Loading

Journal of Human Nutrition and Food Science

In-Depth Functional Characterization of Bacillus subtilis PLSSC Revealing its Robust Probiotic Attributes

Research Article | Open Access | Volume 12 | Issue 2

  • 1. Department of Probiotics, Advanced Enzyme Technologies Ltd, India
+ Show More - Show Less
Corresponding Authors
Aruna Inamdar, Probiotics Department, Advanced Enzyme Technologies Ltd., Thane (W), Maharashtra - 400 604, India, Tel: +91-22-7506161194; 9004641267
Abstract

Functional assessment of a probiotic strain is imperative to substantiate its potential to offer health benefits to the host upon adequate consumption. Present study was designed to study probiotic attributes, safety aspects of Bacillus subtilis PLSSC in vitro with detailed investigation on antimicrobial activity, aligned to the regulatory guidance. We found that Bacillus subtilis PLSSC spore powder preparation is stable at real time ICH stability conditions for 30 months (99.71% viability). It remained viable in acid and bile stress and during in vitro simulated gastrointestinal digestion (99.48-100% viability). It tolerated thermal exposure well up to 80°C (86.18 % viability) up to 6 h and survived (100.0% viability) pasteurization. It showed excellent aqueous stability under ICH recommended storage conditions for one year with 96.21-99.9 % survival. Cell surface adhesion properties indicated affinity towards non-polar solvents and ability to aggregate with pathogens. Bacillus subtilis PLSSC showed broad spectrum antimicrobial activity including anti-listerial with 125 mg L-1 MIC. Its antimicrobial compound identified as subtilosin A (3108 Da by mass spectrometry) showed good stability in broad pH range (1.0-11.0) and temperatures (40-100°C). Peptide was sensitive to pepsin treatment but remained stable when exposed to trypsin. Thus, it showed excellent in vitro probiotic potential with well characterized antimicrobial activity as well as suitability for industrial processing implying wide applications.

Keywords

• Bacillus subtilis PLSSC

• Probiotic

• Antimicrobial

• Stability

• Subtilosin A

CITATION

Dixit Y, Bhingardeve N, InamdarA, Saroj D (2024) In-Depth Functional Characterization of Bacillus subtilis PLSSC Revealing its Robust Probiotic Attributes. J Hum Nutr Food Sci 12(1): 1182.

INTRODUCTION

Probiotics have gained considerable attention in recent years for their potential health benefits and therapeutic applications. These live microorganisms, when administered in adequate amounts, confer a beneficial effect on the host by improving the microbial balance in the gut and promoting various physiological functions [1]. Among the diverse group of probiotic bacteria, Bacillus subtilis has emerged as a promising candidate due to its remarkable probiotic attributes and well-established safety profile [2-6].

Bacillus subtilis (B. subtilis), a Gram-positive, endosporeforming bacterium, has a long history of safe use in food and feed applications [7]. It possesses several characteristics that make it an attractive probiotic candidate. First, B. subtilis exhibits robust survivability under harsh conditions, including the acidic environment of the stomach and the bile-rich environment of the small intestine, enabling it to reach the colon in viable form [8]. Second, it has a wide range of antimicrobial properties, including the production of antimicrobial peptides, enzymes, and secondary metabolites, which contribute to its ability to inhibit the growth of pathogens and modulate the gut microbiota [9]. Third, B. subtilis has demonstrated immunomodulatory effects,promoting a balanced immune response and potentially reducing the risk of certain inflammatory conditions [10,11].

Despite the extensive use of B. subtilis in various industries, comprehensive characterization and evaluation of specific strains for their probiotic potential are essential for their successful application in human health. The present study on characterization of B. subtilis PLSSC strain as a potential probiotic candidate will contribute to the growing body of knowledge on probiotics and provide insights into the selection and development of effective probiotic formulations. B. subtilis PLSSC is a safe probiotic strain with GRAS status from USFDA [7]. The strain has been proven safe for human use through a validated phase 1 clinical trial [12].

This study aims to thoroughly characterize a B. subtilis PLSSC strain and evaluate its probiotic properties, including its survival and stability in simulated gastrointestinal conditions, suitability for industrial processes, production of antimicrobial compound, and its characterization.

MATERIALS AND METHODS

Bacterial strains, media and chemicals

Bacillus subtilis PLSSC (B. subtilis PLSSC) was made available from the in-house culture collection at Advanced Enzyme Technologies ltd., Maharashtra, India. The culture was identified using 16S rRNA sequencing (Data not shown). Bacterial and fungal strains used as pathogenic strains for assessing antimicrobial activity are given in Table 1 with their growth and assay media. Cultures were maintained in 20% glycerol and stored at -80?C. B. subtilis PLSSC was revived in nutrient broth at 37 °C, 160 rpm for experimental purpose wherever applicable. All the chemicals and reagents were purchased from Sigma-Aldrich, India and microbiological media from Hi-Media Labs India Pvt. Ltd.

Spore preparation and viable spore count

Spray dried preparation of spores of B. subtilis PLSSC used in the present study was carried out at Advanced Enzyme Technologies Ltd. (India) following a proprietary good manufacturing process. Viable spore count was determined using pour plate method. Approximately, 1.0 g (or 1.0 mL) sample was withdrawn at predetermined time interval from each of the test conditions and serially diluted in tween peptone water [composition, % (w v-1): proteose peptone 1.00, sodium chloride 0.50, disodium phosphate 0.35, monosodium phosphate 0.15, and tween-80 0.20, pH 7.2±0.2]. Spores were then activated via heat shock treatment for 15 min at 70ºC and immediately cooled to about 45-50°C. Activated spore suspension (1.0 mL) was plated and poured with pre-sterilized molten nutrient agar (M001, HiMedia, Mumbai, India). Solidified media plates were incubated at 37°C for 24 h. Viable spore activity was expressed in colony forming units per mL or g (Log10CFU mL-1 or g-1) by taking the mean of three independent analyses.

Powder stability

Spray dried powder preparation of B. subtilis PLSSC spores was assessed for stability over 30 months under real time stability conditions (25±2°C, 60%±5% RH) in stability chamber [13]. Viable spore count was determined as described earlier.

Acid and bile stability

Ability of B. subtilis PLSSC spores to survive in acid and bile was studied as described by Maity C et al. [14]. Briefly, B. subtilis PLSSC spore suspension (5×108 CFU) was prepared in sterile miliQ water with pH adjusted to 1.5, 2.5, 3.0, 5.0 & 7.0. In another set, spores (2×109 CFU) were exposed to different concentrations of bile salts (SRL) as 0.01, 0.1, 0.2, 0.3, 0.5, 0.7, and 1.0%, w v-1. Both the sets were incubated at 37°C for 6 h. Viable spore count was determined every hour as described earlier.

In vitro stability by static gut model

Stability of B. subtilis PLSSC spores was assessed under simulated in vitro conditions of human gastrointestinal (GI) tract as per standard harmonized procedure based on an international consensus developed by the COST INFOGEST network [15]. Fasting (free spores) and fed conditions along with different diets- pasteurized milk (PM), powdered baby food (PBF), standard American diet (SAD) and standard European diet (SED) were created for the assessment. Briefly, the suspension (1×109 CFU mL-1) was sequentially exposed (50 rpm, 37°C) to simulated salivary fluid (oral phase = 2min, pH = 7.0) followed by simulated gastric fluid (gastric phase = 2h, pH = 3.0) and simulated intestinal fluid (intestinal phase = 2h, pH = 7.0). Samples (1 mL) were withdrawn at specific interval (up to 240 min) and analyzed as described earlier. Each experiment was carried out with freshly prepared digestive juices under aseptic conditions.

Thermal and aqueous stability

B. subtilis PLSSC spores (1×109 CFU) were tested for stability at various temperatures (0, 40, 60, 80, 90°C) for 6 h. Samples were taken every hour, immediately cooled in ice-cold water and analysed. Additionally, B. subtilis PLSSC spores (2×109 CFU) were suspended per 100 mL serving of phosphate buffer (pH 7.2 ± 0.05), milk (pH 6.7 ± 0.05), and juice (freshly prepared orange juice, pH 4.5 ± 0.05). The prepared suspensions were exposed to three different pasteurization temperatures such as 63°C, 72°C and 90°C in oil bath. Samples were taken at specific time intervals [16] and immediately cooled in ice-cold water. Viable spore count was determined as described earlier. An aqueous spore suspension of B. subtilis PLSSC (1.5×108 CFU) was prepared in sterile DW (pH 7.0) and stored in accordance with ICH guidelines [13] for refrigerated (5 ±3°C), intermediate (25 ±2°C, 60% ±5% RH) and accelerated (at 40 ±2°C, NMT 75% RH) stability. Viable spore count was determined at specific intervals and as described earlier.

Bacterial adhesion to hydrophobic solvents (BATH)

Cell surface hydrophobicity was assessed as described by Lim H et al. [17], with slight modifications. Briefly, B. subtilis PLSSC was grown in nutrient broth overnight at 37°C, centrifuged at 3500 rpm for 15 min. Cell pellet was washed twice and resuspended with phosphate buffered saline (PBS; pH 7.4) and OD600 was recorded (A0 ). Culture suspension and hydrophobic solvents (xylene, toluene, and ethyl acetate) were mixed in 1:1 proportion and vortexed for 5 min (Labquest Borosil, MTV012). The mixtures were then allowed to rest for 30 min at ambient temperature. From the two separated phases aqueous layer was removed and OD600 (A1 ) was recorded. The cell surface hydrophobicity percentage was calculated according to the formula:

Hydrophobicity\left ( % \right )=\frac{A_{0}-A_{1}}{A_{0}}\times 100

A0 = Optical density before mixing with solvents; A1 = Optical density after mixing with solvents

Aggregation and co-aggregation ability

Autoaggregation and co-aggregation was assessed as described by Lim et al. [17] with slight modifications. Briefly, B. subtilis PLSSC was grown in nutrient broth overnight at 37 °C, centrifuged at 3500 rpm for 15 min. Cell pellet was washed twice and resuspended with phosphate buffered saline (PBS; pH 7.4) and OD600 was recorded (A0). The suspension was allowed to stand at ambient temperature and OD600 was recorded at 6 h (At). The autoaggregation percentage was calculated according to the formula:

Autoaggregation\left ( % \right )=\frac{A_{0}-A_{1}}{A_{0}}\times 100

At = Optical density at 6 h at 600 nm; A0 = Optical density of 0 h at 600 nm.

To assess co-aggregation ability with pathogens, initially OD600 of suspension of B. subtilis PLSSC and pathogens under study was recorded individually. Then, equal proportion of the two was mixed. The mixtures were incubated at ambient temperature under static conditions. OD600 of the mixtures was recorded at 0 and 6 h. The co-aggregation was calculated according to the formula:

Co-aggregation(%)=(\frac{A_{path}+A_{pro}}{2}-A_{Mix})\div \left ( \frac{A_{path}+A_{Pro}}{2} \right )\times 100

Apath, Apro = Optical density of the pathogen and the probiotic strain suspension at t, respectively;

Amix = Optical density of the mixed suspension at t.

Antimicrobial activity and characterization

Screening and production of antimicrobial compound (AMC): The production of AMC by B. subtilis PLSSC was studied as described by Ahire JJ, et al. [18]. Briefly, a single axenic colony of B. subtilis PLSSC was inoculated in 10 mL of clarified Mueller Hinton (MH) Broth. After incubation at 37°C, 120 rpm for 24 h, inoculum (1%) was transferred to 100 mL clarified MH Broth and incubated under same conditions. The culture was then mixed with 4 % activated sterile XAD16N beads and evenly poured onto sterile MH agar plates. The plates were incubated at 37°C for 5 days. The beads were then collected, washed several times with MilliQ water, followed by 30 % ethanol and again with MilliQ water. The AMC was eluted with 80 % isopropanol (IPA) containing 0.1 % trifluoroacetic acid (TFA). The eluted sample was filtered (0.2 µ cellulose acetate, Axiva) and concentrated using Rotavapor (Rotavapor® R-300, Buchi, Switzerland). The antimicrobial activity of the concentrate was determined against M. luteus by spot-on-the-lawn assay [19]. Briefly, 50 µL of M. luteus culture grown overnight from single colony was mixed with 25 mL molten MH agar and poured in petri dish. After drying, 25 µL of sample was spotted onto the agar surface and the plates were incubated overnight at 37?C. Antimicrobial activity was measured as zone of inhibition in mm.

Purification of AMC: The concentrated sample showing antimicrobial activity was purified using reverse phase C18 cartridge (Sep-Pak, 35 cc Vac Cartridge, 10 g Sorbent, Waters, United States). The C18 column was preconditioned as per manufacturer’s protocol. The sample was loaded, washed with water and eluted with 10 % to 90 % gradient of IPA containing 0.1 % TFA with 10 % increment at a flow rate of 1 mL min-1. The antimicrobial activity of all the eluted fractions was determined against M. luteus by spot-on-the-lawn assay as described earlier. Eluted fractions showing antimicrobial activity were pooled together and concentrated using Rotavapor and lyophilized (Lyophilizer, Lyo Lab, United States). Lyophilized sample was stored in amber color bottle at -20?C until further use [18].

Characterization of AMC: Molecular characterization: Molecular mass of purified AMC (1 mg mL-1 in acetonitrile) was determined using triple quadrupole mass spectrometry (Shimadzu LCMS 8040). Approximately 1 mg mL-1 of lyophilized AMC was prepared in filter sterilized DW. Absorption spectrum (190 – 800 nm) was recorded at data interval of 0.2 nm and scan speed 100 nm min-1 (UV spectrophotometer JASCO V-730).

Antimicrobial spectrum and MIC: Antimicrobial activity of lyophilized AMC (5 mg mL-1) was determined against a set of 17 pathogens (Table 1) by spot-on-the-lawn assay as described earlier.

Table 1: Bacterial and fungal pathogenic strains used in the study

Pathogen

Cultivation conditions

Spot assay medium

Bacillus cereus ATCC 33019

Nutrient broth, 37 °C,

aerobic

Mueller Hinton agar

Bacillus circulans ATCC 4516

Nutrient broth, 37 °C,

aerobic

Mueller Hinton agar

Bacillus subtilis subsp. Spizizenii

ATCC 6633

Brain heart infusion broth, 37 °C, aerobic

Brain heart infusion

agar

Candida albicans ATCC 90028

Potato dextrose broth, 25 °C, aerobic

Mueller Hinton agar

Clostridium sporogenes NCIM- 5125(Equivalent to ATCC 19404)

Reinforced clostridium

medium, 37 °C, aerobic

Mueller Hinton agar

Enterobacter cloacae ATCC 13047

Nutrient broth, 37 °C,

aerobic

Mueller Hinton agar

 

Escherichia coli ATCC 700728

Nutrient broth, 37 °C,

aerobic

Mueller Hinton agar fortified with yeast extract (5 g L-1)

Escherichia coli ATCC 9002 NCTC

Nutrient broth, 37 °C,

aerobic

Mueller Hinton agar

Klebsiella pneumoniae ATCC BAA-1144

Soyabean casein digest broth

Mueller Hinton agar

Listeria monocytogenes ATCC 19115

Brain heart infusion broth, 37 °C, aerobic

Brain Heart Infusion

agar

 

Micrococcus luteus MTCC 106T

Mueller Hinton Broth, 37 °C, aerobic (MH Broth)

 

Mueller Hinton agar

Pasteurella multocida ATCC 12945

Brain heart infusion broth, 37 °C, aerobic

Brain heart infusion

agar

Pseudomonas aeruginosa ATCC 9027

Nutrient broth, 37 °C,

aerobic

Mueller Hinton agar

Salmonella abony NCIM- 2257(Equivalent to ATCC 6017 NCTC)

Nutrient broth, 37 °C,

aerobic

 

Mueller Hinton agar

Salmonella enterica ATCC 14028

Nutrient broth, 37 °C,

aerobic

Mueller Hinton agar

Staphylococcus aureus ATCC 6538P

Nutrient broth, 37 °C,

aerobic

Mueller Hinton agar

Vibrio parahaemolyticus ATCC 17802

Nutrient broth, 37 °C,

aerobic

Mueller Hinton agar fortified with yeast extract (5 g L-1)

Minimum Inhibitory Concentration (MIC) was determined against L. monocytogenes by agar dilution method according to Wiegand I et al. [20]. L. monocytogenes was grown overnight in Brain Heart Infusion Broth (BHIB) at 37?C under shaking conditions (120 rpm). Different concentrations of lyophilized AMC (1 – 1000 mg L-1) were prepared in 15 mL sterile molten MH-agar; five µL of overnight grown culture of L. monocytogenes was spotted on the agar surface. The plates were incubated at 37?C for 48 h. The lowest concentration of AMC in the agar medium that inhibited visible growth was considered MIC.

Stability of AMC: Stability of AMC was determined under various pH, temperature and enzyme conditions [18]. For pH stability, pH of DW was adjusted to different pH viz. 1,3,5,7,9,11 and 14 using 0.1 N NaOH/HCl, and mixed with 5 mg mL-1 AMC. After incubation at 37?C for 30 min, the pH was readjusted to 7.0 ± 0.2. To determine temperature stability, 1 mg mL-1 of AMC was prepared in 0.1% TFA water and kept at 40?C, 60?C, 80?C and 100?C. Samples were collected at 10, 30 and 60 min incubation. In addition, a set of samples was autoclaved at 121?C for 15 min. Stability of AMC in presence of proteolytic Enzymes namely trypsin and pepsin was determined. One mg mL-1 AMC was mixed with 1 mg mL-1 pepsin and trypsin prepared in 0.1% TFA water and Tris HCl (pH 8.0) respectively. After 4 h of incubation at 37?C, the enzyme was inactivated by heating at 95?C for 5 min, and cooled. In all the 3 sets of experiments an untreated AMC (1 mg mL-1) was used as control. Antimicrobial activity of filter sterilized (0.2µ) samples against L. monocytogenes was determined by spot-on-the-lawn assay as described earlier.

Statistical analysis

Viable activity of B. subtilis PLSSC spores was expressed as mean Log10 CFU mL-1 or g-1 of three repeated measurements. Statistical analysis was carried out using GraphPad Prism version 9.5.1 (GraphPad Software Inc., USA, https:// www.graph pad. com/ scien tific- softw are/ prism/). Significant differences between the means were calculated at p<0.05 using Twoway analysis of variance (ANOVA) followed by Tukey’s HSD or Dunnett multiple comparison test.

The whole genome sequencing data for strain B. subtilis PLSSC is submitted to GenBank with accession no. CP031129.1.

RESULTS

Powder stability During

long term storage of spray dried powder of B. subtilis PLSSC, initial viable spore count (10.41±0.78 Log10CFU g-1) reduced by 0.03 Log10CFU over 30 months (10.38±0.78 Log10CFU g-1) indicating excellent stability (Table 2).

Table 2: Real time long term stability of B. subtilis PLSSC spore powder preparation

Time (months)

Viability (Log10CFU g-1)

0 day

10.41±0.78

3

10.41±0.79

6

10.40±0.79

9

10.40±0.79

12

10.40±0.79

18

10.40±0.79

24

10.39±0.79

30

10.38±0.78

Stability data for three independent batches of B. subtilis PLSSC spore powder preparation is provided in supplementary table (Supplementary Table S1).

Table S1: Real time long term stability of B. subtilis PLSSC spore powder preparation carried out for three independent batches (Log10 CFU g-1)

Batch I

Batch II

Batch III

9.94

9.98

11.32

9.94

9.97

11.31

9.93

9.97

11.31

9.92

9.97

11.31

9.92

9.97

11.31

9.92

9.96

11.31

9.91

9.96

11.30

Acid and bile stability

B. subtilis PLSSC spores remained viable when exposed to acidic environment up to 5 h (Figure 1a).

Stability of B. subtilis PLSSC spores at (a) different pH, (b) bile(%) and (c) in simulated gastrointestinal tract as free spores; in Milk, PBF-Powdered baby  food, SAD-Standard American diet, SED-Standard European Diet.

Figure 1: Stability of B. subtilis PLSSC spores at (a) different pH, (b) bile(%) and (c) in simulated gastrointestinal tract as free spores; in Milk, PBF-Powdered baby food, SAD-Standard American diet, SED-Standard European Diet.

Viability up to 5 h was least affected at pH 7.0, 5.0 and 3.5 when compared with initial count. At pH 1.5 and 2.5, significant difference in viability was seen from 1 h (P =0.0003; P=0.0001). Five h exposure led to 1.15 and 0.47 Log10CFU reduction at pH 1.5 and 2.5 respectively, whereas 0.15-0.30 Log10CFU reduction was observed in case of pH 3.5, 5.0 and 7.0. Viability at different bile salt concentrations was maintained for B. subtilis PLSSC spores for up to 5 h (Figure 1b). Among the different concentrations studied maximum reduction in viability of 0.07 Log10CFU was seen at 1.0 % bile after 5 h; albeit negligible as compared with the initial count.

In vitro stability by static gut model

Stability under in vitro static gut model showed ability of B. subtilis PLSSC spores to survive adverse conditions in human gastrointestinal tract. Free spores remained viable under all simulated phases of GI digestion (Figure 1c). During gastric phase of digestion viability of free spores showed reduction of 0.5 Log10CFU which was statistically non-significant (P=0.5250). Presence of food matrices did not alter the viable activity; initial viability of approximately 9.00 Log10CFU was maintained until the end of the intestinal phase in presence of powdered baby food (P>0.9999), milk (P=0.9973), standard American diet (P>0.9999) and standard European diet (P=0.9973).

Thermal and aqueous stability

B. subtilis PLSSC spores exhibited thermal stability from 0 to 80°C up to 6 h (P=0.0609). The viability at 90°C significantly reduced over 6 h duration. Statistically significant reduction in viability (2.60 Log10CFU) was seen after 3 h exposure (P<0.0001). As depicted in Figure 2a viability was completely lost after 6 h (9.00 Log10CFU reduction).

Stability of B. subtilis PLSSC spores after exposure to (a) different temperatures, (b) pasteurization at 63°C in PBS(63-P), Milk(63-M), and orange juice(63- ORJ), (c) pasteurization at 72°C in PBS(72-P), Milk(72-M), and orange juice(72-ORJ), at 90°C in PBS(90-P), Milk(90-M), and orange juice(90-ORJ), (d) aqueous  stability of B. subtilis PLSSC under ICH recommended conditions.

Figure 2: Stability of B. subtilis PLSSC spores after exposure to (a) different temperatures, (b) pasteurization at 63°C in PBS(63-P), Milk(63-M), and orange juice(63- ORJ), (c) pasteurization at 72°C in PBS(72-P), Milk(72-M), and orange juice(72-ORJ), at 90°C in PBS(90-P), Milk(90-M), and orange juice(90-ORJ), (d) aqueous stability of B. subtilis PLSSC under ICH recommended conditions.

At pasteurization temperatures B. subtilis PLSSC spores remained viable in all the 3 matrices. Viability of 9.38±0.02 Log10CFU in PBS, 9.43±0.01 Log10CFU in milk and 9.43±0.02 Log10CFU in orange juice was observed after 30 min at 63°C (Figure 2b). Viable count remained 2×109 per serving for all the 3 matrices at 72°C and 90°C till 30 sec (99.8-100.0% viability) (Figure 2c). Viability of B. subtilis PLSSC spores was maintained as aqueous suspension throughout the stability duration of 1 year under ICH conditions (Figure 2d). Long term stability under shelf and refrigerated storage showed negligible Log10CFU reduction in 1 year.. No significant change in viability (0.04 Log10CFU reduction; P=0.4486) was seen during accelerated stability under refrigerated conditions [25°C ± 2°C/60% RH ± 5% RH]. Viability was affected under accelerated conditions on the shelf [40°C ± 2°C/75% RH ± 5% RH] with 0.31 Log10CFU reduction (P=0.0234) at the end of 6 months.

Aggregation ability and BATH

We observed bacterial adhesion to non-polar solvents in the range of 21-25 % with maximum adhesion to xylene and least to ethyl acetate in 6 h (Figure 3a).

Figure 3: Cell surface hydrophobicity of B. subtilis PLSSC (a) Bacterial adhesion to hydrophobic solvents, (b) autoaggregation and co-aggregation ability of B. subtilis PLSSC

We observed 23.81% autoaggregation for B. subtilis PLSSC in 6 h (Figure 3b). As depicted in Figure 3b,c co-aggregation of B. subtilis PLSSC was highest (23.11%) with S. aureus ATCC 6538P, 20.09% with C. albicans ATCC 90028 and lowest (4.48%) with B. cereus ATCC 33019.

Antimicrobial activity and characterization

Antimicrobial activity of probiotic B. subtilis PLSSC was examined in the present study. XAD16N beads acted as an adsorbent for the AMC produced by B. subtilis PLSSC. The AMC was further eluted with 80% IPA – 0.1% TFA and its rotavap concentrate showed 11 mm zone of inhibition against indicator organism M. luteus. The Sep-Pak C18 column eluted fractions with 50%- 90% IPA-TFA were found to be active fractions showing effective zones of inhibition against M. luteus (Figure 4). The active fractions were pooled, concentrated and freeze-dried to obtain 150 mg of AMC powder.

Figure 4: Antimicrobial activity of IPA extracts of antimicrobial compound produced by B. subtilis PLSSC

The UV-VIS spectrum analysis of the freeze-dried AMC powder revealed presence of absorption peaks in region 200 – 230 nm with intense peak at 221 (shoulder peak), 225 and 228 nm. Mass spectrometric analysis (LCMS) showed presence of precursor ion corresponding to 1037 m/z in positive ionization mode (Figure 5).

Mass spectrum (Triple quadruple mass spectrometry) for antimicrobial compound produced by B. subtilis PLSSC

Figure 5: Mass spectrum (Triple quadruple mass spectrometry) for antimicrobial compound produced by B. subtilis PLSSC.

Antimicrobial spectrum against 17 pathogens revealed broad spectrum antimicrobial activity of AMC produced by B. subtilis PLSSC (Table 3).

Table 3: Inhibition zones (mm) exhibited by AMC produced by B. subtilis PLSSC

Sr.

No.

Pathogen

PLSSC AMC

(5mg mL-1)

Positive control (10 µg mL-1)

1

Escherichia coli ATCC 9002 NCTC

10

15C

2

Salmonella abony NCIM-2257 (Equivalent to ATCC 6017 NCTC)

7

15C

3

Clostridium sporogenes NCIM-5125 (Equivalent to ATCC 19404)

9

16C

4

Pseudomonas aeruginosa ATCC 9027

0

4N

5

Salmonella enterica ATCC 14028

9

17C

6

Enterobacter cloacae ATCC 13047

15

7N

7

Klebsiella pneumoniae ATCC BAA-1144

10

20C

8

Escherichia coli ATCC 700728

0

18C

9

Vibrio parahaemolyticus ATCC 17802

25

12N

10

Pasteurella multocida ATCC 12945

24

33C

11

Listeria monocytogenes ATCC 19115

15

15C

112

Bacillus cereus ATCC 33019

0

12N

13

Staphylococcus aureus ATCC 6538P

14

20C

14

Candida albicans ATCC 90028

0

nd

15

Micrococcus Luteus MTCC 106T

8

19N

16

Bacillus subtilis subsp. Spizizenii ATCC 6633

13

13C

17

Bacillus circulans ATCC 4516

15

11C

C: Cefixime; N: Neomycin; nd: not done; Due to the partially purified nature of AMC slightly high concentration (5 mg.ml-1) was used for determining antimicrobial spectrum

Antimicrobial activity was not observed against C. albicans, P. aeruginosa, E. coli 700728, B. cereus while rest all pathogens were inhibited by AMC. Maximum inhibition at 5 mg mL-1 concentration was seen for E. cloacae (15 mm), L. monocytogenes (15 mm), B. circulans (15 mm), S. aureus (14 mm), and B. subtilis subsp. Spizizenii 6633 (13 mm). The MIC of AMC produced by B. subtilis PLSSC determined against L monocytogenes was found to be 125 mg L-1.

Table 4: Stability of AMC produced by B. subtilis PLSSC under conditions of extremes of pH, temperature and proteolysis

Treatment

Zone of inhibition (mm)

Activity(%)

Control (without any treatment)

14

100

Enzymes

 

Trypsin (1 mg mL-1)

14

100

Pepsin (1mg mL-1)

11

78.57

pH

 

1

14

100

3

14

100

5

15

100

7

15

100

9

14

100

11

13

92.85

14

0

0

Temperature

 

40?C, 60 min

12

85.71

60?C, 60 min

11

78.57

80?C, 60 min

11

78.57

100?C, 60 min

11

78.57

121?C, 15 min

0

0

PLSSC AMC was found to be tolerant to proteolytic treatment; when treated with trypsin 100 % activity was retained and with pepsin 78.57%. Antimicrobial activity remained unaltered in the pH range of 1 to 11. Complete inactivation of AMC occurred at pH 14.0. Thermal tolerance of AMC was also observed where antimicrobial activity was retained at temperatures 40, 60, 80 and 100?C for 60 min. Antimicrobial activity disappeared when AMC was autoclaved for 15 min at 121?C.

DISCUSSION

Probiotics are gaining a lot of attention owing to a plethora of health benefits they offer to the host upon consumption. Extensive screening is required to obtain a good probiotic candidate that can reach the target site in adequate numbers. In the present study, we substantiated probiotic characteristics of B. subtilis PLSSC in vitro and assessed its antimicrobial potential followed by characterizing the AMC.

Spray dried powder of B. subtilis PLSSC spores showed excellent stability and 100.00 % viability (Log10CFU) was maintained for ICH stability conditions for 30 months. Stability of B. subtilis PLSSC spores is similar to the previously reported stability for other probiotic strains of B. subtilis- DE111 [4], SG 188 [5], MB40 [6]. Good stability of a probiotic strain eliminates the need to add overages into formulations for probiotics to reach in adequate numbers at the target site in vivo. Oral probiotics encounter exposure to gastric acid and bile during their passage from oral cavity to colon. Hence, it is desirable for a good probiotic strain to be able to withstand the stress. Acid and bile stress can adversely affect bacterial survival via proton accumulation and disruption of pH gradient, disruption of essential enzyme functions or macromolecules such as DNA [21]. In the present study, acid and bile exposure did not affect the viability of B. subtilis PLSSC from pH 3.5 to 7.0; we observed minor reduction similar to the literature reports at pH 1.5 and 2.5 [17-23]. Other B. subtilis strains have shown roughly 0.10 to 0.60 Log10CFU reduction [22]. About 90 and 40% survival has been reported for B. subtilis MKHJ 1-1 when exposed to pH 2.5 and 0.3% bile salt [17]. B. subtilis FTC01 survived to 80 and 96% at pH 2 and 3 for 2 h [23].

B. subtilis PLSSC spores in presence of various food matrices (mimicking fed conditions) as well as free spores (fasting condition) remained viable throughout all the phases of simulated GI digestion. B. subtilis MA139 [24] and few other strains of B. subtilis have been studied for their tolerance and stability in simulated gastrointestinal conditions and were found to remain unaffected by gastric or intestinal conditions [25,26].

Temperature tolerance of B. subtilis PLSSC was found to be reasonably good as it maintained viability (99.33%) up to high temperatures of 80°C for 6 h. Guo X, et al. [22] have also reported similar tolerance for four B. subtilis strains but for a shorter exposure of 5 to 10 min. Viability at pasteurization temperatures (63,72,90°C) in 3 matrices (PBS, milk, orange juice) was 100.0%. Our results are in agreement with a recent report by Mazhar S et al. [27] on Bacillus subtilis DE111® spores’ viability across three pasteurization processing temperatures (45, 75, and 90°C) in PBS, Oat milk and Apple juice. Our results indicate that B. subtilis PLSSC can be considered to be the ideal probiotic candidate for incorporation in foods and nutraceuticals undergoing high temperature processing.

Our results indicate ability of B. subtilis PLSSC spores to remain viable in aqueous environment under humid conditions for one year. It implies potential to incorporate the strain in aqueous suspensions. The primary reason for a good stability under adverse environmental or processing conditions is the ability of B. subtilis PLSSC to form endospores. The combination of protective layers, reduced metabolic activity, and molecular mechanisms for DNA protection and repair enables spores to withstand the harshest environments until conditions become favorable for germination and growth [28].

BATH, autoaggregation and co-aggregation ability of B. subtilis PLSSC strongly indicate its adherence and survival potential in the gut. BATH as reported by other researchers is comparable to our results; it varies from 5 to 57% with other strains of B. subtilis and non-polar solvents [17,23]. Autoaggregation ability enables a probiotic strain to establish itself in the intestinal lumen. B. subtilis PLSSC showed comparable autoaggregation and co-aggregation ability. The variation in aggregation abilities arises as this property is strain dependent; it ranges from 63% in 5 h to 88% in 4 h for various other strains of B. subtilis [17,29].

Co-aggregation of B. subtilis P223 with S. aureus ATCC 6538P (57.09%) and B. subtilis MKHJ 1-1 with S. aureus KCTC 3881 (24.75%) is reported in the literature [17,21].

Ability to antagonize pathogens is a well established mechanism through which probiotics offer protection to the host from various infections. B. subtilis is known to produce plethora of AMCs of various types including ribosomal peptides (RPs), volatile compounds, polyketides (PKs), non-ribosomal peptides (NRPs), and hybrids between PKs and NRPs [9]. We studied the ability of B. subtilis PLSSC to produce AMC on solid media. Adsorptive polymeric resin Amberlite® XAD16N was used for their property to retain hydrophobic compounds. It is a nonionic, hydrophobic, crosslinked polymer with macroporous structure, high surface area, and the aromatic nature. With its characteristic pore size distribution, it is known to adsorb hydrophobic molecules from polar solvents such as recovery and purification of antimicrobial substances from fermentation broth [30,31]. The compounds adsorbed onto XAD16N were eluted with 80% IPA-0.1% TFA which showed zone of inhibition confirming the antimicrobial nature of the extracted compounds. AMC was purified by column chromatography and lyophilized for further analysis. Khochamit N et al. [30], and Epparti P et al. [32], have reported triply charged precursor ions of 1134.863+ m/z and 1140.193+ m/z respectively, corresponding to subtilosin A by quadrupole-time-of-flight (QTOF) analysis. We obtained precursor ion with 1037 m/z in LC-MS using triple quadruple as a detector. This precursor ion could be triply charged computing to molecular mass of 3111 corresponding to subtilosin A. However, multiple charges cannot be confirmed using triple quadruple detector as it is limited by unit mass resolution. For accurate mass determination and compound identification High Resolution Mass Spectrometry (HRMS) technology (Q-TOF, MALDI-TOF) would be required. Additionally, our results are supported by whole genome sequencing data for B. subtilis PLSSC (GenBank: CP031129.1) where genes for subitlosin A family bacteriocin and subtilosin A maturase are annotated. Subtilosin A is a sactipeptide produced by B. subtilis known to possess antibacterial activity against both Gram-positive and Gram-negative pathogens. Subtilosin A attaches to a membrane receptor, and binds plasma membrane via electrostatic interaction. Transmembrane pH gradient gets disrupted as a result of electrostatic binding and effluxes intracellular ATP. Eventually cell dies due to starvation. Subtilosin A can block quorum sensing and inhibit biofilm formation [33].

Subtilosin A produced by B. subtilis PLSSC showed broad spectrum antimicrobial activity. Notably, it was able to inhibit Gram positive food borne pathogen Listeria monocytogenes and is a prominent characteristic of our strain B. subtilis PLSSC. Additionally, subtilosin A showed clear inhibition of two poultry pathogens E. coli ATCC 9002 NCTC and S. enterica ATCC 14028 indicating its potential application in poultry feed. Bacteriocins bind to specific receptors in the target cells prior to initiating antimicrobial effect [34]. Precisely for this reason, though broad spectrum, strain specificity has been noted for each independent bacteriocin. Partly, this could be the reason in differences in antimicrobial effect seen for different strains of the same species of a target pathogen e.g. E. coli ATCC 9002 NCTC (Poultry pathogen) and Escherichia coli ATCC 700728 (Human pathogen) used in the present study. Broad-spectrum antimicrobial activity of B. subtilis and anti-listerial potential of subtilosin A is well established [23, 27, reviewed in 35, 36]. MIC of subtilosin A against L. monocytogenes was found to be 125 µg mL-1 which is higher than the MIC reported in previous literature (19-26 µg mL-1) and could be due to its partially purified nature [32,37]. Subtilosin A exhibited good antimicrobial activity across a broad pH range including pH as low as 1.0 and up to pH 11.0. This trend is similar to the one reported by Karagiota A et al. [38] where AMC fractions from B. subtilis subsp. subtilis NCIB 3610 showed stable antimicrobial activity on acidic range of pH but was adversely affected at extreme alkaline pH such as pH 10.0. Similar observation is showed in other reports [39,40]. Thermal stability for subtilosin A coincides with earlier reports [38, 39]. We observed no activity for subtilosin A after autoclaving. Kim SY et al. [41], has also reported complete loss of antimicrobial activity for subtilin KU43 when autoclaved. On the contrary there are few reports of B. subtilis AMCs namely subtilin JS-4 [42], Subpeptin JM-4A & B [43] to have retained their activity after autoclaving. The AMC produced by B. subtilis PLSSC was resistant to proteolytic enzymes trypsin and pepsin; it was more sensitive to pepsin than trypsin indicating its peptide nature. Ample of evidence on adverse effect of pepsin on AMC produced by B. subtilis is available [30,38,39,42].

CONCLUSION

The present study comprised of probiotic characterization of B. subtilis PLSSC strain in terms of its ability to survive adverse gastrointestinal conditions to reach colon in adequate amounts. Its stability during thermal processing and ICH recommended storage was established. The strain was also studied for production of AMC which was identified as subtilosin A. Antimicrobial spectrum and stability of subtilosin A was also assessed. The findings of this study will provide valuable information for further research and contribute to the development of evidencebased probiotic interventions for promoting human health and well-being .

ACKNOWLEDGEMENTS

Authors gratefully acknowledge the support of Mr. V.L. Rathi and Mr. M. Kabra at Advanced Enzyme Technologies Ltd., India. Authors thank Mr. Atul Biyani for his support and contribution in long term stability studies for spore powder. Authors are also thankful to Shimadzu Analytical Pvt. Ltd. (India) for their assistance in molecular mass determination.

AUTHOR CONTRIBUTION

Project was conceived and manuscript was reviewed by Dina Saroj. Manuscript was reviewed and revised by Aruna Inamdar. Experiments and data collection was performed by Yogini Dixit, Namrata Bhingardeve. Data was analysed and first draft of the manuscript was written by Yogini Dixit. All authors have read and agreed to the manuscript.

DATA AVAILABILITY

All data supporting the findings of this study are available within the paper and its Supplementary Information.

REFERENCES
  1. Hill C, Guarner F, Reid G, Gibson GR, Merenstein DJ, Pot B, et al. Expert consensus document: The International Scientific Association for Probiotics and Prebiotics consensus statement on the scope and appropriate use of the term probiotic. Nat Rev Gastroenterol Hepatol. 2014: 506–514.
  2. GRN 1007 (FDA’s letter pending). Determination for the Use of Bacillus subtilis Strain R0179.
  3. GRN 969 (2021) Bacillus subtilis “Bss-19” spore preparation.
  4. GRN 831 (2019) Bacillus subtilis DE111.
  5. GRN 905 (2020) Bacillus subtilis SG188.
  6. GRN 955 (2021) Bacillus subtilis strain BS-MB40 PTA-122264 spore preparation.
  7. GRN 956 (2021) Bacillus subtilis ATCC SD-7280
  8. Gao C, Major A, Rendon D, Lugo M, Jackson V, Shi Z, Mori-Akiyama Y, Versalovic J. Histamine H2 receptor-mediated suppression of intestinal inflammation by probiotic Lactobacillus reuteri. MBio. 2015; 6: 10-128.
  9. Caulier S, Nannan C, Gillis A, Licciardi F, Bragard C, Mahillon J. Overview of the antimicrobial compounds produced by members of the Bacillus subtilis group. Front Microbiol. 2019; 10: 302.
  10. Freedman KE, Hill JL, Wei Y, Vazquez AR, Grubb DS, Trotter RE, et al. Examining the Gastrointestinal and Immunomodulatory Effects of the Novel Probiotic Bacillus subtilis DE111. Int J Mol Sci. 2021; 22: 2453.
  11. Lee NK, Kim WS, Paik HD. Bacillus strains as human probiotics: characterization, safety, microbiome, and probiotic carrier. Food Sci Biotechnol. 2019; 28: 1297-1305.
  12. Clinical safety study of Bacillus subtilis PLSSC probiotic. International Clinical Trial Registry platform. 2023
  13. Guideline ICH. Stability testing of new drug substances and products. Q1A (R2), current step. 2003 Feb; 4 (1-24). Accessed 11 January 2022
  14. Maity C, Gupta AK, Saroj DB, Biyani A, Bagkar P, Kulkarni J, et al. Impact of a Gastrointestinal Stable Probiotic Supplement Bacillus coagulans LBSC on Human Gut Microbiome Modulation. J Diet Suppl. 2021; 18: 577-596.
  15. Brodkorb A, Egger L, Alminger M, Alvito P, Assunção R, Ballance S, et al. INFOGEST static in vitro simulation of gastrointestinal food digestion. Nat Protoc. 2019; 14: 991-1014.
  16. International Dairy Foods Association (IDFA). Pasteurization. 2023.
  17. Lim H, Oh S, Yu S, Kim M. Isolation and characterization of probiotic Bacillus subtilis MKHJ 1-1 possessing L-asparaginase activity. Appl Sci. 2021; 11: 4466.
  18. Ahire JJ, Kashikar MS, Lakshmi SG, Madempudi R. Identification and characterization of antimicrobial peptide produced by indigenously isolated Bacillus paralicheniformis UBBLi30 strain. 3 Biotech. 2020; 10: 112.
  19. Sabo SD, Mendes MA, Araujo ED, Muradian LB, Makiyama EN, LeBlanc JG, et al. Bioprospecting of probiotics with antimicrobial activities against Salmonella Heidelberg and that produce B-complex vitamins as potential supplements in poultry nutrition. Scientific reports. 2020; 10: 7235.
  20. Wiegand I, Hilpert K, Hancock RE. Agar and broth dilution methods to determine the minimal inhibitory concentration (MIC) of antimicrobial substances. Nature protocols. 2008; 3: 163-75.
  21. Guan N, Liu L. Microbial response to acid stress: mechanisms and applications. Appl Microbiol Biotechnol. 2020; 104: 51-65.
  22. Guo X, Chen DD, Peng KS, Cui ZW, Zhang XJ, Li S, et al. Identification and characterization of Bacillus subtilis from grass carp (Ctenopharynodon idellus) for use as probiotic additives in aquatic feed. Fish Shellfish Immunol. 2016; 52: 74-84.
  23. Ritter AC, Paula A, Correa F, Veras FF, Brandelli A. Characterization of Bacillus subtilis available as probiotics. Journal of Microbiology Research. 2018; 8: 23-32.
  24. Guo X, Li D, Lu W, Piao X, Chen X. Screening of Bacillus strains as potential probiotics and subsequent confirmation of the in vivo effectiveness of Bacillus subtilis MA139 in pigs. Antonie van leeuwenhoek. 2006; 90: 139-146.
  25. Soares MB, Martinez RC, Pereira EP, Balthazar CF, Cruz AG, Ranadheera CS, et al. The resistance of Bacillus, Bifidobacterium, and Lactobacillus strains with claimed probiotic properties in different food matrices exposed to simulated gastrointestinal tract conditions. Food research international. 2019; 125: 108542.
  26. Duc LH, Hong HA, Cutting SM. Germination of the spore in the gastrointestinal tract provides a novel route for heterologous antigen delivery. Vaccine. 2003; 21: 4215-4224.
  27. Mazhar S, Khokhlova E, Colom J, Simon A, Deaton J, Rea K. In vitro and in silico assessment of probiotic and functional properties of Bacillus subtilis DE111®. Frontiers in Microbiology. 2023; 13: 1101144.
  28. McKenney PT, Driks A, Eichenberger P. The Bacillus subtilis endospore: assembly and functions of the multilayered coat. Nat Rev Microbiol. 2013; 11: 33-44.
  29. Jeon HL, Lee NK, Yang SJ, Kim WS, Paik HD. Probiotic characterization of Bacillus subtilis P223 isolated from kimchi. Food Sci Biotechnol. 2017; 26: 1641-1648.
  30. Khochamit N, Siripornadulsil S, Sukon P, Siripornadulsil W. Antibacterial activity and genotypic–phenotypic characteristics of bacteriocin-producing Bacillus subtilis KKU213: potential as a probiotic strain. Microbiological Research. 2015; 170: 36-50.
  31. AmberLite™ XAD™16N Polymeric Adsorbent. DuPont Product data sheet. 2020.
  32. Epparti P, Eligar SM, Sattur AP, Kumar BG, Halami PM. Characterization of dual bacteriocins producing Bacillus subtilis SC3.7 isolated from fermented food. LWT. 2022; 154: 112854.
  33. Tran C, Cock IE, Chen X, Feng Y. Antimicrobial Bacillus: metabolites and their mode of action. Antibiotics. 2022; 11: 88.
  34. Weaver S, Casamassima E, Nguyen AE, Baltrus DA. Broad intraspecies killing activity in Pseudomonas syringae due to the combinatorial action of LPS-interacting bacteriocins. bioRxiv. 2023.
  35. Nayak SK. Multifaceted applications of probiotic Bacillus species in aquaculture with special reference to Bacillus subtilis. Reviews in Aquaculture. 2021; 13: 862-906.
  36. Ramachandran R, Chalasani AG, Lal R, Roy U. A Broad-Spectrum Antimicrobial Activity of Bacillus subtilis RLID 12.1. The scientific world journal. 2014.
  37. Van Kuijk S, Noll KS, Chikindas ML. The species-specific mode of action of the antimicrobial peptide subtilosin against Listeria monocytogenes Scott A. Letters in applied microbiology. 2012; 54: 52-58.
  38. Karagiota A, Tsitsopoulou H, Tasakis RN, Zoumpourtikoudi V, Touraki M. Characterization and quantitative determination of a diverse group of Bacillus subtilis subsp. subtilis NCIB 3610 antibacterial peptides. Probiotics & Antimicro. Prot. 2021; 13: 555-570.
  39. Sutyak KE, Wirawan RE, Aroutcheva AA, Chikindas ML. Isolation of the Bacillus subtilis antimicrobial peptide subtilosin from the dairy product-derived Bacillus amyloliquefaciens. J Appl Microbiol. 2008; 104: 1067-1074.
  40. Liu X, Lee JY, Jeong SJ, Cho KM, Kim GM, Shin JH, et al. Properties of a bacteriocin produced by Bacillus subtilis EMD4 isolated from ganjang (soy sauce). J Microbiol Biotechnol. 2015; 25: 1493-1501.
  41. Kim SY, Lee NK, Han EJ, Paik HD. Characterization of subtilin KU43 produced by Bacillus subtilis KU43 isolated from traditional Korean fermented foods. Food Science and Biotechnology. 2012; 21: 1433-1438.
  42. Wei Z, Shan C, Zhang L, Wang Y, Xia X, Liu X, et al. A novel subtilin- like lantibiotics subtilin JS-4 produced by Bacillus subtilis JS-4, and its antibacterial mechanism against Listeria monocytogenes. LWT. 2021; 142: 110993.
  43. Wu S, Jia S, Sun D, Chen M, Chen X, Zhong J, et al. Purification and characterization of two novel antimicrobial peptides subpeptin JM4-A and subpeptin JM4-B produced by Bacillus subtilis JM4. Current microbiology. 2005; 51: 292-296.

Dixit Y, Bhingardeve N, InamdarA, Saroj D (2024) In-Depth Functional Characterization of Bacillus subtilis PLSSC Revealing its Robust Probiotic Attributes. J Hum Nutr Food Sci 12(1): 1182.

Received : 25 Jan 2024
Accepted : 28 Feb 2024
Published : 29 Feb 2024
Journals
Annals of Otolaryngology and Rhinology
ISSN : 2379-948X
Launched : 2014
JSM Schizophrenia
Launched : 2016
Journal of Nausea
Launched : 2020
JSM Internal Medicine
Launched : 2016
JSM Hepatitis
Launched : 2016
JSM Oro Facial Surgeries
ISSN : 2578-3211
Launched : 2016
JSM Regenerative Medicine and Bioengineering
ISSN : 2379-0490
Launched : 2013
JSM Spine
ISSN : 2578-3181
Launched : 2016
Archives of Palliative Care
ISSN : 2573-1165
Launched : 2016
JSM Nutritional Disorders
ISSN : 2578-3203
Launched : 2017
Annals of Neurodegenerative Disorders
ISSN : 2476-2032
Launched : 2016
Journal of Fever
ISSN : 2641-7782
Launched : 2017
JSM Bone Marrow Research
ISSN : 2578-3351
Launched : 2016
JSM Mathematics and Statistics
ISSN : 2578-3173
Launched : 2014
Journal of Autoimmunity and Research
ISSN : 2573-1173
Launched : 2014
JSM Arthritis
ISSN : 2475-9155
Launched : 2016
JSM Head and Neck Cancer-Cases and Reviews
ISSN : 2573-1610
Launched : 2016
JSM General Surgery Cases and Images
ISSN : 2573-1564
Launched : 2016
JSM Anatomy and Physiology
ISSN : 2573-1262
Launched : 2016
JSM Dental Surgery
ISSN : 2573-1548
Launched : 2016
Annals of Emergency Surgery
ISSN : 2573-1017
Launched : 2016
Annals of Mens Health and Wellness
ISSN : 2641-7707
Launched : 2017
Journal of Preventive Medicine and Health Care
ISSN : 2576-0084
Launched : 2018
Journal of Chronic Diseases and Management
ISSN : 2573-1300
Launched : 2016
Annals of Vaccines and Immunization
ISSN : 2378-9379
Launched : 2014
JSM Heart Surgery Cases and Images
ISSN : 2578-3157
Launched : 2016
Annals of Reproductive Medicine and Treatment
ISSN : 2573-1092
Launched : 2016
JSM Brain Science
ISSN : 2573-1289
Launched : 2016
JSM Biomarkers
ISSN : 2578-3815
Launched : 2014
JSM Biology
ISSN : 2475-9392
Launched : 2016
Archives of Stem Cell and Research
ISSN : 2578-3580
Launched : 2014
Annals of Clinical and Medical Microbiology
ISSN : 2578-3629
Launched : 2014
JSM Pediatric Surgery
ISSN : 2578-3149
Launched : 2017
Journal of Memory Disorder and Rehabilitation
ISSN : 2578-319X
Launched : 2016
JSM Tropical Medicine and Research
ISSN : 2578-3165
Launched : 2016
JSM Head and Face Medicine
ISSN : 2578-3793
Launched : 2016
JSM Cardiothoracic Surgery
ISSN : 2573-1297
Launched : 2016
JSM Bone and Joint Diseases
ISSN : 2578-3351
Launched : 2017
JSM Bioavailability and Bioequivalence
ISSN : 2641-7812
Launched : 2017
JSM Atherosclerosis
ISSN : 2573-1270
Launched : 2016
Journal of Genitourinary Disorders
ISSN : 2641-7790
Launched : 2017
Journal of Fractures and Sprains
ISSN : 2578-3831
Launched : 2016
Journal of Autism and Epilepsy
ISSN : 2641-7774
Launched : 2016
Annals of Marine Biology and Research
ISSN : 2573-105X
Launched : 2014
JSM Health Education & Primary Health Care
ISSN : 2578-3777
Launched : 2016
JSM Communication Disorders
ISSN : 2578-3807
Launched : 2016
Annals of Musculoskeletal Disorders
ISSN : 2578-3599
Launched : 2016
Annals of Virology and Research
ISSN : 2573-1122
Launched : 2014
JSM Renal Medicine
ISSN : 2573-1637
Launched : 2016
Journal of Muscle Health
ISSN : 2578-3823
Launched : 2016
JSM Genetics and Genomics
ISSN : 2334-1823
Launched : 2013
JSM Anxiety and Depression
ISSN : 2475-9139
Launched : 2016
Clinical Journal of Heart Diseases
ISSN : 2641-7766
Launched : 2016
Annals of Medicinal Chemistry and Research
ISSN : 2378-9336
Launched : 2014
JSM Pain and Management
ISSN : 2578-3378
Launched : 2016
JSM Women's Health
ISSN : 2578-3696
Launched : 2016
Clinical Research in HIV or AIDS
ISSN : 2374-0094
Launched : 2013
Journal of Endocrinology, Diabetes and Obesity
ISSN : 2333-6692
Launched : 2013
Journal of Substance Abuse and Alcoholism
ISSN : 2373-9363
Launched : 2013
JSM Neurosurgery and Spine
ISSN : 2373-9479
Launched : 2013
Journal of Liver and Clinical Research
ISSN : 2379-0830
Launched : 2014
Journal of Drug Design and Research
ISSN : 2379-089X
Launched : 2014
JSM Clinical Oncology and Research
ISSN : 2373-938X
Launched : 2013
JSM Bioinformatics, Genomics and Proteomics
ISSN : 2576-1102
Launched : 2014
JSM Chemistry
ISSN : 2334-1831
Launched : 2013
Journal of Trauma and Care
ISSN : 2573-1246
Launched : 2014
JSM Surgical Oncology and Research
ISSN : 2578-3688
Launched : 2016
Annals of Food Processing and Preservation
ISSN : 2573-1033
Launched : 2016
Journal of Radiology and Radiation Therapy
ISSN : 2333-7095
Launched : 2013
JSM Physical Medicine and Rehabilitation
ISSN : 2578-3572
Launched : 2016
Annals of Clinical Pathology
ISSN : 2373-9282
Launched : 2013
Annals of Cardiovascular Diseases
ISSN : 2641-7731
Launched : 2016
Journal of Behavior
ISSN : 2576-0076
Launched : 2016
Annals of Clinical and Experimental Metabolism
ISSN : 2572-2492
Launched : 2016
Clinical Research in Infectious Diseases
ISSN : 2379-0636
Launched : 2013
JSM Microbiology
ISSN : 2333-6455
Launched : 2013
Journal of Urology and Research
ISSN : 2379-951X
Launched : 2014
Journal of Family Medicine and Community Health
ISSN : 2379-0547
Launched : 2013
Annals of Pregnancy and Care
ISSN : 2578-336X
Launched : 2017
JSM Cell and Developmental Biology
ISSN : 2379-061X
Launched : 2013
Annals of Aquaculture and Research
ISSN : 2379-0881
Launched : 2014
Clinical Research in Pulmonology
ISSN : 2333-6625
Launched : 2013
Journal of Immunology and Clinical Research
ISSN : 2333-6714
Launched : 2013
Annals of Forensic Research and Analysis
ISSN : 2378-9476
Launched : 2014
JSM Biochemistry and Molecular Biology
ISSN : 2333-7109
Launched : 2013
Annals of Breast Cancer Research
ISSN : 2641-7685
Launched : 2016
Annals of Gerontology and Geriatric Research
ISSN : 2378-9409
Launched : 2014
Journal of Sleep Medicine and Disorders
ISSN : 2379-0822
Launched : 2014
JSM Burns and Trauma
ISSN : 2475-9406
Launched : 2016
Chemical Engineering and Process Techniques
ISSN : 2333-6633
Launched : 2013
Annals of Clinical Cytology and Pathology
ISSN : 2475-9430
Launched : 2014
JSM Allergy and Asthma
ISSN : 2573-1254
Launched : 2016
Journal of Neurological Disorders and Stroke
ISSN : 2334-2307
Launched : 2013
Annals of Sports Medicine and Research
ISSN : 2379-0571
Launched : 2014
JSM Sexual Medicine
ISSN : 2578-3718
Launched : 2016
Annals of Vascular Medicine and Research
ISSN : 2378-9344
Launched : 2014
JSM Biotechnology and Biomedical Engineering
ISSN : 2333-7117
Launched : 2013
Journal of Hematology and Transfusion
ISSN : 2333-6684
Launched : 2013
JSM Environmental Science and Ecology
ISSN : 2333-7141
Launched : 2013
Journal of Cardiology and Clinical Research
ISSN : 2333-6676
Launched : 2013
JSM Nanotechnology and Nanomedicine
ISSN : 2334-1815
Launched : 2013
Journal of Ear, Nose and Throat Disorders
ISSN : 2475-9473
Launched : 2016
JSM Ophthalmology
ISSN : 2333-6447
Launched : 2013
Journal of Pharmacology and Clinical Toxicology
ISSN : 2333-7079
Launched : 2013
Annals of Psychiatry and Mental Health
ISSN : 2374-0124
Launched : 2013
Medical Journal of Obstetrics and Gynecology
ISSN : 2333-6439
Launched : 2013
Annals of Pediatrics and Child Health
ISSN : 2373-9312
Launched : 2013
JSM Clinical Pharmaceutics
ISSN : 2379-9498
Launched : 2014
JSM Foot and Ankle
ISSN : 2475-9112
Launched : 2016
JSM Alzheimer's Disease and Related Dementia
ISSN : 2378-9565
Launched : 2014
Journal of Addiction Medicine and Therapy
ISSN : 2333-665X
Launched : 2013
Journal of Veterinary Medicine and Research
ISSN : 2378-931X
Launched : 2013
Annals of Public Health and Research
ISSN : 2378-9328
Launched : 2014
Annals of Orthopedics and Rheumatology
ISSN : 2373-9290
Launched : 2013
Journal of Clinical Nephrology and Research
ISSN : 2379-0652
Launched : 2014
Annals of Community Medicine and Practice
ISSN : 2475-9465
Launched : 2014
Annals of Biometrics and Biostatistics
ISSN : 2374-0116
Launched : 2013
JSM Clinical Case Reports
ISSN : 2373-9819
Launched : 2013
Journal of Cancer Biology and Research
ISSN : 2373-9436
Launched : 2013
Journal of Surgery and Transplantation Science
ISSN : 2379-0911
Launched : 2013
Journal of Dermatology and Clinical Research
ISSN : 2373-9371
Launched : 2013
JSM Gastroenterology and Hepatology
ISSN : 2373-9487
Launched : 2013
Annals of Nursing and Practice
ISSN : 2379-9501
Launched : 2014
JSM Dentistry
ISSN : 2333-7133
Launched : 2013
Author Information X