This article was downloaded by: [Swinburne University of Technology] On: 06 September 2014, At: 09:09 Publisher: Taylor & Francis Informa Ltd Registered in England and Wales Registered Number: 1072954 Registered office: Mortimer House, 37-41 Mortimer Street, London W1T 3JH, UK

Preparative Biochemistry and Biotechnology Publication details, including instructions for authors and subscription information: http://www.tandfonline.com/loi/lpbb20

Comparison of Two Methods for Purification of Enterocin B, A Bacteriocin Produced by Enterococcus Faecium W3 a

b

b

b

c

Halil Dündar , Mehmet Atakay , Ömür Çelikbiçak , Bekir Salih & Faruk Bozoğlu a

Department of Biotechnology, Middle East Technical University, Ankara, Turkey

b

Department of Chemistry, Hacettepe University, Beytepe, Ankara, Turkey

c

Department of Food Engineering, Middle East Technical University, Ankara, Turkey Accepted author version posted online: 02 Sep 2014.

To cite this article: Halil Dündar, Mehmet Atakay, Ömür Çelikbiçak, Bekir Salih & Faruk Bozoğlu (2014): Comparison of Two Methods for Purification of Enterocin B, A Bacteriocin Produced by Enterococcus Faecium W3, Preparative Biochemistry and Biotechnology, DOI: 10.1080/10826068.2014.958165 To link to this article: http://dx.doi.org/10.1080/10826068.2014.958165

Disclaimer: This is a version of an unedited manuscript that has been accepted for publication. As a service to authors and researchers we are providing this version of the accepted manuscript (AM). Copyediting, typesetting, and review of the resulting proof will be undertaken on this manuscript before final publication of the Version of Record (VoR). During production and pre-press, errors may be discovered which could affect the content, and all legal disclaimers that apply to the journal relate to this version also.

PLEASE SCROLL DOWN FOR ARTICLE Taylor & Francis makes every effort to ensure the accuracy of all the information (the “Content”) contained in the publications on our platform. However, Taylor & Francis, our agents, and our licensors make no representations or warranties whatsoever as to the accuracy, completeness, or suitability for any purpose of the Content. Any opinions and views expressed in this publication are the opinions and views of the authors, and are not the views of or endorsed by Taylor & Francis. The accuracy of the Content should not be relied upon and should be independently verified with primary sources of information. Taylor and Francis shall not be liable for any losses, actions, claims, proceedings, demands, costs, expenses, damages, and other liabilities whatsoever or howsoever caused arising directly or indirectly in connection with, in relation to or arising out of the use of the Content. This article may be used for research, teaching, and private study purposes. Any substantial or systematic reproduction, redistribution, reselling, loan, sub-licensing, systematic supply, or distribution in any form to anyone is expressly forbidden. Terms & Conditions of access and use can be found at http:// www.tandfonline.com/page/terms-and-conditions

COMPARISON OF TWO METHODS FOR PURIFICATION OF ENTEROCIN B, A BACTERIOCIN PRODUCED By Enterococcus faecium W3 Halil Dündar1, Mehmet Atakay2, Ömür Çelikbiçak2, Bekir Salih2, Faruk Bozoğlu3 1

Department of Biotechnology, Middle East Technical University, Ankara, Turkey, Department of Chemistry, Hacettepe University, Beytepe, Ankara, Turkey, 3Department of Food Engineering, Middle East Technical University, Ankara, Turkey

cr ip t

2

Abstract

an us

This study aimed to compare two different approaches for the purification of enterocin B from Enterococcus faecium strain W3 based on the observation that the bacteriocin was found both in cell associated form and in culture supernatant. The first approach employed ammonium sulfate precipitation, cation-exchange chromatography and

M

sequential reverse-phase high performance liquid chromatography. The latter approach exploited pH mediated cell adsorption-desorption method to extract cell bound

ep te d

bacteriocin, and one run of reverse-phase chromatography. The first method resulted in purification of enterocin B with a recovery of 4% of the initial bacteriocin activity found in culture supernatant. MALDI-TOF MS analysis and de novo peptide sequencing of the purified bacteriocin confirmed that the active peptide was enterocin B. The second method achieved the purification of enterocin B with a higher recovery (16%) and

Ac c

Downloaded by [Swinburne University of Technology] at 09:09 06 September 2014

Correspondence Addressed to Halil Dündar: E-mail: [email protected]

enabled us to achieve pure bacteriocin within a shorter period of time by avoiding time consuming purification protocols. The purity and identity of the active peptide was confirmed again by MALDI-TOF MS analysis. Although both approaches were satisfactory to obtain sufficient amount of enterocin B for use in MS and amino acid

1

sequence analysis, the latter was proved to be applicable in large-scale and rapid purification of enterocin B.

INTRODUCTION

an us

Bacteriocins from lactic acid bacteria (LAB) are of great importance because of their use in food preservation.[1] Bacteriocins are ribosomally synthesized antimicrobial peptides displaying antagonistic effect against other bacteria.[2] They exhibit bactericidal activity on the sensitive cells through depolarization of cell membrane or through the inhibition

M

of cell wall synthesis. The bacteriocins from LAB have received considerable attention regarding food safety because of the generally recognized as safe (GRAS) status of these

ep te d

bacteria.[3] Bacteriocins produced from Enterococcus spp. are the most widely produced and characterized among LAB bacteriocins.[4] Among them, enterocin B shows important properties such as stability and solubility over a wide pH range, heat stability and inhibition spectrum, making it an attractive candidate for food preservation. Enterocin B was shown to inhibit the growth of Listeria monocytogenes, Enterococcus faecalis,

Ac c

Downloaded by [Swinburne University of Technology] at 09:09 06 September 2014

chromatography, MALDI-TOF, peptide sequencing

cr ip t

KEYWORDS: bacteriocin, enterocin B, bacteriocin purification, reverse-phase

Clostridium sporogenes, Clostridium tyrobutyricum, Staphylococcus aureus.[5] It showed significant antilisterial effect in minced pork meat, pork liver patè cooked ham and debondened chicken breasts[6] as well as inhibiting the production of slime by L. sakei CTC746 in vacuum packaged sliced cooked pork ham.[7] The synergistic action of enterocin B with high hydrostatic pressure (HHP) was shown in cooked ham model with

2

reduction in the intensity of HHP treatment.[8] The first attempts to purify enterocin B employed its cationic and hydrophobic nature and hence, cation-exchange and hydrophobic interaction chromatography with repeated reverse-phase chromatography steps were applied on the concentrated culture supernatant.[5] Various methods were

cr ip t

reported for the purification of enterocins: enterocin A purification; ammonium sulfate

exchange column), hydrophobic interaction chromatography (Octyl-Sepharose CL-4B

an us

column), followed by C 2/C18 reverse-phase chromatography,[9] enterocin B purification;

extraction with Amberlite XAD-16, cation-exchange chromatography (SP-Sepharose Fast Flow cation-exchange column), hydrophobic interaction chromatography (OctylSepharose CL-4B column), followed by C2/C18 reverse-phase chromatography,[5]

M

enterocins L50A and L50B purification; extraction with Amberlite XAD-16, cationexchange chromatography (SP-Sepharose Fast Flow cation-exchange column),

ep te d

hydrophobic interaction chromatography (Octyl-Sepharose CL-4B column), followed by C2/C18 reverse-phase chromatography,[10] enterocin I purification; ammonium sulfate precipitation, cation-exchange chromatography (SP-Sepharose Fast-Flow column), hydrophobic interaction chromatography (Phenyl-Sepharose CL-4B column), followed by C2/C18 reverse-phase chromatography,[11] enterocin P purification; ammonium sulfate

Ac c

Downloaded by [Swinburne University of Technology] at 09:09 06 September 2014

precipitation, cation-exchange chromatography (SP-Sepharose Fast Flow cation-

precipitation, gel filtration (G25 PD10 columns), cation exchange chromatography (SPSepharose Fast Flow column), hydrophobic-interaction chromatography (OctylSepharose CL-4B column), followed by C2/C18 reverse-phase chromatography.[12] Although these methods worked to achieve purification to homogeneity for elucidation of primary structure, the yields were variable and low, the protocols being time consuming

3

and not suitable for large-scale applications. Therefore, methods for rapid and large-scale purification of bacteriocins need to be devised for use in controlling food spoilage and in clinical applications.

cr ip t

Micro-Cel,[13] Rice hull ash and silicic acid[14] were exploited for extraction of

was studied also.[15,16] Bacteriocins of lactic acid bacteria are adsorbed on the cells of

an us

producer strain depending of pH of the environment, adsorption occurs at around pH 6.0 and desorption occurs around pH 2.0. Based on this property of LAB bacteriocins,

pediocin AcH, leuconocin Lcm1 and sakacin A, and the lantibiotic nisin were purified on

M

a large-scale.[17]

In this study we tested the utility of pH mediated cell adsorption-desorption method with

ep te d

the subsequent reverse-phase high performance liquid chromatography to purify enterocin B. The method was compared with the usual purification protocol consisting of ammonium sulfate precipitation, cation-exchange chromatography and sequential reverse-phase high performance liquid chromatography.

Ac c

Downloaded by [Swinburne University of Technology] at 09:09 06 September 2014

bacteriocins from culture supernatant. Extraction with aqueous two-phase partitioning

MATERIALS AND METHODS

Microorganisms

Bacteriocin producer Enterococcus faecium W3 was a food strain previously isolated in our laboratory Middle East Technical University, Food Microbiology Labotatory) and

4

identified as E. faecium by gram staining, endospore staining, catalase reaction, heterofermentative/homofermentative distinguishing and 16S rDNA analysis.

Bacteriocin Activity Assay

cr ip t

Bacteriocin activity was evaluated by a microtiter plate assay. Two-fold serial dilutions

μL de Man Rogasa Sharp (MRS) broth. 150 μL of indicator strain Lactobacillus

an us

delbrueckii subsp. delbrueckii RSSK 498 (100-fold diluted) was added to the bacteriocin containing wells and the plate was incubated at 30°C for 6 h. Bacteriocin activity was evaluated by measuring the growth at 620 nm by a microtiter plate reader reader (Multiscan Ascent; Labsystems, Helsinki, Finland). One bacteriocin unit (BU) was

M

defined as the amount of bacteriocin inhibiting the growth of the indicator strain by 50%.

ep te d

Temperature And Ph Optimization For Bacteriocin Production MRS broth adjusted to an initial pH of 6.5 was inoculated (1%, v/v) with an overnight culture of strain W3 and incubated at 10, 15, 20, 25, 30 and 37°C without pH control for 16 h. For determination of the effect of initial pH on bacteriocin production, MRS broth adjusted to pH 4.0, 4.5, 5.0, 5.5, 6.0, 6.5, 7.0 was inoculated (1%, v/v) with an overnight

Ac c

Downloaded by [Swinburne University of Technology] at 09:09 06 September 2014

of preparations containing bacteriocin were added to microtiter plate wells containing 50

culture of strain W3 and incubated at 30°C without pH regulation for 16 h. Bacteriocin activity was measured as described above.

Bacteriocin Purification

5

Purification By Ammonium Sulfate Precipitation, Cation Exchange Chromatography On SP-Sepharose Fast Flow Column And Reverse-Phase High Performance Liquid Chromatography On Akta Purifier. 1000 mL of MRS broth was inoculated with an overnight culture of E. faecium W3 (1%

cr ip t

v/v) and incubated for 16 h with an initial pH of 6.5 at 30°C. Bacteriocin containing

4°C. Ammonium sulfate (40 g per 100 mL) was added to 500 mL of culture supernatant

an us

in centrifuge tubes and then shaked at 4°C for 40 min. The mixture was centrifugated

(13000 rpm for 30 min at 4°C) to precipitate the bacteriocin from supernatant. The pellet was dissolved in 25 mL of sterile distilled water and diluted to 100 mL with sterile distilled water. This sample was applied to 5 mL of SP-Sepharose Fast Flow (GE

M

Healthcare Biosciences) column, which was equilibrated with 10 mM acetic acid. The column was washed with 2 column volumes (CV) of 10 mM sodium phosphate buffer

ep te d

(pH 6.0) and 2 CV of sodium phosphate buffer (10 mM, pH 6.0) containing 100 mM NaCI. Subsequently, the column was eluted with a step-wise gradient of 2 CV of 300 mM NaCI and 2 CV of 1.0 M NaCI. Fractions of 2.5 mL at a flow rate of 1 mL were collected and assayed for bacteriocin activity. Fractions with the highest bacteriocin activity from SP Sepharose Fast flow column, the last 7.5 mL (300 mM NaCI) and the first 5 mL (1.0

Ac c

Downloaded by [Swinburne University of Technology] at 09:09 06 September 2014

supernatant was obtained after centrifugation of the culture at 13000 rpm for 30 min at

M NaCI), were combined and applied to Resource 15 RPC 3 mL reverse-phase column (100 mm length, 6.4 mm i.d., and with 15 µm pore size) equilibrated with 0.1% (v/v) (TFA) in distilled water. Elution was carried out with a 5 CV linear gradient of 15 to 55% 2-propanol with 0.1% TFA at a flow rate of 1 mL/min using Äkta Purifier fast protein liquid chromatography system. Fractions of 1 mL were collected and assayed for

6

bacteriocin activity. Active fractions from first reverse-phase chromatography were applied to reverse-phase chromatography under the same conditions and bacteriocin activity was assayed as described above. Purified bacteriocin was subjected to MALDI-

cr ip t

TOF mass spectrometry and amino acid sequencing analysis.

Phase High Performance Liquid Chromatography

an us

The second purification protocol employed cell-associated form of the bacteriocin as an initial sample to extract relatively pure bacteriocin based on our observation that the bacteriocin produced by E. faecium W3 was detected in cell associated form (bound to

producer cell surface) as well as in culture supernatant fluid. The pelleted cells of the 1 L

M

of culture was mixed with 500 mL of culture supernatant left from the same fermentation flask. The pH of the heat treated culture was adjusted to 6.0 by addition of 1.0 M NaOH

ep te d

solution. The culture was stirred for 1 h at 4°C to allow the bacteriocin to adsorb onto bacterial cells. The cells were collected by centrifugation and washed with 200 mL of 5.0 mM sodium phosphate buffer (pH 6.0) to remove impurities. After centrifugation for 30 min at 4°C (13000 rpm), the pelleted cells were resuspended in 100 mL of 5.0 mM sodium phosphate buffer (pH 6.0) containing 100 mM NaCI, and the pH was adjusted to

Ac c

Downloaded by [Swinburne University of Technology] at 09:09 06 September 2014

Purification By Ph Mediated Cell Adsorption-Desorption Method And Reverse-

2.0 by gradual addition of HCI. The cell suspension was stirred in cold room (4°C) overnight, centrifuged and filtered. The extract was concentrated with boiling to a volume of 12 mL and assayed for bacteriocin activity. After freeze-drying of this fraction, it was resuspended in 1 mL of 0.1% TFA and applied to reverse-phase high performance liquid chromatography by injecting 100 µL aliquots into Vydac C18 column (150 mm length,

7

4.6 mm i.d., and with 5 µm pore size) incorporated in Agilent 1200 Series Quaternary HPLC system. Detection was carried out at wavelength range between 190-700 nm. Mobile phase was composed using varying amounts of water with 0.1% TFA (v/v) (A) and acetonitrile (ACN) with 0.085% TFA (v/v) (B). Mobile phase was varied in the

cr ip t

following gradients: 0 to 5 min, 96% to 4% (v/v); 5 to 80 min, 60% to 40% (v/v); 80 to

collected in separate vials for 1 min intervals and assayed for bacteriocin activity. Protein

an us

concentrations were measured spectrophotometrically at 280 nm. Purified bacteriocin

was subjected to MALDI-TOF mass spectrometry and amino acid sequencing analysis.

MALDI-TOF Analysis

M

Active fractions were dried in water bath at 70°C, and finally in vacuum oven at 30°C. Mass spectrometric analyses were performed by dissolving the dried peptide in 200 μL

ep te d

ACN:H2O mixture in 50:50 (v/v) containing 0.1% TFA (v/v). Mass spectra of purified bacteriocin were acquired on a Voyager-DETM PRO MALDI-TOF mass spectrometer (Applied Biosystems, USA) as described.[18]

Amino Acid Sequencing

Ac c

Downloaded by [Swinburne University of Technology] at 09:09 06 September 2014

90 min, 10% to 90% (v/v); and 90 to 100 min, 10% to 90% (v/v). Peptide fractions were

Mass spectrometry data were acquired using a Q-STAR Elite Q-TOF mass spectrometer (Applied Biosystems) with a nano-ESI source. MS/MS data were obtained via Information Dependent Acquisition (IDA) method, where doubly and triply charged parent ions were selected for fragmentation by collision induced dissociation (CID) using nitrogen as collision gas. Protein identification was performed by searching the NCBI

8

database using the Mascot search engine with the following search criteria: monoisotopic mass, peptide charge +2, 50 ppm mass accuracy, trypsin as digesting enzyme with 1 missed cleavage allowed, carbamidomethylation of cysteine as a fixed modification, oxidation of methionine as allowable variable modification. For all de novo algorithms,

cr ip t

methionine oxidation was selected as variable modification and carbamidomethylation of

tolerance and MS/MS tolerance of 0.3 Da. A maximum of one missed-cleavage was

an us

allowed. All data were searched for tryptic peptides.

Mode Of Action Of Ph Mediated Cell Surface Adsorption-Desorption Fraction Cell suspension of Listeria monocytogenes RSSK 475 and Enterococcus faecalis was

M

grown to exponential growth phase. Monitoring of bacterial growth was carried out by determining the absorbance at 600 nm (A600). Exponentially growing culture of the

ep te d

organism was divided into two equal parts and pH mediated cell surface adsorptiondesorption fraction from E. faecium W3 was added to each part with a final concentration of 300 AU mL. Control culture and bacteriocin treated culture were incubated and at selected intervals samples were taken from both cultures for total viable colony counting and A600 measurement. Total viable counts (CFU/mL) were determined by pour plating

Ac c

Downloaded by [Swinburne University of Technology] at 09:09 06 September 2014

cysteines as fixed modification. QSTAR data were de novo sequenced using peptide

technique using the serial dilutions of indicator organism. A600 values were measured by a spectrophotometer (path length, 1 cm; UV-160 UV-visible spectrophotometer; Shimadsu). All assays were performed in duplicate.

RESULTS AND DISCUSSION

9

Effect Of Temperature And Ph On Bacteriocin Production Growth and bacteriocin production occurred at temperatures between 20 and 37 oC with an initial pH of 6.5-7.0 under uncontrolled conditions. Production of bacteriocin by the producer organism resulted in 320 AU/mL bacteriocin activity at 30 and 37 oC in CSF and

cr ip t

160 AU/mL at 20 and 25 oC). The producer organism produced the same amount of

an us

Nevertheless, no bacterial growth and bacteriocin production were detected under pH 5.0.

Purification By Ammonium Sulfate Precipitation, Cation-Exchange

Chromatography On SP-Sepharose Fast Flow, And Three Consecutive ReversePhase HPLC On Resource RPC 3 Ml Column Using Äkta Purifier.

M

The culture supernatant had an activity of 320 BU/mL and ammonium sulfate precipitation resulted in 80% recovery of the initial bacteriocin activity. The precipitate

ep te d

was subjected to cation-exchange chromatography on SP Sepharose Fast Flow column. The recovery by cation-exchange chromatography was low (14%) due to activity loss in flow-through. The results of the protocol is shown in Table 1. The highest bacteriocin activity from SP Sepharose Fast flow column, the last 7.5 mL (300 mM NaCl) and the first 5 mL (1.0 M NaCl), were pooled (19200 BU/mL) and applied to Resource 15 RPC 3

Ac c

Downloaded by [Swinburne University of Technology] at 09:09 06 September 2014

bacteriocin whent the initial pH was between 5.5 and 7.0 (320 AU/mL) at 30 oC.

mL reverse-phase column equilibrated with 0.1% (v/v) (TFA) in distilled water. Bacteriocin activity was detected in fractions 11, 12, and 13 (Figure 1). Fraction 12 with the highest bacteriocin activity (6400 BU/mL) was diluted ten times with 0.1% (v/v) TFA and run on Resource 15 RPC 3 mL column with Akta purifier under the same conditions. The second run of reverse-phase HPLC resulted in one symmetrical peak with bacteriocin

10

activity in fraction 18 (6400 BU/mL) (Figure 2), which was concentrated by SpeedVac, applied for MALDI-TOF and de novo peptide sequencing. MALDI-TOF analysis of this active peak resulted in a mass of 5462 Da (Figure 3) and its de novo sequencing produced the following amino acid sequence from position 8 to 21, position 23 to 35 and position

cr ip t

37 to 50: NH2--------NEI/LNRPNNI/LS-----CGAAI/LAGGI/LFGI/LP-

Thus, the sequence and MALDI-TOF mass spectrometry data confirmed that active peak

an us

is identical to enterocin B with the following sequence:

ENDHRMPNELNRPNNLSKGGAKCGAAIAGGLFGIPKGPLAWAAGLANVYSKCN.

Although the protocol worked well enough to be able to identify the bacteriocin, it caused

M

substantial loss of bacteriocin activity with a final recovery of 4%. The remarkable activity loss was observed during cation-exchange chromatography where much

ep te d

bacteriocin activity was lost in flow through (Table 1). Reverse-phase chromatography also contributed to the loss of bacteriocin activity during the first run.

Cell-associated form of the bacteriocin was used as initial extract in the second purification method where the bacteriocin was eluted from the cell surface by low pH

Ac c

Downloaded by [Swinburne University of Technology] at 09:09 06 September 2014

GPI/LAWAAGI/LANVYS where I/L denotes that the residue is isoleucine or leucine.

(pH 2.0) and 150 mM NaCI. This extraction method recovered 48% of the bacteriocin activity found in culture supernatant. The pH 2 extract was purified to homogeneity with the application of reverse-phase HPLC on Vydac C18 column incorporated in Agilent 1200 Series Quaternary HPLC system. The pH dependent adsorption-desorption method resulted in purification with 21.5 fold increase but the largest increase in specific activity

11

was provided by reverse-phase chromatography with a 4408 fold increase (Table 2). RPHPLC resulted in six peaks, the fourth peak showed bacteriocin activity (Figure 4). The retention time of the bacteriocin on Vydac C18 column was about 80 min, corresponding to 90% acetonitrile. MALDI-TOF MS analysis confirmed the purity and molecular mass

cr ip t

of enterocin B (Figure 5). MALDI mass spectrum of active peptide was obtained using

mass spectrum of the sample was obtained in α-cyano-4-hydroxycinnamic acid matrix

an us

and is given in Figure 5. Three mass value for the active peptide were observed by MALDI-TOF analysis including 5444, 5466 and 5482 Da as a result of the second

purification protocol (Figure 5). The main peak with a molecular mass of 5465 Da is the same as the calculated molecular mass (5465.2 Da). The peak with a molecular mass of

M

5482 Da probably represents Enterocin B with an oxidized methionine residue as reported previously.[5,19] The related molecular mass values of enterocin B including 5466

ep te d

and 5482 Da were observed both in pH mediated cell surface adsorption-desorption extract and in the active fraction from reverse-phase chromatography. MALDI-MS spectra of this active fraction were also carried out at low and at very high mass ranges up to 200000 Da. No other peptide and proteins were observed in the same active peptide fraction (Figure 5). Amino acid sequence analysis of the active fraction showed that it

Ac c

Downloaded by [Swinburne University of Technology] at 09:09 06 September 2014

positive ion and linear mode in various novel MALDI matrices but the suitable MALDI

was identical to the sequence obtained from the first purification protocol. The pH mediated cell adsorption-desorption method was carried out with some modifications as described previously.[17] It is known that bacteriocin producing strains of lactic acid bacteria adsorb their own bacteriocins around pH 6.0 and release around pH 2.0.[20] Dextranicin 24 produced by Leuconostoc sp.,[21] pediocin AcM produced by

12

Pediococcus acidilactici,[22] leucocins A-, B- and C-TA33a produced by Leuconostoc mesenteroides TA33a[23] and plantaricin C19 produced by Lactobacillus plantarum C19[24] were purified by pH mediated cell adsorption-desorption method and reversephase chromatography. Bovicin 255 produced by Streptococcus sp. was purified by pH

cr ip t

mediated cell adsorption-desorption method, ammonium sulphate precipitation, reverse-

by Pediococcus pentosaceus was purified by pH mediated cell adsorption-desorption

an us

method and gel filtration chromatography on FPLC system.[26] In this study, pH mediated cell adsorption-desorption method provided relatively pure enterocin B as seen after running of the sample by reverse-phase chromatography on Vydac column. Enterocin B obtained by pH mediated cell adsorption-desorption method can be used in ex situ

M

applications taken into account its purity and higher yield without performing expensive purification protocols. Most purification schemes devised for purification of enterocins

ep te d

and other bacteriocins from lactic acid bacteria take advantage of the cationic and relatively hydrophobic natures of these molecules, and hence involve applications of cation-exchange chromatography, hydrophobic interaction chromatography and reversephase high performance liquid chromatography. Although these methods has been successful for the purification of many bacteriocins for identification, they are time

Ac c

Downloaded by [Swinburne University of Technology] at 09:09 06 September 2014

phase and gel filtration chromatography on FPLC system.[25] Pediocin ACCEL produced

consuming, costly and not suitable for large-scale production.

Inhibition Spectrum Of Cell Surface Adsorbed-Desorbed Enterocin B Both partially purified bacteriocin (pH mediated cell surface adsorption desorption fraction) and its processed product through reverse-phase chromatography was tested

13

against a set of Gram-positive and Gram-negative bacteria. Pure enterocin B and cell surface adsorption-desorption fraction showed the same inhibitory spectrum and inhibited the growth of Lc. mesenteroides, L. delbrueckii, L. cremoris, L. monocytogenes, L. innocua, E. faecalis, B. cereus and some lactic acid bacteria. However, no Gram-negative

cr ip t

bacteria tested were inhibited by pure enterocin B and the cell surface adsorption-

an us

Mode Of Action

Turbidity of the bacteriocin treated culture of L. monocytogenes declined rapidly after the first 20 min (Figure 6). The decrease in A600 continued throughout the incubation as opposed the control culture. L. monocytogenes RSSK 475 lost its viability by 99.99 %

M

within the first 20 min; viable colony count dropped rapidly from 8x10 8 to 104 (a 5.9 log reduction). After 4 hours, the viable colony count of the bacteriocin treated culture was

ep te d

400 CFU/mL, that is a 6.3 log reduction in viability (Figure 6).

CONCLUSION

Comparison of the two purification approach indicated that pH mediated cell surface adsorption-desorption method together with RP-HPLC can be used successfully for rapid

Ac c

Downloaded by [Swinburne University of Technology] at 09:09 06 September 2014

desorption fraction (Table 3).

purification of enterocin B, which is a non-pediocin like one peptide bacteriocin (class IIc), as performed for many pediocin-like bacteriocins. The application of pH mediated cell surface adsorption-desorption method avoided time consuming ammonium sulfate precipitation, cation-exchange chromatography and hydrophobic interaction chromatography steps which might cause activity losses during the purification. Although

14

both methods achieved purification of enterocin B with a similar specific activity, a significant improvement in the yield of enterocin B was achieved (16%) by pH mediated cell surface adsorption with a following reverse-phase chromatography compared with the yield obtained by ammonium sulfate precipitation, cation-exchange chromatography

cr ip t

and final sequential reverse-phase chromatography (4%). Both purification approaches is

high purity and yield of the bacteriocin preparation obtained from pH mediated cell

an us

surface adsorption-desorption method make it possible its use in a concentrated form as a

food preservative. The cell of the producer strain functioned as a specific ligand to adsorb the enterocin B from culture supernatant, and provided both purification and concentation of the molecule. It is known that some stains of E. faecium produces enterocin A together

M

with enterocin B which shows ynergistic action towards pathogenic and food-spoilage bacteria when used in combination. The rapid purification method mentioned here can be

ep te d

exploited for the large-scale purification of enterocin A and enterocin B together as well as for the bacteriocins of multiple bacteriocin producer strains. Combined and partial purification of different bacteriocins might be an alternative approach to avoid development of bacteriocin resistant strains. The feasibility of pH mediated cell surface adsorption and desorption method can be analysed for the multiple bacteriocins from

Ac c

Downloaded by [Swinburne University of Technology] at 09:09 06 September 2014

successful to the extent that primary structure of the bacteriocin was obtained. However,

different LAB bacteria by mixing relavant bacteriocin supernatants and optimization of adsorption onto a suitable producer cell, which can provide large-scale and concomitant purification of a bacteriocin to achieve a strong bacteriocin cocktail targeting the resistant cells.

15

REFERENCES 1.

Gálvez, A.; Abriouel, H.; López, R.L.; Omar, N.B. 2007. Bacteriocin-based

strategies for food biopreservation. Int. J. Food Microbiol. 2007, 120, 51–70. Garneau, S.; Martin, N.I.; Vederas, J.C. Two-peptide bacteriocins produced by

lactic acid bacteria. Biochimie. 2002, 84, 577–592.

Mills, S.; Stanton, C.; Hill, C.; Ross, R.P. New developments and applications of

bacteriocins and peptides in foods. Annu. Rev. Food Sci. Technol. 2011, 2, 299–329.

Batdorj, B.; Dalgalarrondo, M.; Choiset, Y.; Pedroche, J.; Métro, F.; Prévost, H.;

an us

4.

Chobert, J.M.; Haertlé, T. Purification and characterization of two bacteriocins produced by lactic acid bacteria isolated from Mongolian airag. J. Appl. Microbiol. 2006, 101, 837– 848.

Casaus, P.; Nilsen, T.; Cintas, L.M.; Nes, I.F.; Hernández, P.E.; Holo, H.

M

5.

Enterocin B, a new bacteriocin from Enterococcus faecium T136 which can act

6.

ep te d

synergistically with enterocin A. Microbiol. 1997, 143, 2287–2294. Aymerich, M.T.; Garriga, M.; Ylla, J.; Vallier, J.; Monfort, J.M.; Hugas, M.

Application of enterocins as biopreservatives against Listeria innocua in meat products. J. Food Prot. 2000, 63, 721–726. 7.

Aymerich, M.T.; Garriga, M.; Costa, S.; Monfort, J.M.; Hugas, M. Prevention of

Ac c

Downloaded by [Swinburne University of Technology] at 09:09 06 September 2014

3.

cr ip t

2.

ropiness in cooked pork by bacteriocinogenic cultures. Int. Dairy J. 2002, 12, 239–246. 8.

Garriga, M.; Aymerich, M.T.; Costa, S.; Monfort, J.M.; Hugas, M. Bactericidal

synergism through bacteriocins and high pressure in a meat model system during storage. Food Microbiol. 2002, 19, 509–518.

16

9.

Aymerich, T.; Holo, H.; Håvarstein, L.S.; Hugas, M.; Garriga, M.; Nes, I.F.

Biochemical and genetic characterization of enterocin A from Enterococcus faecium, a new antilisterial bacteriocin in the pediocin family of bacteriocins. Appl. Environ. Microbiol. 1996, 62, 1676–1682. Cintas, L.M.; Casaus, P.; Holo, H.; Hernandez, P.E.; Nes, I.F.; Havarstein, L.S.

cr ip t

10.

related to staphylococcal hemolysins. J. Bacteriol. 1998, 180, 1988–1994.

Floriano, B.; Ruiz-Barba, J.L.; Jiménez-Díaz, R. Purification and genetic

an us

11.

characterization of enterocin I from Enterococcus faecium 6T1a, a novel antilisterial plasmid-encoded bacteriocin which does not belong to the pediocin family of bacteriocins. Appl. Environ. Microbiol. 1998, 64, 4883–4890.

Herranz, C.; Mukhopadhyay, S.; Casaus, P.; Martínez, J.M.; Rodríguez, J.M.;

M

12.

Nes, I.F.; Cintas, L.M.; Hernández, P.E. Biochemical and genetic evidence of enterocin P

ep te d

production by two Enterococcus faecium-like strains isolated from fermented sausages. Curr. Microbiol. 1999, 39, 282–290. 13.

Coventry, M.J.; Gordon, J.B.; Alexander, M.; Hickey, M.W.; Wan, J. A food-

grade process for isolation and partial purification of bacteriocins of lactic acid bacteria that uses diatomite calcium silicate. Appl. Environ. Microbiol. 1996, 62, 1764–1769.

Ac c

Downloaded by [Swinburne University of Technology] at 09:09 06 September 2014

Enterocins L50A and L50B, two novel bacteriocins from Enterococcus faecium L50, are

14.

Janes, M.E.; Nannapaneni, R.; Proctor, A.; Johnson, M.G. Rice hull ash and

silicic acid as adsorbents for concentration of bacteriocins. Appl. Environ. Microbiol. 1998, 64, 4403–4409.

17

15.

Métivier, A.; Boyaval, P.; Duffes, F.; Dousset, X.; Compoint, J.P.; Marion, D.

Triton X-114 phase partitioning for the isolation of a pediocin-like bacteriocin from Carnobacterium divergens. Lett. Appl. Microbiol. 2000, 30, 42–46. 16.

Li, C.; Bai, J.; Li, W.; Cai, Z.; Ouyang, F. Optimization of conditions for

17.

Yang, R.; Johnson, M.C.; Ray, B. Novel method to extract large amounts of

18.

an us

bacteriocins from lactic acid bacteria. Appl. Environ. Microbiol. 1992, 58, 3355–3359. Ceyhan, T.; Yüksek, M.; Yağlıoğlu, H.G.; Salih, B.; Erbil, M.K.; Elmalı, A.;

Bekaroğlu, Ö. Synthesis, characterization and nonlinear absorption of novel octakisPOSS substituted metallophthalocyanines and strong optical limiting property of CuPc.

19.

M

Dalton Trans. 2008, 2407–2413.

Franz, C.M.A.P.; Worobo, R.W.; Quadri, L.E.N.; Schillinger, U.; Holzapfel,

ep te d

W.H.; Vederas, J.C.; Stiles, M.E. Atypical genetic locus associated with constitutive production of enterocin B by Enterococcus faecium BFE 900. Appl. Environ. Microbiol. 1999, 65, 2170–2178. 20.

Klaenhammer, T.R. Genetics of bacteriocins produced by lactic acid bacteria.

FEMS Microbiol. Rev. 1993, 12, 39–85.

Ac c

Downloaded by [Swinburne University of Technology] at 09:09 06 September 2014

experimental designs). Biotechnol. Prog. 2001, 17, 366–368.

cr ip t

bacteriocin extraction in PEG (Salt aqueous two-phase systems using statistical

21.

Revol-Junelles, A.M.; Lefebvre, G. Purification and N-terminal amino acid

sequence of dextranicin 24, a bacteriocin of Leuconostoc sp. Curr. Microbiol. 1996, 33, 136–137.

18

22.

Elegado, F.B.; Kim, W.J.; Kwon, D.Y. Rapid purification, partial characterization,

and antimicrobial spectrum of the bacteriocin, Pediocin AcM, from Pediococcus acidilactici M. Int. J. Food Microbiol. 1997, 37, 1–11. 23.

Papathanasopoulos, M.A.; Dykes, G.A.; Revol-Junelles, A.M.; Delfour, A.; Von

cr ip t

Holy, A.; Hastings, J.W. Sequence and structural relationships of leucocins A-, B- and C-

24.

Atrih, A.; Rekhif, N.; Moir, A.J.G.; Lebrihi, A.; Lefebvre, G. Mode of action,

an us

purification and amino acid sequence of plantaricin C19, an anti-Listeria bacteriocin

produced by Lactobacillus plantarum C19. Int. J. Food Microbiol. 2001, 68, 93-104. 25.

Whitford, M.F.; McPherson, M.A.; Forster, R.J.; Teather, R.M. Identification of

bacteriocin-like inhibitors from rumen Streptococcus spp. and isolation and

26.

M

characterization of bovicin 255. Appl. Environ. Microbiol. 2001, 67, 569–574. Wu, C.W.; Yın, L.J.; Jiang, S.T. Purification and characterization of bacteriocin

ep te d

from Pediococcus pentosaceus ACCEL. J. Agric. Food Chem. 2004, 52, 1146–1151.

Ac c

Downloaded by [Swinburne University of Technology] at 09:09 06 September 2014

TA33a from Leuconostoc mesenteroides TA33a. Microbiol. 1998, 144, 1343–1348.

19

TABLE 1. First purification protocol of enterocin B

Stage Culture

Total act

Protein

Sp acta

Yield

(BU)

(mg/mL)

(BU/mg)

(%)

500

160000

30.83

10.37

25

128000

6.22

823.15

12.5

22400

0.42

4266

First run

3

12800

Second run

1

6400

supernatant

SPSepharose

a

14

411

4740

8

457

0.12

53333

4

5143

ep te d

Specific activity is total BU divided by the total protein.

b

79

0.9

M

RP-FPLC b

1

80

an us

(NH4)2SO4

RP-FPLC, fast-performance liquid chromatography.

Ac c

Downloaded by [Swinburne University of Technology] at 09:09 06 September 2014

Precipit.

100

Fold

cr ip t

Purification Vol (mL)

20

TABLE 2 Second purification protocol of enterocin B Purification

Vol

Total

Stage

(mL)

act

Protein

Sp acta

(mg/mL) (BU/mg)

Yield

fold

(%)

500

160000

30.83

10.37

100

1

pH 2 extract

12

76800

28.66

223.3

48

21.5

RP-HPLC b

1

25600

0.56

45714

16

4408

a

an us

Specific activity is total BU divided by the total protein.

cr ip t

Culture sup.

b

ep te d

M

RP-FPLC, fast-performance liquid chromatography.

Ac c

Downloaded by [Swinburne University of Technology] at 09:09 06 September 2014

(BU)

21

TABLE 3. Inhibition spectrum of enterocin B and enterocin pool from E. faecium W3. pH 2 extract

Purified entB

Medium b

a

a

temp(oC)

L. fermentum

-

-

L. casei

-

-

L. acidophilus

-

-

L. plantarum LP73

-

-

L. plantarum Z11L

-

-

L. plantarum HU

-

L. plantarum RSSK 675

+

L. plantarum RSSK 10

+

L. plantarum 80B

+

L. delbrueckii RSSK 498

+

sensitivity

MRS

30

MRS

37

MRS

30

MRS

30

-

MRS

30

+

MRS

30

+

MRS

30

+

MRS

30

+

MRS

30

an us

MRS

M

ep te d

30

L. cremoris RSSK 708

+

+

MRS

30

L. salivarious TNO M7

-

-

MRS

30

L. plantarum LMG 2003

+

+

MRS

30

L. sakei LMG 2313

+

+

MRS

30

+

+

MRS

30

E. faecalis LMG 2602

+

+

GM17

30

L. innocua LMG 2813

+

+

GM17

30

L. monocytogenes RSSK

+

+

GM17

30

Ac c

Downloaded by [Swinburne University of Technology] at 09:09 06 September 2014

sensitivity

Growth

cr ip t

Indicator organism

L. mesenteroides RSSK 923

22

471 L. monocytogenes RSSK

+

+

GM17

30

+

+

GM17

30

+

+

B. cereus LMG 2732

+

+

S. aureus

+

P. pentosaceous LMG

-

L. monocytogenes RSSK 475

30

+

GM17

30

-

TSB

30

-

TSB

30

-

-

TSB

30

-

-

TSB

30

-

-

TSB

30

-

-

TSB

30

-

-

TSB

30

-

ep te d

E. coli LMG 3083

M

30

-

S. paratyphi GATA

GM17 GM17

P. fluorescens LMG 3020

3085

30

+

2001

S. enterica typh LMG

GM17

an us

478

A2961

S. paratyphi GATA

Ac c

Downloaded by [Swinburne University of Technology] at 09:09 06 September 2014

L. monocytogenes RSSK

cr ip t

472

B33653

S. paratyphi GATA B33656 S. paratyphi GATA B34323

23

S. paratyphi GATA

-

-

TSB

30

-

-

TSB

30

-

-

TSB

30

-

-

TSB

30

B38359 S. paratyphi GATA

S. paratyphi GATA

S. paratyphi GATA

-

-

TSB

30

-

TSB

30

-

TSB

30

-

-

TSB

30

-

-

TSB

30

38383 S. enteritidis GATA

-

38055 -

ep te d

S. enteritidis GATA

M

S. enteritidis GATA

an us

B4620

33280

S. cholerasuis GATA 34088

S. cholerasuis GATA 21 a

+, presence of inhibition; -, absence of inhibition; -+,presence of weak inhibition;

Ac c

Downloaded by [Swinburne University of Technology] at 09:09 06 September 2014

B38518

cr ip t

B38470

b

MRS: de Man-Rogosa-Sharpe broth (Merck); GM17: M17+0.5% glucose; TSB:

Tryptone soy broth (Oxoid); NB: Nutrient Broth (Oxoid). ATCC: American type Culture Collection, Rockville, Maryland, USA; DSM: Deutsche Sammlung von Mikroorganismen, Braunschweig, Germany; TNO: Netherlands Organisation for Applied Scientific Research; LMG: Laboratory of Microbial Gene

24

Technology, Department of Chemistry, Biotechnology, and Food Science, Norwegian University of Life Sciences, As, Norway; RSSK: Refik Saydam National Type Culture Collection, Ankara, Turkey; GATA: Gülhane Military Medical Academy, Ankara,

cr ip t an us M ep te d Ac c

Downloaded by [Swinburne University of Technology] at 09:09 06 September 2014

Turkey.

25

Figure 1. First run of RP-HPLC profile of the first purification protocol. Bacteriocin

cr ip t an us M ep te d Ac c

Downloaded by [Swinburne University of Technology] at 09:09 06 September 2014

activity was highest in fractions 12 (6400 BU/mL).

26

Figure 2. Second run of RP-HPLC profile of the first purification protocol. Bacteriocin

cr ip t an us M ep te d Ac c

Downloaded by [Swinburne University of Technology] at 09:09 06 September 2014

activity was highest in fraction 18 (3200 BU/mL).

27

Figure 3. MALDI-TOF/MS analysis of the active fraction (fraction 18) obtained from the second run RP-HPLC of the first purification protocol. The m/z value of 5462 Da

cr ip t an us M ep te d Ac c

Downloaded by [Swinburne University of Technology] at 09:09 06 September 2014

corresponds to [M + H] + indicating a molecular mass of 5461 Da.

28

Figure 4. Reverse-phase HPLC profile of pH dependent cell adsorption-desorption extract (pH 2 extract) with Vydac C18 column incorporated in Agilent 1200 Series Quaternary HPLC system. Bacteriocin activity was detected in the fraction indicated with

cr ip t an us M ep te d Ac c

Downloaded by [Swinburne University of Technology] at 09:09 06 September 2014

arrow.

29

Figure 5. MALDI-TOF/MS analysis of the active fraction from reverse-phase HPLC of the second purification protocol. The m/z value of 5466 Da corresponds to [M + H] +

cr ip t an us M ep te d Ac c

Downloaded by [Swinburne University of Technology] at 09:09 06 September 2014

indicating a molecular mass of 5465 Da.

30

Figure 6. Effect of pH mediated cell surface adsorption-desorption fraction containing enterocin B against growing cells of L. monocytogenes RSSK 475 with 300 BU/mL final concentration. Symbols: ● and ○, cell viability (log10 CFU/mL); ▲ and Δ, Turbidity (A600 nm) (appropriate broth); ○ and Δ, culture with bacteriocin; ● and ▲, culture

cr ip t an us M

ep te d Ac c

Downloaded by [Swinburne University of Technology] at 09:09 06 September 2014

without bacteriocin.

31

Comparison of two methods for purification of enterocin B, a bacteriocin produced by Enterococcus faecium W3.

This study aimed to compare two different approaches for the purification of enterocin B from Enterococcus faecium strain W3 based on the observation ...
538KB Sizes 0 Downloads 8 Views