.1172/JCI163447

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  65. Cruz N, Herculano-Carvalho M, Roque D, Faria CC, Cascão R, Ferreira HA et al (2023) Highlighted advances in therapies for difficult-to-treat brain tumours such as glioblastoma. Pharmaceutics 15(3):928. https://doi.org/10.3390/pharmaceutics15030928

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  66. Xu W-H, Xu Y, Wang J, Wan F-N, Wang H-K, Cao D-L et al (2019) Prognostic value and immune infiltration of novel signatures in clear cell renal cell carcinoma microenvironment. Aging (Albany NY) 11(17):6999–7020. https://doi.org/10.18632/aging.102233

    Article  CAS  PubMed  Google Scholar 

  67. Zhu Z, Yu Z, Wang J, Zhou L, Zhang J, Yao B et al (2018) Krüppel-like factor 4 inhibits pancreatic cancer epithelial-to-mesenchymal transition and metastasis by down-regulating caveolin 1 expression. Cell Physiol Biochem 46(1):23852. https://doi.org/10.1159/000488426

    Article  CAS  Google Scholar 

  68. Chang K, Xue R, Zhao M, Zhao Y, Yu W, Zhao Z, Liu X (2021) EP300/CBP is crucial for cAMP-PKA pathway to alleviate podocyte dedifferentiation via targeting Notch3 signaling. Exp Cell Res 407(2):112825. https://doi.org/10.1016/j.yexcr.2021.112825

    Article  CAS  PubMed  Google Scholar 

  69. Mallipattu SK, Liu R, Zheng F, Narla G, Ma’ayan A, Dikman S et al (2012) Krüppel-like factor 15 (KLF15) is a key regulator of podocyte differentiation. J Biol Chem 287(23):19122–19135. https://doi.org/10.1074/jbc.M112.345983

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  70. Khatibi SMH, Ardalan M, Abediazar S, Vahed SZ (2020) The impact of steroids on the injured podocytes in nephrotic syndrome. J Steroid Biochem Mol Biol 196:105490. https://doi.org/10.1016/j.jsbmb.2019.105490

    Article  CAS  Google Scholar 

  71. Zhang Y, Yao C, Ju Z, Jiao D, Hu D, Qi L et al (2023) Krüppel-like factors in tumors: Key regulators and therapeutic avenues. Front Oncol 13:1080720. https://doi.org/10.3389/fonc.2023.1080720

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  72. Liu Y, Zhao X, Wang B, Liu Z, Zhang M, Wang J et al (2021) miR-376a provokes rectum adenocarcinoma via CTC1 depletion-induced telomere dysfunction. Front Cell Dev Biol 9:649328. https://doi.org/10.3389/fcell.2021.649328

    Article  PubMed  PubMed Central  Google Scholar 

  73. Wang J, Wang W, Zhu F, Duan Q (2022) The role of branched-chain amino acids metabolic disorders in tumorigenesis and progression. Biomed Pharmacother 153:113390. https://doi.org/10.1016/j.biopha.2022.113390

    Article  CAS  PubMed  Google Scholar 

  74. He Z, Wang C, Xue H, Zhao R, Li G (2020) Identification of a metabolism-related risk signature associated with clinical prognosis in glioblastoma using integrated bioinformatic analysis. Front Oncol 10:1631. https://doi.org/10.3389/fonc.2020.01631

    Article  PubMed  PubMed Central  Google Scholar 

  75. Ray SK (2016) The transcription regulator Kruppel-like factor 4 and its dual roles of oncogene in glioblastoma and tumor suppressor in neuroblastoma. For Immunopathol Dis Therap 7(2):127–139. https://doi.org/10.1615/ForumImmunDisTher.2016017227

    Article  PubMed  PubMed Central  Google Scholar 

  76. Li J, Shen J, Zhao Y, Du F, Li M, Wu X et al (2023) Role of miR-181a-5p in cancer. Int J Oncol 63(4):1–18. https://doi.org/10.3892/ijo.2023.5556

    Article  CAS  Google Scholar 

  77. Zhu L, Ma N, Wang B, Wang L, Zhou C, Yan Y et al (2019) Significant prognostic values of aquaporin mRNA expression in breast cancer. Cancer Manag Res 11:1503–1515. https://doi.org/10.2147/CMAR.S193396

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  78. Abulizi A, Dawuti A, Yang B (2023) Aquaporins in tumor. Aquaporins Adv Exp Med Biol 1398:303–315. https://doi.org/10.1007/978-981-19-7415-1_21

    Article  CAS  PubMed  Google Scholar 

  79. Charlestin V, Fulkerson D, Arias Matus CE, Walker ZT, Carthy K, Littlepage LE (2022) Aquaporins: new players in breast cancer progression and treatment response. Front Oncol 12:988119. https://doi.org/10.3389/fonc.2022.988119

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  80. Chow PH, Bowen J, Yool AJ (2020) Combined systematic review and transcriptomic analyses of mammalian aquaporin classes 1 to 10 as biomarkers and prognostic indicators in diverse cancers. Cancers 2(7):1911. https://doi.org/10.3390/cancers12071911

    Article  CAS  Google Scholar 

  81. Kushwaha PP, Verma S, Gupta S (2023) Aquaporins as prognostic biomarker in prostate cancer. Cancers 15(2):331. https://doi.org/10.3390/cancers15020331

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  82. Jang SJ, Moon C (2023) Aquaporin 5 (AQP5) expression in breast cancer and its clinicopathological characteristics. PLoS ONE 18(1):e0270752. https://doi.org/10.1371/journal.pone.0270752

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  83. Edamana S, Login FH, Riishede A, Dam VS, Tramm T, Nejsum LN (2023) The cell polarity protein Scribble is downregulated by the water channel aquaporin-5 in breast cancer cells. Am J Physiol Cell Physiol 324(2):C307–C319. https://doi.org/10.1152/ajpcell.00311.2022

    Article  CAS  PubMed  Google Scholar 

  84. Bystrup M, Login FH, Edamana S, Borgquist S, Tramm T, Kwon TH, Nejsum LN (2022) Aquaporin-5 in breast cancer. APMIS 130(5):253–260. https://doi.org/10.1111/apm.13212

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  85. Zhou Y, Huang H, Zhou D, Li Q, Pan R, Yuan C, Cui Y (2023) Cloning and bioinformatics analysis of aquaporin proteins in the European house dust mite, Dermatophagoides pteronyssinus. Euro Zool J 90(1):344–353. https://doi.org/10.1080/24750263.2023.2205866

    Article  CAS  Google Scholar 

  86. Ahmed B, Alam M, Aktar N, Hossain MS, Ullah MW, Bashar KK et al (2023) Genome-wide investigation of aquaporin genes in Corchorus spp and their role in organ development and abiotic stress tolerance. Plant Gene 34:100410. https://doi.org/10.1016/j.plgene.2023.100410

    Article  CAS  Google Scholar 

  87. Zhou C, Lin Z-s, Shi Y, Feng J, Hu Z, Yang M-j et al (2023) Genome-wide identification, structural and evolutionary characteristics, and expression analysis of aquaporin gene family members in Mercenaria mercenaria. Front Marine Sci 10:1138074. https://doi.org/10.3389/fmars.2023.1138074

    Article  Google Scholar 

  88. Mutchler SM, Shi S, Whelan SCM, Kleyman TR (2023) Validation of commercially available antibodies directed against subunits of the epithelial Na+ channel. Physiol Rep 11(1):e15554. https://doi.org/10.14814/phy2.15554

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  89. Korbecki J, Bosiacki M, Gutowska I, Chlubek D, Baranowska-Bosiacka I (2023) Biosynthesis and significance of fatty acids, glycerophospholipids, and triacylglycerol in the processes of glioblastoma tumorigenesis. Cancers 15(7):2183. https://doi.org/10.3390/cancers15072183

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  90. Li J, Zhang S, Chen S, Yuan Y, Zuo M, Li T et al (2023) Lipid metabolism-related gene signature predicts prognosis and depicts tumor microenvironment immune landscape in gliomas. Front Immunol 14:1021678. https://doi.org/10.3389/fimmu.2023.1021678

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  91. Xiong Y, Xiong Z, Cao H, Li C, Wanggou S, Li X (2020) Multi-dimensional omics characterization in glioblastoma identifies the purity-associated pattern and prognostic gene signatures. Cancer Cell Int 20(1):1–12. https://doi.org/10.1186/s12935-020-1116-3

    Article  Google Scholar 

  92. Zhang S-H, Zeng N, Sun J-X, Liu C-Q, Xu J-Z, Xu M-Y et al (2023) Pan-cancer analysis reveals the prognostic and immunologic roles of cereblon and its significance for PROTAC design. Heliyon 29(6):e16644. https://doi.org/10.1016/j.heliyon.2023.e16644

    Article  Google Scholar 

  93. Xu H, Zhu Q, Tang L, Jiang J, Yuan H, Zhang A, Lou M (2021) Prognostic and predictive value of FCER1G in glioma outcomes and response to immunotherapy. Cancer Cell Int 21:1–16. https://doi.org/10.1186/s12935-021-01804-3

    Article  CAS  Google Scholar 

  94. Huang X, Tian T, Zhang Y, Zhou S, Hu P, Zhang J (2021) Age-associated changes in adverse events arising from anti-PD-(L) 1 therapy. Front Oncol 11:619385. https://doi.org/10.3389/fonc.2021.619385

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  95. Malta TM, Sokolov A, Gentles AJ, Burzykowski T, Poisson L, Weinstein JN et al (2018) Machine learning identifies stemness features associated with oncogenic dedifferentiation. Cell 173(2):338–54.e15. https://doi.org/10.1016/j.cell.2018.03.034.

  96. Liang Y, Li Z, Yuan H, Wang L (2021) Gao L-H (2021) Poly (p-phenylenevinylene) nanoparticles modified with antiEGFRvIII for specific glioblastoma therapy. Sci Rep 11(1):4449. https://doi.org/10.1038/s41598-021-83931-5

    Article  ADS  CAS  PubMed  PubMed Central  Google Scholar 

  97. Zhou Z, Zhan J, Cai Q, Xu F, Chai R, Lam K et al (2022) The water transport system in astrocytes-aquaporins. Cells 11(16):2564. https://doi.org/10.3390/cells11162564

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  98. da Silva IV, Garra S, Calamita G, Soveral G (2022) The multifaceted role of aquaporin-9 in health and its potential as a clinical biomarker. Biomolecules 12(7):897. https://doi.org/10.3390/biom12070897

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  99. Roma-Rodrigues C, Mendes R, Baptista PV, Fernandes AR (2019) Targeting tumor microenvironment for cancer therapy. Int J Mol Sci 20(4):840. https://doi.org/10.3390/ijms20040840

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  100. Mahajan S, Schmidt MH, Schumann U (2023) The glioma immune landscape: a double-edged sword for treatment regimens. Cancers 15(7):2024. https://doi.org/10.3390/cancers15072024

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  101. Simmen FA, Alhallak I, Simmen RC (2023) Krüppel-like factor-9 and Krüppel-like factor-13: highly related, multi-functional, transcriptional repressors and activators of oncogenesis. Cancers 15(23):5667. https://doi.org/10.3390/cancers15235667

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  102. Shu L, Tang J, Liu S, Tao Y (2023) Plasma cell signatures predict prognosis and treatment efficacy for lung adenocarcinoma. Cell Oncol. https://doi.org/10.1007/s13402-023-00883-w

    Article  Google Scholar 

  103. Shbeer AM (2023) Current state of knowledge and challenges for harnessing the power of dendritic cells in cancer immunotherapy. Pathol-Res Pract. https://doi.org/10.1016/j.prp.2023.155025

    Article  PubMed  Google Scholar 

  104. Hornero RA, Idoyaga J (2023) Plasmacytoid dendritic cells: a dendritic cell in disguise. Mol Immunol 159:38–45. https://doi.org/10.1016/j.molimm.2023.05.007

    Article  CAS  PubMed Central  Google Scholar 

  105. Dey M, Chang AL, Miska J, Wainwright DA, Ahmed AU, Balyasnikova IV et al (2015) Dendritic cell–based vaccines that utilize myeloid rather than plasmacytoid cells offer a superior survival advantage in malignant glioma. J Immunol 195(1):367–376. https://doi.org/10.4049/jimmunol.1401607

    Article  CAS  PubMed  Google Scholar 

  106. Zhang Y (ed) (2024) Dendritic cell vaccine in cancer immunotherapy. In: Third international conference on biological engineering and medical science (ICBioMed2023) SPIE. https://doi.org/10.1007/s11684-011-0172-4

  107. Hwang HS, Shin H, Han J, Na K (2018) Combination of photodynamic therapy (PDT) and anti-tumor immunity in cancer therapy. J Pharma Invest 48:143–151. https://doi.org/10.1007/s40005-017-0377-x

    Article  CAS  Google Scholar 

  108. Cascão R, Faria CC (2023) Optimizing the role of immunotherapy for the treatment of glioblastoma. In: New insights glioblastoma. Elsevier, New York, pp 553–91. https://doi.org/10.1016/b978-0-323-99873-4.00012-8

  109. Zhang S-C, Hu Z-Q, Long J-H, Zhu G-M, Wang Y, Jia Y et al (2019) Clinical implications of tumor-infiltrating immune cells in breast cancer. J Cancer 10(24):6175. https://doi.org/10.7150/jca.35901

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  110. Kwon Y (2022) Potential pro-tumorigenic effect of bisphenol A in breast cancer via altering the tumor microenvironment. Cancers 14(12):3021. https://doi.org/10.3390/cancers14123021

    Article  CAS  PubMed  PubMed Central  Google Scholar 

Download references

Acknowledgements

Not applicable.

Funding

There is no funding.

Author information

Authors and Affiliations

Authors

Contributions

AMO wrote the manuscript. AMO and OSW performed the experiments, collected, and analyzed data. AKO, RMH, AJ, AA, AMA, and AIA revised the manuscript. AMO conceived and designed the study and revised the manuscript for improved intellectual content. All authors read and approved the final manuscript.

Corresponding author

Correspondence to Ayobami Matthew Olajuyin.

Ethics declarations

Ethics approval and consent to participate

Not applicable.

Consent for publication

Not applicable.

Competing interests

The authors declare that they do not have competing interests.

Additional information

Publisher's Note

Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Rights and permissions

Open Access This article is licensed under a Creative Commons Attribution 4.0 International License, which permits use, sharing, adaptation, distribution and reproduction in any medium or format, as long as you give appropriate credit to the original author(s) and the source, provide a link to the Creative Commons licence, and indicate if changes were made. The images or other third party material in this article are included in the article's Creative Commons licence, unless indicated otherwise in a credit line to the material. If material is not included in the article's Creative Commons licence and your intended use is not permitted by statutory regulation or exceeds the permitted use, you will need to obtain permission directly from the copyright holder. To view a copy of this licence, visit http://creativecommons.org/licenses/by/4.0/.

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Olajuyin, A.M., Nwachukwu, O.S., Olajuyin, A.K. et al. Prognostic values and immune infiltration of KLF15, AQP7, AGPAT9 in glioma and glioblastoma. Futur J Pharm Sci 10, 32 (2024). https://doi.org/10.1186/s43094-024-00608-2

Download citation

  • Received:

  • Accepted:

  • Published:

  • DOI: https://doi.org/10.1186/s43094-024-00608-2

Keywords