Open Access

Lipid metabolic rewiring in glioma‑associated microglia/macrophages (Review)

  • Authors:
    • Yixuan Ma
    • Yimin Huang
    • Feng Hu
    • Kai Shu
  • View Affiliations

  • Published online on: September 16, 2024     https://doi.org/10.3892/ijmm.2024.5426
  • Article Number: 102
  • Copyright: © Ma et al. This is an open access article distributed under the terms of Creative Commons Attribution License [CC BY_NC 4.0].

Metrics: Total Views: 0 (Spandidos Publications: | PMC Statistics: )
Total PDF Downloads: 0 (Spandidos Publications: | PMC Statistics: )


Abstract

Gliomas are the most prevailing brain malignancy in both children and adults. Microglia, which are resident in the central nervous system (CNS), are distributed throughout the brain and serve an important role in the immunity of the CNS. Microglial cells exhibit varying phenotypic and metabolic properties during different stages of glioma development, making them a highly dynamic cell population. In particular, glioma‑associated microglia/macrophages (GAMs) can alter their metabolic characteristics and influence malignancies in response to the signals they receive. The significance of macrophage metabolic reprogramming in tumor growth is becoming increasingly acknowledged in recent years. However, to the best of our knowledge, there is currently a scarcity of data from investigations into the lipid metabolic profiles of microglia/macrophages in the glioma setting. Therefore, the present review aims to provide a thorough review of the role that lipid metabolism serves in tumor‑associated macrophages. In addition, it outlines potential targets for therapy based on lipid metabolism. The present review aims to serve as a reference source for future investigations into GAMs.

Introduction

Gliomas are the most common type of brain malignancy in both children and adults. According to the World Health Organization (WHO), glioma can be classified into the low-grade (grades 1 and 2) and high-grade (grades 3 and 4) categories, based on the degree of malignancy from lowest to highest (1-3). The prognosis for gliomas remains poor despite the existence of multiple treatment strategies, including surgery, radiation, chemotherapy and targeted therapy. Specifically, glioblastoma (GBM) multiforme (WHO grade 4) has a 5-year survival rate of only 5.5% (4), which may be due to chemoresistance, heterogeneity and infiltrative properties, making the tumor difficult to remove completely (5). By contrast, low-grade gliomas (WHO grades 1-2) have a relatively favorable prognosis, with an overall survival of ~7 years (6).

Glioma tissues can consist not only of cancer cells but can also contain various non-cancerous cell types, such as resident microglia from the brain and monocytes (macrophages) from the circulating bloodstream. In particular, macrophages and microglia are highly heterogeneous and plastic, such that they become cells of different phenotypes after in vitro stimulation (7). Toll-like receptor 4 (TLR4) ligands and IFN-γ stimulation typically result in the pro-inflammatory M1 phenotype, whereas IL-4, IL-10 and IL-13 stimulation typically produce an anti-inflammatory M2 phenotype (8). In addition, macrophages can be selectively activated further and then subdivided into the M2a [type II T-helper cell (Th2) response, type II inflammation, pathogen killing and allergic response], M2b (Th2 activation, immunomodulation) and M2c (immunomodulation, matrix deposition and tissue remodeling) states (8,9). These macrophage subpopulations differ in their receptor expression, effector function, as well as cytokine and chemokine expression profiles. However, this phenotype definition was proposed based on data from mainly in vitro research, meaning that they cannot be used to fully reflect the in vivo situation of the different pathological conditions.

Metabolic reprogramming refers to the process by which cells adjust their metabolic pathways and energy production methods to adapt to environmental changes under specific conditions. However, this process is not merely a simple metabolic change. Instead, it typically involves profound systemic adjustments aimed at meeting the specific physiological needs of the cells (10,11). Metabolic reprogramming in tumors will likely involve significant changes in the energy production and metabolic pathways being activated. Changes that have been previously reported include the Warburg effect, enhanced lipid synthesis and abnormal amino acid metabolism (12,13). However, the majority of such previous studies have mainly focused on tumor cells. Metabolic reprogramming in other cell types that reside in the tumor microenvironment, such as glioma-associated microglia/macrophages (GAMs), should also be considered. Tumor cell metabolic reprogramming can mediate macrophage phenotypic alterations through various mechanisms, such as epigenetic modifications, leading to altered macrophage metabolism and in turn tumor progression (14). Studies over the past decade have demonstrated that altered lipid metabolism in tumor-associated macrophages (TAMs) can serve an important role in tumor progression, though to the best of our knowledge, there have been few similar studies on GAMs. Therefore, the present review aims to systematically summarize the research progress on metabolic reprogramming in GAMs. Based on existing studies on TAMs (15), hypotheses regarding the role of GAMs in glioma are proposed, emphasizing their potential metabolic similarities. In addition, the complex regulatory mechanisms potentially driving these metabolic changes and their implications for tumor progression and immune evasion are summarized. The present review also explores potential therapeutic targets within lipid metabolism, aiming to facilitate future strategies for inhibiting glioma tumor growth by modulating GAM metabolism. The novelty of the present review lies in its comprehensive focus on the underexplored area of GAM lipid metabolism and integration of recent findings to propose novel research directions and clinical applications.

Metabolic microenvironment of glioma

Metabolic characteristics of glioma

Similar to other rapidly proliferating cells, glioma cells typically metabolize glucose into lactate even in the presence of oxygen (the 'Warburg' effect). This allows tumor cells to use glucose-derived carbon to synthesize essential cellular components whilst simultaneously producing sufficient ATP to support its substantial metabolic demands (16-18). In addition, glioma cells can increase their own intracellular stores of fats, amino acids and nucleotides through various pathways. These include extracellular uptake, de novo synthesis and the delivery of carbon or nitrogen via multiple routes (17,19).

Effect of cells in the tumor microenvironment on metabolism

The brain is a highly metabolically active organ that relies on glucose as its major energy substrate. However, lactate, ketone bodies, fatty acids (FAs) and amino acids can also serve as its energy source (20-22). In addition, astrocytes, neurons and microglia can all regulate the nutrient uptake processes of each other (18). Specifically, neurons can absorb lactate, cholesterol and FAs produced by astrocytes, whilst astrocytes can take up glutamate produced by neurons (20). Gliomas develop in a complex and frequently hypoxic environment, which significantly influences the metabolic decisions of glioma cells, driving tumor growth, reproduction and invasion (23-26).

GAMs

Sources

Microglia are macrophages that reside in the central nervous system (CNS) and are distributed throughout the brain. They serve as the key immune effector cell type in the CNS. GAMs typically originate from two cell types, namely brain-resident microglia (BRM) and bone marrow-derived monocytes (BMDM) (27). The debate over the origin of microglia remains to the present day after it was first proposed by del Rio-Hortega (28). It has been suggested that increased microglial density after CNS injury involves both BRM proliferation and active recruitment of BMDM progenitors from the bloodstream (29-32). By contrast, it has also been suggested that the increase in microglial density originates primarily from the BRM (33). The reason for this controversy may lie in the experimental methodology used. To avoid the influence of the blood-brain barrier, the method used to distinguish microglia from monocytes is to first destroy the hematopoietic system of the recipient's bone marrow with radiation and then transplant the labeled hematopoietic stem cells into the recipient, before observing the infiltration of the labeled monocytes into the tumor tissue of the brain. However, irradiation can damage the blood-brain barrier in mice whilst disrupting the immune system and non-specific infiltration of immune cells into the brain, compromising the accuracy of the experiment (34). This debate continued until it was resolved when a chimeric animal was generated by a form of heterologous symbiosis that required neither irradiation nor transplantation. Both axotomy and neurodegeneration models failed to recruit microglia from the circulation (33). In addition, similar results were observed in a mouse model of experimental allergic encephalomyelitis (35).

In high-grade gliomas, BMDM accounts for >85% of GAMs, whereas BRM is predominantly distributed in peritumoral tissues (36,37). In the past, the expression levels of CD45 were typically used to differentiate between BRM (CD45 high expression)-derived and BMDM (CD45 low expression)-derived GAMs (38). However, different views have emerged in recent years. A previous study has shown that although the expression level of CD48 can distinguish BRM-GAMs from BMDM-GAMs to a certain extent, the cell type-specific CD45 expression profiles of humans and mice are different. In addition, the differentiation effect of CD45 is not precise, necessitating the use of more sensitive and specific methods, such as RNA sequencing and flow cytometry, to accurately distinguish between BRM-derived and BMDM-derived GAMs (39).

A large-scale RNA sequencing analysis has previously revealed the existence of BRM-derived GAMs and BMDM-derived GAMs with distinct gene expression patterns. In particular, subpopulations of GAMs from different origins may perform different functions (36). Another previous study found that transmembrane protein 119 (TMEM119) was stably expressed only in BRM-derived GAMs. Subsequently, RNA-sequencing was performed in this previous study based on the expression profile of TMEM119 and differences in the transcript fragments of BRM- and BMDM-derived GAMs were found. It was also observed that the gene expression pattern of BRM may differ at different stages of development, such that, as microglia mature, the expression of their specifically expressed genes (such as TMEM119, purinergic receptor P2Y12 and olfactomedin-like 3) increases, but their proliferative capacity decreases (40). Using genealogical tracer techniques and a mouse model of glioma, Bowman et al (39) previously found that the transcriptional profiles and epigenetic landscapes between the two major subgroups of GAMs differed markedly, whereby CD49d was proposed as a distinguishing marker. Furthermore, Müller et al (41) previously performed single-cell sequencing on clinical glioma specimens and found that the levels of immunosuppressive cytokines, M2 activation markers (IL-10 and TGF-βII), phagocytosis and tricarboxylic acid (TCA) cycle activity were all upregulated in BMDM compared with those in BRM.

Phenotypic changes

Macrophages and microglia belong to the same monocyte type. Therefore, they can have a high degree of diversity and plasticity, allowing them to exhibit various phenotypes when exposed to different in vitro stimuli (7). Stimulation with Toll-like receptor (TLR)4 ligands and IFN-γ typically produces the pro-inflammatory M1 phenotype, whereas stimulation with IL-4, IL-10 and IL-13 produces an anti-inflammatory M2 phenotype (8). In a previous study, RNA microarray was applied to compare the expression profiles of microglia, macrophages and control microglia obtained by CD11b antibody-mediated magnetic beads sorting. The results showed that ~1,000 transcripts were differentially expressed in GAMs twice or more compared with those in control microglia. This expression pattern overlapped only partially with the reported gene profiles of the M1, M2a, M2b and M2c phenotypes (42). It has also been shown that GAMs can exhibit a different expression profile from the M1 and M2 phenotypes whilst highly expressing glycoprotein non-metastatic melanoma protein B and secreted phosphoprotein 1.

According to previous histological investigations that focused on single cells, the characteristics of GAMs are not limited to only M1 and M2 phenotypes. Instead, a wide range of variations have been noted. At present, no one superior typing method has been found compared with M1/M2. Since the majority of the relevant studies have continued to concentrate on M1/M2, discussion of data related to this topic will also center around M1/M2. GAMs express a number of markers that characterize the M1 or M2 phenotype (7). It has been previously shown that glioma-derived macrophage colony-stimulating factor (CSF) can induce microglia and macrophages to shift into an M2 phenotype, thereby promoting tumor growth (43). Similarly, mTOR and CSF-1 was found to inhibit microglia transformation into the M1 phenotype (43,44). Dopamine, microRNA (miR)-142-3p, prolyl 4-hydroxylase subunit α 1 downregulation and anti-programmed cell death protein 1 also showed similar anti-tumor effects (45-48). Based on these previous aforementioned studies, it has been proposed that targeted therapy aiming at converting the M2 phenotype into the M1 phenotype is a potential therapeutic strategy to inhibit glioma growth. However, other previous studies have also shown that M1-specific markers or associated pathways (IL1-β) are positively associated with glioma growth (49). It has also been indicated that sterile α and HEAT/armadillo motif can inhibit glioma progression by inducing the M2 polarization of GAMs (50).

Effect of GAMs on glioma

In the glioma microenvironment, microglia act through two main mechanisms. Microglia first become active upon glioma stimulation, producing cytokines, growth factors and MMPs to promote tumor growth and invasion (51). Subsequently, tumor cells secrete chemotactic agents and chemokines to recruit another population of microglia for activation, creating a continuous cycle (7,52,53). It has been previously shown that several common chemokines and receptors are upregulated in gliomas, including monocyte chemoattractant protein-1 (MCP-1), granulocyte-macrophage (GM)-CSF and fractalkine (54). MCP-1 has been considered to serve a key role in recruiting microglia to gliomas, where IL-33 may also be involved (Fig. 1) (51,54,55). In addition, microglia can have an important effect on angiogenesis, an effect associated with VEGF, which stimulates angiogenesis and promotes tumor growth (8). It has also been shown that inflammation is a key factor in brain tumor progression. Inflammation leads to the production of chemokines, such as C-X-C-motif chemokine ligand (CXCL)12, CXCL18 and reactive oxygen species (ROS), which promote tumor development by damaging DNA, proteins and lipids (56). Another previous study showed that the programmed cell death 10 protein serves an important role in the CXCL2/C-X-C chemokine receptor type 2 signaling pathway (57).

Lipid metabolic reprogramming and tumor-associated microglia/macrophages

Lipid metabolic reprogramming is a major feature of tumorigenesis and progression, serving a crucial role in GAMs. Initially, during tumor development, GAMs exhibit an M1 phenotype. However, as the tumor progresses, GAMs predominantly show the M2 phenotype. This metabolic reprogramming has been indicated to be regulated by hypoxia-inducible factor 1α and its downstream components (58-64). However, due to the limited availability of pertinent studies on gliomas, this section will discuss lipid metabolic reprogramming in TAMs of other tumor types. From these insights, the potential impact of GAMs of gliomas will be speculated.

Reprogramming of lipid metabolism in TAMs
FA metabolism in TAMs

Altered FA metabolism in tumor cells increases lipid accumulation in the TAM, which in turn promotes TAM activation and polarization (65). It has been previously found that M2 polarization is associated with FA oxidation (FAO). The scavenger receptor CD36 is highly expressed in TAMs, through which they take up and accumulate lipids (66). Results from a previous in vivo experiment corroborated this finding, where TAMs from tumor-bearing mice were found to have a higher lipid content compared with macrophages from tumor-free mice (67). High levels of FAO can promote mitochondrial oxidative phosphorylation and downstream signaling, accompanied by activation of the TCA cycle, which in turn promotes the M2 polarization of TAMs (15,60,66,68-70). Other previous studies have also shown that the metabolic efficiency of FAO serves an important role in regulating the polarization of TAMs, whereby β-oxidation is closely associated with the phenotype of TAMs (60,70). The peroxisome proliferator-activated receptor (PPAR) system regulates FAO and significantly influences the metabolic reprogramming of TAMs and their polarization towards the M2 phenotype (71,72). Specifically, the PPAR system enhances FAO metabolic efficiency mediated by STAT6 and PPARγ coactivator-1β (73,74).

However, other potentially noteworthy pathways have not been intensively studied. In particular, IFN-γ, GM-CSF and lipopolysaccharide (LPS) are factors that can induce M1 polarization (75). Previous studies have shown that there may be associations between the aforementioned factors and the FAO (76,77). In addition, proposals of regulating FAO by targeting IFN-γ, GM-CSF and LPS to in turn achieve a desired anti-tumor effect have been made (78-81).

PPAR is an indispensable component in the FA metabolic pathway (82). To date, three PPAR isoforms have been identified, namely PPARα, PPARγ and PPARβ/δ. PPARγ serves an important role in lipid synthesis, whilst PPARα and PPARδ mainly regulate oxidative phosphorylation, substrate transport and energy homeostasis (83). PPAR can regulate the M2 polarization of TAM through multiple pathways, which in turn promotes tumor proliferation, angiogenesis and immunosuppression (84).

PPARβ/δ can promote TAM polarization toward M2, tumor invasion and angiogenesis. A previous lipidomic analysis of ovarian cancer ascites has revealed that high concentrations of polyunsaturated FAs (PUFAs), particularly linoleic acid, can function as potent PPARβ/δ agonists in macrophages, thereby promoting the M2 polarization of TAMs (85). Sirtuin 4 (SIRT4) is a member of the SIRT family that can regulate cell proliferation and metabolism. It has been previously shown that upon downregulation of SIRT4 in human hepatocellular carcinoma, TAMs can activate the FAO/PPARβ/δ-STAT3 signaling pathway, which leads to M2 polarization (86,87).

A series of studies have shown that the intact structural PPAR system is required for the regulation of FAO. Caspase-1 activation generates a 41-kDa PPARγ fragment by cleaving PPARγ on Asp64. This fragment can then enter the mitochondria and inhibit medium-chain acyl-CoA dehydrogenase activity, reducing the efficiency of FAO and leading to lipid droplet accumulation, which in turn promotes M2 polarization (88-90). In addition, receptor-interacting protein kinase 3 (RIPK3) is another key factor mediating macrophage necrosis. It has been shown that in human and mouse hepatocellular carcinoma tissues, downregulation of RIPK3 can inhibit caspase-1-mediated PPAR cleavage, promote FAO, polarize TAMs toward the M2 phenotype and enhance tumor immunosuppression (89).

In addition, the FA binding protein (FABP) family serves another important role in FA metabolism, where its intracellular localization is involved in glioma progression (91). It has been previously shown that epidermal FABP is significantly overexpressed in mouse mammary carcinoma TAMs, which promotes the production of IFN-β by modulating lipid droplets, thereby recruiting immune effector cells and inhibiting tumor progression (92,93). By contrast, adipocyte/macrophage FABP is highly expressed in mouse and human breast cancer TAMs, where it promotes breast cancer cell proliferation and metastasis through the NF-κB/miR-29b/IL-6/STAT3 pathway (92,94). This suggests the different roles of different subtypes of FABP in cancer, where some types can promote tumor growth and metastasis, whilst others have oncolytic effects.

CD36 is a scavenger receptor that mediates lipid uptake, immune recognition, inflammation, molecular adhesion and apoptosis. This protein is a transmembrane glycoprotein and can bind to a variety of ligands, including FAs, to exert its effects (95). TAMs highly express CD36 and extensively utilize FAO for their energy supply. This process promotes mitochondrial oxidative phosphorylation and the production of ROS, leading to the activation of STAT6 and modulation of TAM polarization (66). S100A4 is another well-established pre-metastatic oncoprotein that is primarily expressed by macrophages in the tumor microenvironment. S100A4 has been shown to enhance CD36-mediated FA uptake through the PPARγ pathway, thereby promoting and polarizing TAMs towards the M2 phenotype (96).

Overall, the aforementioned previous observations have demonstrated that FA metabolism serves a role in promoting M2 polarization in TAMs to a certain degree. Therefore, it can be hypothesized that a comparable phenomenon may be present in GAMs. However, it must be emphasized that the existing body of experimental evidence on GAMs is insufficient to substantiate such a hypothesis. Further studies in this area are required for further advancement.

Phospholipid metabolism in TAM

Arachidonic acid (AA) is a membrane phospholipid produced by phospholipase A2 and is released into the cytosol. Known enzymes involved in AA metabolism include cytochrome P450, cyclooxygenase (COX) and lipoxygenase (LOX), which breaks AA down into hydroxyeicosatraenoic acids, prostaglandins and leukotrienes, respectively (97). In addition, AA or phospholipid metabolism in TAM mainly regulates the immune escape and proliferation of tumor cells.

A previous study has shown that TAMs can increase the expression of COX2 and prostaglandin E2 (PGE2) through the PI3K/Akt/mTOR pathway, leading to tamoxifen resistance and enhanced endocrine resistance in breast cancer (98). Meanwhile, PGE2 stimulates angiogenesis, suppresses immune function, promotes cancer cell migration and inhibits CD80 expression on tumor-associated phagocytes, thereby promoting cancer progression (99,100). TAM-derived osteopontin binds to α9β1 integrins, which upregulates COX2 expression, then increases the expression of PGE2 and MMP9 and accelerates angiogenesis (101). Another study showed that blocking the microsomal prostaglandin E synthase-1 and COX2 promoted TAM polarization toward M2 in colon cancer, thereby inhibiting tumor progression (102). Furthermore, it has also been previously shown that 5-LOX serves an important role in TAMs. In a metastatic lung cancer model, 5-LOX-expressing macrophages were observed to promote tumor cell proliferation by upregulating leukotriene B4 expression, whereas 5-LOX-suppressed macrophages exhibited reduced tumor proliferation (103,104). Similarly, reduced 5-LOX expression in human breast cancer TAMs can lead to decreased leukotriene synthesis and reduced effector T-cell recruitment, thereby promoting tumor progression (105).

Triglyceride metabolism in TAMs

Triglycerides are produced by the esterification of three hydroxyl groups on glycerol with three long-chain FA molecules and are involved in anabolism and catabolism. Anabolism is mainly regulated by diacylglycerol O-acyltransferases and monoacylglycerol O-acyltransferases, whilst catabolism is mainly regulated by hormone-sensitive lipase, abhydrolase domain-containing (ABHD)5, adipose triglyceride lipase and monoglyceride lipase (MGLL) (106,107). It has been shown that TAM can affect tumor development by regulating triglyceride metabolizing enzymes, where ABHD and MGLL serve a key role in this process.

ABHD is a key enzyme in triglyceride catabolism whilst also being able to inhibit autophagy and apoptosis in tumor cells (108). A previous study found that ABHD5 can promote the expression of spermine synthase (SRM) in TAMs of human and mouse colon cancer tissues. Spermine promotes apoptosis in tumor cells. Single-cell sequencing results also showed that high expression of ABHD5 in TAMs can promote tumor growth. Therefore, targeting the ABHD5/SRM/spermine axis in TAM may serve as a potential therapeutic strategy for colon cancer (109). Furthermore, it has been shown that ABHD5 in TAMs can increase MMP9 expression through the NF-κB pathway, thereby promoting the lung metastasis of colorectal cancer (110).

MGLL is another important component of the triglyceride catabolic pathway, which hydrolyzes triglycerides into free FAs. It has been previously found in mouse models of colon and breast cancer that MGLL deficiency can cause lipid accumulation in TAMs and promotes endocannabinoid receptor-2/TLR4 activation in TAMs, which enhances immunosuppression and promotes tumor progression (111).

Cholesterol metabolism in TAMs

Cholesterol is an important component of biological membranes. It regulates cell membrane fluidity and participates in various signaling pathways as a solubilizer of other lipids. Cholesterol metabolic reprogramming in TAMs has been previously shown to serve an important role in tumor development through TAM activation and recruitment, whilst promoting M2 polarization. It has been indicated that cholesterol metabolic reprogramming in TAMs mainly focuses on the alteration of the cholesterol efflux pathway. Therefore, targeting cholesterol efflux may be a potential method of controlling or treating cancer (112,113).

An ATP-binding cassette transporter (ABC) is a type of ATP-powered pump that consists of two transmembrane structural domains and two ATP-binding domains on the cytoplasmic side. ABC proteins scavenge surplus cholesterol within cells and regulate the balance of cholesterol to maintain homeostasis (84). Various cancers have been found to have elevated cholesterol levels (114). A recent study discovered that Apolipoprotein A (ApoA1) can enhance the removal of cholesterol from GAMs, decreasing intracellular cholesterol levels; this process was found to activate CD8+ T cells, enhancing anti-tumor immunity in a mouse GBM model (115). In another study, it was discovered that ABC-mediated cholesterol efflux from TAM membranes facilitated M2 polarization in a mouse model of metastatic ovarian cancer; this in turn led to IL-4-associated immunosuppression and invasive metastasis, whilst also inhibiting the IFN-γ-induced antitumor effects (116). Studies on bladder cancer and melanoma mouse models have also revealed a similar phenomenon, whereby ABCG1 can facilitate the removal of cholesterol to control the balance of cholesterol within cells. The absence of ABCG1 in mice led to activation of the NF-κB pathway and a transformation of macrophages from M2 to M1, resulting in enhanced direct cytotoxic effects on tumors, which hinders the growth of malignancies (114). However, further studies are required to clarify the effects of cholesterol metabolism in a cell type-specific context.

27-Hydroxycholesterol (27-HC) is a major metabolite of cholesterol that is catalyzed by its cytochrome P450 oxidase (CYP27A1). CYP27A1 is highly expressed in M2 macrophages and activates M2 polarization, thereby promoting tumor progression (117). It has been previously found that CYP27A1 is highly expressed in mouse breast cancer TAMs, whilst the 27-HC catabolic enzyme CYP27B1 is not expressed at high levels in breast cancer cells, a setup that results in the accumulation of 27-HC in the tumor cells. This accumulation of 27-HC in turn promotes the proliferation of the tumor cells and facilitates the expression of several chemokines by the TAMs, including chemokine (C-C motif) ligand (CCL)2 and CCL3, which then recruit monocytes to the tumor site to promote tumor progression (118).

The lipid metabolic reprogramming in TAMs and its regulatory mechanism for tumor progression are shown in Fig. 2.

Metabolite-driven phenotypic changes in TAM
Short-chain FAs (SCFAs)

The role of SCFAs in macrophages in inflammation has been extensively studied, but their role in TAMs has remained elusive. Therefore, this section will focus on their role in inflammation and, by extension, their role in tumor regulation.

During the inflammatory response, SCFAs can mediate both pro-inflammatory and anti-inflammatory effects. This phenomenon may be due to the expression and local concentration profiles of the different SCFA receptors and SCFAs themselves, respectively. It has been previously shown that in macrophages, SCFAs (namely butyrate) can bind to and activate free FA receptor (FFAR)3 to downregulate the levels of proinflammatory factors (including inducible nitric oxide synthase, TNF, MCP-1 and IL-6), thereby exerting anti-inflammatory effects (119,120). In addition, during airway inflammation, SCFAs can downregulate IL-8 expression by targeting FFAR2 and FFAR3 in macrophages, thereby exerting an anti-inflammatory effect and improving patient symptoms (121). These results suggest that SCFAs can exert potent anti-inflammatory effects that are realized through FFAR2 and FFAR3. Therefore, inhibitors of FFAR2/3 may mediate both proinflammatory and anti-tumor effects, to inhibit tumor progression. However, other studies have found that SCFAs can exert proinflammatory effects. It has been previously found that when FFAR2/3 is activated, it further activates mTOR, PI3K and MAPK signaling pathways downstream to mediate proinflammatory effects (122,123). In addition, SCFAs (acetate) were found to upregulate the production of proinflammatory cytokines and chemokines, such as CXCL1/2 and IL-6, by activating FFAR3/FFAR2 and ERK1/2 downstream. These aforementioned studies suggest that SCFAs can have opposite roles in inflammation. This phenomenon is associated with the local concentration of SCFAs.

It has been demonstrated that SCFAs can also regulate inflammation through the binding of hydroxycarboxylic acid receptor 2 (GPR109A), a butyrate receptor present in intestinal epithelial cells and immune cells that serves an important role in inflammation and immunity. Stimulation with IFN-γ has been shown to upregulate GPR109A expression in macrophages (124). GPR109A activation is involved in IL-8 and IL-10 production downstream, which affects regulatory T cells to reduce inflammation (125-128). Therefore, GPR109A may exert anti-inflammatory and immunomodulatory effects. However, to the best of our knowledge, there are relatively few relevant studies in TAMs, meaning that the relationship between GPR109A and tumors requires further exploration.

SCFAs can not only exert anti-inflammatory effects through signaling but also participate in inflammatory regulation by inhibiting histone deacetylase (HDAC). In macrophages, SCFAs (propionate and butyrate) have been observed to exert anti-inflammatory effects by inhibiting the TNF and NF-κB signaling pathways, in addition to inhibiting HDAC and promoting IL-10 production (129-131). However, it remains elusive which HDAC is inhibited and further studies are warranted.

To conclude, SCFAs serve a role in controlling inflammation in macrophages mainly through two primary modes of behavior. One method likely involves attaching to G protein-coupled receptors (such as FFAR2/3 and GPR109A) to trigger signaling pathways further downstream. Another method may involve the inhibition of HDAC once it has entered the cell, resulting in anti-inflammatory effects. Therefore, it would be of benefit to study whether a similar mechanism exists in GAMs in future studies, where SCFAs can potentially exert anti-inflammatory effects in addition to promoting tumor growth.

Long-chain PUFAs

Omega-3 FAs are a family of long-chain PUFAs that also includes α-linolenic acid, eicosapentaenoic acid (EPA) and docosahexaenoic acid (DHA). G-protein coupled receptor 120 (GPR120) is a G-protein coupled receptor that is involved in the regulation of metabolic, endocrine and immune functions. GPR120 can be activated by long-chain PUFAs (132). It has been previously shown that GPR120 is highly expressed in adipose tissue and proinflammatory macrophages in mice fed a high saturated fat diet, where the use of fish oil containing DHA and EPA can exert anti-inflammatory effects through GPR120 (133). In addition, mice fed an omega-3 diet had a significant reduction in the number of M2-like TAMs and expression of M2-associated cytokines, chemokines and growth factors in tumor tissues, compared with those in mice fed on an omega-6 diet (134). Data from another previous study corroborated this finding, as DHA was found to combine with ethanolamine to generate DHEA in breast cancer cells, reducing the secretion of CCL5 and affecting TAM recruitment and tumor progression. In addition, omega-3 FAs have been found to inhibit prostate cancer progression through a variety of mechanisms, including inhibition of COX2-mediated PGE2 formation, LOX activity, TLRs, formation of pro-resolvin metabolites, activation of PPARγ and inhibition of NF-κB (135).

Fig. 3 briefly shows that SCFAs and long-chain PUFAs can drive phenotypic changes in macrophages, which in turn can regulate inflammation and tumor progression.

Possibility of lipid metabolic modulation-based therapy

Traditional views suggest that M1-like TAMs prefer glycolysis as an energy source, whilst M2-like TAMs favor FAO (66,136). Therefore, regulating FAO may offer a strategy to inhibit tumor progression.

S100A4 is a well-known pre-metastatic oncoprotein that is primarily expressed by macrophages in the tumor microenvironment. S100A4 can enhance CD36-mediated FA uptake through the PPARγ pathway, promoting the polarization of TAMs towards the M2 phenotype (96). Previous studies have shown that injecting S100A4-knockout macrophages can significantly reduce tumor growth in mice (71). Similarly, using VT1021 to block CD36 lipid uptake was found to inhibit the M2 polarization of TAMs, thereby suppressing cancer (137,138).

The caspase-1/PPARγ/FAO axis is another crucial target for cancer therapy. A study has previously found that the caspase-1 inhibitor (Tyr-Val-Ala-Asp) can inhibit breast cancer progression by blocking caspase-1-mediated PPARγ cleavage (90). In addition, RIPK3 deficiency was found to inhibit caspase-1-mediated cleavage of PPARα and PPARγ in TAMs, leading to increased FAO and promoting hepatocellular carcinoma. This process can be inhibited by using the RIPK3 agonist decitabine (90). It has also been shown that lipid metabolism can be reprogrammed and TAM polarization reversed by blocking FAO using etomoxir (66,139).

Rofecoxib, a specific COX-2 inhibitor, has been documented to restore the adhesion and antitumor activity of TAMs (140). Celecoxib was also observed to exert similar effects (141). Another study previously showed that a selective COX-2 inhibitor, LM-1685, significantly reduced the level of arginase 1 in M2 macrophages, thereby inhibiting tumor progression (142).

Zileuton, a 5-LOX inhibitor, was found to decrease MMP7 expression whilst reducing TAM migration and infiltration (143,144).

A recombinant tumor lysing adenovirus carrying ApoA1 was previously designed to overexpress ApoA1 in the tumor microenvironment. This led to an increase in cholesterol removal from GAMs and a significant decrease in cholesterol levels within GAMs. As a result, GAMs were able to regain their ability to engulf and present antigens, which enhanced the effectiveness of CD8+ T cells in eliminating GBM. Furthermore, this treatment also induced a long-lasting immune response (115). In another study, ATR101, an inhibitor of ABC, was found to inhibit the M2 polarization of TAM by inhibiting cholesterol efflux from TAM, leading to cholesterol accumulation in cells (145).

Therapies that have been experimentally proven to be feasible through metabolic modulation are listed in Table I.

Table I

Possible therapies based on lipid metabolic modulation.

Table I

Possible therapies based on lipid metabolic modulation.

Target Drug/interventionMechanismTumor type(Refs.)
S100A4S100A4-KOInhibition of IL-4/S100A4/PPARγ/CD36/FAOBreast cancer(71,96)
CD36VT1021Inhibition of FA intake/FAOGBM(137,138)
Caspase-1YVADInhibition of caspase-1/PPARγ/FAOBreast cancer(90)
RIPK3DecitabineInhibition of caspase-1/PPARγ/FAO(89)
FAOEtomoxir; 25-HCInhibition of FAOHepatocellular carcinoma(66,139)
COX-2 Rofecoxib/CelecoxibRestoration of the adhesion and antitumor activity of TAMHead and neck squamous cell carcinoma/Breast cancer(140,141)
LM-1685Reduction of Arg1 levels in M2Colon carcinoma(142)
5-LOXZileutonReduction of migration and invasion of TAMLung cancer(143,144)
ApoA1 AdVAPOA1Increase of cholesterol removal from GAMsGBM(115)
ABCATR101Inhibition of the efflux of TAM cholesterolLung cancer(145)

[i] TAM, tumor-associated macrophage. All abbreviations used in the table are defined in Table SI.

However, to the best of our knowledge, few therapies targeting the metabolic pathway of GAMs are available at present and the aforementioned drugs have not yet been tested in clinical trials in patients with glioma or animal models of glioma. It remains speculative whether similar drugs may be effective in GAMs. Further research is needed to explore these possibilities.

Summary and prospect

GAMs play an important role in the tumor microenvironment, influencing glioma growth, invasion and angiogenesis through specific signaling molecules like MCP-1, GM-CSF and VEGF. They exhibit high plasticity, allowing them to differentiate into various phenotypes under different stimuli, adapting their metabolic pathways to support tumor progression.

While recent studies have highlighted significant alterations in lipid metabolism within TAMs, literature on lipid metabolic reprogramming in GAMs remains scarce. More in-depth studies focusing on GAMs are essential to understanding their unique metabolic adaptations and roles in glioma. Targeted therapies modulating lipid metabolism in GAMs hold promise for inhibiting tumor progression. Inhibiting FAO and targeting pathways involving COX-2 and PGE2 have shown potential in preclinical studies. Developing drugs specifically modulating GAM metabolic pathways may provide more effective treatment options for glioma.

Future research should focus on specific drug development for GAMs, targeting lipid metabolism and other pathways unique to these cells. Testing potential metabolic modulation drugs in preclinical or clinical trials is urgently needed to evaluate their efficacy and safety. Considering the complexity of tumor metabolic pathways, multi-targeted therapeutic strategies may enhance therapeutic outcomes by disrupting the tumor's metabolic network comprehensively.

In addition, identifying and validating biomarkers for monitoring metabolic reprogramming and therapeutic response is crucial. Biomarkers will help optimize treatment regimens, improve efficacy and provide critical information for personalized therapy. Further research into GAM metabolic reprogramming mechanisms will provide a foundation for developing novel therapeutic approaches, potentially in combination with existing treatments like immunotherapy and chemotherapy.

By addressing these areas, future research can advance the understanding of GAM metabolism, leading to effective therapies for glioma, ultimately improving patient outcomes.

Supplementary Data

Availability of data and materials

Not applicable.

Authors' contributions

Writing-original draft preparation: YM and YH; writing-review and editing: FH and KS; visualization: YM and YH; supervision: FH and KS. All authors have read and agreed to the published version of the manuscript. Data authentication is not applicable.

Ethics approval and consent to participate

Not applicable.

Patient consent for publication

Not applicable.

Competing interests

The authors declare that they have no competing interests.

Acknowledgements

Not applicable.

Funding

This research was funded by the National Natural Science Foundation of China (grant no. 82203683), the National Key Research and Development Program of China (grant no. 2023YFC2510001) and the Chinese Society of Clinical Oncology Foundation-Zai Lab Cancer Treatment Research Foundation (grant no. Y-zai2021/qn-0217).

References

1 

Laug D, Glasgow SM and Deneen B: A glial blueprint for gliomagenesis. Nat Rev Neurosci. 19:393–403. 2018. View Article : Google Scholar : PubMed/NCBI

2 

Wang J, Leavenworth JW, Hjelmeland AB, Smith R, Patel N, Borg B, Si Y and King PH: Deletion of the RNA regulator HuR in tumor-associated microglia and macrophages stimulates anti-tumor immunity and attenuates glioma growth. Glia. 67:2424–2439. 2019. View Article : Google Scholar : PubMed/NCBI

3 

Jiang Y, Marinescu VD, Xie Y, Jarvius M, Maturi NP, Haglund C, Olofsson S, Lindberg N, Olofsson T, Leijonmarck C, et al: Glioblastoma cell malignancy and drug sensitivity are affected by the cell of origin. Cell Rep. 18:977–990. 2017. View Article : Google Scholar : PubMed/NCBI

4 

Ostrom QT, Gittleman H, Liao P, Vecchione-Koval T, Wolinsky Y, Kruchko C and Barnholtz-Sloan JS: CBTRUS statistical report: Primary brain and other central nervous system tumors diagnosed in the United States in 2010-2014. Neuro Oncol. 19(Suppl 5): V1–V88. 2017. View Article : Google Scholar : PubMed/NCBI

5 

Uddin MS, Mamun AA, Alghamdi BS, Tewari D, Jeandet P, Sarwar MS and Ashraf GM: Epigenetics of glioblastoma multiforme: From molecular mechanisms to therapeutic approaches. Semin Cancer Biol. 83:100–120. 2022. View Article : Google Scholar

6 

Claus EB, Walsh KM, Wiencke JK, Molinaro AM, Wiemels JL, Schildkraut JM, Bondy ML, Berger M, Jenkins R and Wrensch M: Survival and low-grade glioma: The emergence of genetic information. Neurosurg Focus. 38:E62015. View Article : Google Scholar : PubMed/NCBI

7 

Hambardzumyan D, Gutmann DH and Kettenmann H: The role of microglia and macrophages in glioma maintenance and progression. Nat Neurosci. 19:20–27. 2016. View Article : Google Scholar :

8 

Mantovani A, Sozzani S, Locati M, Allavena P and Sica A: Macrophage polarization: Tumor-associated macrophages as a paradigm for polarized M2 mononuclear phagocytes. Trends Immunol. 23:549–555. 2002. View Article : Google Scholar : PubMed/NCBI

9 

Mantovani A, Sica A, Sozzani S, Allavena P, Vecchi A and Locati M: The chemokine system in diverse forms of macrophage activation and polarization. Trends Immunol. 25:677–686. 2004. View Article : Google Scholar : PubMed/NCBI

10 

Xia L, Oyang L, Lin J, Tan S, Han Y, Wu N, Yi P, Tang L, Pan Q, Rao S, et al: The cancer metabolic reprogramming and immune response. Mol Cancer. 20:282021. View Article : Google Scholar : PubMed/NCBI

11 

Yang K, Wang X, Song C, He Z, Wang R, Xu Y, Jiang G, Wan Y, Mei J and Mao W: The role of lipid metabolic reprogramming in tumor microenvironment. Theranostics. 13:1774–1808. 2023. View Article : Google Scholar : PubMed/NCBI

12 

Hanahan D and Weinberg RA: Hallmarks of cancer: The next generation. Cell. 144:646–674. 2011. View Article : Google Scholar : PubMed/NCBI

13 

Fernandez LP, Gomez de Cedron M and Ramirez de Molina A: Alterations of lipid metabolism in cancer: Implications in prognosis and treatment. Front Oncol. 10:5774202020. View Article : Google Scholar : PubMed/NCBI

14 

Chen JQ and Russo J: Dysregulation of glucose transport, glycolysis, TCA cycle and glutaminolysis by oncogenes and tumor suppressors in cancer cells. Biochim Biophys Acta. 1826:370–384. 2012.PubMed/NCBI

15 

Xiang Y and Miao H: Lipid metabolism in tumor-associated macrophages. Adv Exp Med Biol. 1316:87–101. 2021. View Article : Google Scholar : PubMed/NCBI

16 

Vander Heiden MG and DeBerardinis RJ: Understanding the intersections between metabolism and cancer biology. Cell. 168:657–669. 2017. View Article : Google Scholar : PubMed/NCBI

17 

Venneti S and Thompson CB: Metabolic reprogramming in brain tumors. Annu Rev Pathol. 12:515–545. 2017. View Article : Google Scholar : PubMed/NCBI

18 

Bi J, Chowdhry S, Wu S, Zhang W, Masui K and Mischel PS: Altered cellular metabolism in gliomas-an emerging landscape of actionable co-dependency targets. Nat Rev Cancer. 20:57–70. 2020. View Article : Google Scholar

19 

Pavlova NN and Thompson CB: The emerging hallmarks of cancer metabolism. Cell Metab. 23:27–47. 2016. View Article : Google Scholar : PubMed/NCBI

20 

Belanger M, Allaman I and Magistretti PJ: Brain energy metabolism: Focus on astrocyte-neuron metabolic cooperation. Cell Metab. 14:724–738. 2011. View Article : Google Scholar : PubMed/NCBI

21 

Magistretti PJ and Allaman I: A cellular perspective on brain energy metabolism and functional imaging. Neuron. 86:883–901. 2015. View Article : Google Scholar : PubMed/NCBI

22 

Zielke HR, Zielke CL and Baab PJ: Direct measurement of oxidative metabolism in the living brain by microdialysis: A review. J Neurochem. 109(Suppl 1): S24–S29. 2009. View Article : Google Scholar

23 

Kaur B, Khwaja FW, Severson EA, Matheny SL, Brat DJ and Van Meir EG: Hypoxia and the hypoxia-inducible-factor pathway in glioma growth and angiogenesis. Neuro Oncol. 7:134–153. 2005. View Article : Google Scholar : PubMed/NCBI

24 

Kayama T, Yoshimoto T, Fujimoto S and Sakurai Y: Intratumoral oxygen pressure in malignant brain tumor. J Neurosurg. 74:55–59. 1991. View Article : Google Scholar : PubMed/NCBI

25 

Kucharzewska P, Christianson HC and Belting M: Global profiling of metabolic adaptation to hypoxic stress in human glioblastoma cells. PLoS One. 10:e01167402015. View Article : Google Scholar : PubMed/NCBI

26 

Li Z, Bao S, Wu Q, Wang H, Eyler C, Sathornsumetee S, Shi Q, Cao Y, Lathia J, McLendon RE, et al: Hypoxia-Inducible factors regulate tumorigenic capacity of glioma stem cells. Cancer Cell. 15:501–513. 2009. View Article : Google Scholar : PubMed/NCBI

27 

Ricard C, Tchoghandjian A, Luche H, Grenot P, Figarella-Branger D, Rougon G, Malissen M and Debarbieux F: Phenotypic dynamics of microglial and monocyte-derived cells in glioblastoma-bearing mice. Sci Rep. 6:263812016. View Article : Google Scholar : PubMed/NCBI

28 

del Rio-Hortega P: The third element of the nerve centers. Bulletin of the Spanish Society of Biology. 9:69–129. 1919.In Spanish.

29 

Simard AR and Rivest S: Bone marrow stem cells have the ability to populate the entire central nervous system into fully differentiated parenchymal microglia. FASEB J. 18:998–1000. 2004. View Article : Google Scholar : PubMed/NCBI

30 

Priller J, Flugel A, Wehner T, Boentert M, Haas CA, Prinz M, Fernández-Klett F, Prass K, Bechmann I, de Boer BA, et al: Targeting gene-modified hematopoietic cells to the central nervous system: Use of green fluorescent protein uncovers microglial engraftment. Nat Med. 7:1356–1361. 2001. View Article : Google Scholar : PubMed/NCBI

31 

Flugel A, Bradl M, Kreutzberg GW and Graeber MB: Transformation of donor-derived bone marrow precursors into host microglia during autoimmune CNS inflammation and during the retrograde response to axotomy. J Neurosci Res. 66:74–82. 2001. View Article : Google Scholar : PubMed/NCBI

32 

Hickey WF and Kimura H: Perivascular microglial cells of the CNS are bone marrow-derived and present antigen in vivo. Science. 239:290–292. 1988. View Article : Google Scholar : PubMed/NCBI

33 

Massengale M, Wagers AJ, Vogel H and Weissman IL: Hematopoietic cells maintain hematopoietic fates upon entering the brain. J Exp Med. 201:1579–1589. 2005. View Article : Google Scholar : PubMed/NCBI

34 

De Leo A, Ugolini A and Veglia F: Myeloid cells in glioblastoma microenvironment. Cells. 10:182020. View Article : Google Scholar : PubMed/NCBI

35 

Ajami B, Bennett JL, Krieger C, McNagny KM and Rossi FM: Infiltrating monocytes trigger EAE progression, but do not contribute to the resident microglia pool. Nat Neurosci. 14:1142–1249. 2011. View Article : Google Scholar : PubMed/NCBI

36 

Chen Z, Feng X, Herting CJ, Garcia VA, Nie K, Pong WW, Rasmussen R, Dwivedi B, Seby S, Wolf SA, et al: Cellular and molecular identity of tumor-associated macrophages in glioblastoma. Cancer Res. 77:2266–2278. 2017. View Article : Google Scholar : PubMed/NCBI

37 

Landry AP, Balas M, Alli S, Spears J and Zador Z: Distinct regional ontogeny and activation of tumor associated macrophages in human glioblastoma. Sci Rep. 10:195422020. View Article : Google Scholar : PubMed/NCBI

38 

Xu C, Xiao M, Li X, Xin L, Song J, Zhan Q, Wang C, Zhang Q, Yuan X, Tan Y and Fang C: Origin, activation, and targeted therapy of glioma-associated macrophages. Front Immunol. 13:9749962022. View Article : Google Scholar : PubMed/NCBI

39 

Bowman RL, Klemm F, Akkari L, Pyonteck SM, Sevenich L, Quail DF, Dhara S, Simpson K, Gardner EE, Iacobuzio-Donahue CA, et al: Macrophage ontogeny underlies differences in tumor-specific education in brain malignancies. Cell Rep. 17:2445–2459. 2016. View Article : Google Scholar : PubMed/NCBI

40 

Bennett ML, Bennett FC, Liddelow SA, Ajami B, Zamanian JL, Fernhoff NB, Mulinyawe SB, Bohlen CJ, Adil A, Tucker A, et al: New tools for studying microglia in the mouse and human CNS. Proc Natl Acad Sci USA. 113:E1738–E1746. 2016. View Article : Google Scholar : PubMed/NCBI

41 

Müller S, Kohanbash G, Liu SJ, Alvarado B, Carrera D, Bhaduri A, Watchmaker PB, Yagnik G, Di Lullo E, Malatesta M, et al: Single-cell profiling of human gliomas reveals macrophage ontogeny as a basis for regional differences in macrophage activation in the tumor microenvironment. Genome Biol. 18:2342017. View Article : Google Scholar : PubMed/NCBI

42 

Szulzewsky F, Pelz A, Feng X, Synowitz M, Markovic D, Langmann T, Holtman I R, Wang X, Eggen BJ, Boddeke HW, et al: Glioma-Associated microglia/macrophages display an expression profile different from M1 and M2 polarization and highly express Gpnmb and Spp1. PLoS One. 10:e01166442015. View Article : Google Scholar : PubMed/NCBI

43 

Pyonteck SM, Akkari L, Schuhmacher AJ, Bowman RL, Sevenich L, Quail DF, Olson OC, Quick ML, Huse JT, Teijeiro V, et al: CSF-1R inhibition alters macrophage polarization and blocks glioma progression. Nat Med. 19:1264–1272. 2013. View Article : Google Scholar : PubMed/NCBI

44 

Lisi L, Laudati E, Navarra P and Dello Russo C: The mTOR kinase inhibitors polarize glioma-activated microglia to express a M1 phenotype. J Neuroinflammation. 11:1252014. View Article : Google Scholar : PubMed/NCBI

45 

Qin T, Wang C, Chen X, Duan C, Zhang X, Zhang J, Chai H, Tang T, Chen H, Yue J, et al: Dopamine induces growth inhibition and vascular normalization through reprogramming M2-polarized macrophages in rat C6 glioma. Toxicol Appl Pharmacol. 286:112–123. 2015. View Article : Google Scholar : PubMed/NCBI

46 

Xu S, Wei J, Wang F, Kong LY, Ling XY, Nduom E, Gabrusiewicz K, Doucette T, Yang Y, Yaghi NK, et al: Effect of miR-142-3p on the M2 macrophage and therapeutic efficacy against murine glioblastoma. J Natl Cancer Inst. 106:dju1622014. View Article : Google Scholar : PubMed/NCBI

47 

Wang Q, Zhang J, Fang S, Wang J, Han X, Liu F and Jin G: P4HA1 down-regulation inhibits glioma invasiveness by promoting M1 microglia polarization. Onco Targets Ther. 14:1771–1782. 2021. View Article : Google Scholar : PubMed/NCBI

48 

Rao G, Latha K, Ott M, Sabbagh A, Marisetty A, Ling X, Zamler D, Doucette TA, Yang Y, Kong LY, et al: Anti-PD-1 Induces M1 polarization in the glioma microenvironment and exerts therapeutic efficacy in the absence of CD8 Cytotoxic T cells. Clin Cancer Res. 26:4699–4712. 2020. View Article : Google Scholar : PubMed/NCBI

49 

Feng X, Szulzewsky F, Yerevanian A, Chen Z, Heinzmann D, Rasmussen RD, Alvarez-Garcia V, Kim Y, Wang B, Tamagno I, et al: Loss of CX3CR1 increases accumulation of inflammatory monocytes and promotes gliomagenesis. Oncotarget. 6:15077–15094. 2015. View Article : Google Scholar : PubMed/NCBI

50 

Zhou C, Li T, Dong Q, Liang H and Xu L: SARM suppresses glioma progression in GL261 glioma cells and regulates microglial polarization. Cell Biol Int. 46:1927–1936. 2022. View Article : Google Scholar : PubMed/NCBI

51 

Wei J, Gabrusiewicz K and Heimberger A: The controversial role of microglia in malignant gliomas. Clin Dev Immunol. 2013:2852462013. View Article : Google Scholar : PubMed/NCBI

52 

Gutmann DH and Kettenmann H: Microglia/Brain macrophages as central drivers of brain tumor pathobiology. Neuron. 104:442–449. 2019. View Article : Google Scholar : PubMed/NCBI

53 

Zeppellini A, Galimberti S, Leone BE, Pacifico C, Riva F, Cicchiello F, Capici S, Maggioni C, Sala L and Cazzaniga ME: Comparison of tumor microenvironment in primary and paired metastatic ER+/HER2-breast cancers: Results of a pilot study. BMC Cancer. 21:2602021. View Article : Google Scholar

54 

Lailler C, Louandre C, Morisse MC, Lhossein T, Godin C, Lottin M, Constans JM, Chauffert B, Galmiche A and Saidak Z: ERK1/2 signaling regulates the immune microenvironment and macrophage recruitment in glioblastoma. Biosci Rep. 39:BSR201914332019. View Article : Google Scholar : PubMed/NCBI

55 

De Boeck A, Ahn BY, D'Mello C, Lun X, Menon SV, Alshehri MM, Szulzewsky F, Shen Y, Khan L, Dang NH, et al: Glioma-derived IL-33 orchestrates an inflammatory brain tumor microenvironment that accelerates glioma progression. Nat Commun. 11:49972020. View Article : Google Scholar : PubMed/NCBI

56 

Greten FR and Grivennikov SI: Inflammation and cancer: Triggers, mechanisms, and consequences. Immunity. 51:27–41. 2019. View Article : Google Scholar : PubMed/NCBI

57 

Zhang Q, Wang J, Yao X, Wu S, Tian W, Gan C, Wan X, You C, Hu F, Zhang S, et al: Programmed Cell Death 10 Mediated CXCL2-CXCR2 signaling in regulating tumor-associated microglia/macrophages recruitment in glioblastoma. Front Immunol. 12:6370532021. View Article : Google Scholar : PubMed/NCBI

58 

Anagnostakis F and Piperi C: Targeting options of tumor-associated macrophages (TAM) activity in gliomas. Curr Neuropharmacol. 21:457–470. 2023. View Article : Google Scholar :

59 

Sun L, Zhang H and Gao P: Metabolic reprogramming and epigenetic modifications on the path to cancer. Protein Cell. 13:877–919. 2022. View Article : Google Scholar :

60 

Liu Y, Xu R, Gu H, Zhang E, Qu J, Cao W, Huang X, Yan H, He J and Cai Z: Metabolic reprogramming in macrophage responses. Biomark Res. 9:12021. View Article : Google Scholar : PubMed/NCBI

61 

Wang Y, Wang D, Yang L and Zhang Y: Metabolic reprogramming in the immunosuppression of tumor-associated macrophages. Chin Med J (Engl). 135:2405–2416. 2022.PubMed/NCBI

62 

Muri J and Kopf M: Redox regulation of immunometabolism. Nat Rev Immunol. 21:363–381. 2021. View Article : Google Scholar

63 

Blouin CC, Pagé EL, Soucy GM and Richard DE: Hypoxic gene activation by lipopolysaccharide in macrophages: Implication of hypoxia-inducible factor 1alpha. Blood. 103:1124–1130. 2004. View Article : Google Scholar

64 

Van den Bossche J, Baardman J, Otto NA, van der Velden S, Neele AE, van den Berg SM, Luque-Martin R, Chen HJ, Boshuizen MC, Ahmed M, et al: Mitochondrial dysfunction prevents repolarization of inflammatory macrophages. Cell Rep. 17:684–696. 2016. View Article : Google Scholar : PubMed/NCBI

65 

Liu S, Liu J, Ma Q, Cao L, Fattah RJ, Yu Z, Bugge TH, Finkel T and Leppla SH: Solid tumor therapy by selectively targeting stromal endothelial cells. Proc Natl Acad Sci USA. 113:E4079–E4087. 2016.PubMed/NCBI

66 

Su P, Wang Q, Bi E, Ma X, Liu L, Yang M, Qian J and Yi Q: enhanced lipid accumulation and metabolism are required for the differentiation and activation of tumor-associated macrophages. Cancer Res. 80:1438–1450. 2020. View Article : Google Scholar : PubMed/NCBI

67 

Luo Q, Zheng NS, Jiang L, Wang T, Zhang P, Liu Y, Zheng P, Wang W, Xie G, Chen L, et al: Lipid accumulation in macrophages confers protumorigenic polarization and immunity in gastric cancer. Cancer Sci. 111:4000–4011. 2020. View Article : Google Scholar : PubMed/NCBI

68 

Huang SC, Everts B, Ivanova Y, O'Sullivan D, Nascimento M, Smith AM, Beatty W, Love-Gregory L, Lam WY, O'Neill CM, et al: Cell-intrinsic lysosomal lipolysis is essential for alternative activation of macrophages. Nat Immunol. 15:846–855. 2014. View Article : Google Scholar : PubMed/NCBI

69 

Teng Y, Xu L, Li W, Liu P, Tian L and Liu M: Targeting reactive oxygen species and fat acid oxidation for the modulation of tumor-associated macrophages: A narrative review. Front Immunol. 14:12244432023. View Article : Google Scholar : PubMed/NCBI

70 

Kumar S, Mittal S, Gupta P, Singh M, Chaluvally-Raghavan P and Pradeep S: Metabolic reprogramming in tumor-associated macrophages in the ovarian tumor microenvironment. Cancers (Basel). 14:52242022. View Article : Google Scholar : PubMed/NCBI

71 

Liu S, Zhang H, Li Y, Zhang Y, Bian Y, Zeng Y, Yao X, Wan J, Chen X, Li J, et al: S100A4 enhances protumor macrophage polarization by control of PPAR-γ-dependent induction of fatty acid oxidation. J Immunother Cancer. 9:e0025482021. View Article : Google Scholar

72 

Zhou D, Ji L and Chen Y: TSPO Modulates IL-4-Induced Microglia/Macrophage M2 Polarization via PPAR-gamma Pathway. J Mol Neurosci. 70:542–549. 2020. View Article : Google Scholar

73 

Dubey S, Ghosh S, Goswami D, Ghatak D and De R: Immunometabolic attributes and mitochondria-associated signaling of Tumor-Associated Macrophages in tumor microenvironment modulate cancer progression. Biochem Pharmacol. 208:1153692023. View Article : Google Scholar

74 

Puthenveetil A and Dubey S: Metabolic reprograming of tumor-associated macrophages. Ann Transl Med. 8:10302020. View Article : Google Scholar : PubMed/NCBI

75 

Shapouri-Moghaddam A, Mohammadian S, Vazini H, Taghadosi M, Esmaeili SA, Mardani F, Seifi B, Mohammadi A, Afshari JT and Sahebkar A: Macrophage plasticity, polarization, and function in health and disease. J Cell Physiol. 233:6425–6440. 2018. View Article : Google Scholar : PubMed/NCBI

76 

Lee LY, Oldham WM, He H, Wang R, Mulhern R, Handy DE and Loscalzo J: Interferon-ү impairs human coronary artery endothelial glucose metabolism by tryptophan catabolism and activates fatty acid oxidation. Circulation. 144:1612–1628. 2021. View Article : Google Scholar : PubMed/NCBI

77 

Friedmann Angeli JP, Xavier da Silva TN and Schilling B: CD8+ T cells PUF(A)ing the flames of cancer ferroptotic cell death. Cancer Cell. 40:346–348. 2022. View Article : Google Scholar : PubMed/NCBI

78 

Yerrapragada MR and Mampallil D: Interferon-γ detection in point of care diagnostics: Short review. Talanta. 245:1234282022. View Article : Google Scholar

79 

Lin CY, Chen WL, Huang YC, Lim CL and Yang CH: Gum Arabic in combination with IFN-γ promotes the M1 polarization in macrophage. Int J Biol Macromol. 209:506–512. 2022. View Article : Google Scholar : PubMed/NCBI

80 

Abdi K, Laky K, Abshari M, Hill EM, Lantz L, Singh NJ and Long EO: Dendritic cells Trigger IFN-γ secretion by NK cells independent of IL-12 and IL-18. Eur J Immunol. 52:1431–1440. 2022. View Article : Google Scholar : PubMed/NCBI

81 

Zhao X, Peng T, Cao X, Hou Y, Li R, Han T, Fan Z, Zhao M, Chang Y, Chen H, et al: In vivo G-CSF treatment activates the GR-SOCS1 axis to suppress IFN-y secretion by natural killer cells. Cell Rep. 40:1113422022. View Article : Google Scholar

82 

Chawla A: Control of Macrophage Activation and Function by PPARs. Circ Res. 106:1559–1569. 2010. View Article : Google Scholar : PubMed/NCBI

83 

Christofides A, Konstantinidou E, Jani C and Boussiotis VA: The role of peroxisome proliferator-activated receptors (PPAR) in immune responses. Metabolism. 114:1543382021. View Article : Google Scholar

84 

Qiao X, Hu Z, Xiong F, Yang Y, Peng C, Wang D and Li X: Lipid metabolism reprogramming in tumor-associated macrophages and implications for therapy. Lipids Health Dis. 22:452023. View Article : Google Scholar : PubMed/NCBI

85 

Schumann T, Adhikary T, Wortmann A, Finkernagel F, Lieber S, Schnitzer E, Legrand N, Schober Y, Nockher WA, Toth PM, et al: Deregulation of PPARβ/δ target genes in tumor-associated macrophages by fatty acid ligands in the ovarian cancer microenvironment. Oncotarget. 6:13416–13433. 2015. View Article : Google Scholar : PubMed/NCBI

86 

Fernandez-Marcos PJ and Serrano M: Sirt4: The glutamine gatekeeper. Cancer Cell. 23:427–428. 2013. View Article : Google Scholar : PubMed/NCBI

87 

Li Z, Li H, Zhao ZB, Zhu W, Feng PP, Zhu XW and Gong JP: SIRT4 silencing in tumor-associated macrophages promotes HCC development via PPARδ signalling-mediated alternative activation of macrophages. J Exp Clin Cancer Res. 38:4692019. View Article : Google Scholar

88 

Mojsilovic SS, Mojsilovic S, Villar VH and Santibanez JF: The metabolic features of tumor-associated macrophages: Opportunities for immunotherapy? Anal Cell Pathol (Amst). 2021:55230552021.PubMed/NCBI

89 

Wu L, Zhang X, Zheng L, Zhao H, Yan G, Zhang Q, Zhou Y, Lei J, Zhang J, Wang J, et al: RIPK3 orchestrates fatty acid metabolism in tumor-associated macrophages and hepatocarcinogenesis. Cancer Immunol Res. 8:710–721. 2020. View Article : Google Scholar : PubMed/NCBI

90 

Niu Z, Shi Q, Zhang W, Shu Y, Yang N, Chen B, Wang Q, Zhao X, Chen J, Cheng N, et al: Caspase-1 cleaves PPARγ for potentiating the pro-tumor action of TAMs. Nat Commun. 8:7662017. View Article : Google Scholar

91 

McKillop LH, Girardi CA and Thompson KJ: Role of fatty acid binding proteins (FABPs) in cancer development and progression. Cell Signal. 62:1093362019. View Article : Google Scholar : PubMed/NCBI

92 

Zhang Y, Sun Y, Rao E, Yan F, Li Q, Zhang Y, Silverstein KA, Liu S, Sauter E, Cleary MP and Li B: Fatty Acid-Binding Protein E-FABP restricts tumor growth by promoting IFN-β responses in tumor-associated macrophages. Cancer Res. 74:2986–2998. 2014. View Article : Google Scholar : PubMed/NCBI

93 

Furuhashi M and Hotamisligil GS: Fatty acid-binding proteins: role in metabolic diseases and potential as drug targets. Nat Rev Drug Discov. 7:489–503. 2008. View Article : Google Scholar : PubMed/NCBI

94 

Hao J, Yan F, Zhang Y, Triplett A, Zhang Y, Schultz DA, Sun Y, Zeng J, Silverstein KAT, Zheng Q, et al: Expression of adipocyte/macrophage fatty acid-binding protein in tumor-associated macrophages promotes breast cancer progression. Cancer Res. 78:2343–2355. 2018. View Article : Google Scholar : PubMed/NCBI

95 

Wang J and Li Y: CD36 tango in cancer: Signaling pathways and functions. Theranostics. 9:4893–4908. 2019. View Article : Google Scholar : PubMed/NCBI

96 

Nath A and Chan C: Genetic alterations in fatty acid transport and metabolism genes are associated with metastatic progression and poor prognosis of human cancers. Sci Rep. 6:186692016. View Article : Google Scholar : PubMed/NCBI

97 

Harizi H, Corcuff JB and Gualde N: Arachidonic-acid-derived eicosanoids: Roles in biology and immunopathology. Trends Mol Med. 14:461–469. 2008. View Article : Google Scholar : PubMed/NCBI

98 

Qin Q, Ji H, Li D, Zhang H, Zhang Z and Zhang Q: Tumor-associated macrophages increase COX-2 expression promoting endocrine resistance in breast cancer via the PI3K/Akt/mTOR pathway. Neoplasma. 68:938–946. 2021. View Article : Google Scholar : PubMed/NCBI

99 

Rabold K, Netea MG, Adema GJ and Netea-Maier RT: Cellular metabolism of tumor-associated macrophages-functional impact and consequences. FEBS Lett. 591:3022–3041. 2017. View Article : Google Scholar : PubMed/NCBI

100 

Olesch C, Sha W, Angioni C, Sha LK, Açaf E, Patrignani P, Jakobsson PJ, Radeke HH, Grösch S, Geisslinger G, et al: MPGES-1-derived PGE2 suppresses CD80 expression on tumor-associated phagocytes to inhibit anti-tumor immune responses in breast cancer. Oncotarget. 6:10284–10296. 2015. View Article : Google Scholar : PubMed/NCBI

101 

Kale S, Raja R, Thorat D, Soundararajan G, Patil TV and Kundu GC: Osteopontin signaling upregulates cyclooxygenase-2 expression in tumor-associated macrophages leading to enhanced angiogenesis and melanoma growth via alpha9beta1 integrin. Oncogene. 33:2295–2306. 2014. View Article : Google Scholar

102 

Nakanishi Y, Nakatsuji M, Seno H, Ishizu S, Akitake-Kawano R, Kanda K, Ueo T, Komekado H, Kawada M, Minami M and Chiba T: COX-2 inhibition alters the phenotype of tumor-associated macrophages from M2 to M1 in ApcMin/+ mouse polyps. Carcinogenesis. 32:1333–1339. 2011. View Article : Google Scholar : PubMed/NCBI

103 

Nosaka T, Baba T, Naito T, et al: Leukotriene B 4 generated by alveolar macrophages drive hepatocellular carcinoma lung metastasis. Hepatology. 66:85A. 2017.

104 

Hall Z, Ament Z, Wilson CH, Burkhart DL, Ashmore T, Koulman A, Littlewood T, Evan GI and Griffin JL: Myc expression drives aberrant lipid metabolism in lung cancer. Cancer Res. 76:4608–4618. 2016. View Article : Google Scholar : PubMed/NCBI

105 

Ringleb J, Strack E, Angioni C, Geisslinger G, Steinhilber D, Weigert A and Brüne B: Apoptotic cancer cells suppress 5-lipoxygenase in tumor-associated macrophages. J Immunol. 200:857–868. 2018. View Article : Google Scholar

106 

Cheng C, Geng F, Cheng X and Guo D: Lipid metabolism reprogramming and its potential targets in cancer. Cancer Commun (Lond). 38:272018.PubMed/NCBI

107 

Ou J, Miao H, Ma Y, Guo F, Deng J, Wei X, Zhou J, Xie G, Shi H, Xue B, et al: Loss of Abhd5 promotes colorectal tumor development and progression by inducing aerobic glycolysis and epithelial-mesenchymal transition. Cell Rep. 9:1798–1811. 2014. View Article : Google Scholar : PubMed/NCBI

108 

Yen CL, Nelson DW and Yen MI: Intestinal triacylglycerol synthesis in fat absorption and systemic energy metabolism. J Lipid Res. 56:489–501. 2015. View Article : Google Scholar :

109 

Miao H, Ou J, Peng Y, Zhang X, Chen Y, Hao L, Xie G, Wang Z, Pang X, Ruan Z, et al: Macrophage ABHD5 promotes colorectal cancer growth by suppressing spermidine production by SRM. Nat Commun. 7:117162016. View Article : Google Scholar : PubMed/NCBI

110 

Shang S, Ji X, Zhang L, Chen J, Li C, Shi R, Xiang W, Kang X, Zhang D, Yang F, et al: Macrophage ABHD5 Suppresses NFκB-Dependent matrix metalloproteinase expression and cancer metastasis. Cancer Res. 79:5513–5526. 2019. View Article : Google Scholar : PubMed/NCBI

111 

Xiang W, Shi R, Kang X, Zhang X, Chen P, Zhang L, Hou A, Wang R, Zhao Y, Zhao K, et al: Monoacylglycerol lipase regulates cannabinoid receptor 2-dependent macrophage activation and cancer progression. Nat Commun. 9:25742018. View Article : Google Scholar : PubMed/NCBI

112 

King RJ, Singh PK and Mehla K: The cholesterol pathway: Impact on immunity and cancer. Trends Immunol. 43:78–92. 2022. View Article : Google Scholar :

113 

van der Vorst EPC, Theodorou K, Wu Y, Hoeksema MA, Goossens P, Bursill CA, Aliyev T, Huitema LFA, Tas SW, Wolfs IMJ, et al: High-Density lipoproteins exert pro-inflammatory effects on macrophages via passive cholesterol depletion and PKC-NF-κB/STAT1-IRF1 signaling. Cell Metab. 25:197–207. 2017. View Article : Google Scholar

114 

Sag D, Cekic C, Wu R, Linden J and Hedrick CC: The cholesterol transporter ABCG1 links cholesterol homeostasis and tumour immunity. Nat Commun. 6:63542015. View Article : Google Scholar : PubMed/NCBI

115 

Wang S, Yan W, Kong L, Zuo S, Wu J, Zhu C, Huang H, He B, Dong J and Wei J: Oncolytic viruses engineered to enforce cholesterol efflux restore tumor-associated macrophage phagocytosis and anti-tumor immunity in glioblastoma. Nat Commun. 14:43672023. View Article : Google Scholar : PubMed/NCBI

116 

Goossens P, Rodriguez-Vita J, Etzerodt A, Masse M, Rastoin O, Gouirand V, Ulas T, Papantonopoulou O, Van Eck M, Auphan-Anezin N, et al: Membrane cholesterol efflux drives tumor-associated macrophage reprogramming and tumor progression. Cell Metab. 29:1376–1389.e4. 2019. View Article : Google Scholar : PubMed/NCBI

117 

Nelson ER, Wardell SE, Jasper JS, Park S, Suchindran S, Howe MK, Carver NJ, Pillai RV, Sullivan PM, Sondhi V, et al: 27-Hydroxycholesterol links hypercholesterolemia and breast cancer pathophysiology. Science. 342:1094–1098. 2013. View Article : Google Scholar : PubMed/NCBI

118 

Shi SZ, Lee EJ, Lin YJ, Chen L, Zheng HY, He XQ, Peng JY, Noonepalle SK, Shull AY, Pei FC, et al: Recruitment of monocytes and epigenetic silencing of intratumoral CYP7B1 primarily contribute to the accumulation of 27-hydroxycholesterol in breast cancer. Am J Cancer Res. 9:2194–2208. 2019.PubMed/NCBI

119 

Ohira H, Fujioka Y, Katagiri C, Mamoto R, Aoyama-Ishikawa M, Amako K, Izumi Y, Nishiumi S, Yoshida M, Usami M and Ikeda M: Butyrate attenuates inflammation and lipolysis generated by the interaction of adipocytes and macrophages. J Atheroscler Thromb. 20:425–442. 2013. View Article : Google Scholar : PubMed/NCBI

120 

Yao Y, Cai X, Fei W, Ye Y, Zhao M and Zheng C: The role of short-chain fatty acids in immunity, inflammation and metabolism. Crit Rev Food Sci Nutr. 62:1–12. 2022. View Article : Google Scholar

121 

Masui R, Sasaki M, Funaki Y, Ogasawara N, Mizuno M, Iida A, Izawa S, Kondo Y, Ito Y, Tamura Y, et al: G protein-coupled receptor 43 moderates gut inflammation through cytokine regulation from mononuclear cells. Inflamm Bowel Dis. 19:2848–2856. 2013. View Article : Google Scholar : PubMed/NCBI

122 

Seljeset S and Siehler S: Receptor-specific regulation of ERK1/2 activation by members of the 'free fatty acid receptor' family. J Recept Signal Transduct Res. 32:196–201. 2012. View Article : Google Scholar : PubMed/NCBI

123 

Tan J, McKenzie C, Potamitis M, Thorburn AN, Mackay CR and Macia L: The role of short-chain fatty acids in health and disease. Adv Immunol. 121:91–119. 2014. View Article : Google Scholar : PubMed/NCBI

124 

Schaub A, Fütterer A and Pfeffer K: PUMA-G, an IFN-gamma-inducible gene in macrophages is a novel member of the seven transmembrane spanning receptor superfamily. Eur J Immunol. 31:3714–3725. 2001. View Article : Google Scholar : PubMed/NCBI

125 

Singh N, Gurav A, Sivaprakasam S, Brady E, Padia R, Shi H, Thangaraju M, Prasad PD, Manicassamy S, Munn DH, et al: Activation of Gpr109a, receptor for niacin and the commensal metabolite butyrate, suppresses colonic inflammation and carcinogenesis. Immunity. 40:128–139. 2014. View Article : Google Scholar : PubMed/NCBI

126 

Karunaratne TB, Okereke C, Seamon M, Purohit S, Wakade C and Sharma A: Niacin and Butyrate: Nutraceuticals targeting dysbiosis and intestinal permeability in parkinson's disease. Nutrients. 13:282020. View Article : Google Scholar : PubMed/NCBI

127 

Bach Knudsen KE, Laerke HN, Hedemann MS, Nielsen TS, Ingerslev AK, Gundelund Nielsen DS, Theil PK, Purup S, Hald S, Schioldan AG, et al: Impact of diet-modulated butyrate production on intestinal barrier function and inflammation. Nutrients. 10:14992018. View Article : Google Scholar : PubMed/NCBI

128 

Chai JT, Digby JE and Choudhury RP: GPR109A and vascular inflammation. Curr Atheroscler Rep. 15:3252013. View Article : Google Scholar : PubMed/NCBI

129 

Aoyama M, Kotani J and Usami M: Butyrate and propionate induced activated or non-activated neutrophil apoptosis via HDAC inhibitor activity but without activating GPR-41/GPR-43 pathways. Nutrition. 26:653–661. 2010. View Article : Google Scholar

130 

Chang PV, Hao L, Offermanns S and Medzhitov R: The microbial metabolite butyrate regulates intestinal macrophage function via histone deacetylase inhibition. Proc Natl Acad Sci USA. 111:2247–2252. 2014. View Article : Google Scholar : PubMed/NCBI

131 

Usami M, Kishimoto K, Ohata A, Miyoshi M, Aoyama M, Fueda Y and Kotani J: Butyrate and trichostatin A attenuate nuclear factor kappaB activation and tumor necrosis factor α secretion and increase prostaglandin E2 secretion in human peripheral blood mononuclear cells. Nutr Res. 28:321–328. 2008. View Article : Google Scholar : PubMed/NCBI

132 

Hara T, Hirasawa A, Ichimura A, Kimura I and Tsujimoto G: Free fatty acid receptors FFAR1 and GPR120 as novel therapeutic targets for metabolic disorders. J Pharm Sci. 100:3594–3601. 2011. View Article : Google Scholar : PubMed/NCBI

133 

Oh DY, Talukdar S, Bae EJ, Imamura T, Morinaga H, Fan W, Li P, Lu WJ, Watkins SM and Olefsky JM: GPR120 is an omega-3 fatty acid receptor mediating potent anti-inflammatory and insulin-sensitizing effects. Cell. 142:687–698. 2010. View Article : Google Scholar : PubMed/NCBI

134 

Liang P, Henning SM, Guan J, Grogan T, Elashoff D, Olefsky JM, Cohen P and Aronson WJ: Role of Host GPR120 in mediating dietary omega-3 fatty acid inhibition of prostate cancer. J Natl Cancer Inst. 111:52–59. 2019. View Article : Google Scholar :

135 

Liang P, Henning SM, Schokrpur S, Wu L, Doan N, Said J, Grogan T, Elashoff D, Cohen P and Aronson WJ: Effect of dietary omega-3 fatty acids on tumor-associated macrophages and prostate cancer progression. Prostate. 76:1293–1302. 2016. View Article : Google Scholar : PubMed/NCBI

136 

Zhu L, Zhao Q, Yang T, Ding W and Zhao Y: Cellular metabolism and macrophage functional polarization. Int Rev Immunol. 34:82–100. 2015. View Article : Google Scholar

137 

Wu H, Han Y, Rodriguez Sillke Y, Deng H, Siddiqui S, Treese C, Schmidt F, Friedrich M, Keye J, Wan J, et al: Lipid droplet-dependent fatty acid metabolism controls the immune suppressive phenotype of tumor-associated macrophages. EMBO Mol Med. 11:e106982019. View Article : Google Scholar : PubMed/NCBI

138 

Ahluwalia M, Battiste J, Bockorny B, Bullock A, Patel MR, Wen P, Shepard D, Vaickus L, Vincent M, Vincent M, et al: Clinical efficacy and biomarker assessment of VT1021, A CD36/CD47 dual-targeting agent, in recurrent glioblastoma. Neuro-Oncology. 23:50. 2021. View Article : Google Scholar

139 

Zhang Q, Wang H, Mao C, Sun M, Dominah G, Chen L and Zhuang Z: Fatty acid oxidation contributes to IL-1β secretion in M2 macrophages and promotes macrophage-mediated tumor cell migration. Mol Immunol. 94:27–35. 2018. View Article : Google Scholar

140 

Lang S, Tiwari S, Andratschke M, Loehr I, Lauffer L, Bergmann C, Mack B, Lebeau A, Moosmann A, Whiteside TL and Zeidler R: Immune restoration in head and neck cancer patients after in vivo COX-2 inhibition. Cancer Immunol Immunother. 56:1645–1652. 2007. View Article : Google Scholar : PubMed/NCBI

141 

Zheng X, Mansouri S, Krager A, Grimminger F, Seeger W, Pullamsetti SS, Wheelock CE and Savai R: Metabolism in tumour-associated macrophages: A quid pro quo with the tumour microenvironment. Eur Respir Rev. 29:2001342020. View Article : Google Scholar : PubMed/NCBI

142 

Eruslanov E, Daurkin I, Ortiz J, Vieweg J and Kusmartsev S: Tumor-mediated induction of myeloid-derived suppressor cells and M2-polarized macrophages by altering intracellular PGE2 catabolism in myeloid cells. J Leukoc Biol. 88:839–848. 2010. View Article : Google Scholar : PubMed/NCBI

143 

Wen Z, Liu H, Li M, Li B, Gao W, Shao Q, Fan B, Zhao F, Wang Q, Xie Q, et al: Increased metabolites of 5-lipoxygenase from hypoxic ovarian cancer cells promote tumor-associated macrophage infiltration. Oncogene. 34:1241–1252. 2015. View Article : Google Scholar

144 

Nosaka T, Baba T, Tanabe Y, Sasaki S, Nishimura T, Imamura Y, Yurino H, Hashimoto S, Arita M, Nakamoto Y and Mukaida N: Alveolar macrophages drive hepatocellular carcinoma lung metastasis by generating leukotriene B4. J Immunol. 200:1839–1852. 2018. View Article : Google Scholar : PubMed/NCBI

145 

Hoppstadter J, Dembek A, Horing M, Schymik HS, Dahlem C, Sultan A, Wirth N, Al-Fityan S, Diesel B, Gasparoni G, et al: Dysregulation of cholesterol homeostasis in human lung cancer tissue and tumour-associated macrophages. Ebiomedicine. 72:1035782021. View Article : Google Scholar : PubMed/NCBI

Related Articles

Journal Cover

November-2024
Volume 54 Issue 5

Print ISSN: 1107-3756
Online ISSN:1791-244X

Sign up for eToc alerts

Recommend to Library

Copy and paste a formatted citation
x
Spandidos Publications style
Ma Y, Huang Y, Hu F and Shu K: Lipid metabolic rewiring in glioma‑associated microglia/macrophages (Review). Int J Mol Med 54: 102, 2024.
APA
Ma, Y., Huang, Y., Hu, F., & Shu, K. (2024). Lipid metabolic rewiring in glioma‑associated microglia/macrophages (Review). International Journal of Molecular Medicine, 54, 102. https://doi.org/10.3892/ijmm.2024.5426
MLA
Ma, Y., Huang, Y., Hu, F., Shu, K."Lipid metabolic rewiring in glioma‑associated microglia/macrophages (Review)". International Journal of Molecular Medicine 54.5 (2024): 102.
Chicago
Ma, Y., Huang, Y., Hu, F., Shu, K."Lipid metabolic rewiring in glioma‑associated microglia/macrophages (Review)". International Journal of Molecular Medicine 54, no. 5 (2024): 102. https://doi.org/10.3892/ijmm.2024.5426