1
|
Han G, Gu BH, Park SY, Park DS, Hwang SM, Ji W, Kim SB, Kim M. Topical administration of coumarin derivatives alleviates skin inflammatory symptoms in atopic dermatitis model. Biomed Pharmacother 2025; 186:118004. [PMID: 40157005 DOI: 10.1016/j.biopha.2025.118004] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2025] [Revised: 03/06/2025] [Accepted: 03/18/2025] [Indexed: 04/01/2025] Open
Abstract
The prevalence of atopic dermatitis (AD), a chronic inflammatory skin condition, is increasing. Coumarin derivatives, plant secondary metabolites, possess anti-inflammatory properties, but their specific role in AD treatment remain unclear. This study investigated the therapeutic potential of 7-geranyloxycoumarin (C#6), a selective coumarin derivative, in alleviating AD-like symptoms through multifaceted mechanisms. Among various coumarin derivatives tested, C#6 demonstrated remarkable efficacy in both in vitro and in vivo models. Notably, C#6 significantly suppressed interleukin-8 and thymic stromal lymphopoietin production in stimulated HaCaT cells. Experiments on an MC903-induced mouse model of AD revealed that topical administration of C#6 for 10 days led to a significant reduction in ear and epidermal thickness. Flow cytometry analysis showed a significant decrease in CD45 + leukocytes, eosinophils, and Th2 cells in C#6-treated AD mice. Importantly, 16S rRNA sequencing indicated that C#6 restored the disrupted skin microbiome by increasing the abundance of beneficial Lactobacillus and reducing pathogenic bacteria such as Enterobacteriaceae, Corynebacteriaceae, and Corynebacterium, contributing to maintaining skin microbiome balance. Molecular docking studies revealed high binding affinities of C#6 to key regulators, including NOD1, TLR2, PAR2, and TLR3, suggesting a role in modulating critical inflammatory pathways. Additionally, co-culture experiments revealed that C#6 treatment of TNF-α and IFN-γ-stimulated HaCaT cells suppressed inflammatory cytokines expression by THP-1 cells. Collectively, these findings demonstrate that C#6 exerts its anti-atopic effects by suppressing Th2-driven inflammation, reducing eosinophilic infiltration, modulating immune-epidermal crosstalk, and maintaining skin microbiome homeostasis, highlighting its potential as a promising therapeutic agent for AD management.
Collapse
Affiliation(s)
- Goeun Han
- Laboratory of Animal Immunology, Department of Animal Science, College of Natural Resource & Life Science, Pusan National University, Miryang, Republic of Korea; Institute for Future Earth, JYS Institute for Basic Science, Pusan National University, Busan, Republic of Korea
| | - Bon-Hee Gu
- Life and Industry Convergence Research Institute, Pusan National University, Miryang, Republic of Korea
| | - Sun Young Park
- Institute for Future Earth, JYS Institute for Basic Science, Pusan National University, Busan, Republic of Korea
| | - Da Som Park
- Development Division, GC Cell Corp., Yongin, Republic of Korea
| | - Su Mi Hwang
- Department of Biomedical Laboratory Science, College of Health and Medical Science, Sangji University, Wonju, Republic of Korea
| | - Woonhak Ji
- Laboratory of Animal Immunology, Department of Animal Science, College of Natural Resource & Life Science, Pusan National University, Miryang, Republic of Korea; Institute for Future Earth, JYS Institute for Basic Science, Pusan National University, Busan, Republic of Korea
| | - Seon Beom Kim
- Institute for Future Earth, JYS Institute for Basic Science, Pusan National University, Busan, Republic of Korea; Department of Food Science & Technology, College of Natural Resource & Life Science, Pusan National University, Miryang, Republic of Korea.
| | - Myunghoo Kim
- Laboratory of Animal Immunology, Department of Animal Science, College of Natural Resource & Life Science, Pusan National University, Miryang, Republic of Korea; Institute for Future Earth, JYS Institute for Basic Science, Pusan National University, Busan, Republic of Korea.
| |
Collapse
|
2
|
Do AD, Quang HP, Phan QK. Probiotic cell-free supernatant as effective antimicrobials against Klebsiella pneumoniae and reduce antibiotic resistance development. Int Microbiol 2025; 28:623-632. [PMID: 39117894 DOI: 10.1007/s10123-024-00575-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2024] [Revised: 08/01/2024] [Accepted: 08/06/2024] [Indexed: 08/10/2024]
Abstract
This study evaluated the antimicrobial activity, resistance development, and synergistic potential of cell-free supernatant (CFSs) derived from Levilactobacillus brevis (Lb-CFS) and Lactiplantibacillus plantarum (Lp-CFS) against Klebsiella pneumoniae. Both CFSs exhibited potent growth inhibition, with minimum inhibitory concentrations (MICs) of 128 μg/mL and 64 μg/mL for Lb-CFS and Lp-CFS, respectively, and demonstrated dose-dependent bactericidal activity, achieving complete bacterial eradication at minimum bactericidal concentrations (MBC) within 6 h. The CFSs suppressed the expression of virulence genes (galF, wzi, and manC) and biofilm formation in a dose-dependent manner. Synergistic interactions were observed when combining CFSs with antibiotics, resulting in 2- to fourfold reductions in antibiotic MICs and MBCs. Notably, adaptive evolution experiments revealed significantly slower resistance development in K. pneumoniae against CFSs (twofold MIC/MBC increase) compared to antibiotics (16- to 128-fold increase) after 21 days. Furthermore, CFS-adapted strains exhibited increased antibiotic susceptibility, while antibiotic-adapted strains displayed cross-resistance to multiple antibiotics. No cross-resistance occurred between Lb-CFS and Lp-CFS, suggesting distinct adaptive mechanisms. These findings highlight the potential of probiotic-derived CFSs as effective antimicrobials with a lower propensity for inducing rapid resistance compared to conventional antibiotics, suggesting their promise in combating multidrug-resistant infections.
Collapse
Affiliation(s)
- Anh Duy Do
- Department of Biotechnology, NTT Hi-Tech Institute, Nguyen Tat Thanh University, Ho Chi Minh City, 700000, Vietnam.
| | - Hoa Pham Quang
- Department of Biotechnology, NTT Hi-Tech Institute, Nguyen Tat Thanh University, Ho Chi Minh City, 700000, Vietnam
| | - Quang Khai Phan
- Department of Biotechnology, NTT Hi-Tech Institute, Nguyen Tat Thanh University, Ho Chi Minh City, 700000, Vietnam
| |
Collapse
|
3
|
Lam LN, Savage KE, Shakir CN, Lemos JA. Genome-wide analysis of Enterococcus faecalis genes that facilitate interspecies competition with Lactobacillus crispatus. J Bacteriol 2025; 207:e0043824. [PMID: 39902966 PMCID: PMC11925238 DOI: 10.1128/jb.00438-24] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2024] [Accepted: 01/15/2025] [Indexed: 02/06/2025] Open
Abstract
Enterococci are opportunistic pathogens notorious for causing a variety of infections. While both Enterococcus faecalis and Lactobacillus crispatus are commensal residents of the vaginal tract, the molecular mechanisms that enable E. faecalis to take advantage of a vaginal biome with lower counts of lactobacilli to colonize the vaginal tract and induce aerobic vaginitis remain unknown. Here, we show that L. crispatus eradicates E. faecalis in a contact-independent manner. Using transposon sequencing to identify E. faecalis OG1RF transposon (Tn) mutants that are either under-represented or over-represented when co-cultured with L. crispatus, we found that Tn mutants with disruption in the dltABCD operon, that encodes the proteins responsible for the D-alanylation of teichoic acids, and OG1RF_11697 encoding for an uncharacterized hypothetical protein are more susceptible to killing by L. crispatus. Inversely, Tn mutants with disruption in ldh1, which encodes for L-lactate dehydrogenase, are more resistant to L. crispatus killing. Using the Galleria mellonella infection model, we show that co-injection of L. crispatus with E. faecalis OG1RF enhances larvae survival while this L. crispatus-mediated protection was lost in larvae co-infected with either L. crispatus and E. faecalisΔldh1 or Δldh1Δldh2 strains. Last, using RNA sequencing to identify E. faecalis genes that are differently expressed in the presence of L. crispatus, we found major changes in the expression of genes associated with glycerophospholipid metabolism, central metabolism, and general stress responses. The findings in this study provide insights into how E. faecalis mitigate assaults by L. crispatus.IMPORTANCEEnterococcus faecalis is an opportunistic pathogen notorious for causing a multitude of infections. As vaginal commensals, E. faecalis must interact with Lactobacillus crispatus, but how E. faecalis overcomes or mitigate assaults by L. crispatus killing remains unknown. We show that L. crispatus eradicates E. faecalis temporally in a contact-independent manner. Using high-throughput molecular approaches, we identified genetic determinants that enable E. faecalis to compete with L. crispatus. This study represents an important first step for the identification of adaptive genetic traits required for enterococci to tolerate assaults by lactobacilli.
Collapse
Affiliation(s)
- Ling Ning Lam
- Department of Oral Biology, College of Dentistry, University of Florida, Gainesville, Florida, USA
| | - Kathryn E. Savage
- Department of Oral Biology, College of Dentistry, University of Florida, Gainesville, Florida, USA
| | - Camille N. Shakir
- Department of Oral Biology, College of Dentistry, University of Florida, Gainesville, Florida, USA
| | - José A. Lemos
- Department of Oral Biology, College of Dentistry, University of Florida, Gainesville, Florida, USA
| |
Collapse
|
4
|
Bedir E, Ay H, Kotil ES, Ozbek T. Isolation, genomic characterization and biotechnological evaluation of lactobacilli strains from chicken gastrointestinal tract. Microb Pathog 2025; 199:107142. [PMID: 39617076 DOI: 10.1016/j.micpath.2024.107142] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2024] [Revised: 11/15/2024] [Accepted: 11/20/2024] [Indexed: 12/10/2024]
Abstract
During a study investigating possible probiotics from chicken gut microbiota, strains C1-4 and C2-3 were isolated and identified as members of the genus Ligilactobacillus. The strains formed a well-supported cluster with Ligilactobacillus salivarius and Ligilactobacillus hayatikensis in phylogenetic trees. Their genomes, sized 1.8 Mb with G + C content of 32 %, were related to "Candidatus Avacholeplasma faecigallinarum" with a dDDH level of 95.4 %, indicating the strains were the first culturable members of the uncultured taxon. Furthermore, a dDDH value of 78.9 % with L. salivarius DSM 20555T suggested that the strains may represent a novel subspecies of L. salivarius. The functional analysis of the genomes revealed that the strains harbour genes associated with probiotic traits, including lactate utilization, acetoin and butanediol metabolism, pH homeostasis and exopolysaccharide biosynthesis. The genome annotation for the secondary metabolite biosynthesis gene clusters showed that the strains have a type III polyketide gene cluster and a bacteriocin immunity protein gene. The strains exhibited phenotypic features compatible with their potential use as probiotics, such as tolerance to low pH and NaCl, ability to achieve high auto-aggregation, and hydrophobicity properties. In addition, the strains exhibited strong antibacterial activity against pathogenic MRSA (Meticillin-resistant Staphylococcus aureus ATCC 67101), S. aureus ATCC 25923, Listeria monocytogenes MBG16, and VRSA (Vancomycin-resistant Staphylococcus aureus MBG89), while showing moderate activity against Salmonella Typhimurium MBG15, Escherichia coli ATCC 25922, Klebsiella pneumoniae ATCC 13833, and Pseudomonas aeruginosa ATCC 27853. The cell-free supernatant of the strains notably affected Lis. monocytogenes and S. aureus, possibly due to possible bacteriocin production. In conclusion, the strains isolated from chicken gut microbiota have a high potential to be used as probiotics in agriculture and medicine.
Collapse
Affiliation(s)
- Esra Bedir
- Yildiz Technical University, Faculty of Arts and Sciences, Department of Molecular Biology and Genetics, Istanbul, Turkey
| | - Hilal Ay
- Yildiz Technical University, Faculty of Arts and Sciences, Department of Molecular Biology and Genetics, Istanbul, Turkey
| | - Enes Seyfullah Kotil
- Bogazici University, Faculty of Arts and Sciences, Department of Molecular Biology and Genetics, Istanbul, Turkey
| | - Tulin Ozbek
- Yildiz Technical University, Faculty of Arts and Sciences, Department of Molecular Biology and Genetics, Istanbul, Turkey.
| |
Collapse
|
5
|
Acharyya S, Majumder S, Nandi S, Ghosh A, Saha S, Bhattacharya M. Uncovering mercury accumulation and the potential for bacterial bioremediation in response to contamination in the Singalila National Park. Sci Rep 2025; 15:3664. [PMID: 39881141 PMCID: PMC11779926 DOI: 10.1038/s41598-024-81927-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2024] [Accepted: 11/30/2024] [Indexed: 01/31/2025] Open
Abstract
Several recent investigations into montane regions have reported on excess mercury accumulation in high-altitude forest ecosystems. This study explored the Singalila National Park, located on the Singalila ridge of the Eastern Himalayas, revealing substantial mercury contamination. Particular focus was on Sandakphu (3636 m), the highest peak in West Bengal, India. It harboured 6.77 ± 0.01 mg/kg of total mercury in its topsoil. Further evidence was provided by accumulation in leaves (0.040 ± 0.01 mg/kg), and roots (0.150 ± 0.008 mg/kg) of local vegetation, litterfall (0.234 ± 0.019 mg/kg), mosses (0.367 ± 0.043 mg/kg), surface water from local lakes and waterbodies (0.010 ± 0.005 mg/l), fresh snow (0.014 ± 0.004 mg/l), and sleet (0.019 ± 0.009 mg/l). Samples from other points of varying elevation in the park also demonstrated contamination. The soil displayed a range of 0.068-5.28 mg/kg, while the mean concentration in leaves was 0.153 ± 0.105 mg/kg, roots was 0.106 ± 0.054 mg/kg, and leaf litter was 0.240 ± 0.112 mg/kg. Additionally, the microbial consortia isolated from the contaminated soil displayed a high tolerance to mercuric chloride, presumably gained through repeated and consistent exposure. Four high tolerance bacterial strains, MTS2C, MTS3A, MTS4B and MTS6A, were further characterized for potential use in bioremediation strategies. Their mercury removal capacities were determined to be 82.35%, 75.21%, 61.95%, and 37.47%, respectively. Overall, the findings presented provide evidence for a highly contaminated environment in the Singalila National Park, that poses significant ecological risk to the flora, fauna and local inhabitants of this biodiversity hotspot. This research also highlights the need for further exploration and monitoring of the Eastern Himalayas for its role as a sink for atmospheric mercury.
Collapse
Affiliation(s)
- Sukanya Acharyya
- Molecular Biology and Tissue Culture Laboratory, Department of Tea Science, University of North Bengal, Siliguri, West Bengal, India
| | - Soumya Majumder
- Molecular Biology and Tissue Culture Laboratory, Department of Tea Science, University of North Bengal, Siliguri, West Bengal, India
| | - Sudeshna Nandi
- Molecular Biology and Tissue Culture Laboratory, Department of Tea Science, University of North Bengal, Siliguri, West Bengal, India
| | - Arindam Ghosh
- Molecular Biology and Tissue Culture Laboratory, Department of Tea Science, University of North Bengal, Siliguri, West Bengal, India
| | - Sumedha Saha
- Molecular Biology and Tissue Culture Laboratory, Department of Tea Science, University of North Bengal, Siliguri, West Bengal, India
| | - Malay Bhattacharya
- Molecular Biology and Tissue Culture Laboratory, Department of Tea Science, University of North Bengal, Siliguri, West Bengal, India.
| |
Collapse
|
6
|
Li LQ, Yan JK, Tai WCS, Kwok KWH, Wu JY. Anti-bacterial effects, and metabolites derived from bifidobacterial fermentation of an exopolysaccharide of Cs-HK1 medicinal fungus. Carbohydr Polym 2024; 345:122577. [PMID: 39227109 DOI: 10.1016/j.carbpol.2024.122577] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2024] [Revised: 05/31/2024] [Accepted: 08/02/2024] [Indexed: 09/05/2024]
Abstract
This study was to investigate the antibacterial effects and metabolites derived from bifidobacterial fermentation of an exopolysaccharide EPS-LM produced by a medicinal fungus Cordyceps sinensis, Cs-HK1. EPS-LM was a partially purified polysaccharide fraction which was mainly composed of Man, Glc and Gal at 7.31:12.95:1.00 mol ratio with a maximum molecular weight of 360 kDa. After fermentation of EPS-LM in two bifidobacterial cultures, B. breve and B. longum, the culture digesta showed significant antibacterial activities, inhibiting the proliferation and biofilm formation of Escherichia coli. Based on untargeted metabolomic profiling of the digesta, the levels of short chain fatty acids, carboxylic acids, benzenoids and their derivatives were all increased significantly (p < 0.01), which probably contributed to the enhanced antibacterial activity by EPS-LM. Since EPS-LM was only slightly consumed for the bifidobacterial growth, it mainly stimulated the biosynthesis of bioactive metabolites in the bifidobacterial cells. The results also suggested that EPS-LM polysaccharide may have a regulatory function on the bifidobacterial metabolism leading to production of antibacterial metabolites, which may be of significance for further exploration.
Collapse
Affiliation(s)
- Long Qing Li
- Engineering Research Center of Health Food Design & Nutrition Regulation, Dongguan Key Laboratory of Typical Food Precision Design, China National Light Industry Key Laboratory of Healthy Food Development and Nutrition Regulation, School of Life and Health Technology, Dongguan University of Technology, Dongguan 523808, China; Research Institute for Future Food, Department of Food Science and Nutrition, The Hong Kong Polytechnic University, Hung Hom, Kowloon, Hong Kong
| | - Jing Kun Yan
- Engineering Research Center of Health Food Design & Nutrition Regulation, Dongguan Key Laboratory of Typical Food Precision Design, China National Light Industry Key Laboratory of Healthy Food Development and Nutrition Regulation, School of Life and Health Technology, Dongguan University of Technology, Dongguan 523808, China
| | - William Chi-Shing Tai
- Engineering Research Center of Health Food Design & Nutrition Regulation, Dongguan Key Laboratory of Typical Food Precision Design, China National Light Industry Key Laboratory of Healthy Food Development and Nutrition Regulation, School of Life and Health Technology, Dongguan University of Technology, Dongguan 523808, China
| | - Kevin Wing-Hin Kwok
- Engineering Research Center of Health Food Design & Nutrition Regulation, Dongguan Key Laboratory of Typical Food Precision Design, China National Light Industry Key Laboratory of Healthy Food Development and Nutrition Regulation, School of Life and Health Technology, Dongguan University of Technology, Dongguan 523808, China
| | - Jian Yong Wu
- Research Institute for Future Food, Department of Food Science and Nutrition, The Hong Kong Polytechnic University, Hung Hom, Kowloon, Hong Kong.
| |
Collapse
|
7
|
Liu T, Lei C, Huang Q, Song W, Li C, Sun N, Liu Z. Hesperidin and Fecal Microbiota Transplantation Modulate the Composition of the Gut Microbiota and Reduce Obesity in High Fat Diet Mice. Diabetes Metab Syndr Obes 2024; 17:3643-3656. [PMID: 39398388 PMCID: PMC11468570 DOI: 10.2147/dmso.s474034] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/17/2024] [Accepted: 09/30/2024] [Indexed: 10/15/2024] Open
Abstract
Introduction Obesity, which is associated with gut microbiota dysbiosis, low-grade chronic inflammation and intestinal barrier dysfunction, can cause a variety of chronic metabolic diseases. Phytochemical flavonoids have a variety of biological activities, among which there may be safe and effective anti-obesity solutions. Methods We tested a plant-derived flavonoid hesperidin and fecal microbiota transplantation (FMT) to alleviate diet-induced obesity. High-fat diet (HFD)-fed mice were treated with hesperidin (100 and 200 mg/kg BW) and FMT. Results Results indicated that hesperidin had the effects of reducing obesity as indicated by reduction of body weight, fat accumulation and blood lipids, reducing inflammation as indicated by reduction of pro-inflammation factors including TNFα, IL-6, IL-1βand iNOS, and improving gut integrity as indicated by increasing colon length, reducing plasma gut permeability indicators iFABP and LBP, increased mRNA expression of mucus protein Muc2, tight junction p Claudin 2, Occludin and ZO-1 in the HFD-fed mice. The anti-obesity effects of hesperidin treatment have a dose-dependent manner. In addition, 16S rRNA-based gut microbiota analysis revealed that hesperidin selectively promoted the growth of Lactobacillus salivarius, Staphylococcus sciuri and Desulfovibrio C21_c20 while inhibiting Bifidobacterium pseudolongum, Mucispirillum schaedleri, Helicobacter ganmani and Helicobacter hepaticus in the HFD-fed mice. Horizontal feces transfer from the normal diet (ND)-fed mice to the HFD-fed mice conferred anti-obesity effects and transmitted some of the HFD-modulated microbes. Conclusion We concluded that hesperidin and FMT both affect the reduction of body weight and improve HFD-related disorders in the HFD-fed mice possibly through modulating the composition of the gut microbiota.
Collapse
Affiliation(s)
- Ting Liu
- Key Laboratory of Biological Targeting Diagnosis, Therapy and Rehabilitation of Guangdong Higher Education Institutes, the Fifth Affiliated Hospital of Guangzhou Medical University, Guangzhou, Guangdong, 510799, People’s Republic of China
| | - Chao Lei
- Key Laboratory of Biological Targeting Diagnosis, Therapy and Rehabilitation of Guangdong Higher Education Institutes, the Fifth Affiliated Hospital of Guangzhou Medical University, Guangzhou, Guangdong, 510799, People’s Republic of China
| | - Qinhong Huang
- The First Clinical College, Guangzhou Medical University, Guangzhou, 511400, People’s Republic of China
| | - Weiqi Song
- Department of Public Health, Guangzhou Medical University, Guangzhou, People’s Republic of China
| | - Chen Li
- Key Laboratory of Biological Targeting Diagnosis, Therapy and Rehabilitation of Guangdong Higher Education Institutes, the Fifth Affiliated Hospital of Guangzhou Medical University, Guangzhou, Guangdong, 510799, People’s Republic of China
| | - Ning Sun
- Guangzhou 11th People’s Hospital, Guangzhou Cadre and Talent Health Management Center, Guangzhou, 510530, People’s Republic of China
- The State Key Laboratory of Chemical Biology and Drug Discovery, Department of Applied Biology and Chemical Technology, the Hong Kong Polytechnic University, 999077, Hong Kong, China
| | - Zhihua Liu
- Department of Anorectal Surgery, the Tenth Affiliated Hospital of Southern Medical University (Dongguan People’s Hospital), Dongguan, People’s Republic of China
| |
Collapse
|
8
|
Dobreva L, Atanasova N, Donchev P, Krumova E, Abrashev R, Karakirova Y, Mladenova R, Tolchkov V, Ralchev N, Dishliyska V, Danova S. Candidate-Probiotic Lactobacilli and Their Postbiotics as Health-Benefit Promoters. Microorganisms 2024; 12:1910. [PMID: 39338583 PMCID: PMC11434380 DOI: 10.3390/microorganisms12091910] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2024] [Revised: 09/17/2024] [Accepted: 09/17/2024] [Indexed: 09/30/2024] Open
Abstract
Lactobacillus species are widely recognized for their probiotic potential, focusing on their mechanisms of health benefits and protection. Here we conducted an in vitro investigation of the probiotic potential with a role in microbiome homeostasis of four strains: Lactiplantibacillus plantarum L6 and F53, Ligilactobacillus salivarius 1, and Lactobacillus helveticus 611. A broad spectrum of antibacterial and antifungal activity was determined. The strain-specific inhibition of Staphylococcus aureus, Streptococcus mutans, Escherichia coli, Pseudomonas aeruginosa, and saprophytic/toxigenic fungi makes them promising as protective cultures. DPPH (2,2-diphenyl-1-picrylhydrazyl) and ABTS (2,2'-azino-bis-(3-ethylbenzothiazoline-6-sulfonic) acid) measurements showed that tested samples had strain-specific capacity for scavenging of radicals. The molecular base for the antioxidant potential of two lyophilized forms of active strains was investigated by electron paramagnetic resonance spectroscopy. The MTT (3-(4,5-dimethylthiazol-2-yl)-2,5-diphenyltetrazolium bromide) assay, with fractions of the most active postbiotics obtained by SEC-FPLC (fast protein liquid chromatography) analysis, showed a wide variety of effects on the growth of a K562 myeloid leukemia cell line. The IC50 (half-maximal inhibitory concentration) of L. salivarius 1 was determined to be 46.15 mg/mL. The proven in vitro functionality of the selected lactobacilli make them suitable for development of target probiotics with specific beneficial effects expected in vivo. Further investigations on produced postbiotics and safety have to be completed before they can be considered as scientifically proven probiotic strains.
Collapse
Affiliation(s)
- Lili Dobreva
- The Stephan Angeloff Institute of Microbiology, Bulgarian Academy of Sciences, 26 Acad. G. Bonchev Str., 1113 Sofia, Bulgaria
| | - Nikoleta Atanasova
- The Stephan Angeloff Institute of Microbiology, Bulgarian Academy of Sciences, 26 Acad. G. Bonchev Str., 1113 Sofia, Bulgaria
| | - Petar Donchev
- The Stephan Angeloff Institute of Microbiology, Bulgarian Academy of Sciences, 26 Acad. G. Bonchev Str., 1113 Sofia, Bulgaria
| | - Ekaterina Krumova
- The Stephan Angeloff Institute of Microbiology, Bulgarian Academy of Sciences, 26 Acad. G. Bonchev Str., 1113 Sofia, Bulgaria
| | - Radoslav Abrashev
- The Stephan Angeloff Institute of Microbiology, Bulgarian Academy of Sciences, 26 Acad. G. Bonchev Str., 1113 Sofia, Bulgaria
| | - Yordanka Karakirova
- Institute of Catalysis, Bulgarian Academy of Sciences, 11 Acad. G. Bonchev Str., 1113 Sofia, Bulgaria
| | - Ralitsa Mladenova
- Institute of Catalysis, Bulgarian Academy of Sciences, 11 Acad. G. Bonchev Str., 1113 Sofia, Bulgaria
| | - Vladimir Tolchkov
- National Center of Infectious and Parasitic Diseases, Yanko Sakuzov Blvd 26, 1504 Sofia, Bulgaria
| | - Nikola Ralchev
- The Stephan Angeloff Institute of Microbiology, Bulgarian Academy of Sciences, 26 Acad. G. Bonchev Str., 1113 Sofia, Bulgaria
| | - Vladislava Dishliyska
- The Stephan Angeloff Institute of Microbiology, Bulgarian Academy of Sciences, 26 Acad. G. Bonchev Str., 1113 Sofia, Bulgaria
| | - Svetla Danova
- The Stephan Angeloff Institute of Microbiology, Bulgarian Academy of Sciences, 26 Acad. G. Bonchev Str., 1113 Sofia, Bulgaria
| |
Collapse
|
9
|
Subbarayudu S, Namasivayam SKR, Arockiaraj J. Immunomodulation in Non-traditional Therapies for Methicillin-resistant Staphylococcus aureus (MRSA) Management. Curr Microbiol 2024; 81:346. [PMID: 39240286 DOI: 10.1007/s00284-024-03875-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2024] [Accepted: 08/26/2024] [Indexed: 09/07/2024]
Abstract
The rise of methicillin-resistant Staphylococcus aureus (MRSA) poses a significant challenge in clinical settings due to its ability to evade conventional antibiotic treatments. This overview explores the potential of immunomodulatory strategies as alternative therapeutic approaches to combat MRSA infections. Traditional antibiotics are becoming less effective, necessitating innovative solutions that harness the body's immune system to enhance pathogen clearance. Recent advancements in immunotherapy, including the use of antimicrobial peptides, phage therapy, and mechanisms of immune cells, demonstrate promise in enhancing the body's ability to clear MRSA infections. However, the exact interactions between these therapies and immunomodulation are not fully understood, underscoring the need for further research. Hence, this review aims to provide a broad overview of the current understanding of non-traditional therapeutics and their impact on immune responses, which could lead to more effective MRSA treatment strategies. Additionally, combining immunomodulatory agents with existing antibiotics may improve outcomes, particularly for immunocompromised patients or those with chronic infections. As the landscape of antibiotic resistance evolves, the development of effective immunotherapeutic strategies could play a vital role in managing MRSA infections and reducing reliance on traditional antibiotics. Future research must focus on optimizing these approaches and validating their efficacy in diverse clinical populations to address the urgent need for effective MRSA management strategies.
Collapse
Affiliation(s)
- Suthi Subbarayudu
- Toxicology and Pharmacology Laboratory, Department of Biotechnology, Faculty of Science and Humanities, SRM Institute of Science and Technology, Kattankulathur, Chengalpattu District, Tamil Nadu, 603203, India
| | - S Karthick Raja Namasivayam
- Centre for Applied Research, Saveetha School of Engineering, Saveetha Institute of Medical and Technical Sciences (SIMATS), Chennai, Tamil Nadu, 602105, India.
| | - Jesu Arockiaraj
- Toxicology and Pharmacology Laboratory, Department of Biotechnology, Faculty of Science and Humanities, SRM Institute of Science and Technology, Kattankulathur, Chengalpattu District, Tamil Nadu, 603203, India.
| |
Collapse
|
10
|
Saini P, Ayyanna R, Kumar R, Bhowmick SK, Bhaskar V, Dey B. Restriction of growth and biofilm formation of ESKAPE pathogens by caprine gut-derived probiotic bacteria. Front Microbiol 2024; 15:1428808. [PMID: 39135871 PMCID: PMC11317286 DOI: 10.3389/fmicb.2024.1428808] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2024] [Accepted: 07/05/2024] [Indexed: 08/15/2024] Open
Abstract
The accelerated rise in antimicrobial resistance (AMR) poses a significant global health risk, necessitating the exploration of alternative strategies to combat pathogenic infections. Biofilm-related infections that are unresponsive to standard antibiotics often require the use of higher-order antimicrobials with toxic side effects and the potential to disrupt the microbiome. Probiotic therapy, with its diverse benefits and inherent safety, is emerging as a promising approach to prevent and treat various infections, and as an alternative to antibiotic therapy. In this study, we isolated novel probiotic bacteria from the gut of domestic goats (Capra hircus) and evaluated their antimicrobial and anti-biofilm activities against the 'ESKAPE' group of pathogens. We performed comprehensive microbiological, biochemical, and molecular characterizations, including analysis of the 16S-rRNA gene V1-V3 region and the 16S-23S ISR region, on 20 caprine gut-derived lactic acid bacteria (LAB). Among these, six selected Lactobacillus isolates demonstrated substantial biofilm formation under anaerobic conditions and exhibited robust cell surface hydrophobicity and autoaggregation, and epithelial cell adhesion properties highlighting their superior enteric colonization capability. Notably, these Lactobacillus isolates exhibited broad-spectrum growth inhibitory and anti-biofilm properties against 'ESKAPE' pathogens. Additionally, the Lactobacillus isolates were susceptible to antibiotics listed by the European Food Safety Authority (EFSA) within the prescribed Minimum Inhibitory Concentration limits, suggesting their safety as feed additives. The remarkable probiotic characteristics exhibited by the caprine gut-derived Lactobacillus isolates in this study strongly endorse their potential as compelling alternatives to antibiotics and direct-fed microbial (DFM) feed supplements in the livestock industry, addressing the escalating need for antibiotic-free animal products.
Collapse
Affiliation(s)
- Prerna Saini
- National Institute of Animal Biotechnology, Hyderabad, India
- Regional Centre for Biotechnology, Faridabad, India
| | - Repally Ayyanna
- National Institute of Animal Biotechnology, Hyderabad, India
| | - Rishi Kumar
- National Institute of Animal Biotechnology, Hyderabad, India
- Regional Centre for Biotechnology, Faridabad, India
| | - Sayan Kumar Bhowmick
- National Institute of Animal Biotechnology, Hyderabad, India
- Regional Centre for Biotechnology, Faridabad, India
| | - Vinay Bhaskar
- National Institute of Animal Biotechnology, Hyderabad, India
| | - Bappaditya Dey
- National Institute of Animal Biotechnology, Hyderabad, India
- Regional Centre for Biotechnology, Faridabad, India
| |
Collapse
|
11
|
Prema P, Ali D, Nguyen VH, Pradeep BV, Veeramanikandan V, Daglia M, Arciola CR, Balaji P. A Response Surface Methodological Approach for Large-Scale Production of Antibacterials from Lactiplantibacillus plantarum with Potential Utility against Foodborne and Orthopedic Infections. Antibiotics (Basel) 2024; 13:437. [PMID: 38786166 PMCID: PMC11118495 DOI: 10.3390/antibiotics13050437] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2024] [Revised: 05/06/2024] [Accepted: 05/09/2024] [Indexed: 05/25/2024] Open
Abstract
A variety of bacteria, including beneficial probiotic lactobacilli, produce antibacterials to kill competing bacteria. Lactobacilli secrete antimicrobial peptides (AMPs) called bacteriocins and organic acids. In the food industry, bacteriocins, but even whole cell-free supernatants, are becoming more and more important as bio-preservatives, while, in orthopedics, bacteriocins are introducing new perspectives in biomaterials technologies for anti-infective surfaces. Studies are focusing on Lactiplantibacillus plantarum (previously known as Lactobacillus plantarum). L. plantarum exhibits great phenotypic versatility, which enhances the chances for its industrial exploitation. Importantly, more than other lactobacilli, it relies on AMPs for its antibacterial activity. In this study, Response Surface Methodology (RSM) through a Box-Behnken experimental design was used to estimate the optimal conditions for the production of antibacterials by L. plantarum. A temperature of 35 °C, pH 6.5, and an incubation time of 48 h provided the highest concentration of antibacterials. The initial pH was the main factor influencing the production of antibacterials, at 95% confidence level. Thanks to RSM, the titer of antibacterials increased more than 10-fold, this result being markedly higher than those obtained in the very few studies that have so far used similar statistical methodologies. The Box-Behnken design turned out to be a valid model to satisfactorily plan a large-scale production of antibacterials from L. plantarum.
Collapse
Affiliation(s)
- Paulpandian Prema
- Department of Zoology, VHN Senthikumar Nadar College, Virudhunagar 626001, TN, India;
| | - Daoud Ali
- Department of Zoology, College of Science, King Saud University, P.O. Box 2455, Riyadh 11451, Saudi Arabia;
| | - Van-Huy Nguyen
- Centre for Herbal Pharmacology and Environmental Sustainability, Chettinad Hospital and Research Institute, Chettinad Academy of Research and Education, Kelambakkam 603103, TN, India;
| | - Bhathini Vaikuntavasan Pradeep
- Centre for Microbial Technology, Department of Microbiology, Karpagam Academy of Higher Education, Coimbatore 641021, TN, India; (B.V.P.); (V.V.)
| | - Veeramani Veeramanikandan
- Centre for Microbial Technology, Department of Microbiology, Karpagam Academy of Higher Education, Coimbatore 641021, TN, India; (B.V.P.); (V.V.)
| | - Maria Daglia
- Department of Pharmacy, University of Naples Federico II, Via D. Montesano 49, 80131 Naples, Italy;
- International Research Center for Food Nutrition and Safety, Jiangsu University, Zhenjiang 212013, China
| | - Carla Renata Arciola
- Laboratory of Immunorheumatology and Tissue Regeneration, Laboratory of Pathology of Implant Infections, IRCCS Istituto Ortopedico Rizzoli, Via di Barbiano 1/10, 40136 Bologna, Italy
- Department of Medical and Surgical Sciences (DIMEC), University of Bologna, Via San Giacomo 14, 40126 Bologna, Italy
| | - Paulraj Balaji
- PG and Research Centre in Biotechnology, MGR College, Hosur 635130, TN, India
| |
Collapse
|
12
|
Li J, Zhang Q, Zhao J, Zhang H, Chen W. Lactobacillus-derived components for inhibiting biofilm formation in the food industry. World J Microbiol Biotechnol 2024; 40:117. [PMID: 38429597 DOI: 10.1007/s11274-024-03933-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2023] [Accepted: 02/19/2024] [Indexed: 03/03/2024]
Abstract
Biofilm, a microbial community formed by especially pathogenic and spoilage bacterial species, is a critical problem in the food industries. It is an important cause of continued contamination by foodborne pathogenic bacteria. Therefore, removing biofilm is the key to solving the high pollution caused by foodborne pathogenic bacteria in the food industry. Lactobacillus, a commonly recognized probiotic that is healthy for consumer, have been proven useful for isolating the potential biofilm inhibitors. However, the addition of surface components and metabolites of Lactobacillus is not a current widely adopted biofilm control strategy at present. This review focuses on the effects and preliminary mechanism of action on biofilm inhibition of Lactobacillus-derived components including lipoteichoic acid, exopolysaccharides, bacteriocins, secreted protein, organic acids and some new identified molecules. Further, the review discusses several modern biofilm identification techniques and particularly interesting new technology of biofilm inhibition molecules. These molecules exhibit stronger inhibition of biofilm formation, playing a pivotal role in food preservation and storage. Overall, this review article discusses the application of biofilm inhibitors produced by Lactobacillus, which would greatly aid efforts to eradicate undesirable bacteria from environment in the food industries.
Collapse
Affiliation(s)
- Jiaxun Li
- State Key Laboratory of Food Science and Resources, Jiangnan University, Wuxi, Jiangsu, 214122, China
- School of Food Science and Technology, Jiangnan University, Wuxi, Jiangsu, 214122, China
| | - Qiuxiang Zhang
- State Key Laboratory of Food Science and Resources, Jiangnan University, Wuxi, Jiangsu, 214122, China.
- School of Food Science and Technology, Jiangnan University, Wuxi, Jiangsu, 214122, China.
| | - Jianxin Zhao
- State Key Laboratory of Food Science and Resources, Jiangnan University, Wuxi, Jiangsu, 214122, China
- School of Food Science and Technology, Jiangnan University, Wuxi, Jiangsu, 214122, China
| | - Hao Zhang
- State Key Laboratory of Food Science and Resources, Jiangnan University, Wuxi, Jiangsu, 214122, China
- School of Food Science and Technology, Jiangnan University, Wuxi, Jiangsu, 214122, China
- National Engineering Research Center for Functional Food, Jiangnan University, Wuxi, Jiangsu, 214122, China
| | - Wei Chen
- State Key Laboratory of Food Science and Resources, Jiangnan University, Wuxi, Jiangsu, 214122, China
- School of Food Science and Technology, Jiangnan University, Wuxi, Jiangsu, 214122, China
- National Engineering Research Center for Functional Food, Jiangnan University, Wuxi, Jiangsu, 214122, China
| |
Collapse
|
13
|
Poothong S, Tanasupawat S, Chanpongsang S, Kingkaew E, Nuengjamnong C. Anaerobic flora, Selenomonas ruminis sp. nov., and the bacteriocinogenic Ligilactobacillus salivarius strain MP3 from crossbred-lactating goats. Sci Rep 2024; 14:4838. [PMID: 38418870 PMCID: PMC10901824 DOI: 10.1038/s41598-024-54686-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2023] [Accepted: 02/15/2024] [Indexed: 03/02/2024] Open
Abstract
This study aimed to examine the distribution of anaerobic bacteria in the rumen fluid of Thai crossbred goats and to screen potential probiotic strains capable of producing antimicrobial compounds and inhibiting bacteria that cause milk fat depression. Thirty-four strains of bacteria from the rumen fluid were divided into 13 groups within 12 genera based on 16S rRNA gene sequences. The RF1-5 and RF5-12 were identified as Streptococcus luteliensis and Bacillus licheniformis, respectively, and demonstrated non-ropy exopolysaccharide. Furthermore, mPRGC5T was closely related to Selenomonas caprae JCM 33725 T (97.8% similarity) based on 16S rRNA gene sequences. It exhibited low average nucleotide identity, digital DNA-DNA hybridization, and average amino acid identity values with related type strains ranging from 84.9 to 86.0%, 21.3 to 21.8%, and 73.8 to 76.1%, respectively. The genotypic and phenotypic characteristics of mPRGC5T strongly support this strain as a new species of the genus Selenomonas for which the name Selenomonas ruminis mPRGC5T was proposed. The type strain is mPRGC5T (= JCM 33724 T = KCTC 25177 T). Ligilactobacillus salivarius MP3 showed antibacterial activity against Cutibacterium acnes subsp. acnes DSM 1897 T and Kocuria rhizophila MIII. The enterolysin A cluster gene was identified in its genome. The auto-aggregation of L. salivarius MP3 was 93.6 ± 0.2%. Additionally, co-aggregation of L. salivarius MP3 with C. acnes DSM 1897 T and K. rhizophila MIII had 92.2 ± 3.4% and 87.3 ± 4.5%, respectively. The adhesion capacity of strain MP3 was 76.11 ± 2.2%. Probiogenomic analysis revealed that L. salivarius MP3 was nonhazardous to animal supplementation and included acid- and bile-tolerant ability. However, strain MP3 contained three antibiotic resistance genes. Thus, the supplementation of L. salivarius MP3 could increase the milk fat content by suppressing C. acnes DSM 1897 T with antibiotic resistance gene horizontal transfer awareness.
Collapse
Affiliation(s)
- Saranporn Poothong
- Department of Animal Husbandry, Faculty of Veterinary Science, Chulalongkorn University, Bangkok, 10330, Thailand
| | - Somboon Tanasupawat
- Department of Biochemistry and Microbiology, Faculty of Pharmaceutical Sciences, Chulalongkorn University, Bangkok, 10330, Thailand.
| | - Somchai Chanpongsang
- Department of Animal Husbandry, Faculty of Veterinary Science, Chulalongkorn University, Bangkok, 10330, Thailand
| | - Engkarat Kingkaew
- Department of Biology, School of Sciences, King Mongkut's Institute of Technology Ladkrabang, Bangkok, 10520, Thailand
| | - Chackrit Nuengjamnong
- Department of Animal Husbandry, Faculty of Veterinary Science, Chulalongkorn University, Bangkok, 10330, Thailand.
- Center of Excellence for Food and Water Risk Analysis (FAWRA), Faculty of Veterinary Science, Chulalongkorn University, Bangkok, 10330, Thailand.
| |
Collapse
|
14
|
Carbonne C, Chadi S, Kropp C, Molimard L, Chain F, Langella P, Martin R. Ligilactobacillus salivarius CNCM I-4866, a potential probiotic candidate, shows anti-inflammatory properties in vitro and in vivo. Front Microbiol 2023; 14:1270974. [PMID: 38094624 PMCID: PMC10716304 DOI: 10.3389/fmicb.2023.1270974] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2023] [Accepted: 10/23/2023] [Indexed: 04/19/2024] Open
Abstract
INTRODUCTION The aim of this work was to characterize a new strain of Ligilactobacillus salivarius (CNCM I-4866) (CNCM I-4866) to address its potential as probiotic with a special focus on intestinal inflammation. Potential anti-inflammatory abilities of this strain were evaluated through in vivo and in vitro experiments. METHODS Firstly, the strain was tested in a murine acute inflammation colitis model induced by DNBS. In vitro characterization was then performed with diverse tests: modulation capability of intestinal permeability; study of the impact on immunity profile through cytokines dosage; capacity to inhibit pathogens and adhere to intestinal cells lines. Production of metabolites, antibiotic resistance and survival to gastro-intestinal tract conditions were also tested. RESULTS In vitro assay has shown a reduction of colonic damage and markers of inflammation after treatment with CNCM I-4866. Transcriptomic analysis performed on colons showed the capacity of the strain to down-regulate pro-inflammatory cytokines. L. salivarius CNCM I-4866 exerted anti-inflammatory profile by reducing IL-8 production by TNF-α stimulated cell and modulated cytokines profile on peripheral blood mononuclear cells (PBMC). It protected intestinal integrity by increasing trans-epithelial electrical resistance (TEER) on Caco-2 TNF-α inflamed cells. Additionally, L. salivarius CNCM I-4866 displayed inhibition capacity on several intestinal pathogens and adhered to eukaryotic cells. Regarding safety and technical concerns, CNCM I-4866 was highly resistant to 0.3% of bile salts and produced mainly L-lactate. Finally, strain genomic characterization allowed us to confirm safety aspect of our strain, with no antibiotic gene resistance found. DISCUSSION Taken together, these results indicate that L. salivarius CNCM I-4866 could be a good probiotic candidate for intestinal inflammation, especially with its steady anti-inflammatory profile.
Collapse
Affiliation(s)
| | | | | | | | | | | | - Rebeca Martin
- Micalis Institute, AgroParisTech, INRAE, Université Paris-Saclay, Jouy-en-Josas, France
| |
Collapse
|
15
|
Miao M, Li S, Yu Y, Li F. LysM-containing proteins function in the resistance of Litopenaeus vannamei against Vibrio parahaemolyticus infection. DEVELOPMENTAL AND COMPARATIVE IMMUNOLOGY 2023; 148:104900. [PMID: 37536402 DOI: 10.1016/j.dci.2023.104900] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/03/2023] [Revised: 07/27/2023] [Accepted: 07/30/2023] [Indexed: 08/05/2023]
Abstract
Lysin motif (LysM) is a functional domain that can bind to peptidoglycans, chitin and their derivatives. The LysM-containing proteins participate in multiple biological processes, such as the hydrolysis of bacterial cell walls and the perception of PAMPs in plants and high animals. In the present study, two genes encoding LysM-containing proteins, designated as LvLysM1 and LvLysM2, were identified in the Pacific white shrimp, Litopenaeus vannamei, and their functions during Vibrio infection were analyzed. The open-reading frame (ORF) of LvLysM1 was 795 bp, only encoding a LysM domain at the N-terminal region. The ORF of LvLysM2 was 834 bp, encoding a LysM domain at the central region and a transmembrane region at the C-terminal region. Both LvLysM1 and LvLysM2 were widely transcribed in all tested shrimp tissues. Enzyme-linked immunosorbent assay (ELISA) showed that the recombinant protein of LvLysM2 could bind to different bacterial polysaccharides, while LvLysM1 showed no direct binding activity. The transcripts of LvLysMs in gills increased significantly after infection with Vibrio parahaemolyticus. When LvLysM1 or LvLysM2 was knocked down by dsRNA, the mortality of shrimp was significantly increased after infection with Vibrio parahaemolyticus. Interestingly, some SNPs existed in these two genes were apparently correlated with the VpAHPND resistance of shrimp. These results suggested that LvLysM1 and LvLysM2 might contribute to the disease resistance of shrimp. The data provide new knowledge about the function of LysM-containing proteins in shrimp and potential genetic markers for disease resistance breeding.
Collapse
Affiliation(s)
- Miao Miao
- CAS and Shandong Province Key Laboratory of Experimental Marine Biology, Institute of Oceanology, Chinese Academy of Sciences, Qingdao, 266071, China; University of Chinese Academy of Sciences, Beijing, 100049, China
| | - Shihao Li
- CAS and Shandong Province Key Laboratory of Experimental Marine Biology, Institute of Oceanology, Chinese Academy of Sciences, Qingdao, 266071, China; Center for Ocean Mega-Science, Chinese Academy of Sciences, Qingdao, 266071, China.
| | - Yang Yu
- CAS and Shandong Province Key Laboratory of Experimental Marine Biology, Institute of Oceanology, Chinese Academy of Sciences, Qingdao, 266071, China; Center for Ocean Mega-Science, Chinese Academy of Sciences, Qingdao, 266071, China
| | - Fuhua Li
- CAS and Shandong Province Key Laboratory of Experimental Marine Biology, Institute of Oceanology, Chinese Academy of Sciences, Qingdao, 266071, China; Center for Ocean Mega-Science, Chinese Academy of Sciences, Qingdao, 266071, China; The Innovation of Seed Design, Chinese Academy of Sciences, Wuhan, 430072, China.
| |
Collapse
|
16
|
Huang Q, Zhang H, Zhang L, Xu B. Bacterial microbiota in different types of processed meat products: diversity, adaptation, and co-occurrence. Crit Rev Food Sci Nutr 2023; 65:287-302. [PMID: 37905560 DOI: 10.1080/10408398.2023.2272770] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/02/2023]
Abstract
As a double-edged sword, some bacterial microbes can improve the quality and shelf life of meat products, but others mainly responsible for deterioration of the safety and quality of meat products. This review aims to present a landscape of the bacterial microbiota in different types of processed meat products. After demonstrating a panoramic view of the bacterial genera in meat products, the diversity of bacterial microbiota was evaluated in two dimensions, namely different types of processed meat products and different meats. Then, the influence of environmental factors on bacterial communities was evaluated according to the storage temperature, packaging conditions, and sterilization methods. Furthermore, microbes are not independent. To explore interactions among those genera, co-occurrence patterns were examined. In these respects, this review highlighted the recent advances in fundamental principles that underlie the environmental adaption tricks and why some species tend to occur together frequently, such as metabolic cross-feeding, co-aggregate at microscale, and the intercellular signaling system. Further investigations are required to unveil the underlying molecular mechanisms that govern microbial community systems, ultimately contributing to developing new strategies to harness beneficial microorganisms and control harmful microorganisms.
Collapse
Affiliation(s)
- Qianli Huang
- Engineering Research Center of Bio-process, Ministry of Education, Hefei University of Technology, Hefei, China
- School of Food and Biological Engineering, Hefei University of Technology, Hefei, China
| | - Huijuan Zhang
- Engineering Research Center of Bio-process, Ministry of Education, Hefei University of Technology, Hefei, China
- School of Food and Biological Engineering, Hefei University of Technology, Hefei, China
| | - Li Zhang
- Engineering Research Center of Bio-process, Ministry of Education, Hefei University of Technology, Hefei, China
- School of Food and Biological Engineering, Hefei University of Technology, Hefei, China
| | - Baocai Xu
- Engineering Research Center of Bio-process, Ministry of Education, Hefei University of Technology, Hefei, China
- School of Food and Biological Engineering, Hefei University of Technology, Hefei, China
| |
Collapse
|
17
|
Wang Y, Xu X, Chen H, Yang F, Xu B, Wang K, Liu Q, Liang G, Zhang R, Jiao X, Zhang Y. Assessment of beneficial effects and identification of host adaptation-associated genes of Ligilactobacillus salivarius isolated from badgers. BMC Genomics 2023; 24:530. [PMID: 37679681 PMCID: PMC10483869 DOI: 10.1186/s12864-023-09623-8] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2023] [Accepted: 08/25/2023] [Indexed: 09/09/2023] Open
Abstract
BACKGROUND Ligilactobacillus salivarius has been frequently isolated from the gut microbiota of humans and domesticated animals and has been studied as a candidate probiotic. Badger (Meles meles) is known as a "generalist" species that consumes complex foods and exhibits tolerance and resistance to certain pathogens, which can be partly attributed to the beneficial microbes such as L. salivarius in the gut microbiota. However, our understanding of the beneficial traits and genomic features of badger-originated L. salivarius remains elusive. RESULTS In this study, nine L. salivarius strains were isolated from wild badgers' feces, one of which exhibited good probiotic properties. Complete genomes of the nine L. salivarius strains were generated, and comparative genomic analysis was performed with the publicly available complete genomes of L. salivarius obtained from humans and domesticated animals. The strains originating from badgers harbored a larger genome, a higher number of protein-coding sequences, and functionally annotated genes than those originating from humans and chickens. The pan-genome phylogenetic tree demonstrated that the strains originating from badgers formed a separate clade, and totally 412 gene families (12.6% of the total gene families in the pan-genome) were identified as genes gained by the last common ancestor of the badger group. The badger group harbored significantly more gene families responsible for the degradation of complex carbohydrate substrates and production of polysaccharides than strains from other hosts; many of these were acquired by gene gain events. CONCLUSIONS A candidate probiotic and nine L. salivarius complete genomes were obtained from the badgers' gut microbiome, and several beneficial genes were identified to be specifically present in the badger-originated strains that were gained in the evolution. Our study provides novel insights into the adaptation of L. salivarius to the intestinal habitat of wild badgers and provides valuable strain and genome resources for the development of L. salivarius as a probiotic.
Collapse
Affiliation(s)
- Yu Wang
- Jiangsu Co-Innovation Center for Prevention and Control of Important Animal Infectious Diseases and Zoonoses, Yangzhou University, Yangzhou, 225009, China
- Jiangsu Key Laboratory of Zoonosis, Yangzhou University, Yangzhou, 225009, China
- Joint International Research Laboratory of Agriculture and Agri-product Safety of the Ministry of Education, Yangzhou University, Yangzhou, 225009, China
- Key Laboratory of Prevention and Control of Biological Hazard Factors (Animal Origin) for Agrifood Safety and Quality, Ministry of Agriculture of China, Yangzhou University, Yangzhou, 225009, China
| | - Xiaomeng Xu
- Jiangsu Co-Innovation Center for Prevention and Control of Important Animal Infectious Diseases and Zoonoses, Yangzhou University, Yangzhou, 225009, China
- Jiangsu Key Laboratory of Zoonosis, Yangzhou University, Yangzhou, 225009, China
- Joint International Research Laboratory of Agriculture and Agri-product Safety of the Ministry of Education, Yangzhou University, Yangzhou, 225009, China
- Key Laboratory of Prevention and Control of Biological Hazard Factors (Animal Origin) for Agrifood Safety and Quality, Ministry of Agriculture of China, Yangzhou University, Yangzhou, 225009, China
| | - Huan Chen
- Jiangsu Co-Innovation Center for Prevention and Control of Important Animal Infectious Diseases and Zoonoses, Yangzhou University, Yangzhou, 225009, China
- Jiangsu Key Laboratory of Zoonosis, Yangzhou University, Yangzhou, 225009, China
- Joint International Research Laboratory of Agriculture and Agri-product Safety of the Ministry of Education, Yangzhou University, Yangzhou, 225009, China
- Key Laboratory of Prevention and Control of Biological Hazard Factors (Animal Origin) for Agrifood Safety and Quality, Ministry of Agriculture of China, Yangzhou University, Yangzhou, 225009, China
| | - Fang Yang
- Jiangsu Co-Innovation Center for Prevention and Control of Important Animal Infectious Diseases and Zoonoses, Yangzhou University, Yangzhou, 225009, China
- Jiangsu Key Laboratory of Zoonosis, Yangzhou University, Yangzhou, 225009, China
- Joint International Research Laboratory of Agriculture and Agri-product Safety of the Ministry of Education, Yangzhou University, Yangzhou, 225009, China
- Key Laboratory of Prevention and Control of Biological Hazard Factors (Animal Origin) for Agrifood Safety and Quality, Ministry of Agriculture of China, Yangzhou University, Yangzhou, 225009, China
| | - Bo Xu
- Jiangsu Co-Innovation Center for Prevention and Control of Important Animal Infectious Diseases and Zoonoses, Yangzhou University, Yangzhou, 225009, China
- Jiangsu Key Laboratory of Zoonosis, Yangzhou University, Yangzhou, 225009, China
- Joint International Research Laboratory of Agriculture and Agri-product Safety of the Ministry of Education, Yangzhou University, Yangzhou, 225009, China
- Key Laboratory of Prevention and Control of Biological Hazard Factors (Animal Origin) for Agrifood Safety and Quality, Ministry of Agriculture of China, Yangzhou University, Yangzhou, 225009, China
| | - Kun Wang
- Jiangsu Co-Innovation Center for Prevention and Control of Important Animal Infectious Diseases and Zoonoses, Yangzhou University, Yangzhou, 225009, China
- Jiangsu Key Laboratory of Zoonosis, Yangzhou University, Yangzhou, 225009, China
- Joint International Research Laboratory of Agriculture and Agri-product Safety of the Ministry of Education, Yangzhou University, Yangzhou, 225009, China
- Key Laboratory of Prevention and Control of Biological Hazard Factors (Animal Origin) for Agrifood Safety and Quality, Ministry of Agriculture of China, Yangzhou University, Yangzhou, 225009, China
| | - Qianwen Liu
- Jiangsu Co-Innovation Center for Prevention and Control of Important Animal Infectious Diseases and Zoonoses, Yangzhou University, Yangzhou, 225009, China
- Jiangsu Key Laboratory of Zoonosis, Yangzhou University, Yangzhou, 225009, China
- Joint International Research Laboratory of Agriculture and Agri-product Safety of the Ministry of Education, Yangzhou University, Yangzhou, 225009, China
- Key Laboratory of Prevention and Control of Biological Hazard Factors (Animal Origin) for Agrifood Safety and Quality, Ministry of Agriculture of China, Yangzhou University, Yangzhou, 225009, China
| | - Guixin Liang
- Jiangsu Co-Innovation Center for Prevention and Control of Important Animal Infectious Diseases and Zoonoses, Yangzhou University, Yangzhou, 225009, China
- Jiangsu Key Laboratory of Zoonosis, Yangzhou University, Yangzhou, 225009, China
- Joint International Research Laboratory of Agriculture and Agri-product Safety of the Ministry of Education, Yangzhou University, Yangzhou, 225009, China
- Key Laboratory of Prevention and Control of Biological Hazard Factors (Animal Origin) for Agrifood Safety and Quality, Ministry of Agriculture of China, Yangzhou University, Yangzhou, 225009, China
| | - Ruiqi Zhang
- Jiangsu Co-Innovation Center for Prevention and Control of Important Animal Infectious Diseases and Zoonoses, Yangzhou University, Yangzhou, 225009, China
- Jiangsu Key Laboratory of Zoonosis, Yangzhou University, Yangzhou, 225009, China
- Joint International Research Laboratory of Agriculture and Agri-product Safety of the Ministry of Education, Yangzhou University, Yangzhou, 225009, China
- Key Laboratory of Prevention and Control of Biological Hazard Factors (Animal Origin) for Agrifood Safety and Quality, Ministry of Agriculture of China, Yangzhou University, Yangzhou, 225009, China
| | - Xin'an Jiao
- Jiangsu Co-Innovation Center for Prevention and Control of Important Animal Infectious Diseases and Zoonoses, Yangzhou University, Yangzhou, 225009, China.
- Jiangsu Key Laboratory of Zoonosis, Yangzhou University, Yangzhou, 225009, China.
- Joint International Research Laboratory of Agriculture and Agri-product Safety of the Ministry of Education, Yangzhou University, Yangzhou, 225009, China.
- Key Laboratory of Prevention and Control of Biological Hazard Factors (Animal Origin) for Agrifood Safety and Quality, Ministry of Agriculture of China, Yangzhou University, Yangzhou, 225009, China.
| | - Yunzeng Zhang
- Jiangsu Co-Innovation Center for Prevention and Control of Important Animal Infectious Diseases and Zoonoses, Yangzhou University, Yangzhou, 225009, China.
- Jiangsu Key Laboratory of Zoonosis, Yangzhou University, Yangzhou, 225009, China.
- Joint International Research Laboratory of Agriculture and Agri-product Safety of the Ministry of Education, Yangzhou University, Yangzhou, 225009, China.
- Key Laboratory of Prevention and Control of Biological Hazard Factors (Animal Origin) for Agrifood Safety and Quality, Ministry of Agriculture of China, Yangzhou University, Yangzhou, 225009, China.
| |
Collapse
|
18
|
Sodré MTC, Ferraz FA, Alencar AKV, Silva KF, Silva DHDS, Silva LDS, Araújo Carneiro JSDS, Monteiro CA, Silva LCN, Monteiro ADS. The Potential of Lactiplantibacillus plantarum ATCC 14917 in the Development of Alginate-Based Gel Formulations with Anti- Staphylococcus aureus Properties. Pharmaceuticals (Basel) 2023; 16:1112. [PMID: 37631027 PMCID: PMC10458396 DOI: 10.3390/ph16081112] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2023] [Revised: 06/04/2023] [Accepted: 06/15/2023] [Indexed: 08/27/2023] Open
Abstract
This study aimed to evaluate the potential of lactic acid bacteria (LAB) in developing alginate-based gel formulations to inhibit Staphylococcus aureus. Initially, the antagonistic actions of three lactic acid bacteria (LAB) (Lacticaseibacillus rhamnosus ATCC 10863, Lactiplantibacillus plantarum ATCC 14917, Limosilactobacillus fermentum ATCC 23271) were evaluated against S. aureus ATCC 25923. All tested LAB inhibited S. aureus, but the highest activity was observed for L. plantarum ATCC 14917 (p < 0.05). The antimicrobial effects of L. plantarum ATCC 14917 cell suspensions, sonicate cells extract, and cell-free supernatants (pH 5 or 7) were analyzed using a broth-based assay. The cell suspensions inhibited S. aureus at concentrations ≥ 10%, and these effects were confirmed by a time-kill assay. Alginate-based gels were formulated with cell suspensions, sonicate cells extract, and cell-free supernatant (pH 5). These formulations inhibited S. aureus growth. Based on the results, the alginate gel with cell suspensions at 10% was selected for further characterization. L. plantarum ATCC 14917 survived in the alginate-based gel, especially when stored at 5 °C. At this temperature, the L. plantarum-containing alginate gel was stable, and it was in compliance with microbiological standards. These findings suggest it can be a promising agent for the topical treatment of infections induced by S. aureus.
Collapse
Affiliation(s)
| | | | | | - Karinny Farias Silva
- Laboratory of Applied Microbiology, CEUMA University, São Luís 65075-120, MA, Brazil
| | | | | | | | - Cristina Andrade Monteiro
- Laboratory of Microbiology Research, Federal Institute of Education, Science and Technology of Maranhão (IFMA), São Luís 65030-005, MA, Brazil
| | | | | |
Collapse
|
19
|
Neidhöfer C, Rathore K, Parčina M, Sieber MA. ESKAPEE Pathogen Biofilm Control on Surfaces with Probiotic Lactobacillaceae and Bacillus species. Antibiotics (Basel) 2023; 12:871. [PMID: 37237774 PMCID: PMC10215598 DOI: 10.3390/antibiotics12050871] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2023] [Revised: 04/21/2023] [Accepted: 05/05/2023] [Indexed: 05/28/2023] Open
Abstract
Combatting the rapidly growing threat of antimicrobial resistance and reducing prevalence and transmission of ESKAPEE pathogens in healthcare settings requires innovative strategies, one of which is displacing these pathogens using beneficial microorganisms. Our review comprehensively examines the evidence of probiotic bacteria displacing ESKAPEE pathogens, with a focus on inanimate surfaces. A systematic search was conducted using the PubMed and Web of Science databases on 21 December 2021, and 143 studies were identified examining the effects of Lactobacillaceae and Bacillus spp. cells and products on the growth, colonization, and survival of ESKAPEE pathogens. While the diversity of study methods limits evidence analysis, results presented by narrative synthesis demonstrate that several species have the potential as cells or their products or supernatants to displace nosocomial infection-causing organisms in a variety of in vitro and in vivo settings. Our review aims to aid the development of new promising approaches to control pathogen biofilms in medical settings by informing researchers and policymakers about the potential of probiotics to combat nosocomial infections. More targeted studies are needed to assess safety and efficacy of different probiotic formulations, followed by large-scale studies to assess utility in infection control and medical practice.
Collapse
Affiliation(s)
- Claudio Neidhöfer
- Institute of Medical Microbiology, Immunology and Parasitology, University Hospital Bonn, Venusberg-Campus 1, 53127 Bonn, Germany
| | - Kamni Rathore
- Institute of Medical Microbiology, Immunology and Parasitology, University Hospital Bonn, Venusberg-Campus 1, 53127 Bonn, Germany
- Institute for Functional Gene Analytics, Bonn-Rhein-Sieg University of Applied Sciences, 53757 Sankt Augustin, Germany
| | - Marijo Parčina
- Institute of Medical Microbiology, Immunology and Parasitology, University Hospital Bonn, Venusberg-Campus 1, 53127 Bonn, Germany
| | - Martin A. Sieber
- Institute for Functional Gene Analytics, Bonn-Rhein-Sieg University of Applied Sciences, 53757 Sankt Augustin, Germany
| |
Collapse
|
20
|
Dash J, Sethi M, Deb S, Parida D, Kar S, Mahapatra S, Minz AP, Pradhan B, Prasad P, Senapati S. Biochemical, functional and genomic characterization of a new probiotic Ligilactobacillus salivarius F14 from the gut of tribes of Odisha. World J Microbiol Biotechnol 2023; 39:171. [PMID: 37101059 DOI: 10.1007/s11274-023-03626-z] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2022] [Accepted: 04/18/2023] [Indexed: 04/28/2023]
Abstract
Characterization of new potential probiotics is desirable in the field of research on probiotics for their extensive use in health and disease. Tribes could be an unusual source of probiotics due to their unique food habits and least dependence on medications and consumption of antibiotics. The aim of the present study is to isolate lactic acid bacteria from tribal fecal samples of Odisha, India, and characterize their genetic and probiotic attributes. In this context one of the catalase-negative and Gram-positive isolates, identified using 16S rRNA sequencing as Ligilactobacillus salivarius, was characterized in vitro for its acid and bile tolerance, cell adhesion and antimicrobial properties. The whole genome sequence was obtained and analyzed for strain level identification, presence of genomic determinants for probiotic-specific features, and safety. Genes responsible for its antimicrobial and immunomodulatory functions were detected. The secreted metabolites were analyzed using high resolution mass spectroscopy; the results indicated that the antimicrobial potential could be due to the presence of pyroglutamic acid, propionic acid, lactic acid, 2-hydroxyisocaproic acid, homoserine, and glutathione, and the immuno-modulating activity, contributed by the presence of short chain fatty acids such as acetate, propionate, and butyrate. So, to conclude we have successfully characterized a Ligilactobacillus salivarius species with potential antimicrobial and immunomodulatory ability. The health-promoting effects of this probiotic strain and/or its derivatives will be investigated in future.
Collapse
Affiliation(s)
- Jayalaxmi Dash
- Institute of Life Sciences, Nalco Square, Bhubaneswar, Odisha, 751023, India
| | - Manisha Sethi
- Institute of Life Sciences, Nalco Square, Bhubaneswar, Odisha, 751023, India
- Regional Centre for Biotechnology, Faridabad, Haryana, India
| | - Sushanta Deb
- Institute of Life Sciences, Nalco Square, Bhubaneswar, Odisha, 751023, India
- Department of Microbiology, AIIMS, New Delhi, India
| | - Deepti Parida
- Institute of Life Sciences, Nalco Square, Bhubaneswar, Odisha, 751023, India
- Regional Centre for Biotechnology, Faridabad, Haryana, India
| | - Salona Kar
- Institute of Life Sciences, Nalco Square, Bhubaneswar, Odisha, 751023, India
- Regional Centre for Biotechnology, Faridabad, Haryana, India
| | - Soumendu Mahapatra
- Institute of Life Sciences, Nalco Square, Bhubaneswar, Odisha, 751023, India
| | - Aliva P Minz
- Institute of Life Sciences, Nalco Square, Bhubaneswar, Odisha, 751023, India
- Regional Centre for Biotechnology, Faridabad, Haryana, India
| | - Biswaranjan Pradhan
- S. K. Dash Center of Excellence of Biosciences and Engineering & Technology (SKBET), Indian Institute of Technology Bhubaneswar, Bhubaneswar, Odisha, India
| | - Punit Prasad
- Institute of Life Sciences, Nalco Square, Bhubaneswar, Odisha, 751023, India
| | | |
Collapse
|
21
|
Yao Y, Zhu HY, Zeng M, Liu Z. Immunological mechanisms and treatable traits of chronic rhinosinusitis in Asia: A narrative review. Clin Otolaryngol 2023; 48:363-370. [PMID: 36317525 DOI: 10.1111/coa.14001] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2022] [Revised: 09/23/2022] [Accepted: 10/16/2022] [Indexed: 11/15/2022]
Abstract
OBJECTIVES To review the current literature on immunological mechanisms and treatable traits of chronic rhinosinusitis (CRS) in Asia. DESIGN This is a narrative review of published data on the immunological mechanisms and treatable traits of CRS in Asia. Published English literature on CRS in Asian and Western countries was reviewed. Where available, the data extracted included epidemiology, immunology, bacterium, phenotype, endotype and treatment. RESULTS AND CONCLUSION CRS is a heterogeneous disease characterised by persistent locoregional mucosal inflammation of the paranasal sinuses. The inflammatory signatures of CRS vary across patients with distinct racial and ethnic backgrounds and geographic areas. Compared to CRS patients in Western countries, Asian CRS patients display less eosinophilic and Type 2 inflammation, which is associated with lower asthma and allergic rhinitis comorbidities. In contrast, Asian patients with CRS have more prominent non-eosinophilic inflammation than those in Western countries. In addition, Asian CRS patients may have different bacterial colonisation than patients in Western countries. Our review suggests that the distinct immunological mechanisms between Asian and Western CRS patients may influence the clinical phenotype, responses to treatment and outcomes. The treatable trait is a new strategy and therapeutic target identified by phenotype or endotype and has been proposed as a new paradigm for the management of diseases. Improved understanding of CRS phenotypic and endotypic heterogeneity and incorporation of treatable traits into clinical care pathways may facilitate more effective selections of therapeutic interventions, including surgery and biologics.
Collapse
Affiliation(s)
- Yin Yao
- Department of Otolaryngology-Head and Neck Surgery, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, People's Republic of China.,Hubei Clinical Research Center for Nasal Inflammatory Diseases, Wuhan, Hubei, People's Republic of China
| | - Hong-Yu Zhu
- Department of Otolaryngology-Head and Neck Surgery, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, People's Republic of China
| | - Ming Zeng
- Department of Otolaryngology-Head and Neck Surgery, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, People's Republic of China.,Hubei Clinical Research Center for Nasal Inflammatory Diseases, Wuhan, Hubei, People's Republic of China
| | - Zheng Liu
- Department of Otolaryngology-Head and Neck Surgery, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, People's Republic of China.,Hubei Clinical Research Center for Nasal Inflammatory Diseases, Wuhan, Hubei, People's Republic of China
| |
Collapse
|
22
|
Jalalifar S, Mirzaei R, Motallebirad T, Razavi S, Talebi M. The Emerging Role of Probiotics and their Derivatives against Biofilm-Producing MRSA: A Scoping Review. BIOMED RESEARCH INTERNATIONAL 2022; 2022:4959487. [PMID: 36605101 PMCID: PMC9810406 DOI: 10.1155/2022/4959487] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/13/2022] [Revised: 11/20/2022] [Accepted: 12/01/2022] [Indexed: 12/29/2022]
Abstract
BACKGROUND Methicillin-resistant Staphylococcus aureus (MRSA) is one of the main bacterial pathogens causing chronic infections, mainly because of its capacity to produce biofilm. Biofilm production is one of the underlying strategies for antibacterial drug resistance. Accordingly, preventing and attenuating biofilm production has become an emerging approach to controlling persistent infections. Therefore, this scoping review is aimed at surveying the published literature describing the usage of probiotics and their derivatives against biofilm-producing MRSA. METHODS Updated literature searches were conducted across seven electronic databases including Web of Science, PubMed, Scopus, Cochrane Library, ProQuest, Embase, and Google Scholar to identify all original published articles about probiotics against MRSA. In this regard, studies were summarized and analyzed in the present review. RESULTS In the reviewed studies, various microorganisms and compounds were used as probiotics as follows: Lactobacillus species (8 studies), Enterococcus species (4 studies), Bacillus species (2 studies), Streptomyces species (2 studies), Saccharomyces cerevisiae (1 study), Corynebacterium accolens (1 study), and Lactococcus lactis derived Nisin (3 studies). Based on our comprehensive search, 21 studies with eligibility criteria were included in the present review including 12 studies on clinical strains, 6 studies on ATCC, 2 studies simultaneously on clinical and standard strains, and finally 1 study on food sample. CONCLUSIONS Our study showed that there was an increasing trend in the number of publications reporting probiotics against biofilm-producing MRSA. The results of this scoping review could use to guide the undertaking of the subsequent systematic reviews. In summary, probiotics with antimicrobial and antibiofilm properties can use as an embedded agent in food products or as a biopharmaceutical in the prevention and treatment of MRSA infections.
Collapse
Affiliation(s)
- Saba Jalalifar
- Microbial Biotechnology Research Center, Iran University of Medical Sciences, Tehran, Iran
- Department of Microbiology, School of Medicine, Iran University of Medical Sciences, Tehran, Iran
| | - Rasoul Mirzaei
- Venom and Biotherapeutics Molecules Lab, Medical Biotechnology Department, Biotechnology Research Center, Pasteur Institute of Iran, Tehran, Iran
| | - Tahereh Motallebirad
- Department of Microbiology, School of Medicine, Isfahan University of Medical Sciences, Isfahan, Iran
| | - Shabnam Razavi
- Microbial Biotechnology Research Center, Iran University of Medical Sciences, Tehran, Iran
- Department of Microbiology, School of Medicine, Iran University of Medical Sciences, Tehran, Iran
| | - Malihe Talebi
- Microbial Biotechnology Research Center, Iran University of Medical Sciences, Tehran, Iran
- Department of Microbiology, School of Medicine, Iran University of Medical Sciences, Tehran, Iran
| |
Collapse
|
23
|
Hossain TJ, Mozumder HA, Ali F, Akther K. Inhibition of Pathogenic Microbes by the Lactic Acid Bacteria Limosilactobacillus Fermentum Strain LAB-1 and Levilactobacillus Brevis Strain LAB-5 Isolated from the Dairy Beverage Borhani. CURRENT RESEARCH IN NUTRITION AND FOOD SCIENCE JOURNAL 2022. [DOI: 10.12944/crnfsj.10.3.10] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/29/2022]
Abstract
Lactic acid bacteria (LAB) with prominent antimicrobial effects against pathogens have been reported in several milk-based and plant-based foods. Borhani is a popular beverage prepared from the ingredients of both dairy and plant origins and is believed to be highly beneficial for health. Herein, we report the pathogen-inhibitory activity of two borhani-associated lactic acid bacteria (LAB), Limosilactobacillus fermentum strain LAB-1 and Levilactobacillus brevis strain LAB-5. Their antimicrobial activity was primarily assessed using the cell free supernatant (CFS) by agar diffusion technique in which both strains showed strong antimicrobial effects against several pathogenic and spoilage microorganisms including Acinetobacter baumannii, Bacillus cereus, Bacillus subtilis, Escherichia coli, Klebsiella pneumonia, Pseudomonas aeruginosa, Salmonella abony, Salmonella typhi, Shigella flexneri, and Staphylococcus aureus. The L. fermentum strain with its ability to inhibit all the target pathogens appeared to be more effective with larger inhibition-zone formation as compared to the L. brevis strain which also successfully inhibited all pathogens but had relatively little effects on A. baumannii. The extent of their inhibitory effect was further assessed by co-culture inhibition assay in which growth of the test microbes was monitored for 24 hours in presence of the CFS. The CFS of both lactic acid bacteria could effectively inhibit growth of the pathogenic microbes for a significant period of time. While the L. fermentum strain could almost completely stop growth of all test organisms, the L. brevis strain was particularly effective against Shigella flexneri and the Salmonella species. Our study, therefore, suggests the presence of beneficial lactic acid bacteria in borhani which can be of important use as antimicrobial agents in functional foods and therapeutics to help acquire protection against drug resistant pathogens.
Collapse
Affiliation(s)
- Tanim Jabid Hossain
- 1Department of Biochemistry and Molecular Biology, University of Chittagong, Chattogram, Bangladesh
| | | | - Ferdausi Ali
- 3Department of Microbiology, University of Chittagong, Chattogram Bangladesh
| | - Khadiza Akther
- 2Biochemistry and Pathogenesis of Microbes Research Group, Chattogram, Bangladesh
| |
Collapse
|
24
|
Lactiplantibacillus plantarum KAU007 Extract Modulates Critical Virulence Attributes and Biofilm Formation in Sinusitis Causing Streptococcus pyogenes. Pharmaceutics 2022; 14:pharmaceutics14122702. [PMID: 36559194 PMCID: PMC9780990 DOI: 10.3390/pharmaceutics14122702] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2022] [Revised: 11/25/2022] [Accepted: 11/29/2022] [Indexed: 12/09/2022] Open
Abstract
Streptococcus pyogenes is one of the most common bacteria causing sinusitis in children and adult patients. Probiotics are known to cause antagonistic effects on S. pyogenes growth and biofilm formation. In the present study, we demonstrated the anti-biofilm and anti-virulence properties of Lactiplantibacillus plantarum KAU007 against S. pyogenes ATCC 8668. The antibacterial potential of L. plantarum KAU007 metabolite extract (LME) purified from the cell-free supernatant of L. plantarum KAU007 was evaluated in terms of minimum inhibitory concentrations (MIC) and minimum bactericidal concentrations (MBC). LME was further analyzed for its anti-biofilm potential using crystal violet assay and microscopic examination. Furthermore, the effect of LME was tested on the important virulence attributes of S. pyogenes, such as secreted protease production, hemolysis, extracellular polymeric substance production, and cell surface hydrophobicity. Additionally, the impact of LME on the expression of genes associated with biofilm formation and virulence attributes was analyzed using qPCR. The results revealed that LME significantly inhibited the growth and survival of S. pyogenes at a low concentration (MIC, 9.76 µg/mL; MBC, 39.06 µg/mL). Furthermore, LME inhibited biofilm formation and mitigated the production of extracellular polymeric substance at a concentration of 4.88 μg/mL in S. pyogenes. The results obtained from qPCR and biochemical assays advocated that LME suppresses the expression of various critical virulence-associated genes, which correspondingly affect various pathogenicity markers and were responsible for the impairment of virulence and biofilm formation in S. pyogenes. The non-hemolytic nature of LME and its anti-biofilm and anti-virulence properties against S. pyogenes invoke further investigation to study the role of LME as an antibacterial agent to combat streptococcal infections.
Collapse
|
25
|
Naskar A, Cho H, Kim KS. A Nanocomposite with Extracellular Vesicles from Lactobacillus paracasei as a Bioinspired Nanoantibiotic Targeting Staphylococcus aureus. Pharmaceutics 2022; 14:2273. [PMID: 36365092 PMCID: PMC9692410 DOI: 10.3390/pharmaceutics14112273] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2022] [Revised: 10/14/2022] [Accepted: 10/21/2022] [Indexed: 08/29/2023] Open
Abstract
The utilization of biomimetic materials that merge functional nanoparticles (NPs) with a cell-derived nanosized membrane is a state-of-the-art approach to harnessing cellular properties for biomedical applications. However, the development of biocompatible and species-selective biomimetic agents against hazardous pathogens threatening human health is still in its early stages. Herein, we report the synthesis and functional analysis of a novel nanoplatform in which a PEGylated MoS2-ZnO (MZ) nanocomposite was cloaked with a generally regarded as safe (GRAS)-grade Lactobacillus paracasei-derived extracellular vesicle (LPEV) for MZ-LPEV nanocomposite and evaluated its activity against Staphylococcus aureus. The MZ nanocomposite was characterized via X-ray diffraction, transmission electron microscopy, and X-ray photoelectron spectroscopy. The coating of MZ with LPEV was confirmed through nanoparticle tracking analysis and zeta potential measurements. MZ-LPEV exhibited 5- to 20-fold higher antibacterial activity than that of ZO NPs and MZ nanocomposite against S. aureus. Reactive oxygen species (ROS) production and bacterial membrane disruption were confirmed as antibacterial mechanisms of MZ-LPEV. Finally, MZ-LPEV exhibited enhanced biocompatibility and selectivity for S. aureus. All our results showed that LPEV could be utilized for developing synergistic nanoantibiotics against S. aureus.
Collapse
Affiliation(s)
| | | | - Kwang-sun Kim
- Department of Chemistry and Chemistry Institute for Functional Materials, Pusan National University, Busan 46241, Korea
| |
Collapse
|
26
|
Jara J, Jurado R, Almendro-Vedia VG, López-Montero I, Fernández L, Rodríguez JM, Orgaz B. Interspecies relationships between nosocomial pathogens associated to preterm infants and lactic acid bacteria in dual-species biofilms. Front Cell Infect Microbiol 2022; 12:1038253. [PMID: 36325465 PMCID: PMC9618709 DOI: 10.3389/fcimb.2022.1038253] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2022] [Accepted: 09/26/2022] [Indexed: 12/08/2023] Open
Abstract
The nasogastric enteral feeding tubes (NEFTs) used to feed preterm infants are commonly colonized by bacteria with the ability to form complex biofilms in their inner surfaces. Among them, staphylococci (mainly Staphylococcus epidermidis and Staphylococcus aureus) and some species belonging to the Family Enterobacteriaceae are of special concern since they can cause nosocomial infections in this population. NETF-associated biofilms can also include lactic acid bacteria (LAB), with the ability to compete with pathogenic species for nutrients and space. Ecological interactions among the main colonizers of these devices have not been explored yet; however, such approach could guide future strategies involving the pre-coating of the inner surfaces of NEFTs with well adapted LAB strains in order to reduce the rates of nosocomial infections in neonatal intensive care units (NICUs). In this context, this work implied the formation of dual-species biofilms involving one LAB strain (either Ligilactobacillus salivarius 20SNG2 or Limosilactobacillus reuteri 7SNG3) and one nosocomial strain (either Klebsiella pneumoniae 9SNG3, Serratia marcescens 10SNG3, Staphylococcus aureus 45SNG3 or Staphylococcus epidermidis 46SNG3). The six strains used in this study had been isolated from the inner surface of NEFTs. Changes in adhesion ability of the pathogens were characterized using a culturomic approach. Species interactions and structural changes of the resulting biofilms were analyzed using scanning electron microscopy (SEM) and confocal laser scanning microscopy (CLSM). No aggregation was observed in dual-species biofilms between any of the two LAB strains and either K. pneumoniae 9SNG3 or S. marcescens 10SNG3. In addition, biofilm thickness and volume were reduced, suggesting that both LAB strains can control the capacity to form biofilms of these enterobacteria. In contrast, a positive ecological relationship was observed in the combination L. reuteri 7SNG3-S. aureus 45SNG3. This relationship was accompanied by a stimulation of S. aureus matrix production when compared with its respective monospecies biofilm. The knowledge provided by this study may guide the selection of potentially probiotic strains that share the same niche with nosocomial pathogens, enabling the establishment of a healthier microbial community inside NEFTs.
Collapse
Affiliation(s)
- Josué Jara
- Departamento de Nutrición y Ciencia de los Alimentos, Facultad de Veterinaria, Universidad Complutense de Madrid, Madrid, Spain
| | - Rubén Jurado
- Departamento de Farmacia Galénica y Tecnología de los Alimentos, Facultad de Veterinaria, Universidad Complutense de Madrid, Madrid, Spain
| | - Víctor G. Almendro-Vedia
- Departamento de Farmacia Galénica y Tecnología de los Alimentos, Facultad de Veterinaria, Universidad Complutense de Madrid, Madrid, Spain
| | - Iván López-Montero
- Departamento de Química Física, Facultad de Químicas, Universidad Complutense de Madrid, Madrid, Spain
- Instituto de Investigación Biomédica Hospital Doce de Octubre (imas12), Madrid, Spain
- Instituto Pluridisciplinar, Universidad Complutense de Madrid, Madrid, Spain
| | - Leonides Fernández
- Departamento de Farmacia Galénica y Tecnología de los Alimentos, Facultad de Veterinaria, Universidad Complutense de Madrid, Madrid, Spain
| | - Juan Miguel Rodríguez
- Departamento de Nutrición y Ciencia de los Alimentos, Facultad de Veterinaria, Universidad Complutense de Madrid, Madrid, Spain
| | - Belén Orgaz
- Departamento de Farmacia Galénica y Tecnología de los Alimentos, Facultad de Veterinaria, Universidad Complutense de Madrid, Madrid, Spain
| |
Collapse
|
27
|
Wan Z, Zhang X, Jia X, Qin Y, Sun N, Xin J, Zeng Y, Jing B, Fang J, Pan K, Zeng D, Bai Y, Wang H, Ma H, Ni X. Lactobacillus johnsonii YH1136 plays a protective role against endogenous pathogenic bacteria induced intestinal dysfunction by reconstructing gut microbiota in mice exposed at high altitude. Front Immunol 2022; 13:1007737. [PMID: 36304467 PMCID: PMC9592553 DOI: 10.3389/fimmu.2022.1007737] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2022] [Accepted: 09/06/2022] [Indexed: 11/13/2022] Open
Abstract
Background Intestinal microbiota plays an important role in maintaining the microecological balance of the gastrointestinal tract in various animals. Disturbances in the intestinal microbiota may lead to the proliferation of potentially pathogenic bacteria that become the dominant species, leading to intestinal immune disorders, intestinal inflammation, and other intestinal diseases. Numerous studies have been confirmed that high-altitude exposure affects the normal function of the intestine and the composition of the intestinal microbiota. However, it is still necessary to reveal the changes in intestinal microbiota in high-altitude exposure environments, and clarify the relationship between the proliferation of potentially pathogenic bacteria and intestinal injury in this environment. In addition, explored probiotics that may have preventive effects against intestinal diseases. Methods and results C57BL/6 mice were randomly divided into three groups, a high-altitude group (HA), control group (C), and high-altitude probiotic group (HAP). The HA and HAP groups were subjected to hypoxia modeling for 14 days in a low-pressure oxygen chamber with daily gavage of 0.2 mL of normal saline (HA) and Lactobacillus johnsonii YH1136 bacterial fluid (HAP), while the control group was fed normally. L. johnsonii YH1136 was isolated from feces of a healthy Tibetan girl in Baingoin county, the Nagqu region of the Tibet Autonomous Region, at an altitude of 5000 meters. Our observations revealed that gavage of YH1136 was effective in improving the damage to the intestinal barrier caused by high-altitude exposure to hypoxic environments and helped to reduce the likelihood of pathogenic bacteria infection through the intestinal barrier. It also positively regulates the intestinal microbiota to the extent of Lactobacillus being the dominant microbiome and reducing the number of pathogenic bacteria. By analyzing the expression profile of ileal microRNAs and correlation analysis with intestinal microbiota, we found that Staphylococcus and Corynebacterium1 cooperated with miR-196a-1-3p and miR-3060-3p, respectively, to play a regulatory role in the process of high-altitude hypoxia-induced intestinal injury. Conclusion These findings revealed the beneficial effect of L. johnsonii YH1136 in preventing potential endogenous pathogenic bacteria-induced intestinal dysfunction in high-altitude environments. The mechanism may be related to the regulation of intestinal injury from the perspective of the gut microbiota as well as miRNAs.
Collapse
Affiliation(s)
- Zhiqiang Wan
- Animal Microecology Institute, College of Veterinary, Sichuan Agricultural University, Chengdu, China
| | - Xufei Zhang
- Plateau Brain Science Research Center, Tibet University, Lhasa, China
| | - Xianhao Jia
- Plateau Brain Science Research Center, Tibet University, Lhasa, China
| | - Yuhua Qin
- College of Basic Medical Sciences, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Ning Sun
- Animal Microecology Institute, College of Veterinary, Sichuan Agricultural University, Chengdu, China
| | - Jinge Xin
- Guangdong Provincial Key Laboratory of Gastroenterology, Department of Gastroenterology, Institute of Gastroenterology of Guangdong Province, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Yan Zeng
- Animal Microecology Institute, College of Veterinary, Sichuan Agricultural University, Chengdu, China
| | - Bo Jing
- Animal Microecology Institute, College of Veterinary, Sichuan Agricultural University, Chengdu, China
| | - Jing Fang
- Animal Microecology Institute, College of Veterinary, Sichuan Agricultural University, Chengdu, China
| | - Kangcheng Pan
- Animal Microecology Institute, College of Veterinary, Sichuan Agricultural University, Chengdu, China
| | - Dong Zeng
- Animal Microecology Institute, College of Veterinary, Sichuan Agricultural University, Chengdu, China
| | - Yang Bai
- Guangdong Provincial Key Laboratory of Gastroenterology, Department of Gastroenterology, Institute of Gastroenterology of Guangdong Province, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Hesong Wang
- Guangzhou Beneco Biotechnology Co. Ltd., Guangzhou, China
| | - Hailin Ma
- Plateau Brain Science Research Center, Tibet University, Lhasa, China
| | - Xueqin Ni
- Animal Microecology Institute, College of Veterinary, Sichuan Agricultural University, Chengdu, China
| |
Collapse
|
28
|
Antimicrobial activity against Staphylococcus aureus and genome features of Lactiplantibacillus plantarum LR-14 from Sichuan pickles. Arch Microbiol 2022; 204:637. [PMID: 36127470 DOI: 10.1007/s00203-022-03232-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2021] [Revised: 08/27/2022] [Accepted: 08/30/2022] [Indexed: 11/02/2022]
Abstract
The persistence of Staphylococcus aureus within biofilm can lead to contamination of medical devices and life-threatening infections. Luckily, lactic acid bacteria (LAB) have an inhibitory effect on the growth of these bacteria. This study aims to select LAB strains from fermented vegetables, and analyze their potential inhibition activities against S. aureus. In total, 45 isolates of LAB were successfully isolated from Sichuan pickles, and the CFS of Lactiplantibacillus plantarum LR-14 exerted the strongest inhibitory effect against S. aureus. Moreover, S. aureus cells in planktonic and biofilm states both wrinkled and damaged when treated with the CFS of L. plantarum LR-14. In addition, whole genome sequencing analysis indicates that L. plantarum LR-14 contains various functional genes, including predicted extracellular polysaccharides (EPS) biosynthesis genes, and genes participating in the synthesis and metabolism of fatty acid, implying that L. plantarum LR-14 has the potential to be used as a probiotic with multiple functions.
Collapse
|
29
|
Sudan S, Li J. Dispersal of Pathogen-associated Multispecies Biofilm by Novel Probiotic Bacillus subtilis in a Contact Dependent Manner. J Appl Microbiol 2022; 133:2501-2515. [PMID: 35858688 DOI: 10.1111/jam.15721] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2022] [Revised: 06/20/2022] [Accepted: 06/27/2022] [Indexed: 11/28/2022]
Abstract
AIMS Biofilms are involved in pathogenesis of various bacterial infections. Treatment of biofilm related bacterial infection remains a major challenge due to the reduced efficacy of antibiotics and associated antibiotic resistance. Given the increasing prevalence of Enterotoxic Escherichia. coli (ETEC), Salmonella Typhimurium (S. Typhimurium), and Methicillin-resistant Staphylococcus aureus (MRSA) related infections and associated drug resistance, it is imperative to develop alternative strategies for treatment and prevention. The current study investigated antibiofilm activity of a recently isolated Bacillus subtilis (B. subtilis-9) against these pathogens. METHODS AND RESULTS Crystal violet staining showed that treatment with B. subtilis-9 significantly reduced biofilm biomass of ETEC (60-80%), S. Typhimurium (68-73%) and MRSA (66-82%). In addition, B. subtilis-9 significantly reduced pre-formed biofilm biomass of ETEC (59%), S. Typhimurium (62%), MRSA (65%) and multispecies (58%). Fluorescence microscopy revealed that B. subtilis-9 treatment significantly reduced the thickness of biofilm and viability of the embedded bacteria. Additionally, B. subtilis-9 significantly reduced planktonic cell growth of ETEC (92%), S. Typhimurium (94%) and MRSA (93%). Interestingly, transwell assay showed that B. subtilis-9 exhibited antibiofilm properties in a cell-to-cell contact dependant manner and significantly reduced mRNA expression of biofilm related genes, bssS, luxS and ihfB in ETEC. CONCLUSION Novel B. subtilis-9 exhibits a strong inhibitory activity against ETEC, S. Typhimurium and MRSA biofilm formation and adhesion to abiotic surfaces. With further investigations, our study could bring forward a novel Bacillus based probiotic intervention strategy to combat pathogenic biofilms, in clinical and agricultural settings. SIGNIFICANCE AND IMPACT OF THE STUDY Probiotic bacteria propose a potential alternative in combating biofilm related infections, however, data on the efficacy and strain selection is limited. Data from this study is critical in further developing a Bacillus-based novel probiotic applications that may reduce the use of antibiotics in biofilm related infections in humans and animals.
Collapse
Affiliation(s)
- Sudhanshu Sudan
- Department of Animal Biosciences, University of Guelph, Guelph, Ontario, Canada
| | - Julang Li
- Department of Animal Biosciences, University of Guelph, Guelph, Ontario, Canada
| |
Collapse
|
30
|
Improvements in Human Keratinocytes and Antimicrobial Effect Mediated by Cell-Free Supernatants Derived from Probiotics. FERMENTATION 2022. [DOI: 10.3390/fermentation8070332] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022] Open
Abstract
The skin acts as a physical and physiological barrier, thereby protecting the body from various environmental components and stimuli. Cell-free supernatants (CFS) derived from probiotics can improve skin functions and retain moisture. In this study, to assess the efficacy of CFS derived from Ligilactobacillus salivarius and Limosilactobacillus fermentum, we investigated the barrier strengthening and moisturizing effects of CFS in keratinocytes along with their antibacterial effects. We also determined the adhesive effects of probiotics on colorectal cells. To confirm improvements in moisturization and barrier function mediated by CFS in keratinocytes, hyaluronic acid (HA) production, and mRNA expression of HA synthases (HAS)2, HAS3, and FLG were measured. The results showed that CFS from L. salivarius MG242 and L. fermentum MG901 increased the expression of these genes along with the production of HA (2.40- and 1.95-fold of control). Additionally, CFS derived from L. salivarius MG242 and L. fermentum MG901 inhibited the growth of S. aureus and E. coli, thereby demonstrating inhibitory effects against harmful pathogens observed on the skin. These results indicate that the use of CFS derived from L. salivarius MG242 and L. fermentum MG901 may increase moisturization in the skin and improve barrier function of keratinocytes along with elimination of potential pathogens.
Collapse
|
31
|
Abd Ellatif SA, Bouqellah NA, Abu-Serie MM, Razik ESA, Al-Surhanee AA, Askary AE, Daigham GE, Mahfouz AY. Assessment of probiotic efficacy and anticancer activities of Lactiplantibacillus plantarum ESSG1 (MZ683194.1) and Lactiplantibacillus pentosus ESSG2 (MZ683195.1) isolated from dairy products. ENVIRONMENTAL SCIENCE AND POLLUTION RESEARCH INTERNATIONAL 2022; 29:39684-39701. [PMID: 35112259 DOI: 10.1007/s11356-022-18537-z] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/17/2021] [Accepted: 01/03/2022] [Indexed: 06/14/2023]
Abstract
Resistance to antibiotics is on the rise, and its indiscriminate usage has resulted in human and animal management constraints. In the research for an innovative treatment to diminish antimicrobial resistance, lactic acid bacteria (LAB) throw light on diminishing this problem in public health. As a result, this paper looked at the efficacy of LAB isolates and their active metabolites to combat pathogens, reduce antibiotic use in clinical settings, and explore the anticancer potential of 8 strains of LAB isolated from dairy products. Antifungal and antibacterial potential of LAB isolates against selected crop pathogenic fungi and food pathogenic bacteria had been estimated. Results revealed that all isolates exert antioxidant efficacy relating to DPPH, NO scavenging ability, reducing power, superoxide anion, hydroxyl radical, and anti-lipid peroxidation potential. Additionally, 12B isolate exert the highest anticancer upshot with IC50 values of 43.98 ± 0.4; 36.7 ± 0.6, 43.1 ± 0.8, and 35.1 ± 0.3 μg/ml, versus Caco-2, MCF-7, HepG-2, and PC3 cell lines respectively, whereas 13B isolate significantly had the highest selectivity index between peripheral blood mononuclear cells (PBMCs) and the tested human cancer cell lines compared to 5-fluorouracil. 13B was the most apoptosis-dependent death inducer for all human cancer cell lines besides exerting the lowest percentage of apoptosis against PBMCs suggesting its safety against PBMCs. The most promising strains 12B and 13B were identified by 16S rRNA sequencing as Lactiplantibacillus plantarum ESSG1 (MZ683194.1) and Lactiplantibacillus pentosus ESSG2 (MZ683195.1). LAB and their extracts are superb substitutive, safe, and efficient antimicrobial, antioxidant, and antitumor curative agents.
Collapse
Affiliation(s)
- Sawsan A Abd Ellatif
- Bioprocess development Department, Genetic Engineering and Biotechnology Research Institute (GEBRI), City for Scientific Research and Technology Applications, New Borg El-Arab, 21934, Alexandria, Egypt
| | - Nahla Alsayed Bouqellah
- Science College, Biology Department, Taibah University, 42317- 8599, Al-Madinah Al-Munawara, Kingdom of Saudi Arabia
| | - Marwa M Abu-Serie
- Medical Biotechnology Department, Genetic Engineering and Biotechnology Research Institute (GEBRI), the City of Scientific Research and Technology Applications, New Borg El-Arab, 21934, Alexandria, Egypt
| | - Elsayed S Abdel Razik
- Plant Protection and Biomolecular Diagnosis Department, Arid Lands Cultivation Research Institute, City for Scientific Research and Technology Applications, New Borg El-Arab, 21934, Alexandria, Egypt
| | - Ameena A Al-Surhanee
- Biology Department, College of Science, Jouf University, Sakaka, 2014, Kingdom of Saudi Arabia
| | - Ahmad El Askary
- Department of Clinical Laboratory Sciences, College of Applied Medical Sciences, Taif University, P.O. Box 11099, Taif, 21944, Saudi Arabia
| | - Ghadir E Daigham
- Botany and Microbiology Department, Faculty of Science, Al-Azhar University (Girls Branch), Cairo, Egypt
| | - Amira Y Mahfouz
- Botany and Microbiology Department, Faculty of Science, Al-Azhar University (Girls Branch), Cairo, Egypt.
| |
Collapse
|
32
|
Squarzanti DF, Zanetta P, Ormelli M, Manfredi M, Barberis E, Vanella VV, Amoruso A, Pane M, Azzimonti B. An animal derivative-free medium enhances Lactobacillus johnsonii LJO02 supernatant selective efficacy against the methicillin (oxacillin)-resistant Staphylococcus aureus virulence through key-metabolites. Sci Rep 2022; 12:8666. [PMID: 35606510 PMCID: PMC9126979 DOI: 10.1038/s41598-022-12718-z] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2022] [Accepted: 05/09/2022] [Indexed: 12/15/2022] Open
Abstract
The spread of multidrug-resistant bacteria, such as the skin commensal Staphylococcus aureus, is a worldwide health challenge; new methods to counteract opportunistic pathogen growth and virulence are urgent. We compared the activity of Lacticaseibacillus rhamnosus LR06 (DSM 21981) and Lactobacillus johnsonii LJO02 (DSM 33828) cell-free supernatants (CFSs) produced in the conventional animal derivative-based MRS medium and an innovative animal derivative-free broth (TIL) versus the MDR S. aureus (ATCC 43300). CFS influence was assessed towards the viability, metabolic activity, and ability to form biofilm of the MDR strain through optical density, alamarBlue assay, and crystal violet staining; their content in short-chain fatty acids, lactic acid, and proteins was analysed via high-resolution mass spectrometry and gas chromatography. All CFSs reduce viable and metabolically active S. aureus, being TIL more efficient compared to MRS in stimulating lactic acid bacteria metabolism and decreasing S. aureus biofilm formation. Particularly, the CFS from LJO02 grown in TIL has the best efficacy, revealing a high amount of lactic acid and 59 peculiar proteins; its effectiveness is partially maintained upon trypsin and proteinase K treatments, but not by pepsin and pH basification. Therefore, antagonistic CFSs may represent a strategic prevention approach, with bacteriotherapeutic and bio-repair potential.
Collapse
Affiliation(s)
- Diletta Francesca Squarzanti
- Laboratory of Applied Microbiology, Department of Health Sciences (DiSS), Center for Translational Research on Allergic and Autoimmune Diseases (CAAD), School of Medicine, Università del Piemonte Orientale (UPO), Corso Trieste 15/A, 28100, Novara, Italy
| | - Paola Zanetta
- Laboratory of Applied Microbiology, Department of Health Sciences (DiSS), Center for Translational Research on Allergic and Autoimmune Diseases (CAAD), School of Medicine, Università del Piemonte Orientale (UPO), Corso Trieste 15/A, 28100, Novara, Italy
| | - Margherita Ormelli
- Laboratory of Applied Microbiology, Department of Health Sciences (DiSS), Center for Translational Research on Allergic and Autoimmune Diseases (CAAD), School of Medicine, Università del Piemonte Orientale (UPO), Corso Trieste 15/A, 28100, Novara, Italy
| | - Marcello Manfredi
- Laboratory of Biological Mass Spectrometry, Department of Translational Medicine (DiMeT), Center for Translational Research On Allergic and Autoimmune Diseases (CAAD), School of Medicine, Università del Piemonte Orientale (UPO), Corso Trieste 15/A, 28100, Novara, Italy
| | - Elettra Barberis
- Laboratory of Biological Mass Spectrometry, Department of Translational Medicine (DiMeT), Center for Translational Research On Allergic and Autoimmune Diseases (CAAD), School of Medicine, Università del Piemonte Orientale (UPO), Corso Trieste 15/A, 28100, Novara, Italy
| | - Virginia Vita Vanella
- Laboratory of Biological Mass Spectrometry, Department of Translational Medicine (DiMeT), Center for Translational Research On Allergic and Autoimmune Diseases (CAAD), School of Medicine, Università del Piemonte Orientale (UPO), Corso Trieste 15/A, 28100, Novara, Italy
| | - Angela Amoruso
- Probiotical Research S.R.L, Via Mattei 3, 28100, Novara, Italy
| | - Marco Pane
- Probiotical Research S.R.L, Via Mattei 3, 28100, Novara, Italy
| | - Barbara Azzimonti
- Laboratory of Applied Microbiology, Department of Health Sciences (DiSS), Center for Translational Research on Allergic and Autoimmune Diseases (CAAD), School of Medicine, Università del Piemonte Orientale (UPO), Corso Trieste 15/A, 28100, Novara, Italy.
| |
Collapse
|
33
|
Asadi A, Lohrasbi V, Abdi M, Mirkalantari S, Esghaei M, Kashanian M, Oshaghi M, Talebi M. The probiotic properties and potential of vaginal Lactobacilli spp. isolated from healthy women against some vaginal pathogens. Lett Appl Microbiol 2022; 74:752-764. [DOI: 10.1111/lam.13660] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2022] [Accepted: 01/21/2022] [Indexed: 11/26/2022]
Affiliation(s)
- Arezoo Asadi
- Department of Microbiology School of Medicine Iran University of Medical Sciences Tehran Iran
| | - Vahid Lohrasbi
- Department of Microbiology School of Medicine Iran University of Medical Sciences Tehran Iran
| | - Milad Abdi
- Department of Microbiology School of Medicine Iran University of Medical Sciences Tehran Iran
| | - Shiva Mirkalantari
- Department of Microbiology School of Medicine Iran University of Medical Sciences Tehran Iran
| | - Maryam Esghaei
- Department of Virology School of Medicine Iran University of Medical Sciences Tehran Iran
| | - Maryam Kashanian
- Department of Obstetrics & Gynecology Akbarabadi Teaching Hospital, Iran University of Medical Sciences Tehran Iran
| | - Mozhgan Oshaghi
- Department of Lab Sciences Faculty of Allied Medicine Iran University of Medical Sciences Tehran Iran
| | - Malihe Talebi
- Department of Microbiology School of Medicine Iran University of Medical Sciences Tehran Iran
- Microbial Biotechnology Research Centre Iran University of Medical Sciences Shahid Hemmat Highway Tehran Iran
| |
Collapse
|
34
|
Giordani B, Parolin C, Vitali B. Lactobacilli as Anti-biofilm Strategy in Oral Infectious Diseases: A Mini-Review. FRONTIERS IN MEDICAL TECHNOLOGY 2022; 3:769172. [PMID: 35047965 PMCID: PMC8757881 DOI: 10.3389/fmedt.2021.769172] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2021] [Accepted: 09/20/2021] [Indexed: 12/14/2022] Open
Abstract
The spread of biofilm-related diseases in developed countries has led to increased mortality rates and high health care costs. A biofilm is a community of microorganisms that is irreversibly attached to a surface, behaving very differently from planktonic cells and providing resistance to antimicrobials and immune response. Oral diseases are an excellent example of infection associated with the formation of highly pathogenic biofilms. It is generally accepted that, when the oral homeostasis is broken, the overgrowth of pathogens is facilitated. Among them, Porphyromonas gingivalis and Aggregatibacter actinomycetemcomitans are the main etiological agents of periodontitis, while Streptococcus mutans is strongly associated with the onset of dental caries. Other microorganisms, such as the fungus Candida albicans, may also be present and contribute to the severity of infections. Since the common antibiotic therapies usually fail to completely eradicate biofilm-related oral diseases, alternative approaches are highly required. In this regard, the topical administration of probiotics has recently gained interest in treating oral diseases. Thus, the present mini-review focuses on the possibility of using Lactobacillus spp. as probiotics to counteract biofilm-mediated oral infections. Many evidence highlight that Lactobacillus living cells can impede the biofilm formation and eradicate mature biofilms of different oral pathogens, by acting through different mechanisms. Even more interestingly, lactobacilli derivatives, namely postbiotics (soluble secreted products) and paraprobiotics (cell structural components) are able to trigger anti-biofilm effects too, suggesting that they can represent a novel and safer alternative to the use of viable cells in the management of biofilm-related oral diseases.
Collapse
Affiliation(s)
- Barbara Giordani
- Department of Pharmacy and Biotechnology, University of Bologna, Bologna, Italy
| | - Carola Parolin
- Department of Pharmacy and Biotechnology, University of Bologna, Bologna, Italy
| | - Beatrice Vitali
- Department of Pharmacy and Biotechnology, University of Bologna, Bologna, Italy
| |
Collapse
|
35
|
Dong J, Li W, Wang Q, Chen J, Zu Y, Zhou X, Guo Q. Relationships Between Oral Microecosystem and Respiratory Diseases. Front Mol Biosci 2022; 8:718222. [PMID: 35071321 PMCID: PMC8767498 DOI: 10.3389/fmolb.2021.718222] [Citation(s) in RCA: 50] [Impact Index Per Article: 16.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2021] [Accepted: 12/09/2021] [Indexed: 02/05/2023] Open
Abstract
Oral microecosystem is a very complicated ecosystem that is located in the mouth and comprises oral microbiome, diverse anatomic structures of oral cavity, saliva and interactions between oral microbiota and between oral microbiota and the host. More and more evidence from studies of epidemiology, microbiology and molecular biology is establishing a significant link between oral microecosystem and respiratory diseases. Microbiota settling down in oral microecosystem is known as the main source of lung microbiome and has been associated with the occurrence and development of respiratory diseases like pneumonia, chronic obstructive pulmonary disease, lung cancer, cystic fibrosis lung disease and asthma. In fact, it is not only indigenous oral microbes promote or directly cause respiratory infection and inflammation when inhaled into the lower respiratory tract, but also internal environment of oral microecosystem serves as a reservoir for opportunistic respiratory pathogens. Moreover, poor oral health and oral diseases caused by oral microecological dysbiosis (especially periodontal disease) are related with risk of multiple respiratory diseases. Here, we review the research status on the respiratory diseases related with oral microecosystem. Potential mechanisms on how respiratory pathogens colonize oral microecosystem and the role of indigenous oral microbes in pathogenesis of respiratory diseases are also summarized and analyzed. Given the importance of oral plaque control and oral health interventions in controlling or preventing respiratory infection and diseases, we also summarize the oral health management measures and attentions, not only for populations susceptible to respiratory infection like the elderly and hospitalized patients, but also for dentist or oral hygienists who undertake oral health care. In conclusion, the relationship between respiratory diseases and oral microecosystem has been established and supported by growing body of literature. However, etiological evidence on the role of oral microecosystem in the development of respiratory diseases is still insufficient. Further detailed studies focusing on specific mechanisms on how oral microecosystem participate in the pathogenesis of respiratory diseases could be helpful to prevent and treat respiratory diseases.
Collapse
Affiliation(s)
- Jiajia Dong
- Department of Pulmonary and Critical Care Medicine, West China Hospital, Sichuan University, Chengdu, China
| | - Wei Li
- State Key Laboratory of Oral Diseases, National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, China
| | - Qi Wang
- State Key Laboratory of Oral Diseases, National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, China
| | - Jiahao Chen
- State Key Laboratory of Oral Diseases, National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, China
| | - Yue Zu
- State Key Laboratory of Oral Diseases, National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, China
| | - Xuedong Zhou
- State Key Laboratory of Oral Diseases, National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, China
| | - Qiang Guo
- State Key Laboratory of Oral Diseases, National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, China
| |
Collapse
|
36
|
Guerrero Sanchez M, Passot S, Campoy S, Olivares M, Fonseca F. Ligilactobacillus salivarius functionalities, applications, and manufacturing challenges. Appl Microbiol Biotechnol 2021; 106:57-80. [PMID: 34889985 DOI: 10.1007/s00253-021-11694-0] [Citation(s) in RCA: 45] [Impact Index Per Article: 11.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2021] [Revised: 11/11/2021] [Accepted: 11/15/2021] [Indexed: 11/25/2022]
Abstract
Ligilactobacillus salivarius is a lactic acid bacteria that has been gaining attention as a promising probiotic. Numerous strains exhibit functional properties with health benefits such as antimicrobial activity, immunological effects, and the ability to modulate the intestinal microbiota. However, just a small number of them are manufactured at an industrial scale and included in commercial products. The under exploitation of L. salivarius strains that remain in the freezer of companies is due to their incapacity to overcome the environmental stresses induced by production and stabilization processes.The present study summarizes the functionalities and applications of L. salivarius reported to date. It aims also at providing a critical evaluation of the literature available on the manufacturing steps of L. salivarius concentrates, the bacterial quality after each step of the process, and the putative degradation and preservation mechanisms. Here, we highlight the principal issues and future research challenges for improving the production and long-term preservation at the industrial scale of this microorganism, and probably of other probiotics.Key points• L. salivarius beneficial properties and commercialized products.• Production conditions and viability of L. salivarius after stabilization processes.• Prospects for identifying preservation mechanisms to improve L. salivarius stability.
Collapse
Affiliation(s)
| | - S Passot
- Université Paris-Saclay, INRAE, AgroParisTech, UMR SayFood, 78850, Thiverval-Grignon, France
| | - S Campoy
- R&D Department, Biosearch Life, 18004, Granada, Spain
| | - M Olivares
- R&D Department, Biosearch Life, 18004, Granada, Spain
| | - F Fonseca
- Université Paris-Saclay, INRAE, AgroParisTech, UMR SayFood, 78850, Thiverval-Grignon, France.
| |
Collapse
|
37
|
Kang MS, Park GY. In Vitro Evaluation of the Effect of Oral Probiotic Weissella cibaria on the Formation of Multi-Species Oral Biofilms on Dental Implant Surfaces. Microorganisms 2021; 9:microorganisms9122482. [PMID: 34946084 PMCID: PMC8707126 DOI: 10.3390/microorganisms9122482] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2021] [Revised: 11/19/2021] [Accepted: 11/29/2021] [Indexed: 11/16/2022] Open
Abstract
Oral probiotics are beneficial bacteria that can help prevent periodontal disease. However, little is known about the effects of oral probiotics on the formation of implant biofilms. This study aimed to evaluate the effects of oral probiotics Weissella cibaria CMU and CMS1 in an in vitro complex biofilm model on titanium implant surfaces. First, it was identified through colony biofilm assay that W. cibaria CMU and CMS1 inhibit the formation of multi-species biofilms formed by eight types of bacteria. Two types of saliva-coated titanium discs inoculated with early (Streptococcus gordonii, Streptococcus oralis, Streptococcus sanguinis, Actinomyces naeslundii, and Veillonella parvula), secondary (Fusobacterium nucleatum and Prevotella intermedia), and late (Porphyromonas gingivalis) colonizers were treated with the oral probiotics and then incubated anaerobically for three days. The effects of oral probiotics on titanium disc biofilm formation were analyzed using culture methods, quantitative polymerase chain reaction (qPCR), and microscopic analysis. Both probiotics significantly inhibited the formation of biofilm, and all eight bacterial species were significantly reduced. The effectiveness of both probiotic strains was confirmed by all the methods used. Oral probiotics may have dramatically reduced the biofilm formation of secondary colonizers that act as bridges, thus inhibiting biofilm formation on the titanium surface. Our results suggest that the probiotic W. cibaria offers new possibilities for the prevention of peri-implant mucositis.
Collapse
|
38
|
Abstract
Microbes in the 21st century are understood as symbionts ‘completing’ the human ‘superorganism’ (Homo sapiens plus microbial partners-in-health). This paper addresses a significant paradox: despite the vast majority of our genes being microbial, the lack of routine safety testing for the microbiome has led to unintended collateral side effects from pharmaceuticals that can damage the microbiome and inhibit innate ‘colonization resistance’ against pathobionts. Examples are discussed in which a Microbiome First Medicine approach provides opportunities to ‘manage our microbes’ holistically, repair dysbiotic superorganisms, and restore health and resilience in the gut and throughout the body: namely, managing nosocomial infections for Clostridioides difficile and Staphylococcus aureus and managing the gut and neural systems (gut–brain axis) in autism spectrum disorder. We then introduce a risk analysis tool: the evidence map. This ‘mapping’ tool was recently applied by us to evaluate evidence for benefits, risks, and uncertainties pertaining to the breastmilk ecosystem. Here, we discuss the potential role of the evidence map as a risk analysis methodology to guide scientific and societal efforts to: (1) enhance ecosystem resilience, (2) ‘manage our microbes’, and (3) minimize the adverse effects of both acute and chronic diseases.
Collapse
|
39
|
Exoproteome Analysis of Antagonistic Interactions between the Probiotic Bacteria Limosilactobacillus reuteri LR1 and Lacticaseibacillus rhamnosus F and Multidrug Resistant Strain of Klebsiella pneumonia. Int J Mol Sci 2021; 22:ijms222010999. [PMID: 34681658 PMCID: PMC8537075 DOI: 10.3390/ijms222010999] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2021] [Revised: 10/06/2021] [Accepted: 10/08/2021] [Indexed: 12/21/2022] Open
Abstract
The expansion of multiple drug resistant (MDR) strains of Klebsiella pneumoniae presents an immense threat for public health. Annually, this microorganism causes thousands of lethal nosocomial infections worldwide. Currently, it has been shown that certain strains of lactic acid bacteria (LAB) can efficiently inhibit growth of K. pneumoniae and the formation of its biofilms; however, the active principle of such action remains unknown. In the current article, the growth inhibition of MDR K. pneumoniae by two LAB—Limosilactobacillus reuteri LR1 and Lacticaseibacillus rhamnosus F—is demonstrated, and the nature of this inhibition studied at the level of exoproteome. This article shows that the exoproteomes of studied LAB contains both classically and non-classically secreted proteins. While for L. reuteri LR1 the substantial portion of classically secreted proteins was presented by cell-wall-degrading enzymes, for L. rhamnosus F only one out of four classically secreted proteins was presented by cell-wall hydrolase. Non-classically secreted proteins of both LAB were primarily metabolic enzymes, for some of which a possible moonlighting functioning was proposed. These results contribute to knowledge regarding antagonistic interaction between LAB and pathogenic and opportunistic microorganisms and set new perspectives for the use of LAB to control the spread of these microorganisms.
Collapse
|
40
|
Comparative Genomic Analysis Determines the Functional Genes Related to Bile Salt Resistance in Lactobacillus salivarius. Microorganisms 2021; 9:microorganisms9102038. [PMID: 34683359 PMCID: PMC8539994 DOI: 10.3390/microorganisms9102038] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2021] [Revised: 09/17/2021] [Accepted: 09/18/2021] [Indexed: 11/23/2022] Open
Abstract
Lactobacillus salivarius has drawn attention because of its promising probiotic functions. Tolerance to the gastrointestinal tract condition is crucial for orally administrated probiotics to exert their functions. However, previous studies of L. salivarius have only focused on the bile salt resistance of particular strains, without uncovering the common molecular mechanisms of this species. Therefore, in this study, we expanded our research to 90 L. salivarius strains to explore their common functional genes for bile salt resistance. First, the survival rates of the 90 L. salivarius strains in 0.3% bile salt solutions were determined. Comparative genomics analysis was then performed to screen for the potential functional genes related to bile salt tolerance. Next, real-time polymerase chain reaction and gene knockout experiments were conducted to further verify the tolerance-related functional genes. The results indicated that the strain-dependent bile salt tolerance of L. salivarius was mainly associated with four peptidoglycan synthesis-related genes, seven phosphotransferase system-related genes, and one chaperone-encoding gene involved in the stress response. Among them, the GATase1-encoding gene showed the most significant association with bile salt tolerance. In addition, four genes related to DNA damage repair and substance transport were redundant in the strains with high bile salt tolerance. Besides, cluster analysis showed that bile salt hydrolases did not contribute to the bile salt tolerance of L. salivarius. In this study, we determined the global regulatory genes, including LSL_1568, LSL_1716 and LSL_1709, for bile salt tolerance in L. salivarius and provided a potential method for the rapid screening of bile salt-tolerant L. salivarius strains, based on PCR amplification of functional genes.
Collapse
|
41
|
The Influence of Environmental Conditions on the Antagonistic Activity of Lactic Acid Bacteria Isolated from Fermented Meat Products. Foods 2021; 10:foods10102267. [PMID: 34681316 PMCID: PMC8534964 DOI: 10.3390/foods10102267] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2021] [Revised: 09/17/2021] [Accepted: 09/22/2021] [Indexed: 02/01/2023] Open
Abstract
The aim of this study was to determine the impact of environmental conditions on the antimicrobial properties of 21 lactic acid bacteria strains in the selected indicator bacteria. To assess the antimicrobial activity of the whole bacteria culture (WBC), the agar well diffusion method was used. The interference of LAB strains with the growth of the selected indicator bacteria was evaluated by incubating co-cultures in the food matrix. Based on the conducted research, it was found that environmental conditions have a significant impact on the antimicrobial activity of lactic acid bacteria strains. The highest antimicrobial activity was recorded under optimal conditions for the development of LAB, the incubation time being different depending on the indicator strain used. The tested LAB strains were characterized by a high ability to inhibit indicator strains, especially in the food matrix. These results led us to further characterize and purify the antimicrobial compound produced by lactic acid bacteria taking into account changing environmental conditions.
Collapse
|
42
|
Amirkhanova Z, Akhmetova S, Kozhakhmetov S, Kushugulova A, Bodeeva R, Issina Z, Tusbayev M. Screening of Antimicrobial and Adhesive Activity of Lactobacilli Isolated from the National Food Products from Different Districts of the Karaganda Region (Kazakhstan). Open Access Maced J Med Sci 2021. [DOI: 10.3889/oamjms.2021.7053] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/05/2022] Open
Abstract
BACKGROUND: It is a national priority to look for new probiotic bacteria with highly active biological properties to create a new generation of probiotics, ferments, therapeutic, and prophylactic fermented milk products, taking into account ethnocultural and regional characteristics.
AIM: The aim of the study is to assess probiotic properties of strains of lactobacilli (antimicrobial and adhesive), which are isolated from national lactic acid products from different districts of the Karaganda region (Kazakhstan).
MATERIALS AND METHODS: There were modern microbiological methods applied during the experiment. To determine the morpho-cultural properties, the following methods were used: Gram staining, a catalase test, serial dilutions. The Matrix Supported Laser Desorption/Ionization Flight Time Mass Spectrometry was used for identification, and the deferred-antagonism method was used to determine the antimicrobial activity. The buccal epithelial cells were used for the cell object as a test system to determine the adhesive activity.
RESULTS: In this experiment, 26 lactobacillus isolates were isolated from 68 samples of national lactic acid products produced in a traditional homemade way in different districts of the Karaganda region (Kazakhstan). As a result of the studies carried out on the cultural and morphological characteristics and identification by the mass spectrometer, the following lactobacilli were obtained: Lactobacillus acidophilus (two strains), Lactobacillus delbrueckii subsp. bulgaricum (two strains), Lactobacillus rhamnosus (seven strains), Lactobacillus plantarum (two strains), Lactobacillus paracasei (11 strains), and Lactobacillus fermentum (two strains). Twenty-six isolates of lactobacilli were tested for antimicrobial activity, 13 isolates of which showed an inhibitory effect, but the degree of antagonism varied among lactobacillus isolates. In general, the inhibitory activity of lactobacillus isolates was shown against the Gram-negative indicator microorganisms Salmonella typhimurium NCTC 12023, Escherichia coli NCTC 12923. The antibacterial activity was shown against the Staphylococcus aureus NCTC 12973 indicator microorganism in nine isolates of lactobacilli. Only six isolates of lactobacilli showed antifungal activity against the test strain of Candida albicans NCPF 3179. Out of 13 isolates of lactobacilli, nine isolates of medium and high activity competed for binding to buccal epithelial cells.
CONCLUSION: The obtained isolates from traditional dairy products are considered to be promising candidates and competitive isolates with some probiotic potential. This study calls for further researches to be made in this area.
Collapse
|
43
|
Paniágua AL, Correia AF, Pereira LC, de Alencar BM, Silva FBA, Almeida RM, de Medeiros Nóbrega YK. Inhibitory effects of Lactobacillus casei Shirota against both Candida auris and Candida spp. isolates that cause vulvovaginal candidiasis and are resistant to antifungals. BMC Complement Med Ther 2021; 21:237. [PMID: 34556109 PMCID: PMC8461885 DOI: 10.1186/s12906-021-03405-z] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2020] [Accepted: 09/01/2021] [Indexed: 12/25/2022] Open
Abstract
BACKGROUND Vulvovaginal candidiasis (VVC), the second leading cause of genital infection in women of reproductive age, is caused by yeasts of the genus Candida. Treatment is usually empirical and performed with azoles, which have shown increasing ineffectiveness due to resistance from these species. This therapeutic challenge has led to the search for new treatment strategies. Lactobacillus spp. produce several components with microbicidal effects, such as lactic acid. These species are the main components of a healthy vaginal microbiota and have been used as probiotics. The aim of this work was to investigate the in vitro inhibitory effects of Lactobacillus casei Shirota on both the Candida spp. that cause VVC and on C. auris. METHODS The microbicidal effects of L. casei Shirota on the main VVC-causing species, C. albicans, C. tropicalis, C. norvegensis and C. parapsilosis, in addition to C. auris were investigated by counting the Colony-forming Units (CFUs) after cocultivation. The antifungal activity of lactic acid against these Candida strains was assessed using the microtiter broth dilution method to determine the minimum inhibitory concentrations (MICs). The effects of L. casei Shirota on hyphal and early biofilm formation was measured by optical microscopy. RESULTS L. casei Shirota showed inhibitory action against all tested Candida spp., ranging from 66.9 to 95.6% inhibition depending on the species. This inhibition is possibly related to the production of lactic acid, since lactic acid has shown microbicidal action against these same Candida spp. at a concentration of 5 mg/mL, which corresponds to half of the normal physiological concentration. In addition, L. casei Shirota was able to reduce the formation of C. albicans hyphae and early biofilms, showing strong anti-Candida effects. CONCLUSIONS These results suggest that L. casei Shirota has antifungal activity against the Candida species that cause VVC. L. casei also has microbicidal action against C. auris.
Collapse
Affiliation(s)
- Aline Lorenzoni Paniágua
- Clinical Microbiology and Immunology Laboratory, Department of Pharmacy, University of Brasilia, Campus Darcy Ribeiro, Brasilia, DF, 70910-900, Brazil
| | - Amabel Fernandes Correia
- Central Public Health Laboratory of the District Federal (LACEN-DF), Medical Biology Management, Center of Parasitology and Mycology, Brasília, DF, Brazil
| | - Lívia Custódio Pereira
- Vulvar Pathology Clinic, Department of Gynecology, Brasilia University Hospital, University of Brasilia, Brasilia, DF, Brazil
| | - Bruna Maciel de Alencar
- Clinical Microbiology and Immunology Laboratory, Department of Pharmacy, University of Brasilia, Campus Darcy Ribeiro, Brasilia, DF, 70910-900, Brazil
| | - Fabiana Brandão Alves Silva
- Clinical Microbiology and Immunology Laboratory, Department of Pharmacy, University of Brasilia, Campus Darcy Ribeiro, Brasilia, DF, 70910-900, Brazil
| | - Rosane Mansan Almeida
- Clinical Microbiology and Immunology Laboratory, Department of Pharmacy, University of Brasilia, Campus Darcy Ribeiro, Brasilia, DF, 70910-900, Brazil.
| | - Yanna Karla de Medeiros Nóbrega
- Clinical Microbiology and Immunology Laboratory, Department of Pharmacy, University of Brasilia, Campus Darcy Ribeiro, Brasilia, DF, 70910-900, Brazil
| |
Collapse
|
44
|
Effect of Novel Bacteriocinogenic Lactobacillus fermentum BZ532 on Microbiological Shelf-Life and Physicochemical and Organoleptic Properties of Fresh Home-Made Bozai. Foods 2021; 10:foods10092120. [PMID: 34574232 PMCID: PMC8470737 DOI: 10.3390/foods10092120] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2021] [Revised: 08/24/2021] [Accepted: 08/27/2021] [Indexed: 11/17/2022] Open
Abstract
Bacteriocinogenic Lactobacillus fermentum BZ532 with novel bacteriocin LF-BZ532 was originally isolated from Chinese cereal fermented drink, showing an antimicrobial characteristic during fermentation. This study aimed to explore the in situ antimicrobial activities of L. fermentum BZ532 and co-culturing investigation against key food pathogens, i.e., Staphylococcus aureus and Escherichia coli K-12, was conducted during fresh bozai production. The growth of spoilage bacteria was suppressed and bacterial count was reduced to a significantly low level during 48 h of co-cultures. In situ production of antimicrobial compounds expressed positive activity against S. aureus and E. coli K-12, but negative acitivity against Salmonella sp. D104. The total viable count of bozai BZ-Lf (bozai fermented with BZ532 strain) had a comparatively lower viable count than bozai BZ-C (bozai as an experimental control without BZ532) during storage of 7 days. Titratable acidity of bozai treatments (BZ-C, BZ-Lf) was increased, while pH declined accordingly during storage of 7 days. The organoleptic quality of bozai BZ-C had low sensorial scores as compared with BZ-Lf during storage. In comparison with naturally fermented bozai (BZ-C), L. fermentum BZ532 (BZ-Lf) could significantly reduce the microbial spoilage and extend the shelf-life based on microbiological examination. Conclusively, L. fermentum BZ532 can be used as a bio-protective culture for improving the safety of bozai.
Collapse
|
45
|
Sornsenee P, Singkhamanan K, Sangkhathat S, Saengsuwan P, Romyasamit C. Probiotic Properties of Lactobacillus Species Isolated from Fermented Palm Sap in Thailand. Probiotics Antimicrob Proteins 2021; 13:957-969. [PMID: 33595830 DOI: 10.1007/s12602-021-09754-y] [Citation(s) in RCA: 25] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 02/04/2021] [Indexed: 10/22/2022]
Abstract
Lactic acid bacteria (LAB), which are the most frequently used probiotics in foods, confer health benefits such as antimicrobial activity, immune stimulation, and anticancer activity. Fermented palm sap is a potential source of LAB. This study aimed to evaluate in vitro antimicrobial and probiotic properties of LAB isolated from traditional fermented palm sap in Thailand. Among 40 isolated LAB species, 10 were preliminarily selected for their antimicrobial activity. These 10 isolates were identified and confirmed by matrix-assisted laser desorption ionization time-of-flight mass spectrometry (MALDI-TOF MS) and 16S rRNA sequencing as Lactobacillus paracasei (8/10), Lactobacillus fermentum (1/10), and Lactobacillus brevis (1/10). They were evaluated for probiotic characteristics and antimicrobial activities against pathogens. These isolates were tolerant toward simulated gastrointestinal tract conditions, including low pH, pepsin, pancreatin, and bile salts. The 10 isolates retained strong auto-aggregation and cell surface hydrophobicity, and they adhered tightly to human intestinal epithelial cells. The isolates were susceptible to ampicillin, erythromycin, clindamycin, tetracycline, and chloramphenicol but resistant to vancomycin, kanamycin, and streptomycin. Moreover, all isolates exhibited no hemolytic activity. All isolates exhibited good antibacterial activity against nine pathogenic bacteria. Thus, these 10 Lactobacillus isolates from fermented palm sap are promising potential candidates for use as probiotics in functional fermented foods and pharmaceutical products.
Collapse
Affiliation(s)
- Phoomjai Sornsenee
- Department of Family and Preventive Medicine, Faculty of Medicine, Prince of Songkla University, Songkhla, 90110, Thailand
| | - Kamonnut Singkhamanan
- Department of Biomedical Sciences, Faculty of Medicine, Prince of Songkla University, Songkhla, 90110, Thailand
| | - Surasak Sangkhathat
- Department of Biomedical Sciences, Faculty of Medicine, Prince of Songkla University, Songkhla, 90110, Thailand
- Department of Surgery, Faculty of Medicine, Prince of Songkla University, Songkhla, 90110, Thailand
| | - Phanvasri Saengsuwan
- Department of Biomedical Sciences, Faculty of Medicine, Prince of Songkla University, Songkhla, 90110, Thailand
| | - Chonticha Romyasamit
- Department of Medical Technology, School of Allied Health Sciences, Walailak University, Nakhon Si Thammarat, 80161, Thailand.
| |
Collapse
|
46
|
Antibacterial and Antifungal Activity of the Human Endometrial Fluid during the Natural Cycle. Infect Dis Obstet Gynecol 2021; 2021:8849664. [PMID: 34220191 PMCID: PMC8221874 DOI: 10.1155/2021/8849664] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2020] [Revised: 04/26/2021] [Accepted: 06/02/2021] [Indexed: 11/18/2022] Open
Abstract
Purpose Some microbiota patterns have been associated with favorable IVF prognosis and others with pathological conditions. The endometrial fluid aspirate (EFA) contains antibacterial proteins that are enriched in implantative IVF cycles, but the antimicrobial effect of EFA has not been addressed. We aimed to evaluate the antimicrobial activity of the human endometrial fluid during the natural cycle. Methods EFA was obtained through an embryo transfer catheter in 38 women, aged 18-40 years, with regular cycles attending to a fertility clinic. The antimicrobial activity of EFAs was tested against two strains of Staphylococcus aureus; one strain each of Streptococcus agalactiae, Enterococcus faecalis, Escherichia coli, and Klebsiella pneumoniae; and three yeasts (Candida albicans, Candida glabrata, and Candida krusei). Results All samples exhibited antibacterial activity against S. aureus. In addition, 32.4% of EFAs were active against one of the other microorganisms assayed, 16.2% against two, and 5.4% against four of them. In contrast, none exhibited antibacterial activity against E. coli or K. pneumoniae. The antimicrobial activity differs considerably between EFA samples, and we failed to observe a cycle-related pattern. Conclusions EFA presented two antimicrobial activity patterns: (a) one common to all the samples, exhibiting activity against S. aureus and lack of activity against E. coli and K. pneumoniae, and (b) an individualized pattern, showing activity against some of the other microorganisms tested. The intensity of antibacterial activity differs between EFA samples. Our data suggest that the uterine microbiota is controlled by means of endometrial fluid components.
Collapse
|
47
|
Yeu JE, Lee HG, Park GY, Lee J, Kang MS. Antimicrobial and Antibiofilm Activities of Weissella cibaria against Pathogens of Upper Respiratory Tract Infections. Microorganisms 2021; 9:1181. [PMID: 34070813 PMCID: PMC8229644 DOI: 10.3390/microorganisms9061181] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2021] [Revised: 05/27/2021] [Accepted: 05/27/2021] [Indexed: 01/15/2023] Open
Abstract
Recently discovered preventive effects of probiotics on oral health have attracted interest to their use for the prevention and treatment of various diseases. This study aimed to evaluate the antimicrobial and antibiofilm properties of Weissella cibaria against Streptococcus pyogenes, Staphylococcus aureus, S. pneumoniae, and Moraxella catarrhalis, the major pathogens of upper respiratory tract infections (URTIs). The antimicrobial activities of W. cibaria were compared with those of other oral probiotics using a competitive inhibition assay and the determination of the minimum inhibitory concentrations (MICs). In addition, a time-kill assay, spectrophotometry, and confocal laser scanning microscopy were used to confirm the antimicrobial and antibiofilm abilities of W. cibaria CMU (oraCMU) and CMS1 (oraCMS1). Both live cells and cell-free supernatants of all tested probiotics, except Streptococcus salivarius, showed excellent antimicrobial activities. All target pathogens were killed within 4 to 24 h at twice the MIC of oraCMU and oraCMS1, which showed the highest antimicrobial activities against M. catarrhalis. The antimicrobial substances that affected different target pathogens were different. Both oraCMU and oraCMS1 showed excellent abilities to inhibit biofilm formation and remove preformed biofilms. Our results suggest that the W. cibaria probiotics offer new possibilities for the prevention and treatment of bacterial URTIs.
Collapse
Affiliation(s)
- Ji-Eun Yeu
- R&D Center, OraPharm, Inc., Seoul 04782, Korea; (J.-E.Y.); (G.-Y.P.)
- Department of Food and Nutrition, Hanyang University, Seoul 04763, Korea;
| | - Hyeon-Gyu Lee
- Department of Food and Nutrition, Hanyang University, Seoul 04763, Korea;
| | - Geun-Yeong Park
- R&D Center, OraPharm, Inc., Seoul 04782, Korea; (J.-E.Y.); (G.-Y.P.)
| | - Jisun Lee
- Bio-Healthcare Food Science Interdisciplinary Major, School of Humanities, Art & Technology, Kookmin University, Seoul 02707, Korea;
| | - Mi-Sun Kang
- R&D Center, OraPharm, Inc., Seoul 04782, Korea; (J.-E.Y.); (G.-Y.P.)
| |
Collapse
|
48
|
Limosilactobacillus fermentum CECT5716: Mechanisms and Therapeutic Insights. Nutrients 2021; 13:nu13031016. [PMID: 33801082 PMCID: PMC8003974 DOI: 10.3390/nu13031016] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2021] [Revised: 03/09/2021] [Accepted: 03/18/2021] [Indexed: 12/12/2022] Open
Abstract
Probiotics microorganisms exert their health-associated activities through some of the following general actions: competitive exclusion, enhancement of intestinal barrier function, production of bacteriocins, improvement of altered microbiota, and modulation of the immune response. Among them, Limosilactobacillus fermentum CECT5716 has become one of the most promising probiotics and it has been described to possess potential beneficial effects on inflammatory processes and immunological alterations. Different studies, preclinical and clinical trials, have evidenced its anti-inflammatory and immunomodulatory properties and elucidated the precise mechanisms of action involved in its beneficial effects. Therefore, the aim of this review is to provide an updated overview of the effect on host health, mechanisms, and future therapeutic approaches.
Collapse
|
49
|
Zielińska D, Łepecka A, Ołdak A, Długosz E, Kołożyn-Krajewska D. Growth and adhesion inhibition of pathogenic bacteria by live and heat-killed food-origin Lactobacillus strains or their supernatants. FEMS Microbiol Lett 2021; 368:6149457. [PMID: 33629723 DOI: 10.1093/femsle/fnab024] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2020] [Accepted: 02/23/2021] [Indexed: 12/27/2022] Open
Abstract
The study aimed to evaluate qualitatively and quantitatively the antimicrobial capacity of 10 potential probiotic Lactobacillus strains against model enteropathogens and spoilage microorganisms. The probiotic strains (live and heat-killed forms) were also assessed for their ability to inhibit adhesion of selected pathogens to Caco-2 cells. The largest inhibition zones (the diffusion method) were connected with the usage of whole bacteria cultures (WBC), also high and moderate with cell-free supernatant (CFS) and the lowest with cell-free neutralized supernatant (CNS). The highest antagonistic activity of Lactobacillus strains was observed against L. monocytogenes strains, moderate activity against Salmonella, Shigella, Escherichia coli, Pseudomonas and, the lowest against S.aureus, Bacillus and Enterococcus. The inhibition of adhesion to Caco-2 cells was very high in the case of E. coli, Salmonella and L. monocytogenes, and moderate in the case of S.aureus. On average, the inhibition effect was higher when pathogenic bacteria were treated by WBC, than heat-killed Lactobacillus. Although, in most samples, the effect was not significantly different (P> 0.05). The strains Lb. brevis O24 and Lb. rhamnosus K3 showed the biggest overall antimicrobial properties, and were most effective in adherence inhibition of investigated indicator strains. These bacteria or their metabolites can be used for the production of various foods or pharmaceutical products.
Collapse
Affiliation(s)
- Dorota Zielińska
- Department of Food Gastronomy and Food Hygiene, Institute of Human Nutrition Sciences, Warsaw University of Life Science - SGGW, Nowoursynowska 159, 02-776 Warsaw, Poland
| | - Anna Łepecka
- Department of Food Gastronomy and Food Hygiene, Institute of Human Nutrition Sciences, Warsaw University of Life Science - SGGW, Nowoursynowska 159, 02-776 Warsaw, Poland
| | - Aleksandra Ołdak
- Department of Food Gastronomy and Food Hygiene, Institute of Human Nutrition Sciences, Warsaw University of Life Science - SGGW, Nowoursynowska 159, 02-776 Warsaw, Poland
| | - Ewa Długosz
- Department of Preclinical Sciences, Institute of Veterinary Medicine, Warsaw University of Life Science - SGGW, Nowoursynowska 159, 02-776 Warsaw, Poland
| | - Danuta Kołożyn-Krajewska
- Department of Food Gastronomy and Food Hygiene, Institute of Human Nutrition Sciences, Warsaw University of Life Science - SGGW, Nowoursynowska 159, 02-776 Warsaw, Poland
| |
Collapse
|
50
|
Sevin S, Karaca B, Haliscelik O, Kibar H, OmerOglou E, Kiran F. Postbiotics secreted by Lactobacillus sakei EIR/CM-1 isolated from cow milk microbiota, display antibacterial and antibiofilm activity against ruminant mastitis-causing pathogens. ITALIAN JOURNAL OF ANIMAL SCIENCE 2021. [DOI: 10.1080/1828051x.2021.1958077] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/20/2022]
Affiliation(s)
- Sedat Sevin
- Department of Pharmacology and Toxicology, Ankara University, Ankara, Turkey
- The Company of Sentezfarma, Ankara University Technopolis, Ankara, Turkey
| | - Basar Karaca
- Microbiology Research Laboratory, Department of Biology, Ankara University, Ankara, Turkey
| | - Ozan Haliscelik
- Pharmabiotic Technologies Research Laboratory, Department of Biology, Ankara University, Ankara, Turkey
| | - Hazal Kibar
- Pharmabiotic Technologies Research Laboratory, Department of Biology, Ankara University, Ankara, Turkey
| | - Emine OmerOglou
- Pharmabiotic Technologies Research Laboratory, Department of Biology, Ankara University, Ankara, Turkey
| | - Fadime Kiran
- Pharmabiotic Technologies Research Laboratory, Department of Biology, Ankara University, Ankara, Turkey
| |
Collapse
|