Next Article in Journal
Lactococcus garvieae FUA009, a Novel Intestinal Bacterium Capable of Producing the Bioactive Metabolite Urolithin A from Ellagic Acid
Next Article in Special Issue
Bioactive Compounds, Health Benefits and Food Applications of Grape
Previous Article in Journal
Physicochemical and Functional Properties of Texturized Vegetable Proteins and Cooked Patty Textures: Comprehensive Characterization and Correlation Analysis
Previous Article in Special Issue
Phytochemical Profile and Chemopreventive Properties of Cooked Glutinous Purple Rice Extracts Using Cell-Based Assays and Rat Model
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

In Vitro Toxicity Studies of Bioactive Organosulfur Compounds from Allium spp. with Potential Application in the Agri-Food Industry: A Review

by
Antonio Cascajosa-Lira
1,
Pedro Andreo-Martínez
2,
Ana Isabel Prieto
1,*,
Alberto Baños
3,
Enrique Guillamón
3,
Angeles Jos
1 and
Ana M. Cameán
1
1
Área de Toxicología, Facultad de Farmacia, Universidad de Sevilla, Profesor García González n 2, 41012 Seville, Spain
2
Department of Agricultural Chemistry, Faculty of Chemistry, Campus of Espinardo, University of Murcia, 30100 Murcia, Spain
3
DMC Research Center, Camino de Jayena, 82, 18620 Alhendín, Spain
*
Author to whom correspondence should be addressed.
Foods 2022, 11(17), 2620; https://doi.org/10.3390/foods11172620
Submission received: 20 July 2022 / Revised: 5 August 2022 / Accepted: 25 August 2022 / Published: 29 August 2022
(This article belongs to the Special Issue Bioactive Compounds, Antioxidants, and Health Benefits)

Abstract

:
Organosulfur compounds (OSCs) are secondary metabolites produced by different Allium species which present important biological activities such as antimicrobial, antioxidant, anti-inflammatory antidiabetic, anticarcinogenic, antispasmodic, etc. In recent years, their use has been promoted in the agri-food industry as a substitute for synthetic preservatives, increasing potential accumulative exposure to consumers. Before their application in the food industry, it is necessary to pass a safety assessment as specified by the European Food Safety Authority (EFSA). This work reviews the scientific literature on OSCs regarding their in vitro toxicity evaluation following PRISMA guidelines for systematic reviews. Four electronic research databases were searched (Web of Science, Scopus, Science Database and PubMed) and a total of 43 works were selected according to predeterminate inclusion and exclusion criteria. Different data items and the risk of bias for each study were included. Currently, there are very few in vitro studies focused on investigating the potential toxicity of OSCs. Most research studies aimed to evaluate the cytotoxicity of OSCs to elucidate their antiproliferative effects focusing on their therapeutic aspects using cancer cell lines as the main experimental model. The results showed that diallyl disulfide (DADS) is the compound most studied, followed by diallyl trisulfide (DATS), diallyl sulfide (DAS), Allicin and Ajoene. Only 4 studies have been performed specifically to explore the safety of OSCs for agri-food applications, and genotoxicity studies are limited. More toxicity studies of OSCs are necessary to ensure consumers safety and should mainly be focused on the evaluation of genotoxicity and long-term toxicity effects.

Graphical Abstract

1. Introduction

The name Allium is derived from the Greek word “aleo” defined as “to avoid” due to its strong odor [1]. The Allium genus includes about 600 to 700 species, with onion (A. cepa) and garlic (A. sativum) as the most well-known characteristic and edible species [2]. They were among the first domestic plants documented [3].
The main interest of these species lies in their organosulfur compounds (OSCs), which are secondary metabolites (e.g., ajoenes and thiosulphinates) with biological action and a distinctive smell of Allium species. When tissues are damaged, a cascade of enzymatic reactions by alliinase occurs, resulting in a degradation of initial compounds, and thus, sequentially, new highly reactive and biologically active compounds appear, giving rise to a great variety of byproducts [4]. These phytochemical compounds are biosynthesized for two main purposes: as a defense mechanism against biotic stresses, and as a mediator for pollination [1]. The Allium spp. are also well known for their multiple biological effects, such as antiviral, antimicrobial, antioxidant, antiprotozoal, antidiabetic, anticarcinogenic, antispasmodic, antimutagenic, anti-amnesic, antiasthmatic, anti-inflammatory, neuroprotective, hepatoprotective, hypotensive, immunomodulatory, hypoglycemic, as well as their prebiotic properties [2,4,5,6,7,8,9,10,11,12]. In relation to antibacterial activity, gram-positive bacteria seem to be more sensitive than gram-negative bacteria to these compounds. This could be due to lipopolysaccharides present in the outer membrane of Gram-negative bacteria. However, the mechanism of action is not yet sufficiently clarified, and it seems that it is associated with the inactivation of thiolic bacterial enzymes. Moreover, the mechanism of the anti-inflammatory activity of some OSCss can be associated with the inhibition of TNF-α-initiated secretion of pro-inflammatory cytokines from epithelial digestive cells. In addition, compounds such DADS, DATS and SAC inhibit the formation of inflammatory lipopolysaccharide by repressing NF-κB and MAPK signaling pathways [2].
Recently, Rochetti et al. [13] carried out an extensive phytochemical investigation of nine Allium species, highlighting a promising nutraceutical potential of these species. These properties give the Allium genus diverse uses in several areas of knowledge, and although traditionally this genus has already been used medicinally, in recent years its use has incremented in the agri-food industry [14]. Thus, Allium extracts and OSCss can be potentially used in food and feed sectors as a substitute for synthetic preservatives due to their significant antimicrobial properties [11,15]; nevertheless, their use in food is limited due to their organoleptic properties [1]. The use of OSCs and Allium extracts to control food spoilage have also been studied [16,17]. Several OSCs, such as propyl-propane-thiosulfinate (PTS) and propyl-propane thiosulfonate (PTSO) have a potential application in maize storage, mainly due to their antifungal and antimycotoxigenic activity. They could be used in concentrations in the order of parts per million to reduce up to 90% the concentrations of mycotoxins produced by Fusarium [18]. Moreover, due to their antioxidant and antibacterial activities, PTSO and PTS inserted in a synthetic matrix have been also proposed for usage in active food-packaging to better preserve salads [19,20]. As a consequence of their new applications, the potential accumulative exposure to consumers has increased, and a safe range of concentrations for their use in the industry should be established to reach the market and avoid risks for consumers (Table 1).
Furthermore, synthetic chemical additives, such as butylhydroxytoluene (BHT) and Butylhydroxyanisole (BHA), have shown problems regarding their safety, including carcinogenicity and sensitization in consumers [43]. For this reason, there is a call for replacement with natural and safe alternatives of natural origin, such as the bacteriocins [44] like Nisin (E234), a peptide produced from Lactococcus lactis, vegetable extracts such as citrus extracts rich in flavonoids [44] or essential oils (EO) with preservative properties, which have been reported to have antimicrobial and antioxidant properties [45]. In the case of garlic EOs, some authors demonstrated their preservation and antimicrobial capacities (including PTSO and PTS in its composition) [46,47]. However, important adverse effects produced by these compounds have also been described such as allergic reactions, gastrointestinal tract injury, weight loss, anemia, and toxicity to the liver, heart, and kidney [16]. Thus, garlic has been classified as a type I allergen.
Because of this all, to carry out all possible applications in the agri-food industry, it is necessary that OSCs pass a safety assessment as specified by the European Food Safety Authority (EFSA) [48,49,50,51]. This safety evaluation includes a wide array of tests both in vitro and in vivo. The “Guidance for submission for food additive evaluations” by EFSA [48] described the need of a toxicological evaluation in the following core areas: genotoxicity, toxicokinetics, toxicity comprising chronic, subchronic and carcinogenicity, developmental and reproductive toxicity. For the toxicological research, a tiered approach is followed, initially using fewer complex tests to obtain hazard data. These are then evaluated to determine if they are sufficient for risk assessment or, if not, for designing studies at higher tiers [48]. Therefore, in vitro studies are usually the initial step in the toxicological evaluation of any compound.
Thus, the objective of this work is to provide a systematic overview of the scientific literature of OSCs with antibacterial, antifungal, and other properties present in the Allium genus (Table 1) in regard to their in vitro toxicity evaluation. To achieve this purpose, the authors have followed the PRISMA 2020 guidelines for systematic reviews. This information could contribute to the safe use of these compounds in the agri-food sector.

2. Materials and Methods

This research was performed according to the Preferred Reporting Items for a Systematic Review and Meta-analysis (PRISMA) statement [52]. The question to be answered was: Do Allium compounds have toxic activity in vitro?

2.1. Protocol and Registration

The protocol for the present systematic review was not registered in any Systematic Review and Meta-analysis database.

2.2. Eligibility and Exclusion Criteria

International studies were considered. The eligibility criteria of the present systematic review were as follows: Inclusion criteria: (1) articles on Allium toxicity in vitro; (2) articles published prior to 21 September 2021; and (3) articles reporting comprehensive results and/or information on the field. Exclusion criteria: (1) unsystematic and narrative reviews; (2) articles published in a language other than English; (3) proceedings of conferences and dissertations; (4) books or book chapters; (5) editorial material; (6) articles dealing with Allium in vitro in which the test item is not a naturally occurring alliaceous compound (e.g., synthetically modified allicin) in the genus Allium or when the test has no toxicological relevance (e.g., protective effects of organosulfur compounds towards N-nitrosamine-induced DNA damage) as represented in the flowchart (Figure 1).

2.3. Information Sources and Search Strategy

The electronic research databases Web of Science, Scopus, Science Database and PubMed were searched on 21 September 2021. The search identified articles published from inception to 21 September 2021 inclusive. The Boolean strings chosen were: (“Propyl thiosulfinate oxide” OR “propyl-propane-thiosulfonate” OR “propyl propane thiosulfinate” OR “propyl-propane-thiosulfinate” OR “organosulfur compound*” OR diallyl* OR allicin OR alliin OR ajoene OR “dipropyl disulphide” OR “dipropyl sulphide” OR propiin) AND (toxicity OR cytotoxicity OR genotoxicity) AND (“in vitro” OR “cell line”). The searches included works published in all languages. The Web of Science database option search was “theme” in all databases. The Scopus database options search were: “title, abstract and keywords”. The Science Database option search was “all fields except full text (NOFT)” and the PubMed option search was “all fields”.

2.4. Study Selection

Once the selection criteria have been established, a three-step process was performed to review all records according to the eligibility criteria: first was reading the title, second, the abstract, and third, the entire text of the publication. The works obtained by the four databases were crossed with the EndNote X9 (Bld 12062) software to identify possible duplicates and to classify the works according to the exclusion and inclusion criteria. Two authors (PA-M and AC-L) formed the review team to implement measures to reduce random mistakes and bias at all review phases and independently examined titles, abstracts and full texts of the articles for possible addition. Conflicts on whether a given reference should be incorporated or not were determined through discussion.

2.5. Data Extraction and Data Items

The data items included for data extraction were: Assays performed, experimental model, concentration ranges and time exposure, and main results. This data extraction form is presented in Table 2.

2.6. Risk of Bias in Individual Studies

Bias can be judged to be a systematic mistake that can lead to an underestimation or overestimation of the true result [53]. The risk of bias for each incorporated work was evaluated using The Methods Guide for Comparative Effectiveness Reviews [54]. The characteristics of bias considered are shown in Table 3. Each risk of bias question scored 2 (fully reported), 1 (partially reported) or 0 (unclear/not reported). Scores for each risk of bias question were added together to give a total score between 0 and 10. A score of 0–4 was considered a high risk of bias; 5–7 was considered a moderate risk of bias; 8–10 was considered a low risk of bias. The risk of bias assessment was performed by the reviewer team. Differences were resolved by discussion between the three reviewers (AC-L, AC and AIP).

2.7. Results Construction and Statistical Analysis

Compiled information and findings in the studies were examined independently by the reviewer team in order to find conflicts in the extracted data, should they be present.

3. Results

3.1. Search Results

The procedure of article selection is shown in the flow chart of Figure 1. The research database Web of Science returned 461 works; Scopus returned 190 works; Science Database returned 9 works and PubMed returned 287 works. The 947 works found by the four databases combined were crossed with the EndNote X9 (Bld 12062) software to identify possible duplicates. A total of 307 works were eliminated in this stage. After evaluating the abstract of each of the remaining articles, those that were associated with the subject of the study (43) were selected. According with the flow diagram (Figure 1), some articles were excluded since they had no toxicological importance, e.g., “Effect of allicin on promastigotes and intracellular amastigotes of Leishmania donovani and L. infantum”. It should be noted that some of the studies could be introduced in more than one exclusion group but the final criterion was agreed by discussion of the review team. Finally, a total of 43 works were found to be eligible for the present systematic review following the full-text eligibility assessment.

3.2. Study Characteristics and Results of Individual Studies

The characteristics and main findings of the 43 selected articles in the present systematic review, such as Assays performed, experimental model, concentration ranges and time exposure and main results, are summarized in Table 2. Thus, the main tests carried out have focused on investigating cytotoxicity, cell apoptosis and ROS production against antioxidant assays. The MTT assay highlights how to determine the EC50 of these compounds in different cellular models. The flow cytometry is used for different determinations such as apoptosis, cell cycle, mitochondrial membrane potential and ROS. On the other hand, assays related to the mutagenicity and genotoxicity showed less interest. In this sense, Ames test, MN, comet assay and MLA have been performed. Regarding the experimental models, different cell lines have been used. Melanoma, lymphoma and gastrointestinal tract cells were the ones most often selected (Figure 2). The 43 selected articles were published between 1990 and 2021. According to the corresponding authors addresses, the articles were published in 19 different countries: 8 in China, 6 in Korea, 4 in USA, 3 in Spain, 3 in Poland, 2 in India, 2 in Taiwan, 2 in Germany, 2 in UK, 2 in Venezuela, 1 in Brazil, 1 in Israel, 1 in Egypt, 1 in Japan, 1 in Iran, 1 in France, 1 in Croatia, 1 in Turkey and 1 in Italy. The 43 included articles were published in 33 different journals, Food and Chemical Toxicology being the journals with most publications.

3.3. In Vitro Studies of OSCs from Allium spp. Focused on Safety Evaluation for Agri-Food Application

Of the 43 studies selected, only 4 have been performed specifically to explore the safety of OSCs from Allium spp. for agri-food applications [16,17,55,56] (see Table 2). Mellado-García et al. [17,18,19,20,21,22,23,24,25,26,27,28,29,30,31,32,33,34,35,36,37,38,39,40,41,42,43,44,45,46,47,48,49,50,51,52,53,54,55] studied the genotoxicity potential of PTSO and PTS using in vitro test battery including Salmonella typhimurium reverse mutation assay (Ames Test), The cytokinesis-block micronucleus cytome assay (MN) and single cell gel electrophoresis assay (comet assay). Both compounds reported negative results by Ames Test and genotoxic effects were described in similar concentration for PTSO and PTS by MN assay. However, MLA and comet assays showed contradictory results between these compounds. No cytotoxicity or mutagenicity of DPS, DPDS, and mixtures were reported by Llana-Ruiz-Cabello et al. [16]. In relation to polymer films of Allium cepa L., no induction of MN was observed, although the HTP films showed signs of mutagenicity by the Ames Test [56].
Table 2. Overview of the studies reporting the in vitro toxicological evaluation of OSCs from Allium spp.
Table 2. Overview of the studies reporting the in vitro toxicological evaluation of OSCs from Allium spp.
Allium Products (Pure Compound or Extract)Assays PerformedExperimental ModelConcentration Ranges and Time ExposureMain ResultsReference
Ajoene
Allicin
Survival by MTT assay
Total cell protein
FS4
BHK21
BJA-B
5–20 µg/mL for 48 h.EC50(ajoene): FS4 (36 µM) > BHK21 (30 µM) > BJA-B (12 µM).
ED50 (ajoene): FS4 (450 fmol/cell) > BHK21 (190 fmol/cell) > BJA-B (85 fmol/cell).
ED50(allicin): FS4 (720 fmol/cell) > BHK21 (430 fmol/cell) > BJA-B (185 fmol/cell).
[57]
AjoeneMetabolic activity by MTT assay
Cell death by trypan blue assay
Levels of GSH, GSSG and acidic aminoacids Glu and Asp
BJA-B cells173 and 82 nmol/mL for 0–6 h.


385 and 150 fmol/cell for 48 h.
In both experiments, ↓ cell viability in a dose and time dependent manner.
In the first minutes after exposure, GSH decreased and GSSG increased. The further course strongly depended on the dose.
The Glu pool showed an immediate increase and in a later stage decreased. The Asp pool showed the contrary pattern.
[58]
DDS
DAS
Clonal survival
Mass growth rate.
Anchorage-Independent Growth.
Control and differentiated HT29 cells1–3 µg/mL DDS 24 h
100–300 µg/mL DAS 24 h after which the cells were incubated in fresh medium for 10–12 days.

Cells were exposed to 24-h intervals for seven days.
Dq (concentration at which no cell killing occurs) 0.10 ± 0.03 µg/mL for DAS and was not found for DDS.
D37 (dose required to reduce survival from 100% to 37%) are 2.93 ± 0.14 µg/mL for DAS and 164 ± 12 µg/mL for DDS.
[59]
DAS, DADSClonal survival
Induction of chromosome aberrations
Induction of SCEs
CHO100–600 µg/mL DAS
2–10 µg/mL DDS
Cytotoxicity DADS > DAS.
Both compounds induced chromosome aberrations and SCEs (DADS > DAS)
+S9: reduction of the induction of SCEs by both compounds, and enhanced the generation of aberrations by DADS.
[60]
DAS, DADS, DPS, DPDSAmes testAmes test: Salmonella typhimurium strains TA98 or TA100 Ames test was performed with S9 and microsomes from DAS-, DADS-, DPS-, DPDS-treated rats (1 mmol/Kg) DAS, DPS, DPDS: increased activation of BaP, CP, N-PiP and PhIP, while DADS only increased mutagenicity of PhIP.
In contrast, some OCS inhibited the mutagenicity of different mutagens, while other enhanced it.
[61]
DAS, DADS, garlic extractCytotoxicity by MTT assay
Western blot analysis of Bcl-2, Bax and p53 expression
Northern blot analysis
Apoptosis by acridine orange staining
p-53-wild type H460 and p-53-null type H1299 non-small cell lung cancer cells (NSCLC)DAS and DADS (0–25 µM) and garlic extract (0–200 µg/mL) for 1 hThe cell growth was significantly inhibited by DAS and DADS and slightly inhibited by garlic extract. The OSCs compounds and garlic extract have apoptotic potential on lung cancer cells, and the mechanism was regulated through p53-dependent or p-53 independent related Bax/Bcl-2 dual pathway. [62]
DADSCell viability and apoptosis by flow cytometry
Oxidative stress (carbonylated proteins, MDA)
Western blot analysis
SH-SY5Y 50 µM for 12 and 24 h
50 µM up to 2 h
Blockage in G2/M phase
DADS induced a ROS-mediated activation of JNK/c-Jun pathway in neuroblastoma cells, and this activation led to apoptosis.
[63]
DADS Survival by MTT assay
Apoptosis by flow cytometry



Cell signals by western blot analysis of phosphorylated forms of GSK-3β and Akt, and p85a PI3K

Free radical levels and membrane lipid peroxidation
N18D3 10–200 µM for 2 h.
10, 25 µM for 2 h pretreatment and treated with 100 µM H2O2 for 30 min.

25, 100 µM for 2 h treatment with and without post-treatment of 100 µM H2O2 for 30 min.
Cellular viability was not affected up to 25 µM DAS.
↑ apoptotic cells at 100 µM of treatment and at 25 µM of pretreatment versus H2O2 treatment in these cells.

↑ the expressions of p85a PI3K, phosphorylated Akt and phosphorylated GSK-3 in N18D3 cells pretreated with 25 µM (2 h) and subsequently exposed to 100 µM H2O2 (30 min).
Treatment with 100 µM reduced these biomarkers in N18D3 cells.

↑ the levels of free radicals and membrane lipid peroxidation a concentration-dependent manner.
[64]
AllicinCell proliferation by MTT assay
Morphological apoptosis
DNA fragmentation assay
Cell cycle by flow cytometry
Caspase-3 determination
Expression of Cyt c, Bax and AIF by Western blot analysis
AGS 5–100 µg/mL for 6, 12, 24 and 48 h.Allicin caused inhibition of cellular growth in a concentration- and time-dependent manner.

DNA fragmentation and morphological changes (degeneration of neuritis, shrinkage of cell bodies and condensation of nuclei) in cells exposed to 5–20 µg/mL for 24 h.

45.2% apoptotic cells.
↑ in the sub-G1 DNA content.
↓ in the proportion in cells of S phase after exposure to 20 µg/mL of allicin for 24 h.

Allicin results in the release of Cyt c and in increase of mitochondrial Bax protein level. Caspase-3 activation and cleavage of PARP were not detected.
[65]
AjoeneMTS/PMS chromogenic assay








Cell adhesion assay
B16/BL6
HT-29
A549
MDA-MB-231
PANC-1
SKBR-3
NIH 3T3
3T3/HER2
Splenocytes

B16/BL6-LEC1
10–100 µM for 24 h









1–100 µM for 24 h
IC50(B16/BL6) = 18 µM
IC50(HT-29) = 19 µM
IC50(A549) = 41 µM
IC50(MDA-MB-231) = 7 µM
IC50(PANC-1) = 38 µM
IC50(SKBR-3) = 19 µM
IC50(NIH 3T) = 17 µM
IC50(3T3/HER2) = 9 µM
IC50(Splenocytes) ≥100 µM

B16/BL6 29% inhibition at 10 µM
[66]
DAS, DADS, DATSCell viability by trypan blue exclusion assay
Wright staining and ApopTag assay for apoptosis
ROS production
Intracellular free [Ca2+] by Fura-2 assay
mRNA expression of β-actin, bax, bcl-2, calpastatin
phosphorylation of stress kinases analysis
Western Blot analysis analysis
Mitocondrial membrane potential
Caspase 3 and 9 activity
GST activity
T98G
U87MG
100 µM DAS
100 µM DADS
25 µM DATS for 24 h
The three garlic compounds induced cytotoxic effects via ROS production, increase in endoplasmic reticulum (ER) stress, decrease mitochondrial membrane potential, activation of stress kinases and cysteine proteases. [67]
AllicinIn vitro tubulin polymerization assay and image analysisNIH-3T30.2–25 µMDepolymerizing effect of allicin in a concentration-dependent manner until 25 µM.
Disruptive effect of allicin increases with the duration of incubation.
[68]
Ajoene (≥98%)Cell viability by MTS assay
Apoptosis by flow cytometry
Microscopic evaluation
B16F10 1, 5, 10 µmol/L for 24 h5 and 10 µmol/L ↓ cell viability and this cytotoxic effect was not prevented by the addition of mevalonate or GGPP.

Ajoene (5 µmol/L) in combination with atorvastatin (0.1 mmol/L) or pravastatin (0.1 mmol/L) ↓ cell viability in a synergistic way.

Apoptosis observed by diminution in cell volume, condensation of cytoplasm.
[69]
DADSCell viability by MTT assay
Apoptosis by fluorescence microscopy and flow cytometry
p-ERK and ERK protein levels by Western blot analysis
MCF-750–400 μmol/L for 24 h.


200 μmol/L for 6, 12, 24 and 48 h.
↓ cellular viability and ↑ apoptosis in concentration-dependent manner. These effects were observed mainly from 200 μmol/L of treatment.

Inhibition of ERK and activation of SAPK/JNK and p38.
[70]
DASCell viability by trypan blue exclusion assay
ALP and LDH assays
ROS generation
Apoptosis by flow cytometry analysis
Cell cycle analysis
DNA fragmentation analysis
Immunoblot analysis of caspase-3, NF-κB, ERK-2
Colo 320 DM5–75 µM for 0–24 h

50 µM for 6, 12 h
50 µM for 12 h
50 µM for 12 h
50 µM ↓ cell viabilityALP and LDH decreased with time. ROS increased.~55% apoptosis. Cell cycle arrest at G2/M Oligonucleosomal-laddering, characteristic of apoptosis.Higher protein expression of caspase-3 and NF-κB and lower expression of ERK-2.[71]
DADSMTT assayApoptosis by flow cytometry, DNA fragmentation assay and morphology analysisWestern blot analysis of the expression of phosphor-MAPKs (ERK, p38)HL-601.25–20 mg/L for 96 h
5–20 mg/L for 24 h

10 mg/L for 24 h
10 mg/L ↓59.6% cell viability
Apoptosis was evidenced in a concentration-dependent manner by different assays.
Inhibition of ERK and activation of p38
[72]
DATSMTT assay

Apoptosis by flow cytometry


Expression of Bcl-2, Bax, Bcl-xL/Bcl-xS, Cyt c, caspase 9 and poly(ADP-ribose) polymerase by western blotting.
Activity of caspase 3
A375
M14
5–60 µM for 24, 48, 72 h
Exposure to IC50 for 72 h: A375 11.7 µM and M14 14.1 µM
IC50 for 24, 48, 72 h




IC50 for 16, 24, 36, 48 h
IC50(A375) = 11.7 µM
IC50(M14) = 14.1 µM
↑ percentage of apoptosis
Reduced Bcl-2 and Bcl-xL expression
Increase in caspase-3 activity with time
[73]
n-DADS
s-DADS
MTT assay

Cell cycle analysis

Apoptosis by flow cytometry and by fluorescence microscopy after staining with Hoechst 33,258
MCF-70.01–2.00 mmol/L 48 for 72 h.
0.01, 0.05, 0.25 mmol/L for 48 h.

0.05 mmol/L for 48 h, microscopy
s- and n-DADS present concentration- and time-dependent inhibitory effects and similar cytotoxicity in MCF-7 cells.
Apoptosis from 0.01 mmol/L for 48 h.
The percentages of cells in G0/G1-, S- and G2/Mphase did not differ from each other.
[74]
DATSCell viability by trypan blue assay
ROS by fluorescence microscopy and flow cytometry
Mitochondrial ROS levels assay
Immunoblot analysis
MDA-MB-231 10–100 μM for 16 h

10–80 μM for 1 h


10, 50 μM for 1 h

10–100 μM for 16 h
Apoptotic cell death in concentration- and time-dependent manner was observed with cell shrinkage and cytoplasmic membrane blebbing.

↑ ROS with activation of ASK1 and a downstream signal transduction JNK (C-Jun N-terminal kinase)-Bim pathway at 50–80 μM.
[75]
DASCell viability, cell cycle and apoptosis by PI staining by flow cytometric assay
DNA damage by Comet assay and DAPI staining
Flow cytometric assay for the production of Ca2+ and the level of mitochondrial membrane potential
Western blot of apoptotic associated proteins
Microarray assay
HeLa 25–100 µM for 24 h




75 µM for 0–2 h



75 µM for 0–72 h

5 μM DAS for 24 h
DNA damage and fragmentation.
Induced apoptosis and decreased the viability in concentration- and time-dependent manner.
Induced cell cycle arrest increasing G0/G1 cell population and decreasing G2/M and S cell population.
Decreased levels of mitochondrial membrane potential and promoted the levels of Ca2+
DAS promoted the levels of Fas, FasL and caspase-8, Bax, cytochrome c, Apaf-1, Bid, caspase-9 and -3.
28 genes were expressed at least by 2-fold compared with the untreated control cells.
[76]
DATSComet assay
Apoptosis and ROS by flow cytometry
Immunoblotting for phosphoolorect-ERK1/2 (P-ERK1/2
PC-3 cells transfected with the plasmid encoding p66ShcS36A or an empty pcDNA3.1 vector After 24 of transfection, cells were treated with DATS (0–40 µM)DATS increased p66Shc phosphorylation at serine 36, which was abolished by JNK inhibitor, and DATS-induced ROS formation was abolished in cells expressing p66ShcS36A variant. In cells expressing this variant, DATS-induced Akt dephosphorilation was reduced. The signaling pathway with P66Shc could be indispensable for DATS-induced prostate cancer cell death by modulating the Akt activity and ROS generation.[77]
DATSROS by flow cytometryProtein levels by ImmunoblottingLabile iron poolDNA damage by comet assay and microscopy PC-340 µM for 4 h
40 µM for 12 h
DATS-mediated increase in labile iron pool is regulated by JNK1 but not JNK2. Ferritin degradation in PC-3 cells treated with DATS is controlled by JNK1. DATS-induced increase in ROS formation is JNK1-dependent. Iron is not involved in DATS-induced cell death. DATS-induced DNA damage is not ameliorated by iron chelation.[78]
DATSCell viability by MTT assay
Apoptosis by DAPI staining
ROS and Mitochondrial membrane potential by flow cytometry
Caspase-9 and -3 activities
Apoptosis associated proteins by Western blotting
Primary colorectal cancer cells10–40 µM for 24 h

20 µM for 6,12 h


20 µM for 24 h
Viability inhibition in a concentration-dependent way.
Apoptosis induction. Nuclear shrinkage/condensation and nuclear fragmentation.
ROS production induction and decreased level of mitochondrial membrane potential.
Activation of caspase 9 and 3.
Increased protein levels of cytochrome c, caspase -9 and caspase-3.
[79]
DATS Cell survival by sulforhodamine B assay
ROS by flow cytometry
Protein level by immunoblotting
PC-3
PNT1A
40 µM for 24 hPNT1A cells are more resistant to cytotoxic effects than PC-3 cells. In these cells, reduction of induced p66Shc
hosphorylation and ferritin degradation, reduction Akt inactivation, and ROS generation was nearly abolished in PNT1A cells.
[80]
Allium sivasicum aqueous extractCytotoxicity by Trypan blue exclusion assay and MTT assay
Apoptosis by flow cytometry
MCF-7
MDA-MB-468
MDA-MD231
10–100 µg/mL, 48 h


MCF-7 21 ± 1.4 µg/mL
MDA-MB-468 22 ± 1.4 µg/mL
MDA-MB-231 24 ± 1.3 µg/mL
(24 h for all)
IC50(MCF-7) = 21 ± 1.4 µg/mL
IC50(MDA-MB-468) = 22 ± 1.4 µg/mL
IC50(MDA-MB-231) = 24 ± 1.3 µg/mL

↑ percentage of apoptosis
[81]
S-AllylmercaptocyteineCell proliferation by [3H] thymidine incorporation assay
DNA fragmentation assay
Free SH groups

Cell cycle by flow cytometry

Cytotoxicity by MTS assay
HEL
OCIM-1
0.02, 0.05, 0.1, 0.25 mM 24 h in HEL cells

0.05 or 0.1 mM for 2 days

0.1, 0.25, 0.5, 1 mM for 1,2, 3 days
0.25 mM for 6 h and 0.1 mM for 72 h
0.002–2 mM
Significant reduction in [3H] thymidine incorporation
Signs of DNA fragmentation
Initial increase of free SH groups followed by progressive decrease with extended incubation
Accumulation of cells in G2/M phase
OCIM-1 more sensitive. LD50 (HEL) = 0.1 mM and LD50 (OCIM-1) = 0.046 mM
[82]
AllicinCell proliferation by MTT assay
Apoptosis and cell cycle by flow cytometry
SGG-7901Not revealed
Apoptosis: 3 mg/l for 12, 24, 48 h
Cell cycle: 3, 6, 12 mg/L for 24 and 48 h
Growth inhibition in a concentration-dependent manner
Increased apoptosis
Cell cycle arrest in G2/M
[83]
DADSCell viability by MTT assay
Apoptosis by phase contrast microscopy and flow cytometry
ECA109
L02
10–60 µg/mL for 24 h
20–80 µg/mL for 24 h
Cell viability inhibition in a concentration-dependent manner in ECA109. Less toxic in L02
Membrane blebbing and formation of apoptotic bodies. Cellular shrinkage. Apoptosis induction in a concentration-dependent manner
[84]
DADSCell viability by MTT assay
Cell cycle and apoptosis by flow cytometry
PCR to investigate G2/M phase relative molecular pathway
Protein expression by Western blot
ECA109
L02
10–60 µg/mL for 24–72 h
20–60 µg/mL for 24 h
Cell viability Inhibition in a concentration-dependent manner.
Apoptosis induction in a concentration-dependent manner. G2/M phase arrest.
Upregulated levels of p21 and p53
Protein levels of caspase-3 and cleaved caspase-3 upregulated in a concentration-dependent way.
Induced apoptosis through upregulation of Bax mRNA, downregulation of Bcl-2 mRNA and a shift of Bax/Bcl-2 ratio.
Expression levels of MEK1 and ERK1/2 did not change, but p-MEK1 and p- ERK1/2 decreased
[85]
PTSOAmes test
MN test
MLA assay
comet assays (with and without Endo III and FPG enzymes)
Salmonella typhimurium strains L5178Ytk+/- Caco-25–100 µM for the different assays, depending on the viability of the cells (Trypan blue exclusion test)PTSO was not mutagenic in the Ames test, although it was weak mutagenic in the MLA assay after 24 of treatment (2.5–20.0 µM). The parent compound did not induce MN on mammalian cells, although in presence S9, induced positive results (20 µM). PTSO did not induce DNA breaks or oxidative damage in the comet assays.[17]
DPS, DPDS, and mixturesCell viability by PC, NR, MTS
ROS, GSH
Morphology study
Ames test
Caco-2 cells
S. typhimurium strains
0–200 µM for 2, 4, 8 hNo cytotoxicity or mutagenicity and no significant adverse effects were reported. ROS scavenger activity was observed for both compounds.[16]
AllicinCell viability by MTT assay
Apoptosis by Hoechst staining and flow cytometry
Expression levels of apoptosis-associated proteins by western blotting
MGC-803
BGC-823
SGC-7901
0.5–10 µg/mL for 48 h
1 µg/mL for 12, 24 and 48 h
0.01–10 µg/mL for 48 h
Cell viability is affected in a concentration and time-dependent manner.
Apoptosis induction
Enhanced expression levels of cleaved caspase 3
[86]
DAS, DADS, DATSCytotoxicity assay by cell counting kit-8Protein expression by western blottingCaspase-8 and 9 activityImmunofluorescence analysisLuciferase reporter assayRT-PCRBC3BCBL1HBL6BC2Ramos
DG75
1–50 µM for 24 hDAS and DADS slightly decreased viability
DAT:
IC50(BC3) = 13.7 ± 0.8
IC50(BCBL1) = 15.5 ± 1.0
IC50(HBL6) = 17.7 ± 0.6
IC50(BC2) = 14.6 ± 0.4
IC50(Ramos) = 43.4 ± 1.4
IC50(DG75) = 48.0 ± 0.9
Apoptosis by activation of caspases
Suppression

of NF-κB signaling
[87]
PTSAmes test
MN assay
MLA assay
comet assays (with and without Endo III and FPG enzymes)
S. typhimurium strains for Ames test; L5178Ytk+/− cells for MN and MLA assays; and Caco 2 cells for comet tests0–280 µM for the different assays, depending on the viability of the cells (total protein, NRU, MTS) Not mutagenic neither in the Ames test nor in MLA. Genotoxic effects were reported in the MN test at the highest concentration assayed (17.25 µM) without S9, and also its metabolites (+S9, from 20 µM).
↑ breaks damage on CaCO2 cells at the highest concentration tested (280 µM) but it did not induce oxidative DNA damage.
[55]
DATSCell viability by MTT assay
Cell cycle and apoptosis by flow cytometry
Protein expression by western blot
Nuclear morphological changes
ROS and MMP
AGS
Chang liver cells
0–50 µM for 0–24 h


50 µM, 0–24 h
Concentration- and time-dependent decrease of cell viability in AGS cells. No effect on Chang liver cells.
In AGS cells DATS induced G2/M arrest and apoptosis by blocking cell cycle into G1 phase, mitotic arrest, caspase-dependent apoptosis, and ROS-dependent AMPK activation
[88]
DATSCell viability by trypan blue exclusion assay
Clonogenic assay
ROS
Expression of DR4 and DR5 by flow cytometry
Immunocytochemistry
Apoptosis by flow cytometry
Immunoblotting.
U87MG
A172
U343
T98 G
25–50 µM for 30 min

25 µM 24 h





5–50 µM for 24 h and 25 µM for 0–24 h
Up-regulated DR5 receptor expression, and enhanced TRAIL-induced apoptosis through the downregulation of anti-apoptotic protein Mcl-1 and the upregulation of DR5 receptors through actions on the ROS-induced-p53[89]
Allicin Cytotoxicity by MTT assay


Cell proliferation and colony formation assays
Protein expression by western blot analysis
Gene expression by RT-qPCR
Caspase activity
Morphology study
Apoptosis by flow cytometry
U25115–90 µg/mL for 24 h.

5–90 µg/mL for 24, 48, 72 h.
30, 60 µg/mL for 48 h

30, 60 µg/mL

30, 60 µg/mL, 24 h

30, 60 µg/mL, 48 h
Cytotoxic effect in a concentration-dependent manner and nuclear morphology changes in U251 cells. IC50 = 41.97 µg allicin/mL for 24 h.
Increased apoptosis
Morphological changes of apoptotic cells (condensation of chromatin, nuclear fragmentation)
Proliferation inhibition
↑ caspase-3, -8 and -9 activities and Fas/FasL and Bax mRNA expression levels.
↓ Bcl-2 expression levels in a dose-dependent manner.
↑ the activation of both intrinsic and extrinsic apoptosis signaling pathways in U251 cells.
[90]
DASCell viability by MTT assay
The extend of lipid accumulation
ROS by flow cytometry
qRT-PCR of inflammatory genes
3T3L1
RAW 264.7
100 mM ethanol and treated with 50–500 µM DAS for 24 and 48 h.↑ viability in ethanol-exposed 3T3L1 cells treated with 200–500 µM for 24 h and 50–500 µM for 48 h.
↓ ROS production, reduces expression of pro-inflammatory cytokines, and enhance anti-inflammatory cytokine production in ethanol-exposed 3T3L1 cells treated with 50–100 µM for 24 or 48 h.
100 µM for 24h ↑ expression of M2 phenotype- specific genes in ethanol-exposed RAW 264.7 cells.
[91]
AllicinMTT assay
Cell cycle by flow cytometry
RT-PCR of cyclin D1, MMP-9 and RARβ
CD44+
CD117+ cells
CD44+: 4–32 µg allicin/mL or 8–125 µg ATRA/mL or 5 µg/mL of allicin during 4 h followed by 8–125 µg ATRA/mL. Total time of exposure 48 h.
CD117+: 0.5–24 µg allicin/mL or 4–64 µg ATRA/mL or 5 µg/mL of allicin during 4 h followed by 4–64 µg ATRA/mL. Total time of exposure 48 h.

IC50 for 48 h.
IC50 CD44+: allicin/ATRA (17.53 µg/mL) ˂ allicin (29.19 µg/mL) ˂ ATRA (37.43 µg/mL)
IC50 CD117+: ATRA (8.09 µg/mL) ˂ allicin (10.75 µg/mL) ˂ allicin/ATRA (13.65 µg/mL)

↑ of cells at the G2/M and G0/G1 phases in the CD44+ and CD117+ cells, respectively. The combination treatment caused the inhibition of CD44+ and CD117+ melanoma cells at the S phases compared to ATRA alone.

↑ cyclin D1 mRNA expression by all treatments and reduction of MMP-9 mRNA expression by allicin treatment both CD44+ and CD117+ cells.
↑ mRNA level of RARβ expression by allicin/ATRA treatment in CD117+ cells.
Increased MMP-9 gene expression by allicin/ATRA and ATRA treatments in CD44+ cells.
Allicin reinforces the ATRA-mediated inhibitory effects on CD44+ and CD117+ melanoma cells
[92]
DADSCell viability by trypan blue assay
SiRNA
Immunoblotting assay
Apoptosis by flow cytometry
DNA fragmentation assay
Caspase-3/7 activity assay
HCT116
DLD-1
HT29
SW620
FHC
5–100 µM for 24 h.

0–25 µM 20 h + 50 ng/mL TRAIL for 4 h.
0–10 µM caused ˂20% CRC cell deaths.


DADS + TRAIL produced concentration-dependent decreased of % survival in SW620 cells, but not in FHC cells.

0–10 µM did not alter the expression of pro-apoptotic proteins (Bax and Bid) or antiapoptotic proteins (XIAP and olorecta) and Bcl-2 were down-regulated in CRC cell lines.
[93]
Polymer films of Allium cepa L. Cell viability by MTT assayAmes testMN assayHepG2 GM-07492
S. typhimurium strains
Eluates from HTP-films and W-HTP films containing onion pulp were used at different concentrationsCytotoxicity: HTP > W-HTP.
No induction of MN was observed in both type of films, although the HTP films showed signs of mutagenicity in the Ames test.
[56]
Triploid onium Allium cornutum Clementi ex Visiani, 1842, and common onion Allium cepa L.Proliferation assay by MTS
DNA fragmentation assay
PCR of p53, Bax, Caspase 3
Hela, HCT116, and U2OS human cancer cell linesSerial dilutions of extracts from both Allium species (containing sulfides) were added to the 3 cell lines.Antiproliferative effects of both species were reported in the three cell lines. They induced apoptosis in HeLa cells.[94]
AllicinDetermination of LC50
DNA fragmentation assay
Schistosoma mansoniNot revealed
LC50 = 315 µL/L
No DNA fragmentation
[95]

3.4. Risk of Bias

Studies were considered to have a low, moderate or high risk of bias in terms of score out of 10. A moderate risk of bias was found in each of the 43 works chosen for the present systematic review. When reviewing the quality of selection, the studies show more limitations in “reproducibility” and “adequate statistical analysis” items. Full details are given in Table 3.
Table 3. Risk of bias for the methodological quality of studies reporting the toxicological evaluation in vitro of OSCs from Allium spp. 0: not reported; 1: not appropriately or clearly evaluated; 2: appropriately evaluated. M: medium (5–7); L: low (8–10); H: high (0–4).
Table 3. Risk of bias for the methodological quality of studies reporting the toxicological evaluation in vitro of OSCs from Allium spp. 0: not reported; 1: not appropriately or clearly evaluated; 2: appropriately evaluated. M: medium (5–7); L: low (8–10); H: high (0–4).
ReferenceClear ObjectiveWell Characterized ProductReproducibility of the AssayComparabilityAdequate Statistical AnalysisTotalRisk of BiasGeneral Risk of Bias
[57]2212073M
[58]2212073M
[59]1211273M
[60]2221182L
[61]2112282L
[62]2211282L
[63]2022282L
[64]22222100L
[65]2122291L
[66]2221073M
[67]2211282L
[68]22222100L
[69]1122064M
[70]2212055M
[71]2022282L
[72]2212291L
[75]1212282L
[73]22222100L
[74]2212291L
[76]2112064M
[77]2022282L
[78]2222191L
[79]2202282L
[80]2212291L
[81]2222082L
[82]2012273M
[83]2211282L
[84]1211273L
[85]2221291L
[16]22222100L
[17]22222100L
[86]2211282L
[87]2212291L
[88]22222100L
[89]2222082L
[55]22222100L
[91]2221291L
[92]2111282L
[90]22222100L
[93]2112064M
[56]2021273M
[94]22222100L
[95]2200046H

3.5. Limitations

The present systematic review was restricted by the databases used, the search conditions, and the recognized inclusion/exclusion principles chosen. However, the exploration strategy was quite comprehensive, so it is expected that relatively, only a few important studies could not be identified and considered. Only works reported in English were included, and this point could indicate bias in the source searching and selection process [96]. Finally, the lack of sufficient statistical information made impossible to combine the results of different studies into a meta-analysis section that it had to be divided by subheadings. It should provide a concise and precise description of the experimental results, their interpretation, as well as the experimental conclusions that can be drawn.

3.6. In Vivo Studies Excluded

Several in vivo studies (n = 26, Figure 1), despite being excluded by the criteria of this systematic review, have been analyzed by the authors. As in in vitro studies, many of them deal with the anticancer properties of natural organosulfur compounds, mainly assayed in mice. In this sense, Sundaram et al. [97] studied the growth inhibitory properties of DADS against colon cancer, Chu et al. [98] studied the compound S-Allylcysteine against prostate cancer and Nishikawa et al. [99] studied the inhibitory properties of ajoene against skin cancer. Other in vivo studies focused on measuring biological markers, such as catalase and monooxygenase activity, or their protective properties against toxic substances in animals treated with alliaceous compounds. In this sense, Zhang et al. [100] studied the protective effect of allicin against acrylamide. Only a few in vivo studies have focused on evaluating the toxicity of alliaceous compounds. Thus, acute studies [20] or subchronic toxicity studies of isolated substances (such as PTSO) [101] or extracts from plants of the Allium genus [102] were found. Among the in vivo toxicological studies, the genotoxicity tests (MN and comet) in rats are highlighted [103,104]. In general, no significant signs of toxicity neither genotoxic effect were observed in the subchronically studies or genotoxicity endpoints.

4. Discussion

The beneficial effects of OSCs compounds have been reviewed by different authors. In addition, their phytochemical profile has been well described [5,12]. However, as far as we know, the safety evaluation and toxicity effects of these compounds have not been reported.
The number of scientific publications dealing with the in vitro toxicity of OSCs that meet the criteria established in this review amounts to 43. In vitro studies play an important role in the toxicity evaluation of compounds. They can give valuable hints about mechanisms of toxicity, providing rapid and cost-effective screening, and allow one to reduce the use of live animal models in research.
Among the OSCs investigated, most studies have focused on DADS, DATS and DAS, followed by DATS, DAS, Allicin and Ajoene, whereas for others, the existing reports were limited (i.e., DPS, DPDS, PTSO, PTS) (see Figure 2). Moreover, most of the toxicity studies of OSCs are reported from 1990s and early 2000s, and only 14 of them have been published after 2015, so there are few current toxicological studies focusing on the toxicological effects of these compounds.
The most frequently used assays included cell viability determination, mechanisms of cell death (apoptosis), cell cycle analysis, oxidative stress biomarkers, mitochondria membrane potential (MMP), gene expression by PCR and protein expression by Western blotting. Cytotoxicity has been tested mainly by the MTT test [56,57]. Apoptosis has been investigated mainly by flow cytometry [65,93], but also by microscopic evaluation [63,66,70,80,84,86,93]. Moreover, expression of related genes and proteins (such as Bcl-2, Bax, p53, etc.) has also been explored [63,68,94] as well as caspases activity [68,74,80,87,93], but to a lesser extent.
Most of these studies aimed to evaluate the cytotoxicity of OSCs on different cellular models and tried to elucidate the mechanisms involved. Indeed, the vast majority of them explored the antiproliferative effects of OSCs to justify their potential as chemoprotectants against carcinogenesis (see Table 2). Several studies that reported the anti-cancer effect of black garlic on the cancer cell line showed inhibition of tumor activity by regulating metabolism [12]. Furthermore, DADS has been proposed as a therapeutic strategy for oxidative stress-injury in neurodegenerative diseases [65] and DAS has proved to be effective in reducing ethanol induced injury of cells (Kema et al., 2018). Apart from therapeutic aspects, there are a limited number of papers dealing with genotoxicity [16,17,55,56,60,62,95]. Finally, only 4 out of 43 studies have been performed specifically to explore the safety of OSCs for further agrifood applications [16,17,55,56]
Regarding results obtained for specific OSCs, DADS in relation to genotoxicity aspects has been reported to induce chromosome aberrations and sister chromatid exchanges in a Chinese hamster’s ovary cell line (CHO) [60]. Additionally, it increased the mutagenicity of 2-amino-1-methyl-6-phenylimidazol [4,5-b] pyridine when the Ames test was performed with S9 fraction from rats exposed to DADS. Filomeni et al., [64] and Kim et al. [65] explored the effects of DADS on two different cellular models of the nervous system and obtained different results. Whereas the first one suggested a pivotal role for oxidative stress in DADS-induced apoptosis on SH-SY5Y cells and pointed out a potential use as antiproliferative agent in cancer therapy, the second one observed opposite results on N18D3 cells, depending on the concentration used, with a protective effect at low concentrations. Recently, important toxic effects have been reported of this compound associated to high doses [105]. For these reasons, more DADS toxicity studies are necessary to guarantee its safe use as an anticancer agent.
Two other studies [70,75] investigated its effects on a breast cancer cell line (MCF-7) and obtained similar results, inhibition of cell proliferation and apoptosis induction, with Lei et al. [70] providing mechanistic clues (inhibition of ERK and activation of SAPK/JNK and p38 pathways). Apoptosis was also observed in other different cell types where the antiproliferative effects of DADS were investigated such as p53-wild type H460 and p53-null type H1299 non-small-cell lung cancer cells [63], in human glioblastoma cells [68], human leukemia cells [72], human esophageal carcinoma cells [84,85] or primary effusion lymphoma cells [87]. All these reports support the potential use of DADS as chemotherapeutic agent.
Results reported for DATS are similar to those discussed for DADS, as DATS have also shown to induce cytotoxicity, ROS production or apoptosis (also evidenced by changes in the expression of related genes and proteins) in different cell types such as NIH-3T3, MCF-7, PC-3, AGS, U87M6, PNT-1A, MDA-MB468, MDA-MD231, A172, U343 and T98G. Only Das et al. [68] and Shigemi et al. [87] evaluated both DADS and DATS, and compared the results obtained. Both of them observed similar results: DATS was more potent than DADS and DAS for induction of cell death with involvement of mitochondria and ROS production.
With respect to DAS, it induced genotoxic effects similarly to DADS [60,62], but it was less cytotoxic to CHO cells and it increased the activation of a higher number of mutagens. Again, several studies in a variety of cell lines showed its antiproliferative effect [59,63,68,71,77,87]). It has been reported that garlic compounds (DAS, DADS, DATS) do not require a p53-dependent pathway for mediation of apoptosis [68]. Moreover, its potential to reduce the tissue injury caused by ethanol was also demonstrated [92]. In order to compare the effects produced by each of these compounds, differences have been found, mainly due to the diverse experimental models and conditions used (concentration, time of exposure, biomarkers, etc.).
Studies evaluating allicin mainly reported cytotoxicity [57], and apoptosis [66,83,86,90] as adverse outcomes, mediated by different key events such as altered genes and protein expression or cell cycle changes. Scharfenberg et al. [57] were the only ones that studied not only allicin but also its decomposition product ajoene and observed that allicin was less toxic than ajoene in three different cell lines. This compound, ajoene, was also investigated by different authors [58,67,73] that observed cytotoxicity (with different cell lines showing different sensitivity) and apoptosis.
Regarding cytotoxicity assays, there are only a few cell lines listed in these studies that are included in the guidelines by OECD guidance for toxicological evaluation of chemicals. Most of them are cancer cell lines, and the effects reported in these findings correspond to a therapeutic anticancer effect and not to a cytotoxic evaluation. In this sense, more studies focused on the toxic effect of OSCs isolated are necessary.
Moreover, those OSCs with the fewer number of studies available in the scientific literature were also those mainly focused on safety issues in relation to their use in the agri-food sector. Thus, neither mutagenicity by the Ames test nor cytotoxicity in the human intestinal carcinoma Caco-2 cell line was observed for DPS and DPDS [16]. Additionally, a complete battery of genotoxicity tests were performed for PTSO [17] and PTS [55].
Genotoxicity assessment plays a key role in the safety evaluation required by EFSA guidelines for the submission of dossiers of different substances, such as food and feed additives, etc. [48,50], with the basic battery performed with in vitro tests. However, results of this review showed that only few assays have been carried out and some studies do not include the basic battery of tests required by the EFSA. In this regard and taking into account that specific OSCs that have been mainly investigated for their chemotherapeutic potential such as DADS, DATS, DAS, etc., show also interesting activities for their use in the food industry (antimicrobial, antioxidant or antifungal activities, among others, see Table 1), the thorough study of their genotoxicity would be worthy of research. Moreover, advanced in vitro models (i.e., 3D) could provide new data to support in vitro-in vivo data extrapolation for OSCs in general, and the testing of relevant concentrations used in the agri-food sector would allow to consider both efficacy and safety aspects. Thus, in vitro assays on their own can still provide valuable information to contribute to the commercial use of OSCs.

5. Conclusions

In general, there are very few in vitro studies focused on investigating the potential toxicity of OSCs. Most research studies aimed at evaluating only the cytotoxicity of OSCs on different cellular models to elucidate antiproliferative effects of these compounds and justify their potential as chemoprotective agent against carcinogenesis. This makes it difficult to assess the safety of the use of these compounds for a correct risk assessment. In addition, it limits the preliminary information needed to proceed with an in vivo toxicity assessment. Therefore, other cellular models such non-cancer cell lines should be included to ensure a correct in vitro toxicity evaluation of these compounds. Specifically, considering that genotoxicity assessment plays a key role in the safety evaluation required by EFSA; more genotoxicity studies of OSCs are necessary to guarantee consumer safety before their use as a potential natural additive in the food industry.

Author Contributions

Conceptualization, A.C.-L., A.B., E.G., A.J. and A.M.C.; methodology, A.C.-L. and P.A.-M.; formal analysis, A.C.-L., P.A.-M., A.I.P., A.J. and A.M.C., investigation, A.C.-L., P.A.-M., A.I.P., A.J. and A.M.C.; data curation, A.C.-L., P.A.-M., A.I.P., A.J. and A.M.C.; writing—original draft preparation, A.C.-L., P.A.-M., A.I.P., A.B., E.G., A.J. and A.M.C.; writing—review and editing, A.C.-L., A.I.P., A.B., E.G., A.J. and A.M.C.; supervision, A.J.; project administration, A.M.C.; funding acquisition, A.M.C. All authors have read and agreed to the published version of the manuscript.

Funding

This research was funded by the Consejería de Economía y Conocimiento of Junta de Andalucía (AT17_5323_USE and P18-TP-2147) and Antonio Cascajosa Lira thanks the Spanish Ministerio de Universidades for the funding FPU grant (FPU2019/01247).

Institutional Review Board Statement

Not applicable.

Informed Consent Statement

Not applicable.

Data Availability Statement

The data presented in this study is contained within the article.

Acknowledgments

The authors wish to thank the Consejería de Economía y Conocimiento of Junta de Andalucía (AT17_5323_USE and P18-TP-2147) for the financial support. Antonio Cascajosa Lira thanks the Spanish Ministerio de Universidades for the funding FPU grant (FPU2019/01247).

Conflicts of Interest

The authors declare no conflict of interest. The co-authors Alberto Baños and Enrique Guillamón affiliated to DMC Research Center declare that they have no conflict of interest.

Abbreviations

3T3-HER2: Mice fibroblast cell line; 3T3-L1: Mice fibroblast cell line; 5-Fu: 5-fluorouracil; A172: Human brain cell line; A375: Human melanoma cells; A549: Adenocarcinomic human alveolar basal epithelial cells; AGS: Human gastric carcinoma cell line; AIF: Apoptosis inducing factor; AMPK: AMP-activated protein kinase; ALP: Alkaline phosphatase activity; Asp: Aspartate; ATRA: All-trans retinoic acid; B16/BL6: Melanoma mouse cell line; B16F10: Murine melanoma cell; BC2: Human hepatoma cell line; BC3: Primary effusion lymphoma cell line; BCBL1: Human lymphoma cell line; BGC-823: Human gastric cancer cell line; BHK21: Baby hamster kidney-derived cell line; BJA-B: Burkitt lymphoma-derived cell line; Caco-2: Human colorectal adenocarcinoma cell line; CHO: Chinese hamster ovary cells; CRC: Human colon cancer cell line; CTX: Cyclophosphamide; Cyt c: Cytochrome c; DADS: Diallyl disulfide; DAS: Diallyl sulfide; DATS: Diallyl trisulfide; DLD-1: Colorectal adenocarcinoma cell line; DG75: Human lymphoma cell line; DPS: Dipropyl sulphide; DPDS: Dipropyl disulfide; ECA109: Human esophageal carcinoma cell line; ED50: amount of test compound required to reduce cell viability to 50%; ERK: Stress-activated protein kinase extracellular signal-regulated kinase; ERK-2: Extracellular regulatory kinase-2; EROD: Ethoxyresorufin O-deethylase; FHC: Normal human colon cell line; FS4: Human foreskin-derived cell line; GGPP: Geranylgeranylpyrophosphate; Glu: L-glutamate; GM-07492: Primary human skin fibroblasts; GSH: Reduced Glutathione; GSH-Px: Glutathione peroxidase; GSK-3: Glycogen syntethase kinase-3; GSSG: Oxidized glutathione; HBL6: Human lymphoma cell line; HEL: Human erythroleukemia cell line; HeLa: Cervical cancer cell line; HepG2: Human liver cancer cell line; HL-60: Human leukemia cell line; HT29: Human colorectal cell line; HTC116: Human colon cancer cell line; Http: Unwashed hydrothermally treated pulp; JNK/c-Jun: c-jun terminal kinase; M14: Human melanoma cell line; MAPKs; Mitogen-activated protein kinases; MCB: Minimal Bactericidal Concentration; MCF-7: Human breast cancer cell line; MDA: Malondialdehyde; MDA-MB-231: Human breast cell line; MDA-MB-468: Human breast cell line; MFC: Minimum fungicidal concentration; MIC: Minimal Inhibitory Concentration; MGC-803: Human gastric cancer cell line; MLA: Mouse lymphoma thymidine-kinase assay; MMP: Mitocondrial membrane potential; MN: Micronucleus; MROD: Methoxyresorufin O-demethylase; MTS: 3-(4,5-dimethylthiazol-2-yl)-5-(3-carboxymethoxyphenyl)-2-(4-sulfophenyl)-2H-tetrazolium, inner salt; NF-κB: Nuclear factor enhancing the kappa light chains of activated B cells; NIH-3T3: Mice fibroblasts cell line; NR: neutral red; L02: Human normal liver cell line; LDH: Lactate dehydrogenase activity; LEC1: Leuco-phytohemagglutinin resistant cell line; OCIM-1: Human leukemia cell line; p38: Mitogen-activated protein kinase; PAGE: Protein extracts and polyacrylamide gel electrophoresis; PANC-1: Human pancreatic carcinoma; PC: Protein content; PC-3: Human prostate adenocarcinoma cell line; PI3K/Akt: Phosphatidylinositol 3-kinase; PNPH: p-Nitrophenol hydroxylase; PROD: pentoxyresorufin O-dealkylase; PTS: Propyl-propane thiosulphinate; PTSO: Propyl-propane thiosulphonate; ROS: Reactive oxygen species; Ramos: Epstein-Barr virus-negative Burkitt lymphoma cell line; RAW 264.7: Mouse macrophage cell line; ROS: reactive oxygen species; RT-PCR: Quantitative realtime polymerase chain reaction; SAPK: stress-activated protein kinase; SCEs: sister chromatid exchanges; SGG-7901: Human gastric cancer cell line; SiRNA: small interfering RNA; SKBR-3: Human breast cancer cell line; SOD: Superoxide dismutase; SW620: Human colorectal cancer cell line; T98G: Glioblastoma cell line; TRAIL: Tumor necrosis factor-related apoptosis-induced ligand; U251: Human glioma cell line; U2OS: Human osteosarcoma cell line; U343: Human glioblastoma cell line; U87MG: Human glioblastoma cell line http:HTP: Washed hydrothermally treated pulp.

References

  1. Poojary, M.M.; Putnik, P.; Bursać Kovačević, D.; Barba, F.J.; Lorenzo, J.M.; Dias, D.A.; Shpigelman, A. Stability and extraction of bioactive sulfur compounds from Allium genus processed by traditional and innovative technologies. J. Food Compos. Anal. 2017, 61, 28–39. [Google Scholar] [CrossRef]
  2. Putnik, P.; Gabrić, D.; Roohinejad, S.; Barba, F.J.; Granato, D.; Mallikarjunan, K.; Lorenzo, J.M.; Bursać Kovačević, D. An overview of organosulfur compounds from Allium spp.: From processing and preservation to evaluation of their bioavailability, antimicrobial, and anti-inflammatory properties. Food Chem. 2019, 276, 680–691. [Google Scholar] [CrossRef] [PubMed]
  3. Nicastro, H.L.; Ross, S.A.; Milner, J.A. Garlic and onions: Their cancer prevention properties. Cancer Prev. Res. 2015, 8, 181–189. [Google Scholar] [CrossRef] [PubMed]
  4. Ramirez, D.A.; Locatelli, D.A.; González, R.E.; Cavagnaro, P.F.; Camargo, A.B. Analytical methods for bioactive sulfur compounds in Allium: An integrated review and future directions. J. Food Compos. Anal. 2017, 61, 4–19. [Google Scholar] [CrossRef]
  5. Ariza, J.J.; García-López, D.; Sánchez-Nieto, E.; Guillamón, E.; Baños, A.; Martínez-Bueno, M. Antilisterial effect of a natural formulation based on citrus extract in ready-to-eat foods. Foods 2021, 10, 1475. [Google Scholar] [CrossRef]
  6. Deka, B.; Manna, P.; Borah, J.C.; Talukdar, N.C. A review on phytochemical, pharmacological attributes and therapeutic uses of Allium hookeri. Phytomed. Plus 2022, 2, 100262. [Google Scholar] [CrossRef]
  7. Guillamón, E.; Andreo-Martínez, P.; Mut-Salud, N.; Fonollá, J.; Baños, A. Beneficial effects of organosulfur compounds from Allium cepa on gut health: A systematic review. Foods 2021, 10, 1680. [Google Scholar] [CrossRef] [PubMed]
  8. Rabelo-Ruiz, M.; Teso-Pérez, C.; Peralta-Sánchez, J.M.; Ariza, J.J.; Martín-Platero, A.M.; Casabuena-Rincón, Ó.; Vázquez-Chas, P.; Guillamón, E.; Aguinaga-Casañas, M.A.; Maqueda, M.; et al. Allium extract implements weaned piglet’s productive parameters by modulating distal gut microbiota. Antibiotics 2021, 10, 269. [Google Scholar] [CrossRef]
  9. Rabelo-Ruiz, M.; Ariza-Romero, J.J.; Zurita-González, M.J.; Martín-Platero, A.M.; Baños, A.; Maqueda, M.; Valdivia, E.; Martínez-Bueno, M.; Peralta-Sánchez, J.M. Allium-based phytobiotic enhances egg production in laying hens through microbial composition changes in ileum and cecum. Animals 2021, 11, 448. [Google Scholar] [CrossRef]
  10. Sharma, P.; Hajam, Y.A.; Kumar, R.; Rai, S. Complementary and alternative medicine for the treatment of diabetes and associated complications: A review on therapeutic role of polyphenols. Phytomed. Plus 2022, 2, 100188. [Google Scholar] [CrossRef]
  11. Sorlozano-Puerto, A.; Albertuz-Crespo, M.; Lopez-Machado, I.; Ariza-Romero, J.J.; Baños-Arjona, A.; Exposito-Ruiz, M.; Gutierrez-Fernandez, J. In vitro antibacterial activity of propyl-propane-thiosulfinate and propyl-propane-thiosulfonate derived from Allium spp. against gram-negative and gram-positive multidrug-resistant bacteria isolated from human samples. Biomed Res. Int. 2018, 2018, 7861207. [Google Scholar] [CrossRef] [PubMed]
  12. Vinayagam, R.; Lee, K.E.; Ambati, R.R.; Gundamaraju, R.; Ramadan, M.F.; Kang, S.G. Recent development in black garlic: Nutraceutical applications and health-promoting phytoconstituents. Food Rev. Int. 2021, 1–22. [Google Scholar] [CrossRef]
  13. Rochetti, G.; Chang, L.; Bocchi, S.; Giuberti, G.; Ak, G.; Elbasan, F.; Yildiztugay, E.; Ceylam, R.; Caree Nancy Picot-Allain, M.; Fawzi Mahomoodally, M.; et al. The functional potential of nine Allium species related to their untargeted phytochemical characterization, antioxidant capacity and enzyme inhibitory ability. Food Chem. 2022, 368, 130782. [Google Scholar] [CrossRef] [PubMed]
  14. Teshika, J.D.; Zakariyyah, A.M.; Zaynab, T.; Zengin, G.; Rengasamy, K.R.; Pandian, S.K.; Fawzi, M.M. Traditional and modern uses of onion bulb (Allium cepa L.): A systematic review. Crit. Rev. Food Sci. Nutr. 2019, 59, S39–S70. [Google Scholar] [CrossRef] [PubMed]
  15. Ribeiro-Santos, R.; Andrade, M.; Melo, N.R.; de Sanches-Silva, A. Use of essential oils in active food packaging: Recent advances and future trends. Trends Food Sci. Technol. 2017, 61, 132–140. [Google Scholar] [CrossRef]
  16. Llana-Ruiz-Cabello, M.; Maisanaba, S.; Gutiérrez-Praena, D.; Prieto, A.I.; Pichardo, S.; Jos, Á.; Moreno, F.J.; Cameán, A.M. Cytotoxic and mutagenic in vitro assessment of two organosulfur compounds derived from onion to be used in the food industry. Food Chem. 2015, 166, 423–431. [Google Scholar] [CrossRef]
  17. Mellado-García, P.; Maisanaba, S.; Puerto, M.; Llana-Ruiz-Cabello, M.; Prieto, A.I.; Marcos, R.; Pichardo, S.; Cameán, A.M. Genotoxicity assessment of propyl thiosulfinate oxide, an organosulfur compound from Allium extract, intended to food active packaging. Food Chem. Toxicol. 2015, 86, 365–373. [Google Scholar] [CrossRef]
  18. Mylona, K.; Garcia-Cela, E.; Sulyok, M.; Medina, A.; Naresh, M. Influence of Two Garlic-Derived Compounds, Propyl Propane Thiosulfonate (PTS) and Propyl Propane Thiosulfinate (PTSO), on Growth and Mycotoxin Production by Fusarium Species In Vitro and in Stored Cereals. Toxins 2019, 11, 495. [Google Scholar] [CrossRef] [PubMed]
  19. Llana-Ruíz-Cabello, M.; Pichardo, S.; Jiménez-Morillo, N.T.; Abad, P.; Guillamón, E.; González-Vila, F.J.; Cameán, A.M.; González-Pérez, J.A. Characterisation of a bio-based packaging containing a natural additive from Allium spp. using analytical pyrolysis and carbon stable isotopes. J. Anal. Appl. Pyrolysis. 2016, 120, 334–340. [Google Scholar] [CrossRef]
  20. Llana-Ruiz-Cabello, M.; Gutiérrez-Praena, D.; Puerto, M.; Pichardo, S.; Moreno, F.J.; Baños, A.; Nuñez, C.; Guillamón, E.; Cameán, A.M. Acute toxicological studies of the main organosulfur compound derived from Allium sp. intended to be used in active food packaging. Food Chem. Toxicol. 2015, 82, 1–11. [Google Scholar] [CrossRef]
  21. Yoshida, H.; Iwata, N.; Katsuzaki, H.; Naganawa, R.; Ishikawa, K.; Fukuda, H.; Fujino, T.; Suzuki, A. Antimicrobial activity of a compound isolated from an oil-macerated garlic extract. Biosci. Biotechnol. Biochem. 1998, 62, 1014–1017. [Google Scholar] [CrossRef] [PubMed]
  22. Cho, S.; Ryu, J.; Surh, Y. Ajoene, a Major Organosulfide Found in Crushed Garlic, Induces NAD(P)H:quinone Oxidoreductase Expression Through Nuclear Factor E2-related Factor-2 Activation in Human Breast Epithelial Cells. J. Cancer Prev. 2019, 2, 112. [Google Scholar] [CrossRef] [PubMed]
  23. Chung, L.Y. The antioxidant properties of garlic compounds: Alyl cysteine, alliin, allicin, and allyl disulfide. J. Med. Food. 2006, 9, 205–213. [Google Scholar] [CrossRef] [PubMed]
  24. Bachrach, G.; Jamil, A.; Naor, R.; Tal, G.; Ludmer, Z.; Steinberg, D. Garlic allicin as a potential agent for controlling oral pathogens. J. Med. Food. 2014, 14, 1338–1343. [Google Scholar] [CrossRef] [PubMed]
  25. O’Gara, E.A.; Hill, D.J.; Maslin, D.J. Activities of garlic oil, garlic powder, and their diallyl constituents against Helicobacter pylori. Appl. Env. Microbiol. 2000, 66, 2269–2273. [Google Scholar] [CrossRef] [PubMed]
  26. Wallock-Richards, D.; Doherty, C.J.; Doherty, L.; Clarke, D.J.; Place, M.; Govan, J.R.W.; Campopiano, D.J. Garlic revisited: Antimicrobial activity of allicin-containing garlic extracts against Burkholderia cepacia complex. PLoS ONE 2014, 9, e112726. [Google Scholar] [CrossRef]
  27. Yamada, Y.; Azuma, K. Evaluation of the in vitro antifungal activity of allicin. Antimicrob. Agents Chemother. 1977, 11, 743–749. [Google Scholar] [CrossRef]
  28. Gong-chen, W.; Lu-lu, H.; Jing, W.; Wan-nan, L.; Chuan-yi, P.; Yan-fei, L. Effects of Allicin on Lipid Metabolism and Antioxidant Activity in Chickens. J. Northeast. Agric. Univ. 2014, 21, 46–49. [Google Scholar] [CrossRef]
  29. Li, F.; Li, Q.; Wu, S.; Tan, Z. Salting-out extraction of allicin from garlic (Allium sativum L.) based on ethanol/ammonium sulfate in laboratory and pilot scale. Food Chem. 2017, 217, 91–97. [Google Scholar] [CrossRef] [PubMed]
  30. Tsao, S.M.; Yin, M.C. In-vitro antimicrobial activity of four diallyl sulphides occurring naturally in garlic and Chinese leek oils. J. Med. Microbiol. 2001, 50, 646–649. [Google Scholar] [CrossRef] [Green Version]
  31. Feng, C.; Luo, Y.; Nian, Y.; Liu, D.; Yin, X.; Wu, J.; Di, J.; Zhang, R.; Zhang, J. Diallyl Disulfide Suppresses the Inflammation and Apoptosis Resistance Induced by DCA through ROS and the NF-κB Signaling Pathway in Human Barrett’s Epithelial Cells. Inflammation 2017, 40, 818–831. [Google Scholar] [CrossRef] [PubMed]
  32. Mathan Kumar, M.; Tamizhselvi, R. Protective effect of diallyl disulfide against cerulein-induced acute pancreatitis and associated lung injury in mice. Int. Immunopharmacol. 2000, 80, 106136. [Google Scholar] [CrossRef]
  33. Kalayarasan, S.; Prabhu, P.N.; Sriram, N.; Manikandan, R.; Arumugam, M.; Sudhandiran, G. Diallyl sulfide enhances antioxidants and inhibits inflammation through the activation of Nrf2 against gentamicin-induced nephrotoxicity in Wistar rats. Eur. J. Pharmacol. 2009, 606, 162–171. [Google Scholar] [CrossRef]
  34. Tsai, C.Y.; Wang, C.C.; Lai, T.Y.; Tsu, H.N.; Wang, C.H.; Liang, H.Y.; Kuo, W.W. Antioxidant effects of diallyl trisulfide on high glucose-induced apoptosis are mediated by the PI3K/Akt-dependent activation of Nrf2 in cardiomyocytes. Int. J. Cardiol. 2013, 168, 1286–1297. [Google Scholar] [CrossRef] [PubMed]
  35. Peinado, M.J.; Ruiz, R.; Echávarri, A.; Aranda-Olmedo, I.; Rubio, L.A. Garlic derivative PTS-O modulates intestinal microbiota composition and improves digestibility in growing broiler chickens. Anim. Feed Sci. Technol. 2013, 181, 87–92. [Google Scholar] [CrossRef]
  36. Peinado, M.J.; Ruiz, R.; Echávarri, A.; Rubio, L.A. Garlic derivative propyl propane thiosulfonate is effective against broiler enteropathogens in vivo. Poult. Sci. 2012, 91, 2148–2157. [Google Scholar] [CrossRef]
  37. Falcón-Piñeiro, A.; Remesal, E.; Noguera, M.; Ariza, J.J.; Guillamón, E.; Baños, A.; Navas-Cortes, J.A. Antifungal activity of propyl-propane-thiosulfinate (PTS) and propyl-propane-thiosulfonate (PTSO) from Allium cepa against Verticillium dahliae: In vitro and in planta assays. J. Fungi 2021, 7, 736. [Google Scholar] [CrossRef] [PubMed]
  38. Bravo, D.; Lillehoj, H. Use of At Least One Dialkyl Thosulfonate or Thosulfinate for Reducing the Number of Apcomplexan an Animal. U.S. Patent 2013/0079402 A1, 28 March 2013. Available online: https://patentimages.storage.googleapis.com/5b/df/ca/82319693e99358/US20130079402A1.pdf (accessed on 19 July 2022).
  39. Nuñez Lechado, C.; Baños Arjona, A.; Guillamon Ayala, E.; Valero López, A.; Navarro Moll, M.C.; Sanz, A. Use of Propyl Propanethosulfinate and Propyl Propanethosulfonate for the Prevention and Reduction of Parasites in Aquatic Animals. U.S. Patent 9,271,947 B2, 1 March 2016. Available online: https://patentimages.storage.googleapis.com/db/7d/6d/a1e24b67f01c40/US9271947.pdf (accessed on 19 July 2022).
  40. Sorlozano-Puerto, A.; Albertuz-Crespo, M.; Lopez-Machado, I.; Gil-Martinez, L.; Ariza-Romero, J.J.; Maroto-Tello, A.; Baños-Arjona, A.; Gutierrez-Fernandez, J. Antibacterial and antifungal activity of propyl-propane-thiosulfinate and propyl-propane-thiosulfonate, two organosulfur compounds from Allium cepa: In vitro antimicrobial effect via the gas phase. Pharmaceuticals 2021, 14, 21. [Google Scholar] [CrossRef] [PubMed]
  41. Maldonado, P.D.; Barrera, D.; Rivero, I.; Mata, R.; Medina-Campos, O.N.; Hernández-Pando, R.; Pedraza-Chaverrí, J. Antioxidant S-allylcysteine prevents gentamicin-induced oxidative stress and renal damage. Free Radic. Biol. Med. 2003, 35, 317–324. [Google Scholar] [CrossRef]
  42. Higuchi, O.; Tateshita, K.; Nishimura, H. Antioxidative Activity of Sulfur-Containing Compounds in Allium Species for Human Low-Density Lipoprotein (LDL) Oxidation in vitro. J. Agric. Food Chem. 2003, 51, 7208–7214. [Google Scholar] [CrossRef]
  43. Kahl, R.; Kappus, H. Toxikologie der synthetischen Antioxidantien BHA und BHT im Vergleich mit dem natürlichen Antioxidans Vitamin E. Z. Lebensm. Unters. Forsch. 1993, 196, 329–338. [Google Scholar] [CrossRef]
  44. Baños, A.; García, J.D.; Núñez, C.; Mut-Salud, N.; Ananou, S.; Martínez-Bueno, M.; Maqueda, M.; Valdivia, E. Subchronic toxicity study in BALBc mice of enterocin AS-48, an anti-microbial peptide produced by Enterococcus faecalis UGRA10. Food Chem. Toxicol. 2019, 132, 110667. [Google Scholar] [CrossRef] [PubMed]
  45. Maisanaba, S.; Llana-Ruiz-Cabello, M.; Gutiérrez-Praena, D.; Pichardo, S.; Puerto, M.; Prieto, A.I.; Jos, A.; Cameán, A.M. New advances in active packaging incorporated with essential oils or their main components for food preservation. Food Rev. Int. 2017, 33, 447–515. [Google Scholar] [CrossRef]
  46. Llana-Ruiz-Cabello, M.; Pichardo, S.; Bermudez, J.M.; Baños, A.; Ariza, J.J.; Guillamón, E.; Aucejo, S.; Cameán, A.M. Characterisation and antimicrobial activity of active polypropylene films containing oregano essential oil and Allium extract to be used in packaging for meat products. Food Addit. Contam.-Part A Chem. Anal. Control. Expo. Risk Assess. 2018, 35, 782–791. [Google Scholar] [CrossRef] [PubMed]
  47. Seydim, A.C.; Sarikus, G. Antimicrobial activity of whey protein based edible films incorporated with oregano, rosemary and garlic essential oils. Food Res. Int. 2006, 39, 639–644. [Google Scholar] [CrossRef]
  48. EFSA Panel on Food Additives and Nutrient Sources added to Food (ANS). Guidance for submission for food additive evaluations. EFSA J. 2012, 10, 2760. Available online: https://www.efsa.europa.eu/en/efsajournal/pub/2760 (accessed on 3 January 2022).
  49. EFSA CEF Panel (EFSA Panel on Food Contact Materials, Enzymes, Flavourings and Processing Aids). Scientific opinion on recent developments in the risk assessment of chemicals in food and their potential impact on the safety assessment of sub-stances used in food contact materials. EFSA J. 2016, 14, 4357. Available online: https://www.efsa.europa.eu/en/efsajournal/pub/4357 (accessed on 3 January 2022).
  50. EFSA FEEDAP Panel (EFSA Panel on Products or Substances used in Animal Feed). Guidance on the assessment of the safety of feed additives for the consumer. EFSA J. 2017, 15, e05022. Available online: https://www.efsa.europa.eu/en/efsajournal/pub/5022 (accessed on 3 January 2022).
  51. EFSA Scientific Committee. Scientific Opinion on the clarification of some aspects related to genotoxicity assessment. EFSA J. 2017, 15, 5113. Available online: https://www.efsa.europa.eu/en/efsajournal/pub/5113 (accessed on 3 January 2022).
  52. Page, M.J.; McKenzie, J.E.; Bossuyt, P.M.; Boutron, I.; Hoffmann, T.C.; Mulrow, C.D.; Shamseer, L.; Tetzlaff, J.M.; Akl, E.A.; Brennan, S.E.; et al. The PRISMA 2020 statement: An updated guideline for reporting systematic reviews. Int. J. Surg. 2021, 88, 89. [Google Scholar] [CrossRef] [PubMed]
  53. Higgins, J.P.T.; Thomas, J.; Chandler, J.; Cumpston, M.; Li, T.; Page, M.J.; Welch, V.A. Cochrane Handbook for Systematic Reviews of Interventions; John Wiley & Sons: Hoboken, NJ, USA, 2019. [Google Scholar] [CrossRef]
  54. Viswanathan, M.; Patnode, C.D.; Berkman, N.D.; Bass, E.B.; Chang, S.; Hartling, L.; Murad, M.H.; Treadwell, J.R.; Kane, R.L. Recommendations for assessing the risk of bias in systematic reviews of health-care interventions. J. Clin. Epidemiol. 2018, 97, 26–34. [Google Scholar] [CrossRef] [PubMed]
  55. Mellado-García, P.; Maisanaba, S.; Puerto, M.; Prieto, A.I.; Marcos, R.; Pichardo, S.; Cameán, A.M. In vitro toxicological assessment of an organosulfur compound from Allium extract: Cytotoxicity, mutagenicity and genotoxicity studies. Food Chem. Toxicol. 2017, 99, 231–240. [Google Scholar] [CrossRef] [PubMed]
  56. Rodrigues-Barreto, M.; Aleixo, N.A.; Silvestre, R.B.; Fregonezi, N.F.; Barud, H.D.S.; Dias, D.D.S.; Ribeiro, C.A.; Resende, F.A. Genotoxicological safety assessment of puree-only edible films from onion bulb (Allium cepa L.) for use in food packaging-related applications. J. Food Sci. 2019, 85, 201–208. [Google Scholar] [CrossRef]
  57. Scharfenberg, K.; Wagner, R.; Wagner, K.G. The cytotoxic effect of ajoene, a natural product from garlic, investigated with different cell lines. Cancer Lett. 1990, 53, 103–108. [Google Scholar] [CrossRef]
  58. Scharfenberg, K.; Ryll, T.; Wagner, R.; Wagner, K.G. Injuries to cultivated BJA-B cells by ajoene, a garlic-derived natural compound: Cell viability, glutathione metabolism, and pools of acidic amino acids. J. Cell. Physiol. 1994, 158, 55–60. [Google Scholar] [CrossRef]
  59. Musk, S.R.R.; Smith, T.K.; Stening, P.; Fyfe, D.; Johnson, I.T.; Stephenson, P. Selective Toxicity of Compounds Naturally Present in Food Toward the Transformed Phenotype of Human Colorectal Cell Line HT29. Nutr. Cancer 1990, 24, 289–298. [Google Scholar] [CrossRef]
  60. Musk, S.R.R.; Clapham, P.; Johnson, I.T. Cytotoxicity and Genotoxicity of Diallyl Sulfide and Diallyl Disulfide Towards Chinese Hamster Ovary Cells. Food Chem. Toxicol. 1997, 35, 379–385. [Google Scholar] [CrossRef]
  61. Sigounas, G.; Hooker, J.L.; Li, W.; Anagnostou, A.; Steiner, M. S-allylmercaptocysteine, a stable thioallyl compound, induces apoptosis in erythroleukemia cell lines. Nutr. Cancer 1997, 28, 153–159. [Google Scholar] [CrossRef]
  62. Guyonnet, D.; Belloir, C.; Suschetet, M.; Siess, M.H.; Le Bon, A.M. Liver subcellular fractions from rats treated by organosulfur compounds from Allium modulate mutagen activation. Mutat. Res.-Genet. Toxicol. Env. Mutagen. 2000, 466, 17–26. [Google Scholar] [CrossRef]
  63. Hong, Y.S.; Ham, Y.A.; Choi, J.H.; Kim, J. Effects of allyl sulfur compounds and garlic extract on the expressions of Bcl-2, Bax, and p53 in non small cell lung cancer cell lines. Exp. Mol. Med. 2000, 32, 127–134. [Google Scholar] [CrossRef] [PubMed]
  64. Filomeni, G.; Aquilano, K.; Rotilio, G.; Ciriolo, M.R. Reactive oxygen species-dependent c-Jun NH2-terminal kinase/c-Jun signaling cascade mediates neuroblastoma cell death induced by diallyl disulfide. Cancer Res. 2003, 63, 5940–5949. [Google Scholar] [PubMed]
  65. Kim, J.G.; Koh, S.H.; Lee, Y.J.; Lee, K.Y.; Kim, Y.; Kim, S.; Lee, M.K.; Kim, S.H. Differential effects of diallyl disulfide on neuronal cells depend on its concentration. Toxicology 2005, 211, 86–96. [Google Scholar] [CrossRef] [PubMed]
  66. Park, S.Y.; Cho, S.J.; Kwon, H.; Lee, K.R.; Rhee, D.K.; Pyo, S. Corrigendum to “Caspase-independent cell death by allicin in human epithelial carcinoma cells: Involvement of PKA”. Cancer Lett. 2005, 224, 123–132. [Google Scholar] [CrossRef]
  67. Taylor, P.; Noriega, R.; Farah, C.; Abad, M.J.; Arsenak, M.; Apitz, R. Ajoene inhibits both primary tumor growth and metastasis of B16/BL6 melanoma cells in C57BL/6 mice. Cancer Lett. 2006, 239, 298–304. [Google Scholar] [CrossRef] [PubMed]
  68. Das, A.; Banik, N.L.; Ray, S.K. Garlic compounds generate reactive oxygen species leading to activation of stress kinases and cysteine proteases for apoptosis in human glioblastoma T98G and U87MG cells. Cancer 2007, 110, 1083–1095. [Google Scholar] [CrossRef] [PubMed]
  69. Prager-Khoutorsky, M.; Goncharov, I.; Rabinkov, A.; Mirelman, D.; Geiger, B.; Bershadsky, A.D. Allicin inhibits cell polarization, migration and division via its direct effect on microtubules. Cell Motil. Cytoskeleton. 2007, 64, 321–337. [Google Scholar] [CrossRef] [PubMed]
  70. Lei, X.Y.; Yao, S.Q.; Zu, X.Y.; Huang, Z.X.; Liu, L.J.; Zhong, M.; Zhu, B.Y.; Tang, S.S.; Liao, D.F. Apoptosis induced by diallyl disulfide in human breast cancer cell line MCF-7. Acta Pharmacol. Sin. 2008, 29, 1233–1239. [Google Scholar] [CrossRef]
  71. Sriram, N.; Kalayarasan, S.; Ashokkumar, P.; Sureshkumar, A.; Sudhandiran, G. Diallyl sulfide induces apoptosis in Colo 320 DM human colon cancer cells: Involvement of caspase-3, NF-κB, and ERK-2. Mol. Cell. Biochem. 2008, 311, 157–165. [Google Scholar] [CrossRef]
  72. Tan, H.; Ling, H.; He, J.; Yi, L.; Zhou, J.; Lin, M.; Su, Q. Inhibition of ERK and activation of p38 are involved in diallyl disulfide induced apoptosis of leukemia HL-60 cells. Arch. Pharm. Res. 2008, 31, 786–793. [Google Scholar] [CrossRef]
  73. Ledezma, E.; Wittig, O.; Alonso, J.; Cardier, J.E. Potentiated cytotoxic effects of statins and ajoene in murine melanoma cells. Melanoma Res. 2009, 19, 69–74. [Google Scholar] [CrossRef]
  74. Zhou, C.; Mao, X.P.; Guo, Q.; Zeng, F.Q. Diallyl trisulphide-induced apoptosis in human melanoma cells involves downregulation of Bcl-2 and Bcl-xL expression and activation of caspases. Clin. Exp. Dermatol. 2009, 34, 537–543. [Google Scholar] [CrossRef] [PubMed]
  75. Jun, Z.; Suzuki, M.; Xiao, J.; Wen, J.; Talbot, S.G.; Li, G.C.; Xu, M. Comparative effects of natural and synthetic diallyl disulfide on apoptosis of human breast-cancer MCF-7 cells. Biotechnol. Appl. Biochem. 2009, 52, 113. [Google Scholar] [CrossRef] [PubMed]
  76. Lee, B.-C.; Park, B.-H.; Kim, S.-Y.; Lee, Y.J. Role of Bim in diallyl trisulfide-induced cytotoxicity in human cancer cells. J. Cell. Biochem. 2011, 112, 118–127. [Google Scholar] [CrossRef] [Green Version]
  77. Wu, P.P.; Chung, H.W.; Liu, K.C.; Wu, R.S.C.; Yang, J.S.; Tang, N.Y.; Lo, C.; Hsia, T.E.C.; Yu, C.C.; Chueh, F.U.S.; et al. Diallyl sulfide induces cell cycle arrest and apoptosis in HeLa human cervical cancer cells through the p53, caspase- and mitochondria-dependent pathways. Int. J. Oncol. 2011, 38, 1605–1613. [Google Scholar] [CrossRef] [PubMed]
  78. Borkowska, A.; Sielicka-Dudzin, A.; Herman-Antosiewicz, A.; Wozniak, M.; Fedeli, D.; Falcioni, G.; Antosiewicz, J. Diallyl trisulfide-induced prostate cancer cell death is associated with Akt/PKB dephosphorylation mediated by P-p66shc. Eur. J. Nutr. 2012, 51, 817–825. [Google Scholar] [CrossRef]
  79. Sielicka-Dudzin, A.; Borkowska, A.; Herman-Antosiewicz, A.; Wozniak, M.; Jozwik, A.; Fedeli, D.; Antosiewicz, J. Impact of JNK1, JNK2, and ligase Itch on reactive oxygen species formation and survival of prostate cancer cells treated with diallyl trisulfide. Eur. J. Nutr. 2012, 51, 573–581. [Google Scholar] [CrossRef] [PubMed]
  80. Yu, C.S.; Huang, A.C.; Lai, K.C.; Huang, Y.P.; Lin, M.W.; Yang, J.S.; Chung, J.G. Diallyl trisulfide induces apoptosis in human primary colorectal cancer cells. Oncol. Rep. 2012, 28, 949–954. [Google Scholar] [CrossRef]
  81. Borkowska, A.; Knap, N.; Antosiewicz, J. Diallyl trisulfide is more cytotoxic to prostate cancer cells PC-3 than to noncancerous epithelial cell line PNT1A: A possible role of p66Shc signaling axis. Nutr. Cancer 2013, 65, 711–717. [Google Scholar] [CrossRef]
  82. Tepe, B.; Tuncer, E.; Saraydän, S.U.; Özer, H.; Şen, M.; Karadayi, K.; Inan, D.S.; Karadayi, S.; Polat, Z.; Akpulat, A.; et al. Antitumoral effects of Allium sivasicum on breast cancer in vitro and in vivo. Mol. Biol. Rep. 2013, 40, 597–604. [Google Scholar] [CrossRef]
  83. Tao, M.; Gao, L.; Pan, J.; Wang, X. Study on the inhibitory effect of allicin on human gastric cancer cell line SGC-7901 and its mechanism. Afr. J. Tradit. Complement. Altern. Med. 2014, 11, 176–179. [Google Scholar] [CrossRef] [PubMed]
  84. Yin, X.; Zhang, J.; Li, X.; Liu, D.; Feng, C.; Liang, R.; Zhuang, K.; Cai, C.; Xue, X.; Jing, F.; et al. DADS suppresses human esophageal xenograft tumors through RAF/MEK/ERK and mitochondria-dependent pathways. Int. J. Mol. Sci. 2014, 15, 12422–12441. [Google Scholar] [CrossRef] [PubMed]
  85. Yin, X.; Zhang, R.; Feng, C.; Zhang, J.; Liu, D.; Xu, K.; Wang, X.; Zhang, S.; Li, Z.; Liu, X.; et al. Diallyl disulfide induces G2/M arrest and promotes apoptosis through the p53/p21 and MEK-ERK pathways in human esophageal squamous cell carcinoma. Oncol. Rep. 2014, 32, 1748–1756. [Google Scholar] [CrossRef] [PubMed]
  86. Zhang, X.; Zhu, Y.; Duan, W.; Feng, C.; He, X. Allicin induces apoptosis of the MGC-803 human gastric carcinoma cell line through the p38 mitogen-activated protein kinase/caspase-3 signaling pathway. Mol. Med. Rep. 2015, 11, 2755–2760. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  87. Shigemi, Z.; Furukawa, Y.; Hosokawa, K.; Minami, S.; Matsuhiro, J.; Nakata, S.; Watanabe, T.; Kagawa, H.; Nakagawa, K.; Takeda, H.; et al. Diallyl trisulfide induces apoptosis by suppressing NF-B signaling through destabilization of TRAF6 in primary effusion lymphoma. Int. J. Oncol. 2016, 48, 293–304. [Google Scholar] [CrossRef]
  88. Choi, Y.H. Diallyl trisulfide induces apoptosis and mitotic arrest in AGS human gastric carcinoma cells through reactive oxygen species-mediated activation of AMP-activated protein kinase. Biomed. Pharmacother. 2017, 94, 63–71. [Google Scholar] [CrossRef]
  89. Hwang, J.S.; Lee, Y.Y.; Lee, D.H.; Kwon, K.H. DATS sensitizes glioma cells to TRAIL-mediated apoptosis by up-regulation of death receptor 5 via ROS. Food Chem. Toxicol. 2017, 106, 514–521. [Google Scholar] [CrossRef]
  90. Li, C.; Jing, H.; Ma, G.; Liang, P. Allicin induces apoptosis through activation ofboth intrinsic and extrinsic pathways in glioma cells. Mol. Med. Rep. 2018, 17, 5976–5981. [Google Scholar] [CrossRef]
  91. Kema, V.H.; Khan, I.; Kapur, S.; Mandal, P. Evaluating the effect of diallyl sulfide on regulation of inflammatory mRNA expression in 3T3L1 adipocytes and RAW 264.7 macrophages during ethanol treatment. Drug Chem. Toxicol. 2018, 41, 302–313. [Google Scholar] [CrossRef]
  92. Jobani, B.M.; Najafzadeh, N.; Mazani, M.; Arzanlou, M.; Vardin, M.M. Molecular mechanism and cytotoxicity of allicin and all-trans retinoic acid against CD44+ versus CD117+ melanoma cells. Phytomedicine 2018, 48, 161–169. [Google Scholar] [CrossRef]
  93. Kim, H.J.; Kang, S.; Kim, D.Y.; You, S.; Park, D.; Oh, S.C.; Lee, D.H. Diallyl disulfide (DADS) boosts TRAIL-Mediated apoptosis in colorectal cancer cells by inhibiting Bcl-2. Food Chem. Toxicol. 2019, 125, 354–360. [Google Scholar] [CrossRef]
  94. Fredotović, Ž.; Soldo, B.; Šprung, M.; Marijanović, Z.; Jerković, I.; Puizina, J. Comparison of organosulfur and amino acid composition between triploid onion Allium cornutum clementi ex visiani, 1842, and common onion Allium cepa L., and evidences for antiproliferative activity of their extracts. Plants 2020, 9, 98. [Google Scholar] [CrossRef] [PubMed]
  95. Rashed, H.A.E.H.; Abu Almaaty, A.H.; Soliman, M.F.M.; El-Shenawy, N.S. The in vitro antischistosomal activity and genotoxicity of the active ingredients of Allium sativum (Allicin) and Curcuma longa (curcumin). Iran. J. Parasitol. 2021, 16, 101–110. [Google Scholar] [CrossRef]
  96. Ferreira González, I.; Urrútia, G.; Alonso-Coello, P. Systematic Reviews and Meta-Analysis: Scientific Rationale and Interpretation. Rev. Esp. Cardiol. 2011, 64, 688–696. [Google Scholar] [CrossRef] [PubMed]
  97. Sundaram, S.G.; Milner, J.A. Diallyl disulfide suppresses the growth of human colon tumor cell xenografts in athymic nude mice. J. Nutr. 1996, 126, 1355–1361. [Google Scholar] [CrossRef] [PubMed]
  98. Chu, Q.; Lee, D.T.W.; Tsao, S.W.; Wang, X.; Wong, Y.C. S-allylcysteine, a water-soluble garlic derivative, suppresses the growth of a human androgen-independent prostate cancer xenograft, CWR22R, under in vivo conditions. BJU Int. 2007, 9, 925–932. [Google Scholar] [CrossRef] [PubMed]
  99. Nishikawa, T.; Yamada, N.; Hattori, A.; Fukuda, H.; Fujino, T. Inhibition by ajoene of skin-tumor promotion in mice. Biosci. Biotechnol. Biochem. 2022, 66, 2221–2223. [Google Scholar] [CrossRef] [PubMed]
  100. Zhang, L.; Wang, E.; Chen, F.; Yana, H.; Yuan, Y. Potential protective effects of oral administration of allicin on acrylamide-induced toxicity in male mice. Food Funct. 2013, 4, 1229–1236. [Google Scholar] [CrossRef]
  101. Cascajosa Lira, A.; Prieto, A.I.; Baños, A.; Guillamón, E.; Moyano, R.; Jos, Á.; Cameán, A.M. Safety assessment of Propil-propane-thiosulfonate (PTSO): 90-days oral subchronic toxicity study in rats. Food Chem. Toxicol. 2020, 144, 111612. [Google Scholar] [CrossRef]
  102. Mellado-Garcia, P.; Puerto, M.; Pichardo, S.; Llana-Ruiz-Cabello, M.; Moyano, R.; Blanco, A.; Jos, A.; Camean, A.M. Toxicological evaluation of an Allium-based commercial product in a 90-day feeding study in Sprague-Dawley rats. Food Chem. Toxicol. 2016, 90, 18–29. [Google Scholar] [CrossRef]
  103. Cascajosa-Lira, A.; Puerto, M.; Prieto, A.I.; Pichardo, S.; Díez-Quijada Jiménez, L.; Baños, A.; Guillamón, E.; Moyano, R.; Molina-Hernández, V.; Jos, Á.; et al. Genotoxicity Evaluation of Propyl-Propane-Thiosulfinate (PTS) from Allium genus Essential Oils by a Combination of Micronucleus and Comet Assays in Rats. Foods 2021, 10, 989. [Google Scholar] [CrossRef]
  104. Mellado-García, P.; Puerto, M.; Prieto, A.I.; Pichardo, S.; Martín-Cameán, A.; Moyano, R.; Blanco, A.; Cameán, A.M. Genotoxicity of a thiosulfonate compound derived from Allium sp. Intended to be used in active food packaging: In vivo comet assay and micronucleus test. Mutat Res Genet. Toxicol Env. Mutagen 2016, 800–801, 1–11. [Google Scholar] [CrossRef] [PubMed]
  105. Dutta, A.; Dahiya, A.; Prakash, A.; Agrawala, P.K. Acute toxicity of diallyl sulfide derived from Allium sativum (garlic) in mice and its possible mechanisms. Phytomed. Plus 2021, 1, 100084. [Google Scholar] [CrossRef]
Figure 1. PRISMA flowchart of article selection.
Figure 1. PRISMA flowchart of article selection.
Foods 11 02620 g001
Figure 2. Graphical model: (A) reported beneficial health effects, (B) in vitro toxicological evaluation and (C) reported results.
Figure 2. Graphical model: (A) reported beneficial health effects, (B) in vitro toxicological evaluation and (C) reported results.
Foods 11 02620 g002
Table 1. Organosulfur compounds present in Allium species, chemical structure and main properties.
Table 1. Organosulfur compounds present in Allium species, chemical structure and main properties.
NameChemical StructureMode of ActionReference
E-Ajoene
Z-Ajoene
Foods 11 02620 i001Antibacterial: in vitro activity against Gram +:
MIC = 5–250 µg/mL and Gram –: MIC = 100–500 µg/mL
[21]
Antifungal: MIC = 15–50 µg/mL
Antioxidant enzyme induction: NAD(P)H: quinone oxidoreductase-1 (NQO1)[22]
AlliinFoods 11 02620 i002Antioxidant: superoxide scavenging activity[23]
AllicinFoods 11 02620 i003Antibacterial: in vitro activity against Staphylococcus strains MIC = 600 µg/mL and Gram-bacteria: MIC = 4–300 µg/mL[24,25,26]
Antifungal: MIC = 1.52–6.25 µg/mL[27]
Antioxidant activity in vivo: SOD and GSH-Px activities
increased.
Radical scavenging ability of hydroxyl radical increase with Allicin
[24,28,29]
Di-allyl-disulphide
(DADS)
Foods 11 02620 i004Antibacterial activity against S. aureus:
MIC = 4 µg/mL
and Helicobacter pylori MIC = 200 µg/mL
[25,30]
Antifungal activity against Aspergillus spp:
MIC = 8–12 µg/mL and Candida spp: MIC = 4–12 µg/mL
[30]
Antioxidant in vivo activity[31,32]
Di-allyl-sulphide
(DAS)
Foods 11 02620 i005Antibacterial activity against S. aureus:
MIC = 20 µg/mL
and H. pylory MIC = 4 µg/mL
[25,30]
Antifungal activity against Aspergillus spp:
MIC= 40–64 µg/mL and Candida spp: MIC= 32–72 µg/mL
[30]
Antioxidant in vivo activity[33]
Di-allyl-trisulphide
(DATS)
Foods 11 02620 i006Antibacterial activity against S. aureus:
MIC = 2 µg/mL
and H. pylory MIC = 25 µg/mL
[25,30]
Antifungal activity against Aspergillus spp:
MIC = 2–8 µg/mL and Candida spp: MIC = 1–8 µg/mL
[30]
Antioxidant in vitro activity[34]
Dipropyl disulphide
(DPDS)
Foods 11 02620 i007Antioxidant in vitro activity [16]
Dipropyl sulphide
(DPS)
Foods 11 02620 i008Antioxidant in vitro activity[16]
Propyl-propane-tiosulphonate
(PTSO)
Foods 11 02620 i009Antibacterial activity against Gram +: MCB = 0.5–10 µg/mL and Gram-: MCB = 1.25–10 µg/mL[20,35,36]
Antifungal:
MFC against Verticillium dahliae = 19.53–39.06 µg/mL
MFC90 against Candida spp. = 64–128 µg/mL
[11,37]
Antiprotozoal and Antiparasitic: reduce the number of apicomplexa in monogastric animals.
Reducing a plurality of aquatic parasites in aquatic animals.
[38,39]
Antioxidant[20]
Propyl-propane-tiosulphinate
(PTS)
Foods 11 02620 i010Antibacterial activity against Gram -: MCB = 128–1024 µg/mL and Gram +: MCB = 128 µg/mL[11]
Antifungal activity
MFC against Verticillium dahliae = 78.13 µg/mL
MFC90 against Candida spp = 128 µg/mL
[34,40]
Antiprotozoal and antiparasitic: reduce the number of Apicomplexa in monogastric animals.
Reducing a plurality of aquatic parasites in aquatic animals.
[38,39]
S-allylcysteine
(SAC)
Foods 11 02620 i011Antioxidant activity by scavenging ROS[41]
VinyldithiinFoods 11 02620 i012Antioxidant activity[42]
Publisher’s Note: MDPI stays neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Share and Cite

MDPI and ACS Style

Cascajosa-Lira, A.; Andreo-Martínez, P.; Prieto, A.I.; Baños, A.; Guillamón, E.; Jos, A.; Cameán, A.M. In Vitro Toxicity Studies of Bioactive Organosulfur Compounds from Allium spp. with Potential Application in the Agri-Food Industry: A Review. Foods 2022, 11, 2620. https://doi.org/10.3390/foods11172620

AMA Style

Cascajosa-Lira A, Andreo-Martínez P, Prieto AI, Baños A, Guillamón E, Jos A, Cameán AM. In Vitro Toxicity Studies of Bioactive Organosulfur Compounds from Allium spp. with Potential Application in the Agri-Food Industry: A Review. Foods. 2022; 11(17):2620. https://doi.org/10.3390/foods11172620

Chicago/Turabian Style

Cascajosa-Lira, Antonio, Pedro Andreo-Martínez, Ana Isabel Prieto, Alberto Baños, Enrique Guillamón, Angeles Jos, and Ana M. Cameán. 2022. "In Vitro Toxicity Studies of Bioactive Organosulfur Compounds from Allium spp. with Potential Application in the Agri-Food Industry: A Review" Foods 11, no. 17: 2620. https://doi.org/10.3390/foods11172620

APA Style

Cascajosa-Lira, A., Andreo-Martínez, P., Prieto, A. I., Baños, A., Guillamón, E., Jos, A., & Cameán, A. M. (2022). In Vitro Toxicity Studies of Bioactive Organosulfur Compounds from Allium spp. with Potential Application in the Agri-Food Industry: A Review. Foods, 11(17), 2620. https://doi.org/10.3390/foods11172620

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