1
|
Dolivo DM, Rodrigues AE, Mustoe TA, Galiano RD, Hong SJ. Ethacrynic Acid Mitigates Skin Fibrosis through Downregulation of S100 Family Damage-Associated Molecular Pattern Expression in the Epidermis. J Invest Dermatol 2025:S0022-202X(25)00396-3. [PMID: 40210113 DOI: 10.1016/j.jid.2025.03.029] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2024] [Revised: 03/07/2025] [Accepted: 03/23/2025] [Indexed: 04/12/2025]
Affiliation(s)
- David M Dolivo
- Department of Surgery, Northwestern University Feinberg School of Medicine, Chicago, Illinois, USA
| | - Adrian E Rodrigues
- Department of Surgery, Northwestern University Feinberg School of Medicine, Chicago, Illinois, USA
| | - Thomas A Mustoe
- Department of Surgery, Northwestern University Feinberg School of Medicine, Chicago, Illinois, USA
| | - Robert D Galiano
- Department of Surgery, Northwestern University Feinberg School of Medicine, Chicago, Illinois, USA.
| | - Seok Jong Hong
- Department of Surgery, Northwestern University Feinberg School of Medicine, Chicago, Illinois, USA.
| |
Collapse
|
2
|
Hahn JM, Combs KA, Phillips CM, Warner PM, Qazi UA, Powell HM, Supp DM. CYP24A1 is overexpressed in keloid keratinocytes and its inhibition alters profibrotic gene expression. BURNS & TRAUMA 2025; 13:tkae063. [PMID: 39822648 PMCID: PMC11736898 DOI: 10.1093/burnst/tkae063] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/24/2024] [Revised: 09/20/2024] [Accepted: 09/24/2024] [Indexed: 01/19/2025]
Abstract
Background Keloids are disfiguring, fibrotic scar-like lesions that are challenging to treat and commonly recur after therapy. A deeper understanding of the mechanisms driving keloid formation is necessary for the development of more effective therapies. Reduced vitamin D receptor (VDR) expression has been observed in keloids, implicating vitamin D signaling in keloid pathology. Vitamin D exhibits anti-proliferative and anti-inflammatory properties, suggesting it could have therapeutic utility in keloid disorder. The current study investigated vitamin D-regulated gene expression in keloid keratinocytes and the effects of inhibiting an enzyme involved in vitamin D metabolism on the phenotype of keloid-derived keratinocytes. Methods Normal and keloid-derived primary keratinocytes were isolated from normal skin and keloid lesions, respectively, and were cultured in the absence or presence of vitamin D. In some experiments, inhibitors of the vitamin D metabolizing enzyme CYP24A1, ketoconazole or VID400 were added in the absence or presence of vitamin D. Cellular proliferation, migration and gene expression were measured. Results We observed significant overexpression of CYP24A1 mRNA in keloid versus normal keratinocytes and increased CYP24A1 protein levels in keloids versus normal skin. CYP24A1 encodes 24 hydroxylase and is induced by vitamin D in a feedback loop that regulates vitamin D levels; thus, inhibition of CYP24A1 activity may locally increase active vitamin D levels. Ketoconazole, a non-specific cytochrome P-450 inhibitor, reduced proliferation of keloid and normal keratinocytes, but VID400, a specific CYP24A1 inhibitor, only significantly affected keloid keratinocyte proliferation. Neither inhibitor significantly reduced keratinocyte migration. The two inhibitors had different effects on vitamin D target gene expression in keratinocytes. Specifically, ketoconazole treatment reduced CYP24A1 expression in normal and keloid keratinocytes, whereas VID400 increased CYP24A1 expression. Both inhibitors decreased expression of profibrotic genes, including periostin and hyaluronan synthase 2, in keloid-derived cells. Combined treatment of keloid keratinocytes with vitamin D and ketoconazole or VID400 increased the effects of vitamin D treatment on target genes, although the effects were gene- and cell type-specific. Conclusions The data suggest that reduction of vitamin D inactivation with CYP24A1 inhibitors may reduce profibrotic gene expression in keloid-derived cells. Therefore, CYP24A1 inhibitors may serve as adjunctive therapies to suppress keloid-associated gene expression changes.
Collapse
Affiliation(s)
- Jennifer M Hahn
- Department of Surgery, University of Cincinnati College of Medicine, 231 Albert Sabin Way, Cincinnati, OH, 45267, USA
| | - Kelly A Combs
- Department of Surgery, University of Cincinnati College of Medicine, 231 Albert Sabin Way, Cincinnati, OH, 45267, USA
| | - Caitlin M Phillips
- Clinical Research Department, Shriners Children’s Ohio, One Children's Plaza - 2 West, Dayton, OH, 45404, USA
| | - Petra M Warner
- Department of Surgery, University of Cincinnati College of Medicine, 231 Albert Sabin Way, Cincinnati, OH, 45267, USA
| | - Uzair A Qazi
- Department of Surgery, University of Cincinnati College of Medicine, 231 Albert Sabin Way, Cincinnati, OH, 45267, USA
| | - Heather M Powell
- Clinical Research Department, Shriners Children’s Ohio, One Children's Plaza - 2 West, Dayton, OH, 45404, USA
- Department of Materials Science and Engineering, The Ohio State University, 140 W. 19th Avenue, Columbus, OH, 43210, USA
- Department of Biomedical Engineering, The Ohio State University, 140 W. 19th Avenue, Columbus, OH, 43210, USA
| | - Dorothy M Supp
- Department of Surgery, University of Cincinnati College of Medicine, 231 Albert Sabin Way, Cincinnati, OH, 45267, USA
- Clinical Research Department, Shriners Children’s Ohio, One Children's Plaza - 2 West, Dayton, OH, 45404, USA
- Center for Stem Cell and Organoid Medicine (CuSTOM), Cincinnati Children’s Hospital Medical Center, 3333 Burnet Avenue, Cincinnati, OH, 45229, USA
| |
Collapse
|
3
|
Simmons J, Gallo RL. The Central Roles of Keratinocytes in Coordinating Skin Immunity. J Invest Dermatol 2024; 144:2377-2398. [PMID: 39115524 PMCID: PMC11920965 DOI: 10.1016/j.jid.2024.06.1280] [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: 11/27/2023] [Revised: 06/06/2024] [Accepted: 06/14/2024] [Indexed: 10/25/2024]
Abstract
The function of keratinocytes (KCs) to form a barrier and produce cytokines is well-known, but recent progress has revealed many different roles for KCs in regulation of skin immunity. In this review, we provide an update on the current understanding of how KCs communicate with microbes, immunocytes, neurons, and other cells to form an effective immune barrier. We catalog the large list of genes and metabolites of KCs that participate in host defense and discuss the mechanisms of immune crosstalk, addressing how KCs simultaneously form a physical barrier, communicate with fibroblasts, and control immune signals. Overall, the signals sent and received by KCs are an exciting group of therapeutic targets to explore in the treatment of dermatologic disorders.
Collapse
Affiliation(s)
- Jared Simmons
- Department of Dermatology, School of Medicine, University of California San Diego, La Jolla, California, USA
| | - Richard L Gallo
- Department of Dermatology, School of Medicine, University of California San Diego, La Jolla, California, USA.
| |
Collapse
|
4
|
Chang N, Liu Y, Li W, Ma Y, Zhou X, Zhao X, Yang L, Li L. Neutrophil-secreted S100A8/A9 participates in fatty liver injury and fibrosis by promoting myofibroblast migration. J Mol Med (Berl) 2024; 102:1117-1133. [PMID: 38995368 DOI: 10.1007/s00109-024-02469-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2023] [Revised: 06/17/2024] [Accepted: 07/04/2024] [Indexed: 07/13/2024]
Abstract
Fatty liver, which is induced by abnormal lipid metabolism, is one of the most common causes of chronic liver disease globally and causes liver fibrosis. During this process, bone marrow-derived mesenchymal stromal cells (BMSCs) and hepatic stellate cells (HSCs) migrate toward the injured liver and participate in fibrogenesis by transdifferentiating into myofibroblasts. S100A8/A9 is a powerful inducer of cell migration and is involved in liver injury. But there are few reports about the effects of S100A8/A9 on BMSC/HSC migration. In the current study, we found that S100A8/A9 expression was increased during fatty liver injury/fibrogenesis. Moreover, S100A8/A9 expression had a positive correlation with fibrosis marker gene expressions in the injured liver. S100A8/A9 was mainly produced by neutrophils in the fibrotic liver. In vitro, neutrophil-secreted S100A8/A9 promoted BMSC/HSC migration via remodeling of microfilaments. Using specific siRNA and inhibitor, we proved that S100A8/A9-induced BMSC/HSC migration is dependent on TLR4/Rho GTPases signaling. Moreover, S100A8/A9 knock-down alleviated liver injury and fibrogenesis in vivo, while injection of S100A9 neutralizing antibody performed similar roles. We proved that S100A8/A9 was involved in liver injury and fibrogenesis via inducing BMSC/HSC migration. Our research reveals a new mechanism underlying BMSC/HSC migration in liver fibrosis and suggests S100A8/A9 as a potential therapeutic target of liver fibrosis. KEY MESSAGES: S100A8/A9 is secreted by neutrophils and increased in fatty liver injury. Neutrophil-secreted S100A8/A9 is a mediator of BMSC/HSC migration in vitro. S100A8/A9-induced BMSC/HSC migration is dependent on TLR4/Rho GTPases signaling. S100A8/A9 blockade alleviates liver injury and fibrogenesis in vivo.
Collapse
Affiliation(s)
- Na Chang
- Department of Cell Biology, Laboratory for Clinical Medicine, Municipal Laboratory for Liver Protection and Regulation of Regeneration, Capital Medical University, No. 10 Xitoutiao, You An Men, Beijing, 100069, China
| | - Yuran Liu
- Department of Cell Biology, Laboratory for Clinical Medicine, Municipal Laboratory for Liver Protection and Regulation of Regeneration, Capital Medical University, No. 10 Xitoutiao, You An Men, Beijing, 100069, China
| | - Weiyang Li
- Department of Cell Biology, Laboratory for Clinical Medicine, Municipal Laboratory for Liver Protection and Regulation of Regeneration, Capital Medical University, No. 10 Xitoutiao, You An Men, Beijing, 100069, China
| | - Yuehan Ma
- Department of Cell Biology, Laboratory for Clinical Medicine, Municipal Laboratory for Liver Protection and Regulation of Regeneration, Capital Medical University, No. 10 Xitoutiao, You An Men, Beijing, 100069, China
| | - Xuan Zhou
- Department of Cell Biology, Laboratory for Clinical Medicine, Municipal Laboratory for Liver Protection and Regulation of Regeneration, Capital Medical University, No. 10 Xitoutiao, You An Men, Beijing, 100069, China
| | - Xinhao Zhao
- Department of Cell Biology, Laboratory for Clinical Medicine, Municipal Laboratory for Liver Protection and Regulation of Regeneration, Capital Medical University, No. 10 Xitoutiao, You An Men, Beijing, 100069, China
| | - Lin Yang
- Department of Cell Biology, Laboratory for Clinical Medicine, Municipal Laboratory for Liver Protection and Regulation of Regeneration, Capital Medical University, No. 10 Xitoutiao, You An Men, Beijing, 100069, China
| | - Liying Li
- Department of Cell Biology, Laboratory for Clinical Medicine, Municipal Laboratory for Liver Protection and Regulation of Regeneration, Capital Medical University, No. 10 Xitoutiao, You An Men, Beijing, 100069, China.
| |
Collapse
|
5
|
Chen S, Xiong Y, Yang F, Hu Y, Feng J, Zhou F, Liu Z, Liu H, Liu X, Zhao J, Zhang Z, Chen L. Approaches to scarless burn wound healing: application of 3D printed skin substitutes with dual properties of anti-infection and balancing wound hydration levels. EBioMedicine 2024; 106:105258. [PMID: 39068733 PMCID: PMC11332815 DOI: 10.1016/j.ebiom.2024.105258] [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: 03/04/2024] [Revised: 07/09/2024] [Accepted: 07/09/2024] [Indexed: 07/30/2024] Open
Abstract
BACKGROUND Severe burn wounds face two primary challenges: dysregulated cellular impairment functions following infection and an unbalanced wound hydration microenvironment leading to excessive inflammation and collagen deposition. These results in hypertrophic scar contraction, causing significant deformity and disability in survivors. METHODS A three-dimensional (3D) printed double-layer hydrogel (DLH) was designed and fabricated to address the problem of scar formation after burn injury. DLH was developed using methacrylated silk fibroin (SFMA) and gelatin methacryloyl (GelMA) for the upper layer, and GelMA and hyaluronic acid methacryloyl (HAMA) for the lower layer. To combat infection, copper-epigallocatechin gallate (Cu-EGCG) was incorporated into the lower layer bioink, collectively referred to as DLS. To balance wound hydration levels, HaCaT cells were additionally encapsulated in the upper layer, designed as DLS/c. FINDINGS DLH demonstrated suitable porosity, appropriate mechanical properties, and excellent biocompatibility. DLS exhibited potent antimicrobial properties, exerted anti-inflammatory effects by regulating macrophage polarisation, and may enhance angiogenesis through the HIF-1α/VEGF pathway. In the DLS/c group, animal studies showed significant improvements in epidermal formation, barrier function, and epidermal hydration, accompanied by reduced inflammation. In addition, Masson's trichrome and Sirius red staining revealed that the structure and ratio of dermal collagen in DLS/c resembled that of normal skin, indicating considerable potential for scarless wound healing. INTERPRETATION This biomimetic matrix shows promise in addressing the challenges of burn wounds and aiming for scarless repair, with benefits such as anti-infection, epidermal hydration, biological induction, and optimised topological properties. FUNDING Shown in Acknowledgements.
Collapse
Affiliation(s)
- Shuying Chen
- Department of Burn, Wound Repair & Reconstruction, The First Affiliated Hospital of Sun Yat-sen University, Guangzhou 510080, China; Guangdong Provincial Engineering Technology Research Center of Burn and Wound Accurate Diagnosis and Treatment Key Technology and Series of Products, Sun Yat-sen University, Guangzhou 510080, China; Institute of Precision Medicine, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou 510080, China
| | - Yahui Xiong
- Department of Burn, Wound Repair & Reconstruction, The First Affiliated Hospital of Sun Yat-sen University, Guangzhou 510080, China; Guangdong Provincial Engineering Technology Research Center of Burn and Wound Accurate Diagnosis and Treatment Key Technology and Series of Products, Sun Yat-sen University, Guangzhou 510080, China; Institute of Precision Medicine, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou 510080, China
| | - Fan Yang
- Department of Burn, Wound Repair & Reconstruction, The First Affiliated Hospital of Sun Yat-sen University, Guangzhou 510080, China; Guangdong Provincial Engineering Technology Research Center of Burn and Wound Accurate Diagnosis and Treatment Key Technology and Series of Products, Sun Yat-sen University, Guangzhou 510080, China; Institute of Precision Medicine, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou 510080, China
| | - Yanke Hu
- Department of Burn, Wound Repair & Reconstruction, The First Affiliated Hospital of Sun Yat-sen University, Guangzhou 510080, China; Guangdong Provincial Engineering Technology Research Center of Burn and Wound Accurate Diagnosis and Treatment Key Technology and Series of Products, Sun Yat-sen University, Guangzhou 510080, China; Institute of Precision Medicine, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou 510080, China
| | - Jinghao Feng
- Guangzhou Panyu Central Hospital, Guangzhou 511400, China
| | - Fei Zhou
- Department of Burn, Wound Repair & Reconstruction, The First Affiliated Hospital of Sun Yat-sen University, Guangzhou 510080, China; Guangdong Provincial Engineering Technology Research Center of Burn and Wound Accurate Diagnosis and Treatment Key Technology and Series of Products, Sun Yat-sen University, Guangzhou 510080, China; Institute of Precision Medicine, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou 510080, China
| | - Zhonghua Liu
- South China Agricultural University, Guangzhou 510642, China
| | - Hengdeng Liu
- Department of Burn, Wound Repair & Reconstruction, The First Affiliated Hospital of Sun Yat-sen University, Guangzhou 510080, China; Guangdong Provincial Engineering Technology Research Center of Burn and Wound Accurate Diagnosis and Treatment Key Technology and Series of Products, Sun Yat-sen University, Guangzhou 510080, China; Institute of Precision Medicine, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou 510080, China
| | - Xiaogang Liu
- Department of Burn, Wound Repair & Reconstruction, The First Affiliated Hospital of Sun Yat-sen University, Guangzhou 510080, China; Guangdong Provincial Engineering Technology Research Center of Burn and Wound Accurate Diagnosis and Treatment Key Technology and Series of Products, Sun Yat-sen University, Guangzhou 510080, China; Institute of Precision Medicine, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou 510080, China
| | - Jingling Zhao
- Department of Burn, Wound Repair & Reconstruction, The First Affiliated Hospital of Sun Yat-sen University, Guangzhou 510080, China; Guangdong Provincial Engineering Technology Research Center of Burn and Wound Accurate Diagnosis and Treatment Key Technology and Series of Products, Sun Yat-sen University, Guangzhou 510080, China; Institute of Precision Medicine, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou 510080, China
| | - Zhaoqiang Zhang
- Department of Oral and Maxillofacial Surgery, Stomatological Hospital, School of Stomatology, Southern Medical University, Guangzhou 510280, China.
| | - Lei Chen
- Department of Burn, Wound Repair & Reconstruction, The First Affiliated Hospital of Sun Yat-sen University, Guangzhou 510080, China; Guangdong Provincial Engineering Technology Research Center of Burn and Wound Accurate Diagnosis and Treatment Key Technology and Series of Products, Sun Yat-sen University, Guangzhou 510080, China; Institute of Precision Medicine, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou 510080, China.
| |
Collapse
|
6
|
Mazio C, Scognamiglio LS, Casale C, Panzetta V, Urciuolo F, Galietta LJV, Imparato G, Netti PA. A functional 3D full-thickness model for comprehending the interaction between airway epithelium and connective tissue in cystic fibrosis. Biomaterials 2024; 308:122546. [PMID: 38552367 DOI: 10.1016/j.biomaterials.2024.122546] [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: 11/07/2023] [Revised: 02/22/2024] [Accepted: 03/20/2024] [Indexed: 05/03/2024]
Abstract
Patients with cystic fibrosis (CF) experience severe lung disease, including persistent infections, inflammation, and irreversible fibrotic remodeling of the airways. Although therapy with transmembrane conductance regulator (CFTR) protein modulators reached optimal results in terms of CFTR rescue, lung transplant remains the best line of care for patients in an advanced stage of CF. Indeed, chronic inflammation and tissue remodeling still represent stumbling blocks during treatment, and underlying mechanisms are still unclear. Nowadays, animal models are not able to fully replicate clinical features of the human disease and the conventional in vitro models lack a stromal compartment undergoing fibrotic remodeling. To address this gap, we show the development of a 3D full-thickness model of CF with a human bronchial epithelium differentiated on a connective airway tissue. We demonstrated that the epithelial cells not only underwent mucociliary differentiation but also migrated in the connective tissue and formed gland-like structures. The presence of the connective tissue stimulated the pro-inflammatory behaviour of the epithelium, which activated the fibroblasts embedded into their own extracellular matrix (ECM). By varying the composition of the model with CF epithelial cells and a CF or healthy connective tissue, it was possible to replicate different moments of CF disease, as demonstrated by the differences in the transcriptome of the CF epithelium in the different conditions. The possibility to faithfully represent the crosstalk between epithelial and connective in CF through the full thickness model, along with inflammation and stromal activation, makes the model suitable to better understand mechanisms of disease genesis, progression, and response to therapy.
Collapse
Affiliation(s)
- Claudia Mazio
- Istituto Italiano di Tecnologia-IIT, Center for Advanced Biomaterials for Healthcare, Largo Barsanti e Matteucci 53, 80125, Napoli, Italy
| | - Laura Sara Scognamiglio
- Istituto Italiano di Tecnologia-IIT, Center for Advanced Biomaterials for Healthcare, Largo Barsanti e Matteucci 53, 80125, Napoli, Italy
| | - Costantino Casale
- Interdisciplinary Research Centre on Biomaterials-CRIB, University of Napoli Federico II, P.le Tecchio 80, 80125, Napoli, Italy
| | - Valeria Panzetta
- Interdisciplinary Research Centre on Biomaterials-CRIB, University of Napoli Federico II, P.le Tecchio 80, 80125, Napoli, Italy; Department of Chemical, Materials and Industrial Production Engineering-DICMAPI, University of Naples Federico II, P.le Tecchio 80, 80125, Naples, Italy
| | - Francesco Urciuolo
- Interdisciplinary Research Centre on Biomaterials-CRIB, University of Napoli Federico II, P.le Tecchio 80, 80125, Napoli, Italy; Department of Chemical, Materials and Industrial Production Engineering-DICMAPI, University of Naples Federico II, P.le Tecchio 80, 80125, Naples, Italy
| | - Luis J V Galietta
- Telethon Institute of Genetics and Medicine-TIGEM, Via Campi Flegrei 34, 80078, Pozzuoli, NA, Italy
| | - Giorgia Imparato
- Istituto Italiano di Tecnologia-IIT, Center for Advanced Biomaterials for Healthcare, Largo Barsanti e Matteucci 53, 80125, Napoli, Italy.
| | - Paolo A Netti
- Istituto Italiano di Tecnologia-IIT, Center for Advanced Biomaterials for Healthcare, Largo Barsanti e Matteucci 53, 80125, Napoli, Italy; Interdisciplinary Research Centre on Biomaterials-CRIB, University of Napoli Federico II, P.le Tecchio 80, 80125, Napoli, Italy; Department of Chemical, Materials and Industrial Production Engineering-DICMAPI, University of Naples Federico II, P.le Tecchio 80, 80125, Naples, Italy
| |
Collapse
|
7
|
Martin L, Simpson K, Brzezinski M, Watt J, Xu W. Cellular response of keratinocytes to the entry and accumulation of nanoplastic particles. Part Fibre Toxicol 2024; 21:22. [PMID: 38685063 PMCID: PMC11057139 DOI: 10.1186/s12989-024-00583-9] [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: 09/18/2023] [Accepted: 04/14/2024] [Indexed: 05/02/2024] Open
Abstract
Plastic accumulation in the environment is rapidly increasing, and nanoplastics (NP), byproducts of environmental weathering of bulk plastic waste, pose a significant public health risk. Particles may enter the human body through many possible routes such as ingestion, inhalation, and skin absorption. However, studies on NP penetration and accumulation in human skin are limited. Loss or reduction of the keratinized skin barrier may enhance the skin penetration of NPs. The present study investigated the entry of NPs into a human skin system modeling skin with compromised barrier functions and cellular responses to the intracellular accumulations of NPs. Two in vitro models were employed to simulate human skin lacking keratinized barriers. The first model was an ex vivo human skin culture with the keratinized dermal layer (stratum corneum) removed. The second model was a 3D keratinocyte/dermal fibroblast cell co-culture model with stratified keratinocytes on the top and a monolayer of skin fibroblast cells co-cultured at the bottom. The penetration and accumulation of the NPs in different cell types were observed using fluorescent microscopy, confocal microscopy, and cryogenic electron microscopy (cryo-EM). The cellular responses of keratinocytes and dermal fibroblast cells to stress induced by NPs stress were measured. The genetic regulatory pathway of keratinocytes to the intracellular NPs was identified using transcript analyses and KEGG pathway analysis. The cellular uptake of NPs by skin cells was confirmed by imaging analyses. Transepidermal transport and penetration of NPs through the skin epidermis were observed. According to the gene expression and pathway analyses, an IL-17 signaling pathway was identified as the trigger for cellular responses to internal NP accumulation in the keratinocytes. The transepidermal NPs were also found in co-cultured dermal fibroblast cells and resulted in a large-scale transition from fibroblast cells to myofibroblast cells with enhanced production of α-smooth muscle actin and pro-Collagen Ia. The upregulation of inflammatory factors and cell activation may result in skin inflammation and ultimately trigger immune responses.
Collapse
Affiliation(s)
- Leisha Martin
- Department of Life Sciences, College of Science, Texas A&M University - Corpus Christi, 6300 Ocean Dr, 78412, Corpus Christi, TX, USA
| | - Kayla Simpson
- Department of Life Sciences, College of Science, Texas A&M University - Corpus Christi, 6300 Ocean Dr, 78412, Corpus Christi, TX, USA
| | - Molly Brzezinski
- Department of Life Sciences, College of Science, Texas A&M University - Corpus Christi, 6300 Ocean Dr, 78412, Corpus Christi, TX, USA
| | - John Watt
- Center for Integrated Nanotechnologies, Los Alamos National Laboratory, Albuquerque, NM, USA
| | - Wei Xu
- Department of Life Sciences, College of Science, Texas A&M University - Corpus Christi, 6300 Ocean Dr, 78412, Corpus Christi, TX, USA.
| |
Collapse
|
8
|
Paramasivam S, Perumal SS, Ekambaram SP. Computational Deciphering of the Role of S100A8 and S100A9 Proteins and Their Changes in the Structure Assembly Influences Their Interaction with TLR4, RAGE, and CD36. Protein J 2024; 43:243-258. [PMID: 38431537 DOI: 10.1007/s10930-024-10186-0] [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] [Accepted: 02/20/2024] [Indexed: 03/05/2024]
Abstract
S100A8 and S100A9 belong to the calcium-binding, damage associated molecular pattern (DAMP) proteins shown to aggravate the pathogenesis of rheumatoid arthritis (RA) through their interaction with the TLR4, RAGE and CD36 receptors. S100A8 and S100A9 proteins tend to exist in monomeric, homo and heterodimeric forms, which have been implicated in the pathogenesis of RA, via interacting with Pattern Recognition receptors (PRRs). The study aims to assess the influence of changes in the structure and biological assembly of S100A8 and S100A9 proteins as well as their interaction with significant receptors in RA through computational methods and surface plasmon resonance (SPR) analysis. Molecular docking analysis revealed that the S100A9 homodimer and S100A8/A9 heterodimer showed higher binding affinity towards the target receptors. Most S100 proteins showed good binding affinity towards TLR4 compared to other receptors. Based on the 50 ns MD simulations, TLR4, RAGE, and CD36 formed stable complexes with the monomeric and dimeric forms of S100A8 and S100A9 proteins. However, SPR analysis showed that the S100A8/A9 heterodimers formed stable complexes and exhibited high binding affinity towards the receptors. SPR data also indicated that TLR4 and its interactions with S100A8/A9 proteins may play a primary role in the pathogenesis of RA, with additional contributions from CD36 and RAGE interactions. Subsequent in vitro and in vivo investigations are warranted to corroborate the involvement of S100A8/A9 and the expression of TLR4, RAGE, and CD36 in the pathophysiology of RA.
Collapse
Affiliation(s)
- Sivasakthi Paramasivam
- Department of Pharmaceutical Technology, Bharathidasan Institute of Technology Campus, University College of Engineering, Anna University, Tiruchirappalli, Tamil Nadu, 620 024, India
| | - Senthamil Selvan Perumal
- Department of Pharmaceutical Technology, Bharathidasan Institute of Technology Campus, University College of Engineering, Anna University, Tiruchirappalli, Tamil Nadu, 620 024, India
| | - Sanmuga Priya Ekambaram
- Department of Pharmaceutical Technology, Bharathidasan Institute of Technology Campus, University College of Engineering, Anna University, Tiruchirappalli, Tamil Nadu, 620 024, India.
| |
Collapse
|
9
|
Zhang X, Han P, Qiu J, Huang F, Luo Q, Cheng J, Shan K, Yang Y, Zhang C. Single-cell RNA sequencing reveals the complex cellular niche of pterygium. Ocul Surf 2024; 32:91-103. [PMID: 38290663 DOI: 10.1016/j.jtos.2024.01.013] [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: 10/12/2023] [Revised: 01/26/2024] [Accepted: 01/26/2024] [Indexed: 02/01/2024]
Abstract
PURPOSE Pterygium is a vision-threatening conjunctival fibrovascular degenerated disease with a high global prevalence up to 12 %, while no absolute pharmacotherapy has been applied in clinics. In virtue of single-cell RNA sequencing (scRNA-seq) technique, our study investigated underlying pathogeneses and potential therapeutic targets of pterygium from the cellular transcriptional level. METHODS A total of 45605 cells from pterygium of patients and conjunctiva of normal controls (NC) were conducted with scRNA-seq, and then analyzed via integrated analysis, pathway enrichment, pseudotime trajectory, and cell-cell communications. Besides, immunofluorescence and western blot were performed in vivo and in vitro to verify our findings. RESULTS In brief, 9 major cellular types were defined, according to canonical markers. Subsequently, we further determined the subgroups of each major cell lineages. Several newly identified cell sub-clusters could promote pterygium, including immuno-fibroblasts, epithelial mesenchymal transition (EMT)-epithelial cells, and activated vascular endothelial cells (activated-vEndo). Besides, we also probed the enrichment of immune cells in pterygium. Particularly, macrophages, recruited by ACKR1+activated-vEndo, might play an important role in the development of pterygium by promoting angiogenesis, immune suppression, and inflammation. CONCLUSION An intricate cellular niche was revealed in pterygium via scRNA-seq analysis and the interactions between macrophages and ACKR1+ activated-vEndo might be the key part in the development of pterygia.
Collapse
Affiliation(s)
- Xueling Zhang
- Department of Ophthalmology, Eye Ear Nose and Throat Hospital of Fudan University, Shanghai, 200031, China; Department of Ophthalmology, Shanghai Medical College, Fudan University, Shanghai, 200031, China; NHC Key Laboratory of Myopia (Fudan University), Laboratory of Myopia, Chinese Academy of Medical Sciences, China
| | - Peizhen Han
- Department of Head and Neck Surgery, Fudan University Shanghai Cancer Center, Shanghai, China; Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China
| | - Jini Qiu
- Department of Ophthalmology, Eye Ear Nose and Throat Hospital of Fudan University, Shanghai, 200031, China; Department of Ophthalmology, Shanghai Medical College, Fudan University, Shanghai, 200031, China; NHC Key Laboratory of Myopia (Fudan University), Laboratory of Myopia, Chinese Academy of Medical Sciences, China
| | - Feifei Huang
- Department of Ophthalmology, Eye Ear Nose and Throat Hospital of Fudan University, Shanghai, 200031, China; Department of Ophthalmology, Shanghai Medical College, Fudan University, Shanghai, 200031, China; NHC Key Laboratory of Myopia (Fudan University), Laboratory of Myopia, Chinese Academy of Medical Sciences, China
| | - Qiting Luo
- Department of Ophthalmology, Eye Ear Nose and Throat Hospital of Fudan University, Shanghai, 200031, China; Department of Ophthalmology, Shanghai Medical College, Fudan University, Shanghai, 200031, China; NHC Key Laboratory of Myopia (Fudan University), Laboratory of Myopia, Chinese Academy of Medical Sciences, China
| | - Jingyi Cheng
- Department of Ophthalmology, Eye Ear Nose and Throat Hospital of Fudan University, Shanghai, 200031, China; Department of Ophthalmology, Shanghai Medical College, Fudan University, Shanghai, 200031, China; NHC Key Laboratory of Myopia (Fudan University), Laboratory of Myopia, Chinese Academy of Medical Sciences, China
| | - Kun Shan
- Department of Ophthalmology, Eye Ear Nose and Throat Hospital of Fudan University, Shanghai, 200031, China; Department of Ophthalmology, Shanghai Medical College, Fudan University, Shanghai, 200031, China; NHC Key Laboratory of Myopia (Fudan University), Laboratory of Myopia, Chinese Academy of Medical Sciences, China.
| | - Yujing Yang
- Department of Ophthalmology, Eye Ear Nose and Throat Hospital of Fudan University, Shanghai, 200031, China; Department of Ophthalmology, Shanghai Medical College, Fudan University, Shanghai, 200031, China; NHC Key Laboratory of Myopia (Fudan University), Laboratory of Myopia, Chinese Academy of Medical Sciences, China.
| | - Chaoran Zhang
- Department of Ophthalmology, Eye Ear Nose and Throat Hospital of Fudan University, Shanghai, 200031, China; Department of Ophthalmology, Shanghai Medical College, Fudan University, Shanghai, 200031, China; NHC Key Laboratory of Myopia (Fudan University), Laboratory of Myopia, Chinese Academy of Medical Sciences, China.
| |
Collapse
|
10
|
Ding D, Luan R, Xue Q, Yang J. Prognostic significance of peripheral blood S100A12, S100A8, and S100A9 concentrations in idiopathic pulmonary fibrosis. Cytokine 2023; 172:156387. [PMID: 37826869 DOI: 10.1016/j.cyto.2023.156387] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2023] [Revised: 09/19/2023] [Accepted: 09/29/2023] [Indexed: 10/14/2023]
Abstract
BACKGROUND S100A12, S100A8, and S100A9 are inflammatory disease biomarkers whose functional significance in idiopathic pulmonary fibrosis (IPF) remains unclear. We evaluated the significance of S100A12, S100A8, and S100A9 levels in IPF development and prognosis. METHODS The dataset was collected from the Gene Expression Omnibus (GEO) database and differentially expressed genes were screened using GEO2R. We conducted a retrospective study of 106 patients with IPF to explore the relationships between different biomarkers and poor outcomes. Pearson's correlation coefficient, Kaplan-Meier, Cox regression, and functional enrichment analyses were used to evaluate relationships between these biomarkers' levels and clinical parameters or prognosis. RESULTS Serum levels of S100A12, S100A8, and S100A9 were significantly elevated in patients with IPF. The two most significant co-expression genes of S100A12 were S100A8 and S100A9. Patients with levels of S100A12 (median 231.21 ng/mL), S100A9 (median 57.09 ng/mL) or S100A8 (median 52.20 ng/mL), as well as combined elevated S100A12, S100A9, and S100A8 levels, exhibited shorter progression-free survival and overall survival. Serum S100A12 and S100A8, S100A12 and S100A9, S100A9 and S100A8 concentrations also displayed a strong positive correlation (rs2 = 0.4558, rs2 = 0.4558, rs2 = 0.6373; P < 0.001). S100A12 and S100A8/9 concentrations were independent of FVC%, DLCO%, and other clinical parameters (age, laboratory test data, and smoking habit). Finally, in multivariate analysis, the serum levels of S100A12, S100A8, and S100A9 were significant prognostic factors (hazard ratio 1.002, P = 0.032, hazard ratio 1.039, P = 0.001, and hazard ratio 1.048, P = 0.003). CONCLUSIONS S100A12, S100A8, and S100A9 are promising circulating biomarkers that may aid in determining IPF patient prognosis. Multicenter clinical trials are needed to confirm their clinical value.
Collapse
Affiliation(s)
- Dongyan Ding
- Department of Respiratory Medicine, The Second Hospital of Jilin University, Changchun, China
| | - Rumei Luan
- Department of Respiratory Medicine, The Second Hospital of Jilin University, Changchun, China
| | - Qianfei Xue
- Department of Respiratory Medicine, The University Hospital of Jilin University, Changchun, China
| | - Junling Yang
- Department of Respiratory Medicine, The Second Hospital of Jilin University, Changchun, China.
| |
Collapse
|
11
|
Zhang Y, Song Y, Du J, Liu W, Dong C, Huang Z, Zhang Z, Yang L, Wang T, Xiong S, Dong L, Guo Y, Dang J, He Q, Yu Z, Ma X. S100 calcium-binding protein A9 promotes skin regeneration through toll-like receptor 4 during tissue expansion. BURNS & TRAUMA 2023; 11:tkad030. [PMID: 37936894 PMCID: PMC10627002 DOI: 10.1093/burnst/tkad030] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 08/21/2022] [Revised: 02/17/2023] [Indexed: 11/09/2023]
Abstract
Background In plastic surgery, tissue expansion is widely used for repairing skin defects. However, low expansion efficiency and skin rupture caused by thin, expanded skin remain significant challenges in promoting skin regeneration during expansion. S100 calcium-binding protein A9 (S100A9) is essential in promoting wound healing; however, its effects on skin regeneration during tissue expansion remain unclear. The aim of the present study was to explore the role of S100A9 in skin regeneration, particularly collagen production to investigate its importance in skin regeneration during tissue expansion. Methods The expression and distribution of S100A9 and its receptors-toll-like receptor 4 (TLR-4) and receptor for advanced glycation end products were studied in expanded skin. These characteristics were investigated in skin samples of rats and patients. Moreover, the expression of S100A9 was investigated in stretched keratinocytes in vitro. The effects of S100A9 on the proliferation and migration of skin fibroblasts were also observed. TAK-242 was used to inhibit the binding of S100A9 to TLR-4; the levels of collagen I (COL I), transforming growth factor beta (TGF-β), TLR-4 and phospho-extracellular signal-related kinase 1/2 (p-ERK1/2) in fibroblasts were determined. Furthermore, fibroblasts were co-cultured with stretched S100A9-knockout keratinocytes by siRNA transfection and the levels of COL I, TGF-β, TLR-4 and p-ERK1/2 in fibroblasts were investigated. Additionally, the area of expanded skin, thickness of the dermis, and synthesis of COL I, TGF-β, TLR-4 and p-ERK1/2 were analysed to determine the effects of S100A9 on expanded skin. Results Increased expression of S100A9 and TLR-4 was associated with decreased extracellular matrix (ECM) in the expanded dermis. Furthermore, S100A9 facilitated the proliferation and migration of human skin fibroblasts as well as the expression of COL I and TGF-β in fibroblasts via the TLR-4/ERK1/2 pathway. We found that mechanical stretch-induced S100A9 expression and secretion of keratinocytes stimulated COL I, TGF-β, TLR-4 and p-ERK1/2 expression in skin fibroblasts. Recombined S100A9 protein aided expanded skin regeneration and rescued dermal thinning in rats in vivo as well as increasing ECM deposition during expansion. Conclusions These findings demonstrate that mechanical stretch promoted expanded skin regeneration by upregulating S100A9 expression. Our study laid the foundation for clinically improving tissue expansion using S100A9.
Collapse
Affiliation(s)
- Yu Zhang
- Department of Plastic Surgery, Xijing Hospital, Fourth Military Medical University, No.127 Changle West Road, Xi’an, Shaanxi Province 710032, China
| | - Yajuan Song
- Department of Plastic Surgery, Xijing Hospital, Fourth Military Medical University, No.127 Changle West Road, Xi’an, Shaanxi Province 710032, China
| | - Jing Du
- Department of Plastic Surgery, Xijing Hospital, Fourth Military Medical University, No.127 Changle West Road, Xi’an, Shaanxi Province 710032, China
| | - Wei Liu
- Department of Plastic Surgery, Xijing Hospital, Fourth Military Medical University, No.127 Changle West Road, Xi’an, Shaanxi Province 710032, China
| | - Chen Dong
- Department of Plastic Surgery, Xijing Hospital, Fourth Military Medical University, No.127 Changle West Road, Xi’an, Shaanxi Province 710032, China
| | - Zhaosong Huang
- Department of Plastic Surgery, Xijing Hospital, Fourth Military Medical University, No.127 Changle West Road, Xi’an, Shaanxi Province 710032, China
| | - Zhe Zhang
- Department of Plastic Surgery, Xijing Hospital, Fourth Military Medical University, No.127 Changle West Road, Xi’an, Shaanxi Province 710032, China
| | - Liu Yang
- Department of Plastic Surgery, Xijing Hospital, Fourth Military Medical University, No.127 Changle West Road, Xi’an, Shaanxi Province 710032, China
| | - Tong Wang
- Department of Plastic Surgery, Xijing Hospital, Fourth Military Medical University, No.127 Changle West Road, Xi’an, Shaanxi Province 710032, China
| | - Shaoheng Xiong
- Department of Plastic Surgery, Xijing Hospital, Fourth Military Medical University, No.127 Changle West Road, Xi’an, Shaanxi Province 710032, China
| | - Liwei Dong
- Department of Plastic Surgery, Xijing Hospital, Fourth Military Medical University, No.127 Changle West Road, Xi’an, Shaanxi Province 710032, China
| | - Yaotao Guo
- Department of Plastic Surgery, Xijing Hospital, Fourth Military Medical University, No.127 Changle West Road, Xi’an, Shaanxi Province 710032, China
| | - Juanli Dang
- Department of Plastic Surgery, Xijing Hospital, Fourth Military Medical University, No.127 Changle West Road, Xi’an, Shaanxi Province 710032, China
| | - Qiang He
- Department of Plastic Surgery, Xijing Hospital, Fourth Military Medical University, No.127 Changle West Road, Xi’an, Shaanxi Province 710032, China
| | - Zhou Yu
- Department of Plastic Surgery, Xijing Hospital, Fourth Military Medical University, No.127 Changle West Road, Xi’an, Shaanxi Province 710032, China
| | - Xianjie Ma
- Department of Plastic Surgery, Xijing Hospital, Fourth Military Medical University, No.127 Changle West Road, Xi’an, Shaanxi Province 710032, China
| |
Collapse
|
12
|
Sänger CS, Cernakova M, Wietecha MS, Garau Paganella L, Labouesse C, Dudaryeva OY, Roubaty C, Stumpe M, Mazza E, Tibbitt MW, Dengjel J, Werner S. Serine protease 35 regulates the fibroblast matrisome in response to hyperosmotic stress. SCIENCE ADVANCES 2023; 9:eadh9219. [PMID: 37647410 PMCID: PMC10468140 DOI: 10.1126/sciadv.adh9219] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/24/2023] [Accepted: 07/28/2023] [Indexed: 09/01/2023]
Abstract
Hyperosmotic stress occurs in several diseases, but its long-term effects are largely unknown. We used sorbitol-treated human fibroblasts in 3D culture to study the consequences of hyperosmotic stress in the skin. Sorbitol regulated many genes, which help cells cope with the stress condition. The most robustly regulated gene encodes serine protease 35 (PRSS35). Its regulation by hyperosmotic stress was dependent on the kinases p38 and JNK and the transcription factors NFAT5 and ATF2. We identified different collagens and collagen-associated proteins as putative PRSS35 binding partners. This is functionally important because PRSS35 affected the extracellular matrix proteome, which limited cell proliferation. The in vivo relevance of these findings is reflected by the coexpression of PRSS35 and its binding partners in human skin wounds, where hyperosmotic stress occurs as a consequence of excessive water loss. These results identify PRSS35 as a key regulator of the matrisome under hyperosmotic stress conditions.
Collapse
Affiliation(s)
- Catharina S. Sänger
- Institute of Molecular Health Sciences, Department of Biology, ETH Zurich, Otto-Stern-Weg 7, 8093 Zurich, Switzerland
| | - Martina Cernakova
- Institute of Molecular Health Sciences, Department of Biology, ETH Zurich, Otto-Stern-Weg 7, 8093 Zurich, Switzerland
| | - Mateusz S. Wietecha
- Institute of Molecular Health Sciences, Department of Biology, ETH Zurich, Otto-Stern-Weg 7, 8093 Zurich, Switzerland
- Department of Oral Biology, College of Dentistry, University of Illinois Chicago, Chicago, IL 60612, USA
| | - Lorenza Garau Paganella
- Institute for Mechanical Systems, Department of Mechanical and Process Engineering, ETH Zurich, Leonhardstrasse 21, 8092 Zurich, Switzerland
- Institute for Energy and Process Engineering, Department of Mechanical and Process Engineering, ETH Zurich, Sonneggstrasse 3, 8092 Zurich, Switzerland
| | - Céline Labouesse
- Institute for Energy and Process Engineering, Department of Mechanical and Process Engineering, ETH Zurich, Sonneggstrasse 3, 8092 Zurich, Switzerland
| | - Oksana Y. Dudaryeva
- Institute for Energy and Process Engineering, Department of Mechanical and Process Engineering, ETH Zurich, Sonneggstrasse 3, 8092 Zurich, Switzerland
| | - Carole Roubaty
- Faculty of Science and Medicine, Department of Biology, University of Fribourg, Ch. du Musée 10, 1700 Fribourg, Switzerland
| | - Michael Stumpe
- Faculty of Science and Medicine, Department of Biology, University of Fribourg, Ch. du Musée 10, 1700 Fribourg, Switzerland
| | - Edoardo Mazza
- Institute for Mechanical Systems, Department of Mechanical and Process Engineering, ETH Zurich, Leonhardstrasse 21, 8092 Zurich, Switzerland
| | - Mark W. Tibbitt
- Institute for Energy and Process Engineering, Department of Mechanical and Process Engineering, ETH Zurich, Sonneggstrasse 3, 8092 Zurich, Switzerland
| | - Jörn Dengjel
- Faculty of Science and Medicine, Department of Biology, University of Fribourg, Ch. du Musée 10, 1700 Fribourg, Switzerland
| | - Sabine Werner
- Institute of Molecular Health Sciences, Department of Biology, ETH Zurich, Otto-Stern-Weg 7, 8093 Zurich, Switzerland
| |
Collapse
|
13
|
Liu B, Kong Y, Alimi OA, Kuss MA, Tu H, Hu W, Rafay A, Vikas K, Shi W, Lerner M, Berry WL, Li Y, Carlson MA, Duan B. Multifunctional Microgel-Based Cream Hydrogels for Postoperative Abdominal Adhesion Prevention. ACS NANO 2023; 17:3847-3864. [PMID: 36779870 PMCID: PMC10820954 DOI: 10.1021/acsnano.2c12104] [Citation(s) in RCA: 36] [Impact Index Per Article: 18.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/14/2023]
Abstract
Postoperative abdominal adhesions are a common problem after surgery and can produce serious complications. Current antiadhesive strategies focus mostly on physical barriers and are unsatisfactory and inefficient. In this study, we designed and synthesized advanced injectable cream-like hydrogels with multiple functionalities, including rapid gelation, self-healing, antioxidation, anti-inflammation, and anti-cell adhesion. The multifunctional hydrogels were facilely formed by the conjugation reaction of epigallocatechin-3-gallate (EGCG) and hyaluronic acid (HA)-based microgels and poly(vinyl alcohol) (PVA) based on the dynamic boronic ester bond. The physicochemical properties of the hydrogels including antioxidative and anti-inflammatory activities were systematically characterized. A mouse cecum-abdominal wall adhesion model was implemented to investigate the efficacy of our microgel-based hydrogels in preventing postoperative abdominal adhesions. The hydrogels, with a high molecular weight HA, significantly decreased the inflammation, oxidative stress, and fibrosis and reduced the abdominal adhesion formation, compared to the commercial Seprafilm group or Injury-only group. Label-free quantitative proteomics analysis demonstrated that S100A8 and S100A9 expressions were associated with adhesion formation; the microgel-containing hydrogels inhibited these expressions. The microgel-containing hydrogels with multifunctionality decreased the formation of postoperative intra-abdominal adhesions in a murine model, demonstrating promise for clinical applications.
Collapse
Affiliation(s)
- Bo Liu
- Mary & Dick Holland Regenerative Medicine Program, University of Nebraska Medical Center, Omaha, NE, 68198, USA
- Division of Cardiology, Department of Internal Medicine, University of Nebraska Medical Center, Omaha, NE, 68198, USA
| | - Yunfan Kong
- Mary & Dick Holland Regenerative Medicine Program, University of Nebraska Medical Center, Omaha, NE, 68198, USA
- Division of Cardiology, Department of Internal Medicine, University of Nebraska Medical Center, Omaha, NE, 68198, USA
| | - Olawale A. Alimi
- Mary & Dick Holland Regenerative Medicine Program, University of Nebraska Medical Center, Omaha, NE, 68198, USA
- Division of Cardiology, Department of Internal Medicine, University of Nebraska Medical Center, Omaha, NE, 68198, USA
| | - Mitchell A. Kuss
- Mary & Dick Holland Regenerative Medicine Program, University of Nebraska Medical Center, Omaha, NE, 68198, USA
- Division of Cardiology, Department of Internal Medicine, University of Nebraska Medical Center, Omaha, NE, 68198, USA
| | - Huiyin Tu
- Department of Emergency Medicine, College of Medicine, University of Nebraska Medical Center, Omaha, NE, 68198, USA
| | - Wenfeng Hu
- Department of Emergency Medicine, College of Medicine, University of Nebraska Medical Center, Omaha, NE, 68198, USA
| | - Abu Rafay
- Mass Spectrometry & Proteomics Core, University of Nebraska Medical Center, Omaha, NE, 68198, USA
| | - Kumar Vikas
- Mass Spectrometry & Proteomics Core, University of Nebraska Medical Center, Omaha, NE, 68198, USA
- Department of Genetics, Cell Biology and Anatomy, University of Nebraska Medical Center, Omaha, NE, 68198, USA
| | - Wen Shi
- Mary & Dick Holland Regenerative Medicine Program, University of Nebraska Medical Center, Omaha, NE, 68198, USA
- Division of Cardiology, Department of Internal Medicine, University of Nebraska Medical Center, Omaha, NE, 68198, USA
| | - Megan Lerner
- Department of Surgery, University of Oklahoma Health Sciences Center, Oklahoma City, OK, 73104, USA
| | - William L. Berry
- Department of Surgery, University of Oklahoma Health Sciences Center, Oklahoma City, OK, 73104, USA
| | - Yulong Li
- Department of Emergency Medicine, College of Medicine, University of Nebraska Medical Center, Omaha, NE, 68198, USA
| | - Mark A. Carlson
- Department of Genetics, Cell Biology and Anatomy, University of Nebraska Medical Center, Omaha, NE, 68198, USA
- Department of Surgery-General Surgery, University of Nebraska Medical Center, Omaha, NE, 68198, USA
| | - Bin Duan
- Mary & Dick Holland Regenerative Medicine Program, University of Nebraska Medical Center, Omaha, NE, 68198, USA
- Division of Cardiology, Department of Internal Medicine, University of Nebraska Medical Center, Omaha, NE, 68198, USA
- Department of Surgery-General Surgery, University of Nebraska Medical Center, Omaha, NE, 68198, USA
- Department of Mechanical and Materials Engineering, University of Nebraska-Lincoln, Lincoln, NE, 68588, USA
| |
Collapse
|
14
|
Dolivo DM, Sun LS, Rodrigues AE, Galiano RD, Mustoe TA, Hong SJ. Epidermal Potentiation of Dermal Fibrosis: Lessons from Occlusion and Mucosal Healing. THE AMERICAN JOURNAL OF PATHOLOGY 2023; 193:510-519. [PMID: 36740181 DOI: 10.1016/j.ajpath.2023.01.008] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/02/2022] [Revised: 01/23/2023] [Accepted: 01/24/2023] [Indexed: 02/05/2023]
Abstract
Fibrotic skin conditions, such as hypertrophic and keloid scars, frequently result from injury to the skin and as sequelae to surgical procedures. The development of skin fibrosis may lead to patient discomfort, limitation in range of motion, and cosmetic disfigurement. Despite the frequency of skin fibrosis, treatments that seek to address the root causes of fibrosis are lacking. Much research into fibrotic pathophysiology has focused on dermal pathology, but less research has been performed to understand aberrations in fibrotic epidermis, leading to an incomplete understanding of dermal fibrosis. The literature on occlusion, a treatment modality known to reduce dermal fibrosis, in part through accelerating wound healing and regulating aberrant epidermal inflammation that otherwise drives fibrosis in the dermis, is reviewed. There is a focus on epidermal-dermal crosstalk, which contributes to the development and maintenance of dermal fibrosis, an underemphasized interplay that may yield novel strategies for treatment if understood in more detail.
Collapse
Affiliation(s)
- David M Dolivo
- Department of Surgery, Northwestern University Feinberg School of Medicine, Chicago, Illinois
| | - Lauren S Sun
- Department of Surgery, Northwestern University Feinberg School of Medicine, Chicago, Illinois
| | - Adrian E Rodrigues
- Department of Surgery, Northwestern University Feinberg School of Medicine, Chicago, Illinois
| | - Robert D Galiano
- Department of Surgery, Northwestern University Feinberg School of Medicine, Chicago, Illinois
| | - Thomas A Mustoe
- Department of Surgery, Northwestern University Feinberg School of Medicine, Chicago, Illinois
| | - Seok Jong Hong
- Department of Surgery, Northwestern University Feinberg School of Medicine, Chicago, Illinois.
| |
Collapse
|
15
|
Du L, Chen Y, Shi J, Yu X, Zhou J, Wang X, Xu L, Liu J, Gao J, Gu X, Wang T, Yin Z, Li C, Yan M, Wang J, Yin X, Lu Q. Inhibition of S100A8/A9 ameliorates renal interstitial fibrosis in diabetic nephropathy. Metabolism 2022:155376. [PMID: 36521551 DOI: 10.1016/j.metabol.2022.155376] [Citation(s) in RCA: 35] [Impact Index Per Article: 11.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/28/2022] [Revised: 11/29/2022] [Accepted: 12/05/2022] [Indexed: 12/14/2022]
Abstract
BACKGROUND Renal interstitial fibrosis (RIF) is one of the main features of diabetic nephropathy (DN), but the molecular mechanisms mediating RIF in DN has yet been fully understood. S100A8 and S100A9 are the proteins associated with immune and inflammation response. Here we reported the expression of S100A8 and S100A9 were significantly increased on tubular epithelial cells in diabetic kidneys through a proteomic analysis. METHODS We detected the expression of S100A8/A9 in diabetic kidneys by using immunoblotting, real-time PCR and immunostaining. RNA silencing and overexpression were performed by using S100A8/A9 expression/knockdown lentivirus to investigate the connection between S100A8/A9 and epithelial to mesenchymal transition (EMT) process. We also identify the expression of TLR4/NFκB pathway-related molecules in the case mentioned above. Afterwards a CO-IP assay was used to verify that compound AB38b ameliorates the EMT by interfering S100A8/A9 expression. RESULTS The expression of S100A8 and S100A9 were significantly increased on tubular epithelial cells in diabetic kidneys. S100A8/A9 knocking-down alleviate and over-expression promote the renal interstitial fibrosis of diabetic mice. Mechanically, high levels of S100A8/A9 expression in tubular epithelial cells during diabetic condition activated the TLR4/NF-κB signal pathway which promoted the EMT process and finally led to RIF progression. S100A8/A9 knockdown ameliorated RIF of diabetic mice. Further experiments revealed that compound AB38b inhibited the EMT progression of tubular epithelial cells induced by S100A8/A9 through interfering the expressions of S100A8/A9. CONCLUSIONS Our study suggest that abnormal expression of S100A8/A9 in the disease condition promotes EMT process and RIF through TLR4/NF-κB signal pathway. Using small molecular inhibitor AB38b to inhibit the abnormal expressions of S100A8/A9 might be a novel therapeutic strategy in treating DN.
Collapse
Affiliation(s)
- Lei Du
- Jiangsu Key Laboratory of New Drug Research and Clinical Pharmacy, Xuzhou Medical University, China
| | - Yibing Chen
- Jiangsu Key Laboratory of New Drug Research and Clinical Pharmacy, Xuzhou Medical University, China; State Key Laboratory of Bioactive Substance and Function of Natural Medicines, Institute of Materia Medica, Chinese Academy of Medical Sciences and Peking Union Medical College, China
| | - Jiasen Shi
- Jiangsu Key Laboratory of New Drug Research and Clinical Pharmacy, Xuzhou Medical University, China
| | - Xiujuan Yu
- Jiangsu Key Laboratory of New Drug Research and Clinical Pharmacy, Xuzhou Medical University, China
| | - Jieling Zhou
- Jiangsu Key Laboratory of New Drug Research and Clinical Pharmacy, Xuzhou Medical University, China
| | - Xue Wang
- Jiangsu Key Laboratory of New Drug Research and Clinical Pharmacy, Xuzhou Medical University, China
| | - Liu Xu
- Jiangsu Key Laboratory of New Drug Research and Clinical Pharmacy, Xuzhou Medical University, China
| | - Junjie Liu
- Department of Urology, Affiliated Hospital of Xuzhou Medical University, China
| | - Jian Gao
- Jiangsu Key Laboratory of New Drug Research and Clinical Pharmacy, Xuzhou Medical University, China
| | - Xiaoke Gu
- Jiangsu Key Laboratory of New Drug Research and Clinical Pharmacy, Xuzhou Medical University, China
| | - Tao Wang
- Department of Clinical Pharmacy, Affiliated Hospital of Xuzhou Medical University, China
| | - Zeyuan Yin
- Jiangsu Key Laboratory of New Drug Research and Clinical Pharmacy, Xuzhou Medical University, China; Department of Clinical Medicine, Xuzhou Medical University, China
| | - Chenglin Li
- Jiangsu Key Laboratory of New Drug Research and Clinical Pharmacy, Xuzhou Medical University, China
| | - Meng Yan
- Jiangsu Key Laboratory of New Drug Research and Clinical Pharmacy, Xuzhou Medical University, China
| | - Jianyun Wang
- Jiangsu Key Laboratory of New Drug Research and Clinical Pharmacy, Xuzhou Medical University, China
| | - Xiaoxing Yin
- Jiangsu Key Laboratory of New Drug Research and Clinical Pharmacy, Xuzhou Medical University, China
| | - Qian Lu
- Jiangsu Key Laboratory of New Drug Research and Clinical Pharmacy, Xuzhou Medical University, China.
| |
Collapse
|
16
|
Liu Q, Lei X, Cao Z, Zhang J, Yan L, Fu J, Tong Q, Qin W, Shao Y, Liu C, Liu Z, Wang Z, Chu Y, Xu G, Liu S, Wen X, Yamamoto H, Mori M, Liang XM, Xu X. TRPM8 deficiency attenuates liver fibrosis through S100A9-HNF4α signaling. Cell Biosci 2022; 12:58. [PMID: 35525986 PMCID: PMC9080211 DOI: 10.1186/s13578-022-00789-4] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/26/2021] [Accepted: 04/18/2022] [Indexed: 12/03/2022] Open
Abstract
Background Liver fibrosis represent a major global health care burden. Data emerging from recent advances suggest TRPM8, a member of the transient receptor potential (TRP) family of ion channels, plays an essential role in various chronic inflammatory diseases. However, its role in liver fibrosis remains unknown. Herein, we assessed the potential effect of TRPM8 in liver fibrosis. Methods The effect of TRPM8 was evaluated using specimens obtained from classic murine models of liver fibrosis, namely wild-type (WT) and TRPM8−/− (KO) fibrotic mice after carbon tetrachloride (CCl4) or bile duct ligation (BDL) treatment. The role of TRPM8 was systematically evaluated using specimens obtained from the aforementioned animal models after various in vivo and in vitro experiments. Results Clinicopathological analysis showed that TRPM8 expression was upregulated in tissue samples from cirrhosis patients and fibrotic mice. TRPM8 deficiency not only attenuated inflammation and fibrosis progression in mice but also helped to alleviate symptoms of cholangiopathies. Moreover, reduction in S100A9 and increase in HNF4α expressions were observed in liver of CCl4- and BDL- treated TRPM8−/− mice. A strong regulatory linkage between S100A9 and HNF4α was also noticed in L02 cells that underwent siRNA-mediated S100A9 knockdown and S100A9 overexpressing plasmid transfection. Lastly, the alleviative effect of a selective TRPM8 antagonist was confirmed in vivo. Conclusions These findings suggest TRPM8 deficiency may exert protective effects against inflammation, cholangiopathies, and fibrosis through S100A9-HNF4α signaling. M8-B might be a promising therapeutic candidate for liver fibrosis. Graphical Abstract ![]()
Supplementary Information The online version contains supplementary material available at 10.1186/s13578-022-00789-4.
Collapse
|
17
|
Liu X, Lu F, Chen X. Examination of the role of necroptotic damage-associated molecular patterns in tissue fibrosis. Front Immunol 2022; 13:886374. [PMID: 36110858 PMCID: PMC9468929 DOI: 10.3389/fimmu.2022.886374] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2022] [Accepted: 08/08/2022] [Indexed: 11/29/2022] Open
Abstract
Fibrosis is defined as the abnormal and excessive deposition of extracellular matrix (ECM) components, which leads to tissue or organ dysfunction and failure. However, the pathological mechanisms underlying fibrosis remain unclear. The inflammatory response induced by tissue injury is closely associated with tissue fibrosis. Recently, an increasing number of studies have linked necroptosis to inflammation and fibrosis. Necroptosis is a type of preprogrammed death caused by death receptors, interferons, Toll-like receptors, intracellular RNA and DNA sensors, and other mediators. These activate receptor-interacting protein kinase (RIPK) 1, which recruits and phosphorylates RIPK3. RIPK3 then phosphorylates a mixed lineage kinase domain-like protein and causes its oligomerization, leading to rapid plasma membrane permeabilization, the release of cellular contents, and exposure of damage-associated molecular patterns (DAMPs). DAMPs, as inflammatory mediators, are involved in the loss of balance between extensive inflammation and tissue regeneration, leading to remodeling, the hallmark of fibrosis. In this review, we discuss the role of necroptotic DAMPs in tissue fibrosis and highlight the inflammatory responses induced by DAMPs in tissue ECM remodeling. By summarizing the existing literature on this topic, we underscore the gaps in the current research, providing a framework for future investigations into the relationship among necroptosis, DAMPs, and fibrosis, as well as a reference for later transformation into clinical treatment.
Collapse
Affiliation(s)
| | - Feng Lu
- *Correspondence: Feng Lu, ; Xihang Chen,
| | | |
Collapse
|
18
|
Veronese S, Beatini A, Urbani C, Lanza E, Paz OM, Saussaye Y, Lomuto M, Sbarbati A. V-EMF treatment of facial scar: First results. J Tissue Viability 2022; 31:614-618. [PMID: 35853796 DOI: 10.1016/j.jtv.2022.07.006] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2021] [Accepted: 07/12/2022] [Indexed: 10/17/2022]
Abstract
AIM OF STUDY This is a retrospective study aimed at evaluating the effectiveness of the use of electromagnetic fields and negative pressure treatment (V-EMF) for facial scars, from an aesthetic and functional point of view, and considering the variations in the levels of hydration. MATERIAL AND METHODS 25 subjects with facial scarring were re-evaluated after being treated with the V-EMF method. The hydration levels of the scars before and after treatment were compared. The results were evaluated considering the satisfaction levels of the patients with the VAS, and of the medical specialists who performed the treatment, and of 3 independent dermatologists with the Likert scale. RESULTS Mean hydration levels of scars went from 41.8 to 53.3, with mean hydration levels of healthy reference points equal to 54.6. The minimum patient satisfaction level was 2 in the VAS. The minimum level of satisfaction of specialists and dermatologists was equal to IV on the Likert scale for all patients, except for 1 subject in which it was III for the specialist who had treated him. Anti-aging and re-pigmentation effects were also noted as secondary results. CONCLUSIONS From an aesthetic and functional point of view, and for the overall anti-aging effect of the treated area, V-EMF applied to facial scars has shown extremely promising results.
Collapse
Affiliation(s)
- Sheila Veronese
- Department of Neuroscience, Biomedicine and Movement Sciences, University of Verona, 37134, Verona, Italy.
| | | | | | - Eliana Lanza
- Estederm Studio di Dermatologia, Medicina Estetica e Chirurgia Plastica, 95127, Catania, Italy
| | | | | | - Michele Lomuto
- Ospedale Casa Sollievo della Sofferenza IRCCS, 71013, San Giovanni Rotondo, Foggia, Italy
| | - Andrea Sbarbati
- Department of Neuroscience, Biomedicine and Movement Sciences, University of Verona, 37134, Verona, Italy
| |
Collapse
|
19
|
Reduced hydration regulates pro-inflammatory cytokines via CD14 in barrier function-impaired skin. Biochim Biophys Acta Mol Basis Dis 2022; 1868:166482. [DOI: 10.1016/j.bbadis.2022.166482] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2022] [Revised: 06/28/2022] [Accepted: 06/29/2022] [Indexed: 11/23/2022]
|
20
|
He Y, Zou C, Cai Z. Construction and Comprehensive Analysis of the ceRNA Network to Reveal Key Genes for Benign Tracheal Stenosis. Front Genet 2022; 13:891741. [PMID: 35812753 PMCID: PMC9261475 DOI: 10.3389/fgene.2022.891741] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2022] [Accepted: 05/24/2022] [Indexed: 11/21/2022] Open
Abstract
Objective: To explore the possible biological functions of the differentially expressed genes in patients with benign tracheal stenosis, and to provide a valuable molecular basis for investigating the pathogenesis of benign tracheal stenosis. Method: Whole transcriptome sequencing was performed on blood samples collected from patients with benign tracheal stenosis and normal controls. Differentially expressed mRNA, lncRNA, and circRNA were analyzed using the DESeq2 package. The protein interaction networks for differentially expressed mRNAs were constructed by STRING. The results of gene co-expression network analysis, Starbase database prediction, and differential gene expression were combined to construct a competing endogenous RNA network. The transcription factors of key genes were predicted using the Network Analyst database and a transcription factor-mRNA regulatory network was constructed. The classical pathways, intermolecular interaction networks, and upstream regulatory components of key genes were analyzed using Ingenuity Pathway Analysis (IPA). Finally, the DGIDB database was used to predict the potential therapeutic drugs to target the identified key genes. Result: Based on mRNA, lncRNA and circRNA expression data, we found that differentially expressed mRNAs were enriched in oxygen transport, neutrophil activation, immune response, and oxygen binding. Then the pearson correlation between mRNAs of 46 key genes and lncRNAs and cricRNAs were calculated, and the correlation greater than 0.9 were selected to construct the co-expression network of “mRNA-lncRA” and “mRNA-cricRNA.” Moreover, a “lncRNA-miRNA-mRNA” network and a “circRNA-miRNA-mRNA” network were constructed. IPA analysis showed that the 46 key genes were significantly associated with inflammatory activation and acute respiratory distress syndrome. The constructed TF-mRNA regulatory network was composed of 274 nodes and 573 interacting pairs. 251 potential therapeutic drugs were identified from the DGIDB database. Conclusion: This study analyzed the differential genes associated with benign tracheal stenosis and explored the potential regulatory mechanisms, providing a scientific reference for further studies on the pathogenesis of benign tracheal stenosis.
Collapse
Affiliation(s)
- Yanpeng He
- The First Department of Pulmonary and Critical Care Medicine, The Second Hospital of Hebei Medical University, Heibei Key Laboratory of Respiratory Critical Care, Shijiazhuang, China
- Department of Pulmonary and Critical Care Medicine, The First Hospital of Qinhuangdao, Qinhuangdao, China
| | - Chunyan Zou
- Department of Pulmonary and Critical Care Medicine, The First Hospital of Qinhuangdao, Qinhuangdao, China
| | - Zhigang Cai
- The First Department of Pulmonary and Critical Care Medicine, The Second Hospital of Hebei Medical University, Heibei Key Laboratory of Respiratory Critical Care, Shijiazhuang, China
- *Correspondence: Zhigang Cai,
| |
Collapse
|
21
|
Parikh UM, Mentz J, Collier I, Davis MJ, Abu-Ghname A, Colchado D, Short WD, King A, Buchanan EP, Balaji S. Strategies to Minimize Surgical Scarring: Translation of Lessons Learned from Bedside to Bench and Back. Adv Wound Care (New Rochelle) 2022; 11:311-329. [PMID: 34416825 DOI: 10.1089/wound.2021.0010] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
Significance: An understanding of the physiology of wound healing and scarring is necessary to minimize surgical scar formation. By reducing tension across the healing wound, eliminating excess inflammation and infection, and encouraging perfusion to healing areas, surgeons can support healing and minimize scarring. Recent Advances: Preoperatively, newer techniques focused on incision placement to minimize tension, skin sterilization to minimize infection and inflammation, and control of comorbid factors to promote a healing process with minimal scarring are constantly evolving. Intraoperatively, measures like layered closure, undermining, and tissue expansion can be taken to relieve tension across the healing wound. Appropriate suture technique and selection should be considered, and finally, there are new surgical technologies available to reduce tension across the closure. Postoperatively, the healing process can be supported as proliferation and remodeling take place within the wound. A balance of moisture control, tension reduction, and infection prevention can be achieved with dressings, ointments, and silicone. Vitamins and corticosteroids can also affect the scarring process by modulating the cellular factors involved in healing. Critical Issues: Healing with no or minimal scarring is the ultimate goal of wound healing research. Understanding how mechanical tension, inflammation and infection, and perfusion and hypoxia impact profibrotic pathways allows for the development of therapies that can modulate cytokine response and the wound extracellular microenvironment to reduce fibrosis and scarring. Future Directions: New tension-off loading topical treatments, laser, and dermabrasion devices are under development, and small molecule therapeutics have demonstrated scarless wound healing in animal models, providing a promising new direction for future research aimed to minimize surgical scarring.
Collapse
Affiliation(s)
- Umang M. Parikh
- Division of Pediatric Surgery, Michael E. DeBakey Department of Surgery, Texas Children's Hospital and Baylor College of Medicine, Houston, Texas, USA
| | - James Mentz
- Division of Plastic Surgery, Michael E. DeBakey Department of Surgery, Texas Children's Hospital and Baylor College of Medicine, Houston, Texas, USA
| | - Ian Collier
- Division of Plastic Surgery, Michael E. DeBakey Department of Surgery, Texas Children's Hospital and Baylor College of Medicine, Houston, Texas, USA
| | - Matthew J. Davis
- Division of Plastic Surgery, Michael E. DeBakey Department of Surgery, Texas Children's Hospital and Baylor College of Medicine, Houston, Texas, USA
| | - Amjed Abu-Ghname
- Division of Plastic Surgery, Michael E. DeBakey Department of Surgery, Texas Children's Hospital and Baylor College of Medicine, Houston, Texas, USA
| | - Daniel Colchado
- Division of Pediatric Surgery, Michael E. DeBakey Department of Surgery, Texas Children's Hospital and Baylor College of Medicine, Houston, Texas, USA
- Division of Plastic Surgery, Michael E. DeBakey Department of Surgery, Texas Children's Hospital and Baylor College of Medicine, Houston, Texas, USA
| | - Walker D. Short
- Division of Pediatric Surgery, Michael E. DeBakey Department of Surgery, Texas Children's Hospital and Baylor College of Medicine, Houston, Texas, USA
| | - Alice King
- Division of Pediatric Surgery, Michael E. DeBakey Department of Surgery, Texas Children's Hospital and Baylor College of Medicine, Houston, Texas, USA
| | - Edward P. Buchanan
- Division of Plastic Surgery, Michael E. DeBakey Department of Surgery, Texas Children's Hospital and Baylor College of Medicine, Houston, Texas, USA
| | - Swathi Balaji
- Division of Pediatric Surgery, Michael E. DeBakey Department of Surgery, Texas Children's Hospital and Baylor College of Medicine, Houston, Texas, USA
| |
Collapse
|
22
|
Rochette L, Dogon G, Rigal E, Zeller M, Cottin Y, Vergely C. Involvement of Oxidative Stress in Protective Cardiac Functions of Calprotectin. Cells 2022; 11:cells11071226. [PMID: 35406797 PMCID: PMC8997643 DOI: 10.3390/cells11071226] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2022] [Revised: 03/24/2022] [Accepted: 04/01/2022] [Indexed: 02/01/2023] Open
Abstract
Calprotectin (CLP) belonging to the S-100 protein family is a heterodimeric complex (S100A8/S100A9) formed by two binding proteins. Upon cell activation, CLP stored in neutrophils is released extracellularly in response to inflammatory stimuli and acts as damage-associated molecular patterns (DAMPs). S100A8 and S100A9 possess both anti-inflammatory and anti-bacterial properties. The complex is a ligand of the toll-like receptor 4 (TLR4) and receptor for advanced glycation end (RAGE). At sites of infection and inflammation, CLP is a target for oxidation due to its co-localization with neutrophil-derived oxidants. In the heart, oxidative stress (OS) responses and S100 proteins are closely related and intimately linked through pathophysiological processes. Our review summarizes the roles of S100A8, S100A9 and CLP in the inflammation in relationship with vascular OS, and we examine the importance of CLP for the mechanisms driving in the protection of myocardium. Recent evidence interpreting CLP as a critical modulator during the inflammatory response has identified this alarmin as an interesting drug target.
Collapse
Affiliation(s)
- Luc Rochette
- Equipe d’Accueil (EA 7460): Physiopathologie et Epidémiologie Cérébro-Cardiovasculaires (PEC2), Faculté des Sciences de Santé, Université de Bourgogne—Franche Comté, 7 Bd Jeanne d’Arc, 21000 Dijon, France; (G.D.); (E.R.); (M.Z.); (C.V.)
- Correspondence:
| | - Geoffrey Dogon
- Equipe d’Accueil (EA 7460): Physiopathologie et Epidémiologie Cérébro-Cardiovasculaires (PEC2), Faculté des Sciences de Santé, Université de Bourgogne—Franche Comté, 7 Bd Jeanne d’Arc, 21000 Dijon, France; (G.D.); (E.R.); (M.Z.); (C.V.)
| | - Eve Rigal
- Equipe d’Accueil (EA 7460): Physiopathologie et Epidémiologie Cérébro-Cardiovasculaires (PEC2), Faculté des Sciences de Santé, Université de Bourgogne—Franche Comté, 7 Bd Jeanne d’Arc, 21000 Dijon, France; (G.D.); (E.R.); (M.Z.); (C.V.)
| | - Marianne Zeller
- Equipe d’Accueil (EA 7460): Physiopathologie et Epidémiologie Cérébro-Cardiovasculaires (PEC2), Faculté des Sciences de Santé, Université de Bourgogne—Franche Comté, 7 Bd Jeanne d’Arc, 21000 Dijon, France; (G.D.); (E.R.); (M.Z.); (C.V.)
| | - Yves Cottin
- Service de Cardiologie, CHU-Dijon, 21000 Dijon, France;
| | - Catherine Vergely
- Equipe d’Accueil (EA 7460): Physiopathologie et Epidémiologie Cérébro-Cardiovasculaires (PEC2), Faculté des Sciences de Santé, Université de Bourgogne—Franche Comté, 7 Bd Jeanne d’Arc, 21000 Dijon, France; (G.D.); (E.R.); (M.Z.); (C.V.)
| |
Collapse
|
23
|
Termer M, Jaeger A, Carola C, Salazar A, Keck CM, Kolmar H, von Hagen J. Methoxy-Monobenzoylmethane Protects Skin from UV-Induced Damages in a Randomized, Placebo Controlled, Double-Blinded Human In Vivo Study and Prevents Signs of Inflammation While Improving the Skin Barrier. Dermatol Ther (Heidelb) 2022; 12:435-449. [PMID: 34939179 PMCID: PMC8850519 DOI: 10.1007/s13555-021-00652-3] [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: 10/15/2021] [Accepted: 11/18/2021] [Indexed: 11/24/2022] Open
Abstract
INTRODUCTION Sun protection is important in skin care and requires special attention as inefficient protection might trigger skin pathologies including polymorphic light eruption (PLE). The reduce-improve-protect (RIP) concept to avoid the onset of ultraviolet (UV) irradiation-induced diseases or damage to human skin is important. Methoxy-monobenzoylmethane (MeO-MBM), which is neither a UVB nor a UVA filter, converts to the UV filter avobenzone under UV irradiation and further acts as a photoantioxidant during its conversion process and initially as an antioxidant material. The aim of this study was to understand the mechanisms by which MeO-MBM improves the condition of UV-stressed skin through its photoantioxidant properties. The improvement of the skin condition by the activity of MeO-MBM as active ingredient was also investigated. METHODS Potential molecular targets were identified by in silico docking to numerous cellular membrane receptors on the cell surface or nuclear membrane, followed by microarray analysis of 164 genes after MeO-MBM treatment of normal human epidermal keratinocytes (NHEK). We conducted randomized, double-blinded, intra-individual comparison vs. placebo studies on ten volunteers, aged between 34 and 65 years, to assess the effect of MeO-MBM in vivo. The effect after UV-induced inflammation was assessed in a protective and curative set-up with 2% MeO-MBM vs. 1% hydrocortisone and placebo based on the change in blood flow. The barrier function of the skin was assessed by the change in transepidermal water loss (TEWL), skin scaling and skin thickness after the treatment with MeO-MBM. Additionally, the effect of MeO-MBM after UV-induced stress on the activation of ferritin in human explants was determined ex vivo. RESULTS A docking simulation of MeO-MBM showed a potential interaction with the retinoic acid receptor gamma and further revealed downregulation of proteins related to inflammation. In the protective treatment set-up, after 24 h MeO-MBM significantly reduced the delta blood flow compared to placebo, while this reduction was more prominent with hydrocortisone. In the curative treatment set-up, a greater reduction in delta blood flow was also observed with MeO-MBM compared to placebo and similar to hydrocortisone. Treatment with MeO-MBM revealed an improvement in skin barrier function as a result of decreased TEWL, reduced skin scaling and increased skin thickness. Immunohistochemistry staining of ferritin on human skin explants further showed that the treatment with MeO-MBM reduced the ferritin expression. CONCLUSION Based on these results, MeO-MBM is capable of exerting an anti-aging activity via the retinoic acid receptor gamma. Its anti-inflammatory and anti-oxidative activity manifested via the downregulation of multiple anti-inflammatory genes as well as the reduction of ferritin in skin tissue. This study shows that the multidimensional functionality of MeO-MBM offers an effective approach to combat acute and chronic deleterious effects of oxidative UV damage while simultaneously enhancing the skin barrier function.
Collapse
Affiliation(s)
- Michael Termer
- Department of Pharmaceutics and Biopharmaceutics, Philipps University of Marburg, Marburg, Germany
| | - Anita Jaeger
- Department of Applied Biochemistry, Technical University of Darmstadt, Darmstadt, Germany
| | | | | | - Cornelia M. Keck
- Department of Pharmaceutics and Biopharmaceutics, Philipps University of Marburg, Marburg, Germany
| | - Harald Kolmar
- Department of Applied Biochemistry, Technical University of Darmstadt, Darmstadt, Germany
| | | |
Collapse
|
24
|
Pang B, Zhu Z, Xiao C, Luo Y, Fang H, Bai Y, Sun Z, Ma J, Dang E, Wang G. Keratin 17 Is Required for Lipid Metabolism in Keratinocytes and Benefits Epidermal Permeability Barrier Homeostasis. Front Cell Dev Biol 2022; 9:779257. [PMID: 35096815 PMCID: PMC8790522 DOI: 10.3389/fcell.2021.779257] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2021] [Accepted: 11/29/2021] [Indexed: 12/20/2022] Open
Abstract
The epidermal barrier refers to the stratum corneum, the uppermost layer of the skin, and constitutes the first line of defense against invasion by potentially harmful pathogens, diminishes trans-epidermal water loss, and plays a crucial role in the maintenance of skin homeostasis. Keratin 17 (K17) is a type I epithelial keratin with multiple functions, including in skin inflammation, epithelial cell growth, protein synthesis, and tumorigenesis. However, the relationship between K17 and the skin barrier has yet to be systematically investigated. In this study, we found that acute disruption of the epidermal permeability barrier led to a rapid increase in epidermal K17 expression in vivo. Krt17 gene deficiency in mice resulted in decreased expression of lipid metabolism-related enzymes and antimicrobial peptides, while also delaying epidermal permeability barrier recovery after acute disruption. Adenovirus-mediated overexpression of K17 enhanced, whereas siRNA-mediated knockdown of Krt17 inhibited, the expression of fatty acid synthase (FASN) and that of the transcription factors SREBP-1 and PPARγ in vitro. We further confirmed that K17 can facilitate the nuclear transportation of SREBP-1 and PPARγ and promote lipid synthesis in keratinocytes. This study demonstrated that K17 contributes to the restoration of the epidermal permeability barrier via stabilizing lipid metabolism in keratinocytes.
Collapse
Affiliation(s)
- Bingyu Pang
- Department of Dermatology, Xijing Hospital, Fourth Military Medical University, Xi'an, China
| | - Zhenlai Zhu
- Department of Dermatology, Xijing Hospital, Fourth Military Medical University, Xi'an, China
| | - Chunying Xiao
- Department of Dermatology, Xijing Hospital, Fourth Military Medical University, Xi'an, China
| | - Yixin Luo
- Department of Dermatology, Xijing Hospital, Fourth Military Medical University, Xi'an, China
| | - Hui Fang
- Department of Dermatology, Xijing Hospital, Fourth Military Medical University, Xi'an, China
| | - Yaxing Bai
- Department of Dermatology, Xijing Hospital, Fourth Military Medical University, Xi'an, China
| | - Zhongbin Sun
- Department of Dermatology, Xijing Hospital, Fourth Military Medical University, Xi'an, China
| | - Jingyi Ma
- Department of Dermatology, Xijing Hospital, Fourth Military Medical University, Xi'an, China
| | - Erle Dang
- Department of Dermatology, Xijing Hospital, Fourth Military Medical University, Xi'an, China
| | - Gang Wang
- Department of Dermatology, Xijing Hospital, Fourth Military Medical University, Xi'an, China
| |
Collapse
|
25
|
Amiri N, Golin AP, Jalili RB, Ghahary A. Roles of cutaneous cell-cell communication in wound healing outcome: An emphasis on keratinocyte-fibroblast crosstalk. Exp Dermatol 2021; 31:475-484. [PMID: 34932841 DOI: 10.1111/exd.14516] [Citation(s) in RCA: 37] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2021] [Revised: 11/28/2021] [Accepted: 12/19/2021] [Indexed: 01/13/2023]
Abstract
Tissue repair is a very complex event and involves a continuously orchestrated sequence of signals and responses from platelets, fibroblasts, epithelial, endothelial and immune cells. The details of interaction between these signals, which are mainly growth factors and cytokines, have been widely discussed. However, it is still not clear how activated cells at wound sites lessen their activities after epithelialization is completed. Termination of the wound healing process requires a fine balance between extracellular matrix (ECM) deposition and degradation. Maintaining this balance requires highly accurate epithelial-mesenchymal communication and correct information exchange between keratinocytes and fibroblasts. As it has been reported in the literature, a disruption in epithelialization during the process of wound healing increases the frequency of developing chronic wounds or fibrotic conditions, as seen in a variety of clinical cases. Conversely, the potential stop signal for wound healing should have a regulatory role on both ECM synthesis and degradation to reach a successful wound healing outcome. This review briefly describes the potential roles of growth factors and cytokines in controlling the early phase of wound healing and predominantly explores the role of releasable factors from epithelial-mesenchymal interaction in controlling during and the late stage of the healing process. Emphasis will be given on the crosstalk between keratinocytes and fibroblasts in ECM modulation and the healing outcome following a brief discussion of the wound healing initiation mechanism. In particular, we will review the termination of acute dermal wound healing, which frequently leads to the development of hypertrophic scarring.
Collapse
Affiliation(s)
- Nafise Amiri
- Department of Surgery, International Collaboration on Repair Discoveries (ICORD), University of British Columbia, Vancouver, British Columbia, Canada
| | - Andrew P Golin
- Faculty of Medicine, University of British Columbia, Vancouver, British Columbia, Canada
| | - Reza B Jalili
- Department of Surgery, International Collaboration on Repair Discoveries (ICORD), University of British Columbia, Vancouver, British Columbia, Canada
| | - Aziz Ghahary
- Department of Surgery, International Collaboration on Repair Discoveries (ICORD), University of British Columbia, Vancouver, British Columbia, Canada
| |
Collapse
|
26
|
Goto T, Sapio MR, Maric D, Robinson JM, Domenichiello AF, Saligan LN, Mannes AJ, Iadarola MJ. Longitudinal peripheral tissue RNA-Seq transcriptomic profiling, hyperalgesia, and wound healing in the rat plantar surgical incision model. FASEB J 2021; 35:e21852. [PMID: 34499774 PMCID: PMC9293146 DOI: 10.1096/fj.202100347r] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2021] [Revised: 07/24/2021] [Accepted: 07/28/2021] [Indexed: 02/06/2023]
Abstract
Postoperative pain and delayed healing in surgical wounds, which require complex management strategies have understudied complicated mechanisms. Here we investigated temporal changes in behavior, tissue structure, and transcriptomic profiles in a rat model of a surgical incision, using hyperalgesic behavioral tests, histological analyses, and next‐generation RNA sequencing, respectively. The most rapidly (1 hour) expressed genes were the chemokines, Cxcl1 and Cxcl2. Consequently, infiltrating leukocytes were abundantly observed starting at 6 and peaking at 24 hours after incising which was supported by histological analysis and appearance of the neutrophil markers, S100a8 and S100a9. At this time, hyperalgesia was at a peak and overall transcriptional activity was most highly activated. At the 1‐day timepoint, Nppb, coding for natriuretic peptide precursor B, was the most strongly upregulated gene and was localized by in situ hybridization to the epidermal keratinocytes at the margins of the incision. Nppb was basically unaffected in a peripheral inflammation model transcriptomic dataset. At the late phase of wound healing, five secreted, incision‐specific peptidases, Mmp2, Aebp1, Mmp23, Adamts7, and Adamtsl1, showed increased expression, supporting the idea of a sustained tissue remodeling process. Transcripts that are specifically upregulated at each timepoint in the incision model may be potential candidates for either biomarkers or therapeutic targets for wound pain and wound healing. This study incorporates the examination of longitudinal temporal molecular responses, corresponding anatomical localization, and hyperalgesic behavioral alterations in the surgical incision model that together provide important and novel foundational knowledge to understand mechanisms of wound pain and wound healing.
Collapse
Affiliation(s)
- Taichi Goto
- Symptoms Biology Unit, National Institute of Nursing Research, National Institutes of Health, Bethesda, MD, USA
| | - Matthew R Sapio
- Department of Perioperative Medicine, Clinical Center, National Institutes of Health, Bethesda, MD, USA
| | - Dragan Maric
- Flow and Imaging Cytometry Core Facility, National Institute of Neurological Disorders and Stroke, National Institutes of Health, Bethesda, MD, USA
| | - Jeffrey M Robinson
- Translational Life Science Technology Program, University of Maryland, Baltimore County, Baltimore, MD, USA
| | - Anthony F Domenichiello
- Lipid Peroxidation Unit, Laboratory of Clinical Investigation, National Institute on Aging, National Institutes of Health, Baltimore, MD, USA
| | - Leorey N Saligan
- Symptoms Biology Unit, National Institute of Nursing Research, National Institutes of Health, Bethesda, MD, USA
| | - Andrew J Mannes
- Department of Perioperative Medicine, Clinical Center, National Institutes of Health, Bethesda, MD, USA
| | - Michael J Iadarola
- Department of Perioperative Medicine, Clinical Center, National Institutes of Health, Bethesda, MD, USA
| |
Collapse
|
27
|
Perkins TN, Oury TD. The perplexing role of RAGE in pulmonary fibrosis: causality or casualty? Ther Adv Respir Dis 2021; 15:17534666211016071. [PMID: 34275342 PMCID: PMC8293846 DOI: 10.1177/17534666211016071] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022] Open
Abstract
Idiopathic pulmonary fibrosis (IPF) is a progressive and fatal lung disease in which most patients die within 3 years of diagnosis. With an unknown etiology, IPF results in progressive fibrosis of the lung parenchyma, diminishing normal lung function, which results in respiratory failure, and eventually, death. While few therapies are available to reduce disease progression, patients continue to advance toward respiratory failure, leaving lung transplantation the only viable option for survival. As incidence and mortality rates steadily increase, the need for novel therapeutics is imperative. The receptor for advanced glycation endproducts (RAGE) is most highly expressed in the lungs and plays a significant role in a number of chronic lung diseases. RAGE has long been linked to IPF; however, confounding data from both human and experimental studies have left an incomplete and perplexing story. This review examines the present understanding of the role of RAGE in human and experimental models of IPF, drawing parallels to recent advances in RAGE biology. Moreover, this review discusses the role of RAGE in lung injury response, type 2 immunity, and cellular senescence, and how such mechanisms may relate to RAGE as both a biomarker of disease progression and potential therapeutic target in IPF.The reviews of this paper are available via the supplemental material section.
Collapse
Affiliation(s)
- Timothy N Perkins
- Department of Pathology, University of Pittsburgh School of Medicine, 3550 Terrace Street, S-784 Scaife Hall, Pittsburgh, PA 15261, USA
| | - Tim D Oury
- Department of Pathology, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
| |
Collapse
|
28
|
Dolivo D, Rodrigues A, Sun L, Li Y, Hou C, Galiano R, Hong SJ, Mustoe T. The Na x (SCN7A) channel: an atypical regulator of tissue homeostasis and disease. Cell Mol Life Sci 2021; 78:5469-5488. [PMID: 34100980 PMCID: PMC11072345 DOI: 10.1007/s00018-021-03854-2] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2021] [Revised: 04/15/2021] [Accepted: 05/08/2021] [Indexed: 12/15/2022]
Abstract
Within an articulately characterized family of ion channels, the voltage-gated sodium channels, exists a black sheep, SCN7A (Nax). Nax, in contrast to members of its molecular family, has lost its voltage-gated character and instead rapidly evolved a new function as a concentration-dependent sensor of extracellular sodium ions and subsequent signal transducer. As it deviates fundamentally in function from the rest of its family, and since the bulk of the impressive body of literature elucidating the pathology and biochemistry of voltage-gated sodium channels has been performed in nervous tissue, reports of Nax expression and function have been sparse. Here, we investigate available reports surrounding expression and potential roles for Nax activity outside of nervous tissue. With these studies as justification, we propose that Nax likely acts as an early sensor that detects loss of tissue homeostasis through the pathological accumulation of extracellular sodium and/or through endothelin signaling. Sensation of homeostatic aberration via Nax then proceeds to induce pathological tissue phenotypes via promotion of pro-inflammatory and pro-fibrotic responses, induced through direct regulation of gene expression or through the generation of secondary signaling molecules, such as lactate, that can operate in an autocrine or paracrine fashion. We hope that our synthesis of much of the literature investigating this understudied protein will inspire more research into Nax not simply as a biochemical oddity, but also as a potential pathophysiological regulator and therapeutic target.
Collapse
Affiliation(s)
- David Dolivo
- Department of Surgery, Northwestern University-Feinberg School of Medicine, Chicago, USA
| | - Adrian Rodrigues
- Department of Surgery, Northwestern University-Feinberg School of Medicine, Chicago, USA
| | - Lauren Sun
- Department of Surgery, Northwestern University-Feinberg School of Medicine, Chicago, USA
| | - Yingxing Li
- Department of Surgery, Northwestern University-Feinberg School of Medicine, Chicago, USA
| | - Chun Hou
- Department of Surgery, Northwestern University-Feinberg School of Medicine, Chicago, USA
- Department of Plastic and Cosmetic Surgery, First Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
| | - Robert Galiano
- Department of Surgery, Northwestern University-Feinberg School of Medicine, Chicago, USA
| | - Seok Jong Hong
- Department of Surgery, Northwestern University-Feinberg School of Medicine, Chicago, USA.
- , 300 E. Superior St., Chicago, IL, 60611, USA.
| | - Thomas Mustoe
- Department of Surgery, Northwestern University-Feinberg School of Medicine, Chicago, USA.
- , 737 N. Michigan Ave., Chicago, IL, 60611, USA.
| |
Collapse
|
29
|
Identification of differentially expressed proteins involved in fetal scarless wound healing using a rat model of cleft lip. Mol Med Rep 2021; 24:596. [PMID: 34165164 PMCID: PMC8240453 DOI: 10.3892/mmr.2021.12235] [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: 05/06/2020] [Accepted: 12/07/2020] [Indexed: 11/05/2022] Open
Abstract
In early pregnancy, fetal skin wounds can heal quickly and undergo a transition period from scarless healing to scar formation. The aim of the present study was to identify potential biomarkers associated with scarless repair of cleft lips, in order to determine the intrinsic factors leading to scar formation in embryonic tissue. A stable model of cleft lip was established using microsurgery by constructing a wedge-shaped cleft lip-like defect in fetal rats at gestational age (GA) 16.5 and GA18.5. The GA16.5 and GA18.5 groups were used to model scarless healing and scar formation, respectively. The fetuses were returned to the uterus following surgery, then removed 72 h after the procedure. Macroscopic observation of the cleft defect and histological examination were carried out. Reverse transcription-quantitative (RT-q) PCR and parallel reaction monitoring (PRM) were used to detect mRNA and protein expression levels, respectively. The upper-left lip completely healed 72 h after surgery in the GA16.5 group of fetal rats. However, this was not the case in the GA18.5 group. Histological examination indicated new follicles visible under the epidermis of the scarless group (GA16.5). Scarring was visible on the upper-left cleft lip wound of the fetal rats in the GA18.5 group. The expression of some growth and pro-inflammatory factors, including TNF-α, were also different between two groups. Label-free quantification was used to identified differentially expressed proteins and five differentially expressed proteins (Smad4, Fabp5, S100a4, S100a8 and S100a9) were identified. The relative expression of these molecules at the mRNA and protein levels were measured using RT-qPCR and PRM. These molecules may represent potential biomarkers for the scarless repair of fetal rat cleft lip wounds.
Collapse
|
30
|
Tanaka K, Enomoto N, Hozumi H, Isayama T, Naoi H, Aono Y, Katsumata M, Yasui H, Karayama M, Suzuki Y, Furuhashi K, Fujisawa T, Inui N, Nakamura Y, Suda T. Serum S100A8 and S100A9 as prognostic biomarkers in acute exacerbation of idiopathic pulmonary fibrosis. Respir Investig 2021; 59:827-836. [PMID: 34154976 DOI: 10.1016/j.resinv.2021.05.008] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2021] [Revised: 04/16/2021] [Accepted: 05/19/2021] [Indexed: 10/21/2022]
Abstract
BACKGROUND Acute exacerbation of idiopathic pulmonary fibrosis (AE-IPF) is a devastating and life-threatening condition during its clinical course. Biomarkers for precisely anticipating the prognosis of AE-IPF remain to be fully established. The objective of this study was to clarify whether S100A8 and S100A9, which are calcium-binding proteins mainly produced by activated neutrophils, are significant prognostic biomarkers in AE-IPF. METHODS Thirty-seven patients with AE-IPF who were diagnosed and treated at our hospital were retrospectively evaluated. The serum levels of S100A8 and S100A9 were measured using enzyme-linked immunosorbent assay, and the relationships between these levels and clinical parameters or prognosis were evaluated. RESULTS The serum levels of S100A8 (median 386.5 ng/mL) and S100A9 (median 60.2 ng/mL) in patients with AE-IPF were significantly higher than those in age-matched healthy controls and in patients at IPF diagnosis (p < 0.001 for all combinations). The serum levels of S100A8 negatively correlated with percent forced vital capacity (r = -0.356, p = 0.049) and positively correlated with peripheral white blood cell number (r = 0.509, p = 0.002). Immunohistochemical staining of autopsy lung specimens showed that neutrophils, present mainly in the alveolar septum, were positive for S100A8 and S100A9. Patients with AE-IPF with higher levels of S100A8 or S100A9 showed significantly worse 3-month survival than those with lower levels (log-rank test, both p = 0.028). Finally, in multivariate analysis, the serum levels of both S100A8 and S100A9 were significant prognostic factors (hazard ratio 4.032, p = 0.023 and hazard ratio 4.327, p = 0.012). CONCLUSION The serum levels of S100A8 and S100A9 at AE were significant prognostic biomarkers in patients with AE-IPF.
Collapse
Affiliation(s)
- Kazuki Tanaka
- Second Division, Department of Internal Medicine, Hamamatsu University School of Medicine, Hamamatsu, Japan
| | - Noriyuki Enomoto
- Second Division, Department of Internal Medicine, Hamamatsu University School of Medicine, Hamamatsu, Japan; Health Administration Center, Hamamatsu University School of Medicine, Hamamatsu, Japan.
| | - Hironao Hozumi
- Second Division, Department of Internal Medicine, Hamamatsu University School of Medicine, Hamamatsu, Japan
| | - Takuya Isayama
- Medical & Biological Laboratories Co., Ltd., Nagoya, Japan
| | - Hyogo Naoi
- Second Division, Department of Internal Medicine, Hamamatsu University School of Medicine, Hamamatsu, Japan
| | - Yuya Aono
- Second Division, Department of Internal Medicine, Hamamatsu University School of Medicine, Hamamatsu, Japan
| | - Mineo Katsumata
- Second Division, Department of Internal Medicine, Hamamatsu University School of Medicine, Hamamatsu, Japan
| | - Hideki Yasui
- Second Division, Department of Internal Medicine, Hamamatsu University School of Medicine, Hamamatsu, Japan
| | - Masato Karayama
- Second Division, Department of Internal Medicine, Hamamatsu University School of Medicine, Hamamatsu, Japan
| | - Yuzo Suzuki
- Second Division, Department of Internal Medicine, Hamamatsu University School of Medicine, Hamamatsu, Japan
| | - Kazuki Furuhashi
- Second Division, Department of Internal Medicine, Hamamatsu University School of Medicine, Hamamatsu, Japan
| | - Tomoyuki Fujisawa
- Second Division, Department of Internal Medicine, Hamamatsu University School of Medicine, Hamamatsu, Japan
| | - Naoki Inui
- Second Division, Department of Internal Medicine, Hamamatsu University School of Medicine, Hamamatsu, Japan; Department of Clinical Pharmacology and Therapeutics, Hamamatsu University School of Medicine, Hamamatsu, Japan
| | - Yutaro Nakamura
- Second Division, Department of Internal Medicine, Hamamatsu University School of Medicine, Hamamatsu, Japan
| | - Takafumi Suda
- Second Division, Department of Internal Medicine, Hamamatsu University School of Medicine, Hamamatsu, Japan
| |
Collapse
|
31
|
Shi A, Li J, Qiu X, Sabbah M, Boroumand S, Huang TCT, Zhao C, Terzic A, Behfar A, Moran SL. TGF-β loaded exosome enhances ischemic wound healing in vitro and in vivo. Theranostics 2021; 11:6616-6631. [PMID: 33995680 PMCID: PMC8120220 DOI: 10.7150/thno.57701] [Citation(s) in RCA: 80] [Impact Index Per Article: 20.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2020] [Accepted: 04/06/2021] [Indexed: 01/01/2023] Open
Abstract
Rationale: With over seven million infections and $25 billion treatment cost, chronic ischemic wounds are one of the most serious complications in the United States. The controlled release of bioactive factor enriched exosome from finbrin gel was a promising strategy to promote wound healing. Methods: To address this unsolved problem, we developed clinical-grade platelets exosome product (PEP), which was incorporate with injectable surgical fibrin sealant (TISSEEL), to promote chronic wound healing and complete skin regeneration. The PEP characterization stimulated cellular activities and in vivo rabbit ischemic wound healing capacity of TISSEEL-PEP were performed and analyzed. Results: PEP, enriched with transforming growth factor beta (TGF-β), possessed exosomal characteristics including exosome size, morphology, and typical markers including CD63, CD9, and ALG-2-interacting protein X (Alix). In vitro, PEP significantly promoted cell proliferation, migration, tube formation, as well as skin organoid formation. Topical treatment of ischemic wounds with TISSEEL-PEP promoted full-thickness healing with the reacquisition of hair follicles and sebaceous glands. Superior to untreated and TISSEEL-only treated controls, TISSEEL-PEP drove cutaneous healing associated with collagen synthesis and restoration of dermal architecture. Furthermore, PEP promoted epithelial and vascular cell activity enhancing angiogenesis to restore blood flow and mature skin function. Transcriptome deconvolution of TISSEEL-PEP versus TISSEEL-only treated wounds prioritized regenerative pathways encompassing neovascularization, matrix remodeling and tissue growth. Conclusion: This room-temperature stable, lyophilized exosome product is thus capable of delivering a bioactive transforming growth factor beta to drive regenerative events.
Collapse
Affiliation(s)
- Ao Shi
- Van Cleve Cardiac Regenerative Medicine Program, Center for Regenerative Medicine, Mayo Clinic, Rochester, MN, USA
- Department of Cardiovascular Medicine, Mayo Clinic, Rochester, MN, USA
- Department of Biochemistry and Molecular Biology, Mayo Graduate School of Biomedical Science, Mayo Clinic, Rochester, MN, USA
| | - Jialun Li
- Division of Plastic Surgery, Mayo Clinic, Rochester, MN, USA
- Department of Plastic Surgery, Union Hospital, Huazhong University of Science and Technology, Wuhan, China
| | - Xinyuan Qiu
- Department of Biology and Chemistry, College of Liberal Arts and Sciences, National University of Defense Technology, Changsha, China
| | - Michael Sabbah
- Van Cleve Cardiac Regenerative Medicine Program, Center for Regenerative Medicine, Mayo Clinic, Rochester, MN, USA
- Department of Cardiovascular Medicine, Mayo Clinic, Rochester, MN, USA
| | - Soulmaz Boroumand
- Van Cleve Cardiac Regenerative Medicine Program, Center for Regenerative Medicine, Mayo Clinic, Rochester, MN, USA
| | | | - Chunfeng Zhao
- Department of Orthopedic Surgery, Mayo Clinic, Rochester, MN, USA
| | - Andre Terzic
- Van Cleve Cardiac Regenerative Medicine Program, Center for Regenerative Medicine, Mayo Clinic, Rochester, MN, USA
- Department of Cardiovascular Medicine, Mayo Clinic, Rochester, MN, USA
- Department of Molecular Pharmacology and Experimental Therapeutics, Mayo Clinic, Rochester, MN, USA
- Department of Clinical Genomics, Mayo Clinic, Rochester, MN, USA
| | - Atta Behfar
- Van Cleve Cardiac Regenerative Medicine Program, Center for Regenerative Medicine, Mayo Clinic, Rochester, MN, USA
- Department of Cardiovascular Medicine, Mayo Clinic, Rochester, MN, USA
- Department of Molecular Pharmacology and Experimental Therapeutics, Mayo Clinic, Rochester, MN, USA
| | - Steven L Moran
- Division of Plastic Surgery, Mayo Clinic, Rochester, MN, USA
| |
Collapse
|
32
|
Defrêne J, Berrazouane S, Esparza N, Pagé N, Côté MF, Gobeil S, Aoudjit F, Tessier PA. Deletion of S100a8 and S100a9 Enhances Skin Hyperplasia and Promotes the Th17 Response in Imiquimod-Induced Psoriasis. JOURNAL OF IMMUNOLOGY (BALTIMORE, MD. : 1950) 2021; 206:505-514. [PMID: 33361205 DOI: 10.4049/jimmunol.2000087] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/24/2020] [Accepted: 11/26/2020] [Indexed: 02/04/2023]
Abstract
High concentrations of the damage-associated molecular patterns S100A8 and S100A9 are found in skin and serum from patients suffering from psoriasis, an IL-17-related disease. Notably, although the expression of these proteins correlates with psoriatic disease severity, the exact function of S100A8 and S100A9 in psoriasis pathogenesis remains unclear. In this study, we investigated the role of S100A8 and S100A9 in psoriasis-associated skin hyperplasia and immune responses using S100a8-/- and S100a9-/- mice in an imiquimod-induced model of psoriasis. We found that S100a8-/- and S100a9-/- psoriatic mice exhibit worsened clinical symptoms relative to wild-type mice and increased expression of S100A9 and S100A8 proteins in keratinocytes, respectively. In addition, the loss of S100A8 enhances proliferation of keratinocytes and disrupts keratinocyte differentiation. We further detected elevated production of IL-17A and -F from CD4+ T cells in the absence of S100A8 and S100A9, as well as increased infiltration of neutrophils in the skin. In addition, treatment with anti-IL-17A and -F was found to reduce psoriasis symptoms and skin hyperplasia in S100a8-/- and S100a9-/- mice. These data suggest that S100A8 and S100A9 regulate psoriasis by inhibiting production of IL-17A and -F, thereby, to our knowledge, providing new insights into their biological functions.
Collapse
Affiliation(s)
- Joan Defrêne
- Axe de Recherche sur les Maladies Infectieuses et Immunitaires, Centre de Recherche du Centre Hospitalier Universitaire de Québec-Université Laval, Quebec City, Quebec G1V 4G2, Canada
| | - Sofiane Berrazouane
- Axe de Recherche sur les Maladies Infectieuses et Immunitaires, Centre de Recherche du Centre Hospitalier Universitaire de Québec-Université Laval, Quebec City, Quebec G1V 4G2, Canada
| | - Nayeli Esparza
- Axe de Recherche sur les Maladies Infectieuses et Immunitaires, Centre de Recherche du Centre Hospitalier Universitaire de Québec-Université Laval, Quebec City, Quebec G1V 4G2, Canada
| | - Nathalie Pagé
- Axe de Recherche sur les Maladies Infectieuses et Immunitaires, Centre de Recherche du Centre Hospitalier Universitaire de Québec-Université Laval, Quebec City, Quebec G1V 4G2, Canada
| | - Marie-France Côté
- Axe Endocrinologie et Néphrologie, Centre de Recherche du Centre Hospitalier Universitaire de Québec-Université Laval, Quebec City, Quebec G1V 4G2, Canada
| | - Stéphane Gobeil
- Axe Endocrinologie et Néphrologie, Centre de Recherche du Centre Hospitalier Universitaire de Québec-Université Laval, Quebec City, Quebec G1V 4G2, Canada
- Département de Médecine Moléculaire, Faculté de Médecine, Université Laval, Quebec City, Quebec G1V 0A6, Canada; and
| | - Fawzi Aoudjit
- Axe de Recherche sur les Maladies Infectieuses et Immunitaires, Centre de Recherche du Centre Hospitalier Universitaire de Québec-Université Laval, Quebec City, Quebec G1V 4G2, Canada
- Département de Microbiologie-Infectiologie et d'Immunologie, Faculté de Médecine, Université Laval, Quebec City, Quebec G1V 0A6, Canada
| | - Philippe A Tessier
- Axe de Recherche sur les Maladies Infectieuses et Immunitaires, Centre de Recherche du Centre Hospitalier Universitaire de Québec-Université Laval, Quebec City, Quebec G1V 4G2, Canada;
- Département de Microbiologie-Infectiologie et d'Immunologie, Faculté de Médecine, Université Laval, Quebec City, Quebec G1V 0A6, Canada
| |
Collapse
|
33
|
Machahua C, Guler SA, Horn MP, Planas-Cerezales L, Montes-Worboys A, Geiser TK, Molina-Molina M, Funke-Chambour M. Serum calprotectin as new biomarker for disease severity in idiopathic pulmonary fibrosis: a cross-sectional study in two independent cohorts. BMJ Open Respir Res 2021; 8:8/1/e000827. [PMID: 33451989 PMCID: PMC7813379 DOI: 10.1136/bmjresp-2020-000827] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2020] [Revised: 12/11/2020] [Accepted: 12/13/2020] [Indexed: 12/16/2022] Open
Abstract
BACKGROUND Non-invasive biomarkers for the assessment of disease severity in idiopathic pulmonary fibrosis (IPF) are urgently needed. Calprotectin belongs to the S-100 proteins produced by neutrophils, which likely contribute to IPF pathogenesis. Calprotectin is a well-established biomarker in inflammatory bowel diseases. In this cross-sectional study, we aimed to establish the potential role of calprotectin as a biomarker in IPF. Specifically, we hypothesised that patients with IPF have higher serum calprotectin levels compared with healthy controls, and that calprotectin levels are associated with disease severity. METHODS Blood samples were obtained from healthy volunteers (n=26) and from two independent IPF cohorts (derivation cohort n=26, validation cohort n=66). Serum calprotectin levels were measured with a commercial kit adapted for that purpose and compared between healthy controls and patients with IPF. Clinical parameters, including forced vital capacity, diffusing capacity for carbon monoxide (DLCO) and the Composite Physiologic Index (CPI), were correlated with calprotectin serum levels. RESULTS The IPF derivation cohort showed increased serum calprotectin levels compared with healthy controls (2.47±1.67 vs 0.97±0.53 µg/mL, p<0.001). In addition, serum calprotectin levels correlated with DLCO% predicted (r=-0.53, p=0.007) and with CPI (r=0.66, p=0.007). These findings were confirmed in an independent IPF validation cohort. CONCLUSION Serum calprotectin levels are significantly increased in patients with IPF compared with healthy controls and correlate with DLCO and CPI. Calprotectin might be a potential new biomarker for disease severity in IPF.
Collapse
Affiliation(s)
- Carlos Machahua
- Department for Pulmonary Medicine, Inselspital, Bern University Hospital, Department for BioMedical Research (DBMR), University of Bern, Bern, Switzerland.,Department of Pulmonary Medicine, Inselspital, Bern University Hospital, University of Bern, Bern, Switzerland
| | - Sabina A Guler
- Department for Pulmonary Medicine, Inselspital, Bern University Hospital, Department for BioMedical Research (DBMR), University of Bern, Bern, Switzerland.,Department of Pulmonary Medicine, Inselspital, Bern University Hospital, University of Bern, Bern, Switzerland
| | - Michael P Horn
- Department of Clinical Chemistry, Inselspital University Hospital Bern, Bern, Switzerland
| | - Lurdes Planas-Cerezales
- Unit of interstitial lung disease, Department of Pneumology, University Hospital of Bellvitge L'Hospitalet de Llobregat, Barcelona, Spain.,Pneumology Research Group, Institut D'Investigació Biomedica de Bellvitge IDIBELL, L'Hospitalet de Llobregat, Barcelona, Spain
| | - Ana Montes-Worboys
- Pneumology Research Group, Institut D'Investigació Biomedica de Bellvitge IDIBELL, L'Hospitalet de Llobregat, Barcelona, Spain
| | - Thomas K Geiser
- Department for Pulmonary Medicine, Inselspital, Bern University Hospital, Department for BioMedical Research (DBMR), University of Bern, Bern, Switzerland.,Department of Pulmonary Medicine, Inselspital, Bern University Hospital, University of Bern, Bern, Switzerland
| | - Maria Molina-Molina
- Unit of interstitial lung disease, Department of Pneumology, University Hospital of Bellvitge L'Hospitalet de Llobregat, Barcelona, Spain.,Pneumology Research Group, Institut D'Investigació Biomedica de Bellvitge IDIBELL, L'Hospitalet de Llobregat, Barcelona, Spain
| | - Manuela Funke-Chambour
- Department for Pulmonary Medicine, Inselspital, Bern University Hospital, Department for BioMedical Research (DBMR), University of Bern, Bern, Switzerland .,Department of Pulmonary Medicine, Inselspital, Bern University Hospital, University of Bern, Bern, Switzerland
| |
Collapse
|
34
|
Suzuki M, Ikari J, Anazawa R, Tanaka N, Katsumata Y, Shimada A, Suzuki E, Tatsumi K. PAD4 Deficiency Improves Bleomycin-induced Neutrophil Extracellular Traps and Fibrosis in Mouse Lung. Am J Respir Cell Mol Biol 2021; 63:806-818. [PMID: 32915635 DOI: 10.1165/rcmb.2019-0433oc] [Citation(s) in RCA: 86] [Impact Index Per Article: 21.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022] Open
Abstract
Excessive release of neutrophil extracellular traps (NETs) has been implicated in several organ fibrosis, including pulmonary fibrosis. NETs constitute a phenomenon in which decorated nuclear chromatin with cytosolic proteins is released into the extracellular space. PAD4 (peptidylarginine deiminase 4) plays an important role in the formation of NETs. However, the role of NETs in the pathogenesis of pulmonary fibrosis remains undefined. Here, we identified NETs in the alveolar and interstitial lung space of mice undergoing bleomycin (BLM)-induced lung fibrosis, which was suppressed by a pan-PAD inhibitor, Cl-amidine. In vitro, BLM directly induced NETs in blood neutrophils, which was also inhibited by Cl-amidine. Furthermore, Padi4 gene knockout (PAD4-KO) in mice led to the alleviation of BLM-induced NETs and pulmonary fibrosis and to the expression of inflammatory and fibrotic genes. PAD4 deficiency prevented decreases in alveolar epithelial and pulmonary vascular endothelial cell numbers and increases in ACTA2-positive mesenchymal cells and S100A4-positive fibroblasts in the lung. Hematopoietic cell grafts from PAD4-KO mice, not wild-type mice, resolved BLM-induced lung fibrosis and fibrotic gene expression in wild-type and PAD4-KO mice, suggesting that expression of PAD4 in hematopoietic cells may be involved in the development of lung fibrosis. These data suggest that PAD4 deficiency could ameliorate BLM-induced formation of NETs and lung fibrosis, suggesting that this pathway could serve as a therapeutic target for pulmonary fibrosis treatment.
Collapse
Affiliation(s)
- Masaki Suzuki
- Department of Respirology, Graduate School of Medicine, Chiba University, Chuo-ku, Chiba City, Chiba, Japan
| | - Jun Ikari
- Department of Respirology, Graduate School of Medicine, Chiba University, Chuo-ku, Chiba City, Chiba, Japan
| | - Rie Anazawa
- Department of Respirology, Graduate School of Medicine, Chiba University, Chuo-ku, Chiba City, Chiba, Japan
| | - Nozomi Tanaka
- Department of Respirology, Graduate School of Medicine, Chiba University, Chuo-ku, Chiba City, Chiba, Japan
| | - Yusuke Katsumata
- Department of Respirology, Graduate School of Medicine, Chiba University, Chuo-ku, Chiba City, Chiba, Japan
| | - Ayako Shimada
- Department of Respirology, Graduate School of Medicine, Chiba University, Chuo-ku, Chiba City, Chiba, Japan
| | - Eiko Suzuki
- Department of Respirology, Graduate School of Medicine, Chiba University, Chuo-ku, Chiba City, Chiba, Japan
| | - Koichiro Tatsumi
- Department of Respirology, Graduate School of Medicine, Chiba University, Chuo-ku, Chiba City, Chiba, Japan
| |
Collapse
|
35
|
Hou C, Dolivo D, Rodrigues A, Li Y, Leung K, Galiano R, Hong SJ, Mustoe T. Knockout of sodium channel Na x delays re-epithelializathion of splinted murine excisional wounds. Wound Repair Regen 2020; 29:306-315. [PMID: 33378794 DOI: 10.1111/wrr.12885] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2020] [Revised: 12/02/2020] [Accepted: 12/07/2020] [Indexed: 01/29/2023]
Abstract
Mammalian wound healing is a carefully orchestrated process in which many cellular and molecular effectors respond in concert to perturbed tissue homeostasis in order to close the wound and re-establish the skin barrier. The roles of many of these molecular effectors, however, are not entirely understood. Our lab previously demonstrated that the atypical sodium channel Nax (encoded by Scn7a) responds to wound-induced epidermal dehydration, resulting in molecular cascades that drive pro-inflammatory signaling. Acute inhibition of Nax was sufficient to attenuate dermatopathological symptoms in models of hypertrophic scar and dermatitis. To date, however, the role of Nax in excisional wound healing has not been demonstrated. Here we report development of a knockout mouse that lacks expression of functional Nax , and we demonstrate that lack of functional Nax results in deficient wound healing in a murine splinted excisional wound healing model. This deficiency in wound healing was reflected in impaired re-epithelialization and decreased keratinocyte proliferation, a finding which was further supported by decreased proliferation upon Nax knockdown in HaCaT cells in vitro. Defective wound healing was observed alongside increased expression of inflammatory genes in the wound epidermis of Nax -/- mice, suggesting that mice lacking functional Nax retain the ability to undergo skin inflammation. Our observations here motivate further investigation into the roles of Nax in wound healing and other skin processes.
Collapse
Affiliation(s)
- Chun Hou
- Department of Surgery, Northwestern University-Feinberg School of Medicine, Chicago, Illinois, USA.,Department of Plastic and Cosmetic Surgery, First Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
| | - David Dolivo
- Department of Surgery, Northwestern University-Feinberg School of Medicine, Chicago, Illinois, USA
| | - Adrian Rodrigues
- Department of Surgery, Northwestern University-Feinberg School of Medicine, Chicago, Illinois, USA
| | - Yingxing Li
- Department of Surgery, Northwestern University-Feinberg School of Medicine, Chicago, Illinois, USA
| | - Kai Leung
- Division of Combat Wound Repair, US Army Institute of Surgical Research, JB Fort Sam Houston, San Antonio, Texas, USA
| | - Robert Galiano
- Department of Surgery, Northwestern University-Feinberg School of Medicine, Chicago, Illinois, USA
| | - Seok Jong Hong
- Department of Surgery, Northwestern University-Feinberg School of Medicine, Chicago, Illinois, USA
| | - Thomas Mustoe
- Department of Surgery, Northwestern University-Feinberg School of Medicine, Chicago, Illinois, USA
| |
Collapse
|
36
|
Wilgus TA. Inflammation as an orchestrator of cutaneous scar formation: a review of the literature. PLASTIC AND AESTHETIC RESEARCH 2020; 7:54. [PMID: 33123623 PMCID: PMC7592345 DOI: 10.20517/2347-9264.2020.150] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
Inflammation is a key phase in the cutaneous wound repair process. The activation of inflammatory cells is critical for preventing infection in contaminated wounds and results in the release of an array of mediators, some of which stimulate the activity of keratinocytes, endothelial cells, and fibroblasts to aid in the repair process. However, there is an abundance of data suggesting that the strength of the inflammatory response early in the healing process correlates directly with the amount of scar tissue that will eventually form. This review will summarize the literature related to inflammation and cutaneous scar formation, highlight recent discoveries, and discuss potential treatment modalities that target inflammation to minimize scarring.
Collapse
Affiliation(s)
- Traci A Wilgus
- Department of Pathology, Ohio State University, Columbus, OH 43210, USA
| |
Collapse
|
37
|
Zhao J, Xie P, Galiano RD, Qi S, Mao R, Mustoe TA, Hong SJ. Imiquimod-induced skin inflammation is relieved by knockdown of sodium channel Na x. Exp Dermatol 2020; 28:576-584. [PMID: 30903711 DOI: 10.1111/exd.13917] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2018] [Revised: 02/22/2019] [Accepted: 02/28/2019] [Indexed: 12/30/2022]
Abstract
Nax is an atypical sodium channel that mediates inflammatory pathways in pathological conditions of the skin. In this study, we developed a skin inflammation model in the rabbit ear through application of imiquimod (IMQ). Knockdown of Nax using RNAi attenuated IMQ-induced skin inflammation, including skin erythema, scaling and papule formation. Histologic analysis showed that thickening and insufficient differentiation of the epidermis found in psoriasis-like skin were normalized by administration of Nax -RNAi. Excessive infiltration of inflammatory cells found in inflammatory lesions, such as mast cells, eosinophils, neutrophils, T cells and macrophages, was reduced by Nax -RNAi. Expression of S100A9, which is a downstream gene of Nax and a mediator of inflammation, was decreased by Nax -RNAi. Our results demonstrated that knockdown of Nax ameliorated IMQ-induced psoriasis-like skin inflammation in vivo. Thus, targeting of Nax may represent a potential therapeutic option for the treatment of psoriasis.
Collapse
Affiliation(s)
- Jingling Zhao
- Department of Burns, The First Affiliated Hospital of Sun Yat-sen University, Guangzhou, Guangdong, China.,Department of Surgery/Plastic Surgery Division, Laboratory for Tissue Repair and Regenerative Surgery, Feinberg School of Medicine, Northwestern University, Chicago, Illionis
| | - Ping Xie
- Department of Surgery/Plastic Surgery Division, Laboratory for Tissue Repair and Regenerative Surgery, Feinberg School of Medicine, Northwestern University, Chicago, Illionis
| | - Robert D Galiano
- Department of Surgery/Plastic Surgery Division, Laboratory for Tissue Repair and Regenerative Surgery, Feinberg School of Medicine, Northwestern University, Chicago, Illionis
| | - Shaohai Qi
- Department of Burns, The First Affiliated Hospital of Sun Yat-sen University, Guangzhou, Guangdong, China
| | - Renxiang Mao
- Department of Dermatology, The First Affiliated Hospital of Sun Yat-sen University, Guangzhou, Guangdong, China
| | - Thomas A Mustoe
- Department of Surgery/Plastic Surgery Division, Laboratory for Tissue Repair and Regenerative Surgery, Feinberg School of Medicine, Northwestern University, Chicago, Illionis
| | - Seok Jong Hong
- Department of Surgery/Plastic Surgery Division, Laboratory for Tissue Repair and Regenerative Surgery, Feinberg School of Medicine, Northwestern University, Chicago, Illionis
| |
Collapse
|
38
|
Sreejit G, Abdel Latif A, Murphy AJ, Nagareddy PR. Emerging roles of neutrophil-borne S100A8/A9 in cardiovascular inflammation. Pharmacol Res 2020; 161:105212. [PMID: 32991974 DOI: 10.1016/j.phrs.2020.105212] [Citation(s) in RCA: 42] [Impact Index Per Article: 8.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/27/2020] [Revised: 09/11/2020] [Accepted: 09/21/2020] [Indexed: 02/07/2023]
Abstract
Elevated neutrophil count is associated with higher risk of major adverse cardiac events including myocardial infarction and early development of heart failure. Neutrophils contribute to cardiac damage through a number of mechanisms, including attraction of other immune cells and release of inflammatory mediators. Recently, a number of independent studies have reported a causal role for neutrophil-derived alarmins (i.e. S100A8/A9) in inducing inflammation and cardiac injury following myocardial infarction (MI). Furthermore, a positive correlation between serum S100A8/A9 levels and major adverse cardiac events (MACE) in MI patients was also observed implying that targeting neutrophils or their inflammatory cargo could be beneficial in reducing heart failure. However, contradictory to this idea, neutrophils and neutrophil-derived S100A8/A9 also seem to play a vital role in the resolution of inflammation. Thus, a better understanding of how neutrophils balance these seemingly contrasting functions would allow us to develop effective therapies that preserve the inflammation-resolving function while restricting the damage caused by inflammation. In this review, we specifically discuss the mechanisms behind neutrophil-derived S100A8/A9 in promoting inflammation and resolution in the context of MI. We also provide a perspective on how neutrophils could be potentially targeted to ameliorate cardiac inflammation and the ensuing damage.
Collapse
Affiliation(s)
- Gopalkrishna Sreejit
- Division of Cardiac Surgery, Department of Surgery, The Ohio State University Wexner Medical Center, Columbus, OH, USA
| | - Ahmed Abdel Latif
- Division of Cardiovascular Medicine, Department of Medicine, University of Kentucky, Lexington, KY, USA
| | - Andrew J Murphy
- Baker Heart and Diabetes Institute, Division of Immunometabolism, Melbourne, Australia
| | - Prabhakara R Nagareddy
- Division of Cardiac Surgery, Department of Surgery, The Ohio State University Wexner Medical Center, Columbus, OH, USA.
| |
Collapse
|
39
|
Havlikova J, May RC, Styles IB, Cooper HJ. Direct identification of bacterial and human proteins from infected wounds in living 3D skin models. Sci Rep 2020; 10:11900. [PMID: 32681099 PMCID: PMC7368034 DOI: 10.1038/s41598-020-68233-6] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2019] [Accepted: 06/19/2020] [Indexed: 11/23/2022] Open
Abstract
Trauma is one of the leading causes of death in people under the age of 49 and complications due to wound infection are the primary cause of death in the first few days after injury. The ESKAPE pathogens are a group of bacteria that are a leading cause of hospital-acquired infections and a major concern in terms of antibiotic resistance. Here, we demonstrate a novel and highly accurate approach for the rapid identification of ESKAPE pathogens (Enterococcus faecium, Staphylococcus aureus, Klebsiella pneumoniae, Acinetobacter baumannii, Pseudomonas aeruginosa, and Enterobacter spp.) directly from infected wounds in 3D in vitro skin models. Wounded skin models were inoculated with bacteria and left to incubate. Bacterial proteins were identified within minutes, directly from the wound, by liquid extraction surface analysis mass spectrometry. This approach was able to distinguish closely related strains and, unlike genomic approaches, can be modified to provide dynamic information about pathogen behaviour at the wound site. In addition, since human skin proteins were also identified, this method offers the opportunity to analyse both host and pathogen biomarkers during wound infection in near real-time.
Collapse
Affiliation(s)
- Jana Havlikova
- EPSRC Centre for Doctoral Training in Physical Sciences for Health, University of Birmingham, Edgbaston, Birmingham, B15 2TT, UK.,School of Biosciences, University of Birmingham, Edgbaston, Birmingham, B15 2TT, UK
| | - Robin C May
- School of Biosciences, University of Birmingham, Edgbaston, Birmingham, B15 2TT, UK.,Institute of Microbiology and Infection, University of Birmingham, Edgbaston, Birmingham, B15 2TT, UK
| | - Iain B Styles
- EPSRC Centre for Doctoral Training in Physical Sciences for Health, University of Birmingham, Edgbaston, Birmingham, B15 2TT, UK.,School of Computer Science, University of Birmingham, Edgbaston, Birmingham, B15 2TT, UK.,Centre of Membrane Proteins and Receptors, The Universities of Birmingham and Nottingham, The Midlands, Birmingham, UK.,Alan Turing Institute, 96 Euston Road, London, NW1 2DB, UK
| | - Helen J Cooper
- School of Biosciences, University of Birmingham, Edgbaston, Birmingham, B15 2TT, UK.
| |
Collapse
|
40
|
Xu W, Vebrosky EN, Armbrust KL. Potential toxic effects of 4-OH-chlorothalonil and photodegradation product on human skin health. JOURNAL OF HAZARDOUS MATERIALS 2020; 394:122575. [PMID: 32289622 DOI: 10.1016/j.jhazmat.2020.122575] [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: 02/10/2020] [Revised: 03/20/2020] [Accepted: 03/21/2020] [Indexed: 06/11/2023]
Abstract
Chlorothalonil (CHT) is widely used in agriculture as a fungicide and has been detected in various ecosystems along with its degradation products. A primary intermediate product of degradation, 4-hydroxychlorothalonil (4-OH-CHT) has demonstrated toxic effects on aquatic organisms. However, the toxic effects of 4-OH-CHT on human health and the impacts of environmental factors on the toxicity remain unclear. To understand the environmental modification on the toxicity of 4-OH-CHT to human health, we used a three-dimensional human skin culture model. 4-OH-CHT and irradiated 4-OH-CHT were applied to the model for the dermatoxicity analyses. Although neither the 4-OH-CHT nor the irradiated 4-OH-CHT inhibited the cell proliferation, the 4-OH-CHT significantly attenuated the keratinocyte migration by 26% at a concentration of 20 ppb and by 44 % at 100 ppb. The 4-OH-CHT also demonstrated inhibitory effects on keratinocyte differentiation at both 20 ppb and 100 ppb. In contrast, photodegraded 4-OH-CHT did not show inhibitory effects on the migration and differentiation of the keratinocytes at any concentration. Similarly, the 4-OH-CHT treated 3D keratinocyte culture dramatically activated the co-cultured dermal fibroblast cells by increasing the production of α smooth muscle actin (α-SMA) and pro-Collagen Iα. The mRNA levels of these two proteins were upregulated by 1.13 and 10.97 folds with the stimulation of 100 ppb 4-OH-CHT. The protein level of pro-Collagen Iα in dermal fibroblast cells was increased by 68 % with 100 ppb 4-OH-CHT. The photodegraded 4-OH-CHT failed to activate the co-cultured fibroblast cells. The 4-OH-CHT also enhanced pro-inflammatory cytokine production in keratinocytes compared to the photodegraded products. These results suggest that exposure to environmental 4-OH-CHT could increase the risk of inflammatory skin diseases in humans.
Collapse
Affiliation(s)
- Wei Xu
- Department of Life Sciences, College of Science and Engineering, Texas A&M University Corpus Christi, Corpus Christi, TX, USA.
| | - Emily N Vebrosky
- Department of Environmental Sciences, College of the Coast & Environment, Louisiana State University, Baton Rouge, LA, USA
| | - Kevin L Armbrust
- Department of Environmental Sciences, College of the Coast & Environment, Louisiana State University, Baton Rouge, LA, USA
| |
Collapse
|
41
|
Li J, Liang C, Yang KY, Huang X, Han MY, Li X, Chan VW, Chan KS, Liu D, Huang ZP, Zhou B, Lui KO. Specific ablation of CD4 + T-cells promotes heart regeneration in juvenile mice. Am J Cancer Res 2020; 10:8018-8035. [PMID: 32724455 PMCID: PMC7381734 DOI: 10.7150/thno.42943] [Citation(s) in RCA: 44] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2019] [Accepted: 05/20/2020] [Indexed: 12/21/2022] Open
Abstract
Unlike adult cardiomyocytes, neonatal cardiomyocytes can readily proliferate that contributes to a transient regenerative potential after myocardial injury in mice. We have recently reported that CD4+ regulatory T-cells promote this process; however, the role of other CD4+ T-cell subsets as well as CD8+ T-cells in postnatal heart regeneration has been less studied. Methods: by comparing the regenerating postnatal day (P) 3 and the non-regenerating P8 heart after injury, we revealed the heterogeneity of CD4+ and CD8+ T-cells in the myocardium through single cell analysis. We also specifically ablated CD4+ and CD8+ T-cells using the lytic anti-CD4 and -CD8 monoclonal antibodies, respectively, in juvenile mice at P8 after myocardial injury. Results: we observe significantly more CD4+FOXP3- conventional T-cells in the P8 heart when compared to that of the P3 heart within a week after injury. Surprisingly, such a difference is not seen in CD8+ T-cells that appear to have no function as their depletion does not reactivate heart regeneration. On the other hand, specific ablation of CD4+ T-cells contributes to mitigated cardiac fibrosis and increased cardiomyocyte proliferation after injury in juvenile mice. Single-cell transcriptomic profiling reveals a pro-fibrotic CD4+ T-cell subset in the P8 but not P3 heart. Moreover, there are likely more Th1 and Th17 cells in the P8 than P3 heart. We further demonstrate that cytokines of Th1 and Th17 cells can directly reduce the proliferation and increase the apoptosis of neonatal cardiomyocytes. Moreover, ablation of CD4+ T-cells can directly or indirectly facilitate the polarization of macrophages away from the pro-fibrotic M2-like signature in the juvenile heart. Nevertheless, ablation of CD4+ T-cells alone does not offer the same protection in the adult heart after myocardial infarction, suggesting a developmental change of immune cells including CD4+ T-cells in the regulation of age-related mammalian heart repair. Conclusions: our results demonstrate that ablation of CD4+ but not CD8+ T-cells promotes heart regeneration in juvenile mice; and CD4+ T-cells play a distinct role in the regulation of heart regeneration and repair during development.
Collapse
|
42
|
Hu W, Liu Y, Kan S, Zhang T, Jiang Z, Zhu R. The correlation between imaging expression of P16 and S100 in hypertrophic ligamentum flavum. BMC Musculoskelet Disord 2020; 21:359. [PMID: 32513152 PMCID: PMC7282051 DOI: 10.1186/s12891-020-03395-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/28/2020] [Accepted: 06/02/2020] [Indexed: 11/10/2022] Open
Abstract
Background Lumbar spinal stenosis (LSS) is a common degenerative disease, which can lead to neurological dysfunction and requires surgical treatment. In the previous study, we used H&E staining and immunohistochemistry to qualitatively analyze the expression of S100 and P16 in the pathological process of ligamentum flavum (LF) hypertrophy in patients with LSS. To further explore the relationship between P16, S100 and LF hypertrophy in patients with LSS, we quantitatively detected S100 and P16 and their expressed products based on molecular biology techniques, and analyzed their imaging correlation. Methods Before posterior lumbar surgery, LF thickness was measured by Magnetic Resonance Imaging (MRI). Through the operation, we obtained the specimens of LF from 120 patients, all of whom were L4/5 LF. They were designated: simple lumbar disc herniation (LDH), single-segment spinal stenosis (SLSS), and double-segment LSS (DLSS). The detection of each side of LF was assessed. S100 and P16 and their expression products were detected by western blot and quantitative polymerase chain reaction (qPCR). Results The dorsal mRNA expression of P16 in DLSS group was significantly higher than that in SLSS group. On the dorsal and dural side of LF, the expression of P16 mRNA and proteins in the LDH group was significantly lower than that in SLSS and DLSS groups. We found a correlation between the thickness of LF and the expression of P16. However, there was no significant difference in the expression of S100 mRNA and S100 protein on both sides of the ligament and among the three groups, and no significant correlation between the expression of S100 and the thickness of LF. Conclusions P16 is involved in the process of LF hypertrophy in patients with LSS, and the imaging thickness of LF is related to the expression of P16. No obvious evidence proves that S100 may be related to the hypertrophy of LF in patients with LSS.
Collapse
Affiliation(s)
- Wei Hu
- Department of Spine Surgery, Tianjin Union Medical Center, Tianjin, 300121, China
| | - Yidong Liu
- Department of Spine Surgery, Tianjin Union Medical Center, Tianjin, 300121, China
| | - Shunli Kan
- Department of Spine Surgery, Tianjin Union Medical Center, Tianjin, 300121, China
| | - Tengfei Zhang
- Department of Orthopaedics, Dagang Hospital of Tianjin Binhai New Area, Tianjin, 300270, China
| | - Zehua Jiang
- Department of Spine Surgery, Tianjin Union Medical Center, Tianjin, 300121, China
| | - Rusen Zhu
- Department of Spine Surgery, Tianjin Union Medical Center, Tianjin, 300121, China.
| |
Collapse
|
43
|
Muzumdar S, Koch M, Hiebert H, Bapst A, Gravina A, Bloch W, Beer HD, Werner S, Schäfer M. Genetic activation of Nrf2 reduces cutaneous symptoms in a murine model of Netherton syndrome. Dis Model Mech 2020; 13:dmm042648. [PMID: 32457102 PMCID: PMC7286291 DOI: 10.1242/dmm.042648] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2019] [Accepted: 03/16/2020] [Indexed: 01/08/2023] Open
Abstract
Netherton syndrome is a monogenic autosomal recessive disorder primarily characterized by the detachment of the uppermost layer of the epidermis, the stratum corneum It results from mutations in the SPINK5 gene, which codes for a kallikrein inhibitor. Uncontrolled kallikrein activity leads to premature desquamation, resulting in a severe epidermal barrier defect and subsequent life-threatening systemic infections and chronic cutaneous inflammation. Here, we show that genetic activation of the transcription factor nuclear factor (erythroid-derived 2)-like 2 (Nfe2l2/Nrf2) in keratinocytes of Spink5 knockout mice, a model for Netherton syndrome, significantly alleviates their cutaneous phenotype. Nrf2 activation promoted attachment of the stratum corneum and concomitant epidermal barrier function, and reduced the expression of pro-inflammatory cytokines such as tumor necrosis factor α and thymic stromal lymphopoietin. Mechanistically, we show that Nrf2 activation induces overexpression of secretory leukocyte protease inhibitor (Slpi), a known inhibitor of kallikrein 7 and elastase 2, in mouse and human keratinocytes in vivo and in vitro, respectively. In the Spink5-deficient epidermis, the upregulation of Slpi is likely to promote stabilization of corneodesmosomes, thereby preventing premature desquamation. Our results suggest pharmacological NRF2 activation as a promising treatment modality for Netherton syndrome patients.This article has an associated First Person interview with the first author of the paper.
Collapse
Affiliation(s)
- Sukalp Muzumdar
- Department of Biology, Institute of Molecular Health Sciences, ETH Zurich, 8093 Zurich, Switzerland
| | - Michael Koch
- Department of Biology, Institute of Molecular Health Sciences, ETH Zurich, 8093 Zurich, Switzerland
| | - Hayley Hiebert
- Department of Biology, Institute of Molecular Health Sciences, ETH Zurich, 8093 Zurich, Switzerland
| | - Andreas Bapst
- Department of Biology, Institute of Molecular Health Sciences, ETH Zurich, 8093 Zurich, Switzerland
| | - Alessia Gravina
- Department of Biology, Institute of Molecular Health Sciences, ETH Zurich, 8093 Zurich, Switzerland
| | - Wilhelm Bloch
- Department of Molecular and Cellular Sport Medicine, German Sport University Cologne, 50933 Cologne, Germany
| | - Hans-Dietmar Beer
- Department of Dermatology, University Hospital of Zurich, Gloriastrasse 3, 8091 Zurich, Switzerland
| | - Sabine Werner
- Department of Biology, Institute of Molecular Health Sciences, ETH Zurich, 8093 Zurich, Switzerland
| | - Matthias Schäfer
- Department of Biology, Institute of Molecular Health Sciences, ETH Zurich, 8093 Zurich, Switzerland
| |
Collapse
|
44
|
Steffens S, Van Linthout S, Sluijter JPG, Tocchetti CG, Thum T, Madonna R. Stimulating pro-reparative immune responses to prevent adverse cardiac remodelling: consensus document from the joint 2019 meeting of the ESC Working Groups of cellular biology of the heart and myocardial function. Cardiovasc Res 2020; 116:1850-1862. [DOI: 10.1093/cvr/cvaa137] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/22/2020] [Revised: 03/31/2020] [Accepted: 05/05/2020] [Indexed: 12/14/2022] Open
Abstract
Abstract
Cardiac injury may have multiple causes, including ischaemic, non-ischaemic, autoimmune, and infectious triggers. Independent of the underlying pathophysiology, cardiac tissue damage induces an inflammatory response to initiate repair processes. Immune cells are recruited to the heart to remove dead cardiomyocytes, which is essential for cardiac healing. Insufficient clearance of dying cardiomyocytes after myocardial infarction (MI) has been shown to promote unfavourable cardiac remodelling, which may result in heart failure (HF). Although immune cells are integral key players of cardiac healing, an unbalanced or unresolved immune reaction aggravates tissue damage that triggers maladaptive remodelling and HF. Neutrophils and macrophages are involved in both, inflammatory as well as reparative processes. Stimulating the resolution of cardiac inflammation seems to be an attractive therapeutic strategy to prevent adverse remodelling. Along with numerous experimental studies, the promising outcomes from recent clinical trials testing canakinumab or colchicine in patients with MI are boosting the interest in novel therapies targeting inflammation in cardiovascular disease patients. The aim of this review is to discuss recent experimental studies that provide new insights into the signalling pathways and local regulators within the cardiac microenvironment promoting the resolution of inflammation and tissue regeneration. We will cover ischaemia- and non-ischaemic-induced as well as infection-related cardiac remodelling and address potential targets to prevent adverse cardiac remodelling.
Collapse
Affiliation(s)
- Sabine Steffens
- Institute for Cardiovascular Prevention (IPEK), Ludwig-Maximilians-Universität, Munich, Germany
- German Centre for Cardiovascular Research (DZHK), partner site Munich Heart Alliance, Munich, Germany
| | - Sophie Van Linthout
- Berlin Institute of Health Center for Regenerative Therapies (BCRT), Charité, University Medicine Berlin, Berlin, Germany
- German Centre for Cardiovascular Research (DZHK), partner site Berlin, Germany
| | - Joost P G Sluijter
- Department of Cardiology, Laboratory of Experimental Cardiology, University Medical Center Utrecht, Utrecht, the Netherlands
- Regenerative Medicine Center, Circulatory Health Laboratory, University Medical Center Utrecht, University Utrecht, Utrecht, The Netherlands
| | - Carlo Gabriele Tocchetti
- Department of Translational Medical Sciences and Interdepartmental Center of Clinical and Translational Sciences (CIRCET), Federico II University, Naples, Italy
| | - Thomas Thum
- Institute for Molecular and Translational Therapeutic Strategies (IMTTS), Hannover Medical School, Hannover, Germany
| | - Rosalinda Madonna
- Institute of Cardiology, University of Pisa, Via Paradisa, Pisa 56124, Italy
| |
Collapse
|
45
|
Russo B, Brembilla NC, Chizzolini C. Interplay Between Keratinocytes and Fibroblasts: A Systematic Review Providing a New Angle for Understanding Skin Fibrotic Disorders. Front Immunol 2020; 11:648. [PMID: 32477322 PMCID: PMC7232541 DOI: 10.3389/fimmu.2020.00648] [Citation(s) in RCA: 85] [Impact Index Per Article: 17.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2020] [Accepted: 03/23/2020] [Indexed: 12/20/2022] Open
Abstract
Background/Objective: Skin fibrosis is the result of aberrant processes leading to abnormal deposition of extracellular matrix (ECM) in the dermis. In healthy skin, keratinocytes participate to maintain skin homeostasis by actively crosstalking with fibroblasts. Within the wide spectrum of fibrotic skin disorders, relatively little attention has been devoted to the role of keratinocytes for their capacity to participate to skin fibrosis. This systematic review aims at summarizing the available knowledge on the reciprocal interplay of keratinocytes with fibroblasts and their soluble mediators in physiological states, mostly wound healing, and conditions associated with skin fibrosis. Methods: We performed a systematic literature search on PubMed to identify in vitro and ex vivo human studies investigating the keratinocyte characteristics and their interplay with fibroblasts in physiological conditions and within fibrotic skin disorders including hypertrophic scars, keloids, and systemic sclerosis. Studies were selected according to pre-specified eligibility criteria. Data on study methods, models, stimuli and outcomes were retrieved and summarized according to pre-specified criteria. Results: Among the 6,271 abstracts retrieved, 73 articles were included, of which 14 were specifically dealing with fibrotic skin pathologies. Fifty-six studies investigated how keratinocyte may affect fibroblast responses in terms of ECM-related genes or protein production, phenotype modification, and cytokine production. Most studies in both physiological conditions and fibrosis demonstrated that keratinocytes stimulate fibroblasts through the production of interleukin 1, inducing keratinocyte growth factor (KGF) and metalloproteinases in the fibroblasts. When the potential of keratinocytes to modulate collagen synthesis by healthy fibroblasts was explored, the results were controversial. Nevertheless, studies investigating keratinocytes from fibrotic skin, including keloids, hypertrophic scar, and scleroderma, suggested their potential involvement in enhancing ECM deposition. Twenty-three papers investigated keratinocyte proliferation differentiation and production of soluble mediators in response to interactions with fibroblasts. Most studies showed that fibroblasts modulate keratinocyte viability, proliferation, and differentiation. The production of KGF by fibroblast was identified as key for these functions. Conclusions: This review condenses evidence for the active interaction between keratinocytes and fibroblasts in maintaining skin homeostasis and the altered homeostatic interplay between keratinocytes and dermal fibroblasts in scleroderma and scleroderma-like disorders.
Collapse
Affiliation(s)
- Barbara Russo
- Department of Pathology and Immunology, School of Medicine, University of Geneva, Geneva, Switzerland
| | - Nicolò C Brembilla
- Department of Pathology and Immunology, School of Medicine, University of Geneva, Geneva, Switzerland.,Dermatology, School of Medicine, University Hospital, Geneva, Switzerland
| | - Carlo Chizzolini
- Department of Pathology and Immunology, School of Medicine, University of Geneva, Geneva, Switzerland
| |
Collapse
|
46
|
Knockdown of sodium channel Na x reduces dermatitis symptoms in rabbit skin. J Transl Med 2020; 100:751-761. [PMID: 31925326 DOI: 10.1038/s41374-020-0371-1] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2019] [Revised: 12/02/2019] [Accepted: 12/22/2019] [Indexed: 12/19/2022] Open
Abstract
The skin plays a critical role in maintenance of water homeostasis. Dysfunction of the skin barrier causes not only delayed wound healing and hypertrophic scarring, but it also contributes to the development of various skin diseases. Dermatitis is a chronic inflammatory skin disorder that has several different subtypes. Skin of contact dermatitis and atopic dermatitis (AD) show epidermal barrier dysfunction. Nax is a sodium channel that regulates inflammatory gene expression in response to perturbation of barrier function of the skin. We found that in vivo knockdown of Nax using RNAi reduced hyperkeratosis and keratinocyte hyperproliferation in rabbit ear dermatitic skin. Increased infiltration of inflammatory cells (mast cells, eosinophils, T cells, and macrophages), a characteristic of dermatitis, was reduced by Nax knockdown. Upregulation of PAR-2 and thymic stromal lymphopoietin (TSLP), which induce Th2-mediated allergic responses, was inhibited by Nax knockdown. In addition, expression of COX-2, IL-1β, IL-8, and S100A9, which are downstream genes of Nax and are involved in dermatitis pathogenesis, were also decreased by Nax knockdown. Our data show that knockdown of Nax relieved dermatitis symptoms in vivo and indicate that Nax is a novel therapeutic target for dermatitis, which currently has limited therapeutic options.
Collapse
|
47
|
Vinaik R, Abdullahi A, Barayan D, Jeschke MG. NLRP3 inflammasome activity is required for wound healing after burns. Transl Res 2020; 217:47-60. [PMID: 31843468 PMCID: PMC7036017 DOI: 10.1016/j.trsl.2019.11.002] [Citation(s) in RCA: 48] [Impact Index Per Article: 9.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/30/2019] [Revised: 11/25/2019] [Accepted: 11/25/2019] [Indexed: 12/11/2022]
Abstract
Survival of burn patients is contingent on effective wound healing, a complex process that requires coordinated responses of myeloid cells and inflammatory pathways. NLRP3, which serves as a platform for secretion of proinflammatory cytokines, is implicated as a central regulator of wound healing. However, its role during the acute dermal and epidermal regeneration in the context of burns is unknown. Wild-type (WT) and NLRP3-/- mice were exposed to a 30% TBSA scald burn. Gene expression was conducted via real-time polymerase chain reaction. Trichrome staining was used to assess collagen deposition and granulation tissue formation. F4/80 immunostaining compared macrophage infiltration. Flow cytometric analysis was used to characterize skin macrophage distribution and profile. NLRP3, IL1β and IL18 expression was upregulated in skin after burn, and these changes were nonexistent in NLRP3-/-. NLRP3-/- had decreased expression of proinflammatory cytokines, chemokines, inflammatory markers, and growth factors at 3 days (P < 0.05). NLRP3-/- burn skin demonstrated significantly less macrophage infiltration and higher expression of anti-inflammatory markers Arg1 and Fizz1 (P < 0.05) compared to WT. Trichrome staining showed decreased collagen deposition compared to WT. We show that NLRP3 is protective in burn wound healing, primarily through production of inflammatory mediators, macrophage recruitment, and polarization to a proinflammatory phenotype. Our findings highlight a central role of NLRP3 in wound healing through regulation of inflammation and macrophage polarization after burns.
Collapse
Affiliation(s)
| | | | | | - Marc G Jeschke
- Department of Surgery, Division of Plastic Surgery, University of Toronto, Toronto, Canada; Department of Immunology, University of Toronto, Toronto, Canada; Ross Tilley Burn Centre, Sunnybrook Health Sciences Centre, Toronto, Canada; Sunnybrook Research Institute, Toronto, Canada.
| |
Collapse
|
48
|
Sreejit G, Flynn MC, Patil M, Krishnamurthy P, Murphy AJ, Nagareddy PR. S100 family proteins in inflammation and beyond. Adv Clin Chem 2020; 98:173-231. [PMID: 32564786 DOI: 10.1016/bs.acc.2020.02.006] [Citation(s) in RCA: 59] [Impact Index Per Article: 11.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
The S100 family proteins possess a variety of intracellular and extracellular functions. They interact with multiple receptors and signal transducers to regulate pathways that govern inflammation, cell differentiation, proliferation, energy metabolism, apoptosis, calcium homeostasis, cell cytoskeleton and microbial resistance. S100 proteins are also emerging as novel diagnostic markers for identifying and monitoring various diseases. Strategies aimed at targeting S100-mediated signaling pathways hold a great potential in developing novel therapeutics for multiple diseases. In this chapter, we aim to summarize the current knowledge about the role of S100 family proteins in health and disease with a major focus on their role in inflammatory conditions.
Collapse
Affiliation(s)
| | - Michelle C Flynn
- Division of Immunometabolism, Baker Heart and Diabetes Institute, Melbourne, VIC, Australia
| | - Mallikarjun Patil
- Department of Biomedical Engineering, Schools of Medicine and Engineering, University of Alabama at Birmingham, Birmingham, AL, United States
| | - Prasanna Krishnamurthy
- Department of Biomedical Engineering, Schools of Medicine and Engineering, University of Alabama at Birmingham, Birmingham, AL, United States
| | - Andrew J Murphy
- Division of Immunometabolism, Baker Heart and Diabetes Institute, Melbourne, VIC, Australia; Department of Immunology, Monash University, Melbourne, VIC, Australia
| | | |
Collapse
|
49
|
Meyer M, Ben-Yehuda Greenwald M, Rauschendorfer T, Sänger C, Jukic M, Iizuka H, Kubo F, Chen L, Ornitz DM, Werner S. Mouse genetics identifies unique and overlapping functions of fibroblast growth factor receptors in keratinocytes. J Cell Mol Med 2019; 24:1774-1785. [PMID: 31830366 PMCID: PMC6991627 DOI: 10.1111/jcmm.14871] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2019] [Revised: 11/07/2019] [Accepted: 11/11/2019] [Indexed: 12/13/2022] Open
Abstract
Fibroblast growth factors (FGFs) are key regulators of tissue development, homeostasis and repair, and abnormal FGF signalling is associated with various human diseases. In human and murine epidermis, FGF receptor 3 (FGFR3) activation causes benign skin tumours, but the consequences of FGFR3 deficiency in this tissue have not been determined. Here, we show that FGFR3 in keratinocytes is dispensable for mouse skin development, homeostasis and wound repair. However, the defect in the epidermal barrier and the resulting inflammatory skin disease that develops in mice lacking FGFR1 and FGFR2 in keratinocytes were further aggravated upon additional loss of FGFR3. This caused fibroblast activation and fibrosis in the FGFR1/FGFR2 double‐knockout mice and even more in mice lacking all three FGFRs, revealing functional redundancy of FGFR3 with FGFR1 and FGFR2 for maintaining the epidermal barrier. Taken together, our study demonstrates that FGFR1, FGFR2 and FGFR3 act together to maintain epidermal integrity and cutaneous homeostasis, with FGFR2 being the dominant receptor.
Collapse
Affiliation(s)
- Michael Meyer
- Department of Biology, Institute of Molecular Health Sciences, ETH Zurich, Zurich, Switzerland
| | | | - Theresa Rauschendorfer
- Department of Biology, Institute of Molecular Health Sciences, ETH Zurich, Zurich, Switzerland
| | - Catharina Sänger
- Department of Biology, Institute of Molecular Health Sciences, ETH Zurich, Zurich, Switzerland
| | - Marko Jukic
- Department of Biology, Institute of Molecular Health Sciences, ETH Zurich, Zurich, Switzerland
| | - Haruka Iizuka
- Department of Biology, Institute of Molecular Health Sciences, ETH Zurich, Zurich, Switzerland
| | - Fumimasa Kubo
- Department of Biology, Institute of Molecular Health Sciences, ETH Zurich, Zurich, Switzerland
| | - Lin Chen
- Center of Bone Metabolism and Repair, Department of Rehabilitation Medicine, State Key Laboratory of Trauma, Burns and Combined Injury, Trauma Center, Research Institute of Surgery, Daping Hospital, Third Military Medical University, Chongqing, China
| | - David M Ornitz
- Department of Developmental Biology, Washington University School of Medicine, St. Louis, Missouri
| | - Sabine Werner
- Department of Biology, Institute of Molecular Health Sciences, ETH Zurich, Zurich, Switzerland
| |
Collapse
|
50
|
Hu W, Kan S, Liu G, Cao Z, Zhu R. The expression of P16 and S100 associated with elastin degradation and fibrosis of the Ligamentum Flavum hypertrophy. BMC Musculoskelet Disord 2019; 20:458. [PMID: 31638980 PMCID: PMC6805382 DOI: 10.1186/s12891-019-2825-4] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/30/2019] [Accepted: 09/12/2019] [Indexed: 01/15/2023] Open
Abstract
BACKGROUND One of the characteristics of lumbar spinal stenosis (LSS) is elastin degradation and fibrosis in the ligamentum flavum (LF). However, the biochemical factors that cause these histologic changes is unclear. P16 and S100 participate in scar formation and collagen development in wound healing and fibrosis diseases. In this study, we investigate the association between P16 and S100 expression and the fibrosis of the hypertrophic LF in LSS. METHODS The LF specimens were surgically obtained from 30 patients with single-segment LSS (SLSS), 30 patients with double-segment LSS (DLSS) and 30 patients with L4/5 lumbar disc herniation (LDH). The LF thickness was measured by axial T1-weighted MRI. The extent of LF elastin degradation and fibrosis were graded based on hematoxylin-eosin (HE) and Verhoff's Van Gieson's (VVG) stain, respectively. The localization of P16 and S100 was determined by immunohistochemistry. RESULTS The Absolute and relative LF thickness were greater in the DLSS group compared with the SLSS and LDH groups (p < 0.05). The elastic tissue from the dorsal aspect to the dural aspect in SLSS and DLSS groups was significantly increased. The amount of collagen deposition and elastic tissue is significantly higher in the DLSS group compared with the SLSS and LDH groups (p < 0.05). The specimens in the DLSS group showed positive staining of P16, especially in the dorsal layer. Almost all samples in the SLSS group were partially positive for P16. The LDH group showed negative staining of P16 in both the dural and dorsal layers. All the three groups were stained with S100 in the dorsal layer of the LF. On the contrary, S100 staining was absent in the dural layer of the LF in the three groups. CONCLUSIONS Elastin degradation and fibrosis of the LF in the DLSS patients is more severe compared with the SLSS and LDH patients. Increased expression of P16 associated with LF fibrosis and thickness, suggested that the expression of P16 may related to LF hypertrophy in the patients who suffer with LSS. LF hypertrophy process may not be associated with high expression of S100.
Collapse
Affiliation(s)
- Wei Hu
- Department of Spine Surgery, Tianjin Union Medical Center, Tianjin, 300121, China
| | - Shunli Kan
- Department of Spine Surgery, Tianjin Union Medical Center, Tianjin, 300121, China
| | - Guang Liu
- Department of Pathology, Tianjin Union Medical Center, Tianjin, 300121, China
| | - Zegang Cao
- Department of Spine Surgery, Tianjin Union Medical Center, Tianjin, 300121, China
| | - Rusen Zhu
- Department of Spine Surgery, Tianjin Union Medical Center, Tianjin, 300121, China.
| |
Collapse
|