1
|
Chen D, Wu J, Zhang F, Lyu R, You Q, Qian Y, Cai Y, Tian X, Tao H, He Y, Nawaz W, Wu Z. Trained immunity of intestinal tuft cells during infancy enhances host defense against enteroviral infections in mice. EMBO Mol Med 2024; 16:2516-2538. [PMID: 39261649 PMCID: PMC11479266 DOI: 10.1038/s44321-024-00128-9] [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: 11/16/2023] [Revised: 07/23/2024] [Accepted: 08/12/2024] [Indexed: 09/13/2024] Open
Abstract
Innate immune cells have been acknowledged as trainable in recent years. While intestinal tuft cells are recognized for their crucial roles in the host defense against intestinal pathogens, there remains uncertainty regarding their trainability. Enterovirus 71 (EV71), a prevalent enterovirus that primarily infects children but rarely infects adults. At present, there is a significant expansion of intestinal tuft cells in the EV71-infected mouse model, which is associated with EV71-induced interleukin-25 (IL-25) production. Further, we found that IL-25 pre-treatment at 2 weeks old mouse enabled tuft cells to acquire immune memory. This was evidenced by the rapid expansion and stronger response of IL-25-trained tuft cells in response to EV71 infection at 6 weeks old, surpassing the reactivity of naïve tuft cells in mice without IL-25-trained progress. Interestingly, IL-25-trained intestinal tuft cells exhibit anti-enteroviral effect via producing a higher level of IL-25. Mechanically, IL-25 treatment upregulates spermidine/spermine acetyl-transferase enzyme (SAT1) expression, mediates intracellular polyamine deficiency, further inhibits enterovirus replication. In summary, tuft cells can be trained by IL-25, which supports faster and higher level IL-25 production in response to EV71 infection and further exhibits anti-enteroviral effect via SAT1-mediated intracellular polyamine deficiency. Given that IL-25 can be induced by multiple gut microbes during human growth and development, including shifts in gut flora abundance, which may partially explain the different susceptibility to enteroviral infections between adults and children.
Collapse
Affiliation(s)
- Deyan Chen
- Anhui Key Laboratory of Infection and Immunity, Bengbu Medical University, Bengbu, China
- Medical School of Nanjing University, Nanjing, Jiangsu, China
| | - Jing Wu
- Medical School of Nanjing University, Nanjing, Jiangsu, China
| | - Fang Zhang
- Department of Burn and Plastic Surgery, Affiliated Hospital of Zunyi Medical University, Zunyi, Guizhou, China
| | - Ruining Lyu
- Medical School of Nanjing University, Nanjing, Jiangsu, China
| | - Qiao You
- Medical School of Nanjing University, Nanjing, Jiangsu, China
| | - Yajie Qian
- Nanjing Stomatological Hospital, Medical School of Nanjing University, Nanjing, Jiangsu, China
| | - Yurong Cai
- School of Life Science, Ningxia University, Yinchuan, China
| | - Xiaoyan Tian
- Medical School of Nanjing University, Nanjing, Jiangsu, China
| | - Hongji Tao
- Medical School of Nanjing University, Nanjing, Jiangsu, China
| | - Yating He
- Medical School of Nanjing University, Nanjing, Jiangsu, China
| | - Waqas Nawaz
- Hȏpital Maisonneuve-Rosemont, School of medicine, University of Montreal, Montreal, Canada
| | - Zhiwei Wu
- Medical School of Nanjing University, Nanjing, Jiangsu, China.
- State Key Laboratory of Analytical Chemistry for Life Science, Nanjing University, Nanjing, Jiangsu, China.
- Yunnan Provincial Key Laboratory of Entomological Biopharmaceutical R&D, College of Pharmacy, Dali University, Dali, Yunnan, China.
| |
Collapse
|
2
|
Chong Y, Yu D, Han R, Li Y, Gu Y, Lu Z, Nie F, Wang L, Cui H. Preparation of Luvangetin Nanoemulsions: Antimicrobial Mechanism and Role in Infected Wound Healing. Int J Nanomedicine 2024; 19:5493-5509. [PMID: 38882542 PMCID: PMC11178095 DOI: 10.2147/ijn.s457322] [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: 12/30/2023] [Accepted: 05/07/2024] [Indexed: 06/18/2024] Open
Abstract
Purpose Incorporation of luvangetin in nanoemulsions for antimicrobial and therapeutic use in infected wound healing. Patients and Methods Luvangetin nanoemulsions were prepared by high-speed shear method and characterized based on their appearance structure, average droplet size, polydispersity index (PDI), electric potential, storage stability. Optimized formulation of luvangetin nanoemulsion by Box-Behnken design (BBD). The antimicrobial activity and antimicrobial mechanism of luvangetin nanoemulsions against common hospital pathogens, ie, Staphylococcus aureus (S. aureus) and Escherichia coli (E. coli), were investigated using luvangetin nanoemulsions. The biosafety of luvangetin nanoemulsion was evaluated through cytotoxicity, apoptosis, and reactive oxygen species (ROS) assay experiments using human normal epidermal cells and endothelial cells. Finally, the effect of luvangetin nanoemulsion on healing of infected wounds was investigated in B6 mice. Results Luvangetin nanoemulsion formulation consists of 2.5% sunflower seed oil, 10% emulsifier Span-20 and 7 minutes of shear time, and with good stability. Luvangetin nanoemulsion produces antibacterial activity against S. aureus and E. coli by disrupting the structure of bacterial cell membranes. Luvangetin nanoemulsion are biologically safe for HaCat and HUVEC. Luvangetin nanoemulsion showed good therapeutic effect on MRSA infected wounds in mice. Conclusion For the first time, developed a new formulation called luvangetin nanoemulsion, which exhibited superior antibacterial effects against Gram-positive bacteria. Luvangetin nanoemulsion has a favorable effect in promoting infected wound healing. We have combined luvangetin, which has multiple activities, with nanoemulsions to provide a new topical fungicidal formulation, and have comprehensively evaluated its effectiveness and safety, opening up new possibilities for further applications of luvangetin.
Collapse
Affiliation(s)
- Yang Chong
- Department of Traditional Chinese Medicine, The Affiliated Hospital of Yangzhou University, Yangzhou University, Yangzhou, Jiangsu, 225000, People's Republic of China
- Department of General Surgery, The Affiliated Hospital of Yangzhou University, Yangzhou University, Yangzhou, Jiangsu, 225000, People's Republic of China
| | - Dong Yu
- Department of Traditional Chinese Medicine, The Affiliated Hospital of Yangzhou University, Yangzhou University, Yangzhou, Jiangsu, 225000, People's Republic of China
- Department of General Surgery, The Affiliated Hospital of Yangzhou University, Yangzhou University, Yangzhou, Jiangsu, 225000, People's Republic of China
| | - Rui Han
- Institute of Translational Medicine, Medical College, Yangzhou University, Yangzhou, 225009, People's Republic of China
| | - Yanxu Li
- Department of General Surgery, The Affiliated Hospital of Yangzhou University, Yangzhou University, Yangzhou, Jiangsu, 225000, People's Republic of China
| | - Yali Gu
- Institute of Translational Medicine, Medical College, Yangzhou University, Yangzhou, 225009, People's Republic of China
| | - Zhaoyu Lu
- Department of Traditional Chinese Medicine, The Affiliated Hospital of Yangzhou University, Yangzhou University, Yangzhou, Jiangsu, 225000, People's Republic of China
- Department of General Surgery, The Affiliated Hospital of Yangzhou University, Yangzhou University, Yangzhou, Jiangsu, 225000, People's Republic of China
| | - Fengsong Nie
- Department of Traditional Chinese Medicine, The Affiliated Hospital of Yangzhou University, Yangzhou University, Yangzhou, Jiangsu, 225000, People's Republic of China
- Department of General Surgery, The Affiliated Hospital of Yangzhou University, Yangzhou University, Yangzhou, Jiangsu, 225000, People's Republic of China
| | - Lingli Wang
- Department of Traditional Chinese Medicine, The Affiliated Hospital of Yangzhou University, Yangzhou University, Yangzhou, Jiangsu, 225000, People's Republic of China
| | - Hengmi Cui
- Institute of Epigenetics and Epigenomics and College of Animal Science and Technology, Yangzhou University, Yangzhou, Jiangsu, 225009, People's Republic of China
| |
Collapse
|
3
|
Chansoria P, Chaudhari A, Etter EL, Bonacquisti EE, Heavey MK, Le J, Maruthamuthu MK, Kussatz CC, Blackwell J, Jasiewicz NE, Sellers RS, Maile R, Wallet SM, Egan TM, Nguyen J. Instantly adhesive and ultra-elastic patches for dynamic organ and wound repair. Nat Commun 2024; 15:4720. [PMID: 38830847 PMCID: PMC11148085 DOI: 10.1038/s41467-024-48980-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2022] [Accepted: 05/21/2024] [Indexed: 06/05/2024] Open
Abstract
Bioadhesive materials and patches are promising alternatives to surgical sutures and staples. However, many existing bioadhesives do not meet the functional requirements of current surgical procedures and interventions. Here, we present a translational patch material that exhibits instant adhesion to tissues (2.5-fold stronger than Tisseel, an FDA-approved fibrin glue), ultra-stretchability (stretching to >300% its original length without losing elasticity), compatibility with rapid photo-projection (<2 min fabrication time/patch), and ability to deliver therapeutics. Using our established procedures for the in silico design and optimization of anisotropic-auxetic patches, we created next-generation patches for instant attachment to tissues while conforming to a broad range of organ mechanics ex vivo and in vivo. Patches coated with extracellular vesicles derived from mesenchymal stem cells demonstrate robust wound healing capability in vivo without inducing a foreign body response and without the need for patch removal that can cause pain and bleeding. We further demonstrate a single material-based, void-filling auxetic patch designed for the treatment of lung puncture wounds.
Collapse
Affiliation(s)
- Parth Chansoria
- Division of Pharmacoengineering and Molecular Pharmaceutics, Eshelman School of Pharmacy, University of North Carolina at Chapel Hill, Chapel Hill, NC, 27599, USA
| | - Ameya Chaudhari
- Division of Pharmacoengineering and Molecular Pharmaceutics, Eshelman School of Pharmacy, University of North Carolina at Chapel Hill, Chapel Hill, NC, 27599, USA
| | - Emma L Etter
- Division of Pharmacoengineering and Molecular Pharmaceutics, Eshelman School of Pharmacy, University of North Carolina at Chapel Hill, Chapel Hill, NC, 27599, USA
| | - Emily E Bonacquisti
- Division of Pharmacoengineering and Molecular Pharmaceutics, Eshelman School of Pharmacy, University of North Carolina at Chapel Hill, Chapel Hill, NC, 27599, USA
| | - Mairead K Heavey
- Division of Pharmacoengineering and Molecular Pharmaceutics, Eshelman School of Pharmacy, University of North Carolina at Chapel Hill, Chapel Hill, NC, 27599, USA
| | - Jiayan Le
- Division of Pharmacoengineering and Molecular Pharmaceutics, Eshelman School of Pharmacy, University of North Carolina at Chapel Hill, Chapel Hill, NC, 27599, USA
| | - Murali Kannan Maruthamuthu
- Division of Pharmacoengineering and Molecular Pharmaceutics, Eshelman School of Pharmacy, University of North Carolina at Chapel Hill, Chapel Hill, NC, 27599, USA
| | - Caden C Kussatz
- Division of Pharmacoengineering and Molecular Pharmaceutics, Eshelman School of Pharmacy, University of North Carolina at Chapel Hill, Chapel Hill, NC, 27599, USA
| | - John Blackwell
- Division of Cardiothoracic Surgery, Department of Surgery, School of Medicine, University of North Carolina at Chapel Hill, Chapel Hill, NC, 27599, USA
| | - Natalie E Jasiewicz
- Division of Pharmacoengineering and Molecular Pharmaceutics, Eshelman School of Pharmacy, University of North Carolina at Chapel Hill, Chapel Hill, NC, 27599, USA
| | - Rani S Sellers
- Pathology and Laboratory Medicine, Department of Medicine, University of North Carolina, Chapel Hill, NC, 27599, USA
| | - Robert Maile
- Department of Surgery, University of North Carolina at Chapel Hill, Chapel Hill, NC, 27599, USA
- Department of Microbiology and Immunology, School of Medicine, University of North Carolina at Chapel Hill, Chapel Hill, NC, 27599, USA
| | - Shannon M Wallet
- Division of Oral and Craniofacial Health Sciences, University of North Carolina at Chapel Hill, Chapel Hill, NC, 27599, USA
| | - Thomas M Egan
- Division of Cardiothoracic Surgery, Department of Surgery, School of Medicine, University of North Carolina at Chapel Hill, Chapel Hill, NC, 27599, USA
- Joint Department of Biomedical Engineering, University of North Carolina at Chapel Hill, Chapel Hill, NC, 27599, USA
- North Carolina State University, Raleigh, NC, 27695, USA
| | - Juliane Nguyen
- Division of Pharmacoengineering and Molecular Pharmaceutics, Eshelman School of Pharmacy, University of North Carolina at Chapel Hill, Chapel Hill, NC, 27599, USA.
- Joint Department of Biomedical Engineering, University of North Carolina at Chapel Hill, Chapel Hill, NC, 27599, USA.
| |
Collapse
|
4
|
An N, Wang R, Li L, Wang B, Wang H, Peng G, Zhou H, Chen G. Celastrol alleviates diabetic vascular injury via Keap1/Nrf2-mediated anti-inflammation. Front Pharmacol 2024; 15:1360177. [PMID: 38881873 PMCID: PMC11176472 DOI: 10.3389/fphar.2024.1360177] [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: 12/22/2023] [Accepted: 05/10/2024] [Indexed: 06/18/2024] Open
Abstract
Introduction: Celastrol (Cel) is a widely used main component of Chinese herbal medicine with strong anti-inflammatory, antiviral and antitumor activities. In the present study, we aimed to elucidate the cellular molecular protective mechanism of Cel against diabetes-induced inflammation and endothelial dysfunction. Methods: Type 2 diabetes (T2DM) was induced by db/db mice, and osmotic pumps containing Cel (100 μg/kg/day) were implanted intraperitoneally and were calibrated to release the drug for 28 days. In addition, human umbilical vein endothelial cells (HUVECs) were cultured in normal or high glucose and palmitic acid-containing (HG + PA) media in the presence or absence of Cel for 48 h. Results: Cel significantly ameliorated the hyperglycemia-induced abnormalities in nuclear factor (erythroid-derived 2)-like protein 2 (Nrf2) pathway activity and alleviated HG + PA-induced oxidative damage. However, the protective effect of Cel was almost completely abolished in HUVECs transfected with short hairpin (sh)RNA targeting Nrf2, but not by nonsense shRNA. Furthermore, HG + PA reduced the phosphorylation of AMP-activated protein kinase (AMPK), the autophagic degradation of p62/Kelch-like ECH-associated protein 1 (Keap1), and the nuclear localization of Nrf2. However, these catabolic pathways were inhibited by Cel treatment in HUVECs. In addition, compound C (AMPK inhibitors) and AAV9-sh-Nrf2 reduced Cel-induced Nrf2 activation and angiogenesis in db/db mice. Discussion: Taking these findings together, the endothelial protective effect of Cel in the presence of HG + PA may be at least in part attributed to its effects to reduce reactive oxygen species (ROS) and inflammation through p62/Keap1-mediated Nrf2 activation.
Collapse
Affiliation(s)
- Ning An
- The Affiliated Li Huili Hospital of Ningbo University, Health Science Center, Ningbo University, Ningbo, China
| | - Rixiang Wang
- The Affiliated Li Huili Hospital of Ningbo University, Health Science Center, Ningbo University, Ningbo, China
| | - Lin Li
- The Affiliated Li Huili Hospital of Ningbo University, Health Science Center, Ningbo University, Ningbo, China
| | - Bingyu Wang
- The Affiliated Li Huili Hospital of Ningbo University, Health Science Center, Ningbo University, Ningbo, China
| | - Huiting Wang
- Department of Pharmacology, Health Science Center, Ningbo University, Ningbo, China
| | - Ganyu Peng
- Department of Pharmacology, Health Science Center, Ningbo University, Ningbo, China
| | - Hua Zhou
- The Affiliated Li Huili Hospital of Ningbo University, Health Science Center, Ningbo University, Ningbo, China
| | - Gen Chen
- The Affiliated Li Huili Hospital of Ningbo University, Health Science Center, Ningbo University, Ningbo, China
- Department of Pharmacology, Health Science Center, Ningbo University, Ningbo, China
| |
Collapse
|
5
|
Mu X, Gu R, Tang M, Wu X, He W, Nie X. IL-17 in wound repair: bridging acute and chronic responses. Cell Commun Signal 2024; 22:288. [PMID: 38802947 PMCID: PMC11129447 DOI: 10.1186/s12964-024-01668-w] [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: 03/07/2024] [Accepted: 05/18/2024] [Indexed: 05/29/2024] Open
Abstract
Chronic wounds, resulting from persistent inflammation, can trigger a cascade of detrimental effects including exacerbating inflammatory cytokines, compromised blood circulation at the wound site, elevation of white blood cell count, increased reactive oxygen species, and the potential risk of bacterial infection. The interleukin-17 (IL-17) signaling pathway, which plays a crucial role in regulating immune responses, has been identified as a promising target for treating inflammatory skin diseases. This review aims to delve deeper into the potential pathological role and molecular mechanisms of the IL-17 family and its pathways in wound repair. The intricate interactions between IL-17 and other cytokines will be discussed in detail, along with the activation of various signaling pathways, to provide a comprehensive understanding of IL-17's involvement in chronic wound inflammation and repair.
Collapse
Affiliation(s)
- Xingrui Mu
- College of Pharmacy, Zunyi Medical University, Zunyi, 563006, China
- Key Lab of the Basic Pharmacology of the Ministry of Education & Joint International Research Laboratory of Ethnomedicine of Ministry of Education, Zunyi Medical University, Zunyi, 563006, China
| | - Rifang Gu
- School Medical Office, Zunyi Medical University, Zunyi, 563006, China
| | - Ming Tang
- Department of Structural Biology, St. Jude Children's Research Hospital, Memphis, TN, 38105, USA
| | - Xingqian Wu
- College of Pharmacy, Zunyi Medical University, Zunyi, 563006, China
- Key Lab of the Basic Pharmacology of the Ministry of Education & Joint International Research Laboratory of Ethnomedicine of Ministry of Education, Zunyi Medical University, Zunyi, 563006, China
| | - Wenjie He
- College of Pharmacy, Zunyi Medical University, Zunyi, 563006, China
- Key Lab of the Basic Pharmacology of the Ministry of Education & Joint International Research Laboratory of Ethnomedicine of Ministry of Education, Zunyi Medical University, Zunyi, 563006, China
| | - Xuqiang Nie
- College of Pharmacy, Zunyi Medical University, Zunyi, 563006, China.
- Key Lab of the Basic Pharmacology of the Ministry of Education & Joint International Research Laboratory of Ethnomedicine of Ministry of Education, Zunyi Medical University, Zunyi, 563006, China.
| |
Collapse
|
6
|
Cheng Y, Ren L, Niyazi A, Sheng L, Zhao Y. Identification of potential immunologic resilience in the healing process of diabetic foot ulcers. Int Wound J 2024; 21:e14465. [PMID: 37926487 PMCID: PMC10898407 DOI: 10.1111/iwj.14465] [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: 09/07/2023] [Accepted: 10/15/2023] [Indexed: 11/07/2023] Open
Abstract
Diabetic foot ulcers (DFUs) are one of the most common and challenging complications of diabetes, yet our understanding of their pathogenesis remains limited. We collected gene expression data of DFU patients from public databases. Bioinformatics tools were applied for systematic analysis, including the identification of differentially expressed genes (DEGs), weighted gene co-expression network analysis (WGCNA) and enrichment analysis. We further used single-cell RNA sequencing to identify the distribution of different cell populations in DFU. Finally, key results were validated using reverse transcription-quantitative polymerase chain reaction (RT-qPCR) and flow cytometry. We identified 217 DEGs between ulcerated and healthy skin, and 37 DEGs between healing ulcers and ulcers. WGCNA revealed that the cyan module had the highest positive correlation with healthy skin and negative correlation with ulcers. The black module had the highest negative correlation with healthy skin and positive correlation with ulcers. Enrichment analysis showed that the genes in the cyan module were mainly associated with complement and coagulation cascades, while the genes in the black module were mainly associated with the IL-17 signalling pathway. In addition, CD8 T cells were significantly lower in ulcers than in healthy and healing ulcers. By comparing marker genes of CD8 T cells, we identified key genes in the cyan and black modules and validated their expression using RT-qPCR. The proportion of CD8 T cells was increased in healing ulcers. Flow cytometry detected increased levels of CD8 T, B and natural killer cells in healing ulcers. CD8 T cells and related key genes play an important role in the healing process of DFU. The results of this study provide a new perspective for understanding the pathogenesis and treatment of DFU.
Collapse
Affiliation(s)
- Yifeng Cheng
- Department of BurnsThe First Affiliated Hospital of Xinjiang Medical UniversityXinjiangChina
| | - Lei Ren
- Department of BurnsThe First Affiliated Hospital of Xinjiang Medical UniversityXinjiangChina
| | - Aihemaitijiang Niyazi
- Department of BurnsThe First Affiliated Hospital of Xinjiang Medical UniversityXinjiangChina
| | - Li Sheng
- Department of BurnsThe First Affiliated Hospital of Xinjiang Medical UniversityXinjiangChina
| | - Yang Zhao
- Department of BurnsThe First Affiliated Hospital of Xinjiang Medical UniversityXinjiangChina
| |
Collapse
|
7
|
Lyu R, Wu J, He Y, You Q, Qian Y, Jiang N, Cai Y, Chen D, Wu Z. Folate supports IL-25-induced tuft cell expansion following enteroviral infections. FASEB J 2024; 38:e23430. [PMID: 38243751 DOI: 10.1096/fj.202301928r] [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: 09/20/2023] [Revised: 12/11/2023] [Accepted: 01/03/2024] [Indexed: 01/21/2024]
Abstract
Intestinal tuft cells, a kind of epithelial immune cells, rapidly expand in response to pathogenic infections, which is associated with infection-induced interleukin 25 (IL-25) upregulation. However, the metabolic mechanism of IL-25-induced tuft cell expansion is largely unknown. Folate metabolism provides essential purine and methyl substrates for cell proliferation and differentiation. Thus, we aim to investigate the roles of folate metabolism playing in IL-25-induced tuft cell expansion by enteroviral infection and recombinant murine IL-25 (rmIL-25) protein-stimulated mouse models. At present, enteroviruses, such as EV71, CVA16, CVB3, and CVB4, upregulated IL-25 expression and induced tuft cell expansion in the intestinal tissues of mice. However, EV71 did not induce intestinal tuft cell expansion in IL-25-/- mice. Interestingly, compared to the mock group, folate was enriched in the intestinal tissues of both the EV71-infected group and the rmIL-25 protein-stimulated group. Moreover, folate metabolism supported IL-25-induced tuft cell expansion since both folate-depletion and anti-folate MTX-treated mice had a disrupted tuft cell expansion in response to rmIL-25 protein stimulation. In summary, our data suggested that folate metabolism supported intestinal tuft cell expansion in response to enterovirus-induced IL-25 expression, which provided a new insight into the mechanisms of tuft cell expansion from the perspective of folate metabolism.
Collapse
Affiliation(s)
- Ruining Lyu
- Center for Public Health Research, Medical School, Nanjing University, Nanjing, China
| | - Jing Wu
- Center for Public Health Research, Medical School, Nanjing University, Nanjing, China
| | - Yating He
- Center for Public Health Research, Medical School, Nanjing University, Nanjing, China
| | - Qiao You
- Center for Public Health Research, Medical School, Nanjing University, Nanjing, China
| | - Yajie Qian
- Nanjing Stomatological Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing, China
| | - Na Jiang
- Center for Public Health Research, Medical School, Nanjing University, Nanjing, China
| | - Yurong Cai
- School of Life Science, Ningxia University, Yinchuan, China
| | - Deyan Chen
- Center for Public Health Research, Medical School, Nanjing University, Nanjing, China
| | - Zhiwei Wu
- Center for Public Health Research, Medical School, Nanjing University, Nanjing, China
- School of Life Science, Ningxia University, Yinchuan, China
- State Key Laboratory of Analytical Chemistry for Life Science, Nanjing University, Nanjing, China
- Jiangsu Key Laboratory of Molecular Medicine, Medical School, Nanjing University, Nanjing, China
| |
Collapse
|
8
|
Zhao Y, Zhao Y, Xu B, Liu H, Chang Q. Microenvironmental dynamics of diabetic wounds and insights for hydrogel-based therapeutics. J Tissue Eng 2024; 15:20417314241253290. [PMID: 38818510 PMCID: PMC11138198 DOI: 10.1177/20417314241253290] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2023] [Accepted: 04/22/2024] [Indexed: 06/01/2024] Open
Abstract
The rising prevalence of diabetes has underscored concerns surrounding diabetic wounds and their potential to induce disability. The intricate healing mechanisms of diabetic wounds are multifaceted, influenced by ambient microenvironment, including prolonged hyperglycemia, severe infection, inflammation, elevated levels of reactive oxygen species (ROS), ischemia, impaired vascularization, and altered wound physicochemical properties. In recent years, hydrogels have emerged as promising candidates for diabetic wound treatment owing to their exceptional biocompatibility and resemblance to the extracellular matrix (ECM) through a three-dimensional (3D) porous network. This review will first summarize the microenvironment alterations occurring in the diabetic wounds, aiming to provide a comprehensive understanding of its pathogenesis, then a comprehensive classification of recently developed hydrogels will be presented, encompassing properties such as hypoglycemic effects, anti-inflammatory capabilities, antibacterial attributes, ROS scavenging abilities, promotion of angiogenesis, pH responsiveness, and more. The primary objective is to offer a valuable reference for repairing diabetic wounds based on their unique microenvironment. Moreover, this paper outlines potential avenues for future advancements in hydrogel dressings to facilitate and expedite the healing process of diabetic wounds.
Collapse
Affiliation(s)
- Ying Zhao
- Department of Plastic Surgery, The First Affiliated Hospital of Jinan University, Jinan University, Guangzhou, China
- Department of Burn and Plastic surgery, Jinan University Affiliated Shunde Hospital, Jinan University, Foshan, China
| | - Yulan Zhao
- Department of Nephropathy Rheumatology, Guizhou Medical University Affiliated Zhijin Hospital, Zhijin, China
| | - Bing Xu
- Department of Burn and Plastic surgery, Jinan University Affiliated Shunde Hospital, Jinan University, Foshan, China
| | - Hongwei Liu
- Department of Plastic Surgery, The First Affiliated Hospital of Jinan University, Jinan University, Guangzhou, China
| | - Qiang Chang
- Department of Plastic and Reconstruction Surgery, Nanfang Hospital, Southern Medical University, Guangzhou, China
| |
Collapse
|
9
|
Ma Z, Mo R, Yang P, Ding Y, Zhang H, Dong Z, Chen Y, Tan Q. PDK4 facilitates fibroblast functions and diabetic wound healing through regulation of HIF-1α protein stability and gene expression. FASEB J 2023; 37:e23215. [PMID: 37737961 DOI: 10.1096/fj.202300874rr] [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: 05/02/2023] [Revised: 09/06/2023] [Accepted: 09/12/2023] [Indexed: 09/23/2023]
Abstract
Fibroblast activation disorder is one of the main pathogenic characteristics of diabetic wounds. Orchestrated fibroblast functions and myofibroblast differentiation are crucial for wound contracture and extracellular matrix (ECM) formation. Pyruvate dehydrogenase kinase 4 (PDK4), a key enzyme regulating energy metabolism, has been implicated in modulating fibroblast function, but its specific role in diabetic wounds remains poorly understood. In this study, we investigated the impact of PDK4 on diabetic wounds and its underlying mechanisms. To assess the effect of PDK4 on human dermal fibroblasts (HDFs), we conducted CCK-8, EdU proliferation assay, wound healing assay, transwell assay, flow cytometry, and western blot analyses. Metabolic shifts were analyzed using the Seahorse XF analyzer, while changes in metabolite expression were measured through LC-MS. Local recombinant PDK4 administration was implemented to evaluate its influence on wound healing in diabetic mice. Finally, we found that sufficient PDK4 expression is essential for a normal wound-healing process, while PDK4 is low expressed in diabetic wound tissues and fibroblasts. PDK4 promotes proliferation, migration, and myofibroblast differentiation of HDFs and accelerates wound healing in diabetic mice. Mechanistically, PDK4-induced metabolic reprogramming increases the level of succinate that inhibits PHD2 enzyme activity, thus leading to the stability of the HIF-1α protein, during which process the elevated HIF-1α mRNA by PDK4 is also indispensable. In conclusion, PDK4 promotes fibroblast functions through regulation of HIF-1α protein stability and gene expression. Local recombinant PDK4 administration accelerates wound healing in diabetic mice.
Collapse
Affiliation(s)
- Zhouji Ma
- Department of Burns and Plastic Surgery, Nanjing Drum Tower Hospital Clinical College of Nanjing Medical University, Nanjing, China
| | - Ran Mo
- Department of Burns and Plastic Surgery, Nanjing Drum Tower Hospital, the Affiliated Hospital of Nanjing University Medical School, Nanjing, China
| | - Ping Yang
- Department of Burns and Plastic Surgery, Nanjing Drum Tower Hospital, the Affiliated Hospital of Nanjing University Medical School, Nanjing, China
| | - Youjun Ding
- Department of Burns and Plastic Surgery, Nanjing Drum Tower Hospital Clinical College of Jiangsu University, Nanjing, China
- Department of Emergency Surgery, The Fourth Affiliated Hospital of Jiangsu University (Zhenjiang Fourth People's Hospital), Zhenjiang, China
| | - Hao Zhang
- Department of Burns and Plastic Surgery, Nanjing Drum Tower Hospital, the Affiliated Hospital of Nanjing University Medical School, Nanjing, China
| | - Zheng Dong
- Department of Burns and Plastic Surgery, Nanjing Drum Tower Hospital, the Affiliated Hospital of Nanjing University Medical School, Nanjing, China
| | - Yutong Chen
- Department of Burns and Plastic Surgery, Nanjing Drum Tower Hospital, the Affiliated Hospital of Nanjing University Medical School, Nanjing, China
| | - Qian Tan
- Department of Burns and Plastic Surgery, Nanjing Drum Tower Hospital Clinical College of Nanjing Medical University, Nanjing, China
- Department of Burns and Plastic Surgery, Nanjing Drum Tower Hospital, the Affiliated Hospital of Nanjing University Medical School, Nanjing, China
- Department of Burns and Plastic Surgery, Anqing Shihua Hospital, Nanjing Drum Tower Hospital Group, Anqing, China
| |
Collapse
|
10
|
Zhang M, Yao A, Ai F, Lin J, Fu Q, Wang D. Cobalt-containing borate bioactive glass fibers for treatment of diabetic wound. JOURNAL OF MATERIALS SCIENCE. MATERIALS IN MEDICINE 2023; 34:42. [PMID: 37530851 PMCID: PMC10397116 DOI: 10.1007/s10856-023-06741-3] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/16/2023] [Accepted: 07/10/2023] [Indexed: 08/03/2023]
Abstract
Impaired angiogenesis is one of the predominant reasons for non-healing diabetic wounds. Cobalt is well known for its capacity to induce angiogenesis by stabilizing hypoxia-inducible factor-1α (HIF-1α) and subsequently inducing the production of vascular endothelial growth factor (VEGF). In this study, Co-containing borate bioactive glasses and their derived fibers were fabricated by partially replacing CaO in 1393B3 borate glass with CoO. Fourier transform infrared (FTIR) spectroscopy and nuclear magnetic resonance (NMR) analyses were performed to characterize the effect of Co incorporation on the glass structure, and the results showed that the substitution promoted the transformation of [BO3] into [BO4] units, which endow the glass with higher chemical durability and lower reaction rate with the simulated body fluid (SBF), thereby achieving sustained and controlled Co2+ ion release. In vitro biological assays were performed to assess the angiogenic potential of the Co-containing borate glass fibers. It was found that the released Co2+ ion significantly enhanced the proliferation, migration and tube formation of the Human Umbilical Vein Endothelial Cells (HUVECs) by upregulating the expression of angiogenesis-related proteins such as HIF-1α and VEGF. Finally. In vivo results demonstrated that the Co-containing fibers accelerated full-thickness skin wound healing in streptozotocin (STZ)-induced diabetic rat model by promoting angiogenesis and re-epithelialization.
Collapse
Affiliation(s)
- Minhui Zhang
- School of Materials Science and Engineering, Tongji University, 200092, Shanghai, China
| | - Aihua Yao
- School of Materials Science and Engineering, Tongji University, 200092, Shanghai, China
- Key Laboratory of Advanced Civil Engineering Materials, Ministry of Education, 200092, Shanghai, China
| | - Fanrong Ai
- School of Mechatronics Engineering, Nanchang University, 330031, Nanchang, China
| | - Jian Lin
- School of Materials Science and Engineering, Tongji University, 200092, Shanghai, China.
- Key Laboratory of Advanced Civil Engineering Materials, Ministry of Education, 200092, Shanghai, China.
| | - Qingge Fu
- Department of Orthopedic trauma, Changhai Hospital, Second Military Medical University, 200433, Shanghai, China.
| | - Deping Wang
- School of Materials Science and Engineering, Tongji University, 200092, Shanghai, China
- Key Laboratory of Advanced Civil Engineering Materials, Ministry of Education, 200092, Shanghai, China
| |
Collapse
|
11
|
Lecron JC, Charreau S, Jégou JF, Salhi N, Petit-Paris I, Guignouard E, Burucoa C, Favot-Laforge L, Bodet C, Barra A, Huguier V, Mcheik J, Dumoutier L, Garnier J, Bernard FX, Ryffel B, Morel F. IL-17 and IL-22 are pivotal cytokines to delay wound healing of S. aureus and P. aeruginosa infected skin. Front Immunol 2022; 13:984016. [PMID: 36275755 PMCID: PMC9585169 DOI: 10.3389/fimmu.2022.984016] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2022] [Accepted: 09/20/2022] [Indexed: 11/13/2022] Open
Abstract
IntroductionAlthough the presence of pathogens in skin wounds is known to delay the wound healing process, the mechanisms underlying this delay remain poorly understood. In the present study, we have investigated the regulatory role of proinflammatory cytokines on the healing kinetics of infected wounds.MethodsWe have developed a mouse model of cutaneous wound healing, with or without wound inoculation with Staphylococcus aureus and Pseudomonas aeruginosa, two major pathogens involved in cutaneous wound bacterial infections.ResultsAseptic excision in C57BL/6 mouse skin induced early expression of IL-1β, TNFα and Oncostatin M (OSM), without detectable expression of IL-22 and IL-17A/F. S. aureus and P. aeruginosa wound inoculation not only increased the expression of IL-1β and OSM, but also induced a strong cutaneous expression of IL-22, IL-17A and IL-17F, along with an increased number of infiltrating IL-17A and/or IL-22-producing γδ T cells. The same cytokine expression pattern was observed in infected human skin wounds. When compared to uninfected wounds, mouse skin infection delayed the wound healing process. Injection of IL-1α, TNFα, OSM, IL-22 and IL-17 together in the wound edges induced delayed wound healing similar to that induced by the bacterial infection. Wound healing experiments in infected Rag2KO mice (deficient in lymphocytes) showed a wound healing kinetic similar to uninfected Rag2KO mice or WT mice. Rag2KO infected-skin lesions expressed lower levels of IL-17 and IL-22 than WT, suggesting that the expression of these cytokines is mainly dependent on γδ T cells in this model. Wound healing was not delayed in infected IL-17R/IL-22KO, comparable to uninfected control mice. Injection of recombinant IL-22 and IL-17 in infected wound edges of Rag2KO mice re-establish the delayed kinetic of wound healing, as in infected WT mice.ConclusionThese results demonstrate the synergistic and specific effects of IL-22 and IL-17 induced by bacterial infection delay the wound healing process, regardless of the presence of bacteria per se. Therefore, these cytokines play an unexpected role in delayed skin wound healing.
Collapse
Affiliation(s)
- Jean-Claude Lecron
- Laboratoire Inflammation, Tissus Epithéliaux et Cytokines, UR15560, Université de Poitiers, Poitiers, France
- Laboratoire Immunologie et Inflammation, Centre Hospitalier et Universitaire (CHU) de Poitiers, Poitiers, France
- *Correspondence: Jean-Claude Lecron,
| | - Sandrine Charreau
- Laboratoire Inflammation, Tissus Epithéliaux et Cytokines, UR15560, Université de Poitiers, Poitiers, France
- Qima-Bioalternatives (Qima Life Sciences), Gençay, France
| | - Jean-François Jégou
- Laboratoire Inflammation, Tissus Epithéliaux et Cytokines, UR15560, Université de Poitiers, Poitiers, France
| | - Nadjet Salhi
- Laboratoire Inflammation, Tissus Epithéliaux et Cytokines, UR15560, Université de Poitiers, Poitiers, France
| | - Isabelle Petit-Paris
- Laboratoire Inflammation, Tissus Epithéliaux et Cytokines, UR15560, Université de Poitiers, Poitiers, France
| | - Emmanuel Guignouard
- Laboratoire Inflammation, Tissus Epithéliaux et Cytokines, UR15560, Université de Poitiers, Poitiers, France
| | - Christophe Burucoa
- Laboratoire Inflammation, Tissus Epithéliaux et Cytokines, UR15560, Université de Poitiers, Poitiers, France
- Laboratoire de Bactériologie, Centre Hospitalier et Universitaire (CHU) de Poitiers, Poitiers, France
| | - Laure Favot-Laforge
- Laboratoire Inflammation, Tissus Epithéliaux et Cytokines, UR15560, Université de Poitiers, Poitiers, France
| | - Charles Bodet
- Laboratoire Inflammation, Tissus Epithéliaux et Cytokines, UR15560, Université de Poitiers, Poitiers, France
| | - Anne Barra
- Laboratoire Inflammation, Tissus Epithéliaux et Cytokines, UR15560, Université de Poitiers, Poitiers, France
- Laboratoire Immunologie et Inflammation, Centre Hospitalier et Universitaire (CHU) de Poitiers, Poitiers, France
| | - Vincent Huguier
- Laboratoire Inflammation, Tissus Epithéliaux et Cytokines, UR15560, Université de Poitiers, Poitiers, France
- Service de Chirurgie Plastique, Centre Hospitalier et Universitaire (CHU) de Poitiers, Poitiers, France
| | - Jiad Mcheik
- Laboratoire Inflammation, Tissus Epithéliaux et Cytokines, UR15560, Université de Poitiers, Poitiers, France
- Service de Chirurgie Pédiatrique, Centre Hospitalier et Universitaire CHU) de Poitiers, Poitiers, France
| | - Laure Dumoutier
- De Duve Institute, Université catholique de Louvain, Brussels, Belgium
| | - Julien Garnier
- Qima-Bioalternatives (Qima Life Sciences), Gençay, France
| | - François-Xavier Bernard
- Laboratoire Inflammation, Tissus Epithéliaux et Cytokines, UR15560, Université de Poitiers, Poitiers, France
- Qima-Bioalternatives (Qima Life Sciences), Gençay, France
| | - Bernhard Ryffel
- Laboratoire d'Immunologie et Neurogénétique Expérimentales et Moléculaire (INEM) - Unité Mixte de Recherche (UMR) 7355, Centre National de la Recherche Scientifique (CNRS) et Université d’Orléans, Orléans, France
| | - Franck Morel
- Laboratoire Inflammation, Tissus Epithéliaux et Cytokines, UR15560, Université de Poitiers, Poitiers, France
| |
Collapse
|
12
|
Chen L, Wang M, Zhong Z, Liu B, Zhang W, Zhu B, Jiao C, Yu C, Guan B. Role of Exosomes in Pharyngucutaneous Fistula After Total Laryngectomy. Int J Nanomedicine 2022; 17:4119-4135. [PMID: 36118178 PMCID: PMC9480600 DOI: 10.2147/ijn.s372042] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2022] [Accepted: 09/01/2022] [Indexed: 11/23/2022] Open
Abstract
Pharyngocutaneous fistula is the most common complication after total laryngectomy and is difficult to heal. Although conservative treatment and surgical repair are effective, they often take longer and additional trips to the operating room, which undoubtedly increases the financial burden on patients. Especially in combination with diseases such as diabetes and hypertension, which affect the efficacy of surgery. Adding growth factors into the repair material can promote fibroblast proliferation, angiogenesis, and accelerate wound healing. A substantial number of studies have shown that a type of nanoscale extracellular vesicle, called exosomes, facilitates organization repair by promoting blood vessel production, protein polysaccharides, and collagen deposition, thereby representing a new type of cellular therapy. At present, there is little research on the application of exosomes in pharyngocutaneous fistula regeneration after total laryngectomy. In this review, we summarize the biological characteristics of exosomes and their application in biomedical science, and highlight their application prospects in pharyngocutaneous fistula regeneration after total laryngectomy.
Collapse
Affiliation(s)
- Li Chen
- Department of Otolaryngology, Head and Neck Surgery, Dalian Medical University, Dalian, 116000, People's Republic of China
| | - Maohua Wang
- Department of Otolaryngology, Head and Neck Surgery, The First People's Hospital of Foshan, Foshan, 528000, People's Republic of China
| | - Zhenhua Zhong
- Department of Otolaryngology, Head and Neck Surgery, The Affiliated Hospital of Yangzhou University, Yangzhou University, Yangzhou, 225000, People's Republic of China
| | - Baoxu Liu
- Department of Otolaryngology, Head and Neck Surgery, Dalian Medical University, Dalian, 116000, People's Republic of China
| | - Wentao Zhang
- Department of Otolaryngology, Head and Neck Surgery, Clinical Medical College, Yangzhou University, Yangzhou, 225001, People's Republic of China
| | - Bin Zhu
- Department of Otolaryngology, Head and Neck Surgery, Clinical Medical College, Yangzhou University, Yangzhou, 225001, People's Republic of China
| | - Cheng Jiao
- Department of Otolaryngology, Head and Neck Surgery, Clinical Medical College, Yangzhou University, Yangzhou, 225001, People's Republic of China
| | - Chenjie Yu
- Department of Otolaryngology, Head and Neck Surgery, Affiliated Drum Tower Hospital of Nanjing University Medical School, Jiangsu Provincial Key Medical Discipline (Laboratory), Nanjing, 210008, People's Republic of China
| | - Bing Guan
- Department of Otolaryngology, Head and Neck Surgery, Clinical Medical College, Yangzhou University, Yangzhou, 225001, People's Republic of China
| |
Collapse
|
13
|
Hsieh MCW, Wang WT, Lin CY, Kuo YR, Lee SS, Hou MF, Wu YC. Stem Cell-Based Therapeutic Strategies in Diabetic Wound Healing. Biomedicines 2022; 10:biomedicines10092085. [PMID: 36140185 PMCID: PMC9495374 DOI: 10.3390/biomedicines10092085] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2022] [Revised: 08/10/2022] [Accepted: 08/23/2022] [Indexed: 11/16/2022] Open
Abstract
Impaired wound healing and especially the “all-too-common” occurrence of associated diabetic foot ulcers (DFU) are becoming an increasingly urgent and deteriorating healthcare issue, which drastically impact the quality of life and further heighten the risks of infection and amputation in patients with diabetes mellitus. Amongst the multifactorial wound healing determinants, glycemic dysregulation has been identified to be the primary casual factor of poor wound healing. Unfortunately, current therapeutic modalities merely serve as moderate symptomatic relieves but often fail to completely restore the wound site to its pre-injury state and prevent further recurrence. Stem cell-based therapeutics have been employed for its promising potential to address the root of the problem as they not only exhibit the capacity for self-renewal and differentiation towards multiple lineages, but also have been disclosed to participate in mediating variant growth factors and cytokines. Herein we review the current literatures on the therapeutic benefits of using various kinds of stem cells, including embryonic stem cells (ESCs), induced pluripotent stem cells (iPSCs), mesenchymal stem cells (MSCs), and adipose-derived stem cells (ASCs) in diabetic wound healing by searching on the PubMed® Database for publications. This study shall serve as an overview of the current body of research with particular focus on autologous ASCs and the laboratory expandable iPSCs in hope of shedding more light on this attractive therapy so as to elevate the efficacy of wound healing that is almost always compromised in diabetic patients.
Collapse
Affiliation(s)
- Meng-Chien Willie Hsieh
- Department of Surgery, Division of Plastic Surgery, Kaohsiung Medical University Hospital, Kaohsiung 80708, Taiwan
- Department of Plastic Surgery, Kaohsiung Municipal Ta-Tung Hospital, Kaohsiung 80145, Taiwan
| | - Wei-Ting Wang
- Department of Surgery, Division of Plastic Surgery, Kaohsiung Medical University Hospital, Kaohsiung 80708, Taiwan
| | - Chuang-Yu Lin
- Department of Biomedical Science and Environmental Biology, College of Life Science, Kaohsiung Medical University, Kaohsiung 80708, Taiwan
| | - Yur-Ren Kuo
- Department of Surgery, Division of Plastic Surgery, Kaohsiung Medical University Hospital, Kaohsiung 80708, Taiwan
- Department of Surgery, School of Medicine, Kaohsiung Medical University, Kaohsiung 80708, Taiwan
- Regenerative Medicine and Cell Therapy Research Center, Kaohsiung Medical University, Kaohsiung 80708, Taiwan
| | - Su-Shin Lee
- Department of Surgery, Division of Plastic Surgery, Kaohsiung Medical University Hospital, Kaohsiung 80708, Taiwan
- Department of Surgery, School of Medicine, Kaohsiung Medical University, Kaohsiung 80708, Taiwan
- Regenerative Medicine and Cell Therapy Research Center, Kaohsiung Medical University, Kaohsiung 80708, Taiwan
- Department of Surgery, Kaohsiung Municipal Siaogang Hospital, Kaohsiung 81267, Taiwan
| | - Ming-Feng Hou
- Department of Surgery, School of Medicine, Kaohsiung Medical University, Kaohsiung 80708, Taiwan
- Department of Surgery, Division of Breast Oncology and Surgery, Kaohsiung Medical University Hospital, Kaohsiung 80708, Taiwan
| | - Yi-Chia Wu
- Department of Surgery, Division of Plastic Surgery, Kaohsiung Medical University Hospital, Kaohsiung 80708, Taiwan
- Department of Plastic Surgery, Kaohsiung Municipal Ta-Tung Hospital, Kaohsiung 80145, Taiwan
- Department of Surgery, School of Medicine, Kaohsiung Medical University, Kaohsiung 80708, Taiwan
- Regenerative Medicine and Cell Therapy Research Center, Kaohsiung Medical University, Kaohsiung 80708, Taiwan
- Department of Surgery, Division of Breast Oncology and Surgery, Kaohsiung Medical University Hospital, Kaohsiung 80708, Taiwan
- Correspondence: ; Tel.: +886-7-312-1101 (ext. 7675)
| |
Collapse
|
14
|
Hasegawa T, Oka T, Demehri S. Alarmin Cytokines as Central Regulators of Cutaneous Immunity. Front Immunol 2022; 13:876515. [PMID: 35432341 PMCID: PMC9005840 DOI: 10.3389/fimmu.2022.876515] [Citation(s) in RCA: 24] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2022] [Accepted: 03/08/2022] [Indexed: 12/13/2022] Open
Abstract
Skin acts as the primary interface between the body and the environment. The skin immune system is composed of a complex network of immune cells and factors that provide the first line of defense against microbial pathogens and environmental insults. Alarmin cytokines mediate an intricate intercellular communication between keratinocytes and immune cells to regulate cutaneous immune responses. Proper functions of the type 2 alarmin cytokines, thymic stromal lymphopoietin (TSLP), interleukin (IL)-25, and IL-33, are paramount to the maintenance of skin homeostasis, and their dysregulation is commonly associated with allergic inflammation. In this review, we discuss recent findings on the complex regulatory network of type 2 alarmin cytokines that control skin immunity and highlight the mechanisms by which these cytokines regulate skin immune responses in host defense, chronic inflammation, and cancer.
Collapse
Affiliation(s)
| | - Tomonori Oka
- Center for Cancer Immunology and Cutaneous Biology Research Center, Department of Dermatology and Center for Cancer Research, Massachusetts General Hospital and Harvard Medical School, Boston, MA, United States
| | - Shadmehr Demehri
- Center for Cancer Immunology and Cutaneous Biology Research Center, Department of Dermatology and Center for Cancer Research, Massachusetts General Hospital and Harvard Medical School, Boston, MA, United States
| |
Collapse
|