Next Article in Journal
Pharmacological Properties and Molecular Targets of Alisol Triterpenoids from Alismatis Rhizoma
Next Article in Special Issue
The Role of Non-Coding RNAs in Glioma
Previous Article in Journal
The Multifaceted Roles of Autophagy in Infectious, Obstructive, and Malignant Airway Diseases
Previous Article in Special Issue
The Intricate Epigenetic and Transcriptional Alterations in Pediatric High-Grade Gliomas: Targeting the Crosstalk as the Oncogenic Achilles’ Heel
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Lipid Metabolism in Glioblastoma: From De Novo Synthesis to Storage

1
Department of Radiation Oncology, Ohio State Comprehensive Cancer Center, Arthur G. James Cancer Hospital and Richard J. Solove Research Institute, College of Medicine at The Ohio State University, Columbus, OH 43012, USA
2
Center for Cancer Metabolism, James Comprehensive Cancer Center at The Ohio State University, Columbus, OH 43210, USA
*
Author to whom correspondence should be addressed.
Biomedicines 2022, 10(8), 1943; https://doi.org/10.3390/biomedicines10081943
Submission received: 12 July 2022 / Revised: 1 August 2022 / Accepted: 6 August 2022 / Published: 11 August 2022
(This article belongs to the Special Issue Glioma Metabolism, Epigenetics, and Microenvironment)

Abstract

:
Glioblastoma (GBM) is the most lethal primary brain tumor. With limited therapeutic options, novel therapies are desperately needed. Recent studies have shown that GBM acquires large amounts of lipids for rapid growth through activation of sterol regulatory element-binding protein 1 (SREBP-1), a master transcription factor that regulates fatty acid and cholesterol synthesis, and cholesterol uptake. Interestingly, GBM cells divert substantial quantities of lipids into lipid droplets (LDs), a specific storage organelle for neutral lipids, to prevent lipotoxicity by increasing the expression of diacylglycerol acyltransferase 1 (DGAT1) and sterol-O-acyltransferase 1 (SOAT1), which convert excess fatty acids and cholesterol to triacylglycerol and cholesteryl esters, respectively. In this review, we will summarize recent progress on our understanding of lipid metabolism regulation in GBM to promote tumor growth and discuss novel strategies to specifically induce lipotoxicity to tumor cells through disrupting lipid storage, a promising new avenue for treating GBM.

1. Introduction

Fatty acids and cholesterol are two essential lipids that constitute the basic structure of cellular membranes [1]. Their derivatives can act as important signaling molecules to regulate cellular function [2]. However, dysregulation of fatty acid and cholesterol metabolism can cause various metabolic diseases, i.e., obesity, fatty liver, atherosclerosis, neutral lipid storage disease (NLSD), and cholesterol storage disease (CESD) [3,4]. Increasing evidence shows that lipid alterations also accelerate neurodegenerative disease progression by promoting oligomerization of pathogenic proteins and brain inflammation [5,6]. In recent years, following extensive investigation on energy metabolism [7], the role of abnormal lipid metabolism in cancer cells has rapidly received increasing attention [1,8,9,10]. New observations indicate addictive de novo lipid synthesis in various types of cancers [1]. Thus, targeting lipid synthesis has become a promising new direction for cancer therapy, as demonstrated by the many preclinical tumor models and Phase I/II clinical trials (Table 1) [1,9].
Glioblastoma (GBM) is the most lethal primary brain tumor [11]. Unfortunately, there has not been significant progress to improve GBM outcome over the past two decades despite extensive clinical testing [12]. Recent studies show that GBM needs abundant lipids for rapid growth [13,14,15,16], but accumulation of intracellular lipids, especially free fatty acids and cholesterol, can cause lipotoxicity [17,18]. How GBM cells protect themselves against excess lipid accumulation and lipotoxicity is an intriguing question. Our recent studies demonstrated that GBM cells prevent lipotoxicity and maintain tumor growth by dynamically storing excess lipids into lipid droplets (LDs), a specific lipid storage organelle [19,20,21,22]. In this review, we will summarize recent progress on our understanding of lipid metabolism regulation in GBM to maintain cellular homeostasis and tumor growth via coordination of lipid synthesis, uptake and storage processes, and will discuss a new strategy to target GBM involving disruption of lipid storage specifically in tumor cells.

2. Standard GBM Therapy

GBM is a WHO grade IV glioma, with a median survival post-diagnosis of only approximately 15 months despite extensive treatments [23]. GBM represents more than 60% of all brain tumors in adults and occurs in 2–3 cases per 100,000 individuals in Europe and North America [24]. Approximately 90% of GBM cases occur as primary tumors that have wild-type isocitrate dehydrogenase (IDH) and less than three months of clinical symptoms before diagnosis, while less than 5% of all cases are secondary tumors with IDH mutations, progressing from WHO grade II/III gliomas and with better prognosis [12,25,26,27]. The standard treatment for GBM is to first perform maximal surgical resection, followed by concomitant radiotherapy and alkylating agent Temozolomide (TMZ) for 6 weeks, and then continuation of TMZ alone every 4 weeks for six cycles of maintenance treatment [28]. Unfortunately, this treatment is only effective for a few months and almost all GBM tumors unavoidably recur after treatment [29]. The 5-year survival for patients with GBM is only around 6.8% in the United States [30].
In the clinic, except for TMZ, only Bevacizumab, a humanized monoclonal antibody against vascular endothelial growth factor (VEGF), has been approved by the FDA for the treatment of recurrent GBM and not for newly diagnosed GBM [31,32]. It is effective at reducing symptoms and improving the quality of life only for a short time [25]. However, Bevacizumab has no combinatory effect with TMZ and radiotherapy [25]. Moreover, it reduces the uptake of TMZ, fails to increase the overall survival, and has significant side effects such as hypertension, venous thrombosis, and infections [25].
Current investigations for new approaches targeting GBM have centered on molecularly targeted therapies, immunotherapy, and brain-targeted drug delivery [33]. For example, 57% of GBM cases show evidence of amplification or gain of function mutation of EGFR (EGFRvIII) [34]. Unfortunately, results from clinical trials for monoclonal antibodies and tyrosine kinase inhibitors (TKI) targeting EGFR have been disappointing, with no therapeutic benefit or being no better than TMZ [12,35]. Moreover, although 61% [36] to 88% [37] of GBM patients have tumors expressing PD-L1, clinical trials inhibiting PD-L1 or PD1 have also shown disappointing results [38]. Collectively, the failure to make significant progress in targeting GBM indicates that more efforts are needed to understand the biology of GBM, which is foundational to identify effective approaches antagonizing GBM.

3. Lipid Metabolism Regulation in GBM

3.1. Lipogenesis

Glucose is the main source for lipid production through the de novo synthesis pathway [1]. Glucose through glycolysis generates pyruvate that is converted to acetyl-CoA in the mitochondria by the pyruvate dehydrogenase (PDH) [1,39]. Condensation of acetyl-CoA with oxaloacetate (OAA) forms citrate, which is then released into the cytosol by the SLC25A1 transporter and converted back to acetyl-CoA by ATP-citrate lyase (ACLY). Acetyl-CoA then serves as a precursor for fatty acid and cholesterol synthesis [1,40] (Figure 1). In addition, acetyl-CoA synthetase 2 (ACSS2) can convert cytosolic acetate to acetyl-CoA for de novo fatty acid and cholesterol synthesis (Figure 1) to promote tumor growth [1]. ACSS2 is upregulated in GBM [41] and in hepatocellular carcinoma (HCC), myeloma, prostate, and bladder cancers [42]. Furthermore, acetyl-CoA is also involved in epigenetic regulation as it enters into the nucleus to modify histone proteins by direct acetylation [43].
De novo fatty acid synthesis is commonly observed in the liver, adipose tissue, and lactating breast, and normal cells usually assimilate fatty acid through the uptake of extracellular lipids [44]. However, actively proliferating cells, such as cancer cells and effector T cells, increase their fatty acid synthesis, uptake, and oxidation (FAO) for the synthesis of structural lipids and energy expenditure [9,45]. Expression of the enzymes controlling de novo fatty acid synthesis, such as ACLY, ACSS2, acetyl-CoA carboxylases (ACC), fatty acid synthase (FASN), and stearoyl-CoA desaturase 1 (SCD1), and mitochondria citrate transporter SLC25A1, is mainly regulated by sterol regulatory element-binding protein 1 (SREBP-1), a master transcription factor that regulates fatty acid and cholesterol synthesis (Figure 1) [13,14,15,46,47]. Inhibitors targeting these enzymes have been developed for the treatment of a broad type of cancers (Table 1) [9]. Carnitine palmitoyltransferase 1 (CPT1), the rate-limiting enzyme involved in mitochondrial fatty acid oxidation, which allows fatty acyl-CoA to enter into the mitochondria matrix from the cytosol, is also an important target of lipid metabolism for cancer therapy [48,49].
Like in most cancers, metabolism is reprogrammed in GBM and plays an important role during tumor initiation and progression, with increased glycolysis, altered nucleotide metabolism, and glutamine addiction [50,51]. We were the first to report that fatty acid synthesis is highly elevated in GBM to promote rapid tumor growth [1,13,14,15]. We found that the oncogenic EGFR/PI3K/AKT pathway increases fatty acid synthesis by stimulating the activation of SREBP-1 [14,39]. Inactivating SREBP-1 pharmacologically with its inhibitor Fatostatin or by shRNA knockdown significantly suppresses GBM tumor growth in vitro and in vivo [14,19,52]. Directly targeting FASN, a downstream transcriptional target of SREBP-1 and a key enzyme in controlling de novo fatty acid synthesis, with its inhibitor C75 effectively suppresses GBM growth [14]. Moreover, inhibiting fatty acid and cholesterol synthesis with the AMP-activated protein kinase (AMPK) agonist AICAR (5-aminoimidazole-4-carboxamide-1-β-D-ribofuranoside) also reduces GBM growth in a xenograft mouse model [13,16]. Furthermore, our studies revealed that a small non-coding RNA, miR-29, mediates a negative feedback loop to control SREBP expression and lipogenesis [53,54]. SREBP-1 transcriptionally upregulates pre-miR-29 expression, while mature miR-29 can inversely inhibit SREBP-1 expression via binding to the 3′-untranslational region (3-UTR) of SREBP-1 mRNA to induce its degradation and suppress SREBP translation. Delivering miR-29 to GBM cells significantly suppresses tumorigenesis in orthotopic mouse model by inhibiting SREBP-1 and lipogenesis [53,54]. Collectively, these data demonstrate that inhibiting lipogenesis is a promising new direction for GBM therapy.
Interestingly, a recent study reports that the brain metastasis from seven breast cell lines that harbor PI3K activation is dependent on the SREBF1 gene that encodes the SREBP-1 protein [55]. A significant inhibition of metastatic brain is observed in SREBF1 knockout breast cancer cell lines [55], demonstrating the importance of SREBP-1 in brain metastasis from breast cancers. Whether other types of brain metastatic cell lines have a similar SREBF1 dependency needs further investigation. In addition, directly targeting SREBP-1 downstream targets should be tested in these cell lines to determine whether fatty acid or cholesterol synthesis play major roles for breast cancer brain metastasis.

3.2. SREBP Activation, Connecting Glucose and Glutamine to Lipid Synthesis

GBM growth consumes large amounts of glucose and amino acids [56], but the mechanism by which tumor cells sense their levels to trigger lipid synthesis has been left unanswered for a long time. Our most recent study identified that glucose and glutamine coordinate to activate SREBP-1 to trigger de novo lipid synthesis in GBM and various other cancer cells [57]. The SREBP family includes three isoforms, SREBP-1a, -1c, and -2 [58,59,60]. SREBP-1c mainly regulates the expression of genes controlling fatty acid synthesis, while SREBP-2 regulates cholesterol synthesis and uptake, and SREBP-1a, which has the highest transcriptional activity, regulates all three processes [1,61,62,63]. SREBPs are synthesized as ~125 kD inactive precursors, which are spatially restrained in the endoplasmic reticulum (ER) membrane and are activated through a tightly controlled ER–Golgi–nucleus translocation process [59,62]. SREBPs bind to SREBP-cleavage activating protein (SCAP), which further binds to COPII-coated vesicles to move from the ER to the Golgi [62,64]. In the Golgi, SREBPs are sequentially cleaved by site-1 and -2 proteases to release their N-terminal forms (~65 kD) that then enter the nucleus to activate lipogenic gene expression [65,66,67,68,69]. Interestingly, SCAP/SREBP trafficking is inhibited by an ER-resident protein, insulin-inducible gene protein (Insig), which includes two isoforms, Insig-1 and -2 [70,71]. Insig binds to SCAP to retain the SCAP/SREBP complex in the ER (Figure 1) [59,64]. Cholesterol or 25-hydroxycholesterol (25-HC) can bind to SCAP or Insig to further enhance their association, which represents a negative feedback loop to modulate SREBP activation (Figure 1) [70,72,73].
The key step activating the SCAP/Insig dissociation for subsequent SREBP translocation has just been elucidated in a recent study by Cheng et al. [57]. Our previous study showed that glucose stimulates SREBP activation and lipogenesis by promoting SCAP N-glycosylation and stability [46,74,75,76]. Unexpectedly, our most recent study shows that when glutamine is removed from the medium, glucose alone is unable to activate SREBPs and lipogenesis, even with low cholesterol levels and in the presence of SCAP N-glycosylation [57]. We uncovered an unprecedented role of ammonia, which is released by glutaminolysis and acts as a key activator of the dissociation of N-glycosylated SCAP from Insig by inducing dramatic conformational changes in the SCAP transmembrane domain through interaction via hydrogen bonds with the side chains of three residues, i.e., aspartate D428, serine S326 and S330, eventually leading to SREBP activation and lipid synthesis (Figure 1) [57]. We further unveiled the competitive role of 25-HC that blocks ammonia binding to SCAP, thereby keeping SCAP bound to Insig and suppressing SREBP activation. Blocking ammonia binding to SCAP by mutating D428 to alanine (D428A) dramatically suppressed both GBM and lung cancer growth in orthotopic xenograft models. In addition, we provided further physiological evidence for a connection between glutaminolysis and lipogenesis by showing the molecular link between glutaminase (GLS) expression and SREBP-1 activation in human lung cancer and glioma tissues. Moreover, the study demonstrated that the activation of SREBPs and lipogenesis by glutamine/ammonia in concert with glucose also occurs in melanoma, liver and breast cancer cells in addition to lung cancer and GBM cells, suggesting that this is a common mechanism at play in a wide range of cancer types [57]. Thus, this study revealed the essential role of ammonia in the regulation of SCAP/Insig dissociation, SREBP activation and lipid metabolism, and identified SCAP as a critical sensor connecting glutamine, glucose, and lipid metabolism to promote tumor growth (Figure 1) [57]. The study also suggests that targeting the key molecular link between glutaminolysis, glucose and lipid metabolism might be a promising strategy for treating malignancies and metabolic syndromes.

3.3. Membrane Phospholipid Remodeling

Phospholipids (PLs) constitute the main membrane structure as they form lipid bilayers through their amphiphilic characteristics [77]. Elevated fatty acid synthesis promotes PL formation and membrane expansion to facilitate rapid tumor growth [1,78,79]. The composition of the fatty acid chains in PLs is not static, but is dynamically remodeled through the coordinated activities of phospholipase A (PLA) and lysophosphatidylcholine acyltransferases (LPCATs) [80]. PLA removes an esterified fatty acid from the sn-2 position of PLs to convert them to lysophosphatidylipids, while LPCATs add back fatty acids to lysophosphatidylipids, thereby remaking PLs [81,82]. Thus, fatty acid chains in membrane PLs are modified by these two enzymes to adjust to the dynamic alterations of the cellular environment [82]. The LPCAT family contains four members (LPCAT1-4) [83,84]. LPCAT1 mainly regulates the incorporation of saturated fatty acids into lysophosphatidylcholine (LPC) to form PLs, thereby increasing the levels of saturated PLs. Interestingly, LPCAT1 has been shown to be upregulated in various cancers, including GBM [85], HCC [86], clear cell renal cell cancer [87], esophageal squamous cell carcinoma (ESCC) [88], gastric [89], and breast cancer [90]. Its upregulation regulates oncogenic EGFR signaling in GBM by affecting EGFR and its constitutively active mutant EGFRvIII protein stability [85]. Targeting LPCAT1 effectively suppresses GBM [85] and ESCC growth [88]. LPCAT1 elevation across cancer types suggests that the increase in membrane saturated PL levels might prevent lipid peroxidation and ferroptosis in cancer cells, thereby facilitating tumor growth. LPCAT3 regulates the incorporation of polyunsaturated fatty acids, such as arachidonic acid, thereby increasing membrane fluidity and flexibility [84]. LPCAT3 has been shown to be a major player to facilitate lipid peroxidation and ferroptosis via increasing membrane polyunsaturated PL level [91,92,93,94]. Nevertheless, there has no study clearly demonstrating the expression levels of LPCPAT3 in tumor tissues and its role in tumorigenesis.

3.4. Cholesterol Uptake

Cholesterol, which is an indispensable lipid molecule for maintaining cell viability [95], is inserted between phospholipid molecules to control membrane integrity and fluidity [96], and accounts for 30–40% of membrane lipids in the plasma membrane [97,98,99,100]. Cholesterol is esterified with fatty acids to convert to neutral cholesteryl esters (CE), which are carried by low-density lipoprotein (LDL) to circulate in the bloodstream and be absorbed by different tissues [101]. LDL is endocytosed into cells as mediated by the LDL receptor (LDLR) and is then delivered to the lysosomes, where LDL is broken down by lysosomal lipases to release free cholesterol, which then egresses from the lysosomes for maintenance and expansion of the plasma membrane but also of the membranes of all cellular organelles such as the mitochondria and endoplasmic reticulum (ER) (Figure 1) [15,102,103,104,105,106].
Recent studies from our group showed that GBM cells acquire abundant cholesterol for rapid growth by elevating LDL uptake [15]. GBM increases the expression of the LDL receptor (LDLR), which enhances LDL absorption to provide sufficient cholesterol [15]. We further showed that EGFR signaling activates SREBP-1 to upregulate LDLR expression and promote cholesterol uptake for GBM growth [15]. Inducing LDLR degradation by increasing expression of its E3 ligase Idol (inducible degrader of LDLR) with the LXR agonist GW3965 inhibits GBM growth [15]. Therefore, limiting LDL/cholesterol uptake appears as a promising new direction for GBM therapy.

3.5. Lipid storage and Energy Homeostasis

Excess free fatty acids and cholesterol can cause lipotoxicity, leading to cell death when the levels are uncontrolled [107,108]. In the human body, excess fatty acids and cholesterol are sequestered into the adipocytes of fat tissues within specialized organelles known as lipid droplets (LDs) after being converted to triacylglycerol (TAG) and cholesteryl esters (CE) (Figure 1) [109]. Interestingly, in 2016, Geng et al. reported for the first time that LDs are highly prevalent in human GBM tumor tissues, while not observed in normal brain tissues [19,20]. Using transmission electronic microscopy, they clearly showed that LDs are present in the cytosol of GBM tumor cells. In following studies, the group demonstrated that when glucose level decreases, GBM cells mobilize LD-stored fatty acids into the mitochondria, a process mediated by autophagy for beta-oxidation and energy production, which maintain energy homeostasis and GBM survival [21]. Thus, LDs serve two functions in GBM. First, they act as a lipotoxicity-preventing organelle, buffering excess fatty acids and cholesterol [17,22]. Second, they also serve as an energy reservoir, providing fuel to maintain energy homeostasis when glucose levels are reduced [21]. Thus, targeting LD formation could be a promising new approach for GBM therapy, which is discussed in the sections below.

4. The Basics of LDs

4.1. LD Formation

LDs are organelles containing a core of neutral lipids, mainly TAG and CE, which is covered by a monolayer of phospholipids [4,109,110,111,112]. LDs exist in almost all organisms, from bacteria, yeast, and plants to mammals [113]. Mammalian LDs were first described in 1886 with improvement in microscopy technology but were ignored for more than a century [114]. In the 1990s, the discovery of perilipin, the first LD membrane protein to be identified, and that of Chanarin–Dorfman Syndrome, the first genetic LD disorder [114], incited scientists to begin exploring the characteristics of LDs and their roles in metabolic diseases. Nevertheless, despite major advances in recent years, the mechanism of LD formation is still unclear [109]. Three hypotheses have been advanced to explain LD formation, and the “budding model” is the most widely accepted [115]. The first step in LD biogenesis is the synthesis of TAG and CE derived from fatty acids and cholesterol in the ER, which are catalyzed by diacylglycerol acyltransferases (DGAT1 and DGAT2) and sterol O-acyltransferases (SOAT1 and SOAT2), also named acyl-CoA:cholesterol acyltransferases (ACAT1 and ACAT2) [109]. As the concentration of TAG and CE increases in the interspace between the bilayer leaflets of the ER membrane, the neutral lipids eventually bud from the ER membrane when the surface tension reaches a critical point and get covered with a phospholipid monolayer derived from the ER two leaflets [109,116,117,118]. This budding process is facilitated by multiple ER proteins, such as the fat-storage-inducing transmembrane (FIT) proteins and Seipin [109,119,120,121,122,123,124]. Because of the instability of the phospholipid monolayer, perilipin traffics from the ER to the surface of LDs to reduce the membrane surface tension. It is still unclear whether LDs bud from ER randomly or from preferential sites. Finally, LDs grow and mature through droplet-to-droplet fusion or lipid synthesis on the LD surface [109,124,125,126,127,128].

4.2. LD Size and Composition

The most predominant features of LDs are their heterogeneity and diversity. The number, size, and composition of LDs vary widely between cells or even within the same cell depending on different conditions [109,112,129,130,131]. Only one LD with a diameter often >100 µm is found in most adipocytes, whereas a greater number of LDs are found in non-adipocyte cells and are usually <1 µm in diameter [132,133]. However, the size and number of LDs change dramatically under pathophysiological conditions. For example, during the progression of non-alcoholic steatohepatitis (NASH), the increased number and size of LDs in hepatocytes causes the activation of hepatic stellate cells, leading to inflammation and liver fibrosis [134]. Interestingly, LDs are loaded with excess oleic acid and cholesterol in adrenal and liver cells, and two subpopulations of LDs, i.e., TG-rich and CE-rich LDs, are found in the cytosol and coated with different members of the perilipin family [135]. Translation inhibitors only induce formation of CE-rich LDs in 3Y1 cells by an unknown mechanism [136]. The preferential lipid composition of LDs and how it is established needs further investigation.

4.3. LD Interaction and Proteomes

LDs are cytoplasmic organelles that tightly communicate with other cellular organelles, such as the ER, mitochondria, peroxisomes, Golgi apparatus, and lysosomes [137]. LDs extensively form membrane contact sites to exchange materials such as lipids, metabolites, and ions with other organelles [109,138,139], as well as to regulate organelle division, trafficking, and inheritance [109,140,141]. Dysregulation of LD contact site proteins are found in many genetic, infectious, and metabolic diseases [137]. Recently, LDs were found to reside also in the cell nucleus and were termed “nuclear LDs (nLD)” [142,143,144,145]. Although the mechanism of nLD formation is still unclear, their function in gene transcription regulation has been described in yeast [146], hepatic cell lines [147], and primary hepatocytes [148].
Because of the extensive contact of LDs with other organelles, purification by optimized fractionation approaches still contain contaminants from other associated organelles, especially the ER [149], which has hampered the identification of the profile of LD proteomes. However, the recent development of the proximity labeling strategy enables high-confidence cataloguing of LD proteomes, which are composed of 100–150 proteins in mammalian cells [112,150]. These proteins are categorized into several distinct functional classes, involving structure maintenance, lipid metabolism, signaling, and membrane trafficking [4]. They can also be classified into two classes according to differences in trafficking pathways, i.e., class I LD proteins that are first inserted into the ER through hydrophobic hairpins and then traffic to the surface of LDs through membrane bridges, and class II LD proteins that are synthesized in the cytosol and inserted into LDs through a lipid anchor or amphipathic α-helices [109,112]. LD protein degradation is mediated by the ubiquitin-dependent pathway or the autophagic pathway [112].

5. Inhibiting LD Formation for GBM Therapy

LDs had been reported to be abundant in many cancer types, although not in every subtype [111]. In the early 1960s, LDs were found in the mammary carcinoma of a female patient [151] and abundantly in more than 100 cases of malignant lymphoma [152]. However, few people realized the importance of LDs in cancer research at that time. LDs have now been suggested to be involved in all processes of cancer development, including initiation, promotion, and progression [111]. The hypotheses regarding LD function in cancer cells are numerous, i.e., promoting tumor proliferation [153,154,155], driving epithelial–mesenchymal transition (EMT) [115,156,157], mediating radio-resistance [158,159] and chemo-resistance [160,161,162,163], maintaining cancer cell stemness [164,165,166], and avoiding immune destruction [167,168,169]. Nevertheless, these hypotheses need to be experimentally validated.
Our group reported for the first time that human patients with glioma exhibit LDs in large numbers that correlated with GBM progression and poor survival [19]. Our studies suggested that targeting SOAT1 and DGAT1 could be a promising new strategy for GBM therapy as neutral lipid synthesis catalyzed by SOAT1 and DGAT1 initiates the formation of LDs in GBM

5.1. Targeting SOAT1 to Disrupt Cholesterol Homeostasis for GBM Therapy

Cholesterol is an essential lipid, but excess cholesterol is toxic to cells; thus, the intracellular cholesterol levels need to be precisely balanced [1,20]. Cholesterol homeostasis is regulated through a complex network, including uptake, biosynthesis, efflux, storage, and trafficking [1,96,170]. Disrupting this network could trigger toxicity for cells. The ER is the key cellular organelle that senses changes in cholesterol levels. A previous study using Chinese hamster ovary (CHO) cells as a model showed that even a low 5% increase in ER cholesterol can trigger a feedback reaction that blocks SREBPs from trafficking to the Golgi and being activated, which slows down cholesterol synthesis and uptake in order to maintain cholesterol homeostasis [58,171]. Intracellular cholesterol levels can also be reduced by promoting cholesterol efflux through the ATP-binding cassette (ABC) transporter superfamily, including the ABC subfamily A 1 (ABCA1) and ABC subfamily G (ABCG) 1, 5, and 8 members in a cell-type dependent manner [96,172,173]. Our previous study showed that promoting cholesterol efflux and at the same time reducing cholesterol uptake by activating the liver X receptor (LXR) inhibits GBM growth by disturbing cholesterol balance [15].
Another strategy developed by cells to overcome lipotoxicity from excess cholesterol is to convert free cholesterol to CE in the ER and then store CE in LDs, a process controlled by SOATs [174,175]. SOATs belong to the membrane-bound O-acyltransferase (MBOAT) superfamily [176] and include two isoenzymes, i.e., SOAT1 and SOAT2. SOAT1 is mainly located in the ER and has been detected in most cell types and tissues, while SOAT2 is expressed predominantly in fetal liver and intestine cells and at low levels in other tissues [177,178,179]. SOATs, especially SOAT1, have long been studied as a therapeutic target for atherosclerosis and Alzheimer’s disease due to their importance in cholesterol metabolism [179,180].
In recent years, SOAT1 has started to gain significant consideration in the cancer field as accumulating evidence suggests that SOAT1 is a novel and promising drug target in many cancer types. Yue et al. reported that inhibition of cholesterol esterification by the SOAT1 inhibitors Avasimibe and Sandoz 58-035 suppresses prostate cancer cell proliferation and tumor growth in vivo [181]. Other groups also reported that SOAT1 is upregulated in prostate cancer tissues and is associated with earlier recurrence [182,183], and genetic inhibition of SOAT1 leads to the suppression of tumor growth in mice [182]. In hepatocellular carcinoma, SOAT1 is highly expressed in tumor tissues, and inhibition of SOAT1 by Avasimibe significantly inhibits tumor growth in vivo [184,185,186,187]. SOAT1 has also been shown to play an important role in pancreatic, colorectal, gastric, and lung cancers [188,189,190,191]. A novel SOAT1 inhibitor, Nevanimibe, also known as ATR-101, has been tested in a Phase I study in advanced adrenocortical carcinoma, although it showed only limited efficacy (Table 1) [192].
Our group discovered that SOAT1 expression is highly upregulated in tumor tissues from GBM patients, while SOAT2 is undetectable in tumor tissues [19,20]. Genetic or pharmacological inhibition of SOAT1 suppresses cholesterol storage and LD/CE formation, causing an increase in ER cholesterol, which suppresses SREBP-1 activation and reduces fatty acid synthesis, leading to suppression of GBM growth in vitro and in an orthotopic mouse model (Figure 2) [19,20]. As development of clinically effective pharmacological SREBP-1 inhibitors has failed in the past decades [193], indirect inhibition of SREBP-1 such as inhibition of SOAT1 may be an alternative strategy. Recently, high expression of SOAT1 was also found in glioma-associated macrophages, suggesting that inhibition of SOAT1 might also trigger an immune response to GBM cells [194]. One of the current challenges for SOAT1 inhibition in the clinic is that most SOAT1 inhibitors were manufactured as candidate drugs to treat atherosclerosis; thus, it is still unknown whether they could cross the blood–brain barrier (BBB). Therefore, development of novel SOAT1 inhibitors that could efficiently cross the BBB are worth developing and testing.

5.2. Targeting DGAT1 to Disrupt Fatty Acid Homeostasis for GBM Therapy

Fatty acids (FAs) are not only essential components of the membrane phospholipids but also serve as energy source through mitochondria-mediated β-oxidation [195]. Nevertheless, excess free fatty acid can impair cellular homeostasis and disrupts tissue function [107]. In 2003, Listenberger et al. reported that excess accumulation of palmitic acid (C16:0 saturated fatty acid) induces apoptosis in cultured CHO cells, while oleic acid (C18:1 monounsaturated fatty acid) supplementation leads to TAG accumulation [196]. Moreover, oleic acid rescues palmitate-induced apoptosis by channeling palmitate into TG pools, away from pathways leading to apoptosis. They suggested that palmitate-induced apoptosis in CHO cells occurs though a mechanism involving an increase in reactive oxygen species (ROS) and ceramide. The same year, another group reported that saturated fatty acids induce apoptosis in breast cancer cells through triggering cardiolipin turnover in mitochondria and cytochrome c release [197]. Interestingly, other studies reported that ceramide-mediated apoptosis could be prevented by generating acyl-ceramide and sequestrating it in LDs [198,199].
DGATs convert excessive free FAs into TAG that are stored in LDs [17,22]. In humans, there are two DGAT enzymes, DGAT1 and DGAT2, which are members of two different gene families and differ in their tissue distribution [200,201]. DGAT1 is highly expressed in the small intestine and DGAT2 is mainly expressed in liver and adipose tissues [202,203]. Loss of function mutations in DGAT1 were identified in patients with congenital diarrheal disorders [204] and protein-losing enteropathy [205]. DGAT2 knockout mice died soon after birth [206], while DGAT1 knockout mice only showed less TAG in liver and mammary glands [207]. Both DGAT1 and DGAT2 are located in the ER membrane and DGAT2 is a class I LD protein [109]. The two enzymes also have non-canonical acyltransferase activities with different substrate preference [201].
Our recent study demonstrated that targeting DGAT1 to block TAG synthesis causes severe FA metabolism dysregulation in GBM cells, leading to mitochondria damage and ROS production that trigger tumor cell apoptosis (Figure 3) [17,22]. We found that DGAT1 is overexpressed in tumor tissues from GBM patients and genetic or pharmacological inhibition of DGAT1, but not DGAT2, suppresses LD/TAG formation and reduces GBM tumor growth (Figure 3) [17]. Interestingly, inhibition of DGAT1 and DGAT2 is required to block LD/TAG formation and induce cell death in HepG2 cells [17]. It is still unclear why DGAT1 and DGAT2 behave differently in various cell types and which transcription factor regulates their expression in cancer cells. In addition, our study showed that the DGAT1 inhibitor A922500 dramatically suppresses GBM tumor growth in a subcutaneous mouse model (Table 1) [17]. Thus, it will be worthwhile to test if it effectively crosses the BBB. Targeting DGAT1 to block FA storage and disrupt homeostasis emerges as a promising new direction for GBM therapy. Furthermore, many publications have shown that DGAT1 is a better target than DGAT2 for cancer therapy [157,208,209,210,211,212,213].

6. Other Targets in LDs

Gene expression analysis shows that hypoxia-inducible lipid droplet associated (HILPDA), one of the LD-associated proteins, is highly expressed in bladder, colon, kidney, lung, pancreas, and uterine cancers as well as in GBM [214]. HILPDA increases LD accumulation by inhibiting TG hydrolysis by the adipose triglyceride lipase (ATGL) and stimulating TG synthesis via DGAT1 [210,215]. Mao et al. reported that HILPDA overexpression induced by hypoxia contributed to Bevacizumab resistance in GBM [216]. It may be a potential novel therapeutic target for GBM. Analysis of The Cancer Genome Atlas (TCGA) database shows that CGI-58, a co-activator of ATGL that stimulates TAG lipolysis and decreases LD numbers [217], is overexpressed in GBM and cervical cancer. Shi et al. reported that CGI-58 is upregulated in endometrial cancer and high CGI-58 expression was a poor prognostic marker for endometrial cancer [218]. However, a recent study showed that CGI-58 is a tumor suppressor, inducing cell cycle arrest in the G1 phase and causing growth retardation in a panel of prostate cancer cells [219]. The function of CGI-58 in GBM and whether it can affect tumor growth in GBM need to be explored further.

7. Conclusions and Future Directions

It is only recently that more scientists have realized the importance of lipid metabolism reprogramming in cancer cells. Our recent reports unveiled that lipid rewiring is a hallmark of GBM [1,22]. Drugs targeting lipid metabolism pathways such as de novo synthesis, uptake, storage, and catabolism are now being actively developed for cancer therapy, including therapy based on LD formation and regulation. Resolving the structures of SOAT1 [220,221,222] and DGAT1 [223,224] certainly will accelerate the discovery of potent inhibitors targeting these two enzymes for GBM therapy. Another alternative option is to develop new brain-targeted drug delivery systems based on nanoparticles to transfer effective drugs to the brain, possibly reducing the unexpected side effects due to offsite targeting.
Many questions remain regarding lipid metabolism regulation in GBM. Which transcription factors regulate the expression of SOAT1 and DGAT1? How GBM cells balance lipid synthesis, lipid storage, and lipolysis? Could potent SREBP-1 inhibitors be developed and tested clinically for cancer therapy? Are LDs playing a role in therapy resistance and tumor recurrence? Are there other targets regulating the formation of LDs in GBM as there are over 100 proteins locating or interacting with LDs [112,150]? Could other proteins in addition to DGAT1 and SOAT1 serve as druggable targets for cancer therapy? Addressing these questions certainly will provide new insights into the understanding of lipid metabolism in GBM and will help to develop novel therapeutic approaches.
Table 1. Molecular targets and inhibitors in the lipid metabolism pathway for cancer therapy.
Table 1. Molecular targets and inhibitors in the lipid metabolism pathway for cancer therapy.
TargetInhibitorsTypes of CancerPreclinical EvidenceClinical TrialsReferences
SREBPsFatostatin, Betulin, PF-429242, dipyridamoleGBM, breast, uterine, prostate, liver, lung,
kidney, pancreatic and colon
Xenografts [1,19,225,226,227,228,229,230,231,232,233,234,235]
ACLYSB-201076, Hydroxycitric acid, Bempedoic acid, BMS-303141, SB-204990,Lung, prostateXenografts [1,236,237]
ACCsND-646, ND-654, soraphen A, TOFALung, liver, breastXenografts,
DEN-injured rats
[17,156,238,239,240,241,242,243,244]
FASNTVB-2640, C75, cerulenin, orlistatLung, colon, breast, GBM, astrocytoma, prostate, uterineXenograftsNCT03808558
NCT02223247
NCT02980029
NCT03179904
NCT05118776
NCT03032484
[185,187,192,245,246,247,248,249]
SCD1A939572, MF-438, CAY10566
MK-8245
Lung, pancreatic, kidney, GBM, ovarianXenograft,
Pdx1Cre; LSL-KrasG12D mouse
NCT00972322
(type 2 diabetes)
[164,250,251,252,253,254,255,256]
ACSS2Tetrazoles, Pyridine Derivatives [257,258]
SLC25A1CTPi2, BTA, CNASB [259,260]
CPT1AEtomoxir, Perhexiline, ST1326GBM, breast, leukemia, prostate, colonXenografts,
Transgenic mice
[261,262,263,264,265,266,267,268]
DGAT1A922500, AZD3988,
PF-04620110
GBM, liver, uterine, prostateXenografts [17,156,242,243,244,269]
SOAT1Avasimibe, ATR-101, K604,Liver, colon, GBM, kidneyP53-deficient mice, AOM/DSS-treated and ApcMin/+ mice, xenografts NCT01898715[185,187,192,246,247]

Funding

This work was supported by National Institute of Neurological Disorders and Stroke (NINDS) and National Cancer Institute (NCI) of United States grants R01NS104332, R01NS112935, R01CA227874, and R01CA240726 to D.G. and American Cancer Society (United States) Research Scholar grant RSG-14-228-01-CSM to D.G. We also appreciate the support from OSUCCC-Pelotonia (United States) Idea grant and Urban and Shelly Meyer Foundation to D.G. And The APC was funded by OSUCCC start-up funds.

Institutional Review Board Statement

Not applicable.

Informed Consent Statement

Not applicable.

Data Availability Statement

Not applicable.

Acknowledgments

The authors wish to thank Martine Torres for her careful reading of the manuscript and editorial assistance. All the figures were created with BioRender.com (accessed on 5 May 2022).

Conflicts of Interest

The authors declare no conflict of interest.

Abbreviation

3-UTR3′-untranslational region
25-HC25-hydroxycholesterol
α-KGα-ketoglutaric acid
ABCATP-binding cassette
ABCA1ATP-binding cassette subfamily A member 1
ACATacyl-CoA: cholesterol acyltransferase
ACCacetyl-CoA carboxylases
ACLYATP-citrate lyase
ACSLlong-chain acyl-CoA synthetase
ACSS2acetyl-CoA synthetase 2
AICAR5-aminoimidazole-4-carboxamide-1-β-D-ribofuranoside
AKTprotein kinase B
AMPKAMP-activated protein kinase
ASCT2alanine/serine/cysteine-preferring transporter 2
ATGLadipose triglyceride lipase
BBBblood–brain barrier
COPIIcoat protein complex II
CPT1carnitine palmitoyltransferase 1
CEcholesteryl esters
CESDcholesterol storage disease
DGAT1diacylglycerol acyltransferase 1
EGFRepidermal growth factor receptor
EMTepithelial–mesenchymal transition
ERendoplasmic reticulum
ESCCesophageal squamous cell carcinoma
FAsfatty acids
FAOfatty acid oxidation
FASNfatty acid synthase
FDPSfarnesyldiphosphate synthase
FITfat-storage-inducing transmembrane
GBMglioblastoma
GLSglutaminase
Glutsglucose transporters
HBPhexosamine biosynthesis pathway
HCChepatocellular carcinoma
HMGCShydroxymethylglutaryl-CoA synthase
HMGCRhydroxymethylglutaryl-CoA reductase
HILPDAhypoxia-inducible lipid droplet associated
Insiginsulin-inducible gene protein
IDHisocitrate dehydrogenase
LDslipid droplets
LDLlow-density lipoprotein
LDLRLDL receptor
IDOLinducible degrader of LDLR
LPClysophosphatidylcholine
LPCATslysophosphatidylcholine acyltransferases
LXRliver X receptor
MBOATmembrane-bound O-acyltransferase
MCTsmonocarboxylate transporters
NASHnon-alcoholic steatohepatitis
NLSDneutral lipid storage disease
OAAoxaloacetate
PD1programmed cell death protein 1
PDHpyruvate dehydrogenase
PD-L1programmed death-ligand 1
PI3Kphosphoinositide 3-kinases
PLsphospholipids
PLAphospholipase A
ROSreactive oxygen species
S1Psite 1 protease
S2Psite 2 protease
SCAPSREBP-cleavage activating protein
SCD1stearoyl-CoA desaturase 1
SLC25A1tricarboxylate transport protein, mitochondrial
SOAT1sterol-O-acyltransferase 1
SQLEsqualene monooxygenase
SREsterol regulatory element
SREBF1sterol regulatory element binding transcription factor 1
SREBPsterol regulatory element-binding protein
TAGtriacylglycerol
TCA cycletricarboxylic acid cycle
TCGAthe cancer genome atlas
TKItyrosine kinase inhibitors
TMZtemozolomide
VEGFvascular endothelial growth factor

References

  1. Cheng, C.; Geng, F.; Cheng, X.; Guo, D. Lipid metabolism reprogramming and its potential targets in cancer. Cancer Commun. 2018, 38, 27. [Google Scholar] [CrossRef]
  2. Cockcroft, S. Mammalian lipids: Structure, synthesis and function. Essays Biochem. 2021, 65, 813–845. [Google Scholar] [CrossRef]
  3. Cisa-Wieczorek, S.; Hernandez-Alvarez, M.I. Deregulation of Lipid Homeostasis: A Fa(c)t in the Development of Metabolic Diseases. Cells 2020, 9, 2605. [Google Scholar] [CrossRef]
  4. Onal, G.; Kutlu, O.; Gozuacik, D.; Dokmeci Emre, S. Lipid Droplets in Health and Disease. Lipids Health Dis. 2017, 16, 128. [Google Scholar] [CrossRef] [Green Version]
  5. Estes, R.E.; Lin, B.; Khera, A.; Davis, M.Y. Lipid Metabolism Influence on Neurodegenerative Disease Progression: Is the Vehicle as Important as the Cargo? Front. Mol. Neurosci. 2021, 14, 788695. [Google Scholar] [CrossRef] [PubMed]
  6. Teixeira, V.; Maciel, P.; Costa, V. Leading the way in the nervous system: Lipid Droplets as new players in health and disease. Biochim. Biophys. Acta Mol. Cell Biol. Lipids 2021, 1866, 158820. [Google Scholar] [CrossRef] [PubMed]
  7. Hanahan, D.; Weinberg, R.A. Hallmarks of cancer: The next generation. Cell 2011, 144, 646–674. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  8. Snaebjornsson, M.T.; Janaki-Raman, S.; Schulze, A. Greasing the Wheels of the Cancer Machine: The Role of Lipid Metabolism in Cancer. Cell Metab. 2020, 31, 62–76. [Google Scholar] [CrossRef]
  9. Broadfield, L.A.; Pane, A.A.; Talebi, A.; Swinnen, J.V.; Fendt, S.M. Lipid metabolism in cancer: New perspectives and emerging mechanisms. Dev. Cell 2021, 56, 1363–1393. [Google Scholar] [CrossRef] [PubMed]
  10. Bian, X.; Liu, R.; Meng, Y.; Xing, D.; Xu, D.; Lu, Z. Lipid metabolism and cancer. J. Exp. Med. 2021, 218, e20201606. [Google Scholar] [CrossRef]
  11. Cloughesy, T.F.; Cavenee, W.K.; Mischel, P.S. Glioblastoma: From molecular pathology to targeted treatment. Annu. Rev. Pathol. 2014, 9, 1–25. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  12. Cruz Da Silva, E.; Mercier, M.C.; Etienne-Selloum, N.; Dontenwill, M.; Choulier, L. A Systematic Review of Glioblastoma-Targeted Therapies in Phases II, III, IV Clinical Trials. Cancers 2021, 13, 1795. [Google Scholar] [CrossRef] [PubMed]
  13. Guo, D.; Hildebrandt, I.J.; Prins, R.M.; Soto, H.; Mazzotta, M.M.; Dang, J.; Czernin, J.; Shyy, J.Y.; Watson, A.D.; Phelps, M.; et al. The AMPK agonist AICAR inhibits the growth of EGFRvIII-expressing glioblastomas by inhibiting lipogenesis. Proc. Natl. Acad. Sci. USA 2009, 106, 12932–12937. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  14. Guo, D.; Prins, R.M.; Dang, J.; Kuga, D.; Iwanami, A.; Soto, H.; Lin, K.Y.; Huang, T.T.; Akhavan, D.; Hock, M.B.; et al. EGFR signaling through an Akt-SREBP-1-dependent, rapamycin-resistant pathway sensitizes glioblastomas to antilipogenic therapy. Sci. Signal. 2009, 2, ra82. [Google Scholar] [CrossRef] [Green Version]
  15. Guo, D.; Reinitz, F.; Youssef, M.; Hong, C.; Nathanson, D.; Akhavan, D.; Kuga, D.; Amzajerdi, A.N.; Soto, H.; Zhu, S.; et al. An LXR agonist promotes glioblastoma cell death through inhibition of an EGFR/AKT/SREBP-1/LDLR-dependent pathway. Cancer Discov. 2011, 1, 442–456. [Google Scholar] [CrossRef] [Green Version]
  16. Guo, D.; Cloughesy, T.F.; Radu, C.G.; Mischel, P.S. AMPK: A metabolic checkpoint that regulates the growth of EGFR activated glioblastomas. Cell Cycle 2010, 9, 211–212. [Google Scholar] [CrossRef] [Green Version]
  17. Cheng, X.; Geng, F.; Pan, M.; Wu, X.; Zhong, Y.; Wang, C.; Tian, Z.; Cheng, C.; Zhang, R.; Puduvalli, V.; et al. Targeting DGAT1 Ameliorates Glioblastoma by Increasing Fat Catabolism and Oxidative Stress. Cell Metab. 2020, 32, 229–242.e8. [Google Scholar] [CrossRef]
  18. Rudalska, R.; Harbig, J.; Snaebjornsson, M.T.; Klotz, S.; Zwirner, S.; Taranets, L.; Heinzmann, F.; Kronenberger, T.; Forster, M.; Cui, W.; et al. LXRalpha activation and Raf inhibition trigger lethal lipotoxicity in liver cancer. Nat. Cancer 2021, 2, 201–217. [Google Scholar] [CrossRef]
  19. Geng, F.; Cheng, X.; Wu, X.; Yoo, J.Y.; Cheng, C.; Guo, J.Y.; Mo, X.; Ru, P.; Hurwitz, B.; Kim, S.H.; et al. Inhibition of SOAT1 Suppresses Glioblastoma Growth via Blocking SREBP-1-Mediated Lipogenesis. Clin. Cancer Res. Off. J. Am. Assoc. Cancer Res. 2016, 22, 5337–5348. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  20. Geng, F.; Guo, D. Lipid droplets, potential biomarker and metabolic target in glioblastoma. Intern. Med. Rev. 2017, 3. [Google Scholar] [CrossRef] [Green Version]
  21. Wu, X.; Geng, F.; Cheng, X.; Guo, Q.; Zhong, Y.; Cloughesy, T.F.; Yong, W.H.; Chakravarti, A.; Guo, D. Lipid Droplets Maintain Energy Homeostasis and Glioblastoma Growth via Autophagic Release of Stored Fatty Acids. iScience 2020, 23, 101569. [Google Scholar] [CrossRef] [PubMed]
  22. Cheng, X.; Geng, F.; Guo, D. DGAT1 protects tumor from lipotoxicity, emerging as a promising metabolic target for cancer therapy. Mol. Cell. Oncol. 2020, 7, 1805257. [Google Scholar] [CrossRef] [PubMed]
  23. Stupp, R.; Mason, W.P.; van den Bent, M.J.; Weller, M.; Fisher, B.; Taphoorn, M.J.; Belanger, K.; Brandes, A.A.; Marosi, C.; Bogdahn, U.; et al. Radiotherapy plus concomitant and adjuvant temozolomide for glioblastoma. N. Engl. J. Med. 2005, 352, 987–996. [Google Scholar] [CrossRef]
  24. Rock, K.; McArdle, O.; Forde, P.; Dunne, M.; Fitzpatrick, D.; O’Neill, B.; Faul, C. A clinical review of treatment outcomes in glioblastoma multiforme--the validation in a non-trial population of the results of a randomised Phase III clinical trial: Has a more radical approach improved survival? Br. J. Radiol. 2012, 85, e729–e733. [Google Scholar] [CrossRef]
  25. Janjua, T.I.; Rewatkar, P.; Ahmed-Cox, A.; Saeed, I.; Mansfeld, F.M.; Kulshreshtha, R.; Kumeria, T.; Ziegler, D.S.; Kavallaris, M.; Mazzieri, R.; et al. Frontiers in the treatment of glioblastoma: Past, present and emerging. Adv. Drug Deliv. Rev. 2021, 171, 108–138. [Google Scholar] [CrossRef] [PubMed]
  26. Ohgaki, H.; Kleihues, P. The definition of primary and secondary glioblastoma. Clin. Cancer Res. Off. J. Am. Assoc. Cancer Res. 2013, 19, 764–772. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  27. Li, Z.H.; Guan, Y.L.; Liu, Q.; Wang, Y.; Cui, R.; Wang, Y.J. Astrocytoma progression scoring system based on the WHO 2016 criteria. Sci. Rep. 2019, 9, 96. [Google Scholar] [CrossRef]
  28. Weller, M.; van den Bent, M.; Tonn, J.C.; Stupp, R.; Preusser, M.; Cohen-Jonathan-Moyal, E.; Henriksson, R.; Le Rhun, E.; Balana, C.; Chinot, O.; et al. European Association for Neuro-Oncology (EANO) guideline on the diagnosis and treatment of adult astrocytic and oligodendroglial gliomas. Lancet Oncol. 2017, 18, e315–e329. [Google Scholar] [CrossRef] [Green Version]
  29. Ringel, F.; Pape, H.; Sabel, M.; Krex, D.; Bock, H.C.; Misch, M.; Weyerbrock, A.; Westermaier, T.; Senft, C.; Schucht, P.; et al. Clinical benefit from resection of recurrent glioblastomas: Results of a multicenter study including 503 patients with recurrent glioblastomas undergoing surgical resection. Neuro-Oncology 2016, 18, 96–104. [Google Scholar] [CrossRef] [Green Version]
  30. Ostrom, Q.T.; Cioffi, G.; Waite, K.; Kruchko, C.; Barnholtz-Sloan, J.S. CBTRUS Statistical Report: Primary Brain and Other Central Nervous System Tumors Diagnosed in the United States in 2014–2018. Neuro-Oncology 2021, 23 (Suppl. 2), iii1–iii105. [Google Scholar] [CrossRef]
  31. Friedman, H.S.; Prados, M.D.; Wen, P.Y.; Mikkelsen, T.; Schiff, D.; Abrey, L.E.; Yung, W.K.; Paleologos, N.; Nicholas, M.K.; Jensen, R.; et al. Bevacizumab alone and in combination with irinotecan in re-current glioblastoma. J. Clin. Oncol. 2009, 27, 4733–4740. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  32. Gilbert, M.R.; Dignam, J.J.; Armstrong, T.S.; Wefel, J.S.; Blumenthal, D.T.; Vogelbaum, M.A.; Colman, H.; Chakravarti, A.; Pugh, S.; Won, M.; et al. A randomized trial of bevacizumab for newly diagnosed glioblastoma. N. Engl. J. Med. 2014, 370, 699–708. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  33. Wu, W.; Klockow, J.L.; Zhang, M.; Lafortune, F.; Chang, E.; Jin, L.; Wu, Y.; Daldrup-Link, H.E. Glioblastoma multiforme (GBM): An overview of current therapies and mechanisms of resistance. Pharmacol. Res. 2021, 171, 105780. [Google Scholar] [CrossRef] [PubMed]
  34. Brennan, C.W.; Verhaak, R.G.; McKenna, A.; Campos, B.; Noushmehr, H.; Salama, S.R.; Zheng, S.; Chakravarty, D.; Sanborn, J.Z.; Berman, S.H.; et al. The somatic genomic landscape of glioblastoma. Cell 2013, 155, 462–477. [Google Scholar] [CrossRef]
  35. Mellinghoff, I.K.; Schultz, N.; Mischel, P.S.; Cloughesy, T.F. Will kinase inhibitors make it as glioblastoma drugs? Curr. Top. Microbiol. Immunol. 2012, 355, 135–169. [Google Scholar]
  36. Nduom, E.K.; Wei, J.; Yaghi, N.K.; Huang, N.; Kong, L.Y.; Gabrusiewicz, K.; Ling, X.; Zhou, S.; Ivan, C.; Chen, J.Q.; et al. PD-L1 expression and prognostic impact in glioblastoma. Neuro-Oncology 2016, 18, 195–205. [Google Scholar] [CrossRef] [Green Version]
  37. Berghoff, A.S.; Kiesel, B.; Widhalm, G.; Rajky, O.; Ricken, G.; Wöhrer, A.; Dieckmann, K.; Filipits, M.; Brandstetter, A.; Weller, M.; et al. Programmed death ligand 1 expression and tumor-infiltrating lymphocytes in glioblastoma. Neuro-Oncology 2015, 17, 1064–1075. [Google Scholar] [CrossRef] [Green Version]
  38. Muir, M.; Gopakumar, S.; Traylor, J.; Lee, S.; Rao, G. Glioblastoma multiforme: Novel therapeutic targets. Expert Opin. Targets 2020, 24, 605–614. [Google Scholar] [CrossRef]
  39. Guo, D.; Bell, E.H.; Mischel, P.; Chakravarti, A. Targeting SREBP-1-driven lipid metabolism to treat cancer. Curr. Pharm. Des. 2014, 20, 2619–2626. [Google Scholar] [CrossRef] [Green Version]
  40. Bhagavan, N.V.; Ha, C.-E. Chapter 16—Lipids I: Fatty Acids and Eicosanoids. In Essentials of Medical Biochemistry, 2nd ed.; Bhagavan, N.V., Ha, C.-E., Eds.; Academic Press: San Diego, CA, USA, 2015; pp. 269–297. [Google Scholar]
  41. Mashimo, T.; Pichumani, K.; Vemireddy, V.; Hatanpaa Kimmo, J.; Singh Dinesh, K.; Sirasanagandla, S.; Nannepaga, S.; Piccirillo Sara, G.; Kovacs, Z.; Foong, C.; et al. Acetate Is a Bioenergetic Substrate for Human Glioblastoma and Brain Metastases. Cell 2014, 159, 1603–1614. [Google Scholar] [CrossRef] [Green Version]
  42. Liu, M.; Liu, N.; Wang, J.; Fu, S.; Wang, X.; Chen, D. Acetyl-CoA Synthetase 2 as a Therapeutic Target in Tumor Metabolism. Cancers 2022, 14, 2896. [Google Scholar] [CrossRef] [PubMed]
  43. Etchegaray, J.P.; Mostoslavsky, R. Interplay between Metabolism and Epigenetics: A Nuclear Adaptation to Environmental Changes. Mol. Cell 2016, 62, 695–711. [Google Scholar] [CrossRef] [Green Version]
  44. Rohrig, F.; Schulze, A. The multifaceted roles of fatty acid synthesis in cancer. Nat. Rev. Cancer 2016, 16, 732–749. [Google Scholar] [CrossRef] [PubMed]
  45. Shen, H.; Shi, L.Z. Metabolic regulation of TH17 cells. Mol. Immunol. 2019, 109, 81–87. [Google Scholar] [CrossRef] [PubMed]
  46. Cheng, C.; Ru, P.; Geng, F.; Liu, J.; Yoo, J.Y.; Wu, X.; Cheng, X.; Euthine, V.; Hu, P.; Guo, J.Y.; et al. Glucose-Mediated N-glycosylation of SCAP Is Essential for SREBP-1 Activation and Tumor Growth. Cancer Cell 2015, 28, 569–581. [Google Scholar] [CrossRef] [Green Version]
  47. Horton, J.D. Sterol regulatory element-binding proteins: Transcriptional activators of lipid synthesis. Biochem. Soc. Trans. 2002, 30 Pt 6, 1091–1095. [Google Scholar] [CrossRef] [Green Version]
  48. Melone, M.A.B.; Valentino, A.; Margarucci, S.; Galderisi, U.; Giordano, A.; Peluso, G. The carnitine system and cancer metabolic plasticity. Cell Death Dis. 2018, 9, 228. [Google Scholar] [CrossRef] [Green Version]
  49. Schlaepfer, I.R.; Joshi, M. CPT1A-mediated Fat Oxidation, Mechanisms, and Therapeutic Potential. Endocrinology 2020, 161, bqz046. [Google Scholar] [CrossRef]
  50. Martinez-Reyes, I.; Chandel, N.S. Cancer metabolism: Looking forward. Nat. Rev. Cancer 2021, 21, 669–680. [Google Scholar] [CrossRef]
  51. Zhou, W.; Wahl, D.R. Metabolic Abnormalities in Glioblastoma and Metabolic Strategies to Overcome Treatment Resistance. Cancers 2019, 11, 1231. [Google Scholar] [CrossRef] [Green Version]
  52. Williams, K.J.; Argus, J.P.; Zhu, Y.; Wilks, M.Q.; Marbois, B.N.; York, A.G.; Kidani, Y.; Pourzia, A.L.; Akhavan, D.; Lisiero, D.N.; et al. An essential requirement for the SCAP/SREBP signaling axis to protect cancer cells from lipotoxicity. Cancer Res. 2013, 73, 2850–2862. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  53. Ru, P.; Hu, P.; Geng, F.; Mo, X.; Cheng, C.; Yoo, J.Y.; Cheng, X.; Wu, X.; Guo, J.Y.; Nakano, I.; et al. Feedback Loop Regulation of SCAP/SREBP-1 by miR-29 Modulates EGFR Signaling-Driven Glioblastoma Growth. Cell Rep. 2016, 16, 1527–1535. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  54. Ru, P.; Guo, D. microRNA-29 mediates a novel negative feedback loop to regulate SCAP/SREBP-1 and lipid metabolism. RNA Dis. 2017, 4, e1525. [Google Scholar] [PubMed] [Green Version]
  55. Jin, X.; Demere, Z.; Nair, K.; Ali, A.; Ferraro, G.B.; Natoli, T.; Deik, A.; Petronio, L.; Tang, A.A.; Zhu, C.; et al. A metastasis map of human cancer cell lines. Nature 2020, 588, 331–336. [Google Scholar] [CrossRef]
  56. Venneti, S.; Dunphy, M.P.; Zhang, H.; Pitter, K.L.; Zanzonico, P.; Campos, C.; Carlin, S.D.; La Rocca, G.; Lyashchenko, S.; Ploessl, K.; et al. Glutamine-based PET imaging facilitates enhanced metabolic evaluation of gliomas in vivo. Sci. Transl. Med. 2015, 7, 274ra217. [Google Scholar] [CrossRef] [Green Version]
  57. Cheng, C.; Geng, F.; Li, Z.; Zhong, Y.; Wang, H.; Cheng, X.; Zhao, Y.; Mo, X.; Horbinski, C.; Duan, W.; et al. Ammonia stimulates SCAP/Insig dissociation and SREBP-1 activation to promote lipogenesis and tumour growth. Nat. Metab. 2022, 4, 575–588. [Google Scholar] [CrossRef]
  58. Goldstein, J.L.; DeBose-Boyd, R.A.; Brown, M.S. Protein sensors for membrane sterols. Cell 2006, 124, 35–46. [Google Scholar] [CrossRef] [Green Version]
  59. Goldstein, J.L.; Brown, M.S. A century of cholesterol and coronaries: From plaques to genes to statins. Cell 2015, 161, 161–172. [Google Scholar] [CrossRef] [Green Version]
  60. Jeon, T.I.; Osborne, T.F. SREBPs: Metabolic integrators in physiology and metabolism. Trends Endocrinol. Metab. 2012, 23, 65–72. [Google Scholar] [CrossRef] [Green Version]
  61. Horton, J.D.; Goldstein, J.L.; Brown, M.S. SREBPs: Activators of the complete program of cholesterol and fatty acid synthesis in the liver. J. Clin. Investig. 2002, 109, 1125–1131. [Google Scholar] [CrossRef]
  62. Brown, M.S.; Radhakrishnan, A.; Goldstein, J.L. Retrospective on Cholesterol Homeostasis: The Central Role of Scap. Annu. Rev. Biochem. 2018, 87, 783–807. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  63. Bennett, M.K.; Lopez, J.M.; Sanchez, H.B.; Osborne, T.F. Sterol regulation of fatty acid synthase promoter. Coordinate feedback regulation of two major lipid pathways. J. Biol. Chem. 1995, 270, 25578–25583. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  64. Sun, L.P.; Seemann, J.; Goldstein, J.L.; Brown, M.S. Sterol-regulated transport of SREBPs from endoplasmic reticulum to Golgi: Insig renders sorting signal in Scap inaccessible to COPII proteins. Proc. Natl Acad. Sci. USA 2007, 104, 6519–6526. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  65. Espenshade, P.J.; Cheng, D.; Goldstein, J.L.; Brown, M.S. Autocatalytic processing of site-1 protease removes propeptide and permits cleavage of sterol regulatory element-binding proteins. J. Biol. Chem. 1999, 274, 22795–22804. [Google Scholar] [CrossRef] [Green Version]
  66. Cheng, D.; Espenshade, P.J.; Slaughter, C.A.; Jaen, J.C.; Brown, M.S.; Goldstein, J.L. Secreted site-1 protease cleaves peptides corresponding to luminal loop of sterol regulatory element-binding proteins. J. Biol. Chem. 1999, 274, 22805–22812. [Google Scholar] [CrossRef] [Green Version]
  67. Rawson, R.B.; Zelenski, N.G.; Nijhawan, D.; Ye, J.; Sakai, J.; Hasan, M.T.; Chang, T.Y.; Brown, M.S.; Goldstein, J.L. Complementation cloning of S2P, a gene encoding a putative metalloprotease required for intramembrane cleavage of SREBPs. Mol. Cell 1997, 1, 47–57. [Google Scholar] [CrossRef]
  68. Duncan, E.A.; Dave, U.P.; Sakai, J.; Goldstein, J.L.; Brown, M.S. Second-site cleavage in sterol regulatory element-binding protein occurs at transmembrane junction as determined by cysteine panning. J. Biol. Chem. 1998, 273, 17801–17809. [Google Scholar] [CrossRef] [Green Version]
  69. Hua, X.; Sakai, J.; Brown, M.S.; Goldstein, J.L. Regulated cleavage of sterol regulatory element binding proteins requires sequences on both sides of the endoplasmic reticulum membrane. J. Biol. Chem. 1996, 271, 10379–10384. [Google Scholar] [CrossRef] [Green Version]
  70. Yang, T.; Espenshade, P.J.; Wright, M.E.; Yabe, D.; Gong, Y.; Aebersold, R.; Goldstein, J.L.; Brown, M.S. Crucial step in cholesterol homeostasis: Sterols promote binding of SCAP to INSIG-1, a membrane protein that facilitates retention of SREBPs in ER. Cell 2002, 110, 489–500. [Google Scholar] [CrossRef] [Green Version]
  71. Yabe, D.; Brown, M.S.; Goldstein, J.L. Insig-2, a second endoplasmic reticulum protein that binds SCAP and blocks export of sterol regulatory element-binding proteins. Proc. Natl. Acad. Sci. USA 2002, 99, 12753–12758. [Google Scholar] [CrossRef] [Green Version]
  72. Espenshade, P.J.; Li, W.P.; Yabe, D. Sterols block binding of COPII proteins to SCAP, thereby controlling SCAP sorting in ER. Proc. Natl. Acad. Sci. USA 2002, 99, 11694–11699. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  73. Adams, C.M.; Goldstein, J.L.; Brown, M.S. Cholesterol-induced conformational change in SCAP enhanced by Insig proteins and mimicked by cationic amphiphiles. Proc. Natl. Acad. Sci. USA 2003, 100, 10647–10652. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  74. Guo, D. SCAP links glucose to lipid metabolism in cancer cells. Mol. Cell. Oncol. 2016, 3, e1132120. [Google Scholar] [CrossRef] [Green Version]
  75. Shao, W.; Espenshade, P.J. Sugar Makes Fat by Talking to SCAP. Cancer Cell 2015, 28, 548–549. [Google Scholar] [CrossRef] [Green Version]
  76. Cheng, C.; Guo, J.Y.; Geng, F.; Wu, X.; Cheng, X.; Li, Q.; Guo, D. Analysis of SCAP N-glycosylation and Trafficking in Human Cells. J. Vis. Exp. 2016, 117, e54709. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  77. Yang, Y.; Hu, B. 21-Bio-based chemicals from biorefining: Lipid and wax conversion and utilization. In Advances in Biorefineries; Waldron, K., Ed.; Woodhead Publishing: Thorston, UK, 2014; pp. 693–720. [Google Scholar]
  78. Currie, E.; Schulze, A.; Zechner, R.; Walther Tobias, C.; Farese Robert, V. Cellular Fatty Acid Metabolism and Cancer. Cell Metab. 2013, 18, 153–161. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  79. Hoy, A.J.; Nagarajan, S.R.; Butler, L.M. Tumour fatty acid metabolism in the context of therapy resistance and obesity. Nat. Rev. Cancer 2021, 21, 753–766. [Google Scholar] [CrossRef]
  80. Pérez-Chacón, G.; Astudillo, A.M.; Balgoma, D.; Balboa, M.A.; Balsinde, J. Control of free arachidonic acid levels by phospholipases A2 and lysophospholipid acyltransferases. Biochim. Bi-Ophysica Acta (BBA)-Mol. Cell Biol. Lipids 2009, 1791, 1103–1113. [Google Scholar] [CrossRef] [Green Version]
  81. Cifarelli, V.; Abumrad, N.A. Chapter 48—Enterocyte Fatty Acid Handling Proteins and Chylomicron Formation. In Physiology of the Gastrointestinal Tract, 6th ed.; Said, H.M., Ed.; Academic Press: Cambridge, MA, USA, 2018; pp. 1087–1107. [Google Scholar]
  82. Wang, B.; Tontonoz, P. Phospholipid Remodeling in Physiology and Disease. Annu. Rev. Physiol. 2019, 81, 165–188. [Google Scholar] [CrossRef]
  83. Saito, R.F.; Rangel, M.C.; Halman, J.R.; Chandler, M.; de Sousa Andrade, L.N.; Odete-Bustos, S.; Furuya, T.K.; Carrasco, A.G.M.; Chaves-Filho, A.B.; Yoshinaga, M.Y.; et al. Simultaneous silencing of lyso-phosphatidylcholine acyltransferases 1–4 by nucleic acid nanoparticles (NANPs) improves radiation response of melanoma cells. Nanomed. Nanotechnol. Biol. Med. 2021, 36, 102418. [Google Scholar] [CrossRef]
  84. Shi, C.; Qiao, S.; Wang, S.; Wu, T.; Ji, G.J.I.J.C.E.M. Recent progress of lysophosphatidylcholine acyltransferases in metabolic disease and cancer. Int. J. Clin. Exp. Med. 2018, 11, 8941–8953. [Google Scholar]
  85. Bi, J.; Ichu, T.-A.; Zanca, C.; Yang, H.; Zhang, W.; Gu, Y.; Chowdhry, S.; Reed, A.; Ikegami, S.; Turner, K.M.; et al. Oncogene Amplification in Growth Factor Signaling Pathways Renders Cancers De-pendent on Membrane Lipid Remodeling. Cell Metab. 2019, 30, 525–538.e8. [Google Scholar] [CrossRef] [PubMed]
  86. He, R.-Q.; Li, J.-D.; Du, X.-F.; Dang, Y.-W.; Yang, L.-J.; Huang, Z.-G.; Liu, L.-M.; Liao, L.-F.; Yang, H.; Chen, G. LPCAT1 overexpression promotes the progression of hepatocellular carcinoma. Cancer Cell Int. 2021, 21, 442. [Google Scholar] [CrossRef] [PubMed]
  87. Du, Y.; Wang, Q.; Zhang, X.; Wang, X.; Qin, C.; Sheng, Z.; Yin, H.; Jiang, C.; Li, J.; Xu, T. Lysophosphatidylcholine acyltransferase 1 upregulation and concomitant phospholipid alterations in clear cell renal cell carcinoma. J. Exp. Clin. Cancer Res. CR 2017, 36, 66. [Google Scholar] [CrossRef] [Green Version]
  88. Tao, M.; Luo, J.; Gu, T.; Yu, X.; Song, Z.; Jun, Y.; Gu, H.; Han, K.; Huang, X.; Yu, W.; et al. LPCAT1 reprogramming cholesterol metabolism promotes the progression of esophageal squamous cell car-cinoma. Cell Death Dis. 2021, 12, 845. [Google Scholar] [CrossRef]
  89. Uehara, T.; Kikuchi, H.; Miyazaki, S.; Iino, I.; Setoguchi, T.; Hiramatsu, Y.; Ohta, M.; Kamiya, K.; Morita, Y.; Tanaka, H.; et al. Overexpression of Lysophosphatidylcholine Acyltransferase 1 and Con-comitant Lipid Alterations in Gastric Cancer. Ann. Surg. Oncol. 2016, 23 (Suppl. 2), S206–S213. [Google Scholar] [CrossRef] [Green Version]
  90. Lebok, P.; von Hassel, A.; Meiners, J.; Hube-Magg, C.; Simon, R.; Höflmayer, D.; Hinsch, A.; Dum, D.; Fraune, C.; Göbel, C.; et al. Up-regulation of lysophosphatidylcholine acyltransferase 1 (LPCAT1) is linked to poor prognosis in breast cancer. Aging 2019, 11, 7796–7804. [Google Scholar] [CrossRef]
  91. Li, D.; Li, Y. The interaction between ferroptosis and lipid metabolism in cancer. Signal Transduct. Target. Ther. 2020, 5, 108. [Google Scholar] [CrossRef]
  92. Mbah, N.E.; Lyssiotis, C.A. Metabolic regulation of ferroptosis in the tumor microenvironment. J. Biol. Chem. 2022, 298, 101617. [Google Scholar] [CrossRef]
  93. Lei, P.; Bai, T.; Sun, Y. Mechanisms of Ferroptosis and Relations with Regulated Cell Death: A Review. Front. Physiol. 2019, 10, 139. [Google Scholar] [CrossRef] [Green Version]
  94. Kagan, V.E.; Mao, G.; Qu, F.; Angeli, J.P.; Doll, S.; Croix, C.S.; Dar, H.H.; Liu, B.; Tyurin, V.A.; Ritov, V.B.; et al. Oxidized arachidonic and adrenic PEs navigate cells to ferroptosis. Nat. Chem. Biol. 2017, 13, 81–90. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  95. Yeagle, P. L: Modulation of membrane function by cholesterol. Biochimie 1991, 73, 1303–1310. [Google Scholar] [CrossRef]
  96. Luo, J.; Yang, H.; Song, B.L. Mechanisms and regulation of cholesterol homeostasis. Nat. Rev. Mol. Cell Biol. 2020, 21, 225–245. [Google Scholar] [CrossRef] [PubMed]
  97. Holthuis, J.C.; Menon, A.K. Lipid landscapes and pipelines in membrane homeostasis. Nature 2014, 510, 48–57. [Google Scholar] [CrossRef]
  98. Ikonen, E. Cellular cholesterol trafficking and compartmentalization. Nat. Rev. Mol. Cell Biol. 2008, 9, 125–138. [Google Scholar] [CrossRef]
  99. van Meer, G.; Voelker, D.R.; Feigenson, G.W. Membrane lipids: Where they are and how they behave. Nat. Rev. Mol. Cell Biol. 2008, 9, 112–124. [Google Scholar] [CrossRef]
  100. Iaea, D.B.; Maxfield, F.R. Cholesterol trafficking and distribution. Essays Biochem. 2015, 57, 43–55. [Google Scholar]
  101. Holmes, M.V.; Ala-Korpela, M. What is ‘LDL cholesterol’? Nat. Rev. Cardiol. 2019, 16, 197–198. [Google Scholar] [CrossRef]
  102. Maxfield, F.R.; van Meer, G. Cholesterol, the central lipid of mammalian cells. Curr. Opin. Cell Biol. 2010, 22, 422–429. [Google Scholar] [CrossRef] [Green Version]
  103. Segrest, J.P.; Jones, M.K.; De Loof, H.; Dashti, N. Structure of apolipoprotein B-100 in low density lipoproteins. J. Lipid Res. 2001, 42, 1346–1367. [Google Scholar] [CrossRef]
  104. Goldstein, J.L.; Brown, M.S. The LDL receptor. Arterioscler. Thromb. Vasc. Biol. 2009, 29, 431–438. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  105. Goldstein, J.L.; Anderson, R.G.; Brown, M.S. Receptor-mediated endocytosis and the cellular uptake of low density lipoprotein. Ciba Found. Symp. 1982, 92, 77–95. [Google Scholar]
  106. Goldstein, J.L.; Basu, S.K.; Brown, M.S. Receptor-mediated endocytosis of low-density lipoprotein in cultured cells. Methods Enzymol. 1983, 98, 241–260. [Google Scholar] [PubMed]
  107. Ertunc, M.E.; Hotamisligil, G.S. Lipid signaling and lipotoxicity in metaflammation: Indications for metabolic disease pathogenesis and treatment. J. Lipid Res. 2016, 57, 2099–2114. [Google Scholar] [CrossRef] [Green Version]
  108. Lipke, K.; Kubis-Kubiak, A.; Piwowar, A. Molecular Mechanism of Lipotoxicity as an Interesting Aspect in the Development of Pathological States—Current View of Knowledge. Cells 2022, 11, 844. [Google Scholar] [CrossRef]
  109. Olzmann, J.A.; Carvalho, P. Dynamics and functions of lipid droplets. Nat. Rev. Mol. Cell Biol. 2019, 20, 137–155. [Google Scholar] [CrossRef]
  110. Cohen, S. Lipid Droplets as Organelles. Int. Rev. Cell Mol. Biol. 2018, 337, 83–110. [Google Scholar]
  111. Cruz, A.L.S.; Barreto, E.A.; Fazolini, N.P.B.; Viola, J.P.B.; Bozza, P.T. Lipid droplets: Platforms with multiple functions in cancer hallmarks. Cell Death Dis. 2020, 11, 105. [Google Scholar] [CrossRef] [Green Version]
  112. Roberts, M.A.; Olzmann, J.A. Protein Quality Control and Lipid Droplet Metabolism. Annu. Rev. Cell Dev. Biol. 2020, 36, 115–139. [Google Scholar] [CrossRef]
  113. Lundquist, P.K.; Shivaiah, K.K.; Espinoza-Corral, R. Lipid droplets throughout the evolutionary tree. Prog. Lipid Res. 2020, 78, 101029. [Google Scholar] [CrossRef]
  114. Coleman, R.A. The “discovery” of lipid droplets: A brief history of organelles hidden in plain sight. Biochim. Biophys. Acta Mol. Cell Biol. Lipids 2020, 1865, 158762. [Google Scholar] [CrossRef] [PubMed]
  115. Shang, C.; Qiao, J.; Guo, H. The dynamic behavior of lipid droplets in the pre-metastatic niche. Cell Death Dis. 2020, 11, 990. [Google Scholar] [CrossRef] [PubMed]
  116. Thiam, A.R.; Forêt, L. The physics of lipid droplet nucleation, growth and budding. Biochim. Biophys. Acta (BBA)-Mol. Cell Biol. Lipids 2016, 1861 Pt A, 715–722. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  117. Ben M’barek, K.; Ajjaji, D.; Chorlay, A.; Vanni, S.; Forêt, L.; Thiam, A.R. ER Membrane Phospholipids and Surface Tension Control Cellular Lipid Droplet Formation. Dev. Cell 2017, 41, 591–604.e7. [Google Scholar] [CrossRef] [Green Version]
  118. Chorlay, A.; Thiam, A.R. An Asymmetry in Monolayer Tension Regulates Lipid Droplet Budding Direction. Biophys. J. 2018, 114, 631–640. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  119. Choudhary, V.; Ojha, N.; Golden, A.; Prinz, W.A. A conserved family of proteins facilitates nascent lipid droplet budding from the ER. J. Cell Biol. 2015, 211, 261–271. [Google Scholar] [CrossRef]
  120. Gross, D.A.; Zhan, C.; Silver, D.L. Direct binding of triglyceride to fat storage-inducing transmembrane proteins 1 and 2 is important for lipid droplet formation. Proc. Natl. Acad. Sci. USA 2011, 108, 19581–19586. [Google Scholar] [CrossRef] [Green Version]
  121. Pagac, M.; Cooper, D.E.; Qi, Y.; Lukmantara, I.E.; Mak, H.Y.; Wu, Z.; Tian, Y.; Liu, Z.; Lei, M.; Du, X.; et al. SEIPIN Regulates Lipid Droplet Expansion and Adipocyte Development by Modulating the Activity of Glycerol-3-phosphate Acyltransferase. Cell Rep. 2016, 17, 1546–1559. [Google Scholar] [CrossRef] [Green Version]
  122. Fei, W.; Shui, G.; Gaeta, B.; Du, X.; Kuerschner, L.; Li, P.; Brown, A.J.; Wenk, M.R.; Parton, R.G.; Yang, H. Fld1p, a functional homologue of human seipin, regulates the size of lipid droplets in yeast. J. Cell Biol. 2008, 180, 473–482. [Google Scholar] [CrossRef]
  123. Szymanski, K.M.; Binns, D.; Bartz, R.; Grishin, N.V.; Li, W.P.; Agarwal, A.K.; Garg, A.; Anderson, R.G.; Goodman, J.M. The lipodystrophy protein seipin is found at endoplasmic reticulum lipid droplet junctions and is important for droplet morphology. Proc. Natl. Acad. Sci. USA 2007, 104, 20890–20895. [Google Scholar] [CrossRef] [Green Version]
  124. Wang, H.; Becuwe, M.; Housden, B.E.; Chitraju, C.; Porras, A.J.; Graham, M.M.; Liu, X.N.; Thiam, A.R.; Savage, D.B.; Agarwal, A.K.; et al. Seipin is required for converting nascent to mature lipid droplets. eLife 2016, 5, e16582. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  125. Xu, D.; Li, Y.; Wu, L.; Li, Y.; Zhao, D.; Yu, J.; Huang, T.; Ferguson, C.; Parton, R.G.; Yang, H.; et al. Rab18 promotes lipid droplet (LD) growth by tethering the ER to LDs through SNARE and NRZ interactions. J. Cell Biol. 2018, 217, 975–995. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  126. Wu, L.; Xu, D.; Zhou, L.; Xie, B.; Yu, L.; Yang, H.; Huang, L.; Ye, J.; Deng, H.; Yuan, Y.A.; et al. Rab8a-AS160-MSS4 regulatory circuit controls lipid droplet fusion and growth. Dev. Cell 2014, 30, 378–393. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  127. Gong, J.; Sun, Z.; Wu, L.; Xu, W.; Schieber, N.; Xu, D.; Shui, G.; Yang, H.; Parton, R.G.; Li, P. Fsp27 promotes lipid droplet growth by lipid exchange and transfer at lipid droplet contact sites. J. Cell Biol. 2011, 195, 953–963. [Google Scholar] [CrossRef]
  128. Wilfling, F.; Wang, H.; Haas, J.T.; Krahmer, N.; Gould, T.J.; Uchida, A.; Cheng, J.X.; Graham, M.; Christiano, R.; Fröhlich, F.; et al. Triacylglycerol synthesis enzymes mediate lipid droplet growth by relocalizing from the ER to lipid droplets. Dev. Cell 2013, 24, 384–399. [Google Scholar] [CrossRef] [Green Version]
  129. Bermúdez, M.A.; Balboa, M.A.; Balsinde, J. Lipid Droplets, Phospholipase A2, Arachidonic Acid, and Atherosclerosis. Biomedicines 2021, 9, 1891. [Google Scholar] [CrossRef]
  130. Rinia, H.A.; Burger, K.N.J.; Bonn, M.; Müller, M. Quantitative label-free imaging of lipid composition and packing of individual cellular lipid droplets using multiplex CARS microscopy. Biophys. J. 2008, 95, 4908–4914. [Google Scholar] [CrossRef] [Green Version]
  131. Zhang, W.; Xu, L.; Zhu, L.; Liu, Y.; Yang, S.; Zhao, M. Lipid Droplets, the Central Hub Integrating Cell Metabolism and the Immune System. Front. Physiol. 2021, 12, 746749. [Google Scholar] [CrossRef]
  132. Suzuki, M.; Shinohara, Y.; Ohsaki, Y.; Fujimoto, T. Lipid droplets: Size matters. J. Electron. Microsc. 2011, 60 (Suppl. 1), S101–S116. [Google Scholar] [CrossRef]
  133. Yang, H.; Galea, A.; Sytnyk, V.; Crossley, M. Controlling the size of lipid droplets: Lipid and protein factors. Curr. Opin. Cell Biol. 2012, 24, 509–516. [Google Scholar] [CrossRef]
  134. Scorletti, E.; Carr, R.M. A new perspective on NAFLD: Focusing on lipid droplets. J. Hepatol. 2022, 76, 934–945. [Google Scholar] [CrossRef] [PubMed]
  135. Hsieh, K.; Lee, Y.K.; Londos, C.; Raaka, B.M.; Dalen, K.T.; Kimmel, A.R. Perilipin family members preferentially sequester to either triacylglycerol-specific or cholesteryl-ester-specific intracellular lipid storage droplets. J. Cell Sci. 2012, 125 Pt 17, 4067–4076. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  136. Suzuki, M.; Ohsaki, Y.; Tatematsu, T.; Shinohara, Y.; Maeda, T.; Cheng, J.; Fujimoto, T. Translation inhibitors induce formation of cholesterol ester-rich lipid droplets. PLoS ONE 2012, 7, e42379. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  137. Herker, E.; Vieyres, G.; Beller, M.; Krahmer, N.; Bohnert, M. Lipid Droplet Contact Sites in Health and Disease. Trends Cell Biol. 2021, 31, 345–358. [Google Scholar] [CrossRef]
  138. Prinz, W.A. Bridging the gap: Membrane contact sites in signaling, metabolism, and organelle dynamics. J. Cell Biol. 2014, 205, 759–769. [Google Scholar] [CrossRef] [Green Version]
  139. Prinz, W.A.; Toulmay, A.; Balla, T. The functional universe of membrane contact sites. Nat. Rev. Mol. Cell Biol. 2020, 21, 7–24. [Google Scholar] [CrossRef]
  140. Valm, A.M.; Cohen, S.; Legant, W.R.; Melunis, J.; Hershberg, U.; Wait, E.; Cohen, A.R.; Davidson, M.W.; Betzig, E.; Lippincott-Schwartz, J. Applying systems-level spectral imaging and analysis to reveal the organelle interactome. Nature 2017, 546, 162–167. [Google Scholar] [CrossRef]
  141. Shai, N.; Yifrach, E.; van Roermund, C.W.T.; Cohen, N.; Bibi, C.; Ijlst, L.; Cavellini, L.; Meurisse, J.; Schuster, R.; Zada, L.; et al. Systematic mapping of contact sites reveals tethers and a function for the peroxisome-mitochondria contact. Nat. Commun. 2018, 9, 1761. [Google Scholar] [CrossRef]
  142. Wang, L.; Wang, Y.; Liang, Y.; Li, J.; Liu, Y.; Zhang, J.; Zhang, A.; Fu, J.; Jiang, G. Specific accumulation of lipid droplets in hepatocyte nuclei of PFOA-exposed BALB/c mice. Sci. Rep. 2013, 3, 2174. [Google Scholar] [CrossRef] [Green Version]
  143. Uzbekov, R.; Roingeard, P. Nuclear lipid droplets identified by electron microscopy of serial sections. BMC Res. Notes 2013, 6, 386. [Google Scholar] [CrossRef] [Green Version]
  144. Layerenza, J.P.; Gonzalez, P.; Garcia de Bravo, M.M.; Polo, M.P.; Sisti, M.S.; Ves-Losada, A. Nuclear lipid droplets: A novel nuclear domain. Biochim. Biophys. Acta 2013, 1831, 327–340. [Google Scholar] [CrossRef] [PubMed]
  145. Barbosa, A.D.; Siniossoglou, S. New kid on the block: Lipid droplets in the nucleus. FEBS J. 2020, 287, 4838–4843. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  146. Romanauska, A.; Kohler, A. The Inner Nuclear Membrane Is a Metabolically Active Territory that Generates Nuclear Lipid Droplets. Cell 2018, 174, 700–715.e8. [Google Scholar] [CrossRef] [Green Version]
  147. Ohsaki, Y.; Kawai, T.; Yoshikawa, Y.; Cheng, J.; Jokitalo, E.; Fujimoto, T. PML isoform II plays a critical role in nuclear lipid droplet formation. J. Cell Biol. 2016, 212, 29–38. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  148. Soltysik, K.; Ohsaki, Y.; Tatematsu, T.; Cheng, J.; Fujimoto, T. Nuclear lipid droplets derive from a lipoprotein precursor and regulate phosphatidylcholine synthesis. Nat. Commun. 2019, 10, 473. [Google Scholar] [CrossRef] [PubMed]
  149. Zhang, S.; Wang, Y.; Cui, L.; Deng, Y.; Xu, S.; Yu, J.; Cichello, S.; Serrero, G.; Ying, Y.; Liu, P. Morphologically and Functionally Distinct Lipid Droplet Subpopulations. Sci. Rep. 2016, 6, 29539. [Google Scholar] [CrossRef] [Green Version]
  150. Bersuker, K.; Peterson, C.W.H.; To, M.; Sahl, S.J.; Savikhin, V.; Grossman, E.A.; Nomura, D.K.; Olzmann, J.A. A Proximity Labeling Strategy Provides Insights into the Composition and Dynamics of Lipid Droplet Proteomes. Dev. Cell 2018, 44, 97–112.e7. [Google Scholar] [CrossRef]
  151. Aboumrad, M.H.; Horn, R.C., Jr.; Fine, G. Lipid-secreting mammary carcinoma. Report of a case associated with Paget’s disease of the nipple. Cancer 1963, 16, 521–525. [Google Scholar] [CrossRef]
  152. Wright, D.H. Lipid content of malignant lymphomas. J. Clin. Pathol. 1968, 21, 643–649. [Google Scholar] [CrossRef] [Green Version]
  153. Qi, W.; Fitchev, P.S.; Cornwell, M.L.; Greenberg, J.; Cabe, M.; Weber, C.R.; Roy, H.K.; Crawford, S.E.; Savkovic, S.D. FOXO3 growth inhibition of colonic cells is dependent on intraepithelial lipid droplet density. J. Biol. Chem. 2013, 288, 16274–16281. [Google Scholar] [CrossRef] [Green Version]
  154. Fazolini, N.P.; Cruz, A.L.; Werneck, M.B.; Viola, J.P.; Maya-Monteiro, C.M.; Bozza, P.T. Leptin activation of mTOR pathway in intestinal epithelial cell triggers lipid droplet formation, cytokine production and increased cell proliferation. Cell Cycle 2015, 14, 2667–2676. [Google Scholar] [CrossRef] [PubMed]
  155. Penrose, H.; Heller, S.; Cable, C.; Makboul, R.; Chadalawada, G.; Chen, Y.; Crawford, S.E.; Savkovic, S.D. Epidermal growth factor receptor mediated proliferation depends on increased lipid droplet density regulated via a negative regulatory loop with FOXO3/Sirtuin6. Biochem. Biophys. Res. Commun. 2016, 469, 370–376. [Google Scholar] [CrossRef] [Green Version]
  156. Corbet, C.; Bastien, E.; Santiago de Jesus, J.P.; Dierge, E.; Martherus, R.; Vander Linden, C.; Doix, B.; Degavre, C.; Guilbaud, C.; Petit, L.; et al. TGFβ2-induced formation of lipid droplets supports acidosis-driven EMT and the metastatic spreading of cancer cells. Nat. Commun. 2020, 11, 454. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  157. Giudetti, A.M.; De Domenico, S.; Ragusa, A.; Lunetti, P.; Gaballo, A.; Franck, J.; Simeone, P.; Nicolardi, G.; De Nuccio, F.; Santino, A.; et al. A specific lipid metabolic profile is associated with the epithelial mesenchymal transition program. Biochim. Biophys. Acta Mol. Cell Biol. Lipids 2019, 1864, 344–357. [Google Scholar] [CrossRef] [PubMed]
  158. Tirinato, L.; Marafioti, M.G.; Pagliari, F.; Jansen, J.; Aversa, I.; Hanley, R.; Nistico, C.; Garcia-Calderon, D.; Genard, G.; Guerreiro, J.F.; et al. Lipid droplets and ferritin heavy chain: A devilish liaison in human cancer cell radioresistance. eLife 2021, 10, e72943. [Google Scholar] [CrossRef]
  159. Nistico, C.; Pagliari, F.; Chiarella, E.; Fernandes Guerreiro, J.; Marafioti, M.G.; Aversa, I.; Genard, G.; Hanley, R.; Garcia-Calderon, D.; Bond, H.M.; et al. Lipid Droplet Biosynthesis Impairment through DGAT2 Inhibition Sensitizes MCF7 Breast Cancer Cells to Radiation. Int. J. Mol. Sci. 2021, 22, 10102. [Google Scholar] [CrossRef]
  160. Lettiero, B.; Inasu, M.; Kimbung, S.; Borgquist, S. Insensitivity to atorvastatin is associated with increased accumulation of intracellular lipid droplets and fatty acid metabolism in breast cancer cells. Sci. Rep. 2018, 8, 5462. [Google Scholar] [CrossRef]
  161. Schlaepfer, I.R.; Hitz, C.A.; Gijon, M.A.; Bergman, B.C.; Eckel, R.H.; Jacobsen, B.M. Progestin modulates the lipid profile and sensitivity of breast cancer cells to docetaxel. Mol. Cell Endocrinol. 2012, 363, 111–121. [Google Scholar] [CrossRef] [Green Version]
  162. Rak, S.; De Zan, T.; Stefulj, J.; Kosovic, M.; Gamulin, O.; Osmak, M. FTIR spectroscopy reveals lipid droplets in drug resistant laryngeal carcinoma cells through detection of increased ester vibra-tional bands intensity. Analyst 2014, 139, 3407–3415. [Google Scholar] [CrossRef]
  163. Verbrugge, S.E.; Al, M.; Assaraf, Y.G.; Kammerer, S.; Chandrupatla, D.M.; Honeywell, R.; Musters, R.P.; Giovannetti, E.; O’Toole, T.; Scheffer, G.L.; et al. Multifactorial resistance to aminopeptidase inhibitor prodrug CHR2863 in myeloid leukemia cells: Down-regulation of carboxylesterase 1, drug sequestration in lipid droplets and pro-survival activation ERK/Akt/mTOR. Oncotarget 2016, 7, 5240–5257. [Google Scholar] [CrossRef] [Green Version]
  164. Li, J.; Condello, S.; Thomes-Pepin, J.; Ma, X.; Xia, Y.; Hurley, T.D.; Matei, D.; Cheng, J.X. Lipid Desaturation Is a Metabolic Marker and Therapeutic Target of Ovarian Cancer Stem Cells. Cell Stem Cell 2017, 20, 303–314.e5. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  165. Singh, S.R.; Zeng, X.; Zhao, J.; Liu, Y.; Hou, G.; Liu, H.; Hou, S.X. The lipolysis pathway sustains normal and transformed stem cells in adult Drosophila. Nature 2016, 538, 109–113. [Google Scholar] [CrossRef] [PubMed]
  166. Hershey, B.J.; Vazzana, R.; Joppi, D.L.; Havas, K.M. Lipid Droplets Define a Sub-Population of Breast Cancer Stem Cells. J. Clin. Med. 2019, 9, 87. [Google Scholar] [CrossRef] [Green Version]
  167. 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. 2019, 11, e10698. [Google Scholar] [CrossRef]
  168. Siddiqui, S.; Glauben, R. Fatty Acid Metabolism in Myeloid-Derived Suppressor Cells and Tumor-Associated Macrophages: Key Factor in Cancer Immune Evasion. Cancers 2022, 14, 250. [Google Scholar] [CrossRef] [PubMed]
  169. Veglia, F.; Tyurin, V.A.; Mohammadyani, D.; Blasi, M.; Duperret, E.K.; Donthireddy, L.; Hashimoto, A.; Kapralov, A.; Amoscato, A.; Angelini, R.; et al. Lipid bodies containing oxidatively truncated lipids block antigen cross-presentation by dendritic cells in cancer. Nat. Commun. 2017, 8, 2122. [Google Scholar] [CrossRef] [PubMed]
  170. Schade, D.S.; Shey, L.; Eaton, R.P. Cholesterol Review: A Metabolically Important Molecule. Endocr. Pract. 2020, 26, 1514–1523. [Google Scholar] [CrossRef] [PubMed]
  171. Radhakrishnan, A.; Goldstein, J.L.; McDonald, J.G.; Brown, M.S. Switch-like control of SREBP-2 transport triggered by small changes in ER cholesterol: A delicate balance. Cell Metab. 2008, 8, 512–521. [Google Scholar] [CrossRef] [Green Version]
  172. Zelcer, N.; Tontonoz, P. Liver X receptors as integrators of metabolic and inflammatory signaling. J. Clin. Investig. 2006, 116, 607–614. [Google Scholar] [CrossRef] [Green Version]
  173. Calkin, A.C.; Tontonoz, P. Liver x receptor signaling pathways and atherosclerosis. Arterioscler. Thromb. Vasc. Biol. 2010, 30, 1513–1518. [Google Scholar] [CrossRef] [Green Version]
  174. Chang, T.Y.; Chang, C.C.; Ohgami, N.; Yamauchi, Y. Cholesterol sensing, trafficking, and esterification. Annu. Rev. Cell Dev. Biol. 2006, 22, 129–157. [Google Scholar] [CrossRef] [PubMed]
  175. Hai, Q.; Smith, J.D. Acyl-Coenzyme A: Cholesterol Acyltransferase (ACAT) in Cholesterol Metabolism: From Its Discovery to Clinical Trials and the Genomics Era. Metabolites 2021, 11, 543. [Google Scholar] [CrossRef] [PubMed]
  176. Hofmann, K. A superfamily of membrane-bound O-acyltransferases with implications for wnt signaling. Trends Biochem. Sci. 2000, 25, 111–112. [Google Scholar] [CrossRef]
  177. Oelkers, P.; Behari, A.; Cromley, D.; Billheimer, J.T.; Sturley, S.L. Characterization of two human genes encoding acyl coenzyme A:cholesterol acyltransferase-related enzymes. J. Biol. Chem. 1998, 273, 26765–26771. [Google Scholar] [CrossRef] [Green Version]
  178. Chang, C.C.; Sakashita, N.; Ornvold, K.; Lee, O.; Chang, E.T.; Dong, R.; Lin, S.; Lee, C.Y.; Strom, S.C.; Kashyap, R.; et al. Immunological quantitation and localization of ACAT-1 and ACAT-2 in human liver and small intestine. J. Biol. Chem. 2000, 275, 28083–28092. [Google Scholar] [CrossRef]
  179. Chang, T.Y.; Li, B.L.; Chang, C.C.; Urano, Y. Acyl-coenzyme A: Cholesterol acyltransferases. Am. J. Physiol. Endocrinol. Metab. 2009, 297, E1–E9. [Google Scholar] [CrossRef] [Green Version]
  180. Shibuya, Y.; Chang, C.C.; Chang, T.Y. ACAT1/SOAT1 as a therapeutic target for Alzheimer’s disease. Future Med. Chem. 2015, 7, 2451–2467. [Google Scholar] [CrossRef] [Green Version]
  181. Yue, S.; Li, J.; Lee, S.Y.; Lee, H.J.; Shao, T.; Song, B.; Cheng, L.; Masterson, T.A.; Liu, X.; Ratliff, T.L.; et al. Cholesteryl ester accumulation induced by PTEN loss and PI3K/AKT activation underlies human prostate cancer aggressiveness. Cell Metab. 2014, 19, 393–406. [Google Scholar] [CrossRef] [Green Version]
  182. Liu, Y.; Wang, Y.; Hao, S.; Qin, Y.; Wu, Y. Knockdown of sterol O-acyltransferase 1 (SOAT1) suppresses SCD1-mediated lipogenesis and cancer procession in prostate cancer. Prostaglandins Other Lipid Mediat. 2021, 153, 106537. [Google Scholar] [CrossRef]
  183. Eckhardt, C.; Sbiera, I.; Krebs, M.; Sbiera, S.; Spahn, M.; Kneitz, B.; Joniau, S.; Fassnacht, M.; Kubler, H.; Weigand, I.; et al. High expression of Sterol-O-Acyl transferase 1 (SOAT1), an enzyme involved in cholesterol metabolism, is associated with earlier biochemical recurrence in high risk prostate cancer. Prostate Cancer Prostatic Dis. 2021. [Google Scholar] [CrossRef]
  184. Jiang, Y.; Sun, A.; Zhao, Y.; Ying, W.; Sun, H.; Yang, X.; Xing, B.; Sun, W.; Ren, L.; Hu, B.; et al. Proteomics identifies new therapeutic targets of early-stage hepatocellular carcinoma. Nature 2019, 567, 257–261. [Google Scholar] [CrossRef] [PubMed]
  185. Ren, M.; Xu, H.; Xia, H.; Tang, Q.; Bi, F. Simultaneously targeting SOAT1 and CPT1A ameliorates hepatocellular carcinoma by disrupting lipid homeostasis. Cell Death Discov. 2021, 7, 125. [Google Scholar] [CrossRef] [PubMed]
  186. Chen, Y.; Yang, X.; Chen, Y.; Chen, G.; Winkler, C.A.; An, P.; Lyu, J. Impacts of the SOAT1 genetic variants and protein expression on HBV-related hepatocellular carcinoma. BMC Cancer 2021, 21, 615. [Google Scholar] [CrossRef] [PubMed]
  187. Zhu, Y.; Gu, L.; Lin, X.; Zhou, X.; Lu, B.; Liu, C.; Li, Y.; Prochownik, E.V.; Karin, M.; Wang, F.; et al. P53 deficiency affects cholesterol esterification to exacerbate hepatocarcinogenesis. Hepatology 2022. [Google Scholar] [CrossRef] [PubMed]
  188. Oni, T.E.; Biffi, G.; Baker, L.A.; Hao, Y.; Tonelli, C.; Somerville, T.D.D.; Deschenes, A.; Belleau, P.; Hwang, C.I.; Sanchez-Rivera, F.J.; et al. SOAT1 promotes mevalonate pathway dependency in pancreatic cancer. J. Exp. Med. 2020, 217, e20192389. [Google Scholar] [CrossRef] [PubMed]
  189. Wang, X.C.; Luo, L.M.; Huang, T.S.; Feng, L.F. SOAT1 is a new prognostic factor of colorectal cancer. Ir. J. Med. Sci. 2021, 191, 1549–1554. [Google Scholar] [CrossRef]
  190. Zhu, T.; Wang, Z.; Zou, T.; Xu, L.; Zhang, S.; Chen, Y.; Chen, C.; Zhang, W.; Wang, S.; Ding, Q.; et al. SOAT1 Promotes Gastric Cancer Lymph Node Metastasis Through Lipid Synthesis. Front. Pharm. 2021, 12, 769647. [Google Scholar] [CrossRef]
  191. Mo, Y.; Lin, L.; Zhang, J.; Yu, C. SOAT1 enhances lung cancer invasiveness by stimulating AKT-mediated mitochondrial fragmentation. Biochem. Cell Biol. 2022, 100, 68–74. [Google Scholar] [CrossRef]
  192. Smith, D.C.; Kroiss, M.; Kebebew, E.; Habra, M.A.; Chugh, R.; Schneider, B.J.; Fassnacht, M.; Jafarinasabian, P.; Ijzerman, M.M.; Lin, V.H.; et al. A phase 1 study of nevanimibe HCl, a novel adrenal-specific sterol O-acyltransferase 1 (SOAT1) inhibitor, in adrenocortical carcinoma. Investig. New Drugs 2020, 38, 1421–1429. [Google Scholar] [CrossRef]
  193. Cheng, X.; Li, J.; Guo, D. SCAP/SREBPs are Central Players in Lipid Metabolism and Novel Metabolic Targets in Cancer Therapy. Curr. Top. Med. Chem. 2018, 18, 484–493. [Google Scholar] [CrossRef]
  194. Lohr, M.; Hartig, W.; Schulze, A.; Kroiss, M.; Sbiera, S.; Lapa, C.; Mages, B.; Strobel, S.; Hundt, J.E.; Bohnert, S.; et al. SOAT1: A Suitable Target for Therapy in High-Grade Astrocytic Glioma? Int. J. Mol. Sci. 2022, 23, 3726. [Google Scholar] [CrossRef] [PubMed]
  195. Yoon, H.; Shaw, J.L.; Haigis, M.C.; Greka, A. Lipid metabolism in sickness and in health: Emerging regulators of lipotoxicity. Mol. Cell 2021, 81, 3708–3730. [Google Scholar] [CrossRef] [PubMed]
  196. Listenberger, L.L.; Han, X.; Lewis, S.E.; Cases, S.; Farese, R.V., Jr.; Ory, D.S.; Schaffer, J.E. Triglyceride accumulation protects against fatty acid-induced lipotoxicity. Proc. Natl. Acad. Sci. USA 2003, 100, 3077–3082. [Google Scholar] [CrossRef] [Green Version]
  197. Hardy, S.; El-Assaad, W.; Przybytkowski, E.; Joly, E.; Prentki, M.; Langelier, Y. Saturated fatty acid-induced apoptosis in MDA-MB-231 breast cancer cells. A role for cardiolipin. J. Biol. Chem. 2003, 278, 31861–31870. [Google Scholar] [CrossRef] [Green Version]
  198. Senkal, C.E.; Salama, M.F.; Snider, A.J.; Allopenna, J.J.; Rana, N.A.; Koller, A.; Hannun, Y.A.; Obeid, L.M. Ceramide Is Metabolized to Acylceramide and Stored in Lipid Droplets. Cell Metab. 2017, 25, 686–697. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  199. Obeid, L.M.; Linardic, C.M.; Karolak, L.A.; Hannun, Y.A. Programmed cell death induced by ceramide. Science 1993, 259, 1769–1771. [Google Scholar] [CrossRef]
  200. Bhatt-Wessel, B.; Jordan, T.W.; Miller, J.H.; Peng, L. Role of DGAT enzymes in triacylglycerol metabolism. Arch. Biochem. Biophys. 2018, 655, 1–11. [Google Scholar] [CrossRef]
  201. Hernandez-Corbacho, M.J.; Obeid, L.M. A novel role for DGATs in cancer. Adv. Biol. Regul. 2019, 72, 89–101. [Google Scholar] [CrossRef]
  202. Cases, S.; Smith, S.J.; Zheng, Y.W.; Myers, H.M.; Lear, S.R.; Sande, E.; Novak, S.; Collins, C.; Welch, C.B.; Lusis, A.J.; et al. Identification of a gene encoding an acyl CoA:diacylglycerol acyltransferase, a key enzyme in triacylglycerol synthesis. Proc. Natl. Acad. Sci. USA 1998, 95, 13018–13023. [Google Scholar] [CrossRef] [Green Version]
  203. Cases, S.; Stone, S.J.; Zhou, P.; Yen, E.; Tow, B.; Lardizabal, K.D.; Voelker, T.; Farese, R.V., Jr. Cloning of DGAT2, a second mammalian diacylglycerol acyltransferase, and related family members. J. Biol. Chem. 2001, 276, 38870–38876. [Google Scholar] [CrossRef] [Green Version]
  204. van Rijn, J.M.; Ardy, R.C.; Kuloglu, Z.; Harter, B.; van Haaften-Visser, D.Y.; van der Doef, H.P.J.; van Hoesel, M.; Kansu, A.; van Vugt, A.H.M.; Thian, M.; et al. Intestinal Failure and Aberrant Lipid Metabolism in Patients With DGAT1 Deficiency. Gastroenterology 2018, 155, 130–143.e5. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  205. Stephen, J.; Vilboux, T.; Haberman, Y.; Pri-Chen, H.; Pode-Shakked, B.; Mazaheri, S.; Marek-Yagel, D.; Barel, O.; Di Segni, A.; Eyal, E.; et al. Congenital protein losing enteropathy: An inborn error of lipid metabolism due to DGAT1 mutations. Eur. J. Hum. Genet. 2016, 24, 1268–1273. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  206. Stone, S.J.; Myers, H.M.; Watkins, S.M.; Brown, B.E.; Feingold, K.R.; Elias, P.M.; Farese, R.V., Jr. Lipopenia and skin barrier abnormalities in DGAT2-deficient mice. J. Biol. Chem. 2004, 279, 11767–11776. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  207. Smith, S.J.; Cases, S.; Jensen, D.R.; Chen, H.C.; Sande, E.; Tow, B.; Sanan, D.A.; Raber, J.; Eckel, R.H.; Farese, R.V., Jr. Obesity resistance and multiple mechanisms of triglyceride synthesis in mice lacking Dgat. Nat. Genet 2000, 25, 87–90. [Google Scholar] [CrossRef]
  208. Xia, L.; Wang, Y.; Cai, S.; Xu, M. DGAT1 Expression Promotes Ovarian Cancer Progression and Is Associated with Poor Prognosis. J. Immunol. Res. 2021, 2021, 6636791. [Google Scholar] [CrossRef]
  209. Nardi, F.; Franco, O.E.; Fitchev, P.; Morales, A.; Vickman, R.E.; Hayward, S.W.; Crawford, S.E. DGAT1 Inhibitor Suppresses Prostate Tumor Growth and Migration by Regulating Intracellular Lipids and Non-Centrosomal MTOC Protein GM130. Sci. Rep. 2019, 9, 3035. [Google Scholar] [CrossRef]
  210. de la Rosa Rodriguez, M.A.; Deng, L.; Gemmink, A.; van Weeghel, M.; Aoun, M.L.; Warnecke, C.; Singh, R.; Borst, J.W.; Kersten, S. Hypoxia-inducible lipid droplet-associated induces DGAT1 and promotes lipid storage in hepatocytes. Mol. Metab. 2021, 47, 101168. [Google Scholar] [CrossRef]
  211. Morales, A.; Greenberg, M.; Nardi, F.; Gil, V.; Hayward, S.W.; Crawford, S.E.; Franco, O.E. Loss of ephrin B2 receptor (EPHB2) sets lipid rheostat by regulating proteins DGAT1 and ATGL inducing lipid droplet storage in prostate cancer cells. Lab. Investig. 2021, 101, 921–934. [Google Scholar] [CrossRef]
  212. Mitra, R.; Le, T.T.; Gorjala, P.; Goodman, O.B., Jr. Positive regulation of prostate cancer cell growth by lipid droplet forming and processing enzymes DGAT1 and ABHD5. BMC Cancer 2017, 17, 631. [Google Scholar] [CrossRef]
  213. He, P.; Cheng, S.; Hu, F.; Ma, Z.; Xia, Y. Up-regulation of DGAT1 in cancer tissues and tumor-infiltrating macrophages influenced survival of patients with gastric cancer. BMC Cancer 2021, 21, 252. [Google Scholar] [CrossRef]
  214. de la Rosa Rodriguez, M.A.; Kersten, S. Regulation of lipid droplet homeostasis by hypoxia inducible lipid droplet associated HILPDA. Biochim. Biophys. Acta Mol. Cell Biol. Lipids 2020, 1865, 158738. [Google Scholar] [CrossRef] [PubMed]
  215. Povero, D.; Johnson, S.M.; Liu, J. Hypoxia, hypoxia-inducible gene 2 (HIG2)/HILPDA, and intracellular lipolysis in cancer. Cancer Lett. 2020, 493, 71–79. [Google Scholar] [CrossRef] [PubMed]
  216. Mao, X.G.; Wang, C.; Liu, D.Y.; Zhang, X.; Wang, L.; Yan, M.; Zhang, W.; Zhu, J.; Li, Z.C.; Mi, C.; et al. Hypoxia upregulates HIG2 expression and contributes to bevacizumab resistance in glioblastoma. Oncotarget 2016, 7, 47808–47820. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  217. Yu, L.; Li, Y.; Grisé, A.; Wang, H. CGI-58: Versatile Regulator of Intracellular Lipid Droplet Homeostasis. Adv. Exp. Med. Biol. 2020, 1276, 197–222. [Google Scholar] [PubMed]
  218. Shi, Z.; Luo, X.; Zhao, H.; Huang, B.; Wang, Y.; Chen, X.; Yu, J. Clinicalpathologic and Prognostic Significance of CGI-58 in Endometrial Cancer. J. Cancer 2021, 12, 7374–7379. [Google Scholar] [CrossRef]
  219. Chen, G.; Zhou, G.; Lotvola, A.; Granneman, J.G.; Wang, J. ABHD5 suppresses cancer cell anabolism through lipolysis-dependent activation of the AMPK/mTORC1 pathway. J. Biol. Chem. 2021, 296, 100104. [Google Scholar] [CrossRef]
  220. Long, T.; Sun, Y.; Hassan, A.; Qi, X.; Li, X. Structure of nevanimibe-bound tetrameric human ACAT1. Nature 2020, 581, 339–343. [Google Scholar] [CrossRef]
  221. Qian, H.; Zhao, X.; Yan, R.; Yao, X.; Gao, S.; Sun, X.; Du, X.; Yang, H.; Wong, C.C.L.; Yan, N. Structural basis for catalysis and substrate specificity of human ACAT1. Nature 2020, 581, 333–338. [Google Scholar] [CrossRef]
  222. Guan, C.; Niu, Y.; Chen, S.C.; Kang, Y.; Wu, J.X.; Nishi, K.; Chang, C.C.Y.; Chang, T.Y.; Luo, T.; Chen, L. Structural insights into the inhibition mechanism of human sterol O-acyltransferase 1 by a competitive inhibitor. Nat. Commun. 2020, 11, 2478. [Google Scholar] [CrossRef]
  223. Sui, X.; Wang, K.; Gluchowski, N.L.; Elliott, S.D.; Liao, M.; Walther, T.C.; Farese, R.V., Jr. Structure and catalytic mechanism of a human triacylglycerol-synthesis enzyme. Nature 2020, 581, 323–328. [Google Scholar] [CrossRef]
  224. Wang, L.; Qian, H.; Nian, Y.; Han, Y.; Ren, Z.; Zhang, H.; Hu, L.; Prasad, B.V.V.; Laganowsky, A.; Yan, N.; et al. Structure and mechanism of human diacylglycerol O-acyltransferase 1. Nature 2020, 581, 329–332. [Google Scholar] [CrossRef] [PubMed]
  225. Wen, Y.-A.; Xiong, X.; Zaytseva, Y.Y.; Napier, D.L.; Vallee, E.; Li, A.T.; Wang, C.; Weiss, H.L.; Evers, B.M.; Gao, T. Downregulation of SREBP inhibits tumor growth and initiation by altering cellular metabolism in colon cancer. Cell Death Dis. 2018, 9, 265. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  226. Jie, Z.; Xie, Z.; Xu, W.; Zhao, X.; Jin, G.; Sun, X.; Huang, B.; Tang, P.; Wang, G.; Shen, S.; et al. SREBP-2 aggravates breast cancer associated osteolysis by promoting osteoclastogenesis and breast cancer metastasis. Biochim. Biophys. Acta Mol. Basis Dis. 2019, 1865, 115–125. [Google Scholar] [CrossRef] [PubMed]
  227. Yao, L.; Chen, S.; Li, W. Fatostatin inhibits the development of endometrial carcinoma in endometrial carcinoma cells and a xenograft model by targeting lipid metabolism. Arch. Bio-Chem. Biophys. 2020, 684, 108327. [Google Scholar] [CrossRef] [PubMed]
  228. Brovkovych, V.; Izhar, Y.; Danes, J.M.; Dubrovskyi, O.; Sakallioglu, I.T.; Morrow, L.M.; Atilla-Gokcumen, G.E.; Frasor, J. Fatostatin induces pro- and anti-apoptotic lipid accumulation in breast cancer. Oncogenesis 2018, 7, 66. [Google Scholar] [CrossRef] [PubMed]
  229. Li, X.; Chen, Y.T.; Hu, P.; Huang, W.C. Fatostatin displays high antitumor activity in prostate cancer by blocking SREBP-regulated metabolic pathways and androgen receptor signaling. Mol. Cancer Ther. 2014, 13, 855–866. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  230. Yin, F.; Feng, F.; Wang, L.; Wang, X.; Li, Z.; Cao, Y. SREBP-1 inhibitor Betulin enhances the antitumor effect of Sorafenib on hepatocellular carcinoma via restricting cellular glycolytic activity. Cell Death Dis. 2019, 10, 672. [Google Scholar] [CrossRef] [Green Version]
  231. Li, J.; Yan, H.; Zhao, L.; Jia, W.; Yang, H.; Liu, L.; Zhou, X.; Miao, P.; Sun, X.; Song, S.; et al. Inhibition of SREBP increases gefitinib sensitivity in non-small cell lung cancer cells. Oncotarget 2016, 7, 52392–52403. [Google Scholar] [CrossRef] [Green Version]
  232. Caruana, B.T.; Skoric, A.; Brown, A.J.; Lutze-Mann, L.H. Site-1 protease, a novel metabolic target for glioblastoma. Biochem. Biophys. Res. Commun. 2017, 490, 760–766. [Google Scholar] [CrossRef]
  233. Wang, T.-B.; Geng, M.; Jin, H.; Tang, A.-G.; Sun, H.; Zhou, L.-Z.; Chen, B.-H.; Shen, G.; Sun, Q. SREBP1 site 1 protease inhibitor PF-429242 suppresses renal cell carcinoma cell growth. Cell Death Dis. 2021, 12, 717. [Google Scholar] [CrossRef]
  234. Siqingaowa Sekar, S.; Gopalakrishnan, V.; Taghibiglou, C. Sterol regulatory element-binding protein 1 inhibitors decrease pancreatic cancer cell viability and proliferation. Biochem. Biophys. Res. Commun. 2017, 488, 136–140. [Google Scholar] [CrossRef] [PubMed]
  235. Esquejo, R.M.; Roqueta-Rivera, M.; Shao, W.; Phelan, P.E.; Seneviratne, U.; Am Ende, C.W.; Hershberger, P.M.; Machamer, C.E.; Espenshade, P.J.; Osborne, T.F. Dipyridamole Inhibits Lipogenic Gene Expression by Retaining SCAP-SREBP in the Endoplasmic Reticulum. Cell Chem. Biol. 2021, 28, 169–179.e7. [Google Scholar] [CrossRef] [PubMed]
  236. Granchi, C. ATP-citrate lyase (ACLY) inhibitors as therapeutic agents: A patenting perspective. Expert Opin. Ther. Pat. 2022, 32, 731–742. [Google Scholar] [CrossRef]
  237. Hatzivassiliou, G.; Zhao, F.; Bauer, D.E.; Andreadis, C.; Shaw, A.N.; Dhanak, D.; Hingorani, S.R.; Tuveson, D.A.; Thompson, C.B. ATP citrate lyase inhibition can suppress tumor cell growth. Cancer Cell 2005, 8, 311–321. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  238. Svensson, R.U.; Parker, S.J.; Eichner, L.J.; Kolar, M.J.; Wallace, M.; Brun, S.N.; Lombardo, P.S.; Van Nostrand, J.L.; Hutchins, A.; Vera, L.; et al. Inhibition of acetyl-CoA carboxylase suppresses fatty acid synthesis and tumor growth of non-small-cell lung cancer in preclinical models. Nat. Med. 2016, 22, 1108–1119. [Google Scholar] [CrossRef] [Green Version]
  239. Lally, J.S.V.; Ghoshal, S.; DePeralta, D.K.; Moaven, O.; Wei, L.; Masia, R.; Erstad, D.J.; Fujiwara, N.; Leong, V.; Houde, V.P.; et al. Inhibition of Acetyl-CoA Carboxylase by Phosphorylation or the Inhibitor ND-654 Suppresses Lipogenesis and Hepatocellular Carcinoma. Cell Metab. 2019, 29, 174–182.e175. [Google Scholar] [CrossRef] [Green Version]
  240. Tan, K. Investigating the Role of Acetyl-CoA Carboxylases in Breast Cancer Progression. Doctoral Dissertation, The University of Sydney, Camperdown, Australia, 2021. [Google Scholar]
  241. Pizer, E.S.; Thupari, J.; Han, W.F.; Pinn, M.L.; Chrest, F.J.; Frehywot, G.L.; Townsend, C.A.; Kuhajda, F.P. Malonyl-coenzyme-A is a potential mediator of cytotoxicity induced by fatty-acid synthase inhibition in human breast cancer cells and xenografts. Cancer Res. 2000, 60, 213–218. [Google Scholar]
  242. Qi, J.; Lang, W.; Geisler, J.G.; Wang, P.; Petrounia, I.; Mai, S.; Smith, C.; Askari, H.; Struble, G.T.; Williams, R.; et al. The use of stable isotope-labeled glycerol and oleic acid to differentiate the hepatic functions of DGAT1 and-2. J. Lipid Res. 2012, 53, 1106–1116. [Google Scholar] [CrossRef] [Green Version]
  243. Balaban, S.; Nassar, Z.D.; Zhang, A.Y.; Hosseini-Beheshti, E.; Centenera, M.M.; Schreuder, M.; Lin, H.-M.; Aishah, A.; Varney, B.; Liu-Fu, F.; et al. Extracellular fatty acids are the major contributor to lipid synthesis in prostate cancer. Mol. Cancer Res. 2019, 17, 949–962. [Google Scholar] [CrossRef]
  244. Dow, R.L.; Li, J.C.; Pence, M.P.; Gibbs, E.M.; LaPerle, J.L.; Litchfield, J.; Piotrowski, D.W.; Munchhof, M.J.; Manion, T.B.; Zavadoski, W.J.; et al. Discovery of PF-04620110, a Potent, Selective, and Orally Bioavailable Inhibitor of DGAT-1. ACS Med. Chem. Lett. 2011, 2, 407–412. [Google Scholar] [CrossRef] [Green Version]
  245. Falchook, G.; Infante, J.; Arkenau, H.T.; Patel, M.R.; Dean, E.; Borazanci, E.; Brenner, A.; Cook, N.; Lopez, J.; Pant, S.; et al. First-in-human study of the safety, pharmacokinetics, and pharmacodynamics of first-in-class fatty acid synthase inhibitor TVB-2640 alone and with a taxane in advanced tumors. EClinicalMedicine 2021, 34, 100797. [Google Scholar] [CrossRef] [PubMed]
  246. Zhu, Y.; Gu, L.; Lin, X.; Zhang, J.; Tang, Y.; Zhou, X.; Lu, B.; Lin, X.; Liu, C.; Prochownik, E.V.; et al. Ceramide-mediated gut dysbiosis enhances cholesterol esterification and promotes colorectal tu-morigenesis in mice. JCI Insight 2022, 7, e150607. [Google Scholar] [CrossRef] [PubMed]
  247. Ohmoto, T.; Nishitsuji, K.; Yoshitani, N.; Mizuguchi, M.; Yanagisawa, Y.; Saito, H.; Sakashita, N. K604, a specific acyl-CoA:cholesterol acyltransferase 1 inhibitor, suppresses proliferation of U251-MG glioblastoma cells. Mol. Med. Rep. 2015, 12, 6037–6042. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  248. Rae, C.; Fragkoulis, G.I.; Chalmers, A.J. Cytotoxicity and Radiosensitizing Activity of the Fatty Acid Synthase Inhibitor C75 Is Enhanced by Blocking Fatty Acid Uptake in Prostate Cancer Cells. Adv. Radiat. Oncol. 2020, 5, 994–1005. [Google Scholar] [CrossRef]
  249. Grube, S.; Dünisch, P.; Freitag, D.; Klausnitzer, M.; Sakr, Y.; Walter, J.; Kalff, R.; Ewald, C. Overexpression of fatty acid synthase in human gliomas correlates with the WHO tumor grade and inhibition with Orlistat reduces cell viability and triggers apoptosis. J. Neuro-Oncol. 2014, 118, 277–287. [Google Scholar] [CrossRef]
  250. Hu, X.; Xiang, J.; Li, Y.; Xia, Y.; Xu, S.; Gao, X.; Qiao, S. Inhibition of Stearoyl-CoA Desaturase 1 Potentiates Anti-tumor Activity of Amodiaquine in Non-small Cell Lung Cancer. Biol. Pharm. Bull. 2022, 45, 438–445. [Google Scholar] [CrossRef]
  251. Skrypek, K.; Balog, S.; Eriguchi, Y.; Asahina, K. Inhibition of Stearoyl-CoA Desaturase Induces the Unfolded Protein Response in Pancreatic Tumors and Suppresses Their Growth. Pancreas 2021, 50, 219–226. [Google Scholar] [CrossRef]
  252. She, K.; Fang, S.; Du, W.; Fan, X.; He, J.; Pan, H.; Huang, L.; He, P.; Huang, J. SCD1 is required for EGFR-targeting cancer therapy of lung cancer via re-activation of EGFR/PI3K/AKT signals. Cancer Cell Int. 2019, 19, 103. [Google Scholar] [CrossRef]
  253. von Roemeling, C.A.; Marlow, L.A.; Wei, J.J.; Cooper, S.J.; Caulfield, T.R.; Wu, K.; Tan, W.W.; Tun, H.W.; Copland, J.A. Stearoyl-CoA desaturase 1 is a novel molecular therapeutic target for clear cell renal cell carcinoma. Clin. Cancer Res. Off. J. Am. Assoc. Cancer Res. 2013, 19, 2368–2380. [Google Scholar] [CrossRef] [Green Version]
  254. Pisanu, M.E.; Noto, A.; De Vitis, C.; Morrone, S.; Scognamiglio, G.; Botti, G.; Venuta, F.; Diso, D.; Jakopin, Z.; Padula, F.; et al. Blockade of Stearoyl-CoA-desaturase 1 activity reverts resistance to cisplatin in lung cancer stem cells. Cancer Lett. 2017, 406, 93–104. [Google Scholar] [CrossRef]
  255. Pisanu, M.E.; Maugeri-Saccà, M.; Fattore, L.; Bruschini, S.; De Vitis, C.; Tabbì, E.; Bellei, B.; Migliano, E.; Kovacs, D.; Camera, E.; et al. Inhibition of Stearoyl-CoA desaturase 1 reverts BRAF and MEK inhibition-induced selection of cancer stem cells in BRAF-mutated melanoma. J. Exp. Clin. Cancer Res. CR 2018, 37, 318. [Google Scholar] [CrossRef] [PubMed]
  256. Oballa, R.M.; Belair, L.; Black, W.C.; Bleasby, K.; Chan, C.C.; Desroches, C.; Du, X.; Gordon, R.; Guay, J.; Guiral, S.; et al. Development of a liver-targeted stearoyl-CoA desaturase (SCD) inhibitor (MK-8245) to establish a therapeutic window for the treatment of diabetes and dyslipidemia. J. Med. Chem. 2011, 54, 5082–5096. [Google Scholar] [CrossRef] [PubMed]
  257. Sabnis, R.W. Novel Substituted Tetrazoles as ACSS2 Inhibitors for Treating Cancer. ACS Med. Chem. Lett. 2021, 12, 1894–1895. [Google Scholar] [CrossRef] [PubMed]
  258. Sabnis, R.W. Amide-Substituted Condensed Pyridine Derivatives as ACSS2 Inhibitors for Treating Cancer. ACS Med. Chem. Lett. 2021, 12, 1870–1871. [Google Scholar] [CrossRef] [PubMed]
  259. Kalthoff, C.; Xiang, K.; Muench, C.; Hlouschek, J.; Jendrossek, V.; Matschke, J. Abstract PO-022: Inhibition of the citrate carrier SLC25A1 affects repair of radiation-induced DNA damage, increases radiosensitivity, and enhances lethality of IR in combination with DNA repair defects or DSB inhibitors. Clin. Cancer Res. 2021, 27 (Suppl. 8), PO-022. [Google Scholar] [CrossRef]
  260. Hlouschek, J.; Hansel, C.; Jendrossek, V.; Matschke, J. The Mitochondrial Citrate Carrier (SLC25A1) Sustains Redox Homeostasis and Mitochondrial Metabolism Supporting Radioresistance of Cancer Cells With Tolerance to Cycling Severe Hypoxia. Front. Oncol. 2018, 8, 170. [Google Scholar] [CrossRef]
  261. Reis, L.M.D.; Adamoski, D.; Ornitz Oliveira Souza, R.; Rodrigues Ascenção, C.F.; Sousa de Oliveira, K.R.; Corrêa-da-Silva, F.; Malta de Sá Patroni, F.; Meira Dias, M.; Consonni, S.R.; Mendes de Moraes-Vieira, P.M.; et al. Dual inhibition of glutaminase and carnitine palmitoyltransferase decreases growth and migration of glutaminase inhibition-resistant triple-negative breast cancer cells. J. Biol. Chem. 2019, 294, 9342–9357. [Google Scholar] [CrossRef]
  262. Jiang, N.; Xie, B.; Xiao, W.; Fan, M.; Xu, S.; Duan, Y.; Hamsafar, Y.; Evans, A.C.; Huang, J.; Zhou, W.; et al. Fatty acid oxidation fuels glioblastoma radioresistance with CD47-mediated immune evasion. Nat. Commun. 2022, 13, 1511. [Google Scholar] [CrossRef]
  263. Galicia-Vázquez, G.; Aloyz, R. Ibrutinib Resistance Is Reduced by an Inhibitor of Fatty Acid Oxidation in Primary CLL Lymphocytes. Front. Oncol. 2018, 8, 411. [Google Scholar] [CrossRef]
  264. Flaig, T.W.; Salzmann-Sullivan, M.; Su, L.J.; Zhang, Z.; Joshi, M.; Gijón, M.A.; Kim, J.; Arcaroli, J.J.; Van Bokhoven, A.; Lucia, M.S.; et al. Lipid catabolism inhibition sensitizes prostate cancer cells to antiandrogen blockade. Oncotarget 2017, 8, 56051–56065. [Google Scholar] [CrossRef] [Green Version]
  265. Schlaepfer, I.R.; Rider, L.; Rodrigues, L.U.; Gijón, M.A.; Pac, C.T.; Romero, L.; Cimic, A.; Sirintrapun, S.J.; Glodé, L.M.; Eckel, R.H.; et al. Lipid catabolism via CPT1 as a therapeutic target for prostate cancer. Mol. Cancer Ther. 2014, 13, 2361–2371. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  266. Dhakal, B.; Li, C.M.Y.; Li, R.; Yeo, K.; Wright, J.A.; Gieniec, K.A.; Vrbanac, L.; Sammour, T.; Lawrence, M.; Thomas, M.; et al. The Antianginal Drug Perhexiline Displays Cytotoxicity against Colorectal Cancer Cells In Vitro: A Potential for Drug Repurposing. Cancers 2022, 14, 1043. [Google Scholar] [CrossRef] [PubMed]
  267. Kant, S.; Kesarwani, P.; Guastella, A.R.; Kumar, P.; Graham, S.F.; Buelow, K.L.; Nakano, I.; Chinnaiyan, P. Perhexiline Demonstrates FYN-mediated Antitumor Activity in Glioblastoma. Mol. Cancer Ther. 2020, 19, 1415–1422. [Google Scholar] [CrossRef] [PubMed]
  268. Ricciardi, M.R.; Mirabilii, S.; Allegretti, M.; Licchetta, R.; Calarco, A.; Torrisi, M.R.; Foà, R.; Nicolai, R.; Peluso, G.; Tafuri, A. Targeting the leukemia cell metabolism by the CPT1a inhibition: Functional preclinical effects in leukemias. Blood 2015, 126, 1925–1929. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  269. Zhou, J.; Simon, J.M.; Liao, C.; Zhang, C.; Hu, L.; Zurlo, G.; Liu, X.; Fan, C.; Hepperla, A.; Jia, L.; et al. An oncogenic JMJD6-DGAT1 axis tunes the epigenetic regulation of lipid droplet formation in clear cell renal cell carcinoma. Mol. Cell, 2022; in press. [Google Scholar]
Figure 1. Lipid metabolism reprogramming in cancer cells. (1) Combined elevation of glucose and glutamine consumption promotes lipogenesis by activating the SREBP/SCAP pathway. Glucose produces N-glycans through HBP (Hexosamine Biosynthesis Pathway), which stabilizes SCAP (SREBP-cleavage activating protein) through N-linked glycosylation of its luminal loops (1 and 7). Glutamine enters the mitochondria and ammonia (NH3) is released by GLS (glutaminase). NH3 is protonated and converted to NH4+, which directly binds to the aspartate D428 and Serine S326/330 in the core of the SCAP transmembrane domains, forming stable hydrogen bonds. This binding triggers a dramatic conformation change in SCAP, leading to its dissociation from Insig (insulin-induced gene), an endoplasmic reticulum (ER)-resident protein. Subsequently, SCAP escorts SREBP (sterol regulatory element-binding protein) to the Golgi, where it is cleaved by two enzymes S1P (site 1 protease) and S2P (site 2 protease) to release its active N-terminal fragment. Finally, the N-terminal domain goes into the nucleus, binds to the SRE (sterol regulatory element) motif located in the promoters of gene involved in lipogenesis to activate their transcription and promote de novo lipid synthesis and tumor growth. (2) Glucose is the main source for lipid synthesis. Glucose via glycolysis breaks down into pyruvate, which enters the mitochondria and is converted to acetyl-CoA by PDH (pyruvate dehydrogenase), followed by condensation with OAA (oxaloacetate) to form citrate to enter the TCA cycle (tricarboxylic acid cycle). Citrate is released to the cytosol via its mitochondria transporter, SLC25A1. Citrate is then cleaved to acetyl-CoA by ACLY (ATP citrate lyase), which serves as a precursor for fatty acid and cholesterol biosynthesis catalyzed by a series of enzymes that are the main transcriptional targets of SREBPs, as shown in the Figure. In addition, cytosol acetate can be converted to acetyl-CoA for lipid synthesis by ACCS2 (acetyl-CoA synthetase 2). Besides that, acetyl-CoA is also the substrate for the acetylation of histones, which is involved in epigenetic regulation. Moreover, glutamine through glutaminolysis contributes as an anaplerotic substrate to replenish tricarboxylic acid (TCA) cycle intermediates. Glutamate, the product of the first step of glutaminolysis, is converted to α-KG (α-ketoglutaric acid) and then enters into the TCA cycle. (3) SREBPs upregulates the expression of LDLR (low-density lipoprotein receptor), which binds to LDL and transports it into cells through the endocytosis process. LDL is then hydrolyzed in the lysosomes and cholesterol is released, promoting tumor growth. (4) Excess fatty acids and cholesterol are converted to TAG (triacylglycerol) and CE (cholesteryl ester) by DGAT1 (diacylglycerol O-acyltransferase 1) and sterol O acyl-transferase 1 (SOAT1) to form LDs (lipid droplets) and prevent toxicity from high lipid levels. Under conditions of nutrient deficiency, LDs are hydrolyzed by autophagy to release free fatty acids and cholesterol for tumor survival.
Figure 1. Lipid metabolism reprogramming in cancer cells. (1) Combined elevation of glucose and glutamine consumption promotes lipogenesis by activating the SREBP/SCAP pathway. Glucose produces N-glycans through HBP (Hexosamine Biosynthesis Pathway), which stabilizes SCAP (SREBP-cleavage activating protein) through N-linked glycosylation of its luminal loops (1 and 7). Glutamine enters the mitochondria and ammonia (NH3) is released by GLS (glutaminase). NH3 is protonated and converted to NH4+, which directly binds to the aspartate D428 and Serine S326/330 in the core of the SCAP transmembrane domains, forming stable hydrogen bonds. This binding triggers a dramatic conformation change in SCAP, leading to its dissociation from Insig (insulin-induced gene), an endoplasmic reticulum (ER)-resident protein. Subsequently, SCAP escorts SREBP (sterol regulatory element-binding protein) to the Golgi, where it is cleaved by two enzymes S1P (site 1 protease) and S2P (site 2 protease) to release its active N-terminal fragment. Finally, the N-terminal domain goes into the nucleus, binds to the SRE (sterol regulatory element) motif located in the promoters of gene involved in lipogenesis to activate their transcription and promote de novo lipid synthesis and tumor growth. (2) Glucose is the main source for lipid synthesis. Glucose via glycolysis breaks down into pyruvate, which enters the mitochondria and is converted to acetyl-CoA by PDH (pyruvate dehydrogenase), followed by condensation with OAA (oxaloacetate) to form citrate to enter the TCA cycle (tricarboxylic acid cycle). Citrate is released to the cytosol via its mitochondria transporter, SLC25A1. Citrate is then cleaved to acetyl-CoA by ACLY (ATP citrate lyase), which serves as a precursor for fatty acid and cholesterol biosynthesis catalyzed by a series of enzymes that are the main transcriptional targets of SREBPs, as shown in the Figure. In addition, cytosol acetate can be converted to acetyl-CoA for lipid synthesis by ACCS2 (acetyl-CoA synthetase 2). Besides that, acetyl-CoA is also the substrate for the acetylation of histones, which is involved in epigenetic regulation. Moreover, glutamine through glutaminolysis contributes as an anaplerotic substrate to replenish tricarboxylic acid (TCA) cycle intermediates. Glutamate, the product of the first step of glutaminolysis, is converted to α-KG (α-ketoglutaric acid) and then enters into the TCA cycle. (3) SREBPs upregulates the expression of LDLR (low-density lipoprotein receptor), which binds to LDL and transports it into cells through the endocytosis process. LDL is then hydrolyzed in the lysosomes and cholesterol is released, promoting tumor growth. (4) Excess fatty acids and cholesterol are converted to TAG (triacylglycerol) and CE (cholesteryl ester) by DGAT1 (diacylglycerol O-acyltransferase 1) and sterol O acyl-transferase 1 (SOAT1) to form LDs (lipid droplets) and prevent toxicity from high lipid levels. Under conditions of nutrient deficiency, LDs are hydrolyzed by autophagy to release free fatty acids and cholesterol for tumor survival.
Biomedicines 10 01943 g001
Figure 2. Targeting SOAT1 to disrupt cholesterol homeostasis for GBM therapy. (A) SOAT1 catalyzes the esterification of cholesterol to store excess free cholesterol into LDs, and thus is involved in maintaining cellular cholesterol homeostasis. Cholesterol negatively regulates SCAP/SREBP trafficking and SREBP activation. Cholesterol reduction in the ER activates SCAP dissociation from Insig, resulting in SREBP translocation and activation to promote lipogenesis. (B) Inhibition of SOAT1 reduces CE formation, which in turn results in the accumulation of cholesterol in the ER membrane, subsequently leading to the SCAP/SREBP complex remaining in the ER and to reduction of lipogenesis, along with enhanced ER stress, resulting in tumor suppression.
Figure 2. Targeting SOAT1 to disrupt cholesterol homeostasis for GBM therapy. (A) SOAT1 catalyzes the esterification of cholesterol to store excess free cholesterol into LDs, and thus is involved in maintaining cellular cholesterol homeostasis. Cholesterol negatively regulates SCAP/SREBP trafficking and SREBP activation. Cholesterol reduction in the ER activates SCAP dissociation from Insig, resulting in SREBP translocation and activation to promote lipogenesis. (B) Inhibition of SOAT1 reduces CE formation, which in turn results in the accumulation of cholesterol in the ER membrane, subsequently leading to the SCAP/SREBP complex remaining in the ER and to reduction of lipogenesis, along with enhanced ER stress, resulting in tumor suppression.
Biomedicines 10 01943 g002
Figure 3. Inhibition of DGAT1 as a therapy strategy to induce cancer cell death. (1) Left: Tumor cells acquire fatty acids through uptake or de novo synthesis via the activation of SREBP-1. Fatty acids are converted to acyl-CoAs, which are the substrates for phospholipid (PL) synthesis and produce energy via entering into mitochondria to undergo β-oxidation and oxidative phosphorylation. Excessive acyl-CoA is stored in LDs as catalyzed by DGAT1 in tumor cells. (2) Right: Inhibiting DGAT1 causes an imbalance of fatty acid catabolism, leading to cell death. Inhibiting DGAT1 forces more acyl-CoAs to enter the mitochondria through carnitine palmitate transferase 1 (CPT1) for β-oxidation, leading to high levels of reactive oxygen species (ROS), which trigger apoptosis and ferroptosis to kill tumor cells. Moreover, the profile of PLs is altered upon DGAT1 inhibition, contributing to further cellular stress.
Figure 3. Inhibition of DGAT1 as a therapy strategy to induce cancer cell death. (1) Left: Tumor cells acquire fatty acids through uptake or de novo synthesis via the activation of SREBP-1. Fatty acids are converted to acyl-CoAs, which are the substrates for phospholipid (PL) synthesis and produce energy via entering into mitochondria to undergo β-oxidation and oxidative phosphorylation. Excessive acyl-CoA is stored in LDs as catalyzed by DGAT1 in tumor cells. (2) Right: Inhibiting DGAT1 causes an imbalance of fatty acid catabolism, leading to cell death. Inhibiting DGAT1 forces more acyl-CoAs to enter the mitochondria through carnitine palmitate transferase 1 (CPT1) for β-oxidation, leading to high levels of reactive oxygen species (ROS), which trigger apoptosis and ferroptosis to kill tumor cells. Moreover, the profile of PLs is altered upon DGAT1 inhibition, contributing to further cellular stress.
Biomedicines 10 01943 g003
Publisher’s Note: MDPI stays neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Share and Cite

MDPI and ACS Style

Kou, Y.; Geng, F.; Guo, D. Lipid Metabolism in Glioblastoma: From De Novo Synthesis to Storage. Biomedicines 2022, 10, 1943. https://doi.org/10.3390/biomedicines10081943

AMA Style

Kou Y, Geng F, Guo D. Lipid Metabolism in Glioblastoma: From De Novo Synthesis to Storage. Biomedicines. 2022; 10(8):1943. https://doi.org/10.3390/biomedicines10081943

Chicago/Turabian Style

Kou, Yongjun, Feng Geng, and Deliang Guo. 2022. "Lipid Metabolism in Glioblastoma: From De Novo Synthesis to Storage" Biomedicines 10, no. 8: 1943. https://doi.org/10.3390/biomedicines10081943

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

Article Metrics

Back to TopTop