Skip to main content

Postbiotic metabolites produced by Lactobacillus plantarum strains exert selective cytotoxicity effects on cancer cells

Abstract

Background

Lactobacillus plantarum, a major species of Lactic Acid Bacteria (LAB), are capable of producing postbiotic metabolites (PM) with prominent probiotic effects that have been documented extensively for rats, poultry and pigs. Despite the emerging evidence of anticancer properties of LAB, very limited information is available on cytotoxic and antiproliferative activity of PM produced by L. plantarum. Therefore, the cytotoxicity of PM produced by six strains of L. plantarum on various cancer and normal cells are yet to be evaluated.

Methods

Postbiotic metabolites (PM) produced by six strains of L. plantarum were determined for their antiproliferative and cytotoxic effects on normal human primary cells, breast, colorectal, cervical, liver and leukemia cancer cell lines via MTT assay, trypan blue exclusion method and BrdU assay. The toxicity of PM was determined for human and various animal red blood cells via haemolytic assay. The cytotoxicity mode was subsequently determined for selected UL4 PM on MCF-7 cells due to its pronounced cytotoxic effect by fluorescent microscopic observation using AO/PI dye reagents and flow cytometric analyses.

Results

UL4 PM exhibited the lowest IC50 value on MCF-7, RG14 PM on HT29 and RG11 and RI11 PM on HL60 cell lines, respectively from MTT assay. Moreover, all tested PM did not cause haemolysis of human, dog, rabbit and chicken red blood cells and demonstrated no cytotoxicity on normal breast MCF-10A cells and primary cultured cells including human peripheral blood mononuclear cells, mice splenocytes and thymocytes. Antiproliferation of MCF-7 and HT-29 cells was potently induced by UL4 and RG 14 PM respectively after 72 h of incubation at the concentration of 30% (v/v). Fluorescent microscopic observation and flow cytometric analyses showed that the pronounced cytotoxic effect of UL4 PM on MCF-7 cells was mediated through apoptosis.

Conclusion

In conclusion, PM produced by the six strains of L. plantarum exhibited selective cytotoxic via antiproliferative effect and induction of apoptosis against malignant cancer cells in a strain-specific and cancer cell type-specific manner whilst sparing the normal cells. This reveals the vast potentials of PM from L. plantarum as functional supplement and as an adjunctive treatment for cancer.

Peer Review reports

Background

Lactic acid bacteria (LAB) are generally accepted as safe, active and functional ingredients for foods owing to their long history of consumption together with fermented foods [1]. Furthermore, their metabolic end products, such as lactic acid and bacteriocin, can be used as natural preservatives and antimicrobial agents against contamination and food spoilage [2]. The beneficial effects of LAB have been reported extensively, such as the prevention of urogenital infections [3], control of inflammatory bowel diseases [4], immunomodulating functions [5, 6], control of serum cholesterol [7] and prevent certain types of cancer [8]. In addition, numerous in vivo, in vitro, human and epidemiological studies have provided increasing evidence of LAB effects on colon, bladder, liver, breast, and gastric cancers [8,9,10,11,12]. These effects are mediated by various mechanisms, such as the alteration of gastrointestinal microflora, enhancement of the host’s immune response, antioxidative, antiproliferative, and induction of apoptosis [13, 14]. Nevertheless, various LAB species employed as starter cultures in food fermentation may act as a reservoir for antibiotic resistance genes with the potential to be transferred to humans, animals and pathogenic microbes via food chain [15,16,17,18,19]. Antibiotic resistance is a major safety concern when the resistance is transferrable, especially to pathogenic bacteria [20]. Another concern for the application of living probiotic cultures is their viability below expected standards. This may affect their efficacy as viable commercial probiotic strains [21]. In addition, the functionality of probiotic cultures might be affected without changes in cell viability [22]. Several clinical studies demonstrated the use of live bacteria might possess adverse effects such as acute inflammation and increased mortality [23, 24].

Recently, there is growing interest in probiotic effects exerted by microbial metabolites known as bioactive postbiotic metabolites (PM), particularly in maintaining gut health, curing gut diseases and enhancing innate immunity [25,26,27,28,29]. PM ranges from soluble or secreted factors, metabolites, bacteriocins, and cell-free supernatant was reported with equivalent efficacy as live probiotics vastly related to Lactobacilli [24, 27]. Konstantinov et al. [28] summarised the role of postbiotics in maintaining colonic health and proposed that postbiotics can be a safer alternative in comparison to live bacteria. They further suggested the possibility of postbiotics in improving the patients’ quality of life in the later stage of colorectal cancer, while Tsilingiri et al. [24] proposed the utilisation of postbiotic in the treatment and prevention of gut-related diseases such as inflammatory bowel disease. Prominent probiotic effects of bacteriocin-containing postbiotic produced by the six strains of Lactobacillus plantarum employed in this study have been documented extensively for rats [29,30,31], poultry [32, 33] and pigs [34] with growth- and health-promoting effects observed with the supplementation of postbiotics as antibiotic replacer in their feeds. These L. plantarum strains produce several PM such as lactic acid, acetic acid and bacteriocin [35, 36]. Interestingly, Moghadam et al. [37] and Tai et al. [38] reported L. plantarum I-UL4 harbours a novel combination of two bacteriocin genes, the plantaricin W and plantaricin EF with broad inhibition activity against pathogenic Gram-positive and Gram-negative bacteria [35, 36].

The effects of different components (culture supernatants, cytoplasmic extracts, cell-wall extracts and live cells) of Lactobacillus gasseri and Lactobacillus crispatus on proliferative and apoptotic responses of normal and tumour cervical cells have been reported by Motevaseli et al. [39]. In addition, the exopolysaccharide from L. plantarum 70,180 have been reported to exert antitumor activity against colon carcinoma cells [40]. The culture supernatants of Lactobacillus casei and Lactobacillus paracasei strains isolated from human breast milk that have bioactivity of cytotoxicity and apoptosis against cervical cancer cells were suggested to have the potential as natural antitumour drugs [41]. Therefore, the cytotoxicity of PM produced by six strains of L. plantarum on various cancer and normal cells was determined in this study. In addition, the toxicity of the selected PM on human, dog, chicken and rabbit red blood cells (RBC) was subsequently verified. Cancer is a global epidemic disease affecting people at all ages and socio-economic groups. According to the report of GLOBOCAN (2012) [42], breast cancer incidence, mortality and 5-year (2012–2016) prevalence was estimated to be the highest for women, whereas colorectal cancer incidence, mortality and 5-year (2012–2016) prevalence were ranked second and third for women and men, respectively. Hence, the cytotoxic effect of PM produced by six strains of L. plantarum was further investigated on breast and colon cancer cell lines. The mode of cell death induced by the selected PM produced by L. plantarum I-UL4 was subsequently verified on breast cancer cells.

Materials and methods

Bacterial cultures

Six bacteriocin-producing L. plantarum identified as I-UL4, TL1, RS5, RI11, RG11 and RG14 strains [43] were isolated from Malaysian foods [44] and obtained from the Laboratory of Industrial Biotechnology, Department of Bioprocess Technology, Faculty of Biotechnology and Biomolecular Sciences, Universiti Putra Malaysia (UPM). They were maintained in De Man, Rogosa and Sharpe (MRS) medium (Merck, Germany) and propagated twice in MRS broth for 24 h at 30 °C under anaerobic condition prior to each experiment. The stock cultures were kept at -80 °C in MRS broth containing 20% (v/v) sterile glycerol.

Cell culture and maintenance

Human breast cancer cells MCF-7 (ATCC-HTB-22), colorectal cancer cells HT-29 (ATCC HTB-38), cervical cancer cells HeLa (ATCC CCL2), liver cancer cells Hep-G2 (ATCC HB-8065), leukemia cells HL60 (ATCC CCL-240) and K562 (ATCC CCL-243) were provided by Animal Tissue Culture Laboratory of UPM. The nonmalignant MCF-10A cells (reference of normal glandular epithelium) were purchased from American Type Culture Collection (ATCC). All cells were maintained at 37 °C under 5% CO2 atmosphere using ATCC recommended medium supplemented with 10% (v/v) heat-inactivated fetal bovine serum and 100 IU/ml penicillin-streptomycin.

Preparation of normal mice splenocytes and thymocytes, human peripheral blood mononuclear cells

The protocols of animal and human cell experiments were conducted according to the guidelines of the Institutional Animal Care and Use Committee (IACUC) of UPM. The 7–8 week old male mice were sacrificed by cervical dislocation. A transverse cut was made in the middle of the abdominal area to remove the spleen. The rib cage was cut off to remove the thymus. The spleen and thymus were then smashed separately by using wire mesh in phosphate buffer saline (PBS) and centrifuged at 200×g for 10 min. The cell pellet of splenocyte was re-suspended in 4 ml of lysis buffer and left on ice for 5 min prior to centrifugation at 200×g for 10 min. The splenocyte was then washed with 4 ml of PBS. For the thymocytes, the cell pellet was washed with PBS twice.

Human peripheral blood mononuclear cells and blood samples were withdrawn from healthy donors and kept in preservative-free heparin tubes (BD Biosciences, USA). Anticoagulated blood was diluted with an equal volume of PBS (pH 7.5) and slowly layered over Ficoll-Paque Plus (GE Healthcare, Sweden), followed by centrifugation at 400×g for 40 min at 18–20 °C. Plasma, mononuclear, Ficoll-Paque Plus, granulocytes and erythrocytes were separated into four layers. The mononuclear layer was transferred to a centrifuge tube and washed with 4 ml of PBS (pH 7.5) twice after the plasma layer was aspirated off.

The washed cell pellets of mice splenocytes, thymocytes and human peripheral blood mononuclear cells (PBMC) were re-suspended in complete growth media to a density of 5 × 105 cells ml− 1 for MTT assay.

Preparation of PM

Respective L. plantarum [1% (v/v)] was grown in MRS broth for 24 h at 30 °C. The cells were then separated by centrifugation at 10,000×g for 10 min at 4 °C. The supernatant was collected as PM. The pH of each PM was then adjusted to physiological pH (pH 7.2–7.4) using 5 M sodium hydroxide and all tested PM were then filtered through 0.22 μm polyethersulfone membrane syringe filter (Millipore, USA) prior to cytotoxicity, haemolysis, antiproliferation, and cell dead mode assays.

Cytotoxic effect of PM on various cancer and normal cells

Cancer cells were plated onto 96-well microplates at 1 × 105 cells ml− 1 and incubated at 37 °C in a 5% CO2 incubator for cytotoxicity assay. After 24 h, two-fold dilution of PM produced by L. plantarum strains were administered in complete growth medium with concentrations ranging from 0.47–30% (v/v). After each respective incubation interval (24, 48 and 72 h), 20 μl of 3-(4,5-dimethylthiazol-2-yl)-2,5-diphenyl tetrazolium bromide (MTT) solution (Sigma, USA) (5 mg ml− 1 in PBS) were added to each well and the plates were incubated in the dark for 4 h prior to centrifugation at 200×g for 5 min to separate the formazan crystals. The resultant formazan crystals were dissolved in 100 μl of dimethylsulfoxide (Fisher Scientific, UK) for 15–30 min after removing 170 μl of the growth medium from each well. The absorbance of formazan dye was quantified by a μ Quant ELISA reader (Biotek EL340, USA) at 570 nm with a reference wavelength of 630 nm. The experiment was repeated three times with triplicate samples. The following equation was used to calculate the percentage of cell viability: (Asample-Ablank)/(Acontrol-Ablank) × 100%, where Asample represents the absorbance of cells treated with PM; Ablank represents absorbance of PM and Acontrol represents absorbance of untreated cells. The concentration for 50% of growth (IC50) was determined by plotting the percentage of cell viability versus the concentration of PM.

Haemolytic effect of PM on red blood cells

Haemolysis of red blood cell (RBC) was determined as described by Reddy et al. [45] with minor modifications. The fresh animal blood samples were obtained directly from the slaughter house of UPM. The collection of human peripheral blood mononuclear cells and blood samples were also approved by the IACUC of UPM. Human blood samples were withdrawn from healthy donors after consent was informed according to the protocol of IACUC of UPM. Washed RBC [2% (v/v)] were incubated with 100 μL of two-fold increasing concentrations [1.56–100% (v/v)] of PM prepared by using PBS in 96-round bottom well plates for 10 min.

The intact RBC were then separated by centrifugation at 400×g for 5 min and the absorbance of the supernatant was determined at 575 nm with a reference wavelength at 540 nm. Percentage of haemolysis was calculated by using the following formula: (Asample-Acontrol-Ablank)/(A100% lysis-Acontrol-Ablank) × 100%. Zero and 100% haemolysis were determined with isotonic PBS and deionised water. Two-fold diluted PM in PBS without RBC was used as blank, while 1% (v/v) sodium dodecyl sulfate (SDS) was served as positive control. The experiments were repeated three times with triplicate samples. The concentration of PM resulting in 50% lysis (L50) was determined from dose-response curve.

Bromodeoxyuridine cell proliferation assay

Bromodeoxyuridine (BrdU) cell proliferation assay was conducted to study the antiproliferation effect of PM on breast and colon cancer cells. A cell population of 5 × 103 cells well− 1 was seeded in a 96-well plate. Two controls were prepared: treatment blank (without cells) and background control (containing cells without BrdU reagent). The growth medium was aspirated after 24 h and replaced with fresh medium containing 0, 15 and 30% (v/v) PM and incubated for 24, 48 and 72 h, respectively. Further procedure was carried out according to the protocol of manufacturer (Millipore). The colored reaction product was quantified by measuring the absorbance at 450 nm (reference wavelength 540 nm) using a microplate reader. Three independent experiments were performed with triplicate samples. The following formula was used to calculate the percentage of cell proliferation: (Asample-Ablank)/(Acontrol-Ablank) × 100%. The percentage of antiproliferation was obtained by subtracting 100 with the percentage of cell proliferation.

Trypan blue exclusion assay

Cells were plated in 6-well tissue culture plate (2.5 × 105 cells well− 1) and incubated for 24 h. The medium was then aspirated and replenished with fresh medium containing 0, 15 and 30% (v/v) PM. The cells were trypsinised at 0, 24, 48 and 72 h of incubations and collected by centrifugation at 200×g for 5 min. The supernatant was aspirated and the cell pellet was re-suspended in 100 μl of complete growth media.

Cell counting was performed by using hemocytometer. Ten microliter of cells was added with 10 μl of trypan blue dye and viewed under inverted microscope. The viable cells that did not stain blue were counted. The experiment was repeated three times with triplicate samples. Results were expressed as cell population, where it equals to the viable cell concentration in a single well multiplied with 100 μl of complete media that used to re-suspend the cell pellet.

Fluorescent microscopy using acridine orange/propidium iodide staining

MCF-7 cells (2 × 105 cells well− 1) seeded in six well plates were treated with 0% (v/v), 15% (v/v) and 30% (v/v) of PM and incubated in 5% CO2 atmospheric condition at 37 °C for 24, 48 and 72 h. After each incubation time, the detached cells in the medium were collected and mixed with trypsinised cells. The cells were then washed with PBS and incubated for 10 min with 10 μl of AO (100 μg ml− 1) and PI (100 μg ml− 1) at a ratio of 1:1 in 1 ml of cells. The stained cells were collected by centrifugation at 200×g for 5 min. The supernatant was aspirated and left 50 μl of remnant supernatant. The cell pellet was re-suspended and 10 μl of cell suspension was placed on slide and viewed under fluorescent microscope (Nikon FC-35DX, Japan) within 30 min using an excitation filter and barrier filter at 450–490 nm and long pass filter of 520 nm. Viable (green intact cells), apoptotic (green shrinking cells with condensed or fragmented nucleus), late apoptotic and necrotic (red) cells were observed. The experiment was repeated for at least three times with triplicate samples.

Flow cytometry analysis

Cell cycle analysis and Annexin V was performed to determine the cell cycle perturbation of MCF-7 cells when treated with postbiotic UL4 at different concentrations [15 and 30% (v/v)] and different incubation periods (24, 48 and 72 h). Phosphatidylserine exposure on the cell surface of MCF-7 cells treated with postbiotic UL4 was detected using Annexin V-FITC Apoptosis Detection Kit (BD Bioscience, USA) according to manufacturer’s protocol.

Statistical analysis

Data of cytotoxicity and antiproliferative effects were analysed by using ANOVA with all paired-wise multiple comparison procedure (Tukey) and presented as the mean ± standard error of the mean (SEM). The results were analysed by Minitab Statistical Software at differences of P < 0.05.

Results

Cytotoxic effect of PM on various cancer and normal cells

The inhibition concentration of 50% growth (IC50) were determined for various cancer cell lines to compare the cytotoxicity effects of PM produced by L. plantarum I-UL4, TL1, RS5, RG14, RG11 and RI11, which was designated as UL4 PM, TL1 PM, RS5 PM, RG14 PM, RG11 PM and RI11 PM, respectively. The cytotoxic effect of the PM was time- and dose-dependent [41]. No cytotoxicity activity was noted for MRS medium against various cancer cell lines in this study (results not shown). Table 1 shows the PM produced by L. plantarum strains exhibited profound cytotoxic effect at 72 h of incubation on all tested cancer (breast, cervical, colorectal, liver and leukemia) cell lines, although different IC50 values were exhibited by different PM. Moreover, the tested PM exhibited more potent cytotoxic effect on suspension cancer cells such as HL60 and K562 as compared to anchorage-dependent cancer cells (such as MCF-7, HeLa, Hep-G2 and HT-29). This was particularly evident on HL60 in comparison to K562. Interestingly, amongst the anchorage-dependent cells, the PM produced by all L. plantarum strains was more cytotoxic against MCF-7 cells. Among them, UL4 PM exhibited the most potent cytotoxic effect against MCF-7 cells with IC50 of 10% (v/v) at 72 h of incubation (Table 1).

Table 1 IC50 values of PM produced by six strains of Lactobacillus plantarum on various cancer cells

Despite the profound cytotoxic effects of PM on various cancer cells, no IC50 value was detected on rapidly-dividing normal MCF-10A cells, with the exception that limited cytotoxicity [IC50 value of 26% (v/v)] was detected when treated with UL4 PM for 72 h (results not shown).

Haemolytic effect of PM on red blood cells

The haemolytic effect of PM produced by six strains of L. plantarum was determined with red blood cells (RBC) of humans, dogs, rabbits and chickens. With respect to lytic concentration at equilibrium (which was usually expressed as L50 concentration), toxicity on the basis of RBC haemolysis was not detected for all tested PM although the concentration was increased to 100% (v/v) as compared to 1% (v/v) SDS (Fig. 1).

Fig. 1
figure 1

Haemolysis effect of PM produced by six stains of Lactobacillus plantarum on (a) human, (b) rabbit, (c) chicken and (d) dog RBC. T1: 100% (v/v); T2: 50% (v/v): T3: 25% (v/v); T4: 12.5% (v/v); T5: 6.25% (v/v); T6: 3.13% (v/v); T7: 1.56% (v/v); 1% SDS: 1% (w/v) SDS. Each bar represents the mean ± SEM of nine replicates

Antiproliferative activities of PM on breast and colon cancer cells

In this study, the antiproliferative activities evaluated by Bromodeoxyuridine(BrdU) proliferation assay of PM produced by L. plantarum I-UL4, TL1, RS5, RG14, RG11 and RI11 were particularly focused on the breast and colon cancer cells due to the high incidence and mortality of both cancers as reported by GLOBOCAN (2012) [42], in addition to extensive studies reported on LAB and their anticancer effects on colorectal cancer [8, 14].

Significant antiproliferative effect (P < 0.05) on MCF-7 (Fig. 2 panel a) and HT-29 (Fig. 2 panel b) were detected for all PM at the concentrations of 15% (v/v) (results not shown) and 30% (v/v), respectively, whereby continuous reduction of BrdU incorporation was noted with increased concentrations of PM and incubation time as shown in Fig. 2. In comparison, all tested PM decreased the rate of DNA synthesis in MCF-7 cells (Fig. 2 panel a) more profoundly than in HT-29 cells (Fig. 2 panel b) throughout the incubation times used in this study. At 72 h of incubation, 30% (v/v) concentration of PM produced by L. plantarum I-UL4 ceased the DNA synthesis of MCF-7 cells remarkably, whereas small amount of BrdU was incorporated into MCF-7 cells when treated with other PM at the concentration of 30% (v/v) (Fig. 2 panel a). As for HT-29 cells, RG14 PM demonstrated the highest reduction (89%) of DNA synthesis in HT-29 cells as compared to the PM produced by the other L. plantarum strains (Fig. 2 panel b).

Fig. 2
figure 2

Antiproliferative effect of 30% (v/v) of PM produced by six strains of Lactobacillus plantarum on (a) MCF-7 cells and (b) HT-29 cells determined by BrdU cell proliferation assay. Values within the same row and experiment having an asterisk indicate significant differences (P < 0.05) from the untreated control groups. Error bars represent the standard error of the mean of nine replicates

Further verification of antiproliferative effects of PM produced by L. plantarum was conducted for growth arrest study via trypan blue cell counting. PM produced by L. plantarum I-UL4 and RG14 with distinct antiproliferative property on MCF-7 cells and HT-29 cells respectively were selected and the viable cell count was enumerated. Figure 3 shows that PM produced by L. plantarum I-UL4 exhibited time and dose-dependent inhibition on proliferation of MCF-7 cells. Proliferation of MCF-7 cells was significantly arrested by UL4 PM at the concentration of 30% (v/v). Pronounced differences in cell population were observed among 15% (v/v) and 30% (v/v) PM-treated MCF-7 cells and the untreated cells. This difference became more significant (P < 0.05) with prolong incubation time. After 72 h of incubation, the ratio of viable population of MCF-7 cells incubated with 15% (v/v) and 30% (v/v) of UL4 PM to the viable population of untreated cells were 1:2 and 1:1.3 at 24 h; 1:3 and 1:1.9 at 48 h; and 1:7.7 and 1:2.2 at 72 h, respectively.

Fig. 3
figure 3

Antiproliferative effect of PM produced by Lactobacillus plantarum I-UL4 on MCF-7 cells determined via trypan blue staining and cell count. Cell population equals to the viable cell concentration of a single well multiplied with 100 μl of complete growth media used to suspend the cell pellet. Values within the same row and experiment having an asterisk indicate significant difference (P < 0.05) from the untreated control groups. Error bars represent the standard error of the mean

As for HT-29 cells, significant difference in cell population was observed for HT-29 cells treated with 15% (v/v) and 30% (v/v) RG14 PM, as well as the untreated cells. RG14 PM exhibited time-and dose-dependent antiproliferative effects on HT-29 cells, whereby the ratio of viable population of HT-29 cells treated with 15% (v/v) and 30% (v/v) of RG14 PM to the viable population of untreated population were 1:1.6 and 1:1.3 at 24 h; 2.1 and 1:1.3 at 48 h; and 1:4.3 and 1:1.5 at 72 h, respectively (Fig. 4).

Fig. 4
figure 4

Antiproliferative effect of PM produced by Lactobacillus plantarum RG14 on HT-29 cells determined via trypan blue staining and cell count. Cell population equals to the viable cell concentration of a single well multiplied with 100 μl of complete growth media used to suspend the cell pellet. Values within the same row and experiment having an asterisk indicate significant difference (P < 0.05) from the untreated control groups. Error bars represent the standard error of the mean

Apoptosis induced by UL4 PM

Results obtained in this time course study showed that 15 and 30% (v/v) UL4 PM induced apoptotic cell death in MCF-7 cells. Figure 5 shows different morphologies of MCF-7 cells when treated with UL4 PM as detected by using acridine orange (AO) and propidium iodide (PI) staining. Cell shrinkage, masses of condensed chromatin aggregation at the nuclear membrane as shown by bright fluorescence at the membrane periphery, membrane blebbing and cytoplasmic and nuclear fragmentation leading to the formation of apoptotic bodies were among the striking apoptotic cellular changes noted in the treated MCF-7 cells observed under fluorescence microscope.

Fig. 5
figure 5

Apoptosis induction by PM of Lactobacillus plantarum I-UL4 on MCF-7 cells. Panel a: 15% (v/v), 24 h; b: 15% (v/v), 48 h; c: 15% (v/v), 72 h; d: 30% (v/v), 24 h; e: 30% (v/v), 48 h; and f: 30% (v/v), 72 h. Morphological changes following exposure to PM produced by L. plantarum I-UL4 were typical of apoptosis, i.e. cell shrinkage (a), membrane blebbing (b), perinuclear chromatin condensation (c), apoptotic bodies formation (d), and secondary necrotic cells (e). (Bar: 10 μm; magnification: 400×)

Flow cytometric assessments via cell cycle perturbation and phospatidylserine exposure using Annexin V verified further the apoptotic effect of UL4 metabolites on MCF-7 cells. Cell cycle was predominantly arrested at G0/G1 phase after 48 h of incubation for both treatment groups of 15% (v/v) and 30% (v/v) (Table 2), while apoptotic cells were majorly predominating the cell population in a dose- and time-dependent manner especially when treated with 30% (v/v) UL4 PM (Table 3).

Table 2 Effect of postbiotic UL4 on cell cycle phases distribution of MCF-7 cells
Table 3 Cell death modes of MCF-7 cells induced by postbiotic UL4

Discussion

Food has long been known to be a vital source to maintain well-being and health status. Nowadays, the functional food market is in the limelight and gains more attention from consumer on the functionality of foods. However, some of these foods are costly and the unclear safety of the food components becomes drawbacks to consumers [46]. Postbiotic containing bacteriocins, for example, are constituted with small peptides, which are mostly non-immunogenic and biodegradable, thus safe for consumption and would not contaminate the environment when disposed [47]. Furthermore, the utilisation of postbiotic are not limited to functional foods, probiotic-mediated suppression of cancer or inflamed cells have also been reported for postbiotic [10, 24, 25]. Postbiotic from lactobacilli offers multitude possibilities for utilisation in cancer cells and inflammatory studies [48]. This offers a safer alternative to the current treatment of radio- and chemotherapies that is limited by toxicities associated with the latter therapies. In this present study, we investigated the cytotoxic effect of PM produced by six strains of L. plantarum on cancer and normal cell line to determine the potential of the PM as a functional supplement and as an adjunctive treatment for cancer.

As shown in this study, RG11 PM and RI11 PM exhibited remarkable cytotoxic effect on all tested suspension cancer cells (K562 and HL60) but they did not exhibit any significant toxicity on epithelial-origin cancer cells (HeLa, Hep-G2 and HT-29) except for MCF-7 breast cancer cells. In addition, only RG14 PM and RS5 PM showed cytotoxic effects on HT-29 cells with IC50 values of 22 and 28% (v/v), respectively. The strain-specific and cell type-dependent cytotoxic effects of PM were in agreement with the finding of Chumchalova and Smarda, whereby the inhibitory effects of colicins were cell-, species- and strain-specific and act in dose-dependent manner [49]. Furthermore, a study conducted by Tan et al. [50] demonstrated that different media compositions of reconstituted MRS with the supplementation of polysorbate 80 affected significantly the selective cytotoxic and antiproliferative effects of PM produced by L. plantarum I-UL4 on MCF-7 cells.

Assessment of potential risk to human health is essential for the products including those for food, home care, personal care, pharmaceutical use and pesticides [51]. Most cytotoxic agents are not only targeting to the rapidly dividing cancer cells, but also simultaneously giving deleterious side effect to some rapidly dividing normal cells. Some normal tissues are also sensitive to the induction of cell death by cytotoxic agents [52, 53]. In this study, no marked toxicity on normal breast MCF-10A cells was noted, which agreed with Motasevali et al. [39] that have reported that culture supernatants of L. gasseri and L. crispatus are non-toxic to normal cervical cells. Furthermore, inert towards actively proliferating primary mice splenocyte, thymocytes and human peripheral mononuclear cells were detected for the tested PM, indicating their selectivity on malignant cells.

Other than normal cell line and primary cultured cells, erythrocytes or RBC are employed as a prime candidate for the determination of membranolytic or cytolytic activities. The PM produced by L. plantarum I-UL4 was employed for the haemolysis study since it exhibited profound cytotoxic effects on all tested suspension cancer cells and epithelial-origin cancer cells, except for HT-29 cells (Table 1). The results of haemolytic effect showed that there was no haemolysis observed for all RBC treated with UL4 PM, which agreed with the findings of Thirabunyanon et al. [54]. The haemolytic results implied that UL4 PM has no adverse effect on membrane permeability properties and the cytotoxic effect of PM observed in this study was not attributed to the osmotic pressure and acute toxicity effect on the treated cancer cells.

In addition, a myriad of health effects has been associated vastly to LAB and most of the studies on anticancer effects of LAB were demonstrated extensively on colorectal cancer [55]. Thus, MCF-7 and HT-29 cells were selected subsequently on the basis of their reported prevalence on the global population, for the determination of antiproliferation effect of PM. The results of the antiproliferative effect correlated well with cytotoxic effect of tested PM, which was also strain-specific and cell type-dependent. UL4 PM manifested the most profound antiproliferative effect on MCF-7 cells, whereas RG14 PM showed the most significant (P < 0.05) antiproliferative effect on HT-29 cells. In comparison, the antiproliferative effect on MCF-7 cells induced by UL4 PM was more profound that the RG14 PM on HT-29 cells. Nonetheless, the antiproliferative effects of UL4 PM on MCF-7 cells and RG14 PM on HT-29 cells were further verified by trypan blue assay and similar results were also obtained as shown in BrdU cell proliferation assay.

The mode of cell death induced by UL4 PM against MCF-7 cells was subsequently determined in this study since the UL4 PM exhibited the most potent cytotoxic effect on MCF-7 cells in comparison to cytotoxic effect manifested on HT-29 cells as well as the effect induced by the RG14 PM. Staining with fluorescence dyes such as AO and PI is considered as an appropriate method for evaluating the changes of nuclear morphology [56] associated with cytotoxic activity, which can be used to facilitate the differentiation of cell death mode, as well as the characteristic morphological changes in chromatin and cell membrane involved in cytotoxic activity induced by cytotoxic agent, as well as the detection of the cell cycle perturbation via flow cytometric analyses. Based on nuclear morphology (perinuclear chromatin condensation, nuclear collapse and eventually fragmentation), several subpopulations of apoptotic cells could be distinguished from viable and necrotic cells [57]. Morphological changes in a transition from membrane blebbing and nucleus condensation, cell shrinkage and apoptotic body formation to secondary necrotic bodies (which nucleus was stained orange-red but the cytoplasm was stained green) as observed in this study inferring that apoptosis rather than necrosis was induced by the UL4 PM. The cell cycle arrest at G0/G1 phase as well as detection of exposed phosphatidylserine on the cell surface via flow cytometric analyses supported further the notion of the apoptotic cell death mode of MCF-7 cells exerted by postbiotic UL4.

It is noteworthy that PM produced by the six strains of L. plantarum were selective on various human cancer cells, implying the vast potential of the PM as an alternative nutraceutical supplements that possess promising anticancer activity due to their low cytotoxicity to normal mammalian cells. Notably, PM produced by L. plantarum I-UL4 specifically induced apoptosis, resulting in the breakdown of apoptosised cells into smaller apoptotic bodies, which are then reported to be cleared by phagocytic cells without evoking any inflammatory reaction in vivo [58], a preferable cell mode death as compared to necrosis [59, 60] for the treatment of cancers.

Conclusion

In conclusion, the results obtained in present study indicate that PM produced by L. plantarum exhibited selective cytotoxic effect on various cancer cells in dose- and time-dependent manners without causing toxic effect or haemolysis on normal cells. Fluorescent microscopic observation and flow cytometric assessments showed that UL4 PM reduced viability of MCF-7 cells via antiproliferative effect and induction of apoptosis. The selectivity of the PM produced by L. plantarum strains to various tumorigenic cells inferring that they hold interesting anti-cancer properties and hence possesses vast potential as a functional supplement and as an adjuncctive treatment against cancer which can be further explored.

Availability of data and materials

The datasets used and/or analysed during this study are available from the corresponding author on reasonable request.

Abbreviations

ANOVA:

Analysis of variance

AO:

Acridine orange

ATCC:

American Type Culture Collection

BrdU:

Bromodeoxyuridine

DNA:

Deoxyribonucleic acid

ELISA:

Enzyme-linked immunoabsorbent assay

h:

Hours

IACUC:

Institute of Animal Care and Use Committee

ICR:

Institute of Cancer Research

LAB:

Lactic acid bacteria

MRS:

de Man Rogosa Sharpe

MTT:

3-(4,5-dimethylthiazol-2-yl)-2,5-diphenyl tetrazolium bromide

PBMC:

Peripheral blood mononuclear cells

PBS:

Phosphate buffer saline

PI:

Propidium iodide

PM:

Postbiotic metabolites

RBC:

Red blood cells

UPM:

Universiti Putra Malaysia

References

  1. Karovičová J, Kohajdová Z. Lactic acid fermentation of various vegetable juices. Acta Aliment. 2005;34:237–46.

    Article  Google Scholar 

  2. Jama YH, Varadaraj MC. Antibacterial effect of plantaricin LP84 on foodborne pathogenic bacteria occurring as contaminants during idli batter fermentation. World J Microbiol Biotechnol. 1999;15:33–8.

    Article  CAS  Google Scholar 

  3. Reid G, Bruce AW. Urogenital infections in women: can probiotics help? Postgrad Med J. 2003;79:428–32.

    Article  CAS  Google Scholar 

  4. Lammers KM, Vergopoulos A, Babel N, Gionchetti P, Rizzello F, Morselli C, et al. Probiotic therapy in the prevention of pouchitis onset: decreased interleukin-1b, interleukin-8, and interferon-g gene expression. Inflamm Bowel Dis. 2005;11:447–54.

    Article  Google Scholar 

  5. Kato-Mori Y, Orihashi T, Kanai Y, Sato M, Sera K, Hagiwara K. Fermentation metabolites from Lactobacillus gasser and Propionibacterium freudenreichii exerts beneficial effects in mice. J Med Food. 2010;13:1460–7.

    Article  CAS  Google Scholar 

  6. Ratajczak C, Duez C, Grangette C, Pochard P, Tonnel A-B, Pestel J. Impact of lactic acid bacteria on dendritic cells from allergic patients in an experimental model of intestinal epithelium. J Biomed Biotechnol. 2007;2007:71921.

    Article  Google Scholar 

  7. Kießling G, Schneider J, Jahreis G. Long-term consumption of fermented dairy products over 6 months increases HDL cholesterol. Eur J Clin Nutr. 2002;56:843–9.

    Article  Google Scholar 

  8. Ishikawa H, Akedo I, Otani T, Suzuki T, Nakamura T, Takeyama I, et al. Randomized trial of dietary fiber and Lactobacillus casei administration for prevention of colorectal tumors. Int J Cancer. 2005;116:762–7.

    Article  CAS  Google Scholar 

  9. Seow SW, Rahmat JN, Kanakkappillai Mohamed AA, Lee YK, Bay BH. Lactobacillus species is more cytotoxic to human bladder cancer cells than Mycobacterium bovis(bacillus Calmette-Guerin). J Urol. 2002;168:2236–9.

    Article  CAS  Google Scholar 

  10. de LeBlanc A de M, Matar C, Thériault C, Perdigón G. Effects of milk fermented by Lactobacillus helveticus R389 on immune cells associated to mammary glands in normal and a breast cancer model. Immunobiology. 2005;210:349–58.

    Article  Google Scholar 

  11. El-Nezami HS, Polychronaki NN, Ma J, Zhu HL, Ling WH, Salminen EK, et al. Probiotic supplementation reduces a biomarker for increased risk of liver cancer in young men from southern China. Am J Clin Nutr. 2006;83:1199–203.

    Article  CAS  Google Scholar 

  12. Park K-B, Oh S-H, Kim N-S, Oh C-H, Jeon J-I. Kimchi fermented in a kimchi refrigerator showed enhanced anti-cancer effects on human leukemia and gastric cancer cells (LB405). FASEB J Federation Am Soc Exp Biol. 2014;2(8):LB405 http://www.fasebj.org/content/28/1_Supplement/LB405. Accessed 25 Dec 2017.

    Google Scholar 

  13. Rafter J. Lactic acid bacteria and cancer: mechanistic perspective. Br J Nutr. 2002;88:S89 Zhong L, Zhang X, Covasa M. Emerging roles of lactic acid bacteria in protection against colorectal cancer. World J. Gastroenterol. 2014;20:7878–86.

    Article  CAS  Google Scholar 

  14. Gfeller KY, Roth M, Meile L, Teuber M. Sequence and genetic organization of the 19.3-kb erythromycin- and dalfopristin-resistance plasmid pLME300 from Lactobacillus fermentum ROT1. Plasmid. 2003;50:190–201.

    Article  CAS  Google Scholar 

  15. Mathur S, Singh R. Antibiotic resistance in food lactic acid bacteria - a review. Int J Food Microbiol. 2005;105:281–95.

    Article  CAS  Google Scholar 

  16. Masco L, Van Hoorde K, De Brandt E, Swings J, Huys G. Antimicrobial susceptibility of Bifidobacterium strains from humans, animals and probiotic products. J Antimicrob Chemother. 2006;58:85–94.

    Article  CAS  Google Scholar 

  17. Hummel AS, Hertel C, Holzapfel WH, Franz CMAP. Antibiotic resistances of starter and probiotic strains of lactic acid bacteria. Appl Environ Microbiol. 2007;73:730–9.

    Article  CAS  Google Scholar 

  18. Aarestrup FM, Wegener HC, Collignon P. Resistance in bacteria of the food chain: epidemiology and control strategies. Expert Rev Anti-Infect Ther. 2008;6:733–50.

    Article  Google Scholar 

  19. Gueimonde M, Sánchez B, de los Reyes-Gavilán CG, Margolles A. Antibiotic resistance in probiotic bacteria. Front Microbiol. 2013;4:1–6.

    Article  Google Scholar 

  20. Fasoli S, Marzotto M, Rizzotti L, Rossi F, Dellaglio F, Torriani S. Bacterial composition of commercial probiotic products as evaluated by PCR-DGGE analysis. Int J Food Microbiol. 2003;82:59–70.

    Article  CAS  Google Scholar 

  21. Vinderola G, Binetti A, Burns P, Reinheimer J. Cell viability and functionality of probiotic bacteria in dairy products. Front Microbiol. 2011;2:1–6.

    Article  Google Scholar 

  22. Besselink MG, van Santvoort HC, Buskens E, Boermeester MA, van Goor H, Timmerman HM, et al. Probiotic prophylaxis in predicted severe acute pancreatitis: a randomised, double-blind, placebo-controlled trial. Lancet. 2008;371:651–9.

    Article  Google Scholar 

  23. Klaenhammer TR, Kleerebezem M, Kopp MV, Rescigno M. The impact of probiotics and prebiotics on the immune system. Nat Rev Immunol Nature Publishing Group. 2012;12:728–34.

    Article  CAS  Google Scholar 

  24. Tsilingiri K, Barbosa T, Penna G, Caprioli F, Sonzogni A, Viale G, et al. Probiotic and postbiotic activity in health and disease: comparison on a novel polarised ex-vivo organ culture model. Gut. 2012;61:1007–15.

    Article  CAS  Google Scholar 

  25. Cicenia A, Scirocco A, Carabotti M, Pallotta L, Marignani M, Severi C. Postbiotic activities of lactobacilli-derived factors. J Clin Gastroenterol. 2014;48:S18–22.

    Article  Google Scholar 

  26. Jin D, Zhang H, Sun J. Manipulation of microbiome, a promising therapy for inflammatory bowel diseases. J Clin Cell Immunol. 2014;5.

  27. Gosálbez L, Ramón D. Probiotics in transition: novel strategies. Trends Biotechnol. 2015;33:195–6.

    Article  Google Scholar 

  28. Konstantinov SR, Kuipers EJ, Peppelenbosch MP. Functional genomic analyses of the gut microbiota for crc screening. Nat Rev Gastroenterol. 2013;10:741–5 Foo HL, Loh TC, Law FL, Lim YZ, Kufli CN, Rusul G. Hepatol. Nature publishing group.

    Article  CAS  Google Scholar 

  29. Effects of feeding Lactobacillus plantarum I-UL4 isolated from Malaysian tempeh on growth performance, faecal flora and lactic acid bacteria and plasma cholesterol concentrations in postweaning rats. Food Sci Biotechnol 2003;12.

  30. Loh TC, Chong SW, Foo HL, Law FL. Effects on growth performance, faecal microflora and plasma cholesterol after supplementation of spray-dried metabolite to postweaning rats. Czech J Anim Sci. 2009;54:10–6.

    Article  CAS  Google Scholar 

  31. Foo HL, Loh TC, Lai PW, Lim YZ, Kufli CN, Rusul G. Effects of adding Lactobacillus plantarum I-UL4 metabolites in drinking water of rats. Pakistan J Nutr. 2003;2:283–8.

    Article  Google Scholar 

  32. Thanh NT, Loh TC, Foo HL, Hair-Bejo M, Azhar K. Effects of feeding metabolite combinations produced by Lactobacillus plantarum on growth performance, faecal microbial population, small intestine villus height and faecal volatile fatty acids in broilers. Br Poult Sci. 2009;50:298–306.

    Article  CAS  Google Scholar 

  33. Loh TC, Thanh NT, Foo HL, Hair-Bejo M, Azhar BK. Feeding of different levels of metabolite combinations produced by Lactobacillus plantarum on growth performance, fecal microflora, volatile fatty acids and villi height in broilers. Anim Sci J. 2010;81:205–14.

    Article  CAS  Google Scholar 

  34. Thu TV, Loh TC, Foo HL, Yaakub H, Bejo MH. Effects of liquid metabolite combinations produced by Lactobacillus plantarum on growth performance, faeces characteristics, intestinal morphology and diarrhoea incidence in postweaning piglets. Trop Anim Health Prod. 2011;43:69–75.

    Article  CAS  Google Scholar 

  35. Van TT, Foo HL, Loh TC, Bejo MH. Inhibitory activity and organic acid concentrations of metabolite combinations produced by various strains of Lactobacillus plantarum. African J Biotechnol. 2011;10:1359–63.

    Google Scholar 

  36. Choe DW, Foo HL, Loh TC, Hair-Bejo M, Awis QS. Inhibitory property of metabolite combinations produced from Lactobacillus plantarum strains. Pertanika J Trop Agric Sci. 2013;36:79–88.

    Google Scholar 

  37. Moghadam MS, Foo HL, Leow TC, Rahim RA, Loh TC. Novel bacteriocinogenic Lactobacillus plantarum strains and their differentiation by sequence analysis of 16S rDNA , 16S-23S and 23S-5S intergenic spacer regions and randomly amplified polymorphic DNA analysis. Food Technol Biotechnol. 2010;48:476–83.

    CAS  Google Scholar 

  38. Tai HF, Foo HL, Abdul Rahim R, Loh TC, Abdullah MP, Yoshinobu K. Molecular characterisation of new organisation of plnEF and plw loci of bacteriocin genes harbour concomitantly in Lactobacillus plantarum I-UL4. Microb Cell Fact BioMed Central. 2015;14:89.

    Article  Google Scholar 

  39. Motevaseli E, Shirzad M, Akrami SM, Mousavi AS, Mirsalehian A, Modarressi MH. Normal and tumour cervical cells respond differently to vaginal lactobacilli, independent of pH and lactate. J Med Microbiol. 2013;62:1065–1072. -0.

    Article  Google Scholar 

  40. Wang K, Li W, Rui X, Chen X, Jiang M, Dong M. Characterisation of a novel exopolysaccharide with antitumor activity from Lactobacillus plantarum 70810. Int J Biol Macromol. 2014;63:133–9.

    Article  CAS  Google Scholar 

  41. Shahid M, Rajoka R, Zhao H, Lu Y, Lian Z, Li N, Hussain N, Shao D, Jin M, Li Q, Shi J. Anticancer potential against cervix cancer (HeLa) cell line of probiotic Lactobacillus casei and Lactobacillus paracasei strains isolated from human breast milk. Food Funct. 2018;9:2705–15.

    Article  Google Scholar 

  42. GLOBOCAN. Estimated Cancer incidence, mortality and prevalence Worlwide in 2012. Available from: http://globocan.iarc.fr/Pages/fact_sheets_population.aspx. Accessed 24 Dec 2017.

  43. Foo Hooi Ling, Loh Teck Chwen, Chuah Li Oon, Noorjahan Banu Alitheen RAR. Tumour cytotoxic agent and methods thereof. US 2016/0030492. 2016;

  44. Sze LY. Probiotic characteristics of bacteriocinogenic Lactobacillus plantarum strains isolated from Malaysian foods. 2010. Masters thesis. Universiti Putra Malaysia.

  45. Reddy KVR, Aranha C, Gupta SM, Yedery RD. Evaluation of antimicrobial peptide nisin as a safe vaginal contraceptive agent in rabbits: in vitro and in vivo studies. Reproduction. 2008;128:117–26.

    Article  Google Scholar 

  46. Palou A, Serra F, Pico C. General aspects on the assessment of functional foods in the European Union. Eur J Clin Nutr. 2003;57:S12–7.

    Article  CAS  Google Scholar 

  47. Nishio K, Ma J, Arora S, Kaur S, Kaur S. Bacteriocins as potential anticancer agents. Front Pharmacol. 2015;6:272.

    Google Scholar 

  48. Soa SSY, Wana MLY, El-Nezami H. Probiotics-mediated suppression of cancer. Curr Opin Oncol. 2017;29:62–72.

    Article  Google Scholar 

  49. Chumchalová J, Šmarda J. Human tumor cells are selectively inhibited by colicins. Folia Microbiol. 2003;48:111–5.

    Article  Google Scholar 

  50. Tan HK, Foo HL, Loh TC, Banu N, Alitheen M, Abdul Rahim R. Cytotoxic effect of proteinaceous postbiotic metabolites produced by Lactobacillus plantarum I-UL4 cultivated in different media composition on MCF-7 breast cancer cell. Malays J Microbiol. 2015;11:207–14.

    CAS  Google Scholar 

  51. Westmoreland C, Holmes AM. Assuring consumer safety without animals: applications for tissue engineering. Organogenesis. 2009;5:67–72.

    Article  Google Scholar 

  52. Aquino Esperanza JA, Aguirre MV, Aispuru GR, Lettieri CN, Juaristi JA, Alvarez MA, et al. In vivo 5-flourouracil-induced apoptosis on murine thymocytes: involvement of FAS, Bax and Caspase3. Cell Biol Toxicol. 2008;24:411–22.

    Article  CAS  Google Scholar 

  53. Potten CS, Grant HK. The relationship between ionizing radiation-induced apoptosis and stem cells in the small and large intestine. Br J Cancer. 1998;78:993–1003.

    Article  CAS  Google Scholar 

  54. Thirabunyanon M, Boonprasom P, Niamsup P. Probiotic potential of lactic acid bacteria isolated from fermented dairy milks on antiproliferation of colon cancer cells. Biotechnol Lett. 2009;31:571–6.

    Article  CAS  Google Scholar 

  55. Hirayama K, Rafter J. The role of probiotic bacteria in cancer prevention. Microbes Infect. 2000;2:681–6.

    Article  CAS  Google Scholar 

  56. Savitskiy VP, Shman TV, Potapnev MP. Comparative measurement of spontaneous apoptosis in pediatric acute leukemia by different techniques. Cytometry B Clin Cytom. 2003;56:16–22.

    Article  Google Scholar 

  57. Baskić D, Popović S, Ristić P, Arsenijević NN. Analysis of cycloheximide-induced apoptosis in human leukocytes: fluorescence microscopy using annexin V/propidium iodide versus acridin orange/ethidium bromide. Cell Biol Int. 2006;30:924–32.

    Article  Google Scholar 

  58. Krysko DV, Denecker G, Festjens N, Gabriels S, Parthoens E, D’Herde K, et al. Macrophages use different internalization mechanisms to clear apoptotic and necrotic cells. Cell Death Differ. 2006;13:2011–22.

    Article  CAS  Google Scholar 

  59. Festjens N, Vanden Berghe T, Vandenabeele P. Necrosis, a well-orchestrated form of cell demise: Signalling cascades, important mediators and concomitant immune response. Biochim Biophys Acta Bioenerg. 2006;1757:1371–87.

    Article  CAS  Google Scholar 

  60. Denecker G, Vercammen D, Declercq W, Vandenabeele P. Apoptotic and necrotic cell death induced by death domain receptors. Cell Mol Life Sci. 2001;58:356–70.

    Article  CAS  Google Scholar 

Download references

Acknowledgements

The authors would like to thank Ministry of Education Malaysia for funding the grants under FRGS and LRGS.

Funding

This work was supported by the Fundamental Research Grant Scheme (FRGS) and the Long-Term Research Grant Scheme (LRGS) of the Ministry of Education of Malaysia.

Author information

Authors and Affiliations

Authors

Contributions

LOC carried out the cytotoxicity, antiproliferation and AOPI studies and drafted the manuscript. HLF participated in the design, conceived and coordination of this study; and helped to draft and revised the manuscript. TCL participated in the design of the study and performed the statistical analysis. NBA participated in the design of the antiproliferation and AOPI study. SKY participated in the design and analysis of haemolysis and AOPI studies. NEAM helped to draft and revise the manuscript. RAR participated in the cytotoxicity study and helped to revise the manuscript. KY helped to draft and revised the manuscript. All authors read and approved the final manuscript.

Authors’ information

LOC was a PhD student who has conducted the cytotoxicity, antiproliferation and AOPI studies. HLF, TCL, NBA, SKY, NEAM, RAR and KY are collaborators of the research funding used for the experimental work presented in this report. LOC, HLF, TCL, NBA, NEAM, RAR and KY are researchers of UPM, while SKY is the researcher from Xiamen Malaysia University.

Corresponding author

Correspondence to Hooi Ling Foo.

Ethics declarations

Ethics approval and consent to participate

The in vitro protocols of animal and human cell experiments were conducted according to the guidelines of the Institutional Animal Care and Use Committee (IACUC) of UPM. The human blood was withdrawn from healthy donors by a nurse at the Health Center of UPM after consent was informed according to the protocol of IACUC of UPM.

Consent for publication

Not applicable.

Competing interests

The authors declare that they have no competing interests.

Additional information

Publisher’s Note

Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Rights and permissions

Open Access This article is distributed under the terms of the Creative Commons Attribution 4.0 International License (http://creativecommons.org/licenses/by/4.0/), which permits unrestricted use, distribution, and reproduction in any medium, provided you give appropriate credit to the original author(s) and the source, provide a link to the Creative Commons license, and indicate if changes were made. The Creative Commons Public Domain Dedication waiver (http://creativecommons.org/publicdomain/zero/1.0/) applies to the data made available in this article, unless otherwise stated.

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Chuah, LO., Foo, H.L., Loh, T.C. et al. Postbiotic metabolites produced by Lactobacillus plantarum strains exert selective cytotoxicity effects on cancer cells. BMC Complement Altern Med 19, 114 (2019). https://doi.org/10.1186/s12906-019-2528-2

Download citation

  • Received:

  • Accepted:

  • Published:

  • DOI: https://doi.org/10.1186/s12906-019-2528-2

Keywords