Abstract
sulfur-containing amino acids have been reported to patriciate in gene regulation, DNA methylation, protein synthesis and other physiological or pathological processes. In recent years, metabolism-related molecules of sulfur-containing amino acids affecting the occurrence, development and treatment of tumors have been implicated in various disorders, especially in leukemia. Here, we summarize current knowledge on the sulfur-containing amino acid metabolism pathway in leukemia and examine ongoing efforts to target this pathway, including treatment strategies targeting (a) sulfur-containing amino acids, (b) metabolites of sulfur-containing amino acids, and (c) enzymes and cofactors related to sulfur-containing amino acid metabolism in leukemia. Future leukemia therapy will likely involve innovative strategies targeting the sulfur-containing amino acid metabolism pathway.
Keywords: Sulfur-containing amino acids, Metabolites, Methylation, Trans-sulfuration, Leukemia
Introduction
Hematopoiesis is the process that involves the development and maturation of all types of hematopoietic cells. All blood cell lineages originate from hematopoietic stem cells. Based on the morphological and functional characteristics of hematopoietic cells, the hematopoietic process is typically divided into three stages: hematopoietic stem cells, lineage-committed progenitor cells, and morphologically recognizable precursor cells. Progenitor cells committed to the erythroid lineage are termed erythroid progenitor cells, while those committed to the granulocyte-monocyte lineage are called colony-forming unit-granulocyte-macrophage (CFU-GM), megakaryocytic progenitor cells are termed colony-forming units-megakaryocyte (CFU-MK), and progenitor cells of the lymphoid lineage are known as lymphoid progenitor cells. Morphologically identifiable immature cells of various lineages, exhibiting specific morphological characteristics, are termed precursor cells. These precursor cells undergo further differentiation and maturation into distinct types of terminally differentiated blood cells with specialized functions, which are subsequently released into the circulation (Fig. 1).
Fig. 1.
Process of hematopoietic stem cell differentiation. The figure illustrates the differentiation of hematopoietic stem cells from multipotent progenitor cells through committed progenitor cells and precursor cells to mature blood cells
Leukemia is a type of malignant clonal disorder characterized by disrupted hematopoietic homeostasis, abnormal differentiation and the excessive accumulation of leukemic blasts (Culp-Hill et al. 2021; Kuek et al. 2021). The production of blood cell is inhibited by these leukemic blasts in the bone marrow and other related organs (Culp-Hill et al. 2021; Kuek et al. 2021). Numerous studies have demonstrated that amino acids metabolism plays a critical role in the onset, progression, and prognosis of leukemia. Specifically, the metabolism of sulfur-containing amino acids is intricately linked to the pathology of leukemia and its targeted therapies (Sarno et al. 2020).
Sulfur-containing amino acids and their metabolites
Elemental sulfur is present in various essential bioorganic and inorganic small molecules. Sulfur can donate or accept electrons and is the atom most commonly involved in altering the oxidation state of oxygen atoms in cells. Sulfur-containing amino acids, such as methionine, homocysteine, cysteine, and taurine, are crucial molecules required by the human body (Brosnan and Brosnan 2006). Methionine, an essential amino acid, undergoes a series of metabolic steps to form homocysteine, cysteine and taurine (Stipanuk 2004). The metabolic pathway of sulfur-containing amino acids is depicted in Fig. 2.
Fig. 2.
Summary of sulfur-containing amino acid metabolism. The figure explains the sulfur-containing amino acid metabolic pathway and its associated enzymes. MAT: Methionine adenosyltransferase; MT: Methyltransferase; SAHH: S-adenosylhomocysteine hydrolase; CBS: Cystathionine β-synthase; CSE: Cystathionine-γ-lyase; GCS: γ-glutamylcysteine synthase; GSS: Glutathione synthetase; MS: Methionine synthase; BHMT: Betaine homocysteine methyltransferase; CH3: Methyl; VitB12: vVitaminB12; THF: Tetrahydrofolate; 5-CH3-THF: 5-methyltetrahydrofolate; CSD: Cysteine sulfonate decarboxylase; HD: Hypotaurine dehydrogenase; H2S: Hydrogen sulfide; DHF: Dihydrofolate; MTHFR: Methylenetetrahydrofolate reductase; DHFR: Dihydrofolate reductase
Methionine plays a crucial role in metabolism as an active methyl donor in the human body. Methionine is converted to S-adenosylmethionine (SAM) by methionine adenosyltransferase (MAT), which is subsequently demethylated by methyltransferase (MT) to produce S-adenosylhomocysteine (Portillo et al. 2020). S-adenosylhomocysteine is then hydrolyzed to form homocysteine by S-adenosylhomocysteine hydrolase (SAHH) (Brosnan and Brosnan 2006). The conversion of homocysteine to methionine, known as remethylation, occurs primarily through two pathways. One pathway involves betaine homocysteine methyltransferase (BHMT), which utilizes a methyl group from betaine (Brosnan and Brosnan 2006; Portillo et al. 2020). Betaine is primarily derived from choline. Choline is synthesized from phosphatidylcholine, a product of phosphatidylethanolamine methylation by SAM (Blachier et al. 2020). The other pathway involves 5-methyltetrahydrofolate (5-CH3-THF) and methionine synthase (MS), with vitamin B12 (VitB12) as a coenzyme, producing tetrahydrofolate (THF) (Blachier et al. 2020). Intracellular folic acid is converted to THF from dihydrofolate (DHF) by dihydrofolate reductase (DHFR) (Oosterom et al. 2019). THF either enters the 5-CH3-THF pool for the methylation cycle via methylenetetrahydrofolate reductase (MTHFR) or is utilized in the purine cycle for DNA/RNA biosynthesis (Oosterom et al. 2019). Cystathionine-β-synthase (CBS) and cystathionine γ-lyase (CSE) are responsible for synthesizing cysteine from homocysteine in the transsulfuration pathway (Townsend et al. 2004; Wang et al. 2022). Additionally, cysteine can be used to synthesize other sulfur-containing amino acids and bioactive compounds, such as glutathione (GSH), hydrogen sulfide (H2S) and cysteine sulfinic acid (Blachier et al. 2020). Cysteine forms GSH through γ-glutamylcysteine synthase (GCS) and glutathione synthetase (GSS), utilizing glutamate and glycine, following the intermediate formation of γ-glutamylcysteine (Blachier et al. 2020; Wu et al. 2004). Cysteine sulfinic acid utimately produces taurine via cysteine sulfinic acid decarboxylase (CSD) and hypotaurine dehydrogenase (HD) in two steps (Blachier et al. 2020; Duszka 2022; Ramírez-Guerrero et al. 2022).
Methionine and leukemia
The metabolism of amino acids is essential for the survival of leukemia cells. A screen for amino acid deprivation in primary leukemia stem cells and progenitor cells revealed significant dependence on methionine (Cunningham et al. 2022). The research found that methionine is primarily utilized in protein translation and for the provision of methyl groups to histones via SAM. Methionine depletion significantly impacts H3K36me3, and is accompanied by reduced RNA levels, enhanced apoptosis, and cell cycle arrest. However, methionine depletion did not affect the redox balance control in acute myeloid leukemia (AML) (Cunningham et al. 2022). Disruption of methionine/SAM metabolism either by methionine deprivation or by pharmacological inhibition of downstream pathways reduces the overall cellular methylation potential, decreases relative cell numbers, and selectively induces apoptosis in established mixed lineage leukemia (MLL)-AF4 or MLL-AF6- expressing patient blasts (Barve et al. 2019). In a leukemia mouse model, administration of L-methionine significantly increased homocysteine levels, decreased tissue plasminogen activator (tPA) levels, reversed hyperfibrinolysis, and ultimately reduced hemorrhagic symptoms in acute promyelocytic leukemia (APL) (Jácomo et al. 2012). Additionally, methionine dependence was also experimentally verified in HL-60 and Jurkat acute leukemia cells. The proliferation, cell cycle, and apoptosis of HL-60 and Jurkat cells were markedly affected by methionine restriction (He et al. 2023).
SAM and leukemia
SAM, a universal methyl donor in human cells, is synthesized from methionine by MAT. Two distinct isoforms of MAT are encoded by the genes methionine adenosyltransferase 1 A (MAT1A) and methionine adenosyltransferase 2 A (MAT2A), with MAT1A expressed in the liver and MAT2A expressed in extrahepatic tissues (Nordgren et al. 2011) The use of the MAT2A inhibitor PF-9366 and the knockdown of MAT2A via small interfering RNA (siRNA) in a human CRISPR/Cas9-MLL-rearranged (CRISPR/Cas9-MLLr) leukemia model resulted in alterations to several cellular processes, including proliferation, viability, differentiation, apoptosis, the cell cycle, and histone methylation (Secker et al. 2020). These findings provide corroborating evidence for the hypothesis that targeted therapies against the SAM-dependent methyltransferases of the MLL protein may be a viable therapeutic approach for acute leukemia with the MLL fusion gene or MLL leukemia (Chern et al. 2020). Arsenic trioxide (As2O3, also known as ATO) was initially employed in the clinical management of APL by Chinese researchers in the 20th century (Chen and Ding 2024; Zhang et al. 2018). Subsequently, it has been established as a primary therapeutic option for APL, as indicated by numerous clinical guidelines for leukemia (Chen and Ding 2024). However, As2O3 may alter the levels of S-adenosylhomocysteine but not SAM in the treatment of APL (Khaleghian et al. 2014).
Homocysteine and leukemia
The homocysteine-treated human acute monocytic leukemia cell line (THP-1) exhibited inhibition of autophagosome formation via the AMPK-mTOR-TFEB signaling pathway (Yang et al. 2022). Elevated plasma homocysteine levels are also used to assess the risk of toxicity associated with methotrexate (MTX), whether used alone or in combination, in acute lymphoblastic leukemia and Burkitt lymphoma (Kubota et al. 2014; Pinnix et al. 2017; Zahra et al. 2020). Furthermore, melatonin treatment has been shown to mitigate MTX toxicity, as evidenced by reduced homocysteine level (Chen et al. 2020).
Cysteine and leukemia
Intracellular cysteine comes from two pathways, one involves conversion from intracellular methionine, and the other involves uptake via the cystine-glutamate antiporter known as system xc− and the alanine/serine/cysteine/threonine (ASCT) neutral amino acid transporter encoded by SLC1A4 (Cunningham et al. 2024; Liu et al. 2020; Ma et al. 2023). The ASCT transporter imports cysteine from the extracellular environment, whereas this is not the primary pathway for intracellular cysteine due to its chemical instability in the extracellular environment (Fu et al. 2019; Liu et al. 2020; Yin et al. 2016). The system xc− cystine/glutamate exchange antiporter is a heterodimer consisting of a light chain subunit (xCT or SLC7A11) and a heavy chain subunit (CD98hc or SLC3A2), which imports cystine and exports glutamate (Liu et al. 2020). Subsequently, cystine is reduced to cysteine for the formation of GSH (Liu et al. 2020). Literature reported that cysteine cannot be synthesized from methionine in human lymphocytes and leukemia cells and are therefore highly dependent on the uptake pathway from the microenvironment (Liu et al. 2020; Ma et al. 2023). As a result, cystine import is the primary source of intracellular cysteine, particularly for hematopoietic tumor cells (Liu et al. 2020; Ma et al. 2023).
Blockade of the system xc−-GSH axis has proven remarkably effective against a wide range of tumors, including hematopoietic malignancies (Liu et al. 2020; Ma et al. 2023; Pardieu et al. 2022; Zhang et al. 2012). SLC3A2 is implicated in the glutamate dehydrogenase 1 (GDH1)-dependent progression of AML cells (Ma et al. 2023). When GDH1 is downregulated, intracellular cysteine is diminished due to disruption of the cystine-glutamate transporter resulting from decreased SLC3A2 expression. This exacerbates GSH depletion and disrupts redox homeostasis, ultimately leading to ferroptosis in AML cells (Ma et al. 2023).
Targeting key amino acid metabolic pathways through matrix-leukemia interactions is an effective means for treating residual leukemia cells (Boutter et al. 2014; Tabe et al. 2019). One such phenomenon is the dependence of chronic lymphocytic leukemia (CLL) cells on extracellular cysteine for survival. The study by Zhang et al. illustrates that patients’ CLL cells lack xCT expression, restricting the inward flow of cystine. Coincidentally, however, bone marrow stromal cells expressing high levels of xCT can efficiently utilize the cystine antiporter to intake cystine and synthesize cysteine within themselves. Cysteine is then released into the microenvironment and taken up by CLL cells leading to increased GSH synthesis, which enhances the survival of the leukemia cells and protects them from drug-induced cytotoxicity (Zhang et al. 2012). Furthermore, inhibition of SLC7A11 affects the function of AML cell lines in a cysteine-dependent manner (Pardieu et al. 2022).
Leukemia stem cells (LSCs) primarily dependent on cysteine to sustain energy metabolism for survival (Jones et al. 2019). Cysteine deficiency impairs glutathionylation of succinate dehydrogenase A (SDHA), thereby impairing the activity of the electron transport chain complex II (ETC II), inhibiting oxidative phosphorylation, reducing ATP levels, and leading to LSC death (Jones et al. 2019). These results have been confirmed by studies showing that cysteine depletion induces cell death in chronic myeloid leukemia (CML) and AML through ferroptosis, involving thioredoxin reductase 1 (TXNRD1) and glutathione peroxidase 4 (GPX4) (Cunningham et al. 2024; Liu et al. 2021). An encouraging study has developed an AML treatment using GSH-responsive cysteine polymer-based ferroptosis-inducing nanomedicine (GCFN), which acts as an efficient ferroptosis inducer and chemotherapeutic drug nanocarrier. GCFN not only depletes intracellular GSH and GSH-dependent GPX4, inducing ferroptosis in AML cells through lipid peroxidation, but also prolongs the lifespan of mice in leukemia models with minimal side effects (Yu et al. 2023). These results made it clear that cysteine has the potential to serve as a therapeutic strategy for combating leukemia.
GSH and leukemia
GSH is a major antioxidant in cells. Multidrug-resistant (MDR) K562/adriamycin (ADM) leukemia cells have higher GSH levels and iron-regulatory protein 2 (IRP2), transferrin receptor, ferritin heavy chain 1 (FTH1), and GPX4 expression, which may enhance their antioxidantcapacity and protect K562/ADM cells from ferroptosis (Zhang et al. 2022). Another study confirmed the drug-resistant effect of GSH, showing that GSH levels are associated with MTX resistance. The authors identified two genes involved in GSH metabolism, gamma-glutamyltransferase 1 (GGT1) and thioredoxin reductase 3 (TXNRD3), as contributors to MTX resistance (Canevarolo et al. 2022).
In the study of the effect of compound 9b (2-chloro-3-ethyl-5,6,7-trimethoxy-1,4-naphthoquinone) on HL-60 leukemia cells, it was found that compound 9b induced mitochondrial DNA damage, cell cycle arrest, and ROS generation in HL-60 cells thereby resisting leukemia, with GSH playing a crucial role in the anti-leukemia mechanism of compound 9b (Li et al. 2018). In addition, elevated GSH levels were detected in the plasma of patients with primary or relapsed acute lymphoblastic leukemia (ALL) (Mahmoud et al. 2017).
AML is a highly aggressive malignancy characterized by oxidative stress resulting in abnormal levels of reduced and oxidized GSH (Abbas et al. 2023). The ratio of GSH to oxidized glutathione (GSSG) is a key indicator of cellular oxidative stress. The real-time, dynamic and highly sensitive detection of GSH/GSSG in AML cells is crucial for the clinical diagnosis and treatment of leukemia. Additionally, researchers have innovatively developed subcellular compartment-specific sensors to monitor GSH/GSSG and combined with high-resolution fluorescence microscopy to provide insights into the level of GSH/GSSG in cytoplasm, mitochondria, nucleus, and endoplasmic reticulum, revealing substantial heterogeneity in the dynamics of GSH/GSSG levels in different subcellular compartments (Abbas et al. 2023). Furthermore, drugs such as cytarabine and doxorubicin can alter GSH/GSSG levels in different subcellular compartments. The authors demonstrated, using live cell imaging, that different cellular compartments responded differently to drugs such as CB-839, parthenolide (PTL) and piperlongumine (PLM) (Abbas et al. 2023). This research will provide insights for AML treatment.
GSS and leukemia
GSS is an important enzyme in the methionine metabolic pathway and in GSH synthesis. Cholez et al. reported that the downregulation of STAT5A, an important transcription factor, through stable transfection, siRNA, or TAT-fusion proteins, increased spontaneous cell death and Fas-promoted apoptosis in the leukemic pre-B cell lines NALM6 and Reh (Cholez et al. 2012). The targets of STAT5A might include the GSS, as that there are several binding sites in the promoter of the GSS for STAT5 (Lee et al. 2005). GSS was significantly downregulated following the decreased expression of STAT5A, leading to a decline in the protective effect against ROS, which might regulate apoptosis in NALM6 and Reh cells (Cholez et al. 2012).
In addition, GSS is involved in the mechanism of action of anti-tumor drugs that kill leukemia cells. Hamdoun et al. have shown that the anthelmintic niclosamide is more lethal to three hematopoietic tumor cell lines—CCRF-CEM, CEM/ADR5000 leukemia, and RPMI-8226 multiple myeloma—than to some solid cancer cell lines Hamdoun et al. 2017). GSS, a key molecule in the pathway by which niclosamide exerts its inhibitory effect on leukemia cell growth, may play a role in inhibiting leukemia cell growth by promoting ROS accumulation in leukemia cells ( Hamdoun et al. 2017). GSS catalyzes the biosynthesis of GSH from γ-glutamylcysteine and glycine in an ATP-dependent manner (Okada and Kimura 2022). Niclosamide, having a higher affinity for GSS due to its lower binding energy, competitively binds to GSS in leukemia cells, reducing the binding of GSS to ATP, thereby decreasing GSH synthesis and promoting ROS accumulation (Hamdoun et al. 2017).
Taurine and leukemia
Taurine is nonproteinogenic amino acid generated from cysteine. In the present study, Zhou et al. used gas chromatography time-of-flight mass spectrometry of plasma and bone marrow supernatants to analyze the metabolic characteristics of patients with AML with maturation (AML-M2) (Zhou et al. 2020). The study showed that taurine not only had a higher plasma abundance in AML-M2 patients but also that the plasma level of taurine was positively associated with AML-M2 risk status (Zhou et al. 2020). However, the expression level of taurine transporter was negatively correlated with AML-M2 malignancy (Zhou et al. 2020).These results suggest that the plasma abundance of taurine has the potential to serve as a prognostic marker for patients with AML-M2.
In addition, patients with ALL often experience side effects during chemotherapy. The use of taurine increases white blood cell counts during chemotherapy in patients with ALL, resulting in a decrease in febrile episodes (Islambulchilar et al. 2015b). Furthermore, taurine supplementation attenuated chemotherapy-induced adverse effects, such as nausea and vomiting, and improved liver and kidney functions. Moreover, taurine significantly reduced serum malondialdehyde and superoxide dismutase levels (Islambulchilar et al. 2015a, c).
These valuable results suggest that taurine is not only a biomarker for malignant progression, but its application also has the potential to mitigate toxic side effects due to its antioxidant capacity in leukemia.
H2S and leukemia
In mammals, H2S is primarily produced through the metabolism of sulfur-containing amino acids. CBS and CSE are two enzymes that catabolize L-cysteine to produce H2S, constituting the main pathway for H2S production in vivo (Fig. 2). Wang et al. reported that CBS and H2S levels were elevated in bone marrow mononuclear cells and K562 cells from pediatric CML patients. Inhibition of CBS reduced the malignancy of CML, accompanied by alterations in bone marrow mononuclear cells, apoptosis, cell-cycle progression, and migration in K562 cells and tumor xenografts. Endogenous H2S levels were reduced following shRNA knockdown of CBS and inhibition of CBS activity with aminooxyacetic acid (AOAA), leading to mitochondria-associated apoptosis and suppression of NF-κB-mediated gene expression (Wang et al. 2021). This study suggests that H2S and CBS could be potential targets for the treatment of CML.
Folate, THF, 5-CH3-THF and leukemia
Intracellular folate levels can be categorized into two THF pools: (1) for the methylation cycle, and (2) for the purine cycle involved in DNA/RNA biosynthesis (Oosterom et al. 2019; Zarou et al. 2021). Numerous reports on hematological malignancies have demonstrated alterations in the folate metabolism pathway in leukemia cells (Köse et al. 2021; Mahmood and Emadi 2021; Metayer et al. 2016, 2023). A significant milestone in the therapeutic approach to childhood leukemia was the introduction of antifolate drugs by Sidney Farber over 70 years ago (Stine et al. 2022). MTX is a crucial antifolate used in the treatment of pediatric ALL, primarily by inhibiting the activities of several folate-associated enzymes (Winter et al. 2018; Xu et al. 2022; Yousef et al. 2019). The levels of 5-CH3-THF and THF reflect the efficacy and side effects of MTX, either alone or in combination, in the treatment of ALL, as the quantities of these folates determine overall folate levels (Oosterom et al. 2019).
DHFR and leukemia
The expression level of DHFR in leukemia cell lines (Jurkat, Nalm-6, Reh, K562, and Molt-4) was significantly higher compared to normal hematopoietic cells. Therefore, DHFR and its gene amplification are potential targets for anti-leukemia drugs (Eyilcim et al. 2023; Gervasini et al. 2017; Kim et al. 2021; Kodidela et al. 2015; Milosevic et al. 2019; Oiwa et al. 2021; Wińska et al. 2019).
In a separate study, DHFR expression was measured in 96 children with ALL and 100 control individuals. The results indicated that DHFR mRNA expression was significantly higher in children with ALL compared to the controls (P < 0.001). Furthermore, DHFR mRNA levels were elevated in patients with B-cell lineage ALL compared to those with T-cell lineage ALL (P < 0.05). Survival analysis revealed that elevated DHFR mRNA levels were negatively correlated with overall survival in children with ALL (Organista-Nava et al. 2018).
MTHFR and leukemia
MTHFR is a crucial enzyme influencing the pharmacokinetics of anti-leukemia drugs, such as MTX (Umerez et al. 2017). A meta-analysis including 26 studies involving 4,682 children with ALL and 7144 controls showed that the C677T polymorphism of MTHFR was associated with ALL in children, especially in Asian populations (Li et al. 2020). This conclusion was confirmed by a report by Frikha et al. (Frikha et al. 2021). Another systematic review and meta-analysis on patients with CLL found that the MTHFR polymorphism A1298C, but not C677T, might be related to the tumorigenesis of CLL under an allelic model (Raoufi et al. 2021). The authors suggested that other factors such as age, gender, ethnicity, gene-gene interactions and environmental conditions need to be considered to clarify the true association of MTHFR polymorphisms with CLL, and further experiments are necessary (Raoufi et al. 2021). Interestingly, a study suggested that the MTHFR 677T-1298 C haplotype is common in ALL and might be a valuable biomarker indicating high-dose MTX chemotherapy-related adverse effects (Frikha et al. 2022). Collectively, these studies indicate that genetic polymorphisms in MTHFR are widely associated with the occurrence and development of leukemia (Aly et al. 2014; Bănescu et al. 2015; Cwiklinska et al. 2020; Gutiérrez-Álvarez et al. 2016; Huang et al. 2015; Sazawal et al. 2014).
VitB12 and leukemia
VitB12 is a crucial cofactor in the methionine cycle. VitB12 deficiency is a significant reversible cause of myelosuppression, megaloblastic anemia, poor growth, and increased susceptibility to infections (Kitaz et al. 2022; Konda et al. 2019; Sharma et al. 2021; Sutton and Mba 2017). Furthermore, VitB12 deficiency was significantly associated with toxic deaths in ALL (Tandon et al. 2015). Sharma et al. (Sharma et al. 2021) reported a case of a 14-month-old child with VitB12 deficiency presenting with symptoms suggestive of acute leukemia, including pancytopenia and severe anemia, and 19% reactive/atypical cells in the peripheral blood. After treatment with VitB12, the symptoms of acute leukemia improved dramatically (Sharma et al. 2021).
BHMT and leukemia
Utilizing Tetra-primer Amplification Refractory Mutation System PCR (T-ARMS-PCR) and DNA sequencing, Bellampalli et al. (Bellampalli et al. 2017) evaluated the association of BHMT with ALL (Bellampalli et al. 2017). The results showed that although 18 nonsynonymous single nucleotide polymorphisms (nsSNPs) were deleterious, rs59109725, rs116634518, and rs138578732 in 3’-UTR altered the miRNA-binding site, and 11 copy number variations (CNVs) were present in the BHMT gene. Consequently, BHMT polymorphism resulted in alterations in free energy. Unfortunately, the association of BHMT polymorphism with both childhood and adult ALL was not significant (Bellampalli et al. 2017). The study noted that further evaluation with larger sample sizes, and exploration of the effects of other SNPs, CNVs, and miRNAs, are needed to fully understand the role of the BHMT gene in the pathogenesis of ALL (Bellampalli et al. 2017).
Conclusion
The occurrence, progression, and therapeutic targets of leukemia are influenced by multiple factors. This review uniquely focuses on the impact of sulfur-containing amino acid metabolism on leukemia development and associated therapeutic targets.
Although there remain many uncertainties regarding the detailed mechanisms of sulfur-containing amino acids and their metabolites in leukemia, therapies targeting the metabolism of these amino acids hold significant potential for improving leukemia patient survival.
Abbreviations
- MAT
Methionine adenosyltransferase
- MT
Methyltransferase
- SAHH
S-adenosylhomocysteine hydrolase
- CBS
Cystathionine β-synthase
- CSE
Cystathionine-γ-lyase
- GCS
γ-glutamylcysteine synthetase
- GSS
Glutathione synthetase
- MS
Methionine synthase
- BHMT
Betaine homocysteine methyltransferase
- CH3
Methyl
- VitB12
VitaminB12
- THF
Tetrahydrofolate
- 5-CH3-THF
5-methyltetrahydrofolate
- CSD
Cysteine sulfonate decarboxylase
- HD
Hypotaurine dehydrogenase
- H2S
Hydrogen sulfide
- DHF
Dihydrofolate
- MTHFR
Methylenetetrahydrofolate reductase
- DHFR
Dihydrofolate reductase
- SAM
S-adenosylmethionine
- GSH
Glutathione
- CSAD
Cysteine sulfinic acid decarboxylase
- HD
Hypotaurine dehydrogenase
- AML
Acute myeloid leukemia
- MLL
Mixed lineage leukemia
- tPA
Tissue plasminogen activator
- APL
Acute promyelocytic leukemia
- MAT1A
Methionine adenosyltransferase 1 A
- MAT2A
Methionine adenosyltransferase 2 A
- siRNA
Small interfering RNA
- As2O3
Arsenic trioxide
- THP-1
Acute monocytic leukemia cell line
- ASCT
Alanine/serine/cysteine/threonine
- GDH1
Glutamate dehydrogenase 1
- CLL
Chronic lymphocytic leukemia
- LSCs
Leukemia stem cells
- SDHA
Succinate dehydrogenase A
- ETC II
Electron transport chain complex II
- TXNRD1
Thioredoxin reductase 1
- GPX4
Glutathione peroxidase 4
- MDR
Multidrug-resistant
- ADM
Adriamycin
- IRP2
Iron-regulatory protein 2
- FTH1
Ferritin heavy chain 1
- MTX
Methotrexate
- GGT1
Gamma-glutamyltransferase 1
- TXNRD3
Thioredoxin reductase 3
- ROS
Reactive oxygen species
- PTL
Parthenolide
- PLM
Piperlongumine
- AOAA
Aminooxyacetic acid
- nsSNPs
Nonsynonymous single nucleotide polymorphisms
- CNVs
Copy number variations
Author contributions
Xiaoyan Chen, Yanfei Li: had the idea for the article. Xiaoyan Chen, Jiahui Jin and Yanfei Li: performed the literature search, data analysis and drafted paper. Rui Chang, Xing Yang, Na Li, Xi Zhu, Linlin Ma: proof final content. All authors have read and approved the final manuscript.
Funding
This work was supported by Biomedical Technology Support Special Project of “Shanghai Science and Technology Innovation Action Plan” [21S11901700], Shanghai Natural Science Foundation [21ZR1428400], Shanghai Pujiang Program [2021PJD027], and Pudong New District Academic Leaders Programme [PWRd2021-17].
Data availability
No datasets were generated or analysed during the current study.
Declarations
Competing interests
The authors have no relevant financial or non-financial interests to disclose.
Footnotes
Publisher’s Note
Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional affiliations.
Xiaoyan Chen and Jiahui Jin contributed equally to this work.
References
- Abbas G, Cui MM, Wang DB et al (2023) Construction of genetically encoded biosensors to monitor subcellular compartment-specific glutathione response to chemotherapeutic drugs in acute myeloid leukemia cells. Anal Chem 95(5):2838–2847. 10.1021/acs.analchem.2c04255 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Aly RM, Taalab MM, Ghazy HF (2014) MTHFR A1298C and C677T gene polymorphisms and susceptibility to chronic myeloid leukemia in Egypt. Int J Clin Exp Pathol 7(5):2571–2578 [PMC free article] [PubMed] [Google Scholar]
- Bănescu C, Iancu M, Trifa AP et al (2015) The methylenetetrahydrofolate reductase (MTHFR) 677 C > T polymorphism increases the risk of developing chronic myeloid leukemia-a case-control study. Tumour Biol 36(4):3101–3107. 10.1007/s13277-014-2946-1 [DOI] [PubMed] [Google Scholar]
- Barve A, Vega A, Shah PP et al (2019) Perturbation of methionine/S-adenosylmethionine metabolism as a novel vulnerability in MLL rearranged leukemia. Cells 8(11):1322. 10.3390/cells8111322 [DOI] [PMC free article] [PubMed]
- Bellampalli R, Vohra M, Sharma K et al (2017) Acute lymphoblastic leukemia and genetic variations in BHMT gene: case-control study and computational characterization. Cancer Biomark 19(4):393–401. 10.3233/CBM-160186 [DOI] [PubMed] [Google Scholar]
- Blachier F, Andriamihaja M, Blais A (2020) Sulfur-containing amino acids and lipid metabolism. J Nutr 150(Suppl 1):2524S–2531S. 10.1093/jn/nxaa243 [DOI] [PubMed] [Google Scholar]
- Boutter J, Huang Y, Marovca B et al (2014) Image-based RNA interference screening reveals an individual dependence of acute lymphoblastic leukemia on stromal cysteine support. Oncotarget 5(22):11501–11512. 10.18632/oncotarget.2572 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Brosnan JT, Brosnan ME (2006) The sulfur-containing amino acids: an overview. J Nutr 136(6 Suppl):1636S–1640S. 10.1093/jn/136.6.1636S [DOI] [PubMed] [Google Scholar]
- Canevarolo RR, Melo CPS, Cury NM et al (2022) Glutathione levels are associated with methotrexate resistance in acute lymphoblastic leukemia cell lines. Front Oncol 12:1032336. 10.3389/fonc.2022.1032336 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Chen JX, Ding ZQ (2024) Natural products as potential drug treatments for acute promyelocytic leukemia. Chin Med 19(1):57. 10.1186/s13020-024-00928-8 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Chen YC, Sheen JM, Hsu MH et al (2020) Melatonin rescued methotrexate-induced spatial deficit and hyperhomocysteinemia and increased asymmetric dimethylarginine in plasma and dorsal hippocampus in developing rats. Life Sci 242:116931. 10.1016/j.lfs.2019.116931 [DOI] [PubMed] [Google Scholar]
- Chern TR, Liu L, Petrunak E et al (2020) Discovery of potent small-molecule inhibitors of MLL methyltransferase. ACS Med Chem Lett 11(6):1348–1352. 10.1021/acsmedchemlett.0c00229 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Cholez E, Debuysscher V, Bourgeais J et al (2012) Evidence for a protective role of the STAT5 transcription factor against oxidative stress in human leukemic pre-B cells. Leukemia 26(11):2390–2397. 10.1038/leu.2012.112 [DOI] [PubMed] [Google Scholar]
- Culp-Hill R, D’Alessandro A, Pietras EM (2021) Extinguishing the embers: targeting AML metabolism. Trends Mol Med 27(4):332–344. 10.1016/j.molmed.2020.10.001 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Cunningham A, Erdem A, Alshamleh I et al (2022) Dietary methionine starvation impairs acute myeloid leukemia progression. Blood 140(19):2037–2052. 10.1182/blood.2022017575 [DOI] [PubMed] [Google Scholar]
- Cunningham A, Oudejans LL, Geugien M et al (2024) The nonessential amino acid cysteine is required to prevent ferroptosis in acute myeloid leukemia. Blood Adv 8(1):56–69. 10.1182/bloodadvances.2023010786 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Cwiklinska M, Czogala M, Kwiecinska K et al (2020) Polymorphisms of SLC19A1 80 G > A, MTHFR 677 C > T, and tandem TS repeats influence pharmacokinetics, acute liver toxicity, and vomiting in children with acute lymphoblastic leukemia treated with high doses of methotrexate. Front Pediatr 8:307. 10.3389/fped.2020.00307 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Duszka K (2022) Versatile triad alliance: bile acid, taurine and microbiota. Cells 11(15):2337. 10.3390/cells11152337 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Eyilcim O, Gunay F, Gunkara OT et al (2023) Design and synthesis of novel 1,2,3,4-tetrazines as new anti-leukemia cancer agents. Chem Biol Drug Des 102(5):1186–1201. 10.1111/cbdd.14328 [DOI] [PubMed] [Google Scholar]
- Frikha R, Elloumi M, Rebai T et al (2022) MTHFR 677T-1298 C haplotype in acute lymphoblastic leukemia: impact on methotrexate therapy. J Oncol Pharm Pract 28(5):1031–1034. 10.1177/10781552211017193 [DOI] [PubMed] [Google Scholar]
- Frikha R, Turki F, Frikha F et al (2021) Involvement of MTHFR rs1801133 in the susceptibility of acute lymphoblastic leukemia: a preliminary study. J Pediatr Hematol Oncol 43(6):e816–e818. 10.1097/MPH.0000000000001970 [DOI] [PubMed] [Google Scholar]
- Fu XY, Cate SA, Dominguez M et al (2019) Cysteine disulfides (cys-ss-X) as sensitive plasma biomarkers of oxidative stress. Sci Rep 9(1):115. 10.1038/s41598-018-35566-2 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Gervasini G, de Murillo SG, Jiménez M et al (2017) Dihydrofolate reductase genetic polymorphisms affect methotrexate dose requirements in pediatric patients with acute lymphoblastic leukemia on maintenance therapy. J Pediatr Hematol Oncol 39(8):589–595. 10.1097/MPH.0000000000000908 [DOI] [PubMed] [Google Scholar]
- Gutiérrez-Álvarez O, Lares-Asseff I, Galaviz-Hernández C et al (2016) Involvement of MTHFR and TPMT genes in susceptibility to childhood acute lymphoblastic leukemia (ALL) in mexicans. Drug Metab Pers Ther 31(1):41–46. 10.1515/dmpt-2015-0036 [DOI] [PubMed] [Google Scholar]
- Hamdoun S, Jung P, Efferth T (2017) Drug repurposing of the anthelmintic niclosamide to treat multidrug-resistant leukemia. Front Pharmacol 8:110. 10.3389/fphar.2017.00110 [DOI] [PMC free article] [PubMed] [Google Scholar]
- He YJ, Yu SS, Zhang B et al (2023) [Effects of methionine restriction on proliferation, cell cycle, and apoptosis of human acute leukemia cells]. Zhongguo Shi Yan Xue Ye Xue Za Zhi 31(5):1290–1295. 10.19746/j.cnki.issn.1009-2137.2023.05.006 [DOI] [PubMed] [Google Scholar]
- Huang LL, Deng DH, Peng ZG et al (2015) Polymorphisms in the methylenetetrahydrofolate reductase gene (MTHFR) are associated with susceptibility to adult acute myeloid leukemia in a Chinese population. Cancer Epidemiol 39(3):328–333. 10.1016/j.canep.2015.02.012 [DOI] [PubMed] [Google Scholar]
- Islambulchilar M, Asvadi I, Sanaat Z et al (2015a) Effect of taurine on attenuating chemotherapy-induced adverse effects in acute lymphoblastic leukemia. J Cancer Res Ther 11(2):426–432. 10.4103/0973-1482.151933 [DOI] [PubMed] [Google Scholar]
- Islambulchilar M, Asvadi I, Sanaat Z et al (2015b) Effect of taurine on febrile episodes in acute lymphoblastic leukemia. Adv Pharm Bull 5(1):103–108. 10.5681/apb.2015.014 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Islambulchilar M, Asvadi I, Sanaat Z et al (2015c) Taurine attenuates chemotherapy-induced nausea and vomiting in acute lymphoblastic leukemia. Amino Acids 47(1):101–109. 10.1007/s00726-014-1840-x [DOI] [PubMed] [Google Scholar]
- Jácomo RH, Santana-Lemos BA, Lima ASG et al (2012) Methionine-induced hyperhomocysteinemia reverts fibrinolytic pathway activation in a murine model of acute promyelocytic leukemia. Blood 120(1):207–213. 10.1182/blood-2011-04-347187 [DOI] [PubMed] [Google Scholar]
- Jones CL, Stevens BM, D’Alessandro A et al (2019) Cysteine depletion targets leukemia stem cells through inhibition of electron transport complex II. Blood 134(4):389–394. 10.1182/blood.2019898114 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Khaleghian A, Ghaffari SH, Ahmadian S et al (2014) Metabolism of arsenic trioxide in acute promyelocytic leukemia cells. J Cell Biochem 115(10):1729–1739. 10.1002/jcb.24838 [DOI] [PubMed] [Google Scholar]
- Kim A, Shin JY, Seo JS (2021) Genomic and transcriptomic analyses reveal a tandem amplification unit of 11 genes and mutations in mismatch repair genes in methotrexate-resistant HT-29 cells. Exp Mol Med 53(9):1344–1355. 10.1038/s12276-021-00668-x [DOI] [PMC free article] [PubMed] [Google Scholar]
- Kitaz MN, Zazo R, Alsabbagh M et al (2022) A rare combination of acute myeloid leukemia with vit B12 deficiency: case report. Ann Med Surg (Lond) 81:104500. 10.1016/j.amsu.2022.104500 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Kodidela S, Pradhan SC, Dubashi B et al (2015) Influence of dihydrofolate reductase gene polymorphisms rs408626 (-317A > G) and rs442767 (-680C > A) on the outcome of methotrexate-based maintenance therapy in south Indian patients with acute lymphoblastic leukemia. Eur J Clin Pharmacol 71(11):1349–1358. 10.1007/s00228-015-1930-z [DOI] [PubMed] [Google Scholar]
- Konda M, Godbole A, Pandey S et al (2019) Vitamin B12 deficiency mimicking acute leukemia. Proc (Bayl Univ Med Cent) 32(4):589–592. 10.1080/08998280.2019.1641045 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Köse V, Bilir ÖA, Kara A et al (2021) The impact of pretreatment serum cobalamin and folate levels on complications and peripheral blood recovery during induction chemotherapy of leukemia: a cross-sectional study. Support Care Cancer 29(4):2225–2230. 10.1007/s00520-020-05688-3 [DOI] [PubMed] [Google Scholar]
- Kubota M, Nakata R, Adachi S et al (2014) Plasma homocysteine, methionine and S-adenosylhomocysteine levels following high-dose methotrexate treatment in pediatric patients with acute lymphoblastic leukemia or Burkitt lymphoma: association with hepatotoxicity. Leuk Lymphoma 55(7):1591–1595. 10.3109/10428194.2013.850684 [DOI] [PubMed] [Google Scholar]
- Kuek V, Hughes AM, Kotecha RS et al (2021) Therapeutic targeting of the leukaemia microenvironment. Int J Mol Sci 22(13):6888. 10.3390/ijms22136888 [DOI] [PMC free article] [PubMed]
- Lee TD, Yang HP, Whang J et al (2005) Cloning and characterization of the human glutathione synthetase 5’-flanking region. Biochem J 390(Pt 2):521–528. 10.1042/BJ20050439 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Li K, Yang K, Zheng LF et al (2018) Anti-acute myeloid leukemia activity of 2-chloro-3-alkyl-1,4-naphthoquinone derivatives through inducing mtDNA damage and GSH depletion. Bioorg Med Chem 26(14):4191–4200. 10.1016/j.bmc.2018.07.010 [DOI] [PubMed] [Google Scholar]
- Li Y, Pei YX, Wang LN et al (2020) MTHFR-C677T gene polymorphism and susceptibility to acute lymphoblastic leukemia in children: a meta-analysis. Crit Rev Eukaryot Gene Expr 30(2):125–136. 10.1615/CritRevEukaryotGeneExpr.2020033468 [DOI] [PubMed] [Google Scholar]
- Liu JY, Xia XJ, Huang P (2020) xCT: a critical molecule that links cancer metabolism to redox signaling. Mol Ther 28(11):2358–2366. 10.1016/j.ymthe.2020.08.021 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Liu SH, Wu W, Chen QQ et al (2021) TXNRD1: a key regulator involved in the ferroptosis of CML cells induced by cysteine depletion in vitro. Oxid Med Cell Longev 2021:7674565. 10.1155/2021/7674565 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Ma ZX, Ye WL, Wang JH et al (2023) Glutamate dehydrogenase 1: a novel metabolic target in inhibiting acute myeloid leukaemia progression. Br J Haematol 202(3):566–577. 10.1111/bjh.18884 [DOI] [PubMed] [Google Scholar]
- Mahmood K, Emadi A (2021) 1-C metabolism-serine, glycine, folates-in acute myeloid leukemia. Pharmaceuticals (Basel) 14(3):190. 10.3390/ph14030190 [DOI] [PMC free article] [PubMed]
- Mahmoud LB, Mdhaffar M, Ghozzi H et al (2017) Oxidative stress in Tunisian patients with acute lymphoblastic leukemia and its involvement in leukemic relapse. J Pediatr Hematol Oncol 39(3):e124–e130. 10.1097/MPH.0000000000000793 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Metayer C, Dahl G, Wiemels J et al (2016) Childhood leukemia: a preventable disease. Pediatrics 138(Suppl 1):S45–S55. 10.1542/peds.2015-4268H [DOI] [PMC free article] [PubMed] [Google Scholar]
- Metayer C, Imani P, Dudoit S et al (2023) One-carbon (folate) metabolism pathway at birth and risk of childhood acute lymphoblastic leukemia: a biomarker study in newborns. Cancers (Basel) 15(4):1011. 10.3390/cancers15041011 [DOI] [PMC free article] [PubMed]
- Milosevic G, Kotur N, Lazic J et al (2019) Influence of variants in folate metabolism genes on 6-mercaptopurine induced toxicity during treatment for childhood acute lymphocytic leukemia. J BUON 24(5):2075–2083 [PubMed] [Google Scholar]
- Nordgren KKS, Peng Y, Pelleymounter LL et al (2011) Methionine adenosyltransferase 2A/2B and methylation: gene sequence variation and functional genomics. Drug Metab Dispos 39(11):2135–2147. 10.1124/dmd.111.040857 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Okada M, Kimura Y (2022) Characterization of glutamate-cysteine ligase and glutathione synthetase from the δ-proteobacterium myxococcus xanthus. Proteins 90(8):1547–1560. 10.1002/prot.26333 [DOI] [PubMed] [Google Scholar]
- Oiwa K, Hosono N, Nishi R et al (2021) Characterization of newly established pralatrexate-resistant cell lines and the mechanisms of resistance. BMC Cancer 21(1):879. 10.1186/s12885-021-08607-9 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Oosterom N, de Jonge R, Smith DEC et al (2019) Changes in intracellular folate metabolism during high-dose methotrexate and leucovorin rescue therapy in children with acute lymphoblastic leukemia. PLoS ONE 14(9):e0221591. 10.1371/journal.pone.0221591 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Organista-Nava J, Gómez-Gómez Y, Illades-Aguiar B et al (2018) Overexpression of dihydrofolate reductase is a factor of poor survival in acute lymphoblastic leukemia. Oncol Lett 15(6):8405–8411. 10.3892/ol.2018.8429 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Pardieu B, Pasanisi J, Ling F et al (2022) Cystine uptake inhibition potentiates front-line therapies in acute myeloid leukemia. Leukemia 36(6):1585–1595. 10.1038/s41375-022-01573-6 [DOI] [PubMed] [Google Scholar]
- Pinnix CC, Chi L, Jabbour EJ et al (2017) Dorsal column myelopathy after intrathecal chemotherapy for leukemia. Am J Hematol 92(2):155–160. 10.1002/ajh.24611 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Portillo F, Vázquez J, Pajares MA (2020) Protein-protein interactions involving enzymes of the mammalian methionine and homocysteine metabolism. Biochimie 173:33–47. 10.1016/j.biochi.2020.02.015 [DOI] [PubMed] [Google Scholar]
- Ramírez-Guerrero S, Guardo-Maya S, Medina-Rincón GJ et al (2022) Taurine and astrocytes: a homeostatic and neuroprotective relationship. Front Mol Neurosci 15:937789. 10.3389/fnmol.2022.937789 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Raoufi A, Kelarijani BR, Ahadi HR et al (2021) Association of MTHFR C677T and A1298C polymorphisms with susceptibility to chronic lymphocytic leukemia: a systematic review and meta-analysis. Iran J Public Health 50(1):83–92. 10.18502/ijph.v50i1.5074 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Sarno F, Nebbioso A, Altucci L (2020) DOT1L: a key target in normal chromatin remodelling and in mixed-lineage leukaemia treatment. Epigenetics 15(5):439–453. 10.1080/15592294.2019.1699991 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Sazawal S, Chaubey R, Kaur P et al (2014) MTHFR gene polymorphisms and the risk of acute lymphoblastic leukemia in adults and children: a case control study in India. Indian J Hematol Blood Transfus 30(4):219–225. 10.1007/s12288-013-0295-7 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Secker KA, Bloechl B, Keppeler H et al (2020) MAT2A as key regulator and therapeutic target in MLLr leukemogenesis. Cancers (Basel) 12(5):1342. 10.3390/cancers12051342 [DOI] [PMC free article] [PubMed]
- Sharma N, Kunwar S, Shrestha AK (2021) Vitamin B12 deficiency resembling acute leukemia: a case report. JNMA J Nepal Med Assoc 59(243):1182–1184. 10.31729/jnma.6600 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Stine ZE, Schug ZT, Salvino JM et al (2022) Targeting cancer metabolism in the era of precision oncology. Nat Rev Drug Discov 21(2):141–162. 10.1038/s41573-021-00339-6 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Stipanuk MH (2004) Sulfur amino acid metabolism: pathways for production and removal of homocysteine and cysteine. Annu Rev Nutr 24:539–577. 10.1146/annurev.nutr.24.012003.132418 [DOI] [PubMed] [Google Scholar]
- Sutton L, Mba N (2017) Hematogones detected by flow cytometry in a child with vitamin B12 deficiency. Pediatr Dev Pathol 20(2):172–175. 10.1177/1093526616686241 [DOI] [PubMed] [Google Scholar]
- Tabe Y, Lorenzi PL, Konopleva M (2019) Amino acid metabolism in hematologic malignancies and the era of targeted therapy. Blood 134(13):1014–1023. 10.1182/blood.2019001034 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Tandon S, Moulik NR, Kumar A et al (2015) Effect of pre-treatment nutritional status, folate and vitamin B12 levels on induction chemotherapy in children with acute lymphoblastic leukemia. Indian Pediatr 52(5):385–389. 10.1007/s13312-015-0642-x [DOI] [PubMed] [Google Scholar]
- Townsend DM, Tew KD, Tapiero H (2004) Sulfur containing amino acids and human disease. Biomed Pharmacother 58(1):47–55. 10.1016/j.biopha.2003.11.005 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Umerez M, Gutierrez-Camino A, Muñoz-Maldonado C et al (2017) MTHFR polymorphisms in childhood acute lymphoblastic leukemia: influence on methotrexate therapy. Pharmgenomics Pers Med 10:69–78. 10.2147/PGPM.S107047 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Wang D, Yang H, Zhang Y et al (2021) Inhibition of cystathionine beta-synthase promotes apoptosis and reduces cell proliferation in chronic myeloid leukemia. Signal Transduct Target Ther 6(1):52. 10.1038/s41392-020-00410-5 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Wang R, Qi YF, Ding CH et al (2022) Sulfur-containing amino acids and their metabolites in atrial fibrosis. Front Pharmacol 13:1063241. 10.3389/fphar.2022.1063241 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Wińska P, Widło L, Skierka K et al (2019) Simultaneous inhibition of protein kinase CK2 and dihydrofolate reductase results in synergistic effect on acute lymphoblastic leukemia cells. Anticancer Res 39(7):3531–3542. 10.21873/anticanres.13499 [DOI] [PubMed] [Google Scholar]
- Winter SS, Dunsmore KP, Devidas M et al (2018) Improved survival for children and young adults with T-lineage acute lymphoblastic leukemia: results from the children’s oncology group AALL0434 methotrexate randomization. J Clin Oncol 36(29):2926–2934. 10.1200/JCO.2018.77.7250 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Wu GY, Fang YZ, Yang S et al (2004) Glutathione metabolism and its implications for health. J Nutr 134(3):489–492. 10.1093/jn/134.3.489 [DOI] [PubMed] [Google Scholar]
- Xu M, Wu S, Wang Y et al (2022) Association between high-dose methotrexate-induced toxicity and polymorphisms within methotrexate pathway genes in acute lymphoblastic leukemia. Front Pharmacol 13:1003812. 10.3389/fphar.2022.1003812 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Yang YP, Ren YG, Cai BQ et al (2022) Homocysteine suppresses autophagy through AMPK-mTOR-TFEB signaling in human THP-1 macrophages. J Cardiovasc Pharmacol 79(5):730–738. 10.1097/FJC.0000000000001232 [DOI] [PubMed] [Google Scholar]
- Yin J, Ren WK, Yang G et al (2016) L-cysteine metabolism and its nutritional implications. Mol Nutr Food Res 60(1):134–146. 10.1002/mnfr.201500031 [DOI] [PubMed] [Google Scholar]
- Yousef AM, Farhad R, Alshamaseen D et al (2019) Folate pathway genetic polymorphisms modulate methotrexate-induced toxicity in childhood acute lymphoblastic leukemia. Cancer Chemother Pharmacol 83(4):755–762. 10.1007/s00280-019-03776-8 [DOI] [PubMed] [Google Scholar]
- Yu YH, Meng YB, Xu X et al (2023) A ferroptosis-inducing and leukemic cell-targeting drug nanocarrier formed by redox-responsive cysteine polymer for acute myeloid leukemia therapy. ACS Nano 17(4):3334–3345. 10.1021/acsnano.2c06313 [DOI] [PubMed] [Google Scholar]
- Zahra FT, Nahid NA, Islam MR et al (2020) Pharmacogenetic variants in MTHFR gene are significant predictors of methotrexate toxicities in Bangladeshi patients with acute lymphoblastic leukemia. Clin Lymphoma Myeloma Leuk 20(2):e58–e65. 10.1016/j.clml.2019.11.020 [DOI] [PubMed] [Google Scholar]
- Zarou MM, Vazquez A, Helgason GV (2021) Folate metabolism: a re-emerging therapeutic target in haematological cancers. Leukemia 35(6):1539–1551. 10.1038/s41375-021-01189-2 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Zhang W, Trachootham D, Liu JY et al (2012) Stromal control of cystine metabolism promotes cancer cell survival in chronic lymphocytic leukaemia. Nat Cell Biol 14(3):276–286. 10.1038/ncb2432 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Zhang J, Zhang Y, Wang WY et al (2018) Double-sided personality: effects of arsenic trioxide on inflammation. Inflammation 41(4):1128–1134. 10.1007/s10753-018-0775-x [DOI] [PubMed] [Google Scholar]
- Zhang XY, Ai ZY, Zhang ZW et al (2022) Dihydroartemisinin triggers ferroptosis in multidrug-resistant leukemia cells. DNA Cell Biol 41(8):705–715. 10.1089/dna.2021.1145 [DOI] [PubMed] [Google Scholar]
- Zhou X, Zheng M, Wang Q et al (2020) Metabolomics analysis identifies lysine and taurine as candidate prognostic biomarkers for AML-M2 patients. Int J Hematol 111(6):761–770. 10.1007/s12185-020-02836-7 [DOI] [PubMed] [Google Scholar]
Associated Data
This section collects any data citations, data availability statements, or supplementary materials included in this article.
Data Availability Statement
No datasets were generated or analysed during the current study.