Molecules 30 02108
Molecules 30 02108
                                         Abstract: Curcumin, a major phytochemical derived from Curcuma longa, has been shown
                                         to enhance the efficacy of chemotherapeutic agents such as doxorubicin, 5-fluorouracil, and
                                         cisplatin by overcoming drug resistance, making it a promising adjunct in the treatment
                                         of glioblastoma. However, the global gene-expression changes triggered by curcumin in
                                         glioblastoma remain underexplored. In this study, we investigated the effects of curcumin
                                         on human glioblastoma (U87 MG) cells, where it significantly reduced cell viability and
                                         proliferation in a dose- and time-dependent manner and induced apoptosis without af-
                                         fecting senescence. Transcriptomic analysis revealed 5036 differentially expressed genes,
                                         with pathway enrichment identifying 13 dysregulated cancer-associated pathways. No-
                                         tably, curcumin modulated several key regulators involved in MAPK, Ras, TGF-β, Wnt,
                                         Cytokine, and TNF signaling pathways. Several apoptosis and cell cycle-associated genes,
Academic Editors: Marcus                 including PRKCG, GDF7, GDF9, GDF15, GDF5, FZD1, FZD2, FZD8, AIFM3, TP53AIP1,
Tullius Scotti and Luciana Scotti        CRD14, NIBAN3, BOK, BCL2L10, BCL2L14, BNIPL, FASLG, GZMM, TNFSF10, TNFSF11,
Received: 3 April 2025                   and TNFSF4, were significantly altered. Several pro-apoptotic and anti-BCL, cell-cycle-
Revised: 6 May 2025                      regulated genes were modulated following curcumin treatment, emphasizing its potential
Accepted: 8 May 2025                     role in curcumin-mediated anti-tumor effects. This study provides insight into the molecu-
Published: 9 May 2025
                                         lar mechanisms underlying curcumin’s action against glioblastoma.
Citation: Mashozhera, N.T.; Reddy,
C.S.; Ranasinghe, Y.N.; Natarajan, P.;   Keywords: glioblastoma-U87 cells; natural compound; curcumin; apoptosis; RNA-seq
Reddy, U.K.; Hankins, G.
Curcumin-Induced Molecular
Mechanisms in U-87 MG
Glioblastoma Cells: Insights from
Global Gene Expression Profiling.
                                         1. Introduction
Molecules 2025, 30, 2108.                     Glioblastoma multiforme is the most common primary malignancy of the brain, with
https://doi.org/10.3390/                 an incidence of 3.2 in 100,000 [1]. Glioblastoma remains a formidable challenge, with
molecules30102108
                                         a five-year survival rate below 10% despite aggressive treatment [2]. The hallmarks of
Copyright: © 2025 by the authors.        glioblastoma include aggressive and diffuse proliferation, and resistance to apoptosis-
Licensee MDPI, Basel, Switzerland.       inducing drugs [3]. Tumor drug resistance is a major cause of treatment failure, driven by
This article is an open access article
                                         cellular heterogeneity, diverse molecular signatures, and variable drug responsiveness [4].
distributed under the terms and
                                         Conventional chemotherapy fails to achieve complete remission and induces toxicity,
conditions of the Creative Commons
Attribution (CC BY) license
                                         harming normal cells and causing severe side effects [5].
(https://creativecommons.org/                 Plant-derived compounds, known for their pleiotropic anti-cancer effects, have gained
licenses/by/4.0/).                       interest in overcoming multi-drug resistance [6,7]. Natural products like resveratrol,
                           quercetin, EGCG, and curcumin enhance chemotherapy by sensitizing tumor cells through
                           antioxidant, anti-inflammatory, immune-modulatory, and apoptosis-inducing mechanisms,
                           boosting efficacy without added toxicity [8].
                                 Curcumin, a yellow pigment from Curcuma longa, is a well-studied anti-cancer
                           agent with low toxicity and therapeutic potential [9]. Preclinical studies show that cur-
                           cumin enhances the efficacy of chemotherapeutic drugs like sulfinosine, 5-FU, doxorubicin,
                           oxaliplatin, and cisplatin, often sensitizing drug-resistant cells [10]. In prostate cancer,
                           curcumin with docetaxel improved anti-proliferative and apoptotic effects [11]. It also
                           boosted 5-FU efficacy in colon, breast, and gastric cancers [12,13]. In a pancreatic cancer
                           xenograft, curcumin with gemcitabine significantly reduced tumor volume by enhancing
                           anti-proliferative and anti-angiogenic pathways [14]. Clinical trials suggest daily curcumin
                           supplementation exerts anti-inflammatory and immune-modulatory effects in various
                           cancers [15].
                                 Curcumin modulates key pathways in tumor initiation, promotion, and progression.
                           It suppresses carcinogenesis by inducing apoptosis, arresting the cell cycle, and inhibit-
                           ing metastasis, invasion, and angiogenesis [16]. In glioblastoma, curcumin suppresses
                           growth and chemoresistance via AP-1 and NFκB transcription factors, which regulate
                           cell proliferation, apoptosis, and inflammation. In lung cancer, it inhibits metastasis by
                           targeting matrix metalloproteinases (MMP-2 and MMP-9) and vascular endothelial growth
                           factor (VEGF), crucial factors in tumor invasion. For hepatocellular carcinoma, curcumin’s
                           suppression of proliferation and induction of apoptosis occurs through modulation of the
                           Wnt signaling pathway. Similarly, in non-small-cell lung cancer, it impedes migration and
                           invasion through the up-regulation of miR-206 and suppression of the PI3K/AKT/mTOR
                           pathway. Across these studies, curcumin’s multifaceted role in regulating critical molecular
                           signaling pathways and ncRNAs make it a potential multi-targeted therapeutic agent that
                           can be leveraged in cancer prevention and treatment, enhancing efficacy and overcoming
                           resistance mechanisms [17–21].
                                 Although extensive research has revealed curcumin’s anti-carcinogenic properties and
                           its potential to support cancer therapy, studies examining its impact on global gene expres-
                           sion via transcriptome profiling in various cancer cells remain limited. High-throughput
                           RNA sequencing (RNA-seq) of curcumin-treated cells can elucidate the anti-proliferative
                           and cell-death pathways associated with curcumin [22]. Recent studies have employed
                           RNA-seq to profile the most significantly up- and down-regulated genes following cur-
                           cumin treatment, followed by protein–protein interaction analysis and in silico molecular
                           docking to identify novel curcumin targets [23]. Compared with targeted proteomic meth-
                           ods, integrating global pathway enrichment analyses with systemic studies yields a more
                           comprehensive insight into the regulatory and functional dynamics driving curcumin-
                           induced cytotoxicity in cancer, thereby enhancing its therapeutic application [24]. In two
                           breast cancer cell lines, transcriptome profiling showed that curcumin primarily induced
                           cell death via ferroptosis rather than apoptosis, suggesting that ferroptosis may be a more
                           promising therapeutic target [25]. In contrast, in adrenocortical carcinoma cells and their
                           xenograft model, curcumin induced apoptosis mainly through ER stress pathways with
                           accompanying up-regulation of p38 and JNK/MAPK signaling [24].
                                 Given the limited number of transcriptome profiling studies on curcumin, further
                           research could enhance our understanding of its cytotoxic mechanisms in cancer [25].
                           Global gene expression analyses can identify novel genetic targets of curcumin, guiding
                           future research efforts [26]. Accordingly, this study evaluated the impact of curcumin on
                           global gene expression in the U87-MG human glioblastoma cell line using RNA-seq, with
                           its cytotoxic effects confirmed via CCK-8 proliferation, apoptosis, and senescence assays.
                           its cytotoxic effects confirmed via CCK-8 proliferation, apoptosis, and senescence
                                Figure 2. Effect of curcumin on migration of U87 MG cells. (A) The cell monolayer was scratched
                                with a 20 µL pipette tip, photographed at 0 h, and treated for 24 h. (B) Gap closure was quantified
                                by comparing gap width at 0 h and 24 h post-treatment with vehicle or curcumin. Data are mean
                                gap width of six replicates ± SD. One-way ANOVA was used to examine statistical differences fol-
                                lowed by Tukey’s honestly significant difference test for post hoc testing for differences between
                                treatment groups. **** Significant at p < 0.0001.
                                     Senescent cells are identified through elevated lysosomal activity, which can be bio-
                                 chemically assayed through the activity of β-galactosidase enzyme [27]. In this study,
                                 curcumin-induced senescence was assayed using the SA-β-galactosidase assay kit. Cur-
                                 cumin treatment did not significantly induce senescence in U87 MG cells (p > 0.05 when
                                 compared to control) (Figure 3B).
                            Figure 4. Enrichment analysis of up-regulated genes. (A) Bubble plot for KEGG pathway enrichment
                           Figure 4. Enrichment analysis of up-regulated genes. (A) Bubble plot for KEGG pathway enrich-
                            of the top 50 up-regulated genes. Each circle represents an enriched function. The gene enrichment
                           ment
                            ratio of  the top 50
                                  is assigned  to up-regulated
                                                    the x-axis, andgenes.  Each circle
                                                                    the descriptions  of represents
                                                                                         the pathways   anare
                                                                                                           enriched
                                                                                                              assignedfunction.   The gene
                                                                                                                         to the y-axis. The enrich-
                           ment   ratio
                            area of  the is assigned
                                         circles        to the x-axis,
                                                  is proportional       and
                                                                   to the    the descriptions
                                                                          number                 of thetopathways
                                                                                   of genes assigned                   are assigned
                                                                                                            the term, and             tothe
                                                                                                                           the color to  the y-axis.
                           The  area p-value.
                            adjusted             (B) GOisof
                                       of the circles       three ontologies
                                                           proportional       enriched
                                                                          to the number in curcumin-induced
                                                                                            of genes assigned     up-regulated
                                                                                                                    to the term,genes.
                                                                                                                                   and The
                                                                                                                                        the color to
                            width   of each  bar  represents  enrichment   score of genes  ( − log 10 (p-value)). BP  = biological
                           the adjusted p-value. (B) GO of three ontologies enriched in curcumin-induced up-regulated genes.        process,
                            CC = cellular component, MF = molecular function.
                           The width of each bar represents enrichment score of genes (−log10(p-value)). BP = biological process,
                           CC = cellular component,
                                Conversely,         MF = molecular
                                             KEGG pathway          function.
                                                            analysis of the top 50 down-regulated genes revealed
                            involvement in pathways including O-glycan biosynthesis; Notch signaling; glycine, serine,
                            and threonine metabolism; glycolysis and gluconeogenesis; and cytokine–cytokine receptor
                            interaction (Figure 5A). GO analysis further highlighted enrichment in molecular functions,
                            including DNA-binding transcription repressor activity, DNA-binding transcription factor
                            binding, transcription coactivator activity, and catalytic activity acting on RNA (Figure 5B).
                           receptor interaction (Figure 5A). GO analysis further highlighted enrichment in molecular
                           functions, including DNA-binding transcription repressor activity, DNA-binding tran-
Molecules 2025, 30, 2108   scription factor binding, transcription coactivator activity, and catalytic activity
                                                                                                             6 ofacting
                                                                                                                 16     on
                           RNA (Figure 5B).
                            Figure 5. Enrichment analysis of down-regulated genes. (A) Bubble plot for KEGG pathway en-
                           Figure 5. Enrichment analysis of down-regulated genes. (A) Bubble plot for KEGG pathway enrich-
                            richment of the top down-regulated genes. Each circle represents an enriched function. The gene
                           ment   of the ratio
                            enrichment   top down-regulated
                                                is assigned to thegenes.  Eachthecircle
                                                                   x-axis and           represents
                                                                                   description  of theanpathways
                                                                                                          enrichedtofunction.
                                                                                                                     the y-axis.The
                                                                                                                                 Thegene en-
                           richment   ratio
                            area of the      is assigned
                                        circles           to thetox-axis
                                                is proportional          and the
                                                                   the number      description
                                                                                of genes         of to
                                                                                          assigned   the
                                                                                                       thepathways
                                                                                                           term, andto
                                                                                                                     thethe y-axis.
                                                                                                                         color to theThe area
                           ofadjusted p-value.
                              the circles       (B) GO of three
                                           is proportional       ontologies
                                                             to the  number enriched
                                                                              of genesin curcumin-induced
                                                                                         assigned to the term,down-regulated   genes.
                                                                                                                   and the color   to the ad-
                            The width of each bar represents enrichment score of genes (−log10 (p-value)). BP = biological process,
                           justed p-value. (B) GO of three ontologies enriched in curcumin-induced down-regulated genes. The
                            CC = cellular component, MF = molecular function.
                           width of each bar represents enrichment score of genes (−log10(p-value)). BP = biological process, CC
                           = cellular component,
                                  Further   analysisMFof=the
                                                           molecular
                                                               50 mostfunction.
                                                                        significantly altered KEGG pathways identified
                            13 pathways strongly linked to tumorigenesis, cancer progression, and immune evasion.
                            TheseFurther
                                    includedanalysis   of the 50 most
                                              cytokine–cytokine          significantly
                                                                   receptor interaction; altered
                                                                                         pathways KEGG     pathways
                                                                                                     in cancer;         identified 13
                                                                                                                MAPK signal-
                           pathways      strongly
                            ing; regulation         linked
                                              of stem         to tumorigenesis,
                                                        cell pluripotency;  TGF-β cancer
                                                                                   signaling;progression,   and
                                                                                               breast cancer;     immune
                                                                                                                Ras          evasion.
                                                                                                                    signaling;
                            IL-17  signaling; PD-L1   expression   and PD-1  checkpoint   in cancer; proteoglycans
                           These included cytokine–cytokine receptor interaction; pathways in cancer; MAPK sig-      in cancer;
                            Wnt signaling;
                           naling;            gastric
                                     regulation       cancer;
                                                  of stem   celland viral proteinTGF-β
                                                                 pluripotency;    interaction  with cytokines
                                                                                         signaling;             and cytokine
                                                                                                      breast cancer;   Ras signaling;
                            receptors. Among these, several tumor-suppressive pathways stood out. Genes within
                           IL-17 signaling; PD-L1 expression and PD-1 checkpoint in cancer; proteoglycans in cancer;
                            MAPK, Ras, TGF-β, and Wnt pathways, and cytokine–cytokine receptor signaling, such
                           Wnt signaling; gastric cancer; and viral protein interaction with cytokines and cytokine
                            as PRKCG (+8.81), GDF7 (+7.78), GDF5 (−2.68), FZD1 (+2.62), FZD2 (+2.95), and FZD8
                           receptors. Among these, several tumor-suppressive pathways stood out. Genes within
                            (−3.18) are hypothesized to have contributed to curcumin’s anti-tumorigenic effects in
                           MAPK,      Ras,
                            this study.     TGF-β, and
                                         Additionally,   theWnt  pathways,
                                                             well-known       andsuppressor
                                                                          tumor    cytokine–cytokine
                                                                                              gene RUNX3  receptor   signaling, such
                                                                                                            was significantly
                            up-regulated (+8.93) in curcumin-treated cells, further supporting its potential role in
                            curcumin-mediated tumor suppression.
Molecules 2025, 30, 2108                                                                                            7 of 16
                           3. Discussion
                                Curcumin, a polyphenol from Curcuma longa, is a well-studied natural compound
                           that modulates tumor initiation, progression, and promotion in various cancers [28]. In
                           glioblastoma, curcumin suppresses oncogenic signaling, induces multimodal cell death,
                           and modulates immune components within the tumor microenvironment (TME) [29].
                           This study examines the impact of curcumin on global gene expression and its effects on
                           proliferation, migration, apoptosis, and senescence in U87MG glioblastoma cells.
                                  Theeffect
                                 The    effectof of  curcumin
                                                 curcumin         on senescence
                                                             on senescence          inMG
                                                                               in U87   U87cells
                                                                                              MGwascells was examined
                                                                                                      examined               using
                                                                                                                 using β-galac-
                           tosidase activity. While glioma senescence can both suppress and promote malignancyma-
                            β-galactosidase     activity.  While   glioma    senescence  can  both  suppress  and   promote     in
                            lignancy   in vivo   [27], curcumin   (10–20   µmol/L)   did not induce  senescence
                           vivo [27], curcumin (10–20 µmol/L) did not induce senescence (p > 0.05). Prior studies(p >  0.05). Prior
                            studiescurcumin’s
                           suggest   suggest curcumin’s        effectson
                                                    effects depend       depend   on concentration—low
                                                                          concentration—low     doses (≤10doses   (≤10
                                                                                                            µmol/L)      µmol/L)
                                                                                                                      induce   se-
                            induce  senescence     without  cell death,   while higher  doses trigger apoptosis due
                           nescence without cell death, while higher doses trigger apoptosis due to excessive stress to  excessive
                            stress
                           [45].   [45].
                                 The     The
                                      lack oflack   of senescence
                                               senescence    in thisinstudy
                                                                        this study suggests
                                                                              suggests      cytotoxicity
                                                                                       cytotoxicity      at these
                                                                                                    at these      concentrations,
                                                                                                             concentrations,    as
                            as confirmed    by   poly-caspase   and   viability
                           confirmed by poly-caspase and viability assays.      assays.
                           1× antibiotic/antimycotic solution. Once cells reached 80% confluence, they were treated
                           with ethanol (vehicle control) or 10 or 20 µmol/L curcumin. Staurosporine (5 µmol/L)
                           (ImmunoChemistry Technologies, Bloomington, MN, USA) served as a positive control.
                           After 24 h of treatment, cells were collected, stained with FLICA, and incubated for one
                           hour at 37 ◦ C. Following washing, a 100 µL aliquot of the cell suspension, at a density of
                           greater than 2 × 106 cells/mL, was transferred to a 96-well flat-bottom microplate. Fluores-
                           cence was measured at an excitation wavelength of 488 nm and an emission wavelength of
                           520 nm using a microplate reader.
                           5. Conclusions
                                In conclusion, this study elucidated that curcumin acts as a promising therapeutic
                           agent against U87 glioblastoma cells by targeting multiple cancer-related processes, in-
                           cluding regulation of the cell cycle, suppression of cell viability, induction of pro-apoptotic
                           genes, down-regulation of anti-apoptotic genes, and activation of anti-metastatic genes.
                           Curcumin exerts these effects through modulation of several critical signaling pathways,
                           such as the calcium signaling pathway; the PI3K-Akt signaling pathway; TNF signaling;
Molecules 2025, 30, 2108                                                                                                     12 of 16
                                 MAPK, Ras, TGF-β, Wnt, and cytokine signaling, and regulation of the actin cytoskele-
                                 ton. Furthermore, curcumin influences the expression of several pivotal genes involved
                                 in apoptosis, metastasis, and stem cell dynamics, including AIFM3, TP53AIP1, CRD14,
                                 NIBAN3, BOK, BCL2L10, BCL2L14, BNIPL, FASLG, GZMM, TNFSF10, TNFSF11, TNFSF4,
                                 FOS, PRKCG, GDF7, GDF9, GDF15, GDF5, FZD1, FZD2, FZD8, SMAD5, APC, WNT7B,
                                 and HOXA1. By modulating these interconnected molecular networks, curcumin impairs
                                 the survival of cancer stem cells, reduces therapy resistance, inhibits metastatic potential,
                                 and sensitizes tumor cells to conventional treatments. Collectively, these findings highlight
                                 curcumin’s multifaceted role in cancer prevention and therapy, emphasizing its potential
                                 as a powerful adjunct in glioblastoma management.
                                 Supplementary Materials: The following supporting information can be downloaded at: https://
                                 www.mdpi.com/article/10.3390/molecules30102108/s1, Figure S1: Cell viability was assessed at
                                 24 h following treatment with various concentrations of curcumin, and the corresponding IC50 value
                                 was determined based on the dose–response relationship. Figure S2: Principal Component Analysis
                                 (PCA) of RNA-seq data. PCA shows the variance in normalized gene expression profiles across
                                 samples. The first two principal components, PC1 and PC2, explain 85% and 5% of the total variance,
                                 respectively. Samples are color- and shape-coded by group: U87C (control) in red circles and U87T
                                 (treated) in cyan triangles.
                                 Author Contributions: Conceptualization, G.H. and U.K.R.; experimentation, N.T.M., Y.N.R. and
                                 C.S.R.; RNA-seq analysis, P.N.; writing—original draft preparation, N.T.M., C.S.R., G.H. and U.K.R.;
                                 writing—review and editing, C.S.R. and U.K.R. All authors have read and agreed to the published
                                 version of the manuscript.
                                 Funding: This material is based on work supported by the National Science Foundation (NSF) under
                                 award number 2242771.
                                 Data Availability Statement: The raw paired-end Illumina RNA-sequencing reads generated in
                                 the current study are available in the Sequence Read Archive (SRA) at NCBI under the Bio project
                                 accession numbers PRJNA1243709.
References
1.    Agosti, E.; Zeppieri, M.; De Maria, L.; Tedeschi, C.; Fontanella, M.M.; Panciani, P.P.; Ius, T. Glioblastoma immunotherapy: A
      systematic review of the present strategies and prospects for advancements. Int. J. Mol. Sci. 2023, 24, 15037. [CrossRef]
2.    Ren, X.; Qin, L. Glioblastoma Management in the Post-COVID-19 Era: Challenges, Strategies, and Adaptations; Intechopen: London,
      UK, 2025.
3.    Jackson, G.A.; Adamson, D.C. Similarities in Mechanisms of Ovarian Cancer Metastasis and Brain Glioblastoma Multiforme
      Invasion Suggest Common Therapeutic Targets. Cells 2025, 14, 171. [CrossRef]
4.    Aleksakhina, S.N.; Kashyap, A.; Imyanitov, E.N. Mechanisms of acquired tumor drug resistance. Biochim. Et Biophys. Acta
      (BBA)-Rev. Cancer 2019, 1872, 188310. [CrossRef] [PubMed]
5.    Srirangan, P.; Sabina, E.P. Protective effects of herbal compounds against cyclophosphamide-induced organ toxicity: A pathway-
      centered approach. Drug Chem. Toxicol. 2025, 1–43. [CrossRef] [PubMed]
6.    Reddy, C.S.; Natarajan, P.; Nimmakayala, P.; Hankins, G.R.; Reddy, U.K. From Fruit Waste to Medical Insight: The Comprehensive
      Role of Watermelon Rind Extract on Renal Adenocarcinoma Cellular and Transcriptomic Dynamics. Int. J. Mol. Sci. 2023,
      24, 15615. [CrossRef]
7.    Chinreddy, S.R.; Mashozhera, N.T.; Rashrash, B.; Flores-Iga, G.; Nimmakayala, P.; Hankins, G.R.; Harris, R.T.; Reddy, U.K.
      Unraveling TRPV1’s Role in Cancer: Expression, Modulation, and Therapeutic Opportunities with Capsaicin. Molecules 2024,
      29, 4729. [CrossRef] [PubMed]
Molecules 2025, 30, 2108                                                                                                             13 of 16
8.    Datta, S.; Saha, P.; Dey, S.; Sinha, D. Natural Products as Chemosensitizers for Adjunct Therapy in Cancer Management.
      Pharmacother. Bot. Cancer Chemoprev. 2020, 154, 67–119.
9.    Islam, M.R.; Rauf, A.; Akash, S.; Trisha, S.I.; Nasim, A.H.; Akter, M.; Dhar, P.S.; Ogaly, H.A.; Hemeg, H.A.; Wilairatana, P.
      Targeted therapies of curcumin focus on its therapeutic benefits in cancers and human health: Molecular signaling pathway-based
      approaches and future perspectives. Biomed. Pharmacother. 2024, 170, 116034. [CrossRef]
10.   Banerjee, S.; Singh, S.K.; Chowdhury, I.; Lillard, J.W., Jr.; Singh, R. Combinatorial effect of curcumin with docetaxel modulates
      apoptotic and cell survival molecules in prostate cancer. Front. Biosci. 2017, 9, 235.
11.   Tiryaki, S.; Macit, M.; Zemheri, I.E.; Süt, P.A.; Duman, G.; Telci, D. Anticancer Activity of Soy Lecithin-Based Curcumin in Prostate
      Cancer. J. Appl. Polym. Sci. 2025, 142, e56816. [CrossRef]
12.   Du, B.; Jiang, L.; Xia, Q.; Zhong, L. Synergistic inhibitory effects of curcumin and 5-fluorouracil on the growth of the human colon
      cancer cell line HT-29. Chemotherapy 2005, 52, 23–28. [CrossRef] [PubMed]
13.   Yang, Z.; Cespedes-Acuña, C.L.; Yang, Z.; Abu Bakar, M.Z.; Chan, K.W.; Deng, X. Plant foods and their bioactives as dietary
      enhancers for colon cancer treatment with 5-fluorouracil. Food Rev. Int. 2025, 1–50. [CrossRef]
14.   Casarcia, N.; Rogers, P.; Guld, E.; Iyer, S.; Li, Y.; Burcher, J.T.; Deliberto, L.K.; Banerjee, S.; Bishayee, A. Phytochemicals for the
      prevention and treatment of pancreatic cancer: Current progress and future prospects. Br. J. Pharmacol. 2023, 182, 2181–2234.
      [CrossRef] [PubMed]
15.   Allegra, A.; Mirabile, G.; Ettari, R.; Pioggia, G.; Gangemi, S. The impact of curcumin on immune response: An immunomodulatory
      strategy to treat sepsis. Int. J. Mol. Sci. 2022, 23, 14710. [CrossRef]
16.   Shanmugam, M.K.; Rane, G.; Kanchi, M.M.; Arfuso, F.; Chinnathambi, A.; Zayed, M.; Alharbi, S.A.; Tan, B.K.; Kumar, A.P.; Sethi,
      G. The multifaceted role of curcumin in cancer prevention and treatment. Molecules 2015, 20, 2728–2769. [CrossRef]
17.   Dhandapani, K.M.; Mahesh, V.B.; Brann, D.W. Curcumin suppresses growth and chemoresistance of human glioblastoma cells
      via AP-1 and NFκB transcription factors. J. Neurochem. 2007, 102, 522–538. [CrossRef]
18.   Lin, S.-S.; Lai, K.-C.; Hsu, S.-C.; Yang, J.-S.; Kuo, C.-L.; Lin, J.-P.; Ma, Y.-S.; Wu, C.-C.; Chung, J.-G. Curcumin inhibits the
      migration and invasion of human A549 lung cancer cells through the inhibition of matrix metalloproteinase-2 and-9 and Vascular
      Endothelial Growth Factor (VEGF). Cancer Lett. 2009, 285, 127–133. [CrossRef] [PubMed]
19.   Xu, M.X.; Zhao, L.; Deng, C.; Yang, L.; Wang, Y.; Guo, T.; Li, L.; Lin, J.; Zhang, L. Curcumin suppresses proliferation and induces
      apoptosis of human hepatocellular carcinoma cells via the wnt signaling pathway. Int. J. Oncol. 2013, 43, 1951–1959. [CrossRef]
      [PubMed]
20.   Wang, N.; Feng, T.; Liu, X.; Liu, Q. Curcumin inhibits migration and invasion of non-small cell lung cancer cells through
      up-regulation of miR-206 and suppression of PI3K/AKT/mTOR signaling pathway. Acta Pharm. 2020, 70, 399–409. [CrossRef]
21.   Wang, H.; Zhang, K.; Liu, J.; Yang, J.; Tian, Y.; Yang, C.; Li, Y.; Shao, M.; Su, W.; Song, N. Curcumin regulates cancer progression:
      Focus on ncRNAs and molecular signaling pathways. Front. Oncol. 2021, 11, 660712. [CrossRef]
22.   Khazei, K.; Jamali, M.; Sarhadi, S.; Dadashpour, M.; Shokrollahzade, S.; Zarghami, N. Transcriptome profiling of curcumin-treated
      T47D human breast cancer cells by a system-based approach. Gene Rep. 2022, 27, 101556. [CrossRef]
23.   Wang, R.; Yu, H.; Chen, P.; Yuan, T.; Zhang, J. Integrated transcriptome and molecular docking to identify the hub superimposed
      attenuation targets of curcumin in breast cancer cells. Int. J. Mol. Sci. 2023, 24, 12479. [CrossRef] [PubMed]
24.   Huang, X.; Liang, C.; Yang, H.; Li, X.; Deng, X.; Liang, X.; Li, L.; Huang, Z.; Lu, D.; Ma, Y. Curcumin induces apoptosis and
      inhibits the growth of adrenocortical carcinoma: Identification of potential candidate genes and pathways by transcriptome
      analysis. Oncol. Lett. 2021, 21, 476. [CrossRef]
25.   Li, R.; Zhang, J.; Zhou, Y.; Gao, Q.; Wang, R.; Fu, Y.; Zheng, L.; Yu, H. Transcriptome investigation and in vitro verification
      of curcumin-induced HO-1 as a feature of ferroptosis in breast cancer cells. Oxidative Med. Cell. Longev. 2020, 2020, 3469840.
      [CrossRef] [PubMed]
26.   Mo, F.; Xiao, Y.; Zeng, H.; Fan, D.; Song, J.; Liu, X.; Luo, M.; Ma, X. Curcumin-induced global profiling of transcriptomes in small
      cell lung cancer cells. Front. Cell Dev. Biol. 2021, 8, 588299. [CrossRef]
27.   Salam, R.; Saliou, A.; Bielle, F.; Bertrand, M.; Antoniewski, C.; Carpentier, C.; Alentorn, A.; Capelle, L.; Sanson, M.; Huillard, E.
      Cellular senescence in malignant cells promotes tumor progression in mouse and patient Glioblastoma. Nat. Commun. 2023,
      14, 441. [CrossRef]
28.   Giordano, A.; Tommonaro, G. Curcumin and cancer. Nutrients 2019, 11, 2376. [CrossRef]
29.   Wong, S.C.; Kamarudin, M.N.A.; Naidu, R. Anticancer mechanism of curcumin on human glioblastoma. Nutrients 2021, 13, 950.
      [CrossRef]
30.   Zoi, V.; Kyritsis, A.P.; Galani, V.; Lazari, D.; Sioka, C.; Voulgaris, S.; Alexiou, G.A. The role of curcumin in cancer: A focus on the
      PI3K/Akt pathway. Cancers 2024, 16, 1554. [CrossRef]
31.   Hesari, A.; Rezaei, M.; Rezaei, M.; Dashtiahangar, M.; Fathi, M.; Rad, J.G.; Momeni, F.; Avan, A.; Ghasemi, F. Effect of curcumin
      on glioblastoma cells. J. Cell. Physiol. 2019, 234, 10281–10288. [CrossRef]
Molecules 2025, 30, 2108                                                                                                                  14 of 16
32.   Yang, C.-L.; Liu, Y.-Y.; Ma, Y.-G.; Xue, Y.-X.; Liu, D.-G.; Ren, Y.; Liu, X.-B.; Li, Y.; Li, Z. Curcumin blocks small cell lung cancer cells
      migration, invasion, angiogenesis, cell cycle and neoplasia through Janus kinase-STAT3 signalling pathway. PLoS ONE 2012,
      7, e37960. [CrossRef]
33.   Chen, Q.-Y.; Zheng, Y.; Jiao, D.-M.; Chen, F.-Y.; Hu, H.-Z.; Wu, Y.-Q.; Song, J.; Yan, J.; Wu, L.-J.; Lv, G.-Y. Curcumin inhibits lung
      cancer cell migration and invasion through Rac1-dependent signaling pathway. J. Nutr. Biochem. 2014, 25, 177–185. [CrossRef]
      [PubMed]
34.   Li, W.; Sun, L.; Lei, J.; Wu, Z.; Ma, Q.; Wang, Z. Curcumin inhibits pancreatic cancer cell invasion and EMT by interfering with
      tumor-stromal crosstalk under hypoxic conditions via the IL-6/ERK/NF-κB axis. Oncol. Rep. 2020, 44, 382–392. [CrossRef]
      [PubMed]
35.   Weissenberger, J.; Priester, M.; Bernreuther, C.; Rakel, S.; Glatzel, M.; Seifert, V.; Kögel, D. Dietary curcumin attenuates glioma
      growth in a syngeneic mouse model by inhibition of the JAK1, 2/STAT3 signaling pathway. Clin. Cancer Res. 2010, 16, 5781–5795.
      [CrossRef]
36.   Li, J.; Ye, L.; Parr, C.; Douglas-Jones, A.; Kynaston, H.G.; Mansel, R.E.; Jiang, W.G. The aberrant expression of bone morphogenetic
      protein 12 (BMP-12) in human breast cancer and its potential prognostic value. Gene Ther. Mol. Biol. 2009, 13, 186–193. [CrossRef]
      [PubMed]
37.   Abdullah Thani, N.A.; Sallis, B.; Nuttall, R.; Schubert, F.R.; Ahsan, M.; Davies, D.; Purewal, S.; Cooper, A.; Rooprai, H.K. Induction
      of apoptosis and reduction of MMP gene expression in the U373 cell line by polyphenolics in Aronia melanocarpa and by
      curcumin. Oncol. Rep. 2012, 28, 1435–1442. [CrossRef]
38.   Karmakar, S.; Banik, N.L.; Ray, S.K. Curcumin suppressed anti-apoptotic signals and activated cysteine proteases for apoptosis in
      human malignant glioblastoma U87MG cells. Neurochem. Res. 2007, 32, 2103–2113. [CrossRef]
39.   Cheng, C.; Jiao, J.T.; Qian, Y.; Guo, X.Y.; Huang, J.; Dai, M.C.; Zhang, L.; Ding, X.P.; Zong, D.; Shao, J.F. Curcumin induces G2/M
      arrest and triggers apoptosis via FoxO1 signaling in U87 human glioma cells. Mol. Med. Rep. 2016, 13, 3763–3770. [CrossRef]
40.   Wang, P.; Hao, X.; Li, X.; Yan, Y.; Tian, W.; Xiao, L.; Wang, Z.; Dong, J. Curcumin inhibits adverse psychological stress-induced
      proliferation and invasion of glioma cells via down-regulating the ERK/MAPK pathway. J. Cell. Mol. Med. 2021, 25, 7190–7203.
      [CrossRef]
41.   Wang, L.; Hu, R.; Dai, A. Curcumin Increased the Sensitivity of Non-Small-Cell Lung Cancer to Cisplatin through the Endoplasmic
      Reticulum Stress Pathway. Evid.-Based Complement. Altern. Med. 2022, 2022, 6886366. [CrossRef]
42.   Huang, Y.-F.; Zhu, D.-J.; Chen, X.-W.; Chen, Q.-K.; Luo, Z.-T.; Liu, C.-C.; Wang, G.-X.; Zhang, W.-J.; Liao, N.-Z. Curcumin
      enhances the effects of irinotecan on colorectal cancer cells through the generation of reactive oxygen species and activation of the
      endoplasmic reticulum stress pathway. Oncotarget 2017, 8, 40264. [CrossRef] [PubMed]
43.   Su, X.; Chen, S.; Lu, H.; Li, H.; Qin, C. Study on the inhibitory effect of curcumin on GBM and its potential mechanism. Drug Des.
      Dev. Ther. 2021, 15, 2769–2781. [CrossRef]
44.   Huang, T.-Y.; Tsai, T.-H.; Hsu, C.-W.; Hsu, Y.-C. Curcuminoids suppress the growth and induce apoptosis through caspase-3-
      dependent pathways in glioblastoma multiforme (GBM) 8401 cells. J. Agric. Food Chem. 2010, 58, 10639–10645. [CrossRef]
45.   Mosieniak, G.; Adamowicz, M.; Alster, O.; Jaskowiak, H.; Szczepankiewicz, A.A.; Wilczynski, G.M.; Ciechomska, I.A.; Sikora, E.
      Curcumin induces permanent growth arrest of human colon cancer cells: Link between senescence and autophagy. Mech. Ageing
      Dev. 2012, 133, 444–455. [CrossRef]
46.   Zhang, H.; Wang, R.; Yu, Y.; Liu, J.; Luo, T.; Fan, F. Glioblastoma treatment modalities besides surgery. J. Cancer 2019, 10, 4793.
      [CrossRef]
47.   Cheng, J.; Zeng, M.; Peng, B.; Li, P.; Zhao, S. Transient receptor potential vanilloid-1 (TRPV1) channels act as suppressors of the
      growth of glioma. Brain Res. Bull. 2024, 211, 110950. [CrossRef]
48.   Pucci, C.; Martinelli, C.; Ciofani, G. Innovative approaches for cancer treatment: Current perspectives and new challenges.
      ecancermedicalscience 2019, 13, 961. [CrossRef] [PubMed]
49.   Aggarwal, B.B.; Sundaram, C.; Malani, N.; Ichikawa, H. Curcumin: The Indian solid gold. Mol. Targets Ther. Uses Curcumin Health
      Dis. 2007, 595, 1–75.
50.   Avci, C.; Dodurga, Y.; Susluer, S.; Sıgva, Z.; Yucebas, M.; Caglar, H.; Akalin, T.; Dalbasti, T.; Oktar, N.; Gunduz, C. Promoter
      hypermethylation-mediated down-regulation of RUNX3 gene in human brain tumors. Ir. J. Med. Sci. 2014, 183, 259–264.
      [CrossRef] [PubMed]
51.   Chuang, L.; Ito, Y. RUNX3 is multifunctional in carcinogenesis of multiple solid tumors. Oncogene 2010, 29, 2605–2615. [CrossRef]
52.   Li, X.; Zheng, J.; Diao, H.; Liu, Y. RUNX 3 is down-regulated in glioma by Myc-regulated miR-4295. J. Cell. Mol. Med. 2016, 20,
      518–525. [CrossRef]
53.   Steponaitis, G.; Kazlauskas, A.; Vaitkienė, P.; Deltuva, V.P.; Mikuciunas, M.; Skiriutė, D. Oncosuppressive role of RUNX3 in
      human astrocytomas. J. Oncol. 2019, 2019, 1232434. [CrossRef]
54.   Mei, P.-J.; Bai, J.; Liu, H.; Li, C.; Wu, Y.-P.; Yu, Z.-Q.; Zheng, J.-N. RUNX3 expression is lost in glioma and its restoration causes
      drastic suppression of tumor invasion and migration. J. Cancer Res. Clin. Oncol. 2011, 137, 1823–1830. [CrossRef]
Molecules 2025, 30, 2108                                                                                                            15 of 16
55.   Sun, J.; Jia, Z.; Li, B.; Zhang, A.; Wang, G.; Pu, P.; Chen, Z.; Wang, Z.; Yang, W. MiR-19 regulates the proliferation and invasion of
      glioma by RUNX3 via β-catenin/Tcf-4 signaling. Oncotarget 2017, 8, 110785. [CrossRef]
56.   Sun, J.; Li, B.; Jia, Z.; Zhang, A.; Wang, G.; Chen, Z.; Shang, Z.; Zhang, C.; Cui, J.; Yang, W. RUNX3 inhibits glioma survival and
      invasion via suppression of the β-catenin/TCF-4 signaling pathway. J. Neuro-Oncol. 2018, 140, 15–26. [CrossRef]
57.   Newton, A.C.; Brognard, J. Reversing the paradigm: Protein kinase C as a tumor suppressor. Trends Pharmacol. Sci. 2017, 38,
      438–447. [CrossRef]
58.   Pilo, C.A.; Newton, A.C. Two sides of the same coin: Protein kinase C γ in cancer and neurodegeneration. Front. Cell Dev. Biol.
      2022, 10, 929510. [CrossRef]
59.   Satow, R.; Suzuki, Y.; Asada, S.; Ota, S.; Idogawa, M.; Kubota, S.; Ikeo, N.; Yoneda, A.; Fukami, K. Downregulation of protein
      kinase C gamma reduces epithelial property and enhances malignant phenotypes in colorectal cancer cells. Iscience 2022,
      25, 105501. [CrossRef]
60.   Liu, L.; Wang, G.; Wang, L.; Yu, C.; Li, M.; Song, S.; Hao, L.; Ma, L.; Zhang, Z. Computational identification and characterization
      of glioma candidate biomarkers through multi-omics integrative profiling. Biol. Direct 2020, 15, 10. [CrossRef]
61.   Wang, J.-Y.; Wang, X.; Wang, X.-J.; Zheng, B.-Z.; Wang, Y.; Liang, B. Curcumin inhibits the growth via Wnt/β-catenin pathway in
      non-small-cell lung cancer cells. Eur. Rev. Med. Pharmacol. Sci. 2018, 22, 7492–7499.
62.   Zhang, X.; Dong, S.; Xu, F. Structural and druggability landscape of frizzled G protein-coupled receptors. Trends Biochem. Sci.
      2018, 43, 1033–1046. [CrossRef]
63.   Huang, K.; Xu, H.; Han, L.; Xu, R.; Xu, Z.; Xie, Y. Identification of therapeutic targets and prognostic biomarkers among frizzled
      family genes in glioma. Front. Mol. Biosci. 2023, 9, 1054614. [CrossRef] [PubMed]
64.   Hirano, H.; Yonezawa, H.; Yunoue, S.; Habu, M.; Uchida, H.; Yoshioka, T.; Kishida, S.; Kishida, M.; Oyoshi, T.; Fujio, S. Immunore-
      activity of Wnt5a, Fzd2, Fzd6, and Ryk in glioblastoma: Evaluative methodology for DAB chromogenic immunostaining. Brain
      Tumor Pathol. 2014, 31, 85–93. [CrossRef] [PubMed]
65.   Ran, Y.; Han, S.; Gao, D.; Chen, X.; Liu, C. Interference of FZD2 suppresses proliferation, vasculogenic mimicry and stemness in
      glioma cells via blocking the Notch/NF-κB signaling pathway. Exp. Ther. Med. 2024, 28, 373. [CrossRef] [PubMed]
66.   Jia, F.; Zhang, L.; Jiang, Z.; Tan, G.; Wang, Z. FZD1/KLF10-hsa-miR-4762–5p/miR-224–3p-circular RNAs axis as prognostic
      biomarkers and therapeutic targets for glioblastoma: A comprehensive report. BMC Med. Genom. 2023, 16, 21. [CrossRef]
67.   Ma, S.; Guo, Z.; Wang, B.; Yang, M.; Yuan, X.; Ji, B.; Wu, Y.; Chen, S. A Computational Framework to Identify Biomarkers for
      Glioma Recurrence and Potential Drugs Targeting Them. Front. Genet. 2022, 12, 832627. [CrossRef]
68.   Ohka, F.; Shinjo, K.; Deguchi, S.; Matsui, Y.; Okuno, Y.; Katsushima, K.; Suzuki, M.; Kato, A.; Ogiso, N.; Yamamichi, A. Pathogenic
      epigenetic consequences of genetic alterations in IDH-wild-type diffuse astrocytic gliomas. Cancer Res. 2019, 79, 4814–4827.
      [CrossRef]
69.   Li, Y.; Liu, Z.; Zhang, Y. Expression and prognostic impact of FZDs in pancreatic adenocarcinoma. BMC Gastroenterol. 2021, 21,
      1–14. [CrossRef]
70.   Sun, Y.; Wang, W.; Zhao, C. Frizzled receptors in tumors, focusing on signaling, roles, modulation mechanisms, and targeted
      therapies. Oncol. Res. 2021, 28, 661. [CrossRef]
71.   Yin, S.; Xu, L.; Bonfil, R.D.; Banerjee, S.; Sarkar, F.H.; Sethi, S.; Reddy, K.B. Tumor-initiating cells and FZD8 play a major role in
      drug resistance in triple-negative breast cancer. Mol. Cancer Ther. 2013, 12, 491–498. [CrossRef]
72.   Caja, L.; Bellomo, C.; Moustakas, A. Transforming growth factor β and bone morphogenetic protein actions in brain tumors.
      FEBS Lett. 2015, 589, 1588–1597. [CrossRef] [PubMed]
73.   Tenen, D.G. Disruption of differentiation in human cancer: AML shows the way. Nat. Rev. Cancer 2003, 3, 89–101. [CrossRef]
74.   Hanavadi, S.; Martin, T.; Watkins, G.; Mansel, R.; Jiang, W. The role of growth differentiation factor-9 (GDF-9) and its analog,
      GDF-9b/BMP-15, in human breast cancer. Ann. Surg. Oncol. 2007, 14, 2159–2166. [CrossRef]
75.   Varadaraj, A.; Patel, P.; Serrao, A.; Bandyopadhay, T.; Lee, N.Y.; Jazaeri, A.A.; Huang, Z.; Murphy, S.K.; Mythreye, K. Epigenetic
      regulation of GDF2 suppresses anoikis in ovarian and breast epithelia. Neoplasia 2015, 17, 826–838. [CrossRef]
76.   Du, P.; Ye, L.; Li, H.; Ruge, F.; Yang, Y.; Jiang, W.G. Growth differentiation factor-9 expression is inversely correlated with an
      aggressive behaviour in human bladder cancer cells. Int. J. Mol. Med. 2012, 29, 428–434. [PubMed]
77.   Codó, P.; Weller, M.; Kaulich, K.; Schraivogel, D.; Silginer, M.; Reifenberger, G.; Meister, G.; Roth, P. Control of glioma cell
      migration and invasiveness by GDF-15. Oncotarget 2016, 7, 7732. [CrossRef] [PubMed]
78.   Peng, H.; Li, Z.; Fu, J.; Zhou, R. Growth and differentiation factor 15 regulates PD-L1 expression in glioblastoma. Cancer Manag.
      Res. 2019, 11, 2653–2661. [CrossRef]
79.   Kadowaki, M.; Yoshioka, H.; Kamitani, H.; Watanabe, T.; Wade, P.A.; Eling, T.E. DNA methylation-mediated silencing of
      nonsteroidal anti-inflammatory drug-activated gene (NAG-1/GDF15) in glioma cell lines. Int. J. Cancer 2012, 130, 267–277.
      [CrossRef]
80.   Bolger, A.; Giorgi, F. Trimmomatic: A flexible read trimming tool for Illumina NGS data. Bioinformatics 2014, 30, 2114–2120.
      [CrossRef]
Molecules 2025, 30, 2108                                                                                                           16 of 16
81.   Dobin, A.; Davis, C.A.; Schlesinger, F.; Drenkow, J.; Zaleski, C.; Jha, S.; Batut, P.; Chaisson, M.; Gingeras, T.R. STAR: Ultrafast
      universal RNA-seq aligner. Bioinformatics 2013, 29, 15–21. [CrossRef]
82.   Anders, S.; Pyl, P.T.; Huber, W. HTSeq—A Python framework to work with high-throughput sequencing data. Bioinformatics 2015,
      31, 166–169. [CrossRef] [PubMed]
83.   Love, M.; Anders, S.; Huber, W. Differential analysis of count data–the DESeq2 package. Genome Biol 2014, 15, 10–1186.
84.   Xie, C.; Mao, X.; Huang, J.; Ding, Y.; Wu, J.; Dong, S.; Kong, L.; Gao, G.; Li, C.-Y.; Wei, L. KOBAS 2.0: A web server for annotation
      and identification of enriched pathways and diseases. Nucleic Acids Res. 2011, 39 (Suppl. S2), W316–W322. [CrossRef] [PubMed]
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.