Está en la página 1de 30

Accepted Manuscript

Cholesterol-lowering effects of a putative probiotic strain Lactobacillus plantarum EM


isolated from kimchi
Eun A. Choi, Hae Choon Chang
PII:

S0023-6438(15)00035-3

DOI:

10.1016/j.lwt.2015.01.019

Reference:

YFSTL 4400

To appear in:

LWT - Food Science and Technology

Received Date: 30 June 2014


Revised Date:

2 January 2015

Accepted Date: 13 January 2015

Please cite this article as: Choi, E.A, Chang, H.C., Cholesterol-lowering effects of a putative probiotic
strain Lactobacillus plantarum EM isolated from kimchi, LWT - Food Science and Technology (2015),
doi: 10.1016/j.lwt.2015.01.019.
This is a PDF file of an unedited manuscript that has been accepted for publication. As a service to
our customers we are providing this early version of the manuscript. The manuscript will undergo
copyediting, typesetting, and review of the resulting proof before it is published in its final form. Please
note that during the production process errors may be discovered which could affect the content, and all
legal disclaimers that apply to the journal pertain.

ACCEPTED MANUSCRIPT

1
2

Cholesterol-lowering effects of a putative probiotic strain

Lactobacillus plantarum EM isolated from kimchi

RI
PT

4
5
6

Eun A Choi Hae Choon Chang*

SC

M
AN
U

8
9
10

Department of Food and Nutrition, Kimchi Research Center, Chosun University, 309

12

Pilmun-daero, Dong-gu, Gwangju 501-759, South Korea

13

TE
D

11

14

15

e-mail: hcchang@chosun.ac.kr

AC
C

EP

Corresponding author: Tel.: +82 62 2307345; fax: +82 62 2228086

ACCEPTED MANUSCRIPT

16

Abstract

17
Lactobacillus plantarum EM, which was isolated from kimchi, showed high cholesterol removal by

19

growing, resting, and even dead cells. Moreover, cell wall fraction of L. plantarum EM removed

20

cholesterol in a cell wall concentration-dependent pattern. Lactobacillus acidophilus ATCC 43121 as a

21

control showed high cholesterol removal by growing cells, whereas resting and dead cells showed less

22

cholesterol removal. Scanning electron micrographs showed that large amounts of cholesterol adhered to

23

surfaces of growing and dead cells of L. plantarum EM without changes in cell morphology. On the other

24

hand, only a small amount of cholesterol adhered to the surface of L. acidophilus ATCC 43121 cells, and

25

growing cells of L. acidophilus ATCC 43121 showed morphological changes with a thinner and longer

26

shape. Based on these results, cholesterol removal mechanisms by L. plantarum EM could be attributed to

27

enzymatic assimilation including bile salt hydrolase activity and cell surface-binding. Further, L.

28

plantarum EM showed reasonable tolerance to acid and bile stresses and displayed antagonistic activity

29

against pathogens. Moreover, L. plantarum EM did not represent a health risk due to antibiotic resistance.

30

The results of this study indicate that L. plantarum EM may be a promising probiotic candidate and

31

adjunct culture for reduction of serum cholesterol level regardless of its viability.

35

SC

M
AN
U

TE
D

34

EP

33

Keywords: Cholesterol-lowering effect, Lactobacillus plantarum, Probiotic, Kimchi

AC
C

32

RI
PT

18

ACCEPTED MANUSCRIPT

36

1. Introduction

37
Lactic acid bacteria (LAB) are industrially important microorganisms worldwide for the fermentation

39

of foods. Moreover, LAB as probiotics have become the focus of intensive international research for their

40

health-promotion effects, which include reduction of serum cholesterol level, stimulation of immune

41

responses, cancer prevention, and alleviation of diarrhea (De Vrese & Schrezenmeir, 2008). The

42

numerous health benefits of LAB have made them into promising probiotic candidates that are studied for

43

their desirable properties. The World Health Organization (WHO) has predicted that cardiovascular

44

disease will remain the leading cause of death through 2030, affecting approximately 23.6 million people

45

worldwide (WHO, 2009). Increased serum cholesterol correlates highly with coronary heart disease

46

(Kumar et al., 2012). Thus, investigators are paying close attention to the cholesterol-lowering effects of

47

LAB among their many functional effects.

M
AN
U

SC

RI
PT

38

Studies have indicated that several LAB strains, mainly Lactobacillus spp., have cholesterol-reducing

49

effects in vitro or in vivo (Kumar et al., 2012; Ooi & Liong, 2010). However, the exact mechanisms of

50

serum cholesterol reduction by probiotic bacteria are not completely clear. Several mechanisms have been

51

proposed in vitro, including assimilation (Pereira & Gibson, 2002), surface binding (Liong & Shah, 2005),

52

incorporation into cellular membranes (Lye, Rusul, & Liong, 2010a), co-precipitation with deconjugated

53

bile (Liong & Shah, 2006), enzymatic deconjugation of bile acids by bile salt hydrolase (BSH) (Lambert,

54

Bongers, de Vos, & Kleerebezem, 2008), conversion of cholesterol into coprostanol (Lye, Rusul, & Liong,

55

2010b), and production of short-chain fatty acids by probiotics (De Preter et al., 2007).

EP

TE
D

48

The aim of this study was to investigate high cholesterol removal by dead cells of the selected LAB

57

strain as well as its possible mechanisms of action. Moreover, the selected strain was identified and

58

evaluated in terms of its probiotic properties, such as acid and bile tolerance, antimicrobial activity

59

against pathogens, and antibiotic resistance.

AC
C

56

60
61

2. Materials and methods

62
63

2.1. Bacterial cultures and screening of LAB strains for cholesterol removal

64
3

ACCEPTED MANUSCRIPT

65
66

Screening of LAB strains from kimchi for cholesterol removal was carried out by determination of
BSH activity (Dashkevicz & Feighner, 1989) and cholesterol assimilation (Rudel & Morris, 1973).

67
68

2.1.1. BSH activity

RI
PT

69

For BSH activity measurement, 10 l of culture grown in MRS broth was spotted onto BSH screening

71

medium, which consisted of de Man Rogosa and Sharpe (MRS; Difco, Sparks, MD, USA) agar

72

supplemented with 0.5% (w/v) sodium salt taurodeoxycholic acid (TDCA, Sigma-Aldrich, St Louis, MO,

73

USA) and 0.37 g/l of CaCl2 (Dashkevicz & Feighner, 1989). Plates were incubated anaerobically at 37 C

74

using GasPak EZ anaerobe container systems (Becton, Dickinson and Company, Sparks, MD, USA),

75

after which BSH activity was determined by measuring the diameters of precipitation zones. BSH activity

76

was expressed based on the diameters of precipitation zones on BSH screening medium: -, no

77

precipitation; +, precipitation zone of up to 10 mm; ++, precipitation zone of up to 15 mm; and +++,

78

precipitation zone of up to 20 mm.

80
81

TE
D

79

M
AN
U

SC

70

2.1.2. Cholesterol assimilation

Cholesterol assimilation was determined using the method of Rudel and Morris (Rudel & Morris,

83

1973). LAB cells grown overnight were inoculated (1%) and cultivated anaerobically using GasPak EZ

84

anaerobe container systems (Becton, Dickinson and Company) at 37 C in MRS broth supplemented with

85

0.5% (w/v) oxgall (Sigma-Aldrich) and 0.1 g/l of water-soluble cholesterol (Sigma-Aldrich). Following

86

incubation, cells were harvested (9,950 g, 5 min, 4 C), after which 1 ml of supernatant was added to

87

2 ml of 33% (w/v) potassium hydroxide and 3 ml of 95% (v/v) ethanol. The mixture was shaken well for

88

1 min and then heated for 10 min in a 60 C water bath. After cooling in cold water, 5 ml of hexane was

89

added, followed by mixing and addition of 1 ml of distilled water. The tube was allowed to stand for 10

90

min at room temperature for phase separation, after which 3 ml of the hexane phase was transferred to a

91

clean tube. The hexane phase was then evaporated under a nitrogen stream. The concentrated phase was

92

added to 4 ml of freshly prepared O-phthalaldehyde (Sigma-Aldrich, 0.5 mg of O-phthalaldehyde/ml of

AC
C

EP

82

ACCEPTED MANUSCRIPT

93

acetic acid), mixed, and permitted to stand at room temperature for 10 min. Following the addition of

94

2 ml of concentrated sulfuric acid and incubation for 10 min, the absorbance at 550 nm was read using a

95

spectrophotometer (Amersham Biosciences, Uppsala, Sweden). Absorbance values were compared to

96

those obtained using cholesterol standard.

98

RI
PT

97
2.2. Identification of isolate

99

The LAB strain showing the highest BSH activity and cholesterol assimilation during measurement of

101

cholesterol removal was identified for further study. Identification was carried out by Gram staining,

102

catalase test, and morphological observation under a microscope, and 16S rRNA gene sequences were

103

determined using an API prism 3730 DNA analyzer (Applied Biosystems, Foster City, CA, USA)

104

according to the method described by Yang and Chang (Yang & Chang, 2008). The determined 16S rRNA

105

gene sequences were compared with sequences available in the GenBank database using the Clustal W

106

program (Thompson, Higgins, & Gibson, 1994).

M
AN
U

SC

100

108

TE
D

107
2.3. Growth in bile acids and cholesterol

109

Growth of LAB under bile acids and cholesterol was determined. LAB grown overnight in MRS were

111

inoculated (1%) into MRS broth supplemented with 0.3% (w/v) oxgall and 0.1 g/l of water-soluble

112

cholesterol, 0.5% (w/v) oxgall and 0.1 g/l of water-soluble cholesterol, 0.3% (w/v) TDCA and 0.1

113

g/l of water-soluble cholesterol, and 0.5% (w/v) TDCA and 0.1 g/l of water-soluble cholesterol.

114

Growth of LAB in MRS broth without bile was used as a control. Initial pHs of the prepared media were

115

pH 6.2-6.4. The culture was incubated anaerobically for 24 h at 37 C, after which the absorbance at 600

116

nm was measured.

AC
C

EP

110

117
118

2.4. Acid and bile tolerance

119
120

Tolerance levels of LAB to acid and bile salts were determined as previously described (Ryu & Chang,
5

ACCEPTED MANUSCRIPT

2013). LAB were cultivated at 37 C for 24 h in MRS broth and harvested by centrifugation (9,950 g, 5

122

min, 4 C), after which cell pellets were resuspended in phosphate-buffered saline (PBS, pH 2.5; Hyclone,

123

Logan, UT, USA), simulated gastric juice (SGJ; 3 mg of pepsin dissolved in 1 ml of 0.5% saline buffer,

124

pH 2.5), or bile salt (0.3% oxgall dissolved in PBS, pH 8.0) with approximately 9.8 log CFU/ml of cell

125

numbers. The suspensions were incubated at 37 C for 1 h in PBS or SGJ for 3 h in bile salt. Thereafter,

126

suspensions were harvested (9,950 g, 5 min, 4 C) and resuspended in MRS broth, after which viable

127

cell counts were enumerated on MRS agar after incubation at 37 C for 48 h. LAB suspended in MRS

128

broth without acid or bile salt were considered as controls.

SC

129
2.5. Antibiotic susceptibility

M
AN
U

130

RI
PT

121

131

Antibiotic susceptibility of the selected LAB isolate was determined according to the technical

133

guidelines of the European Food Safety Authority (EFSA) (EFSA, 2012) using a previously described

134

method (Ryu & Chang, 2013). The minimal inhibitory concentrations (MIC) of antibiotics were

135

measured. After culturing LAB in MRS broth for 24 h, cells were centrifuged and resuspended in Muller

136

Hinton (MH; Difco) broth containing 0.5% dextrose. Resultant cell suspensions were diluted in the same

137

medium to a cell density of 5.0 log CFU/ml (A600=0.4-0.5). Each antibiotic was added to the diluted cell

138

suspension, which was incubated anaerobically at 37 C for 24-48 h. Cell growth was observed visually

139

and measured based on absorbance at 600 nm (Amersham Biosciences). MIC values were defined as the

140

lowest antibiotic concentrations where no bacterial growth occurred. All antibiotics were purchased from

141

Sigma-Aldrich.

143
144

EP

AC
C

142

TE
D

132

2.6. Antimicrobial activity

145

LAB grown overnight were inoculated (1%) into MRS broth and incubated anaerobically at 37 C for

146

24 h. The culture was centrifuged, after which the supernatant was membrane-filtered (0.4 m membrane

147

filter; Advantec MFS, Dublin, CA, USA) and then freeze-dried (Samwon, Busan, South Korea). The

ACCEPTED MANUSCRIPT

148

freeze-dried culture supernatant was concentrated (5-fold) in 20 mM sodium acetate (pH 4.0).

149

Concentrated (5-fold) MRS broth was used as a control.


Antimicrobial activities against seven pathogens were investigated using spot-on-the-lawn assay

151

(Hoover & Harlander, 1993). Pathogens were grown at 37 C on Luria-Bertani (LB; Difco) agar for

152

E.coli, Pseudomonas aeruginosa, and Salmonella enterica serovar Typhi, nutrient agar (NA; Difco)

153

supplemented with 2% (w/v) NaCl for Vibrio parahaemolyticus, and tryptic soy agar (TSA; Difco) for

154

Bacillus cereus, Micrococcus luteus, and Staphylococcus aureus. ATCC strains were purchased from the

155

American Type Culture Collection (Manassas, VA, USA), whereas KCTC strains were purchased from

156

the Korean Collection for Type Cultures (Daejeon, South Korea). LB, TSA, or NA plates were spread

157

with each pathogen at a concentration of 6 log CFU/ml. An aliquot (10 l) of LAB culture was spotted

158

onto sensitive pathogen plates for each antimicrobial sample. Antimicrobial activity, expressed as

159

arbitrary units (AU) per milliliter, was defined as the reciprocal of the highest dilution at which pathogen

160

growth was inhibited (Ammor, Tauveron, Dufour, & Cherallier, 2006). The antimicrobial titer was

161

calculated as (1000/d) D, where D is the dilution factor and d is the dose (amount of antimicrobial

162

samples pipetted onto each spot). The above experiment was performed in triplicate.

164
165

SC

M
AN
U

TE
D

163

RI
PT

150

2.7. Cholesterol removal by growing, resting, and dead cells

For cholesterol assimilation by growing cells, LAB cells were inoculated (1%) and grown

167

anaerobically at 37 C in MRS broth supplemented with 0.5% (w/v) oxgall and 0.1 g/l of water-soluble

168

cholesterol as well as in MRS broth containing 0.5% (w/v) TDCA and 0.1 g/l of water-soluble cholesterol.

169

To prepare resting and dead cells (Lye, Rusul, & Liong, 2010a), LAB strains were incubated

170

anaerobically at 37 C in MRS broth containing 0.5% oxgall as well as in MRS broth containing 0.5%

171

TDCA. The cell pellet obtained from the media was washed twice with sterile-distilled water. For

172

cholesterol removal by resting cells, the cell pellet was suspended in 0.05 M phosphate buffer (pH 6.8)

173

containing 0.5% oxgall and 0.1 g/l of water-soluble cholesterol as well as in 0.5% TDCA and 0.1 g/l of

174

water-soluble cholesterol. For dead cell assay, the cell pellet was suspended in saline and heat-killed at

175

121 C for 15 min. The dead cells were harvested, after which the pellet was suspended in MRS

AC
C

EP

166

ACCEPTED MANUSCRIPT

176

containing 0.5% oxgall and 0.1 g/l of water-soluble cholesterol as well as in 0.5% TDCA and 0.1 g/l

177

water-soluble cholesterol.
All strains were incubated at 37 C for 24 h, after which the mixture was centrifuged (9,950 g, 5 min,

179

4 C). The remaining cholesterol concentration of broth was determined using the method of Rudel and

180

Morris (Rudel & Morris, 1973) described in 2.1.1. Cholesterol assimilation (Ooi & Liong, 2010) was

181

expressed as {0.1 g/l of cholesterol (control) concentration of remaining cholesterol in cultures}/control

182

100.

RI
PT

178

184

SC

183
2.8. Cholesterol removal by LAB cell wall

186

2.8.1. Preparation of cell wall

187

M
AN
U

185

Isolation of LAB cell wall was carried out as previously described with modifications (Piuri, Sanchez-

189

Rivas, & Ruzal, 2005). Exponential phase of LAB cultures (20 ml) was harvested, washed twice with

190

PBS (pH 7.0-7.2, Hyclone), and suspended in 20 ml of PBS at 4 C. Ice-jacketed cell suspensions were

191

sonicated (Amp 60, 10 min, pulse 2 sec; Sonics, Newton, CT, USA) twice, centrifuged, suspended in 5%

192

(w/v) sodium dodecyl sulfate (SDS, Roche, Indianapolis, IN, USA), and then incubated at 60 C for 30

193

min. The crude cell wall fraction was centrifuged (9,950 g, 30 min, 4 C), washed twice with PBS,

194

suspended in 10 mM Tris-HCl (pH 7.5) with 10 mM MgCl2, and subsequently incubated with a mixture

195

of 0.01 g/ml of DNase (Sigma-Aldrich), 0.01 mg/ml of RNase A (Sigma-Aldrich), and 0.01 g/ml of

196

trypsin (Sigma-Aldrich) for 4 h at 37 C. Finally, the cell wall fraction was centrifuged, washed three

197

times with PBS, suspended in 20 ml of PBS, and then homogenized twice by sonication (Amp 60, 10 min,

198

pulse 2 sec). The purified cell wall fraction was then freeze-dried.

200

EP

AC
C

199

TE
D

188

2.8.2. Cholesterol removal by LAB cell wall fraction

201
202

The prepared cell wall fraction was suspended in MRS containing 0.5% TDCA and 0.1 g/l of water-

203

soluble cholesterol along with A600=1, 2, 3, 4, and 5 mg/ml of the LAB cell wall fraction. The suspension
8

ACCEPTED MANUSCRIPT

204

was incubated anaerobically at 37 C for 24 h, after which the mixture was centrifuged (9,950 g, 30 min,

205

4 C). The remaining cholesterol concentration of the supernatant was measured using the method of

206

Rudel and Morris (Rudel & Morris, 1973), and its cholesterol assimilation (%) was determined.

208

RI
PT

207
2.9. Scanning electron microscopy (SEM)

Growing and dead cells as well as cell wall fraction were prepared as described in sections 2.7 and 2.8.

210

The prepared growing and dead cells (A6008.0) as well as cell wall fraction (A60012.0) were suspended

211

in MRS broth supplemented with 0.5% TDCA and 0.1 g/l of cholesterol incubated at 37 C for 24 h. The

212

prepared LAB cultures in MRS broth without any supplementation were used as a control. After

213

incubation, LAB cultures were centrifuged and then washed twice with PBS (pH 7.0-7.2, Hyclone).

M
AN
U

SC

209

Cholesterol attached to LAB cells or cell wall fraction was observed using field emission scanning

215

electron microscopy (FE-SEM; Hitachi, Tokyo, Japan) as described previously with slight modifications

216

(Kalchayanand, Dunne, Sikes, & Ray, 2004). LAB cell or cell wall fraction was prefixed with 0.1 M

217

sodium cacodylate buffer (pH 7.4) containing 2.5% (v/v) glutaldehyde (pH 7.4; Sigma-Aldrich) for 2 h at

218

4 C, followed by washing three times with 0.1 M sodium cacodylate buffer (pH 7.4). The samples were

219

then postfixed with 1% (w/v) osmium tetroxide in 0.1 M sodium cacodylate buffer (pH 7.4) for 1-2 h,

220

washed three times with 0.1 M sodium cacodylate buffer (pH 7.4), and dehydrated in an ascending

221

ethanol series [(%, v/v): 50, 60, 70, 80, 90, 95, and 100 ethanol] for 2 min each. Finally, the samples were

222

immersed two times in 100% tert-butanol for 20 min and then coated with platinum using a sputter coater

223

(E-1030 Ion sputter, Hitachi, Tokyo, Japan). The resultant chips were viewed in a FE-SE microscope.

225
226

EP

AC
C

224

TE
D

214

2.10. Statistical analysis

227

Data are presented as the means and standard deviations (means SD) of three independent

228

experiments performed in duplicate or triplicate. Statistical analysis was performed on the data using

229

Duncans Multiple Range Test (DMRT) by SPSS ver. 21.0 for Windows (SPSS, Chicago, IL, USA) with

230

statistical significance determined at p < 0.05.

ACCEPTED MANUSCRIPT

231
232

3. Results and discussion

233
234

3.1. Isolation and identification of cholesterol-lowering LAB

RI
PT

235

Seventy LAB cultures were isolated from 28 kimchi samples collected from 17 cities in South Korea.

237

All isolates were grown well at 30 C in MRS medium. However, only 50 LAB strains among the 70 total

238

LAB strains could be grown at 37 C. These 50 LAB strains were preselected for further experimentation.

239

BSH activity and cholesterol assimilation of the 50 preselected LAB strains were examined (Table 1). Of

240

the 50 tested LAB strains, 16 LAB strains expressed BSH activity, and their cholesterol assimilation

241

varied from 3.51-80.86%. Based on this result, we selected two strains (EM and DC1) for further

242

experimentation. Among the 50 strains, EM showed the strongest cholesterol assimilation (80.86%) and

243

BSH activity (17.2 mm precipitation zone), whereas DC1 showed the weakest cholesterol assimilation

244

(3.51%) and BSH activity (5.3 mm precipitation zone).

M
AN
U

SC

236

The EM and DC1 strains were Gram-positive, rod-shaped, and catalase-negative. When 16S rRNA

246

gene sequences of the EM (1,881 bp) and DC1 (1,471 bp) isolates were determined and compared with

247

those of type strains in GenBank, EM and DC1 sequences showed 99.9% and 99.9% homologies with

248

those of L. plantarum JCM 1149T and L. sakei DSM 20017T, respectively. Thus, EM and DC1 isolates

249

were finally designated as L. plantarum EM and Lactobacillus sakei DC1, respectively, and their gene

250

sequences were deposited in GenBank (accession numbers KC 422389 for EM and KJ 812206 for DC1).

252
253

EP

AC
C

251

TE
D

245

3.2. Acid and bile tolerance

254

It is well known that probiotic bacteria should be capable of surviving passage through the

255

gastrointestinal tract based on acid tolerance to human gastric juice as well as bile tolerance for survival

256

in the small intestine (Saarela, Mogensen, Fondn, Mtt, & Mattila-Sandholm, 2000). Therefore, these

257

characteristics may serve as suitable criteria for probiotic bacteria selection. L. plantarum EM showed

258

high tolerance to acidic conditions at pH 2.5 (PBS and SGJ) after 1 h of exposure at 37 C (Table 2).

10

ACCEPTED MANUSCRIPT

When L. plantarum EM was added to PBS containing 0.3% oxgall (pH 8.0) for 3 h at 37 C, initial cell

260

counts (9.78 log CFU/ml) of L. plantarum EM decreased to 5.53 log CFU/ml. In this study, acid and bile

261

treatments to L. plantarum EM were carried out under nutritional deficit. Considering the human

262

gastrointestinal tract is full of food, the actual survival of L. plantarum EM in the gastrointestinal tract

263

will actually be greater than the result shown in Table 2. This suggests that L. plantarum EM has

264

reasonable tolerance to acid and bile and should be active in the gastrointestinal tract.

RI
PT

259

265
3.3. Antibiotic resistance

SC

266
267

L. plantarum EM was assessed for its resistance to nine antibiotics, including those highlighted by the

269

EFSA (Florez et al., 2006). As shown in Table 3, L. plantarum EM was susceptible to all of the antibiotics

270

tested, except vancomycin. The technical guidelines of the EFSA have demonstrated that no breakpoints

271

for vancomycin and streptomycin are required for L. plantarum (EFSA, 2012). Therefore, it seems

272

reasonable to conclude that consumption of L. plantarum EM in this study does not represent a health risk

273

to humans due to antibiotic resistance. It is recognized that probiotics lack undesirable properties such as

274

antibiotic resistance (Saarela, Mogensen, Fondn, Mtt, & Mattila-Sandholm, 2000; Florez et al., 2006).

275

Thus, any evaluation of new probiotic candidates, including those traditionally used in food fermentation,

276

should carefully confirm their safety status.

279

TE
D

EP

278

3.4. Antimicrobial activity

AC
C

277

M
AN
U

268

280

L. plantarum EM showed strong activities against the seven tested pathogens (Table 4). In particular, L.

281

plantarum EM showed its strongest activity against Vibrio parahaemolyticus ATCC 17802 (25,600 units)

282

as well as its weakest activity against Staphylococcus aureus ATCC 29123 (200 units). L. plantarum EM

283

showed a broad antimicrobial spectrum against Gram-positive and Gram-negative pathogens (Table 4).

284

Antimicrobial substances produced from LAB are known to include H2O2, CO2, lactic and acetic acids,

285

bacteriocin, and others (Usman & Hosono, 1999). However, most of these antimicrobial substances from

286

LAB strains have a narrow antimicrobial spectrum, and thus their inhibitory effects are only against

287

closely related species (Holzapfel, Geisen, & Schillinger, 1995). One of the most important functional
11

ACCEPTED MANUSCRIPT

288

requirements of probiotics is a broad antimicrobial spectrum as well as antagonism against pathogenic

289

bacteria with strong antimicrobial activity (Saarela, Mogensen, Fondn, Mtt, & Mattila-Sandholm,

290

2000). Thus, the results represented in Table 4 for L. plantarum EM fulfill the beneficial requirements of

291

probiotics.

293

RI
PT

292
3.5. Growth in the presence of bile acid

294

Growth in the presence of bile acid (oxgall or TDCA) was examined using L. plantarum EM and L.

296

sakei DC1 (Table 5). L. acidophilus ATCC 43121, which is characterized by high cholesterol assimilation

297

(Gilliand & Walker, 1989; Gilliand, Nelson, & Maxwell, 1985), was used as a control strain. Generally,

298

all three strains grew well in the presence of both bile acids. However, growth of the three LAB strains

299

slightly decreased in TDCA- or oxgall-containing media. Oxgall showed a slightly higher toxic effect on

300

LAB growth than TDCA. The antimicrobial action of bile is well known; bile salt at high concentrations

301

can rapidly dissolve membrane lipids and cause dissociation of integral membrane proteins, resulting in

302

leakage of cell contents and cell death (Begley, Gahan, & Hill, 2005). The results represented in Table 5

303

suggest that the three LAB strains are moderately resistant to bile toxicity.

304

307
308

EP

306

3.6. Cholesterol removal

3.6.1. Cholesterol removal by growing, resting, and dead cells

AC
C

305

TE
D

M
AN
U

SC

295

309

As shown in Table 6, cholesterol removal according to cell state was in the order of growing, resting,

310

and dead cells. L. plantarum EM showed the highest cholesterol removal among the three tested strains

311

regardless of cell state. As already shown in Table 5, L. sakei DC1 showed approximately 1/2 growth

312

(A6005) compared to the other two LAB strains (A6008-10). However, cholesterol removal of L. sakei

313

DC1 was much lower (1/3-1/30) than that of L. plantarum EM or L. acidophilus ATCC 43121 (Table 6).

314

Cholesterol removal by growing cells of L. acidophilus ATCC 43121 was as high as that of L. plantarum

315

EM. However, cholesterol removal by dead cells of L. acidophilus ATCC 43121 was significantly (p <
12

ACCEPTED MANUSCRIPT

316

0.05) lower than that of L. plantarum EM.

317
318

3.6.2. Cholesterol removal by LAB cell wall fraction and SEM observation

319
To determine the reason for such high cholesterol removal by dead cells of L. plantarum EM,

321

cholesterol removal by cell wall fractions of both L. plantarum EM and L. acidophilus ATCC 43121 was

322

investigated. As shown in Fig. 1, the cell wall fraction of L. plantarum EM removed cholesterol in a cell

323

wall concentration-dependent pattern. On the other hand, the cell wall fraction of L. acidophilus ATCC

324

43121 showed significantly (p < 0.05) lower cholesterol removal than that of L. plantarum EM. This

325

result indicates that cholesterol can be removed by the cell wall fraction itself without any metabolic

326

process.

M
AN
U

SC

RI
PT

320

Thus, we tried to observe cholesterol attachment to LAB cells according to cell state (growing, dead

328

cell, and cell wall fraction) using SEM. As shown in Fig. 2, large amounts of cholesterol adhered to the

329

surfaces of growing and dead cells of L. plantarum EM. Adherence of cholesterol to the cell wall fraction

330

was observed only with L. plantarum EM, whereas no cholesterol adherence or alteration of cell

331

morphology could be observed with L. sakei DC1. On the other hand, only small amounts of cholesterol

332

adhered to surfaces of growing and dead cells of L. acidophilus ATCC 43121. Interestingly, L.

333

acidophilus ATCC 43121 cells underwent morphological changes in a growing state (Fig. 2 - growing cell

334

B), as evidenced by a longer and thinner cell shape (arrow indicated) compared to control growing cells

335

of L. acidophilus ATCC 43121 (Fig. 2 - growing cell A). Further, dead cells of L. acidophilus ATCC

336

43121 did not show any morphological changes. Noh et al. previously reported the incorporation of

337

cholesterol into the cellular membrane of L. acidophilus ATCC 43121 in the presence of oxgall and

338

cholesterol (Noh, Kim, & Gilliand, 1997). Namely, a portion of the assimilated cholesterol was recovered

339

in the membrane fractions of cells grown in media containing cholesterol, which suggests that cholesterol

340

may have altered the cellular membrane or wall of L. acidophilus ATCC 43121.

AC
C

EP

TE
D

327

341

Cholesterol removal (%) by growing cells was almost the same between L. plantarum EM and L.

342

acidophilus ATCC 43121 (Table 6). However, examination of cholesterol adherence onto these two LAB

343

using SEM showed that their configurations were quite different. These data indicate that the cholesterol

344

removal mechanism of L. plantarum EM is different from that of L. acidophilus ATCC 43121. As


13

ACCEPTED MANUSCRIPT

reported already by Noh et al. (Noh, Kim, & Gilliand, 1997), incorporation of cholesterol into the cellular

346

membrane of L. acidophilus ATCC 43121 during growth might result in morphological changes, as

347

represented in this study (Fig. 2). On the other hand, Hosono and Tano-oka reported cholesterol binding

348

to LAB varies among strains and species and hypothesized that these differences in binding abilities can

349

be attributed to chemical and structural properties of cell wall peptidoglycans containing amino acids

350

capable of binding to cholesterol (Hosono & Tano-oka, 1995). Thus, strong cholesterol attachment to L.

351

plantarum EM regardless of its cell state might be due to the unique in chemical and structural properties

352

of the L. plantarum EM cell wall compared to those of other LAB cell types. However, direct

353

investigation of the chemical and structural properties of the L. plantarum EM cell wall that promote

354

binding to cholesterol is required. Cholesterol removal by dead cells of LAB strains has been reported

355

previously, even though the degree of removal is significantly less compared to that of growing cells

356

(Zeng, Pan, & Guo, 2010; Liong & Shah, 2005; Mahdieh, Hamid, & Naheed, 2014; Sirilun, Chaiyasut,

357

Kantachote, & Luxananil, 2010; Emami & Bazargani, 2014). Further, cholesterol removal by dead cells

358

due to cholesterol attachment to the cellular membrane has been investigated based on scanning electron

359

microscopic observation of cholesterol binding to growing cells (Lye, Rusul, & Liong, 2010a; Ooi &

360

Liong, 2010). To the best of our knowledge, there is no direct evidence verifying cholesterol removal by

361

dead cells via binding of cholesterol to dead cells or the cell wall fraction itself.

TE
D

M
AN
U

SC

RI
PT

345

Based on the results in Table 6 and Fig. 2, the cholesterol-lowering effect of L. plantarum EM can be

363

considered to be a major part of enzymatic assimilation, including BSH activity. Further, L. plantarum

364

EM showed a cholesterol-lowering effect with strong binding capability to cell surfaces regardless of its

365

viability. Cholesterol bound to LAB strains has been shown to inhibit intestinal absorption of cholesterol

366

in previous investigations (Usman & Hosono, 1999). Thus, the high cholesterol-binding capability of L.

367

plantarum EM might be more effective in reducing the serum cholesterol level in humans. However, in

368

vivo evaluation of this strain in humans is required to assess the usage of probiotics for reducing serum

369

cholesterol.

AC
C

EP

362

370
371

4. Conclusions

372
373

As the term probiotics means for life, viable probiotics are required to have beneficial effects on their
14

ACCEPTED MANUSCRIPT

host. In this study, L. plantarum EM showed high cholesterol removal by growing, resting, and even dead

375

cells based on the high cholesterol-binding capacity of its cell wall fraction. Therefore, L. plantarum EM

376

could be a potent probiotic to reduce serum cholesterol regardless of its viability. Moreover, L. plantarum

377

EM also appeared to meet the functional criteria required for beneficial probiotics, such as acid and bile

378

tolerance, antagonistic activity against pathogens, and antibiotic susceptibility.

RI
PT

374

379
380

SC

381
382

M
AN
U

383
384
385

Acknowledgment

386

TE
D

389

Agriculture, Food and Rural Affairs, South Korea.

EP

388

This research was supported by the Technology Development Program for Food, Ministry of

AC
C

387

15

ACCEPTED MANUSCRIPT

390

References

391
Ammor, S., Tauveron, G., Dufour, E., & Cherallier, I. (2006). Antibacterial activity of lactic acid bacteria

393

against spoilage and pathogenic bacteria isolated from the same meat small-scale facility 1-

394

Screening and characterization of the antibacterial compounds. Food Control, 17, 454-461.

396
397
398

Begley, M., Gahan, C. G., & Hill, C. (2005). The interaction between bacteria and bile. FEMS
Microbiology Reviews, 29, 625-651.

Dashkevicz, M. P., & Feighner, S. D. (1989). Development of a differential medium for bile salt

SC

395

RI
PT

392

hydrolase-active Lactobacillus spp. Applied and Environmental Microbiology, 55, 11-16.


De Preter, V., Vanhoutte. T., Huys, G., Swings, J., De Vuyst, L., Rutgeerts, P., et al. (2007). Effects of

400

Lactobacillus casei shirota, Bifidobacterium breve, and oligofructose-enriched inulin on colonic

401

nitrogen-protein metabolism in healthy humans. American Journal of Physiology - Gastrointestinal

402

and Liver Physiology, 292, G358-G368.

404
405
406

De Vrese, M., & Schrezenmeir, J. (2008). Probiotics, prebiotics, and synbiotics. Advances in Biochemical
Engineering Biotechnology, 111, 1-66.

TE
D

403

M
AN
U

399

EFSA (2012). Guidance on the assessment of bacterial susceptibility to antimicrobials of human or


veterinary importance. EPSA Journal, 10, 27-40.
Emami, A., & Bazargani, A. (2014). Dual effects of Lactobacilli as a cholesterol assimilator and an

408

inhibitor of gastrointestinal pathogenic bacteria. International Journal of Enteric Pathogens, 2,

409

e15768

411
412
413
414
415
416

Florez, A. B., Egervrn, M., Danielsen, M., Tosi, L., Morelli, L., Lindgren, S., et al. (2006). Susceptibility

AC
C

410

EP

407

of Lactobacillus plantarum strains to six antibiotics and definition of new susceptibility-resistance


cut-off values. Microbial Drug Resistance, 12, 252-256.

Gilliand, S. E., Nelson, C. R., & Maxwell, C. (1985). Assimilation of cholesterol by Lactobacillus
acidophilus. Applied and Environmental Microbiology, 49, 377-381.
Gilliand, S. E., & Walker, D. K. (1989). Cholesterol uptake by culture of Lactobacillus acidophilus used
for nonfermented acidophilus milk. Animal Science Papers and Reports, 11, 258-262.

16

ACCEPTED MANUSCRIPT

417

Holzapfel, W. H., Geisen, R., & Schillinger, U. (1995). Biological preservation of food with reference to

418

protective cultures, bacteriocins and food-grade enzymes. International Journal of Food

419

Microbiology, 24, 343-362.


Hoover, D. G., & Harlander, S. K. (1993). Screening methods for detecting bacteriocin activity. In: D. G.

421

Hoover, & L. R. Steenson (Eds.), In Bacteriocins of lactic acid bacteria (pp.23-29). San Diego:

422

Academic press

424

Hosono, A., & Tono-oka, T. (1995). Binding of cholesterol with lactic acid bacteria cells.
Milchwissenschaft, 50, 556-560.

SC

423

RI
PT

420

Kalchayanand, N., Dunne, P., Sikes, A., & Ray, B. (2004). Viability loss and morphology change of

426

foodborne pathogens following exposure to hydrostatic pressures in the presence and absence of

427

bacteriocin. International Journal of Food Microbiology, 91, 91-98.

M
AN
U

425

428

Kumar, M., Nagpal, R., Kumar, R., Hemalatha, R., Verma, V., Kumar, A., et al. (2012). Cholesterol-

429

lowering probiotics as potential biotherapeutics for metabolic diseases. Experimental Diabetes

430

Research, 2012, 1-14.

Lambert, J. M., Bongers R. S., de Vos, W. M., & Kleerebezem, M. (2008). Functional analysis of four

432

bile salt hydrolase and penicillin acylase family members in Lactobacillus plantarum WCFS1.

433

Applied and Environmental Microbiology, 74, 4719-4726.

436
437

Lactobacilli strains. Journal of Dairy Science, 88, 55-66.

EP

435

Liong, M. T., & Shah, N. P. (2005). Acid and bile tolerance and cholesterol removal ability of

Liong, M. T., & Shah, N. P. (2006). Effects of a Lactobacillus casei synbiotic on serum lipoprotein,
intestinal microflora, and organic acids in rats. Journal of Dairy Science, 89, 1390-1399.

AC
C

434

TE
D

431

438

Lye, H. S., Rusul, G., & Liong, M. T. (2010a). Mechanisms of cholesterol removal by lactobacilli under

439

conditions that mimic the human gastrointestinal tract. International Dairy Journal, 20, 169-175.

440
441

Lye, H. S., Rusul, G., & Liong, M. T. (2010b). Removal of cholesterol by Lactobacilli via incorporation
of and conversion to coprostanol. Journal of Dairy Science, 93, 1383-1392.

442

Mahdieh, I., Hamid, E., & Naheed, M. (2014). Antibacterial activity and cholesterol assimilation of lactic

443

acid bacteria isolated from traditional Iranian dairy products. LWT-Food Science and Technology, 58,

444

355-359.

17

ACCEPTED MANUSCRIPT

445
446
447
448

Noh, D. O., Kim, S. H., & Gilliand, S. E. (1997). Incorporation of cholesterol into the cellular membrane
of Lactobacillus acidophilus ATCC 43121. Journal of Dairy Science, 80, 3107-3113.
Ooi, L. G., & Liong, M. T. (2010). Cholesterol-lowering effects of probiotics and prebiotics: a review of
in vivo and in vitro findings. International Journal of Molecular Sciences, 11, 2499-2522.
Pereira, D. I. A., & Gibson, G. R. (2002). Cholesterol assimilation by lactic acid bacteria and

450

Bifidobacteria isolated from the human gut. Applied and Environmental Microbiology, 68, 4689-

451

4693.

454
455
456
457
458
459

SC

453

Piuri, M., Sanchez-Rivas, C., & Ruzal, S. M. (2005). Cell wall modifications during osmotic stress in
Lactobacillus casei. Journal of Applied Microbiology, 98, 84-95.

Rudel, L. L., & Morris, M. D. (1973). Determination of cholesterol using O-phthalaldehyde. Journal of
Lipid Research, 14, 364-366.

M
AN
U

452

RI
PT

449

Ryu, E. H., & Chang, H. C. (2013). In vitro study of potentially probiotic lactic acid bacteria strains
isolated from kimchi. Annals of Microbiology, 63, 1387-1395.

Saarela, M., Mogensen, G., Fondn, R., Mtt, J., & Mattila-Sandholm, T. (2000). Probiotic bacteria:
Safety, functional and technological properties. Journal of Biotechnology, 84, 197-215.
Sirilun, S., Chaiyasut, C., Kantachote, D., & Luxananil, A. (2010). Characterisation of non human origin

461

probiotic Lactobacillus plantarum with cholesterol-lowering property. African Journal of

462

Mcirobiology Research, 4, 994-1000.

TE
D

460

Thompson, J. D., Higgins, D. G., & Gibson, T. J. (1994). CLUSTAL W: improving the sensitivity of

464

progressive multiple sequence alignment through sequence weighting, position-specific gap

465

pantalties and weight matrix choice. Nucleic Acids Research, 22, 4673-4680.

AC
C

EP

463

466

Usman, & Hosono, A. (1999). Viability of Lactobacillus gasseri and its cholesterol-binding

467

antimutagenic activities during subsequent refrigerated storage in nonfermented milk. Journal of

468
469
470
471
472
473

Dairy Science, 82, 2536-2542.

WHO. (2009). Cardiovascular Disease (CVDs). Fact sheet N 317. http://www.who.int/mediacentre/


factsheets/fs317/en/print.html.
Yang, E. J., & Chang, H. C. (2008). Antifungal activity of Lactobacillus plantarum isolated from kimchi.
Korean Journal of Microbiology and Biotechnology, 36, 276-284.
Zeng, X. Q., Pan, D. D., & Guo, Y. X. (2010). The probiotic properties of Lactobacillus buchneri P2.
18

ACCEPTED MANUSCRIPT

474

Journal of Applied Microbiology, 108, 2059-2066.

AC
C

EP

TE
D

M
AN
U

SC

RI
PT

475

19

ACCEPTED MANUSCRIPT

Table 1
Cholesterol assimilation and BSH activities of LAB strains isolated from kimchi.
Strain

AB

rod

BC

coccus

5.95

BY

rod

8.33

C12

rod

10.54

C14

rod

15.50

CC

rod

8.33

CM1

short rod

8.92

CM3

short rod

33.78
11.94

CS

short rod

DC1

rod

3.51

11

DC2

rod

5.23

12

DC3

short rod

6.98

13

DJ1

coccus

14

DM1

coccus

15

DS

rod

+
-

SC

BSH activity

RI
PT

Shape

10

13.20

+
-

5.14

10.72

80.86

+++

53.83

EM

rod

GJ2

coccus

18

GJ6

short rod

13.29

19

GJ7

rod

17.70

20

GS

rod

7.07

21

HA1

short rod

22

HC

M
AN
U

16
17

8.78

coccus

10.54

77.61

+++

23

HD1

rod

24

HJ

short rod

10.72

25

JD

short rod

10.09

++
-

JH

short rod

8.92

JS

coccus

9.41

28

JT

coccus

10.81

29

JW

short rod

10.86

30

KO

short rod

7.39

31

KW

10.45

32

MA

33

MH

34

MO1

35
37

coccus

short rod

9.73

short rod

9.59

short rod

17.25

MP1

coccus

6.80

MS1

short rod

9.14

NG1

coccus

15.32

NJ1

short rod

4.91

39

NJ6

short rod

13.15

40

NM1

short rod

15.86

++

41

NO1

rod

78.11

++
+

AC
C

38

TE
D

26
27

36

478
479
480

Cholesterol
Assimilation (%)
11.71

No.

EP

476
477

42

NP

coccus

13.87

43

OS

rod

16.49

44

PH2

rod

11.98

45

R1

coccus

9.37

46

SC

coccus

3.83

47

SH

rod

9.68

48

SM

rod

11.08

49

UC

short rod

3.78

50

W1

coccus

8.29

*BSH activity was expressed based on the diameters of precipitation zones on BSH screening medium: -,
no precipitation; +, precipitation zone of up to 10 mm; ++, precipitation zone of up to 15 mm; and +++,
precipitation zone of up to 20 mm.
20

ACCEPTED MANUSCRIPT

Table 2
Acid and bile tolerance of L. plantarum EM.

SGJc

Survival after 3 h at pH 8.0


0.3% oxgall

log CFU/ml

log CFU/ml

%a

log CFU/ml

%a

log CFU/ml

%a

9.780.09a

8.090.50b

82.7

8.520.07b

87.1

5.531.20c

56.5

TE
D

M
AN
U

All values are means standard deviation.


a
% Survival: final (CFU/ml)/control (CFU/ml) 100. 100% survival indicates that the growth rate of the strain was not affected by the treatment.
b
PBS : Phosphate-buffered saline.
c
SGJ : Simulated gastric juice.
* Means in the same row with different superscript letters are significantly different (p < 0.05) by Duncans multiple range test.

EP

483
484
485
486
487

RI
PT

L. plantarum EM

PBSb

SC

Strain

Survival after 1 h at pH 2.5

Initial
mean counts

AC
C

481
482

21

ACCEPTED MANUSCRIPT

488
489

Table 3
Minimum inhibitory concentrations of antibiotics for L. plantarum EM.
MIC (g/ ml)b
Strain
GEN

KAN

STR

ERY

CLI

TET

CHL

L. plantarum EM

>512

0.5

16

0.125

16

Break points for L. plantaruma

n.r.c

16

64

n.r.

RI
PT

VAN

32

Breakpoints are according to the guidelines of the EFSA (EFSA 2012).


Strains with MICs lower than or equal to the breakpoints are considered susceptible. AMP: ampicillin;
VAN: vancomycin; GEN: gentamycin; KAN: kanamycin; STR: streptomycin, ERY: erythromycin; CLI:
clindamycin; TET: tetracycline; CHL: chloramphenicol.
c
n.r. Not required.

EP

TE
D

M
AN
U

SC

AC
C

490
491
492
493
494

AMP

22

ACCEPTED MANUSCRIPT

495
496

Table 4
Antimicrobial activity of L. plantarum EM.
Microorganisms

Antimicrobial
activity (AU/mL)

Sensitive strains
Bacillus cereus KCTC 3624
Micrococcus luteus ATCC 15307

800

Staphylococcus aureus ATCC 29123


E. coli O157:H ATCC 43895
Pseudomonas aeruginosa ATCC 27853

Salmonella enterica serovar Typhi ATCC 19430


Vibrio parahaemolyticus ATCC 17802

EP

TE
D

M
AN
U

Antimicrobial activity was measured by the spot-on-the-lawn test.

AC
C

497

23

200

1,500

SC

Gram-negative
bacteria

RI
PT

Gram-positive
bacteria

3,100

3,100

3,200
25,600

ACCEPTED MANUSCRIPT

498
499

Table 5
Growth of lactic acid bacteria in the presence of bile acid.
A600
Media

L. plantarum
EM

L. acidophilus
ATCC 43121

5.750.02aC

9.870.01aB

MRS+0.3% TDCA+0.1 g/L water-soluble cholesterol

9.740.01bA

4.980.07bC

9.180.09bB

MRS+0.5% TDCA+0.1 g/L water-soluble cholesterol

9.040.01cA

4.910.02bcB

9.090.09bA

MRS+0.3% Oxgall+0.1 g/L water-soluble cholesterol

8.570.03dA

4.830.04bcB

8.490.11cA

MRS+0.5% Oxgall+0.1 g/L water-soluble cholesterol

7.960.06eB

4.740.20cC

8.480.11cA

SC

RI
PT

10.470.01aA

MRS

EP

TE
D

M
AN
U

All values are means standard deviation.


Means in the same column for a single strain with different lowercase superscript letters are significantly
different (p < 0.05) by Duncans multiple range test. Means in the same row with different uppercase
superscript letters are significantly different (p < 0.05) by Duncans multiple range test.

AC
C

500
501
502
503

L. sakei
DC1

24

ACCEPTED MANUSCRIPT

Table 6
Cholesterol removal by growing, resting, and dead cells of LAB.

Cholesterol
removal (%)

Cholesterol
removal (%)

Growing cell

88.122.80a

47.660.00b

Resting cell

60.590.83c

Dead cell

39.020.00d

Cell state

L. plantarum EM

L. sakei DC1

33.330.00d

6.442.10g

Resting cell

5.292.50g

Dead cell

1.220.00h

0.000.00g

80.690.70b

51.700.30a

32.350.83e

15.450.40e

29.270.00f

15.150.00e

Resting cell
Dead cell

6.381.20f
5.900.40f

EP

TE
D

Growing, resting, and dead cells were incubated at 37 C for 24 h in 0.5% oxgall- as well as 0.5% TDCAcontaining cholesterol (0.1 g/l) media.
* Means in the same column with different superscript letters are significantly different (p < 0.05) by
Duncans multiple range test.

AC
C

506
507
508
509

38.200.00c

Growing cell

Growing cell
L. acidophilus
ATCC 43121

RI
PT

0.5% TDCA

SC

Strains

0.5% oxgall

M
AN
U

504
505

25

ACCEPTED MANUSCRIPT

510

Figure captions

511
Fig. 1. Cholesterol removal by cell wall fractions of L. plantarum EM (
) and L. acidophilus ATCC

513
514
515

43121 (
).
Fig. 2. Scanning electron micrographs of LAB in media containing no cholesterol (A) and media
containing cholesterol (B).

AC
C

EP

TE
D

M
AN
U

SC

RI
PT

512

26

EP

TE
D

Fig. 1

AC
C

516
517

M
AN
U

SC

RI
PT

ACCEPTED MANUSCRIPT

27

ACCEPTED MANUSCRIPT

LAB

L. plantarum EM

Cell state

L. sakei DC1

L. acidophilus ATCC 43121

RI
PT

Growing
cell

M
AN
U

SC

TE
D

Dead
cell

Cell wall
fraction

AC
C

EP

518

Fig. 2

28

ACCEPTED MANUSCRIPT
Highlights
L. plantarum EM showed high cholesterol removal by growing and even dead cells.
Cell wall fraction of L. plantarum EM removed cholesterol.
L. plantarum EM met the functional criteria required for beneficial probiotics.

AC
C

EP

TE
D

M
AN
U

SC

RI
PT

L. plantarum EM could be a promising probiotic candidate regardless of its viability.

También podría gustarte