Cervicovaginal microflora isolated from healthy women exhibited probiotic properties and antimicrobial activity against pathogens isolated from cervical cancer patients

DOI: https://doi.org/10.21203/rs.3.rs-1418233/v1

Abstract

Abnormal cervicovaginal microflora plays an important role in HPV persistence and progression to cervical cancer. The present study aimed at isolating and identifying probiotics from vaginal swabs of healthy women and evaluating their activity against vaginal pathogens isolated from cervical cancer patients. Based on probiotic, acid-bile tolerance and antimicrobial properties, 13 lactic acid bacteria (LAB) from the healthy group were identified by MALDI TOF MS. Among these, four strains, Lactobacillus gasseri P36Mops, Limosilactobacillus fermentum P37Mws, Lactobacillus delbrueckii P31Mcs and Enterococcus faecium P26Mcm, exhibited significant antimicrobial activity against 8 vaginal pathogens (Staphylococcus haemolyticus P41Tcs, Escherichia coli P30Tcs, E. coli P79Bcm, Enterococus faecalis P29Mops, E. faecalis P50Tws, E. faecalis P68Tcb, S. haemolyticus P48Bcb and S. haemolyticus P58Bcb) isolated from precancerous and cervical cancer patients. 16S rRNA sequencing of four potential probiotics revealed congruency with the MALDI-TOF MS (Matrix Assisted Laser Desorption and Ionisation, Time Of Flight Mass Spectrometry) identification and phylogenetic analysis showed genetic relationship with previously reported LAB strains. The selected LAB showed strain specific hydrophobicity (35.88-56.70%), auto-aggregation (35.26-61.39%) and antibiotic susceptibility. Interestingly, L. gasseri P36Mops was resistant to five standard antibiotics routinely used against urogenital or vaginal infections. LCMS (Liquid Chromatography Mass Spectrometry) analyses of the CFS (cell-free supernatant) of the four probiotics revealed presence of metabolites  such as N-(1-deoxy-1-fructosyl)valine, hygroline, acetoxy-2-hydroxy-16-heptadecen-4-one, avocadyne 4-acetate, avocadyne 2-acetate, taraxinic acid glucosyl ester, 6-hydroxypentadecanedioic acid, with reported antimicrobial activity. The overall data suggests the bio-therapeutic potential of the identified vaginal probiotics against cervical cancer associated pathogens.

Introduction

Cervical cancer is the second leading cause of cancer death among Indian women (Siegel et al. 2018). Infection with high-risk human papilloma viruses (HPVs) such as HPV16 and 18 is the major cause for developing cervical cancer (Siegel et al. 2018). Majority of such infections do not progress to cancer, but instead get cleared spontaneously. Recent evidences suggest that the local cervico-vaginal microbiota play a critical role in development of pre-cancerous cervical intraepithelial neoplasia (CIN) and progression to invasive cervical carcinoma (ICC).

Healthy vaginal microflora is mostly dominated by beneficial microbes such as Lactobacillus and opportunistic pathogens. Changes in the vaginal microflora could modulate immune responses and favour the growth of pathogens, thereby, facilitating development of several diseases (Kalia et al. 2020; Mitra et al. 2020). Depletion of beneficial Lactobacillus could increase infection with diverse aerobic (Staphylococcus sp, Pseudomonas sp. E coli, and E. faecalis) and anaerobic (Gardnerella sp., Atopobium sp., Eggerthella sp., Sneathia sp. and Prevotella sp.) bacteria. Such vaginal dysbiosis can lead to either bacterial vaginosis (BV) and/ or invasive cervical carcinoma (ICC) (Biswal et al. 2014; Mitra et al. 2020). The exact role of pathogenic bacteria in cervical carcinogenesis is unknown, however, it is anticipated that they produce toxins and carcinogenic metabolites (nitrosamines) that could trigger increased production of proinflammatory cytokines and DNA adducts, which could lead to CIN and ICC (Kyrgiou et al. 2017).

The current standard of care (SOC) drugs for treating vaginal infections during cervical cancer therapy usually include metronidazole or clindamycin, norfloxacin and cotrimoxazole (Mulu et al. 2015; Petrina et al. 2017; Thulkar et al. 2012; Turovskiy et al. 2009). The indiscriminate use of antibiotics mostly results into development of resistant micro-organisms (Ahire et al. 2021), thereby increasing the risk of secondary infections in the cervical cancer patients (Gao et al. 2020). Use of antibiotics may also be associated with vaginal pain, metrorrhagia, nausea, vomiting, diarrhoea and renal failure (Mubarak and Kazi 2014). To overcome such issues, probiotics are being used as adjuncts during treatment of cervical cancer associated bacterial infections (Mejía-Caballero et al. 2021) or BV (Happel et al. 2020; Mejía-Caballero et al. 2021). In the present study, we have isolated and identified vaginal Lactobacillus and non-Lactobacillus (Enterococcus) species from healthy women. The isolates were evaluated for probiotic potential and antibacterial activity against vaginal pathogens isolated from cervical cancer patients.

Materials And Method

Subject selection and sampling

The study was approved by Institutional Ethics Committees of Bharati Vidyapeeth (Deemed to be) University Medical College (BVMC) (Ref: BVDU/MC/57), and B. J. Government Medical College and Sassoon General Hospitals (BJGMCSGH), Pune (Ref No. BJGMC/IEC/Pharmac/ND-Dept 0119007-007). Vaginal swabs from healthy (n꓿45); low grade squamous intraepithelial lesion, LSIL (n=1); high grade squamous intraepithelial lesion, HSIL (n=1) and invasive cervical carcinoma ICC (n꓿6) patients (aged between 18-55 years) were collected by the clinicians at the respective clinical sites. All the patients provided informed written consent. The patients were screened for eligibility criteria based upon Pap test, vaginal cytology or colposcopy findings and were categorized under healthy, LSIL, HSIL and ICC groups. Post collection, the samples were immediately placed on ice and taken to IRSHA for further analysis within 2 hours of the collection.

Culture and reagents

The clinical strain, Pseudomonas aeruginosa MCC 2081, was procured from National Centre for Microbial Resource (NCMR), NCCS, Pune. All the media components such as de Man, Rogosa and Sharpe (MRS), Tryptone Soya (TSA) or Brain Heart Infusion (BHI), L-cysteine, Bile, Sheep blood agar plates were purchased from HiMedia Laboratories, Mumbai, India. AnaeroGasPak, antibiotics discs and sterile cotton swabs were procured from Himedia Laboratories, Mumbai, India. Pepsin and pancreatin were purchased from Sigma‑Aldrich (St. Louis, MO, USA). Plasticware was purchased from Tarson Products Private Limited, India. Ethanol (99% v/v) was procured from Changsu Hongsheng Fine Chemical Co. Ltd. (China).

Isolation and cultivation of microorganisms

For isolation of microflora, aliquots from the collected swabs were plated on de Man, Rogosa and Sharpe (MRS), Tryptone Soya (TSA) or Brain Heart Infusion (BHI) media. Based on the morphology and biochemical tests, Lactobacillus and non-Lactobacillus colonies were separated and purified. All the Lactobacillus strains grown on MRS media, were supplemented with 0.05% L-cysteine at 37°C for 48 h in anaerobic jars, supplemented with anaeroGasPak (Himedia, Mumbai). Non-Lactobacillus strains were grown either on TSA or BHI media and incubated at 37°C for 24 h. The pure colonies were stored in 20% glycerol at -80oC until further use.

Hemolysis assay

All the isolates were determined for hemolytic activity by streaking the strains on sheep blood agar plates (HiMedia, Mumbai) and incubated at 37°C for 24-48 h. The hemolytic activity was detected by observing either a clear zone of hydrolysis around the colonies (beta hemolysis), or a greenish zone (alpha hemolysis), or no zone (gamma hemolysis).

Acid and bile tolerance

The tolerance assay was done as reported earlier (Azat et al. 2016; Ehrmann 2002). Briefly, the isolates were grown, harvested and the pellet was washed and resuspended in 1X PBS (pH 3) with a final concentration of 8-9 logCFU/ml or PBS (pH 7.4) containing 0.3 % bile, followed by incubation at 37°C for 3 h. Appropriate dilutions from each sample were spread plated on the respective agar plates and incubated anaerobically using GasPack system at 37°C for 24-48 h. The tolerance was evaluated by total viable count method (log CFU/ml) after 3 h of incubation. The survival rate (%) was calculated as per the following equation:

Survival rate (%) = ,

where, N0 and N1 are the total viable counts of the selected strains before and after treatment, respectively.

MALDI-TOF MS analysis

The identification of isolates by MALDI-TOF MS was carried out at National Centre for Microbial Resources (NCMR), National Centre for Cell Sciences (NCCS), Pune. Sample preparation for MALDI-TOF MS analysis was performed as described earlier (Kurli et al. 2018). The samples were loaded onto the MALDI-TOF MS instrument (AUTOFLEX speed, Bruker Daltonics, GmbH, Germany) and MALDI biotyper software 3.1 (Bruker Daltonik GmbH, Germany) was used to identify the isolates and visualize the mass spectra. The strain showing ≥1.7 log value with the strain in the database were confirmed as the member of that genus and strains showing ≥2.0 log values were confirmed to be the member of that species.

Preparation of cell-free supernatant (CFS)

The LAB strains were grown for 24-48 h and centrifuged at 3000 rpm for 15 min. The CFS was collected and filter-sterilized with 0.22 µm syringe filter. The resultant CFS was used for antimicrobial assays and for LCMS, lyophilized powder was stored until further use.

Antibacterial activity

The antibacterial activity of CFS of the selected isolates was determined by agar well diffusion method (Reuben et al. 2019). Briefly, cultures of overnight grown indicator pathogens were adjusted to OD600nm of 0.5 McFarland. 100 µL adjusted pathogen culture was aseptically spread on the nutrient agar plates. Wells of 8 mm diameter were cut out with sterile cork borer and 150 µl CFS was loaded. The plates were kept at 4ºC for diffusion and incubated at 37°C for 24 h. After incubation, the diameter (mm) of the zone of inhibition (ZOI) around the well was measured. Pseudomonas aeruginosa MCC 2081 was used as the standard clinical pathogen. The pathogens (with beta and alpha hemolysis) used in this study were isolated from cervical cancer patients and identified by MALDI TOF MS method. Antibacterial activity of the eight standard antibiotics [carbencillin (100𝜇g), cefoxitin (30𝜇g), clindamycin (2 𝜇g), chloramphenicol (30 𝜇g), erythromycin (15𝜇g), metronidazole (5𝜇g), penicillin G (10 unit) and tetracycline (30 𝜇g)] was also evaluated against pathogens by disc diffusion method (Bayer et al. 1966).

Liquid chromatography mass spectroscopy (LCMS)

LCMS analysis of the lyophilized CFS of the selected probiotics was carried out at Center for Applications of Mass Spectrometry (CAMS), Venture Centre, Pune. Samples were analyzed by non‑targeted LCMS QTOF using Agilent 1290 HPLC system as described previously (Aphale et al. 2018). Briefly, 8 µl of sample was injected onto an Agilent 1290 HPLC system having Zorbax Eclipse Plus C18 column (2.1 mm × 50 mm, 1.8 µm particle sizes). The mobile phases consisted of (A) water and (B) acetonitrile (LCMS grade, J. T. Baker) with flow rate of 0.3 ml/min and 95:5 acetonitrile/water. Both mobile phases were modified with 0.1% (v/v) formic acid for MS analysis in positive mode and with 5 mm ammonium acetate for analysis in negative mode. The chromatographic conditions included first 18 min run of B from 95% to 5% gradient, applied from 18 to 30 min, followed by 3 min isocratically at 100%. MS analysis was performed on an Agilent 6530 Quadrupole time‑of‑flight spectrometer fitted with an electrospray ionization source in both positive and negative mode. Data were analysed by using Mass Hunter Qualitative Analysis Software Package (Agilent Technologies) and online database Metlin. Compound lists were screened against online mass databases; METLIN Metabolomics Database and MassBank Database.

16S rRNA gene sequencing and phylogenetic analysis

The identification of isolates was carried out at the sequencing facility of NCMR, Pune. The genomic DNA was isolated by the standard phenol/chloroform extraction method (Sambrook et al.1989), followed by PCR amplification of the 16S rRNA gene using universal primers 16F27 [5'-CCA GAG TTT GAT CMT GGC TCA G-3'] and 16R1492 [5'-TAC GGY TAC CTT GTT ACG ACT T-3']. The amplified 16S rRNA PCR product was purified by PEG-NaCl precipitation and directly sequenced on ABI® 3730XL automated DNA sequencer (Applied Biosystems, Inc., Foster City, CA) as per the manufacturer’s instructions. Assembly was carried out using Lasergene package followed by identification using the EzBioCloud database (Yoon et al. 2017). The phylogenic trees were constructed using the neighbor-joining method (Tamura et al. 2013) with MEGA6 software.

Tolerance to simulated gastrointestinal conditions

The ability of Lactobacilli to tolerate the simulated gastric conditions was determined as described previously (Pino et al. 2019; Pithva et al. 2014). Briefly, simulated gastric juice (SGJ) consisting of 0.3% pepsin in NaCl solution was adjusted to pH 2.5 by adding 1 M HCl. On the other hand, simulated intestinal fluid (SIF) comprising of 0.1% (w/v) pancreatin and 0.3% (w/v) bile in NaCl solution was adjusted to pH 8 by adding 1 M NaOH. Both the solutions were prepared immediately before use and sterilized using 0.22 µm filter. Probiotics were grown for 48 h, centrifuged for 3000 × g for 15 min. The pellet was washed with saline, resuspended in gastric fluid and incubated at 37°C for 3 h. After incubation, the cells were pelleted down by centrifugation, washed with 1X PBS and resuspended in SIF, followed by incubation at 37 °C for 3 h. The cells were washed with 1X PBS, serially diluted and plated on MRS agar for determining viability by total viable count method. The survival rate (%) was calculated as described for the acid and bile tolerance test.

Hydrophobicity and auto-aggregation properties

Hydrophobicity (H) (Kang et al. 2018) and auto‐aggregation (A) (Juárez Tomás et al. 2005) of the selected probiotic strains were determined as described earlier. Overnight grown bacterial suspensions in MRS broth were harvested by centrifugation at 3000 rpm for 15 min at room temperature. The cells were washed twice with 1X PBS and adjusted to an optical density (OD) of 0.5 ± 0.1 (A0) at 600 nm. To determine the hydrophobicity of the cell surface, xylene was used as a solvent. It was added to each bacterial suspension in the ratio of 1:1 and the mixtures were vortexed for 1 min and incubated for 5 h at 37°C. After phase stabilization and separation, the aqueous phase was removed, followed by measurement of absorbance (At) at 600 nm. Hydrophobicity percentage was calculated from the formula, H(%) =A0 − At/A0×100, where, A0 and At are the optical densities before and after extraction with xylene, respectively. For the auto-aggregation assay, each bacterial suspension (initial OD600nm = 0.5 ± 0.1) was vortexed for 10 s and incubated at 37°C for 5 h without agitation. The absorbance (At) was measured at 600 nm in microplate reader (Epoch, BioTek Instruments, Inc, USA). The percentage of auto-aggregation was calculated from the formula A(%) =A0 − At/A0×100, where, A0 is the OD at initial time (0 h) and At is the OD at final time (5 h) of the assay.

Biofilm formation

For biofilm formation (Terraf et al. 2012), the selected probiotic strains were inoculated in MRS media at 37°C for 24 h. Around 200 μl of the grown bacterial culture (OD 0.5) was added into the each well of 96-well plate and incubated at 37 °C for 72 h. The biofilm formed in the wells was washed twice with 200 μl 1X PBS and dried for 30 min at 37°C, followed by incubation with 200 μl of 0.1% (w/v) crystal violet for 30 min at room temperature (RT). The well was washed twice with 200 μl distilled water, dried for 10 min at RT. The residual crystal violet was dissolved in 200 μl solution containing 95% ethanol and 0.1% acetic acid in water, followed by measurement of the absorbance at 570 nm in multiplate reader.

Antibiotic susceptibility

Susceptibility of the selected probiotics to antibiotics was determined by the agar diffusion method by using two different antibiotics disks (Combi II and Combi III, Himedia, Mumbai). A total of 16 antibiotics were tested that included carbencillin (100𝜇g), cefoxitin (30𝜇g), clindamycin (2 𝜇g), chloramphenicol (30 𝜇g), erythromycin (15𝜇g), metronidazole (5𝜇g), penicillin G (10 unit), tetracycline (30 𝜇g), ampicillin (10 𝜇g), norfloxacin (10 𝜇g), nitrofurantoin (300 𝜇g), nalidixic acid (30𝜇g), gentamicin (10 𝜇g), cotrimoxazole (25𝜇g), cefalotin (30 𝜇g), and cefotaxime (30𝜇g).

Statistical analysis

The data has been presented as mean ± standard deviation (SD) and was analyzed for statistical significance by one-way and two-way analysis of variance (ANOVA) by using Tukey's multiple comparisons test. Statistical significance level was defined at p value < 0.05. All the statistical analyses were performed by using Graph Pad Prism version 9 (GraphPad, San Diego, USA).

Accession number

The nucleotide sequences of 16S rRNA of the four strains were deposited at the GenBank database under the following accession numbers: Lactobacillus delbrueckii P31Mcs (OM049479), Lactobacillus gasseri P36Mops (OM049480), Limosilactobacillus fermentum P37Mws (OM049481) and Enterococcus faecium P15Mcm (OM049482).

Results

Isolation and characterization of vaginal microflora

From the vaginal swabs of 45 healthy women, 111 microflora were isolated and characterized for their microscopic structure, Gram staining and catalase activity (Supplementary Table S1). From 40 healthy vaginal swabs, most of the isolates showed Lactobacillus related morphology (rods, Gram positive and catalase negative) (88.89%). Other 5 vaginal swabs showed presence of cocci and non-Lactobacilli. All the isolates were further evaluated for hemolysis. 79 isolates revealed non-hemolytic nature (Supplementary Table S1), out of which four were omitted that showed morphological features similar to yeast and Gram negative cocci. From the vaginal swabs of LSIL (1), HSIL (1) and ICC (6) patients (Supplementary Table S2), 29 microflora were isolated. Among these, 17.24% isolates showed Lactobacillus-related morphology, rest were cocci with either catalase negative or positive activity, and few were Gram-negative rods. Out of 29 isolates, 13 exhibited beta hemolysis and 5 showed alpha hemolysis (Supplementary Table S2) while the remaining 11 showed gamma hemolysis.

Non-hemolytic microflora exhibited acid and bile tolerance 

The non-hemolytic strains (75) from healthy controls were evaluated for acid bile tolerance. Around 90.66% strains showed tolerance to acidic pH (Table 1). After incubation with bile (0.3%), the viability of 40 isolates was reduced by approximately more than 2-log with survival rates less than 75% (Table 1). Another 22 isolates were totally inhibited in the presence of bile. Interestingly, only thirteen isolates showed tolerance to both acid and bile with less than 2-log reductions and were taken further for antibacterial studies.

MALDI TOF MS identification of the isolates 

Table 1

Acid and bile tolerance activity of non-hemolytic isolates

Sr. no.

Isolates

Acid (pH 3.0)

Bile (0.3%, w/v)

Log CFU/ml

(0 h)

Log CFU/ml

(3 h)

Survival rate

(%)

Log CFU/ml

(0 h)

Log CFU/ml

(3 h)

Survival rate

(%)

1.

P1Mcs

9.22 ± 0.05

9.03 ± 0.05

97.94

9.18 ± 0.03

8.44 ± 0.09

91.94

2.

P1Bws

8.97 ± 0.04

7.05 ± 0.12

78.59

8.91 ± 0.04

5.28 ± 0.31

59.26

3.

P2Mct

8.91 ± 0.05

0

0

8.90 ± 0.03

0

0

4.

P2Mcs

8.90 ± 0.03

8.24 ± 0.11

92.58

8.89 ± 0.02

6.64 ± 0.05

74.69

5.

P3Bws

8.93 ± 0.03

7.44 ± 0.04

83.31

8.91 ± 0.03

0

0

6.

P3Bwm

9.20 ± 0.04

8.18 ± 0.13

88.91

9.15 ± 0.07

5.18 ± 0.21

56.61

7.

P3Bwb

8.98 ± 0.04

8.63 ± 0.06

96.10

9.01 ± 0.03

0

0

8.

P5Mcs

8.83 ± 0.04

8.47 ± 0.09

95.92

8.81 ± 0.07

6.23 ± 0.07

70.71

9.

P5Bws

8.73 ± 0.07

7.13 ± 0.21

81.67

8.75 ± 0.04

5.16 ± 0.25

58.97

10.

P6MS

8.69 ± 0.05

8.31 ± 0.09

95.27

8.71 ± 0.05

5.8 ± 0.22

66.59

11.

P6Bops

8.82 ± 0.05

8.25 ± 0.14

93.51

8.83 ± 0.03

0

0

12.

P7Bop

8.63 ± 0.06

7.38 ± 0.33

85.51

8.70 ± 0.05

5.00 ± 0.00

57.47

13.

P7Mcs

9.27 ± 0.04

9.26 ± 0.03

99.89

9.34 ± 0.02

5.18 ± 0.21

55.46

14.

P8Mcs

9.24 ± 0.03

9.06 ± 0.05

98.05

9.25 ± 0.01

5.13 ± 0.21

55.45

15.

P9Mcs

8.57 ± 0.06

0.00

0

8.59 ± 0.09

0.00

0

16.

P10Tct

9.21 ± 0.02

9.15 ± 0.02

99.34

9.14 ± 0.04

5.00

54.70

17.

P10Mcb

9.06 ± 0.04

8.71 ± 0.05

96.13

9.07 ± 0.02

5.10 ± 0.16

56.22

18.

P11Mcb

9.00 ± 0.03

8.91 ± 0.04

99.00

8.97 ± 0.02

0 ± 0.00

0

19.

P11Bws

8.89 ± 0.03

8.40 ± 0.09

94.48

8.86 ± 0.04

5.25 ± 0.43

59.25

20.

P12Bops

8.78 ± 0.04

8.06 ± 0.08

91.79

8.80 ± 0.05

5.27 ± 0.26

59.88

21.

P13Bops

8.87 ± 0.05

0

0

8.89 ± 0.04

0

0

22.

P13Mcs

8.17 ± 0.18

8.00 ± 0.14

97.91

8.39 ± 0.08

7.05 ± 0.12

84.02

23.

P14Mcs

8.61 ± 0.06

8.17 ± 0.11

94.88

8.64 ± 0.05

7.05 ± 0.12

81.59

24.

P15Mcm

9.20 ± 0.02

8.98 ± 0.04

97.60

9.29 ± 0.02

8.75 ± 0.04

94.18

25.

P15Mcs

9.10 ± 0.03

8.67 ± 0.06

95.27

9.04 ± 0.03

6.48 ± 0.09

71.68

26.

P16Mws

8.93 ± 0.06

8.61 ± 0.07

96.41

8.92 ± 0.05

7.88 ± 0.10

88.34

27.

P17Mws

9.16 ± 0.03

0

0

9.12 ± 0.03

0

0

28.

P18Mops

8.99 ± 0.08

0

0

8.90 ± 0.07

0

0

29.

P18Topb

8.75 ± 0.08

0

0

8.70 ± 0.06

0

0

30.

P18Mct

9.04 ± 0.03

8.76 ± 0.04

93.19

9.06 ± 0.03

0

0

31.

P18Mcs

9.03 ± 0.03

8.77 ± 0.04

97.12

9.01 ± 0.03

0

0

32.

P19Mws

8.90 ± 0.11

7.89 ± 0.12

88.65

8.88 ± 0.09

0

0

33.

P20 Mop

9.01 ± 0.05

8.31 ± 0.18

92.23

8.96 ± 0.04

0

0

34.

P21Mcb

8.58 ± 0.19

7.42 ± 0.33

86.48

8.79 ± 0.05

6.5 ± 0.11

73.94

35.

P22Mws

8.50 ± 0.11

0

0

8.63 ± 0.06

0

0

36.

22Mops

9.18 ± 0.03

8.94 ± 0.04

97.38

9.15 ± 0.04

7.15 ± 0.16

78.14

37.

P23Mcs

9.76 ± 0.01

9.68 ± 0.04

99.18

9.71 ± 0.03

6.56 ± 0.15

67/56

38.

P24Mwt

9.11 ± 0.03

8.86 ± 0.05

97.25

9.06 ± 0.02

7.05 ± 0.12

77.81

39.

P25Mws

9.41 ± 0.05

8.89 ± 0.08

94.47

9.5 ± 0.06

6.56 ± 0.09

69.05

40.

P26Mcm

8.96 ± 0.05

8.59 ± 0.08

95.87

9.29 ± 0.04

8.55 ± 0.12

92.03

41.

P26Mct

9.04 ± 0.05

8.64 ± 0.09

95.57

9.01 ± 0.06

0

0

42.

P28Mws

8.92 ± 0.00

8.10 ± 0.24

90.80

9.10 ± 0.05

5.35 ± 0.12

58.79

43.

P31Mcs

9.32 ± 0.023

9.07 ± 0.05

97.37

9.33 ± 0.03

8.49 ± 0.10

90.37

44.

P33Mcm

9.19 ± 0.03

8.40 ± 0.11

91.40

9.16 ± 0.03

5.37 ± 0.31

55.93

45.

P34Mws

9.03 ± 0.03

8.91 ± 0.03

98.67

9.04 ± 0.04

5.43 ± 0.16

60.06

46.

P34Mcs

8.24 ± 0.2

7.96 ± 0.15

96.60

8.38 ± 0.11

6.23 ± 0.12

74.34

47.

P35Mws

8.88 ± 0.06

8.54 ± 0.07

96.17

8.94 ± 0.03

5.18 ± 0.21

57.94

48.

P36Mops

8.61 ± 0.08

8.32 ± 0.10

96.63

8.63 ± 0.06

7.81 ± 0.11

90.49

49.

P36Mws

9.40 ± 0.05

9.18 ± 0.05

97.65

9.36 ± 0.03

6.66 ± 0.12

71.15

50.

P37Mcb

9.22 ± 0.01

8.40 ± 0.11

91.10

9.23 ± 0.02

0

0

51.

P37Mws

9.29 ± 0.03

8.87 ± 0.06

95.47

9.26 ± 0.04

8.55 ± 0.08

92.33

52.

P38Mops

8.80 ± 0.07

8.29 ± 0.10

94.20

8.78 ± 0.05

5.10 ± 0.16

58.08

53.

P39Mops

8.79 ± 0.03

8.52 ± 0.09

96.92

8.78 ± 0.04

5.3 ± 0.27

60.36

54.

P42Mcm

8.85 ± 0.06

7 ± 0.00

79.09

8.84 ± 0.04

0

0

55.

P43Mops

8.72 ± 0.05

8.41 ± 0.09

96.44

8.64 ± 0.07

6.65 ± 0.08

76.96

56.

P44Mcm

9.18 ± 0.02

8.62 ± 0.10

98.73

9.07 ± 0.03

5.38 ± 0.03

59.31

57.

P44Mcs

9.08 ± 0.05

8.99 ± 0.09

99.00

9.07 ± 0.04

6.04 ± 0.10

66.59

58.

P44Tcs

9.02 ± 0.03

8.43 ± 0.08

93.45

9.15 ± 0.33

5.10 ± 0.16

55.73

59.

P45Bws

9.13 ± 0.03

8.09 ± 0.11

88.60

9.13 ± 0.02

5.13 ± 0.21

56.18

60.

P45Tcm

9.21 ± 0.03

9.10 ± 0.02

98.91

9.18 ± 0.03

0

0

61.

P46Mopb

8.74 ± 0.07

8.44 ± 0.09

96.56

8.65 ± 0.11

5.21 ± 0.24

60.23

62.

P46Mops

8.72 ± 0.09

8.30 ± 0.11

95.18

8.75 ± 0.10

6.27 ± 0.10

71.65

63.

P47Mops

8.91 ± 0.04

8.42 ± 0.07

94.50

8.79 ± 0.03

6.74 ± 0.05

76.67

64.

P47Bws

9.05 ± 0.03

8.48 ± 0.11

93.90

9.06 ± 0.02

5.10 ± 0.16

56.29

65.

P49Mopb

8.87 ± 0.06

8.16 ± 0.09

91.99

8.90 ± 0.05

5.23 ± 0.19

58.76

66.

P51Mcm

9.16 ± 0.02

8.16 ± 0.12

89.08

9.11 ± 0.04

5.18 ± 0.21

56.86

67.

P51Mwt

9.17 ± 0.03

9.03 ± 0.03

98.47

9.18 ± 0.02

6.23 ± 0.08

67.86

68.

P51Tops

8.97 ± 0.03

8.39 ± 0.07

93.53

8.97 ± 0.03

5 ± 0.00

55.74

69.

P52Mcm

9.20 ± 0.02

9.05 ± 0.02

98.36

9.16 ± 0.02

5.48 ± 0.20

59.82

70.

P52Tcb

9.16 ± 0.03

9.02 ± 0.03

98.47

9.16 ± 0.02

0

0

71.

P53Mcs

9.02 ± 0.02

8.47 ± 0.06

93.90

8.97 ± 0.04

5.19 ± 0.45

57.85

72.

P53Tcm

9.2 ± 0.03

8.98 ± 0.04

97.60

9.18 ± 0.03

0

0

73.

P54Mcs

9.18 ± 0.02

9.08 ± 0.03

98.91

9.16 ± 0.03

5.15 ± 0.16

56.22

74.

P54Tcb

9.17 ± 0.05

9.10 ± 0.02

99.23

9.12 ± 0.04

0

0

75.

P54Tcm

9.16 ± 0.04

9.09 ± 0.05

99.23

9.14 ± 0.04

6.78 ± 0.07

74.17

The data has been presented as mean ± SD of three independent experiments, each performed in duplicates.

All the thirteen isolates showing acid and bile tolerance were further identified by MALDI-TOF MS (Table 2). For each LAB strain, the score value was in the range of 1.7-2. Among the 13 identified LAB from healthy group, Lactobacillus gasseri (53.85%) was found to be dominant, followed by Limosilactobacillus fermentum (15.38%), Enterococcus faecium (15.38%), Lactobacillus reuteri (7.69%) and Lactobacillus delbrueckii (7.69%). Out of 18 pathogenic isolates, 8 strains were identified by MALDI TOF MS (score value above 2.0) (Table 3), S. haemolyticus P41Tcs and E. coli P79Bcm were isolated from the swabs of LSIL and HSIL patients, respectively. E. coli P30Tcs, E. faecalis P29Mops, E. faecalis P50Tws, E. faecalis P68Tcb, S. haemolyticus P48Bcb and S. haemolyticus P58Bcb were isolated from the swabs of 6 cervical cancer patients. Among these, S. haemolyticus (37.5%) and E. faecalis (37.5%) were found to be the dominant strains, followed by E. coli(25%). The identified pathogenic strains were used as indicators for further antimicrobial studies. 

Table 2

Identification of thirteen isolates from healthy individuals by MALDI-TOF MS

Sr. no.

Isolate from healthy individuals

Organism(best match)

Score

1.

P1Mcs

Limosilactobacillus fermentum DSM 20055

1.745

2.

P13Mws

Lactobacillus gasseri DSM 20243T

1.843

3.

P14Mcs

Lactobacillus gasseri DSM 20243T

2.314

4.

P15Mcm

Enterococcus faecium DSM 2146

2.302

5.

P16Mws

Lactobacillus gasseri DSM 20604

1.771

6.

P22Mops

Lactobacillus gasseri DSM 20243T

2.439

7.

P24Mwt

Lactobacillus reuteri DSM 20053

1.729

8.

P26Mcm

Enterococcus faecium 20218_1

1.989

9.

P31Mcs

Lactobacillus delbrueckii DSM 20073

1.888

10.

P36Mops

Lactobacillus gasseri DSM 20243T

1.856

11.

P37Mws

Lactobacillus fermentum DSM 20055

2.020

12.

P43Mops

Lactobacillus gasseri DSM 20243T

2.442

13.

P47Mops

Lactobacillus gasseri CIP 101909

2.304

Table 3

MALDI-TOF MS Identification of pathogenic isolates from precancerous (LSIL and HSIL) and ICC patients

Sr. No.

Isolate

Organism

Score

Patient group

1.

P41Tcs

Staphylococcus haemolyticus

2.02

LSIL

2.

P79Bcm

Escherichia coli

2.08

HSIL

3.

P30Tcs

E. coli

2.20

ICC

4.

P29Mops

Enterococcus faecalis

2.43

ICC

5.

P50Tws

E. faecalis

2.03

ICC

6.

P68Tcb

E. faecalis

2.12

ICC

7.

P48Bcb

S. haemolyticus

2.10

ICC

8.

P58Bcb

S. haemolyticus

2.05

ICC

CFS of selected strains from healthy group exhibited antimicrobial activity 

The cell free supernatant (CFS) of the acid-bile tolerant isolates (13) from healthy individuals were evaluated for antimicrobial activity against standard (commercially obtained) pathogen, P. aeruginosa 2081 and eight isolated pathogenic strains from cervical cancer patients (Table 4). Among the thirteen isolates, L. fermentum P37Mws exhibited significantly high (p < 0.05) antimicrobial activity against four pathogenic strains, P. aeruginosa, E. coli P79Bcm, S. haemolyticus P48Bcb and P58Bcb. L. gasseri P36Mops also showed antibacterial activity (p < 0.05) against four pathogens, E. coli P30Tcs and P79Bcm; S. haemolyticus P41Tcs and P58Bcb. L. delbrueckii P31Mcs showed significant (p < 0.05) zone of inhibition against (three pathogens) E. coli P30Tcs, S. haemolyticus P48Bcb and 58Bcb. Interestingly, E. faecium P15Mcm showed significantly higher zone of inhibition (> 18 mm) against three pathogenic strains of E. faecalis P29Mops, P50Tws and P68Tcb. Thus, overall data showed that four strains (L. fermentum P37Mws, L. delbrueckii P31Mcs, L. gasseri P36Mops and E. faecium P15Mcm) exhibited probiotic characteristics and antibacterial activity against more than two pathogens. The vaginal pathogens were also tested for their susceptibility to eight standard antibiotics that included carbencillin, cefoxitin, clindamycin, chloramphenicol, erythromycin, metronidazole, penicillin G and tetracycline. Tetracycline exhibited antibacterial activity against all the eight vaginal pathogens whereas metronidazole did not show any activity against the pathogens (Supplementary Table S3). On the other hand, carbencillin displayed antibacterial activity only against the strains of E. faecalis.Rest of the antibiotics displayed varied spectrum of antibacterial activity against the pathogens.

Table 4

Antimicrobial activity of selected isolates from healthy group against pathogens

Sr. no.

Isolates (CFSs)

Zone of inhibition (mm) against different pathogens

P. aeruginosa

2081

E. coliP30Tcs

E. coliP79Bcm

E. faecalis

P29Mops

E. faecalis

P50Tws

E. faecalis

P68Tcb

S. haemolyticus

P41Tcs

S. haemolyticus

P58Bcb

S. haemolyticus

P48Bcb

1.

L. fermentum P1Mcs

13.56 ± 0.73e

14.56 ± 0.53a

12.56 ± 0.53e

-

-

-

-

15.33 ± 0.50a

14.89 ± 0.60b

2.

L. gasseri P13Mcs

11.56 ± 0.53e

12.44 ± 0.73e

-

-

-

-

-

12.89 ± 0.60e

13.56 ± 0.53e

3.

L. gasseri P14Mcs

14.00 ± 0.71e

13.44 ± 0.53e

-

-

-

-

-

14.44 ± 0.88c

12.78 ± 0.44e

4.

E. faecium P15Mcm

13.78 ± 0.44e

-

-

21.00 ± 1.32a

18.79 ± 0.97a

19.67 ± 0.50a

-

-

-

5.

L. gasseri P16Mws

15.33 ± 0.5e

14.78 ± 0.44a

-

-

-

-

-

15.00 ± 0.71a

14.44 ± 0.53e

6.

L. gasseri P22Mops

16.11 ± 0.60a

-

-

-

-

-

-

11.33 ± 0.50e

13.78 ± 0.44e

7.

L. reuteri P24Mwt

13.11 ± 0.93e

13.67 ± 0.71e

-

-

-

-

-

11.67 ± 0.50e

11.89 ± 0.33e

4

8.

E. faecium P26Mcm

12.56 ± 0.53e

-

-

14.44 ± 0.53e

14.11 ± 0.60e

14.00 ± 0.50e

-

-

-

9.

L. delbrueckii P31Mcs

15.67 ± 0.71a

15.11 ± 0.60a

12.89 ± 0.33e

11.67 ± 0.50e

-

-

13.44 ± 0.53a

15.11 ± 0.60a

14.89 ± 0.60b

10.

L. gasseri P36Mops

15.11 ± 0.60d

14.44 ± 0.73a

12.00 ± 0.50e

12.67 ± 0.50e

-

-

11.22 ± 0.97e

15.00 ± 0.71a

15.67 ± 0.50a

11.

L. fermentum P37Mws

15.56 ± 0.53a

13.78 ± 0.44e

14.00 ± 0.5a

14.33 ± 0.71e

14.89 ± 0.33e

-

11.89 ± 0.33e

15.33 ± 0.50a

15.22 ± 0.44a

12.

L. gasseri P43Mops

14.00 ± 0.87e

13.33 ± 0.71e

-

-

-

-

-

13.78 ± 0.44e

14.11 ± 0.6e

13.

L. gasseri P47Mops

13.67 ± 0.71e

11.67 ± 0.50e

-

-

-

-

-

14.44 ± 0.53c

14.33 ± 0.50e

“-‘ no activity observed. The data has been presented as mean ± SD of three independent experiments, each performed in triplicates. Statistically different results within the column were labelled with various lowercase letter (a:p > 0.05; b:p < 0.05; c:p < 0.005; d:p < 0.001; e:p < 0.0001).
The inhibition zones (ZOI) were classified as: no activity (< 9 mm); weak (< 14 mm); good (15–19 mm) and strong (> 20 mm) (Reuben et al 2019).

CFS showed presence of metabolites with reported antimicrobial activity

LCMS analysis (non-targeted) of the CFS from the selected probiotics was carried out to identify the potential antibacterial compounds secreted by the respective strains. Around 119 compounds from L. gasseri, 95 from L. fermentum, 106 from L. delbrueckii P31Mcs and 117 from E. faecium P15Mcm were identified. The major metabolites with antibacterial activity have been shown in Table 5 and their respective chromatograms have been included in Supplementary Fig. S1. The antibacterial metabolites from CFSs of L. gasseri P36Mops included N-(1-deoxy-1-fructosyl)valine, homoarecoline, 2-Isopropyl-1,4-benzenediol, 1-Acetoxy-2-hydroxy-16-heptadecen-4-one, avocadyne 4-acetate, avocadyne 2-acetate, grandidentatin, taraxinic acid glucosyl ester and nigellicine. The metabolites detected from L. fermentum P37Mws included methylarmepavine, (-)-hygroline, avocadyne 4-acetate, avocadyne 2-acetate and 1-acetoxy-2-hydroxy-16-heptadecen-4-one. The CFS of L. delbrueckii P31Mcs showed the presence of metabolites such as (+)-O-methylarmepavine, 6-methylquinoline, quinaldine, 1-acetoxy-2-hydroxy-16-heptadecen-4-one, 3-O-sulfogalactosylceramide. The metabolites such as nepetalactam, hordenine, cuminaldehyde, 4-phenyl-3-buten-2-ol, 6-hydroxypentadecanedioic acid, hygroline, taraxinic acid glucosyl ester, avermectin A2b aglycone were detected from E. faeciumP15Mcm. Thus, the CFS of the probiotic strains showed presence of antimicrobial metabolites, which could be responsible for their observed antibacterial activity.

Table 5

Antibacterial metabolites present in the CFSs of selected probiotics identified by LCMS

Name of identified compounds

Empirical formula

Observed RT

M/Z ratio

Observed mass

Score

L. gasseri P36Mops

N-(1-Deoxy-1-fructosyl)valine

C11 H21 N O7

0.805

280.1383

279.131

96.74

Homoarecoline

C9 H15 N O2

1.158

170.1172

169.1099

98.96

2-Isopropyl-1,4-benzenediol

C9 H12 O2

1.158

170.1172

152.0834

98.92

(-)-Hygroline

C8 H17 N O

3.191

166.1207

143.1316

93.34

1-Acetoxy-2-hydroxy-16- heptadecen-4-one

C19 H34 O4

6.573

344.2793

326.2451

91.02

Avocadyne 4-acetate

C19 H34 O4

6.573

344.2793

326.2451

91.02

Avocadyne 2-acetate

C19 H34 O4

6.573

344.2793

326.2451

91.02

Grandidentatin

C21 H28 O9

6.584

442.2053

424.1717

92.5

Taraxinic acid glucosyl ester

C21 H28 O9

6.598

442.2056

424.172

93.3

Nigellicine

C13 H15 N2 O3

8.962

930.6324

247.108

94.77

L. fermentum P37Mws

Methylarmepavine

C20 H25 N O3

2.12

328.1897

327.1826

91.85

6-Hydroxypentadecanedioic acid

C15 H28 O5

2.906

289.2015

288.1943

93.50

(-)-Hygroline

C8 H17 N O

4.081

166.1208

143.1317

93.65

Taraxinic acid glucosyl ester

C21 H28 O9

6.803

442.2056

424.1717

92.45

1-Acetoxy-2-hydroxy-16-

heptadecen-4-one

C19 H34 O4

6.814

344.2794

326.2452

93.81

Avocadyne 4-acetate

C19 H34 O4

6.814

344.2794

326.2452

93.81

Avocadyne 2-acetate

C19 H34 O4

6.814

344.2794

326.2452

93.81

L. delbrueckii P31Mcs

(+)-O-Methylarmepavine

C20 H25 N O3

2.075

328.1898

327.1826

94.22

6-Methylquinoline

C10 H9 N

2.979

144.0811

143.0737

93.17

Quinaldine

C10 H9 N

2.979

144.0811

143.0737

93.17

1-Acetoxy-2-hydroxy-16-

heptadecen-4-one

C19 H34 O4

6.692

344.2796

326.245

95.36

Avocadyne 4-acetate

C19 H34 O4

6.692

344.2796

326.245

95.36

Avocadyne 2-acetate

C19 H34 O4

6.692

344.2796

326.245

95.36

Taraxinic acid glucosyl ester

C21 H28 O9

6.694

442.2056

424.172

92.95

E. faecium P15Mcm

Ephedrine

C10 H15 N O

1.841

166.1219

165.1148

93.25

Nepetalactam

C10 H15 N O

1.841

166.1219

165.1148

93.25

Hordenine

C10 H15 N O

1.841

166.1219

165.1148

93.25

Cuminaldehyde

C10 H12 O

1.841

166.1219

148.0882

93.12

Anethole

C10 H12 O

1.841

166.1219

148.0882

93.12

Estragole

C10 H12 O

1.841

166.1219

148.0882

93.12

6-Hydroxypentadecanedioic acid

C15 H28 O5

2.201

289.2012

288.1941

96.85

Hygroline

C8 H17 N O

3.089

166.1207

143.1316

94.48

Taraxinic acid glucosyl ester

C21 H28 O9

6.598

442.2056

424.172

93.3

Avermectin A2b aglycone

C34 H50 O9

7.009

603.3528

602.3459

93.29

Avermectin B2a aglycone

C34 H50 O9

7.009

603.3528

602.3459

93.29

RT: Retention time

16s rRNA sequencing and phylogenetic analysis 

The selected probiotic strains were subjected to molecular identification and phylogenetic analysis by the Sanger sequencing method and EzBioCloud database, respectively (Table 6). A comparative 16S rRNA gene based phylogenetic analysis of the four vaginal strains, P36Mops, P37Mws, P31Mcs and P15Mcm revealed their closest similarity (ranging from 100 to 99%) to the sequences of the type strains, L. gasseri ATCC 33323 (Azcarate-Peril et al. 2008), L. fermentum CECT562 (Zheng et al. 2020), L. delbrueckii DSM 20072 (Schoch et al. 2020) and E. lactis BT159 (Morandi et al. 2012), respectively, from GeneBank database (Supplementary Fig. S2-S5).

Table 6

Molecular identification of selected vaginal probiotics by 16S rRNA sequencing

Sr. No

Strain

Closest Neighbour %

% Similarity

Taxonomic Designation

Accession no.

1

P36Mops

Lactobacillus gasseri ATCC 33323( T)

CP000413

99.86

2

P37Mws

Limosilactobacillus fermentum CECT 562(T)

AJ575812

99.85

3

P31Mcs

Lactobacillus delbrueckii subsp. lactis DSM 20072(T)

AEXU01000148

99.92

4

P15Mcm

Enterococcus faecium LMG 11423(T)

AJ301830

99.78

Probiotics survived the simulated gastric juice (SGJ) and intestinal fluid (SIF) conditions

All the selected strains displayed > 80% survival in 0.3% pepsin (Table 7), representing SGJ. L. fermentum P37Mws exhibited significant (p < 0.0001) survival (96.3%) with viable count of 8.86 ± 0.09 log CFU/ml, followed by E. faecium P15Mcm (94.16%). On the other hand, L. gasseri P36Mops and L. delbrueckii P31Mcs showed survival upto 85.94 to 84.86%, respectively. All the four probiotics showed 60.09–76.30% survival upto 3 h in the presence of pancreatin enzyme. The viable count for L. fermentum P37Mws and L. gasseri P36Mops did not differ significantly (p > 0.05) under SIF environment. The viable count was decreased to approximately 6 logs CFU/ml from the initial count (approx. 8–9 logs CFU/ml) in all, except in L. delbrueckiiP31Mcs that showed viable count of 5.24 log CFU/ml. Thus, the probiotic strains survived the gastrointestinal (GI) transit, which is a prerequisite for colonization to the host epithelial cells for providing health benefits. 

Table 7

Effect of simulated human gastric (SGJ) and intestinal fluid (SIF) on the survival of selected vaginal probiotics

Probiotic strains

(log CFU/ml)

0 h

SGJ

(log CFU/ml)

3 h

SR (%)

SIF

(log CFU/ml)

3 h

SR (%)

L. gasseri P36Mops

8.89 ± 0.04A

7.64 ± 0.19A

85.94

6.15 ± 0.12A

69.18

L. fermentum P37Mws

9.20 ± 0.02AcB

8.86 ± 0.09AcB

96.30

6.18 ± 0.18AaB

67.17

L. delbrueckii P31Mcs

8.72 ± 0.10AaBcC

7.40 ± 0.30AbBcC

84.86

5.24 ± 0.19AcBcC

60.09

E. faecium P15Mcm

9.07 ± 0.04AaBaCc

8.54 ± 0.11AcBcCc

94.16

6.92 ± 0.09AcBcCc

76.30

The data has been presented as mean ± SD of three independent experiments, each performed in triplicates. Statistically (Tukey's multiple comparisons test) different results within the column were labelled with a various lowercase letter (a:p > 0.05; b:p < 0.005; c:p < 0.0001)

Probiotics exhibited properties of hydrophobicity, auto-aggregation and biofilm formation

All the selected probiotics, after 5 h of incubation with the xylene, showed moderate hydrophobicity (35.88–56.70%). L. gasseri P36Mops showed highest (56.70%±1.63) hydrophobicity (Table 8), followed by L. delbrueckii P31Mcs (55.92%±2.41), E. faecium P15Mcm (39.69%±2.83) and L. fermentum P37Mws (35.88%±1.97). There was no significant difference (P > 0.05) in the hydrophobicity between L. gasseri P36Mops and L. delbrueckii P31Mcs. The tested probiotics showed highest auto-aggregation with E. faecium P15Mcm (61.39%±1.49; p < 0.0001), followed by L. fermentum P37Mws (52.20%±0.93; p < 0.0001), L. delbrueckii P31Mcs (43.12%±2.27; p < 0.0001) and L. gasseri P36Mops (35.26%±2.03; p < 0.0001) (Table 8). All the four probiotic strains showed higher optical density (ODT) at 570 nm that was 4 times greater (4ODc < ODT; p < 0.0001), than the OD of control media (ODc = 0.321 ± 0.04). Based on biofilm production, the strains were divided into non- (ODT ≤ ODc); weak (ODc < ODT ≤ 4ODc); and strong (4ODc < ODT) producers, where ODc is OD of control media (Terraf et al. 2012). It was noted that L. fermentum P37Mws produced more biofilm, (3.25 ± 0.15; p < 0.0001), which was followed by E. faecium P15Mcm (2.57 ± 0.43; p < 0.0001), L. delbrueckii P31Mcs (2.40 ± 0.41; p < 0.0001) and L. gasseri P36Mops (1.41 ± 0.35; p < 0.0001) (Fig. 1). Thus, the selected strains exhibited probiotic properties of hydrophobicity, auto aggregation and biofilm production.

Table 8

Hydrophobicity and auto-aggregation properties of selected probiotics

Probiotic strains

Hydrophobicity

(%)

Auto-aggregation

(%)

L. gasseri P36Mops

56.70 ± 1.63A

35.26 ± 2.03A

L. fermentum P37Mws

35.88 ± 1.97AcB

52.20 ± 0.93AcB

L delbrueckii P31Mcs

55.92 ± 2.41AaBcC

43.12 ± 2.27AcBcC

E. faecium P15Mcm

39.69 ± 2.83AcBbCc

61.39 ± 1.49AcBcCc

The data has been presented as mean ± SD of three independent experiments, each performed in triplicates. Statistically (Tukey's multiple comparisons test) different results within the column were labelled with a various lowercase letter (a:p > 0.05; b:p < 0.05; c:p < 0.0001)

Probiotics exhibited resistance towards standard antibiotics 

The selected probiotics were evaluated for their resistance towards 16 different antibiotics (carbencillin, cefoxitin, clindamycin, chloramphenicol, erythromycin, metronidazole, penicillin G, tetracycline, ampicillin, norfloxacin, nitrofurantoin, nalidixic acid, gentamicin, cotrimoxazole, cefalotin and cefotaxime), mostly prescribed for bacterial vaginosis or urogenital infections. L. gasseri exhibited resistance towards five antibiotics that included nalidixic acid, norfloxacin, nitrofurantoin, metronidazole and cotrimoxazole (Table 9). L. fermentum P37Mws showed resistance to four antibiotics namely, cefoxitin, metronidazole, nalidixic acid and cotrimoxazole. E. faecium P15Mcm exhibited resistance to four antibiotics (carbencillin, metronidazole, nalidixic acid and cotrimoxazole) and L. delbrueckii showed resistance towards two antibiotics (nalidixic acid and cotrimoxazole).

Table 9

Antibiotics susceptibility of selected probiotics

Antibiotics

Antibiotic susceptibility

L. gasseri

P36Mops

L. fermentum P37Mws

L. delbrueckii P31Mcs

E. faecium P15Mcm

Carbecillin

S

S

S

R

Cefoxitin

S

R

S

S

Clindamycin

S

S

S

S

Chloramphenicol

S

S

S

S

Erythromycin

S

S

S

S

Metronidazole

R

R

S

R

Penicillin G

S

S

S

S

Tetracycline

S

S

S

S

Gentamycin

S

S

S

S

Nalidixic acid

R

R

R

R

Nitrofuramion

R

S

S

S

Norfloxacin

R

S

S

S

Ampicillin

S

S

S

S

Cefotaxime

S

S

S

S

Cephathnin

S

S

S

S

Cotrimoxazole

R

R

R

R

Note: S: susceptible, R: resistance

Discussion

In the present study, microflora isolated from the vaginal swabs of healthy women exhibited probiotic properties along with antimicrobial activity against pathogenic bacteria isolated from vaginal swabs of precancerous and cervical cancer patients. Depletion of beneficial vaginal Lactobacilli with increased load of pathogenic bacteria is one of the high-risk factors for cervical cancer progression (Adebamowo et al. 2017; Mitra et al. 2020). Vaginal microflora of cervical cancer patients is predominantly colonised with different anaerobic (Ureaplasma parvum, Atopobium vaginae, Megasphaera, Prevotella, Gardnerella, Sneathia sanguinegens, Fusobacteria, Fastidiosipila and Dialister) and aerobic (S. epidermidis, E. faecalis, E. coli) pathogens (Adebamowo et al. 2017; Caselli et al. 2020; Kalia et al. 2020; Mitra et al. 2020). Such microflora produce toxins and carcinogenic metabolites (nitrosamines) that trigger oxidation of DNA and production of pro-inflammatory cytokines, which together aid in the progression of cervical cancer (Chen et al. 2019; Mitra et al. 2020).Thus, a balanced vaginal microbiome may not only prevent vulvo-vaginal infections but could also regulate cancer development (Godoy-Vitorino et al. 2018; Hatta et al. 2021).

The present data showed that out of 75 non-hemolytic isolates from healthy group, 13 strains survived gastro-intestinal transit (GIT) with more than 75% survival, which is an important criteria for development of oral probiotics. For oral delivery, probiotics should a) be non-pathogenic; b) tolerate the unfavourable conditions of the gastrointestinal tract (acidic pH and bile acid concentrations) and c) reach the intestine in a viable state (Dinçer and Kıvanç 2021; Oh et al. 2018). The tolerance of other vaginal probiotics such as L. rhamnosus, L. helveticus and L. salivarius (Pithva et al. 2014), L. gasseri and L. plantarum (Bouridane et al. 2016), L. fermentum (Brandt et al. 2020) to simulated human gastrointestinal tract conditions has been reported earlier. Acid-bile tolerance for L. gasseri, L. fermentum has been reported with above 70–80% survival (Archer and Halami 2015; Oh et al. 2018). Thus, the thirteen strains from healthy individuals exhibited probiotic properties of survival in GI tract.

Initially, MALDI-TOF MS analysis was used to identify the thirteen stains, among which L. gasseri was found to be the dominant strain, followed by L. fermentum, E. faecium, L. reuteri and L. delbrueckii. The species level identification of the LAB strains was further confirmed by 16S rRNA identification, with scores in the range 1.7–2.0. Similar observations have been reported earlier with vaginal LAB (Anderson et al. 2014). The isolates from LSIL, HSIL and ICC vaginal swabs were either cocci or Gram negative rods and only few had Lactobacilli-like morphology. Among these isolates, three strains of E. faecalis exhibited alpha hemolytic activity whereas three strains of S. haemolyticus and two strains of E. coli exhibited beta hemolytic activity, thereby confirming their pathogenic nature. E. faecalis isolated from patients with intestinal infection has been reported to exhibit alpha hemolysis (Bello Gonzalez et al. 2017). Beta hemolysis has been reported for several clinical strains of E. coli (Navidinia et al. 2012; Navidinia 2014; Toval et al. 2014) and S. haemolyticus (Pinheiro et al. 2015), isolated from patients with urinary tract and blood infections, respectively.

In the current study, the CFSs of all the thirteen strains inhibited the growth of the standard pathogen, P. aeruginosa MCC 2081. CFS of four isolates, L. fermentum P37Mws, L. gasseri P36Mops, L. delbrueckii P31Mcs and E. faecium P15Mcm exhibited good to strong antibacterial activity against vaginal pathogens isolated from vaginal swabs of LSIL, HSIL and ICC patients. The antibacterial activity of vaginal probiotic L. fermentum, isolated from healthy Algerian women was reported against the vaginal pathogens E. coli, Staphylococcus sp., Enterococcus sp., and Candida sp. (Ouarabi et al. 2019). CFS from Lactobacillus strain VLb3 has shown antibacterial activity against the vaginal pathogen G. vaginalis ATCC14018 (Andreeva et al. 2016). Bacteriocin extracted from vaginal probiotic Lactobacillus has shown activity against Salmonella, Gardnerella, Chlamydia, Trichomonas and Neisseria, isolated from patients with cervicovaginal infection (Dasari et al. 2014). The healthy vaginal microflora is mostly dominated by Lactobacillus species and opportunistic pathogens (Kyrgiou et al. 2017). However, Lactobacillus dominant communities protect the host against genital pathogenic infections through the production of antimicrobial compounds and short chain fatty acids (SCFA) that acidify the local microenvironment by keeping vaginal pH below 4.5 (Kyrgiou et al. 2017). Depletion of beneficial Lactobacillus could increase vaginal pH, thereby increasing its susceptibility to infection with diverse aerobic (Staphylococcus sp, Pseudomonas sp. E. coli, and E. faecalis) and anaerobic (Gardnerella sp., Atopobium sp., Eggerthella sp., Sneathia sp. and Prevotella sp.) bacteria. Such dysbiosis could modulate immune responses and thus lead to pathogenesis of several diseases including cervical cancer.

The CFS from L. gasseri P36Mops was found to be rich in antimicrobial compounds such as N-(1-deoxy-1-fructosyl)valine (organic acid) (Fuochi et al. 2019); alkaloids, homoarecoline (Matsumoto et al. 2012) and hygroline (Jurica et al. 2017), 2-isopropyl-1,4-benzenediol (hydroquinone) (Cretton et al. 2021); long chain fatty alcohols, 1-acetoxy-2-hydroxy-16-heptadecen-4-one, avocadyne 4-acetate and avocadyne 2-acetate (Rodríguez-Sánchez et al. 2019); grandidentatin (cinnamate ester) (Tyśkiewicz et al. 2019); taraxinic acid glucosyl ester (sesquiterpene lactone); (Cartagena et al. 2008) and alkaloid, nigellicine (Mohammed et al. 2019). L. fermentum P37Mws CFS showed presence of methylarmepavine (benzylisoquinolines) with reported anti-Leishmanial and antibacterial activities (Do Nascimento et al. 2015); and antimicrobial compounds such as 6-hydroxypentadecanedioic acid (long-chain fatty acid) (Rocchetti et al. 2020), hygroline (Jurica et al. 2017), avocadyne 4-acetate, avocadyne 2-acetate and 1-acetoxy-2-hydroxy-16-heptadecen-4-one (Rodríguez-Sánchez et al. 2019). CFS from L. delbrueckii P31Mcs showed the presence of antimicrobial metabolites such as (+)-O-methylarmepavine (Do Nascimento et al. 2015), 1-acetoxy-2-hydroxy-16-heptadecen-4-one, avocadyne 4-acetate, avocadyne 2-acetate (Rodríguez-Sánchez et al. 2019) and quinoline derivatives, 6-methylquinoline and quinaldine (Bawa et al. 2009; Jeon et al. 2009). E. faecium P15Mcm CFS had antimicrobial compounds such as alkaloids, ephedrine (Tulgar et al. 2018) and hordenine (Zhou et al. 2018), nepetalactam (tetrahydropyridine) (Aridoss et al. 2008); cuminaldehyde (benzaldehyde) (Wongkattiya et al. 2019); anethole (phenylpropanoid) (Esfandyari-Manesh et al. 2013); estragole (olefinic compound) (Song et al. 2016); 6-hydroxypentadecanedioic acid (Rocchetti et al. 2020); hygroline (Jurica et al. 2017); taraxinic acid glucosyl ester (Cartagena et al. 2008; Tyśkiewicz et al. 2019); oxane and tertiary allylic alcohol derivatives, avermectin A2b aglycone and avermectin B2a aglycone (El-Saber Batiha et al. 2020). Some antimicrobial metabolites were common in CFS of all the four probiotics. Antimicrobial activity of L. fermentum TcUESC01 having valine and benzeneacetic acid as metabolites have shown activity against Streptococcus mutans UA159 (de Souza Rodrigues et al. 2020). Different strains of L. fermentum and L. gasseri, isolated from human (oral and vaginal) samples, have shown antibacterial activity against P. aeruignosa (Fuochi 2016) and Legionella pneumophila. Hydroquinone has shown antibacterial activity against E. faecalis (Jurica et al. 2017). Different species of Lactobacillus producing metabolites such as 2,4-hexadienoic acid and hydroxypentadecanedioic acid have shown antimicrobial activity against Candida vini (Lipinska-Zubrycka et al. 2020). L. rhamnosus and L. salivarius, producing valine, acetate, ethanol, 2-3-butanediol, uridine, 3 hydroxyphenylacetate have shown antibacterial activity against L. pneumophila (Fuochi et al. 2019). L. plantarum, producing organic acids such as 1,2-benzenedicarboxylic, palmitic, oleic, pentadecanoic acid, inhibited the growth of E. coli (Kanjan and Hongpattarakere 2016). Lactiplantibacillus plantarum producing the different metabolites showed antibacterial activity against S. aureus (Ray Mohapatra et al. 2022).

The selected four probiotic strains showed moderate hydrophobicity (35.88–56.70%) towards xylene (apolar solvent), with L. delbrueckii P31Mcs (56.70%) showing highest hydrophobicity. Cell surface hydrophobicity and auto-aggregation properties correlate with adhesion of the probiotics to the epithelial cells for a longer time, a prerequisite for preventing colonization of pathogens at the epithelial surface and for ensuring health benefits (Krausova et al. 2019). Hydrophobicity of any probiotic is measured by evaluating their affinity to hydrocarbon solvent by microbial adhesion to hydrocarbon (MATH) method. Hydrophobicity can be divided into low (< 33%), moderate (33–66%), or high (> 66%) (Fonseca et al. 2021). Bacteria with higher hydrophobicity can bind better to epithelial cells and thus prevent colonization of pathogens. Interestingly, E. faecium P15Mcm exhibited strong auto-aggregation and others showed moderate auto-aggregation. Different vaginal L. fermentum species have shown 60–80% auto-aggregation, whereas vaginal L. gasseri UBLG36 has shown 32.98% auto-aggregation (Ahire et al. 2021). The selected probiotic strains produced strong biofilm on the plastic surface of the 96-well microplate with higher production by L. fermentum P37Mws. Biofilm formation by probiotic bacteria is considered to be a beneficial property because it prevents the colonization of pathogenic bacteria (Salas-Jara et al. 2016). Biofilm producing probiotics such as L. rhamnosus and L. reuteri have been successfully used in adjuvant treatments of bacterial vaginosis (Ventolini 2015). Earlier, two strains of L. plantarum, LSC3 and LSC22 have shown biofilm formation, whereas three strains of L. plantarum LSC did not produce biofilm on plastic surfaces (Gheziel et al. 2019). L. delbrueckii HY5 and three strains of L. fermentum (RGM3, RCM11 and RCM13) have been reported to produce strong biofilm (Aziz et al. 2019). Our results are in agreement with the reported biofilm production by L. plantarum and L. fermentum (Gheziel et al. 2019; Aziz et al. 2019).

L. gasseri P36Mops, L. fermentum P37Mws and E. faecium P15Mcm showed resistance to most of the antibiotics such as norfloxacin, nitrofuramion, metronidazole, nalidixic acid and cotrimoxazole. These antimicrobials are generally used for the treatment of BV (Bradshaw and Sobel 2016; Larsson et al. 2011; Schwebke and Desmond 2007) and urinary tract infections (Anger et al. 2019). Most of the vaginal Lactobacillus sp. has been reported to show resistance towards norfloxacin, nitrofuramion, nalidixic acid (Fonseca et al. 2021) and co-trimoxazole (Salas-Jara et al. 2016) and metronidazole (Mastromarino et al. 2002; Pithva et al. 2014). Resistance to antibiotics is considered to be a positive feature in vaginal microbiota restoration therapy (Wiik et al. 2019). Decrease in Lactobacilli load favours the growth of HPV and microorganisms in bacterial vaginosis (Happel et al. 2020). A significant correlation has been reported between BV and cervical cancer (Muñoz et al. 2006). Recent evidences have suggested that besides HPV infection, cervical microflora play an important role in the development of cervical cancer (Muñoz et al. 2006). Thus, the resistant probiotics strains, L. gasseri P36Mops, L. fermentum P37Mws, L. delbrueckii P31Mcs and E. faecium P15Mcm could be used to restore the normal cervico-vaginal microflora during the vaginal infections.

Probiotics work through different mechanisms through 1) production of organic acids (lactic acid and short chain fatty acids) that maintain vaginal pH below 4.5 and antimicrobial agents such as hydrogen peroxide, bacteriocins and peptides (Tachedjian et al. 2017); 2) increase in the mucosal viscosity in the vagina (Di Cerbo 2016); 3) stimulation of the immune system (Di Cerbo 2016); and 4) formation of biofilms at the epithelial layer, thus inhibiting colonization of pathogens at the epithelial surface of the cells (Di Cerbo 2016; Mitra et al. 2020). Probiotic supplementation in cervical cancer patients has been reported to reduce the radiotherapy associated side effects such as diarrhoea, vaginal dryness, itching and increased risk of vaginal infections (Linn et al. 2019). The present probiotics hold a great promise in managing cervical cancer-associated bacterial infections and may also help in preventing chemo/radio therapy-associated side effects in cervical cancer patients.

Conclusion

The present study indicated that healthy vaginal ecosystem is an excellent source of Lactobacillus and Enteroccocus sp. with promising probiotic characteristics that could be explored to manage cervical cancer-associated infections. Probiotic LAB supplementations in cervical cancer patients can help in restoration of abnormal vaginal flora, prevent vaginal dysbiosis and help in attenuating therapy associated side effects. However, extensive studies need to be done to identify the most beneficial and safe probiotic strains for the treatment of cervical cancer associated pathogens as well as management of cervical cancer.

Declarations

Acknowledgements

The authors would like to thank Director, IRSHA, Dr A C Mishra for constant support and encouragement.  

Funding information

This study was funded by the Department of Science and Technology Women Scientist Scheme-A (DST WOS-A), Government of India. 

Data availability 

The data generated in this study has been included in the article and in the supplementary information files.

Compliance with ethical standards

The study was conducted according to the guidelines of the Helsinki Declaration and all the procedures involving human patients was approved by the Institutional Ethics Committee (IEC)s of Bharati Vidyapeeth (Deemed to be) University Medical College (Ref: BVDU/MC/57) and B. J. Government Medical College Sassoon General Hospitals (Ref No. BJGMC/IEC/Pharmac/ND-Dept 0119007-007], Pune. 

Conflicts of interest

The authors declare that there are no conflicts of interest.

References

  1. Adebamowo SN, Ma B, Zella D, Famooto A, Ravel J, ,AdebamowoC,ACCME Research Group (2017) Mycoplasma hominis and Mycoplasma genitalium in the vaginal microbiota and persistent high-risk human papillomavirus infection. Front Public Health. 5:140.doi: 10.3389/fpubh.2017.00140
  2. Ahire JJ, Sahoo S, Kashikar MS, Heerekar A, Lakshmi SG, Madempudi RS (2021) In Vitro Assessment of Lactobacillus crispatus UBLCp01, Lactobacillus gasseri UBLG36, and Lactobacillus johnsonii UBLJ01 as a potential vaginal probiotic candidate. Probiotics Antimicrob Proteins 20:1–2. https://doi.org/10.1007/s12602-021-09838-9
  3. Anderson AC, Sanunu M, Schneider C, Clad A, Karygianni L, Hellwig E, Al-Ahmad A (2014) Rapid species-level identification of vaginal and oral lactobacilli using MALDI-TOF MS analysis and 16S rDNA sequencing. BMC Microbiol 14:1–9. https://dx.doi.org/10.1186%2Fs12866-014-0312-5
  4. Andreeva P, Shterev A, Danova S (2016) Antimicrobial activity of vaginal lactobacilli against Gardnerella vaginalis and pathogens. Int J Adv Res Biol Sci 3:200–207. SOI: http://s-o-i.org/1.15/ijarbs-2016-3-5-29
  5. Anger J, Lee U, Ackerman AL, Chou R, Chughtai B, Clemens JQ, Hickling D et al (2019) Recurrent uncomplicated urinary tract infections in women: AUA/CUA/SUFU guideline. J Urol 202:282–289. doi: 10.1097/JU.0000000000000296
  6. Aphale S, Pandita S, Raina P, Mishra JN, Kaul-Ghanekar R (2018) Phytochemical standardization of panchavalkala: An ayurvedic formulation and evaluation of its anticancer activity in cervical cancer cell lines. Phcog Mag 14:554–560. DOI: 10.4103/pm.pm_252_18
  7. Archer AC, Halami PM (2015) Probiotic attributes of Lactobacillus fermentum isolated from human feces and dairy products. Appl Microbiol Biotechnol 99:8113–8123. https://doi.org/10.1080/14712598.2021.1828858
  8. Aridoss G, Amirthaganesan S, Kumar NA, Kim JT, Lim KT, Kabilan S, Jeong YT (2008) A facile synthesis, antibacterial, and antitubercular studies of some piperidin-4-one and tetrahydropyridine derivatives. Bioorg Med Chem Lett 18:6542–6548. doi: 10.1016/j.bmcl.2008.10.045
  9. Azat R, Liu Y, Li W, Kayir A, Lin DB, Zhou WW, Zheng XD (2016) Probiotic properties of lactic acid bacteria isolated from traditionally fermented Xinjiang cheese. J Zhejiang Univ Sci B 17:597–609. doi: 10.1631/jzus.B1500250
  10. Azcarate-Peril MA, Altermann E, Goh YJ, Tallon R, Sanozky-Dawes RB, Pfeiler EA et al (2008) Analysis of the genome sequence of Lactobacillus gasseri ATCC 33323 reveals the molecular basis of an autochthonous intestinal organism. Appl Environ Microbiol 74:4610–4625. doi: 10.1128/AEM.00054-08
  11. Aziz K, Tariq M, Zaidi A (2019) Biofilm development in L. fermentum under shear flow & sequential GIT digestion. FEMS Microbiol Lett 366:fnz064. doi: 10.1093/femsle/fnz064
  12. Bawa S, Kumar S, Drabu S, Kumar R (2009) Synthesis and antimicrobial activity of 2-chloro-6-methylquinoline hydrazone derivatives. J Pharm Bioall Sci 1(1):27. doi: 10.4103/0975-7406.62683
  13. Bayer AW, Kirby WM, Sherris JC, Turck M (1966) Antibiotic susceptibility testing by a standardized single disc method. Am J Clin Pathol 45:493–496. doi:10.1093/ajcp/45.4_ts.493
  14. Bello Gonzalez TdJ, Pham P, Top J, Willems RJL, van Schaik W, van Passel MWJ, Smidt H (2017) Characterization of Enterococcus isolates colonizing the intestinal tract of intensive care unit patients receiving selective digestive decontamination. Front Microbiol 8:1596. doi: 10.3389/fmicb.2017.01596
  15. Biswal BM, Singh KK, Ismail MB, Jalal MI, Safruddin EI (2014) Current concept of bacterial vaginosis in cervical cancer. J Clin Gynecol Obstet 3:1–7. http://dx.doi.org/10.14740/jcgo175w
  16. Bouridane H, Sifour M, Idoui T, Annick L, Thonard P (2016) Technological and probiotic traits of the Lactobacilli isolated from vaginal tract of the healthy women for probiotic use. Iran J Biotechnol 14:192–201. doi: 10.15171/ijb.1432
  17. Bradshaw CS, Sobel JD (2016) Current treatment of bacterial vaginosis limitations and need for innovation. J Infect Dis 214(suppl1):S14–S20. https://doi.org/10.1093/infdis/jiw159
  18. Brandt K, Nethery MA, O’Flaherty S, Barrangou R (2020) Genomic characterization of Lactobacillus fermentum DSM 20052. BMC Genomics 21:328. https://doi.org/10.1186/s12864-020-6740-8
  19. Cartagena E, Montanaro S, Bardón A (2008) Improvement of the antibacterial activity of sesquiterpene lactones. Rev Latinoam Química 36:43–50
  20. Caselli E, D’Accolti M, Santi E, Soffritti I, Conzadori S, Mazzacane S, Greco P et al (2020) Vaginal microbiota and cytokine microenvironment in HPV clearance/persistence in women surgically treated for cervical intraepithelial neoplasia: an observational prospective study. Front Cell Infect Microbiol 10:540900. doi: 10.3389/fcimb.2020.540900
  21. Chen Y, Hong Z, Wang W, Gu L, Gao H, Qiu L, Di W (2019) Association between the vaginal microbiome and high-risk human papillomavirus infection in pregnant Chinese women. BMC Infect Dis 19:677. https://doi.org/10.1186/s12879-019-4279-6
  22. Cretton S, Genta-Jouve G, Kaiser M, Mäser P, Muñoz O, Bürgi T, Cuendet M et al (2021) Hygroline derivatives from Schizanthus tricolor and their anti-trypanosomatid and antiplasmodial activities. Phytochem 192:112957. https://doi.org/10.1016/j.phytochem.2021.112957
  23. Dasari S, Shouri RN, Wudayagiri R, Valluru L (2014) Antimicrobial activity of Lactobacillus against microbial flora of cervicovaginal infections. Asian Pac J Trop Dis 4:18–24. doi: 10.1016/S2222-1808(14)60307-8
  24. de Souza Rodrigues JZ, Passos MR, de Macêdo Neres NS, Almeida RS, Pita LS, Santos IA Silveira PH(2020) Antimicrobial activity of Lactobacillus fermentum TcUESC01 against Streptococcus mutans UA159.Microb Pathog142:104063. https://doi.org/10.1016/j.micpath.2020.104063
  25. Di Cerbo A, Palmieri B, Aponte M, Morales-Medina JC, Iannitti T (2016) Mechanisms and therapeutic effectiveness of lactobacilli. J Clin Pathol 69:187–203. doi: 10.1136/jclinpath-2015-202976
  26. Dinçer E, Kıvanç M (2021) In vitro evaluation of probiotic potential of Enterococcus faecium strains isolated from Turkish pastırma. Arch Microbiol 203:2831–2841. doi: 10.1007/s00203-021-02273-y
  27. Do Nascimento RF, De Sales IR, de Oliveira Formiga R, Barbosa-Filho JM, Sobral MV, Tavares JF, Diniz MD et al (2015) Activity of alkaloids on peptic ulcer: what’s new? Molecules20:929-950.10.3390/molecules20010929
  28. Ehrmann MA, Kurzak P, Bauer J, Vogel RF (2002) Characterization of lactobacilli towards their use as probiotic adjuncts in poultry. J Appl Microbiol 92:966–975. doi: 10.1046/j.1365-2672.2002.01608.x
  29. El-Saber Batiha G, Alqahtani A, Ilesanmi OB, Saati AA, El-Mleeh A, Hetta HF, Magdy Beshbishy A (2020) Avermectin derivatives, pharmacokinetics, therapeutic and toxic dosages, mechanism of action, and their biological effects. Pharmaceuticals 13(8):196. https://doi.org/10.3390/ph13080196
  30. Esfandyari-Manesh M, Ghaedi Z, Asemi M, Khanavi M, Manayi A, Jamalifar H, Atyabi F et al (2013) Study of antimicrobial activity of anethole and carvone loaded PLGA nanoparticles. J Pharm Res 7:290–295. https://doi.org/10.1016/j.jopr.2013.04.019
  31. Fonseca HC, de Sousa Melo D, Ramos CL, Dias DR, Schwan RF (2021) Probiotic properties of lactobacilli and their ability to inhibit the adhesion of enteropathogenic bacteria to Caco-2 and HT-29 cells. Probiotics Antimicrob Proteins 13:102–112. https://doi.org/10.1007/s12602-020-09659-2
  32. Fuochi V (2016) Amensalistic activity of Lactobacillus spp., isolated from human samples. Università degli Studi di Catania, Catania
  33. Fuochi V, Coniglio MA, Laghi L, Rescifina A, Caruso M, Stivala A, Furneri PM (2019) Metabolic characterization of supernatants produced by Lactobacillus spp. with in vitro anti-Legionella activity. Front Microbiol 10:1403. doi: 10.3389/fmicb.2019.01403
  34. Gao Y, Shang Q, Li W, Guo W, Stojadinovic A, Mannion C, Man YG et al (2020) Antibiotics for cancer treatment: A double-edged sword. J Cancer 11:5135–5149. doi: 10.7150/jca.47470
  35. Gheziel C, Russo P, Arena MP, Spano G, Ouzari HI, Kheroua O, Saidi D (2019) Evaluating the probiotic potential of Lactobacillus plantarum strains from Algerian infant feces: towards the design of probiotic starter cultures tailored for developing countries. Probiotics Antimicrob Proteins 11:113–123. doi: 10.1007/s12602-018-9396-9
  36. Godoy-Vitorino F, Romaguera J, Zhao C, Vargas-Robles D, Ortiz-Morales G, Vázquez-Sánchez F, Sanchez-Vázquez M et al (2018) Cervicovaginal fungi and bacteria associated with cervical intraepithelial neoplasia and high-risk human papillomavirus infections in a hispanic population. Front Microbiol 9:2533. doi: 10.3389/fmicb.2018.02533
  37. Happel AU, Kullin B, Gamieldien H, Wentzel N, Zauchenberger CZ, Jaspan HB, Dabee S et al (2020) Exploring potential of vaginal Lactobacillus isolates from South African women for enhancing treatment for bacterial vaginosis. PLoS Pathog 16:e1008559. doi: 10.1371/journal.ppat.1008559
  38. Hatta MN, Mohamad Hanif EA, Chin SF, Neoh HM (2021) Pathogens and Carcinogenesis: A Review. Biology 10:533. https://doi.org/10.3390/biology10060533
  39. Jeon JH, Lee CH, Lee HS (2009) Antimicrobial activities of 2-methyl-8-hydroxyquinoline and its derivatives against human intestinal bacteria. J Korean Soc Appl Biol Chem 52:202–205. https://doi.org/10.3839/jksabc.2009.037
  40. Juárez Tomás MS, Wiese B, Nader-Macías ME (2005) Effects of culture conditions on the growth and auto‐aggregation ability of vaginal Lactobacillus johnsonii CRL 1294. J Appl Microbiol 99:1383–1391. doi:10.1111/j.1365-2672.2005.02726.x
  41. Jurica K, Gobin I, Kremer D, Čepo DV, Grubešić RJ, Karačonji IB, Kosalec I (2017) Arbutin and its metabolite hydroquinone as the main factors in the antimicrobial effect of strawberry tree (Arbutus unedo L.) leaves. J Herb Med 8:17–23. doi.org/10.1016/j.hermed.2017.03.006
  42. Kalia N, Singh J, Kaur M (2020) Microbiota in vaginal health and pathogenesis of recurrent vulvovaginal infections: a critical review. Ann Clin Microbiol Antimicrob 19:1–19. https://doi.org/10.1186/s12941-020-0347-4
  43. Kang CH, Han SH, Kim Y, Paek NS, So JS (2018) In vitro probiotic properties of Lactobacillus salivarius MG242 isolated from human vagina. Probiotics Antimicrob Proteins 10:343–349. doi 10.1007/s12602-017-9323-5
  44. Kanjan P, Hongpattarakere T (2016) Antibacterial metabolites secreted under glucose-limited environment of the mimicked proximal colon model by lactobacilli abundant in infant feces. Appl Microbiol Biotechnol 100:7651–7664. https://doi.org/10.1007/s00253-016-7606-5
  45. Krausova G, Hyrslova I, Hynstova I (2019) In vitro evaluation of adhesion capacity, hydrophobicity, and auto-aggregation of newly isolated potential probiotic strains. Fermentation 5:1–11. doi:10.3390/fermentation5040100
  46. Kurli R, Chaudhari D, Pansare AN, Khairnar M, Shouche YS, Rahi P (2018) Cultivable microbial diversity associated with cellular phones. Front Microbiol 9:1229. doi: 10.3389/fmicb.2018.01229
  47. Kyrgiou M, Mitra A, Moscicki AB (2017) Does the vaginal microbiota play a role in the development of cervical cancer? Transl Res 179:168–182. doi:10.1016/j.trsl.2016.07.004
  48. Larsson PG, Brandsborg E, Forsum U, Pendharkar S, Andersen KK, Nasic S, Hammarström L et al (2011) Extended antimicrobial treatment of bacterial vaginosis combined with human lactobacilli to find the best treatment and minimize the risk of relapses. BMC Infect Dis 11:1–4. doi.org/10.1186/1471-2334-11-223
  49. Linn YH, Thu KK, Win NH (2019) Effect of probiotics for the prevention of acute radiation-induced diarrhoea among cervical cancer patients: a randomized double-blind placebo-controlled study. Probiotics Antimicrob Proteins 11(2):638–647. https://doi.org/10.1007/s12602-018-9408-9
  50. Lipinska-Zubrycka L, Klewicki R, Sojka M, Bonikowski R, Milczarek A, Klewicka E (2020) Anticandidal activity of Lactobacillus spp. in the presence of galactosyl polyols. Microbiol Res 240:126540. https://doi.org/10.1016/j.micres.2020.126540
  51. Mastromarino P, Brigidi P, Macchia S, Maggi L, Pirovano F, Trinchieri V, Conte U et al (2002) Characterization and selection of vaginal Lactobacillus strains for the preparation of vaginal tablets. J Appl Microbiol 93:884–893. doi: 10.1046/j.1365-2672.2002.01759.x
  52. Matsumoto Y, Kaihatsu K, Nishino K, Ogawa M, Kato N, Yamaguchi A (2012) Antibacterial and antifungal activities of new acylated derivatives of epigallocatechin gallate. Front Microbiol 3:53. doi: 10.3389/fmicb.2012.00053
  53. Mejía-Caballero A, Salas-Villagrán VA, Jiménez-Serna A, Farrés A (2021) Challenges in the production and use of probiotics as therapeuticals in cancer treatment or prevention. J Ind Microbiol Biotechnol 48(9–10). :kuab052.https://doi.org/10.1093/jimb/kuab052
  54. Mitra A, MacIntyre DA, Ntritsos G, Smith A, Tsilidis KK, Marchesi JR, Bennett PR et al (2020) The vaginal microbiota associates with the regression of untreated cervical intraepithelial neoplasia 2 lesions. Nat Commun 11:1–3. https://doi.org/10.1038/s41467-020-15856-y
  55. Mohammed SJ, Amin HH, Aziz SB, Sha AM, Hassan S, Abdul Aziz JM, Rahman HS (2019) Structural characterization, antimicrobial activity, and in vitro cytotoxicity effect of black seed oil. Evid Based Complement Alternat Med2019:1–9. https://doi.org/10.1155/2019/6515671
  56. Morandi S, Cremonesi P, Povolo M, Brasca M (2012) Enterococcus lactis sp. nov., from Italian raw milk cheeses. Int J Syst Evol Microbiol 62:1992–1996. doi: 10.1099/ijs.0.030825-0
  57. Mubarak M, Kazi JI (2014) Acute renal failure in a young female with vaginal bleeding with partial recovery. J Nephropathol 3:45–47. doi: 10.12860/jnp.2014.10
  58. Mulu W, Yimer M, Zenebe Y, Abera B (2015) Common causes of vaginal infections and antibiotic susceptibility of aerobic bacterial isolates in women of reproductive age attending at Felegehiwot Referral Hospital, Ethiopia: a cross sectional study. BMC Womens Health 15:1–9. doi: 10.1186/s12905-015-0197-y
  59. Muñoz N, Castellsagué X, de González AB, Gissmann L (2006) HPV in the etiology of human cancer. Vaccine 24:S1–S0. https://doi.org/10.1016/j.vaccine.2006.05.115
  60. Navidinia M, Karimi A, Rahbar M, Fallah F, Ahsani RR, Malekan MA, Jahromi MH et al (2012) Study prevalence of verotoxigenic E. coli isolated from urinary tract infections (UTIs) in an Iranian children hospital. Open Microbiol J 6:1–4. doi: 10.2174/1874285801206010001
  61. Navidinia M, Peerayeh SN, Fallah F, Bakhshi B, Sajadinia RS (2014) Phylogenetic grouping and pathotypic comparison of urine and fecal Escherichia coli isolates from children with urinary tract infection. Brazilian J Microbiol 45:509–514. doi: 10.1590/s1517-83822014000200019
  62. Oh NS, Joung JY, Lee JY, Kim Y (2018) Probiotic and anti-inflammatory potential of Lactobacillus rhamnosus 4B15 and Lactobacillus gasseri 4M13 isolated from infant feces. PLoS ONE 14:13:e0192021. doi: 10.1371/journal.pone.0192021
  63. Oh NS, Joung JY, Lee JY, Kim Y (2018) Probiotic and anti-inflammatory potential of Lactobacillus rhamnosus 4B15 and Lactobacillus gasseri 4M13 isolated from infant feces. PLoS ONE 14:13:e0192021. doi: 10.1371/journal.pone.0192021
  64. Ouarabi L, Chait YA, Seddik HA, Drider D, Bendali F (2019) Newly isolated lactobacilli strains from Algerian human vaginal microbiota: Lactobacillus fermentum strains relevant probiotic’s candidates. Probiotics Antimicrob Proteins 11:43–54. doi: 10.1007/s12602-017-9360-0
  65. Petrina MA, Cosentino LA, Rabe LK, Hillier SL (2017) Susceptibility of bacterial vaginosis (BV)-associated bacteria to secnidazole compared to metronidazole, tinidazole and clindamycin. Anaerobe 47:115–119. doi: 10.1016/j.anaerobe.2017.05.005
  66. Pinheiro L, Brito CI, De Oliveira A, Martins PY, Pereira VC, Da Cunha MD (2015) Staphylococcus epidermidis and Staphylococcus haemolyticus: molecular detection of cytotoxin and enterotoxin genes. Toxins 7(9):3688–3699. doi: 10.3390/toxins7093688
  67. Pino A, Bartolo E, Caggia C, Cianci A, Randazzo CL (2019) Detection of vaginal lactobacilli as probiotic candidates. Sci Rep 9:3355. doi: 10.1038/s41598-019-40304-3
  68. Pithva S, Shekh S, Dave J, Vyas BR (2014) Probiotic attributes of autochthonous Lactobacillus rhamnosus strains of human origin. Appl Biochem Biotech 173:259–277. doi: 10.1007/s12010-014-0839-9
  69. Ray Mohapatra A, Harikrishnan A, Lakshmanan D, Jeevaratnam K (2022) Targeting Staphylococcus aureus and its biofilms with novel antibacterial compounds produced by Lactiplantibacillus plantarum SJ33. Arch Microbiol 204:1–5. doi: 10.1007/s00203-021-02630-x
  70. Reuben RC, Roy PC, Sarkar SL, Alam RU, Jahid IK (2019) Isolation, characterization, and assessment of lactic acid bacteria toward their selection as poultry probiotics. BMC Microbiol 19:1–20. ttps://doi.org/10.1186/s12866-019-1626-0
  71. Rocchetti G, Bernardo L, Pateiro M, Barba FJ, Munekata PE, Trevisan M, Lorenzo JM et al (2020) Impact of a pitanga leaf extract to prevent lipid oxidation processes during shelf life of packaged pork burgers: An untargeted metabolomic approach. Foods9:1668 10.3390/foods9111668
  72. Rodríguez-Sánchez DG, Pacheco A, Villarreal-Lara R, Ramos-González MR, Ramos-Parra PA, Granados-Principal S, de la Díaz RI et al (2019) Chemical profile and safety assessment of a food-grade acetogenin-enriched antimicrobial extract from avocado seed. Molecules 24:2354. doi: 10.3390/molecules24132354
  73. Salas-Jara MJ, Ilabaca A, Vega M, García A (2016) Biofilm forming Lactobacillus: new challenges for the development of probiotics. Microorganisms 4:1–14. doi: 10.3390/microorganisms4030035
  74. Sambrook J, Fritsch EF, Maniatis T (1989) Molecular Cloning a laboratory manual. Cold Spring Harbor Laboratory Press, New York
  75. Schoch CL, Ciufo S, Domrachev M, Hotton CL, Kannan S, Khovanskaya R et al (2020) NCBI Taxonomy: a comprehensive update on curation, resources and tools. Database (Oxford). 2020: baaa062. doi: 10.1093/database/baaa062
  76. Schwebke JR, Desmond R (2007) A randomized trial of metronidazole in asymptomatic bacterial vaginosis to prevent the acquisition of sexually transmitted diseases. Am J Obstet Gynecol 196:517–516. doi: 10.1016/j.ajog.2007.02.048
  77. Siegel RL, Miller KD, Jemal A (2018) Cancer statistics. CA Cancer J Clin 68:7–30. https://doi.org/10.3322/caac.21442
  78. Song YR, Choi MS, Choi GW, Park IK, Oh CS (2016) Antibacterial activity of cinnamaldehyde and estragole extracted from plant essential oils against Pseudomonas syringae pv. actinidiae causing bacterial canker disease in kiwifruit. Plant Pathol J 32:363–370. doi: 10.5423/PPJ.NT.01.2016.0006
  79. Tachedjian G, Aldunate M, Bradshaw CS, Cone RA (2017) The role of lactic acid production by probiotic Lactobacillus species in vaginal health. Res Microbiol 168:782–792. https://doi.org/10.1016/j.resmic.2017.04.001
  80. Tamura K, Stecher G, Peterson D, Filipski A, Kumar S (2013) MEGA6: molecular evolutionary genetics analysis version 6.0. Mol Biol Evol 30:2725–2729. doi: 10.1093/molbev/mst197
  81. Terraf ML, Juárez Tomás MS, Nader-Macías ME, Silva C (2012) Screening of biofilm formation by beneficial vaginal lactobacilli and influence of culture media components. J Appl Microbiol 113:1517–1529. doi: 10.1111/j.1365-2672.2012.05429.x
  82. Thulkar J, Kriplani A, Agarwal N (2012) A comparative study of oral single dose of metronidazole, tinidazole, secnidazole and ornidazole in bacterial vaginosis. Indian J Pharmacol 44:243–245. doi: 10.4103/0253-7613.93859
  83. Toval F, Köhler CD, Vogel U, Wagenlehner F, Mellmann A, Fruth A, Schmidt MA et al (2014) Characterization of Escherichia coli isolates from hospital inpatients or outpatients with urinary tract infection. J Clin Microbiol 52:407–418. doi: 10.1128/JCM.02069-13
  84. Tulgar S, Alasehir EA, Selvi O (2018) The antimicrobial activity of ephedrine and admixture of ephedrine and propofol: an in vitro study. Braz J Anesthesiol 68(1):69–74. doi: 10.1016/j.bjan.2017.08.001
  85. Turovskiy Y, Ludescher RD, Aroutcheva AA, Faro S, Chikindas ML (2009) Lactocin 160, a bacteriocin produced by vaginal Lactobacillus rhamnosus, targets cytoplasmic membranes of the vaginal pathogen, Gardnerella vaginalis. Probiotics Antimicrob Proteins 1:67–74. doi: 10.1007/s12602-008-9003-6
  86. Tyśkiewicz K, Konkol M, Kowalski R, Rój E, Warmiński K, Krzyżaniak M, Gil Ł et al (2019) Characterization of bioactive compounds in the biomass of black locust, poplar and willow, vol 33. Trees, pp 1235–1263. https://doi.org/10.1007/s00468-019-01837-2
  87. Ventolini G (2015) Vaginal Lactobacillus: biofilm formation in vivo–clinical implications. Int J Womens Health 7:243–247. doi: 10.2147/IJWH.S77956
  88. Wiik J, Sengpiel V, Kyrgiou M, Nilsson S, Mitra A, Tanbo T, Jonassen CM (2019) Cervical microbiota in women with cervical intra-epithelial neoplasia, prior to and after local excisional treatment, a Norwegian Cohort study. BMC Womens Health 19:1–9. doi: 10.1186/s12905-019-0727-0
  89. Wongkattiya N, Sanguansermsri P, Fraser IH, Sanguansermsri D (2019) Antibacterial activity of cuminaldehyde on food-borne pathogens, the bioactive component of essential oil from Cuminum cyminum L. collected in Thailand. J Complement Integr Med 16:20180195. doi: 10.1515/jcim-2018-0195
  90. Yoon SH, Ha SM, Kwon S, Lim J, Kim Y, Seo H, Chun J (2017) Introducing EzBioCloud: A taxonomically united database of 16S rRNA and whole genome assemblies. Int J Syst Evol Microbiol 67:1613–1617. doi: 10.1099/ijsem.0.001755
  91. Zheng J, Wittouck S, Salvetti E, Franz CM, Harris H, Mattarelli P et al (2020) A taxonomic note on the genus Lactobacillus: Description of 23 novel genera, emended description of the genus Lactobacillus Beijerinck 1901, and union of Lactobacillaceae and Leuconostocaceae. Int J Syst Evol Microbiol 70:2782–2858. doi 10.1099/ijsem.0.004107
  92. Zhou JW, Luo HZ, Jiang H, Jian TK, Chen ZQ, Jia AQ (2018) Hordenine: a novel quorum sensing inhibitor and antibiofilm agent against Pseudomonas aeruginosa. J Agric Food Chem 66:1620–1628. doi: 10.1021/acs.jafc.7b05035