Next Article in Journal
Herbal Tea for the Management of Pharyngitis: Inhibition of Streptococcus pyogenes Growth and Biofilm Formation by Herbal Infusions
Previous Article in Journal
Perspective on Adenoviruses: Epidemiology, Pathogenicity, and Gene Therapy
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

The Glucosinolates: A Sulphur Glucoside Family of Mustard Anti-Tumour and Antimicrobial Phytochemicals of Potential Therapeutic Application

1
Honorary Senior Research Associate, Raymond Purves Bone and Joint Research Laboratory, Kolling Institute of Medical Research, Royal North Shore Hospital, Faculty of Medicine and Health, The University of Sydney, St. Leonards, NSW 2065, Australia
2
Graduate School of Biomedical Engineering, Faculty of Engineering, University of New South Wales, Sydney, NSW 2052, Australia
3
Sydney Medical School, Northern, Royal North Shore Hospital, St. Leonards, NSW 2065, Australia
Biomedicines 2019, 7(3), 62; https://doi.org/10.3390/biomedicines7030062
Submission received: 25 July 2019 / Revised: 15 August 2019 / Accepted: 17 August 2019 / Published: 19 August 2019
(This article belongs to the Section Drug Discovery and Development)

Abstract

:
This study reviewed aspects of the biology of two members of the glucosinolate family, namely sinigrin and glucoraphanin and their anti-tumour and antimicrobial properties. Sinigrin and glucoraphanin are converted by the β-sulphoglucosidase myrosinase or the gut microbiota into their bioactive forms, allyl isothiocyanate (AITC) and sulphoraphanin (SFN) which constitute part of a sophisticated defence system plants developed over several hundred million years of evolution to protect them from parasitic attack from aphids, ticks, bacteria or nematodes. Delivery of these components from consumption of cruciferous vegetables rich in the glucosinolates also delivers many other members of the glucosinolate family so the dietary AITCs and SFN do not act in isolation. In vitro experiments with purified AITC and SFN have demonstrated their therapeutic utility as antimicrobials against a range of clinically important bacteria and fungi. AITC and SFN are as potent as Vancomycin in the treatment of bacteria listed by the World Health Organisation as antibiotic-resistant “priority pathogens” and also act as anti-cancer agents through the induction of phase II antioxidant enzymes which inactivate potential carcinogens. Glucosinolates may be useful in the treatment of biofilms formed on medical implants and catheters by problematic pathogenic bacteria such as Pseudomonas aeruginosa and Staphylococcus aureus and are potent antimicrobials against a range of clinically important bacteria and fungi. The glucosinolates have also been applied in the prevention of bacterial and fungal spoilage of food products in advanced atmospheric packaging technology which improves the shelf-life of these products.

1. Introduction

Plants produce a myriad of phytochemicals and many of these have valuable nutritive, medicinal and health promoting properties [1,2,3] (Table 1, Table 2 and Table 3). Anecdotal evidence often points to these beneficial properties however in this report we will concentrate on two members of the glucosinolates (Figure 1), Glucoraphanin and Sinigrin, with a very extensive scientific and nutritional literature illustrating their potential therapeutic applications [3,4,5,6,7,8,9,10,11,12] (Table 2).
Cruciferous plants such as those listed in Table 1 represent important components of a healthy diet and have characteristic spicy flavor profiles which are appealing to many and have important effects in several physiological processes.

2. The Natural Anti-Microbial Activity of Glucosinolate Rich Foods

When plant tissues are damaged, myrosinase, a β-thioglucosidase converts the glucosinolates (Figure 1) to nitriles, thiocyanates and isothiocyanates (Figure 2) which have potent antimicrobial activity with the isothiocyanates in particular displaying potent antibacterial and anti-fungal activity profiles (Figure 3). The inclusion of dietary cruciferous vegetables rich in the glucosinolates may counter antibiotic-resistant bacteria in the food chain arising from the overuse of antibiotics in animal rearing practices. The traditional use of mustard-derived flavoring condiments, while contributing desirable flavor profiles to cooked food items, also provides food preservative properties which traditional societies have relied upon in the prevention of microbial spoilage of foods [13]. This is particularly important in climatic conditions and ambient temperatures conducive to microbial growth leading to food spoilage. Until relatively recently, these societies did not have access to refrigerated storage facilities thus mustard seed products played an important role in food preservation. Mustard seed oil is a potent source of bioactive glucosinolates and represents approximately 30% of the edible oil market in SE Asia. The widespread use of this oil has positively contributed to food storage properties and protection from microbial infection [13,14]. The glucosinolates and their derivatives are volatile compounds and this property has been applied in modern gaseous food packaging technology to extend the shelf-life of food products [14,15]. All cereals have fungal spores associated with the grain surface and the husk, thus whole milled cereal flours used for bread production contain fungal spores. These are inactivated during the baking process; however, fungal spoilage of bread and bakery products can still occur in the post-baking storage and or processing of bakery products. Rape seed oil or mustard flour have been evaluated in bread production to minimize fungal spoilage [16], the major active glucosinolates in rape seed brown mustard (Brassica juncea) oil are AITC (85%) and butenyl isothiocyanate (10%) [13] and these have broad fungicidal activity (Figure 3). In the bakery environment, 2 ppm AITC inhibited the growth of Penicillium roqueforti, P. corylophilum, Eurotium repens, A. flavus and Endomyces fibuliger on rye bread stored in an airtight environment [17]. Modified atmospheric packaging formats (85% CO2, 1% O2) combined with mustard oil vapour packaging has been used to extend the storage properties of bread and bakery products [18] and enhances the potency of AITC as a preservative [18,19].

3. The Brassicaceae Family of Plants

As already indicated, the Brassicaceae are a rich source of sulphur glucoside glucosinolates, these impart a characteristic spicy flavor profile to these vegetables. Glucosinolates have been classified into three categories on the basis of their amino acid precursors (i) aliphatic (e.g., glucoraphanin; Ala, Leu, Ileu, Val, Met), (ii) indole (e.g., glucobrassicin; Trp), and (iii) aromatic (e.g., gluconasturtiin; Phe, Tyr). While ~130 glucosinolates have been identified to date, in a survey of 2,121 German participants in the European Prospective Investigation into Cancer and Nutrition (EPIC study), only five of these glucosinolates were commonly found in the human diet, glucobrassicin, sinigrin, glucoraphasatin (dehydroerucin), glucoraphanin, and glucoiberin [20].
Glucosinolates have only been found in dicotyledonous plants occurring mainly in the Capparales order (Figure 1 and Figure 2) including cruciferous vegetables and the mustards Brassica juncea (brown mustard) [21], Brassica napus. (rape seed) and the popular Japanese condiment horseradish Wasabi (Eutrema japonicum or Wasabia japonica) [22,23] (Table 1). The glucosinolates are stored in a concentrated form in the seed heads and are extracted in cold pressed oils but are also components of the stem and leaves of these plants (Figure 4).
The mustard plant, rapeseed, yellow, white and brown mustards are widely distributed and have a characteristic yellow flower head (Figure 5). Rapeseed (Brassica napus), also known as rape, oilseed rape [25] is a member of the Brassicacea, mustard or cabbage family named from the Latin word for turnip, rapum [26]. This is an ancient plant known of since Biblical times, which has even been identified in the fossil record of the Mesozoic era/mid-Devonian period in Western China. Identification of fossil remains in food cooking implements suggest that mustard seeds may have been the first ever condiment used to flavor food by prehistoric man [27]. Plant evolutionary studies show that the mustard seed plant was of fundamental importance to the subsequent evolution of most other modern day cultivated plants. The leaves, seeds, and roots of wild mustard Cleome viscosa have all been widely used in traditional and folkloric medicine for generations. In Ayurvedic medicine mustard was reported to have many beneficial properties, subsequent scientific and pharmacological studies verified it’s antimicrobial, analgesic, anti-inflammatory, antipyretic, anti-diabetic and hepatoprotective qualities [28,29,30,31]. Subsequent studies have identified the phytochemicals responsible for these activities as shown in the present study; glucosinolates are prominently represented on this list of bioactive compounds.
Brassica napus was botanically described and published in Species Plantarum by Carl Linnaeus, who introduced the binomial name Brassica napus for the first time in 1753 [25] (Figure 5).
Rapeseed oil is one of the oldest known vegetable oils, but historically has been used in limited quantities as a food item due to its high levels of erucic acid, natural rapeseed oil can contain up to 54% w/v erucic acid [32]. Rapeseed cultivated for food production typically contains ~0.5–5% w/v erucic acid. Erucic acid (C22H42O2) is a C22 chain mono-unsaturated omega-9-fatty acid. A strain of mustard subsequently developed with low erucic acid and glucosinolate levels, Canola, a contraction of the terms “Canada” and “ola”, is a low erucic acid, low glucosinolate rapeseed [33]. Canola oil is limited by government regulation to a maximum of 2% w/v erucic acid in the USA and 5% w/v in the EU. In 1992, the health promoting properties of rapeseed oil gained publicity in the George Miller feature film “Lorenzo’s Oil” starring Nick Nolte and Susan Sarandon, which documented the work of a British chemist, Don Suddaby, and Augusto Odone in 1985 who developed a blend of rapeseed and olive oils which halted the progression of Adrenoleukodystrophy, a genetic disorder characterized by an enzyme abnormality resulting in the build-up of toxic fatty acid levels in the brain damaging the myelin sheaths impairing neuronal function and resulting in convulsions, seizures and hyperactivity. The antioxidant properties of activated glucosinolate compounds are also conducive to the maintenance of brain health [34,35,36,37,38,39,40,41,42,43,44]. The brain is a fatty acid rich tissue and particularly prone to redox ROS mediated mitochondrial damage during neuroinflammation [45,46].

4. Public Health Concern over the Impact of Antibiotic-Resistant Bacteria

There is considerable current-day public concern about the overuse of antibiotics in husbandry practice in order to maintain animal health and commercial output levels. The emergence of antibiotic-resistant organisms in humans is related to this agricultural practice. This has been acknowledged by the WHO and by the publication of government guidelines on the use and abuse of antibiotics in agricultural practice. The publication of a list of antibiotic-resistant pathogenic bacteria of particular concern by the WHO (Table 3), and the allocation of major research funds to national agencies in the USA, Canada and Australia to address the problem of antibiotic-resistant bacteria, testifies to the significant threat these organisms represent to human health.

4.1. Treatment of Antibiotic-Resistant Bacterial Infections

Antibiotics and antimicrobial medications have been a mainstay in the treatment of infectious diseases for over 70 years and have been an essential component in healthcare practice to combat bacterial and fungal infections. Widespread use of antibiotics and indeed their over-prescription in healthcare circles, plus an overuse of antibiotics in animal rearing practices in agriculture, has led to infectious organisms being widely exposed to these compounds and, as a consequence, this has actually selected for organisms which have developed a resistance to these compounds and these strains of bacteria and fungi now represent a significant healthcare risk on a global scale. An estimated 2 million patients have become infected with antibiotic-resistant bacterial strains in the USA and as a consequence 23,000 deaths were recorded directly arising from these bacterial infections. Multi-drug-resistant bacterial infections were also responsible for an estimated 25,000 deaths per year in the EEC in 2015–2017 and these cost €1.5 billion per year in healthcare treatment and lost productivity. If these current infection rates are not reversed then 10 million deaths globally per year are predicted by 2050 (317,000 in the USA; 392,000 in S. America; 392,000 in EEC; 4.1 million in Africa; 4.7 million in Asia and 22,000 in Australia). Moreover, it is estimated that additional hospital costs per patient will be in the order of 10,000–40,000 $US in OECD countries. Furthermore, the associated impact of lost economic output due to increased mortality, prolonged sickness and reduced labour efficiency may effectively double this figure. In vitro studies on the activated thiocyanates, isothiocyanates and nitrile compounds generated from the glucosinolates by myrosinase demonstrate these are suitable compounds for antibacterial and anti-fungal evaluations in the treatment of such infections (Figure 3). Furthermore, some of these plant compounds synergise with existing antibiotic treatment protocols (gentamycin, vancomycin) and may represent a useful adjunct to these treatments [47]. Listeria monocytogenes and Staphylococcus aureus in particular were significantly inhibited by benzylisothiocyanate and 2-phenylethylisothiocyanate in isolation or in phytochemical-antibiotic combinations.

4.2. Commercial Development of Antibiotics

Antimicrobial resistance is a global crisis that threatens the public healthcare system. Development of novel antibiotic products is a critical component to combatting antimicrobial infections [48]. A survey of all participants and interested parties in antibiotic research in 2015 in the European Union was undertaken to develop new economic incentives to stimulate greater antibacterial drug innovation [48]. Unfortunately, the financial incentives to undertake such research have proven to be insufficient for many major pharmaceutical companies to maintain investment in antibiotic research programs. The announcement by Novartis of its intention to exit from all antibiotic research in 2018 joined AstraZeneca, Sanofi, and Allergan who also exited from this type of research due to the high cost of undertaking such research coupled with a lack of financial return. This leaves Merck, Roche, GlaxoSmithKline, and Pfizer as the remaining pharmaceutical companies that have continuing active antibiotic research programs. An editorial in Nature Biotechnology in 2018 entitled “Wanted: a reward for antibiotic development” summarised findings of the Nature conference “Countering Antimicrobial Resistance” held in Beijing, China in 2018 which showcased a biodiverse array of discovery approaches currently being undertaken globally to combat drug-resistant bacteria [49]. Even so, development of new antibiotics has dwindled to dangerously low levels in the past three decades. It is well recognised that this lack of innovation has perilous consequences on available treatments for pathogenic infections to the detriment of patient care. This has resulted in the formation of several government agencies and collaborative platforms to support the discovery of new antibiotics.
The European Observatory on Health Systems and Policy has been assembled to support the development of new therapeutic antibiotics and contains members from The European WHO Regional Office, European Governments (Austria, Belgium, Finland, Ireland, Norway, Slovenia, Sweden, the UK, Veneto Region in Italy), European Commission, World Bank, National Union of Health Insurance Funds in France, Schools of Economics and Political Science; Hygiene and Tropical Medicine in London, UK. The Observatory has a secretariat in Brussels and hubs in London and Technical University of Berlin. A 133-page report issued in 2016 by Renwick, Simpkin and Mossialos entitled “Targeting innovation in antibiotic drug discovery and development: The need for a One Health – One Europe – One World Framework” [50] recommends several initiatives to promote antibiotic research including financial incentives, R&D support, effective coordination and dissemination of findings from European agencies, effective collaboration with outside agencies and preclinical support [50].
The pharmaceutical industry has not released any new antibiotic formulations for over three decades, and this has resulted in an alarming incidence of deaths resulting directly from antibiotic-resistant bacteria. The World Health Organisation (WHO) has publicised this as a major public healthcare issue, indicating the real possibility that without new antibiotic treatments becoming available, we may be entering an era when even previously treatable bacterial infections will become life-threatening. The repurposing of anti-cancer drugs for the treatment of bacterial infections has been suggested since some of these have proven to be effective in vitro for the elimination of recalcitrant, multidrug tolerant bacteria, while other antibiotics are useful as anti-cancer compounds [51,52,53,54]. Among the most harmful human pathogenic bacteria, Staphylococcus aureus (Golden Staph) stands out as one of the most virulent and troublesome due to its ability to cause life-threatening infections and to readily adapt to changing environmental conditions [55,56]. The ability of S.aureus to establish itself in various community home and hospital environments, and its resistance to antibiotic treatment make this an important healthcare threat [57]. The emergence of methicillin resistant S.aureus (MRSA) almost five decades ago demonstrates the serious nature of such infections. Hospital environments are conducive to S.aureus colonisation and its virulence is a major threat particularly to patients with reduced immune function. Particularly virulent strains of Enterococcus, resistant to conventional antibiotic treatment, have also emerged in hospitalized patients [58]. Of particular concern are the vancomycin-resistant enterococci (VRE), that lead to infections of the urinary tract associated with prolonged catheter use or to catheter mediated bloodstream infections [59]. There is therefore an increasing global interest in the identification of bioactive compounds from plant sources, which display antibacterial and anti-fungal properties that are pharmacologically effective but which display limited or no side effects. The glucosinolates produced by the Brassicacea family, order Capparales contain compounds with potent antibacterial, anti-fungal, anti-nematodicidal, anti-viral and insecticidal properties making them obvious candidates in the search for compounds to counter bacterial infections [4,10,11,60,61,62,63,64,65,66]. Morever, many of the glucosinolates act synergistically with existing antibiotic regimens improving their effectiveness [47,63]. A list of antibiotic-resistant “priority pathogens” published by WHO in 2017 covers 12 bacterial families posing the greatest threat to human health [67] and highlights Gram-negative bacteria resistant to multiple antibiotics which threaten global public health, these have been referred to as Super-bugs [68,69,70].
The effective antibiotics available for the treatment of bacterial infections are relatively small in number and in many cases have become largely ineffective. The last time a new antibiotic was released on to the world market was approximately 30 years ago, there is a strong need for antibiotic development and a world market eagerly awaiting this product. The WHO has established three treatment categories based on the urgency for new antibiotics: these are critical, high and medium priority (Table 3). The WHO have categorized critical, high, and medium priority treatment areas for antibiotic-resistant bacterial strains (Table 3) and have identified hospital and nursing home patients with reduced immune function as being a particularly susceptible group to such infections. Patients who regularly use ventilators, dialysis machines and medications requiring long-term administration by catheter are liable to become infected with strains of Acinetobacter, Pseudomonas, Klebsiella, E. coli, Serratia, and Proteus that are capable of causing severe and often deadly bloodstream infections and the development of pneumonia. Unfortunately, bacterial strains have emerged which are no longer responsive to the carbapanem and third generation cephalosporins, which were previously the most effective compounds used to treat such infections. Gonorrhoea is rapidly becoming a condition which will soon become untreatable.
The common practice of routine administration of broad-spectrum antibiotics to treat an infection before identification of the specific pathogen responsible for an infection has proven to be an ill-advised practice. By removing the normal bacterial microflora, antibiotics actually provide an opportunistic niche for the emergence of antibiotic-resistant bacterial strains which no longer have to compete with the normal bacterial populations present in the body [71,72]. Klebsiella pneumoniae is a Gram negative, facultative anaerobic commensal microorganism that can cause chronic urinary tract and soft tissue infections, pneumonia, and sepsis, and mostly occurs in immunocompromised patients [73]. Klebsiella pneumoniae normally colonises the mouth, skin, and intestines. Illness predominantly affects middle-aged and older men with debilitating diseases but infants are increasingly now being reported with this organism in urinary tract and intestinal infections [74,75,76]. The emergence of multi-drug-resistant bacterial infections in hospitalized patients with underlying morbidity is of particular concern [77]. In a recent US epidemiology study [72] 25% of K. pneumoniae infections in long-term acute care hospitals were resistant to carbapenems, which are currently used to treat penicillin-resistant Gram-negative pathogens. The most common condition caused by Klebsiella bacteria outside the hospital environment is pneumonia, bronchopneumonia and bronchitis and has a death rate around 50%, even when antimicrobial therapy is administered. Gram negative Enterobacteriaceae bacteria such as Klebsiella have evolved β-lactamase genes which counter the effectiveness of carbapanem as an antibiotic and have resulted in the rapid spread of such antibiotic-resistant bacteria worldwide [78]. Transmissible carbapenem-resistant Enterobacteriaceae have been recognised for the last two decades; however, the global dissemination of these bacteria is a more recent pandemic and is now recognised to be occurring at an alarming pace [79]. Identification of Klebsiella pneumoniae carbapenamase-producing K. pneumonia as a deadly pathogen is of particular concern due to the rise of its global incidence in pediatric and neonatal intensive care units [80,81,82].

5. WHO, United Nation and World Bank Programmes and Coordinated Interagency Collaborations Designed to Combat Antibiotic-Resistant Bacteria

A number of National and International Government initiated and private sector organisations have emerged which aim to investigate better methods to combat the threat of drug-resistant bacterial strains. Stated operational areas, research objectives and areas of expertise for each organisation demonstrate a diverse approach to combating this global public health concern and some areas of overlap, however in order to avoid duplication, redundancy and areas of wasted effort further agencies have also been set up to co-ordinate and disseminate research findings to all interested parties. Independent private commercial organisations are also actively engaged in the search for effective treatments for antibiotic-resistant organisms to service an immense global market.
The Global Antimicrobial Resistance Surveillance System (GLASS) is a WHO initiative which was established to support a systematic approach to the collection, analysis and dissemination of antimicrobial resistance data at a global level to facilitate informed decision-making, at the local, national and regional action areas. GARDP, The Global Antibiotic Research and Development Partnership was also formed by WHO and DNDi, Drugs for Neglected Diseases initiative. GARDP undertakes research and development through public-private partnerships. IACG, Interagency Coordination Group on Antimicrobial Resistance is an initiative of the United Nations Secretary-General which was established to improve coordinated efforts between international organisations and to ensure effective global health security activity [83].
CARB-X has indicated its intention to release up to four new antibacterial treatments based on modifications of current antibiotics in the next 4 years. The UN Deputy Secretary-General and WHO Director General are co-chairs on IACG along with executive members from several other UN and international agencies and acknowledged experts from several industry sectors.
The Centers for Disease Control and Prevention (CDC) and related US agencies are also actively involved in several measures to combat antibiotic-resistant bacterial infections through a collaborative global approach across all government and private sector agencies. CDC has published “CDC. The Core Elements of Human Antibiotic Stewardship Programs in Resource -Limited Settings: National and Hospital Levels. Atlanta, GA: US Department of Health and Human Services, CDC; 2018. Available at: https://www.cdc.gov/antibiotic-use/healthcare/implementation.html” (accessed 14 July 2019) to help improve guidelines for antibiotic use in healthcare settings worldwide. The Food and Drug Administration (FDA) has also announced plans to combat antibiotic resistance through innovative antibiotic developments and the coordinated use of antibiotics in human medicine and in animal husbandry practice. CARB-X, a global non-profit partnership launched in 2016, is dedicated to accelerating antibacterial research to counter the global impact of drug-resistant bacteria This organisation, led by Boston University is currently funding 33 projects in N. America, Europe and Asia. CARB-X is funded by BARDA, The US Department of Health and Human Services Biomedical Advanced Research and Development Authority which is part of the Office of the Assistant Secretary for Preparedness and Response (ASPR). Other organisations which form part of the CARB-X initiative include The Wellcome Trust, a global UK based charity working to improve global health. BMBF, Germany’s Federal Ministry of Education and Research and the United Kingdom, The Global Antimicrobial Resistance Innovation Fund based in the United Kingdom (UK GAMRIF), and world’s largest humanitarian Foundation (Bill and Melinda Gates Foundation) also contribute funding to CARB-X and National Institute of Allergy and Infectious Diseases (NIAID) and US National Institutes of Health (NIH) also provide in-kind support. CARB-X has reported that they will invest >$500 US million by 2021 into the development of antibiotics to combat the deadliest super-bugs, and develop vaccines, rapid diagnostics, and other life-saving products to aid in the treatment of antibiotic-resistant bacteria. This supports The US National Action Plan for Combatting Antibiotic-Resistant Bacteria, AR: https://www.cdc.gov/DrugResistance/us-activities.html. (accessed 2 July 2019) Strategies being developed in Australia to combat bacterial resistant infections involve a unified approach by all government and private agencies to combat the threat of antibiotic overuse and development of antibiotic-resistant bacterial infections.

6. Application of the Myrosinase-Glucosinolate System in Biomedicine

The bioactivity of glucosinolate hydrolysis products and potential biomedical applications are well documented (Table 3, Table 4, Table 5 and Table 6). SFN has roles in cancer prevention, high blood pressure, macular degeneration and stomach ulcers and is a potent inducer of mammalian phase II detoxication enzyme systems which deactivate and excrete many carcinogens. The induction of NAD(P)H quinone reductase, heme oxygenase 1 (HO-1), glutamate-cysteine ligase catalytic subunit, and glutathione S transferases occurs through the Keap1-Nrf2-ARE cell signaling pathway [84,85,86]. Numerous studies in human colon, leukemia, pancreatic, lung, and skin cancer cell lines have demonstrated SFN’s inhibitory effects on cell cycle arrest [12,87,88,89] and elevated apoptosis in human bladder [90] and prostate [91] cell lines. Sulforaphane’s ability to disrupt tubulin and actin polymerization, inhibits mitotic spindle formation and tumour cell growth in animal models of breast cancer [92,93] and also inhibits histone deacetylase, increasing apoptosis in human colon, prostate, and kidney cell lines [94,95,96,97].

6.1. The Bioactivity of Glucosinolates

The glucosinolates are benign molecules requiring conversion by myrosinase to bioactive thiocyanate, isothiocyanate and nitrile derivatives (Figure 2). Therefore, glucoraphanin and sinigrin are converted into bioactive SFN and AITCs with fungicidal, bactericidal, nematocidal, antioxidant and anti-cancer properties. Biofilm formation on medical devices and implants such as catheters, mechanical heart valves, pacemakers, prosthetic joints, and contact lenses pose a critical medical problem. The most common biofilm-forming bacteria include Enterococcus faecalis, Staphylococcus aureus, Staphylococcus epidermidis, Streptococcus viridans, Escherichia coli, Klebsiella pneumoniae, Proteus mirabilis, and Pseudomonas aeruginosa [98,99,100,101,102,103], S. aureus and S. epidermidis are most commonly found on cardiovascular devices [104,105,106], it estimated that 40%–50% of prosthetic heart valve infections, and 50%–70% of catheter biofilm infections are due to these bacteria [107,108]. Despite the evaluation of a wide range of anti-fouling compounds [103,109,110] improvements are still required in this area. Glucosinolates have proven useful in the prevention of biofilm development by Pseudomonas aeruginosa [5,111,112,113].
Cooking of cruciferous vegetables inactivates myrosinase activity however the gut microbiota in humans may provide myrosinase activity and lead to absorption of SFN and AITCs in the intestine. A diet rich in cruciferous vegetables is associated with a lower risk of developing breast, lung, prostate, and colorectal cancer [114,115,116,117,118]. It is important to control the redox balance in the human brain to control neuronal mitochondrial activity, oxidant stress on mitochondria can diminish neuronal energetics and promote neurodegeneration in Parkinson’s and Alzheimers’s disease [119]. Brain tissue is very rich in fatty acids and is especially sensitive to the action of free radical oxidant activity. The GSTs are ROS scavengers and are neuroprotective [119,120,121].

6.2. Cancer and Dietary SFN and AITC Levels

Meta analyses of clinical trials on dietary glucosinolates have generally provided promising but not compelling evidence of the efficacy of these as anti-oxidants or anti-cancer agents despite positive in vitro findings in cell culturing experiments and may reflect the inefficiencies of the dietary route for delivery of these compounds. Positive effects are generally achieved in vitro with concentrations of the active glucosinolate components in the 1–40 µmol range. It is unlikely that this level of therapeutic agent would be delivered successfully to the target tumour cells in vivo by the diet. Attempts have been made to increase the glucosinolate content of broccoli hybrids, broccoli sprouts are also richer sources of the glucosinolates, particularly since these are consumed uncooked thus endogenous myrosinase is not inactivated by the cooking process and it has time to convert the glucosinolates to bioactive forms during food mastication. The detection of SFN and AITCs excreted in urine and faecal matter following consumption of cooked cruciferous vegetables where the endogenous myrosinase is inactivated in the initial cooking stages, indicates that the gut microbiota are another source of myrosinase activity. Therefore, therapeutic doses of SFN and AITCs are likely achievable to target tumour cells in the colon [104,210,211], prostate [91,95,171,172,173] and bladder [162,188,189,190,191,192,193]. Dietary glucosinolates are also effective in the treatment of gastric H.Pylori infections and gastric cancer. The delivery of therapeutic doses of dietary SFN and AITCs through the systemic circulation to pancreatic, ovarian, breast and liver cancer and melanoma, however, is less likely to be as effective and may explain the relatively poor findings of meta analyses of dietary clinical trials on the glucosinolates as anti-cancer agents. In many cases, the statistical power achieved in these analyses has also been reduced by low sample sizes or no associations were established. More high-quality cohort studies with larger sample sizes and well controlled confounding factors are required to confirm the benefit of dietary cruciferous vegetable consumption; initial studies have delivered sufficient evidence to warrant such studies. The bioavailability of glucosinolates following different food processing methods has also been evaluated in order to improve the dietary content of bioactive forms of the glucosinolates [217] Supplementation of the diet with broccoli sprouts or myrosinase containing mustard products have also been examined as a means of increasing the dietary SFN and AITC content [218]. The effective delivery of SFN and AITCs to the target cells in solid tumours is a difficult proposition. Delivery systems based on hyaluronan as a carrier molecule have been developed for several steroids and cytotoxic compounds and successfully treated solid tumours however this methodology has yet to be applied to the delivery of SFN or AITCs in these problematic cancers (reviewed in [219]).

6.3. The Beneficial Bioactivities of Sinigrin and Their Applications in Biomedicine

Although the scientific literature on sinigrin (Table 7) is not as extensive as that of SFN, they share similar bioactivities and areas of application and if supplied as a dietary component will not be acting in isolation anyway [113].

7. Concluding Remarks

The myrosinase-glucosinolate system in plants is a sophisticated protective system that developed over several hundred million years of evolution. With a greater understanding of the system’s component parts, it is now possible to apply this knowledge to human physiological processes, an advance that is of potential benefit in biomedicine. Some of these compounds may be useful in the prevention of fouling of plant equipment, sterilisation of medical implants, wound healing and the prevention of some forms of cancer. The extensive literature documenting the biodiversity of glucosinolate applications in biomedicine indicates considerable promise in future areas of investigation in:
  • Antibiotics, anti-fungal and anti-viral agents
  • Biofilm prevention in medical implants, catheters and industrial plant equipment
  • Nutritive additives with anti-cancer properties
  • Advanced food packaging technology to improve shelf-life of food products.

Funding

No funding was received for this study.

Conflicts of Interest

The author states that he has no financial disclosures or conflicts to report. The author declares that the research was conducted in the absence of any commercial or financial relationships that could be construed as a potential conflict of interest.

Abbreviations

ADAlzheimer’s disease
AKTA serine/threonine-specific protein kinase
AREAntioxidant response element
EPAEnvironment Protection Agency
ESBLExtended Spectrum Beta-Lactamases
Keap-1-Nrf2-AREKelch-like erythroid cell-derived protein with CNC homology (ECH)-associated protein 1–anti-oxidant response element
AITCAllyl isothiocyanate
GARDPGlobal Antibiotic Research and Development Partnership
GSKGlycogen Synthase Kinase
GSTGlutathione-S-transferase
DNDIDrugs for Neglected Diseases initiative
IACGInteragency Coordination Group on Antimicrobial Resistance
LPSLipopolysaccharide
MAPKA mitogen-activated protein kinase
NFκBNuclear factor kappa light chain enhancer of activated B cells
NLRPR3Nucleotide-binding domain and leucine-rich repeat–containing protein 3
NSAIDNon-Steroidal anti-inflammatory
PDGFPlatelet derived growth factor
ROSReactive oxygen species
SMCSmooth muscle cell
TNFTumour necrosis factor-alpha

References

  1. Briones-Herrera, A.; Eugenio-Perez, D.; Reyes-Ocampo, J.G.; Rivera-Mancia, S.; Pedraza-Chaverri, J. New highlights on the health-improving effects of sulforaphane. Food. Funct. 2018, 9, 2589–2606. [Google Scholar] [CrossRef] [PubMed]
  2. Palliyaguru, D.L.; Yuan, J.M.; Kensler, T.W.; Fahey, J.W. Isothiocyanates: Translating the Power of Plants to People. Mol. Nutr. Food Res. 2018, 62, e1700965. [Google Scholar] [CrossRef] [PubMed]
  3. Vanduchova, A.; Anzenbacher, P.; Anzenbacherova, E. Isothiocyanate from Broccoli, Sulforaphane, and Its Properties. J. Med. Food 2019, 22, 121–126. [Google Scholar] [CrossRef]
  4. Aires, A.; Mota, V.R.; Saavedra, M.J.; Rosa, E.A.; Bennett, R.N. The antimicrobial effects of glucosinolates and their respective enzymatic hydrolysis products on bacteria isolated from the human intestinal tract. J. Appl. Microbiol. 2009, 106, 2086–2095. [Google Scholar] [CrossRef] [PubMed]
  5. Baskar, V.; Park, S.W.; Nile, S.H. An Update on Potential Perspectives of Glucosinolates on Protection against Microbial Pathogens and Endocrine Dysfunctions in Humans. Crit. Rev. Food Sci. Nutr. 2016, 56, 2231–2249. [Google Scholar] [CrossRef]
  6. Becker, T.M.; Juvik, J.A. The Role of Glucosinolate Hydrolysis Products from Brassica Vegetable Consumption in Inducing Antioxidant Activity and Reducing Cancer Incidence. Diseases 2016, 4, 22. [Google Scholar] [CrossRef]
  7. Borges, A.; Abreu, A.C.; Ferreira, C.; Saavedra, M.J.; Simoes, L.C.; Simoes, M. Antibacterial activity and mode of action of selected glucosinolate hydrolysis products against bacterial pathogens. J. Food Sci. Technol. 2015, 52, 4737–4748. [Google Scholar] [CrossRef]
  8. Carpenter, E.; Mai, N.; Miranda, C.L.; Reed, R.L.; Stevens, J.F.; Indra, A.K.; Indra, G. Photoprotective Properties of Isothiocyanate and Nitrile Glucosinolate Derivatives from Meadowfoam (Limnanthes alba) against UVB Irradiation in Human Skin Equivalent. Front. Pharmacol. 2018, 9, 477. [Google Scholar] [CrossRef]
  9. Dinkova-Kostova, A.T.; Kostov, R.V. Glucosinolates and isothiocyanates in health and disease. Trends Mol. Med. 2012, 18, 337–347. [Google Scholar] [CrossRef]
  10. Dufour, V.; Alazzam, B.; Ermel, G.; Thepaut, M.; Rossero, A.; Tresse, O.; Baysse, C. Antimicrobial activities of isothiocyanates against Campylobacter jejuni isolates. Front. Cell Infect. Microbiol. 2012, 2, 53. [Google Scholar] [CrossRef]
  11. Dufour, V.; Stahl, M.; Baysse, C. The antibacterial properties of isothiocyanates. Microbiology 2015, 161, 229–243. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  12. Gamet-Payrastre, L.; Li, P.; Lumeau, S.; Cassar, G.; Dupont, M.A.; Chevolleau, S.; Gasc, N.; Tulliez, J.; Terce, F. Sulforaphane, a naturally occurring isothiocyanate, induces cell cycle arrest and apoptosis in HT29 human colon cancer cells. Cancer Res. 2000, 60, 1426–1433. [Google Scholar] [PubMed]
  13. Attokaran, M. Mustard, Natural Food Flavors and Colorants; Wiley-Blackwell: Hoboken, NJ, USA, 2011. [Google Scholar]
  14. Ekanayake, A.; Strife, R.J.; Zehentbauer, G.N.; David, J.R.D. Chapter 98 - Yellow or White Mustard (Sinapis alba L.) Oils. In Essential Oils in Food Preservation, Flavor and Safety; Preedy, V., Ed.; Academic Press: Cambridge, MA, USA, 2016; pp. 857–863. [Google Scholar]
  15. Hyldgaard, M.; Mygind, T.; Meyer, R.L. Essential oils in food preservation: Mode of action, synergies, and interactions with food matrix components. Front. Microbiol. 2012, 3, 12. [Google Scholar] [CrossRef]
  16. Quiles, J.M.; Manyes, L.; Luciano, F.B.; Manes, J.; Meca, G. Effect of the oriental and yellow mustard flours as natural preservative against aflatoxins B1, B2, G1 and G2 production in wheat tortillas. J. Food Sci. Technol. 2015, 52, 8315–8321. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  17. Suhr, K.I.; Nielsen, P.V. Antifungal activity of essential oils evaluated by two different application techniques against rye bread spoilage fungi. J. Appl. Microbiol. 2003, 94, 665–674. [Google Scholar] [CrossRef] [PubMed]
  18. Suhr, K.I.; Nielsen, P.V. Inhibition of Fungal Growth on Wheat and Rye Bread by Modified Atmosphere Packaging and Active Packaging Using Volatile Mustard Essential Oil. J. Food Sci. 2005, 70, M37–M44. [Google Scholar] [CrossRef]
  19. Nielsen, P.V.; Rios, R. Inhibition of fungal growth on bread by volatile components from spices and herbs, and the possible application in active packaging, with special emphasis on mustard essential oil. Int. J. Food Microbiol. 2000, 60, 219–229. [Google Scholar] [CrossRef]
  20. Steinbrecher, A.; Linseisen, J. Dietary intake of individual glucosinolates in participants of the EPIC-Heidelberg cohort study. Ann. Nutr. Metab. 2009, 54, 87–96. [Google Scholar] [CrossRef]
  21. Ngala, B.M.; Haydock, P.P.; Woods, S.; Back, M.A. Biofumigation with Brassica juncea, Raphanus sativus and Eruca sativa for the management of field populations of the potato cyst nematode Globodera pallida. Pest Manag. Sci. 2015, 71, 759–769. [Google Scholar] [CrossRef]
  22. Eutrema japonicum (Miq) Koidz—The plant list. Available online: http://www.theplantlist.org/tpl1.1/record/kew-2805492 (accessed on 31 May 2019).
  23. Wasabia japonica. Multilingual multiscript plant nama database—University of Melbourne. Available online: http://www.plantnames.unimelb.edu.au/ (accessed on 31 May 2019).
  24. Verboven, P.; Herremans, E.; Borisjuk, L.; Helfen, L.; Ho, Q.T.; Tschiersch, H.; Fuchs, J.; Nicolai, B.M.; Rolletschek, H. Void space inside the developing seed of Brassica napus and the modelling of its function. New Phytol. 2013, 199, 936–947. [Google Scholar] [CrossRef]
  25. Linnaeus, C. Classification of Brassica napus. Species Plantarum 1753, 2, 666. [Google Scholar]
  26. Harper, D. Derivation of the name rape-seed. OnlineEtymology Dictionary. 2016. Available online: www.etymonline.com (accessed on 12 May 2019).
  27. Saul, H.; Madella, M.; Fischer, A.; Glykou, A.; Hartz, S.; Craig, O.E. Phytoliths in pottery reveal the use of spice in European prehistoric cuisine. PLoS ONE 2013, 8, e70583. [Google Scholar] [CrossRef] [PubMed]
  28. Grover, J.K.; Yadav, S.; Vats, V. Medicinal plants of India with anti-diabetic potential. J. Ethnopharmacol. 2002, 81, 81–100. [Google Scholar] [CrossRef]
  29. Gupta, R.; Bajpai, K.G.; Johri, S.; Saxena, A.M. An overview of Indian novel traditional medicinal plants with anti-diabetic potentials. Afr. J. Tradit. Complement. Altern. Med. 2007, 5, 1–17. [Google Scholar] [PubMed]
  30. Mali, R.G. Cleome viscosa (wild mustard): A review on ethnobotany, phytochemistry, and pharmacology. Pharm. Biol. 2010, 48, 105–112. [Google Scholar] [CrossRef] [PubMed]
  31. Sheikh, Y.; Maibam, B.C.; Biswas, D.; Laisharm, S.; Deb, L.; Talukdar, N.C.; Borah, J.C. Anti-diabetic potential of selected ethno-medicinal plants of north east India. J. Ethnopharmacol. 2015, 171, 37–41. [Google Scholar] [CrossRef] [PubMed]
  32. Sahasrabudhe, M. Crismer values and erucic scid contents of rapeseed oils. J. Am. Oil Chem. Soc. 1977, 54, 323–324. [Google Scholar] [CrossRef]
  33. Potts, D.; Rakow, G.W.; Males, D.R. Canola quality Brassica juncea, a new oilseed crop for the Canadian prairies. New Horizons for an old crop. In Proceedings of the 10th International Rapeseed Congress, Canberra, Australia, 26–29 September 1999. [Google Scholar]
  34. Chen, N.G.; Chen, K.T.; Lu, C.C.; Lan, Y.H.; Lai, C.H.; Chung, Y.T.; Yang, J.S.; Lin, Y.C. Allyl isothiocyanate triggers G2/M phase arrest and apoptosis in human brain malignant glioma GBM 8401 cells through a mitochondria-dependent pathway. Oncol. Rep. 2010, 24, 449–455. [Google Scholar] [Green Version]
  35. Giacoppo, S.; Galuppo, M.; Iori, R.; De Nicola, G.R.; Bramanti, P.; Mazzon, E. (RS)-glucoraphanin purified from Tuscan black kale and bioactivated with myrosinase enzyme protects against cerebral ischemia/reperfusion injury in rats. Fitoterapia 2014, 99, 166–177. [Google Scholar] [CrossRef]
  36. Giacoppo, S.; Galuppo, M.; Iori, R.; De Nicola, G.R.; Bramanti, P.; Mazzon, E. The protective effects of bioactive (RS)-glucoraphanin on the permeability of the mice blood-brain barrier following experimental autoimmune encephalomyelitis. Eur. Rev. Med. Pharmacol. Sci. 2014, 18, 194–204. [Google Scholar]
  37. Giacoppo, S.; Galuppo, M.; Montaut, S.; Iori, R.; Rollin, P.; Bramanti, P.; Mazzon, E. An overview on neuroprotective effects of isothiocyanates for the treatment of neurodegenerative diseases. Fitoterapia 2015, 106, 12–21. [Google Scholar] [CrossRef] [PubMed]
  38. Guerrero-Beltran, C.E.; Calderon-Oliver, M.; Pedraza-Chaverri, J.; Chirino, Y.I. Protective effect of sulforaphane against oxidative stress: Recent advances. Exp. Toxicol. Pathol. 2012, 64, 503–508. [Google Scholar] [CrossRef] [PubMed]
  39. Sanadgol, N.; Zahedani, S.S.; Sharifzadeh, M.; Khalseh, R.; Barbari, G.R.; Abdollahi, M. Recent Updates in Imperative Natural Compounds for Healthy Brain and Nerve Function: A Systematic Review of Implications for Multiple Sclerosis. Curr. Drug Targets 2017, 18, 1499–1517. [Google Scholar] [CrossRef] [PubMed]
  40. Shirai, Y.; Fujita, Y.; Hashimoto, R.; Ohi, K.; Yamamori, H.; Yasuda, Y.; Ishima, T.; Suganuma, H.; Ushida, Y.; Takeda, M.; et al. Dietary Intake of Sulforaphane-Rich Broccoli Sprout Extracts during Juvenile and Adolescence Can Prevent Phencyclidine-Induced Cognitive Deficits at Adulthood. PLoS ONE 2015, 10, e0127244. [Google Scholar] [CrossRef] [PubMed]
  41. Tarozzi, A.; Angeloni, C.; Malaguti, M.; Morroni, F.; Hrelia, S.; Hrelia, P. Sulforaphane as a potential protective phytochemical against neurodegenerative diseases. Oxid. Med. Cell Longev. 2013, 2013, 415078. [Google Scholar] [CrossRef] [PubMed]
  42. Vauzour, D.; Buonfiglio, M.; Corona, G.; Chirafisi, J.; Vafeiadou, K.; Angeloni, C.; Hrelia, S.; Hrelia, P.; Spencer, J.P. Sulforaphane protects cortical neurons against 5-S-cysteinyl-dopamine-induced toxicity through the activation of ERK1/2, Nrf-2 and the upregulation of detoxification enzymes. Mol. Nutr. Food Res. 2010, 54, 532–542. [Google Scholar] [CrossRef] [PubMed]
  43. Yao, W.; Zhang, J.C.; Ishima, T.; Dong, C.; Yang, C.; Ren, Q.; Ma, M.; Han, M.; Wu, J.; Suganuma, H.; et al. Role of Keap1-Nrf2 signaling in depression and dietary intake of glucoraphanin confers stress resilience in mice. Sci. Rep. 2016, 6, 30659. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  44. Zhang, J.C.; Yao, W.; Dong, C.; Yang, C.; Ren, Q.; Ma, M.; Han, M.; Wu, J.; Ushida, Y.; Suganuma, H.; et al. Prophylactic effects of sulforaphane on depression-like behavior and dendritic changes in mice after inflammation. J. Nutr. Biochem. 2017, 39, 134–144. [Google Scholar] [CrossRef] [PubMed]
  45. Freitas, H.R.; Ferreira, G.D.C.; Trevenzoli, I.H.; Oliveira, K.J.; de Melo Reis, R.A. Fatty Acids, Antioxidants and Physical Activity in Brain Aging. Nutrients 2017, 9, 1263. [Google Scholar] [CrossRef] [PubMed]
  46. Singh, A.; Kukreti, R.; Saso, L.; Kukreti, S. Oxidative Stress: A Key Modulator in Neurodegenerative Diseases. Molecules 2019, 24, 1583. [Google Scholar] [CrossRef] [PubMed]
  47. Dias, C.; Aires, A.; Bennett, R.N.; Rosa, E.A.; Saavedra, M.J. First study on antimicriobial activity and synergy between isothiocyanates and antibiotics against selected Gram-negative and Gram-positive pathogenic bacteria from clinical and animal source. Med. Chem. 2012, 8, 474–480. [Google Scholar] [CrossRef] [PubMed]
  48. Ardal, C.; Baraldi, E.; Theuretzbacher, U.; Outterson, K.; Plahte, J.; Ciabuschi, F.; Rottingen, J.A. Insights into early stage of antibiotic development in small- and medium-sized enterprises: A survey of targets, costs, and durations. J. Pharm. Policy Pract. 2018, 11, 8. [Google Scholar] [CrossRef]
  49. Anonymous. Wanted: A reward for antibiotic development. Nat. Biotechnol. 2018, 36, 555. [Google Scholar] [CrossRef] [PubMed]
  50. Renwick, M.J.; Simpkin, V.; Mossialos, E. Targeting innovation in antibiotic drug discovery and development: The need for a One Health – One Europe – One World Framework; European Observatory on Health Systems and Policies: Copenhagen, Denmark, 2016. Available online: http://www.ncbi.nlm.nih.gov/books/NBK447337/ (accessed on 19 August 2019).
  51. Cruz-Muniz, M.Y.; Lopez-Jacome, L.E.; Hernandez-Duran, M.; Franco-Cendejas, R.; Licona-Limon, P.; Ramos-Balderas, J.L.; Martinez-Vazquez, M.; Belmont-Diaz, J.A.; Wood, T.K.; Garcia-Contreras, R. Repurposing the anticancer drug mitomycin C for the treatment of persistent Acinetobacter baumannii infections. Int. J. Antimicrob. Agents 2017, 49, 88–92. [Google Scholar] [CrossRef] [PubMed]
  52. Rangel-Vega, A.; Bernstein, L.R.; Mandujano-Tinoco, E.A.; Garcia-Contreras, S.J.; Garcia-Contreras, R. Drug repurposing as an alternative for the treatment of recalcitrant bacterial infections. Front. Microbiol. 2015, 6, 282. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  53. Soo, V.W.; Kwan, B.W.; Quezada, H.; Castillo-Juarez, I.; Perez-Eretza, B.; Garcia-Contreras, S.J.; Martinez-Vazquez, M.; Wood, T.K.; Garcia-Contreras, R. Repurposing of Anticancer Drugs for the Treatment of Bacterial Infections. Curr. Top. Med. Chem. 2017, 17, 1157–1176. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  54. Van Nuffel, A.M.; Sukhatme, V.; Pantziarka, P.; Meheus, L.; Sukhatme, V.P.; Bouche, G. Repurposing Drugs in Oncology (ReDO)-clarithromycin as an anti-cancer agent. Ecancermedicalscience 2015, 9, 513. [Google Scholar] [CrossRef] [PubMed]
  55. Chambers, H.F.; Deleo, F.R. Waves of resistance: Staphylococcus aureus in the antibiotic era. Nat. Rev. Microbiol. 2009, 7, 629–641. [Google Scholar] [CrossRef] [PubMed]
  56. Fry, D.E.; Barie, P.S. The changing face of Staphylococcus aureus: A continuing surgical challenge. Surg. Infect. 2011, 12, 191–203. [Google Scholar] [CrossRef]
  57. Velazquez-Meza, M.E.; Hernandez-Salgado, M.; Contreras-Cordero, J.F.; Perez-Cortes, P.; Villarreal-Trevino, L. Surveillance of methicillin-resistant Staphylococcus aureus causing nosocomial infections in five medical centers of Monterrey, Nuevo Leon, Mexico from 2005-2009. Arch. Med. Res. 2013, 44, 570–574. [Google Scholar] [CrossRef]
  58. Reid, K.C.; Cockerill, I.F.; Patel, R. Clinical and epidemiological features of Enterococcus casseliflavus/flavescens and Enterococcus gallinarum bacteremia: A report of 20 cases. Clin. Infect. Dis. 2001, 32, 1540–1546. [Google Scholar] [CrossRef] [PubMed]
  59. Tang, H.J.; Chen, C.C.; Zhang, C.C.; Su, B.A.; Li, C.M.; Weng, T.C.; Chiang, S.R.; Ko, W.C.; Chuang, Y.C. In vitro efficacy of fosfomycin-based combinations against clinical vancomycin-resistant Enterococcus isolates. Diagn. Microbiol. Infect. Dis. 2013, 77, 254–257. [Google Scholar] [CrossRef] [PubMed]
  60. Abreu, A.C.; Borges, A.; Simoes, L.C.; Saavedra, M.J.; Simoes, M. Antibacterial activity of phenyl isothiocyanate on Escherichia coli and Staphylococcus aureus. Med. Chem. 2013, 9, 756–761. [Google Scholar] [CrossRef] [PubMed]
  61. Galuppo, M.; Nicola, G.R.; Iori, R.; Dell’utri, P.; Bramanti, P.; Mazzon, E. Antibacterial activity of glucomoringin bioactivated with myrosinase against two important pathogens affecting the health of long-term patients in hospitals. Molecules 2013, 18, 14340–14348. [Google Scholar] [CrossRef] [PubMed]
  62. Olaimat, A.N.; Holley, R.A. Inhibition of Listeria monocytogenes and Salmonella by combinations of oriental mustard, malic acid, and EDTA. J. Food Sci. 2014, 79, M614–M621. [Google Scholar] [CrossRef] [PubMed]
  63. Saavedra, M.J.; Borges, A.; Dias, C.; Aires, A.; Bennett, R.N.; Rosa, E.S.; Simoes, M. Antimicrobial activity of phenolics and glucosinolate hydrolysis products and their synergy with streptomycin against pathogenic bacteria. Med. Chem. 2010, 6, 174–183. [Google Scholar] [CrossRef] [PubMed]
  64. Saavedra, M.J.; Dias, C.S.; Martinez-Murcia, A.; Bennett, R.N.; Aires, A.; Rosa, E.A. Antibacterial effects of glucosinolate-derived hydrolysis products against enterobacteriaceae and enterococci isolated from pig ileum segments. Foodborne Pathog. Dis. 2012, 9, 338–345. [Google Scholar] [CrossRef] [PubMed]
  65. Sotelo, T.; Lema, M.; Soengas, P.; Cartea, M.E.; Velasco, P. In vitro activity of glucosinolates and their degradation products against brassica-pathogenic bacteria and fungi. Appl. Environ. Microbiol. 2015, 81, 432–440. [Google Scholar] [CrossRef]
  66. Fahey, J.W.; Haristoy, X.; Dolan, P.M.; Kensler, T.W.; Scholtus, I.; Stephenson, K.K.; Talalay, P.; Lozniewski, A. Sulforaphane inhibits extracellular, intracellular, and antibiotic-resistant strains of Helicobacter pylori and prevents benzo[a]pyrene-induced stomach tumors. Proc. Natl. Acad. Sci. USA 2002, 99, 7610–7615. [Google Scholar] [CrossRef]
  67. World Health Organisation. Guidelines for the prevention and control of carbapenem-resistant Enterobacteriaceae, Acinetobacter baumannii and Pseudomonas aeruginosa in health care facilities. 2017. Available online: https://www.who.int/infection-prevention/publications/guidelines-cre/en/ (accessed on 19 August 2019).
  68. Adegoke, A.A.; Faleye, A.C.; Singh, G.; Stenstrom, T.A. Antibiotic Resistant Superbugs: Assessment of the Interrelationship of Occurrence in Clinical Settings and Environmental Niches. Molecules 2016, 22, 29. [Google Scholar] [CrossRef]
  69. Mohammed, N.; Savardekar, A.R.; Patra, D.P.; Narayan, V.; Nanda, A. The 21st-century challenge to neurocritical care: The rise of the superbug Acinetobacter baumannii. A meta-analysis of the role of intrathecal or intraventricular antimicrobial therapy in reduction of mortality. Neurosurg. Focus 2017, 43, E8. [Google Scholar] [CrossRef] [PubMed]
  70. Rello, J.; Kalwaje Eshwara, V.; Lagunes, L.; Alves, J.; Wunderink, R.G.; Conway-Morris, A.; Rojas, J.N.; Alp, E.; Zhang, Z. A global priority list of the TOp TEn resistant Microorganisms (TOTEM) study at intensive care: A prioritization exercise based on multi-criteria decision analysis. Eur. J. Clin. Microbiol. Infect. Dis. 2019, 38, 319–323. [Google Scholar] [CrossRef] [PubMed]
  71. Bassetti, M.; Righi, E.; Carnelutti, A.; Graziano, E.; Russo, A. Multidrug-resistant Klebsiella pneumoniae: Challenges for treatment, prevention and infection control. Expert. Rev. Anti-Infect. Ther. 2018, 16, 749–761. [Google Scholar] [CrossRef] [PubMed]
  72. Han, J.H.; Goldstein, E.J.; Wise, J.; Bilker, W.B.; Tolomeo, P.; Lautenbach, E. Epidemiology of Carbapenem-Resistant Klebsiella pneumoniae in a Network of Long-Term Acute Care Hospitals. Clin. Infect. Dis. 2017, 64, 839–844. [Google Scholar] [CrossRef] [PubMed]
  73. Ruiz, J.; Gordon, M.; Villarreal, E.; Frasquet, J.; Sanchez, M.A.; Martin, M.; Castellanos, A.; Ramirez, P. Influence of antibiotic pressure on multi-drug resistant Klebsiella pneumoniae colonisation in critically ill patients. Antimicrob. Resist. Infect. Control. 2019, 8, 38. [Google Scholar] [CrossRef]
  74. Akturk, H.; Sutcu, M.; Somer, A.; Aydin, D.; Cihan, R.; Ozdemir, A.; Coban, A.; Ince, Z.; Citak, A.; Salman, N. Carbapenem-resistant Klebsiella pneumoniae colonization in pediatric and neonatal intensive care units: Risk factors for progression to infection. Braz. J. Infect. Dis. 2016, 20, 134–140. [Google Scholar] [CrossRef] [PubMed]
  75. Justo-da-Silva, L.H.; De-Azeredo, A.N.; Bueno, A.C.; Montezzi, L.F.; Leobons, M.; Alves, M.S.; de Souza Inhaquite, P.; Santos, R.R.; Girao, V.B.C.; da Cunha, A.; et al. Diversity of clonal types of Klebsiella pneumoniae causing infections in intensive care neonatal patients in a large urban setting. Braz. J. Microbiol. 2019. [Google Scholar] [CrossRef] [PubMed]
  76. Vergadi, E.; Bitsori, M.; Maraki, S.; Galanakis, E. Community-onset carbapenem-resistant Klebsiella pneumoniae urinary tract infections in infancy following NICU hospitalisation. J. Pediatr. Urol. 2017, 13, 495.e491–495.e496. [Google Scholar] [CrossRef] [PubMed]
  77. Ferreira, R.L.; da Silva, B.C.M.; Rezende, G.S.; Nakamura-Silva, R.; Pitondo-Silva, A.; Campanini, E.B.; Brito, M.C.A.; da Silva, E.M.L.; Freire, C.C.M.; da Cunha, A.F.; et al. High Prevalence of Multidrug-Resistant Klebsiella pneumoniae Harboring Several Virulence and beta-Lactamase Encoding Genes in a Brazilian Intensive Care Unit. Front. Microbiol. 2018, 9, 3198. [Google Scholar] [CrossRef] [PubMed]
  78. Logan, L.K.; Weinstein, R.A. The Epidemiology of Carbapenem-Resistant Enterobacteriaceae: The Impact and Evolution of a Global Menace. J. Infect. Dis. 2017, 215, S28–S36. [Google Scholar] [CrossRef] [Green Version]
  79. Shankar, C.; Nabarro, L.E.; Anandan, S.; Ravi, R.; Babu, P.; Munusamy, E.; Jeyaseelan, V.; Rupali, P.; Verghese, V.P.; Veeraraghavan, B. Extremely High Mortality Rates in Patients with Carbapenem-resistant, Hypermucoviscous Klebsiella pneumoniae Blood Stream Infections. J. Assoc. Physicians India 2018, 66, 13–16. [Google Scholar] [PubMed]
  80. Aygun, F.; Aygun, F.D.; Varol, F.; Durak, C.; Cokugras, H.; Camcioglu, Y.; Cam, H. Infections with Carbapenem-Resistant Gram-Negative Bacteria are a Serious Problem Among Critically Ill Children: A Single-Centre Retrospective Study. Pathogens 2019, 8, 69. [Google Scholar] [CrossRef]
  81. Brinkac, L.M.; White, R.; D’Souza, R.; Nguyen, K.; Obaro, S.K.; Fouts, D.E. Emergence of New Delhi Metallo-beta-Lactamase (NDM-5) in Klebsiella quasipneumoniae from Neonates in a Nigerian Hospital. mSphere 2019, 4. [Google Scholar] [CrossRef]
  82. Karampatakis, T.; Tsergouli, K.; Politi, L.; Diamantopoulou, G.; Iosifidis, E.; Antachopoulos, C.; Karyoti, A.; Mouloudi, E.; Tsakris, A.; Roilides, E. Molecular Epidemiology of Endemic Carbapenem-Resistant Gram-Negative Bacteria in an Intensive Care Unit. Microb. Drug Resist. 2019, 25, 712–716. [Google Scholar] [CrossRef] [PubMed]
  83. Sirijatuphat, R.; Sripanidkulchai, K.; Boonyasiri, A.; Rattanaumpawan, P.; Supapueng, O.; Kiratisin, P.; Thamlikitkul, V. Implementation of global antimicrobial resistance surveillance system (GLASS) in patients with bacteremia. PLoS ONE 2018, 13, e0190132. [Google Scholar] [CrossRef] [PubMed]
  84. Deshmukh, P.; Unni, S.; Krishnappa, G.; Padmanabhan, B. The Keap1-Nrf2 pathway: Promising therapeutic target to counteract ROS-mediated damage in cancers and neurodegenerative diseases. Biophys. Rev. 2017, 9, 41–56. [Google Scholar] [CrossRef] [PubMed]
  85. Jaramillo, M.C.; Zhang, D.D. The emerging role of the Nrf2-Keap1 signaling pathway in cancer. Genes Dev. 2013, 27, 2179–2191. [Google Scholar] [CrossRef] [PubMed]
  86. Kansanen, E.; Kuosmanen, S.M.; Leinonen, H.; Levonen, A.L. The Keap1-Nrf2 pathway: Mechanisms of activation and dysregulation in cancer. Redox Biol. 2013, 1, 45–49. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  87. Fimognari, C.; Nusse, M.; Cesari, R.; Iori, R.; Cantelli-Forti, G.; Hrelia, P. Growth inhibition, cell-cycle arrest and apoptosis in human T-cell leukemia by the isothiocyanate sulforaphane. Carcinogenesis 2002, 23, 581–586. [Google Scholar] [CrossRef] [PubMed]
  88. Liang, H.; Lai, B.; Yuan, Q. Sulforaphane induces cell-cycle arrest and apoptosis in cultured human lung adenocarcinoma LTEP-A2 cells and retards growth of LTEP-A2 xenografts in vivo. J. Nat. Prod. 2008, 71, 1911–1914. [Google Scholar] [CrossRef] [PubMed]
  89. Pham, N.A.; Jacobberger, J.W.; Schimmer, A.D.; Cao, P.; Gronda, M.; Hedley, D.W. The dietary isothiocyanate sulforaphane targets pathways of apoptosis, cell cycle arrest, and oxidative stress in human pancreatic cancer cells and inhibits tumor growth in severe combined immunodeficient mice. Mol. Cancer Ther. 2004, 3, 1239–1248. [Google Scholar] [PubMed]
  90. Tang, L.; Zhang, Y.; Jobson, H.E.; Li, J.; Stephenson, K.K.; Wade, K.L.; Fahey, J.W. Potent activation of mitochondria-mediated apoptosis and arrest in S and M phases of cancer cells by a broccoli sprout extract. Mol. Cancer Ther. 2006, 5, 935–944. [Google Scholar] [CrossRef] [PubMed]
  91. Singh, A.V.; Xiao, D.; Lew, K.L.; Dhir, R.; Singh, S.V. Sulforaphane induces caspase-mediated apoptosis in cultured PC-3 human prostate cancer cells and retards growth of PC-3 xenografts in vivo. Carcinogenesis 2004, 25, 83–90. [Google Scholar] [CrossRef] [PubMed]
  92. Azarenko, O.; Okouneva, T.; Singletary, K.W.; Jordan, M.A.; Wilson, L. Suppression of microtubule dynamic instability and turnover in MCF7 breast cancer cells by sulforaphane. Carcinogenesis 2008, 29, 2360–2368. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  93. Jackson, S.J.; Singletary, K.W. Sulforaphane: A naturally occurring mammary carcinoma mitotic inhibitor, which disrupts tubulin polymerization. Carcinogenesis 2004, 25, 219–227. [Google Scholar] [CrossRef] [PubMed]
  94. Dashwood, R.H.; Ho, E. Dietary agents as histone deacetylase inhibitors: Sulforaphane and structurally related isothiocyanates. Nutr. Rev. 2008, 66 (Suppl. 1), S36–S38. [Google Scholar] [CrossRef]
  95. Gibbs, A.; Schwartzman, J.; Deng, V.; Alumkal, J. Sulforaphane destabilizes the androgen receptor in prostate cancer cells by inactivating histone deacetylase 6. Proc. Natl. Acad. Sci. USA 2009, 106, 16663–16668. [Google Scholar] [CrossRef] [Green Version]
  96. Myzak, M.C.; Hardin, K.; Wang, R.; Dashwood, R.H.; Ho, E. Sulforaphane inhibits histone deacetylase activity in BPH-1, LnCaP and PC-3 prostate epithelial cells. Carcinogenesis 2006, 27, 811–819. [Google Scholar] [CrossRef]
  97. Myzak, M.C.; Karplus, P.A.; Chung, F.L.; Dashwood, R.H. A novel mechanism of chemoprotection by sulforaphane: Inhibition of histone deacetylase. Cancer Res. 2004, 64, 5767–5774. [Google Scholar] [CrossRef]
  98. Desai, J.V.; Mitchell, A.P.; Andes, D.R. Fungal biofilms, drug resistance, and recurrent infection. Cold Spring Harb. Perspect. Med. 2014, 4, a019729. [Google Scholar] [CrossRef]
  99. Deva, A.K.; Adams, W.P., Jr.; Vickery, K. The role of bacterial biofilms in device-associated infection. Plast Reconstr. Surg. 2013, 132, 1319–1328. [Google Scholar] [CrossRef] [PubMed]
  100. Hall, M.R.; McGillicuddy, E.; Kaplan, L.J. Biofilm: Basic principles, pathophysiology, and implications for clinicians. Surg. Infect. 2014, 15, 1–7. [Google Scholar] [CrossRef] [PubMed]
  101. Holban, A.M.; Gestal, M.C.; Grumezescu, A.M. New molecular strategies for reducing implantable medical devices associated infections. Curr. Med. Chem. 2014, 21, 3375–3382. [Google Scholar] [CrossRef] [PubMed]
  102. Nicolle, L.E. Urinary catheter-associated infections. Infect. Dis. Clin. North Am. 2012, 26, 13–27. [Google Scholar] [CrossRef] [PubMed]
  103. Chen, M.; Yu, Q.; Sun, H. Novel strategies for the prevention and treatment of biofilm related infections. Int. J. Mol. Sci. 2013, 14, 18488–18501. [Google Scholar] [CrossRef] [PubMed]
  104. Otto, M. Staphylococcal biofilms. Curr. Top. Microbiol. Immunol. 2008, 322, 207–228. [Google Scholar] [PubMed]
  105. Otto, M. Staphylococcal infections: Mechanisms of biofilm maturation and detachment as critical determinants of pathogenicity. Annu. Rev. Med. 2013, 64, 175–188. [Google Scholar] [CrossRef] [PubMed]
  106. Otto, M. Staphylococcus epidermidis pathogenesis. Methods Mol. Biol. 2014, 1106, 17–31. [Google Scholar]
  107. Agarwal, A.; Singh, K.P.; Jain, A. Medical significance and management of staphylococcal biofilm. FEMS Immunol. Med. Microbiol. 2010, 58, 147–160. [Google Scholar] [CrossRef]
  108. Kleinschmidt, S.; Huygens, F.; Faoagali, J.; Rathnayake, I.U.; Hafner, L.M. Staphylococcus epidermidis as a cause of bacteremia. Future Microbiol. 2015, 10, 1859–1879. [Google Scholar] [CrossRef]
  109. Shah, S.; Tatara, A.M.; D’Souza, R.N.; Mikos, A.G.; Kurtis, F.K. Evolving strategies for preventing biofilm on implantable materials. Mater. Today 2013, 16, 177–182. [Google Scholar] [CrossRef]
  110. Sulemankhil, I.; Ganopolsky, J.G.; Dieni, C.A.; Dan, A.F.; Jones, M.L.; Prakash, S. Prevention and treatment of virulent bacterial biofilms with an enzymatic nitric oxide-releasing dressing. Antimicrob. Agents Chemother. 2012, 56, 6095–6103. [Google Scholar] [CrossRef] [PubMed]
  111. Kaiser, S.J.; Mutters, N.T.; Blessing, B.; Gunther, F. Natural isothiocyanates express antimicrobial activity against developing and mature biofilms of Pseudomonas aeruginosa. Fitoterapia 2017, 119, 57–63. [Google Scholar] [CrossRef] [PubMed]
  112. Lara-Lledo, M.; Olaimat, A.; Holley, R.A. Inhibition of Listeria monocytogenes on bologna sausages by an antimicrobial film containing mustard extract or sinigrin. Int. J. Food Microbiol. 2012, 156, 25–31. [Google Scholar] [CrossRef] [PubMed]
  113. Mazumder, A.; Dwivedi, A.; du Plessis, J. Sinigrin and Its Therapeutic Benefits. Molecules 2016, 21, 416. [Google Scholar] [CrossRef] [PubMed]
  114. Feskanich, D.; Ziegler, R.G.; Michaud, D.S.; Giovannucci, E.L.; Speizer, F.E.; Willett, W.C.; Colditz, G.A. Prospective study of fruit and vegetable consumption and risk of lung cancer among men and women. J. Natl. Cancer Inst. 2000, 92, 1812–1823. [Google Scholar] [CrossRef] [PubMed]
  115. Joseph, M.A.; Moysich, K.B.; Freudenheim, J.L.; Shields, P.G.; Bowman, E.D.; Zhang, Y.; Marshall, J.R.; Ambrosone, C.B. Cruciferous vegetables, genetic polymorphisms in glutathione S-transferases M1 and T1, and prostate cancer risk. Nutr. Cancer 2004, 50, 206–213. [Google Scholar] [CrossRef]
  116. Neuhouser, M.L.; Patterson, R.E.; Thornquist, M.D.; Omenn, G.S.; King, I.B.; Goodman, G.E. Fruits and vegetables are associated with lower lung cancer risk only in the placebo arm of the beta-carotene and retinol efficacy trial (CARET). Cancer Epidemiol. Biomarkers Prev. 2003, 12, 350–358. [Google Scholar]
  117. Verhoeven, D.T.; Goldbohm, R.A.; van Poppel, G.; Verhagen, H.; van den Brandt, P.A. Epidemiological studies on brassica vegetables and cancer risk. Cancer Epidemiol. Biomarkers Prev. 1996, 5, 733–748. [Google Scholar]
  118. Voorrips, L.E.; Goldbohm, R.A.; van Poppel, G.; Sturmans, F.; Hermus, R.J.; van den Brandt, P.A. Vegetable and fruit consumption and risks of colon and rectal cancer in a prospective cohort study: The Netherlands Cohort Study on Diet and Cancer. Am. J. Epidemiol. 2000, 152, 1081–1092. [Google Scholar] [CrossRef]
  119. Smith, G.A.; Lin, T.H.; Sheehan, A.E.; Van der Goes van Naters, W.; Neukomm, L.J.; Graves, H.K.; Bis-Brewer, D.M.; Zuchner, S.; Freeman, M.R. Glutathione S-Transferase Regulates Mitochondrial Populations in Axons through Increased Glutathione Oxidation. Neuron 2019. [Google Scholar] [CrossRef] [PubMed]
  120. Agbas, A.; Krishnamurthy, P.; Michaelis, M.L.; Michaelis, E.K. Mitochondrial Electron Transfer Cascade Enzyme Activity Assessment in Cultured Neurons and Select Brain Regions. Curr. Protoc. Toxicol. 2019. [Google Scholar] [CrossRef] [PubMed]
  121. Young, A.; Gill, R.; Mailloux, R.J. Protein S-glutathionylation: The linchpin for the transmission of regulatory information on redox buffering capacity in mitochondria. Chem. Biol. Interact. 2019, 299, 151–162. [Google Scholar] [CrossRef]
  122. Krug, P.; Mielczarek, L.; Wiktorska, K.; Kaczynska, K.; Wojciechowski, P.; Andrzejewski, K.; Ofiara, K.; Szterk, A.; Mazur, M. Sulforaphane-conjugated selenium nanoparticles: Towards a synergistic anticancer effect. Nanotechnology 2019, 30, 065101. [Google Scholar] [CrossRef] [PubMed]
  123. Kan, S.F.; Wang, J.; Sun, G.X. Sulforaphane regulates apoptosis- and proliferationrelated signaling pathways and synergizes with cisplatin to suppress human ovarian cancer. Int. J. Mol. Med. 2018, 42, 2447–2458. [Google Scholar] [PubMed]
  124. Rakariyatham, K.; Wu, X.; Tang, Z.; Han, Y.; Wang, Q.; Xiao, H. Synergism between luteolin and sulforaphane in anti-inflammation. Food Funct. 2018, 9, 5115–5123. [Google Scholar] [CrossRef] [PubMed]
  125. Lubecka, K.; Kaufman-Szymczyk, A.; Fabianowska-Majewska, K. Inhibition of breast cancer cell growth by the combination of clofarabine and sulforaphane involves epigenetically mediated CDKN2A upregulation. Nucleosides Nucleotides Nucleic Acids 2018, 37, 280–289. [Google Scholar] [CrossRef]
  126. Bose, C.; Awasthi, S.; Sharma, R.; Benes, H.; Hauer-Jensen, M.; Boerma, M.; Singh, S.P. Sulforaphane potentiates anticancer effects of doxorubicin and attenuates its cardiotoxicity in a breast cancer model. PLoS ONE 2018, 13, e0193918. [Google Scholar] [CrossRef]
  127. Chirumbolo, S.; Bjorklund, G. Sulforaphane and 5-fluorouracil synergistically inducing autophagy in breast cancer: A possible role for the Nrf2-Keap1-ARE signaling? Food Chem. Toxicol. 2018, 112, 414–415. [Google Scholar] [CrossRef]
  128. Schillheim, B.; Jansen, I.; Baum, S.; Beesley, A.; Bolm, C.; Conrath, U. Sulforaphane Modifies Histone H3, Unpacks Chromatin, and Primes Defense. Plant Physiol. 2018, 176, 2395–2405. [Google Scholar] [CrossRef]
  129. Royston, K.J.; Udayakumar, N.; Lewis, K.; Tollefsbol, T.O. A Novel Combination of Withaferin A and Sulforaphane Inhibits Epigenetic Machinery, Cellular Viability and Induces Apoptosis of Breast Cancer Cells. Int. J. Mol. Sci. 2017, 18, 1092. [Google Scholar] [CrossRef] [PubMed]
  130. Chaiprasongsuk, A.; Lohakul, J.; Soontrapa, K.; Sampattavanich, S.; Akarasereenont, P.; Panich, U. Activation of Nrf2 Reduces UVA-Mediated MMP-1 Upregulation via MAPK/AP-1 Signaling Cascades: The Photoprotective Effects of Sulforaphane and Hispidulin. J. Pharmacol. Exp. Ther. 2017, 360, 388–398. [Google Scholar] [CrossRef] [PubMed]
  131. Chatterjee, S.; Rhee, Y.H.; Ahn, J.C. Sulforaphene-Carboplatin Combination Synergistically Enhances Apoptosis by Disruption of Mitochondrial Membrane Potential and Cell Cycle Arrest in Human Non-Small Cell Lung Carcinoma. J. Med. Food 2016, 19, 860–869. [Google Scholar] [CrossRef] [PubMed]
  132. Huang, J.; Tao, C.; Yu, Y.; Yu, F.; Zhang, H.; Gao, J.; Wang, D.; Chen, Y.; Zhang, G.; Zhou, G.; et al. Simultaneous Targeting of Differentiated Breast Cancer Cells and Breast Cancer Stem Cells by Combination of Docetaxel- and Sulforaphane-Loaded Self-Assembled Poly(D, L-lactide-co-glycolide)/Hyaluronic Acid Block Copolymer-Based Nanoparticles. J. Biomed. Nanotechnol. 2016, 12, 1463–1477. [Google Scholar] [CrossRef] [PubMed]
  133. Kaczynska, A.; Herman-Antosiewicz, A. Combination of lapatinib with isothiocyanates overcomes drug resistance and inhibits migration of HER2 positive breast cancer cells. Breast Cancer 2017, 24, 271–280. [Google Scholar] [CrossRef] [PubMed]
  134. Erzinger, M.M.; Bovet, C.; Hecht, K.M.; Senger, S.; Winiker, P.; Sobotzki, N.; Cristea, S.; Beerenwinkel, N.; Shay, J.W.; Marra, G.; et al. Sulforaphane Preconditioning Sensitizes Human Colon Cancer Cells towards the Bioreductive Anticancer Prodrug PR-104A. PLoS ONE 2016, 11, e0150219. [Google Scholar] [CrossRef] [PubMed]
  135. Gao, J.; Xiong, B.; Zhang, B.; Li, S.; Huang, N.; Zhan, G.; Jiang, R.; Yang, L.; Wu, Y.; Miao, L.; et al. Sulforaphane Alleviates Lipopolysaccharide-induced Spatial Learning and Memory Dysfunction in Mice: The Role of BDNF-mTOR Signaling Pathway. Neuroscience 2018, 388, 357–366. [Google Scholar] [CrossRef] [PubMed]
  136. Lucarini, E.; Micheli, L.; Trallori, E.; Citi, V.; Martelli, A.; Testai, L.; De Nicola, G.R.; Iori, R.; Calderone, V.; Ghelardini, C.; et al. Effect of glucoraphanin and sulforaphane against chemotherapy-induced neuropathic pain: Kv7 potassium channels modulation by H2 S release in vivo. Phytother. Res. 2018, 32, 2226–2234. [Google Scholar] [CrossRef] [PubMed]
  137. Sohel, M.M.H.; Amin, A.; Prastowo, S.; Linares-Otoya, L.; Hoelker, M.; Schellander, K.; Tesfaye, D. Correction to: Sulforaphane protects granulosa cells against oxidative stress via activation of NRF2-ARE pathway. Cell Tissue Res. 2018, 374, 679–685. [Google Scholar] [CrossRef] [Green Version]
  138. Wang, Y.; Mandal, A.K.; Son, Y.O.; Pratheeshkumar, P.; Wise, J.T.F.; Wang, L.; Zhang, Z.; Shi, X.; Chen, Z. Roles of ROS, Nrf2, and autophagy in cadmium-carcinogenesis and its prevention by sulforaphane. Toxicol. Appl. Pharmacol. 2018, 353, 23–30. [Google Scholar] [CrossRef]
  139. Corssac, G.B.; Campos-Carraro, C.; Hickmann, A.; da Rosa Araujo, A.S.; Fernandes, R.O.; Bello-Klein, A. Sulforaphane effects on oxidative stress parameters in culture of adult cardiomyocytes. Biomed. Pharmacother. 2018, 104, 165–171. [Google Scholar] [CrossRef] [PubMed]
  140. Jaafaru, M.S.; Abd Karim, N.A.; Enas, M.E.; Rollin, P.; Mazzon, E.; Abdull Razis, A.F. Protective Effect of Glucosinolates Hydrolytic Products in Neurodegenerative Diseases (NDDs). Nutrients 2018, 10, 580. [Google Scholar] [CrossRef] [PubMed]
  141. Lee, S.; Choi, B.R.; Kim, J.; LaFerla, F.M.; Park, J.H.Y.; Han, J.S.; Lee, K.W. Sulforaphane Upregulates the Heat Shock Protein Co-Chaperone CHIP and Clears Amyloid-beta and Tau in a Mouse Model of Alzheimer’s Disease. Mol. Nutr. Food Res. 2018, 62, e1800240. [Google Scholar] [CrossRef] [PubMed]
  142. Moustafa, P.E.; Abdelkader, N.F.; El Awdan, S.A.; El-Shabrawy, O.A.; Zaki, H.F. Extracellular Matrix Remodeling and Modulation of Inflammation and Oxidative Stress by Sulforaphane in Experimental Diabetic Peripheral Neuropathy. Inflammation 2018, 41, 1460–1476. [Google Scholar] [CrossRef] [PubMed]
  143. Silva Rodrigues, J.F.; Silva, E.S.C.; Franca Muniz, T.; de Aquino, A.F.; Neuza da Silva Nina, L.; Fialho Sousa, N.C.; Nascimento da Silva, L.C.; de Souza, B.; da Penha, T.A.; Abreu-Silva, A.L.; et al. Sulforaphane Modulates Joint Inflammation in a Murine Model of Complete Freund’s Adjuvant-Induced Mono-Arthritis. Molecules 2018, 23, 988. [Google Scholar] [CrossRef] [PubMed]
  144. Pu, D.; Zhao, Y.; Chen, J.; Sun, Y.; Lv, A.; Zhu, S.; Luo, C.; Zhao, K.; Xiao, Q. Protective Effects of Sulforaphane on Cognitive Impairments and AD-like Lesions in Diabetic Mice are Associated with the Upregulation of Nrf2 Transcription Activity. Neuroscience 2018, 381, 35–45. [Google Scholar] [CrossRef] [PubMed]
  145. Jhang, K.A.; Park, J.S.; Kim, H.S.; Chong, Y.H. Sulforaphane rescues amyloid-beta peptide-mediated decrease in MerTK expression through its anti-inflammatory effect in human THP-1 macrophages. J. Neuroinflammation 2018, 15, 75. [Google Scholar] [CrossRef] [PubMed]
  146. Ma, T.; Zhu, D.; Chen, D.; Zhang, Q.; Dong, H.; Wu, W.; Lu, H.; Wu, G. Sulforaphane, a Natural Isothiocyanate Compound, Improves Cardiac Function and Remodeling by Inhibiting Oxidative Stress and Inflammation in a Rabbit Model of Chronic Heart Failure. Med. Sci. Monit. 2018, 24, 1473–1483. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  147. Choi, S.Y.; Kee, H.J.; Jin, L.; Ryu, Y.; Sun, S.; Kim, G.R.; Jeong, M.H. Inhibition of class IIa histone deacetylase activity by gallic acid, sulforaphane, TMP269, and panobinostat. Biomed. Pharmacother. 2018, 101, 145–154. [Google Scholar] [CrossRef] [PubMed]
  148. Zhou, Y.; Yang, G.; Tian, H.; Hu, Y.; Wu, S.; Geng, Y.; Lin, K.; Wu, W. Sulforaphane metabolites cause apoptosis via microtubule disruption in cancer. Endocr. Relat. Cancer 2018, 25, 255–268. [Google Scholar] [CrossRef] [PubMed]
  149. Eren, E.; Tufekci, K.U.; Isci, K.B.; Tastan, B.; Genc, K.; Genc, S. Sulforaphane Inhibits Lipopolysaccharide-Induced Inflammation, Cytotoxicity, Oxidative Stress, and miR-155 Expression and Switches to Mox Phenotype through Activating Extracellular Signal-Regulated Kinase 1/2-Nuclear Factor Erythroid 2-Related Factor 2/Antioxidant Response Element Pathway in Murine Microglial Cells. Front. Immunol. 2018, 9, 36. [Google Scholar] [PubMed]
  150. Qin, S.; Yang, C.; Huang, W.; Du, S.; Mai, H.; Xiao, J.; Lu, T. Sulforaphane attenuates microglia-mediated neuronal necroptosis through down-regulation of MAPK/NF-kappaB signaling pathways in LPS-activated BV-2 microglia. Pharmacol. Res. 2018, 133, 218–235. [Google Scholar] [CrossRef] [PubMed]
  151. Dulull, N.K.; Dias, D.A.; Thrimawithana, T.R.; Kwa, F.A.A. L-Sulforaphane Confers Protection Against Oxidative Stress in an In Vitro Model of Age-Related Macular Degeneration. Curr Mol Pharmacol 2018, 11, 237–253. [Google Scholar] [CrossRef] [PubMed]
  152. Dokumacioglu, E.; Iskender, H.; Aktas, M.S.; Hanedan, B.; Dokumacioglu, A.; Sen, T.M.; Musmul, A. The effect of sulforaphane on oxidative stress and inflammation in rats with toxic hepatitis induced by acetaminophene. Bratisl Lek Listy 2017, 118, 453–459. [Google Scholar] [CrossRef] [PubMed]
  153. Sun, B.; Zhang, X.; Yin, Y.; Sun, H.; Ge, H.; Li, W. Effects of sulforaphane and vitamin E on cognitive disorder and oxidative damage in lead-exposed mice hippocampus at lactation. J. Trace Elem. Med. Biol. 2017, 44, 88–92. [Google Scholar] [CrossRef]
  154. Son, Y.H.; Jang, E.J.; Kim, Y.W.; Lee, J.H. Sulforaphane prevents dexamethasone-induced muscle atrophy via regulation of the Akt/Foxo1 axis in C2C12 myotubes. Biomed. Pharmacother. 2017, 95, 1486–1492. [Google Scholar] [CrossRef] [PubMed]
  155. Lan, H.; Yuan, H.; Lin, C. Sulforaphane induces p53deficient SW480 cell apoptosis via the ROSMAPK signaling pathway. Mol. Med. Rep. 2017, 16, 7796–7804. [Google Scholar] [CrossRef]
  156. Dacosta, C.; Bao, Y. The Role of MicroRNAs in the Chemopreventive Activity of Sulforaphane from Cruciferous Vegetables. Nutrients 2017, 9, 902. [Google Scholar] [CrossRef]
  157. Psurski, M.; Janczewski, L.; Switalska, M.; Gajda, A.; Goszczynski, T.M.; Oleksyszyn, J.; Wietrzyk, J.; Gajda, T. Novel phosphonate analogs of sulforaphane: Synthesis, in vitro and in vivo anticancer activity. Eur. J. Med. Chem. 2017, 132, 63–80. [Google Scholar] [CrossRef]
  158. Yanaka, A. Role of Sulforaphane in Protection of Gastrointestinal Tract Against H. pylori and NSAID-Induced Oxidative Stress. Curr. Pharm. Des. 2017, 23, 4066–4075. [Google Scholar] [CrossRef] [Green Version]
  159. Nazmy, E.A.; El-Khouly, O.A.; Atef, H.; Said, E. Sulforaphane protects against sodium valproate-induced acute liver injury. Can. J. Physiol. Pharmacol. 2017, 95, 420–426. [Google Scholar] [CrossRef] [PubMed]
  160. Bi, M.; Li, Q.; Guo, D.; Ding, X.; Bi, W.; Zhang, Y.; Zou, Y. Sulphoraphane Improves Neuronal Mitochondrial Function in Brain Tissue in Acute Carbon Monoxide Poisoning Rats. Basic Clin. Pharmacol. Toxicol. 2017, 120, 541–549. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  161. Lan, A.; Li, W.; Liu, Y.; Xiong, Z.; Zhang, X.; Zhou, S.; Palko, O.; Chen, H.; Kapita, M.; Prigge, J.R.; et al. Chemoprevention of oxidative stress-associated oral carcinogenesis by sulforaphane depends on NRF2 and the isothiocyanate moiety. Oncotarget 2016, 7, 53502–53514. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  162. Liu, C.; Xu, H.; Fu, S.; Chen, Y.; Chen, Q.; Cai, Z.; Zhou, J.; Wang, Z. Sulforaphane Ameliorates Bladder Dysfunction through Activation of the Nrf2-ARE Pathway in a Rat Model of Partial Bladder Outlet Obstruction. Oxid. Med. Cell Longev. 2016, 2016, 7598294. [Google Scholar] [CrossRef] [PubMed]
  163. Miao, X.; Bai, Y.; Sun, W.; Cui, W.; Xin, Y.; Wang, Y.; Tan, Y.; Miao, L.; Fu, Y.; Su, G.; et al. Sulforaphane prevention of diabetes-induced aortic damage was associated with the up-regulation of Nrf2 and its down-stream antioxidants. Nutr. Metab. 2012, 9, 84. [Google Scholar] [CrossRef]
  164. An, Y.W.; Jhang, K.A.; Woo, S.Y.; Kang, J.L.; Chong, Y.H. Sulforaphane exerts its anti-inflammatory effect against amyloid-beta peptide via STAT-1 dephosphorylation and activation of Nrf2/HO-1 cascade in human THP-1 macrophages. Neurobiol. Aging. 2016, 38, 1–10. [Google Scholar] [CrossRef]
  165. Koolivand, M.; Ansari, M.; Piroozian, F.; Moein, S.; MalekZadeh, K. Alleviating the progression of acute myeloid leukemia (AML) by sulforaphane through controlling miR-155 levels. Mol. Biol. Rep. 2018, 45, 2491–2499. [Google Scholar] [CrossRef]
  166. Misiewicz, I.; Skupinska, K.; Kasprzycka-Guttman, T. Sulforaphane and 2-oxohexyl isothiocyanate induce cell growth arrest and apoptosis in L-1210 leukemia and ME-18 melanoma cells. Oncol. Rep. 2003, 10, 2045–2050. [Google Scholar] [CrossRef]
  167. Prata, C.; Facchini, C.; Leoncini, E.; Lenzi, M.; Maraldi, T.; Angeloni, C.; Zambonin, L.; Hrelia, S.; Fiorentini, D. Sulforaphane Modulates AQP8-Linked Redox Signalling in Leukemia Cells. Oxid. Med. Cell Longev. 2018, 2018, 4125297. [Google Scholar] [CrossRef]
  168. Shang, H.S.; Shih, Y.L.; Lee, C.H.; Hsueh, S.C.; Liu, J.Y.; Liao, N.C.; Chen, Y.L.; Huang, Y.P.; Lu, H.F.; Chung, J.G. Sulforaphane-induced apoptosis in human leukemia HL-60 cells through extrinsic and intrinsic signal pathways and altering associated genes expression assayed by cDNA microarray. Environ. Toxicol. 2017, 32, 311–328. [Google Scholar] [CrossRef]
  169. Shih, Y.L.; Wu, L.Y.; Lee, C.H.; Chen, Y.L.; Hsueh, S.C.; Lu, H.F.; Liao, N.C.; Chung, J.G. Sulforaphane promotes immune responses in a WEHI3induced leukemia mouse model through enhanced phagocytosis of macrophages and natural killer cell activities in vivo. Mol. Med. Rep. 2016, 13, 4023–4029. [Google Scholar] [CrossRef] [PubMed]
  170. Xue, X.; Chen, F.; Liu, A.; Sun, D.; Wu, J.; Kong, F.; Luan, Y.; Qu, X.; Wang, R. Reversal of the multidrug resistance of human ileocecal adenocarcinoma cells by acetyl-11-keto-beta-boswellic acid via downregulation of P-glycoprotein signals. Biosci. Trends 2016, 10, 392–399. [Google Scholar] [CrossRef] [PubMed]
  171. Dogan Sigva, Z.O.; Balci Okcanoglu, T.; Biray Avci, C.; Yilmaz Susluer, S.; Kayabasi, C.; Turna, B.; Dodurga, Y.; Nazli, O.; Gunduz, C. Investigation of the synergistic effects of paclitaxel and herbal substances and endemic plant extracts on cell cycle and apoptosis signal pathways in prostate cancer cell lines. Gene 2019, 687, 261–271. [Google Scholar] [CrossRef] [PubMed]
  172. Singh, K.B.; Kim, S.H.; Hahm, E.R.; Pore, S.K.; Jacobs, B.L.; Singh, S.V. Prostate cancer chemoprevention by sulforaphane in a preclinical mouse model is associated with inhibition of fatty acid metabolism. Carcinogenesis 2018, 39, 826–837. [Google Scholar] [CrossRef] [PubMed]
  173. Vyas, A.R.; Moura, M.B.; Hahm, E.R.; Singh, K.B.; Singh, S.V. Sulforaphane Inhibits c-Myc-Mediated Prostate Cancer Stem-Like Traits. J. Cell Biochem. 2016, 117, 2482–2495. [Google Scholar] [CrossRef] [Green Version]
  174. Tsai, J.Y.; Tsai, S.H.; Wu, C.C. The chemopreventive isothiocyanate sulforaphane reduces anoikis resistance and anchorage-independent growth in non-small cell human lung cancer cells. Toxicol. Appl. Pharmacol. 2019, 362, 116–124. [Google Scholar] [CrossRef]
  175. Zuryn, A.; Litwiniec, A.; Safiejko-Mroczka, B.; Klimaszewska-Wisniewska, A.; Gagat, M.; Krajewski, A.; Gackowska, L.; Grzanka, D. The effect of sulforaphane on the cell cycle, apoptosis and expression of cyclin D1 and p21 in the A549 non-small cell lung cancer cell line. Int. J. Oncol. 2016, 48, 2521–2533. [Google Scholar] [CrossRef]
  176. Carrasco-Pozo, C.; Tan, K.N.; Gotteland, M.; Borges, K. Sulforaphane Protects against High Cholesterol-Induced Mitochondrial Bioenergetics Impairments, Inflammation, and Oxidative Stress and Preserves Pancreatic beta-Cells Function. Oxid. Med. Cell Longev. 2017, 2017, 3839756. [Google Scholar] [CrossRef]
  177. Chen, X.; Jiang, Z.; Zhou, C.; Chen, K.; Li, X.; Wang, Z.; Wu, Z.; Ma, J.; Ma, Q.; Duan, W. Activation of Nrf2 by Sulforaphane Inhibits High Glucose-Induced Progression of Pancreatic Cancer via AMPK Dependent Signaling. Cell Physiol. Biochem. 2018, 50, 1201–1215. [Google Scholar] [CrossRef]
  178. Dong, Z.; Shang, H.; Chen, Y.Q.; Pan, L.L.; Bhatia, M.; Sun, J. Sulforaphane Protects Pancreatic Acinar Cell Injury by Modulating Nrf2-Mediated Oxidative Stress and NLRP3 Inflammatory Pathway. Oxid. Med. Cell Longev. 2016, 2016, 7864150. [Google Scholar] [CrossRef]
  179. Park, Y.K.; Ramalingam, M.; Kim, S.; Jang, B.C.; Park, J.W. Sulforaphane inhibits the interferon-gamma-induced expression of MIG, IP-10 and I-TAC in INS1 pancreatic beta-cells through the downregulation of IRF-1, STAT-1 and PKB. Int. J. Mol. Med. 2017, 40, 907–912. [Google Scholar] [CrossRef] [PubMed]
  180. Cao, C.; Wu, H.; Vasilatos, S.N.; Chandran, U.; Qin, Y.; Wan, Y.; Oesterreich, S.; Davidson, N.E.; Huang, Y. HDAC5-LSD1 axis regulates antineoplastic effect of natural HDAC inhibitor sulforaphane in human breast cancer cells. Int. J. Cancer 2018, 143, 1388–1401. [Google Scholar] [CrossRef] [PubMed]
  181. Danafar, H.; Sharafi, A.; Kheiri Manjili, H.; Andalib, S. Sulforaphane delivery using mPEG-PCL co-polymer nanoparticles to breast cancer cells. Pharm. Dev. Technol. 2017, 22, 642–651. [Google Scholar] [CrossRef] [PubMed]
  182. Gianfredi, V.; Nucci, D.; Vannini, S.; Villarini, M.; Moretti, M. In vitro Biological Effects of Sulforaphane (SFN), Epigallocatechin-3-gallate (EGCG), and Curcumin on Breast Cancer Cells: A Systematic Review of the Literature. Nutr. Cancer 2017, 69, 969–978. [Google Scholar] [CrossRef] [PubMed]
  183. Gianfredi, V.; Vannini, S.; Moretti, M.; Villarini, M.; Bragazzi, N.L.; Izzotti, A.; Nucci, D. Sulforaphane and Epigallocatechin Gallate Restore Estrogen Receptor Expression by Modulating Epigenetic Events in the Breast Cancer Cell Line MDA-MB-231: A Systematic Review and Meta-Analysis. J. Nutrigenet. Nutrigenomics 2017, 10, 126–135. [Google Scholar] [CrossRef] [PubMed]
  184. Jaman, M.S.; Sayeed, M.A. Ellagic acid, sulforaphane, and ursolic acid in the prevention and therapy of breast cancer: Current evidence and future perspectives. Breast Cancer 2018, 25, 517–528. [Google Scholar] [CrossRef] [PubMed]
  185. Kamal, M.M.; Nazzal, S. Novel sulforaphane-enabled self-microemulsifying delivery systems (SFN-SMEDDS) of taxanes: Formulation development and in vitro cytotoxicity against breast cancer cells. Int. J. Pharm. 2018, 536, 187–198. [Google Scholar] [CrossRef] [PubMed]
  186. Lewinska, A.; Adamczyk-Grochala, J.; Deregowska, A.; Wnuk, M. Sulforaphane-Induced Cell Cycle Arrest and Senescence are accompanied by DNA Hypomethylation and Changes in microRNA Profile in Breast Cancer Cells. Theranostics 2017, 7, 3461–3477. [Google Scholar] [CrossRef]
  187. Yang, F.; Wang, F.; Liu, Y.; Wang, S.; Li, X.; Huang, Y.; Xia, Y.; Cao, C. Sulforaphane induces autophagy by inhibition of HDAC6-mediated PTEN activation in triple negative breast cancer cells. Life Sci. 2018, 213, 149–157. [Google Scholar] [CrossRef]
  188. Abbaoui, B.; Lucas, C.R.; Riedl, K.M.; Clinton, S.K.; Mortazavi, A. Cruciferous Vegetables, Isothiocyanates, and Bladder Cancer Prevention. Mol. Nutr. Food Res. 2018, 62, e1800079. [Google Scholar] [CrossRef]
  189. Bhattacharya, A.; Li, Y.; Wade, K.L.; Paonessa, J.D.; Fahey, J.W.; Zhang, Y. Allyl isothiocyanate-rich mustard seed powder inhibits bladder cancer growth and muscle invasion. Carcinogenesis 2010, 31, 2105–2110. [Google Scholar] [CrossRef] [PubMed]
  190. He, C.; Huang, L.; Lei, P.; Liu, X.; Li, B.; Shan, Y. Sulforaphane Normalizes Intestinal Flora and Enhances Gut Barrier in Mice with BBN-Induced Bladder Cancer. Mol. Nutr. Food Res. 2018, 62, e1800427. [Google Scholar] [CrossRef] [PubMed]
  191. Jin, C.Y.; Molagoda, I.M.N.; Karunarathne, W.; Kang, S.H.; Park, C.; Kim, G.Y.; Choi, Y.H. TRAIL attenuates sulforaphane-mediated Nrf2 and sustains ROS generation, leading to apoptosis of TRAIL-resistant human bladder cancer cells. Toxicol. Appl. Pharmacol. 2018, 352, 132–141. [Google Scholar] [CrossRef] [PubMed]
  192. Leone, A.; Diorio, G.; Sexton, W.; Schell, M.; Alexandrow, M.; Fahey, J.W.; Kumar, N.B. Sulforaphane for the chemoprevention of bladder cancer: Molecular mechanism targeted approach. Oncotarget 2017, 8, 35412–35424. [Google Scholar] [CrossRef] [PubMed]
  193. Veeranki, O.L.; Bhattacharya, A.; Tang, L.; Marshall, J.R.; Zhang, Y. Cruciferous vegetables, isothiocyanates, and prevention of bladder cancer. Curr. Pharmacol. Rep. 2015, 1, 272–282. [Google Scholar] [CrossRef] [PubMed]
  194. Kntayya, S.B.; Ibrahim, M.D.; Mohd Ain, N.; Iori, R.; Ioannides, C.; Abdull Razis, A.F. Induction of Apoptosis and Cytotoxicity by Isothiocyanate Sulforaphene in Human Hepatocarcinoma HepG2 Cells. Nutrients 2018, 10, 718. [Google Scholar] [CrossRef] [PubMed]
  195. Liu, P.; Wang, W.; Zhou, Z.; Smith, A.J.O.; Bowater, R.P.; Wormstone, I.M.; Chen, Y.; Bao, Y. Chemopreventive Activities of Sulforaphane and Its Metabolites in Human Hepatoma HepG2 Cells. Nutrients 2018, 10, 585. [Google Scholar] [CrossRef] [PubMed]
  196. Ren, J.; Yuan, L.; Wang, Y.; Chen, G.; Hu, K. Benzyl sulforaphane is superior to sulforaphane in inhibiting the Akt/MAPK and activating the Nrf2/ARE signalling pathways in HepG2 cells. J. Pharm. Pharmacol. 2018, 70, 1643–1653. [Google Scholar] [CrossRef]
  197. Ren, K.; Li, Z.; Li, Y.; Zhang, W.; Han, X. Sulforaphene enhances radiosensitivity of hepatocellular carcinoma through suppression of the NF-kappaB pathway. J. Biochem. Mol. Toxicol. 2017, 31, e21917. [Google Scholar] [CrossRef]
  198. Zou, X.; Qu, Z.; Fang, Y.; Shi, X.; Ji, Y. Endoplasmic reticulum stress mediates sulforaphane-induced apoptosis of HepG2 human hepatocellular carcinoma cells. Mol. Med. Rep. 2017, 15, 331–338. [Google Scholar] [CrossRef]
  199. Choi, Y.H. ROS-mediated activation of AMPK plays a critical role in sulforaphane-induced apoptosis and mitotic arrest in AGS human gastric cancer cells. Gen Physiol. Biophys. 2018, 37, 129–140. [Google Scholar] [CrossRef] [PubMed]
  200. Kiani, S.; Akhavan-Niaki, H.; Fattahi, S.; Kavoosian, S.; Babaian Jelodar, N.; Bagheri, N.; Najafi Zarrini, H. Purified sulforaphane from broccoli (Brassica oleracea var. italica) leads to alterations of CDX1 and CDX2 expression and changes in miR-9 and miR-326 levels in human gastric cancer cells. Gene 2018, 678, 115–123. [Google Scholar] [CrossRef] [PubMed]
  201. Elkashty, O.A.; Ashry, R.; Elghanam, G.A.; Pham, H.M.; Su, X.; Stegen, C.; Tran, S.D. Broccoli extract improves chemotherapeutic drug efficacy against head-neck squamous cell carcinomas. Med. Oncol. 2018, 35, 124. [Google Scholar] [CrossRef] [PubMed]
  202. Saha, K.; Fisher, M.L.; Adhikary, G.; Grun, D.; Eckert, R.L. Sulforaphane suppresses PRMT5/MEP50 function in epidermal squamous cell carcinoma leading to reduced tumor formation. Carcinogenesis 2017, 38, 827–836. [Google Scholar] [CrossRef] [PubMed]
  203. Li, X.; Zhao, Z.; Li, M.; Liu, M.; Bahena, A.; Zhang, Y.; Nambiar, C.; Liu, G. Sulforaphane promotes apoptosis, and inhibits proliferation and self-renewal of nasopharyngeal cancer cells by targeting STAT signal through miRNA-124-3p. Biomed. Pharmacother. 2018, 103, 473–481. [Google Scholar] [CrossRef] [PubMed]
  204. Arcidiacono, P.; Ragonese, F.; Stabile, A.; Pistilli, A.; Kuligina, E.; Rende, M.; Bottoni, U.; Calvieri, S.; Crisanti, A.; Spaccapelo, R. Antitumor activity and expression profiles of genes induced by sulforaphane in human melanoma cells. Eur. J. Nutr. 2018, 57, 2547–2569. [Google Scholar] [CrossRef]
  205. Kumar, R.; de Mooij, T.; Peterson, T.E.; Kaptzan, T.; Johnson, A.J.; Daniels, D.J.; Parney, I.F. Modulating glioma-mediated myeloid-derived suppressor cell development with sulforaphane. PLoS ONE 2017, 12, e0179012. [Google Scholar] [CrossRef]
  206. Wu, S.; Zhou, Y.; Yang, G.; Tian, H.; Geng, Y.; Hu, Y.; Lin, K.; Wu, W. Sulforaphane-cysteine induces apoptosis by sustained activation of ERK1/2 and caspase 3 in human glioblastoma U373MG and U87MG cells. Oncol. Rep. 2017, 37, 2829–2838. [Google Scholar] [CrossRef] [Green Version]
  207. Sita, G.; Hrelia, P.; Graziosi, A.; Morroni, F. Sulforaphane from Cruciferous Vegetables: Recent Advances to Improve Glioblastoma Treatment. Nutrients 2018, 10, 1755. [Google Scholar] [CrossRef]
  208. Liu, K.C.; Shih, T.Y.; Kuo, C.L.; Ma, Y.S.; Yang, J.L.; Wu, P.P.; Huang, Y.P.; Lai, K.C.; Chung, J.G. Sulforaphane Induces Cell Death Through G2/M Phase Arrest and Triggers Apoptosis in HCT 116 Human Colon Cancer Cells. Am. J. Chin. Med. 2016, 44, 1289–1310. [Google Scholar] [CrossRef]
  209. Martin, S.L.; Kala, R.; Tollefsbol, T.O. Mechanisms for the Inhibition of Colon Cancer Cells by Sulforaphane through Epigenetic Modulation of MicroRNA-21 and Human Telomerase Reverse Transcriptase (hTERT) Down-regulation. Curr. Cancer Drug Targets 2018, 18, 97–106. [Google Scholar] [CrossRef] [PubMed]
  210. Tao, S.; Rojo de la Vega, M.; Chapman, E.; Ooi, A.; Zhang, D.D. The effects of NRF2 modulation on the initiation and progression of chemically and genetically induced lung cancer. Mol. Carcinog. 2018, 57, 182–192. [Google Scholar] [CrossRef] [PubMed]
  211. Zhu, J.; Wang, S.; Chen, Y.; Li, X.; Jiang, Y.; Yang, X.; Li, Y.; Wang, X.; Meng, Y.; Zhu, M.; et al. miR-19 targeting of GSK3beta mediates sulforaphane suppression of lung cancer stem cells. J. Nutr. Biochem. 2017, 44, 80–91. [Google Scholar] [CrossRef] [PubMed]
  212. Kim, B.G.; Fujita, T.; Stankovic, K.M.; Welling, D.B.; Moon, I.S.; Choi, J.Y.; Yun, J.; Kang, J.S.; Lee, J.D. Sulforaphane, a natural component of broccoli, inhibits vestibular schwannoma growth in vitro and in vivo. Sci. Rep. 2016, 6, 36215. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  213. Lubelska, K.; Wiktorska, K.; Mielczarek, L.; Milczarek, M.; Zbroinska-Bregisz, I.; Chilmonczyk, Z. Sulforaphane Regulates NFE2L2/Nrf2-Dependent Xenobiotic Metabolism Phase II and Phase III Enzymes Differently in Human Colorectal Cancer and Untransformed Epithelial Colon Cells. Nutr. Cancer 2016, 68, 1338–1348. [Google Scholar] [CrossRef] [PubMed]
  214. Cheng, Y.M.; Tsai, C.C.; Hsu, Y.C. Sulforaphane, a Dietary Isothiocyanate, Induces G(2)/M Arrest in Cervical Cancer Cells through CyclinB1 Downregulation and GADD45beta/CDC2 Association. Int. J. Mol. Sci. 2016, 17, 1530. [Google Scholar] [CrossRef] [PubMed]
  215. Bauman, J.E.; Zang, Y.; Sen, M.; Li, C.; Wang, L.; Egner, P.A.; Fahey, J.W.; Normolle, D.P.; Grandis, J.R.; Kensler, T.W.; et al. Prevention of Carcinogen-Induced Oral Cancer by Sulforaphane. Cancer Prev. Res. 2016, 9, 547–557. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  216. Sharma, D.; Sukumar, S. Big punches come in nanosizes for chemoprevention. Cancer Prev. Res. 2013, 6, 1007–1010. [Google Scholar] [CrossRef] [PubMed]
  217. Barba, F.J.; Nikmaram, N.; Roohinejad, S.; Khelfa, A.; Zhu, Z.; Koubaa, M. Bioavailability of Glucosinolates and Their Breakdown Products: Impact of Processing. Front. Nutr. 2016, 3, 24. [Google Scholar] [CrossRef]
  218. Okunade, O.; Niranjan, K.; Ghawi, S.K.; Kuhnle, G.; Methven, L. Supplementation of the Diet by Exogenous Myrosinase via Mustard Seeds to Increase the Bioavailability of Sulforaphane in Healthy Human Subjects after the Consumption of Cooked Broccoli. Mol. Nutr. Food Res. 2018, 62, e1700980. [Google Scholar] [CrossRef]
  219. Hayes, A.; Melrose, J. Glycosaminoglycan and Proteoglycan Biotherapeutics in Articular Cartilage Protection and Repair Strategies: Novel Approaches to Visco-supplementation in Orthobiologics. Adv. Ther. 2019. [Google Scholar] [CrossRef]
  220. Kim, J.; Bang, H.; Ahn, M.; Choi, Y.; Kim, G.O.; Shin, T. Allyl isothiocyanate reduces liver fibrosis by regulating Kupffer cell activation in rats. J. Vet. Med. Sci. 2018, 80, 893–897. [Google Scholar] [CrossRef] [PubMed]
  221. Lee, H.W.; Lee, C.G.; Rhee, D.K.; Um, S.H.; Pyo, S. Sinigrin inhibits production of inflammatory mediators by suppressing NF-kappaB/MAPK pathways or NLRP3 inflammasome activation in macrophages. Int. Immunopharmacol. 2017, 45, 163–173. [Google Scholar] [CrossRef] [PubMed]
  222. Mazumder, A.; Dwivedi, A.; du Preez, J.L.; du Plessis, J. In vitro wound healing and cytotoxic effects of sinigrin-phytosome complex. Int. J. Pharm. 2016, 498, 283–293. [Google Scholar] [CrossRef]
  223. Awasthi, S.; Saraswathi, N.T. Elucidating the molecular interaction of sinigrin, a potent anticancer glucosinolate from cruciferous vegetables with bovine serum albumin: Effect of methylglyoxal modification. J. Biomol. Struct. Dyn. 2016, 34, 2224–2232. [Google Scholar] [CrossRef] [PubMed]
  224. Jie, M.; Cheung, W.M.; Yu, V.; Zhou, Y.; Tong, P.H.; Ho, J.W. Anti-proliferative activities of sinigrin on carcinogen-induced hepatotoxicity in rats. PLoS ONE 2014, 9, e110145. [Google Scholar] [CrossRef] [PubMed]
  225. Hwang, E.S.; Lee, H.J. Allyl isothiocyanate and its N-acetylcysteine conjugate suppress metastasis via inhibition of invasion, migration, and matrix metalloproteinase-2/-9 activities in SK-Hep 1 human hepatoma cells. Exp. Biol. Med. 2006, 231, 421–430. [Google Scholar] [CrossRef]
  226. Smith, T.K.; Lund, E.K.; Clarke, R.G.; Bennett, R.N.; Johnson, I.T. Effects of Brussels sprout juice on the cell cycle and adhesion of human colorectal carcinoma cells (HT29) in vitro. J. Agric. Food Chem. 2005, 53, 3895–3901. [Google Scholar] [CrossRef]
  227. Smith, T.K.; Lund, E.K.; Parker, M.L.; Clarke, R.G.; Johnson, I.T. Allyl-isothiocyanate causes mitotic block, loss of cell adhesion and disrupted cytoskeletal structure in HT29 cells. Carcinogenesis 2004, 25, 1409–1415. [Google Scholar] [CrossRef] [Green Version]
  228. Musk, S.R.; Smith, T.K.; Johnson, I.T. On the cytotoxicity and genotoxicity of allyl and phenethyl isothiocyanates and their parent glucosinolates sinigrin and gluconasturtiin. Mutat. Res. 1995, 348, 19–23. [Google Scholar] [CrossRef]
  229. Brabban, A.D.; Edwards, C. The effects of glucosinolates and their hydrolysis products on microbial growth. J. Appl. Bacteriol. 1995, 79, 171–177. [Google Scholar] [CrossRef] [PubMed]
  230. Morse, M.A.; Wang, C.X.; Amin, S.G.; Hecht, S.S.; Chung, F.L. Effects of dietary sinigrin or indole-3-carbinol on O6-methylguanine-DNA-transmethylase activity and 4-(methylnitrosamino)-1-(3-pyridyl)-1-butanone-induced DNA methylation and tumorigenicity in F344 rats. Carcinogenesis 1988, 9, 1891–1895. [Google Scholar] [CrossRef] [PubMed]
Figure 1. Generic structure of the Glucosinolates showing glucose, sulphation and the aglycone side chain (R) used to categorize the aliphatic, indolic or aromatic glucosinolates.
Figure 1. Generic structure of the Glucosinolates showing glucose, sulphation and the aglycone side chain (R) used to categorize the aliphatic, indolic or aromatic glucosinolates.
Biomedicines 07 00062 g001
Figure 2. Enzymatic processing of the glucosinolates by myrosinase into bioactive components.
Figure 2. Enzymatic processing of the glucosinolates by myrosinase into bioactive components.
Biomedicines 07 00062 g002
Figure 3. The antimicrobial activities of glucosinolate thiocyanate, iso-thiocyanate and nitrile derivatives.
Figure 3. The antimicrobial activities of glucosinolate thiocyanate, iso-thiocyanate and nitrile derivatives.
Biomedicines 07 00062 g003
Figure 4. Lipid and moisture storage in Brassica napus seeds (a,b) and hypocotyl (c,d) visualized by non-invasive MRI. The concentration of water and oil are colour coded red (high); blue, (low). Crucifern immunolocalisation (e) and iodine stained starch (f) modified from [24] under Creative Commons Deed Attribution licence 2.5. oc,/ic outer/inner cotyledon; el, endosperm; hy, hypocotyl; sc, seed coat; cc, central cylinder.
Figure 4. Lipid and moisture storage in Brassica napus seeds (a,b) and hypocotyl (c,d) visualized by non-invasive MRI. The concentration of water and oil are colour coded red (high); blue, (low). Crucifern immunolocalisation (e) and iodine stained starch (f) modified from [24] under Creative Commons Deed Attribution licence 2.5. oc,/ic outer/inner cotyledon; el, endosperm; hy, hypocotyl; sc, seed coat; cc, central cylinder.
Biomedicines 07 00062 g004
Figure 5. Anatomical description of a mustard (Brassica napus) plant showing its characteristic four petal flower head, stamen, seed pods, leaf arrangements and seeds. Image from Franz Eugen Koehler archive, Koehlers Medicinal Plants, Germany 1887. Image reproduced from Wikimedia Commons Repository, https://en.wikipedia.org/wiki/Rapeseed (accessed 19 August 2019), copyright lapsed.
Figure 5. Anatomical description of a mustard (Brassica napus) plant showing its characteristic four petal flower head, stamen, seed pods, leaf arrangements and seeds. Image from Franz Eugen Koehler archive, Koehlers Medicinal Plants, Germany 1887. Image reproduced from Wikimedia Commons Repository, https://en.wikipedia.org/wiki/Rapeseed (accessed 19 August 2019), copyright lapsed.
Biomedicines 07 00062 g005
Table 1. Examples of Glucosinolate rich Cruciferous plants of the Brassicacea family order Capparales.
Table 1. Examples of Glucosinolate rich Cruciferous plants of the Brassicacea family order Capparales.
Plant Types
Brocolli
Brocolli Sprouts
Cabbage
Brussell Sprouts
Cauliflower
Daikon (Japanese radish)
Daikon sprouts
Garden Cress (Lepidum sativum)
Kale
Rapeseed (Brassica napus)
Wasabi (Wasabia japonica)
White Mustard (Sinapis alba)
Yellow Mustard (Brassica juncea)
Bok Choi
Arugula, Rocket (Eruca sativa)
Collard Greens
Horseradish
Kohlrabi
Radish
Rutabaga/turnip
Watercress
Mustard Greens
Table 2. Aliphatic (A), Indolic (B) and Aromatic (C) Glucosinolates and their contents in Brassica vegetables Capparales Order (μmol/100 g wet wt tissue) (data modified from [24,25,26]).
Table 2. Aliphatic (A), Indolic (B) and Aromatic (C) Glucosinolates and their contents in Brassica vegetables Capparales Order (μmol/100 g wet wt tissue) (data modified from [24,25,26]).
Glucosinolate Trivial NameAglycone
Chemical Name
Aglycone
Structure
Vegetable SourceGlucosinolate Content μmol/100 g)
A. Aliphatic
Glucoibervirin3-Methylthiopropyl Biomedicines 07 00062 i001Green Cauliflower
White Cauliflower
0–11.8
1.5–7.1
Glucoerucin4-Methylthiobutyl Biomedicines 07 00062 i002Rocket52–109
Glucoiberin3-Methylsulfinylbutyl Biomedicines 07 00062 i003Brocolli Sprouts
Savoy Cabbage
59–181
24–50
Glucoraphanin4-Methylsulfinylbutyl Biomedicines 07 00062 i004Brocolli Sprouts
Brocolli
233–676
24–285
SinigrinProp-2-enyl Biomedicines 07 00062 i005Brussels Sprouts
White Cauliflower
46–91
57–121
GluconapinBut-2-enyl Biomedicines 07 00062 i006Pak Choi24–157
GlucovrassicanapinPent-2-enyl Biomedicines 07 00062 i007Chinese Cabbage
Pak Choi
2.3–25
27–69
Progoitrin(2R)-Hydroxybut-3-enyl Biomedicines 07 00062 i008Turnip
Chinese Brocolli
18–41
49
B. Indolic
GlucobrassicinIndol-3-ylmethyl Biomedicines 07 00062 i009Brocolli
White Cauliflower
13–29
11–33
4-Hydroxy-Glucobrassicin4-Hydroxy-3-indolylmethyl Biomedicines 07 00062 i010Brocolli
White Cauliflower
0.1–3.3
0.2–2.8
4-Methoxy–Glucobrassicin4-Methoxy-3-indolylmethyl Biomedicines 07 00062 i011Brocolli
White Cauliflower
0.9–2.8
0.7–3.2
Neo-Glucobrassicin4-Methoxyindol-3-ylmethyl Biomedicines 07 00062 i012Brocolli
White Cauliflower
1.8–13
0.9–3.0
C. Aromatic
GlucotropaeolinBenzyl Biomedicines 07 00062 i013Garden CressNA
GluconasturtiinPenylethyl Biomedicines 07 00062 i014Water CressNA
Table 3. The WHO Dirty Dozen Pathogen List of Problematic Super Bugs *.
Table 3. The WHO Dirty Dozen Pathogen List of Problematic Super Bugs *.
Priority CategoryBacteriumDrug Resistance
CriticalAcinetobacter baumannii
Pseudomonas aeruginosaCarbapanem
ESBL** producing members of the Enterobacteriaceae
HighEnterococcus faeciumVancomycin
Staphylococcus aureusMethicillin/Vancomycin
Helicobacter pyloriClathrimycin
Campylobacter sppFluoroquinolone
SalmonellaeFluoroquinolone
Neisseria gonorrhoeaeCepalosporin/Fluoriquinolone
MediumStreptococcus pneumoniaPenicillin
Haemophilus influenzaeAmpicillin
Shigella sppFluoriquinolone
* http://www.who.int/news-room/detail/27-02-2017-who-publishes-list-of-bacteria-for-which-new-antibiotics-are-urgently-needed (accessed 12 January 2018); ** Certain strains of bacteria are resistant to treatments with commonly used antibiotics such as penicillin and cephalosporins. These bacteria produce enzymes known as Extended Spectrum Beta-Lactamases (ESBL). ESBL producing bacteria are resistant to most types of third generation antibiotics and include strains of Klebsiella pneumoniae, Klebsiella oxytoca and Escherichia coli. Enterobacter spp., Salmonella spp., Morganella morganii, Proteus mirabilis, Serratia marcescens and Pseudomonas aeruginosa produce ESBLs relatively infrequently.
Table 4. Combination Therapies of Sulphoraphane and Conventional Anti-Cancer and Antibacterial Drugs.
Table 4. Combination Therapies of Sulphoraphane and Conventional Anti-Cancer and Antibacterial Drugs.
Compound Used in Combination TherapyReference
SFN-Selenium nanoparticles[122]
Paclitaxel[9]
Cisplatin[123]
Luteolin[124]
Clofarabine[125]
Doxorubicin[126]
5-fluorouracil[127]
HistoneH3[128]
Withaferin A[129]
Hispidulin[130]
Carboplatin[131]
Docetaxel[132]
Lapatinib[133]
PR-104A[134]
Table 5. Some Examples of The Diverse Therapeutic Applications of Sulphoraphane.
Table 5. Some Examples of The Diverse Therapeutic Applications of Sulphoraphane.
Medical Conditions Treated with Sulphoraphane
Spatial learning and memory dysfunction [135]
Chemotherapy-induced neuropathic pain [136]
Protection of granulosa cells against oxidative stress[137]
Cadmium-mediated carcinogenesis[138]
Oxidative stress in cultured adult cardiomyocytes [139]
Protective effects of glucosinolate hydrolysis products in neurodegenerative diseases[140]
Clearance of Amyloid-β and Tau protein in a mouse model of AD[141]
Experimental diabetic peripheral neuropathy [142]
Joint inflammation in a murine adjuvant-induced mono-arthritis [143]
Protection against cognitive impairment in AD-like lesions in diabetes [144]
Anti-inflammatory effect of SFN on human THP-1 macrophages in a murine AD model[145]
Inhibition of oxidative stress/inflammation improves cardiac function in a Rabbit Model of Chronic Heart Failure [146]
Inhibition of class IIa histone deacetylase activity [147]
Apoptosis via microtubule disruption in cancer [148]
Inhibition of LPS-Induced Inflammation/cytotoxicity/oxidative microglial stress [149]
Down-regulation of MAPK/NF-κB signaling in LPS-activated BV-2 microglia[150]
Inhibition of oxidative stress in an in vitro model of age-related macular degeneration[151]
Modification of Histone H3, unpacking of chromatin, to prime defence [128]
Modulation of oxidative stress and inflammation in rats with toxic hepatitis [152]
Modulation of oxidative damage in lead exposed rat hippocampus [153]
Prevention of dexamethasone-induced myotube atrophy via Akt/Foxo1 [154]
Induction of p53 deficient SW480 cell apoptosis by ROS MAPK signaling [155]
Role of microRNAs in the chemopreventive activity of SFN [156]
Novel phosphonate analogs of SFN with in vitro and in vivo anti-cancer activity[157]
Gastrointestinal protection against H. pylori and NSAID-Induced Oxidative Stress[158]
Protection against sodium valproate-induced acute liver injury [159]
Enhanced SFN cardioprotection against oxidative stress by 17β-Estradiol[159]
Photoprotective Effects of SFN and Hispidulin[130]
Improvement of neuronal mitochondrial function in brain tissue[160]
Chemoprevention of oxidative stress-associated with oral carcinogenesis [161]
Amelioration of bladder dysfunction via activation of Nrf2-ARE Pathway[162]
Protection against aortic complications in diabetes[163]
Anti-inflammatory effect against amyloid-β peptide via STAT-1 dephosphorylation and activation of Nrf2/HO-1[164]
Table 6. Sulphoraphane Applications in Cancer Models.
Table 6. Sulphoraphane Applications in Cancer Models.
Cancer TypeReference
Leukemia[87,165,166,167,168,169,170]
Prostate cancer [91,95,171,172,173]
non-small cell lung cancer cells [131,174,175]
Pancreatic cancer[176,177,178,179]
Breast cancer[92,93,125,126,127,129,132,133,180,181,182,183,184,185,186,187]
Bladder cancer[162,188,189,190,191,192,193]
Ovarian cancer[123]
HepG2 Carcinoma Cells[194,195,196,197,198]
Gastric cancer[199,200]
Squamous cell carcinoma[201,202]
Nasopharangeal cancer[203]
Melanoma[204]
Glioma[163,205,206,207]
Colon cancer[134,208,209]
Lung cancer[210,211]
Schwannoma[212]
Colorectal cancer[213]
Cervical cancer[214]
Oral cancer[215,216]
Table 7. The Varied Applications of Sinigrin in Biomedicine.
Table 7. The Varied Applications of Sinigrin in Biomedicine.
ApplicationReference
Reduction of liver fibrosis[220]
Suppression of NF-κB/MAPK and NLRP3 inflammasome activation in macrophages[221]
Promotion of wound healing [113,222]
Anti-cancer properties in methyl glyoxal modification[223]
Anti-proliferative activity on carcinogen-induced hepatotoxicity[224]
Biofumigation of potato cyst nematode[21]
Inhibition of Listeria monocytogenes on bologna sausages[112]
inhibition of invasion, migration, MMP-2/-9 activities in SK-Hep 1 human hepatoma cells[225]
Brussel sprout juice mediated effects on cell cycle and adhesion of human colorectal carcinoma cells (HT29) in vitro[226]
AITC mediated mitotic block, loss of cell adhesion/disrupted cytoskeleton in HT29 cells[227]
Cytotoxicity and genotoxicity of allyl and phenethyl isothiocyanates, glucosinolates, sinigrin and gluconasturtiin[228]
Inhibition of microbial growth[47,65,229]
Effects of dietary sinigrin or indole-3-carbinol on O6-methylguanine-DNA-transmethylase activity and 4-(methylnitrosamino)-1-(3-pyridyl)-1-butanone-induced DNA methylation and tumorigenicity in F344 rats[230]

Share and Cite

MDPI and ACS Style

Melrose, J. The Glucosinolates: A Sulphur Glucoside Family of Mustard Anti-Tumour and Antimicrobial Phytochemicals of Potential Therapeutic Application. Biomedicines 2019, 7, 62. https://doi.org/10.3390/biomedicines7030062

AMA Style

Melrose J. The Glucosinolates: A Sulphur Glucoside Family of Mustard Anti-Tumour and Antimicrobial Phytochemicals of Potential Therapeutic Application. Biomedicines. 2019; 7(3):62. https://doi.org/10.3390/biomedicines7030062

Chicago/Turabian Style

Melrose, James. 2019. "The Glucosinolates: A Sulphur Glucoside Family of Mustard Anti-Tumour and Antimicrobial Phytochemicals of Potential Therapeutic Application" Biomedicines 7, no. 3: 62. https://doi.org/10.3390/biomedicines7030062

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