1
|
Piazzesi A, Scanu M, Ciprandi G, Putignani L. Modulations of the skin microbiome in skin disorders: A narrative review from a wound care perspective. Int Wound J 2024; 21:e70087. [PMID: 39379177 PMCID: PMC11461044 DOI: 10.1111/iwj.70087] [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: 06/26/2024] [Revised: 09/19/2024] [Accepted: 09/19/2024] [Indexed: 10/10/2024] Open
Abstract
The cutaneous microbiome represents a highly dynamic community of bacteria, fungi and viruses. Scientific evidence, particularly from the last two decades, has revealed that these organisms are far from being inconsequential microscopic hitchhikers on the human body, nor are they all opportunistic pathogens waiting for the chance to penetrate the skin barrier and cause infection. In this review, we will describe how dermatological diseases have been found to be associated with disruptions and imbalances in the skin microbiome and how this new evidence had shaped the diagnosis and clinical practice relating to these disorders. We will identify the microbial agents which have been found to directly exacerbate skin diseases, as well as those which can ameliorate many of the symptoms associated with dermatological disorders. Furthermore, we will discuss the studies which suggest that bacteriotherapy, either by topical use of probiotics or by bacteria-derived compounds, can rectify skin microbial imbalances, thereby offering a promising alternative to antibiotic treatment and reducing the risks of antibiotic resistance.
Collapse
Affiliation(s)
- Antonia Piazzesi
- Immunology, Rheumatology and Infectious Diseases Research Unit, Unit of the MicrobiomeBambino Gesù Children's Hospital, IRCCSRomeItaly
| | - Matteo Scanu
- Immunology, Rheumatology and Infectious Diseases Research Unit, Unit of the MicrobiomeBambino Gesù Children's Hospital, IRCCSRomeItaly
| | - Guido Ciprandi
- Research Institute Division of Plastic and Maxillofacial Surgery, Department of SurgeryBambino Gesu' Children's Hospital, IRCCSRomeItaly
| | - Lorenza Putignani
- Unit of Microbiology and Diagnostic Immunology, Unit of Microbiomics; and Immunology, Rheumatology and Infectious Diseases Research Unit, Unit of the MicrobiomeBambino Gesù Children's Hospital, IRCCSRomeItaly
| |
Collapse
|
2
|
Altves S, Celik FS, Gunes CE, Bilecen K. Exploring the therapeutic potential of Leuconostoc mesenteroides lysates in wound healing and immune modulation on keratinocyte cells. Arch Dermatol Res 2024; 316:548. [PMID: 39162738 DOI: 10.1007/s00403-024-03324-2] [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: 06/26/2024] [Revised: 06/26/2024] [Accepted: 08/05/2024] [Indexed: 08/21/2024]
Abstract
The skin, being the body's largest organ, primarily functions as a formidable defense mechanism against potential microbial infections. The skin's microbiota, consisting of a complex assembly of microorganisms, exerts a pivotal influence on skin homeostasis by modulating keratinocytes and their cytokine secretion, thereby playing an integral role in promoting optimal cutaneous health. Leuconostoc mesenteroides finds extensive application in the production of fermented foods and bacteriocins. Empirical studies validate the effectiveness of L. mesenteroides treatments in enhancing immune function and demonstrating notable antioxidant characteristics. This study investigates the potential of L. mesenteroides in improving skin health and wound healing. It also aims to comprehend their impact on wound healing markers, cytokine production, and cell cycle regulation compared to ferulic acid, known for its wound healing effects. Our findings indicate that L. mesenteroides lysate possesses antibacterial properties against Staphylococcus aureus and Pseudomonas aeruginosa, along with the ability to mitigate their toxic effects in a pathogen-simulating model employing HaCaT keratinocyte cells. Additionally, the lysate demonstrated noteworthy wound closure after a 24-hour treatment, along with a significant reduction in interleukin-6 levels and oxidative stress index. Modulation of the cell cycle is evident by decreasing G0/G1 phases and increasing S and G2/M phases and enhanced expression of wound healing marker genes and proteins CDH1. In conclusion, L. mesenteroides lysate exhibits immune-modulating and antibacterial properties, offering potential alternatives to conventional treatments for various skin conditions. These findings contribute to the exploration of innovative approaches to enhancing human life through skin health and wound healing.
Collapse
Affiliation(s)
- Safaa Altves
- Science and Technology Research and Application Center (BITAM), Necmettin Erbakan University, Konya, Türkiye.
- Department of Medical Biology, Faculty of Medicine, Necmettin Erbakan University, Konya, Türkiye.
| | - Fatma Secer Celik
- Department of Medical Biology, Faculty of Medicine, Ankara Medipol University, Ankara, Türkiye
| | - Canan Eroglu Gunes
- Department of Medical Biology, Faculty of Medicine, Necmettin Erbakan University, Konya, Türkiye
| | - Kivanc Bilecen
- Department of Molecular Biology and Genetics, Faculty of Agriculture and Natural Sciences, Konya Food and Agriculture University, Konya, Türkiye
| |
Collapse
|
3
|
Dinić M, Burgess JL, Lukić J, Catanuto P, Radojević D, Marjanović J, Verpile R, Thaller SR, Gonzalez T, Golić N, Strahinić I, Tomic-Canic M, Pastar I. Postbiotic lactobacilli induce cutaneous antimicrobial response and restore the barrier to inhibit the intracellular invasion of Staphylococcus aureus in vitro and ex vivo. FASEB J 2024; 38:e23801. [PMID: 39018106 PMCID: PMC11258854 DOI: 10.1096/fj.202400054rr] [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: 01/08/2024] [Revised: 06/20/2024] [Accepted: 06/25/2024] [Indexed: 07/18/2024]
Abstract
Intracellular pathogens including Staphylococcus aureus contribute to the non-healing phenotype of chronic wounds. Lactobacilli, well known as beneficial bacteria, are also reported to modulate the immune system, yet their role in cutaneous immunity remains largely unknown. We explored the therapeutic potential of bacteria-free postbiotics, bioactive lysates of lactobacilli, to reduce intracellular S. aureus colonization and promote healing. Fourteen postbiotics derived from various lactobacilli species were screened, and Latilactobacillus curvatus BGMK2-41 was selected for further analysis based on the most efficient ability to reduce intracellular infection by S. aureus diabetic foot ulcer clinical isolate and S. aureus USA300. Treatment of both infected keratinocytes in vitro and infected human skin ex vivo with BGMK2-41 postbiotic cleared S. aureus. Keratinocytes treated in vitro with BGMK2-41 upregulated expression of antimicrobial response genes, of which DEFB4, ANG, and RNASE7 were also found upregulated in treated ex vivo human skin together with CAMP exclusively upregulated ex vivo. Furthermore, BGMK2-41 postbiotic treatment has a multifaceted impact on the wound healing process. Treatment of keratinocytes stimulated cell migration and the expression of tight junction proteins, while in ex vivo human skin BGMK2-41 increased expression of anti-inflammatory cytokine IL-10, promoted re-epithelialization, and restored the epidermal barrier via upregulation of tight junction proteins. Together, this provides a potential therapeutic approach for persistent intracellular S. aureus infections.
Collapse
Affiliation(s)
- Miroslav Dinić
- Wound Healing and Regenerative Medicine Research Program, Dr. Phillip Frost Department of Dermatology and Cutaneous Surgery, University of Miami Miller School of Medicine, Miami, FL, USA
- Group for Probiotics and Microbiota-Host Interaction, Laboratory for Molecular Microbiology, Institute of Molecular Genetics and Genetic Engineering, University of Belgrade, Belgrade, Serbia
| | - Jamie L. Burgess
- Wound Healing and Regenerative Medicine Research Program, Dr. Phillip Frost Department of Dermatology and Cutaneous Surgery, University of Miami Miller School of Medicine, Miami, FL, USA
- Molecular and Cellular Pharmacology Graduate Program, University of Miami Miller School of Medicine, Miami FL, USA
| | - Jovanka Lukić
- Group for Probiotics and Microbiota-Host Interaction, Laboratory for Molecular Microbiology, Institute of Molecular Genetics and Genetic Engineering, University of Belgrade, Belgrade, Serbia
| | - Paola Catanuto
- Wound Healing and Regenerative Medicine Research Program, Dr. Phillip Frost Department of Dermatology and Cutaneous Surgery, University of Miami Miller School of Medicine, Miami, FL, USA
| | - Dušan Radojević
- Group for Probiotics and Microbiota-Host Interaction, Laboratory for Molecular Microbiology, Institute of Molecular Genetics and Genetic Engineering, University of Belgrade, Belgrade, Serbia
| | - Jelena Marjanović
- Wound Healing and Regenerative Medicine Research Program, Dr. Phillip Frost Department of Dermatology and Cutaneous Surgery, University of Miami Miller School of Medicine, Miami, FL, USA
| | - Rebecca Verpile
- Wound Healing and Regenerative Medicine Research Program, Dr. Phillip Frost Department of Dermatology and Cutaneous Surgery, University of Miami Miller School of Medicine, Miami, FL, USA
| | - Seth R. Thaller
- DeWitt Daughtry Family Department of Surgery, Division of Plastic Surgery, University of Miami Miller School of Medicine, Miami, FL, USA
| | - Tammy Gonzalez
- Wound Healing and Regenerative Medicine Research Program, Dr. Phillip Frost Department of Dermatology and Cutaneous Surgery, University of Miami Miller School of Medicine, Miami, FL, USA
| | - Nataša Golić
- Group for Probiotics and Microbiota-Host Interaction, Laboratory for Molecular Microbiology, Institute of Molecular Genetics and Genetic Engineering, University of Belgrade, Belgrade, Serbia
| | - Ivana Strahinić
- Group for Probiotics and Microbiota-Host Interaction, Laboratory for Molecular Microbiology, Institute of Molecular Genetics and Genetic Engineering, University of Belgrade, Belgrade, Serbia
| | - Marjana Tomic-Canic
- Wound Healing and Regenerative Medicine Research Program, Dr. Phillip Frost Department of Dermatology and Cutaneous Surgery, University of Miami Miller School of Medicine, Miami, FL, USA
- Molecular and Cellular Pharmacology Graduate Program, University of Miami Miller School of Medicine, Miami FL, USA
| | - Irena Pastar
- Wound Healing and Regenerative Medicine Research Program, Dr. Phillip Frost Department of Dermatology and Cutaneous Surgery, University of Miami Miller School of Medicine, Miami, FL, USA
| |
Collapse
|
4
|
Maslova E, EisaianKhongi L, Rigole P, Coenye T, McCarthy RR. Carbon source competition within the wound microenvironment can significantly influence infection progression. NPJ Biofilms Microbiomes 2024; 10:52. [PMID: 38918415 PMCID: PMC11199515 DOI: 10.1038/s41522-024-00518-4] [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: 11/30/2023] [Accepted: 05/08/2024] [Indexed: 06/27/2024] Open
Abstract
It is becoming increasingly apparent that commensal skin bacteria have an important role in wound healing and infection progression. However, the precise mechanisms underpinning many of these probiotic interactions remain to be fully uncovered. In this work, we demonstrate that the common skin commensal Cutibacterium acnes can limit the pathogenicity of the prevalent wound pathogen Pseudomonas aeruginosa in vivo. We show that this impact on pathogenicity is independent of any effect on growth, but occurs through a significant downregulation of the Type Three Secretion System (T3SS), the primary toxin secretion system utilised by P. aeruginosa in eukaryotic infection. We also show a downregulation in glucose acquisition systems, a known regulator of the T3SS, suggesting that glucose availability in a wound can influence infection progression. C. acnes is well known as a glucose fermenting organism, and we demonstrate that topically supplementing a wound with glucose reverses the probiotic effects of C. acnes. This suggests that introducing carbon source competition within the wound microenvironment may be an effective way to prevent or limit wound infection.
Collapse
Affiliation(s)
- Evgenia Maslova
- Division of Biosciences, Department of Life Sciences, College of Health and Life Sciences, Brunel University London, Uxbridge, UK
| | - Lara EisaianKhongi
- Division of Biosciences, Department of Life Sciences, College of Health and Life Sciences, Brunel University London, Uxbridge, UK
| | - Petra Rigole
- Laboratory of Pharmaceutical Microbiology, Ghent University, 9000, Ghent, Belgium
| | - Tom Coenye
- Laboratory of Pharmaceutical Microbiology, Ghent University, 9000, Ghent, Belgium
| | - Ronan R McCarthy
- Division of Biosciences, Department of Life Sciences, College of Health and Life Sciences, Brunel University London, Uxbridge, UK.
| |
Collapse
|
5
|
Alhubail M, McBain AJ, O'Neill CA. A survey of multiple candidate probiotic bacteria reveals specificity in the ability to modify the effects of key wound pathogens. Microbiol Spectr 2024; 12:e0034724. [PMID: 38700333 PMCID: PMC11237428 DOI: 10.1128/spectrum.00347-24] [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: 02/06/2024] [Accepted: 04/12/2024] [Indexed: 05/05/2024] Open
Abstract
We have evaluated the inhibitory effects of supernatants and lysates derived from several candidate probiotics, on the growth and biofilm formation of wound pathogens, and their ability to protect human primary epidermal keratinocytes from the toxic effects of pathogens. Supernatants (neutralized and non-neutralized) and lysates (via sonication) from Lactiplantibacillus plantarum, Limosilactobacillus reuteri, Bifidobacterium longum, Lacticaseibacillus rhamnosus GG, and Escherichia coli Nissle 1917 were tested for their inhibitory effects against Staphylococcus aureus, Streptococcus pyogenes, Escherichia coli, Pseudomonas aeruginosa, and Acinetobacter baumanni. The supernatants of L. plantarum, L. rhamnosus, B. longum, and L. rhamnosus GG reduced the growth of S. aureus, E. coli, and A. baumanni. B. longum additionally inhibited P. aeruginosa growth. However, neutralized Lactobacillus supernatants did not inhibit growth and in some cases were stimulatory. Lysates of L. plantarum and L. reuteri inhibited S. pyogenes while B. longum lysates inhibited E. coli and S. aureus growth. E. coli Nissle 1917 lysates enhanced the growth of S. pyogenes and P. aeruginosa. Biofilm formation by E. coli was reduced by lysates of L. reuteri and neutralized supernatants of all candidate probiotics. P. aeruginosa biofilm formation was reduced by E. coli Nissle supernatant but increased by L. plantarum, L. reuteri, and Bifidobacterium longum lysates. L. reuteri decreased the toxic effects of S. aureus on keratinocytes while E. coli Nissle 1917 lysates protected keratinocytes from S. pyogenes toxicity. In conclusion, lactobacilli and E. coli Nissle lysates confer inhibitory effects on pathogenic growth independently of acidification and may beneficially alter the outcome of interactions between host cell-pathogen in a species-specific manner.IMPORTANCEOne of the attributes of probiotics is their ability to inhibit pathogens. For this reason, many lactobacilli have been investigated for their effects as potential topical therapeutics against skin pathogens. However, this field is in its infancy. Even though probiotics are known to be safe when taken orally, the potential safety concerns when applied to potentially compromised skin are unknown. For this reason, we believe that extracts of probiotics will offer advantages over the use of live bacteria. In this study, we have surveyed five candidate probiotics, when used as extracts, in terms of their effects against common wound pathogens. Our data demonstrate that some probiotic extracts promote the growth of pathogens and highlight the need for careful selection of species and strains when probiotics are to be used topically.
Collapse
Affiliation(s)
- Muna Alhubail
- Faculty of Biology, Medicine and Health, School of Biological Sciences, The University of Manchester, Manchester, United Kingdom
| | - Andrew J. McBain
- Faculty of Biology, School of Health Sciences, Medicine and Health, The University of Manchester, Manchester, United Kingdom
| | - Catherine A. O'Neill
- Faculty of Biology, Medicine and Health, School of Biological Sciences, The University of Manchester, Manchester, United Kingdom
| |
Collapse
|
6
|
Dubey AK, Sharma M, Parul, Raut S, Gupta P, Khatri N. Healing wounds, defeating biofilms: Lactiplantibacillus plantarum in tackling MRSA infections. Front Microbiol 2023; 14:1284195. [PMID: 38116526 PMCID: PMC10728654 DOI: 10.3389/fmicb.2023.1284195] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2023] [Accepted: 11/01/2023] [Indexed: 12/21/2023] Open
Abstract
Introduction Methicillin-resistant Staphylococcus aureus (MRSA) infections are well-known hospital-borne infections and are a major contributing factor to global health concerns of antimicrobial resistance due to the formation of biofilms. Probiotics are known to assist in the healing of wounds through immunomodulation and also possess anti-pathogen properties via competitive inhibition. The probiotic bacterium, Lactiplantibacillus plantarum MTCC 2621 and its cell-free supernatant (Lp2621) have previously been reported to have antibacterial, excellent antioxidant, and wound healing activity in in vitro conditions and wounds contaminated with S. aureus in mice. Methods In the current study, we evaluated its anti-MRSA, biofilm inhibition and eradication efficacy, immunomodulatory activity in THP-1 cells, and wound healing potential in wounds contaminated with MRSA infection in mice. Results In agar well diffusion assay, Lp2621 showed anti-MRSA activity and revealed dose-dependent inhibition and eradication of biofilm by crystal violet assay as well as by Confocal Scanning Laser Microscopy (CLSM) analysis. Further, Lp2621 showed immunomodulatory activity at varied concentrations as measured by IL-6 and IL-10 gene expression in THP-1 cells. Similar findings were observed in serum samples of mice after treatment of excision wound contaminated with MRSA infection by Lp2621 gel, as evident by expression of IL-6 (pro-inflammatory) and IL-10 (anti-inflammatory) cytokines. Conclusions Overall, our results show that Lp2621 has potent anti-MRSA and antioxidant properties and can prevent and eliminate biofilm formation. It also showed promise when applied to mice with MRSA-infected wounds.
Collapse
Affiliation(s)
- Ashish Kumar Dubey
- IMTech Centre for Animal Resources & Experimentation (iCARE), CSIR-Institute of Microbial Technology (IMTECH), Chandigarh, India
- Academy of Scientific and Innovative Research (AcSIR), Ghaziabad, Uttar Pradesh, India
| | - Mohini Sharma
- IMTech Centre for Animal Resources & Experimentation (iCARE), CSIR-Institute of Microbial Technology (IMTECH), Chandigarh, India
| | - Parul
- IMTech Centre for Animal Resources & Experimentation (iCARE), CSIR-Institute of Microbial Technology (IMTECH), Chandigarh, India
| | - Sachin Raut
- IMTech Centre for Animal Resources & Experimentation (iCARE), CSIR-Institute of Microbial Technology (IMTECH), Chandigarh, India
- Academy of Scientific and Innovative Research (AcSIR), Ghaziabad, Uttar Pradesh, India
| | - Pawan Gupta
- Academy of Scientific and Innovative Research (AcSIR), Ghaziabad, Uttar Pradesh, India
- Department of Molecular Biology, CSIR-Institute of Microbial Technology (IMTECH), Chandigarh, India
| | - Neeraj Khatri
- IMTech Centre for Animal Resources & Experimentation (iCARE), CSIR-Institute of Microbial Technology (IMTECH), Chandigarh, India
- Academy of Scientific and Innovative Research (AcSIR), Ghaziabad, Uttar Pradesh, India
| |
Collapse
|
7
|
Gruber JV, Holtz R. Living, quiescent Lactobacillus plantarum Lp90 probiotic, delivered topically to full thickness tissues in vitro via a just-add-water cream delivery system, stimulates the expression of elastin protein. J Cosmet Dermatol 2023; 22:2852-2860. [PMID: 37470208 DOI: 10.1111/jocd.15927] [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: 04/22/2023] [Revised: 06/08/2023] [Accepted: 06/29/2023] [Indexed: 07/21/2023]
Abstract
INTRODUCTION Delivering living probiotics to the skin can be challenging as most water-containing cosmetic products require preservatives to maintain product stability. A recently introduced powdered technology [Stratabiosys™, Vantage Personal Care] allows for quiescent probiotic powders to be stored for extended periods of time. The powders can then be reconstituted to creams at the point of use by adding water and mixing and were examined in vitro on reconstructed human full thickness tissues to see if the probiotic had any influence of several important biomolecules expressed in the skin. MATERIALS AND METHODS A probiotic powder containing 200 M CFU/gram of living quiescent Lactobacillus plantarum Lp90 was reconstituted to a cream by adding ultrapure water and gently mixing the components at room temperature to quickly produce a cream. The resulting cream was tested topically on Epiderm® Full Thickness Tissues by treating the tissues for 24 h, removing the cream with a PBS rinse and then repeating the treatment for another 24 h. The resulting tissues were examined for four strategically important skin biomolecules including Type 1A collagen, elastin, filaggrin and hyaluronic acid. The probiotic-containing powder was tested against untreated tissues and powders not containing probiotics and powders containing measured amounts of one of two cryoprotectants known to be used to maintain the integrity of the quiescent probiotics during drying of the quiescent probiotic powders. RESULTS It was found that topical treatment on Epiderm® tissues with creams containing 2 M (1%), 4 M (2%) and 6 M (3%) CFU/gram prepared from a base powder containing 200 M CFU/gram of Lactobacillus plantarum Lp90 stimulated elastin expression in a dose dependent fashion. There was no effect on the other biomolecules examined in the studies. In addition, it was found that creams made from powders containing only the known cryoprotectants, not bacteria, had no influence on elastin expression. CONCLUSION The results of this study demonstrate that topical delivery of probiotics is possible from powders containing quiescent probiotic powders converted to creams just prior to application to the tissues. In the case of a powder containing Lactobacillus plantarum Lp90, topical application significantly increased expression of elastin in the skin replicants after 48 h of exposure to the cream made with the probiotic. The elastin-stimulating effects are not coming from the oligosaccharide cryoprotectants used to maintain the probiotic powders in their quiescent, dried state. The results indicate that it is the living Lactobacillus plantarum probiotic that is stimulating the elastin expression in the skin tissues.
Collapse
Affiliation(s)
| | - Robert Holtz
- Bioinnovation Laboratories, Inc., Lakewood, Colorado, USA
| |
Collapse
|
8
|
Menni A, Moysidis M, Tzikos G, Stavrou G, Tsetis JK, Shrewsbury AD, Filidou E, Kotzampassi K. Looking for the Ideal Probiotic Healing Regime. Nutrients 2023; 15:3055. [PMID: 37447381 DOI: 10.3390/nu15133055] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2023] [Revised: 06/30/2023] [Accepted: 07/05/2023] [Indexed: 07/15/2023] Open
Abstract
Wound healing is a multi-factorial response to tissue injury, aiming to restore tissue continuity. Numerous recent experimental and clinical studies clearly indicate that probiotics are applied topically to promote the wound-healing process. However, the precise mechanism by which they contribute to healing is not yet clear. Each strain appears to exert a distinctive, even multi-factorial action on different phases of the healing process. Given that a multi-probiotic formula exerts better results than a single strain, the pharmaceutical industry has embarked on a race for the production of a formulation containing a combination of probiotics capable of playing a role in all the phases of the healing process. Hence, the object of this review is to describe what is known to date of the distinctive mechanisms of each of the most studied probiotic strains in order to further facilitate research toward the development of combinations of strains and doses, covering the whole spectrum of healing. Eleven probiotic species have been analyzed, the only criterion of inclusion being a minimum of two published research articles.
Collapse
Affiliation(s)
- Alexandra Menni
- Department of Surgery, Aristotle University of Thessaloniki, 54636 Thessaloniki, Greece
| | - Moysis Moysidis
- Department of Surgery, Aristotle University of Thessaloniki, 54636 Thessaloniki, Greece
| | - Georgios Tzikos
- Department of Surgery, Aristotle University of Thessaloniki, 54636 Thessaloniki, Greece
| | - George Stavrou
- Department of Colorectal Surgery, Addenbrooke's Hospital, Cambridge CB2 2QQ, UK
| | | | - Anne D Shrewsbury
- Department of Surgery, Aristotle University of Thessaloniki, 54636 Thessaloniki, Greece
| | - Eirini Filidou
- Laboratory of Pharmacology, Faculty of Medicine, Democritus University of Thrace, 68100 Alexandroupolis, Greece
| | - Katerina Kotzampassi
- Department of Surgery, Aristotle University of Thessaloniki, 54636 Thessaloniki, Greece
| |
Collapse
|
9
|
Maslova E, Osman S, McCarthy RR. Using the Galleria mellonella burn wound and infection model to identify and characterize potential wound probiotics. MICROBIOLOGY (READING, ENGLAND) 2023; 169:001350. [PMID: 37350463 PMCID: PMC10333784 DOI: 10.1099/mic.0.001350] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/14/2023] [Accepted: 06/07/2023] [Indexed: 06/24/2023]
Abstract
Burn wound infection is the leading cause of mortality among burn wound patients. One of the most commonly isolated bacterial burn wound pathogens is Pseudomonas aeruginosa, a notorious nosocomial multidrug-resistant pathogen. As a consequence of its recalcitrance to frontline antibiotic therapy, there is an urgent need to develop alternative treatment avenues to tackle this pathogen. One potential alternative infection prevention measure is to seed the wound bed with probiotic bacteria. Several species of Lactobacillus, a common commensal bacterium, have been previously reported to display growth inhibition activity against wound pathogens. Various species of this genus have also been shown to augment the wound healing process, which makes it a promising potential therapeutic agent. Due to the complexity of the burn wound trauma and burn wound infection, an in vivo model is required for the development of novel therapeutics. There are multiple in vivo models that are currently available, the most common among them being the murine model. However, mammalian burn wound infection models are logistically challenging, do not lend themselves to screening approaches and come with significant concerns around ethics and animal welfare. Recently, an invertebrate burn wound and infection model using G. mellonella has been established. This model addresses several of the challenges of more advanced animal models, such as affordability, maintenance and reduced ethical concerns. This study validates the capacity of this model to screen for potential wound probiotics by demonstrating that a variety of Lactobacillus spp. can limit P. aeruginosa burn wound infection and improve survival.
Collapse
Affiliation(s)
- Evgenia Maslova
- Division of Biosciences, Department of Life Sciences, Centre of Inflammation Research and Translational Medicine, College of Health, Medicine and Life Sciences, College of Health and Life Sciences, Brunel University London, Uxbridge, UK
| | - Shanga Osman
- Division of Biosciences, Department of Life Sciences, Centre of Inflammation Research and Translational Medicine, College of Health, Medicine and Life Sciences, College of Health and Life Sciences, Brunel University London, Uxbridge, UK
| | - Ronan R. McCarthy
- Division of Biosciences, Department of Life Sciences, Centre of Inflammation Research and Translational Medicine, College of Health, Medicine and Life Sciences, College of Health and Life Sciences, Brunel University London, Uxbridge, UK
| |
Collapse
|
10
|
Yu Z, Chen J, Liu Y, Meng Q, Liu H, Yao Q, Song W, Ren X, Chen X. The role of potential probiotic strains Lactobacillus reuteri in various intestinal diseases: New roles for an old player. Front Microbiol 2023; 14:1095555. [PMID: 36819028 PMCID: PMC9932687 DOI: 10.3389/fmicb.2023.1095555] [Citation(s) in RCA: 12] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2022] [Accepted: 01/03/2023] [Indexed: 02/05/2023] Open
Abstract
Lactobacillus reuteri (L. reuteri), a type of Lactobacillus spp., is a gut symbiont that can colonize many mammals. Since it was first isolated in 1962, a multitude of research has been conducted to investigate its function and unique role in different diseases as an essential probiotic. Among these, the basic functions, beneficial effects, and underlying mechanisms of L. reuteri have been noticed and understood profoundly in intestinal diseases. The origins of L. reuteri strains are diverse, with humans, rats, and piglets being the most common. With numerous L. reuteri strains playing significant roles in different intestinal diseases, DSM 17938 is the most widely used in humans, especially in children. The mechanisms by which L. reuteri improves intestinal disorders include protecting the gut barrier, suppressing inflammation and the immune response, regulating the gut microbiota and its metabolism, and inhibiting oxidative stress. While a growing body of studies focused on L. reuteri, there are still many unknowns concerning its curative effects, clinical safety, and precise mechanisms. In this review, we initially interpreted the basic functions of L. reuteri and its related metabolites. Then, we comprehensively summarized its functions in different intestinal diseases, including inflammatory bowel disease, colorectal cancer, infection-associated bowel diseases, and pediatric intestinal disorders. We also highlighted some important molecules in relation to the underlying mechanisms. In conclusion, L. reuteri has the potential to exert a beneficial impact on intestinal diseases, which should be further explored to obtain better clinical application and therapeutic effects.
Collapse
Affiliation(s)
- Zihan Yu
- Department of Gastroenterology and Hepatology, Tianjin Medical University General Hospital, Tianjin, China,Tianjin Institute of Digestive Disease, Tianjin Medical University General Hospital, Tianjin, China
| | - Jihua Chen
- Department of Gastroenterology and Hepatology, Tianjin Medical University General Hospital, Tianjin, China,Tianjin Institute of Digestive Disease, Tianjin Medical University General Hospital, Tianjin, China
| | - Yaxin Liu
- Department of Gastroenterology and Hepatology, Tianjin Medical University General Hospital, Tianjin, China,Tianjin Institute of Digestive Disease, Tianjin Medical University General Hospital, Tianjin, China
| | - Qingguo Meng
- Department of Gastroenterology and Hepatology, Tianjin Medical University General Hospital, Tianjin, China,Tianjin Institute of Digestive Disease, Tianjin Medical University General Hospital, Tianjin, China
| | - Hang Liu
- Department of Gastroenterology and Hepatology, Tianjin Medical University General Hospital, Tianjin, China,Tianjin Institute of Digestive Disease, Tianjin Medical University General Hospital, Tianjin, China
| | - Qinyan Yao
- Department of Gastroenterology and Hepatology, Tianjin Medical University General Hospital, Tianjin, China,Tianjin Institute of Digestive Disease, Tianjin Medical University General Hospital, Tianjin, China
| | - Wenxuan Song
- Department of Gastroenterology and Hepatology, Tianjin Medical University General Hospital, Tianjin, China,Tianjin Institute of Digestive Disease, Tianjin Medical University General Hospital, Tianjin, China
| | - Xiangfeng Ren
- Department of Gastroenterology and Hepatology, Tianjin Medical University General Hospital, Tianjin, China,Tianjin Institute of Digestive Disease, Tianjin Medical University General Hospital, Tianjin, China
| | - Xin Chen
- Department of Gastroenterology and Hepatology, Tianjin Medical University General Hospital, Tianjin, China,Tianjin Institute of Digestive Disease, Tianjin Medical University General Hospital, Tianjin, China,*Correspondence: Xin Chen ✉
| |
Collapse
|
11
|
The Antimicrobial Effect of Various Single-Strain and Multi-Strain Probiotics, Dietary Supplements or Other Beneficial Microbes against Common Clinical Wound Pathogens. Microorganisms 2022; 10:microorganisms10122518. [PMID: 36557771 PMCID: PMC9781324 DOI: 10.3390/microorganisms10122518] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2022] [Revised: 12/09/2022] [Accepted: 12/13/2022] [Indexed: 12/24/2022] Open
Abstract
The skin is the largest organ in the human body and is colonized by a diverse microbiota that works in harmony to protect the skin. However, when skin damage occurs, the skin microbiota is also disrupted, and pathogens can invade the wound and cause infection. Probiotics or other beneficial microbes and their metabolites are one possible alternative treatment for combating skin pathogens via their antimicrobial effectiveness. The objective of our study was to evaluate the antimicrobial effect of seven multi-strain dietary supplements and eleven single-strain microbes that contain probiotics against 15 clinical wound pathogens using the agar spot assay, co-culturing assay, and agar well diffusion assay. We also conducted genera-specific and species-specific molecular methods to detect the DNA in the dietary supplements and single-strain beneficial microbes. We found that the multi-strain dietary supplements exhibited a statistically significant higher antagonistic effect against the challenge wound pathogens than the single-strain microbes and that lactobacilli-containing dietary supplements and single-strain microbes were significantly more efficient than the selected propionibacteria and bacilli. Differences in results between methods were also observed, possibly due to different mechanisms of action. Individual pathogens were susceptible to different dietary supplements or single-strain microbes. Perhaps an individual approach such as a 'probiogram' could be a possibility in the future as a method to find the most efficient targeted probiotic strains, cell-free supernatants, or neutralized cell-free supernatants that have the highest antagonistic effect against individual clinical wound pathogens.
Collapse
|
12
|
Saha UB, Saroj SD. Lactic acid bacteria: prominent player in the fight against human pathogens. Expert Rev Anti Infect Ther 2022; 20:1435-1453. [PMID: 36154442 DOI: 10.1080/14787210.2022.2128765] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
INTRODUCTION The human microbiome is a unique repository of diverse bacteria. Over 1000 microbial species reside in the human gut, which predominantly influences the host's internal environment and plays a significant role in host health. Lactic acid bacteria have long been employed for multiple purposes, ranging from food to medicines. Lactobacilli, which are often used in commercial food fermentation, have improved to the point that they might be helpful in medical applications. AREAS COVERED This review summarises various clinical and experimental evidence on efficacy of lactobacilli in treating a wide range of infections. Both laboratory based and clinical studies have been discussed. EXPERT OPINION Lactobacilli are widely accepted as safe biological treatments and host immune modulators (GRAS- Generally regarded as safe) by the US Food and Drug Administration and Qualified Presumption of Safety. Understanding the molecular mechanisms of lactobacilli in the treatment and pathogenicity of bacterial infections can help with the prediction and development of innovative therapeutics aimed at pathogens which have gained resistance to antimicrobials. To formulate effective lactobacilli based therapy significant research on the effectiveness of different lactobacilli strains and its association with demographic distribution is required. Also, the side effects of such therapy needs to be evaluated.
Collapse
Affiliation(s)
- Ujjayni B Saha
- Symbiosis School of Biological Sciences, Symbiosis International (Deemed University), Symbiosis Knowledge Village, Lavale, Pune, India
| | - Sunil D Saroj
- Symbiosis School of Biological Sciences, Symbiosis International (Deemed University), Symbiosis Knowledge Village, Lavale, Pune, India
| |
Collapse
|
13
|
Lizardo M, Magalhães RM, Tavaria FK. Probiotic Adhesion to Skin Keratinocytes and Underlying Mechanisms. BIOLOGY 2022; 11:1372. [PMID: 36138851 PMCID: PMC9495785 DOI: 10.3390/biology11091372] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/28/2022] [Revised: 09/08/2022] [Accepted: 09/14/2022] [Indexed: 11/17/2022]
Abstract
The effects of probiotics on the skin are not yet well understood. Their topical application and benefits derived thereafter have recently been investigated. Improvements in different skin disorders such as atopic dermatitis, acne, eczema, and psoriasis after their use have, however, been reported. One of the mechanisms through which such benefits are documented is by inhibiting colonization by skin pathogens. Bacterial adhesion is the first step for colonization to occur; therefore, to avoid pathogenic colonization, inhibiting adhesion is crucial. In this study, invasion and adhesion studies have been carried out using keratinocytes. These showed that Escherichia coli is not able to invade skin keratinocytes, but adhered to them. Lacticaseibacillus rhamnosus and Propioniferax innocua decreased the viable counts of the three pathogens under study. L. rhamnosus significantly inhibited S. aureus adhesion. P. innocua did not inhibit pathogenic bacteria adhesion, but when added simultaneously with S. aureus (competition assay) a significant adhesion reduction (1.12 ± 0.14 log10CFU/mL) was observed. Probiotic bacteria seem to use carbohydrates to adhere to the keratinocytes, while S. aureus uses proteins. Lacticaseibacillus rhamnosus showed promising results in pathogen inhibition in both in vitro and ex vivo experiments and can potentially be used as a reinforcement of conventional therapies for skin dysbiosis.
Collapse
Affiliation(s)
| | | | - Freni Kekhasharú Tavaria
- Centro de Biotecnologia e Química Fina–Laboratório Associado, Escola Superior de Biotecnologia, Universidade Católica Portuguesa/Porto, Rua Diogo Botelho, 1327, 4169-005 Porto, Portugal
| |
Collapse
|
14
|
Idrees M, Imran M, Atiq N, Zahra R, Abid R, Alreshidi M, Roberts T, Abdelgadir A, Tipu MK, Farid A, Olawale OA, Ghazanfar S. Probiotics, their action modality and the use of multi-omics in metamorphosis of commensal microbiota into target-based probiotics. Front Nutr 2022; 9:959941. [PMID: 36185680 PMCID: PMC9523698 DOI: 10.3389/fnut.2022.959941] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2022] [Accepted: 08/24/2022] [Indexed: 11/13/2022] Open
Abstract
This review article addresses the strategic formulation of human probiotics and allows the reader to walk along the journey that metamorphoses commensal microbiota into target-based probiotics. It recapitulates what are probiotics, their history, and the main mechanisms through which probiotics exert beneficial effects on the host. It articulates how a given probiotic preparation could not be all-encompassing and how each probiotic strain has its unique repertoire of functional genes. It answers what criteria should be met to formulate probiotics intended for human use, and why certain probiotics meet ill-fate in pre-clinical and clinical trials? It communicates the reasons that taint the reputation of probiotics and cause discord between the industry, medical and scientific communities. It revisits the notion of host-adapted strains carrying niche-specific genetic modifications. Lastly, this paper emphasizes the strategic development of target-based probiotics using host-adapted microbial isolates with known molecular effectors that would serve as better candidates for bioprophylactic and biotherapeutic interventions in disease-susceptible individuals.
Collapse
Affiliation(s)
- Maryam Idrees
- Department of Microbiology, Quaid-i-Azam University, Islamabad, Pakistan
- National Agricultural Research Centre (NARC), National Institute for Genomics and Advanced Biotechnology (NIGAB), Islamabad, Pakistan
| | - Muhammad Imran
- Department of Microbiology, Quaid-i-Azam University, Islamabad, Pakistan
| | - Naima Atiq
- Department of Microbiology, Quaid-i-Azam University, Islamabad, Pakistan
| | - Rabaab Zahra
- Department of Microbiology, Quaid-i-Azam University, Islamabad, Pakistan
| | - Rameesha Abid
- National Agricultural Research Centre (NARC), National Institute for Genomics and Advanced Biotechnology (NIGAB), Islamabad, Pakistan
- Department of Biotechnology, University of Sialkot, Sialkot, Pakistan
| | - Mousa Alreshidi
- Department of Biology, College of Science, University of Hail, Hail, Saudi Arabia
- Molecular Diagnostics and Personalized Therapeutics Unit, University of Ha’il, Ha’il, Saudi Arabia
| | - Tim Roberts
- Metabolic Research Group, Faculty of Science, School of Environmental and Life Sciences, The University of Newcastle, University Drive, Callaghan, NSW, Australia
| | - Abdelmuhsin Abdelgadir
- Department of Biology, College of Science, University of Hail, Hail, Saudi Arabia
- Molecular Diagnostics and Personalized Therapeutics Unit, University of Ha’il, Ha’il, Saudi Arabia
| | | | - Arshad Farid
- Gomal Center of Biochemistry and Biotechnology, Gomal University, Dera Ismail Khan, Pakistan
| | | | - Shakira Ghazanfar
- National Agricultural Research Centre (NARC), National Institute for Genomics and Advanced Biotechnology (NIGAB), Islamabad, Pakistan
| |
Collapse
|
15
|
Development and metabolic profiling of a postbiotic complex exhibiting antibacterial activity against skin microorganisms and anti-inflammatory effect on human keratinocytes. Food Sci Biotechnol 2022; 31:1325-1334. [PMID: 35992320 PMCID: PMC9385932 DOI: 10.1007/s10068-022-01123-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2022] [Revised: 06/01/2022] [Accepted: 06/16/2022] [Indexed: 11/04/2022] Open
Abstract
Beyond probiotics, the interest in the application of postbiotics to various fields has been growing. We aimed to develop a novel postbiotic complex (PC) with antibacterial and anti-inflammatory properties. Through antibacterial activity testing against Staphylococcus aureus or Cutibacterium acnes, a PC [a mixture of cell-free supernatants (postbiotics) from probiotic Lactobacillus helveticus (HY7801) and Lactococcus lactis (HY449)] was developed. Anti-inflammatory activity of the PC was investigated using HaCaT keratinocytes treated with S. aureus or C. acnes. PC significantly decreased IL-8 levels and increased hyaluronic acid levels in HaCaT cells cultured with S. aureus or C. acnes. GC-MS based metabolic profiling suggested 2-hydroxyisocaproic acid, hypoxanthine, succinic acid, ornithine, and γ-aminobutyric acid as potential contributing metabolites for the antibacterial and anti-inflammatory effects of PC. The PC developed in this study could be utilized in food, cosmetics, and pharmaceutical products as an alternative or complementary resources of probiotics. Supplementary Information The online version contains supplementary material available at 10.1007/s10068-022-01123-x.
Collapse
|
16
|
Smythe P, Efthimiou G. In Silico Genomic and Metabolic Atlas of Limosilactobacillus reuteri DSM 20016: An Insight into Human Health. Microorganisms 2022; 10:microorganisms10071341. [PMID: 35889060 PMCID: PMC9320016 DOI: 10.3390/microorganisms10071341] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2022] [Revised: 06/29/2022] [Accepted: 06/30/2022] [Indexed: 11/23/2022] Open
Abstract
Probiotics are bacterial strains that are known to provide host health benefits. Limosilactobacillus reuteri is a well-documented lactic acid bacterium that has been cultured from numerous human sites. The strain investigated was L. reuteri DSM 20016, which has been found to produce useful metabolites. The strain was explored using genomic and proteomic tools, manual searches, and databases, including KEGG, STRING, BLAST Sequence Similarity Search, and UniProt. This study located over 200 key genes that were involved in human health benefit pathways. L. reuteri DSM 20016 has metabolic pathways to produce acetate, propionate, and lactate, and there is evidence of a pathway for butanoate production through a FASII mechanism. The bacterium produces histamine through the hdc operon, which may be able to suppress proinflammatory TNF, and the bacterium also has the ability to synthesize folate and riboflavin, although whether they are secreted is yet to be explored. The strain can bind to human Caco2 cells through srtA, mapA/cnb, msrB, and fbpA and can compete against enteric bacteria using reuterin, which is an antimicrobial that induces oxidative stress. The atlas could be used for designing metabolic engineering approaches to improve beneficial metabolite biosynthesis and better probiotic-based cures.
Collapse
Affiliation(s)
- Paisleigh Smythe
- Centre for Atherothrombosis and Metabolic Disease, Hull York Medical School, Castle Hill Hospital, Daisy Building, Hull HU16 5JQ, UK;
| | - Georgios Efthimiou
- Department of Biomedical and Forensic Sciences, University of Hull, Cottingham Road, Hardy Building, Hull HU6 7RX, UK
- Correspondence: ; Tel.: +44-(0)1482-465970
| |
Collapse
|
17
|
El-Chami C, Choudhury R, Mohammedsaeed W, McBain AJ, Kainulainen V, Lebeer S, Satokari R, O'Neill CA. Multiple Proteins of Lacticaseibacillus rhamnosus GG Are Involved in the Protection of Keratinocytes From the Toxic Effects of Staphylococcus aureus. Front Microbiol 2022; 13:875542. [PMID: 35633665 PMCID: PMC9134637 DOI: 10.3389/fmicb.2022.875542] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2022] [Accepted: 03/22/2022] [Indexed: 11/21/2022] Open
Abstract
We have previously shown that lysates of Lacticaseibacillus rhamnosus GG confer protection to human keratinocytes against Staphylococcus aureus. L. rhamnosus GG inhibits the growth of S. aureus as well as competitively excludes and displaces the pathogen from keratinocytes. In this study, we have specifically investigated the anti-adhesive action. We have tested the hypothesis that this activity is due to quenching of S. aureus binding sites on keratinocytes by molecules within the Lacticaseibacillus lysate. Trypsinisation or heat treatment removed the protective effect of the lysate suggesting the involvement of proteins as effector molecules. Column separation of the lysate and analysis of discrete fractions in adhesion assays identified a fraction of moderate hydrophobicity that possessed all anti-adhesive functions. Immunoblotting demonstrated that this fraction contained the pilus protein, SpaC. Recombinant SpaC inhibited staphylococcal adhesion to keratinocytes in a dose-dependent manner and improved keratinocyte viability following challenge with viable S. aureus. However, SpaC did not confer the full anti-adhesive effects of the LGG lysate and excluded but did not displace S. aureus from keratinocytes. Further purification produced four protein-containing peaks (F1–F4). Of these, F4, which had the greatest column retention time, was the most efficacious in anti-staphylococcal adhesion and keratinocyte viability assays. Identification of proteins by mass spectrometry showed F4 to contain several known “moonlighting proteins”—i.e., with additional activities to the canonical function, including enolase, Triosephosphate isomerase (TPI), Glyceraldehyde 3 phosphate dehydrogenase (G3P) and Elongation factor TU (EF-Tu). Of these, only enolase and TPI inhibited S. aureus adhesion and protected keratinocytes viability in a dose-dependent manner. These data suggest that inhibition of staphylococcal binding by the L. rhamnosus GG lysate is mediated by SpaC and specific moonlight proteins.
Collapse
Affiliation(s)
- Cecile El-Chami
- Faculty of Biology, Medicine and Health, School of Biological Sciences, University of Manchester, Manchester, United Kingdom
| | - Rawshan Choudhury
- Faculty of Biology, Medicine and Health, School of Biological Sciences, University of Manchester, Manchester, United Kingdom
| | - Walaa Mohammedsaeed
- Faculty of Biology, Medicine and Health, School of Biological Sciences, University of Manchester, Manchester, United Kingdom
| | - Andrew J McBain
- Faculty of Biology, School of Health Sciences, Medicine and Health, University of Manchester, Manchester, United Kingdom
| | - Veera Kainulainen
- Faculty of Medicine, Human Microbiome Research Program, University of Helsinki, Helsinki, Finland
| | - Sarah Lebeer
- Department of Bioscience Engineering, University of Antwerp, Antwerp, Belgium
| | - Reetta Satokari
- Faculty of Medicine, Human Microbiome Research Program, University of Helsinki, Helsinki, Finland
| | - Catherine A O'Neill
- Faculty of Biology, Medicine and Health, School of Biological Sciences, University of Manchester, Manchester, United Kingdom
| |
Collapse
|
18
|
Mei L, Zhang D, Shao H, Hao Y, Zhang T, Zheng W, Ji Y, Ling P, Lu Y, Zhou Q. Injectable and Self-Healing Probiotics-Loaded Hydrogel for Promoting Superbacteria-Infected Wound Healing. ACS APPLIED MATERIALS & INTERFACES 2022; 14:20538-20550. [PMID: 35471815 DOI: 10.1021/acsami.1c23713] [Citation(s) in RCA: 43] [Impact Index Per Article: 21.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/14/2023]
Abstract
Superbacteria-induced skin wound infections are huge health challenges, resulting in significant financial and medical costs due to notable morbidity and mortality worldwide. Probiotics are found in the skin and are effective in treating bacterial infection, moderating the microbial dysbiosis and inflammation induced by pathogens, regulating the immune system, as well as even promoting tissue repair. However, improving their colonization efficiency and viability remains a large obstacle for proper applications. Inspired by probiotic therapy and the natural extracellular matrix structure, hyaluronate-adipic dihydrazide/aldehyde-terminated Pluronic F127/fucoidan hydrogels loaded with Lactobacillus rhamnosus (HPF@L.rha) with unique (bio)physicochemical characteristics were developed through the dynamic Schiff-base reaction for superbacteria-infected trauma management. The developed HPF@L.rha exhibit a shortened gelation time, enhanced mechanical strength, and excellent self-healing and liquid-absorption abilities. Importantly, their anti-superbacteria (Pseudomonas aeruginosa) effect was greatly increased in a dose-dependent fashion. Additionally, in vitro evaluation shows that the prepared HPF@L.rha containing appropriate probiotic concentrations (less than 1 × 107 CFU/mL) possess satisfactory cytocompatibility and blood compatibility. Further, compared to the HPF hydrogel, in vivo the hydrogel combined with probiotics significantly inhibits P. aeruginosa infection and inflammation, promotes the formation of re-epithelialization and collagen, and thus accelerates full-thickness superbacteria-infected wound repair, which is comparable to commercial Prontosan gel formulation. This work suggests that the combination of biomimicking hydrogels and probiotic therapy displays the great potential to manage superbug-infected trauma.
Collapse
Affiliation(s)
- Li Mei
- Institute for Translational Medicine, Department of Stomatology, The Affiliated Hospital of Qingdao University, Qingdao University, Qingdao 266003, China
- Tianjin Enterprise Key Laboratory for Application Research of Hyaluronic Acid, Tianjin 300038, China
- School of Stomatology, Qingdao University, Qingdao 266003, China
| | - Dongjie Zhang
- Department of Gastroenterology, The Affiliated Hospital of Qingdao University, Qingdao 266003, China
| | - Huarong Shao
- Key Laboratory of Biopharmaceuticals, Shandong Academy of Pharmaceutical Sciences, Jinan 250101, China
| | - Yuanping Hao
- Institute for Translational Medicine, Department of Stomatology, The Affiliated Hospital of Qingdao University, Qingdao University, Qingdao 266003, China
| | - Ting Zhang
- Institute for Translational Medicine, Department of Stomatology, The Affiliated Hospital of Qingdao University, Qingdao University, Qingdao 266003, China
- School of Stomatology, Qingdao University, Qingdao 266003, China
| | - Weiping Zheng
- Institute for Translational Medicine, Department of Stomatology, The Affiliated Hospital of Qingdao University, Qingdao University, Qingdao 266003, China
- School of Stomatology, Qingdao University, Qingdao 266003, China
| | - Yanjing Ji
- Institute for Translational Medicine, Department of Stomatology, The Affiliated Hospital of Qingdao University, Qingdao University, Qingdao 266003, China
- School of Stomatology, Qingdao University, Qingdao 266003, China
| | - Peixue Ling
- Key Laboratory of Biopharmaceuticals, Shandong Academy of Pharmaceutical Sciences, Jinan 250101, China
| | - Yun Lu
- Department of Gastroenterology, The Affiliated Hospital of Qingdao University, Qingdao 266003, China
| | - Qihui Zhou
- Institute for Translational Medicine, Department of Stomatology, The Affiliated Hospital of Qingdao University, Qingdao University, Qingdao 266003, China
- Tianjin Enterprise Key Laboratory for Application Research of Hyaluronic Acid, Tianjin 300038, China
| |
Collapse
|
19
|
Mohammedsaeed W. Identification of skin microbiota in Saudi female community and their effects on keratinocytes viability (in vitro). JOURNAL OF TAIBAH UNIVERSITY FOR SCIENCE 2022. [DOI: 10.1080/16583655.2021.2015899] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/19/2022]
Affiliation(s)
- Walaa Mohammedsaeed
- Medical Laboratories Technology Department, Taibah University, AL-Madinah AL-Menorah, Saudi Arabia
| |
Collapse
|
20
|
Mohammedsaeed W, Manzoor N. An in vitro investigation of the protective role of Staphylococcus Epidermidis extracts on Staphylococcus Aureus induced toxicity in human keratinocytes. Indian J Med Microbiol 2022; 40:239-244. [DOI: 10.1016/j.ijmmb.2022.01.005] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2021] [Revised: 12/30/2021] [Accepted: 01/16/2022] [Indexed: 11/05/2022]
|
21
|
Ostapchenko D, Korotkyi O, Penchyk Y, Tsyryuk O, Sichel L. ANTIMICROBIAL POTENTIAL OF LACTIC ACID BACTERIA LACTOBACILLUS RHAMNOSUS LYSATE. BULLETIN OF TARAS SHEVCHENKO NATIONAL UNIVERSITY OF KYIV. SERIES: BIOLOGY 2022. [DOI: 10.17721/1728.2748.2022.91.19-23] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/01/2023]
Abstract
According to World Health Organization, antibiotic resistance is rising to dangerously high levels in all parts of the world. New resistance mechanisms are emerging and spreading globally, threatening our ability to treat common infectious diseases. Therefore, searching for new antimicrobial agents of natural origin is an extraordinary global problem. The work aimed to determine the antimicrobial activity of lyophilized enzymatic lysate of cells of the Lactobacillus rhamnosus V strain of lactic acid bacteria. The object of the study was the drug Del-Imun V®, which hasanti-allergican dimmuno stimulating activity. The researchers' efforts aimed to fully reveal the drug's potential, particularlyitsanti microbialaction. Antimicrobial activity was determined by the minimum inhibitory concentration (MIC). Determination of MIC was carried out by the method of twotime serial dilutions in meat-peptone broth (MPB) for bacteria and liquid wort for yeast. Gram-negative (Escherichia coli IEM-1, Proteus vulgaris PA-12, Pseudomonas sp. MI-2) and Gram-positive (Bacillus subtilis BТ-2, Staphylococcus aureus BМС-1) bacteria, as well as yeast (Candida albicans D-6, Candida tropicalis PE-2, Candida utilis BVS-65). It was shown that MIC valuesof the native preparation for the bacterial test cultures (EscherichiacoliIEM-1, Bacillussubtilis BT-2, Staphylococcusaureus BMS-1, Proteusvulgaris PA-12, Pseudomonassp. MI-2) were 8 time slower, than those of the thermally in activated preparation, forthe yeasts (Candidaalbicans D-6, Candidatropicalis PE-2, Candidautilis BVS-65) – 4-8 time slower. As a result of the conducted research, the antibacterial and antifungal activity of the drugDel-Imun V® was established. The spectrum of antimicrobial activity concerned gram-positiveand gram-negative bacteria and yeast-like fungi of the genus Candida. The minimum inhibitory concentrations were quite low: from 1.0 to 4.0 μg/ml for bacterial cultures and from 62.5 to 125 μg/ml for yeast. The culture of B. subtilis BT-2 was the least sensitive to the drug's action (MIC – 12.5 μg/ml). There fore, it can be concluded that the lysate of Lactobacillus rhamnosus V lacticacid bacteriahasanti bacteria landanti fungal properties.
Collapse
|
22
|
Stanbro J, Park JM, Bond M, Stockelman MG, Simons MP, Watters C. Topical Delivery of Lactobacillus Culture Supernatant Increases Survival and Wound Resolution in Traumatic Acinetobacter baumannii Infections. Probiotics Antimicrob Proteins 2021; 12:809-818. [PMID: 31741312 DOI: 10.1007/s12602-019-09603-z] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
Species of Lactobacillus have been proposed as potential candidates for treating wound infections due to their ability to lower pH, decrease inflammation, and release antimicrobial compounds. This study investigated the impact of lactobacilli (Lactobacillus acidophilus ATCC 4356, Lactobacillus casei ATCC 393, Lactobacillus reuteri ATCC 23272) secreted products on wound pathogens in vitro and in a murine wound infection model. Evaluation of 1-5 day lactobacilli conditioned media (CM) revealed maximal inhibition against wound pathogens using the 5-day CM. The minimum inhibitory concentration (MIC) of 5-day Lactobacillus CMs was tested by diluting CM in Mueller-Hinton (MH) broth from 0 to 25% and was found to be 12.5% for A. baumannii. Concentrating the CM to 10× with a 3 kDa centrifuge filter decreased the CM MIC to 6.25-12.5% for A. baumannii planktonic cells. Minimal impact of 5-day CMs was observed against bacterial biofilms. No toxicity was observed when these Lactobacillus CMs were injected into Galleria melonella waxworms. For the murine A. baumannii wound infection studies, improved survival was observed following topical treatment with L. acidophilus ATCC 4356 or L. reuteri ATCC 23272, while L. reuteri ATCC 23272 treatment alone improved wound resolution. Overall, this study suggests that the topical application of certain Lactobacillus species byproducts could be effective against gram-negative multi-drug resistant (MDR) wound pathogens, such as A. baumannii.
Collapse
Affiliation(s)
- Josh Stanbro
- Wound Infections Department, Naval Medical Research Center, 503 Robert Grant Avenue, Silver Spring, MD, 20910, USA
| | - Ju Me Park
- Wound Infections Department, Naval Medical Research Center, 503 Robert Grant Avenue, Silver Spring, MD, 20910, USA
| | - Matthew Bond
- Wound Infections Department, Naval Medical Research Center, 503 Robert Grant Avenue, Silver Spring, MD, 20910, USA
| | - Michael G Stockelman
- Wound Infections Department, Naval Medical Research Center, 503 Robert Grant Avenue, Silver Spring, MD, 20910, USA
| | - Mark P Simons
- Wound Infections Department, Naval Medical Research Center, 503 Robert Grant Avenue, Silver Spring, MD, 20910, USA
| | - Chase Watters
- Wound Infections Department, Naval Medical Research Center, 503 Robert Grant Avenue, Silver Spring, MD, 20910, USA.
| |
Collapse
|
23
|
Mørch MGM, Møller KV, Hesselager MO, Harders RH, Kidmose CL, Buhl T, Fuursted K, Bendixen E, Shen C, Christensen LG, Poulsen CH, Olsen A. The TGF-β ligand DBL-1 is a key player in a multifaceted probiotic protection against MRSA in C. elegans. Sci Rep 2021; 11:10717. [PMID: 34021197 PMCID: PMC8139972 DOI: 10.1038/s41598-021-89831-y] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2021] [Accepted: 04/30/2021] [Indexed: 02/04/2023] Open
Abstract
Worldwide the increase in multi-resistant bacteria due to misuse of traditional antibiotics is a growing threat for our health. Finding alternatives to traditional antibiotics is thus timely. Probiotic bacteria have numerous beneficial effects and could offer safer alternatives to traditional antibiotics. Here, we use the nematode Caenorhabditis elegans (C. elegans) to screen a library of different lactobacilli to identify potential probiotic bacteria and characterize their mechanisms of action. We show that pretreatment with the Lactobacillus spp. Lb21 increases lifespan of C. elegans and results in resistance towards pathogenic methicillin-resistant Staphylococcus aureus (MRSA). Using genetic analysis, we find that Lb21-mediated MRSA resistance is dependent on the DBL-1 ligand of the TGF-β signaling pathway in C. elegans. This response is evolutionarily conserved as we find that Lb21 also induces the TGF-β pathway in porcine epithelial cells. We further characterize the host responses in an unbiased proteome analysis and identify 474 proteins regulated in worms fed Lb21 compared to control food. These include fatty acid CoA synthetase ACS-22, aspartic protease ASP-6 and vitellogenin VIT-2 which are important for Lb21-mediated MRSA resistance. Thus, Lb21 exerts its probiotic effect on C. elegans in a multifactorial manner. In summary, our study establishes a mechanistic basis for the antimicrobial potential of lactobacilli.
Collapse
Affiliation(s)
- Maria G M Mørch
- Department of Chemistry and Bioscience, Aalborg University, Aalborg, Denmark
| | - Katrine V Møller
- Department of Chemistry and Bioscience, Aalborg University, Aalborg, Denmark
| | | | - Rikke H Harders
- Department of Chemistry and Bioscience, Aalborg University, Aalborg, Denmark
| | - Caroline L Kidmose
- Department of Chemistry and Bioscience, Aalborg University, Aalborg, Denmark
| | - Therese Buhl
- Department of Chemistry and Bioscience, Aalborg University, Aalborg, Denmark
| | | | - Emøke Bendixen
- Department of Molecular Biology and Genetics, Aarhus University, Aarhus, Denmark
| | - Chong Shen
- Gut Immunology Lab, Health & Biosciences , IFF , Brabrand , Denmark
| | | | | | - Anders Olsen
- Department of Chemistry and Bioscience, Aalborg University, Aalborg, Denmark.
| |
Collapse
|
24
|
Antibiotic Resistance Crisis: An Update on Antagonistic Interactions between Probiotics and Methicillin-Resistant Staphylococcus aureus (MRSA). Curr Microbiol 2021; 78:2194-2211. [PMID: 33881575 DOI: 10.1007/s00284-021-02442-8] [Citation(s) in RCA: 25] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2020] [Accepted: 03/01/2021] [Indexed: 02/07/2023]
Abstract
Antimicrobial resistance (AMR) havoc is a global multifaceted crisis endowing a significant challenge for the successful eradication of devastating pathogens. Methicillin-Resistant Staphylococcus aureus (MRSA) is an enduring superbug involved in causing devastating infections. Although MRSA is a frequent colonizer of human skin, wound, and anterior nares, the intestinal colonization of MRSA has greatly increased the risk of inducing MRSA-associated colitis besides creating a conducive environment for horizontal transfer of resistant genes to commensal microbes. On the other hand, staphylococcal resistance to last-resort antibiotics has urged the development of novel antimicrobial agents for the effective decolonization of MRSA. In response, probiotics and their metabolites (postbiotics) have been proposed as the adjunct therapeutic avenues. Probiotics exhibit a multitude of anti-MRSA actions (anti-bacterial, anti-biofilm, anti-virulence, anti-drug resistance, co-aggregation, and anti-quorum sensing) through the production of numerous antagonistic compounds such as organic acids, hydrogen peroxide, low molecular weight compounds, biosurfactants, bacteriocins, and bacteriocins like inhibitory substances. Besides, probiotics stabilize the epithelial barrier function and positively modulate the host immune system via regulating various signal transduction mechanisms. Preclinical and human intervention studies have suggested that probiotics outcompete with MRSA by exhibiting anti-colonization mechanisms via protective, competitive, and displacement mode. In this review, we aim to highlight the dynamics of MRSA associated virulence and drug resistance properties, and how probiotics antagonize MRSA through various mechanism of action.
Collapse
|
25
|
Salimi F, Mohammadipanah F. Nanomaterials Versus The Microbial Compounds With Wound Healing Property. FRONTIERS IN NANOTECHNOLOGY 2021. [DOI: 10.3389/fnano.2020.584489] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/27/2023] Open
Abstract
Age and diabetes related slow-healing or chronic wounds may result in morbidity and mortality through persistent biofilms infections and prolonged inflammatory phase. Nano-materials [metal/metal oxide NPs (39%), lipid vehicles (21%), polymer NPs (19%), ceramic nanoparticles (NPs) (14%), and carbon nanomaterials (NMs) (7%)] can be introduced as a possible next-generation therapy because of either their intrinsic wound healing activity or via carrying bioactive compounds including, antibiotics, antioxidants, growth factor or stem cell. The nanomaterials have been shown to implicate in all four stages of wound healing including hemostasis (polymer NPs, ceramic NPs, nanoceria-6.1%), inflammation (liposome/vesicles/solid lipid NPs/polymer NPs/ceramic NPs/silver NPs/gold NPs/nanoceria/fullerenes/carbon-based NPs-32.7%), proliferation (vesicles/liposome/solid lipid NPs/gold NPs/silver NPs/iron oxide NPs/ceramic NPs/copper NPs/self-assembling elastin-like NPs/nanoceria/micelle/dendrimers/polymer NPs-57.1%), remodeling (iron oxide NPs/nanoceria-4.1%). Natural compounds from alkaloids, flavonoids, retinoids, volatile oil, terpenes, carotenoids, or polyphenolic compounds with proven antioxidant, anti-inflammatory, immunomodulatory, or antimicrobial characteristics are also well known for their potential to accelerate the wound healing process. In the current paper, we survey the potential and properties of nanomaterials and microbial compounds in improving the process of wound and scar healing. Finally, we review the potential biocompounds for incorporation to nano-material in perspective to designate more effective or multivalent wound healing natural or nano-based drugs.
Collapse
|
26
|
Accorsi EK, Franzosa EA, Hsu T, Joice Cordy R, Maayan-Metzger A, Jaber H, Reiss-Mandel A, Kline M, DuLong C, Lipsitch M, Regev-Yochay G, Huttenhower C. Determinants of Staphylococcus aureus carriage in the developing infant nasal microbiome. Genome Biol 2020; 21:301. [PMID: 33308267 PMCID: PMC7731505 DOI: 10.1186/s13059-020-02209-7] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2020] [Accepted: 11/19/2020] [Indexed: 12/26/2022] Open
Abstract
BACKGROUND Staphylococcus aureus is a leading cause of healthcare- and community-associated infections and can be difficult to treat due to antimicrobial resistance. About 30% of individuals carry S. aureus asymptomatically in their nares, a risk factor for later infection, and interactions with other species in the nasal microbiome likely modulate its carriage. It is thus important to identify ecological or functional genetic elements within the maternal or infant nasal microbiomes that influence S. aureus acquisition and retention in early life. RESULTS We recruited 36 mother-infant pairs and profiled a subset of monthly longitudinal nasal samples from the first year after birth using shotgun metagenomic sequencing. The infant nasal microbiome is highly variable, particularly within the first 2 months. It is weakly influenced by maternal nasal microbiome composition, but primarily shaped by developmental and external factors, such as daycare. Infants display distinctive patterns of S. aureus carriage, positively associated with Acinetobacter species, Streptococcus parasanguinis, Streptococcus salivarius, and Veillonella species and inversely associated with maternal Dolosigranulum pigrum. Furthermore, we identify a gene family, likely acting as a taxonomic marker for an unclassified species, that is significantly anti-correlated with S. aureus in infants and mothers. In gene content-based strain profiling, infant S. aureus strains are more similar to maternal strains. CONCLUSIONS This improved understanding of S. aureus colonization is an important first step toward the development of novel, ecological therapies for controlling S. aureus carriage.
Collapse
Affiliation(s)
- Emma K. Accorsi
- Harvard T. H. Chan School of Public Health, 677 Huntington Avenue, Boston, MA 02115 USA
| | - Eric A. Franzosa
- Harvard T. H. Chan School of Public Health, 677 Huntington Avenue, Boston, MA 02115 USA
- Broad Institute, 415 Main St., Cambridge, MA 02142 USA
| | - Tiffany Hsu
- Harvard T. H. Chan School of Public Health, 677 Huntington Avenue, Boston, MA 02115 USA
- Broad Institute, 415 Main St., Cambridge, MA 02142 USA
| | - Regina Joice Cordy
- Harvard T. H. Chan School of Public Health, 677 Huntington Avenue, Boston, MA 02115 USA
- Wake Forest University, 1834 Wake Forest Rd., Winston-Salem, NC 27109 USA
| | - Ayala Maayan-Metzger
- Sackler Faculty of Medicine, Tel Aviv University, 69978 Ramat Aviv, Tel Aviv, Israel
- Sheba Medical Center, Derech Sheba 2, Ramat Gan, Israel
| | - Hanaa Jaber
- Sheba Medical Center, Derech Sheba 2, Ramat Gan, Israel
| | | | - Madeleine Kline
- Harvard Medical School, 25 Shattuck St., Boston, MA 02115 USA
| | - Casey DuLong
- Harvard T. H. Chan School of Public Health, 677 Huntington Avenue, Boston, MA 02115 USA
| | - Marc Lipsitch
- Harvard T. H. Chan School of Public Health, 677 Huntington Avenue, Boston, MA 02115 USA
| | - Gili Regev-Yochay
- Sackler Faculty of Medicine, Tel Aviv University, 69978 Ramat Aviv, Tel Aviv, Israel
- Sheba Medical Center, Derech Sheba 2, Ramat Gan, Israel
| | - Curtis Huttenhower
- Harvard T. H. Chan School of Public Health, 677 Huntington Avenue, Boston, MA 02115 USA
- Broad Institute, 415 Main St., Cambridge, MA 02142 USA
| |
Collapse
|
27
|
Mourenza Á, Gil JA, Mateos LM, Letek M. Alternative Anti-Infective Treatments to Traditional Antibiotherapy against Staphylococcal Veterinary Pathogens. Antibiotics (Basel) 2020; 9:antibiotics9100702. [PMID: 33076497 PMCID: PMC7602553 DOI: 10.3390/antibiotics9100702] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2020] [Revised: 10/13/2020] [Accepted: 10/14/2020] [Indexed: 12/29/2022] Open
Abstract
The genus Staphylococcus encompasses many species that may be pathogenic to both humans and farm animals. These bacteria have the potential to acquire multiple resistant traits to the antimicrobials currently used in the veterinary or medical settings. These pathogens may commonly cause zoonoses, and the infections they cause are becoming difficult to treat due to antimicrobial resistance. Therefore, the development of novel alternative treatments to traditional antibiotherapy has gained interest in recent years. Here, we reviewed the most promising therapeutic strategies developed to control staphylococcal infections in the veterinary field to overcome antibiotic resistance.
Collapse
Affiliation(s)
- Álvaro Mourenza
- Departamento de Biología Molecular, Área de Microbiología, Universidad de León, 24071 León, Spain; (Á.M.); (J.A.G.)
| | - José A. Gil
- Departamento de Biología Molecular, Área de Microbiología, Universidad de León, 24071 León, Spain; (Á.M.); (J.A.G.)
- Instituto de Biología Molecular, Genómica y Proteómica (INBIOMIC), Universidad de León, 24071 León, Spain
| | - Luis M. Mateos
- Departamento de Biología Molecular, Área de Microbiología, Universidad de León, 24071 León, Spain; (Á.M.); (J.A.G.)
- Instituto de Biología Molecular, Genómica y Proteómica (INBIOMIC), Universidad de León, 24071 León, Spain
- Correspondence: (L.M.M.); (M.L.)
| | - Michal Letek
- Departamento de Biología Molecular, Área de Microbiología, Universidad de León, 24071 León, Spain; (Á.M.); (J.A.G.)
- Instituto de Desarrollo Ganadero y Sanidad Animal (INDEGSAL), Universidad de León, 24071 León, Spain
- Correspondence: (L.M.M.); (M.L.)
| |
Collapse
|
28
|
Exploring the wound healing, anti-inflammatory, anti-pathogenic and proteomic effects of lactic acid bacteria on keratinocytes. Sci Rep 2020; 10:11572. [PMID: 32665600 PMCID: PMC7360600 DOI: 10.1038/s41598-020-68483-4] [Citation(s) in RCA: 56] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2020] [Accepted: 06/25/2020] [Indexed: 12/26/2022] Open
Abstract
The topical application of lactic acid bacteria (LAB) is recognized as a useful approach to improve skin health. This work aims to characterize by a multidisciplinary approach, the wound healing, anti-inflammatory, anti-pathogens and proteomic effects of six LAB lysates, belonging to the genus Lactobacillus. Our results demonstrated that the lysates of tested LAB stimulated the proliferation of keratinocytes, and that L. plantarum SGL 07 and L. salivarius SGL 19 accelerated the re-epithelization by inducing keratinocyte migration. The bacterial lysates also reduced the secretion of specific pro-inflammatory mediators from keratinocytes. Furthermore, viable L. salivarius SGL 19 and L. fermentum SGL 10 had anti-pathogenic effects against S. aureus and S. pyogenes, while L. brevis SGL 12 and L. paracasei SGL 04 inhibited S. aureus and S. pyogenes, respectively. The tested lactobacilli lysates also induced specific proteome modulation of the exposed keratinocytes, involving dysregulation of proteins (such as interleukin enhancer-binding factor 2 and ATP-dependent RNA helicase) and pathways (such as cytokine, NF-kB, Hedgehog, and RUNX signaling) associated with their specific wound healing and anti-inflammatory effects. This study indicates the different potential of selected lactobacilli, suggesting that they may be successfully used in the future together with conventional therapies to bring relief from skin disorders.
Collapse
|
29
|
Butler É, Lundqvist C, Axelsson J. Lactobacillus reuteri DSM 17938 as a Novel Topical Cosmetic Ingredient: A Proof of Concept Clinical Study in Adults with Atopic Dermatitis. Microorganisms 2020; 8:microorganisms8071026. [PMID: 32664536 PMCID: PMC7409218 DOI: 10.3390/microorganisms8071026] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2020] [Revised: 07/07/2020] [Accepted: 07/08/2020] [Indexed: 12/14/2022] Open
Abstract
Atopic Dermatitis (AD) is a chronically relapsing skin condition characterized by dry, itchy, and inflamed skin where sufferers can frequently be subject to infections. Probiotics are known to be potent immune-modulators, and live Lactobacillus reuteri DSM 17938 has shown to be anti-inflammatory but also to possess antimicrobial and barrier function properties. This study aimed to investigate and compare two investigational ointment products (topical probiotic and control) for cutaneous acceptability, safety, and efficacy under normal conditions of use, in adult subjects with atopic dermatitis. The products were applied twice daily for 8 weeks, and cutaneous acceptability, SCORAD index, local SCORAD, and adverse events were evaluated after 4 and 8 weeks of treatment. At the end of the observations, it was demonstrated that both the probiotic-containing and probiotic-free ointments were both cutaneously acceptable and safe. It importantly showed a statistically and clinically significant improvement of the SCORAD index and local SCORAD in adult subjects with AD after 4 and 8 weeks of continuous use. In conclusion, we show evidence that the probiotic product, containing live L. reuteri DSM 17938 as an extra ingredient, is safe and promising as a novel topical cosmetic ointment and with further testing could be a standard topical product for the management of atopic dermatitis or other disorders associated with the skin.
Collapse
Affiliation(s)
- Éile Butler
- BioGaia AB, Mobilvägen 10, 223 62 Lund, Sweden; (É.B.); (C.L.)
- Faculty of health and society, Department of Health Biomedical, Malmö University, Jan Waldenströms Gata 25, 214 28 Malmö, Sweden
| | | | - Jakob Axelsson
- BioGaia AB, Mobilvägen 10, 223 62 Lund, Sweden; (É.B.); (C.L.)
- Correspondence:
| |
Collapse
|
30
|
Moskovicz V, Gross A, Mizrahi B. Extrinsic Factors Shaping the Skin Microbiome. Microorganisms 2020; 8:E1023. [PMID: 32664353 PMCID: PMC7409027 DOI: 10.3390/microorganisms8071023] [Citation(s) in RCA: 20] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2020] [Revised: 07/07/2020] [Accepted: 07/07/2020] [Indexed: 12/13/2022] Open
Abstract
Human skin, our most environmentally exposed organ, is colonized by a vast array of microorganisms constituting its microbiome. These bacterial communities are crucial for the fulfillment of human physiological functions such as immune system modulation and epidermal development and differentiation. The structure of the human skin microbiome is established during the early life stages, starting even before birth, and continues to be modulated throughout the entire life cycle, by multiple host-related and environmental factors. This review focuses on extrinsic factors, ranging from cosmetics to the environment and antibacterial agents, as forces that impact the human skin microbiome and well-being. Assessing the impact of these factors on the skin microbiome will help elucidate the forces that shape the microbial populations we coexist with. Furthermore, we will gain additional insight into their tendency to stimulate a healthy environment or to increase the propensity for skin disorder development.
Collapse
Affiliation(s)
| | | | - Boaz Mizrahi
- Faculty of Biotechnology and Food Engineering, Technion, Haifa 3200003, Israel; (V.M.); (A.G.)
| |
Collapse
|
31
|
Onbas T, Osmanagaoglu O, Kiran F. Potential Properties of Lactobacillus plantarum F-10 as a Bio-control Strategy for Wound Infections. Probiotics Antimicrob Proteins 2020; 11:1110-1123. [PMID: 30523603 DOI: 10.1007/s12602-018-9486-8] [Citation(s) in RCA: 40] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/27/2023]
Abstract
In this study, Lactobacillus plantarum F-10, a promising probiotic strain isolated from fecal microbiota of healthy breastfed infant, was assessed as a bio-control strategy for wound infections. Pseudomonas aeruginosa PAO1/ATCC 27853, methicillin-resistant Staphylococcus aureus ATCC 43300, and their hospital-derived strains isolated from skin chronic wound samples were used as important skin pathogens. The cell-free extract (CFE) of the strain F-10 was shown to inhibit the growth of all pathogens tested, while no inhibition was observed when CFE was neutralized. The strain displayed no hemolysis and exhibited a strong auto-aggregating phenotype (51.48 ± 1.45%, 5 h) as well as co-aggregation. Antibiotic resistance profile was found to be safe according to the European Food Safety Authority. Biofilm formation was measured by crystal violet assay and visualized with scanning electron microscopy and confocal laser scanning microscopy. One hundred percent reduction in biofilm formation of all pathogens tested was obtained by sub-MIC value (12.5 mg/ml) of CFE following 24-h co-incubation. Inhibition of quorum sensing-controlled virulence factors (motility, protease and elastase activity, production of pyocyanin and rhamnolipid) of P. aeruginosa strains was also observed. DPPH radical scavenging activity of the CFE was determined as 88.57 ± 0.49%. In conclusion, our results suggest that L. plantarum F-10 may represent an alternative bio-control strategy against skin infections with its antimicrobial, anti-biofilm, anti-quorum sensing, and antioxidant activity.
Collapse
Affiliation(s)
- Tugce Onbas
- Faculty of Science, Department of Biology, Ankara University, Tandogan, 06100, Ankara, Turkey
| | - Ozlem Osmanagaoglu
- Faculty of Science, Department of Biology, Ankara University, Tandogan, 06100, Ankara, Turkey
| | - Fadime Kiran
- Faculty of Science, Department of Biology, Ankara University, Tandogan, 06100, Ankara, Turkey.
| |
Collapse
|
32
|
Huang HC, Lee IJ, Huang C, Chang TM. Lactic Acid Bacteria and Lactic Acid for Skin Health and Melanogenesis Inhibition. Curr Pharm Biotechnol 2020; 21:566-577. [DOI: 10.2174/1389201021666200109104701] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2019] [Revised: 07/26/2019] [Accepted: 12/11/2019] [Indexed: 12/21/2022]
Abstract
Lactic acid bacteria are beneficial to human health. Lactic acid bacteria have wide applications
in food, cosmetic and medicine industries due to being Generally Recognized As Safe (GRAS)
and a multitude of therapeutic and functional properties. Previous studies have reported the beneficial
effects of lactic acid bacteria, their extracts or ferments on skin health, including improvements in skin
conditions and the prevention of skin diseases. Lipoteichoic acid isolated from Lactobacillus plantarum
was reported to inhibit melanogenesis in B16F10 melanoma cells. In particular, lipoteichoic acid
also exerted anti-photoaging effects on human skin cells by regulating the expression of matrix metalloproteinase-
1. The oral administration of Lactobacillus delbrueckii and other lactic acid bacteria has
been reported to inhibit the development of atopic diseases. Additionally, the clinical and histologic
evidence indicates that the topical application of lactic acid is effective for depigmentation and improving
the surface roughness and mild wrinkling of the skin caused by environmental photo-damage. This
review discusses recent findings on the effects of lactic acid bacteria on skin health and their specific
applications in skin-whitening cosmetics.
Collapse
Affiliation(s)
- Huey-Chun Huang
- Department of Medical Laboratory Science and Biotechnology, China Medical University, Taichung, Taiwan
| | - I. Jung Lee
- Department of Kampo Medicine, Yokohama University of Pharmacy, Yokohama, Japan
| | - Chen Huang
- Office of Paradigm Industrial- Academic R & D Headquarter, Hungkuang University, Taichung, Taiwan
| | - Tsong-Min Chang
- Department of Applied Cosmetology, Hungkuang University, Taichung City, Taiwan
| |
Collapse
|
33
|
Moman R, O'Neill CA, Ledder RG, Cheesapcharoen T, McBain AJ. Mitigation of the Toxic Effects of Periodontal Pathogens by Candidate Probiotics in Oral Keratinocytes, and in an Invertebrate Model. Front Microbiol 2020; 11:999. [PMID: 32612578 PMCID: PMC7308727 DOI: 10.3389/fmicb.2020.00999] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2020] [Accepted: 04/24/2020] [Indexed: 12/23/2022] Open
Abstract
The larvae of the wax moth Galleria mellonella and human oral keratinocytes were used to investigate the protective activity of the candidate oral probiotics Lactobacillus rhamnosus GG (LHR), Lactobacillus reuteri (LR), and Streptococcus salivarius K-12 (SS) against the periodontal pathogens Fusobacterium nucleatum (FN), Porphyromonas gingivalis (PG), and Aggregatibacter actinomycetemcomitans (AA). Probiotics were delivered to the larvae (i) concomitantly with the pathogen in the same larval pro-leg; (ii) concomitantly with the pathogen in different pro-legs, and (iii) before inoculation with the pathogen in different pro-legs. Probiotics were delivered as viable cells, cell lysates or cell supernatants to the oral keratinocytes concomitantly with the pathogen. The periodontal pathogens killed at least 50% of larvae within 24 h although PG and FN were significantly more virulent than AA in the order FN > PG > AA and were also significantly lethal to mammalian cells. The candidate probiotics, however, were not lethal to the larvae or human oral keratinocytes at doses up to 107 cells/larvae. Wax worm survival rates increased up to 60% for some probiotic/pathogen combinations compared with control larvae inoculated with pathogens only. SS was the most effective probiotic against FN challenge and LHR the least, in simultaneous administration and pre-treatment, SS and LR were generally the most protective against all pathogens (up to 60% survival). For P. gingivalis, LR > LHR > SS, and for A. actinomycetemcomitans SS > LHR and LR. Administering the candidate probiotics to human oral keratinocytes significantly decreased the toxic effects of the periodontal pathogens. In summary, the periodontal pathogens were variably lethal to G. mellonella and human oral keratinocytes and the candidate probiotics had measurable protective effects, which were greatest when administrated simultaneously with the periodontal pathogens, suggesting protective effects based on bacterial interaction, and providing a basis for mechanistic studies.
Collapse
Affiliation(s)
- Raja Moman
- Department of Microbiology and Immunology, Faculty of Pharmacy, University of Tripoli, Tripoli, Libya
| | - Catherine A O'Neill
- Division of Musculoskeletal and Dermatological Sciences, School of Biological Sciences, The University of Manchester, Manchester, United Kingdom
| | - Ruth G Ledder
- Division of Pharmacy and Optometry, School of Health Sciences, Faculty of Biology, Medicine and Health, The University of Manchester, Manchester, United Kingdom
| | - Tanaporn Cheesapcharoen
- Division of Pharmacy and Optometry, School of Health Sciences, Faculty of Biology, Medicine and Health, The University of Manchester, Manchester, United Kingdom
| | - Andrew J McBain
- Division of Pharmacy and Optometry, School of Health Sciences, Faculty of Biology, Medicine and Health, The University of Manchester, Manchester, United Kingdom
| |
Collapse
|
34
|
Chien AL, Tsai J, Leung S, Mongodin EF, Nelson AM, Kang S, Garza LA. Association of Systemic Antibiotic Treatment of Acne With Skin Microbiota Characteristics. JAMA Dermatol 2020; 155:425-434. [PMID: 30758497 DOI: 10.1001/jamadermatol.2018.5221] [Citation(s) in RCA: 53] [Impact Index Per Article: 13.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
Importance Given the widespread use of systemic antibiotics for treatment of moderate to severe acne, it is important to understand the associations of such antibiotic use with changes not only in Cutibacterium acnes (formerly Propionibacterium acnes) but also in the complete bacterial community of the skin. Objective To examine the composition, diversity, and resilience of skin microbiota associated with systemic antibiotic perturbation in individuals with acne. Design, Setting, and Participants This longitudinal cohort study conducted at an academic referral center in Maryland from February 11 to September 23, 2014, included 4 female participants who had received a recent diagnosis of acne vulgaris, showed comedonal and inflammatory acne on the face, were at least 18 years old, and had no recent use of systemic or topical treatments for acne, including antibiotics and retinoids. Data analysis was performed between July 5, 2017, and November 7, 2018. Interventions Participants were prescribed oral minocycline, 100 mg, twice daily for 4 weeks. Skin areas on the forehead, cheek, and chin were sampled for 16S ribosomal RNA gene sequencing at baseline, 4 weeks after starting minocycline treatment, and then 1 week and 8 weeks after discontinuation of treatment. Main Outcomes and Measures Skin microbiota examined with respect to relative abundance of bacterial taxa, α diversity (represents within-sample microbial diversity), and β diversity (represents between-sample microbial diversity). Acne status evaluated with photography and lesion count. Results Of the 4 patients included in this study, 2 were 25 years old, 1 was 29 years old, and 1 was 35 years old; 2 were white women, 1 was an African American woman, and 1 was an Asian woman. Across all 4 patients, antibiotic treatment was associated with a 1.4-fold reduction in the level of C acnes (difference, -10.3%; 95% CI, -19.9% to -0.7%; P = .04) with recovery following cessation of treatment. Distinct patterns of change were identified in multiple bacterial genera, including a transient 5.6-fold increase in the relative abundance of Pseudomonas species (difference, 2.2%; 95% CI, 0.9%-3.4%; P < .001) immediately following antibiotic treatment, as well as a persistent 1.7-fold increase in the relative abundance of Streptococcus species (difference, 5.4%; 95% CI, 0.3%-10.6%; P = .04) and a 4.7-fold decrease in the relative abundance of Lactobacillus species (difference, -0.8%; 95% CI, -1.4% to -0.2%; P = .02) 8 weeks following antibiotic treatment withdrawal. In general, antibiotic administration was associated with an initial decrease from baseline of bacterial diversity followed by recovery. Principal coordinates analysis results showed moderate clustering of samples by patient (analysis of similarity, R = 0.424; P = .001) and significant clustering of samples by time in one participant (analysis of similarity, R = 0.733; P = .001). Conclusions and Relevance In this study, systemic antibiotic treatment of acne was associated with changes in the composition and diversity of skin microbiota, with variable rates of recovery across individual patients and parallel changes in specific bacterial populations. Understanding the association between systemic antibiotic use and skin microbiota may help clinicians decrease the likelihood of skin comorbidities related to microbial dysbiosis.
Collapse
Affiliation(s)
- Anna L Chien
- Department of Dermatology, Johns Hopkins School of Medicine, Baltimore, Maryland
| | - Jerry Tsai
- Department of Dermatology, Johns Hopkins School of Medicine, Baltimore, Maryland
| | - Sherry Leung
- Department of Dermatology, Johns Hopkins School of Medicine, Baltimore, Maryland
| | - Emmanuel F Mongodin
- Institute for Genome Sciences, University of Maryland School of Medicine, Baltimore
| | - Amanda M Nelson
- Department of Dermatology, Johns Hopkins School of Medicine, Baltimore, Maryland.,Now with the Department of Dermatology, Penn State College of Medicine, Hershey, Pennsylvania
| | - Sewon Kang
- Department of Dermatology, Johns Hopkins School of Medicine, Baltimore, Maryland
| | - Luis A Garza
- Department of Dermatology, Johns Hopkins School of Medicine, Baltimore, Maryland
| |
Collapse
|
35
|
Cell-Free Culture Supernatants of Lactobacilli Modify the Expression of Virulence Factors Genes in Staphylococcus aureus. Jundishapur J Microbiol 2020. [DOI: 10.5812/jjm.96806] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/18/2023] Open
|
36
|
Efficacy of Using Probiotics with Antagonistic Activity against Pathogens of Wound Infections: An Integrative Review of Literature. BIOMED RESEARCH INTERNATIONAL 2019; 2019:7585486. [PMID: 31915703 PMCID: PMC6930797 DOI: 10.1155/2019/7585486] [Citation(s) in RCA: 68] [Impact Index Per Article: 13.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/22/2019] [Accepted: 10/03/2019] [Indexed: 02/06/2023]
Abstract
The skin and its microbiota serve as physical barriers to prevent invasion of pathogens. Skin damage can be a consequence of illness, surgery, and burns. The most effective wound management strategy is to prevent infections, promote healing, and prevent excess scarring. It is well established that probiotics can aid in skin healing by stimulating the production of immune cells, and they also exhibit antagonistic effects against pathogens via competitive exclusion of pathogens. Our aim was to conduct a review of recent literature on the efficacy of using probiotics against pathogens that cause wound infections. In this integrative review, we searched through the literature published in the international following databases: PubMed, ScienceDirect, Web of Science, and Scopus using the search terms “probiotic” AND “wound infection.” During a comprehensive review and critique of the selected research, fourteen in vitro studies, 8 animal studies, and 19 clinical studies were found. Two of these in vitro studies also included animal studies, yielding a total of 39 articles for inclusion in the review. The most commonly used probiotics for all studies were well-known strains of the species Lactobacillus plantarum, Lactobacillus casei, Lactobacillus acidophilus, and Lactobacillus rhamnosus. All in vitro studies showed successful inhibition of chosen skin or wound pathogens by the selected probiotics. Within the animal studies on mice, rats, and rabbits, probiotics showed strong opportunities for counteracting wound infections. Most clinical studies showed slight or statistically significant lower incidence of surgical site infections, foot ulcer infection, or burn infections for patients using probiotics. Several of these studies also indicated a statistically significant wound healing effect for the probiotic groups. This review indicates that exogenous and oral application of probiotics has shown reduction in wound infections, especially when used as an adjuvant to antibiotic therapy, and therefore the potential use of probiotics in this field remains worthy of further studies, perhaps focused more on typical skin inhabitants as next-generation probiotics with high potential.
Collapse
|
37
|
Khan MA, Hussain Z, Ali S, Qamar Z, Imran M, Hafeez FY. Fabrication of Electrospun Probiotic Functionalized Nanocomposite Scaffolds for Infection Control and Dermal Burn Healing in a Mice Model. ACS Biomater Sci Eng 2019; 5:6109-6116. [PMID: 33405664 DOI: 10.1021/acsbiomaterials.9b01002] [Citation(s) in RCA: 20] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
The importance of microbiota paves the way to use microbial cells as medicines to treat pathobiomic diseases. This study reported the fabrication of probiotic (Enterococcus mundtii QAUEM2808)-functionalized nanocomposite scaffolds of poly(vinyl alcohol)/poly(vinylpyrrolidone)/glycerol via electrospinning. Scanning electron microscopy, Fourier transform infrared spectroscopy, and thermogravimetric analysis resolved the living composite structure and supported the encapsulation of E. mundtii throughout the nanostructured (318 ± 12 nm) fibers of bioscaffold membranes. The shelf life evaluation of 4-week-old samples supported that bioscaffolds showed an enhancement in probiotic survival count by 2.78 ± 0.10 log10 colony-forming units (cfu) versus counterpart biodispersion. The swelling and antagonistic evaluation showed that a bioscaffold is degradable in a simulated wound fluid which is essential for activation of probiotic strains to antagonize infection-causing Gram-positive and Gram-negative pathogens. A second-degree contact burn was made on the dorsum of male BALB/c mice (n = 30). The wounds were left open for 2 days to mimic burn contamination, and the mice were randomized into negative (untreated), positive (silver sulfadiazine cream), vehicle (biodispersion and nanoscaffold), and experimental bioscaffold groups (n = 6/group). These treatments were applied on 2, 6, 10, and 14 days postburn. A comparative wound closure, histopathology, and wound microbial evaluation demonstrated that the bioscaffolds accelerate epithelialization, collagen deposition, and hair follicle formation, inhibit harmful bacteria, and provide interference benefits. In particular, the probiotic active bioscaffold membrane could serve as a novel candidate to control infections and speed up the healing of burn wounds.
Collapse
Affiliation(s)
- Muhammad Ali Khan
- Applied Microbiology and Biotechnology Laboratory, Department of Biosciences, COMSATS University Islamabad (CUI), Park Road, Tarlai Kalan, Islamabad, Islamabad Capital Territory 45550, Pakistan
| | - Zahid Hussain
- Applied Microbiology and Biotechnology Laboratory, Department of Biosciences, COMSATS University Islamabad (CUI), Park Road, Tarlai Kalan, Islamabad, Islamabad Capital Territory 45550, Pakistan
| | - Sakhawat Ali
- National Veterinary Laboratories (NVL), Park Road, Islamabad, Islamabad Capital Territory 45550, Pakistan
| | - Zahid Qamar
- Nano-Scale Physics Laboratory, Department of Physics, Air University, Sector E-9, Islamabad, Islamabad Capital Territory 44200, Pakistan
| | - Muhammad Imran
- Department of Microbiology, Faculty of Biological Sciences, Quaid-i-Azam University (QAU), Islamabad, Islamabad Capital Territory 45320, Pakistan
| | - Fauzia Yusuf Hafeez
- Applied Microbiology and Biotechnology Laboratory, Department of Biosciences, COMSATS University Islamabad (CUI), Park Road, Tarlai Kalan, Islamabad, Islamabad Capital Territory 45550, Pakistan
| |
Collapse
|
38
|
Abstract
Scientific and commercial interest of probiotics, prebiotics and their effect on human health and disease has increased in the last decade. The aim of this review article is to evaluate the role of pro- and prebiotics on the normal function of healthy skin as well as their role in the prevention and therapy of skin disease. Lactobacilli and Bifidobacterium are the most commonly used probiotics and thought to mediate skin inflammation, treat atopic dermatitis (AD) and prevent allergic contact dermatitis (ACD). Probiotics are shown to decolonise skin pathogens (e.g., P. aeruginosa, S. aureus, A. Vulgaris, etc.) while kefir is also shown to support the immunity of the skin and treat skin pathogens through the production of antimicrobial substances and prebiotics. Finally, prebiotics (e.g., Fructo-oligosaccharides, galacto-oligosaccharides and konjac glucomannan hydrolysates) can contribute to the treatment of diseases including ACD, acne and photo aging primarily by enhancing the growth of probiotics.
Collapse
|
39
|
Melgaço ACC, Blohem Pessoa WF, Freire HP, Evangelista de Almeida M, Santos Barbosa M, Passos Rezende R, Timenetsky J, Miranda Marques L, Romano CC. Potential of Maintaining a Healthy Vaginal Environment by Two Lactobacillus Strains Isolated from Cocoa Fermentation. BIOMED RESEARCH INTERNATIONAL 2018; 2018:7571954. [PMID: 30364031 PMCID: PMC6186379 DOI: 10.1155/2018/7571954] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/16/2018] [Revised: 08/28/2018] [Accepted: 09/12/2018] [Indexed: 11/25/2022]
Abstract
Bacteria in the genera Mycoplasma and Ureaplasma do not have cell walls and therefore interact with host cells through lipid-associated membrane proteins (LAMP). These lipoproteins are important for both surface adhesion and modulation of host immune responses. Mycoplasma and Ureaplasma have been implicated in cases of bacterial vaginosis (BV), which can cause infertility, abortion, and premature delivery. In contrast, bacteria of the genus Lactobacillus, which are present in the vaginal microbiota of healthy women, are thought to inhibit local colonization by pathogenic microorganisms. The aim of the present study was to evaluate the in vitro interactions between lipoproteins of Mycoplasma and Ureaplasma species and vaginal lineage (HMVII) cells and to study the effect of Lactobacillus isolates from cocoa fermentation on these interactions. The tested Lactobacillus strains showed some important probiotic characteristics, with autoaggregation percentages of 28.55% and 31.82% for L. fermentum FA4 and L. plantarum PA3 strains, respectively, and percent adhesion values of 31.66 and 41.65%, respectively. The two strains were hydrophobic, with moderate to high hydrophobicity values, 65.33% and 71.12% for L. fermentum FA4 and L. plantarum PA3 in toluene. Both strains secreted acids into the culture medium with pH=4.32 and pH=4.33, respectively, and showed antibiotics susceptibility profiles similar to those of other lactobacilli. The strains were also able to inhibit the death of vaginal epithelial cells after incubation with U. parvum LAMP from 41.03% to 2.43% (L. fermentum FA4) and 0.43% (L. plantarum PA3) and also managed to significantly decrease the rate of cell death caused by the interaction with LAMP of M. hominis from 34.29% to 14.06% (L. fermentum FA4) and 14.61% (L. plantarum PA3), thus demonstrating their potential for maintaining a healthy vaginal environment.
Collapse
Affiliation(s)
- Ana Clara Correia Melgaço
- Departamento de Ciências Biológicas, Laboratório de Imunologia, Centro de Biotecnologia e Genética, Universidade Estadual de Santa Cruz (UESC), Campus Soane Nazaré de Andrade, Salobrinho, Rodovia Jorge Amado, Km 16, 45662-900 Ilhéus, BA, Brazil
| | - Wallace Felipe Blohem Pessoa
- Departamento de Ciências Biológicas, Laboratório de Imunologia, Centro de Biotecnologia e Genética, Universidade Estadual de Santa Cruz (UESC), Campus Soane Nazaré de Andrade, Salobrinho, Rodovia Jorge Amado, Km 16, 45662-900 Ilhéus, BA, Brazil
| | - Herbert Pina Freire
- Departamento de Ciências Biológicas, Laboratório de Imunologia, Centro de Biotecnologia e Genética, Universidade Estadual de Santa Cruz (UESC), Campus Soane Nazaré de Andrade, Salobrinho, Rodovia Jorge Amado, Km 16, 45662-900 Ilhéus, BA, Brazil
| | - Milena Evangelista de Almeida
- Departamento de Ciências Biológicas, Laboratório de Imunologia, Centro de Biotecnologia e Genética, Universidade Estadual de Santa Cruz (UESC), Campus Soane Nazaré de Andrade, Salobrinho, Rodovia Jorge Amado, Km 16, 45662-900 Ilhéus, BA, Brazil
| | - Maysa Santos Barbosa
- Instituto de Ciências Biomédicas, Departamento de Microbiologia, Laboratório de Micoplasmas, Universidade de São Paulo (USP), São Paulo, Brazil
| | - Rachel Passos Rezende
- Departamento de Ciências Biológicas, Laboratório de Biotecnologia Microbiana, Centro de Biotecnologia e Genética, Universidade Estadual de Santa Cruz (UESC), Campus Soane Nazaré de Andrade, Salobrinho, Rodovia Jorge Amado, Km 16, 45662-900 Ilhéus, BA, Brazil
| | - Jorge Timenetsky
- Instituto de Ciências Biomédicas, Departamento de Microbiologia, Laboratório de Micoplasmas, Universidade de São Paulo (USP), São Paulo, Brazil
| | - Lucas Miranda Marques
- Instituto de Ciências Biomédicas, Departamento de Microbiologia, Laboratório de Micoplasmas, Universidade de São Paulo (USP), São Paulo, Brazil
- Instituto Multidisciplinar em Saúde/Campus Anísio Teixeira, Universidade Federal da Bahia, IMS/CAT-UFBA, Vitória da Conquista, Brazil
| | - Carla Cristina Romano
- Departamento de Ciências Biológicas, Laboratório de Imunologia, Centro de Biotecnologia e Genética, Universidade Estadual de Santa Cruz (UESC), Campus Soane Nazaré de Andrade, Salobrinho, Rodovia Jorge Amado, Km 16, 45662-900 Ilhéus, BA, Brazil
| |
Collapse
|
40
|
Zommiti M, Cambronel M, Maillot O, Barreau M, Sebei K, Feuilloley M, Ferchichi M, Connil N. Evaluation of Probiotic Properties and Safety of Enterococcus faecium Isolated From Artisanal Tunisian Meat "Dried Ossban". Front Microbiol 2018; 9:1685. [PMID: 30127770 PMCID: PMC6088202 DOI: 10.3389/fmicb.2018.01685] [Citation(s) in RCA: 66] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2018] [Accepted: 07/06/2018] [Indexed: 12/19/2022] Open
Abstract
Enterococcus faecium strains were isolated from an original biotope, artisanal dried Tunisian meat “Dried Ossban,” and evaluated for safety and capacity as probiotics. Gram-positive, catalase negative, and bacteriocin-producing bacteria were screened using selective microbiological media. All isolates were identified by phenotypic and molecular tools. Five E. faecium strains (MZF1, MZF2, MZF3, MZF4, and MZF5) were selected and further assessed for their probiotic properties. They were found to be resistant to the physiological concentrations of bile salts, and the harsh conditions of the gastrointestinal tract, and showed autoaggregation and adhesion ability. All these isolates possess at least one enterocin and could efficiently inhibit the growth of Listeria innocua HPB13. The analysis of their safety profile revealed for almost all the strains the absence of cytotoxicity and virulence determinants, and susceptibility to clinically important antibiotics such as vancomycin. These data suggest that these bacteria, isolated from “Dried Ossban,” do not present a risk to human health, and may be considered as interesting candidates for future use as probiotics and bioprotective cultures for application in the food and/or feed industries.
Collapse
Affiliation(s)
- Mohamed Zommiti
- Unité de Protéomique Fonctionnelle et Potentiel Nutraceutique de la Biodiversité de Tunisie, Institut Supérieur des Sciences Biologiques Appliquées de Tunis, Université de Tunis El Manar, Tunis, Tunisia
| | - Mélyssa Cambronel
- Laboratoire de Microbiologie Signaux et Microenvironnement, Université de Rouen Normandie, Évreux, France
| | - Olivier Maillot
- Laboratoire de Microbiologie Signaux et Microenvironnement, Université de Rouen Normandie, Évreux, France
| | - Magalie Barreau
- Laboratoire de Microbiologie Signaux et Microenvironnement, Université de Rouen Normandie, Évreux, France
| | - Khaled Sebei
- Unité de Protéomique Fonctionnelle et Potentiel Nutraceutique de la Biodiversité de Tunisie, Institut Supérieur des Sciences Biologiques Appliquées de Tunis, Université de Tunis El Manar, Tunis, Tunisia
| | - Marc Feuilloley
- Laboratoire de Microbiologie Signaux et Microenvironnement, Université de Rouen Normandie, Évreux, France
| | - Mounir Ferchichi
- Unité de Protéomique Fonctionnelle et Potentiel Nutraceutique de la Biodiversité de Tunisie, Institut Supérieur des Sciences Biologiques Appliquées de Tunis, Université de Tunis El Manar, Tunis, Tunisia.,Clinical Laboratory Department, College of Applied Medical Sciences, King Faisal University, Al-Ahsa, Saudi Arabia
| | - Nathalie Connil
- Laboratoire de Microbiologie Signaux et Microenvironnement, Université de Rouen Normandie, Évreux, France
| |
Collapse
|
41
|
Chan AP, Choi Y, Brinkac LM, Krishnakumar R, DePew J, Kim M, Hinkle MK, Lesho EP, Fouts DE. Multidrug resistant pathogens respond differently to the presence of co-pathogen, commensal, probiotic and host cells. Sci Rep 2018; 8:8656. [PMID: 29872152 PMCID: PMC5988826 DOI: 10.1038/s41598-018-26738-1] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2018] [Accepted: 05/18/2018] [Indexed: 11/19/2022] Open
Abstract
In light of the ongoing antimicrobial resistance crisis, there is a need to understand the role of co-pathogens, commensals, and the local microbiome in modulating virulence and antibiotic resistance. To identify possible interactions that influence the expression of virulence or survival mechanisms in both the multidrug-resistant organisms (MDROs) and human host cells, unique cohorts of clinical isolates were selected for whole genome sequencing with enhanced assembly and full annotation, pairwise co-culturing, and transcriptome profiling. The MDROs were co-cultured in pairwise combinations either with: (1) another MDRO, (2) skin commensals (Staphylococcus epidermidis and Corynebacterium jeikeium), (3) the common probiotic Lactobacillus reuteri, and (4) human fibroblasts. RNA-Seq analysis showed distinct regulation of virulence and antimicrobial resistance gene responses across different combinations of MDROs, commensals, and human cells. Co-culture assays demonstrated that microbial interactions can modulate gene responses of both the target and pathogen/commensal species, and that the responses are specific to the identity of the pathogen/commensal species. In summary, bacteria have mechanisms to distinguish between friends, foe and host cells. These results provide foundational data and insight into the possibility of manipulating the local microbiome when treating complicated polymicrobial wound, intra-abdominal, or respiratory infections.
Collapse
Affiliation(s)
- Agnes P Chan
- J. Craig Venter Institute (JCVI), 9605 Medical Center Drive, Suite 150, Rockville, MD, 20850, United States.
| | - Yongwook Choi
- J. Craig Venter Institute (JCVI), 9605 Medical Center Drive, Suite 150, Rockville, MD, 20850, United States
| | - Lauren M Brinkac
- J. Craig Venter Institute (JCVI), 9605 Medical Center Drive, Suite 150, Rockville, MD, 20850, United States
| | - Radha Krishnakumar
- J. Craig Venter Institute (JCVI), 9605 Medical Center Drive, Suite 150, Rockville, MD, 20850, United States
| | - Jessica DePew
- J. Craig Venter Institute (JCVI), 9605 Medical Center Drive, Suite 150, Rockville, MD, 20850, United States
| | - Maria Kim
- J. Craig Venter Institute (JCVI), 9605 Medical Center Drive, Suite 150, Rockville, MD, 20850, United States
| | - Mary K Hinkle
- Multidrug-resistant organism Repository and Surveillance Network, Bacterial Diseases Branch, Walter Reed Army Institute of Research, Silver Spring, MD, 20910, United States
| | - Emil P Lesho
- Multidrug-resistant organism Repository and Surveillance Network, Bacterial Diseases Branch, Walter Reed Army Institute of Research, Silver Spring, MD, 20910, United States.,Infectious Diseases Unit, Rochester Regional Health, Rochester, NY, 14621, United States
| | - Derrick E Fouts
- J. Craig Venter Institute (JCVI), 9605 Medical Center Drive, Suite 150, Rockville, MD, 20850, United States.
| |
Collapse
|
42
|
Nwanodi O. Skin Protective Nutraceuticals: The Current Evidence in Brief. Healthcare (Basel) 2018; 6:healthcare6020040. [PMID: 29734688 PMCID: PMC6023352 DOI: 10.3390/healthcare6020040] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2018] [Revised: 04/16/2018] [Accepted: 05/01/2018] [Indexed: 01/02/2023] Open
Abstract
Nutraceuticals are important for healthy skin maintenance. Probiotics, phenolics, and vitamins are just a few of the nutraceuticals meant to potentially prevent and assist medical management of dermatologic conditions. Among these, probiotics, vitamin E, and green tea catechins may offer the broadest array of skin protective mechanisms with probiotics having the greatest clinical range. Probiotics’ amelioration of atopic dermatitis and opportunistic infections of skin burns has been targeted in recent research efforts. This includes the improvement of Scoring Atopic Dermatitis index scores, p = 0.02, with intact Lactobacillus rhamnosus Goldin and Gorbach (LGG) in comparison to heat inactivated LGG or placebo. Lactobacillus reuteri used prior to or concurrently with Staphylococcus aureus infection can increase epidermal keratinocyte survival, p < 0.01. Phenolics may not have been extensively studied for atopic dermatitis or skin burns. However, phenolics do have a role in photoprotection. The phenolic rutin increases ultraviolet B radiation filter reactive oxygen species scavenging at 75%, p < 0.002, and peak wavelength absorption, p < 0.001. While oral and topical probiotics have untapped potential for atopic dermatitis amelioration and skin infection prevention, phenolics will be increasingly used for photoprotection. With optimized bioavailability, dosage, and formulation, nutraceuticals will become crucial for healthy skin maintenance.
Collapse
Affiliation(s)
- Oroma Nwanodi
- Obstetrics and Gynecology Locum Tenens, Salinas, CA 93902, USA.
| |
Collapse
|
43
|
Tapia-Paniagua ST, Ceballos-Francisco D, Balebona MC, Esteban MÁ, Moriñigo MÁ. Mucus glycosylation, immunity and bacterial microbiota associated to the skin of experimentally ulcered gilthead seabream (Sparus aurata). FISH & SHELLFISH IMMUNOLOGY 2018; 75:381-390. [PMID: 29421587 DOI: 10.1016/j.fsi.2018.02.006] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/29/2017] [Revised: 01/24/2018] [Accepted: 02/02/2018] [Indexed: 05/25/2023]
Abstract
Interest in fish skin immunity and its associated microbiota has greatly increased among immunologists. The objective of this study is to know if skin ulcers may be associated with changes in the mucus composition and microbial diversity. The abundance of terminal carbohydrates, several enzymes (protease, antiprotease, peroxidase, lysozyme) and total immunoglobulin M levels were evaluated in skin mucus of experimentally ulcered gilthead seabream (Sparus aurata L.). Furthermore, the composition of the microbiota of ulcered and non-ulcered skin has been determined using Illumina Miseq technology. Significant decreases of terminal abundance of α-D-mannose, α-D-glucose and N-acetyl-galactosamine in skin mucus of ulcered fish, compared to control fish were detected. The levels of IgM and all the tested enzymes in mucus were decreased in ulcered fish (compared to control fish) although the observed decreases were only statistically significant for proteases and antiproteases. Concomitantly, the analysis of the composition of the skin microbiota showed clear differences between ulcered and non-ulcered areas. The genus taxonomic analysis showed that Staphylococcus and Lactobacillus were more abundant in non-ulcered skin whereas in ulcered area were Streptococcus and Granulicatella. Important decreases of the number of sequences related to Alteromonas, Thalassabius and Winogradskyella were detected in ulcered skin whilst slight increases of sequences related to Flavobacterium, Chryseobacterium and Tenacibaculum genera were observed. Overall these results demonstrated that the presence of skin ulcers provide microenvironments that perturb both the mucus composition and microbial biodiversity of this important external surface which seem to be more vulnerable to diseases.
Collapse
Affiliation(s)
- Silvana Teresa Tapia-Paniagua
- Group of Prophylaxis and Biocontrol of Fish Diseases, Departamento de Microbiología, Campus de Teatinos s/n, Universidad de Málaga, 29071 Málaga, Spain
| | - Diana Ceballos-Francisco
- Department of Cell Biology and Histology, Faculty of Biology, Campus Regional de Excelencia Internacional "Campus Mare Nostrum", University of Murcia, 30100, Murcia, Spain
| | - M Carmen Balebona
- Group of Prophylaxis and Biocontrol of Fish Diseases, Departamento de Microbiología, Campus de Teatinos s/n, Universidad de Málaga, 29071 Málaga, Spain
| | - María Ángeles Esteban
- Department of Cell Biology and Histology, Faculty of Biology, Campus Regional de Excelencia Internacional "Campus Mare Nostrum", University of Murcia, 30100, Murcia, Spain
| | - Miguel Ángel Moriñigo
- Group of Prophylaxis and Biocontrol of Fish Diseases, Departamento de Microbiología, Campus de Teatinos s/n, Universidad de Málaga, 29071 Málaga, Spain.
| |
Collapse
|
44
|
Lagrafeuille R, Miquel S, Balestrino D, Vareille-Delarbre M, Chain F, Langella P, Forestier C. Opposing effect of Lactobacillus on in vitro Klebsiella pneumoniae in biofilm and in an in vivo intestinal colonisation model. Benef Microbes 2018; 9:87-100. [DOI: 10.3920/bm2017.0002] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/06/2023]
Abstract
Beneficial bacteria represent potential sources of therapy, particularly in the battle against antibiotic-resistant pathogens. The Gram-negative bacillus Klebsiella pneumoniae is not only a paradigm of multi-resistant opportunistic pathogen, but it is also able to colonise the human intestine and displays a high capacity to form biofilm. In this study, the anti-biofilm activity of 140 neutralised Lactobacillus supernatants was assessed against K. pneumoniae. Among the 13 strains whose supernatant significantly impaired biofilm formation, Lactobacillus plantarum CIRM653 was selected because it was also able to impair K. pneumoniae preformed biofilm, independently of a bactericidal effect. Mixed K. pneumoniae/L. plantarum CIRM653 biofilms had reduced tridimensional structures associated with a significant decrease in K. pneumoniae biomass. Further investigation showed that L. plantarum CIRM653 supernatant induced transcriptional modifications of K. pneumoniae biofilm-related genes, including down-regulation of the quorum sensing-related lsr operons and over-expression of type 3 pili structure genes. Increased production of type 3 pili was validated by Western-blot, hemagglutination and adhesion assays. L. plantarum CIRM653 activity against K. pneumoniae was also assessed in a murine intestinal colonisation model: a constant faecal pathogen burden was observed, as against a gradual decrease in the control group. These results reveal that an in vitro a priori attracting anti-biofilm activity of Lactobacillus might be counterbalanced by an in vivo behaviour in a complex microbiota environment with potential deleterious dispersal of highly adherent K. pneumoniae cells, raising the question of the accuracy of in vitro assays in screening of beneficial microbes.
Collapse
Affiliation(s)
- R. Lagrafeuille
- Université Clermont Auvergne, CNRS UMR 6023 Laboratoire Microorganismes: Génome et Environnement (LMGE), 63000 Clermont-Ferrand, France
| | - S. Miquel
- Université Clermont Auvergne, CNRS UMR 6023 Laboratoire Microorganismes: Génome et Environnement (LMGE), 63000 Clermont-Ferrand, France
| | - D. Balestrino
- Université Clermont Auvergne, CNRS UMR 6023 Laboratoire Microorganismes: Génome et Environnement (LMGE), 63000 Clermont-Ferrand, France
| | | | - F. Chain
- Commensal and Probiotics-Host Interactions Laboratory/AgroParisTech, UMR 1319 Micalis, INRA, 78352 Jouy-en-Josas, France
| | - P. Langella
- Commensal and Probiotics-Host Interactions Laboratory/AgroParisTech, UMR 1319 Micalis, INRA, 78352 Jouy-en-Josas, France
| | - C. Forestier
- Université Clermont Auvergne, CNRS UMR 6023 Laboratoire Microorganismes: Génome et Environnement (LMGE), 63000 Clermont-Ferrand, France
| |
Collapse
|
45
|
Mukherjee S, Ramesh A. Dual-label flow cytometry-based host cell adhesion assay to ascertain the prospect of probiotic Lactobacillus plantarum in niche-specific antibacterial therapy. MICROBIOLOGY-SGM 2017; 163:1822-1834. [PMID: 29091578 DOI: 10.1099/mic.0.000561] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Abstract
Host cell adhesion assays that provide quantitative insight on the potential of lactic acid bacteria (LAB) to inhibit adhesion of intestinal pathogens can be leveraged for the development of niche-specific anti-adhesion therapy. Herein, we report a dual-colour flow cytometry (FCM) analysis to assess the ability of probiotic Lactobacillus plantarum strains to impede adhesion of Enterococcus faecalis, Listeria monocytogenes and Staphylococcus aureus onto HT-29 cells. FCM in conjunction with a hierarchical cluster analysis could discern the anti-adhesion potential of L. plantarum strains, wherein the efficacy of L. plantarum DF9 was on a par with the probiotic L. rhamnosus GG. Combination of FCM with principal component analysis illustrated the relative influence of LAB strains on adhesion parameters kd and em of the pathogen and identified probiotic LAB suitable for anti-adhesion intervention. The analytical merit of the FCM analysis was captured in host cell adhesion assays that measured relative elimination of adhered LAB vis-à-vis pathogens, on exposure to either LAB bacteriocins or therapeutic antibiotics. It is envisaged that the dual-colour FCM-based adhesion assay described herein would enable a fundamental understanding of the host cell adhesion process and stimulate interest in probiotic LAB as safe anti-adhesion therapeutic agents against gastrointestinal pathogens.
Collapse
Affiliation(s)
- Sandipan Mukherjee
- Department of Biosciences and Bioengineering, Indian Institute of Technology Guwahati, Guwahati 781039, Assam, India
| | - Aiyagari Ramesh
- Department of Biosciences and Bioengineering, Indian Institute of Technology Guwahati, Guwahati 781039, Assam, India
| |
Collapse
|
46
|
Lukic J, Chen V, Strahinic I, Begovic J, Lev-Tov H, Davis SC, Tomic-Canic M, Pastar I. Probiotics or pro-healers: the role of beneficial bacteria in tissue repair. Wound Repair Regen 2017; 25:912-922. [PMID: 29315980 PMCID: PMC5854537 DOI: 10.1111/wrr.12607] [Citation(s) in RCA: 81] [Impact Index Per Article: 11.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2017] [Accepted: 12/15/2017] [Indexed: 12/20/2022]
Abstract
Probiotics are beneficial microorganisms, known to exert numerous positive effects on human health, primarily in the battle against pathogens. Probiotics have been associated with improved healing of intestinal ulcers, and healing of infected cutaneous wounds. This article reviews the latest findings on probiotics related to their pro-healing properties on gut epithelium and skin. Proven mechanisms by which probiotic bacteria exert their beneficial effects include direct killing of pathogens, competitive displacement of pathogenic bacteria, reinforcement of epithelial barrier, induction of fibroblasts, and epithelial cells' migration and function. Beneficial immunomodulatory effects of probiotics relate to modulation and activation of intraepithelial lymphocytes, natural killer cells, and macrophages through induced production of cytokines. Systemic effects of beneficial bacteria and link between gut microbiota, immune system, and cutaneous health through gut-brain-skin axes are discussed as well. In light of growing antibiotic resistance of pathogens, antibiotic use is becoming less effective in treating cutaneous and systemic infections. This review points to a new perspective and therapeutic potential of beneficial probiotic species as a safe alternative approach for treatment of patients affected by wound healing disorders and cutaneous infections.
Collapse
Affiliation(s)
- Jovanka Lukic
- University of Belgrade, Institute of Molecular Genetics and Genetic Engineering, Laboratory for Molecular Microbiology, Belgrade, Serbia
| | - Vivien Chen
- University of Miami Miller School Of Medicine, Wound Healing and Regenerative Medicine Research Program, Department of Dermatology and Cutaneous Surgery, Miami, FL, USA
| | - Ivana Strahinic
- University of Belgrade, Institute of Molecular Genetics and Genetic Engineering, Laboratory for Molecular Microbiology, Belgrade, Serbia
| | - Jelena Begovic
- University of Belgrade, Institute of Molecular Genetics and Genetic Engineering, Laboratory for Molecular Microbiology, Belgrade, Serbia
| | - Hadar Lev-Tov
- University of Miami Miller School Of Medicine, Wound Healing and Regenerative Medicine Research Program, Department of Dermatology and Cutaneous Surgery, Miami, FL, USA
| | - Stephen C Davis
- University of Miami Miller School Of Medicine, Wound Healing and Regenerative Medicine Research Program, Department of Dermatology and Cutaneous Surgery, Miami, FL, USA
| | - Marjana Tomic-Canic
- University of Miami Miller School Of Medicine, Wound Healing and Regenerative Medicine Research Program, Department of Dermatology and Cutaneous Surgery, Miami, FL, USA
| | - Irena Pastar
- University of Miami Miller School Of Medicine, Wound Healing and Regenerative Medicine Research Program, Department of Dermatology and Cutaneous Surgery, Miami, FL, USA
| |
Collapse
|
47
|
Veljović K, Popović N, Miljković M, Tolinački M, Terzić-Vidojević A, Kojić M. Novel Aggregation Promoting Factor AggE Contributes to the Probiotic Properties of Enterococcus faecium BGGO9-28. Front Microbiol 2017; 8:1843. [PMID: 29018422 PMCID: PMC5622976 DOI: 10.3389/fmicb.2017.01843] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2017] [Accepted: 09/08/2017] [Indexed: 01/07/2023] Open
Abstract
The understanding of mechanisms of interactions between various bacterial cell surface proteins and host receptors has become imperative for the study of the health promoting features of probiotic enterococci. This study, for the first time, describes a novel enterococcal aggregation protein, AggE, from Enterococcus faecium BGGO9-28, selected from a laboratory collection of enterococcal isolates with auto-aggregation phenotypes. Among them, En. faecium BGGO9-28 showed the strongest auto-aggregation, adhesion to components of ECM and biofilm formation. Novel aggregation promoting factor AggE, a protein of 178.1 kDa, belongs to the collagen-binding superfamily of proteins and shares similar architecture with previously discovered aggregation factors from lactic acid bacteria (LAB). Its expression in heterologous enterococcal and lactococcal hosts demonstrates that the aggE gene is sufficient for cell aggregation. The derivatives carrying aggE exhibited the ten times higher adhesion ability to collagen and fibronectin, possess about two times higher adhesion to mucin and contribute to the increase of biofilm formation, comparing to the control strains. Analysis for the presence of virulence factors (cytolysin and gelatinase production), antibiotic resistance (antibiotic susceptibility) and genes (cylA, agg, gelE, esp, hylN, ace, efaAfs, and efaAfm) showed that BGGO9-28 was sensitive to all tested antibiotics, without hemolytic or gelatinase activity. This strain does not carry any of the tested genes encoding for known virulence factors. Results showed that BGGO9-28 was resistant to low pH and high concentrations of bile salts. Also, it adhered strongly to the Caco-2 human epithelial cell line. In conclusion, the results of this study indicate that the presence of AggE protein on the cell surface in enterococci is a desirable probiotic feature.
Collapse
Affiliation(s)
- Katarina Veljović
- Institute of Molecular Genetics and Genetic Engineering, University of Belgrade, Belgrade, Serbia
| | - Nikola Popović
- Institute of Molecular Genetics and Genetic Engineering, University of Belgrade, Belgrade, Serbia
| | - Marija Miljković
- Institute of Molecular Genetics and Genetic Engineering, University of Belgrade, Belgrade, Serbia
| | - Maja Tolinački
- Institute of Molecular Genetics and Genetic Engineering, University of Belgrade, Belgrade, Serbia
| | - Amarela Terzić-Vidojević
- Institute of Molecular Genetics and Genetic Engineering, University of Belgrade, Belgrade, Serbia
| | - Milan Kojić
- Institute of Molecular Genetics and Genetic Engineering, University of Belgrade, Belgrade, Serbia
| |
Collapse
|
48
|
Blanchet-Réthoré S, Bourdès V, Mercenier A, Haddar CH, Verhoeven PO, Andres P. Effect of a lotion containing the heat-treated probiotic strain Lactobacillus johnsonii NCC 533 on Staphylococcus aureus colonization in atopic dermatitis. Clin Cosmet Investig Dermatol 2017; 10:249-257. [PMID: 28721083 PMCID: PMC5501445 DOI: 10.2147/ccid.s135529] [Citation(s) in RCA: 54] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/23/2022]
Abstract
Objective Staphylococcus aureus dominates the skin microbiota in patients with atopic dermatitis (AD), with bacterial loads correlating with disease severity. The aim of this exploratory study was to investigate the effect of a cosmetic lotion containing heat-treated Lactobacillus johnsonii NCC 533 (HT La1) on S. aureus colonization in AD patients. Methods This open-label, multicenter study was performed in AD patients in Germany. First, detection of S. aureus was performed in all patients using the swab or scrub-wash method of sampling, followed by quantitative culture or quantitative polymerase chain reaction. Repeatability and reproducibility of all method combinations were evaluated to select the best combination of sampling and quantification. Second, a lotion containing HT La1 was applied to lesional skin twice daily for 3 weeks. Scoring using local objective SCORing Atopic Dermatitis (SCORAD), measurement of S. aureus load, and lesional microbiome analysis were performed before and after the 3-week treatment period. Results Thirty-one patients with AD were included in the study. All sampling and quantification methods were found to be robust, reproducible, and repeatable for assessing S. aureus load. For simplicity, a combination of swab and quantitative polymerase chain reaction was chosen to assess the efficacy of HT La1. Following application of a lotion containing HT La1 to AD lesions for 3 weeks, a reduction in S. aureus load was observed in patients, which correlated with a decrease in local objective SCORAD. Interestingly, high baseline skin concentrations of S. aureus were associated with good responses to the lotion. Conclusion This study demonstrated that the application of a lotion containing HT La1 to the lesional skin of patients with AD for 3 weeks controlled S. aureus colonization and was associated with local clinical improvement (SCORAD). These findings support further development of topical treatments containing heat-treated nonreplicating beneficial bacteria for patients with AD.
Collapse
Affiliation(s)
- Sandrine Blanchet-Réthoré
- CUTIS (Clinical Unit for Tests and Imaging of Skin), Evaluation Department, Nestlé Skin Health/Galderma Research and Development, Sophia-Antipolis, France
| | - Valérie Bourdès
- CUTIS (Clinical Unit for Tests and Imaging of Skin), Evaluation Department, Nestlé Skin Health/Galderma Research and Development, Sophia-Antipolis, France
| | - Annick Mercenier
- Host Microbiome Interaction Group, Gut Ecosystem Department, Institute of Nutritional Science, Nestlé Research Center, Lausanne, Switzerland
| | - Cyrille H Haddar
- Laboratory of Infectious Agents and Hygiene, University Hospital of Saint-Etienne and GIMAP (Mucosal Immunity and Pathogen Agents Group), Saint-Etienne, France
| | - Paul O Verhoeven
- Laboratory of Infectious Agents and Hygiene, University Hospital of Saint-Etienne and GIMAP (Mucosal Immunity and Pathogen Agents Group), Saint-Etienne, France
| | - Philippe Andres
- Medical Affairs Department, Galderma International, Paris, France
| |
Collapse
|
49
|
Calonghi N, Parolin C, Sartor G, Verardi L, Giordani B, Frisco G, Marangoni A, Vitali B. Interaction of vaginal Lactobacillus strains with HeLa cells plasma membrane. Benef Microbes 2017; 8:625-633. [PMID: 28618863 DOI: 10.3920/bm2016.0212] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/27/2023]
Abstract
Vaginal lactobacilli offer protection against recurrent urinary and vaginal infections. The precise mechanisms underlying the interaction between lactobacilli and the host epithelium remain poorly understood at the molecular level. Deciphering such events can provide valuable information on the mode of action of commensal and probiotic bacteria in the vaginal environment. We investigated the effects exerted by five Lactobacillus strains of vaginal origin (Lactobacillus crispatus BC1 and BC2, Lactobacillus gasseri BC9 and BC11 and Lactobacillus vaginalis BC15) on the physical properties of the plasma membrane in a cervical cell line (HeLa). The interaction of the vaginal lactobacilli with the cervical cells determined two kinds of effects on plasma membrane: (1) modification of the membrane polar lipid organisation and the physical properties (L. crispatus BC1 and L. gasseri BC9); (2) modification of α5β1 integrin organisation (L. crispatus BC2, L. gasseri BC11 and L. vaginalis BC15). These two mechanisms can be at the basis of the protective role of lactobacilli against Candida albicans adhesion. Upon stimulation with all Lactobacillus strains, we observed a reduction of the basal oxidative stress in HeLa cells that could be related to modifications in physical properties and organisation of the plasma membrane. These results confirm the strictly strain-specific peculiarities of Lactobacillus and deepen the understanding of the mechanisms underlying the health-promoting role of this genus within the vaginal ecosystem.
Collapse
Affiliation(s)
- N Calonghi
- 1 Department of Pharmacy and Biotechnology, University of Bologna, Via San Donato 15, 40127 Bologna, Italy
| | - C Parolin
- 1 Department of Pharmacy and Biotechnology, University of Bologna, Via San Donato 15, 40127 Bologna, Italy
| | - G Sartor
- 1 Department of Pharmacy and Biotechnology, University of Bologna, Via San Donato 15, 40127 Bologna, Italy
| | - L Verardi
- 1 Department of Pharmacy and Biotechnology, University of Bologna, Via San Donato 15, 40127 Bologna, Italy
| | - B Giordani
- 1 Department of Pharmacy and Biotechnology, University of Bologna, Via San Donato 15, 40127 Bologna, Italy
| | - G Frisco
- 1 Department of Pharmacy and Biotechnology, University of Bologna, Via San Donato 15, 40127 Bologna, Italy
| | - A Marangoni
- 2 Department of Experimental, Diagnostic and Specialty Medicine, University of Bologna, Via Massarenti 9, 40138 Bologna, Italy
| | - B Vitali
- 1 Department of Pharmacy and Biotechnology, University of Bologna, Via San Donato 15, 40127 Bologna, Italy
| |
Collapse
|
50
|
Message in a Bottle: Dialog between Intestine and Skin Modulated by Probiotics. Int J Mol Sci 2017; 18:ijms18061067. [PMID: 28598354 PMCID: PMC5485927 DOI: 10.3390/ijms18061067] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2017] [Revised: 05/10/2017] [Accepted: 05/11/2017] [Indexed: 12/14/2022] Open
Abstract
At the beginning, probiotics were used exclusively for gastrointestinal conditions. However, over the years, evidence has shown that probiotics exert systemic effects. In this review article, we will summarize recent reports that postulate probiotic treatment as an efficient one against skin pathologies, such as cancer, allergy, photoaging and skin infections. The focus will be restricted to oral probiotics that could potentially counteract the ultraviolet irradiation-induced skin alterations. Moreover, the possible underlying mechanisms by which probiotics can impact on the gut and exert their skin effects will be reviewed. Furthermore, how the local and systemic immune system is involved in the intestine-cutaneous crosstalk will be analyzed. In conclusion, this article will be divided into three core ideas: (a) probiotics regulate gut homeostasis; (b) gut and skin homeostasis are connected; (c) probiotics are a potentially effective treatment against skin conditions.
Collapse
|