Highlights of the Manuscript
Glioma Stem-like Cells (GSCs), derived from progenitor cells, are crucial in initiating and sustaining glioblastoma (GB), contributing to tumor recurrence and resistance due to their plasticity and self-renewal.
Genetic mutations and epigenetic modifications convert progenitor cells into tumor-initiating GSCs, forming a therapy-resistant cell population.
GB cells recruit microglia to create an immunosuppressive microenvironment, which promotes tumor growth and complicates therapeutic response.
Cytokines and growth factors facilitate communication between microglia and GSCs, supporting GSC survival and enhancing resistance to treatment.
miRNAs and lncRNAs regulate genes that impact tumor progression, cellular interactions, and immune responses, supporting the maintenance of the stem-like phenotype in GSCs.
Due to the diversity of cellular and molecular interactions, effective GB treatment requires strategies that target multiple pathways and cell types.
Tailoring therapies to individual tumor profiles, specifically ncRNA expression patterns, could improve treatment outcomes.
Combining therapies to address GSCs, microglia, and ncRNAs may help overcome resistance mechanisms and improve therapeutic efficacy.
Novel drug delivery systems, such as nanoparticles and exosomes, are needed to cross the blood-brain barrier and deliver targeted treatments effectively.
Targeting this axis with pathway inhibitors, immunomodulatory agents, and ncRNA-based therapies is a promising approach to disrupt tumor growth, reduce resistance, and extend patient survival.
1. Introduction
Glioblastoma (GB) is the most aggressive and deadly form of primary brain tumor in adults, accounting for over 50% of all gliomas [
1]. It is characterized by rapid growth, strong invasion of the surrounding brain tissue, and marked cellular heterogeneity, making treatment difficult [
2]. Current therapeutic approaches include maximal surgical resection followed by concurrent chemoradiotherapy and chemotherapy, usually with temozolomide [
3,
4]. However, even with aggressive treatment, the prognosis remains dismal, with a median survival of 12-15 months and a 5-year survival rate of less than 10% [
1]. A major challenge is incomplete surgical removal of the tumor due to its diffuse infiltration into healthy brain tissue, as well as the development of resistance to radiation and chemotherapy [
5]. Furthermore, the tumor microenvironment (TME) and molecular complexity, including genetic and epigenetic alterations, contribute to poor treatment outcomes. Understanding the intricate cellular and molecular mechanisms that drive GB pathogenesis and resistance to therapy is crucial for developing new and effective treatment strategies.
This review provides a comprehensive overview of three key components of GB progression: progenitor cells, microglia, and non-coding RNAs (ncRNAs). Progenitor cells, particularly glioma stem-like cells (GSCs), are a source of tumor recurrence and resistance because of their ability to self-renew and differentiate into various cell types within the tumor. Microglia, the resident immune cells of the brain, are often reprogrammed by the tumor to create an immunosuppressive environment, aiding in invasiveness and resistance to therapies. In addition, ncRNAs, such as microRNAs and lncRNAs, have emerged as crucial regulators of gene expression, modulating pathways that drive GB malignancy and resistance to conventional treatments.
By focusing on these components, we discuss how their interactions contribute to the development and persistence of GB and how they can be targeted for novel therapeutic interventions. The overarching goal is to highlight promising research avenues that could lead to more effective targeted therapies, improve patient outcomes, and overcome current therapeutic challenges.
2. Progenitor Cells in GB: Drivers of Tumorigenesis and Resistance
2.1. Progenitor Cells in the Central Nervous System
Neural progenitor cells (NPCs) are essential components of the central nervous system (CNS) and are responsible for generating various types of neural cells during brain development. They exhibit the capacity for self-renewal and differentiation, and play a critical role in maintaining brain homeostasis and responding to injury [
6,
7]. However, in the GB, certain progenitor cells, particularly GSCs, undergo an aberrant transformation, becoming key drivers of tumorigenesis (
Figure 1). GSCs share many characteristics with normal NPCs, including self-renewal and multipotency, but they also exhibit enhanced survival, proliferation, and resistance to standard therapies [
7]. This malignant stem-like population is thought to be responsible for the initiation, growth, and recurrence of GB, making them pivotal players in the aggressive nature [
7].
2.2. Transformation of Progenitor Cells into GSCs
The transformation of progenitor cells into GSCs is a complex process driven by both genetic and epigenetic alterations [
8]. Mutations in key oncogenes and tumor suppressor genes such as
TP53 and
IDH1 are common in gliomas and contribute to the malignant transformation of these cells [
9].
TP53 mutations, a hallmark of many cancers, disrupt normal cell cycle regulation and promote genomic instability. Meanwhile, mutations in
IDH1, particularly in lower-grade astrocytomas and oligodendrogliomas, are linked to the production of the oncometabolite 2-hydroxyglutarate, which interferes with cellular differentiation and promotes a stem-like state [
10,
11,
12].
Epigenetic modifications such as DNA methylation and histone modification further contribute to the stem-like phenotype of GB cells. These changes alter gene expression patterns and maintain progenitor cells in an undifferentiated and proliferative state [
13]. In addition, the TME plays a crucial role in the transformation and maintenance of GSCs. Hypoxia is a hallmark of most malignancies, including GB-TME, and has been connected to worse patient outcomes and aggressive metastatic features. GSC survival and stemness are supported by the hypoxic condition they occasionally encounter, which is thought to be regulated by hypoxia-inducible factor (HIF) signaling [
14]. Cytokines and growth factors within the TME, including interleukin-6 (IL-6) and transforming growth factor-beta (TGF-β), further promote the plasticity and self-renewal capabilities of progenitor cells, enabling their adaptation to the tumor niche [
15,
16,
17].
2.3. Progenitor Cells and Tumor Growth
GSCs are pivotal not only in tumor initiation but also in driving the continuous growth and invasion of GB [
18]. These cells display enhanced proliferative potential and are highly invasive, contributing to the diffuse infiltration of the GB throughout the brain [
19,
20]. Unlike more differentiated tumor cells, GSCs possess the ability to migrate along white matter tracts and blood vessels, enabling them to evade surgical resection and seed new tumor foci [
21].
The presence of GSCs within the tumor mass also contributes to its heterogeneity, which is a defining feature of GB. GSCs can differentiate into various cell types within the tumor, leading to a heterogeneous population of cells with varying levels of susceptibility to treatment [
2,
22]. This heterogeneity is a significant factor in the development of therapy resistance, as GSCs can survive treatments that effectively eliminate differentiated tumor cells. As a result, GSCs are often implicated in tumor recurrence as they can repopulate the tumor following therapy [
5,
23].
2.4. Progenitor Cells and Therapy Resistance
Progenitor cells, particularly GSCs, are highly resistant to conventional therapies including radiation and chemotherapy. This resistance is attributed to several factors, including enhanced DNA repair mechanisms, slow cell cycle progression, and activation of survival pathways that protect cells from apoptosis [
24,
25,
26]. For instance, GSCs exhibit increased expression of DNA repair proteins such as MGMT (O6-methylguanine-DNA methyltransferase), which confers resistance to temozolomide, the standard chemotherapeutic agent used in GB treatment. Furthermore, GSCs often reside in protective niches within tumors, such as perivascular or hypoxic regions, where they are shielded from therapeutic agents and radiation [
5,
24,
25,
26].
The cellular plasticity of GSCs also plays a significant role in resistance to therapy. These cells can dynamically switch between stem-like and differentiated states in response to therapeutic pressure, allowing them to survive treatment and re-establish the tumor. This adaptability makes targeting GSCs a crucial focus for the development of new therapeutic strategies [
22,
27,
28].
Emerging therapies targeting key signaling pathways involved in GSC maintenance and self-renewal, such as the Notch, Wnt, and Sonic Hedgehog (SHH) pathways, have shown promise in preclinical studies [
26,
29]. These pathways are critical for the regulation of stemness and differentiation of both normal progenitor cells and GSCs. Inhibitors of these pathways, such as gamma-secretase inhibitors (targeting Notch signaling), are being investigated as potential therapies to specifically target the GSC population, thereby overcoming resistance and reducing the likelihood of tumor recurrence [
26,
29,
30,
31].
Figure 1.
Role of Progenitors Cells in Glioblastoma (GB). Cellular plasticity, genetic mutations, self-renewal capacity, and interactions with the microenvironment are key drivers of tumorigenesis, while DNA repair mechanisms, drug efflux transporters, and hypoxia-induced stemness and regulatory non-coding RNAs contribute to therapeutic resistance in GB. Progenitor cells play a dual role in GB by promoting aggressiveness and survival challenges.
Figure 1.
Role of Progenitors Cells in Glioblastoma (GB). Cellular plasticity, genetic mutations, self-renewal capacity, and interactions with the microenvironment are key drivers of tumorigenesis, while DNA repair mechanisms, drug efflux transporters, and hypoxia-induced stemness and regulatory non-coding RNAs contribute to therapeutic resistance in GB. Progenitor cells play a dual role in GB by promoting aggressiveness and survival challenges.
3. Microglia in GB: Tumor-Associated Immune Cells
3.1. Microglia and Their Role in Brain Homeostasis
Microglial brain (CNS) resident immune cells are pivotal in maintaining the health and homeostasis of the central nervous system (CNS). Originating from yolk sac progenitors, these unique cells constitute approximately 10-15% of the total cells in the brain. Unlike other immune cells that circulate in the bloodstream, microglia are strategically positioned throughout the CNS, enabling them to respond quickly to various stimuli including injury, infection, and disease [
32,
33]. In their resting state, microglia exhibit a characteristic ramified morphology with long, thin processes that extend into the surrounding environment. This morphology allows them to constantly survey the CNS for changes or damages. Through their highly motile processes, microglia engage in the active surveillance of synapses, contributing to synaptic pruning, a critical process during development that eliminates excess synapses to optimize neural circuit functions [
33,
34]. In addition to synaptic pruning, microglia play an essential role in neurogenesis, supporting the survival and maturation of new neurons. When faced with injury or pathological changes, the microglia undergo rapid activation. This activation leads to a transformation from a resting ramified state to an amoeboid shape, which enhances their ability to engulf cellular debris, dead cells, and pathogens. Activated microglia release a variety of pro-inflammatory cytokines including tumor necrosis factor-alpha (TNF-α), interleukin-1 beta (IL-1β), and interleukin-6 (IL-6). These cytokines play essential roles in recruiting other immune cells to the injury site, thereby initiating the healing process [
33,
35]. While acute microglial activation is crucial for repair and recovery, prolonged or dysregulated activation can lead to chronic neuroinflammation, which is associated with various neurodegenerative diseases, such as Alzheimer’s disease and multiple sclerosis. Thus, maintaining a balance in microglial activation is critical for brain health, underscoring their dual roles as protectors and potential contributors to pathology [
34,
35].
3.2. Microglial Infiltration into the GB Microenvironment
In the context of GB, which is one of the most aggressive forms of brain cancer, the role of microglia has become increasingly complex. GB is characterized by its highly infiltrative nature, extensive cellular heterogeneity, and unique TME that includes not only tumor cells, but also stromal cells, vascular components, and immune cells, particularly microglia. Microglial infiltration into the GB microenvironment is a dynamic process influenced by multiple factors. GB cells release a plethora of signaling molecules, including cytokines, chemokines, and extracellular vesicles, which play critical roles in recruiting and reprogramming microglia [
35,
36,
37]. For instance, the secretion of transforming growth factor-beta (TGF-β) by GB cells is a key factor that drives microglial activation and polarization toward a tumor-promoting phenotype. Once recruited, microglia undergo significant reprogramming, altering their function and phenotype in response to TME. This reprogramming often results in a shift from a protective role to pro-tumor activities [
33,
34]. Factors such as interleukin-10 (IL-10), a cytokine with anti-inflammatory properties, and the release of extracellular vesicles carrying microRNAs (miRNAs) and other bioactive molecules from tumor cells can further influence microglial behavior. This interaction results in a population of tumor-associated microglia that is markedly different from their resting counterparts. These tumor-associated microglia often exhibit features characteristic of the M2 phenotype, which is associated with immune suppression, tissue repair, and promotion of tumor growth [
34,
35]. In this altered state, microglia can support GB progression by enhancing the survival and proliferation of tumor cells, promoting angiogenesis, and facilitating the invasion of the surrounding brain tissue. Moreover, the tumor microenvironment can create a feedback loop that perpetuates microglial activation. As microglia become more involved in supporting tumor growth, they may release additional signals that further enhance GB cell proliferation and survival, creating a vicious cycle that complicates treatment efforts [
37,
38].
3.3. Microglial Polarization and GB Progression
Microglia are well-known for their remarkable plasticity, which enables them to adopt various functional states in response to environmental cues. In the context of GB, microglial polarization can be broadly classified into two main phenotypes, M1 and M2. The balance between these two phenotypes plays a critical role in determining the overall outcome of tumor-host interaction [
35,
38].
M1 Microglia: Pro-Inflammatory Phenotype
M1 microglia are classically activated in response to pro-inflammatory signals and are associated with antitumor immune responses. They secrete a variety of pro-inflammatory cytokines such as IL-12 and interferon-gamma (IFN-γ), which can enhance the activity of other immune cells, including T cells and natural killer (NK) cells. This pro-inflammatory environment can inhibit tumor growth and promote tumor cell apoptosis. However, in GB, the M1 response is often overshadowed by the predominance of M2 microglia, which limits the effectiveness of this anti-tumor response. The transition from M1 to M2 is facilitated by the tumor microenvironment, which is rich in immunosuppressive factors [
34,
39].
3.4. Microglia and Therapeutic Resistance
The interplay between GB cells and microglia significantly contributes to therapeutic resistance, which is a major challenge in the treatment of aggressive cancer. GBs are notoriously resistant to conventional therapies including surgery, chemotherapy, and radiation, and microglia play several roles in this resistance.
Mechanisms of Immune Evasion
One of the primary mechanisms by which microglia contribute to immune evasion is the secretion of growth factors that enhance GB cell survival. For example, insulin-like growth factor 1 (IGF-1) is a potent survival factor that can be released by microglia, promoting resistance to apoptosis in GB cells even when subjected to chemotherapeutic agents [
42,
43]. This interaction effectively enables tumor cells to withstand treatments that would typically induce cell death. Additionally, microglia can upregulate immune checkpoint proteins such as programmed death-ligand 1 (PD-L1), which inhibits T-cell activation and promotes an immunosuppressive environment. By expressing PD-L1, microglia can contribute to the evasion of immune surveillance, allowing GB cells to proliferate unchecked [
44,
45].
Radiation Resistance
Microglia have also been implicated in radiation resistance, which is a significant concern for GB therapy. Following radiation treatment, activated microglia secrete neuroprotective factors and cytokines that aid tumor cell survival. For instance, microglial release of IL-6 can activate signaling pathways in GB cells that promote survival and proliferation, thereby counteracting the intended effects of radiation therapy. This radiation-induced activation of microglia can lead to a vicious cycle, wherein tumor cells stimulate microglial activation and, in turn, activated microglia support the survival of tumor cells. This cycle not only undermines the efficacy of radiation therapy but also creates a challenging environment for the development of novel treatment strategies [
5,
23,
44].
Potential Therapeutic Strategies Targeting Microglia
Given the significant role of microglia in GB progression and therapeutic resistance, targeting these cells is a promising avenue for improving treatment outcomes. Several strategies have been proposed:
CSF1R Inhibitors: Colony-stimulating factor 1 receptor (CSF1R) inhibitors aim to disrupt the recruitment and activation of microglia in the tumor microenvironment. By inhibiting CSF1R, these agents can reduce the population of pro-tumor M2 microglia, potentially restoring a more protective immune environment [
46].
Immune Checkpoint Blockade: Combining immune checkpoint inhibitors with strategies to modulate microglial behavior may enhance anti-tumor immunity. For example, by blocking PD-L1 interactions, immune checkpoint blockade can reinvigorate T-cell responses, potentially overcoming the immunosuppressive effects of tumor-associated microglia [
47,
48].
Reprogramming Microglia: Approaches aimed at reprogramming microglia from the M2 to the M1 phenotype hold promise for enhancing antitumor responses. Therapeutic agents that promote M1 polarization or inhibit M2 signaling pathways could shift the balance toward a more favorable immune environment for combating GB [
49].
Combination therapies: Combination therapies that incorporate standard treatments (such as chemotherapy and radiation) with microglial-targeted therapies could provide synergistic effects, improve treatment efficacy, and overcome resistance mechanisms [
50,
51].
4. Non-coding RNAs (ncRNAs) in GB: Key Regulators of Pathogenesis and Resistance
4.1. NcRNAs
ncRNAs are a vital component of the genome, encompassing a wide array of RNA molecules that do not translate into proteins but play crucial regulatory roles in gene expression and cellular functions. Their importance in various biological processes, particularly cancer, has garnered significant attention in recent years. Below, we explore the types of ncRNAs, their mechanisms of action, and their implications in diseases, such as GB, cancer, and TB [
52,
53,
54,
55].
They are short, typically 20-22 nucleotides in length, single-stranded RNA molecules that primarily function in post-transcriptional regulation. MiRNAs bind to complementary sequences in target messenger RNAs (mRNAs), leading to mRNA degradation or translational repression. This process modulates gene expression and can have profound effects on various cellular functions including proliferation, differentiation, and apoptosis. MicroRNAs are emerging as vital regulators in GB, influencing many aspects of tumor biology [
52,
56,
57]. Research has identified several key miRNAs that are significantly implicated in GB pathogenesis, including miR-21: Often referred to as an "oncomiR," miR-21 is frequently overexpressed in GB tissues and is associated with aggressive tumor behavior [
58]. It promotes cell proliferation and invasion by targeting tumor suppressor genes, such as phosphatase and tensin homolog (PTEN), and RECK (reversion-inducing cysteine-rich protein with Kazal motifs). Upregulation of miR-21 correlates with poorer patient prognosis, highlighting its potential as a therapeutic target. miR-10b: This miRNA enhances the invasive properties of GB cells. By downregulating HOXD10, a gene known for its tumor-suppressive functions, miR-10b facilitates tumor cell migration and invasion, thereby contributing to the aggressive nature of GB [
59]. miR-34a: Acting as a tumor suppressor, miR-34a regulates critical pathways involved in cell cycle control and apoptosis. Its expression is frequently downregulated in GB, leading to unchecked cell proliferation and enhanced survival of tumor cells in response to stress [
60,
61].
The mechanisms by which miRNAs exert their effects on GB include the following.
Regulation of Proliferation: MiRNAs, such as miR-34a target genes, are involved in cell cycle progression. By inhibiting these targets, miR-34a can prevent tumor cells from progressing through the cell cycle, thereby reducing their proliferation. Conversely, loss of miR-34a expression can lead to enhanced cell growth. Promotion of Invasion: MiRNAs such as miR-10b facilitate GB invasion by targeting cell adhesion molecules and extracellular matrix components [
60,
61]. This regulation allows tumor cells to detach from their primary site and invade the surrounding tissues, a hallmark of GB aggressiveness. Maintenance of Stemness: MiRNAs are crucial for the maintenance of cancer stem cell characteristics that are linked to tumor recurrence and treatment resistance. For example, miR-21 promotes stemness in GB cells, enabling them to survive in harsh microenvironments and resist therapy [
60,
62].
MicroRNAs also play significant roles in GB resistance to therapies:
Drug Efflux Mechanisms: miRNAs influence the expression of ATP-binding cassette (ABC) transporters, which are responsible for drug efflux. The overexpression of specific miRNAs can enhance the expression of these transporters, leading to decreased intracellular concentrations of chemotherapeutic agents and reduced drug efficacy.
Evasion of Apoptosis: By down-regulating pro-apoptotic factors and up-regulating anti-apoptotic factors, miRNAs enable GB cells to evade programmed cell death. This mechanism is particularly important in the context of chemotherapy and radiation, where the induction of apoptosis is a primary therapeutic goal [
60,
63].
LncRNAs and Their Role in GB: Defined as ncRNAs longer than 200 nucleotides, lncRNAs exhibit a wide range of biological activities. They can interact with chromatin, transcription factors, and other RNA molecules, influencing gene expression at multiple levels. LncRNAs are involved in regulating cellular processes such as cell cycle progression, differentiation, and responses to stress [
53,
54,
55]. LncRNAs are increasingly recognized for their roles in GB pathogenesis. Key lncRNAs involved in GB include:
HOTAIR: Homeobox transcript antisense intergenic RNA (HOTAIR) is a well-studied lncRNA associated with poor prognosis in GB[
64,
65]. HOTAIR facilitates tumor metastasis through chromatin remodeling, which alters the expression of genes involved in invasion and migration. Its overexpression correlates with increased tumor aggressiveness and enhanced metastatic potential [
66].
MALAT1: Metastasis-associated lung adenocarcinoma transcript 1 (MALAT1) is overexpressed in GB and plays a critical role in regulating cell proliferation and migration. MALAT1 acts as a sponge for several miRNAs, thereby modulating the expression of target genes that control cell cycle progression and tumor growth [
67].
NEAT1: Nuclear paraspeckle assembly transcript 1 (NEAT1) is essential for the formation of paraspeckles, nuclear structures involved in gene expression regulation. NEAT1 contributes to GB progression by promoting cell survival, influencing the stress response, and modulating immune responses within the tumor microenvironment [
68,
69].
LncRNAs exert their regulatory functions through several mechanisms: Tumor Growth: By interacting with chromatin-modifying complexes, lncRNAs like HOTAIR can enhance the expression of oncogenes, driving tumor growth. They can also recruit transcription factors to specific gene loci, influencing the transcriptional landscape of GB cells. Stem Cell Maintenance: Certain lncRNAs are involved in maintaining cancer stem cell populations, which contribute to tumor heterogeneity and the capacity for self-renewal. This characteristic is crucial for the resilience of GB against therapeutic interventions. Immune Modulation: LncRNAs can influence the immune landscape within the tumor microenvironment. By regulating the expression of cytokines and immune checkpoint molecules, lncRNAs may affect the recruitment and activity of immune cells, allowing GB to evade immune surveillance [
70].
Circular RNAs and Their Emerging Role in GB (circRNAs): These are unique, covalently closed RNA molecules formed by back-splicing of exons. CircRNAs are often stable and resistant to degradation, allowing them to serve as important regulators in the cell. They can function as sponges for miRNAs, binding to them and preventing their interaction with target mRNAs, thus modulating gene expression [
71,
72]. The study of ncRNAs has revealed their critical involvement in the pathogenesis of various cancers, including GB, where they play key roles in tumor growth, metastasis, and therapeutic resistance. Circular RNAs (circRNAs) are a novel class of ncRNAs that have garnered attention for their unique structures and regulatory roles in GB. Key examples of circRNAs include circ-FBXW7: This CircRNA acts as a sponge for miR-197, thus enhancing the expression of FBXW7, a tumor suppressor that plays a vital role in regulating cell proliferation and survival by down-regulating miR-197 [
73,
74]. circ-FBXW7 promotes the degradation of oncogenic proteins, thereby inhibiting GB progression. circHIPK3: Another important CircRNA, circHIPK3, is involved in regulating cell proliferation and apoptosis in GB. It sponges several miRNAs, influencing the expression of genes that control these critical cellular processes [
75].
Mechanism of regulatory role of CircRNAs in sponging miRNA: CircRNAs primarily function as miRNA sponges, sequestering miRNAs and preventing them from binding to their target mRNAs. This sponging activity can lead to the upregulation of oncogenes or downregulation of tumor suppressors, thereby impacting GB biology. By modulating miRNA availability, circRNAs can significantly influence gene expression and contribute to the aggressive nature of GB.
4.5. NcRNAs in Therapeutic Resistance
The role of ncRNAs in therapeutic resistance is a critical area of research in GB. ncRNAs influence resistance mechanisms through various pathways:
Chemotherapy Resistance: NcRNAs can regulate the expression of genes involved in drug metabolism, efflux, and apoptosis. For instance, certain lncRNAs can enhance the expression of drug transporters, leading to decreased efficacy of chemotherapeutic agents. Additionally, miRNAs may target pro-apoptotic genes, promoting cell survival and resistance to chemotherapy [
60,
63,
65].
Radiotherapy Resistance: NcRNAs are implicated in the cellular response to radiation therapy. Some miRNAs can enhance DNA repair pathways, allowing GB cells to survive radiation exposure and continue proliferating [
65].
Targeted Therapy Resistance: NcRNAs can also influence the expression of targets for specific therapies. For example, lncRNAs may modulate the expression of receptor tyrosine kinases, affecting the sensitivity of GB to targeted therapies [
76].
Exosomal ncRNAs in GB: Exosomal ncRNAs play a crucial role in regulating GB pathways, and influencing tumor progression and therapeutic responses. For instance, miR-21 is often upregulated in GB, promoting cell proliferation by targeting tumor suppressor genes like PTEN, which enhances survival and growth. Similarly, miR-221/222 inhibits pro-apoptotic factors, further aiding cell survival, while exosomal lncRNAs such as H19 can drive cell cycle progression [
76,
77]. In terms of invasion, miR-10b enhances the migratory capacity of GB cells by targeting genes involved in cell adhesion, and exosomes can promote epithelial-mesenchymal transition (EMT), facilitating local invasion. Exosomal ncRNAs also contribute to angiogenesis, with miR-125b promoting blood vessel formation, and they play a role in immune evasion by modulating immune responses through miR-155, which helps the tumor escape immune detection [
63,
76]. Additionally, ncRNAs are implicated in therapeutic resistance; for example, downregulation of miR-34a can lead to increased resistance to chemotherapy. Finally, ncRNAs can enhance cancer stem cell properties, contributing to tumor maintenance and recurrence [
63]. The complex interactions of exosomal ncRNAs in these pathways highlight their potential as targets for therapeutic intervention and as biomarkers for GB management.
5. Interplay Between Progenitor Cells, Microglia, and NcRNAs in GB [AH]
5.1. Progenitor Cell-Microglia Cross-talk
The interaction between progenitor cells and microglia within the GB-TME is a crucial factor driving tumor progression [
78]. GSCs, which originate from transformed neural progenitor cells, actively influence microglial behavior through direct and indirect signaling mechanisms [
79]. Roles of progenitor cells, microglia, and ncRNAs in pathogenesis and therapeutic resistance in GB are summarized in
Table 1.
Progenitor Cell Influence on Microglia: Progenitor cells release various signaling molecules, including chemokines and cytokines, which affect microglial polarization [
80]. This polarization shifts microglia towards a tumor-supportive phenotype, often described as M2-like polarization [
33]. These M2-polarized microglia support the immunosuppressive and pro-tumorigenic environment by releasing factors that promote glioma growth and inhibit anti-tumor immune responses [
33,
35].
Microglial Support for GSCs: Microglia, in response to progenitor cell signals, secrete various cytokines (IL-6, TGF-β) and growth factors (CSF-1, VEGF) that enhance the survival, self-renewal, and proliferation of GSCs [
32,
35]. This reciprocal interaction between progenitor cells and microglia fosters a symbiotic relationship where both cell types promote each other’s survival, facilitating tumor growth, invasion, and resistance to therapies [
35,
36,
80].
5.2. ncRNAs as Mediators of Cellular Interactions
NcRNAs, which include microRNAs (miRNAs), lncRNAs, and circular RNAs (circRNAs), have emerged as key regulators of cellular communication within the GB-TME [
64]. They modulate the cross-talk between progenitor cells and microglia, influencing the course of GB development [
64,
65].
ncRNA-Mediated Modulation of Communication: ncRNAs can act as molecular bridges, modulating signaling pathways and transcriptional networks between progenitor cells and microglia [
66,
81]. For instance, miRNAs such as miR-124 and miR-21 are known to regulate microglial polarization and progenitor cell behavior, either suppressing anti-tumor responses or promoting the M2-like phenotype that supports tumor growth [
56,
57,
81].
Regulatory Feedback Loops: NcRNAs can establish complex regulatory feedback loops. For example, miRNAs may inhibit the expression of specific transcription factors that would otherwise limit progenitor cell proliferation, while lncRNAs and circRNAs may act as "sponges" for these miRNAs, reducing their activity and thus maintaining the stem-like state of glioma cells. These regulatory interactions create a finely tuned system that promotes GB progression [
57,
71,
77,
82].
Impact on Tumor Microenvironment: NcRNAs not only affect individual cells but also modulate the broader TME [
83]. They influence the secretion of cytokines and growth factors, reshape immune cell recruitment, and alter the extracellular matrix composition, thereby facilitating tumor-promoting conditions. The dysregulation of ncRNAs amplifies cellular cross-talk, reinforcing GB malignancy [
56,
65,
77].
5.3. Implications for Tumor Progression and Resistance
The intricate interplay between progenitor cells, microglia, and ncRNAs creates a feedback system that accelerates GB progression and strengthens therapeutic resistance.
Tumor Progression: The continuous cross-talk between progenitor cells and microglia, mediated by ncRNAs, enhances the invasive capacity of GSCs, and promotes tumor heterogeneity [
23,
35,
69]. This complex cellular and molecular environment supports the creation of a highly adaptive and aggressive tumor. GSCs, supported by microglial-derived factors and ncRNA signaling, maintain their self-renewal and invasive properties, contributing to the relentless growth of GB [
5,
15,
19,
20].
Synergistic Roles in Therapeutic Resistance: This tripartite interaction is also a major contributor to therapy resistance. Progenitor cells and GSCs exhibit high plasticity, which allows them to survive conventional treatments such as radiotherapy and chemotherapy [
20,
27,
84,
85]. Microglial-derived cytokines further protect these cells from therapy-induced apoptosis [
35]. Moreover, ncRNAs can upregulate resistance-related genes, such as those involved in DNA repair and drug efflux, reinforcing the tumor's ability to withstand therapeutic pressure [
55,
82]. The synergistic action of these elements thus creates a robust, multi-layered defense against current treatment strategies [
22,
86]
Table 1.
Roles of progenitor cells, microglia, and ncRNAs in GB pathogenesis and resistance.
Table 1.
Roles of progenitor cells, microglia, and ncRNAs in GB pathogenesis and resistance.
Progenitor Cells |
Types |
Role in GB Pathogenesis |
Mechanism of Resistance |
Ref |
Neural Progenitor Cells (NPCs) |
Provide cells with self-renewal and differentiation potential; Mutations can trigger tumorigenic transformation. |
High drug-efflux pump activity, enhanced DNA repair, and maintenance of stemness properties. |
[87] |
Glioma Stem-Like Cells (GSCs) |
Promote tumor growth and recurrence with stem-like properties and contribute to GB heterogeneity. |
Quiescence, increased DNA repair, hypoxic niche protection |
[88] |
Oligodendrocyte Progenitor Cells (OPCs) |
Potential cell of origin in the proneural GB; Dysregulation of OPCs promotes tumor progression |
Activation of PI3K/Akt/mTOR signaling pathways; Adaptation to microenvironmental stressors |
[89,90] |
Mesenchymal Progenitor Cells (MPCs) |
Differentiation into tumor-associated stromal cells; supports aggressive growth of the mesenchymal subtype. |
Enhance invasion, angiogenesis, and immune evasion |
[91] |
Endothelial Progenitor Cells (EPCs) |
Support neovascularization, increase blood supply to the tumor, and facilitate invasion. |
Maintain a hypoxic environment, protect from radiotherapy, and support angiogenesis. |
[92] |
Microglia |
Tumor-Associated Microglia/Macrophages (TAMs) |
Support tumor growth through secretion of growth factors and cytokines; promote GB invasion and vascularization. |
Immunosuppressive environment, increased secretion of anti-inflammatory cytokines |
[93] |
M1 Microglia (Pro-inflammatory) |
Transiently suppress GB progression by releasing pro-inflammatory cytokines (e.g., TNF-α, IL-1β) |
Reduced activity due to tumor-derived immunosuppressive signaling and metabolic reprogramming |
[94] |
M2 Microglia (Anti-inflammatory) |
Promote tumor growth by enhancing angiogenesis, immunosuppression, and extracellular matrix remodeling. |
High resistance through secretion of growth factors (e.g. TGF-β) and anti-inflammatory cytokines |
[33,95] |
Reactive Microglia |
Activated in response to GB-induced inflammation; secrete factors promoting GB proliferation and matrix remodeling. |
Secrete matrix metalloproteinases (MMPs) that support tumor invasion |
[96] |
Perivascular Microglia |
Facilitate the invasion of GB cells along blood vessels and contribute to the formation of the perivascular niche. |
Protect tumor cells by promoting a supportive niche and maintaining BBB integrity. |
[97] |
Glioma-Associated Microglia (GAMs) |
Specialized microglia in GB; interact closely with GSCs and tumor cells to promote proliferation and invasion |
Promote therapeutic resistance by maintaining stemness and supporting immune evasion. |
[98] |
Non-Coding RNAs |
miR-21 (microRNA-21) |
Promotes GB cell proliferation, and invasion, and inhibits apoptosis by targeting tumor suppressor genes (e.g., PTEN, PDCD4). |
Increases resistance by activating anti-apoptotic signaling pathways and reduces sensitivity to chemotherapy |
[99,100] |
miR-10b |
Facilitates tumor cell invasion and promotes stem cell-like properties |
Induces therapeutic resistance through upregulation of pro-survival pathways and inhibition of apoptosis |
[101] |
lncRNA HOTAIR |
Enhances GB cell migration, invasion, and epithelial-to-mesenchymal transition (EMT) |
Contributes to radioresistance by promoting DNA damage repair and enhancing stemness properties |
[102,103,104] |
lncRNA MALAT1 |
Supports tumor growth and angiogenesis through modulation of gene expression |
Enhances resistance by modulating autophagy and promoting anti-apoptotic mechanisms |
[105,106,107] |
circRNA circHIPK3 |
Promotes GB proliferation and invasiveness by sponging tumor-suppressive miRNAs (e.g., miR-124) |
Mediates chemoresistance through PI3K/AKT signaling activation |
[75,108] |
SNHG12 (Small Nucleolar RNA Host Gene 12) |
Enhances GB proliferation, migration, and immune evasion |
Increases resistance by modulating immune checkpoints and enhancing anti-apoptotic signaling |
[109,110] |
miR-155 |
Promotes tumor progression by targeting tumor suppressor genes and facilitating immunosuppression |
Contributes to radioresistance and chemoresistance by improving DNA repair mechanisms |
[111,112] |
6. Therapeutic Implications and Future Directions
6.1. Current Therapeutic Strategies
GB treatment remains a significant challenge, particularly due to the involvement of progenitor cells, microglia, and ncRNAs in tumor progression and therapeutic resistance [
86,
113,
114]. Current GB management includes surgical resection followed by adjuvant radiotherapy with temozolomide, an alkylating agent (the most widely used chemotherapeutic drug for glioma management), and followed by chemotherapy alone [
3,
4,
115,
116]. Emerging therapeutic strategies target progenitor cells, microglia, and ncRNAs to disrupt the GB microenvironment and limit tumor growth [
30].
Treatments Targeting Progenitor Cells: Therapeutic approaches aimed at progenitor cells and GSCs primarily focus on differentiation therapy and the inhibition of key signaling pathways [
26,
117,
118]. Differentiation therapy attempts to drive GSCs into more differentiated, less tumorigenic states, thereby reducing their proliferative capacity [
119,
120]. Drugs that target critical signaling pathways, such as the Notch, Wnt, and Hedgehog (SHH) pathways, aim to inhibit the self-renewal and maintenance of stem cells [
121,
122].
Microglia-Targeted Therapies: Therapies targeting microglia seek to reprogram these immune cells from a pro-tumorigenic to an anti-tumorigenic state [
48,
123]. One promising approach involves using CSF1R inhibitors to block signals that promote microglial support for GB growth. In addition, strategies to polarize microglia towards an M1-like phenotype (anti-tumor) or prevent their recruitment into the tumor microenvironment are being explored [
33,
46].
ncRNA-Based Therapies: The therapeutic potential of ncRNAs lies in their regulatory roles in gene expression and tumorigenesis [
124,
125]. Antisense oligonucleotides (ASOs), miRNA mimics, and miRNA inhibitors have been developed to target oncogenic ncRNAs or restore the function of tumor-suppressive ncRNAs [
126,
127,
128]. For instance, miRNA mimics can be introduced to restore miRNA levels that suppress glioma growth, while inhibitors can block oncogenic miRNAs that contribute to tumorigenesis. Clinical trials are ongoing to assess the efficacy of ncRNA-based therapies in GB [
126,
129,
130,
131,
132,
133].
6.2. Challenges in Targeting the Progenitor Cells-Microglia-ncRNA Axis
Despite advances in therapeutic approaches, targeting the progenitor cells-microglia-ncRNA axis presents several significant challenges:
Blood-Brain Barrier (BBB): The BBB is a major obstacle in delivering therapeutic agents to the brain. Its highly selective permeability limits the efficacy of many treatments, including small molecule inhibitors, antibodies, and nucleic acid-based therapies such as ASOs and miRNA mimics. Overcoming the BBB remains a critical hurdle in developing effective GB therapies [
134,
135,
136].
Tumor Heterogeneity: GB is characterized by extreme tumor heterogeneity, not only in its genetic and epigenetic landscape but also in the behavior of GSCs and microglia [
84,
137]. This heterogeneity results in diverse treatment responses, with different tumor cell populations exhibiting varying levels of resistance [
2]. Microglia and GSCs can adapt to therapeutic pressure, leading to recurrence even after aggressive treatment. These adaptive responses significantly complicate the development of effective therapies that can target all tumor subpopulations [
5,
20,
35].
6.3. Emerging Therapeutic Approaches
To overcome these challenges, several emerging therapeutic approaches are being developed, to address the multifaceted nature of GB pathogenesis.
Combination Therapies: Single-agent therapies have shown limited success due to the complex and adaptive nature of GB [
50]. Combination therapies, which target multiple components of the tumor microenvironment simultaneously, are being explored to improve treatment outcomes [
50,
51,
138]. For instance, co-targeting progenitor cell pathways (e.g., Notch or Wnt) along with microglia modulation (CSF1R inhibitors or immunomodulators) and ncRNA-based interventions could potentially address both the cellular and molecular components driving GB [
46,
121,
139].
Personalized Therapies Based on ncRNA Profiles: The advent of precision medicine offers the possibility of tailoring treatments based on the specific ncRNA expression profiles of individual tumors [
140]. Personalized therapeutic approaches could involve the use of miRNA mimics or inhibitors specifically chosen to target the dysregulated ncRNAs driving the patient's tumor [
141]. This approach could help overcome tumor heterogeneity by targeting the unique molecular characteristics of each tumor [
72,
142,
143].
Advances in Drug Delivery Systems: Recent advances in nanoparticle and exosome-based delivery systems show promise in enhancing drug delivery across the BBB and directly targeting glioma cells [
135,
144]. Nanoparticles can be engineered to carry therapeutic agents such as pathway inhibitors or ncRNAs, improving their bioavailability and specificity [
145,
146]. Exosomes, natural carriers of RNA and proteins, have emerged as a potential vehicle for delivering miRNA-based therapies to the tumor site, offering a novel approach to overcoming the BBB [
147,
148,
149,
150].
7. Conclusion
Glioma stem-like cells (GSCs), which arise from progenitor cells, are central to tumor initiation, recurrence, and resistance, due to their plasticity and self-renewal capabilities. The transformation of progenitor cells into GSCs, driven by genetic mutations and epigenetic changes, creates a pool of tumor-initiating cells that exhibit resistance to conventional therapies. Microglia, co-opted by GB cells, play a pivotal role in creating an immunosuppressive microenvironment that fosters tumor growth. The cross-talk between microglia and GSCs, mediated by cytokines and growth factors, enhances GSC survival and therapy resistance. Furthermore, ncRNAs, particularly microRNAs (miRNAs) and long non-coding RNAs (lncRNAs), regulate the expression of key genes involved in tumor growth and the cellular interactions between progenitor cells and microglia. These ncRNAs also contribute to the maintenance of the stem-like phenotype in GSCs and modulate immune responses, further promoting tumor progression and resistance to treatments.
The complexity of GB, with its diverse and interconnected cellular and molecular components, necessitates the development of multi-targeted therapeutic approaches. Future research should focus on understanding the dynamic interactions between progenitor cells, microglia, and ncRNAs. Identifying the precise mechanisms by which these components communicate within the tumor microenvironment is crucial for designing more effective therapies. One promising avenue is the personalization of therapies based on individual tumor profiles, particularly ncRNA expression patterns. Moreover, combination therapies that simultaneously target GSCs, microglia, and ncRNAs hold the potential for overcoming resistance mechanisms. Advancements in drug delivery systems that can effectively cross the blood-brain barrier, such as nanoparticles and exosomes, will be pivotal in improving the therapeutic efficacy of these multi-targeted approaches.
Understanding the roles of progenitor cells, microglia, and ncRNAs in GB pathogenesis opens new therapeutic avenues that go beyond conventional treatments. Targeting the progenitor cells-microglia-ncRNA axis can potentially disrupt the cellular and molecular networks that drive tumor growth and therapy resistance. Novel therapies such as pathway inhibitors for progenitor cells, immunomodulatory agents targeting microglia, and ncRNA-based therapeutics are being developed to improve patient outcomes. These therapies, combined with advanced drug delivery technologies, could lead to more effective treatments that minimize resistance, slow tumor progression, and ultimately, extend survival in patients with GB.
Conflicts of Interest
All authors hereby confirm that there is no conflict of interest to declare.
Ethical Approval
Since the current study did not include any human or animal material, the ethics committee does not need to approve this publication.
Data Availability Statement
The article does not fall under the category of data sharing because no datasets were created or examined in this study.
Author Contributions
Conceptualization: Adil Husain; Data curation: Adil Husain; Firoz Ahmad; Resources: Software, Supervision: Adil Husain; Firoz Ahmad; Tarun Kumar Upadhyay; Roles/Writing - Original draft: Adil Husain; Firoz Ahmad; Sandeep Pandey
Writing - Review & Editing: Adil Husain; Firoz Ahmad; Sandeep Pandey; Sojin Kang; Min; Choi; Jinwon Choi; Moon Nyeo Park; Bonglee Kim.
Funding
This research was supported by Basic Science Research Program through the National Research Foundation of Korea (NRF) funded by the Ministry of Education (NRF-2020R1I1A2066868), the National Research Foundation of Korea (NRF) grant funded by the Korea government (MSIT) (No. 2020R1A5A2019413), a grant of the Korea Health Technology R&D Project through the Korea Health Industry Development Institute (KHIDI), funded by the Ministry of Health & Welfare, Republic of Korea (Grant Number: RS-2020-KH087790) and the National Research Foundation of Korea (NRF) grant funded by the Korea government (MSIT) (RS-2024-00350362).
References
- Ostrom, Q.T.; Price, M.; Neff, C.; Cioffi, G.; Waite, K.A.; Kruchko, C.; Barnholtz-Sloan, J.S. CBTRUS Statistical Report: Primary Brain and Other Central Nervous System Tumors Diagnosed in the United States in 2016—2020. Neuro-Oncology 2023, 25, iv1–iv99. [CrossRef]
- Nicholson, J.G.; Fine, H.A. Diffuse Glioma Heterogeneity and Its Therapeutic Implications. Cancer Discov 2021, 11, 575–590. [CrossRef]
- Stupp, R.; Weller, M.; Belanger, K.; Bogdahn, U.; Ludwin, S.K.; Lacombe, D.; Mirimanoff, R.O. Radiotherapy plus Concomitant and Adjuvant Temozolomide for Glioblastoma. The New England Journal of Medicine 2005, 10.
- Stupp, R.; Brada, M.; van den Bent, M.J.; Tonn, J.C.; Pentheroudakis, G. High-Grade Glioma: ESMO Clinical Practice Guidelines for Diagnosis, Treatment and Follow-Up. Annals of Oncology 2014, 25, 93–101. [CrossRef]
- Bao, S.; Wu, Q.; McLendon, R.E.; Hao, Y.; Shi, Q.; Hjelmeland, A.B.; Dewhirst, M.W.; Bigner, D.D.; Rich, J.N. Glioma Stem Cells Promote Radioresistance by Preferential Activation of the DNA Damage Response. Nature 2006, 444, 756–760. [CrossRef]
- Martínez-Cerdeño, V.; Noctor, S.C. Neural Progenitor Cell Terminology. Frontiers in Neuroanatomy 2018, 12, 104. [CrossRef]
- Finkel, Z.; Esteban, F.; Rodriguez, B.; Fu, T.; Ai, X.; Cai, L. Diversity of Adult Neural Stem and Progenitor Cells in Physiology and Disease. Cells 2021, 10, 2045. [CrossRef]
- Zhang, S.; Xiao, X.; Yi, Y.; Wang, X.; Zhu, L.; Shen, Y.; Lin, D.; Wu, C. Tumor Initiation and Early Tumorigenesis: Molecular Mechanisms and Interventional Targets. Sig Transduct Target Ther 2024, 9, 1–36. [CrossRef]
- Louis, D.N.; Perry, A.; Wesseling, P.; Brat, D.J.; Cree, I.A.; Figarella-Branger, D.; Hawkins, C.; Ng, H.K.; Pfister, S.M.; Reifenberger, G.; et al. The 2021 WHO Classification of Tumors of the Central Nervous System: A Summary. Neuro-Oncology 2021, 23, 1231–1251. [CrossRef]
- Avsar, T.; Kose, T.B.; Oksal, M.D.; Turan, G.; Kilic, T. IDH1 Mutation Activates mTOR Signaling Pathway, Promotes Cell Proliferation and Invasion in Glioma Cells. Molecular Biology Reports 2022, 49, 9241–9249. [CrossRef]
- Bhavya, B.; Anand, C.R.; Madhusoodanan, U.K.; Rajalakshmi, P.; Krishnakumar, K.; Easwer, H.V.; Deepti, A.N.; Gopala, S. To Be Wild or Mutant: Role of Isocitrate Dehydrogenase 1 (IDH1) and 2-Hydroxy Glutarate (2-HG) in Gliomagenesis and Treatment Outcome in Glioma. Cellular and Molecular Neurobiology 2020, 40, 53–63. [CrossRef]
- Parsons, D.W.; Jones, S.; Zhang, X.; Lin, J.C.-H.; Leary, R.J.; Angenendt, P.; Mankoo, P.; Carter, H.; Siu, I.-M.; Gallia, G.L.; et al. An Integrated Genomic Analysis of Human Glioblastoma Multiforme. Science 2008, 321, 1807. [CrossRef]
- Kreth, S.; Thon, N.; Kreth, F.W. Epigenetics in Human Gliomas. Cancer Letters 2014, 342, 185–192. [CrossRef]
- Pandey, S.; Singh, R.; Habib, N.; Tripathi, R.; Kushwaha, R.; Mahdi, A. Regulation of Hypoxia Dependent Reprogramming of Cancer Metabolism: Role of HIF-1 and Its Potential Therapeutic Implications in Leukemia. Asian Pac J Cancer Prev 2024, 25, 1121–1134. [CrossRef]
- Bikfalvi, A.; Da Costa, C.A.; Avril, T.; Barnier, J.-V.; Bauchet, L.; Brisson, L.; Cartron, P.F.; Castel, H.; Chevet, E.; Chneiweiss, H.; et al. Challenges in Glioblastoma Research: Focus on the Tumor Microenvironment. Trends in Cancer 2023, 9, 9–27. [CrossRef]
- El-Tanani, M.; Rabbani, S.A.; Babiker, R.; Rangraze, I.; Kapre, S.; Palakurthi, S.S.; Alnuqaydan, A.M.; Aljabali, A.A.; Rizzo, M.; El-Tanani, Y.; et al. Unraveling the Tumor Microenvironment: Insights into Cancer Metastasis and Therapeutic Strategies. Cancer Letters 2024, 591, 216894. [CrossRef]
- Emami Nejad, A.; Najafgholian, S.; Rostami, A.; Sistani, A.; Shojaeifar, S.; Esparvarinha, M.; Nedaeinia, R.; Haghjooy Javanmard, S.; Taherian, M.; Ahmadlou, M.; et al. The Role of Hypoxia in the Tumor Microenvironment and Development of Cancer Stem Cell: A Novel Approach to Developing Treatment. Cancer Cell International 2021, 21, 62. [CrossRef]
- Prager, B.C.; Bhargava, S.; Mahadev, V.; Hubert, C.G.; Rich, J.N. Glioblastoma Stem Cells: Driving Resiliency through Chaos. Trends in cancer 2020, 6, 223. [CrossRef]
- Gimple, R.C.; Bhargava, S.; Dixit, D.; Rich, J.N. Glioblastoma Stem Cells: Lessons from the Tumor Hierarchy in a Lethal Cancer. Genes Dev. 2019, 33, 591–609. [CrossRef]
- Alves, A.L.V.; Gomes, I.N.F.; Carloni, A.C.; Rosa, M.N.; Da Silva, L.S.; Evangelista, A.F.; Reis, R.M.; Silva, V.A.O. Role of Glioblastoma Stem Cells in Cancer Therapeutic Resistance: A Perspective on Antineoplastic Agents from Natural Sources and Chemical Derivatives. Stem Cell Res Ther 2021, 12, 206. [CrossRef]
- Li, Y.; Wang, J.; Song, S.-R.; Lv, S.-Q.; Qin, J.; Yu, S.-C. Models for Evaluating Glioblastoma Invasion along White Matter Tracts. Trends in Biotechnology 2024, 42, 293–309. [CrossRef]
- Eckerdt, F.; Platanias, L.C. Emerging Role of Glioma Stem Cells in Mechanisms of Therapy Resistance. Cancers 2023, 15, 3458. [CrossRef]
- Gu, J.; Mu, N.; Jia, B.; Guo, Q.; Pan, L.; Zhu, M.; Zhang, W.; Zhang, K.; Li, W.; Li, M.; et al. Targeting Radiation-Tolerant Persister Cells as a Strategy for Inhibiting Radioresistance and Recurrence in Glioblastoma. Neuro-Oncology 2022, 24, 1056–1070. [CrossRef]
- Dymova, M.A.; Kuligina, E.V.; Richter, V.A. Molecular Mechanisms of Drug Resistance in Glioblastoma. International Journal of Molecular Sciences 2021, 22, 6385. [CrossRef]
- Auffinger, B.; Spencer, D.; Pytel, P.; Ahmed, A.U.; Lesniak, M.S. The Role of Glioma Stem Cells in Chemotherapy Resistance and Glioblastoma Multiforme Recurrence. Expert review of neurotherapeutics 2015, 15, 741. [CrossRef]
- Kang, H.; Lee, H.; Kim, D.; Kim, B.; Kang, J.; Kim, H.Y.; Youn, H.; Youn, B. Targeting Glioblastoma Stem Cells to Overcome Chemoresistance: An Overview of Current Therapeutic Strategies. Biomedicines 2022, 10, 1308. [CrossRef]
- da Silva-Diz, V.; Lorenzo-Sanz, L.; Bernat-Peguera, A.; Lopez-Cerda, M.; Muñoz, P. Cancer Cell Plasticity: Impact on Tumor Progression and Therapy Response. Seminars in Cancer Biology 2018, 53, 48–58. [CrossRef]
- Yuan, S.; Norgard, R.J.; Stanger, B.Z. Cellular Plasticity in Cancer. Cancer Discov 2019, 9, 837–851. [CrossRef]
- Jin, J.; Grigore, F.; Chen, C.C.; Li, M. Self-renewal Signaling Pathways and Differentiation Therapies of Glioblastoma Stem Cells (Review). Int J Oncol 2021, 59, 45. [CrossRef]
- Safa, A.R.; Saadatzadeh, M.R.; Cohen-Gadol, A.A.; Pollok, K.E.; Bijangi-Vishehsaraei, K. Emerging Targets for Glioblastoma Stem Cell Therapy. J Biomed Res 2016, 30, 19–31. [CrossRef]
- Cherry, A.E.; Stella, N. G Protein-Coupled Receptors as Oncogenic Signals in Glioma: Emerging Therapeutic Avenues. Neuroscience 2014, 0, 222. [CrossRef]
- Da, M.; D, G.-N. Microglial Dynamics During Human Brain Development. Frontiers in immunology 2018, 9. [CrossRef]
- Orihuela, R.; McPherson, C.A.; Harry, G.J. Microglial M1/M2 Polarization and Metabolic States. British Journal of Pharmacology 2015, 173, 649. [CrossRef]
- Kuntzel, T.; Bagnard, D. Manipulating Macrophage/Microglia Polarization to Treat Glioblastoma or Multiple Sclerosis. Pharmaceutics 2022, 14, 344. [CrossRef]
- D, H.; Dh, G.; H, K. The Role of Microglia and Macrophages in Glioma Maintenance and Progression. Nature neuroscience 2016, 19. [CrossRef]
- Li, W.; Graeber, M.B. The Molecular Profile of Microglia under the Influence of Glioma. Neuro-Oncology 2012, 14, 958. [CrossRef]
- Ling, E.A.; Wong, W.C. The Origin and Nature of Ramified and Amoeboid Microglia: A Historical Review and Current Concepts. Glia 1993, 7, 9–18. [CrossRef]
- Szulzewsky, F.; Pelz, A.; Feng, X.; Synowitz, M.; Markovic, D.; Langmann, T.; Holtman, I.R.; Wang, X.; Eggen, B.J.L.; Boddeke, H.W.G.M.; et al. Glioma-Associated Microglia/Macrophages Display an Expression Profile Different from M1 and M2 Polarization and Highly Express Gpnmb and Spp1. PLoS ONE 2015, 10, e0116644. [CrossRef]
- Zeiner, P.S.; Preusse, C.; Blank, A.-E.; Zachskorn, C.; Baumgarten, P.; Caspary, L.; Braczynski, A.K.; Weissenberger, J.; Bratzke, H.; Reiß, S.; et al. MIF Receptor CD74 Is Restricted to Microglia/Macrophages, Associated with a M1-Polarized Immune Milieu and Prolonged Patient Survival in Gliomas. Brain Pathology 2014, 25, 491. [CrossRef]
- Sj, C.; E, E.; K, D.; Er, S.; Bl, W.; Mh, S.; Je, S. Microglial Stimulation of Glioblastoma Invasion Involves Epidermal Growth Factor Receptor (EGFR) and Colony Stimulating Factor 1 Receptor (CSF-1R) Signaling. Molecular medicine (Cambridge, Mass.) 2012, 18. [CrossRef]
- I, B.; S, T.; W, P. Microglia Promote Glioma Migration. Acta neuropathologica 2002, 103. [CrossRef]
- Markovic, D.S.; Glass, R.; Synowitz, M.; Rooijen, N. van; Kettenmann, H. Microglia Stimulate the Invasiveness of Glioma Cells by Increasing the Activity of Metalloprotease-2. J Neuropathol Exp Neurol 2005, 64, 754–762. [CrossRef]
- Zhang, J.; Sarkar, S.; Cua, R.; Zhou, Y.; Hader, W.; Yong, V.W. A Dialog between Glioma and Microglia That Promotes Tumor Invasiveness through the CCL2/CCR2/Interleukin-6 Axis. Carcinogenesis 2012, 33, 312–319. [CrossRef]
- Hu, F.; A Dzaye, O.D.; Hahn, A.; Yu, Y.; Scavetta, R.J.; Dittmar, G.; Kaczmarek, A.K.; Dunning, K.R.; Ricciardelli, C.; Rinnenthal, J.L.; et al. Glioma-Derived Versican Promotes Tumor Expansion via Glioma-Associated Microglial/Macrophages Toll-like Receptor 2 Signaling. Neuro-Oncology 2015, 17, 200–210. [CrossRef]
- Wick, W.; Platten, M.; Weller, M. [No Title Found]. Journal of Neuro-Oncology 2001, 53, 177–185. [CrossRef]
- Wen, J.; Wang, S.; Guo, R.; Liu, D. CSF1R Inhibitors Are Emerging Immunotherapeutic Drugs for Cancer Treatment. European Journal of Medicinal Chemistry 2023, 245, 114884. [CrossRef]
- Liu, H.; Zhao, Q.; Tan, L.; Wu, X.; Huang, R.; Zuo, Y.; Chen, L.; Yang, J.; Zhang, Z.-X.; Ruan, W.; et al. Neutralizing IL-8 Potentiates Immune Checkpoint Blockade Efficacy for Glioma. Cancer Cell 2023, 41, 693-710.e8. [CrossRef]
- Ye, Z.; Ai, X.; Yang, K.; Yang, Z.; Fei, F.; Liao, X.; Qiu, Z.; Gimple, R.C.; Yuan, H.; Huang, H.; et al. Targeting Microglial Metabolic Rewiring Synergizes with Immune-Checkpoint Blockade Therapy for Glioblastoma. Cancer Discovery 2023, 13, 974–1001. [CrossRef]
- Wang, T.; Zhou, Y.; Fan, Y.; Duan, H.; Guo, X.; Chang, J.; Jiang, Y.; Li, C.; Fu, Z.; Gao, Y.; et al. PERK-Mediated Cholesterol Excretion from IDH Mutant Glioma Determines Anti-Tumoral Polarization of Microglia. Advanced Science 2023, 10, 2205949. [CrossRef]
- Chandran, M.; Candolfi, M.; Shah, D.; Mineharu, Y.; Yadav, V.N.; Koschmann, C.; Asad, A.S.; Lowenstein, P.R.; Castro, M.G. Single vs. Combination Immunotherapeutic Strategies for Glioma. Expert Opinion on Biological Therapy 2017, 17, 543–554. [CrossRef]
- Ding, A.S.; Routkevitch, D.; Jackson, C.; Lim, M. Targeting Myeloid Cells in Combination Treatments for Glioma and Other Tumors. Front. Immunol. 2019, 10, 1715. [CrossRef]
- Balandeh, E.; Mohammadshafie, K.; Mahmoudi, Y.; Hossein Pourhanifeh, M.; Rajabi, A.; Bahabadi, Z.R.; Mohammadi, A.H.; Rahimian, N.; Hamblin, M.R.; Mirzaei, H. Roles of Non-Coding RNAs and Angiogenesis in Glioblastoma. Front. Cell Dev. Biol. 2021, 9, 716462. [CrossRef]
- Mahinfar, P.; Baradaran, B.; Davoudian, S.; Vahidian, F.; Cho, W.C.-S.; Mansoori, B. Long Non-Coding RNAs in Multidrug Resistance of Glioblastoma. Genes 2021, 12, 455. [CrossRef]
- Mousavi, S.M.; Derakhshan, M.; Baharloii, F.; Dashti, F.; Mirazimi, S.M.A.; Mahjoubin-Tehran, M.; Hosseindoost, S.; Goleij, P.; Rahimian, N.; Hamblin, M.R.; et al. Non-Coding RNAs and Glioblastoma: Insight into Their Roles in Metastasis. Molecular Therapy - Oncolytics 2022, 24, 262–287. [CrossRef]
- Subaiea, G.M.; Syed, R.U.; Afsar, S.; Alhaidan, T.M.S.; Alzammay, S.A.; Alrashidi, A.A.; Alrowaili, S.F.; Alshelaly, D.A.; Alenezi, A.M.S.R.A. Non-Coding RNAs (ncRNAs) and Multidrug Resistance in Glioblastoma: Therapeutic Challenges and Opportunities. Pathology - Research and Practice 2024, 253, 155022. [CrossRef]
- Brower, J.V.; Clark, P.A.; Lyon, W.; Kuo, J.S. MicroRNAs in Cancer: Glioblastoma and Glioblastoma Cancer Stem Cells. Neurochemistry International 2014, 77, 68–77. [CrossRef]
- Sati, I.S.E.E.; Parhar, I. MicroRNAs Regulate Cell Cycle and Cell Death Pathways in Glioblastoma. IJMS 2021, 22, 13550. [CrossRef]
- D’Asti, E.; Chennakrishnaiah, S.; Lee, T.H.; Rak, J. Extracellular Vesicles in Brain Tumor Progression. Cell Mol Neurobiol 2016, 36, 383–407. [CrossRef]
- Turra, L.P.; Rodrigues, A.R.; Lizarte Neto, F.S.; Novais, P.C.; Nunes, M.J.; Tirapelli, V.C.; Peria, F.M.; Carneiro, V.M.; Cirino, M.L.D.A.; Carlotti Jr, C.G.; et al. Expression of microRNAs miR-21 and miR-326 Associated with HIF-1α Regulation in Neurospheres of Glioblastoma Submitted to Ionizing Radiation Treatment. Rep Pract Oncol Radiother. 2022, 27, 215–225. [CrossRef]
- Khan, M.B.; Ruggieri, R.; Jamil, E.; Tran, N.L.; Gonzalez, C.; Mugridge, N.; Gao, S.; MacDiarmid, J.; Brahmbhatt, H.; Sarkaria, J.N.; et al. Nanocell-Mediated Delivery of miR-34a Counteracts Temozolomide Resistance in Glioblastoma. Mol Med 2021, 27, 28. [CrossRef]
- Stepanović, A.; Nikitović, M.; Stanojković, T.P.; Grujičić, D.; Bukumirić, Z.; Srbljak, I.; Ilić, R.; Milošević, S.; Arsenijević, T.; Petrović, N. Association between microRNAs 10b/21/34a and Acute Toxicity in Glioblastoma Patients Treated with Radiotherapy and Temozolomide. Sci Rep 2022, 12, 7505. [CrossRef]
- Jesionek-Kupnicka, D.; Braun, M.; Trąbska-Kluch, B.; Czech, J.; Szybka, M.; Szymańska, B.; Kulczycka-Wojdala, D.; Bieńkowski, M.; Kordek, R.; Zawlik, I. MiR-21, miR-34a, miR-125b, miR-181d and miR-648 Levels Inversely Correlate with MGMT and TP53 Expression in Primary Glioblastoma Patients. aoms 2019, 15, 504–512. [CrossRef]
- Yin, J.; Zeng, A.; Zhang, Z.; Shi, Z.; Yan, W.; You, Y. Exosomal Transfer of miR-1238 Contributes to Temozolomide-Resistance in Glioblastoma. EBioMedicine 2019, 42, 238–251. [CrossRef]
- Goenka, A.; Tiek, D.M.; Song, X.; Iglesia, R.P.; Lu, M.; Hu, B.; Cheng, S.-Y. The Role of Non-Coding RNAs in Glioma. Biomedicines 2022, 10, 2031. [CrossRef]
- Rezaei, O.; Tamizkar, K.H.; Sharifi, G.; Taheri, M.; Ghafouri-Fard, S. Emerging Role of Long Non-Coding RNAs in the Pathobiology of Glioblastoma. Front. Oncol. 2021, 10, 625884. [CrossRef]
- Vecera, M.; Sana, J.; Lipina, R.; Smrcka, M.; Slaby, O. Long Non-Coding RNAs in Gliomas: From Molecular Pathology to Diagnostic Biomarkers and Therapeutic Targets. IJMS 2018, 19, 2754. [CrossRef]
- Yang, J.; Sun, G.; Hu, Y.; Yang, J.; Shi, Y.; Liu, H.; Li, C.; Wang, Y.; Lv, Z.; Niu, J.; et al. Extracellular Vesicle lncRNA Metastasis-Associated Lung Adenocarcinoma Transcript 1 Released From Glioma Stem Cells Modulates the Inflammatory Response of Microglia After Lipopolysaccharide Stimulation Through Regulating miR-129-5p/High Mobility Group Box-1 Protein Axis. Front. Immunol. 2020, 10, 3161. [CrossRef]
- Chen, J.; Wang, H.; Wang, J.; Niu, W.; Deng, C.; Zhou, M. LncRNA NEAT1 Enhances Glioma Progression via Regulating the miR-128-3p/ITGA5 Axis. Mol Neurobiol 2021, 58, 5163–5177. [CrossRef]
- Yu, H.; Xu, A.; Wu, B.; Wang, M.; Chen, Z. Long Noncoding RNA NEAT1 Promotes Progression of Glioma as a ceRNA by Sponging miR-185-5p to Stimulate DNMT1/mTOR Signaling. Journal Cellular Physiology 2021, 236, 121–130. [CrossRef]
- Liu, S.J.; Dang, H.X.; Lim, D.A.; Feng, F.Y.; Maher, C.A. Long Noncoding RNAs in Cancer Metastasis. Nat Rev Cancer 2021, 21, 446–460. [CrossRef]
- Molavand, M.; Ebrahimnezhade, N.; Kiani, A.; Yousefi, B.; Nazari, A.; Majidinia, M. Regulation of Autophagy by Non-Coding RNAs in Human Glioblastoma. Med Oncol 2024, 41, 260. [CrossRef]
- Sandhanam, K.; Tamilanban, T. Unraveling the Noncoding RNA Landscape in Glioblastoma: From Pathogenesis to Precision Therapeutics. Naunyn-Schmiedeberg’s Arch Pharmacol 2024. [CrossRef]
- Ghadami, E.; Gorji, A.; Pour-Rashidi, A.; Noorbakhsh, F.; Kabuli, M.; Razipour, M.; Choobineh, H.; Maghsudlu, M.; Damavandi, E.; Ghadami, M. CircZNF609 and circNFIX as Possible Regulators of Glioblastoma Pathogenesis via miR-145-5p/EGFR Axis. Sci Rep 2024, 14, 13551. [CrossRef]
- Yang, Y.; Gao, X.; Zhang, M.; Yan, S.; Sun, C.; Xiao, F.; Huang, N.; Yang, X.; Zhao, K.; Zhou, H.; et al. Novel Role of FBXW7 Circular RNA in Repressing Glioma Tumorigenesis. JNCI: Journal of the National Cancer Institute 2018, 110, 304–315. [CrossRef]
- Stella, M.; Falzone, L.; Caponnetto, A.; Gattuso, G.; Barbagallo, C.; Battaglia, R.; Mirabella, F.; Broggi, G.; Altieri, R.; Certo, F.; et al. Serum Extracellular Vesicle-Derived circHIPK3 and circSMARCA5 Are Two Novel Diagnostic Biomarkers for Glioblastoma Multiforme. Pharmaceuticals 2021, 14, 618. [CrossRef]
- Fattahi, M.; Alamdari-palangi, V.; Rahimi Jaberi, K.; Ehtiati, S.; Ojaghi, S.; Rahimi-Jaberi, A.; Samavarchi Tehrani, S.; Dang, P.; Movahedpour, A.; Hossein Khatami, S. Exosomal Long Non-Coding RNAs in Glioblastoma. Clinica Chimica Acta 2024, 553, 117705. [CrossRef]
- Bouzari, B.; Mohammadi, S.; Bokov, D.O.; Krasnyuk, I.I.; Hosseini-Fard, S.R.; Hajibaba, M.; Mirzaei, R.; Karampoor, S. Angioregulatory Role of miRNAs and Exosomal miRNAs in Glioblastoma Pathogenesis. Biomedicine & Pharmacotherapy 2022, 148, 112760. [CrossRef]
- Crivii, C.-B.; Boșca, A.B.; Melincovici, C.S.; Constantin, A.-M.; Mărginean, M.; Dronca, E.; Suflețel, R.; Gonciar, D.; Bungărdean, M.; Șovrea, A. Glioblastoma Microenvironment and Cellular Interactions. Cancers 2022, 14, 1092. [CrossRef]
- Lai, Y.; Lu, X.; Liao, Y.; Ouyang, P.; Wang, H.; Zhang, X.; Huang, G.; Qi, S.; Li, Y. Crosstalk between Glioblastoma and Tumor Microenvironment Drives Proneural–Mesenchymal Transition through Ligand-Receptor Interactions. Genes & Diseases 2024, 11, 874–889. [CrossRef]
- Hu, Y.; Tao, W. Current Perspectives on Microglia-Neuron Communication in the Central Nervous System: Direct and Indirect Modes of Interaction. Journal of Advanced Research 2024. [CrossRef]
- Ramón Y Cajal, S.; Segura, M.F.; Hümmer, S. Interplay Between ncRNAs and Cellular Communication: A Proposal for Understanding Cell-Specific Signaling Pathways. Front Genet 2019, 10, 281. [CrossRef]
- Sánchez-Marín, D.; Trujano-Camacho, S.; Pérez-Plasencia, C.; De León, D.C.; Campos-Parra, A.D. LncRNAs Driving Feedback Loops to Boost Drug Resistance: Sinuous Pathways in Cancer. Cancer Letters 2022, 543, 215763. [CrossRef]
- Slack, F.J.; Chinnaiyan, A.M. The Role of Non-Coding RNAs in Oncology. Cell 2019, 179, 1033–1055. [CrossRef]
- Yabo, Y.A.; Niclou, S.P.; Golebiewska, A. Cancer Cell Heterogeneity and Plasticity: A Paradigm Shift in Glioblastoma. Neuro-Oncology 2022, 24, 669–682. [CrossRef]
- Mosher, K.I.; Andres, R.H.; Fukuhara, T.; Bieri, G.; Hasegawa-Moriyama, M.; He, Y.; Guzman, R.; Wyss-Coray, T. Neural Progenitor Cells Regulate Microglia Functions and Activity. Nat Neurosci 2012, 15, 1485–1487. [CrossRef]
- Noch, E.K.; Ramakrishna, R.; Magge, R. Challenges in the Treatment of Glioblastoma: Multisystem Mechanisms of Therapeutic Resistance. World Neurosurgery 2018, 116, 505–517. [CrossRef]
- Llaguno, S.R.A.; Parada, L.F. Cell of Origin of Glioma: Biological and Clinical Implications. British Journal of Cancer 2016, 115, 1445. [CrossRef]
- Singh, S.K.; Hawkins, C.; Clarke, I.D.; Squire, J.A.; Bayani, J.; Hide, T.; Henkelman, R.M.; Cusimano, M.D.; Dirks, P.B. Identification of Human Brain Tumour Initiating Cells. Nature 2004, 432, 396–401. [CrossRef]
- Jones, D.T.W.; Mulholland, S.A.; Pearson, D.M.; Malley, D.S.; Openshaw, S.W.S.; Lambert, S.R.; Liu, L.; Bäcklund, L.M.; Ichimura, K.; Collins, V.P. Adult Grade II Diffuse Astrocytomas Are Genetically Distinct from and More Aggressive than Their Paediatric Counterparts. Acta Neuropathol 2011, 121, 753–761. [CrossRef]
- Liu, C.; Sage, J.C.; Miller, M.R.; Verhaak, R.G.W.; Hippenmeyer, S.; Vogel, H.; Foreman, O.; Bronson, R.T.; Nishiyama, A.; Luo, L.; et al. Mosaic Analysis with Double Markers Reveals Tumor Cell of Origin in Glioma. Cell 2011, 146, 209–221. [CrossRef]
- Wang, Q.; Hu, B.; Hu, X.; Kim, H.; Squatrito, M.; Scarpace, L.; deCarvalho, A.C.; Lyu, S.; Li, P.; Li, Y.; et al. Tumor Evolution of Glioma-Intrinsic Gene Expression Subtypes Associates with Immunological Changes in the Microenvironment. Cancer Cell 2017, 32, 42-56.e6. [CrossRef]
- Ricci-Vitiani, L.; Pallini, R.; Biffoni, M.; Todaro, M.; Invernici, G.; Cenci, T.; Maira, G.; Parati, E.A.; Stassi, G.; Larocca, L.M.; et al. Tumour Vascularization via Endothelial Differentiation of Glioblastoma Stem-like Cells. Nature 2010, 468, 824–828. [CrossRef]
- Poon, C.C.; Sarkar, S.; Yong, V.W.; Kelly, J.J.P. Glioblastoma-Associated Microglia and Macrophages: Targets for Therapies to Improve Prognosis. Brain 2017, 140, 1548–1560. [CrossRef]
- Hambardzumyan, D.; Bergers, G. Glioblastoma: Defining Tumor Niches. Trends in cancer 2015, 1, 252. [CrossRef]
- Matsuzaki, H.; Pan, C.; Komohara, Y.; Yamada, R.; Yano, H.; Fujiwara, Y.; Kai, K.; Mukasa, A. The Roles of Glioma-Associated Macrophages/Microglia and Potential Targets for Anti-Glioma Therapy. Immunological Medicine 0, 1–9. [CrossRef]
- Saha, D.; Martuza, R.L.; Rabkin, S.D. Macrophage Polarization Contributes to Glioblastoma Eradication by Combination Immunovirotherapy and Immune Checkpoint Blockade. Cancer Cell 2017, 32, 253-267.e5. [CrossRef]
- Matias, D.; Balça-Silva, J.; Graça, G.C. da; Wanjiru, C.M.; Macharia, L.W.; Nascimento, C.P.; Roque, N.R.; Coelho-Aguiar, J.M.; Pereira, C.M.; Santos, M.F.D.; et al. Microglia/Astrocytes–Glioblastoma Crosstalk: Crucial Molecular Mechanisms and Microenvironmental Factors. Frontiers in Cellular Neuroscience 2018, 12, 235. [CrossRef]
- Roesch, S.; Rapp, C.; Dettling, S.; Herold-Mende, C. When Immune Cells Turn Bad—Tumor-Associated Microglia/Macrophages in Glioma. International Journal of Molecular Sciences 2018, 19, 436. [CrossRef]
- Masoudi, M.S.; Mehrabian, E.; Mirzaei, H. MiR-21: A Key Player in Glioblastoma Pathogenesis. J Cell Biochem 2018, 119, 1285–1290. [CrossRef]
- Ivo D’Urso, P.; Fernando D’Urso, O.; Damiano Gianfreda, C.; Mezzolla, V.; Storelli, C.; Marsigliante, S. miR-15b and miR-21 as Circulating Biomarkers for Diagnosis of Glioma. Curr Genomics 2015, 16, 304–311. [CrossRef]
- Teplyuk, N.M.; Mollenhauer, B.; Gabriely, G.; Giese, A.; Kim, E.; Smolsky, M.; Kim, R.Y.; Saria, M.G.; Pastorino, S.; Kesari, S.; et al. MicroRNAs in Cerebrospinal Fluid Identify Glioblastoma and Metastatic Brain Cancers and Reflect Disease Activity. Neuro-Oncology 2012, 14, 689–700. [CrossRef]
- Ahmadov, U.; Picard, D.; Bartl, J.; Silginer, M.; Trajkovic-Arsic, M.; Qin, N.; Blümel, L.; Wolter, M.; Lim, J.K.M.; Pauck, D.; et al. The Long Non-Coding RNA HOTAIRM1 Promotes Tumor Aggressiveness and Radiotherapy Resistance in Glioblastoma. Cell Death Dis 2021, 12, 885. [CrossRef]
- Ke, J.; Yao, Y.; Zheng, J.; Wang, P.; Liu, Y.; Ma, J.; Li, Z.; Liu, X.; Li, Z.; Wang, Z.; et al. Knockdown of Long Non-Coding RNA HOTAIR Inhibits Malignant Biological Behaviors of Human Glioma Cells via Modulation of miR-326. Oncotarget 2015, 6, 21934. [CrossRef]
- Cheng, S.; Zhang, Y.; Chen, S.; Zhou, Y. LncRNA HOTAIR Participates in Microglia Activation and Inflammatory Factor Release by Regulating the Ubiquitination of MYD88 in Traumatic Brain Injury. J Mol Neurosci 2021, 71, 169–177. [CrossRef]
- Baspinar, Y.; Elmaci, I.; Ozpinar, A.; Altinoz, M.A. Long Non-Coding RNA MALAT1 as a Key Target in Pathogenesis of Glioblastoma. Janus Faces or Achilles’ Heal? Gene 2020, 739, 144518. [CrossRef]
- Chen, W.; Xu, X.-K.; Li, J.-L.; Kong, K.-K.; Li, H.; Chen, C.; He, J.; Wang, F.; Li, P.; Ge, X.-S.; et al. MALAT1 Is a Prognostic Factor in Glioblastoma Multiforme and Induces Chemoresistance to Temozolomide through Suppressing miR-203 and Promoting Thymidylate Synthase Expression. Oncotarget 2017, 8, 22783–22799. [CrossRef]
- Fu, S.; Wang, Y.; Li, H.; Chen, L.; Liu, Q. Regulatory Networks of LncRNA MALAT-1 in Cancer. CMAR 2020, Volume 12, 10181–10198. [CrossRef]
- Fu, Y.; Sun, H. Biogenesis, Cellular Effects, and Biomarker Value of circHIPK3. Cancer Cell Int 2021, 21, 256. [CrossRef]
- Gandhi, S.; Bhushan, A.; Shukla, U.; Pundir, A.; Singh, S.; Srivastava, T. Downregulation of lncRNA SNHG1 in Hypoxia and Stem Cells Is Associated with Poor Disease Prognosis in Gliomas. Cell Cycle 2023, 22, 1135–1153. [CrossRef]
- Chen, M.; Yang, Y.; Zhang, W.; Li, X.; Wu, J.; Zou, X.; Zeng, X. Long Noncoding RNA SNHG5 Knockdown Alleviates Neuropathic Pain by Targeting the miR-154-5p/CXCL13 Axis. Neurochem Res 2020, 45, 1566–1575. [CrossRef]
- Tripathy, D.K.; Panda, L.P.; Biswal, S.; Barhwal, K. Insights into the Glioblastoma Tumor Microenvironment: Current and Emerging Therapeutic Approaches. Frontiers in Pharmacology 2024, 15, 1355242. [CrossRef]
- Chen, G.; Chen, Z.; Zhao, H. MicroRNA-155-3p Promotes Glioma Progression and Temozolomide Resistance by Targeting Six1. Journal of Cellular and Molecular Medicine 2020, 24, 5363. [CrossRef]
- Yalamarty, S.S.K.; Filipczak, N.; Li, X.; Subhan, M.A.; Parveen, F.; Ataide, J.A.; Rajmalani, B.A.; Torchilin, V.P. Mechanisms of Resistance and Current Treatment Options for Glioblastoma Multiforme (GBM). Cancers (Basel) 2023, 15, 2116. [CrossRef]
- Lowe, S.; Bhat, K.P.; Olar, A. Current Clinical Management of Patients with Glioblastoma. Cancer Rep (Hoboken) 2019, 2, e1216. [CrossRef]
- Aparicio-Blanco, J.; Sanz-Arriazu, L.; Lorenzoni, R.; Blanco-Prieto, M.J. Glioblastoma Chemotherapeutic Agents Used in the Clinical Setting and in Clinical Trials: Nanomedicine Approaches to Improve Their Efficacy. International Journal of Pharmaceutics 2020, 581, 119283. [CrossRef]
- Blumenthal, D.T.; Gorlia, T.; Gilbert, M.R.; Kim, M.M.; Burt Nabors, L.; Mason, W.P.; Hegi, M.E.; Zhang, P.; Golfinopoulos, V.; Perry, J.R.; et al. Is More Better? The Impact of Extended Adjuvant Temozolomide in Newly Diagnosed Glioblastoma: A Secondary Analysis of EORTC and NRG Oncology/RTOG. Neuro-Oncology 2017, 19, 1119–1126. [CrossRef]
- Tang, X.; Zuo, C.; Fang, P.; Liu, G.; Qiu, Y.; Huang, Y.; Tang, R. Targeting Glioblastoma Stem Cells: A Review on Biomarkers, Signal Pathways and Targeted Therapy. Front Oncol 2021, 11, 701291. [CrossRef]
- Wang, Z.; Zhang, H.; Xu, S.; Liu, Z.; Cheng, Q. The Adaptive Transition of Glioblastoma Stem Cells and Its Implications on Treatments. Sig Transduct Target Ther 2021, 6, 1–13. [CrossRef]
- Campos, B.; Wan, F.; Farhadi, M.; Ernst, A.; Zeppernick, F.; Tagscherer, K.E.; Ahmadi, R.; Lohr, J.; Dictus, C.; Gdynia, G.; et al. Differentiation Therapy Exerts Antitumor Effects on Stem-like Glioma Cells. Clin Cancer Res 2010, 16, 2715–2728. [CrossRef]
- Arima, Y.; Nobusue, H.; Saya, H. Targeting of Cancer Stem Cells by Differentiation Therapy. Cancer Science 2020, 111, 2689. [CrossRef]
- Takebe, N.; Miele, L.; Harris, P.J.; Jeong, W.; Bando, H.; Kahn, M.; Yang, S.X.; Ivy, S.P. Targeting Notch, Hedgehog, and Wnt Pathways in Cancer Stem Cells: Clinical Update. Nat Rev Clin Oncol 2015, 12, 445–464. [CrossRef]
- Xia, R.; Xu, M.; Yang, J.; Ma, X. The Role of Hedgehog and Notch Signaling Pathway in Cancer. Molecular Biomedicine 2022, 3, 44. [CrossRef]
- Andersen, R.S.; Anand, A.; Harwood, D.S.L.; Kristensen, B.W. Tumor-Associated Microglia and Macrophages in the Glioblastoma Microenvironment and Their Implications for Therapy. Cancers 2021, 13, 4255. [CrossRef]
- Nemeth, K.; Bayraktar, R.; Ferracin, M.; Calin, G.A. Non-Coding RNAs in Disease: From Mechanisms to Therapeutics. Nat Rev Genet 2024, 25, 211–232. [CrossRef]
- Zhu, P.; Liu, B.; Fan, Z. Noncoding RNAs in Tumorigenesis and Tumor Therapy. Fundamental Research 2023, 3, 692–706. [CrossRef]
- Kim, T.; Croce, C.M. MicroRNA: Trends in Clinical Trials of Cancer Diagnosis and Therapy Strategies. Exp Mol Med 2023, 55, 1314–1321. [CrossRef]
- Toden, S.; Zumwalt, T.J.; Goel, A. Non-Coding RNAs and Potential Therapeutic Targeting in Cancer. Biochimica et biophysica acta. Reviews on cancer 2020, 1875, 188491. [CrossRef]
- Saenz-Pipaon, G.; Dichek, D.A. Targeting and Delivery of microRNA-Targeting Antisense Oligonucleotides in Cardiovascular Diseases. Atherosclerosis 2023, 374, 44–54. [CrossRef]
- Valerius, A.R.; Webb, L.M.; Thomsen, A.; Lehrer, E.J.; Breen, W.G.; Campian, J.L.; Riviere-Cazaux, C.; Burns, T.C.; Sener, U. Review of Novel Surgical, Radiation, and Systemic Therapies and Clinical Trials in Glioblastoma. International Journal of Molecular Sciences 2024, 25, 10570. [CrossRef]
- Rong, L.; Li, N.; Zhang, Z. Emerging Therapies for Glioblastoma: Current State and Future Directions. J Exp Clin Cancer Res 2022, 41, 142. [CrossRef]
- Ito, M.; Miyata, Y.; Okada, M. Current Clinical Trials with Non-Coding RNA-Based Therapeutics in Malignant Diseases: A Systematic Review. Translational Oncology 2023, 31, 101634. [CrossRef]
- Shergalis, A.; Armand Bankhead, I.I.I.; Luesakul, U.; Muangsin, N.; Neamati, N. Current Challenges and Opportunities in Treating Glioblastoma. Pharmacological Reviews 2018, 70, 412. [CrossRef]
- Benmelouka, A.Y.; Munir, M.; Sayed, A.; Attia, M.S.; Ali, M.M.; Negida, A.; Alghamdi, B.S.; Kamal, M.A.; Barreto, G.E.; Ashraf, G.M.; et al. Neural Stem Cell-Based Therapies and Glioblastoma Management: Current Evidence and Clinical Challenges. Int J Mol Sci 2021, 22, 2258. [CrossRef]
- Achar, A.; Myers, R.; Ghosh, C. Drug Delivery Challenges in Brain Disorders across the Blood–Brain Barrier: Novel Methods and Future Considerations for Improved Therapy. Biomedicines 2021, 9, 1834. [CrossRef]
- Husain, A.; Pandey, N.; Singh, D.; Ahmad, F.; Sharma, R.; Siddiqui, M.H. Drug Discovery in Glioblastoma: Current Status and Future Perspectives. Biointerface Research in Applied Chemistry 2023, 13. [CrossRef]
- Cha, G.D.; Kang, T.; Baik, S.; Kim, D.; Choi, S.H.; Hyeon, T.; Kim, D.-H. Advances in Drug Delivery Technology for the Treatment of Glioblastoma Multiforme. Journal of Controlled Release 2020, 328, 350–367. [CrossRef]
- Qazi, M.A.; Vora, P.; Venugopal, C.; Sidhu, S.S.; Moffat, J.; Swanton, C.; Singh, S.K. Intratumoral Heterogeneity: Pathways to Treatment Resistance and Relapse in Human Glioblastoma. Annals of Oncology 2017, 28, 1448–1456. [CrossRef]
- Jin, X.; Kim, L.J.Y.; Wu, Q.; Wallace, L.C.; Prager, B.C.; Sanvoranart, T.; Gimple, R.C.; Wang, X.; Mack, S.C.; Miller, T.E.; et al. Targeting Glioma Stem Cells through Combined BMI1 and EZH2 Inhibition. Nat Med 2017, 23, 1352–1361. [CrossRef]
- Kumar, V.; Vashishta, M.; Kong, L.; Wu, X.; Lu, J.J.; Guha, C.; Dwarakanath, B.S. The Role of Notch, Hedgehog, and Wnt Signaling Pathways in the Resistance of Tumors to Anticancer Therapies. Frontiers in Cell and Developmental Biology 2021, 9, 650772. [CrossRef]
- Adewunmi, O.; Shen, Y.; Zhang, X.H.-F.; Rosen, J.M. Targeted Inhibition of lncRNA Malat1 Alters the Tumor Immune Microenvironment in Preclinical Syngeneic Mouse Models of Triple-Negative Breast Cancer. Cancer Immunology Research 2023, 11, 1462–1479. [CrossRef]
- Rončević, A.; Koruga, N.; Soldo Koruga, A.; Rončević, R.; Rotim, T.; Šimundić, T.; Kretić, D.; Perić, M.; Turk, T.; Štimac, D. Personalized Treatment of Glioblastoma: Current State and Future Perspective. Biomedicines 2023, 11, 1579. [CrossRef]
- Iyer, K.; Saini, S.; Bhadra, S.; Kulavi, S.; Bandyopadhyay, J. Precision Medicine Advancements in Glioblastoma: A Systematic Review. BioMedicine 2023, 13, 1. [CrossRef]
- Karimi-Sani, I.; Molavi, Z.; Naderi, S.; Mirmajidi, S.-H.; Zare, I.; Naeimzadeh, Y.; Mansouri, A.; Tajbakhsh, A.; Savardashtaki, A.; Sahebkar, A. Personalized mRNA Vaccines in Glioblastoma Therapy: From Rational Design to Clinical Trials. Journal of Nanobiotechnology 2024, 22, 601. [CrossRef]
- Hersh, A.M.; Bhimreddy, M.; Weber-Levine, C.; Jiang, K.; Alomari, S.; Theodore, N.; Manbachi, A.; Tyler, B.M. Applications of Focused Ultrasound for the Treatment of Glioblastoma: A New Frontier. Cancers 2022, 14, 4920. [CrossRef]
- Elumalai, K.; Srinivasan, S.; Shanmugam, A. Review of the Efficacy of Nanoparticle-Based Drug Delivery Systems for Cancer Treatment. Biomedical Technology 2024, 5, 109–122. [CrossRef]
- Sun, L.; Liu, H.; Ye, Y.; Lei, Y.; Islam, R.; Tan, S.; Tong, R.; Miao, Y.-B.; Cai, L. Smart Nanoparticles for Cancer Therapy. Sig Transduct Target Ther 2023, 8, 1–28. [CrossRef]
- Kalluri, R.; LeBleu, V.S. The Biology, Function, and Biomedical Applications of Exosomes. Science 2020, 367, eaau6977. [CrossRef]
- Jiang, X.-C.; Gao, J.-Q. Exosomes as Novel Bio-Carriers for Gene and Drug Delivery. International Journal of Pharmaceutics 2017, 521, 167–175. [CrossRef]
- Tenchov, R.; Sasso, J.M.; Wang, X.; Liaw, W.-S.; Chen, C.-A.; Zhou, Q.A. Exosomes─Nature’s Lipid Nanoparticles, a Rising Star in Drug Delivery and Diagnostics. ACS Nano 2022, 16, 17802–17846. [CrossRef]
- Iqbal, Z.; Rehman, K.; Mahmood, A.; Shabbir, M.; Liang, Y.; Duan, L.; Zeng, H. Exosome for mRNA Delivery: Strategies and Therapeutic Applications. Journal of Nanobiotechnology 2024, 22, 395. [CrossRef]
|
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. |
© 2024 by the authors. Licensee MDPI, Basel, Switzerland. This article is an open access article distributed under the terms and conditions of the Creative Commons Attribution (CC BY) license (http://creativecommons.org/licenses/by/4.0/).