Next Article in Journal
Up-Cycling Grape Pomace through Sourdough Fermentation: Characterization of Phenolic Compounds, Antioxidant Activity, and Anti-Inflammatory Potential
Previous Article in Journal
Dietary Supplementation with a Cocoa–Carob Blend Modulates Gut Microbiota and Prevents Intestinal Oxidative Stress and Barrier Dysfunction in Zucker Diabetic Rats
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Targeting Homocysteine and Hydrogen Sulfide Balance as Future Therapeutics in Cancer Treatment

by
Avisek Majumder
Department of Medicine, University of California, San Francisco, CA 94143, USA
Antioxidants 2023, 12(8), 1520; https://doi.org/10.3390/antiox12081520
Submission received: 24 June 2023 / Revised: 24 July 2023 / Accepted: 28 July 2023 / Published: 29 July 2023
(This article belongs to the Section Health Outcomes of Antioxidants and Oxidative Stress)

Abstract

:
A high level of homocysteine (Hcy) is associated with oxidative/ER stress, apoptosis, and impairment of angiogenesis, whereas hydrogen sulfide (H2S) has been found to reverse this condition. Recent studies have shown that cancer cells need to produce a high level of endogenous H2S to maintain cell proliferation, growth, viability, and migration. However, any novel mechanism that targets this balance of Hcy and H2S production has yet to be discovered or exploited. Cells require homocysteine metabolism via the methionine cycle for nucleotide synthesis, methylation, and reductive metabolism, and this pathway supports the high proliferative rate of cancer cells. Although the methionine cycle favors cancer cells for their survival and growth, this metabolism produces a massive amount of toxic Hcy that somehow cancer cells handle very well. Recently, research showed specific pathways important for balancing the antioxidative defense through H2S production in cancer cells. This review discusses the relationship between Hcy metabolism and the antiapoptotic, antioxidative, anti-inflammatory, and angiogenic effects of H2S in different cancer types. It also summarizes the historical understanding of targeting antioxidative defense systems, angiogenesis, and other protective mechanisms of cancer cells and the role of H2S production in the genesis, progression, and metastasis of cancer. This review defines a nexus of diet and precision medicine in targeting the delicate antioxidative system of cancer and explores possible future therapeutics that could exploit the Hcy and H2S balance.

Graphical Abstract

1. Introduction

Cancer is the second leading cause of death after cardiovascular disease [1]. Current understanding characterizes cancer into six hallmarks: maintaining proliferative signaling, bypassing growth suppressors, resisting apoptosis, enabling replicative immortality, inducing angiogenesis, and initiating invasion and metastasis [2]. Due to the high proliferative rate, cancer cells depend on many nutrient sources from the diet [3]. Methionine is one of the nutrients that cancer cells require to maintain cell proliferation, growth, survival, and metastasis [4]. Methionine was the first amino acid used in protein synthesis in the eucaryotic system [5]. As an essential amino acid, methionine is not produced in our bodies, so it must be consumed from the diet [6]. Methionine is not only essential for the formation of all proteins, but it also provides lots of other metabolites that are required in multiple other metabolic processes [7]. Via the methionine cycle (Figure 1), methionine can be converted to S-Adenosyl methionine (SAM), the only methyl group donor in DNA, RNA, and histone methylation reactions. DNA, RNA, and histone methylation are dynamic; these regulate gene expression and alter cellular signaling [8]. After transferring methyl group, SAM converts to S-Adenosyl homocysteine (SAH), which then converts to homocysteine (Hcy) [7]. Hcy is a sulfur-containing nonproteinogenic amino acid; after production, half of the Hcy goes to the transsulfuration pathway to produce cysteine (a semi-essential amino acid), and another half of the Hcy can be remethylated back into methionine with the help of the folate cycle [7]. Cysteine is a semi-essential amino acid obtained from the diet or by de novo synthesis from the methionine cycle [9]. In the transsulfuration pathway, when cysteine is produced, other than helping in protein formation, it is also used for hydrogen sulfide (H2S) and glutathione (GSH) production [7]. Due to the high proliferative rate, cancer cells mainly depend on the methionine cycle for methylation reaction as well as the production of H2S and GSH [10]. A study found that cancer cells express high levels of methionine transporter SLC43A2 for the consumption of more methionine, which causes cancer progression [11]. Many studies found that a methionine restriction diet can reduce cancer risk and progression through various molecular processes [10,12,13]. A phase 1 trial also showed that it is tolerable for metastatic cancer patients to be on a methionine-restricted diet to reduce tumor growth [14].
Cancer cells depend highly on the methionine cycle, producing a massive amount of Hcy [16]. As high levels of intracellular Hcy can be secreted to blood, so many studies correlate high Hcy levels with cancer [17,18,19]. More than 15 μmol/L of Hcy in the blood is clinically termed hyperhomocysteinemia (HHcy) [7]. HHcy has been associated with multiple disease conditions, including cancer [20]. Elevated levels of Hcy are connected with oxidative stress, ER stress, apoptosis, protein oxidation, inflammation, and impaired angiogenesis [17,20]. Moreover, many previous studies found variable associations of polymorphisms of Hcy metabolism genes with cancer [21,22,23], suggesting the possible role of gene–environment interactions in the causation of cancer [24]. Some studies suggested that together with genetic polymorphisms, dietary methionine, folate, vitamin B12, B6, and alcohol consumption play an essential role in the genesis of tumors [25,26,27]. Also, different studies showed that specific genetic polymorphisms may induce risk for specific cancer types [28,29]. More studies are needed on a large number of patients in order to understand which genetic polymorphisms predispose to which types of cancer and how lifestyle modifications could be helpful in reducing cancer risk.
Cancer cells depend on the methionine cycle for their cellular turnover, producing toxic Hcy [20]. Cancer patients show high Hcy levels, but that does not mean that high Hcy is a risk factor for cancer, rather that cancer cells shuttle more Hcy to the transsulfuration pathway [20]. As high Hcy levels lead to many cellular pathogeneses, cancer cells transfer excess Hcy to the transsulfuration pathway for the production of H2S [20]. Recent studies revealed that cancer cells increased the expression of CBS (the rate-limiting enzyme in transsulfuration reaction) to reduce excess Hcy levels and produce H2S [30]. Previously, H2S was considered a toxic gas; however, recent research found that H2S has beneficial effects in reversing cellular pathophysiology [7]. H2S emerged as a third gasotransmitter after NO and CO [31], and it has been shown to have beneficial effects in reducing oxidative and ER stress, apoptosis, and inflammation, and improving neoangiogenesis [17]. Studies in colon and ovarian cancer mainly showed that this higher production of H2S induced tumor growth via inducing cell proliferation and angiogenesis [30]. Also, suppressing CBS expression led to a reduction in tumor growth [32]. This suggests that cancer cells maintain a balance of H2S and Hcy levels for their cellular growth and metastasis. Very limited studies have exploited this delicate balance of H2S and Hcy as a therapeutic opportunity for cancer treatment. Many studies showed that H2S reverses all the pathophysiological effects of Hcy [17]. Many antifolate drugs and drugs targeting Hcy metabolism have long been used to treat cancer; however, all showed limited clinical efficacy due to multiple reasons [19]. In the future, more research is needed that exploits the Hcy and H2S balance to treat cancer patients. This review article summarizes the Hcy metabolism and how Hcy metabolism and H2S production are associated with cancer. This review also discusses the current therapeutics and future therapeutic opportunities that target these pathways in cancer treatment.

2. Homocysteine Production and Hyperhomocysteinemia

As discussed in the above section, after production, half of the Hcy goes to the transsulfuration pathway, and the other half of the Hcy remethylates back to methionine with the help of the folate cycle [33,34,35]. As Hcy can be secreted into the blood, so different forms of Hcy can be found in blood circulation, as shown in Figure 2 [36]. In the transsulfuration pathway, Hcy is converted to cystathionine with the help of cystathionine β-synthase (CBS), where vitamin B6 (pyridoxine) is an essential co-factor [8]. This is the rate-limiting step of the transsulfuration pathway [8]. After production, cystathionine is further converted to cysteine by cystathionine γ-lyase (CSE), and this cysteine further produces GSH [37].
In normal conditions, cells maintain a delicate balance of Hcy production (through the methionine cycle) and elimination of Hcy (via the transsulfuration pathway) [37]. The normal range of plasma Hcy levels for young adults (~30 years) is 4.6–8.1 μM, and for older adults (30 years and above) is 4–15 μM [38]. In different disease conditions, the balance between the production and elimination of Hcy becomes affected [39,40]. High levels of Hcy in the blood circulation are called hyperhomocysteinemia (HHcy), a systemic disorder [7]. Patients with HHcy show more than 15 uM plasma Hcy [41]. HHcy has been classified as moderate (15–30 μM), intermediate (30–100 μM), and severe (>100 μM) [41]. Genetic mutations in the CBS and methylenetetrahydrofolate reductase (MTHFR) genes (involved in the folate cycle) can lead to HHcy [42,43,44,45]. Different genetic variants of the CBS and MTHFR genes that lead to HHcy can also be associated with other disease conditions (Table 1). Other than genetic factors, people also develop HHcy via various environmental factors, including consuming excess amounts of a methionine-rich diet, vitamin B12/folate deficiency, alcohol intake, diabetes, and the obstruction of renal clearance [46,47]. Under HHcy conditions, the methionine cycle is generally dysregulated [48], so this leads to the disruption of multiple signaling pathways because it is the only pathway that gives rise to the production of essential methyl groups needed for the subsequent biosynthesis of cellular compounds (for example, creatine, epinephrine, carnitine, phospholipids, proteins, and polyamines) and also methylation of DNA, RNA, and histones [8,49].

3. Homocysteine Metabolism in Cancer

Hcy metabolism depends on several factors, including the intake of methionine in the diet, the production of SAM, and the type of cells in which methionine metabolism occurs [70]. Previous studies showed that high SAM levels can act as an allosteric inhibitor of methylenetetrahydrofolate reductase (MTHFR) [17]. MTHFR enzymes convert the 5,10-MTHF to 5-MTHF in the remethylation reaction [71]. So, high SAM levels prevent Hcy from entering the remethylation pathway. Interestingly a high SAM level also acts as an allosteric activator for the CBS, a rate-limiting enzyme of the transsulfuration pathway [72,73]. This suggests that high SAM levels favor Hcy entering the transsulfuration pathway. Cancer cells depend on the methionine cycle for their methylation reaction, producing high SAM, which leads to more production of GSH and H2S via the transsulfuration pathway. Hcy metabolism also depends on the dietary methionine load, which can affect SAM synthesis, suggesting a link between diet and cancer risk [10,74,75,76]. Studies suggest that when our diet contains a basal methionine level, Hcy goes to the remethylation pathway about 1.5–2.0 times more than the transsulfuration pathway [77]. Alternatively, when we take high methionine levels from the diet, Hcy cycling through remethylation is reduced by about 1.5-fold [77]. HHcy is found in ~5–7% of the general population and is associated with other disorders [7], including cancer [8,78,79,80,81]. Hcy metabolism pathways, including the transsulfuration and remethylation pathways, are associated with several types of cancer [82,83,84,85,86,87,88,89,90,91]. Recent advancements in research found a close link between HHcy and cancer that is discussed in the following paragraph.

4. Association of Hyperhomocysteinemia and Cancer

A study by Lily L Wu and James T Wu showed that patients (who were not taking antifolate drugs) with breast, ovarian and pancreatic carcinoma had elevated serum Hcy levels [92]. Elevated Hcy is also associated with a rapid proliferation rate of tumors in leukemia patients [93] and ovarian cancer [94]. Cancer cells have a high proliferation rate, so they depend more on the methionine cycle for the DNA, RNA, and histone methylation reactions. This methionine dependency or overproduction of Hcy could be a phenotypic expression of malignancy. This suggests that elevated Hcy could be an early carcinogenesis marker and a sensitive marker for detecting recurrence. Serum tumor markers have been used most frequently for monitoring cancer patients during therapy [95].

4.1. High Plasma Hcy Levels and Cancer

High homocysteine levels have been associated with various types of cancer, as summarized in Table 2. These studies (in Table 2) suggest that patients with advanced-stage cancer show higher Hcy levels than patients with early-stage cancer. This suggests that high Hcy levels can lead to apoptosis, and cancer cells in the late stage are more proliferative, so they secrete Hcy outside the cells.
Additionally, patients who underwent surgery or chemotherapy showed increased Hcy levels in their blood. As most of the chemotherapy drugs (alkylating agents, antimetabolites, methotrexate, hormones, and antagonists) are antifolate, folate deficiency can increase Hcy levels in these patients [119]. Another study showed that older cancer patients have a higher risk of developing HHcy than younger [120], suggesting age is another causing factor for high Hcy levels in cancer patients. Venous thromboembolism (VTE) is the most common complication associated with cancer, and it is also shown to be the most common cause of death in cancer. Interestingly, HHcy patients also developed venous thromboembolism, suggesting a link between cancer-associated complications and high Hcy levels. Moreover, a study showed that cancer patients without HHcy did not show venous thromboembolism [121].

4.2. Alteration in Hcy Metabolism Gene and Risk of Cancer

Previous studies have identified numerous enzyme mutations and polymorphisms (MTHFR, CBS, MTRR, MTR, MTHFD, BHMT, TYMS, TCN 2) that regulate Hcy metabolism [122,123,124,125,126,127,128]. These mutations and polymorphisms are often linked to HHcy and different cancer types (Table 2). The most common mutations in MTHFR 677C->T transition at codon 222 and 1298A->C transversion at codon 429 have been associated with cervical [68], colorectal [129], endometrial [130], and esophageal cancer [131]. Interestingly, the 677TT and 1298CC homozygotes have been found to have reduced prostate cancer risk, as the frequencies are very low (9 and 11%, respectively) [90,99], suggesting the risk factor of specific polymorphism depends on the types of cancer. In addition to genetic polymorphisms, many environmental factors, including folate status, methionine, and the effects of alcohol consumption, play a vital role in the causation of cancer. This understanding gives rise to the targeted therapy approach, where specific mutation types can be targeted with a specific drug. Similarly, MTRR gene A66G Ile22Met is found to be associated with colorectal cancer [87] and leukemia [21]. It is also noted that homozygotes (GG) have a three-fold higher risk of colorectal cancer than with that heterozygote (AG) polymorphism. As this allelic frequency varies between the different ethnic groups, this suggests that some populations may have a higher risk for certain types than others. Likewise, one significant polymorphism (MTR A2756G; Asp919Gly) has been documented in MTR [28]. A 1958G->A; Ala653Gly polymorphism in the MTHFD-1 gene was associated with acute lymphoid leukemia [132], but no association was reported with lung cancer [133]; an inverse association was reported with colon cancer.

4.3. Homocysteine-Mediated Epigenetic Alterations and Risk of Cancer

Epigenetics are the process of changes in phenotype without alteration of the DNA sequence; this can be heritable or achieved through gene–environmental interaction [134]. There are three types of epigenetic modification: (1) DNA methylation, (2) histone modification, and (3) RNA interference. Methylation can occur in DNA, RNA, and histone protein, and this process is mediated via the methionine cycle. There are three DNA methyltransferase (DNMT) types: DNMT1, DNMT3a, and DNMT3b. SAMs act as a crucial substrate methylation reaction via DNMTs. SAM levels can be changed via environmental factors like a high methionine diet, folate deficiency, vitamin B6, and vitamin B12. Many studies have connected Global DNA hypomethylation to cancer [135], suggesting that cancer cells show differential signaling than normal cells due to high SAM levels.
Gene activation or deactivation depends upon the methylation pattern of the N-terminal tail of histones [136]. Moreover, crosstalk between these histone tail modifications (methylation, acetylation, and homocysteinylation) may have mechanistic linkages with different types of cancer [137]. Although many studies showed that high Hcy levels are associated with different epigenetic alterations and associated with cellular pathology [8,138], minimal studies have shown the role of these modifications in cancer. A study noted that Hcy in various concentrations might alter gene silencing and activation in different patterns [139]. Studies suggest that severe HHcy may induce more injurious effects via alteration of the methylation reaction [140].
Global genomic hypomethylation has been found in many types of cancer, including metastatic prostate, chronic lymphocytic, and hepatocellular carcinoma [141,142,143,144]. Regional hypomethylation of DNA sequences is also often observed during the early stages of tumorigenesis and in abnormal nonneoplastic tissue, such as hyperplasia [145]. DNA hypomethylation leads to the decondensation of pericentromeric heterochromatin and the activation of retrotransposon elements [146]; these have been associated with activating some oncogene and deactivating some tumor suppressor genes [147].

4.3.1. Hcy-Mediated DNA Methylation and Cancer

A previous study showed that methionine-rich food induces intra-cellular SAM levels, and as a consequence, global hypermethylation occurs and induces Hcy levels [148]. Where another study noted elevated Hcy-induced SAH levels, this induced SAH, in turn, inhibited SAM-dependent methyltransferases (such as DNMTs) via a negative feedback mechanism [140]. These studies suggest high Hcy levels may result in DNA hyper/hypomethylation. Moreover, many researchers using human and animal models proposed that HHcy leads to hyper/hypomethylation in a tissue-specific manner [149,150,151]. Cancer patients often show high Hcy levels, suggesting a possible link between Hcy-mediated hyper/hypomethylation and the causation of different types of cancer. Indeed, a study found HHcy-mediated hypermethylation of CpG islands located in the promoter of the ERα gene in breast cancer cell cells [152]. Interestingly, Zhang et al. showed that 10 and 30 μmol/L Hcy levels induced hypomethylation, whereas 100 and 300 μmol/L Hcy levels induced hypermethylation in the promoter of the Dimethylarginine Dimethylaminohydrolase 2 (DDAH2) gene [153]; this result suggests that hyper/hypomethylation may also depend on levels of Hcy production. Additionally, the methylation pattern also depends on many other factors such as DNA replication, chromatin accessibility, local availability of SAM, nutritional factors (folate supplementation), and aging [154]. Although hypo/hypermethylation of DNA depends on the HHcy state and tissue types [62,149,150,151], very limited studies have been carried out so far to show the association of HHcy and causation, progression and metastasis of cancer.

4.3.2. Hcy-Mediated Histone Modification and Cancer

Histone protein is present in the nucleosomes, where DNA molecules warp around at specific intervals [155]. Many post-translation modifications (acetylation, methylation, phosphorylation, ubiquitination, and sumoylation) of histones lead to gene activation and inactivation [156]. These modifications are dynamic; one set of enzymes (called writers) can put down these activation/repressive marks, and another group of enzymes (erasers) can reverse these marks [156]. Although alteration of histone modifications can cause upregulation or downregulation of specific gene expression, minimal studies have been conducted on HHcy-mediated histone modification and its associated pathology in cancer. Since HHcy can inhibit SAM-dependent methyltransferases via a negative feedback mechanism [157], it can be concluded that HHcy can also alter histone methylation patterns that might influence tumor formations. Indeed, some studies found that these histone modifications act as drivers for different types of cancer, as reviewed by Levi A Garraway et al. and Kristian Helin et al. [158,159]. However, histone modifications also vary between cell types [160], so various histone modifications may lead to different types of cancer. Which factors and how these modifications have been regulated in different cell types that lead to different types of cancers is something that needs to be explored in the near future.

4.3.3. Hcy-Mediated RNA Interference and Cancer

Earlier researchers used to think that RNA had only a housekeeping function (tRNAs and rRNAs) and a messenger function (mRNA) [161]; however, recent studies have found many new classes of regulatory non-coding RNAs. Some important non-coding RNAs are micro-RNA, endogenous small interfering RNAs (endo-siRNAs), PIWI-associated RNAs (piRNAs), and long non-coding RNAs. The discovery of non-coding RNAs has completely updated our understanding of cancer research [162]. The prognosis value of microRNA (miRNA) and long non-coding RNA (lncRNA) are widely reported in cancers [163,164]. Many studies showed that HHcy interferes with microRNA regulation and long non-coding RNA (lncRNA) [165], suggesting a link between HHcy and abnormal gene expression in cancer progression. Although most cancer research has focused on the abnormal expression of oncogenes or tumor suppressor genes, 97% of the human genome consists of non-coding sequences, leading researchers to investigate this dark matter of tumorigenesis. Non-coding RNAs can induce tumorigenesis and tumor progression via transcriptional and post-transcriptional modification, chromatin remodeling, and signal transduction. Although, to date, most of the integration of non-coding RNAs and tumorigenesis is still unknown, current research has started uncovering the complex network of the interaction of non-coding RNAs and how they modify the expression of oncogenes and tumor suppressor genes. These non-coding RNAs present in a tissue-specific manner and are considered as diagnostic, prognostic, and therapeutic targets in different diseases. There is growing research about the dysregulation of Circular RNAs (circRNAs) in cancer [166,167,168,169]. Recent reports show that circRNAs play essential roles in prostate cancer’s progression, proliferation, and epithelial–mesenchymal transition (EMT) [170]. In our previous studies, we noticed that under HHcy conditions circRNAs profile differently than in normal conditions [166,167,171]. HHcy-mediated, non-coding RNAs vary in different tissue types, suggesting more research is needed to identify specific changes in non-coding RNA based on the cancer types.

4.4. Hcy-Mediated H2S Production and Risk of Cancer

H2S was previously thought of as a toxic gas. However, recent studies found that other than from gut microbiota, H2S is produced inside a cell via Hcy metabolism [172]. H2S acts as a gasotransmitter like other gasotransmitters (for example, nitric oxide) and has a cytoprotective role [17]. H2S plays a crucial role in reducing oxidative and ER stress during HHcy conditions [7], suggesting a favorable role of H2S in cancer progression. Many studies also reported the role of H2S in cell proliferation, viability, and migration of cancer cells [173]. The CBS gene in Hcy metabolism typically catalyzes the condensation of serine with Hcy to produce cystathionine (in a transsulfuration reaction), whereas it produces H2S via β-elimination and β-replacement reactions [174]. Both β-elimination (catalysis of cysteine) and β-replacement (reaction of L-cysteine and 2-mercaptoethanol) reactions produce H2S. Many clinical studies have shown that there is CBS overexpression and increased H2S production in many cancer types [175,176]. Previous studies showed that tumor cells have a high proliferative rate, producing a massive amount of reactive oxygen species (ROS) [177] and needing neoangiogenesis [178]. In contrast, many studies have suggested that H2S reduces oxidative stress, induces cell proliferation and viability, and improves neoangiogenesis [7,48,172,179]. As SAM is an allosteric activator of CBS that binds to the regulatory domain of CBS and regulates H2S production, indeed, it helps in the growth of tumor cells [180]. Therefore, future strategies to treat cancer patients should involve modulation of CBS and H2S levels.

5. Multifactorial Role of H2S in Cancer

Recent studies showed that H2S production helps induce cancer cell proliferation, viability, invasion, and metastasis [18]. Increasing levels of H2S have been proposed to induce cancer development by regulating a wide variety of cancer-related processes; this suggests that targeting H2S production could be a beneficial tool for cancer treatment. This section focuses on how H2S plays a role in cancer progression by targeting different processes, including oxidative stress, anti-apoptosis, DNA repair, tumor growth, cancer metabolism, metastasis, and angiogenesis (summarized in Figure 3).

5.1. H2S Production via Dysregulation of CBS, CSE, and 3MST Genes in Cancer

H2S is produced endogenously through the transsulfuration pathway (involving CBS, CSE, and 3MST enzymes) of Hcy metabolism, as shown in Figure 4 [181]. Three enzymes that catalyze H2S production are often found dysregulated in cancer, as shown in Table 3.

5.1.1. Dysregulation of CBS in Cancer

The main rate-limiting enzyme of the trans-sulphuration reaction of Hcy-metabolism is CBS, which catalyzes H2S production by driving the beta-replacement. The CBS gene is often found to be upregulated in colon cancer, ovarian cancer, breast cancer, thyroid cancer, and gallbladder adenocarcinoma tissues [182,184,185,197]. A study found that DNA methylation of the CBS promoter favors colon cancer progression [198]. SAM can allosterically activate the CBS gene to favor the cell proliferation of colon cancer cells [199]. Additionally, CBS can also be controlled via its redox sensitivity through the 272CXXC275 motif [200]. The high proliferation of cancer cells creates redox stress conditions inside the cells, which activates the CBS gene to produce H2S through the 272CXXC275 motif [200]. Although some studies showed that CBS expression is downregulated in glioma tumor cells, gastrointestinal cancer cells [186,187,201], and hepatocellular carcinoma, alternatively, reduced CBS expression upregulates the 3-MST gene in glioma tumor cells [202].

5.1.2. Dysregulation of CSE in Cancer

CSE is one of the three H2S-producing enzymes in the transsulfuration pathway of Hcy metabolism. CSE has been upregulated in multiple cancer types, including prostate cancer, gastric cancer, and melanoma cells [190,203,204]. CSE was found to be induced by oxidative stress, ER stress, Golgi stress, inflammation, and starvation [205]. Unlike CBS, CSE can be upregulated transcriptionally via cellular stress response [206]. Under oxidative stress condition, nuclear factor (erythroid-derived 2)-like 2 (Nrf2) induces CSE expression through binding to its antioxidant response element (ARE) at 5′-untranslated regions (UTR), which in turn induce H2S production [207]. Overexpression of another transcription factor, specificity protein (SP) 1, induces H2S generation via binding to the CSE promoter [208]. Similarly, another study showed that tumor necrosis factor α (TNFα) induces H2S production through SP1-mediated CSE promoter binding [209]. In prostate cancer, a study found that CSE over-expression increased H2S production that led to the activation of nuclear factor-κB (NF-κB)-mediated interleukin 1β (IL-1β) signaling, resulting in enhanced cell invasion, angiogenesis, lymphangiogenesis, tumor growth, and metastasis [210]. Moreover, the upregulation of CSE by the STAT3 pathway increased breast cancer cell proliferation, growth, and migration [188]. Similarly, the upregulation of CSE by the Wnt/β-catenin pathway increased cell proliferation in colon cancer [193] whereas by extracellular signal-regulated protein kinase 1/2 (ERK1/2) pathway increased cell proliferation in liver cancer [192].

5.1.3. Dysregulation of 3MST in Cancer

3MST is another H2S-producing enzyme primarily regulated through a redox-sensitive mechanism [206]. As oxidative stress is one of the characteristics of cancer cells, it seems that cancer cells primarily depend on 3MST for H2S production. During oxidative stress condition, 3MST becomes activated via oxidation at Cys247 and subsequently produces H2S to control cellular homeostasis [211]. Indeed, pharmacological inhibition of 3MST has been found to reduce cell proliferation, migration, and bioenergetics in colon cancer cells [212]. More research is needed to understand how different cancer types regulate 3MST to maintain their cellular redox balance.

5.2. H2S-Mediated Redox Balance in Cancer

As cancer cells have a high proliferative rate, they produce many free radicals [213]. There is the possibility that cancer cells upregulate H2S-producing enzymes in order to handle oxidative stress. Multiple studies demonstrated the cytoprotective effects of H2S in different in vitro models, all relating to its ability to neutralize a variety of reactive species [214,215,216] and reduction of a disulfide bond in proteins [217,218]. H2S in water dissociates into H+, HS, and S2− ions. HS has the capacity to scavenge ROS. H2S itself has also been recognized to be a reducing agent, as it can react directly with and quench the superoxide anion (O2−) [219,220] and free radicals like peroxynitrite [221] as well as other ROS in vitro. Micro concentrations of H2S generated from Na2S/NaHS were found to neutralize free oxyradicals [222], peroxynitrite [214], hypochlorous acid [215], and Hcy [216] in in vitro conditions. There is no sulfide receptor in mammalian cells that is responsible for the biological actions of sulfide; hence sulfide, as a thiol with strong reducing activities, may also be a redox-controlling molecule similar to other small thiols, such as cysteine and GSH [214,223]. A study using primary cultures of neurons found that H2S increases cellular GSH levels by enhancing gamma-glutamylcysteine synthetase activity and upregulating cystine transport [223]. Similarly, another study reported that 100 μM NaHS induces glutamate uptake by assisting glial glutamate transporter-1 (GLT-1) and enhances cysteine transport and GSH synthesis [224]. In support of this effect, multiple studies demonstrated that H2S induces cellular GSH in the brain [225], spinal cord [226], heart [227], lung [228], kidney [229], liver [228], and gastrointestinal tract [230,231]. Moreover, recent reports suggested that H2S could attenuate cellular oxidative stress by improving the activities of catalase [227,232,233,234] and glutathione peroxidase [235,236,237].

5.3. H2S-Mediated Recovery of Hypoxia in Cancer

Hypoxia is one of the hallmarks of solid tumors. H2S has been widely studied for its effects on the regulation of oxygen homeostasis via inhibiting HIF-1α activation [238]. Different studies found upregulation of H2S-producing enzymes under hypoxia conditions and its associated cancer progression [239,240]. In addition to this, our previous studies found that under hypoxia conditions, H2S induces neoangiogenesis via upregulation of the PPAR-c/HIF-1α signaling pathway [48]. Similarly, another study identified that H2S enhances HIF-1α expression via the downregulation of miR-640 [241]. In non-small cell lung cancer, a study proposed that H2S might activate HIF-1α via the PI3K/AKT pathway leading to angiogenesis [242]. Similarly, another study showed that under hypoxia, cancer cells produce H2S via induction of CSE to facilitate angiogenesis [243].

5.4. H2S-Mediated Recovery of Apoptosis in Cancer

Apoptosis is the process of cell death that happens naturally due to physiological or environmental stress [2]. Inhibition of apoptosis is one of the hallmarks of cancer progression that allows cancer cells to survive under various stresses [244]. Recent studies found that H2S has an antiapoptotic property in various cell types [17]. Different studies also found that cancer cells produce more H2S to evade apoptosis [245,246,247,248]. These studies suggest that, like classical antioxidants (for example, GSH), H2S inhibits apoptosis in cancer cells via scavenging ROS and reactive nitrogen species (RNS). The cancer cell has a high metabolic activity due to the high proliferative rate, and this leads to the generation of ROS and RNS. So, to recover from this oxidative stress condition, cancer cells need to produce more antioxidants like H2S to create profound antioxidant protection [206].
H2S not only suppresses apoptosis through the reduction in oxidative stress but is also found to activate various antiapoptotic pathways, including NF-κB [209], kelch-like ECH-associated protein 1 (Keap1) [249], and mitogen-activated protein kinase kinase 1 (MEK1) [250]. When NF-κB signaling becomes activated, it further activates multiple antiapoptotic genes, including the X-linked inhibitor of apoptosis protein (XIAP), cellular Inhibitors of Apoptosis Proteins (cIAPs), and the B-cell lymphoma 2 (Bcl-2) [251]. In contrast, Keap1 is mediated by persulfidation by H2S; after persulfidation, Keap1 acts as an adaptor for the Keap1-Cul3-RBX1 E3 ligase complex, which targets Nrf2 to proteasomal degradation [252]. Nrf2 acts as a transcription factor for genes containing antioxidant response elements (AREs) to suppress apoptosis in cancer cells [252]. The other process of H2S-mediated inhibition of apoptosis is via the activation of MEK1, which is one of the classical MAP kinase family proteins. MEK1 generally suppresses apoptosis via inhibition of the expressions of apoptotic-related proteins, including Bad, Bim-EL, Caspase 9, MCL-1, and TNFR [253].

5.5. H2S-Mediated DNA Repair in Cancer

H2S has been found to activate the DNA repair process via MEK1/Protein poly [ADP-ribose] polymerase 1 (PARP1)-mediated signaling pathways in cancer cells [250]. After the persulfidation of MEK1 at Cys341 residue by H2S, MEK1 translocates to the nucleus to stimulate PARP-1. PARP-1 is widely known as a sensor of DNA single- or double-strand breaks [254]. This suggests that cancer cells may use H2S to recover from DNA damage during proliferation. Moreover, H2S has also been found to help in mitochondrial DNA (mtDNA) repair via persulfidation on mt-specific DNA repair enzymes EXOG at Cys76 [255], which suggests that cancer cells may skip the apoptosis process via H2S-mediated recovery of DNA damage.

5.6. H2S-Mediated Tumor Growth and Metastasis in Cancer

Different studies found that higher levels of H2S in multiple cancer types [182,188,210,212] and inhibition of H2S production via suppression of CBS or CSE activities cause a reduction in tumor growth in multiple cancer types [182,210,255]. This suggests the critical role of H2S in the growth, proliferation, and survival of cancer cells. In addition, many studies found that endogenous H2S promotes cancer cell migration and invasion in multiple cancer types [210,242,256,257]. These studies showed that H2S promotes the metastasis process via various mechanisms, which include induction of epithelial-to-mesenchymal transition (EMT). Moreover, NF-κB is a key player in cancer metastasis; as H2S induces the persulfidation of NF-κB, it helps p65 to translocate into the nucleus and induce expressions of the metastatic promoting gene [210].

5.7. H2S-Mediated Metabolism in Cancer

Cancer cells have a very high proliferative rate, so they require more ATP production to maintain cellular energetics [258]. Endogenous H2S production was shown to act as a metabolic substrate for mitochondrial ATP production in cancer cells [199]. Moreover, H2S was found to increase the catalytic activity of mitochondria ATP synthase via persulfidation of ATP synthase (ATP5A1), which may induce mitochondrial ATP production [258]. To support their high growth rates, cancer cells preferentially convert glucose to lactate by aerobic glycolysis even in sufficient O2 (Warburg effect). In this process, lactate dehydrogenase A (LDHA) acts as a key player, and it is found that cancer cells induce LDHA activity via the persulfidation of LDHA at Cys163. Consistent with this, depletion of H2S production via knockdown of CBS was also found to reduce ATP production in cancer cells [176,182].

5.8. H2S-Mediated Angiogenesis in Cancer

Angiogenesis is one of the hallmarks of cancer; during tumor growth and metastasis, tumor cells secret proangiogenic factors such as VEGF [259]. Previous studies found that H2S induced angiogenesis under different disease conditions [48], including cancer [243]. Similarly, suppressing H2S production via silencing the CBS gene reduced angiogenesis in colon and ovarian cancer [176,182]. Additionally, suppression of H2S production via silencing another H2S-producing enzyme, CSE, was found to block angiogenesis [210]. Moreover, H2S was found to promote angiogenesis via activation of HIF-1α [197]. Additionally, H2S-mediated induction of angiogenesis has been found via NF-κB/IL-1β, PI3K/AKT, and MAPK signaling pathways [199,210].

5.9. H2S-Mediated Reduction in ER Stress in Cancer

As cancer cells have a high proliferation rate, they create different gene mutations, produce more misfolded proteins, and induce ER stress response [260]. As H2S was found to reduce ER stress in different disease conditions [7,261,262,263], it suggests that cancer cells may produce more H2S to recover from ER stress. In addition, as cancer cells mainly depend on the methylation cycle, this also produces a high amount of Hcy, which induces homocysteinylation of protein and further activates ER stress [264]. There is also the possibility that H2S can reverse protein homocysteinylation [265].

6. Current Cancer Therapeutics Targeting the Hcy and H2S Signaling and Their Limitations

Current treatment options for cancer are based on specific types of cancer and the stage of cancer; these include chemotherapy, radiation therapy, immunotherapy, and targeted therapy [266]. While treatment increases the lifespan of many patients, it is also associated with many side effects that will determine the health consequences. Also, the efficacy of these treatment options is limited by the resistance that patients develop [267]. As chemotherapy has many side effects due to the property of killing normal healthy cells, recent treatment is shifting gears toward targeted therapy approaches.
Methionine is an essential amino acid, and many tumor cells show dependence on exogenous sources of methionine [4]. Studies showed that methionine restriction inhibits cancer cell growth proliferation while normal cells remain unaffected [4]. In addition, methionine restriction showed enhanced efficiency of chemotherapy and radiotherapy in animal models [268]. A previous study showed that methionine restriction for an average of 17 weeks is safe and feasible in patients with advanced metastatic cancer [14].
Moreover, there have been many drugs developed that target the methionine cycle, but none of them showed clinical success. Antifolate drugs (for example, methotrexate) that interfere with the folate cycle have shown limited clinical efficiency due to side effects and resistance [269,270]. As the methylation cycle is essential for normal cells, these drugs kill both cancer and healthy cells. However, small molecule inhibitors that target serine synthesis pathways have been successful in in vitro and animal studies [271,272]. However, in order to reduce side effects, the drug used has to be specific to the particular cancer. For small molecule inhibitors that inhibit PHGDH (for example, NCT-503, CBR-5884), the tumor has to be fully addicted to PHGDH. Also, it needs to be ensured that this drug does not interfere with any other signaling pathways critical for signaling. If any type of tumor is not fully dependent on a specific pathway, that means the cancer cells may be using another source for that specific pathway. For targeting the serine synthesis pathway, if any specific cancer cell lines do not respond to the drug, these cells may be using exogenous serine supplementation. So, to target this cancer type with this drug, we should also consider the external source of nutrients/diet. Another type of mechanism is called the compensatory mechanism, by which cancer cells become resistant to specific drugs. For example, any drug targeting mitochondrial methylenetetrahydrofolate dehydrogenase (MTHFD1L) cancer cells can compensate using cytoplasmic MTHFD1 [273].
There are variable reports so far documented when it comes to targeting H2S metabolism. Lower doses of H2S donor compounds were found to have pro-cancer activity by different mechanisms [274], whereas higher doses had anticancer activity due to uncontrolled intracellular acidification [275,276,277]. Various H2S donor compounds (for example, NaHS, Na2S, GYY4137) tested preclinically for their anticancer property [275,276,277]. In contrast, as many studies showed, endogenous H2S has beneficial effects for tumor growth and metastasis, and inhibiting endogenous production of H2S (via targeting H2S-producing enzymes) may be a good strategy. DL-propargylglycine (PAG) is an inhibitor of CSE that showed limited cell permeability [218] and non-selective inhibition of other enzymes [278,279,280]. Another inhibitor of H2S-producing enzymes is aminooxy acetic acid (AOAA), which is also found to inhibit cysteine aminotransferase (CAT) [281]. HMPSNE is an inhibitor that targets the 3rd H2S-producing enzyme 3MST, showed the highest selectivity for 3MST [31,282], and was found to inhibit cell proliferation of colon cancer [212,283]. In order to make a more efficient drug that inhibits H2S production, more research is needed.
Although targeting H2S production showed promise, there are a few limitations. Firstly, many previous studies that targeted H2S production used the pharmacological inhibitor AOAA (a CBS inhibitor). However, AOAA showed nonspecific inhibition of CSE, 3MST, and over thirty other cellular enzymes [284]. Similarly, another H2S production inhibitor, β-cyano-alanine, showed suboptimum specificity [285]. Secondly, there are functional differences between H2S production enzymes in different cancer types. For example, in prostate cancer, mutant CSE was found to be lowly invasive but did not interfere with cell migration capacity [210], suggesting the mutation-specific targeting of H2S production will be necessary for future therapeutics.

A Hypothesis of Targeting the Hcy and H2S Balance for Cancer Treatment and Its Application

Due to the high proliferative rate, cancer cells require an external source of methionine for protein formation, methylation reaction (epigenetic alteration), and the production of H2S (antioxidant). When cancer cells use the methionine cycle, it produces a massive amount of Hcy, which is toxic for the growth of the tumors. As previous studies found that high SAM levels act as an allosteric inhibitor of MTHFR (involved in the folate cycle) and activator of CBS, so when cancer cells produce high SAM levels, it prevents Hcy from entering the remethylation pathway rather than allowing excess Hcy to shuttle to the transculturation pathway to produce H2S [71,72,73]. This production of high H2S in cancer cells helps them to survive, proliferate, grow, and metastasize. Although many therapeutic strategies have been developed either by methionine restriction or targeting different enzymes of the methionine cycle, folate cycle, and transsulfuration pathway, none of these treatment strategies showed effectiveness in clinical studies. So, in the future, more research is required where we can utilize their dependence on the methionine cycle and target specific enzymes to treat cancer. So far, we have noticed that cancer cells depend on the methionine cycle more, so if we can target both the transsulfuration pathway (via CBS) and the remethylation pathway at the same time, as shown in Figure 5, then these cells will build up toxic Hcy and inhibit the production of H2S. High levels of Hcy will induce apoptosis, protein oxidation, and oxidative and ER stress and inhibit angiogenesis, whereas low levels of H2S will inhibit cancer growths; as a result, tumor progression will be inhibited due to the effects of high Hcy and low levels of antioxidants like H2S.
Efforts to interfere with the methylation cycle in different cancers have reached a plateau, with only incrementally effective inhibitors developed to date. In order to overcome this barrier and develop highly effective inhibitors, we need to understand how to target this Hcy and H2S signaling more precisely with minimal side effects. Based on the above discussion, although it is apparent that targeting Hcy and H2S balance is beneficial for cancer treatment, there are however no studies have been carried out that exploit this circuit. This extensive review article will likely lead us and many in academia and industry to develop next-generation therapeutic agents targeting the blocking of H2S production and Hcy remethylation. The significance and impact would be profound in increasing efficacy and reducing toxicity for a large number of cancer patients where targeted therapy was shown to be non-effective. For example, in treating triple-negative breast cancer, this treatment strategy will be a good option as there is no other oncogene-driven monotherapy available. Similarly, cancer types that are more dependent on the methionine cycle will be the best option to use this strategy. Another exciting cancer treatment aspect that was not covered in this review is Hcy-mediated epigenetic alteration and H2S-mediated polysulfide production in cancer. So, a better understanding of their signaling in cancer will undoubtedly facilitate better treatment for cancer patients.

7. Conclusions

Given that cancer cells depend on the methionine cycle for their methylation reaction and H2S production, many researchers tried different strategies to target these signaling pathways. Unfortunately, none of the strategies turned out beneficial for cancer treatment. Based on current understanding, it is noted that indefinite targeting of the methionine cycle, either via a methionine restriction diet or targeting different enzymes of the methionine cycle, is not feasible due to the development of resistance, non-responsiveness, and numerous side effects. However, targeting H2S production showed to be somewhat promising based on its effects on cancer progression via inhibition apoptosis, oxidative stress, ER stress, and stimulation of the DNA repair process, cancer metabolism, tumor growth, and metastasis. Again, this strategy did not benefit cancer treatment due to nonspecific targeting. Therefore, for future prospects, it is necessary to target the transsulfuration pathway for blocking H2S production and the remethylation pathway to build up toxic Hcy. As we noticed, Hcy has detrimental effects on cells via apoptosis, protein oxidation, lipid peroxidation, poor angiogenesis, oxidative stress, and ER stress. So, excess Hcy build-up will not be able to recover via simultaneous blocking of H2S production, which leads to the regression of tumor growth. However, more research is anticipated to test this proof of concept for cancer treatment.

Funding

This research received no external funding.

Conflicts of Interest

The author declares no conflict of interest.

Abbreviations

SAHS-Adenosyl Homocysteine
HcyHomocysteine
HHcyHyperhomocysteinemia
SAMS-Adenosyl Methionine
H2SHydrogen Sulfide
GSHGlutathione
ROSReactive Oxygen Species
EREndoplasmic Reticulum
MATMethionine Adenosyl Transferase
CBSCystathionine Β-Synthase
CSECystathionine Γ-Lyase
MTHFRMethylenetetrahydrofolate Reductase
MTRRMethionine Synthase Reductase
MTRMethionine Synthase
MTHFDMethylenetetrahydrofolate Dehydrogenase
BHMTBetaine Homocysteine Methyltransferase
TYMSThymidylate Synthase
TCN2Transcobalamin 2
MTHFD1LMethylenetetrahydrofolate dehydrogenase
DNMTSDNA Methyltransferases
VSMCSVascular Smooth Muscle Cells
DDAH2Dimethylarginine Dimethylaminohydrolase 2
HMTHistone Methyltransferase
lncRNALong Non-Coding RNA
miRNAMicroRNA
circRNACircularRNA
NF-κBNuclear factor kappa-light-chain-enhancer of activated B cells
Keap1Kelch-like ECH-associated protein 1
MEK1Mitogen-activated protein kinase kinase1
XIAPX-linked inhibitor of apoptosis protein
cIAPsCellular Inhibitors of Apoptosis Proteins
Bcl-2B-cell lymphoma 2 gene
AREsAntioxidant Response Elements

References

  1. Global, regional, and national life expectancy, all-cause mortality, and cause-specific mortality for 249 causes of death, 1980–2015: A systematic analysis for the Global Burden of Disease Study 2015. Lancet 2016, 388, 1459–1544. [CrossRef] [Green Version]
  2. Hanahan, D.; Weinberg, R.A. Hallmarks of cancer: The next generation. Cell 2011, 144, 646–674. [Google Scholar] [CrossRef] [Green Version]
  3. Altea-Manzano, P.; Cuadros, A.M.; Broadfield, L.A.; Fendt, S.M. Nutrient metabolism and cancer in the in vivo context: A metabolic game of give and take. EMBO Rep. 2020, 21, e50635. [Google Scholar] [CrossRef]
  4. Cellarier, E.; Durando, X.; Vasson, M.P.; Farges, M.C.; Demiden, A.; Maurizis, J.C.; Madelmont, J.C.; Chollet, P. Methionine dependency and cancer treatment. Cancer Treat. Rev. 2003, 29, 489–499. [Google Scholar] [CrossRef] [PubMed]
  5. Dunn, R.; McCoy, J.; Simsek, M.; Majumdar, A.; Chang, S.H.; Rajbhandary, U.L.; Khorana, H.G. The bacteriorhodopsin gene. Proc. Natl. Acad. Sci. USA 1981, 78, 6744–6748. [Google Scholar] [CrossRef] [PubMed]
  6. Navik, U.; Sheth, V.G.; Khurana, A.; Jawalekar, S.S.; Allawadhi, P.; Gaddam, R.R.; Bhatti, J.S.; Tikoo, K. Methionine as a double-edged sword in health and disease: Current perspective and future challenges. Ageing Res. Rev. 2021, 72, 101500. [Google Scholar] [CrossRef]
  7. Majumder, A.; Singh, M.; Behera, J.; Theilen, N.T.; George, A.K.; Tyagi, N.; Metreveli, N.; Tyagi, S.C. Hydrogen sulfide alleviates hyperhomocysteinemia-mediated skeletal muscle atrophy via mitigation of oxidative and endoplasmic reticulum stress injury. Am. J. Physiol. Cell Physiol. 2018, 315, C609–C622. [Google Scholar] [CrossRef]
  8. Majumder, A.; Behera, J.; Jeremic, N.; Tyagi, S.C. Hypermethylation: Causes and Consequences in Skeletal Muscle Myopathy. J. Cell. Biochem. 2017, 118, 2108–2117. [Google Scholar] [CrossRef]
  9. Combs, J.A.; DeNicola, G.M. The Non-Essential Amino Acid Cysteine Becomes Essential for Tumor Proliferation and Survival. Cancers 2019, 11, 678. [Google Scholar] [CrossRef] [Green Version]
  10. Wanders, D.; Hobson, K.; Ji, X. Methionine Restriction and Cancer Biology. Nutrients 2020, 12, 684. [Google Scholar] [CrossRef] [Green Version]
  11. Peng, H.; Yan, Y.; He, M.; Li, J.; Wang, L.; Jia, W.; Yang, L.; Jiang, J.; Chen, Y.; Li, F.; et al. SLC43A2 and NFκB signaling pathway regulate methionine/cystine restriction-induced ferroptosis in esophageal squamous cell carcinoma via a feedback loop. Cell Death Dis. 2023, 14, 347. [Google Scholar] [CrossRef] [PubMed]
  12. Khairan, P.; Sobue, T.; Eshak, E.S.; Zha, L.; Kitamura, T.; Sawada, N.; Iwasaki, M.; Inoue, M.; Yamaji, T.; Shimazu, T.; et al. Association of dietary intakes of vitamin B12, vitamin B6, folate, and methionine with the risk of esophageal cancer: The Japan Public Health Center-based (JPHC) prospective study. BMC Cancer 2021, 21, 982. [Google Scholar] [CrossRef] [PubMed]
  13. Guo, H.; Lishko, V.K.; Herrera, H.; Groce, A.; Kubota, T.; Hoffman, R.M. Therapeutic tumor-specific cell cycle block induced by methionine starvation in vivo. Cancer Res. 1993, 53, 5676–5679. [Google Scholar]
  14. Epner, D.E.; Morrow, S.; Wilcox, M.; Houghton, J.L. Nutrient intake and nutritional indexes in adults with metastatic cancer on a phase I clinical trial of dietary methionine restriction. Nutr. Cancer 2002, 42, 158–166. [Google Scholar] [CrossRef]
  15. George, A.K.; Singh, M.; Homme, R.P.; Majumder, A.; Sandhu, H.S.; Tyagi, S.C. A hypothesis for treating inflammation and oxidative stress with hydrogen sulfide during age-related macular degeneration. Int. J. Ophthalmol. 2018, 11, 881–887. [Google Scholar] [CrossRef]
  16. Kaiser, P. Methionine Dependence of Cancer. Biomolecules 2020, 10, 568. [Google Scholar] [CrossRef] [Green Version]
  17. Majumder, A.; Singh, M.; George, A.K.; Tyagi, S.C. Restoration of skeletal muscle homeostasis by hydrogen sulfide during hyperhomocysteinemia-mediated oxidative/ER stress condition. Can. J. Physiol. Pharmacol. 2019, 97, 441–456. [Google Scholar] [CrossRef]
  18. Hellmich, M.R.; Szabo, C. Hydrogen Sulfide and Cancer. Handb. Exp. Pharmacol. 2015, 230, 233–241. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  19. Newman, A.C.; Maddocks, O.D.K. One-carbon metabolism in cancer. Br. J. Cancer 2017, 116, 1499–1504. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  20. Hasan, T.; Arora, R.; Bansal, A.K.; Bhattacharya, R.; Sharma, G.S.; Singh, L.R. Disturbed homocysteine metabolism is associated with cancer. Exp. Mol. Med. 2019, 51, 1–13. [Google Scholar] [CrossRef] [Green Version]
  21. Skibola, C.F.; Smith, M.T.; Kane, E.; Roman, E.; Rollinson, S.; Cartwright, R.A.; Morgan, G. Polymorphisms in the methylenetetrahydrofolate reductase gene are associated with susceptibility to acute leukemia in adults. Proc. Natl. Acad. Sci. USA 1999, 96, 12810–12815. [Google Scholar] [CrossRef] [PubMed]
  22. He, L.; Shen, Y. MTHFR C677T polymorphism and breast, ovarian cancer risk: A meta-analysis of 19,260 patients and 26,364 controls. OncoTargets Ther. 2017, 10, 227–238. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  23. Li, X.L.; Xu, J.H. MTHFR polymorphism and the risk of prostate cancer: A meta-analysis of case-control studies. Prostate Cancer Prostatic Dis. 2012, 15, 244–249. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  24. Mbemi, A.; Khanna, S.; Njiki, S.; Yedjou, C.G.; Tchounwou, P.B. Impact of Gene-Environment Interactions on Cancer Development. Int. J. Environ. Res. Public Health 2020, 17, 8089. [Google Scholar] [CrossRef]
  25. Chen, J.; Giovannucci, E.; Kelsey, K.; Rimm, E.B.; Stampfer, M.J.; Colditz, G.A.; Spiegelman, D.; Willett, W.C.; Hunter, D.J. A methylenetetrahydrofolate reductase polymorphism and the risk of colorectal cancer. Cancer Res. 1996, 56, 4862–4864. [Google Scholar]
  26. Ma, E.; Iwasaki, M.; Junko, I.; Hamada, G.S.; Nishimoto, I.N.; Carvalho, S.M.; Motola, J., Jr.; Laginha, F.M.; Tsugane, S. Dietary intake of folate, vitamin B6, and vitamin B12, genetic polymorphism of related enzymes, and risk of breast cancer: A case-control study in Brazilian women. BMC Cancer 2009, 9, 122. [Google Scholar] [CrossRef] [Green Version]
  27. Suzuki, T.; Matsuo, K.; Sawaki, A.; Mizuno, N.; Hiraki, A.; Kawase, T.; Watanabe, M.; Nakamura, T.; Yamao, K.; Tajima, K.; et al. Alcohol drinking and one-carbon metabolism-related gene polymorphisms on pancreatic cancer risk. Cancer Epidemiol. Biomark. Prev. 2008, 17, 2742–2747. [Google Scholar] [CrossRef] [Green Version]
  28. Chen, K.; Song, L.; Jin, M.J.; Fan, C.H.; Jiang, Q.T.; Yu, W.P. Association between genetic polymorphisms in folate metabolic enzyme genes and colorectal cancer: A nested case-control study. Zhonghua Zhong Liu Za Zhi 2006, 28, 429–432. [Google Scholar]
  29. Chen, J.; Giovannucci, E.; Hankinson, S.E.; Ma, J.; Willett, W.C.; Spiegelman, D.; Kelsey, K.T.; Hunter, D.J. A prospective study of methylenetetrahydrofolate reductase and methionine synthase gene polymorphisms, and risk of colorectal adenoma. Carcinogenesis 1998, 19, 2129–2132. [Google Scholar] [CrossRef] [Green Version]
  30. Ascenção, K.; Szabo, C. Emerging roles of cystathionine β-synthase in various forms of cancer. Redox Biol. 2022, 53, 102331. [Google Scholar] [CrossRef]
  31. Ibrahim, H.; Serag, A.; Farag, M.A. Emerging analytical tools for the detection of the third gasotransmitter H2S, a comprehensive review. J. Adv. Res. 2021, 27, 137–153. [Google Scholar] [CrossRef]
  32. Wang, D.; Yang, H.; Zhang, Y.; Hu, R.; Hu, D.; Wang, Q.; Liu, Y.; Liu, M.; Meng, Z.; Zhou, W.; et al. Inhibition of cystathionine β-synthase promotes apoptosis and reduces cell proliferation in chronic myeloid leukemia. Signal Transduct. Target. Ther. 2021, 6, 52. [Google Scholar] [CrossRef]
  33. Stipanuk, M.H.; Ueki, I. Dealing with methionine/homocysteine sulfur: Cysteine metabolism to taurine and inorganic sulfur. J. Inherit. Metab. Dis. 2011, 34, 17–32. [Google Scholar] [CrossRef] [Green Version]
  34. Veeranki, S.; Tyagi, S.C. Defective homocysteine metabolism: Potential implications for skeletal muscle malfunction. Int. J. Mol. Sci. 2013, 14, 15074–15091. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  35. Cascella, M.; Arcamone, M.; Morelli, E.; Viscardi, D.; Russo, V.; De Franciscis, S.; Belli, A.; Accardo, R.; Caliendo, D.; De Luca, E.; et al. Erratum to: Multidisciplinary approach and anesthetic management of a surgical cancer patient with methylene tetrahydrofolate reductase deficiency: A case report and review of the literature. J. Med. Case Rep. 2015, 9, 218. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  36. Hankey, G.J.; Eikelboom, J.W. Homocysteine and vascular disease. Lancet 1999, 354, 407–413. [Google Scholar] [CrossRef] [PubMed]
  37. Majumder, A.; Singh, M.; George, A.K.; Homme, R.P.; Laha, A.; Tyagi, S.C. Remote ischemic conditioning as a cytoprotective strategy in vasculopathies during hyperhomocysteinemia: An emerging research perspective. J. Cell. Biochem. 2019, 120, 77–92. [Google Scholar] [CrossRef] [Green Version]
  38. Lahiri, K.D.; Datta, H.; Das, H.N. Reference interval determination of total plasma homocysteine in an Indian population. Indian J. Clin. Biochem. 2014, 29, 74–78. [Google Scholar] [CrossRef] [Green Version]
  39. Tiahou, G.; Dupuy, A.M.; Jaussent, I.; Sees, D.; Cristol, J.P.; Badiou, S. Determinants of homocysteine levels in Ivorian rural population. Int. J. Vitam. Nutr. Res. 2009, 79, 319–327. [Google Scholar] [CrossRef] [Green Version]
  40. Maddocks, O.D.; Labuschagne, C.F.; Adams, P.D.; Vousden, K.H. Serine Metabolism Supports the Methionine Cycle and DNA/RNA Methylation through De Novo ATP Synthesis in Cancer Cells. Mol. Cell 2016, 61, 210–221. [Google Scholar] [CrossRef] [Green Version]
  41. Lehotsky, J.; Tothova, B.; Kovalska, M.; Dobrota, D.; Benova, A.; Kalenska, D.; Kaplan, P. Role of Homocysteine in the Ischemic Stroke and Development of Ischemic Tolerance. Front. Neurosci. 2016, 10, 538. [Google Scholar] [CrossRef] [Green Version]
  42. Banerjee, I.; Gupta, V.; Ganesh, S. Association of gene polymorphism with genetic susceptibility to stroke in Asian populations: A meta-analysis. J. Hum. Genet. 2007, 52, 205–219. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  43. Gao, S.; Li, H.; Xiao, H.; Yao, G.; Shi, Y.; Wang, Y.; Zhou, X.; Yu, H. Association of MTHFR 677T variant allele with risk of intracerebral haemorrhage: A meta-analysis. J. Neurol. Sci. 2012, 323, 40–45. [Google Scholar] [CrossRef] [PubMed]
  44. Kang, S.; Zhao, X.; Liu, L.; Wu, W.; Zhang, D. Association of the C677T polymorphism in the MTHFR gene with hemorrhagic stroke: A meta-analysis. Genet. Test. Mol. Biomark. 2013, 17, 412–417. [Google Scholar] [CrossRef] [Green Version]
  45. Yu, H.H.; Zhang, W.L.; Shi, J.P. Relationship between methylenetetrahydrofolate reductase gene C677T polymorphism and susceptibility of ischemic stroke: A meta-analysis. Zhonghua Yi Xue Za Zhi 2011, 91, 2060–2064. [Google Scholar]
  46. Sen, U.; Mishra, P.K.; Tyagi, N.; Tyagi, S.C. Homocysteine to hydrogen sulfide or hypertension. Cell Biochem. Biophys. 2010, 57, 49–58. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  47. Diakoumopoulou, E.; Tentolouris, N.; Kirlaki, E.; Perrea, D.; Kitsou, E.; Psallas, M.; Doulgerakis, D.; Katsilambros, N. Plasma homocysteine levels in patients with type 2 diabetes in a Mediterranean population: Relation with nutritional and other factors. Nutr. Metab. Cardiovasc. Dis. 2005, 15, 109–117. [Google Scholar] [CrossRef] [PubMed]
  48. Majumder, A.; Singh, M.; George, A.K.; Behera, J.; Tyagi, N.; Tyagi, S.C. Hydrogen sulfide improves postischemic neoangiogenesis in the hind limb of cystathionine-β-synthase mutant mice via PPAR-γ/VEGF axis. Physiol. Rep. 2018, 6, e13858. [Google Scholar] [CrossRef]
  49. Laha, A.; Majumdar, A.; Singh, M.; Tyagi, S.C. Connecting homocysteine and obesity through pyroptosis, gut microbiome, epigenetics, peroxisome proliferator-activated receptor gamma, and zinc finger protein 407. Can. J. Physiol. Pharmacol. 2018, 96, 971–976. [Google Scholar] [CrossRef]
  50. Orendac, M.; Muskova, B.; Richterova, E.; Zvarova, J.; Stefek, M.; Zaykova, E.; Kraus, J.P.; Stribrny, J.; Hyanek, J.; Kozich, V. Is the common 844ins68 polymorphism in the cystathionine beta-synthase gene associated with atherosclerosis? J. Inherit. Metab. Dis. 1999, 22, 674–675. [Google Scholar] [CrossRef]
  51. Ding, R.; Lin, S.; Chen, D. The association of cystathionine beta synthase (CBS) T833C polymorphism and the risk of stroke: A meta-analysis. J. Neurol. Sci. 2012, 312, 26–30. [Google Scholar] [CrossRef]
  52. de Franchis, R.; Fermo, I.; Mazzola, G.; Sebastio, G.; Di Minno, G.; Coppola, A.; Andria, G.; D’Angelo, A. Contribution of the cystathionine beta-synthase gene (844ins68) polymorphism to the risk of early-onset venous and arterial occlusive disease and of fasting hyperhomocysteinemia. Thromb. Haemost. 2000, 84, 576–582. [Google Scholar] [CrossRef] [PubMed]
  53. McGimpsey, S.J.; Woodside, J.V.; Cardwell, C.; Cahill, M.; Chakravarthy, U. Homocysteine, methylenetetrahydrofolate reductase C677T polymorphism, and risk of retinal vein occlusion: A meta-analysis. Ophthalmology 2009, 116, 1778–1787. [Google Scholar] [CrossRef] [PubMed]
  54. Ray, J.G.; Shmorgun, D.; Chan, W.S. Common C677T polymorphism of the methylenetetrahydrofolate reductase gene and the risk of venous thromboembolism: Meta-analysis of 31 studies. Pathophysiol. Haemost. Thromb. 2002, 32, 51–58. [Google Scholar] [CrossRef]
  55. Cai, W.; Yin, L.; Yang, F.; Zhang, L.; Cheng, J. Association between Hcy levels and the CBS844ins68 and MTHFR C677T polymorphisms with essential hypertension. Biomed. Rep. 2014, 2, 861–868. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  56. Heifetz, E.M.; Birk, R.Z. MTHFR C677T polymorphism affects normotensive diastolic blood pressure independently of blood lipids. Am. J. Hypertens. 2015, 28, 387–392. [Google Scholar] [CrossRef] [Green Version]
  57. Yang, K.M.; Jia, J.; Mao, L.N.; Men, C.; Tang, K.T.; Li, Y.Y.; Ding, H.X.; Zhan, Y.Y. Methylenetetrahydrofolate reductase C677T gene polymorphism and essential hypertension: A meta-analysis of 10,415 subjects. Biomed. Rep. 2014, 2, 699–708. [Google Scholar] [CrossRef] [Green Version]
  58. Hua, Y.; Zhao, H.; Kong, Y.; Ye, M. Association between the MTHFR gene and Alzheimer’s disease: A meta-analysis. Int. J. Neurosci. 2011, 121, 462–471. [Google Scholar] [CrossRef]
  59. Ibrahim, S.; Maqbool, S.; Azam, M.; Iqbal, M.P.; Qamar, R. CBS mutations and MTFHR SNPs causative of hyperhomocysteinemia in Pakistani children. Mol. Biol. Rep. 2018, 45, 353–360. [Google Scholar] [CrossRef]
  60. Castro, R.; Rivera, I.; Blom, H.J.; Jakobs, C.; Tavares de Almeida, I. Homocysteine metabolism, hyperhomocysteinaemia and vascular disease: An overview. J. Inherit. Metab. Dis. 2006, 29, 3–20. [Google Scholar] [CrossRef]
  61. White, R.H. The epidemiology of venous thromboembolism. Circulation 2003, 107, I4–I8. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  62. Liang, R.; Zhou, Y.; Xie, J.; Lv, W.; Kang, B.; Liang, Y.; Chen, Y.; Li, Y. Association of C677T gene polymorphisms of methylenetetrahydrofolate reductase and plasma homocysteine level with hyperlipidemia. J. South. Med. Univ. 2014, 34, 1195–1198. [Google Scholar]
  63. Al-Rubeaan, K.; Siddiqui, K.; Saeb, A.T.; Nazir, N.; Al-Naqeb, D.; Al-Qasim, S. ACE I/D and MTHFR C677T polymorphisms are significantly associated with type 2 diabetes in Arab ethnicity: A meta-analysis. Gene 2013, 520, 166–177. [Google Scholar] [CrossRef]
  64. Chang, W.W.; Zhang, L.; Yao, Y.S.; Su, H.; Jin, Y.L.; Chen, Y. Methylenetetrahydrofolate reductase (MTHFR) C677T polymorphism and susceptibility to diabetic nephropathy in Chinese type 2 diabetic patients: A meta-analysis. Ren. Fail. 2013, 35, 1038–1043. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  65. Yang, S.; Zhang, J.; Feng, C.; Huang, G. MTHFR 677T variant contributes to diabetic nephropathy risk in Caucasian individuals with type 2 diabetes: A meta-analysis. Metab. Clin. Exp. 2013, 62, 586–594. [Google Scholar] [CrossRef] [PubMed]
  66. Zhang, D.; Zhou, Y.; Han, L.; Ji, H.; Li, J. The effect of MTHFR C677T polymorphism on type 2 diabetes mellitus with vascular complications in Chinese Han population: A meta-analysis. Endocr. J. 2014, 61, 717–726. [Google Scholar] [CrossRef]
  67. Gouveia, L.O.; Canhao, P. MTHFR and the risk for cerebral venous thrombosis—A meta-analysis. Thromb. Res. 2010, 125, e153–e158. [Google Scholar] [CrossRef]
  68. Wu, C.Y.; Yang, M.; Lin, M.; Li, L.P.; Wen, X.Z. MTHFR C677T polymorphism was an ethnicity-dependent risk factor for cervical cancer development: Evidence based on a meta-analysis. Arch. Gynecol. Obstet. 2013, 288, 595–605. [Google Scholar] [CrossRef]
  69. Wu, Y.L.; Ding, X.X.; Sun, Y.H.; Yang, H.Y.; Sun, L. Methylenetetrahydrofolate reductase (MTHFR) C677T/A1298C polymorphisms and susceptibility to Parkinson’s disease: A meta-analysis. J. Neurol. Sci. 2013, 335, 14–21. [Google Scholar] [CrossRef]
  70. Laster, L.; Mudd, S.H.; Finkelstein, J.D.; Irreverre, F. Homocystinuria due to cystathionine synthase deficiency: The metabolism of L-methionine. J. Clin. Investig. 1965, 44, 1708–1719. [Google Scholar] [CrossRef]
  71. Finkelstein, J.D.; Martin, J.J. Methionine metabolism in mammals. Distribution of homocysteine between competing pathways. J. Biol. Chem. 1984, 259, 9508–9513. [Google Scholar] [CrossRef] [PubMed]
  72. Finkelstein, J.D.; Kyle, W.E.; Martin, J.L.; Pick, A.M. Activation of cystathionine synthase by adenosylmethionine and adenosylethionine. Biochem. Biophys. Res. Commun. 1975, 66, 81–87. [Google Scholar] [CrossRef] [PubMed]
  73. Stabler, S.P.; Steegborn, C.; Wahl, M.C.; Oliveriusova, J.; Kraus, J.P.; Allen, R.H.; Wagner, C.; Mudd, S.H. Elevated plasma total homocysteine in severe methionine adenosyltransferase I/III deficiency. Metab. Clin. Exp. 2002, 51, 981–988. [Google Scholar] [CrossRef] [PubMed]
  74. Stolzenberg-Solomon, R.Z.; Miller, E.R., 3rd; Maguire, M.G.; Selhub, J.; Appel, L.J. Association of dietary protein intake and coffee consumption with serum homocysteine concentrations in an older population. Am. J. Clin. Nutr. 1999, 69, 467–475. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  75. Selhub, J. Homocysteine metabolism. Annu. Rev. Nutr. 1999, 19, 217–246. [Google Scholar] [CrossRef] [Green Version]
  76. Selhub, J.; Jacques, P.F.; Rosenberg, I.H.; Rogers, G.; Bowman, B.A.; Gunter, E.W.; Wright, J.D.; Johnson, C.L. Serum total homocysteine concentrations in the third National Health and Nutrition Examination Survey (1991–1994): Population reference ranges and contribution of vitamin status to high serum concentrations. Ann. Intern. Med. 1999, 131, 331–339. [Google Scholar] [CrossRef] [Green Version]
  77. Eloranta, T.O.; Martikainen, V.; Smith, T.K. Adaptation of adenosylmethionine metabolism and methionine recycling to variations in dietary methionine in the rat. Proc. Soc. Exp. Biol. Med. 1990, 194, 364–371. [Google Scholar] [CrossRef]
  78. Veeranki, S.; Winchester, L.J.; Tyagi, S.C. Hyperhomocysteinemia associated skeletal muscle weakness involves mitochondrial dysfunction and epigenetic modifications. Biochim. Biophys. Acta 2015, 1852, 732–741. [Google Scholar] [CrossRef] [Green Version]
  79. Veeranki, S.; Tyagi, S.C. Mechanisms of hyperhomocysteinemia induced skeletal muscle myopathy after ischemia in the CBS-/+ mouse model. Int. J. Mol. Sci. 2015, 16, 1252–1265. [Google Scholar] [CrossRef] [Green Version]
  80. Winchester, L.; Veeranki, S.; Givvimani, S.; Tyagi, S.C. Exercise mitigates the adverse effects of hyperhomocysteinemia on macrophages, MMP-9, skeletal muscle, and white adipocytes. Can. J. Physiol. Pharmacol. 2014, 92, 575–582. [Google Scholar] [CrossRef]
  81. Ishii, I.; Akahoshi, N.; Yamada, H.; Nakano, S.; Izumi, T.; Suematsu, M. Cystathionine gamma-Lyase-deficient mice require dietary cysteine to protect against acute lethal myopathy and oxidative injury. J. Biol. Chem. 2010, 285, 26358–26368. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  82. Giovannucci, E.; Rimm, E.B.; Ascherio, A.; Stampfer, M.J.; Colditz, G.A.; Willett, W.C. Alcohol, low-methionine--low-folate diets, and risk of colon cancer in men. J. Natl. Cancer Inst. 1995, 87, 265–273. [Google Scholar] [CrossRef] [PubMed]
  83. Ma, J.; Stampfer, M.J.; Christensen, B.; Giovannucci, E.; Hunter, D.J.; Chen, J.; Willett, W.C.; Selhub, J.; Hennekens, C.H.; Gravel, R.; et al. A polymorphism of the methionine synthase gene: Association with plasma folate, vitamin B12, homocyst(e)ine, and colorectal cancer risk. Cancer Epidemiol. Biomark. Prev. 1999, 8, 825–829. [Google Scholar]
  84. Bravatà, V. Controversial roles of methylenetetrahydrofolate reductase polymorphisms and folate in breast cancer disease. Int. J. Food Sci. Nutr. 2015, 66, 43–49. [Google Scholar] [CrossRef] [PubMed]
  85. Kato, I.; Dnistrian, A.M.; Schwartz, M.; Toniolo, P.; Koenig, K.; Shore, R.E.; Akhmedkhanov, A.; Zeleniuch-Jacquotte, A.; Riboli, E. Serum folate, homocysteine and colorectal cancer risk in women: A nested case-control study. Br. J. Cancer 1999, 79, 1917–1922. [Google Scholar] [CrossRef] [Green Version]
  86. de Jong, M.M.; Nolte, I.M.; te Meerman, G.J.; van der Graaf, W.T.; de Vries, E.G.; Sijmons, R.H.; Hofstra, R.M.; Kleibeuker, J.H. Low-penetrance genes and their involvement in colorectal cancer susceptibility. Cancer Epidemiol. Biomark. Prev. 2002, 11, 1332–1352. [Google Scholar]
  87. Matsuo, K.; Hamajima, N.; Hirai, T.; Kato, T.; Inoue, M.; Takezaki, T.; Tajima, K. Methionine Synthase Reductase Gene A66G Polymorphism is Associated with Risk of Colorectal Cancer. Asian Pac. J. Cancer Prev. 2002, 3, 353–359. [Google Scholar]
  88. Robien, K.; Ulrich, C.M. 5,10-Methylenetetrahydrofolate reductase polymorphisms and leukemia risk: A HuGE minireview. Am. J. Epidemiol. 2003, 157, 571–582. [Google Scholar] [CrossRef] [Green Version]
  89. Krajinovic, M.; Lamothe, S.; Labuda, D.; Lemieux-Blanchard, E.; Theoret, Y.; Moghrabi, A.; Sinnett, D. Role of MTHFR genetic polymorphisms in the susceptibility to childhood acute lymphoblastic leukemia. Blood 2004, 103, 252–257. [Google Scholar] [CrossRef] [Green Version]
  90. Singal, R.; Ferdinand, L.; Das, P.M.; Reis, I.M.; Schlesselman, J.J. Polymorphisms in the methylenetetrahydrofolate reductase gene and prostate cancer risk. Int. J. Oncol. 2004, 25, 1465–1471. [Google Scholar] [CrossRef]
  91. Matsuo, K.; Ito, H.; Wakai, K.; Hirose, K.; Saito, T.; Suzuki, T.; Kato, T.; Hirai, T.; Kanemitsu, Y.; Hamajima, H.; et al. One-carbon metabolism related gene polymorphisms interact with alcohol drinking to influence the risk of colorectal cancer in Japan. Carcinogenesis 2005, 26, 2164–2171. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  92. Wu, L.L.; Wu, J.T. Hyperhomocysteinemia is a risk factor for cancer and a new potential tumor marker. Clin. Chim. Acta 2002, 322, 21–28. [Google Scholar] [CrossRef] [PubMed]
  93. Ueland, P.M.; Refsum, H. Plasma homocysteine, a risk factor for vascular disease: Plasma levels in health, disease, and drug therapy. J. Lab. Clin. Med. 1989, 114, 473–501. [Google Scholar]
  94. Corona, G.; Toffoli, G.; Fabris, M.; Viel, A.; Zarrelli, A.; Donada, C.; Boiocchi, M. Homocysteine accumulation in human ovarian carcinoma ascitic/cystic fluids possibly caused by metabolic alteration of the methionine cycle in ovarian carcinoma cells. Eur. J. Cancer 1997, 33, 1284–1290. [Google Scholar] [CrossRef] [PubMed]
  95. Santotoribio, J.D.; Cañavate-Solano, C.; Garcia-de la Torre, A.; Del Valle-Vazquez, L.; Arce-Matute, F.; Cuadros-Muñoz, J.F.; Sanchez del Pino, M.J.; Bandez-Ruiz, M.J.; Piñuela-Rojas, C.; Perez-Ramos, S. Homocysteine: New tumor marker in pleural fluid. Tumor Biol. 2015, 36, 7941–7945. [Google Scholar] [CrossRef]
  96. Shujuan, Y.; Jianxing, Z.; Xin-Yue, C. Methylenetetrahydrofolate reductase genetic polymorphisms and esophageal squamous cell carcinoma susceptibility: A meta-analysis of case-control studies. Pak. J. Med. Sci. 2013, 29, 693–698. [Google Scholar] [PubMed]
  97. Marugame, T.; Tsuji, E.; Inoue, H.; Shinomiya, S.; Kiyohara, C.; Onuma, K.; Hamada, H.; Koga, H.; Handa, K.; Hayabuchi, H.; et al. Methylenetetrahydrofolate reductase polymorphism and risk of colorectal adenomas. Cancer Lett. 2000, 151, 181–186. [Google Scholar] [CrossRef]
  98. Paynter, R.A.; Hankinson, S.E.; Hunter, D.J.; De Vivo, I. No association between MTHFR 677 C->T or 1298 A->C polymorphisms and endometrial cancer risk. Cancer Epidemiol. Biomark. Prev. 2004, 13, 1088–1089. [Google Scholar] [CrossRef]
  99. Safarinejad, M.R.; Shafiei, N.; Safarinejad, S. Relationship between three polymorphisms of methylenetetrahydrofolate reductase (MTHFR C677T, A1298C, and G1793A) gene and risk of prostate cancer: A case-control study. Prostate 2010, 70, 1645–1657. [Google Scholar] [CrossRef]
  100. Fang, D.H.; Ji, Q.; Fan, C.H.; An, Q.; Li, J. Methionine synthase reductase A66G polymorphism and leukemia risk: Evidence from published studies. Leuk. Lymphoma 2014, 55, 1910–1914. [Google Scholar] [CrossRef]
  101. Wang, P.; Li, S.; Wang, M.; He, J.; Xi, S. Association of MTRR A66G polymorphism with cancer susceptibility: Evidence from 85 studies. J. Cancer 2017, 8, 266–277. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  102. Wu, P.P.; Tang, R.N.; An, L. A meta-analysis of MTRR A66G polymorphism and colorectal cancer susceptibility. J. BUON 2015, 20, 918–922. [Google Scholar] [PubMed]
  103. Yoo, J.Y.; Kim, S.Y.; Hwang, J.A.; Hong, S.H.; Shin, A.; Choi, I.J.; Lee, Y.S. Association Study between Folate Pathway Gene Single Nucleotide Polymorphisms and Gastric Cancer in Koreans. Genom. Inform. 2012, 10, 184–193. [Google Scholar] [CrossRef] [PubMed]
  104. Wu, X.; Zou, T.; Cao, N.; Ni, J.; Xu, W.; Zhou, T.; Wang, X. Plasma homocysteine levels and genetic polymorphisms in folate metablism are associated with breast cancer risk in chinese women. Hered. Cancer Clin. Pract. 2014, 12, 2. [Google Scholar] [CrossRef] [Green Version]
  105. Cui, L.H.; Song, Y.; Si, H.; Shen, F.; Shin, M.H.; Kim, H.N.; Choi, J.S. Folate metabolism-related gene polymorphisms and susceptibility to primary liver cancer in North China. Med. Oncol. 2012, 29, 1837–1842. [Google Scholar] [CrossRef]
  106. Tao, M.H.; Shields, P.G.; Nie, J.; Marian, C.; Ambrosone, C.B.; McCann, S.E.; Platek, M.; Krishnan, S.S.; Xie, B.; Edge, S.B.; et al. DNA promoter methylation in breast tumors: No association with genetic polymorphisms in MTHFR and MTR. Cancer Epidemiol. Biomark. Prev. 2009, 18, 998–1002. [Google Scholar] [CrossRef] [Green Version]
  107. Semmler, A.; Simon, M.; Moskau, S.; Linnebank, M. The methionine synthase polymorphism c.2756A>G alters susceptibility to glioblastoma multiforme. Cancer Epidemiol. Biomark. Prev. 2006, 15, 2314–2316. [Google Scholar] [CrossRef] [Green Version]
  108. Ott, N.; Geddert, H.; Sarbia, M. Polymorphisms in methionine synthase (A2756G) and cystathionine beta-synthase (844ins68) and susceptibility to carcinomas of the upper gastrointestinal tract. J. Cancer Res. Clin. Oncol. 2008, 134, 405–410. [Google Scholar] [CrossRef]
  109. Zhao, Y.; Chen, Z.; Ma, Y.; Xia, Q.; Zhang, F.; Fu, D.; Wang, X.F. Lack of association between methionine synthase A2756G polymorphism and digestive system cancer risk: Evidence from 39327 subjects. PLoS ONE 2013, 8, e61511. [Google Scholar] [CrossRef] [Green Version]
  110. Wang, L.; Ke, Q.; Chen, W.; Wang, J.; Tan, Y.; Zhou, Y.; Hua, Z.; Ding, W.; Niu, J.; Shen, J.; et al. Polymorphisms of MTHFD, plasma homocysteine levels, and risk of gastric cancer in a high-risk Chinese population. Clin. Cancer Res. 2007, 13, 2526–2532. [Google Scholar] [CrossRef] [Green Version]
  111. Zhang, H.; Ma, H.; Li, L.; Zhang, Z.; Xu, Y. Association of methylenetetrahydrofolate dehydrogenase 1 polymorphisms with cancer: A meta-analysis. PLoS ONE 2013, 8, e69366. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  112. da Silva, L.M.; Galbiatti, A.L.; Ruiz, M.T.; Raposo, L.S.; Maniglia, J.V.; Pavarino, E.C.; Goloni-Bertollo, E.M. MTHFD1 G1958A, BHMT G742A, TC2 C776G and TC2 A67G polymorphisms and head and neck squamous cell carcinoma risk. Mol. Biol. Rep. 2012, 39, 887–893. [Google Scholar] [CrossRef] [PubMed]
  113. Xu, X.; Gammon, M.D.; Zeisel, S.H.; Lee, Y.L.; Wetmur, J.G.; Teitelbaum, S.L.; Bradshaw, P.T.; Neugut, A.I.; Santella, R.M.; Chen, J. Choline metabolism and risk of breast cancer in a population-based study. FASEB J. 2008, 22, 2045–2052. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  114. Mostowska, A.; Myka, M.; Lianeri, M.; Roszak, A.; Jagodziński, P.P. Folate and choline metabolism gene variants and development of uterine cervical carcinoma. Clin. Biochem. 2011, 44, 596–600. [Google Scholar] [CrossRef]
  115. Pawlik, P.; Mostowska, A.; Lianeri, M.; Sajdak, S.; Kędzia, H.; Jagodzinski, P.P. Folate and choline metabolism gene variants in relation to ovarian cancer risk in the Polish population. Mol. Biol. Rep. 2012, 39, 5553–5560. [Google Scholar] [CrossRef]
  116. Hazra, A.; Wu, K.; Kraft, P.; Fuchs, C.S.; Giovannucci, E.L.; Hunter, D.J. Twenty-four non-synonymous polymorphisms in the one-carbon metabolic pathway and risk of colorectal adenoma in the Nurses’ Health Study. Carcinogenesis 2007, 28, 1510–1519. [Google Scholar] [CrossRef] [Green Version]
  117. Kurzwelly, D.; Knop, S.; Guenther, M.; Loeffler, J.; Korfel, A.; Thiel, E.; Hebart, H.; Simon, M.; Weller, M.; Linnebank, M.; et al. Genetic variants of folate and methionine metabolism and PCNSL incidence in a German patient population. J. Neurooncol. 2010, 100, 187–192. [Google Scholar] [CrossRef] [Green Version]
  118. Gao, C.M.; Takezaki, T.; Wu, J.Z.; Liu, Y.T.; Ding, J.H.; Li, S.P.; Su, P.; Hu, X.; Kai, H.T.; Li, Z.Y.; et al. Polymorphisms in thymidylate synthase and methylenetetrahydrofolate reductase genes and the susceptibility to esophageal and stomach cancer with smoking. Asian Pac. J. Cancer Prev. 2004, 5, 133–138. [Google Scholar]
  119. Stathopoulou, A.; Vlachonikolis, I.; Mavroudis, D.; Perraki, M.; Kouroussis, C.; Apostolaki, S.; Malamos, N.; Kakolyris, S.; Kotsakis, A.; Xenidis, N.; et al. Molecular detection of cytokeratin-19-positive cells in the peripheral blood of patients with operable breast cancer: Evaluation of their prognostic significance. J. Clin. Oncol. 2002, 20, 3404–3412. [Google Scholar] [CrossRef]
  120. Refsum, H.; Nurk, E.; Smith, A.D.; Ueland, P.M.; Gjesdal, C.G.; Bjelland, I.; Tverdal, A.; Tell, G.S.; Nygård, O.; Vollset, S.E. The Hordaland Homocysteine Study: A community-based study of homocysteine, its determinants, and associations with disease. J. Nutr. 2006, 136, 1731s–1740s. [Google Scholar] [CrossRef] [Green Version]
  121. Gatt, A.; Makris, A.; Cladd, H.; Burcombe, R.J.; Smith, J.M.; Cooper, P.; Thompson, D.; Makris, M. Hyperhomocysteinemia in women with advanced breast cancer. Int. J. Lab. Hematol. 2007, 29, 421–425. [Google Scholar] [CrossRef] [PubMed]
  122. Goyette, P.; Sumner, J.S.; Milos, R.; Duncan, A.M.; Rosenblatt, D.S.; Matthews, R.G.; Rozen, R. Human methylenetetrahydrofolate reductase: Isolation of cDNA, mapping and mutation identification. Nat. Genet. 1994, 7, 195–200. [Google Scholar] [CrossRef]
  123. Goyette, P.; Frosst, P.; Rosenblatt, D.S.; Rozen, R. Seven novel mutations in the methylenetetrahydrofolate reductase gene and genotype/phenotype correlations in severe methylenetetrahydrofolate reductase deficiency. Am. J. Hum. Genet. 1995, 56, 1052–1059. [Google Scholar] [PubMed]
  124. Kluijtmans, L.A.; Wendel, U.; Stevens, E.M.; van den Heuvel, L.P.; Trijbels, F.J.; Blom, H.J. Identification of four novel mutations in severe methylenetetrahydrofolate reductase deficiency. Eur. J. Hum. Genet. 1998, 6, 257–265. [Google Scholar] [CrossRef]
  125. Weisberg, I.; Tran, P.; Christensen, B.; Sibani, S.; Rozen, R. A second genetic polymorphism in methylenetetrahydrofolate reductase (MTHFR) associated with decreased enzyme activity. Mol. Genet. Metab. 1998, 64, 169–172. [Google Scholar] [CrossRef]
  126. Sibani, S.; Christensen, B.; O’Ferrall, E.; Saadi, I.; Hiou-Tim, F.; Rosenblatt, D.S.; Rozen, R. Characterization of six novel mutations in the methylenetetrahydrofolate reductase (MTHFR) gene in patients with homocystinuria. Hum. Mutat. 2000, 15, 280–287. [Google Scholar] [CrossRef]
  127. Tonetti, C.; Amiel, J.; Munnich, A.; Zittoun, J. Impact of new mutations in the methylenetetrahydrofolate reductase gene assessed on biochemical phenotypes: A familial study. J. Inherit. Metab. Dis. 2001, 24, 833–842. [Google Scholar] [CrossRef]
  128. Yano, H.; Nakaso, K.; Yasui, K.; Wakutani, Y.; Nakayasu, H.; Kowa, H.; Adachi, Y.; Nakashima, K. Mutations of the MTHFR gene (428C>T and [458G>T+459C>T]) markedly decrease MTHFR enzyme activity. Neurogenetics 2004, 5, 135–140. [Google Scholar] [CrossRef]
  129. Ma, J.; Stampfer, M.J.; Giovannucci, E.; Artigas, C.; Hunter, D.J.; Fuchs, C.; Willett, W.C.; Selhub, J.; Hennekens, C.H.; Rozen, R. Methylenetetrahydrofolate reductase polymorphism, dietary interactions, and risk of colorectal cancer. Cancer Res. 1997, 57, 1098–1102. [Google Scholar]
  130. Esteller, M.; Garcia, A.; Martinez-Palones, J.M.; Xercavins, J.; Reventos, J. Germ line polymorphisms in cytochrome-P450 1A1 (C4887 CYP1A1) and methylenetetrahydrofolate reductase (MTHFR) genes and endometrial cancer susceptibility. Carcinogenesis 1997, 18, 2307–2311. [Google Scholar] [CrossRef] [Green Version]
  131. Song, C.; Xing, D.; Tan, W.; Wei, Q.; Lin, D. Methylenetetrahydrofolate reductase polymorphisms increase risk of esophageal squamous cell carcinoma in a Chinese population. Cancer Res. 2001, 61, 3272–3275. [Google Scholar] [PubMed]
  132. Krajinovic, M.; Lemieux-Blanchard, E.; Chiasson, S.; Primeau, M.; Costea, I.; Moghrabi, A. Role of polymorphisms in MTHFR and MTHFD1 genes in the outcome of childhood acute lymphoblastic leukemia. Pharmacogenom. J. 2004, 4, 66–72. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  133. Zhang, X.D.; Li, Y.T.; Yang, S.Y.; Li, W. Meta-analysis on MTHFR polymorphism and lung cancer susceptibility in East Asian populations. Biomed. Rep. 2013, 1, 440–446. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  134. Dick, D.M. Gene-environment interaction in psychological traits and disorders. Annu. Rev. Clin. Psychol. 2011, 7, 383–409. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  135. Ehrlich, M. DNA hypomethylation in cancer cells. Epigenomics 2009, 1, 239–259. [Google Scholar] [CrossRef] [Green Version]
  136. Martin, C.; Zhang, Y. The diverse functions of histone lysine methylation. Nat. Rev. Mol. Cell Biol. 2005, 6, 838–849. [Google Scholar] [CrossRef]
  137. Yuan, C.; Zhang, J.; Deng, C.; Xia, Y.; Li, B.; Meng, S.; Jin, X.; Cheng, L.; Li, H.; Zhang, C.; et al. Crosstalk of Histone and RNA Modifications Identified a Stromal-Activated Subtype with Poor Survival and Resistance to Immunotherapy in Gastric Cancer. Front. Pharmacol. 2022, 13, 868830. [Google Scholar] [CrossRef]
  138. Handy, D.E.; Castro, R.; Loscalzo, J. Epigenetic modifications: Basic mechanisms and role in cardiovascular disease. Circulation 2011, 123, 2145–2156. [Google Scholar] [CrossRef] [Green Version]
  139. Santos-Rebouças, C.B.; Pimentel, M.M. Implication of abnormal epigenetic patterns for human diseases. Eur. J. Hum. Genet. 2007, 15, 10–17. [Google Scholar] [CrossRef] [Green Version]
  140. Zhou, S.; Zhang, Z.; Xu, G. Notable epigenetic role of hyperhomocysteinemia in atherogenesis. Lipids Health Dis. 2014, 13, 134. [Google Scholar] [CrossRef] [Green Version]
  141. Jiang, L.; Gonda, T.A.; Gamble, M.V.; Salas, M.; Seshan, V.; Tu, S.; Twaddell, W.S.; Hegyi, P.; Lazar, G.; Steele, I.; et al. Global hypomethylation of genomic DNA in cancer-associated myofibroblasts. Cancer Res. 2008, 68, 9900–9908. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  142. Berchuck, J.E.; Baca, S.C.; McClure, H.M.; Korthauer, K.; Tsai, H.K.; Nuzzo, P.V.; Kelleher, K.M.; He, M.; Steinharter, J.A.; Zacharia, S.; et al. Detecting Neuroendocrine Prostate Cancer Through Tissue-Informed Cell-Free DNA Methylation Analysis. Clin. Cancer Res. 2022, 28, 928–938. [Google Scholar] [CrossRef] [PubMed]
  143. Cahill, N.; Rosenquist, R. Uncovering the DNA methylome in chronic lymphocytic leukemia. Epigenetics 2013, 8, 138–148. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  144. Zhao, P.; Malik, S.; Xing, S. Epigenetic Mechanisms Involved in HCV-Induced Hepatocellular Carcinoma (HCC). Front. Oncol. 2021, 11, 677926. [Google Scholar] [CrossRef] [PubMed]
  145. Ehrlich, M. DNA hypomethylation, cancer, the immunodeficiency, centromeric region instability, facial anomalies syndrome and chromosomal rearrangements. J. Nutr. 2002, 132, 2424s–2429s. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  146. Pappalardo, X.G.; Barra, V. Losing DNA methylation at repetitive elements and breaking bad. Epigenetics Chromatin 2021, 14, 25. [Google Scholar] [CrossRef]
  147. Burns, K.H. Transposable elements in cancer. Nat. Rev. Cancer 2017, 17, 415–424. [Google Scholar] [CrossRef]
  148. Zhang, N. Role of methionine on epigenetic modification of DNA methylation and gene expression in animals. Anim. Nutr. 2018, 4, 11–16. [Google Scholar] [CrossRef]
  149. Lund, G.; Andersson, L.; Lauria, M.; Lindholm, M.; Fraga, M.F.; Villar-Garea, A.; Ballestar, E.; Esteller, M.; Zaina, S. DNA methylation polymorphisms precede any histological sign of atherosclerosis in mice lacking apolipoprotein E. J. Biol. Chem. 2004, 279, 29147–29154. [Google Scholar] [CrossRef] [Green Version]
  150. Devlin, A.M.; Arning, E.; Bottiglieri, T.; Faraci, F.M.; Rozen, R.; Lentz, S.R. Effect of Mthfr genotype on diet-induced hyperhomocysteinemia and vascular function in mice. Blood 2004, 103, 2624–2629. [Google Scholar] [CrossRef] [Green Version]
  151. Chaturvedi, P.; Kalani, A.; Givvimani, S.; Kamat, P.K.; Familtseva, A.; Tyagi, S.C. Differential regulation of DNA methylation versus histone acetylation in cardiomyocytes during HHcy in vitro and in vivo: An epigenetic mechanism. Physiol. Genom. 2014, 46, 245–255. [Google Scholar] [CrossRef]
  152. Izadi, P.; Noruzinia, M.; Karimipoor, M.; Karbassian, M.H.; Akbari, M.T. Promoter hypermethylation of estrogen receptor alpha gene is correlated to estrogen receptor negativity in Iranian patients with sporadic breast cancer. Cell J. 2012, 14, 102–109. [Google Scholar]
  153. Zhang, J.G.; Liu, J.X.; Li, Z.H.; Wang, L.Z.; Jiang, Y.D.; Wang, S.R. Dysfunction of endothelial NO system originated from homocysteine-induced aberrant methylation pattern in promoter region of DDAH2 gene. Chin. Med. J. 2007, 120, 2132–2137. [Google Scholar] [CrossRef]
  154. Lister, R.; Pelizzola, M.; Dowen, R.H.; Hawkins, R.D.; Hon, G.; Tonti-Filippini, J.; Nery, J.R.; Lee, L.; Ye, Z.; Ngo, Q.M.; et al. Human DNA methylomes at base resolution show widespread epigenomic differences. Nature 2009, 462, 315–322. [Google Scholar] [CrossRef] [Green Version]
  155. Mariño-Ramírez, L.; Kann, M.G.; Shoemaker, B.A.; Landsman, D. Histone structure and nucleosome stability. Expert Rev. Proteom. 2005, 2, 719–729. [Google Scholar] [CrossRef] [PubMed]
  156. Millán-Zambrano, G.; Burton, A.; Bannister, A.J.; Schneider, R. Histone post-translational modifications—Cause and consequence of genome function. Nat. Rev. Genet. 2022, 23, 563–580. [Google Scholar] [CrossRef] [PubMed]
  157. Ouyang, Y.; Wu, Q.; Li, J.; Sun, S.; Sun, S. S-adenosylmethionine: A metabolite critical to the regulation of autophagy. Cell Prolif. 2020, 53, e12891. [Google Scholar] [CrossRef] [PubMed]
  158. Helin, K.; Dhanak, D. Chromatin proteins and modifications as drug targets. Nature 2013, 502, 480–488. [Google Scholar] [CrossRef] [Green Version]
  159. Garraway, L.A.; Lander, E.S. Lessons from the cancer genome. Cell 2013, 153, 17–37. [Google Scholar] [CrossRef] [Green Version]
  160. Halsall, J.A.; Andrews, S.; Krueger, F.; Rutledge, C.E.; Ficz, G.; Reik, W.; Turner, B.M. Histone modifications form a cell-type-specific chromosomal bar code that persists through the cell cycle. Sci. Rep. 2021, 11, 3009. [Google Scholar] [CrossRef]
  161. Yang, J.X.; Rastetter, R.H.; Wilhelm, D. Non-coding RNAs: An Introduction. Adv. Exp. Med. Biol. 2016, 886, 13–32. [Google Scholar] [CrossRef]
  162. Slack, F.J.; Chinnaiyan, A.M. The Role of Non-coding RNAs in Oncology. Cell 2019, 179, 1033–1055. [Google Scholar] [CrossRef]
  163. Ratti, M.; Lampis, A.; Ghidini, M.; Salati, M.; Mirchev, M.B.; Valeri, N.; Hahne, J.C. MicroRNAs (miRNAs) and Long Non-Coding RNAs (lncRNAs) as New Tools for Cancer Therapy: First Steps from Bench to Bedside. Target. Oncol. 2020, 15, 261–278. [Google Scholar] [CrossRef] [PubMed]
  164. Volovat, S.R.; Volovat, C.; Hordila, I.; Hordila, D.A.; Mirestean, C.C.; Miron, O.T.; Lungulescu, C.; Scripcariu, D.V.; Stolniceanu, C.R.; Konsoulova-Kirova, A.A.; et al. MiRNA and LncRNA as Potential Biomarkers in Triple-Negative Breast Cancer: A Review. Front. Oncol. 2020, 10, 526850. [Google Scholar] [CrossRef] [PubMed]
  165. Kalani, A.; Kamat, P.K.; Tyagi, S.C.; Tyagi, N. Synergy of homocysteine, microRNA, and epigenetics: A novel therapeutic approach for stroke. Mol. Neurobiol. 2013, 48, 157–168. [Google Scholar] [CrossRef] [PubMed]
  166. George, A.K.; Master, K.; Majumder, A.; Homme, R.P.; Laha, A.; Sandhu, H.S.; Tyagi, S.C.; Singh, M. Circular RNAs constitute an inherent gene regulatory axis in the mammalian eye and brain. Can. J. Physiol. Pharmacol. 2019, 97, 463–472. [Google Scholar] [CrossRef]
  167. Singh, M.; George, A.K.; Homme, R.P.; Majumder, A.; Laha, A.; Sandhu, H.S.; Tyagi, S.C. Circular RNAs profiling in the cystathionine-β-synthase mutant mouse reveals novel gene targets for hyperhomocysteinemia induced ocular disorders. Exp. Eye Res. 2018, 174, 80–92. [Google Scholar] [CrossRef] [PubMed]
  168. George, A.K.; Homme, R.P.; Majumder, A.; Tyagi, S.C.; Singh, M. Effect of MMP-9 gene knockout on retinal vascular form and function. Physiol. Genom. 2019, 51, 613–622. [Google Scholar] [CrossRef]
  169. Homme, R.P.; Singh, M.; Majumder, A.; George, A.K.; Nair, K.; Sandhu, H.S.; Tyagi, N.; Lominadze, D.; Tyagi, S.C. Remodeling of Retinal Architecture in Diabetic Retinopathy: Disruption of Ocular Physiology and Visual Functions by Inflammatory Gene Products and Pyroptosis. Front. Physiol. 2018, 9, 1268. [Google Scholar] [CrossRef] [Green Version]
  170. Liu, X.; Tong, Y.; Xia, D.; Peng, E.; Yang, X.; Liu, H.; Ye, T.; Wang, X.; He, Y.; Ye, Z.; et al. Circular RNAs in prostate cancer: Biogenesis, biological functions, and clinical significance. Mol. Ther.-Nucleic Acids 2021, 26, 1130–1147. [Google Scholar] [CrossRef]
  171. Singh, M.; George, A.K.; Homme, R.P.; Majumder, A.; Laha, A.; Sandhu, H.S.; Tyagi, S.C. Expression Analysis of the Circular RNA Molecules in the Human Retinal Cells Treated with Homocysteine. Curr. Eye Res. 2019, 44, 287–293. [Google Scholar] [CrossRef] [PubMed]
  172. George, A.K.; Homme, R.P.; Majumder, A.; Laha, A.; Metreveli, N.; Sandhu, H.S.; Tyagi, S.C.; Singh, M. Hydrogen sulfide intervention in cystathionine-β-synthase mutant mouse helps restore ocular homeostasis. Int. J. Ophthalmol. 2019, 12, 754–764. [Google Scholar] [CrossRef] [PubMed]
  173. Shackelford, R.E.; Mohammad, I.Z.; Meram, A.T.; Kim, D.; Alotaibi, F.; Patel, S.; Ghali, G.E.; Kevil, C.G. Molecular Functions of Hydrogen Sulfide in Cancer. Pathophysiology 2021, 28, 437–456. [Google Scholar] [CrossRef] [PubMed]
  174. Kimura, H. Hydrogen sulfide as a neuromodulator. Mol. Neurobiol. 2002, 26, 13–19. [Google Scholar] [CrossRef]
  175. Guo, H.; Gai, J.W.; Wang, Y.; Jin, H.F.; Du, J.B.; Jin, J. Characterization of hydrogen sulfide and its synthases, cystathionine β-synthase and cystathionine γ-lyase, in human prostatic tissue and cells. Urology 2012, 79, 483.e1–483.e5. [Google Scholar] [CrossRef] [PubMed]
  176. Bhattacharyya, S.; Saha, S.; Giri, K.; Lanza, I.R.; Nair, K.S.; Jennings, N.B.; Rodriguez-Aguayo, C.; Lopez-Berestein, G.; Basal, E.; Weaver, A.L.; et al. Cystathionine beta-synthase (CBS) contributes to advanced ovarian cancer progression and drug resistance. PLoS ONE 2013, 8, e79167. [Google Scholar] [CrossRef]
  177. Arfin, S.; Jha, N.K.; Jha, S.K.; Kesari, K.K.; Ruokolainen, J.; Roychoudhury, S.; Rathi, B.; Kumar, D. Oxidative Stress in Cancer Cell Metabolism. Antioxidants 2021, 10, 642. [Google Scholar] [CrossRef] [PubMed]
  178. Li, S.; Xu, H.X.; Wu, C.T.; Wang, W.Q.; Jin, W.; Gao, H.L.; Li, H.; Zhang, S.R.; Xu, J.Z.; Qi, Z.H.; et al. Angiogenesis in pancreatic cancer: Current research status and clinical implications. Angiogenesis 2019, 22, 15–36. [Google Scholar] [CrossRef]
  179. Cai, W.J.; Wang, M.J.; Ju, L.H.; Wang, C.; Zhu, Y.C. Hydrogen sulfide induces human colon cancer cell proliferation: Role of Akt, ERK and p21. Cell Biol. Int. 2010, 34, 565–572. [Google Scholar] [CrossRef]
  180. Untereiner, A.A.; Pavlidou, A.; Druzhyna, N.; Papapetropoulos, A.; Hellmich, M.R.; Szabo, C. Drug resistance induces the upregulation of H2S-producing enzymes in HCT116 colon cancer cells. Biochem. Pharmacol. 2018, 149, 174–185. [Google Scholar] [CrossRef]
  181. Shibuya, N.; Mikami, Y.; Kimura, Y.; Nagahara, N.; Kimura, H. Vascular endothelium expresses 3-mercaptopyruvate sulfurtransferase and produces hydrogen sulfide. J. Biochem. 2009, 146, 623–626. [Google Scholar] [CrossRef]
  182. Szabo, C.; Coletta, C.; Chao, C.; Módis, K.; Szczesny, B.; Papapetropoulos, A.; Hellmich, M.R. Tumor-derived hydrogen sulfide, produced by cystathionine-β-synthase, stimulates bioenergetics, cell proliferation, and angiogenesis in colon cancer. Proc. Natl. Acad. Sci. USA 2013, 110, 12474–12479. [Google Scholar] [CrossRef] [PubMed]
  183. Sen, S.; Kawahara, B.; Gupta, D.; Tsai, R.; Khachatryan, M.; Roy-Chowdhuri, S.; Bose, S.; Yoon, A.; Faull, K.; Farias-Eisner, R.; et al. Role of cystathionine β-synthase in human breast Cancer. Free Radic. Biol. Med. 2015, 86, 228–238. [Google Scholar] [CrossRef] [PubMed]
  184. Turbat-Herrera, E.A.; Kilpatrick, M.J.; Chen, J.; Meram, A.T.; Cotelingam, J.; Ghali, G.; Kevil, C.G.; Coppola, D.; Shackelford, R.E. Cystathione β-Synthase Is Increased in Thyroid Malignancies. Anticancer Res. 2018, 38, 6085–6090. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  185. Li, D.; Yang, Z.; Liu, Z.; Zou, Q.; Yuan, Y. Clinical Significance of CBS and CCL21 in Gallbladder Adenocarcinomas and Squamous Cell/Adenosquamous Carcinomas. Appl. Immunohistochem. Mol. Morphol. 2020, 28, 103–110. [Google Scholar] [CrossRef]
  186. Kim, J.; Hong, S.J.; Park, J.H.; Park, S.Y.; Kim, S.W.; Cho, E.Y.; Do, I.G.; Joh, J.W.; Kim, D.S. Expression of cystathionine beta-synthase is downregulated in hepatocellular carcinoma and associated with poor prognosis. Oncol. Rep. 2009, 21, 1449–1454. [Google Scholar] [CrossRef]
  187. Zhao, H.; Li, Q.; Wang, J.; Su, X.; Ng, K.M.; Qiu, T.; Shan, L.; Ling, Y.; Wang, L.; Cai, J.; et al. Frequent epigenetic silencing of the folate-metabolising gene cystathionine-beta-synthase in gastrointestinal cancer. PLoS ONE 2012, 7, e49683. [Google Scholar] [CrossRef]
  188. You, J.; Shi, X.; Liang, H.; Ye, J.; Wang, L.; Han, H.; Fang, H.; Kang, W.; Wang, T. Cystathionine-γ-lyase promotes process of breast cancer in association with STAT3 signaling pathway. Oncotarget 2017, 8, 65677–65686. [Google Scholar] [CrossRef] [Green Version]
  189. Pei, Y.; Wu, B.; Cao, Q.; Wu, L.; Yang, G. Hydrogen sulfide mediates the anti-survival effect of sulforaphane on human prostate cancer cells. Toxicol. Appl. Pharmacol. 2011, 257, 420–428. [Google Scholar] [CrossRef]
  190. Zhang, L.; Qi, Q.; Yang, J.; Sun, D.; Li, C.; Xue, Y.; Jiang, Q.; Tian, Y.; Xu, C.; Wang, R. An Anticancer Role of Hydrogen Sulfide in Human Gastric Cancer Cells. Oxid. Med. Cell. Longev. 2015, 2015, 636410. [Google Scholar] [CrossRef] [Green Version]
  191. Gai, J.W.; Qin, W.; Liu, M.; Wang, H.F.; Zhang, M.; Li, M.; Zhou, W.H.; Ma, Q.T.; Liu, G.M.; Song, W.H.; et al. Expression profile of hydrogen sulfide and its synthases correlates with tumor stage and grade in urothelial cell carcinoma of bladder. Urol. Oncol. 2016, 34, 166.e15. [Google Scholar] [CrossRef] [PubMed]
  192. Pan, Y.; Ye, S.; Yuan, D.; Zhang, J.; Bai, Y.; Shao, C. Hydrogen sulfide (H2S)/cystathionine γ-lyase (CSE) pathway contributes to the proliferation of hepatoma cells. Mutat. Res. 2014, 763–764, 10–18. [Google Scholar] [CrossRef] [PubMed]
  193. Fan, K.; Li, N.; Qi, J.; Yin, P.; Zhao, C.; Wang, L.; Li, Z.; Zha, X. Wnt/β-catenin signaling induces the transcription of cystathionine-γ-lyase, a stimulator of tumor in colon cancer. Cell. Signal. 2014, 26, 2801–2808. [Google Scholar] [CrossRef]
  194. Breza, J., Jr.; Soltysova, A.; Hudecova, S.; Penesova, A.; Szadvari, I.; Babula, P.; Chovancova, B.; Lencesova, L.; Pos, O.; Breza, J.; et al. Endogenous H2S producing enzymes are involved in apoptosis induction in clear cell renal cell carcinoma. BMC Cancer 2018, 18, 591. [Google Scholar] [CrossRef]
  195. Wróbel, M.; Czubak, J.; Bronowicka-Adamska, P.; Jurkowska, H.; Adamek, D.; Papla, B. Is development of high-grade gliomas sulfur-dependent? Molecules 2014, 19, 21350–21362. [Google Scholar] [CrossRef] [Green Version]
  196. Oláh, G.; Módis, K.; Törö, G.; Hellmich, M.R.; Szczesny, B.; Szabo, C. Role of endogenous and exogenous nitric oxide, carbon monoxide and hydrogen sulfide in HCT116 colon cancer cell proliferation. Biochem. Pharmacol. 2018, 149, 186–204. [Google Scholar] [CrossRef] [PubMed]
  197. Cao, X.; Ding, L.; Xie, Z.Z.; Yang, Y.; Whiteman, M.; Moore, P.K.; Bian, J.S. A Review of Hydrogen Sulfide Synthesis, Metabolism, and Measurement: Is Modulation of Hydrogen Sulfide a Novel Therapeutic for Cancer? Antioxid. Redox Signal. 2019, 31, 1–38. [Google Scholar] [CrossRef] [Green Version]
  198. Tu, X.H.; Huang, S.X.; Li, W.S.; Song, J.X. Correlation of methylation of CpG island in cystathionine beta synthase promoter and clinicopathological features in colorectal cancer. Zhonghua Zhong Liu Za Zhi 2013, 35, 351–355. [Google Scholar] [CrossRef]
  199. Módis, K.; Coletta, C.; Asimakopoulou, A.; Szczesny, B.; Chao, C.; Papapetropoulos, A.; Hellmich, M.R.; Szabo, C. Effect of S-adenosyl-L-methionine (SAM), an allosteric activator of cystathionine-β-synthase (CBS) on colorectal cancer cell proliferation and bioenergetics in vitro. Nitric Oxide 2014, 41, 146–156. [Google Scholar] [CrossRef] [Green Version]
  200. Niu, W.; Wang, J.; Qian, J.; Wang, M.; Wu, P.; Chen, F.; Yan, S. Allosteric control of human cystathionine β-synthase activity by a redox active disulfide bond. J. Biol. Chem. 2018, 293, 2523–2533. [Google Scholar] [CrossRef] [Green Version]
  201. Takano, N.; Sarfraz, Y.; Gilkes, D.M.; Chaturvedi, P.; Xiang, L.; Suematsu, M.; Zagzag, D.; Semenza, G.L. Decreased expression of cystathionine β-synthase promotes glioma tumorigenesis. Mol. Cancer Res. 2014, 12, 1398–1406. [Google Scholar] [CrossRef] [Green Version]
  202. Kimura, Y.; Toyofuku, Y.; Koike, S.; Shibuya, N.; Nagahara, N.; Lefer, D.; Ogasawara, Y.; Kimura, H. Identification of H2S3 and H2S produced by 3-mercaptopyruvate sulfurtransferase in the brain. Sci. Rep. 2015, 5, 14774. [Google Scholar] [CrossRef] [Green Version]
  203. Mustafa, A.K.; Gadalla, M.M.; Sen, N.; Kim, S.; Mu, W.; Gazi, S.K.; Barrow, R.K.; Yang, G.; Wang, R.; Snyder, S.H. H2S signals through protein S-sulfhydration. Sci. Signal. 2009, 2, ra72. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  204. d’Emmanuele di Villa Bianca, R.; Mitidieri, E.; Esposito, D.; Donnarumma, E.; Russo, A.; Fusco, F.; Ianaro, A.; Mirone, V.; Cirino, G.; Russo, G.; et al. Human Cystathionine-β-Synthase Phosphorylation on Serine227 Modulates Hydrogen Sulfide Production in Human Urothelium. PLoS ONE 2015, 10, e0136859. [Google Scholar] [CrossRef] [Green Version]
  205. Sbodio, J.I.; Snyder, S.H.; Paul, B.D. Regulators of the transsulfuration pathway. Br. J. Pharmacol. 2019, 176, 583–593. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  206. Murphy, B.; Bhattacharya, R.; Mukherjee, P. Hydrogen sulfide signaling in mitochondria and disease. FASEB J. 2019, 33, 13098–13125. [Google Scholar] [CrossRef] [Green Version]
  207. Hourihan, J.M.; Kenna, J.G.; Hayes, J.D. The gasotransmitter hydrogen sulfide induces nrf2-target genes by inactivating the keap1 ubiquitin ligase substrate adaptor through formation of a disulfide bond between cys-226 and cys-613. Antioxid. Redox Signal. 2013, 19, 465–481. [Google Scholar] [CrossRef]
  208. Yang, G.; Pei, Y.; Teng, H.; Cao, Q.; Wang, R. Specificity protein-1 as a critical regulator of human cystathionine gamma-lyase in smooth muscle cells. J. Biol. Chem. 2011, 286, 26450–26460. [Google Scholar] [CrossRef] [Green Version]
  209. Sen, N.; Paul, B.D.; Gadalla, M.M.; Mustafa, A.K.; Sen, T.; Xu, R.; Kim, S.; Snyder, S.H. Hydrogen sulfide-linked sulfhydration of NF-κB mediates its antiapoptotic actions. Mol. Cell 2012, 45, 13–24. [Google Scholar] [CrossRef] [Green Version]
  210. Wang, Y.H.; Huang, J.T.; Chen, W.L.; Wang, R.H.; Kao, M.C.; Pan, Y.R.; Chan, S.H.; Tsai, K.W.; Kung, H.J.; Lin, K.T.; et al. Dysregulation of cystathionine γ-lyase promotes prostate cancer progression and metastasis. EMBO Rep. 2019, 20, e45986. [Google Scholar] [CrossRef]
  211. Nagahara, N.; Katayama, A. Post-translational regulation of mercaptopyruvate sulfurtransferase via a low redox potential cysteine-sulfenate in the maintenance of redox homeostasis. J. Biol. Chem. 2005, 280, 34569–34576. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  212. Augsburger, F.; Randi, E.B.; Jendly, M.; Ascencao, K.; Dilek, N.; Szabo, C. Role of 3-Mercaptopyruvate Sulfurtransferase in the Regulation of Proliferation, Migration, and Bioenergetics in Murine Colon Cancer Cells. Biomolecules 2020, 10, 447. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  213. Hayes, J.D.; Dinkova-Kostova, A.T.; Tew, K.D. Oxidative Stress in Cancer. Cancer Cell 2020, 38, 167–197. [Google Scholar] [CrossRef]
  214. Whiteman, M.; Armstrong, J.S.; Chu, S.H.; Jia-Ling, S.; Wong, B.S.; Cheung, N.S.; Halliwell, B.; Moore, P.K. The novel neuromodulator hydrogen sulfide: An endogenous peroxynitrite ‘scavenger’? J. Neurochem. 2004, 90, 765–768. [Google Scholar] [CrossRef] [PubMed]
  215. Whiteman, M.; Cheung, N.S.; Zhu, Y.Z.; Chu, S.H.; Siau, J.L.; Wong, B.S.; Armstrong, J.S.; Moore, P.K. Hydrogen sulphide: A novel inhibitor of hypochlorous acid-mediated oxidative damage in the brain? Biochem. Biophys. Res. Commun. 2005, 326, 794–798. [Google Scholar] [CrossRef]
  216. Yan, S.K.; Chang, T.; Wang, H.; Wu, L.; Wang, R.; Meng, Q.H. Effects of hydrogen sulfide on homocysteine-induced oxidative stress in vascular smooth muscle cells. Biochem. Biophys. Res. Commun. 2006, 351, 485–491. [Google Scholar] [CrossRef]
  217. Greiner, R.; Palinkas, Z.; Basell, K.; Becher, D.; Antelmann, H.; Nagy, P.; Dick, T.P. Polysulfides link H2S to protein thiol oxidation. Antioxid. Redox Signal. 2013, 19, 1749–1765. [Google Scholar] [CrossRef] [Green Version]
  218. Szabo, C. Hydrogen sulphide and its therapeutic potential. Nat. Rev. Drug Discov. 2007, 6, 917–935. [Google Scholar] [CrossRef]
  219. Al-Magableh, M.R.; Kemp-Harper, B.K.; Ng, H.H.; Miller, A.A.; Hart, J.L. Hydrogen sulfide protects endothelial nitric oxide function under conditions of acute oxidative stress in vitro. Naunyn-Schmiedeberg’s Arch. Pharmacol. 2014, 387, 67–74. [Google Scholar] [CrossRef]
  220. Predmore, B.L.; Lefer, D.J.; Gojon, G. Hydrogen sulfide in biochemistry and medicine. Antioxid. Redox Signal. 2012, 17, 119–140. [Google Scholar] [CrossRef] [Green Version]
  221. Whiteman, M.; Li, L.; Kostetski, I.; Chu, S.H.; Siau, J.L.; Bhatia, M.; Moore, P.K. Evidence for the formation of a novel nitrosothiol from the gaseous mediators nitric oxide and hydrogen sulphide. Biochem. Biophys. Res. Commun. 2006, 343, 303–310. [Google Scholar] [CrossRef] [PubMed]
  222. Geng, B.; Chang, L.; Pan, C.; Qi, Y.; Zhao, J.; Pang, Y.; Du, J.; Tang, C. Endogenous hydrogen sulfide regulation of myocardial injury induced by isoproterenol. Biochem. Biophys. Res. Commun. 2004, 318, 756–763. [Google Scholar] [CrossRef] [PubMed]
  223. Kimura, Y.; Kimura, H. Hydrogen sulfide protects neurons from oxidative stress. FASEB J. 2004, 18, 1165–1167. [Google Scholar] [CrossRef] [PubMed]
  224. Lu, M.; Hu, L.F.; Hu, G.; Bian, J.S. Hydrogen sulfide protects astrocytes against H2O2-induced neural injury via enhancing glutamate uptake. Free Radic. Biol. Med. 2008, 45, 1705–1713. [Google Scholar] [CrossRef]
  225. Tyagi, N.; Moshal, K.S.; Sen, U.; Vacek, T.P.; Kumar, M.; Hughes, W.M., Jr.; Kundu, S.; Tyagi, S.C. H2S protects against methionine-induced oxidative stress in brain endothelial cells. Antioxid. Redox Signal. 2009, 11, 25–33. [Google Scholar] [CrossRef] [Green Version]
  226. Kesherwani, V.; Nelson, K.S.; Agrawal, S.K. Effect of sodium hydrosulphide after acute compression injury of spinal cord. Brain Res. 2013, 1527, 222–229. [Google Scholar] [CrossRef]
  227. Huang, C.; Kan, J.; Liu, X.; Ma, F.; Tran, B.H.; Zou, Y.; Wang, S.; Zhu, Y.Z. Cardioprotective effects of a novel hydrogen sulfide agent-controlled release formulation of S-propargyl-cysteine on heart failure rats and molecular mechanisms. PLoS ONE 2013, 8, e69205. [Google Scholar] [CrossRef] [Green Version]
  228. Wang, C.; Wang, H.Y.; Liu, Z.W.; Fu, Y.; Zhao, B. Effect of endogenous hydrogen sulfide on oxidative stress in oleic acid-induced acute lung injury in rats. Chin. Med. J. 2011, 124, 3476–3480. [Google Scholar]
  229. Sen, U.; Basu, P.; Abe, O.A.; Givvimani, S.; Tyagi, N.; Metreveli, N.; Shah, K.S.; Passmore, J.C.; Tyagi, S.C. Hydrogen sulfide ameliorates hyperhomocysteinemia-associated chronic renal failure. Am. J. Physiol. Ren. Physiol. 2009, 297, F410–F419. [Google Scholar] [CrossRef] [Green Version]
  230. Guo, C.; Liang, F.; Shah Masood, W.; Yan, X. Hydrogen sulfide protected gastric epithelial cell from ischemia/reperfusion injury by Keap1 s-sulfhydration, MAPK dependent anti-apoptosis and NF-kappaB dependent anti-inflammation pathway. Eur. J. Pharmacol. 2014, 725, 70–78. [Google Scholar] [CrossRef]
  231. Cui, J.; Liu, L.; Zou, J.; Qiao, W.; Liu, H.; Qi, Y.; Yan, C. Protective effect of endogenous hydrogen sulfide against oxidative stress in gastric ischemia-reperfusion injury. Exp. Ther. Med. 2013, 5, 689–694. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  232. Zhu, X.; Tang, Z.; Cong, B.; Du, J.; Wang, C.; Wang, L.; Ni, X.; Lu, J. Estrogens increase cystathionine-gamma-lyase expression and decrease inflammation and oxidative stress in the myocardium of ovariectomized rats. Menopause 2013, 20, 1084–1091. [Google Scholar] [CrossRef] [PubMed]
  233. Fu, Z.; Liu, X.; Geng, B.; Fang, L.; Tang, C. Hydrogen sulfide protects rat lung from ischemia-reperfusion injury. Life Sci. 2008, 82, 1196–1202. [Google Scholar] [CrossRef]
  234. Wen, Y.D.; Wang, H.; Kho, S.H.; Rinkiko, S.; Sheng, X.; Shen, H.M.; Zhu, Y.Z. Hydrogen sulfide protects HUVECs against hydrogen peroxide induced mitochondrial dysfunction and oxidative stress. PLoS ONE 2013, 8, e53147. [Google Scholar] [CrossRef] [Green Version]
  235. Liu, Y.Y.; Nagpure, B.V.; Wong, P.T.; Bian, J.S. Hydrogen sulfide protects SH-SY5Y neuronal cells against d-galactose induced cell injury by suppression of advanced glycation end products formation and oxidative stress. Neurochem. Int. 2013, 62, 603–609. [Google Scholar] [CrossRef] [PubMed]
  236. Benetti, L.R.; Campos, D.; Gurgueira, S.A.; Vercesi, A.E.; Guedes, C.E.; Santos, K.L.; Wallace, J.L.; Teixeira, S.A.; Florenzano, J.; Costa, S.K.; et al. Hydrogen sulfide inhibits oxidative stress in lungs from allergic mice in vivo. Eur. J. Pharmacol. 2013, 698, 463–469. [Google Scholar] [CrossRef] [Green Version]
  237. Su, Y.W.; Liang, C.; Jin, H.F.; Tang, X.Y.; Han, W.; Chai, L.J.; Zhang, C.Y.; Geng, B.; Tang, C.S.; Du, J.B. Hydrogen sulfide regulates cardiac function and structure in adriamycin-induced cardiomyopathy. Circ. J. 2009, 73, 741–749. [Google Scholar] [CrossRef] [Green Version]
  238. Kai, S.; Tanaka, T.; Daijo, H.; Harada, H.; Kishimoto, S.; Suzuki, K.; Takabuchi, S.; Takenaga, K.; Fukuda, K.; Hirota, K. Hydrogen sulfide inhibits hypoxia- but not anoxia-induced hypoxia-inducible factor 1 activation in a von hippel-lindau- and mitochondria-dependent manner. Antioxid. Redox Signal. 2012, 16, 203–216. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  239. Wang, M.; Guo, Z.; Wang, S. Regulation of cystathionine γ-lyase in mammalian cells by hypoxia. Biochem. Genet. 2014, 52, 29–37. [Google Scholar] [CrossRef]
  240. Giuffrè, A.; Tomé, C.S.; Fernandes, D.G.F.; Zuhra, K.; Vicente, J.B. Hydrogen Sulfide Metabolism and Signaling in the Tumor Microenvironment. Adv. Exp. Med. Biol. 2020, 1219, 335–353. [Google Scholar] [CrossRef]
  241. Zhou, Y.; Li, X.H.; Zhang, C.C.; Wang, M.J.; Xue, W.L.; Wu, D.D.; Ma, F.F.; Li, W.W.; Tao, B.B.; Zhu, Y.C. Hydrogen sulfide promotes angiogenesis by downregulating miR-640 via the VEGFR2/mTOR pathway. Am. J. Physiol. Cell Physiol. 2016, 310, C305–C317. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  242. Wang, M.; Yan, J.; Cao, X.; Hua, P.; Li, Z. Hydrogen sulfide modulates epithelial-mesenchymal transition and angiogenesis in non-small cell lung cancer via HIF-1α activation. Biochem. Pharmacol. 2020, 172, 113775. [Google Scholar] [CrossRef] [PubMed]
  243. Szabó, C.; Papapetropoulos, A. Hydrogen sulphide and angiogenesis: Mechanisms and applications. Br. J. Pharmacol. 2011, 164, 853–865. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  244. Wong, R.S. Apoptosis in cancer: From pathogenesis to treatment. J. Exp. Clin. Cancer Res. 2011, 30, 87. [Google Scholar] [CrossRef] [Green Version]
  245. Rose, P.; Moore, P.K.; Ming, S.H.; Nam, O.C.; Armstrong, J.S.; Whiteman, M. Hydrogen sulfide protects colon cancer cells from chemopreventative agent beta-phenylethyl isothiocyanate induced apoptosis. World J. Gastroenterol. 2005, 11, 3990–3997. [Google Scholar] [CrossRef] [PubMed]
  246. Zheng, D.; Chen, Z.; Chen, J.; Zhuang, X.; Feng, J.; Li, J. Exogenous hydrogen sulfide exerts proliferation, anti-apoptosis, migration effects and accelerates cell cycle progression in multiple myeloma cells via activating the Akt pathway. Oncol. Rep. 2016, 36, 1909–1916, Corrigendum in Oncol. Rep. 2021, 45, 1315. [Google Scholar] [CrossRef] [Green Version]
  247. Zhen, Y.; Pan, W.; Hu, F.; Wu, H.; Feng, J.; Zhang, Y.; Chen, J. Exogenous hydrogen sulfide exerts proliferation/anti-apoptosis/angiogenesis/migration effects via amplifying the activation of NF-κB pathway in PLC/PRF/5 hepatoma cells. Int. J. Oncol. 2015, 46, 2194–2204. [Google Scholar] [CrossRef] [Green Version]
  248. Tiong, C.X.; Lu, M.; Bian, J.S. Protective effect of hydrogen sulphide against 6-OHDA-induced cell injury in SH-SY5Y cells involves PKC/PI3K/Akt pathway. Br. J. Pharmacol. 2010, 161, 467–480. [Google Scholar] [CrossRef] [Green Version]
  249. Yang, G.; Zhao, K.; Ju, Y.; Mani, S.; Cao, Q.; Puukila, S.; Khaper, N.; Wu, L.; Wang, R. Hydrogen sulfide protects against cellular senescence via S-sulfhydration of Keap1 and activation of Nrf2. Antioxid. Redox Signal. 2013, 18, 1906–1919. [Google Scholar] [CrossRef]
  250. Zhao, K.; Ju, Y.; Li, S.; Altaany, Z.; Wang, R.; Yang, G. S-sulfhydration of MEK1 leads to PARP-1 activation and DNA damage repair. EMBO Rep. 2014, 15, 792–800. [Google Scholar] [CrossRef] [Green Version]
  251. Hoesel, B.; Schmid, J.A. The complexity of NF-κB signaling in inflammation and cancer. Mol. Cancer 2013, 12, 86. [Google Scholar] [CrossRef] [Green Version]
  252. Rojo de la Vega, M.; Chapman, E.; Zhang, D.D. NRF2 and the Hallmarks of Cancer. Cancer Cell 2018, 34, 21–43. [Google Scholar] [CrossRef] [PubMed]
  253. Lu, Z.; Xu, S. ERK1/2 MAP kinases in cell survival and apoptosis. IUBMB Life 2006, 58, 621–631. [Google Scholar] [CrossRef] [PubMed]
  254. Ray Chaudhuri, A.; Nussenzweig, A. The multifaceted roles of PARP1 in DNA repair and chromatin remodelling. Nat. Rev. Mol. Cell Biol. 2017, 18, 610–621. [Google Scholar] [CrossRef] [PubMed]
  255. Szczesny, B.; Marcatti, M.; Zatarain, J.R.; Druzhyna, N.; Wiktorowicz, J.E.; Nagy, P.; Hellmich, M.R.; Szabo, C. Inhibition of hydrogen sulfide biosynthesis sensitizes lung adenocarcinoma to chemotherapeutic drugs by inhibiting mitochondrial DNA repair and suppressing cellular bioenergetics. Sci. Rep. 2016, 6, 36125. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  256. Ascenção, K.; Dilek, N.; Augsburger, F.; Panagaki, T.; Zuhra, K.; Szabo, C. Pharmacological induction of mesenchymal-epithelial transition via inhibition of H2S biosynthesis and consequent suppression of ACLY activity in colon cancer cells. Pharmacol. Res. 2021, 165, 105393. [Google Scholar] [CrossRef] [PubMed]
  257. Phillips, C.M.; Zatarain, J.R.; Nicholls, M.E.; Porter, C.; Widen, S.G.; Thanki, K.; Johnson, P.; Jawad, M.U.; Moyer, M.P.; Randall, J.W.; et al. Upregulation of Cystathionine-β-Synthase in Colonic Epithelia Reprograms Metabolism and Promotes Carcinogenesis. Cancer Res. 2017, 77, 5741–5754. [Google Scholar] [CrossRef] [Green Version]
  258. Módis, K.; Ju, Y.; Ahmad, A.; Untereiner, A.A.; Altaany, Z.; Wu, L.; Szabo, C.; Wang, R. S-Sulfhydration of ATP synthase by hydrogen sulfide stimulates mitochondrial bioenergetics. Pharmacol. Res. 2016, 113, 116–124. [Google Scholar] [CrossRef] [Green Version]
  259. Folkman, J. Angiogenesis: An organizing principle for drug discovery? Nat. Rev. Drug Discov. 2007, 6, 273–286. [Google Scholar] [CrossRef]
  260. Dicks, N.; Gutierrez, K.; Michalak, M.; Bordignon, V.; Agellon, L.B. Endoplasmic reticulum stress, genome damage, and cancer. Front. Oncol. 2015, 5, 11. [Google Scholar] [CrossRef] [Green Version]
  261. Wei, H.; Zhang, R.; Jin, H.; Liu, D.; Tang, X.; Tang, C.; Du, J. Hydrogen sulfide attenuates hyperhomocysteinemia-induced cardiomyocytic endoplasmic reticulum stress in rats. Antioxid. Redox Signal. 2010, 12, 1079–1091. [Google Scholar] [CrossRef]
  262. Li, C.; Hu, M.; Wang, Y.; Lu, H.; Deng, J.; Yan, X. Hydrogen sulfide preconditioning protects against myocardial ischemia/reperfusion injury in rats through inhibition of endo/sarcoplasmic reticulum stress. Int. J. Clin. Exp. Pathol. 2015, 8, 7740–7751. [Google Scholar] [PubMed]
  263. Li, X.; Zhang, K.Y.; Zhang, P.; Chen, L.X.; Wang, L.; Xie, M.; Wang, C.Y.; Tang, X.Q. Hydrogen sulfide inhibits formaldehyde-induced endoplasmic reticulum stress in PC12 cells by upregulation of SIRT-1. PLoS ONE 2014, 9, e89856. [Google Scholar] [CrossRef]
  264. Bełtowski, J. Protein homocysteinylation: A new mechanism of atherogenesis? Adv. Hyg. Exp. Med. 2005, 59, 392–404. [Google Scholar]
  265. Chen, S.M.; Tang, X.Q. Homocysteinylation and Sulfhydration in Diseases. Curr. Neuropharmacol. 2022, 20, 1726–1735. [Google Scholar] [CrossRef]
  266. Majumder, A.; Singh, M.; Tyagi, S.C. Post-menopausal breast cancer: From estrogen to androgen receptor. Oncotarget 2017, 8, 102739–102758. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  267. Majumder, A.; Sandhu, M.; Banerji, D.; Steri, V.; Olshen, A.; Moasser, M.M. The role of HER2 and HER3 in HER2-amplified cancers beyond breast cancers. Sci. Rep. 2021, 11, 9091. [Google Scholar] [CrossRef]
  268. Gao, X.; Sanderson, S.M.; Dai, Z.; Reid, M.A.; Cooper, D.E.; Lu, M.; Richie, J.P., Jr.; Ciccarella, A.; Calcagnotto, A.; Mikhael, P.G.; et al. Dietary methionine influences therapy in mouse cancer models and alters human metabolism. Nature 2019, 572, 397–401. [Google Scholar] [CrossRef]
  269. Mitsuboshi, S.; Niimura, T.; Kanda, M.; Ishida, S.; Zamami, Y.; Ishizawa, K. Risk of Hematologic Events With Coadministration of Methotrexate and the Breast Cancer Resistance Protein Inhibitor Febuxostat. Ann. Pharmacother. 2022, 56, 910–915. [Google Scholar] [CrossRef]
  270. Diddens, H.; Niethammer, D.; Jackson, R.C. Patterns of cross-resistance to the antifolate drugs trimetrexate, metoprine, homofolate, and CB3717 in human lymphoma and osteosarcoma cells resistant to methotrexate. Cancer Res. 1983, 43, 5286–5292. [Google Scholar]
  271. Mullarky, E.; Lucki, N.C.; Beheshti Zavareh, R.; Anglin, J.L.; Gomes, A.P.; Nicolay, B.N.; Wong, J.C.; Christen, S.; Takahashi, H.; Singh, P.K.; et al. Identification of a small molecule inhibitor of 3-phosphoglycerate dehydrogenase to target serine biosynthesis in cancers. Proc. Natl. Acad. Sci. USA 2016, 113, 1778–1783. [Google Scholar] [CrossRef] [PubMed]
  272. Pacold, M.E.; Brimacombe, K.R.; Chan, S.H.; Rohde, J.M.; Lewis, C.A.; Swier, L.J.; Possemato, R.; Chen, W.W.; Sullivan, L.B.; Fiske, B.P.; et al. A PHGDH inhibitor reveals coordination of serine synthesis and one-carbon unit fate. Nat. Chem. Biol. 2016, 12, 452–458. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  273. Ducker, G.S.; Chen, L.; Morscher, R.J.; Ghergurovich, J.M.; Esposito, M.; Teng, X.; Kang, Y.; Rabinowitz, J.D. Reversal of Cytosolic One-Carbon Flux Compensates for Loss of the Mitochondrial Folate Pathway. Cell Metab. 2016, 23, 1140–1153. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  274. Hellmich, M.R.; Coletta, C.; Chao, C.; Szabo, C. The therapeutic potential of cystathionine β-synthetase/hydrogen sulfide inhibition in cancer. Antioxid. Redox Signal. 2015, 22, 424–448. [Google Scholar] [CrossRef] [Green Version]
  275. Lee, Z.W.; Zhou, J.; Chen, C.S.; Zhao, Y.; Tan, C.H.; Li, L.; Moore, P.K.; Deng, L.W. The slow-releasing hydrogen sulfide donor, GYY4137, exhibits novel anti-cancer effects in vitro and in vivo. PLoS ONE 2011, 6, e21077. [Google Scholar] [CrossRef] [Green Version]
  276. Lu, S.; Gao, Y.; Huang, X.; Wang, X. GYY4137, a hydrogen sulfide (H2S) donor, shows potent anti-hepatocellular carcinoma activity through blocking the STAT3 pathway. Int. J. Oncol. 2014, 44, 1259–1267. [Google Scholar] [CrossRef] [Green Version]
  277. Sakuma, S.; Minamino, S.; Takase, M.; Ishiyama, Y.; Hosokura, H.; Kohda, T.; Ikeda, Y.; Fujimoto, Y. Hydrogen sulfide donor GYY4137 suppresses proliferation of human colorectal cancer Caco-2 cells by inducing both cell cycle arrest and cell death. Heliyon 2019, 5, e02244. [Google Scholar] [CrossRef] [Green Version]
  278. Tanase, S.; Morino, Y. Irreversible inactivation of aspartate aminotransferases during transamination with L-propargylglycine. Biochem. Biophys. Res. Commun. 1976, 68, 1301–1308. [Google Scholar] [CrossRef]
  279. Burnett, G.; Marcotte, P.; Walsh, C. Mechanism-based inactivation of pig heart L-alanine transaminase by L-propargylglycine. Half-site reactivity. J. Biol. Chem. 1980, 255, 3487–3491. [Google Scholar] [CrossRef]
  280. Mitra, J.; Bhattacharyya, D. Irreversible inactivation of snake venom l-amino acid oxidase by covalent modification during catalysis of l-propargylglycine. FEBS Open Bio 2013, 3, 135–143. [Google Scholar] [CrossRef] [Green Version]
  281. Yadav, P.K.; Yamada, K.; Chiku, T.; Koutmos, M.; Banerjee, R. Structure and kinetic analysis of H2S production by human mercaptopyruvate sulfurtransferase. J. Biol. Chem. 2013, 288, 20002–20013. [Google Scholar] [CrossRef] [Green Version]
  282. Hanaoka, K.; Sasakura, K.; Suwanai, Y.; Toma-Fukai, S.; Shimamoto, K.; Takano, Y.; Shibuya, N.; Terai, T.; Komatsu, T.; Ueno, T.; et al. Discovery and Mechanistic Characterization of Selective Inhibitors of H2S-producing Enzyme: 3-Mercaptopyruvate Sulfurtransferase (3MST) Targeting Active-site Cysteine Persulfide. Sci. Rep. 2017, 7, 40227. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  283. Bantzi, M.; Augsburger, F.; Loup, J.; Berset, Y.; Vasilakaki, S.; Myrianthopoulos, V.; Mikros, E.; Szabo, C.; Bochet, C.G. Novel Aryl-Substituted Pyrimidones as Inhibitors of 3-Mercaptopyruvate Sulfurtransferase with Antiproliferative Efficacy in Colon Cancer. J. Med. Chem. 2021, 64, 6221–6240. [Google Scholar] [CrossRef]
  284. Zuhra, K.; Augsburger, F.; Majtan, T.; Szabo, C. Cystathionine-β-Synthase: Molecular Regulation and Pharmacological Inhibition. Biomolecules 2020, 10, 697. [Google Scholar] [CrossRef] [PubMed]
  285. Asimakopoulou, A.; Panopoulos, P.; Chasapis, C.T.; Coletta, C.; Zhou, Z.; Cirino, G.; Giannis, A.; Szabo, C.; Spyroulias, G.A.; Papapetropoulos, A. Selectivity of commonly used pharmacological inhibitors for cystathionine β synthase (CBS) and cystathionine γ lyase (CSE). Br. J. Pharmacol. 2013, 169, 922–932. [Google Scholar] [CrossRef] [PubMed] [Green Version]
Figure 1. Schematic diagram of Hcy production through the methionine and folate cycle. (A) Dietary methionine is converted to homocysteine (Hcy) through S-adenosyl methionine (SAM) and S-adenosyl homocysteine (SAH) and then back to methionine (MET) via the remethylation pathway. Half of Hcy goes to the transsulfuration pathway, where it is converted to cysteine with the help of cystathionine-β synthase (CBS) and cystathionine-γ lyase (CSE). Then cysteine is further converted to glutathione (GSH); (B) Conversion of cobalamin (vitamin B12) to methyl-B12 in the presence of methionine synthase reductase (MTR) is necessary for remethylation of 5-methyl-tetrahydrofolate (THF) to THF; (C) Dietary folic acid (vitamin B9) enters the folate cycle after its conversion first to dihydrofolate (DHF) and then to THF. The 5, 10-methyltetrahydrofolate reductase (MTHFR) is a key enzyme that converts 5, 10-methylene-THF to 5-methyl-THF [15].
Figure 1. Schematic diagram of Hcy production through the methionine and folate cycle. (A) Dietary methionine is converted to homocysteine (Hcy) through S-adenosyl methionine (SAM) and S-adenosyl homocysteine (SAH) and then back to methionine (MET) via the remethylation pathway. Half of Hcy goes to the transsulfuration pathway, where it is converted to cysteine with the help of cystathionine-β synthase (CBS) and cystathionine-γ lyase (CSE). Then cysteine is further converted to glutathione (GSH); (B) Conversion of cobalamin (vitamin B12) to methyl-B12 in the presence of methionine synthase reductase (MTR) is necessary for remethylation of 5-methyl-tetrahydrofolate (THF) to THF; (C) Dietary folic acid (vitamin B9) enters the folate cycle after its conversion first to dihydrofolate (DHF) and then to THF. The 5, 10-methyltetrahydrofolate reductase (MTHFR) is a key enzyme that converts 5, 10-methylene-THF to 5-methyl-THF [15].
Antioxidants 12 01520 g001
Figure 2. Different forms of homocysteine found in blood circulation: Hcy can be secreted from the cells and can be detected in blood in three different forms: around 1% as free thiol, 70–80% present bound with plasma proteins, and the remaining 20–30% present as homo/heterodimerized with other thiols.
Figure 2. Different forms of homocysteine found in blood circulation: Hcy can be secreted from the cells and can be detected in blood in three different forms: around 1% as free thiol, 70–80% present bound with plasma proteins, and the remaining 20–30% present as homo/heterodimerized with other thiols.
Antioxidants 12 01520 g002
Figure 3. Different signaling pathways showing the multifactorial effects of H2S in cancer development. The cartoons represent the six cancer hallmarks influenced by H2S/H2S-mediated protein persulfidation.
Figure 3. Different signaling pathways showing the multifactorial effects of H2S in cancer development. The cartoons represent the six cancer hallmarks influenced by H2S/H2S-mediated protein persulfidation.
Antioxidants 12 01520 g003
Figure 4. Signaling pathways of H2S production via Hcy metabolism. In the cytoplasm, H2S is produced from Hcy with the help of the CBS and CSE enzymes, whereas in mitochondria, H2S is produced with the help of the 3-MST enzyme.
Figure 4. Signaling pathways of H2S production via Hcy metabolism. In the cytoplasm, H2S is produced from Hcy with the help of the CBS and CSE enzymes, whereas in mitochondria, H2S is produced with the help of the 3-MST enzyme.
Antioxidants 12 01520 g004
Figure 5. Cartoon diagram showing how homocysteine and H2S balance can be targeted for cancer treatment. Targeting the transsulfuration and remethylation pathway in cancer cells will build up highly toxic homocysteine inside the cells. Consequently, cancer progression will be inhibited via apoptosis, poor angiogenesis, protein oxidation, and oxidative and ER stress.
Figure 5. Cartoon diagram showing how homocysteine and H2S balance can be targeted for cancer treatment. Targeting the transsulfuration and remethylation pathway in cancer cells will build up highly toxic homocysteine inside the cells. Consequently, cancer progression will be inhibited via apoptosis, poor angiogenesis, protein oxidation, and oxidative and ER stress.
Antioxidants 12 01520 g005
Table 1. Association of Hyperhomocysteinemia with different disorders.
Table 1. Association of Hyperhomocysteinemia with different disorders.
GenesPolymorphismsConditionAssociated ComplicationsReferences
CBS844INS68HHcyPeripheral artery occlusive disease[50]
T833CHHcyStroke[51]
844INS68HHcyThrombosis[52]
MTHFRC677THHcyRetinal vein occlusion[53]
C677THHcyStroke[42,43,44,45]
C677THHcyVenous thromboembolism[54]
C677THHcyHypertension[55,56,57]
C677THHcyAlzheimer’s Disease[58]
A1298CHHcyCerebral venous sinus thrombosis[59,60,61]
C677THHcyHyperlipidemia[62]
C677THHcyDiabetic nephropathy[63,64,65,66]
C677THHcyCerebral venous thrombosis[67]
C677THHcyParkinson’s Disease[68,69]
Table 2. Association of polymorphisms homocysteine metabolism genes with cancer risk. (odds ratio is indicated as OR).
Table 2. Association of polymorphisms homocysteine metabolism genes with cancer risk. (odds ratio is indicated as OR).
GenesPolymorphismsCancer TypesSignificant Association (OR)References
MTHFR677C-> TBreast CancerPositive Association (1.19)[22]
Ovarian CancerNo association (1.03)[22]
Esophageal Squamous Cell CarcinomaPositive Association (1.47)[96]
Acute Lymphocytic LeukemiaNegative Association (0.99)[21]
Prostate CancerNegative association (0.78)[23]
Colorectal AdenomasNegative association (0.76)[97]
Late-stage colorectal tumorigenesisPositive Association (1.32)[29]
Endometrial CancerNo association (1.10)[98]
1298A->CProstate CancerNegative Association (0.58)[99]
Acute Lymphocytic LeukemiaNegative Association (0.33)[21]
Acute Myeloid LeukemiaNo association (1.00)[88]
Endometrial CancerNo association (1.00)[98]
MTRR66A->GAcute Myeloid LeukemiaPositive association for Asian population (1.40)[100]
Head and Neck CancerPositive Association (1.24)[101]
Colorectal CancerPositive Association (2.77, 1.15)[87,102]
Gastric CancerPositive Association (1.39)[103]
Breast CancerPositive Association (4.45)[104]
MTRb2756A->GColorectal CancerPositive Association (2.04)[28]
Primary Liver CancerNo association (1.00)[105]
Breast CancerNo association (1.00)[106]
Glioblastoma MultiformeNo association (1.00)[107]
Upper Gastrointestinal Tract cancerNo association (1.00)[108]
Digestive System CancerNo association (1.00)[109]
MTHFD11958G->AGastric CancerPositive Association (2.05)[110]
G1958AColon CancerNegative Association (0.89)[111]
BHMT742G->AHead and Neck Squamous Cell CarcinomaPositive Association (1.34)[112]
Breast CancerNo association (0.98)[113]
Cervical CancerNegative Association (0.433)[114]
Ovarian CancerNo association (1.00)[115]
Colorectal AdenomaPositive Association (1.09)[116]
TCN 2776G>CGlioblastoma MultiformeNo association (1.00)[107]
Primary Central Nervous System LymphomaNo association (1.00)[117]
TYMSTS 3′-UTREsophageal and Stomach CancerNo association (1.00)[118]
Table 3. Association of different H2S-producing enzymes in different cancer types.
Table 3. Association of different H2S-producing enzymes in different cancer types.
EnzymesCancer TypesUpregulation/DownregulationReference
CBSColon CancerUpregulation[182]
Ovarian CancerUpregulation[176]
Breast CancerUpregulation[183]
Thyroid CancerUpregulation[184]
Gallbladder AdenocarcinomaUpregulation[185]
Hepatocellular CarcinomaDownregulation[186]
Gastrointestinal CancerDownregulation[187]
CSEBreast CancerUpregulation[188]
Prostate CancerUpregulation[189]
Gastric CancerUpregulation[190]
Bladder CancerUpregulation[191]
HepatomaUpregulation[192]
Colon CancerUpregulation[193]
Renal Cell CarcinomaDownregulation[194]
3MSTGlioma TumorUpregulation[195]
Colon CancerUpregulation[196]
Disclaimer/Publisher’s Note: The statements, opinions and data contained in all publications are solely those of the individual author(s) and contributor(s) and not of MDPI and/or the editor(s). MDPI and/or the editor(s) disclaim responsibility for any injury to people or property resulting from any ideas, methods, instructions or products referred to in the content.

Share and Cite

MDPI and ACS Style

Majumder, A. Targeting Homocysteine and Hydrogen Sulfide Balance as Future Therapeutics in Cancer Treatment. Antioxidants 2023, 12, 1520. https://doi.org/10.3390/antiox12081520

AMA Style

Majumder A. Targeting Homocysteine and Hydrogen Sulfide Balance as Future Therapeutics in Cancer Treatment. Antioxidants. 2023; 12(8):1520. https://doi.org/10.3390/antiox12081520

Chicago/Turabian Style

Majumder, Avisek. 2023. "Targeting Homocysteine and Hydrogen Sulfide Balance as Future Therapeutics in Cancer Treatment" Antioxidants 12, no. 8: 1520. https://doi.org/10.3390/antiox12081520

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

Article Metrics

Back to TopTop