1
|
Kim DY, Lee TS, Lee YJ, Ahn SY, Chu B, Jung DH, Kim YJ, Seo IS, Kim WG, Cho YJ, Hong JJ, Park JH. Lactobacillus reuteri NCHBL-005 improves wound healing by promoting the activation of fibroblasts through TLR2/MAPK signaling. Inflamm Regen 2025; 45:10. [PMID: 40211423 PMCID: PMC11983859 DOI: 10.1186/s41232-025-00370-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2024] [Accepted: 02/23/2025] [Indexed: 04/14/2025] Open
Abstract
BACKGROUND Wound healing is a complex physiological process essential for restoring tissue integrity following various injuries, ranging from minor, everyday incidents to post-surgical complications. Emerging studies have demonstrated that lactic acid bacteria (LAB) can offer benefits beyond gut health, extending their positive effects on skin health. This study investigated the potential of Lactobacillus reuteri NCHBL-005, a honeybee-derived probiotic strain, to enhance fibroblast-mediated wound healing. METHOD L929 cells and mouse embryonic fibroblasts (MEFs) were utilized as models to specifically target fibroblasts. To assess the wound healing potential in vitro, a scratch assay was performed, providing insights into wound closure. Additionally, we created wound models in mice to evaluate the in vivo effects of the treatment. RESULTS Our results showed that L. reuteri NCHBL-005 significantly accelerated wound closure in L929 fibroblast compared to other lactobacilli and exhibited superior efficacy in activating the mitogen-activated protein kinase (MAPK) pathway. Through MAPK inhibition assays, we confirmed that the wound healing effects of L. reuteri NCHBL-005 were MAPK-dependent, promoting fibroblast proliferation and differentiation. Notably, L. reuteri NCHBL-005 treatment did not facilitate wound healing in MEF cells derived from Toll-like-receptor 2 knockout (TLR2-/-) mice, highlighting the critical role of TLR2 in this mechanism. In vivo studies further corroborated these findings, in which topical administration of L. reuteri NCHBL-005 enhanced wound healing and stimulated fibroblast proliferation and activation, as confirmed by histopathological analysis. CONCLUSION These findings revealed that L. reuteri NCHBL-005 activates fibroblasts through TLR2 stimulation and subsequent MAPK pathway activation, suggesting its potential as a promising therapeutic candidate for wound management.
Collapse
Affiliation(s)
- Dong-Yeon Kim
- National Primate Research Centre, Korea Research Institute of Bioscience and Biotechnology (KRIBB), Cheongju, Chungcheongbuk, 28116, Republic of Korea
- Laboratory Animal Medicine, Animal Medical Institute, College of Veterinary Medicine, Chonnam National University, Buk-Gu, Gwangju, 61186, Republic of Korea
| | - Tae-Sung Lee
- Laboratory Animal Medicine, Animal Medical Institute, College of Veterinary Medicine, Chonnam National University, Buk-Gu, Gwangju, 61186, Republic of Korea
| | - Yun-Ji Lee
- Laboratory Animal Medicine, Animal Medical Institute, College of Veterinary Medicine, Chonnam National University, Buk-Gu, Gwangju, 61186, Republic of Korea
| | - So-Yeon Ahn
- Laboratory Animal Medicine, Animal Medical Institute, College of Veterinary Medicine, Chonnam National University, Buk-Gu, Gwangju, 61186, Republic of Korea
| | - Byeongsam Chu
- Nodcure, Inc., Buk-Gu, Gwangju, 61186, Republic of Korea
| | - Do-Hyeon Jung
- Laboratory Animal Medicine, Animal Medical Institute, College of Veterinary Medicine, Chonnam National University, Buk-Gu, Gwangju, 61186, Republic of Korea
| | - Yeong-Jun Kim
- Laboratory Animal Medicine, Animal Medical Institute, College of Veterinary Medicine, Chonnam National University, Buk-Gu, Gwangju, 61186, Republic of Korea
| | - In-Su Seo
- Laboratory Animal Medicine, Animal Medical Institute, College of Veterinary Medicine, Chonnam National University, Buk-Gu, Gwangju, 61186, Republic of Korea
| | - Wan-Gyu Kim
- Laboratory Animal Medicine, Animal Medical Institute, College of Veterinary Medicine, Chonnam National University, Buk-Gu, Gwangju, 61186, Republic of Korea
| | - Young Jin Cho
- Laboratory Animal Medicine, Animal Medical Institute, College of Veterinary Medicine, Chonnam National University, Buk-Gu, Gwangju, 61186, Republic of Korea
| | - Jung Joo Hong
- National Primate Research Centre, Korea Research Institute of Bioscience and Biotechnology (KRIBB), Cheongju, Chungcheongbuk, 28116, Republic of Korea
| | - Jong-Hwan Park
- Laboratory Animal Medicine, Animal Medical Institute, College of Veterinary Medicine, Chonnam National University, Buk-Gu, Gwangju, 61186, Republic of Korea.
- Nodcure, Inc., Buk-Gu, Gwangju, 61186, Republic of Korea.
| |
Collapse
|
2
|
Demirhan HK, Aksoy ZB, Karaca B, Kankilic T, Akcali KC, Kiran F. In vitro wound healing effects of postbiotics derived from the gut microbiota of long-lived blind mole rats, a model of healthy ageing. Wound Repair Regen 2025; 33:e70023. [PMID: 40247718 PMCID: PMC12006833 DOI: 10.1111/wrr.70023] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2024] [Revised: 03/01/2025] [Accepted: 03/25/2025] [Indexed: 04/19/2025]
Abstract
Chronic wounds represent a global public health burden to patients and healthcare professionals worldwide. Considering the unmet need for safe and effective therapeutic approaches for wound healing, research on discovering new bioactive materials that support all stages of wound healing is gaining importance. In this study, the wound-healing activity of postbiotics obtained from Limosilactobacillus reuteri EIR/Spx-2, isolated from the gut microbiota of long-lived blind mole rats (Nannospalax xanthodon), was investigated. Our results demonstrated that postbiotics exhibited a strong inhibitory effect against important skin pathogens, eliminated their biofilm formation, and downregulated the expression of genes involved in their quorum-sensing regulatory mechanisms. Furthermore, treatment with postbiotics resulted in a significant increase (23.82% ± 2.11%) in L929 fibroblast cell proliferation. Additionally, postbiotics applied on scratched fibroblast monolayer significantly accelerated the re-epithelialization by 66.78% ± 3.74%. The treatment also increased the mRNA expression and protein levels of COL1A1 in the early healing phase. Moreover, the intracellular ROS levels of L929 cells suppressed by H2O2 were significantly reduced, which could be attributed to the content of flavonoids (4.8 mg/g) and phenolic compounds (7.12 mg/g) in postbiotics, as well as their DPPH scavenging activity. After treatment with postbiotics, the mRNA levels of IL-6 (5.77-fold) and TNF-α (1.76-fold) and the amount of NO (79.25% ± 3.18%) were significantly decreased in LPS-induced murine macrophages. The diverse metabolite profile of postbiotics, as characterised using chromatographic techniques, exhibited a strong correlation with their biological activity across all stages of the wound healing process, highlighting their potential as promising candidates for wound healing applications.
Collapse
Affiliation(s)
- Hazal K. Demirhan
- Pharmabiotic Technologies Research Laboratory, Department of Biology, Faculty of ScienceAnkara UniversityAnkaraTurkey
- Graduate School of Natural and Applied SciencesAnkara UniversityAnkaraTurkey
| | | | - Basar Karaca
- Microbiology Research Laboratory, Department of Biology, Faculty of ScienceAnkara UniversityAnkaraTurkey
| | - Teoman Kankilic
- Department of Biotechnology, Faculty of ScienceNigde Omer Halisdemir UniversityNigdeTurkey
| | - Kamil C. Akcali
- Stem Cell InstituteAnkara UniversityAnkaraTurkey
- Department of Biophysics, Faculty of MedicineAnkara UniversityAnkaraTurkey
| | - Fadime Kiran
- Pharmabiotic Technologies Research Laboratory, Department of Biology, Faculty of ScienceAnkara UniversityAnkaraTurkey
| |
Collapse
|
3
|
Mercer SD, Doherty C, Singh G, Willmott T, Cheesapcharoen T, Teanpaisan R, O'Neill C, Ledder RG, McBain AJ. Lactobacillus lysates protect oral epithelial cells from pathogen-associated damage, increase secretion of pro-inflammatory cytokines and enhance barrier integrity. Sci Rep 2025; 15:5894. [PMID: 39966408 PMCID: PMC11836205 DOI: 10.1038/s41598-025-86914-y] [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: 07/26/2024] [Accepted: 01/14/2025] [Indexed: 02/20/2025] Open
Abstract
Periodontitis is a chronic gum disease characterised by inflammation and the loss of bone. We have explored the potential prophylactic effects of lysates from four Lactobacillus strains against the toxic effects of three periodontal pathogens (Porphyromonas gingivalis, Fusobacterium nucleatum, and Aggregatibacter actinomycetemcomitans). TR146 oral epithelial cells were pre-treated with Lactobacillus lysates (L. rhamnosus - GG, L. rhamnosus - SD11, L. reuteri and L. plantarum) and then challenged with pathogenic material (live cells, lysates, or supernatants). Cytokine analysis was performed on supernatants of cells treated with probiotic lysates from 1.5 h to 24 h. Effects of probiotic lysates on re-epithelialisation were determined using keratinocyte scratch assays, monitoring both migration and proliferation. Epithelial barrier function was observed after lysate addition by trans-epithelial electrical resistance (TEER) and by quantifying claudin-1 expression. Treatment of host cells with Lactobacillus lysates before pathogen exposure conferred significant protection against viability loss. Although extended pre-treatment did not generally increase protection, against live Aggregatibacter actinomycetemcomitans, significant increases in viability were seen after 24 h of pre-treatment for GG, SD11 and L. plantarum. Pro-inflammatory cytokines TNF-α, IP-10, IL-6, and IL-8 increased significantly with extended probiotic treatment, while IL-1β and IL-1α secretion significantly increased but remained constant over time. Secretion of the growth-promoting cytokine TGF-β increased after 3 h of treatment, however no increases in the regulatory cytokine IL-10 were recorded. Only exposure to SD11 significantly enhanced re-epithelialisation, TEER and claudin-1 expression while GG increased TEER but decreased claudin-1 expression. L. plantarum significantly inhibited re-epithelialisation but did not impact TEER or claudin-1 expression. All lysates significantly improved TEER in the presence of pathogenic material, demonstrating a protective effect on barrier function.
Collapse
Affiliation(s)
- Steven D Mercer
- Division of Pharmacy and Optometry, School of Health Sciences, Faculty of Biology, Medicine and Health, The University of Manchester, Manchester, UK.
- Division of Musculoskeletal and Dermatological Sciences, School of Health Sciences, Faculty of Biology, Medicine and Health, The University of Manchester, Manchester, UK.
| | - Christopher Doherty
- Division of Pharmacy and Optometry, School of Health Sciences, Faculty of Biology, Medicine and Health, The University of Manchester, Manchester, UK
| | - Gurdeep Singh
- Division of Musculoskeletal and Dermatological Sciences, School of Health Sciences, Faculty of Biology, Medicine and Health, The University of Manchester, Manchester, UK
- Lydia Becker Institute of Immunology and Inflammation, Faculty of Biology, Medicine and Health, The University of Manchester, Manchester, UK
| | - Thomas Willmott
- Division of Pharmacy and Optometry, School of Health Sciences, Faculty of Biology, Medicine and Health, The University of Manchester, Manchester, UK
- Institute of Infection, Veterinary and Ecological Sciences, Clinical Infection, Microbiology & Immunology, University of Liverpool, Liverpool, UK
| | - Tanaporn Cheesapcharoen
- Department of Conservative Dentistry, Faculty of Dentistry, Prince of Songkla University, Hat- Yai, Thailand
| | - Rawee Teanpaisan
- Medical Science Research and Innovation Institute, Prince of Songkla University, Hat-Yai, Thailand
| | - Catherine O'Neill
- Division of Musculoskeletal and Dermatological Sciences, School of Health Sciences, Faculty of Biology, Medicine and Health, The University of Manchester, Manchester, UK
| | - Ruth G Ledder
- Division of Pharmacy and Optometry, School of Health Sciences, Faculty of Biology, Medicine and Health, The University of Manchester, Manchester, UK
| | - Andrew J McBain
- Division of Pharmacy and Optometry, School of Health Sciences, Faculty of Biology, Medicine and Health, The University of Manchester, Manchester, UK.
| |
Collapse
|
4
|
Liu Z, Cao Q, Wang W, Wang B, Yang Y, Xian CJ, Li T, Zhai Y. The Impact of Lactobacillus reuteri on Oral and Systemic Health: A Comprehensive Review of Recent Research. Microorganisms 2024; 13:45. [PMID: 39858814 PMCID: PMC11767923 DOI: 10.3390/microorganisms13010045] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2024] [Revised: 12/24/2024] [Accepted: 12/26/2024] [Indexed: 01/27/2025] Open
Abstract
Oral diseases, particularly dental caries and periodontal disease, pose significant global health challenges. The imbalance of the oral microbiota plays a key role in the occurrence of these diseases, prompting researchers to seek new strategies to restore oral ecological balance. Lactobacillus reuteri is a Gram-positive rod-shaped bacterium that exists in various body parts of humans, including the gastrointestinal tract, urinary tract, skin, and so on. This species has a potentially positive impact on oral health and plays an important role in maintaining systemic health. Recent studies have explored the application of Lactobacillus reuteri in the prevention and treatment of oral diseases, and its impact on systemic health has also been preliminarily revealed. The current review summarizes the role of Lactobacillus reuteri in oral health and systemic health and outlines its potential applications in the future. Lactobacillus reuteri has shown promising prospects in treating non-communicable biofilm-dependent oral diseases, but its mechanism of action and efficacy still need further research. In addition, Lactobacillus reuteri has also displayed some potential benefits in promoting overall health. Future research should focus on revealing the specific pathways of action of Lactobacillus reuteri, screening for the most beneficial strains, determining the most effective drug delivery strategies, developing oral and systemic health products based on Lactobacillus reuteri, and ensuring their safety in clinical applications.
Collapse
Affiliation(s)
- Zihui Liu
- School of Stomatology, Henan University, Kaifeng 475004, China; (Z.L.); (Q.C.); (W.W.); (B.W.); (Y.Y.); (T.L.)
- Kaifeng Key Laboratory of Periodontal Tissue Engineering, Kaifeng 475000, China
| | - Qing Cao
- School of Stomatology, Henan University, Kaifeng 475004, China; (Z.L.); (Q.C.); (W.W.); (B.W.); (Y.Y.); (T.L.)
- Kaifeng Key Laboratory of Periodontal Tissue Engineering, Kaifeng 475000, China
| | - Wenqing Wang
- School of Stomatology, Henan University, Kaifeng 475004, China; (Z.L.); (Q.C.); (W.W.); (B.W.); (Y.Y.); (T.L.)
- Kaifeng Key Laboratory of Periodontal Tissue Engineering, Kaifeng 475000, China
| | - Bowen Wang
- School of Stomatology, Henan University, Kaifeng 475004, China; (Z.L.); (Q.C.); (W.W.); (B.W.); (Y.Y.); (T.L.)
- Kaifeng Key Laboratory of Periodontal Tissue Engineering, Kaifeng 475000, China
| | - Yilun Yang
- School of Stomatology, Henan University, Kaifeng 475004, China; (Z.L.); (Q.C.); (W.W.); (B.W.); (Y.Y.); (T.L.)
- Kaifeng Key Laboratory of Periodontal Tissue Engineering, Kaifeng 475000, China
| | - Cory J. Xian
- UniSA Clinical and Health Sciences, University of South Australia, Adelaide, SA 5001, Australia;
| | - Tiejun Li
- School of Stomatology, Henan University, Kaifeng 475004, China; (Z.L.); (Q.C.); (W.W.); (B.W.); (Y.Y.); (T.L.)
- Department of Oral Pathology, Peking University School and Hospital of Stomatology, Beijing 100081, China
| | - Yuankun Zhai
- School of Stomatology, Henan University, Kaifeng 475004, China; (Z.L.); (Q.C.); (W.W.); (B.W.); (Y.Y.); (T.L.)
- Kaifeng Key Laboratory of Periodontal Tissue Engineering, Kaifeng 475000, China
| |
Collapse
|
5
|
Moysidis M, Chorti A, Cheva A, Abba Deka I, Tzikos G, Kosmidis C, Koutelidakis I, Tsetis JK, Papavramidis T, Kotzampassi K. L. plantarum UBLP-40 Versus the Combined Formula of L. rhamnosus UBLP-58 and B. longum UBBL-64 in Excisional Wound Healing: A Cellular Perspective. Pharmaceuticals (Basel) 2024; 17:1414. [PMID: 39598326 PMCID: PMC11597307 DOI: 10.3390/ph17111414] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2024] [Revised: 10/13/2024] [Accepted: 10/14/2024] [Indexed: 11/29/2024] Open
Abstract
INTRODUCTION The utilization of probiotics in enhancing the active healing of skin wounds represents a burgeoning trend in contemporary medicine. Previous research has extensively explored wound healing mechanisms involving the strains of Lactiplantibacillus plantarum, Lacticaseibacillus rhamnosus, and Bifidobacterium longum. This study seeks to compare and interpret cellular findings derived from immunohistochemical and pathological applications. METHODS Three groups (the control, Lactiplantibacillus plantarum (RO1) group, and Lacticaseibacillus rhamnosus and Bifidobacterium longum (PRO2) group) underwent histological analysis, and microscopic cell counting were employed, offering insights into dynamic changes among neutrophils, lymphocytes, plasmacytes, mast cells, fibroblasts, and newly formed vessels across distinct treatment groups and temporal intervals. RESULTS The neutrophil count was found to be elevated in PRO2 on day 2, while the same group resulted in the highest decline on day 15. The number of fibroblasts peaked on day 4 for the PRO1 group, compared to the other two groups, which peaked on day 8. The lymphocyte count was the highest in the control group, while they peaked on day 4 in PRO2. The mast cells and plasmacytes were variable and sparse among all groups and time frames. Neovascularization was promoted by PRO1 and PRO2 groups on day 4 and remained high on day 8 for PRO2. CONCLUSIONS Probiotic strains can be beneficial to the human population and in assisting skin wound healing, each strain working differently and more effectively in different healing phases. Thus, a combined formula containing different probiotics to modulate various healing phases is desirable.
Collapse
Affiliation(s)
- Moysis Moysidis
- Department of Surgery, Aristotle University of Thessaloniki, 54636 Thessaloniki, Greece; (A.C.); (T.P.)
| | - Angeliki Chorti
- Department of Surgery, Aristotle University of Thessaloniki, 54636 Thessaloniki, Greece; (A.C.); (T.P.)
| | - Angeliki Cheva
- Department of Pathology, Medical School, Faculty of Health Sciences, Aristotle University of Thessaloniki, 54124 Thessaloniki, Greece (I.A.D.)
| | - Ioanna Abba Deka
- Department of Pathology, Medical School, Faculty of Health Sciences, Aristotle University of Thessaloniki, 54124 Thessaloniki, Greece (I.A.D.)
| | - Georgios Tzikos
- Department of Surgery, Aristotle University of Thessaloniki, 54636 Thessaloniki, Greece; (A.C.); (T.P.)
| | - Christoforos Kosmidis
- 3th Department of Surgery, AHEPA University Hospital, Medical School, Faculty of Health Sciences, Aristotle University of Thessaloniki, 54124 Thessaloniki, Greece
| | - Ioannis Koutelidakis
- 2nd Department of Surgery, G. Gennimatas University Hospital, Medical School, Faculty of Health Sciences, Aristotle University of Thessaloniki, 54124 Thessaloniki, Greece
| | - Joulia K. Tsetis
- Department of Surgery, Aristotle University of Thessaloniki, 54636 Thessaloniki, Greece; (A.C.); (T.P.)
| | - Theodossis Papavramidis
- Department of Surgery, Aristotle University of Thessaloniki, 54636 Thessaloniki, Greece; (A.C.); (T.P.)
| | - Katerina Kotzampassi
- Department of Surgery, Aristotle University of Thessaloniki, 54636 Thessaloniki, Greece; (A.C.); (T.P.)
| |
Collapse
|
6
|
Wang J, Li X, Zhao X, Yuan S, Dou H, Cheng T, Huang T, Lv Z, Tu Y, Shi Y, Ding X. Lactobacillus rhamnosus GG-derived extracellular vesicles promote wound healing via miR-21-5p-mediated re-epithelization and angiogenesis. J Nanobiotechnology 2024; 22:644. [PMID: 39427198 PMCID: PMC11490139 DOI: 10.1186/s12951-024-02893-8] [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: 06/01/2024] [Accepted: 10/02/2024] [Indexed: 10/21/2024] Open
Abstract
Extracellular vesicles (EVs), especially those derived from stem cells, have emerged as a novel treatment for promoting wound healing in regenerative medicine. However, the clinical application of mammalian cells-derived EVs is hindered by their high cost and low yields. Inspired by the ability of EVs to mediate interkingdom communication, we explored the therapeutic potential of EVs released by the probiotic strain Lactobacillus rhamnosus GG (LGG) in skin wound healing and elucidated the underlying mechanism involved. Using full-thickness skin wound-healing mouse models, we found that LGG-EVs accelerated wound healing procedures, including increased re-epithelialization and promoted angiogenesis. Using in vitro experiments, we further demonstrated that LGG-EVs boosted the proliferation and migration capacities of both epithelial and endothelial cells, as well as promoted endothelial tube formation. miRNA profiling analysis revealed that miR-21-5p was highly enriched in LGG-EVs and LGG-EV treatment significantly increased miR-21-5p level in recipient cells. Mechanically, LGG-EVs induced regulatory effects via miR-21-5p mediated metabolic signaling rewiring. Our results suggest that EVs derived from LGG could serve as a promising candidate for accelerating wound healing and possibly for treating chronic and impaired healing conditions.
Collapse
Affiliation(s)
- Juan Wang
- Institute of Geriatrics (Shanghai University), Affiliated Nantong Hospital of Shanghai University (The Sixth People's Hospital of Nantong), School of Medicine, Shanghai University, Nantong, 226011, China
- Shanghai Engineering Research Center of Organ Repair, School of Medicine, Shanghai University, Shanghai, 200444, China
| | - Xiaojie Li
- Institute of Geriatrics (Shanghai University), Affiliated Nantong Hospital of Shanghai University (The Sixth People's Hospital of Nantong), School of Medicine, Shanghai University, Nantong, 226011, China
- Shanghai Engineering Research Center of Organ Repair, School of Medicine, Shanghai University, Shanghai, 200444, China
| | - Xinyue Zhao
- Institute of Geriatrics (Shanghai University), Affiliated Nantong Hospital of Shanghai University (The Sixth People's Hospital of Nantong), School of Medicine, Shanghai University, Nantong, 226011, China
- Shanghai Engineering Research Center of Organ Repair, School of Medicine, Shanghai University, Shanghai, 200444, China
| | - Siqi Yuan
- Institute of Geriatrics (Shanghai University), Affiliated Nantong Hospital of Shanghai University (The Sixth People's Hospital of Nantong), School of Medicine, Shanghai University, Nantong, 226011, China
- Shanghai Engineering Research Center of Organ Repair, School of Medicine, Shanghai University, Shanghai, 200444, China
| | - Hanyu Dou
- Institute of Geriatrics (Shanghai University), Affiliated Nantong Hospital of Shanghai University (The Sixth People's Hospital of Nantong), School of Medicine, Shanghai University, Nantong, 226011, China
- Shanghai Engineering Research Center of Organ Repair, School of Medicine, Shanghai University, Shanghai, 200444, China
| | - Ting Cheng
- Institute of Translational Medicine, Shanghai University, Shanghai, 200444, China
| | - Taomin Huang
- Eye & ENT Hospital, Fudan University, Shanghai, 200031, China
| | - Zhi Lv
- Shanghai Inoherb R&D Center, Shanghai, 200444, China
| | - Yidong Tu
- Shanghai Inoherb R&D Center, Shanghai, 200444, China
| | - Yejiao Shi
- Institute of Translational Medicine, Shanghai University, Shanghai, 200444, China.
| | - Xiaolei Ding
- Institute of Geriatrics (Shanghai University), Affiliated Nantong Hospital of Shanghai University (The Sixth People's Hospital of Nantong), School of Medicine, Shanghai University, Nantong, 226011, China.
- Shanghai Engineering Research Center of Organ Repair, School of Medicine, Shanghai University, Shanghai, 200444, China.
- Department of Dermatology, Huashan Hospital, Fudan University, Shanghai, 200040, China.
| |
Collapse
|
7
|
Yin Z, Wang Y, Feng X, Liu C, Guan X, Liu S, Long Z, Miao Z, He F, Cheng R, Han Y, Li K. Lactobacillus rhamnosus GG and Bifidobacterium animalis subsp. lactis BB-12 promote infected wound healing via regulation of the wound microenvironment. Microb Biotechnol 2024; 17:e70031. [PMID: 39422648 PMCID: PMC11488118 DOI: 10.1111/1751-7915.70031] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2024] [Accepted: 09/30/2024] [Indexed: 10/19/2024] Open
Abstract
Infected wounds can result in complex clinical complications and delayed healing, presenting a significant global public health challenge. This study explored the effects of topical application of two probiotics, Lactobacillus rhamnosus GG (LGG) and Bifidobacterium animalis subsp. lactis BB-12, on the microenvironment of infected wounds and their impact on wound healing. LGG and BB-12 were applied separately and topically on the Staphylococcus aureus (S. aureus)-infected skin wounds of the rat model on a daily basis. Both probiotics significantly accelerated wound healing, demonstrated by enhanced granulation tissue formation and increased collagen deposition, with BB-12 showing superior efficacy. LGG and BB-12 both effectively inhibited neutrophil infiltration and decreased the expression of pro-inflammatory cytokines tumor necrosis factor-α (TNF-α) and interleukin-6 (IL-6). Notably, BB-12 markedly reduced IL-6 levels, while LGG significantly lowered TNF-α, transforming growth factor-β (TGF-β) and vascular endothelial growth factor (VEGF). Additionally, both probiotics promoted macrophage polarization towards the anti-inflammatory M2 phenotype. Microbiota analysis revealed that LGG and BB-12 significantly decreased the abundance of pathogenic bacteria (e.g. Staphylococcus and Proteus) and increased the proportion of beneficial bacteria (e.g. Corynebacterium). Particularly, BB-12 was more effective in reducing Staphylococcus abundance, whereas LGG excelled in promoting Corynebacterium growth. These findings suggest the ability of LGG and BB-12 to modulate the wound microenvironment, enhance wound healing and provide valuable insights for the management of infected wounds.
Collapse
Affiliation(s)
- Zhe Yin
- Sichuan University—The Hong Kong Polytechnic University Institute for Disaster Management and ReconstructionChengduChina
- Medicine and Engineering Interdisciplinary Research Laboratory of Nursing & Materials, West China Hospital, Sichuan University/West China School of NursingSichuan UniversityChengduChina
- Department of GastroenterologyAffiliated Tumor Hospital of Xinjiang Medical UniversityUrumqiChina
| | - Yilin Wang
- Medicine and Engineering Interdisciplinary Research Laboratory of Nursing & Materials, West China Hospital, Sichuan University/West China School of NursingSichuan UniversityChengduChina
| | - Xiaojuan Feng
- Department of General SurgeryWest China Hospital, Sichuan UniversityChengduChina
| | - Changqing Liu
- Medicine and Engineering Interdisciplinary Research Laboratory of Nursing & Materials, West China Hospital, Sichuan University/West China School of NursingSichuan UniversityChengduChina
| | - Xiaoyang Guan
- School of Fashion and TextilesThe Hong Kong Polytechnic UniversityKowloonHong Kong SARChina
| | - Shuyan Liu
- Medicine and Engineering Interdisciplinary Research Laboratory of Nursing & Materials, West China Hospital, Sichuan University/West China School of NursingSichuan UniversityChengduChina
| | - Zhanyi Long
- Medicine and Engineering Interdisciplinary Research Laboratory of Nursing & Materials, West China Hospital, Sichuan University/West China School of NursingSichuan UniversityChengduChina
| | - Zhonghua Miao
- Department of Clinical Nutrition, West China Second HospitalSichuan UniversityChengduChina
| | - Fang He
- Department of Nutrition and Food Hygiene, West China School of Public Health and West China Fourth HospitalSichuan UniversityChengduChina
| | - Ruyue Cheng
- Department of Nutrition and Food Hygiene, West China School of Public Health and West China Fourth HospitalSichuan UniversityChengduChina
| | - Yanting Han
- Medicine and Engineering Interdisciplinary Research Laboratory of Nursing & Materials, West China Hospital, Sichuan University/West China School of NursingSichuan UniversityChengduChina
| | - Ka Li
- Medicine and Engineering Interdisciplinary Research Laboratory of Nursing & Materials, West China Hospital, Sichuan University/West China School of NursingSichuan UniversityChengduChina
| |
Collapse
|
8
|
Karimi F, Montazeri-Najafabady N, Mohammadi F, Azadi A, Koohpeyma F, Gholami A. A potential therapeutic strategy of an innovative probiotic formulation toward topical treatment of diabetic ulcer: an in vivo study. Nutr Diabetes 2024; 14:66. [PMID: 39164243 PMCID: PMC11335896 DOI: 10.1038/s41387-024-00320-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/29/2023] [Revised: 07/22/2024] [Accepted: 07/24/2024] [Indexed: 08/22/2024] Open
Abstract
BACKGROUND The probiotic potential of Lacticacid bacteria has been studied in various medical complications, from gastrointestinal diseases to antibiotic resistance infections recently. Moreover, diabetic ulcer (DU) is known as one of the most significant global healthcare concerns, which comprehensively impacts the quality of life for these patients. Given that the conventional treatments of DUs have failed to prevent later complications completely, developing alternative therapies seems to be crucial. METHODS We designed the stable oleogel-based formulation of viable probiotic cells, including Lactobacillus rhamnosus (L. rhamnosus), Lactobacillus casei (L. casei), Lactobacillus fermentum (L. fermentum), and Lactobacillus acidophilus (L. acidophilus) individually to investigate their effect on wound healing process as an in vivo study. The wound repair process was closely monitored regarding morphology, biochemical, and histopathological changes over two weeks and compared it with the effects of topical tetracycline as an antibiotic approach. Furthermore, the antibiofilm activity of probiotic bacteria was assessed against some common pathogens. RESULTS The findings indicated that all tested lactobacillus groups (excluded L. casei) included in the oleogel-based formulation revealed a high potential for repairing damaged skin due to the considerably more levels of hydroxyproline content of tissue samples along with the higher numerical density of mature fibroblasts cell and volume density of hair follicles, collagen fibrils, and neovascularization in comparison with antibiotic and control groups. L. acidophilus and L. rhamnosus showed the best potential of wound healing among all lactobacillus species, groups treated by tetracycline and control groups. Besides, L. rhamnosus showed a significant biofilm inhibition activity against tested pathogens. CONCLUSIONS This experiment demonstrated that the designed formulations containing probiotics, particularly L. acidophilus and L. rhamnosus, play a central role in manipulating diabetic wound healing. It could be suggested as an encouraging nominee for diabetic wound-healing alternative approaches, though further studies in detailed clinical trials are needed.
Collapse
Affiliation(s)
- Farkhonde Karimi
- Biotechnology Research Center, Shiraz University of Medical Sciences, Shiraz, Iran
- Department of Pharmaceutical Biotechnology, School of Pharmacy, Shiraz University of Medical Sciences, Shiraz, Fars, Iran
| | - Nima Montazeri-Najafabady
- Biotechnology Research Center, Shiraz University of Medical Sciences, Shiraz, Iran
- Endocrine and Metabolism Research Center, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Fatemeh Mohammadi
- Biotechnology Research Center, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Amir Azadi
- Department of Pharmaceutics, School of Pharmacy, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Farhad Koohpeyma
- Endocrine and Metabolism Research Center, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Ahmad Gholami
- Biotechnology Research Center, Shiraz University of Medical Sciences, Shiraz, Iran.
- Department of Pharmaceutical Biotechnology, School of Pharmacy, Shiraz University of Medical Sciences, Shiraz, Fars, Iran.
| |
Collapse
|
9
|
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
|
10
|
Li Y, Zhang L, He M, Zhao Y. Sequence analysis of microbiota in clinical human cases with diabetic foot ulcers from China. Heliyon 2024; 10:e34368. [PMID: 39104504 PMCID: PMC11298921 DOI: 10.1016/j.heliyon.2024.e34368] [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: 03/24/2024] [Revised: 07/03/2024] [Accepted: 07/08/2024] [Indexed: 08/07/2024] Open
Abstract
Background Diabetic foot ulcers (DFU) seriously threaten the health and quality of life of patients. The microbiota is the primary reason for the refractory and high recurrence of DFU. This study aimed to determine the wound microbiota at different DFU stages. Methods Wound samples were collected from 48 patients with DFU and divided into three phases: inflammatory (I, n = 49), proliferation (P, n = 22), and remodeling (R, n = 19). The wound samples obtained at different stages were then subjected to 16S rRNA gene sequencing. The number of operational taxonomic units (OTUs) in the different groups was calculated according to the criterion of 97 % sequence similarity. The diversity of the microbiota differentially presented bacterial taxa at the phylum and genus levels, and important phyla and genera in the different groups were further explored. Results After sequencing, 3351, 925, and 777 OTUs were observed in groups I, P, and R, respectively, and 175 OTUs overlapped. Compared with the inflammatory stage, the α-diversity of wound microbiota at proliferation and remodeling stages was significantly decreased (P < 0.05). At the phylum level, Firmicutes, Proteobacteria, Actinobacteriota, and Bacteroidota were the dominant phyla, accounting for more than 90 % of all the phyla. At the genus level, Random Forest and linear discriminant analysis effect size analyses showed that Peptoniphilus, Lactobacillus, Prevotella, Veillonella, Dialister, Streptococcus, and Ruminococcus were the signature wound microbiota for the inflammatory stage; Anaerococcus, Ralstonia, Actinomyces, and Akkermansia were important species for the proliferation stage; and the crucial genera for the remodeling stage were Enterobacter, Pseudomonas, Sondgrassella, Bifidobacterium, and Faecalibacterium. Conclusions There were significant differences in the composition and structure of the wound microbiota in patients with DFU at different stages, which may lay a foundation for effectively promoting wound healing in DFU.
Collapse
Affiliation(s)
- Ying Li
- The Ninth Clinical School of Shanxi Medical University, Taiyuan Central Hospital, Taiyuan, 030000, Shanxi, China
| | - Li Zhang
- Department of Endocrinology, Taiyuan Central Hospital, Taiyuan, 030000, Shanxi, China
| | - Meifang He
- Department of Endocrinology, Taiyuan Central Hospital, Taiyuan, 030000, Shanxi, China
| | - Yuebin Zhao
- Department of Endocrinology, Taiyuan Central Hospital, Taiyuan, 030000, Shanxi, China
| |
Collapse
|
11
|
Catella J, Guillot N, Nader E, Skinner S, Poutrel S, Hot A, Connes P, Fromy B. Controversies in the pathophysiology of leg ulcers in sickle cell disease. Br J Haematol 2024; 205:61-70. [PMID: 38867511 DOI: 10.1111/bjh.19584] [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: 12/22/2023] [Accepted: 05/27/2024] [Indexed: 06/14/2024]
Abstract
Patients with sickle cell disease (SCD) often experience painful vaso-occlusive crises and chronic haemolytic anaemia, as well as various acute and chronic complications, such as leg ulcers. Leg ulcers are characterized by their unpredictability, debilitating pain and prolonged healing process. The pathophysiology of SCD leg ulcers is not well defined. Known risk factors include male gender, poor social conditions, malnutrition and a lack of compression therapy when oedema occurs. Leg ulcers typically start with spontaneous pain, followed by induration, hyperpigmentation, blister formation and destruction of the epidermis. SCD is characterized by chronic haemolysis, increased oxidative stress and decreased nitric oxide bioavailability, which promote ischaemia and inflammation and consequently impair vascular function in the skin. This cutaneous vasculopathy, coupled with venostasis around the ankle, creates an ideal environment for local vaso-occlusive crises, which can result in the development of leg ulcers that resemble arterial ulcers. Following the development of the ulcer, healing is hindered as a result of factors commonly observed in venous ulceration, including venous insufficiency, oedema and impaired angiogenesis. All of these factors are modulated by genetic factors. However, our current understanding of these genetic factors remains limited and does not yet enable us to accurately predict ulceration susceptibility.
Collapse
Affiliation(s)
- Judith Catella
- Service de Médecine Interne et Vasculaire, Hôpital Edouard Herriot, Hospices Civils de Lyon, Lyon, France
- Laboratoire d'Excellence du Globule Rouge (Labex GR-Ex), Sorbonne, Paris, France
- Laboratoire Interuniversitaire de Biologie de la Motricité (LIBM) EA7424, Equipe "Biologie Vasculaire et du Globule Rouge", Université Claude Bernard Lyon 1, Université de Lyon, Lyon, France
- Laboratoire de Biologie Tissulaire et Ingénierie Thérapeutique (LBTI UMR 5305), CNRS/Université Claude Bernard Lyon 1, Lyon, France
| | - Nicolas Guillot
- Laboratoire d'Excellence du Globule Rouge (Labex GR-Ex), Sorbonne, Paris, France
- Laboratoire Interuniversitaire de Biologie de la Motricité (LIBM) EA7424, Equipe "Biologie Vasculaire et du Globule Rouge", Université Claude Bernard Lyon 1, Université de Lyon, Lyon, France
| | - Elie Nader
- Laboratoire d'Excellence du Globule Rouge (Labex GR-Ex), Sorbonne, Paris, France
- Laboratoire Interuniversitaire de Biologie de la Motricité (LIBM) EA7424, Equipe "Biologie Vasculaire et du Globule Rouge", Université Claude Bernard Lyon 1, Université de Lyon, Lyon, France
| | - Sarah Skinner
- Clinical Research and Epidemiology Unit, Montpellier University, Montpellier, France
| | - Solène Poutrel
- Service de Médecine Interne et Vasculaire, Hôpital Edouard Herriot, Hospices Civils de Lyon, Lyon, France
- Laboratoire d'Excellence du Globule Rouge (Labex GR-Ex), Sorbonne, Paris, France
- Laboratoire Interuniversitaire de Biologie de la Motricité (LIBM) EA7424, Equipe "Biologie Vasculaire et du Globule Rouge", Université Claude Bernard Lyon 1, Université de Lyon, Lyon, France
| | - Arnaud Hot
- Service de Médecine Interne et Vasculaire, Hôpital Edouard Herriot, Hospices Civils de Lyon, Lyon, France
- Laboratoire d'Excellence du Globule Rouge (Labex GR-Ex), Sorbonne, Paris, France
- Laboratoire Interuniversitaire de Biologie de la Motricité (LIBM) EA7424, Equipe "Biologie Vasculaire et du Globule Rouge", Université Claude Bernard Lyon 1, Université de Lyon, Lyon, France
| | - Philippe Connes
- Laboratoire d'Excellence du Globule Rouge (Labex GR-Ex), Sorbonne, Paris, France
- Laboratoire Interuniversitaire de Biologie de la Motricité (LIBM) EA7424, Equipe "Biologie Vasculaire et du Globule Rouge", Université Claude Bernard Lyon 1, Université de Lyon, Lyon, France
| | - Berengère Fromy
- Laboratoire de Biologie Tissulaire et Ingénierie Thérapeutique (LBTI UMR 5305), CNRS/Université Claude Bernard Lyon 1, Lyon, France
| |
Collapse
|
12
|
Leser T, Baker A. Molecular Mechanisms of Lacticaseibacillus rhamnosus, LGG ® Probiotic Function. Microorganisms 2024; 12:794. [PMID: 38674738 PMCID: PMC11051730 DOI: 10.3390/microorganisms12040794] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2024] [Revised: 04/10/2024] [Accepted: 04/12/2024] [Indexed: 04/28/2024] Open
Abstract
To advance probiotic research, a comprehensive understanding of bacterial interactions with human physiology at the molecular and cellular levels is fundamental. Lacticaseibacillus rhamnosus LGG® is a bacterial strain that has long been recognized for its beneficial effects on human health. Probiotic effector molecules derived from LGG®, including secreted proteins, surface-anchored proteins, polysaccharides, and lipoteichoic acids, which interact with host physiological processes have been identified. In vitro and animal studies have revealed that specific LGG® effector molecules stimulate epithelial cell survival, preserve intestinal barrier integrity, reduce oxidative stress, mitigate excessive mucosal inflammation, enhance IgA secretion, and provide long-term protection through epigenetic imprinting. Pili on the cell surface of LGG® promote adhesion to the intestinal mucosa and ensure close contact to host cells. Extracellular vesicles produced by LGG® recapitulate many of these effects through their cargo of effector molecules. Collectively, the effector molecules of LGG® exert a significant influence on both the gut mucosa and immune system, which promotes intestinal homeostasis and immune tolerance.
Collapse
Affiliation(s)
- Thomas Leser
- Future Labs, Human Health Biosolutions, Novonesis, Kogle Alle 6, 2970 Hoersholm, Denmark;
| | | |
Collapse
|
13
|
Al-Najjar MAA, Abdulrazzaq SB, Alzaghari LF, Mahmod AI, Omar A, Hasen E, Athamneh T, Talib WH, Chellappan DK, Barakat M. Evaluation of immunomodulatory potential of probiotic conditioned medium on murine macrophages. Sci Rep 2024; 14:7126. [PMID: 38531887 DOI: 10.1038/s41598-024-56622-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2023] [Accepted: 03/08/2024] [Indexed: 03/28/2024] Open
Abstract
Probiotics are a mixture of beneficial live bacteria and/or yeasts that naturally exist in our bodies. Recently, numerous studies have focused on the immunostimulatory effects of single-species or killed multi-species probiotic conditioned mediums on macrophages. This study investigates the immunostimulatory effect of commercially available active, multi-species probiotic conditioned medium (CM) on RAW264.7 murine macrophages. The probiotic CM was prepared by culturing the commercially available probiotic in a cell-culture medium overnight at 37 °C, followed by centrifugation and filter-sterilization to be tested on macrophages. The immunostimulatory effect of different dilution percentages (50%, 75%, 100%) of CM was examined using the MTT assay, proinflammatory cytokine (tumor necrosis factor TNF-alpha) production in macrophages, migration, and phagocytosis assays. For all the examined CM ratios, the percentages of cell viability were > 80%. Regarding the migration scratch, TNF-alpha and phagocytosis assays, CM demonstrated a concentration-dependent immunostimulatory effect. However, the undiluted CM (100%) showed a significant (p-value < 0.05) stimulatory effect compared to the positive and negative controls. The findings suggest that the secretions and products of probiotics, as measured in the CM, may be closely associated with their immune-boosting effects. Understanding this relationship between probiotic secretions and immune function is crucial for further exploring the potential benefits of probiotics in enhancing overall health and well-being.
Collapse
Affiliation(s)
| | | | | | | | - Amin Omar
- Faculty of Pharmacy, Applied Science Private University, 11937, Amman, Jordan
| | - Eliza Hasen
- MEA Research Center, Middle East University, Amman, Jordan
| | - Tamara Athamneh
- Institute of Nanotechnology, Jordan University of Science and Technology, Irbid, Jordan
| | - Wamidh H Talib
- Faculty of Allied Medical Sciences, Applied Science Private University, 11937, Amman, Jordan
| | - Dinesh Kumar Chellappan
- Department of Life Sciences, School of Pharmacy, International Medical University, 57000, Bukit Jalil, Kuala Lumpur, Malaysia
| | - Muna Barakat
- Faculty of Pharmacy, Applied Science Private University, 11937, Amman, Jordan.
| |
Collapse
|
14
|
Yang Y, Huang J, Zeng A, Long X, Yu N, Wang X. The role of the skin microbiome in wound healing. BURNS & TRAUMA 2024; 12:tkad059. [PMID: 38444635 PMCID: PMC10914219 DOI: 10.1093/burnst/tkad059] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/11/2023] [Revised: 08/05/2023] [Accepted: 11/21/2023] [Indexed: 03/07/2024]
Abstract
The efficient management of skin wounds for rapid and scarless healing represents a major clinical unmet need. Nonhealing skin wounds and undesired scar formation impair quality of life and result in high healthcare expenditure worldwide. The skin-colonizing microbiota contributes to maintaining an intact skin barrier in homeostasis, but it also participates in the pathogenesis of many skin disorders, including aberrant wound healing, in many respects. This review focuses on the composition of the skin microbiome in cutaneous wounds of different types (i.e. acute and chronic) and with different outcomes (i.e. nonhealing and hypertrophic scarring), mainly based on next-generation sequencing analyses; furthermore, we discuss the mechanistic insights into host-microbe and microbe-microbe interactions during wound healing. Finally, we highlight potential therapeutic strategies that target the skin microbiome to improve healing outcomes.
Collapse
Affiliation(s)
- Yuyan Yang
- Department of Plastic and Reconstructive Surgery, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, No. 1, Shuaifuyuan, Dongcheng District, Beijing, 100005, China
| | - Jiuzuo Huang
- Department of Plastic and Reconstructive Surgery, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, No. 1, Shuaifuyuan, Dongcheng District, Beijing, 100005, China
| | - Ang Zeng
- Department of Plastic and Reconstructive Surgery, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, No. 1, Shuaifuyuan, Dongcheng District, Beijing, 100005, China
| | - Xiao Long
- Department of Plastic and Reconstructive Surgery, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, No. 1, Shuaifuyuan, Dongcheng District, Beijing, 100005, China
| | - Nanze Yu
- Department of Plastic and Reconstructive Surgery, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, No. 1, Shuaifuyuan, Dongcheng District, Beijing, 100005, China
| | - Xiaojun Wang
- Department of Plastic and Reconstructive Surgery, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, No. 1, Shuaifuyuan, Dongcheng District, Beijing, 100005, China
| |
Collapse
|
15
|
Lee YJ, Choi JH, Kang KK, Sung SE, Lee S, Sung M, Seo MS, Park JH. Antioxidant and Antimelanogenic Activities of Lactobacillus kunkeei NCHBL-003 Isolated from Honeybees. Microorganisms 2024; 12:188. [PMID: 38258014 PMCID: PMC10818717 DOI: 10.3390/microorganisms12010188] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/28/2023] [Revised: 01/11/2024] [Accepted: 01/14/2024] [Indexed: 01/24/2024] Open
Abstract
Excessive reactive oxygen species production can detrimentally impact skin cell physiology, resulting in cell growth arrest, melanogenesis, and aging. Recent clinical studies have found that lactic acid bacteria have a special effect directly or indirectly on skin organs, but the exact mechanism has not been elucidated. In this study, we investigated the mechanisms underlying the antioxidant protective effect and the inhibitory effect on melanin synthesis of Lactobacillus kunkeei culture supernatant (CSK), isolated from Apis mellifera Linnaeus (the Western honeybee). CSK exhibited notable efficacy in promoting cell migration and wound healing under oxidative stress, surpassing the performance of other strains. CSK pretreatment significantly upregulated the expression of Nrf2/HO-1 (nuclear factor erythroid 2-related factor 2/heme oxygenase-1), a key player in cellular defenses against oxidative stress, relative to the control H2O2-treated cells. The DCF-DA (dichloro-dihydro-fluorescein diacetate) assay results confirmed that CSK's ability to enhance Nrf2 and HO-1 expression aligns with its robust ability to remove H2O2-induced reactive oxygen species. Furthermore, CSK upregulated MAPK (mitogen-activated protein kinase) phosphorylation, an upstream signal for HO-1 expression, and MAPK inhibitors compromised the wound-healing effect of CSK. Additionally, CSK exhibited inhibitory effects on melanin synthesis, downregulating melanogenesis-related genes in B16F10 cells. Thus, the present study demonstrated that CSK exhibited antioxidant effects by activating the Nrf2/HO-1 pathway through MAPK phosphorylation, thereby restoring cell migration and demonstrating inhibitory effects on melanin production. These findings emphasize the antioxidant and antimelanogenic potential of CSK, suggesting its potential use as a therapeutic agent, promoting wound healing, and as an active ingredient in skin-lightening cosmetics.
Collapse
Affiliation(s)
- Yeon-Ji Lee
- Preclinical Research Center, Daegu-Gyeongbuk Medical Innovation Foundation, Daegu 41061, Republic of Korea; (Y.-J.L.); (J.-H.C.); (K.-K.K.); (S.-E.S.); (S.L.); (M.S.)
| | - Joo-Hee Choi
- Preclinical Research Center, Daegu-Gyeongbuk Medical Innovation Foundation, Daegu 41061, Republic of Korea; (Y.-J.L.); (J.-H.C.); (K.-K.K.); (S.-E.S.); (S.L.); (M.S.)
| | - Kyung-Ku Kang
- Preclinical Research Center, Daegu-Gyeongbuk Medical Innovation Foundation, Daegu 41061, Republic of Korea; (Y.-J.L.); (J.-H.C.); (K.-K.K.); (S.-E.S.); (S.L.); (M.S.)
| | - Soo-Eun Sung
- Preclinical Research Center, Daegu-Gyeongbuk Medical Innovation Foundation, Daegu 41061, Republic of Korea; (Y.-J.L.); (J.-H.C.); (K.-K.K.); (S.-E.S.); (S.L.); (M.S.)
| | - Sijoon Lee
- Preclinical Research Center, Daegu-Gyeongbuk Medical Innovation Foundation, Daegu 41061, Republic of Korea; (Y.-J.L.); (J.-H.C.); (K.-K.K.); (S.-E.S.); (S.L.); (M.S.)
| | - Minkyoung Sung
- Preclinical Research Center, Daegu-Gyeongbuk Medical Innovation Foundation, Daegu 41061, Republic of Korea; (Y.-J.L.); (J.-H.C.); (K.-K.K.); (S.-E.S.); (S.L.); (M.S.)
| | - Min-Soo Seo
- Laboratory of Veterinary Tissue Engineering, College of Veterinary Medicine, Kyungpook National University, 80 Daehak-ro, Buk-gu, Daegu 41566, Republic of Korea
| | - Jong-Hwan Park
- Laboratory Animal Medicine, College of Veterinary Medicine and the Brain Korea 21 PLUS Project Team, Chonnam National University, 77 Yongbong-ro, Buk-gu, Gwangju 61186, Republic of Korea
| |
Collapse
|
16
|
Kuhn T, Aljohmani A, Frank N, Zielke L, Mehanny M, Laschke MW, Koch M, Hoppstädter J, Kiemer AK, Yildiz D, Fuhrmann G. A cell-free, biomimetic hydrogel based on probiotic membrane vesicles ameliorates wound healing. J Control Release 2024; 365:969-980. [PMID: 38070602 DOI: 10.1016/j.jconrel.2023.12.011] [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: 03/31/2023] [Revised: 11/21/2023] [Accepted: 12/04/2023] [Indexed: 12/22/2023]
Abstract
Probiotic bacteria, such as Lactobacilli, have been shown to elicit beneficial effects in various tissue regeneration applications. However, their formulation as living bacteria is challenging, and their therapeutic use as proliferating microorganisms is especially limited in immunocompromised patients. Here, we propose a new therapeutic avenue to circumvent these shortcomings by developing a bacteriomimetic hydrogel based on membrane vesicles (MVs) produced by Lactobacilli. We coupled MVs from Lactobacillus plantarum and Lactobacillus casei, respectively, to the surface of synthetic microparticles, and embedded those bacteriomimetics into a pharmaceutically applicable hydrogel matrix. The wound microenvironment changes during the wound healing process, including adaptions of the pH and changes of the oxygen supply. We thus performed proteomic characterization of the MVs harvested under different culture conditions and identified characteristic proteins related to the biological effect of the probiotics in every culture state. In addition, we highlight a number of unique proteins expressed and sorted into the MVs for every culture condition. Using different in vitro models, we demonstrated that increased cell migration and anti-inflammatory effects of the bacteriomimetic microparticles were dependent on the culture condition of the secreting bacteria. Finally, we demonstrated the bacteriomimetic hydrogel's ability to improve healing in an in vivo mouse full-thickness wound model. Our results create a solid basis for the future application of probiotic-derived vesicles in the treatment of inflammatory dispositions and stimulates the initiation of further preclinical trials.
Collapse
Affiliation(s)
- Thomas Kuhn
- Helmholtz Institute for Pharmaceutical Research Saarland (HIPS), Helmholtz Centre for Infection Research (HZI), Campus E8.1, Saarbrücken 66123, Germany; Department of Pharmacy, Saarland University, Campus E8.1, 66123 Saarbrücken, Germany
| | - Ahmad Aljohmani
- Institute of Experimental and Clinical Pharmacology and Toxicology, PZMS, Saarland University, 66421 Homburg, Germany
| | - Nicolas Frank
- Helmholtz Institute for Pharmaceutical Research Saarland (HIPS), Helmholtz Centre for Infection Research (HZI), Campus E8.1, Saarbrücken 66123, Germany; Department of Pharmacy, Saarland University, Campus E8.1, 66123 Saarbrücken, Germany
| | - Lina Zielke
- Helmholtz Institute for Pharmaceutical Research Saarland (HIPS), Helmholtz Centre for Infection Research (HZI), Campus E8.1, Saarbrücken 66123, Germany
| | - Mina Mehanny
- Helmholtz Institute for Pharmaceutical Research Saarland (HIPS), Helmholtz Centre for Infection Research (HZI), Campus E8.1, Saarbrücken 66123, Germany; Department of Pharmaceutics and Industrial Pharmacy, Faculty of Pharmacy, Ain Shams University, 11566 Cairo, Egypt
| | - Matthias W Laschke
- Institute for Clinical and Experimental Surgery, Saarland University, Homburg, Germany
| | - Marcus Koch
- INM - Leibniz Institute for New Materials, Campus D2 2, 66123, Saarbrücken, Germany
| | - Jessica Hoppstädter
- Department of Pharmacy, Saarland University, Campus E8.1, 66123 Saarbrücken, Germany
| | - Alexandra K Kiemer
- Department of Pharmacy, Saarland University, Campus E8.1, 66123 Saarbrücken, Germany
| | - Daniela Yildiz
- Institute of Experimental and Clinical Pharmacology and Toxicology, PZMS, Saarland University, 66421 Homburg, Germany.
| | - Gregor Fuhrmann
- Helmholtz Institute for Pharmaceutical Research Saarland (HIPS), Helmholtz Centre for Infection Research (HZI), Campus E8.1, Saarbrücken 66123, Germany; Department of Pharmacy, Saarland University, Campus E8.1, 66123 Saarbrücken, Germany; Friedrich-Alexander-Universität Erlangen-Nürnberg, Department of Biology, Pharmaceutical Biology, Staudtstr. 5, 91058 Erlangen, Germany; FAU NeW - Research Center New Bioactive Compounds, Nikolaus-Fiebiger-Str. 10, 91058 Erlangen, Germany.
| |
Collapse
|
17
|
Hong J, Son M, Sin J, Kim H, Chung DK. Nanoparticles of Lactiplantibacillus plantarum K8 Reduce Staphylococcus aureus Respiratory Infection and Tumor Necrosis Factor Alpha- and Interferon Gamma-Induced Lung Inflammation. Nutrients 2023; 15:4728. [PMID: 38004123 PMCID: PMC10675637 DOI: 10.3390/nu15224728] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2023] [Revised: 10/26/2023] [Accepted: 11/07/2023] [Indexed: 11/26/2023] Open
Abstract
Multiple studies have confirmed that Lactiplantibacillus plantarum has beneficial effects in respiratory diseases, including respiratory tract infections, asthma, and chronic obstructive pulmonary disease. However, the role of L. plantarum lysates in respiratory diseases is unclear. Staphylococcus aureus infects the lungs of mice, recruits immune cells, and induces structural changes in alveoli. Lung diseases can be further aggravated by inflammatory cytokines such as CCL2 and interleukin (IL)-6. In in vivo studies, L. plantarum K8 nanoparticles (K8NPs) restored lung function and prevented lung damage caused by S. aureus infection. They inhibited the S. aureus infection and the infiltration of immune cells and prevented the increase in goblet cell numbers in the lungs of S. aureus-infected mice. K8NPs suppressed the expression of CCL2 and IL-6, which were increased by the combination treatment of tumor necrosis factor alpha and interferon gamma (TI), in a dose-dependent manner. In in vitro studies, the anti-inflammatory effect of K8NPs in TI-treated A549 cells and TI-injected mice occurred through the reduction in activated mitogen-activated protein kinases and nuclear factor kappa-B. These findings suggest that the efficacy of K8NPs in controlling respiratory inflammation and infection can be used to develop functional materials that can prevent or alleviate respiratory diseases.
Collapse
Affiliation(s)
- Jonghyo Hong
- Graduate School of Biotechnology, Kyung Hee University, Yongin 17104, Republic of Korea; (J.H.); (M.S.); (J.S.)
| | - Minseong Son
- Graduate School of Biotechnology, Kyung Hee University, Yongin 17104, Republic of Korea; (J.H.); (M.S.); (J.S.)
| | - Jaeeun Sin
- Graduate School of Biotechnology, Kyung Hee University, Yongin 17104, Republic of Korea; (J.H.); (M.S.); (J.S.)
| | - Hangeun Kim
- Research and Development Center, Skin Biotechnology Center Co., Ltd., Yongin 17104, Republic of Korea
| | - Dae-Kyun Chung
- Graduate School of Biotechnology, Kyung Hee University, Yongin 17104, Republic of Korea; (J.H.); (M.S.); (J.S.)
| |
Collapse
|
18
|
Li Z, Zhang S, Zuber F, Altenried S, Jaklenec A, Langer R, Ren Q. Topical application of Lactobacilli successfully eradicates Pseudomonas aeruginosa biofilms and promotes wound healing in chronic wounds. Microbes Infect 2023; 25:105176. [PMID: 37406851 DOI: 10.1016/j.micinf.2023.105176] [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: 03/07/2023] [Revised: 06/18/2023] [Accepted: 06/22/2023] [Indexed: 07/07/2023]
Abstract
Chronic wounds are difficult to treat due to the presence of biofilm which prevents wound healing. Pseudomonas aeruginosa is one of the most common pathogens found in chronic wounds and conventional treatment strategies have been ineffective in the eradication of its biofilm, without harming the surrounding healthy tissue at the same time. Here, we introduced an innovative approach applying the probiotic product Bio-K+ (containing three lactobacilli) topically as an antimicrobial and antibiofilm agent. We identified lactic acid as the main active component. While antibiotics and antiseptics such as silver-ions only demonstrated limited efficacy, Bio-K+ was able to completely eradicate mature P. aeruginosa biofilms established in an in-vitro and ex-vivo human skin model. Furthermore, it demonstrated biocompatibility in the co-culture with human dermal fibroblasts and accelerated the migration of fibroblasts in a cell migration assay promoting wound healing. To enhance clinical practicability, we introduced Bio-K+ into the hydrocolloid dressing Aquacel, achieving sustained release of lactic acid and biofilm eradication. This new treatment approach applying probiotics could represent a major improvement in the management of chronic wounds and can be extended in treating other biofilm-associated infections.
Collapse
Affiliation(s)
- Zhihao Li
- Swiss Federal Laboratories for Materials Science and Technology, Lerchenfeldstrasse 5, 9014 St. Gallen, Switzerland.
| | - Sixuan Zhang
- Swiss Federal Laboratories for Materials Science and Technology, Lerchenfeldstrasse 5, 9014 St. Gallen, Switzerland
| | - Flavia Zuber
- Swiss Federal Laboratories for Materials Science and Technology, Lerchenfeldstrasse 5, 9014 St. Gallen, Switzerland
| | - Stefanie Altenried
- Swiss Federal Laboratories for Materials Science and Technology, Lerchenfeldstrasse 5, 9014 St. Gallen, Switzerland
| | - Ana Jaklenec
- David H. Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, 500 Main Street, Cambridge, MA 02139, USA
| | - Robert Langer
- David H. Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, 500 Main Street, Cambridge, MA 02139, USA
| | - Qun Ren
- Swiss Federal Laboratories for Materials Science and Technology, Lerchenfeldstrasse 5, 9014 St. Gallen, Switzerland.
| |
Collapse
|
19
|
Ersanli C, Tzora A, Voidarou C(C, Skoufos S, Zeugolis DI, Skoufos I. Biodiversity of Skin Microbiota as an Important Biomarker for Wound Healing. BIOLOGY 2023; 12:1187. [PMID: 37759587 PMCID: PMC10525143 DOI: 10.3390/biology12091187] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/28/2023] [Revised: 08/24/2023] [Accepted: 08/29/2023] [Indexed: 09/29/2023]
Abstract
Cutaneous wound healing is a natural and complex repair process that is implicated within four stages. However, microorganisms (e.g., bacteria) can easily penetrate through the skin tissue from the wound bed, which may lead to disbalance in the skin microbiota. Although commensal and pathogenic bacteria are in equilibrium in normal skin, their imbalance in the wound area can cause the delay or impairment of cutaneous wounds. Moreover, skin microbiota is in constant crosstalk with the immune system and epithelial cells, which has significance for the healing of a wound. Therefore, understanding the major bacteria species in the cutaneous wound as well as their communication with the immune system has gained prominence in a way that allows for the emergence of a new perspective for wound healing. In this review, the major bacteria isolated from skin wounds, the role of the crosstalk between the cutaneous microbiome and immune system to heal wounds, the identification techniques of these bacteria populations, and the applied therapies to manipulate the skin microbiota are investigated.
Collapse
Affiliation(s)
- Caglar Ersanli
- Laboratory of Animal Science, Nutrition and Biotechnology, Department of Agriculture, University of Ioannina, 47100 Arta, Greece; (C.E.); (I.S.)
- Laboratory of Animal Health, Food Hygiene and Quality, Department of Agriculture, University of Ioannina, 47100 Arta, Greece; (C.V.)
- Regenerative, Modular & Developmental Engineering Laboratory (REMODEL), Charles Institute of Dermatology, Conway Institute of Biomolecular and Biomedical Research, School of Mechanical and Materials Engineering, University College Dublin, D04 V1W8 Dublin, Ireland;
| | - Athina Tzora
- Laboratory of Animal Health, Food Hygiene and Quality, Department of Agriculture, University of Ioannina, 47100 Arta, Greece; (C.V.)
| | - Chrysoula (Chrysa) Voidarou
- Laboratory of Animal Health, Food Hygiene and Quality, Department of Agriculture, University of Ioannina, 47100 Arta, Greece; (C.V.)
| | - Stylianos Skoufos
- Laboratory of Animal Health, Food Hygiene and Quality, Department of Agriculture, University of Ioannina, 47100 Arta, Greece; (C.V.)
| | - Dimitrios I. Zeugolis
- Regenerative, Modular & Developmental Engineering Laboratory (REMODEL), Charles Institute of Dermatology, Conway Institute of Biomolecular and Biomedical Research, School of Mechanical and Materials Engineering, University College Dublin, D04 V1W8 Dublin, Ireland;
| | - Ioannis Skoufos
- Laboratory of Animal Science, Nutrition and Biotechnology, Department of Agriculture, University of Ioannina, 47100 Arta, Greece; (C.E.); (I.S.)
| |
Collapse
|
20
|
Nicholas-Haizelden K, Murphy B, Hoptroff M, Horsburgh MJ. Bioprospecting the Skin Microbiome: Advances in Therapeutics and Personal Care Products. Microorganisms 2023; 11:1899. [PMID: 37630459 PMCID: PMC10456854 DOI: 10.3390/microorganisms11081899] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2023] [Revised: 07/20/2023] [Accepted: 07/25/2023] [Indexed: 08/27/2023] Open
Abstract
Bioprospecting is the discovery and exploration of biological diversity found within organisms, genetic elements or produced compounds with prospective commercial or therapeutic applications. The human skin is an ecological niche which harbours a rich and compositional diversity microbiome stemming from the multifactorial interactions between the host and microbiota facilitated by exploitable effector compounds. Advances in the understanding of microbial colonisation mechanisms alongside species and strain interactions have revealed a novel chemical and biological understanding which displays applicative potential. Studies elucidating the organismal interfaces and concomitant understanding of the central processes of skin biology have begun to unravel a potential wealth of molecules which can exploited for their proposed functions. A variety of skin-microbiome-derived compounds display prospective therapeutic applications, ranging from antioncogenic agents relevant in skin cancer therapy to treatment strategies for antimicrobial-resistant bacterial and fungal infections. Considerable opportunities have emerged for the translation to personal care products, such as topical agents to mitigate various skin conditions such as acne and eczema. Adjacent compound developments have focused on cosmetic applications such as reducing skin ageing and its associated changes to skin properties and the microbiome. The skin microbiome contains a wealth of prospective compounds with therapeutic and commercial applications; however, considerable work is required for the translation of in vitro findings to relevant in vivo models to ensure translatability.
Collapse
Affiliation(s)
- Keir Nicholas-Haizelden
- Institute of Infection, Veterinary and Ecological Sciences, University of Liverpool, Liverpool L69 3BX, UK;
| | - Barry Murphy
- Unilever Research & Development, Port Sunlight, Wirral CH63 3JW, UK; (B.M.); (M.H.)
| | - Michael Hoptroff
- Unilever Research & Development, Port Sunlight, Wirral CH63 3JW, UK; (B.M.); (M.H.)
| | - Malcolm J. Horsburgh
- Institute of Infection, Veterinary and Ecological Sciences, University of Liverpool, Liverpool L69 3BX, UK;
| |
Collapse
|
21
|
Xu J, Chen C, Gan S, Liao Y, Fu R, Hou C, Yang S, Zheng Z, Chen W. The Potential Value of Probiotics after Dental Implant Placement. Microorganisms 2023; 11:1845. [PMID: 37513016 PMCID: PMC10383117 DOI: 10.3390/microorganisms11071845] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2023] [Revised: 07/15/2023] [Accepted: 07/18/2023] [Indexed: 07/30/2023] Open
Abstract
Dental implantation is currently the optimal solution for tooth loss. However, the health and stability of dental implants have emerged as global public health concerns. Dental implant placement, healing of the surgical site, osseointegration, stability of bone tissues, and prevention of peri-implant diseases are challenges faced in achieving the long-term health and stability of implants. These have been ongoing concerns in the field of oral implantation. Probiotics, as beneficial microorganisms, play a significant role in the body by inhibiting pathogens, promoting bone tissue homeostasis, and facilitating tissue regeneration, modulating immune-inflammatory levels. This review explores the potential of probiotics in addressing post-implantation challenges. We summarize the existing research regarding the importance of probiotics in managing dental implant health and advocate for further research into their potential applications.
Collapse
Affiliation(s)
- Jia Xu
- State Key Laboratory of Oral Diseases and National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu 610041, China
- Department of Oral Prosthodontics, West China Hospital of Stomatology, Sichuan University, Chengdu 610041, China
| | - Chenfeng Chen
- State Key Laboratory of Oral Diseases and National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu 610041, China
- Department of General Dentistry, West China Hospital of Stomatology, Sichuan University, Chengdu 610041, China
| | - Shuaiqi Gan
- State Key Laboratory of Oral Diseases and National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu 610041, China
- Department of Oral Prosthodontics, West China Hospital of Stomatology, Sichuan University, Chengdu 610041, China
| | - Yihan Liao
- State Key Laboratory of Oral Diseases and National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu 610041, China
- Department of Oral Prosthodontics, West China Hospital of Stomatology, Sichuan University, Chengdu 610041, China
| | - Ruijie Fu
- State Key Laboratory of Oral Diseases and National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu 610041, China
- Department of Oral Prosthodontics, West China Hospital of Stomatology, Sichuan University, Chengdu 610041, China
| | - Chuping Hou
- State Key Laboratory of Oral Diseases and National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu 610041, China
- Department of Oral Prosthodontics, West China Hospital of Stomatology, Sichuan University, Chengdu 610041, China
| | - Shuhan Yang
- State Key Laboratory of Oral Diseases and National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu 610041, China
- Department of Oral Prosthodontics, West China Hospital of Stomatology, Sichuan University, Chengdu 610041, China
| | - Zheng Zheng
- State Key Laboratory of Oral Diseases and National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu 610041, China
- Department of Oral Prosthodontics, West China Hospital of Stomatology, Sichuan University, Chengdu 610041, China
| | - Wenchuan Chen
- State Key Laboratory of Oral Diseases and National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu 610041, China
- Department of Oral Prosthodontics, West China Hospital of Stomatology, Sichuan University, Chengdu 610041, China
- Jinjiang Out-Patient Section, West China Hospital of Stomatology, Sichuan University, Chengdu 610041, China
| |
Collapse
|
22
|
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 PMCID: PMC10346906 DOI: 10.3390/nu15133055] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [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 (M.M.); (G.T.); (A.D.S.)
| | - Moysis Moysidis
- Department of Surgery, Aristotle University of Thessaloniki, 54636 Thessaloniki, Greece (M.M.); (G.T.); (A.D.S.)
| | - Georgios Tzikos
- Department of Surgery, Aristotle University of Thessaloniki, 54636 Thessaloniki, Greece (M.M.); (G.T.); (A.D.S.)
| | - 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 (M.M.); (G.T.); (A.D.S.)
| | - 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 (M.M.); (G.T.); (A.D.S.)
| |
Collapse
|
23
|
Chin JD, Zhao L, Mayberry TG, Cowan BC, Wakefield MR, Fang Y. Photodynamic Therapy, Probiotics, Acetic Acid, and Essential Oil in the Treatment of Chronic Wounds Infected with Pseudomonas aeruginosa. Pharmaceutics 2023; 15:1721. [PMID: 37376169 PMCID: PMC10301549 DOI: 10.3390/pharmaceutics15061721] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2023] [Revised: 06/05/2023] [Accepted: 06/07/2023] [Indexed: 06/29/2023] Open
Abstract
As a prevalent medical problem that burdens millions of patients across the world, chronic wounds pose a challenge to the healthcare system. These wounds, often existing as a comorbidity, are vulnerable to infections. Consequently, infections hinder the healing process and complicate clinical management and treatment. While antibiotic drugs remain a popular treatment for infected chronic wounds, the recent rise of antibiotic-resistant strains has hastened the need for alternative treatments. Future impacts of chronic wounds are likely to increase with aging populations and growing obesity rates. With the need for more effective novel treatments, promising research into various wound therapies has seen an increased demand. This review summarizes photodynamic therapy, probiotics, acetic acid, and essential oil studies as developing antibiotic-free treatments for chronic wounds infected with Pseudomonas aeruginosa. Clinicians may find this review informative by gaining a better understanding of the state of current research into various antibiotic-free treatments. Furthermore. this review provides clinical significance, as clinicians may seek to implement photodynamic therapy, probiotics, acetic acid, or essential oils into their own practice.
Collapse
Affiliation(s)
- Jaeson D. Chin
- Department of Microbiology, Immunology & Pathology, Des Moines University, Des Moines, IA 50312, USA
| | - Lei Zhao
- The Department of Respiratory Medicine, The Second People’s Hospital of Hefei and Hefei Hospital Affiliated to Anhui Medical University, Hefei 230002, China
| | - Trenton G. Mayberry
- Department of Surgery, University of Missouri School of Medicine, Columbia, MO 65212, USA
| | - Braydon C. Cowan
- Department of Surgery, University of Missouri School of Medicine, Columbia, MO 65212, USA
| | - Mark R. Wakefield
- Department of Surgery, University of Missouri School of Medicine, Columbia, MO 65212, USA
- Ellis Fischel Cancer Center, University of Missouri, Columbia, MO 65212, USA
| | - Yujiang Fang
- Department of Microbiology, Immunology & Pathology, Des Moines University, Des Moines, IA 50312, USA
| |
Collapse
|
24
|
Magnifico I, Perna A, Cutuli MA, Medoro A, Pietrangelo L, Guarnieri A, Foderà E, Passarella D, Venditti N, Vergalito F, Petronio Petronio G, Di Marco R. A Wall Fragment of Cutibacterium acnes Preserves Junctional Integrity Altered by Staphylococcus aureus in an Ex Vivo Porcine Skin Model. Pharmaceutics 2023; 15:pharmaceutics15041224. [PMID: 37111709 PMCID: PMC10145065 DOI: 10.3390/pharmaceutics15041224] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2023] [Revised: 04/07/2023] [Accepted: 04/07/2023] [Indexed: 04/29/2023] Open
Abstract
(1) Background alteration of the skin microbiota, dysbiosis, causes skin barrier impairment resulting in disease development. Staphylococcus aureus, the main pathogen associated with dysbiosis, secretes several virulence factors, including α-toxin that damages tight junctions and compromises the integrity of the skin barrier. The use of members of the resident microbiota to restore the skin barrier, bacteriotherapy, represents a safe treatment for skin conditions among innovative options. The aim of this study is the evaluation of a wall fragment derived from a patented strain of Cutibacterium acnes DSM28251 (c40) alone and conjugated to a mucopolysaccharide carrier (HAc40) in counteracting S. aureus pathogenic action on two tight junction proteins (Claudin-1 and ZO-1) in an ex vivo porcine skin infection model. Methods: skin biopsies were infected with live S. aureus strains ATCC29213 and DSM20491. Tissue was pre-incubated or co-incubated with c40 and HAc40. (3) Results: c40 and HAc40 prevent and counteract Claudin-1 and Zo-1 damage (4) Conclusions: c40 and the functional ingredient HAc40 represent a potential non-pharmacological treatment of skin diseases associated with cutaneous dysbiosis of S. aureus. These findings offer numerous avenues for new research.
Collapse
Affiliation(s)
- Irene Magnifico
- Department of Medicine and Health Science "V. Tiberio", Università degli Studi del Molise, 8600 Campobasso, Italy
| | - Angelica Perna
- Department of Medicine and Health Science "V. Tiberio", Università degli Studi del Molise, 8600 Campobasso, Italy
| | - Marco Alfio Cutuli
- Department of Medicine and Health Science "V. Tiberio", Università degli Studi del Molise, 8600 Campobasso, Italy
| | - Alessando Medoro
- Department of Medicine and Health Science "V. Tiberio", Università degli Studi del Molise, 8600 Campobasso, Italy
| | - Laura Pietrangelo
- Department of Medicine and Health Science "V. Tiberio", Università degli Studi del Molise, 8600 Campobasso, Italy
| | - Antonio Guarnieri
- Department of Medicine and Health Science "V. Tiberio", Università degli Studi del Molise, 8600 Campobasso, Italy
| | - Emanuele Foderà
- Department of Medicine and Health Science "V. Tiberio", Università degli Studi del Molise, 8600 Campobasso, Italy
| | - Daniela Passarella
- Department of Medicine and Health Science "V. Tiberio", Università degli Studi del Molise, 8600 Campobasso, Italy
| | - Noemi Venditti
- Department of Medicine and Health Science "V. Tiberio", Università degli Studi del Molise, 8600 Campobasso, Italy
| | - Franca Vergalito
- Department of Agricultural, Environmental and Food Sciences (DiAAA), Università degli Studi del Molise, 8600 Campobasso, Italy
| | - Giulio Petronio Petronio
- Department of Medicine and Health Science "V. Tiberio", Università degli Studi del Molise, 8600 Campobasso, Italy
| | - Roberto Di Marco
- Department of Medicine and Health Science "V. Tiberio", Università degli Studi del Molise, 8600 Campobasso, Italy
| |
Collapse
|
25
|
Michels M, Córneo E, Rocha LBG, Dias R, Voytena APL, Rossetto M, Ramlov F, Dal-Pizzol F, Jesus GFA. Paraprobiotics strains accelerate wound repair by stimulating re-epithelialization of NIH-3T3 cells, decreasing inflammatory response and oxidative stress. Arch Microbiol 2023; 205:134. [PMID: 36959516 DOI: 10.1007/s00203-023-03469-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2022] [Accepted: 03/06/2023] [Indexed: 03/25/2023]
Abstract
The present study aimed to evaluate the potential and specificity of the inflammatory and antioxidant response of Microbe-Associated Molecular Patterns (MAMPs) in NIH-3T3 fibroblast cells, as well as in the healing process of skin wounds. Cells (NIH-3T3) were cultivated in supplemented specific medium. NIH-3T3 cells were treated with MAMPs (Bifidobacterium lactis or Lactobacillus casei or Lactobacillus gasseri or Lactobacillus paracasei or Streptococcus thermophilus), at two concentrations and insulted with LPS or H2O2. Cell viability, myeloperoxidase activity, nitrite/nitrate, oxidative damage and inflammatory parameters were measured. In addition, scratch assay was performed. Significant scratch closure was observed after 24 h and 48 h, and the effect of 0.1 g/mL MAMPs on wound healing was found to be highly statistically significant. In the viability cellular assay, Lactobacillus showed better response in 0.1 g/mL dose, whereas B. lactis and S. thermophilus showed better response in 0.01 g/mL dose. There was reduction in IL-6 and IL-1β levels in all treatments insulted with LPS. MAMP's showed preventive efficacy in reducing the effects caused by LPS. The MAMP's action in decreasing the production of ROS, inflammatory activity and increasing cell viability, besides significant cell proliferation during wound healing processes suggests remodeling mechanisms and new possibilities for wound healing.
Collapse
Affiliation(s)
- Monique Michels
- Gabbia Biotechnology, Barra Velha, Santa Catarina, Brazil.
- Biohall Research and Innovation, Itajaí, Santa Catarina, Brazil.
| | - Emily Córneo
- Laboratory of Experimental Pathophysiology-Graduate Program in Health Sciences, University of Southern Santa Catarina (UNESC), Criciúma, Brazil
| | - Luana Bezerra Gonçalves Rocha
- Laboratory of Experimental Pathophysiology-Graduate Program in Health Sciences, University of Southern Santa Catarina (UNESC), Criciúma, Brazil
| | - Rodrigo Dias
- Laboratory of Experimental Pathophysiology-Graduate Program in Health Sciences, University of Southern Santa Catarina (UNESC), Criciúma, Brazil
| | | | | | | | - Felipe Dal-Pizzol
- Laboratory of Experimental Pathophysiology-Graduate Program in Health Sciences, University of Southern Santa Catarina (UNESC), Criciúma, Brazil
| | - Gabriel Fernandes Alves Jesus
- Gabbia Biotechnology, Barra Velha, Santa Catarina, Brazil
- Biohall Research and Innovation, Itajaí, Santa Catarina, Brazil
| |
Collapse
|
26
|
Al-Romaih S, Harati O, Mfuna LE, Filali-Mouhim A, Pelletier A, Renteria Flores A, Desrosiers M. Response to intranasal Lactococcus lactis W136 probiotic supplementation in refractory CRS is associated with modulation of non-type 2 inflammation and epithelial regeneration. FRONTIERS IN ALLERGY 2023; 4:1046684. [PMID: 37007649 PMCID: PMC10050565 DOI: 10.3389/falgy.2023.1046684] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2022] [Accepted: 02/20/2023] [Indexed: 03/17/2023] Open
Abstract
JustificationWe have previously documented that in individuals with chronic rhinosinusitis (CRS) refractory to surgery, intranasal application of live Lactococcus lactis W136, a probiotic bacterium, improves sinus-specific symptoms, SNOT-22, and mucosal aspect on endoscopy, accompanied by a reduction in sinus pathogens and an increase in protective bacteria. The present work explores the molecular mechanisms underpinning these observations using transcriptomics of the sinus mucosa.MethodEpithelial brushings collected prospectively as a sub-study of the L. lactis W136 clinical trial were used to probe epithelial responses to microbiome supplementation using a hypothesis-free bioinformatic analysis of gene expression analysis. Samples from twenty-four patients with CRS refractory to medical and surgical management were prospectively collected during a clinical trial assessing the effect of 14 days of BID nasal irrigation with 1.2 billion CFU of live L. lactis W136 probiotic bacteria (CRSwNP = 17, CRSsNP = 7). Endoscopically guided sinus brushings were collected as part of the initial study, with brushings performed immediately before and after treatment. Following RNA extraction, samples were assessed using the Illumina HumanHT-12 V4 BeadChip. Differential gene expression was calculated, and pathway enrichment analysis was performed to identify potentially implicated processes.ResultsDifferentially identified transcripts and pathways were assessed for the overall population and the clinical phenotypes of CRSwNP and CRSsNP. Patterns of response to treatment were similar across all groups, implicating pathways for the regulation of immunity and epithelial cell regulation. These resemble the patterns of improvement observed following successful treatment with endoscopic sinus surgery or azithromycin.ConclusionGene expression profiling following the application of live bacteria to the diseased sinus epithelium highlights the implication of multiple components of the inflammation-microbiome-epithelial barrier axis implicated in CRS. These effects appear to involve both epithelial restoration and modulation of innate and adaptive immunity, supporting the potential interest of targeting the sinus epithelium and the microbiome as potential CRS therapies.
Collapse
Affiliation(s)
- Saud Al-Romaih
- Department of Otolaryngology, Head and Neck Surgery, Faculty of Medicine, King Saud University, Riyadh, Saudi Arabia
| | - Oumkaltoum Harati
- Centre de Recherche du Centre Hospitalier de l’Université de Montréal (CRCHUM), Montreal, QC, Canada
| | - Leandra Endam Mfuna
- Centre de Recherche du Centre Hospitalier de l’Université de Montréal (CRCHUM), Montreal, QC, Canada
| | - Ali Filali-Mouhim
- Centre de Recherche du Centre Hospitalier de l’Université de Montréal (CRCHUM), Montreal, QC, Canada
| | - Audrey Pelletier
- Centre de Recherche du Centre Hospitalier de l’Université de Montréal (CRCHUM), Montreal, QC, Canada
| | - Axel Renteria Flores
- Centre de Recherche du Centre Hospitalier de l’Université de Montréal (CRCHUM), Montreal, QC, Canada
- Division of Otolaryngology-Head & Neck Surgery, Centre Hospitalier de l'Université de Montréal, Montréal, QC, Canada
| | - Martin Desrosiers
- Centre de Recherche du Centre Hospitalier de l’Université de Montréal (CRCHUM), Montreal, QC, Canada
- Division of Otolaryngology-Head & Neck Surgery, Centre Hospitalier de l'Université de Montréal, Montréal, QC, Canada
- Correspondence: Martin Desrosiers
| |
Collapse
|
27
|
Investigating the effect of the probiotic Lactobacillus plantarum and the prebiotic fructooligosaccharides on Pseudomonas aeruginosa metabolome, virulence factors and biofilm formation as potential quorum sensing inhibitors. Microb Pathog 2023; 177:106057. [PMID: 36878335 DOI: 10.1016/j.micpath.2023.106057] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2022] [Revised: 02/27/2023] [Accepted: 03/03/2023] [Indexed: 03/07/2023]
Abstract
Pseudomonas aeruginosa (P. aeruginosa) uses quorum sensing signaling (QS) molecules to control the expression of virulence factors and biofilm formation. In this study, the effects of the probiotic's (Lactobacillus plantarum (L. plantarum)) lysate and cell-free supernatant and the prebiotic (Fructooligosaccharides (FOS)) on the levels of P. aeruginosa QS molecules, virulence factors, biofilm density and metabolites were observed. These effects were investigated using exofactor assays, crystal violet and liquid chromatography-mass spectrometry (LC-MS)-based metabolomics approach. Results showed that in comparison to untreated P. aeruginosa, the L. plantarum cell-free supernatant (5%) and FOS (2%) significantly reduced the levels of the virulence factor pyoverdine (PVD) and several metabolites in the QS pathway including Pseudomonas autoinducer-2 (PAI-2). Metabolomics study revealed that the level of different secondary metabolites involved in the biosynthesis of vitamins, amino acids and the tricarboxylic acid (TCA) cycle were also affected. L. Plantarum was found to have a higher impact on the metabolomics profile of P. aeruginosa and its QS molecules compared to FOS. Lastly, a decrease in the formation of the P. aeruginosa biofilm was observed in a time-dependent pattern upon treatment with either cell-free supernatant of L. plantarum (5%), FOS (2%) or a combination of both treatments (5% + 2%). The latter showed the highest effect with 83% reduction in biofilm density at 72 h incubation. This work highlighted the important role probiotics and prebiotics play as potential QS inhibitors for P. aeruginosa. Moreover, it demonstrated the significant role of LC-MS metabolomics for investigating the altered biochemical and QS pathways in P. aeruginosa.
Collapse
|
28
|
Bubnov R, Spivak M. Pathophysiology-Based Individualized Use of Probiotics and Prebiotics for Metabolic Syndrome: Implementing Predictive, Preventive, and Personalized Medical Approach. ADVANCES IN PREDICTIVE, PREVENTIVE AND PERSONALISED MEDICINE 2023:133-196. [DOI: 10.1007/978-3-031-19564-8_6] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/03/2025]
|
29
|
Mohammedsaeed W, Bukhari S, Manzoor N, Almaramhy HH. An ex-vivo study to investigate the potential of Staphylococcus epidermidis lysate to improve wound healing in diabetic patients. JOURNAL OF TAIBAH UNIVERSITY FOR SCIENCE 2022. [DOI: 10.1080/16583655.2022.2112489] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/14/2022]
Affiliation(s)
- Walaa Mohammedsaeed
- Department of Medical Laboratory Technology, Faculty of Applied Medical Science, Taibah University, Medina, Saudi Arabia
| | - Samyah Bukhari
- Endocrinologist in King Fahd Hospital, Prince Abdelaziz Ben Maged Ben Abdelaziz Diabetic Centre, Al Madinah, Saudi Arabia
| | - Nikhat Manzoor
- Department of Biosciences, Jamia Millia Islamia, New Delhi-, Delhi, India
| | | |
Collapse
|
30
|
Srivastava P, Sondak T, Sivashanmugam K, Kim KS. A Review of Immunomodulatory Reprogramming by Probiotics in Combating Chronic and Acute Diabetic Foot Ulcers (DFUs). Pharmaceutics 2022; 14:2436. [PMID: 36365254 PMCID: PMC9699442 DOI: 10.3390/pharmaceutics14112436] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2022] [Revised: 11/02/2022] [Accepted: 11/08/2022] [Indexed: 08/29/2023] Open
Abstract
Diabetic foot ulcers (DFUs) are characterized by a lack of angiogenesis and distal limb diabetic neuropathy. This makes it possible for opportunistic pathogens to protect the biofilm-encased micro-communities, causing a delay in wound healing. The acute and chronic phases of DFU-associated infections are distinguished by the differential expression of innate proinflammatory cytokines and tumor necrosis factors (TNF-α and -β). Efforts are being made to reduce the microbial bioburden of wounds by using therapies such as debridement, hyperbaric oxygen therapy, shock wave therapy, and empirical antibiotic treatment. However, the constant evolution of pathogens limits the effectiveness of these therapies. In the wound-healing process, continuous homeostasis and remodeling processes by commensal microbes undoubtedly provide a protective barrier against diverse pathogens. Among commensal microbes, probiotics are beneficial microbes that should be administered orally or topically to regulate gut-skin interaction and to activate inflammation and proinflammatory cytokine production. The goal of this review is to bridge the gap between the role of probiotics in managing the innate immune response and the function of proinflammatory mediators in diabetic wound healing. We also highlight probiotic encapsulation or nanoformulations with prebiotics and extracellular vesicles (EVs) as innovative ways to tackle target DFUs.
Collapse
Affiliation(s)
- Prakhar Srivastava
- Department of Chemistry and Chemistry Institute of Functional Materials, Pusan National University, Busan 46241, Korea
| | - Tesalonika Sondak
- Department of Chemistry and Chemistry Institute of Functional Materials, Pusan National University, Busan 46241, Korea
| | - Karthikeyan Sivashanmugam
- School of Biosciences and Technology, High Throughput Screening Lab, Vellore Institute of Technology, Vellore 632014, Tamil Nadu, India
| | - Kwang-sun Kim
- Department of Chemistry and Chemistry Institute of Functional Materials, Pusan National University, Busan 46241, Korea
| |
Collapse
|
31
|
Li C, Li Y, Li S, Chen S, Liu G, Deng X, Chang W, Cai H. Bacillus subtilis Protects the Ducks from Oxidative Stress Induced by Escherichia coli: Efficacy and Molecular Mechanism. Antioxidants (Basel) 2022; 11:1951. [PMID: 36290674 PMCID: PMC9598806 DOI: 10.3390/antiox11101951] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2022] [Revised: 09/25/2022] [Accepted: 09/26/2022] [Indexed: 01/03/2025] Open
Abstract
Bacillus subtilis has been widely used in animal husbandry as a potential alternative to antibiotics due to its excellent bacteriostasis and antioxidant activity. This study aims to investigate the effects of Bacillus subtilis on the protection of ducks from Escherichia coli infection and its mechanism. The four experimental groups include the negative control group, positive control group, antibiotic group and Bacillus subtilis group. Ducks in positive, antibiotic and Bacillus subtilis groups are orally administered with Escherichia coli and equivalent saline solution for the negative group. The results show that supplements with Bacillus subtilis enhances the performance and health status of the infected ducks. Moreover, Bacillus subtilis alleviates the increase in globulin, LPS and MDA, and the decrease in albumin, T-AOC and T-SOD in the serum caused by Escherichia coli infection. Bacillus subtilis also attenuates injury in the intestine and partially reverses the increase in ROS production and the depletion of ATP in the jejunum. These effects are accompanied with the change of related genes of the ribosome (13.54%) and oxidative phosphorylation (6.68%). Collectively, Bacillus subtilis alleviates the damage caused by Escherichia coli infection in ducks by activating ribosome and oxidative phosphorylation signaling to regulate antioxidant and energy metabolism.
Collapse
Affiliation(s)
- Chong Li
- Key Laboratory for Feed Biotechnology of the Ministry of Agriculture and Rural Affairs, Institute of Feed Research, Chinese Academy of Agriculture Sciences, Beijing 100081, China
- Precision Livestock and Nutrition Laboratory, Teaching and Research Centre (TERRA), Gembloux Agro-Bio Tech, University of Liège, 5030 Gembloux, Belgium
| | - Yang Li
- Key Laboratory for Feed Biotechnology of the Ministry of Agriculture and Rural Affairs, Institute of Feed Research, Chinese Academy of Agriculture Sciences, Beijing 100081, China
| | - Shuzhen Li
- Key Laboratory for Feed Biotechnology of the Ministry of Agriculture and Rural Affairs, Institute of Feed Research, Chinese Academy of Agriculture Sciences, Beijing 100081, China
| | - Si Chen
- Department of Molecular Cell Biology, Samsung Medical Center, Sungkyunkwan University School of Medicine, Suwon 16419, Korea
| | - Guohua Liu
- Key Laboratory for Feed Biotechnology of the Ministry of Agriculture and Rural Affairs, Institute of Feed Research, Chinese Academy of Agriculture Sciences, Beijing 100081, China
| | - Xuejuan Deng
- National Engineering Research Center of Biological Feed, Beijing 100081, China
| | - Wenhuan Chang
- Key Laboratory for Feed Biotechnology of the Ministry of Agriculture and Rural Affairs, Institute of Feed Research, Chinese Academy of Agriculture Sciences, Beijing 100081, China
| | - Huiyi Cai
- Key Laboratory for Feed Biotechnology of the Ministry of Agriculture and Rural Affairs, Institute of Feed Research, Chinese Academy of Agriculture Sciences, Beijing 100081, China
- National Engineering Research Center of Biological Feed, Beijing 100081, China
| |
Collapse
|
32
|
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: 54] [Impact Index Per Article: 18.0] [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
|
33
|
Moysidis M, Stavrou G, Cheva A, Abba Deka I, Tsetis JK, Birba V, Kapoukranidou D, Ioannidis A, Tsaousi G, Kotzampassi K. The 3-D configuration of excisional skin wound healing after topical probiotic application. Injury 2022; 53:1385-1393. [PMID: 35148901 DOI: 10.1016/j.injury.2022.02.006] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/27/2021] [Revised: 02/01/2022] [Accepted: 02/03/2022] [Indexed: 02/02/2023]
Abstract
Nowadays, there is an increasing knowledge that probiotic bacteria, topically applied, affects skin pathology. The objective of this study is to evaluate the effect on wound healing of locally applied probiotics by calculating the 3-D configuration of a standardized excisional wound. Fifty-two male Wistar rats were randomly allocated into groups: control, PRO1 [L. plantarum] and PRO2 [L. rhamnosus, B. longum]. Six excisional full-thickness wounds were created on each dorsum by an 8-mm circular biopsy punch; probiotics or saline were applied on days 0, 2, 4, 8, 16, photos of the wounds taken and specimens excised for histology [4 rats/group/time-point]. Both probiotic-groups exhibited accelerated healing significantly faster than the control, throughout, PRO2 exhibiting finally the best results [day 16]. However, only on day 2, did PRO1 exhibit the best results [wounded area, borders distance and epitheliazation line]. The results clearly demonstrate that the topical application of probiotics significantly improves the healing process, each strain working differently and more effectively in different healing phases. Thus, a combined formula containing different probiotics to modulate various healing phases is desirable. To this end our research continous.
Collapse
Affiliation(s)
- Moysis Moysidis
- Department of Surgery, Aristotle University of Thessaloniki, AHEPA University Hospital, Thessaloniki, Greece
| | - George Stavrou
- Department of Surgery, Aristotle University of Thessaloniki, AHEPA University Hospital, Thessaloniki, Greece
| | - Aggeliki Cheva
- Department of Pathology, Faculty of Medicine, Aristotle University of Thessaloniki, Thessaloniki, Greece
| | - Ioanna Abba Deka
- Department of Pathology, Faculty of Medicine, Aristotle University of Thessaloniki, Thessaloniki, Greece
| | | | - Vasiliki Birba
- Department of Surgery, Aristotle University of Thessaloniki, AHEPA University Hospital, Thessaloniki, Greece
| | - Dorothea Kapoukranidou
- Department of Physiology, Faculty of Medicine, Aristotle University of Thessaloniki, Thessaloniki, Greece
| | - Aris Ioannidis
- Department of Surgery, Aristotle University of Thessaloniki, AHEPA University Hospital, Thessaloniki, Greece
| | - Georgia Tsaousi
- Department of Surgery, Aristotle University of Thessaloniki, AHEPA University Hospital, Thessaloniki, Greece
| | - Katerina Kotzampassi
- Department of Surgery, Aristotle University of Thessaloniki, AHEPA University Hospital, Thessaloniki, Greece.
| |
Collapse
|
34
|
Moraffah F, Kiani M, Abdollahi M, Yoosefi S, Vatanara A, Samadi N. In Vitro-In Vivo Correlation for the Antibacterial Effect of Lactiplantibacillus plantarum as a Topical Healer for Infected Burn Wound. Probiotics Antimicrob Proteins 2022; 14:675-689. [PMID: 35349102 DOI: 10.1007/s12602-022-09934-4] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 03/14/2022] [Indexed: 01/20/2023]
Abstract
Difficulties in delivering antimicrobial agents to wound areas and emersion of multiple drug resistant organisms (MDROs) have converted managing burn infections into a complicated task in medicine. Probiotics emerged not only as a probable solution for burn infections but also as an accelerator in the healing process. The probability of in vitro-in vivo correlation (IVIVC) in probiotic activity leads to lower costs in finding new therapeutic options. Simulated wound fluid (SWF) was used to evaluate the antibacterial function of Lactiplantibacillus plantarum in wounds. The growth parameters in SWF were evaluated using a logistic model to predict growth behavior in the wound area. In addition, probiotic antimicrobial activity and secretion of antibacterial substances in SWF were also studied. Data were used to select the initial dose and apply frequency for in vivo study. The wound models were infected by two main pathogens (Pseudomonas aeruginosa or Staphylococcus aureus). In vitro results showed less lag time associated with considerable acid production in SWF. In the following, secretion of antimicrobial substances and co-aggregation with pathogens became more important. The susceptibility of pathogens to these factors was different, and culture medium affected the yield of each factor involved in eliminating pathogens. Histological analysis and macroscopic examination of wounds revealed probiotics as effective as positive control or more. There were some differences in the antibacterial functions of probiotics in simulated and real wound environments. The in vitro effect of probiotics on removal of pathogens was not the same as the trend seen in vivo.
Collapse
Affiliation(s)
- Fatemeh Moraffah
- Department of Pharmaceutics, Faculty of Pharmacy, Tehran University of Medical Sciences, Tehran, Iran
| | - Melika Kiani
- Department of Pharmaceutics, Faculty of Pharmacy, Tehran University of Medical Sciences, Tehran, Iran
| | - Mohammad Abdollahi
- Department of Toxicology and Pharmacology, Faculty of Pharmacy, Tehran University of Medical Sciences, Tehran, Iran
| | - Sepideh Yoosefi
- Department of Drug and Food Control, Faculty of Pharmacy, Tehran University of Medical Sciences, Tehran, Iran
| | - Alireza Vatanara
- Department of Pharmaceutics, Faculty of Pharmacy, Tehran University of Medical Sciences, Tehran, Iran.
| | - Nasrin Samadi
- Department of Drug and Food Control, Faculty of Pharmacy, Tehran University of Medical Sciences, Tehran, Iran.
- Pharmaceutical Quality Assurance Research Center, The Institute of Pharmaceutical Sciences (TIPS), Tehran University of Medical Sciences, Tehran, Iran.
| |
Collapse
|
35
|
Colonization of Lactobacillus rhamnosus GG in Cirrhinus molitorella (Mud Carp) Fingerling: Evidence for Improving Disease Resistance and Growth Performance. Appl Microbiol 2022. [DOI: 10.3390/applmicrobiol2010012] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
The use of probiotic bacteria can not only enhance the nutritional utilization of fish feeds to produce more biomass but can also provide a practically “safer” alternative to the fish farming industry to reduce the abuse of antibiotics and drugs. This study investigated the possibility of colonizing Lactobacillus rhamnosus strain GG (LGG) to the intestine of Cirrhinus molitorella (mud carp) fingerling. Colonization of LGG was observed in gut tissue after 14 days of administration with a diet supplemented with 1 × 108 CFU/mL LGG. Moreover, growth performance parameters of the LGG-supplemented diet group, including relative weight gain, feed conversion ratio and feed efficiency, were found about two-fold higher than the control group after 60 days. In addition, fish fed with an LGG-supplemented diet for 60 days showed substantial resistance against the infection of pathogenic bacterial Aeromonas hydrophila, with a relative survival rate of up to 57% compared to the control group. In summary, the results indicated that LGG as dietary supplement for mud carp fingerling can enhance nutrition utilization and better protect fish against the infection of Aeromonas hydrophila. The results provide an insight to the fish farming industry, encouraging a reduction in the use of antibiotics and drugs and the production of “safer” mud carp for the market at a manageable cost.
Collapse
|
36
|
Lebeer S, Oerlemans EFM, Claes I, Henkens T, Delanghe L, Wuyts S, Spacova I, van den Broek MFL, Tuyaerts I, Wittouck S, De Boeck I, Allonsius CN, Kiekens F, Lambert J. Selective targeting of skin pathobionts and inflammation with topically applied lactobacilli. Cell Rep Med 2022; 3:100521. [PMID: 35243421 PMCID: PMC8861818 DOI: 10.1016/j.xcrm.2022.100521] [Citation(s) in RCA: 30] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2021] [Revised: 10/29/2021] [Accepted: 01/16/2022] [Indexed: 01/04/2023]
Abstract
Tailored skin microbiome modulation approaches with probiotics are highly challenging. Here, we show that lactobacilli are underestimated members of the skin microbiota. We select specific strains of nomadic lactobacilli for their functional applicability on the skin and capacity to inhibit growth and inflammation by skin pathobionts. The strains are formulated as microcapsules for topical formulations and tested in patients with mild-to-moderate acne. The selected lactobacilli are able to reduce inflammatory lesions in a pilot and placebo-controlled study. Daily application for 8 weeks is associated with an in vivo temporary modulation of the microbiome, including a reduction in relative abundance of staphylococci and Cutibacterium acnes, and an increase in lactobacilli. The reduction in inflammatory lesions is still apparent 4 weeks after the topical application of the lactobacilli ended, indicating a possible additional immunomodulatory effect. This study shows that carefully selected and formulated lactobacilli are a viable therapeutic option for common acne lesions.
Collapse
Affiliation(s)
- Sarah Lebeer
- University of Antwerp, Department of Bioscience Engineering, Groenenborgerlaan 171, B-2020 Antwerp, Belgium
| | - Eline F M Oerlemans
- University of Antwerp, Department of Bioscience Engineering, Groenenborgerlaan 171, B-2020 Antwerp, Belgium
| | - Ingmar Claes
- University of Antwerp, Department of Bioscience Engineering, Groenenborgerlaan 171, B-2020 Antwerp, Belgium
| | - Tim Henkens
- University of Antwerp, Department of Pharmaceutical, Biomedical and Veterinary Sciences, Laboratory of Pharmaceutical Technology and Biopharmacy, Universiteitsplein 1, B-2610 Wilrijk, Belgium
| | - Lize Delanghe
- University of Antwerp, Department of Bioscience Engineering, Groenenborgerlaan 171, B-2020 Antwerp, Belgium
| | - Sander Wuyts
- University of Antwerp, Department of Bioscience Engineering, Groenenborgerlaan 171, B-2020 Antwerp, Belgium
| | - Irina Spacova
- University of Antwerp, Department of Bioscience Engineering, Groenenborgerlaan 171, B-2020 Antwerp, Belgium
| | - Marianne F L van den Broek
- University of Antwerp, Department of Bioscience Engineering, Groenenborgerlaan 171, B-2020 Antwerp, Belgium
| | - Ines Tuyaerts
- University of Antwerp, Department of Bioscience Engineering, Groenenborgerlaan 171, B-2020 Antwerp, Belgium
| | - Stijn Wittouck
- University of Antwerp, Department of Bioscience Engineering, Groenenborgerlaan 171, B-2020 Antwerp, Belgium
| | - Ilke De Boeck
- University of Antwerp, Department of Bioscience Engineering, Groenenborgerlaan 171, B-2020 Antwerp, Belgium
| | - Camille N Allonsius
- University of Antwerp, Department of Bioscience Engineering, Groenenborgerlaan 171, B-2020 Antwerp, Belgium
| | - Filip Kiekens
- University of Antwerp, Department of Pharmaceutical, Biomedical and Veterinary Sciences, Laboratory of Pharmaceutical Technology and Biopharmacy, Universiteitsplein 1, B-2610 Wilrijk, Belgium
| | - Julien Lambert
- University Hospital Antwerp/University of Antwerp, Department of Dermatology and Venereology, Wilrijkstraat 10, 2650 Edegem, Belgium
| |
Collapse
|
37
|
de Souza da Motta A, Nespolo CR, Breyer GM. Probiotics in milk and dairy foods. PROBIOTICS 2022:103-128. [DOI: 10.1016/b978-0-323-85170-1.00004-x] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/06/2025]
|
38
|
Does Probiotic Consumption Enhance Wound Healing? A Systematic Review. Nutrients 2021; 14:nu14010111. [PMID: 35010987 PMCID: PMC8746682 DOI: 10.3390/nu14010111] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2021] [Revised: 12/21/2021] [Accepted: 12/23/2021] [Indexed: 01/04/2023] Open
Abstract
The use of probiotics is one of the emerging lines of treatment for wound healing. This systematic review aimed to summarize currently available evidence on the effect of oral or enteral probiotic therapy on skin or oral mucosal wound healing in humans. To verify the developments in this field and the level of available scientific evidence, we applied a broad search strategy with no restrictions on wound type, target population, probiotic strain, or intervention protocol used. This review included seven studies involving 348 individuals. Four studies reported positive outcomes for healing improvement after probiotic therapy, and none of the studies reported adverse effects or a marked increase in wound healing time. The positive or neutral results observed do not generate strong evidence regarding the effectiveness of probiotics for wound healing. However, they suggest a promising field for future clinical research where the probiotic strains used, type of wounds, and target population are controlled for.
Collapse
|
39
|
Albuquerque-Souza E, Ishikawa KH, Amado PP, Nicoli JR, Holzhausen M, Mayer MPA. Probiotics improve re-epithelialization of scratches infected by Porphyromonas gingivalis through up-regulating CXCL8-CXCR1/CXCR2 axis. Anaerobe 2021; 72:102458. [PMID: 34547426 DOI: 10.1016/j.anaerobe.2021.102458] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2021] [Revised: 09/15/2021] [Accepted: 09/16/2021] [Indexed: 10/20/2022]
Abstract
Porphyromonas gingivalis inhibits the release of CXCL8 by gingival epithelial cells and reduces their proliferation. We previously reported that Bifidocaterium sp. and Lactobacillus sp. immunomodulate gingival epithelial cells response to this periodontal pathogen, but their effects on re-epithelialization properties are still unknown. Herein we explored these activities of potential probiotics on gingival epithelial cells and clarified their mechanisms. The immortalized OBA-9 lineage was used to perform in vitro scratches. Twelve clinical isolates and commercially available strains of Bifidobacterium sp. and Lactobacillus sp. were screened. L. casei 324 m and B. pseudolongum 1191A were selected to perform mechanistic assays with P. gingivalis W83 infection and the following parameters were measured: percentage of re-epithelialization by DAPI immunofluorescence area measurement; cell number by Trypan Blue exclusion assay; CXCL8 regulation by ELISA and RT-qPCR; and expression of CXCL8 cognate receptors-CXCR1 and CXCR2 by Flow Cytometry. Complementary mechanistic assays were performed with CXCL8, in the presence or absence of the CXCR1/CXCR2 inhibitor-reparixin. L. casei 324 m and B. pseudolongum 1191A enhanced re-epithelialization/cell proliferation as well as inhibited the harmful effects of P. gingivalis W83 on these activities through an increase in the expression and release of CXCL8 and in the number of cells positive for CXCR1/CXCR2. Further, we revealed that the beneficial effects of these potential probiotics were dependent on activation of the CXCL8-CXCR1/CXCR2 axis. The current findings indicate that these potential probiotics strains may improve wound healing in the context of the periodontal tissues by a CXCL8 dependent mechanism.
Collapse
Affiliation(s)
- Emmanuel Albuquerque-Souza
- Division of Periodontics, Department of Stomatology, School of Dentistry, University of São Paulo, SP, Brazil; Department of Microbiology, Institute of Biomedical Sciences, University of São Paulo, SP, Brazil
| | - Karin Hitomi Ishikawa
- Division of Periodontics, Department of Stomatology, School of Dentistry, University of São Paulo, SP, Brazil
| | - Pâmela Penas Amado
- Department of Microbiology, Institute of Biomedical Sciences, University of São Paulo, SP, Brazil
| | - Jacques Robert Nicoli
- Department of Microbiology, Biological Science Institute, Federal University of Minas Gerais, Belo Horizonte, MG, Brazil
| | - Marinella Holzhausen
- Division of Periodontics, Department of Stomatology, School of Dentistry, University of São Paulo, SP, Brazil
| | - Marcia P A Mayer
- Division of Periodontics, Department of Stomatology, School of Dentistry, University of São Paulo, SP, Brazil; Department of Microbiology, Institute of Biomedical Sciences, University of São Paulo, SP, Brazil.
| |
Collapse
|
40
|
Byeon J, Blizinsky KD, Persaud A, Findley K, Lee J, Buscetta AJ, You S, Bittinger K, Minniti CP, Bonham VL, Grice EA. Insights into the skin microbiome of sickle cell disease leg ulcers. Wound Repair Regen 2021; 29:801-809. [PMID: 33964094 PMCID: PMC8410641 DOI: 10.1111/wrr.12924] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2020] [Revised: 03/10/2021] [Accepted: 04/14/2021] [Indexed: 01/04/2023]
Abstract
Leg ulcers are estimated to occur in 1%-10% of North American patients with sickle cell disease (SCD). Their pathophysiology remains poorly defined, but as with other chronic wounds, it is hypothesised that the microbial milieu, or microbiome, contributes to their healing and clinical outcomes. This study utilises 16S ribosomal RNA (rRNA) gene sequencing to describe, for the first time, the microbiome of the SCD leg ulcer and its association with clinical factors. In a cross-sectional analysis of 42 ulcers, we recovered microbial profiles similar to other chronic wounds in the predominance of anaerobic bacteria and opportunistic pathogens including Staphylococcus, Corynebacterium, and Finegoldia. Ulcers separated into two clusters: one defined by predominance of Staphylococcus and smaller surface area, and the other displaying a greater diversity of taxa and larger surface area. We also find that the relative abundance of Porphyromonas is negatively associated with haemoglobin levels, a key clinical severity indicator for SCD, and that Finegoldia relative abundance is negatively associated with CD19+ B cell count. Finally, ratios of Corynebacterium:Lactobacillus and Staphylococcus:Lactobacillus are elevated in the intact skin of individuals with a history of SCD leg ulcers, while the ratio of Lactobacillus:Bacillus is elevated in that of individuals without a history of ulcers. Investigations of the skin microbiome in relation to SCD ulcer pathophysiology can inform clinical guidelines for this poorly understood chronic wound, as well as enhance broader understanding about the role of the skin microbiome in delayed wound healing.
Collapse
Affiliation(s)
- Julia Byeon
- Social and Behavioral Research BranchNational Human Genome Research Institute, National Institutes of HealthBethesdaMarylandUSA
| | - Katherine D. Blizinsky
- All of Us Research ProgramNational Institutes of HealthBethesdaMarylandUSA
- NeurologyRush Alzheimer's Disease Center, Rush Medical CollegeChicagoIllinoisUSA
| | - Anitra Persaud
- Social and Behavioral Research BranchNational Human Genome Research Institute, National Institutes of HealthBethesdaMarylandUSA
| | - Keisha Findley
- Social and Behavioral Research BranchNational Human Genome Research Institute, National Institutes of HealthBethesdaMarylandUSA
| | - Jung‐Jin Lee
- The Children's Hospital of PhiladelphiaMicrobiome Center Roberts Center for Pediatric ResearchPhiladelphiaPennsylvaniaUSA
| | - Ashley J. Buscetta
- Social and Behavioral Research BranchNational Human Genome Research Institute, National Institutes of HealthBethesdaMarylandUSA
| | - Shuo You
- Department of Medicine, Division of HematologyMontefiore Medical Center, Albert Einstein College of MedicineBronxNew YorkUSA
| | - Kyle Bittinger
- The Children's Hospital of PhiladelphiaMicrobiome Center Roberts Center for Pediatric ResearchPhiladelphiaPennsylvaniaUSA
| | - Caterina P. Minniti
- Albert Einstein College of Medicine, Center for Sickle Cell Disease ResearchMontefiore Medical CenterBronxNew YorkUSA
| | - Vence L. Bonham
- Social and Behavioral Research BranchNational Human Genome Research Institute, National Institutes of HealthBethesdaMarylandUSA
| | - Elizabeth A. Grice
- Department of Dermatology and MicrobiologyUniversity of Pennsylvania Perelman School of MedicinePhiladelphiaPennsylvaniaUSA
| |
Collapse
|
41
|
Nam Y, Kim J, Baek J, Kim W. Improvement of Cutaneous Wound Healing via Topical Application of Heat-Killed Lactococcus chungangensis CAU 1447 on Diabetic Mice. Nutrients 2021; 13:nu13082666. [PMID: 34444827 PMCID: PMC8401197 DOI: 10.3390/nu13082666] [Citation(s) in RCA: 24] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2021] [Revised: 07/28/2021] [Accepted: 07/29/2021] [Indexed: 12/13/2022] Open
Abstract
Cutaneous wound healing comprises a complex systemic network. Probiotics, naturally extracted substances, medicine, and chemical compounds have been used for wound healing, but the application of postbiotics as therapeutic agents has yet to be explored. Our study shows potential beneficial effects of heat-killed Lactococcus chungangensis CAU 1447 on type 1 diabetic mice. The postbiotic strain significantly decreased the skin wound size. The activity of myeloperoxidase secreted from neutrophils also decreased. The molecular mechanism of wound healing was adjusted by important mediators, growth factors, chemokines, and cytokines. These elements regulated the anti-inflammatory activity and accelerated wound healing. To determine the role of the postbiotic in wound repair, we showed a similar taxonomic pattern as compared to the diabetic mice using skin microbiome analysis. These findings demonstrated that heat-killed Lactococcus chungangensis CAU 1447 had beneficial effects on wound healing and can be utilized as postbiotic therapeutic agents.
Collapse
|
42
|
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: 12] [Impact Index Per Article: 3.0] [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
|
43
|
Effect of probiotic Lactobacillus rhamnosus by-products on gingival epithelial cells challenged with Porphyromonas gingivalis. Arch Oral Biol 2021; 128:105174. [PMID: 34058722 DOI: 10.1016/j.archoralbio.2021.105174] [Citation(s) in RCA: 29] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2021] [Revised: 05/04/2021] [Accepted: 05/25/2021] [Indexed: 02/06/2023]
Abstract
OBJECTIVE Probiotics are usually given as living cells, but their effects may be also achieved by postbiotics. We hypothesized that probiotics products (spent media and lysate) altered the response induced by P. gingivalis in gingival epithelial cells (GECS). METHODS Immortalized human OBA-9 GECs (∼2,5 × 105cells/well) were challenged with P. gingivalis ATCC33277, and co-infected with L. rhamnosus Lr-32 for 4 h. L. rhamnosus Lr-32 spent medium or cells lysate was added to GECs co-infected with P. gingivalis. Another set of OBA-9 GECs were first exposed to P. gingivalis ATCC 33277 and then to the living probiotic or probiotic products. Transcription of genes encoding inflammatory mediators (IL-1β, TNF-α, IL-6, and CXCL-8) and receptors (TLR2 and TLR4) were evaluated by RT-qPCR. P. gingivalis growth under L. rhamnosus Lr-32 postbiotics was also evaluated. RESULTS L. rhamnosus Lr-32 spent media decreased cell viability, while living cells and cell lysates did not. L. rhamnosus Lr-32 lysate, but not spent media, upregulated transcription of inflammatory mediators (IL-1β, TNF-α, IL-6, and CXCL-8) in GECs infected with P. gingivalis. Transcription of TRL2 was upregulated in all experimental groups compared to control, whereas TLR4 was upregulated by the probiotic or its postbiotics in P. gingivalis infected cells. Spent media and lysates reduced the growth of P. gingivalis. CONCLUSION L. rhamnosus Lr-32 cell components rather than live probiotic enhanced the expression of inflammatory mediators in P. gingivalis infected gingival epithelial cells. The increased potential of Lr-32 cell lysates to promote immune response to the periodontopathogen may favor pathogen elimination but may also lead to additional deleterious effects of the exacerbated inflammation.
Collapse
|
44
|
The Ambivalent Role of Skin Microbiota and Adrenaline in Wound Healing and the Interplay between Them. Int J Mol Sci 2021; 22:ijms22094996. [PMID: 34066786 PMCID: PMC8125934 DOI: 10.3390/ijms22094996] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2021] [Revised: 04/27/2021] [Accepted: 05/06/2021] [Indexed: 02/07/2023] Open
Abstract
After skin injury, wound healing sets into motion a dynamic process to repair and replace devitalized tissues. The healing process can be divided into four overlapping phases: hemostasis, inflammation, proliferation, and maturation. Skin microbiota has been reported to participate in orchestrating the wound healing both in negative and positive ways. Many studies reported that skin microbiota can impose negative and positive effects on the wound. Recent findings have shown that many bacterial species on human skin are able to convert aromatic amino acids into so-called trace amines (TAs) and convert corresponding precursors into dopamine and serotonin, which are all released into the environment. As a stress reaction, wounded epithelial cells release the hormone adrenaline (epinephrine), which activates the β2-adrenergic receptor (β2-AR), impairing the migration ability of keratinocytes and thus re-epithelization. This is where TAs come into play, as they act as antagonists of β2-AR and thus attenuate the effects of adrenaline. The result is that not only TAs but also TA-producing skin bacteria accelerate wound healing. Adrenergic receptors (ARs) play a key role in many physiological and disease-related processes and are expressed in numerous cell types. In this review, we describe the role of ARs in relation to wound healing in keratinocytes, immune cells, fibroblasts, and blood vessels and the possible role of the skin microbiota in wound healing.
Collapse
|
45
|
Ho CC, Ng SC, Chuang HL, Chen JY, Wen SY, Kuo CH, Mahalakshmi B, Le QV, Huang CY, Kuo WW. Seven traditional Chinese herbal extracts fermented by Lactobacillus rhamnosus provide anti-pigmentation effects by regulating the CREB/MITF/tyrosinase pathway. ENVIRONMENTAL TOXICOLOGY 2021; 36:654-664. [PMID: 33314651 DOI: 10.1002/tox.23069] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/05/2020] [Accepted: 11/20/2020] [Indexed: 06/12/2023]
Abstract
Skin pigmentation is resulted from several processes, such as melanin synthesis transportation and abnormal melanin accumulation in keratinocytes. Various studies have suggested that seven traditional Chinese herbal extracts from Atractylodes macrocephala, Paeonia lactiflora, Bletilla striata, Poria cocos, Dictamnus dasycarpus, Ampelopsis japonica and Tribulus terrestris (which we collectively named ChiBai), show several protective effects toward skin-related diseases. Lactobacillus rhamnosus, a lactic acid bacterium, has been reported to treat skin inflammation and atopic dermatitis. In this study, the broth produced by the cofermentation of ChiBai with Lactobacillus rhamnosus was studied for its effects on skin pigmentation through in vitro and in vitro experiments. In the in vitro experiments, we found that the fermented broth of ChiBai (FB-ChiBai) suppressed alpha-melanocyte stimulating hormone (α-MSH)-induced melanogenesis in B16F0 murine melanoma cells without any cytotoxicity at a concentration of 0.5%. FB-ChiBai significantly attenuated melanin production, tyrosinase activities and melanogenesis-related signaling pathways. Treatment with FB-ChiBai also reduced the nuclear translocation and promoter binding activities of MITF. In the in vivo experiments, FB-ChiBai was topically applied to the dorsal skin of C57BL/6J nude mice and concurrently irradiated with UVB, three times a week for 8 weeks. The results indicated that FB-ChiBai alleviated UVB-induced hyperpigmentation by reducing epidermal hyperplasia and inhibiting the CREB/MITF/tyrosinase pathway. In conclusion, our data indicated that the anti-melanogenic effects of FB-ChiBai are mediated by the inhibition of CREB/MITF/tyrosinase signaling pathway. The findings suggest that FB-ChiBai can protect against UV-B irradiation and that it might be used as an agent in cosmetic products to protect against UVB-induced hyperpigmentation.
Collapse
Affiliation(s)
- Chih-Chu Ho
- Department of Biological Science and Technology, College of Life Sciences, China Medical University, Taichung, Taiwan
| | - Shang-Chuan Ng
- Department of Biological Science and Technology, College of Life Sciences, China Medical University, Taichung, Taiwan
| | - Ho-Lin Chuang
- Department of Biological Science and Technology, College of Life Sciences, China Medical University, Taichung, Taiwan
| | - Jia-Yi Chen
- Department of Biological Science and Technology, College of Life Sciences, China Medical University, Taichung, Taiwan
| | - Su-Ying Wen
- Department of Dermatology, Taipei City Hospital, Renai Branch, Taipei, Taiwan
- Department of Cosmetic Applications and Management, Mackay Junior College of Medicine, Nursing, and Management, Taipei, Taiwan
- Department of Health Care Management, National Taipei University of Nursing and Health Sciences, Taipei, Taiwan
| | - Chia-Hua Kuo
- Department of Sports Sciences, University of Taipei, Taipei, Taiwan
| | - B Mahalakshmi
- Institute of Research and Development, Duy Tan University, Da Nang, Vietnam
| | - Quoc-Vu Le
- Department of Biological Science and Technology, College of Life Sciences, China Medical University, Taichung, Taiwan
| | - Chih-Yang Huang
- Graduate Institute of Biomedical Sciences, China Medical University, Taichung, Taiwan
- Cardiovascular and Mitochondrial Related Disease Research Center, Hualien Tzu Chi Hospital, Buddhist Tzu Chi Medical Foundation, Hualien, Taiwan
- Center of General Education, Buddhist Tzu Chi Medical Foundation, Tzu Chi University of Science and Technology, Hualien, Taiwan
- Department of Medical Research, China Medical University Hospital, China Medical University, Taichung, Taiwan
- Department of Biotechnology, Asia University, Taichung, Taiwan
| | - Wei-Wen Kuo
- Department of Biological Science and Technology, College of Life Sciences, China Medical University, Taichung, Taiwan
| |
Collapse
|
46
|
The role of lactobacilli in inhibiting skin pathogens. Biochem Soc Trans 2021; 49:617-627. [PMID: 33704415 DOI: 10.1042/bst20200329] [Citation(s) in RCA: 28] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2020] [Revised: 02/12/2021] [Accepted: 02/17/2021] [Indexed: 12/25/2022]
Abstract
The human skin microbiota forms a key barrier against skin pathogens and is important in modulating immune responses. Recent studies identify lactobacilli as endogenous inhabitants of healthy skin, while inflammatory skin conditions are often associated with a disturbed skin microbiome. Consequently, lactobacilli-based probiotics are explored as a novel treatment of inflammatory skin conditions through their topical skin application. This review focuses on the potential beneficial role of lactobacilli (family Lactobacillaceae) in the skin habitat, where they can exert multifactorial local mechanisms of action against pathogens and inflammation. On one hand, lactobacilli have been shown to directly compete with skin pathogens through adhesion inhibition, production of antimicrobial metabolites, and by influencing pathogen metabolism. The competitive anti-pathogenic action of lactobacilli has already been described mechanistically for common different skin pathogens, such as Staphylococcus aureus, Cutibacterium acnes, and Candida albicans. On the other hand, lactobacilli also have an immunomodulatory capacity associated with a reduction in excessive skin inflammation. Their influence on the immune system is mediated by bacterial metabolites and cell wall-associated or excreted microbe-associated molecular patterns (MAMPs). In addition, lactobacilli can also enhance the skin barrier function, which is often disrupted as a result of infection or in inflammatory skin diseases. Some clinical trials have already translated these mechanistic insights into beneficial clinical outcomes, showing that topically applied lactobacilli can temporarily colonize the skin and promote skin health, but more and larger clinical trials are required to generate in vivo mechanistic insights and in-depth skin microbiome analysis.
Collapse
|
47
|
Puebla-Barragan S, Reid G. Probiotics in Cosmetic and Personal Care Products: Trends and Challenges. Molecules 2021; 26:1249. [PMID: 33652548 PMCID: PMC7956298 DOI: 10.3390/molecules26051249] [Citation(s) in RCA: 46] [Impact Index Per Article: 11.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2021] [Revised: 02/20/2021] [Accepted: 02/22/2021] [Indexed: 12/11/2022] Open
Abstract
Probiotics, defined as "live microorganisms that, when administered in adequate amounts, confer a health benefit on the host," are becoming increasingly popular and marketable. However, too many of the products currently labelled as probiotics fail to comply with the defining characteristics. In recent years, the cosmetic industry has increased the number of products classified as probiotics. While there are several potential applications for probiotics in personal care products, specifically for oral, skin, and intimate care, proper regulation of the labelling and marketing standards is still required to guarantee that consumers are indeed purchasing a probiotic product. This review explores the current market, regulatory aspects, and potential applications of probiotics in the personal care industry.
Collapse
Affiliation(s)
- Scarlett Puebla-Barragan
- Centre for Human Microbiome and Probiotics, Lawson Health Research Institute, London, ON N6C 2R5, Canada
- Departments of Microbiology & Immunology and Surgery, University of Western Ontario, London, ON N6A 3K7, Canada;
| | - Gregor Reid
- Centre for Human Microbiome and Probiotics, Lawson Health Research Institute, London, ON N6C 2R5, Canada
- Departments of Microbiology & Immunology and Surgery, University of Western Ontario, London, ON N6A 3K7, Canada;
| |
Collapse
|
48
|
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: 1.3] [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
|
49
|
Masunaga A, Kawahara T, Morita H, Nakazawa K, Tokunaga Y, Akita S. Fatty acid potassium improves human dermal fibroblast viability and cytotoxicity, accelerating human epidermal keratinocyte wound healing in vitro and in human chronic wounds. Int Wound J 2021; 18:467-477. [PMID: 33433959 PMCID: PMC8273623 DOI: 10.1111/iwj.13547] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2020] [Revised: 12/19/2020] [Accepted: 12/31/2020] [Indexed: 12/11/2022] Open
Abstract
Effective cleaning of a wound promotes wound healing and favours wound care as it can prevent and control biofilms. The presence of biofilm is associated with prolonged wound healing, increased wound propensity to infection, and delayed wound closure. Anionic potassium salts of fatty acids are tested with commonly used anionic surfactants, such as sodium laureth sulphate (SLES) and sodium lauryl sulphate/sodium dodecyl sulphate (SLS/SDS). The normal human dermal cells demonstrated significantly greater viability in fatty acid potassium, including caprylic acid (C8), capric acid (C10), lauric acid (C12), oleic acid (C18:1), and linoleic acid (C18:2), than in SLES or SLS after a 24-hour incubation. Cytotoxicity by LDH assay in a 5-minute culture in fatty acid potassium was significantly lower than in SLES or SLS. in vitro wound healing of human epidermal keratinocytes during the scratch assay in 24-hour culture was more significantly improved by fatty acid treatment than by SLES or SLS/SDS. In a live/dead assay of human epidermal keratinocytes, C8K and C18:1K demonstrated only green fluorescence, indicating live cells, whereas synthetic surfactants, SLES and SLS, demonstrated red fluorescence on staining with propidium iodide, indicating dead cells after SLES and SLS/SDS treatment. Potassium salts of fatty acids are useful wound cleaning detergents that do not interfere with wound healing, as observed in the scratch assay using human epidermal keratinocytes. As potassium salts of fatty acids are major components of natural soap, which are produced by natural oil and caustic potash using a saponification method, this may be clinically important in wound and peri-wound skin cleaning. In human chronic wounds, natural soap containing fatty acid potassium increased tissue blood flow based on laser speckle flowgraphs after 2 weeks (P < .05), in addition to removing the eschars and debris. Wound cleansing by natural soap of fatty acid potassium is beneficial for wound healing.
Collapse
Affiliation(s)
| | | | | | - Kohji Nakazawa
- Department of Life and Environment Engineering, The University of Kitakyushu, Kitakyushu, Japan
| | - Yuto Tokunaga
- Department of Life and Environment Engineering, The University of Kitakyushu, Kitakyushu, Japan
| | - Sadanori Akita
- Department of Plastic Surgery, Wound Repair and Regeneration, Fukuoka University, Fukuoka, Japan
| |
Collapse
|
50
|
Moreira CF, Cassini-Vieira P, Canesso MCC, Felipetto M, Ranfley H, Teixeira MM, Nicoli JR, Martins FS, Barcelos LS. Lactobacillus rhamnosus CGMCC 1.3724 (LPR) Improves Skin Wound Healing and Reduces Scar Formation in Mice. Probiotics Antimicrob Proteins 2021; 13:709-719. [PMID: 33433898 DOI: 10.1007/s12602-020-09713-z] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 09/24/2020] [Indexed: 01/16/2023]
Abstract
Skin wounds are an important clinical problem which affects millions of people worldwide. The search for new therapeutic approaches to improve wound healing is needed. The present study aimed to evaluate the effects of the oral treatment with the skin-related probiotics Lactobacillus johnsonii LA1 (LJ), L. paracasei ST11 (LP), and L. rhamnosus LPR (LR) in a model of excisional skin wounds in Swiss mice. The animals received daily oral gavage of PBS or 1 × 107 colony-forming units of LJ, LP, or LR, singly, beginning just after the creation of wounds until euthanasia. Blood flow was evaluated by laser Doppler perfusion imaging. Myeloperoxidase and N-acetyl-β-D-glucosaminidase activities were used to assess the accumulation of neutrophils and macrophages, respectively. The wound tissue was also collected for histological analyses (H&E, Toluidine blue, and Picrosirius red staining). The macroscopic wound closure rate was faster only in mice treated with LR, but not with LJ and LP, when compared to mice treated with PBS. Histological evaluations showed that treatment with LR stimulated wound epithelization when compared to PBS. Further analyses showed that wounds from LR-treated mice presented a significant decrease in macrophage (p < 0.001) and mast cell (p < 0.001) infiltration, along with improved angiogenesis (p < 0.001) and blood flow (p < 0.01). Of note, collagen deposition and scarring were reduced in LR-treated mice when compared to PBS-treated mice. In conclusion, our results show that the oral treatment with Lactobacillus rhamnosus accelerates skin wound closure and reduces scar, besides to reducing inflammation and fibrogenesis and improving angiogenesis in the wounded skin.
Collapse
Affiliation(s)
- Camila Francisco Moreira
- Department of Physiology and Biophysics, Institute of Biological Sciences, Federal University of Minas Gerais (UFMG), Av. Antônio Carlos, 6627 - Pampulha, Minas Gerais, 31270-901, Belo Horizonte, Brazil
| | - Puebla Cassini-Vieira
- Department of Physiology and Biophysics, Institute of Biological Sciences, Federal University of Minas Gerais (UFMG), Av. Antônio Carlos, 6627 - Pampulha, Minas Gerais, 31270-901, Belo Horizonte, Brazil
| | - Maria Cecília Campos Canesso
- Department of Physiology and Biophysics, Institute of Biological Sciences, Federal University of Minas Gerais (UFMG), Av. Antônio Carlos, 6627 - Pampulha, Minas Gerais, 31270-901, Belo Horizonte, Brazil
| | - Mariane Felipetto
- Department of Physiology and Biophysics, Institute of Biological Sciences, Federal University of Minas Gerais (UFMG), Av. Antônio Carlos, 6627 - Pampulha, Minas Gerais, 31270-901, Belo Horizonte, Brazil
| | - Hedden Ranfley
- Department of Physiology and Biophysics, Institute of Biological Sciences, Federal University of Minas Gerais (UFMG), Av. Antônio Carlos, 6627 - Pampulha, Minas Gerais, 31270-901, Belo Horizonte, Brazil
| | - Mauro Martins Teixeira
- Department of Biochemistry and Immunology, Institute of Biological Sciences, Federal University of Minas Gerais, Minas Gerais, Belo Horizonte, Brazil
| | - Jacques Robert Nicoli
- Department of Microbiology, Institute of Biological Sciences, Federal University of Minas Gerais, Minas Gerais, Belo Horizonte, Brazil
| | - Flaviano Santos Martins
- Department of Microbiology, Institute of Biological Sciences, Federal University of Minas Gerais, Minas Gerais, Belo Horizonte, Brazil
| | - Lucíola Silva Barcelos
- Department of Physiology and Biophysics, Institute of Biological Sciences, Federal University of Minas Gerais (UFMG), Av. Antônio Carlos, 6627 - Pampulha, Minas Gerais, 31270-901, Belo Horizonte, Brazil.
| |
Collapse
|