Abstract
Malic enzymes (MEs) are metabolic enzymes that catalyze the oxidation of malate to pyruvate and NAD(P)H. While researchers have well established the physiological metabolic roles of MEs in organisms, recent research has revealed a link between MEs and carcinogenesis. This review collates evidence of the molecular mechanisms by which MEs promote cancer occurrence, including transcriptional regulation, post-transcriptional regulation, post-translational protein modifications, and protein-protein interactions. Additionally, we highlight the roles of MEs in reprogramming energy metabolism, suppressing senescence, and modulating the tumor immune microenvironment. We also discuss the involvement of these enzymes in mediating tumor resistance and how the development of novel small-molecule inhibitors targeting MEs might be a good therapeutic approach. Insights through this review are expected to provide a comprehensive understanding of the intricate relationship between MEs and cancer, while facilitating future research on the potential therapeutic applications of targeting MEs in cancer management.
Keywords: Malic enzymes, Cancer, Drug resistance, Cancer therapy, Malic enzyme inhibitors
Graphical abstract
1. Introduction
In 1947, Kornberg et al. discovered an enzyme in pigeon liver that could convert malate to pyruvate using manganese ions (Mn2+). Subsequently, this enzyme was named malic enzyme (ME) [1]. As detection techniques advanced, scientists revealed the widespread distribution of MEs and their roles in animals, plants, and microorganisms [[2], [3], [4], [5], [6], [7], [8], [9]]. Mammalian MEs are classified into three subtypes based on differences in subcellular localization and auxiliary factors: cytoplasmic NADP+-dependent ME1, mitochondrial NAD(P)+-dependent ME2, and mitochondrial NADP+-dependent ME3 [10,11]. In the conversion of malate to pyruvate, MEs reduce NAD(P)+ to NAD(P)H. Pyruvate serves as a feeder molecule to initiate the metabolism of major nutrients, whereas NAD(P)H neutralizes reactive oxygen species (ROS) and is involved in lipid synthesis [[12], [13], [14], [15]]. MEs play crucial roles in regulating the redox balance, molecular synthesis, and energy metabolism in organisms [[16], [17], [18], [19]].
The link between MEs and cancer has garnered considerable interest recently. Dysregulation of MEs has been implicated in several types of cancers, highlighting their potential as important prognostic biomarkers for anticancer treatment outcomes [[20], [21], [22], [23]]. Despite being a group of metabolic enzymes, MEs not only promote the metabolic reprogramming of tumor cells but also contribute to ferroptosis, senescence, and alterations in the tumor immune microenvironment [[24], [25], [26]]. We summarize novel regulatory mechanisms of MEs in cancer, provide an overview of recent findings on how MEs mediate tumor resistance, and highlight the current status of ME inhibitors in cancer management. This review will provide novel insights into new targets and research strategies for the diagnosis and treatment of cancer.
2. Subcellular localization and structure of MEs
Three subtypes of MEs have been identified in mammals, namely ME1, ME2, and ME3, each exhibiting distinct subcellular localization and biological functions [10,11,27] (Fig. 1A). ME1 is an NADP+-dependent metabolic enzyme that primarily localizes to the cytoplasm and participates in cytoplasmic NADPH generation and fatty acid metabolism [[27], [28], [29], [30], [31]]. ME2 is the most abundant and active subtype of MEs, predominantly present in the mitochondria. ME2 regulates cellular energy metabolism by modulating mitochondrial function, the tricarboxylic acid cycle, and glutamine catabolism [22,32,33]. The conventional mitochondrial ME2 lacks an N-terminal chain of 18 amino acids, whereas the cytoplasmic full-length ME2 has this amino acid chain in its structure. Therefore, ME2 can exist in two distinct structural forms and can be found in both the cytoplasm and mitochondria [34]. ME3 is predominantly present in the mitochondria and catalyzes the oxidative decarboxylation of malate to pyruvate in the tricarboxylic acid cycle. Moreover, ME3 plays a crucial role in NADPH synthesis and maintains intracellular homeostasis in ROS levels [11,35].
Fig. 1.
Subcellular localization and structure of MEs
(A) The three subtypes of MEs exhibit distinct subcellular localization. ME1 is predominantly present in the cytoplasm; ME2 can be localized in both the cytoplasm and mitochondria; ME3 is predominantly present in the mitochondria. (B) ME monomers are composed of 572–604 amino acids, consisting of four domains (A, B, C, and D), with domain B further divided into two peptide fragments (B1 and B2).
Malic enzymes are homotetrameric proteins with a dimer-of-dimers structure, and each monomer comprises 572 to 604 amino acids. ME monomers have a molecular weight of 64–68 kDa, comprising four domains (A, B, C, and D), with domain B further divided into two peptide fragments (B1 and B2) (Fig. 1B). The formation of tetramers is facilitated primarily by domains A and D, whereas domains B and C, along with some residues in domain A, play a crucial role in enzyme catalysis [36,37]. The active site of MEs lies within a deep cleft between domains B and C, with highly conserved amino acid residues in the active site region that facilitate substrate binding, divalent cation coordination, NAD(P)+ cofactor binding, and enzyme catalysis regulation [10,37,38].
3. Regulatory mechanisms of MEs in cancer
We outline four key regulatory mechanisms of MEs in cancer: interaction with transcription factors, modulation by miRNAs affecting mRNA levels, involvement in various post-translational modifications influencing cancer progression, and direct protein-protein interactions altering metabolic pathways and proliferation capabilities (Fig. 2).
Fig. 2.
Regulatory mechanisms of MEs in cancer
(A) Interaction of MEs with transcription factors. MEs interact with transcription factors, such as MYC and p53, forming interaction mechanisms, or are directly activated by transcription factors such as NRF2 and ETV4, thereby mediating subsequent functions. (B) MicroRNAs negatively regulate MEs. The mRNA levels of MEs are regulated by miRNAs, such as miR-30a and miR-885-5p, thereby affecting tumor cell proliferation. (C) Post-translational modifications of MEs. MEs are involved in various post-transcriptional modifications such as phosphorylation, acetylation, methylation, and desuccinylation. (D) Interactions between MEs and proteins. MEs directly bind to proteins or form complexes, linking multiple metabolic pathways and affecting tumor progression.
Abbreviations: 6PGD: 6-phosphogluconate dehydrogenase; ACAT1: acetyl-CoA acetyltransferase 1; AKT1: AKT serine/threonine kinase 1; AML: acute myeloid leukemia; AMPK: AMP-activated protein kinase; CRC: colorectal cancer; ETV4: ETS variant transcription factor 4; GC: gastric cancer; HCC: hepatocellular carcinoma; HTC: hydride transfer complex; LKB1: liver kinase B1; LSCC: laryngeal squamous cell carcinoma; MDH1: malate dehydrogenase 1; MDM2: murine double minute 2; ME1: malic enzyme 1; ME2: malic enzyme 2; ME3: malic enzyme 3; mTORC1: mammalian target of rapamycin complex 1; MYC: MYC proto-oncogene, bHLH transcription factor; NEK1: NIMA-related kinase 1; NRF2: nuclear factor erythroid 2-related factor 2; OS: osteosarcoma; OV: ovarian cancer; PC: pyruvate carboxylase; PGAM5: PGAM family member 5; PRAD: prostate adenocarcinoma; PRMT1: protein arginine methyltransferase 1; ROS: reactive oxygen species; SIRT5: sirtuin 5; TCL: T cell lymphoma; TF: transcription factor.
3.1. Interaction of MEs with transcription factors
Transcription factors target MEs based on the cellular context, meticulously adjusting their activity to address diverse environmental and physiological challenges. This precise regulatory mechanism is crucial for optimizing cellular metabolism to suit specific needs, thereby influencing cancer progression and treatment outcomes [21,22,35,43]. These regulatory factors are essential in human physiology and disease management, as they modulate gene expression by recognizing specific DNA sequences, significantly affecting various cancer phenotypes and functions [[44], [45], [46]].
In certain tumors, specific transcription factors activate MEs and speed up cancer progression [21,43] (Fig. 2A). For example, in hepatocellular carcinoma, nuclear factor erythroid 2-related factor 2 (NRF2) is activated by extracellular oxidative stress and subsequently enhances the transcription of ME1. This mechanism is tailored to mitigate ROS damage and bolster the antioxidant defenses in hepatocellular carcinoma cells, which inadvertently supports their proliferation [21]. In gastric cancer, ME1 is upregulated by ETS variant transcription factor 4 (ETV4) transcription, leading to increased resistance to apoptosis and enhanced proliferation of gastric cancer cells. Moreover, ME1 promotes tumor growth and metastasis of gastric cancer in vivo [43].
MEs are also regulated by multiple transcriptional regulatory factors through feedback loop mechanisms, thereby participating in tumorigenesis and progression [22,35] (Fig. 2A). For instance, in a mouse model with both ME2 gene knockout and MYC overexpression, MYC upregulated ME2 and stimulated mTORC1 activity by enhancing glutamine metabolism [22]. This promoted MYC translation, forming a positive feedback loop that maintained redox homeostasis and enhanced the proliferation of T cells in lymphoma [22]. In another study, p53, activated by DNA damage signals, inhibited the transcription of ME1 in colorectal cancer cells, and downregulation of ME1 activates p53 through the inhibition of murine double minute 2 (MDM2) expression. Simultaneously, p53 transcriptionally suppressed ME2, upregulating AMP-activated protein kinase (AMPK) expression and leading to further phosphorylation and activation of p53. In summary, ME1/ME2 inhibited by p53, in turn activates p53, ultimately enhancing the senescence phenotype and reducing the proliferation of colorectal cancer cells [35].
These studies indicate that the specific transcriptional regulatory mechanisms associated with MEs affect the redox balance, energy metabolism, and pathways such as cell senescence or apoptosis, in cancer cells and play pivotal roles in cancer onset and progression. Interventions targeting MEs or their transcriptional regulatory pathways may offer new options for treating malignant tumors.
3.2. MicroRNAs negatively regulate MEs
MicroRNAs (miRNAs) are post-transcriptional regulators that mediate mRNA degradation or translational inhibition through complementary pairing with target mRNA, thereby regulating the development of drug resistance in cancer [[47], [48], [49]]. Some miRNAs have been identified to inhibit the mRNA levels of MEs and subsequently attenuate cancer progression [[50], [51], [52]] (Fig. 2B). For example, in KRAS-mutant colorectal cancer cells, miR-30a suppressed ME1 expression by binding to its 3′ untranslated region. Downregulation of ME1 expression can reduce NADPH synthesis, lipid synthesis, and fatty acid uptake in cells, inhibiting the proliferation of colorectal cancer cells both in vitro and in vivo [50]. Another miRNA, miR-885-5p, was reported to reduce the proliferation, migration, and invasion of laryngeal squamous cell carcinoma cells by suppressing ME1 expression [52]. In gastric cancer cells, miR-885-5p inhibits the expression of ME1 and that of its downstream molecules, vimentin and fibronectin. As a result, the migration, invasion, and metastasis of gastric cancer are inhibited [51].
Current research on miRNAs and MEs has focused on the ME1 subtype, which contributes to our understanding of the involvement of MEs in cancer at the post-transcriptional level [[50], [51], [52]]. These studies have emphasized the importance of ME1 as an adverse prognostic factor in various cancers, suggesting that the downregulation of ME1 expression may hold therapeutic potential for cancer management.
3.3. Post-translational modifications of MEs
Post-translational modifications occur on specific amino acids within the regulatory domains of target proteins, increasing the functional diversity of the proteome in complex ways. Such modifications have emerged as therapeutic targets in cancer [[53], [54], [55]]. MEs can influence cancer progression after undergoing various post-translational modifications, including phosphorylation, acetylation, methylation, and desuccinylation [20,23,32,34,56] (Fig. 2C).
One such modification is phosphorylation, which is crucial for ME activity. AKT serine/threonine kinase 1 (AKT1) phosphorylates the Ser9 site of cytoplasmic ME2, enhancing its enzymatic activity and preventing its mitochondrial translocation. Cytoplasmic ME2 assembles various glycolytic enzymes, including PFKL, GAPDH, PKM2, and LDHA, ultimately facilitating the metabolic switch from mitochondrial metabolism to glycolysis, thereby resulting in tumorigenesis [34].
Acetylation is another pronounced post-translational modification. The dynamic regulation of phosphorylation and acetylation in ME1 plays a crucial role in energy metabolism and disease progression in colorectal cancer [20]. NIMA-related kinase 1 (NEK1) phosphorylated ME1 at the S336 site, inhibiting its activity and suppressing tumor growth in colorectal cancer [20]. In contrast, PGAM family member 5 (PGAM5) dephosphorylated the S336 site, allowing acetyl-CoA acetyltransferase 1 (ACAT1) to acetylate the adjacent K337 site [20]. After acetylation, ME1 underwent dimerization, which enhanced its ability to catalyze NADPH and lipid production, promoting the proliferation of colorectal cancer cells [20]. Acetylation is the process of transferring and adding acetyl groups to lysine residues or N-termini of proteins, which to some extent influences the occurrence and development of tumors [[57], [58], [59]]. The dynamic balance between phosphorylation at S336 and acetylation at K337 of ME1 suggests a potential mechanism through which metabolic enzymes compete for modifications, thereby governing tumor signaling [20]. Moreover, research on ME activity emphasizes the importance of NADPH production. In osteosarcoma cells, silencing ME1/ME2 reduced NADPH levels resulting in disrupted histone acetylation and transcription processes [56].
Another form of modification is methylation, which is closely associated with tumorigenesis [[60], [61], [62]]. Mitochondrial overdrive is a main contributor to acute myeloid leukemia development, and ME2 enhances the invasiveness of leukemia cells and promotes tumor progression in vivo [32]. Methylation of ME2 by protein arginine methyltransferase-1 (PRMT1) at the fumarate-binding site (R67) of ME2 inhibits its dimerization and activity, suppressing fumarate signaling transduction and mitosis, thereby reducing the invasiveness of acute myeloid leukemia [32].
Desuccinylation has been found to significantly impact cancer progression, as evidenced by reports that desuccinylation of ME2 influences this process [23]. Succinylation involves attaching succinyl groups to lysine residues of substrate proteins, thereby altering protein structure and function. This modification has been found to regulate tumor growth [[63], [64], [65]]. The succinylation of ME2 is considered a physiological post-translational modification in colorectal cancer cells. Protein lysine desuccinylase 5 (SIRT5) desuccinylates the K346 site of ME2, activating it and subsequently enhancing mitochondrial respiration and cell proliferation [23]. Moreover, desuccinylation of the K346 site of ME2 promotes tumor growth in vivo [23].
These data suggest that post-translational modifications of MEs regulate tumor progression by influencing mitochondrial function, NADPH synthesis, and metabolic reprogramming.
3.4. Interactions between MEs and proteins
MEs can influence tumor development by directly binding to proteins or forming complexes with other molecules [24,66,67] (Fig. 2D). For example, ME1 can directly bind to and activate 6-phosphogluconate dehydrogenase (6PGD) and promote NADPH generation, pentose phosphate pathway flux, and the proliferation of osteosarcoma cells, suggesting direct interactions between MEs and the pentose phosphate pathway [66]. In the cytoplasm of prostate cancer cells, the tricarboxylic acid cycle (TCA) involves ME1, pyruvate carboxylase (PC), and malate dehydrogenase 1 (MDH1), resulting in the formation of intermediate transfer complex (HTC). This complex provides cells with NAD+ and NADPH, driving metabolic reprogramming to suppress senescence and induce tumorigenesis [24]. However, MEs are not always carcinogenic. In ovarian cancer cells, liver kinase B1 (LKB1) promotes the function of ME3 by binding to its N-terminus. Upregulation of ME3 expression enhances the transcription of p21 and p53 while inhibiting the transcription of NF-κB, promoting apoptosis and inhibiting cancer cell proliferation [67]. The regulatory effect of MEs on the development of pelvic malignancies and osteosarcoma is complex, and interventions targeting these targets may provide more therapeutic opportunities for patients with cancer.
4. Biological functions of MEs in cancer
MEs are dysregulated in various types of cancers and are closely linked to cancer prognosis (Fig. 3, Table 1). MEs are upregulated in several cancers, and are usually associated with shorter overall survival. However, MEs are found to be downregulated in certain types of cancers, such as bladder cancer, which is associated with shorter overall survival duration. To provide a more detailed description of malic enzymes in these cancers and their association with overall survival, we have supplemented our analysis with additional data from the UALCAN database (presented in Fig. 3). MEs play vital roles in cancer development. Owing to the recent advancement in understanding the complex biological role of MEs in cancer, this section focuses on the impact of MEs on tumor metabolic reprogramming, programmed cell death, senescence and cell cycle progression, and the tumor immune microenvironment (Fig. 4).
Fig. 3.
Dysregulation and survival relevance of MEs in various cancer types
Triangles represent the expression levels of MEs in cancer, with red indicating upregulation, blue indicating downregulation, and no color indicating no statistically significant difference in expression levels or N/A. Circles represent the relationship between expression levels of MEs and survival outcomes. Red circles indicate that high expression predicts unfavorable survival, blue circles indicate that high expression predicts favorable survival, and circles with no color and diagonal lines indicate that expression levels are not significantly associated with survival. Content within the dashed lines is supplementary data from the UALCAN database. (For interpretation of the references to color in this figure legend, the reader is referred to the Web version of this article.)
Table 1.
The role of MEs in cancer.
| ME | Cancer types | Expression | Effect on cancer | Prognosis | Biological functions | Ref |
|---|---|---|---|---|---|---|
| ME1 | Lung cancer | Upregulated | Promote | / | Proliferation, metabolic reprogramming | [82] |
| Lung cancer | Upregulated | Promote | Poor | Proliferation | [104] | |
| hepatocellular carcinoma | Upregulated | Promote | Poor | Proliferation, migration, invasion, metastasis, metabolic reprogramming | [21] | |
| hepatocellular carcinoma | Upregulated | Promote | / | / | [109] | |
| hepatocellular carcinoma | / | Promote | / | Proliferation, metabolic reprogramming | [81] | |
| Osteosarcoma | / | Promote | / | Proliferation, metabolic reprogramming | [66] | |
| Laryngeal squamous cell carcinoma | Upregulated | Promote | / | / | [52] | |
| Synovial sarcoma | Downregulated | / | / | Ferroptosis, metabolic reprogramming | [25] | |
| Colorectal cancer | / | Promote | / | Senescence, proliferation, metabolic reprogramming | [35] | |
| Colorectal cancer | Upregulated | Promote | / | Proliferation, metabolic reprogramming | [20] | |
| Colorectal cancer | Upregulated | Promote | / | [96] | ||
| Colorectal cancer | Upregulated | Promote | / | Proliferation, metabolic reprogramming | [50] | |
| Colorectal cancer | Upregulated | Promote | / | Senescence, apoptosis, proliferation, metabolic reprogramming | [85] | |
| Oral squamous cell carcinoma | Upregulated | Promote | / | Proliferation, invasion | [102] | |
| Oral squamous cell carcinoma | Upregulated | Promote | / | EMT, metabolic reprogramming | [110] | |
| Oral squamous cell carcinoma | Upregulated | Promote | Poor | Proliferation, invasion, EMT, stemness | [111] | |
| Prostate cancer | Upregulated | Promote | / | Senescence, proliferation, metabolic reprogramming | [24] | |
| Breast cancer | / | Promote | / | Proliferation | [112] | |
| Breast cancer | Upregulated | Promote | Poor | Proliferation, metabolic reprogramming | [76] | |
| Breast cancer | / | Promote | / | Proliferation, apoptosis, metabolic reprogramming | [113] | |
| Gastric cancer | Upregulated | Promote | Poor | Proliferation, apoptosis, metastasis | [43] | |
| Gastric cancer | Upregulated | Promote | Poor | Migration, invasion, metastasis | [51] | |
| Ovarian cancer | Upregulated | Promote | / | Senescence, apoptosis, proliferation, metabolic reprogramming | [85] | |
| Acute myeloid leukemia | Upregulated | Promote | Poor | Proliferation, metabolic reprogramming | [103] | |
| ME2 | Acute myeloid leukemia | Upregulated | Promote | Poor | Proliferation, metabolic reprogramming | [32] |
| Acute myeloid leukemia | Upregulated | Promote | / | Apoptosis, metabolic reprogramming | [77] | |
| Lymphoma | Upregulated | Promote | Poor | Proliferation, metabolic reprogramming | [22] | |
| Lung cancer | Upregulated | Promote | / | Migration, invasion | [27] | |
| Lung cancer | Upregulated | Promote | / | Proliferation, metabolic reprogramming | [34] | |
| hepatocellular carcinoma | Upregulated | Promote | Poor | Proliferation, cell cycle | [93] | |
| hepatocellular carcinoma | Upregulated | Promote | Poor | Proliferation, metabolic reprogramming | [33] | |
| hepatocellular carcinoma | / | Promote | / | Migration, invasion | [114] | |
| hepatocellular carcinoma | Upregulated | Promote | Poor | Proliferation, migration, EMT, cell cycle, apoptosis | [92] | |
| Melanoma | Upregulated | Promote | / | Proliferation, migration, invasion, metabolic reprogramming | [115] | |
| Glioblastoma | Upregulated | Promote | Poor | Proliferation, migration, invasion, PMT, metabolic reprogramming | [80] | |
| Colorectal cancer | / | Promote | / | Senescence, proliferation, metabolic reprogramming | [35] | |
| Colorectal cancer | Upregulated | Promote | Poor | Proliferation, metabolic reprogramming | [23] | |
| Oral squamous cell carcinoma | Upregulated | Promote | Poor | [116] | ||
| Breast cancer | Upregulated | Promote | Poor | Metastasis | [117] | |
| Salivary gland cancer | Upregulated | Promote | / | Proliferation | [118] | |
| ME3 | Bladder cancer | Downregulated | Suppress | Good | [119] | |
| Ovarian cancer | / | Suppress | / | Proliferation, apoptosis | [67] | |
| Pancreatic cancer | Upregulated | Promote | / | Apoptosis, metabolic reprogramming | [16] | |
| Pancreatic cancer | Upregulated | Promote | Poor | Proliferation, migration, invasion, EMT | [120] | |
| Acute myeloid leukemia | Upregulated | Promote | Poor | Proliferation, metabolic reprogramming | [103] | |
| hepatocellular carcinoma | Upregulated | Promote | Poor | Proliferation, migration, invasion, metastasis, metabolic reprogramming | [21] |
Fig. 4.
Biological functions of MEs in cancer
The outer circle delineates the subtypes of MEs, with ME1 denoted by a blue circle, ME2 by a purple circle, and ME3 by a yellow circle. (A) MEs in tumor metabolic reprogramming. MEs reprogram the metabolism of tumor cells by mediating aerobic glycolysis, fatty acid metabolism, and glutamine degradation. The mitochondrial activity and redox balance of tumor cells are altered as a result. (B) MEs in programmed cell death. Silencing of MEs induces apoptosis by increasing the activity of caspase-3,7 or ROS. The absence of ME1 leads to the accumulation of ROS and active iron, thereby increasing ferroptosis. (C) MEs in senescence and cell cycle progression. MEs modulate the senescence phenotype by forming complexes with other molecules. MEs play a role in regulating cell cycle progression through direct or indirect mechanisms. (D) MEs in the tumor immune microenvironment. ME1 overexpression in CD8+ T cells increases mitochondrial respiration rate and ATP production, contributing partially to the enhanced cytotoxicity of CD8+ T cells. Downregulation of ME1 sensitizes tumor cells to the toxicity of natural killer cells-derived HMGB1. (For interpretation of the references to color in this figure legend, the reader is referred to the Web version of this article.)
4.1. MEs in tumor metabolic reprogramming
Cancer cells have the ability regulate their energy metabolism, favoring their proliferation and differentiation. Therefore, reprogramming of energy metabolism is a hallmark of malignant cancers [[68], [69], [70]]. Typically, metabolic reprogramming in cancer occurs in pathways of aerobic glycolysis, fatty acid metabolism, and glutamine metabolism [71,72]. MEs can mediate tumor onset and progression by promoting aerobic glycolysis, fatty acid metabolism, and glutamine metabolism, as discussed in subsequent sections (Fig. 4A).
4.1.1. MEs in aerobic glycolysis
Aerobic glycolysis, also known as the Warburg effect, refers to the phenomenon in which tumor cells convert glucose to lactate even in the presence of oxygen, to satisfy their own needs for proliferation and metastasis. It is the most extensively studied phenomenon in the metabolic reprogramming of malignant cancers [[73], [74], [75]]. MEs can mediate aerobic glycolysis and influence tumor progression. For example, ME1 overexpression increases the production of lactate in breast cancer cells, which increases the extracellular acidification rate (ECAR) and decreases the oxygen consumption rate (OCR), thereby promoting aerobic glycolysis and facilitating tumor growth [76]. Downregulation of ME2 in acute myeloid leukemia decreases the expression of glycolytic intermediates, effectively inhibits glycolysis, and suppresses acute myeloid leukemia cell proliferation [77].
4.1.2. MEs in fatty acid metabolism
The metabolic reprogramming of fatty acid metabolism is reported to occur in several cancer types, which activates the biosynthesis of fatty acids to satisfy the proliferation demands of cancer cells [71,78,79]. MEs can promote tumor cell fatty acid metabolism [50,80,81]. For instance, ME2 enhances the de novo synthesis of fatty acids via the AMPK-SREBP-1-ACSS2 pathway, thereby promoting the proliferation, migration, and invasiveness of glioblastoma multiforme cells [80]. Knocking out ME1 significantly reduces triglyceride and fatty acid levels in KRAS-mutant colorectal cancer cells, thereby inhibiting their tumorigenicity [50]. PGAM5 mediates ME1 dephosphorylation and enhances ME1 activity, thereby promoting NADPH production, fatty acid metabolism, and tumor growth in hepatocellular carcinoma cells [81]. Considering that NADPH provides the essential reducing power for lipid metabolism, an increase in NADPH production through the action of ME1 has been reported to enhance the growth of various tumors [21,24,82].
4.1.3. MEs in glutamine catabolism
Glutamine is another important nutrient in addition to glucose and fats. Glutamine is broken down into various metabolites to satisfy the energy demands of tumor cells without compromising the redox balance in cancer cells [83,84]. Previous studies have indicated that silencing ME1 or ME2 can reduce glutamine breakdown in colorectal cancer cells, suggesting that MEs play crucial roles in glutamine catabolism [35]. Glutamine, rather than glucose, is the main nutrient for MYC/ME2-driven T-cell lymphoma cells, and ME2 overexpression can restore glutamine consumption in MYC-deleted cells, thereby promoting cell proliferation [22]. ME2 also maintains the levels of 2-hydroxyglutarate in glioblastoma cells by regulating glutamine breakdown. The direct binding of 2-hydroxyglutarate with mutant p53 inhibits the MDM2-mediated ubiquitination and degradation of mutant p53, thereby promoting tumor growth [33].
MEs play important roles in maintaining mitochondrial function and cellular redox balance [21,32]. For instance, in acute myeloid leukemia, ME2 can target and activate deoxyuridine triphosphate nucleotidohydrolase, elevating the mitochondrial levels of dNTP and mtDNA and consequently increasing mitochondrial biomass in acute myeloid leukemia cells [32]. In hepatocellular carcinoma, ME3 facilitates the production of a large amount of mitochondrial NADPH, which helps maintain redox balance; conversely, silencing ME3 directly reduces mitochondrial activity in hepatocellular carcinoma cells [21].
In summary, MEs accelerate tumor progression by modulating nutrient metabolism reprogramming and mitochondrial activity. Therefore, targeting MEs may offer a novel therapeutic strategy for inhibiting tumor metabolism.
4.2. MEs in programmed cell death
MEs inhibit tumor cell apoptosis through various mechanisms, promoting tumor development [43,77,85]. For instance, silencing ME1 in gastric cancer cells induces apoptosis and significantly inhibits tumor growth [43]. In non-small cell lung cancer cells, silencing ME1 significantly increases the activity of caspase-3, 7 and induces apoptosis [85]. Furthermore, ME2 downregulation leads to increased ROS, thus resulting in apoptosis of acute myeloid leukemia cells [77].
Unlike apoptosis, ferroptosis is a newly identified iron-dependent form of programmed cell death that generates large amounts of lipid peroxidation products, ultimately inducing cell death and playing a crucial role in cancer progression [[86], [87], [88]]. The absence of ME1 in synovial sarcoma cells leads to the accumulation of ROS and active iron, increasing the occurrence of ferroptosis and enhancing the sensitivity of cells to cystine transporter SLC7A11/xCT inhibitors that induce ferroptosis [25]. Loss of ME1 expression exacerbates ischemia-reperfusion-induced hepatocyte ferroptosis, whereas supplementation of the ME1 substrate l-malate restores NADPH and glutathione levels, thereby inhibiting ischemia-reperfusion-induced hepatocyte ferroptosis [17].
In summary, MEs regulate different types of programmed cell death, thereby affecting tumor progression and treatment resistance (Fig. 4B). Therefore, interventions that target MEs may, at least, partially address the challenge of treatment resistance in cancer therapy.
4.3. MEs in senescence and cell cycle progression
Senescence is a state of cell cycle arrest and a protective mechanism against tumorigenesis, with microenvironmental stress and imbalances in cellular signaling networks as triggers of senescence [[89], [90], [91]]. MEs are reported to promote tumor growth by inhibiting cancer cell senescence [24,85] (Fig. 4C). For instance, murine and human prostate cancer models exhibit overexpression of the hydrogen transfer complex (HTC), consisting of ME1, PC, and MDH1. Silencing any of these three enzymes in the HTC induces senescence and leads to the arrest of proliferation [24]. Similarly, prostate cells lacking ME1 exhibit a higher proportion of SA-β-Gal-positive cells, another hallmark of senescence, suggesting that knocking out ME1 induces senescence and inhibits cancer cell growth [85]. The mutual regulation between p53 and MEs plays a major role in regulating senescence in normal and cancer cells [35].
MEs also mediate tumor progression by regulating the cell cycle process [92,93] (Fig. 4C). ME2 promotes the binding of α-ketoglutaric acid to RNA polymerase II, indirectly increasing cyclin D1 transcription and thus promoting cell cycle progression and the proliferation of hepatocellular carcinoma cells [93]. An increase in the number of non-proliferating hepatocellular carcinoma cells in the G0 phase has been reported after silencing ME2, indicating that ME2 downregulation can inhibit cell cycle progression and proliferation [92]. These data suggest that MEs mediate tumor progression either by inhibiting senescence or by promoting cell cycle progression, highlighting that cellular metabolism is a major predictor of the irreversible fate of cells.
4.4. MEs in the tumor microenvironment
Malignant tumors are not simply a cluster of isolated cell types but rather a complex ecosystem composed of immune cells, stromal cells, blood vessels, nerves, lymphatic vessels, and other types of cells [94,95]. ME1 regulates the activity of CD8+ T cells and natural killer cells through different mechanisms and alters the tumor microenvironment (TME) [26,96] (Fig. 4D). For instance, salvage therapy sometimes can activate cytotoxic T cells in patients with refractory advanced cancer, leading to tumor regression [26]. Analysis of peripheral blood lymphocytes from patients with late-stage lung metastasis patients who received salvage therapy with spatially fractionated radiotherapy showed that the treatment increased the toxicity of CD8+ T cells by upregulating ME1. ME1 overexpression in CD8+ T cells significantly increased mitochondrial respiration rate and ATP production, which contributed partially to the enhanced cytotoxicity of CD8+ T cells through the type I interferon pathway [26]. Natural killer cells-derived high-mobility group box 1 (HMGB1) can also effectively induce cell death in colorectal cancer cells, and downregulation of ME1 sensitizes glutamine-dependent colorectal cancer cells to the toxicity of HMGB1 [96].
TME, comprised of diverse immune cell subsets, plays a critical role in tumor development and metastasis. Ongoing advancements in combined immunotherapy, built upon understanding the regulatory mechanisms within the TME, are being continuously refined [97,98]. Research exploring the link between malic enzymes and the TME provides valuable insights into combined therapies for cancer. Further investigation in this field will deepen our understanding of the underlying molecular mechanisms.
5. ME-mediated drug resistance
Drug resistance poses a major challenge that prevents us from achieving favorable outcomes despite advancements in cancer treatment strategies, and genetic and non-genetic mechanisms are reported to contribute to resistance to cancer treatment [[99], [100], [101]]. MEs can influence drug resistance in hematologic and epithelial malignancies through various pathways [21,82,102,103] (Fig. 5). For instance, in highly invasive acute myeloid leukemia, ME1/ME3 modulates cellular oxidative metabolism by balancing the levels of NADPH and ROS, thereby mediating resistance to cytarabine. Targeting ME1/ME3 effectively reduces the tumorigenicity of acute myeloid leukemia and helps overcome resistance [103]. In hepatocellular carcinoma, ME1/ME3 downregulation leads to increased apoptosis, reduced proliferation, and ROS induction after sorafenib treatment, suggesting that interference with the ME1/ME3 pathway sensitizes hepatocellular carcinoma to sorafenib therapy [21]. ME1 and G6PD serve as primary NADPH contributors in tyrosine kinase inhibitor-resistant non-small cell lung cancer cells, and the administration of ME1 inhibitors effectively overcomes cancer resistance, especially when combined with osimertinib [82]. The high expression of ME1 maintains lactate fermentation levels by promoting glutamine metabolism, enhancing proliferation, and increasing resistance in oral squamous cell carcinoma [102].
Fig. 5.
ME-mediated drug resistance
MEs influence the sensitivity of cancers originating from epithelial tissues, mesenchymal tissues, and the hematopoietic system, to chemotherapy drugs, targeting agents, and ferroptosis inducers.
Depletion of ME1 expression makes mesenchymal tissue-derived malignant tumors more sensitive to ACXT-3102, which is an erastin compound that induces ferroptosis by inhibiting xCT-mediated cystine uptake [25]. Depletion of ME1 expression also alters the antioxidant defense system in synovial sarcoma cells, leading to the accumulation of ROS and labile iron, thereby increasing the sensitivity of these cells to ACXT-3102-induced ferroptosis. This further supports the development of therapeutic strategies targeting ferroptosis pathways in synovial sarcoma [25]. Moreover, patients with non-small cell lung cancer who exhibit increased ME1 expression typically experience poorer clinical outcomes following radiotherapy. This relationship underscores the potential of targeting ME1 to enhance radiotherapy efficacy, offering a promising direction for future therapeutic strategies [104].
In summary, MEs mediate tumor resistance by influencing cellular redox balance, glutamine metabolism, NADPH synthesis, and other processes in malignant tumors of the hematopoietic system and epithelial tissues. Conversely, in tumors originating from mesenchymal tissues, MEs affect tumor resistance by modulating the sensitivity of these cancer cells to ferroptosis induction. This highlights the potential of MEs as emerging biological markers in cancer therapy and provides new directions and insights for future research endeavors.
6. ME inhibitors
The various subtypes of human MEs share a similar quaternary structure, forming a homotetrameric protein (Fig. 6A). ME2 has a complex allosteric system, with each enzyme monomer containing two independent allosteric sites: the fumarate site at the dimer interface and an exosite at the tetramer interface. ME2 can shift between two open and two closed forms in equilibrium [10,[39], [40], [41], [42]]. ME1 and ME3 are non-allosteric enzymes with relatively stable tetrameric structures [37,38]. From the first identification of the crystal structure of human ME in 1999 to the elucidation of the crystal structure of ME3 in 2022, significant strides have been made in characterizing the structure of this enzyme family, establishing a basis for subsequent small-molecule drug development [36,38].
Fig. 6.
The quaternary structure of MEs and their inhibitors
(A) The various subtypes of human MEs share a similar quaternary structure, forming a homotetrameric protein. The quaternary structure of proteins shown inFig. 6A was obtained from the Protein Data Bank (ME1: PDB ID7X11, ME2: PDB ID1DO8, ME3: PDB ID8E76). (B) Chemical molecular structures of ME inhibitors.
The role of MEs in mediating tumor progression and drug resistance has garnered considerable interest in recent years. Therefore, targeting MEs could potentially become a novel approach in cancer therapy (Fig. 6B–Table 2). A virtual screening against homologous models of MEs conducted in 2006 led to the design and synthesis of novel ME inhibitors based on a piperazine-1-pyrrolidine-2,5-dione scaffold [105]. AS1134900 is a small-molecule inhibitor targeting ME1 (with an IC50 value of 0.73 μM) with high selectivity. Upon binding, it reduces the flexibility of domain C of ME1 and competitively inhibits its activity in the presence of substrates NADP+ and malate [106]. However, it fails to effectively inhibit the proliferation of pancreatic cancer cells, possibly due to its limited cell permeability. Optimization of this inhibitor could offer new directions for ME1-related cancer therapy in the future [106].
Table 2.
ME inhibitors.
| ME | Inhibitors | Cancer types | IC50(μM) | Target site | Advantages | Disadvantages | Ref |
|---|---|---|---|---|---|---|---|
| ME1 | AS1134900 | Pancreatic cancer | 0.73 | Between the α-helices of domains B and C | High specificity | Low cell permeability; Requiring further optimization | [106] |
| ME2 | NPD389 | / | 4.63 ± 0.36 (No Brij-35), 5.59 ± 0.38 (0.01 % Brij-35) |
/ | Fast-binding | Requiring NAD+ for binding; Mixed-type inhibition | [107] |
| ME2 | NPD387 | / | 11.84 ± 0.57 (No Brij-35), 18.27 ± 1.46 (0.01 % Brij-35) |
/ | Novel structure | Lower efficiency; Structural instability | [107] |
| ME2 | MDSA | Lung cancer | 0.51 | The dimer interface's allosteric fumarate-binding site | High specificity | Varying efficacy | [27] |
| ME2 | EA | Lung cancer | 0.1 | The dimer interface's allosteric fumarate-binding site | High specificity | Limited inhibition | [27] |
| ME2 | Na2EA | Acute myeloid leukemia | 120 | / | High specificity | Dose-dependent effects | [77] |
| ME3 | Several analogs of 6-piperazin-1-ylpyridin-3-ol amides | Pancreatic cancer | 0.06–0.58 | Compound 10b binds the hydrophobic pocket of ME3 | High specificity; Stable tetramer structure | Lacking allosteric regulation | [108] |
ME2 inhibitors have garnered popularity in recent years. In 2014, a 2,5-dihydroxyl benzoquinone derivative, NPD387, was identified through high-throughput screening as a moderate ME2 inhibitor. It exhibits specificity by binding to the 2,5-dihydroxybenzoquinone skeleton of ME2 [107]. However, it has lower efficacy and structural instability. A more potent derivative, NPD389, was also discovered, which binds rapidly and non-competitively to the NAD+ binding site of ME2. NPD389 has fast-binding properties, is a highly effective inhibitor, and shows specificity for ME2. Nevertheless, it requires NAD+ for binding and demonstrates mixed-type inhibition [107]. Two ME2-specific inhibitors, 5,5′-methylenedisalicylic acid (MDSA) and embonic acid (EA), were reported to inhibit ME2 activity in non-small cell lung cancer cells (with IC50 values of 0.51 μM and 1.1 μM, respectively), leading to tumor cell death [27]. Specifically, MDSA and EA bind to the fumarate binding site of ME2 and inhibit ME2 by inducing conformational changes, disrupting cellular energy metabolism, and inhibiting the migration and invasion abilities of non-small cell lung cancer cells [27]. The fumarate binding site is located at the dimer interface of the enzyme, approximately 30 Å from the active site [40]. Similarly, disodium salt of embonic acid (Na2EA) inhibits ME2 activity by triggering conformational changes, disrupting redox homeostasis, reducing energy metabolism, inducing apoptosis, and inhibiting tumor growth in acute myeloid leukemia, suggesting that targeting ME2 could be an effective strategy for treating acute myeloid leukemia [77]. Moreover, treatment effectively reduces ME2 activity and ATP levels in non-cancerous cells, demonstrating that ME2 inhibitors impact the energy metabolism of both cancerous and normal cells [27]. This emphasizes the necessity for the future advancement of ME2 inhibitors that specifically target tumor cells while minimizing effects on normal cells [27].
ME3 inhibitors have recently emerged. Several analogs of 6-piperazin-1-ylpyridin-3-ol amides possessing potent inhibitory activity against ME3 have been synthesized and identified [108]. Notably, the in silico binding analysis of compound 10b showed that ring B was aligned toward the hydrophobic pocket of ME3. Furthermore, analogs of AS1134900 (ME1 inhibitor) are speculated to be effective against ME3, based on the premise that ME3 has a non-allosteric structure and an AS1134900-binding site that is highly conserved between ME1 and ME3 [38].
In summary, research on ME inhibitors is still in its infancy. In the face of several challenges, further in-depth research on MEs is expected to assist future development activities.
7. Conclusions and perspectives
MEs are metabolic enzymes located in the cytoplasm or mitochondria of cells. They drive various critical physiological metabolic processes in both eukaryotes and prokaryotes. A close association has been identified between MEs in mammalian cells and the onset and progression of cancer. This review has detailed the molecular regulatory mechanisms and biological functions of MEs in cancer and summarized recent research on their involvement in tumor drug resistance and the development of inhibitors. At the molecular level, MEs first influence the cellular redox balance, proliferation, and metastatic ability of tumors by inducing transcriptional changes in the expression of several key molecules. Subsequently, MEs affect post-transcriptional modifications through miRNAs, altering the energy metabolism programming, proliferation, and migration abilities of cancer cells. Post-translational modifications of MEs are complex and interconnected. A dynamic link exists between phosphorylation and acetylation, whereas methylation and desuccinylation alter mitosis or mitochondrial function. MEs can also directly bind to proteins or form complexes, linking multiple metabolic pathways and thus influencing tumor progression. In terms of biological functions, we highlight the role of MEs in tumor energy metabolism reprogramming, programmed cell death, senescence and cell cycle progression, and TME modulation. While MEs predominantly exhibit a pro-carcinogenic effect in most malignant tumors, they also play a role in suppressing the growth of certain tumors.
By regulating cell death pathways, redox balance, and energy metabolism, MEs can also influence the extent to which tumor cells are resistant to chemotherapeutic drugs, such as cytarabine, targeting agents like sorafenib and osimertinib, and the iron-induced cell death inducer ACXT-3102. The multidimensional role of MEs in malignant tumors makes them potential targets for cancer therapy. Consequently, research on ME inhibitors has been on the rise. These inhibitors can competitively inhibit ME1 activity or allosterically inhibit ME2 activity by binding to its malate-binding site. Overall, research on MEs and malignant tumors is still in its early stages, and the application of MEs-based therapy in clinical settings faces several challenges. However, gaining a deeper understanding of its specific molecular mechanisms and biological functions, and emphasizing the role that MEs play in tumorigenesis, will highlight the therapeutic potential of using ME inhibitors in cancer management. Future in-depth studies on the mechanisms of MEs, multidisciplinary integration, and the availability of clinical trial evidence will contribute to optimizing cancer treatment modalities, ultimately improving the efficacy of cancer treatment for patients.
Availability of data and materials
Not applicable.
Ethics approval and consent to participate
Not applicable.
Consent for publication
All authors consent to publication.
CRediT authorship contribution statement
Huan Wang: Writing – original draft, Funding acquisition. Wanlin Cui: Writing – original draft. Song Yue: Investigation. Xianglong Zhu: Investigation. Xiaoyan Li: Investigation. Lian He: Investigation. Mingrong Zhang: Investigation. Yan Yang: Investigation. Minjie Wei: Writing – review & editing. Huizhe Wu: Writing – review & editing. Shuo Wang: Writing – review & editing.
Declaration of competing interest
The authors declare that they have no known competing financial interests or personal relationships that could have appeared to influence the work reported in this paper.
Acknowledgements
This work was supported by grants from the National Natural Science Foundation of China (82272915, 82073884), the project of the fourth batch of science and technology plan of Liaoning province (2021JH210300133), the Fundamental Research Funds for the Central Universities (LD202211), “Oncology+” Open Project of Liaoning Cancer Hospital (2024-ZLKF-03).
Contributor Information
Huan Wang, Email: wanghuan2@cancerhosp-ln-cmu.com.
Wanlin Cui, Email: cwl530@163.com.
Song Yue, Email: yuesong567@163.com.
Minjie Wei, Email: wangshuo@cancerhosp-ln-cmu.com.
Huizhe Wu, Email: wuhz@cmu.edu.cn.
Shuo Wang, Email: mjwei@cmu.edu.cn.
Abbreviations
- 6PGD
6-phosphogluconate dehydrogenase
- ACAT1
acetyl-CoA acetyltransferase 1
- ACSS2
acetyl-CoA synthetase short-chain family member 2
- AKT1
AKT serine/threonine kinase 1
- AMPK
AMP-activated protein kinase
- dNTP
deoxy-ribonucleoside triphosphate
- EA
embonic acid
- ECAR
extracellular acidification rate
- ETV4
ETS variant transcription factor 4
- GAPDH
glyceraldehyde-3-phosphate dehydrogenase
- HMGB1
high-mobility group box 1
- HTC
hydride transfer complex
- KRAS
kirsten rat sarcoma viral oncogene homolog
- LDHA
lactate dehydrogenase A
- LKB1
liver kinase B1
- MDH1
malate dehydrogenase 1
- MDM2
murine double minute 2
- MDSA
5,5′-Methylenedisalicylic acid
- ME
malic enzyme
- ME1
malic enzyme 1
- ME2
malic enzyme 2
- ME3
malic enzyme 3
- MEs
malic enzymes
- miRNA
microRNA
- mtDNA
mitochondrial DNA
- mTORC1
mammalian target of rapamycin complex 1
- MYC
MYC proto-oncogene, bHLH transcription factor
- Na2EA
disodium salt of embonic acid
- NEK1
NIMA-related kinase 1
- NRF2
nuclear factor erythroid 2-related factor 2
- NF-κB
nuclear factor kappa-light-chain-enhancer of activated B cells
- OCR
oxygen consumption rate
- PC
pyruvate carboxylase
- PFKL
phosphofructokinase
- PGAM5
PGAM family member 5
- PKM2
pyruvate kinase M2
- PRMT1
protein arginine methyltransferase 1
- ROS
reactive oxygen species
- SIRT5
sirtuin 5
- SLC7A11
solute carrier family 7 member 11
- SREBP-1
sterol regulatory element binding protein 1
- TCA
tricarboxylic acid cycle
- TME
tumor microenvironment
Data availability
No data was used for the research described in the article.
References
- 1.Ochoa S., Mehler A., Kornberg A. Reversible oxidative decarboxylation of malic acid. J. Biol. Chem. 1947;167(3):871–872. [PubMed] [Google Scholar]
- 2.Olson D.G., Horl M., Fuhrer T., et al. Glycolysis without pyruvate kinase in Clostridium thermocellum. Metab. Eng. 2017;39:169–180. doi: 10.1016/j.ymben.2016.11.011. [DOI] [PubMed] [Google Scholar]
- 3.Shin K.S., Lee S.K. Introduction of an acetyl-CoA carboxylation bypass into Escherichia coli for enhanced free fatty acid production. Bioresour. Technol. 2017;245(Pt B):1627–1633. doi: 10.1016/j.biortech.2017.05.169. [DOI] [PubMed] [Google Scholar]
- 4.Begara-Morales J.C., Sanchez-Calvo B., Gomez-Rodriguez M.V., et al. Short-term low temperature induces nitro-oxidative stress that deregulates the NADP-malic enzyme function by tyrosine nitration in Arabidopsis thaliana. Antioxidants. 2019;8(10) doi: 10.3390/antiox8100448. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 5.Wang L., Zhang H., Zhang Y., et al. (13)C metabolic flux analysis on roles of malate transporter in lipid accumulation of Mucor circinelloides. Microb. Cell Factories. 2019;18(1):154. doi: 10.1186/s12934-019-1207-9. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 6.Hidese R., Matsuda M., Osanai T., et al. Malic enzyme facilitates d-lactate production through increased pyruvate supply during anoxic dark fermentation in synechocystis sp. PCC 6803. ACS Synth. Biol. 2020;9(2):260–268. doi: 10.1021/acssynbio.9b00281. [DOI] [PubMed] [Google Scholar]
- 7.Fazili A.B.A., Shah A.M., Albeshr M.F., et al. Overexpression of the mitochondrial malic enzyme genes (malC and malD) improved the lipid accumulation in mucor circinelloides WJ11. Front. Microbiol. 2022;13 doi: 10.3389/fmicb.2022.919364. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 8.Wu B., Li P., Hong X., et al. The receptor-like cytosolic kinase RIPK activates NADP-malic enzyme 2 to generate NADPH for fueling ROS production. Mol. Plant. 2022;15(5):887–903. doi: 10.1016/j.molp.2022.03.003. [DOI] [PubMed] [Google Scholar]
- 9.Yang H., Lu L., Chen Y., et al. Transcriptomic analysis reveals the response of the bacterium priestia aryabhattai SK1-7 to interactions and dissolution with potassium feldspar. Appl. Environ. Microbiol. 2023;89(5) doi: 10.1128/aem.02034-22. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 10.Chang G.G., Tong L. Structure and function of malic enzymes, a new class of oxidative decarboxylases. Biochemistry. 2003;42(44):12721–12733. doi: 10.1021/bi035251+. [DOI] [PubMed] [Google Scholar]
- 11.Pongratz R.L., Kibbey R.G., Shulman G.I., et al. Cytosolic and mitochondrial malic enzyme isoforms differentially control insulin secretion. J. Biol. Chem. 2007;282(1):200–207. doi: 10.1074/jbc.M602954200. [DOI] [PubMed] [Google Scholar]
- 12.Vacanti N.M., Divakaruni A.S., Green C.R., et al. Regulation of substrate utilization by the mitochondrial pyruvate carrier. Mol. Cell. 2014;56(3):425–435. doi: 10.1016/j.molcel.2014.09.024. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 13.Liu L., Shah S., Fan J., et al. Malic enzyme tracers reveal hypoxia-induced switch in adipocyte NADPH pathway usage. Nat. Chem. Biol. 2016;12(5):345–352. doi: 10.1038/nchembio.2047. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 14.Lahey R., Carley A.N., Wang X., et al. Enhanced redox state and efficiency of glucose oxidation with miR based suppression of maladaptive NADPH-dependent malic enzyme 1 expression in hypertrophied hearts. Circ. Res. 2018;122(6):836–845. doi: 10.1161/CIRCRESAHA.118.312660. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 15.Yiew N.K.H., Vazquez J.H., Martino M.R., et al. Hepatic pyruvate and alanine metabolism are critical and complementary for maintenance of antioxidant capacity and resistance to oxidative insult. Mol. Metabol. 2023;77 doi: 10.1016/j.molmet.2023.101808. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 16.Dey P., Baddour J., Muller F., et al. Genomic deletion of malic enzyme 2 confers collateral lethality in pancreatic cancer. Nature. 2017;542(7639):119–123. doi: 10.1038/nature21052. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 17.Fang X., Zhang J., Li Y., et al. Malic enzyme 1 as a novel anti-ferroptotic regulator in hepatic ischemia/reperfusion injury. Adv. Sci. 2023;10(13) doi: 10.1002/advs.202205436. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 18.Ryan E.M., Sadiku P., Coelho P., et al. NRF2 activation reprograms defects in oxidative metabolism to restore macrophage function in chronic obstructive pulmonary disease. Am. J. Respir. Crit. Care Med. 2023;207(8):998–1011. doi: 10.1164/rccm.202203-0482OC. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 19.Zhang F., Yang S., Jiang L., et al. Melatonin-mediated malic enzyme 2 orchestrates mitochondrial fusion and respiratory functions to promote odontoblastic differentiation during tooth development. J. Pineal Res. 2023;74(4) doi: 10.1111/jpi.12865. [DOI] [PubMed] [Google Scholar]
- 20.Zhu Y., Gu L., Lin X., et al. Dynamic regulation of ME1 phosphorylation and acetylation affects lipid metabolism and colorectal tumorigenesis. Mol. Cell. 2020;77(1):138–149 e5. doi: 10.1016/j.molcel.2019.10.015. [DOI] [PubMed] [Google Scholar]
- 21.Lee D., Zhang M.S., Tsang F.H., et al. Adaptive and constitutive activations of malic enzymes confer liver cancer multilayered protection against reactive oxygen species. Hepatology. 2021;74(2):776–796. doi: 10.1002/hep.31761. [DOI] [PubMed] [Google Scholar]
- 22.Li W., Kou J., Zhang Z., et al. Cellular redox homeostasis maintained by malic enzyme 2 is essential for MYC-driven T cell lymphomagenesis. Proc. Natl. Acad. Sci. U. S. A. 2023;120(23) doi: 10.1073/pnas.2217869120. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 23.Teng P., Cui K., Yao S., et al. SIRT5-mediated ME2 desuccinylation promotes cancer growth by enhancing mitochondrial respiration. Cell Death Differ. 2024;31(1):65–77. doi: 10.1038/s41418-023-01240-y. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 24.Igelmann S., Lessard F., Uchenunu O., et al. A hydride transfer complex reprograms NAD metabolism and bypasses senescence. Mol. Cell. 2021;81(18):3848–3865 e19. doi: 10.1016/j.molcel.2021.08.028. [DOI] [PubMed] [Google Scholar]
- 25.Brashears C.B., Prudner B.C., Rathore R., et al. Malic enzyme 1 absence in synovial sarcoma shifts antioxidant system dependence and increases sensitivity to ferroptosis induction with ACXT-3102. Clin. Cancer Res. 2022;28(16):3573–3589. doi: 10.1158/1078-0432.CCR-22-0470. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 26.Gicobi J.K., Mao Z., Defranco G., et al. Salvage therapy expands highly cytotoxic and metabolically fit resilient CD8(+) T cells via ME1 up-regulation. Sci. Adv. 2023;9(46) doi: 10.1126/sciadv.adi2414. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 27.Hsieh J.Y., Chen K.C., Wang C.H., et al. Suppression of the human malic enzyme 2 modifies energy metabolism and inhibits cellular respiration. Commun. Biol. 2023;6(1):548. doi: 10.1038/s42003-023-04930-y. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 28.Frenkel R. Regulation and physiological functions of malic enzymes. Curr. Top. Cell. Regul. 1975;9:157–181. doi: 10.1016/b978-0-12-152809-6.50012-3. [DOI] [PubMed] [Google Scholar]
- 29.Chen L., Zhang Z., Hoshino A., et al. NADPH production by the oxidative pentose-phosphate pathway supports folate metabolism. 2019;1(3):404–415. [PMC free article] [PubMed] [Google Scholar]
- 30.Balsa E., Perry E.A., Bennett C.F., et al. Defective NADPH production in mitochondrial disease complex I causes inflammation and cell death. Nat. Commun. 2020;11(1):2714. doi: 10.1038/s41467-020-16423-1. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 31.Simmen F.A., Pabona J.M.P., Al-Dwairi A., et al. Malic enzyme 1 (ME1) promotes adiposity and hepatic steatosis and induces circulating insulin and leptin in obese female mice. Int. J. Mol. Sci. 2023;24(7) doi: 10.3390/ijms24076613. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 32.Wang Y.P., Sharda A., Xu S.N., et al. Malic enzyme 2 connects the Krebs cycle intermediate fumarate to mitochondrial biogenesis. Cell Metabol. 2021;33(5):1027–1041 e8. doi: 10.1016/j.cmet.2021.03.003. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 33.Zhao M., Yao P., Mao Y., et al. Malic enzyme 2 maintains protein stability of mutant p53 through 2-hydroxyglutarate. Nat. Metab. 2022;4(2):225–238. doi: 10.1038/s42255-022-00532-w. [DOI] [PubMed] [Google Scholar]
- 34.Chen T., Xie S., Cheng J., et al. AKT1 phosphorylation of cytoplasmic ME2 induces a metabolic switch to glycolysis for tumorigenesis. Nat. Commun. 2024;15(1):686. doi: 10.1038/s41467-024-44772-8. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 35.Jiang P., Du W., Mancuso A., et al. Reciprocal regulation of p53 and malic enzymes modulates metabolism and senescence. Nature. 2013;493(7434):689–693. doi: 10.1038/nature11776. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 36.Xu Y., Bhargava G., Wu H., et al. Crystal structure of human mitochondrial NAD (P)+-dependent malic enzyme: a new class of oxidative decarboxylases. 1999;7(8):877–889. [PubMed] [Google Scholar]
- 37.Hsieh J.Y., Li S.Y., Chen M.C., et al. Structural characteristics of the nonallosteric human cytosolic malic enzyme. Biochim. Biophys. Acta. 2014;1844(10):1773–1783. doi: 10.1016/j.bbapap.2014.06.019. [DOI] [PubMed] [Google Scholar]
- 38.Grell T. a J., Mason M., Thompson A.A., et al. Integrative structural and functional analysis of human malic enzyme 3: a potential therapeutic target for pancreatic cancer. Heliyon. 2022;8(12) doi: 10.1016/j.heliyon.2022.e12392. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 39.Yang Z., Lanks C.W., Tong L. Molecular mechanism for the regulation of human mitochondrial NAD(P)+-dependent malic enzyme by ATP and fumarate. Structure. 2002;10(7):951–960. doi: 10.1016/s0969-2126(02)00788-8. [DOI] [PubMed] [Google Scholar]
- 40.Hung H.C., Kuo M.W., Chang G.G., et al. Characterization of the functional role of allosteric site residue Asp102 in the regulatory mechanism of human mitochondrial NAD(P)+-dependent malate dehydrogenase (malic enzyme) Biochem. J. 2005;392(Pt 1):39–45. doi: 10.1042/BJ20050641. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 41.Hsieh J.Y., Hung H.C. Engineering of the cofactor specificities and isoform-specific inhibition of malic enzyme. J. Biol. Chem. 2009;284(7):4536–4544. doi: 10.1074/jbc.M807008200. [DOI] [PubMed] [Google Scholar]
- 42.Hsieh J.Y., Shih W.T., Kuo Y.H., et al. Functional roles of metabolic intermediates in regulating the human mitochondrial NAD(P)(+)-Dependent malic enzyme. Sci. Rep. 2019;9(1):9081. doi: 10.1038/s41598-019-45282-0. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 43.Lu Y.X., Ju H.Q., Liu Z.X., et al. ME1 regulates NADPH homeostasis to promote gastric cancer growth and metastasis. Cancer Res. 2018;78(8):1972–1985. doi: 10.1158/0008-5472.CAN-17-3155. [DOI] [PubMed] [Google Scholar]
- 44.Lambert S.A., Jolma A., Campitelli L.F., et al. The human transcription factors. Cell. 2018;172(4):650–665. doi: 10.1016/j.cell.2018.01.029. [DOI] [PubMed] [Google Scholar]
- 45.Ulasov A.V., Rosenkranz A.A., Sobolev A.S. Transcription factors: time to deliver. J. Contr. Release. 2018;269:24–35. doi: 10.1016/j.jconrel.2017.11.004. [DOI] [PubMed] [Google Scholar]
- 46.Saitoh M. Transcriptional regulation of EMT transcription factors in cancer. Semin. Cancer Biol. 2023;97:21–29. doi: 10.1016/j.semcancer.2023.10.001. [DOI] [PubMed] [Google Scholar]
- 47.Liu S.H., Hsu K.W., Lai Y.L., et al. Systematic identification of clinically relevant miRNAs for potential miRNA-based therapy in lung adenocarcinoma. Mol. Ther. Nucleic Acids. 2021;25:1–10. doi: 10.1016/j.omtn.2021.04.020. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 48.Cocks A., Del Vecchio F., Martinez-Rodriguez V., et al. Pro-tumoral functions of tumor-associated macrophage EV-miRNA. Semin. Cancer Biol. 2022;86(Pt 1):58–63. doi: 10.1016/j.semcancer.2021.08.001. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 49.Dragomir M.P., Knutsen E., Calin G.A. Classical and noncanonical functions of miRNAs in cancers. Trends Genet. 2022;38(4):379–394. doi: 10.1016/j.tig.2021.10.002. [DOI] [PubMed] [Google Scholar]
- 50.Shen H., Xing C., Cui K., et al. MicroRNA-30a attenuates mutant KRAS-driven colorectal tumorigenesis via direct suppression of ME1. Cell Death Differ. 2017;24(7):1253–1262. doi: 10.1038/cdd.2017.63. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 51.Jiang Z., Cui H., Zeng S., et al. miR-885-5p inhibits invasion and metastasis in gastric cancer by targeting malic enzyme 1. DNA Cell Biol. 2021;40(5):694–705. doi: 10.1089/dna.2020.6478. [DOI] [PubMed] [Google Scholar]
- 52.Xu Q., Yu B., Chen W., et al. CircSERPINA3 promoted cell proliferation, migration, and invasion of laryngeal squamous cell carcinoma by targeting miR-885-5p. Cell Biol. Int. 2022;46(11):1852–1863. doi: 10.1002/cbin.11872. [DOI] [PubMed] [Google Scholar]
- 53.Choo J., Heo G., Pothoulakis C., et al. Posttranslational modifications as therapeutic targets for intestinal disorders. Pharmacol. Res. 2021;165 doi: 10.1016/j.phrs.2020.105412. [DOI] [PubMed] [Google Scholar]
- 54.Lee J.M., Hammaren H.M., Savitski M.M., et al. Control of protein stability by post-translational modifications. Nat. Commun. 2023;14(1):201. doi: 10.1038/s41467-023-35795-8. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 55.Wang Y., Hu J., Wu S., et al. Targeting epigenetic and posttranslational modifications regulating ferroptosis for the treatment of diseases. Signal Transduct. Targeted Ther. 2023;8(1):449. doi: 10.1038/s41392-023-01720-0. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 56.Li W., Kou J., Qin J., et al. NADPH levels affect cellular epigenetic state by inhibiting HDAC3–Ncor complex. Nat. Metab. 2021;3(1):75–89. doi: 10.1038/s42255-020-00330-2. [DOI] [PubMed] [Google Scholar]
- 57.Jia X., Liu H., Ren X., et al. Nucleolar protein NOC4L inhibits tumorigenesis and progression by attenuating SIRT1-mediated p53 deacetylation. Oncogene. 2022;41(39):4474–4484. doi: 10.1038/s41388-022-02447-y. [DOI] [PubMed] [Google Scholar]
- 58.Li Y., Bie J., Zhao L., et al. SLC25A51 promotes tumor growth through sustaining mitochondria acetylation homeostasis and proline biogenesis. Cell Death Differ. 2023;30(8):1916–1930. doi: 10.1038/s41418-023-01185-2. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 59.Li D.J., Cheng Y.W., Pan J.M., et al. KAT8/SIRT7-mediated Fascin-K41 acetylation/deacetylation regulates tumor metastasis in esophageal squamous cell carcinoma. J. Pathol. 2024;263:74–88. doi: 10.1002/path.6261. [DOI] [PubMed] [Google Scholar]
- 60.He Y., Korboukh I., Jin J., et al. Targeting protein lysine methylation and demethylation in cancers. Acta Biochim. Biophys. Sin. 2012;44(1):70–79. doi: 10.1093/abbs/gmr109. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 61.Huang L., Zhang X.O., Rozen E.J., et al. PRMT5 activates AKT via methylation to promote tumor metastasis. Nat. Commun. 2022;13(1):3955. doi: 10.1038/s41467-022-31645-1. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 62.Zheng Q., Zhang W., Rao G.W. Protein lysine methyltransferase SMYD2: a promising small molecule target for cancer therapy. J. Med. Chem. 2022;65(15):10119–10132. doi: 10.1021/acs.jmedchem.2c00325. [DOI] [PubMed] [Google Scholar]
- 63.Bringman-Rodenbarger L.R., Guo A.H., Lyssiotis C.A., et al. Emerging roles for SIRT5 in metabolism and cancer. Antioxidants Redox Signal. 2018;28(8):677–690. doi: 10.1089/ars.2017.7264. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 64.Liu X., Rong F., Tang J., et al. Repression of p53 function by SIRT5-mediated desuccinylation at Lysine 120 in response to DNA damage. Cell Death Differ. 2022;29(4):722–736. doi: 10.1038/s41418-021-00886-w. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 65.Hu Q., Xu J., Wang L., et al. SUCLG2 regulates mitochondrial dysfunction through succinylation in lung adenocarcinoma. Adv. Sci. 2023;10(35) doi: 10.1002/advs.202303535. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 66.Yao P., Sun H., Xu C., et al. Evidence for a direct cross-talk between malic enzyme and the pentose phosphate pathway via structural interactions. J. Biol. Chem. 2017;292(41):17113–17120. doi: 10.1074/jbc.M117.810309. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 67.Rho S.B., Byun H.J., Kim B.R., et al. Liver kinase B1 mediates its anti-tumor function by binding to the N-terminus of malic enzyme 3. Biomol Ther (Seoul) 2023;31(3):330–339. doi: 10.4062/biomolther.2023.041. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 68.Hanahan D., Weinberg R.A. Hallmarks of cancer: the next generation. Cell. 2011;144(5):646–674. doi: 10.1016/j.cell.2011.02.013. [DOI] [PubMed] [Google Scholar]
- 69.Li Z., Sun C., Qin Z. Metabolic reprogramming of cancer-associated fibroblasts and its effect on cancer cell reprogramming. Theranostics. 2021;11(17):8322–8336. doi: 10.7150/thno.62378. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 70.Raggi C., Taddei M.L., Rae C., et al. Metabolic reprogramming in cholangiocarcinoma. J. Hepatol. 2022;77(3):849–864. doi: 10.1016/j.jhep.2022.04.038. [DOI] [PubMed] [Google Scholar]
- 71.Vriens K., Christen S., Parik S., et al. Evidence for an alternative fatty acid desaturation pathway increasing cancer plasticity. Nature. 2019;566(7744):403–406. doi: 10.1038/s41586-019-0904-1. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 72.Faubert B., Solmonson A., Deberardinis R.J. Metabolic reprogramming and cancer progression. Science. 2020;368(6487) doi: 10.1126/science.aaw5473. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 73.Warburg O.J.S. On the origin of cancer cells. 1956;123(3191):309–314. doi: 10.1126/science.123.3191.309. [DOI] [PubMed] [Google Scholar]
- 74.Hou Y., Zhang Q., Pang W., et al. YTHDC1-mediated augmentation of miR-30d in repressing pancreatic tumorigenesis via attenuation of RUNX1-induced transcriptional activation of Warburg effect. Cell Death Differ. 2021;28(11):3105–3124. doi: 10.1038/s41418-021-00804-0. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 75.Yue S.W., Liu H.L., Su H.F., et al. m6A-regulated tumor glycolysis: new advances in epigenetics and metabolism. Mol. Cancer. 2023;22(1):137. doi: 10.1186/s12943-023-01841-8. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 76.Liao R., Ren G., Liu H., et al. ME1 promotes basal-like breast cancer progression and associates with poor prognosis. Sci. Rep. 2018;8(1) doi: 10.1038/s41598-018-35106-y. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 77.Chen K.C., Hsiao I.H., Huang Y.N., et al. Targeting human mitochondrial NAD(P)(+)-dependent malic enzyme (ME2) impairs energy metabolism and redox state and exhibits antileukemic activity in acute myeloid leukemia. Cell. Oncol. 2023;46(5):1301–1316. doi: 10.1007/s13402-023-00812-x. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 78.Jin H.R., Wang J., Wang Z.J., et al. Lipid metabolic reprogramming in tumor microenvironment: from mechanisms to therapeutics. J. Hematol. Oncol. 2023;16(1):103. doi: 10.1186/s13045-023-01498-2. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 79.Xu K., Xia P., Chen X., et al. ncRNA-mediated fatty acid metabolism reprogramming in HCC. Trends Endocrinol. Metabol. 2023;34(5):278–291. doi: 10.1016/j.tem.2023.02.007. [DOI] [PubMed] [Google Scholar]
- 80.Yang M., Chen X., Zhang J., et al. ME2 promotes proneural-mesenchymal transition and lipogenesis in glioblastoma. Front. Oncol. 2021;11 doi: 10.3389/fonc.2021.715593. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 81.Fu G., Li S.T., Jiang Z., et al. PGAM5 deacetylation mediated by SIRT2 facilitates lipid metabolism and liver cancer proliferation. Acta Biochim. Biophys. Sin. 2023;55(9):1370–1379. doi: 10.3724/abbs.2023155. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 82.Ran M., Zhou Y., Guo Y., et al. Cytosolic malic enzyme and glucose-6-phosphate dehydrogenase modulate redox balance in NSCLC with acquired drug resistance. FEBS J. 2023;290(19):4792–4809. doi: 10.1111/febs.16897. [DOI] [PubMed] [Google Scholar]
- 83.Gorrini C., Harris I.S., Mak T.W. Modulation of oxidative stress as an anticancer strategy. Nat. Rev. Drug Discov. 2013;12(12):931–947. doi: 10.1038/nrd4002. [DOI] [PubMed] [Google Scholar]
- 84.Li Y., Li B., Xu Y., et al. GOT2 silencing promotes reprogramming of glutamine metabolism and sensitizes hepatocellular carcinoma to glutaminase inhibitors. Cancer Res. 2022;82(18):3223–3235. doi: 10.1158/0008-5472.CAN-22-0042. [DOI] [PubMed] [Google Scholar]
- 85.Murai S., Ando A., Ebara S., et al. Inhibition of malic enzyme 1 disrupts cellular metabolism and leads to vulnerability in cancer cells in glucose-restricted conditions. Oncogenesis. 2017;6(5):e329. doi: 10.1038/oncsis.2017.34. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 86.Ye Z., Liu W., Zhuo Q., et al. Ferroptosis: final destination for cancer? Cell Prolif. 2020;53(3) doi: 10.1111/cpr.12761. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 87.Wang H., Lin D., Yu Q., et al. A promising future of ferroptosis in tumor therapy. Front. Cell Dev. Biol. 2021;9 doi: 10.3389/fcell.2021.629150. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 88.Kim R., Taylor D., Vonderheide R.H., et al. Ferroptosis of immune cells in the tumor microenvironment. Trends Pharmacol. Sci. 2023;44(8):542–552. doi: 10.1016/j.tips.2023.06.005. [DOI] [PubMed] [Google Scholar]
- 89.Gorgoulis V., Adams P.D., Alimonti A., et al. Cellular senescence: defining a path forward. Cell. 2019;179(4):813–827. doi: 10.1016/j.cell.2019.10.005. [DOI] [PubMed] [Google Scholar]
- 90.Wang B., Kohli J., Demaria M. Senescent cells in cancer therapy: friends or foes? Trends. Cancer. 2020;6(10):838–857. doi: 10.1016/j.trecan.2020.05.004. [DOI] [PubMed] [Google Scholar]
- 91.Schmitt C.A., Wang B., Demaria M. Senescence and cancer - role and therapeutic opportunities. Nat. Rev. Clin. Oncol. 2022;19(10):619–636. doi: 10.1038/s41571-022-00668-4. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 92.Zhang S., Cheng Z.M., Yu J.L., et al. Malic enzyme 2 promotes the progression of hepatocellular carcinoma via increasing triglyceride production. Cancer Med. 2021;10(19):6795–6806. doi: 10.1002/cam4.4209. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 93.Yang Y., Zhang Z., Li W., et al. alphaKG-driven RNA polymerase II transcription of cyclin D1 licenses malic enzyme 2 to promote cell-cycle progression. Cell Rep. 2023;42(7) doi: 10.1016/j.celrep.2023.112770. [DOI] [PubMed] [Google Scholar]
- 94.Binnewies M., Roberts E.W., Kersten K., et al. Understanding the tumor immune microenvironment (TIME) for effective therapy. Nat. Med. 2018;24(5):541–550. doi: 10.1038/s41591-018-0014-x. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 95.Fu T., Dai L.J., Wu S.Y., et al. Spatial architecture of the immune microenvironment orchestrates tumor immunity and therapeutic response. J. Hematol. Oncol. 2021;14(1):98. doi: 10.1186/s13045-021-01103-4. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 96.Gdynia G., Sauer S.W., Kopitz J., et al. The HMGB1 protein induces a metabolic type of tumour cell death by blocking aerobic respiration. Nat. Commun. 2016;7 doi: 10.1038/ncomms10764. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 97.Charpentier M., Spada S., Van Nest S.J., et al. Radiation therapy-induced remodeling of the tumor immune microenvironment. Semin. Cancer Biol. 2022;86(Pt 2):737–747. doi: 10.1016/j.semcancer.2022.04.003. [DOI] [PubMed] [Google Scholar]
- 98.Xu L., Zou C., Zhang S., et al. Reshaping the systemic tumor immune environment (STIE) and tumor immune microenvironment (TIME) to enhance immunotherapy efficacy in solid tumors. J. Hematol. Oncol. 2022;15(1):87. doi: 10.1186/s13045-022-01307-2. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 99.Nussinov R., Tsai C.J., Jang H. Anticancer drug resistance: an update and perspective. Drug Resist. Updates. 2021;59 doi: 10.1016/j.drup.2021.100796. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 100.Zhou H.M., Zhang J.G., Zhang X., et al. Targeting cancer stem cells for reversing therapy resistance: mechanism, signaling, and prospective agents. Signal Transduct. Targeted Ther. 2021;6(1):62. doi: 10.1038/s41392-020-00430-1. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 101.Cheng J., Xie W., Chen Y., et al. Drug resistance mechanisms in dopamine agonist-resistant prolactin pituitary neuroendocrine tumors and exploration for new drugs. Drug Resist. Updates. 2024;73 doi: 10.1016/j.drup.2024.101056. [DOI] [PubMed] [Google Scholar]
- 102.Nakashima C., Fujiwara-Tani R., Mori S., et al. An Axis between the long non-coding RNA HOXA11-AS and NQOs enhances metastatic ability in oral squamous cell carcinoma. Int. J. Mol. Sci. 2022;23(18) doi: 10.3390/ijms231810704. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 103.Huang D., Zhang C., Xiao M., et al. Redox metabolism maintains the leukemogenic capacity and drug resistance of AML cells. Proc. Natl. Acad. Sci. U. S. A. 2023;120(13) doi: 10.1073/pnas.2210796120. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 104.Chakrabarti G. Mutant KRAS associated malic enzyme 1 expression is a predictive marker for radiation therapy response in non-small cell lung cancer. Radiat. Oncol. 2015;10:145. doi: 10.1186/s13014-015-0457-x. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 105.Zhang Y.J., Wang Z., Sprous D., et al. In silico design and synthesis of piperazine-1-pyrrolidine-2,5-dione scaffold-based novel malic enzyme inhibitors. Bioorg. Med. Chem. Lett. 2006;16(3):525–528. doi: 10.1016/j.bmcl.2005.10.065. [DOI] [PubMed] [Google Scholar]
- 106.Yoshida T., Kawabe T., Cantley L.C., et al. Discovery and characterization of a novel allosteric small-molecule inhibitor of NADP(+)-Dependent malic enzyme 1. Biochemistry. 2022;61(15):1548–1553. doi: 10.1021/acs.biochem.2c00123. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 107.Wen Y., Xu L., Chen F.-L., et al. Discovery of a novel inhibitor of NAD(P)+-dependent malic enzyme (ME2) by high-throughput screening. Acta Pharmacol. Sin. 2014;35(5):674–684. doi: 10.1038/aps.2013.189. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 108.Sheth G., Shah S.R., Sengupta P., et al. In the quest for potent and selective malic enzyme 3 inhibitors for the treatment of pancreatic ductal adenocarcinoma. ACS Med. Chem. Lett. 2023;14(1):41–50. doi: 10.1021/acsmedchemlett.2c00369. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 109.Mihara Y., Akiba J., Ogasawara S., et al. Malic enzyme 1 is a potential marker of combined hepatocellular cholangiocarcinoma, subtype with stem-cell features, intermediate-cell type. Hepatol. Res. 2019;49(9):1066–1075. doi: 10.1111/hepr.13365. [DOI] [PubMed] [Google Scholar]
- 110.Nakashima C., Kirita T., Yamamoto K., et al. Malic enzyme 1 is associated with tumor budding in oral squamous cell carcinomas. Int. J. Mol. Sci. 2020;21(19) doi: 10.3390/ijms21197149. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 111.Nakashima C., Yamamoto K., Fujiwara-Tani R., et al. Expression of cytosolic malic enzyme (ME1) is associated with disease progression in human oral squamous cell carcinoma. Cancer Sci. 2018;109(6):2036–2045. doi: 10.1111/cas.13594. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 112.Kourtidis A., Jain R., Carkner R.D., et al. An RNA interference screen identifies metabolic regulators NR1D1 and PBP as novel survival factors for breast cancer cells with the ERBB2 signature. Cancer Res. 2010;70(5):1783–1792. doi: 10.1158/0008-5472.CAN-09-1550. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 113.Ying M., You D., Zhu X., et al. Lactate and glutamine support NADPH generation in cancer cells under glucose deprived conditions. Redox Biol. 2021;46 doi: 10.1016/j.redox.2021.102065. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 114.Yang Y., Zhang Z., Li W., et al. ME2 promotes hepatocellular carcinoma cell migration through pyruvate. Metabolites. 2023;13(4) doi: 10.3390/metabo13040540. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 115.Chang Y.L., Gao H.W., Chiang C.P., et al. Human mitochondrial NAD(P)(+)-dependent malic enzyme participates in cutaneous melanoma progression and invasion. J. Invest. Dermatol. 2015;135(3):807–815. doi: 10.1038/jid.2014.385. [DOI] [PubMed] [Google Scholar]
- 116.Zhou J.J., Xiao Y., Li H., et al. Overexpression of malic enzyme 2 indicates pathological and clinical significance in oral squamous cell carcinoma. Int. J. Med. Sci. 2020;17(6):799–806. doi: 10.7150/ijms.43832. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 117.You D., Du D., Zhao X., et al. Mitochondrial malic enzyme 2 promotes breast cancer metastasis via stabilizing HIF-1alpha under hypoxia. Chin. J. Cancer Res. 2021;33(3):308–322. doi: 10.21147/j.issn.1000-9604.2021.03.03. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 118.Wu C.C., Li H., Xiao Y., et al. Expression levels of SIX1, ME2, and AP2M1 in adenoid cystic carcinoma and mucoepidermoid carcinoma. Oral Dis. 2020;26(8):1687–1695. doi: 10.1111/odi.13506. [DOI] [PubMed] [Google Scholar]
- 119.Vantaku V., Donepudi S.R., Piyarathna D.W.B., et al. Large-scale profiling of serum metabolites in African American and European American patients with bladder cancer reveals metabolic pathways associated with patient survival. Cancer. 2019;125(6):921–932. doi: 10.1002/cncr.31890. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 120.Zhang Q., Li J., Tan X.P., et al. Effects of ME3 on the proliferation, invasion and metastasis of pancreatic cancer cells through epithelial-mesenchymal transition. Neoplasma. 2019;66(6):896–907. doi: 10.4149/neo_2019_190119N59. [DOI] [PubMed] [Google Scholar]
Associated Data
This section collects any data citations, data availability statements, or supplementary materials included in this article.
Data Availability Statement
Not applicable.
No data was used for the research described in the article.







