Abstract
In recent years, metabolic reprogramming has emerged as a significant breakthrough in elucidating the onset and progression of gastrointestinal (GI) malignancies. As central regulatory hubs for lipid metabolism, sterol regulatory element binding proteins (SREBPs) integrate dietary metabolic signals and carcinogenic stimuli through subtype-specific mechanisms, thereby promoting malignant tumour phenotypes. In this review, we first present the molecular background, structural characteristics, and posttranscriptional regulatory networks associated with SREBPs. We subsequently describe a systematic analysis of the distinct activation patterns of SREBPs in liver, gastric, colorectal, and other gastrointestinal cancers. Furthermore, we explore targeted intervention strategies for different SREBP subtypes, including small molecule inhibitors (such as fatostatin, which inhibits SREBP cleavage), natural compounds (such as berberine, which modulates the AMPK/mTOR pathway), and statin-mediated inhibition of the mevalonic acid pathway. These strategies may enhance tumour cell sensitivity to chemotherapeutic agents (such as 5-FU, gezil, and tabine) and improve the response to synergistic chemoradiotherapy by reversing adaptive metabolic resistance driven by the tumour microenvironment. Through this review, we hope to provide new insights into precise interventions targeting various subtypes of the SREBP molecule.
Keywords: SREBPs, GI tumours, Lipid metabolism reprogramming, Lipogenesis, Targeted therapy
Introduction
Gastrointestinal (GI) tumours, which include malignancies such as oesophageal, stomach, colorectal, pancreatic, and liver cancer, have emerged as prevalent, life-threatening diseases that significantly impact quality of life in humans [1–4]. Although the pathogenesis of GI tumours remains largely elusive, modifications in lipid metabolism have been identified as distinctive characteristics of this type of cancer. Increasing attention has also been given to changes in lipid metabolism in cancer [5, 6]. Cancer cells rely on significant lipid synthesis and uptake to support their replication. A diverse array of enzymes, proteins, and transcription factors are involved in the reprogramming of lipid metabolism in GI tumours [6, 7]. These molecules, when expressed abnormally, can enhance lipid production and promote the clustering of lipid droplets through various mechanisms, thus affecting the development, spread, and growth of GI tumours.
Sterol regulatory element-binding proteins (SREBPs) serve as primary regulators of lipogenesis and significantly contribute to the maintenance of appropriate cellular lipid levels, including triglycerides and sterols, via intricate proteolytic processing mechanisms [8]. When highly expressed, SREBPs have been observed to be instrumental in lipid reprogramming across a diverse range of cancers [9]. Recent research has indicated a notable increase in lipid synthesis and uptake within gastrointestinal tumours, and this factor substantiates tumour growth. The range of cancers affected by the high expression of SREBPs includes prostate, liver, colorectal, stomach, and pancreatic malignancies [9, 10]. SREBPs function as pivotal transcription factors that govern lipid metabolism and modulate the expression of essential genes implicated in lipid synthesis and uptake, processes that may also be linked to oncogenesis [11]. Consequently, the utilisation of therapeutic strategies focusing on SREBPs in gastrointestinal tumours could provide promising molecular targets for cancer treatment.
In this review, we conducted a comprehensive search of PubMed for articles pertaining to the three subtypes of SREBPs (SREBP-1a, SREBP-1c, and SREBP-2) in relation to various gastrointestinal tumours. The search utilised both MeSH terms and free-text keywords, with the review being current as of July 2024. By examining the relationship between the aberrant regulation of SREBP molecular subtypes and the progression of gastrointestinal tumours, we offer new insights into their significant role in cancer biology and their potential as therapeutic targets in cancer treatment.
Biochemical background of SREBPs
SREBPs are integral to the regulation of lipid homeostasis, glycogenesis, and the biosynthesis of fatty acids, triglycerides, and cholesterol. In addition to functioning as transcription factors, they also control the expression of enzymes involved in endogenous cholesterol synthesis, fatty acid synthesis, triacylglycerol synthesis, and phospholipid synthesis. Moreover, as members of the bHLH-Zip transcription factor family, SREBPs possess basic helix-loop-helix-leucine zip structures, which are similar to those observed in their gene products [12].
In general, the SREBP precursors (each possessing approximately 1150 amino acids) are classified into the following three domains [13, 14]: (1) an NH2 amino acid terminal domain that binds DNA, including the bHLH-Zip motif, which modulates the DNA-binding and dimerisation activities of mature SREBP transcription factors; (2) a short ring of approximately 30 amino acids between the two hydrophobic transmembrane segments that extends into the cavity of the endoplasmic reticulum, which is known as the membrane binding region; and (3) an approximately 590 amino acid COOH-terminal domain that regulates SREBP localisation and translocation within the cell (Fig. 2).
Fig. 2.
Structural diagram of different SREBP molecules (SREBP-1a, SREBP-1c, and SREBP-2) along with documented types of post-translational modifications
Currently, the SREBP family of proteins consists of two genes (SREBP1 and SREBP2), which are translated into three SREBP isoforms known as SREBP-1a, SREBP-1c, and SREBP-2 [15]. Among these isoforms, SREBP-1a and SREBP-1c are both products of the SREBP1 gene. However, due to alternative splicing at the transcription initiation site, the NH2-terminal domain of SREBP-1a is 24 amino acids longer than that of SREBP-1c [16]. Consequently, these isoforms exhibit functional differences. Both SREBP-1a and SREBP-1c are involved in regulating fatty acid synthesis genes, whereas SREBP-2 regulates cholesterol synthesis genes [17–20]. Moreover, SREBPs are differentially distributed in mice and humans, with SREBP-1c most commonly expressed in the liver, white adipose tissue, adrenal gland and brain [21]. In contrast, SREBP-1a and SREBP-2 are predominantly expressed in cell lines, the nervous system and intestinal tissues [22, 23]. Unlike SREBP-1a, SREBP-1c possesses four unique amino acids rather than 28 amino acids. Therefore, SREBP-1c has a shorter N-terminal sequence, and its transcriptional activity is weaker [24, 25]. Furthermore, SREBP-2 possesses a robust activation domain analogous to that of SREBP-1a, whereas the shorter SREBP-1c isoform exhibits a comparatively weaker transcriptional activation domain than SREBP-1a [26].
Transcriptional regulation of SREBPs
To monitor sterol levels in the ER, two membrane-based proteins are involved in the SREBP pathway, including SREBP cleavage activation protein (SCAP) and insulin-inducing gene protein (INSIG) [27]. Sterol-sensing domains are present in SCAP molecules. The SCAP confines SREBPs to the endoplasmic reticulum when sterol levels are high in the cell; however, it escorts them to the Golgi apparatus when sterol levels are low [14]. Moreover, INSIG (as a regulatory protein) binds to the SCAP/SREBP complex and participates in the regulation of SREBP molecular function [28].
Notably, SREBPs synthesised in the ER are in their precursor form of 125 kDa, and enzyme processing is required for the precursor to become the mature form of 68 kDa, with the mature enzyme playing a role in lipid metabolism. Specifically, a precursor SREBP known as pSREBP is initially synthesised and subsequently inserted into the ER membrane, where it binds to the COOH terminal region of the SCAP [29]. This process also involves INSIG family proteins, which retain SREBPs in the ER along with cholesterol [30]. As a nuclear SREBP (nSREBP), the NH2 terminal domain activates transcription by binding to nonpalindrosterol sterol response elements (SREs) in multiple target gene promoters/enhancers [13] (Fig. 1). The ACLY, ACC, FASN, and SCD genes (among others) regulated by SREBPs are highly expressed in cancer tissues compared with nontumour tissues and play important roles in cancer cell growth, metastasis, and survival. In summary, the modulation of the transcriptional activation process of SREBPs or the regulation of their downstream target molecules may represent promising therapeutic strategies in clinical settings.
Fig. 1.
Transcriptional activation diagram of SREBPs molecule
PTMs in SREBPs
Significantly, the regulation of SREBPs'transcriptional activity is also influenced by posttranslational modifications (PTMs) (Fig. 2). Most studies have focused on SREBP-2 and SREBP-1a, which are sterol-dependent regulators of posttranslational proteolysis.
Protein phosphorylation, the most prevalent form of posttranslational modification, regulates a diverse array of biological processes. SREBP-1 is also regulated by phosphorylation. A recent study revealed that SREBP-1 is activated during cell transformation in Ras/PI3K mutants and that a reduction in SREBP-1 in cancer cells leads to decreased proliferation [31]. In addition, insulin-induced phosphorylation of SREBP-1c enhances transport from the endoplasmic reticulum (ER) to the Golgi and proteolytic processing [32]. For example, glycogen synthase kinase 3 directly phosphorylates Ser443 on SREBP-2 to mediate FBW7-induced ubiquitination and degradation [33]. Similarly, when nuclear SREBP-1a is phosphorylated at sites S430 and T426, FBW7 is able to interact with it and promotes its degradation and ubiquitination. Furthermore, FBW7 degrades nuclear SREBP-2 and SREBP-1c.
Transforming growth factor-β activated kinase 1 (TAK1), which is an inflammatory signalling protein, binds and phosphorylates full-length SREBPs in the ER or Golgi bodies in the cytoplasm, thus further reducing their DNA-binding capacity and weakening their transcription [34]. Via the ubiquitin–proteasome pathway, SREBPs are rapidly degraded in the nucleus. Additionally, the glycogen synthetase kinase-3B phosphorylates serine and/or threonine residues near the ubiquitination site in SREBP-1 and SREBP-2, which enhances their ubiquitination [35].
As a type II PRMT enzyme, protein arginine methyltransferase 5 (PRMT5) modifies target proteins through SDMA to cause tumour progression. Mass spectrometry has revealed that PRMT5 can symmetrically dimethylate mature SREBP1 (mSREBP1a, 68 kDa) at the R321 site, thereby increasing its transcriptional activity and stabilising SREBP-1a [36]. Methylated stable SREBP-1a enhances lipid synthesis and promotes the proliferation of cancer cells both in living organisms and under laboratory conditions. Conversely, AMPK prevents TDG-mediated DNA demethylation of the SREBP-1 promoter, thus indicating that TDG regulated by insulin or metformin could be a promising therapeutic target for cancers linked to type 2 diabetes [37].
SREBPs can undergo modification by a different protein known as small ubiquitin-like modifier (SUMO). The process of SUMOylation, which affects transcription factors such as SREBPs, generally reduces the ability of these factors to activate transcription [38, 39]. Mitsumi et al. demonstrated that SREBP-2 is modified by SUMO-2 or SUMO-3 molecules at Lys 464 and that SUMOylation reduces SREBP transcriptional activity by recruiting a corepressor complex containing HDAC3 [40]. Similarly, two major SUMO sites (Lys 123 and Lys 418) were identified in SREBP-1a. The absence of one or two SUMOylation sites also increases the transactivation capacity of SREBP-responsive promoters [41].
SREBP-1a and SREBP-2 can undergo acetylation mediated by the histone acetyltransferases known as p300 and CREB-binding protein (CBP), and this process is contingent upon the presence of the N-terminal p300/CBP binding domain. Further studies have revealed that acetylation affects the stability of K324 and K333 in the DNA-binding domain of SREBP-1a, thereby regulating their transcriptional activities [42]. Conversely, sirtuin-1 (SIRT1), which is a member of the deacetylase family, deacetylates SREBP-2, thereby reducing the abundance of SREBP-2 in the nucleus [43]. SREBP-1 has also been identified as a substrate for neddylation by UBC12. In addition to increasing the stability of SREBP-1, neddylation reduces ubiquitination, which promotes the aggressive characteristics of HCC and breast cancer [44].
The SREBPs in hepatocellular carcinoma
Hepatocellular carcinoma (HCC) represents one of the most prevalent liver malignancies worldwide and is characterised by a poor prognosis and a dearth of potent and efficacious treatment modalities. Between 2010 and 2021, the global incidence and mortality rates of liver cancer increased by approximately 25%, with hepatitis B virus (HBV) persisting as the predominant cause of liver cancer-related morbidity and mortality worldwide [45]. Concurrently, nonalcoholic fatty liver disease (NAFLD) has emerged as a significant public health concern in Asia, with a prevalence rate of 34%; additionally, the incidence of NAFLD-associated HCC is increasing [46]. In the liver, SREBPs are involved in the pathogenesis of nonalcoholic fatty liver disease, nonalcoholic steatohepatitis, hepatitis, and liver cancer. Moreover, SREBPs serve as central regulators in numerous physiological and pathophysiological cellular processes, thereby influencing both transcriptional and posttranscriptional regulation [17]. Numerous studies have demonstrated that de novo adipogenesis (which is regulated by the transcription factor SREBP-1) plays a crucial role in the progression of HCC [47, 48].
In cancer cells, genes and enzymes involved in lipid metabolism are significantly altered. By controlling the transcription of genes involved in lipid biosynthesis and absorption, SREBPs play crucial roles in regulating lipid metabolism in HCC [49]. ATP1A1/CA2, which has been identified as the target molecule of bufalin, exhibits significant antitumour activity against hepatocellular carcinoma. Bufalin exerts its effects by modulating the ATP1A1/CA2 axis, thereby leading to the downregulation of the SREBP-1/FASN/ACLY pathway in liver cancer [50]. This regulatory mechanism results in the inhibition of lipogenesis and tumourigenesis. Phosphorylation, which is recognised as the most prevalent type of posttranslational modification, is crucial in the pathogenesis and progression of cancer. Comprehensive investigations by Xu et al. have revealed that the phosphorylation of cytoplasmic phosphoenolpyruvate carboxykinase 1 (PCK1) at Ser90, as well as INSIG1 at Ser207 and INSIG2 at Ser151, is positively associated with the nuclear accumulation of SREBP-1 in HCC samples [51]. Moreover, this phosphorylation is significantly correlated with poor prognosis in HCC patients. Furthermore, the interaction between pyruvate kinase M2 (PKM2) and nuclear SREBP-1a facilitates the phosphorylation of SREBP-1a at threonine 59 residues [52]. This SREBP-1a/PKM2 complex is posited to play a significant role in the pathogenesis of hepatocellular carcinoma. The rapid growth of HCC creates an oxygen-poor tumour environment. Additionally, EFNA3 enhances HCC cell self-renewal, proliferation, and migration, with EphA2 acting as a crucial downstream mediator. The Ephrin-A3/EphA2 axis aids in SREBP-1 maturation and can regulate HCC cell metabolism under hypoxia [53]. Moreover, SLC25A47, which is a downregulated carrier protein in HCC mitochondria, affects energy metabolism by influencing the mitochondrial NAD+-dependent deacetylase known as SIRT3. The loss of SLC25A47 decreases mitochondrial NAD+ levels, thereby reducing SIRT3 activity and promoting HCC progression via the SIRT3-AMPKα-SREBPs pathway, which regulates lipid metabolism [54].
SREBP-1 is upregulated in hepatic carcinoma tissues, and its overexpression is correlated with unfavourable clinicopathological characteristics. The knockdown of SREBP-1 has been shown to inhibit the proliferation, migration, and invasion of hepatocellular carcinoma cells [55]. Additionally, peroxidase 5 (Prx5) functions as an activator of AMP-activated protein kinase (AMPK), thus leading to the downregulation of SREBP-1 expression [56]. This regulatory mechanism can mitigate hepatic steatosis induced by excessive energy intake, thereby serving as a protective factor in the context of fatty liver diseases. Moreover, circPRKAA1, which is a circular RNA (circRNA) originating from the α1 subunit of AMPK, has the capacity to selectively recruit mSREBP-1, thereby increasing its transcription, enhancing fatty acid synthesis, and facilitating tumour growth [57]. Moreover, the membrane-bound transcription factor site-1 protease inhibitor known as PF-429242 induces autophagy-dependent cell death in HCC cells. Additionally, SREBP-1a/c are able to mitigate the cytotoxic effects induced by PF-429242 (to a certain extent) [58].
Rapidly proliferating cancer cells fulfil their lipid and cholesterol requirements by augmenting the uptake of dietary lipids or by reprogramming intracellular lipid biosynthesis. Hyperglycaemia induces the production of reactive oxygen species (ROS), which subsequently increase the nuclear translocation of SREBP-1 through a series of signalling pathways, thereby promoting the progression of hepatocellular carcinoma [59]. Conversely, the inhibition of SREBP-1 activation not only impedes aerobic glycolysis in HCC cells but also augments the antitumour efficacy of radiofrequency ablation in xenograft tumours [60]. Therefore, the targeting of SREBP-1 activation may represent a promising therapeutic strategy for the treatment of advanced HCC. Additionally, several cytokines have been implicated in regulating hepatocellular cancer development and prognosis. The oncogene pleiotrophin has been demonstrated to upregulate the expression of SREBP-1c and adipogenic genes, thereby playing a critical role in HCC proliferation and metastasis [61]. Moreover, hepatoma-derived growth factor (HDGF) functions as a coactivator in SREBP-1-mediated lipogenic gene transcription. The coexpression of SREBP-1 and nuclear HDGF is associated with poor prognoses in HCC patients [62]. Additionally, ficolin 3 (FCN3) is a constituent of the complement system that functions as a recognition molecule targeting pathogens within the lectin pathway. FCN3 has the capacity to directly induce inactivation of insulin receptor β, thereby inhibiting the expression of SREBP-1c and promoting ferroptosis in hepatocellular carcinoma [63]. Drug resistance poses a significant challenge in treating HCC. Sorafenib (SFN), which is a drug used for advanced HCC, experiences resistance due to metabolic dysregulation in HCC cells, including increased mTOR activation and disrupted SREBP-1c inhibition [64]. Energy deficiency in susceptible HepG2 cells inhibits mTOR and SREBP-1c, thus contributing to SFN resistance and poor clinical outcomes. Furthermore, the inhibition of SREBP-1c may also help in treating tamoxifen-induced hepatocyte steatosis [65].
Recent proteomics research has indicated that key proteins involved in maintaining cholesterol homeostasis (such as SREBP-2) are significantly upregulated in the early stages of HCC [66]. NS5ABP37, which is a novel hepatitis C virus nonstructural protein (NS)5A-associated binding protein, has been observed to inhibit the promoter activities of both SREBP-2 and SREBP-1c [67]. The downregulation of these proteins at both the mRNA and protein levels has demonstrated antitumour effects. Pan et al. demonstrated that METTL3 facilitates the m6A modification of SREBP-2 cleavage-activating protein mRNA, thereby increasing its translation and subsequently activating cholesterol biosynthesis, which promotes HCC progression [68]. Concurrently, sapienate metabolism in cancer cells is regulated by mTOR and SREBPs. Similar to the well-characterised desaturase SCD1, fatty acid desaturase 2 (FADS2) serves as a target for both SREBP-1 and SREBP-2. The expression of FADS2 mRNA and protein can be modulated via mTOR signalling, thereby influencing cancer progression [69]. Emodin, which is an active component of Chinese herbal medicine, has been demonstrated to reduce cholesterol biosynthesis by inhibiting SREBP-2 transcriptional activity in HCC cells [70]. Additionally, the modulation of SIRT6 levels in HCC cells has been shown to influence SREBP activity, which is specifically achieved by inhibiting both SREBP-1 and SREBP-2, thereby limiting lipogenesis and reducing fibrosis [71]. Under typical physiological conditions, large tumour suppressor kinase 2 (LATS2) tumour suppressors function as regulators of SREBP activity, thereby maintaining cholesterol and lipid homeostasis. The downregulation of LATS2 results in the activation of SREBP-2 and subsequent accumulation of excess cholesterol [72]. Moreover, Liang et al. reported that the haploinsufficient tumour suppressor known as apoptosis-stimulating protein of p53-2 (ASPP2) interacts with SREBP-2 within the nucleus, thereby inhibiting the transcriptional activation of genes in the mevalonate pathway by SREBP-2 [73]. Intrahepatic cholangiocarcinoma (ICC) constitutes a significant component of primary liver cancer, and its high recurrence rate poses substantial challenges for clinical management. Recent research has indicated that elevated expression of myelin and lymphocyte protein 2 (MAL2) in ICC is correlated with unfavourable prognostic outcomes. The underlying mechanism of this effect involves the promotion of lipid accumulation within ICC cells via the stabilisation of EGFR membrane localisation and the subsequent activation of the PI3K/AKT/SREBP-1 signalling axis [74].
NAFLD has been identified as an early indicator of cirrhosis and HCC. Nonetheless, lipid metabolism is pivotal in regulating the balance between lipogenesis and fatty acid oxidation, which are processes that are advantageous for the therapeutic management of NAFLD.
Yang et al. demonstrated that exosomes derived from human umbilical cord mesenchymal stem cells (MSC-exs) inhibit fatty acid synthesis via calcium/calmodulin-dependent protein kinase 1 (CAMKK1) and SREBP-1c while simultaneously enhancing fatty acid oxidation via PPARα [75]. This dual regulatory mechanism leads to a reduction in lipid accumulation in an AMP-activated protein kinase (AMPK)-dependent manner. Similarly, SREBP-1 also directly upregulates histone deacetylase 8 (HDAC8), thereby increasing carcinogenic activity in NAFLD-associated hepatocellular carcinoma [76]. SREBP-1 is a key activator of apoptosis antagonising transcription factor (AATF) in the context of NAFLD, and therapeutic agents that reduce SREBP-1 activity can also reduce the risk of HCC development by reducing AATF [77]. Conversely, the phytochemical known as tetrahydropalmatine (THP) has been shown to ameliorate hepatic steatosis in NAFLD by activating the AMPK-SREBP-1c-Sirt1 signalling axis, thereby enhancing lipid oxidation [78]. In addition, octyl gallate (OG), which is a widely utilised food antioxidant, promotes the accumulation of lipid droplets in HepG2 cells via the modulation of SREBP-1c gene expression, thus potentially contributing to the pathogenesis of NAFLD when it is employed as a food additive [79]. Furthermore, ursodeoxycholic acid (UDCA) within bile acids has been demonstrated to mitigate lipid metabolism disorders induced by oleic acid (OA) via the inhibition of the AKT/mTOR/SREBP-1 signalling pathway [80]. In the hepatic tissues of obese patients with NAFLD, the intake of polyunsaturated fatty acids is correlated with increased levels of SREBP-1c, thereby potentially initiating a pro-lipogenic program that exacerbates hepatic lipid accumulation in these individuals [81]. In conclusion, the data derived from cellular and animal models, as well as clinical specimens, indicate that SREBPs play a critical role in the progression of pre-nonalcoholic fatty liver disease (pre-NAFLD). The targeting of SREBPs via various signalling pathways (including CAMKK1 and Sirt1) is anticipated to mitigate the metabolic transition from NAFLD to hepatocellular carcinoma.
In summary, the development and progression of HCC are linked to SREBPs (particularly SREBP-1 and SREBP-2), which increase tumour growth by activating lipid production pathways such as FASN/ACLY and cholesterol synthesis pathways such as SCAP-m6A modification (Table 1). Notably, during the transformation from NAFLD to HCC, SREBPs play crucial roles in mediating lipid metabolism imbalance via the coordination of the AMPK/mTOR/Sirt1 signalling pathways. Targeted intervention strategies, including the use of AMPK activators and METTL3 inhibitors, have demonstrated efficacy in disrupting this process. Future approaches that involve the multipathway regulation of SREBPs, along with their upstream and downstream effector molecules such as HDAC8 and AATF, may offer promising avenues for overcoming drug resistance in HCC treatment and achieving precise metabolic intervention.
Table 1.
Related mechanisms of SREBPs molecular types in hepatocellular carcinoma
| Type of Cancer | Targets of SREBPs | Key molecules/mechanism pathways | Main cell lines/sample sizes | Reference |
|---|---|---|---|---|
| Hepatocellular carcinoma | SREBP-1 | Bufalin/ATP1A1/CA2 | PLC/PRF/5, Huh6, Huh7 | [50] |
| SREBP-1 | PCK1/INSIG1/INSIG2 | Huh7, Hep3B, SNU-398/HL7702, THLE-2/CHL-1, U87, H1993/90 human HCC tissue samples | [51] | |
| SREBP-1 | Ephrin-A3/EphA2 axis | Huh7, PLC/PRF/5, MHCC97L, HepG2/97 paired HCC/non-tumourous liver samples | [53] | |
| SREBP-1 | Prx5/circPRKAA1 | HepG2 | [56] | |
| SREBP-1 | ROS | HepG2, Hep3B, HuH-7 | [59] | |
| SREBP-1 | HDGF | HepG2, HEK293 T,7721 | [62] | |
| SREBP-1 | HDAC8 | LO2, HepG2, Bel-7404, PLC5/24 paired NAFLD-associated HCC samples | [76] | |
| SREBP-1 | AATF | HepG2, Hepa1–6, Huh7, QGY-7703/18–20 human samples | [77] | |
| SREBP-1 | UDCA/AKT/mTOR | L02 | [80] | |
| SREBP-1a | PKM2 | HEK293 T, HepG2, SW480, A549, MCF7/75 patient samples | [52] | |
| SREBP-1c | Pleiotrophin | HepG2, Huh-7, LX-2, LO2/80 HCC patient tissue samples | [61] | |
| SREBP-1c | FCN3/insulin receptor β | Huh7, HepG2, HEK-293 T, SM-386, YY-8103, MHCC97-H/133 human samples | [63] | |
| SREBP-1c | SFN, Tamoxifen | HepG2, Huh7, Hep3B/126 patients who received SFN therapy | [64] | |
| SREBP-1c | Exosomes derived from MSC-ex | L02, HFL1, AML12, HEK293 T | [75] | |
| SREBP-1c | THP/AMPK/SIRT1 | SMMC-7721, BEL-7402 | [78] | |
| SREBP-1c | OG | HepG2 | [79] | |
| SREBP-1a/c | PF-429242 | PLC/PRF/5, HepG2 | [58] | |
| SREBP-2 | METTL3 | HKCI2, HKCI10, RIL-175, Hepa1-6 | [68] | |
| SREBP-2 | Emodin | Hep3B, HepG2, SK-HEP-1, Huh7, PLC/PRF5 | [70] | |
| SREBP-2 | LATS2 | HepG2 | [72] | |
| SREBP-2 | ASPP2/MVA | HEK293 T, HCC-LM3, HepG2, Huh-7 | [73] | |
| SREBP-1/2 | FADS2/mTOR | HUH7, U87 MG glioblastoma cells, U2OS osteosarcoma cells | [69] | |
| SREBP-1/2 | SIRT6 | HepG2, Huh-7 | [71] | |
| SREBP-1/2 | SLC25A47/SIRT3/AMPK | L02, HepG2, Huh7, Hela, HEK293 T | [54] | |
| SREBP-1c/2 | NS5ABP37 | HepG2, L02/10 HCC cases of different stages and 10 normal pancreatic tissue samples | [67] |
SREBPs in colorectal cancer
Lipid metabolic reprogramming is thought to be an important aspect of tumour metabolism, thus suggesting that SREBPs may be effective targets for cancer therapy. Many studies have demonstrated that SREBPs are elevated in many cancers, including colorectal cancer (CRC); moreover, their high expression has been observed to accelerate cancer progression. According to statistical data from 2020, CRC ranks as the second leading cause of cancer-related mortality in the United States. Additionally, both the incidence and mortality rates of CRC are increasing annually in Asian countries, with a pronounced increase being observed in East Asia [82]. Protein tyrosine phosphatase receptor type O (PTPRO) functions as a tumour suppressor across various cancer types. The silencing of PTPRO facilitates AKT/mTOR phosphorylation, which subsequently induces the expression of SREBP-1 and its downstream target gene ACC1, thereby increasing fatty acid synthesis and lipid accumulation; these effects may further contribute to the tumourigenesis and progression of CRC [83]. Aspirin enhances the susceptibility of CRC cells to ferroptosis by inhibiting the PI3K/AKT/mTOR signalling pathway, which subsequently suppresses SREBP-1/SCD1-mediated lipogenesis [84]. Furthermore, the genetic ablation of SREBP-1 or SCD1 increases the sensitivity of cancer cells to ferroptosis induction. In alignment with prior research, the selective PI3Kδ inhibitor known as TYM-3–98 has been demonstrated to inhibit CRC by inducing ferroptosis via the suppression of SREBP1 and its downstream targets. Notably, the overexpression of SREBP-1 diminishes the efficacy of TYM-3–98. These findings indicate that TYM-3–98 exhibits promise as a potential therapeutic agent for targeting CRC [85]. Moreover, brexpiprazole has been shown to markedly suppress the proliferation, lipogenesis, and cell cycle progression of colorectal cancer cells. The underlying mechanism of these effects likely involves the inhibition of colorectal cancer cell proliferation and adipogenesis via the AMPK/SREBP-1 signalling pathway [86].
SREBP-1 expression is elevated in colorectal tumour tissues, particularly at the invasive front and tumour budding sites, in contrast to normal tissues. This differential expression implies a potential role for SREBP-1 in the invasion and metastasis of colorectal cancer. Mechanistically, SREBP-1 facilitates endothelial cell angiogenesis through the augmentation of ROS and enhances the invasive and metastatic capabilities of colon cancer cells by upregulating matrix metalloproteinase 7 (MMP7) via p65 phosphorylation [87]. Liver metastasis often leads to death in advanced colorectal cancer patients. Yuan et al. reported that the cyclic RNA known as circNOLC1 (which is linked to the oxidative pentose phosphate pathway) interacts with zinc-alpha2-glycoprotein (AZGP1) to activate mTOR/SREBP-1 signalling [88]. This effect correspondingly induces glucose‐6‐phosphate dehydrogenase (G6PD), thus further activating the pathway and playing a crucial role in colorectal cancer liver metastasis.
Radiation resistance (both acquired and intrinsic) poses a significant challenge in treating CRC. High cholesterol levels enhance this resistance effect. External cholesterol intake increases radioresistance in CRC cells; however, the inhibition of the SREBP-1/FASN pathway reduces cholesterol synthesis and increases radiation-induced cell death in CRC [89]. The knockdown of SREBP1 has been shown to significantly inhibit the proliferation, migration, and invasion of CRC cells by controlling lipid metabolism and enhancing their apoptotic sensitivity [90]. This observation is corroborated by the study conducted by Gao et al., which demonstrated that SREBP-1 can inhibit apoptosis. Furthermore, the inhibition of SREBP1 overexpression also results in the suppression of apoptosis. The underlying mechanism of this effect likely involves the regulatory effect of SREBP-1 on the caspase-7 apoptosis signalling pathway and the concurrent inhibition of poly(ADP-ribose) polymerase-1 (PARP1) cleavage, thereby mitigating the apoptosis of colon cancer cells [91]. The knockdown of SREBP1 or SREBP2 significantly hampers cell proliferation and tumour growth; alters cell metabolism by reducing glycolysis, mitochondrial respiration, and fatty acid oxidation; and diminishes the tumour-forming ability of colon cancer cells while also reducing cancer stem cell gene expression [92]. Moreover, ZNFX1 antisense RNA 1 (ZFAS1), which is overexpressed in various cancers, functions as an oncogene by regulating miRNAs and is vital for CRC proliferation and metastasis. In CRC cells, ZFAS1 enhances fat metabolism reprogramming by binding to poly(A)-binding protein 2 (PABP2), thus facilitating the interaction of PABP2 with SREBP-1 [93]. This effect stabilises SREBP-1 mRNA and activates its downstream genes SCD1 and FASN, thus promoting CRC progression. Additionally, acyl-CoA synthetase 3 (ACSL3) is an enzyme that converts long-chain fatty acids into fatty acyl-CoA esters for lipid synthesis and fatty acid oxidation. Transforming growth factor beta 1 (TGFβ1) can upregulate ACSL3 via the SREBP-1 pathway, thereby leading to energy metabolic reprogramming in CRC cells [94]. This process supports EMT and metastasis and provides energy for cancer cell invasion and distant metastasis.
Increasing evidence also suggests that SREBP-1 may play an important role in tumour progression and malignancy [95]. SREBP-1 promotes CRC cell growth, migration, and invasion by upregulating snail family transcriptional repressor 1 (SNAIL) and accelerating EMT. This process relies on the oncogene known as c-Myc, with which SREBP-1 interacts to increase the binding of c-Myc to the SNAIL promoter, thereby increasing SNAIL levels and enhancing EMT and cell migration [96]. Drug resistance continues to pose a significant challenge to the prognoses and survival rates of patients with colorectal cancer. SREBP-1 is also overexpressed in CRC patient samples that are resistant to chemotherapy. SREBP-1 negatively regulates caspase-7 and sensitises CRC cells to chemotherapy [97]. Conversely, low SREBP-1 expression in CRC samples is associated with elevated caspase-7 levels. Therefore, the targeting of SREBP-1 may increase the sensitivity of colorectal cancer cells to gemcitabine. Similarly, when CRC cells are cultured in adipocyte-conditioned media, SREBP-1 levels increase via the Akt and p70S6K pathways, thereby reducing sensitivity to 5-FU and increasing drug resistance [98]. Similarly, mutations in PIK3 CA-mutated tumours lead to sustained activation of the PI3K/Akt signalling pathway, which correspondingly promotes the nuclear accumulation of SREBP-1 and the transcription of apolipoprotein A5 (APOA5) [99]. This transcriptional activation can further stimulate the peroxisome proliferator-activated receptor γ (PPARγ) signalling pathway, thus resulting in a reduction in the production of reactive oxygen species and consequently enhancing the chemoresistance of CRC.
Oridonin, which is a diterpenoid compound isolated from Rabdosia rubescens, exhibits notable anti-proliferative activity against cancer. Empirical evidence has demonstrated that oridonin effectively suppresses the expression of FAS and SREBP1 mRNAs and proteins in human colorectal cancer cells, thereby exerting its anticancer effects [100]. Berberine, which is traditionally utilised in Chinese medicine, has been demonstrated to inhibit the growth and lipogenesis of colon cancer xenografts via a SCAP-dependent mechanism [101]. This inhibition occurs via the suppression of SREBP-1 activation and SCAP expression, thus ultimately leading to the downregulation of associated lipogenic enzymes. Furthermore, the natural cyclic peptide known as RA-XII suppresses the expression of the novel fatty acid-synthesising proteins FASN and SCD by downregulating SREBP-1 expression, thereby inhibiting the proliferation and metastasis of colorectal tumours [102]. Ilexgenin A, which is known for its substantial lipid-lowering properties, has been demonstrated to reduce the expression of SREBP-1 and its target genes in the colon tissues of colorectal cancer model mice [103]. This compound may facilitate the early prevention of colorectal cancer by modulating lipid metabolism via the HIF1α/SREBP-1 pathway.
B7H3, which is a member of the B7 cosignaling molecule family, enhances resistance to ferroptosis by modulating SREBP-2-mediated cholesterol metabolism [104]. Both the administration of exogenous cholesterol and the application of the SREBP-2 inhibitor betulin effectively counteracted the influence of B7H3 on iron-induced apoptosis in colorectal cancer cells. Furthermore, the interaction between YAP-mediated zinc finger MYND-type containing 8 (ZMYND8) and SREBP-2 facilitates enhancer-promoter interactions and promotes the recruitment of the mediator complex, thereby upregulating genes associated with the mevalonate (MVA) pathway, which correspondingly increases the susceptibility of patients to colorectal cancer [105]. Yu et al. reported that increased SREBP-2 and cholesterol due to lower atypical protein kinase C (PKC) levels are crucial for managing metaplasia and producing aggressive cell subpopulations in serrated tumours [106]. Thus, the targeting of cholesterol biosynthesis could serve as a chemoprophylaxis strategy. Similarly, polyunsaturated fatty acids such as docosahexaenoic acid and oleic acid may enhance CRC progression by activating SREBP-2 and inhibiting its target genes, which can occur independently of cholesterol levels and ER stress [107]. Sorting nexin 10 (SNX10) is crucial for maintaining intestinal stem cells (ISCs). Moreover, the deletion of SNX10 increases SREBP-2 activation, increases cholesterol production and aids in ISC recovery for mucosal healing [108]. Additionally, studies have demonstrated that the interaction between polyamine metabolism and the cholesterol synthesis pathway via SREBP-2 regulates colorectal cancer cell growth and malignancy [109]. Curcumin, a constituent of traditional Chinese medicine, induces activation of the transient receptor potential cation channel subfamily A member 1 (TRPA1), leading to enhanced calcium influx. This process subsequently upregulates peroxisome proliferator-activated receptor gamma (PPARγ) while concurrently downregulating the SP-1/SREBP-2/NPC1L1 pathway, thereby ultimately inhibiting cholesterol absorption [110].
SREBPs are central to CRC metabolic reprogramming and promote malignancy via lipid synthesis, cholesterol balance, and signalling activities. The targeting of SREBP and effectors such as SCD1 and NPC1L1 can hinder metabolic adaptation, EMT, and drug resistance (Table 2). Moreover, the combination of natural compounds with synthetic inhibitors could offer new CRC treatment strategies. Future research should explore the interactions of SREBPs with the tumour microenvironment to enable precise metabolic interventions and personalised therapies.
Table 2.
Related mechanisms of SREBPs molecular types in colorectal cancer
| Type of Cancer | Targets of SREBPs | Key molecules/mechanism pathways | Main cell lines/sample sizes | Reference |
|---|---|---|---|---|
| Colorectal cancer | SREBP-1 | PTPRO/AKT/mTOR | DLD1, HCT116, SW480, SW620, LoVo, RKO/276 patients with CRC | [83] |
| SREBP-1 | Aspirin | DLD-1, HCT 116, HepG2, PANC-1, AGS | [84] | |
| SREBP-1 | TYM-3–98 | HCT 116, LoVo, SW620 | [85] | |
| SREBP-1 | Brexpiprazole | HCT116 | [86] | |
| SREBP-1 | MMP7/AMPK | HT29, SW620, HUVEC/60 primary carcinoma specimens | [87] | |
| SREBP-1 | circNOLC1, AZGP1/mTOR, G6PD | HCT116, LoVo, LS174 T | [88] | |
| SREBP-1 | FASN | HT‐29, HCT‐8 | [89] | |
| SREBP-1 | Caspase-7, PARP1 | HT29, SW620/30 primary carcinoma specimens | [91] | |
| SREBP-1 | ZFAS1, PABP2 | SW480/30 CRC and adjacent tissues, blood samples were obtained from 218 patients with CRC and 238 healthy people | [93] | |
| SREBP-1 | TGFβ1, ACSL3 | HCT116, LoVo | [94] | |
| SREBP-1 | c-Myc, SNAIL/EMT | SW480, HT29, HCT116, SW620, LS180/44 patient fresh CRC and matched adjacent non-cancerous tissues | [96] | |
| SREBP-1 | Akt/p70S6K, 5-FU | DLD-1, SW480 | [98] | |
| SREBP-1 | APOA5, PPARγ | HCT15, HCT116, SW480, SW620, LOVO | [99] | |
| SREBP-1 | Oridonin | SW480, SW620 | [100] | |
| SREBP-1 | Berberine, SCAP | DLD-1, Caco-2 | [101] | |
| SREBP-1 | RA-XII | HCT116 | [102] | |
| SREBP-1 | Ilexgenin A/HIF1α | HT29, HCT 116 | [103] | |
| SREBP-2 | B7H3 | NCM460, HCT116, HT29, SW480, SW620 | [104] | |
| SREBP-2 | ZMYND8/MVA | HEK293 T, HCT116, DLD1 | [105] | |
| SREBP-2 | PKC | HEK293 T, HCT116/483 CRC tissues from patients | [106] | |
| SREBP-2 | SNX10 | HCT116 | [108] | |
| SREBP-2 | Curcumin/SP-1, NPC1L1 | Caco-2 | [110] |
SREBPs in pancreatic ductal adenocarcinoma
The prevalence and mortality of pancreatic ductal adenocarcinoma (PDAC) vary throughout the world, particularly in Asia. The Global Burden of Disease Study indicated a notable increase in pancreatic cancer cases and deaths from 1990 to 2017, especially in high-income areas [111]. In Southeast Asia, both the incidence and mortality of this type of cancer are increasing and are projected to significantly increase by 2050 [112]. The initiation of abnormally activated lipid metabolic reprogramming plays a key role in the development of PDAC, which can develop into a deadly malignant tumour with a high probability of recurrence and distant metastasis [69]. The expression of oxysterol binding protein-associated protein 5 (ORP5) is correlated with increased invasiveness and poor prognoses in patients with pancreatic cancer. ORP5 upregulates the expression of SREBP-2 and activates downstream genes associated with sterol response elements [113]. This regulatory mechanism influences the expression levels of tumour suppressor genes, thereby promoting the invasion and proliferation of pancreatic cancer cells. Peroxisome proliferator-activated receptor-gamma coactivator-1 (PGC-1) alpha and beta play key roles in regulating intermediate metabolism. Hannes et al. reported that PGC-1α increases SREBP-1c and SREBP-2 expression in beta cells, thus impairing insulin secretion by increasing UCP2 transcription [114]. Conversely, a reduction of PGC-1β decreases the impact of SREBP-1c on granulophilin transcription, thereby enhancing glucose-stimulated insulin release. Moreover, sodium medroglutarate is enzymatically converted to farnesyl pyrophosphate (FPP), which is a precursor of cholesterol and sterols, and to geranyl pyrophosphate. These lipids are involved in tumourigenesis and progression. Dysregulation of the MVA pathway may activate SCAP and transcription factors such as SREBPs and HIF-1 [115]. As inhibitors of the MVA pathway, statins have the ability to reduce cancer risk and/or treat cancer by interfering with essential cell functions such as proliferation and differentiation. Kubota et al. demonstrated that synergistic apoptosis can be achieved when PDAC cells are treated with both statins and SREBP inhibitors and that this effect is dependent on geranylgeranyl diphosphate (GGPP), which is produced in the MVA pathway [116].
Diabetes is a significant risk factor for PDAC, with over 50% of individuals who are diagnosed with PDAC also presenting with diabetes. Research has demonstrated that elevated insulin secretion is correlated with the induction of SREBP-1c and augmented lipid synthesis in murine islets cultured under high-glucose conditions [117]. Type 2 diabetes mellitus (T2DM) is characterised by impaired glucose-stimulated insulin secretion (GSIS) due to glycolipid toxicity. The downregulation of SREBP-1c partially reverses GSIS injury induced by palmitate [118]. According to a recent report, transcolloin-2 expression is higher in PDAC tissues than in normal tissues [119]. Additionally, insulin activates SREBP-1-mediated transcription in PDAC cells to upregulate transgelin-2 expression. Moreover, the aberrant expression of cellular retinoic acid binding protein II (CRABP-II) enhances the stability of SREBP-1c mRNA and upregulates the downstream genes of SREBP-1c, thereby facilitating the absorption and accumulation of cholesterol in lipid rafts [120]. This increased accumulation of cholesterol in lipid rafts subsequently promotes AKT survival signalling and contributes to PDAC resistance. In addition, in the context of pancreatic cancer, liver X receptor (LXR) and SREBP-1 levels are notably lower in tumour tissues than in adjacent tissues. Furthermore, triptonide can further reduce LXR and SREBP-1 levels in tumours by activating p53 and causing DNA strand breaks, thus resulting in cell death [121].
In summary, the targeting of SREBPs and related pathways (such as MVA and CRABP-II) can effectively hinder metabolic adaptation and resistance (Table 3). Additionally, the combination of statins with natural compounds (such as triptonide) could enhance the effectiveness of PDAC treatment. Upcoming studies should concentrate on the dynamic interactions between SREBPs and the metabolic environments of tumours, including those associated with diabetes and cholesterol metabolism, to create targeted interventions and improve clinical treatments.
Table 3.
Related mechanisms of SREBPs molecular types in pancreatic ductal adenocarcinoma
| Type of Cancer | Targets of SREBPs | Key molecules/mechanism pathways | Main cell lines/sample sizes | Reference |
|---|---|---|---|---|
| Pancreatic ductal adenocarcinoma | SREBP-1 | Triptonide/P53 | Patu8988, Panc-1/100 primary PC tumour tissues and 80 paired normal pancreatic tissues | [121] |
| SREBP-1c | CRABP-II | BxPC3, Capan-1, Panc-1, Panc10.05, HEK293 T/12 primary PDACs samples | [120] | |
| SREBP-2 | ORP5 | Capan-1, Capan-2, Hs700 T, MiaPaCa2, Panc-1 | [113] | |
| SREBP-1c/2 | PGC-1α/β/UCP2 | INS-1E | [114] | |
| SREBPs | MVA/GGPP | Pa02c, Pa03c, Pa16c, Pa20c, HEK293 T | [116] |
SREBPs in gastric cancer
The prevalence and mortality rates associated with gastric cancer (GC) in Asia represent a significant public health concern. Empirical studies have indicated that Asia is among the regions with the highest incidence and mortality rates of gastric cancer, particularly regarding East Asian countries such as China, Japan, and South Korea [122]. Moreover, lipid metabolism plays a crucial role in the pathogenesis of GC, which is complex and diverse. SREBP-1c is activated in human gastric cancer and promotes the expression of a series of fatty acid synthesis-related genes, such as SCD1 and FASN [123]. SREBP-1c gene knockdown can restore the migration and invasion defects of gastric cancer cells. A similar increase in FASN and SREBP-1c protein and gene expression in gastric adenocarcinoma (GA) patients is associated with the overexpression of the hypoxia-inducing factor known as HIF-1α. The activation of SREBP-1c by HIF-1α can facilitate the upregulation of FASN in GA, which has diagnostic and prognostic significance [124]. The transcriptional cofactor RPRD1B (regulation of nuclear pre-mRNA domain containing 1B) has been identified as being a promoter of lymph node metastasis in gastric cancer both in vivo and in vitro. The underlying mechanism of this phenomenon likely involves the upregulation of fatty acid uptake and synthesis via transcriptional activation of the c-Jun/c-Fos pathway and subsequent activation of the c-Jun/c-Fos/SREBP1 axis [125]. Consequently, the targeting of this pathway represents a promising therapeutic strategy for the treatment of gastric cancer. Indeed, many types of drugs are also involved in treating gastric cancer via the lipid metabolism pathway related to SREBPs. For example, apatinib, which is a vascular endothelial growth factor 2 (VEGFR2) inhibitor, has antiangiogenic and anticancer effects. Specifically, it induces lipid peroxidation via the antioxidant enzyme glutathione peroxidase 4 (GPX4), which is mediated by SREBP-1a, thereby negatively regulating iron-induced apoptosis in GC cells, including those cells resistant to multiple drugs [126]. Natural compounds such as flavonoids, alkaloids and saponins can also inhibit the de novo synthesis of lipids in GC by regulating SREBP-1, reducing lipid accumulation levels, and subsequently inhibiting the occurrence and development of GC [127]. Statins can inhibit the MVA pathway, trigger apoptosis in cancer cells in vitro, and reduce tumour incidence in animals. Research has demonstrated that statins induce apoptosis in HGT-1 human gastric cancer cells, which is regulated by SREBP-1 and SREBP-2, with caspase-7 being activated under mevalonic acid restriction [128]. In summary, SREBPs are key regulators of lipid reprogramming in gastric cancer and promote malignant traits by integrating hypoxia, metastasis pathways, and drug responses. The targeting of SREBPs and their downstream effectors (such as FASN and GPX4) can effectively inhibit metabolic adaptation and metastasis (Table 4).
Table 4.
Related mechanisms of SREBPs molecular types in gastric cancer
| Type of Cancer | Targets of SREBPs | Key molecules/mechanism pathways | Main cell lines/sample sizes | Reference |
|---|---|---|---|---|
| Gastric cancer | SREBP-1 | RPRD1B/c-Jun/c-Fos/SREBP1 axis | HGC27, AGS/42 paired fresh specimens of tumour and adjacent non-tumour tissues | [125] |
| SREBP-1a | Apatinib | MGC803, AGS, GES-1 | [126] | |
| SREBP-1c | HIF-1α | AGS/112 GA patients and 156 control cases | [124] | |
| SREBP-1/2 | MVA pathway | HGT-1, HCT116, HepG2, L50 | [128] |
SREBPs in oesophageal cancer
Oesophageal cancer is linked to risk factors such as smoking and obesity. The Global Burden of Cancer report estimated 511,054 new cases of oesophageal cancer and 445,391 deaths worldwide in 2022, with approximately 75% of the cases occurring in Asia [129]. Obesity is frequently linked to the dysregulation of lipid metabolism, whereas cancer cells are commonly characterised by metabolic reprogramming. Tumour cells require high levels of cholesterol during development to perform membrane biosynthesis and undergo rapid proliferation. In oesophageal tumours, SREBP1 overexpression is associated with lower survival rates and levels of miR142-5p, which is a tumour suppressor [130]. The inhibition or silencing of SREBP-1 in oesophageal cancer cells resulted in significant reductions in tributary characteristics, decreased expression of EMT markers, and increased expression of miR-142-5p. Additionally, the targeting of SREBP-1 molecules with the SREBP-1 inhibitor fatostatin can significantly inhibit tumourigenesis both in vitro and in vivo. Recently, AKT-dependent PCK1 was demonstrated to promote SREBP1-dependent adipogenesis. In patients with oesophageal squamous cell carcinoma (ESCC), PCK1 pS90, INSIG1 pS207/INSIG2 pS151 or nuclear SREBP-1 levels were observed to be positively associated with a poor prognosis [131]. A combined expression value for these indicators demonstrates better prognostic value for oesophageal cancer compared with a single protein expression value. By disrupting redox homeostasis, trace zinc can increase the level of intracellular Fe2+ and prevent ferroptosis in oesophageal cancer cells. A mechanism explaining this change could be related to the inhibition of p-AMPK and the induction of SREBP1 and SCD1, which increase the production of monounsaturated fatty acids exhibiting antiferroptotic characteristics [132]. In addition, the upregulation of SREBP1 in ESCC tumours and cells is closely related to proliferation and metastasis. As a result of SREBP1 depletion, SCD1 and Wnt/β-catenin signalling pathway activity are inhibited, thereby diminishing in vivo metastasis [133]. In ESCC, lysophosphatidylcholine acyltransferase 1 (LPCAT1) is highly expressed and reprograms cholesterol metabolism. Additionally, LPCAT1 regulates cholesterol synthesis by two mechanisms: the activation of PI3K to facilitate SP1/SREBP2 nuclear entry and the activation of EGFR to reduce INSIG-1 expression and promote SREBP-1 nuclear entry [134]. Moreover, the reduction of SREBP-2 expression in oesophageal cancer cells decreases their survival, migration, and colony formation. This scenario occurs via the interaction of SREBP-2 with c-Myc and the joint activation of HMG-CoA reductase (HMGCR) expression [135]. Thus, a focus on SREBPs and their effectors (such as SCD1 and HMGCR) can collectively impede metabolic adaptation and metastasis, whereas interventions such as fatostatin and zinc metabolism provide potential methods to combat drug resistance. Future research should focus on the interaction between SREBPs and activities in the tumour microenvironment (such as redox balance and EGFR/PI3K signalling) to develop multitarget therapies for overcoming clinical treatment challenges in oesophageal cancer (Table 5).
Table 5.
Related mechanisms of SREBPs molecular types in oesophageal cancer
| Type of Cancer | Targets of SREBPs | Key molecules/mechanism pathways | Main cell lines/sample sizes | Reference |
|---|---|---|---|---|
| oesophageal cancer | SREBP-1 | miR-142-5p | OE21, OE33 | [130] |
| SREBP-1 | PCK1/INSIG1/INSIG2 | 200 ESCC patient samples | [131] | |
| SREBP-1 | Trace zinc | EC109, EC9706, Het-1 A/31 pairs of ESCC patients and healthy control | [132] | |
| SREBP-1 | SCD1/Wnt/β-catenin signaling pathway | TE-1, ECA-109, KYSE-150/77 paired ESCC tumour and adjacent normal tissue samples | [133] | |
| SREBP-1/2 | LPCAT1/PI3K/EGFR | HEEC, EC9706, TE1/Tumour tissue from 154 patients with ESCC | [134] | |
| SREBP-2 | c-Myc/HMGCR | Het-1 A, HEK 293 T, Eca109, KYSE150/samples from 52 patients(oesophageal cancer and adjacent non-tumour samples) | [135] |
Strengths and limitations
The exploration of SREBPs in GI tumour metabolism has unearthed a wealth of knowledge, yet it also confronts certain challenges.
One of the significant strengths lies in the comprehensive understanding of SREBPs' role in lipid metabolic reprogramming across multiple GI tumours. In HCC, SREBPs intricately regulate lipid-related genes, with SREBP-1 playing a pivotal part in tumour progression by modulating pathways like FASN/ACLY for lipid production. In CRC, SREBPs are involved in various processes such as angiogenesis, metastasis, and drug resistance. This in-depth understanding of their functions provides a solid foundation for developing targeted therapies.
Furthermore, the identification of various intervention strategies targeting SREBPs represents a significant advancement in the field. Small molecule inhibitors have the capacity to directly inhibit SREBP activation, thereby disrupting lipid synthesis associated with tumour promotion. Natural compounds, with their multi-target regulatory capabilities, can modulate pathways related to SREBPs, providing a more comprehensive approach to treatment. Additionally, statins, through their interference with the mevalonate pathway, not only impact lipid metabolism but also demonstrate potential in augmenting the effectiveness of other anticancer therapies.
However, the current body of research is not without limitations. A major drawback is the heavy reliance on cellular and animal models for mechanistic investigations. These models, while valuable, cannot fully recapitulate the complexity of the human tumour microenvironment. The differences in metabolic regulation, immune-tumour interactions, and genetic backgrounds between models and human patients may lead to inaccurate predictions of treatment efficacy in the clinical setting.
Another limitation is the incomplete understanding of the functional divergence among SREBP subtypes. SREBP-1a, SREBP-1c, and SREBP-2 demonstrate distinct functions and expression patterns across various gastrointestinal cancers. Their differential roles in lipid metabolism, such as the variable influence of SREBP-1c on fatty acid and cholesterol metabolism in different tumours, necessitate more comprehensive investigation. This gap in understanding impedes the development of highly specific and effective targeted therapies. Clinically, the utility of SREBPs as reliable biomarkers is challenged. Although their expression levels correlate with patient prognosis, the presence of metabolic disorders, such as NAFLD and T2DM, complicates this relationship. Furthermore, certain SREBP-targeting intervention strategies may induce unintended side effects in normal tissues, thereby limiting their clinical applicability.
In conclusion, although substantial advancements have been made in understanding the role of SREBPs in gastrointestinal tumour metabolism, it is imperative to address existing limitations by employing more sophisticated research models, conducting comprehensive subtype analyses, and enhancing biomarker validation. These steps are crucial for the successful translation of research findings into effective clinical therapies.
Summary
As central regulators of lipid metabolic reprogramming, SREBPs exhibit a multifaceted cancer-promoting mechanism across various gastrointestinal solid tumours. The role of SREBPs extends beyond facilitating the synthesis of fatty acids and cholesterol. Furthermore, a dynamic regulatory network is established by integrating metabolic signals with classical oncogenic pathways, such as the AKT/mTOR, Wnt/β-catenin, and epithelial-mesenchymal transition (EMT) pathways. For example, in HCC, SREBP-1 enhances nuclear translocation through its interaction with PKM2 and the Ephrin-A3/EphA2 axis. In CRC, SREBP-1 collaborates with c-Myc to activate the SNAIL/EMT pathway. Additionally, in ESCC, LPCAT1 activates SREBP-1/2 via a dual pathway to reprogram cholesterol metabolism. The shared metabolism-signalling interactions in various cancers suggest that the targeting of SREBPs could be widely effective against tumours. Intervention strategies include small molecule inhibitors such as fatostatin, which block SREBP activation; natural compounds such as lamonidin and berberine, which inhibit SREBP via pathways such as the AMPK/mTOR pathway; and statins, which disrupt the substrate supply via the MVA pathway. The combination of SREBP targeting with other metabolic targets (such as SCD1 or HMGCR) or treatments such as chemotherapy/radiotherapy has demonstrated promise in overcoming resistance, such as 5-FU resistance in CRC and iron death resistance in ESCC.
Nevertheless, the current body of research is subject to notable limitations. First, a substantial portion of the mechanistic evidence is derived from cellular or animal models, which possess metabolic microenvironments that are distinct from those of human tumours. This discrepancy may lead to an overestimation of the therapeutic efficacy of the targeting of a single pathway. Second, the functional heterogeneity among members of the SREBP family (including SREBPs-1a/1c-2) that is observed across various cancer types remains inadequately understood. For example, SREBP-1c predominantly influences fatty acid synthesis in GC, whereas it is more closely linked to cholesterol metabolism in PDAC, thus necessitating precise targeting strategies to differentiate between the subtypes. From a clinical translation perspective, although SREBP expression is significantly correlated with prognosis, it lacks specificity as a biomarker, particularly in patients with metabolic disorders such as NAFLD and T2DM. Furthermore, certain intervention strategies (such as cholesterol deprivation) may induce off-target toxicity in normal tissues. Thus, the advancement of metabolic targeted therapy necessitates the interdisciplinary integration of metabolic biology, pharmaceutical engineering, and clinical oncology to overcome existing limitations and facilitate a paradigm shift from"pathway inhibition"to"metabolic microenvironment reshaping".
Acknowledgements
Not applicable.
Abbreviations
- GI
Gastrointestinal tumour
- SREBPs
Sterol regulatory element-binding proteins
- SCAP
SREBP cleavage activation protein
- INSIG
Insulin-inducing gene protein
- SREs
Nonpalindrosterol response elements
- ACLY
ATP citrate lyase
- ACC
Acetyl-CoA carboxylase
- FASN
FA synthase
- SCD
Stearoyl-CoA desaturase
- ER
Endoplasmic reticulum
- Golgi
Golgi apparatus
- FBW7
F-Box And WD Repeat Domain Containing 7
- TAK1
Transforming growth factor-β activated kinase 1
- PRMT5
Protein arginine methyltransferase 5
- SUMO
Small ubiquitin-like modifier
- HDAC3
Histone deacetylase 3
- CBP
CREB-binding protein
- SIRT1
Sirtuin-1
- UBC12
Ubiquitin Conjugating Enzyme E2 M
- HCC
Hepatocellular Carcinoma
- ATP1A1/CA2
ATPase Na+/K+ Transporting Subunit Alpha 1/ Carbonic Anhydrase 2
- PCK1
Phosphoenolpyruvate carboxykinase 1
- PKM2
Pyruvate kinase M2
- EFNA3
Ephrin A3
- EphA2
Ephrin Type-A Receptor 2
- SLC25A47
Solute Carrier Family 25 Member 47
- SIRT3
Sirtuin 3
- Prx5
Peroxidase 5
- AMPK
AMP-activated protein kinase
- ROS
Reactive oxygen species
- HDGF
Hepatoma-derived growth factor
- FCN3
Ficolin 3
- SFN
Sorafenib
- NS5ABP37
Novel hepatitis C virus non-structural protein 5A-associated binding protein
- LATS2
Large Tumour Suppressor Kinase 2
- NAFLD
Non-alcoholic fatty liver disease
- MSC-ex
Exosomes derived from human umbilical cord mesenchymal stem cells
- CAMKK1
Calcium/calmodulin-dependent protein kinase 1
- AATF
Apoptosis antagonizing transcription factor
- THP
The phytochemical tetrahydropalmatine
- UDCA
Ursodeoxycholic acid
- OA
Oleic acid
- OG
Octyl gallate
- CRC
Colorectal cancer
- PTPRO
Protein tyrosine phosphatase receptor Type O
- MMP7
Matrix metalloproteinase 7
- AZGP1
Alpha-2-Glycoprotein 1, Zinc-Binding
- ZFAS1
ZNFX1 antisense RNA1
- ACSL3
Acyl-CoA synthetase 3
- 5-FU
5-Fluorocrail
- PPARγ
Peroxisome Proliferator Activated Receptor Gamma
- B7H3
B7 homolog 3 protein
- ZMYND8
Zinc finger MYND-type containing 8
- MVA
Mevalonate
- PKC
Atypical protein kinase C
- ISCs
Intestinal stem cells
- SNX10
Sorting nexin 10
- TRPA1
Transient receptor potential ankyrin 1
- NPC1L1
Niemann-Pick C1-like 1
- ORP5
Oxysterol binding protein-associated protein 5
- PGC-1
Peroxisome proliferator-activated receptor-gamma co-activator-1
- UCP2
Uncoupling Protein 2
- FPP
Farnesyl pyrophosphate
- GGPP
Geranylgeranyl diphosphate
- PDAC
Pancreatic ductal adenocarcinoma
- T2DM
Type 2 diabetes mellitus
- GSIS
Glucose-stimulated insulin secretion
- CRABP-II
Cellular retinoic acid binding protein II
- LXRs
Liver X receptors
- HIF-1α
Hypoxia Inducible Factor 1 Subunit Alpha
- RPRD1B
Regulation Of Nuclear Pre-MRNA Domain Containing 1B
- VEGFR2
Vascular endothelial growth factor 2
- GPX4
Glutathione Peroxidase 4
- GC
Gastric cancer
- LPCAT1
Lysophosphatidylcholine Acyltransferase 1
- EGFR
Epidermal Growth Factor Receptor
- HMGCR
3-Hydroxy-3-Methylglutaryl-CoA Reductase
- ESCC
Oesophageal squamous cell carcinoma
- ORP5
Oxysterol binding related proteins 5
Authors’ contributions
HWT conceived the idea and wrote the manuscript. YTZ, DNZ and MJG drew the diagrams and tables. XY and XW (corresponding author) revised and edited the manuscript. The author(s) read and approved the final manuscript.
Funding
This study was supported by the National Natural Science Foundation of China (No. 32170910) and the Projects from Social Development of Zhenjiang (SH2024024).
Data availability
No datasets were generated or analysed during the current study.
Declarations
Ethics approval and consent to participate
Not applicable.
Consent for publication
Not applicable.
Competing interests
The authors declare no competing interests.
Footnotes
Publisher’s Note
Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional affiliations.
Contributor Information
Xiao Yuan, Email: yuanxiao07seven@163.com.
Xu Wang, Email: jsdxwx@126.com.
References
- 1.Caini S, Del Riccio M, Vettori V, Raimondi S, Assedi M, Vignati S, Bonaccorsi G, Cattaruzza MS, Bellerba F, Vagnoni G, et al. The prognostic impact of quitting smoking at or around diagnosis on the survival of patients with gastrointestinal cancers: a systematic literature review. Cancers (Basel). 2022;14(16):3857. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 2.Ding L, Zhao Y, Dang S, Wang Y, Li X, Yu X, Li Z, Wei J, Liu M, Li G. Circular RNA circ-DONSON facilitates gastric cancer growth and invasion via NURF complex dependent activation of transcription factor SOX4. Mol Cancer. 2019;18(1):45. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 3.Kang MH, Choi H, Oshima M, Cheong JH, Kim S, Lee JH, Park YS, Choi HS, Kweon MN, Pack CG, et al. Estrogen-related receptor gamma functions as a tumor suppressor in gastric cancer. Nat Commun. 2018;9(1):1920. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 4.Wu Q, Qian W, Sun X, Jiang S. Small-molecule inhibitors, immune checkpoint inhibitors, and more: FDA-approved novel therapeutic drugs for solid tumors from 1991 to 2021. J Hematol Oncol. 2022;15(1):143. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 5.German NJ, Yoon H, Yusuf RZ, Murphy JP, Finley LW, Laurent G, Haas W, Satterstrom FK, Guarnerio J, Zaganjor E, et al. PHD3 loss in cancer enables metabolic reliance on fatty acid oxidation via deactivation of ACC2. Mol Cell. 2016;63(6):1006–20. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 6.Wu D, Yang Y, Hou Y, Zhao Z, Liang N, Yuan P, Yang T, Xing J, Li J. Increased mitochondrial fission drives the reprogramming of fatty acid metabolism in hepatocellular carcinoma cells through suppression of Sirtuin 1. Cancer Commun (Lond). 2022;42(1):37–55. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 7.Zhu Y, Lin X, Zhou X, Prochownik EV, Wang F, Li Y. Posttranslational control of lipogenesis in the tumor microenvironment. J Hematol Oncol. 2022;15(1):120. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 8.Porstmann T, Santos CR, Griffiths B, Cully M, Wu M, Leevers S, Griffiths JR, Chung YL, Schulze A. SREBP activity is regulated by mTORC1 and contributes to Akt-dependent cell growth. Cell Metab. 2008;8(3):224–36. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 9.Alizadeh J, Kavoosi M, Singh N, Lorzadeh S, Ravandi A, Kidane B, Ahmed N, Mraiche F, Mowat MR, Ghavami S. Regulation of Autophagy Via Carbohydrate And Lipid Metabolism In Cancer. Cancers (Basel). 2023;15(8):2195. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 10.Currie E, Schulze A, Zechner R, Walther TC, Farese RV Jr. Cellular fatty acid metabolism and cancer. Cell Metab. 2013;18(2):153–61. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 11.Cheng C, Geng F, Cheng X, Guo D. Lipid metabolism reprogramming and its potential targets in cancer. Cancer Commun (Lond). 2018;38(1):27. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 12.Goldstein JL, Rawson RB, Brown MS. Mutant mammalian cells as tools to delineate the sterol regulatory element-binding protein pathway for feedback regulation of lipid synthesis. Arch Biochem Biophys. 2002;397(2):139–48. [DOI] [PubMed] [Google Scholar]
- 13.Horton JD, Goldstein JL, Brown MS. SREBPs: activators of the complete program of cholesterol and fatty acid synthesis in the liver. J Clin Invest. 2002;109(9):1125–31. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 14.Weber LW, Boll M, Stampfl A. Maintaining cholesterol homeostasis: sterol regulatory element-binding proteins. World J Gastroenterol. 2004;10(21):3081–7. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 15.Harada N, Yonemoto H, Yoshida M, Yamamoto H, Yin Y, Miyamoto A, Hattori A, Wu Q, Nakagawa T, Nakano M, et al. Alternative splicing produces a constitutively active form of human SREBP-1. Biochem Biophys Res Commun. 2008;368(3):820–6. [DOI] [PubMed] [Google Scholar]
- 16.Eberle D, Hegarty B, Bossard P, Ferre P, Foufelle F. SREBP transcription factors: master regulators of lipid homeostasis. Biochimie. 2004;86(11):839–48. [DOI] [PubMed] [Google Scholar]
- 17.Brown MS, Goldstein JL. The SREBP pathway: regulation of cholesterol metabolism by proteolysis of a membrane-bound transcription factor. Cell. 1997;89(3):331–40. [DOI] [PubMed] [Google Scholar]
- 18.Horton JD, Shah NA, Warrington JA, Anderson NN, Park SW, Brown MS, Goldstein JL. Combined analysis of oligonucleotide microarray data from transgenic and knockout mice identifies direct SREBP target genes. Proc Natl Acad Sci U S A. 2003;100(21):12027–32. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 19.Horton JD, Shimomura I, Ikemoto S, Bashmakov Y, Hammer RE. Overexpression of sterol regulatory element-binding protein-1a in mouse adipose tissue produces adipocyte hypertrophy, increased fatty acid secretion, and fatty liver. J Biol Chem. 2003;278(38):36652–60. [DOI] [PubMed] [Google Scholar]
- 20.Osborne TF, Espenshade PJ. Evolutionary conservation and adaptation in the mechanism that regulates SREBP action: what a long, strange tRIP it’s been. Genes Dev. 2009;23(22):2578–91. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 21.Deng X, Cagen LM, Wilcox HG, Park EA, Raghow R, Elam MB. Regulation of the rat SREBP-1c promoter in primary rat hepatocytes. Biochem Biophys Res Commun. 2002;290(1):256–62. [DOI] [PubMed] [Google Scholar]
- 22.Ferre P, Foufelle F. SREBP-1c transcription factor and lipid homeostasis: clinical perspective. Horm Res. 2007;68(2):72–82. [DOI] [PubMed] [Google Scholar]
- 23.Kim JH, Ong WY. Localization of the transcription factor, sterol regulatory element binding protein-2 (SREBP-2) in the normal rat brain and changes after kainate-induced excitotoxic injury. J Chem Neuroanat. 2009;37(2):71–7. [DOI] [PubMed] [Google Scholar]
- 24.Shimano H, Horton JD, Shimomura I, Hammer RE, Brown MS, Goldstein JL. Isoform 1c of sterol regulatory element binding protein is less active than isoform 1a in livers of transgenic mice and in cultured cells. J Clin Invest. 1997;99(5):846–54. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 25.Toth JI, Datta S, Athanikar JN, Freedman LP, Osborne TF. Selective coactivator interactions in gene activation by SREBP-1a and -1c. Mol Cell Biol. 2004;24(18):8288–300. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 26.Huang G, Huang PJ, Jiang CS, Liu ZD. Variations during developmental process of AchE isoenzyme in susceptible strain and dipterex-resistant strain of the Culex pipiens pallens Coq. Yi Chuan Xue Bao. 1988;15(2):129–37. [PubMed] [Google Scholar]
- 27.Yan R, Cao P, Song W, Qian H, Du X, Coates HW, Zhao X, Li Y, Gao S, Gong X, et al. A structure of human Scap bound to Insig-2 suggests how their interaction is regulated by sterols. Science. 2021;371(6533):eabb2224. [DOI] [PubMed] [Google Scholar]
- 28.Rong S, Xia M, Vale G, Wang S, Kim CW, Li S, McDonald JG, Radhakrishnan A, Horton JD. DGAT2 inhibition blocks SREBP-1 cleavage and improves hepatic steatosis by increasing phosphatidylethanolamine in the ER. Cell Metab. 2024;36(3):617–29 e617. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 29.Shechter I, Dai P, Huo L, Guan G. IDH1 gene transcription is sterol regulated and activated by SREBP-1a and SREBP-2 in human hepatoma HepG2 cells: evidence that IDH1 may regulate lipogenesis in hepatic cells. J Lipid Res. 2003;44(11):2169–80. [DOI] [PubMed] [Google Scholar]
- 30.Tiscione SA, Vivas O, Ginsburg KS, Bers DM, Ory DS, Santana LF, Dixon RE, Dickson EJ. Disease-associated mutations in Niemann-Pick type C1 alter ER calcium signaling and neuronal plasticity. J Cell Biol. 2019;218(12):4141–56. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 31.Ricoult SJ, Yecies JL, Ben-Sahra I, Manning BD. Oncogenic PI3K and K-Ras stimulate de novo lipid synthesis through mTORC1 and SREBP. Oncogene. 2016;35(10):1250–60. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 32.Yellaturu CR, Deng X, Cagen LM, Wilcox HG, Mansbach CM 2nd, Siddiqi SA, Park EA, Raghow R, Elam MB. Insulin enhances post-translational processing of nascent SREBP-1c by promoting its phosphorylation and association with COPII vesicles. J Biol Chem. 2009;284(12):7518–32. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 33.Sundqvist A, Bengoechea-Alonso MT, Ye X, Lukiyanchuk V, Jin J, Harper JW, Ericsson J. Control of lipid metabolism by phosphorylation-dependent degradation of the SREBP family of transcription factors by SCF(Fbw7). Cell Metab. 2005;1(6):379–91. [DOI] [PubMed] [Google Scholar]
- 34.Morioka S, Sai K, Omori E, Ikeda Y, Matsumoto K, Ninomiya-Tsuji J. TAK1 regulates hepatic lipid homeostasis through SREBP. Oncogene. 2016;35(29):3829–38. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 35.Sato R. SREBPs: protein interaction and SREBPs. FEBS J. 2009;276(3):622–7. [DOI] [PubMed] [Google Scholar]
- 36.Liu L, Zhao X, Zhao L, Li J, Yang H, Zhu Z, Liu J, Huang G. Arginine Methylation of SREBP1a via PRMT5 Promotes De Novo Lipogenesis and Tumor Growth. Cancer Res. 2016;76(5):1260–72. [DOI] [PubMed] [Google Scholar]
- 37.Zhao Q, Lin X, Wang G. Targeting SREBP-1-Mediated Lipogenesis as Potential Strategies for Cancer. Front Oncol. 2022;12:952371. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 38.Bies J, Markus J, Wolff L. Covalent attachment of the SUMO-1 protein to the negative regulatory domain of the c-Myb transcription factor modifies its stability and transactivation capacity. J Biol Chem. 2002;277(11):8999–9009. [DOI] [PubMed] [Google Scholar]
- 39.Hirano Y, Yoshida M, Shimizu M, Sato R. Direct demonstration of rapid degradation of nuclear sterol regulatory element-binding proteins by the ubiquitin-proteasome pathway. J Biol Chem. 2001;276(39):36431–7. [DOI] [PubMed] [Google Scholar]
- 40.Arito M, Horiba T, Hachimura S, Inoue J, Sato R. Growth factor-induced phosphorylation of sterol regulatory element-binding proteins inhibits sumoylation, thereby stimulating the expression of their target genes, low density lipoprotein uptake, and lipid synthesis. J Biol Chem. 2008;283(22):15224–31. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 41.Hirano Y, Murata S, Tanaka K, Shimizu M, Sato R. Sterol regulatory element-binding proteins are negatively regulated through SUMO-1 modification independent of the ubiquitin/26 S proteasome pathway. J Biol Chem. 2003;278(19):16809–19. [DOI] [PubMed] [Google Scholar]
- 42.Giandomenico V, Simonsson M, Gronroos E, Ericsson J. Coactivator-dependent acetylation stabilizes members of the SREBP family of transcription factors. Mol Cell Biol. 2003;23(7):2587–99. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 43.Walker AK, Yang F, Jiang K, Ji JY, Watts JL, Purushotham A, Boss O, Hirsch ML, Ribich S, Smith JJ, et al. Conserved role of SIRT1 orthologs in fasting-dependent inhibition of the lipid/cholesterol regulator SREBP. Genes Dev. 2010;24(13):1403–17. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 44.Heo MJ, Kang SH, Kim YS, Lee JM, Yu J, Kim HR, Lim H, Kim KM, Jung J, Jeong LS, et al. UBC12-mediated SREBP-1 neddylation worsens metastatic tumor prognosis. Int J Cancer. 2020;147(9):2550–63. [DOI] [PubMed] [Google Scholar]
- 45.Tan EY, Danpanichkul P, Yong JN, Yu Z, Tan DJH, Lim WH, Koh B, Lim RYZ, Tham EKJ, Mitra K, et al. Liver cancer in 2021: Global Burden of Disease study. J Hepatol. 2025;82(5):851–60. [DOI] [PubMed] [Google Scholar]
- 46.Yip TC, Lee HW, Chan WK, Wong GL, Wong VW. Asian perspective on NAFLD-associated HCC. J Hepatol. 2022;76(3):726–34. [DOI] [PubMed] [Google Scholar]
- 47.Voigt H, Vetter-Kauczok CS, Schrama D, Hofmann UB, Becker JC, Houben R. CD147 impacts angiogenesis and metastasis formation. Cancer Invest. 2009;27(3):329–33. [DOI] [PubMed] [Google Scholar]
- 48.Yahagi N, Shimano H, Hasegawa K, Ohashi K, Matsuzaka T, Najima Y, Sekiya M, Tomita S, Okazaki H, Tamura Y, et al. Co-ordinate activation of lipogenic enzymes in hepatocellular carcinoma. Eur J Cancer. 2005;41(9):1316–22. [DOI] [PubMed] [Google Scholar]
- 49.Li N, Li X, Ding Y, Liu X, Diggle K, Kisseleva T, Brenner DA. SREBP regulation of lipid metabolism in liver disease, and therapeutic strategies. Biomedicines. 2023;11(12):3280. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 50.Huang CJ, Zhang CY, Zhao YK, Wang D, Zhuang L, Qian L, Xie L, Zhu Y, Meng ZQ. Bufalin Inhibits Tumorigenesis and SREBP-1-Mediated Lipogenesis in Hepatocellular Carcinoma via Modulating the ATP1A1/CA2 Axis. Am J Chin Med. 2023;51(2):461–85. [DOI] [PubMed] [Google Scholar]
- 51.Xu D, Wang Z, Xia Y, Shao F, Xia W, Wei Y, Li X, Qian X, Lee JH, Du L, et al. The gluconeogenic enzyme PCK1 phosphorylates INSIG1/2 for lipogenesis. Nature. 2020;580(7804):530–5. [DOI] [PubMed] [Google Scholar]
- 52.Zhao X, Zhao L, Yang H, Li J, Min X, Yang F, Liu J, Huang G. Pyruvate kinase M2 interacts with nuclear sterol regulatory element-binding protein 1a and thereby activates lipogenesis and cell proliferation in hepatocellular carcinoma. J Biol Chem. 2018;293(17):6623–34. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 53.Husain A, Chiu YT, Sze KM, Ho DW, Tsui YM, Suarez EMS, Zhang VX, Chan LK, Lee E, Lee JM, et al. Ephrin-A3/EphA2 axis regulates cellular metabolic plasticity to enhance cancer stemness in hypoxic hepatocellular carcinoma. J Hepatol. 2022;77(2):383–96. [DOI] [PubMed] [Google Scholar]
- 54.Cheng L, Deepak R, Wang G, Meng Z, Tao L, Xie M, Chi W, Zhang Y, Yang M, Liao Y, et al. Hepatic mitochondrial NAD + transporter SLC25A47 activates AMPKalpha mediating lipid metabolism and tumorigenesis. Hepatology. 2023;78(6):1828–42. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 55.Li C, Yang W, Zhang J, Zheng X, Yao Y, Tu K, Liu Q. SREBP-1 has a prognostic role and contributes to invasion and metastasis in human hepatocellular carcinoma. Int J Mol Sci. 2014;15(5):7124–38. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 56.Kim MH, Seong JB, Huh JW, Bae YC, Lee HS, Lee DS. Peroxiredoxin 5 ameliorates obesity-induced non-alcoholic fatty liver disease through the regulation of oxidative stress and AMP-activated protein kinase signaling. Redox Biol. 2020;28:101315. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 57.Li Q, Yao H, Wang Y, Wu Y, Thorne RF, Zhu Y, Wu M, Liu L. circPRKAA1 activates a Ku80/Ku70/SREBP-1 axis driving de novo fatty acid synthesis in cancer cells. Cell Rep. 2022;41(8):111707. [DOI] [PubMed] [Google Scholar]
- 58.Lin JC, Liu TP, Chen YB, Yang PM. PF-429242 exhibits anticancer activity in hepatocellular carcinoma cells via FOXO1-dependent autophagic cell death and IGFBP1-dependent anti-survival signaling. Am J Cancer Res. 2023;13(9):4125–44. [PMC free article] [PubMed] [Google Scholar]
- 59.Athavale D, Chouhan S, Pandey V, Mayengbam SS, Singh S, Bhat MK. Hepatocellular carcinoma-associated hypercholesterolemia: involvement of proprotein-convertase-subtilisin-kexin type-9 (PCSK9). Cancer Metab. 2018;6:16. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 60.Zou XZ, Hao JF, Zhou XH. Inhibition of SREBP-1 activation by a novel small-molecule inhibitor enhances the sensitivity of hepatocellular carcinoma tissue to radiofrequency ablation. Front Oncol. 2021;11:796152. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 61.Bai PS, Xia N, Sun H, Kong Y. Pleiotrophin, a target of miR-384, promotes proliferation, metastasis and lipogenesis in HBV-related hepatocellular carcinoma. J Cell Mol Med. 2017;21(11):3023–43. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 62.Min X, Wen J, Zhao L, Wang K, Li Q, Huang G, Liu J, Zhao X. Role of hepatoma-derived growth factor in promoting de novo lipogenesis and tumorigenesis in hepatocellular carcinoma. Mol Oncol. 2018;12(9):1480–97. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 63.Yuan Y, Xu J, Jiang Q, Yang C, Wang N, Liu X, Piao HL, Lu S, Zhang X, Han L, et al. Ficolin 3 promotes ferroptosis in HCC by downregulating IR/SREBP axis-mediated MUFA synthesis. J Exp Clin Cancer Res. 2024;43(1):133. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 64.Narahara S, Watanabe T, Nagaoka K, Fujimoto N, Furuta Y, Tanaka K, Tokunaga T, Kawasaki T, Yoshimaru Y, Setoyama H, et al. Clusterin and related scoring index as potential early predictors of response to sorafenib in hepatocellular carcinoma. Hepatol Commun. 2022;6(5):1198–212. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 65.Zhao F, Xie P, Jiang J, Zhang L, An W, Zhan Y. The effect and mechanism of tamoxifen-induced hepatocyte steatosis in vitro. Int J Mol Sci. 2014;15(3):4019–30. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 66.Yang PM, Hong YH, Hsu KC, Liu TP. p38alpha/S1P/SREBP2 activation by the SAM-competitive EZH2 inhibitor GSK343 limits its anticancer activity but creates a druggable vulnerability in hepatocellular carcinoma. Am J Cancer Res. 2019;9(10):2120–39. [PMC free article] [PubMed] [Google Scholar]
- 67.Feng S, Han M, Zhou L, Wang Q, Li Z, Li Y, Lu H, Liu T, Ma Y, Liu S, et al. NS5ABP37 inhibits liver cancer by impeding lipogenesis and cholesterogenesis. Cancer Sci. 2017;108(1):12–22. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 68.Pan Y, Chen H, Zhang X, Liu W, Ding Y, Huang D, Zhai J, Wei W, Wen J, Chen D, et al. METTL3 drives NAFLD-related hepatocellular carcinoma and is a therapeutic target for boosting immunotherapy. Cell Rep Med. 2023;4(8):101144. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 69.Triki M, Rinaldi G, Planque M, Broekaert D, Winkelkotte AM, Maier CR, Janaki Raman S, Vandekeere A, Van Elsen J, Orth MF, et al. mTOR signaling and SREBP activity increase FADS2 expression and can activate sapienate biosynthesis. Cell Rep. 2020;31(12):107806. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 70.Kim YS, Lee YM, Oh TI, Shin DH, Kim GH, Kan SY, Kang H, Kim JH, Kim BM, Yim WJ, et al. Emodin Sensitizes Hepatocellular Carcinoma Cells to the Anti-Cancer Effect of Sorafenib through Suppression of Cholesterol Metabolism. Int J Mol Sci. 2018;19(10):3127. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 71.Xiang X, Ohshiro K, Zaidi S, Yang X, Bhowmick K, Vegesna AK, Bernstein D, Crawford JM, Mishra B, Latham PS, et al. Impaired reciprocal regulation between SIRT6 and TGF-beta signaling in fatty liver. FASEB J. 2022;36(6):e22335. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 72.Aylon Y, Gershoni A, Rotkopf R, Biton IE, Porat Z, Koh AP, Sun X, Lee Y, Fiel MI, Hoshida Y, et al. The LATS2 tumor suppressor inhibits SREBP and suppresses hepatic cholesterol accumulation. Genes Dev. 2016;30(7):786–97. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 73.Liang B, Chen R, Song S, Wang H, Sun G, Yang H, Jing W, Zhou X, Fu Z, Huang G, et al. ASPP2 inhibits tumor growth by repressing the mevalonate pathway in hepatocellular carcinoma. Cell Death Dis. 2019;10(11):830. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 74.Huang T, Cao H, Liu C, Sun X, Dai S, Liu L, Wang Y, Guo C, Wang X, Gao Y, et al. MAL2 reprograms lipid metabolism in intrahepatic cholangiocarcinoma via EGFR/SREBP-1 pathway based on single-cell RNA sequencing. Cell Death Dis. 2024;15(6):411. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 75.Yang F, Wu Y, Chen Y, Xi J, Chu Y, Jin J, Yan Y. Human umbilical cord mesenchymal stem cell-derived exosomes ameliorate liver steatosis by promoting fatty acid oxidation and reducing fatty acid synthesis. JHEP Rep. 2023;5(7):100746. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 76.Tian Y, Wong VW, Wong GL, Yang W, Sun H, Shen J, Tong JH, Go MY, Cheung YS, Lai PB, et al. Histone deacetylase HDAC8 promotes insulin resistance and beta-catenin activation in NAFLD-associated hepatocellular carcinoma. Cancer Res. 2015;75(22):4803–16. [DOI] [PubMed] [Google Scholar]
- 77.Kumar DP, Santhekadur PK, Seneshaw M, Mirshahi F, Uram-Tuculescu C, Sanyal AJ. A regulatory role of apoptosis antagonizing transcription factor in the pathogenesis of nonalcoholic fatty liver disease and hepatocellular carcinoma. Hepatology. 2019;69(4):1520–34. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 78.Yin X, Liu Z, Wang J. Tetrahydropalmatine ameliorates hepatic steatosis in nonalcoholic fatty liver disease by switching lipid metabolism via AMPK-SREBP-1c-Sirt1 signaling axis. Phytomedicine. 2023;119:155005. [DOI] [PubMed] [Google Scholar]
- 79.Lima KG, Schneider Levorse VG, Rosa Garcia MC, de Souza BB, Pasqualotto Costa B, Antunes GL, Luft C, Haute GV, Leal Xavier L, Donadio MVF, et al. Octyl gallate induces hepatic steatosis in HepG2 cells through the regulation of SREBP-1c and PPAR-gamma gene expression. EXCLI J. 2020;19:962–71. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 80.Hu J, Hong W, Yao KN, Zhu XH, Chen ZY, Ye L. Ursodeoxycholic acid ameliorates hepatic lipid metabolism in LO2 cells by regulating the AKT/mTOR/SREBP-1 signaling pathway. World J Gastroenterol. 2019;25(12):1492–501. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 81.Bissig-Choisat B, Alves-Bezerra M, Zorman B, Ochsner SA, Barzi M, Legras X, Yang D, Borowiak M, Dean AM, York RB, et al. A human liver chimeric mouse model for non-alcoholic fatty liver disease. JHEP Rep. 2021;3(3):100281. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 82.Siegel RL, Miller KD, Goding Sauer A, Fedewa SA, Butterly LF, Anderson JC, Cercek A, Smith RA, Jemal A. Colorectal cancer statistics, 2020. CA Cancer J Clin. 2020;70(3):145–64. [DOI] [PubMed] [Google Scholar]
- 83.Dai W, Xiang W, Han L, Yuan Z, Wang R, Ma Y, Yang Y, Cai S, Xu Y, Mo S, et al. PTPRO represses colorectal cancer tumorigenesis and progression by reprogramming fatty acid metabolism. Cancer Commun (Lond). 2022;42(9):848–67. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 84.Chen J, Lv S, Huang B, Ma X, Fu S, Zhao Y. Upregulation of SCD1 by ErbB2 via LDHA promotes breast cancer cell migration and invasion. Med Oncol. 2022;40(1):40. [DOI] [PubMed] [Google Scholar]
- 85.Zheng YN, Lou SY, Lu J, Zheng FL, Tang YM, Zhang EJ, Cui SL, Zhao HJ. Selective PI3Kdelta inhibitor TYM-3-98 suppresses AKT/mTOR/SREBP1-mediated lipogenesis and promotes ferroptosis in KRAS-mutant colorectal cancer. Cell Death Dis. 2024;15(7):474. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 86.Li T, Liu X, Long X, Li Y, Xiang J, Lv Y, Zhao X, Shi S, Chen W. Brexpiprazole suppresses cell proliferation and de novo lipogenesis through AMPK/SREBP1 pathway in colorectal cancer. Environ Toxicol. 2023;38(10):2352–60. [DOI] [PubMed] [Google Scholar]
- 87.Gao Y, Nan X, Shi X, Mu X, Liu B, Zhu H, Yao B, Liu X, Yang T, Hu Y, et al. SREBP1 promotes the invasion of colorectal cancer accompanied upregulation of MMP7 expression and NF-kappaB pathway activation. BMC Cancer. 2019;19(1):685. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 88.Yuan M, Zhang X, Yue F, Zhang F, Jiang S, Zhou X, Lv J, Zhang Z, Sun Y, Chen Z, et al. CircNOLC1 promotes colorectal cancer liver metastasis by interacting with AZGP1 and sponging miR-212-5p to regulate reprogramming of the oxidative pentose phosphate pathway. Adv Sci (Weinh). 2023;10(33):e2205229. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 89.Jin Y, Chen Z, Dong J, Wang B, Fan S, Yang X, Cui M. SREBP1/FASN/cholesterol axis facilitates radioresistance in colorectal cancer. FEBS Open Bio. 2021;11(5):1343–52. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 90.Qiu Z, Deng W, Hong Y, Zhao L, Li M, Guan Y, Su Y, Chen C, Shi Q, Yu J, et al. Biological behavior and lipid metabolism of colon cancer cells are regulated by a combination of sterol regulatory element-binding protein 1 and ATP citrate lyase. Onco Targets Ther. 2021;14:1531–42. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 91.Gao Y, Zhao Q, Mu X, Zhu H, Liu B, Yao B, Liu X, Xue W, Wang B, Liu S. SREBP1 promotes 5-FU resistance in colorectal cancer cells by inhibiting the expression of caspase7. Int J Clin Exp Pathol. 2019;12(3):1095–100. [PMC free article] [PubMed] [Google Scholar]
- 92.Wen YA, Xiong X, Zaytseva YY, Napier DL, Vallee E, Li AT, Wang C, Weiss HL, Evers BM, Gao T. Downregulation of SREBP inhibits tumor growth and initiation by altering cellular metabolism in colon cancer. Cell Death Dis. 2018;9(3):265. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 93.Wang H, Chen Y, Liu Y, Li Q, Luo J, Wang L, Chen Y, Sang C, Zhang W, Ge X, et al. The lncRNA ZFAS1 regulates lipogenesis in colorectal cancer by binding polyadenylate-binding protein 2 to stabilize SREBP1 mRNA. Mol Ther Nucleic Acids. 2022;27:363–74. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 94.Quan J, Cheng C, Tan Y, Jiang N, Liao C, Liao W, Cao Y, Luo X. Acyl-CoA synthetase long-chain 3-mediated fatty acid oxidation is required for TGFbeta1-induced epithelial-mesenchymal transition and metastasis of colorectal carcinoma. Int J Biol Sci. 2022;18(6):2484–96. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 95.Yao C, Su L, Shan J, Zhu C, Liu L, Liu C, Xu Y, Yang Z, Bian X, Shao J, et al. IGF/STAT3/NANOG/Slug signaling axis simultaneously controls epithelial-mesenchymal transition and stemness maintenance in colorectal cancer. Stem Cells. 2016;34(4):820–31. [DOI] [PubMed] [Google Scholar]
- 96.Zhai D, Cui C, Xie L, Cai L, Yu J. Sterol regulatory element-binding protein 1 cooperates with c-Myc to promote epithelial-mesenchymal transition in colorectal cancer. Oncol Lett. 2018;15(4):5959–65. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 97.Shen W, Xu T, Chen D, Tan X. Targeting SREBP1 chemosensitizes colorectal cancer cells to gemcitabine by caspase-7 upregulation. Bioengineered. 2019;10(1):459–68. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 98.Lee KC, Wu KL, Yen CK, Chen CN, Chang SF, Huang WS. 6-Shogaol antagonizes the adipocyte-conditioned medium-initiated 5-fluorouracil resistance in human colorectal cancer cells through controlling the SREBP-1 Level. Life (Basel). 2021;11(10):1067. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 99.Liu YL, Xiang Z, Zhang BY, Zou YW, Chen GL, Yin L, Shi YL, Xu LL, Bi J, Wang Q. APOA5 alleviates reactive oxygen species to promote oxaliplatin resistance in PIK3CA-mutated colorectal cancer. Aging (Albany NY). 2024;16(11):9410–36. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 100.Kwan HY, Yang Z, Fong WF, Hu YM, Yu ZL, Hsiao WL. The anticancer effect of oridonin is mediated by fatty acid synthase suppression in human colorectal cancer cells. J Gastroenterol. 2013;48(2):182–92. [DOI] [PubMed] [Google Scholar]
- 101.Liu Y, Hua W, Li Y, Xian X, Zhao Z, Liu C, Zou J, Li J, Fang X, Zhu Y. Berberine suppresses colon cancer cell proliferation by inhibiting the SCAP/SREBP-1 signaling pathway-mediated lipogenesis. Biochem Pharmacol. 2020;174:113776. [DOI] [PubMed] [Google Scholar]
- 102.Wang Y, Guo D, He J, Song L, Chen H, Zhang Z, Tan N. Inhibition of fatty acid synthesis arrests colorectal neoplasm growth and metastasis: anti-cancer therapeutical effects of natural cyclopeptide RA-XII. Biochem Biophys Res Commun. 2019;512(4):819–24. [DOI] [PubMed] [Google Scholar]
- 103.Zhang L, Qiao X, Chen M, Li P, Wen X, Sun M, Ma X, Hou Y, Yang J. Ilexgenin A prevents early colonic carcinogenesis and reprogramed lipid metabolism through HIF1alpha/SREBP-1. Phytomedicine. 2019;63:153011. [DOI] [PubMed] [Google Scholar]
- 104.Jin H, Zhu M, Zhang D, Liu X, Guo Y, Xia L, Chen Y, Chen Y, Xu R, Liu C, et al. B7H3 increases ferroptosis resistance by inhibiting cholesterol metabolism in colorectal cancer. Cancer Sci. 2023;114(11):4225–36. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 105.Pan Q, Zhong S, Wang H, Wang X, Li N, Li Y, Zhang G, Yuan H, Lian Y, Chen Q, et al. The ZMYND8-regulated mevalonate pathway endows YAP-high intestinal cancer with metabolic vulnerability. Mol Cell. 2021;81(13):2736–51 e2738. [DOI] [PubMed] [Google Scholar]
- 106.Muta Y, Linares JF, Martinez-Ordonez A, Duran A, Cid-Diaz T, Kinoshita H, Zhang X, Han Q, Nakanishi Y, Nakanishi N, et al. Enhanced SREBP2-driven cholesterol biosynthesis by PKClambda/iota deficiency in intestinal epithelial cells promotes aggressive serrated tumorigenesis. Nat Commun. 2023;14(1):8075. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 107.Storvold GL, Fleten KG, Olsen CG, Follestad T, Krokan HE, Schonberg SA. Docosahexaenoic acid activates some SREBP-2 targets independent of cholesterol and ER stress in SW620 colon cancer cells. Lipids. 2009;44(8):673–83. [DOI] [PubMed] [Google Scholar]
- 108.Bao W, You Y, Ni J, Hou H, Lyu J, Feng G, Wang Y, You K, Zhang S, Zhang L, et al. Inhibiting sorting nexin 10 promotes mucosal healing through SREBP2-mediated stemness restoration of intestinal stem cells. Sci Adv. 2023;9(35):eadh5016. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 109.Kakimoto M, Yamamoto H, Tanaka AR. Spermine synthesis inhibitor blocks 25-hydroxycholesterol-induced- apoptosis via SREBP2 upregulation in DLD-1 cell spheroids. Biochem Biophys Rep. 2020;22:100754. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 110.Qin S, Su Q, Li X, Shao M, Zhang Y, Yu F, Ni Y, Zhong J. Curcumin suppresses cell proliferation and reduces cholesterol absorption in Caco-2 cells by activating the TRPA1 channel. Lipids Health Dis. 2023;22(1):6. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 111.Collaborators GBDPC. The global, regional, and national burden of pancreatic cancer and its attributable risk factors in 195 countries and territories, 1990–2017: a systematic analysis for the Global Burden of Disease Study 2017. Lancet Gastroenterol Hepatol. 2019;4(12):934–47. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 112.Dee EC, Laversanne M, Bhoo-Pathy N, Ho FDV, Feliciano EJG, Eala MAB, Ting FIL, Ginsburg O, Moraes FY, Gyawali B, et al. Cancer incidence and mortality estimates in 2022 in southeast Asia: a comparative analysis. Lancet Oncol. 2025;26(4):516–28. [DOI] [PubMed] [Google Scholar]
- 113.Ishikawa S, Nagai Y, Masuda T, Koga Y, Nakamura T, Imamura Y, Takamori H, Hirota M, Funakosi A, Fukushima M, et al. The role of oxysterol binding protein-related protein 5 in pancreatic cancer. Cancer Sci. 2010;101(4):898–905. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 114.Oberkofler H, Hafner M, Felder T, Krempler F, Patsch W. Transcriptional co-activator peroxisome proliferator-activated receptor (PPAR)gamma co-activator-1beta is involved in the regulation of glucose-stimulated insulin secretion in INS-1E cells. J Mol Med (Berl). 2009;87(3):299–306. [DOI] [PubMed] [Google Scholar]
- 115.Bathaie SZ, Ashrafi M, Azizian M, Tamanoi F. Mevalonate pathway and human cancers. Curr Mol Pharmacol. 2017;10(2):77–85. [DOI] [PubMed] [Google Scholar]
- 116.Kubota CS, Myers SL, Seppala TT, Burkhart RA, Espenshade PJ. In vivo CRISPR screening identifies geranylgeranyl diphosphate as a pancreatic cancer tumor growth dependency. Mol Metab. 2024;85:101964. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 117.Diraison F, Ravier MA, Richards SK, Smith RM, Shimano H, Rutter GA. SREBP1 is required for the induction by glucose of pancreatic beta-cell genes involved in glucose sensing. J Lipid Res. 2008;49(4):814–22. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 118.Shao S, Liu Z, Yang Y, Zhang M, Yu X. SREBP-1c, Pdx-1, and GLP-1R involved in palmitate-EPA regulated glucose-stimulated insulin secretion in INS-1 cells. J Cell Biochem. 2010;111(3):634–42. [DOI] [PubMed] [Google Scholar]
- 119.Sun Y, He W, Luo M, Zhou Y, Chang G, Ren W, Wu K, Li X, Shen J, Zhao X, et al. Role of transgelin-2 in diabetes-associated pancreatic ductal adenocarcinoma. Oncotarget. 2017;8(30):49592–604. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 120.Yu S, Wang L, Che D, Zhang M, Li M, Naito M, Xin W, Zhou L. Targeting CRABP-II overcomes pancreatic cancer drug resistance by reversing lipid raft cholesterol accumulation and AKT survival signaling. J Exp Clin Cancer Res. 2022;41(1):88. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 121.Yang B, Zhang B, Cao Z, Xu X, Huo Z, Zhang P, Xiang S, Zhao Z, Lv C, Meng M, et al. The lipogenic LXR-SREBF1 signaling pathway controls cancer cell DNA repair and apoptosis and is a vulnerable point of malignant tumors for cancer therapy. Cell Death Differ. 2020;27(8):2433–50. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 122.Rahman R, Asombang AW, Ibdah JA. Characteristics of gastric cancer in Asia. World J Gastroenterol. 2014;20(16):4483–90. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 123.Sun Q, Yu X, Peng C, Liu N, Chen W, Xu H, Wei H, Fang K, Dong Z, Fu C, et al. Activation of SREBP-1c alters lipogenesis and promotes tumor growth and metastasis in gastric cancer. Biomed Pharmacother. 2020;128:110274. [DOI] [PubMed] [Google Scholar]
- 124.Ezzeddini R, Taghikhani M, Somi MH, Samadi N, Rasaee MJ. Clinical importance of FASN in relation to HIF-1alpha and SREBP-1c in gastric adenocarcinoma. Life Sci. 2019;224:169–76. [DOI] [PubMed] [Google Scholar]
- 125.Jia Y, Yan Q, Zheng Y, Li L, Zhang B, Chang Z, Wang Z, Tang H, Qin Y, Guan XY. Long non-coding RNA NEAT1 mediated RPRD1B stability facilitates fatty acid metabolism and lymph node metastasis via c-Jun/c-Fos/SREBP1 axis in gastric cancer. J Exp Clin Cancer Res. 2022;41(1):287. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 126.Zhao L, Peng Y, He S, Li R, Wang Z, Huang J, Lei X, Li G, Ma Q. Apatinib induced ferroptosis by lipid peroxidation in gastric cancer. Gastric Cancer. 2021;24(3):642–54. [DOI] [PubMed] [Google Scholar]
- 127.Guo C, Zhang L, Zhao M, Ai Y, Liao W, Wan L, Liu Q, Li S, Zeng J, Ma X, et al. Targeting lipid metabolism with natural products: a novel strategy for gastrointestinal cancer therapy. Phytother Res. 2023;37(5):2036–50. [DOI] [PubMed] [Google Scholar]
- 128.Gibot L, Follet J, Metges JP, Auvray P, Simon B, Corcos L, Le Jossic-Corcos C. Human caspase 7 is positively controlled by SREBP-1 and SREBP-2. Biochem J. 2009;420(3):473–83. [DOI] [PubMed] [Google Scholar]
- 129.Teng Y, Xia C, Cao M, Yang F, Yan X, He S, Cao M, Zhang S, Li Q, Tan N, et al. Esophageal cancer global burden profiles, trends, and contributors. Cancer Biol Med. 2024;21(8):656–66. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 130.Huang CM, Huang CS, Hsu TN, Huang MS, Fong IH, Lee WH, Liu SC. Disruption of cancer metabolic SREBP1/miR-142–5p suppresses epithelial-mesenchymal transition and stemness in esophageal carcinoma. Cells. 2019;9(1):7. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 131.Shao F, Bian X, Jiang H, Zhao G, Zhu L, Xu D, Wang S, Guo W, Xing D, Xue Q, et al. Association of phosphoenolpyruvate carboxykinase 1 protein kinase activity-dependent sterol regulatory element-binding protein 1 activation with prognosis of oesophageal carcinoma. Eur J Cancer. 2021;142:123–31. [DOI] [PubMed] [Google Scholar]
- 132.Yang P, Li H, Sun M, Guo X, Liao Y, Hu M, Ye P, Liu R. Zinc deficiency drives ferroptosis resistance by lactate production in esophageal squamous cell carcinoma. Free Radic Biol Med. 2024;213:512–22. [DOI] [PubMed] [Google Scholar]
- 133.Wang J, Ling R, Zhou Y, Gao X, Yang Y, Mao C, Chen D. SREBP1 silencing inhibits the proliferation and motility of human esophageal squamous carcinoma cells via the Wnt/beta-catenin signaling pathway. Oncol Lett. 2020;20(3):2855–69. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 134.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 carcinoma. Cell Death Dis. 2021;12(9):845. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 135.Zhong C, Fan L, Li Z, Yao F, Zhao H. SREBP2 is upregulated in esophageal squamous cell carcinoma and co-operates with c-Myc to regulate HMGCR expression. Mol Med Rep. 2019;20(4):3003–10. [DOI] [PMC free article] [PubMed] [Google Scholar]
Associated Data
This section collects any data citations, data availability statements, or supplementary materials included in this article.
Data Availability Statement
No datasets were generated or analysed during the current study.


