1
|
Park I, Nam H, Ravichandran S, Wall EH, Lillehoj HS. Phytochemicals act holistically to enhance host defenses during poultry coccidiosis. Poult Sci 2025; 104:105042. [PMID: 40120241 PMCID: PMC11987653 DOI: 10.1016/j.psj.2025.105042] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2025] [Revised: 03/12/2025] [Accepted: 03/15/2025] [Indexed: 03/25/2025] Open
Abstract
This study was conducted to investigate the effects of a phytochemical mixture containing full spectrum cinnamon, clove, and oregano essential oils (CCO) on the growth performance, intestinal immunity, and intestinal integrity of broilers infected with coccidiosis. In chicken macrophage cells (CMCs), inflammation was induced with 1.0 µg/mL LPS, followed by stimulating with CCO at three concentrations (1.0, 10.0, and 100 µg/mL) and measuring the gene expression levels of IL-1β and IL-8. In chicken intestinal epithelial cells (IECs), CCO was added and cultured, and the gene expression levels of occludin, ZO-1, and MUC2 were measured. In the in vivo experiment, one hundred and twenty male broiler chickens (0-day-old) were allocated into three treatment groups: (1) basal diet without infection (NC), (2) basal diet with E. maxima infection (PC), and (3) CCO at 4.5 mg/kg feed with E. maxima infection (CCO). Body weight (BW) was measured on days 0, 7, 14, 20, and 22. PC and CCO groups were orally infected with E. maxima on day 14. Jejunal samples were collected on day 22 to conduct gene expression analysis of cytokines, TJ proteins, and antioxidant enzymes. CCO significantly decreased IL-1β and IL-8 in CMCs and increased ZO-1 and MUC2 in IECs in a dose-dependent manner. In the E. maxima-infected groups, dietary CCO tended to mitigate BW loss due to infection. Upon infection, proinflammatory cytokines were suppressed in the CCO group compared to the PC group. Dietary CCO also increased the expression of occludin and JAM-2 in the jejunum. However, CCO did not reduce the oocyst number in coccidiosis-infected chickens. These results suggest that dietary CCO supplementation may improve intestinal immunity and permeability, helping to reduce productivity losses in E. maxima-infected broilers through gut physiological responses, rather than direct antimicrobial effects. These results show the advantage of using in vitro screening based on host-mediated responses, and not on direct pathogen killing, when exploring new phytochemicals to mitigate disease response to reduce economic losses due to coccidiosis.
Collapse
Affiliation(s)
- Inkyung Park
- Animal Bioscience and Biotechnology Laboratory, United States Department of Agriculture, Beltsville Agricultural Research Center, Beltsville, MD 20705, USA
| | - Hyoyoun Nam
- Animal Bioscience and Biotechnology Laboratory, United States Department of Agriculture, Beltsville Agricultural Research Center, Beltsville, MD 20705, USA
| | | | - Emma H Wall
- Nutreco Exploration, Nutreco Switzerland GmbH, 9320 Arbon, Switzerland
| | - Hyun S Lillehoj
- Animal Bioscience and Biotechnology Laboratory, United States Department of Agriculture, Beltsville Agricultural Research Center, Beltsville, MD 20705, USA.
| |
Collapse
|
2
|
Asayama S, Igarashi T, Abe Y, Iwasaki A, Kubo M, Ikeda A, Akiyama K, Okamoto T, Yagi M, Niki Y, Ando H, Ichihashi M, Mizutani KI. Rosae multiflorae fructus extracts regulate the differentiation and vascular endothelial cell-mediated proliferation of keratinocytes. Biosci Biotechnol Biochem 2025; 89:750-760. [PMID: 39848919 DOI: 10.1093/bbb/zbaf007] [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: 12/31/2024] [Accepted: 01/17/2025] [Indexed: 01/25/2025]
Abstract
Keratinocytes are the primary component of the epidermis, so maintaining the precise balance between proliferation and differentiation is essential for conserving epidermal structure and function. Rosae multiflorae fructus extract (RMFE) has wide application in the cosmetic industry, but the molecular mechanisms underlying beneficial effects on keratinocytes are still not fully understood. In this study, we found that RMFE promoted epidermal differentiation and enhanced the barrier function of normal human epidermal keratinocytes (NHEKs) and three-dimensional epidermis model in culture. In addition, RMFE promoted human umbilical vein endothelial cell (HUVEC) proliferation and angiogenesis, whereas the conditioned medium from RMFE-treated HUVECs further promoted NHEK proliferation and increased wound healing ability. Analysis of constituent bioactivities identified a quercetin derivative as a potential mediator of NHEK and HUVEC responses to RMFE. Taken together, these results suggest that RMFE enhances epidermal functions through both direct effects on keratinocytes and indirect effects mediated by endothelial cells.
Collapse
Affiliation(s)
- Sae Asayama
- Laboratory of Stem Cell Biology, Graduate School of Pharmaceutical Sciences, Kobe Gakuin University, Kobe, Japan
- Laboratory of Biochemistry, Faculty of Pharmaceutical Sciences, Kobe Gakuin University, Kobe, Japan
| | - Tami Igarashi
- Research & Development Headquarters, Rosette Co., Ltd., Tokyo, Japan
| | - Yosihimi Abe
- Laboratory of Stem Cell Biology, Graduate School of Pharmaceutical Sciences, Kobe Gakuin University, Kobe, Japan
| | - Ayaka Iwasaki
- Laboratory of Stem Cell Biology, Graduate School of Pharmaceutical Sciences, Kobe Gakuin University, Kobe, Japan
- Laboratory of Biochemistry, Faculty of Pharmaceutical Sciences, Kobe Gakuin University, Kobe, Japan
| | - Mizuki Kubo
- Laboratory of Stem Cell Biology, Graduate School of Pharmaceutical Sciences, Kobe Gakuin University, Kobe, Japan
- Laboratory of Biochemistry, Faculty of Pharmaceutical Sciences, Kobe Gakuin University, Kobe, Japan
| | - Ayaka Ikeda
- Laboratory of Stem Cell Biology, Graduate School of Pharmaceutical Sciences, Kobe Gakuin University, Kobe, Japan
- Laboratory of Biochemistry, Faculty of Pharmaceutical Sciences, Kobe Gakuin University, Kobe, Japan
| | - Kouki Akiyama
- Laboratory of Stem Cell Biology, Graduate School of Pharmaceutical Sciences, Kobe Gakuin University, Kobe, Japan
- Laboratory of Biochemistry, Faculty of Pharmaceutical Sciences, Kobe Gakuin University, Kobe, Japan
| | - Tadashi Okamoto
- Laboratory of Biochemistry, Faculty of Pharmaceutical Sciences, Kobe Gakuin University, Kobe, Japan
| | - Masayuki Yagi
- Research & Development Headquarters, Rosette Co., Ltd., Tokyo, Japan
| | - Yoko Niki
- Cosmetic Science Laboratory, School of Pharmacy and Pharmaceutical Sciences, Mukogawa Women's University, Nishinomiya, Japan
| | - Hideya Ando
- Department of Bioscience, Okayama University of Science, Okayama, Japan
| | | | - Ken-Ichi Mizutani
- Laboratory of Stem Cell Biology, Graduate School of Pharmaceutical Sciences, Kobe Gakuin University, Kobe, Japan
| |
Collapse
|
3
|
Yaron JR, Pallod S, Grigaitis-Esman N, Singh V, Rhodes S, Patel DM, Ghosh D, Rege K. Histamine receptor agonism differentially induces immune and reparative healing responses in biomaterial-facilitated tissue repair. Biomaterials 2025; 315:122967. [PMID: 39586217 DOI: 10.1016/j.biomaterials.2024.122967] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2024] [Revised: 11/04/2024] [Accepted: 11/14/2024] [Indexed: 11/27/2024]
Abstract
Tissue repair is a highly regulated process involving immune, stromal, vascular, and parenchymal cell responses. Mediators of cellular responses at different phases of the healing process stimulate transitions through the continuum of repair. Histamine is an early mediator of healing, which, in skin, is released by resident cells (e.g., mast cells) after cutaneous injury, and acts to stimulate diverse responses in multiple cell populations. Histamine signaling is regulated by four distinct cell surface G-protein coupled receptors (HRH1-4 in humans, Hrh1-4 in mice) which initiate different downstream signaling cascades upon activation, but the specific effect of each receptor on tissue repair is poorly understood. Here, we systematically investigated the effect of selective histamine receptor agonism in laser-activated sealing and tissue repair of incisional skin wounds in immunocompetent mice. Although all four histamine receptors exhibited wound responsiveness in the epidermis, we find that activation of Hrh1, Hrh2, and Hrh4 stimulate a pro-healing immune response characterized by increased pro-resolution macrophages, reduced pro-inflammatory macrophages, and suppressed neutrophil responses. Further, activation of Hrh1 and Hrh4 stimulate angiogenesis after injury. Lastly, although Hrh1 activation resulted in enhanced epidermal epithelial-to-mesenchymal transition (EMT) in vivo and epithelialization in vitro, activation of Hrh2 suppressed both epidermal EMT and epithelialization. Activation of Hrh3, primarily found on neuronal cells, had no effect on any measure in our study. Selective histamine receptor agonism, specifically of histamine receptors Hrh-1 and 4, is a potential reparative approach to promote the efficacy of biomaterial-mediated repair of tissues, including skin.
Collapse
Affiliation(s)
- Jordan R Yaron
- Center for Biomaterials Innovation and Translation, The Biodesign Institute, Arizona State University, Tempe, AZ, 85287, USA; Chemical Engineering, School for Engineering of Matter, Transport, and Energy, Ira A. Fulton Schools of Engineering, Arizona State University, Tempe, AZ, 85287, USA
| | - Shubham Pallod
- Center for Biomaterials Innovation and Translation, The Biodesign Institute, Arizona State University, Tempe, AZ, 85287, USA; Biological Design Graduate Program, School for Engineering of Matter, Transport, and Energy, Ira A. Fulton Schools of Engineering, Arizona State University, Tempe, AZ, 85287, USA
| | - Nicole Grigaitis-Esman
- Center for Biomaterials Innovation and Translation, The Biodesign Institute, Arizona State University, Tempe, AZ, 85287, USA; Biological Design Graduate Program, School for Engineering of Matter, Transport, and Energy, Ira A. Fulton Schools of Engineering, Arizona State University, Tempe, AZ, 85287, USA
| | - Vanshika Singh
- Center for Biomaterials Innovation and Translation, The Biodesign Institute, Arizona State University, Tempe, AZ, 85287, USA; Biomedical Engineering, School for Biological and Health Systems Engineering, Ira A. Fulton Schools of Engineering, Arizona State University, Tempe, AZ, 85287, USA
| | - Samantha Rhodes
- Center for Biomaterials Innovation and Translation, The Biodesign Institute, Arizona State University, Tempe, AZ, 85287, USA; Biological Design Graduate Program, School for Engineering of Matter, Transport, and Energy, Ira A. Fulton Schools of Engineering, Arizona State University, Tempe, AZ, 85287, USA
| | - Dirghau Manishbhai Patel
- Center for Biomaterials Innovation and Translation, The Biodesign Institute, Arizona State University, Tempe, AZ, 85287, USA; Biomedical Engineering, School for Biological and Health Systems Engineering, Ira A. Fulton Schools of Engineering, Arizona State University, Tempe, AZ, 85287, USA
| | - Deepanjan Ghosh
- Center for Biomaterials Innovation and Translation, The Biodesign Institute, Arizona State University, Tempe, AZ, 85287, USA; Biological Design Graduate Program, School for Engineering of Matter, Transport, and Energy, Ira A. Fulton Schools of Engineering, Arizona State University, Tempe, AZ, 85287, USA
| | - Kaushal Rege
- Center for Biomaterials Innovation and Translation, The Biodesign Institute, Arizona State University, Tempe, AZ, 85287, USA; Chemical Engineering, School for Engineering of Matter, Transport, and Energy, Ira A. Fulton Schools of Engineering, Arizona State University, Tempe, AZ, 85287, USA; Biological Design Graduate Program, School for Engineering of Matter, Transport, and Energy, Ira A. Fulton Schools of Engineering, Arizona State University, Tempe, AZ, 85287, USA.
| |
Collapse
|
4
|
Serra D, Garroni G, Cruciani S, Coradduzza D, Pashchenko A, Amler E, Pintore G, Parisse P, Satta R, Martini F, Tognon M, Brunetti A, Ventura C, Maioli M. PVA and PVP nanofibers combined with Helichrysum italicum oil preserve skin cell interactions, elasticity and proliferation. Sci Rep 2025; 15:10864. [PMID: 40158043 PMCID: PMC11954863 DOI: 10.1038/s41598-025-95788-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2024] [Accepted: 03/24/2025] [Indexed: 04/01/2025] Open
Abstract
Development of electrospun nanofibers with suitable properties to promote wound healing is an advantage in developing non-invasive skin treatments. We showed the potential application of Polyvinyl acetate (PVA) and Polyvinylpyrrolidone (PVP) combined with Helichrysum italicum oil (HO) in wound healing. During this process, Tight junctions (TJs) play a crucial role in maintaining skin integrity. TJs are intercellular junctions composed of a variety of transmembrane proteins, including Occludin (OCLN), observed also in migrating epithelial cells. Changes in OCLN expression affect epidermal permeability, indicating an active role in the healing process. Within this context, we studied the OCLN expression during healing after scratch assay on Keratinocytes (HaCaT), by a confocal microscopic analysis. In addition, we evaluated the effect of treatment after scratch on cell elasticity by Atomic Force Microscopy (AFM) analysis. All results show a positive trend in cell proliferation and viability on HaCaT treated with functionalized nanofibers. These results were confirmed by the expression of genes involved in the early stages of the regenerative process. Understanding the cell mechanisms involved in skin changes during repair process would allow future application of nanomaterials combined with HO in vivo.
Collapse
Affiliation(s)
- Diletta Serra
- Department of Biomedical Sciences, University of Sassari, Viale San Pietro 43/B, 07100, Sassari, Italy
- R&D Laboratory Center, InoCure s.r.o., Politických Veziu 935/13, 110 00, Prague, Czech Republic
| | - Giuseppe Garroni
- Department of Biomedical Sciences, University of Sassari, Viale San Pietro 43/B, 07100, Sassari, Italy
| | - Sara Cruciani
- Department of Biomedical Sciences, University of Sassari, Viale San Pietro 43/B, 07100, Sassari, Italy
| | - Donatella Coradduzza
- Department of Biomedical Sciences, University of Sassari, Viale San Pietro 43/B, 07100, Sassari, Italy
| | - Aleksei Pashchenko
- Department of Biomedical Sciences, University of Sassari, Viale San Pietro 43/B, 07100, Sassari, Italy
- Department of Biophysics, Second Faculty of Medicine, Charles University, V Uvalu 84, 150 06, Prague, Czech Republic
- University Centre for Energy Efficient Buildings, Czech Technical University in Prague, Trinecka 1024, 273 43, Bustehrad, Czech Republic
| | - Evzen Amler
- University Centre for Energy Efficient Buildings, Czech Technical University in Prague, Trinecka 1024, 273 43, Bustehrad, Czech Republic
- Student Science, Národních hrdinů 279, 190 12, Praha 9, Czech Republic
| | - Giorgio Pintore
- Department of Medicine, Surgery and Pharmacy, University of Sassari, 07100, Sassari, Italy
| | - Pietro Parisse
- Institute of Materials (IOM-CNR), Area Science Park, 34149, Basovizza, Trieste, Italy
| | - Rosanna Satta
- Department of Medicine, Surgery and Pharmacy, University of Sassari, 07100, Sassari, Italy
| | - Fernanda Martini
- Department of Medical Sciences, University of Ferrara, Ferrara, Italy
| | - Mauro Tognon
- Department of Medical Sciences, University of Ferrara, Ferrara, Italy
| | - Antonio Brunetti
- Department of Biomedical Sciences, University of Sassari, Viale San Pietro 43/B, 07100, Sassari, Italy
| | - Carlo Ventura
- Laboratory of Molecular Biology and Stem Cell Engineering - Eldor Lab Istituto Nazionale Biostrutture e Biosistemi, Via Di Corticella 183, 40128, Bologna, Italy
| | - Margherita Maioli
- Department of Biomedical Sciences, University of Sassari, Viale San Pietro 43/B, 07100, Sassari, Italy.
- Department of Biomedical Sciences, Center for Developmental Biology and Reprogramming-CEDEBIOR, University of Sassari, Viale San Pietro 43/B, 07100, Sassari, Italy.
| |
Collapse
|
5
|
Chen Y, Gao B, Cai W, Lai J, Lai K, Wang Y. Oral mucosa: anti-inflammatory function, mechanisms, and applications. J Mater Chem B 2025; 13:4059-4072. [PMID: 40062381 DOI: 10.1039/d4tb02845g] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/27/2025]
Abstract
Large soft tissue injuries require several weeks to heal and frequently leave fibrotic scars that can negatively impact tissue function. However, the applicability of traditional skin and mucous membrane transplantation for the treatment of lesions in the ocular surface and urethra is limited owing to the unique locations and functions of these tissues. Oral mucosa has been widely used in the repair of such injuries owing to its reduced propensity for inducing an inflammatory response, angiogenesis, and scarring. Enhancing chronic wound healing while avoiding scar formation requires a broader understanding of the cellular and molecular pathways that drive wound repair in the oral mucosa. This review integrates current knowledge on the mechanisms underlying the resistance of the oral mucosa to inflammation and its application as a graft material, highlighting its challenges and potential advancements. The aim of this review is to offer insights into future therapeutic strategies for wound healing and related conditions.
Collapse
Affiliation(s)
- Yani Chen
- Stomatology Hospital, School of Stomatology, Zhejiang University School of Medicine Zhejiang Provincial Clinical Research Center for Oral Diseases, Key Laboratory of Oral Biomedical Research of Zhejiang Province, Cancer Center of Zhejiang University, Engineering Research Center of Oral Biomaterials and Devices of Zhejiang Province, Hangzhou 310000, P. R. China.
| | - Bicong Gao
- Stomatology Hospital, School of Stomatology, Zhejiang University School of Medicine Zhejiang Provincial Clinical Research Center for Oral Diseases, Key Laboratory of Oral Biomedical Research of Zhejiang Province, Cancer Center of Zhejiang University, Engineering Research Center of Oral Biomaterials and Devices of Zhejiang Province, Hangzhou 310000, P. R. China.
| | - Wenjin Cai
- Stomatology Hospital, School of Stomatology, Zhejiang University School of Medicine Zhejiang Provincial Clinical Research Center for Oral Diseases, Key Laboratory of Oral Biomedical Research of Zhejiang Province, Cancer Center of Zhejiang University, Engineering Research Center of Oral Biomaterials and Devices of Zhejiang Province, Hangzhou 310000, P. R. China.
| | - Junhong Lai
- Stomatology Hospital, School of Stomatology, Zhejiang University School of Medicine Zhejiang Provincial Clinical Research Center for Oral Diseases, Key Laboratory of Oral Biomedical Research of Zhejiang Province, Cancer Center of Zhejiang University, Engineering Research Center of Oral Biomaterials and Devices of Zhejiang Province, Hangzhou 310000, P. R. China.
| | - Kaichen Lai
- Stomatology Hospital, School of Stomatology, Zhejiang University School of Medicine Zhejiang Provincial Clinical Research Center for Oral Diseases, Key Laboratory of Oral Biomedical Research of Zhejiang Province, Cancer Center of Zhejiang University, Engineering Research Center of Oral Biomaterials and Devices of Zhejiang Province, Hangzhou 310000, P. R. China.
| | - Ying Wang
- Stomatology Hospital, School of Stomatology, Zhejiang University School of Medicine Zhejiang Provincial Clinical Research Center for Oral Diseases, Key Laboratory of Oral Biomedical Research of Zhejiang Province, Cancer Center of Zhejiang University, Engineering Research Center of Oral Biomaterials and Devices of Zhejiang Province, Hangzhou 310000, P. R. China.
| |
Collapse
|
6
|
Han C, Yuan H, Chen AK, DiPietro LA, Chen L. Differential Temperature-Induced Responses in Immortalized Oral and Skin Keratinocytes. Int J Mol Sci 2025; 26:2851. [PMID: 40243437 PMCID: PMC11988828 DOI: 10.3390/ijms26072851] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2025] [Revised: 03/17/2025] [Accepted: 03/18/2025] [Indexed: 04/18/2025] Open
Abstract
The epidermis of the skin and oral mucosa is constantly exposed to various environmental stimuli, including temperature changes. In particularly extreme conditions, such as excess heat or cold, significant injury may occur. Oral and skin keratinocytes exhibit tissue-specific differences in wound healing outcomes and the transcriptomic response to injury. This study investigated if skin and oral keratinocytes also have differential responses to heat- and cold-induced injury. Oral keratinocytes (TIGKs) were found to exhibit an enhanced viability following heat-induced injury compared to skin keratinocytes (HaCaTs). However, there were no discernible differences between skin and oral keratinocyte viability following cold-induced injury. To examine the transcriptomic differences between skin and oral keratinocytes in response to temperature-induced injury, we generated an mRNA-sequencing gene expression dataset. Differentially expressed genes (DEGs) including heat shock proteins (HSPs) were identified between HaCaTs and TIGKs at baseline (37 °C) and after heat- (60 °C) or cold-induced (-25 °C) injury. Our comparative analyses suggest that skin and oral keratinocytes exhibit transcriptomic differences at baseline and in their responses to heat or cold exposure. The enhanced heat tolerance of TIGKs relative to HaCaTs may be due to an advantageous expression of a subset of HSPs at baseline in TIGKs. Our work also provides a source of skin and oral keratinocyte gene expression data following heat- and cold-induced injury that can be used for future analyses.
Collapse
Affiliation(s)
| | | | | | | | - Lin Chen
- Center for Wound Healing and Tissue Regeneration, University of Illinois Chicago, Chicago, IL 60612, USA; (C.H.); (H.Y.); (A.K.C.); (L.A.D.)
| |
Collapse
|
7
|
Park I, Nam H, Ravichandran S, Wall EH, Lillehoj HS. Molecular responses to clove and oregano essential oils are associated with reduced inflammation and improved gut barrier function in broiler chickens. Poult Sci 2025; 104:104713. [PMID: 39721262 PMCID: PMC11732532 DOI: 10.1016/j.psj.2024.104713] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2024] [Revised: 12/17/2024] [Accepted: 12/19/2024] [Indexed: 12/28/2024] Open
Abstract
In vitro tests were conducted to characterize the host-mediated responses of chickens to Clove Essential Oil (CEO) and Oregano Essential Oil (OEO). Chicken macrophage cells (CMCs), chicken intestinal epithelial cells (IECs), quail muscle cells (QMCs), and chicken embryonic muscle cells (EMCs) were utilized in these assays. EMCs were collected from the 13-day-old embryo during egg incubation and all cell lines were seeded at 2 × 105/mL in a 24-well plate. In CMCs, an inflammatory response was induced by stimulating with 1.0 µg/mL of Lipopolysaccharide (LPS). To induce muscle cell differentiation, 0.5 % FBS was used in QMCs and 2.0 % FBS in EMCs. Three different concentrations (1.0, 10.0, and 100 µg/mL) of CEO and OEO were administered. qRT-PCR was used to measure gene expression levels of IL-1β and IL-8 from CMCs, occludin, ZO-1, and MUC2 from IECs, and Pax7 and MyoG from QMCs and EMCs. Cytotoxic effects of CEO and OEO were determined using an MTT assay; CEO and OEO did not show cytotoxicity at concentrations below 0.1 mg/mL in CMCs, IECs, QMCs, and EMCs. CEO reduced (P < 0.05) the LPS-induced increase of IL-1β and IL-8 in CMCs and increased (P < 0.05) ZO-1 and MUC2 in IECs. OEO suppressed (P < 0.05) the release of IL-8, increased ZO-1, and Pax7. Both CEO and OEO demonstrated microbicidal activity against sporozoite of E. tenella and C. perfringens bacteria, but only at doses 10-100 × higher than those that would be used in feed. These findings support our previous findings on other phytochemicals; both CEO and OEO are promising candidates for improved resilience in chickens not due to their direct antimicrobial effects, but due to gut physiological responses that take place at the level of the host.
Collapse
Affiliation(s)
- Inkyung Park
- Animal Bioscience and Biotechnology Laboratory United States Department of Agriculture-Agricultural Research Service, Beltsville Agricultural Research Center, Beltsville, MD 20705, USA
| | - Hyoyoun Nam
- Animal Bioscience and Biotechnology Laboratory United States Department of Agriculture-Agricultural Research Service, Beltsville Agricultural Research Center, Beltsville, MD 20705, USA
| | | | - Emma H Wall
- Nutreco Exploration, Nutreco, the Netherlands
| | - Hyun S Lillehoj
- Animal Bioscience and Biotechnology Laboratory United States Department of Agriculture-Agricultural Research Service, Beltsville Agricultural Research Center, Beltsville, MD 20705, USA.
| |
Collapse
|
8
|
Ren X, Li K, Fang X, Zhu Z, Chen Q, Li C, Hua H. Oral mucosal changes in tight junction proteins in patients with oral lichen planus. Oral Dis 2024; 30:4367-4375. [PMID: 38439057 DOI: 10.1111/odi.14912] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2023] [Revised: 01/30/2024] [Accepted: 02/19/2024] [Indexed: 03/06/2024]
Abstract
OBJECTIVE This study aimed to investigate the expression of tight junction, its distribution pattern in oral lichen planus samples and its potential association with the severity of oral lichen planus. MATERIALS AND METHODS Cross-sectional study designs were conducted. Transcriptome sequencing was conducted using oral mucosal tissues from 22 patients with oral lichen planus and 11 healthy controls. Immunohistochemistry and quantitative reverse transcription PCR were performed to verify the expression of claudin-1, claudin-4, occludin and zonula occludens-1 in oral mucosal tissues from another 30 patients with oral lichen planus and 26 healthy controls. The relationship between tight junction protein expression and oral lichen planus severity was explored using correlation analysis. RESULTS 5603 and 2475 differentially expressed genes were upregulated and downregulated respectively, in oral lichen planus tissues. KEGG analysis showed that tight junctions including CLDN1, CLDN4, OCLN and TJP1 were downregulated in oral lichen planus. Claudin-1, claudin-4, occludin and zonula occludens-1 expression was verified to be significantly lower in oral lichen planus. Furthermore, correlation analyses showed that decreased occludin expression was positively related to oral lichen planus severity. CONCLUSION Decreased expression of TJ barrier proteins may be associated with the development of oral lichen planus.
Collapse
Affiliation(s)
- Xiaomeng Ren
- Department of Oral Medicine, Peking University School and Hospital of Stomatology, National Center of Stomatology, National Clinical Research Center for Oral Diseases, National Engineering Research Center of Oral Biomaterials and Digital Medical Devices, Beijing, China
| | - Kaiyi Li
- Department of Oral Medicine, Peking University School and Hospital of Stomatology, National Center of Stomatology, National Clinical Research Center for Oral Diseases, National Engineering Research Center of Oral Biomaterials and Digital Medical Devices, Beijing, China
- Department of Oral Mucosa, Shanghai Stomatological Hospital, Shanghai Key Laboratory of Craniomaxillofacial Development and Diseases, Fudan University, Shanghai, China
| | - Xin Fang
- Department of Oral Medicine, Peking University School and Hospital of Stomatology, National Center of Stomatology, National Clinical Research Center for Oral Diseases, National Engineering Research Center of Oral Biomaterials and Digital Medical Devices, Beijing, China
- Department of Oral Medicine, Yunnan Key Laboratory of Stomatology, Kunming Medical University School and Hospital of Stomatology, Kunming, China
| | - Zhengda Zhu
- Department of Oral Medicine, Peking University School and Hospital of Stomatology, National Center of Stomatology, National Clinical Research Center for Oral Diseases, National Engineering Research Center of Oral Biomaterials and Digital Medical Devices, Beijing, China
- Department of Oral Medicine, Capital Medical University Affiliated Beijing Friendship Hospital, Beijing, China
| | - Qianming Chen
- Department of Oral Medicine, West China School of Stomatology, Sichuan University, Chengdu, Sichuan, China
| | - Chunlei Li
- Department of Oral Medicine, Peking University School and Hospital of Stomatology, National Center of Stomatology, National Clinical Research Center for Oral Diseases, National Engineering Research Center of Oral Biomaterials and Digital Medical Devices, Beijing, China
| | - Hong Hua
- Department of Oral Medicine, Peking University School and Hospital of Stomatology, National Center of Stomatology, National Clinical Research Center for Oral Diseases, National Engineering Research Center of Oral Biomaterials and Digital Medical Devices, Beijing, China
| |
Collapse
|
9
|
Rui S, Dai L, Zhang X, He M, Xu F, Wu W, Armstrong DG, You Y, Xiao X, Ma Y, Chen Y, Deng W. Exosomal miRNA-26b-5p from PRP suppresses NETs by targeting MMP-8 to promote diabetic wound healing. J Control Release 2024; 372:221-233. [PMID: 38909697 DOI: 10.1016/j.jconrel.2024.06.050] [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: 03/25/2024] [Revised: 05/28/2024] [Accepted: 06/20/2024] [Indexed: 06/25/2024]
Abstract
The utilization of platelet-rich plasma (PRP) has exhibited potential as a therapeutic approach for the management of diabetic foot ulcers (DFUs). However, it is currently not well understood how the diabetic environment may influence PRP-derived exosomes (PRP-Exos) and their potential impact on neutrophil extracellular traps (NETs). This study aims to investigate the effects of the diabetic environment on PRP-Exos, their communication with neutrophils, and the subsequent influence on NETs and wound healing. Through bulk-seq and Western blotting, we confirmed the increased expression of MMP-8 in DFUs. Additionally, we discovered that miRNA-26b-5p plays a significant role in the communication between DFUs and PRP-Exos. In our experiments, we found that PRP-Exos miR-26b-5p effectively improved diabetic wound healing by inhibiting NETs. Further tests validated the inhibitory effect of miR-26b-5p on NETs by targeting MMP-8. Both in vitro and in vivo experiments showed that miRNA-26b-5p from PRP-Exos promoted wound healing by reducing neutrophil infiltration through its targeting of MMP-8. This study establishes the importance of miR-26b-5p in the communication between DFUs and PRP-Exos, disrupting NETs formation in diabetic wounds by targeting MMP-8. These findings provide valuable insights for developing novel therapeutic strategies to enhance wound healing in individuals suffering from DFUs.
Collapse
Affiliation(s)
- Shunli Rui
- Department of Endocrinology and Metabolism, School of Medicine, Chongqing University Central Hospital, Chongqing Emergency Medical Centre, Chongqing 400014, China
| | - Linrui Dai
- Department of Endocrinology and Metabolism, School of Medicine, Chongqing University Central Hospital, Chongqing Emergency Medical Centre, Chongqing 400014, China
| | - Xiaoshi Zhang
- Department of Endocrinology and Metabolism, School of Medicine, Chongqing University Central Hospital, Chongqing Emergency Medical Centre, Chongqing 400014, China
| | - Min He
- Department of Endocrinology and Metabolism, School of Medicine, Chongqing University Central Hospital, Chongqing Emergency Medical Centre, Chongqing 400014, China
| | - Fan Xu
- Department of Endocrinology and Metabolism, School of Medicine, Chongqing University Central Hospital, Chongqing Emergency Medical Centre, Chongqing 400014, China
| | - Wei Wu
- Key Laboratory of Biorheological Science and Technology, Ministry of Education, College of Bioengineering, Chongqing University, Chongqing 400044, China
| | - David G Armstrong
- Department of Surgery, Keck School of Medicine of University of Southern California, Los Angeles, CA 90033, USA
| | - Yuehua You
- The Chongqing Key Laboratory of Translational Medicine in Major Metabolic Diseases, the First Affiliated Hospital of Chongqing Medical University, Chongqing 400010, China
| | - Xiaoqiu Xiao
- The Chongqing Key Laboratory of Translational Medicine in Major Metabolic Diseases, the First Affiliated Hospital of Chongqing Medical University, Chongqing 400010, China
| | - Yu Ma
- Department of Endocrinology and Metabolism, School of Medicine, Chongqing University Central Hospital, Chongqing Emergency Medical Centre, Chongqing 400014, China.
| | - Yan Chen
- Department of Endocrinology and Metabolism, School of Medicine, Chongqing University Central Hospital, Chongqing Emergency Medical Centre, Chongqing 400014, China.
| | - Wuquan Deng
- Department of Endocrinology and Metabolism, School of Medicine, Chongqing University Central Hospital, Chongqing Emergency Medical Centre, Chongqing 400014, China.
| |
Collapse
|
10
|
Meng Y, Liu Y, Guo J, Guo X, Han X, Zhang L, Di T, Zhao J, Wang Y, Li P. Qing-Re-Chu-shi decoction ameliorates 2,4-dinitrochlorobenzene-induced atopic dermatitis in NC/Nga mice through anti-inflammation and immunoregulatory mechanisms. JOURNAL OF ETHNOPHARMACOLOGY 2024; 323:117702. [PMID: 38176665 DOI: 10.1016/j.jep.2024.117702] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/09/2023] [Revised: 12/22/2023] [Accepted: 01/02/2024] [Indexed: 01/06/2024]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE Qing-Re-Chu-Shi Decoction (QRCSD), a traditional Chinese herbal formula, has been employed as a complementary and alternative therapy for inflammatory skin diseases. However, its active constituents and the mechanistic basis of its action on atopic dermatitis remain in adequately understood. AIM OF THE STUDY Atopic dermatitis (AD) is an allergic dermatitis marked by eczematous lesions and pruritus. The study aimed to elucidate the underlying effects of QRCSD on AD and to identify the components responsible for its therapeutic efficacy in a mouse model. MATERIALS AND METHODS Network pharmacology and UPLC-mass analysis were used to anticipate the pharmacological mechanisms and to identify active components of QRCSD, respectively. A DNCB-induced AD-like model was established in NC/Nga mice. QRCSD or prednisolone (as a positive control) was administered via gavage every other day from day14 to day 21. Dermatitis severity score, scratching behavior, skin barrier function, spleen index, Th1/Th2 lymphocyte ratio, and serum IgE levels were evaluated. Protein arrays, including 40 inflammatory cytokines, were performed on skin lesions, followed by confirmation experiments of Western blotting in dorsal skin lesions. RESULTS The construction of a QRCSD-AD-Network and topological analysis firstly proposed potential targets of QRCSD acting on AD. Animal experiments demonstrated that oral administration of QRCSD ameliorated AD-like lesions, reduced epidermal thickness and mast cell count, decreased serum IgE levels, augmented tight junction protein (Claudin 1, Occludin) levels, and regulated the Th1/Th2 balance in the spleen, as well as spleen index. Elevated levels of interleukin (IL)-4, IL-5, IL-6, IL-17, and Eotaxin were revealed in AD-like skin lesions by protein arrays. Western blotting confirmed that the phosphorylation levels of ERK, P38, JNK, STAT3 and P65 were downregulated, and IL-6 expression was also reduced following QRCSD treatment. CONCLUSIONS The study enhances the understanding of the anti-inflammatory and immunomodulatory effects of QRCSD, showcasing its significant protective role against atopic dermatitis. Treatment with QRCSD may be considered as a viable candidate for complementary and alternative therapy in managing atopic dermatitis.
Collapse
Affiliation(s)
- YuJiao Meng
- Beijing Hospital of Traditional Chinese Medicine, Capital Medical University, Beijing Institute of Traditional Chinese Medicine, Beijing Key Laboratory of Clinic and Basic Research with Traditional Chinese Medicine on Psoriasis (BZ0375), Beijing, 100010, China; Beijing University of Chinese Medicine, Beijing, 100105, China
| | - Yu Liu
- Beijing Hospital of Traditional Chinese Medicine, Capital Medical University, Beijing Institute of Traditional Chinese Medicine, Beijing Key Laboratory of Clinic and Basic Research with Traditional Chinese Medicine on Psoriasis (BZ0375), Beijing, 100010, China; Beijing University of Chinese Medicine, Beijing, 100105, China
| | - Jianning Guo
- Beijing Hospital of Traditional Chinese Medicine, Capital Medical University, Beijing Institute of Traditional Chinese Medicine, Beijing Key Laboratory of Clinic and Basic Research with Traditional Chinese Medicine on Psoriasis (BZ0375), Beijing, 100010, China; Beijing University of Chinese Medicine, Beijing, 100105, China
| | - Xiaoyao Guo
- Beijing Hospital of Traditional Chinese Medicine, Capital Medical University, Beijing Institute of Traditional Chinese Medicine, Beijing Key Laboratory of Clinic and Basic Research with Traditional Chinese Medicine on Psoriasis (BZ0375), Beijing, 100010, China; Beijing University of Chinese Medicine, Beijing, 100105, China
| | - Xuyang Han
- Beijing Hospital of Traditional Chinese Medicine, Capital Medical University, Beijing Institute of Traditional Chinese Medicine, Beijing Key Laboratory of Clinic and Basic Research with Traditional Chinese Medicine on Psoriasis (BZ0375), Beijing, 100010, China
| | - Lu Zhang
- Beijing Hospital of Traditional Chinese Medicine, Capital Medical University, Beijing Institute of Traditional Chinese Medicine, Beijing Key Laboratory of Clinic and Basic Research with Traditional Chinese Medicine on Psoriasis (BZ0375), Beijing, 100010, China
| | - Tingting Di
- Beijing Hospital of Traditional Chinese Medicine, Capital Medical University, Beijing Institute of Traditional Chinese Medicine, Beijing Key Laboratory of Clinic and Basic Research with Traditional Chinese Medicine on Psoriasis (BZ0375), Beijing, 100010, China
| | - Jingxia Zhao
- Beijing Hospital of Traditional Chinese Medicine, Capital Medical University, Beijing Institute of Traditional Chinese Medicine, Beijing Key Laboratory of Clinic and Basic Research with Traditional Chinese Medicine on Psoriasis (BZ0375), Beijing, 100010, China
| | - Yan Wang
- Beijing Hospital of Traditional Chinese Medicine, Capital Medical University, Beijing Institute of Traditional Chinese Medicine, Beijing Key Laboratory of Clinic and Basic Research with Traditional Chinese Medicine on Psoriasis (BZ0375), Beijing, 100010, China
| | - Ping Li
- Beijing Hospital of Traditional Chinese Medicine, Capital Medical University, Beijing Institute of Traditional Chinese Medicine, Beijing Key Laboratory of Clinic and Basic Research with Traditional Chinese Medicine on Psoriasis (BZ0375), Beijing, 100010, China; Beijing University of Chinese Medicine, Beijing, 100105, China.
| |
Collapse
|
11
|
Bui CM, Vuong HG, Le MK, Rybski KJ, Zengin HB, Tang H, Smoller BR. Claudin-4 Upregulation in Acantholytic and Autoimmune-Mediated Bullous Disorders. Dermatopathology (Basel) 2023; 11:1-7. [PMID: 38534263 DOI: 10.3390/dermatopathology11010001] [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: 09/19/2023] [Revised: 12/09/2023] [Accepted: 12/14/2023] [Indexed: 03/28/2024] Open
Abstract
Claudin-4 is a key component of tight junctions, which play an important role in the formation of the epidermal barrier by forming a circumferential network in the granular layer that serves as a gatekeeper of the paracellular pathway. The aim of this study is to illustrate claudin-4 immunohistochemical staining patterns of different blistering disorders. We collected 35 cases, including two Hailey-Hailey disease, one Darier disease, three Grover disease, one acantholytic acanthoma, two warty dyskeratoma, 11 pemphigus vulgaris (PV) including six mucosal PV, and two pemphigus foliaceus. For comparison, we included five cases of normal skin, five eczema, and three bullous pemphigoid cases. Claudin-4 demonstrated weak-to-moderate expression in keratinocytes located in the stratum granulosum, keratinocytes surrounding hair follicles, and adnexal glands. Further, claudin-4 exhibited moderate-to-strong membranous staining in disrupted keratinocytes surrounding and within the acantholytic and bullous areas in 16/22 of the acantholytic cases (not seen in the six cases of mucosal PV) and all three bullous pemphigoids. This finding suggests that claudin-4 is upregulated in these conditions, which may be a compensatory response to the disrupted barrier function. This finding could shed light on the molecular mechanisms underlying disrupted barrier function in blistering disorders, independent of the specific underlying disease mechanism.
Collapse
Affiliation(s)
- Chau M Bui
- Department of Pathology and Laboratory Medicine, University of Rochester, Rochester, NY 14642, USA
| | - Huy G Vuong
- Department of Pathology, University of Iowa, Iowa City, IA 52242, USA
| | - Minh-Khang Le
- Department of Pathology, University of Yamanashi, Yamanashi 409-3898, Japan
| | - Kristin J Rybski
- Department of Pathology and Laboratory Medicine, University of Rochester, Rochester, NY 14642, USA
| | - Hatice B Zengin
- Department of Pathology and Laboratory Medicine, University of Rochester, Rochester, NY 14642, USA
| | - Haiming Tang
- Department of Pathology, Yale School of Medicine, New Haven, CT 06510, USA
| | - Bruce R Smoller
- Department of Pathology and Laboratory Medicine, University of Rochester, Rochester, NY 14642, USA
| |
Collapse
|
12
|
Ren L, Jiang Z, Zhang H, Chen Y, Zhu D, He J, Chen Y, Wang Y, Yang G. Biomaterials derived from hard palate mucosa for tissue engineering and regenerative medicine. Mater Today Bio 2023; 22:100734. [PMID: 37636987 PMCID: PMC10458294 DOI: 10.1016/j.mtbio.2023.100734] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2023] [Revised: 07/07/2023] [Accepted: 07/19/2023] [Indexed: 08/29/2023] Open
Abstract
Autologous materials have superior biosafety and are widely used in clinical practice. Due to its excellent trauma-healing ability, the hard palate mucosa (HPM) has become a hot spot for autologous donor area research. Multiple studies have conducted an in-depth analysis of the healing ability of the HPM at the cellular and molecular levels. In addition, the HPM has good maneuverability as a donor area for soft tissue grafts, and researchers have isolated various specific mesenchymal stem cells (MSCs) from HPM. Free soft tissue grafts obtained from the HPM have been widely used in the clinic and have played an essential role in dentistry, eyelid reconstruction, and the repair of other specific soft tissue defects. This article reviews the advantages of HPM as a donor area and its related mechanisms, classes of HPM-derived biomaterials, the current status of clinical applications, challenges, and future development directions.
Collapse
Affiliation(s)
- Lingfei Ren
- Stomatology Hospital, School of Stomatology, Zhejiang University School of Medicine, Zhejiang Provincial Clinical Research Center for Oral Diseases, Key Laboratory of Oral Biomedical Research of Zhejiang Province, Cancer Center of Zhejiang University, Engineering Research Center of Oral Biomaterials and Devices of Zhejiang Province, Hangzhou, 310000, China
| | - Zhiwei Jiang
- Stomatology Hospital, School of Stomatology, Zhejiang University School of Medicine, Zhejiang Provincial Clinical Research Center for Oral Diseases, Key Laboratory of Oral Biomedical Research of Zhejiang Province, Cancer Center of Zhejiang University, Engineering Research Center of Oral Biomaterials and Devices of Zhejiang Province, Hangzhou, 310000, China
| | - Hui Zhang
- Stomatology Hospital, School of Stomatology, Zhejiang University School of Medicine, Zhejiang Provincial Clinical Research Center for Oral Diseases, Key Laboratory of Oral Biomedical Research of Zhejiang Province, Cancer Center of Zhejiang University, Engineering Research Center of Oral Biomaterials and Devices of Zhejiang Province, Hangzhou, 310000, China
| | - Yani Chen
- Stomatology Hospital, School of Stomatology, Zhejiang University School of Medicine, Zhejiang Provincial Clinical Research Center for Oral Diseases, Key Laboratory of Oral Biomedical Research of Zhejiang Province, Cancer Center of Zhejiang University, Engineering Research Center of Oral Biomaterials and Devices of Zhejiang Province, Hangzhou, 310000, China
| | - Danji Zhu
- Stomatology Hospital, School of Stomatology, Zhejiang University School of Medicine, Zhejiang Provincial Clinical Research Center for Oral Diseases, Key Laboratory of Oral Biomedical Research of Zhejiang Province, Cancer Center of Zhejiang University, Engineering Research Center of Oral Biomaterials and Devices of Zhejiang Province, Hangzhou, 310000, China
| | - Jin He
- Stomatology Hospital, School of Stomatology, Zhejiang University School of Medicine, Zhejiang Provincial Clinical Research Center for Oral Diseases, Key Laboratory of Oral Biomedical Research of Zhejiang Province, Cancer Center of Zhejiang University, Engineering Research Center of Oral Biomaterials and Devices of Zhejiang Province, Hangzhou, 310000, China
| | - Yunxuan Chen
- Stomatology Hospital, School of Stomatology, Zhejiang University School of Medicine, Zhejiang Provincial Clinical Research Center for Oral Diseases, Key Laboratory of Oral Biomedical Research of Zhejiang Province, Cancer Center of Zhejiang University, Engineering Research Center of Oral Biomaterials and Devices of Zhejiang Province, Hangzhou, 310000, China
| | - Ying Wang
- Stomatology Hospital, School of Stomatology, Zhejiang University School of Medicine, Zhejiang Provincial Clinical Research Center for Oral Diseases, Key Laboratory of Oral Biomedical Research of Zhejiang Province, Cancer Center of Zhejiang University, Engineering Research Center of Oral Biomaterials and Devices of Zhejiang Province, Hangzhou, 310000, China
| | - Guoli Yang
- Stomatology Hospital, School of Stomatology, Zhejiang University School of Medicine, Zhejiang Provincial Clinical Research Center for Oral Diseases, Key Laboratory of Oral Biomedical Research of Zhejiang Province, Cancer Center of Zhejiang University, Engineering Research Center of Oral Biomaterials and Devices of Zhejiang Province, Hangzhou, 310000, China
| |
Collapse
|
13
|
Ha P, Liu TP, Li C, Zheng Z. Novel Strategies for Orofacial Soft Tissue Regeneration. Adv Wound Care (New Rochelle) 2023; 12:339-360. [PMID: 35651274 DOI: 10.1089/wound.2022.0037] [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] [Indexed: 11/12/2022] Open
Abstract
Significance: Orofacial structures are indispensable for speech and eating, and impairment disrupts whole-body health through malnutrition and poor quality of life. However, due to the unique and highly specialized cell populations, tissue architecture, and healing microenvironments, regeneration in this region is challenging and inadequately addressed to date. Recent Advances: With increasing understanding of the nuanced physiology and cellular responses of orofacial soft tissue, novel scaffolds, seeded cells, and bioactive molecules were developed in the past 5 years to specifically target orofacial soft tissue regeneration, particularly for tissues primarily found within the orofacial region such as oral mucosa, taste buds, salivary glands, and masseter muscles. Critical Issues: Due to the tightly packed and complex anatomy, orofacial soft tissue injury commonly implicates multiple tissue types, and thus functional unit reconstruction in the orofacial region is more important than single tissue regeneration. Future Directions: This article reviews the up-to-date knowledge in this highly translational topic, which provides insights into novel biologically inspired and engineered strategies for regenerating orofacial component tissues and functional units.
Collapse
Affiliation(s)
- Pin Ha
- David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, California, USA
| | - Timothy P Liu
- David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, California, USA
| | - Chenshuang Li
- Department of Orthodontics, School of Dental Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Zhong Zheng
- David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, California, USA
- School of Dentistry, University of California, Los Angeles, Los Angeles, California, USA
| |
Collapse
|
14
|
Lee EJ, Kim Y, Salipante P, Kotula AP, Lipshutz S, Graves DT, Alimperti S. Mechanical Regulation of Oral Epithelial Barrier Function. Bioengineering (Basel) 2023; 10:bioengineering10050517. [PMID: 37237587 DOI: 10.3390/bioengineering10050517] [Citation(s) in RCA: 18] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2023] [Revised: 04/18/2023] [Accepted: 04/21/2023] [Indexed: 05/28/2023] Open
Abstract
Epithelial cell function is modulated by mechanical forces imparted by the extracellular environment. The transmission of forces onto the cytoskeleton by modalities such as mechanical stress and matrix stiffness is necessary to address by the development of new experimental models that permit finely tuned cell mechanical challenges. Herein, we developed an epithelial tissue culture model, named the 3D Oral Epi-mucosa platform, to investigate the role mechanical cues in the epithelial barrier. In this platform, low-level mechanical stress (0.1 kPa) is applied to oral keratinocytes, which lie on 3D fibrous collagen (Col) gels whose stiffness is modulated by different concentrations or the addition of other factors such as fibronectin (FN). Our results show that cells lying on intermediate Col (3 mg/mL; stiffness = 30 Pa) demonstrated lower epithelial leakiness compared with soft Col (1.5 mg/mL; stiffness = 10 Pa) and stiff Col (6 mg/mL; stiffness = 120 Pa) gels, indicating that stiffness modulates barrier function. In addition, the presence of FN reversed the barrier integrity by inhibiting the interepithelial interaction via E-cadherin and Zonula occludens-1. Overall, the 3D Oral Epi-mucosa platform, as a new in vitro system, will be utilized to identify new mechanisms and develop future targets involved in mucosal diseases.
Collapse
Affiliation(s)
- Eun-Jin Lee
- Department of Biochemistry and Molecular & Cellular Biology, School of Medicine, Georgetown University, Washington, DC 20057, USA
- Microsystems and Nanotechnology Division, Physical Measurement Laboratory, National Institute of Standards and Technology, Gaithersburg, MD 20899, USA
- Department of Chemistry and Biochemistry, College of Computer, Mathematical and Natural Sciences, University of Maryland, College Park, MD 20742, USA
| | - Yoontae Kim
- Department of Biochemistry and Molecular & Cellular Biology, School of Medicine, Georgetown University, Washington, DC 20057, USA
| | - Paul Salipante
- Materials Science and Engineering Division, Material Measurement Laboratory, National Institute of Standards and Technology, Gaithersburg, MD 20899, USA
| | - Anthony P Kotula
- Materials Science and Engineering Division, Material Measurement Laboratory, National Institute of Standards and Technology, Gaithersburg, MD 20899, USA
| | - Sophie Lipshutz
- Department of Biochemistry and Molecular & Cellular Biology, School of Medicine, Georgetown University, Washington, DC 20057, USA
| | - Dana T Graves
- Department of Periodontics, School of Dental Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Stella Alimperti
- Department of Biochemistry and Molecular & Cellular Biology, School of Medicine, Georgetown University, Washington, DC 20057, USA
| |
Collapse
|
15
|
Nemoto S, Kubota T, Ohno H. Exploring body weight-influencing gut microbiota by elucidating the association with diet and host gene expression. Sci Rep 2023; 13:5593. [PMID: 37019989 PMCID: PMC10076326 DOI: 10.1038/s41598-023-32411-z] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2022] [Accepted: 03/27/2023] [Indexed: 04/07/2023] Open
Abstract
We aimed to identify gut microbiota that influences body weight by elucidating the association with diets and host genes. Germ-free (GF) mice with and without fecal microbiota transplant (FMT) were fed a normal, high-carbohydrate, or high-fat diet. FMT mice exhibited greater total body weight; adipose tissue and liver weights; blood glucose, insulin, and total cholesterol levels; and oil droplet size than the GF mice, regardless of diet. However, the extent of weight gain and metabolic parameter levels associated with gut microbiota depended on the nutrients ingested. For example, a disaccharide- or polysaccharide-rich diet caused more weight gain than a monosaccharide-rich diet. An unsaturated fatty acid-rich diet had a greater microbial insulin-increasing effect than a saturated fatty acid-rich diet. Perhaps the difference in microbial metabolites produced from substances taken up by the host created metabolic differences. Therefore, we analyzed such dietary influences on gut microbiota, differentially expressed genes between GF and FMT mice, and metabolic factors, including body weight. The results revealed a correlation between increased weight gain, a fat-rich diet, increased Ruminococcaceae abundance, and decreased claudin 22 gene expression. These findings suggest that weight regulation might be possible through the manipulation of the gut microbiota metabolism using the host's diet.
Collapse
Affiliation(s)
- Shino Nemoto
- Laboratory for Intestinal Ecosystem, RIKEN Center for Integrative Medical Sciences, Kanagawa, Japan.
| | - Tetsuya Kubota
- Laboratory for Intestinal Ecosystem, RIKEN Center for Integrative Medical Sciences, Kanagawa, Japan
- Division of Diabetes and Metabolism, The Institute of Medical Science, Asahi Life Foundation, Tokyo, Japan
- Department of Clinical Nutrition, National Institutes of Biomedical Innovation, Health and Nutrition, Tokyo, Japan
- Division of Cardiovascular Medicine, Toho University Ohashi Medical Center, Tokyo, Japan
- Department of Diabetes and Metabolic Diseases, Graduate School of Medicine, The University of Tokyo, Tokyo, Japan
| | - Hiroshi Ohno
- Laboratory for Intestinal Ecosystem, RIKEN Center for Integrative Medical Sciences, Kanagawa, Japan
- Laboratory for Immune Regulation, Graduate School of Medical and Pharmaceutical Sciences, Chiba University, Chiba, Japan
- Immunobiology Laboratory, Graduate School of Medical Life Science, Yokohama City University, Kanagawa, Japan
| |
Collapse
|
16
|
Kim WH, Kim JE, Kim S, Na Y, Hong YD, Choi J, Park WS, Shim SM. Bioconversion of BIOGF1K, a compound-K-rich fraction from ginseng root and its effect on epidermal barrier function. Heliyon 2023; 9:e14803. [PMID: 37025761 PMCID: PMC10070716 DOI: 10.1016/j.heliyon.2023.e14803] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2022] [Revised: 03/10/2023] [Accepted: 03/17/2023] [Indexed: 03/29/2023] Open
Abstract
BIOGF1K, the ginseng root-based and hydrolyzed ginsenoside-rich fraction, is known to improve skin damage, but there are rare studies on the kinetic of ginsenosides in the epidermis and their effects on epidermal barrier function. The current study investigated the effect of BIOGF1K on epidermal barrier function and its kinetics on epidermal transport. HPLC and LC/MS were used to verify the ginsenosides and the metabolites of BIOGF1K. Human immortalized keratinocytes (HaCaT) and epidermis-dermis artificial skin were treated with BIOGF1K and their metabolites were analyzed by HPLC and LC/MS. The epidermal barrier function was evaluated by transepithelial electrical resistance (TEER). In BIOGF1K, ginsenoside Rg1, Rd, F1, F2, compound Mc, compound Y (CY), and compound K (CK) were detected and CK and CY were the most and second abundant ginsenosides. TEER of HaCaT with 100 and 200 μg/mL BIOGF1K treatment was significantly higher than the control during 600 min of incubation. CK was permeated to the epidermis in a time-dependent manner and its maximum transported rate was observed at 600 min. In the case of artificial skin, CY and CK were permeated to the epidermis-dermis skin as time-dependent. Also, 24 h after treatment of CY, CK was detected as 19.59% of CY. It was proposed that CY was hydrolyzed into CK while permeating the epidermis. Results from the current study suggest that bioconversion of BIOGF1K rich in CK effectively enhances epidermal barrier function and it could be a useful cosmeceutical to exhibit its functionality to the skin.
Collapse
Affiliation(s)
- Woo-Hyun Kim
- Department of Food Science and Biotechnology, Sejong University, 98 Gunja-dong, Seoul 05006, South Korea
| | - Jeong-Eun Kim
- Department of Food Science and Biotechnology, Sejong University, 98 Gunja-dong, Seoul 05006, South Korea
| | - Sehyun Kim
- AMOREPACIFIC Research and Innovation Center, 1920, Yonggu-daero, Giheung-gu, Yongin-si 17074, Gyeonggi-do, South Korea
| | - Yongjoo Na
- AMOREPACIFIC Research and Innovation Center, 1920, Yonggu-daero, Giheung-gu, Yongin-si 17074, Gyeonggi-do, South Korea
| | - Yong-Deok Hong
- AMOREPACIFIC Research and Innovation Center, 1920, Yonggu-daero, Giheung-gu, Yongin-si 17074, Gyeonggi-do, South Korea
| | - Joonho Choi
- AMOREPACIFIC Research and Innovation Center, 1920, Yonggu-daero, Giheung-gu, Yongin-si 17074, Gyeonggi-do, South Korea
| | - Won-Seok Park
- AMOREPACIFIC Research and Innovation Center, 1920, Yonggu-daero, Giheung-gu, Yongin-si 17074, Gyeonggi-do, South Korea
- Corresponding author.
| | - Soon-Mi Shim
- Department of Food Science and Biotechnology, Sejong University, 98 Gunja-dong, Seoul 05006, South Korea
- Corresponding author.
| |
Collapse
|
17
|
Abstract
The pentapeptide L-R5 has previously been shown to transiently increase the permeability of nasal epithelial cell layers in vitro, allowing paracellular transport of molecules of up to 4 kDa. Protein kinase C zeta (PKC ζ), a member of a family of serine/threonine kinases was shown to be involved in tight junction modulation induced by L-R5. We show here that the ability of L-R5 to modulate tight junctions is comparable to other permeability enhancers such as bilobalide, latrunculin A or C10. Interaction of the peptide with the target protein occurs via electrostatic interaction, with the presence of positive charges being essential for its functionality. L-R5 is myristoylated to allow quick cell entry and onset of activity. While no epithelial cytotoxicity was detected, the hydrophobic myristoyl rest was shown to cause haemolysis. Taken together, these data show that a structural optimization of L-R5 may be possible, both from a toxicological and an efficacy point of view.
Collapse
Affiliation(s)
- Joël Brunner
- Section of Pharmaceutical Sciences, Institute of Pharmaceutical Sciences of Western Switzerland (ISPSO), University of Geneva, Switzerland
| | - Gerrit Borchard
- Section of Pharmaceutical Sciences, Institute of Pharmaceutical Sciences of Western Switzerland (ISPSO), University of Geneva, Switzerland,CONTACT Gerrit Borchard Section of Pharmaceutical Sciences, Institute of Pharmaceutical Sciences of Western Switzerland (ISPSO), University of Geneva, Switzerland
| |
Collapse
|
18
|
Zhao M, Rolandi M, Isseroff RR. Bioelectric Signaling: Role of Bioelectricity in Directional Cell Migration in Wound Healing. Cold Spring Harb Perspect Biol 2022; 14:a041236. [PMID: 36041786 PMCID: PMC9524286 DOI: 10.1101/cshperspect.a041236] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/24/2022]
Abstract
In wound healing, individual cells' behaviors coordinate movement toward the wound center to restore small or large barrier defects. The migration of epithelial cells as a continuous sheet structure is one of the most important processes by which the skin barrier is restored. How such multicellular and tissue level movement is initiated upon injury, coordinated during healing, and stopped when wounds healed has been a research focus for decades. When skin is wounded, the compromised epithelial barrier generates endogenous electric fields (EFs), produced by ion channels and maintained by cell junctions. These EFs are present across wounds, with the cathodal pole at the wound center. Epithelial cells detect minute EFs and migrate directionally in response to electrical signals. It has long been postulated that the naturally occurring EFs facilitate wound healing by guiding cell migration. It is not until recently that experimental evidence has shown that large epithelial sheets of keratinocytes or corneal epithelial cells respond to applied EFs by collective directional migration. Although some of the mechanisms of the collective cell migration are similar to those used by isolated cells, there are unique mechanisms that govern the coordinated movement of the cohesive sheet. We will review the understanding of wound EFs and how epithelial cells and other cells important to wound healing respond to the electric signals individually as well as collectively. Mounting evidence suggests that wound bioelectrical signaling is an important mechanism in healing. Critical understanding and proper exploitation of this mechanism will be important for better wound healing and regeneration.
Collapse
Affiliation(s)
- Min Zhao
- Department of Ophthalmology & Vision Science, University of California, Davis, Sacramento, California 95817, USA
- Department of Dermatology, University of California, Davis, California 95616, USA
| | - Marco Rolandi
- Department of Electrical and Computer Engineering, University of California Santa Cruz, Santa Cruz, California 95064, USA
| | - R Rivkah Isseroff
- Department of Dermatology, University of California, Davis, California 95616, USA
| |
Collapse
|
19
|
El-Serafi AT, El-Serafi I, Steinvall I, Sjöberg F, Elmasry M. A Systematic Review of Keratinocyte Secretions: A Regenerative Perspective. Int J Mol Sci 2022; 23:7934. [PMID: 35887279 PMCID: PMC9323141 DOI: 10.3390/ijms23147934] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2022] [Revised: 07/03/2022] [Accepted: 07/15/2022] [Indexed: 02/01/2023] Open
Abstract
Cell regenerative therapy is a modern solution for difficult-to-heal wounds. Keratinocytes, the most common cell type in the skin, are difficult to obtain without the creation of another wound. Stem cell differentiation towards keratinocytes is a challenging process, and it is difficult to reproduce in chemically defined media. Nevertheless, a co-culture of keratinocytes with stem cells usually achieves efficient differentiation. This systematic review aims to identify the secretions of normal human keratinocytes reported in the literature and correlate them with the differentiation process. An online search revealed 338 references, of which 100 met the selection criteria. A total of 80 different keratinocyte secretions were reported, which can be grouped mainly into cytokines, growth factors, and antimicrobial peptides. The growth-factor group mostly affects stem cell differentiation into keratinocytes, especially epidermal growth factor and members of the transforming growth factor family. Nevertheless, the reported secretions reflected the nature of the involved studies, as most of them focused on keratinocyte interaction with inflammation. This review highlights the secretory function of keratinocytes, as well as the need for intense investigation to characterize these secretions and evaluate their regenerative capacities.
Collapse
Affiliation(s)
- Ahmed T. El-Serafi
- Department of Biomedical and Clinical Sciences, Linköping University, 58183 Linkoping, Sweden; (I.S.); (F.S.); (M.E.)
- Department of Hand Surgery, Plastic Surgery and Burns, Linköping University, 58183 Linkoping, Sweden;
| | - Ibrahim El-Serafi
- Department of Hand Surgery, Plastic Surgery and Burns, Linköping University, 58183 Linkoping, Sweden;
- Basic Medical Sciences Department, College of Medicine, Ajman University, Ajman P.O. Box 346, United Arab Emirates
| | - Ingrid Steinvall
- Department of Biomedical and Clinical Sciences, Linköping University, 58183 Linkoping, Sweden; (I.S.); (F.S.); (M.E.)
- Department of Hand Surgery, Plastic Surgery and Burns, Linköping University, 58183 Linkoping, Sweden;
| | - Folke Sjöberg
- Department of Biomedical and Clinical Sciences, Linköping University, 58183 Linkoping, Sweden; (I.S.); (F.S.); (M.E.)
- Department of Hand Surgery, Plastic Surgery and Burns, Linköping University, 58183 Linkoping, Sweden;
| | - Moustafa Elmasry
- Department of Biomedical and Clinical Sciences, Linköping University, 58183 Linkoping, Sweden; (I.S.); (F.S.); (M.E.)
- Department of Hand Surgery, Plastic Surgery and Burns, Linköping University, 58183 Linkoping, Sweden;
| |
Collapse
|
20
|
Clare AJ, Liu J, Copland DA, Theodoropoulou S, Dick AD. Unravelling the therapeutic potential of IL-33 for atrophic AMD. Eye (Lond) 2022; 36:266-272. [PMID: 34531552 PMCID: PMC8807696 DOI: 10.1038/s41433-021-01725-5] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2021] [Revised: 07/08/2021] [Accepted: 07/27/2021] [Indexed: 02/08/2023] Open
Abstract
Age-related macular degeneration (AMD), a degenerative disease affecting the retinal pigment epithelium (RPE) and photoreceptors in the macula, is the leading cause of central blindness in the elderly. AMD progresses to advanced stages of the disease, atrophic AMD (aAMD), or in 15% of cases "wet" or neovascular AMD (nAMD), associated with substantial vision loss. Whilst there has been advancement in therapies treating nAMD, to date, there are no licenced effective treatments for the 85% affected by aAMD, with disease managed by changes to diet, vitamin supplements, and regular monitoring. AMD has a complex pathogenesis, involving highly integrated and common age-related disease pathways, including dysregulated complement/inflammation, impaired autophagy, and oxidative stress. The intricacy of AMD pathogenesis makes therapeutic development challenging and identifying a target that combats the converging disease pathways is essential to provide a globally effective treatment. Interleukin-33 is a cytokine, classically known for the proinflammatory role it plays in allergic disease. Recent evidence across degenerative and inflammatory disease conditions reveals a diverse immune-modulatory role for IL-33, with promising therapeutic potential. Here, we will review IL-33 function in disease and discuss the future potential for this homeostatic cytokine in treating AMD.
Collapse
Affiliation(s)
- Alison J. Clare
- grid.5337.20000 0004 1936 7603Academic Unit of Ophthalmology, Translational Health Sciences, University of Bristol, Bristol, UK
| | - Jian Liu
- grid.5337.20000 0004 1936 7603Academic Unit of Ophthalmology, Translational Health Sciences, University of Bristol, Bristol, UK
| | - David A. Copland
- grid.5337.20000 0004 1936 7603Academic Unit of Ophthalmology, Translational Health Sciences, University of Bristol, Bristol, UK
| | - Sofia Theodoropoulou
- grid.5337.20000 0004 1936 7603Academic Unit of Ophthalmology, Translational Health Sciences, University of Bristol, Bristol, UK
| | - Andrew D. Dick
- grid.5337.20000 0004 1936 7603Academic Unit of Ophthalmology, Translational Health Sciences, University of Bristol, Bristol, UK ,grid.5337.20000 0004 1936 7603School of Cellular and Molecular Medicine, University of Bristol, Bristol, UK ,grid.439257.e0000 0000 8726 5837NIHR Biomedical Research Centre of Ophthalmology, Moorfields Eye Hospital, London, UK ,grid.83440.3b0000000121901201UCL Institute of Ophthalmology, London, UK
| |
Collapse
|
21
|
Deng L, Lu H, Tu C, Zhou T, Cao W, Gao C. A tough synthetic hydrogel with excellent post-loading of drugs for promoting the healing of infected wounds in vivo. MATERIALS SCIENCE & ENGINEERING. C, MATERIALS FOR BIOLOGICAL APPLICATIONS 2021; 134:112577. [PMID: 35525747 DOI: 10.1016/j.msec.2021.112577] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/20/2021] [Revised: 11/18/2021] [Accepted: 11/25/2021] [Indexed: 12/24/2022]
Abstract
Bacterial infection is a major obstacle to the wound healing process. The hydrogel dressings with a simpler structure and good antibacterial and wound healing performance are appealing for clinical application. Herein, a robust hydrogel was synthesized from acrylamide (AM), acrylic acid (AA) and N,N'-methylene diacrylamide (MBA) via a redox initiating polymerization. The polymerization conditions were optimized to obtain the hydrogel with minimum unreacted monomers, which were 0.25% and 0.12% for AM and AA, respectively. The hydrogel had good mechanical strength, and could effectively resist damage by external forces and maintain a good macroscopic shape. It showed large water uptake capacity, and could post load a wide range of molecules via hydrogen bonding and electrostatic interaction. Loading of antibiotic doxycycline (DOX) enabled the hydrogel with good antibacterial activity against both Gram-positive bacteria and Gram-negative bacteria in vitro and in vivo. In a rat model of methicillin-resistant Staphylococcus aureus (MRSA)-infected full-thickness skin defect wound, the DOX-loaded hydrogel showed good therapeutic effect. It could significantly promote the wound closure, increased the collagen coverage area, down-regulate the expressions of pro-inflammatory TNF-α and IL-1β factors, and up-regulate the expressions of anti-inflammatory IL-4 factor and CD31 neovascularization factor.
Collapse
Affiliation(s)
- Liwen Deng
- College of Biomedical Engineering and Instrument Science, Zhejiang University, Hangzhou 310027, China; MOE Key Laboratory of Macromolecular Synthesis and Functionalization, Department of Polymer Science and Engineering, Zhejiang University, Hangzhou 310027, China
| | - Huidan Lu
- Zhejiang University School of Medicine Second Affiliated Hospital, Hangzhou 310009, China
| | - Chenxi Tu
- Shanxi-Zheda Institute of Advanced Materials and Chemical Engineering, Taiyuan 030000, China; MOE Key Laboratory of Macromolecular Synthesis and Functionalization, Department of Polymer Science and Engineering, Zhejiang University, Hangzhou 310027, China
| | - Tong Zhou
- MOE Key Laboratory of Macromolecular Synthesis and Functionalization, Department of Polymer Science and Engineering, Zhejiang University, Hangzhou 310027, China
| | - Wangbei Cao
- MOE Key Laboratory of Macromolecular Synthesis and Functionalization, Department of Polymer Science and Engineering, Zhejiang University, Hangzhou 310027, China
| | - Changyou Gao
- College of Biomedical Engineering and Instrument Science, Zhejiang University, Hangzhou 310027, China; Shanxi-Zheda Institute of Advanced Materials and Chemical Engineering, Taiyuan 030000, China; MOE Key Laboratory of Macromolecular Synthesis and Functionalization, Department of Polymer Science and Engineering, Zhejiang University, Hangzhou 310027, China.
| |
Collapse
|
22
|
Pitak-Arnnop P, Auychai P, Subbalekha K, Malakul A, Meningaud JP, Neff A. An approach to a repeated self-biting tongue injury in a toddler. Arch Pediatr 2021; 28:576-579. [PMID: 34507865 DOI: 10.1016/j.arcped.2021.08.006] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2020] [Revised: 03/22/2021] [Accepted: 08/09/2021] [Indexed: 10/20/2022]
Abstract
BACKGROUND Children with tongue injuries often visit the pediatric emergency department. The vast majority of cases can be conservatively treated, while some injuries require operative repair. The aim of this article was to demonstrate a "back-to-basics" approach to a refractory bifid tongue injury in a toddler. CASE DESCRIPTION A 1-year-old toddler with a tongue injury was unsuccessfully treated three times within a week by a surgeon in another specialty; all reconstructions were mucosal only. The case was then referred to our maxillofacial unit for proper management. On examination, the recurrent injury seemed to occur because of the patient's self-biting habit. We performed the fourth reconstruction of the tongue muscles and mucosa, and because of no dental prosthetic laboratory available in our hospital, we used transparent adhesive drapes fixed by resorbable sutures to cover the patient's anterior teeth instead of bite guards. The toddler was fed via a nasogastric tube for 1 week under 2-day antibiotic prophylaxis and routine oral care. The patient was discharged without any complications 1 week later. CONCLUSIONS AND PRACTICAL IMPLICATIONS The causes of repeated orofacial injuries should be identified and require particular attention to establish a proper treatment. For intraoral injuries in pediatric patients, self-biting habits should not be overlooked. The application of materials in an operating theater can help the treating clinicians improve the treatment outcomes.
Collapse
Affiliation(s)
- Poramate Pitak-Arnnop
- Department of Oral and Maxillofacial Surgery, University Hospital of Giessen and Marburg UKGM GmbH, Campus Marburg, Faculty of Medicine, Philipps University, Marburg 35043, Germany.
| | - Prim Auychai
- Department of Pediatric Dentistry, Faculty of Dentistry, Chulalongkorn University, Bangkok, Thailand
| | - Keskanya Subbalekha
- Department of Oral and Maxillofacial Surgery, Faculty of Dentistry, Chulalongkorn University, Bangkok, Thailand
| | - Atinuch Malakul
- Department of Occlusion, Faculty of Dentistry, Chulalongkorn University, Bangkok, Thailand
| | - Jean-Paul Meningaud
- Department of Plastic, Reconstructive, Aesthetic and Maxillofacial Surgery, Henri Mondor University Hospital, AP-HP, Faculty of Medicine, University Paris-Est Créteil Val de Marne (Paris XII), Créteil, France
| | - Andreas Neff
- Department of Oral and Maxillofacial Surgery, University Hospital of Giessen and Marburg UKGM GmbH, Campus Marburg, Faculty of Medicine, Philipps University, Marburg 35043, Germany
| |
Collapse
|
23
|
An Overview of Physical, Microbiological and Immune Barriers of Oral Mucosa. Int J Mol Sci 2021; 22:ijms22157821. [PMID: 34360589 PMCID: PMC8346143 DOI: 10.3390/ijms22157821] [Citation(s) in RCA: 90] [Impact Index Per Article: 22.5] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2021] [Revised: 07/15/2021] [Accepted: 07/21/2021] [Indexed: 02/07/2023] Open
Abstract
The oral mucosa, which is the lining tissue of the oral cavity, is a gateway to the body and it offers first-line protection against potential pathogens, exogenous chemicals, airborne allergens, etc. by means of its physical and microbiological-immune barrier functions. For this reason, oral mucosa is considered as a mirror to the health of the individual as well as a guard or early warning system. It is organized in two main components: a physical barrier, which consists of stratified epithelial cells and cell-cell junctions, and a microbiological-immune barrier that keeps the internal environment in a condition of homeostasis. Different factors, including microorganism, saliva, proteins and immune components, have been considered to play a critical role in disruption of oral epithelial barrier. Altered mucosal structure and barrier functions results in oral pathologies as well as systemic diseases. About 700 kinds of microorganisms exist in the human mouth, constituting the oral microbiota, which plays a significant role on the induction, training and function of the host immune system. The immune system maintains the symbiotic relationship of the host with this microbiota. Crosstalk between the oral microbiota and immune system includes various interactions in homeostasis and disease. In this review, after reviewing briefly the physical barriers of oral mucosa, the fundamentals of oral microbiome and oral mucosal immunity in regard to their barrier properties will be addressed. Furthermore, their importance in development of new diagnostic, prophylactic and therapeutic strategies for certain diseases as well as in the application for personalized medicine will be discussed.
Collapse
|
24
|
Suárez LJ, Arboleda S, Angelov N, Arce RM. Oral Versus Gastrointestinal Mucosal Immune Niches in Homeostasis and Allostasis. Front Immunol 2021; 12:705206. [PMID: 34290715 PMCID: PMC8287884 DOI: 10.3389/fimmu.2021.705206] [Citation(s) in RCA: 33] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2021] [Accepted: 06/22/2021] [Indexed: 12/15/2022] Open
Abstract
Different body systems (epidermis, respiratory tract, cornea, oral cavity, and gastrointestinal tract) are in continuous direct contact with innocuous and/or potentially harmful external agents, exhibiting dynamic and highly selective interaction throughout the epithelia, which function as both a physical and chemical protective barrier. Resident immune cells in the epithelia are constantly challenged and must distinguish among antigens that must be either tolerated or those to which a response must be mounted for. When such a decision begins to take place in lymphoid foci and/or mucosa-associated lymphoid tissues, the epithelia network of immune surveillance actively dominates both oral and gastrointestinal compartments, which are thought to operate in the same immune continuum. However, anatomical variations clearly differentiate immune processes in both the mouth and gastrointestinal tract that demonstrate a wide array of independent immune responses. From single vs. multiple epithelia cell layers, widespread cell-to-cell junction types, microbial-associated recognition receptors, dendritic cell function as well as related signaling, the objective of this review is to specifically contrast the current knowledge of oral versus gut immune niches in the context of epithelia/lymphoid foci/MALT local immunity and systemic output. Related differences in 1) anatomy 2) cell-to-cell communication 3) antigen capture/processing/presentation 4) signaling in regulatory vs. proinflammatory responses and 5) systemic output consequences and its relations to disease pathogenesis are discussed.
Collapse
Affiliation(s)
- Lina J Suárez
- Departamento de Ciencias Básicas y Medicina Oral, Universidad Nacional de Colombia, Bogotá, Colombia
| | - Silie Arboleda
- Department of Periodontics and Dental Hygiene, School of Dentistry, University of Texas Health Science Center at Houston, Houston, TX, United States
| | - Nikola Angelov
- Department of Periodontics and Dental Hygiene, School of Dentistry, University of Texas Health Science Center at Houston, Houston, TX, United States
| | - Roger M Arce
- Department of Periodontics and Dental Hygiene, School of Dentistry, University of Texas Health Science Center at Houston, Houston, TX, United States
| |
Collapse
|
25
|
Zhang MY, Fang S, Gao H, Zhang X, Gu D, Liu Y, Wan J, Xie J. A critical role of AREG for bleomycin-induced skin fibrosis. Cell Biosci 2021; 11:40. [PMID: 33622407 PMCID: PMC7903615 DOI: 10.1186/s13578-021-00553-0] [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: 01/18/2021] [Accepted: 02/10/2021] [Indexed: 12/16/2022] Open
Abstract
We report our discovery of an important player in the development of skin fibrosis, a hallmark of scleroderma. Scleroderma is a fibrotic disease, affecting 70,000 to 150,000 Americans. Fibrosis is a pathological wound healing process that produces an excessive extracellular matrix to interfere with normal organ function. Fibrosis contributes to nearly half of human mortality. Scleroderma has heterogeneous phenotypes, unpredictable outcomes, no validated biomarkers, and no effective treatment. Thus, strategies to slow down scleroderma progression represent an urgent medical need. While a pathological wound healing process like fibrosis leaves scars and weakens organ function, oral mucosa wound healing is a scarless process. After re-analyses of gene expression datasets from oral mucosa wound healing and skin fibrosis, we discovered that several pathways constitutively activated in skin fibrosis are transiently induced during oral mucosa wound healing process, particularly the amphiregulin (Areg) gene. Areg expression is upregulated ~ 10 folds 24hrs after oral mucosa wound but reduced to the basal level 3 days later. During bleomycin-induced skin fibrosis, a commonly used mouse model for skin fibrosis, Areg is up-regulated throughout the fibrogenesis and is associated with elevated cell proliferation in the dermis. To demonstrate the role of Areg for skin fibrosis, we used mice with Areg knockout, and found that Areg deficiency essentially prevents bleomycin-induced skin fibrosis. We further determined that bleomycin-induced cell proliferation in the dermis was not observed in the Areg null mice. Furthermore, we found that inhibiting MEK, a downstream signaling effector of Areg, by selumetinib also effectively blocked bleomycin-based skin fibrosis model. Based on these results, we concluded that the Areg-EGFR-MEK signaling axis is critical for skin fibrosis development. Blocking this signaling axis may be effective in treating scleroderma.
Collapse
Affiliation(s)
- Mary Yinghua Zhang
- Department of Pediatrics, The Wells Center for Pediatric Research, Indiana University School of Medicine, Indianapolis, IN, USA
| | - Shuyi Fang
- Department of BioHealth Informatics, School of Informatics and Computing At IUPUI, Indiana University, Indianapolis, IN, USA
| | - Hongyu Gao
- The IU Simon Comprehensive Cancer Center, Indiana University, Indianapolis, IN, USA
| | - Xiaoli Zhang
- Department of Pediatrics, The Wells Center for Pediatric Research, Indiana University School of Medicine, Indianapolis, IN, USA
| | - Dongsheng Gu
- Department of Pediatrics, The Wells Center for Pediatric Research, Indiana University School of Medicine, Indianapolis, IN, USA
| | - Yunlong Liu
- Department of BioHealth Informatics, School of Informatics and Computing At IUPUI, Indiana University, Indianapolis, IN, USA
- The IU Simon Comprehensive Cancer Center, Indiana University, Indianapolis, IN, USA
- The Center for Computational Biology and Bioinformatics, Indiana University School of Medicine, Indianapolis, IN, USA
- Department of Medical and Molecular Genetics, Indiana University School of Medicine, Indianapolis, IN, USA
| | - Jun Wan
- Department of BioHealth Informatics, School of Informatics and Computing At IUPUI, Indiana University, Indianapolis, IN, USA
- The IU Simon Comprehensive Cancer Center, Indiana University, Indianapolis, IN, USA
- The Center for Computational Biology and Bioinformatics, Indiana University School of Medicine, Indianapolis, IN, USA
- Department of Medical and Molecular Genetics, Indiana University School of Medicine, Indianapolis, IN, USA
| | - Jingwu Xie
- Department of Pediatrics, The Wells Center for Pediatric Research, Indiana University School of Medicine, Indianapolis, IN, USA.
- The IU Simon Comprehensive Cancer Center, Indiana University, Indianapolis, IN, USA.
| |
Collapse
|