1
|
Chen RY, Shi JJ, Liu YJ, Yu J, Li CY, Tao F, Cao JF, Yang GJ, Chen J. The State-of-the-Art Antibacterial Activities of Glycyrrhizin: A Comprehensive Review. Microorganisms 2024; 12:1155. [PMID: 38930536 PMCID: PMC11206003 DOI: 10.3390/microorganisms12061155] [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: 05/11/2024] [Revised: 05/30/2024] [Accepted: 06/03/2024] [Indexed: 06/28/2024] Open
Abstract
Licorice (Glycyrrhiza glabra) is a plant of the genus Glycyrrhiza in the family Fabaceae/Leguminosae and is a renowned natural herb with a long history of medicinal use dating back to ancient times. Glycyrrhizin (GLY), the main active component of licorice, serves as a widely utilized therapeutic agent in clinical practice. GLY exhibits diverse medicinal properties, including anti-inflammatory, antibacterial, antiviral, antitumor, immunomodulatory, intestinal environment maintenance, and liver protection effects. However, current research primarily emphasizes GLY's antiviral activity, while providing limited insight into its antibacterial properties. GLY demonstrates a broad spectrum of antibacterial activity via inhibiting the growth of bacteria by targeting bacterial enzymes, impacting cell membrane formation, and altering membrane permeability. Moreover, GLY can also bolster host immunity by activating pertinent immune pathways, thereby enhancing pathogen clearance. This paper reviews GLY's inhibitory mechanisms against various pathogenic bacteria-induced pathological changes, its role as a high-mobility group box 1 inhibitor in immune regulation, and its efficacy in combating diseases caused by pathogenic bacteria. Furthermore, combining GLY with other antibiotics reduces the minimum inhibitory concentration, potentially aiding in the clinical development of combination therapies against drug-resistant bacteria. Sources of information were searched using PubMed, Web of Science, Science Direct, and GreenMedical for the keywords "licorice", "Glycyrrhizin", "antibacterial", "anti-inflammatory", "HMGB1", and combinations thereof, mainly from articles published from 1979 to 2024, with no language restrictions. Screening was carried out by one author and supplemented by others. Papers with experimental flaws in their experimental design and papers that did not meet expectations (antifungal papers, etc.) were excluded.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | - Guan-Jun Yang
- State Key Laboratory for Managing Biotic and Chemical Threats to the Quality and Safety of Agro-Products, School of Marine Sciences, Ningbo University, Ningbo 315211, China (J.-J.S.); (Y.-J.L.); (J.Y.); (C.-Y.L.); (F.T.); (J.-F.C.)
| | - Jiong Chen
- State Key Laboratory for Managing Biotic and Chemical Threats to the Quality and Safety of Agro-Products, School of Marine Sciences, Ningbo University, Ningbo 315211, China (J.-J.S.); (Y.-J.L.); (J.Y.); (C.-Y.L.); (F.T.); (J.-F.C.)
| |
Collapse
|
2
|
Luan J, Zhu Y, Lin J, Zhang Y, Xu Q, Zhan L, Tian X, Zhao G, Peng X. Quercetin protects against Aspergillus fumigatus keratitis by reducing fungal load and inhibiting TLR-4 induced inflammatory response. Cytokine 2023; 171:156356. [PMID: 37677994 DOI: 10.1016/j.cyto.2023.156356] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2023] [Revised: 08/01/2023] [Accepted: 08/30/2023] [Indexed: 09/09/2023]
Abstract
PURPOSE To investigate the antifungal and anti-inflammatory effects of quercetin in Aspergillus fumigatus (A. fumigatus) keratitis. METHODS Draize eye test was performed in mice to evaluate the toxicity of quercetin, and the antifungal effects on A. fumigatus were assessed via scanning electron microscopy (SEM), propidium iodide uptake, and adherence assay. In fungal keratitis (FK) mouse models, immunostaining was performed for investigating toll-like receptor 4 (TLR-4) expression and macrophage infiltration. Real-time PCR, ELISA, and Western blot were used to evaluate the expression of pro-inflammatory factors IL-1β, TNF-α, and IL-6 in infected RAW264.7 cells. Cells were also treated with TLR-4 siRNA or agonist CRX-527 to investigate mechanisms underlying the anti-inflammatory activity of quercetin. RESULTS Quercetin at 32 μM was non-toxic to corneal epithelial and significantly inhibited A. fumigatus growth and adhesion, and also altered the structure and reduced the number of mycelia. Quercetin significantly reduced macrophage infiltration in the mouse cornea, and attenuated the expression of TLR-4 in the corneal epithelium and stroma of mice with keratitis caused by A. fumigatus. In RAW264.7 cells infected by A. fumigatus, quercetin downregulated TLR-4 along with pro-inflammatory factors IL-1β, TNF-α, and IL-6. RAW cells with TLR-4 knockdown had reduced expression of factors after A. fumigatus infection, which was decreased even further with quercetin treatment. In contrast, cells with CRX-527 had elevated inflammatory factors compared to control, which was significantly attenuated in the presence of quercetin. CONCLUSION Quercetin plays a protective role in mouse A. fumigatus keratitis by inhibiting fungal load, disrupting hyphae structure, macrophage infiltration, and suppressing inflammation response in macrophages via TLR-4 mediated signaling pathway.
Collapse
Affiliation(s)
- Junjie Luan
- Department of Ophthalmology, The Affiliated Hospital of Qingdao University, Qingdao, China.
| | - Yunan Zhu
- Department of Ophthalmology, The Affiliated Hospital of Qingdao University, Qingdao, China; Department of Ophthalmology, Cangzhou Central Hospital, Cangzhou, China.
| | - Jing Lin
- Department of Ophthalmology, The Affiliated Hospital of Qingdao University, Qingdao, China.
| | - Yingxue Zhang
- Department of Biochemistry, Microbiology, and Immunology, Wayne State University School of Medicine, Detroit, MI 48201, United States.
| | - Qiang Xu
- Department of Ophthalmology, The Affiliated Hospital of Qingdao University, Qingdao, China.
| | - Lu Zhan
- Department of Ophthalmology, The Affiliated Hospital of Qingdao University, Qingdao, China.
| | - Xue Tian
- Department of Ophthalmology, The Affiliated Hospital of Qingdao University, Qingdao, China.
| | - Guiqiu Zhao
- Department of Ophthalmology, The Affiliated Hospital of Qingdao University, Qingdao, China.
| | - Xudong Peng
- Department of Ophthalmology, The Affiliated Hospital of Qingdao University, Qingdao, China; Department of Ophthalmology, University of Washington, Seattle, WA, United States.
| |
Collapse
|
3
|
Hazlett LD, Xu S, Somayajulu M, McClellan SA. Host-microbe interactions in cornea. Ocul Surf 2023; 28:413-423. [PMID: 34619389 PMCID: PMC8977393 DOI: 10.1016/j.jtos.2021.09.008] [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: 08/20/2021] [Revised: 09/23/2021] [Accepted: 09/26/2021] [Indexed: 11/23/2022]
Abstract
Corneal infections result through interaction between microbes and host innate immune receptors. Damage to the cornea occurs as a result of microbial virulence factors and is often exacerbated by lack of a controlled host immune response; the latter contributing to bystander damage to corneal structure. Understanding mechanisms involved in host microbial interactions is critical to development of novel therapeutic targets, ultimate control of microbial pathogenesis, and restoration of tissue homeostasis. Studies on these interactions continue to provide exciting findings directly related to this ultimate goal.
Collapse
Affiliation(s)
- Linda D Hazlett
- Department of Ophthalmology, Visual and Anatomical Sciences, Wayne State University School of Medicine, Detroit, MI, 48201, USA.
| | - Shunbin Xu
- Department of Ophthalmology, Visual and Anatomical Sciences, Wayne State University School of Medicine, Detroit, MI, 48201, USA
| | - Mallika Somayajulu
- Department of Ophthalmology, Visual and Anatomical Sciences, Wayne State University School of Medicine, Detroit, MI, 48201, USA
| | - Sharon A McClellan
- Department of Ophthalmology, Visual and Anatomical Sciences, Wayne State University School of Medicine, Detroit, MI, 48201, USA
| |
Collapse
|
4
|
Glycyrrhizin-Based Hydrogels Accelerate Wound Healing of Normoglycemic and Diabetic Mouse Skin. Pharmaceutics 2022; 15:pharmaceutics15010027. [PMID: 36678656 PMCID: PMC9861362 DOI: 10.3390/pharmaceutics15010027] [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: 11/14/2022] [Revised: 12/01/2022] [Accepted: 12/08/2022] [Indexed: 12/24/2022] Open
Abstract
Efficient wound repair is crucial for mammalian survival. Healing of skin wounds is severely hampered in diabetic patients, resulting in chronic non-healing wounds that are difficult to treat. High-mobility group box 1 (HMGB1) is an important signaling molecule that is released during wounding, thereby delaying regenerative responses in the skin. Here, we show that dissolving glycyrrhizin, a potent HMGB1 inhibitor, in water results in the formation of a hydrogel with remarkable rheological properties. We demonstrate that these glycyrrhizin-based hydrogels accelerate cutaneous wound closure in normoglycemic and diabetic mice by influencing keratinocyte migration. To facilitate topical application of glycyrrhizin hydrogels on cutaneous wounds, several concentrations of glycyrrhizinic acid in water were tested for their rheological, structural, and biological properties. By varying the concentration of glycyrrhizin, these hydrogel properties can be readily tuned, enabling customized wound care.
Collapse
|
5
|
Lin H, Wang Q, Niu Y, Gu L, Hu L, Li C, Zhao G. Antifungal and Anti-inflammatory Effect of Punicalagin on Murine Aspergillus fumigatus Keratitis. Curr Eye Res 2021; 47:517-524. [PMID: 34797193 DOI: 10.1080/02713683.2021.2008982] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/19/2022]
Abstract
PURPOSE This study aimed to investigate the anti-inflammatory effect and antifungal effect of punicalagin in murine fungal keratitis. METHODS We used in vitro and in vivo protocols to assess the anti-inflammatory effect and antifungal effect of punicalagin. In vitro, time kill and mycelial stain were done. In vivo, murine fungal keratitis was established and treated with PBS or PUN. Clinical scores were taken on days 1, 3, and 5 post infection. The mRNA and protein levels of inflammatory factors were detected by RT-PCR and Western blot, and the number and location of macrophages were analyzed by flow cytometry and immunofluorescence. Also, fungal plate counting was used to assess the antifungal effect. The DCFH-DA fluorescence probe detected the ROS level. RESULTS In vitro, PUN showed activity against A.fumigatus. (A.F.), with MIC90 values of 250 μg/ml, and significantly reduced A.F. biofilm formation (p < .001). In vivo, the mouse fungal keratitis model after punicalagin treatment exhibited less disease, lower clinical scores (p < .05), lower reduced macrophage infiltrate (p < .001), and fungal load (p < .001) than those treated with PBS. Treatment with punicalagin also reduced the mRNA expression and protein level of pro-inflammatory factors. At the cellular level, PUN significantly reduced the mRNA expression of inflammatory factors and ROS production caused by the stimulation of mycelia in RAW264.7 (p < .001). CONCLUSIONS The results show that punicalagin is beneficial in the treatment of murine fungal keratitis. The mechanism of its anti-inflammatory effect was synthetical, including antifungal activity, an inhibitory effect of proinflammatory factor and macrophages, and anti-oxidation.
Collapse
Affiliation(s)
- Hao Lin
- Department of Ophthalmology, The Affiliated Hospital of Qingdao University, Qingdao, China
| | - Qian Wang
- Department of Ophthalmology, The Affiliated Hospital of Qingdao University, Qingdao, China
| | - Yawen Niu
- Department of Ophthalmology, The Affiliated Hospital of Qingdao University, Qingdao, China
| | - Lingwen Gu
- Department of Ophthalmology, The Affiliated Hospital of Qingdao University, Qingdao, China
| | - Liting Hu
- Department of Ophthalmology, The Affiliated Hospital of Qingdao University, Qingdao, China
| | - Cui Li
- Department of Ophthalmology, The Affiliated Hospital of Qingdao University, Qingdao, China
| | - Guiqiu Zhao
- Department of Ophthalmology, The Affiliated Hospital of Qingdao University, Qingdao, China
| |
Collapse
|
6
|
Zhang R, Peng X, Lin J, Zhang Y, Zhan L, Tian X, Yin J, Zhao G. The Role of SREC-Ⅰ in Innate Immunity to Aspergillus fumigatus Keratitis. Invest Ophthalmol Vis Sci 2021; 62:12. [PMID: 34236384 PMCID: PMC8267217 DOI: 10.1167/iovs.62.9.12] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
Purpose To determine the role of scavenger receptor expressed by endothelial cell-1 (SREC-Ⅰ) in vitro and in a mouse model of Aspergillus fumigatus keratitis. Methods SREC-Ⅰ mRNA and protein expression were tested in both normal and A fumigatus stimulated human corneal epithelial cells (HCECs). Immunofluorescence was used to detect SREC-Ⅰ expression in human corneas with or without A fumigatus infection. HCECs were incubated with SREC-Ⅰ small interfering RNA, then the mRNA levels of LOX-1, IL-1β, and TNF-α were detected after A fumigatus stimulation. A mouse fungal keratitis (FK) model was established and SREC-Ⅰ mRNA and protein expression were detected by RT-PCR, Western blot and immunofluorescence. The severity of FK was evaluated by clinical score. CLCX1, LOX-1, IL-1β, and TNF-α mRNA expression levels were tested before and after anti-SREC-Ⅰ treatment. Results SREC-Ⅰ expressed in normal and A fumigatus treated HCECs and human corneal epithelium. In vitro experiment showed that SREC-Ⅰ mRNA and protein levels were significantly increased after A fumigatus stimulation. SREC-Ⅰ small interfering RNA treatment inhibited the expressions of LOX-1, IL-1β, and TNF-α in HCECs. The expressions of CLCX1, LOX-1, IL-1β, and TNF-α were elevated in mice with A fumigatus keratitis, which could be decreased by SREC-Ⅰ-neutralizing antibody treatment. Conclusions SREC-Ⅰ is a key mediator in inflammatory response induced by A fumigatus keratitis. SREC-Ⅰ blockade could be a potential therapeutic approach for FK.
Collapse
Affiliation(s)
- Ranran Zhang
- Department of Ophthalmology, The Affiliated Hospital of Qingdao University, Qingdao, Shandong Province, China
| | - Xudong Peng
- Department of Ophthalmology, The Affiliated Hospital of Qingdao University, Qingdao, Shandong Province, China
| | - Jing Lin
- Department of Ophthalmology, The Affiliated Hospital of Qingdao University, Qingdao, Shandong Province, China
| | - Yingxue Zhang
- Department of Biochemistry, Microbiology and Immunology, Wayne State University School of Medicine, Detroit, Michigan, United States
| | - Lu Zhan
- Department of Ophthalmology, The Affiliated Hospital of Qingdao University, Qingdao, Shandong Province, China
| | - Xue Tian
- Department of Ophthalmology, The Affiliated Hospital of Qingdao University, Qingdao, Shandong Province, China
| | - Jiao Yin
- Department of Ophthalmology, The Affiliated Hospital of Qingdao University, Qingdao, Shandong Province, China
| | - Guiqiu Zhao
- Department of Ophthalmology, The Affiliated Hospital of Qingdao University, Qingdao, Shandong Province, China
| |
Collapse
|
7
|
Zhu Y, Peng X, Zhang Y, Lin J, Zhao G. Baicalein Protects Against Aspergillus fumigatus Keratitis by Reducing Fungal Load and Inhibiting TSLP-Induced Inflammatory Response. Invest Ophthalmol Vis Sci 2021; 62:26. [PMID: 34038512 PMCID: PMC8164373 DOI: 10.1167/iovs.62.6.26] [Citation(s) in RCA: 36] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
Abstract
Purpose To investigate the antifungal and anti-inflammatory effects of baicalein on Aspergillus fumigatus (A. fumigatus) keratitis and the underlying mechanisms. Methods The noncytotoxic antifungal concentration of baicalein was determined using CCK8, cell scratch assay, minimum inhibitory concentration, biofilm formation, scanning electron microscopy, propidium iodide uptake test and adherence assay in vitro and Draize test in vivo. In fungal keratitis (FK) mouse models, clinical score and plate count were used to evaluate FK severity, and myeloperoxidase assay and immunofluorescence staining were performed to examine neutrophil infiltration and activity. Real-time PCR, ELISA, and Western blot were performed to explore the anti-inflammatory activity of baicalein and the underlying mechanisms in vivo and in vitro. Results Baicalein at 0.25 mM (noncytotoxic) significantly inhibited A. fumigatus growth, biofilm formation, and adhesion in vitro. In A. fumigatus keratitis mice, baicalein mitigated FK severity, reduced fungal load, and inhibited neutrophil infiltration and activity. Baicalein not only suppressed mRNA and protein levels of proinflammatory factors IL-1β, IL-6, and TNF-α, but also inhibited the expression of thymic stromal lymphopoietin (TSLP) and TSLP receptor (TSLPR) in vivo and in vitro. In HCECs, mRNA and protein levels of IL-1β, IL-6, and TNF-α were significantly lower in the TSLP siRNA–treated group, while higher in the rTSLP-treated group than in the corresponding control. Baicalein treatment significantly inhibited rTSLP induced the expression of IL-1β, IL-6, and TNF-α. Conclusions Baicalein plays a protective role in mouse A. fumigatus keratitis by inhibiting fungal growth, biofilm formation, and adhesion, and suppressing inflammatory response via downregulation of the TSLP/TSLPR pathway.
Collapse
Affiliation(s)
- Yunan Zhu
- Department of Ophthalmology, The Affiliated Hospital of Qingdao University, Qingdao, Shandong Province, China
| | - Xudong Peng
- Department of Ophthalmology, The Affiliated Hospital of Qingdao University, Qingdao, Shandong Province, China
| | - Yingxue Zhang
- Department of Biochemistry, Microbiology and Immunology, Wayne State University School of Medicine, Detroit, Michigan, United States
| | - Jing Lin
- Department of Ophthalmology, The Affiliated Hospital of Qingdao University, Qingdao, Shandong Province, China
| | - Guiqiu Zhao
- Department of Ophthalmology, The Affiliated Hospital of Qingdao University, Qingdao, Shandong Province, China
| |
Collapse
|
8
|
Al-Radadi NS. Facile one-step green synthesis of gold nanoparticles (AuNp) using licorice root extract: Antimicrobial and anticancer study against HepG2 cell line. ARAB J CHEM 2021. [DOI: 10.1016/j.arabjc.2020.102956] [Citation(s) in RCA: 18] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/22/2023] Open
|
9
|
Fan Y, Li C, Peng X, Jiang N, Hu L, Gu L, Zhu G, Zhao G, Lin J. Perillaldehyde Ameliorates Aspergillus fumigatus Keratitis by Activating the Nrf2/HO-1 Signaling Pathway and Inhibiting Dectin-1-Mediated Inflammation. Invest Ophthalmol Vis Sci 2021; 61:51. [PMID: 32579678 PMCID: PMC7415897 DOI: 10.1167/iovs.61.6.51] [Citation(s) in RCA: 36] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022] Open
Abstract
Purpose The purpose of this study was to investigate the therapeutic effect of perillaldehyde (PAE) on Aspergillus fumigatus (A. fumigatus) keratitis. Methods Human corneal epithelial cells (HCECs) were pretreated with PAE and stimulated with A. fumigatus mycelium. C57BL/6 mice were infected with A. fumigatus and treated with or without PAE 1 day after infection. Clinical scores, PCR, ELISA, and Western blotting were used to detect the expression of pro-inflammatory mediators, dendritic cell-associated c-type lectin-1 (Dectin-1), nuclear factor (erythroid-derived 2) like 2 (Nrf2), and heme oxygenase (HO-1). Nrf2 expression in HCECs pretreated with PAE was observed by immunofluorescence. NIMP-R14 protein expression and localization in mouse corneas were observed by immunofluorescence staining after treatment with PAE. Corneal colony counting, time-kill tests, and mycelial transformation inhibition tests were used to evaluate the antifungal effect of PAE. Results C57BL/6 mice treated with PAE at 1 day after infection had a lower clinical score and decreased IL-1β, TNF-α, IL-6, Dectin-1, and MPO levels. PAE treatment significantly reduced neutrophil recruitments to the corneal stroma. Compared with the DMSO-treated group, PAE treatment significantly decreased mRNA and protein levels of pro-inflammatory cytokines and Dectin-1 in HCECs. PAE pretreatment before A. fumigatus stimulation obviously elevated the mRNA and protein levels of components of the Nrf2/HO-1 axis. HCECs pretreated with PAE before infection showed a weakened ability to inhibit inflammation in the presence of brusatol (BT; an Nrf2 inhibitor) or ZnPP (an HO-1 inhibitor). PAE treatment significantly reduced the fungal load of C57BL/6 mouse corneas and inhibited fungal growth in vitro. Conclusions These data proved that PAE may ameliorate A. fumigatus keratitis by activating the Nrf2/HO-1 signaling pathway and inhibiting the Dectin-1 mediated inflammatory response and neutrophil recruitment. Furthermore, PAE exerts direct fungicidal activity on A. fumigatus.
Collapse
|
10
|
Somayajulu M, McClellan SA, Pitchaikannu A, Bessert D, Liu L, Steinle J, Hazlett LD. Effects of Glycyrrhizin Treatment on Diabetic Cornea. J Ocul Pharmacol Ther 2020; 37:12-23. [PMID: 33347772 DOI: 10.1089/jop.2020.0105] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022] Open
Abstract
Purpose: To test how glycyrrhizin (GLY) affects mouse corneal epithelial cells (MCEC) and the diabetic murine cornea. Methods: Viability of MCEC grown under normal or high glucose (HG) with/without GLY was tested by an MTT assay. In addition, C57BL/6 mice were injected with streptozotocin and a subset of control and diabetic mice received GLY in their drinking water. mRNA and protein levels of proinflammatory and oxidative stress molecules were tested by reverse transcription-polymerase chain reaction (RT-PCR) in both models. Ex vivo studies using human diabetic versus control corneas analyzed proinflammatory and oxidative stress markers using RT-PCR and enzyme-linked immunosorbent assay. Results: GLY protected against loss of cell viability induced by HG and significantly reduced HMGB1, IL-1β, TLR2, TLR4, NLRP3, COX2, SOD2, HO-1, GPX2, and GR1. In vivo, corneas of GLY-treated diabetic mice showed significantly decreased mRNA expression for CXCL2, iNOS, and all molecules listed above; GLY also lowered HMGB1 and IL-1β proteins (in vitro and in vivo). Ex vivo studies using diabetic human corneas revealed elevated mRNA levels of inflammatory and oxidative stress molecules (as listed above for in vivo) versus normal age-matched controls. Protein levels for HMGB1 and IL-1β also were elevated in diabetic human versus control corneas. Conclusions: The data provide evidence that GLY treatment attenuates inflammation and oxidative stress in vitro in MCEC and in vivo in the cornea of diabetic mice. Ex vivo data support the similarities of proinflammatory and oxidative stress data in mouse compared to human, suggesting that GLY treatment would have relevancy to patient care.
Collapse
Affiliation(s)
- Mallika Somayajulu
- Department of Ophthalmology, Visual and Anatomical Sciences, Wayne State University School of Medicine, Detroit, Michigan, USA
| | - Sharon A McClellan
- Department of Ophthalmology, Visual and Anatomical Sciences, Wayne State University School of Medicine, Detroit, Michigan, USA
| | - Ahalya Pitchaikannu
- Department of Ophthalmology, Visual and Anatomical Sciences, Wayne State University School of Medicine, Detroit, Michigan, USA
| | - Denise Bessert
- Department of Ophthalmology, Visual and Anatomical Sciences, Wayne State University School of Medicine, Detroit, Michigan, USA
| | - Li Liu
- Department of Ophthalmology, Visual and Anatomical Sciences, Wayne State University School of Medicine, Detroit, Michigan, USA
| | - Jena Steinle
- Department of Ophthalmology, Visual and Anatomical Sciences, Wayne State University School of Medicine, Detroit, Michigan, USA
| | - Linda D Hazlett
- Department of Ophthalmology, Visual and Anatomical Sciences, Wayne State University School of Medicine, Detroit, Michigan, USA
| |
Collapse
|
11
|
Carruthers NJ, McClellan SA, Somayajulu M, Pitchaikannu A, Bessert D, Peng X, Huitsing K, Stemmer PM, Hazlett LD. Effects of Glycyrrhizin on Multi-Drug Resistant Pseudomonas aeruginosa. Pathogens 2020; 9:pathogens9090766. [PMID: 32962036 PMCID: PMC7557769 DOI: 10.3390/pathogens9090766] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2020] [Accepted: 09/16/2020] [Indexed: 02/06/2023] Open
Abstract
The effects of glycyrrhizin (GLY) on multi-drug resistant (MDR) systemic (MDR9) vs. ocular (B1045) Pseudomonas aeruginosa clinical isolates were determined. Proteomes of each isolate with/without GLY treatment were profiled using liquid chromatography mass spectrometry (LC-MS/MS). The effect of GLY on adherence of MDR isolates to immortalized human (HCET) and mouse (MCEC) corneal epithelial cells, and biofilm and dispersal was tested. Both isolates were treated with GLY (0.25 minimum inhibitory concentration (MIC), 10 mg/mL for MDR9 and 3.75 mg/mL for B1045) and subjected to proteomic analysis. MDR9 had a greater response to GLY (51% of identified proteins affected vs. <1% in B1045). In MDR9 vs. controls, GLY decreased the abundance of proteins for: antibiotic resistance, biofilm formation, and type III secretion. Further, antibiotic resistance and type III secretion proteins had higher control abundances in MDR9 vs. B1045. GLY (5 and 10 mg/mL) significantly reduced binding of both isolates to MCEC, and B1045 to HCET. MDR9 binding to HCET was only reduced at 10 mg/mL GLY. GLY (5 and 10 mg/mL) enhanced dispersal for both isolates, at early (6.5 h) but not later times (24–72 h). This study provides evidence that GLY has a greater effect on the proteome of MDR9 vs. B1045, yet it was equally effective at disrupting adherence and early biofilm dispersal.
Collapse
Affiliation(s)
- Nicholas J. Carruthers
- Institute of Environmental Health Sciences, Wayne State University School of Medicine, 540 E. Canfield Avenue, Detroit, MI 48201, USA; (N.J.C.); (P.M.S.)
| | - Sharon A. McClellan
- Department of Ophthalmology, Visual and Anatomical Sciences, Wayne State University, School of Medicine, Detroit, MI 48201, USA; (S.A.M.); (M.S.); (A.P.); (D.B.); (K.H.)
| | - Mallika Somayajulu
- Department of Ophthalmology, Visual and Anatomical Sciences, Wayne State University, School of Medicine, Detroit, MI 48201, USA; (S.A.M.); (M.S.); (A.P.); (D.B.); (K.H.)
| | - Ahalya Pitchaikannu
- Department of Ophthalmology, Visual and Anatomical Sciences, Wayne State University, School of Medicine, Detroit, MI 48201, USA; (S.A.M.); (M.S.); (A.P.); (D.B.); (K.H.)
| | - Denise Bessert
- Department of Ophthalmology, Visual and Anatomical Sciences, Wayne State University, School of Medicine, Detroit, MI 48201, USA; (S.A.M.); (M.S.); (A.P.); (D.B.); (K.H.)
| | - Xudong Peng
- Department of Ophthalmology, The Affiliated Hospital of Qingdao University, Qingdao 266071, China;
| | - Kylie Huitsing
- Department of Ophthalmology, Visual and Anatomical Sciences, Wayne State University, School of Medicine, Detroit, MI 48201, USA; (S.A.M.); (M.S.); (A.P.); (D.B.); (K.H.)
| | - Paul M. Stemmer
- Institute of Environmental Health Sciences, Wayne State University School of Medicine, 540 E. Canfield Avenue, Detroit, MI 48201, USA; (N.J.C.); (P.M.S.)
| | - Linda D. Hazlett
- Department of Ophthalmology, Visual and Anatomical Sciences, Wayne State University, School of Medicine, Detroit, MI 48201, USA; (S.A.M.); (M.S.); (A.P.); (D.B.); (K.H.)
- Correspondence: ; Tel.: +1-313-577-1079; Fax: +1-313-577-3125
| |
Collapse
|
12
|
Somayajulu M, Ekanayaka S, McClellan SA, Bessert D, Pitchaikannu A, Zhang K, Hazlett LD. Airborne Particulates Affect Corneal Homeostasis and Immunity. Invest Ophthalmol Vis Sci 2020; 61:23. [PMID: 32301974 PMCID: PMC7401652 DOI: 10.1167/iovs.61.4.23] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022] Open
Abstract
Purpose To determine the effects of airborne particulate matter (PM) <2.5 µm in vitro and on the normal and Pseudomonas aeruginosa (PA)-infected cornea. Methods An MTT viability assay tested the effects of PM2.5 on mouse corneal epithelial cells (MCEC) and human corneal epithelial cells (HCET). MCEC were tested for reactive oxygen species using a 2′,7′-dichlorodihydrofluorescein assay; RT-PCR determined mRNA levels of inflammatory and oxidative stress markers in MCEC (HMGB1, toll-like receptor 2, IL-1β, CXCL2, GPX1, GPX2, GR1, superoxide dismutase 2, and heme oxygenase 1) and HCET (high mobility group box 1, CXCL2, and IL-1β). C57BL/6 mice also were infected and after 6 hours, the PM2.5 was topically applied. Disease was graded by clinical score and evaluated by histology, plate count, myeloperoxidase assay, RT-PCR, ELISA, and Western blot. Results After PM2.5 (25–200 µg/mL), 80% to 90% of MCEC and HCET were viable and PM exposure increased reactive oxygen species in MCEC and mRNA expression levels for inflammatory and oxidative stress markers in mouse and human cells. In vivo, the cornea of PA+PM2.5 exposed mice exhibited earlier perforation over PA alone (confirmed histologically). In cornea, plate counts were increased after PA+PM2.5, whereas myeloperoxidase activity was significantly increased after PA+PM2.5 over other groups. The mRNA levels for several proinflammatory and oxidative stress markers were increased in the cornea in the PA+PM2.5 over other groups; protein levels were elevated for high mobility group box 1, but not toll-like receptor 4 or glutathione reductase 1. Uninfected corneas treated with PM2.5 did not differ from normal. Conclusions PM2.5 triggers reactive oxygen species, upregulates mRNA levels of oxidative stress, inflammatory markers, and high mobility group box 1 protein, contributing to perforation in PA-infected corneas.
Collapse
|
13
|
Hazlett LD, Ekanayaka SA, McClellan SA, Francis R. Glycyrrhizin Use for Multi-Drug Resistant Pseudomonas aeruginosa: In Vitro and In Vivo Studies. Invest Ophthalmol Vis Sci 2019; 60:2978-2989. [PMID: 31311033 PMCID: PMC6944246 DOI: 10.1167/iovs.19-27200] [Citation(s) in RCA: 23] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2019] [Accepted: 06/14/2019] [Indexed: 02/07/2023] Open
Abstract
Purpose Our purpose was to test glycyrrhizin (GLY) effects and ciprofloxacin interactions on multidrug resistant (MDR) isolates of Pseudomonas aeruginosa in vitro and in vivo in a mouse model of keratitis. Methods A Hardy-disk tested antibiotic sensitivity of isolates MDR9 (nonocular) and B1045 (ocular). GLY MIC (both isolates) and ciprofloxacin was determined spectrophotometrically. A live/dead assay using confocal microscopy and plate count, tested GLY effects on bacterial permeabilization/viability. Proteomics profiled bacterial efflux pumps (MDR9 vs. PAO1); RT-PCR comparatively tested GLY effects on their mRNA expression levels. The activity of efflux pumps was tested using ethidium bromide (EB); and scanning electron microscopy (SEM) visualized the effects of GLY treatment of bacteria. A combination of GLY and ciprofloxacin was tested in C57BL/6 mice (begun 18 hours after infection) and disease scored, photographed and MPO and plate counts done. Results MDR9 was resistant to 6/12 and B1045 to 7/12 antibiotics (both to ciprofloxacin). MIC GLY for MDR9 was 40 mg/mL and 15 mg/mL for B1045. Ciprofloxacin MIC (32 μg/mL) was reduced 2-fold to 16 μg/mL when ciprofloxacin and GLY were combined. GLY altered bacterial membrane permeability and reduced viability. Proteomics revealed increased efflux pumps in MDR9 versus PAO1; GLY reduced their mRNA expression levels and EB suggested decreased activity. In C57BL/6 mice, treatment with GLY and ciprofloxacin versus ciprofloxacin, significantly reduced clinical scores, plate count, and MPO. Conclusions GLY decreases MDR by: altering bacterial parameters, including viability and efflux pump activity. In vivo, it increases the effectiveness of ciprofloxacin, reducing ocular disease, plate count, and MPO activity.
Collapse
MESH Headings
- Animals
- Anti-Bacterial Agents/therapeutic use
- Anti-Inflammatory Agents/therapeutic use
- Ciprofloxacin/therapeutic use
- Corneal Ulcer/drug therapy
- Corneal Ulcer/microbiology
- Drug Resistance, Multiple, Bacterial/drug effects
- Drug Therapy, Combination
- Eye Infections, Bacterial/drug therapy
- Eye Infections, Bacterial/microbiology
- Female
- Glycyrrhizic Acid/therapeutic use
- Mice
- Mice, Inbred C57BL
- Microbial Sensitivity Tests
- Microscopy, Confocal
- Microscopy, Electron, Scanning
- Pseudomonas Infections/drug therapy
- Pseudomonas Infections/microbiology
- Pseudomonas aeruginosa/drug effects
- Pseudomonas aeruginosa/metabolism
- Pseudomonas aeruginosa/ultrastructure
- RNA, Bacterial/genetics
- RNA, Messenger/genetics
- Real-Time Polymerase Chain Reaction
Collapse
Affiliation(s)
- Linda D. Hazlett
- Department of Ophthalmology, Visual and Anatomical Sciences, Wayne State University School of Medicine, Detroit, Michigan, United States
| | - Sandamali A. Ekanayaka
- Department of Ophthalmology, Visual and Anatomical Sciences, Wayne State University School of Medicine, Detroit, Michigan, United States
| | - Sharon A. McClellan
- Department of Ophthalmology, Visual and Anatomical Sciences, Wayne State University School of Medicine, Detroit, Michigan, United States
| | - Rebecca Francis
- Department of Ophthalmology, Visual and Anatomical Sciences, Wayne State University School of Medicine, Detroit, Michigan, United States
| |
Collapse
|
14
|
Xu L, Xu Y, He L, Zhang M, Wang L, Li Z, Li X. Immunomodulatory effects of chicken egg yolk antibodies (IgY) against experimental Shewanella marisflavi AP629 infections in sea cucumbers (Apostichopus japonicus). FISH & SHELLFISH IMMUNOLOGY 2019; 84:108-119. [PMID: 30266604 DOI: 10.1016/j.fsi.2018.09.073] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/14/2018] [Revised: 08/17/2018] [Accepted: 09/25/2018] [Indexed: 06/08/2023]
Abstract
Skin ulceration syndrome in sea cucumbers is an infectious bacterial disease with fast and high mortality. This study investigated the protection of chicken egg yolk antibodies (IgY) on skin ulcer syndrome in sea cucumbers induced by intraperitoneally injecting Shewanella marisflavi AP629. Inactivated whole S. marisflavi AP629 cells were used as an immunogen to immunize laying hens. The highest titer of the obtained specific IgY by ELISA was 1:90000. Specific IgY significantly inhibited the growth of S. marisflavi AP629 in a liquid medium, dose-dependent manner at concentrations ranging from 0.5 to 2 mg/mL. Results obtained from scanning electron microscopy and confocal laser scanning microscopy showed that specific IgY could make bacteria agglutinate and damage the cell membrane of S. marisflavi AP629, resulting in a decrease of bacterial viability. Sea cucumbers treated with 25, 5, and 1 mg/mL anti-S. marisflavi AP629 IgY could achieve survival rates of 77.5%, 50%, and 22.5% at day 12 when the infection and injection therapy were carried out at the same time, respectively. However, survival rates of sea cucumbers treated with 25 mg/mL of nonspecific IgY were only 7.5% at day 12. All sea cucumbers in the positive control group died within twelve days after bacterial inoculation. Levels of the five humoral immune factors (LYZ, ACP, NOS, SOD, CAT) released by coelomocytes were significantly increased in the specific IgY group compared to the nonspecific IgY and positive control groups within 12 h. However, the activities of LYZ, ACP, and SOD decreased rapidly at the 48 h time point in the specific IgY group, indicating that specific IgY treatment could shorten the time needed to restore balance in sea cucumber immune systems. Oral prophylaxis with egg yolk powders was that all sea cucumbers were challenged with 4.2 × 106 CFU S. marisflavi AP629 by intraperitoneal injection after 60 days of feeding. Survival rates of diets containing 10%, 5%, and 1% specific egg yolk powder were 57.5%, 52.5%, and 30% by day 12, respectively, and the survival rate was 27.5% for the nonspecific group and 22.5% for the positive control group. After feeding for 60 days, enzyme activities of LZY, NOS, and SOD were all significantly enhanced in sea cucumbers fed with specific egg yolk powder when compared to the control group (p < 0.05). This study demonstrated that the phagocytic activities of coelomocytes were significantly stimulated after specific IgY treatment over that of nonspecific IgY or without IgY treatments in sea cucumbers (p < 0.05). Overall, our results revealed that anti-S. marisflavi AP629 IgY has a positive immunomodulatory effect on sea cucumbers infected with S. marisflavi AP629.
Collapse
Affiliation(s)
- Le Xu
- School of Life Science and Biotechnology, Dalian University of Technology, Dalian, 116024, China
| | - Yongping Xu
- School of Life Science and Biotechnology, Dalian University of Technology, Dalian, 116024, China; Center for Food Safety of Animal Origin, Ministry of Education, Dalian University of Technology, Dalian, 116600, China
| | - Liangyu He
- School of Life Science and Biotechnology, Dalian University of Technology, Dalian, 116024, China
| | - Meixia Zhang
- School of Life Science and Biotechnology, Dalian University of Technology, Dalian, 116024, China
| | - Lili Wang
- School of Life Science and Biotechnology, Dalian University of Technology, Dalian, 116024, China
| | - Zhen Li
- School of Life Science and Biotechnology, Dalian University of Technology, Dalian, 116024, China
| | - Xiaoyu Li
- School of Life Science and Biotechnology, Dalian University of Technology, Dalian, 116024, China; Center for Food Safety of Animal Origin, Ministry of Education, Dalian University of Technology, Dalian, 116600, China.
| |
Collapse
|
15
|
Ekanayaka SA, McClellan SA, Peng X, Barrett RP, Francis R, Hazlett LD. HMGB1 Antagonist, Box A, Reduces TLR4, RAGE, and Inflammatory Cytokines in the Cornea of P. aeruginosa-Infected Mice. J Ocul Pharmacol Ther 2018; 34:659-669. [PMID: 30407111 PMCID: PMC6302910 DOI: 10.1089/jop.2018.0073] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2018] [Accepted: 08/20/2018] [Indexed: 12/12/2022] Open
Abstract
PURPOSE High mobility group box 1 (HMGB1) contributes to adverse disease outcome in Pseudomonas aeruginosa keratitis. This study tests Box A, an HMGB1 antagonist, in a model of the disease. METHODS C57BL/6 mice (B6) were injected subconjunctivally (1 day before infection) with Box A or phosphate-buffered saline (PBS), infected with P. aeruginosa strain ATCC 19660, and injected intraperitoneally with Box A or PBS at 1 and 3 days postinfection (p.i.). Clinical scores, photographs with a slit lamp camera, real-time polymerase chain reaction (RT-PCR), western blot, immunohistochemistry, enzyme-linked immunosorbent assay (ELISA), myeloperoxidase (MPO), and bacterial plate count were used to assess disease outcome. In separate experiments, the therapeutic potential of Box A was tested as described above, but with treatment begun at 6 h p.i. RESULTS Box A versus PBS prophylactic treatment significantly reduced clinical scores, MPO activity, bacterial load, and expression of TLR4, RAGE, IL-1β, CXCL2, and TNF-α in the infected cornea. Box A blocked co-localization of HMGB1/TLR4 in infiltrated cells in the stroma at 3 and 5 days p.i., but only at 5 days p.i. for HMGB1/RAGE. Box A versus PBS therapeutic treatment significantly reduced clinical scores, MPO activity, bacterial load, and protein levels of IL-1β, CXCL2, and IL-6 in the infected cornea. CONCLUSION Overall, Box A lessens the severity of Pseudomonas keratitis in mice by decreasing expression of TLR4, RAGE (their interaction with HMGB1), IL-1β, CXCL2 (decreasing neutrophil infiltrate), and bacterial plate count when given prophylactically. Therapeutic treatment was not as effective at reducing opacity (disease), but shared similar features with pretreatment of the mice.
Collapse
Affiliation(s)
- Sandamali A. Ekanayaka
- Department of Ophthalmology, Visual and Anatomical Sciences, Wayne State University School of Medicine, Detroit, Michigan
| | - Sharon A. McClellan
- Department of Ophthalmology, Visual and Anatomical Sciences, Wayne State University School of Medicine, Detroit, Michigan
| | - Xudong Peng
- Department of Ophthalmology, The Affiliated Hospital of Qingdao University, Qingdao, China
| | - Ronald P. Barrett
- Department of Ophthalmology, Visual and Anatomical Sciences, Wayne State University School of Medicine, Detroit, Michigan
| | - Rebecca Francis
- Department of Ophthalmology, Visual and Anatomical Sciences, Wayne State University School of Medicine, Detroit, Michigan
| | - Linda D. Hazlett
- Department of Ophthalmology, Visual and Anatomical Sciences, Wayne State University School of Medicine, Detroit, Michigan
| |
Collapse
|