Open Access

Glutathione‑degrading enzymes in the complex landscape of tumors (Review)

  • This article is part of the special Issue: Going to the discovery of new biomarkers in cancer
  • Authors:
    • Tianyi Zhang
    • Chongjie Yao
    • Xu Zhou
    • Shimin Liu
    • Li Qi
    • Shiguo Zhu
    • Chen Zhao
    • Dan Hu
    • Weidong Shen
  • View Affiliations

  • Published online on: June 3, 2024     https://doi.org/10.3892/ijo.2024.5660
  • Article Number: 72
  • Copyright: © Zhang et al. This is an open access article distributed under the terms of Creative Commons Attribution License.

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


Abstract

Glutathione (GSH)‑degrading enzymes are essential for starting the first stages of GSH degradation. These enzymes include extracellular γ‑glutamyl transpeptidase (GGT) and intracellular GSH‑specific γ‑glutamylcyclotransferase 1 (ChaC1) and 2. These enzymes are essential for cellular activities, such as immune response, differentiation, proliferation, homeostasis regulation and programmed cell death. Tumor tissue frequently exhibits abnormal expression of GSH‑degrading enzymes, which has a key impact on the development and spread of malignancies. The present review summarizes gene and protein structure, catalytic activity and regulation of GSH‑degrading enzymes, their vital roles in tumor development (including regulation of oxidative and endoplasmic reticulum stress, control of programmed cell death, promotion of inflammation and tumorigenesis and modulation of drug resistance in tumor cells) and potential role as diagnostic biomarkers and therapeutic targets.

1. Introduction

Glutathione (GSH), also known as γ-glutamylcysteylglycine, is the most common small molecular weight thiol molecule generated in living cells (1). It is extensively present in all eukaryotes and is particularly concentrated in the liver. GSH manifests in two forms, mercaptan reduction (GSH) and disulfide oxidation (GSSG). GSH is the more common form, with a concentration >100 times higher than that of GSSG (2). Synthesized from glutamic acid, cysteine, and glycine, GSH serves a pivotal role in the pathway using niacinamide adenine dinucleotide to establish a reducing environment key for cellular function. A previous study (3) underscored the crucial involvement of GSH in diverse cellular processes, including immunological function, cell proliferation, differentiation and programmed cell death. As a regulator agent in key signal transduction pathways, GSH is involved in maintaining cellular homeostasis. The dysregulation of GSH expression strongly correlates with onset and progression of numerous types of disease, including tumors (4), liver disease (5), diabetes (6) and neurodegenerative disease (7,8). Tumor cells require elevated GSH levels to combat reactive oxygen species (ROS) and detoxify carcinogens. Thus, decreasing intracellular GSH renders tumor cells more susceptible to oxidative stress and chemotherapeutic drugs (9).

While existing studies (2,4) predominantly focused on the anabolic aspects of GSH metabolism, the catabolic process of GSH has received limited attention (10,11). Previously, the cytoplasm was hypothesized to have no role in GSH catabolism. However, the identification of novel GSH degradation pathways in the cytosol (12,13) underscores the importance of exploring GSH degradation. GSH-degrading enzymes are essential for maintaining GSH homeostasis in cells. Dysregulation of these enzymes significantly impacts GSH homeostasis, leading to pathological changes. Such dysregulation is frequently observed in tumor tissue and has been shown to play an essential role in tumor development (14,15). Since different GSH-degrading enzymes are oriented to either intracellular or extracellular GSH pools, intracellular degrading enzymes directly decrease intracellular levels of GSH. By contrast, extracellular degrading enzyme produces cysteine, providing an additional rate-limiting amino acid for resynthesis of intracellular GSH (16). Therefore, different GSH-degrading enzymes exhibit different effects on cancer, either promoting or suppressing it, and their specific functions varies according to the type of tissue and tumor. In addition, the levels of GSH degradation products glutamic acid, cysteine and glycine serve as growth factors, proliferation stimulators and signal transducers of tumor cells (17,18). Thus, understanding of GSH-degrading enzymes and their roles in cancer is imperative for developing more effective therapeutic interventions.

The present review summarizes the crucial role of enzymes in GSH degradation in tumors, as well as their potential as biomarkers and targets for tumor therapy and their potential directions for clinical translation in tumor therapies.

2. Extracellular and intracellular GSH-degrading enzymes

Initiation steps in the mammalian GSH degradation pathway fall into two categories, intracellular and extracellular degradation (Fig. 1) (10). The first category is classical GSH degradation, which commences with extracellular enzyme γ-glutamyl transpeptidase (GGT) (11). Intracellular GSH is released into the extracellular space via multidrug-resistant protein 1-mediated transporter (19). Once outside the cell, GSH is hydrolyzed to Cys-Gly and glutamate by plasma membrane-bound GGT, marking the initial step in extracellular GSH degradation. Discoveries in the cytoplasmic cation transport regulator homolog (ChaC) family of γ-glutamylcyclotransferases have expanded the understanding of GSH degradation (12,13). This family, including ChaC1 and ChaC2, directly breaks down GSH within the cell into Cys-Gly and 5-oxoproline (11,20).

GGT: Classical perspective

GGT, a core component of the γ-glutamyl cycle (21), has long been associated with GSH degradation, predating the discovery of the ChaC family. Initially considered the sole enzyme capable of degrading GSH, GGT hydrolyzes the γ-glutamyl bond of extracellular reduced and oxidized GSH (22). This results in cleaved glutamate, cysteine and glycine while facilitating the transfer of γ-glutamyl moiety of GSH to either water (hydrolysis) or substrates such as peptides (transpeptidation). Consequently, GGT is classified as a bisubstrate enzyme (23).

GGT is a glycosylated heterodimer protein formed by the non-covalent combination of a heavy chain subunit (relative molecular mass, 50,000-62,000) and a light chain subunit (relative molecular mass 22,000-30,000; Fig. 2) (24). The human GGT family members are synthesized and cleaved by an autocatalytic processing reaction. They have a conserved 'sandwich-like' three-dimensional domain with four layers of αββα folds (23). The catalytic site of GGT consists of two successive regions: Well-characterized donor site, specifying the substrates to which donor γ-glutamyl groups bind, and the acceptor site, about which little is currently known regarding the involved residues (24).

Anchored in the plasma membrane by the N-terminus of the heavy chain across the membrane segment, GGT protein, under physiological conditions, is typically confined to the plasma membrane. It is distributed on the apical surface of epithelial and endothelial cells in glands and lumens. A unique characteristic of GGT is that it is located on the extracellular surface of mammalian cells with a catalytic active site oriented to the extracellular environment (11). The kidney expresses GGT at the highest levels, while notable expression is also found in bile canaliculi of hepatocytes, ducts within the pancreas, the apical surface of the intestinal epithelium and the luminal surface epithelium of many reproductive organs (25).

GGT gene family and proteins

A GGT gene family exists in the human genome (Table I), suggesting that regulating GGT activity may be associated with activating different GGT genes rather than identifying distinct gene loci (26).

Table I

GGT-homologous sequences.

Table I

GGT-homologous sequences.

GeneaPrevious namesFunctional proteinAbnormal expression
GGT1Gene 6; GGT type IFunctional proteinDysregulated in various tumors
GGT2Clone F15; Gene 3 (L10396); GGT type IIInactive propeptide, 94% homologous to part of GGT1Low expression in glioblastoma multiforme
GGT3PClone F11; GGT3PseudogeneNot reported
GGT4PGene 12 (L10398); clone F30PseudogeneNot reported
GGT5GGL, γ-glutamyl leukotrienase; GGTLA1/GGT-rel; GGT5 precursor; GGTLA1Functional protein, 40% homologous to GGT1, exhibits <1/46 activity of GGT1 in hydrolyzing GSH, GSSG and leukotriene C4High expression of GGT5 is beneficial to the prognosis of hepatocellular carcinoma
GGT6Rat GGT6 homologNot characterizedOverexpressed in low-grade glioma
GGT7GGTL3, GGT4, GGTL5; GC20M032896Not characterizedLow expression in gastric cancer; high expression may lead to poor overall survival in hepatocellular carcinoma; GGT7 polymorphic loci rs6119534 and rs11546155 are associated with risk of pancreatic disease
GGT8P/PseudogeneNot reported
GGTLC1GGTL6; GGTLA4; GGTLA4Encode only the light chain part of GGTNot reported
GGTLC2Gene 1 (L10394); GGTL4; GGTL4Encode only the light chain part of GGTNot reported
GGTLC3γ-glutamyl transferase light chain 3; LOC728226May encode only the light chain of GGTNot reported
GGTLC4Pγ-glutamyl transferase light chain 4 pseudogenePseudogeneNot reported
GGTLC5Pγ-glutamyl transferase light chain 5 pseudogenePseudogeneNot reported

a Heisterkamp et al (2008) combed a summary of the human γ-glutamyl transferase gene family (27). GGT, γ-glutamyl transpeptidase; GGTLA, γ-glutamyl transferase-like activity.

The human genome sequence contains 13 GGT homologs, the most active of which is GGT. The GGT gene was first discovered on human chromosome 22 at q11.1-q11.2 (27), although it was also subsequently discovered on additional autosomes (26). In addition, two homologs, GGTLC1 (previously GGTL6, GGTLA4) and GGTLC2, which may only encode the light-chain portion of GGT, as well as at least three other homologs, exhibit activity: GGT5 (formerly GGL, GGTLA1/GGT-rel), GGT6 (formerly rat GGT6 homologous) and GGT7 (formerly GGTL3, GGT4) (28). While GGT5 and GGT1 share 40% of the amino acid sequence (22), GGT5 is not as active in hydrolyzing GSH, GSSG and leukotriene C4 as GGT1 is (29). Despite the absence of verified protein-coding activity, GGT6 and GGT7 exhibit aberrant expression in conditions such as tumors (30-32) and pancreatic disease (33). Furthermore, proteins expressed by the human GGT2 gene share 94% of the amino acid sequence encoded by GGT1 (34), even though GGT2 only encodes inactive pro-peptides. GGT2 also exhibits abnormal expression in tumors (30) and upregulating GGT2 can overcome H2O2-induced apoptosis (35). These findings suggest that GGTs play potential roles in disease physiology and pathology.

Catalytic activity of GGT

As a member of the N-terminal nucleophilic hydrolase superfamily (Ntn), GGT uses a highly conserved catalytic mechanism. An N-terminal Thr residue is essential for substrate priming in human GGT (22), with the substrate binding site featuring a key Thr side chain. This facilitates conversion of the γ-glutamyl bond of GSH into an acyl bond, releasing Cys-Gly and glutamate (11).

GGT gene expression

While the human GGT gene is not fully characterized, evidence suggests its existence in multiple copies within the human genome (36). As GGT mRNAs share a common coding sequence and have 59 untranslated regions (UTRs), its structural complexity is evident (37). Understanding of the human GGT promoter remains limited (38).

A number of promoters control human GGT transcription, and the resulting transcripts undergo selective splicing in untranslated regions and coding sequences (37). The initiation of GGT mRNA transcription involves cis-reactive elements, including the cis-regulatory element (TRE) and the binding element for activator protein 1 (AP-1) (38). TRE, also known as 12-O-tetracylacylphobolol 13 acetic acid reactive element, incorporates binding sites for activating protein 2 (AP-2) and specific protein 1 (Sp1; Fig. 3).

A study (39) on HeLa cells highlighted that phorbol 12-myric acid 13-acetic acid enhance the expression of human GGT, pinpointing its binding site at the AP-1 binding site 2,214/2,225 nucleotides upstream from the transcription start site. More research is necessary to understand the transcriptional mechanism of the human GGT gene and promoters.

Although rat and human GGT promoters may have similar structures, the human promoter is more complex. As the rat GGT gene is single-copy, replicating unique genes after species transfer between rats and humans likely results in multiple human GGT genes (40). Therefore, rat GGT may provide insight into human GGT expression and regulation.

In rats, GGT expression is controlled by a tandem P1-P5 promoter, facilitated by variable splicing. This yields transcripts sharing the same coding region and diverse 5'-UTRs (38). These unique promoters have high tissue stage-specificity (38).

GGT regulation

Upregulation of GGT activity is primarily dependent on the Ras protein and its downstream effectors, which include extracellular signal-regulated kinase 1/2 (ERK1/2), p38 mitogen-activated protein kinase (MAPK), phosphatidylinositol 3-kinase (PI3K)/AKT and c-Jun N-terminal kinase (JNK) signaling pathways (Fig. 3) (41). The Ras protein is a key regulator of signaling pathways key for normal cell proliferation. Malignant phenotype of tumor cells is caused by the Ras gene, often mutated or active in tumor cells, and leads to aberrant tumor cell proliferation, programmed cell death, invasion and angiogenesis (42).

Through the electrophile response element (EpRE), ERK1/2 and p38MAPK signaling pathways actively contribute to upregulation of GGT promoter 5 (GP5) activity in response to 4-hydroxynonenal (HNE) (43). Following activation by redox signaling downstream of the MAPK signal pathway, the EpRE binding protein, also an oxidative stress-associated transcription factor, induces the dissociation of Nrf2 from Keap1 via redox modification and/or phosphorylation of Nrf2. Subsequently, Nrf2 translocates from cytoplasm to the nucleus, forming heterodimers with other proteins to bind to EpRE. This leads to the amplification of GGT transcription. In alveolar type II (L2) cells, EpRE motif in the proximal region of GP5 EpRE induces the expression of the major GGT transcript mRNA V-2 in the lung (38). However, pretreatment with ERK1/2 pathway inhibitor (PD98059) or p38MAPK inhibitor (SB203580) partly decreases the expression of GGT mRNA V-2 induced by HNE in L2 cells (38). Furthermore, activated Ras is also implicated in inducing activation of GP2 and increasing expression of GGT transcriptional products and protein in colon cancer cells treated with naphthoquinone during acute oxidative stress (Fig. 3) (41).

Moreover, inflammatory conditions significantly enhance GGT levels by activating the NF-κB pathway. NF-κB, the core transcription factor in the NF-κB signaling pathway, is a dimer family formed by p50/p105/NF-κB1, p52/p100/NF-κB2, c-Rel, p65/RelA and RelB (44). It regulates the expression of chemokines, cytokines, transcription factors and regulatory proteins, playing a crucial role in inflammation and immunity (44). Upon cell stimulation by an external signal, the NF-κB dimer is released from its inhibitor (IκB) and freely transferred into the nucleus. When the NF-κB pathway is activated, it triggers production of pro-inflammatory proteins downstream, such as tumor necrosis factor-α (TNF-α), which in turn causes inflammatory responses and pain. Furthermore, by serving as NF-κB activators, these inflammatory cytokines intensify inflammation by further triggering the NF-κB pathway (45). In the 536 bp site of the proximal promoter of GGT, a binding site exists between p50, TNF-α and Sp1, regulated by the activation of the NF-κB signaling pathway, thereby promoting expression of GGT and inducing inflammatory response (Fig. 3) (46). Moreover, it has been reported (46) that inhibitors of the NF-κB pathway can effectively block the trans-activation of GGT promoters at different levels. For example, remicade, a clinically used anti-TNF-α antibody targeting the p50 and p65 NF-κB subtype of small interfering RNA and curcumin, a well-characterized natural NF-κB inhibitor that is also a dominant negative inhibitor of κBα (IκBα), can inhibit GGT activation through distinct mechanisms. This suggests the involvement of the NF-κB pathway in regulating GGT expression. Therefore, inflammatory conditions may increase GGT synthesis, potentially acting as a cellular protective mechanism under increased oxidative stress or promoting inflammatory progression. Further research is necessary to explore these possibilities.

ChaC1/ChaC2: Additional perspective

In addition to the well-established extracellular GSH degradation mechanism, studies have shown an additional intracellular hydrolysis pathway for GSH degradation (47,48). ChaC protein features a BtrG/γ-GCT fold and distinctive β-barrels surrounded by α-helices (Fig. 4) (13). Mammals exhibit two isoforms of ChaC: Mammalian pro-apoptotic factor ChaC1 (formerly MGC4504) and its homologous counterpart ChaC2. Conversely, only one ChaC member is present in lower eukaryotes, especially in unicellular eukaryotes (47).

Gene and protein structure of ChaC1/ChaC2

The human ChaC1 gene is on chromosome 15q15.1 and comprises three exons, encoding a protein with 222 amino acid residues and a molecular weight of ~25 kDa (49). ChaC1, 30% identical to mammalian and prokaryotic genes, serves a crucial role in basic physiology (12).

ChaC2 is on chromosome 2p16.2 and encodes a protein with 184 amino acid residues and a molecular weight of ~20.9 kDa (20,47,50). A phylogenetic study (47) highlighted that ChaC2 evolved earlier than ChaC1 and shares key structural similarities with ChaC proteins of lower eukaryotes. Human ChaC2 and ChaC1 share 50% of their protein identity (47). ChaC2 typically exists in dimer crystals with a unique flexible loop 2 structure, with an open conformation that can facilitate close contact with crystallographically adjacent ChaC2 molecules. Additionally, ChaC2 E74Q/E83Q active site mutants exhibit a closed conformation, regulating the degradation activity of ChaC2 to GSH (20).

Catalytic activity of ChaC1/ChaC2

ChaC1, an inducible enzyme, can be expressed under specific stresses or pathological conditions (12,47). It selectively hydrolyzes GSH, producing Cys-Gly and 5-oxoproline (a cyclized form of glutamate) (11,51), thereby accelerating formation of the cellular oxidative environment. The Michaelis constant of ChaC1 for GSH is ~2.2±0.4 mM (47), comparable with the concentration of intracellular GSH (1-10 mM) under physiological conditions. ChaC1 often forms dimers or tetramers, with dimerization being key for regulating enzyme activity and substrate specificity. Under stress or tumor growth, there is an increased need for enzyme breakdown, which leads to formation of dimers or longer oligomers of ChaC1 (20).

ChaC2 is constitutively expressed and exhibits catalytic efficiency for GSH 10-20 times weaker than that of ChaC1 (47). The lower activity of ChaC2 may be partly attributed to flexible loop 2, acting as a gating function to achieve specificity for GSH binding and regulate a constant GSH degradation rate. In addition, the Glu74 and Glu83 residues of ChaC2 are key for directing the conformation of the enzyme and regulating enzyme activity (20).

Expression and regulation of ChaC1/ChaC2

Various signals, including endoplasmic reticulum (ER) and oxidative stress and viral infection, activate ChaC1 promoters in different cell types, cellular processes and diseases through the unfolded protein response (UPR) (12,48,52). ChaC1 is downstream of the protein kinase R-like ER kinase (PERK)/eukaryotic initiation factor-2α (eIF2α)/activating transcription factor (ATF) 4/ATF3/CCAAT/enhancer binding protein (C/EBP) homologous protein (CHOP) pathway, serving as a pro-apoptotic and pro-ferroptosis component downstream of UPR. ATF4, ATF3, CHOP and C/EBP-β upregulate ChaC1 transcription (Fig. 5). The activation of ChaC1 by UPR primarily relies on ATF4, while the involvement of CHOP, C/EBP-β, and ATF3 is indirect (12). A direct relationship between ATF4 and regulatory elements within the ChaC1 promoter has been identified. A-267 ATF/cAMP response element (CRE) in conjunction with a novel-248 ATF/CRE modifier (ACM), serving as a binding site for ATF4 and ATF3 transcription factors, regulates the activity of the basic ChaC1 promoter. Among these elements, ATF3 predominantly regulates the basal and stress-induced expression of ChaC1 through ATF/CRE, while ATF4 primarily regulates stress-induced ChaC1 expression through ATF/CRE and ACM (48). Additionally, conserved-209 CEBP-ATF response element has a limited impact on regulating human ChaC1 transcription (48,53).

Amino acid starvation can induce the expression of ChaC1 (Fig. 5). The amino acid starvation response activates ATF4 via the general control nonderepressible 2 (GCN2)/eIF2a/ATF4/ATF3 pathway (54). Both ER stress and amino acid starvation induce stress synergistically by activating ATF4 and ChaC1 is one of the downstream targets of ATF4. In regulating ChaC1 expression, C/EBP-β has been observed to recruit ATF4 to the ChaC1 promoter in response to ER stress (48). However, the precise C/EBP-β response element on the ChaC1 promoter remains unclear (48). Further studies are necessary to elucidate the detailed mechanism of C/EBP-β-mediated ATF4 recruitment and its impact on ChaC1 expression. These findings highlight the intricate nature of ChaC1 transcriptional regulation and underscore the importance of maintaining appropriate redox balance in cells (48). The mechanism governing ChaC1 protein expression requires further characterization.

Understanding of the regulation mechanism of ChaC2 is limited. A previous study (47) suggested that ChaC2 is expressed at higher basal levels under physiological conditions than ChaC1. However, under cellular stress such as ER stress or amino acid starvation, ChaC1 is upregulated, while ChaC2 expression remains unaffected (47). Thus, ChaC2 serves as a constitutively expressed protein for basal hydrolysis of GSH, acting as a steward for slow and continuous GSH turnover (47).

3. GSH-degrading enzymes in tumorigenesis and progression

Tumor cells enhance GGT expression across the entire cell membrane, facilitating the acquisition of additional cysteine and cystine from GSH in blood and interstitial fluid to replenish intracellular GSH levels (22). Consequently, aberrant GGT expression is observed in various types of cancer, including ovarian (55), renal cell (56), lung (57), stomach (58) and pancreatic cancer (59). Elevated GGT expression is generally associated with poor prognosis, as patients with high levels of GGT in tumors exhibit shorter overall and progression-free survival (60). However, some breast tumor tissues exhibit GGT loss (61).

Studies (54,62,63) of the ChaC family have yielded conflicting findings regarding ChaC expression and its role in tumor tissues, emphasizing the complexity of GSH regulation and function. ChaC1, as a tumor-influencing factor, enhances ER stress, contributing to necroptosis and ferroptosis of multiple cancers, including metastatic melanoma (64), breast (54), prostate (65) and primary liver cancer (66), Burkitt's Lymphoma (67), head and neck squamous cell carcinoma (68), glioblastoma multiforme (GBM) (69), oral squamous cell carcinoma, T lymphoblastic leukemia Molt4 cells and colitis-associated carcinogenesis (15). Decreased ChaC1 expression is an indicator of poor prognosis in kidney renal clear cell carcinoma (KIRC) (70) and certain types of gastric cancer (71,72). Conversely, reports suggest that ChaC1 overexpression may be associated with tumor cell dedifferentiation, proliferation, invasion and migration, leading to lower patient survival rates (73,74). ChaC1 serves as a reliable indicator for poor prognosis of certain types of gastric cancer (63) and melanoma (75,76), as well as an independent indicator for elevated risk for female germ line tumors (including breast and ovarian cancer) (77). Therefore, different effects of ChaC1 may be linked to the specific functions of GSH in different types of tumor tissues.

ChaC2 may be implicated in numerous vital physiological functions, including DNA replication and repair, cell cycle regulation, RNA and damaged DNA binding, oocyte meiosis and maturation (50). Its important physiological role was initially discerned in undifferentiated human embryonic stem cells (hESCs), where ChaC2 is prominently expressed and maintains cell self-renewal and pluripotency by modulating GSH homeostasis. Conversely, downregulation of ChaC2 decelerates the cell cycle progression of hESCs and triggers cell death (78), underscoring its pivotal role in regulating human growth and development. In pathological functions, ChaC2 exhibits a multifaceted role in tumor tissue. Generally acting as a tumor suppressor, ChaC2 decreases GSH levels in tumor cells, instigates mitochondrial apoptosis and autophagy via UPR and hinders tumor cell proliferation and migration in vitro and in vivo (79). Therefore, ChaC2 expression is commonly downregulated in tumor tissues, such as gastric and colorectal cancer (79). ChaC2 may exert a tissue-specific function, promoting the survival of tumor cells in specific contexts (50,81,82). The expression of ChaC2 increases with progression of lymph node metastasis and the stage of breast cancer, aligning with findings of GGT loss (61) and increased ChaC1 expression (77) in breast cancer cells. The increased expression of ChaC2 is associated with the high expression of p53 (50), and ChaC2 is the upstream regulator of the main antioxidant regulator Nrf2 (78). In addition mutated p53 regulates NRF2-dependent antioxidant responses that are critical for supporting cancer cell survival (83). Therefore, increased ChaC2 expression may be related to regulating the binding of p53 mutants to Nrf2. When transcription and activity of GSH synthase is increased, the GSH level of the tissue is increased (80). The GSH overexpression then foster the survival and proliferation of tumor cells. Additionally, increased ChaC2 expression targets the Cadherin 1 gene (encoding E-cadherin) mutation, resulting in E-cadherin loss, increased epithelial-mesenchymal transformation and lymph node metastasis, ultimately contributing to the low differentiation of breast cancer (50). Consequently, overall ChaC2 expression is associated with lymph node metastasis and stage progression of breast cancer. ChaC2 promotes lung adenocarcinoma growth by elevating ROS levels and activating MAPK signaling pathways (81).

GGT serves as an extracellular degradation enzyme of GSH, catalyzing enzymatic degradation products to enter cells and providing an additional cysteine source for intracellular GSH synthesis. Intracellular degrading enzymes ChaC1 and ChaC2 play a key role in downregulating intracellular GSH levels, exerting a counteractive effect to regulate intracellular GSH homeostasis. The functions of GGT and ChaC1/ChaC2 include regulation of oxidative and ER stress (38,84), modulation of programmed cell death (85), promotion of inflammation and cell drug resistance (24,86) (Fig. 6).

GSH degradation products such as glutamic acid, glycine and cysteine also play key roles in the metabolic network of the tumor environment. For example, glutamate regulates proliferation, migration and survival of neuroprogenitor cells and immature neurons. While the ability to proliferate and migrate uncontrollably is characteristic of tumor cells, glutamate has been shown to serve as a growth factor and signaling medium in certain types of tumor tissues in both an autocrine and paracrine manner (17). Glycine is involved in cell transformation and tumorigenesis via cleavage into one-carbon metabolism (18). Therefore, exploring the function of GSH-degrading enzymes in tumors may clarify the role of the complex metabolic network of tumors.

Regulating oxidative and ER stress

GSH-degrading enzymes key central to coordinating cellular metabolism by regulating amino acid availability under physiological conditions (11). The modulation of the cellular stress environment relies on regulation of GSH metabolism (4). Tumor cells, in their quest for survival and proliferation, generate abnormally high levels of oxidative stress, partly due to increased cellular redox buffer GSH (87). Therefore, the role of GSH-degrading enzymes is key to regulate the cell stress environment.

The GSH degradation pathway initiated by extracellular GGT effectively controls the intracellular oxidative stress environment (38). GGT deficiency leads to oxidative stress and cellular vulnerability to oxidative damage. Animal studies (88-90) have indicated that GGT knockout mice exhibit 20% of the plasma cysteine concentration in wild-type mice. GGT knockout mice experience increased accumulation of DNA oxidative damage, decreased intracellular GSH levels, elevated oxidative stress and death by 10 weeks of age due to cysteine deficiency (88). Patients with partial GGT homozygous deletions report glutathionuria and neurodevelopmental disorder (91). Hence, maintaining regular GGT expression is essential for cellular GSH homeostasis and protecting cells against oxidative stress.

Cells overexpressing GGT gain an advantage in environments with physiological and limited cysteine concentrations by efficiently utilizing extracellular GSH as a source of cysteine (92). Tumor cells with elevated intracellular GSH levels often induce overexpression of GGT. It has been reported (22) that both GSH depletion and GGT inhibition significantly enhance cytotoxicity under oxidative stress in tumor cells. Tumor cells with high GGT expression demonstrate notable oxidative stress tolerance without DNA damage, while clones with low GGT expression exhibit increased sensitivity to oxidative stress and apoptosis (93). As a marker of oxidative stress, GGT expression in advanced tumor cells surpasses that in early tumor cells. The increased oxidative stress and impaired immune responses may be key for promoting cancer progression to advanced stages and may be induced by inflammatory mediators within the tumor (94). Therefore, upregulation of GGT in tumor cells provides a potential mechanism to resist oxidative stress and foster tumor progression.

Simultaneously, changes in the tumor microenvironment generate persistent ER stress signals in various types of tumor (95) such as colorectal (96), pancreatic cancer (97) and so on. This state has a dual effect on tumor cells, on one hand controlling several tumor-promoting features, on the other hand dynamically reprogramming immune cells and inducing tumor cells autophagy, apoptosis and ferroptosis (95). ChaC1, a component of the UPR pathway, is a target of ferroptosis induced by ER stress signals (12). Thus, ChaC1 is as a key regulator of tumor development, metastasis and responses to chemotherapy, targeted therapy and immunotherapy.

Previous research (78) has also revealed a broader range of functions for ChaC2 than previously understood. ChaC2 inhibits ChaC1-mediated GSH degradation, indicating competition with ChaC1 to maintain GSH homeostasis (78). ChaC2 directly regulates GSH production via a ChaC1-independent pathway (78). ChaC2 enhances GSH production by upregulating Nrf2, a key regulator of antioxidation, and its downstream glutamate-cysteine ligase (78). These diverse actions underscore the importance of ChaC2 in maintaining cellular redox homeostasis and antioxidation mechanisms.

Extensive crosstalk exists between oxidative and ER stress as oxidative stress can disrupt redox homeostasis in the ER, triggering ER stress (95). GGT and ChaC1 may be key factors in this mechanism. A study (98) demonstrated that GGT1 and GGT7 stimulate induction of ER stress-related protein, CHOP-10 and immunoglobulin heavy chain binding protein BiP, indicating specific roles for these GGT protein subtypes in ER stress response. This suggests possible crosstalk between GGTDelta1, GGTDelta7 and ChaC1 via the ER stress/CHOP pathway. A recent study (99) proposed that the ATF4/CHOP/ChaC1 signaling pathway might be vital for apoptosis induced by crosstalk between oxidative and ER stress. Under extreme heat stress, cells produce a large amount of ROS, leading to oxidative stress and protein misfolding in the ER, resulting in ER stress and triggering ChaC1-associated UPR. Moreover, induction of ChaC1 serves an essential regulatory role in ER stress-mediated apoptosis of cancer cells induced by the anticancer monosaccharide xylitol, leading to secondary induction of oxidative stress in treated cells and apoptosis (100). This evidence collectively demonstrates the key role of GGT and ChaC1 in mediating crosstalk between oxidative and ER stress.

Modulating programmed cell death

Programmed cell death, encompassing apoptosis, ferroptosis, necrotic apoptosis and autophagy, is instigated by a series of intracellular processes (101). GGT- and ChaC1/ChaC2-mediated intracellular GSH depletion can concurrently or sequentially initiate multiple forms of programmed cell death. Studies (101,102) have indicated that the GSH/GSSG redox status serves as a vital indicator of tumor programmed cell death, consistently associating programmed cell death with a decrease in the GSH/GSSG ratio. Therefore, targeting GGT and ChaC1/ChaC2 to modulate programmed cell death holds implications for tumor therapy.

Regulating apoptosis

Apoptosis, the quintessential programmed cell death process, primarily relies on the caspase family for initiation and is typically characterized by membrane contraction, chromatin concentration and apoptotic body formation (103). In tumor cells, the apoptosis pathway is often impeded by various mechanisms, many of which contribute to intrinsic resistance to chemotherapy, the most prevalent anticancer therapy (104). Reducing GSH impairs cellular antioxidant regulation, increasing ROS production, thereby accelerating mitochondrial damage and apoptosis induction (101). Consequently, inhibiting GGT1 in tumor cells facilitates induction of apoptotic phenotypes (105). Simultaneously, research (106) on another member of the GGT family has shown that low GGT7 expression may elevate cell ROS levels, inhibiting apoptosis and fostering tumor proliferation. This suggests variations in regulation of ROS levels within the GGT family. ChaC1 overexpression can augment apoptosis by activating caspase-3/9, degradation of poly (ADP ribose) polymerase, induction of autophagy, ROS generation, increased intracellular calcium and loss of mitochondrial membrane potential (69).

Regulating ferroptosis

Ferroptosis, a distinct iron-dependent form of cell death, arises from lethal accumulation of lipid peroxides (107). In tumor cells, evasion of ferroptosis mediated by oncogenes or carcinogenic signaling contributes to tumor onset, progression, metastasis and resistance to treatment. Simultaneously, some tumor cells, owing to specific mutations, elevated ROS levels and other unique biological features, exhibit ferroptosis susceptibility, with their survival hinging on the ferroptosis defense system (108). For example, ferroptosis resistance is conferred by frequently occurring PI3K activating mutations or loss of phosphatase and tensin homolog deleted on chromosome 10 function in human cancer such as lung adenocarcinoma (109) and breast cancer (110) and so on. Conversely, inhibition of the PI3K/AKT/mTOR signaling axis sensitizes cancer cells to ferroptosis induction (111). Consequently, targeting ferroptosis regulation holds implications for cancer immunotherapy and tumor suppression.

Regarding ferroptosis regulation, GGT-activated extracellular GSH catabolism produces iron-derived ROS, inducing lipid peroxidation via NF-κB pathway activation (112). GGT-mediated GSH catabolism via lipid peroxidation enhances NF-κB DNA binding capacity in tumor cells (113). GGT increased intracellular GSH levels (114), restores the reduced GSH/GSSG ratio and reactivates GSH peroxidase 4, a core ferroptosis regulator. Therefore, elevated GGT expression increases tumor cell resistance to ferroptosis, safeguarding cells from ROS and lipid peroxidation, thus driving tumor cell proliferation, metastasis and chemotherapy drug resistance (85,107). In addition, GGT activates the mTORC1 pathway and inhibits integrated stress response (ISR) by modulating cystine-GSH crosstalk. This inhibition of ferroptosis promotes cancer development and other cysteine-deficient diseases (115). Inhibiting GGT impairs GSH ability to restore mTORC1 signaling and ISR, inducing ferroptosis. This implies that the role of GGT in inducing capacity of GSH to release cysteine, rather than GSH itself, modulates the mTORC1 pathway, ISR and ferroptosis. By contrast, ChaC1 induces ferroptosis in tumor cells by activating the GCN2/EIF2α/ATF4 pathway to intensify cystine depletion (116). ChaC1 overexpression depletes GSH, initiating and executing ferroptosis. The deletion of ATF4, an upstream factor of ChaC1, in embryonic fibroblasts, results in a ferric oxide-dependent death phenotype, emphasizing the role of ATF4 as a downstream molecule of the eIF2α/ATF4 pathway (117). Hence, ferroptosis control is associated with ChaC1 expression.

Regulating necroptosis

Necroptosis, a regulated form of cell death primarily dependent on receptor-interacting protein kinase 3 and mixed lineage kinase domain-like, is characterized by widespread cytoplasm and organelle swelling, plasma membrane rupture and release of cell components into the microenvironment (118). This pro-inflammatory form of cell death holds implication for combating pathogen infection, inflammatory progression (119), and therefore may also contribute to early prevention of inflammatory cancer transformation.

Understanding of the impact of GGT and ChaC1/ChaC2 on necroptosis is limited. GGT, one of the virulence factors of Helicobacter pylori, has been demonstrated to induce necroptosis in gastric epithelial cells (119). In the early stage of infection, necroptosis may serve a protective role in the mucosa by triggering an immune response (119). However, as the disease progresses, uncontrolled necroptosis exacerbates mucosal inflammation and contributes to the transformation of inflammation into cancer (119). Additionally, ChaC1, by stimulating the GCN2/eIF2α/ATF4 pathway, enhances necroptosis induced by cystine deprivation (54).

Potential regulation of autophagy

Autophagy, a highly conserved cellular degradation process, involves breaking down cytoplasmic components and damaged organelles via lysosomes, recycling resulting macromolecules to shield cells from diverse stressful conditions. Traditionally viewed as a cytoprotective mechanism, autophagy, when excessive, can also instigate cell death and contribute to tumor suppression (120). GSH is implicated in inducing autophagy, where low GSH levels serve as a signal activating autophagy as an adaptive stress response (101). Inhibition of GGT (105) and elevated expression of ChaC1/ChaC2 (69,79) are associated with autophagy phenotype. Nevertheless, evidence (79,121) elucidating the precise mechanisms and interactions between GGT and ChaC1/ChaC2 and initiation and promotion of autophagy remains limited.

Promoting inflammation

Chronic non-specific inflammation is pivotal in tumorigenesis (122) and is a primary environmental factor contributing to the onset and metastasis of specific types of cancer such as non-small cell lung cancer and colorectal cancer (122,123). Various blood tests, either individually or in combination, reflecting local or systemic inflammation, are valuable prognostic indicators for multiple tumor types (124).

GGT serves as a well-established inflammatory marker associated with inflammatory environments and malignancy (125,126). Combining serum albumin and GGT levels serves as an inflammatory indicator for assessing the prognosis of hepatocellular carcinoma (HCC). Patients with elevated GGT and decreased albumin expression exhibit poorer prognosis, revealing significant differences in tumor characteristics, including larger maximum tumor diameters, more tumor nodules and potential for macroscopic vascular invasion and higher serum tumor marker levels (124).

Furthermore, although there is limited research on the effect of human GGT on the colonization of H. pylori, it is known that H. pylori GGT is a bacterial virulence factor that contributes to the colonization of H. pylori in human stomach parietal cells, hence inducing inflammation and gastric parietal cell carcinogenesis (127). In H. pylori infection, stimulation of H. pylori GGT accelerates the decrease of GSH levels in gastric epithelial cells, thereby exacerbating ROS production, leading to DNA damage and playing a key role in the emergence of chronic gastritis and gastric cancer (127). H. pylori GGT is also key for the tolerogenic effect of dendritic cells in H. pylori infection, ensuring bacterial persistence and cross-protection from chronic inflammation and autoimmune diseases by promoting H. pylori to reprogram dendritic cells into tolerogenic phenotypes (128). More research is needed to determine whether human GGT functions similarly in H. pylori-infected gastric parietal cells.

Human ChaC1 has potential ability to promote inflammation (129). ChaC1 is highly expressed in gastric cancer associated with H. pylori infection (129). Infection with H. pylori triggers ChaC1 overexpression in gastric epithelial cells, leading to GSH degradation and ROS accumulation, suppressing nucleotide alterations in TP53 that induce tumor suppressor gene expression (130). Overexpression of ChaC1 in H. pylori-infected parietal cells may also lead to H. pylori-induced somatic mutation, thereby promoting the development of gastric cancer (131). In summary, high expression of human ChaC1 is involved in inducing development of gastric cancer.

ChaC1 expression varies in normal and cystic fibrosis bronchial epithelial cells, with low ChaC1 expression hypothesized to play a significant role in regulating the chronic inflammatory response induced by Pseudomonas aeruginosa (Pa) (132). When exposed to Pa and its virulence components, normal bronchial epithelial cells preferentially produce ChaC1. Conversely, low ChaC1 expression is associated with increased secretion of inflammatory markers interleukin-8, interleukin-6 and prostaglandin E2 in the presence of lipopolysaccharide and flagellin stimulation (132). Low ChaC1 expression also promotes increased phosphorylation of NF-κB p65, possibly contributing to the exacerbation of characteristic inflammation in the lungs of patients with cystic fibrosis following Pa infection (132). Cystic fibrosis itself is a risk factor for various cancers, including lung cancer (133).

Drug resistance

High GGT and low ChaC1 expression in cancer cells are pivotal factors in developing drug-resistant phenotypes in tumors (86). GGT expression provides cells with an additional supply of cysteine, while low ChaC1 expression hinders degradation of intracellular GSH. Both factors contribute to GSH consumption in tumor cells during anticancer chemotherapy, leading to drug resistance. Maintaining high intracellular GSH expression preserves redox status, allowing cells to respond to proliferative and differentiation signals in tissue after toxin injury (22) and rapidly supplement GSH during pro-oxidant anticancer therapy.

Chemotherapy-resistant tumors often exhibit high GGT expression, exemplified by cisplatin resistance (134). Platinum (II) class antitumor drugs such as cisplatin and oxaliplatin, widely used in cancer chemotherapy, target DNA damage and overall cytotoxicity in tumor cells, resulting in cell death (135). GGT-related detoxification of Pt(II) medication is a key mechanism of drug resistance (136). Cisplatin can strongly bind to mercaptan metabolites produced by GGT-mediated GSH cleavage, reducing Pt ion entry into cells and inactivating Pt drugs outside the cell (136). GGT-transfected cells show decreased DNA platinization, resulting in decreased sensitivity to cisplatin and lower susceptibility to DNA damage (114). GGT-transfected HeLa cells exposed to cisplatin exhibit a 10-fold increase in cisplatin resistance (134). Systematic inhibition of GGT expression is conducive to suppressing nephrotoxic side effects of cisplatin without diminishing its intracellular toxicity to tumor cells (137). These findings underscore the key role of high GGT expression in promoting drug-resistant phenotypes in tumor cells.

By contrast with GGT, high ChaC1 expression, combined with drugs such as bortezomib and docetaxel, inhibits tumor cell viability by blocking cell cycle progression from the G1 phase to the mitotic S phase. Additionally, high ChaC1 expression increases tumor cell sensitivity to anti-tumor drugs by inducing ER stress and ferroptosis (65,69,138). GSH depletion triggered by the Nrf2/ATF3/4/ChaC1 pathway appears to be the primary factor inducing death in drug-resistant tumor cells (138), positioning ChaC1 as a key target for certain potential anti-tumor drugs such as busatol (139) and glaucocalyxin A (140). Low expression of ChaC1 appears in the drug-resistant phenotype of tumor cells (141), which may be related to enhanced intracellular GSH levels.

4. Role of GSH-degrading enzymes in medicine

Promising tumor biomarkers

Early tumor screening and diagnosis are key for effective treatment and favorable prognosis. Reliable and specific tumor biomarkers are key for accurate screening and diagnosis. The aberrant expression of GSH-degrading enzymes is associated with the prognosis of certain cancers such as gastric adenocarcinoma (72), breast cancer (74) and so on, making them promising tumor biomarkers.

GGT as a biomarker

GGT has been extensively studied and is widely used as a biomarker in clinical practice (142-144). Serum GGT activity is a rapid, reliable and cost-effective method to assess liver function (145). Therefore, GGT has the potential to serve as a tumor biomarker. Elevated GGT expression can indicate the early-stage risk of tumor development, as suggested by an epidemiological study linking high GGT expression with increased risk of prostate cancer development (145). Moreover, high GGT expression indicates poor prognosis in various types of tumors, including renal cell carcinoma and prostate and urothelial cancer (145). The Cancer antigen 19-9/GGT ratio serves as an independent prognostic predictor following radical resection in ampulla carcinoma (146). GGT6 and GGT2 are novel synergistic prognostic biomarkers for low-grade glioma and GBM, potentially aiding early detection (30).

GGT probes have been developed to detect GGT activity accurately for tumor imaging. However, the imaging process often requires organic solvents, posing risks of damage to the enzyme and body. A water-soluble fluorescent probe, TCF-GGT (147), has shown promise by producing red fluorescence during GGT catalytic hydrolysis without interference from the background. This water-soluble compound holds clinical value due to its practical imaging, quick metabolic cycle and excellent water solubility. Due to shallow tissue penetration of many GGT-targeted fluorescent probes, their clinical application is limited. A novel positron emission tomography imaging probe, ([18F]GCPA)2, has been designed to sensitively and precisely monitor GGT levels in living subjects because of the high sensitivity and intense tissue penetration of positron emission tomography (148). In addition, Cy-GSH, a zero-crosstalk ratio near-infrared GGT fluorescent probe, has also been designed to visualize deep cancer in vivo. The probe accurately visualizes tumors and metastases in mice, suggesting that it could be a convenient tool for fluorescence-guided cancer surgery (149). More clinically applicable GGT-targeted probes for visualizing deep cancer need further studies, which may contribute to the early diagnosis of clinical tumors.

ChaC1/ChaC2 as biomarkers

ChaC1, a GSH-degrading enzyme, is biomarker for certain types of tumors. In vitro, ChaC1 induces cell death in KIRC cell lines, signifying its potential as an effective marker for poor prognosis in KIRC (70). High ChaC1 expression also serves as a biomarker for adverse outcomes in gastric adenocarcinoma (72), corpus endometrial (73) and breast cancer (74) and uveal melanoma (75). Notably, a positive correlation exists between ChaC1 expression and immune infiltrating cells in corpus endometrial carcinoma (73). In uveal melanoma (75,150), ChaC1 is associated with poor overall, progression-free and disease-specific survival and progression-free interval, making it a promising indicator of unfavorable tumor prognosis. In aggressive breast tumor subtypes such as triple-negative breast cancer, ChaC1 exhibits significant upregulation (151). In addition, malignant breast cancer tissue with active lymph node metastases and high proliferation rates demonstrates elevated levels of ChaC1, supporting its potential for defining tumor progression and metastasis (Table II).

Table II

Tumors with altered expression of GGT and ChaC1/ChaC2.

Table II

Tumors with altered expression of GGT and ChaC1/ChaC2.

First author/s (year)TumorExpression(Refs.)
Chen, et al, 2023; Hayashima, et al, 2022; Chen, et al, 2017; Xu, et al, 2022GliomaIncreased ChaC1; increased GGT(69,85,154,180)
Wen, et al, 2017Nasopharyngeal carcinomaIncreased GGT(181)
Wang, et al, 2022; Mujawar, et al, 2020Oral squamous cell carcinomaDecreased ChaC1; increased GGT(140,182)
Mizushima, et al, 2016Head and neck squamous cell carcinomaIncreased GGT(183)
Lee, et al, 2021Laryngeal cancerIncreased GGT(184)
Gu, et al, 2022Thyroid cancerIncreased GGT(185)
Foddis, et al, 2022Malignant pleural mesotheliomaIncreased GGT(186)
Peng, et al, 2023; Lee, et al, 2021Lung cancerIncreased GGT and ChaC2(81,184)
Huang et al, 2017; Choi, et al, 2017Esophagus cancerIncreased GGT(187,188)
Tian, et al, 2021; Tian, et al, 2022Hepatocellular carcinomaIncreased GGT and ChaC2(32,82)
Chen, et al, 2021Intrahepatic cholangiocarcinomaIncreased GGT(189)
Catalano, et al, 2023; Liao, et al, 2023Pancreatic cancerIncrease GGT(190,191)
Wu, et al, 2021; Zhang, et al, 2022; Tseng, et al, 2021; Liu, et al, 2017 Ogawa, et al, 2019; Hong, et al, 2021; Yang, et al, 2019Gastric cancerDecreased ChaC1 and ChaC2; increased GGT and ChaC1(62,63,71,79, 131,192,193)
Xiao et al, 2022Stomach adenocarcinomaDecreased ChaC1(72)
Liu, et al, 2017; Hong, et al, 2021; Hong, et al, 2020Colorectal cancerDecreased ChaC2; increased GGT(79,192,194)
Li et al, 2021; Yang, 2022; Horie, et al, 2020Renal clear cell carcinomaDecreased ChaC1; increased GGT(70,195,196)
Nguyen, et al, 2019; Chand, et al, 2022; Mehta, et al, 2022; Goebel, et al, 2012; Mehta et al, 2022; Pankevičiūtė-Bukauskienė, et al, 2023; Seol, et al, 2021Breast cancerIncreased GGT and ChaC1 and 2(20,50,74, 77, 151,197,198)
Goebel et al, 2012; Shi1 et al, 2018Ovarian cancerIncreased GGT and ChaC1(77,199)
Liu, et al, 2022Uterine corpus endometrial carcinomaIncreased ChaC1(73)
Schwameis, et al, 2016Uterine leiomyosarcomaIncreased GGT(200)
Polterauer, et al, 2011Cervical cancerIncreased GGT(201)
He, et al, 2021; Kawakami, et al, 2017Prostate cancerDecreased ChaC1; increased GGT(65,202)
Su et al, 2021Bladder cancerIncreased GGT(203)
Takemura, et al, 2019Urothelial carcinomaIncreased GGT(204)
Liu, et al, 2022Cutaneous melanomaIncreased ChaC1(76)
Liu, et al, 2019; Jin, et al, 2021;Uveal melanomaIncreased ChaC1(75,150)
Song et al, 2021Acute myeloid leukemiaDecreased ChaC1(205)

[i] GGT, γ-glutamyl transpeptidase; ChaC1, glutathione-specific γ-glutamylcyclotransferase 1.

The abnormal expression of ChaC2 as a tumor biomarker has garnered recent attention (50,81). As an independent marker of poor prognosis for certain types of tumors such as breast cancer (50) and hepatocellular carcinoma (82), ChaC2 can monitor early tumor occurrence and treatment effects. Low expression of ChaC2 independently indicates poor prognosis in gastrointestinal tumors. ChaC2 induces mitochondrial apoptosis and autophagy via UPR, serving a pivotal role as a tumor suppressor gene in the onset, proliferation and metastasis of gastric and colorectal cancer (79). Immunohistochemistry and western blot analysis reveal ChaC2 downregulation in most tumor tissue, with ChaC2 expression positively correlating with 3-year survival rate (79). However, recent findings indicate that high ChaC2 expression is inversely correlated with overall survival in patients with breast cancer (50). Elevated ChaC2 expression is also associated with poor prognosis in HCC (82). This underscores the tissue-specific effect of ChaC2 on tumors, necessitating further study (Table II).

Further, the previous study (152) integrates traditional predictors and ChaC1, a novel biomarker, to develop a prognostic score for patients with tumors. The score can be used as a reference for clinical chemotherapy decision-making (152). In evaluating patients with primary breast cancer, including ChaC1 mRNA expression levels in the scoring model led to changes in chemotherapy decisions in 16% of patients (152). In addition, ChaC1 has been included in prognostic models of renal cell carcinoma (153) and glioblastoma (154). Including ChaC1 in tumor prognosis predictions may guide personalized treatment options. ChaC1/ChaC2 may be promising targets for precision medicine.

While studies (50,73,74,151,82) have reported ChaC1/ChaC2 as tumor biomarkers, the development of ChaC1/ChaC2 tracer fluorescent probes remains unexplored. Fluorescent probes are the foundation for tumor-specific imaging in clinical applications. Therefore, developing optical probes to track ChaC1/ChaC2 in vivo or in vitro is key for fully realizing the clinical potential of ChaC1/ChaC2 as tumor markers.

Therapeutic targets for tumors

Drug selection and emerging drug development. Exploration of GSH-degrading enzyme modulators presents a promising avenue for impeding cancer progression, overcoming tumor resistance to pro-oxidative therapy and preserving tumor sensitivity to chemotherapy (Table III).

Table III

List of the classical and promising therapeutics affecting GGT or ChaC1/ChaC2.

Table III

List of the classical and promising therapeutics affecting GGT or ChaC1/ChaC2.

First author (year)Functional categoryTherapySource Mechanism/pathways(Refs.)
Lyons, et al, 1990GGT inhibitorGlutamine analogues (acivicin, 6-diazo-5-oxo-norleucine and azaserine)Fermentation products of StreptomycesGlutamine analogs are competitive inhibitors that directly modify active site nucleophiles. Due to neurotoxicity, they are no longer used in the clinic(161)
Han, et al, 2007; Watanabe, et al, 2017GGsTopChemical synthesisA phosphonate-basedpotent, non-toxic, highly selective and irreversible GGT inhibitor. Human GGT recognizes the negative charge of GGsTop instead of the C-terminal carboxy group of glutathione by a positively charged critical residue located in the Cys-Gly binding site(206,207)
King, et al, 2009OU749Chemical synthesis Species-specifically non-competitively inhibiting human GGT. OU749 binds to the covalent E-γ-glutamyl complex, the F form of the enzyme(208)
Azouz, et al, 2020AmlodipineChemical synthesis; fully substituted dialkyl 1,4-dihydropyridine-3,5-dicarboxylate derivativeCurrently unclear(164)
Brancaccio, et al, 2019OvothiolsMarine metazoansInhibit membrane-bound GGT of human cells non-competitively and reduce proliferation in GGT-positive cell lines with simultaneous occurrence of a non-protective/cytotoxic form of autophagy, indicating inhibition of GGT activity is likely involved in the modulation of autophagic mechanisms(105)
Joo, et al, 2012ChaC1 activatorNisinBacterium Lactococcus lactisCurrently unclear(68)
Stevens, et al, 2019AtovaquoneChemical synthesis of hydroxynaphthoquinone or analog of ubiquinoneIncreased EIF2α/ATF4/ChaC1 pathway activity(158)
Tomonobu, et al, 2020XylitolFruits and vegetablesInduction of CHAC1 by xylitol triggers endoplasmic reticulum stress(100)
Wang, et al, 2019ArtesunateArtemisia apiaceaIncreased EIF2α/ATF4/ChaC1 pathway activity(67)
Wang, et al, 2021 DihydroartemisininArtemisia apiaceaIncreased EIF2α/ATF4/ChaC1 pathway activity(66)
Wang, et al, 2022Glaucocalyxin ARabdosia japonicaIncreased EIF2α/ATF4/ChaC1 pathway activity(140)
Guan, et al, 2020Tanshinone IIASalvia miltiorrhiza BungeCurrently unclear(162)
Chen, et al, 2021Omega-3 fatty acids docosahexaenoic acid or eicosapentaenoic acidMarine metazoansIncreased EIF2α/ATF4/ChaC1 pathway activity(138)
Tseng, et al, 2021Negatively regulating CHAC1 expression factorMetforminGalega officinalisRegulating the Loc100506691-miR-26a-5p-miR-330-5p-ChaC1 axis signaling induces cell cycle arrest in G2/M phase, inhibiting cancer cell proliferation(71)
Zhai et al, 2022ChaC2 activatorNaringinCitrus fruitUpregulating CHAC2 via activation of the Nrf2 signaling pathway(209)

[i] GGT, γ-glutamyl transpeptidase; ChaC1, glutathione-specific γ-glutamylcyclotransferase 1; EIF2α, eukaryotic initiation factor-2α; ATF4, activating transcription factor 4; miR, microRNA; GGsTop, 2-amino-4-[(3-(carboxymethyl)phenoxy)(methoyl)phosphoryl]butanoic acid.

Diminishing GGT expression is beneficial for tumor treatment. Classical GGT inhibitors, including glutamate analogs such as acivicin, 6-diazo-5-oxo-L-norleucine, and azaserine (155), have shown clinical toxicity against embryonic cells (156). γ phosphonoglutamate analogs such as GGsTop and derivatives of the lead compound N-[5-(4-methoxybenzyl)-1,3,4-thiadiazol-2-yl]benzenesulfonamide, OU749, represent another class of GGT inhibitors (22). GGsTop targets GGT, can reduce the immunosuppressive function of anti-tumor drugs when used in combination with anti-tumor drugs (157). OU749, a species-specific uncompetitive GGT inhibitor, exhibits low toxicity and a broad therapeutic window for humans.

The development of ChaC1 modulators introduces innovative approaches to cancer treatment. Various potential ChaC1 modulators have been reported, targeting tumor cell cycle arrest and apoptosis (68,158). Metformin, by regulating the Loc100506691-miR-26a-5p-miR-330-5p-CHAC1 axis, induces cell cycle arrest in the G2/M phase, inhibiting cancer cell proliferation (71). Nisin, an apoptotic bacteriocin, induces ChaC1 activation, calcium influx and cell cycle arrest in the G2 phase, leading to increased apoptosis and decreased cell proliferation in vitro and in vivo (68). Atovaquone, an anti-parasitic agent, induces eIF2a phosphorylation at serine 51, amplifying the eIF2α/ATF/CHOP/ChaC1 signaling pathway under ER stress (158).

Natural extracts and their bioactive compounds have recently gained recognition as potential lead molecules in drug discovery for cancer treatment (159,160). They offer an alternative to chemical synthetic drugs, potentially minimizing toxic side effects and holding promise for enhancing clinical anti-tumor therapy. GGT and ChaC1 modulators derived from natural extracts have been explored. Ovothiol, a marine-derived 5(N)-methyl-thiohistidine, is a more effective non-competitive-like GGT inhibitor than traditional counterparts such as acivicin, 6-diazo-5-oxo-norleucine and azaserine (105,161). Ovothiol (105) induces apoptosis and autophagy in GGT-overexpressing cells. The anticancer monosaccharide xylitol (100) and natural Chinese herbal extracts, including dihydroartemisinin (66), artesunate (67), glaucocalyxin A (140), and tanshinone IIA (162), upregulate Prostaglandin-Endoperoxide Synthase 2, p53 and ChaC1 expression. This amplifies the ATF4/CHOP/ChaC1 cascade under ER stress, resulting in decreased intracellular GSH and cysteine, increased intracellular ROS, redox homeostasis disruption, secondary oxidative stress in cancer cells and selective ferroptosis of cancer cells (66,100).

In clinical treatment of tumors, a key goal is to decrease toxicity and resistance of anti-tumor drugs by regulating low expression of GGT (22) and high expression of ChaC1 (65). Blocking nephrotoxicity induced by the anti-tumor drug cisplatin is achieved by inhibiting GGT (137). The unique renal sulfhydryl acid metabolic pathway, involving degradation of GSH by GGT, contributes to cisplatin nephrotoxicity (163). Supramolecular Pt prodrug nano-assemblies inhibiting c-glutamyl transferase prove beneficial for overcoming tumor resistance (136). GGT inhibitor amlodipine, through its anti-inflammatory effect, suppresses p38 MAPK-triggered pro-inflammatory signaling, decreasing expression of TNF-α and other downstream targets while upregulating expression of the transcription factor Nrf2 and the antioxidant protein heme oxygenase-1 (164). Amlodipine diminishes the pro-apoptotic effector/anti-apoptotic protein expression ratio induced by cisplatin, preventing inflammation, oxidative stress and apoptotic damage (165).

Combining ChaC1 overexpression with chemotherapy is advantageous in decreasing cell drug resistance. Temozolomide, for example, induces binding of ChaC1 to the Notch3 protein, inhibiting activation of Notch3. This weakens the Notch3-mediated downstream signaling pathway, inducing glioma cell death and indicating that ChaC1 can influence the cytotoxicity of tumor cells induced by temozolomide (69). The omega-3 fatty acids docosahexaenoic acid and eicosapentaenoic acid serve a role in reducing the resistance of tumor cells to bortezomib by activating the serine synthesis pathway, mitochondrial folate cycle, methionine cycle-associated GSH synthesis and ChaC1-mediated GSH degradation in tumor cells, ultimately promoting GSH degradation (138).

5. Conclusion

Recent years have seen notable advancements in understanding the role of the intracellular degrading enzyme ChaC family in regulating cellular GSH levels (165,166). Notably, coordinated actions of GGT and ChaC1/ChaC2 have been identified in modulating GSH levels both within and outside the cell (11). These changes in intracellular GSH levels affect key biological processes, including signal transduction, cell survival, proliferation (167) and various forms of cell death (168,169).

The present review underscores the pivotal role of GSH-degrading enzymes, specifically GGT and ChaC1, in cancer development. GGT, via degradation of extracellular GSH, provides cysteine for intracellular GSH synthesis and regeneration, elevating intracellular GSH levels (170). Consequently, GGT can modulate the oxidative stress state of cancer cells, inhibiting programmed cell death (170). GSH regeneration also counteracts the depletion induced by cancer chemotherapeutic agents, potentially leading to development of cancer drug resistance (171). By contrast, intracellular degradation enzyme ChaC1, differentially expressed in tumor cells, serves as a pro-apoptotic molecule by downregulating intracellular GSH levels under amino acid linkage and ER stress (99). ChaC1 induces various forms of cell death, enhancing ER stress and influencing the tumor microenvironment (70,151). Moreover, ChaC1 has a mitigating effect on cell resistance to cancer drugs (69). Together, GGT and ChaC1 regulate both intracellular and extracellular GSH degradation, emerging as promising prognostic markers and therapeutic targets for specific types of tumor such as liver cancer (172,173), breast cancer (74) and so on. Leveraging these enzymes for targeted anti-tumor therapy holds the advantage of precision targeting, minimal damage to healthy cells and reduced toxic side effects, thereby enhancing and reversing tumor drug resistance.

Despite advancements, unanswered questions persist regarding GSH-degrading enzymes. The determinants of functional and regulatory changes in GGT and ChaC1/ChaC2 across different malignancies or settings remain elusive. Although external stimuli induce ChaC1/ChaC2 gene and GGT expression (15,174,175), the specific regulatory mechanisms governing these inductions remain unclear. Additionally, crosstalk effect of different pathways in GGT and ChaC1/ChaC2 regulation requires further investigation. Furthermore, while the roles of GGT and ChaC1/ChaC2 in tumor tissue are known, these enzymes represent only the initial steps in GSH degradation. The interaction between GGT, ChaC1/ChaC2 and downstream enzymes in the complete degradation of GSH, Cys-Gly degrading enzymes and 5-oxo-proline (176-179) remains unclear. The development of tumor treatment strategies targeting GSH-degrading enzymes faces challenges, including different roles of these enzymes in different types of tumor and their impact on the immune response or tumor microenvironment.

In conclusion, GGT and the ChaC family of GSH degrading enzymes, both intracellularly and extracellularly, are key for maintaining GSH homeostasis and serve key roles in normal cellular processes and tumor-related stress conditions. A more comprehensive understanding of these mechanisms may clarify the potential of GSH-degrading enzymes as targets for tumor diagnosis and therapeutic interventions.

Availability of data and materials

Not applicable.

Authors' contributions

TZ and CY wrote and revised the manuscript. CY, DH and WS conceived and designed the review. XZ, SL, LQ, SZ and CZ performed the literature review. DH and WS revised the manuscript. Data authentication is not applicable. All authors have read and approved the final manuscript.

Ethics approval and consent to participate

Not applicable.

Patient consent for publication

Not applicable.

Competing interests

The authors declare that they have no competing interests.

Abbreviations:

ACM

a novel-248 ATF/CRE modifier

AP-1

activator protein 1

AP-2

activating protein 2

ATF

activating transcription factor

C/EBP-β

CCAAT/enhancer binding protein β

ChaC1

cation transport regulator homolog glutathione specific γ-glutamylcyclotransferase 1

CHOP

C/EBP homologous protein

eIF2α

eukaryotic initiation factor-2α

EpRE

electrophile response element

ER

endoplasmic reticulum

GBM

glioblastoma multiforme

GCN2

general control nonderepressible 2

GGT

γ-glutamyl transpeptidase

GP5

GGT promoter 5

HCC

hepatocellular carcinoma

hESC

human embryonic stem cell

HNE

4-hydroxynonenal

ISR

integrated stress response

IκB

inhibitor of κB

KIRC

kidney renal clear cell carcinoma

L2

alveolar type II

Pa

Pseudomonas aeruginosa

ROS

reactive oxygen species

Sp1

specific protein 1

UPR

unfolded protein response

Acknowledgements

Not applicable.

Funding

The present study was supported by the Shanghai Style TCM Inheritance and Innovation Team Building Project (grant no. 2021LPTD-004), Natural Science Foundation of Shanghai (grant nos. 19ZR1457500, 19ZR1460800 and 18ZR1440300) and Research Project of Shanghai Health Commission (grant no. 202140348).

References

1 

Lushchak VI: Glutathione homeostasis and functions: potential targets for medical interventions. J Amino Acids. 2012:7368372012. View Article : Google Scholar : PubMed/NCBI

2 

Lu SC: Regulation of glutathione synthesis. Mol Aspects Med. 30:42–59. 2009. View Article : Google Scholar :

3 

Kennedy L, Sandhu JK, Harper ME and Cuperlovic-Culf M: Role of glutathione in cancer: From mechanisms to therapies. Biomolecules. 10:14292020. View Article : Google Scholar : PubMed/NCBI

4 

Bansal A and Simon MC: Glutathione metabolism in cancer progression and treatment resistance. J Cell Biol. 217:2291–2298. 2018. View Article : Google Scholar : PubMed/NCBI

5 

Fernández-Checa J, Hirano T, Tsukamoto H and Kaplowitz N: Mitochondrial glutathione depletion in alcoholic liver disease. Alcohol. 10:469–475. 1993. View Article : Google Scholar : PubMed/NCBI

6 

Guarino MP, Afonso RA, Raimundo N, Raposo JF and Macedo MP: Hepatic glutathione and nitric oxide are critical for hepatic insulin-sensitizing substance action. Am J Physiol Gastrointest Liver Physiol. 284:G588–G594. 2003. View Article : Google Scholar

7 

Mandal PK, Roy RG and Samkaria A: Oxidative stress: Glutathione and its potential to protect methionine-35 of abeta peptide from oxidation. ACS Omega. 7:27052–27061. 2022. View Article : Google Scholar : PubMed/NCBI

8 

Charisis S, Ntanasi E, Stamelou M, Xiromerisiou G, Maraki M, Veskoukis AS, Yannakoulia M, Kosmidis MH, Anastasiou CA, Giagkou N, et al: Plasma glutathione and prodromal parkinson's disease probability. Mov Disord. 37:200–205. 2022. View Article : Google Scholar

9 

Niu B, Liao K, Zhou Y, Wen T, Quan G, Pan X and Wu C: Application of glutathione depletion in cancer therapy: Enhanced ROS-based therapy, ferroptosis, and chemotherapy. Biomaterials. 277:1211102021. View Article : Google Scholar : PubMed/NCBI

10 

Bachhawat AK and Yadav S: The glutathione cycle: Glutathione metabolism beyond the γ-glutamyl cycle. IUBMB Life. 70:585–592. 2018. View Article : Google Scholar : PubMed/NCBI

11 

Bachhawat AK and Kaur A: Glutathione degradation. Antioxid Redox Signal. 27:1200–1216. 2017. View Article : Google Scholar : PubMed/NCBI

12 

Mungrue I, Pagnon J, Kohannim O, Gargalovic P and Lusis A: CHAC1/MGC4504 is a novel proapoptotic component of the unfolded protein response, downstream of the ATF4-ATF3-CHOP cascade. J Immunol. 182:466–476. 2009. View Article : Google Scholar

13 

Kumar A, Tikoo S, Maity S, Sengupta S, Sengupta S, Kaur A and Bachhawat AK: Mammalian proapoptotic factor ChaC1 and its homologues function as γ-glutamyl cyclotransferases acting specifically on glutathione. EMBO Rep. 13:1095–1101. 2012. View Article : Google Scholar : PubMed/NCBI

14 

Zhang Y, Ma S, Zhang J, Lou L, Liu W, Gao C, Miao L, Sun F, Chen W, Cao X and Wei J: MicroRNA-142-3p promotes renal cell carcinoma progression by targeting RhoBTB3 to regulate HIF-1 signaling and GGT/GSH pathways. Sci Rep. 13:59352023. View Article : Google Scholar : PubMed/NCBI

15 

Zhang X, Li W, Ma Y, Zhao X, He L, Sun P and Wang H: High-fat diet aggravates colitis-associated carcinogenesis by evading ferroptosis in the ER stress-mediated pathway. Free Radic Biol Med. 177:156–166. 2021. View Article : Google Scholar : PubMed/NCBI

16 

Alborzinia H, Flórez AF, Kreth S, Brückner LM, Yildiz U, Gartlgruber M, Odoni DI, Poschet G, Garbowicz K, Shao C, et al: MYCN mediates cysteine addiction and sensitizes neuroblastoma to ferroptosis. Nat Cancer. 3:471–485. 2022. View Article : Google Scholar : PubMed/NCBI

17 

Stepulak A, Rola R, Polberg K and Ikonomidou C: Glutamate and its receptors in cancer. J Neural Transm (Vienna). 121:933–944. 2014. View Article : Google Scholar : PubMed/NCBI

18 

Locasale JW: Serine, glycine and one-carbon units: Cancer metabolism in full circle. Nat Rev Cancer. 13:572–583. 2013. View Article : Google Scholar : PubMed/NCBI

19 

Cole SP and Deeley RG: Transport of glutathione and glutathione conjugates by MRP1. Trends Pharmacol Sci. 27:438–446. 2006. View Article : Google Scholar : PubMed/NCBI

20 

Nguyen YTK, Park JS, Jang JY, Kim KR, Vo TTL, Kim KW and Han BW: Structural and functional analyses of human ChaC2 in glutathione metabolism. Biomolecules. 10:312019. View Article : Google Scholar : PubMed/NCBI

21 

Orlowski M and Meister A: The gamma-glutamyl cycle: A possible transport system for amino acids. Proc Natl Acad Sci USA. 67:1248–1255. 1970. View Article : Google Scholar : PubMed/NCBI

22 

Hanigan MH: Gamma-glutamyl transpeptidase: Redox regulation and drug resistance. Adv Cancer Res. 122:103–141. 2014. View Article : Google Scholar : PubMed/NCBI

23 

Verma VV, Gupta R and Goel M: Phylogenetic and evolutionary analysis of functional divergence among gamma glutamyl transpeptidase (GGT) subfamilies. Biol Direct. 10:492015. View Article : Google Scholar

24 

Mistry D and Stockley RA: Gamma-glutamyl transferase: The silent partner? COPD. 7:285–290. 2010. View Article : Google Scholar : PubMed/NCBI

25 

Hanigan MH and Frierson HF Jr: Immunohistochemical detection of gamma-glutamyl transpeptidase in normal human tissue. J Histochem Cytochem. 44:1101–1108. 1996. View Article : Google Scholar : PubMed/NCBI

26 

Figlewicz DA, Delattre O, Guellaen G, Krizus A, Thomas G, Zucman J and Rouleau GA: Mapping of human gamma-glutamyl transpeptidase genes on chromosome 22 and other human autosomes. Genomics. 17:299–305. 1993. View Article : Google Scholar : PubMed/NCBI

27 

Morris C, Courtay C, van Kessel AG, ten Hoeve J, Heisterkamp N and Groffen J: Localization of a gamma-glutamyl-transferase-related gene family on chromosome 22. Hum Genet. 91:31–36. 1993. View Article : Google Scholar : PubMed/NCBI

28 

Heisterkamp N, Groffen J, Warburton D and Sneddon TP: The human gamma-glutamyltransferase gene family. Hum Genet. 123:321–232. 2008. View Article : Google Scholar : PubMed/NCBI

29 

Wickham S, West MB, Cook PF and Hanigan MH: Gamma-glutamyl compounds: Substrate specificity of gamma-glutamyl transpeptidase enzymes. Anal Biochem. 414:208–214. 2011. View Article : Google Scholar : PubMed/NCBI

30 

Mottaghitalab F, Lanjanian H and Masoudi-Nejad A: Revealing transcriptional and post-transcriptional regulatory mechanisms of γ-glutamyl transferase and keratin isoforms as novel cooperative biomarkers in low-grade glioma and glioblastoma multiforme. Genomics. 113:2623–2633. 2021. View Article : Google Scholar : PubMed/NCBI

31 

Wang X, Zhang L, Chan FKL, Ji J, Yu J and Liang JQ: Gamma-glutamyltransferase 7 suppresses gastric cancer by cooperating with RAB7 to induce mitophagy. Oncogene. 41:3485–3497. 2022. View Article : Google Scholar : PubMed/NCBI

32 

Tian S, Li J, Guo Y, Dong W and Zheng X: Expression status and prognostic significance of gamma-glutamyl transpeptidase family genes in hepatocellular carcinoma. Front Oncol. 11:7311442021. View Article : Google Scholar : PubMed/NCBI

33 

Samgina TA and Lazarenko VA: The role of polymorphic variants rs11546155 and rs6119534 of the GGT7 gene and risk factors in the development of acute pancreatitis. Vopr Pitan. 91:43–50. 2022.In Russian.

34 

West MB, Wickham S, Parks EE, Sherry DM and Hanigan MH: Human GGT2 does not autocleave into a functional enzyme: A cautionary tale for interpretation of microarray data on redox signaling. Antioxid Redox Signal. 19:1877–1888. 2013. View Article : Google Scholar : PubMed/NCBI

35 

Moon DO, Kim BY, Jang JH, Kim MO, Jayasooriya RG, Kang CH, Choi YH, Moon SK, Kim WJ, Ahn JS and Kim GY: K-RAS transformation in prostate epithelial cell overcomes H2O2-induced apoptosis via upregulation of gamma-glutamyltransferase-2. Toxicol In Vitro. 26:429–434. 2012. View Article : Google Scholar : PubMed/NCBI

36 

Courtay C, Heisterkamp N, Siest G and Groffen J: Expression of multiple gamma-glutamyltransferase genes in man. Biochem J. 297:503–508. 1994. View Article : Google Scholar : PubMed/NCBI

37 

Taniguchi N and Ikeda Y: Gamma-glutamyl transpeptidase: Catalytic mechanism and gene expression. Adv Enzymol Relat Areas Mol Biol. 72:239–278. 1998.PubMed/NCBI

38 

Zhang H and Forman HJ: Redox regulation of gamma-glutamyl transpeptidase. Am J Respir Cell Mol Biol. 41:509–515. 2009. View Article : Google Scholar : PubMed/NCBI

39 

Daubeuf S, Duvoix A, Wellman-Rousseau M, Diederich M and Visvikis A: Phorbol ester regulation of the human gamma-glutamyltransferase gene promoter. Biochem Biophys Res Commun. 313:300–307. 2004. View Article : Google Scholar

40 

Pawlak A, Lahuna O, Bulle F, Suzuki A, Ferry N, Siegrist S, Chikhi N, Chobert MN, Guellaen G and Laperche Y: Gamma-glutamyl transpeptidase: A single copy gene in the rat and a multigene family in the human genome. J Biol Chem. 263:9913–9916. 1988. View Article : Google Scholar : PubMed/NCBI

41 

Pandur S, Pankiv S, Johannessen M, Moens U and Huseby NE: Gamma-glutamyltransferase is upregulated after oxidative stress through the Ras signal transduction pathway in rat colon carcinoma cells. Free Radic Res. 41:1376–1384. 2007. View Article : Google Scholar : PubMed/NCBI

42 

Downward J: Targeting RAS signalling pathways in cancer therapy. Nat Rev Cancer. 3:11–22. 2003. View Article : Google Scholar : PubMed/NCBI

43 

Zhang H, Liu H, Iles KE, Liu RM, Postlethwait EM, Laperche Y and Forman HJ: 4-Hydroxynonenal induces rat gamma-glutamyl transpeptidase through mitogen-activated protein kinase-mediated electrophile response element/nuclear factor erythroid 2-related factor 2 signaling. Am J Respir Cell Mol Biol. 34:174–181. 2006. View Article : Google Scholar

44 

Mussbacher M, Derler M, Basilio J and Schmid JA: NF-κB in monocytes and macrophages-an inflammatory master regulator in multitalented immune cells. Front Immunol. 14:11346612023. View Article : Google Scholar

45 

Yao C, Ren J, Huang R, Tang C, Cheng Y, Lv Z, Kong L, Fang S, Tao J, Fu Y, et al: Transcriptome profiling of microRNAs reveals potential mechanisms of manual therapy alleviating neuropathic pain through microRNA-547-3p-mediated Map4k4/NF-κb signaling pathway. J Neuroinflammation. 19:2112022. View Article : Google Scholar

46 

Reuter S, Schnekenburger M, Cristofanon S, Buck I, Teiten MH, Daubeuf S, Eifes S, Dicato M, Aggarwal BB, Visvikis A and Diederich M: Tumor necrosis factor alpha induces gamma-glutamyltransferase expression via nuclear factor-kappaB in cooperation with Sp1. Biochem Pharmacol. 77:397–411. 2009. View Article : Google Scholar

47 

Kaur A, Gautam R, Srivastava R, Chandel A, Kumar A, Karthikeyan S and Bachhawat AK: ChaC2, an enzyme for slow turnover of cytosolic glutathione. J Biol Chem. 292:638–651. 2017. View Article : Google Scholar :

48 

Crawford RR, Prescott ET, Sylvester CF, Higdon AN, Shan J, Kilberg MS and Mungrue IN: Human CHAC1 protein degrades glutathione, and mRNA induction is regulated by the transcription factors ATF4 and ATF3 and a bipartite ATF/CRE regulatory element. J Biol Chem. 290:15878–15891. 2015. View Article : Google Scholar : PubMed/NCBI

49 

Zhang B, Jiang M and Zheng M: Biological characteristics and functions of ChaC1-catalyzed clutathione degradation in the cytoplasm. Chin J Biochem Mol Biol. 38:284–289. 2022.

50 

Chand S, Mehta V, Sharma RK, Anvikar AR and Chander H: Cancer informatics analysis indicates high CHAC2 associated with unfavorable prognosis in breast cancer. Front Oncol. 12:10589312022. View Article : Google Scholar : PubMed/NCBI

51 

Tsunoda S, Avezov E, Zyryanova A, Konno T, Mendes-Silva L, Pinho Melo E, Harding HP and Ron D: Intact protein folding in the glutathione-depleted endoplasmic reticulum implicates alternative protein thiol reductants. Elife. 3:e034212014. View Article : Google Scholar : PubMed/NCBI

52 

Suyal S, Choudhury C and Bachhawat AK: The ChaC1 active site: Defining the residues and determining the role of ChaC1-exclusive residues in the structural and functional stability. Proteins. 91:567–580. 2023. View Article : Google Scholar

53 

Oh-Hashi K, Nomura Y, Shimada K, Koga H, Hirata Y and Kiuchi K: Transcriptional and post-translational regulation of mouse cation transport regulator homolog 1. Mol Cell Biochem. 380:97–106. 2013. View Article : Google Scholar : PubMed/NCBI

54 

Chen MS, Wang SF, Hsu CY, Yin PH, Yeh TS, Lee HC and Tseng LM: CHAC1 degradation of glutathione enhances cystine-starvation-induced necroptosis and ferroptosis in human triple negative breast cancer cells via the GCN2-eIF2α-ATF4 pathway. Oncotarget. 8:114588–114602. 2017. View Article : Google Scholar

55 

Grimm C, Hofstetter G, Aust S, Mutz-Dehbalaie I, Bruch M, Heinze G, Rahhal-Schupp J, Reinthaller A, Concin N and Polterauer S: Association of gamma-glutamyltransferase with severity of disease at diagnosis and prognosis of ovarian cancer. Br J Cancer. 109:610–614. 2013. View Article : Google Scholar : PubMed/NCBI

56 

Hofbauer SL, Stangl KI, de Martino M, Lucca I, Haitel A, Shariat SF and Klatte T: Pretherapeutic gamma-glutamyltransferase is an independent prognostic factor for patients with renal cell carcinoma. Br J Cancer. 111:1526–1531. 2014. View Article : Google Scholar : PubMed/NCBI

57 

Wei JR, Dong J and Li L: Cancer-associated fibroblasts-derived gamma-glutamyltransferase 5 promotes tumor growth and drug resistance in lung adenocarcinoma. Aging (Albany NY). 12:13220–13233. 2020. View Article : Google Scholar : PubMed/NCBI

58 

Huang Y, Zhou H, Zou J and Wang D: GGT5 is an independent prognostic biomarker in stomach adenocarcinoma. Can J Gastroenterol Hepatol. 2022:99833512022. View Article : Google Scholar : PubMed/NCBI

59 

Xiao Y, Yang H, Lu J, Li D, Xu C and Risch HA: Serum gamma-glutamyltransferase and the overall survival of metastatic pancreatic cancer. BMC Cancer. 19:10202019. View Article : Google Scholar : PubMed/NCBI

60 

Wang Q, Shu X, Dong Y, Zhou J, Teng R, Shen J, Chen Y, Dong M, Zhang W, Huang Y, et al: Tumor and serum gamma-glutamyl transpeptidase, new prognostic and molecular interpretation of an old biomarker in gastric cancer. Oncotarget. 8:361712017. View Article : Google Scholar : PubMed/NCBI

61 

Durham JR, Frierson HF and Hanigan MH: Gamma-glutamyl transpeptidase immunoreactivity in benign and malignant breast tissue. Breast Cancer Res Treat. 45:55–62. 1997. View Article : Google Scholar : PubMed/NCBI

62 

Wu J, Wang X, Wang N, Ma L, Xie X, Zhang H, Kang H and Zhou Z: Identification of novel antioxidant gene signature to predict the prognosis of patients with gastric cancer. World J Surg Oncol. 19:2192021. View Article : Google Scholar : PubMed/NCBI

63 

Zhang L, Li C, Zhang Y, Zhang J and Yang X: Ophiopogonin B induces gastric cancer cell death by blocking the GPX4/xCT-dependent ferroptosis pathway. Oncol Lett. 23:1042022. View Article : Google Scholar : PubMed/NCBI

64 

Gagliardi M, Cotella D, Santoro C, Cora D, Barlev NA, Piacentini M and Corazzari M: Aldo-keto reductases protect metastatic melanoma from ER stress-independent ferroptosis. Cell Death Dis. 10:9022019. View Article : Google Scholar : PubMed/NCBI

65 

He S, Zhang M, Ye Y, Zhuang J, Ma X, Song Y and Xia W: ChaC glutathione specific γ-glutamylcyclotransferase 1 inhibits cell viability and increases the sensitivity of prostate cancer cells to docetaxel by inducing endoplasmic reticulum stress and ferroptosis. Exp Ther Med. 22:9972021. View Article : Google Scholar

66 

Wang Z, Li M, Liu Y, Qiao Z, Bai T, Yang L and Liu B: Dihydroartemisinin triggers ferroptosis in primary liver cancer cells by promoting and unfolded protein response-induced upregulation of CHAC1 expression. Oncol Rep. 46:2402021. View Article : Google Scholar :

67 

Wang N, Zeng GZ, Yin JL and Bian ZX: Artesunate activates the ATF4-CHOP-CHAC1 pathway and affects ferroptosis in Burkitt's Lymphoma. Biochem Biophys Res Commun. 519:533–539. 2019. View Article : Google Scholar : PubMed/NCBI

68 

Joo NE, Ritchie K, Kamarajan P, Miao D and Kapila YL: Nisin, an apoptogenic bacteriocin and food preservative, attenuates HNSCC tumorigenesis via CHAC1. Cancer Med. 1:295–305. 2012. View Article : Google Scholar

69 

Chen PH, Shen WL, Shih CM, Ho KH, Cheng CH, Lin CW, Lee CC, Liu AJ and Chen KC: The CHAC1-inhibited Notch3 pathway is involved in temozolomide-induced glioma cytotoxicity. Neuropharmacology. 116:300–314. 2017. View Article : Google Scholar

70 

Li D, Liu S, Xu J, Chen L, Xu C, Chen F, Xu Z, Zhang Y, Xia S, Shao Y and Wang Y: Ferroptosis-related gene CHAC1 is a valid indicator for the poor prognosis of kidney renal clear cell carcinoma. J Cell Mol Med. 25:3610–3621. 2021. View Article : Google Scholar : PubMed/NCBI

71 

Tseng HH, Chen YZ, Chou NH, Chen YC, Wu CC, Liu LF, Yang YF, Yeh CY, Kung ML, Tu YT and Tsai KW: Metformin inhibits gastric cancer cell proliferation by regulation of a novel Loc100506691-CHAC1 axis. Mol Ther Oncolytics. 22:180–194. 2021. View Article : Google Scholar : PubMed/NCBI

72 

Xiao R, Wang S, Guo J, Liu S, Ding A, Wang G, Li W, Zhang Y, Bian X, Zhao S and Qiu W: Ferroptosis-related gene NOX4, CHAC1 and HIF1A are valid biomarkers for stomach adenocarcinoma. J Cell Mol Med. 26:1183–1193. 2022. View Article : Google Scholar : PubMed/NCBI

73 

Liu Q, Yu M and Zhang T: Construction of oxidative stress-related genes risk model predicts the prognosis of uterine corpus endometrial cancer patients. Cancers (Basel). 14:55722022. View Article : Google Scholar : PubMed/NCBI

74 

Mehta V, Suman P and Chander H: High levels of unfolded protein response component CHAC1 associates with cancer progression signatures in malignant breast cancer tissues. Clin Transl Oncol. 24:2351–2365. 2022. View Article : Google Scholar : PubMed/NCBI

75 

Liu Y, Li M, Shi D and Zhu Y: Higher expression of cation transport regulator-like protein 1 (CHAC1) predicts of poor outcomes in uveal melanoma (UM) patients. Int Ophthalmol. 39:2825–2832. 2019. View Article : Google Scholar : PubMed/NCBI

76 

Liu C, Liu Y, Yu Y, Zhao Y and Yu A: Comprehensive analysis of ferroptosis-related genes and prognosis of cutaneous melanoma. BMC Med Genomics. 15:392022. View Article : Google Scholar : PubMed/NCBI

77 

Goebel G, Berger R, Strasak AM, Egle D, Müller-Holzner E, Schmidt S, Rainer J, Presul E, Parson W, Lang S, et al: Elevated mRNA expression of CHAC1 splicing variants is associated with poor outcome for breast and ovarian cancer patients. Br J Cancer. 106:189–198. 2012. View Article : Google Scholar :

78 

Wang CK, Yang SC, Hsu SC, Chang FP, Lin YT, Chen SF, Cheng CL, Hsiao M, Lu FL and Lu J: CHAC2 is essential for self-renewal and glutathione maintenance in human embryonic stem cells. Free Radic Biol Med. 113:439–451. 2017. View Article : Google Scholar : PubMed/NCBI

79 

Liu S, Fei W, Shi Q, Li Q, Kuang Y, Wang C, He C and Hu X: CHAC2, downregulated in gastric and colorectal cancers, acted as a tumor suppressor inducing apoptosis and autophagy through unfolded protein response. Cell Death Dis. 8:e30092017. View Article : Google Scholar : PubMed/NCBI

80 

Tchantchou F, Graves M, Ashline D, Morin A, Pimenta A, Ortiz D, Rogers E and Shea TB: Increased transcription and activity of glutathione synthase in response to deficiencies in folate, vitamin E, and apolipoprotein E. J Neurosci Res. 75:508–515. 2004. View Article : Google Scholar : PubMed/NCBI

81 

Peng W, Wen L, Jiang R, Deng J and Chen M: CHAC2 promotes lung adenocarcinoma by regulating ROS-mediated MAPK pathway activation. J Cancer. 14:1309–1320. 2023. View Article : Google Scholar : PubMed/NCBI

82 

Tian Y, Lu J and Qiao Y: A metabolism-associated gene signature for prognosis prediction of hepatocellular carcinoma. Front Mol Biosci. 9:9883232022. View Article : Google Scholar : PubMed/NCBI

83 

Lisek K, Campaner E, Ciani Y, Walerych D and Del Sal G: Mutant p53 tunes the NRF2-dependent antioxidant response to support survival of cancer cells. Oncotarget. 9:20508–20523. 2018. View Article : Google Scholar : PubMed/NCBI

84 

Joyce-Brady M, Jean JC and Hughey RP: Gamma-glutamyltransferase and its isoform mediate an endoplasmic reticulum stress response. J Biol Chem. 276:9468–9477. 2001. View Article : Google Scholar

85 

Hayashima K and Katoh H: Expression of gamma-glutamyltransferase 1 in glioblastoma cells confers resistance to cystine deprivation-induced ferroptosis. J Biol Chem. 298:1017032022. View Article : Google Scholar : PubMed/NCBI

86 

Ramsay EE and Dilda PJ: Glutathione S-conjugates as prodrugs to target drug-resistant tumors. Front Pharmacol. 5:1812014. View Article : Google Scholar : PubMed/NCBI

87 

Liu Y, Hyde AS, Simpson MA and Barycki JJ: Emerging regulatory paradigms in glutathione metabolism. Adv Cancer Res. 122:69–101. 2014. View Article : Google Scholar : PubMed/NCBI

88 

Lieberman MW, Wiseman AL, Shi ZZ, Carter BZ, Barrios R, Ou CN, Chévez-Barrios P, Wang Y, Habib GM, Goodman JC, et al: Growth retardation and cysteine deficiency in gamma-glutamyl transpeptidase-deficient mice. Proc Natl Acad Sci USA. 93:7923–7926. 1996. View Article : Google Scholar : PubMed/NCBI

89 

Barrios R, Shi ZZ, Kala SV, Wiseman AL, Welty SE, Kala G, Bahler AA, Ou CN and Lieberman MW: Oxygen-induced pulmonary injury in gamma-glutamyl transpeptidase-deficient mice. Lung. 179:319–330. 2001. View Article : Google Scholar

90 

Rojas E, Valverde M, Kala SV, Kala G and Lieberman MW: Accumulation of DNA damage in the organs of mice deficient in gamma-glutamyltranspeptidase. Mutat Res. 447:305–316. 2000. View Article : Google Scholar

91 

Darin N, Leckström K, Sikora P, Lindgren J, Almén G and Asin-Cayuela J: γ-glutamyl transpeptidase deficiency caused by a large homozygous intragenic deletion in GGT1. Eur J Hum Genet. 26:808–817. 2018. View Article : Google Scholar : PubMed/NCBI

92 

Alessandro C, Maria F, Aldo P and Alfonso P: Gamma-glutamyltransferase of cancer cells at the crossroads of tumor progression, drug resistance and drug targeting. Anticancer Res. 30:1169–1181. 2010.

93 

Giommarelli C, Corti A, Supino R, Favini E, Paolicchi A, Pompella A and Zunino F: Cellular response to oxidative stress and ascorbic acid in melanoma cells overexpressing gamma-glutamyltransferase. Eur J Cancer. 44:750–759. 2008. View Article : Google Scholar : PubMed/NCBI

94 

Panis C, Victorino VJ, Herrera AC, Freitas LF, De Rossi T, Campos FC, Simão AN, Barbosa DS, Pinge-Filho P, Cecchini R and Cecchini AL: Differential oxidative status and immune characterization of the early and advanced stages of human breast cancer. Breast Cancer Res Treat. 133:881–888. 2012. View Article : Google Scholar

95 

Chen X and Cubillos-Ruiz JR: Endoplasmic reticulum stress signals in the tumour and its microenvironment. Nat Rev Cancer. 21:71–88. 2021. View Article : Google Scholar :

96 

Shi Y, Jiang B and Zhao J: Induction mechanisms of autophagy and endoplasmic reticulum stress in intestinal ischemia-reperfusion injury, inflammatory bowel disease, and colorectal cancer. Biomed Pharmacother. 170:1159842024. View Article : Google Scholar

97 

Chen H, Xu N, Xu J, Zhang C, Li X, Xu H, Zhu W, Li J, Liang D and Zhou W: A risk signature based on endoplasmic reticulum stress-associated genes predicts prognosis and immunity in pancreatic cancer. Front Mol Biosci. 10:12980772023. View Article : Google Scholar : PubMed/NCBI

98 

Joyce-Brady M, Jean JC and Hughey RP: Gamma-glutamyltransferase and its isoform mediate an endoplasmic reticulum stress response. J Biol Chem. 276:9468–9477. 2001. View Article : Google Scholar

99 

Cui Y, Zhou X, Chen L, Tang Z, Mo F, Li XC, Mao H, Wei X, Wang C and Wang H: Crosstalk between endoplasmic reticulum stress and oxidative stress in heat exposure-induced apoptosis is dependent on the ATF4-CHOP-CHAC1 signal pathway in IPEC-J2 cells. J Agric Food Chem. 69:15495–15511. 2021. View Article : Google Scholar : PubMed/NCBI

100 

Tomonobu N, Komalasari NLGY, Sumardika IW, Jiang F, Chen Y, Yamamoto KI, Kinoshita R, Murata H, Inoue Y and Sakaguchi M: Xylitol acts as an anticancer monosaccharide to induce selective cancer death via regulation of the glutathione level. Chem Biol Interact. 324:1090852020. View Article : Google Scholar : PubMed/NCBI

101 

Lv H, Zhen C, Liu J, Yang P, Hu L and Shang P: Unraveling the potential role of glutathione in multiple forms of cell death in cancer therapy. Oxid Med Cell Longev. 2019:31501452019. View Article : Google Scholar : PubMed/NCBI

102 

Wen RJ, Dong X, Zhuang HW, Pang FX, Ding SC, Li N, Mai YX, Zhou ST, Wang JY and Zhang JF: Baicalin induces ferroptosis in osteosarcomas through a novel Nrf2/xCT/GPX4 regulatory axis. Phytomedicine. 116:1548812023. View Article : Google Scholar : PubMed/NCBI

103 

Elmore S: Apoptosis: A review of programmed cell death. Toxicol Pathol. 35:495–516. 2007. View Article : Google Scholar : PubMed/NCBI

104 

Pfeffer CM and Singh ATK: Apoptosis: A target for anticancer therapy. Int J Mol Sci. 19:4482018. View Article : Google Scholar : PubMed/NCBI

105 

Brancaccio M, Russo M, Masullo M, Palumbo A, Russo GL and Castellano I: Sulfur-containing histidine compounds inhibit γ-glutamyl transpeptidase activity in human cancer cells. J Biol Chem. 294:14603–14614. 2019. View Article : Google Scholar : PubMed/NCBI

106 

Bui TT, Nitta RT, Kahn SA, Razavi SM, Agarwal M, Aujla P, Gholamin S, Recht L and Li G: γ-Glutamyl transferase 7 is a novel regulator of glioblastoma growth. BMC Cancer. 15:2252015. View Article : Google Scholar

107 

Hirschhorn T and Stockwell BR: The development of the concept of ferroptosis. Free Radic Biol Med. 133:130–143. 2019. View Article : Google Scholar :

108 

Lei G, Zhuang L and Gan B: Targeting ferroptosis as a vulnerability in cancer. Nat Rev Cancer. 22:381–396. 2022. View Article : Google Scholar : PubMed/NCBI

109 

Wu J, Wen T, Marzio A, Song D, Chen S, Yang C, Zhao F, Zhang B, Zhao G, Ferri A, et al: FBXO32-mediated degradation of PTEN promotes lung adenocarcinoma progression. Cell Death Dis. 15:2822024. View Article : Google Scholar : PubMed/NCBI

110 

Zhan CH, Ding DS, Zhang W, Wang HL, Mao ZY and Liu GJ: The cancer-testis antigen a-kinase anchor protein 3 facilitates breast cancer progression via activation of the PTEN/PI3K/AKT/mTOR signaling. Bioengineered. 13:8478–8489. 2022. View Article : Google Scholar : PubMed/NCBI

111 

Yi J, Zhu J, Wu J, Thompson CB and Jiang X: Oncogenic activation of PI3K-AKT-mTOR signaling suppresses ferroptosis via SREBP-mediated lipogenesis. Proc Natl Acad Sci USA. 117:31189–31197. 2020. View Article : Google Scholar : PubMed/NCBI

112 

Accaoui MJ, Enoiu M, Mergny M, Masson C, Dominici S, Wellman M and Visvikis A: Gamma-glutamyltranspeptidase-dependent glutathione catabolism results in activation of NF-kB. Biochem Biophys Res Commun. 276:1062–1067. 2000. View Article : Google Scholar : PubMed/NCBI

113 

Djavaheri-Mergny M, Accaoui MJ, Rouillard D and Wietzerbin J: Gamma-glutamyl transpeptidase activity mediates NF-kappaB activation through lipid peroxidation in human leukemia U937 cells. Mol Cell Biochem. 232:103–111. 2002. View Article : Google Scholar : PubMed/NCBI

114 

Franzini M, Corti A, Lorenzini E, Paolicchi A, Pompella A, De Cesare M, Perego P, Gatti L, Leone R, Apostoli P and Zunino F: Modulation of cell growth and cisplatin sensitivity by membrane gamma-glutamyltransferase in melanoma cells. Eur J Cancer. 42:2623–2630. 2006. View Article : Google Scholar : PubMed/NCBI

115 

Yu X and Long YC: Crosstalk between cystine and glutathione is critical for the regulation of amino acid signaling pathways and ferroptosis. Sci Rep. 6:300332016. View Article : Google Scholar : PubMed/NCBI

116 

Tang X, Ding CK, Wu J, Sjol J, Wardell S, Spasojevic I, George D, McDonnell DP, Hsu DS, Chang JT and Chi JT: Cystine addiction of triple-negative breast cancer associated with EMT augmented death signaling. Oncogene. 36:4235–4242. 2017. View Article : Google Scholar

117 

Dixon SJ, Patel DN, Welsch M, Skouta R, Lee ED, Hayano M, Thomas AG, Gleason CE, Tatonetti NP, Slusher BS and Stockwell BR: Pharmacological inhibition of cystine-glutamate exchange induces endoplasmic reticulum stress and ferroptosis. Elife. 3:e025232014. View Article : Google Scholar : PubMed/NCBI

118 

Galluzzi L, Kepp O, Chan FKM and Kroemer G: Necroptosis: Mechanisms and relevance to disease. Annu Rev Pathol. 12:103–130. 2017. View Article : Google Scholar

119 

Zhang G, Wang J, Zhao Z, Xin T, Fan X, Shen Q, Raheem A, Lee CR, Jiang H and Ding J: Regulated necrosis, a proinflammatory cell death, potentially counteracts pathogenic infections. Cell Death Dis. 13:6372022. View Article : Google Scholar : PubMed/NCBI

120 

Yang Z and Klionsky DJ: An overview of the molecular mechanism of autophagy. Curr Top Microbiol Immunol. 335:1–32. 2009.PubMed/NCBI

121 

Xu G, Wang J, Zhang Y, Chen Z and Deng R: GGT1 suppresses the development of ferroptosis and autophagy in mouse retinal ganglion cell through targeting GCLC. Eye Brain. 15:139–151. 2023. View Article : Google Scholar : PubMed/NCBI

122 

Sohrab SS, Raj R, Nagar A, Hawthorne S, Paiva-Santos AC, Kamal MA, El-Daly MM, Azhar EI and Sharma A: Chronic inflammation's transformation to cancer: A nanotherapeutic paradigm. Molecules. 28:44132023. View Article : Google Scholar : PubMed/NCBI

123 

Ying HQ, Liao YC, Luo YR, Xiong G, Huang Y, Nie RW, Xiong CF and Cheng XX: Cancer-elicited inflammation attenuates response and outcome in tyrosine kinase inhibitor naive patients with advanced NSCLC. Pharmacol Res. 170:1057342021. View Article : Google Scholar : PubMed/NCBI

124 

Carr B and Guerra V: Serum inflammation parameters and survival in hepatocellular carcinoma patients: Importance of albumin and gamma-glutamyltranspeptidase. Oncology. 101:313–320. 2023. View Article : Google Scholar : PubMed/NCBI

125 

Ozcelik F: Prognostic value of gamma-glutamyl transpeptidase in liver cirrhosis and hepatocellular cancer regardless of other parameters. Clin Res Hepatol Gastroenterol. 45:1017082021. View Article : Google Scholar : PubMed/NCBI

126 

Singh J, Chander J, Singh S, Singh G and Atal CK: Gamma-glutamyl transpeptidase: A novel biochemical marker in inflammation. Biochem Pharmacol. 35:3753–3760. 1986. View Article : Google Scholar : PubMed/NCBI

127 

Ricci V, Giannouli M, Romano M and Zarrilli R: Helicobacter pylori gamma-glutamyl transpeptidase and its pathogenic role. World J Gastroenterol. 20:630–638. 2014. View Article : Google Scholar : PubMed/NCBI

128 

Oertli M, Noben M, Engler DB, Semper RP, Reuter S, Maxeiner J, Gerhard M, Taube C and Müller A: Helicobacter pylori γ-glutamyl transpeptidase and vacuolating cytotoxin promote gastric persistence and immune tolerance. Proc Natl Acad Sci USA. 110:3047–3052. 2013. View Article : Google Scholar

129 

Li N, Ouyang Y, Chen S, Peng C, He C, Hong J, Yang X, Zhu Y and Lu NH: Integrative analysis of differential lncRNA/mRNA expression profiling in Helicobacter pylori infection-associated gastric carcinogenesis. Front Microbiol. 11:8802020. View Article : Google Scholar : PubMed/NCBI

130 

Wada Y, Takemura K, Tummala P, Uchida K, Kitagaki K, Furukawa A, Ishige Y, Ito T, Hara Y, Suzuki T, et al: Helicobacter pylori induces somatic mutations in TP53 via overexpression of CHAC1 in infected gastric epithelial cells. FEBS Open Bio. 8:671–679. 2018. View Article : Google Scholar : PubMed/NCBI

131 

Ogawa T, Wada Y, Takemura K, Board PG, Uchida K, Kitagaki K, Tamura T, Suzuki T, Tokairin Y, Nakajima Y and Eishi Y: CHAC1 overexpression in human gastric parietal cells with Helicobacter pylori infection in the secretory canaliculi. Helicobacter. 24:e125982019. View Article : Google Scholar : PubMed/NCBI

132 

Perra L, Balloy V, Foussignière T, Moissenet D, Petat H, Mungrue IN, Touqui L, Corvol H, Chignard M and Guillot L: CHAC1 is differentially expressed in normal and cystic fibrosis bronchial epithelial cells and regulates the inflammatory response induced by Pseudomonas aeruginosa. Front Immunol. 9:28232018. View Article : Google Scholar : PubMed/NCBI

133 

Rousset-Jablonski C, Dalon F, Reynaud Q, Lemonnier L, Dehillotte C, Jacoud F, Berard M, Viprey M, Van Ganse E, Durieu I and Belhassen M: Cancer incidence and prevalence in cystic fibrosis patients with and without a lung transplant in France. Front Public Health. 10:10436912022. View Article : Google Scholar : PubMed/NCBI

134 

Daubeuf S, Leroy P, Paolicchi A, Pompella A, Wellman M, Galteau MM and Visvikis A: Enhanced resistance of HeLa cells to cisplatin by overexpression of gamma-glutamyltransferase. Biochem Pharmacol. 64:207–216. 2002. View Article : Google Scholar : PubMed/NCBI

135 

Zhang C, Xu C, Gao X and Yao Q: Platinum-based drugs for cancer therapy and anti-tumor strategies. Theranostics. 12:2115–2132. 2022. View Article : Google Scholar : PubMed/NCBI

136 

Wang L, Liu Z, He S, He S and Wang Y: Fighting against drug-resistant tumors by the inhibition of γ-glutamyl transferase with supramolecular platinum prodrug nano-assemblies. J Mater Chem B. 9:4587–4595. 2021. View Article : Google Scholar : PubMed/NCBI

137 

Hanigan MH, Frierson HF Jr, Abeler VM, Kaern J and Taylor PT Jr: Human germ cell tumours: Expression of gamma-glutamyl transpeptidase and sensitivity to cisplatin. Br J Cancer. 81:75–79. 1999. View Article : Google Scholar : PubMed/NCBI

138 

Chen J, Zaal EA, Berkers CR, Ruijtenbeek R, Garssen J and Redegeld FA: Omega-3 fatty acids DHA and EPA reduce bortezomib resistance in multiple myeloma cells by promoting glutathione degradation. Cells. 10:22872021. View Article : Google Scholar : PubMed/NCBI

139 

Yu X, He Z, Wang Z, Ke S, Wang H, Wang Q and Li S: Brusatol hinders the progression of bladder cancer by Chac1/Nrf2/SLC7A11 pathway. Exp Cell Res. 438:1140532024. View Article : Google Scholar : PubMed/NCBI

140 

Wang X, He MJ, Chen XJ, Bai YT and Zhou G: Glaucocalyxin A impairs tumor growth via amplification of the ATF4/CHOP/CHAC1 cascade in human oral squamous cell carcinoma. J Ethnopharmacol. 290:1151002022. View Article : Google Scholar : PubMed/NCBI

141 

Zhou H, Shen Q, Fu J, Jiang F, Wang L and Wang Y: Analysis of lncRNA UCA1-related downstream pathways and molecules of cisplatin resistance in lung adenocarcinoma. J Clin Lab Anal. 34:e233122020. View Article : Google Scholar : PubMed/NCBI

142 

Ha Y, Chon YE, Kim MN, Lee JH and Hwang SG: Gamma-glutamyl transpeptidase dynamics as a biomarker for advanced fibrosis in non-alcoholic fatty liver disease. J Gastroenterol Hepatol. 37:1624–1632. 2022. View Article : Google Scholar : PubMed/NCBI

143 

Zhang S, Xu L and Xu M: Gamma-glutamyl transpeptidase to albumin ratio holds a prognostic significance after hepatectomy in patients with hepatocellular carcinoma and liver cirrhosis. Asian J Surg. 46:1327–1328. 2023. View Article : Google Scholar

144 

Yamada Y, Ishizaki M, Kido T, Honda R, Tsuritani I, Ikai E and Yamaya H: Alcohol, high blood pressure, and serum gamma-glutamyl transpeptidase level. Hypertension. 18:819–826. 1991. View Article : Google Scholar : PubMed/NCBI

145 

Takemura K, Board PG and Koga F: A systematic review of serum γ-glutamyltransferase as a prognostic biomarker in patients with genitourinary cancer. Antioxidants (Basel). 10:5492021. View Article : Google Scholar

146 

Chen RQ, Zhang ZL, Jia YM, Chen RX and Peng L: Preoperative CA19-9 and GGT ratio as a prognostic indicator in ampullary carcinoma. BMC Gastroenterol. 23:722023. View Article : Google Scholar : PubMed/NCBI

147 

Xie H, Gao J, Sun X, Song Y, Zhang Q, Zhang P and Ding C: A water-soluble fluorescent probe for the determination of γ-glutamyltransferase activity and its application in tumor imaging. Talanta. 253:1239432023. View Article : Google Scholar

148 

Gao D, Miao Y, Ye S, Lu C, Lv G, Li K, Yu C, Lin J and Qiu L: A fluorine-18 labeled radiotracer for PET imaging of γ-glutamyltranspeptidase in living subjects. RSC Adv. 11:18738–18747. 2021. View Article : Google Scholar

149 

Ou-Yang J, Li Y, Jiang WL, He SY, Liu HW and Li CY: Fluorescence-guided cancer diagnosis and surgery by a zero cross-talk ratiometric near-infrared γ-glutamyltranspeptidase fluorescent probe. Anal Chem. 91:1056–1063. 2019. View Article : Google Scholar

150 

Jin Y, Wang Z, He D, Zhu Y, Gong L, Xiao M, Chen X and Cao K: Analysis of ferroptosis-mediated modification patterns and tumor immune microenvironment characterization in uveal melanoma. Front Cell Dev Biol. 9:6851202021. View Article : Google Scholar : PubMed/NCBI

151 

Mehta V, Meena J, Kasana H, Munshi A and Chander H: Prognostic significance of CHAC1 expression in breast cancer. Mol Biol Rep. 49:8517–8526. 2022. View Article : Google Scholar : PubMed/NCBI

152 

Jahn B, Arvandi M, Rochau U, Fiegl H, Goebel G, Marth C and Siebert U: Development of a novel prognostic score for breast cancer patients using mRNA expression of CHAC1. J Comp Eff Res. 6:563–574. 2017. View Article : Google Scholar : PubMed/NCBI

153 

Lin D, Hu B, Zhu S and Wu Y: Exploring a ferroptosis and oxidative stress-based prognostic model for clear cell renal cell carcinoma. Front Oncol. 13:11314732023. View Article : Google Scholar : PubMed/NCBI

154 

Chen W, Lei C, Wang Y, Guo D, Zhang S, Wang X, Zhang Z, Wang Y and Ma W: Prognostic prediction model for glioblastoma: A ferroptosis-related gene prediction model and independent external validation. J Clin Med. 12:13412023. View Article : Google Scholar : PubMed/NCBI

155 

Ahluwalia GS, Grem JL, Hao Z and Cooney DA: Metabolism and action of amino acid analog anti-cancer agents. Pharmacol Ther. 46:243–271. 1990. View Article : Google Scholar : PubMed/NCBI

156 

Viña JR, Palacin M, Puertes IR, Hernandez R and Viña J: Role of the gamma-glutamyl cycle in the regulation of amino acid translocation. Am J Physiol. 257:E916–E922. 1989.PubMed/NCBI

157 

Xie Z, Kawasaki T, Zhou H, Okuzaki D, Okada N and Tachibana M: Targeting GGT1 eliminates the tumor-promoting effect and enhanced immunosuppressive function of myeloid-derived suppressor cells caused by G-CSF. Front Pharmacol. 13:8737922022. View Article : Google Scholar : PubMed/NCBI

158 

Stevens AM, Xiang M, Heppler LN, Tošić I, Jiang K, Munoz JO, Gaikwad AS, Horton TM, Long X, Narayanan P, et al: Atovaquone is active against AML by upregulating the integrated stress pathway and suppressing oxidative phosphorylation. Blood Adv. 3:4215–4227. 2019. View Article : Google Scholar : PubMed/NCBI

159 

U Ferreira MJ: Natural product-derived compounds for targeting multidrug resistance in cancer and microorganisms. Int J Mol Sci. 24:143212023. View Article : Google Scholar : PubMed/NCBI

160 

Ashrafizadeh M, Aref AR, Sethi G, Ertas YN and Wang L: Natural product/diet-based regulation of macrophage polarization: Implications in treatment of inflammatory-related diseases and cancer. J Nutr Biochem. 1096472024. View Article : Google Scholar : PubMed/NCBI

161 

Lyons SD, Sant ME and Christopherson RI: Cytotoxic mechanisms of glutamine antagonists in mouse L1210 leukemia. J Biol Chem. 265:11377–11381. 1990. View Article : Google Scholar : PubMed/NCBI

162 

Guan Z, Chen J, Li X and Dong N: Tanshinone IIA induces ferroptosis in gastric cancer cells through p53-mediated SLC7A11 down-regulation. Biosci Rep. 40:BSR202018072020. View Article : Google Scholar : PubMed/NCBI

163 

Townsend DM, Deng M, Zhang L, Lapus MG and Hanigan MH: Metabolism of cisplatin to a nephrotoxin in proximal tubule cells. J Am Soc Nephrol. 14:1–10. 2003. View Article : Google Scholar

164 

Azouz AA, Abdel-Nassir Abdel-Razek E and Abo-Youssef AM: Amlodipine alleviates cisplatin-induced nephrotoxicity in rats through gamma-glutamyl transpeptidase (GGT) enzyme inhibition, associated with regulation of Nrf2/HO-1, MAPK/NF-κB, and Bax/Bcl-2 signaling. Saudi Pharm J. 28:1317–1325. 2020. View Article : Google Scholar : PubMed/NCBI

165 

Xue Y, Lu F, Chang Z, Li J, Gao Y, Zhou J, Luo Y, Lai Y, Cao S, Li X, et al: Intermittent dietary methionine deprivation facilitates tumoral ferroptosis and synergizes with checkpoint blockade. Nat Commun. 14:47582023. View Article : Google Scholar : PubMed/NCBI

166 

Sumi D, Taguchi H, Takeuchi K and Fujishiro H: CHAC1 exacerbates arsenite cytotoxicity by lowering intracellular glutathione levels. J Toxicol Sci. 48:487–494. 2023. View Article : Google Scholar : PubMed/NCBI

167 

Chen S, Fan J, Xie P, Ahn J, Fernandez M, Billingham LK, Miska J, Wu JD, Wainwright DA, Fang D, et al: CD8+ T cells sustain antitumor response by mediating crosstalk between adenosine A2A receptor and glutathione/GPX4. J Clin Invest. 134:e1700712024. View Article : Google Scholar : PubMed/NCBI

168 

Xiong Y, Liu T and Chen J: Anisomycin has the potential to induce human ovarian cancer stem cell ferroptosis by influencing glutathione metabolism and autophagy signal transduction pathways. J Cancer. 14:1202–1215. 2023. View Article : Google Scholar : PubMed/NCBI

169 

Liu CX, Gao Y, Xu XF, Jin X, Zhang Y, Xu Q, Ding HX, Li BJ, Du FK, Li LC, et al: Bile acids inhibit ferroptosis sensitivity through activating farnesoid X receptor in gastric cancer cells. World J Gastroenterol. 30:485–498. 2024. View Article : Google Scholar : PubMed/NCBI

170 

Mitrić A and Castellano I: Targeting gamma-glutamyl transpeptidase: A pleiotropic enzyme involved in glutathione metabolism and in the control of redox homeostasis. Free Radic Biol Med. 208:672–683. 2023. View Article : Google Scholar

171 

Pompella A, Corti A, Paolicchi A, Giommarelli C and Zunino F: Gamma-glutamyltransferase, redox regulation and cancer drug resistance. Curr Opin Pharmacol. 7:360–366. 2007. View Article : Google Scholar : PubMed/NCBI

172 

Bai C, Zhang M, Zhang Y, He Y, Dou H, Wang Z, Wang Z, Li Z and Zhang L: Gamma-glutamyltransferase activity (GGT) is a long-sought biomarker of redox status in blood circulation: A retrospective clinical study of 44 types of human diseases. Oxid Med Cell Longev. 2022:84940762022. View Article : Google Scholar : PubMed/NCBI

173 

Hamano M, Tomonaga S, Osaki Y, Oda H, Kato H and Furuya S: Transcriptional activation of Chac1 and other Atf4-target genes induced by extracellular l-serine depletion is negated with glycine consumption in Hepa1-6 hepatocarcinoma cells. Nutrients. 12:30182020. View Article : Google Scholar : PubMed/NCBI

174 

Ge X, Cai Q, Zhang S, Wu X, Ying P, Ke J and Yang Z: Treatment with paraquat affects the expression of ferroptosis-related genes. Hum Exp Toxicol. 42:96032712311675852023. View Article : Google Scholar : PubMed/NCBI

175 

Dosumu OA, Rotimi SO, Adeleye OO, Akamo AJ, Osinuga KT, Taiwo OA, Omotosho OO and Sani LO: Vitamin K protects against 7,12-dimethylbenz(A)anthracene induced hepatotoxicity in Wistar rats. Environ Toxicol. 36:362–373. 2021. View Article : Google Scholar

176 

Schreiber CL and Smith BD: Molecular imaging of aminopeptidase N in cancer and angiogenesis. Contrast Media Mol Imaging. 2018:53151722018. View Article : Google Scholar : PubMed/NCBI

177 

Amin SA, Adhikari N and Jha T: Design of aminopeptidase N inhibitors as anti-cancer agents. J Med Chem. 61:6468–6490. 2018. View Article : Google Scholar : PubMed/NCBI

178 

Guzman-Rojas L, Rangel R, Salameh A, Edwards JK, Dondossola E, Kim YG, Saghatelian A, Giordano RJ, Kolonin MG, Staquicini FI, et al: Cooperative effects of aminopeptidase N (CD13) expressed by nonmalignant and cancer cells within the tumor microenvironment. Proc Natl Acad Sci USA. 109:1637–1642. 2012. View Article : Google Scholar : PubMed/NCBI

179 

Wang X, Shi L, Deng Y, Qu M, Mao S, Xu L, Xu W and Fang C: Inhibition of leucine aminopeptidase 3 suppresses invasion of ovarian cancer cells through down-regulation of fascin and MMP-2/9. Eur J Pharmacol. 768:116–122. 2015. View Article : Google Scholar : PubMed/NCBI

180 

Xu Y, Zhang N, Chen C, Xu X, Luo A, Yan Y, Lu Y, Liu J, Ou X, Tan Y, et al: Sevoflurane induces ferroptosis of glioma cells through activating the ATF4-CHAC1 pathway. Front Oncol. 12:8596212022. View Article : Google Scholar : PubMed/NCBI

181 

Wen YF, Yang XZ, Zeng LS, Peng HH, Huang WJ, Cai LM, Zhou TC and Lin XD: Prognostic impact of pretherapeutic gamma-glutamyltransferase on patients with nasopharyngeal carcinoma. PLoS One. 12:e01723452017. View Article : Google Scholar : PubMed/NCBI

182 

Mujawar SJ, Suchitra G, Kosandal KA, Choudhari S, Inamdar NA and Ahmed KB: Evaluation of salivary gamma-glutamyl transpeptidase as a biomarker in oral squamous cell carcinoma and precancerous lesions. J Oral Maxillofac Pathol. 24:5842020. View Article : Google Scholar

183 

Mizushima T, Ohnishi S, Shimizu Y, Hatanaka Y, Hatanaka KC, Hosono H, Kubota Y, Natsuizaka M, Kamiya M, Ono S, et al: Fluorescent imaging of superficial head and neck squamous cell carcinoma using a γ-glutamyltranspeptidase-activated targeting agent: A pilot study. BMC Cancer. 16:4112016. View Article : Google Scholar

184 

Lee YJ, Han KD, Kim DH and Lee CH: Determining the association between repeatedly elevated serum gamma-glutamyltransferase levels and risk of respiratory cancer: A nationwide population-based cohort study. Cancer Med. 10:1366–1376. 2021. View Article : Google Scholar : PubMed/NCBI

185 

Gu J, Xie R, Zhao Y, Zhao Z, Xu D, Ding M, Lin T, Xu W, Nie Z, Miao E, et al: A machine learning-based approach to predicting the malignant and metastasis of thyroid cancer. Front Oncol. 12:9382922022. View Article : Google Scholar

186 

Foddis R, Franzini M, Bonotti A, Marino R, Silvestri R, Fallahi P, Chiappino D, Emdin M, Paolicchi A and Cristaudo A: Big and free fractions of gamma-glutamyltransferase: New diagnostic biomarkers for malignant mesothelioma? Diagnostics (Basel). 12:3112022. View Article : Google Scholar : PubMed/NCBI

187 

Huang H, Wang XP, Li XH, Chen H, Zheng X, Lin JH, Kang T, Zhang L and Chen PS: Prognostic value of pretreatment serum alanine aminotransferase/aspartate aminotransferase (ALT/AST) ratio and gamma glutamyltransferase (GGT) in patients with esophageal squamous cell carcinoma. BMC Cancer. 17:5442017. View Article : Google Scholar : PubMed/NCBI

188 

Choi YJ, Lee DH, Han KD, Yoon H, Shin CM, Park YS and Kim N: Elevated serum gamma-glutamyltransferase is associated with an increased risk of oesophageal carcinoma in a cohort of 8,388,256 Korean subjects. PLoS One. 12:e01770532017. View Article : Google Scholar : PubMed/NCBI

189 

Chen Y, Liu H, Zhang J, Wu Y, Zhou W, Cheng Z, Lou J, Zheng S, Bi X, Wang J, et al: Prognostic value and predication model of microvascular invasion in patients with intrahepatic cholangiocarcinoma: A multicenter study from China. BMC Cancer. 21:12992021. View Article : Google Scholar : PubMed/NCBI

190 

Catalano M, Roviello G, Aprile G, Ramello M, Conca R, Petrioli R, Perrone G, Ianza A and Mini E: Prognostic value of alkaline phosphatase and gamma-glutamyl transferase in patients with metastatic pancreatic cancer. Future Oncol. 19:937–946. 2023. View Article : Google Scholar : PubMed/NCBI

191 

Liao W, Yang Y, Yang H, Qu Y, Song H and Li Q: Circulating gamma-glutamyl transpeptidase and risk of pancreatic cancer: A prospective cohort study in the UK Biobank. Cancer Med. 12:7877–7887. 2023. View Article : Google Scholar

192 

Hong SW, Lee HJ, Han K, Moon JM, Park S, Soh H, Kang EA, Chun J, Im JP and Kim JS: Risk of gastrointestinal cancer in patients with an elevated level of gamma-glutamyltransferase: A nationwide population-based study. PLoS One. 16:e02450522021. View Article : Google Scholar : PubMed/NCBI

193 

Yang S, He X, Liu Y, Ding X, Jiang H, Tan Y and Lu H: Prognostic significance of serum uric acid and gamma-glutamyltransferase in patients with advanced gastric cancer. Dis Markers. 2019:14154212019. View Article : Google Scholar : PubMed/NCBI

194 

Hong TC, Yang HC, Chen CL, Kao JH, Liu CJ, Chen MJ, Wang HY, Kuo YC, Yu LY and Hu KC: Relationship between serum gamma-glutamyl transferase level and colorectal adenoma. PLoS One. 15:e02404452020. View Article : Google Scholar : PubMed/NCBI

195 

Yang LH, Xu LZ, Huang ZJ, Pan HH, Wu M, Wu QY, Lu T, Zhang YP, Zhu YB, Wu JB, et al: Comprehensive analysis of immune ferroptosis gene in renal clear cell carcinoma: Prognosis and influence of tumor microenvironment. Am J Transl Res. 14:5982–6010. 2022.PubMed/NCBI

196 

Horie K, Kawakami K, Fujita Y, Matsuda Y, Arai T, Suzui N, Miyazaki T, Koie T, Mizutani K and Ito M: Serum exosomal gamma-glutamyltransferase activity increased in patients with renal cell carcinoma with advanced clinicopathological features. Oncology. 98:734–742. 2020. View Article : Google Scholar : PubMed/NCBI

197 

Pankevičiūtė-Bukauskienė M, Mikalayeva V, Žvikas V, Skeberdis VA and Bordel S: Multi-omics analysis revealed increased de novo synthesis of serine and lower activity of the methionine cycle in breast cancer cell lines. Molecules. 28:45352023. View Article : Google Scholar

198 

Seol A, Wang W, Kim SI, Han Y, Park IS, Yoo J, Jo H, Han KD and Song YS: Enhanced susceptibility to breast cancer in Korean women with elevated serum gamma-glutamyltransferase levels: A nationwide population-based cohort study. Front Oncol. 11:6686242021. View Article : Google Scholar : PubMed/NCBI

199 

Shi B, Zhang Z, Jin Q, Wang Z, Tang J, Xu G, Zhu T, Gong X, Tang X and Zhao C: Selective tracking of ovarian-cancer-specific γ-glutamyltranspeptidase using a ratiometric two-photon fluorescent probe. J Mater Chem B. 6:7439–7443. 2018. View Article : Google Scholar : PubMed/NCBI

200 

Schwameis R, Grimm C, Brodowicz T, Petru E, Hefler-Frischmuth K, Staudigl C, Reinthaller A, Heinze G, Polterauer S and Polterauer M: Gamma-glutamyltransferase as novel biomarker in patients with uterine leiomyosarcoma. Sci Rep. 6:337572016. View Article : Google Scholar : PubMed/NCBI

201 

Polterauer S, Hofstetter G, Grimm C, Rahhal J, Mailath-Pokorny M, Kohl M, Concin N, Tempfer C, Marth C and Reinthaller A: Relevance of gamma-glutamyltransferase-a marker for apoptotic balance-in predicting tumor stage and prognosis in cervical cancer. Gynecol Oncol. 122:590–594. 2011. View Article : Google Scholar : PubMed/NCBI

202 

Kawakami K, Fujita Y, Matsuda Y, Arai T, Horie K, Kameyama K, Kato T, Masunaga K, Kasuya Y, Tanaka M, et al: Gamma-glutamyltransferase activity in exosomes as a potential marker for prostate cancer. BMC Cancer. 17:3162017. View Article : Google Scholar : PubMed/NCBI

203 

Su S, Liu L, Sun C, Nie Y, Guo H, Hu Y, Guo S and Pang S: Preoperative serum gamma-glutamyltransferase as a prognostic biomarker in patients undergoing radical cystectomy for bladder cancer. Front Oncol. 11:6489042021. View Article : Google Scholar : PubMed/NCBI

204 

Takemura K, Fukushima H, Ito M, Kataoka M, Nakanishi Y, Sakamoto K, Suzuki H, Tobisu KI and Koga F: Prognostic significance of serum γ-glutamyltransferase in patients with advanced urothelial carcinoma. Urol Oncol. 37:108–115. 2019. View Article : Google Scholar

205 

Song Y, Tian S, Zhang P, Zhang N, Shen Y and Deng J: Construction and validation of a novel ferroptosis-related prognostic model for acute myeloid leukemia. Front Genet. 12:7086992022. View Article : Google Scholar : PubMed/NCBI

206 

Watanabe B, Tabuchi Y, Wada K and Hiratake J: Synthesis and evaluation of the inhibitory activity of the four stereoisomers of the potent and selective human γ-glutamyl transpeptidase inhibitor GGsTop. Bioorg Med Chem Lett. 27:4920–4924. 2017. View Article : Google Scholar : PubMed/NCBI

207 

Han L, Hiratake J, Kamiyama A and Sakata K: Design, synthesis, and evaluation of gamma-phosphono diester analogues of glutamate as highly potent inhibitors and active site probes of gamma-glutamyl transpeptidase. Biochemistry. 46:1432–1447. 2007. View Article : Google Scholar : PubMed/NCBI

208 

King JB, West MB, Cook PF and Hanigan MH: A novel, species-specific class of uncompetitive inhibitors of gamma-glutamyl transpeptidase. J Biol Chem. 284:9059–9065. 2009. View Article : Google Scholar : PubMed/NCBI

209 

Zhai X, Dai T, Chi Z, Zhao Z, Wu G, Yang S and Dong D: Naringin alleviates acetaminophen-induced acute liver injury by activating Nrf2 via CHAC2 upregulation. Environ Toxicol. 37:1332–1342. 2022. View Article : Google Scholar : PubMed/NCBI

Related Articles

Journal Cover

July-2024
Volume 65 Issue 1

Print ISSN: 1019-6439
Online ISSN:1791-2423

Sign up for eToc alerts

Recommend to Library

Copy and paste a formatted citation
x
Spandidos Publications style
Zhang T, Yao C, Zhou X, Liu S, Qi L, Zhu S, Zhao C, Hu D and Shen W: Glutathione‑degrading enzymes in the complex landscape of tumors (Review). Int J Oncol 65: 72, 2024
APA
Zhang, T., Yao, C., Zhou, X., Liu, S., Qi, L., Zhu, S. ... Shen, W. (2024). Glutathione‑degrading enzymes in the complex landscape of tumors (Review). International Journal of Oncology, 65, 72. https://doi.org/10.3892/ijo.2024.5660
MLA
Zhang, T., Yao, C., Zhou, X., Liu, S., Qi, L., Zhu, S., Zhao, C., Hu, D., Shen, W."Glutathione‑degrading enzymes in the complex landscape of tumors (Review)". International Journal of Oncology 65.1 (2024): 72.
Chicago
Zhang, T., Yao, C., Zhou, X., Liu, S., Qi, L., Zhu, S., Zhao, C., Hu, D., Shen, W."Glutathione‑degrading enzymes in the complex landscape of tumors (Review)". International Journal of Oncology 65, no. 1 (2024): 72. https://doi.org/10.3892/ijo.2024.5660