297
Views
68
CrossRef citations to date
0
Altmetric
Review

Chemopreventive activity of sulforaphane

, , , , , & show all
Pages 2905-2913 | Published online: 11 Sep 2018

Abstract

Cancer is one of the major causes of morbidity and mortality in the world. Carcinogenesis is a multistep process induced by genetic and epigenetic changes that disrupt pathways controlling cell proliferation, apoptosis, differentiation, and senescence. In this context, many bioactive dietary compounds from vegetables and fruits have been demonstrated to be effective in cancer prevention and intervention. Over the years, sulforaphane (SFN), found in cruciferous vegetables, has been shown to have chemopreventive activity in vitro and in vivo. SFN protects cells from environmental carcinogens and also induces growth arrest and/or apoptosis in various cancer cells. In this review, we will discuss several potential mechanisms of the chemopreventive activity of SFN, including regulation of Phase I and Phase II drug-metabolizing enzymes, cell cycle arrest, and induction of apoptosis, especially via regulation of signaling pathways such as Nrf2-Keap1 and NF-κB. Recent studies suggest that SFN can also affect the epigenetic control of key genes and greatly influence the initiation and progression of cancer. This research may provide a basis for the clinical use of SFN for cancer chemoprevention and enable us to design preventive strategies for cancer management, reduce cancer development and recurrence, and thus improve patient survival.

Introduction

Cancer is one of the major causes of morbidity and mortality in the world. Tumor development is a multistep process, including initiation, promotion, and progression, and involves genetic and epigenetic changes that disrupt pathways controlling cell proliferation, apoptosis, differentiation, and senescence.Citation1Citation3

Cancer chemoprevention is defined as the use of dietary or pharmacological agents to prevent, block, or even reverse the process of tumor development before clinical manifestation of the disease.Citation4 The major goal of chemoprevention is to delay the onset of cancer and to decrease its incidence. Therefore, effective chemoprevention requires the use of compounds that inhibit specific molecular steps in the carcinogenic pathway. These preventive measures that target the various steps involved in cancer initiation and progression could significantly decrease the incidence and mortality of cancer. In particular, the use of dietary chemoprevention strategies has gained significant interest.Citation5 The use of chemopreventive compounds may have a significant impact on establishing recommendations for high-risk cancer patients, thereby increasing their survival through simple dietary choices with easily accessible foods.Citation6 Some studies suggest that cruciferous vegetable intake may lower overall cancer risk, including colon, lung, and prostate cancer, particularly during the early stages.Citation4,Citation5,Citation7 This inverse relationship is strongest for the consumption of cruciferous vegetables, especially those of the Brassica genus (including Brussels sprouts, broccoli, cabbage, cauliflower, and bokchoy).Citation8 Thus, there is a growing interest in identifying specific chemoprotective constituents in cruciferous vegetables and their mechanisms of action. Sulforaphane (SFN), which is converted from a major glucosinolate in broccoli/broccoli sprouts, has been shown to prevent chemically induced cancers in animal models and to inhibit the growth of established tumors.Citation9

The mechanisms whereby SFN exerts chemopreventive activity include modulation of Phase I and II xenobiotic-metabolizing enzymes and direct inhibition of binding of carcinogens to DNA. As a result, SFN inhibits DNA adduct formation and reduces the mutation rate. SFN also has IL-10. In addition, SFN can arrest cell cycle progression, particularly in the G2/M phase,Citation11 and high concentrations of SFN can activate proapoptotic pathways.Citation12 Recent studies suggest that SFN can also regulate the epigenetic control of key genes including CDKs, p21, Bax, and Nrf2, and thus greatly influence the initiation and progression of cancer.Citation13

There is growing experimental evidence to support the efficacy of SFN in regulating the prevention and treatment of cancer through several different mechanisms. The aim of this review is to summarize the chemopreventive activity of SFN.

The characteristics and biological activity of SFN

Isothiocyanates (ITCs), plant-derived chemoprotective constituents, are formed by the hydrolysis of their precursor parent compounds, glucosinolates. The levels of glucosinolate vary greatly within members of the Cruciferae family, depending on the environment and genotype.Citation14 SFN is an important and well-studied ITC derived from cruciferous vegetables, including broccoli, cauliflower, cabbage, and kale, with the highest concentration found in broccoli and broccoli sprouts.Citation14 SFN has antioxidant, antiproliferative, and anticarcinogenic properties.Citation15 The epidemiological surveys in the US, Europe, China, and Singapore have established the associations between consumption of cruciferous vegetables and downtrend of carcinogenic risk.Citation16,Citation17 Numerous experimental studies also confirm the preventive effect of SFN in chemically induced lung, breast, renal, prostate, and colon cancers.Citation8,Citation9,Citation11,Citation18,Citation19

There are three key stages in the development of cancer: initiation, maintenance, and progression. SFN can inhibit the initiation of tumor development or halt the progression of cancer. Prevention of cancer initiation can be achieved by minimizing the exposure of cells to environmental carcinogenic factors through inhibition of their activation or by promoting their detoxification.Citation16 In addition, SFN can also exhibit chemopreventive behavior by interfering with various signaling pathways that regulate oxidative stress, inflammation, cell proliferation, differentiation, and apoptosis. Thus, SFN can impact all stages of tumor development.Citation2,Citation17

In rats, the pharmacokinetics of SFN was assessed following an oral dose of 50 μmol of SFN. The plasma concentration of SFN can be detected at 1 hour and it peaks at 20 μM at 4 hours. The concentration of SFN in plasma increases with the activation of various clusters of genes that are important in cellular defense mechanisms and cell cycle regulation, such as metallothionein, GSTA3, and MAPK in rat livers.Citation20 It has been reported that glucosinolate or ITC preparations (the main components of SFN) were administered to the rats with dimethylbenzanthracene-induced mammary tumors by daily gavage. The mammary tumor development was significantly retarded in terms of both tumor multiplicity and incidence.Citation21 SFN can also inhibit skin tumorigenesis in mice and inhibits the growth of PC-3 cell xenografts in nude mice.Citation22,Citation23 In addition, the ability of SFN to inhibit tumor growth, metastasis, and angiogenesis and to enhance the therapeutic potential of tumor necrosis factor-related apoptosis inducing ligand (TRAIL) suggests that SFN alone or in combination with TRAIL can be used for the management of embryonal rhabdomyosarcomas.Citation24

Traditionally, the major mechanism of protection against the initiation of carcinogenesis by SFN includes modulation of Phase I and Phase II xenobiotic-metabolizing enzymes that increase cell defenses against oxidative damage and promote the removal of carcinogens. However, there is ample evidence suggesting that SFN can have chemoprotective effects through multiple mechanisms, including the modulation of biotransformation enzymes, induction of apoptotic signaling pathways, arrest of cell cycle progression, as well as inhibition of angiogenesis and inflammation.Citation25 In addition, recent studies suggest that SFN can also affect the epigenetic control of key genes and, thus, greatly influence the initiation and progression of cancer.Citation13,Citation26

SFN inhibits Phase I and Phase II drug-metabolizing enzymes

Virtually, all carcinogens are exposed to metabolism when they enter the body. These metabolic processes mainly include oxidation, reduction, hydrolysis, and conjugation reactions, which cause the chemical carcinogens to become more hydrophilic. Phase I (cytochrome P450, CYP) drug-metabolizing enzymes (DMEs) are frequently involved in the metabolic processing of carcinogenic chemicals.Citation24 The formation of electrophilic reactive metabolites or procarcinogens is often catalyzed by CYP DMEs through two electron oxidations to a hydroxylated or epoxidized medium.Citation27 These chemical reaction processes have been defined as the bioactivation of carcinogens.Citation2 Numerous studies have demonstrated that SFN can inhibit the DNA adduct and chemical carcinogenic processes through regulation of certain CYP isoforms via a competitive mechanism and a direct covalent modification in rodents.Citation28 SFN alone did not significantly alter the activity and expression of the studied DMEs, except for GSTA1 at the mRNA level, which was significantly enhanced.Citation29 However, SFN dose-dependently inhibited the activities of CYP1A1 and 2B1/2 in rat hepatocytes.Citation29 In other reports, SFN also reduced CYP3A4 activity by decreasing CYP3A4 transcript level in rat hepatocytes.Citation30 It has also been shown that SFN is a potent competitive inhibitor of CYP2E1 with a Ki of 37.0±4.5 mM in microsomes from livers of acetone-treated rats, and SFN is able to inhibit the genotoxicity of N-nitrosodimethylamine.Citation31 Overall, these findings suggest that SFN must be able to inhibit the activities of numerous CYPs and potentially alleviate the activation of procarcinogens. SFN has also been shown to effectively inhibit the chemically induced DNA adduct formation and tumorigenesis following exposure to benzo[a]pyrene and 1,6-dinitropyrene in the human mammary epithelial cell line MCF-10F.Citation32 All these reports provide evidence that SFN protects against carcinogen-induced DNA damage, and support that the inhibition of Phase I enzymes plays a role in the chemopreventive activity of SFN.

Another major mechanism not involving the modulation of Phase I DMEs is the induction of Phase II DMEs, which transform carcinogens to inactive metabolites that are easily eliminated from the body, thus preventing their reaction with DNA. Over the past few decades, SFN has attracted a lot of attention in cancer chemoprevention since it is among the most potent naturally occurring inducers of Phase II enzymes, where a strong inverse relationship exists between the tissue levels of these enzymes and susceptibility to chemical carcinogenesis.Citation33 For example, the Phase II enzyme of GSTs, typically leading to carcinogen detoxification, plays a direct role in the carcinogenic process. Human deficiencies in GSTs expression have been associated with increased cancer risk, such as bladder, oral, and lung cancers.Citation34 Conversely, overexpression of GST-P1 protected human prostate cells from the cytotoxic and DNA damaging action of the prostate carcinogen.Citation35 Most work has focused on Phase II enzyme induction via antioxidant response element (ARE)-driven gene expression.Citation36 ARE-driven targets include NQO1, HO-1, UGT, and gamma-glutamylcysteine synthetase.Citation37

The modulation of Phase II enzyme activity and gene expression by SFN has been confirmed in a series of cell lines, of which the most commonly used are liver hepatoma cell lines HepG2 and Hepa1c1c7. Some studies indicate that NQO1 and GST activities were increased in the fore stomach, duodenum, and bladder of rats treated with SFN (40 μM/kg/day), while a higher dose of SFN (200–1,000 μM/kg/day) increased NQO1 and GST activities in the liver, colon, and pancreas of rats.Citation38,Citation39 The effect of SFN on Phase II enzyme regulation has also been studied generally in prostate cancer where SFN was shown to significantly induce the expression and activity of Phase II enzyme in the human prostate cell lines LNCaP, MDA PCa 2a, MDA PCa 2b, PC-3, and TSU-Pr1 treated with 0.1–15 mM of SFN.Citation40

Keap1-ARE signaling pathway is an important modifier of susceptibility to electrophilic and oxidative stresses, factors central to the processes of chemical carcinogenesis and other chronic degenerative diseases.Citation41 A number of studies have revealed that the effects of SFN on Nrf2 and its downstream cytoprotective genes are through modification of Keap1 cysteine residues;Citation42 activation of MAPK, PI3K, and PKB/Akt pathways; and epigenetic modifications, which result in the phosphorylation, nuclear accumulation, and increased transcription and stability of Nrf2.Citation43,Citation44 SFN can react with the thiol groups of Keap1 to form thionoacyl adducts, which enables Nrf2 to escape Keap1-dependent ubiquitination and degradation, leading to nuclear localization of Nrf2.Citation45,Citation46 The nuclear Nrf2 binds to ARE and activates Phase II detoxifying and antioxidant genes.Citation41 Shan et al reported that p38 MAPK activation could regulate Nrf2-ARE–driven enzymes, thereby facilitating the role of SFN in chemoprevention of bladder cancer.Citation47 The expression and activity of HO-1 by SFN regulation has been shown to be Nrf2 dependent in mouse peritoneal macrophages.Citation48 Also, activation of PI3K-PKB/Akt signaling regulated cell survival and Nrf2-driven HO-1 expression in SFN-treated human mesothelioma MSTO-211H cells.Citation49 Wu et al also indicated that SFN singly or in combination with estrogen increased Nrf2 activity through activation of the PI3K pathway in breast cancer cell line MCF-7.Citation50 SFN exerted an anticancer effect in a mouse skin tumor model, which involved the epigenetic reprogramming of Nrf2 leading to epigenetic reactivation of Nrf2 and subsequent induction of downstream target genes HO-1, NQO1, and UGT1A1.Citation51

Taken together, it appears that SFN, at least in part, inhibits tumor initiation by inhibiting Phase I enzymes and stimulating Phase II enzymes.

The anti-inflammatory activity of SFN

Chronic inflammation and carcinogenesis are thought to be closely related, and SFN has been found to have anti-inflammatory properties.Citation45 Constitutive activation of NF-κB is common in various human malignancies, including breast and prostate cancer, and leads to the upregulation of genes encoding adhesion molecules, inflammatory cytokines, growth factors, and antiapoptotic genes.Citation52

NF-κB is a transcription factor that regulates the expression of several proinflammatory genes, most notably including nitric oxide, inducible nitric oxide synthase, Cox-2, and TNF-α. Elevated levels of Cox-2 have been monitored in various tumors and may account for excessive production of prostaglandin. In human malignant glioblastoma cells, SFN can also significantly decrease NF-κB expression compared to control cells, suggesting that NF-κB is an important molecular target of SFN.Citation53

SFN can reduce the DNA-binding activity of NF-κB directly by indirectly interacting with the thiol groups leading to dithiocarbamate formation and directly uniting with essential cysteine residues of NF-κB subunits, thereby reducing their DNA-binding capacity. In addition, SFN can also interact with glutathione and other redox regulators such as thioredoxin and Ref-1, which indirectly intervenes with NF-κB DNA-binding activity.Citation54 These findings further confirm that thiol reactivity and redox modulation are important in the regulation of NF-κB–dependent transcription by SFN. Recently, the functions of SFN on natural killer cells and cell-mediated immune response were also researched in normal and tumor-bearing BALB/c mice, where administration of SFN significantly enhanced natural killer cell activity, antibody-dependent cellular cytotoxicity, and the production of IL-2 and interferon gamma in both normal as well as tumor-bearing mice.Citation55

Thus, the inactivation of NF-κB is an important chemopreventive mechanism of SFN. This suggests that SFN manifests anticarcinogenic effects not only through the regulation of biotransformation enzymes, but also by modulation of inflammation.

The apoptosis-inducing properties of SFN

There are various options by which cells are able to undergo cell death, and their respective mechanisms can be described based on their unique morphological features. Apoptosis, in contrast to other forms of cell death such as necrosis and autophagy, is the most prominent form of programmed cell death during animal development and maintenance of tissue homeostasis. Apoptosis is regarded as a “silent” mechanism of cell elimination projected to digest the contents of damaged cells and ensure the elimination of cells that are no longer necessary for the organism.Citation56 Apoptosis is an essential function during the development and homeostasis of animals and cells, but inappropriate regulation of apoptosis may cause serious disorders. For example, excessive apoptosis has been related to neurodegenerative diseases, organ failure, autoimmune diseases, and cancers resulting from exposure to toxins.Citation57 Many factors, including various genotoxic compounds and complicated environmental stresses, are responsible for the initiation and execution of apoptosis.

Data clearly show that SFN is a powerful inducer of apoptosis both in vitro and in vivo.Citation58 Twenty years ago, the first evidence of SFN having proapoptotic activity was provided in a report of an antitumor drug in human colon cancer cells, where SFN could decrease the viability of HT29 and Caco-2 cells.Citation59 Then, several other reports using in vitro models also demonstrated that SFN was able to mediate apoptosis by regulating multiple targets in the apoptotic pathway.Citation60Citation62 SFN has been shown to target several steps involved in apoptosis, including downregulation of the expression of antiapoptotic factors Bcl-2 and Bcl-XL, upregulation of proapoptotic Bax, proteolytic activation of caspase-3, and degradation and/or cleavage of poly(ADP-ribose) polymerase.Citation61,Citation62 Additionally, it has been demonstrated that SFN can trigger the activation of Bax, downregulate inhibitor of apoptosis proteins (IAP) family (including cIAP1, cIAP2, and XIAP), and induce the activation of Apaf-1 in human prostate cancer cells.

The mechanisms whereby SFN regulates apoptosis are diverse. It was demonstrated that SFN-induced apoptosis is mediated by reactive oxygen species (ROS)-mediated activation of AMPK in human gastric cancer cells.Citation60 Treatment of PC-3 cells with SFN can lead to ROS generation and break the mitochondrial membrane potential, which causes cytosolic release of cytochrome c and apoptosis. Apart from ROS, the MAPK pathway has also been reported to be activated by SFN in human prostate cancer PC-3 cells. SFN has also been shown to activate AP-1, which requires the activation of ERK and JNK signaling pathways, during the regulation of cell death elicited by SFN in PC-3 cells.Citation63

Mitochondrial signaling pathways play an important role in apoptosis, and SFN can activate the intrinsic/mitochondrial apoptotic pathway. This apoptosis involves release of cytochrome c from the mitochondria into the cytosol, which then binds Apaf-1 and leads to activation of the “apoptosis initiator” caspase-9.Citation63 Furthermore, decreased mitochondrial potential has been detected in prostate and bladder cancer cells in response to SFN.Citation64,Citation65 Apart from the activation of a caspase-dependent apoptotic pathway, SFN has also been shown to induce apoptosis in a caspase-independent manner. Subsequent release of mitochondrial proteins including cytochrome c, Smac, and AIF has also been observed. Treatment of glioblastoma cells with SFN (40 μM) for 24 hours can cause a significant increase in the cytosolic protein expression levels of AIF.Citation66 All of the above evidence demonstrates that SFN can activate the mitochondrial apoptotic pathway.

A number of studies have also revealed that SFN can activate the death receptor/extrinsic pathway of apoptosis.Citation67,Citation68 Shankar et al demonstrated that SFN could upregulate the protein expressions of TRAIL receptor-1 (TRAIL-R1/DR4), TRAIL-R2/DR5, Bax, and Bak, and could inhibit the activation of NF-κB, P13K/AKT, and MEK/ERK pathways in prostate tumor tissues.Citation69 However, the activation of the downstream effector caspase-3 has been well documented for SFN in many pathways.Citation70,Citation71

SFN-induced apoptosis is also involved in the regulation of proteins in the IAP family, which can inhibit the activity of caspase signaling pathway. After treatment of prostate cancer PC-3 and LNCaP cells with SFN, the protein expression of all three members of the IAP family, including XIAP, c-IAP1, and c-IAP2, was significantly reduced.Citation72 Furthermore, the expression of the IAP family members c-IAP1 and c-IAP2 was also downregulated with SFN treatment in human glioblastoma cells.Citation66

The induction of cell cycle arrest of SFN

The progression of the cell cycle through all four phases, G1, S, G2, and M, is adjusted by CDKs and cyclins.Citation73 It has been shown that SFN can arrest the cell cycle at various stages of its progression, thereby inhibiting the growth of cancer cells. Induction of cell cycle arrest in G0/G1, S, and G2/M phases upon treatment with SFN has been reported in breast,Citation74 bladder,Citation65 colon,Citation75 and prostate cancer cells.Citation76,Citation77 Arrest at the G0/G1 phase of the cell cycle in response to SFN was associated with a higher depolarization of the mitochondrial membrane potential and intracellular ROS generation in human non-small-cell lung cancer cells,Citation78 as well as a decreased phosphorylation of tumor suppressor RB and protection of the RB–E2F-1 complex in epithelial ovarian cancer cell lines of MDAH 2774 and SkOV-3.Citation79 Another study demonstrated that G2/M phase arrest by SFN was achieved in human ovarian cancer cell line PA-1 via CDK1 downregulation and dissociation of the cyclin B1/CDK1 complex.Citation80 However, the ability of SFN to specifically cause cytotoxicity in cancer cells, not normal cells, is an important factor in determining its safety and clinical relevance as a chemoprevention agent. It has been proved by Clarke et al’s team that SFN selectively induced cell cycle arrest and apoptosis in hyperplastic and cancerous prostate cells, but not PrEC normal cells.Citation76 The differences in sensitivity to SFN in PrEC and cancerous prostate cells are likely due to a decrease in several class I HDAC proteins, induction of histone acetylation at the P21 promoter, and ultimately, induction of G2/M cell cycle arrest and apoptosis, and not due to differences in SFN metabolism or differences in Phase II enzyme induction.Citation76

Substantial evidence indicates that the concentration of SFN is responsible for these different effects. G2/M arrest was observed with treatment of SFN at a dose of 20 μM, whereas concentrations of 100 μM induced the accumulation of cells in the sub-G1 phase, cell death, and dissipation of mitochondrial membrane potential in human colon adenocarcinoma cell Caco-2.Citation81 SFN at 10 μM can reduce cell viability and induce G2/M phase arrest in prostate cancer DU145 cells.Citation82 Another G2/M phase arrest of SFN was observed in ovarian cancer PA-1 cells with 12.5 μM SFN.Citation80 Other evidence also indicates that the growth inhibitory effect of SFN is highly time dependent.Citation83 Inhibition of cell growth by SFN followed a biphasic pattern in human colon cancer cells: transient exposure of human colon carcinoma cell line HT-29 to SFN for up to 6 hours resulted in reversible G2/M cell cycle arrest and cytostatic growth inhibition even at high concentrations.Citation59 After a minimum continuous exposure time of 12 hours, SFN could irreversibly induce cell cycle arrest in the G2/M phase and the cells appeared to subsequently undergo apoptosis.Citation84 These data indicate that SFN can influence cell cycle arrest through varying complex regulatory mechanisms.

Epigenetic regulation of cancer genes by SFN

Many natural dietary agents have been confirmed to be effective in cancer prevention. Treatment of cancer cells with these nutraceuticals often mediates favorable epigenetic changes.Citation85 There are three common types of epigenetic mechanisms in mammals, including changes in DNA methylation, histone modifications, and non-coding RNAs.

DNA methylation is the most extensively researched of the epigenetic processes. DNMT and HDAC often work together in larger protein complexes to strip chromatin of active acetylation marks and lay down DNA methylation for stable gene repression. Enzymes that regulate the epigenetic signature of cancer cells have proven to be a viable target in cancer prevention and cancer therapeutic research.Citation5 Hsu et al confirmed that SFN could significantly decrease the expression of DNMTs and repressed methylation-silenced cyclin D2 in prostate cancer cells. These results demonstrate that SFN has the ability to epigenetically modulate cyclin D2 expression, and provide novel insights into the mechanisms by which SFN may regulate the gene expression as a chemopreventive agent in prostate cancer.Citation77 Another paper also revealed that SFN can promote breast cancer cell death by adjusting the levels of DNMT and HDAC and downregulating the levels of cyclin D1, CDK4, and pRB.Citation74

Many cancers often exhibit aberrant patterns of histone modification and non-coding RNAs. SFN-induced suppression of tumors in Apc-minus mice was associated with inhibition of HDAC activity and concomitant increases in acetylation of histone H3 and H4 located in the promoter region of p21 and Bax genes.Citation86 Zhang et al showed that SFN could increase mRNA and protein expressions of Nrf2 and its downstream target gene NQO1 by decreasing the protein levels of DNMT1 and DNMT3a.Citation87 SFN also attenuated the protein expression levels of HDACs 1, 4, 5, and 7, while increasing the level of active chromatin marker acetyl-histone 3(Ac-H3) during tumorigenesis in vivo (using TRAMP mice) and in vitro (using TRAMP C1 cells).Citation87 These results suggest that SFN may exert its chemopreventive effect partly via epigenetic modifications, and via expression of the Nrf2 gene with subsequent induction of its downstream antioxidative stress pathway.

Conclusion

Human clinical studies have supported the chemopreventive effects of SFN on carcinogenesis. The clinical Phase I and Phase II studies showed that broccoli sprout extracts containing SFN were well tolerated and caused no significant adverse effects in healthy volunteers, women with breast cancer, and men with recurrent prostate cancer.Citation88Citation90 In addition, a recent study reported that SFN effectively inhibited tumor growth and increased the sensitivity of cancer cells to chemotherapeutics in patients with advanced pancreatic ductal adenocarcinoma.Citation91

Based on the above-mentioned studies, it is clear that the SFN is a safe and relatively nontoxic chemopreventive agent, and exerts anticancer activities through multiple mechanisms, including regulation of Phase I and Phase II DMEs, anti-inflammatory activity, cell cycle arrest, induction of apoptosis, and the epigenetic regulation on Nrf2-Keap1, cyclins, and CDKs. Further understanding of the cancer chemopreventive activities of SFN will allow us to assess its efficacy in human cancers as a single agent or as part of combination strategies in various types of human cancers.

Acknowledgments

This study was supported in part by grants from the National Science Foundation of China (81570344 to Ying Xin), the Norman Bethne Program of Jilin University (2015225 to Ying Xin, 2015203 to Xin Jiang), the Fundamental Research Funds for the Jilin University (450060501512 to Xin Jiang), the Education Department of Jilin Province Foundations (2016-448 to Xin Jiang), and the Health and Family Planning Commission of Jilin Province Foundations (2016Q034 to Ying Xin and 2015Q010 to Xin Jiang).

Disclosure

The authors report no conflicts of interest in this work.

References

  • AnfossiSFuXNagvekarRCalinGAMicroRNAs, Regulatory messengers inside and outside cancer cellsAdv Exp Med Biol201810568710829754176
  • CheungKLKongA-NMolecular targets of dietary phenethyl isothiocyanate and sulforaphane for cancer chemopreventionAAPS J2010121879720013083
  • ZhouYYangGTianHSulforaphane metabolites cause apoptosis via microtubule disruption in cancerEndocr Relat Cancer201825325526829431641
  • LinsalataMOrlandoARussoFPharmacological and dietary agents for colorectal cancer chemoprevention: effects on polyamine metabolism (review)Int J Oncol20144551802181225119812
  • W WatsonGM BeaverLE WilliamsDH DashwoodRHoEPhytochemicals from cruciferous vegetables, epigenetics, and prostate cancer preventionAAPS J201315495196123800833
  • RamirezCNLiWZhangCIn vitro-in vivo dose response of ursolic acid, sulforaphane, PEITC, and curcumin in cancer preventionAAPS J201820119
  • MoriNShimazuTSasazukiSCruciferous vegetable intake is inversely associated with lung cancer risk among current nonsmoking men in the Japan Public Health Center (JPHC) StudyJ Nutr2017147584184928381528
  • QjWYangYWangJHanLHXiangYBCruciferous vegetable consumption and gastric cancer risk: a meta-analysis of epidemiological studiesCancer Sci201310481067107323679348
  • ShwterANAbdullahNAAlshawshMAChemoprevention of colonic aberrant crypt foci by Gynura procumbens in ratsJ Ethnopharmacol201415131194120124393787
  • BesslerHDjaldettiMBroccoli and human health: immunomodulatory effect of sulforaphane in a model of colon cancerInt J Food Sci Nutr201820118
  • JuengelEEulerSMaxeinerSSulforaphane as an adjunctive to everolimus counteracts everolimus resistance in renal cancer cell linesPhytomedicine2017271728314474
  • XuCShenGYuanXERK and JNK signaling pathways are involved in the regulation of activator protein 1 and cell death elicited by three isothiocyanates in human prostate cancer PC-3 cellsCarcinogenesis200627343744516272172
  • RoystonKUdayakumarNLewisKTollefsbolTA novel combination of withaferin A and sulforaphane inhibits epigenetic machinery, cellular viability and induces apoptosis of breast cancer cellsInt J Mol Sci20171851092
  • SivapalanTMelchiniASahaSBioavailability of glucoraphanin and sulforaphane from high-glucoraphanin broccoliMol Nutr Food Res Epub20171221
  • YangLPalliyaguruDLKenslerTWFrugal chemoprevention: targeting Nrf2 with foods rich in sulforaphaneSemin Oncol201643114615326970133
  • HanausekMWalaszekZSlagaTJDetoxifying cancer causing agents to prevent cancerIntegr Cancer Ther20032213914415035900
  • KarinMNuclear factor-κB in cancer development and progressionNature2006441709243143616724054
  • PereiraLSilvaPDuarteMTargeting colorectal cancer proliferation, stemness and metastatic potential using Brassicaceae extracts enriched in isothiocyanates: a 3D cell model-based studyNutrients201794368
  • FrydoonfarHRMcgrathDRSpigelmanADSulforaphane inhibits growth of a colon cancer cell lineColorectal Dis200461283114692949
  • HuRHebbarVKimBRIn vivo pharmacokinetics and regulation of gene expression profiles by isothiocyanate sulforaphane in the ratJ Pharmacol Exp Ther2004310126327114988420
  • AtwellLLZhangZMoriMSulforaphane bioavailability and chemopreventive activity in women scheduled for breast biopsyCancer Prev Res (Phila)20158121184119126511489
  • FranklinSJDickinsonSEKarlageKLBowdenGTMyrdalPBStability of sulforaphane for topical formulationDrug Dev Ind Pharm201440449450223611476
  • MyzakMCTongPDashwoodWMDashwoodRHHoESulforaphane retards the growth of human PC-3 xenografts and inhibits HDAC activity in human subjectsExp Biol Med (Maywood)2007232222723417259330
  • BergantinEQuartaCNanniCSulforaphane induces apoptosis in rhabdomyosarcoma and restores TRAIL-sensitivity in the aggressive alveolar subtype leading to tumor elimination in miceCancer Biol Ther20141591219122524971463
  • TomczykJOlejnikASulforaphane – a possible agent in prevention and therapy of cancerPostepy Hig Med Dosw (Online)20106459060321160094
  • SahaKHornyakTJEckertRLEpigenetic cancer prevention mechanisms in skin cancerAAPS J20131541064107123904153
  • MeunierBde VisserSPShaikSMechanism of oxidation reactions catalyzed by cytochrome p450 enzymesChem Rev200410493947398015352783
  • PeroccoPBronzettiGCanistroDGlucoraphanin, the bioprecursor of the widely extolled chemopreventive agent sulforaphane found in broccoli, induces Phase-I xenobiotic metabolizing enzymes and increases free radical generation in rat liverMutat Res20065951–212513616442570
  • LnenickovaKDymakovaASzotakovaBSulforaphane alters beta-naphthoflavone-induced changes in activity and expression of drug-metabolizing enzymes in rat hepatocytesMolecules20172211E198329144397
  • Gross-SteinmeyerKEatonDLDietary modulation of the biotransformation and genotoxicity of aflatoxin B1Toxicology20122992–3697922640941
  • BarceloSGardinerJMGescherAChipmanJKCYP2E1-mediated mechanism of anti-genotoxicity of the broccoli constituent sulforaphaneCarcinogenesis19961722772828625450
  • SingletaryKMacdonaldCInhibition of benzo[a]pyrene- and 1,6-dinitropyrene-DNA adduct formation in human mammary epithelial cells bydibenzoylmethane and sulforaphaneCancer Lett20001551475410814878
  • KwakMKEgnerPADolanPMRole of Phase 2 enzyme induction in chemoprotection by dithiolethionesMutat Res2001480–481305315
  • LandiSMammalian class theta GST and differential susceptibility to carcinogens: a reviewMutat Res2000463324728311018744
  • NelsonCPKiddLCSauvageotJProtection against 2-hydroxyamino-1-methyl-6-phenylimidazo[4,5-b]pyridine cytotoxicity and DNA adduct formation in human prostate by glutathione S-transferase P1Cancer Res200161110310911196146
  • Anwar-MohamedAElshenawyOHSoshilovAAMethylated pentavalent arsenic metabolites are bifunctional inducers, as they induce cytochrome P450 1A1 and NAD(P)H:quinone oxidoreductase through AhR- and Nrf2-dependent mechanismsFree Radic Biol Med20146717118724161444
  • LiuLWuWLiJTwo sesquiterpene aminoquinones protect against oxidative injury in HaCaT keratinocytes via activation of AMPKα/ERK-Nrf2/ARE/HO-1 signalingBiomed Pharmacother201810041742529471244
  • MundayRMundayCMInduction of Phase II detoxification enzymes in rats by plant-derived isothiocyanates: comparison of allyl isothiocyanate with sulforaphane and related compoundsJ Agric Food Chem20045271867187115053522
  • MatusheskiNVJefferyEHComparison of the bioactivity of two glucoraphanin hydrolysis products found in broccoli, sulforaphane and sulforaphane nitrileJ Agric Food Chem200149125743574911743757
  • BrooksJDPatonVGVidanesGPotent induction of Phase 2 enzymes in human prostate cells by sulforaphaneCancer Epidemiol Biomarkers Prev200110994995411535546
  • XingH-YCaiY-QWangX-FThe cytoprotective effect of hyperoside against oxidative stress is mediated by the Nrf2-ARE signaling pathway through GSK-3β inactivationPLoS One20151012e014518326674355
  • HuCEgglerALMesecarADvan BreemenRBModification of Keap1 cysteine residues by sulforaphaneChem Res Toxicol2011244515521 2011/04/1821391649
  • QinSHouD-XMultiple regulations of Keap1/Nrf2 system by dietary phytochemicalsMol Nutr Food Res20166081731175527523917
  • MageshSChenYHuLSmall molecule modulators of Keap1-Nrf2-ARE pathway as potential preventive and therapeutic agentsMed Res Rev201232468772622549716
  • ShimizuTInoueK-IchiHachiyaHShibuyaNAokiTKubotaKAccumulation of phosphorylated p62 is associated with NF-E2-related factor 2 activation in hepatocellular carcinomaJ Hepatobiliary Pancreat Sci201623846747127246794
  • FurukawaMXiongYBTB protein Keap1 targets antioxidant transcription factor Nrf2 for ubiquitination by the Cullin 3-Roc1 ligaseMol Cell Biol200525116217115601839
  • ShanYWangXWangWHeCBaoYp38 MAPK plays a distinct role in sulforaphane-induced up-regulation of ARE-dependent enzymes and down-regulation of COX-2 in human bladder cancer cellsOncol Rep20102341133113820204301
  • LinWWuRTWuTKhorT-OWangHKongA-NSulforaphane suppressed LPS-induced inflammation in mouse peritoneal macrophages through Nrf2 dependent pathwayBiochem Pharmacol200876896797318755157
  • LeeYJJeongHYKimYBReactive oxygen species and PI3K/Akt signaling play key roles in the induction of Nrf2-driven heme oxygenase-1 expression in sulforaphane-treated human mesothelioma MSTO-211H cellsFood Chem Toxicol201250211612322019695
  • WuJWilliamsDWalterGAThompsonWESidellNEstrogen increases Nrf2 activity through activation of the PI3K pathway in MCF-7 breast cancer cellsExp Cell Res2014328235136025172557
  • ZySZhangCLeeJHRequirement and epigenetics reprogramming of Nrf2 in suppression of tumor promoter TPA-induced mouse skin cell transformation by sulforaphaneCancer Prev Res (Phila)20147331932924441674
  • KolbergMPedersenSBastaniNECarlsenHBlomhoffRPaurITomato paste alters NF-kappaB and cancer-related mRNA expression in prostate cancer cells, xenografts, and xenograft microenvironmentNutr Cancer201567230531525664890
  • KarmakarSWeinbergMSBanikNLPatelSJRaySKActivation of multiple molecular mechanisms for apoptosis in human malignant glioblastoma T98G and U87MG cells treated with sulforaphaneNeuroscience200614131265128016765523
  • HeissEHerhausCKlimoKBartschHGerhäuserCNuclear factor kappa B is a molecular target for sulforaphane-mediated anti-inflammatory mechanismsJ Biol Chem200127634320083201511410599
  • ThejassPKuttanGAugmentation of natural killer cell and antibody-dependent cellular cytotoxicity in BALB/c mice by sulforaphane, a naturally occurring isothiocyanate from broccoli through enhanced production of cytokines IL-2 and IFN-gammaImmunopharmacol Immunotoxicol200628344345716997793
  • Pérez-GarijoAStellerHSpreading the word: non-autonomous effects of apoptosis during development, regeneration and diseaseDevelopment2015142193253326226443630
  • JugeNMithenRFTrakaMMolecular basis for chemoprevention by sulforaphane: a comprehensive reviewCell Mol Life Sci20076491105112717396224
  • LenziMFimognariCHreliaPSulforaphane as a promising molecule for fighting cancerCancer Treat Res201415920722324114482
  • ParnaudGLiPCassarGMechanism of sulforaphane-induced cell cycle arrest and apoptosis in human colon cancer cellsNutr Cancer200448219820615231455
  • ChoiYHROS-mediated activation of AMPK plays a critical role in sulforaphane-induced apoptosis and mitotic arrest in AGS human gastric cancer cellsGen Physiol Biophys2018370212914029593120
  • LanHYuanHLinCSulforaphane induces p53-deficient SW480 cell apoptosis via the ROS-MAPK signaling pathwayMol Med Rep20171657796780428944886
  • ParkSKimGBaeS-JYooYChoiYInduction of apoptosis by isothiocyanate sulforaphane in human cervical carcinoma HeLa and hepatocarcinoma HepG2 cells through activation of caspase-3Oncol Rep200718118118717549366
  • LeeC-SUChoH-JIJeongYUN-JIsothiocyanates inhibit the invasion and migration of C6 glioma cells by blocking FAK/JNK-mediated MMP-9 expressionOncol Rep20153462901290826397194
  • Negrette-GuzmánMHuerta-YepezSVegaMISulforaphane induces differential modulation of mitochondrial biogenesis and dynamics in normal cells and tumor cellsFood Chem Toxicol20171009010227993529
  • IslamSSMokhtariRBAkbariPHatinaJYegerHFarhatWASimultaneous targeting of bladder tumor growth, survival, and epithelial-to-mesenchymal transition with a novel therapeutic combination of acetazolamide (AZ) and sulforaphane (SFN)Target Oncol201611220922726453055
  • KarmakarSChoudhuryS RoyBanikNLRaySKActivation of multiple molecular mechanisms for increasing apoptosis in human glioblastoma T98G xenograftJ Cancer Sci Ther20100204107113
  • JinC-YMolagodaIMNKarunarathneWAHMTRAIL attenuates sulforaphane-mediated Nrf2 and sustains ROS generation, leading to apoptosis of TRAIL-resistant human bladder cancer cellsToxicol Appl Pharmacol201835213214129792947
  • ŻuryńALitwiniecASafiejko-MroczkaBThe effect of sulforaphane on the cell cycle, apoptosis and expression of cyclin D1 and p21 in the A549 non-small cell lung cancer cell lineInt J Oncol20164862521253327035641
  • ShankarSGanapathySSrivastavaRKSulforaphane enhances the therapeutic potential of TRAIL in prostate cancer orthotopic model through regulation of apoptosis, metastasis, and angiogenesisClin Cancer Res200814216855686618980980
  • ParkHSHanMHKimG-YSulforaphane induces reactive oxygen species-mediated mitotic arrest and subsequent apoptosis in human bladder cancer 5637 cellsFood Chem Toxicol20146415716524296129
  • WangNWangWLiuCJinJShaoBShenLInhibition of growth and induction of apoptosis in A549 cells by compounds from oxheart cabbage extractJ Sci Food Agric201696113813382026679410
  • ChoiSLewKLXiaoHD,L-Sulforaphane-induced cell death in human prostate cancer cells is regulated by inhibitor of apoptosis family proteins and Apaf-1Carcinogenesis200728115116216920735
  • GubernAJoaquinMMarquèsMThe N-terminal phosphorylation of RB by p38 bypasses its inactivation by CDKs and prevents proliferation in cancer cellsMol Cell2016641253627642049
  • RoystonKJPaulBNozellSRajbhandariRTollefsbolTOWithaferin A and sulforaphane regulate breast cancer cell cycle progression through epigenetic mechanismsExp Cell Res20183681677429689276
  • PsurskiMJanczewskiŁukaszŚwitalskaMNovel phosphonate analogs of sulforaphane: synthesis, invitro and invivo anticancer activityEur J Med Chem2017132638028342398
  • ClarkeJDHsuAYuZDashwoodRHHoEDifferential effects of sulforaphane on histone deacetylases, cell cycle arrest and apoptosis in normal prostate cells versus hyperplastic and cancerous prostate cellsMol Nutr Food Res2011557999100921374800
  • HsuAWongCPYuZWilliamsDEDashwoodRHHoEPromoter de-methylation of cyclin D2 by sulforaphane in prostate cancer cellsClin Epigenetics2011313:322303414
  • ChatterjeeSRheeY-HAhnJ-CSulforaphene–carboplatin combination synergistically enhances apoptosis by disruption of mitochondrial membrane potential and cell cycle arrest in human non-small cell lung carcinomaJ Med Food201619986086927467015
  • BryantCSKumarSChamalaSSulforaphane induces cell cycle arrest by protecting RB–E2F-1 complex in epithelial ovarian cancer cellsMol Cancer2010914720196847
  • ChangC-CHungC-MYangY-RLeeM-JHsuY-CSulforaphane induced cell cycle arrest in the G2/M phase via the blockade of cyclin B1/CDC2 in human ovarian cancer cellsJ Ovarian Res2013614123799914
  • JakubíkováJSedlákJMithenRBaoYRole of PI3K/Akt and MEK/ERK signaling pathways in sulforaphane- and erucin-induced Phase II enzymes and MRP2 transcription, G2/M arrest and cell death in Caco-2 cellsBiochem Pharmacol200569111543155215896333
  • YangSHChienCMChangLSLinSRInvolvement of c-jun N-terminal kinase in G2/M arrest and caspase-mediated apoptosis induced by cardiotoxin III (Naja naja atra) in K562 leukemia cellsToxicon200749796697417368702
  • PengXZhouYANTianHUASulforaphane inhibits invasion by phosphorylating ERK1/2 to regulate E-cadherin and CD44v6 in human prostate cancer DU145 cellsOncol Rep20153431565157226134113
  • PappaGBartschHGerhäuserCBiphasic modulation of cell proliferation by sulforaphane at physiologically relevant exposure times in a human colon cancer cell lineMol Nutr Food Res200751897798417628879
  • PandeyMKGuptaSCNabavizadehAAggarwalBBRegulation of cell signaling pathways by dietary agents for cancer prevention and treatmentSemin Cancer Biol20174615818128823533
  • MyzakMCDashwoodWMOrnerGAHoEDashwoodRHSulforaphane inhibits histone deacetylase in vivo and suppresses tumorigenesis in Apc-minus miceFaseb J200620350650816407454
  • ZhangCSuZ-YKhorTOShuLKongA-NTSulforaphane enhances Nrf2 expression in prostate cancer TRAMP C1 cells through epigenetic regulationBiochem Pharmacol20138591398140423416117
  • ShapiroTAFaheyJWDinkova-KostovaATSafety, tolerance, and metabolism of broccoli sprout glucosinolates and isothiocyanates: a clinical Phase I studyNutr Cancer2006551536216965241
  • CornblattBSYeLDinkova-KostovaATPreclinical and clinical evaluation of sulforaphane for chemoprevention in the breastCarcinogenesis20072871485149017347138
  • AlumkalJJSlottkeRSchwartzmanJA Phase II study of sulforaphane-rich broccoli sprout extracts in men with recurrent prostate cancerInvest New Drugs201533248048925431127
  • LozanovskiVJHoubenPHinzUHackertTHerrISchemmerPPilot study evaluating broccoli sprouts in advanced pancreatic cancer (POUDER trial) – study protocol for a randomized controlled trialTrials201415120424894410